Tissue Engineering of Vascular Prosthetic Grafts
Tissue Engineering of Vascular Prosthetic Grafts
ENGINEERING
INTELLIGENCE
UNIT 1
Tissue Engineering
of Vascular Prosthetic Grafts
R.G. LANDES
COMPANY
AUSTIN, TEXAS
U.S.A.
TISSUE ENGINEERING INTELLIGENCE UNIT
Tissue Engineering of Vascular Prosthetic Grafts
ISBN: 1-57059-549-6
Tissue engineering of prosthetic vasxcular grafts [edited by] Peter Zilla, Howard P. Greisler.
p. cm.--(Tissue engineering intelligence unit)
Includes bibliographical referecnces and index.
ISBN 1-57059-549-6 (alk. paper)
1. Blood vessel prosthesis. 2. Vascular grafts. 3. Biomedical materials. I. Zilla, P.P. (Peter Paul) II. Greisler,
Howard P. III. Series.
[DNLM: 1. Blood Vessel Prosthesis. 2. Blood Vessels--transplantation. 3. Cell Transplantation. 4. Endothelium,
Vascular--cytology. 5. Host vs Graft reaction--immunology. 6. Biocompatyible Materials. WG 170 T616 1999]
RD598.55.T57 1999
617.4’130592--dc21
DNLM/DLC 99-24890
for Library of Congress CIP
While the authors, editors and publisher believe that drug selection and dosage and the specifications and usage of equipment
and devices, as set forth in this book, are in accord with current recommendations and practice at the time of publication, they
make no warranty, expressed or implied, with respect to material described in this book. In view of the ongoing research,
equipment development, changes in governmental regulations and the rapid accumulation of information relating to the
biomedical sciences, the reader is urged to carefully review and evaluate the information provided herein.
CONTENTS
PART I
Bio-Inert Prostheses: Insufficient Healing
1. The Lack of Healing in Conventional Vascular Grafts ........................................................................................... 3
Lester Davids, Terri Dower, Peter Zilla
Introduction ..................................................................................................................................................................... 3
Midgraft Healing .............................................................................................................................................................. 4
ePTFE Grafts ..................................................................................................................................................................... 5
Dacron Grafts ................................................................................................................................................................... 9
Transanastomotic Healing ............................................................................................................................................. 15
Pannus Tissue ................................................................................................................................................................. 15
Transanastomotic Endothelialization ........................................................................................................................... 17
Porosity ........................................................................................................................................................................... 18
Fibrin ............................................................................................................................................................................... 26
Macrophages ................................................................................................................................................................... 30
Prosthetic Wall Vascularization ..................................................................................................................................... 34
Conclusion ...................................................................................................................................................................... 37
2. Noncompliance: The Silent Acceptance of a Villain ............................................................................................. 45
Alexander M. Seifalian, Alberto Giudiceandrea, Thomas Schmitz-Rixen, George Hamilton
Introduction ................................................................................................................................................................... 45
Historical Overview ........................................................................................................................................................ 45
Physical Properties of the Vessel Wall ........................................................................................................................... 46
Assessment of Compliance ............................................................................................................................................ 46
Synthetic and Biological Grafts ..................................................................................................................................... 49
Causes of Graft Failure ................................................................................................................................................... 50
Tubular Compliance ....................................................................................................................................................... 51
Anastomotic Compliance Mismatch ............................................................................................................................. 51
Signal Transduction Pathways and Flow ....................................................................................................................... 52
Development of a Graft with Better Compliance ......................................................................................................... 52
The Future ...................................................................................................................................................................... 55
PART II
Biolized Prostheses: Surface Healing
3. Endothelial Cell Seeding: A Review ...................................................................................................................... 61
Steven P. Schmidt, Gary L. Bowlin
Introduction ................................................................................................................................................................... 61
4. Surface Precoating in the 1980s:
A First Taste of Cell-Matrix Interactions .......................................................................................................... 69
J. Vincent Smyth, Michael G. Walker
Endothelial Cell Attachment to Prosthetic Grafts ........................................................................................................ 70
Cell Migration ................................................................................................................................................................ 72
Endothelial Cell Retention Under Flow Conditions .................................................................................................... 72
Seeding of Native Vascular Surfaces .............................................................................................................................. 73
Mechanism of Cell Adherence ....................................................................................................................................... 73
Nonendothelial Cell Lines .............................................................................................................................................. 75
Future Directions ........................................................................................................................................................... 75
5. Surface Precoating in the 1990s:
The Fine Tuning of Endothelial Cell Transplantation ..................................................................................... 79
Mark M. Samet, Victor V. Nikolaychik, Peter I. Lelkes
Introduction ................................................................................................................................................................... 79
Endothelialization: Past and Current Approaches ....................................................................................................... 80
Fine Tuning of Endothelialization via Surface Coating ............................................................................................... 80
Morphological Aspects of the Endothelialized Surfaces .............................................................................................. 84
Concluding Remarks ...................................................................................................................................................... 86
Microvascular Endothelial Cell Transplantation ..................................................................................91
6. Microvascular Endothelial Cell Transplantation: A Review ................................................................................ 93
Stuart K. Williams
Early Development of Endothelial Cell Transplantation Technology ........................................................................ 93
Cultured Endothelium for Transplantation ................................................................................................................. 94
Current Considerations in Endothelial Cell Transplantation ..................................................................................... 94
Markers for Isolated Endothelial Cells .......................................................................................................................... 95
Cell Deposition on Graft Surfaces: Seeding vs. Sodding .............................................................................................. 96
Animal Models of Endothelial Cell Transplantation. .................................................................................................. 97
Mechanisms Underlying the Formation of a Neointima in Tissue Engineered Vascular Grafts
Using Microvascular Endothelial Cell Transplantation ............................................................................................ 97
Future of Microvessel Endothelial Cell Transplantation ............................................................................................. 99
7. Morphological Aspects of Microvascular Cell Isolates ...................................................................................... 101
Manuela Vici
Introduction ................................................................................................................................................................. 101
Microvascular Cells ...................................................................................................................................................... 102
Electron Microscopic and Immunocytochemical Profiles of Microvascular Cell Isolates ...................................... 103
Discussion ..................................................................................................................................................................... 110
8. Functional Aspects of Microvascular Cell Isolates ............................................................................................. 115
M. Fittkau, Teddy Fischlein
Antiaggregatory Properties of Microvascular Endothelial Cells ............................................................................... 116
Anticoagulative Properties ........................................................................................................................................... 116
Profibrinolytic and Antifibrinolytic Properties ......................................................................................................... 117
Conclusion .................................................................................................................................................................... 119
9. Automated Seeding Devices ................................................................................................................................ 121
Dominic Dodd, J. Vincent Smyth, Michael G. Walker
Introduction ................................................................................................................................................................. 121
Early Seeding Techniques ............................................................................................................................................. 122
Optimal Conditions for Seeding ................................................................................................................................. 122
Choice of Seeding Technique ....................................................................................................................................... 123
Summary ....................................................................................................................................................................... 125
10. Healing Patterns Following Microvascular Seeding—
A Clinical Evaluation of Microvascular-Seeded A-V Access Grafts ............................................................... 127
Steven P. Schmidt, Sharon O. Meerbaum, Duane L. Donovan
Introduction ................................................................................................................................................................. 127
Materials and Methods ................................................................................................................................................ 128
Results ........................................................................................................................................................................... 129
Discussion ..................................................................................................................................................................... 129
11. Neointimal Hyperplasia in Small Diameter Prosthetic Vascular Grafts:
Influence of Endothelial Cell Seeding with Microvascular
Omental Cells in a Canine Model .................................................................................................................... 131
Miralem Pasic, Werner Müller-Glauser, Marko Turina
Introduction ................................................................................................................................................................. 131
Experimental Studies with Endothelial Cell Seeding at the University Hospital Zurich ......................................... 132
Endothelial Cells Reduce Late Neointimal Proliferation ........................................................................................... 134
Intimal and Neointimal Hyperplasia .......................................................................................................................... 137
Omental Microvascular Cells ...................................................................................................................................... 139
Limitations of Experimental Studies ........................................................................................................................... 140
12. Human Clinical Trials of Microvascular Endothelial Cell Sodding .................................................................. 143
Stuart K. Williams
History of EC Transplantation .................................................................................................................................... 143
Human Trial of Microvascular Endothelial Cell Transplantation ............................................................................. 144
Conclusions .................................................................................................................................................................. 147
Macrovascular Endothelial Cell Transplantation ................................................................................149
13. In Vitro Endothelialization:
Its Contribution Towards an Ideal Vascular Replacement ............................................................................. 151
Peter Zilla
Historical Perspective ................................................................................................................................................... 152
Current and Future Perspective ................................................................................................................................... 153
14. Serial Cultivation of Human Endothelial Cells .................................................................................................. 157
Caroline Gillis-Hægerstrand, Anders Hægerstrand
Introduction ................................................................................................................................................................. 157
Characteristics of the Endothelium ............................................................................................................................ 157
Sources of Endothelial Cells ........................................................................................................................................ 158
Culture Techniques for Human ECs ........................................................................................................................... 158
Does the Culture Technique Matter from a Clinical Perspective? ............................................................................. 159
Future Possibilities of Cultured ECs ........................................................................................................................... 160
15. Risk Factors for Autologous Endothelial Cell Cultures ..................................................................................... 163
Johann Meinhart, Manfred Deutsch, Peter Zilla
Introduction ................................................................................................................................................................. 163
Standard Cell Culture Technique for Autologous Endothelial Cell Cultures ........................................................... 163
Procedure Related Risk Factors ................................................................................................................................... 164
Patient Related Risk Factors ........................................................................................................................................ 165
Summary and Future Aspects ...................................................................................................................................... 166
16. Adhesion Molecule Expression Following In Vitro Lining ................................................................................ 171
Caroline Gillis-Hægerstrand
Introduction ................................................................................................................................................................. 171
Vascular Grafts .............................................................................................................................................................. 171
Autologous Grafts ......................................................................................................................................................... 171
Prosthetic Grafts ........................................................................................................................................................... 171
Glutaraldehyde-Tanned Grafts .................................................................................................................................... 172
Heart Valve Prostheses ................................................................................................................................................. 172
Inflammation ................................................................................................................................................................ 172
Leukocyte Adhesion Under Flow Conditions ............................................................................................................. 173
Endothelialization Reduces Monocyte Adhesion
to Xenogeneic Tissue in a Time Dependent Manner ............................................................................................... 174
Future Prospect: Antiadhesive Therapy? .................................................................................................................... 175
Concluding Remarks .................................................................................................................................................... 175
17. In Vitro Endothelialization Elicits Tissue Remodeling
Emulating Native Artery Structures ................................................................................................................ 179
Manfred Deutsch, Johann Meinhart, Peter Zilla
Case Report ................................................................................................................................................................... 180
Conclusions .................................................................................................................................................................. 183
18. In Vitro Endothelialization of Synthetic Vascular Grafts
in Long Term Clinical Use ................................................................................................................................ 189
Manfred Deutsch, Johann Meinhart, Peter Zilla
Introduction ................................................................................................................................................................. 189
Laboratory Procedure .................................................................................................................................................. 189
Surgical Procedure and Clinical Follow Up ................................................................................................................ 190
Randomized Clinical Study ......................................................................................................................................... 190
Clinical Routine Endothelialization ............................................................................................................................ 191
Conclusion .................................................................................................................................................................... 192
PART III
Biointeractive Prostheses: Complete Healing
Biological Components
Taming of Adverse Responses ...................................................................................................................... 197
Prevention of the Inflammatory Reaction ............................................................................................................ 197
19. Inflammatory Reaction: The Nemesis of Implants ............................................................................................ 197
James M. Anderson
Introduction ................................................................................................................................................................. 197
Inflammation and the Healing Response .................................................................................................................... 197
Foreign Body Reaction ................................................................................................................................................. 199
Macrophage Motility, Adhesion and Activation ......................................................................................................... 201
Foreign Body Giant Cell Formation ............................................................................................................................ 204
Future Perspectives on Inflammatory Responses
to Tissue Engineered Prosthetic Vascular Grafts ..................................................................................................... 205
Taming of Adverse Responses ...................................................................................................................... 207
Prevention of the Inflammatory Reaction ............................................................................................................ 207
20. Molecular Determinants of Acute Inflammatory Responses to Biomaterials ................................................. 207
Liping Tang, John W. Eaton
Introduction ................................................................................................................................................................. 207
Surface-Protein Interactions ........................................................................................................................................ 207
Fibrin(ogen) Is Necessary for Phagocyte Accumulation on Biomaterial Implants .................................................. 209
The Mechanism of Biomaterial-Mediated Inflammatory Responses ....................................................................... 212
Conclusions .................................................................................................................................................................. 214
Taming of Adverse Responses ...................................................................................................................... 219
Prevention of Fibrosis ............................................................................................................................................ 219
21. The Accumulation of Inflammatory Mediators:
A Target for the Prevention of Fibrosis ........................................................................................................... 219
John Zagorski, Sharon M. Wahl
Introduction ................................................................................................................................................................. 219
Inflammation ................................................................................................................................................................ 220
Regulation of Leukocyte Recruitment ........................................................................................................................ 220
Mitigation of Recruitment ........................................................................................................................................... 223
Summary and Perspectives .......................................................................................................................................... 224
Taming of Adverse Responses ...................................................................................................................... 229
Prevention of a Hyperplastic Intimal Response ................................................................................................... 229
22. Pathobiology of Hyperplastic Intimal Responses .............................................................................................. 229
Erik L. Owens, Alexander W. Clowes
Introduction ................................................................................................................................................................. 229
Arteries .......................................................................................................................................................................... 230
Grafts ............................................................................................................................................................................. 235
Summary ....................................................................................................................................................................... 236
Taming of Adverse Responses ...................................................................................................................... 241
Prevention of a Hyperplastic Intimal Response ................................................................................................... 241
23. Cell Cycle Interruption to Inhibit Intimal Hyperplasia .................................................................................... 241
Michael J. Mann, Ruediger C. Braun-Dullaeus, Victor J. Dzau
The Molecular and Cellular Biology of Neointimal Vascular Graft Disease............................................................. 242
Cell Cycle Progression: A Careful Orchestration ....................................................................................................... 243
Cell Cycle Arrest and Neointimal hyperplasia ............................................................................................................ 245
Genetic Engineering of Vein Graft Resistance to Atherosclerosis
via Cell Cycle Gene Blockade .................................................................................................................................... 246
Summary ....................................................................................................................................................................... 247
Facilitation of Healing.................................................................................................................................. 251
Chemotaxis ............................................................................................................................................................ 251
24. Signaling Mechanisms for Vascular Cell Migration ........................................................................................... 251
Ian Zachary
Introduction ................................................................................................................................................................. 251
Migration Factors ......................................................................................................................................................... 252
Future Perspectives ....................................................................................................................................................... 262
Facilitation of Healing.................................................................................................................................. 271
Chemotaxis ............................................................................................................................................................ 271
25. Adhesion Molecules: Potent Inducers of Endothelial Cell Chemotaxis ........................................................... 271
Zoltan Szekanecz, Alisa E. Koch
Introduction ................................................................................................................................................................. 271
The Role of Extracellular Matrix Macromolecule
in Endothelial Cell Motility During Vessel Formation ........................................................................................... 272
Adhesion Molecules in Endothelial Cell Migration and Angiogenesis ..................................................................... 272
Interactions of Soluble Mediators with Cellular Adhesion Molecules
and Extracellular Matrix Components During Endothelial Cell Recruitment
and Neovascularization ............................................................................................................................................. 273
The Relevance of Angiogenesis Studies in Vascular Surgery:
Aortic Aneurysms and Wound Healing .................................................................................................................... 274
A Regulatory Network in Sites of Endothelial Cell Migration:
Potential Target Promoting Graft Healing ............................................................................................................... 275
Facilitation of Healing.................................................................................................................................. 279
Angiogenesis ........................................................................................................................................................... 279
26. Angiogenesis in Tissues and Vascular Grafts ...................................................................................................... 279
Paula K. Shireman, Howard P. Greisler
Overview of Angiogenesis ............................................................................................................................................ 279
Angiogenesis in Vascular Grafts .................................................................................................................................. 282
Therapeutic Angiogenesis in Ischemic Tissues ........................................................................................................... 283
Future Directions ......................................................................................................................................................... 284
Facilitation of Healing.................................................................................................................................. 287
Angiogenesis ........................................................................................................................................................... 287
27. Polypeptide Growth Factors with a Collagen Binding Domain:
Their Potential for Tissue Repair and Organ Regeneration........................................................................... 287
Bo Han, Lynn L.H. Huang, David Cheung, Fabiola Cordoba, Marcel Nimni
Introduction ................................................................................................................................................................. 287
The Collagen Derived Matrix ...................................................................................................................................... 287
Interactions of Biosynthetic Matrices with Cells ....................................................................................................... 289
Modified Growth Factors: TGF-! With a Collagen Binding Domain ....................................................................... 291
Summary and Conclusion ........................................................................................................................................... 297
Facilitation of Healing.................................................................................................................................. 301
Angiogenesis ........................................................................................................................................................... 301
28. Fibroblast Growth Factors in Angiogenesis and Tissue Engineering ............................................................... 301
Karin A. Blumofe, Timothy J. Heilizer, Paula K. Shireman, Howard P. Greisler
Introduction ................................................................................................................................................................. 301
History .......................................................................................................................................................................... 301
Angiogenesis ................................................................................................................................................................. 302
FGF Characterization ................................................................................................................................................... 303
Structure ....................................................................................................................................................................... 303
FGF Secretion ............................................................................................................................................................... 303
Interaction with Heparin ............................................................................................................................................. 304
FGF and Vascular Grafts .............................................................................................................................................. 307
FGF and Ischemia ......................................................................................................................................................... 309
Conclusions .................................................................................................................................................................. 309
Facilitation of Healing .................................................................................................................................. 313
Matrix Degradation .............................................................................................................................................. 313
29. Role of Urokinase-Type Plasminogen Activator (uPA)
in In Vitro Angiogenesis in Fibrin Matrices .................................................................................................... 313
Pieter Koolwijk, Victor W.M. van Hinsbergh
Summary ....................................................................................................................................................................... 313
Introduction. ................................................................................................................................................................ 313
Components of the Plasmin/Plasminogen Activator System .................................................................................... 314
The Regulation of Plasminogen Activation and Pericellular Proteolysis
by Inflammatory and Angiogenic Mediators ........................................................................................................... 316
Interaction Between the uPA/Plasmin System and Matrix-Degrading Metalloproteases ....................................... 316
Involvement of uPA and uPA Receptor in Cell Migration
and Realignment of Endothelial Cells ...................................................................................................................... 317
Role of the Plasmin/Plasminogen Activator System
in the Formation of Endothelial Tubes in a Fibrin Matrix ...................................................................................... 318
Perspective .................................................................................................................................................................... 320
Facilitation of Healing .................................................................................................................................. 325
Matrix Modulation ................................................................................................................................................ 325
30. How Does Extracellular Matrix Control Capillary Morphogenesis? ................................................................ 325
Robert B. Vernon, E. Helene Sage
Angiogenesis: An Introduction .................................................................................................................................... 325
Models of Angiogenesis: The Essential Role of ECM ................................................................................................. 326
Interactions Between ECs and ECM that Mediate Angiogenesis .............................................................................. 330
Facilitation of Healing .................................................................................................................................. 343
Matrix Modulation ................................................................................................................................................ 343
! .......................................................................... 343
31. Collagen Matrices Attenuate Fibroblast Response to TGF-!
Richard R. Clark, John M. McPherson
Introduction ................................................................................................................................................................. 343
Results ........................................................................................................................................................................... 344
Discussion ..................................................................................................................................................................... 348
Facilitation of Healing .................................................................................................................................. 353
Matrix Modulation ................................................................................................................................................ 353
32. Extracellular Matrix Proteins Are Potent Agonists
of Human Smooth Muscle Cell Migration ...................................................................................................... 353
Terry L. Kaiura, K. Craig Kent
Introduction ................................................................................................................................................................. 353
Cell Migration .............................................................................................................................................................. 353
Extracellular Matrix Proteins ...................................................................................................................................... 354
Extracellular Matrix Protein and Migration ............................................................................................................... 355
Integrins ........................................................................................................................................................................ 356
Cell Signaling ................................................................................................................................................................ 357
Conclusion .................................................................................................................................................................... 359
Facilitation of Healing .................................................................................................................................. 363
Matrix Modulation ................................................................................................................................................ 363
33. Extracellular Matrix Effect on Endothelial Control
of Smooth Muscle Cell Migration and Matrix synthesis ............................................................................... 363
Richard J. Powell
Introduction ................................................................................................................................................................. 363
Model ............................................................................................................................................................................ 364
Effect of Matrix on Endothelial Cell Control of Smooth Muscle Cell Migration .................................................... 364
Matrix Effect on Endothelial Cell Control of Smooth Muscle Cell Matrix Synthesis .............................................. 366
Facilitation of Healing.................................................................................................................................. 371
Cell Entrapment from the Circulation .................................................................................................................. 371
34. Circulating Stem Cells: A Fourth Source for the Endothelialization
of Cardiovascular Implants ............................................................................................................................. 371
Willie R. Koen
Introduction ................................................................................................................................................................. 371
The History of Spontaneously Healing Surfaces ........................................................................................................ 371
Properties of Hematopoietic Stem Cells and Progenitor Cells .................................................................................. 372
The Location of Hematopoietic Progenitor Cells ...................................................................................................... 372
Recognition of Stem Cells ............................................................................................................................................ 373
Control of Stem Cells ................................................................................................................................................... 373
Speculative Application ................................................................................................................................................ 375
A Practical Vision for Future Cardiovascular Implants ............................................................................................. 376
Facilitation of Healing.................................................................................................................................. 379
Cell Entrapment from the Circulation .................................................................................................................. 379
35. Surface Population with Blood-Borne Cells ....................................................................................................... 379
William P. Hammond
Background ................................................................................................................................................................... 379
Previous Studies ........................................................................................................................................................... 380
Present Studies .............................................................................................................................................................. 383
Related Findings ........................................................................................................................................................... 383
Implications .................................................................................................................................................................. 384
Facilitation of Healing.................................................................................................................................. 387
Cell Entrapment from the Circulation .................................................................................................................. 387
36. Cellular Population of the Textured-Surface Left Ventricular Assist Devices
Leads to Sustained Activation of a Procoagulant and Proinflammatory Systemic Response ...................... 387
Talia B. Spanier, Ann Marie Schmidt, Mehmet C. Oz
Overview ....................................................................................................................................................................... 387
Thrombin Generation and Fibrinolysis ...................................................................................................................... 388
LVAD Surface Cellularization ...................................................................................................................................... 389
Cellular Activation ....................................................................................................................................................... 394
LVAD as Inflammatory/Immune Organ ..................................................................................................................... 395
NF-∀∀B Is a Marker of Cellular Activation in the LVAD Surface Milieu
and a Target for Anti-Inflammatory Intervention ................................................................................................... 398
Conclusion .................................................................................................................................................................... 400
Facilitation of Healing.................................................................................................................................................. 403
Transdifferentiation .............................................................................................................................................. 403
37. Transdifferentiation and the Vascular Wall ........................................................................................................ 403
William A. Beresford
Endothelial Cells ........................................................................................................................................................... 403
Fibroblasts ..................................................................................................................................................................... 406
Smooth Muscle Cells .................................................................................................................................................... 408
Pericytes ........................................................................................................................................................................ 410
Mast Cells ...................................................................................................................................................................... 410
Macrophages ................................................................................................................................................................. 410
Conclusions .................................................................................................................................................................. 410
Facilitation of Healing.................................................................................................................................. 417
Transdifferentiation .............................................................................................................................................. 417
38. Endothelial Cells Transformed from Fibroblasts During Angiogenesis ........................................................... 417
Takashi Fujiwara, Kazunori Kon
Introduction ................................................................................................................................................................. 417
Angiogenesis in Rabbit Ear Chamber ......................................................................................................................... 417
Discussion ..................................................................................................................................................................... 419
Concluding Remarks .................................................................................................................................................... 422
Facilitation of Healing .................................................................................................................................. 425
Transdifferentiation .............................................................................................................................................. 425
39. Mechanical Forces and Cell Differentiation ....................................................................................................... 425
Ira Mills, Bauer E. Sumpio
Introduction ................................................................................................................................................................. 425
PART III
Biointeractive Prostheses: Complete Healing
Engineering Components .....................................................................................................................439
Scaffold Engineering .................................................................................................................................... 441
Structural Designs ................................................................................................................................................. 441
40. An Integral Mathematical Approach
to Tissue Engineering of Vascular Grafts ........................................................................................................ 441
Greg R. Starke, A.S. Douglas, D.J. Conway
Introduction ................................................................................................................................................................. 441
Inflammation and the Healing Response .................................................................................................................... 441
The Mechanics of Arteries ........................................................................................................................................... 442
Material Laws for the Native Artery and Graft Materials .......................................................................................... 445
Finite Element Implementation .................................................................................................................................. 452
Conclusions .................................................................................................................................................................. 456
Scaffold Engineering .............................................................................................................................459
Material Aspects .................................................................................................................................................... 459
41. Bioinertness: An Outdated Principle .................................................................................................................. 459
David F. Williams
The Convention of Inertness ....................................................................................................................................... 459
The Impossibility of Inertness ..................................................................................................................................... 460
The Requirement for Controlled Reactivity ............................................................................................................... 461
The Essential Requirement of Reactivity in Tissue Engineering ............................................................................... 462
Conclusions .................................................................................................................................................................. 462
Scaffold Engineering .................................................................................................................................... 463
Material Aspects .................................................................................................................................................... 463
42. Bioinert Biomaterials: Are Their Properties Irreplaceable? ............................................................................... 463
Patrick T. Cahalan
Scaffold Engineering .................................................................................................................................... 469
Material Aspects .................................................................................................................................................... 469
43. Biostable Polymers as Durable Scaffolds
for Tissue Engineered Vascular Prostheses ..................................................................................................... 469
Arthur J. Coury
Introduction ................................................................................................................................................................. 469
Design Concept ............................................................................................................................................................ 469
Material Requirements and Limitations ..................................................................................................................... 469
Modes of Polymer Degradation .................................................................................................................................. 470
Polymers with Potential to Serve as Biostable Scaffolds ............................................................................................ 472
Concluding Comments ................................................................................................................................................ 477
Scaffold Engineering .................................................................................................................................... 481
Material Aspects .................................................................................................................................................... 481
44. Biophilic Polymers: What’s on the Horizon? ...................................................................................................... 481
Patrick T. Cahalan
Physical Methods .......................................................................................................................................................... 484
Chemical Methods ........................................................................................................................................................ 486
Summary ....................................................................................................................................................................... 487
Scaffold Engineering .................................................................................................................................... 489
Material Aspects .................................................................................................................................................... 489
45. Bioresorbable Grafts: A Counterintuitive Approach ......................................................................................... 489
David Fox, David A. Vorp, Howard P. Greisler
Overview ....................................................................................................................................................................... 489
Definitions .................................................................................................................................................................... 489
Introduction ................................................................................................................................................................. 489
Single Component Resorbable Grafts ......................................................................................................................... 490
Preventing Aneurysmal Dilatation .............................................................................................................................. 498
Resorbable Outer Wraps .............................................................................................................................................. 500
Biopolymers: Resorbable Prostheses of Biologic Origin ........................................................................................... 500
Conclusion .................................................................................................................................................................... 501
Scaffold Engineering .................................................................................................................................... 505
Material Aspects .................................................................................................................................................... 505
46. Biodegradable Materials ...................................................................................................................................... 505
K.J.L. Burg, S.W. Shalaby
Introduction ................................................................................................................................................................. 505
Criteria for Useful Tissue Engineering Materials ....................................................................................................... 505
Methods of Fabrication of Synthetic Polymeric Materials for Use in Tissue Engineering ...................................... 507
Biodegradable Polymers for Vascular Grafts—Past, Present, and Future ................................................................. 508
Newly Tailored Materials for Tissue Engineering ....................................................................................................... 510
Future Prospectives ...................................................................................................................................................... 510
Scaffold Engineering .................................................................................................................................... 513
Material Aspects .................................................................................................................................................... 513
47. The Influence of Porosity and Surface Roughness on Biocompatibility .......................................................... 513
J.M. Schakenraad, K.H. Lam
Introduction ................................................................................................................................................................. 513
Evaluation Models ........................................................................................................................................................ 514
Interpretation of Results .............................................................................................................................................. 525
Summary and Conclusions .......................................................................................................................................... 528
Scaffold Engineering .................................................................................................................................... 531
Surface Modification ............................................................................................................................................. 531
48. Microgroove Driven Tissue Ingrowth ................................................................................................................ 531
Edwin T. Den Braber, John A. Jansen
Implants, Tissue Engineering, and Biomaterials ........................................................................................................ 531
Surface Micropatterns Manipulating Cellular Behavior ............................................................................................ 532
Changed Cell Shape and Cellular Orientation ........................................................................................................... 533
Cell Attachment on Microtextured Surfaces ............................................................................................................... 534
Other Surface Topography Induced Cell Behavior Alterations In Vitro ................................................................... 538
Microtextured Surfaces In Vivo ................................................................................................................................... 538
The Hypotheses ............................................................................................................................................................ 540
Future Perspectives: Vascular Grafts and Microtextured Surfaces ............................................................................ 540
Scaffold Engineering .................................................................................................................................... 545
Surface Modification ............................................................................................................................................. 545
49. Surface Bonding of Heparin ................................................................................................................................ 545
Patrick T. Cahalan ......................................................................................................................................................................... 545
Scaffold Engineering .................................................................................................................................... 553
Surface Modification ............................................................................................................................................. 553
50. Covalent Grafting of RGD Peptides to Synthetic Surfaces ................................................................................ 553
Nina M.K. Lamba, S.L. Cooper
Introduction ................................................................................................................................................................. 553
Methods to Improve Endothelialization ..................................................................................................................... 553
Immobilization of RGD Peptides ................................................................................................................................ 554
Summary ....................................................................................................................................................................... 558
Matrix Engineering ...................................................................................................................................... 561
51. Hydrogels in Biological Control During Graft Healing ..................................................................................... 561
Jeffrey A. Hubbell
Introduction—Why Hydrogels? .................................................................................................................................. 561
Hydrogel Structure and Synthesis ............................................................................................................................... 562
In Situ Transformations ............................................................................................................................................... 563
Protein and Cell Interactions with Hydrogels ............................................................................................................ 564
Designed Cell Adhesiveness of Hydrogels .................................................................................................................. 566
Nonenzymatic Degradation of Hydrogels .................................................................................................................. 566
Enzymatic Degradation of Hydrogels ......................................................................................................................... 567
Controlled Release ........................................................................................................................................................ 567
Design Principles, Hydrogels for Biological Control During Graft Healing ............................................................ 568
Matrix Engineering ...................................................................................................................................... 571
52. In Vivo Synthesis of Organs
Using Collagen-GAG Copolymers ................................................................................................................... 571
Ioannis V. Yannas
Introduction ................................................................................................................................................................. 571
Summary of Evidence for In Vivo Synthesis of Organs ............................................................................................. 571
Methodological Principles of Induced Regeneration ................................................................................................. 572
Models of the Mechanism of Regeneration ................................................................................................................ 573
Conclusions .................................................................................................................................................................. 575
Matrix Engineering ...................................................................................................................................... 577
53. Artificial Extracellular Matrix Proteins for Graft Design ................................................................................. 577
Alyssa Panitch, David A. Tirrell
Introduction ................................................................................................................................................................. 577
Artificial Proteins ......................................................................................................................................................... 578
Artificial ECM Proteins ................................................................................................................................................ 578
Current and Future Directions .................................................................................................................................... 580
Matrix Engineering ...................................................................................................................................... 583
54. Cell-Extracellular Matrix Interactions Relevant
to Vascular Tissue Engineering ........................................................................................................................ 583
Stephen P. Massia
Introduction ................................................................................................................................................................. 583
Cell-Extracellular Surface Interactions at the Molecular Level ................................................................................. 584
Biomimetic Cell-Adhesive Biomaterials ..................................................................................................................... 586
Cell-ECM Interactions in Vascular Biology ................................................................................................................ 589
Modulating Cell-Extracellular Interactions for Vascular Tissue Engineering .......................................................... 591
Conclusion .................................................................................................................................................................... 593
Matrix Engineering ...................................................................................................................................... 599
55. Use of Hydroxypropylchitosan Acetate as a Carrier
for Growth Factor Release ................................................................................................................................ 599
Keiko Yamamura, Toshitaka Nabeshima, Tsunehisa Sakurai
Introduction ................................................................................................................................................................. 599
Use of HPCHA as a Carrier for bFGF ......................................................................................................................... 600
Influence of HPCHA on Release of bFGF ................................................................................................................... 600
Effect of bFGF-HPCHA on the Endothelialization of Grafts .................................................................................... 602
Concluding Remarks .................................................................................................................................................... 604
Cell Engineering ........................................................................................................................................... 605
56. Transplantation of Transduced Smooth Muscle Cells:
A Vehicle For Local and Systemic Gene Therapy ............................................................................................ 605
Randolph L. Geary, Alexander W. Clowes, Monika M. Clowes
Introduction ................................................................................................................................................................. 605
Smooth Muscle Cells as Targets for Gene Transfer .................................................................................................... 606
Vectors for SMC Gene Transfer. .................................................................................................................................. 606
Retroviral Vectors for Ex Vivo Arterial Gene Transfer ............................................................................................... 606
Retroviral Vectors for SMC Gene Transfer in Prosthetic Vascular Grafts ................................................................. 608
Index.............................................................................................................................................................................. 613
EDITORS
Peter Zilla, M.D., Ph.D.
Cardiovascular Research Unit
University of Cape Town Medical School
Cape Town, South Africa
Chapters 1, 13, 15, 17, 18
CONTRIBUTORS
James M. Anderson, M.D., Ph.D. David Cheung
Institute of Pathology University of Southern California
University Hospital of Cleveland School of Medicine
Cleveland, Ohio, U.S.A. Childrens Hospital Los Angeles
Chapter 19 Division of Surgical Research
Los Angeles, California, U.S.A.
William A. Beresford Chapter 27
Department of Anatomy
Robert C. Byrd Health Science Center Richard R. Clark, M.D.
West Virginia University Department of Dermatology
School of Medicine State University of New York at Stony Brook
Morgantown, West Virginia, U.S.A. Stony Brook, New York, U.S.A.
Chapter 37 Chapter 31
S.L. Cooper
Chapter 50
Fabiola Cordoba Ruediger C. Braun-Dullaeus
University of Southern California Department of Medicine
School of Medicine Brigham & Women’s Hospital
Childrens Hospital Los Angeles Harvard Medical School
Division of Surgical Research Boston, Massachusetts, U.S.A.
Los Angeles, California, U.S.A. Chapter 23
Chapter 27
Victor J. Dzau
Arthur J. Coury, Ph.D. Chairman, Department of Medicine
Focal, Inc. Brigham & Women’s Hospital
Lexington, Massachusetts, U.S.A. Harvard Medical School
Chapter 43 Boston, Massachusetts, U.S.A.
Chapter 23
Lester Davids, M.Sc.
Cardiovascular Research Unit John W Eaton, Ph.D.
University of Cape Town Medical School Department of Pediatrics
Cape Town, South Africa Baylor College of Medicine
Chapter 1 Houston, Texas, U.S.A.
Chapter 20
Edwin T. Den Braber, Ph.D.
Katolieke Universiteit Nijmegen Teddy Fischlein, M.D.
Faculty of Medical Sciences Johann Wolfgang Goethe-Universität Frankfurt
Dental School Klinik für Thorax-, Herz und Gefäßchirurgie
Nijmegen, The Netherlands Frankfurt, Germany
Chapter 48 Chapters 8, 18
Introduction
S eldom has any prosthetic implant become the nemesis of so many manufacturers as small
diameter synthetic vascular grafts. In the era of microsphere-encapsulated cell transplants
and human gene therapy it is astonishing that we still do not fully understand why the in-
corporation of prosthetic cardiovascular grafts does not even remotely resemble healing.
Even if the postnatal organism has mostly forgotten the “restitutio ad integrum” healing of
the fetal period—the complete regeneration of highly complex tissue with its original cell
components—it is nevertheless capable of repairing every nonlethal injury other than chronic
infection with a quiescent scar tissue. In the case of synthetic implants, however, the tissue
response is a far cry from any concluded repair event. The majority of contemporary pros-
thetic vascular grafts are not only continually lacking typical components of a healthy native
artery such as a midgraft endothelium, contractile arrangements of smooth muscle cells,
functionally differentiated elastin formations and other specialized components of a vessel’s
extracellular matrix. Synthetic vascular grafts are also lacking the much simpler ability to
restore tissue integrity through a quiescent scar formation. Even after prolonged periods of
implantation, chronic inflammation dominates the interstices of these prostheses while the
luminal patency is endangered by thrombotic surface appositions or the uncontrolled pro-
liferation of a so-called “neointima”.
Today’s dilemma is certainly the consequence of a multitude of developments of the
past three decades. When modern cardiovascular surgery first set out to replace arteries with
synthetic grafts, a graft had simply to act as a nonleaking blood conduit. When it soon be-
came obvious that the surface thrombogenicity of these prostheses was significantly higher
than that of native vessels, this complication was seen as a shortcoming of imperfect materi-
als rather than the lack of active physiological functions. This material-centered era only
slowly gave way to the realization that the right cells need to be an integral part of a synthetic
substitute vessel. However, a full commitment to truly “healing” grafts was further delayed
by the industry’s fear of disproportionate delays for new products resulting from a new
dimension of complexity.
This historical retrospect makes it clear that the most consequential reason for the
scanty patchwork of data available today lies in the fundamentally different concepts of
graft designs in this earlier era. Instead of a holistic engineering approach which defined all
biological components separately before sensibly combining them, grafts were mostly manu-
factured on the basis of material choices and mechanical strength. Therefore, completed
products were investigated with the intention to fulfill regulatory requirements rather than
learn about principal mechanisms. As a consequence, a multitude of poorly defined variants
such as entirely different synthetics, a wide range of interstitial space dimensions and a variety
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
4 Tissue Engineering of Prosthetic Vascular Grafts
of animal models make it extremely difficult today to ex- ity of studies of the last three decades focused unintention-
tract significant information from decades of prosthetic graft ally on exactly this type of healing. By choosing animal mod-
research. At the same time, biology has only recently provided els which far exceed the human ability of transanastomotic
us with the necessary diagnostic means to analyze tissue re- healing, as well as graft lengths which were in average one
actions with regard to the identification of cell types as well tenth of those clinically used, midgraft healing under ex-
as secretion products. Thus, the emergence of basic biologi- perimental conditions occurred predominantly through
cal tools and engineering technologies in the recent past— anastomotic outgrowth rather than transmural ingrowth.
promising the development of implants which may eventu- Considering the fact, however, that prosthetic arterial graft
ally heal “ad integrum”—does not build on a comprehen- endothelialization in humans—mainly restricted to the anas-
sive knowledge regarding the shortcomings of previous tomotic region—often involves less than one thirtieth of the
concepts. entire surface, facilitated anastomotic outgrowth will most
Since the retrospective deduction of biological prin- likely continue to play a limited role.
ciples from data which had mostly been obtained with a dif- Therefore, in spite of the countless studies which in-
ferent intention will not be able to create a full understand- vestigated prosthetic arterial grafts, a description of midgraft
ing of the apparent absence of physiological healing patterns healing—clearly distinguishable from transanastomotic
in prosthetic vascular grafts, a systematic experimental ap- events—is the exception rather than the rule. Nevertheless,
proach will need to supplement the erratic experience of by putting the sporadic observations of all these studies into
the past thirty years. For any eventual conclusions preced- a framework, a surprisingly consistent pattern of healing
ing serious tissue engineering approaches of the future, well became apparent. A further confusion arising from the mul-
planned, biologically oriented in vivo and in vitro experi- titude of animal models used also proved to be less of a prob-
ments will need to be performed. In this chapter we will lem than expected. As with any other aspect of graft heal-
nevertheless attempt to retrospectively identify certain cri- ing, by and large midgraft healing also confirmed the previ-
teria which may have influenced the mitigated healing re- ous belief that it is not the healing pattern itself but its time
sponse of prosthetic vascular grafts in the past. In order to course which distinguishes the various animal models from
avoid dealing with an infinite number of variables, we have each other. However, there are healing aspects like the im-
concentrated our efforts on those two basic graft types which penetrable inner fibrin capsule of Dacron grafts in humans
have been dominating the field of synthetic vascular pros- which may well be based on principal differences between
theses for the past decades: Dacron and expanded the species rather than simply on a protracted time course
polytetrafluoroethylene. of common events. Nevertheless, these principal differences
may again reflect differences in the time course of common
Midgraft Healing sequences, as certain biological phenomena may only occur
No other aspect of prosthetic graft research missed at a certain point in time. Therefore, a solution to graft heal-
the point as fundamentally as did midgraft healing. In spite ing could well lie in the facilitation of the ideal timing of
of a unanimous agreement with regard to the limits of crucial biological events.
transanastomotic tissue outgrowth in man, the vast major-
of cellular coverage.6,9-11 However, anecdotal descriptions multinucleated giant cells13,18,19 and macrophages8-20 demar-
of multilayered cell islands in the central segments of grafts1,5 cating the outer graft surface against a well developed fibro-
add to the decades old controversy concerning the origin of elastic perigraft tissue,1,7 no persistent inflammatory reac-
intimal cells.12 tion is found around the graft.7 However, there is a distinct
Similar to surface healing, transmural tissue ingrowth difference between wrap-reinforced and nonreinforced grafts
remains incomplete. Until week 2 most of the interstices of with regard to the tissue interaction with the outside envi-
the grafts are devoid of any recognizable cellular material,1 ronment. In nonwrapped grafts, relatively few foreign body
with only scanty macrophages beginning to migrate into the giant cells (FBGC) adhere to the PTFE nodes on the outside
micropores.13 After 3 weeks the fibrin matrix in the graft surface, while single macrophages have infiltrated the inter-
wall has slowly become populated with inflammatory cells,2 stices. In wrapped grafts, the narrow structure of the wrap
but there is hardly any connective tissue ingrowth into the limits the infiltration of the interstices with cells. Therefore,
graft.13-17 In the chacma baboon model we find a typical the almost acellular fibrin matrix usually found in the cen-
triple layer picture, with macrophages invading from the ter of the prosthetic wall extends all the way to the outside
blood surface and the adventitia into an otherwise almost surface. This outside surface, in return, borders upon a dis-
acellular central fibrin matrix (Fig. 1.4). Apart from scanty tinct layer of macrophages and FBGC (Fig. 1.4). This indi-
cates once more that the formation of FBGC may be signifi- a more amorphous protein matrix densely covered by plate-
cantly influenced by surface structures, because the mate- lets and a few white blood cells (Fig. 1.7). Overall, the thick-
rial is PTFE in both instances. In the case of the ePTFE wrap ness of the surface thrombus is moderate, because one can
it is difficult to say whether the higher density of FBGC is a recognize the nodal ePTFE structure throughout the graft
result of the finer porosity and structure of the PTFE or the (Fig. 1.8). Histologically, a homogenous fibrin layer covered
overall result of an impenetrable barrier plane compared to by a carpet of platelets enshrouds the ePTFE surfaces. Ex-
the relatively open spaced single nodes in unwrapped ePTFE. cept for very few occasional polymorphnuclear granulocytes,
Eventually, after 6 weeks of implantation, connective tissue this fibrin layer is practically acellular (Fig. 1.7). The pres-
cells occasionally begin to grow into the interstices of un- ence of a layer of macrophages underneath this fibrin ma-
wrapped prosthesis from outside.13,14,17,21-23 Six months af- trix—which is demarcated for the depth of the graft by a
ter implantation, however, connective tissue ingrowth from zone of equally acellular fibrin—indicates that the surface
outside still remains moderate and mostly limited to the fibrin has previously been permeable to inflammatory cells.
outer part of the graft wall,18-20,24-26 while the majority of In contrast to the senescent chacma baboon, high porosity
interstitial graft spaces continue to be primarily occupied ePTFE grafts lead to early and spontaneous
by fibrin and a mild inflammatory infiltrate in the proxim- endothelialization in juvenile recipients such as young yel-
ity of the surfaces.25 In contrast to the early days of implan- low baboons. Patches of endothelial cells30 and capillary ori-
tation, however, these cells lie in a very fine fibrillar extra- fices—approximately 100-500 ∝m apart31—appear on the
cellular meshwork which has replaced the solid acellular fi- blood surface as early as 1-2 weeks after implantation, lead-
brin. If present, this scanty and loose connective tissue con- ing to confluence shortly thereafter.30-35 Since the endothe-
tains hardly any capillaries.113,25 lium soon rests on a well developed layer of actin positive
cells3-33,35 which contains only a few macrophages,36 the fi-
High Porosity ePTFE Grafts brin matrix which initially covers the inner surface and pro-
( > 45 ∝m Internodal Distance) vides the outgrowth substrate for the endothelium repre-
In contrast to low porosity ePTFE grafts, there is a dis- sents a very transient matrix under these circumstances.
tinct difference in high porosity ePTFE grafts with regard to These actin positive cells appearing underneath the endot-
the time course of healing events in humans and different helium9,33 also exhibit the ultrastructural characteristics of
animal models. arterial smooth muscle cells.30 Although proliferating
Initially, the blood surface resembles that of low po- smooth muscle cells resemble wound fibroblasts, there is
rosity ePTFE grafts. It is covered by a thin fibrin layer which currently more evidence against fibroblasts trans-differen-
over time develops into a variable coagulum of fibrin, plate- tiating into smooth muscle cells19,37-40 than there is for it.
lets and erythrocytes.27 In dogs and other more senescent Therefore, it is likely that these cells are derived from
animal models, this stage lacking endothelial coverage per- pericytes accompanying EC.9,33 Over time the endothelium
sists well into the sixth week of implantation.27-29 In the se- and its subendothelial tissue layer develop into a stable
nescent chacma baboon, for instance, we find a six week pan- neointima20,31,35,41,42 with a higher density of actin positive
nus ingrowth of only 7.8 ± 3.5 mm reaching onto otherwise cells adjacent to the endothelium than to the graft sur-
nonendothelialized grafts (Fig. 1.6). The surface of these face.20,26,31,40,43-49 This intimal thickening is evenly distrib-
grafts is covered either by a relatively thin layer of smooth uted along the entire graft surface and not confined to the
fibrin containing white blood cells and a few platelets or by anastomotic region as in low porosity ePTFE grafts.30 It is
particularly noteworthy that these smooth muscle cells only macrophage products stimulate SMC proliferation and high
started to appear in the intima and began to proliferate after ones are inhibitory.69 Eventually the discovery of PDGF-A
endothelial cells had covered the luminal surface.30 With mRNA in the overlying endothelial cells resolved the issue
SMCs multiplying in the inner one third of the intima adja- of the source of SMC mitogens.114b This cytokine is not only
cent to an endothelium33 and without the concomitant pres- one of the strongest currently known proliferative agents
ence of platelets33 the situation is the opposite of trauma- for smooth muscle cells, but was previously already shown
tized arteries. Therefore, it is conceivable that endothelial in vitro to be secreted by endothelial cells vectorially into
cells themselves produce growth promoting substances for the abluminal basal compartment.228 Its strong presence also
smooth muscle cells. This is further supported by the fact explains the 5-100 times higher proliferative activity of SMCs
that after 3 months, SMCs are predominantly found in the and the 10-100 times higher one of endothelial cells50,51 in
subendothelial intima relatively distant from the macroph- grafts than in normal arteries. Nevertheless, the presence of
ages in the interstices.33 Apart from hinting at the involve- higher levels of PDGF and the proof of a higher mitotic ac-
ment of endothelial cells in intimal smooth muscle cell pro- tivity does not indicate whether this is an adaptive response
liferation, this observation of an increasing smooth muscle or a primary phenomenon. On the one hand one could ar-
cell proliferation with increasing distance from the macroph- gue that a high cycling rate may simply be the consequence
ages is inconsistent with the hypothesis that macrophages of an upregulated PDGF production. On the other hand,
are an important source of SMC mitogens,134,189 unless an however, it is also quite likely that a poor anchorage of en-
inverse gradient applies in which low concentrations of dothelial cells at this stage leads to mechanical detachment
The Lack of Healing in Conventional Vascular Grafts 9
and this in turn upregulates PDGF production as a repara- baboon, it takes 2-5 weeks and longer until sprouting capil-
tive response. The latter explanation is supported by the laries begin to reach into the macrophage dominated outer
morphological appearance of endothelial cells. Typical for one third to one half of the graft wall27-29 (Fig. 1.9). Dis-
endothelial surfaces affected by a distinct cell loss,5 these tinctly different from Dacron grafts, these microvessels re-
endothelial cells lack the regular spindle shaped pattern30 semble mature arterioles which are small in diameter (23.06
and are larger than those in arteries.30 This sequence of a ± 13.11 ∝m; personal observation) and regularly accompa-
higher cell loss preceding a higher mitotic activity, rather nied by at least one layer of smooth muscle cells. However,
than a higher cycling rate, being the reason for higher en- in spite of this initial vascularization, the histological pic-
dothelial cell shedding would coincide with the finding for ture of these implants remains dominated by a triple layer
in vitro endothelialized grafts that a distinct degree of en- appearance even after 6 weeks of implantation: While the
dothelial cell detachment occurs during early implantation blood and the adventitial part of the prostheses is infiltrated
in spite of continual endothelial integrity.5 However, since by inflammatory cells, the central zone of the prosthetic wall
the overall thickness of this neointima may also represent often remains filled with an almost acellular amorphous
an adaptation attempt of the tissue to flow conditions in an matrix (Fig. 1.10). Within this triple layered structure the
otherwise rigid graft, the PDGF upregulation in the surface cellularity of the outer third is regularly higher than that of
endothelium could as a third explanation also be a natural the inner third of the graft, although it does not change much
mechanism of macrovascular endothelial cell response to over time. Ham 56 positive macrophages continue to domi-
environmental changes such as flow. This would explain why nate over connective tissue cells,55 while foreign body giant
a many-fold thicker neointima is found under low flow con- cells are conspicuously absent.56 In spite of the scanty pres-
ditions than under high flow conditions.54 ence of connective tissue, the homogenous fibrin matrix ini-
When surface healing occurs so early and indepen- tially filling the interstices gradually gets replaced by a loose
dently of transanastomotic tissue ingrowth, the cell source transparent extracellular meshwork wherever inflammatory
for it can only be perigraft tissue which reaches the blood cell infiltrates reach into the interstices (Fig. 1.11). On the
surface through apt transmural ingrowth. However, since outside, high porosity ePTFE grafts are embedded into a
complete transmural ingrowth does not readily occur in moderately developed mature fibrous tissue which contains
other graft types, it is particularly interesting to see whether hardly any inflammatory cells. Only the direct interface be-
it differs substantially in high porosity ePTFE grafts com- tween adventitial tissue and graft occasionally shows a few
pared with other prostheses. single FBGCs.
Comparably to other grafts, the basically acellular ear-
liest interstitial fibrin matrix gets progressively populated Dacron Grafts
with macrophages and polymorphnuclear leukocytes28,33 In Dacron grafts the issue of porosity is slightly com-
during the initial period of graft implantation followed by plicated through the optional combination of the grafts with
capillary ingrowth. In the juvenile yellow baboon, these cap- a velour surface which differs from the basic texture.
illaries reach through the entire wall as early as after 2 weeks
of implantation. Although fibroblasts from the perigraft re- Low Porosity Dacron Grafts (Woven)
gion soon follow the capillaries,16 the majority of the graft Immediately after implantation a thin layer of fibrin,
interstices still remain populated with macrophages.33 In erythrocytes, white blood cells and platelets is deposited on
more senescent animal models like the dog or the chacma the blood surface of the prosthesis.40,57 During the first few
hours to days this thrombus layer slowly starts thickening appear.58,59 These endothelial islands always rest on the com-
until it reaches a stable equilibrium of compacted fibrin.45 pacted fibrin rather than on the scanty islands of connec-
In dogs this stage is reached within 6 months,45 whereas in tive tissue.
humans it is observed after 1 1/2 years.223 Exceptionally, fi- Underneath this apparently impenetrable fibrin layer,
brous tissue begins to organize the basis of the fibrin cap- healing is most likely to take place; at least with regard to
sule in a few scanty areas of the graft.40,45 This process is perigraft tissue ingrowth, the initial events of graft incorpo-
observed after 4-8 weeks in dogs40,45 and after 1 1/2 years in ration are similar to ePTFE. A fibrin matrix fills the narrow
humans.58 Those few areas of a pseudoneointima where tis- prosthetic interstices within minutes of implantation,57 and
sue reaches the blood surface40,58,59 have a thickness which organizing tissue begins to invade the fibrin layer surround-
is comparable to that of the surrounding compacted fibrin ing the graft to form the outer fibrous tissue capsule.45,57 In
(400-500 ∝ m). 58 In humans these rare and localized the interface between this fibrous tissue and the Dacron yarns
neointimal spots extend over less than 1 mm and are col- a variable degree of foreign body giant cells begins to build
lagen rich, with increasing cellularity towards the graft sur- up.57 Subsequently, a few capillaries and fibroblasts start
face.47 The situation is similar with regard to EC. In areas growing into the tight interstitial spaces of the woven
which represent true midgraft regions—unaffected by anas- Dacron.40,57 This process is quite variable in its extent and
tomotic ingrowth—no endothelial cells are found within the time course. In dogs it can be seen as early as 2-3 weeks after
first 1-2 years.40,58,59 After prolonged implantation periods implantation,40 whereas in humans it may be observed after
of up to 11 years—which are naturally restricted to hu- 5 months11 but may also be absent after up to 18 years.47,58,59
mans—small islands of endothelial cells may occasionally However, even if tissue does occasionally grow through the
interstices of the prostheses, it does not break through the gions in particularly long grafts in which the inner fibrin
compacted fibrin of the inner capsule to reach the blood capsule appears to be both impenetrable for the ingrowing
surface43 even after decades of implantation. interstitial graft tissue43,44,48,59,60,71,72 and nonendothe-
lialized. 60,72,73 This lack of surface healing oc-
High Porosity Dacron Grafts (Knitted) curs43,44,48,59,60,71,72,74,75 although transmural tissue reaches
There is a distinct difference regarding the healing re- the compacted fibrin of the blood surface within 3-4 weeks
sponse to variations in porosity between Dacron grafts and in the calf,60 the yellow baboon35 and the dog,2,76 and 3-6
ePTFE grafts. In ePTFE grafts, the fibrin matrix may get rap- months in humans.48,60 However, in spite of this dominant
idly replaced by inflammatory and connective tissue if the healing deficit, complete tissue replacement of the inner fi-
porosity is increased. In Dacron grafts, the mighty inner fi- brin capsule may sporadically occur in the absence of a sur-
brin capsule mostly persists in both high and low porosity face endothelium and with a poorly differentiated fibroblast
grafts although interstitial healing is more accelerated in the matrix rather than with smooth muscle cells.61 Occasion-
higher porosity prostheses. ally, even complete endothelialization of very long prosthe-
In knitted dacron prostheses the initial surface net of ses is seen46,77 comparable to those in highly porous ePTFE
fibrin, white blood cells, erythrocytes and platelets48 increases grafts. In the yellow baboon, for instance, knitted Dacron
during the first week of implantation to a thickness of grafts may form complete endothelial linings, although not
100-120 ∝m.44,60 During the subsequent 5 weeks of implan- as readily and consistently as in 60 ∝m ePTFE.32,35,78,79 They
tation it appears as if the inner fibrin capsule has not yet initially show small islands of endothelium after 2 weeks,
reached an equilibrium and therefore holds a wide range of and in a minority of implants even confluence after
thickness: 100-500 ∝m in humans,60 30-210 ∝m in dogs2,61-66 4 weeks.32
and 290-300 ∝m in the yellow baboon.35,91 After a subse- However, in contrast to the case of high porosity
quent further insignificant increase,60-62 the thickness of the ePTFE grafts, it remains unclear whether this endothelium
internal capsule eventually reaches an equilibrium before is derived from transmural tissue ingrowth, facilitated
sixth months of implantation.61,62 Although a superficial transanastomotic outgrowth or fall-out healing.12,80 The lat-
screening of the literature suggests that it is only a matter of ter is a phenomenon predominantly observed in Dacron
time until this entire layer of compacted fibrin gets orga- grafts where—apparently independently of transmural tis-
nized by ingrowing tissue, deeper analysis makes it likely that sue ingrowth and with some delay—endothelial islands
this may well be a misinterpretation based on the confusion emerge on the surface of the compacted fibrin. These is-
of midgraft healing with anastomotic outgrowth. It is true lands are well separated from the transmurally ingrowing
that the vast majority of studies describe a mature endothe- fibrous tissue by a distinct layer of compacted fi-
lium and a well developed intimal smooth muscle layer, rest- brin.43,48,59,60,71 They either rest directly on the fibrin43,59,72
ing directly on the graft surface,2,67-69 but the graft length or on a few layers of actin positive cells with no other tissue
and implantation period in these studies make it almost cer- connection,59,71 an observation which can be made in dogs
tain that this mature neointimal tissue represents nothing after 2-6 months72 and in humans perhaps in every fourth
but extended anastomotic outgrowth.2,22,32,35,44,68-70 This graft after 1-11 years.43,59,71 However, although sporadic
suspicion is confirmed by descriptions of true midgraft re- endothelialization occasionally occurs in limited areas of the
grafts, the majority of the surface remains covered by its basal lamina (Fig. 1.11). Those few capillaries which some-
fibrin only. times reach through the entire graft wall and reach the in-
If one focuses on the transmural tissue ingrowth it- ner capsule also remain free of smooth muscle cells.
self, it also begins with the infiltration of the surrounding However, in areas where transanastomotic ingrowth
outer fibrin matrix by granulation tissue.2,48 Initially it is has resulted in a mighty neointima covering the surface, one
primarily macrophages which begin to invade the intersti- occasionally finds true multilayered vessels with smooth
tial fibrin,14 populating the entire depth of the graft at the muscle cells underneath the endothelium.
end of the first month of implantation.60 Early giant cell In summary, the porosity of knitted, and to some ex-
formation surrounding the yarns and—if present—the ve- tent even woven, Dacron prostheses eventually allows a cer-
lour fibrils commences after 7 days and increases, predomi- tain degree of tissue ingrowth from outside. At the same time,
nantly in the outer half of the graft wall,62 during the however, it seems that the compacted inner fibrin capsule
following months.61,62 The subsequent ingrowth of blood on the blood surface of Dacron grafts represents an ingrowth
vessels and fibroblasts varies not only between the different barrier for transmural connective tissue regardless of
animal models but also within one and the same recipient whether the grafts are knitted or woven. Although this bar-
species. In dogs for instance, the interstitial tissue ingrowth rier may eventually be overcome by ingrowth of undiffer-
can either reach the inner fibrin capsule as early as 2-4 weeks entiated fibrous tissue, capillaries remain unconnected to
after implantation2,61,63,76 or be absent as late as after 2 the often poorly endothelialized blood surface.75 The pecu-
months.72,81 Similarly, clinical implants in humans show all liar coincidence of zones which are densely packed with for-
stages of ingrowth for up to three months.179 From then eign body giant cells and hugely dilated capillary sinuses in
onwards, ingrowing adventitial tissue reaches the internal the vicinity is particularly obvious in Dacron prostheses.
fibrous capsule without exception in all animal spe- Moreover, it is also interesting that the fibrin coverage on
cies.2,11,43,59,60,71,81,82 This tissue contains a moderate num- Dacron grafts seems to have a higher propensity to capture
ber of capillaries, fibroblasts and collagen2,11,43,59,71,82 and blood-borne cells than that on ePTFE grafts.80 In this con-
eventually extends along the inside surface of the graft.60,62,71 text it is also noteworthy that the resulting isolated endot-
However, a clear, layered structure of tissue elements still helial islands often rest on equally isolated smooth muscle
dominates the histological picture. The Dacron fibers ap- layers.43,59 However, in spite of the scientific excitement aris-
pear to demarcate themselves expressively against the sur- ing from this observation, the rare and late occurrence of
rounding tissue by a particularly distinct accumulation of the phenomenon makes it the least typical healing charac-
foreign body giant cells (FBGC). Between this outer layer of teristic of Dacron grafts.
the prosthetic wall which is packed with FBGC and the sur-
rounding connective tissue capsule with its longitudinally High Porosity Dacron Velour
aligned collagen bundles,59,60,81 hugely dilated capillary si- The rationale behind a filamentous coverage of Dacron
nuses are usually present during the early weeks of implan- fabric was the attempt to improve the healing between
tation. These sinuses typically consist of a single endothelial perigraft tissue and the prosthesis. Initially only used as an
cell layer which contains no smooth muscle cells or cell types external cover,60 it was soon also applied as an internal
other than macrophages and FBGC, which snuggle against cover.83 Interestingly, it was thought that a finer and more
The Lack of Healing in Conventional Vascular Grafts 13
even porosity would cover the coarser structure of the knit- contains white blood cells in deeper layers which one can
ted fabric underneath and thus achieve facilitated healing. still recognize from the surface (Fig. 1.15). Nevertheless, one
As is obvious from our porosity analysis (see “Porosity”) the also finds areas of densely packed fibrin, covered with mor-
opposite is true, and velour materials actually provide sig- phologically mostly unactivated platelets and white cells
nificantly wider ingrowth spaces than knitted or woven struc- comparable to ePTFE (Fig. 1.16). Very rarely does one see
tures. This wider porosity does result in a firmer tissue inte- vessel openings on the surface (Fig. 1.17). The relatively freely
gration on the outside,15,84 but reduces platelet survival and lying single fibers of the material are completely engulfed
leads to less pronounced pseudointimal development on the by conglomerates of foreign body giant cells, whereas the
inside.85 In order to study the healing pattern of entire ve- interstices are dominated by a loose fibrin matrix contain-
lour structures, we have fabricated graft tubes out of pure ing lymphocytes, polymorphonuclear granulocytes, macro-
velour material and studied the tissue reaction in the chacma phages and FBGCs. In spite of the wide open porosity, hardly
baboon. Similarly to previous results,86 the transanastomotic any fibroblasts or collagen are found in the interstices. The
outgrowth of a neointima is significantly less distinct than huge and dilated capillary sinuses, which were all localized
in nonveloured knitted Dacron, even after weeks of implan- between the outer layer of the graft and the surrounding
tation. The fibrin matrix itself often appears looser than on tissue in knitted prostheses, are equally present but scattered
ePTFE and, moreover, often covered with relatively densely throughout the meshwork of the grafts. These often enor-
adherent macrophages (Fig. 1.14). The loose fibrin often mously dilated capillary sacs are all factor VIII positive, and
equally as devoid of smooth muscle cells as knitted prosthe- region, the widely used term “neointima” in these studies
ses (Fig. 1.18). Occasionally, the simultaneous infiltration actually refers to anastomotic pannus tissue.
mode from inside and outside can still be detected after 6 However, in spite of the poorly defined criteria with
weeks, with a central cell free zone of fibrin wedged between regard to anastomotic tissue ingrowth and the scanty data
the inner and outer graft thirds, the outer being dominated available for some of these criteria, certain trends concern-
by foreign body giant cells. ing animal models, material and porosity still become
These observations indicate that in spite of wide open apparent.
interstitial spaces, there seems to be a distinct inhibition for
connective tissue ingrowth in Dacron scaffolds. It is even Pannus Tissue
more obvious from velour explants than from the narrower Within less than 2 days of implantation, a straighten-
structures of knitted Dacron that capillarization is rather ing and disruption of the internal elastic membrane of the
overshooting than underdeveloped. These widely dilated adjacent native artery occurs.87 Simultaneously, the smooth
endothelial sacs, however, neither reach the blood surface muscle cells of the inner one-third of the media begin to
nor attract concomitantly ingrowing smooth muscle cells. proliferate87 and migrate through the ruptured internal elas-
tic membrane into the intima. It is primarily this hyperplas-
Transanastomotic Healing tic intimal tissue which—together with the endothelium—
The assessment of anastomotic tissue ingrowth is an- migrates and proliferates onto the graft surface, thereby
other ambiguous task within the attempt to interpret the slowly replacing the inner fibrinous capsule from the anas-
healing of prosthetic vascular grafts. Although surface tomosis towards the midgraft. Typically, endothelial cells
endothelialization—which under experimental circum- precede this outgrowth,19,28,88 followed by connective tissue
stances overwhelmingly happens through transanastomotic cells. This means that the foremost edge of the outgrowing
ingrowth—was indirectly the focus of almost every single endothelium initially rests on the fibrin matrix of the inner
study in the past, there is hardly any other aspect of pros- capsule6,28,88 before a few layers of smooth muscle cells ap-
thetic graft healing which is so vaguely described. pear underneath.14,25,27,13 This edge of outgrowing endot-
When all studies included in this review are taken into helium often does not resemble a solidly migrating forma-
account, the average graft length is 10.8 ± 2.6 cm. This value tion of endothelial cells, but rather deeply meandered, tat-
includes the 11% of grafts which were longer than 10 cm. If tered and irregularly shaped islands and tongues of endot-
those grafts—which were substantially longer than the oth- helium (Fig. 1.19). Very often, the foremost edge consists of
ers (36.6 ± 2.4 cm)—are excluded, the mean length of the loosely lying endothelial cells, surrounded by platelet-cov-
remaining 89% of prostheses is only 5.5 ± 1.2 cm. With a ered fibrin (Fig. 1.20). Eventually, a multilayered connective
median implantation period of 91.8 ± 17.3 days in Dacron tissue, which increases in thickness over time, slowly builds
grafts and 60.0 ± 9.36 days in PTFE grafts, it does not come up between the graft and the blood surface.19 This tissue
as a surprise that transanastomotic ingrowth was frequently does not consist exclusively of secretory smooth muscle
completed before the prostheses were retrieved. This often cells69 but also of fibroblasts, collagen and a few macroph-
makes it impossible to determine retrospectively the point ages.30,40,52,55,61,62,68 After a few months a layered structure
in time when the surface coverage through transanastomotic develops with a hypercellular myofibroblast-dominated lu-
tissue ingrowth was completed. To create further uncertainty, minal aspect and a fibro-collagenous and glucosamino-
measurements of tissue ingrowth vary widely amongst those glycan-rich aspect near the graft surface.68,89 However, even
studies where surface endothelialization was not completed at that stage this subendothelial tissue lacks features of na-
prior to explantation, and which could thus be admitted to tive arteries like elastic fibers between the smooth muscle
a comparative ingrowth assessment. cell layers20 and an internal elastic membrane69 separating
Another point of confusion relates to the terms the endothelium from the bulk of smooth muscle cells. Al-
“neointima” and “pannus”. As we have argued under the topic together, the undifferentiated pannus near the anastomosis
“midgraft healing”, prior to the use of high porosity ePTFE of a prosthetic graft has not much in common with the or-
grafts neointimal tissue formation was mostly restricted to ganized intimal structures of a native vessel. Thus,
transanastomotic ingrowth, even if many of the investiga- transanastomotic tissue outgrowth remains a far cry from a
tors were not aware of it. As opposed to the mostly fibrin- self-terminating healing process leading to true neo-artery
ous “pseudoneointima” of midgraft regions, “neointima” structures on the inside of a prosthetic graft.
could almost synonymously be used as the term for In spite of the undifferentiated hyperplastic nature of
transanastomotically ingrowing tissue covering the blood this pannus tissue, however, there are various circumstances
surface of the graft. However, with truly transmurally de- which significantly affect at least its tissue composition and
rived neointimas observed in certain contemporary pros- thickness. Anatomical position, for instance,72,81 as well as
theses, it seems necessary to restrict the term “neointima” to the difference between distal and proximal anasto-
surface tissue which results from transmural midgraft heal- moses64,81,90 are known to influence the thickness of pan-
ing and to keep describing the anastomotic tissue ingrowth nus tissue. Moreover, tissue hyperplasia is also a means of
as “pannus”. As necessary as this restriction of the term compensating for the lack of compliance and contractility
“neointima” is, it eliminates further bits of assumed clarity. in prosthetic grafts. Therefore, variations in flow8,52 and shear
Since the majority of studies describe prostheses in which stress52 result in an adaptive reduction or increase in thick-
the anastomotic pannus has already reached the midgraft ness of the subendothelial tissue.
16 Tissue Engineering of Prosthetic Vascular Grafts
Apart from these influences, it appears as if the thick- and transmural tissue ingrowth does not proceed into the
ness of the anastomotic pannus is to a certain degree also inner capsule of either at this stage, pannus outgrowth onto
predetermined by an almost template function of the initial the graft surface seems to be much faster in knitted
fibrinous inner capsule. Even at an early stage when trans- grafts,2,46,60,68,72,77,81 with their almost continual tissue layer
mural tissue ingrowth has hardly reached the inner fibrin- underneath the fibrin capsule compared with the scanty
ous capsule, pannus formation replicates the dimensions of transmural tissue ingrowth beneath the fibrin matrix of
the fibrin layer which it replaces. Two weeks after implanta- woven grafts.40 Similarly, a significantly faster transan-
tion for instance, the surface thrombus on ePTFE grafts mea- astomotic tissue outgrowth occurs in high porosity ePTFE
sures about one-eighth to one-fourth of that on Dacron grafts if interstitial tissue development is facilitated through
grafts.1,2,44,60 Similarly, the pannus replacing this thrombus external tissue wrapping. This facilitated outgrowth is ob-
in the anastomotic region not only measures one-eighth to served before transmural tissue ingrowth could account
one-fourth on ePTFE grafts compared with Dacron pros- for it.28
theses,2,35 but actually has the same thickness as the preex- As far as the cellular and matrix composition of the
isting fibrin layer.2,8,64 The alacrity, however, with which this pannus tissue is concerned, it appears as if the absence of a
pannus layer grows towards the midgraft region may well mature and confluent endothelium at the time of sub-en-
be accelerated by the presence of tissue underneath the in- dothelial tissue development may be one reason for its lack
ner fibrin capsule, as is the case with high porosity grafts. of differentiation. As early as 1975, Mansfield and Sauvage92
Although the fibrin layer on the blood surface of woven and described the prevention of undifferentiated pannus out-
knitted Dacron grafts is of equal thickness2,45,58,60-62,64-66,91 growth through in vitro endothelialization of grafts in calves.
The Lack of Healing in Conventional Vascular Grafts 17
Almost 20 years later clinical in vitro endothelialization dem- aiming at overcoming this limitation uses animal models
onstrated the same phenomenon: In endothelial lined grafts which characteristically show premature and rapid surface
in which the formation of a confluent endothelium precedes coverage with anastomotic endothelium. In the past this
that of the subendothelial tissue,5 smooth muscle cells rather rapid transanastomotic endothelialization was usually the
than the occasionally observed fibroblasts61 dominate a key parameter of investigations when primarily surface
mature matrix which even contains an internal elastic mem- endothelialization was compared between different pros-
brane.93 The use of purified cultured autologous endothe- thetic grafts. However, if one agrees that future grafts for
lial cells for the in vitro lining process makes it quite un- clinical use will require transmural tissue ingrowth as a main
likely that smooth muscle cell coseeding was the reason for cell source for surface endothelialization because of their
the development of such a well differentiated subendothe- extraordinary length, a better understanding of transan-
lial tissue rather than the interaction with the preexisting astomotic endothelialization is indeed necessary, but for a
endothelium. Since a tissue which lies beneath an internal different reason than before: To be able to clearly define for
elastic membrane qualifies per definition as neo-media each animal model a biological phenomenon which would
rather than hyperplastic neo-intima,93 such a differentiation otherwise continue to blur the distinction line between trans-
means a huge step towards true healing and therefore needs mural and transanastomotic tissue ingrowth.
to be understood with regard to the underlying biological
mechanisms. In another study, similar degrees of pannus Differences Between Animal Models
maturation were observed following endothelial cell seed- Alerted by the human situation of complete ingrowth
ing with mixed microvascular cells.61 In this case, fibroblasts stoppage, we can hardly assume that transanastomotic endo-
were initially coseeded with other cell types at similar pro- thelialization is a linearly progressing event in animal mod-
portions to those normally seen in anastomotic pannus tis- els. Therefore, extrapolation of pointwise observations to a
sue. Nevertheless, the subendothelial matrix—which ma- long term situation, or even to a constant daily outgrowth
tured after the endothelium reached confluence—consisted distance,2,94 seems obsolete, although it would have provided
of mature muscle cells and also elastin rather than the typi- valuable additional data for an ingrowth curve. Even in the
cal poorly differentiated collagen-rich pannus tissue.61 These yellow baboon with its vigorous angiogenic capacity, a simi-
observations support the presumption that the early pres- lar trend of ingrowth stoppage as in humans can be observed:
ence of an endothelium may influence the degree of differ- On 30 ∝m ePTFE prostheses—which do not allow transmu-
entiation of the subendothelial connective tissue of both pan- ral endothelialization—transanastomotic endothelial in-
nus and neo-intimal tissue. growth progressively slows down, to stop approximately 2 cm
from the anastomosis.95-97 Furthermore, high porosity
Transanastomotic Endothelialization ePTFE grafts, as well as a few knitted Dacron grafts, cannot
Endothelial outgrowth from the adjacent artery onto be admitted into this comparison because of complete trans-
the surface of a prosthetic vascular graft is another enigma mural surface endothelialization. Considering these exemp-
of prosthetic graft research. For unknown reasons, tions and the fact that only a minority of studies describe
transanastomotic endothelialization stops shortly after the ingrowth margins rather than percentages of surface cover-
anastomosis in humans.11,47,60 Yet, the experimental set-up age, it is difficult to compare accurately the various animal
models with regard to transanastomotic endothelial cell are usually also senescent at implantation,74 whereas pigs
outgrowth. Nevertheless, since most of the experiments in and calves can be expected to be juvenile. In the rat model,
the past were based on the canine model, a relatively com- the issue of dimensions complicates interpretation. A
plete chronology can be estimated for standard grafts in the transanastomotic endothelial outgrowth of approximately
dog. Single data obtained in other animal models can then 2.2 mm after 6 weeks13,16,17,88 in 30 ∝m ePTFE makes it im-
be related to the time course of surface healing in dogs. How- possible to relate these data to the 7.8-13.6 mm of other large
ever, the accuracy of even this narrowed appraisal decreases animal models.
rapidly after the 12th week of implantation when grafts in In summary, transanastomotic endothelialization is
most of the studies were fully endothelialized and thus can at least 7.5 times more pronounced in any animal model
not be used for the assessment of further transanastomotic than in man. Within the experimental set-up, however, the
endothelial cell outgrowth. senescent primate model of the chacma baboon at least
During the first 3 months of implantation promises to eventually reveal certain biological principles
transanastomotic endothelialization in dogs consecutively which make the human healing pattern of prosthetic grafts
covers a distance of approximately 2 cm in ePTFE so unique.
grafts1-3,10,21,86,98-102 and more than 3 cm in Dacron pros-
theses.2,60,62-64,66,72,76 These ingrowth distances also apply to Differences Between Grafts
the second most commonly used animal model, the yellow Since the endothelial margin represents the frontier
baboon (Papio cynocephalus).9,19,32,35,52,55,91 Taking into ac- of transanastomotically ingrowing tissue,19 it may well be a
count that the ingrowth margin of endothelium is often very key player in a synergistic interaction with sub-fibrinous tis-
irregular with endothelial tongues reaching far into the graft, sue. If one compares the two low porosity prostheses (30 ∝m
a minimal length of 15 cm in all animal models therefore ePTFE and woven Dacron), endothelial outgrowth seems
seems a reasonable choice for future 3 month studies. to be twice as high on ePTFE1-3,10,21,78,86,98-102 than on
In spite of this general guideline, there is still a dis- Dacron.40,43 This difference is not too surprising, because
tinct difference between the respective animal models with the mighty inner fibrinous capsule of Dacron seems to have
regard to transanastomotic ingrowth behavior. However, features of an ingrowth barrier for transmural capillar-
since the in vitro proliferative capacity of endothelial cells ies.43,44,48,59,60,71,72 One can imagine that the same biological
from humans and experimental animals hardly differs,103 reasons which inhibit capillary ingrowth from outside may
the distinctly prostrate transanastomotic outgrowth poten- inhibit endothelial outgrowth from the anastomosis. How-
tial of human endothelial cells must be a consequence of ever, although this fibrin matrix seems to be similar in high
the complex overall situation of the human model rather porosity2,35,44,60-66,91 and low porosity grafts,45,58 it allows
than a principal mitotic sluggishness of human endothelial better transanastomotic endothelial ingrowth under high po-
cells. Therefore, one should not downplay the importance rosity 40,46,47,60,68,72,81 than low porosity condi-
of differences between animal models. If one animal model tions.1,2,21,40,43,86,98-102 Since this observation only relates to
comes significantly closer to the human situation than an- grafts in which the transmural tissue ingrowth did not
other, it may well hold the key to understanding the biologi- traverse the fibrinous capsule and the time would have been
cal reasons for the human failure of graft healing. If too short for fallout endothelialization, the endothelium can
transanastomotic ingrowth values of different species on be assumed to be of transanastomotic origin. If however, a
standard ePTFE grafts are related to those of the dog, the main difference for transanastomotic endothelial ingrowth
coverage which is reached in humans after 56 weeks occurs on an otherwise not ideal surface seems to be a well devel-
in dogs after 3.5 weeks,1,2,21,98,86,100 in the yellow baboon af- oped tissue layer underneath the fibrin capsule, it is reason-
ter 5.6 weeks and in the chacma baboon after 7.6 weeks. Thus, able to speculate that this distant tissue layer may somehow
the transanastomotic outgrowth is 2.1 times slower in the affect surface endothelialization.
chacma baboon than in the dog. The difference between the
two primate species may well be due to the fact that the Porosity
chacma baboon represents a senescent model with a body Porosity was long known to be a determining factor
mass of up to 40 kg, whereas the yellow baboon is a juvenile behind the fate of prosthetic vascular grafts. Initial attempts
model which is on average 2 years old and weighs 10 kg. In to replace arteries with solid tubes of synthetic material soon
this context, it is interesting to note that the difference be- demonstrated that porosity is a prerequisite for graft pa-
tween the two kinds of baboons is much more pronounced tency.45,49 Subsequently, pioneers of prosthetic vascular
in transmural than transanastomotic ingrowth. Transmu- grafts60,71,74 unsuccessfully tried to achieve complete graft
ral capillarization leads to surface endothelialization of high healing through porosity. However, only with the advent of
porosity ePTFE grafts after 2 weeks in the yellow baboon, highly porous ePTFE grafts did it become apparent that com-
whereas perigraft tissue has usually not even grown into the plete surface healing of arterial prostheses was theoretically
outer two-thirds of these prostheses in the chacma baboon possible beyond a certain porosity, and this only in a very
(personal observation). juvenile animal model. In the endothelium-centered 1980s,
Other animal models like rats, calves104 and pigs105 are excitement arose primarily from the fact that higher poros-
either too different or not sufficiently documented. How- ity may allow surface endothelialization.30,32 However, if tis-
ever, pigs are usually large enough for vascular implants at a sue engineering of prosthetic vascular grafts ever intends to
young age, but dog puppies are too small. Therefore, dogs truly emulate functional arteries, the tandem of deep-seated
The Lack of Healing in Conventional Vascular Grafts 19
suspicion of the presence of smooth muscle cells and the water that passes through the material per unit time, assum-
narrow focus on endothelial cells will need to give way to a ing that the thickness of the membrane remains constant.
more comprehensive mode of tissue ingrowth. Neverthe- The resultant measurement has been misleadingly called
less, our current biological understanding validates the view “water porosity” by surgeons and manufacturers alike.
that the ability of a prosthetic graft to allow capillary in-
growth still holds the key to today’s grander picture. Porosity
Researchers were long puzzled as to why interstitial Porosity, in contrast, refers to the void spaces (pores)
graft spaces of particular prostheses were sufficient to allow within the boundaries of a solid material, compared to its
macrophages and other inflammatory cells to immigrate, total volume. Although one could argue that the voids within
whereas connective tissue cells of similar dimensions were a material will allow water to flow through them, numerous
often conspicuously absent.2,18,22,24 Today we understand pores may well be cul-de-sacs and thus fail to provide an
that certain of these connective tissue cells, like smooth open corridor from one side of the graft to the other. More-
muscle cells, mainly follow the ingrowing endothelial over, vascular prostheses are not designed to be rigid struc-
cells.106,107 Therefore, one important determining dimen- tures. They are compressible for ease of handling and can be
sion for complete graft healing is that which mechanically readily deformed by radial and longitudinal tensions as well
allows capillaries to sprout into the meshwork of a synthetic as by internal pressures, even in the relatively noncompliant
graft and still leave some space for accompanying cells. Since contemporary grafts. Such stresses not only change the di-
the average diameter of a capillary is ~10 ∝m,108-110 it would mensions of the prostheses, but also lead to a redistribution
seem as if the minimum area required for ingrowth of cap- of the yarns and fibers within the material. Naturally, the
illaries is at least 20-80 ∝m.2 The diameter of a functional change in textile structure by both circulatory stress and
vessel, i.e., an endothelium and at least one layer of smooth compression by ingrowing tissue itself affects the distribu-
muscle cells, is approximately 23.06 ± 13.11 ∝m in diameter tion, size and tortuosity of the channels that run from one
(personal observations). side of the graft to the other. Since the limiting factor for
The attempt to determine porosity requirements for tissue ingrowth is the bottleneck of the narrowest part of a
graft healing is no doubt complicated by material specific transmural space, it is further important to look at intersti-
characteristics and structural uniqueness. The complex three tial spaces throughout their three dimensional course.
dimensional structure of contemporary grafts results in a Unfortunately, hardly any information is available re-
wide range of voids between the synthetic surfaces, which garding the dimensions of porous structures in vascular
hardly allows a precise definition of ingrowth spaces. For grafts. In order to get a better insight into this critical
manufacturers, a way out of this dilemma was the introduc- parameter for tissue incorporation, we have evaluated in-
tion of water permeability as a means of defining porosity. terstitial space dimensions in a few typical contemporary
With this method, materials could be separated on the basis grafts, both before and after implantation. Moreover, we have
of low and high porosity. For example, materials such as investigated these prostheses regarding their true maximum
Dacron were distinguished not only on the basis of being interstitial ingrowth spaces and their resulting theoretical
knitted and woven, but further characterized as being of high ability to accommodate transmural microvessels.
porosity (1500 to 4000 ml/cm 2/min) or low porosity Microvessels were decided to be the dimension against which
(200-1000 ml/cm2/min).111 The same principle applied to measurements were compared, based on their central role
ePTFE on the basis of distances between the nodes of the in a variety of healing aspects from surface endothelialization
expanded material. However, although these values served to neo-media formation.
as useful parameters in characterizing the materials, they
were not helpful with regard to healing. The poorly defined Theoretical Considerations for Healing
space dimensions made it impossible to come to a conclu- of Vascular Grafts
sive answer concerning the role porosity plays in the mitiga- Although the process of expanding teflon material to
tion of tissue ingrowth into prosthetic vascular grafts. If one the typical nodes-and-fiber structure of ePTFE grafts
attempts to gain a better understanding of the relationship (Fig. 1.22A) allows for a wide range of internodal distances,
of porosity and healing, one needs to make a clear distinc- the resulting changes in ingrowth spaces are moderate. With
tion between porosity and permeability in order to avoid a well-defined anchor points and a rigid material like teflon,
confusion of terminology. It is important to understand that the distensibility of the fibrils is limited (Fig. 1.22B), thus
these terms are not synonymous, because a highly porous making disproportional increases in internodal distance
material may have a low permeability and vice versa. necessary to achieve increases in fibril length and hence an
increase of ingrowth dimensions of a few micrometers. To
Permeability try to correlate true ingrowth spaces with internodal dis-
The permeability of a material can be defined as its tances as defined by the manufacturers of ePTFE grafts is
ability to allow other substances (either gaseous, liquid or insufficient, therefore, if not combined with the assessment
solid) to pass through its pores or interstices. The custom- of fibril dimensions. Furthermore, in order to accurately de-
ary method for measuring permeability is using water and termine ingrowth spaces, one needs to assess these struc-
measuring the area and the applied hydrostatic pressure gra- tures throughout the entire thickness of the graft wall. In a
dient across a porous membrane. The permeability value is similar way, one needs to measure more than just bundle
therefore directly proportional to the measured volume of densities and fiber diameters in knitted (Fig. 1.24) and woven
20 Tissue Engineering of Prosthetic Vascular Grafts
Dacron materials (Fig. 1.25) to obtain an accurate measure- grafts was based on the combined image analyses of histo-
ment of cell ingrowth spaces. These assessments are further logical cross-sections and scanning electron microscopy
complicated by the introduction of externally wrapped PTFE (SEM) of surface structures (see figure legends). Based on
grafts (Fig. 1.23) and veloured Dacron surfaces (Fig. 1.26).60 these measurements (Table 1.1), we attempted to reconstruct
These coverings are intended to prevent the grafts from an- the maximum ingrowth space dimensions throughout the
eurysmal dilatations and to increase the tissue bond between graft wall and draw conclusions about whether the graft
graft and perigraft tissues, with a subsequent better healing materials would sufficiently allow transmural ingrowth for
response.111 microvessels invading from the adventitia. In order to cal-
Taking all these factors into account, our approach culate ingrowth spaces, we measured a number of param-
toward the assessment of true ingrowth spaces in commer- eters related to the material characteristics (Table 1.1).
cially obtained both low porosity (< 30 ∝m) and high po- Scanning electron microscopic examination of the two
rosity (60 ∝m) ePTFE, Dacron velour and knitted Dacron different porosities of ePTFE used in this study gave a good
The Lack of Healing in Conventional Vascular Grafts 21
indication of surface dimensions with respect to internodal the node-to-fibril structure and dimensions. Based on this,
distances, internodal spaces, mean fibril lengths and mean we measured internodal fibril lengths in 6 contemporary
interfibril space (Table 1.1). Internodal distance was mea- 30 ∝ m ePTFE grafts with varying internal diameters
sured from the center of one node to the center of another, (Table 1.2). Only the inner surface was measured, as all the
whereas internodal space width was defined as the actual grafts contained dense outer circumferential wraps some-
space between the margins of two adjacent nodes. No sig- times additionally reinforced with rings. The results showed
nificant difference was found between the inside and out- a reasonably consistent fibril length irrespective of internal
side surfaces for each respective graft. However, the intern- diameter (Table 1.2). The values of 24.28 ± 3.48 ∝m and
odal distance stated by the manufacturers did not precisely 20.69 ± 2.35 ∝m for the 6 and 10 mm I.D. grafts (W.L. Gore
match the internodal distance we measured. and Associates, Flagstaff, Ariz.) respectively, are in agreement
It could be argued that the internal diameter of ePTFE with earlier studies.1 However, although there were hardly
grafts may influence the manufacturing process and thus any differences in internodal distance between grafts of
22 Tissue Engineering of Prosthetic Vascular Grafts
different inner diameter from the same manufacturer, there sumption of a worst scenario was that, beginning at the ad-
were distinct differences in internodal distance between com- ventitial surface, each successive layer of fibrils lies in the
parable grafts of different manufacturers. In 30 ∝m ePTFE center of the preceding layer, thereby creating the greatest
grafts from Impra (Table 1.1), a mean inner fibril length of possible obstruction. Therefore, this approach provides the
17.79 ± 5.62 ∝m was opposed by 23.57 ± 6.21 ∝m measured narrowest possible channel spaces for continual tissue in-
in W.L. Gore products (Table 1.2). growth from outside to inside.
In order to calculate the minimum continual trans- Figures 1.27 and 1.28 illustrate the theoretical com-
mural ingrowth channels through the wall of both ePTFE parison between the minimum continual transmural in-
grafts (Figs. 1.27 and 1.28), a mathematical approach was growth channel and the maximum available ingrowth space
chosen relating three dimensional structures of nodes and of a given layer throughout the walls of the 30 and 60 ∝m
fibers to factors such as the obstruction caused by intern- PTFE grafts. The reason for the discrepancy between the two
odal fibrils from one layer to the next. The underlying as- curves in the 30 ∝m PTFE (Fig. 1.27) is the fact that the
The Lack of Healing in Conventional Vascular Grafts 23
Table 1.1. Material characteristics of 5 mm internal diameter ePTFE grafts (Impra, Inc.)
Table 1.2. Comparative values of contemporary 30∝m ePTFE grafts (W.L. Gore and Associates)
3 21.12±5.56
5 23.57±6.21
6 24.28±3.48
10 20.69±2.35
15 32.09±5.10
22 26.17±2.43
Counts were done on 5 random fields on the inner surface of all the grafts using calibrated NIH image analysis software. The outer surface of
all the grafts was covered with a circumferentially dense wrap. The 5 and 15mm ID grafts were further reinforced with external rings. Results
are presented as mean±SD. A clear discrepancy exists between the 5mm ID internodal fibril lengths of the two manufactured grafts.
maximum space (Fig. 1.27, line marked with solid squares) in contrast, would be permitted by 100% of the transmural
has been worked out on the basis of the maximum ingrowth spaces.
space available between two fibrils without taking the inter-
action of the neighboring fibrils into account. These spaces Assessment of Ingrowth Spaces in Dacron Grafts
are therefore ingrowth spaces in isolation. If one compares If the task of assessing interstitial graft spaces is con-
the maximum ingrowth space available through the graft sidered difficult in ePTFE, it gets even more challenging when
(assuming that each histological layer equals 200 true layers Dacron prostheses are concerned. In the era prior to pro-
as previously established by SEM, resulting in a total of 2000 tein pretreatment of vascular prostheses, porosity was di-
layers throughout the entire wall thickness) to the theoreti- rectly proportional to bleeding. Therefore, textured grafts
cally available transmural ingrowth channel (Fig. 1.27, lined made of polyester braids aimed at providing surgeons with
marked by open diamonds), it becomes obvious that the an instantly hemostatic alternative. In contrast to ePTFE,
available space is in fact much less than it appears initially however, high or low porosity is not primarily achieved
on the basis of isolated layers of nodes and fibrils. Interest- through a variation of the same manufacturing process, but
ingly, the minimal ingrowth channel from outside for 30 ∝m rather through the different procedures of knitting and weav-
PTFE grafts progressively diminishes in size over the first 10 ing. Moreover, an additional variable is introduced by the
layers, as a result of the obstruction caused by the successive needle density used in the knitting process and by optional
layers of fibrils. Thereafter, the values remain consistent be- combination with a velour surface, which further influences
fore increasing again till layer 200. Throughout the rest of the size of the open spaces between the braids.
the graft, the predicted channel size would not accommo-
date capillaries, although the ingrowth space of each iso- Woven Dacron
lated layer of nodes and fibrils would remain theoretically Commercially, woven fabrics consist of two sets of
sufficient for capillary ingrowth. This calculation corre- yarns, the warp and the weft, which are interlaced at right
sponds with the histological observation that only a few cap- angles to each other (Fig. 1.25). Woven material is often pre-
illaries are found in the wall of nonwrapped 30 ∝m PTFE ferred by surgeons, as it is known for its high bursting
grafts, and those few are restricted to the outer third. strengths, low permeability to liquids, minimal tendency to
In 60 ∝m PTFE grafts (Fig. 1.28), the minimum di- deform under stress and less proneness to kinking. How-
mension of transmural ingrowth channels from outside also ever, the woven structure also has poor compliance, limited
progressively diminishes in size in the first 20 layers, after elongation, a tendency to fray and, above all, a very low po-
which the values become consistent throughout the rest of rosity. In spite of this low porosity, tissue ingrowth into the
the graft wall till layer 200. From then onwards the maxi- tight interstitial spaces of the fabric is occasionally ob-
mum diameter of a continual transmural ingrowth channel served.40,57
decreases again as a consequence of the narrowing of the However, since the available space does not appear to
internodal space. Despite the fact that the calculated chan- allow for cell infiltration in the manufactured configuration,
nel size represents the “worst” scenario in terms of ingrowth one must assume that cellular ingrowth is a consequence of
dimensions, as it nears the lumen it still provides sufficient a rearrangement of the relatively loosely lying interstitial fi-
space for ingrowing arterioles within the 5% confidence in- bers. Theoretically, therefore, the number of fibers, fiber di-
terval. Therefore, complete transmural vascularization would ameter and bundle area could be measured to obtain a rea-
still be possible in these grafts. Transmural capillarization, sonable measurement of ingrowth spaces. This information
The Lack of Healing in Conventional Vascular Grafts 25
would then be used to determine a packing factor for the Dacron. We therefore analyzed explanted histological sec-
material. The main shortfall of this approach, however, is tions for knitted as well as pure velour prostheses (Fig. 1.26)
that although the native graft material seems flexible and through ten layered areas from the adventitial surface to the
reorganization of fibers seems theoretically feasible, the in- lumen. The details of the measurements are described in
terstices of the graft upon implantation are soon filled with the figure legends. The commercial knitted graft (Vascutek)
a fibrin matrix which polymerizes into a fairly rigid struc- used in this study had a water porosity of 1350 ml/cm2/min,
ture, thus embedding the fibers and bundles and allowing falling into the lower range of porosities for knitted materi-
very little room for movement. It seems likely that the pro- als.15 On the basis of interfiber measurements through the
teolytic process required for this rearrangement needs a criti- graft wall, a graphical representation showing the mean val-
cal tissue presence with its associated degrading capacity, ues for maximum ingrowth spaces could be obtained
which in turn would need a higher initial porosity. This may (Fig. 1.29). In spite of the fact that this material lies in the
explain why single cells such as macrophages and fibroblasts lower range of porosity within knitted grafts, it was still sur-
are found near the surface of woven prostheses, but only prising to see how narrow the maximum ingrowth spaces
very occasionally capillaries.40,57 through the knitted graft were. When one compares the per-
centage distribution of spaces theoretically permissive for
Knitted Dacron capillaries and/or arterioles (Fig. 1.29B,C), however, the
According to the manufacturers,112 knitted construc- mean size of spaces within the first third of the graft wall
tions contain a set of yarns which are interlooped around (14.44 ± 3.74 ∝m, layers 7-10) would accommodate capil-
each other as opposed to interlaced (Fig. 1.24). The specific laries. In addition, albeit at a low percentage (5.16 ± 0.56%),
type and the compliance behavior of knitted fabrics depends the mean ingrowth space of 24.64 ± 1.75 ∝m would also al-
on the direction in which the yarns are interlooped. If the low a few arterioles to surpass this zone (Fig. 1.29C). To-
yarns lie predominantly in the lengthwise direction then the wards the middle of the graft (layers 4-6) the restriction for
fabric is “warp knit” whereas if they lie in the transverse di- capillaries becomes less pronounced (13.92 ± 2.68 ∝m), de-
rection, the fabric is “weft knit”. A significant difference be- fying the common sense expectation that cells would en-
tween these two types of knit is that weft knit fabrics will counter more tightly packed bundles in this midzone of the
unravel, whereas warp knits will not. This is an important prosthesis. The luminal third of the graft (layers 1-3) has
consideration for the surgeon, as the grafts are often cut at consistently narrow spacing, allowing capillary ingrowth
preimplantation for perfect abutting to the anastomoses. Un- only in 30.4 ± 0.38% with a mean ingrowth space of
doubtedly, once implanted, the difference is negligible. 12.34 ± 2.50 ∝m. None of the spaces in this zone allow arte-
Due to the difference in structure of Dacron grafts as riole ingrowth (Fig. 1.29C).
opposed to PTFE, a slightly different approach was adopted
to calculate ingrowth spaces. Surface measurements made Velour Surfaces
by SEM, and the histological assessment of wall thickness The term “velour” in a textile context refers to a thick-
and ingrowth space dimensions, are the same as in ePTFE. bodied fabric, either woven, knitted or nonwoven, that has a
However, while fibrils cause the main spatial limitation in smooth, soft surface by virtue of additional yarns. All com-
PTFE, fiber orientation and the arrangement of fiber bundles mercial velours, however, are knitted designs. They may be
throughout the graft thickness limit the spaces in knitted distinguished from the earlier knitted prostheses by their
thicker fabric and greater porosity, as well as lower fabric for most of the contemporary vascular prostheses. It becomes
density and packing factor. To assess ingrowth space within clear that, although surface measurements of graft materi-
velour graft types has been very difficult, and most research- als give a good indication of ingrowth characteristics, addi-
ers have relied on methods such as water porosity. However, tional information such as interfiber distances and theoreti-
a few authors113 have attempted to measure its true dimen- cally predicted channel spaces throughout the graft wall are
sions by looking at pile height and the amount of piles in critical for assessing the degree of pure mechanical restric-
the veloured knit. The impetus for using velours stemmed tions for tissue ingrowth into prosthetic vascular grafts.
from a conviction that a rougher and more porous surface
both externally and internally would facilitate the develop- Fibrin
ment of neointimal and perigraft tissue. Prior to implantation, conventional nonpretreated
In order to assess the qualities of pure velour struc- grafts represent simply synthetic scaffolds. Therefore, all bio-
tures, we manufactured velour tubes made of Dacron ve- logical components which eventually fill the interstices of
lour felt (Fig. 1.26, inset). In sharp contrast to the seemingly these scaffolds are host derived. Within this process of host
restrictive interfiber spaces of the knitted material, the incorporation, proteinaceous ingrowth matrices naturally
Dacron velour showed mean ingrowth spaces ranging from precede infiltration with cellular components. Since the
30.02 ± 2.95 ∝m at the adventitia, to 36.88 ± 20.06 ∝m at the implantation of a vascular prostheses inflicts a surgical
lumen, with an overall mean ingrowth space of wound, this proteinaceous matrix is fibrinous. In low po-
27.97 ± 4.80 ∝m (Fig. 1.30A). These mean ingrowth spaces, rosity grafts, the surrounding fibrin coagulum derives en-
as in the case of the knitted grafts, were assessed on the basis tirely from iatrogenically injured vessels. The cut ends of
of histological measurements of interfiber distances through these vessels constrict within a few minutes and get sealed
ten consecutive areas of explanted samples (see figure leg- with aggregating blood platelets. As a result, the hematoma
end). Interestingly, although the mean space seems sufficient around the graft usually remains moderate. Within a few
for arteriole ingrowth, the wall profile (Fig. 1.30A) shows minutes tissue thromboplastin generates thrombin, which,
that the space becomes narrowed towards the central part together with platelet factor 3 (PF 3) released by the few
of the graft and may have a restrictive effect on ingrowth. entrapped platelets, leads to the transformation of fibrino-
The reason for this central restriction is the presence of nu- gen into fibrin. Particularly in nonpretreated porous grafts,
merous bundles in this area causing an overall smaller in- blood may also extravasate from the graft lumen to the out-
terfiber space. Despite this restriction, the calculated maxi- side. In this case the activation of the clotting cascade oc-
mum spaces would still allow the ingrowth of capillaries and curs through contact of factors XII, prekallikrein, XI and
arterioles at all levels. Percentage distribution profiles for high molecular weight kinogen114 and, again, through re-
both the capillaries and arterioles (Fig. 1.30B,C) show a simi- leased platelet PF 3. Although blood does not seep through
lar distribution pattern as the mean ingrowth space profile the wall of less porous grafts, plasma still clots within the
(Fig. 1.30A), with the majority of spaces of more than 23 ∝m synthetic meshwork through a similar mechanism. There-
(Fig. 1.30C) being in the outer and inner one-third of the fore, one can assume that the entire fibrinous matrix ini-
graft. tially surrounding the graft and filling its interstices is rep-
Taking all these results into account, it seems that the resentative of typical wound fibrin, which distinctly favors
dimensions of graft porosity may have been overestimated tissue ingrowth.
In contrast to this loose initial fibrin matrix, an en- the responsiveness of cells to PDGF.120 This is an important
tirely different fibrin clot may be continually generated on aspect of the triggering of angiogenesis at such an early stage.
the blood surface. While the interstitial and external clot- Furthermore, TGF-!—which is a chemotactically highly po-
ting environment is characterized by a small blood pool with tent cytokine for macrophages—is potentially active in fi-
limited availability of coagulation factors and platelets, brin,121 whereas it is inactivated in collagen matrices by
undiminishing amounts of fibrinogen and platelets are decorin.120 The relatively low platelet concentration in
present at the blood-contacting surface of the prosthesis. wound fibrin makes it likely that TGF-! augments macro-
Therefore, this consecutively increasing inner layer of throm- phage infiltration without inhibiting vessel ingrowth. This
bus obviously differs in both its mode of generation and its can be explained by the concentration dependence of TGF-!,
composition. which is only inhibitory for angiogenesis at high concentra-
tions 108 but chemotactic for macrophages in trace
Outer Fibrin Matrix amounts.122 This early fibrin matrix also facilitates protease
Since tissue ingrowth into prosthetic grafts occurs pri- upregulation and the resulting degradation of the fibrin clot
marily from outside, the outer fibrin clot surrounding the to enable tissue ingrowth. The initial events in this process
grafts serves as a provisional matrix for cell migration from are again regulated through an integrin-ligand mechanism.
the adjacent tissue. However, in order to understand pos- Ingrowing endothelial cells for instance, induce their pro-
sible adverse qualities of the inner fibrin matrix, one needs tease secretion through the binding of #5!1 to fibronectin
to understand the principal interactions occurring between and fibronectin fragments in the fibrin matrix.117,120 The
cells and the more natural wound-type fibrin on the out- relatively low number of platelets contributing to this initial
side of the graft. This blood clot filling the adventitial wound clot also makes it likely that the concentration of platelet-
space consists mainly of crosslinked fibrin, plasma derived thrombospondin (TSP1)—a potent inhibitor of pro-
fibronectin, vitronectin and thrombospondin (TSP),115 con- teases145—does not result in a mitigation of tissue ingrowth.
taining low levels of platelet derived cytokines like TGF-!
and PDGF. This fibronectin-rich fibrin matrix offers bind- Inner Fibrin Matrix
ing sites for integrins of ingrowing cells. For macrophages it Although the mechanism of ingrowth inhibition into
is primarily #5!1,116 whereas it is #v!3 for new blood ves- prosthetic vascular grafts may well be a multifactorial event,
sels.117 The integrin #v!5 has also been implicated in the for- it is conspicuous that the most impenetrable part of the graft
mation of new blood vessels.118 However, whether invasion wall in humans seems to be the inner fibrinous capsule. This
of endothelial cells into a fibrin matrix is #v!5-dependent phenomenon has been reported for more than 20 years in
remains to be established, as the role of this integrin points porous Dacron grafts.43,44,48,59,60,71,72,74,75 We have also ob-
to it interacting selectively with a vitronectin-rich matrix.119 served it in 60 ∝m ePTFE prostheses in the chacma baboon
The interaction between integrins and the matrix also where the impenetrability of the inner fibrin layer even ap-
influences other regulatory mechanisms. For instance, a plies to inflammatory cells. After a few weeks of implanta-
“stressed” fibrin matrix as a result of contraction increases tion, we found the fibrin coverage of the blood surface to be
28 Tissue Engineering of Prosthetic Vascular Grafts
almost completely acellular (Fig. 1.10). It is therefore tempt- affect the structure and/or composition of fibrin in such a
ing to ask whether it is possible to recognize obvious differ- way that it becomes hostile to tissue ingrowth?
ences in the composition or structure of the inner fibrin
capsule which may be responsible for this distinct ingrowth Fibrin Structure
inhibition. In pursuing this question it seems as if the mode It is a relatively recent discovery that fibrin structure
of fibrin generation on the inside of grafts already provides influences cell growth. At lower fiber density a fibrin matrix
significant clues with regard to growth inhibitory proper- strongly stimulates capillary morphogenesis,126 whereas it
ties. First of all, there is a uniqueness about this particular only stimulates endothelial migration without three dimen-
fibrin clot: Both clotting factors and platelets are not con- sional capillary formation at higher fiber density. On an ul-
sumed from a predefined quantity as is the case in a he- trastructural level, a similar inverse correlation between cell
matoma, but rather replenished on an ongoing basis. On growth and fiber density was found.127 The higher the den-
the one hand, a process of platelet-enrichment through sur- sity of fibrin fibers and the lower the average thickness of
face adherence results in an disproportionate density of these fiber bundles, the lower the penetrability for cells.127
thrombocytes which undergo activation through surface Since in vascular grafts cellular ingrowth seems more inhib-
contact and shear stress. As a consequence of this augmented ited in the inner than the outer fibrin matrix, it is therefore
platelet release reaction, high concentrations of #-granule of interest whether inside circumstances favor the genera-
products like platelet factor 3, fibrinogen and von Willebrand tion of a denser fibrin matrix with lower fiber bundle thick-
factor, as well as dense granule contents such as calcium, ness. It was recently demonstrated that an increase in fibrino-
continually sustain the coagulation process.123-125 On the gen concentration by a factor of 3 resulted in a decrease in
other hand, the rapid replenishment of blood plasma next fiber bundle thickness and an increase in fiber bundle den-
to the platelet carpet on the graft surface further guarantees sity, also by a factor of 3.127 Considering the high platelet
undiminishing concentrations of fibrinogen and other clot- density on the blood surface of grafts and the undiminishing
ting factors. Thus, one can assume that the fibrin generated pool of clotting factors, one can assume that the fibrinogen
on the inner surface of vascular prostheses has a high con- concentration inside this surface clot is higher than outside,
tent of fibrinogen. Since TGF-! is also liberated from #-gran- thus resulting in a fibrin matrix of higher fiber density and
ules, and its incorporation into fibrin matrices correlates with lower fiber diameter. The suspicion that higher fibrinogen
its preexisting concentration, this fibrinogen rich fibrin contents inhibit cellular ingrowth was further substantiated
matrix can also be expected to be particularly rich in TGF-!. by other studies using fibroblasts128 and endothelial cells.61,129
Based on these considerations, one needs to ask the initial
question differently: Can high fibrinogen concentrations as Cytokine Content
well as other #-granule release products like TGF-! and The high density of platelets adherent to the blood
thrombospondin and dense granule products like calcium surface, as well as their ongoing activation,5,130 suggest a high
The Lack of Healing in Conventional Vascular Grafts 29
concentration of platelet #-granule contents at the time of incorporation into the LAP-LTBP-TGF-! complex may well
coagulation. Amongst other substances, the #-granules of make it immunohistochemically unrecognizable.
platelets contain platelet derived growth factor (PDGF) and
transformting growth factor-beta (TGF-!).5 PDGF is a Inhibition of Matrix Degradation
dimeric growth factor which is mitogenic for endothelial Apart from directly inhibiting ingrowing endothelial
cells and smooth muscle cells. TGF-!, in contrast, is a po- cells, capillarization and tissue ingrowth into the inner fi-
tent inhibitor of cell proliferation, migration and protein- brin layer may also be indirectly inhibited through a par-
ase production in vascular endothelial cells.131 In view of ticularly high PAI-1 concentration in the matrix. Fibrinoly-
the suspected inhibitory effect of the inner fibrin matrix on sis of the fibrin matrix in normal wound healing is prima-
angiogenesis it is reasonable to hypothesize that particularly rily due to plasmin activation from its precursor molecule,
high levels of TGF-! contribute to the mitigated vessel out- plasminogen. In vivo, this conversion to plasmin is medi-
growth. However, the TGF-! content of human platelet #- ated by tissue plasminogen activator (tPA) and urokinase
granules exists exclusively in the !1 isoform, stored in a la- plasminogen activator (uPA), both of which are derived from
tent form in which the precursor peptide (the latency-asso- endothelial cells, macrophages and giant cells. The action of
ciated peptide LAP) remains noncovalently associated with these proteases is inhibited by PAI-1, secreted by endothe-
the mature 25 kDa TGF-!1 dimer.132 In addition, the latent lial cells and platelets. If one assumes that platelets play an
TGF-! complex from platelets has an additional protein, the disproportionate role in fibrin generation on the inside of
latent TGF-! binding protein (LTB-P) which is linked with the graft, one must also reckon with an disproportionately
a disulphide bond to the LAP.133 Since the activation of high level of platelet-derived PAI-1.139
TGF-! only occurs when the mature 25 kDa dimer is re-
leased from the LAP121 and the entire complex is incorpo- Angiogenesis Inhibitors
rated into the fibrin structure, fibrin degradation is a pre- Another platelet-derived protein which may detrimen-
requisite for liberating inhibitory TGF-! concentrations from tally affect capillarization of the inner fibrin capsule is the
the matrix. Coincidentally, the release of the active 25 kDa extracellular matrix molecule thrombospondin (TSP1).
TGF-! dimer needs the same proteases, such as plasmin, TSP1 is a large, multi-functional ECM glycoprotein that can
which are needed to degrade the fibrin matrix.134 There- influence endothelial cell function in vitro140 and angiogen-
fore, a likely explanation for the late inhibition of esis both in vitro and in vivo.141,142 The role of TSP1 in
capillarization through TGF-! would be the following sce- modulating endothelial cell function has been widely inves-
nario: Initially, endothelial cells from the adventitial region tigated. TSP1 was initially identified and characterized as a
of the vascular prosthesis are attracted into its interstices by protein released from the #-granules of platelets upon acti-
the PDGF and VEGF released by platelets and vation by thrombin. TSP1 is now known to also be produced
polymorphnuclear granulocytes.135,136 Since the fibrin ma- by many other cell types such as endothelial cells, fibroblasts,
trix represents a more or less normal wound matrix at this smooth muscle cells and monocytes/macrophages.
stage, cell receptors involved in initial migration and prolif- Thrombospondin occurs in two forms—soluble and
eration, such as the integrins #v!3 and #v!5, are expressed matrix-bound. The effect of the matrix-bound TSP1 is of-
together with the upregulation of serine proteases (uPA, tPA), ten different from that of the soluble form. Soluble TSP1,
enabling the matrix degradation required for migration. for instance, has been demonstrated to inhibit the prolifera-
Moreover, the interstices of the prostheses contain a mix- tion of endothelial cells143 while matrix-bound TSP1 appears
ture of macrophages and foreign body giant cells (FBGC) to be a permissive substrate for endothelial cell prolifera-
which are generators of large amounts of pro-angiogenic tion. Migratory, invading endothelial cells adhere to in-
cytokines such as bFGF (predominantly under hypoxic con- soluble, matrix-bound TSP1 while soluble TSP1 may satu-
ditions), and only minor amounts of inhibitory TGF-!.56 rate surface receptors and produce an anti-adhesive effect.144
Under those favorable circumstances, capillary ingrowth The anti-adhesive activity of soluble TSP1 may interfere with
slowly proceeds towards the inner fibrin layer with its higher the angiogenic process by preventing the cell-to-substrate
fibrinogen and TGF-! content, as well as its denser fibrin or cell-to-cell interactions necessary for endothelial cell mi-
structure. With increasing TGF-! concentrations as well as gration and capillary formation. Furthermore, recent reports
changing fibrin composition, angiogenesis slows down. The have revealed that TSP1 can also function as a protease in-
increase of TGF-! may be further augmented by macro- hibitor.145 These findings suggest that, apart from its direct
phages, which degrade fibrin matrices through the effect on cells, soluble TSP1 may also influence angiogen-
upregulation of endogenous serine proteases without con- esis by affecting ECM turnover and composition. There is
comitant release of plasminogen activator inhibitor (PAI-1). further evidence that TSP1 may inhibit the proteolytic en-
Therefore, matrix degradation by macrophages is much less zymes of the fibrinolytic pathway, including plasmin and
controlled than degradation by endothelial cells, where a urokinase plasminogen activator.146 The modulation of pro-
balanced, focalized proteolysis of the pericellular matrix tease activity therefore provides a potentially crucial role for
through the up or downregulation of proteases and PAI-1 TSP1 in regulating angiogenesis, as it seems to be a factor in
occurs.137,138 Eventually, both fibrin structure and liberated correcting the balance of protease activity, which is essential
and activated TGF-! amounts may be sufficient to bring in- for angiogenesis.
growing capillaries to a complete halt. A main challenge in Another important interaction of TSP1, particularly
substantiating this scenario will be the detection of high with respect to capillarization of vascular prostheses, is its
TGF-! amounts in the inner fibrous capsule, because the
30 Tissue Engineering of Prosthetic Vascular Grafts
interaction with transformting growth factor-beta (TGF-!). pression of tissue factor, a transmembrane protein.151 This
TSP1 has been shown to bind and activate secreted and factor is capable of initiating the extrinsic pathway of co-
ECM-sequested transformting growth factor-b.147 It follows agulation,152 leading to fibrin generation.153 In vitro studies
therefore that the interaction between soluble TSP1 and have shown that, in particular, the adherence to Dacron ini-
TGF-! may prove to be crucial in inhibiting cell growth and tiates more procoagulant activity than adherence to ePTFE.
proliferation. Even more interesting is the fact that TGF-! This seems to relate to the ability of Dacron to adhere more
and TSP1 are both found in the #-granules of platelets,148,149 monocytes than ePTFE,154 rather than to its surface charac-
increasing their presence in equal proportions in an teristics. Hence, the increased deposition of fibrin onto the
environment characterized by a significant platelet release luminal surface of Dacron grafts over time, as opposed to
reaction. Since thrombospondin released by platelets is not ePTFE, may relate to the initially higher adherence of mono-
incorporated into a secreted extracellular matrix, it would cytes to Dacron. In addition, macrophages are known to se-
provide another explanation for the inhibitory effect of a crete both Il-1! and TNF-# following adherence to synthetic
platelet-rich fibrin matrix, since the anti-proliferative effect materials. These cytokines also enhance tissue factor expres-
of TSP1 is determined by the soluble form of this sion,155 leading to a further induction of PCA and further
secreted protein. fibrin deposition. Since Dacron induces higher levels of se-
The one paradox that remains unresolved is the find- cretion of these cytokines than ePTFE, this may further fa-
ing that macrophages in wounds, as well as in other inflam- cilitate fibrin deposition on the Dacron surface. It would
matory settings, actively produce TSP1. The finding that the appear, therefore, that differences in the ability to mediate
macrophages are releasing a molecule that inhibits angio- monocyte adhesion and activation and so differentially
genesis is at first surprising. However, TSP1 release from upregulate procoagulant activity could partially explain the
macrophages may represent a “direct” control of angiogen- observed differences between the luminal fibrin layer of
esis, instead of an “indirect” control of angiogenesis through Dacron and ePTFE. In addition, tissue factor activity has
the protease activation of latent TGF-!. been associated with membrane vesicles that apparently are
shed concomitantly with procoagulant activity.156,157 Mac-
Fibrinogenic Effect of Macrophages rophages in the depth, therefore, could theoretically influ-
One of the most puzzling questions is why the inner ence fibrin deposition on the surface by shedding vesicles
fibrin capsule of Dacron grafts differs from that of ePTFE containing transmembrane tissue factor. The higher num-
grafts although the blood exposure of the material itself is ber of macrophages present in the depth of Dacron grafts as
only a transient initial event. From the first thin layer of fi- opposed to ePTFE grafts would, therefore, augment
brin which covers the synthetic material, one would expect coagulation.
no further difference between the surfaces with regards to In summary, although fibrin forms a pro-angiogenic
the thrombus formation. This, however, is not the case. If matrix and seems to be an “ideal” scaffold for wound heal-
the exposed surfaces themselves do not differ because of the ing, it may also present its Janus face under different cir-
capping effect of the fibrin, what are the most likely differ- cumstances. It seems likely that the unusual situation of
ences which could be responsible for the qualitative and ongoing surface thrombogenicity within the blood stream
quantitative composition of the inner fibrin capsule? One also produces an unusual fibrin matrix which is rich in fi-
such difference may well be the surface structure. Prior to brinogen and ingrowth inhibitory cytokines.
any cell accumulation, the relatively coarse surface of knit-
ted Dacron is covered by a thrombus of more than Macrophages
100 ∝m,44,60 whereas the fine-structured surface of ePTFE Macrophages dominate the tissue reaction against
lies under a thin fibrin coagulum of only 15 ∝m.1,2 However, prosthetic vascular implants more persistently than any other
the fate of crimped surfaces demonstrates that fibrin tends cell type. The deviation from a normal healing process, how-
to equalize surface unevenness. As a consequence, all graft ever, is not their early presence but the tenacity with which
surfaces will eventually have the same smooth fibrin cover- they reside in the graft structure even after years of implan-
age, irrespective of their structure. If there are no differences tation. Their physiological role in early phases of wound
between the initial surface fibrin, and the material itself is healing is opposed by the perseverance of a chronic inflam-
hidden underneath, the most likely explanation for an on- matory reaction to which they significantly contribute in
going difference in thrombogenicity would be the active par- later stages. Although it is difficult to answer the often re-
ticipation of cellular components in the coagulation pro- curring question of whether macrophages are friends or foes
cess. Since the predominant cell type at the time of the most in prosthetic graft healing, it seems that they are both: friends
significant difference in fibrin buildup between the Dacron at the beginning and foes at the end. Their active secretion
and ePTFE are inflammatory cells, and later on specifically of cytokines may act as a key chemotactic and mitogenic
the macrophages, one needs to answer the question of factor for capillary and connective tissue ingrowth in the
whether macrophages may be key players in the formation early phases of wound healing. Their continual presence,
of the inner fibrin capsule. In this context, it is interesting to however, may increasingly derail the delicate chronology of
note that macrophages are able to express procoagulant ac- cytokines required for the successful accomplishment of
tivities (PCA) through their adherence to surfaces.150 Adhe- healing and also result in material degradation. The diffi-
sion of macrophages to biomaterials causes activation and culty in identifying the key events responsible for this ad-
induction of this procoagulant activity by inducing the ex- verse development lies in the multitude of contributing fac-
The Lack of Healing in Conventional Vascular Grafts 31
tors. On the prosthetic side the different materials, struc- Following material contact, one of the most impor-
tures and microstructures of the graft are all capable of sepa- tant determinants in the chronic inflammatory reaction
rately influencing the foreign body reaction against the im- against synthetic vascular grafts is the biomaterial itself.
plant. From the biological standpoint, distinctly different Adherence, spreading and the secretion of inflammatory
environments characterize the luminal and the outer aspect mediators like TNF- #, 167-171 IL-1 ! 154,163,167,172-177 and
of grafts. IL-6167-169,171 have been extensively investigated on all ma-
The dynamics of macrophage infiltration can best be jor biosynthetics. Dacron, for instance, was shown to lead to
studied on ePTFE grafts, with their fairly even wall struc- a higher density of cells with a morphology indicative of
ture. In the first weeks of implantation one can typically activation than ePTFE,154 as well as to higher TNF-#,178
observe a sandwich structure of macrophage populations IL-6168 and Il-1! secretion.168,179 Not only does Dacron in-
in the graft wall with a relatively distinct accumulation of duce a greater response, but it seems to induce an earlier
Ham 56 and CD68 positive cells at the inside and outside response as well, with the IL-1 peak occurring on day 4 on
surface, separated by a cell-free fibrin matrix in be- Dacron but only on day 7 on all other biomaterials.177 Fur-
tween.13,18,54,28,33 This triple layered infiltration pattern thermore, material differences influence not only the ability
clearly proves that recruitment occurs from both sides. At of macrophages to secrete cytokines but also the expression
the luminal surface the extravasation of blood-borne cells of other markers of activation, for example, MHC-II,180
into the porosity of the material occurs simultaneously with Cd11b181 or TGF-!.29 Expanded PTFE and Dacron also dif-
insudation of blood proteins immediately following implan- ferentially induce alteration of expression of subunits of the
182
tation. At the adventitial surface, however, only a few mono- !2 integrins, which mediate both cell-substratum and cell-
cytes may extravasate during the initial bleeding phase prior cell interactions.
to hemostasis. Subsequently, macrophages have to get there However, there is mounting evidence that these effects
by adhering to capillary endothelial cells and transmigrat- are indirect, rather than directly material-mediated. The dif-
ing through basement membranes via the sequential action ferent abilities of the various materials to adsorb plasma
of a number of adhesion molecules on both the monocyte proteins, in particular, appear to be a main determinant be-
(leukocyte specific !2 integrins)158 and the endothelial cell hind the distinct macrophage reaction against certain
surface (ICAMs).159 biomaterials.183-185 On materials such as polyethylene tereph-
It follows, therefore, that macrophages, irrespective of thalate (PET), polytetrafluoroethylene (PTFE) and polyure-
whether they originate from the blood or from transmigra- thanes, the major blood proteins albumin, fibrinogen and
tion into the adventitial tissue, are faced with a similar task IgG are the predominant proteins adsorbed onto the mate-
to begin with, namely, to infiltrate a provisional fibrin ma- rial surface, with other blood proteins adsorbed to a lesser
trix probably not much different from that found in a nor- extent.183-185 Protein adsorption is not dependent, however,
mal wound. During infiltration, macrophage migration is on relative protein concentrations in the blood,184 but is
facilitated by a low level of basic uPA activity. This should determined primarily by the nature of hydrophobic inter-
also be sufficient to liberate and activate platelet derived actions between the material surface and hydrophobic do-
TGF-! from the fibrin matrix160 which was previously in- mains of the protein. Since most proteins have a net nega-
corporated during hemostasis-related fibrinogenesis.139 tive charge, secondary electrostatic interactions are also im-
Since TGF-! upregulates the expression of integrins on the portant in protein-material interactions and are governed
surface of the blood monocytes thereby promoting adhe- by the chemical nature of the material. The overall combi-
sion and migration,161 it acts as a strong chemotactic agent nation of hydrophobic and electrostatic interactions will have
to recruit macrophages to the inflammatory site.122 More- several effects on the process of protein adsorption. Firstly,
over, it also upregulates the uPA activity of macrophages162 different biomaterials adsorb different types and relative con-
to further facilitate migration. Macrophages are also capable centrations of proteins, with more hydrophobic materials
of binding and internalizing fibrin through the Mac-1 re- adsorbing more protein. It has also been shown that the
ceptor, which augments fibrinolysis by a plasmin-indepen- nature of the adsorbed proteins changes over time with vary-
dent mechanism.163 The degradation products of fibrinoly- ing rates of adsorption versus desorption on different ma-
sis, in return, also act as chemoattractants, recruiting still terials.183 Finally, material-dependent conformational
more macrophages into the implant site.164 In addition, traf- changes as a result of these interactions can be detected by
ficking of monocytes to inflammatory sites also involves the differences in elutability of proteins on different chemical
superfamily of chemoattractant cytokines (chemokines ) and structures.186,187 These changes in conformation differen-
their receptors.165 Specifically, the C-C chemokine MCP-1 tially expose hydrophilic domains to the aqueous environ-
has been demonstrated immunohistochemically to be ment, which may or may not contain sites capable of medi-
expressed by macrophages residing at the implant site as early ating cell adhesion. When comparing two hydrophobic sur-
as 48 h postimplantation. 166 This sequence of events faces of different chemical structure, therefore, although a
illustrates how this initial phase of healing is dominated by nonpolar hydrophobic material may adsorb more protein,
self-augmentation of macrophage chemotaxis. Until the the influence that interactions with polar groups on a chemi-
macrophage actually contacts the material surface, the cally different surface will have on the conformation of these
recruitment into the wound fibrin of the prosthetic graft adsorbed proteins will ultimately determine the differences
does not deviate from that taking place in normal in cellular interaction. This may explain why PTFE which,
wound healing. with a surface tension of 18.5 dynes/cm2,188 is certainly more
32 Tissue Engineering of Prosthetic Vascular Grafts
hydrophobic than Dacron (43 dynes/cm2)188 and should thus mitogenic for hypoxic endothelial cells through the release
adsorb more protein,189,190 mediates less cell adhesion. One of the FGFs. This suggests a paracrine mechanism in which
can imagine that the increased polarity of the carboxyl hypoxia stimulates macrophages to release mitogenic fac-
groups of PET compared to the apolar fluorine groups of tors for hypoxically preconditioned endothelial cells, thereby
PTFE affects the conformation of the adsorbed proteins to promoting angiogenesis. Additionally, TNF-#, which is also
the extent that Dacron binds more macrophages than upregulated in macrophages by hypoxia,198 is known to play
PTFE.154 a role in macrophage mediated angiogenesis, presumably
Of the predominant proteins absorbed, fibrinogen has through the activation and recruitment of further mono-
been shown to be a primary mediator of this adhesion as cytes.199 The role of TNF-# in angiogenesis is, however, con-
conformational changes on adsorption expose the P1 epi- troversial and seems to depend on the biological con-
tope which is recognized by the Mac-1 receptor (Cd11b/cd18, text. 200,201 These effects are further potentiated by
191-194
#m!2) of the macrophage. This receptor also binds to upregulation of MIP-1, a potent macrophage chemo-
adsorbed fragments of the complement component C3 attractant, during oxygen deprivation.198 Therefore, mac-
(C3bi),195 but studies in complement—depleted mice indi- rophages residing in the outer half of synthetic grafts to-
cate that this is not a major mechanism of monocyte adher- gether with VEGF-releasing infiltrating neutrophils136 are
ence.191-193 As we have previously noted, the luminal sur- likely to continue to contribute to a pro-angiogenic milieu
face of a vascular prosthesis is exposed to a practically un- at this stage. The low concentration of TGF-! released with
limited pool of fibrinogen derived from the circulating blood migration-associated degradation of the provisional wound
and released by platelets on activation. The insudation of fibrin should not be inhibitory for endothelial cell prolif-
this fibrinogen into the depth of the graft structure facili- eration at this stage.137
tates macrophage adhesion to the material. Thus, the supe- As the organization of the macrophages within the
rior healing pattern of identical implants in the subcutane- graft wall changes and transmural endothelial cell ingrowth
ous position, with limited exposure to circulating or plate- begins, the situation where the outer wall of the graft is domi-
let fibrinogen, may well be partly explained by this nated by single macrophages also slowly begins to change
phenomenon. by the increasing appearance of foreign body giant
Apart from the material effects on adhesion, the struc- cells.13,19,52,55,64 These may persist for the lifetime of the im-
ture of grafts certainly plays another important role with plant. As macrophage adhesion is determined by material
regard to the macrophage response. Increasing diameters of characteristics, so is macrophage fusion to form giant cells
synthetic fibers, for instance, result in increasing numbers dependent on material surface as well as on structure. It has
of adherent, nonspreading macrophages—a morphology been shown that relatively hydrophilic material surfaces in-
indicative of nonactivated cells.196 This effect may be due to duce more fusion than relatively hydrophobic surfaces.202-206
the associated changes in curvature. In addition to micro- Theoretical analyses show that the time-dependent foreign
structure, the overall porosity of the material will also have body giant cell formation is also influenced by the initial
important implications. Ultimately this will be constrained density of adherent macrophages.203,204 Therefore, any sur-
by two factors: high enough porosity to allow tissue ingrowth, face characteristic which influences the number of adherent
yet low enough to limit continuous insudation of fibrino- cells will also influence the extent of giant cell formation.
gen. It seems reasonable to expect a diminished chronic mac- Most conspicuous is the absence of foreign body giant cells
rophage response in grafts where a temporarily sealing in- (FBGCs) inside the wall structure of ePTFE grafts, compared
growth matrix limits the ongoing insudation of plasma and to Dacron and polyurethane. Although a rim of giant cells
hence limits monocyte adhesion to the material through fi- often demarcates these prostheses against the surrounding
brinogen adsorption. tissue19 and the interstices are sometimes packed with single
As the deposition of fibrin onto the graft surface con- macrophages, no fusion occurs. We have regularly observed
tinues, the inner macrophage zone increasingly becomes this phenomenon in both high porosity and low porosity
wedged between two almost acellular fibrin matrices, one ePTFE in the primate. Although the internodal distances of
in the graft center and one on the blood surface.2-4,7,60,66,72,82 a 60 ∝m ePTFE graft provide both wide localized ingrowth
As discussed under “Fibrin”, the most likely explanation for spaces of approximately 47.94 ± 9.90 ∝m and a significant
this mitigated macrophage recruitment from the blood adhesion area on the stretched fibrils, macrophages prefer a
seems to be the changing physical nature of the fibrin ma- solitary existence under these circumstances. Similarly, the
trix of the inner capsule due to different fiber structure and PTFE nodes—which are the adhesion site for foreign body
concentrations of fibrinogen. In the outer wall of the graft, giant cells on their smooth end on the exterior of the grafts—
however, a macrophage-dominated cell infiltrate continues remain free of giant cells inside the graft wall where numer-
to populate the outer one third to one half of the prosthetic ous fibrils subdivide their surface. The absence of giant cells
structure within 1-3 months.27,28 As the macrophage popu- on the nodal surface subdivided by numerous fibrils indi-
lation migrates into the depth of the graft structure, it must cates that giant cell formation may be suppressed by geo-
eventually overstep the maximum distance of a cell from a metrical space limitations. However, given the in vitro evi-
capillary and thus get into a mild hypoxic state. Macrophages dence that fewer monocytes adhere to ePTFE and it is less
exposed to hypoxia, however, are synthesizing and releasing inflammatory than Dacron,168,178,179,182 and that a more hy-
increased amounts of PDGF as well as aFGF and bFGF.197 drophobic surface decreases the propensity of single mac-
Medium conditioned by hypoxic macrophages in culture is rophages to fuse,202-204,206 the inflammatory potential of the
The Lack of Healing in Conventional Vascular Grafts 33
surface may also play a role in giant cell formation where a capillary sinuses and the presence of smooth muscle cells,
more chemically inert, more hydrophobic ePTFE surface re- seems to indicate that the giant cell contributes to the mis-
duces fusion. directing of the process of transmural capillary ingrowth.
Not only do mononuclear cells gradually fuse to form In situ hybridization of human explants has demonstrated
more giant cells in a material-dependent manner, but fur- that interstitial macrophages and foreign body giant cells
ther complexity is added by the changing nature of the are still secreting Il-1! even after numbers of years.213 Al-
mononuclear macrophage population over time. Macroph- though Il-1! is known to stimulate both smooth muscle cell
ages found in intimate contact with synthetic materials in proliferation and fibronectin deposition in atherosclerotic
situ were ED1 and MHCII positive, whereas ED2 positive plaque formation, 214 it has been suggested that this
macrophages were observed lying behind the ED1 positive interleukin can also inhibit vascular smooth muscle prolif-
cells.169 This indicates a pattern of distribution of not only eration in an autocrine fashion through nitric oxide
mono—versus multinuclear macrophages, but also imma- upregulation.215 The giant cells, therefore, may be respon-
ture (ED1 positive) versus mature (ED2 positive), and acti- sible for not only affecting capillarization but also inhibit-
vated (MHCII positive) versus nonactivated macrophages ing smooth muscle cell ingrowth through persistent Il-1!
within the implant site. Theoretical analysis shows that up secretion. In summary, the true character of the FBGC re-
to 5 weeks of implantation, FBGCs are in fact formed from mains elusive. On the one hand, their persistence at the im-
the fusion of a small percentage of the adherent macroph- plant site and their destructive role in biodegradation, as
ages which are already present on the third day of implanta- well as their possible role in the mitigation of vessel forma-
tion.203,204 This raises the issue of fusogenic potential of dif- tion, seem to label them as villains in incomplete graft heal-
ferent subsets of cells. Expression of cell surface receptors ing. Yet, the evidence supporting a role for a typically anti-
may be a prerequisite. For example, inhibitors of mannose inflammatory cytokine—IL-4—in stimulating giant cell for-
receptor activity have been shown to inhibit IL-4 induced mation is fairly convincing both in vitro and in vivo.99,208
fusion in vitro, suggesting that this receptor may play a role Given that inflammation associated with normal wound
in giant cell formation.207 Elaboration of certain cell surface healing is resolved because of the transient nature of the
markers in a material or environment dependent manner macrophage response, it would appear that the inability to
may be required for a higher propensity for giant cell remove the inflammatory stimulus (a nondegradable bio-
formation. material) confounds even the best intentions of these cells
If it is difficult to decide whether macrophages are ben- to mediate healing. The consequence is a chronic ongoing
eficial for graft healing or not, it is even more difficult to inflammation and the derailing of the normal sequence of
decide whether the presence of foreign body giant cells rep- healing events.
resents a state of pacification or a particularly detrimental In summary, two distinct zones develop in prosthetic
variant of chronic inflammation. One indication for the vascular grafts over time: one close to the blood surface which
FBGCs role as a mediator of pacification of inflammation becomes increasingly uninhabitable for macrophages, and
arises from the fact that their fusion can be induced by one in the outer portion of the graft which is densely packed
IL-4.99,208 IL-4 is secreted by Th2 lymphocytes and is known with macrophages and later on with foreign body giant cells.
to downregulate several macrophage inflammatory func- It is well documented that macrophages infiltrating a wound
tions, for example, the release of certain inflammatory me- environment stimulate the proliferation of endothelial cells
diators,209 the secretion of reactive oxygen intermediates210 and, hence, angiogenesis. However, the apparent lack of cor-
and collagenase production,211 to mention a few. In this con- relation between the number of resident macrophages in the
text it has been proposed that giant cells may represent a interstices of grafts and the extent of transmurally ingrow-
attempt to “mop-up” the deleterious effect of persistent ing vessels indicates that macrophages are not alone in af-
chronic foreign body reaction. In contrast, a detrimental role fecting endothelial ingrowth, but that other factors, for ex-
for the giant cell is suggested by the fact that material sur- ample, the nature of the ingrowth matrix, may affect the
face cracking occurs directly beneath adherent foreign body overall extent of capillarization. Aside from an often impen-
giant cells, implicating the secretory products of these cells etrable fibrin matrix, the protracted ingrowth of connective
in the degradation of biosynthetics.212 The presence of ac- tissue, specifically smooth muscle cells from outside, may
tin-containing adhesive structures202 indicates that the ad- well have to do with ongoing macrophage activation in the
hesion of the giant cell to the biomaterial forms a sealed presence of persistent material stimulus. The secretion of
compartment with a high local concentration of degradative inhibitory cytokines like Il-1! by both macrophages and for-
enzymes. It is also peculiar that the accumulation of giant eign body giant cells may be partly responsible. What is be-
cells in Dacron regularly coincides with the development of coming increasingly obvious is that a macrophage is not al-
huge convolutes of capillary sinuses in the vicinity. These ways a macrophage and that the contribution of each of these
widely dilated and irregularly shaped endothelial sacs are subsets of macrophage populations to the lack of healing of
principally devoid of any second cell layer (Figs. 1.11, 1.18). the vascular graft remains to be investigated. Overall, the
In contrast, 60 ∝m ePTFE, with its scanty presence of for- macrophage remains the Jekyll and Hyde of graft healing,
eign body giant cells, shows arterioles in its outer interstitial but as our insight into the mechanisms of healing grows so
spaces with one or more layers of smooth muscle cells. This does our understanding of which critical questions remain
histological observation, where the lack of giant cells corre- to be answered in the elucidation of the role of this
lates with the absence of malformed vessels in the form of enigmatous cell, namely:
34 Tissue Engineering of Prosthetic Vascular Grafts
1. If monocyte adhesion to biomaterials is prevented Apart from the favorable matrix environment, white blood
completely, do we compromise healing in any way; cells provide various pro-angiogenic stimuli at this stage.
2. To what extent does engineering tissue ingrowth Infiltrating neutrophils are a source of VEGF,136 and mac-
modulate the deleterious effect of the inflammatory rophages migrating into a presumably mildly hypoxic envi-
cell by promoting selective cellular infiltration be- ronment secrete acidic and basic FGF as well as PDGF.197
fore the development of chronic inflammation; and These cytokines are both chemotactic and mitogenic for
3. Can we identify secretory products of macrophages endothelial cells. Once again, integrin mediated binding to
or giant cells which specifically mediate the mitiga- the wound matrix is central to these responses, with both
tion of tissue ingrowth, either endothelial sprouts #v!3 and #v!5 involved in growth factor-stimulated angio-
or smooth muscle cells? genesis, although via different mechanisms:59 bFGF-induced
angiogenesis is mediated by the #v!3 integrin, whereas #v!5
Prosthetic Wall Vascularization integrin is crucial in VEGF-stimulated angiogenesis.
The “restitutio ad integrum” healing of a vascular pros- Although the initial phase of vascularization resembles
thesis—the formation of a neo-artery in place of a removed normal wound healing, the subsequent phase of centripetal
segment—would necessitate the formation of vasa vasorum vessel ingrowth through the interstices of the graft is already
in the graft wall, accompanied by the growth of a fully func- substantially slowed and often only reaches the outer half of
tional medium in an inflammation-free environment. The the graft wall.60,78 Considering the fact that capillarization
second best solution—the formation of granulation tissue of several millimeters of wound fibrin is normally accom-
with the resulting conclusion of the healing process through plished in humans within days, the time period of several
scar tissue—would at one stage also require extensive months required for achieving this ingrowth can only be
capillarization. Even if conventional synthetic grafts are lack- explained by a significant deviation from normal wound
ing the sophisticated biological information necessary for healing. One obvious difference between normal and graft
“ad integrum” healing, their porosity would in many cases healing is the nature of the chronic inflammatory response.
at least allow the ingrowth of granulation tissue. From our Specifically, the macrophage-derived giant cells which gradu-
daily surgical practice we are all familiar with the alacrity ally build up in the outer parts of the graft27,28,33,60,62 are
with which the formation of such granulation tissue and only formed in response to a foreign body. In high porosity
the subsequent stabilization through scar tissue is usually Dacron grafts, this foreign body reaction is not only associ-
accomplished. The tissue gap in technically the best of all ated with mitigated ingrowth but also the formation of
surgical wound closures is often more than one millimeter hugely dilated capillary sacs as opposed to regular vessels
wide, but capillarization is not only complete, but scar tis- (Figs. 1.11, 1.18). Moreover, in some models, the conspicu-
sue formation is already beginning, as early as after seven ous lack of smooth muscle cells, and therefore of vessels other
days. It is therefore surprising that an equally short distance than capillaries, indicates that not only angiogenesis but also
in the case of the wall of prosthetic vascular grafts does not musculogenesis is inhibited. Given these unique circum-
accomplish the same degree of full thickness capillarization stances, it should be possible to describe biological interac-
even after years of implantation. In order to further eluci- tions, which at this stage of graft healing, may be respon-
date this major shortcoming in graft healing. one needs to sible for the gradual mitigation of transmural ingrowth. His-
ask two key questions: tological analysis suggests that probably both matrix- and
1. Why does adventitial capillary sprouting into the cell-mediated phenomena are responsible for the inhibition
outer fibrinous capsule and later on into the inter- of angio- and musculogenesis.
stices of the prosthetic graft occur at such a slow pace As far as the matrix is concerned, we have already dis-
and eventually come to a halt; and cussed the possibility of a gradual transition in the nature
2. Why is anastomotic endothelial cell outgrowth so of the fibrin from the outer wound to a fibrinogen-rich blood
futile concerning both surface endothelialization and surface which may gradually slow down the capillarization
capillary sprouting into the depth? from outside to inside. In this regard, it is interesting that
The initial angiogenic response of the surrounding studies on matrix control of angiogenesis217,218 have ob-
tissue to a freshly implanted vascular prosthesis does not served that it is specifically capillary morphogenesis that
seem to differ from any other wound repair. This early phase depends on extracellular matrix (ECM).219 Especially, the
is typically characterized by infiltration into the wound fi- ability of endothelial cells to exert mechanical forces on the
brin of a relatively high proportion of polymorphonuclear surrounding matrix,217 and the subsequent development of
granulocytes, but also lymphocytes and a few macrophages. matrix networks218 to guide capillary sprout formation, are
Capillaries within the adventitia sprout into the freshly matrix determined. Physical and/or biochemical aspects of
formed fibrin matrix within days of injury.2,13,48,62,63,76,94 fibrin formation will, therefore, affect the malleability of the
During this formation of early granulation tissue, the ma- matrix and hence the susceptibility to cell mediated trac-
trix is probably inconspicuous with regard to fibrinogen tion and the realignment of matrix fibers. Hence, the sig-
concentration, fiber constitution and cytokine content and, nificance of the qualitative change in the matrix towards the
as such, is naturally pro-angiogenic. The sprouting endot- lumen may be inhibition of capillary ingrowth.
helial cells bind via #v!3 to the RGD site of the fibrin #-chain In addition to matrix mediated effects, two histologi-
and migrate into the pores of the graft. This also promotes cal observations hint at cellular events playing a role in the
survival of the angiogenic sprout by sustaining an anti- mitigation of vascularization. Firstly, there is the fact that
apoptotic response through inhibition of p53 activation.216 capillary sprouting increasingly comes to a halt towards the
The Lack of Healing in Conventional Vascular Grafts 35
acellular inner fibrinous capsule, while the macrophage- boon, for instance, capillary outgrowth is rapid and thus
dominated chronic inflammatory tissue, which is limited to completed before a changing environment can inhibit
the outer half of the graft, reaches a state of sufficient vascu- it.30,32,34 Under senescent circumstances as in humans78 and
larization.2,11,27-29,60 It would appear that, although hypoxic the chacma baboon, however, the protracted angiogenic
macrophages may initially be responsible for a pro-angio- potential of endothelial cells does not allow completion of
genic stimulus, the vascularization which has taken place in capillary ingrowth prior to the change in the biological en-
the meantime in the outer half of the graft re-establishes a vironment. This seems to explain why the same graft leads
normoxic environment. With an increasing oxygen tension to rapid spontaneous endothelialization in the one model
in the tissue, one would expect a downregulation of the se- but disproportionately fails to achieve this goal in another.
cretion of certain endothelial cell mitogens220,221 and hence While unfavorable matrix characteristics, continual
a downregulation of the angiogenesis. Moreover, the short- macrophage presence and host specific endothelial cell char-
lived neutrophil response may further deprive angiogenesis acteristics offer a possible explanation for the slowing down
of a potent stimulator—VEGF.56,136,222 of capillary ingrowth, the lack of musculogenesis within the
The second and perhaps most unique observation is graft wall is more challenging to explain. With potent tools
not that angiogenesis gradually slows down, but the fact that of tissue engineering available, one needs to overcome the
vessel formation is so derailed that the endothelial cells form deep seated suspicion of the presence of smooth muscle cells
sinuses as opposed to tubes. It seems likely that this sinus based on their role in intimal hyperplasia. This suspicion
formation may be due to the lack of appropriate intercellu- prevails despite reports that smooth muscle cell prolifera-
lar alignment and adhesion events, which are as critical to tion reaches an equilibrium on prosthetic vascular grafts,61,93
the process of angiogenesis as the migration and prolifera- depending partly on functional interactions with physiologi-
tion of endothelial cells.218 One hint of what the mecha- cal regulators like blood flow.54 However, the goal of even-
nism may be comes from an in vitro model of bFGF-in- tually emulating functional arteries through “smart” engi-
duced angiogenesis. It was observed that, in fibrin gels, stimu- neered scaffolds will necessarily need to assign the signifi-
lation of endothelial cells with bFGF alone resulted in large cant role of creating a “healthy” neo-media to smooth muscle
lumen sinuses similar to those seen in grafts, whereas the cells. It is therefore paramount to try to understand mecha-
addition of TGF-! at picogram concentrations reduced the nisms of ingrowth inhibitions for smooth muscle cells also
lumen dimensions.223 Addition of nanograms of TGF-! miti- and not only for endothelial cells. Therefore, one must see
gated lumen formation completely. Given these in vitro ob- two peculiar observations in prosthetic midgraft healing in
servations, it would appear that ultimately the 3-dimensional this context: the absence of smooth muscle cells in most parts
organization of capillary sprouts depends on a balance of of the graft wall2,16,26,27,94,224 and at the same time the pres-
proliferating versus differentiating stimuli, where an over- ence of smooth muscle cells underneath luminal endothe-
riding mitogenic stimulus (in this model, bFGF) results in lial cells,2,14,30-33,35,59,67,77,81 sometimes separated by acellu-
large sinuses but increasing concentrations of a differenti- lar fibrin59 from transmurally ingrowing connective tissue.
ating stimulus (TGF-!) arrests proliferation and so facili- With transdifferentiation of fibroblasts being only a
tates tube formation. Driven to the other extreme, an over- remote possibility, the presence of smooth muscle cells seems
riding differentiating stimulus arrests both proliferation and to be dependent on the differentiation of mural/mesenchy-
tube formation. If one concludes that the sinuses are the mal cells in the wake of angiogenesis.106,107 Embryonic data
result of overriding mitogenic signals in the outer zone of suggests that endothelial tube structures form first and di-
the graft wall, where could these signals originate? As previ- rect the subsequent recruitment of the surrounding cells—
ously mentioned, histologically we observe that sinus for- pericytes in small vessels and smooth muscle cells (SMC) in
mation is associated with the gradual buildup of giant cells. larger vessels. This promotion of mural cell proliferation and
The obvious association of these cells with the capillary si- migration is primarily achieved through the synthesis and
nuses indicates that they may be secreting an abundance of secretion of platelet derived growth factor-BB (PDGF-BB)
endothelial cell mitogens. Although macrophages themselves by endothelial cells, which upregulates smooth muscle cell
are known to stimulate endothelial proliferation,197,220 the expression of collagenase, #v!3 and PDGF-receptors. The
limited information available on the nature of secretory upregulation of these factors allows the mural cells to move
products of the foreign body giant cell makes it difficult to toward the endothelial cell, eventually establishing intercel-
define exactly what paracrine mechanism may be involved. lular contact through adherens junctions. This intimate con-
To further complicate the situation, the success of graft tact between the two cell types drives the expression and
wall vascularization appears to depend not only on the in- activation of TGF-!, resulting in a local high concentration
teraction of these pro- and anti-angiogenic influences of sufficient to inhibit endothelial cell migration and prolif-
inflammatory cells and matrix, but also on the interplay of eration225 as well as to promote differentiation of the
these two factors with the angiogenic vigor of host endo- pericytes/smooth muscle cells. This delicate cytokine bal-
thelial cells. The example of 60 ∝m ePTFE grafts in different ance between endothelial cells and smooth muscle cells re-
animal models illustrates best that there seems to be a net flects the interdependence of these two cell types, further
effect between the endothelial cell’s inherent ability to rap- emphasizing that smooth muscle cells require the presence
idly proliferate and migrate on the one hand and the gradual of endothelial cells for migrating into tissue.
build up of an environment which inhibits angiogenesis on If, on the one hand, capillaries eventually reach the
the other. In the juvenile animal model of the yellow ba- inner fibrinous capsule of prosthetic grafts and, on the other
36 Tissue Engineering of Prosthetic Vascular Grafts
hand, mesenchymal cell differentiation into smooth muscle a species high and the porosity favorable to allow ingrowth
cells normally “follows” the outgrowing endothelial sprouts, together with early sealing of the interstices against continual
why are capillaries often found in abundance but without fibrin/fibrinogen insudation, capillaries may reach across the
accompanying smooth muscle cells? Similarly to the inhibi- entire distance before the biological environment changes.
tion of capillarization, both cell-matrix and cell-cell inter- As a consequence, no premature maturation event would
actions seem to play a role in the inhibition of mus- prevent the endothelial cells from chemotactically attract-
culogenesis. As discussed under “Fibrin”, platelet activation ing mural cells to trail them. This scenario seems to explain
at the blood surface may result in relatively high TGF-! con- best why 60 ∝m ePTFE grafts in the juvenile yellow baboon
centrations in the nonfavorable fibrin matrix. Should show such a distinctly different healing pattern with regard
migration-associated degradation liberate this TGF-!, it may to smooth muscle cell ingrowth compared to other grafts
indeed prematurely inhibit proliferative responses of smooth and species. However, if the fibrinogenic potential of a graft
muscle cells and switch the phenotype of these cells to a rest- is relatively high and the angiogenic potential of the host
ing one. In this way, the biological signals encoding the matu- endothelial cell low, it is reasonable to imagine that the slowly
ration and eventually the arrest of a proliferative event may changing biological environment allows the first part of a
be prematurely transmitted to the smooth muscle cells as a chronological sequence but prevents the second one. As the
result of the changing fibrin environment over time. How- circumstances deteriorate to a point where the entire pro-
ever, as tempting as TGF-! may be as an explanation for the cess begins to halt, the first step—in this case, capillary
mitigation of further capillary ingrowth into the inner fibrin- sprouting—has already materialized while the subsequent
ous capsule, it does not explain the absence of smooth muscle step—the site-directed migration of mural cells—has not
cells in areas of dilated endothelial sinuses towards the ad- even commenced yet.
ventitial surface where the fibrin is assumed to be closer to When both endothelial cells and smooth muscle cells
normal wound fibrin both in structure and cytokine con- do reach the blood surface, additional large vessel signals
tent. Because these sinuses are associated with macrophages appear to play a role in their relationship. Flow conditions
and giant cell formation, one explanation may be the im- and compliance mismatch may result in an overriding stimu-
munohistochemical observation that macrophages and gi- lus for smooth muscle cell proliferation229 despite their di-
ant cells are secreting Il-1! even after years of implanta- rect contact with endothelial cells, which would normally
tion.213 This cytokine has been show to upregulate smooth terminate smooth muscle cell mitogenesis through the ef-
muscle cell nitric oxide production215 and could, as such, fect of TGF-!. It is well established that the endothelium
negatively regulate smooth muscle cell mitogenesis in areas transduces flow-related signals and directs smooth muscle
of macrophage infiltration, in contrast to its role in the cell proliferation when required. It is not surprising, there-
pathogenesis of atherosclerosis.214 More likely, however, is fore, that expression of the smooth muscle cell mitogen
an absence of PDGF-BB secretion by the endothelial cells, PDGF-AA has been detected in the endothelial cells of the
because they never reach maturation associated with vessel well developed intima of grafts implanted in the yellow ba-
formation. boon,52,226 while very little PDGF-B chain is detected. Con-
In addition, we have already discussed the role of a sidering the complexity of, for example, PDGF signaling
slowly changing matrix on the mitigation of full thickness where PDGF is a homo- or heterodimer of two different
wall capillarization. Such arrested endothelial cells may cease chains with three resulting isoforms, not to mention two
to send out chemotactic signals to smooth muscle cells in different receptors selectively binding all or only one of these
form of PDGF. Interestingly, although ingrowing capillaries isoforms, it is possible that these factors may combine to
in high porosity Dacron grafts were completely devoid of generate a wide variety of results and afford the cell a rela-
smooth muscle cells in the chacma baboon model, 60 ∝m tively tight control of the outcome.227,229 This would explain
ePTFE grafts in chacma and yellow baboons showed arteri- why PDGF-AA is present during intimal proliferation al-
oles with layers of actin positive smooth muscle cells though PDGF-BB normally directs smooth muscle cell mi-
(Fig. 1.9). This indicates that in those grafts in which gration and proliferation during angiogenesis.
capillarization was able to overcome ingrowth inhibition at If premature maturation signals are the reason for the
an early stage and proliferation of the endothelial cells was stoppage of transmural ingrowth of capillaries at the inside
not arrested, the smooth muscle cell component is normally of the inner fibrinous capsule, is it possible to apply the same
developed.2,30-33,35,46,77,81 This supports the hypothesis that interpretation to the ingrowth stoppage of anastomotic sur-
mitigation of capillarization influences smooth muscle cell face endothelium? At first thought, events only partially re-
migration. Moreover, it is noticeable that the combination semble those occurring in the graft wall. Since angiogenesis
of high porosity ePTFE with a juvenile and vigorously is a complex process including matrix degradation, migra-
endothelializing animal model achieves the most differenti- tion and proliferation, the earlier termination of endothe-
ated layers of smooth muscle cells.30-33,35 These smooth lial outgrowth in the wall matrix may be due to the inhibi-
muscle cells are observed in immediate proximity to endo- tion of either of these three. Since surface migration does
thelial cells within the initial 2-3 weeks of implantation when not require matrix degradation to the same extent, it must
biological circumstances seem to be more in favor of angio- be either proliferation or migration which is primarily af-
genesis, and hence smooth muscle cell recruitment, than at fected under these circumstances. Extrapolation of the num-
a later stage. Therefore, if the fibrinogenic potential of a graft ber of population doublings230 required to cover the trans-
is relatively low, the angiogenic vigor of endothelial cells of anastomotic ingrowth area clarifies that it is not the divid-
The Lack of Healing in Conventional Vascular Grafts 37
ing potential of the endothelial cell which is the limiting fac- widely held opinion, it would appear that in fact
tor of endothelial outgrowth. Therefore, inhibitory factors transanastomotic endothelialization progressively slows
other than an exhausted mitotic capacity must play a role. down in all models, although at different rates. One can
One explanation could be activated platelets that accumu- therefore not extrapolate short term observations to a long
late in and on the pseudointimal layer throughout the length term situation. Since the cessation of tissue outgrowth can-
of the graft. The complex signaling arising from platelet de- not be due to an exhausted mitotic potential of endothelial
granulation and the high local TGF-! concentration which cells, migration and proliferation appear to get actively ar-
results could affect endothelial cell proliferation at the sur- rested by changing biological circumstances. The fibrino-
face. However, there must be an additional explanation for gen and platelet-rich environment at the blood surface makes
transanastomotic ingrowth stoppage, because it is also seen the buildup of a dense and fine fibrillar fibrin capsule likely,
in grafts with minimal surface fibrin or platelet coverage. containing unusually high concentrations of TGF-!. Such a
The possible involvement of an additional factor becomes matrix seems theoretically capable of prematurely turning
apparent in Dacron grafts, where the outgrowth of endo- the outgrowing endothelium into a noncycling, resting inti-
thelium is more pronounced in a high porosity (knitted) mal tissue.
Dacron40,46,47,60,68,81,107 than in a low porosity (woven) Porosity has always been regarded as a crucial deter-
one.40,43 The difference between the two types of Dacron minant for tissue ingrowth. However, porosity alone is not
grafts obviously lies not in the composition of the fibrinous sufficient to quantify the available space for ingrowth. Even
pseudointima but in the ingrown tissue underlying this fi- in so-called high porosity ePTFE grafts for instance, full-
brin matrix. Could this ingrowing tissue in some way be thickness transmural ingrowth of microarterioles is only pos-
facilitating endothelial ingrowth from the anastomosis so sible in a marginal percentage of interstitial spaces. The graft
that the gradual cessation of transmural ingrowth becomes wall structure may provide the simplest level of inhibition,
reflected in a similar mitigation of outgrowth from the anas- and therefore this factor needs to be considered. The dimen-
tomoses? Paracrine signaling over such distances is known sions of the available ingrowth space in ePTFE grafts have
to occur during embryogenesis, so it is not beyond prob- not previously been assessed in order to determine the di-
ability that ingrowing tissue directs transanastomotic out- mensions of a transmural channel. The mathematical cal-
growth but the underlying mechanism has yet to be culations presented for these grafts in this chapter reveal that
determined. porosity was previously overestimated as a sole determinant
In summary, the most striking shortcomings of pros- for tissue ingrowth. Similarly, the measurements of interfi-
thetic wall vascularization, namely, the failure of the capil- ber spaces in Dacron grafts revealed a maximum space suf-
laries to reach the luminal surface as well as the absence of ficient for capillaries, and limited access to arteriolar in-
mature smooth muscle cells and hence arterioles, could be growth.
partly attributed to both the surrounding matrix and the On the biological level, a continual buildup of seem-
surrounding inflammatory cells. Graft healing therefore ingly adverse conditions eventually prevents the completion
seems to be a race between the endothelial cell’s angiogenic of transmural tissue ingrowth in most of the cases. In those
potential and the buildup of inhibitory influences. Since the instances in which microvessels successfully reach the blood
endothelial cell remains a relative constant in human im- surface, it appears that tissue ingrowth is completed more
plants it makes sense that graft designs should aim at, firstly, rapidly and hence before the environment turns inhibitory.
preventing the deposition of unfavorable, compacted fibrin When identifying inhibitory structures, the compact inner
and, secondly, attenuating the chronic inflammatory re- fibrin capsule and the outer zone, with massive accumula-
sponse. This could well be achieved by incorporating an in- tion of foreign body giant cells, appear to be the primary
growth matrix which not only temporarily seals the poros- culprits. The special composition of the internal fibrinous
ity of the graft wall to protein insudation but at the same capsule provides a reasonable explanation for the fact that it
time selectively promotes vascular cell infiltration while in- is almost impenetrable for capillaries. In contrast, the outer
hibiting the buildup of a prolonged macrophage response. graft zone which is packed with foreign body giant cells seems
rather to affect the quality of angiogenesis by derailing the
Conclusion delicate process of endothelial—and smooth muscle cell—
Healing of prosthetic vascular grafts implies transmu- sprouting. While unrivaled endothelial proliferation leads
ral tissue ingrowth. During the past three decades, however, to the formation of hugely dilated capillary sacs as opposed
the focus of research was almost entirely on transanastomotic to vessels, no smooth muscle cells follow the angiogenic ef-
rather than transmural ingrowth. In spite of this one-sided forts of the endothelial cells. This lack of musculogenesis
approach, an analytic effort combining historical data and occurring in the graft wall corresponds conspicuously with
personal experience with biological principles helped to the presence of foreign body giant cells. The presence of gi-
clarify some main issues involved in the mitigation of the ant cells is in turn a consequence of a pronounced macroph-
entire healing process of contemporary arterial prostheses. age attraction by the synthetic materials. This macrophage
The complete surface endothelialization through infiltration is augmented by fibrinogen insudation from the
transanastomotic ingrowth in animal models under shorter blood stream.
term experimental conditions often suggested that these If one tries to draw any conclusions from these obser-
models differ in principle from the human situation with vations for the tissue engineering of future grafts the fol-
its stoppage of endothelial ingrowth. In contrast to this lowing main guidelines appear to be crucial:
38 Tissue Engineering of Prosthetic Vascular Grafts
• In order to design future experiments with a view to- from sodden expanded polytetrafluorethylene grafts. J
wards transmural rather than transanastomotic Vasc Surg 1997; 25:187-197.
ingrowth, minimal graft lengths of 15 cm should be 11. Sottiurai VS, Yao JST, Flinn WR, Batson RC. Intimal hy-
observed; perplasia and neointima: An ultrastructural analysis of
thrombosed grafts in humans. Surgery 1983; 93: 809-817.
• Any structural design of a graft scaffold should make
12. Stumb MM, Jordan GL, DeBakey ME. Endothelium
provision for continual transmural ingrowth spaces growth from circulating blood on isolated intravascular
capable of accommodating at least arterioles in order Dacron hub. Amer J Path 1963; 43:361-368.
to theoretically allow the ingrowth of both endothe- 13. van der Lei B, Wildevuur RH. Microvascular polytetra-
lial cells and smooth muscle cells; fluorethylene prostheses: The cellular events of healing and
• Considering that tissue ingrowth faces an increasingly prostacyclin production. Plastic and Reconstructive Sur-
adverse microenvironment, a key issue to successful gery 1988; 81:735-741.
healing will be to facilitate rapid completion of vas- 14. Bartels HL, van der Lei B, Robinson PH. Prosthetic
cularization; microvenous grafting in the rat femoral vein. Laboratory
• Since fibrinogen immobilization to the biomaterial is Animals 1993; 27:47-54.
a main mediator of macrophage recruitment, a tem- 15. Guidoin RG, King M, Marois M, Martin L, Marceau D.
New polyester arterial prostheses from Great Britain: An
porarily sealing ingrowth matrix may prevent this
in vitro and in vivo evaluation. Ann Biom Engin 1986;
event and thus reduce the macrophage presence. 14:351-367.
In summary, we eventually begin to understand key 16. Stronck JWS, van der Lei B, Wildevuur CRH. Improved
aspects of the aborted healing events in yesterday’s prosthe- healing of small-caliber polytetrafluoroethylene vascular
ses. This does not only prepare us better for the de novo prostheses by increased hydrophilicity and by enlarged
design of tomorrow’s tissue engineered grafts, but also helps fibril length. An experimental study in rats. J Thorac
us to change the angle from which we approach graft de- Cardiovasc Surg 1992; 103:146-152.
sign: The main emphasis will need to be on the delicate 17. Van Der Lei B, Stronck JW, Wildevuur RH. Enhanced
manipulation of biological chronologies in the context of healing of 30 ∝m Gore-tex PTFE microarterial prostheses
an accurately modeled synthetic scaffold designed to limit by alcohol-pretreatment. British Journal of Plastic Surgery
1991; 44:428-433.
inflammatory responses to material contact.
18. Bellón JM, Buján J, Contreras LA, Jurado F. Similarity in
behaviour of polytetrafluorethylene (ePTFE) prostheses
References implanted into different interfaces. J Biomed Mat Res
1. Graham LM, Burkel W E, Ford JW, Vinter DW, Kahn 1996; 31:1-9.
RH, Stanley JC. Expanded polytetrafluoroethylene vascu- 19. Clowes AW, Gown AM, Hanson SR, Reidy MA. Mecha-
lar prostheses seeded with enzymatically derived and cul- nisms of arterial graft failure: I. Role of cellular prolifera-
tured canine endothelial cells. Surgery 1982; 91:550-559. tion in early healing of PTFE prostheses. Amer J of Path
2. Herring MB, Baughman S, Glover J, Kesler K, Jesseph J, 1985; 118:43-54.
Campbell J, Dilley R, Evan A, Gardner A. Endothelial 20. Florian A, Cohn LH, Dammin GJ, Collins JJ. Small vessel
seeding of dacron and polytetrafluoroethylene grafts: The replacement with Gore-tex (expanded polytetra-
cellular events of healing. Surgery 1984; 96:745-754. fluoroethylene). Arch Surg 1976; 111:267-270.
3. Boyd KL, Schmidt SP, Pippert TR, Sharp WV. Endothe- 21. Hanel KC, McCabe C, Abbott WM, Fallon J, Megerman
lial cell seeding of ULTI carbon-coated small diameter J. A biomechanical, scanning electron, and light micro-
PTFE vascular grafts. ASAIO Transactions 1987; scopic evaluation Ann Surg 1982; 195:456-463.
33:631-635. 22. Mathisen SR, Wu H, Sauvage LR, Usui Y, Walker MW.
4. Kao WJ, McNally AK, Hiltner A, Anderson JM. Role for An experimental study of eight current arterial prosthe-
IL-4 in FBGC formation on a poly(etherurethane urea) ses. J Vasc Surg 1986; 4:33-41.
in vivo. J Biomat Res 1995; 29:1267-1275. 23. Sterpetti AV, Lepidi S, Cucina A, Patrizi AL, Palumbo R,
5. Zilla P, Preiss P, Groscurth P, Rösemeier F, Deutsch M, Taranta A, Stipa F, Cavallaro A, Santoro-D’Angelo L, Stipa
Odell J, Heidinger C, Fasol R, von Oppell U. In vitro- S. Growth factor production after polytetrafluoroethylene
lined endothelium: Initial integrity and ultrastructural and vein arterial grafting: An experimental study. J Vasc
events. Surgery 1994; 116:524-534. Surg 1996; 23:453-460.
6. Friedman EW, Hamilton AJ. Polytetrafluorethylene grafts 24. Campbell CD, Goldfarb D, Roe R. A small arterial sub-
in the peripheral venous circulation of rabbits. The Ameri- stitute: Expanded microporous polytetrafluoroethylene:
can J Surg 1983; 146:355-359. Patency versus porosity. Ann Surg 1975; 182:138-143.
7. Zamora JL, Navarro LT, Ives CL, Weilbaecher DG, Gao 25. Kenney DA, Tu R, Peterson RC. Evaluation of compliant
ZR, Noon GP. Seeding of arteriovenous prostheses with and noncompliant PTFE vascular prostheses. ASAIO
homologous endothelium. J Vasc Surg 1986; 3:860-866. Transactions 1988; 34:661-663.
8. Binns RL, Ku DN, Stewart MT, Ansley JP, Coyle KA. 26. Matsumoto H, Hasegawa T, Fuse MD, Yamamoto M,
Optimal graft diameter: Effect of wall shear stress on vas- Saigusa M. A new vascular prosthesis for a small caliber
cular healing. J Vasc Surg 1989;10:326-337. artery. Surgery 1973; 74:519-523.
9. Golden MA, Hanson SR, Kirkman TR, Schneider PA, 27. Douville EC, Kempczinski RF, Birinyi LK, Ramalanjaona
Clowes AW. Healing of polytetrafluoroethylene arterial GR. Impact of endothelial cell seeding on long-term pa-
grafts is influenced by graft porosity. Journal of Vascular tency and subendothelial proliferation in a small-caliber
Surgery 1990;11:838-845. highly porous polytetrafluoroethylene graft. J Vasc Surg
10. Lewis DA, Lowell RC, Cambria RA, Roche PC, Gloviczki 1987; 5:544-550.
P, Miller VM. Production of endothelium-derived factors
The Lack of Healing in Conventional Vascular Grafts 39
28. Bull DA, Hunter GC, Holubec H, Aguirre ML, Rappaport 46. Noishiki Y. Pattern of arrangement of smooth muscle cells
WD, Putnam CW. Cellular origin and rate of endothelial in neointimae of synthetic vascular prostheses. J Thor and
cell coverage of PTFE grafts. J Surg Res 1995; 58:58-68. Cardiovasc Surg 1978; 75:894-901.
29. Greisler HP, Petsikas D, Cziperle DJ, Murchan PM, 47. Szilagyi DE, Smith RF, Elliott JP, Allen HM. Long-term
Henderson SC, Lam TM. Dacron stimulation of macro- behaviour of a dacron arterial substitute: Clinical roent-
phage transformting growth factor-beta release. Cardio- genologic and histologic correlations. Ann Surg 1965;
vascular Surgery 1996; 4: 169-173. 162:453-475.
30. Clowes AW, Kirkman TR, Clowes MM. Mechanisms of 48. Wesolowski SA, Fries CC, Gennigar G, Fox LM, Sawyer
arterial graft failure. II. Chronic endothelial and smooth PN, Sauvagae LR. Factors contributing to long-term fail-
muscle cell proliferation in healing polytetrafluorethylene ures in human vascular prosthetic grafts. Cardiovas Surg
prostheses. J Vasc Surg 1986; 3:877-884. 1964; 38:544-567.
31. Kohler TR, Kirkman TR, Kraiss LW, Zierler BK, Clowes, 49. Wesolowski SA, Fries CC, Karlson K E, De Bakey M, Saw-
AW. Increased blood flow inhibits neointimal hyperpla- yer PN. Porosity: Primary determinant of ultimate fate of
sia in endothelialized vascular grafts. Circulation Research synthetic vascular grafts. Surgery 1961; 50:91-96.
1991; 69:1557-1565. 50. Martinet Y, Bitterman PB, Morne JF, Grotendorst GR,
32. Clowes AW, Zacharias RK, Kirkman TR. Early endothe- Martin GR, Crystal RG. Activated human monocytes ex-
lial coverage of synthetic arterial grafts: Porosity revisited. press the c-sis proto-oncogene and release a mediator
The Amer J Surg 1987; 153:501-504. showing PDGF-like activity. Nature (Lond) 1986;
33. Golden MA, Au YPT, Kirkman TR, Wilcox JN, Raines 319:158-160.
EW, Ross R, Clowes AW. Platelet-derived growth factor 51. Shimokado K, Raines EW, Madtes DK, Barrett TB, Benditt
activity and mRNA expression in healing vascular grafts EP, Ross R. A significant part of macrophage-derived
in baboons. J Clin Invest 1991; 87:406-414. growth factor consists of at least two forms of PDGF. Cell
34. Lado MD, Knighton DR, Cavallini M, Fiegel,VD, Murray 1985; 43:277-286.
C, Phillips GD. Induction of neointima formation by 52. Kraiss LW, Geary RL, Mattson, EJR, Vergel S, Au YPT,
plataelet derived angiogenesis fraction in a small diam- Clowes AW. Acute reductions in blood flow and shear
eter, wide pore, PTFE graft. Int J Artif Organs 1992; stress induce platelet-derived growth factor—A expression
15:727-736. in baboon prosthetic grafts Circ Res 1996; 79:45-53.
35. Zacharias RK, Kirkman TR, Clowes AW. Mechanisms of 53. Zerwes H, Risau W. Polarized secretion of a platelet-de-
healing in synthetic grafts. J Vasc Surg 1987; 6:429-436. rived growth factor-like chemotactic factor by endothe-
36. Golden MA, Au YPT, Kenagy RD, Clowes AW. Growth lial cells in vitro. J Cell Biol 1987; 105:2037-2041.
factor gene expression by intimal cells in healing 54. Bellón JM, Buján J, Hernando A, Honduvilla NG, Jurado
polytetrafluoroethylene grafts. J Vasc Surg 1990; F. Arterial autografts and PTFE vascular microprostheses:
11:580-585. Similarities in the healing process. Eur J Vasc Surg 1994;
37. Florey HW, Greer SJ, Poole JCF, Werthessen NT. The 8:694-702.
pseudointima lining fabric grafts of the aorta. Br J Exp 55. Kraiss LW, Raines EW, Wilcox JN, Seifert RA, Barrett BT,
Pathol 1961; 42:236-246. Kirkman TR, Hart CE, Bowen-Pope DF, Ross R, Clowes
38. Ghidoni JJ, Liotta D, Hall CW, Adams JG, Lechter A, AW. Regional expression of the platelet-derived growth
Barrionueva M, O’Neal RM, DeBakey ME. Healing of factor and its receptors in a primate graft model of vessel
pseudointimas in velour-lined, impermeable arterial pros- wall assembly. J Clin Invest 1993; 92:338-348.
theses. Am J Pathol 1968; 53:375-390. 56. Anderson JM, Miller KM. Biomaterial biocompatability
39. Lo Gerfo FW, Quist WC, Nowak MD, Crawshaw HM, and the macrophage.Biomaterials 1984; 5:5-10.
Haudenschild CC. Downstream anastomotic hyperplasia: 57. Weselow A. Biological behaviour of tissue and prosthetic
A mechanism of failure in Dacron arterial grafts. Ann Surg grafts. In Haimovici, ed. Vascular Surgery: Principles and
1983; 197:479-483. Techniques. New York: Appleton-Century-Crofts.
40. Stewart GJ, Essa N, Chang KHY, Reichle FA. A scanning 1984:93-118.
and transmission electron microscope study of the limi- 58. Wu MH, Shi Q, Wecheak AR, Clowes AW, Gordon IL,
nal coating on dacron prostheses in the canine thoracic Sauvage LR. Definitive proof of endothelialisation of a
aorta. J Lab Clin Med 1975; 85:208-226. dacron arterial prosthesis in a human being. J Vasc Surg
41. Branson DF, Picha GJ, Desprez J. Expanded 1995; 21:862-867.
polytetrafluoroethylene as a microvascular graft: A study 59. Shi Q, Hong M, Onuki Y, Ghali R, Hunter GC, Johansen
of four fibril lengths. Plastic and Reconstructive Surgery KH, Sauvage LR. Endothelium on the flow surface of
1985; 76:754-763. human aortic dacron vascular grafts. J Vasc Surg 1997;
42. Kogel H, Amselgruber W, Frösch D, Mohr W, Cyba- 25:736-742.
Altunbay S. New techniques of analyzing the healing pro- 60. Sauvage LR, Berger K, Wood SJ, Nakagawa Y, Mansfield
cess of artifical vascularization and endothelialization. Res PB. An external velour surface for porous arterial pros-
Exp Med 1989; 189:61-68. theses. Surgery 1971; 70:940-953.
43. Berger K, Sauvage LR, Rao AM, Wood SJ. Healing of ar- 61. Baitella-Erberle G, Groscurth P, Zilla P, Lachat M, Muller-
terial prostheses in man: Its incompleteness. Ann Surg Glauser W, Schneider J et al. Long term results of tissue
1972; 175: 118-127. development and cell differentiation on Dacron prosthe-
44. Berkowitz HD, Perloff JL, Roberts B. Pseudointimal de- ses seeded with microvascular cells in dogs. J Vasc Surg
velopment on microporous polyurethane lattices. Surgery 1993; 18:1019-1028
1972; 14:888-896. 62. Burkel WE, Ford JW, Vinter DW, Kahn RH, Graham LM,
45. Harrison HJ. Synthetic materials as vascular prostheses. Stanley JC. Fate of knitted dacron velour avascular grafts
Ia. A comparative study in small vessels of nylon, dacron, seeded with enzymatically derived autologous canine en-
orlon, ivalon sponge and teflon. Amer J Surg 1958; dothelium. ASAIO Transactions 1982; 28:178-184.
95:3-15.
40 Tissue Engineering of Prosthetic Vascular Grafts
63. Graham LM, Vinter DW, Ford JW, Kahn RH, Burkel WE, 81. Qu MH, Shi Q, Kouchi Y, Onuki Y, Ghali R, Yoshida H,
Stanley JC. Endothelial cell seeding of prosthetic vascular Kaplan S, Sauvage LR. Implant site influence on arterial
grafts. Early experimental studies with cultured autolo- prosthesis healing: A comparative study with a triple im-
gous canine endothelium. Arch Surg 1980; 115:929-933. plantation model in the same dog. J Vasc Surg 1997;
64. Margolin DA, Kaufman BR, DeLuca DJ, Fox PL, Graham 25:528-536.
LM. Increased platelet-derived growth factor production 82. Gouny P, Hocquet-Cheynel C, Martin-Mondiere C,
and intimal thickening during healing of dacron grafts in Bensenane J, Bonneau M, Nussaume O. Incorporation of
a canine model. J Vasc Surg 1993; 17:858-867. fibronectin-impregnated vascular prostheses in the pig.
65. Schmidt SP, Hunter TJ, Hirko M, Belden TA, Evancho Microscope study. J Cardiovasc Surg 1995; 36:573-580.
MM, Sharp WV, Donovan DL. Small diameter vascular 83. Sauvage LR, Berger K, Mansfield PB. Future directions in
prostheses: Two designs of PTFE and endothelial cell- the development of arterial prostheses for small and me-
seeded and nonseeded dacron. J Vasc Surg 1985; dium caliber arteries. Surgery 1974; 54:213-228.
2:292-297. 84. Liudenauer SM, Weber TR, Miller TA. Velour vascular
66. Schmidt SP, Monajjem N, Evancho MM, Pippert TR, prostheses. Trans Am Soc Artif Intern Org 1974;
Sharp WV. Microvascular endothelial cell seeding of small 20:314-319.
diameter dacron vascular grafts. J Invest Surg 1988; 85. Clagett PC. In vivo evaluation of platelet reactivity with
1:35-44. vascular prostheses. In: Stanley JC, ed. Biologic and Syn-
67. Criado E, Marston WA, Reddick R, Woosley JT. Endot- thetic Vascular Prostheses. New York: Grune & Stratton,
helial coverage of endovascular dacron grafts in dogs. J 1982:131-152.
Vasc Surg 1996; 2:736-737. 86. Köveker GB, Burkel WE, Graham LM, Wakefield TW,
68. Sottiurai VS, Sue SL, Rau DJ, Tran AB. Comparative Stanley JC. Endothelial cell seeding of expanded
analysis of pseudointima biogenesis in gelseal coated polytetrafluoroethylene vena cava conduits: Effects on lu-
dacron knitted graft versus crimped and noncrimped graft. minal production of prostacyclin, platelet adherence, and
J Cardiovasc Surg 1989; 30:902-909. fibrinogen accumulation. J Vasc Surg 1988; 7:600-605.
69. Nomura Y. The ultra-structure of the pseudointima lin- 87. Hamdan AD, Misare B, Contreras M, LoGerfo FW, Quist
ing synthetic arterial grafts in the canine aorta with spe- WC. Evaluation of anastomotic hyperplasia progression
cial reference to the origin of the endothelial cell. using the cyclin specific antibody MIB-1. Am J Surg 1996;
Cardiovasc Surg 1970; 22:282-29. 172:168-171.
70. Mesh CL, Majors A, Mistele D, Graham LM, Ehrhart LA. 88. van der Lei B, Wildevuur RH. Improved healing of mi-
Graft smooth muscle cells specifically synthesise increased crovascular PTFE prostheses by induction of a clot layer:
collagen. J Vasc Surg 1995; 22:142-149. An experimental study in rats. Plastic and Reconstructive
71. De Bakey ME, Jordan Jr GL, Abbott JP, Halbert B, O’Neal Surgery 1989; 84:960-968.
R. The fate of dacron vascular grafts. Arch Surg 1964; 89. Chen C, Ku DN, Kikeri D, Lumsden AB. Tenascin: A
89:757-782. potential role in human arteriovenous PTFE graft failure.
72. Hertzer NR. Regeneration of endothelium in knitted and J Surg Res 1996; 60:409-416.
velour dacron vascular grafts in dogs. J Cardiovas Surg 90. Wu MH, Kouchi Y, Onuki Y, Shi Q, Ghali R, Sauvage
1981; 22:223-230. LR. Effect of differential shear stress on platelet aggrega-
73. Rosenfeld JC, Savarese R, McCombs PR, DeLaurentis DA. tion, surface thrombisis, and endothelialization of
Endothelial infiltration and lining of knitted dacron arte- bilaterial carotid-femoral grafts in the dog. J Vasc Surg
rial grafts. Surgical Forum 1981. 1995; 22:382-392.
74. De Bakey ME, Jordan GL Jr, Beall AC, O’Neal RM, Abbott 91. Shepard AD, Eldrup-Jorgensen J, Keough EM, Foxall TF,
JP, Halpert B. Basic biologic reactions to vascular grafts Ramberg K, Connolly RJ, Mackey WC, Gravis V, Auger
and prostheses. Surg Clin North Am 1965;45:477. KR, Libby P, O’Donnell TF, Callow AD. Endothelial cell
75. Herring MB, Dilley R, Jersild RA Jr, Boxer L, Gardner A, seeding of small-caliber synthetic grafts in the baboon.
Glover J. Seeding arterial prostheses with vascular endot- Surgery 1986; 99:318-325.
helium. Ann Surg 1979; 190:84-90. 92. Mansfield PB, Wechezak AR, Sauvage LR. Preventing
76. Burkel WE, Vinter DW, Ford JW, Kahn RH, Graham LM, thrombus on artificial vascular surfaces: True endothelial
Stanley JC. Sequential studies of healing in endothelial cell linings. ASAIO Transactions 1975; 21:264-271.
seeded vascular prostheses: Histologic and ultrastructure 93. Deutsch M, Meinhart J, Vesely M, Fischlein T, Groscurth
characteristics of graft incorporation. J Surg Res 1981; P, von Oppell U, Zilla P. In vitro endothelialization of
30:305-324. expanded polytetrafluorethylene grafts: A clinical case re-
77. Shi Q, Wu MH, Hayashida N, Wechezak AR, Clowes AW, port after 41 months of implantation. J Vasc Surg 1977;
Sauvage LR. Proof of fallout endothelialization of imper- 25:757-763.
vious dacron grafts in the aorta and inferior vena cava of 94. Zhang H, Williams GM. Capillary and venule prolifera-
the dog. J Vasc Surg 1994; 20:546-557. tion in the healing process of dacron venous grafts in rats.
78. Kohler TR, Stratton JR, Kirkman TR, Johansen KH, Surgery 1992; 111:409-415.
Zierler BK, Clowes AW. Conventional versus high-poros- 95. Goldman MA, Norcott HC, Hawker RJ, Drolc Z,
ity polytetrafluorethylene grafts: Clinical evaluation. Sur- McColum CN. Platelet accumulation on mature Dacron
gery 1992; 112:901-907. grafts in man. Br J Surg 1982; 69:S38-S40.
79. Sauvage LR, Berger KE, Wood SJ, Smith JC, Mansfield 96. Stratton JR, Thiele BL, Ritchie JL. Platelet desposition on
PD. Interspecies healing of porous arterial prostheses. dacron aortic bifurcation grafts in man: Quantitation with
Arch Surg 1974; 109:698-705. indium-III platelet imaging. Circulation 1982;
80. Hammond W. Surface population with blood-borne cells. 66:287-1293.
In: Zilla P, Greisler H, eds. Tissue Engineering of Pros- 97. McCollum CN, Kester R, Rajah SM, Learoyd P, Pepper
thetic Vascular Grafts. Austin: Landes Bioscience, 1998. M. Arterial graft maturation: The duration of thrombotic
The Lack of Healing in Conventional Vascular Grafts 41
activity in Dacron aortobifemoral grafts measured by 114. Colman RW, Scott CF, Schmaier AH, Wachtfogel YT,
platelet and fibrinogen kinetics. Br J Surg 1981; 68:61-64. Pixley RA, Edmunds LH Jr. Initiation of blood coagula-
98. Hussain S, Glover JL, Augelli N, Bendick PJ, Daupin D, tion at artificial surfaces. Ann NY Acad Sci 1987;
McKain M. Host response to autologous endothelial seed- 516:253-267.
ing. J Vasc Surg 1989; 9:656-664. 115. Clark RAF.Mechanisms of cutaneous wound repair. Der-
99. Kaufman BR, DeLuca DJ, Folsom DL, Mansell SL, matology in General Medicine 1993; 473-486.
Gorman ML, Fox PL, Graham LM. Elevated platelet-de- 116. Albelda SM, Buck CA. Integrins and other cell adhesion
rived growth factor production by aortic grafts implanted molecules. FASEB Journal 1990; 4:2868-2880.
on a long-term basis in a canine model. J Vasc Surg 1992; 117. Luscinkas FW, Lawler J. Integrins as dynamic regulators
15:806-816. of vascular function. FASEB Journal 1994; 8:929-938.
100. Ombrellaro MP, Stevens SL, Kerstetter K, Freeman MB, 118. Brooks PC, Clark RAF, Cheresh DA. Requirement for
Goldman MH. Healing characteristics of intraarterial vascular integrin #v!3 for angiogenesis. Science 1994;
stented grafts: Effect of intraluminal position on prosthetic 264:569.
graft healing. Surgery 1996; 120:60-70. 119. Friedlander M, Brooks PC, Shaffer RW, Kincaid CM,
101. Plate G, Hollier LH, Fowl RJ, Sande JR, Kaye MP. En- Varner JA, Cheresh DA. Definition of two angiogenic
dothelial seeding of venous prostheses. Surgery 1984; pathways by distinct # v integrins. Science 1995;
96:929-936. 270:1500-1502.
102. Seeger JM, Klingman, N. Improved in vivo endotheli- 120. Gailit J, Clark RAF. Wound repair in the context of the
alization of prosthetic grafts by surface modification with extracellular matrix. Cur Opin Cell Biol 1994; 6:717-725.
fibronectin. J Vasc Surg 1988; 8:476-482. 121. Grainger DJ, Wakefield L, Bethell HW, Farndale RW,
103. Zilla P, Fasol R, Dudeck U, Kadletz M, Siedler S, Preiss P Metcalfe JC. Release and activation of platelet latent TGF-
et al. In situ cannulation, microgrid follow-up and low ! in blood clots during dissolution with plasmin. Nature
density plating provide first passage endothelial cell mass Medicine 1995; 1:932-937.
cultures for in vitro lining. J Vasc Surg 1990; 12:180-189. 122. Pierce GF, Mustoe TA Lingelbach C, Masakowski VR,
104. Zilla P, Fasol R, Grimm M, Fischlein T, Eberl T, Preiss P, Griffin GL, Senior RM, Deuel TF. Platelet-derived growth
Krupicka O, von Oppell U, Deutsch M. Growth proper- factor and transformting growth factor-b enhance tissue
ties of cultured human endothelial cells on differently repair activities by unique mechanisms. J Cell Biol 1989;
coated artificial heart materials. J Thorac and Cardiovasc 109:429-440.
Surg 1991; 101:671-680. 123. Marcus AJ. Platelet function. N Engl J Med 1969;
105. Hollier LH, Fowl RJ, Pennell RC, Heck CF, Winter KA, 280:1213-1220, 1278-1284, 1330-1335.
Fass DN, Kaye MP. Are seeded endothelial cells the ori- 124. Marcus AJ. The role of lipids in platelet function: With
gin of neointima on prosthetic vascular grafts? J Vasc Surg particular reference to the arachiodonic acid pathway. J
1986; 3:65-73. Lipid Res 1978; 19:793.
106. Beck L Jnr, D’Amore PA. Vascular development: Cellular 125. Walsh PN. Platelet coagulant activities and hemostasis: A
and molecular regulation. FASEB Journal 1997; hypothesis. Blood 1974; 43:597.
11:365-373. 126. Nehls V, Herrmann R. The configuration of fibrin clots
107. Hirschi KK, Rohosky SA, D’Amore PA. Cell-cell interac- determines capillary morphogenesis and endothelial cell
tions in vessel assembly: A model for the fundamentals migration. Microvascular Research 1996; 51(3):347-364.
of vascular remodelling. Thrombosis and Haemostasis 127. Herbert CD, Nagaswami C, Brittner GD, Hubble JA,
1997; 77:894-900. Weisel JW. Effects of fibrin micromorphology on neurite
108. Cheresh DA, Berliner SA, Vicente V, Ruggeri ZM. Recog- growth from dorsal root ganglia cultured within 3-dimen-
nition of distinct adhesive sites on fibrinogen by related sional fibrin gels. J Biomed Mat Res 1998 (in press).
integrins on platelets and endothelial cells. Cell 1989; 128. Henke CA, Roongta U, Mickelson DJ, Knutson JR,
58:945-53. McCarthy JB. CD44-related chondroitin sulphate
109. Dvorak HF, Nagy JA, Berse B, Brown LF, Yeo KT, Yeo proteoglycan, a cell surface receptor implicated with tu-
TK, Dvorak AM, Van De Water L, Sioussat TM, Senger mour cell invasion, mediates endothelial cell migration on
GR. Vascular permeability factor, fibrin, and the patho- fibrinogen and invasion into the fibrin matrix. J Clin In-
genesis of tumor stoma formation. Ann NY Acad Sci 1992; vest 1996; 97:2541-2552.
667:101-111. 129. Dejana E, Lampugnani MG, Giorgi M et al. Fibrinogen
110. Gamble JR, Matthias LJ, MeyerG, Kaur P, Russ G, Faull induces endothelial cell adhesion and spreading via the
R, Berndt MC, Vadas MA. Regulation of in vitro capil- release of endogenous matrix proteins and the recruitment
lary tube formation by anti-integrin antibodies. J Cell Biol of more than one integrin receptor. Blood 1990;
1993; 121:931-943. 75:1509-1517.
111. Tabbara M, White RA. Biologic and prosthetic materials 130. Zilla P, Deutsch M, Meinhart, Puschmann R, Eberl T,
for vascular conduits. In: Veith FJ, Hobson RW, Williams Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I,
RA and Wilson SE, eds. Vascular Surgery: Principles and Fischlein T. Clinical in vitro endotheliaization of
Practice. USA: McGraw-Hill, Inc., 1994:523-535. femoropopliteal bypass grafts: An acturial follow-up over
112. King M, Blais P, Guidoin R, Prowse E, Marcois M, three years. J Vasc Surg 1994; 19:540-548.
Gosselin C, Noel HP. Polyethylene terephthalate 131. Rifkin DB, Kojima S, Abe M and Harpel JG. TGF-!: Struc-
(Dacron®) vascular prostheses—material and fabric con- ture, function and formation. Thrombosis and
struction aspects. Biocompatibility of Clinical Implant Haemostasis 1993; 70:177-179.
Materials, 1981;Vol II:177-207. 132. Wakefield LM, Smith DM, Flanders KC, Sporn MB. La-
113. Herring M, Gardner A and Glover J. Endothelial seeding tent transformting growth factor-b from human platelets.
on vascular prostheses. Arch Surg 1979; 114:679. J Biol Chem 1988; 263:7646-7654.
42 Tissue Engineering of Prosthetic Vascular Grafts
133. Miyazono K, Hollman U, Wornstadt C, Heldin C-H. La- 151. Kalman PG, Rotstein OD, Niven J, Glynn MFX,
tent high molecular weight complex of transforming Romaschin AD. Differential stimulation of macrophage
growth factor !1: Purification from human platelets and procoagulant activity by vascular grafts. J Vasc Surg 1993;
structural characterisation. J Biol Chem 1988; 17:531-537.
263:6407-6417. 152. Bach RR. Initiation of coagulation by tissue factor. CRC
134. Harpel JG, Metz CN, Kojima S, Rifkin DB. Control of Crit Rev Biochem 1988; 23:339-368
transformting growth factor-b activity: Latency vs. acti- 153. Nemerson Y. Tissue factor and haemostasis. Blood 1988;
vation. Progress in Growth Factor Research 1992; 71:1-8.
4:321-335. 154. Miller KM, Huskey RA, Bigby LF, Anderson JM.
135. Mohle R, Green D, Moore MAS, Nachman RL, Rafii S. Characterisation of biomedical polymer-adherent mac-
Constitutive production and thrombin-induced release of rophages: IL-1 generation and SEM studies. Biomaterials
vascular endothelial growth factor by human megakaryo- 1989; 10:187-196.
cytes and platelets. Proc Natl Acad Sci USA, 1997; 155. Schwager I, Jungi TW. Effect of human recombinant
94:663-668. cytokines on the induction of macrophages procoagulant
136. Taichman NS, Young S, Cruchley AT, Taylor P, Paleolog activity. Blood 1994; 83(1):152-160.
E. Human neutrophils secrete vascular endothelial growth 156. Bastida E, Ordinas A, Escolar G, Jamieson GA. Tissue fac-
factor. J Leukoc Biol 1997; 62:397-400. tor in microvesicles shed from U87MG human glioblas-
137. Pepper MS, Vassalli JD, Orci L, Montesano R. Biphasic toma cells induces coagulation, platelet aggregation and
effect of transformting growth factor%b1 on in vitro an- thrombogenesis. Blood 1984; 64:177-184.
giogenesis. Exp Cell Res 1993; 204(2):356-363. 157. Lewis JC, Bennett-Cain AL, DeMars CS, Doellgast GJ,
138. Pepper MS, Vassalli JD, Wilks JW, Schweigerer L, Orci L Grant KW, Jones NL, Gupter M. Procoagulant activity
and Montesano R. Modulation of bovine microvascular after exposure of monocyte-derived macrophages to mini-
endothelial cell proteolytic properties by inhibitors of mally oxidized low density lipoprotien. Colocalization of
angiogenesis. J Cell Biochem 1994; 55(4):419-434. tissue factor antigen and nascent fibrin fibers at the cell
139. Devine DV, Carter CJ. Profibrinolytic and antifibrinolytic surface. Amer J Path 1995; 147:1029-1040.
effects of platelets. Cor Art Dis 1995; 6:915-922. 158. Hogg N, Berlin C. Structure and function of adhesion
140. Murphy-Ullrich JE, Mosher DF. Interactions of receptors in leukocyte trafficking. Immunology Today
thrombospondin with cells in culture: Rapid degradation 1995; 16:327-330.
of both soluble and matrix thrombospondin. Seminars in 159. Mantovani A, Dejana E. Cytokines as cummunication sig-
Thrombosis and Haemostasis 1987; 13(3):343-351. nals between leukocytes and endothelial cells. Immunol-
141. Good DJ, Polverini PJ, Rastinejad F, LeBeau MM, Lem- ogy Today 1989; 10;370-375.
ons RS, Frazier WA and Bouck NP. A tumour suppres- 160. Nunes I, Shapiro RL, Rifkin DB. Characterization of la-
sor-dependent inhibitor of angiogenesis is immunologi- tent TGF-beta activation by murine peritoneal macroph-
cally and functionally indistinguishable from a fragment ages. J Immunol 1995; 155:1450-9.
of thrombospondin. PNAS 1990; 87:6624-6628. 161. Wahl SM, Allen JB, Weeks BS, Wong HL, Klotman
142. Iruela-Arispe ML, Bornstein P, Sage H. Thrombospondin PE.Transforming growth factor beta enhances integrin ex-
exerts an antiangiogenic effect on cord formation by pression and type IV collagenase secretion in human
enothelial cells in vitro. PNAS 1991; 88:5026-5030. monocytes. Proc Natl Acad Sci USA 1993; 90:4577-81.
143. Bagavandross P, Wilks JW. Specific inhibition of endot- 162. Garcia M. Transformting growth factor-beta 1 stimulates
helial cell proliferation by thrombospondin. Biochem macrophage urokinase expression and release of matrix-
Biophys Res Comm 1990; 170:867-872. bound basic fibroblast growth factor. J Cell Physiol 1993;
144. Murphy-Ullrich JE, Hook M. Thrombospondin modulates 155:595-605.
focal adhesions in endothelial cells. J Cell Biol 1989; 163. Loscalzo J. The macrophage and fibrinolysis. Semin
109:1309-1319. Thromb Hemost 1996; 22:503-6.
145. Hogg PJ. Thrombospondin 1 as an enzyme inhibitor. 164. Gross TJ, Leavell KJ, Peterson MW. Cd11b/cd18 medi-
Thrombosis and Haemostasis 1994; 72: 787-792. ates the neutrophil chemotactic activity of fibrin degra-
146. Mosher DF, Misenheimer TM, Stenflo J, Hogg PJ. Modu- dation product D domain. Thromb Haemost 1997;
lation of fibrinolysis by thrombospondin. Ann N Y Acd 5:894-900.
Sci 1992; 667:64-9. 165. Premack BA, Schall TJ. Chemokine receptors: Gateways
147. Schultz-Cherry S, Ribeiro S, Gentry L, Murphy-Ullrich JE. to inflammation and infection. Nature Medicine 1996;
Thrombospondin binds and activates the small and large 2:1174-1178.
forms of latent transformting growth factor%! in a chemi- 166. Rhodes NP, Hunt JA, Williams DF. Macrophage subpopu-
cally defined system. JBC 1994; 269(43):26775-26782. lation differentiation by stimulation with biomaterials. J
148. Assoian RK, Fleurdelys BE, Stevenson HC, Miller PJ, Biomat Res 1997; 37:481-488.
Madtes DK, Raines EW, Ross R and Sporn MB. Expres- 167. Azeez A, Yun J, DeFife K, Colton E, Cahallan L,
sion and secretion of type 1 transforming growth factor Verhoeven M, Cahallan P, Anderson JM, Hiltner A. In
by activated human macrophages. Proc Natl Acad Sci vitro monocyte adhesion and activation on modified FEP
1987; 84:6020-6024. copolymer surfaces. J Appl Poly Sci 1995; 58:1741-1749.
149. DiPietro LA, Polverini PJ. Angiogenic macrophages pro- 168. Bonfield TL, Colton E, Marchant RE, Anderson JM.
duce the angiogenic inhibitor thrombospondin-1. Amer J Cytokine and growth factor production by monocytes/
Pathol 1993; 143:678-684. macrophages on protein preadsorbed polymers. J Biomat
150. Van Ginkel CJW, Van Aken WG, Oh JIH, Vreeken J. Res 1992; 26:837-850.
Stimulation of monocyte procoagulant activity by adher- 169. Hunt JA, Meijs G, Williams DF. Hydrophillicty of poly-
ence to different surfaces. Brit J Haemot 1997; 37:35-45. mers and soft tissue responses; a quantitative analysis. J
Biomat Res 1997; 36:542-549.
The Lack of Healing in Conventional Vascular Grafts 43
170. Hunt JA, Flanagan, BF, McLaughlin PJ, Strickland I, Wil- 186. Rapoza RJ, Horbett TA. Postadsorptive transitions in fi-
liams DF. Effect of biomaterial surface charge on the in- brinogen: Influence of polymer properties. J Biomat Res
flammatory response: Evaluation of cellular infiltration 1990; 24(10):1263-1287.
and TNF-! production. J Biomat Res 1996; 31:139-144. 187. Slack SM, Horbett TA. Changes in fibrinogen adsorbed
171. Yun JK, DeFife, K, Colton E, Stack S, Azeez A, Cahalan to segmented polyurethanes and
L, Verhoeven M, Cahalan P, Anderson JM. Human mono- hydroxyethylmethacrylate-ethylmethacrylate copolymers. J
cyte/macrophage adhesion and cytokine production on Biomat Res 1992; 26(12):1633-1649.
surface modified poly(tetrafluoroethylene/hexafluoro- 188. Zisman WA. Ind Eng Chem 1963; 55:18-25.
propylene) polymers with and without protein 189. Boffa GA, Lucien N, Faure A, Boffa MC. J Biomat Res
preadsorption. J Biomat Res 1995; 29:257-268. 1977; 11:3-17.
172. Bonfield TL, Colton E, Anderson JM. Plasma protein 190. Noishiki Y. Application of immunoperoxidase method to
adsorbed biomedical polymers: Activation of human electron microscopic observation of plasma protein on
monocytes and induction of IL-1. J Biomat Res 1989; polymer surface. J Biomed Mater Res 1982; 16:359-67.
23:535-548. 191. Tang L, Eaton JW. Fibrin(ogen) mediates acute inflam-
173. Bonfield TL, Anderson JM. Functional versus quantita- matory responses to biomaterials. J Exper Med 1993;
tive comparison of Il-1! from monocytes/macrophages on 178:2147-2156.
biomedical polymers. J Biomat Res 1993; 27:1195-1199. 192. Tang L, Lucas AH, Eaton JW. Inflammatory responses to
174. Krause TJ, Robertson FM, Liesch JB, Wasserman AJ, implanted polymeric biomaterials: Role of surface-ab-
Greco RS. Differential production interleukin 1 on the sorbed IgG. J Lab Clin Med 1993; 122:292-300.
surface of biomaterials. Arch Surg 1990; 125:1158-1160. 193. Tang L, Eaton JW. Inflammatory responses to
175. Miller KM, Anderson JM. Human monocyte/macrophage biomaterials. Amer J Clin Path 1995; 103:466-471.
activation and IL-1 generation by biomedical polymers. J 194. Tang L, Ugarova TP, Plow EF, Eaton JW. Molecular de-
Biomat Res 1988; 22:713-731. terminants of acute inflammatory responses to
176. Miller KM, Anderson JM. In vitro stimulation of fibro- biomaterials. J Clin Invest 1996; 97: 1329-1334.
blast activity by factors generated from human monocytes 195. McNally AK, Anderson JM. Complement C3 participa-
activated by biomedical polymers. J Biomat Res 1989; tion in monocyte adhesion to different surfaces. PNAS
23:911-930. 1994; 91:10119-10123.
177. Miller KM, Rose-Caprara V, Anderson JM. Generation of 196. Bernatchez SF, Parks PJ, Gibbons DF. Interaction of
IL-1 like activity in response to biomedical polymer im- macrophages with fibrous materials in vitro. Biomaterials
plants: A comparison of in vitro and in vivo models. J 1996; 17:2077-2086.
Biomat Res 1989; 23:1007-1026. 197. Kuwabara K, Ogawa S, Matsumoto M, Koga S, Clauss M,
178. Swartbol P, Truedsson L, Pärsson, Norgren L. Tumor Pinsky DJ, Lyn P, Leavy J, Witte L, Joseph-Silverstein J,
necrosis factor-# and interleukin-6 release from white Furies MB, Torcia G, Cozzolino F, Kamada T, Stern DM.
blood cells induced by different graft maerials in vitro are Hypoxia-mediated inducation of acidic/basic fibroblast
affected by pentoxifylline and iloprost. J Biomed Mater growth factor and platelet-derived growth factor in mono-
Res 1997; 36:400-406. nuclear phagocytes stimulates growth of hypoxic endot-
179. Bonfield TL, Colton E, Anderson JM. Protein adsorption helial cells. Proc Nat Acad Sci 1995; 92:4606-4610.
of biomedical polymers influences activated monocytes to 198. Van Otteren GM, Standiford TJ, Kunkel SL, Danforth JM,
produce fibroblast stimulating factors. J Biomat Res 1992; Strieter RM. Alterations of ambient oxygen tension modu-
26:457-465. late the expression of tumour necrosis factor and mac-
180. Petillo O, Peluso G, Ambrosio, L, Nicolais L, Kao WJ, rophage inflammatory protein-1 from murine alveolar
Anderson JM. In vivo induction of macrophage Ia anti- macrophages. Amer J Respir Cell Mol Biol 1995;
gen (MHC class II) expression by biomedical polymers 13(4):399-409.
in the cage implant system. J Biomat Res 1994; 28:635- 199. Leibovich SJ, Polverini PJ, Shepard HM, Wiseman DM,
646. Shively V, Nuseir N. Macrophage-induced angiogenesis is
181. Gemmell CH, Black JP, Yeo EL, Sefton MV. Material in- mediated by tumour necrosis factor- # . Nature 1987;
duced up-regulation of leukocyte Cd11b during whole 329:630-632.
blood contact: Material differences and a role for comple- 200. Patterson C, Perrella MA, Endege WO, Yoshizumi M, Lee
ment. J Biomat Res 1996; 32:29-35. M, Haber E. Downregulation of vascular endothelial
182. Swartbol P, Truedsson L, Pärsson, Norgren L. Surface ad- growth factor receptors by tumour necrosis factor-# in
hesion molecule expression on human blood cells induced cultured human vascular cells. Journal of Clinical Inves-
by vascular graft materials in vitro. J Biomat Res 1996; tigation 1996; 98(2):490-496.
32:669-676. 201. Spyridopoulos I, Brogi E, Kearney M, Sullivan AB, Cetrulo
183. Anderson JM, Ziats NP, Azeez A, Brunstedt MR, Stack S, C, Isner JM, Losordo DW. Vascular enothelial growth
Bonfield TL. Protein adsorption and macrophage activa- factor inhibits endothelial cell apoptosis induced by tu-
tion on PDMS and silicone rubber. J of Biomat Sci Poly- mour necrosis factor-#: Balance between growth and death
mer Edition 1995; 7:159-169. signals. J Mol Cell Cardiol 1997; 29:1321-1330.
184. Anderson JM, Bonfield TL, Ziats NP.Protein adsorption 202. Defife KM, Jenney CR, Colton E, Anderson JM. Confocal
and cellular adhesion and activation on biomedical poly- and light microscopic evaluation of silane surface-depen-
mers. Biomaterials 1990; 13:375-382. dent macrophage development and IL-4 induced foreign
185. Ziats NP, Pankowsky DA, Tierney BP, Ratnoff OD, Ander- body giant cell formation. Fifth World Biomaterials Con-
son JM. Absorption of Hageman factor and other human gress 1996.
plasma proteins to biomedical polymers. J Lab Clin Med 203. Kao WJ, Zhao QH, Hiltner A, Anderson JM. Theoretical
1990; 116:687-96. analysis of in vivo macrophage adhesion and FBGC for-
mation on PDMS, low density polyethylene and PEUs. J
Biomat Res 1994; 28:73-79.
44 Tissue Engineering of Prosthetic Vascular Grafts
204. Kao WJ, Hiltner A, Anderson JM, Lodoen GA. Theoreti- 218. Vernon RB, Sage EH. Between molecules and morphol-
cal analysis of in vivo macrophage adhesion and FBGC ogy: Extracellular matrix and creation of vascular form.
formation on strained poly(etherurethane urea) elas- Amer J Pathol 1995; 147(4):873-883.
tomers. J Biomat Res 1994; 28:819-829. 219. Polverini PJ. Cellular adhesion molecules: Newly identi-
205. Mathur A, Collier TO, Kao WJ, Wiggins M, Schubert MA, fied mediators of angiogenesis. Amer J Pathol 1996;
Hiltner A, Anderson JM. In vivo biocompatabiity and 148(4):1023-1029.
biostability of modified polyurethanes. J Biomater Res 220. Iijima K, Yoshikawa N, Connolly DT, Nakamura H. Hu-
1997; 36:246-257. man mesangial cells and peripheral blood mononuclear
206. McNally AK, Anderson JM. The lymphokine IL-4 induces cells produce vascular permeability factor. Kidney Inter-
very large FBGC and syncytia from human macs in a national 1993; 44:959-966.
material surface property dependent manner in vitro. Fifth 221. Polverini PJ, Contran RS, Gimbrone MA Jr, Unanue ER.
World Biomaterials Congress 1996. Activated macrophages induce vascular proliferation. Na-
207. McNally AK, DeFife KM, Anderson JM. IL-4 induced ture 1977; 269:804-806.
macrophage fusion is prevented by inhibitors of mannose 222. Anderson JM. Mechanisms of inflammation and infection
receptor activity. Am J of Pathol 1997; 149:975. with implanted devices. Cardiovasc Pathol 1993;
208. McNally AK, Anderson JM. IL-4 induces foreign body 2:33S-41S.
giant cells from human monocytes/macrophages. Amer J 223. Pepper MS, Belin D, Montesano L, Orci L and Vassalli
Pathol 1995; 47:1487-1499. JD. Transforming growth factor-b1 modulates basic fibro-
209. Essner R, Rhoades K, McBride WH, Morton DL, blast growth factor-induced proteolytic and angiogenic
Economou JS. IL-4 downregulates IL-1 and TNF gene properties of endothelial cells in vitro. J Cell Biol 1990;
expression in human monocytes. J Immunol 1989; 111:743-755.
142:3857-3861. 224. van der Lei B, Dijk F, Bartels H, Jongebloed WL, Robinson
210. Lehn M, Weiser WY, Engelhor S, Gillis S, Remold HG. PH. Healing of microvenous PTFE prostheses implanted
IL-4 inhibits H2O2 production and antileishmanial capac- into the rat femoral vein. Brit J Plastic Surg 1993;
ity of human culture monocytes mediated by fn- &. J 46:110-115.
Immunol 1989; 143:3020-3024. 225. Sato Y, Rifkin DB. Inhibition of endothelial cell move-
211. Lacraz S, Nicod L, Galve-de Rochemonteix B, Bauberger ment by pericytes and smooth muscle cells: Activation of
C, Dayer J-M, Welgus HG. Supression of a latent transforming growth factor—!1-like molecule by
metalloproteinase biosynthesis in human macrophages by plasmin during coculture. J Cell Biol 1989; 109:309-315.
interleukin-4. J Clin Invest 1992; 90:382-388. 226. Clowes AW. Platelet-derived growth factor activity and
212. Zhao Q, Topham N, Anderson JM, Hiltner A, Lodoen G, mRNA expression in healing vascular grafts in baboons. J
Payet CR. Foreign-body giant cells and polyurethane Clin Invest 1991; 87:406-414.
biostability: In vivo correlation of cell adhesion and sur- 227. Jiang B, Yamamura S, Nelson PR, Mureebe L, Kent KC.
face cracking. J Biomat Res 1991; 35:177-183. Differential effects of platelet-derived growth factor
213. Anderson J. Inflammatory reaction: The nemesis of isotypes on human smooth muscle cell proliferation and
implants. In: Zilla P and Greisler H, eds. Tissue Engineer- migration are mediated by distinct signaling pathways.
ing of Prosthetic Vascular Grafts. Austin: Landes Bio- Surgery 1996; 120:427-432.
science, 1998. 228. Koyama N, Hart CE, Clowes AW. Different functions of
214. Forsyth EA, Hamdy MA, Neville RF, Sidawy AN. Prolif- the platelet-derived growth factor-# and—! receptors for
eration and extracellular matrix production by human the migration and proliferation of cultured baboon
infragenicular smooth muscle cells in response to smooth muscle cells. Circ Res 1994; 75:682-691.
interleukin-1!. J Vasc Surg 1997; 26:1002-1008. 229. Sterpetti AV, Cucina A, D’Angelo LS, Cardillo B,
215. Makita S, Nakamura M, Yoshida H, Hiramori K. Carvallaro A. Shear stress modulates the proliferation rate,
Autocrine growth inhibition of IL-1 beta-treated cultured protein synthesis, and mitogenic activity of arterial smooth
human aortic smooth muscle cells: Possible role of nitric muscle cells. Surgery 1993; 113:691-699.
oxide. Heart Vessels 1996; 11:223-8. 230. Watkins MT, Sharefkin JB, Zajtchuk R, Maciag TM,
216. Meredith JE Jr, Schwartz MA. Integrins, adhesion and D’Amore PA, Ryan US, Van Wart H, Rich NM. Adult
apoptosis. Trends in Cell Biol 1997; 7:146-150. human saphenous vein endothelial cells: Assessment of
217. Davis GE, Camarillo CW. Regulation of endothelial cell their reproductive capacity for use in endothelial seedingof
morphogenesis by integrins, mechanical forces and ma- vascular prostheses. J Surg Res 1984; 36:588-96.
trix guidance pathways. Exper Cell Res 1995; 216:113-123.
CHAPTER 2
Noncompliance: The Silent Acceptance of a Villain
Alexander M. Seifalian, Alberto Giudiceandrea,
Thomas Schmitz-Rixen, George Hamilton
Introduction
A rterial occlusive disease is a problem of epidemic proportions in our aging society with
increasing need for vascular reconstructive surgery.1 The best results are achieved with
autologous vein, but because this is often not available, historically there has been extensive
research into production of a suitable substitute graft material.2 At present there are a hand-
ful of such vascular grafts that are either commercially available or under development, but
unfortunately no graft material has yet performed satisfactorily.3,4 Baird and Abbott’s hy-
pothesis of 1976 that a difference in circumferential compliance of a vascular graft and the
host artery is detrimental to graft performance was eventually experimentally verified in
1987.5,6 The nature of compliance mismatch is complex, as it is determined by the compli-
ance differences of the host artery, the anastomosis, and the graft itself. The hemodynamic
consequences of mismatch include increased impedance and decreased distal perfusion as
well as disturbed flow, turbulence and low shear stress rates.5 These changes could lead to
the development of myointimal hyperplasia around the anastomosis, finally resulting in
graft failure, particularly in small diameter vessels.7,8 Some efforts have been made over the
years to engineer prosthetic grafts with compliance similar to that found in human artery.9,10
To date these attempts have failed due to a variety of reasons, explaining our current accep-
tance of highly noncompliant and inferior graft materials. This chapter will give an over-
view of the history of compliance in vascular grafting, a discussion of the nature of compli-
ance and its measurement as a means of understanding the most recent attempts to engineer
a more compliant graft.
Historical Overview
Cardiovascular physiology started in 1628 with Harvey’s “De motu cordis et Sanguinis
in Animalibus” in which he describes the circulation.11 Fifty years later in 1676 Robert Hook
described the proportionality between stress and strain (“Ut Tensio Sic Vis” or, as the exten-
sion so the force) and the concept of elasticity of materials. Isaac Newton and his “Principia
Matematica” was the first to relate elasticity (hence compliance) to wave velocity. His for-
mula (equation 1) showed that velocity in a medium c is equal to the product of elasticity K
and the density of the flowing medium ∋.
(1)
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
46 Tissue Engineering of Prosthetic Vascular Grafts
in 1733 analyzed the role of a compliant vessel in maintain- The deformation undergone by this class of material
ing a continuous blood flow throughout the pulsation of a depends on the magnitude of the stress and on the rate at
cardiac cycle.12 The German translation of his work “He- which it is applied. Vessel compliance, the reciprocal value
mostatic”, introduced the important analogy in understand- of Young’s elastic modulus, is generally defined as the ratio
ing the physiological role of distensibility in arteries, namely of change in diameter over change in blood pressure
the Windkessel. This is an air reservoir which was fitted to (%/mm Hg x 10-2).20 It is defined by equation (2) where D
fire engines of the 18th century in order to smoothen out and P are diameter and pressure, and the d and s subscripts
the oscillations in flow of water due to intermittent pump- denote diastole and systole respectively.
ing, thus ensuring a constant flow to quench the fire.
Otto Frank in 1899 introduced a theoretical approach (2)
to explain the Windkessel effect on pressure waves.13 But it
was not until 1950 through the work of Wormersley and For convenience in comparing different vessels a stan-
McDonald that a rational mathematical approach to pulse dard reporting mean pressure of 100 mm Hg has been
flow and change in diameter over the cardiac cycle was adopted. When thickness is a major fraction of the diameter
reached which allowed comprehensive understanding of the of the vessel the distinction between inner and outer diam-
hemodynamics of arterial flow.14,15 eter becomes important. It must, therefore, be specified
Parallel to these physiological understandings, vascu- which diameter is used in calculating compliance. To com-
lar surgery evolved. The first great step which led to the foun- pute the absolute compliance the pressure should be re-
dation of vascular surgery was the pioneering work of Alexis corded at the exact point of diameter measurement, but this
Carrel in developing a technique for suturing blood vessels.16 is not always the case, especially when calculating compli-
This work published in 1902 lead to his Nobel Laureate in ance in a clinical setting.21,22
1912. The first use of a graft to replace an excised segment
of artery was reported by Goanes in Spain in 1906 when he Assessment of Compliance
bridged the defect with the popliteal vein of the patient.17 There are several strategies used to assess the elastic
Later, homologous grafts were used but these attempts were properties of vascular tissues. Classification of these meth-
frustrated by early graft degeneration and infections. In 1952 ods is to some extent arbitrary and these so-called different
appeared the first report of a synthetic graft made from methods share certain common principles. Two distinct
Vinyon-N, a new synthetic cloth used to manufacture ladies methods are commonly used, namely longitudinal compli-
undergarments.18 In 1954 Vinyon-N was formally intro- ance, which assesses elasticity of a selected length of the vas-
duced to clinical practice. Since then many different materi- cular system, and circumferential compliance. This latter
als have been introduced, with only Darcon and Teflon per- method is the most frequently used and assesses the com-
forming acceptably, such that virtually all modern prosthetic pliance of the cross sectional area of interest of the vessel or
grafts are made from these materials.19 It was not until 1976 graft.
that Baird and Abbott postulated compliance mismatch as a
major factor affecting graft patency.5,6 Since then a consid- Longitudinal Compliance
erable amount of research has elucidated the nature of com- This technique is based on monitoring of pulse wave
pliance mismatch, but as yet has not led to production of a velocity (PWV) of blood down a given arterial pathway.23,24
more compliant graft.9,10,20 The elastic component of the wall plays an important role
in determining the velocity of propagation of a pulse wave.
Physical Properties of the Vessel Wall The Moert-Kortensweg equation (3) describes this relation-
Knowledge of the elastic properties of the arterial wall ship. In this equation E is Elastic module, h is wall thickness,
is essential to study the dynamics of the arterial system. R is radius and ∋ is density of blood.
During systole, with an increase in pressure the arterial wall
circumference increases, with return during diastole to its (3)
previous dimension. This function of the vascular wall is of
crucial physiological importance and due to a combination The compliance C can be computed from the above
of elastic and viscous components inherent to arterial tissue. principle in equation 4.
Solid materials have elasticity to varying extents as an
inherent mechanical feature and this is defined as the ratio (4)
of applied stress to resultant strain. Young’s modulus ex-
presses this quality of elasticity and is measured in units of This approach is based on the assumption that (1/2R)
dyne/cm2. Solid materials which regain their original dimen- is small and that the vessel or grafts are filled with viscous
sions when the stress is withdrawn are perfectly elastic, while liquid. Experimental evaluation of this methodology reveals
those which retain the entire deformation are plastic. The an error of 16-24% in computation of compliance, thus re-
property of viscosity distinguishes a fluid from a solid. When quiring a correction factor.25
stress is applied to a fluid it will undergo viscous flow. The Since the pulse pressure and flow pulse propagates
vessel wall exhibits properties of both an elastic solid and a along a vessel with the same velocity, arterial compliance
viscous fluid and thus can be most properly described as can therefore be indirectly measured by observing the flow
being visco-elastic. pulse with Doppler ultrasound. Values for the time taken
for this pulse to travel a unit length of the arterial pathway,
Noncompliance: The Silent Acceptance of a Villain 47
in the absence of reflection from the periphery, can then be requires recording of systolic and diastolic pressures as well
applied to equation 4 to give the average arterial compli- as noninvasively recording diameter over cardiac cycle.
ance of a section of artery over which the pulse was mea- A wide spectrum of techniques have been used to
sured. This technique requires a simultaneous use of two measure the circumferential compliance in vitro as well as
Doppler devices.26 To further complicate this method, in- in the clinical setting, although less successfully. These meth-
creased PWV due to generalized stiffening of an artery or odologies include electrical, optical, ultrasound, magnetic
local plaque formation cannot be distinguished and ac- resonance imaging and digital angiography.
counted for. As a result, this approach has now been over-
taken by recent noninvasive accurate assessments of cross- Assessment of Compliance Using Electrical Techniques
sectional compliance. Measurement of changes in diameter by placement of
electrical resistance probes on blood vessels was first reported
Circumferential Compliance in 1960.27 The principle of this technique is based on place-
The most popular and acceptable method of comput- ment of transducers on the arterial wall with continuous
ing compliance is from measurement of changes in diam- changes in diameter being electrically produced. The device
eter over a cardiac cycle. which utilizes a miniature differential transformer measures
The first approach for assessment of elasticity, and so changes in external diameter of blood vessels and was used
compliance, is based on measurement of pressure and di- in particular to map the mechanical properties at several
ameter curves over the cardiac cycle. In quasistatic compli- sites along the aorta and some of its major branches.28 This
ance slow inflation and deflation of the blood vessels gener- methodology is highly invasive and effectively could only
ate a pressure diameter curve. The arterial wall, being an be used intraoperatively. In addition, the use of the device
anisotropic material, has a nonlinear elastic behavior. The imposes mechanical restraint on the vessel because of the
incremental elastic modulus is calculated using equation 5. need to apply calipers or coils. Since a typical change in di-
ameter of a blood vessel is in the order of about 5%, the
(5) degree of distortion and frictional force caused by these in-
struments led to acceptably high errors in measurements
Einc is the incremental elastic module, R is radius, o on small blood vessels. Furthermore these instruments mea-
and i subscripts denote inner and outer radius respectively, sure external diameter only, giving no information about
( is Poisson ratio, )R and )P are change in radius and pres- internal diameter movement, and all of this is can further
sure respectively. be compounded by problems in identifying exactly the limit
The second approach is based on assessment of ex- of the external wall in a native artery.
cursions of diameter and pressure during a cardiac cycle and
is known as dynamic compliance. Since strain relates not Optical Techniques
only to the magnitude of stress but also to the rate at which The main principle of this technique is application of
it is applied, it is logical to look at the elastic property in the light to measure diameter of the vessel. The vessel of inter-
physiological setting of pulsatile flow. A potential problem est is placed in a laser scanning system; this consists of a
in simultaneous recordings of changes in diameter and pres- laser light and photocell detectors. In operation the laser tube
sure is the phase lag between the pressure and diameter emits a light source which after passing through appropri-
curves, which is caused by the viscous component of the ate objects sweeps along a particular axis at a known veloc-
vascular wall. A complex elastic modulus has therefore been ity. The presence of an artery in the beam path prevents light
defined to formulate this concept. from reaching a photocell during a particular time. Know-
ing this time and sweep velocity calculations are made to
(6) compute the outside diameter of then artery in continuous
mode.29 This system has been applied to assess the compli-
ance of canine carotid artery, femoral artery and prosthetic
Where is the elastic compo- grafts in vitro by placing the vessels in pulsatile flow cir-
cuits.30 The technique has reported accuracy of detection in
nent and is the viscous component. lm is the order of 13 ∝m changes in outer diameter of the artery.29
The disadvantage of this system is that it is restricted to ex-
the average circumference and qm the average cross section perimental use only. In addition, it detects only changes in
area, P is the amplitude of the applied stress and Dl/l is the outer diameter and gives no information regarding inner
strain, w is the angular frequency, m is viscosity and f phase wall movement, which may be different, particularly in thick-
lag between the pressure and diameter curves. ened vessels.
Changes in diameter and pressure over cardiac cycle
can be measured at selected points on the vessel of interest, Ultrasonic Techniques
giving a more complete picture of the viscoelastic proper- Ultrasound has been the favored method of compli-
ties of the arterial wall. Recording of the pressure in vivo, ance measurement over the past two decades. There are two
however, requires insertion of an arterial catheter with its ways in which ultrasound has been applied to assess the com-
attendant possible complications. In the clinical setting, pliance of blood vessels. The first measures the PWV along
quasistatic compliance is more widely used. This technique a segment while the other is based on the local distention
48 Tissue Engineering of Prosthetic Vascular Grafts
wave form of a local artery (local compliance).20 This latter This technique is applicable clinically as well as experimen-
method requires assessment of diameter of the artery under tally for assessment of compliance. The disadvantage of MRI
investigation at the onset of a cardiac cycle, and thus disten- is that in vascular diseases such as atherosclerosis, focal le-
tion during the cardiac cycle under local pulse pressure. A sions are formed; thus the compliance at one site may be
radio frequency data acquisition system linked to a com- very different from that at another. Further problems may
puter is required and M-mode echo images of the blood ves- occur in follow up studies to monitor the course of the dis-
sel will be displayed. The anterior and posterior walls are ease or response to therapy, since relocation of the original
identified manually or automatically. The radio frequency site of measurement is very difficult. MRI, therefore, while
signals from the ultrasound M-mode output over a cardiac useful in demonstrating accurately pathological disease, has
cycle are digitized and relayed to a wall tracking system31 limitations in these measurements.
(see Fig. 2.1). From these signals end-diastolic and end-sys-
tolic intraluminal diameters can be easily determined and Digital X-ray Techniques
thus the maximum changes in diameter or distention for Angiography, or radiographic imaging of blood ves-
each beat. Blood pressure can be detected noninvasively and sels, has a well established role as the gold standard in the
compliance computed. diagnosis of vascular disease and is widely used in clinical
Ultrasound has inherent inaccuracies relating mainly practice for obtaining high quality vessel images.37,38 Digi-
to problems in recognizing the exact inner or outer diam- tal X-ray angiography has the necessary temporal and spa-
eter of the blood vessel.32,33 In vivo studies of the inter- and tial resolution for volume blood flow measurement and es-
intraobserver variability reveal errors of 5% in measuring timation of diameter over cardiac cycles. Diameter estimates
static diameter, and 10-15% in measuring pulsatile diam- are either based on accurate vessel edge location or by den-
eter changes.34 Increased angle of insonation by the trans- sitometry, i.e., by integrating image brightness perpendicu-
ducer in the longitudinal axis will falsely increase diameter lar to the axis of the vessel.39-41 The latter technique and prin-
and its changes. Due to the tortuous nature of some vessels ciple yield a number proportional to the vessel’s area inde-
in vivo it is difficult to be exactly perpendicular to the vessel pendent of lumen shape, in both healthy and diseased ves-
axis. The variability introduced by this problem in measur- sels. This technique has been used for estimation of compli-
ing diameter can affect the quality of results especially in ance as well as blood flow in the vessel of interest.42,43 Mea-
looking at small caliber vessels and consequently small surement of vascular diameter and volume blood flow in
changes over the cardiac cycle. This uncertainly is partially vessels which are tortuous and which do not lie parallel to
overcome by the use of intravascular ultrasound, which has the imaging plane relies on accurate computation of the 3-di-
the major drawback, however, of being invasive.35 mensional (3D) path length of the vessel, X-ray magnifica-
tion and the angle between the vessel axis and the X-ray
Magnetic Resonance Imaging Techniques beam. These factors can be computed from accurate 3D re-
Magnetic resonance imaging (MRI) is a noninvasive construction of vascular geometry.42,44,45 The authors and
imaging modality that is rapidly gaining clinical acceptance, collaborators have described a novel technique of measure-
although widespread introduction has been delayed by its ment of vessel cross sectional area, and hence diameter, us-
expense. MRI has been used directly to measure regional ing densitometric methods applied to standard intraarterial
aortic compliance as well as total cardiac aortic compliance.36 digital subtraction angiograms.41,43 The technique is based
on image densitometry in which the integral intensity of ization, X-ray angiography is still the modality of choice for
the contrast bolus is computed along a profile perpendicu- critical morphological vascular studies. That X-ray angiog-
lar to the projection of the vessel axis. As illustrated in Fig- raphy has not been widely used for measuring blood flow is
ure 2.2, the true cross-section A is related to the densito- due in part, we believe, to the use of inappropriate algo-
metric measure a by: rithms for processing the imaging digital data.46,47 The
method does, however, have great potential, especially when
(7) combined with lower dose digital subtraction angiography
and the new nonionic contrast agents.48 Mini-puncture
The X-ray magnification factor M and angle ∗ between needles and catheters have led to increased safety of the tech-
the vessel axis and the X-ray axis are computed from the 3D nique and the equipment and expertise is available in most
reconstruction of the vascular configuration from two views. centres.49
The densitometric calibration constant K relates the image
gray value to the mass of iodine integrated along the X-ray Synthetic and Biological Grafts
path from X-ray focus to image. These were obtained from Prosthetic grafts fare well when used in the aortic or
data generated from 3D reconstructions of biplanar X-ray aortofemoral position, but are much less successful for
angiographic data.42 Although it requires vascular catheter- bypasses below the inguinal ligament, with the patient’s own
Table 2.1. Summary of results from literature for five years cumulative patency rates of infrainguinal reconstruction
with autogenous vein study
saphenous vein giving the best long term patency rates.2,50 infrapopliteal reconstructions is dismal.62,63 PTFE grafting
Because of dismal patency rates, prosthetics are not used in into the popliteal artery at the knee shows a slightly better 5
coronary bypass grafting. Primary 5 year patency rates for year patency rate of 40%.61-63 Recently, better patency rates,
autogenous vein bypasses range between 63-75%, and sec- even to the tibial level, have been reported with the use of
ondary patency rates between 80-83%, leading to limb sal- PTFE bypass grafting onto an interposition vein collar or
vage rates which range from 84-92% (Table 2.1).51-53 These patch which is placed onto the distal artery. In part this may
are good clinical results but, unfortunately, in up to 40% of be due to improvement of the compliance match between
patients autogenous vein may not be available or be inad- the host artery and the graft.64
equate because of coexisting disease such as varicose veins Compliance mismatch has an important role in graft
or previous venous thrombosis. failure. Various research workers have shown that compli-
In these circumstances a prosthetic graft is the only ance of biological conduit is significantly greater than that
possible option.3 Since 1950, prosthetic grafts have been used of prosthetic material; this compares well with patency rates
in a variety of locations for arterial reconstruction. Knitted for the two different graft materials (Table 2.2).51,54,63 It ap-
and woven Dacron grafts provided favorable results in high pears that as compliance mismatch increases, patency de-
flow large caliber, vessels but performed poorly in more chal- creases; linear regression analysis of this data shows a highly
lenging small caliber low flow conditions characteristic of significant correlation for this association, as shown in Fig-
infrainguinal bypasses, particularly to the below knee arter- ure 2.3. The patency data for the different grafting materials
ies. To date the most successful prosthetic grafts in this situ- used in the femoro-popliteal position used in this study were
ation have been expanded polytetrafluoroethlylene (PTFE) obtained from several recent large studies and compliance
and the Dardik biograft or human umbilical vein tanned data from a study performed by Abbot et al in Bos-
with glutaraldehyde.54,55 The human umbilical vein graft was ton.51,54,55,63 Finally, prosthetic grafts have poor dynamic
introduced at roughly the same time as PTFE but never compliance profiles in comparison to arteries with no in-
gained broad acceptance, primarily because of a propensity crease in compliance occurring at low pressure, an impor-
for dilatation and aneurysm formation as early as 2 years tant physiological response to shock (Fig 2.4).20
after implantation, occurring in up to 57% of the
grafts.54,56,57 This phenomenon of graft dilatation is com- Causes of Graft Failure
mon to all biografts and is attributed to a combination of Causes of graft failure are complex and include tech-
deterioration of collagen crosslinks with time, and pro- nical failure, poor selection, compliance mismatch, primary
teolytic digestion by host enzymes almost certainly medi- thrombotic failure related to low flow environments, devel-
ated by an immune response.58 In vitro testing of 2 types of opment of intimal hyperplasia, late failure due to graft gen-
biografts, a bovine carotid and the human umbilical vein
graft, with long term perfusion with proteolytic enzymes,
demonstrated changes in the structural integrity of the graft. Table 2.2. Summary of results from literature relationship
This was detected by a degree in compliance, thus providing between compliance vs. patency rate.51,54,63
experimental data for the role of proteolytic digestion in
these grafts failures.59 Despite this, clinical experience with Graft type Compliance Patency %
the human umbilical vein gives a 5 year patency with ap- (%mm Hg x 10-2)
proximately 60% for femoral popliteal grafting with up to
80% in the above knee location.60,61 Thus these grafts per- Host artery 5.9 ± 0.5
form clinically well in the short term and have been recom- Saphenous vein 4.4 ± 0.8 75
mended for use in patients with a short life expectancy. Umbilical vein 3.7 ± 0.5 60
Results achieved with PTFE bypass grafting are less Bovine heterograft 2.6 ± 0.3 59
Dacron 1.9 ± 0.3 50
satisfactory particularly so onto the below knee tibial ves-
PTFE 1.6 ± 0.2 40
sels. On average the long term patency rates (> 3 years) in
◆ Dacron
4 ▼ PTFE
▲ Saphenous vein
3
1
30 40 50 60 70 80
Patency (%)
Noncompliance: The Silent Acceptance of a Villain 51
0
40 60 80 100 120
eration and late failure due to progression of native vessel velocity gradients and turbulence. As a consequence of these
arterial sclerosis. More than one of these factors may be im- mechanical effects, vibratory weakening of the arterial wall
plicated in graft failure. Much research has been focused on has been postulated as a cause of endothelial damage, inti-
these various factors, with particular emphasis on reducing mal hyperplasia and even anastomotic aneurysm formation.
the thrombogenicity of grafts either by modifying the lumi-
nal surface or by endothelial cell seeding. Much less effort Anastomotic Compliance Mismatch
has been directed into the role of compliance in graft fail- Anastomotic compliance mismatch refers to the varia-
ure. Possibly, this is because of limitations in materials and tion in compliance occurring specifically at the anastomotic
material technology. There is much clinical data which em- site. The change in compliance from native artery and pros-
phasizes the importance of compliance mismatch and cer- thetic material is not monotonic because the creation of an
tainly it is established experimentally that current anastomosis always generates a focal decrease in diameter
noncompliant grafts owe their inferior performance to lack and a drop in compliance. This is partially determined by
of distensibility when compared to native arteries.6 It is most the elasticity, or rather rigidity, of the suture material, and
important to consider compliance mismatch as composed partly by the surgical technique, depending on whether a
of two major parts, namely tubular and anastomotic. continuous or interrupted anastomosis is used. Interrupted
anastomotic suture techniques give more compliant anas-
Tubular Compliance tomoses. There is also a paradoxical increase in compliance
Tubular compliance mismatch refers to the imparity of about 50% which occurs within a few millimeters on ei-
of elasticity between the prosthetic conduit and the native ther side of the suture line (Fig. 2.5). This characteristic is
artery. Essentially this results in an adverse hemodynamic known as the para-anastomotic hypercompliance zone
effect and reduced distal perfusion, as was previously dis- (PHZ).20,67,68 It has been hypothesized that PHZ may be re-
cussed. A compliant wall acts as an elastic reservoir absorb- sponsible for the intimal hyperplasia which characteristi-
ing energy during systole, which is released during diastole. cally develops at the same area as this hyper-compliance.
A rigid vessel wall consequently diminishes the pulsatile Thus mismatch of elastic properties around the anastomo-
component of the diastolic recoil, thus reducing the energy sis may act to promote intimal hyperplasia in at least 3 dif-
available for distal perfusion. ferent ways:
Impedance is the term given to resistance to pulsatile 1. Compliance mismatch between the artery and graft
flow. Change in impedance occurs at the interface between may lead to a region of excessive mechanical stress,
a compliant artery and noncompliant graft, and this results possibly resulting in wall injury, a major event in the
in propagation of less than 60% of the pulsatile energy.65 initiation of intimal hyperplasia;6,69,70
Optimal organ perfusion depends on pulsatile flow, and it 2. Cyclic stretching has a positive influence on replica-
has been shown that change from pulsatile to steady state tion of vascular smooth muscle cells and produc-
flow causes peripheral resistance to increase by 10%.66 Wave tion of extracellular matrix.70,71 Experimentally, it has
reflection of the graft artery interface also leads to increased been demonstrated that vascular smooth muscle cell
52 Tissue Engineering of Prosthetic Vascular Grafts
start to produce extracellular matrix and to repli- finding of free calcium concentrations proportional to the
cate when they are subjected to high levels of disten- shear rate suggests an important role of calcium in the sig-
sion.72 Also, animal studies have confirmed that in- nal transduction pathway.81 Shear rate dependent release of
timal thickening is associated with increased growth factor is reflected by a varying expression of mul-
tangential stress;73 tiple growth factor-related genes when wall shear rates are
3. Flow studies show that a sudden increase in compli- changed.82 A possible mechanism underlying the increased
ance is associated with enhanced particle residence expression of growth factor genes may be a recently described
time, flow separation and stasis leading to low shear shear rate-induced transcription factor which may not only
stress conditions.74,75 Also, from animal studies it is regulate the expression of platelet derived growth factor
known that high levels of shear stress have a delete- (PDGF) genes but also of tissue plasminogen activator (tPA),
rious effect on endothelial cells but, most impor- intracellular adhesion molecule 1 (ICAM-1) and transform-
tantly, there is also mounting evidence that intimal ing growth factor-b1 (TGF-!1).83 A shear stress dependent
thickening occurs in areas of low shear rate. An ex- release of PDGE-! mRNA has also been observed in cul-
ample of this phenomenon in humans is of the sud- tured human umbilical vein cells.84 These findings begin to
den increase in compliance found in the carotid bulb illuminate the link between mechanical effects and changes
where the above hemodynamic changes occur. Since and the development of intimal hyperplasia in human ves-
this is a site with predilection for the occurrence of sels and bypass grafts.
atherosclerosis, one could postulate that increased
compliance with prolonged particle residence time, Development of a Graft
flow separation and low shear stress conditions can with Better Compliance
cause damage, allowing initiation of arteriosclerosis The ideal characteristics of a graft are biocom-
in man. patability, a nonthrombogenic surface, physical durability,
elastic or compliant properties similar to those of a native
Signal Transduction Pathways and Flow artery, resistance to infection and ease of implantation. Ad-
Flow is an important modulator of vascular structure vances in manufacture of biomaterials and tissue engineer-
and function, probably mediated through the effects of the ing are leading to developments in the manufacture of more
function on the endothelial cell.76,77 Pathways by which flow compliant grafts. What is not known, however, is the range
acts on the vessel wall are poorly defined, but an essential of compliance required to give the ideal match between the
role is attributed to the endothelial cell.78-80 Nitric oxide graft and the host artery. Arteries change with age with loss
(NO) plays an important role in this pathway, and it has of elastin and consequent reduction of elasticity and thus
been shown that specific potassium channel antagonists can compliance. When the ravages of arteriosclerosis with me-
block flow mediated vasodilatation and nitric oxide release, dial calcification occurring as a result of age are added to
suggesting that the signal transduction pathway underlying these effects, the compliance of the host artery in a patient
flow-mediated vasodilatation may be the activation of the requiring a bypass may be dramatically reduced (Fig. 2.6).20
potassium channel on the endothelial cell membrane.73 The Thus the aim should be to develop a graft which is pulsatile,
Noncompliance: The Silent Acceptance of a Villain 53
4
Compliance
0
30 40 50 60 70 80
Age (years)
but it may even be detrimental to have a graft which is too tion of artificial blood vessels with specific compliance char-
compliant. Study of typical compliances in aged and dis- acteristics (Fig. 2.7).10 In this process the graft material is
eased human arteries, coronary, femoral, popliteal and tibial, made using either a single shot method (Fig. 2.8)9 or a
is required before the ideal graft can be designed. prepolymer technique (Fig. 2.9).9 In a single shot method,
Development of more compliant grafts is currently all reactants are added simultaneously and the final struc-
occurring, either using biomaterials or by tissue engineer- ture is thus dependent on the relative reactivity of the for-
ing of vascular grafts. An example of developments in mulation. In the prepolymer method the ingredients are
biomaterials is the manufacture of grafts using polyurethane. added sequentially, allowing close engineering and design
Grafts have been made from this material for quite some of the final structure. Using a coagulation precipitation tech-
time and the major problem has been biodegradability and nique, porosity of the graft can be varied to form either mi-
subsequent aneurysm formation. A novel formulation of a cro- or macroporous structures. By varying the quantity and
polyurethane graft, namely CronoFlex (Poly Medica Indus- type of chemical groupings within the polyurethane polymer,
tries, Tarvin, Cheshire, UK) has recently allowed produc- it is possible to manipulate the level of crosslinking and thus
54 Tissue Engineering of Prosthetic Vascular Grafts
control mechanical properties. Thus a graft can be designed A further important area in design of such a biomate-
and modulated, varying not only the characteristics of the rial is in selection of optimal luminal characteristics. It is
polymer but also the degree of “sponginess” of the graft wall possible to design the lumen of the graft so that it is inher-
by modulating the size of the pores or bubbles in the wall of ently nonthrombogenic by incorporation, for example, of
the graft. This allows design of a graft to a specific compli- heparin or nonthrombogenic polymers. The alternative ap-
ance. This particular formulation of polyurethane grafts, proach is to make the lumen more attractive for endothelial
known colloquially as the “bubble graft,” has demonstrably cell adhesion and seeding. A simple example of such a meth-
higher compliance characteristics than others (Fig. 2.10).10 odology is the CronoFlex graft lumen, which is constituted
By modulation of the outer wall of the graft using various of many pits and valleys. Experimental evidence suggests
strategies to strengthen it, kinking as the graft crosses the that endothelial cells adhere better and are more resistant to
knee joint can be avoided. Extensive animal studies have the effects of flow shear stress because they can shelter within
shown no tendency for the CronoFlex graft to develop an- the pits, and proliferate upwards to endothelialize the graft
eurysms. (Fig. 2.11).85
Noncompliance: The Silent Acceptance of a Villain 55
Tissue engineering holds much promise for the de- gin, which are said to be nonimmunogenic. Little data is
velopment of more compliant grafts. The relative roles of available as yet on the compliance characteristics of these
collagen, elastin and smooth muscles in the mechanical char- materials. Certainly this approach would hold the greatest
acteristics of an artery are now well understood. Experimen- promise of developing a graft with the necessary visco-elas-
tally, the development of arterial grafts using combinations tic properties comparable to those of human arteries.
of smooth muscle cells, collagen and endothelial cells is pos-
sible. Major problems persist in terms of the source of these The Future
cells. Ideally these should be from the patient, but certainly In the very near future it will be possible to move away
in peripheral vascular grafting the time and logistics involved from the use of the rigid materials, mostly Daron and PTFE,
would mitigate against the widespread use of this new tech- which are currently used in bypass grafting. Initially, pros-
nology. The important role of the immune response in even- thetic biomaterials with suitably designed and compatible
tual biodegradability means that such tissue engineered compliance characteristics will be used, with clinical trials
grafts made from nonautologus cells would require the use expected to begin within 2 years. The further addition of
of either immunosuppressive therapy, or cells of fetal ori- endothelial cell seeding to these more compliant grafts holds
56 Tissue Engineering of Prosthetic Vascular Grafts
7. Kinley CE, Paasche PE, MacDonald AS. Stress at vascular 29. Brant AM, Rodgers VGJ, Borovetz HS. Measurement in
anastomosis in relation to host artery: Synthetic graft di- vitro of pulsatile arterial diameter using a helium-neon
ameter. Surgery 1974; 75(1):28-30. laser. J Appl Physiol 1987; 62:679-83.
8. Madras P, Ward C, Johnson W, Singh P. Anastomotic 30. Megermann JM, Hasson JE, Warnock D, L’Italien GJ,
hyperplasia. Surgery 1981; 90(5):992-3. Abbott WM. Noninvasive measurments of nonlinear ar-
9. Reed AM, Potter J, Szycher M. A solution grade biostable terial elasticity. American Journal of Physiology 1986;
polyurethane elastomer: CronoFlex AR. Journal of 250:H181-8.
Biomaterials Applications 1994; 8:210-36. 31. Hoeks APG, Brands PJ, Smeets FAM, Reneman RS. As-
10. Edwards A, Carson RJ, Bowald S, Quist WC. Develop- sessment of the distensibility of superficial arteries. Ul-
ment of a microporous compliant small bore vascular trasound Med Biol 1990; 16(2):121-8.
graft. Journal of Biomaterials Applications 1995; 32. Jaffe CC. Doppler applications and limit of the method.
10:171-87. Clin Diagn Ultrasound 1984; 13:1-10.
11. Harvey W. (1628) De Motu Cordis, Frankfurt: William 33. Beach KW. The evaluation of velocity and frequency ac-
Fitzer. Translated as movement of the heart and blood in curacy in ultrasound duplex scanners. Journal of Vascu-
animals by Franklin, K.J. (1957). Oxford: Blackwell Sci- lar Technology 1990; 14(5):214-20.
entific Publications. 34. Hansen D, Bergqvist D, Mangell P et al. Non-invasive
12. Hales S. (1733) Statical essays: containing haemastaticks, measurement of pulsatile vessel diameter change and elas-
reprinted 1964, No. 22, History of Medicine Series, Li- tic properties in human arteries—a methodological study.
brary of New York Academy of Medicine. New York: Clin Physiol 1993; 13:631-43.
Hafner Publishing. 35. Hansen ME, Yual EK, Megerman J. et al. In vivo deter-
13. Frank O. Die Theorie der Pulswelle. Z Biol 1927; mination of human arterial compliance: Preliminary in-
85:91-130. vestigation of a new technique. Cardiovascular &
14. Womersley JR. Oscillatory flow in arteries: The con- Interventional Radiology 1994; 17(1):22-6.
strained elastic tube as a model of arterial flow and pulse 36. Mohiaddin RH, Longmore SR. MRI studies of atheroscle-
transmission. Phys Med Biol 1957; 2:313-23. rotic vascular disease: Structural evaluation and physi-
15. McDonald DA. The realtion of pulsatile pressure to flow ological measurements. Brit Med Bull 1989; 45:968-90.
in arteries. J Phsysiol 1955; 127:533-52. 37. Pond GD, Osborne RW, Capp MP et al. Digital subtrac-
16. Carrel A. La technique operatoire des anastomoses tion angiography of peripheral vascular bypass procedures.
vasculaires et la transplantation des visceres. Lyon Med AJR 1982; 2:279-81.
1902; 98:859-64. 38. Guthaner DF, Wexler L, Enzmann DR et al. Evaluation
17. Goyanes J. Nuevos trabajos de cirugia vascular, of peripheral vascular disease using digital subtraction an-
substitucion plastica de las arterias por las venas or giography. Radiology 1983; 147(2):393-8.
arterioplastia venosa, applicada, como nuevo metodo, al 39. Brown BG, Bolson E, Bolson E, Frimer M, Dodge HT.
tratamiento de los aneurismas. Siglo Med 1906; 53:546-9. Quantitative coronary arteriography. Estimation of dimen-
18. Voorhees AB, Jaretzke AL, Blakemore AH. The use of sions hemodynamic resistance and atheroma mass of
tubes constructed from Vinyon-N cloth in briding arte- coronary artery lesions using the arteriogram and digital
rial defects. Ann Surg 1952; 135:332-6. computation. Circulation 1977; 55:329-37.
19. Turner RJ, Sawyer PN. Modern Vascular Grafts. In: Vas- 40. Reiber JHC, Kooijman CJ, Slager CJ. Computer assisted
cular Graft Development: An Industrial Perspective. New analysis of the severity of obstructions from coronary
York: McGraw-Hill,; 1985:75-103. cineangiograms: A methodological review. Automedica
20. Schmitz-Rixen T, Hamilton G. Compliance: A critical 1984; 5:219-38.
parameter for maintenance of arterial reconstruction? In: 41. Hawkes DJ, Colchester ACF, de Belder MA et al. The mea-
Greenhalgh RM, Hollier LH, eds. The maintenance of surement of absolute lumen cross sectional area and lu-
arterial reconstruction. London: WB Saunders, 1991:23-43. men geometry in quantitative angiography. In: Todd-
21. Murgo JP, Westerhof N, Giolma JP, Altobelli SA. Aortic Pokropek AE, Viergever MA, eds. Medical images: For-
input impedance in normal man: Relationship to pres- mation, Handling and Evaluation. Nato ASI Series.
sure wave forms. Circulation 1980; 62:105-16. Heidelburg: Springer-Verlag, 1992:609-26.
22. O’Rourke MF. Systolic blood pressure: Arterial compli- 42. Hawkes DJ, Seifalian AM, Colchester ACF, Iqbal N,
ance and early wave reflection and their modification by Hardingham CR, Bladin CF, Hobbs KEF. Validation of
antihypertensive therapy. J Hum Hypertens 1989; 3:47-52. volume blood flow measurements using three-dimensional
23. O’Rourke M. Arterial compliance and wave reflection. distance-concentration functions derived from digital x-
Arch Mal Coeur 1991; 84(III):45-8. ray angiograms. Investigative Radiology 1994; 29:434-42.
24. Lehmann ED. Elastic properties of the aorta. The Lancet 43. Seifalian AM, Hawkes DJ, Giudiceandra A. et al. A novel
1993; 342:1417. technique of blood flow and compliance measurement
25. Bergel, DH. The visco-elastic properties of the arterial using digital subtraction angiography. In: Greenhalgh RM,
wall. Ph.D. Thesis, London University, 1960. ed. Vascular imaging for surgeons. London: W.B.Saunders
26. Bertram CD. Ultrasonic transit-time measurement for Company Ltd, 1995:51-70.
arterial diameter measurement. Med Biol Eng Comp 1977; 44. Hawkes DJ, Hardingham CR, Seifalian AM et al. Recent
15:489-99. advances in extracting quantitative information from x-
27. Patel DJ, Mallos AJ, Fry DL. Aortic mechanics in the liv- ray angiographic data. Innov Tech Biol Med 1992;
ing dog. J App Physiology 1961; 16:283-99. 13:99-107.
28. Peterson LH, Jenesen RE, Parnell J. Mechanical proper- 45. Hawkes DJ, Mol CB, Colchester ACF. The accurate 3-D
ties of arteries in vivo. Circulation Research 1960; reconstruction of the geometric configuration of vascular
8:622-39. trees from x-ray recordings. In: Guzzardi R, ed. Physics
58 Tissue Engineering of Prosthetic Vascular Grafts
and engineering of medical imaging. NATO ASI. Holland: 66. Giron F, Birtwell WS, Soroff HS, Deterling RA. Hemody-
Martinus Nijhoff Publications, 1987:250-258. namic effects of pulsatile and nopulsatile flow. Arch Surg
46. Seifalian AM, Hawkes DJ, Colchester ACF, Hobbs KEF. 1966; 93(5):802-10.
A new algorithm for deriving pulsatile blood flow wave- 67. Abbott WM, Megermann JM. Adaptives responses of ar-
forms tested using simulated dynamic angiographic data. teries to grafting. Journal of Vascular Surgery 1989;
Neuroradiology 1989; 31:263-9. 9:377-9.
47. Du Boulay GH, Brunt J, Colchester A et al. Volume flow 68. Hasson JE, Megermann JM, Abbott WM. Increased com-
measurement of pulsatile flow by digitised cine angiogra- pliance near vascular anastomosis. Journal of Vascular
phy. Acta Radiologica 1987;Suppl 13:59-62. Surgery 1985; 2:419-23.
48. Bettman MA, Morris TW. Recent advances in contrast 69. Bassiouny HS, White S, Glagov S et al. Anastomotic inti-
agents. RCNA 1986; 24(3):347-57. mal hyperplasia: Mechanical injury or flow induced. Jour-
49. Cope C. Minipuncture angiography. RCNA 1986; nal of Vascular Surgery 1992; 15:708-17.
24(3):359-67. 70. Clowes AW, Clowes MM, Fingerle J, Reidy MA. Kinetics
50. Crawford ES, Bomberger RA, Glaeser DH et al. Aortoiliac of cellular proliferation after arterial injury. Laboratory
occlusive disease: Factors influencing survival and func- Investigation 1989; 60:360-4.
tion following reconstructive operation 0ver a twenty-five 71. Sottiurai VS, Sue SL, Feinberg EL II et al. Distal anasto-
year period. Surgery 1981; 90(6):1055-67. motic intimal hyperplasia: Biogenesis and etiology. Eur J
51. Taylor LM, Edwards JM, Porter JM. Present status of of Vasc Endovasc Surg 1988; 2:245-56.
reversed vein bypass grafting: Five years result of a mod- 72. Predel HG, Yang Z, von Segesser L et al. Implication of
ern series. J Vasc Surg 1990; 11(2):193-205. pulsatile stretch on growth of saphenous vein and mam-
52. Bergamini TM, Towne JB, Bandyk DF. Experience with mary artery smooth muscle. Lancet 1992; 340:878-9.
in situ saphenous vein bypasses during 1981 to 1989: 73. Cooke JP, Rossitch E, Andon NA et al. Flow activates a
Determinant factors of long term patency. J Vasc Surg endothelial potassium channel to relaese an endogenous
1991; 13(1):137-47. nitrovasodilator. Journal of Clinical Investigation 1991;
53. Donaldson MC, Mannick JA, Whittemore AD. Causes of 88:1663-71.
primary graft failure after in situ saphenous vein bypass 74. Stewart S, Lyman DJ. Effects of vascular graft/natural ar-
grafting. J Vasc Surg 1992; 15(1):113-8. tery compliance mismatch on pulsatile flow. J Biomech
54. Dardik H, Ibrahim IM, Dardik I. Evaluation of glutaral- 1992; 25:297-310.
dehyde tanned human umbilical cord vein as a vascular 75. Ojha M, Cobbold RS, Johnston KW. Haemodynamics of
prosthesisfor bypass to the popliteal, tibial and peroneal a side to end proximal arterial anastomosis model. Jour-
arteries. Surgery 1978; 83(5):577-88. nal of Vascular Surgery 1993; 17:646-55.
55. Walden R, L’Italien G, Megerman J. Matched elastic prop- 76. Kamiya A, Ando J, Shibata M, Masuda H. Roles of fluid
erties and successful arterial grafting. Arch Surg 1980; shear stress in physiological regulation of vascular struc-
115:1166-1169. ture and function. Biorheology 1988; 25(1-2):271-8.
56. Dardik H, Ibrahim IM, Sussman B. Biodegradation and 77. Hofstra L, Bergmans DC, Leunissen KM, Hoeks AP,
aneurysm formation in umbilical vein grafts: Observations Kitslaar PJ, Tordoir JH. Prosthetic arteriovenous fistulas
and a realistic strategy. Ann Surg 1984; 199:61-8. and venous anastomotic stenosis: influence of a high flow
57. Hasson JE, Newton WD, Waltman MD et al. Mural velocity on the development of intimal hyperplasia. Blood
degenaration in the glutaraldehyde-tanned umbilical vein Purif 1996; 14(5):345-9.
graft: incidence and implications. J Vasc Surg 1986; 78. Luscher TF. Endothelial control of vascular tone and
4:243-50. growth. Clin Exp Hypertens 1990; 12:897-902.
58. Mattila SP, Fogarty TJ. Antigenicity of vascular hetero- 79. Luscher TF. Imbalance of endothelium-derived relaxing
graft. J Surg Res 1973; 15:81-90. and contracting factors. A new concept in hypertension.
59. Hamilton G, Megerman J, L’Italien GJ et al. Prediction Am J Hypertens 1990; 3:317-30.
of aneurysm formation in vascular grafts of biologic ori- 80. Koo E, Gotlieb AI. Endothelial stimulation of intimal cell
gin. J Vasc Surg 1988; 7(3):400-8. proliferation in a porcine aortic organ culture. American
60. McCollum C, Kenchington G, Alexander C. PTFE or HUV Journal of Pathology 1989; 134:497-503.
for femoro-popliteal bypass: A mnulti-center trial. Eur J 81. Shen J, Luscinskas FW, Conolly A et al. Fluid shear stress
Vasc Surg 1991; 5(4):435-43. modulates cytosolic free calcium in vascular endothelial
61. Aalders GJ, van Vroonhoven TJM. PTFE versus HUV in cells. American Journal of Physiology 1992; 31:c384-c390
above knee femoro-popliteal bypass. Six year results of a 82. Taubmann MB, Rollins BJ, Poon M et al. JE mRNA
randomized clinical trial. J Vasc Surg 1992; 16:816-23. accunulates rapidly in aortic injury and in platlet-derived
62. Witthermore AD, Kent KC, Donaldson MC. What is the growth factor-stimulated vasular smooth muscle cells.
proper role of polytretrafluoroethlene grafts in Circulation Research 1992; 70:314-25.
infrainguinal reconstruction? J Vasc Surg 1998; 83. Resnick N, Collins T, Aktinson W et al. Platlet-derived
10(3):299-305. growth factor beta chain contains a cis-acting fluid shear-
63. Veith FJ, Gupte SK, Ascer E et al. Six-year prospective stress-responsive element. Proc Natl Acad Sci USA 1993;
muticenter randomized comparison of autologous saphe- 90:4591-5.
nous vein and expanded polytetrafluoroethylene graft in 84. Hsieh HJ, Li NQ, Frangos JA. Shear stress increases en-
infrainguinal arterial reconstruction. J Vasc Surg 1986; dothelial-platelet-derived growth factor mRNA levels.
3(1):104-14. American Journal of Physiology 1991; 260:H642-H646.
64. Beard JD, Benveniste GL, Miller JH, Baird RN, Hoskins 85. Giudiceandra A, Seifalian AM, Krijgsman B, Hamilton G.
PR. Haemodynamics of the interposition vein cuff. Br J Effect of prolonged pulsatile shear stress in vitro on en-
Surg 1986; 73:823-5. dothelial cell seeded PTFE and compliant polyurethane
65. Strandness DE, Summer DS. Hemodynamics for Surgeon. vascular grafts. Eur J Vasc Endovasc Surg 1998;
New York: Grune &Stratton, Inc. Pub., 1975. 15:147-154.
PART II
Biolized Prostheses: Surface Healing
CHAPTER 3
Endothelial Cell Seeding: A Review
Steven P. Schmidt, Gary L. Bowlin
Introduction
T he field of Tissue Engineering offers the promise of further elucidating and clarifying the
interactions between endothelial cells and smooth muscle cells. This understanding is
central to the creation of a successful small diameter vascular graft—a goal which has here-
tofore eluded researchers. Small diameter vascular grafts are defined as being less than 6 mm
in internal diameter. These grafts fail in patients from thrombosis and intimal hyperplasia,
processes which may, at least in part, originate from aberrant endothelial and smooth muscle
cell interactions. Clarification of the biology of these two cell types and their interactions is
critical in order to develop therapeutic solutions to the problems of small vessel prosthetic
grafting. These solutions will also undoubtedly involve further elucidation of the roles of
extracellular matrix in endothelial cell and smooth muscle cell biology, as well as of the
responsiveness of these cells to a variety of cytokines.
An endothelial cell lining does not naturally develop on the luminal surfaces of pros-
thetic vascular grafts in humans.1,2 This is in contrast to other animal species that have been
studied in which neoendothelialization of graft luminal surfaces occurs by endothelial cell
proliferation from perianastomotic artery, the microvessels of graft interstices, or circulat-
ing progenitor endothelial cells.3 Because endothelial cells release molecules that modulate
coagulation, platelet aggregation, leukocyte adhesion and vascular tone, the absence of these
cells lining prosthetic grafts, in combination with the inherent hostility of the biomaterial/
blood contacting surface, predisposes these grafts to platelet deposition leading to throm-
bosis. Although the precise mechanisms of the genesis of anastomotic intimal hyperplasia
are still being defined, endothelial cell/smooth muscle cell dysfunctions are thought to be
involved.
Early investigators in the field of small-diameter graft development sought to pro-
mote graft endothelialization by transplantation of autologous endothelial cells onto vascu-
lar grafts prior to their implantation—a process that became known as endothelial cell seed-
ing. The hypothesis underlying this research effort was really quite simple—that is, by pro-
moting the establishment of the patient’s own endothelial cells on the blood-contacting
surface of a vascular prosthesis, a “normal” endothelium-lined neointima would form on
the graft whose biology would counteract rheologic, physiologic, and biomaterial forces
promoting graft failure. In retrospect this simple hypothesis seems deceptively naive.
A review of the history of endothelial cell seeding research has been published which
details early animal and clinical studies.4 The results of these early animal studies were really
quite promising. This early optimism has been tempered, however, by clinical results that
have often been disappointing. It is clear that modifications of traditional methodologies to
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
62 Tissue Engineering of Prosthetic Vascular Grafts
promote endothelialization of vascular grafts are needed if ing and transplantation efficiencies in an appropriate time
endothelial cell transplantation is to be technically success- frame for clinical practicality. We will review briefly the his-
ful. What is even more important, however, is that academi- tory of these studies and then describe the novel technology
cians and physician researchers and investigators have grown of electrostatic cell seeding, which we feel offers a solution
to appreciate the complex biology of the cells forming the to many of the deficiencies of other approaches for trans-
vascular wall, and that technically successful transplantation planting cells onto vascular prostheses.
of endothelial cells does not necessarily presume a positive
outcome clinically in terms of vascular graft performance. Autologous Endothelial Cell Harvesting
The simple hypotheses of early endothelial cell seeding stud- Before endothelial cell seeding or transplantation can
ies have led to investigations providing enormous insights occur, autologous endothelial cells must be harvested from
into the biology of vascular wall cells, as well as stimulating an “appropriate, autologous” source. Sources of endothelial
suggestions for alternative therapeutic avenues that may uti- cells most often reported have been:
lize concepts and techniques of endothelial cell seeding. 1. Nonessential vessels (i.e., saphenous vein); and
The purpose of this chapter is to summarize the his- 2. Omental or subcutaneous adipose tissue (i.e., mi-
toric context and current base of knowledge regarding many crovascular endothelial cells).
of the technical issues relevant to endothelial cell seeding. From the nonessential vessels, endothelial cells have
Special reference will be made to recent exciting research been harvested by two techniques:
methodologies designed to promote endothelial cell attach- 1. Mechanical scraping;13 and
ment to prosthetic graft materials—a technology known as 2. Enzymatic digestion.14,15
electrostatic endothelial cell seeding. The wealth of animal Mechanical scraping uses an abrasive action to remove
studies has been reviewed elsewhere4 and will not be de- the endothelial cells from the vascular wall, which leads to
tailed here. Subsequent chapters in this text will summarize endothelial cell damage as well as the potential for signifi-
clinical outcomes. cant contamination with smooth muscle cells.12,13 The over-
all efficiency of endothelial cell harvest from autologous ves-
Historic Context sels by mechanical methods is < 75%.14 Enzymatic harvest-
The origin of the concept for the vascular prosthesis ing of cells from the vascular tissue requires collagenase or
is credited to Voorhees, Jaretski, and Blakemore, who pre- trypsin digestion to remove the endothelium.16-18 This en-
sented the first published hypothesis on synthetic tubes as zymatic digestion process can create damage to cellular pro-
replacements for natural blood vessel deficits in 1952.5 The teins, which in turn affects cell viability and attachment po-
first clinical applications of this hypothesis were published tential to a vascular prosthetic surface.19,20 The overall effi-
in 1954 and showed that the prosthetic tubes could be used ciency of endothelial cell harvest from autologous vessels
in the arterial setting.6 This publication prompted a race to using the enzymatic harvesting technique varies between
develop the best material for use as a prosthetic vascular graft. 80-100% of the available endothelial cells.14
The two designs of large diameter vascular grafts (> 6 mm Due to the limited availability of nonessential autolo-
I.D.) which are used successfully today clinically are made gous vessels in patients, the ready supply of endothelial cells
of Dacron or expanded polytetrafluoroethylene (ePTFE). harvested by either of the previously mentioned techniques
Patency rates for small diameter (< 6 mm I.D.) vascular pros- is necessarily limited. This limitation has prompted investi-
theses configured from these same materials are unaccept- gators to seek alternative sources which could supply a greater
able when utilized clinically, however, due to acute throm- number of endothelial cells. The alternative sources which
bus formation7,8 and chronic anastomotic hyperplasia9-11 have been most intensively studied include the mesothelium
which consequently prevent clinical usage. Malcolm Her- and the microvasculature, each of which offers the potential
ring, M.D., introduced the hypothesis underlying the tech- for derivation of large cell numbers. Microvascular endot-
nology of endothelial cell seeding in 1978 in an attempt to helial cells can be harvested from microvessels (arterioles,
increase the patency rate of small caliber prosthetic grafts. capillaries, and venules) found in adipose tissue.16,21,22 The
Herring proposed that if these small diameter vascular grafts overall efficiency of isolation of endothelial cells from
could be utilized successfully, they could be of enormous microvessels utilizing enzymatic techniques has been re-
clinical importance in peripheral and coronary bypass graft- ported to be approximately 84% ± 5%.23
ing procedures.12 Tissue culture of endothelial cells offers an alternative
Endothelial cell seeding since its inception has been approach to derive large numbers of cells for transplanta-
largely experimental, with significant technical success in tion from a small inoculum of harvested cells. The tissue
animal models. The technology has not been translated suc- culture approach was first reported by Graham et al.24 Jaffee
cessfully to any extent thus far published in human vascular et al17 initially reported that 92 h were required for an en-
surgery. Surgeons have long expressed concerns about many dothelial cell culture to double in number. However, im-
of the technical issues which must be overcome in order to provements in cell culturing techniques have reduced that
transplant endothelial cells successfully in a relevant time time to approximately 24 h using commercially available
frame for human surgery. Moreover, the long term benefits media (Clonetics Corporation. Unpublished data). Thus,
of a transplanted endothelial cell lining on graft patency in relatively large numbers of endothelial cells can be obtained
patients has not been demonstrated. from low derived inocula of cells in a time span of days to
Many research efforts have focused upon weeks in culture. A number of valid issues with tissue cul-
methodologic issues in an effort to maximize cell harvest- ture have been voiced, however. The exposure of endothe-
Endothelial Cell Seeding: A Review 63
lial cells in culture to an undefined serum (e.g., fetal bovine Hydrostatic seeding techniques use a pressure differ-
serum) as a part of the tissue culturing media presents the ential (internal pressure26,30 or external vacuum31) to force
opportunity for both genotypic and phenotypic modula- harvested endothelial cells resuspended in aliquots of hep-
tion.25 In addition, multiple passages of cells in culture and arinized autologous blood onto the luminal surface of
media changes create vulnerability to cultures becoming microporous graft materials. Experience has revealed at least
contaminated. three major limitations of the hydrostatic seeding techniques.
The first is that this technique is not adequate for fully hep-
Endothelial Cell Seeding Techniques arinized patients due to the graft porosity. In heparinized
Numerous methodologies for endothelial cell trans- patients, heparin interacts with the antithrombin III mol-
plantation onto prosthetic surfaces have been reported in ecule which inhibits the action of thrombin to form subse-
the literature. These investigations can be differentiated and quent clots within the graft pores, which is necessary to main-
summarized by virtue of the unique physical force utilized tain hemostasis.38 Secondly, immediately following the
in each transplantation process to seed endothelial cells to a preclotting technique, the surface may be rough and throm-
vascular prosthetic blood-contacting surface: bogenic.26 The third limitation of this seeding technique is
1. Gravitational;12,26-29 that, as previously mentioned, the endothelial cells adhere
2. Hydrostatic;30,31 and to the graft surface in a spheroid morphology (100% spher-
3. More recently, electrostatic.32 oid shaped) directly from the seeding suspension; thus an
The hypothesis for each of these approaches has been incubation period should be employed (> 2 h) for adhesion
that the transplantation of endothelial cells (initially low maturation prior to implantation.31
surface density, cells/area) on a graft luminal surface will As described above, many investigators have attempted
promote, through time, the development of a mature, physi- to increase the number of endothelial cells adhered to grafts
ologically appropriate, confluent graft luminal endothelial as well as the magnitude of surface adhesion (flattening) by
cell-lined neointima. placing adhesive proteins such as fibronectin on the graft
The most basic of the three seeding techniques that blood-contacting surface to act as a “glue”.39-48 Significant
have been tested utilizes gravitational forces to deliver en- research efforts have focused on glue formulations includ-
dothelial cells to a vascular prosthetic luminal surface. The ing fibronectin, extracellular matrix, collagen, laminin, fi-
basic concept has been to fill the graft with the harvested brin, fibroblast matrix, and plasma.27,40,41,43,45-50 The most
endothelial cells resuspended in tissue culture medium or commonly investigated “glue” has been fibronectin.
blood plasma. The filled graft is maintained horizontally and Fibronectin is an adhesive glycoprotein which is found in
rotated periodically or continuously over a prolonged seed- the basement membrane to which the endothelial cells are
ing time period. Biological glues have commonly been used attached in natural blood vessels. This glycoprotein is re-
with this technique to promote endothelial cell attachment quired to attach the endothelial cells to culture flasks in vitro
to the prosthetic material. One disadvantage of using a glue and it has thus made sense to try it as the primary “glue“ to
is that any nonendothelialized surface becomes more throm- enhance endothelial cell adhesion to other artificial surfaces.
bogenic due to the exposure of blood cells to the glue.8 The problem associated with using fibronectin or any other
A significant disadvantage of these early attempts at “glue” arises from the minimal number of endothelial cells
gravitational endothelial cell seeding was that the trans- which can be harvested relative to the total graft surface area
planted endothelial cells were in a spheroid morphology on to be covered, as well as the inefficiency of the seeding pro-
the graft surface upon completion of the transplantation cedures.51 Any nonendothelialized graft surface or subse-
procedure. Thus, the potential existed for a significant loss quent loss of endothelial cells from the surface upon im-
of endothelial cells from the graft surface when blood flow plantation renders the exposed, fibronectin-treated graft
was restored through the graft if the seeding time was short surface attractive to platelets, thus potentially promoting
(< 2 h) and the cells had minimal time to flatten and ma- thrombotic events that could lead to graft failure. 8
ture. Endothelial cell losses up to 95% in the initial 24 h Ramalanjaona et al40,52 showed that the number of endo-
postimplantation were observed due to this cellular mor- thelial cells adhered was increased and the loss of cells upon
phologic immaturity.33 To overcome this limitation of im- exposure to shear stress was reduced using a fibronectin
mature morphology, attempts were made to endothelialize “glue”. The problem these investigators encountered was that
the graft luminal surface by allowing adhesion to the graft the areas not endothelialized were thrombogenic, and ex-
with subsequent morphological maturation in tissue cul- perimental subjects required anticoagulant therapy to re-
ture (7-14 days in vitro).27,28,34-37 These efforts did in fact duce the complications due to thrombus formation.
result in minimal losses of endothelial cells from the grafts One of the most difficult of the technical issues re-
upon subsequent exposure to blood flow. However, as pre- lated to endothelial cell derivation from autologous sources
viously mentioned, the practicality of this approach remains and subsequent transplantation is the time required for these
of concern to surgeons and researchers. A long tissue cul- processes. It has been our experience that a minimum of
ture period may not be acceptable for a patient who needs 45-60 minutes is required for “immediate” seeding of en-
emergency bypass surgery. In addition, the questions of ge- dothelial cells onto a prosthetic graft being directly implanted
notypic and/or phenotypic changes in cultured endothelial in a patient. This minimum time frame does not allow for
cells as well as the potential for infection (cell culture con- cell flattening and maturation onto the graft prior to resto-
tamination) have been of major concern to investigators in ration of blood flow through the graft. Some investigators
this field. have suggested that a time greater than two hours from
64 Tissue Engineering of Prosthetic Vascular Grafts
harvest to transplantation would most definitely not be ac- these studies indicated that an increasingly positively charged
ceptable clinically, due to the chance of genotypic and/or surface leads to enhanced adhesion, spreading, and
phenotypic changes to the endothelial cells which may oc- magnitude of contact regions. The results on increasingly
cur while they are exposed to media containing undefined negatively charged substrates indicated the inverse, with
serum.25 In addition, it is obvious that longer time periods inhibited adhesion, reduced spreading, and reduced
required for endothelial cell seeding of the vascular pros- contact regions.
theses equates to longer periods of time for patients under
anesthesia, and thus the potential for complications. The Electrostatic Endothelial Cell Seeding
length of time and dosage of anesthesia and its safety are The only conclusion that can be drawn from evaluat-
difficult to generalize due to the fact that exposure compli- ing 15 years of research efforts related to cell seeding tech-
cation encompasses multiple drugs, techniques, anesthetists, niques is that few concrete technical advancements have been
and patients.53,54 made. We have recently proposed and tested a novel device
The necessity for an incubation period for attachment that we feel will be of significant value in improving the ef-
and morphological maturation of transplanted endothelial ficiency of transplanted cell attachment, as well as minimiz-
cells is related to the nature of the electrostatic interactions ing cellular losses upon implantation.32 The technique is
between the endothelial cells and the prosthetic graft mate- called electrostatic endothelial cell seeding. The electrostatic
rials. Endothelial cells are negatively charged.55,56 The clini- seeding technique has been evaluated in vitro using the pro-
cally successful vascular prosthetics (i.e., ePTFE) are highly totype apparatus shown in Figure 3.1. The key to this tech-
negatively charged. This negativity repels platelets which are nique is that it enhances endothelial cell adhesion by induc-
also negatively charged.57-61 Thus, the initial adherence of ing a temporary positive surface charge or a “temporary glue”
transplanted endothelial cells must overcome this negative- on the negatively charged ePTFE graft luminal surface. Fol-
negative charge repulsive force (long range) between cells lowing cell transplantation the ePTFE graft luminal surface
and graft material in order for the seeding procedure to be reverts to its original highly negatively charged surface. Thus,
successful. Experiments using platelets have demonstrated any nonendothelialized graft surfaces or any exposed graft
that the cellular adhesion of platelets on a negatively charged surfaces resultant from endothelial cell losses upon restora-
substrate is one order of magnitude less (ten times) than tion of blood flow remain nonthrombogenic due to the re-
expected by gravitational settling alone due to this electro- stored high negative surface charge of the graft material itself.
static, repulsive, interaction which must be overcome by a The basic issue underlying electrostatic endothelial cell
stochastic process.62-64 Similar short range repulsive inter- seeding is: “How can the surface potential of the graft be
actions alter cellular morphologies by preventing or slow- altered to attract endothelial cells without rendering the sur-
ing morphological maturation even when cells overcome the face thrombogenic?” The electrostatic endothelial cell seed-
repulsive interactions with the material and attach to the ing technique takes advantage of graft material properties
surface. This reality is further demonstrated by endothelial (dielectric material). When a dielectric material is placed
cell and fibroblast studies that have shown the dependence within a capacitor (electrostatic seeding apparatus), the elec-
of cell adherence on the substrate surface charge.65-70 These trons of the atoms and ions which make up the dielectric
studies used varying substrates with varying surface charges material (near surface) are attracted to the capacitor surface
to study cell adhesion, spreading, and contact regions which has accumulated the positive charge. The nuclei of
between the cells and the substrates. The overall results from the dielectric material (near surface) are attracted to the
a
f
Endothelial Cell Seeding: A Review 65
negatively charged surface. These small displacements, or necessary fibronectin (< 3 days) and basement membrane
polarizations, are what induce the surface charge on the graft collagen (< 1 week) to maintain cellular adhesion on the
luminal surface. It should be noted that the electrons in a graft for the long term.75-77 Preliminary in vivo studies
dielectric material are not free and the displacements of the (unpublished) using a canine femoral artery implantation
electrons are very slight. Also, the interior volume of the graft model suggests thromboresistance of the electrostatically
material, dielectric, remains unchanged, thus leaving a net seeded grafts.
charge of zero over the dielectric material.71,72
Several in vitro studies have been performed utilizing Genetically Engineering Endothelial Cells
this electrostatic seeding technique. When human umbili- for Seeding Vascular Prostheses
cal vein endothelial cells were transplanted onto 4 mm I.D. It has recently been suggested that genetically engi-
ePTFE using the electrostatic cell seeding technique, a com- neered endothelial cells could be transplanted, which might
plete nodal area coverage of morphologically mature (com- promote the reduction/prevention of anastomotic hyper-
pletely flattened) endothelial cells (73,540 cells/cm2) was plasia and enhance thromboresistance of small diameter
obtained in 16 minutes (+1.0 Volt applied to apparatus) with vascular prostheses. Additional theoretical therapeutic ap-
minimal cellular membrane damage or effect on endothe- plications using manipulated endothelial cells include the
lial cell viability.32 A section of electrostatically seeded ePTFE possibilities of gene replacement, correction, or augmenta-
is illustrated in the scanning electron micrograph in Fig- tion which may be effective in the treatment of atheroscle-
ure 3.2. These in vitro evaluations of the electrostatic seed- rosis. Localized drug delivery may also be feasible due to the
ing technique revealed no significant losses of endothelial direct contact of transplanted endothelial cells within the
cells upon exposure of the graft to a wall shear stress of blood stream.78,79
15 dynes/cm2 for up to 120 minutes immediately after seed- The initial use of transplanted, genetically modified
ing. The majority of endothelial cell loss (up to 30%) occurs endothelial cells is credited to Zweibel et al.80,81 Since that
within the first 30 minutes of implantation using traditional time, the study of endothelial cell transfection (incorpora-
techniques.33,40 Thus, the electrostatic seeding procedure is tion of foreign DNA) using retroviral and electroporation
superior to the gravitational and hydrostatic seeding proce- methodologies has demonstrated long term cell viability as
dures in terms of the seeding time required, magnitude of well as stable transfection using the reporter genes !-galac-
endothelial cell adhesion (attachment), and cellular reten- tosidase, chloramphenicol acetyltransferase, luciferase and
tion. It is also speculated that the actin microfilaments which human tissue plasminogen activator (tPA), growth hormone,
make up the endothelial cell cytoskeleton and possess an urokinase, and nitric oxide synthase genes.82-93 The efficien-
electrostatic potential will also be rearranged by the electro- cies of retroviral and electroporation transfection of endot-
static endothelial cell seeding technique to assist in the main- helial cells are 1-2% and 10%, respectively.85,89 Adenovirus-
tenance of endothelial cell adhesion (act as an anchoring mediated gene transfer is highly dose dependent and pro-
system), although this has not yet been demonstrated in our duces only transient transfection. 87 Transfection and
experiments.73,74 It is also hoped that the enhanced cell endothelial cell seeding are procedures each of which re-
attachment resultant from electrostatic endothelial cell seed- quires long time periods of laboratory work, which again
ing may allow the seeded endothelial cells to synthesize the raises a red flag among the pragmatists concerned with
19. Gimbrone MA, Cotran RS, Folkman J. Human vascular 37. Budd JS, Allen KE, Hartley G et al. The effect of pre-
endothelial cells in culture. Growth and DNA synthesis. J formed confluent endothelial cell monolayers on the pa-
Cell Biol 1974; 60:673-84. tency and thrombogenicity of small calibre vascular grafts.
20. Sharefkin JB, Van Wart HE, Cruess DF et al. Adult hu- Eur J Vasc Surg 1991; 5:397-405.
man endothelial cell enzymatic harvesting. Estimates of 38. Turgeon ML. Clinical Hematology: Theory and Procedure.
efficiency and comparisons of crude and partially puri- Boston: Little, Brown, and Co., 1988:279-86.
fied bacterial collagenase preparations by replicate 39. Kesler KA, Herring MB, Arnold MP et al. Enhanced
microwell culture and fibronectin degradation measured strength of endothelial attachment on polyester elastomer
by enzyme-linked immunosorbent assay. J Vasc Surg 1986; and polytetrafluoroethylene graft surfaces with fibronectin
4:567-77. substrate. J Vasc Surg 1986; 3:58-64.
21. Jarrell BE, Williams SK, Stokes G et al. Use of freshly iso- 40. Ramalanjaona G, Kempczinski RF, Rosenman JE et al. The
lated capillary endothelial cells for the immediate estab- effect of fibronectin coating on endothelial cell kinetics
lishment of a monolayer on a vascular graft at surgery. on polytetrafluoroethylene grafts. J Vasc Surg 1986;
Surgery 1986; 100:392-9. 3:264-72.
22. Williams SK, Wang TF, Castrillo R et al. Liposuction-de- 41. Seeger JM, Klingman N. Improved in vivo
rived human fat used for vascular graft sodding contains endothelialization of prosthetic grafts by surface modifi-
endothelial cells and not mesothelial cells as the major cation with fibronectin. J Vasc Surg 1988; 8:476-82.
cell type. J Vasc Surg 1994; 19:916-23. 42. Pratt KJ, Jarrell BE, Williams SK et al. Kinetics of endot-
23. Rupnick MA, Hubbard FA, Pratt K et al. helial cell-surface attachment forces. J Vasc Surg 1988;
Endothelialization of vascular prosthetic surfaces after 7:591-9.
seeding or sodding with human microvascular endothe- 43. Van Wachem PB, Vreriks CM, Beugeling T et al. The in-
lial cells. J Vasc Surg 1989; 9:788-95. fluence of protein adsorption on interactions of cultured
24. Graham LM, Vinter DW, Ford JW et al. Endothelial cell human endothelial cells with polymers. J Biomed Mater
seeding of prosthetic vascular grafts. Early experimental Res 1987; 21:701-18.
studies with cultured autologous canine endothelium. 44. Van Wachem PB, Beuqeling T, Mallens BW et al. Depo-
Arch Surg 1980; 115:929-33. sition of endothelial fibronectin on polymeric surfaces.
25. Schmidt SP, Sharp WV, Evancho MM et al. Endothelial Biomaterials 1988; 9:121-3.
cell seeding of prosthetic vascular grafts-current status. In: 45. Schneider A, Melmed RN, Schwalb H et al. An improved
Szycher M, ed. High Performance Biomaterials. Lancaster: method for endothelial cell seeding on
Technomic Publishing Co., 1991:483-96. polytetrafluoroethylene small caliber vascular grafts. J Vasc
26. Yates SG, Barros AAB, Berger K et al. The preclotting of Surg 1992; 15:649-56.
porous arterial prostheses. Ann Surg 1978; 188:611-22. 46. Lee YS, Park DK, Kim YB et al. Endothelial cell seeding
27. Anderson JS, Price TM, Hanson SR et al. In vitro onto the extracellular matrix of fibroblasts for the devel-
endothelialization of small-caliber vascular grafts. Surgery opment of a small diameter polyurethane vessel. ASAIO J
1987; 101:577-86. 1993; 39: M740-5.
28. Foxall TL, Auger KR, Callow AD et al. Adult human 47. Bellon JM, Bujan J, Honduvilla NG et al. Endothelial cell
endothelial cell coverage of small-caliber dacron and seeding of polytetrafluoroethylene vascular prostheses
polytetrafluoroethylene vascular prostheses in vitro. J Surg coated with a fibroblastic matrix. Ann Vasc Surg 1993;
Res 1986; 41:158-72. 7:549-55.
29. Mazzucotelli JP, Roudiere JL, Bernex F et al. A new de- 48. Gosselin C, Vorp DA, Warty V et al. ePTFE coating with
vice for endothelial cell seeding of a small-caliber vascu- fibrin glue, FGF-1, and heparin: Effect on retention of
lar prosthesis. Artif Organs 1993; 17:787-90. seeded endothelial cells. J Surg Res 1996; 60:327-332.
30. Kempczinski RF, Rosenman JE, Pearce WH et al. Endot- 49. Williams SK, Jarrell BE, Friend L et al. Adult human en-
helial cell seeding of a new PTFE vascular prosthesis. J dothelial cell compatibility with prosthetic graft material.
Vasc Surg 1985; 2:424-9. J Surg Res 1985; 38:618-29.
31. Van Wachem PB, Stronck JWS, Koers-Zuideveld R et al. 50. Kaehler J, Zilla P, Fasol R et al. Precoating substrate and
Vacuum cell seeding: A new method for the fast applica- surface configuration determine adherence and spreading
tion of an evenly distributed cell layer on porous vascu- of seeded endothelial cells on polytetrafluoroethylene
lar grafts. Biomaterials 1990; 11:602-6. grafts. J Vasc Surg 1989; 9:535-41.
32. Bowlin GL. Electrostatic endothelial cell seeding of vas- 51. Lindblad B, Burkel WE, Wakefield TW et al. Endothelial
cular prostheses. Doctoral Dissertation, The University of cell seeding efficiency onto expanded
Akron, 1996:1-354. polytetrafluoroethylene grafts with different coatings.
33. Rosenman JE, Kempczinski RF, Pearce WH et al. Kinet- ACTA Chir Scand 1986; 152:653-6.
ics of endothelial cell seeding. J Vasc Surg 1985; 2:778-84. 52. Ramalanjaona GR, Kempczinski RF, Ogle JD et al.
34. Shindo S, Takagi A, Whittemore AD. Improved patency Fibronectin coating of an experimental PTFE vascular
of collagen-impregnated grafts after in vitro autogenous prosthesis. J Surg Res 1986; 41:479-83.
endothelial cell seeding. J Vasc Surg 1987; 6:325-32. 53. Longnecker DE, Murphy FL. Introduction to anesthesia.
35. Prendiville EJ, Coleman JE, Callow AD et al. Increased in 8th ed., Philadelphia: W.B. Saunders Co., Harcourt Brace
vitro incubation time of endothelial cells on fibronectin- Jovanovich, Inc., 1992:419-27.
treated ePTFE increases cell retention in blood flow. Eur 54. Guyton AC. Textbook of medical physiology. 8th ed.
J Vasc Surg 1991; 5:311-9. Philadelphia: W.B. Saunders Co., Harcourt Brace
36. Sentissi JM, Ramberg K, O’Donnell K et al. The effect of Jovanovich, Inc., 1991:269-71.
flow on vascular endothelial cells grown in tissue culture 55. Sawyer PN, Harshaw DH. Electroosmotic characteristics
on polytetrafluoroethylene grafts. Surgery 1986; 99:337-43. of canine aorta and vena cava walls. Biophy J 1966;
6:653-63.
68 Tissue Engineering of Prosthetic Vascular Grafts
56. Srinivasan S, Sawyer PN. Role of surface charge of the 78. Nabel EG, Plautz G, Nabel GJ. Gene transfer into vascu-
blood vessel wall, blood cells, and prosthetic material in lar cells. JACC 1991; 17:189B-94B.
intravascular thrombosis. J Colloid Sci 1970; 32:456-63. 79. Callow AD. The vascular endothelial cell as a vehicle for
57. Sawyer PN, Srinivasan S. Studies on the biophysics of in- gene therapy. J Vasc Surg 1990; 11:793-8.
travascular thrombosis. Am J Surg 967; 114:42-59. 80. Zweibel JA, Freeman SM, Kantoff PW et al. High-level
58. Lowell J. The electrification of polymers by metals. J Phys recombinant gene expression in rabbit endothelial cells
D: Appl Phys 1976; 9:1571-85. transduced by retroviral vectors. Science 1989; 243:220-2.
59. Yu ZZ, Watson PK, Facci JS. The contact charging of 81. Ryan US, Hayes BA, Maxwell G et al. Endothelial cells as
PTFE by mercury: The effect of a thiophene monolayer vehicles for continuous release of recombinant gene prod-
on charge exchange. J Phys D: Appl Phys 1990; ucts. In: Zilla P, Fasol R, Callow A, eds. Applied cardio-
23:1207-11. vascular biology. Basel: S. Karger, 1990:22-9.
60. Yu ZZ, Keith PK. Contact charge accumulation and re- 82. Sackman JE, Freeman MB, Petersen MG et al. Synthetic
versal on polystyrene and PTFE films upon repeated con- vascular grafts seeded with genetically modified endothe-
tacts with mercury. J Phys D: Appl Phys 1989; 22:798-801. lium in the dog: Evaluation of the effect of seeding tech-
61. Abramson HA. The electrophoresis of the blood platelets nique and retroviral vector on cell persistence in vivo. Cell
of the horse with the reference to their origin and to Trans 1995; 4:219-35.
thrombus formation. J Exp Med 1928; 47:677-83. 83. Huber TS, Welling TH, Sarkar R et al. Effects of retroviral-
62. Marmur A, Gill WN, Ruckenstein E. Kinetics of cell depo- mediated tissue plasminogen activator gene transfer and
sition under action of an external force. Bull Math Biol expression on adherence and proliferation of canine en-
1976; 38:713-21. dothelial cells seeded onto expanded
63. Ruckenstein E, Prieve DC. Dynamics of cell deposition polytetrafluoroethylene. J Vasc Surg 1995; 22:795-803.
on surfaces. J Theor Biol 1975; 51:429-38. 84. Powell JT, Klaasse Bos JM, Van Mourik JA. The uptake
64. Ruckenstein E, Marmur A, Rakower SR. Sedimentation and expression of the factor VIII and reporter genes by
and adhesion of platelets onto horizontal glass surface. vascular cells. FEBES 1992; 303:173-7.
Thrombus Haemostas 1976; 36:334-42. 85. Kahn ML, Lee SW, Dichek DA. Optimization of retroviral
65. Van Wachem PB, Hogt AH, Beugeling T et al. Adhesion vector-mediated gene transfer into endothelial cells in
of cultured human endothelial cells onto methacrylate vitro. Circ Res 1992; 71:1508-17.
polymers with varying surface wettability and charge. 86. Hong Z, Guangbin Z, Xiaojun Z et al. Enhanced adeno-
Biomaterials 1987; 8:322-9. associated virus vector expression by adenovirus protein-
66. Macarak EJ, Howard PS. Adhesion of endothelial cells to cationic liposome complex. Chinese Med J 1995;
extracellular matrix proteins. J Cell Phys 1983; 116:76-86. 108:332-7.
67. Sugimoto Y. Effects on the adhesion and locomotion of 87. Hong Z, Guangbin Z, Airu Z et al. Adenovirus mediated
mouse fibroblasts by their interacting with differently gene transfer of vascular smooth muscle cells and endot-
charged substrates. Exp Cell Res 1981; 135:39-45. helial cells in vitro. Chinese Med J 1995; 108:493-6.
68. Schakenraad JM, Arends J, Busscher HJ et al. Kinetics of 88. Von der Leyen HE, Gibbons GH, Morishita R et al. Gene
cell spreading on protein precoated substrata: A study of therapy inhibiting neointimal vascular lesions: In vivo
interfacial aspects. Biomaterials 1989; 10:43-50. transfer of endothelial cell nitric oxide synthase gene. Proc
69. Niu S, Matsuda T, Oka T. Endothelialization on various Natl Acad Sci 1995; 92:1137-41.
segmented polyurethanes: Cellular behavior and its sub- 89. Kotnis RA, Thompson MM, Eady SL et al. Attachment,
strate dependency. ASAIO Trans 1990; 36: M164-8. replication and thrombogenecity of genetically modified
70. Van Wachem PB, Schakenraad JM, Feijen J et al. Adhe- endothelial cells. Eur J Vasc Endovasc Surg 1995; 9:335-40.
sion and spreading of cultured endothelial cells on modi- 90. Wilson JM, Birinyi LK, Salomon RN et al. Implantation
fied and unmodified poly(ethyleneterephthalate): A mor- of vascular grafts lined with genetically modified endot-
phological study. Biomaterials 1989; 10:532-9. helial cells. Science 1989; 244:1344-6.
71. Fink DG, Christiansen D, eds. Electronics engineers’ hand- 91. Dunn PF, Newman KD, Jones M et al. Seeding of vascu-
book. 3rd ed. New York: McGraw-Hill Book Co., lar grafts with genetically modified endothelial cells: Se-
1989:6:29-64. cretion of recombinant TPA results in decreased seeded
72. Halliday D, Resnick R. Fundamentals of physics. 2nd ed. cell retention in vitro and in vivo. Circulation 1996;
New York: John Wiley & Sons, Inc., 1981:484-502. 93:1439-46.
73. Ando T, Kobayashi N, Munekata E. Electrostatic poten- 92. Schwachtgen JL, Ferreira V, Meyer D et al. Optimization
tial around actin. In: Sugi H, Pollack GH, eds. Mecha- of the transfection of human endothelial cells by
nism of myofilament sliding in muscle contraction. New electroporation. Biotechiques 1994; 5:882-7.
York: Plenum Press, 1993:361-76. 93. Kotnis RA, Thompson MM, Eady SL et al. Optimisation
74. Gottlieb AI, Langille BL, Wong MKK et al. Biology of of gene transfer into vascular endothelial cells using
disease: Structure and function of the endothelial electroporation. Eur J Vasc Endovasc Surg 1995; 9:71-9
cytoskelaton. Lab Invest 1991; 65:123-37. 94. Popov SV, Margolis LB. Formation of cell outgrowths by
75. Jaffe EA, Mosher DF. Synthesis of fibronectin by cultured external force: A model study. J Cell Sci 1988; 90:379-89.
human endothelial cells. J Exp Med 1978; 147:1779-91. 95. Spencer SC. Electroporation technique of DNA transfec-
76. Jaffe EA, Minick CR, Adelman B et al. Synthesis of base- tion. In: Murray EJ, ed. Methods in molecular biology.
ment membrane collagen by cultured human endothelial Clifton: The Humana Press, Inc., 1991:45-52.
cells. J Exp Med 1976; 144:209-25. 96. Davis LG, Dibner MD, Battey JF. Basic methods in mo-
77. Howard BV, Macarak EJ, Gunson D et al. Characteriza- lecular biology. Norwalk: Appleton & Lange, 1986:293-95.
tion of the collagen synthesized by endothelial cells in
culture. Proc Natl Acad Sci USA 1976; 73:2361-4.
CHAPTER 4
Surface Precoating in the 1980s:
A First Taste of Cell-Matrix Interactions
J. Vincent Smyth, Michael G. Walker
T echnical difficulties and the frequent lack of available saphenous vein for peripheral arte-
rial reconstruction resulted in the development of a variety of prosthetic materials from
which grafts could be constructed. Of these, polyester elastomer (Dacron) and expanded
polytetrafluorethylene (ePTFE) emerged as the most successful in clinical practice, and re-
main leaders in the field today. However, small caliber prosthetic grafts have consistently
shown lower patency rates, especially when passing across the knee joint, when compared
with autologous vein conduits. The understanding that the endothelial lining of vein grafts
was responsible for the improved patency rates achieved in peripheral bypass naturally led
to attempts to create such a lining on prosthetic grafts, when explants showed that no sig-
nificant endothelialization of such grafts occurs naturally in humans, contrary to the results
seen in animals where virtually complete endothelial linings develop on prosthetic grafts in
a matter of weeks.
Almost all of the work performed up to 1980 utilized animal tissue models such as
bovine, canine or murine endothelium, and extremely promising results had been repeat-
edly achieved both in vivo and in vitro in terms of reduction in graft thrombogenicity, resis-
tance to infection, accelerated re-endothelialization and improved patency.1-6 In order to
make the transition to clinical practice, however, it was clear that human cell lines would be
required, and human umbilical vein endothelial cells (Huvecs) were used as the principal
experimental model until Jarrell showed that human adult endothelial cells (HAECs) could
be harvested from saphenous veins and cultured.7 This work has now advanced so that the
acute enzymatic harvesting of autologous vein can be routinely achieved, with virtually com-
plete denudation of the donor vein segment and a large proportion of harvested cells re-
maining viable for culture. There is now extensive experience with both cell lines toward the
desired end point of successful graft seeding to the rapid development of an endothelial
monolayer that resists the shear stress of flow.8
Almost as soon as the successful culture of Huvecs on prosthetic graft material was
first reported, it was clear that human cell lines were more fragile and fastidious in their
requirements than the canine or bovine endothelial cultures that had principally been used
up to this point. The establishment of a culture was slower even under optimal conditions,
and when achieved these were less hardy and prone to failure from infection. Even so, suc-
cessful culture was reported on all the common graft materials, with typical appearances of
cobblestone morphology and cells expressing endothelium-specific markers such as vWF.
Although human cell lines showed slower cell division and spreading across the culture
surfaces than animal cell lines, even under the optimal experimental conditions of tissue
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
70 Tissue Engineering of Prosthetic Vascular Grafts
culture, the main reason for the delayed development of the grafts, up to 20 times as many cells as were required to form
monolayer was found to be low cell attachment to the graft. a monolayer would be needed for seeding, thus greatly in-
creasing the required cell numbers. Although this could be
Endothelial Cell Attachment accommodated by the use of preculture in the laboratory,18
to Prosthetic Grafts this method is not compatible with routine surgical prac-
The first seeding experiments were carried out in what tice and would necessitate a preliminary operation for vein
we would now classify as a static system, where the cells were harvest and carry risks of disease transmission or graft in-
introduced to the graft surface in a suspension of culture fection. To overcome the problem of limited cell harvests,
medium and allowed to settle on the graft over a period of the use of microvascular (capillary) endothelium19 and me-
time, then cultured in situ. Early work relied on qualitative sothelial20 cells have been proposed, but these only offer a
phase contrast microscopy of the seeded surface at intervals partial solution in terms of available cell numbers, as they
after seeding to assess degree of coverage. Later, the work of share many of the attachment and adhesion difficulties ex-
Sharefkin led to the use of the radio-isotope 111Indium as perienced with Huvecs and HAECs, and raise additional
an endothelial cell label,9 and attachment to grafts could be problems with tissue availability. As a consequence, to achieve
accurately quantified. It was discovered that prosthetic grafts rapid development of confluence in the clinical arena where
exhibit a uniformly poor surface for retention of seeded cells, such large cell numbers could not be obtained acutely using
typically around 4% for ePTFE,10,11 while cell adherence to established methods, either new sources of endothelial cells
plain Dacron was negligible because of its porosity, result- would be required or cell attachment significantly improved
ing in leakage of the seeding solution.12,13 Three approaches to permit successful seeding at subconfluent densities.
to the problem of rapidly developing a monolayer were It had been noticed that the results of human cell seed-
explored. ing work did not parallel those seen in the animal in vivo
The proportion of seeded cells that remained attached models of prosthetic graft seeding. Significantly better at-
to the graft could be increased by increasing the length of tachment was seen in the latter, and it was realized that the
time that the seeding suspension was exposed to the graft preclot typically used for the endothelial cell suspension
material.10,14-16 However, the seeding kinetics indicated that acted as a graft coating, improving cell adherence. By using
the rate of increase fell off after around 30 minutes, and that preclot or serum to coat the graft prior to seeding the en-
even with exposure times of over 60 minutes, the develop- dothelial cells, attachment rates could be dramatically im-
ment of a confluent monolayer was not significantly accel- proved to over 60% of seeded cells in a range of models.10,21
erated. Additionally, prolonged incubation times are incom- As a result, investigators began precoating the culture
patible with the constraints of the operating room. dishes and seeding surfaces with a variety of matrices before
Using more endothelial cells per graft area in the seed- the cells were exposed, and similar improvements in cell at-
ing solution, a higher seeding density, produced a greater tachment were seen11,22-27 (Figs. 4.1-4.3). Comparative stud-
number of cells attaching to the graft, but only in the same ies of a range of potential graft surface coatings were made.
proportion and consequently with an enormous waste of Because of Dacron’s inherent porosity, gelatin or collagen-
seeded cells.15,17 At established attachment rates to plain sealed grafts were frequently used, and this not surprisingly
improved initial cell adherence accordingly, as did all graft plasma show good rates of endothelial attachment, this is
coating matrices compared to the plain graft materials. thought to be due to their high FN content. Laminin and
Histological examination of the vascular endothelium collagens I, III and IV have not proved as effective as FN in
indicated that in the body it was supported by a protein- facilitating endothelial cell attachment. Gelatin-sealed
rich basement membrane layer, and that this layer was pro- Dacron grafts show good initial cell adherence, but the sta-
duced actively at least in part by the endothelium. Examina- bility of the gelatin crosslinking under physiological condi-
tion of endothelial cells in culture showed that the adherent tions is uncertain, as it degrades and may not provide a suit-
cells secreted a similar layer on artificial surfaces28,29 and that able matrix for long enough to allow the cells to establish
a principal component of the basement membrane was their own FN-rich basement membrane layer on the graft.
fibronectin (FN). Fibrin glue has been used successfully in some seeding work,
Following the work of Ramalanjoana21 and others,30-32 but requires fibrinolytic inhibition prior to use without of-
FN has become the most commonly used seeding matrix fering any advantages in terms of cell retention. In the labo-
because of efficacy and availability, giving consistently high ratory model, however, almost equivalent results to those
cell adherences in experiments using a variety of graft types, with FN have been achieved and, in retrospect, it is clear
including ePTFE and Dacron. Preclot, serum, plasma, that one reason for the success of the early animal seeding
laminin, gelatin, collagen and fibrin glue have also been in- experiments using prosthetic grafts had been the serendipi-
vestigated,10,27,33-36 the first three because they are readily tous use of FN-rich preclot as a readily available cell sus-
available in the clinical situation, and the remainder because pension to seed the graft, sealing the porous Dacron used in
they, along with fibronectin, are found as native constitu- this work.
ents of extracellular matrix. Although preclot, serum and
72 Tissue Engineering of Prosthetic Vascular Grafts
Many workers have investigated the attachment kinet- Following flattening, where incomplete confluence
ics of endothelial cells to FN-coated graft surfaces.10,15,27,37 results from seeding, the endothelial cells then migrate across
In the laboratory, the optimum incubation time for cell at- the seeding surface to produce a monolayer. By some elegant
tachment to a coating matrix appears to be 30 minutes with work using removable discs, the migration of saphenous vein
no significant benefit evident beyond this, though some endothelial cells following establishment of healthy cultures
workers have found as little as 15 minutes adequate. As with was measured on a variety of culture surfaces.41 These
uncoated grafts, improved cell attachment is also seen with showed that surface precoating not only improves initial
increasing the seeding density, up to a point whereafter the adherence, but also promotes cell migration and develop-
proportion of seeded cells attaching falls off, though the ment of confluence. Again, fibronectin was the most effec-
absolute number continues to increase. The kinetics of im- tive coating agent, with gelatin and extracellular matrix
proved cell adherence to FN-coated graft surfaces implied showing some advantage over uncoated culture flasks. In-
some specific binding mechanism, and this was confirmed terestingly, laminin, despite being a constituent of the cell
by the finding that endothelial cells express specific FN re- substratum in vivo, did not improve cell migration, imply-
ceptors on their cell membrane. ing that its role in the body is not primarily concerned with
Comparative studies of different grafts seem to sug- maintenance of endothelial monolayer integrity. In addition
gest that ePTFE is a better surface for matrix coating than to enhancement of cell-surface adhesion, seeded cells also
Dacron, although there is some disagreement between indi- proliferate more rapidly on precoated surfaces,22 and some
vidual studies. This may be related to the nature of the graft groups developed variations of the surface coatings specifi-
surface at the fiber level, with a flatter surface being offered cally designed to improve graft endothelialization by pro-
by the multinodal fibrillary construction than the woven or moting cell migration and proliferation.42 Coatings enriched
knitted Dacron. Minor refinements in terms of graft com- with growth factors such as FGF have been shown to en-
position, pore size, internodal distances or woven vs. knit- hance ingrowth through the graft interstices43 and the de-
ted Dacron, do not seem to affect the cell seeding kinetics velopment of an endothelial lining with a minimum of
significantly. On precoated grafts, the differences in endo- smooth muscle hyperplasia.44 The ability of small diameter
thelial cell adherence between graft materials is not striking vascular prostheses to adsorb and retain ECGF under flow
once the porosity of the Dacron is reduced by sealing the conditions has been clearly demonstrated45 though in the
graft with the surface coating, and this can be explained by rabbit model used, no differences in graft re-
the production of essentially similar attachment character- endothelialization was seen at 1 month. Although the com-
istics at the molecular level on the surfaces to which the bination of growth factor-enhanced graft coatings and hu-
seeded endothelial cells are exposed. man endothelial cell seeding would appear to be a promis-
With the establishment of a FN-rich coating as the ing avenue of investigation, there have been no reports of
optimal seeding surface, several workers have investigated such work.
the adsorption kinetics of FN onto prosthetic grafts,38,39
showing that the development of FN-graft binding is pro- Endothelial Cell Retention
portional to the FN concentration and the duration of in- Under Flow Conditions
cubation, though there may be minor differences between The advantages of precoated over plain grafts also ex-
graft materials. The optimum concentration of FN to use as tend to dynamic seeding systems, in which the seeded grafts
the seeding matrix has been investigated,40 and the relation- are exposed to the shear stress of flow, though different
ship between the concentration of FN graft coating and rates groups have achieved wide variation in results. A range of
of endothelial cell adherence approximates first order ki- levels of flow, and thus shear stresses, have been used, in-
netics to 50 ∝g/ml, with no benefit obtained by increasing cluding systems that approximate rates of flow seen in
this to 250 ∝g/ml, further evidence for specific FN-binding grafts.46,47 Electron microscopy of seeded grafts exposed to
sites on the cell surface. pulsatile flow systems show that a proportion of endothelial
loss occurs in areas where the underlying graft is exposed by
Cell Migration loss of surface coating,48 and that this is more marked with
It had been noted that following seeding, cells initially FN-coating than with preclot, which may account for the
attached to the graft in a rounded morphology, later devel- differences some workers have seen in levels of cell reten-
oping a more flattened phenotype responsible for the tion. The monolayer’s resistance to denudation under flow
‘cobblestone’ appearance of the mature endothelial mono- is a combination of the strength of the endothelial cell at-
layer.32 Increased duration of incubation was associated with tachment to the matrix, and of the matrix to the graft. With
progression of the cell morphology from rounded and poorly regard to the former, factors previously observed to improve
attached, to flattened and adherent, and this was generally cell adherence such as duration of incubation, and also de-
accepted to indicate the acclimatization of the seeded cell to gree of confluence and interval prior to commencement of
the seeding surface. The speedier development of an adher- flow, are also relevant to resisting denudation under flow.
ent monolayer following seeding onto prepared graft and Established endothelial monolayers formed by culture on
culture surfaces was thought to correspond with the more the graft resist flow better than acutely attached cells,46,49,50
rapid establishment of the flattened phenotype, as noted although previously noted problems with duration of incu-
microscopically.16,33 bation or graft preculture apply in terms of clinical applica-
bility. Cell loss appears to occur in a biphasic pattern,47,51
Surface Precoating in the 1980s: A First Taste of Cell-Matrix Interactions 73
with an initial rapid rate presumably corresponding to tachment and retention on autologous native surfaces might
loosely attached cells or areas of coating, which settles after be expected to be optimal.
a few minutes to a slower, more steady rate of loss (Fig. 4.4). Seeding after endothelial denudation by angioplasty
However, we have noted that within a flow system sudden balloon has been assessed in a rabbit iliac vein model52 us-
changes in the level of pulsatile shear stress result in a further ing a double balloon catheter-based system and has shown
period of rapid loss which then returns to the slower rate. that acute seeding with autologous cells on a relatively short
The bond between coating and graft has been less thor- time scale can restore a monolayer,53 which is maintained
oughly investigated, but seems to depend on the nature of on restoration of flow. This monolayer retains functional
the graft, with gelatin-coated Dacron performing signifi- integrity54 and may reduce neointimal hyperplasia.55 How-
cantly less well than ePTFE.38,40 Whether the FN-gelatin or ever, the relevance of these results to chronic arterial disease
the gelatin-Dacron interface is the important one is as yet in humans remains uncertain, especially with previous ex-
uncertain, but clinically may not be particularly important, perience of promising animal seeding studies. Endothelial
as ePTFE is the usual graft of choice in the infrainguinal cell seeding following endarterectomy in animals has also
region where it is expected that seeding will be most appli- given hope, demonstrating most of the advantages seen in
cable. Up to 75% of applied matrix may be lost in the first seeded prosthetic grafts.56-60
15 minutes after restoration of flow, but thereafter the graft- Work using human cell lines has demonstrated excel-
matrix bond appears stable within the time scale of the labo- lent cell attachment to both endarterectomy and vein
ratory model. Electron microscopy of coated grafts after angioplasty surfaces,61-63 and in the latter model flow stud-
exposure to flow shows clearly that this early loss occurs at ies have suggested cell retention kinetics similar to coated
least partly by detachment of regions of matrix from the grafts. Following endarterectomy the exposed surface is very
graft, presumably where the matrix-graft bond is less estab- similar to that on which the endothelium normally rests and
lished, with matrix solubility probably contributing more is high in FN, which probably accounts for the optimal at-
towards the later, slow rate of matrix loss. tachment rates seen, and HAECs seeded onto endarter-
ectomized vessels have been shown to be capable of form-
Seeding of Native Vascular Surfaces ing a monolayer,61 again retaining functional integrity.64
The difficulties in achieving an endothelial monolayer
on prosthetic grafts immediately after seeding, given the al- Mechanism of Cell Adherence
most inevitable shortfall in numbers of seeded cells, ethical As a result of the improvements in seeding efficiency
and technical problems with cell or graft preculture, and the produced by surface coatings, increasing interest has devel-
high losses from even FN-coated graft surfaces under flow, oped in the interactions between endothelial cells and the
have prompted workers to examine the seeding potential of matrix. The attachment and migration characteristics of the
native arterial surfaces. The use of autologous cells and na- endothelial cell are due to the presence of a wide range of
tive conduits avoids any potential immune or foreign body adhesion molecules and ligand-specific receptors on the cell
reaction which might result in cell loss, and seeding follow- membrane.65 Many of the receptors have multiple, overlap-
ing angioplasty or endarterectomy have both been consid- ping roles in different pathways, or respond to more than
ered. The advantages of seeding such regions are that the one signal molecule. Variation in ligand affinity and pat-
area for seeding is lower due to shorter segments being in- terns of receptor activation are common and the degree of
volved, thus increasing the seeding density without needing complexity of the intracellular responses to the extracellu-
an increase in the numbers of seeded cells, and that cell at- lar environment is only now becoming more fully
appreciated. Some receptors are endothelial cell specific, tory mediators, ICAM-2 and ICAM-3 are constitutively pro-
some are shared with other vascular cells such as platelets or duced, but all bind to circulating leukocytes. PECAM-1 is
leukocytes, and some also occur on other vessel wall cell types constitutively produced and binds platelets in addition.
including smooth muscle cells and fibroblasts. Because of Again, these molecules are principally involved in the en-
their differential actions, different classes of adhesion mol- dothelial response to inflammation, and these groups are
ecules are frequently asymmetrically arranged between the therefore not further considered.
luminal and abluminal cell membrane (Fig. 4.5). Cell-substratum interactions such as the endothelial
There are three main families of adhesion molecules cell adherence to prosthetic grafts and coatings are medi-
present on endothelial cells, though classification is amended ated by the integrin family of adhesion molecules,66 which
from time to time as new members are continually being are 10- to 100-fold more common on the cell membrane
identified and characterized, and many are known by more than selectins or immunoglobulin-like adhesion molecules,
than one name. The three major adhesion molecules fami- and are present principally on the abluminal surface.
lies consist of the selectins or leukocyte adhesion molecules Integrins are heterodimeric molecules, each subtype being
(ELAMs), responsible for initial capture of circulating leu- composed of a noncovalently bonded pair of # and ! sub-
kocytes in response to inflammatory mediators; the immu- units, each of which has multiple variants. There are several
noglobulin superfamily, also implicated in cell-cell interac- subclasses, distinguished by their ! subunit, though their
tions; and the integrins, involved in both cell-substratum specific ligand is to a large extent determined by the # sub-
and cell-cell interactions. unit. They bind with relatively low affinity to their ligands,
The selectins present on endothelial cells comprise two made up for in large part by their frequency of expression,
major groups; E-selectin or ELAM-1, and P-selectin or gran- allowing multiple weak bonds to the substratum rather than
ule membrane protein-140 (GMP-140). These are induced a few strong links, and thus facilitating cell spread and mi-
by inflammatory mediators and promote the adherence of gration without loss of firm adhesion, the ‘Velcro’ theory of
leukocytes to affected regions of endothelium via leukocyte cell attachment. Ligand binding is cation dependent, Ca2+
surface ligands such as fucose groups. They are located on or Mg2+ depending on the integrin, bound to specific sites
the luminal membrane surface and play little part in endo- on the # subunit.
thelium-substratum interactions. The immunoglobulin su- The VLA group, identified by a !1 subunit, includes
perfamily contains several classes, differing in the length and specific receptors found on the abluminal endothelial cell
composition of a string of immunoglobulin domains which membrane for fibronectin, laminin and collagen. A !2 sub-
lie on the luminal side of the endothelial cell membrane, unit identifies the leucam group which are found on circu-
attached to a peptide chain that extends through this to a lating inflammatory cells and bind to ICAMs on endothe-
cytoplasmic terminal carboxyl group. They include the in- lial cells. The cytoadhesin group of integrins, with a !3 sub-
tercellular adhesion molecules (ICAMs) -1, -2, and -3; vas- unit, includes low specificity, multiligand receptors binding
cular cell adhesion molecule-1 (VCAM-1); and platelet en- fibrinogen, fibronectin, vitronectin, thrombospondin,
dothelial cell adhesion molecule-1 (PECAM-1). ICAM-1 and laminin and vWF. Individual moieties with !4-!8 subunits
VCAM-1 are inducible in response to circulating inflamma- binding a variety of basement membrane proteins, includ-
ing those mentioned previously, have also been identified.
20. Nicholson LJ, Clarke JM, Pittilo RM et al. The mesothe- 39. Budd JS, Allen KE, Bell PRF et al. The effect of varying
lial cell as a non-thrombogenic flow surface. Thromb fibronectin concentration on the attachment of endothe-
Haemost 1984; 52(2):102-104. lial cells to polytetrafluoroethylene grafts. J Vasc Surg
21. Ramalanjoana G, Kempczinski RF, Rosenman JE et al. The 1990; 12:126-130.
effect of fibronectin coating on endothelial cell kinetics 40. Vohra RK, Thompson GJL, Sharma H et al. Fibronectin
in polytetrafluoroethylene grafts. J Vasc Surg 1986; coating of expanded polytetrafluoroethylene (ePTFE)
3:264-272. grafts and its rôle in endothelial seeding. Artif Organs
22. Sank A, Rostami K, Weaver F et al. New evidence and 1990; 14:41-45.
new hope concerning endothelial seeding of vascular 41. Hasson JE, Wiebe DH, Sharefkin JB et al. Migration of
grafts. Am J Surg 1992; 164:199-204. adult human vascular endothelial cells: Effect of extracel-
23. Stansby G, Shukla N, Fuller B et al. Seeding of human lular matrix proteins. Surgery 1986; 100:384-390.
microvascular endothelial cells onto PTFE graft material. 42. Lalka SG, Oelker LM, Malone JM et al. Acellular vascular
Br J Surg 1991; 78:1189-1192. matrix: A natural endothelial cell substrate. Ann Vasc Surg
24. Thomson GJL, Vohra R, Walker MG. Cell seeding for 1989; 3(2):108-117.
small diameter ePTFE grafts: A comparison between adult 43. Greisler HP, Cziperle DJ, Kim DU et al. Enhanced
human endothelial and mesothelial cells. Ann Vasc Surg endothelialization of ePTFE grafts by fibroblast growth
1989; 3(2):140-145. factor type 1 pretreatment. Surgery 1992; 112:244-254.
25. Vohra R, Thomson GJL, Carr HMH et al. In vitro adher- 44. Gray JL, Kang SS, Zenni GC et al. FGF-1 affixation stimu-
ence and kinetics studies of adult human endothelial cell lates ePTFE endothelialization without intimal hyperpla-
seeded polytetrafluoroethylene and gelatin-impregnated sia. J Surg Res 1994; 57:596-612.
Dacron grafts. Eur J Vasc Surg 1991; 5:93-103. 45. Greisler HP, Klosak JJ, Dennis JW et al. Biomaterial pre-
26. Thomson GJL, Vohra RK, Carr HMH et al. Adult human treatment with ECGF to augment endothelial cell prolif-
endothelial cell seeding using expanded eration. J Vasc Surg 1987; 5:393-399.
polytetrafluoroethylene vascular grafts: A comparison of 46. Sentissi JM, Ramberg K, O’Donnell TFJ et al. The effect
four substrates. Surgery 1991; 109:20-27. of flow on vascular endothelial cells grown in tissue cul-
27. Pratt KJ, Jarrell BE, Williams SK et al. Kinetics of endot- ture on polytetrafluoroethylene grafts. Surgery 1986;
helial cell surface attachment forces. J Vasc Surg 1988; 99:337-342.
7:591-599. 47. Vohra RK, Thomson GJL, Sharma H et al. Effects of shear
28. van Wachem PB, Beugeling T, Mallens BW et al. Deposi- stress on endothelial cell monolayers on expanded
tion of endothelial fibronectin on polymeric surfaces. polytetrafluoroethylene (ePTFE) grafts using preclot and
Biomaterials 1988; 9:121-123. fibronectin matrices. Eur J Vasc Surg 1990; 4:33-41.
29. van Wachem PB, Mallens BW, Dekker A et al. Adsorp- 48. James NL, Schindhelm K, Slowiaczek P et al. Endothelial
tion of fibronectin derived from serum and from human cell seeding of small diameter vascular grafts. Artif Or-
endothelial cells on to tissue culture polystyrene. J Biomed gans 1990; 14:355-360.
Mater Res 1987; 21:1317-1327. 49. Miyata T, Conte MS, Trudell LA et al. Delayed exposure
30. Seeger JM, Klingman N. Improved endothelial cell seed- to pulsatile shear stress improves retention of human
ing with cultured cells and fibronectin-coated grafts. J Surg saphenous vein endothelial cells on seeded ePTFE grafts.
Res 1985; 38:641-647. J Surg Res 1991; 50:485-493.
31. Kesler KA, Herring MB, Arnold MP et al. Enhanced 50. Schneider PA, Hanson SR, Price TM et al. Durability of
strength of endothelial attachment on polyester elastomer confluent endothelial cell monolayers on small calibre
and polytetrafluoroethylene graft surfaces with fibronectin vascular prostheses in vitro. Surgery 1988; 103:456-462.
substrate. J Vasc Surg 1986; 3:58-64. 51. Rosenman JE, Kempczinski RF, Pearce WH et al. Kinet-
32. Curti T, Pasquinelli G, Preda P et al. An ultrastructural ics of endothelial cell seeding. J Vasc Surg 1985; 2:778-784.
and immunocytochemical analysis of human endothelial 52. Thompson MM, Budd JS, Eady SL et al. A method to
cell adhesion on coated vascular grafts. Ann Vasc Surg transluminally seed angioplasty sites with endothelial cells
1989; 3(4):351-363. using a double balloon catheter. Eur J Vasc Surg 1993;
33. Kaehler J, Zilla P, Fasol R et al. Precoating substrate and 7:113-121.
surface configuration determine adherence and spreading 53. Thompson MM, Budd JS, Eady SL et al. Platelet deposi-
of seeded endothelial cells on PTFE grafts. J Vasc Surg tion after angioplasty is abolished by restoration of the
1989; 9:535-541. endothelial cell monolayer. J Vasc Surg 1994; 19:478-486.
34. Baker KS, Williams SK, Jarrell BE et al. Endothelialization 54. Thompson MM, Budd JS, Eady SL et al. Endothelial cell
of human collagen surfaces with human adult endothe- seeding of damaged native vascular surfaces: Prostacyclin
lial cells. Am J Surg 1985; 150:197-200. production. Eur J Vasc Surg 1992; 6:487-493.
35. Dalsing MC, Kevorkian M, Raper B et al. An experimen- 55. Thompson MM, Budd JS, Eady SL et al. The effect of
tal collagen-impregnated Dacron graft: Potential for en- transluminal endothelial seeding on myointimal hyperpla-
dothelial seeding. Ann Vasc Surg 1989; 3(2):127-133. sia following angioplasty. Eur J Vasc Surg 1994; 8:423-434.
36. Zilla P, Fasol R, Preiss P et al. Use of fibrin glue as a 56. Bush HL, Jakubowski JA, Curl GR et al. Luminal healing
substrate for in vitro endothelialization of PTFE vascular of arterial endarterectomy: Role of autogenous endothe-
grafts. Surgery 1989; 105:515-522. lial cell seeding. Surg Forum 1985; 36:446-450.
37. Mosquera DA, Goldman M. Endothelial cell seeding. Br J 57. Bush HL, Jakubowski JA, Sentissi JM et al. Neointimal
Surg 1991; 78:656-660. hyperplasia occurring after carotid endarterectomy in a
38. Carr HMH, Vohra R, Welch M et al. Fibronectin binding canine model: Effect of endothelial cell seeding vs
to gelatin-impregnated Dacron (Gelseal) prostheses. Artif perioperative aspirin. J Vasc Surg 1987; 5:118-125.
Organs 1992; 16:342-345. 58. Schneider PA, Hanson SR, Price TM et al. Confluent du-
rable endothelialisation of endarterectomised baboon aorta
Surface Precoating in the 1980s: A First Taste of Cell-Matrix Interactions 77
by early attachment of cultured endothelial cells. J Vasc 72. Eskin SG, Ives CL, Frangos JA et al. Cultured endothe-
Surg 1990; 11:365-372. lium: The response to flow. ASAIO J 1985; 8:109-112.
59. Krupski WC, Bass A, Anderson JS et al. Aspirin-indepen- 73. Kugiyama K, Sakamoto T, Misumi I et al. Transferable
dent antithrombotic effects of acutely attached cultured lipids in oxidized low-density lipoprotein stimulate plas-
endothelial cells on endarterectomised arteries. Surgery minogen activator inhibitor-1 and inhibit tissue-type plas-
1990; 108:283-291. minogen activator release from endothelial cells. Circ Res
60. Sterpetti AV, Schultz RD, Bailey RT. Endothelial cell seed- 1993; 73:335-343.
ing after carotid endarterectomy in a canine model re- 74. Yamamoto C, Kaji T, Sakamoto M et al. Effect of
duces platelet uptake. Eur J Vasc Surg 1992; 6:390-394. endothelin on the release of tissue plasminogen activator
61. Smyth JV, Rooney OB, Dodd PDF et al. Culture of hu- and plasminogen activator inhibitor-1 from cultured hu-
man endothelial cells on endarterectomy surfaces. Eur J man endothelial cells and interaction with thrombin.
Vasc Endovasc Surg 1995; 10:308-315. Thromb Res 1992; 67:619-624.
62. Walluscheck KP, Steinhoff G, Haverich A. Endothelial cell 75. Loskutoff DJ, Ny T, Sawdey M et al. Fibrinolytic system
seeding of native vascular surfaces. Eur J Vasc Endovasc of cultured endothelial cells; regulation by plasminogen
Surg 1996; 11:290-303. activator inhibitor. J Cell Biochem 1986; 32:273-280.
63. Thompson MM, Budd JS, Eady SL et al. Effect of seeding 76. van Hinsbergh VWM, Bertina RM, van Wijngaarden A et
time and density on endothelial cell attachment to dam- al. Activated protein c decreases plasminogen activator
aged vascular surfaces. Br J Surg 1993; 80:359-362. inhibitor activity in endothelial cell-conditioned medium.
64. Smyth JV, Rooney OB, Dodd PD et al. Production of plas- Blood 1985; 65:444-451.
minogen activator inhibitor-1 by human saphenous vein 77. van Hinsbergh VWM, Kooistra T, van den Berg EA et al.
endothelial cells seeded onto endarterectomy specimens Tumor necrosis factor increases the production of plas-
in vitro. J Vasc Surg 1997; 25:722-725. minogen activator inhibitor in human endothelial cells in
65. Mousa SA, Cheresh DA. Recent advances in cell adhesion vitro and in rats in vivo. Blood 1988; 72:1467-1473.
molecules and extracellular matrix proteins: Potential 78. Hanss M, Collen D. Secretion of tissue-type plasminogen
clinical implications. DDT 1997; 2:187-199. activator and plasminogen activator inhibitor by cultured
66. Extracellular matrix receptors on animal cells: The human endothelial cells: Modulation by thrombin, endo-
integrins. In: Alberts B, Bray D, Lewis J et al. Molecular toxin and histamine. J Lab Clin Med 1987; 109:97-104.
biology of the cell. 3rd ed. New York: Garland Publish- 79. Lundgren CH, Herring MB, Arnold MP et al. Fluid shear
ing, Inc, 1994:995-1000. disruption of cultured endothelium: The effect of cell spe-
67. Sterpetti AV, Crucina A, Napoli F et al. Growth factor cies, fibronectin crosslinking and supporting polymer.
release by smooth muscle cells is dependent on ASAIO Trans 1986; 32:334-338.
haemodynamic factors. Eur J Vasc Surg 1992; 6:636-638. 80. Ives CL, Eskin SG, McIntire LV et al. The importance of
68. Greisler HP, Henderson SC, Lam TM. Basic fibroblast cell origin and substrate in the kinetics of endothelial cell
growth factor production in vitro by macrophages exposed alignment in response to steady flow. ASAIO Trans 1983;
to Dacron and polyglactin 910. J Biomater Sci Polym Ed 29:269-274.
1993; 4:415-430. 81. Li JM, Menconi MJ, Wheeler HB et al. Precoating ePTFE
69. Greisler HP, Dennis JW, Endean ED et al. Macrophage/ grafts alters production of endothelial cell derived
biomaterial interactions: The stimulation of endo- thrombomodulators. J Vasc Surg 1992; 15:1010-1017.
thelialization. J Vasc Surg 1989; 9:588-593. 82. Christ G, Seiffert D, Hufnagl P et al. Type 1 plasminogen
70. Sumpio BE, Widmann MD, Ricotta J et al. Increased activator inhibitor synthesis of endothelial cells is
ambient pressure stimulates proliferation and morphologi- downregulated by smooth muscle cells. Blood 1993;
cal changes in cultured endothelial cells. J Cell Physiol 81:1277-1283.
1994; 158:133-139. 83. Scott-Burden T, Vanhoutte PM. Regulation of smooth
71. Iba T, Sumpio BE. Tissue plasminogen activator expres- muscle cell growth by endothelium-derived growth fac-
sion in endothelial cells exposed to cyclic strain in vivo. tors. Tex Heart Inst J 1994; 21:91-97.
Cell Transpl 1992; 1:43-50.
CHAPTER 5
Surface Precoating in the 1990s:
The Fine Tuning of Endothelial Cell Transplantation
Mark M. Samet, Victor V. Nikolaychik, Peter I. Lelkes
Introduction
E ach year 900,000 people in the USA alone suffer from arterial disorders requiring some
form of surgical intervention. Over half of these procedures involve peripheral recon-
struction using vascular grafts to bypass an occluded vessel segment. However, 60% of the
patients undergoing vascular surgery do not have a suitable vessel for grafting and, there-
fore, need synthetic grafts.1 In this realm, about 350,000 artificial vascular grafts are im-
planted annually, and the numbers are expected to rise rapidly in the future. In spite of the
increasing demand for vascular prostheses, the long term success of their use as arterial
substitutes is, at best, mixed.2,3 In artery bypasses, with bore sizes + 5 mm in diameter Dacron®
and ePTFE grafts demonstrate cumulative patency rates similar to those of the natural grafts.1
But, as small artery substitutes, these prostheses have performed poorly and, to date, no
vascular prosthesis of , 4 mm in diameter remains patent for more than a couple of months.1
The high failure rates of small diameter prosthetic grafts are believed to be intimately asso-
ciated with the poor healing and rejection process of the implanted conduit.3-7 One of the
key determinants in this “cause and effect” relationship is the problem of insufficient
hemocompatibility of the polymeric graft material.8
Insufficient hemocompatibility of the biomaterials is also a pressing issue in the field
of cardiac prostheses. Each year, end-stage heart diseases claim some 75,000 victims in the
USA alone. Although 28,000 of these patients qualify for heart transplants, the supply of
donated hearts is limited to approximately 2,000 hearts per year.9,10 Thus, the majority of
these patients has no realistic hope of long term survival, unless permanent total artificial
hearts (TAH) or ventricular assist devices (VAD) become available.
The vision of developing a permanent cardiac prosthesis is more than a half century
old. The first practical implementation of this idea was achieved in the 1950s by W.J. Kolff
and his collaborators, who kept a dog alive for 90 min after successfully replacing the native
heart with an artificial device.11 By the mid-1970s, the first generation of TAHs and VADs
was undergoing advanced clinical trials.11-14 These units were activated externally by pneu-
matic drivers and connected trough transcutaneous pressure lines. Besides restricting the
patient’s postoperative quality of life, these lines served as a major source for bacterial infec-
tion and initiated thromboembolic complications. The newest generation of cardiac pros-
theses, designed to be totally implantable, are powered by a compact electrical source.14 In
spite of significant mechanical and electronic improvements in the new designs, the perma-
nent or even temporary clinical use of cardiac prostheses as a bridge to recovery15 is still
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
80 Tissue Engineering of Prosthetic Vascular Grafts
thwarted by serious obstacles such as infections and throm- lium by mechanical forces.38,39 Many investigators, however,
boembolic complications which are manifested in stroke and turned to surface treatment with select extracellular matrix
multiple organ failures.16,17 As in the case of artificial vascu- (ECM) proteins or blood plasma proteins (“fibrin glue”),
lar grafts, many of the complications with cardiac prosthe- with or without supplements, as a means for improving cel-
ses can be traced back to the problem of inadequate lular performance in grafts.40,41 Our ongoing studies focus
hemocompatibility of the blood-contacting biomaterials in on surface coating as a means for optimizing endo-
these devices. Thus, it follows that if we can successfully over- thelialization.42,43 Specifically, our motivation is to form a
come the blood compatibility barrier, we shall markedly multifunctional protein complex that will perform as a
improve the clinical prospects of both artificial hearts and biointegrator between the living cells and the underlying
small diameter prosthetic grafts. polymeric scaffold in cardiac prostheses.
the particular type of this protein is of secondary impor- biointegrated proteinaceous meshwork at the PU surface
tance for ECs grown in a static environment. which would structurally mimic the topology of the suben-
Subsequently, we examined whether the effectiveness dothelial basement membrane and also provide space for
of a particular surface coating in promoting cell growth may the incorporation of the endogenous, EC-derived extracel-
depend on the method of protein immobilization onto the lular proteins. Such a provisional ECM meshwork might be
polymeric surface. To this end, we seeded BAEC at a density found in fibrin glue, the reaction product of fibrinogen and
of 25,000 cells/cm2 onto PUs coated by different means with thrombin.
ECM proteins, and monitored their growth for several days. In the past, fibrin glue was shown to be beneficial in
The PU samples consisted of 4 separate groups: supporting both EC attachment and growth on synthetic
Group #1: PU coated with FN by adsorption. The materials40,42,43,55-57 and for establishing a stable, shear-re-
starting concentration of FN was 20 ∝g/ml (pH=7.4). sistant monolayer on ePTFE grafts.58 To improve this coat-
After 1 h of incubation at 37°C, the final concentra- ing we substituted purified fibrinogen with plasma cryopre-
tion on the surface, as determined by the cipitate. Indeed, the results of those studies were encourag-
sulforhodamine B assay,53 was 20 ng/cm2. ing: After 4 days in culture the number of cells on fibrin-
Group #2: PU coated with FN by photoimmo- based coating was higher by almost 45% than on TC. Cell
bilization using the method of Clapper et al.54 The coverage was even higher when the fibrin-based coating was
final concentration of FN on the surface was as in supplemented with heparin and insulin.42 Based on these
group #1. findings, we assessed the efficacy of the fibrin-based coating
Group #3: PU coated with Col IV by adsorption. The to promote EC growth on the luminal surface of PU-cov-
starting concentration was 300 ∝g/ml at pH = 3.0. Af- ered “ersatz” ventricles, (T-25 tissue culture flasks61). For
ter 1 h of incubation at 37°C, the final concentration comparison purposes we also used “ersatz” ventricles cov-
on the surface was 50 ng/cm2. ered with either plain or FN-coated PU. In all cases the lu-
Group #4: PU coated with Col IV by photoimmo- minal surface of the PUs used (Biospan™)50 was smooth
bilization. The final concentration of Col IV on the and the coatings were prepared without supplements such
surface was as in group #3. as growth factors, heparin, or insulin.
As shown in Figure 5.1b, the cells grew equally well on The study was conducted in two sequential steps to
FN substrates, whether adsorbed or photoimmobilized. On realistically mimic a full scale endothelialization of the blood
the other hand, when Col IV-based substrates were used, sac of a VAD. First, the cells (BAECs) were seeded with rota-
cell growth was significantly better on PU coated by adsorp- tion for 3 h onto either precoated or plain PU surfaces.61
tion than by photoimmobilization (1.69 ± 0.11 x 105 vs. 1.33 Thereafter, the “ersatz” ventricles were removed from the ro-
± 0.11 x 105, p < 0.05). Thus, for select proteins some meth- tator and kept in an upright position, and the adherent ECs
ods of immobilization may indeed result in better substrates were allowed to spread and proliferate for several days. We
for cell growth than others. However, in our hands, no ECM- monitored cell growth every other day by Alamar Blue as-
derived, monoprotein coating of PU can match the condi- say.50 The results obtained are presented in Figure 5.2a. As
tions offered by a tissue culture-grade polystyrene. Clearly, expected, the lowest rate of growth was obtained on plain
a substrate comprised of two dimensionally assembled, ECM PU. On FN-coated PU the cells grew somewhat better, pre-
adhesive monoproteins does not provide for an adequate sumably because there were more cells attached immedi-
surface coating that can be used as a vehicle for optimizing ately after seeding and so they could interact more effec-
endothelialization of cardiovascular prostheses. Hence, more tively with one another. In the past, several studies had al-
sophisticated approaches are required to generate a ready shown that such interactions play an important role
in cell proliferation in vitro.62,63 By contrast, cells seeded on (p = 0.24). On both CRY-coated PUs, the number of cells
plasma cryoprecipitate-made fibrin substrate (CRY) grew was about 1.5 times higher than on plain, textured PUs. These
rapidly and reached confluence by the seventh day in culture. data suggest that fibrin-based coating is indeed a preferred
Importantly, purified fibrinogen-made fibrin substrate substrate for optimizing EC growth on the luminal surface
(FBG) was not as effective as CRY in promoting cell growth; of cardiovascular prostheses.
even on the ninth day in culture the number of cells on FBG In addition to enhancing cell growth, increasing the
was still significantly lower than on CRY (2.86 ± 0.28 x 105 initial efficiency of cell attachment onto biomaterials is piv-
vs. 3.41 ± 0.25 x 105, p < 0.05). Since both coatings are otal for successfully establishing an EC monolayer in pros-
comprised of a fibrin-based matrix, it is likely that some of thetic devices. In the past, we and others have explored vari-
the intrinsic plasma proteins contained in CRY may have ous ways for increasing the affinity of synthetic surfaces to
contributed to the enhanced rate of cell growth on cells, for example, by glow discharge treatment,68 coating
this substrate. with substrates composed of select proteins,40,69,70 or both.61
Smooth-surface PUs have been used in cardiac pros- Indeed, when BAECs were seeded under static conditions
theses as a means of preventing platelet aggregation and onto various PU substrates, we saw a substantial improve-
thrombus formation on the blood-contacting surfaces. How- ment in attachment efficiency on treated surfaces vs. plain
ever, notwithstanding the improvements in PU manufac- PU (Fig. 5.3a). Our study also indicated that the most suit-
turing and surface processing, the microscopic appearance able surface for cell attachment was TC; CRY-coated PU was
of the luminal surface in these devices has never been com- second to the best (94.1 ± 3.23%, p < 0.05). In our hands,
pletely smooth, and also the hemocompatibility of the the performance of CRY (and FBG) coatings was depen-
smooth surfaces has not met the initial expectations. On the dent upon the concentration of fibrinogen employed. As
other hand, textured PU surfaces have been advocated as a demonstrated in Table 5.1, cell attachment efficacy of puri-
scaffold of choice for enhancing pseudoneointima forma- fied fibrinogen varied in a bell-shaped manner between con-
tion64,65 and for promoting endothelial cell attachment onto centration levels of 0-30 mg/ml. The best performance was
the luminal surface of cardiovascular devices.29,66,67 In adopt- obtained at 5 mg/ml but, for preparing a coating of control-
ing the latter concept for the endothelialization of VAD blood lable thickness on a textured surface, we used only 3.5 mg/ml.
sacs, we evaluated CRY enhancement of cell growth on tex- Optimal adherence of endothelial cells under static
tured PU. The experiments were conducted in the same conditions onto flat PU substrates or a TC surface does not
manner as above with smooth, uncoated PUs serving as con- necessarily translate into efficient attachment of cells dur-
trols. Figure 5.2b exemplifies our findings for plain and CRY- ing endothelialization under rotation of a complex geomet-
coated PUs after seven days in culture. BAECs proliferated ric structure such as a cardiovascular prosthesis. In our
much faster on the uncoated, textured PU than on its smooth hands, there are four equally important factors that deter-
counterpart, (2.17 ± 0.21 x 105 vs. 1.49 ± 0.21 x 105, p < 0.05). mine the efficiency of cell attachment under these unique
Again, the significant difference in the number of cells cov- conditions: number of cells in the inoculum, speed of rota-
ering the surfaces may have resulted, in part, from more ef- tion, duration of seeding and surface treatment by coating.
ficient attachment of cells on textured surface than on By fine tuning these parameters we were able to im-
smooth PU. Application of CRY coating masked the topo- prove the homogeneity of surface coverage, optimize the time
logical difference between the two types of surfaces and en- of residence/contact of cells with the substrate, and thus
hanced cell growth with the same effectiveness. Thus, the promote cell attachment. We established the following as
number of cells on both PUs was virtually the same at all optimal for lining our artificial ventricles: speed at 10 rota-
times; for example, on the seventh day in culture (Fig. 5.2b) tions/h, duration of seeding at 3 h, and inoculation density
we measured 3.24 ± 0.23 x 105 cells/cm2 on smooth PU+CRY at 6.0 x 104 cells/cm2. Utilizing these settings, we examined
vs. 3.56 ± 0.23 x 10 5 cells/cm 2 on textured PU+CRY the efficiency of attachment of cells that were seeded onto
various substrates in smooth PU-covered “ersatz” ventricles. abundant, randomly dispersed cavities of varying dimen-
Plain TC served as controls. The results obtained are shown sions, ranging in size from 30-120 ∝m in diameter. Figure
in Figure 5.3b. In contrast to the findings under static con- 5.4b exemplifies attachment efficiencies on plain and CRY-
ditions, plain TC was the least suitable substrate for cell at- coated PUs. It is apparent from panel A that cell attachment
tachment under rotation, whereas CRY ranked the best onto an uncoated, textured surface was significantly better
(553.1 ± 21% over TC). FBG ranked second to the best (328.2 than onto plain smooth PU (134.8 ± 8% and 108.2 ± 6%,
± 13% over TC) and FN-based substrate was the third with respectively, compared to TC controls, p < 0.05). This im-
a considerably lower efficiency (257.3 ± 11% over TC). provement was likely due to the increase in cell residence
The topography of the luminal surface in most vascu- time at the site of contact and isolation from fluid currents,
lar grafts is primarily rough.71,72 Also, some of the TAHs and rather than resulting from enhancement in surface affinity.
VADs that are currently tested or developed, including the In the case of CRY-coated PUs, the fibrin meshwork com-
Heartmate® and the Milwaukee Heart™, have a rough lu- pletely filled the pores in the textured PU and masked the
minal surface. To gain a more factual representation of the topological differences between rough and smooth surfaces
attachment efficiencies during endothelialization of artifi- (Figs. 5.4 c,d). The resulting substrates had a finely patterned
cial devices, we repeated the above experiments using “er- luminal surface that was enriched in numerous RGD bind-
satz” ventricles covered with textured PU. Figure 5.4a pre- ing sites inherent to the component proteins. As shown in
sents a typical SEM photomicrograph of a textured surface panel B of Figure 5.4b, cells adhered readily onto both these
that was prepared, as previously reported,43 by sprinkling substrates, leading to similar attachment efficiencies of more
NaCl crystals (in this case the mean diameter was 32 ∝m) than 500% above control level. Thus, by using CRY coating,
over a wet PU cast. The entire surface appears corrugated by we completely eliminated the difference in attachment effi-
ciencies between smooth and rough PUs.
Having optimized cell attachment and growth on PUs,
we determined whether the CRY coating could also enhance
the retention of ECs in a monolayer that is exposed to dy-
namic conditions. For these in vitro studies, we used VAD
blood sacs which were fabricated from a commercially avail-
able PU (Biospan™) by the lost wax technique.43 For cell
seeding, the lumina of fully assembled VADs were filled with
125 ml of Air/CO2-presaturated culture medium contain-
ing 4.0 x 107 BAECs, mounted onto the rotating arm of the
seeding apparatus61 and rotated at 10 rotations/h for 3 h.
Following seeding, the VADs were placed, in an upright po-
sition, in a standard CO2 incubator. Cell proliferation was
measured every other day, and the cultures were refed after
conclusion of each Alamar Blue assay.50 Two days after
seeding, the monolayers in the blood sacs had reached
confluence. We then maintained them for eight additional
days under static conditions to allow the cells to synthesize, ing to pulsatile flow resulted in denudation of the monolay-
assemble, and mature their ECM on the underlying scaf- ers by more than 30%, but did not compromise the mono-
fold.73 Following a total of eleven days in a static environ- layers established on CRY coating. In fact, our results indi-
ment, the monolayers were exposed for 24 h to one of the cate that on CRY coating there was minimal, if any, cell loss
following dynamic conditions: due to exposure to the hemodynamic forces encountered in
1. Flexing of the blood sac wall at 1 Hz between systole a pumping artificial ventricle.
and diastole positions, with minimal flow of medium
inside the blood sac; Morphological Aspects
2. Perfusion of the blood sac in an assembled VAD with of the Endothelialized Surfaces
pulsatile flow at the mean rate of 500 ml/min (blood It has long been recognized that the morphology and
sac wall was maintained motionless during the whole function of vascular ECs, as well as the architecture of the
period); and subendothelial matrix, are modulated by hemodynamic
3. Pumping action of the VAD at 1 Hz, mean flow rate forces and by the tissue-specific microenvironment. In most
of 3.2 l/min, and ejection pressure of 150 mm Hg.
For all tests, the VADs were connected aseptically to a
mock loop consisting of a PVC venous reservoir, as previ-
ously described.43 The status of the monolayer lining the Table 5.2. Cell retention under dynamic conditions in
EC monolayers established on either FN or CRY coating
blood sacs was evaluated by the Alamar Blue assay before
and after each run. Our findings for FN and CRY-based coat-
ings are presented in Table 5.2. It is apparent that 24 h of % of cells remaining % of cells remaining
Dynamic Condition on FN coatinga on CRY coatinga
flexing did not dislodge cells from the scaffolds. In fact, the
rhythmic movement of the wall even increased cell density Flexing 120.7 ± 7.6 * 113.1 ± 3.3 *
markedly in all cultures by more than 13%. This observa- Perfusion 69.0 ± 7.1 * 91.6 ± 13.2
tion is in line with the findings previously reported by oth- Pumping 65.0 ± 16.0 * 95.2 ± 6.3
ers.74 On the other hand, exposure of ECs grown on FN coat- a
The data were normalized to the number of cells measured before
the test; * indicates significant difference, p<0.05.
studies, these characteristics have primarily been addressed and/or assembly of the EC-derived ECM, or if it was an ar-
in the context of cellular response to mechanical forces.74-79 tifact of specimen preparation, is unclear. In any event, the
To gain a better insight into adaptation of the cells on subendothelial matrix in either case did not appear to
different surfaces, we focused on morphological aspects of provide sufficient mechanical stability and sturdiness for
the EC monolayer and the subendothelial region on plain supporting the cells during exposure to the dynamic condi-
and coated PUs in postconfluent, static cultures. After seven tions inside a beating ventricle. By contrast to the scanty ECM
days in culture, the cells were removed by treatment with on plain PUs, the subendothelial regions assembled on FN
detergent and the surface was examined by scanning and CRY coatings were more substantial, oftentimes span-
electron microscopy. ning the entire viewing areas (Figs. 5.5c and d, respectively).
A typical electron microscopic view of the subendo- On FN coated PU, (Fig. 5.5c), the network appeared planar
thelial region of cells grown on uncoated, smooth PU is pre- and in many sites it was much denser than on uncoated PUs.
sented in Figure 5.5a. The ECM, made of isolated regions of CRY alone formed a 3-dimensional and continuous
fibers that were interlaced by short fibrils, formed thin sheets meshwork atop the PU, composed of crosslinked fibrin and
of web-like structures of variable density. These regions only other highly organized adhesive plasma proteins such as
partially covered the PU surface and were connected to one fibronectin and vitronectin (Fig. 5.5d). When the cells grown
another by wider filamentous bands. The subendothelial on top this structure were removed, the remaining ECM was
region on uncoated, textured PU (Fig. 5.5b) appeared simi- comprised of thick bands of extremely dense structures,
lar to the ECM on smooth PU, except that oftentimes the closely apposed to the CRY meshwork, resembling the
long fibrillar structures bridged across cavities and recesses amorphous, natural basement membrane. Apparently the
within the surface. On both PUs, however, a large fraction architectural differences between the two coatings, as
of the surface area appeared devoid of any matrix-related manifested in their improved scaffolding capabilities and,
material. Whether this was due to nonuniform deposition ultimately, in their potential to minimize cell loss during
Fig. 5.5. SEM en face view of the subendothelial region after seven days of cell culturing under static conditions
on: (a) uncoated, smooth PU, (b) uncoated, textured PU, (c) FN-coated textured PU, and (d) CRY-coated
textured PU. BAECs were grown in blood sacs as described in the text. On seventh day, select samples of 1 x 1 cm2
were excised from the blood sacs and rinsed with PBS. The cells were removed from the surface by treatment
with 0.1% Triton X-100 and subsequent wash with 0.025 M NH4OH. The samples were rinsed with 0.1 M Na-
cacodylate buffer and, consecutively, with ddH2O. Following serial dehydration (30 sec each) by ethanol and
critical point drying, specimens from each sample were sputter coated with gold-palladium (~210 Å in thick-
ness) and viewed by Philips PW-6700 microscope (Eindhoven, The Netherlands) at a magnification of x2500).
86 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 5.6. Scanning electron microscopic view of: (a) CRY-meshwork before cell seeding (magnification x2500), (b) Confluent and
well-differentiated BAEC monolayer after 7 days in static culture (magnification x650), and (c) Shrinkage gap in the BAEC mono-
layer which was formed during specimen processing in order to provide a detailed insight into the subendothelial region; * indi-
cates a section of the extracellular matrix deposited and organized by BAEC (magnification x2500). On the seventh day, select
samples of 1 x 1 cm2 were excised from the blood sacs and rinsed with PBS. The samples were fixed for 30 min at room temperature
with 1% glutaraldehyde in 0.1 M Na-cacodylate buffer and postfixed with OsO4 for 25 min. Further processing followed the
procedure described in Figure 5.5.
Surface Precoating in the 1990s: The Fine Tuning of Endothelial Cell Transplantation 87
sional meshwork of cryoprecipitate-derived plasma proteins Currently, the focus of our work and that of others is
that serves as a biointegrator between the endothelial cell to further optimize the hemocompatibility of the blood-
monolayer and the underlying synthetic scaffold. In this contacting surfaces in cardiac prostheses by broadening the
chapter we have presented our experience with the plasma concept of “endothelialization”. Over the years, progress in
cryoprecipitate-produced fibrin meshwork as it relates to op- this area has been made largely by extending the idea of a
timizing endothelial cell attachment and growth, and to in- “passive endothelial cell lining”, as applied in the 1980s and
creasing resistance to denudation of the cellular monolayer before, to the concept of biointegration of the textured poly-
upon exposure to the dynamic conditions inside a beating meric scaffold, the 3-dimensional matrix and the “active”
cardiac prosthesis. interaction between the cells and their matrix. This integral
Our in vitro studies establish that denudation of approach requires the coordinated input of a
endothelialized prostheses can be minimized by culturing multidisciplinary team of scientists, and is only one of the
ECs on a 3-dimensional meshwork, rich in both structural many steps needed for the recreation of the entire vascular
and adhesive proteins, as well as by allowing the confluent wall structure lined by a nonthrombogenic cellular lining.
monolayer time for maturation. All these factors, viz, seed- In the past it has been assumed that this cellular lining should
ing of endothelial cells on the three dimensional “provisional be comprised only of endothelial cells, but recently there is
matrix” and subsequent establishment of the subendothe- some evidence that other cell types, either native or geneti-
lial ECM, as well as the functional maturation of the EC cally modified, can also be rendered nonthrombogenic when
monolayer, will likely play vital roles in averting denudation exposed to pulsatile blood flow.
of ECs by mechanical forces. Fine tuning of the surface coat- Reconstruction of the cardiac/vascular wall in vitro
ing will confer added physical stability to the neobasement requires a thorough understanding of how to coculture the
membrane (“shock absorber effect”) and promote functional diverse cell types comprising the entire vascular wall, e.g.,
contacts between the cells in the monolayer. However, once smooth muscle cells, fibroblasts and endothelial cells, in or-
in contact with blood/plasma, other factors may also be of der to achieve a hierarchic, structural and functional orga-
importance for the maintenance of an intact EC monolayer. nization resembling that of the adventitia, media and in-
For example, monolayer integrity may be challenged by vari- tima. Interestingly, preliminary data suggest that exposure
ous proteases, activated plasma proteins and/or circulating of such cocultures to hemodynamic forces such as cyclic
blood cells when the cell culture media is replaced by plasma strain and pulsatile flow will sort out the cells and initiate
or citrated blood during mock loop tests in vitro. Similar, their hierarchic arrangement. Unanswered, as yet, is the ques-
and even more demanding, challenges should also be ex- tion of whether the cells lining the blood-contacting sur-
pected during in vivo trials. faces are indeed functional, viz., whether the cellular lining
will indeed reduce the thrombogenicity of the devices. An
88 Tissue Engineering of Prosthetic Vascular Grafts
exciting approach is to use genetically modified cells, which totypes, policies, and patients. Washington DC: National
would accelerate the hierarchic organization within the ves- Academy Press, 1991:251-261.
sel wall and/or overexpress specific molecules that charac- 10. Marshall E. Artificial heart: The beat goes on. Science
terize the anticoagulant/nonthrombogenic endothelial cells’ 1991; 253:500-502.
11. Kolff WJ. Experiences and practical considerations for the
surface.
future of artificial hearts and of mankind. Artif Organs
Finally, in our opinion, the ultimate cardiovascular 1988; 12:89-111.
“prosthesis” will be a man-made, tissue-engineered replace- 12. Joyce LD, DeVries WC, Hastings WL et al. Response of
ment organ. In the not too distant future we envisage that the human body to the first permanent implant of the
dramatic progress in cell and molecular biology, in concert Jarvik-7 total artificial heart. Trans Am Soc Artif Intern
with novel approaches to 3-dimensional tissue culturing, will Organs 1983; 29:81-87.
yield functional replacement tissue equivalents and, perhaps, 13. Bernhard WF. A fibrillar blood-prosthetic interface for
even entire organs. Some of these exciting procedures, as both temporary and permanent ventricular assist devices:
futuristic as they may sound, are currently being explored Experimental and clinical observations. Artif Organs 1989;
by us and several other groups. Hence, the dream of gener- 13:255-271.
ating ex vivo autologous replacement “spare parts”, has ma- 14. Nose Y. My life with the National Institutes of Health
Artificial Heart program. Artif Organs 1990; 14:174-190.
terialized during the past few years and appears to be achiev-
15. Westaby S. The need for artificial hearts. Heart 1996;
able. We truly believe that our current struggle to optimize 76:200-206.
the design and hemocompatibility of cardiovascular 16. Ward RA, Wellhausen SR, Dobbins JJ et al. Thromboem-
bioprostheses will soon be surpassed by a new area of “as bolic and infectious complications of total artificial heart
good as new” tissue-engineered replacement organs, recre- implantation. Ann N Y Acad Sci 1987; 516:638-650.
ated from the patients’ own cells. 17. Green K, Liska J, Egberg N et al. Hemostatic disturbances
associated with implantation of an artificial heart. Thromb
Acknowledgments Res 1987; 48:349-362.
The present study was supported by the Milwaukee 18. Yozu R, Golding LA, Jacobs G et al. Preclinical evalua-
Heart™ Research Foundation. We thank the entire engineer- tion of a biolized temporary ventricular assist device.
ing group of the Milwaukee Heart™ Project for providing Trans Am Soc Artif Intern Organs 1989; 35:556-558.
19. Gimbrone MA Jr. Vascular endothelium: Nature’s blood-
the polyurethane sheets, blood sacs and VADs for this study.
compatible container. Ann N Y Acad Sci 1987; 516:5-11.
We are also grateful to Mrs. D. M. Wankowski for her skill- 20. Mansfield PB, Wechezak AR, Sauvage LR. Preventing
ful technical assistance in performing the experiments, to thrombus on artificial vascular surfaces: True endothelial
Mrs. I. Hernandez for her technical expertise in conducting cell linings. Trans Am Soc Artif Intern Organs 1975;
the SEM studies, and to Mr. M. D. Silverman for his helpful 21:264-272.
technical suggestions and critical comments. 21. In: Simionescu N, Simionescu M, eds. Endothelial cell
dysfunctions. New York: Plenum Press, 1992:1-565.
References 22. In: Simionescu N, Simionescu M, eds. Endothelial cell
1. Moneta GL, Porter JM. Arterial substitutes in peripheral biology in health and disease. New York: Plenum Press,
vascular surgery: A review. J Long-Term Effects of Medi- 1988:1-458.
cal Implants 1995; 5:47-67. 23. Dunn PF, Newman KD, Jones M et al. Seeding of vascu-
2. Noishiki Y. Progress and problems in the development of lar grafts with genetically modified endothelial cells: Se-
vascular prostheses. Artif Organs 1995; 19:3-6. cretion of recombinant TPA results in decreased seeded
3. Greisler HP. Currently available vascular prostheses. In: cell retention in vitro and in vivo. Circulation 1996;
Greisler HP, ed. New biologic and synthetic vascular pros- 93:1439-1446.
theses. Austin: R.G. Landes Co., 1991:20-32. 24. Williams SK, Rose DG, Jarrell BE. Microvascular endot-
4. Jarrell BE. Vascular and endothelial cell technology. In: helial cell sodding of ePTFE vascular grafts: Improved
Skalak R, Fox CF, eds. Tissue engineering. New York: Alan patency and stability of the cellular lining. J Biomed Mater
R. Liss, Inc., 1988:1-3. Res 1996; 28:203-212.
5. Choi ET, Callow AD. The effect of biomaterials on the 25. Urayama H, Kasashima F, Kawakami T et al. An immu-
host. In: Greco RS, ed. Implantation biology: The host nohistochemical analysis of implanted woven Dacron and
response and biomedical devices. Boca Raton: CRC Press, expanded polytetrafluoroethylene grafts in humans. Artif
1994:39-53. Organs 1996; 20:24-29.
6. Yeager A, Callow AD. New graft materials and current 26. Brunstedt MR, Ziats NP, Rose-Caprara V et al. Attach-
approaches to an acceptable small diameter vascular graft. ment and proliferation of bovine aortic endothelial cells
Trans Am Soc Artif Intern Organs 1988; 34:88-94. onto additive modified poly(ether urethane ureas). J
7. Noishiki Y, Tomizawa Y, Yamane Y, Matsumoto A. The Biomed Mater Res 1993; 27:483-492.
vicious cycle of nonhealing neointima in fabric vascular 27. Park PK, Jarrell BE, Williams SK et al. Thrombus-free,
prostheses. Artif Organs 1995; 19:7-16. human endothelial surface in the midregion of a Dacron
8. Sheppeck RA, LoGrefo FW. Blood and biomaterials. In: vascular graft in the splanchnic venous circuit: Observa-
Greco RS, ed. Implantation biology: The host response tions after nine months of implantation. J Vasc Surg 1990;
and biomedical devices. Boca Raton: CRC Press, 11:468-475.
1994:55-65. 28. Schneider PA, Hanson SR, Price TM, Harker LA.
9. Funk M. Epidemiology of end-stage heart disease. In: Confluent durable endothelialization of endarterectomized
Hognes JR, VanAntwerp M, eds. The artificial heart: Pro- baboon aorta by early attachment of cultured endothelial
cells. J Vasc Surg 1990; 11:365-372.
Surface Precoating in the 1990s: The Fine Tuning of Endothelial Cell Transplantation 89
29. Okoshi T, Soldani G, Goddard M, Galletti PM. Very prosthesis: The effect of growth-support surface chemis-
small-diameter polyurethane vascular prostheses with try, cell seeding density, ECM protein coating, and growth
rapid endothelialization for coronary artery by pass graft- factors. Art Cells Blood Subs and Immob Biotech 1996;
ing. Thorac Cardiovasc Surgeon 1993; 105:791-795. 24:51-63.
30. Baitella-Eberle G, Groscurth P, Zilla P et al. Long-term 49. Lin H-B, Sun W, Mosher DF et al. Synthesis, surface, and
results of tissue development and cell differentiation on cell-adhesion properties of polyurethanes containing co-
Dacron prostheses seeded with microvascular cells in dogs. valently grafted RGD-peptides. J Biomed Mater Res 1994;
J Vasc Surg 1993; 18:1019-1028. 28:329-342.
31. Van Belle E, Tio FO, Couffinhall T et al. Stent 50. Nikolaychik VV, Samet MM, Lelkes PI. A new method
endothelialization: Time course, impact of local catheter for continual quantitation of viable cells on
delivery, feasibility of recombinant protein administration, endothelialized polyurethanes. J Biomater Sci Polymer Edn
and response to cytokine expedition. Circulation 1997; 1996; 7:881-891.
95:438-448. 51. Nichols WK, Gospodarowicz D, Kessler TR, Olsen DB.
32. Langer R, Vacanti JP. Artificial organs. Sci Am 1995; Increased adherence of vascular endothelial cells to Biomer
273:130-133. precoated with extracellular matrix. Trans Am Soc Artif
33. Wilson JM, Birinyi LK, Salomon RN et al. Implantation Intern Organs 1981; 27:208-212.
of vascular grafts lined with genetically modified endot- 52. Seeger JM, Klingman N. Improved in vivo
helial cells. Science 1989; 244:1344-1346. endothelialization of prosthetic grafts by surface modifi-
34. Callow AD. The vascular endothelial cell as a vehicle for cation with fibronectin. J Vasc Surg 1988; 8:476-482.
gene therapy. J Vasc Surg 1990; 11:793-798. 53. Skehan P, Storeng R, Scudiero D et al. New colorimetric
35. Nabel EG, Plautz G, Nabel GJ. Transduction of a foreign cytotoxicity assay for anticancer-drug screening. J Natl
histocompatibility gene into the arterial wall induces vas- Cancer Inst 1990; 13:1107-1112.
culitis. Proc Natl Acad Sci USA 1992; 89:5157-5161. 54. Clapper DL, Daws KM, Guire PE. Photoimmobilized ECM
36. Koh GY, Kim S-J, Klug MG et al. Targeted expression of peptides promote cell attachment and growth on
transforming growth factor-! in intracardiac grafts pro- biomaterials. 20 th Annual Meeting of the Society for
motes vascular endothelial cell DNA synthesis. J Clin In- Biomaterials 1994; 345 (Abstract).
vest 1995; 95:114-121. 55. Zilla P, Fasol R, Preiss P et al. Use of fibrin glue as a
37. Kottke-Marchant K, Veenstra AA, Marchant RE. Human substrate for in vitro endothelialization of PTFE vascular
endothelial cell growth and coagulant function varies with grafts. Surgery 1989; 105:515-522.
respect to interfacial properties of polymeric substrates. J 56. Sirois E, Côte MF, Doillon CJ. Growth factors and bio-
Biomed Mater Res 1996; 30:209-220. logical supports for endothelial cell lining: In vitro study.
38. Ott MJ, Ballermann BJ. Shear stress-conditioned, endot- Int J Artif Organs 1993; 16:609-619.
helial cell-seeded vascular grafts: Improved cell adherence 57. Gosselin C, Vorp DA, Warty VS et al. ePTFE coating with
in response to in vitro shear stress. Surgery 1995; fibrin glue, FGF-1, and heparin: Effect on retention of
117:334-339. seeded endothelial cells. J Surg Res 1996; 60:327-332.
39. Ballermann BJ, Ott MJ. Adhesion and differentiation of 58. Muller-Glauser W, Zilla P, Lachat M et al. Immediate
endothelial cells by exposure to chronic shear stress: A shear stress resistance of endothelial cell monolayers
vascular graft model. Blood Purif 1995; 13:125-134. seeded in vitro on fibrin glue-coated ePTFE prostheses.
40. Kaehler J, Zilla P, Fasol R et al. Precoating substrate and Eur J Vasc Surg 1993; 7:324-328.
surface configuration determine adherence and spreading 59. DePalma L, Criss VR, Luban NLC. The preparation of
of seeded endothelial cells on polytetrafluoroethylene fibrinogen concentrate for use as fibrin glue by four dif-
grafts. J Vasc Surg 1989; 9:535-541. ferent methods. Transfusion 1993; 33:717-720.
41. Gosselin C, Ren D, Ellinger J, Greisler HP. In vivo plate- 60. In: Harris JR, ed. Blood separation and plasma fraction-
let deposition on polytetrafluoroethylene coated with fi- ation. New York: John Wiley & Sons, 1991:1-497.
brin glue containing fibroblast growth factor 1 and hep- 61. Wankowski DM, Samet MM, Nikolaychik VV, Lelkes PI.
arin in a canine model. Am J Surgery 1995; 170:126-130. Endothelial cell seeding with rotation of a ventricular
42. Nikolaychik VV, Samet MM, Lelkes PI. A new, cryopre- blood sac. ASAIO J 1994; 40: M319-M324.
cipitate based coating for improved endothelial cell at- 62. Gimbrone MA, Jr., Cotran RS, Folkman J. Human vascu-
tachment and growth on medical grade artificial surfaces. lar endothelial cells in culture. J Cell Biol 1974; 60:673-
ASAIO J 1994; 40: M846-M852. 684.
43. Nikolaychik VV, Wankowski DM, Samet MM, Lelkes PI. 63. Schor AM, Schor SL, Allen TD. Effects of culture condi-
In vitro testing of endothelial cell monolayers under dy- tions on the proliferation, morphology and migration of
namic conditions inside a beating ventricular prosthesis. bovine aortic endothelial cells. J Cell Sci 1983; 62:267-285.
ASAIO J 1996; 42: M487-M494. 64. Dasse KA, Chipman SD, Sherman CN et al. Clinical ex-
44. Horwitz AF, Thiery JP. Cell-to-cell contact and extracel- perience with textured blood contacting surfaces in ven-
lular matrix. Curr Opin Cell Biol 1994; 6:645-647. tricular assist devices. ASAIO J 1987; 10:418-425.
45. Ruoslahti E. RGD and other recognition sequences for 65. Graham TR, Dasse K, Coumbe A et al. Neo-intimal de-
integrins. Annu Rev Cell Dev Biol 1996; 12:697-715. velopment on textured biomaterial surfaces during clini-
46. In: Hay ED, ed. Cell biology of extracellular matrix. 2nd cal use of an implantable left ventricular assist device. Eur
Edn. New York: Plenum Press, 1991:1-468. J Cardiothorac Surg 1990; 4:182-190.
47. Fasol R, Zilla P. Endothelialization of artificial heart ma- 66. Okoshi T. New concept of microporous structure in small
terials. In: Unger F, ed. Assisted Circulation 3. Berlin: diameter vascular prostheses. Artif Organs 1995; 19:27-31.
Springer-Verlag, 1989:580-598. 67. Okoshi T, Soldani G, Goddard M, Galletti PM.
48. Sipehia R, Martucci G, Lipscombe J. Transplantation of Microporous polyurethane inhibits critical mural throm-
human endothelial cell monolayer on artificial vascular bosis and enhances endothelialization at blood-contacting
90 Tissue Engineering of Prosthetic Vascular Grafts
surface. In: Akutsu T, Koyanagi H, eds. Heart replace- cytoskeletal protein synthesis during cyclic tensional de-
ment: Artificial heart 5. Tokyo: Springer-Verlag, formation. J Vasc Surg 1988; 7:130-138.
1996:47-52. 76. Barbee KA, Davies PF, Lal R. Shear stress-induced reor-
68. Patterson RB, Messier A, Valentini RF. Effects of ganization of the surface topography of living endothelial
radiofrequency glow discharge and oligopeptides on the cells imaged by atomic force microscopy. Circ Res 1994;
attachment of human endothelial cells to polyurethane. 74:163-171.
ASAIO J 1995; 41: M625-M629. 77. Nerem RM, Levesque MJ, Sato M. Vascular dynamics and
69. Greisler HP. Endothelial cell transplantation onto syn- the endothelium. In: Schmid-Schonbein GW, Woo SL-Y,
thetic vascular grafts: Panacea, poison or placebo. In: Zweifach BW, eds. Frontiers in biomechanics. New York:
Greisler HP, ed. New biologic and synthetic vascular pros- Springer-Verlag, 1986:324-341.
theses. Austin: R.G. Landes Co., 1991:47-64. 78. Girard PR, Helmlinger G, Nerem RM. Shear stress effects
70. Thomson GJL, Vohra RK, Carr MH, Walker MG. Adult on the morphology and cytomatrix of cultured vascular
human endothelial cell seeding using expanded endothelial cells. In: Frangos JA, ed. Physical forces and
polytetrafluoroethylene vascular grafts: A comparison of the mammalian cell. San Diego: Academic Press, Inc.,
four substrates. Surgery 1991; 109:20-27. 1993:193-222.
71. Marchant RE, Wang I-W. Physical and chemical aspects 79. Thoumine O, Nerem RM, Girard PR. Changes in organi-
of biomaterials used in humans. In: Greco RS, ed. Im- zation and composition of the extracellular matrix un-
plantation biology: The host response and biomedical derlying cultured endothelial cells exposed to laminar
devices. Boca Raton: CRC Press, 1994:13-38. steady shear stress. Lab Invest 1995; 73:565-576.
72. Frazier OH, Kadipasaoglu KA, Parnis SM, Radovancevic 80. Hirai J, Kanda K, Oka T, Matsuda T. Highly oriented,
B. Biomaterials used in cardiac surgery. In: Greco RS, ed. tubular hybrid vascular tissue for a low pressure circula-
Implantation biology: The host response and biomedical tory system. ASAIO J 1994; 40: M383-M388.
devices. Boca Raton: CRC Press, 1994:165-178. 81. Matsuda T, Takaichi S, Kitamura T et al. A reconstituted
73. Lelkes PI, Samet MM. Endothelialization of the luminal vessel’s wall model on microporous substrates as a small-
sac in artificial cardiac prostheses: A challenge for both caliber artificial graft. In: Akutsu T, Koyanagi H, Takatani
biologists and engineers. J Biomech Eng 1991; S et al., eds. Artificial heart 2. Tokyo: Springer-Verlag,
113:132-142. 1988:65-73.
74. Iba T, Sumpio BE. Morphological response of human 82. Hirai J, Matsuda T. Self-organized, tubular hybrid vascu-
endothelial cells subjected to cyclic strain in vitro. lar tissue composed of vascular cells and collagen for low-
Microvasc Res 1991; 42:245-254. pressure-loaded venous system. Cell Transplantation 1995;
75. Sumpio BE, Banes AJ, Buckley M, Johnson G Jr. Alter- 4:597-608.
ations in aortic endothelial cell morphology and
PART II
Biolized Prostheses: Surface Healing
Microvascular Endothelial Cell Transplantation
CHAPTER 6
Microvascular Endothelial Cell Transplantation: A Review
Stuart K. Williams
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
94 Tissue Engineering of Prosthetic Vascular Grafts
Publication of these studies established the feasibility methods provided a means to produce large quantities of
of performing endothelial cell transplantation and resulted autologous endothelial cells by obtaining a small segment
in extensive work to optimize seeding technology. Numer- of a patient’s blood vessel, isolating and culturing the en-
ous questions were identified which needed to be addressed dothelium in heparin and endothelial cell growth supple-
before endothelial cell transplantation was expected to be ment (ECGS) and transplanting these cells at very high seed-
found clinically acceptable. These questions identified nu- ing densities onto prosthetic grafts.
merous issues related to the source of tissue for subsequent Large vessel-derived endothelial cell seeding at high
endothelial cell transplantation, the function of immediately density was achieved in vitro; however, additional questions
isolated endothelium and methods to improve the interac- have been raised, delaying wide spread clinical utilization of
tion of endothelium with prosthetic graft surfaces. Subse- this technique. Concerns related to the use of cultured en-
quent laboratory work has established the efficacy of intra- dothelium for cell transplantations are surmountable.
operative methods for endothelial cell isolation and deposi- Dr. Peter Zilla has performed a careful series of studies to
tion onto the lumenal surface of vascular grafts. address the major concerns raised about using cultured cells,
as well as providing evidence of the efficacy of this form of
Sources of Endothelium for Transplantation tissue engineering.14-16 Some of these questions included
As described above, Herring first reported the use of concerns relating to the derivation of cells from large vessel
venous-derived endothelium for subsequent transplantation. sources, which requires a separate surgical procedure, most
The yield of cells from a suitable piece of vein segment was likely from patients with an already compromised circula-
approximately 1 x 104 cells.9 Thus, based on the largest piece tory system. This increased risk of a separate vascular com-
of vein which could be obtained from a patient, only 10,000 plication had to be weighed against the need for endothelial
endothelial cells could be obtained. Under even the most cell transplantation. If cultured cells were used for trans-
ideal conditions, this cell number would provide a relatively plantation, the conditions of culture must be evaluated to
sparse coating of cells, requiring extensive cell proliferation determine the effect on subsequent cell function. Bovine
to achieve a monolayer. Kempzinski et al10 reported that even derived serum components such as viruses, and growth fac-
if cells could be isolated and deposited on the surface of tors such as heparin and ECGS, would be carried with trans-
polymeric grafts with optimal efficiency, a large number of planted cells into patients, requiring characterization of their
these cells would be lost from the lumenal surface of the effects. Considering that endothelial cells have an extremely
graft due to the effects of blood flow-induced shear on the low mitotic index, the stimulation of endothelial cell growth
lumenal surface. Endothelial cells seeded onto grafts with must be considered to determine how cell growth may alter
this technology would be required to undergo even more endothelial cell function. Finally, little data is available con-
extensive proliferation or, alternatively, a new source of en- cerning the appropriateness of transplanting vein-derived
dothelial cells obtained in large numbers must be found. endothelial cells into an arterial circulation with respect to
the ability of these cells to differentiate and function under
Cultured Endothelium for Transplantation arterial conditions. Nonetheless, this technique has been
Methods for the isolation and culture of nonhuman successfully implemented in a series of patients undergoing
endothelial cells were first reported in the 1920s, providing peripheral bypass surgery. Initial results indicate both the
methods for the expansion of endothelial cell numbers by safety and efficacy of large vessel endothelial cell
in vitro proliferation.11 However, the same methods used transplantation.16
for nonhuman endothelial cell proliferation had no stimu-
latory effect on human endothelial cell proliferation. Dur- Current Considerations in Endothelial Cell
ing the 1970s a method for human endothelial cell isola- Transplantation
tion, based on the pioneering work of Lewis in 1922, pro-
vided human endothelial cells in culture; however, these cells Use of Microvessel Endothelium for Tissue Engineering
exhibited minimal proliferative capacity. A number of en- of Vascular Grafts
dothelial cell growth factors were discovered that slightly Given the small numbers of cells available if autolo-
stimulated human endothelial cell proliferation. The major gous vein-derived endothelium are used without culture, and
breakthrough in human endothelial cell growth in culture concerns about culturing effects, investigators have been
was work performed by Dr. Susan Thornton, working with considering alternate sources of endothelial cells for trans-
Dr. Elliot Levine, which identified heparin as an integral plantation. In 1986 Jarrell and Williams17 reported methods
cofactor with endothelial cell growth factor for the stimula- for the isolation of autologous microvessel endothelial cells
tion of human umbilical vein endothelial cell growth in cul- from adipose tissue for use in cell transplantation. The source
ture.12 Jarrell and coworkers13 subsequently reported that of fat for this EC isolation was initially fat deposits associ-
heparin and ECGF would stimulate human adult endothe- ated with omentum. This fat is well vascularized, with a
lial cell growth in culture. For the first time, cells from hu- density of endothelial cells in excess of 106 endothelial cells
man artery and vein segments from numerous anatomical per gram of fat isolated. The need to place cells in culture to
positions could be grown in large quantities. Large num- increase cell number is obviated by the large amounts of
bers of human endothelia could be produced from single endothelium available per gram of omental associated fat.
endothelial cells, providing nearly unlimited supplies of However, access to omental-associated fat still would
human adult endothelium. These breakthroughs in culture
Microvascular Endothelial Cell Transplantation: A Review 95
necessitate a separate surgical procedure, with skin incision or trocar puncture to insert the liposuction
related complications. cannula. The liposuction cannulas used for this procedure
The use of omental-associated fat-derived endothe- have been continuously improved for plastic and cosmetic
lial cells was questioned by Visser et al,18 who reported that surgical applications.
omental tissue derived from humans contains predomi- The use of liposuction fat is also advantageous since
nantly mesothelial cells and not endothelium. This report the tissue is effectively minced during the liposuction re-
raised a significant controversy concerning the use of omen- moval process. Scanning electron microscopic evaluation of
tal-associated fat as a source of endothelial cells for trans- liposuction fat (Fig. 6.1) illustrates the morphological char-
plantation. Since this initial report by Visser et al, subsequent acteristics of this tissue. The predominant cell type present
reports have clarified the controversy and provided addi- at this magnification is adipocytes. Recently, a complete char-
tional options for obtaining microvessel endothelial cells.19 acterization of cells present in liposuction-derived fat was
First it was established that investigators have erroneously reported and established that the major cell type, other than
used the terms omentum and omental fat interchangeably. adipocytes, present in this tissue is endothelium.19 Prior to
These two tissues are histologically and anatomically dis- any cell dissociation procedures, human subcutaneous fat
tinct. While omentum as used by Visser et al is a vascular- contains in excess of 85% endothelium based on the total
ized tissue with an extensive number of mesothelial cells cells present per unit volume of fat. Adipocytes account for
present, omental-associated fat, as used by Jarrell and Will- approximately 12% of the cell population, again before any
iams, is composed of predominantly endothelial cells and tissue dissociation and cell isolation techniques are used.
adipocytes. This is not to say that use of omental-associated Therefore, following tissue dissociation and cell isolation,
fat avoids the possibility of mesothelial cells in the primary endothelial cells must account for at least 85% of the cells in
cell isolate. Omental fat deposits are covered by a thin layer the final inoculum.
of mesothelium, and a single cell population isolated from
omental-associated fat will contain a small number of me- Markers for Isolated Endothelial Cells
sothelial cells in addition to endothelium. Careful isolation One significant difficulty in establishing the identity
of omental-associated fat away from omentum will reduce of cells present in the primary isolate following tissue diges-
contamination by mesothelium. tion has been the relative lack of a dependable marker of
Alternate anatomic sources for microvascularized au- endothelial cells. While antibodies directed against von
tologous human fat for endothelial cell transplantation have Willebrand factor or factor VIII-related antigen (FVIIIrAg)
been reported since these original reports using omentum- have been used somewhat routinely for endothelial cell iden-
associated fat. A significant improvement was the identifi- tification, these markers do not unfailingly react with freshly
cation of subcutaneous fat as a source of endothelial cells isolated microvascular endothelial cells. First, upon primary
and the use of liposuction to derive this fat from patients.20 isolation and due in part to the action of proteolytic en-
A patient undergoing endothelial cell transplantation there- zymes, endothelial cells will release these proteins from cy-
fore does not need to undergo a laparotomy to remove omen- toplasmic granules, thus resulting in cells which exhibit nega-
tal fat or undergo a separate vascular procedure to remove a tive staining properties.21 Also confounding is the relative
segment of vein. A small amount (50 cc) of fat can be re- lack of Weibel-palade bodies in fat-derived microvessel en-
moved using a hand held syringe cannula device. This pro- dothelial cells. The cytoplasmic inclusions contain extremely
cedure requires less then five minutes and requires a small high concentrations of vWF, which subsequently stain
extremely well in cell types such as vein-derived endothe- Cell Deposition on Graft Surfaces:
lium. Venous endothelial cells contain a significant number Seeding vs. Sodding
of Weibel-palade bodies, such that investigators who observe The availability of large quantities of autologous en-
vWF staining in microvessel endothelial cells correctly assess dothelial cells does not necessarily overcome another major
cells with limited reaction product as compared to venous concern related to endothelial cell transplantation. The depo-
endothelium. Often investigators will suggest that since the sition of endothelial cells onto the lumenal (blood flow)
bright punctate staining common with Weibel-palade bod- surface of vascular grafts is a critical step in the transplanta-
ies is not observed in microvascular endothelial cells, these tion process. Once placed as an interpositional graft, cells
cells must not be endothelium. On the contrary, microvas- on the lumenal surface must resist the flow of blood and
cular endothelium is phenotypically distinct from venous remain adherent to the polymeric surface during the matu-
or arterial endothelial cells and shows appropriate differ- ration of an endothelial cell monolayer. The original meth-
ences in markers such as vWF and FVIIIrAg. Upon primary ods used to transplant endothelial cells onto graft surfaces
culture of microvessel endothelial cells, the morphology of- were termed seeding procedures, since relatively low con-
ten takes on a fibroblastic appearance and, in concert with centrations of cells were placed in suspension in either whole
the relatively minimal reaction with vWF antibodies, inves- blood or plasma, and this suspension transferred to the in-
tigators will often conclude that the predominant cell type ner lining of vascular grafts. The endothelial cells in this in-
in fat is fibroblasts. What is actually being observed is the oculum were subsequently expected to proliferate and mi-
ability of endothelial cells to phenotypically differentiate grate to the blood flow surface, creating a contiguous lining
under different in vitro and or in vivo conditions. Interest- of cells. An alternate form of transplantation, described us-
ingly, a recent report suggests that fibroblasts can differenti- ing the term sodding, was subsequently suggested and evalu-
ate in vivo into endothelial cells. This phenotypic drift may ated in vitro as well as in preclinical animal and human stud-
be a common characteristic of pluripotent mesenchymal ies.24 Sodding describes a method of endothelial cell depo-
cells, with possible utilization in future cell transplantation sition wherein a concentration of cells which would pro-
technologies. vide a confluent density of cells on the lumenal graft surface
The methods for the isolation of endothelial cells from is forcibly deposited onto the graft lumenal surface. This
fat are essentially modifications of the methods first reported forced deposition is most easily carried out by establishing a
by Wagner in 1972 for the isolation of endothelial cells from pressure gradient across the flow surface of the graft whereby
rat epididymal fat pads.22 Human fat microvessel endothe- the cells are essentially filtered onto the surface of the graft.
lial cells are isolated by first digesting the fat with a pro- While previous methods used gravity to allow cell deposi-
teolytic enzyme mixture composed predominantly of colla- tion, sodding of cells using pressure deposition results in
genase and trypsin. The characteristics of this enzyme are near immediate association of cells with the lumenal sur-
critical for successful isolation of endothelial cells from fat.23 face. The time to prepare a tissue engineered vascular graft
Following collagenase digestion the slurry is centrifuged, for implantation, a period inclusive of tissue isolation, cell
resulting in the separation of buoyant adipocytes from more isolation and cell deposition, has effectively been reduced to
dense endothelium. The endothelial cell rich pellet is then less than 60 minutes.20
used for cell transplantation procedures.
Animal Models of Endothelial formation of an endothelial cell lining, but questions have
Cell Transplantation been raised regarding whether the cells present on the lu-
The availability of methods for endothelial cell isola- menal surface of microvessel sodded grafts represent the
tion and culture, as well as methods to deposit cells onto same cells transplanted during the initial sodding procedure.
graft surfaces, has resulted in numerous investigations to This question has been addressed using a method to trace
evaluate the efficacy of endothelial cell transplantation.25-33 the fate of transplanted endothelial cells using a fluorescent
The earliest animal models were developed to evaluate the tagging method.35 A fluorescent dye is permanently incor-
ability to accelerate the formation of endothelial cell linings porated into the membranes of microvessel endothelial cells
on polymeric grafts. Most of these studies have been at the time they are transplanted onto the lumenal surface
performed in the canine model, due primarily to the accep- of vascular grafts. This dye has unique characteristics in that
tance of this animal model as a predictor of graft function it is long lived and provides a marker of the fate of trans-
in humans. Earliest studies established the ability to planted cells. If, however, fluorescently labeled cells dupli-
accelerate the formation of a continuous monolayer of cate, each daughter cell receives one half the fluorescence of
endothelial cells on the lumenal surface of vascular grafts the initial cell. For cells which undergo multiple duplica-
(Fig. 6.2). In general, monolayer formation is highly depen- tion, a process which could occur following cell transplan-
dent on the density of cells used to treat the graft surface. tation, the subsequent progeny would contain undetectable
When cells are transplanted at confluent densities or greater, amounts of fluorescence. Use of this technique in a canine
monolayer formation appears to be complete in under three model of microvessel endothelial cell sodding revealed that
weeks. Subconfluent densities require more extended peri- after 5 weeks the monolayer of endothelial cells on the flow
ods of time. surface exhibited extensive fluorescence, establishing their
identity as cells present in the original cell inoculum used
Mechanisms Underlying the Formation for sodding. Thus, following sodding, the microvessel en-
of a Neointima in Tissue Engineered Vascular dothelial cells need simply adhere to the polymer surface
Grafts Using Microvascular Endothelial Cell and to each other to form a confluent monolayer without
Transplantation the need to undergo extensive cell proliferation.
A number of preclinical animal studies have estab-
lished that, following the transplantation of microvessel en- Preclinical Animal Trials
dothelial cells onto synthetic grafts, the accelerated forma- of Endothelial Cell Transplantation
tion of an endothelial cell monolayer on the lumenal flow With the development of methods for the isolation of
surface is observed. The earliest time point evaluated is 4 endothelial cells from numerous tissue sources, including
days in a rat aortic graft model of microvessel endothelial macro- and microvascular sources, numerous animal stud-
cell transplantation.34 At this time a confluent layer of en- ies have been performed to evaluate the effects of endothe-
dothelial cells is observed. Studies in dogs have established lial cell transplantation on small diameter vascular graft pa-
that the endothelial cell lining observed in microvessel en- tency. These studies use small diameter grafts (< 6 mm),
dothelial cell sodded grafts remains stable for periods of up most often in the canine species. The predominance of ca-
to one year, the longest published animal implant data.33 nine models for these preclinical evaluations is based on the
Figure 6.3 illustrates the lining on a microvascular endothe- general acceptance of this model as being similar to humans
lial cell sodded ePTFE graft explanted after one year. This with respect to graft healing. While the canine species is com-
morphologic data establishes the ability to accelerate the monly used, techniques for graft placement (e.g., end to end,
Chi-Square
carotid, aortofemoral), use and duration of antiplatelet/an-
2.64
2.07
0.25
0.96
8.98
24.7
2.64
0.7
6.4
*
ticoagulant drugs (e.g., asprinin, persantine) and the type
and use of control grafts (e.g., paired vs. nonpaired grafts)
varies significantly among investigators. The rationale for
95% Lower
Confidence
the choice of each variable is often based on the goals of
-0.20,0.65
-0.09,0.30
0.02,0.62
0.03,0.37
0.06,0.46
0.44,1.00
0.50,0.86
0.01,0.62
0.27,0.89
and Upper
Intervals
individual studies. Quite often decisions are based on the
need to evaluate healing characteristics of grafts as compared
to evaluation of patency. The use of paired grafts (e.g., ca-
rotid or femoral interpositional grafts) is most common,
since cell transplanted grafts can be compared to control
Increase in
Fractional
for Seeded
grafts in the same animal. Variability in animal to animal
Patency
Grafts
0.36
0.17
0.73
0.68
0.11
0.22
0.32
0.58
0.2
thrombotic state is partially accounted for in paired graft
models.
Due to the variability in animal models previously
used by investigators, comparison of data is complicated.
Table 6.1 illustrates a compilation of the results of published
Patent Clotted
Control Grafts
13
16
10
20
11
11
animal trials using 4 mm inner diameter polymer grafts in
8
4
preclinical studies of endothelial cell transplantation. These
studies were chosen for the ability to evaluate patency using
25
26
11
6
1
8
6
1
nonpaired statistical analysis within each study. The signifi-
cance of endothelial cell transplantation can be evaluated
first by assessing the fractional increase in patency between Table 6.1. Endothelial cell-seeded graft versus control graft canine studies using paired 4 mm diameter grafts
Endothelial Cell-Seeded
cell transplanted and control grafts. In this column a posi-
Clotted
tive fractional increase in patency indicates a benefit of cell
7
9
2
2
3
1
1
7
4
transplantation, a zero increase indicates no benefit and a
Grafts
negative fractional increase indicates cell transplantation has
Patent
a negative influence on graft patency. The 95% confidence
12
32
11
27
30
14
9
7
8
limits have been provided for each study to indicate vari-
ability. The major conclusion from this table is the consis-
tently observed positive benefit of endothelial cell transplan-
tation with respect to graft patency. Although not all studies
Antiplatelet
None
None
None
None
6 wk
4 wk
4 wk
2 wk
2 wk
12 wk
16 wk
28 wk
10 wk
4 wk
4 wk
4 wk
Dacron
Dacron
Dacron
Dacron
ePTFE
ePTFE
ePTFE
ePTFE
Graft
Carotid
Carotid
Carotid
Carotid
Williams33
Schmidt28
Stanley31
Belden26
Shindo30
ripheral arterial interpositional grafts. The development of cellular vehicle for gene therapy.41 Endothelial cells reside at
a synthetic peripheral arterial graft is clearly of clinical im- the interface between blood and tissue and therefore main-
portance; however, new designs in synthetic grafts for use in tain a strategic position for the production and release of
other anatomical positions are also of great interest. In the therapeutic materials. Molecular biologists have begun to
USA, prosthetic grafts are presently the most common vas- take advantage of the extensive methodology established for
cular access created for hemodialysis.36 These grafts exhibit endothelial cell transplantation. Reports of genetic modifi-
a failure rate requiring intervention to restore patency at a cation of endothelial cells have exploded recently as the
mean of nine months after patency.37 The estimated annual attractiveness of this cell is realized. The use of genetically
cost to the USA health care system to maintain vascular ac- modified microvascular endothelial cells has moved beyond
cess for hemodialysis is believed to be in excess of $900 mil- prosthetic devices, and studies are ongoing to establish the
lion.37 The reason for the failure of these grafts is common use of these cells in almost every anatomic site under
to all synthetic grafts and includes the development of pre- consideration for gene therapies. Again, the availability of
dominantly distal anastomotic neointimal thickening and methods to easily isolate and transplant endothelial cells,
thrombosis due to the inherent thrombogenicity of poly- especially subcutaneous fat-derived microvessel endothelial
mer materials. Recent studies have begun to evaluate the use cells, provides an exciting new use for endothelial
of microvascular endothelial cell transplantation to improve cell transplantation.
AV graft function.38 These initial studies have demonstrated
that the healing of microvessel endothelial cell sodded grafts References
is quite different, depending on the site of anatomic place- 1. Quist WC, Haudenshield CC, Logerfo FW. Qualitative
ment. While arterial interpositional grafts sodded with en- microscopy of implanted vein grafts: Effects of graft in-
dothelial cells exhibit a relatively thin neointimal lining, sod- tegrity on morphological fate. J Thorac Cardiovasc Surg
ded grafts placed in the AV position in dogs have been re- 1992; 103:671-677.
2. Richardson JV, Wright CB, Hiratzka LF. The role of en-
ported to exhibit a dramatic thickening in the neointima
dothelium in the patency of small venous substitutes. J
throughout the length of the graft. These results suggest that Surg Res 1980; 28:556-562.
the high flow characteristics of AV fistulas create an envi- 3. Cambria RP, Megerman J, Abbot WM. Endothelial pres-
ronment which may stimulate neointimal thickening. ervation in reversed and in situ autogenous vein grafts.
Ann Surg 1985; 202(1):50-55.
Tissue Engineered Coronary Artery Bypass Grafts 4. Angelini GD, Breckenridge IA, Psaila JV, Williams HM,
While synthetic grafts have been used extensively for Henderson AH, Newby AC. Preparation of human saphe-
revascularization in the peripheral arterial position, the use nous vein for coronary artery bypass grafting impairs its
of synthetic, nonautologous vessels for coronary artery by- capacity to produce prostacyclin. Cardiovasc Res 1987;
pass grafting has routinely led to failure. The need for an 21:28-33.
antithrombogenic endothelial cell lining on synthetic CABG 5. Herring M, Gardner A, Glover J. A single staged tech-
nique for seeding vascular grafts with autogenous endot-
is clearly indicated. The development of tissue engineered
helium. Surg 1978; 84:498-504.
CABG using microvessel endothelial cell transplantation has 6. Schmidt SP, Hunter TJ, Sharp WV, Malindzak GS,
used two designs of synthetic grafts. Phillips and coworkers Evancho MM. Endothelial cell seeded four millimeter
report the use of the Possis Perma-Flow CABG as a syn- Dacron vascular grafts. J Vasc Surg 1984; 1:434-441.
thetic conduit for coronary bypass and have evaluated the 7. Stanley JC, Burkel WE, Ford JW. Enanced patency of small
use of microvessel endothelial cell sodding to create an en- diameter, externally supported Dacron iliofemoral grafts
dothelial cell lining on these grafts.39 Arzoumann and co- seeded with endothelial cells. Surg 1982; 92:994.
workers have developed an alternative synthetic CABG us- 8. Graham LM, Vinter DW, Ford JW. Immediate seeding of
ing microvessel endothelial cell sodding.40 This unique enzymatically derived endothelium in Dacron vascular
model involves the initial sodding of ePTFE grafts with place- grafts. Early studies with autologous canine cells. Arch
Surg 1980; 115:1289-1294.
ment and maturation in an AV fistula position first, followed
9. Kesler KA, Herring MB, Arnold MP, Glover JL, Park HM,
by conversion to the coronary position. The maturation in Helmus MN, Bendick PJ. Enhanced strength of endothe-
the AV position was reported to be necessary for the cre- lial attachment on polyester elastomer and
ation of a maximally antithrombogenic lining of endothe- polytetrafluoroethylene graft surfaces with fibronectin sub-
lial cells on the lumenal flow surface prior to conversion to strate. J Vasc Surg 1986; 3:58-64.
the CABG position. These reports suggest that use of tissue 10. Kempczinski RF, Rosenman JE, Pearce WH, Rodersheimer
engineered CABG in humans is on the horizon. LR, Berlatzky Y, Ramalanjaona GR. Endothelial cell seed-
ing of new PTFE vascular prostheses. J Vasc Surg 1985;
Future of Microvessel Endothelial Cell 2:424-429.
Transplantation 11. Lewis WH. Endothelium in tissue culture. Am J Anat
While the original goal of endothelial cell transplan- 1922; 30:39-59.
12. Thornton SC, Mueller SN, Levine EM. Human endothe-
tation studies has focused on small diameter vascular grafts,
lial cells: Cloning and long term serial cultivation employ-
the availability of operating room compatible methods for ing heparin. Science 1983; 222:623-624.
endothelial cell transplantation has expanded the possible 13. Jarrell BE, Levine EM, Shapiro SS, Williams SK, Carabasi
application of these cells. The development of gene therapy RA, Mueller SN, Thornton SC. Human adult endothelial
methodologies has begun to target the endothelial cell as a cell growth in culture. J Vasc Surg 1984; 1:757-764.
100 Tissue Engineering of Prosthetic Vascular Grafts
14. Zilla P, Fasol R, Preiss P, Kadletz M, Deutsch M, Schima 27. Douville EC, Kempczinski RF, Birinyi LK, Ramalanjaona
H, Tsangaris S, Groscurth P. Use of fibrin glue as a sub- GR. Impact of endothelial cell seeding on long term pa-
strate for in vitro endothelialization of ePTFE vascular tency and subendothelial proliferation in a small-caliber
grafts. Surgery 1989; 105(4):515-522. highly porous polytetrafluoroethylene graft. J Vasc Surg
15. Zilla P, Preiss P, Groscurth P, Rosemeier F, Deutsch M, 1987; 5:544.
Odell J, Heidinger C, Fasol R, von Oppel U. In vitro-lined 28. Schmidt SP, Hunter TJ, Hirko M. Small diameter vascu-
endothelium: Initial integrity and ultrastructural events. lar prostheses: Two designs of PTFE and endothelial cell
Surgery 1994; 116(3):524-534. seeded and nonseeded Dacron. J Vasc Surg 1985;
16. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T, 2:292-297.
Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I. 29. Shepard AD, Eldrup-Jorgensen J, Keough EM. Endothe-
Clinical in vitro endothelialization of femoropopliteal by- lial cell seeding of small caliber synthetic grafts in the
pass grafts: An actuarial follow-up over three years. J Vas- baboon. Surg 1986; 99:318.
cular Surgey 1994; 19(3):540-548. 30. Shindo S, Takagi A, Whittemore AD. Improved patency
17. Jarrell BE, Williams SK, Stokes G, Hubbard FA, Carabasi of collagen impregnated grafts after in vitro autogenous
RA, Koolpe E, Greener D, Pratt K, Moritz MJ, Radomski endothelial cell seeding. J Vasc Surg 1987; 6:325.
J, Speicher L. Use of freshly isolated capillary endothelial 31. Stanley JC, Burkel WE, Linbald B. Endothelial cell seed-
cells for the immediate establishment of a monolayer on ing of synthetic vascular prostheses. Acta Chir Scand Suppl
a vascular graft at surgery. Surg 1986; 100(2):392-399. 1985; 529:17-27.
18. Visser MJP, van Bockel JJ, van Muijen GNP, van 32. Tannenbaum G, Ahlborn T, Benvenisty A, Reemstma K,
Hinsbergh VWM. Cells derived from omental fat tissue Nowygrod R. High density seeding of cultured endothe-
and used for seeding vascular prostheses are not endot- lial cells leads to rapid coverage of PTFE grafts. Curr Surg
helial in origin: A study on the origin of epitheloid cells 1983; 222:623.
derived from human omentum. J Vasc Surg 1991; 33. Williams SK, Rose DG, Jarrell BE. Microvascular endot-
13:373-381. helial cell sodding of ePTFE vascular grafts: Improved
19. Williams SK, Wang TF, Castrillo R, Jarrell BE. Liposuction patency and stability of the cellular lining. J Biomed Mater
derived human fat used for vascular graft sodding con- Res 1994; 28:203-212.
tains endothelial cells and not mesothelial cells as the 34. Ahlswede KM, Williams SK. Microvascular endothelial cell
major cell type. J Vasc Surg 1994; 19:916-923. sodding of 1-mm expanded polytetrafluoroethylene vas-
20. Williams SK, Jarrell BE, Rose DG, Pontell J, Kapelan BA, cular grafts. Arteriosclerosis and Thrombosis 1994;
Park PK, Carter TL. Human microvessel endothelial cell 14:25-31.
isolation and vascular graft sodding in the operating room. 35. Williams SK, Kleinert LB, Rose D, McKenney S. Origin
Ann Vasc Surg 1989; 3(2):146-152. of endothelial cells that line expanded
21. Lenzi R, Alpinin G, Liu MH, Rand JH, Tavoloni N. Von polytetrafluoroethylene vascular grafts sodded with cells
willebrand factor antigen is not an accurate marker of rat from microvascularized fat. J Vasc Surg 1994; 19:594-604.
and guinea pig liver endothelial cells. Liver 1990; 36. Palder SB, Kirkman RL, Whittemore AD. Vascular access
10:372-379. for hemodialysis. Ann Surg 1986; 202:235-239.
22. Wagner RC, Matthews MA. The isolation and culture of 37. Windus DW. Permanent vascular access: A nephrologist’s
capillary endothelium from epididymal fat. Microvasc Res view. Am J Kid Dis 1993; 21(5):457-471.
1975; 10:286-297. 38. Williams SK, Jarrell BE, Kleinert LB. Endothelial cell trans-
23. Williams SK, McKenney S, Jarrell BE. Collagenase lot se- plantation onto polymeric arteriovenous grafts evaluated
lection and purification for adipose tissue digestion. Cell using a canine model. J Invest Surg 1994; 7:503-517.
Transplantation. 1995; 4:281-289. 39. Phillips M, Yamaguchi H, Miller V, Williams SK, Morris
24. Williams SK, Carter T, Park PK, Rose DG, Schneider T, JJ, Shaff HJ. Endothelial cell sodding of the Permaflow
Jarrell BE. Formation of a multilayer cellular lining on a prosthetic coronary artery bypass conduit. Annals of Thor
polyurethane vascular graft following endothelial cell sod- Surg 1998; in press.
ding. J Biomed Mat Res 1992; 26:103-117. 40. Arzouman D, Kleinert LB, Patula VB, Phillips M, Will-
25. Allen BT, Long JA, Clark RE, Sicard GA, Hopkins KY, iams SK, Copeland JG. Endothelial cell sodding of ePTFE
Welch MJ. Influence of endothelial cell seeding on plate- coronary artery bypass grafts. Abstract. American Heart
let deposition and patency in small-diameter Dacron ar- Association 70th Scientific Session.
terial grafts. J Vasc Surg 1984; 1:224-232. 41. Stopeck AT, Vahedian M, Williams SK. Transfer and ex-
26. Belden TA, Schmidt SP, Falkow LJ, Sharp WV. Endothe- pression of the interferon gamma gene in human endot-
lial cell seeding of small-diameter vascular grafts. Trans helial cells inhibits vascular smooth muscle cell growth
Am Soc Artif Intern Organs 1982; 28:173. in vitro. Cell Transplant 1997; 6(1):1-8.
CHAPTER 7
Morphological Aspects of Microvascular Cell Isolates
Manuela Vici
Introduction
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
102 Tissue Engineering of Prosthetic Vascular Grafts
For all these reasons we believe morphological infor- method for the isolation of microvascular endothelium from
mation to be very important for studies involved in the tis- first trimester decidua.17
sue engineering of vascular grafts. Moreover, when identi-
fication or localization of certain cell types is also requested, Isolation of Microvascular Endothelial Cells
they can successfully be supplied by immunocytochemical Jarrel and Williams first documented how microvas-
investigations. cular endothelial cells can easily be recovered from enzy-
matically disrupted fat tissue.3
Microvascular Cells In our lab we developed a modification of the Jarrel
Microvessels are considered the alternative to vein as method starting from human subcutaneous fat tissue ex-
a source of endothelial cells for cell transplantation. There cised from the abdominal wall of patients undergoing sur-
are many tissues in humans which are abundant in capillar- gery for abdominal aneurysms.18 The fat tissue is carefully
ies and promptly available either surgically or by noninvasive minced and mixed with a 0.2% collagenase solution in an
methods. During years of experiments the protocols of the Erlenmayer flask and incubated under constant agitation for
isolation of microvascular endothelial cells have become 30 minutes at 37°C. After digestion, the floating adipocytes
increasingly fast and efficient—normally a large number of are discharged and the suspension centrifuged. The pellet is
viable endothelial cells is easily obtainable per gram of tissue. first washed in PBS-BSA and then filtered through a 120 ∝m
nylon filter; the suspension is gently layered onto 45% ster-
Sources of Microvascular Endothelial Cells ile Percoll and endothelial cells are recovered from the milky
Jarrel and Williams first started to harvest microvas- layer formed after centrifugation. Normally a variable num-
cular endothelial cells enzymatically for cell transplantation.3 ber of viable cells ranging from 2.5-8 x 105 is obtained per g
Initially they reported on methods established for the isola- of tissue. Recently we proposed an alternative method of
tion and culture of human adult microvessel endothelial cells microvascular endothelial cell isolation based on the use of
from different sources of human fat such as omental, peri- magnetic polystyrene beads (Dynabeads M-450) coated with
renal and subcutaneous fat. Then they focused on human anti-CD34 monoclonal antibody.19 After the mincing and
subcutaneous fat tissue as the most suitable source of en- collagenase digestion of the subcutaneous fat tissue, the cells
dothelial cells, as it is composed mainly of adipocytes richly filtered and suspended in PBS-BSA as previously described
perfused with microvascular endothelium and pericytes. are incubated with anti-CD34 coated Dynabeads
Human subcutaneous fat is easily obtainable either (Dynabeads M-450 CD34, Dynal) at a ratio of 10 beads to 1
surgically by patients undergoing abdominal surgery, (this endothelial cell for 30 minutes at 4°C under gentle mixing.
is what has been mainly experienced in our lab), or by Rosetted cells are positively collected by means of three wash
liposuction of fat deposits on the abdominal wall.4 cycles performed with a Dynal Magnetic particle concen-
Omentum and omentum-associated fat has also been trator (MPC). Rosetted cells are then resuspended in M199
utilized as a rich source of microvascular endothelial cells, plus 20% FCS containing 10 ∝l DetachaBead (Dynal) per
but recently it has been proved by Williams et al that be- 1 x 107 Dynabeads used for rosetting, and incubated for 1 h
cause of its anatomical location, the primary isolate from at room temperature with constant agitation. After incuba-
omentum often comprises not pure endothelium but a mix- tion cells are negatively recovered using the Dynal MPC.
ture of cells including endothelium, smooth muscle cells and Usually approximately 0.8 x 105 cells per g of fat tissue are
mesothelium.5 obtained.
Microvascular endothelial cells can also be harvested Recently, Chen et al proposed a simple new method
from other sources. Hewett et al developed a method for the for the isolation of microvascular endothelial cells avoiding
isolation and long term culture of human microvessel en- both chemical and mechanical injuries.20 They indicated that
dothelial cells from mammary adipose tissue obtained at when small pieces of lung or muscles of the chest wall of
breast reduction.6 Robinson et al reported on the seeding of rats are placed into a flask, erythrocytes and leukocytes leave
immortalized human dermal microvascular endothelial the tissues first, followed by vascular endothelial cells. Fi-
cells.7 Procedures for the isolation and culture of microvas- broblasts and other mixed cells grow after 72 hours of cul-
cular endothelium from neonatal foreskins have also been ture. When the tissues are discarded within 60 hours the flask
previously described by Sherer et al, Marks et al, and Karasak contains only microvascular endothelial cells and blood cells.
et al.8-10 Hewett et al isolated and cultured microvessel en- The latter can be cleared out after the cells are subcultured.
dothelial cells from lung tissue obtained from lung trans-
plant recipients.11 Haraldsen et al established a method for Microvascular Cell Isolates
the isolation and culture of intestinal microvascular endot- The purity of a microvascular isolate has always been
helial cells.12 Kacemi et al devised a method of isolation and a major concern when microvessel-derived endothelial cells
culture of endothelial cells from villous microvessels from are chosen for cell transplantation. The attempt to charac-
human placenta.13 Masek et al and Schweitzer et al described terize the cell population of a microvascular primary isolate
two simple methods for the isolation and culture of endo- from human fat tissue has lead to controversies. This is a
thelial cells from human bone marrow.14,15 McDouall et al consequence of the fact that the type of cells involved in a
isolated and cultured microvascular endothelial cells from microvascular primary isolate correlates directly with the
the human heart.16 Finally, Grimwood et al developed a novel source of microvessels chosen.
Morphological Aspects of Microvascular Cell Isolates 103
Omentum and omentum fat are rich in mesothelial rience) is that the possibility of collecting information such
cells; consequently the primary isolate comprises a hetero- as the appearance of subcellular components, or the pres-
geneous cell population including endothelial cells, pericytes, ence of intercellular connections, from a primary isolate, can
fibroblasts and mesothelial cells. Human subcutaneous fat give a realistic description of the biological status of the cells
is composed predominantly of adipocytes, pericytes and analyzed and can add an indispensable contribution when
microvascular endothelial cells (Fig. 7.1a,b), and thus the an accurate characterization of the cell types involved
primary isolate is expected to be more homogeneous. Re- is necessary.
cent studies are focusing on the fact that mesothelial cells For example, as already mentioned, the morphologi-
can also be utilized for the seeding of vascular prostheses, cal changes in rough endoplasmic reticulum, Golgi appara-
behaving very much like endothelial cells derived from large tus and mitochondria are correlated to the level of meta-
vessels when seeded.21 bolic activity of the cell; Weibel-palade bodies are undoubt-
Thus, depending on the aim of the study, the charac- edly considered a morphological marker for endothelial cells,
terization of the cell population of a microvascular primary while “fibronexus” is the morphological marker for
isolate can be very important. To do this we focused on myofibroblasts. Moreover, the fact that cells start to develop
morphological studies. Our conviction (derived from expe- intercellular junctions when placed in culture or when
seeded, often gives positive information about growth and
stability. Furthermore, the localization of molecules specifi-
cally present in certain cells by immunohistochemistry is
definitely helpful for the characterization of the different cell
types involved in a primary isolate and can also contribute
to the definition of molecular pathways signaling, when
present, metabolic alterations.
as sites of membrane fusion with adjacent electron-dense acteristic symmetrical clefts. The cytoplasm is rich in in-
“fuzzy” cytoplasm. Specific rod-shaped microtubulated bod- termediate filaments and microfilaments and very few or-
ies (the so-called Weibel-palade bodies) are occasionally seen ganelles are found. Microfilaments are usually disposed par-
but are evident in less than 5% of the cells examined, par- allel to the long axis of the cell, among which are interspersed
ticularly in the clustered elements (Fig. 7.2). Weibel-palade numerous focal densities. These are electron dense struc-
bodies are specifically found in the endothelium (Fig. 7.3B); tures scattered in the cytoplasm which are part, together with
their typical rod shape is contoured by a membrane con- the contractile filaments, of the contractile apparatus. Pre-
taining numerous parallel cylindrical tubules embedded in cisely three different kinds of filaments can be described:
an electron-dense matrix. Immunological studies have myosin filaments (18 nm in diameter); actin filaments
shown that Weibel-palade bodies are sites of storage of the (6-8 nm in diameter) and vimentin and desmin intermedi-
von Willebrand protein. ate filaments. In the subplasmalemmal site there are many
MECs isolated with the help of CD34-coated micropinocytotic vescicles and plasmalemmal introflexions
Dynabeads appear, prior to DetachaBeads application, with called “caveolae” which increase the plasmalemmal surface
1-6 beads bound to the plasma membrane (Fig. 7.4). After by about 25%. SMCs can form gap junctions and under spe-
bead detachment, MECs are present mainly as single cells, cific conditions can change into a “synthetic” phenotype and
morphologically homogeneous and well preserved. Weibel- become myofibroblasts.
palade bodies are occasionally observed in the cytoplasm Myofibroblasts are characterized by nuclear polymor-
together with other typical endothelial features previously phism, cytoplasmic extensions and abundant rough endo-
mentioned (intermediate filaments, micropinocytotic plasmic reticulum, Golgi apparatus and mitochondria. They
vescicles and microcystic lacunae).19 are connected to the extracellular matrix by the so-called
When other sources of microvascular cells are chosen, “fibronexus”, which are transmembrane complexes of intra-
the primary isolate can be more heterogeneous and other cellular microfilaments in apparent continuity with extra-
cell types can be described. Mesothelial cells are character- cellular fibronectin fibers. These cell to stroma attachment
istically identified by their numerous microvilli at the apical sites are more developed and numerous than in smooth
surface; they are heterogeneous in nature and measure up muscle cells, and three types of fibronexus can be described:
to 3 ∝m in length and 0.1 ∝m in diameter. In the cytoplasm plaquelike, tracklike and tandem associations. Within the
there are some pinocytotic vesicles, very few Golgi cytoplasm there are numerous bundles of microfilaments
membranes, scanty granular endoplasmic reticulum and (stress fibers) arranged parallel to the long axis of the cell,
many mitochondria. Junctions of all types can be found in with interspersed numerous dense bodies.
situ. Intermediate filaments are present, sometimes Fibroblasts have no basal lamina and display a slen-
prominent and often arranged in a perinuclear, circumfer- der fusiform and smoothly contoured nucleus; in the cyto-
ential distribution.22 plasm microfilaments are sometimes arranged in bundles
Smooth muscle cells (SMCs) are delimited by a basal beneath the plasma membrane and there is a well developed
lamina and present elongated and lobated nuclei with char- Golgi area and abundant dilated cisternae of rough endo-
Fig. 7.5. Percoll isolated microvascular endothelial cells. (A), left. SEM of a multicell aggregate. (SEM; x4,200). With permission
from: Curti T, Pasquinelli G, Preda P et al. An ultrastructural and immunocytochemical analysis of human endothelial cell adhe-
sion on coated vascular grafts. Ann Vasc Surg, 1989; 3:351-363. (B), right. Immunoelectron microscopic demonstration of FVIII
RA. SEM reveals strong positivity at cell surface. (SEM; x8.500). With permission from: Curti T, Pasquinelli G, Preda P et al. An
ultrastructural and immunocytochemical analysis of human endothelial cell adhesion on coated vascular grafts. Ann Vasc Surg,
1989; 3:351-363.
that EN4 and CD31 monoclonal antibody recognize the ing of cytoplasmic and surface membrane antigens on cell
same 130 kDa antigen.29 smears at optical level. Cells seeded onto fibronectin-coated
Since a primary microvascular isolate can be com- coverslips were fixed in acetone-metanol (7:3 at -20°C for
posed of a mixture of cell types, it might also be useful to 10 minutes) and incubated in the primary antibody, then in
characterize most of them. Vascular smooth muscle cells anti-mouse immunoglobulin and finally in APAAP com-
specifically express vimentin and smooth muscle actin, while plexes. The reaction was revealed by adding the substrate
a low percentage is desmin positive. Cytokeratins such as containing naphtol AS phosphate and new fuchsin. For
CK8 and CK18 can be found in some SMCs. Pericytes also polyclonal antibodies, samples were treated with a bridge
express smooth muscle-specific actin. Nonproliferating me- antibody.
sothelial cells express both vimentin and a variety of Protein a-gold technique was performed on MECs
cytokeratins (CK7, CK8, CK18, CK19).22 Adipocytes express seeded on fibronectin-coated coverslips by incubating cells,
S-100 protein, while monocytes and macrophages can be after the primary antibody, with 18 nm protein a-gold par-
detected using antibodies against specific antigens such as ticles.31,32 The reaction was then silver enhanced. For mono-
150.95 protein and MAC 387. clonal antibodies a secondary antibody was employed.
We studied the immunophenotype of microvascular For immunoelectron microscopy, suspensions of
isolates from human subcutaneous fat tissue at different steps MECs were fixed in 3% paraformaldehyde, then incubated
in the isolation procedure with the help of different immu- with the primary antibody and afterwards with an appro-
nocytochemical techniques using monoclonal and priate dilution of 18 nm protein a-gold complexes. Finally,
polyclonal antibodies. Immunofluorescence was performed MECs were postfixed in 1.7% glutaraldehyde and embed-
by first seeding the isolate onto fibronectin-coated cover- ded in araldite. Immunolabeling specificity was always
slips for 1 hour at 37°C, and then fixing the cells with cold checked by omitting the incubation with the specific pri-
acetone and incubating them with the primary antisera. A mary antibody or replacing the primary antibody with an
second incubation with a fluorescein-conjugated anti-rab- unrelated antibody.
bit or anti-mouse Ig followed. Coverslips were then mounted Our investigation was performed employing the fol-
and observed under a photomicroscope equipped with an lowing antibodies. As endothelial cell markers we used: a
epifluorescent condenser. polyclonal antibody against Factor viii RA; a polyclonal an-
Immunoenzymatic labeling was also performed on tibody specific for the Ulex europaeus type i lectin previ-
MECs cells using the alkaline phosphatase monoclonal ously bound to the endothelial membrane; a monoclonal
antialkaline phosphatase (APAAP) procedure.30 The APAAP antibody against CD31 antigen and QB-End 10, a mono-
technique is particularly suitable for clear and specific stain- clonal antibody specific for the CD34 antigen. Antibodies
108 Tissue Engineering of Prosthetic Vascular Grafts
directed against cytoskeletal proteins (actin, smooth muscle With immunohistochemistry we concluded that en-
actin, desmin and vimentin), monocyte-macrophage spe- zymatically harvested MECs did not contain adipocytes and
cific antigens (MAC 387, anti-protein 150.95) and S-100 pericytes, since immunostaining showed protein S-100 and
protein, which is present in adipocytes, were also used. desmin to be virtually nonexistent. Furthermore, the pres-
Following collagenase digestion approximately 90% ence of pericytes was ruled out by a negative response for
of MECs stained for factor VIII, as well as for CD31, CD34, smooth muscle-specific actin. As demonstrated by
and Ulex. After the Percoll step, cell isolate weakly expressed immunostaining for monocyte-macrophage specific anti-
factor VIII (5%) (Fig. 7.7A). Furthermore, 18% of the iso- gen (150.95 protein and MAC387), the presence of mono-
lated cells stained for CD31 (Fig. 7.7B) and 5% were labeled nuclear blood cells was negligible (1-2%).
with Ulex europaeus. Remarkably these endothelial-specific On the other hand, our results on MECs harvested
antigens were found mostly in cell clumps. Interestingly, from human abdominal subcutaneous fat tissue (which as
CD34 was detected in 90% of the harvested cells compris- previously demonstrated show ultrastructural features typi-
ing the primary isolate irrespective of the separation step cal of endothelium), using common endothelial cell mark-
(Figs. 7.7C and 7.3C). Only 1-2% of cells expressed mono- ers (FVIII RA, Ulex europaeus, CD31) have apparently led
cyte-macrophage specific antigens, 150.95 protein and MAC to some discrepancies that we related to factor VIII release
387. Immunostaining for desmin and S-100 protein was al- and surface antigen rearrangements occurring in vitro dur-
most negative. Smooth muscle actin was expressed in ing procedures of cell isolation, mainly at the Percoll step.
few cells. Interestingly, immunoelectron microscopic demonstration
Morphological Aspects of Microvascular Cell Isolates 109
of factor VIII RA by SEM reveals a moderate to strong posi- to mesodermally derived tissue. The fact that after collage-
tivity at the cell surface (Fig. 7.5B). A possible adverse effect nase isolation the cells expressing vWF and EN4 were fewer
of Percoll on cell function has also been observed by DOT than those observed in intact fat suggests that a portion of
immunoassay. Precisely, DOT immunoassay documented an cells lose vWF and EN4 during the isolation procedure.
intense factor VIII delivery in the washing solutions follow- Stansby et al attempted to characterize microvascular
ing Percoll density gradient, while CD34 was always nega- endothelial cells from human omental tissue using both
tive. Notably, in MECs obtained with the Dynabeads proce- endothelial-specific markers, (namely anti-factor VIII, EN4,
dure, where the Percoll step was omitted, factor VIII RA was PAL-E and QB-End/40) and monoclonals against antigens
expressed by the vast majority of cells (Fig. 7.7D) and DOT specific of cells of connective tissue origin (Thy-1 and
immunoassay of the washing buffers collected after CDw44).34 The authors found that the microvascular cells
Dynabeads application was virtually negative excluding any they harvested from omentum are extremely heterogeneous
loss of factor VIII during the isolation steps. Only CD34 was with respect to their antigenic expression and are probably
constantly expressed by MECs during and after the isola- initially a mixture of several different cell types: only occa-
tion by Percoll density gradient. We concluded that QB- sional cells are positive for factor VIII RA, Thy-1 and CDw44
End/10 monoclonal antibody, which recognizes the highly glycoprotein. No positivity for EN4, PAL-E and QB-End/40
resistant CD34 molecule on the surface of endothelial cells, was reported. After culture it seems likely that one cell type
is particularly well suited for characterizing enzymatically predominates, as the cultures are then homogeneous both
harvested MECs.33 morphologically and immunophenotypically. The majority
Also, Williams et al observed a cellular response to the of cells are then positive for factor VIII RA, Thy-1 and Cdw44
isolation procedure. They studied the cell population of glycoprotein. A small proportion (< 5%) is also positive for
liposuction-derived human fat before and after collagenase the endothelial markers EN4, PAL-E and QB-End/40. The
dispersion with the help of immunocytochemistry and scan- authors suggest that Thy-1 and CDw44 glycoprotein posi-
ning electron microscopy.5 Samples of liposuction-derived tive cells, which were shown to take up acetylated ldl and
human fat have been processed for the presence of FVIII produce prostacyclin, might be pericytes.
RA-von Willebrand factor, #-smooth muscle cell actin, Also, Meerbaum et al performed an immunohis-
cytokeratin (CK18) and EN4. The same study was performed tochemical investigation on the primary isolate enzymati-
on the cells recovered after collagenase dispersion of the cally harvested from subcutaneous fat tissue obtained by
liposuction fat. 86.1% of the cells in intact liposuction-de- liposuction and used for limited clinical trials.35 They found
rived fat expressed vWF, 5.7% were positive for #-smooth 55.8% of cells positive for factor VIII while 42.5% were re-
muscle cell actin, 1% expressed the mesothelial cell-related active to muscle-specific actin. 9.4% of cells reacted with
antigen cytokeratine peptide 18 and 89.6% of the cells in monocyte-macrophage specific markers. They speculated on
intact fat resulted positive for EN4. After collagenase diges- the possibility that contaminating cells might have been
tion of the fat and centrifugal separation of adipocytes from pericytes. They also observed that Percoll separation nega-
vascular and stromal cells, the expression of vWF, #-smooth tively affects cell attachment and growth in culture.
muscle cell actin, and cytokeratin was 77.5%, 5.8%, and 2.1%
respectively. 74.6% of the isolated cells expressed EN4. Discussion
This study demonstrated that the major cell compo- Since our unit was founded in 1962, it has been de-
nent present in liposuction-derived fat, before and after col- voted to the elaboration of a subcellular approach to clini-
lagenase digestion, has been characterized as endothelium, cal manifestation. As soon as the Vascular Surgery unit of
while only a minor fraction is composed of cells common the hospital where we were placed decided to start looking
into the endothelial cell seeding of vascular grafts, it asked One example comes from our own experience. Our
for our participation, finding our experience immediately attempt to characterize microvascular endothelial cells
important to initiate this challenging new task. immunohistochemically yielded some discrepancies. After
Undoubtedly a successful morphological approach Percoll density gradient isolation, we found that typical
correlates well with experience, both for technical skill, which markers (factor VIII, Ulex europaeus, and CD31) were ex-
is always a guarantee for precision and efficiency, and for pressed by only a small percentage of cells. Immuno-
the ability to collect and interpret the results obtained. For histochemically we concluded that microvascular endothe-
example, each lab which focuses on ultrastructure has elabo- lial cells harvested from human subcutaneous fat tissue did
rated its own protocols, and it is always very important to not contain contaminating stromal cells such as adipocytes
learn, when observing samples, how to distinguish artifacts and pericytes, and the presence of mononuclear blood cells
from the “real” structure. was negligible. Moreover, harvested cells showed ultrastruc-
Although the optimization of these aspects could ap- tural features typical of endothelium. Subcellular criteria
pear time consuming, the study of the morphology of single included residual lumina, loosely textured intermediate fila-
cells or of a tissue can give important information about ments, micropinocytotic vesicles, and Weibel-palade bod-
their biology and function. Electron microscopy techniques ies. Interestingly, factor VIII, CD31 and Ulex europaeus al-
represent powerful tools which provide very accurate infor- most exclusively stained cell aggregates. Likewise, TEM dem-
mation about cell ultrastructure and, thanks to combined onstrated Weibel-palade bodies in clustered endothelial cells
immunocytochemistry, about cell biology (related to anti- only, whereas single cells, which also showed the subcellular
gen expression, synthesis of specific molecules, etc.). We features of endothelium, lacked these highly specific endo-
believe that, for all these reasons, morphology can make an thelial organelles.
important contribution to the development of the tissue We hypothesized an adverse effect of Percoll on
engineering of vascular grafts. endothelial cell function. Our hypothesis was supported by
As already reported, submicroscopic studies have been the finding of factor VIII in the washing solutions after
very important in defining the processes involved in the ad- Percoll separation, documented by DOT immunoassay and
hesion of cells to a surface. This information can success- by the detection of factor VIII by immunoSEM on the sur-
fully be applied to the study of vascular cell seeding onto face of some of the cells, which could represent a possible
prosthetic grafts. With the scanning electron microscope we step in the delivery of factor VIII in the washing solutions
can easily follow the different steps of adhesion of the en- following the Percoll treatment. Interestingly, when using
dothelial cells seeded on the vascular substratum that we alternative methods where the Percoll step is omitted the
are testing. First of all, we can perceive a qualitative response: majority of the harvested endothelial cells stain for factor
No interaction with the substratum, as revealed by a steady, VIII.19 Accordingly, the decreased expression of CD31 and
rounded cellular shape, is informative of an unsuitable sur- other membrane glycoproteins, or part of them, could be a
face—cells do not like that substratum. At an intermediate consequence of surface antigen rearrangements also induced
level the endothelial cell has already started to interact with by the Percoll step.
the vascular graft and the cell surface is modifying; endo- Here we have reported on other authors who also per-
thelial cells now appear as skirted cells. If the interaction formed correlated morphological and immunohistochemi-
proceeds, meaning that the vascular substratum is particu- cal studies in order to determine the cellular composition of
larly suitable for the cells, their surface modifies further, be- a microvascular primary isolate; in some cases these studies
coming completely flattened. Depending on the number of have also led to evaluation of the influences of the isolation
cells seeded, they can eventually reach each other and, by procedures chosen over the cell function.
forming intercellular junctions, start to initiate a monolayer. Being correlated to the potential of technological ap-
We can explain all these surface modifications by TEM as paratus, the “limits” of morphological studies are subject to
cytoskeletal rearrangements. constant modification. Computer science, together with in-
TEM has been very helpful for the characterization of novative technology, has permitted the development of very
microvascular primary isolates. We already know that the elaborate microscopes, adding a new perspective to the study
cell types involved in a primary microvascular isolate vary of morphology. Confocal microscopy is an advanced tech-
with the source chosen. Different sources placed in anatomi- nique used to produce multidimensional cellular and sub-
cally different sites, clearly comprise, besides microvessels, cellular structural images. It provides high resolution im-
other cell types. Whether a microvascular isolate contains ages and its optical sectioning ability allows images to be
pure endothelium or not can be of primary importance obtained from different depths within a thick tissue speci-
during clinical trials. TEM with the help of immunocy- men, avoiding processing and sectioning procedures. For this
tochemistry can give a very accurate description of the cell reason, confocal microscopy has made it possible to view
types involved in a microvascular isolate. Ultrastructurally biological tissues under more physiological conditions than
there are morphological markers that can, without doubt, was previously possible. Commonly, its biological applica-
allow different cell types to be recognized. This is supported tion addresses the localization of immunofluorescently la-
by immunocytochemistry which localizes molecules specifi- beled proteins in cell culture or within excised blocks of
cally synthetized by certain cells or groups of cells. More- tissue. Because of its noninvasive optical sectioning capa-
over, together they can also permit the identification of al- bility, it is also ideally suited to the study of living cells in
terations of the biological status. situ and of tissue in intact living animals.36
112 Tissue Engineering of Prosthetic Vascular Grafts
Confocal microscopy immunofluorescence staining ported by very sophisticated machines and, as a consequence,
allows a better resolution than that with traditional immu- their potential is continuously improving. As we have seen,
nofluorescence. It would therefore be possible to perform they have already been successfully employed in the tissue
very detailed in situ or in vivo studies of adhesion molecules engineering of prosthetic vascular grafts, and it is becoming
or other cell products in cultures or, in the near future, in clearer that their future development will be of great value
implanted grafts. The scanning resolution of the confocal in the evolution of this field.
microscope has also remarkably improved on that of the tra-
ditional SEM. More precise evaluations about surface infor- Acknowledgments
mation such as cell lining, for example, can be simultaneously I would like to thank Prof. Massimo Derenzini for
supported, thanks to the optical sectioning ability, by supporting me and giving me the free run of the archives of
“deeper” information regarding thickenings and their bio- the Institute, and Prof. Massimo D’Addato of the Vascular
logical compounds. Understandably, confocal microscopy Surgery Unit, Director of the Center for the Study of Vascu-
is considered to be one of the techniques that in the near lar Prostheses, and his staff for their collaboration. I am also
future will enormously improve the understanding of bio- extremely grateful to Dr. Paola Preda and Dr. Gianandrea
logical events. Pasquinelli for their invaluable assistance and to Mr. Walther
The constant development of technologies for the Mantovani for his indispensable technical skill. Finally,
quantitative analysis of images has made image analysis the thanks to Patrizia and Lorenzo for their patience.
most suitable tool for quantitative studies on cell seeding,
or whenever a precise definition of the number of cells (or References
part of stained cells) is necessary. Supported by microanaly- 1. Langer R, Vacanti JP. Tissue engineering. Science 1993;
sis, quantitative multielemental analysis in small sections of 260:920-926.
samples can be performed routinely. X-ray microanalysis is 2. Versura P, Maltarello MC, Fontana L et al. Ultrastruc-
tural investigation demonstrating reduced cell adhesion
being widely employed in evaluating the biocompatibility
on heparin-surface-modified intraocular lenses. Ophtalmic
and biostability of alternative biomaterials. Precisely, stud- Res 1991; 23:1-11.
ies have been performed in order to describe the influence 3. Williams SK, Jarrel BE, Rose DG. Isolation of human fat-
of biomaterials on organic matrix synthesis and the calcifi- derived microvessel endothelial cells for use in vascular
cation process by using X-ray microanalysis coupled with graft endothelialization. In: Zilla PP, Fasol RD, Deutsch
TEM37 or high resolution electron microscopy.38 The vas- M eds. Endothelialization of Vascular Grafts. Basel: Karger
cular graft field has already gained from this technique. It 1987:211-217.
has allowed the testing of biocompatibility of new polymers 4. Jarrel BE, Williams SK. Microvessel derived endothelial
to be employed in new synthetic grafts.39 It has also made it cell isolation, adherence, and monolayer formation for
possible to assess whether chemical degradation of ePTFE vascular grafts. J Vasc Surg 1991; 13:733-734 (special com-
occurs when such arterial substitutes are implanted munication).
5. Williams SK, Wang TF, Castrillo R et al. Liposuction-de-
in humans.40
rived fat used for vascular graft sodding contains endo-
Lately, techniques have been developed to understand thelial cells and not mesothelial cells as the major cell type.
the state of specific gene expression at cellular and/or J Vasc Surg 1994; 19:916-923.
subcellular levels. In situ hybridization is a powerful and well 6. Hewett PW, Murray JC, Price EA et al. Isolation and char-
established light microscopic technique used to localize acterization of microvessel endothelial cells from human
mRNA, and recent advances have been made with the help mammary adipose tissue. In Vitro Cell Dev Biol Anim
of transmission electron microscopy.41 It is already possible 1993; 29A: 325-331.
to detect, using properly chosen nucleic acid probes 7. Robinson KA, Candal FJ, Scott NA et al. Seeding of vas-
(radioactive or biotinylated), genes or products of genes in cular grafts with an immortalized human dermal mi-
the cell examined. crovascular endothelial cell line. Angiology 1995;
46:107-112.
Recently, genetic manipulation has been introduced
8. Sherer GK, Fitzharris TP, Faulk WP et al. Cultivation of
in order to ameliorate vascular graft implantation. For ex- microvascular endothelial cells from human preputial skin.
ample seeding of small diameter vascular grafts with endo- In Vitro 1980; 16:675-684.
thelial cells genetically engineered to secrete fibrinolytic or 9. Marks RM, Czerniecki M, Penny R. Human dermal mi-
antithrombotic proteins has been performed to improve crovascular endothelial cells: an improved method for tis-
graft patency rates.42 Another example concerns the attempt sue culture and a description of some singular properties
to improve the biomaterial-blood compatibility by induc- in culture. In Vitro Cell Dev Biol 1985; 21:627-635.
ing, with genetical manipulation, the inhibition of SMC pro- 10. Karasek MA. Microvascular cell culture. J Invest Dermatol
liferation.43 TEM-in situ hybridization, with the help of spe- 1989; 93:33S-38S.
cific probes, could theoretically permit the visualization of 11. Hewett PW, Murray JC. Human lung microvessel endot-
genetically engineered cells so that we can obtain informa- helial cells: Isolation, culture, and characterization.
Microvasc Res 1993; 46:89-102.
tion regarding the quantification of seeded genetically engi-
12. Haraldsen G, Rugveit J, Kvale D et al. Isolation and long
neered cells, their proliferation and metabolic activity, and term culture of human intestinal microvascular endothe-
more besides. lial cells. Gut 1995; 37:225-234.
All of these techniques briefly listed are usually pre-
sented in manuals as “innovative microscopy”. They are sup-
Morphological Aspects of Microvascular Cell Isolates 113
13. Kacemi A, Challier JC, Galtier M et al. Culture of endo- 29. Burgio VL, Zupo S, Roncella S et al. Characterization of
thelial cells from human placental microvessels. Cell Tis- EN4 monoclonal antibody: A reagent with CD31 speci-
sue Res 1996; 283:183-190. ficity. Clin Exp Immunol 1994; 96:170-176.
14. Masek LC, Sweetenham JW. Isolation and culture of en- 30. Cordell JL, Falini B, Erber WN et al. Immuno-enzymatic
dothelial cells from human bone marrow. Br J Hematol labeling of monoclonal antibodies using immune com-
1994; 88:855-865. plexes of alkaline phosphatase and monoclonal antialkaline
15. Schweitzer CM, van der Schoot CE, Drager AM et al. Iso- phosphatase (APAAP complexes). J Histochem Cytochem
lation and culture of human bone marrow endothelial 1984; 32:219-229.
cells. Exp Hematol 1995; 23:41-48. 31. Frens G. Controlled nucleation for the regulation of the
16. McDouall RM, Yacoub M, Rose ML. Isolation, culture, particle size in monodisperse gold solution. Nature Phys
and characterization of MHC class II-positive microvas- Sci 1973; 241:20-22.
cular endothelial cells from the human heart. Microvasc 32. Roth J, Bendayan M, Orci L. Ultrastructural localization
Res 1996; 51:137-152. of intracellular antigens by the use of protein a-gold com-
17. Grimwood J, Bicknell R, Rees MC. The isolation, charac- plexes. J Histochem Cytochem 1978; 26:1077-1081.
terization and culture of human decidual endothelium. 33. Vici M, Pasquinelli G, Preda P et al. Electron microscopic
Hum Reprod 1995; 10:2142-2148. and immunocytochemical profiles of human subcutane-
18. Curti T, Pasquinelli G, Preda P et al. An ultrastructural ous fat tissue microvascular endothelial cells. Ann Vasc
and immunocytochemical analysis of human endothelial Surg 1993; 7:541-548.
cell adhesion on coated vascular grafts. Ann Vasc Surg 34. Stansby G, Fuller B, Hamilton G. Human omental mi-
1989; 3:351-363. crovascular endothelial cells: Are they endothelial? In: Zilla
19. Preda P, Pasquinelli G, Vici M et al. Isolation of human PP, Fasol RD, Callow A eds. Applied Cardiovascular bi-
microvascular endothelial cells using monoclonal anti- ology. Basel: Karger 1992:120-123.
CD34 coated dynabeads. VIII International symposium on 35. Meerbaum SO, Sharp Wv, Schmidt SP. Lower extremity
the biology of vascular cells. August 30-September 4 1994; revascularization with polytetrafluoroethylene grafts
Heidelberg, Germany. seeded with microvascular endothelial cells. In: Zilla PP,
20. Chen SF, Fei X, Li SH. A new simple method for isola- Fasol RD, Callow A eds. Applied Cardiovascular biology.
tion of microvascular endothelial cells avoiding both Basel: Karger 1992:107-119.
chemical and mechanical injuries. Microvasc Res 1995; 36. Petroll WM, Jester JV, Cavanagh HD. In vivo confocal
50:119-128. imaging: General principles and application. Scanning
21. Hernando A, Garcìa-Honduvilla N, Bellòn JM et al. Coat- 1994; 16:131-149.
ings for vascular prostheses: Mesothelial cells express spe- 37. Serre CM, Papillard M, Chavassieux P et al. In vitro in-
cific markers for muscle cells and have biological activity duction of a calcifying matrix by biomaterials constituted
similar to that of endothelial cells. Eur J Vasc Surg 1994; of collagen and/or hydroxyapatite: An ultrastructural com-
8:531-536. parison of three types of biomaterials. Biomaterials 1993;
22. Carter D, True L, Otis CN. Serous membranes. In: 14:97-106.
Sternberg SS ed. Histology for pathologists. New York: 38. Lee YS. Morphogenesis of calcification in porcine
Raven Press 1992:499-514. bioprosthesis: Insight from high resolution electron mi-
23. Schurch W, Seemayer TA, Gabbiani G. Myofibroblast. In: croscopic investigation at molecular and atomic resolu-
Sternberg SS ed. Histology for pathologists. New York: tion. J Electron Microsc (Tokyo) 1993; 42:156-165.
Raven Press 1992:109-144. 39. Zhang Z, King M, Guidoin R et al. In vitro exposure of a
24. Brooks JJ, Perosio PM. Adipose tissue. In: Sternberg SS novel polyesterurethane graft to enzymes: A study of the
ed. Histology for pathologists. New York: Raven Press biostability of the Vascugraft arterial prosthesis.
1992:33-60. Biomaterials 1994; 15:1129-1144.
25. Holtfhofer H, Virtanen I, Kariniemi AL et al. Ulex 40. Guidoin R, Maurel S, Chakfé N et al. Expanded
europaeus 1 lectin as a marker for vascular endothelium polytetrafluoroethylene arterial prostheses in humans:
in human tissues. Lab Invest 1982; 47:60-66. Chemical analysis of 79 explanted specimens. Biomaterials
26. Newman PJ, Albelda SM. Cellular and molecular aspects 1993; 14:694-704.
of PECAM-1. Nouv Rev Fr Hematol 1992; 34: S9-S13. 41. Cenacchi G, Musiani M, Gentilomi G et al. In situ hy-
27. Krause DS, Fackler MJ, Civin CI et al. CD34: Structure, bridization at the ultrastructural level: Localization of cy-
biology, and clinical utility. Blood 1996; 87:1-13. tomegalovirus DNA using digoxigenin labelled probes. J
28. Schlingemann RO, Dingjan GM, Emeis JJ et al. Mono- Submicros Cytol Pathol 1993; 25:341-345.
clonal antibody PAL-E specific for endothelium. Lab In- 42. Scott-Burden T, Tock CL, Schwarz et al. Genetically en-
vest 1985; 52:71-76. gineered smooth muscle cells as linings to improve the
biocompatibility of cardiovascular prostheses. Circulation
1996; 94: II235-II238.
CHAPTER 8
Functional Aspects of Microvascular Cell Isolates
M. Fittkau, Teddy Fischlein
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
116 Tissue Engineering of Prosthetic Vascular Grafts
approximately 6-fold higher than in confluent cell cultures.14 for passaging of cultured cells, causes the destruction of those
In human macrovascular endothelial cells (saphenous vein), proteoglycans.
thrombomodulin activation was seen even after transplan-
tation onto ePTFE vascular prostheses.15 Profibrinolytic and Antifibrinolytic Properties
Plasmin
Plasminogen Activator Inhibitor (PAI) 12. Des Rosiers C, Nees S, Gerlach E. Purine metabolism in
PAI is the physiological inhibitor of the plasminogen cultured aortic and coronary endothelial cells. Biochem
activator. Among the stimuli for its release are inflamma- Cell Biol 1989; 67:8-15.
tion, hormones and cytokines. Its molar concentration in 13. Carley WW, Niedbala MJ, Gerritsen ME. Isolation, culti-
vation and partial charaterization of microvascular endot-
plasma is higher than that of tPA. As a potent regulator of
helium derived from human lung. Am J Respir Cell Mol
the fibrinolytic endothelial function, PAI is produced by both Biol 1992; 7:620-630.
macrovascular and microvascular endothelial cells.21-30 14. Anders E, Alles J, Delvos U, Pötzsch B et al. Microvascu-
lar endothelial cells from human omentum tissue. Modi-
Conclusion fied method for long-term cultivation and new aspects of
In summary, the metabolic functional performance characterization. Microvasc Res 1987; 34:239-249.
of microvascular endothelial cells corresponds to that of their 15. Gillis-Haegerstrand C, Frebelius S, Haegerstrand A,
macrovascular counterparts. Differences are mainly found Swedenborg J. Cultured human endothelial cells seeded
in the quantity of the synthetic activities. However, it ap- on expanded polytetrafluoroethylene support thrombin-
pears that following transplantation of microvascular en- mediated activation of protein C. J Vasc Surg 1996;
dothelial cells, a certain adaptation to the new environment 24(2):226-234.
16. Marcum JA, Rosenberg RD. Heparinlike molecules with
will be necessary. Even when mixed cultures were seeded in
anticoagulant activity are synthesized by cultured endot-
the course of animal experiments (dog), a regulation and helial cells. Biochemical and Biophysical Research Com-
adaptation to the physiological requirements could be ob- munications 1985; 126 No 1:365-372.
served.31 Whether the same observations will apply to hu- 17. Van Hinsbergh VWM. Regulation of the synthesis and
mans is questionable. Due to the lack of systematic exami- secretion of plasminogen activators by endothelial cells.
nation following clinical studies, relevant results cannot as Haemostasis 1988; 18:307-327.
yet be supplied. 18. Rijken DC, Van Hinsbergh VWM, Sens EHC.
Quantitation of tissue-type plasminogen activator in hu-
References man endothelial cell cultures by use of an enzyme immu-
1. Jarell BE, Williams SK, Stokes G et al. Use of freshly iso- noassay. Thromb Res 1984; 33:145-153.
lated capillary endothelial cells for the immediate estab- 19. Van Hinsbergh VWM, Binnema D, Scheffer MA,
lishment of a monolayer on a vascular graft at surgery. Sprengers ED et al. Production of plasminogen activators
Surgery 1986; 100:392-399. and inhibitor by serially propagated endothelial cells from
2. Radomski JS, Jarrel BE, Williams SK et al. Initial adher- adult human blood vessels. Arteriosclerosis 1987;
ence of human capillary endothelial cells to Dacron. J Surg 7:389-400.
Res 1987; 42:133-140. 20. Van Hinsbergh VWM, Sprengers ED, Kooistra T. Effect
3. Weiss. Cell tissue biology New York: Urban & of thrombin on the production of plasminogen activators
Schwarzenberg. 1988:191-205. and PA inhibitor-1 by human foreskin microvascular en-
4. Fittkau M, Fischlein T, Reichart B, Juchem G, Nees S. dothelial cells. Thromb. Haemostasis 1987; 57:148-153.
Autologe Endothelialisierung menschlicher Gefä !% 21. Speiser W, Anders E, Preissner KT, Wagner O et al. Dif-
prothesen. Z Kard 1994; 83, Supp:1:38. ferences in coagulant and fibrinolytic activities of cultured
5. Wu KK, Thiagarajan P. Role of endothelium in throm- human endothelial cells derived from omental tissue
bosis and hemostasis. Annu Rev Med 1996; 47:315-331. microvessels and umbilical veins. Blood 1987; 69:964-967.
6. Stansby G, Shukla N, Hamilton G, Jeremy J. Comparison 22. Booyse FM, Osikowicz G, Feder S, Scheinbuks J. Isola-
of prostanoid synthesis in cultured human vascular en- tion and characterization of a urokinase-type plasmino-
dothelial cells derived from omentum and umbilical vein. gen activator (Mr = 54,000) from cultured human endot-
Eur J Vasc Surg 1991; 5:501-506. helial cells indistinguishable from urokinase. J Biol Chem
7. Budd JS, Allen K, Hartley J, Walsh A et al. Prostacyclin 1984; 259:7198-7205.
production from seeded prosthetic vascular grafts. Br J 23. Booyse FM, Scheinbuks J, Radek J, Osikowicz G et al.
Surg 1992; 79:1151-1153. Immunological identification and comparison of plasmi-
8. Sterpetti AV, Schultz RD, Hunter WJ et al. Comparison nogen activator forms in cultured normal human endot-
of two techniques to isolate microvascular endothelial cells helial cells and smooth muscle cells. Thromb Res 1981;
from the omentum. J Surg Res 1990; 48:101-106. 24:495-504.
9. Fischlein T, Lehner G, Lante W, Fittkau M et al. 24. Loskutoff DJ, Van Mourik JA, Erickson LA, Lawrence D.
Endothelialization of cardiac valve bioprotheses. Artif Detection of an unusually stable fibrinolytic inhibitor pro-
Organs 1994; 17 No 6:345-352. duced by bovine endothelial cells. Proc Natn Acad Sci
10. Radomski MW, Palmer PMJ, Moncada S. The anti-aggre- USA 1983; 80:2956-2960.
gating properties of vascular endothelium interactions 25. Emeis JJ, Van Hinsbergh VWM, Verheijen JH, Wijngaards
between prostacyclin and nitric oxide. Br J Pharmacol G. Inhibition of tissue-type plasminogen activator by con-
1987; 92:639-642. ditioned medium from cultured human and porcine vas-
11. Ungureanu-Longrois D, Balligand JL, Okada I et al. Con- cular endothelial cells. Biochem Biophys Res Commun
tractile responsiveness of ventricular myocytes to isopro- 1983; 110:392-398.
terenol is regulated by induction of nitric oxide synthase 26. Philips M, Juul A, Thorsen S. Human endothelial cells
activity in cardiac microvascular endothelial cells in het- produce a plasminogen activator inhibitor and a tissue-
erotypic primary culture. Circ Res 1995; 77 No 3:486-493. type plasminogen activator-inhibitor complex. Biochem
Biophys Acta 1984; 802:99-110.
120 Tissue Engineering of Prosthetic Vascular Grafts
27. Van Hinsbergh VWM, Bertina RM, Van Wijngaarden A, 30. Sprengers ED, Verheijen JH, Van Hinsbergh VWM, Emeis
Van Tilburg NH et al. Activated protein C decreases JJ. Evidence for the presence of two different fibrinolytic
plasminogen activator-inhibitor activity in endothelial cell- inhibitors in human endothelial cells conditioned me-
conditioned medium. Blood 1985; 65:444-451. dium. Biochem Biophys Acta 1984; 801:163-170.
28. Van Mourik JA, Lawrence DA, Loskutoff DJ. Purification 31. Baitella-Eberle G, Groscurth P, Zilla P, Lachat M et al.
of an inhibitor of plasminogen activator (antiactivator) Long-term results of tissue development and cell differ-
synthesized by endothelial cells. J Biol Chem 1984; entiation on Dacron prostheses seeded with microvascu-
259:14914-14921. lar cells in dogs. J Vasc Surg 1993; 18 (6):1019-1028.
29. Dosne AM, Dupuy E, Bodevin E. Production of a fibrin-
olytic inhibitor by cultured endothelial cells derived from
human umbilical vein. Thromb Res 1978; 12:377-387.
CHAPTER 9
Automated Seeding Devices
Dominic Dodd, J. Vincent Smyth, Michael G. Walker
Introduction
A lmost as soon as the application of endothelial seeding to enhance graft patency was
recognized, groups started to develop techniques and equipment to facilitate the process.
The requirements of an ideal seeding system for clinical work include simplicity, efficiency
and speed. These considerations must be applied to the delivery of a high yield of viable
endothelial cells using a readily available tissue source. The maintenance of sterility to avoid
introduction of infective agents; effective seeding onto the graft while retaining the func-
tional integrity of seeded cells; and the rapid development of monolayer within a time-
frame suitable for operating room use are also fundamental. The search for a reproducible,
inexpensive, low-tech, high efficacy technique has been a tortuous and convoluted one in
which the solution to each problem has led to further questions.
In the design of a fully automated seeding device, several key steps must be consid-
ered. The automated seeding process commences with tissue reception. A closed system in
which sterility is maintained is essential. The tissue receiver may be combined with a me-
chanical homogenization process to speed subsequent cell separation by enzymatic diges-
tion. The aim of this stage is to acquire large numbers of endothelial cells (ECs) with rela-
tively few contaminating cell types. Subcutaneous adipose tissue is a ready source of mi-
crovascular endothelial cells and is easily harvested in sufficient quantities from all but the
thinnest of patients. Once ECs have been isolated, maximal graft-cell attachment is needed.
The constraints of the operating room mean that this must be achieved in a relatively short
time. Graft surface coatings clearly have a major role in determining the efficiency of this
stage. Finally, the seeded graft must be presented to the operating team in a package that will
maintain its sterility and preserve the viability of the seeded EC lining. Ideally, a fully auto-
mated system should optimize cell harvest and seeding efficiency; however, a compromise
between these aims and the duration of the procedure must be met. Completion of the
seeding process in approximately one hour is a realistic time scale in a busy operating room.
Much of this time will be taken by cell digestion and separation, leaving little time for cell
attachment or final rinsing.
If vascular graft seeding is to be widely adopted in clinical practice, a reliable, easily
used system is required that can be accommodated in a conventional operating room. It
should be recognized that surgeons’ enthusiasm for the introduction of a complex and time
consuming process into the operating room is limited. Unless the process is automated,
vascular graft seeding will remain a “laboratory” technique, confined to very few surgical
centers. Therefore, in addition to the development of graft materials and coatings for opti-
mal seeding performance, automation of the seeding process must be achieved. This aspect
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
122 Tissue Engineering of Prosthetic Vascular Grafts
of seeding technology is both a biological and engineering onto a fibronectin precoated graft and incubated for at least
challenge. Perhaps the difficulties in successfully achieving 30 minutes.7 To produce a complete monolayer on a graft
this goal are reflected in the relatively small number of pub- would thus require an initial harvest of 1.5 x
lications of techniques and designs investigated. 105−.−d−l−(% Seeding Efficiency) cells, where d=inside diam-
eter and l = length, of the graft. Our own work with imme-
Early Seeding Techniques diately harvested saphenous vein endothelial cells has dem-
At the beginning of the 1980s, seeding methodology onstrated that a seeding efficiency of 30% can be reliably
relied on mechanical harvesting of the endothelial cells by achieved,8 and thus for a standard 30 cm, 6 mm internal
metal pledget from vein segments.1 As well as being labori- diameter graft suitable for a femoropopliteal graft, an initial
ous, the yield of viable cells was relatively low due to harvest harvest of 28 x 106 cells is required. A 60 cm long, 4 mm
trauma. In addition, the low specificity of mechanical har- internal diameter graft such as might be used for femorotibial
vesting resulted in frequent contamination of the endothe- bypass would require 38 x 106 cells. This quantity of endot-
lial culture with fibroblasts and smooth muscle cells. This helial cells can not be derived from autologous saphenous
technique sufficed for laboratory work and was successful vein, since such cell harvest would require a vein segment of
using animal cell lines, but a major step forward was made equivalent size, which would normally be used as the pre-
with the adoption of enzymatic harvesting, leading to the ferred conduit. Microvascular endothelial cells have been
now routine collagenase incubation-based method.2 This proposed as an alternative cell type,9,10 with harvests from
rapidly superseded mechanical harvesting as a straightfor- both omentum and subcutaneous adipose tissue being used
ward method which allowed rapid procurement of pure for seeding experimentally. However, considerably more
endothelial cells, and the promise of seeding prosthetic grafts experimental data have been gained with macrovascular
in terms of patency, resistance to infection and reduced endothelial cells to suggest inhibition of smooth muscle cell
neointimal hyperplasia began to be realized in a range of proliferation in vivo, and acceptable results of retention
experimental laboratory and in vivo models. However, only under arterial levels of shear stress.
animal cell lines had as yet been used extensively and the
next step was clearly to extend this work to clinical practice. Graft Material Considerations
Up to the early 1980s, work with human cell lines had The large majority of EC seeding studies have been
been restricted to umbilical vein endothelium, HUVECs, carried out using commercially available graft materials that
because of difficulties with maintaining adult endothelial were not specifically designed for cell attachment. Indeed,
cell lines, HAECs, in culture. The next breakthrough in en- considerable materials technology has been applied to make
dothelial seeding was the successful culture of HAECs by the luminal surface of these graft materials hydrophobic by
Jarrell, with the recognition of the need for EDGF and hep- negatively-charged fluoropolymer lining and consequently
arin in the culture medium.3 Despite significant problems unsuitable for direct seeding.
with the more fragile human cell cultures and the disap- There has been continued interest over the last de-
pointing results of cell attachment studies detailed in an ear- cade on choice of graft coating materials for optimal cell
lier chapter, initial protocols using harvest, culture and seed- attachment. It is now recognized that the extracellular ma-
ing stages demonstrated that at least some benefit might be trix plays an integral role in cellular function and the choice
realized. In clinical practice, however, the time constraints of graft coating material will influence not only cell attach-
of the operating room meant that cell harvesting would need ment but also cellular activity. Soluble components of the
to be carried out as a preliminary step, or a more rapid tech- extracellular matrix such as fibronectin, collagen and laminin
nique of producing an endothelial monolayer was needed, have all been used to achieve comparable cell attachment
or clinical seeding would be carried out at subconfluent den- rates.11,12 Recently, synthetic EC binding site peptides con-
sities. Subconfluent seeding, whilst effective in the labora- taining arginine-glycine-aspartic acid (RGD) have been used
tory, has been shown to be less able to resist the shear stress to precoat ePTFE, with a significant increase in EC attach-
of flow than an intact monolayer,4 and has been largely aban- ment and retention compared with fibronectin.13 The use
doned. In those clinical studies that have been carried out, of RGD peptides is attractive commercially, as they are simple
the seeding densities achieved were probably subconfluent, to produce and cheaper to use than more complex protein
and did not result in measurable reduction in coatings. EC attachment through focal adhesions is RGD
thrombogenicity measured by labeled platelet scintigraphy,5 dependent; if no RGD sequences are detected in extracellu-
though there was some indirect evidence from serum mark- lar matrix, apoptosis occurs.
ers of a reduction in platelet adhesion and activation.6 In contrast, Sipehia reported the use of gaseous
“plasma” surface modification to enhance cellular attach-
Optimal Conditions for Seeding ment and replication.14 He reported the addition of amino
groups to the surface of a PTFE membrane by anhydrous
Cellular Considerations ammonia in a gaseous plasma increased bovine arterial EC
In order to achieve the maximum coverage from a lim- attachment from 36 to 92% after 96 h in culture.
ited source of cells for seeding, optimal conditions for seed- This suggests that appropriate surface engineering of
ing were extensively investigated. Work with both expanded the prosthetic material to be seeded may have as large an
polytetrafluoroethylene (ePTFE) and polyester graft mate- influence on the rapid development of a healthy neointima
rials suggests the use of at least 150,000 cells/cm2, seeded as cell delivery and modification itself.
Automated Seeding Devices 123
To seed a typical peripheral vascular graft using our cell monolayer of typical endothelial appearance
device, the operator introduces 100-300 g adipose tissue into (Figs. 9.2A,B). Full characterization of the monolayer has
a tissue receiver and starts the seeding program, which is been complicated by the apparent loss of many cell surface
controlled by the machine’s central processing unit. The adi- immunomarkers with the digestion process. Indeed, we have
pose tissue is minced and forced through a metallic mesh by been unable to demonstrate the presence of tPA, vWF, CD34,
an air-driven piston into a tissue digestion flask containing CD31, eNOS or bFGF in our specimens.
300 ml of buffered saline at 37°C. Into this is added 1,000 IU We have used the seeding system for in vivo experi-
collagenase II per gram of adipose tissue and digestion con- mentation using a porcine model. Subcutaneous adipose
tinues with constant agitation for 40 minutes. After this time tissue was harvested from immature Yucatan miniswine and
the digested fat forms a slurry which is coarsely filtered and a segment of 6 mm internal diameter graft implanted im-
seeded under pressure into the interstices of a specially pre- mediately after processing into the infrarenal aorta. After
pared woven polyester graft. The graft has been precoated four weeks, the graft was explanted and compared with
with bovine gelatin and retains a porosity of approximately nonseeded control grafts, also implanted for four weeks. In
1500 ml/cm2/min. After seeding, this porosity is reduced to our study there was no detectable loss in early patency fol-
almost zero at arterial pressure levels. The process takes 50 lowing seeding with a mixed cell population. The presence
to 55 minutes to complete and the graft can be removed of potentially thrombogenic tissue debris in the graft inter-
from the machine and handed to the operating team in a stices did not have a detrimental effect in the immediate
sterile blister pack. This device allows for a fully automated postimplantation phase. However, at four weeks the seeded
seeding process to be undertaken in the operating room samples, whilst remaining patent, demonstrated neointimal
without excessively prolonging the surgical procedure, or hyperplasia, with marked SMC proliferation. We surmise
requiring on-site laboratory facilities or highly skilled cell that, despite endothelial cover, neointimal hyperplasia oc-
culture technicians. Its secondary advantages are the reduc- curs and that this may be due to seeding of SMC which are
tion in risk of pathogen transfer and the avoidance of pre- stimulated in the process. To overcome this problem we pro-
liminary cell harvesting surgery. The main disadvantage with posed the introduction of a cell separation process to mini-
our particular machine is that a heterogeneous cell popula- mize the seeding of nonendothelial cells. The most promis-
tion is initially seeded onto the graft surface. This popula- ing way to include EC purification in an automated seeding
tion is difficult to fully characterize, but is anticipated to in- device would be to use immunomagnetic bead separation.33
clude vascular smooth muscle cells, fibroblasts, adipocytes, Beads coated with antibodies to the EC surface antigen CD34
and pericytes. If omentum is used, then PMC may also be are already commercially available and are approved for clini-
present, although the digestion conditions in the machine cal use in hemopoietic stem cell separation (Baxter
are considerably more rigorous than those normally used Healthcare Ltd, Thetford, UK). However, there is a time pen-
for PMC harvest from omentum and it is likely that most alty incurred with this approach. To offset this, work is in
PMC would be digested. progress to determine the feasibility of replacing enzymatic
In vitro experiments using human subcutaneous adi- tissue digestion with a mechanical homogenization process
pose tissue and omentum and subsequent graft culture in to reduce the overall procedure time. We estimate that a
our laboratory have confirmed the rapid development of a highly selected EC suspension can be seeded onto a pros-
Automated Seeding Devices 125
thesis in under two hours. While this time scale may be un- References
attractive to some vascular surgeons, we feel that the group 1. Herring MB, Gardner AL, Glover J. A single staged tech-
of patients for whom this technology is required are worth nique for seeding vascular grafts with autogenous endot-
the extra time spent in their treatment. helium. Surgery 1978; 84:498-504.
2. Jaffe EA, Nachman RL, Becker CG, Minick CR. Culture
Summary of human endothelial cells derived from umbilical veins.
J Clin Invest 1973; 52:2745-56.
Although our understanding of vascular biology, and
3. Jarrell BE, Levine E, Shapiro S, Williams SK, Carabasi RA,
in particular the elucidation of many cell-matrix signaling Mueller S, Thornton S. Human adult endothelial cell
interactions, have led to dramatic breakthroughs in the labo- growth in culture. J Vasc Surg 1984; 1:757-64.
ratory, endothelial cell seeding of vascular prosthetic grafts 4. Miyata T, Conte MS, Trudell LA, Mason D, Whittemore
is not widely regarded as clinically useful. Until an automated AD, Birinyi LK. Delayed exposure to pulsatile shearstress
harvesting and delivery system is fully developed, it remains improves retention of human saphenous vein endothelial
unlikely that this will change. cells on seeded ePTFE grafts. J Surg Res 1991; 50:485-93.
It is hoped that the development of new surface treat- 5. Jensen N, Lindblad B, Bergqvist D. Endothelial cell seeded
ments will lead to greatly increased rates of seeded cell ad- dacron aortobifurcated grafts: Platelet deposition and
herence, retention and replication. In addition the role of long-term follow-up. J Cardiovasc Surg 1994; 35:425-9.
6. Ortenwall P, Wadenvik H, Kutti J, Risberg B. Endothelial
genetically modified EC with enhanced antithrombotic ac-
cell seeding reduces thrombogenicity of Dacron grafts in
tivity is just now being recognized. It is possible that humans. J Vasc Surg 1990; 11:403-410.
immunomodulated, cryopreserved bioprosthetic grafts will 7. Vohra R, Thomson GJL, Carr HMH, Sharma, Walker MG.
represent the next generation of small diameter arterial by- Comparison of different vascular prostheses and matrices
pass prostheses. in realtion to endothelial seeding. Br J Surg 1991;
78:417-20.
126 Tissue Engineering of Prosthetic Vascular Grafts
8. Carr HMH, Vohra R, Sharma H, Smyth JV, Rooney OB, ing using endothelial cell seeded PTFE grafts: Five-year
Dodd PD, Walker MG. Endothelial cell seeding kinetics clinical experience. Ann Vasc Surg 1995; 9:S15-23.
under chronic flow in prosthetic grafts. Ann Vasc Surg 22. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T,
1996; 10:469-75. Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I,
9. Stansby G, Shukla N, Fuller B, Hamilton G. Seeding of Fischlein T. Clinical in vitro endothelialization of
human microvascular endothelial cells onto femoropopliteal bypass grafts: An actuarial follow-up over
polytetrafluoroethylene graft material. Br J Surg 1991; three years. J Vasc Surg 1994; 19:540-8.
78:1189-92. 23. Jarrell BE, Williams SK, Stokes G, Hubbard FA, Carabasi
10. Schmidt SP,Monajjem N, Evancho MM, Pippert TR, RA, Koolpe E, Greene D, Pratt K, Moritz MJ, Radomski
Sharp WV. Microvascular endothelial cell seeding of J, Speicher L. Use of freshly isolated capillary endothelial
small-diameter Dacron vascular grafts. J Invest Surg 1988; cells for the immediate establishment of a monolayer on
1:35-44. a vascular graft at surgery. Surgery 1986; 100:392-9.
11. Thomson GJ, Vohra RK, Carr HMH, Walker MG. Adult 24. Visser MJ, van Bockel JH, van Muijen GN, van Hinsbergh
human endothelial cell seeding using expanded VW. Cells derived from omental fat tissue are not endot-
poltytetrafluoroethylene vascular grafts: A comparison of helial in origin. A study on the origin of epitheloid cells
four substrates. Surgery 1991; 109:20-7. deirved from omentum. J Vasc Surg 1991; 13:373-81.
12. Koveker GB, Graham LM, Burkel WE, Sell R, Wakefield 25. Williams SK, Kleinert LB, Rose D, McKenney S. Origin
TW, Dietrich K, Stanley JC. Extracellular matrix prepara- of endothelial cells that line expanded
tion of expanded polytetrafluoroethylene grafts seeded polytetrafluoroethylene vascular grafts sodded with cells
with endothelial cells: Influence on early platelet deposi- from microvascularized fat. J Vasc Surg 1994; 19:594-604.
tion, cellular growth and luminal prostacyclin release. 26. Vici M, Pasquinelli G, Preda P, Martinelli GN, Gibellini
Surgery 1991; 109:313-9. D, Freyrie A, Curti T, D’Addato M. Electron microscopic
13. Walluscheck KP, Steinhoff G, Kelm S, Haverich A. Im- and immunocytochemical profiles of human subcutane-
proved endothelial cell attachment on ePTFE vascular ous fat tissue microvascular endothelial cells. Ann Vasc
grafts pretreated with synthetic RGD-containing peptides. Surg 1993; 7:541-8.
Eur J Vasc Endovasc Surg 1996; 12:321-30. 27. Pronk A, Hoynck van Papendrecht AAGM, Leguit P,
14. Sipehia R. The enhanced attachment and growth of Verbrugh HA, Verkooyen RPAJ, van Vroonhoven TJMV.
endothelial cells on anhydrous ammonia gaseous plasma Mesothelial cell adherence to vascular prostheses and their
modified surfaces of polystyrene and subsequent growth in vitro. Cell Transplantation 1994;
poly(tetrafluoroethylene). Biomater Artif Cells Artif Or- 3:41-8.
gans 1990; 18:437-46. 28. Nicholson LJ, Clarke JMF, Pittilo RM, Machin SJ, Woolf
15. Gerlach J, Kreusel KM, Schauwecker HH, Bucherl ES. En- N. The mesothelial cell as a non-thrombogenic surface.
dothelial cell seeding on PTFE vascular prostheses using Thromb Haemostas 1984; 52:102-4.
a standardized seeding technique. Artif Organs 1989; 29. Williams SK, Wang TF, Castrillo R, Jarrell BE.
12:270-5. Liposuction-derived human fat used for vascular graft
16. van Wachem PB, Stronck JW, Koers-Zuideveld R, Dijk F, sodding contains endothelial and not mesothelial cells as
Wildevuur CR. Vacuum cell seeding: A new method for the main cell type. J Vasc Surg 1994; 19:916-23.
the fast application of an evenly distributed cell layer on 30. Noishiki Y, Yamane Y, Tomizawa Y, Okoshi T, Satoh S,
porous vascular grafts. Biomaterials 1990; 11:602-6. Wildevuur CR. Endothelialization of vascular prostheses
17. Nosihiki Y, Yamane Y, Tomizawa Y. Sealing of highly by transplantation of venous tissue fragments. ASAIO
porous fabric vascular prostheses by adipose connective Trans 1990; 36:346-8.
tissue fragments instead of preclotting with fresh blood. J 31. Pasic M, Muller-Glauser W, von Segesser LK, Lachat M,
Invest Surg 1993; 6:231-40. Mihaljevic T, Turina M. Superior late patency of small-
18. Williams SK, Jarrell BE, Rose DG, Pontell J, Kapelan BA, diameter Dacron grafts seeded with omental microvascu-
Park PK, Carter TL. Human microvessel endothelial cell lar cells: An experimental study. Ann Thorac Surg 1994;
isolation and vascular graft sodding in the operating room. 58:677-83.
Ann Vasc Surg 1989; 3:146-52. 32. Pasic M, Muller-Glauser W, Odermatt B, Lachat M, Siefert
19. Mazzutocelli JP, Roudiere JL, Bernex F, Bertrand P, B, Turina M. Seeding with omental cells prevents late
Leandri J, Loisance D. A new device for endothelial cell neointimal hyperplasia in small-diameter Dacron grafts.
seeding of a small-caliber vascular prosthesis. Artif Or- Circulation 1995; 92:2605-16.
gans 1993; 17:787-90. 33. Jackson CJ, Garbett PK, Nissen B, Schrieber L. Binding
20. Budd JS, Allen KE, Bell PRF. Effects of two methods of of human endothelium to Ulex europaeus I-coated
endothelial cell seeding on cell retention during blood Dynabeads: Application to the isolation of microvascular
flow. Br J Surg 1991; 78:878-82. endothelium. J Cell Sci 1990; 96:257-62.
21. Leseche G, Ohan J, Bouttier S, Palombi T, Bertrand P,
Andreassian B. Above-knee femoropopliteal bypass graft-
CHAPTER 10
Healing Patterns Following Microvascular Seeding—
A Clinical Evaluation of Microvascular-Seeded
A-V Access Grafts
Steven P. Schmidt, Sharon O. Meerbaum, Duane L. Donovan
Introduction
T he need for a more successful artificial bypass graft to replace small and medium-diam
eter blood vessels remains an important issue in vascular surgery. Approximately 600,000
patients require arterial reconstructive surgeries in the United States annually, including
20,000 femoropopliteal bypasses. In addition, each year approximately 90,000 new patients
with end stage renal disease (ESRD) present to vascular surgeons for access-related surgeries
for chronic hemodialysis. Surgeons prefer to use autologous native vessels as bypass con-
duits. For peripheral revascularizations, the collective experiences of vascular surgeons have
been that long term performances of bypass grafts constructed from native veins are supe-
rior to those using other biologic or synthetic alternatives. Likewise, autogenous arterio-
venous (AV) fistulas of the Brescia-Ciminio type are the hemodialysis access procedures
that theoretically provide the longest term trouble-free patency compared to synthetic graft
alternatives. Nevertheless, limitations of the use of native veins as bypass grafts have been
described. Less than 15% of the entire dialysis population have well-functioning native AV
fistulas. Eighty percent of dialysis patients receive polytetrafluoroethylene (PTFE) vascular
grafts for access, which is accompanied by the morbidity attendant with implantation of
these grafts. When used as peripheral vascular replacements, one-quarter to one-third of
saphenous vein bypass grafts deteriorate, as assessed by histology, in the relatively higher
flows and pressures of the arterial circulation. It is common, therefore, for patients with no
usable native veins available for grafting (or remaining veins of poor quality) to present to
vascular surgeons for repeated revascularization procedures. The use of synthetic vascular
grafts is imperative in these patients.
The long term performances of commercially available synthetic vascular prosthetics
are poor. Primary patency of PTFE grafts used for AV access is approximately one year;1 the
patency rate for PTFE grafts implanted for femorodistal reconstruction is approximately
20% at three years.2 Failure of vascular grafts at short times postoperatively has been gener-
ally attributed to the combination of low flow hemodynamics plus the inherent
thrombogenicities of the graft materials. Late graft failure most frequently occurs from anas-
tomotic neointimal hyperplasia (NIH) and consequent narrowings at the outflow tract. In
synthetic grafts used for hemodialysis access, midgraft hyperplasia adjacent to frequently
used needle puncture sites may also cause failure. The etiology of NIH remains to be clarified.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
128 Tissue Engineering of Prosthetic Vascular Grafts
One of the early assumptions for the superiority of ferences between seeded and nonseeded graft patencies in
autogenous vascular grafts compared to synthetic grafts was this study and the extent of endothelialization of seeded
the presence of an intact endothelium on the luminal sur- grafts was minimal. Ortenwall et al7,8 reported reduced plate-
faces of the native grafts. Under in vivo conditions of nor- let deposition on both seeded Dacron and polytetra-
mal circulatory physiology, endothelial cells (ECs) are the fluoroethylene grafts implanted in patients. Kadletz et al9
most nonthrombogenic cells known. The blood-contacting also observed little or no platelet accumulation on PTFE
(luminal) surfaces of synthetic vascular grafts implanted in prostheses that had been endothelialized in vitro prior to
animals endothelialize, in time, along their entire lengths implantation in patients. Herring et al10 compared the per-
and typically achieve long term patencies. This endo- formances of 66 seeded PTFE grafts with 53 autologous vein
thelialization occurs, however, only in the anastomotic re- grafts and concluded that at 30 months vein graft patency
gions of human vascular grafts and is limited at most to was superior and that seeding did not improve patency in
2-3 cm of ingrowth. below the knee bypasses. In contrast, Leseche and his col-
Although the issues remain debatable, it has generally leagues11 reported good clinical results when endothelial cell-
been accepted that humans do not heal synthetic bypass seeded PTFE grafts were implanted as above-knee
grafts with an endothelial cell lining as other animals do. As femoropopliteal bypass grafts. These investigators utilized a
a consequence of this failure to heal, in addition to other two stage seeding technique in which a period of in vitro
clinical issues, it is generally regarded as not feasible to uti- incubation was utilized to increase the number of available
lize small caliber vascular grafts (< 6 mm I.D.) clinically in autologous endothelial cells for transplantation onto the
patients, as these grafts fail quickly from accumulated throm- graft.
bus and/or neointimal hyperplasia. The bottom line in this discussion is that the efficacy
The logical assumption of many researchers has been of endothelial cell seeding in improving prosthetic graft pa-
that synthetic graft performances in humans may be en- tency has not been resolved. In fact, proving that graft per-
hanced and patencies extended by promoting formance is indeed enhanced as a result of endothelial cell
endothelialization of the luminal surfaces of these synthetic seeding is complicated in and of itself. Among investigators
grafts. One method of creating such an endothelialized sur- there has been minimal consensus related to many techni-
face is transplantation of autogenous ECs onto the blood- cal issues. One such issue has been the definition of the spe-
contacting graft surface. A new research technology has thus cific tissue source of endothelial cells for transplantation.
evolved called “endothelial cell seeding (ECS)” to investi- Our laboratory program of endothelial cell transplantation
gate this possibility. The underlying logic supporting ECS has focused upon the use of microvascular endothelial cells
investigations is that the presence of transplanted endothe- (MVEC) derived from enzymatic processing of subcutane-
lium mimics the anti-thrombogenic intima of native ves- ous fat. We have been impressed with the relative numbers
sels. In addition, it has been suggested that rapid of endothelial cells that can be derived from fat sources com-
endothelialization in anastomotic regions may normalize pared with the limited endothelial cell harvests typically
aberrant flow patterns, which are thought to contribute to available from autologous blood vessels. Because subcuta-
the genesis of intimal hyperplastic lesions.3 neous fat from which MVEC can be derived is available in
Animal studies have shown that endothelialization of virtually all patients presenting for vascular surgery, we ini-
prosthetic grafts can be encouraged by transplanting endo- tiated a clinical trial of endothelial cell seeding in patients
thelial cells into or onto the graft at the time of implanta- requiring PTFE grafts for A-V access. The study compared
tion. These endothelial cell-seeded grafts typically have im- the histological appearances of biopsies derived from MVEC-
proved patencies versus nonseeded controls when evaluated seeded and nonseeded ePTFE arteriovenous fistulas im-
in animals. To date, however, the prospect from animal stud- planted in patients for blood access for renal dialysis.
ies of successful long term performances of endothelial cell-
seeded grafts in humans has not been realized in the clinical Materials and Methods
trials that have been reported, which are few in number when
compared with the numbers of reported animal studies. AV Fistula Study
The first true clinical trial of endothelial cell seeding Following informed consent, 9 patients (3 males and
in patients was reported by Herring et al.4 Seeded endothe- 6 females; mean age 59.8 years) enrolled in the AV fistula
lial cells were derived by mechanical methods from autolo- study. All of these patients had renal disease requiring long
gous veins and transplanted onto Dacron grafts, which were term hemodialysis. In this study patients were randomized
subsequently implanted in patients as femoropopliteal re- to receive either microvessel cell “seeded” ePTFE grafts or
constructions. Analyses revealed no differences in patencies “nonseeded” ePTFE grafts as AV fistulas. The purpose of the
between seeded and nonseeded grafts; smokers had espe- study was to compare the histological appearances of bi-
cially poor graft patencies. Herring and his colleagues5 were opsy samples of each type of graft at scheduled
able to confirm endothelialization of the luminal graft sur- postimplantation times including 1 month, 3 months and
face from a seeded graft retrieved from a patient via histo- 1 year after surgery. The details of the experimental
logical analysis. Zilla et al6 also reported early clinical data methologies are reported elsewhere.12 An average of 1.1 x 107
from a series of patients who underwent femoropopliteal microvessel cells were transplanted onto the seeded grafts.
bypass using endothelial cell-seeded expanded Records of the length of graft implanted were not kept, so it
polytetrafluoroethylene (ePTFE) grafts. There were no dif-
Healing Patterns Following Microvascular Cell Seeding 129
is impossible to estimate the density of the cells which were seeded graft showed significant concentric fibrous plaque
transplanted onto the graft. formation with a high level of collagenation in the region of
the thickening closest to the lumen. Large vessels could be
Results seen within the fibrous thickening (Fig. 10.1).
There were no operative complications attendant upon The nonseeded graft which was biopsied at one month
derivation of subcutaneous fat. The patients tolerated all postimplantation revealed a luminal surface with exposed
operative procedures well. Harvesting the microvessel cells graft material and some adherant red blood cells. No endo-
added one hour to the total operative time for each patient. thelial cells were seen or identified by factor VIII staining on
A complicating issue in this study was the refusal of the luminal surface of the nonseeded grafts. At 16 months a
patients to comply with the biopsy schedule. One patient biopsied section of a nonseeded graft revealed irregular ar-
agreed to a one month biopsy only. One additional patient eas of intimal fibrous thickening; however, there was no in-
agreed to biopsies at one and three months. A third patient flammatory response or calcification, and the flow-contact-
presented for biopsies at one month and 16 months. And ing surface of the nonseeded graft remained thin.
one patient was biopsied at one month, three months, 20
months and 24 months. Only two patients agreed to one Discussion
year postimplantation biopsies. Therefore, the data from this Ten years of research efforts have investigated the po-
study must be interpreted in light of only limited tential efficacy of ECS in enhancing vascular graft perfor-
postimplantation biopsy samples. mances in in vitro benchtop laboratory models of seeding,
The seeded grafts from biopsied patients at one month animal implants, and preliminary clinical studies. In our
postimplantation revealed few red blood cells and no accu- opinion, however, the definitive studies have yet to be per-
mulating thrombus. The intimal lining consisted of areas of formed and reported that support or refute ECS as a mo-
single cells in thickness to areas with beginning organiza- dality of clinical significance to the vascular surgeon. The
tion of fibrinous material. factor VIII analysis confirmed that research described in this report was intended to accom-
the neointima was lined by endothelial cells. plish that goal. The research was performed in patients re-
Interestingly, the neointimas of seeded grafts obtained quiring placement of synthetic PTFE grafts for access for
at subsequent postimplantation periods were thicker than hemodialysis. We felt that data derived from the analysis of
the neointimas at one month. The thickening consisted of endothelial cell-seeded grafts in hemodialysis patients would
concentric intimal fibrous material, which appeared as a offer insight into the potential efficacy of endothelial cell
mixture of myofibroblasts and fibroblasts with prominent seeding for other synthetic vascular graft applications as well.
collagen deposition. No significant inflammatory cells were Unfortunately patient compliance with this protocol
found. These thickened intimas were also lined by factor VIII and the post operative biopsy schedule was minimal, at best,
positive endothelial cells. At 20 months the biopsied seeded which was disappointing. This was especially disappointing
graft revealed significant concentric intimal thickening, with because in the seeded samples that were analyzed the intima
the beginning of calcification of the intimal plaque. There was greatly thickened compared to the nonseeded grafts. This
were no accumulations of foamy macrophages or choles- would be an important observation if it could be repeated
terol clefts. The luminal surface was smooth and no thrombi in a relatively large number of patients.
were observed. At 24 months, the biopsy sample from a
It is interesting to speculate on the etiology of the in- 5. Herring M, Baughman S, Glover J. Endothelium devel-
timal thickening of the seeded grafts analyzed in this study. ops on seeded human arterial prosthesis: A brief clinical
Certainly the AV fistula is an aberrant hemodynamic envi- note. J Vasc Surg 1985; 2:727-30.
ronment, in which high wall shear stresses may continually 6. Zilla P, Fasol R, Deutsch M et al. Endothelial seeding of
PTFE vascular grafts in humans: A preliminary report. J
perturb transplanted endothelial cells, resulting in a con-
Vasc Surg 1987; 6:535-41.
tinual state of activation. If this is in fact the case, then en- 7. Ortenwall P, Wadenvik H, Risberg B. Reduced platelet
dothelial cell seeding will not enhance graft performance— deposition on seeded versus unseeded segments of ex-
it will only exacerbate the inevitable failure due to intimal panded polytetrafluoroethylene grafts: Clinical observa-
hyperplasia. However, one must caution again that the re- tions after a 6-month follow-up. J Vasc Surg 1989;
sults described herein are from only a limited number of 10:374-380.
patients in the unusual hemodynamic environment of the 8. Ortenwall P, Wadenvik H, Kutti J et al. Endothelial cell
AV fistula, and may not be representative of the fate of seeded seeding reduces thrombogenicity of Dacron grafts in hu-
grafts implanted elsewhere within the vascular tree. mans. J Vasc Surg 1990; 11:403-410.
In summary, we recommend that a well-orchestrated 9. Kadletz M, Magometschnigg H, Minar E et al. Implanta-
study of endothelial cell seeding in AV fistulas be conducted tion of in vitro endothelialized polytetrafluoroethylene
grafts in human beings. A preliminary report. J Thorac
to clarify this issue, with sufficient numbers of patients such
Cardio Surg 1992; 104:736-742.
that the impact of endothelial cell seeding on the perfor- 10. Herring M, Smith J, Dalsing M et al. Endothelial seeding
mance of synthetic AV fistula can be clarified. Transplanta- of polytetrafluoroethylene femoral popliteal bypasses: The
tion of microvessel-derived endothelial cells resulted in fac- failure of low-density seeding to improve patency. J Vasc
tor VIII positive graft sections, but “graft healing” resulted Surg 1994; 20:650-655.
in thickened neointimas which jeopardized blood flow 11. Meerbaum S, Sharp W, Schmidt S. Lower extremity
through the graft. revascularization with polytetrafluoroethylene grafts
seeded with microvascular endothelial cells. In: Zilla P,
References Fasol R, Callow A, eds. Applied Cardiovascular Biology
1. Bell D, Rosenthal J. Arteriovenous graft life in chronic 1990-91. Int Soc Appl Cardiovasc Biol. Basel: Karger,
hemodialysis. Arch Surg 1988; 123:1169. 1992; 2:107-119.
2. Bergen J, Veith F, Bernhard V et al. Randomization of 12. Schmidt S, Meerbaum S, Anderson J et al. Evaluation of
autogenous vein and polytetrafluoroethylene grafts in expanded polytetrafluoroethylene arteriovenous access
femoro-distal reconstructions. Surg 1982; 92:921. grafts onto which microvessel-derived cells were trans-
3. Eguchi H, Okadome K, Mii S et al. Significance of the planted to “improve” graft performace: Preliminary re-
endothelial lining in prevention of intimal thickening of sults. Ann Vasc Surg 1998; 12:405-411.
autogenous vein grafts in dogs. J Surg Res 1991; 50:179.
4. Herring M, Gardner A, Glover J. Seeding human arterial
prostheses with mechanically derived endothelium. The
detrimental effect of smoking. J Vasc Sur 1984; 1:279-89.
CHAPTER 11
Neointimal Hyperplasia in Small Diameter Prosthetic
Vascular Grafts: Influence of Endothelial Cell Seeding
with Microvascular Omental Cells in a Canine Model
Miralem Pasic, Werner Müller-Glauser, Marko Turina
Introduction
Ideal Vascular Graft
Intimal Hyperplasia
Intimal hyperplasia is a characteristic fibromuscular cellular response to vascular in-
jury during vascular reconstruction2,3 and represents—apart from surface thrombogenicity—
the second most important reason for prosthetic graft failure. All forms of vascular recon-
struction cause both injury and a wound healing response,4 resulting in different degrees of
intimal or neointimal proliferation after reconstruction. Neointimal hyperplasia, particu-
larly pronounced in the area of distal anastomoses, accounts for more than 20% of late
failures of infrainguinal prosthetic graft revascularizations.5-8,13 On gross examination, inti-
mal hyperplasia is a white, firm, fibrous lesion.9,10 Histologically, it is characterized by cellu-
lar proliferation and accumulation of extracellular matrix material, occurring as a result of
excessive proliferation and accumulation of smooth muscle cells.11 The formation of
neointimal hyperplasia occurring after implantation of prosthetic vascular grafts is, how-
ever, not identical to intimal proliferation after endarterectomy or angioplasty, in that it is
primarily due to chronic inflammatory responses associated with implanted synthetic grafts.12
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
132 Tissue Engineering of Prosthetic Vascular Grafts
In contrast to occlusions after angioplasty or atherectomy, dothelium. One explanatory finding is that PDGF is also
which tend to appear in the first 6 months,14,15 the stenos- produced in endothelialized grafts.25 For weeks after graft
ing intimal lesions of autologous or prosthetic grafts usu- insertion in dogs, PDGF production of ePTFE grafts seeded
ally appear later.5,13 with autologous venous endothelial cells is even greater than
in nonseeded controls; it correlates positively with endo-
Pathogenesis of Anastomotic Hyperplasia thelial cell coverage and inversely with platelet deposition.25
The pathogenesis of anastomotic hyperplasia, which Although endothelial cells play an important role in
includes the interaction of the various cells and the mito- preventing or inhibiting neointimal hyperplasia by releas-
gens and cytokines they produce, and regulation of cellular ing inhibitors of smooth muscle cells, 35-37 incomplete
growth is not fully understood. Cell migration and prolif- endothelialization seen at the anastomotic areas in
eration during intimal thickening appear to be regulated by nonseeded grafts may induce smooth muscle cell prolifera-
factors from the blood, particularly platelets and leukocytes, tion beneath the endothelium38,39 due to platelet-derived
as well as by interaction with adjacent vascular wall cells.16 growth factor, and other mitogen production by perturbed
Operative manipulation and hemodynamic factors such as endothelial cells.40-42 In contrast to a vein graft, in a seeded
high and low flow velocities, high and low wall shear stress,17 prosthesis there is only a subendothelial layer, without the
and mechanical compliance mismatch 18 also influence medial layer. This might be a reason for early suppression of
smooth muscle cell proliferation and intimal thickening. A smooth muscle cell proliferation after completion of the
possible explanation for the smooth muscle cell prolifera- endothelial layer, which does not occur in vein bypass. The
tion beneath the pannus endothelium in prosthetic grafts is influence of complete endothelialization of a prosthetic graft
the production of smooth muscle cell mitogens by perturbed on the development of late neointimal hyperplasia is un-
anastomotic endothelial cells.19 These endothelial cells are known.
known to produce platelet-derived growth factor (PDGF) The complexity of the pathways leading to formation
and other mitogens.20,21 Furthermore, endothelial cells are of neointimal hyperplasia suggests that one agent alone will
not only actively involved in the regulation of smooth muscle likely not be entirely effective in its prevention. This multi-
cell migration and proliferation during pathologic changes factorial cause seems to be responsible for failed single medi-
such as intimal hyperplasia, but also during normal pro- cal therapies aiming at the prevention of anastomotic
cesses such as vessel development, depending on their state hyperplasia. By achieving a confluent endothelium in a
of activation. It is unknown whether endothelial cells on seeded graft, however, one possible factor for neointimal
implanted prosthetic grafts become permanently activated anastomotic hyperplasia—the nonendothelialized luminal
or if they return to the “normal” inactive state.19 It is, how- surface of the graft—might be excluded, or might be
ever, known that a recovered endothelial layer after arterial converted into an active factor for prevention of late
injury or after endothelial cell lining of a prosthetic graft neointimal hyperplasia.
only decreases or slows the process of intimal proliferation,
without preventing it completely. Experimental Studies with Endothelial Cell
Seeding at the University Hospital Zurich
Endothelial Cells and Neointimal Hyperplasia At the University Hospital Zurich we performed sev-
Endothelial cells possess both inhibitional and prolif- eral studies whose main aim was to test whether complete
erative properties with regard to smooth muscle cell prolif- endothelial-like cell coverage of a small vascular graft—
eration. On the one hand, the degree of myointimal hyper- seeded with omentally derived cells—is capable of reducing
plasia caused by arterial damage is less in areas which are or even preventing late neointimal proliferation and anas-
covered by regenerating endothelium.22,23 This may be due tomotic hyperplasia.43-46 In one study44 we performed long-
to synthesis of heparin proteoglycans.24 Neointimal smooth term tests for up to 1 year in 24 mongrel dogs (weight
muscle cells may even become nonproliferative when the 20-30 kg; age 1-4 years). The dogs were cared for according
overlying endothelial layer is re-established. Therefore, the to the European Convention on Animal Care. All experi-
confluent endothelial cells in a seeded graft may synthesize ments were reviewed and approved by the Ethics Commit-
active substances which inhibit proliferation of smooth tee of the University Hospital Zurich. Anesthesia was induced
muscle cells by releasing inhibitors of smooth muscle cells with sodium thiopental (20 mg/kg IV); the dogs were intu-
such as heparan sulfate,24,26 endothelium-derived relaxing bated and ventilated with an oxygen, nitrous oxide and ha-
factor (EDRF),27-30 and prostacyclin.31,32 Grafts, whether lothane mixture. Intraoperatively the animals received an
seeded or nonseeded, produce prostacyclin (PGI2) in small infusion of lactated Ringer’s solution at the rate of 10 ml/kg
amounts,28,30,33 but seeded grafts have a significantly higher per h during the operation.
production of prostacyclin than the nonseeded ones; it is,
nevertheless, lower than that of the native arteries.27-30 In an Surgical Technique
in vitro model of vascular injury, endothelial cell seeding A small supraumbilical laparotomy was made, and as
significantly increases both basal and stimulated PGI2 release much of the omentum as possible was removed and placed
from damaged vein segments.34 On the other hand, incom- in a sterile 150 ml plate containing 60 ml of HEPES buff-
plete late suppression of smooth muscle cell proliferation ered saline. The omentum was further processed in the cell
may cause late intimal and medial thickening at anastomotic laboratory by the cell-laboratory staff, while the surgical staff
areas despite regeneration of a morphologically intact en- closed the abdominal incision and performed exposure of
Neointimal Hyperplasia in Small Diameter Prosthetic Vascular Grafts 133
the arteries for the implantation. Starch-free gloves were used Cell Identification
throughout the study to avoid the cytotoxic effect of glove An aliquot of approximately 0.5 ml of the cell suspen-
powder.47 A midline neck incision was made and both com- sion was removed for quantification of harvested cells. Cell
mon carotid arteries were dissected free. A 6 cm segment of numbers were assessed by fluorometric deoxyribonucleic
each artery was isolated between the vascular bulldog clamps acid measurements of cell pellets.50 Cell identification re-
and excised, and then both ends were beveled to approxi- lied on specific perinuclear uptake of acetylated low density
mately 45 degrees. A 4 mm internal diameter uncrimped lipoprotein,51 immunohistochemical staining for von
Dacron graft (Sulzer Brother, Ltd., Winterthur, Switzerland) Willebrand factor52 and the determination of cell viability
with an average water porosity of 700 ml/min per cm2 at in culture.53,54 Viability was defined as the number of at-
120 mmHg was simultaneously preclotted with autologous tached cells counted after 18 h in the cell culture, and was
blood and seeded. Immediately thereafter they were im- attributed to those cells which resisted rinsing with PBS. After
planted into the right carotid artery. The oblique end-to- washing, the cell culture was trypsinized and the number of
end anastomoses were performed with a continuous 7-0 cells was calculated by fluorometric deoxyribonucleic acid
polypropylene suture. Paired anatomic sites (the common measurements on cell pellets.50 The density of viable cells
carotid arteries) were used throughout the study; each dog per cm2 of a seeded graft was estimated by the ratio of the
received one seeded (inserted into the right carotid artery) number of viable cells to the seeded graft surface.
and one nonseeded graft (inserted into the left carotid
artery). Graft Surveillance and Explantation
Graft patency was assessed by angiography at 1, 5, 12,
Medication 26, and 52 weeks after surgery. The angiographic catheter
All animals received prophylactic cephalosporine an- was introduced through the common femoral artery and
tibiotics (cefalexinum monohydratum, 1200 mg/day) and than forwarded into the common carotid artery. The
analgesics (buprenorphine chloride 0.9 mg/day) for 4 days angiographic examination was performed by manual injec-
postoperatively, as well as dipyridamole (Dip, 75 mg/day) tion of 10 ml of diluted (1:1 with 0.9% sodium chloride)
and acetylsalicylic acid (ASA, 325 mg/day) orally, beginning nonionic angiographic dye (Iopromidum Ultravist 370,
on 1 and 4 days before surgery, respectively. The antiplatelet Schering, Berlin, Germany).
medication (ASA) was continued for 4 weeks postoperatively. The prostheses were explanted after 5 weeks (n=6
dogs), 12 weeks (n=6 dogs), 26 weeks (n=6 dogs) and 52
Microvascular Endothelial Cell Harvesting weeks (n=6 dogs). After transfemoral angiography, the grafts
The cell harvesting procedure followed the modified were dissected free and then removed with the adjacent 3 cm
method of Schmidt et al.48 The omentum (28.8 g ± 7.3 g of proximal and distal carotid artery. Grafts were histologi-
SD) was rinsed with HBS-2, trimmed, minced with two scal- cally investigated with light microscopy after staining with
pels into very small pieces, and aspirated into a 10 ml pi- hematoxylin-eosin. For scanning electron microscopy, grafts
pette; the tissue was then transferred into a 50 ml Erlenmayer were processed according to the method of Schrotter.55 Im-
flask containing 1500 U/ml collagenase type II (Sigma munohistochemical staining for von Willebrand factor was
Chemical Co., St. Louis, MO, USA). The ratio was 1 g of performed on formalin-fixed paraffin embedded tissue. Sec-
omental tissue to 2 ml of collagenase. The suspension of tions were predigested for 10 min with 0.1% pronase E
omental tissue and collagenase was incubated for 40 min- (Merck, Dreieich, Germany) at 37°C. After blocking endog-
utes in a 37°C shaking water bath at 100 rotations per minute. enous endoperoxidase with methanol-H2O2, rabbit antise-
The digested tissue was homogenized by repetitive pipetting, rum against von Willebrand factor (factor VIII-related an-
transferred into 15 ml tube, and centrifuged twice at 100G tigen) was applied. Rabbit antibodies were detected using
for 5 minutes. The supernatant contained mainly adipocytes the ABC/peroxidase method with diaminobenzidine as a
and the collagenase solution. The cell pellet was resuspended substrate of the color reaction.52 Sections were counter-
in 10 ml PBS, filtered through a 250 micron pore size poly- stained with hemalum. (All immunological reagents were
ester mesh (Bolting Cloth Weaving, Zurich, Switzerland) to from DAKOA/S, Glostrup, Denmark). The maximum thick-
remove nondigested large tissue fragments, and then washed ness of the neointimal tissue within 10 mm of the suture
twice with HBS-2. Finally, a suspension of the endothelial lines, as well as of the central part of the graft, were mea-
cells (2 ml) was added to 15 ml of autologous plasma and sured on hematoxylin-eosin stained cross section by means
centrifuged at 45G for 20 minutes onto the prosthesis using of a computer-assisted automated image analysis system
a rotation device (Sulzer Brothers, Ltd., Winterthur, Swit- (Video planimeter, Zeiss Inc., Switzerland).
zerland). After implantation, the seeded grafts demonstrated Late neointimal hyperplasia was assumed if a signifi-
clusters of cells adhering to the Dacron fibers on the pros- cant difference in neointimal thickness was found when
thetic lumen embedded in red thrombus, preventing the comparing:
wash-out and subsequent loss of the seeded cells.49 Cell seed- 1. The mean neointimal thickness of certain parts of
ing density was 1.75 (0.77 to 2.72, 95% confidence inter- the grafts with the mean neointimal thicknesses of
vals) x 106 cells per square centimeter of graft surface, rang- the other parts at the same time point (e.g., the mean
ing from 0.37 x 106 to 4.54 x 106 cells. value of the neointimal thicknesses of all the distal
parts of all grafts explanted at three months after
134 Tissue Engineering of Prosthetic Vascular Grafts
implantation vs. the mean value of the proximal or 100%, 86%, 49%, 40%, and 13% for nonseeded grafts, re-
central parts of the same group of grafts, etc.); or spectively (Fig. 11.1).
2. The difference between the mean neointimal thick-
nesses of the same parts of the grafts at different time Angiographic Differences of Seeded and Nonseeded
points (e.g., the mean value of the neointimal thick- Grafts
nesses of all the distal parts of all grafts explanted at We found obvious angiographic and histological dif-
three months after implantation vs. the mean value ferences between the seeded and the nonseeded grafts with
of the neointimal thicknesses of all the distal parts regard to neointimal hyperplasia, except when the nonseeded
of all grafts explanted at six months after implanta- grafts were also completely covered with endothelial-like
tion, etc.). cells. According to the fine structural details of the grafts
seen on angiography and later confirmed by histological
Statistics examination, thrombotic processes and neointimal hyper-
Patency data determined by serial angiographic stud- plasia were confirmed as the two main causes of graft occlu-
ies were tabulated and presented in the life table format by sion. Early graft occlusions were obviously caused by throm-
the method of Cutler and Ederer.56 Event-free proportions botic processes, because these grafts showed smooth anas-
at 1, 5, 12, 26, and 52 weeks postoperatively are reported tomotic regions with intraluminal clots on angiography.
with 95% confidence limits. The data are presented as means Contrary to these early findings, progression of late
with 95% nonparametric confidence intervals. Differences neointimal proliferation, predominantly in the anastomotic
in mean values for neointimal thicknesses at different time regions, was the dominant finding in grafts with late occlu-
points and between different locations were assessed by re- sion. The first changes in terms of neointimal hyperplasia
peated ANOVA measures; simple regression was used to were noted by angiographic studies performed at 3 months
evaluate the development of the neointimal thickness dur- after graft insertion, with a trend to progression with time.
ing the time at the same location. A value of p < 0.05 was However, this anastomotic stenosis was not observed in the
considered to be significant. two nonseeded grafts which were totally covered with en-
dothelial-like cells.
Endothelial Cells Reduce Late Neointimal
Proliferation Histological Findings
Based on the combination of three highly conclusive Histological studies of the seeded grafts revealed a
parameters (patency rate, angiographic findings, histologi- neointima with luminal endothelial-like lining and highly
cal examinations), our study strongly supported the hypoth- organized subendothelial layers in the absence of anasto-
esis that complete coverage with endothelial-like cells of the motic neointimal hyperplasia (Fig. 11.2). The subintimal
luminal side of a small vascular graft seeded with omentally- tissue was covered by a luminal monolayer of coherent epi-
derived cells reduces or even prevents late neointimal pro- thelial-like, flat cells expressing von Willebrand factor
liferation and anastomotic hyperplasia. (Fig. 11.3). Microscopic examinations confirmed that
neointimal hyperplasia had caused the anastomotic steno-
Improved Patency of Seeded Grafts sis seen by angiography. Furthermore, histological exami-
The actuarial patency rates at 1, 5, 12, 26, and 52 weeks nations revealed that the stenotic lesions seen on angiogra-
were 100%, 95%, 95%, 95%, and 95% for seeded grafts, and phy in the central parts were predominantly caused by a
thrombotic process, although it was difficult to distinguish
hyperplasia from organized thrombus in some specimens large areas covered with coagulum consisting of fibrin, plate-
of the occluded nonseeded grafts. lets, and occasional leukocytes interspersed with regions of
spontaneous coverage by endothelial-like cells.
Spontaneous Endothelialization
In contrast to the confluent endothelial lining of Neointimal Tissue Thicknesses
seeded grafts, the nonseeded grafts showed a varying degree In endothelial seeded grafts, the measurements of the
of spontaneous coverage with endothelial-like cells maximum thicknesses of the neointimal tissue at the anas-
(Fig. 11.4). The extent of this process was time dependent; tomoses as well as in the central parts of the seeded small
two patent nonseeded grafts, for instance, were completely diameter Dacron grafts showed neither late neointimal pro-
covered by endothelial-like cells after one year. However, liferation nor anastomotic hyperplasia. The mean neointimal
these grafts did not show any neointimal hyperplasia, and tissue thickness of the seeded grafts increased during the
in that resembled seeded grafts. In the other patent initial 26 weeks and then stabilized or slightly regressed af-
nonseeded grafts, scanning electron microscopy revealed ter this period (Fig. 11.5). Simple regression analysis showed
136 Tissue Engineering of Prosthetic Vascular Grafts
progressive reduction of the mean neointimal thickness raphy for routine long-term surveillance, because this type
(-0.5 mm per week). However, this tissue regression was sta- of angiography provided fine anatomical details enabling
tistically not significant. There was no difference when com- us to identify structural changes in grafts.5 Angiograms of
paring any specific part among the different grafts at any the seeded grafts showed a smooth intimal surface without
time point, nor in comparison with two nonseeded grafts luminal narrowing, whereas angiographic examinations of
which were totally covered with endothelial-like cells. most nonseeded grafts demonstrated different degrees of
Repeated ANOVA measures revealed no significant differ- luminal stenosis at the anastomotic regions in the late phase
ence among mean neointimal thicknesses of seeded grafts of implantation.
at different time points (p=0.51) or different locations Thus, the main factors for occlusion of prosthetic small
(Greenhouse-Geisser corrected p=0.16). diameter vascular grafts are increased graft surface
thrombogenicity and technical error in the early postopera-
Graft Occlusion tive course, and progression of the atherosclerotic disease
In summary, both the angiographic studies and his- and neointimal hyperplasia in the late phase.57 In our com-
tological examinations revealed that neointimal prolifera- parative study, technical errors such as incorrectly performed
tion and anastomotic hyperplasia were the main factors lead- anastomoses were excluded by postoperative angiography;
ing to late occlusion of the nonseeded grafts. To obtain fine atherosclerotic process as a possible cause for late occlusion
assessment of our implants, we used conventional angiog- was also excluded by the use of young animals. Therefore,
thrombotic graft occlusion was found to be the main cause endothelial-like cells of a synthetic vascular graft following
of early failure. In all explants, the cause of occlusion was seeding with pure endothelial cells or with a mixture of dif-
determined after considering the results of all diagnostic ferent cells from omentum is an unknown process, too. It
methods (angiographic, histological and macroscopic find- probably requires cell duplication, although it has been
ings), never relying on one single method only. Since grafts shown that endothelial cells remain on the graft surface af-
were explanted at the dates assigned by preoperative ran- ter cell transplantation.59 Due to the similarity of the scan-
domization and not immediately after occlusion was noted, ning microscopic findings between the seeded grafts and the
it was sometimes difficult to distinguish hyperplasia from spontaneously endothelialized regions in nonseeded grafts,
organized thrombus in occluded control grafts. Once throm- it can be speculated that these two different processes prob-
bosis of a graft has occurred, the thrombus rapidly becomes ably involve similar mechanisms leading to surface cover-
organized and fibrotic, such that it is difficult to distinguish age with endothelial-like cells and ultimately to a
from pre-existing hyperplasia. The technique of quantita- thrombogenicity of the graft surface.
tive histologic analysis of neointimal hyperplasia used in our
study permits accurate measurement only in grafts that are Intimal and Neointimal Hyperplasia
patent, so that progression can be observed. However, the It should be emphasized that the term “late neointimal
progressive course of hyperplasia can only be compared be- hyperplasia” does not refer to the absence of any neointimal
tween different animals. Therefore, we performed the two tissue on the luminal side of a seeded prosthetic vascular
types of comparisons described above: firstly, simultaneous graft. It is a well established fact that seeding with omental
measurements and comparisons of the different locations cells produces a neointima on these grafts. Therefore, we
at the same time point and secondly, measurement and com- defined the term “late neointimal hyperplasia” in our study
parisons of the same locations on different specimens at dif- as a significant increase of the neointimal thickness in cer-
ferent times. tain parts of the seeded grafts (proximal, central, distal) as
compared with the other parts at the same time point, or
The Role of Complete Endothelial Coverage the difference between the neointimal thicknesses of the
The second important observation of our study is that same parts of the grafts at different points in time.
complete coverage of the prosthetic surface with endothe- Neointimal hyperplasia in a nonseeded graft is a pro-
lial-like cells per se, regardless of by which method this has cess of smooth muscle cell proliferation which occurs at both
been achieved, was the single most important factor for pre- the proximal and distal anastomoses between a prosthetic
vention of late neointimal hyperplasia and the subsequent nonseeded graft and the adjacent artery. However, it occurs
improvement of graft patency. This conclusion is further to a significantly greater degree at the distal anastomosis than
supported by the fact that late neointimal hyperplasia was the proximal one.60 In our study, we found no difference in
found in nonseeded grafts which were only partly covered neointimal thickness between any parts (proximal, distal,
by endothelial-like cells, but not found in the nonseeded or central) of the seeded grafts at any time point. This makes
grafts which were totally covered with endothelial-like cells late neointimal hyperplasia an unlikely event in seeded pros-
through spontaneous outgrowth. theses because one would expect:
We further conclude that an omental seeding tech- 1. Changing intimal thickness in predefined graft ar-
nique may reduce or prevent late neointimal hyperplasia via eas over time; and
the mechanism of complete luminal coverage with endo- 2. Differences in intimal thickness within one and the
thelial-like cells. However, it should be emphasized that the same graft at the same time point, particularly when
improved overall patency rate of the seeded grafts can only comparing anastomotic regions with midgraft areas.
be partly attributed to the reduction or prevention of late The regular angiographic follow-up and microscopic
neointimal hyperplasia, while other mechanisms such as examination revealed that there was a significant difference
reduced thrombogenicity due to luminal surface coverage between the seeded and the nonseeded grafts with reference
with endothelial-like cells also contribute substantially. to neointimal hyperplasia. The fact that no difference be-
The third important observation of our study is that tween the neointimal thicknesses was found when compar-
we found no scanning electron microscopic differences be- ing the two nonseeded grafts which were patent at 1 year
tween seeded grafts and the regions of the nonseeded grafts (and were totally covered with endothelial-like cells) and the
which were covered with endothelial-like cells. Although seeded grafts does not speak against the hypothesis but rather
spontaneous endothelialization of nonseeded grafts is usu- sheds more light on the impact of complete endotheliali-
ally seen in animals, the precise origin of luminal endothe- zation on neointimal proliferation.
lial-like cells in both endothelialized seeded and nonseeded Intimal hyperplasia (intimal changes in a venous or
grafts remains unclear. The findings of endothelialized anas- arterial graft or native artery) and neointimal hyperplasia
tomotic regions as well as islands of endothelium through- (de novo generation of an intima on a synthetic graft sur-
out the grafts indicate that spontaneous endothelialization face which was previously lacking an intima) are a charac-
can occur by several mechanisms, such as migration of en- teristic fibromuscular cellular response to vascular injury
dothelial cells from the host artery towards the prosthesis, during vascular reconstruction.61 It is characterized by cel-
endothelial cell ingrowth from capillaries originating out- lular proliferation and accumulation of extracellular matrix
side the graft and passing through the interstices of the fab- material which occur as a result of excessive proliferation of
ric, or from circulating blood cells.58 However, coverage with smooth muscle cells.62 All forms of vascular reconstruction
138 Tissue Engineering of Prosthetic Vascular Grafts
cause injury and a wound healing response,4 leading to inti- binding of growth factors can trigger cytoplasmic signal
mal or neointimal proliferation after reconstruction with transduction pathways.77 These regulatory mechanisms of
vein or prosthetic grafts63,64 as well as in a host artery after specific cell behavior are not well understood. Thus, analy-
endarterectomy, atherectomy, or coronary angioplasty.65-68 sis of biological activity of growth factors on smooth muscle
However, formation of neointimal hyperplasia occurring cell growth might be useful for antagonizing pathological
after implantation of prosthetic vascular grafts is not smooth muscle cell migration and proliferation.75,76,82
identical to intimal proliferation after endarterectomy or an- The complexity of the pathways leading to formation
gioplasty, largely due to chronic inflammatory responses of neointimal hyperplasia suggests that one agent alone will
associated with implanted synthetic grafts. Early after im- likely not be entirely effective for its prevention. This multi-
plantation of a prosthetic graft, leukocytes adhere to the graft factorial cause is responsible for failed single medical thera-
surface, followed by monocyte infiltration and formation pies for preventing anastomotic hyperplasia. Since growth
of foreign body giant cells. The latter characterize the chronic factors are the main stimuli for smooth muscle cell migra-
inflammatory or foreign body response to vascular prosthe- tion and proliferation, several pharmacological approaches
ses.69 Neointimal hyperplasia in a synthetic graft had already to smooth muscle cell hyperplasia might be useful.83 Differ-
been recognized in the early phase of peripheral vascular ent agents, such as anti-inflammatories,84 antihypertensive
surgery.8,70 It occurs both in Dacron and polytetrafluoro- drugs,85,86 fish oil,87 prostaglandin analogues,88 #1-adrener-
ethylene grafts71,72 as early as 3 months and as late as 1 year gic blocking agents,89 and anticoagulants90 have been shown
after implantation.7,8,70 In contrast to occlusions after to suppress the process of intimal hyperplasia in animal stud-
angioplasty or atherectomy, which tend to appear in the first ies. The role of anti-platelet therapy in the prevention of
6 months,73 the stenosing intimal lesions of autologous or intimal hyperplasia after arterial reconstruction remains
prosthetic grafts usually appear later after surgery.5,6 Anas- controversial.91,92 Human trials have demonstrated that as-
tomotic hyperplasia involves both the proximal and distal pirin and dipyridamole significantly improve patency of
anastomoses but it has a predilection for the distal anasto- prosthetic, but not saphenous vein, femoropopliteal by-
mosis, with a tendency to progress with time.72 Neointimal passes;93,94 they are particularly beneficial when started be-
hyperplasia in the area of the distal anastomoses accounts fore operation94 or within 24 h of operation.95 However,
for 20%-50% of late lower extremity bypass graft failures.6-8 clinical trials of anti-platelet agents suggest that platelet in-
The regulation of cell migration, proliferation, and hibition alone does not prevent anastomotic intimal
growth during intimal thickening appear to be regulated by hyperplasia.96
factors from the blood, particularly platelets and leukocytes, The present study showed that establishing an endo-
and the interaction of vascular wall cells and grafts them- thelial-like cell coverage on the luminal surface of small di-
selves.16 Operative manipulation and hemodynamic factors ameter prosthetic grafts plays an important role in reduc-
such as high and low flow velocities, high and low wall shear tion or prevention of late neointimal and anastomotic hy-
stress,17 as well as compliance and diameter mismatch be- perplasia. Anastomotic neointimal hyperplasia was sup-
tween graft and host artery,18 also influence smooth muscle pressed in those grafts that were totally covered with endo-
cell proliferation and intimal thickening. thelial-like cells, regardless of whether they had been seeded
It has been proposed that growth factors such as plate- or not. In contrast, occluded control grafts showed clearly
let-derived growth factor and basic fibroblast growth factor visible anastomotic tissue ingrowth in the late phase. There-
released from activated platelets, leukocytes, altered smooth fore, seeding with omental cells may play an important role
muscle cells and endothelial cells may be involved in the in preventing or inhibiting late smooth muscle cell prolif-
smooth muscle cell migration and proliferation after vascu- eration by total coverage of the prosthetic luminal surface
lar injury.74,75 Basic fibroblast growth factor is a prototype with endothelial-like cells. The characteristic of endothelial
of the family of heparin-binding growth factors that regu- cells to possess both inhibitory and proliferative properties
late a variety of cellular responses including cell growth, on smooth muscle cells may thus play an important role in
morphogenesis and differentiation of various cell types, in- pathogenesis of neointimal hyperplasia.97,98 A possible ex-
cluding smooth muscle cells.76 Fibroblast growth factors planation for the smooth muscle cell proliferation beneath
deliver their signals to cells by binding to a dual receptor endothelium in the nonseeded grafts is the production of
system which activates the bound growth factor before its smooth muscle cell mitogens by perturbed endothelial
delivery to the signal-transducer receptors.77,78 Signaling of cells.69 By reaching early luminal confluence of endothelial-
growth and differentiation involves multiple pathways.79 At like cells in a seeded graft, one possible factor for neointimal
least two families of receptors bind basic fibroblast growth anastomotic hyperplasia, namely nonendothelialized lumi-
factor and mediate its response: tyrosine kinase-containing nal surface of the graft, might be excluded, or might be con-
fibroblast growth factor receptors,77 and heparan sulfate verted into an active factor for prevention of late neointimal
proteoglycans.80 Both are known to undergo internalization hyperplasia. Although endothelialized surfaces will not al-
by different pathways.79 Heparan sulfate proteoglycans are ter all factors potentially contributing to anastomotic inti-
obligate partners in binding of basic fibroblast growth fac- mal hyperplasia, they seem to reduce the deposition and
tors to their receptors; fibroblast growth factors do not bind activation of circulating blood elements, including platelets,
to fibroblast growth factor receptors unless heparan sulfate leukocytes, and components of the coagulation and comple-
or its analog, heparin, is present.81 The activation of intrin- ment system. They furthermore seem to decrease the release
sic tyrosine kinase function of receptor tyrosine kinases upon of mitogen factors for smooth muscle cells and prevent
Neointimal Hyperplasia in Small Diameter Prosthetic Vascular Grafts 139
thrombus formation by generation of prostacyclin, immunostaining seen in some of our specimens in the up-
antithrombotic factors and plasminogen activators. per layers of the intima could be caused by previous platelet
deposition and therefore previous thrombosis, which was
Omental Microvascular Cells supposedly prevented by the seeding with omental cells. The
A large number of cells can be harvested from the other methods for identification of these cells, and their dif-
omentum. The cells derived from omental suspension com- ferentiation from mesothelial cells, are the use of endothe-
prise a mixture of microvascular endothelial cells and me- lial cell-specific monoclonal antibodies, such as antibody
sothelial cells, with a few other cell types.48,99 Overall, the BMA 120,113 and the characterization of different biologi-
contamination of a seeding suspension with nonendothelial cal functions typical for these cells.112 Moreover, endothe-
cells depends on the harvesting method. A purer endothe- lial cells express thrombomodulin, a cofactor for activation
lial cell yield can be achieved by lowering the separation den- of protein c, intracellularly; thus they stimulate the activa-
sity during the harvesting procedure, but this leads to a re- tion of protein c by thrombin. 114,115 Furthermore,
duced number of all cells available for seeding. Wang et al100 thrombomodulin has been identified immunologically on
estimated that 10% or less of the total cell number of har- endothelial cells of veins, arteries, and capillaries as well as
vested omentum cells were nonendothelial in origin. Kern on the endothelium of lymphatics and on syncytio-
et al101 obtained a completely pure microvascular endothe- trophoblasts.116
lial cell population harvested from omental tissue with no Comparing cells derived from human omental fat tis-
growth of other cell types in serially passaged cultures, but sue with those of human umbilical vein endothelial cells,
the endothelial cell yield was greatly reduced, at only 103 Visser et al103 concluded that the cells from omental tissue
endothelial cells per gram of omental tissue. were not endothelial but mesothelial in nature. Their state-
ment was based on the observations that cells isolated from
Origin of the Omental Cells human omental tissue did not stain with endothelial spe-
The identification of the cells derived from omentum cific antibodies EN4 and PAL-E; these cells contained abun-
poses a problem and there is no consensus in the literature dant cytokeratins 8 and 18, which were absent in endothe-
regarding their identity and origin.102,103 It is sometimes dif- lial cells.107 The presence of cytokeratins 8 and 18, as deter-
ficult to establish the identity of isolated endothelial cells, mined with the monoclonal antibodies M20 and M9, was
especially their distinction from mesothelial cells,104 because abundant in the cells derived from omentum but not in the
both types of cells are present in omental tissue. Like endo- human umbilical vein endothelial cells.103 Vimenten could
thelial cells, mesothelial cells produce prostacyclin105,106 and be detected (monoclonal antibody V9) in both types of cells,
possess fibrinolytic activity. These cells produce large whereas desmin (monoclonal antibody D33) was present
amounts of tissue-type plasminogen activator in vitro, to- only in omentally derived cells. A faint and diffuse staining
gether with type 1 and 2 plasminogen activator inhibitor.107 of von Willebrand factor was seen in cells from omentum,
Moreover, when seeded onto vascular prostheses these cells whereas microvascular endothelial cells from subcutaneous
acquire a confluent monolayer lining with no adherent plate- fat displayed this factor as indistinct granular structures.107
lets or amorphous material within one month after sur- In contrast to the human umbilical vein endothelial cells,
gery.108,109 In tissue culture, both types of cells show similar which stained with both anti-von Willebrand factor anti-
growth patterns under light microscopy.102,104,105 However, bodies, omentally derived cells stained for von Willebrand
it has been shown that in a culture of mixed cells derived factor only if the antibodies were polyclonal. Moreover, scan-
from omentum, endothelial cells were rapidly displaced by ning electron microscopy revealed that cultured cells derived
mesothelial cells, resulting in a pure culture of mesothelial from omentum contain numerous surface microvilli,
cells.103 Similarly, in a tissue culture of endothelial cells and whereas human umbilical vein endothelial cells did not. All
fibroblasts, endothelial cells are suppressed by fibroblasts that of these findings suggest that the cells derived from omen-
proliferate to form a confluent layer.110 In our study, we did tum were a mixture of predominantly mesothelial cells and
not seed the grafts either with omentum cells immediately a low number of microvascular endothelial cells.103 In con-
after harvesting or with cultured cells. In this way, the sup- trast to these findings, Stansby et al117 were not able to prove
pressive effect of mesothelial cells on endothelial cells in that the omental cells were mesothelial in origin. They con-
culture could be excluded in our study. cluded that human omental microvascular endothelial cells
Although the origin of the omentally derived cells has were pericytic in nature.117 Hernando et al105 proved the
remained controversial, its endothelial nature has usually nonendothelial origin of the cells derived from human
been proven by morphological and functional criteria. The omentum, demonstrating that these cells showed positivity
most widely methods used for identification of endothelial for monoclonal antibodies specific for endothelial cells (anti-
cells are the demonstration of von Willebrand factor by im- CD34 QBEND10), antibodies to intermediate filaments
munofluorescence staining,111 and the uptake of diacetylated (anti-vimentin and anti-desmin) and anti-smooth muscle
low-density lipoprotein. 51 The incorporation of cell antibodies (anti-actin and anti-total actin).
[35S]methionine into von Willebrand factor demonstrates Endothelial cells from different organs show various
the ability of the cultured microvascular endothelial cells differences in tissue culture and in vivo; human endothelial
from human omental tissue to synthesize von Willebrand cells harvested from saphenous or umbilical vein, for in-
factor,112 proving the identity of the cultured cells as endo- stance, exhibit distinctive features in transmission electron
thelial. However, it could be speculated that the extensive microscopic examination if cultivated on precoated PTFE
140 Tissue Engineering of Prosthetic Vascular Grafts
20. DiCorleto PE, Bowen-Pope DF. Cultured endothelial cells of smooth muscle cell growth. J Cell Biol 1981;
produce a platelet-derived growth factor-like protein. Proc 90:372-378.
Natl Acad Sci USA 1983; 80:1919-1923. 37. Jensen N, Brunkwall J, Fält K, Lindblat B, Bergquist D.
21. Miossec P, Cavender D, Ziff M. Production of interleukin Prostacyclin is produced from endothelial cell-seeded
1 by human endothelial cells. J Immunol 1986; grafts: An experimental study in sheep. Eur J Vasc Surg
136:2486-2491. 1992; 6:499-504.
22. Bjorkerud S, Bondjers G. Arterial repair and atheroscle- 38. Angelini GD, Christie MI, Bryan AJ, Lewis MJ. Surgical
rosis after mechanical injury. Part 5. Tissue response af- preparation impairs release of endothelium derived relax-
ter induction of a large superfitial transverse injury. Ath- ing factor from human saphenous vein. Ann Thorac Surg
erosclerosis 1973; 18:235-255. 1989; 48:417-420.
23. Handenschild CC, Schwartz SM. Endothelial regeneration 39. Graham LM, Fox PL. Growth factor production follow-
II. Restoration of endothelial continuity. Lab Invest 1979; ing prosthetic graft implantation. J Vasc Surg 1991;
41:407-418. 13:742-744.
24. Castellot JJ, Addonizio ML, Rosenberg R, Karnovsky MJ. 40. Miossec P, Cavender D, Ziff M. Production of interleukin
Cultured endothelial cells produce heparinlike inhibitor 1 by human endothelial cells. J Immunol 1986;
of smooth muscle cell growth. J Cell Bil 1981; 90:372-378. 136:2486-2491.
25. Kaufman BR, Fox PL, Graham LM. Platelet-derived 41. DiCorleto PE, Bowen-Pope DF. Cultured endothelial cells
growth factor production by canine aortic grafts seeded produce a platelet-derived growth factor-like protein. Proc
with endothelial cells. J Vasc Surg 1992; 15:699-707. Natl Acad Sci USA 1983; 80:1919-1923.
26. Fritze LMS, Reilly CF, Rosenberg RD. An antiproliferative 42. Kaufman BR, Fox PL, Graham LM. Platelet-derived
heparan sulfate species produced by postconfluent smooth growth factor production by canine aortic grafts seeded
muscle cells. J Cell Biol 1985; 100:1041-1049. with endothelial cells. J Vasc Surg 1992; 15:699-707.
27. Schmidt SP, Hunter TJ, Falkow LJ, Evancho MM, Sharp 43. Pasic M, W. Müller-Glauser, Lachat M, Bittmann P, von
WV. Effects of antiplatelet agent on PGI2 production by Segesser L, Turina M. Langzeitresultate in der
endothelial-cell-seeded small diameter Dacron® grafts. Hunderkarotis mit kleinlumigen Gefässprothesen mit
ASAIOJ 1985; 8:99-103. mikrovaskulären Endothelzellen. Helv chir Acta 1993;
28. Boyd KL, Schmidt S, Pippert T, Hite S, Sharp W. The 60:381-385.
effects of pore size and endothelial cell seeding upon per- 44. Pasic M, Müller-Glauser W, Odermatt B, Lachat M, Seifert
formance of small-diameter ePTFE vascular grafts under B, Turina M. Superior late patency of small-diameter
controlled flow conditions. J Biomed Mater Res 1988; Dacron grafts seeding with omental cells prevents late
22:163-177. neointimal hyperplasia in small-diameter dacron grafts.
29. Sterpetti AV, Hunter WJ, Schultz RD, Sugimoto JT, Blair Circulation 1995; 92:2605-2616.
EA, Hacker K, Chasan P, Valentine J. Seeding with en- 45. Pasic M, Müller-Glauser W, von Segesser L, Odermatt B,
dothelial cells derived from the microvessels of the omen- Lachat M, Turina M. Endothelial cell seeding improves
tum and from the jugular vein: A comparative study. J patency of synthetic vascular grafts: Manual versus au-
Vasc Surg 1988; 7:677-684. tomatized method. Eur J Cardio-thorac Surg 1996;
30. Jensen N, Brunkwall J, Fält K, Lindblat B, Bergquist D. 10:372-379.
Prostacyclin is produced from endothelial cell-seeded 46. Pasic M, Müller-Glauser W, von Segesser L, Lachat M,
grafts: An experimental study in sheep. Eur J Vasc Surg Mihaljevic T, Turina M. Superior late patency of small-
1992; 6:499-504. diameter Dacron grafts seeded with omental microvascu-
31. Whitehouse WM Jr, Wakefield TW, Vinter DW, Ford JW, lar cells: An experimental study. Ann Thorac Surg 1994;
Swanson DP, Thrall JH, Froelich JW, Brown LE, Burkel 58:677-684.
WE, Graham LM, Stanley JC. Indium-111-oxine labeled 47. Sharefkin JB, Fairchild KD, Albus RA, Cruess DF, Rich
platelet imaging of endothelial seeded Dacron NM. The cytotoxic effect of surgical glove powder par-
thoracoabdominal vascular prostheses in a canine model. ticles on adult human vascular endothelial cell cultures:
Trans Am Soc Artif Intern Organs 1983; 29:183-187. implications for clinical uses of tissue culture techniques.
32. Clagett GP, Burkel WE, Sharefkin JB, Ford JW, Hufnagel J Surg Res 1986; 41:463-472.
H, Vinter DW, Kahn RH, Graham LM, Stanley JC, 48. Schmidt SP, Monajjem N, Evancho MM, Pippert TR,
Ramwell PM. Platelet reactivity in vivo in dogs with arte- Sharp WV. Microvascular endothelial cell seeding of
rial prostheses seeded with endothelial cells. Circulation small-diameter Dacron vascular grafts. J Invest Surg 1988;
1984; 69:632-639. 1:35-44.
33. Köveker GB, Burkel WE, Graham LM, Wakefield TW, 49. Pasic M. Endothelial-cell seeding with microvascular
Stanley JC. Endothelial cell seeding of expanded omental cells onto small-diameter prosthetic vascular
polytetrafluoroethylene vena cava conduits: Effects on lu- grafts in a canine model. Habilitation. Zurich, Switzer-
minal production of prostacyclin, platelet adherence, and land: The University of Zurich; 1994:79-82.
fibrinogen accumulation. J Vasc Surg 1988; 7:600-605. 50. Anderson JS, Price TM, Hanson SR, Harker LA. In vitro
34. Thompson MM, Budd JS, Eady SL, Allen KE, James M, endothelialization of small-caliber vascular grafts. Surgery
James RFL, Bell PRF. Endothelial cell seeding of damaged 1987; 101:577-586.
native vascular surfaces: Prostacyclin production. Eur J 51. Vojta JC, Via DP, Butterfield CE, Zetter BR. Identifica-
Vasc Surg 1992; 6:487-493. tion and isolation of endothelial cells based on their in-
35. Fritze LMS, Reilly CF, Rosenberg RD. An antiproliferative creased uptake of acetylated-low density lipoprotein. J Cell
heparan sulfate species produced by postconfluent smooth Biol 1984; 99:2034-2040.
muscle cells. J Cell Biol 1985; 100:1041-1049. 52. Hsu SU, Raine L, Fanger H. Use of avidin-biotin-peroxi-
36. Castellot JJ, Addonizio ML, Rosenberg R, Karnovsky MJ. dase complex (ABC) in immunoperoxidase techniques: A
Cultured endothelial cells produce heparinlike inhibitor
142 Tissue Engineering of Prosthetic Vascular Grafts
comparison between ABC and unlabeled antibody (PAP) ing distal anastomotic intimal hyperplasia of 4-mm-I.D.
procedures. J Histochem Cytochem 1981; 29:577-580. Dacron grafts in a canine model. J Biomed Mater Res
53. Müller-Glauser W, Bay U, Lehmann KH, Turina M. An 1991; 25:525-533.
improved procedure for enzymatic harvesting of highly 70. Szilagyi DE, Smith RF, Elliott JP, Allen HM. Long-term
purified canine venous endothelial cells for experimental behavior of a dacron arterial substitute: Clinical, roent-
small diameter vascular prostheses. Ann Vasc Surg 1989; genologic and histologic correlations. Ann Surg 1965;
3:134-139. 162:453-477.
54. Zilla P, Siedler S, Fasol R, Sharefkin JB. Reduced repro- 71. Selman SH, Rhodes RS, Anderson JM, DePalma RG,
ductive capacity of freshly harvested endothelial cells in Clowes AW. Atheromatous changes in expanded
smokers: A possible shortcoming in the success of seed- polytetrafluoroethylene grafts. Surgery 1980; 87:630-637.
ing. J Vasc Surg 1989; 10:143-148. 72. Lo Gerfo FW, Quist WC, Nowak MD, Crawshaw HM,
55. Schrötter D, Spiess E, Paweletz N, Benker R. A procedure Haudenschild CC. Downstream anastomotic hyperplasia.
for rupture free preparation of confluently grown mono- A mechanism of failure in dacron arterial grafts. Ann Surg
layer cells for scanning electron microscopy. J Electr 1983; 197:479-483.
Microsc Techn 1984; 1:219-225. 73. Block PC, Myler RK, Stertzer S, Fallon JT. Morphology
56. Cutler SJ, Ederer F. Minimum utilization of the life table after transluminal angioplasty in human beings. N Engl J
method in analyzing survival. J Chron Dis 1958; Med 1981; 305:382-385.
8:699-712. 74. Ferns GAA, Raines EW, Sprugel KH, Motani AY, Reidy
57. Rutherford RM, Nishikimi N. “Graft Thrombosis and MA, Ross R. Inhibition of neointimal smooth muscle cell
Thormboembolic complications”, 501-510, in Rutherford accumulation after angioplasty by an antibody to PDGF.
RB, ed., Vascular Surgery, third edition, WB Saunders Science 1991; 253:1129-1132.
Company 1989, Philadelphia. 75. Olson NE, Chao S, Lindner V, Reidy M. Intimal smooth
58. Scott SM, Barth MG, Gaddy LR, Ahl Jr ET. The role of muscle cell proliferation after balloon catheter injury. The
circulating cells in the healing of vascular prostheses. J role of basic fibroblast growth factor. Am J Path 1992;
Vasc Surg 1994; 19:585-593. 140:1017-1023.
59. Williams SK, Kleinert LB, Rose D, McKenney S. Origin 76. Burgess WH, Maciag T. The heparin-binding (fibroblast)
of endothelial cells that line expanded growth factor family of proteins. Annu Rev Biochem 1989;
polytetrafluoroethylene vascular grafts sodded with cells 58:576-606.
from microvascularized fat. J Vasc Surg 1994; 19:594-604. 77. Ullrich A, Schlessinger J. Signal tranduction by receptors
60. Cantelmo NL, Quist WC, Lo Gerfo FW, Quantitative with tyrosine kinase activity. Cell 1990; 61:203-212.
analysis of anastomotic intimal hyperplasia in paired 78. Turnbull JE, Gallagher JT. Heparan sulfate: Functional
Dacron and PTFE grafts. J Cardiovasc Surg 1989; role as a modulator of fibroblast growth factor activity.
30:910-915. Biochem Soc Trans 1993; 21:477-482.
61. Chervu A, Moore WS. An overview of intimal hyperpla- 79. Reiland J, Rapraeger AC. Heparan sulphate proteoglycan
sia. Surg Gynecol Obstet 1990; 171:433-447. and FGF receptor target basic FGF to different intracellu-
62. Angelini GD, Bryan AJ, Williams HMJ, Soyombo AA, lar destination. J Cell Sci 1993; 105:1085-1093.
Williams A, Tovey J, Newby AC. Time-course of medial 80. David G. Integral membrane heparan sulfate proteoglcans.
and intimal thickening in pig venous arterial grafts: Rela- FASEB J 1993; 7:1023-1030.
tionship to endothelial injury and cholesterol accumula- 81. Kan M, Wang F, Xu J, Crabb WC, Hou J, McKeehen WL.
tion. J Thorac Cardiovasc Surg 1992; 103:1093-1103. An essential heparin-binding domain in the fibroblast
63. Imparato AM, Bracco A, Geun EK, Zeff R. Intimal and growth factor receptor kinase. Science 1993;
neointimal fibrous proliferation causing failure of arterial 259:1918-1921.
reconstruction. Surgery 1972; 72:1007-1017. 82. Lindner V, Reidy MA. Proliferation of smooth muscle cells
64. Ip JH, Fuster V, Badimon L, Badimon J, Taubman MB, after vascular injury is inhibited by an antibody against
Chesebro JH. Syndromes of accelerated atherosclerosis: fibroblast growth factor. Proc Natl Acad Sci USA 1991;
Role of vascular injury and smooth muscle cell prolifera- 88:3739-3743.
tion. J Am Coll Cardiol 1990; 15:1667-1687. 83. Schoen FJ, Castellot JJ Jr. Vascular graft intimal fibrous
65. Clagett GP. Morphogenesis and clinicopathologic charac- hyperplasia: Prospects for pharmacological inhibition. J
teristics of recurrent carotid disease. J Vasc Surg 1986; Vasc Surg 1991; 13:758-761.
3:10-23. 84. Chervu A, Moore WS, Quinones-Baldrich WJ, Henderson
66. Healy DA, Zierler RE, Nicholls SC et al. Long-term fol- T. Efficacy of corticosteriods in suppression of intimal
low-up and clinical outcome of carotid restenosis. J Vasc hyperplasia. J Vasc Surg 1989; 10:129-134.
Surg 1989; 10:662-669. 85. O’Malley MK, McDermott EW, Mehigan D, O’Higgins NJ.
67. Vroegindeweij D, Kemper FJ, Tielbeek AV, Buth J, Role for prazosin in reducing the development of rabbit
Landman G. Recurrence of stenoses following balloon intimal hyperplasia after endothelial denudation. Br J Surg
angioplasty and Simpson atherectomy of the femoro- 1989; 76:936-938.
popliteal segment. A randomised comparative 1-year fol- 86. El-Sanadiki MN, Cross KS, Murray JJ, Schuman RW,
low-up study using colour flow duplex. Eur J Vasc Surg Mikat E, McCann RL, Hagen P-O. Reduction of intimal
1992; 6:164-171. hyperplasia and enhanced reactivity of experimantal vein
68. Kuntz RE, Safian RD, Levine MJ, Reis GJ, Diver DJ, Baim bypass grafts with verapamil. Ann Surg 1990; 212:87-96.
DS. Novel appproach to the analysis of restenosis after 87. Fanelli C, Arnoff R. Restenosis following coronary
the use of three new coronary devices. J Am Coll Cardiol angioplasty. Am Heart J 1990; 119:357-368.
1992; 19:1493-1499. 88. Kouchi Y, Esato K, O-Hara M, Zempo N. Effect of pros-
69. Graham LM, Brothers TE, Vincent CK, Burkel WE, taglandin I2 analogue TRK-100 on the suppression of in-
Stanley JC. The role of an endothelial cell lining in limit- timal fibrous proliferation. J Vasc Surg 1992; 16:232-238.
CHAPTER 12
Human Clinical Trials of Microvascular Endothelial
Cell Sodding
Stuart K. Williams
History of EC Transplantation
W hile advances in clinical vascular surgery have resulted in significant progress in the
treatment of vascular diseases, a significant frustration has been the inability to sustain
the patency of small diameter (< 6 mm) synthetic vascular grafts. A specific example of the
poor performance of small diameter synthetic grafts is in peripheral bypass procedures.
When compared to saphenous vein autologous grafts, synthetic grafts constructed of either
ePTFE or Dacron exhibit clinically poor outcomes. The clinical experience with synthetic
grafts for coronary artery bypass grafting is even more disappointing, with only rare ex-
amples of successful use of synthetic grafts for coronary revascularization.
The importance of a stable endothelial cell lining on natural blood vessels toward
maintenance of an anti-thrombogenic, metabolically active surface, and as a barrier con-
trolling the selective transport of cellular and plasma protein constituents of blood, has
been well established.1,2 The importance of establishing an endothelial cell lining on syn-
thetic grafts has also been recognized by numerous research laboratories.3,4 Numerous tech-
niques have been proposed toward the accelerated formation of an endothelial cell lining on
synthetic grafts. These techniques generally fall into the two categories of either accelerated
spontaneous endothelialization or accelerated endothelialization through cell transplanta-
tion. Spontaneous endothelialization has been explored through several mechanisms, in-
cluding accelerated pannus ingrowth and stimulated migration of vascular elements di-
rectly though the wall of the graft.5 The efficiency of spontaneous endothelialization is highly
variable, with significant species to species variation. More extensive work has been per-
formed evaluating the use of endothelial cell transplantation toward accelerated
endothelialization of synthetic vascular grafts.6
Numerous variables have been explored during the development of techniques for
endothelial cell transplantation, including source of endothelial cells for transplantation,
the need to culture cells prior to transplantation, methods for cell deposition and graft sur-
face modifications to improve cell-polymer interactions. Each of these variables has been
shown to be critical to the development of operating room compatible methods for endo-
thelial cell transplantation. Prior to the initiation of human clinical trials, endothelial cell
transplantation methods were evaluated in several preclinical animal models where both
the safety of the methods was established and, finally, the effects of endothelial cell trans-
plantation on graft patency were confirmed. This latter point has been critical to support
the hypothesis that the accelerated formation of an endothelial cell monolayer on synthetic
vascular grafts following endothelial cell transplantation will improve graft patency.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
144 Tissue Engineering of Prosthetic Vascular Grafts
Following extensive evaluation of different variables sue culture medium type 199E. This solution was pushed
for endothelial cell transplantation, our laboratory estab- through the interstices of the graft using a pressure gradient
lished methods for the use of autologous endothelial cell equal to 5 psi. Grafts were prewet for at least 30 minutes.
transplantation using adipose tissue-derived microvascular
endothelial cells. Preclinical animal studies established that Cell Deposition
endothelial cells could be isolated from fat and transplanted A suspension of endothelial cells was prepared in
rapidly onto the lumenal surface of synthetic grafts (both medium 199E at a density to provide 2 x 105 cells/cm2 graft
ePTFE and Dacron), and that this form of graft treatment lumenal surface area. The solution was placed into the graft
resulted in a statistically significant improvement in graft and a pressure gradient created across the wall of the graft
patency. The development of these methods was not with- of 5 psi, resulting in the flow of fluid through the graft and
out controversy. Several investigators questioned whether concomitant deposition of endothelial cells. We term this
adipose tissue even contained endothelium, suggesting the process pressure sodding of cells.
cells isolated from fat were mesothelium and not endothe-
lium.7 Extensive evaluation of the cellular components iso- Time to Graft Implantation
lated from adipose tissue following its dissociation using col- Immediately after pressure sodding the grafts were
lagenase established the major contribution of endothelium brought to the operating field and implanted.
to the total cellular makeup of adipose tissue.8 But more As part of this clinical evaluation of endothelial cell
important was the fact that microvascular endothelium transplantation using autologous microvessel endothelial
could be rapidly transplanted onto synthetic vascular grafts, cells, a specialized endothelial cell isolation and cell deposi-
resulting in the accelerated formation of an endothelial cell tion kit was developed. This kit and its use is diagrammed
monolayer and improved patency in the grafts once im- in Figure 12.1. The performance of this kit was modeled af-
planted. A complete discussion of the results of endothelial ter the endothelial cell sodding procedure first developed
cell transplantation studies in preclinical animal models is and used during preclinical animal trails and subsequently
discussed in an accompanying chapter. used during human trials (Fig. 12.2).
from the abdominal wall was performed with a liposuction Overview of Patients
cannula and specially designed 60 cc syringe. Approximately All patients were referred to participating vascular
50 g of fat was removed from each patient and processed for surgeons for amputation.
MVEC isolation. The completely assembled kit, being used Pt. 01. 52 yr old M, diabetic, coronary artery and re-
during a human endothelial cell sodding procedure, is shown nal artery disease, rest pain, severe left lower ischemia, his-
in Figure 12.3. The isolation procedure as outlined above tory of failed bypass graft in leg to receive sodded graft. Re-
resulted in ePTFE grafts with approximately 2 x 105 cells/cm2 ceived ileo-peroneal sodded bypass graft.
graft lumenal surface area. Grafts were immediately im- Pt. 02. 82 yr old M, history of failed bypass grafts in
planted in patients. study leg. Rest pain. Received ileo-peroneal sodded
bypass graft.
146 Tissue Engineering of Prosthetic Vascular Grafts
Pt. 03. 71 yr old F, diabetic with history of coronary Pt. 10. 83 yr old M, diabetic, hypertensive with his-
artery and peripheral vascular disease. Previously received tory of failed bypass grafts in study leg. Received femoral-
several bypass grafts including in situ vein graft. Received tibial sodded bypass graft.
femoral-tibial sodded bypass graft. Pt. 11. 57 yr old M diabetic with hypertension. His-
Pt. 04 69 yr old F, history of failed bypass grafts and tory of peripheral and cardiac bypass grafts including failed
angioplasty. Received ileo-tibial sodded bypass graft. autologous peripheral graft in study leg. Received femoral-
Pt. 05. 75 yr old F, diabetic with hypertension. History tibial sodded bypass graft.
of failed bypass grafts. Received femoral-tibial sodded by-
pass graft. Patient Follow Up
Pt. 06. 74 yr old M. History of cardiovascular disease All patients received routine follow up evaluation of
including CABG, peripheral vascular, renal artery proce- graft function, including color flow Doppler and duplex
dures. Presented with intermittent claudication. Received imaging. Of the original 11 patients, seven were available
femoral-popliteal sodded bypass graft. for follow up at four years while four expired prior to this
Pt. 07. 72 yr old F, diabetic with hypertension. History time due to nongraft related causes. The cumulative patency
of failed bypass grafts including previous posterior tibial in of the study grafts is illustrated in Figure 12.4. As expected
situ bypass graft in study leg which failed after 7 months. of the patient population in this trial, several patients have
Received femoral-posterior tibial sodded bypass graft. experienced other vascular complications including cardiac
Pt. 08. 74 yr old M, history of peripheral and cardiac arrhythmias, myocardial infarctions, nonstudy leg periph-
disease. Received ilio-peroneal sodded bypass graft. eral vascular disease progression, renal disease and pulmo-
Pt. 09. 75 yr old M, received ilio-tibioperoneal sodded nary embolism. Several patients have also undergone treat-
bypass graft. ment for cancer. Follow up evaluation of patients did not
provide information predictive of subsequent graft failure. 2. Richardson JV, Wright CB, Hiratzka LF. The role of en-
One patient exhibited a mid-graft concentric intimal thick- dothelium in the patency of small venous substitutes. J
ening just below the knee approximately 9 months after graft Surg Res 1980; 28:556-562.
implantation. This thickening had disappeared at subsequent 3. Zilla P, Preiss P, Groscurth P, Rosemeier F, Deutsch M,
Odell J, Heidinger C, Fasol R, von Oppel U. In vitro-lined
graft follow-up.
endothelium: Initial integrity and ultrastructural events.
Surg 1994; 116(3):524-534.
Conclusions 4. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T,
This trial was designed as an initial safety study to Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I.
evaluate the ability to isolate endothelial cells from Clinical in vitro endothelialization of femoropopliteal by-
liposuction fat and transplant these cells onto ePTFE grafts. pass grafts: An actuarial follow-up over three years. J Vasc
The initial four year follow up data is quite encouraging and Surg 1994; 19(3):540-548.
suggests endothelial cell transplantation can be performed 5. Clowes AW. Graft endothelialization: The role of angio-
using an automated kit. The patient population selected for genic mechanisms. J Vasc Surg. 1991; 13:734-736.
this trial represents individuals with significant vascular dis- 6. Williams SK. Endothelial cell transplantation. Cell Trans
ease. The lack of significant limb loss in this patient popula- 1995; 4:401-410.
7. Visser MJP, van Bockel JJ, van Muijen GNP, van
tion during the first three years follow up is equally encour-
Hinsbergh VWM. Cells derived fromomental fat tissue and
aging. We conclude that an expanded trial of endothelial cell used for seeding vascular prostheses are not endothelial
transplantation using autologous endothelial cells derived in origin: A study on the origin of epitheloid cells de-
from liposuction derived fat is warranted. rived from human omentum. J Vasc Surg 1991;
13:373-381.
References 8. Williams SK, Wang TF, Castrillo R, Jarrell BE. Liposuction
1. Quist WC, Haudenshield CC, Logerfo FW. Qualitative derived human fat used for vascular graft sodding con-
microscopy of implanted vein grafts: Effects of graft in- tains endothelial cells and notmesothelial cells as the ma-
tegrity on morphological fate. J Thorac Cardiovasc Surg jor cell type. J Vasc Surg 1994; 19:916-923.
1992; 103:671-677.
Macrovascular Endothelial Cell Transplantation
CHAPTER 13
In Vitro Endothelialization: Its Contribution Towards
an Ideal Vascular Replacement
Peter Zilla
E very era in medicine has been driven by one particular discipline which recognized an
exciting new development occurring outside its own sphere as an opportunity for a quan-
tum leap. Although this initial phase of integrating an unfamiliar dimension into a tradi-
tional medical dominion was seldom blessed with clinical success, retrospectively this era is
always perceived as the grand epoch of the particular discipline. For instance, the integra-
tion of technical achievements into a discipline like surgery, previously considered to be
more a skillful art than anything else, made cardiac surgery possible. Those pioneering car-
diac surgeons who dared to let a mechanical pump take over the heart’s function, thus al-
lowing procedures on the open heart, were initially viewed with suspicion by their peers.
Typically, it was not a single technical aspect which was absorbed into this young discipline
but rather a basic openness towards the integration of mechanical and electrical contrap-
tions into the circulatory system, a part of the body which was considered to be untouchable
since the days of Theodor Billroth. It seems also typical that the enthusiasm of such a pio-
neering phase eventually draws the newly discovered, unrelated support discipline further
into its sphere of influence. It was not a coincidence that Earl Bakken developed the first
pacemaker in Minneapolis in the wake of the emerging new discipline of cardiac surgery,
which was plagued by postoperative rhythm problems in this early phase. Retrospectively,
the quantum leap which this pinnacle of cardiac surgery initiated for other medical profes-
sions remains unchallenged.
In vascular surgery, the late 1970s and early 1980s may well, retrospectively, turn out
to have been a similarly fateful period for medicine. However, in contrast to cardiac surgery
twenty years before, it was not the world of sophisticated mechanical devices and electronics
which were integrated into medicine but the amazing world of biology. Symptomatically,
naive amateurism and polarization characterize the early days of this era. Amateurism, be-
cause the first to test the water were surgeons and not biologists. This is the trademark of all
developments of this kind in medicine: Brave and alert medical doctors initially apply dis-
coveries from an alien field to medicine themselves. On the one hand, this effervescent breed
with their urge for knowledge has all the ingredients to overcome initial thresholds and
obstacles. On the other hand, it is often a surgical discipline within the medical profession,
with its desire for clear cut and simple solutions, which drives such initiatives. As a conse-
quence, this surgical mentality may endanger its own achievements when necessary com-
plexity falls victim to impatience. In the case of the awaking biological awareness of the late
1970s, it was indeed surgeons again who were behind it. Almost predictably, they caused
their own setback as they had in previous years with heart transplantation by not handing
the newborn over to scientists in time. In the case of heart transplantation, immune and
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
152 Tissue Engineering of Prosthetic Vascular Grafts
drug research rather than the fury of an ongoing surgical openly admitted, each champion of a particular approach
push could have saved 10 years. The eventual breakthrough towards graft endothelialization therefore hoped for com-
of the cyclosporin era fought an uphill battle against the parable fame. Another aspect of those days in the 1960s was
prejudices arising from the surgical desire for a quick fix that their pioneers demonstrated that almost everything
during the preceding 13 years. In the case of the incorpora- which previously seemed unresolvable became feasible
tion of biology into prosthetic vascular surgery, it was the through the application of new materials and mechanically
endless striving of surgeons to succeed with a simple and determined technologies. Naturally, the emergence of a new
instantly applicable, single-staged procedure right from the dimension like biology caused friction. As a result, at all con-
beginning which almost meant the death knell for the en- ferences in the 1980s, the believers in existing technologies
tire idea 10 years later. A further tragedy which repeats itself asked angrily after each talk on endothelial seeding whether
in the early phases of “quantum leap” eras lies in the typical the speakers were aware that graft patencies depended pri-
negative feedback amongst the alien partners which soon marily on surgical skills. Therefore, they recommended that
follows the initial enthusiasm. Surgeons often feel intimi- the presenter spend his time improving those skills rather
dated by the thought of scientific complexity and scientists than wasting it in the laboratory. The fact that this latter
shy away from the “gung-ho” approach of surgeons. As con- group of surgeons still represents the majority within our
tradictory as it may sound, in the end it is often the perse- ranks proves how successful the previous quantum leap was
verance of a few dedicated surgeons who continue their in getting its new standards generally accepted. In his book
mission regardless, accepting a higher level of complexity. The Structure of Scientific Revolutions, Thomas Kuhn1 ex-
This perseverance allows an idea to survive long enough for plains the mechanisms involved in such paradigm shifts.
its final breakthrough. In heart transplantation, Norman Stephen Hawking2 brought it to the point by arguing that
Shumway fulfilled this role. In the attempt to incorporate people are very reluctant to give up a theory in which they
biological principles into prosthetic vascular surgery, in vitro have invested a lot of time and effort. This theory defining
endothelialization may, in retrospect, have played a similar concepts and procedures is the accepted paradigm, which is
role. If the next decade of research succeeds with tissue en- recognized by all scientists working in that field. If, however,
gineering of physiologically functional vascular prostheses, unexpected developments result in increasing inconsistency
the credit for opening a new era in medicine will undoubt- with the prevailing paradigm, a tense situation ensues
edly be deserved by the early pioneers of single staged en- amongst the scientists. At that stage the majority initially
dothelial cell seeding. However, the role which Norman questions the accuracy of the observations. If that fails, they
Shumway played in heart transplantation—to prevent the try to modify the existing theory in an ad hoc manner. Even-
flickering flame from being extinguished—may well be tually, the old paradigm becomes creaking and ugly and a
granted to those who continuously propagated the complex new one is accepted which explains all the awkward obser-
but eventually successful variant of in vitro vations in an elegant and natural manner. Quantum leaps
endothelialization, to clinically prove the benefit of a prin- in medicine are certainly not such revolutionary shifts in
ciple and thus provide sufficient incentive for today’s efforts scientific paradigms, but their principles and their conse-
towards a broadly acceptable breakthrough. quences are similar. The angry discussant, for instance, ques-
tioning the purpose of merging vascular prosthetic research
Historical Perspective with biology, is not an isolated phenomenon but rather a
If the amateurism of the pioneering years of endothe- typical veteran who may have actively contributed to
lial seeding, with its lack of involvement of basic scientists, yesterday’s paradigm shift. This again has many parallels in
contributed to a delay of today’s drive towards an integrated truly revolutionary paradigm shifts in science. Albert
approach to cardiovascular tissue engineering, one can at Einstein, for instance, who caused a paradigm shift in phys-
least explain this shortcoming by the fundamental differ- ics with his special theory of relativity in 1905 was himself,
ence between the worlds of surgeons and biologists. This many years later, one of the major antagonists to the next
does not apply to the polarization which plagued all of us paradigm shift by resisting the acceptance of quantum me-
who were involved in efforts to “biolize” prosthetic grafts chanics. Surgeons are thus in good company with regard to
from within our own discipline. This polarization was two- the resistance to paradigm shifts. However, the 13 year delay
fold: On the one hand, each different approach to endothe- in achieving this goal in heart transplantation, and the 20
lial seeding was almost religiously upheld by the respective year delay in accepting biology in cardiovascular surgery,
groups which stood for it. On the other hand, a fiercely makes it clear that we are dealing with a particularly conser-
fought confrontation of principal values led to a schism vative discipline.
which continues to divide the surgical community today. Having tried to understand the driving forces behind
Both polarizations may be explained in terms of the previ- the twofold polarization which characterized the past 20
ous quantum leap era, under whose spell the majority of years of attempts to create a biologically functional vascular
cardiovascular surgeons still stood. One aspect of the pre- prosthesis, it seems easier to explain the concrete develop-
ceding grand era of cardiovascular pioneering was that it ments of those two decades as well as today’s situation. The
created heroes as never before. Each facet of the overall quan- internally dividing question, for instance, regarding the prin-
tum leap was associated with a big name, whether it was cipal approach to the endothelialization of a prosthetic sur-
Lillihey, Kirklin, De Bakey, Barnard or Cooley. Even if not face, namely the acceptance of initial complexity versus
In Vitro Endothelialization: Its Contribution Towards an Ideal Vascular Replacement 153
a priori simplicity, was not an issue at the beginning when Current and Future Perspective
no pressure of expectation was exerted by the surgical com- Today’s cumulative experience with clinical endothe-
munity. In the early to mid 1970s, the entire initiative to lial cell lining covers almost a decade and comprises more
surface endothelialization was driven by attempts to culture than 200 patients.52-60 It has clearly demonstrated that in
endothelial cells on synthetic surfaces prior to implanta- vitro endothelialized synthetic prostheses are equal or better
tion.3,4 Pioneers of endothelial seeding, like the groups of than saphenous vein grafts with regard to patency in all
Jim Stanley and Linda Graham were among those who first anatomical positions and any clinical stage other than
cultured autologous endothelial cells on vascular graft sur- stage IV.52-60
faces.5,6 Only subsequently in the mid-1980s, when surgeons Although ePTFE grafts with 30 ∝m internodal distance,
saw the opportunity of implementing their discoveries which are noncompliant and do not allow transmural tis-
through main commercial players, did the focus shift almost sue ingrowth, were used for these studies, the autologous
entirely to single stage procedures. Ironically, when the great- surface endothelium alone was not only sufficient to sig-
est enthusiasm for endothelial seeding seized the surgical nificantly improve patency rates, but also led to the devel-
community at the beginning of the second half of the 1980s, opment of a neomedia between an internal elastic mem-
the death sentence was already sealed through both brane underneath the endothelium and the ePTFE surface.59
premature clinical trials7-11 and “commercial kits” for clini- This observation indicates that the previous fear of “con-
cal single staged procedures. What happened after the ma- taminating” smooth muscle cells was unfounded. Further-
jority of disappointed vascular surgeons turned away from more, it not only supports the current holistic approach to-
this idea really resembled the above mentioned late devel- wards tissue engineered prosthetic vascular grafts which aim
opments in heart transplantation. With the hype of the early at fully functional neoarteries, but also removes the last rem-
1980s over, those who continued were prepared to accept nants of the previous polarization between mixed microvas-
both a long and arduous route of homework prior to imple- cular mass seeding and macrovascular in vitro lining.
mentation and the knowledge that their contribution would The tools which biology offers today often seem like a
be a quiet one. dream to those of us who believed in the idea of integrating
Attitude-wise back to square one, the remaining biology into prosthetic grafts, long before vascular biology
groups focused on the main weakness of graft had entered its maturity. At the same time we also see that
endothelialization, the low cellular inoculum. Mass harvest these tools require a commitment of all parties to a many-
methods for microvascular cells,12-16 as well as mass culture fold higher level of complexity than the one from which most
procedures for macrovascular endothelial cells,17-19 were sci- of us had shied away in the early days of endothelial seed-
entifically refined. One of the main reasons for a more re- ing. Last but not least, we as surgeons are still the majority
laxed approach in this second half of the 1980s was the con- shareholders in this venture, although the scientists will soon
viction that a principle rather than a particular solution take over the supervisory board. This is a necessary and cor-
needed to be proven. At the onset of the era of vascular biol- rect development which should have happened many years
ogy, nobody challenges that a biologically functional pros- ago. However, even if the scientists do take over the lead, in
thesis would eventually come as a product from the shelf, the end there will still be three partners: the industry, the
integrating all encoded signals for spontaneous healing. surgeons and the scientists. The industry will need strongly
Nevertheless, such an undertaking needs a critical mass convincing data to voluntarily accept the replacement of a
which can more easily be achieved on the basis of at least simple and profitable product by a complex and expensive
one proven principle. Although important principles of to- one. The surgeons will need the final painful push to accept
day like intramural contractility, compliance and cellular the shift of a paradigm and the biologists will need both the
quiescence could not yet be tackled, we all focused on the true commitment of the other two parties and a strong
proof that graft endothelialization alone can already dra- motivation with regard to the broad application of their dis-
matically improve synthetic graft performance, even if the coveries before they will wholeheartedly join hands. This may
old prosthetic scaffolds of yesterday are used. In a step by well be the last challenge for the veteran surgeons on board.
step approach, adherence and shear stress resistance of cul- Convincing clinical studies with a method which signifi-
tured endothelial cells on various protein matrices were as- cantly improves graft performance will eventually break the
certained20-37 prior to in vivo experiments ranging from ca- resistance against a paradigm shift amongst our peers and
nine implants20 to primate experiments38,39 and preclinical thus prepare the way for the acceptance of tissue engineer-
primate studies.40 The success of those implants was con- ing. Acceptance of a complex approach by surgeons will
vincing enough to apply the principle to very small diam- mellow the scientists who still see us as “gung-ho” cowboys.
eter grafts41 as well as to bioprosthetic surfaces42-47 and to And finally, a joint front of scientists and surgeons will even-
heart valves.48-51 Eventually, the step into clinical trials had tually force the industry to see beyond today’s profits. Last
to be taken. However, in contrast to the clinical studies with but not least, one needs to keep in mind that even a “Los
single staged endothelial seeding, the situation regarding Alamos” approach to tissue engineering will take many years
clinical trails with in vitro lining was distinctly different: By for the development and many years for clinical trials. This
having accepted the disadvantages of a complex procedure, consideration certainly upgrades clinical in vitro
we had eliminated most of the uncertainties prior to com- endothelialization to a procedure which could benefit an
mencing the trials. uncountable number of patients over many years to come.
154 Tissue Engineering of Prosthetic Vascular Grafts
on different matrices. Eur J Vasc Endovasc Surg 1996; 48. Bengtsson LA, Haegerstrand AN. Endotheliaization of
11:127-133. mechanical heart valves in vitro with cultured adult hu-
35. Gillis-Haegerstrand C, Frebelius S, Haegerstrand A, man cells. J Heart Valve Dis 1993; 2:352-356.
Swedenborg J. Cultured human endothelial cells seeded 49. Bengtsson L, Radegran K, Haegerstrand A. In vitro
on expanded polytetrafluoroethylene support thrombin- endothelialization of commercially available heart valve
mediated activation of protein C. J Vasc Surg 1996; bioprostheses with cultured adult human cells. Eur J
24:226-234 Cardiothorac Surg 1993; 7:383-398.
36. Haegerstrand A, Bengtsson L, Gillis C. Serum proteins 50. Lehner G, Fischlein T, Baretton G, Murphy JG, Reichart
provide a matrix for cultured endothelial cells on ex- B. Endothelialized biological heart valve prostheses in the
panded polytetrafluoroethylene vascular grafts. Scand J non-human primate model. Eur J Cardiothorac Surg 1997;
Thorac Cardiovasc Surg 1993; 27:21-26. 11:498-504.
37. Leseche G, Bikfalvi A, Dupuy E, Tobelem G, Andreassian 51. Fischlein T, Lehner G, Lante W, Fittkau M, Murphy JG,
B, Caen J. Prelining of polytetrafluoroethylene grafts with Weinhold C, Reichart B. Endothelialization of cardiac
cultured human endothelial cells isolated from varicose valve bioprostheses [published erratum appears in Int J
veins. Surgery 1989; 105:36-45. Artif Organs 1994 Jul; 17(7):412].
38. Schneider PA, Hanson SR, Todd M, Price BA, Harker LA. 52. Kadletz M, Magometschnigg H, Minar E, Konig G,
Confluent durable endothelialization of endarterectomized Grabenwoger M, Grimm M, Wolner E. Implantation of
baboon aorta by early attchment of cultured endothelial in vitro endothelialized polytetrafluoroethylene grafts in
cells. J Vasc Surg 1990; 11:365-372. human beings. A preliminary report. J Thorac Cardiovasc
39. Krupski WC, Bass A, Anderson JS, Kelly AB, Harker LA. Surg 1992; 104:736-742.
Aspirin-independent antithrombotic effects of acutely at- 53. Magometschnigg H, Kadletz M, Vodrazka M,
tached cultured endothelial cells on endarterectomized Grabenwoger M, Grimm M, Bock P, Leukauf C, Trubel
arteries. Surgery 1990; 108:283-291. W, Wolner E. Changes following in vitro endothelial cell
40. Zilla P, Preiss P, Groscurth P, Rösemeier F, Deutsch M, lining of ePTFE prostheses: Late morphologic evalulation
Odell J, Heidinger C, Fasol R, von Oppell U. In vitro- of six failed grafts. Eur J Vasc Surg 1994; 8:502-507.
lined endothelium: Initial inegrity and ultrastructural 54. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T,
events. Surgery 1994; 116:524-534. Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I,
41. Gherardini G, Haegerstrand A, Matarasso A, Gurlek A, Fischlein T. Clinical in vitro endothelialization of
Evans GR, Lundeberg T. Cell adhesion and short-term femoropopliteal bypass grafts: An actuarial follow-up over
patency in human endothelium preseeded 1.5mm three years. J Vasc Surg 1994; 19:540-548.
polytetrafluoroethylene vascular grafts: An experimental 55. Magometschnigg H, Kadletz M, Vodrazka M, Dock W,
study. Plast Reconstr Surg 1977; 99:472-478. Grimm M, Minar E, Staudacher M, Fenzl G, Wolner E.
42. Hoch J, Dryjski M, Jarrell BE, Carabasi RA, Williams SK. Prospective clinical study with in vitro endothelial cell lin-
In vitro endothelializaton of an aldehyde-stabilized native ing of expanded polytetrafluoroethylene grafts in crural
vessel. J Surg Res 1988; 44:545-554. repeat reconstruction. J Vasc Surg 1992; 15:527-535.
43. Bengtsson LA, Phillips R, Haegerstrand AN. In vitro 56. Fischlein T, Zilla P, Meinhart J, Puschmann R, Vesely M,
endothelialization of photo-oxidatively stabilized xenoge- Eberl T, Balon R, Deutsch M. In vitro endothelializaation
neic pericardium. Ann Thorac Surg 1995; 60:S365-S368. of mesosystemic shunt: A clinical case report. J Vasc Surg
44. Bengtsson L, Ragnarson B, Haegerstrand A. Lining of vi- 1994; 19:549-554.
able and non-viable allogeneic and xenogeneic cardiovas- 57. Leseche G, Ohan J, Bouttier S, Palombi S, Bertrand P,
cular tisue with cultured adult human venous endothe- Andreassian B. Above-knee femoropopliteal bypass graft-
lium. J Thorac Cardiovasc Surg 1993; 106:434-443. ing using endothelial cell seeded PTFE grafts: Five year
45. Leukauf C, Szeles C, Salaymeh L, Grimm M, Grabenwoger clincial experience. Ann Vasc Surg 1995; 9:S15-S23.
M, Moritz A, Wolner E. In vitro and in vivo 58. Laub HR, Duwe J, Claus M. Autologous endothelial cell
endothelialization of glutaraldehyde treated bovine peri- seeded PTFE vascular grafts for coronary artery bypass:
cardium. J Heart Valve Dis 1993; 2:230-235. First clinical results. 15th Intern Cardiovasc Surg. Sym-
46. Eybl E, Grimm M, Grabenwoger M, Bock P, Muller MM, posium, Zurs, Austria
Wolner E. Endothelial cell lining of bioprosthetic heart 59. Deutsch M, Meinhart J, Vesely M, Fischlein T, Groscurth
valve materials. J Thorac Cardiovasc Surg 1992; P, von Oppell U, Zilla P. In vitro endothelialization of
104:763-769. expanded polytetrafluoroethylene grafts; A clinical case
47. Grabenwoger M, Grimm M, Eybl E, Moritz A, Muller report after 41 months of implantaton. J Vasc Surg 1997;
MM, Bock P, Wolner E. Endothelial cell lining of 25:757-763.
bioprosthetic heart valve material. J Card Surg 1992; 60. Meinhart J, Deutsch M, Zilla P. Eight years of clinical
7:79-84. endothelial cell transplantation. Closing the gap between
prosthetic grafts and vein grafts. ASAIO Journal 1997;
43:M515-M521.
CHAPTER 14
Serial Cultivation of Human Endothelial Cells
Caroline Gillis-Hægerstrand, Anders Hægerstrand
Introduction
U nderstanding of the tremendous capabilities of the endothelial cell has grown consider
ably during the last decades. The use of cell culture techniques has been instrumental in
this development. The single most important breakthrough for endothelial cell (EC) cul-
ture, and still probably the most used technique, was published by Jaffe and coworkers in
1973. They described how to isolate and propagate human umbilical vein endothelial cells
(HUVECs).1 Since then several modifications and improvements have been published,2,3
with the fundamental principle remaining unchanged, i.e., the incubation of a vessel, ideally
clamped at both ends, with a collagenase containing solution, and the subsequent plating
and cultivation in a protein-coated tissue culture flask, the cells being fed with a medium
enriched by relatively large amounts of serum and/or growth factors. Some basic ideas about
the fundamental functions of ECs, e.g., the production of arachidonic acid derivatives, the
expression of adhesion molecules, the tight interactions with leukocyte or smooth muscle
cells and the synthesis of nitric oxide (NO), have been derived from the use of cultured ECs,
mainly HUVECs. It should be remembered that ECs in culture are something very different
than ECs in their natural environment. Compared to many, if not most, other cell types, it is
harder to mimic in vitro what the natural circumstances are under which the ECs operate.
This must be remembered when interpretations of EC functions are made. The in vivo cor-
relate remains the only true measure of the predictive value of in vitro findings.
The idea of culturing endothelial cells for clinical purposes is not new, but only a
limited number of studies have actually been performed. In this review we will discuss some
basic techniques and principles for the culture of human endothelial cells and some of the
future possibilities.
There are differences in morphology, growth rate and types during culture, e.g., fibroblasts, and the long term cul-
protein synthesis between cultured arterial and venous ECs,9 tivation itself to reach the desired amount of cells make EC
as well as between ECs derived from capillaries, arterioles culture for clinical purposes a complicated task.
and venules.10 Venous ECs are larger, thinner and more pleo-
morphic than ECs deriving from aorta. ECs derived from Culture Techniques for Human ECs
veins are considered more difficult to establish in culture A lot can be said about general requirements for cul-
and they grow at a lower growth rate.9 These differences are ture of human cells. Some relatively general components
believed to be due to the different requirements by the he- used during culture of human ECs are summarized in
modynamic environment. Whether this has an impact on Figure 14.1.
utility for transplantation purposes is not known. ECs are continuously exposed to blood, which is
ECs from different sizes and types of blood vessels known to contain growth stimulating factors, although many
show well known differences in metabolic properties, espe- at low concentrations. In our work we have been focusing
cially in their response to vasoactive mediators.11 The physi- on human serum as a source for stimulating growth and
ological significance of this heterogeneity is not clear, but attachment on the prosthetic material, whereas most inves-
may be related to local variations in the function the ECs tigators have used serum containing medium supplemented
serve, for example in arterial versus venous circulation.12 with growth factors and heparin. Serum free alternatives are
When ECs from one source are compared, there is also a commercially available, but these are often quite expensive.
vast difference in basal secretion of, e.g., PGI2 and plasmi- Such media are typically better to introduce after an initial
nogen/plasminogen inhibitors in cultures of ECs from dif- culture period using medium which contains serum. The
ferent patients.13-15 basal medium in most compositions, irrespective of serum
A characteristic that is unique for endothelial cells, dependence or independence, normally contains a relatively
regardless of their origin, is the presence of Weibel-palade high concentration of glucose, and basal media such as M199
bodies.16 In granules inside the Weibel-palade bodies, and MEM are often used. It is generally accepted that some
P-selectin and von Willebrand factor (vWF) are colocalized. matrix component precoated on the tissue culture treated
Other characteristics of the ECs, though not unique, are plastic is beneficial. Most often, as also originally described
prostacyclin production (PGI2),17 angiotensin converting by Jaffe et al, denaturated bovine collagen, i.e., gelatin, can
enzyme (ACE) activity and positive staining for the surface be used. The method by which it is prepared also seems to
antigen PECAM (CD31).18 In most cases, investigators char- fit with current guidelines on the use of bovine/xenogeneic
acterize the ECs by staining with vWF, by showing the pres- material for tissue culture. Fibronectin in concentrations of
ence of Weibel-palade bodies or by showing uptake of acety- approximately 1-10 ∝g per square centimeter is often rec-
lated low density lipoprotein (LDL), the latter being another ommended, but it should be remembered that concentra-
characteristic not restricted to ECs. tion dependent functional alterations can be expected.
Laminin, collagen type I or IV and extracellular matrix
Sources of Endothelial Cells (ECM) have also been used. Shortly after seeding, within
Several sources for isolating ECs during an operation 6 h, ECs deposit matrix components, e.g., laminin and col-
to immediately seed a vascular graft have been demonstrated, lagen type IV, on the plastic, and thereby influence their own
including freshly isolated cells from veins and fat.19-25 Tech- attachment.
niques for isolation and culture of ECs from human tissues
other than large vessels or fatty tissue have been de- Addition of Serum and Growth Factors
scribed.26-29 Others have successfully isolated ECs from Human serum (HS) and fetal calf serum (FCS) pre-
omental tissue,30 although it has been questioned whether pared for cell culture purposes contain growth factors from
they are truly endothelial.31 It is known that mesothelial cells plasma and growth factors released from platelets upon their
share several properties with ECs, especially those related to aggregation. It should be remembered that ECs grow con-
antithrombotic and fibrinolytic functions.32-34 Subcutane- siderably more slowly in blood plasma without platelets or
ous fat is probably a better source of true ECs as compared coagulation factors, which is quite often referred to as se-
to omental fat.25 Sources of ECs for effective and reproduc- rum. Cell culture serum must be prepared according to spe-
ible cultivation are fewer, especially when the cells are in- cific protocols. Serum contains several factors which pro-
tended for clinical purposes. Typically, veins such as the great mote proliferation, platelet derived growth factor (PDGF),
saphenous, basilic, cephalic and external jugular have been epidermal growth factor (EGF), transforming growth fac-
used.35-38 Cultured arterial ECs have not yet been used for tor ! (TGF-!) and insulin-like growth factors (IGFs).39 Fi-
clinical purposes to our knowledge, although that may po- broblast growth factors (FGFs) play many roles in cell
tentially be beneficial, since the purpose is most often to re- growth, differentiation and survival,40 and are well estab-
place nude or denuded tissue in an arterial position. lished growth factors for ECs. Both basic FGF and acidic
In spite of an increasing number of techniques, EC FGF are often used in recombinant form to stimulate EC
research is often hampered by the lack of sufficient num- growth. Endothelial cell growth factor (ECGF), a precursor
bers of ECs of an adequate origin. Even if vessels or tissue form of acidic FGF (AFGF), is a potent mitogen for most
rich in ECs become available, the isolation and cultivation ECs.39,40 A rat brain extract, known as endothelial cell growth
is not easy. The quality and short term storage of the tissue supplement (ECGS), containing different forms of FGFs, is
derived from the patient, the contamination of other cell also commonly used42,43 to improve cell proliferation, and
Serial Cultivation of Human Endothelial Cells 159
was the most frequent additive to EC culture a decade ago, both growth and maintenance of the epitheloid morphol-
before purified and recombinant factors were made com- ogy in human microvascular ECs.26,59,61,62 HUVECs have
mercially available at reasonable prices. also been shown to increase in cell number after stimula-
tion with CT and endogenous peptides which stimulate
Heparin and Endothelial Proliferation cAMP formation.63 Using a medium without cAMP-stimu-
The fact that heparin enhances the stimulatory effect latory compounds, Watkins and coworkers have described
of ECGF on cell proliferation in vitro is well established.43,44 aberrant cell morphologies after repeated passages using
Both heparan sulfate and heparin act by stabilizing growth trypsin.64 In our experience using cAMP-stimulatory fac-
factor activity, partly by preventing proteolytic degradation tors in the medium, the number of cells appearing aberrant,
of the growth factor.45,46 The binding and mitogenic activ- e.g., large and multinuclear, constitutes no more than one-
ity of bFGF are to some degree dependent on heparin-like fiftieth of the cells even after 8-9 passages. This may be due
molecules on the cell surface, or in their absence, on exog- to continuous cAMP stimulation or to the high proportion
enously added heparin-like molecules.47 It has recently been of serum. Cyclic AMP formation has also been shown to
shown that heparin is an FGF-independent activating ligand modulate EC migration,65,66 inhibit hypoxia-induced in-
for FGF receptor 4 (FGFR4) on cultured transfected myo- crease in EC permeability,67 enhance EC thromboresistance68
blasts, fibroblasts and lymphoid cells.48 In our work it was and to amplify agonist-induced release of EDRF.69 Both pro-
shown that heparin alone increased growth of adult human tective and indirect mechanisms may thus be of importance
ECs when heparin was added to a medium containing as for the growth promoting effects of CT/IBMX and db-cAMP.
much as 40% HS. An explanation for these results in com- We have shown that HS is more efficient than FCS in
bination with 40% serum49 is that they may be due to the stimulating growth of HSVECs and that heparin, bFGF and
presence of FGFR4 on ECs or that heparin stabilizes growth cAMP-elevating compounds, together with relatively high
factors in serum. Different growth factors may act differ- concentrations of serum, are efficient in increasing the num-
ently on cultured ECs from various vascular beds. While ber of HSVECs in culture.49 The differences in ability of the
bFGF stimulates proliferation of most ECs, afgf and PDGF different sera and other factors to enhance cell proliferation
have been shown to have no effect on large vessel may depend on the cell type being studied. In our case, it
endothelium but to stimulate microvascular cell growth.50 may be speculated that HS contains factors which are espe-
Bovine microvascular ECs have been shown to express a dif- cially suited for human cells and especially ECs. Human se-
ferent set of growth factor receptors than bovine arterial rum carries the advantage of potential autologous use, which
ECs.51 Interestingly, ECs are known to synthesize both bFGF may increase safety for the patient if the purpose of the cul-
and PDGF, which may act as autocrine and paracrine ture is for transplantation.
growth factors.52-54
Does the Culture Technique Matter
Cyclic AMP and Endothelial Cell Proliferation from a Clinical Perspective?
The addition of purified cholera toxin (CT) to the EC Zilla and coworkers have demonstrated a cell culture
culture medium leads to an increase in intracellular cyclic technique where low density seeding of first passage ECs can
adenosine monophosphate (cAMP) synthesis. The phos- yield 14 x 106 ECs in 26 days.36 The method involves in situ
phodiesterase inhibitor IBMX decreases cAMP degradation, administration of collagenase and basically no passaging of
and both thereby act to increase the intracellular cAMP level. the cells. This may have the advantage of increasing the to-
Cyclic AMP is an ubiquitous messenger which is involved in tal yield and avoidance of exposing the cells to trypsin or
many cellular processes, including changes in morphology, EDTA, which may have negative effects on the cells. In addi-
proliferation55,56 and release of vasoactive substances in re- tion to possible morphological changes, functional changes
sponse to stimulation. 57 When the membrane-bound of the cells after passaging using trypsin, e.g., altered pat-
adenylyl cyclase is activated, ATP is enzymatically converted tern of receptor expression, have also been indicated.70
to cyclic AMP, which leads to activation of protein kinase A In the study which has been used for clinical purposes,
(PKA). These events have been shown to influence cell pro- the medium contained serum, ECGS and heparin. Interest-
liferation,55 and the role of cyclic AMP as a regulator of cell ingly, heparin and ECGS have been shown to supress
proliferation has been studied extensively. Negative control prostacyclin production.44
was demonstrated as early as in the 1970s. Today, however, The choice of culture technique may actually lead to
accumulating data have shown that increase in cAMP levels selection of a selectivley responsive subpopulation of ECs
is also associated with stimulatory effects on several cell types, that will then constitute the majority of the cells available
including epithelial cells.55,58,59 It has also been shown that for transplantation. Direct comparisons of cells from one
activation of cAMP may enhance receptor binding of growth single donor cultured using different methods have not been
factors and progression of the resultant signals.60 performed. Partly differing functions of the cells on the graft,
Db-cAMP is a synthetic cAMP analog which can pen- and thus the clinical result, may become a result of the choice
etrate cell membranes. In our studies cholera toxin, IBMX of culture technique. On the other hand, one may speculate
and db-cAMP have all been shown to increase the prolifera- that the ECs will become functionally normalized once
tion of ECs derived from the great saphenous vein brought back a more natural environment. Nothing but large
(HSVECs).14,49 Karasek and coworkers have shown that com- controlled studies will reveal whether the culture technique
binations of HS and cAMP stimulation are beneficial for will affect the clinical results. Other aspects, e.g., safety and
160 Tissue Engineering of Prosthetic Vascular Grafts
rate of successful cultures using a specific technique, will 2. Gimbrone M, Cotran R, Folkman J. Human vascular en-
probably override such speculations. dothelial cells in culture. Growth and DNA synthesis. J
Cell Biol 1974; 60:673-684.
Future Possibilities of Cultured ECS 3. Maciag T, Hoover G, Stemerman M, Weinstein R. Serial
Transfer of genes for transient or prolonged produc- propagation of human endothelial cells in vitro. J Cell Biol
1981; 91:420-426.
tion of factors which would promote healing of denuded
4. Jaffe E. Cell biology of endothelial cells. Hum Pathol 1987;
vessels, e.g., bFGF or VEGF, is intriguing. Nonspecific transfer 18(3):234-239.
to the cells of the vessel wall seems feasible in animals. The 5. Schwartz S. Dynamic maintenance of the endothelium. In:
use of cultured ECs as recipients of retroviral vectors ex- Freshney, e.d. The endothelial cell—a pluripotent control
pressing secretory enzymes or hormones over long times has of the vessel wall. Basel: Karger, 1983:113-125.
been studied already.71-74 The possibility of transplanting 6. Schwartz S, Gajdusek C, Reidy M, Selden S, Haudenschild
genetically manipulated ECs expressing a substance which C. Maintenance of integrity in aortic endothelium. Fed-
is lacking in patients, such as tPA or insulin, may thus be eration Proc 1980; 39:2618-2625.
within reach. However, retrotviral vectors are not consid- 7. Sumpio BE, Widmann MD, Ricotta J, Awolesi MA,
ered ideal from a safety perspective. Adeno, lenti or herpes Watase M, Increased ambient pressure stimulates prolif-
eration and morphologic changes in cultured endothelial
viruses often carry the same connotation, and most likely
cells. J Cell Physiol 1994; 158(1:133-139.
such will be used when the disease addressed with the gene 8. Haudenschild C, Zahniser D, Folkman J, Klagsbrun M.
therapy is lethal. Since the percentage of successfully trans- Human vascular endothelial cells in culture: Lack of re-
fected cells is often limited with the techniques not involv- sponse to serum growth factors. Exp Cell Res 1976;
ing parts of a viral genome, the genetic manipulation would 98:175-183.
require an efficient culture method and optimal graft ad- 9. Wagner W, Henderssin R, Hicks H, Banes A, Johnson G.
herence to be of practical use. One possibility is to immor- Differences in morphology, growth rate and protein syn-
talize ECs without using retro- or adenoviral components thesis between cultured arterial and venous endothelial
with oncogenes such as the SV-40 Large T construct; this cells. J Vasc Surg 1988; 8(4):509-519.
has already been investigated.75 Furthermore, conditionally 10. Williams S, Jarrel B, Rose D. Isolation of human fat de-
immortalized ECs can potentially be developed, i.e., ECs that rived microvessel endothelial cells for use in vascular graft
endothelialization. In: Zilla P, Fasol R, Deutsch M, eds.
proliferate under the control of a regulatable gene such as
Endothelialization of vascular grafts. Basel: Karger, 1987.
the tetracyclin-regulated expression of Large T antigen. At 11. Shepro D, Dunham B. Endothelial cell metabolism of bio-
the end of the culture period, the oncogene could then be genic amines. Annu Rev Physiol 1986; 48:335-345.
switched, which would increase safety and minimize the risk 12. Manolopolos VG, Lelkes PI. Cyclic strain and forskolin
of transformation of the cells in situ. These latter more specu- differentially induce cAMP production in phenotypically
lative opportunities remain to be investigated and deserve diverse endothelial cells. Biochem Biophys Res Comm
further efforts. 1993; 191(3):1379-1385.
13. Li J, Menconi MJ, Wheeler B, Rohrer MJ, Klassen VA,
References Ansell JE, Appel MC. Precoating expanded polytetra-
1. Jaffe EA, Nachman RL, Becker CG, Minick CR. Culture fluoroethylene grafts alter production of endothelial cell-
of human endothelial cells derived from umbilical veins. derived thrombomodulators. J Vasc Surg 1992; 15(6):
Identification by morphologic and immunologic criteria. 1010-1017.
J Clin Invest 1973; 52:2745-2756.
Serial Cultivation of Human Endothelial Cells 161
14. Hægerstrand A, Gillis C, Bengtsson L. Serial cultivation 31. Stansby G, Shukla N, Fuller B, Hamilton G. Mesothelial
of adult human endothelium from the great saphenous or endothelial? (Letter; Comment) Eur J Vasc Endovasc
vein. J Vasc Surg 1992; 16(2):280-285. 1995; 10(3):387-388.
15. Gillis C, Bengtsson L, Wiman B, Hægerstrand A. Release 32. Pötzsch B, Grulich HJ, Rössing R, Wille D, Müller BG.
of prostacyclin and tissue plasminogen activator and its Close similarity between cultured human omental me-
inhibitor by cultured adult human endothelial cells grown sothelial cells and endothelial cells in cytochemical mark-
on different matrices. J Eur Vasc Surg, 1996; ers and plasminogen activator production. In Vitro Cell
11(2):127-133. Dev Biol 1991.
16. Weibel E, Palade G. New cytoplasmic components in ar- 33. van Hinsbergh V, Koistra T, Scheffer M, van Bockel J,
terial endothelia. J Cell Biol 1964; 23:101-112. van Muijen G. Characterization and fibrinolytic proper-
17. Weksler BB, Ley CW, Jaffe EA. Stimulation of endothe- ties of human omental mesothelial cells. Blood 1990;
lial cell prostacyclin production by thrombin, trypsin and 75:1490-97.
ionophore A 23187. J Clin Invest 1978; 62:923-930. 34. Coene M-C, Solheid C, Claeys M, Herman AG. Prostag-
18. Newman PJ, Berndt MC, Gorski J. PECAM-1 (CD-31) landin production by cultured mesothelial cells. Arch Int
cloning and relation to adhesion molecules of the immu- Pharmacodyn 1981; 249:316-318.
noglobulin gene superfamily. Science 1990; 247:1219-1222. 35. Zilla P, Fasol R, Deutsch M, Fischlein T, Minar E,
19. Herring M, Gardner A, Glover J. A single stage technique Hammarle A, Krapicka O, Kadketz M. Endothelial cell
for seeding vascular grafts with autogeneous endothelium. seeding of polytetrafluoroethylene vascular grafts in hu-
Surgery 1978; 84(4):498-504. mans: A preliminary report. J Vasc Surg 1987; 6:535-541.
20. Herring MB, Gardner AL, Glover JL. Seeding human ar- 36. Zilla P, Fasol R, Dudeck U, Siedler S, Preiss P, Fischlein
terial prostheses with mechanically derived endothelium. T, Müller-Glauser W, Baitella G, Sanan D, Odell J,
The detrimental effect of smoking. J Vasc Surg 1984; 1: Reichart B. In situ cannulation, microgrid follow-up and
279-289. low-density plating provide first passage endothelial cell
21. Herring MB, Compton RS, LeGrand DR, Gardner AL, mass cultures for in vitro lining. J Vasc Surg 1990;
Madison DL, Glover JL. Endothelial seeding of 12:180-89.
polytetrafluoroethylene poplitetal bypasses: A preliminary 37. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T,
report. J Vasc Surg 1987; 6:114-118. Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I,
22. Jarrell B, Levine E, Shapiro S, Williams S, Carabasi A, Fischlein T. Clinical in vitro endothelialization of
Mueller S, Thornton S. Human adult endothelial growth femoropopliteal bypass grafts: An actuarial follow-up over
in culture. J Vasc Surg 1984; 1:757-64. three years. J Vasc Surg 1994; 19(3):540-548.
23. Jarrell B, Williams S, Stokes G, Hubbard A, Carabasi A, 38. Swedenborg J, Bengtsson L, Clyne N, Gillis C, Rosfors S.
Koolpe E, Greener D, Pratt K, Moritz M, Radomski J, Haegerstrand A. In vitro endothelialization of arterio-
Speicher L. Use of freshly isolated capillary endothelial venous loop grafts for hemodialysis. Eur J Vasc Endovasc
cells for immediate establishment of a monolayer on vas- Surg 1997; 13(3):272-277.
cular graft at surgery. Surgery 1986; 100:392-99. 39. Miyazono K, Takaku F. Platelet-derived growth factors,
24. Jarrell BE, Williams SK, Carabasi RA, Hubbard FA. Im- Blood Rev 1989; 3(4):269-276.
mediate vascular graft monolayers using microvessel en- 40. Burgess W, Maciag T. The heparin-binding (fibroblast)
dothelial cells. In: Herring, ed. Endothelial seeding in vas- growth factor family of proteins. Annu Rev Biochem 1989;
cular surgery. Orlando: Grune & Stratton 1987:37-55. 58:575-606.
25. Williams SK, Wang TF, Castrillo R, Jarrell BE. 41. Partanen J, Vainikka JK, Armstrong E, Alitalo K. Diverse
Liposuction-derived human fat used for vascular graft receptors for fibroblast growth factors. Prog Growth Fac-
sodding contains endothelial cells and not mesothelial cells tor Res 1992; 4:69-83.
as the major cell type. J Vasc Surg 1994; 19(5):916-923. 42. Schreiber AB, Kenney J, Kowalski WJ. Interaction of en-
26. Davison PM, Bensch K, Karasek MA. Isolation and growth dothelial cell growth factor with heparin: Characterization
of endothelial cells from the microvessels of the newborn by receptor and antibody recognition. Proc Natl Acad Sci
foreskin in cell culture. J Invest Dermatol 1980; (USA) 1985; 82(18):6138-6142.
75(4):316-321. 43. Thornton S, Mueller S, Levine E. Human umbilical cells:
27. Grafe M, Auch-Schwelk W, Graf K, Terbeek D, Hertel H, Use of heparin in cloning and long-term serial cultiva-
Unkelbach M, Hildebrandt A.E.F. Isolation and charac- tion. Science 1983; 222:623-625.
terization of macrovascular and microvascular endothe- 44. Minter AJ, Dawes J, Chesterman CN. Effects of heparin
lial cells from human hearts. Am J Physiol 1994; 267:2138- and endothelial cell growth supplement on haemostatic
2148. functions of vascular endothelium. Thromb Haemost
28. Grafe M, Graf K, Auch-Schwelk W, Terbeek D, Hertel H, 1992; 67(6):718-723.
Fleck E. Cultivation and characterization of micro- and 45. Saksela O, Moscatelli D, Sommer A, Rifkin DB. Endothe-
macrovascular endothelial cells from the human heart. Eur lial cell-derived heparan sulphate binds basic fibroblast
Heart J 1993; 14:74-81. growth factor and protects it from proteolytic degrada-
29. Hewett PW, Murray JC. Immunomagnetic purification of tion. J Cell Biol 1988; 107(2):743-751.
human endothelial cells using dynabeads coated with 46. Sommer A, Rifkin DB. Interaction of heparin with hu-
monoclonal antibodies to PECAM-1. Eur J Cell Biol 1993; man basic fibroblast growth factor: Protection of the an-
62(2):451-454. giogenic protein from proteolytic degradation by a gly-
30. Williams SK, Jarrell BE. Cells derived from omental fat cosaminoglycan. J Cell Physiol 1989; 138:215-220.
tissue and used for seeding vascular prostheses are not 47. Klagsbrun M, Baird A. A dual receptor system is required
endothelial in origin: (Letter; Comment). J Vasc Surg for basic fibroblast growth factor activity. Cell 1991;
1992; 15(3):583-5. 67:229-231.
162 Tissue Engineering of Prosthetic Vascular Grafts
48. Gao G, Goldfarb M. Heparin can activate a receptor ty- 64. Watkins M, Sharefkin J, Zajtchuk R, Maciag T, D’Amore
rosine kinase. EMBO J 1995; 14(10):2183-2190. P, Ryan U, van Wart H, Rich N. Adult human saphenous
49. Gillis C, Jonzon B, Haegerstrand A. Effects of sera, bFGF, vein endothelial cells: Assessment of their reproductive
heparin and cyclic AMP stimulation on proliferation of capacity for use in endothelial seeding of vascular pros-
human umbilical and saphenous vein endothelial cells. theses. J Surg Res 1985; 36(6):588-596.
Cell Mol Biol 1995; 41:1131-1138. 65. Hackett SF, Friedman Z, Campochiaro PA. Cyclic 3', 5'-
50. D’Amore PA. Mechanisms of endothelial growth control. adenosine monophosphate modulates vascular endothe-
Am J Respir Cell Mol Biol 1992; 6(1):1-8. lial cell migration in vitro. Cell Biol Int Rep 1987;
51. Boes M, Dake BL, Bar RS. Interactions of cultured en- 11(4):279-287.
dothelial cells with TGF-beta, bFGF, PDGF and IGF-I. Life 66. Klein-Soyer C, Archipoff G, Beretz A, Cazenave JP. Op-
Sci 1991; 48(8):811-21. posing effects of heparin with TGF-beta or afgf during
52. Schweigerer L, Neufeld G, Friedman J. Capillary endot- repair of a mechanical wound of human endothelium.
helial cells express basic fibroblast growth factor, a mito- Influence of cAMP on cell migration. Biol Cell 1992;
gen that promotes their own growth. Nature 1987; 75(2):155-162.
325:257-259. 67. Ogawa S, Koga S, Kuwabara K. Hypoxia-induced increased
53. DiCorleto PE, Bowen-Pope DF. Cultured endothelial cells permeability of endothelial monolayers occurs through
produce a platelet-derived factor-like protein. Proc Natl lowering of cellular cAMP, Am J Physiol 1992; 262(3 Pt
Acad Sci (USA) 1983; 80:1919-1923. 1):C 546-554.
54. Vlodavsky I, Friedman R, Sullivan R, Sasse J, Klagsbrun 68. Archipoff G, Beretz A, Bartha K, Brisson C, de la Salle D,
M. Aortic endothelial cells synthesize basic fibroblast Froget-Leon C, Klein-Soyer C, Cazenave JP. Role of cy-
growth factor which remains cell associated and platelet- clic AMP in promoting the thromboresistance of human
derived growth factor-like protein which is secreted. J Cell endothelial cells by enhancing thrombomodulin and de-
Physiol 1987; 131(3):402-408. creasing tissue factor activities. Br J Pharm 1993;
55. Boynton AL, Whitfeld JF. The role of cAMP in cell pro- 109:18-28.
liferation: A critical assessment of the evidence. Adv Cy- 69. Graier WF, Groschner K, Schmidt K, Kukovetz WR. In-
clic Nucleotide Res 1983; 15:193-294. creases in endothelial cAMP levels amplify agonist-induced
56. Dumont J, Jauniaux J-C, Roger P. The cyclic AMP-medi- formation of endothelium derived relaxing factor (EDRF).
ated stimulation of cell proliferation. TIBS 1989; 14:67-71. Biochem J 1992; 288(Pt 2):345-349.
57. Moncada S, Vane JR. Pharmacology and endogenous roles 70. Sung C-P, Arleth A, Shikano K, Zabko-Potapovich B,
of prostaglandin endoperoxides, thromboxane A2 and Berkowitz B. Effects of trypsination in cell culture on
prostacyclin. Pharmacol Rev 1979; 30:292-331. bradykinin receptors in vascular endothelial cells. Bio
58. Green H. Cyclic AMP in relation to proliferation of the Pharm 1989; 38:696-99.
epidermal cell: A new view. Cell 1978; 15:801-811. 71. Flugelman MY, Virmani R, Leon MB, Bowman RL,
59. Davison P, Karasek M. Human dermal microvascular en- Dichek DA. Retroviral vector-mediated gene transfer into
dothelial cells in vitro: Effect of cyclic AMP on cellular endothelial cells. Mol Biol Med 1991; 8(2):257-66.
morphology and proliferation rate. J Cell Physiol 1981; 72. Newman KD, Nguyen N, Dichek DA. Enhancement of the
106:253-58. fibrinolytic activity of sheep endothelial cells by retroviral
60. Olashaw NE, Pledger WJ. Cellular mechanisms regulat- vector-mediated gene transfer. Blood 1991; 77(3):533-41.
ing proliferation. Advances in second messenger and phos- 73. Lee SW, Kahn ML, Dichek DA. Optimization of retroviral
phoprotein research. 1988; 22:139-173. vector-mediated gene transfer into endothelial cells in
61. Kramer R, Fuh G-M, Karachek M. Type IV collagen syn- vitro. Circ Res 1992; 71(6):1508-17.
thesis by cultured human microvascular endothelial cells 74. Dichek DA, Nussbaum O, Degen SJ, Anderson WF. Ge-
and its deposition into the subendothelial basement mem- netically engineered endothelial cells remain adherent and
brane. Biochemistry 1985; 24:7423-30. viable after stent deployment and exposure to flow in
62. Tuder RM, Karasek MA, Bensch KG. Cyclic adenosine vitro. Circ Res 1992; 70(2):348-54.
monophosphate levels and the function of skin microvas- 75. Moldovan F, Bennani H, Fiet J, Cussenot O, Dumas J,
cular endothelial cells. J Cell Physiol 1990; 142(2):272-283. Darbord C, Soliman HR. Establishment of permanent
63. Haegerstrand A, Dalsgaard C-J, Jonzon B, Larsson O, human endothelial cells achieved by transfection with
Nilsson J. Calcitonin gene-related peptide stimulates pro- Large T antigen that retain typical phenotypical and func-
liferation of human endothelial cells. Proc Natl Acad Sci tional characteristics. In vitro Cell Dev Biol Anim 1996;
1990; 87(9):3299-3303. 32(1):16-23.
CHAPTER 15
Risk Factors for Autologous Endothelial Cell Cultures
Johann Meinhart, Manfred Deutsch, Peter Zilla
Introduction
T he history of cell culture started over a hundred years ago, when Wilhelm Roux showed
in 1885 that embryonic cells can survive outside the animal body. Since then the knowl-
edge of how to culture cells in vitro has steadily increased. Today, cell cultures are a powerful
tool to address many fundamental questions in biology and medicine. Recent advances have
made it possible to utilize cultured human cells for therapeutic use. Cultured cells have been
used to reconstruct skin, bone and many other tissues, including the endothelium. As a
consequence, endothelial cells can be used clinically for the physiological masking of syn-
thetic vascular grafts in order to reduce their thrombotic risk. Several studies were performed
in past years with different methods1-7 to achieve endothelialization of otherwise nonhealing
grafts. The presence of endothelial cells on synthetic vascular grafts reportedly diminishes
the thromboembolic risk8,9 and it seems to inhibit the formation of anastomotic hyperpla-
sia.10 Additionally, endothelial cell-covered grafts are probably less prone to graft infection.11
One method of actively facilitating graft endothelialization is in vitro lining with cultured
autologous endothelial cells.12-14 This technique could prove its effectiveness in long term
clinical applications.15-19 However, despite optimized culture and harvesting procedures,
cultures may fail to grow. Since reproducibility of viable autologous cell cultures is a prereq-
uisite for routine clinical use, we have investigated the influence of certain patient-related
risk factors on the growth capability of autologous endothelial cells.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
164 Tissue Engineering of Prosthetic Vascular Grafts
index (“p” index) of a given cell culture area ranging from 0 tion, by their typical morphological appearance. However,
(no cells) to 1.0 (confluent cell culture). Thus, the p index as more information on clinical long term results emerges,
semiquantitatively reflects the growth capacity of primary smooth muscle cells and fibroblasts appear in a different
endothelial cells in culture. light, as they play a substantial role in vessel wall forma-
Apart from the evaluation of endothelial cell growth, tion.18,25 In future, one will probably tend to have a mixture
cultures are also morphologically assessed. Vacuolization and of vascular cells seeded onto the graft surface, which would
cell shape are classified on a scale ranging from 0 (typical de facto mean a convergence of culture and mass harvest
polygonal) to 5 (dendritic) for cell shape and from 0 (no procedures.
vacuoles) to 5 (predominantly vacuoles) for vacuolization. After successful growth of primary cultures, trypsin is
The morphology index (“m”) is therefore m=0 for ideal the standard enzyme for the detachment of cells from the
polygonal, nonvacuolized endothelial cells and m=10 for surface of culture flasks in order to passage them into larger
maximally vacuolized, highly dendritic but still viable cells. containers. Trypsinization, especially of primary human
Continuous light microscopic assessment of morpho- endothelial cell cultures, should be performed very carefully.
logical appearance and quantification of cultures provides a When using 37°C 0.05% trypsin-0.02% EDTA solution, in-
good estimation of cell viability and proliferative capacity. cubation time should not exceed 2 minutes.23
Thus, mitigation of cell growth can be detected at an early For endothelial cell cultures, different media like
stage. The most frequent problems with autologous RPMI, MEM or medium 199 have been used by different
endothelial cell cultures will be described in the authors, with satisfactory results. Medium 199 was originally
following chapters. formulated for the growth of chick fibroblasts, but with Earl’s
salts is now widely used for endothelial cell culture. Liquid
Procedure Related Risk Factors media have a limited shelf life due to the decay of several
compounds over time, even at 4°C. This is especially true
Surgical Vein Harvest for L-glutamine. Complete medium should therefore not
Naturally, autologous human venous endothelial cell be kept at 37°C for a prolonged time, since this will result in
cultures need a sufficient vein segment as a cell source. This pH and osmolality change. Moreover, free radical forma-
vein should be easily accessible for surgical excision in order tion might occur, which would result in severe cell damage.
to keep dissection time short and, therefore, mechanical Substitution of the culture medium with growth fac-
damage low. To further reduce premature mechanical de- tors is necessary to reach optimal proliferation, even when
tachment of endothelial cells, a no touch technique for vein serum is supplemented. A multitude of growth factors for
harvesting is in use.23 In brief, after careful touch-free dis- endothelial cells, like PDGF, EGF and FGFs have been iden-
section, the vessel is cannulated in situ with two free-flow tified and have been described in numerous publications.
vessel cannulas with attached 3-way taps. In order to remove For standard endothelial cell culture, bFGF is the most
remaining blood, the vein is flushed with culture medium frequently used growth factor. The importance of FGF ex-
in a pulsatile manner and filled with a collagenase solution. ceeds mere acceleration of proliferation, as total FGF depri-
The solution is kept under minor pressure within the vein vation in the culture medium also leads to apoptosis of
by closing the 3-way taps. After excision, the cannulated and endothelial cells.26
collagenase-filled vein segment is transferred to the labora- Supplementation with antibiotics might be necessary,
tory in a thermos container at 37°C for further incubation. but they should be used with great care, due to their poten-
If cannulation takes too long, the endothelium might be tial cytotoxicity. Microbial contamination can occur from
damaged by air drying. Thus, if cannulation is difficult due time to time. Serious infections with yeasts, molds and bac-
to a small vessel diameter or other circumstances, the vein teria are macroscopically visible in cultures. However, chroni-
should be flushed intermittently with medium. All manipu- cally infected cultures can be inconspicuous, often only dis-
lations with the vein should be done gently, wearing starch tinguished by decreased cell proliferation. Therefore, micro-
free gloves, since glove powder has been found to be cyto- biological tests should be routinely performed for clinically
toxic for endothelial cell cultures.24 used cultures. Decreased proliferation capacity without sub-
stantial morphological deterioration might also be an indi-
Cell Culture cation for an infection with mycroplasms. Several test kits
A 15 minute incubation time with a 0.1% collagenase are available for mycoplasm detection. In contrast, the de-
solution guarantees a maximal yield of endothelial cells. tection of viruses in cell cultures is more difficult than that
Since all proteolytic enzymes are potentially cytotoxic to cells of bacteria or fungi. Detection methods include coculture
and commercially available collagenases vary in activity, it systems, transmission electron microscopy and immunof-
might be necessary to test each vial separately. Since the ini- luorescence. Some viruses cause cytological changes like cell
tial goal of in vitro endothelialization was to use only pure rounding, vacuolization or syncytium formation. Since these
endothelial cell cultures for the lining of synthetic vascular morphological changes are often similar to other deterio-
prostheses, we tried to avoid the concomitant harvesting of rating influences like cell aging or increased lipid content of
undesirable cells like fibroblasts and smooth muscle cells by serum, the existence of viruses should be kept in mind.27
fine tuning the enzymatic digestion time and enzymatic ac- Standard cell culture flasks made of polystyrene are
tivity. These supposedly “contaminating” cells can be de- surface treated to promote adhesion and proliferation of
tected by immunofluorescence or, in a later stage of cultiva- attachment dependent cells. Endothelial cells cultured in
Risk Factors for Autologous Endothelial Cell Cultures 165
20% autologous serum readily adhere to polystyrene with enrolled in our endothelialization of vascular grafts program
subsequent spreading and growth. This might be facilitated already had quite a long history of vascular disease before
by serum vitronectin and serum fibronectin.28 The role of they had to undergo surgical reconstruction for peripheral
fibronectin as a promoter of cell migration and tight an- arterial occlusion. They were treated conservatively and
chorage of endothelial cells to many biomaterials has been many of them were already operated for other vascular dis-
described extensively.29-31 Therefore, polystyrene culture eases like coronary heart disease or occlusion of the iliac ar-
dishes are frequently precoated with fibronectin, but other tery. A substantial number had already undergone PTA (per-
adhesion substances like collagen or gelatin are also used. cutaneous transluminal angioplasty) before they had to be
The quality of practically all reagents varies from sup- treated surgically. The majority of patients had one or more
plier to supplier. Additionally, quality might even be different risk factors for atherosclerosis. Therefore, these patients
between batches from the same supplier. These facts should formed a quite homogenous group with respect to age, risk
be kept in mind when encountering cell culture problems. factors and clinical stage of disease. Despite this fact, we
Defective machines, like centrifuges, rotation devices and found differences with regard to the in vitro growth behav-
incubators, might also lead to culture problems and should ior of the endothelial cells of these patients. Thirty-four per-
be mentioned briefly here. Above all, incubators should be cent of all cultures showed impaired growth capacity. Cells
checked for accurate temperature, humidity and CO2. In- displayed strong vacuolization and were partially dendritic
cubators might also be a source of repeated contaminations. (Fig. 15.1), a morphological appearance which was previ-
ously shown to lead to imminent cell death.16 When we
Patient Related Risk Factors measured a multitude of serum parameters we found all
Influences of age or genetic constitution on cell vi- three main lipid constituents, triglycerides, cholesterol and
ability and proliferative capacity are well known. In the car- lipoprotein A, significantly (p < 0,05) elevated in sera of pa-
diovascular system, age-associated alterations in vascular cell tients whose cell cultures initially failed to grow (Table 15.1).
function lead to vessel wall thickening, increased dilation Numerous studies have previously shown that hyperlipi-
and aberrant distribution of smooth muscle cells. It is be- demia causes endothelial changes. This includes increased
lieved that these changes play a fundamental role in the on- endocytotic activity34 inhibition of prostacyclin produc-
set of atherosclerosis.33 Besides these intrinsic factors, ac- tion,35 increased monocyte-endothelial cell adhesion,36 dis-
quired factors may alter cellular functions as well. Patients ruption of the barrier function, 37 apoptosis 38 and
Table 15.1. Content of cholesterol, triglycerides, lipoprotein A and glucose in sera of patients with normal growth
characteristics of cells compared to sera of patients with initial growth failure of cells and to nonrecovering cell
cultures after serum change.
cytotoxicity.39 From experimentally induced rabbit athero- that reversibility applied especially to elevated triglyceride
sclerotic aortas, an increased number of multinucleated gi- and cholesterol levels. The fact that those cultures which ir-
ant endothelial cells have been harvested and cultured,40 a reversibly failed to grow had unproportionally increased lev-
finding quite similar to our own observation. Moreover, li- els of lipoprotein A might be an indication that this serum
poprotein A has previously been found to be an indepen- lipid has a particularly damaging effect on endothelial cells.
dent risk factor for atherosclerosis.41
Apart from hyperlipidemia, diabetes is another major Summary and Future Aspects
risk factor for pathological changes of the vasculature. On The successful cultivation of autologous endothelial
the cellular basis it has been shown that endothelial cells cells is endangered by both procedure dependent and pa-
cultured in the presence of high glucose levels have a re- tient dependent risk factors. Reasons for failing cultures due
duced proliferative capacity. Accelerated endothelial cell to procedure-related problems are mechanical damage dur-
death42 is believed to be due to facilitated apoptosis,43 but ing vein dissection, contaminations and inadequate reagents.
may also have osmotic causes. In this context it was interest- However, the most frequent problems with autologous en-
ing that we found higher glucose levels in the serum of pa- dothelial cell cultures are related to patient factors. Most of
tients with normally growing cultures than in sera of pa- the patients who have to undergo bypass surgery have a
tients whose cultures initially failed to grow. Therefore, multitude of major risk factors. Numerous clinical and ex-
moderately increased glucose does not seem to affect viabil- perimental studies have revealed smoking, diabetes and
ity and growth capacity of primary endothelial cell cultures. hyperlipidemia to be major independent risk factors for ath-
The third well established risk factor for the vascula- erosclerosis. The additive detrimental effect of these risk fac-
ture is smoking tobacco. Its negative influence on endothe- tors on the vasculature has been further documented.47-49
lial cells has previously been described.44 Particularly, a lower Our data seem to reflect these findings in vitro. We found
harvest efficiency and a suppressed reproductive capacity significantly different contents of cholesterol, triglycerides,
has been reported for autologous endothelial cell cultures lipoprotein A and glucose in the sera of patients with initial
from smokers.45 This is in accordance with our finding that growth failure as compared to sera of patients with normal
the mean number of harvested cells from smokers was sig- primary growth. However, by replacing the patient’s own
nificantly lower in smokers than nonsmokers. However, since serum as a culture supplement with pooled serum from
the subsequent proliferative behavior of endothelial cells did healthy young donors, the majority of cultures recovered,
not significantly differ, our observation supports the previ- regaining their normal morphological appearance and full
ous interpretation that a considerably thicker basement proliferative capacity. Thus, the success rate of autologous
membrane may be responsible for a stronger endothelial cell endothelial cells cultured for the endothelialization of syn-
attachment in smokers.46 thetic vascular grafts was increased from the previous
However, in contrast to other risk factors influencing 73-95%. This improvement, together with a further simpli-
the successful growth of autologous endothelial cells, the fication of the seeding process, makes in vitro
damaging effects of hyperlipidemia can be reversed. When endothelialization even more clinically practicable. The ben-
serum is changed from autologous to pooled homologous eficial long term results in peripheral revascularization and
serum in cultures with morphological signs of growth fail- the first promising data for clinically used endothelialized
ure, all cultures, with a few exceptions, regain their prolif- synthetic coronary bypass grafts50 provide encouraging ar-
erative capacity together with a typical morphological ap- guments for a wider application.
pearance (Figs. 15.2 and 15.3). It was also interesting to see
It is quite obvious and desirable that tissue engineer- 2. Graham LM, Burkel WE, Ford JW et al. Immediate seed-
ing is directing its effort towards fascinating goals like high ing of enzymatically derived endothelium in dacron vas-
tech, ready to implant grafts with features of a native artery. cular grafts. Arch Surg 1980; 115:289-94.
However, the fact that a substantial number of patients will 3. Zilla P, Fasol R, Deutsch M et al. Endothelial cell seeding
of polytetrafluoroethylene vascular grafts in humans: A
have an impaired endothelium, not able to regenerate or
preliminary report. J Vasc Surg 1987; 6:535-41.
fulfill necessary physiological functions, should be consid- 4. Meerbaum SO, Sharp WV, Schmidt SP. Lower extremity
ered when discussing these future approaches. Until the suc- revascularization with polytetrafluoroethylene grafts
cessful introduction of a fully engineered vascular prosthesis seeded with microvascular endothelial cells. In Zilla P,
which emulates native arteries structurally and functionally, Fasol R, Callow A, eds. Applied Cardiovascular biology
in vitro endothelialization will remain a feasible method of 1990-1991. Basel: Karger, 1992:107-119.
improving the performance of synthetic vascular grafts. 5. Williams SK, Rose DG, Jarrell BE. Microvascular endo-
Centers of excellence could provide endothelialized grafts thelial cell sodding of ePTFE vascular grafts: Improved
to a number of neighboring hospitals. This prospect is even patency and stability of the cellular lining. J Biomed Mat
more exciting in view of the possibility to genetically modify Res 1994; 28:203-212.
endothelial cells.51 Moreover, endothelialization could be 6. Pasic M, Müller-Glauser W, Odermatt B et al. Seeding
with omental cells prevents late intimal hyperplasia in
used as a generalized hemocompatible drug delivery system
small-diameter Dacron grafts. Circulation 1995;
which enables the creation of individual delivery schemes 92:2605-16.
for each single patient. Vascular grafts and endothelialization 7. Greisler HP, Cziperle DJ, Kim DU et al. Enhanced
could also be used for the development of various tissue endothelialization of expanded polytetrafluoroethylene
engineered organs. Different cell types, allogenic or grafts by fibroblast growth factor pretreatment. Surgery
xenogenic, could be seeded on the graft surface. These cells 1992; 112:244-54.
would be subsequently covered confluently by autologous 8. Örtenwall P, Wadenvik H, Risberg B. Reduced platelet
endothelial cells, in order to inhibit the immune response. deposition on seeded versus unseeded segments of ex-
Thus some fascinating prospectives still inhere panded polytetrafluoroethylene grafts: Clinical observa-
to endothelialization. tions after a 6-month follow-up. J Vasc Surg 1989;
10:374-380.
9. Örtenwal P, Wadenvik H, Kutti J. Endothelial cell seed-
References
ing reduces thrombogeneicity of dacron grafts in humans.
1. Herring MB, Gardner AL, Glover JL et al. A single staged
J Vasc Surg 1990; 11:403-10.
technique for seeding vascular grafts with autogenous
10. Graham LM, Brothers TE, Vincent CK et al. The role of
endothelium. Surgery 1978; 4:498-504.
an endothelail cell lining in limiting distal anastomotic
168 Tissue Engineering of Prosthetic Vascular Grafts
intimal hyperplasia of 4-mm-i.d.dacron grafts in a canine 26. Araki S, Shimada Y, Hayashi H. Apoptosis of vascular
model. J Biomed Mat Res 1991; 25:525-533. endothelial cells by fibroblast growth factor deprivation.
11. Rosenman JE, Kempczinski RF, Berlatzky Y et al. Bacterial Biochem Biophys Res Comm 1990; 168:1194-1200.
adherence to endothelial-seeded polytetrafluoroethylene 27. Cell and Tissue Culture: Laboratory Procedures. In: Doyle
grafts. Surgery 1985; 98:816-823. A, Griffiths JB, Newell DG, eds. Chichester: John Wiley
12. Zilla P, Fasol R, Dudeck U, Siedler S, Preiss P, Fischlein & Sons Ltd, 1993; 7:7A-7C.
T, Müller-Glauser W, Baitella G,Sanan D, Odell J, Reichart 28. Steele JG, Johnson G, Underwood PA. Role of serum
B. In situ cannulation, microgrid follow-up and low den- vitronectin and fibronectin in adhesion of fibroblasts fol-
sity plating provide first pasage endothelial cell mass cul- lowing seeding onto tissue culture polystyrene. J Biomed
tures for in vitro lining. J Vasc Surg 1990; 12:180-189. Mat Res 1992; 26:861-884.
13. Zilla P, Fasol R, Preiss P, Kadletz M, Deutsch M, Schima 29. Keseler KA, Herring MB, Arnold MP et al. Enhanced
H, Tsangaris S, Groscurth P. Use of fibrin glue as a sub- strength of endothelial cell seeding using expanded
strate for in vitro endothelialization of PTFE vascular polytetrafluoroethylene vascular grafts: A comparison of
grafts. Surgery 19989; 105:515-522. four substrates. J Vasc Surg 1986; 3:58-64.
14. Zilla P, Preiss P, Groscurth P, Rösemeier F, Deutsch M, 30. Vohra R, Thomson JL, Carr HMH et al In vitro aherence
Odell J, Heidinger C, Fasol R, von Oppell U. In vitro- and kinetics studies of adult human endothelial cell seeded
lined endothelium: Initial integrity and ultrastructural polytetrafluoroethylene and gelatin impregnated Dacron
events. Surgery 1994; 116:524-534. grafts. Eur J Vasc Surg 1991; 5:93-103.
15. Magometschnigg H, Kadletz M,Vodrazka M et al. Pro- 31. Poot A, Beugeling T, Dekker A, Spijkers J et al. Depen-
spective clinical study with in vitro endothelial cell lining dence of endothelial cell growth on substrate bound
of expanded polytetrafluoroethylene grafts in crural re- fibronectin. Clinical Materials 1992; 11:151-155.
peat reconstruction. J Vasc Surg 1992; 15:527-35. 32. Carr HM, Vohra R, Sharma H et al. Endothelial cell ki-
16. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T, netics under chronic flow in prosthetic grafts. Ann Vasc
Minar E, Dudczak R, Lugmaier H, Schmidt P, Noscian I, Surg 1996; 10:469-75.
Fischlein T. Clinical in vitro endothelialization of 33. Crow MT, Boluyt MO, Lakatta EG. The molecular and
femoropopliteal bypass graft: An actuarial follow-up over cellular biology of aging in the cardiovascular system. In:
three years. J Vasc Surg 1994; 19:540-548. Holbrook NJ, Martin GR, Lockshin RA, eds. Cellular ag-
17. Fischlein T, Zilla P, Meinhart J, Puschmann R, Vesely M, ing and cell death. Chichester: John Wiley & Sons,
Eberl T, Balon R, Deutsch M. In vitro endothelialization 1996:81-107.
of a mesosystemic shunt: A clinical case report. J Vasc 34. Holland JA, Pritchard KA, Rogers NJ et al. Atherogenic
Surg 1994: 19:549-554. levels of low density lipoprotein increase endocytotic ac-
18. Deutsch M, Meinhart J, Vesely M, Groscurth P, Von tivity in cultured human endothelial cells. Am J Pathol
Oppell U, Zilla P. In vitro endothelialization of ePTFE 1992; 140:551-558.
grafts: A clinical case report after 41 months of implanta- 35. Peng SK, Hu B, Peng A et al. Effect of cholesterol oxides
tion. J Vasc Surg 1997; 25:757-763. on prostacyclin production and platelet adhesion. Artery
19. Meinhart J, Deutsch M, Zilla P Eight years of clinical en- 1993; 20:122-134.
dothelial cell transplantation. Closing the gap between 36. De Gruijter M, Hoogerbrugge N, van Rijn M et al.
prosthetic grafts and vein grafts. ASAIO J 1997; Patients with combined hypercholesterolemia-hyper-
43:M515-M521. triglyceridemia show an increased monocyte-endothelial
20. Jaffe E, Nachmann R, Becker C et al. Culture of human cell adhesion in vitro: Triglyceride level as a major deter-
endothelial cells derived from umbilical minant. Metabolism 1991; 40:1119-1121.
veins.Identification by morphological and immunological 37. Guretzki HJ, Gerbitz KD, Olgemöller B et al. Atherogenic
criteria. J Clin Invest 1973; 52:2757-2764. levels of low density lipoprotein alter the permeability and
21. Haegerstrand A, Gillis C, Bengtsson L et al. Serial culti- composition of the endothelial barrier. Atherosclerosis
vation of adult human endothelium from the great saphe- 1994; 107:15-24.
nous vein. J Vasc Surg 1992; 16:280-285. 38. Lizard G, Deckert V, Dubrez L et al. Induction of
22. Gillis C, Jonzon B, Haegerstrand A. Effects of sera, basic apoptosis in endothelial cells treated with cholesterol ox-
fibroblast growth factor, heparin and cyclic AMP-stimu- ides. Am J Pathol 1996; 148:1625-1638.
lation on proliferation of human vascular endothelial cells. 39. Sevanian A, Berliner J, Peterson H. Uptake, metabolism,
Cell Mol Biol 1995; 41:1131-1138. and cytotoxicity of isomeric cholesterol-5,6-epoxides in
23. Zilla P, Fasol R, Dudeck U, Siedler S, Preiss P, Fischlein rabbit aortic endothelial cells. J Lipid Res 1991; 32:147-
T, Müller-Glauser W, Baitella G, Sanan D, Odell J, 155.
Reichart B. In situ cannulation, microgrid follow-up and 40. Tashiro K, Shimokama T, Haraoka S et al. Endothelial
low density plating provide first passage endothelial cell cell heterogeneity in experimentally-induced rabbit ath-
mass cultures for in vitro lining. J Vasc Surg 1990; erosclerosis. Demonstration of multinucleated giant en-
12:180-189. dothelial cells by scanning electron microscopy and cell
24. Sharefkin JB, Fairchild KD, Albus RA et al. The cytotoxic culture. Virchows Archiv 1994; 425:521-529.
effects of surgical glove powder particles on adult human 41. Schreiner PJ, Morrisett JD, Sharrett R et al. Lipoprotein
vascular endothelial cell cultures: Implications for use of (a) as a risk factor for preclinical atherosclerosis. Arterio-
tissue culture techniques. J Surg Res 1986; 41:463-72. sclerosis and Thrombosis 1993; 13:826-833.
25. Baitella-Eberle G, Groscurth P, Zilla P et al. Long term 42. Cagliero E, Roth T, Taylor AW, Lorenzi M. The effect of
results of tissue development and cell differentiation on high glucose on human endothelial cell growth and gene
Dacron prostheses seeded with microvascular cells in dogs. expression are not mediated by transformtin growth fac-
J Vasc Surg 1993; 18:1019-28. tor-b. Lab Investig 1995; 73:667-673.
Risk Factors for Autologous Endothelial Cell Cultures 169
43. Baumgartner-Parzer S, Wagner L, Pettermann M et al. 48. Pech-Ansellem MA, Myara I, Storogenko M et al. En-
High-glucose-triggered apoptosis in cultured endothelial hanced modification of low-density lipoproteins (LDL) by
cells. Diabetes 1995; 44:1323-1327. endothelial cells from smokers: A possible mechanism of
44. Strohschneider T, Oberhoff M, Hanke H, Hannekum A, smoking-related atherosclerosis. Cariovascular Research
Karsch KR. Effect of chronic nicotine delivery on the pro- 1996; 31:975-983.
liferation rate of endothelial and smooth muscle cells in 49. Blache D, Bouthillier D, Davignon J. Acute influence of
experimentally induced vascular wall plaques. Clin Investig smoking on platelet behaviour, endothelium and plasma
1994; 72:908-912. lipids and normalization by aspirin. Atherosclerosis 1992;
45. Zilla P, Siedler S, Fasol R et al. Reduced reproductive ca- 93:179-188.
pacity of freshly harvested endothelial cells in smokers: A 50. Laube HR, Duwe J, Claus M et al. Autologous endothe-
possible shortcoming in the success of seeding? J Vasc lial cell seeded PTFE vascular grafts for coronary artery
Surg 1989; 10:143-8. bypass: First clinical results. 15th International Cardiovas-
46. Asmusen I, Kjelsen K. Intima ultrastructure of human cular Surgical Symposion. Zürs: Austria, 1997, March
umbilical arteries: Observations of arteries from newborn 8-15.
children of smoking and non-smoking mothers. Circ Res 51. Dichek DA. Therapeutic potential of genetic engineering:
1975; 36:579-589. Enhancement of endothelial cell fibrinolysis. In: Zilla P,
47. Bartens W, Rader D, Talley G et al. Decreased plasma lev- Fasol R, Callow A, eds. Applied vardiovascular Biology
els of lipoprotein (a) in patients with hypertriglycerdemia. 1990-1991. Basel: Karger, 1992:197-204.
Atherosclerosis 1994; 108:149-157.
CHAPTER 16
Adhesion Molecule Expression Following In Vitro Lining
Caroline Gillis-Hægerstrand
Introduction
T he possibility that human ECs can be seeded on vascular prosthetic grafts to create a
“look-alike” to the natural blood vessel is intriguing. The new field of tissue engineering
has awakened an interest in many scientists from different disciplines which makes the re-
search area fascinating. Important breakthroughs have greatly enhanced our understanding
of the functions of the human endothelial cell. Today it is well accepted that the endothe-
lium is indeed a very active “organ”. The endothelium is crucial in maintenance of hemosta-
sis, coagulation and fibrinolysis, as well as in the regulation of vascular tonus, inflammation,
immune reactions, angiogenesis and vascular permeability.1-6
Long term patency of artificial vascular grafts for hemodialysis access and for bypass
or interposition in small caliber arteries is limited due to neointimal hyperplasia and associ-
ated graft thrombosis. If these grafts could be seeded with well functioning, adherent ECs
which are not activated and expresses adhesion molecules to which leukocytes can adhere,
these problems could partly be overcome. The same reasoning is valid for biological heart
valves.
In this chapter, I will try to cover aspects of how the endothelium interacts with circu-
lating leukocytes and how an endothelial lining of artificial, cell free materials may effect the
adherence of leukocytes.
Vascular Grafts
There are different kinds of grafts to be used for different purposes in vascular surgery.
Autologous Grafts
Autologous vessels are still the preferred vascular substitute. They are superior to all
other available alternatives, although it has been demonstrated that surgical preparation
partly denudes the vessels and damages the endothelium.7 A complete re-endothelialization
after implantation of these grafts has been shown.8 The obvious drawback to using autolo-
gous vessels is the limited supply. Another problem is intimal hyperplasia, which is thought
to be initiated when the endothelium is damaged during the operation. Hyperplasia is a
major reason for re-operation or repeated PTCA procedures after open heart surgery and
peripheral vascular surgery.
Prosthetic Grafts
One feature that distinctly distinguishes the cardiovascular prosthetic graft from its
natural counterpart is its lack of a confluent endothelium. The development of the ideal
prosthetic vascular graft has been one of the major goals of vascular surgery since the first
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
172 Tissue Engineering of Prosthetic Vascular Grafts
grafts were used over 40 years ago. The aim has been to Inflammation
achieve a completely endothelialized prosthetic graft where Inflammation is the reaction of living tissues to all
the cells not only remain when the blood flow is restored, forms of injury. It involves vascular, neurologic, humoral
but also retain their wide range of functions. Synthetic grafts and cellular responses at the site of injury. The adherence of
have acceptable patency rates when used in areas of high neutrophils and monocytes to the vascular endothelium is a
blood flow and good run-off to replace vessels of an inner hallmark of acute inflammation. At sites of inflammation,
diameter greater than 5-6 mm.9 When used in low flow ar- vascular ECs undergo functional and morphological changes
eas, the expectations regarding patency are not currently collectively referred to as endothelial activation. 19,20
being fulfilled. The two types of synthetic vascular grafts that Cytokines such as IL-121 and TNF-#22 induce functional
have gained the widest acceptance are Dacron and ePTFE. changes in human ECs in vitro, including increased adhe-
Glutaraldehyde-tanned homologous grafts are another al- siveness for leukocytes.23 In vivo models have also shown
ternative but their use is under debate. that cytokines induce changes corresponding to endothelial
activation when introduced to tissues.24 When stimulated
Dacron with histamine, thrombin or superoxides the ECs rapidly
The manufacturing process allows defined variation express P-selectin for approximately 20 min. When activated
of porosity. The pores are larger in Dacron grafts than in by cytokines such as IL-1 or TNF-#, the endothelium ex-
ePTFE grafts. After Dacron graft implantation, platelet ad- presses an array of inducible adhesion molecules, e.g.,
hesion and aggregation occurs, followed by fibrin forma- P-selectin, E-selectin, VCAM-1 and ICAM-1 (Fig. 16.1).
tion. Then there is a gradual ingrowth of fibroblasts and The improvement of cardiovascular prosthetic mate-
capillaries, and a deposition of matrix proteins. A fibrous rials, to reduce thrombus formation and inflammatory/im-
tissue is formed on the inner lining of the graft as capillaries munological reactions, is a technological as well as a bio-
grow from the exterior to the interior. logical challenge. By mediating adhesion and by expressing
adhesion molecules to which leukocytes adhere, ECs play a
ePTFE key role in inflammatory reactions. The process of leuko-
ePTFE grafts are made by controlled expansion of the cyte adherence, activation and migration involves an inter-
material by heating. The initial thrombus formation on the play between ECs, leukocyte activation and local cytokine
inner surface of ePTFE grafts when platelets adhere and ag- activity. An activated leukocyte is likely to adhere to the en-
gregate is similar to that in Dacron grafts. However, the dothelium even if the endothelium itself is expressing a “nor-
porosity is much less than that in Dacron grafts, which pre- mal” range of adhesion-promoting molecules on its surface.
vents ingrowth of fibrous tissue and capillaries. No or little A firm adhesion and transmigration into the subendothe-
spontaneous ingrowth of ECs from anastomoses occurs lial tissue requires an interplay between the leukocyte and
in man.10 the ECs, where activation and structural changes occur in
the endothelium. Although the neutrophil is considered
Glutaraldehyde-Tanned Grafts more reactive and has been most extensively studied in this
Glutaraldehyde treatment is implemented to reduce respect, the monocyte expresses a similar range of adhesion
antigenicity, sterilize the graft and stabilize the tissue by molecules as do neutrophils and respond to similar stimuli.
crosslinking of collagen.11 Two of the side effects of this treat- T lymphocytes are likely to play an important role in the
ment are that calcification of the tissue is promoted and a body´s reaction to a foreign material. Less is known about T
spontaneous ingrowth of ECs is prohibited.12,13 The hostile lymphocyte-endothelial interactions.
environment to any adhering or ingrowing cell type due to Endothelial cells express three families of adhesion
toxic residues of glutaraldehyde may delay an inflammatory/ molecules which interact with leukocytes: selectins, immu-
foreign body reaction to the graft. However, inflammatory noglobulins and integrins.
cells, macrophages and T cells in the tissue at explantation
have been described by several groups.14 Selectins
Selectins are cell surface glycoproteins found on the
Heart Valve Prostheses endothelium, leukocytes and platelets. Selectins binds to
Mechanical heart valves, or heart valve bioprostheses, carbohydrate rich domains on white blood cells. Platelet
similar to the native are frequently used in cardiac surgery. selectin (P-selectin) is found in granules of the ECs, known
Patients receiving the former, most often containing one or as Weibel-palade bodies. Upon stimulation with, e.g., throm-
more pyrolytic carbon disks attached to a sewing ring, need bin, P-selectin is rapidly mobilized to the cell surface of ECs
life long anticoagulant medication to prevent thromboem- as well as platelets.25 The expression is short lived, declining
bolic events, which is associated with a higher risk of bleed- substantially within a few minutes.
ing disorders. The degenerative process of a bioprosthetic Endothelial cell selectin (E-selectin) expression is
graft has a multifactorial etiology. Inflammatory,15 mechani- largely restricted to activated ECs.26 Cultured ECs express
cal,16 and metabolic processes17,18 as well as the glutaralde- E-selectin after exposure to interleukin-1 (IL-1), tumor ne-
hyde per se,12,13 have been discussed as contributing factors. crosis factor alpha (TNF-#) or lipopolysacharide (LPS).23,27
These bioprostheses are slowly deteriorated due a complex Maximal surface expression is observed at 4-6 h, followed
reaction involving stepwise calcification, resulting in poor by a decline. The basal levels are usually reached 24-48 h
hemodynamic performance. after stimulation.
Adhesion Molecule Expression Following In Vitro Lining 173
Fig. 16.1. Induction and peak expression of endothelial cell adhesion molecules. H2O2 = superoxide, IL = interleukin, TNF =
tumor necrosis factor, LPS = lipopolysaccharicde (endotoxin), IFN = interferon, ICAM = intercellular adhesion molecule, VCAM
= vascular cell adhesion molecule, MHC = major histomcompatibility complex. Please note that ICAM-2 and LFA-3 are constitu-
tively expressed and that MHC class I is both constitutively expressed and inducible.
divided into several steps: initial contact, primary adhesion ied. The human monocytic cell line U937 was used for
(often referred to as rolling), activation, secondary adhesion adhesion studies. It was shown that monocyte adhesion to
and transmigration. Figure 16.2 shows schematically the nonendothelialized PA was significantly higher than adhe-
several steps involved in neutrophil extravasation in acute sion to endothelialized specimens on days 3 and 7. The spon-
inflammation. Following initial contact, leukocytes often roll taneous monocyte adhesion to EC-seeded porcine aorta was
slowly along the endothelium. Rolling is believed to keep highest 1 day after seeding, declined on day 3 and remained
the cells in close enough contact with the endothelium so low on the 7th day. When the endothelialized tissue was
that they can be stimulated by substances in the local envi- stimulated with IL-1 ! , the adhesion of monocytes
ronment and engage additional binding mechanisms.43 Roll- increased significantly.
ing requires a definite adhesive interaction, as was first dem- Human saphenous vein ECs (HSVECs) seeded on xe-
onstrated by the observation that neuotrophils roll much nogeneic vascular tissue were shown to express E-selectin,
more slowly than predicted for cells tumbling in the fluid ICAM-1, VCAM-1 and MHC class II in a time-dependent
stream adjacent to the vessel wall without any adhesion.44,45 manner. This expression pattern correlates well with the
Stimulated neutrophils adhere to the microvasculature. This adhesion of monocytes. It was found that the expression of
is referred to as firm adhesion. Once firm adhesion has been adhesion molecules and MHC class II was similar on viable
established, the leukocytes migrate to the intercellular junc- and nonviable tissue, as well as on ECs seeded on gelatin-
tions, where they can diapedese.46 coated culture plastic. However, it was also shown that non-
There is a great interest in in vitro endotheli- specific mouse IgG binding to the HSVECs was higher one
alization of cardiovascular prosthetic materials to achieve a day after seeding, although not as high as the adhesion mol-
confluent and flow-resistant vascular lining.14,47-51 But, to ecule expression. A significant increase in cellular adhesion
my knowledge, nothing has been published regarding ad- molecule expression and in MHC class II expression was
hesion molecule expression on EC surface when seeded on observed after stimulation with IL-1! or IFN-&. The non-
prosthetic grafts and exposed to shear stress. More has been specific binding was not affected after cytokine stimulation.
written about neutrophil-endothelial cell interactions. Although mechanical de-endothelialization is a relatively
Quantitative studies of neutrophil adhesion under flow con- imprecise technique, porcine aorta was used as a biological
ditions have shown that overall adhesion decreases with in- model tissue, mainly because of its availability. Furthermore,
creased flow and wall shear stress.52,44 It has been shown the matrix proteins exposed after scraping are reasonably
that the leukocyte adherence is optimal in the postcapillary representative of the matrix on a bioprosthetic graft.
venular wall, where it is about 1-4 dyn per cm2. It decreases The human monocytic cell line U937 is a relatively
rapidly at higher shear stresses. However, there are some re- immature clone derived from a human histiocytoma. How-
sults suggesting that integrin mediated adhesion in the ab- ever, the U937 cells present leukocyte surface integrins to
sence of selectins appears to be effective only at wall shear allow adequate attachment to ECs.56 To our knowledge, the
stresses well below 1 dyn cm2.53,54 expression of U937 integrins, and their role in adhesion to
subendothelial tissue, is not well characterized. However, it
Endothelialization Reduces Monocyte is likely that “normal” integrins are present on U937 and
Adhesion to Xenogeneic Tissue in a Time contribute to the increased binding to de-endothelialized
Dependent Manner collagenous tissue as compared to endothelialized tissue.57
Our group published a paper in 1995 wherein we The time dependent manner in which the adhesion mol-
investigated monocyte adhesion to de-endothelialized and ecule expression declines, which was found in this study, is
re-endothelialized porcine aorta (PA) before and after interesting for several reasons. It indicates that perhaps one
cytokine stimulation.55 In addition, the expression of a should wait 3-7 days before operating an endothelialized
selection of adhesion molecules on the EC surface before graft in a patient. This notion is supported by other find-
and after cytokine stimulation after 1, 3 and 7 days were stud- ings such as that the maturation of the cytoskeleton may be
more pronounced after 7-9 days.58 Furthermore, it has been and organs for transplantation and various gene therapy
shown that ECs exposed to chronic shear stress in vitro, ap- techniques. Because of the integrated systems, and analysis
plied in a stepwise fashion over several days, are induced to required to understand intracellular functions and intercel-
become tightly adherent to the substratum and exhibit more lular interactions, this development should be done in close
differentiated features.59 Most likely this assay also gives a cooperation with researchers with training in biology
better cell retention in seeded grafts after the blood flow has and bioengineers.
been restored. The human body is a marvelous feat of engineering.
There was no difference in the expression of EC adhe- Many vital systems continuously interact with each other in
sion molecules in cells seeded on viable and nonviable tis- ways that are simultaneously extremely complex yet beauti-
sue specimens. This can be explained by the fact that no fully simple. Our understanding at the molecular level has
immunocompetent cells were present. Apparently, the vi- made major advances in the last decade. Because these in-
able smooth muscle cells, fibroblasts or matrix proteins teractions occur dynamically and are subject to various lev-
which were communicating with or exposed to the human els of control, much of our increase in knowledge has been
ECs, did not influence the ECs’ expression of adhesion mol- possible because of the awareness that real advances require
ecules in this model. cross-disciplinary teams of bioengineers and biological sci-
Interestingly, the high levels of monocyte adhesion and entists. This interaction has been and will be extremely ben-
nonspecific binding of antibodies on the first day after seed- eficial in vascular biology.
ing were also true for ECs seeded on gelatin. This may sug-
gest a nonspecfic activation of ECs by passaging. Acknowledgments
In conclusion, there are several findings suggesting that I wish to thank Associate Professor Anders
the endothelium is more ready to cope with and survive the Hægerstrand for fruitful comments on the manuscript. This
conditions in vivo if they are allowed to mature on the graft study was supported by grants from The Swedish Heart Lung
before transplantation. These findings include the adhesion Foundation, The Memorial Foundation of Carl Jeppsson and
molecule expression, at least in vitro. R & E Lundström.
15. Grabenwöger M, Grimm M, Eybl E et al. New aspects of Gatter K. ICAM-3 expression on endothelium in lymphoid
the degeneration of bioprosthetic heart valves after long- malignancy. Am J Path 1993; 143:1040-1043.
term implantation. J Thorac Cardiovasc Surg 1992; 33. Rice GE, Munro JM, Bevilacqua MP. Inducible cell adhe-
104:14-21. sion molecule 110 (INCAM-110) is an endothelial recep-
16. Warnes C, Scott M, Silver G et al. Comparison of late tor for lymphocytes. A CD11/CD18-independent adhesion
degenerative changes in porcine bioprostheses in the mi- mechanism. J Exp Med 1990; 171:1369-1374.
tral and aortic valve position in the same patient. Am J 34. Briscoe D, Shoen F, Rice G et al. Induced expression of
Cardiol 1983; 51:956-68. endothelial-leucocyte adhesion molecules in human car-
17. Sanders S, Levy R, Freed M, Norwood W, Castaneda A. diac allografts. Transplantation 1991; 51:537-539.
Use of Hancock porcine xenografts in children and ado- 35. Galumbeck M, Sanfilippo F, Hagen P-O, Seaver A,
lescents. Am J Cardiol 1980; 46:429-38. Urbaniak J. Inhibition of vessel allograft rejection by en-
18. Schoen F, Kujovich J, Levy R, St John T, Sutton M. dothelial removal. Ann Surg 1987; 206:757-64.
Bioprosthetic valve failure. Cardiovasc Clin 1988; 36. Newman PJ, Albelda SM. Cellular and molecular aspects
18:289-316. of PECAM-1. Nouv Rev Fr Hematol 1992; 34:S7-S11.
19. Cotran R. New roles for the endothelium in inflamma- 37. Albelda SM, Müller WA, Buck C, Newman PJ. Molecular
tion and immunity. Am J Pathol 1987; 129:407-13. and cellular properties of PCAM-1 (endo-CAM/CD-31):
20. Pober JS. Cytokine-mediated activation of vascular endot- A novel vascular cell-cell adhesion molecule. J Cell Biol
helium: Physiology and pathology. Am J Pathol 1988; 1991; 114:1059-1068.
133:426-433. 38. Vaporciyan AA, DeLisser HM, Yan HC et al. Involvement
21. Bevilacqua M, Pober J, Majeau G, Cotran R, Gimbrone of platelet-endothelial cell adhesion molecule-1 in neu-
M. Interleukin (IL-1) induces biosyntesis and cell surface trophil recruitment in vivo. Science 1993; 262:1580-2.
expression of procoagulant activity in human vascular 39. Pober J, Collins T, Gimbrone M, Libby P, Reiss C. In-
endothelial cells. J Exp Med 1984; 160:618-623. ducible expression of class II major histocompatibility
22. Old LJ. Tumour necrosis factor. Nature 1986; 320:584-588. complex antigens and the immunogenicity of vascular
23. Bevilacqua M, Pober J, Wheeler M, Cotran R, Gimbrone endothelium. Transplantation 1986; 41:141-146.
M. Interleukin-1 acts on cultured human vascular endot- 40. Libby P, Hansson GK. Biology of disease. Involvement of
helium to increase the adhesion of polymorphonuclear the immune system in human atherogenesis: Current
leucocytes, monocytes, and related leucocyte cell lines. J knowledge and unanswered questions. Lab Invest 1991;
Clin Invest 1985; 76:2003-2011. 64:5-15.
24. Rosenbaum JT, Howes EL, Rubin RM, Samples JR. Ocu- 41. Basham T, Merigan T. Recombinant interferon-& increases
lar inflammatory effects of intravitreally injected tumor HLA-DR synthesis and expression. J Immunol 1983;
necrosis factor. Am J Pathol 1988; 133:47-53. 130:1492-1494.
25. McEver RP, Beckstead JH, Moore KL, Marshall-Carlson 42. Carlos TM, Harlan JM. Leukocyte-endothelial adhesion
L, Bainton DF. GMP-140, a platelet alpha-granulae mem- molecules. Blood 1994; 84:2068-2101.
brane protein, is also synthesized by vascular endothelial 43. Jones AD, Smith CW, McIntire VL. Leucocyte adhesion
cells and is localized in Weibel-palade bodies. J Clin In- under flow conditions: Principles important in tissue en-
vest 1989; 84:92-99. gineering. Biomaterials 1996; 17:337-347.
26. Gimbrone MAJ, Bevilacqua MP. Vascular endothelim: 44. Atherton ABG. Relationship between the velocity of roll-
Functional modulation at the blood interface. In: ing granulocytes and that of the blood flow in venules. J
Simionescu N, Simionescu M, eds. Endothelial cell biol- Physiol 1973; 233:157-165.
ogy in health and in disease. New York: Plenum Press 45. Goldman AJ, Cox RG, Brenner H. Slow viscious motion
1998:255-273. of a sphere parallel to a plane wall. Chem Engng Sci 1967;
27. Bevilacqua MP, Stengelin S, Gimbrone MA Jr, Seed B. 22:637-660.
Endothelial leukocyte molecule-1: An inducible receptor 46. Smith CW. Endothelial adhesion molecules and their role
for neutrophils related to complement regulatory proteins in inflammation. Can J Physiol Pharmacol 1993; 70:76-87.
and lectins. Science 1989; 243:1160-1165. 47. Zilla P, Preiss P, Groscurth P et al. In vitro lined endot-
28. Pober JS, Gimbrone MA, Lapierre LA et al. Overlapping helium: Initial integrity and ultrastructural events. Sur-
patterns of activation of human endothelial cells by gery 1994; 116:524-534.
interleukin-1, tumor necrosis factor and immune inter- 48. Zilla P, Fasol R, Deutsch M et al. Endothelial cell seeding
feron. J Immunol 1986; 137:1893-1896. of polytetrafluoroethylene vascular grafts in humans: A
29. Staunton DE, Dustin ML, Springer TA. Functional clon- preliminary report. J Vasc Surg 1987; 6:535-541.
ing of ICAM-2, a cell adhesion ligand for LFA-1 homolo- 49. Zilla P, Fasol R, Preiss P et al. Use of fibrin glue as a
gous to ICAM-1. Nature 1989; 339:61-64. substrate for in vitro endothelialization of PTFE vascular
30. van der Vieren M, Le Trong H, Wood CL, Moore PF, St grafts. Surgery 1989; 105:515-522.
John T, Staunton DE, Gallatin WM. A novel leucointegrin, 50. Hægerstrand A, Bengtsson L, Gillis C. Serum proteins
alpha d beta 2, binds preferentially to ICAM-3. Immu- provide a matrix for cultured endothelial cells on ex-
nity 1995; 3:683-690. panded polytetrafluoroethylene vascular grafts. Scand J
31. Holness CL, Bates, PA, Little AJ, Buckley CD, McDowall Thor Cardiovasc Surg 1993; 27:21-26.
A, Bossy D, Hogg N, Simmons DL. Analysis of the bind- 51. Bengtsson L, Rådegran K, Hægerstrand A. A new and
ing site on intercellular adhesion molecule 3 for the leu- simple technique to achieve a confluent and flow resis-
kocyte integrin lymphocyte function-associated antigen 1. tant endothelium on vascular ePTFE-grafts using human
J Biol Chem 1995; 270:877-884. serum. Eur J Vasc Surg 1994; 8:182-187.
32. Doussis-Anagnostopoulou I, Kaklamanis L, Cordell J, 52. Jones DA, McIntire LV, McEver RP, Smith CW. P-selectin
Jones M, Turley H, Pulford K, Simmons D, Mason D, supports neutrophil rolling on histamine-stimulated en-
dothelial cells. Biophys J 1993; 65:1560-1569.
Adhesion Molecule Expression Following In Vitro Lining 177
53. Lawerence MB, Smith CW, Eskin SG, McIntire LV. Ef- 57. Patarroyo M. Short analytical review: Leukocyte adhesion
fect of venous shear stress on CD18-mediated neutrophil in host defence and tissue injury. Clin Immunol
adhesion to cultured endothelium. Blood 1990; Immunopathol 1991; 60:333-348.
75:227-237. 58. Schnittler HJ, Franke RP, Fuhrmann R. Influence of vari-
54. Lawrence MB, Springer TA. Leukocytes roll on a selectin ous substrates on the actin filament exposed to fluid shear
at physiologic flow rates: Distinction from and prerequi- stress. Basel: Karger 183-188.
site for adhesion through integrins. Cell 1991; 65:859-873. 59. Ballermann BJ, Ott MJ. Adhesion and differentiation of
55. Gillis C, Bengtsson L, Hægerstrand A. Reduction of mono- endothelial cells by exposure to chronic shear stress: A
cyte adhesion to xenogeneic tissue by endothelialization: vascular graft model. Blood Purification 1995; 13:125-134.
An adhesion and time-dependent mechanism. J Thorac 60. Cronstein NB, Weissmann G. The adhesion molecules of
Cardiovasc Surg 1995; 110:1583-1589. inflammation. Arthritis and Rheumatism 1993; 36:147155.
56. Prieto J, Eklund A, Patarroyo M. Regulated expression of 61. McIntire LW. 1992 ALZA Distinguished Lecture: Bioengi-
integrins and other adhesion molecules during differen- neering and Vascular Biology. Annals of Biomedical En-
tiation of monocytes into macrophages. Cell Immunol gineering 1994; 22:2-13.
1994; 156:191-211.
CHAPTER 17
In Vitro Endothelialization Elicits Tissue Remodeling
Emulating Native Artery Structures
Manfred Deutsch, Johann Meinhart, Peter Zilla
I n humans, the retrieval of samples from cardiovascular implants is a rare event. Therefore,
it is almost impossible to evaluate the long term healing pattern of cardiovascular pros-
theses. At the same time, animal experiments do not offer a satisfying solution either. The
significantly shorter length of the prostheses and an incomparably short implantation time
make transanastomotic tissue ingrowth their predominant healing mode. Therefore, there
is hardly an opportunity to histologically study blood surfaces of clinical implants which are
not affected by transanastomotic ingrowth. This is particularly frustrating in an exciting
field such as clinical in vitro endothelialization of arterial grafts, in which the patency results
are highly encouraging but the proof for the ongoing existence of functional intimal tissue
has only sporadically been provided.
When the techniques for in vitro lining were refined in the mid 1980s, the seeding
community was haunted by the fear of so called “contaminating” smooth muscle cells. With
the strong emphasis on intimal hyperplasia of those days, the smooth muscle cell was the
declared enemy. As a result, much of the research effort went into purification of endothelial
cell cultures.1,2
Preclinical primate experiments3 confirmed that a mature and functional endothe-
lium continued to form a confluent cell coverage after weeks of implantation. Moreover, the
opportunity to obtain a specimen of a clinically in vitro lined ePTFE graft also showed that
a single layer of pure endothelial cells confluently covered the protein precoated ePTFE sur-
face of the graft 5 weeks after implantation.4 Nevertheless, fears of fibrinolytic degradation
of the underlying matrix, with the concomitant detachment of the endothelium, were justi-
fied in view of the lack of long term specimens. Furthermore, in native arteries endothelial
cells closely interact with their underlying smooth muscle cells in an adaptive attempt to
standardize flow and shear stress conditions.5 Therefore, the presence of a monolayer rest-
ing almost directly on the synthetic graft structure seemed more and more unphysiological,
and perhaps even unsustainable. However, clinical long term studies increasingly indicated
that a functional endothelium continued to cover the graft surface, even after years of im-
plantation. Eventually, we got the chance to morphologically analyze a significant piece of
explanted graft 41 months after clinical in vitro lining. Since the healing pattern of a certain
prosthetic implant does not principally differ from case to case, the extrapolation of a case
report to a principal healing response seems justified, particularly in view of the fact that a
representative number of samples cannot be expected in the clinical setup.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
180 Tissue Engineering of Prosthetic Vascular Grafts
Case Report6 endothelium was still confluent, but the cells were flatter,
A 69 year old man who had undergone a bilateral with better pronounced marginal overlapping.
femoro-popliteal bypass graft procedure with in vitro Forty-one months later at the time of revision, the
endothelialized ePTFE grafts was readmitted 41 months later angiographic appearance of the stenosed area resembled a
for graft revision. The original indication for an in vitro lined native arteriosclerotic lesion. A 21 cm long graft segment
graft was the fact that both saphenous veins had already been was subsequently replaced by an equally in vitro
taken due to previous coronary artery bypass grafting. The endothelialized piece of ePTFE graft. The distance of the
reason for readmission and revision of one graft was the affected segment to the anastomoses was approximately
recurrence of severe claudication and a deterioration of the 15 cm on both sides. On gross examination of the explant,
ankle-brachial index from 0.89-0.59. Furthermore, angiog- the inner surface of the entire specimen was smooth and
raphy demonstrated a significant stenosis in the midsegment gray-white, showing multiple yellow and white plaques again
of the affected graft. resembling native atherosclerosis. Histologically, the inner
The lining procedure with cultured autologous venous surface was completely covered by a single layer of flattened
endothelial cells has been extensively described elsewhere.7-10 cells which were strongly positive for factor VIII, CD31 and
Endothelial cells were harvested by the in situ no-touch tech- CD34 (Fig. 17.1). Between this intima and the graft, a con-
nique previously reported.1 Routine immunohistochemical tinuous tissue layer of 1.21 ± 0.19 mm thickness was de-
screening of the preseeding mass cultures with factor VIII monstrable (Fig. 17.2) showing all histological and immu-
confirmed the purity of endothelial cells. The precoating of nohistochemical criteria of an arterial tunica media. It con-
the ePTFE structure prior to lining was done with a tained numerous actin (Fig.17.3) and desmin-positive cells
fibrinolytically inhibited fibrin glue.3,4,6-10 On light micros- resembling smooth muscle cells. A well developed internal
copy of freshly lined grafts and of preimplantation controls, elastic membrane could regularly be found underneath a
a confluent endothelial cell layer rested on a homogenous collagen iv-positive basement membrane of the endothe-
and evenly covering protein matrix. The postseeding matu- lium (Fig. 17.4). In atheromatous areas the internal elastic
ration of the confluent endothelium on the fibrin matrix membrane was disintegrated to various degrees (Fig. 17.4).
did not result in a diminished thickness of this adhesion The outside of the graft was surrounded by a rim of
matrix (7.3 ∝m versus 7.0 ∝m). By scanning electron mi- histiocytes with multiple multinucleated giant cells. The
croscopy, the freshly lined graft surface showed a completely prosthesis itself did not show any capillary ingrowth. In ad-
covering, confluent endothelium at the end of the lining dition, multiple fatty streaks, fibrous plaques and athero-
procedure. Nine days later, at the time of implantation, the mas (indistinguishable from conventional atherosclerotic
lesions) could be seen. Only in these areas, KP-1 and PG-M1 in diameter. These protrusions were only partially covered
positive foamy macrophages were found (Fig. 17.5). A by intact endothelium, whose cells were polygonal rather
neovascularization of the intima by small capillaries growing than aligned with the blood stream (Fig. 17.7). In contrast
in from the luminal surface of the graft was only found in to the unaffected graft areas, highly polarized leukocytes were
fibrous plaques, but not in the unaffected parts of the graft. adherent. Occasionally, only lamellopodia of these leuko-
On scanning electron microscopy, the entire graft seg- cytes were visible, indicating subendothelial leukocyte mi-
ment was covered by a mature and confluent endothelium gration. Between these endothelial remnants the subendo-
whose cells were mostly elongated parallel to the bloodstream thelial matrix was openly exposed (Fig. 17.7).
(Fig. 17.6). These endothelial cells showed numerous mi- Ultrastructurally, transmission electron microscopy
crovilli and pronounced marginal overlapping. The entire demonstrated a well differentiated endothelium with nu-
surface was free of thrombotic adherences, either fibrin or merous Weibel-Pallade bodies in those areas which were not
platelets. Areas with macroscopically visible plaques were affected by arteriosclerotic changes (Fig. 17.8). Confluence
characterized by surface protrusions of approximately 1 mm was maintained through complex intercellular contacts. In
182 Tissue Engineering of Prosthetic Vascular Grafts
immediate proximity to the endothelial cells a delicate and proximity of a plaque, these cells contained strikingly large
noncontinuous basal membrane was found. The endothe- lipid vacuoles (Fig. 17.10).
lium and its basal membrane rested on a dense network of
collagen fibrils. The subendothelial matrix was character- Conclusions
ized by mature myofibroblasts embedded in densely packed In vitro endothelialization of small diameter vascular
collagen strands (Fig. 17.9). Although these cells had well prostheses has significantly improved prosthetic graft pa-
differentiated peripheral actin filaments they also showed tency.9-12 Due to the difficulty in obtaining specimens from
characteristics of secretory cells: a well developed Golgi com- human implants, it could only be assumed that a persistent
plex and often a pronounced endoplasmic reticulum. In the endothelium was the underlying reason for this better
184 Tissue Engineering of Prosthetic Vascular Grafts
performance. Although the short term integrity of the lined Coseeding During the Initial Endothelialization
endothelium has previously been proved in a 5 week ex- If the progenitor cells of the neo-media were seeded
plant,4 long term developments remained speculative. With together with the endothelial cells, they must have remained
the opportunity to investigate a 41 month implant we were undetectable during cultivation. Even if the assessment of
eventually able to substantiate the clinical success with mor- purity of endothelial cell cultures was based on phenotypic
phological evidence. We could show that in vitro lined grafts characteristics, connective tissue cells can usually be easily
maintain an intact endothelial coverage even after 3 1/2 years detected. Smooth muscle cells and fibroblasts lack the con-
of implantation. Unexpected, however, was the finding that tact inhibition of endothelial cells and therefore tend to over-
a neomedia had developed between the prosthesis and the grow each other in a typical elongated, spindle shape. Since
endothelium throughout the entire length of the graft. The our lining technique only uses first passage mass cultures,1
cells of this neomedia contained actin filaments and were endothelial cells do not show signs of phenotypic senes-
separated from the intima by a true internal elastic mem- cence.14 This renders them even more the typical cobble-
brane which distinguished them from a neointima. Since stone monolayer in their morphology. Furthermore, first
untreated ePTFE grafts do not develop a tissue layer on their passage mass cultures necessitate prolonged cultivation in
luminal side, it is evident that this neomedia was enabled or one and the same flask,1 which usually allows connective
caused by the in vitro lining procedure. Considering the fact tissue cells to develop their typical shape. Therefore, only a
that cell ingrowth on conventional grafts is limited to the very insignificant number of connective tissue cells might
immediate perianastomotic region,13 the most eminent have been inoculated. To achieve an evenly distributed neo-
question is: How did these neomedia cells get there? media with such a low inoculum in a graft of 50-60 cm
The three principal options are: length, cells would need to migrate over long distances. Thus,
1. Through coseeding during the initial endo- even if the neo-media originates from initially coseeded
thelialization process; smooth muscle cells, cell migration would remain a signifi-
2. Subendothelial migration from the adjacent artery; cant phenomenon. Alternatively, a higher inoculum might
or have been achieved by cells which barely differ phenotypi-
3. Colonization from the blood. cally from endothelial cells and were therefore not detected
in the initial culture. This would be feasible because the vas-
cular wall intima contains a stem cell population which is
186 Tissue Engineering of Prosthetic Vascular Grafts
epitheloid in culture but still represents a subpopulation of exactly this concern. In order to clarify this issue we have
smooth muscle cells. However, such a scenario would mean observed tissue development following mixed microvascu-
that two screening methods failed, the immunohistochemi- lar seeding over a long-term period.29 The major outcome
cal staining because of the small sample size and the pheno- of this study was the concurrence of two events: the matu-
typic because of the presence of a rare subpopulation of ration of the subendothelial cell matrix from fibroblastoid
smooth muscle cells. towards myocytoid and the accomplishment of an equilib-
rium of cell proliferation. After the long lasting disputes of
Subendothelial Migration from the Adjacent Artery the 1980s over the method of choice for actively achieving
If the progenitor cells of the neo-media were not graft endothelialization, it is exciting to see that two meth-
seeded but migrated from the anastomotic region, the ques- ods as diverse as in vitro lining and mixed microvascular
tion is: Why was this possible? For reasons not fully under- seeding eventually lead to an identical result: the healing of
stood yet, connective tissue ingrowth onto synthetic vascu- a prosthetic graft accompanied by initial tissue proliferation,
lar prostheses stops shortly after the anastomosis.13 One ex- cell differentiation and endothelialization. This realization
planation may lie in an inhibitory fibrin layer on the blood further underlines the current redefinition of standpoints
surface16 which gradually builds up. If, however, an endot- in prosthetic graft research. In contrast to previous beliefs,
helial cell population of the surface was possible through host tissue appears to be capable of physiologically incor-
apt transmural ingrowth before this change in biological porating synthetic grafts, provided it happens prior to a
surface environment, smooth muscle cells grow well on the buildup of adverse conditions.16 Therefore, the manipula-
same synthetic surface on which they ceased to proliferate tion of the chronology of events may well hold the key to
before. Therefore, it seems as if the presence of a PDGF- ingrowth facilitation. What seems unchallenged is the fact
producing endothelium17 makes the difference with regard that early endothelialization facilitates the subsequent
to the presence of a subendothelial smooth muscle cell popu- musculogenesis within the graft wall. All nonbiological as-
lation. Although quiescent endothelial cells barely produce pects like compliance or porosity—as important as they
any PDGF, a significant PDGF production occurs in regen- might be—were previously overemphasized16 although they
erating endothelium.18,19 In spite of continual integrity of will need to be an integral part of an entirely engineered
the lined endothelium we have seen a significant regenera- arterial graft. Nevertheless, it is quite possible that these
tion in the primate model.3 Others have also found a 8- to nonbiological aspects are the limiting factors of currently
10-fold increase in endothelial replication on ePTFE available methods of graft endothelialization: Both non-
prostheses20 accompanied by a significant production of compliance and low porosity might well be responsible for
growth factors.21 the lack of complete differentiation of myofibroblasts to
myocytes and for the early appearance of true arterioscle-
Colonization from the Blood rotic lesions in endothelialized prosthetic grafts. The most
Finally, as a third option, circulating multipotent stem striking observation, however, was that newly formed vessel
cells must be considered. All four cell types, fibroblasts, structures can emulate a native artery even to the extent that
myocytes, endothelial cells and blood borne cells, are em- they participate in the generalized arteriosclerotic disease
bryologically of the same origin. Since they represent vari- of the arterial tree.
ous differentiations of one common stem cell, transforma-
tion within this cell family appears to be a reasonable op- References
tion. The differentiation of mononuclear blood cells to fi- 1. Zilla P, Fasol R, Dudeck U, Kadletz M, Siedler S, Preiss
broblasts, myocytes and endothelial cells has been de- P, Sanan D, Odell J, Reichart B. In situ canulation,
scribed.22-24 Our finding that white blood cells were not only microgrid follow-up and low density plating provide first
adherent, but also clearly migrated underneath the endot- passage endothelial cell mass cultures for in vitro lining.
helium, might be explained not only with an inflammatory J Vasc Surg 1990; 12:180-9.
2. Haegerstrand A, Gillis C, Bengtsson L. Serial cultivation
reaction, but might well fit into the multipotent blood cell
of adult human endothelium from the great saphenous
theory, which is currently best suited to explain isolated is- vein. J Vasc Surg 1992; 16:280-285.
lands of endothelial cells in the center of prosthetic grafts.25-27 3. Zilla P, Preiss P, Groscurth P, Rösemeier F, Deutsch M,
Whichever theory will eventually explain the forma- Odell J, Heidinger C, Fasol R, Von Oppell U. In vitro
tion of a differentiated neo-media on a 55 cm long pros- lined endotheliaum: Initial integrity and ultrastructural
thetic graft, distinct migration and intermesenchymal trans- events. Surgery 1994; 16:524-534.
formation of cells appear to be pivotal events in this process. 4. Fischlein T, Zilla P, Meinhart J, Puschmann R, Vesely M,
Apart from explaining the origin of this neo-media, Eberl T, Balon R, Deutsch M. In vitro endotheliaization
another question seems to be of imminent practical impor- of a meso-systemic shunt—A clinical case report. J Vasc
tance: Does this neo-media formation represent a patho- Surg 1994; 19:549-554.
5. Kaufman BR, De Luca DJ, Folsom DL, Mansell SL,
logical process in the sense of a nonself-limiting hyperplas-
Gorman ML, Fox PL, Graham LM. Elevated platelet-de-
tic cell proliferation or rather a self-limiting mechanism of rived growth factor production by aortic grafts implanted
tissue remodeling? To answer this question, mixed microvas- on a long-term basis in a canine model. J Vasc Surg 1992;
cular seeding seems to provide an explanatory analogy. When 15:806-816.
Park et al28 presented a case report on a seeded human pros- 6. Deutsch M, Meinhart J, Vesely M, Fischlein T, Groscurth
thesis, an unusually thick subendothelial cell layer had raised P, Von Oppell U, Zilla P. In vitro endothelialization of
In Vitro Endothelialization Elicits Tissue Remodeling Emulating Native Artery Structures 187
expanded polytetrafluoroethylene grafts: A clinical case 18. Kraiss LW, Raines EW, Wilcox JN, Seifert RA, Barraett
report after 41 months of implantation. J Vasc Surg 1997; BT, Kirkman TR, Hart CE, Bowen-Pope DF, Ross R,
25:757-763. Clowes AW. Regional expression of the platelet-derived
7. Zilla P, Fasol R, Preiss P, Kadletz M, Deutsch M, Schima growth factor and its receptors in a primate graft model
H, Tsangaris S, Groscurth P. Use of fibrin glue as a sub- of vessel wall assembly. J Clin Invest 1993; 92:338-348.
strate for in vitro endothelialization of PTFE vascular 19. Golden MA, Au YPT, Kirkman TR, Wilcox JN, Raines
grafts. Surgery 1989; 105:515-522. EW, Ross R, Clowes AW. Platelet-derived growth factor
8. Müller-Glauser W, Zilla P, Lachat M, Bisang B, Rieser F, activity and mRNA expression in healing vascular grafts
von Segesser L, Turina M. Immediate shear stress resis- in baboons. J Clin Invest 1991; 87:406-414.
tance of endothelial cell monolayers lined in vitro on fi- 20. Reidy MA, Chao SS, Kirkman TR, Clowes AW. Endothe-
brin glue-coated ePTFE prostheses. Europ J Vasc Surg lial regeneration: VI: Chronic non-denuding injury in
1994; 7:324-328. baboon vascular grafts. Am J Path 1986; 123:432-439.
9. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T, 21. Zacharis RK, Kirkman TR, Kenagy RD, Bowen-Pope DF,
Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I, Clowes AW. Growth factor production by
Fischlein T. Clinical in vitro endothelialization of femoro- polytetrafluoroethylene grafts. J Vasc Surg 1988; 7:607-610.
popliteal bypass grafts—an actuarial follow-up over 3 22. Lue HJ, Feigl W, Susani M, Odermatt B. Differentiation
years. J Vasc Surg 1994; 19:540-548. of mononuclear blood cels into macrophages, fibroblasts
10. Meinhart J, Deutsch M, Zilla P. Eight years of clinical and endothelial cells in thrombus organization. Exp Cell
endothelial cell transplantation. Closing the gap between Biol 1988; 56:201-210.
prosthetic grafts and vein grafts. ASAIO Journal 1997; 23. Shibuya T, Kambayashi J, Okahara K, Kim DI, Kawasaki
43:M515-M521. T, Shiba E, Mori T. Subendothelial layer of pseudointima
11. Magometschnig H, Kadletz M, Vodrazka M, Dock W, of polytetrafluoroethylene grafts is formed by transforma-
Grimm M, Grabenwöger M, Minar E, Staudacher M, tion of fibroblasts migrated from extravascular space. Eur
Fenzel G, Wolner E. Prospective clinical study with in J Vasc Surg 1994; 8:276-285.
vitro endothelial cell lining of expanded 24. Fujiwara T. Endothelial cells transformed from fibroblasts
polytetrafluoroethylene grafts in crural repeat reconstruc- during angiogenesis. In: Zilla P, Greisler H, eds. Tissue
tion. J Vasc Surg 1992; 15:527-535. Engineering of prosthetic vascular grafts. Georgetown:
12. Leseche G, Ohan J, Bouttier S, Palombi S, Bertrand P, R.G. Landes Co., 1998.
Andreassian B. Above-knee femoropopliteal bypass graft- 25. Shi Q, Hong M, Onuki Y, Ghali R, Huner GC, Johansen
ing using endothelial cell seeded PTFE grafts: Five year KH, Sauvage LR. Endothelium on the flow surface of
clinical experience. Ann Vasc Surg 1995; 9:S15-S23. human aortic dacron vascular grafts. J Vasc Surg 1997;
13. Berger K, Sauvage LR, Rao AM, Wood SJ. Healing of ar- 25:736-742.
terial prostheses in man: Its incompleteness. Ann Surg 26. Hammond W. Surface population with blood-born cells.
1972; 175:118-127. In: Zilla P, Greisler H, eds. Tissue Engineering of pros-
14. Watkins MT, Sharefkin JB, Zajtchuk R. Adult human thetic vascular grafts. Georgetown: R.G. Landes Co., 1998.
saphenous vein endothelial cells: Assessment of their re- 27. Koen W. Entrapment of circulating cells. In: Zilla P,
productive capacity for use in endothelial seeding of vas- Greisler H, eds. Tissue Engineering of prosthetic vascular
cular grafts. J Surg Res 1984; 36:588-596. grafts. Georgetown: R.G. Landes Co., 1998.
15. Schwartz SM, Foy L, Bowen-Pope DF, Ross R. Deriva- 28. Park PK, Jarrell B, Williams SK, Carter TL, Rose DG,
tion and properties of platelet derived growth factor-in- Martinez A, Carabasi RA. Thrombus free human endot-
dependent rat aortic smooth muscle cells. Am J Path 1990; helial surface in the midregion of a Dacron vascular graft
136:1417-1428. in the splanchnic venous circuit—Observation after nine
16. Davids L. The lack of healing in conventional vascular months of implantation. J Vasc Surg 1990; 11:468-475.
grafts. In: Zilla P, Greisler H, eds. Tissue Engineering of 29. Baitella-Eberle G, Groscurth P, Zilla P, Lachmat M,
prosthetic vascular grafts. Georgetown: R.G. Landes Müller-Glauser W, Schneider J, Neudecker A, Segesser L,
Co., 1998. Dardel E, Turina M. Long term results of tissue develop-
17. Clowes AW. Platelet-derived growth factor activity and ment and cell differentiation on Dacron prostheses seeded
MRNA expression in healing vascular grafts in baboons. with microvascular cels in dogs. J Vasc Surg 1993;
J Clin Invest 1991; 87:406-414. 18:1019-1028.
CHAPTER 18
In Vitro Endothelialization of Synthetic Vascular Grafts
in Long Term Clinical Use
Manfred Deutsch, Teddy Fischlein, Johann Meinhart, Peter Zilla
Introduction
P rimary patency of synthetic vascular grafts varies between 30% and 55%, whereas it is
between 68% and 85% for autologous reversed saphenous vein grafts after 5 years of
implantation.1-3 Apart from technical errors and anastomotic intimal hyperplasia, surface
thrombogenicity appears to be a major reason for the limited performance of synthetic con-
duits. The tendency of surgical disciplines towards ready to use products led to efforts aim-
ing at improved synthetic materials, delaying the overdue adoption of biological principles.
When endothelial seeding was eventually introduced as a first attempt to overcome the prob-
lem of graft thrombogenicity with biological tools,4 an elaborate approach soon surren-
dered to the clinicians’ demand for a single staged procedure. Predictably, the first studies in
humans were controversial and mostly disappointing. The most likely reason for the failure
of these approaches was an insufficient seeding density which did not allow the formation
of a sufficient endothelial coverage. One way to overcome a low cell inoculum is by immedi-
ate harvesting of large numbers of cells like microvascular cells from adipose tissue.5-7 The
alternative method of actively facilitating graft endothelialization is in vitro lining with cul-
tured autologous endothelial cells.8-10 After the preclinical proof of this concept in the non-
human primate model,10 our group performed a randomized clinical study involving 49
patients between 1989 and 1991. Based on the encouraging three year results11 and on the
histological proof of an endothelium on a 5 week explant12 and a central graft segment after
41 months of implantation,13 we commenced a clinical program of routine implantation of
in vitro endothelialized grafts.
Laboratory Procedure
In a step by step approach, reliable culture techniques for autologous endothelial cells,8
as well as precoating14 and lining techniques15 which achieved shear stress resistance of the
endothelium,16 were developed. In brief, in vitro endothelialization is a two staged proce-
dure. It needs a laboratory stage before graft implantation in order to provide a high cell
number, sufficient for a complete cellular lining on the luminal graft surface. Therefore, a
small segment of a patient’s subdermal vein (mostly jugular or cephalic) is used as the cell
source and taken out under local anesthesia. After careful, touch-free dissection, the vessel is
cannulated in situ with two free-flow vessel cannulas with attached 3-way taps. In order to
remove remaining blood, the vein is flushed with culture medium in a pulsatile manner. It is
subsequently filled with a 0.1% solution of collagenase. The solution is kept under minor
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
190 Tissue Engineering of Prosthetic Vascular Grafts
pressure within the vein after closing the 3-way taps. Fol- cell-free areas. The whole in vitro procedure from vein exci-
lowing excision, the cannulated and collagenase-filled vein sion to graft implanation took 36.9 days in our first clinical
segment is transferred to the laboratory in a container filled study and could be reduced to 30.3 days in the second phase
with prewarmed phosphate buffered saline. An enzyme in- of clinical routine application. Since most procedures are
cubation time of 15 minutes with collagenase guarantees a elective, this time span appears to be tolerable for a great
maximal yield of endothelial cells without contaminating number of patients.
fibroblasts or smooth muscle cells. After centrifugation of
the cell suspension, the pellet is resuspended in complete Surgical Procedure and Clinical Follow Up
culture medium. Complete culture medium consists of Me- Patients were admitted to the study when their saphe-
dium 199 supplemented with preservative-free heparin, nous veins were unavailable, unsuitable or had to be spared
10 ng/ml bFGF, 50 ∝g/ml gentamycin and 20% autologous for other surgical procedures like aortocoronary bypass graft-
serum. For autologous serum preparation, 250 ml blood was ing. All patients received the same anti-aggregatory treat-
previously taken from each patient and allowed to clot over- ment, namely oral dipyridamole, 75 mg/day, and oral acetyl
night at 4°C in order to guarantee a maximum release of salicylic acid, 330 mg/day, beginning two days prior to im-
growth factors into the serum. Cells are subsequently plated plantation and continuing throughout the observation pe-
in 25 cm2 culture flasks. The culture medium is renewed riod. For endothelialized graft implantation, only minor
twice weekly by replacing half the culture medium with fresh changes were necessary as compared to standard surgical
complete medium. Having reached 90% confluency, primary procedure. Graft implantation was performed as previously
cultures are passaged to two 162 cm2 culture flasks. After described,18 with the exception that the culture medium was
having reached the required cell number, the autologuous kept inside the graft without clamping the prosthesis. Pa-
cells are seeded onto the graft surface by using a rotation tients were anesthetized with a continuous epidural block
device which facilitates an even cell distribution under con- using Bupivacaine-Hydrochloride. Thin-walled reinforced
trolled conditions (Fig. 18.1). However, prior to this, grafts PTFE (6 mm inner diameter) was used as graft material in
are preclotted with a special dilution of clinically approved both phases of implantation.
and fibrinolytically inhibited fibrin glue in order to provide All patients underwent a tight postoperative follow up.
an adequate matrix for endothelial cells. In our pilot clinical During the first year of our randomized clinical study we
study, we further pretreated grafts with virologically tested assessed the resistance of the endothelium by means of 111In-
fibronectin, to increase adhesion and shear stress resistance dium labeling. Patency was determined by clinical evalua-
of endothelial cells. However, in our second phase of clini- tion as well as by Duplex sonography. Angiography was rou-
cal routine application of in vitro endothelialization, no clini- tinely performed on an annual basis and in patients with
cally approved fibronectin was commercially available. In suspected graft thrombosis. This follow up also applied to
order to control the success of the seeding procedure, speci- clinical routine in vitro endothelialization, with the excep-
mens for scanning electron microscopy were taken imme- tion of 111Indium labeling of platelets.
diately after rotation and shortly before implantation
(Fig. 18.2). At the time of implantation, the majority of grafts Randomized Clinical Study
show a confluent endothelial cell lining. Only a few grafts Between June 1989 and December 1991, the clinical
occasionally show a preconfluent endothelium or even small benefit of graft endothelialization was evaluated in a ran-
domized control study comprising 49 patients. After a 1:2 Clinical Routine Endothelialization
randomization pattern, 33 patients were assigned to the These favorable results encouraged us to transfer in
endothelialized group and 16 to the control group. Growth vitro endothelialization to clinical routine in vascular sur-
failure of cell cultures eliminated 9 patients of the gery. So far, 93 in vitro lined femoropopliteal bypass grafts
endothelialized group. The remaining 24 patients received have been implanted in 82 patients. The primary patency
27 endothelialized grafts. Patients were monitored for more rate for all endothelialized femoropopliteal reconstructions
than 7 years. 111Indium labeling revealed a significant re- was 68% after 4 years; this is equivalent to that of reversed
duction of surface thrombogenicity of lined grafts. Through- saphenous vein grafts. Above-knee procedures were done in
out the first year of phase 1 of our endothelialization pro- 71, and below-knee procedures in 22 cases. Analysis of pa-
gram, the difference in platelet activation index (PAI) be- tency clearly revealed that endothelialization especially ex-
tween the endothelialized group and the control was sig- erts its beneficial effect in the below-knee position. Primary
nificant (p < 0.05). Although it was most obvious 9 days patency was 61% in above-knee grafts but was 82% in be-
post implantation, the difference remained significant even low-knee grafts (Fig. 18.3). Secondary patency for all
after one year. In the endothelialized group the PAI (platelet femoropopliteal reconstructions was 94% and limb salvage
activation index) was -2.5 ± 9.4 at 9 days, -4.4 ± 13.5 at 3 rate was 99%, since only one limb had to be amputated
months -0.7 ± 7.1 at 6 months and 7.0 ± 15.2 at 12 months. within the entire observation period.
In contrast, the control group’s values were as high as 37.1 ± However, although the patency rate for in vitro lined
at 9 days, 27.8 ± 22.8 at 3 months, 37.2 ± 22.9 at 6 months ePTFE grafts was comparable to that of vein grafts during
and 29.0 ± 17.4 at 12 months. both phases of our clinical endothelialization program, the
After 3 years a Kaplan-meier analysis showed a pri- primary patency rate of phase 2 was lower than that of phase
mary patency rate of 84.7% for endothelialized grafts and 1. This difference may have several causes:
55.4% for control grafts. After 5 years, it was 73.8% for the 1. Phase 1—as an initial randomized trial—included
endothelialized group and 31.5% for controls.11 After 7 years, considerably fewer patients than phase 2;
primary patency for control grafts dropped to zero, whereas 2. The follow up time of phase 2 was shorter and im-
that for endothelialized grafts remained high, at 73.8%.18 plantations were conducted as clinical routine pro-
One patient who underwent bilateral in vitro endothelialized cedures, including substantially more redo opera-
grafting had a unilateral stenosis after 41 months. We de- tions than phase 1;
cided to remove the stenosed portion of the graft and to 3. Grafts of phase 2 had not been precoated with
implant a newly endothelialized graft. This explant proved fibronectin, a potent adhesion molecule, due to the
the persistence of the transplanted endothelium. Except for unavailability of a clinically approved product. Al-
the immediate atherosclerotic plaque area, a confluent en- though the fibrin glue used for precoating contains
dothelium covered the entire length of the 21 cm long speci- fibronectin and endothelial cells deposit their own
men. Endothelial cells were mostly elongated and orientated fibronectin-rich extracellular matrix with time, ini-
parallel to the bloodstream. They were situated on a tial cell adherence may well have been diminished
continuos tissue layer, which showed many morphologic with a lower density of binding sites. When HFN-
criteria of an arterial tunica media13 (see also chapter 17). pretreated grafts were compared with untreated ones,
192 Tissue Engineering of Prosthetic Vascular Grafts
the difference was indistinct (77.8% versus 72.6%; clinical tool and the motivation for intensified further at-
p = 0.3). However, this difference was more pro- tempts searching for an equally effective or even better prod-
nounced when above-knee grafts were compared uct from the shelf.
(80.0% versus 68.9%, p = 0.17) after 3.5 years of im- With this goal in mind, we were one of the few groups
plantation. Although the differences between which did not abandon the idea of surface endothelialization
fibronectin pretreated or nonpretreated grafts were of vascular grafts in the mid-1980s. We decided to rectify
not statistically significant (which might be due to procedural weaknesses before declaring endothelial cell
the different number of observed grafts), the curve transplantation inefficient. Since a low cell inoculum and
may suggest that nonfibronectin treated grafts have an immature cytoskeleton seemed to be the main culprits
a higher failure risk within the first year of implan- for the failure of single-staged venous seeding, we elected to
tation, a fact probably especially true in the above- defy the surgical aversion towards cell culture procedures
knee position with its high flow, high shear stress by confluently in vitro endothelializing prosthetic grafts with
condition. As a solution, pretreatment of the pro- autologous mass cultures of endothelial cells.
tein matrix with synthetic or recombinant RGD Eight years of experience with clinical in vitro endo-
(arginine-glycine-aspartic acid)-containing mol- thelialization not only provided us with patency data which
ecules could be considered. eventually verified the assumption that surface endo-
Our overall 7 year experience with endothelialized thelialization improves the performance of synthetic grafts.
femoropopliteal ePTFE grafts shows a patency of 66.0%. It also helped us to diminish three major concerns linked to
When pretreated with HFN the seven year patency was in vitro lining: infection, failure rate of cultures and the proof
72.1%. In the above-knee group the 7 year patency was even of a persisting endothelium after graft implantation. In 120
as high as 75.8%20 (Fig. 18.4). in vitro endothelialized ePTFE grafts which have been im-
planted altogether since the start of our in vitro endo-
Conclusion thelialization program, only one graft showed late infection
At the end of this century, biological awareness is in a patient with IDDM and lymph fistula. This graft needed
steadily rising among a new generation of cardiovascular to be removed and amputation could be avoided. However,
surgeons. Today’s research no longer focuses solely on sur- routine microbiological examination of this graft has been
face endothelialization, but rather on the understanding of performed before implantation and was found to be nega-
complex events that might eventually lead to real graft heal- tive for infections. One lined graft which was diagnosed to
ing. Nevertheless, biological events facilitating spontaneous be contaminated during the cultivation period was discarded
graft healing are not yet fully understood and it might take and remained the only occurrence of its kind. The analysis
many more years until such a prosthesis is available for clini- of risk factors for culture conditions enabled us to bring the
cal implantation. Until then, an active method of graft failure rate for autologous endothelial cell cultures down
endothelialization which was shown to significantly improve from 27.3-5% (see chapter 15). Considering the fact that
patency rates could provide us with both an indispensable this two staged procedure provides 93% of patients with an
In Vitro Endothelialization of Synthetic Vascular Grafts in Long Term Clinical Use 193
arterial prosthesis which equals a vein graft, and that the 6. Jarrell B, Williams S, Stokes G, Hubbard A, Carabasi A,
remaining 7% end up with the prosthesis which they would Koolpe E, Greener D, Pratt K, Moritz M, Radomski J,
have had without this procedure, one can accept the low Speicher L. Use of freshly isolated capillary endothelial
probability of growth failure without ethical concerns. Fi- cells for the immediate establishment of a monolayer on
a vascular graft at surgery. Surgery 1986; 100:392-399.
nally, the proof of the existence of a confluently covering
7. Baitell-Eberle G, Groscurth P, Zilla P, Lachat M, Müller-
endothelium 5 weeks and 41 months after in vitro lining Glauser W, Schneider J, Neudecker A, von Segesser L,
dissipated concerns regarding the continual presence of an Dardel E, Turina M. Long term results of tissue develop-
endothelium after implantation. In summary, eight years of ment and cell differentiation on dacron prostheses seeded
clinical in vitro endothelialization were able to prove two with microvascular cells in dogs. 1993; 18:1019-1028.
main hypotheses: Autologous endothelial cell lining signifi- 8. Zilla P, Fasol R, Dudeck U, Siedler S, Preiss P, Fischlein
cantly improves the patency of prosthetic small diameter T, Müller-Glauser W, Baitella G,Sanan D, Odell J, Reichart
vascular grafts, and a cell culture-dependent procedure can B. In situ cannulation, microgrid follow-up and low den-
be carried over into clinical routine. sity plating provide first passage endothelial cell mass cul-
tures for in vitro lining. Vasc Surg 1990; 12:180-189.
References 9. Zilla P, Fasol R, Preiss P, Kadletz M, Deutsch M, Schima
1. Veith FJ, Gupta SK, Ascer E et al. Six year prospective H, Tsangaris S, Groscurth P. Use of fibrin glue as a sub-
multicenter randomized comparison of autologous saphe- strate for in vitro endothelialization of PTFE vascular
nous vein and expanded polytetrafluoroethylene grafts in grafts. Surgery 1989; 105:515-522.
infrainguinal arterial reconstructions. J Vasc Surg 1986; 10. Zilla P, Preiss P, Groscurth P, Rösemeier F, Deutsch M,
3:104-14. Odell J, Heidinger C, Fasol R, von Oppell U. In vitro-
2. Cacciatore R, Inderbitzi R, Stirnemann P. Five years ex- lined endothelium: Initial integrity and ultrastructural
perience with infra-inguinal arterial reconstructions: A events. Surgery 1994; 116:524-534.
comparison of venous with PTFE bypass. VASA 1992; 11. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T,
21:171-176. Minar E, Dudczak R, Lugmaier H, Schmidt P, Noscian I,
3. Quiñones-Baldrich WJ, Prego AA, Ucelay-Gomez R, Fischlein T. Clinical in vitro endothelialization of
Freischlag JA, Ahn SS, Baker JD, Machleder HI, Moor WS. femoropopliteal bypass graft: An actuarial follow-up over
Long-term results of infrainguinal revascularization with three years. Vasc Surg 1994; 19:540-548.
polytetrafluorethylene: A ten-year experience. J Vasc Surg 12. Fischlein T, Zilla P, Meinhart J, Puschmann R, Vesely M,
1992; 16:209-217. Eberl T, Balon R, Deutsch M. In vitro endothelialization
4. Herring M, Gardner A, Glover J. A single-staged technique of a mesosystemic shunt: A clinical case report. Vasc Surg
for seeding vascular grafts with autogenous endothelium. 1994; 19:549-554.
Surgery 1978; 84:498-504. 13. Deutsch M, Meinhart J, Vesely M, Groscurth P, Von
5. Vici M, Pasquinelli G, Preda P, Martinelli G, Gibellini D, Oppell U, Zilla P. In vitro endothelialization of ePTFE
Freyrie A, Curti T, D’Addato M. Electron microscopic and grafts: A clinical case report after 41 months of implanta-
immunocytochemical profiles of human sucutaneous fat tion. Vasc Surg 1997; 25:757-763.
tissue microvascular endothelial cells. Ann Vasc Surg 1993; 14. Kaehler J, Zilla P, Fasol R, Deutsch M, Kadletz M.
7:541-548. Precoating substrate and surface configuration determine
194 Tissue Engineering of Prosthetic Vascular Grafts
adherence and spreading of seeded endothelial cells on 17. Zilla P, Fasol R, Deutsch M, Fischlein T, Minar E,
polytetrafluoroethylene grafts. J Vasc Surg 1989; 9:535-541. Hammerle A, Krupicka O, Kadletz M. Endothelial cell
15. Zilla P, Fasol R, Preiss P, Kadletz M, Deutsch M, Schima seeding of polytetrafluoroethylene vascular grafts in hu-
H, Tsangaris S, Groscurth P. Use of fibrin glue as a sub- mans: A preliminary report. J Vasc Surg 1987; 6:535-541.
strate for in vitro endothelialization of PTFE vascular 18. Meinhart J, Deutsch M, Zilla P. Eight years of clinical
grafts. Surgery 1989; 105:515-522. endothelial cell transplantation. Closing the gap between
16. Franke RP, Graefe M, Schnittler H, Seiffge D, Mittermayer prosthetic grafts and vein grafts. ASAIO J 1997;
C. Induction of human vascular endothelial stress fibres 43:M515-M521.
by fluid shear stress. Nature 1984; 307:648-649.
PART III
Biointeractive Prostheses: Complete Healing
Biological Components
Taming of Adverse Responses
Prevention of the Inflammatory Reaction
CHAPTER 19
Inflammatory Reaction: The Nemesis of Implants
James M. Anderson
Introduction
N emesis is the Greek goddess of retributive justice or vengeance. Thus, the term “nemesis”
has been used to identify one that inflicts retribution or vengeance. Alternatively, nem-
esis has been defined as a source of harm or destruction. Following the implantation of a
medical device, biomaterial or prosthesis, the inflammatory reaction is the first host defense
system which interacts with the medical device, biomaterial or prosthesis, and may be the
source of harm or destruction to the implant or may result in untoward inflammatory and
healing responses which lead to failure of the device in its intended function. In considering
the inflammatory response to prosthetic vascular grafts, the inflammatory response is a com-
plex series of cellular and humoral interactions which are time-dependent and lead to the
healing response. Given these perspectives, the purpose of this chapter is to review the in-
flammatory response to prosthetic vascular grafts and provide a current state of the art
perspective on the inflammatory response to prosthetic vascular grafts and the potential for
inflammatory response interactions with tissue-engineered prosthetic vascular grafts.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
198 Tissue Engineering of Prosthetic Vascular Grafts
recapitulate itself, with the formation of intimal, medial and While little is known regarding the acute and chronic
adventitial components of the vessel wall and the inflammatory responses to the luminal surfaces of vascular
reestablishment of an endothelial lining which separated grafts, some understanding of the series of events following
blood from vascular wall components with their attendant injury in the inflammatory and wound healing responses to
adverse blood-tissue interactions.4,5 This concept has not the porous periadventitial surface of vascular grafts is ap-
been realized in humans to date, although use of highly po- preciated. Acute inflammation is of relatively short dura-
rous ePTFE vascular grafts have produced endothelial lin- tion, lasting from minutes to days, and is characterized by
ings in baboons. In lower species, i.e., rabbits, pigs, dogs and the exudation fluid and plasma proteins and the immigra-
others, the re-endothelialization process does occur on an tion of leukocytes, predominantly neutrophils. Neutrophils
inner capsule (pseudointima) and thus constitutes a are short lived and disintegrate and disappear after 24-48 h,
neointima in these species. depending on the form and topography of the vascular graft.
Immediately following injury, changes occur in vas- Highly porous prostheses may have a longer lived neutro-
cular flow, caliber and permeability. Fluid, proteins and blood phil response, as these cells migrate into the porous body of
cells escape from the vascular system into the injured tissue the vascular graft. This is especially true with loosely knit-
in a process called exudation. Following changes in the vas- ted and velour Dacron vascular grafts and has not been a
cular system, which also include changes induced in blood prominent observation with ePTFE grafts.
and its components, cellular events occur and characterize The accumulation of leukocytes, in particular neutro-
the inflammatory response. Although injury initiates the phils and monocytes, is the most important feature of the
inflammatory response, chemicals and agents released from inflammatory reaction. Leukocytes accumulate through a
plasma, cells and injured tissue, as well as blood, mediate series of processes including margination, adhesion,
the response to vascular grafts. integration, phagocytosis, and extracellular release of
Figure 19.1 illustrates the temporal variation in the leukocyte products. Leukocyte immigration is controlled in
tissue response continuum, i.e., the acute inflammatory re- part by chemotaxis, which is the unidirectional migration
sponse, chronic inflammatory response, granulation tissue of cells along the chemical gradient. A wide variety of exog-
development and foreign body reaction to implanted vas- enous and endogenous substances have been identified as
cular grafts. The intensity and time variables are dependent chemotactic agents. Important to the immigration is move-
upon the extent of injury created in the implantation and ment of leukocytes, as is the presence of specific receptors
the size, shape and topography, and chemical and physical for chemotactic agents on the cell membranes of leukocytes.
properties of the biomaterial as well as the porosity of the These and other receptors may also play a role in the activa-
vascular graft. tion of leukocytes.
Blood protein adsorption to the luminal,
periadventitial and interstitial surfaces of porous vascular
graft biomaterials is a significant early response which is
Table 19.1. Sequence of local events following poorly understood. The protein adsorption phenomenon
implantation occurs prior to any cellular interaction. Prior to the immi-
gration of leukocytes to and within the vascular graft, blood
Injury protein has coated the surfaces of the graft material and thus
Acute inflammation presents a protein-modified surface to cellular interactions.
Chronic inflammation
The physical and chemical characteristics of the vascular
Granulation tissue
Foreign body reaction graft material itself may play little role in subsequent cellu-
Fibrosis lar and humoral responses. Obviously, preclotting of the
vascular graft accomplishes the protein precoating in an ex- studies with human blood and vascular grafts. Heparin was
cessive fashion.6 used as the anticoagulant for these studies; this anticoagu-
To better understand the early blood protein adsorp- lant permits complement activation with subsequent leu-
tion onto vascular graft materials, we have utilized a surface kocyte activation. The utilization of citrate, EDTA or EGTA
radioimmunoassy technique and an immunogold labeling will deplete or bind calcium ions significantly, thus inhibiting
technique with scanning electron microscopy to character- complement activation by the calcium-dependent pathway.
ize human blood protein adsorption onto Dacron and ePTFE With highly porous grafts, i.e., knits and velours, acute
vascular grafts in an in vitro human blood recirculation sys- and chronic inflammatory cell infiltration into the interstices
tem.7-10 The detection of the various proteins deposited on of the vascular grafts occurs. The time dependent nature of
the surfaces of the Dacron, ePTFE and control materials fell these responses and the extent of their duration within the
into two groups: Fibrinogen, IgG and albumin were found interstices of the vascular grafts are dependent upon the
in the highest amounts; Hageman factor (factor XII), porosity and structure of the vascular grafts. As described
factor VIII/vWF, fibronectin and hemoglobin were found earlier, preclotting of Dacron vascular grafts provides a bar-
in moderate amounts. Of the proteins assayed, IgG showed rier to this response and little is known regarding inflam-
the greatest degree of adsorption and affinity to all of the matory cell infiltration into preclotted vascular grafts.
materials tested and particularly to Dacron and ePTFE. Af- In the early 1980s, Meadox Medicals provided a col-
ter exposure of the tested materials for five minutes, IgG ac- lagen coated Dacron vascular graft, Hemashield with
counted for approximately 25% of the radioactivity bound Microvel, in which a thin layer of collagen was present on
to Dacron and to ePTFE. In the recirculation system, sig- the luminal and adventitial surfaces as well as within the
nificant protein adsorption had occurred within 5 minutes interstices of the vascular graft. This simple concept, origi-
of blood contact and did not significantly increase out to nally considered to replace preclotting of the Dacron vascu-
180 minutes. Fibrinogen adsorption to Dacron and ePTFE lar grafts, also had a significant inhibition of the inflamma-
decreased as the time of exposure to blood increased. tory cell infiltrate in the early response. The collagen coat-
Hageman factor (factor XII) significantly decreased on ing converted a highly porous material into a nonporous
Dacron as the time of exposure increased, but its adsorption material which developed its porosity in vivo by the bio-
to other materials, i.e., ePTFE and silica-free PDMS, degradation of the collagen coating. The biodegradation of
remained constant over the 180 minute time period. the collagen coating occurs within 4-12 weeks, and thus po-
Adsorption of IgG to Dacron decreased with time, but IgG rosity develops after the inflammatory responses and dur-
adsorption to ePTFE and PDMS did not change over ing the granulation tissue and fibrosis phases of the healing
180 minutes. response. As the porosity develops, macrophages and fibro-
These studies demonstrate that early adsorption of blasts present at the graft/tissue interface migrate into the
blood proteins which are adhesion receptor ligands vascular graft to produce the foreign body reaction and fi-
(fibronectin, fibrinogen), opsonins which can facilitate brosis without having to contend with the detritus of the
complement activation (IgG), and significant coagulation inflammatory reaction.12 The collagen coated Dacron grafts
cascade proteins (Hageman factor) adsorb quickly and are an early example of vascular graft tissue engineering.
readily to the luminal surfaces, where they may participate
in subsequent inflammatory, coagulation and throm- Foreign Body Reaction
botic processes. The foreign body reaction consisting of macrophages
Our human blood recirculation system has been also and foreign body giant cells develops at the periadventitial
utilized to investigate vascular graft-associated complement tissue-material interface coincidentally with the development
activation and leukocyte adhesion.11 Materials tested were of granulation tissue, with its attendant fibroblast prolifera-
ePTFE, crimped Dacron Bionit (C.R. Bard) and preclotted tion and neovascularization, i.e., capillary formation. At the
Dacron Bionit. A decrease in the total blood leukocyte con- same time, smooth muscle cells and fibroblasts from the wall
centration with perfusion time was seen for all materials of the native artery migrate and proliferate along the sur-
tested, and increasing leukocyte adhesion to the graft sur- face of the vascular graft to produce a pseudointima. En-
face was observed by scanning electron microscopy. The most dothelial cells also migrate and proliferate from the native
dramatic decrease in leukocyte concentration was observed artery at the anastomosis, but in humans their migration
for the interaction of heparinized whole blood with Dacron. into the vascular graft is limited in extent as compared to
This was due to the selective decrease in neutrophils and animal models where endothelialization is extensive. This
monocytes, and was correlated with an increase in both leu- lack of endothelialization of the pseudointimal surface of
kocyte adhesiveness and complement activation, as mea- the vascular graft in humans has remained a challenge and a
sured by C5a elevation. Minimal complement activation or profound problem with human vascular grafts. Of special
leukocyte adhesion was observed for ePTFE and the silicone note is the apparent lack of inflammatory cell interaction at
rubber control. Interestingly, Dacron Bionit exhibited the the luminal surface of human vascular grafts.13 Given the
highest C5a levels when compared to Dacron Bionit which potential for complement activation and leukocyte adhe-
had been preclotted in whole blood. Dacron Bionit sion, one might have expected early monocyte adhesion and
preclotted in platelet-poor plasma gave C5a levels which were differentiation into macrophages which can fuse into for-
slightly greater than ePTFE which was comparable to the eign body giant cells at the luminal surfaces of vascular graft
silicone rubber control. These studies demonstrated the sig- materials. Studies of retrieved human vascular grafts, how-
nificance of utilizing an appropriate anticoagulant in in vitro ever, suggest that this sequence of events does not occur, as
200 Tissue Engineering of Prosthetic Vascular Grafts
foreign body giant cells are not identified at the material- cretion of extracellular matrix materials, and endothelial cell
blood, material-blood protein or material-pseudointimal proliferation, which can affect the hemostatic, thrombotic
interface on the luminal surface of ePTFE vascular grafts. balance, and smooth muscle cell proliferation which may
With Dacron, FBGCs at the luminal surface of the graft participate in anastomotic or pseudointimal hyperplasia.
material are seen following the healing response and This hypothesis developed from two aspects of our research.
neovascularization within the graft interstices. Studies in our First, the consistent and all-inclusive finding that macro-
laboratory have suggested that leukocyte adhesion is depen- phages and foreign body giant cells were present at the in-
dent on both complement C3 and fibronectin deposition, terface of clinically retrieved human vascular grafts regard-
as well as being sensitive to the levels of shear stress in flow- less of implant time pointed toward the importance of the
ing blood.14 High levels of shear stress and/or turbulence, as macrophage and foreign body giant cells in tissue-material
well as low levels of complement C3 and fibronectin depo- interactions. Secondly, our in vivo cage implant studies with
sition, may result in minimal to no leukocyte adhesion, and a wide variety of vascular graft materials, including polyether
thus macrophage development and foreign body giant cell polyurethanes, demonstrated the early—as soon as three
formation at the luminal surfaces is limited. days—adhesion of monocytes/macrophages on the surfaces
The histological evaluation of retrieved human vas- of the materials.
cular grafts shows the presence of macrophages and foreign The second area of research was the first demonstra-
body giant cells interacting with the prosthetic material tion that adherent activated macrophages and foreign body
within the graft interstices and at the periadventitial sur- giant cells on polyether polyurethane surfaces were respon-
face. The foreign body reaction with macrophages and for- sible for the biodegradation of these materials.17,18 This find-
eign body giant cells has been demonstrated as early as sev- ing explained the earlier clinical studies of polyurethane deg-
eral weeks following implantation. In vascular grafts which radation in cardiac pacemaker leads, and also explains the
have been implanted for over two decades, these cells are biodegradation of polyether polyurethanes when they are
still present at the tissue-material interface. As the foreign utilized as vascular graft materials. Numerous academic and
body reaction appears to be virtually an all-inclusive inter- industrial scientists have attempted to utilize polyether poly-
action at soft tissue-material interfaces, we have focused our urethanes as porous and nonporous vascular grafts, but long
attention on the potential roles played by macrophages and term implant studies indicate biodegradation with attendant
foreign body giant cells not only in modulating the wound dilatation of the vascular graft or loss of integrity of the wall
healing response but also their potential participation in fail- of the polyether polyurethane vascular graft. These studies,
ure mechanisms of human vascular grafts, i.e., anastomotic in part, have led to the development of more stable polyure-
hyperplasia and stenosing pseudointimal hyperplasia. thanes by the incorporation of more efficient antioxidants,
In 1984, we began to address the hypothesis that ad- substitution of the degradable polyether soft segments by
herent macrophages which were activated released chemi- more stable soft segment prepolymers such as
cal mediators, in particular cytokines, which in an autocrine, polydimethylsiloxane or polycarbonates, and the chemical
paracrine and endocrine fashion modulated the formation modification of polyether polyurethanes by so-called
of the foreign body reaction at the tissue-material interface “end-capping,” where more stable moieties are bonded to
and the development of the fibrous capsule.3,15,16 Figure 19.2 the chain ends to produce polymers which are inherently
illustrates this early concept, in which activated macrophages less biodegradable.19
on protein-coated biomedical polymer surfaces produce and These new materials are in the early stages of
secrete various cytokines and growth factors which can development and testing for application as vascular graft ma-
modulate platelet activity, fibroblast proliferation and se- terials, and it is anticipated that the desirable elastomeric
and biocompatible properties of the polyurethanes will prove more indicative of the inflammatory reaction at the tissue/
to be efficacious and not undergo inflammatory material interface. Studies under these conditions demon-
cell-mediated biodegradation when utilized in various strated that Dacron and polyethylene exhibited a high reac-
vascular applications. tivity as measured by IL-1 release, ePTFE was intermediate
and Biomer and polydimethylsiloxane were low in their abili-
Macrophage Motility, Adhesion and Activation ties to activate macrophages and release interleukin-1.24 To
The accumulation of macrophages in vascular graft examine the paracrine effect of interleukin-1 on fibroblast
implant sites is achieved through selective cellular and hu- proliferation, we studied the effect of supernatants produced
moral mechanisms that both attract and immobilize mac- by the in vitro interaction of monocytes/macrophages on
rophages and macrophage precursors where their functional various biomedical polymers including Dacron and ePTFE
activity can be directed at resolution and/or repair of tissue. and subsequently measured the fibroblast-stimulating po-
Monocytes contact stimulatory factors within blood vessels tential for these supernatants. For Dacron and ePTFE, the
around the vascular graft and adhere to endothelial cells lin- fibroblast stimulatory potential was equivalent, but the
ing the vessels. These monocytes then pass through the ves- interleukin-1 activity for Dacron was much higher than that
sel wall and into the tissue undergoing transformation in exhibited for ePTFE. Neutralization experiments utilizing
response to the stimuli and differentiate into macrophages. specific antibodies to interleukin-1 in our experiments dem-
Monocyte and macrophage movement within the tissue to- onstrated the potential production and paracrine effects of
ward the site of injury is controlled and directed by chemo- other cytokines and growth factors which may also be pro-
taxis or chemokinesis. Chemokinesis is the accelerated ran- duced in the macrophage activation and secretory process.
dom locomotion in response to, and the speed of turning of Table 19.3 illustrates some monocyte/macrophage
the cells toward, chemical stimuli, whereas chemotaxis is the molecules important in modulation of inflammation and
highly directed movement of cells along a chemical gradi- wound healing with vascular graft materials. The potential
ent. Table 19.2 shows important agents which mediate role for the molecules is shown; these molecules may be ei-
chemotaxis and are integral features of the inflammatory ther secreted to produce an autocrine, paracrine or endo-
process. These agents can both sustain and control the se- crine effect or may be cell membrane associated, such as
verity of the inflammatory response. In addition to dead or important adhesion molecules and receptors which may
dying cells, chemotactic stimuli include fragments derived participate in the adhesion of monocytes/macrophages to
from the activation of complement protein, mediators of protein-coated vascular graft materials or in cell-cell inter-
the kinin, clotting and fibrinolytic systems, and products actions important in the inflammatory and wound healing
produced by leukocytes themselves. processes. Table 19.4 provides a partial list of chemo-
Our early studies on human monocyte/macrophage attractants and growth modulators produced by macro-
activation and interleukin-1 generation by biomedical poly- phages. This table is limited to only those agents thought to
mers were carried out in in vitro cell culture systems where be important in the tissue-material interaction in the in-
the interleukin-1 production was assayed by a biological flammatory response to vascular graft materials. It is note-
thymocyte proliferation biological assay.20-26 In the mid to worthy that interleukin-1 and tissue growth factor-! (TGF-!)
late 1980s, a paucity of reagents was available to study spe- may function as growth agonists or growth antagonists. The
cific cytokines and growth factors produced by macrophages schizophrenic nature of cytokines and growth factors must
and foreign body giant cells in contact with vascular graft be considered when attempting to explain the autocrine,
materials. Later, when radioimmunoassays and ELISAs be- paracrine or endocrine activity of cytokines and growth
came available for interleukin-1 quantitation, these were factors.
incorporated into our studies. It is noteworthy that biomedi- Greisler and colleagues have utilized monocyte/mac-
cal polymers have a very low potential for activating mac- rophage molecules released during the inflammatory and
rophages with the release of interleukin-1. In our studies, wound healing processes to modulate cellular proliferation
we utilized lipopolysaccharide to additionally stimulate the in the wound healing and neointimal response to vascular
adherent monocytes/macrophages, as we believe that this is graft materials.27 In in vitro studies, fibroblast growth fac-
tor type I (FGF-I) and heparin placed into suspensions of
fibrinogen polymerized by addition of thrombin (fibrin
glue) promoted endothelial cell proliferation while suppress-
ing smooth muscle cell proliferation. These studies demon-
Table 19.2. Major mediators of macrophage chemotaxis
strated the potential for controlled release systems coupled
Complement Components
with vascular graft materials which could cellularly engineer
Lymphokines the wound healing response with vascular grafts. Prelimi-
Fibronectin Fragments nary in vivo experiments utilizing growth factor release from
Leukotriene B4 vascular grafts in canines demonstrated this concept and
Interleukins (IL-1, IL-8) identified the potential for the direct control of inflamma-
Endothelial Cell Products tory and healing responses directly by cytokines and growth
Monocyte Chemotactic Peptide 1 (MCP-1) factors.28 From a tissue engineering perspective, this is a
Thrombin
Fibrinopeptides
promising avenue of exploration which requires
additional effort.
202 Tissue Engineering of Prosthetic Vascular Grafts
Table 19.3. Monocyte/macrophage molecules important in the modulation of inflammation and wound healing
Interleukin-1 (IL-1) Receptor Secreted Increased Adhesion Molecule Expression, Smooth Muscle Cell
Migration and Proliferation, Inhibit Endothelial Cell Proliferation
Interleukin-1 Receptor Secreted Decreases Interleukin-1 Response
Antagonist (IL-1Ra)
Interleukin-6 (IL-6) Secreted Activation of Coagulation, Smooth Muscle Cell Secondary Gene
Induction
Interleukin-8 (IL-8) Secreted Recruitment of Lymphocytes and Endothelial Cells
Monocyte Chemoattractant Secreted Monocyte Chemoattractant, Activation of !2 Integrin Receptors
Protein-1 (MCP-1)
Monocyte Colony-Stimulating Secreted Monocyte Chemoattraction, Proliferation, and Survival
Factor (M-CSF)
Granulocyte/Monocyte Colony Secreted Chemoattractant for Monocytes
Stimulating Factor (GM-CSF)
Tumor Necrosis Factor-# (TNF-#) Secreted Increase Endothelial Cell Adhesion Molecule Expression, Monocyte
Chemoattractant, Smooth Muscle Cell Proliferation, Inhibits Endothelial
Cell Proliferation
Tissue Growth Factor-! (TGF-!) Secreted Monocyte Chemoattractant, Increases Smooth Muscle Cell Proliferation
Inhibits Endothelial Cell Proliferation
Interferon-& (INF-&) Secreted Inhibits Endothelial Cell and Smooth Muscle Cell Proliferation
Osteopontin Secreted Cell Adhesion and Migration, Neovessel Formation, and Calcification
SPARC, Osteonectin Secreted Cell Migration, Proliferation, and Neovessel Formation
Thrombospondin Secreted Cell Migration, Neovessel Formation
Oxidized Low Density Secreted Increases Monocyte Chemotaxis and Macrophage Secretion of IL-1 and
TNF-#
Lipoprotein (oxLDL) Increases VCAM-1 Expression on Endothelium
basic Fibroblast Growth Secreted Endothelial Cell Chemoattractant, Increases Endothelial Cell and
Factor (bFGF) Smooth Muscle Cell Proliferation
Vascular Endothelium Growth Secreted Increases Endothelial Cell Proliferation
Factor (VEGF)
Insulin-like growth factor Secreted Smooth Muscle Cell Chemoattractant, Increases Smooth Muscle
(IGF-1) Cell Proliferation
acidic Fibroblast Growth Secreted Endothelial Cell Chemoattraction and Proliferation
Factor (aFGF)
Heparin-Binding Epidermal Secreted Smooth Muscle Cell Chemoattraction and Proliferation
Growth Factor (HB-EGF)
Platelet-Derived Growth Secreted Smooth Muscle Cell Chemoattraction and Proliferation
Factor (PDGF)
Vascular Adhesion Molecule-1 Cell-Associated Monocyte/Macrophage Activation and Retention
(VCAM-1)
Intercellular Cell Adhesion Cell-Associated Monocyte/Macrophage Activation and Retention
Molecule (ICAM-1)
Complement C3bi Receptor Cell-Associated Thrombosis and Macrophage Adhesion
Thrombin Receptor Cell-Associated Monocyte/Macrophage Activation
To complement our in vitro human monocyte/mac- The luminal surface of the vascular graft specimens
rophage activation and cytokine secretion studies, we have showed no to some endothelium at the blood-pseudointima
continued to evaluate retrieved human vascular grafts and interface. The pseudointima (inner capsule) was predomi-
our latest effort involves cytokine and growth factor analy- nantly acellular but isolated areas of fibroblasts and smooth
sis utilizing immunohistochemistry and steady state mRNA muscle cells were identified. In situ hybridization studies
analysis with in situ hybridization techniques. Cytokines and identified IL-1#, IL-1!, TNF-#, TGF-!, and PDGF-A and -B
growth factors/inhibitors studied included interleukin-1# in the pseudointima. The pseudointima-graft interface
(IL-1#), interleukin-1! (IL-1!), tumor necrosis factor-# showed the presence of macrophages and foreign body gi-
(TNF-#), transformtin growth factor-!1 (TGF-!1), and ant cells adjacent to the ePTFE graft nodes and fibrils and
platelet derived growth factor A and B (PDGF-A and -B). Dacron fibers and fiber bundles. These components of the
For analysis, five contiguous zones or areas on or within re- foreign body reaction were localized at the synthetic graft
trieved vascular grafts were evaluated: luminal surface, fiber interface but not in areas distant from the luminal sur-
pseudointima (inner capsule), pseudointima-graft interface face of the graft. The interstitial network of the synthetic
(including the luminal portion of the graft), periadventitia- vascular grafts contained isolated fibroblasts. In situ hybrid-
graft interface (including the outer portion of the graft), and ization and immunohistochemistry analysis identified the
periadventitia (outer capsule). presence of IL-1#, IL-1!, TNF-#, TGF-!, and PDGF-A and
Inflammatory Reaction: The Nemesis of Implants 203
-B in the pseudointima-graft interface. The graft-peri- pressed by macrophages and foreign body giant cells. Steady
adventitial tissue interface exhibited a foreign body reaction state TNF-# mRNA expression was variable, with moderate
with the presence of macrophages and foreign body giant amounts of mRNA and protein being associated with mac-
cells localized at the synthetic graft surface. This interface rophages and foreign body giant cells.
also exhibited the same cytokines and growth factors iden- TGF-!1 mRNA expression was low compared to IL-1#
tified at the pseudointima-graft interface by immunohis- and IL-1! mRNA. Likewise, protein levels of TGF-! as de-
tochemical and in situ hybridization techniques. The termined by immunohistochemistry were also lower than
periadventitial fibrous capsule (outer capsule) showed a well- IL-1# and IL-1!. Fibroblast-like cells in the pseudointima
healed fibrous capsule with fibroblasts and capillaries within and periadventitial fibrous (outer) capsule, and macroph-
a type I collagen matrix. These histopathologic features were ages and foreign body giant cells at the graft-tissue inter-
similar for all types of retrieved human vascular grafts. In faces, were the predominant cells that expressed TGF-!1
situ hybridization and immunohistochemical analysis mRNA. The expression of steady state PDGF-A and -B
showed the presence of IL-1#, IL-1!, TNF-#, TGF-!, and mRNAs was examined, and the relative levels were similar.
PDGF-A and -B within the outer capsule. Relatively low levels were identified, and immunohis-
Overall, for IL-1#, high levels of expression of steady tochemical analysis indicated no or little protein.
state IL-1# mRNA were identified by in situ hybridization, Our preliminary study focused on the utilization of
and protein expression as determined by immunohis- mRNA for transcriptional analysis and cell localization in
tochemistry was also high. Higher levels of steady state IL-1# combination with protein expression by immunohistochem-
mRNA were present in the periadventitial fibrous (outer) istry as an indicator of translational events. Detection of
capsule relative to the pseudointima, the pseudointima-graft IL-1# and -! mRNAs and protein suggests that IL-1 may act
interface and the periadventitia-graft interface. Fibroblast- as a regulatory protein in the induction of intimal/anasto-
like cells were the predominant cell type in the outer cap- motic hyperplasia. In addition, the presence of IL-1# mRNA
sule and expressed higher levels of IL-1# mRNA, as com- in fibroblast-like cells and myofibroblast-like cells suggests
pared to other cell types including smooth muscle cells and that it may function as a paracrine messenger, stimulating
endothelial cells in the capillaries. In the pseudointima, fi- fibroblasts and myofibroblasts to proliferate and differenti-
broblast-like cells expressed high levels of IL-1# mRNA, ate. Activated fibroblasts also produce a number of soluble
whereas smooth muscle cells did not appear to express IL-1# mediators including bFGF, PDGF and IL-1 which can acti-
mRNA. In contrast to the pseudointima and the vate other inflammatory cells in addition to smooth muscle
periadventitial fibrous capsule, macrophages and foreign cells. Although not as strong as IL-1# mRNA, steady state
body giant cells in the pseudointima-graft and IL-1! mRNA was identified and may stimulate smooth
periadventitia-graft interfaces were predominant cell types muscle cell proliferation in a paracrine fashion.
that expressed IL-1# mRNA and protein. Relative levels of Transformtin growth factor-!1 (TGF-!1) was identi-
IL-1! mRNA and protein detected in Dacron and ePTFE fied at low levels of steady state mRNA expression in macro-
vascular graft sections were similar to that of the IL-1# phages and foreign body giant cells. TGF-!1 is an important
mRNA detected. Overall, the periadventitial fibrous (outer) regulator of the duration of the inflammatory response and
capsule expressed higher levels of steady state IL-1! mRNA also is a potent inhibitor of cell proliferation, as well as a
relative to the other areas. However, with the ePTFE vascu- stimulator of connective tissue formation by cells. The en-
lar grafts, very low levels of IL-1! mRNA were found in all hanced activity of macrophages and foreign body giant cells
of the areas studied. IL-1! mRNA was predominantly ex- was exhibited by the detection of steady state TNF-# mRNA.
204 Tissue Engineering of Prosthetic Vascular Grafts
TNF-# is known to enhance the phagocytic activity of macro- fibronectin, for which monocytes also express multiple types
phages and foreign body giant cells. Our in situ hybridiza- of receptors.
tion study found low levels of steady state mRNAs for Because of experimental complexities in dealing with
PDGF-A and -B in macrophages and foreign body giant cells, the three dimensional structures of Dacron and ePTFE, we
and very little to no protein was detected by immuno- initiated our studies utilizing chemically-modified polysty-
histochemistry. rene surfaces to explore the role of surface chemistry in
The detection of various cytokine mRNAs and pro- monocyte adhesion, macrophage phenotypic expression and
tein implicates cytokine-mediated interactions of cellular foreign body giant cell formation.29 Human monocyte in
components in the ePTFE and Dacron vascular grafts. The vitro adhesion with fluorinated, siliconized, nitrogenated and
paracrine interaction mediated by IL-1 in macrophages/ oxygenated surfaces were reduced by 50-100% when comple-
foreign body giant cells and fibroblasts/myofibroblasts with ment component C3-depleted serum was used for adsorp-
vascular smooth muscle cells is strongly suggested by tion. The fluorinated surfaces exhibited the greatest inhibi-
these findings. tion of monocyte adhesion with C3-depleted serum. Mono-
cyte adhesion was restored on all surfaces when C3-depleted
Foreign Body Giant Cell Formation serum was replenished with purified C3. Monocyte adhe-
As part of our ongoing investigation into the role of sion to serum-adsorbed surfaces was inhibited by mono-
the monocyte/macrophage in biocompatibility, a major goal clonal antibodies to the leukocyte integrin beta subunit,
has been to identify the adhesion and fusion mechanisms CD18, and partially inhibited by a monoclonal antibody to
that initiate and promote the observed in vivo morphologic the alpha subunit Cd11b. These findings suggest adhesive
progression of monocyte to macrophage to foreign body interactions between adsorbed C3bi, the hemolytically in-
giant cell on biomaterials and vascular graft materials. Ad- active form of the C3b fragment, and the leukocyte integrin
herent monocyte-derived macrophages and foreign body Cd11b/cd18. Additional studies demonstrated that adsorbed
giant cells (FBGCs), formed by macrophage fusion, are fibrinogen reduced the effectiveness of these inhibiting
prominent and persistent cell types on implanted vascular monoclonal antibodies, indicating that alternative adhesion
grafts and, through their numerous secretory capacities, are mechanisms may operate, depending on the critical adhe-
believed to exert multiple and complex influences on the sion-mediating components adsorbed onto the different
inflammatory response at the implant site and on the surfaces.
biocompatibility of vascular grafts. Although monocyte ad- Recent studies in our laboratory have implicated IL-4
hesion to implanted vascular grafts is critical to and IL-13 cytokines in the in vitro formation of foreign body
biocompatibility outcome, as it initiates macrophage devel- giant cells derived from human monocytes/macro-
opment and FBGC formation, it is unknown how mono- phages.30-33 These studies have resulted in the development
cytes recognize biomaterials and how surface properties of an in vitro culture system that may now be utilized for
might influence this event. testing various materials for their ability to participate in
Material surface property-dependent blood protein foreign body giant cell formation. When IL-4 or IL-13 are
adsorption occurs immediately upon surgical implantation added to human monocyte cultures at day 3, foreign body
of a vascular graft, and it is the protein-modified biomate- giant cells containing as many as 100 nuclei and measuring
rial that inflammatory cells subsequently encounter. Mono- 1.0 mm2 in surface area are formed on various modified sur-
cytes express receptors for various blood components, but faces. Critical to these experiments was the addition of the
they recognize naturally occurring foreign surfaces by re- cytokine at day 3 of the culture and not at day 0 of the cul-
ceptors for opsonins such as fragments of complement com- ture. Simultaneous addition of the cytokine and the cells at
ponent C3. Because complement activation by biomaterials the beginning of the culture does not lead to foreign body
has been well documented, we investigated monocyte inter- giant cell formation and may result in the lack of mono-
actions with foreign surfaces. Exposure to blood during vas- cyte/macrophage adhesion to the substrate. This sequential
cular graft implantation may permit extensive opsonization addition of the cytokine points out the importance of
with the labile fragment C3b and the rapid conversion of temporal variations which may occur in addressing cytokine/
C3b to its hemolytically inactive but nevertheless opsonic cell interactions.
and more stable form, C3bi. C3b is bound by the Cd35 re- IL-4 and IL-13 are lymphokines secreted predomi-
ceptor, but C3bi is recognized by distinct receptors, nantly by Th2 lymphocytes in humans. Thus, lymphocytes
Cd11b/cd18 and Cd11c/cd18. Fibrinogen, a major plasma present transiently in the early chronic inflammatory phase
protein that adsorbs to vascular grafts, is another described of the inflammatory response may participate in the for-
ligand for these molecules, which together with Cd11a/cd18 eign body reaction through facilitation of the formation of
constitutes a subfamily of integrins that is restricted to leu- foreign body giant cells. IL-4 and IL-13 downregulate the
kocytes. Studies with monoclonal antibodies to their com- secretion of proinflammatory cytokines such as IL-1, IL-6
mon !2 subunit (CD18) and distinct # chains (Cd11a,b,c) and TNF-#; growth factors such as granulocyte-macrophage
have implicated Cd11a/cd18 in cell-cell adhesive interactions CSF and granulocyte CSF; and chemokines such as IL-8 and
and Cd11b/cd18 and Cd11c/cd18 in multiple phagocytic cell macrophage inflammatory protein-1#. Fc&R expression, re-
responses. Other potential adhesion-mediating proteins that active oxygen intermediate secretion and cytotoxic activi-
adsorb to biomaterials include IgG, which may interact with ties are also inhibited by these cytokines. Conversely, IL-4
monocytes via receptors to its Fc constant region, and and IL-13 upregulate monocyte/macrophage production of
Inflammatory Reaction: The Nemesis of Implants 205
IL-1R antagonist, antigen-presenting capacity and cell sur- The second major barrier that must be confronted is
face expression of several adhesion molecules including the highly complex and time-dependent nature of the cel-
Cd11b/cd18 and Cd11c/cd18. Although IL-4 and IL-13 nega- lular and humoral components of the inflammatory re-
tively regulate several cytotoxic and inflammatory functions sponses. Our approach has been to utilize human material
of monocytes/macrophages, they are not generally viewed whenever possible, but this limits one to in vitro experiments.
as downregulators of monocyte/macrophage activity. As humans cannot ethically be used in preliminary experi-
The role for IL-4 in foreign body giant cell formation ments to test the safety of devices, animal models must be
in vivo has been demonstrated in a rat cage implant system utilized. The question resulting from the use of animal
with polyurethane.31 Utilization of both recombinant IL-4 models is the equivalency of, and similarity and differences
and anti-IL-4 injected into the cage demonstrates the en- between, the animal model and the response in humans. Not
hancement and inhibition of foreign body giant cell forma- only must the various components of the inflammatory
tion on the polyurethane surface, respectively. The in vivo response be considered, but also the extent and duration of
studies also point out the lack of effect by IL-4 in facilitating the components of the inflammatory response and the
macrophage adhesion to the polyurethane surface. Thus, the time-dependent nature, i.e., kinetics, of the inflammatory
IL-4 appears to facilitate foreign body giant cell formation responses and their components. These factors may or may
through mechanisms not related to the surface density of not vary when comparing animal model responses to
adherent macrophages. human responses.
Complementary experiments with IL-4-induced These barriers and lack of knowledge of inflamma-
macrophage fusion in foreign body giant cell development tory responses offer new challenges that must be met in the
has shown the prevention of macrophage fusion and the for- research and development of tissue-engineered prosthetic
mation of foreign body giant cells by the use of inhibitors of vascular grafts. New paradigms must be developed with new
mannose receptor activity. # -Mannan and synthetic perspectives based on new information, and older para-
neoglycoprotein conjugates inhibit the fusion of macro- digms, while comfortable to some investigators, may not
phages to form foreign body giant cells. Inhibition of mac- adequately and appropriately address the significant scien-
rophage activity in the formation of foreign body giant cells tific issues that are necessary in the development of tissue-
may be facilitated by blocking inhibitors which participate engineered prosthetic vascular grafts.
in the macrophage fusion process. This offers a pathway by A third major barrier facing us in the development of
which the foreign body reaction may be controlled. tissue-engineered prosthetic vascular grafts is the adequate
and appropriate evaluation of new concepts or products
Future Perspectives on Inflammatory from a safety and efficacy perspective. Unique devices will
Responses to Tissue Engineered Prosthetic require unique testing and evaluation protocols. Appropri-
Vascular Grafts ate rationale and justification must be provided for the in-
The future development of tissue engineered pros- clusion or omission of test methods, which will be based on
thetic vascular grafts requires new knowledge and informa- the unique characteristics of the tissue-engineered prosthetic
tion on normal and directed healing mechanisms, biologi- vascular graft under consideration. New materials and de-
cal signals and signal mechanisms, and delivery and pheno- vices designed for deliberate and specific interactions with
typic expression of cells in and on tissue-engineered pros- blood and tissue components must be based on those delib-
thetic vascular grafts. Research efforts in the tissue engineer- erate and specific interactions, which are a component of
ing area must be directed towards developing new informa- the design of the tissue engineered prosthetic vascular graft.
tion, i.e., biologically-based design criteria, and an expan- In the development of tests for tissue-engineered prosthetic
sion of our current knowledge base regarding these processes vascular grafts, it is important that we emphasize the com-
and mechanisms. There is no doubt that we are exception- plex and interactive milieu of the cellular and humoral com-
ally limited in our knowledge base as it applies to the re- ponents that interact or are anticipated to interact with the
search and development of tissue-engineered prosthetic vas- tissue-engineered prosthetic vascular graft. We must appre-
cular grafts. In considering the early responses to implanted ciate the interactive environment provided by the numer-
tissue engineered prosthetic vascular grafts, emphasis must ous humoral enzyme systems (kinin, fibrinolytic, coagula-
be placed on developing an understanding of the inflam- tion and complement), formed elements (platelets), and cells
matory response interactions with the tissue-engineered (polymorphonuclear leukocytes, monocytes, lymphocytes
prosthetic vascular graft with its unique characteristics. As and eosinophils) which provide for positive (activating) and
such, the evaluation must take into consideration the unique negative (inhibiting) interactions. Failure to appreciate these
function and application of the tissue-engineered prosthetic types of interactions and feedback control mechanisms may
vascular graft. A major hurdle to this effort is the apparent lead to false interpretations of results from single or isolated
species differences which exist with prosthetic vascular grafts test methods. Furthermore, emphasis must be placed on the
and in particular the process of endothelialization which quantitative determination of parameters in the biological
occurs in primates and other mammals, but not in human response evaluation. As our knowledge base for the research
prosthetic vascular grafts. Coupled with this problem is the and development of new tissue-engineered prosthetic vas-
lack of knowledge regarding the phenotypic expression of cular grafts increases, enhanced integration of disciplines
cellular components in the inflammatory response in dif- affecting not only the basic research but also the develop-
ferent species. ment of test methods will be necessary.
206 Tissue Engineering of Prosthetic Vascular Grafts
It is our perspective that the inflammatory response and Ratner B, eds. Polymers as Biomaterials. New
to prosthetic vascular grafts and tissue-engineered prosthetic York:Plenum Press, 1984:209-223.
vascular grafts is not necessarily a nemesis. In the past, we 17. Zhao Q, Agger MP, Fitzpatrick M et al. Cellular interac-
have tended to label the inflammatory response as a nem- tions with biomaterials: in vivo cracking of prestressed
Pellethane 2363-80A. J Biomed Mater Res 1990;
esis, i.e., source of harm or destruction, based upon our in-
24:621-637.
complete knowledge of the inflammatory and wound heal- 18. Zhao Q, Topham N, Anderson JM et al. Foreign-body
ing responses. With the expansion of our knowledge base of giant cells and polyurethane biostability: in vivo correla-
inflammatory cell interactions with prosthetic vascular grafts tion of cell adhesion and surface cracking. J Biomed Mater
and tissue-engineered prosthetic vascular grafts, we can an- Res 1991; 25:177-183.
ticipate that the new and expanded knowledge base of in- 19. Mathur AB, Collier TO, Kao WJ et al. In vivo
flammatory responses and interactions will be utilized to biocompatibility and biostability of modified polyure-
modify inflammatory cell responses to these new devices, thanes. J Biomed Mater Res 1997; 36:246-257.
and possibly convert a foe into a friend or ally. 20. Miller KM, Anderson JM. Human monocyte/macrophage
activation and interleukin 1 generation by biomedical
References polymers. J Biomed Mater Res 1988; 22:713-731.
1. Anderson JM. Mechanisms of inflammation and infection 21. Miller KM, Huskey RA, Bigby LF, Anderson JM. Charac-
with implanted devices. Cardiovasc Pathol 1993; terization of biomedical polymer-adherent macrophages:
2:199S-208S. Interleukin 1 generation and scanning electron micros-
2. Anderson JM. Inflammation and the foreign body re- copy studies. Biomaterials 1989; 10:187-196.
sponse. Problems in General Surgery 1994; 11:147-160. 22. Bonfield TL, Colton E, Anderson JM. Plasma protein
3. Anderson JM. Inflammatory response to implants. ASAIO adsorbed biomedical polymers: Activation of human
J 1988; 11:101-107. monocytes and induction of interleukin 1. J Biomed Mater
4. Wesolowski SA. Evalulation of Tissue and Prosthetic Vas- Res 1989; 23:535-548.
cular Grafts. Springfield:Charles C. Thomas, 1962. 23. Miller KM, Anderson JM. In vitro stimulation of fibro-
5. Wesolowski SA and Dennis C, eds. Fundamentals of Vas- blast activity by factors generated from human monocytes
cular Grafting. New York:McGraw-Hill Book Company, activated by biomedical polymers. J Biomed Mater Res
Inc., 1963. 1989; 23:911-930.
6. Yates SG, Barros AAB, Berger K et al. The preclotting of 24. Miller KM, Rose-Caprara V, Anderson JM. Generation of
porous arterial prostheses. Ann Surg 1978; 188:611-622. IL1-like activity in response to biomedical polymer im-
7. Ziats NP, Pankowsky DA, Tierney BP et al. Adsorption plants: A comparison of in vitro and in vivo models. J
of Hageman factor (factor XII) and other human plasma Biomed Mater Res 1989; 23:1007-1026.
proteins to biomedical polymers. J Lab Clin Med 1990; 25. Bonfield TL, Colton E, Marchant RE, Anderson JM.
116:687-696. Cytokine and growth factor production by monocytes/
8. Pankowsky DA, Ziats NP, Topham NS et al. Morphologi- macrophages on protein preadsorbed polymers. J Biomed
cal characteristics of adsorbed human plasma proteins on Mater Res 1992; 26:837-850.
vascular grafts and biomaterials. J Vasc Surg 1990; 26. Bonfield TL, Anderson JM. Functional versus quantita-
11:599-606. tive comparison of IL-1! from monocytes/macrophages
9. Anderson JM, Bonfield TL, Ziats NP. Protein adsorption on biomedical polymers. J Biomed Mater Res 1993;
and cellular adhesion and activation on biomedical poly- 27:1195-1199.
mers. Int J Artif Org 1990; 13:375-382. 27. Greisler HP. Growth factor release from vascular grafts. J
10. Ziats NP, Topham NS, Pankowsky DA, Anderson JM. Contr Rel 1996; 39:267-280.
Analysis of protein adsorption on retrieved human vas- 28. Kang SS, Gosselin C, Ren D, Greisler HP. Selective stimu-
cular grafts using immunogold labelling with silver en- lation of endothelial cell proliferation with inhibition of
hancement. Cells & Materials 1991; 1:73-82. smooth muscle cell proliferation by FGF-1 plus heparin
11. Kottke-Marchant K, Anderson JM, Miller KM et al. Vas- delivered from fibrin glue suspensions. Surgery 1995;
cular graft associated complement activation and leuko- 118:280-287.
cyte adhesion in an artificial circulation. J Biomed Mater 29. McNally AK, Anderson JM. Complement C3 participa-
Res 1987; 21:379-397. tion in monocyte adhesion to different surfaces. Proc Natl
12. Anderson JM. Microvel with Hemashield vascular grafts. Acad Sci USA 1994; 91:10119-10123.
A preliminary report of the healing response in humans. 30. McNally AK, Anderson JM. Interleukin-4 induces foreign
Angio Archiv 1985; 9:73-77. body giant cells from human monocytes/macrophages.
13. Anderson JM, Abbuhl MF, Hering T, Johnston KH. Im- Differential lymphokine regulation of macrophage fusion
munohistochemical identification of components in the leads to morpholgoical variants of multinucleated giant
healing response of human vascular grafts. ASAIO J 1985; cells. Amer J Pathol 1995; 147:1487-1499.
8:79-85. 31. Kao WJ, McNally AK, Hiltner A, Anderson JM. Role for
14. Kao WJ, Sapatnekar S, Hiltner A, Anderson JM. Comple- interleukin-4 in foreign-body giant cell formation on a
ment-mediated leukocyte adhesion on poly(etherurethane poly(etherurethane urea) in vivo. J Biomed Mater Res
ureas) under shear stress in vitro. J Biomed Mater Res 1995; 29:1267-1276.
1996; 32:99-109. 32. McNally AK, DeFife KM, Anderson JM. Interleukin-4-in-
15. Hering TM, Marchant RE and Anderson JM. Type V col- duced macrophage fusion is prevented by inhibitors of
lagen during granulation tissue development. Experimen- mannose receptor activity. Am J Pathol 1996; 149:975-985.
tal and Molecular Pathology 1983; 39:219-229. 33. DeFife KM, McNally AK, Colton E, Anderson JM.
16. Marchant RE, Miller KM, Hiltner A, Anderson JM. Se- Interleukin-13 induces human monocyte/macrophage fu-
lected aspects of cell and molecular biology of in vivo sion and macrophage mannose receptor expression. J
biocompatibility. In: Shalaby S, Hoffman A, Horbett T Immunol 1997; 158:3385-3390.
Taming of Adverse Responses
Prevention of the Inflammatory Reaction
CHAPTER 20
Molecular Determinants of
Acute Inflammatory Responses to Biomaterials
Liping Tang, John W. Eaton
Introduction
D espite the fact that most biomaterials are inert, nontoxic and nonimmunogenic, bioma-
terial implants often cause adverse reactions. Typically, shortly after implantation, bio-
material surfaces attract a layer of phagocytic cells (especially, macrophages/monocytes and
neutrophils)1-4 and fibroblast-like cells.5 This very much resembles the acute inflammatory
response seen with many other types of foreign bodies and, to an extent, with focal bacterial
infections. The acute inflammatory responses to tissue contact biomaterials are very often
followed by chronic inflammation6-8 and the appearance of fibrotic tissue surrounding many
types of implants.7,9-18 Chronic inflammation and fibrosis are also associated with the deg-
radation and failure of many types of implants, including pacemaker leads,19-21 mammary
prostheses,22,23 temporomandibular24,25 and other joint implants.26
At first glance, these inflammatory responses to inert, nonimmunogenic and non-
toxic materials are difficult to understand. Why should such unreactive and so-called
“biocompatible” materials cause recruitment and evident activation of phagocytes? The fol-
lowing represents a brief summary of our present understanding of the inflammatory re-
sponses to implanted biomaterials.
Surface-Protein Interactions
Because biomaterial surfaces spontaneously and rapidly acquire a layer of host pro-
teins upon contact with blood or proteinaceous body fluids, it is generally believed that
these absorbed proteins likely influence, or absolutely dictate, the ensuing adverse
responses.27-30 Because this protein adsorption occurs much faster than the arrival of cells
on the foreign surface, host cells almost certainly interact with surface-adsorbed proteins,
rather than with the material itself.29-31 Therefore, the types and states of adsorbed proteins
are probably critical determinants of biocompatibility.30,32-35
The composition of adsorbed proteins varies with different biomaterials.29,36 Unfor-
tunately, due in part to the bewildering variety of model materials and protein types which
have been used to investigate protein adsorption by biomaterials, the general principles gov-
erning surface-protein interactions under real life conditions are by no means clear. Gener-
ally speaking, albumin, fibrinogen and immunoglobulin G (IgG) predominate on many
types of blood-contact biomaterials, including Dacron® (a woven form of polyester tereph-
thalate; hereafter, ‘PET’), expanded polytetrafluoroethylene, polydimethylsiloxane (‘silastic’),
polyurethanes and polyethylene.29,36,37 Especially on hydrophobic surfaces such as these
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler
©1999 R.G. Landes Company.
208 Tissue Engineering of Prosthetic Vascular Grafts
(which are typical of the majority of implanted biomaterials), dant protein on biomaterial surfaces, can “passivate”
the adsorbed proteins may undergo conformational changes biomaterials, blunting both inflammatory and thrombogenic
over a period of hours, becoming tightly adherent.38-41 These responses.1,33,53,58 We therefore initially hypothesized that
conformational changes of adsorbed proteins have been IgG, which is also adsorbed on biomaterial surfaces in large
detected using many different techniques, including sodium amounts,29,31,36,53 might be the crucial mediator of phago-
dodecyl sulfate (SDS) elution,42,43 scanning angle reflecto- cyte responses to biomaterial implants.
metry,44 scanning force microscopy,45 attenuated total re-
flectance Fourier transform infrared spectroscopy39 and dif- Adsorbed IGg Is Not Required for Phagocyte
ferential scanning calorimetry.46 It has been assumed that, Accumulation on Biomaterial Implants
upon binding to hydrophobic surfaces, many proteins tend The possible involvement of adsorbed IgG in acute
to unfold47 and, within a short period, undergo surface “de- inflammatory responses to biomaterial implants was sup-
naturation” (perhaps via loss of the normal sphere of hy- ported by several observations. First, immunoglobulins are
dration encouraged by contact with the hydrophobic sur- important in the opsonization of foreign particles, which
face).48 The resulting conformational changes of adsorbed makes them more readily ingested by phagocytes.59 Second,
protein may expose hidden epitope(s) which, in turn, help both surface-bound and heat-denatured IgG and IgA are
initiate adverse reactions such as coagulation and inflam- potent activators of human neutrophils, causing the release
mation.49-52 However, a direct link between protein confor- of lysosomal enzymes (lysozyme, !-glucuronidase and
mational changes and these adverse responses has not been myeloperoxidase) and stimulation of phagocyte oxidative
established. metabolism. 60-62 Finally, in our experimental animal
implantation model we found that IgG-coated surfaces ac-
Which Adsorbed Host Protein(s) Might Be Important cumulated as many inflammatory cells (both neutrophils
in Mediating Inflammatory Responses to Biomaterial and macrophages/monocytes) as did uncoated or plasma-
Implants? coated surfaces.53
In order to investigate the mechanisms of biomate- However, further experimental evidence indicates that
rial-mediated inflammatory responses, we developed a adsorbed immunoglobulins are not essential to acute inflam-
simple animal implantation model. Biomaterial films are cut matory responses to implants. We placed implants in mice
into 1.2 cm diameter disks. After repetitive washes with etha- with severe combined immunodeficiency (SCID) which have
nol, the disks are precoated with various types of proteins extremely low plasma IgG levels (< 1 ∝g/ml vs. 4 mg/ml in
and then implanted in the mouse peritoneal cavity for normal, otherwise syngeneic, animals). In SCID mice, the
16-18 h (in this model, the time of maximal phagocyte ac- inflammatory response to uncoated PET disks as evidenced
cumulation).53 The surface associated activities of myelo- by the recruitment of both neutrophils (Fig. 20.1) and
peroxidase (MPO) and nonspecific esterase (NSE) are mea- macrophages (not shown) is almost as great as in immuno-
sured to estimate the numbers of adherent neutrophils and competent animals. The modest decrement in neutrophils
macrophages/monocytes, respectively.53,54 Although a num- and macrophage recruitment in SCID vs. normal mice prob-
ber of other investigators have used subcutaneous implants ably reflects a general defect in phagocyte mobilization, be-
or cage implants (e.g., refs. 2,3,19,20), these experimental cause this difference is apparent on both uncoated and
systems have some shortcomings compared to intraperito- IgG-coated surfaces.
neal implants.7 The peritoneal cavity provides a good site
for studying inflammatory responses because reactions elic- Complement Activation Is Not Required for Phagocyte
ited by the polymer are clearly defined with minimal par- Accumulation on Biomaterial Implants
ticipation of the coagulation system.7,53-57 Furthermore, it A few blood-contact biomaterials may trigger comple-
appears that at least the majority of phagocytic cells found ment activation and (during, e.g., hemodialysis or
adherent to explants are, indeed, recruited de novo and by leukapheresis) transient neutropenia, suggesting that
the implant itself. Thus, mice subjected to sham surgery show complement activation might contribute to the inflamma-
only a small increment in numbers of phagocytes in perito- tory effects of blood-contact biomaterials (e.g., see refs. 32,
neal lavage fluids compared to implant-bearing animals. 63-67). This hypothesis is well supported by the known ef-
As indicated above, albumin, IgG, fibrinogen and (to fects of activated complement components. For example,
a lesser extent) complement are the most abundant proteins surface-induced complement activation will generate C3a
in plasma and are found to predominate on many blood- and C5a fragments,68,69 which are known to mediate granu-
contact and tissue-contact biomaterials, including polyeth- locyte aggregation,67 chemotaxis,70 and adhesiveness to en-
ylene terephthalate, expanded polytetrafluoroethylene, poly- dothelial cells.71 In addition, complement activation by some
dimethylsiloxane, polyurethanes and polyethylene.29,36,37,53,54 polymeric materials has been shown in vitro to cause
Especially on hydrophobic surfaces, these adsorbed proteins phagocyte adhesion.65,72 By extrapolation, one might antici-
tend to assume an altered conformation and become tightly pate that similar events might be involved in acute inflam-
adherent.38-41,53,54 Taking advantage of this property, we were matory responses to tissue contact biomaterials. However,
able to determine the possible role of various plasma pro- complement depleted mice (pretreated with cobra venom
teins—precoated on implant surfaces—in initiating bioma- factor) exhibit normal recruitment of inflammatory cells to
terial-mediated inflammatory responses. In agreement with implant surfaces.53 Consequently, it appears that although
many previous reports, we find that albumin, the most abun- activated complement components may powerfully
Molecular Determinants of Acute Inflammatory Responses to Biomaterials 209
stimulate phagocytes, complement activation is not impor- both neutrophils and macrophages. In order to exclude the
tant in acute inflammatory responses to biomaterial implants possible involvement of activated coagulation factors, simi-
(or, at least to implants of hydrophobic polymers such as lar studies were done using afibrinogenemic plasma in which
polyethylene terephthalate, polyninyl chloride or fibrinogen was undetectable. PET coated with afibrino-
polyether urethane). genemic plasma attracted very few phagocytes (roughly equal
to the numbers which accumulate on albumin-coated ma-
Fibrin(ogen) Is Necessary for Phagocyte terial). However, when physiologic levels (3 mg/ml) of pu-
Accumulation on Biomaterial Implants rified fibrinogen were restored to this afibrinogenemic
The foregoing results indicate that, insofar as our ex- plasma, PET disks incubated in this mixture prompted nor-
perimental murine model is analogous to the human cir- mal recruitment of phagocytes.54
cumstance, neither complement activation nor adsorbed IgG Most importantly, the requirement for fibrinogen ad-
is a necessary mediator of acute inflammatory reactions to sorption to the surface of PET implants would appear to
implanted materials such as Dacron (PET). Together, albu- hold in vivo as well. Mice having undetectable plasma fi-
min, IgG and fibrinogen may comprise > 80% of adsorbed brinogen were produced by repetitive injections of
surface proteins, at least on certain polymers.29,36,73,74 Con- ancrod.75-80 Ancrod infusion causes severe hypofibrino-
sequently, it seemed that fibrinogen—also present in large genemia and hypoplasminogenemia, but does not disturb
amounts on implant surfaces36—might be the host protein the number or turnover of platelets or the levels of other
most important in triggering inflammatory reactions. coagulation factors and plasma proteins (including
This hypothesis was supported by a surprising obser- fibronectin).76,78,81,82 Ancrod-treated mice (having undetect-
vation: Serum coated surfaces, like albumin coated surfaces, able fibrinogen at the time of implantation) show almost
fail to prompt inflammatory responses.54 The major differ- no neutrophil accumulation on untreated disks, but do so if
ence between plasma and serum is that plasma, but not se- the disks are precoated with purified murine fibrinogen
rum, contains fibrinogen. We therefore reconstituted serum (Fig. 20.2). 54 The same dependence on adsorbed
by adding a physiological concentration of fibrinogen and fibrin(ogen) was also observed in similar experiments em-
used this to coat experimental implants. Disks precoated with ploying subcutaneous, rather than intraperitoneal, implants.
fibrinogen-repleted serum attract as many phagocytes as do Finally, we should note that although some studies have
disks coated with fresh, minimally heparinized plasma. In shown that fibronectin can serve as a bridge for phagocyte-
addition, fibrinogen alone is sufficient to trigger inflamma- surface fibrin(ogen) interactions,83,84 our preliminary ex-
tory responses; PET disks preincubated with purified hu- periments have revealed no difference in in vitro phagocyte
man (or murine) fibrinogen also attract large numbers of adherence to materials coated with normal plasma vs.
210 Tissue Engineering of Prosthetic Vascular Grafts
fibronectin-depleted plasma (Tang et al, unpublished ob- epitope(s) responsible for phagocyte accumulation, plasmin
servations). This may, however, reflect the absence of expo- degradation fragments of fibrinogen were generated and
sure of these phagocytes to factors which, e.g., upregulate used to coat implant surfaces (Fig. 20.3). Implants precoated
cell adherence molecules (vide infra). with the plasmin degradation fragment D100
(MW = 105 kDa), but not E50 (MW = 50 kDa), accumulate
Which Portion of Fibrinogen Is Critical large numbers of phagocytes (both neutrophils and mac-
in Mediating Phagocyte Adhesion? rophages/monocytes), as many as fibrinogen-coated disks.
The foregoing results provide strong support for the To determine more precisely the specific portion of fibrino-
proposition that the spontaneous adsorption of host fibrino- gen responsible, D100 was digested to D80 (MW = 80 kDa)
gen to the surfaces of implanted PET is a necessary prece- and further to D30 (MW = 30 kDa). Both D80 and D30 are
dent to the subsequent inflammatory and fibrotic processes. fully active in fostering the in vivo accumulation of both
We reasoned that the adsorbed fibrinogen probably displayed neutrophils and macrophages/monocytes.85 This suggests
one or more epitopes normally occult in the soluble protein that at least one necessary motif for phagocyte accumula-
but exposed by surface-mediated “denaturation” of the pro- tion is within the fibrinogen D30 fragment.
tein. In order to determine the location of the hypothetical
Altieri and colleagues87 had earlier found that one seg- How Does Adsorbed Fibrinogen Become
ment of D30, &190-202 (hereafter abbreviated as ‘P1’), is criti- Pro-Inflammatory?
cally important in mediating phagocyte-fibrinogen interac- The preceding results indicate that fibrinogen
tions. Thus, we thought that this particular short sequence &190-202 is the most important factor in mediating the ac-
might be a crucial determinant of phagocyte interactions cumulation of phagocytes on fibrinogen-bearing implant
with adsorbed fibrinogen on implant surfaces. To test this, surfaces. However, it remains unclear how the simple ad-
P1 and a P1-based scrambled peptide were synthesized and sorption of fibrinogen to implant surfaces might modify the
covalently linked to human albumin (which has been used protein, converting it into a pro-inflammatory signal. It is
widely as a carrier to enhance cellular responses to pep- very likely that the pro-inflammatory nature of adsorbed
tides88-90). Indeed, implant surfaces precoated with P1 pep- fibrinogen reflects the occurrence of a conformational
tide, but not with the scrambled peptide, induce substantial change, probably also reflected by the progressive resistance
phagocyte accumulation, roughly equivalent to that caused of adsorbed fibrinogen to elution by detergents.38 For
by fibrinogen-coated surfaces (Fig. 20.4).
example, after PET film has been incubated with purified also occur naturally in the normal course of hemostasis and
fibrinogen for 4 h, more than 60% of adsorbed protein is wound resolution. The presence of fibrin has long been rec-
resistant to elution by sodium dodecyl sulfate.53 The con- ognized as coeval with inflammatory and fibrotic responses,
formational changes of adsorbed fibrinogen also have been and fibrin deposition is typical of both acute and chronic
recently probed by differential scanning calorimetry inflammatory processes.92-95 Furthermore, patients who are
(DSC),46 based on the assumption that denaturation of hypo- or afibrinogenemic are known to have abnormal in-
adsorbed protein will prevent some or all further thermal flammatory and wound healing responses. Therefore, it may
transitions. The DSC thermograms of native bovine fibrino- be that the body interprets the presence of fibrin as an indi-
gen have two transition peaks at 55°C and 96°C, reflecting cation that vascular barriers have been breached and that
denaturation of the D and E domains of fibrinogen, respec- invasion by microorganisms might have occurred. In this
tively. After prolonged interaction with low temperature iso- light, the attraction of phagocytic cells—important in both
tropic carbon (which has a highly hydrophobic surface), the host defense and in the ultimate dissolution of the coagu-
transition peaks for both domains are diminished, but the lum—makes perfect biological sense.
peak for the fibrinogen D domain is reduced more than that
of the E domain. This suggests that the D domain may be The Mechanism of Biomaterial-Mediated
more susceptible to surface induced denaturation than the Inflammatory Responses
E domain. Because the P1 epitope is within the D domain Although the above may represent some small progress
and the D domain is very susceptible to conformational in understanding the importance of surface protein adsorp-
changes, we now believe that the interaction between fibrino- tion in the response to biomaterial implants, there remain
gen and surfaces mediates the conformational changes of many mysteries concerning the mechanisms involved in the
the D fragment causing the exposure of P1. As a result, oth- overall inflammatory response. The evolution of biomate-
erwise inert and unreactive surfaces displaying this epitope rial-mediated inflammatory responses may be somewhat
are recognized by phagocytes. The exposure of normally arbitrarily divided into three consecutive events (Fig. 20.5):
hidden epitopes as a result of adsorption to polymeric sur- 1. Phagocyte transmigration through the endothelial
faces has some precedent; upon adsorption to plastic tissue barrier;
culture surfaces (which, having been plasma discharge 2. Chemotaxis toward the implants; and, finally,
treated, are not precisely analogous to most hydrophobic 3. Adherence to the biomaterial.
implant surfaces), fibrinogen changes conformation and A fuller understanding of these processes—discussed
exposes multiple receptor-induced binding sites (RIBS) below in reverse order of occurrence—may help in the de-
&112-119 and A#95-98.91 velopment of strategies to moderate biomaterial-mediated
Thus, we now believe that surface fibrin(ogen) un- inflammatory responses and associated tissue responses.
dergoes changes in conformation, becoming fibrin-like and
that this anomalous fibrin(ogen) is critical in the attraction Phagocyte Adherence to Biomaterial Implants
and adherence of phagocytic cells to implant surfaces. The In order to interact with implant surfaces, receptors
ensuing responses of phagocytic cells to this product may on cell surfaces must interact with adsorbed fibrinogen. We
presently think that the phagocyte integrin Mac-1 (Cd11b/ intraperitoneal surface phagocytic cells are obviously pref-
cd18) is required for this interaction. This is partly because erentially accumulated on the implant. We have assumed
earlier investigators had shown that the critical fibrin(ogen) that certain chemokines might participate in attracting the
epitope, P1, is recognized by Mac-1.87 As a partial test of incoming phagocytic cells toward implant surfaces. A num-
this hypothesis, we employed recombinant neutrophil in- ber of chemokines have been reported to foster the recruit-
hibitory factor (hereafter, ‘NIF’), which blocks Cd11b-de- ment of inflammatory cells in a variety of inflammatory dis-
pendent binding of Mac-1 to fibrinogen (which presum- eases.102-105 In searching for possible candidates among many
ably occurs through the P1 epitope) without affecting other potential chemokines we employed reverse transcript poly-
Mac-1 integrin functions such as binding to coagulation fac- merase chain reaction to estimate levels of chemokine mRNA
tor X.96,97 Indeed, when mice were given repetitive injec- produced by implant adherent cells. We observed high lev-
tions of NIF before and during implantation, phagocytes els of mRNA production for two particular chemokines:
failed to accumulate on fibrinogen-coated surfaces. Further macrophage inflammatory protein-1# (MIP-1#) and mono-
support for the importance of Mac-1 in these phagocyte- cyte chemoattractant protein-1 (MCP-1), both of which
implant interactions derives from recent experiments in appear to be involved in phagocyte-implant interactions.
animals lacking this integrin. In Cd11b knockout (KO) and Neutralizing antibodies to both MIP-1# and MCP-1,
CD18 KO mice, the ‘normal’ accumulation of phagocytes but not to MIP-1! and MIP-2, block 70-90% of the accu-
on implant surfaces fails to occur. Interestingly, this impair- mulation of both macrophages and neutrophils on implant
ment of phagocyte accumulation is not caused by decreased surfaces. However, the numbers of recruited phagocytes do
phagocyte recruitment; the numbers of macrophages and not differ amongst control and treated groups.106 Therefore,
neutrophils in peritoneal lavage fluids following implanta- it is likely that the reduction in numbers of adherent phago-
tion in both types of KO animals are roughly equivalent to cytes is caused by impairment of directed phagocyte chemo-
those in wild type controls.98 taxis (although other interpretations are possible at the
Thus, the Mac-1 integrin appears to be crucial in the moment). Indeed, these results are in accord with prior stud-
adhesion of inflammatory cells to implant surfaces. How- ies indicating that MIP-1# and MCP-1 contribute to inflam-
ever, unstimulated phagocytes normally express only low matory and immune tissue responses via promotion of
levels of surface Mac-1 and most of this is in the ‘low-affin- chemotaxis and activation of both phagocytes and T lym-
ity’ state. This raises the question of what factors might phocytes.107-112 Because many different types of cells are able
prompt the apparent upregulation of this phagocyte integrin. to generate either MIP-1# and MCP-1,109,113,114 it is still not
Earlier studies indicated that TNF-# is a potent stimulator clear which cell type is responsible for the production of
of the expression of Mac-1 on cell surfaces.99-101 Further- MIP-1# and MCP-1 in the course of biomaterial-mediated
more, by ELISA assays, we have found that TNF-# is present inflammatory responses.
in peritoneal lavage fluids from implant-bearing mice within Thus, in interim summary, we are gaining some idea
four hours after implantation (unpublished results). This of the mechanisms involved in implant surface-protein
suggested the possibility that TNF-# might be critical in interactions and in phagocyte chemotaxis towards, and
upregulating Mac-1 integrin expression on incoming adherence to, tissue contact biomaterials. However, the
phagocytes. In at least partial support of this possibility, we processes involved in the critical initial step—phagocyte
find that administration of neutralizing anti-TNF-# anti- transmigration through the endothelial barrier—
bodies to implant-bearing mice blocks phagocyte accumu- remain undefined.
lation on implant surfaces. Furthermore, as was the case with
the Mac-1 KO mice described above, we find that neutraliz- Phagocyte Transmigration Through the Endothelial
ing antibodies against TNF-# do not affect the extent of Barrier
phagocyte recruitment to the site of the implant because the For a number of reasons, we hypothesized that the
numbers of recruited cells are roughly equivalent in treated release of histamine might be important for the recruitment
and control animals.98 of inflammatory cells to implantation sites. The most direct
Therefore, the adhesion of inflammatory cells to im- evidence favoring this idea was the observation made by us
plant surfaces appears to depend on three separable events: and others115 that the tissue surrounding biomaterial im-
1. Precedent adherence of fibrin(ogen) accompanied plants is often hyperemic and edematous, features of a clas-
by exposure of the P1 epitope; sical histaminic response. In order to test the possible in-
2. The release of TNF-# in response to implantation; volvement of histamine, we administered the histamine re-
and ceptor antagonists pyrilamine (an H1 receptor antagonist)
3. Consequent TNF-#-mediated upregulation of Mac-1 and famotidine (an H2 receptor antagonist) to animals re-
on incoming phagocytes. ceiving implants. When given separately, neither receptor
antagonist significantly diminishes the accumulation of in-
Phagocyte Chemotaxis Toward Implant Surfaces flammatory cells on implant surfaces. However, combined
The accumulation of phagocytes on implant surfaces treatment of implant recipients with both H1 and H2 re-
is clearly not a random event. In the peritoneum of an ani- ceptor antagonists significantly reduces the numbers of
mal implanted ~16 h previously, ~20% of total neutrophils phagocytes on implant surfaces and the numbers of mac-
and ~50% of total macrophages are found on the implant rophages and neutrophils recruited de novo to the perito-
surface (as opposed to being recovered in peritoneal lavage). neal cavity (down to values of 50% and 40% of untreated
Inasmuch as the implant represents only ~2% of the total
214 Tissue Engineering of Prosthetic Vascular Grafts
controls, respectively).116 We tentatively conclude that his- 1. Implants (and blood-contact devices) made of hy-
tamine enhances the transmigration of phagocytes through drophobic polymers almost instantly acquire a layer
the endothelial barrier via both H1 and H2 receptors. of spontaneously adsorbed host protein;
Mast cells are the most likely source of the histamine 2. The adsorbed protein gradually ‘denatures’;
which may be involved (although macrophages also are re- 3. The presence of the implant is sensed by the host
ported to produce and release histamine).117-119 Indeed, the either through mechanical properties of the mate-
peritoneal space contains relatively large numbers of mast rial itself or through the presence of these ‘denatured’
cells (2-5% of total lavage cells)8,120 and mast cells have the surface proteins;
largest amounts of stored histamine, which can be released 4. Guardian cells—especially mast cells—are activated,
in a short period of time. The importance of mast cells in releasing histamine and, perhaps, other pro-inflam-
the pathogenesis of other types of inflammatory responses matory products;
has been investigated by others using mast cell deficient mice 5. The resultant histaminic response promotes the dia-
(WBB6F1-Wv/W).121-123 In part, the results indicate that pedesis of phagocytic cells across the endothelial
mast cell degranulation is important in neutrophil recruit- barrier;
ment,121 macrophage activation,124 IL-1 production by 6. Acting on the instructions of particular cytokines (es-
monocytes and macrophages,125 fibroblast proliferation and pecially MIP-1#, MCP-1 and TNF-#) the incoming
collagen accumulation,126 fibrin deposition,123 and, not sur- phagocytes migrate towards the implant whilst si-
prisingly, the Arthus reaction.123 In fact, several years ago, multaneously upregulating surface adherence mol-
Christenson and coworkers8 made the prescient suggestion ecules such as Mac-1;
that mast cells might play a key role in the inflammatory 7. Upon encountering the surface of the implant, the
responses to intraperitoneal implants. phagocytic cells bind to the proteinaceous coat of
We therefore tested the possible importance of mast the implant, a binding facilitated by interactions be-
cells in phagocyte recruitment using mast cell deficient tween Mac-1 and a small 13 amino acid epitope dis-
mice.116 After intraperitoneal implantation for 16 h, far fewer played by adsorbed fibrinogen.
PMN (< 50% of normal) and macrophages/monocytes And then? Clearly, a great deal more work will be re-
(< 30% of normal) accumulated on implant surfaces in mast quired to unravel the intricacies involved in the above steps
cell deficient mice than in their congenic controls. Further- and, as importantly, the nature of subsequent responses
more, as was the case with normal animals treated with com- which may eventuate in chronic inflammation and fibrosis.
bined histamine receptor antagonists, the reduction of the Nonetheless, this knowledge, once won, should eventually
numbers of adherent phagocytes is accompanied by greatly enable the purposeful engineering of materials with vastly
decreased recruitment of phagocytes to the peritoneum. improved biocompatibility.
Therefore, the results obtained with two different models in
which histamine-dependent signaling events are disrupted— References
histamine receptor antagonist administration and mast cell 1. Kottke-Marchant K, Anderson JM, Umemura Y, Marchant
deficient animals—are in complete agreement. RE. Effect of albumin coating on the in vitro blood com-
However, once again mysteries remain. It is anything patibility of Dacron arterial prostheses. Biomaterials 1989;
but clear how implant surfaces may cause the activation of 10:147-155.
2. Marchant RE, Phua K, Hiltner A, Anderson JM. In vivo
mast cells and, indeed, whether other released products of
biocompatibility studies: II. Biomer: Preliminary cell ad-
mast cells (such as TNF-#) might be important in the over- hesion and surface characterization studies. J Biomed
all inflammatory response. Nonetheless, we can tentatively Mater Res 1984; 18:309-315.
conclude that biomaterial implants do trigger the activation 3. Marchant RE, Miller KM, Anderson, JM. In vivo
of mast cells, which release histamine. The subsequent his- biocompatibility studies: V. In vivo leukocyte interactions
taminic responses—obvious as localized hyperemia and with biomer. J Biomed Mater Res 1984; 18:1169-1190.
edema—facilitate the arrest and diapedesis of phagocytic 4. Marchant RE, Anderson JM, Dillingham EO. In vivo
cells at sites of biomaterial implantation. biocompatibility studies: VII. Inflammatory responses to
polyethylene and to a cytotoxic polyvinylchloride. J
Conclusions Biomed Mater Res 1986; 20:37-50.
Although typically inert, nontoxic and non- 5. Tang L, Eaton JW. Mechanism of acute inflammatory re-
sponse to biomaterials. Cells Mater 1994; 4:429-436.
immunogenic, tissue-contact biomaterials often prompt a
6. Desai NP, Hubbell JA. Tissue response to intraperitoneal
variety of unwanted host responses. Our present ignorance implants of polyethylene oxide-modified polyethylene
of the mechanisms involved in these implant-mediated host terephthalate. Biomaterials 1992; 13:505-510.
responses hinders the rational development of more 7. Christenson L, Aebischer P, McMillan P, Galletti PM.
biocompatible materials. Despite the efforts of a large Tissue reaction to intraperitoneal polymer implants: Spe-
number of excellent investigators, there remain many more cies difference and effects of corticoids and doxorubicin.
questions than answers. J Biomed Mater Res 1989; 23:705-718.
What little we do know can be very briefly 8. Christenson L, Wahlberg L, Aebischer P. Mast cells and
summarized: tissue reaction to intraperitoneally implanted polymer
capsules. J Biomed Mater Res 1991; 25:1119-1131.
Molecular Determinants of Acute Inflammatory Responses to Biomaterials 215
9. Stark GB, Gobel M, Jaeger K. Intraluminal cyclosporine tificial surfaces and proteins. Ann NY Acad Sci 1987;
A reduces capsular thickness around silicon implants in 516:173-183.
rats. Ann Plast Surg 1990; 24:156-161. 29. Anderson JM, Bonfield TL, Ziats NP. Protein adsorption
10. Behling CA, Spector M. Quantitative characterization of and cellular adhesion and activation on biomedical poly-
cells at the interface of long-term implants of selected mers. Int J Artif Org 1990; 13:375-382.
polymers. J Biomed Mater Res 1986; 20:653-666. 30. Bohnert JL, Horbett TA. Changes in adsorbed fibrinogen
11. Gordon M, Bullough PG. Synovial and osseous inflam- and albumin interactions with polymers indicated by de-
mation in failed silicone rubber prostheses. A report of creases in detergent elutability. J Colloid Interface Sci
six cases. J Bone Joint Surg 1982; 64:574-580. 1986; 111:363-377.
12. Kossovsky N, Millett D, Juma S et al. In vivo character- 31. Pitt WG, Park K, Cooper SL. Sequential protein adsorp-
ization of the inflammatory properties of tion and thrombus deposition on polymeric biomaterials.
poly(tetrafluoroethylene) particulates. J Biomed Mater Res J Colloid Interface Sci 1986; 111:343-362.
1991; 25:1287-1301. 32. Kuwahara T, Markert M, Wauters JP. Protein adsorption
13. Nelson GD. Complications from the treatment of fibrous on dialyzer membranes influences their biocompatibility
capsular contracture of the breast. Plast Reconstr Surg properties. Contrib Nephrol (Basel, Karger) 1989;
1981; 68:969-980. 74:52-57.
14. Smahel J. Foreign material in the capsules around breast 33. Sevastianov VI. Role of protein adsorption in blood
prostheses and the cellular reaction to it. Br J Plast Surg biocompatibility of polymers. CRC Crit Rev Biocompat
1979; 32:35-42. 1988; 4:109-154.
15. Vistness LM, Ksander GA, Kosek J. Study of encapsula- 34. Horbett TA. Adsorption of proteins from plasma to a se-
tion of silicon rubber implants in animals. A foreign re- ries of hydrophilic-hydrophobic copolymers. II. Compo-
action. Plast Reconstr Surg 1978; 62:580-588. sitional analysis with the prelabeled protein technique. J
16. Domanskis EJ, Owsley JQ Jr. Histological investigation of Biomed Mater Res 1981; 15:673-695.
the etiology of capsule contracture following augmenta- 35. Shinoda BA, Mason RG. Reaction of blood with artificial
tion mammaplasty. Plast Reconstr Surg 1976; 58:689-693. surfaces of hemodialyzers. Studies of human blood with
17. Yates SG II, Nakagawa Y, Berger K, Sauvage LR. Surface platelet defects or coagulation factor deficiencies. Biomat
thrombogenicity of arterial prostheses. Surg Gynec Obstet Med Dev Art Org 1978; 6:305-329.
1973; 136:12-16. 36. Pankowsky DA, Ziats NP, Topham NS et al. Morphologic
18. Berger K, Sauvage LR, Rao AM, Wood SJ. Healing of ar- characteristics of adsorbed human plasma proteins on
terial prostheses in man: Its incompleteness. Ann Surg vascular grafts and biomaterials. J Vasc Surg 1990;
1972; 175:118-127. 11:599-606.
19. Zhao Q, Agger MP, Fitzpatrick M et al. Cellular interac- 37. Andrade J, Hlady V. Plasma protein adsorption: The big
tions with biomaterials: in vivo cracking of prestressed twelve. Ann NY Acad Sci 1987; 516:158-172.
pellethane 2363-80A. J Biomed Mater Res 1990; 38. Lenk TJ, Horbett TA, Ratner BD, Chittur KK. Infrared
24:621-637. spectroscopic studies of time-dependent changes in fi-
20. Zhao Q, Topham N, Anderson JM et al. Foreign-body brinogen adsorbed to polyurethanes. Langmuir 1991;
giant cells and polyurethane biostability: In vivo correla- 7:1755-1764.
tion of cell adhesion and surface cracking. J Biomed Mater 39. Lu DR, Park K. Effect of surface hydrophobicity on the
Res 1991; 25:177-183. conformational changes of adsorbed fibrinogen. J Colloid
21. Sutherland K, Mahoney JR II, Coury AJ, Eaton JW. Deg- Interfac Sci 1991; 144:271-281.
radation of biomaterials by phagocyte-derived oxidants. J 40. Slack SM, Horbett TA. Changes in the strength of fibrino-
Clin Invest 1993; 92:2360-2367. gen attachment to solid surfaces: An explanation of the
22. Chan SC, Birdsell DC, Gradeen CY. Detection of influence of surface chemistry on the Vroman effect. J
toluenediamines in the urine of a patient with polyure- Colloid Interfac Sci. 1989; 133:148-165.
thane-covered breast implants. Clin Chem 1991; 41. Vroman L, Adams AL, Klings M et al. Reactions of formed
37:756-758. elements of blood with plasma proteins at interfaces. Ann
23. Picha GJ, Goldstein JA, Stohr E. Natural-Y Meme poly- NY Acad Sci 1977; 283:65-75.
urethane versus smooth silicone: Analysis of the soft-tis- 42. Chinn JA, Posso SE, Horbett TA, Ratner BD.
sue interaction from 3 days to 1 year in the rat animal Postadsorption transition in fibrinogen adsorbed to poly-
model. Plast Reconstr Surg 1990; 85:903-916. urethanes: Changes in antibody binding and sodium
24. Trumpy I, Lyberg T. In vivo deterioration of proplast- dodecyl sulfate elutability. J Biomed Mater Res 1992;
teflon temporomandibular joint interpositional implants: 16:757-778.
A scanning electron microscopic and energy-dispersive X- 43. Rapoza RJ, Horbett TA. Changes in the SDS elutability
ray analysis. J Oral Maxillofac Surg 1993; 51:624-629. of fibrinogen adsorbed from plasma to polymers. J
25. Henry CH, Wolford LM. Treatment outcomes for tem- Biomater Sci Polym Ed 1989; 1:99-110.
poromandibular joint reconstruction after Proplast-Teflon 44. Schaaf P, Dejardin P. Structural changes within an
implant failure. J Oral Maxillofac Surg 1993; 51:352-358. adsorbed fibrinogen layer during the adsorption process:
26. Kozinn SC, Johanson NA, Bullough PG. The biologic in- A study by scanning angle reflectometry. Colloids Surfaces
terface between bone and cementless femoral 1988; 31:89-100.
endoprostheses. J Arthroplasty 1986; 1:249-259. 45. Wigren R, Elwing H, Erlandsson R et al. Structure of
27. Lindsay R. Blood surface interactions. Trans Am Soc Artif adsorbed fibrinogen obtained by scanning force micros-
Internal Org 1980; 26:603-610. copy. FEBS Letts 1991; 280:225-258.
28. Leininger RI, Hutson TB, Jakobsen RJ. Spectroscopic ap- 46. Feng L, Andrade JD. Protein adsorption on low-tempera-
proaches to the investigation of interactions between ar- ture isotropic carbon: I. Protein conformation change
216 Tissue Engineering of Prosthetic Vascular Grafts
probed by differential scanning calorimetry. J Biomed 65. Herzlinger GA, Cumming RD. Role of complement acti-
Mater Res 1994; 28:735-743. vation in cell adhesion to polymer blood contact surface.
47. MacRichie F. The adsorption of proteins at the solid/liq- Trans Am Soc Artif Intern Org 1980; 26:165-171.
uid interface. J Colloid Interfac Sci 1972; 38:484-490. 66. Nusbacher J, Rosefeld SI, MacPherson JL et al. Nylon fi-
48. Lee R. Adsorption of proteins onto hydrophobic polymer ber leukapheresis: Associated complement component
surfaces: Adsorption isotherms and kinetics. J Biomed changes and granulocytopenia. Blood 1978; 51:359-365.
Mater Res 1974; 8:251-259. 67. Craddock PR, Fehr J, Dalmasso AP et al. Hemodialysis
49. Andrade JD, Hlady VL, Van Wagenen RA. Effects of leukopenia. Pulmonary vascular leukostasis resulting from
plasma protein adsorption on protein conformation and complement activation by dialyzer cellophane membrane.
activity. Pure Appl Chem 1984; 56:1345-1350. J Clin Invest 1977; 59:879-888.
50. Absolom DR, Zingg W, Policova Z, Neumann AW. De- 68. Janatova J. C3, C5 and C3a, C4a, and C5a fragments of
termination of the surface tension of protein coated ma- the complement system. Meth Enzymol 1988;
terials by means of the advancing solidification front tech- 162:579-625.
nique. Trans Am Soc Intern Artif Organs 1983; 69. Janatova J, Cheung AK, Parker CJ. Biomedical polymers
29:146-151. differ in their capacity to activate complement. Comple-
51. Morrissey BW, Han CC. The conformation of &-globulin ment Inflamm 1991; 8:61-69.
adsorbed on polystyrene lattices determined by quasielastic 70. Dahlgren C, Hed J, Stendahl O. Chemotaxis of polymor-
light scattering. J Colloid Interfac Sci 1978; 65:423-431. phonuclear leukocytes in response to surface-bound
52. Morrissey BW, Fenstermaker CA. Conformation of complement-derived chemoattractants generated in situ.
adsorbed &-globulin and !-lactoglobulin. Effect of surface Inflammation 1984; 8:201-208.
concentration. Trans Am Soc Artif Intern Org 1976; 71. O’Flaherty JT, Rossi AG, Redman JR, Jacobson DP. Tu-
22:278-284. mor necrosis factor regulates expression of receptors for
53. Tang L, Lucas AH, Eaton JW. Inflammatory responses to formyl-methionyl-leucyl-phenylalanine, leukotriene B4
Dacron: Role of surface adsorbed IgG. J Lab Clin Med and platlet activating factor. Dissociation from priming
1993; 122:292-300. in human polymorphonuclear neutrophils. J Immunol
54. Tang L, Eaton JW. Fibrin(ogen) mediates acute inflam- 1991; 147:3842-3847.
matory responses to biomaterials. J Exp Med 1993; 72. Cumming RD. Important factors affecting initial blood-
178:2147-2156. material interactions. Trans Am Soc Artif Intern Org 1980;
55. Freyria AM, Chignier E, Guidollet J, Louisot P. Perito- 26:304-308.
neal macrophage response: An in vivo model for the study 73. Miale JB. Laboratory Medicine Hematology. Sixth edn. C.
of synthetic materials. Biomaterials 1991; 12:111-118. V. Mosby Co., p. 1084, 1982.
56. Merhi Y, Roy R, Guidoin R et al. Cellular reactions to 74. Dittmer DS (edited), Altman PL (analysis and compila-
polyester arterial prosthesis impregnated with cross-linked tion). Blood and other body fluids. Biology Handbook.
albumin: in vivo studies in mice. Biomaterials 1989; Federation of American Societies for Experimental Biol-
10:56-58. ogy, Washington, DC, p. 540, 1961.
57. Uenoyama K, Kanagawa R, Tamura M et al. Experimen- 75. McRitchie DI, Girotti MJ, Glynn MFX et al. Effect of sys-
tal intraocular lens implantation in the rabbit eye and in temic fibrinogen depletion on intraabdominal abscess for-
the mouse peritoneal space. Part 1. Cellular components mation. J Lab Clin Med 1991; 118:48-55.
observed on the implanted lens surface. J Cat Refract Surg 76. Cole EH, Glynn FX, Laskin CA et al. Ancrod improves
1988; 14:187-191. survival in murine systemic lupus erythematosus. Kidney
58. Guidoin R, Snyder R, Martin L et al. Albumin coating of Internat 1990; 37:29-35.
a knitted polyester arterial prosthesis: An alternative to 77. Pollak VE, Glueck HI, Weiss MA et al. Defibrination with
preclotting. Ann Thorac Surg 1984; 37:457-465. ancrod in glomerulonephritis: Effects on clinical and his-
59. Stossel TP. Medical progress—phagocytosis (first of three tologic findings and on blood coagulation. Am J Nephr
parts). New Engl J Med 1974; 290:717-723. 1982; 2:195-207.
60. Henson PM, Oades ZG. Stimulation of human neutro- 78. Bell WR, Shapiro SS, Martinez J, Nossel HL. The effects
phils by soluble and insoluble immunoglobulin aggregates. of Ancrod, the coagulation enzyme from the venom of
Secretion of granule constituents and increased oxidation Malayan pit viper (A. rhodostoma) on prothrombin and
of glucose. J Clin Invest 1975; 56:1053-1061. fibrinogen metabolism and fibrinopeptide A release in
61. Henson PM, Johnson HB, Spiegelberg HL. The release of man. J Lab Clin Med 1978; 91:592-604.
granule enzymes from human neutrophils stimulated by 79. Silberman S, Bernik MB, Potter EV, Kwuan HC. Effects
aggregated immunoglobulins of different classes and sub- of ancrod (Arvin) in mice: Studies of plasma fibrinogen
classes. J Immunol 1972; 109:1182-1192. and fibrinolytic activity. Br J Hæmat 1973; 24:101-113.
62. Chuang HYK, Mohammad SF, Mason RG. Prostacyclin 80. Reid HA, Chen KE, Thean PC. Prolonged coagulation
(PGI2) inhibits the enhancement of granulocyte adhesion defect (defibrination syndrome) in malayan viper bite.
to cuprophane induced by immunoglobulin G. Thromb Lancet 1963; i:621-626.
Res 1980; 19:1-9. 81. Barcelli U, Rademacher PR, Ooi BS, Pollak VE. Defibrina-
63. Cappelli G, Lucchi L, Bonucchi D et al. Polymorpho- tion with Ancrod: Effect of reticuloendothelial clearance
nuclear oxygen free radical production and complement of circulating immune complexes. Nephron 1982;
activation induced by dialysis membranes as assayed in 30:314-317.
an experimental model. Blood Purif 1989; 7:293-300. 82. Esnouf MP, Tunnah GW. The isolation and properties of
64. Shepard AD, Gelfand JA, Gallow AD, O’Donnell TF Jr. the thrombin-like activity from Ancistrodon rhodostoma
Complement activation by synthetic vascular prostheses. venom. Br J Hæmat 1967; 13:581-590.
J Vasc Surg 1984; 1:829-838. 83. Blystone S, Weston L, Kaplan JE. Fibronectin dependent
macrophage fibrin binding. Blood 1991; 78:2900-2907.
Molecular Determinants of Acute Inflammatory Responses to Biomaterials 217
84. Kaplan JE, Cardarelli PM, Rourke FJ et al. Fibronectin 102. Oppenheim JJ, Zachariae CO, Mukaida N, Matsushima
augments binding of fibrin to macrophages. J Lab Clin K. Properties of the novel proinflammatory supergene
Med 1989; 113:168-176. “intercrine” cytokine family. Ann Rev Immunol 1991;
85. Tang L, Ugarova TP, Plow EF, Eaton JW. Molecular de- 9:617-648.
terminants of acute inflammatory responses to 103. Harrison LC, Campbell IL. Cytokines: An expanding net-
biomaterials. J Clin Invest 1996; 97:1329-1334. work of immuno-inflammatory hormones. Molec
86. Mossesson MW. Fibrin polymerization and its regulatory Endocrin 1988; 2:1151-1156.
role in hemostasis. J Lab Clin Med 1990; 116:8-17. 104. Ward PA. Cytokines, inflammation, and autoimmune dis-
87. Altieri DC, Plescia J, Plow EF. The structural motif gly- eases. Hosp Practice 1995; 30:35-41.
cine190-valine202 of the fibrinogen & chain interacts with 105. Arai K-I, Lee F, Miyajima A et al. Cytokines: Coordina-
Cd11b/cd18 integrin (#M!2, Mac-1) and promotes leu- tors of immune and inflammatory responses. Ann Rev
kocyte adhesion. J Biol Chem 1993; 268:1847-1853. Biochem 1990; 59:783-836.
88. Humphries M, Komoriya A, Akiyama K et al. Identifica- 106. Tang L, Chesney J, Sherry B, Eaton JW. The pivotal role
tion of two distinct regions of the type IIIcs connecting of cytokines in the recruitment and adhesion of inflam-
segment of human plasma fibronectin that promote cell matory cells to implanted biomaterials. Trans Soc Biomat
type-specific adhesion. J Biol Chem 1987; 262:6886-6892. 1997b; 20:127.
89. McCarthy JB, Skubitz APN, Zhao Q, Yi X-Y et al. RGD- 107. Standiford TJ, Kunkel SL, Lukacs NW et al. Macrophage
independent cell adhesion to the carboxy-terminal hep- inflammatory protein-1 alpha mediates lung leukocyte
arin-binding fragments of fibronectin involves heparin recruitment, lung injury, and early mortality in murine
dependent and independent activities. J Cell Biol 1990; endotoxemia. J Immunol 1995; 155:1515-1524.
110:777-787. 108. Kristensen MS, Deleuran BW, Larsen GG et al. Expres-
90. Haugen PKJ, McCarthy JB, Skubitz APN et al. Recogni- sion of monocyte chemotactic and activating factor
tion of the A chain carboxyl-terminal heparin binding (MCAF) in skin related cells: a comparative study.
region of fibronectin involves multiple sites: Two contigu- Cytokines 1995; 5:520-524.
ous sequences act independently to promote neural cell 109. Smith RE, Streiter RM, Zhange K et al. A role of C-C
adhesion. J Cell Biol 1990; 111:2733-2745. chemokines in fibrotic lung diseases. J Leukoc Biol 1995;
91. Ugarova TP, Budzynski AZ, Shattil SJ et al. Conforma- 57:782-787.
tional changes in fibrinogen elicited by its interaction with 110. Alam R, Kumar D, Anderson-Walters D, Forsythe PA.
platelet membrane glycoprotein GPIIb-IIIa. J Biol Chem Macrophage inflammatory protein-1 alpha and monocyte
1993; 268:21080-21087. chemoattractant peptide-1 elicit immediate and late cu-
92. Cooper JA, Lo SK, Malik AB. Fibrin is a determinant of taneous reactions and activate murine mast cells in vivo.
neutrophil sequestration in the lung. Circ Res 1988; J Immunol 1994; 152:1298-1303.
63:735-741. 111. Koch AE, Kunkeln SL, Harlow LA et al. Macrophage in-
93. Behrens BL, Clark RA, Presley DM et al. Comparison of flammatory protein-1 alpha—A novel chemotactic
the evolving histopathology of early and late cutaneous cytokine for macrophages in rheumatoid arthritis. J Clin
and asthmatic responses in rabbits after a single antigen Invest 1994; 93:921-928.
challenge. Lab Invest 1987; 56: 101-113. 112. Strieter RM, Koch AE, Antoney VB et al. The immuno-
94. Rowland TN, Donovan M, Gillies C et al. Fibrin media- pathology of chemotactic cytokines: The role of
tor of in vivo and in vitro injury and inflammation. Curr interleukin-8 and monocyte chemoattractant protein-1. J
Eye Res 1985; 4:537-553. Lab Clin Med 1994; 123:183-197.
95. Colvin RB, Johnson RA, Mihm MC Jr, Dvorak HF. Role 113. Wolpe SD, Cerami A. Macrophage inflammatory proteins
of the clotting system in cell-mediated hypersensitivity. 1 and 2: members of a novel superfamily of cytokines.
1. Fibrin deposition in delayed skin reaction in man. J FASEB J 1989; 3:2565-2573.
Exp Med 1973; 138:686-698. 114. Kasama T, Strieter RM, Standiford TJ et al. Expression
96. Muchowski PG, Zhang L, Chang ER et al. Functional in- and regulation of human neutrophil-derived macrophage
teraction between the integrin antagonist neutrophil in- inflammatory protein-1 alpha (MIP-1 alpha). J Exp Med
hibitory factor and the I domain of the Cd11b/cd18. J 1994; 178:63-72.
Biol Chem 1994; 269:26419-26423. 115. Guo W, Willen R, Andersson R et al. Morphological re-
97. Moyle M, Foster DL, McGrath DE et al. A hookworm sponse of the peritoneum and spleen to intraperitoneal
glycoprotein that inhibits neutrophil function is a ligand biomaterials. Internat J Artif Organs 1993; 16:276-284.
of the integrin Cd11b/cd18. J Biol Chem 1994; 269:10008- 116. Tang L, Jennings TA, Eaton JW. Mast cells mediate acute
10015. inflammatory responses to implanted biomaterials. Proc
98. Tang L, Welty S, Smith CW, Eaton JW. The participation Natl Acad Sci 1998; 95:8841-8846.
of adhesion molecules in inflammatory responses to 117. Riley JF, West GB. The presence of histamine in tissue
biomaterials. Trans Soc Biomat 1997a; 20:261. mast cells. J Physiol 1953; 129:528-537.
99. Lo SK, Detmers PA, Levin SM, Wright SD. Transient ad- 118. Okamoto H, Nakano K. Regulation of interleukin-1 syn-
hesion of neutrophils to endothelium. J Exp Med 1989; thesis by histamine produced by mouse peritoneal mac-
169:1779-1794. rophages per se. Immunology 1990; 69:162-165.
100. Witthaut R, Farhood A, Smith C, Jaeschke H. Comple- 119. Zwadlo-Klarwasser G, Braam U, Muhl-Zurbes P,
ment and TNF# contribute to Mac-1 (Cd11b/cd18) up- Schmutzler W. Macrophages and lymphocytes: Alterna-
regulation and systemic neutrophil activation during tive sources of histamine. Agents Actions 1994;
endotoxemia in vivo. J Leuk Biol 1994; 55:105-111. 41:C99-C100.
101. Nathan C, Sporn M. Cytokines in context. J Cell Biol 120. Franzen L. Further studies on the relationship between
1991; 113:981-986. drug-induced mast cell secretion and local cell prolifera-
tion. Acta Path Microbiol Scand 1981; 89:57-62.
218 Tissue Engineering of Prosthetic Vascular Grafts
121. Zhang Y, Ramos BF, Jakschik BA. Neutrophil recruitment 124. Norrby K, Enestrom S. Cellular and extracellular changes
by tumor necrosis factor from mast cells in immune com- following mast-cell secretion in avascular rat mesentery.
plex peritonitis. Science 1992; 258:1957-1959. Cell Tissue Res 1984; 235:339-345.
122. Ramos BF, Zhang Y, Qureshi R, Jakschik BA. Mast cells 125. Yoffe JR, Taylor DJ, Woolley DE. Mast-cell products and
are critical for the production of leukotrienes responsible heparin stimulate the production of mononuclear-cell fac-
for neutrophil recruitment in immune complex-induced tor by cultured human monocyte/macrophages. Biochem
peritonitis in mice. J Immunol 1991; 147:1636-1641. J 1985; 230:83-88.
123. Ramos BF, Zhang Y, Angkachatchai V, Jakschik BA. Mast 126. Watanabe S, Watanabe K, Ohnishi T et al. Mast cells in
cell mediators regulate vascular permeability changes in the rat alveolar septa undergoing fibrosis after ionizing
Arthus reaction. J Pharmacol Exp Therap 1992; irradiation. Lab Invest 1974; 31:555-567.
262:559-565.
Taming of Adverse Responses
Prevention of Fibrosis
CHAPTER 21
The Accumulation of Inflammatory Mediators:
A Target for the Prevention of Fibrosis
John Zagorski, Sharon M. Wahl
Introduction
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler
©1999 R.G. Landes Company.
220 Tissue Engineering of Prosthetic Vascular Grafts
of microorganisms or antigen removal and subsequent tis- vated endothelial cells while E- and P-selectins, expressed
sue healing, a prolonged response invariably begins to dam- on the surfaces of activated endothelial cells, bind similar
age the inflamed tissue.22 Thus, chronic inflammation is al- ligands on circulating leukocytes37-39 (Fig. 21.1). Leukocyte
most always deleterious, and may in fact be viewed as a pa- rolling on activated endothelia most likely allows the leuko-
thology distinct from the transient and generally benefi- cytes time to survey the local environment for additional
cial acute inflammatory response.23 Expansion of the im- pro-inflammatory stimuli without requiring firm adhesive
mune response is mediated in large part by cytokine net- interactions that might commit the leukocyte to remaining
works which have evolved to regulate inflammation, with at that site. These additional stimuli may in large part be
numerous pro-inflammatory proteins functioning to pro- contributed by locally expressed chemoattractants, prima-
mote, and equally numerous immune-suppressive proteins rily members of the chemokine superfamily.40-43 Exposure
acting to attenuate, the inflammatory response. So long as a of rolling leukocytes to sufficiently high concentrations of
proper balance of pro- and anti-inflammatory mediators is chemoattractants causes a marked alteration in the adhe-
maintained within traumatized tissue, normal clearance of sive interactions between leukocytes and endothelial cells.
pathogens and tissue repair proceed. Dysregulation of the Selectin-mediated adhesion is dramatically reduced, in part,
cytokine balance, by various mechanisms, is at the core of by proteolytic shedding of L-selectin from the leukocyte
persistent, chronic inflammation, leading to the seemingly surface.44 However, prolonged expression of endothelial P-
contradictory results of tissue destruction and selectin has been observed.45
hyperproliferation (fibrosis).24-26 Concurrent with loss of selectin-mediated adhesion,
The grafting of an artificial prosthesis within normal leukocytes begin the second step in vascular emigration,
tissue, the deliberate placement of a foreign object within a adherence to endothelium via tight integrin-mediated in-
body, may be considered as several distinct and deliberate teractions.46-49 The three leukocyte integrins, LFA-1, Mac-1
pathological insults. First, the surgery involved is a controlled and p150,95, are heterodimeric proteins containing a shared
wound which must be managed in such a way as to maxi- !2 subunit (CD18) and one of three different # subunits, #L,
mize normal wound healing. Second, some degree of infec- (Cd11a), #m (Cd11b) or #x (CD11c). Among the multiple
tion often accompanies the grafting procedure and an acute other integrins present on leukocytes, !1 integrins can inter-
inflammatory response is then inevitable. Lastly, and most act with both endothelial cells and extracellular matrix mol-
importantly, the emplacement of a prosthetic device into a ecules.46 For example, #4!1 (VLA-4) receptors bind to re-
host may be interpreted by the host immune system as an gions of fibronectin, but also recognize VCAM, the corre-
immense “pathogen”, and may initiate chronic rejection at- sponding ligand on endothelial cells. Integrin-mediated leu-
tempts by effector cells of the innate and/or adaptive im- kocyte-endothelial adhesion is, at least in part, directly in-
mune system. Ideally, the process of grafting should not in- creased by the actions of chemoattractants on leukocytes.48,49
terfere with the normal acute inflammatory response, but This helps to insure that as leukocytes lose selectin-medi-
also should not evoke such a prolonged and unresolvable ated adhesive interactions in the intimate proximity of en-
stimulus that a chronic response spins out of control. As dothelial cells, a second type of adhesion is rapidly initiated.
such, it is critical that the prosthesis be constructed in such Endothelial ligands for the leukocyte integrins include the
a manner that it evokes as little recognition by the immune ICAM family of immunoglobulin-like adhesion molecules
system as possible. In particular, the biomaterials used in and VCAM.50-51 These molecules are differentially regulated
the construction of the prosthesis should be inert in the sense on the endothelial cell surface and are upregulated by such
that they do not promote activation of immune cells.27-28 In pro-inflammatory mediators as IL-1, TNF-# and IFN-&.
this review we will attempt to review the mechanisms by Integrin-ICAM and -VCAM adhesive interactions serve to
which inflammatory responses become prolonged and at- secure the leukocytes at the inflammatory site and also trans-
tempt to identify pro-inflammatory mediators which may duce signals which are necessary for migration through the
be attractive targets for mitigating some of the side effects endothelial monolayer and underlying basement membrane,
of prostheses and preventing fibrosis. the final step in emigration (diapedesis)52,53 (Fig. 21.1).
of immune cells from the vasculature. It is clear, however, humans, the CC gene cluster is located on chromosome 17,
that mechanisms do exist for the recruitment of specific cell while the CXC cluster is located on human chromosome 4.43
populations, since this has been observed histologically in It seems likely that the various members of each family arose
many pathological conditions.57-59 through recombination-mediated gene duplication of single
Within the past decade, a new group of related ancestral CC and CXC progenitors, a hypothesis with inter-
polypeptides, the chemokines, has emerged as a major source esting implications concerning the evolution of the mam-
of chemotactic activity responsible for promoting immune malian inflammatory response.
cell recruitment in numerous inflammatory pathologies.40-43 Members of the CXC chemokine family include IL-8,
Chemokines are a large superfamily of related proteins char- MIG, NAP-2, IP-10, ENA-78, and the three GRO/MIP-2 pro-
acterized by their close similarities in size and protein ter- teins (GRO#, !, and &). The CC chemokines are a somewhat
tiary structure. This superfamily draws its name from the more diverse family and include MCP-1, -2, -3, -4 and -5,
most important characteristic of the individual proteins, Rantes, I-309, Eotaxin-1 and -2, and MIP-1 #, ! and &
namely, that they are chemotactic cytokines. An additional (Fig. 21.2). The advent of high throughput DNA sequenc-
hallmark of the chemokine superfamily is the presence in ing continues to add putative chemokines to the databases
each protein of four positionally conserved cysteine residues. far faster than they can be characterized for function, and
The organization of the cysteines allows a convenient and the list above is only partial. While most of the current rep-
functionally significant subdivision of the proteins into two ertoire of chemokines were first isolated as cDNAs, either
major chemokine families (Fig. 21.2). In the !-chemokines, from specific or subtracted library screens or from random
these first two cysteines are adjacent (“CC”) while in the sequencing, many of the chemokine proteins were first pu-
#-chemokines the first two cysteines are separated by a single rified by traditional biochemical methods based on their ac-
apparently nonconserved amino acid (“CXC”). The genes tivities.60-63 For two very recent, but rapidly outdated, com-
for CC and CXC chemokines are arranged in clusters and in pilations of chemokines and chemokine ESTs (expressed
222 Tissue Engineering of Prosthetic Vascular Grafts
sequence tags) the reader is referred to Wells and Peitsch42 cells that have migrated through the membrane and become
and Baggiolini et al.43 adherent to its basal surface, or by recovering cells floating
In general, CXC chemokines are chemotactic for neu- in the basal chamber medium.75
trophils while CC chemokines are active on mononuclear Although an oversimplification of what happens in
cells, predominantly monocytes and T lymphocytes and, for vivo, this assay has provided the foundation for understand-
some chemokines, basophils and eosinophils.64-69 It is this ing inflammatory cell recruitment. However, it has long been
cell type selectivity that separates the chemokines from the recognized that one of the properties shared by chemokines
more “traditional” chemoattractants such as complement (and many other proteins) is their affinity for charged mac-
fragments, leukotrienes and bacterial fMLP and that has romolecules. Most chemokines are positively charged pro-
stimulated research on them. In fact, some chemokines have teins and bind with high affinity to negatively charged bio-
exquisite cell type specificity as evidenced by the activities logical macromolecules such as heparin.43 This physical
of the CC chemokines, MIP-1# and MIP-1!. Both of these property of chemokines makes it unlikely that chemokines
proteins attract monocytes but also selectively attract dif- are always found in a “soluble” form at pro-inflammatory
ferent populations of T cells, with MIP-1# active on CD8 sites in vivo. Rather, it is becoming more obvious that
positive cells but MIP-1! active on CD4 positive T cells.70,71 chemokines in vivo may be bound by extracellular matrix
In contrast, the recently reported CC chemokine DC-CK1 (ECM) components and on the surfaces of cells, where they
is chemotactic for naive, CD45RA positive T cells,72 unlike are “presented” to passing leukocytes.76 This phenomenon,
other T cell active chemokines like MIP-1# and MIP-1!, the migration of cells along a gradient of immobilized
which only recruit activated T lymphocytes. The specifici- chemoattractants, is commonly referred to as haptotaxis. The
ties exhibited by chemokines in in vitro assays support the soluble phase chemotaxis induced by chemokines in vitro
numerous observations demonstrating cell type specific re- may only be an approximation of the haptotaxis that is ac-
cruitment in vivo, and offer them as strong candidates for tually generated by solid phase gradients in vivo.
mediators in the associated pathologies. It is possible that The likely physiological significance of chemokine
the unique combination of specificity and redundancy adherence on tissue ECM has important implications for
within the chemokine superfamily may allow therapeutic prostheses. A priori, polymers used for the construction of
inhibition of single chemokines to be efficacious in treating prosthetic devices dictate the electrostatic charge of the pros-
human inflammatory disease without simultaneously sup- thesis in vivo.27 Studies have demonstrated that charged
pressing immune surveillance. polymers are differentially coated with plasma proteins when
Chemotactic activities are usually measured in in vitro implanted into animals and that the kinetics of protein
assays. While many devices are currently used, most are deri- deposition do not necessarily reflect the relative plasma
vations of the Boyden chamber.73,74 However, the physical abundance of the adhering proteins.27,77 Thus, implanted
nature of chemotactic gradients in vitro and in vivo may be polymers are not necessarily preferentially coated with the
very different, leading to some profound physiological con- most abundant serum proteins, namely, albumin
sequences. In Boyden chamber type assays, leukocytes in an and immunoglobulins.
upper chamber are separated from the putative chemo- Circulating or locally expressed proteins with high
attractant in a lower chamber by a polycarbonate membrane affinity for a synthetic matrix may preferentially deposit on
containing 3 ∝m-8 ∝m diameter pores. The membrane mim- the matrix even if they are present at relatively low concen-
ics an endothelial cell layer, separating the leukocytes from trations. Simplistically, since many biopolymers are nega-
the source of chemoattractant and minimizing random cell tively charged, and most chemokines are positively charged,
migration. Cells that migrate in response to the chemo- it is reasonable to speculate that an implanted polymer may
attractant are usually quantitated by staining and counting become coated with significant amounts of chemokine pro-
Inflammatory Mediators: A Target for the Prevention of Fibrosis 223
tein, either directly or indirectly via secondary interactions. response with the generation of IL-4 and IL-10, which can
This effect could easily generate very high and persistent local dampen immune functions.80 Thus, glucocorticoids act as
haptotactic gradients, resulting in sustained leukocyte re- genetic switches, turning off or on entire sets of genes which
cruitment from adjacent microvascular endothelia. Worse control inflammation81,82 and cellular proliferation.83 Un-
still, the very high densities of adherent chemotactic pro- fortunately, chronic use of glucocorticoids is associated with
teins that may decorate implanted biopolymers are likely to severe side effects,84 including antifibrotic effects which may
strongly activate freshly recruited cells (see below). Since the impair wound healing.
biopolymer “matrix” to which the chemokines may become The other major category of drugs, the NSAIDs, in-
immobilized is not likely to turn over, implanted biopoly- hibit the cyclo-oxygenase pathway to reduce biosynthesis of
mers may become “sinks” for concentrating chemotactic fac- prostaglandins, with resulting anti-inflammatory, and an-
tors, resulting in indefinite leukocyte recruitment. It is this algesic, consequences.85,86 Though useful for the treatment
persistence of leukocyte recruitment and activation which of minor inflammatory disease or as part of a combination
results in overproduction of cytokines and growth factors, therapy, NSAIDs such as aspirin, indomethacin, aceta-
leading to chronic inflammatory diseases and fibrosis minophen, and ibuprofen, are relatively weak, and when used
(Fig. 21.1). Although these growth factors are essential for at high doses can have toxic gastrointestinal and renal side
recruitment of fibroblasts, fibroblast proliferation, and ma- effects.87
trix synthesis in the induction of tissue repair, excess quan- There is no doubt that powerful new anti-inflamma-
tities of such factors in the microenvironment of a chronic tory agents are needed which combine the potency of glu-
inflammatory lesion are causative in pathologic fibrotic cocorticoids without the negative side effects. Some such
sequelae. products are now emerging as a consequence of the suc-
cesses of molecular biology and biotechnology over the past
Mitigation of Recruitment two decades. The systematic dissection of the mammalian
Termination of the inflammatory response is as im- immune system has led to the identification of many pro-
portant as its initiation, and the persistence of chronic in- and anti-inflammatory cytokines with clinical potential. In
flammatory states may best be considered as a failure to addition, the numerous adhesion molecules that are a pre-
modulate the immune response rather than its uncontrolled requisite for both normal and aberrant migration of leuko-
initiation. It is notable in this respect that most chemo- cytes between lymphoid and systemic circulatory pathways
attractants have inherent self-limiting properties. In general, and between the circulation and tissues make attractive tar-
the chemotactic dose-response curve for most chemo- gets for synthetic inhibitors.
attractants is bell shaped, and as protein concentration in- An obvious approach for suppressing inflammation
creases beyond a maximum effective concentration, chemo- is by inhibiting leukocyte recruitment to the inflammatory
tactic activity decreases. However, at these higher concen- site. The “three step” model leading to leukocyte diapedesis
trations, chemoattractants tend to activate their target cells, discussed earlier offers several options for intervention. It
inducing in neutrophils, for example, degranulation, oxida- might be possible to inhibit the rolling of cells along acti-
tive burst and increased adhesiveness. Chemotactic dose vated endothelia, thus preventing any possibility of subse-
dependence may automatically trigger cells responding to a quent adhesion and chemotaxis. Synthetic analogs of the
local chemotactic gradient to cease migration near the in- carbohydrate ligand for L-selectin have shown promise in
flammatory site and commence their effector functions preclinical models of acute inflammation.88-90 However,
(Fig. 21.1). Combining chemotactic and activating functions selectins are crucial for normal recirculation of leukocytes
into single molecules may ensure a smooth transition from between the lymphoid and systemic circulations, and anti-
one phenotype to another as leukocytes migrate up the selectin drugs may adversely perturb this process. Integrins
chemokine gradient to their intended destination. This po- are also excellent candidates as targets for inhibitors. Reduc-
tential adaptive advantage is, however, offset by the conse- ing the strong adhesion between leukocytes and endothelial
quence that local conditions supporting leukocyte recruit- cells necessary for diapedesis should limit cellular recruit-
ment and activation may become self-sustaining. ment into inflamed tissue, and many studies have supported
There exists in the current pharmacopia of medicine this concept in vivo.46,91,92 In this regard, synthetic peptides
a wide variety of drugs used for the control of inflamma- derived from the A chain of fibronectin have been shown to
tion. The most commonly used and effective anti- not only block integrin-dependent adhesion,91,92 but also to
inflammatories can probably be assigned to one of two cat- interfere with matrix augmented cytokine signal transduc-
egories, glucocorticosteroids and nonsteroidal anti-inflam- tion and cellular activation. 93 These promising anti-
matory drugs (NSAIDs).78 Steroids, such as prednisolone inflammatory peptides have proven beneficial in animal
and dexamethasone, are very powerful anti-inflammatory models of arthritis, autoimmunity91,92,94,95 and liver fibrosis
agents that exert their effects through regulation of gene (Wahl SM, unpublished data).
expression. Many pro-inflammatory cytokine genes contain Another possibility for suppressing cellular recruit-
glucocorticoid responsive elements within their promoters ment might be to target chemotactic factors themselves, in
which contribute to the regulation of their expression.79 particular by identifying chemoattractant receptor antago-
Exogenous administration of high levels of steroids as thera- nists. If a specific chemokine can be identified as the major
peutics suppresses gene expression of these cytokines, with chemoattractant associated with a given disease, antagonists
the obvious result of limiting accumulation of the proteins of that chemokine might demonstrate efficacy without sig-
themselves. Glucocorticoids can also promote a Th2 cytokine nificant adverse effects or immune suppression. The latter
224 Tissue Engineering of Prosthetic Vascular Grafts
hypothesis derives from the obvious redundancy among shown promise in animal studies. An MCP-1ra has been used
members of the chemokine superfamily, and assumes that to successfully ameliorate experimental arthritis in mice,107
any systemic immune defect caused by specific inhibition and a CINCra (cytokine-induced neutrophil chemo-
of one chemokine will be obscured by the remaining, over- attractant, a rat CXC chemokine) reduced peritoneal inflam-
lapping activities of other chemokines. mation induced by injection of zymosan.106 The specificity
The specificity of chemokines for their respective tar- that chemokine receptor antagonists predictably inherit from
get cells is determined by the relative distribution of their normal counterparts might initiate a new paradigm
chemokine receptors expressed by the various populations for anti-inflammatory selectivity, which is desirable for any
of myeloid cells. Not surprisingly, there exist separate re- new generation of drugs. The need for specificity is exem-
ceptors for the two families of chemokines, CXC receptors plified by observations that similar CXC chemokines such
and CC receptors.96-98 Interestingly, the chemokine recep- as IL-8 (ELR-containing) and IP-10 (non-ELR) exert op-
tors display a wide range of specificities for the chemokines posing influences in idiopathic pulmonary fibrosis, with IL-8
themselves, with some receptors binding a single ligand and promoting, and IP-10 suppressing, angiogenic activity.108
others binding many ligands within a chemokine family. A simpler, but perhaps less obvious approach toward
When all the data are combined, they reveal the existence of antagonizing chemoattractants derives from the spatial re-
networks of redundant ligands and receptors for all types of strictions necessary for generating gradients. In vivo, endo-
myeloid cells, suggesting that recruitment of specific cell thelial layers and basement membranes serve a barrier func-
types in inflammation is dependent on temporal and spa- tion between leukocyte and chemoattractant to generate a
tial sequestration of chemokine expression. chemotactic gradient. With leukocytes circulating in the
Two neutrophil receptors have been identified for CXC vasculature and chemoattractants originating in the tissues,
chemokines which have very different properties (Fig. 21.2). the gradient flows inward towards the blood. It follows that
Both receptors were originally characterized as receptors for by intentionally increasing the concentration of chemo-
IL-8 and were thus named type A and type B IL-8 receptors. attractant in the blood, local gradients associated with in-
However, the type B receptor binds several other CXC flammatory sites should collapse. Simply put, administer-
chemokines besides IL-8, including GRO#, ! and &, NAP-2 ing chemokines or other factors intravenously should have
and ENA-78. In contrast to the type B receptor, the type A a paradoxically anti-inflammatory effect. Some data support
IL-8 receptor is selective for IL-8 and binds other CXC- this concept. Systemic administration of the potent
chemokines with approximately 100-fold lower affinity. chemoattractant TGF-! has been shown to reduce arthritis
Binding of CXC-chemokines to both IL-8 receptors is me- in rats, whereas direct tissue injection of the cytokine into
diated largely, but not entirely, by the important sequence joints aggravated the disease.109,110 Similarly, systemic ad-
motif “ELR” which is located immediately upstream of the ministration of the CC chemokine MCP-1 protected mice
signature CXC domain of these chemokines. Significantly, from lethal endotoxemia, although the mechanism of ac-
the three CXC chemokines that do not bind the type B re- tion was clearly complex, since the treatment notably altered
ceptor, IP-10, MIG and PF4, all lack the ELR domain. plasma TNF-#, IL-10 and IL-12 levels as well.111 Thus, by
The family of CC chemokine receptors is far more blocking the recruitment of leukocytes from the circulation
complicated than that for the two IL-8 receptors. At least to tissue sites of persistent infection, injury and/or implan-
eight receptors for CC chemokines have been identified on tation, it may be possible to attenuate the cascade of events
various mononuclear leukocytes and are designated CCR1 responsible for chronic inflammation, tissue damage and
through CCR8.96-102 Each of the receptors is expressed on fibrotic repair. Once fibropathogenesis has developed, it be-
the surfaces of specific mononuclear cells and binds a spe- comes more difficult to effectively intervene.
cific subset of the CC chemokines, as shown in Figure 21.2,
although the receptors CCR6 and CCR7 and their ligands Summary and Perspectives
are very poorly characterized. Alone among the well char- Technology often outstrips our ability to understand
acterized CC chemokine receptors, CCR8 is apparently and adequately control it. Medical technology, and in par-
unique in having only one well defined ligand, the T lym- ticular surgery, is no exception. For instance, the techniques
phocyte chemoattractant I-309.102 of whole organ transplant were successfully developed dur-
The development of chemokine receptor antagonists ing the 1950s and 1960s, and are now common. However,
(ra) is at the earliest research stage. Structure-function analy- the barrier to successful transplantation was not the physi-
sis of CXC and CC chemokines has revealed that the N-ter- cal act of moving the organ from donor to recipient, but
mini of both families of chemokines are crucial for both convincing the recipient’s immune system to accept, or more
receptor binding and signal transduction.43 Interestingly, precisely ignore, the foreign tissue. It was the advent of im-
chemokine mutants containing N-terminal truncations have munosuppressive anti-rejection drugs like cyclosporin A that
been described which have the desired properties of high finally allowed the full potential of surgical transplants to
affinity receptor binding but negligible receptor activa- be realized.112 Yet, though we can sometimes fool the im-
tion.103-106 These chemokine receptor antagonists generally mune system into accepting our interventions, our level of
retain the receptor type specificities of their normal coun- control remains incomplete.
terparts, suggesting that they might selectively block a lim- The discovery and characterization of an ever increas-
ited number of chemokines in vivo. The potential efficacy ing number of cytokines and their receptors offers the pos-
of chemokine receptor antagonists in treating human dis- sibility that new drugs will be developed that can exercise
ease has yet to be demonstrated, but such proteins have more selective control over inflammation than is currently
Inflammatory Mediators: A Target for the Prevention of Fibrosis 225
possible. Such selectivity almost certainly offers the addi- 18. Hugo NE, Thompson LW, Zook EG et al. Effect of
tional possibility that future therapeutics will have more lim- chronic anemia on the tensile strength of healing wounds.
ited adverse effects to complement their efficacy. While it is Surgery 1969; 66:741-745.
unlikely that we will be able to completely discount drug 19. Simpson DM, Ross R. The neutrophilic leukocyte in
wound repair. A study with antineutrophil serum. J Clin
toxicity in humans, the increasing selectivity of anti-inflam-
Invest 1972; 51:2009-2023.
matory drugs engendered by a better understanding and 20. Wahl SM, Arend WP, Ross R. The effect of complement
control of the immune response will undoubtedly provide depletion on wound healing. Am J Pathol 1974; 74:73-83.
improved therapeutic opportunities. 21. Leibovich SJ, Ross R. The role of the macrophage in
wound repair: A study with hydrocortisone and
References antimacrophage serum. Am J Pathol 1975; 78:71-91.
1. Hircsch JG. Antimicrobial factors in tissues and phago- 22. Border WA, Ruoslahti E. Transformtin growth factor-beta
cytic cells. Bacteriol Rev 1960; 21:133-147. in disease, the dark side of tissue repair. J Clin Invest 1992;
2. Klebanoff SJ. Phagocytic cells: Products of oxygen metabo- 90:1-7.
lism. In: Gallin JI, Goldstein IM, Snyderman R, eds. In- 23. Fassbender, HG. The disease picture in rheumatoid ar-
flammation: basic principles and clinical correlates. New thritis as a result of different pathomechanisms. Z
York: Raven Press, 1988:391-444. Rheumatol 1985; 44:33-46.
3. Babior GL, Rosin RE, McMurrich BJ et al. Arrangement 24. Wahl SM. Fibrosis: Bacterial-cell-wall-induced hepatic
of the respiratory burst oxidase in the plasma membrane granulomas. In: Gallin JI, Goldstein IM, Snyderman R,
of the neutrophil. J Clin Invest 1981; 67:1724-1728. eds. Inflammation: Basic principles and clinical correlates.
4. Elsbach P, Weiss J. A re-evaluation of the roles of the New York: Raven Press, 1988: 841-859.
O2-dependent and O2-independent microbicidal systems 25. Friedman SL. Molecular mechanisms of hepatic fibrosis
of phagocytes. Rev Infect Dis 1983; 5:843-853. and principles of therapy. J Gastroenterol 1997;
5. Elsbach P, Weiss J. Oxygen-dependent and oxygen-inde- 32:424-430.
pendent mechanisms of microbicidal activity of neutro- 26. Morris W, Knauer CM. Cardiopulmonary manifestations
phils. Immunol Lett 1985; 11:159-163. of shistosomiasis. Semin Respir Infect 1997; 12:159-170.
6. Janoff A, Scherer J. Mediators of inflammation in leuko- 27. Marchant RE, Wang IW. Physical and chemical aspects
cyte lysosomes. IX. Elastinolytic activity in granules of of biomaterials used in humans. In: Greco RS, ed. Im-
human polymorphonuclear leukocytes. J Exp Med 1968; plantation Biology: The Host Response to Biomedical
128:1137-1155. Devices. Boca Raton: CRC Press, 1994:13-38.
7. Rabinovitch MC. Elastase and cell matrix interactions in 28. Choi ET, Callow AD. The effect of biomaterials on the
the pathobiology of vascular disease. Acta Paediatr Jpn host. In: Greco RS, ed. Implantation Biology: The Host
1995; 37:657-666. Response to Biomedical Devices. Boca Raton: CRC Press,
8. Doring G. The role of neutrophil elastase in chronic in- 1994:39-54.
flammation. Am J Resp Crit Care Med 1994; 29. Butcher EC. Leukocyte-endothelial cell recognition: Three
150:S114-117. (or more) steps to specificity and diversity. Cell 1991;
9. Lazarus GS, Brown RS, Daniels JR et al. Human granulo- 67:1033-1036.
cyte collagenase. Science 1968; 159:1483-1485. 30. Arfors KE, Lundberg C, Lindborm L et al. A monoclonal
10. Shingleton WD, Hodges DJ, Brick P et al. Collagenase: A antibody to the membrane glycoprotein complex CD18
key enzyme in collagen turnover. Biochem Cell Biol 1996; inhibits polymorphonuclear leukocyte accumulation and
74:759-775. plasma leakage in vivo. Blood 1987; 69:338-340.
11. Yamamoto K. Cathepsin E and cathepsin D: Biosynthe- 31. Ley K, Lundgren E, Berge E et al. Shear dependent inhi-
sis, processing and subcellular location. Adv Exp Med Biol bition of granulocyte adhesion to cultured endothelium
1995; 362:223-229. by dextran sulfate. Blood 1989; 73:1324-1330.
12. Peterson MW, Walter ME, Nygaard SD et al. Effect of 32. Argenbright LW, Letts LG, Rothlein R. Monoclonal anti-
neutrophil mediators on epithelial permeability. Am J bodies to the leukocyte membrane CD18 glycoprotein
Resp Cell Mol Biol 1995; 13:719-727. complex and to intracellular adhesion molecule-1 inhibit
13. Chapman HA, Biese RJ, Shi GP. Emerging roles for cys- leukocyte-endothelial adhesion in rabbits. J Leuk Biol
teine proteases in human biology. Ann Rev Physiol 1997; 1991; 49:253-257.
59:63-88. 33. Springer TA. Traffic signals on endothelium for lympho-
14. Durum SK, Oppenheim JJ. Macrophage derived media- cyte recirculation and leukocyte emigration. Ann Rev
tors: Interleukin 1, tumor necrosis factor, interleukin 6, Physiol 1995; 57:827-872.
and related cytokines. In: Gallin JI, Goldstein IM, 34. McEver RP, Moore KL, Cummings RD. Leukocyte traf-
Snyderman R, eds. Inflammation: Basic Principles and ficking mediated by selectin-carbohydrate interactions. J
Clinical Correlates. New York: Raven Press, 1988:639-662. Biol Chem 1995; 270:11025-11028.
15. Wahl SM. Acute and chronic inflammation. In: Asherson 35. Rossitter H, Alon R, Kupper TS. Selectins, T-cell rolling
GL, Zembala M, eds. Human monocytes. New York: Aca- and inflammation. Mol Med Today 1997; 3:214-222.
demic Press, 1989:361-371. 36. Lasky LA. Selectin-carbohydrate interactions and the ini-
16. Nathan C. Mechanisms and modulation of macrophage tiation of the inflammatory response. Ann Rev Biochem
activation. Behring Inst Mitt 1991; 88:200-207. 1995; 64:113-139
17. Stein JM, Levinson SM. Effect of the inflammatory reac- 37. Varki A. Selectin ligands. Proc Natl Acad Sci USA 1994;
tion on subsequent wound healing. Surg Forum 1966; 91:7390-7397.
17:484-485. 38. Kansas GS. Selectins and their ligands: Current concepts
and controversies. Blood 1996; 88:3259-3287.
226 Tissue Engineering of Prosthetic Vascular Grafts
39. Varki A. Selectin ligands: Will the real ones please stand defense cytokines. Proc Natl Acad Sci USA 1987; 84:9233-
up? J Clin Invest 1997; 99:158-162. 9236.
40. Oppenheim JJ, Zachariae COC, Mukaida N et al. Proper- 61. Matsushima K, Larsen CG, DuBois GC et al. Purification
ties of the novel proinflammatory supergene “intercrine” and characterization of a novel monocyte chemotactic and
family. Ann Rev Immunol 1991; 9:617-648. activating factor produced by a human myelomonocytic
41. Kelvin DJ, Michiel DF, Johnston JA et al. Chemokines cell line. J Exp Med 1989; 169:1485-1490.
and serpentines: the molecular biology of chemokine re- 62. Sherry B, Tekamp-Olson P, Gallegos C et al. Resolution
ceptors. J Leuk Biol 1993; 54:604-612. of the two components of macrophage inflammatory pro-
42. Wells TNC, Peitsch MC. The chemokine information tein 1, and cloning and characterization of one of those
source: Identification and characterization of novel components, macrophage inflammatory protein 1!. J Exp
chemokines using the worldwideweb and expressed se- Med 1988; 168:2251-2259.
quence tag databases. J Leuk Biol 1997; 61:545-550. 63. Nakagawa H, Komorita N, Shibata F et al. Identfication
43. Baggiolini M, Dewald B, Moser B. Human chemokines: of cytokine-induced neutrophil chemoattractants (CINC),
An update. Ann Rev Immunol 1997; 15:675-705. rat GRO/CINC-2# and CINC-2!, produced by granula-
44. Kahn J, Ingraham RH, Shirley F et al. Membrane proxi- tion tissue in culture: Purification, complete amino acid
mal cleavage of L-selectin; identification of the cleavage sequences and characterization. Biochem J 1994;
site and a 6kD transmembrane peptide fragment of L- 301:545-551.
selectin. J Cell Biol 1994; 125:461-470. 64. Geiser T, Dewald B, Ehrengruber MU et al. The
45. Gober JS, Bowen BL, Ebling H et al. Monocyte-endothe- interleukin-8-related chemotactic cytokines GRO#, GRO!,
lial adhesion in chronic rheumatoid arthritis: In situ de- and GRO& activate human neutrophil and basophil leu-
tection of selectin and integrin-dependant interactions. J kocytes. J Biol Chem 1993; 268:15419-15424.
Clin Invest 1993; 91:2609-2619. 65. Weber M, Uguccioni M, Ochensberger B et al. Monocyte
46. Wahl SM, Feldman GM, McCarthy JB. Regulation of leu- chemotactic protein MCP-2 activates human basophil and
kocyte adhesion and signaling in inflammation and dis- eosinophil leukocytes similar to MCP-3. J Immunol 1995;
ease. J Leuk Biol 1996; 59:789-796. 154:4166-4172.
47. Loftus JC, Liddington RC. Cell adhesion in vascular biol- 66. Forssmann U, Uguccioni M, Loetscher P et al. Eotaxin-2,
ogy. New insights into integrin-ligand interaction. J Clin a novel CC chemokine that is selective for the chemokine
Invest 1997; 99:2302-2306. receptor CCR3, and acts like eotaxin on human eosino-
48. Carr MW, Alon R, Springer TA. The C-C chemokine phil and basophil leukocytes. J Exp Med 1997; 185:2171-
MCP-1 differentially modulates the avidity of !1 and !2 2176.
integrins on T lymphocytes. Immunity 1996; 4:179-187. 67. Proost P, Wuyts A, Van Damme J. Human monocyte
49. Burns AR, Simon SI, Kukielka GL et al. Chemotactic fac- chemotactic proteins-2 and -3: Structural and functional
tors stimulate CD18-dependent canine neutrophil adher- comparison with MCP-1. J Leuk Biol 1996; 59:67-74.
ence and motility on lung fibroblasts. J Immunol 1996; 68. Garcia-Zepeda EA, Rothenberg ME, Ownbey RT et al.
156:3389-3401. Human eotaxin is a specific chemoattractant for eosino-
50. Madri JA, Graesser D, Haas T. The roles of adhesion phil cells and provides a new mechanism to explain tis-
molecules and proteinases in lymphocyte transendothelial sue eosinophilia. Nature Med 1996; 2:449-456.
migration. Biochem Cell Biol 1996; 74:749-757. 69. Kita H, Gleich GJ. Chemokines active on eosinophils.
51. Lefkowith, JB. Leukocyte migration in immune complex Potential roles in allergic inflammation. J Exp Med 1996;
glomerulonephritis: Role of adhesion receptors. Kidney Int 183:2421-2426.
1997; 51:1469-1475. 70. Schall TJ, Bacon K, Camp RDR et al. Human macroph-
52. Newton RA, Thiel M, Hogg N. Signaling mechanisms and age inflammatory protein 1 # (MIP-1 # ) and MIP-1 !
the activation of leukocyte integrins. J Leuk Biol 1997; chemokines attract distinct populations of lymphocytes. J
61:422-426. Exp Med 1993; 177:1821-1830.
53. Ruoslahti E. Integrins as signaling molecules and targets 71. Taub D, Conlon K, Lloyd AR et al. Preferential migra-
for tumor therapy. Kidney Int 1997; 51:1413-1417. tion of activated CD4+ and CD8+ T cells in response to
54. Ryan GB. The origin and sequence of the cells found in MIP-1# and MIP-1!. Science 1993; 260:355-358.
the acute inflammatory response. Aust J Exp Biol Med 72. Adema GJ, Hartgers F, Verstraten R et al. A dendritic-
Sci 1967; 45:149-162. cell-derived C-C chemokine that preferentially attracts
55. Wahl SM. Transforming growth factor beta in inflamma- naive T cells. Nature 1997; 387:713-717.
tion. A cause and a cure. J Clin Immunol 1992; 12:61-74. 73. Snyderman R, Altman LC, Hausman MS et al. Human
56. Frank MM, Fries LF. Complement. In: Gallin JI, Goldstein mononuclear leukocyte chemotaxis: A quantitative assay
IM, Snyderman R, eds. Inflammation: Basic principles and for mediators of humoral and cellular chemotactic fac-
clinical correlates. New York: Raven Press, 1988: 679-702. tors. J Immunol 1972; 109:857-860.
57. Standiford TJ, Kunkel SL, Greenberger MJ et al. Expres- 74. Altman LC, Snyderman R, Oppenheim JJ et al. A human
sion and regulation of chemokines in bacterial pneumo- mononuclear leukocyte chemotactic factor: Characteriza-
nia. J Leuk Biol 1996; 59:24-28. tion, specificity and kinetics of production by homolo-
58. Schroder JM, Noso N, Sticherling M et al. Role of eosi- gous leukocytes. J Immunol 1973; 110:801-810.
nophil-chemotactic C-C chemokines in cutaneous inflam- 75. Zagorski J, DeLarco JE. Expression of active human GRO!
mation. J Leuk Biol 1996; 59:1-5. and GRO& neutrophil chemotactic proteins in E. coli. Prot
59. Baggiolini M, Dahinden CA. CC chemokines in allergic Exp Purif 1994; 5:337-345.
inflammation. Immunol Today 1994; 15:127-133. 76. Tanaka Y, Adams DH, Hubscher S et al. T-cell adhesion
60. Yoshimura TK, Matsushima K, Tanaka S et al. Purifica- induced by proteoglycan-immobilized cytokine MIP-1!.
tion of a human monocyte-derived neutrophil chemotac- Nature 1993; 361:79-82.
tic factor that shares sequence homology with other host
Inflammatory Mediators: A Target for the Prevention of Fibrosis 227
77. Andrade JD, Hlady V. Plasma protein adsorption; the big TGF-!1 deficient mice and can be inhibited by adhesion-
twelve. Ann NY Acad Sci 1987; 516:158-172. blocking peptides. J Immunol 1996; 157:1306-1312.
78. Brooks PM, Day RO. Nonsteroidal antiinflammatory 96. Murphy PM. The molecular biology of leukocyte
drugs-differences and similarities. N Engl J Med 1991; chemoattractant receptors. Annu Rev Immunol 1994;
324:1716-1721 12:593-633.
79. Saatcioglu F, Claret FX, Karin M. Negative transcriptional 97. Raport CJ, Schweickart VL, Chantry D et al. New mem-
regulation by nuclear receptors. Seminars Cancer Biol bers of the chemokine receptor gene family. J Leuk Biol
1994; 5:347-359. 1996; 59:18-23.
80. Ramierz F, Fowell DJ, Puklavec M, Simmonds S, Mason 98. Wells TNC, Power CA, Lusti-Narasimhan M et al. Selec-
D. Glucocorticoids promote a TH2 cytokine response by tivity and antagonism of chemokine receptors. J Leuk Biol
CD4+ T cells in vitro. J Immunol 1996;156:2406-2412. 1996; 59:53-60.
81. Brattsand R, Linden M. Cytokine modulation by gluco- 99. Baba M, Imai T, Nishimura M et al. Identification of
corticoids: Mechanisms and actions in cellular studies. CCR6, the specific receptor for a novel lymphocyte-di-
Aliment Pharmacol Ther 1996; 10:81-90. rected CC chemokine LARC. J Biol Chem 1997;
82. Barnes PJ, Karin M. Nuclear factor-kappa!: A pivotal tran- 272:14893-14898.
scription factor in chronic inflammatory disease. N Engl 100. Yoshida R, Imai T, Hieshima K et al. Molecular cloning
J Med 1997; 336:1066-1071. of a novel human CC chemokine EBI1-ligand chemokine
83. Armelin MC, Oliveira ML, Mercado JM et al. Molecular that is a specific functional ligand for EBI1, CCR7. J Biol
genetic approach to cell proliferation control and neopla- Chem 1997; 272:13803-13809.
sia. Braz J Med Biol Res 1996; 29:911-919. 101. Tiffany HL, Lautens LL, Gao JL et al. Identification of
84. Harris ED. The role of glucocorticoids. In: Rheumatoid CCR8: A human monocyte and thymus receptor for the
arthritis. Philadelphia: WB Saunders, 1997:350-363. CC chemokine I-309. J Exp Med 1997; 186:165-170.
85. Lewis RA, Austen KF. Leukotrienes. In: Gallin JI, 102. Roos RS, Loetscher M, Legler DF et al. Identification of
Goldstein IM, Snyderman R, eds. Inflammation: Basic CCR8, the receptor for the human CC chemokine I-309.
Principles and Clinical Correlates. New York: Raven Press, J Biol Chem 1997; 272:17251-17254.
1988:121-128. 103. Moser B, Dewald B, Barella L et al. Interleukin-8 antago-
86. Vane JR, Botting RM. Mechanism of action of aspirin- nists generated by N-terminal modification. J Biol Chem
like drugs. Semin Arthritis Rhem 1997; 26:2-10. 1993; 268:7125-7128.
87. Clements PJ, Paulnott R. Nonsteroidal anti-rheumatic 104. Gong JH, Clark-Lewis I. Antagonists of monocyte
drugs. In: Kelly WN, Harris ED, Ruddy S et al., eds. Text- chemoattractant protein 1 identified by modification of
book of Rhematology, 5th edition. Philadelphia: WB functionally critical NH2-terminal residues. J Exp Med
Saunders, 1996:707-740. 1995; 181:631-640.
88. Kubes P, Jutila M, Payne D. Therapeutic potential of in- 105. Gong JH, Uguccioni M, Dewald B et al. Rantes and MCP-
hibiting leukocyte rolling in ischemia/reperfusion. J Clin 3 antagonists bind multiple chemokine receptors. J Biol
Invest 1995; 95:2510-2519. Chem 1996; 271:10521-10527.
89. Turunen JP, Majuri ML, Seppo A et al. De novo expres- 106. Zagorski J, Wahl SM. Inhibition of acute peritoneal in-
sion of endothelial sialyl Lewis(a) and sialyl Lewis(x) dur- flammation in rats by a cytokine-induced neutrophil
ing cardiac transplant rejection: Superior capacity of a chemoattractant receptor antagonist. J Immunol 1997;
tetravalent sialyl Lewis(x) oligosaccharide in inhibiting L- 159:1059-1062.
selectin-dependent lymphocyte adhesion. J Exp Med 1995; 107. Gong JH, Ratkay LG, Waterfield JD et al. An antagonist
182:1133-1141. of monocyte chemoattractant protein 1 (MCP-1) inhibits
90. Seppo A, Turunen JP, Penttila L et al. Synthesis of a tet- arthritis in the MRL-lpr mouse model. J Exp Med 1997;
ravalent Lewis x glycan, a high affinity inhibitor of L- 186:131-137.
selectin-mediated lymphocyte binding to endothelium. 108. Keane MP, Arenberg DA, Lynch JP et al. The CXC
Glycobiology 1996; 6:65-71. chemokines, IL-8 and IP-10, regulate angiogenic activity
91. Hines KL, Kulkarni AB, McCarthy JB et al. Synthetic in idiopathic pulmonary fibrosis. J Immunol 1997;
fibronectin peptides interrupt inflammatory cell infiltra- 159:1437-1443.
tion in TGF-!1 knockout mice. Proc Natl Acad Sci USA 109. Allen JB, Manthey CL, Hand AR et al. Rapid onset syn-
1994; 91:5187-5191. ovial inflammation and hyperplasia induced by transform-
92. Wahl SM, Allen JB, Hines KL et al. Synthetic fibronectin ing growth factor !. J Exp Med 1990; 171:231-247.
peptides suppress arthritis in rats by interrupting leuko- 110. Brandes ME, Allen JB, Ogawa Y et al. Transforming
cyte adhesion and recruitment. J Clin Invest 1994; growth factor !1 suppresses acute and chronic arthritis
94:655-662. in experimental animals. J Clin Invest 1991; 87:1108-1113.
93. McCarthy JB, Vachhani BV, Wahl SM, Finbloom DS, 111. Zisman DA, Kunkel SL, Strieter RM et al. MCP-1 pro-
Feldman MF. Human monocyte binding to fibronectin tects mice from lethal endotoxemia. J Clin Invest 1997;
enhances IFN-&-induced early signaling events. J Immunol 99:2832-2836.
1997; 159:2424-2430. 112. Rao A, Luo C, Hogan PG. Transcription factors of the
94. McCartney-Francis N, Mizel D, Frazier-Jessen M, Kulkarni NFAT family: Regulation and function. Annu Rev
A, McCarthy J, Wahl SM. Lacrimal gland inflammation Immunol 1997; 15:707-747.
is responsible for ocular pathology in TGF-!1-null mice. 113. Zagorski J, Wahl SM. Recruitment. In: Roitt IM, Delves
Am J Pathol 1997; in press. PJ, eds. Encyclopedia of Immunology, Second Edition.
95. McCartney-Francis N, Mizel D, Redman R et al. Autoim- London: Academic Press, 1998; in press.
mune Sjogren’s-like lesions develop in salivary glands of
Taming of Adverse Responses
Prevention of a Hyperplastic Intimal Response
CHAPTER 22
Pathobiology of Hyperplastic Intimal Responses
Erik L. Owens, Alexander W. Clowes
Introduction
Clinical Relevance
Percutaneous transluminal coronary angioplasty (PTCA) has an excellent primary
success rate (about 90-95%). However, approximately 30% of dilated coronary arteries de-
velop marked restenosis by 6 months.5,6 Coronary vein grafts have a 10 year patency of 50%.7,8
Infra-inguinal saphenous vein grafts have a 24% failure rate at 4 years.9 Polytetra-
fluoroethylene (PTFE) grafts in the same position have a 46% failure rate at 4 years.9 Renal
artery stenting appears to have a restenosis rate of about 44% at 1 year.10 These observations
are just a sampling of the known clinical data related to restenosis and graft failure after
therapeutic intervention. The failure of a vascular graft placed during reconstruction or the
development of restenosis in an artery that has previously undergone angioplasty (or stenting)
exposes the patient to the risk of significant morbidity and mortality. Repeat intervention or
vascular reconstruction is often the only remedy for the situation, further increasing the risk
for the patient. From an economic viewpoint, considerable costs are incurred as a result of
repeat vascular intervention. If one considers the frequency with which vascular procedures
are currently performed and the incidence of graft failure or arterial restenosis after inter-
vention, it becomes clear why such intense interest exists in understanding intimal hyperplasia.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
230 Tissue Engineering of Prosthetic Vascular Grafts
during simple arterial catheterization, endovascular proce- model form the basis for the proposed three phase para-
dures, or vein graft preparation. A rat model of a type II digm for the development of intimal hyperplasia.
injury uses gentle filament endothelial denudation of the Table 22.1 summarizes many of the putative regula-
carotid artery.24 In areas of endothelial loss, platelets may tory factors and their proposed role in intimal lesion for-
adhere and begin to form a thrombus. mation or inhibition.
Type III injuries are inflicted upon countless human
vessels each day in the form of balloon angioplasty, endart- First Phase: Proliferation
erectomy, atherectomy, and other forms of surgical repair The first cellular event after injury is the proliferation
or reconstruction. A type III injury results in transmural of a proportion of medial smooth muscle cells (SMCs).
damage. The endothelium is removed. The IEL is often dis- Medial SMCs in a normal artery proliferate at a very low
rupted, and a significant portion of the medial cells are level (0.06% per day). As soon as 24 hours after injury,
killed.24,28 An inflammatory response is initiated within the 10-30% of the medial SMCs are induced to enter the growth
vessel wall. Upon loss of the endothelium, platelets deposit cycle.29,30 The proportion of proliferating cells remains high
and thrombus forms. for several days but by 4 weeks returns to baseline levels.
Early observations of the events surrounding vascular in-
Molecular Mechanisms of Intimal Hyperplasia jury resulted in the “reaction to injury” hypothesis which
after Injury implicated platelets in the development of intimal hyper-
Much has been learned about the basic pathophysiol- plasia. 31,32 This theory was based upon three
ogy of intimal hyperplasia from the response of a rat ca- key observations:
rotid artery to a type III injury. The model is a modification 1. Platelets appear on the luminal surface of injured
of a method described by Baumgartner.28 A small Fogarty vessels immediately;
embolectomy catheter is passed into the common carotid 2. Platelets deposit #%granules which contain numer-
artery via the external carotid artery. The balloon is inflated ous growth factors for cultured SMCs including
and pulled through the common carotid artery, denuding platelet derived growth factor (PDGF), transform-
the endothelium and damaging the underlying tissue. Pro- ing growth factor (TGF)-!, and epidermal growth
gressive luminal thrombosis does not typically occur. Inti- factor; and
mal thickening occurs within 2 weeks and reaches a maxi- 3. Thrombocytopenic animals experience less intimal
mum at about 3 months. Luminal area declines transiently, thickening after balloon injury.33-35
partly because of intimal thickening and partly because the While overall intimal thickening is reduced in throm-
injured vessel contracts. The histologic and molecular bocytopenic animals, the first wave of SMC proliferation is
changes that have been documented after injury in this rat unaltered.35 Additionally, balloon-injured vessels (a type III
injury) develop more medial SMC proliferation and inti- isoforms of PDGF, PDGF-BB, and stimulation of the
mal thickening than filament-injured vessels (a type II in- PDGF-! receptor as the major migratory signal. While PDGF
jury).35 In both of these models, the endothelial layer is re- was initially isolated from platelets, all vascular wall cells in-
moved and platelets adhere to the exposed subendothelium, cluding endothelial cells, SMCs, and macrophages can ex-
releasing their cytoplasmic granules. Thus, platelet-related press this factor. PDGF is a strong mitogen in vitro, but its
effects in the two models should be similar. The fact that main effect in vivo appears to be on migration. (For a re-
more injury is inflicted upon the medial layer in the balloon view on PDGF, see ref. 60.)
injury model suggests that the specific stimulus for In vitro work in our lab has demonstrated a strong
proliferation is related to the degree of injury in this area. migratory response of baboon SMCs to PDGF-BB but not
The most logical conclusion is that a nonplatelet derived to PDGF-AA.61 Of the three possible isoforms, AA, BB, and
factor released by injury is primarily responsible for the AB, PDGF-BB is the predominant isoform in platelet #-gran-
initial wave of medial SMC proliferation and that the platelet ules in rat, pig, and baboon. Human platelets contain all three
factors must affect a portion of the process other isoforms in equal amounts.62 Since PDGF-BB is able to bind
than proliferation. to both the # and the ! form of the PDGF receptor and
Lindner and Reidy demonstrated that the initial stimu- PDGF-AA is only able to bind to the # receptor,60 this
lus for proliferation is most likely basic fibroblast growth strongly suggests that activation of the ! receptor plays a
factor (bFGF).36-38 In vitro, bFGF is a strong mitogen and is leading role in SMC migration.
released from disrupted cultured SMCs.39,40 Endothelial cells, When a polyclonal antibody to human PDGF is ad-
SMCs, and extracellular matrix all store various amounts of ministered to rats undergoing balloon injury, intimal thick-
bFGF.39 In the extracellular matrix (ECM), bFGF is stored ening is inhibited by 40%.63 Smooth muscle proliferation is
bound to heparan sulfate proteoglycans. Treatment with not affected. When the recombinant form of PDGF-BB is
heparin infusion has been shown to blunt the proliferative administered intravenously or intraarterially, SMC migra-
response, presumably by displacing the bFGF from the ma- tion is markedly stimulated and intimal thickening is in-
trix.41 Proliferating SMCs express the relevant receptor to creased.64 SMC proliferation in the media is only slightly
bFGF.38 SMC proliferation is significantly enhanced by the increased. In our lab, an antibody to the PDGF receptor-!
administration of bFGF and antibodies to bFGF reduce SMC (PDGFR-!) administered to baboons subjected to balloon
proliferation by approximately 80%.36,37,42,43 injury of the saphenous artery or PTFE graft bypass signifi-
The renin-angiotensin system may also play a role in cantly reduces intimal hyperplasia (unpublished results).
the early injury response.44 In vitro, angiotensin ii is a mito- Similar results have been obtained using specific antibodies
gen for SMCs.45 Additionally, AII induces the expression of to the PDGFR-! in baboons and in rats.62 Sirois et al have
other known mitogens.46-48 In vivo, AII receptor antagonists recently shown using immunohistochemical analysis that
and angiotensin converting enzyme (ACE) inhibitors deliv- both PDGF receptors (PDGFR-# and -!) are present in the
ered after injury each reduce SMC proliferation and inhibit normal artery wall and are markedly upregulated after in-
intimal thickening.49 SMC proliferation and intimal thick- jury. 65 Treatment with antisense oligonucleotides to
ening are enhanced by infusion of AII.50,51 PDGFR-! significantly decreases PDGFR-! expression and
Other growth factors might have roles in this early inhibits intimal thickening.
proliferative stage of lesion development including PDGF,52 The roles of PDGF-A chain and the PDGFR%# are not
thrombin,53 endothelin,54-56 insulin-like growth factor-1 yet clear. Animal studies using neutralizing antibodies to
(IGF-1),57 and catecholamines.58 The evidence for their in- PDGF-A have shown no observable effect during injury.62
volvement is, however, not as strong as that for bFGF or AII. In a Boyden chamber used to measure migration, activation
of the PDGFR-# by either PDGF-AA or -BB generates an
Second Phase: Migration inhibitory signal for baboon SMC migration.61 Analysis of
As early as 4 days after injury, SMCs begin to appear human fetal, normal adult, and atherosclerotic aortas sug-
in the intima.59 This results from migration of cells across gests a maintenance role for PDGFR-#, since levels of
the IEL, since normal rat intima contains no SMCs. This PDGF-A message ribonucleic acid (mRNA) by reverse tran-
process continues for up to one month. Only about 50% of scription-polymerase chain reaction (RT-PCR) were 100
migrating cells continue to proliferate.28, 30 times higher in normal vessels than either developing (fe-
Two distinct events must take place for cells to traverse tal) or diseased (atherosclerotic) vessels.66 The idea that
the IEL and enter the forming intima. First, there must be a PDGF-A is a “survival factor” has also been proposed.67-69
chemoattractant or some form of signal for the SMCs to In order for the SMCs to break their attachments with
initiate the journey. Secondly, the cells must be freed from the extracellular matrix, proteases are required. The act of
their surrounding matrix. Multiple factors are certainly detachment itself may serve to further activate the migra-
involved in the migratory process. Some may have dual tory process.35 Plasmin is the main fibrinolytic enzyme in
roles as both chemoattractants and as initiators of the body and is also able to degrade most matrix compo-
matrix detachment. nents. It is derived from plasminogen through the action of
As previously mentioned, platelet factors might play urokinase-like plasminogen activator (uPA) and tissue-type
a role in intimal thickening, perhaps by influencing SMC plasminogen activator (tPA).70 Urokinase-like plasminogen
migration. Platelet #-granules are known to contain PDGF. activator (uPA) is activated within 2-3 hours after injury.71,72
Most of the evidence now seems to point to one of the Tissue-type plasminogen activator (tPA) appears several days
Pathobiology of Hyperplastic Intimal Responses 233
later, around the same time as SMCs are seen entering the The role of SMC migration in the injured atheroscle-
intima.73 Tranexamic acid, a plasmin inhibitor, inhibits SMC rotic lesion is unclear, since SMCs are already present in dis-
migration after injury.74 Knockout mice studies reveal that eased human arteries. It does appear, however, from analy-
deletion of uPA reduces intimal thickening after injury. De- sis of atherectomy specimens, that migration might be the
letion of tPA has no effect and deletion of the naturally oc- predominant mode of intimal formation in human lesions.88
curring plasminogen activator inhibitor (PAI)-1 increases
thickening.75 We have observed that overexpression of PAI- Third Phase: Intimal Expansion
1 in the rat carotid inhibits intimal hyperplasia (unpublished The lesion continues to expand by SMC migration and
results). intimal proliferation. These cells also synthesize and secrete
A second class of enzymes also linked to SMC migra- matrix proteins. The largest contribution to lesion forma-
tion is the matrix metalloproteinases (MMP). The MMPs tion is made by the deposition of these matrix proteins in-
are a group of proteases found in the ECM. They digest col- cluding elastin, collagen, and proteoglycan.89 It would seem
lagen, elastin, fibronectin, laminin, and proteoglycans.76 Plas- logical that some of the factors present in early lesion devel-
min activates MMPs.77 These proteinases have been observed opment might play a role in intimal expansion. Some of the
in stimulated SMCs in culture and in SMCs from injured aforementioned factors, especially TGF-! and PDGF, are
arteries and atherosclerotic plaques. Hours after injury, known to stimulate ECM production.90 Unlike the earlier
MMP-9 (gelatinase b) appears and remains elevated for ap- stages, where a single factor appears to control activity, con-
proximately one week. Several days later, at about the same trol of the later expansion phase appears multifactorial in
time as tPA appears, MMP-2 (gelatinase a), which is present nature. No one stimulatory factor has yet been shown to
in normal artery walls, increases.72 Conditioned media con- predominate.
taining tissue inhibitor of metalloproteinases (TIMP)-1 (a Regarding lesion expansion, studies seem to indicate
natural inhibitor of MMP) completely inhibits the PDGF-BB that bFGF is unlikely to be involved in regulating intimal
induced migration of SMCs in vitro, a process that can be SMC proliferation. Antibodies to bFGF do not affect SMC
overcome by treatment with an antibody to TIMP-1. Local proliferation when administered 5 days after balloon injury
overexpression of TIMP-1 suppresses intimal thickening in of the rat carotid.91 Furthermore, no convincing evidence
the injured rat carotid model.78 In an explant model using exists that PDGF plays a role. PDGF-BB infusion weeks af-
baboon tissue developed by Kenagy, antibodies to urokinase, ter injury does produce a modest but measurable increase
tPA, and MMP-2 and -9 each inhibit migration, while addi- in intimal SMC proliferation.63 However, PDGF-B has not
tion of plasminogen increases migration.22 been detected to any measurable extent in the forming in-
Detachment of the SMCs from their extracellular tima, only in the endothelium near the wound edge.92 Rather,
matrix might be further regulated by integrins, a family of PDGF-A ligand and PDGFR-! predominate in the intima
transmembrane proteins that link the cytoskeleton to the during this time period.52 Since PDGF-A ligand can only
ECM and to adhesion molecules on other cells. Integrins bind to PDGFR-#, it appears unlikely that PDGF plays a sig-
are heterodimers made up of two noncovalently linked sub- nificant role in this phase.
units, designated # and !. Blockade of the integrin receptor As discussed previously, evidence does indicate that
#v!3 reduces PDGF-induced SMC migration in vitro and the angiotensin system may play a role in SMC proliferation
inhibits intimal hyperplasia in a rabbit carotid injury after injury. A possible role in SMC migration49 and intimal
model.79 Blockade of another receptor, #IIb!3, appears to expansion has been demonstrated as well.23 Angiotensin ii
prevent platelet aggregation after injury.80 Human clinical may influence intimal growth indirectly through either its
trials using a monoclonal antibody fragment (c7E3 Fab) that interactions with other growth factors (AII stimulates
blocks both the #v!3 and the #IIb!3 receptors as a result of PDGF-A and TGF-! production) or its stimulation of the
their common !3 component decreases the frequency of is- sympathetic nervous system.50 Catecholamines have been
chemic events after PTCA and reduces restenosis by 26%.81 shown to be SMC mitogens in vitro. Angiotensin ii admin-
Whether this is the result of the inhibitory effect on platelet istration produces a vigorous increase in intimal SMC pro-
aggregation or SMC migration or a combination of the two liferation and lesion area, even when infused at a time when
is unknown. the rat carotid lesion is already well developed.23 Infusion of
New proteins or proteins that are normally present in ACE inhibitors and angiotensin receptor blockers inhibit
only small quantities begin to appear in abundance in the intimal thickening after balloon injury in the rat indepen-
matrix of the newly formed intima of the injured rat ca- dent of blood pressure effects.36-38 Cilazapril, an ACE in-
rotid. It is thought that many of these proteins, including hibitor, as well as other ACE inhibitors, and Dup 753, a spe-
osteopontin,82 versican,83 tenascin,84 thrombospondin,85 and cific angiotensin ii receptor antagonist, block intimal thick-
splice products of fibronectin, may play a role in the adhe- ening in rats after balloon injury.93 A human clinical trial
sion of cells in the injured vascular wall. Peptides with the using Cilazipril during coronary angioplasty, however, has
amino acid sequence arginine-glycine-aspartic acid (RGD) shown no effect on either restenosis or angiographic out-
interfere with integrin binding.86 RGD peptides inhibit mi- come.94,95
gration of SMCs in vitro and reduce intima formation in Messenger RNAs for insulin-like growth factor (IGF)-1
the rat.79 Integrins that bind these peptides have been dem- and TGF-! have been detected in the newly formed intima
onstrated in the injured rat carotid and are also abundant in and could play a role in regulating cell proliferation or ex-
normal human and atherosclerotic human arteries.87 tracellular matrix production.96,97
234 Tissue Engineering of Prosthetic Vascular Grafts
After about 3 months, intimal thickening stops. The In areas that remain devoid of endothelium, the lu-
final composition of the intima, once it achieves a resting minal SMCs may assume some endothelial functions, in-
state, is generally 80% ECM by volume.98 What determines cluding the production of inhibitory factors. For example,
the endpoint at which growth ceases? Most probably, the NO is generated via the inducible form of nitric oxide syn-
lesion reaches the point at which either the stimulatory sig- thase (iNOS) and prostacyclin (PGI2) via cyclooxygenase
nals subside or the inhibitory signals develop strongly (COX).117,118 Smooth muscle cell iNOS is upregulated in
enough to counter the stimulatory signals, or both. areas of injury.117 The administration of a NO synthase in-
Evidence exists that endothelial cells play a role in this hibitor (N-omega-nitro-L-arginine methylester (L-NAME))
aspect of lesion growth. In the rat carotid injury model, an accelerates intimal formation after balloon injury, most likely
endothelial surface is formed by the ingrowth of cells from by inhibiting NO production by iNOS since very little cNOS
uninjured areas proximal and distal to the zone of denuda- is present once the endothelium is removed.119
tion. The central portion remains uncovered indefinitely, and Hemodynamic factors also influence lesion formation.
the growing edge appears to stop 1 cm to 1.2 cm from the Increases in blood flow after balloon injury appear to have a
untraumatized zone.99,100 Smooth muscle cell proliferation protective effect. In a rat carotid injury model, intimal hy-
stops sooner (approximately 2 weeks) in areas covered by a perplasia is reduced in rats that undergo contralateral com-
new endothelial layer. After about 4 weeks, SMC growth re- mon carotid artery ligation (high flow) when compared to
turns to baseline even in areas left devoid of endothelium.89 rats that undergo ipsilateral internal carotid artery ligation
SMC proliferation may slow as a result of growth inhibitor (low flow).120 Since SMC proliferation is not different be-
release by the endothelial cells. tween the two groups of animals, this implies an alteration
Cultured endothelial cells are known to secrete hep- in SMC migration as the possible mechanism.
arin-like heparan sulfates.101 The growth inhibitory prop- Could the ultimate control mechanism in lesion de-
erties of this family of molecules are well known.102,103 He- velopment be related to flow (shear stress)? Endothelial cells
parin has also been shown to alter the composition of the serve as the primary sensor for flow related phenomena
ECM.104 Nitric oxide (NO) is another inhibitory factor pro- through shear stress receptors.121 Activation of these path-
duced by endothelial cells. Nitric oxide here is produced via ways may cause changes in the secretion of growth or in-
constitutive nitric oxide synthase or cNOS, the form of NOS hibitory factors. It has been well documented that increas-
unique to endothelial cells.105 In vitro data suggest that NO ing shear stress increases NO production by endothelial cells
is a potent inhibitor of cell proliferation and migration.106,107 in vitro122 and in vivo.123 In areas of endothelial denuda-
In vivo, NO may control lesion development through both tion, SMCs have been shown to undergo phenotypic changes
its vasoactive properties and its inhibitory effects on other to render themselves more endothelial-like.124 As the lesion
growth factor pathways. Since endothelial cells serve as the size increases, luminal area decreases, resulting in increased
major source of NO, loss of endothelium during injury re- shear stress (assuming constant blood flow and no signifi-
sults in the loss of an important SMC growth inhibitor. Once cant change in vessel contraction or dilation). There is some
endothelial regeneration occurs, the pace of intimal hyper- evidence that the vessel itself dilates in an attempt to com-
plasia and SMC proliferation may slow simply by restoring pensate for the loss of luminal area.18 This response is, how-
the inhibitory NO signal. In both a rat and rabbit balloon ever, probably limited. During the early period of luminal
injury model, L-arginine added to the drinking water in- dilation (relatively low shear stress) and loss of endothelium,
creases NO production and reduces intimal hyperpla- NO production is low. Lack of a strong inhibitory signal al-
sia.108,109 In vivo gene transfer with a plasmid containing a lows continued SMC growth. However, as the luminal area
cDNA encoding for cNOS or an adenovirus containing iNOS decreases, the shear stress begins to increase. As the shear
after rat carotid artery injury restores NO to levels seen in stress approaches some critical level and endothelial cells
normal, untreated vessels and inhibits intimal formation by begin to repopulate the area, NO production occurs, inhib-
70%.110,111 Rats kept in an atmosphere of 80 ppm NO after iting further growth of the lesion. The artery wall’s stimula-
balloon injury also develop significantly less intimal hyper- tory and inhibitory factors once again become balanced and
plasia at both 1 and 2 weeks after injury.112 A single, locally the system returns to an equilibrium that more closely re-
delivered treatment of bovine serum albumin modified to sembles the quiescent artery wall.
carry multiple NO groups inhibits intimal hyperplasia after As we know, this is not the end of the story. Many
balloon injury in a rabbit model in a dose dependent man- lesions continue to grow, ultimately causing significant com-
ner.113 Administration of this molecule decreases platelet promise of the lumen. Most probably, these advanced le-
adhesion and SMC migration in association with elevations sions result from a persistent imbalance of the growth and
in platelet and vascular guanosine 3’,5’-cyclic monophos- inhibitory factors in favor of continued lesion growth. As
phate (cGMP) content, the result of guanylate cyclase acti- an example, endothelial regrowth in an injured area might
vation by NO. Aside from its inhibition of SMC prolifera- not be complete, resulting in the absence of inhibitory sig-
tion and migration, NO also affects vascular smooth muscle nals. It should be noted, however, that endothelial cells also
relaxation,114 platelet inhibition,63 leukocyte adhesion,115 have the ability to produce stimulatory growth factors. This
and vascular permeability.116 It is apparent that NO inhibits suggests that their role in lesion formation is variable de-
lesion formation after arterial injury. Whether this is a re- pending upon their local environment. Additionally, the le-
sult of one specific effect or a combination of several is not sion may create focal turbulence in the flowstream, result-
yet known. ing in persistent growth factor stimulation via the previously
Pathobiology of Hyperplastic Intimal Responses 235
mentioned mechanoreceptors. In humans, atherogenic risk Preexisting venous disease might account for some degree
factors also contribute to altered healing after injury. of lesion formation at nontraumatized sites.134,135 At later
As has been noted before, there is an inflammatory times, vein grafts fail due to the development of frank ath-
response after injury. The role of leukocytes in lesion for- erosclerotic lesions. This is particularly prevalent in the
mation is not entirely clear. It is known, however, that T lym- aortocoronary bypass grafts.136 It is noteworthy that when
phocytes are a major component of human atherosclerotic the internal mammary artery, a conduit which requires no
plaque.125 T lymphocytes are seen in the rat carotid artery adaptation to the arterial circulation, is used for coronary
injury model. The most significant evidence that they play a grafting, a much lower failure rate occurs.7 Renewed inter-
role here is the fact that they express &%interferon, a known est also exists in using the radial artery as a coronary artery
inhibitor of SMC growth. Most of the nondividing SMCs bypass conduit in hopes of achieving similar results. Previ-
seen in the rat intima express the class II major histo- ously, its propensity to spasm and the increased likelihood
compatibility antigen Ia, which is known to be induced only of subclinical atheroma discouraged its use. Recent studies
by &%interferon.126 This raises the possibility that other seem to indicate that if appropriate intraoperative steps are
cytokines, including interleukin-1, might be active in taken to inhibit radial artery vasoreactivity, long term pa-
the wall. tency is excellent.137 The improved success achieved with
arterial conduits suggests that the adaptive process that oc-
Atherosclerotic Artery Injury curs in vein grafts might make them more susceptible to
The arteries of cholesterol-fed rabbits and larger spe- lesion formation.
cies develop atherosclerosis. To accelerate the atherosclerotic Regardless of the actual technique employed (in situ
process and generate lesions that resemble advanced human or reversed), vein graft injury undoubtedly occurs during
disease, investigators have combined cholesterol feeding with reconstruction, even in the best of hands. The immediate,
balloon injury. In the rabbit, these lesions cause stenosis and histologic effects of this injury do not differ significantly from
become restenotic after a second injury (Gruntzig that of arterial injury. Several differences have, however, been
angioplasty). Unfortunately, these lesions are distinct from observed despite this similar starting point. Re-
human lesions, typically consisting of a concentric foam cell endothelialization is fast and complete in injured veins.138
infiltration of the intima and media. Additionally, SMC proliferation appears to take place even
Lesions produced in nonhuman primate models more after endothelial coverage is restored. This is in contrast to
closely resemble those of humans.127,128 These animals first the artery, where endothelial coverage appears to inhibit in-
develop a fatty streak with infiltrates of lipid-filled macro- timal SMC proliferation.28 The time course of wall thicken-
phages and later proceed to develop intermediate and ad- ing differs as well. Maximal arterial wall intima formation
vanced lesions similar to those seen in humans. Balloon in- occurs at 2 weeks. Vein graft thickening continues for up to
jury of diseased, but not stenotic, iliac arteries in hyper- 12 weeks.138 What is responsible for these differences?
cholesterolemic monkeys demonstrate a repair response in- One apparent difference is that vein graft endothe-
volving intimal hyperplasia, recoil, and remodelling.129 These lium appears to possess altered physiologic properties from
three mechanisms are felt to form the basis of the restenotic that of the arterial endothelium. Vein graft endothelium has
pathway in humans. While this model is not characterized impaired vasomotor properties139 and increased permeabil-
by a high degree of stenosis or restenosis, the basic mecha- ity.140 In addition, the arterial circulation exposes the thin
nisms resemble the known human mechanisms more closely venous wall to a continuous type I hemodynamic injury.
than those seen in other animal models.12 This model has While not as dramatic as arterial balloon dilation, this in-
been used especially to characterize remodeling, the process jury provides a persistent stimulus for wall thickening. When
by which the arterial lumen size is preserved by vessel dila- the vein dilates immediately upon exposure to the high pres-
tion during atheroma formation. sure arterial circulation, two phenomena occur. First, the
wall of the vein is exposed to a dramatically higher shear
Grafts stress. Shear stress responsive elements present on the en-
dothelial surface are thought to play a role in the activation
Vein of certain stimulatory pathways. The ultimate result is SMC
Autologous vein is the most widely used conduit in proliferation and migration toward the lumen, possibly
both coronary and peripheral bypass surgery. Vein grafts through some of the same mechanisms that have been char-
develop intimal hyperplasia upon exposure to the arterial acterized in the rat injury model. Second, the increased lu-
circulation.130 During reconstruction, vigorous distention minal blood pressure has been shown to invoke medial thick-
of a vein graft already in spasm or passage of a valvulotome ening, primarily through a response to increased wall
damages the endothelium and the graft wall. However, these stress.141,142 It is not yet clear which of these mechanisms is
defects are relatively minor and are repaired within a few primarily responsible for wall thickening in vein grafts. What
days.131 Restenosis due to intimal hyperplasia occurs in about is clear, however, is that hemodynamic factors play an im-
10% of peripheral vein grafts and usually during the time portant role in vein graft patency.14
period 6-24 months after reconstruction. Intimal thicken-
ing is most pronounced at anastomoses, valve cusps, and Prosthetic
clamp injury sites.132 It is also known to occur in vein grafts The prosthetic graft is a man-made, acellular, fibrous
placed using the in situ technique, despite the fact that this scaffolding upon which the body constructs a modified ves-
technique is thought to be less traumatic to the vessel.133 sel wall. The anastomoses appear to be the areas of maximal
236 Tissue Engineering of Prosthetic Vascular Grafts
intimal hyperplasia as a result of surgical trauma and the the # and ! receptor are increased, but not to a significant
presence of flow disturbance. It is thought that human pros- level. We have recently completed a study using monoclonal
thetic grafts heal via two main mechanisms, capillary in- human/mouse chimeric antibodies to both the PDGFR-#
growth through the graft wall, depending upon the charac- and -! receptor in this model. The # receptor antibody had
teristics of the graft, and growth along the luminal surface no effect, possibly because circulating serum concentrations
from each anastomosis.143 New evidence in a canine model did not achieve desirable levels. Adequate levels of the ! re-
supports fallout endothelialization from the blood stream ceptor antibody were achieved and the intimal thickening
as another possible mode of graft healing.144 No current normally observed after “flow switch” was significantly in-
evidence exists that prosthetic grafts inserted into the hu- hibited in this group of animals (unpublished results). This
man circulation are able to develop an endothelialized in- lends further credence to the theory that PDGF-BB and the
tima along the entire flow surface. Rather, endothelial cells ! receptor play a significant role in intimal development. It
manage to populate the lumen in just the few centimeters is unclear, however, why the PDGF-B ligand has not been
near the anastomoses.145 The remainder of the graft is lined detectable at higher levels in the developing intimal lesion.
by thrombus containing fibrin, platelets, and leukocytes. One possible explanation is that it is being produced
In our experience with prosthetic PTFE grafts placed primarily by the endothelial cells in small but sufficient
in baboons, the development of an endothelial layer is de- quantities and serves mainly as a powerful chemoattractant
pendent upon porosity.146 In low porosity grafts (10 and to attract SMCs towards the lumen. While other factors may
30 ∝m internodal distance), luminal endothelial coverage is participate in the development of intimal hyperplasia in
limited to small areas near the anastomoses. In high poros- PTFE grafts, we feel that the flow-related effects and the
ity grafts, (60 and 90 ∝m), luminal endothelial coverage is resultant production of PDGF and NO play significant roles.
complete. The 90 ∝m grafts, for unknown reasons, develop Further studies are needed to better characterize
areas of endothelial loss. The 60 ∝m grafts perform opti- this relationship.
mally regarding endothelial coverage and intimal layer sta-
bility. Similar results have been obtained in dogs.147 Summary
Once an endothelial layer is present, intimal thicken- Vascular injury can induce intimal hyperplasia. The
ing forms. It is perplexing that in the rat carotid injury model, ultimate response depends upon the severity of the injury
the endothelial layer serves to inhibit intimal thickening, and the change in the overall balance of stimulatory and
while in the prosthetic grafts, intimal thickening occurs only inhibitory factors at the site of injury. Based upon animal
once endothelium is present. This observation appears to models, the essential components of this exuberant healing
confirm the variable role of endothelial cells in the develop- response are SMC proliferation, SMC migration, and extra-
ment of intimal hyperplasia. cellular matrix deposition. SMC proliferation after injury is
Our laboratory has extensively characterized the heal- predominantly stimulated by bFGF released mostly from
ing of prosthetic grafts in baboons. Since there is no signifi- damaged cells but also from the vessel wall matrix. PDGF-BB,
cant thrombotic response, it is unlikely that the growth fac- deposited at the site of injury by platelets and released by
tors responsible for intimal formation are derived from plate- cells in the vessel wall, serves as the primary stimulus for
lets. Endothelial cells, however, can produce growth factors, SMC migration. Release of SMCs from their surrounding
among them, PDGF. Golden observed that healing aortoiliac matrix is accomplished by plasmin and MMPs. Cellular ad-
PTFE grafts removed after 2, 8, and 12 weeks and perfused hesion receptors or integrins are also involved in this pro-
ex vivo released more mitogenic factors than control cess. Control of matrix production is not yet as well defined.
arteries.148 This activity was blocked by an antibody to PDGF. In addition to causing the release and production of stimu-
Furthermore, PDGF-A mRNA and protein levels were latory factors, vascular injury also transiently reduces the
detectable in the graft intima by both Northern analysis and availability of important inhibitory factors such as NO. In-
in situ hybridization. SMC proliferation was seen by timal hyperplasia occurring in atherosclerotic arteries, vein
thymidine labeling in the same region (inner third of the grafts, and prosthetic grafts is not as well understood as in
intima) as PDGF mRNA expression. PDGF-B was the rat carotid artery injury model, although similarities in
nearly undetectable.149 the basic pathophysiology most likely exist. The challenge
As discussed earlier, flow-related effects are seen in vein for the future is to further define the human response in a
graft healing. Studies have also demonstrated flow-depen- systematic manner and to successfully apply this knowledge
dent formation of intima in PTFE grafts in baboons.150 towards improving the effectiveness of our vascular inter-
Grafts placed in the aorto-iliac position under high flow ventions.
conditions created by placement of a distal fistula form
smaller intimal layers than those that heal under normal flow. References
If normal flow is returned to the graft previously allowed to 1. Topol EJ, Leya F, Pinkerton CA et al. A comparison of
heal during high flow, the intima undergoes a vigorous thick- directional atherectomy with coronary angioplasty in pa-
ening. This “flow switch” model has been used in an attempt tients with coronary artery disease. New Engl J Med 1993;
329:221-27.
to identify the stimuli responsible for intimal thickening in
2. Mills JL, Fujitani RM, Taylor SM. The characteristics and
the PTFE graft in primates. anatomic distribution of lesions that cause reversed vein
Four days after flow switch, PDGF-A mRNA and pro- graft failure: A five year prospective study. J Vasc Surg
tein levels are significantly increased.149 Levels of PDGF-B 1993; 17:195-204.
ligand do not appear to change, while mRNA levels for both
Pathobiology of Hyperplastic Intimal Responses 237
3. Lytle BW, Loop FD, Cosgrove DM et al. Long-term (5 to Results of the MARCATOR study: A multicenter, random-
12 years) serial studies of internal mammary artery and ized, double-blind placebo-controlled trial. Circulation
saphenous vein coronary bypass grafts. J Thorac 1992; 86:100-10.
Cardiovasc Surg 1985; 89:248-258. 22. Kenagy RD, Vergel S, Mattsson E et al. The role of plas-
4. Kraiss LW, Clowes AW. Response of the arterial wall to minogen, plasminogen activators, and matrix
injury and intimal hyperplasia. In: Sidawy AN, Sumpio metalloproteinases in primate arterial smooth muscle cell
BE, DePalma RG, eds. The Basic Science of Vascular Dis- migration. Arterioscler Thromb Vasc Biol 1996;
ease. Armonk, NY: Futura Publishing Company, 16:1373-82.
Inc.;1997:289-317. 23. Schwartz SM, deBlois D, O’Brien ERM. The intima: Soil
5. Holmes DR, Vliestra RE, Smith HC et al. Restenosis after for atherosclerosis and restenosis. Circ Res 1995;
percutaneous transluminal coronary angioplasty (PTCA): 77:445-465.
A report from the PTCA Registry of the National Heart, 24. Fingerle J, Au YPT, Clowes et al. Author: Please provide
Lung, and Blood Institute. Am J Cardiol 1984; 53:77C. initials for author Clowes. Intimal lesion formation in rat
6. Fanelli C, Aronoff R. Restenosis following coronary carotid arteries after endothelial denudation in absence
angioplasty. Am Heart J 1990; 119:357-68. of medial injury. Arteriosclerosis 1990; 1082-87.
7. Grondin CM, Campeau L, Lesperance J et al. Compari- 25. Sarembock IJ, LaVeau PJ, Sigal SL et al. Influence of in-
son of late changes in internal mammary artery and saphe- flation pressure and balloon size on the development of
nous vein grafts in two consecutive series of patients 10 intimal hyperplasia after balloon angioplasty: A study in
years after operation. Circulation 1984; 70:1208-12. the atherosclerotic rabbit. Circulation 1989; 80:1029-40.
8. Campeau L, Enjalbert M, Lesperance J et al. The relation 26. Schwartz RS, Huber KC, Murphy JG et al. Restenosis and
of risk factors to the development of atherosclerosis in the proportional neointimal response to coronary artery
saphenous-vein bypass grafts and the progression of dis- injury: Results in a porcine model. J Am Coll Cardiol
ease in the native circulation. N Engl J Med 1984; 1992; 19:267-74.
311:1329-32. 27. Clowes AW, Clowes MM, Fingerle J et al. Regulation of
9. Veith FJ, Gupta SK, Ascer E et al. Six-year prospective smooth muscle cell growth in injured artery. J Cardiovasc
multicenter randomized comparison of autologous saphe- Pharmacol 1989; 14:S12-15.
nous vein and expanded polytetrafluoroethylene grafts in 28. Clowes AW, Reidy MA, Clowes MM. Kinetics of cellular
infrainguinal arterial reconstructions. J Vasc Surg 1986; proliferation after arterial injury. I. Smooth muscle growth
3:104-114. in the absence of endothelium. Lab Invest 1983; 49:327-33.
10. Tullis MJ, Zierler RE, Glickerman DJ et al. Results of per- 29. Majesky MW, Schwartz SM, Clowes AW et al. Heparin
cutaneous transluminal angioplasty for atherosclerotic re- regulates smooth muscle S phase entry in the injured rat
nal artery stenosis: A follow-up study with duplex carotid artery. Circ Res 1987; 61:296-300.
ultrasonography. J Vasc Surg 1977; 25:46-54. 30. Clowes AW, Schwartz SM. Significance of quiescent
11. Healy DA, Clowes AW, Zierler RE et al. Immediate and smooth muscle migration in the injured rat carotid ar-
long-term results of carotid endarterectomy. Stroke 1989; tery. Circ Res 1985; 56:139-145.
20:1138-1142. 31. Ross R, Glomset JA. The pathogenesis of atherosclerosis.
12. Clagett GP. Morphogenesis and clinicopathologic charac- N Engl J Med 1976; 295:369-77.
teristics of recurrent carotid disease. J Vasc Surg 1986; 32. Ross R, Glomset JA. The pathogenesis of atherosclerosis.
3:10-23. N Engl J Med 1976; 295:420-25.
13. Das MB, Hertzer NR, Ratliff NB et al. Recurrent carotid 33. Friedman RJ, Stemerman MB, Wenz B et al. The effect
stenosis: A five-year series of 65 reoperations. Ann Surg of thrombocytopenia on experimental atherosclerotic le-
1985; 202:28-35. sion formation in rabbits. Smooth muscle cell prolifera-
14. Davies MG, Hagen P-O. Pathophysiology of vein graft tion and re-endothelialization. J Clin Invest 1977;
failure: A review. Eur J Vasc Endovasc Surg 1995; 9:7-18. 60:1191-201.
15. Becker GJ. Intravascular stents. General principles and 34. Ross R, Glomset J, Kariya B et al. A platelet-dependent
status of lower-extremity arterial applications. Circulation serum factor that stimulates the proliferation of arterial
1991; 83:I122-36. smooth muscle cells in vitro. Proc Natl Acad Sci USA
16. Mintz GS, Popma JJ, Hong MK et al. Intravascular ultra- 1974; 71:1207-10.
sound to discern device-specific effects and mechanisms 35. Fingerle J, Johnson R, Clowes AW et al. Role of platelets
of restenosis. Am J Cardiol 1996; 78:18-22. in smooth muscle cell migration and proliferation after
17. Bond MG, Adams MR, Bullock BC. Complicating factors vascular injury in rat carotid artery. Proc Natl Acad Sci
in evaluating coronary artery atherosclerotic coronary ar- USA 1989; 86:8412-16.
teries. Artery 1981; 9:21-29. 36. Lindner V, Lappi DA, Baird A et al. Role of basic fibro-
18. Glagov S, Weisenberg E, Zarins CK et al. Compensatory blast growth factor in vascular lesion formation. Circ Res
enlargement of human atherosclerotic coronary arteries. 1991; 68:106-13.
N Engl J Med 1987; 316:1371-75. 37. Lindner V, Reidy MA. Proliferation of smooth muscle cells
19. Kakuta T, Currier JW, Haudenschild CC et al. Differences after vascular injury is inhibited by an antibody against
in compensatory vessel enlargement, not intimal forma- basic fibroblast growth factor. Proc Natl Acad Sci USA
tion, account for restenosis after angioplasty in the hy- 1991; 88:3739-43.
percholesterolemic rabbit model. Circulation 1994; 38. Lindner V, Reidy MA. Expression of basic fibroblast
89:2809-15. growth factor and its receptor by smooth muscle cells and
20. Popma JJ, Califf RM, Topol EJ. Clinical trials of restenosis endothelium in injured rat arteries: An en face study. Circ
after coronary angioplasty. Circulation 1991; 84:1426-36. Res 1993; 73:589-95.
21. MARCATOR Study Group. Does the new angiotensin 39. D’Amore PA. Modes of FGF release in vivo and in vitro.
converting enzyme inhibitor cilazapril prevent restenosis Canc Metastasis Rev 1990; 9:227-38.
after percutaneous transluminal coronary angioplasty?
238 Tissue Engineering of Prosthetic Vascular Grafts
40. Mignatti P, Morimoto T, Rifkin DB. Basic fibroblast 58. Jackson CL, Schwartz SM. Pharmacology of smooth
growth factor, a protein devoid of secretory signal se- muscle cell replication. Hypertension 1992; 20:713-36.
quence, is released by cells via a pathway independent of 59. Thyberg J, Blomgren K, Hedin U et al. Phenotypic modu-
the endoplasmic reticulum-Golgi complex. J Cell Physiol lation of smooth muscle cells during the formation of
1992; 151:81-93. neointimal thickenings in the rat carotid artery after bal-
41. Thompson RW, Whalen GF, Saunders KB et al. Heparin- loon injury: An electronic-microscopic and stereological
mediated release of fibroblast growth factor-like activity study. Cell Tissue Res 1995; 281:421-428.
into the circulation of rabbits. Growth Factors 1990; 60. Raines EW, Bowen-Pope DF, Ross R. Platelet-derived
3:221-29. growth factor. In: Sporn MB, Roberts AB, eds. Handbook
42. Lindner V, Olson NE, Clowes AW et al. Inhibition of of Experimental Pharmacology: Peptide Growth Factors.
smooth muscle cell proliferation in injured rat arteries. New York: Springer-Verlag, Inc; 1990:173-189.
Interaction of heparin with basic fibroblast growth fac- 61. Koyama N, Hart CE, Clowes AW. Different functions of
tor. J Clin Invest 1992; 90:2044-49. the platelet-derived growth factor-# and -! receptors for
43. Lindner V, Majack RA, Reidy MA. Basic fibroblast growth the migration and proliferation of cultured baboon
factor stimulates endothelial regrowth and proliferation smooth muscle cells. Circ Res 1994; 75:682-91.
in denuded arteries. J Clin Invest 1990; 85; 2004-8. 62. Hart CE, Clowes AW. Platelet-derived growth factor and
44. Campbell DJ. Circulating and tissue angiotensin systems. arterial response to injury. Circulation 1997; 95:555-56.
J Clin Invest 1987; 79:1-6. 63. Ferns GAA, Raines EW, Sprugel KH. Inhibition of
45. Campbell-Boswell M, Robertson AL Jr. Effects of angio- neointimal smooth muscle accumulation after angioplasty
tensin ii and vasopressin on human smooth muscle cells by an antibody to PDGF. Science 1991; 253:1129-1132.
in vitro. Exp Mol Pathol 1981; 35:265-76. 64. Jawien A, Bowen-Pope DF, Lindner V et al. Platelet-de-
46. Battegay EJ, Raines EW, Seifert RA et al. TGF-! induces rived growth factor promotes smooth muscle migration
bimodal proliferation of connective tissue cells via com- and intimal thickening in a rat model of balloon
plex control of an autocrine PDGF loop. Cell 1990; angioplasty. J Clin Invest 1992; 89:507-11.
63:515-24. 65. Sirois MG, Simons M, Edelman ER. Antisense oligonucle-
47. Naftilan AJ, Pratt RE, Dzau VJ. Induction of platelet-de- otide inhibition of PDGF-! receptor subunit expression
rived growth factor A-chain and c-myc gene expression directs suppression of intimal thickening. Circulation
by angiotensin ii in cultured rat vascular smooth muscle 1997; 95:669-676.
cells. J Clin Invest 1989; 83:1419-24. 66. Murry CE, Bartosek T, Giachelli CM et al. Platelet-de-
48. Naftilan AJ, Pratt RE, Eldridge CS et al. Angiotensin ii rived growth factor-A mRNA expression in fetal, normal
induces c-fos expression in smooth muscle via transcrip- adult, and atherosclerotic human aortas. Circulation 1996;
tional control. Hypertension 1989; 13:706-11. 93:1095-1106.
49. Prescott MF, Webb RL, Reidy MA. Angiotensin-convert- 67. Barres BA, Hart IK, Coles HSR. Cell death in the oligo-
ing enzyme inhibitor versus angiotensin ii, AT1 receptor dendrocyte lineage. J Neurobiol 1992; 23:1221-1230.
antagonist: Effects on smooth muscle cell migration and 68. Bennett MR, Evan GI, Schwartz SM. Apoptosis of human
proliferation after balloon catheter injury. Am J Pathol vascular smooth muscle cells derived from normal ves-
1991; 139:1291-96. sels and coronary atherosclerotic plaques. J Clin Invest
50. Daemen MJ, Lombardi DM, Bosman FT et al. Angiotensin 1995; 95:2266-74.
ii induces smooth muscle cell proliferation in the normal 69. Harrington EA, Bennett MR, Fanidi A. c-myc-induced
and injured rat arterial wall. Circ Res 1991; 68:450-56. apoptosis in fibroblasts is inhibited by specific cytokines.
51. Laporte S, Escher E. Neointima formation after vascular EMBO J 1994; 13:3286-95.
injury is angiotensin ii mediated. Biochem Biophys Res 70. Wakefield TW. Coagulation and disorders of hemostasis.
Comm 1992; 187:1510-16. In: Sidawy AN, Sumpio BE, DePalma RG, eds. The Basic
52. Majesky MW, Reidy MA, Bowen-Pope DF et al. PDGF Science of Vascular Disease. Armonk, NY Futura Publish-
ligand and receptor gene expression during repair of ar- ing Company, Inc.;1997:289-317.
terial injury. J Cell Biol 1990; 111:2149-58. 71. Clowes AW, Clowes MM, Au YPT et al. Smooth muscle
53. Sarembock IJ, Gertz SD, Gimple LW et al. Effectiveness cells express urokinase during mitogenesis and tissue-type
of recombinant desulphatohirudin in reducing restenosis plasminogen activator during migration in injured rat ca-
after balloon angioplasty of atherosclerotic femoral arter- rotid artery. Circ Res 1990; 67:61-7.
ies in rabbits. Circulation 1991; 84:232-43. 72. Zempo N, Kenagy RD, Au YPT et al. Matrix
54. Douglas SA, Louden C, Vickery-Clark LM et al. A role metalloproteinases of vascular wall cells are increased in
for endogenous endothelin-1 in neointimal formation af- balloon-injured rat carotid artery. J Vasc Surg 1994;
ter rat carotid artery balloon angioplasty. Protective ef- 20:209-17.
fects of the novel nonpeptide endothelin receptor antago- 73. Reidy MA, Irvin C, Lindner V. Migration of arterial wall
nist SB 209670. Circ Res 1994; 75:190-97. cells. Expression of plasminogen activators and inhibitors
55. Trachtenberg JD, Sun S, Choi ET et al. Effect of in injured rat arteries. Circ Res 1996; 78:405-14.
endothelin-1 infusion on the development of intimal hy- 74. Bendeck MP, Zempo N, Clowes AW et al. Smooth muscle
perplasia after balloon catheter injury. J Cardiovasc cell migration and matrix metalloproteinase expression
Pharmacol 1993; 22:S355-39. after arterial injury in the rat. Circ Res 1994; 75:539-45.
56. Takuwa Y, Yanagisawa M, Takuwa N et al. Endothelin, 75. Carmeliet P, Stassen JM, De Mol M et al. Arterial
its diverse biological activities and mechanisms of action. neointima formation after trauma in mice with inactiva-
Prog Growth Factor Res 1989; 1:195-206. tion of the tPA, uPA, or PAI-1 genes. Fibrinolysis 1994;
57. Bornfeldt KE, Arnqvist HJ, Capron L. In vivo prolifera- 8:280a.
tion of rat vascular smooth muscle in relation to diabetes 76. Matrisian LM. Metalloproteinases and their inhibitor in
mellitus insulin-like growth factor I and insulin. matrix remodeling. Trends in Genet 1990; 6(4):121-25.
Diabetologia 1992; 35:104-8.
Pathobiology of Hyperplastic Intimal Responses 239
77. HE CS, Wilhelm SM, Pentland AP. Tissue cooperation in 95. Heyndricks GR, MARCATOR. Angiotensin-converting
a proteolytic cascade activating human interstitial colla- enzyme inhibitor in a human model of restenosis. Basic
genase. Proc Natl Acad Sci USA 1989; 86:2632-36. Res Cardiol 1993; 88:169-82.
78. Forough R, Koyama N, Hasentstab D et al. Overexpression 96. Cercek B, Fishbein MC, Forrester JS et al. Induction of
of tissue inhibitor of matrix metalloproteinase-1 inhibits insulin-like growth factor-! messenger RNA in rat aorta
vascular smooth muscle cell functions in vitro and in vivo. after balloon denudation. Circ Res 1990; 66:1755-60.
Circ Res 1996; 79:812-20. 97. Majesky MW, Lindner V, Twardzik DR et al. Angiotensin
79. Choi ET, Engel L, Callow AD et al. Inhibition of ii induces smooth muscle cell proliferation in the normal
neointimal hyperplasia by blocking #v!3 integrin with a and injured rat arterial wall. Circ Res 1991; 88:904-10.
small peptide antagonist GpenGRGDSPCA. J Vasc Surg 98. Clowes AW, Reidy MA, Clowes MM. Mechanisms of
1994; 19:125-34. stenosis after arterial injury. Lab Invest 1983; 49:208-15.
80. Matsuno H, Hoylaerts MF, Vermylen J et al. Inhibition 99. Reidy MA. A reassessment of endothelial injury and arte-
of integrin function prevents restenosis following vascu- rial lesion formation. Lab Invest 1985; 53:513-20.
lar injury. Nippon Yakurigaku Zasshi 1995; 106:143-55. 100. Schwartz SM, Haudenschild CC, Eddy EM. Endothelial
81. The EPIC Investigation. Use of a monoclonal antibody regeneration. I. Quantitative analysis of endothelial regen-
directed against the platelet glycoprotein IIb/IIIa receptor eration in rat aortic intima. Lab Invest 1978; 38:568-80.
in high-risk coronary angioplasty. New Engl J Med 1994; 101. Castellot JJ Jr, Addonizio ML, Rosenberg R et al. Cul-
330:956-61. tured endothelial cells produce a heparin-like inhibitor of
82. Giachelli CM, Liaw L, Murry CE et al. Osteopontin ex- smooth muscle cell growth. J Cell Biol 1981; 90:372-79.
pression in cardiovascular diseases. Ann N Y Acad Sci 102. Clowes AW, Karnovsky NJ. Suppression by heparin of
1995; 760:109-26. smooth muscle cell proliferation in injured arteries. Na-
83. Wolf YG, Rasmussen LM, Ruoslahti E. Antibodies against ture 1977; 265:625-26.
transformtin growth factor-!-1 suppress intimal hyperpla- 103. Clowes AW, Clowes MM. Kinetics of cellular prolifera-
sia in a rat model. J Clin Invest 1994; 93:1172-78. tion after arterial injury. IV. Heparin inhibits rat smooth
84. Hedin U, Holm J, Hansson GK. Induction of tenascin in muscle mitogenesis and migration. Circ Res 1986;
rat arterial injury; relationship to altered smooth 58:839-45.
muscle cell phenotype. Am J Pathol 1991; 139:649-56. 104 Au YP, Montgomery KF, Clowes AW. Heparin inhibits
85. Majesky MW. Neointima formation after acute vascular collagenase gene expression mediated by phorbol ester-
injury. Role of counteradhesive extracellular matrix pro- responsive element in primate arterial smooth muscle
teins. Texas Heart Inst J 1994; 21:78-85. cells. Circ Res 1992; 70:1062-69.
86. Hynes RO. Integrins: Versatility, modulation, and sig- 105. Lloyd-Jones DM, Bloch KD. The vascular biology of ni-
naling in cell adhesion. Cell 1992; 69:11-25. tric oxide and its role in atherogenesis. Annu Rev Med
87. Hoshiga M, Alpers CE, Smith LL et al. #v!3 integrin ex- 1996; 47:365-75.
pression in normal and atherosclerotic artery. Circ Res 106. Garg UC, Hassid A. Nitric oxide-generating vasodilators
1995; 77:1129-35. and 8-bromo-cyclic guanosine monophosphate inhibit
88. Garratt KN, Edwards WD, Kaufmann UP et al. Differen- mitogenesis and proliferation of cultured rat vascular
tial histopathology of primary atherosclerotic and smooth muscle cells. J Clin Invest;38:1774-77.
restenotic lesions in coronary arteries and saphenous vein 107. Sarkar R, Meinberg EG, Stanley JC et al. Nitric oxide in-
bypass grafts: Analysis of tissue obtained from 73 patients hibits the migration of cultured vascular smooth muscle
by directional atherectomy. J Am Coll Cardiol 1991; cells. Circ Res 1996; 78:225-30.
17:442-48. 108. McNamara DB, Bedi B, Aurora H et al. L-arginine inhib-
89. Clowes AW. Pathologic intimal hyperplasia as a response its balloon catheter-induced intimal hyperplasia. Biochem
to vascular injury and reconstruction. In: Rutherford RB, Biophys Res Commun 1993; 193:291-96.
ed. Vascular Surgery. 4th Edition. Philadelphia: W.B. 109. Taguchi J, Junichi A, Takuwa Y et al. L-arginine inhibits
Saunders, 1995:285-95. neointimal formation following balloon injury. Life Sci
90. Raines EW, Dower SK, Ross R. Interleukin-1 mitogenic 1993; 53:387-92.
activity for fibroblasts and smooth muscle cells is due to 110. von der Leyen HE, Gibbons GH, Morishita R et al. Gene
PDGF-AA. Science 1989; 243:393-96. therapy inhibiting neointimal vascular lesion: In vivo
91. Olson NE, Chao S, Lindner V et al. Intimal smooth transfer of endothelial cell nitric oxide synthase gene. Proc
muscle cell proliferation after balloon catheter injury: Natl Acad Sci USA 1995; 92:1137-1141.
The role of basic fibroblast growth factor. Am J Pathol 111. Tzeng E, Shears LL II, Robbins PD et al. Vascular gene
1992; 140:1017-23. transfer of the human inducible nitric oxide synthase:
92. Lindner V, Reidy MA. Platelet-derived growth factor Characterization of activity and effects on myointimal
ligand and receptor expression by large vessel endothe- hyperplasia. Mol Med 1996; 2:211-25.
lium in vivo. Am J Pathol 1995; 146:1488-97. 112. Lee JS, Adrie C, Jacob HJ et al. Chronic inhalation of ni-
93. Powell, JS, Clozel JP, Muller RKM et al. Inhibitors of tric oxide inhibits neointimal formation after balloon-in-
angiotensin-converting enzyme prevent myointimal pro- duced arterial injury. Circ Res 1996; 78:337-342.
liferation after vascular injury. Science 1989; 245:186-88. 113. Marks DS, Vita JA, Folts JD et al. Inhibition of neointimal
94. Faxon DP. Effect of high dose angiotensin-converting proliferation in rabbits after vascular injury by a single
enzyme inhibition on restenosis: Final results of the treatment with a protein adduct of nitric oxide. J Clin
MARCATOR Study, a multicenter, double-blind, placebo- Invest 1995; 96:2630-38.
controlled trial of cilazapril. The Multicenter American 114. Myers PR, Minor Jr RL, Guerra R et al. Vasorelaxant
Research Trial With Cilazapril After Angioplasty to Pre- properties of the endothelium-derived relaxing factor
vent Transluminal Coronary Obstruction and Restenosis more closely resembles S-nitrosocysteine than nitric ox-
(MARCATOR) Study Group. J Am Coll Cardiol 1995; ide. Nature 1990; 345:161-163.
25:362-69.
240 Tissue Engineering of Prosthetic Vascular Grafts
115. Kubes P, Suzuki M, Granger DN. Nitric oxide: and en- 130. LoGerfo FW, Quist WC, Cantelmo NL et al. Integrity of
dogenous modulator of leukocyte adhesion. Proc Natl vein grafts as a function of initial intimal and medial pres-
Acad Sci USA 1991; 88:4651-4655. ervation. Circulation 1983; 68:II117-24.
116. Kubes P, Granger DN. Nitric oxide modulates microvas- 131. Fuchs JC, Mitchener JS III, Hagen P-O. Postoperative
cular permeability. Am J Physiol 1993; 262:H611-H615. changes in autologous vein grafts. Ann Surg 1978;
117. Hansson GK, Geng Y, Holm J. Arterial smooth muscle 188:1-15.
cells express nitric oxide synthase in response to endo- 132. Bandyk DF, Kaebnick HW, Stewart GW et al. Durability
thelial injury. J Exp Med 1994; 180:733-38. of the in situ saphenous vein arterial bypass. A compari-
117. Pritchard KA Jr, O’Banion MK, Miano JM et al. Induc- son of primary and secondary patency. J Vasc Surg 1987;
tion of cyclooxygenase-2 in rat vascular smooth muscle 5:256-68.
cells in vitro and in vivo. J Biol Chem 1994; 269:8504-9. 133. Panetta TF, Marin ML, Veith FJ et al. Unsuspected pre-
118. Cayatte AJ, Palocino JJ, Horten K et al. Chronic inhibi- existing saphenous vein disease: An unrecognized cause
tion of nitric oxide production accelerates neointimal for- of vein bypass failure. J Vasc Surg 1992; 15:102-12.
mation and impairs endothelial-type function in hyper- 134. Varty K, Allen KE, Bell PRF et al. Infrainguinal vein graft
cholesterolemic rabbits. Arterioscler Thromb 1994; stenosis. Br J Surg 1993; 80:825-33.
14:753-59. 135. Spray TL, Roberts WC. Changes in saphenous used as
119. Kohler TR, Jawien A. Flow affects development of inti- aorto-coronary bypass grafts. Am Heart J; 1977; 94:500-16.
mal hyperplasia after arterial injury in rats. Arterioscler 136. Buxton B, Fuller J, Gaer J et al. Curr Opin Cardiol 1996;
Thromb 1992; 12:963-71. 11:591-98.
120. Berk BC, Corson MA, Peterson TE et al. Protein kinases 137. Kohler TR, Kirkman TR, Clowes AW. Effect of heparin
as mediators of fluid shear stress stimulated signal trans- on adaptation of vein grafts to arterial circulation. Arte-
duction in endothelial cells: A hypothesis for cal- riosclerosis 1989; 9:523-28.
cium-dependent and calcium-independent events activated 138. Luscher TF. Vascular biology of coronary bypass grafts.
by flow. J Biomechanics 1995; 28:1439-50. Coronary Artery Disease 1992; 3:157-65.
121. Corson MA, James NL, Latta SE et al. Phosphorylation 139. Cox JL, Chaisson DA, Gotlieb AI. Stranger in a strange
of endothelial nitric oxide synthase in response to fluid land: The pathogenesis of saphenous vein graft stenosis
shear stress. Circ Res 1996; 79:984-91. with emphasis on structural and functional differences
122. Nadaud S, Philippe M, Arnal JF et al. Sustained increase between veins and arteries. Prog Cardiovasc Dis 1991;
in aortic endothelial nitric oxide synthase expression in 34:45-68.
vivo in a model of chronic high blood flow. Circ Res 1996; 140. Dobrin PB, Littooy FN, Golan J et al. Mechanical and
79:857-63. histologic changes in canine vein grafts. J Surg Res 1988;
123. Davies PF, Remuzzi A, Gordon EJ et al. Turbulent fluid 14:259-60.
shear stress induces vascular endothelial turnover in vitro. 141. Dobrin PB, Litooy FN, Endean ED. Mechanical factors
Proc Natl Acad Sci USA 1986; 83:2114-17. predisposing to intimal hyperplasia and medial thicken-
124. Libby P, Hansson GK. Involvement of the immune sys- ing in autogenous vein grafts. Surgery 1989; 105:393-400.
tem in human atherogenesis: Current knowledge and 142. Clowes AW, Kirkman TR, Reidy MA. Mechanisms of ar-
unanswered questions. Lab Invest 1991; 64:5-15. terial graft healing. Rapid transmural capillary ingrowth
125. Hansson GK, Jonasson L, Holm J et al. Gamma inter- provides a source of intimal endothelium and smooth
feron regulates vascular smooth muscle proliferation and muscle in porous PTFE prostheses. Am J Pathol 1986;
Ia expression in vitro and in vivo. Circ Res 1988 63; 123:220-30.
712-19. 143. Shi Q, Wu MH, Hayashida N et al. Proof of fallout
126. Clarkson TB, Bond MG, Bullock BC et al. A study of ath- endothelialization of impervious Dacron grafts in the aorta
erosclerosis regression in Macaca mulatta, V: and inferior vena cava of the dog. J Vasc Surg 1994;
Changes in abdominal aorta and carotid and coronary 20:546-56.
arteries from animals with atherosclerosis induced for 38 144. Clowes AW, Kohler T. Graft endothelialization: The role
months and then regressed for 24 or 48 months at plasma of angiogenic mechanisms. J Vasc Surg 1991; 13:734-36.
cholesterol concentrations of 300 or 200 mg/dl. Exp Mol 145. Golden MA, Hanson SR, Kirkman TR et al. Healing of
Pathol 1984; 41:96-118. polytetrafluoroethylene arterial grafts is influenced by graft
127. Faggiotto A, Ross R, Harker L. Studies of hyper- porosity. J Vasc Surg 1990; 11:838-45.
cholesterolemia in the nonhuman primate. I. Changes that 146. Tsuchida H, Kashyap A, Cameron BL et al. In vivo study
lead to fatty streak formation. Arteriosclerosis 1984; of a high-porosity polytetrafluoroethylene graft:
4:323-340. Endothelialization, fluid leakage, and the effect of fibrin
128. Geary RL, Williams JK, Golden D et al. Time course of glue sealing. J Invest Surg 1993; 6:509-18.
cellular proliferation, intimal hyperplasia, and remodel- 147. Golden MA, Au YPT, Kenagy RD et al. Growth factor
ing following angioplasty in monkeys with established ath- gene expression by intimal cells in healing
erosclerosis. A nonhuman primate model of restenosis. polytetrafluoroethylene grafts. J Vasc Surg 1990; 11:580-85.
Arterioscl Thromb Vasc Biol 1996; 16:34-43. 148. Kraiss LW, Geary RL, Mattsson EJR et al. Acute reduc-
129. Zwolak RM, Adams MC, Clowes AW. Kinetics of vein tions in blood flow and shear stress induce platelet-de-
graft hyperplasia: Association with tangential stress. J Vasc rived growth factor-A expression in baboon prosthetic
Surg 1987; 5:126-36. grafts. Circ Res 1996; 79:45-53.
149. Kohler TR, Kirkman TR, Kraiss LW et al. Increased blood
flow inhibits neointimal hyperplasia in endothelialized
vascular grafts. Circ Res 1991; 69:1557-65.
Taming of Adverse Responses
Prevention of a Hyperplastic Intimal Response
CHAPTER 23
Cell Cycle Interruption to Inhibit Intimal Hyperplasia
Michael J. Mann, Ruediger C. Braun-Dullaeus, Victor J. Dzau
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
242 Tissue Engineering of Prosthetic Vascular Grafts
autologous vein grafts.8,10-12 We have therefore developed liferation in vitro, including basic fibroblast growth factor
strategies designed to alter patterns of vascular cell gene ex- (bFGF) and platelet derived growth factor (PDGF),18,19 and
pression associated with the onset of this abnormal accu- these factors have been identified in the vessel wall in vivo
mulation of intimal cells. We initially hypothesized that vein in arterial and venous models of neointimal hyperplasia, as
graft failures linked to neointima formation and/or acceler- well.20,21 Increased platelet adherence and degranulation and
ated atherosclerosis were the downstream result of pheno- release from injured vascular cells were initially thought to
typic changes in vascular cells. We subsequently observed be the primary sources of PDGF and bFGF, respectively, al-
that genetic blockade of cell cycle progression during a criti- though it is now recognized that vascular cells themselves
cal phase of vein graft response to the surgical, hemodynamic initiate the autocrine and paracrine production of these fac-
and inflammatory insults of grafting results in a shift of graft tors after arterial injury and vein grafting.18,19
biology toward a more adaptive remodeling process charac- Proliferation among medial VSMC has been docu-
terized by medial hypertrophy and stabilization of endo- mented within 24-48 hours after balloon injury, and migra-
thelial function. tion of VSMC across the internal elastic lamina is established
by days 3 and 4.17,22 Early neointimal cells have demonstrated
The Molecular and Cellular Biology rates of bromo-deoxyuridine incorporation, indicative of
of Neointimal Vascular Graft Disease DNA synthesis, as high as 50%, and this rapid proliferation
Despite progress in the development of artificial vas- establishes approximately 90% of the neointimal cell mass
cular prostheses, autologous vein grafts continue to have within two weeks of injury.22 Cell cycle progression and cell
higher long term patency rates of approximately 70% and division subsequently slows, while further increases in
50% at three and five years, respectively.1,2 Vein graft occlu- neointimal thickness result primarily from continued ex-
sions, however, have been linked to neointimal hyperplasia, tracellular matrix production.23 In addition to PDGF and
and “activated” VSMC are known to express an array of pro- bFGF, other factors, such as transformtin growth factor-!
teins that can contribute to a pro-atherogenic environment (TGF-!), angiotensin II and insulin-like growth factor-1
in the graft wall. These upregulated molecules include (IGF-1), are also expressed in the vessel wall after injury, and
cytokines such as interleukins -1, -2 and -6, and tumor ne- may play important roles in determining the extent of le-
crosis factor (TNF), adhesion molecules such as intercellu- sion formation.24-26
lar adhesion molecule-1 (ICAM-1) and chemoattractants A dramatic increase in VSMC DNA synthesis has also
such as monocyte chemoattractant protein-1 (MCP-1).12-15 been documented during the first week after operation in
In addition to midgraft thrombosis, anastomotic stenosis experimental vein grafts.8,10 In a study of rabbit jugular to
contributes significantly to the failures of artificial graft con- carotid interposition grafts, this proliferative rate returned
duits, and this anastomotic disease is similarly character- to a low level by postoperative weeks 2-4, by which time a
ized by a neointimal proliferative response stemming from thick, highly cellular neointimal layer had been formed.
the adjacent artery.16 Inhibition of neointimal hyperplasia Zwolack and colleagues further demonstrated that
has therefore become a focal point for research aimed at neointimal thickening proceeded over the subsequent 4-8
improving the long term success of surgical revascularization weeks, primarily via increased extracellular matrix produc-
(Fig. 23.1). tion, and that the neointimal layer then remained morpho-
The molecular and cellular biology of neointimal hy- metrically stable up to 12 months, establishing a pattern of
perplasia have been best characterized in animal models of cellular kinetics parallel to those observed after rat carotid
arterial injury that mimic the vessel’s response to balloon balloon injury. Francis et al27 succeeded in demonstrating
angioplasty.17 A primary process of neointimal hyperplasia the release of PDGF in porcine vein grafts at 1 and 4 weeks
is the activation of medial VSMC, and their subsequent pro- after surgery, while Hoch et al12 established the time course
liferation and migration across the internal elastic lamina. of both TGF-!1 and PDGF-A expression by rat vein graft
Numerous factors have been found to stimulate VSMC pro- cells and the relationship of this growth factor expression to
the development of graft neointimal hyperplasia. Further therefore likely to be unsuccessful. There is a final common
studies have since documented the elaboration of cytokines pathway, however, onto which these various proliferative
such as IL-1!, TNF and MCP-1 in the vein graft wall during stimuli and their secondary messengers converge: the mo-
the first 1-4 weeks after operation;14,28 these proteins may lecular and genetic machinery controlling entry into and
play roles in the further “activation” of vascular cells, and progression through the cell cycle (Fig. 23.2). Scientists now
may enhance the infiltration of the vessel wall by pro-in- have a growing understanding of the complex regulation of
flammatory cells. the movement of cells from quiescence (G0-phase) into the
Neointimal growth also occurs in vascular prostheses phases of:
made of artificial materials. Ingrowth of endothelial and 1. Preparation for chromosomal duplication
VSMC from the anastomotic ends of experimental grafts (G1-phase);
comprised of either Dacron or polytetrafluoroethylene 2. DNA synthesis (S-phase);
(PTFE) progresses along the length of the graft to varying 3. Preparation for cell division (G2-phase); and finally
degrees, depending upon the species and material. 29 4. Mitosis (M-phase), and opportunities for targeted
Neointimal thickness, however, is always greatest near the intervention in these processes have therefore
anastomosis, and this heightened perianastomotic thicken- become available.
ing correlates to the clinical phenomenon of anastomotic
stenosis in artificial grafts.30 Increased porosity of the graft Cell Cycle Progression:
material also allows direct ingrowth of capillaries through A Careful Orchestration
the graft wall in animal models, with subsequent accelera- The precision with which cellular growth and prolif-
tion of neointima formation in the grafts’ midportions.31 eration are governed during normal development and heal-
As in the injured artery and the healing vein graft, prosthetic ing has necessitated the development of an elaborate system
vascular graft neointima formation tends to reach a point of checkpoints and regulatory events in the molecular con-
of stable equilibrium, after which cellular proliferation ceases trol of cell cycle progression (Fig. 23.3). Although a number
to contribute to neointimal thickness.30 Several animal mod- of details have been found to vary slightly in different cell
els, including dog and baboon, exhibit complete coverage types and in different species, a remarkably conserved pat-
of Dacron and high porosity PTFE grafts with neointimal tern of gene upregulation and enzymatic activity has been
and endothelial linings, although neointima formation in observed during the movement of cells through the various
human grafts is generally limited to the perianastomotic re- stages of new DNA synthesis and cellular division. In this
gion.29 Clowes and associates have documented the produc- context, several classes of molecules have been described as
tion of both PDGF and TGF-! in the walls of prosthetic vas- playing well defined roles in the management of cell cycle
cular grafts, by both inflammatory and proliferating progression, and studies have confirmed many of these roles
vascular cells.32 in VSMC in vitro.
A wide array of growth factors, such as PDGF, bFGF One of the first responses of cells to stimulation by
and IGF, have all been shown to stimulate the VSMC prolif- growth factors or other mitogens is the upregulated expres-
eration that is a necessary part of neointimal hyperplasia. sion of a series of protooncogenes, also known as immedi-
Strategies to inhibit this proliferative process via blockade ate early genes. Among these genes, c-fos and c-myc are
of any one or two of these redundant molecular targets are known to be important not only for entry of cells into the
cell cycle, but for successful progression through all four protooncogenes such as c-myc, genes encoding enzymes such
stages as well.33,34 The protein products of these genes act as as dihydrofolate reductase, and those for cell cycle regula-
transcription factors that increase the expression of other tory proteins such as Cdk2 and cyclin E. Hyperphos-
molecules (in particular the cyclin-dependent kinases de- phorylation of Rb and activation of E2F comprises the re-
scribed below) whose activities are required to trigger cell striction point R in the cell cycle, beyond which the cell is
cycle switches. The product of another protooncogene, committed to DNA synthesis and after which the cell cycle
c-myb, acts similarly to upregulate cell cycle proteins, whereas can be completed without further stimulation from exter-
the ras gene encodes a membrane-bound, guanine nucle- nal growth factors.39
otide-binding protein that couples signals from surface ty- After DNA replication has taken place in S-phase, an-
rosine kinase growth factor receptors to cytoplasmic second- other cyclin-Cdk complex, that of cyclin B and Cdk1 (also
ary messenger systems.35 known as cell division cycle 2 kinase or cdc2 kinase) is re-
The cyclin-dependent kinases (Cdks) form another sponsible for final progression of the cell through the G2/M
class of molecules that directly regulate the progression of transition. This complex, the mitosis promoting factor
cells through the cell cycle. Other proteins, termed cyclins, (MPF), requires phosphorylation by CAK and dephospho-
act as cofactors for the Cdks, and the binding of specific rylation by cdc25 phosphatase for activation.41,42 An example
cyclins to specific Cdks leads to formation of active holoen- of the coordination of cell cycle events is reflected in the fact
zymes. Both cyclins and Cdks are expressed only at low lev- that c-myc protein not only contributes to the upregulation
els in quiescent cells, and are upregulated at various inter- of Cdk1 protein levels, but also to induction of cdc25 phos-
vals after stimulation of the cell into cell cycle progression. phatase and thereby to Cdk1 activation.43 Activated MPF
Once upregulated, Cdk expression remains high as long as initiates mitosis, and also triggers a proteosomal pathway
the cell continues to progress through rounds of the cell cycle, involving ubiquination that not only facilitates anaphase,
whereas cyclin protein levels fluctuate at different points but also leads to destruction of cyclin B itself in a negative
through cell cycle progression. Early in the G1-phase, cyclin feedback loop.44
D and Cdk4 are upregulated and form a complex together In addition to cell cycle promoting factors, a number
with another cell cycle regulatory protein, proliferating cell of natural inhibitors of cell cycle progression are known to
nuclear antigen (PCNA). Cdk4 further requires phospho- play important roles in the management of cell cycle pro-
rylation by a Cdk activating kinase (CAK) to complete its gression. One group of these molecules is the cyclin-depen-
activation. Cdk2, which forms a holoenzyme with cyclin E, dent kinase inhibitors (CKIs). These inhibitors bind to cyclin/
is the other primary G1-phase Cdk, and it is responsible, Cdk complexes and block their enzymatic activation. In sev-
along with cyclin D/Cdk4, for hyperphosphorylation of the eral cell types, including VSMC, the CKI p27Kip1 is expressed
retinoblastoma gene product Rb.36-38 at high levels during G0-phase, and a drop in this protein
In quiescent cells, Rb exists in its hypophosphorylated level is seen during entry into G1-phase.45 This drop is be-
form, pRb, and resides in a complex with cyclin A, Cdk2 lieved to be necessary for effective phosphorylation of pRb
and the transcription factor E2F. Hyperphosphorylation to at the R point. Interestingly, another CKI, p21Cip1, is ex-
the ppRb state results in a release of E2F from this com- pressed only after cell cycle entry has been initiated, and it is
plex.39 E2F then transactivates a number of other genes believed to play a role in controlling cell cycle progression.45
whose products are essential for progression through the The tumor suppressor p53 acts as a transcription factor to
S-, G2- and M-phases of the cell cycle.40 These genes include inhibit cell cycle progression under certain conditions.46 Beta
particle and ultraviolet light irradiation, for example, lead The first report of in vivo intervention in vascular cell
to a p53-mediated upregulation of p21Cip1 expression, cycle gene expression was made by Simons et al,56 and in-
thereby preventing progression to S-phase and DNA repli- volved antisense ODN inhibition of c-myb expression after
cation in the context of radiation-induced DNA damage.47 rat carotid balloon injury. A porcine arterial balloon injury
In the presence of free E2F protein, p53 has also been linked model was subsequently used to demonstrate a similar re-
to induction of programmed cell death, or apoptosis, pro- duction in neointimal lesion size after treatment of the ves-
viding a mechanism for prevention of propagation of dam- sel wall with antisense ODN against c-myc.57 Whereas single
aged DNA in cells that have passed the R point of the cell antisense ODN targeting PCNA, Cdk1 or Cdk2 have also
cycle.48 The homeobox proteins, such as GAX, are another been shown to reduce neointimal hyperplasia after balloon
class of natural cell cycle inhibitors, and are believed to help injury, the simultaneous inhibition of a combination of cell
maintain differentiated cells in a quiescent state.49 cycle genes, such as Cdk1 and cyclin B or Cdk1 and PCNA,
was found to have a more profound, nearly complete inhi-
Cell Cycle Arrest and Neointimal Hyperplasia bition of neointima formation in a rat carotid injury model
The molecular and genetic basis of cell cycle regula- that lasted up to 8 weeks after injury.54,58 Blockade of the
tion has also been explored in the context of the intact ves- latter combination of genes using ribozymes has also suc-
sel wall during the proliferative response to vascular injury. cessfully inhibited neointimal hyperplasia in a similar arte-
Increases in mRNA levels of the protooncogenes c-myc, c-fos, rial injury model.59
and c-jun have been documented within hours after arterial The synergy observed with inhibition of multiple cell
balloon injury, whereas protein levels of the cell cycle in- cycle regulatory genes led to studies of in vivo inhibition of
hibitors GAX and p27Kip1 are rapidly downregulated.50-52 the transcription factor E2F using decoy ODN transfection
Expression of the “immediate early” genes c-fos and c-jun after carotid injury.60 Fusigenic liposomes comprised of coat
has also been demonstrated in human saphenous vein seg- proteins from inactivated hemagglutinating virus of Japan
ments within hours after harvest, even without re-implan- (HVJ) particles and neutral lipids were used to enhance de-
tation as bypass grafts.53 Expression of c-myc peaks a sec- livery of the decoy ODN. Free E2F is responsible for the
ond time 7 days after arterial injury, when VSMC prolifera- transactivation of up to a dozen cell cycle regulatory pro-
tive activity has been shown to reach its highest levels.50 The teins, including c-myc, Cdk1 and PCNA, and its blockade
upregulation of immediate early genes is followed in turn with the single decoy ODN proved as effective as combina-
by increases in both the mRNA and protein levels of cyclins tions of antisense ODN directed against multiple cell cycle
and Cdks in the arterial wall, within 1-4 days after injury.54,55 gene targets.
Cell cycle gene activation has further been documented in The transfer of genes encoding several bioengineered
medial and neointimal VSMC via the immunohistochemi- or naturally occurring cell cycle inhibitory proteins has also
cal staining of increased levels of PCNA in this time period succeeded in reducing neointimal hyperplasia after arterial
after injury, and increased PCNA and Cdk1 protein levels balloon injury. Overexpression of either p21Cip1 or GAX, for
have similarly been measured in autologous rabbit vein grafts example, has been shown to reduce neointimal hyperplasia
at 4 days after surgery.8 in injured porcine and rabbit arteries, respectively, and
Understanding the control of cell cycle progression at neointima formation has similarly been reduced via trans-
the level of cell cycle regulatory gene expression has led to a duction of porcine femoral arteries with a nonphos-
number of gene-based approaches at inhibiting VSMC pro- phorylatable mutant of Rb and of rat carotid arteries with a
liferation after vascular injury. Genetic manipulation can dominant negative mutant of Ras.61-64 Cell cycle interrup-
involve either the blockade of target genes using oligonucle- tion via gene transfer, however, is likely to require a very
otide transfection, or the introduction of genes using lipids high transduction efficiency to achieve a clinically signifi-
or viral vectors. Antisense oligodeoxynucleotides (ODN) are cant effect on VSMC proliferation and neointimal growth,
short chains of single stranded DNA that bear an antisense, since cell cycle progression will be blocked only in trans-
or Watson-Crick complementary, sequence to that of a re- duced cells. The studies described all used replication-defi-
gion of the target gene mRNA. After delivery to a target cell, cient, recombinant adenoviral vectors to achieve transgene
the ODN bind to this mRNA and prevent its translation into delivery to the target vessels. Although these recombinant
protein. Several mechanisms of antisense activity have been adenoviruses are the most widely studied and efficient vec-
proposed; the most commonly accepted, however, involves tors yet described in vascular tissue, transgene expression is
the destruction of DNA-RNA hybrids via RNAse H. An al- generally limited to approximately 30-50% of cells in an ar-
ternative gene blockade strategy utilizes double-stranded terial wall, and only after some form of vessel wall injury.63,65
“decoy” ODN that bear a consensus binding site for a target Furthermore, infection with these vectors is associated with
transcription factor protein (Fig. 23.4). The factor becomes a prominent inflammatory reaction, transient gene expres-
bound by the excess of decoy ODN, and is prevented from sion and the hazard of possible viral mutation. Nonviral
interacting with its binding site in the promoter region of a means of manipulating cell cycle gene expression may there-
gene under its control. Upregulation of target gene transcrip- fore provide a more practical opportunity at the present time
tion is therefore inhibited. Ribozymes, chains of RNA that for influencing vascular proliferative disease in both human
catalyze cleavage of mRNA in a sequence-specific manner, arteries and vein grafts.
offer another alternative for gene blockade.
246 Tissue Engineering of Prosthetic Vascular Grafts
Genetic Engineering of Vein Graft Resistance significant neointimal hyperplasia, these genetically engi-
to Atherosclerosis via Cell Cycle Gene Blockade neered grafts resisted the accelerated, diet-induced deposi-
Neointimal hyperplasia plays a particularly important tion of foam cells and generation of atherosclerotic plaque
role in the pathogenesis of autologous vein graft failure, not observed in control grafts (Fig. 23.5).
only through direct encroachment upon the vessel lumen, Vein graft susceptibility to accelerated atherosclerosis
but also because it acts as a substrate for accelerated graft is associated with endothelial dysfunction similar to that seen
atherosclerosis. Neointima formation is a well characterized in early arterial atherosclerosis.66 In addition to blockade of
response to multiple forms of vascular injury, ranging from VSMC proliferation during the immediate postoperative
gentle denudation to desiccation to balloon distention. The period, inhibition of cell cycle regulatory gene expression
ischemic, mechanical and inflammatory injuries associated had a long term stabilizing effect on vein graft endothelial
with vein graft harvest and implantation are therefore likely function.9 This phenotypic alteration was documented via
to provide a strong stimulus for neointima formation. The enhanced endothelial cell nitric oxide synthase activity and
authors have hypothesized that blockade of gene expression maintenance of normal jugular vasoreactivity, as well as
necessary for cell cycle progression can prevent the initia- through reduced superoxide anion generation, adhesion
tion of neointimal hyperplasia as a response to these mul- molecule expression and monocyte binding to graft
tiple factors and stimuli. Furthermore, once graft healing endothelium. This preservation of endothelial function
has progressed and the stimulation from these injuries has may represent either a direct effect of cell cycle gene
subsided, the graft can respond to the hemodynamic impe- blockade in endothelial cells, or it may reflect a change in
tus for wall thickening via vascular hypertrophy and medial the influence normally exerted by underlying abnormal
thickening. Stabilization of vascular cell phenotype and func- neointimal VSMC.
tion during the postoperative period of graft healing may Transfection of the vein graft wall with the single agent
therefore allow vein graft adaptation to the hemodynamic E2F decoy ODN has subsequently been shown to similarly
stresses of the arterial circulation without incurring increased influence vein graft biology and prevent both neointimal
susceptibility to graft disease and failure associated with disease and atherosclerosis.67 This observation has been ex-
neointimal hyperplasia. tended to six months after operation, at which time E2F
This hypothesis was tested in a series of experiments decoy ODN-treated grafts remain free of significant
utilizing the delivery of ODN to cells of rabbit jugular veins neointima formation and resist diet-induced atherosclero-
at the time of grafting into the carotid artery. A combina- sis. Studies have also documented that ODN can be deliv-
tion of antisense ODN against both Cdk1 and PCNA inhib- ered efficiently to human saphenous vein segments ex vivo,
ited neointimal hyperplasia by 90%, and this inhibition per- and in an organ culture system these ODN have inhibited
sisted for up to 10 weeks after grafting.8 By week 6, however, target gene function in a sequence specific manner. These
adaptive graft wall thickening approached levels seen in findings have served as the basis for initiation of a large scale
untreated grafts largely through hypertrophy of the medial human clinical trial to test the efficacy of intraoperative vein
layer. This thickening translated into an effective reduction graft engineering via E2F decoy ODN transfection in pre-
in tangential wall stress. Most importantly, in the absence of venting infrainguinal bypass graft failures.68 This study rep-
Cell Cycle Interruption to Inhibit Neoinitmal Hyperplasia 247
resents the first attempt at integrating a gene therapy strat- derway that may establish the practical, long term benefits
egy into a routine cardiovascular surgical procedure, and that a one time intervention in cell cycle regulation may have
the first large scale testing of our ability to influence cell cycle for vascular graft compatibility and adaptation. The results
biology and its role in human vascular pathogenesis. of these basic science and clinical studies will likely yield
vascular biologists, bioengineers and clinicians new tools to
Summary aid in the design of vascular prostheses with enhanced du-
Vascular cell proliferation is clearly a requisite com- rability and performance.
ponent of the neointimal hyperplasia that is known to com-
promise the long term success of autologous and prosthetic References
vascular reconstruction. Cell cycle regulation that is at the 1. Campeau LM, Enjalbert J, Lesperance MC, Bourassa P,
heart of this proliferation is regulated by a complex and well Kwiterowich S, Wacholder A. Sniderman A. The relation
orchestrated series of molecular and genetic events. Inter- of risk factors to the development of atherosclerosis in
ruption of these events at critical points of cell cycle pro- saphenous vein bypass grafts and the progresion of dis-
ease in the native circulation: A study 10 years after
gression has been shown to influence neointimal hyperpla-
aortocoronary bypass surgery. N Engl J Med 1984;
sia in models of arterial injury. Although a powerful tool to 311:1329-1334.
block cellular proliferation, inhibition of cell cycle progres- 2. Grondin CM, Campeau L, Thornton JC, Engle JC, Cross
sion, and blockade of cell cycle regulatory gene expression FS, Schreiber H. Coronary artery bypass grafting with
in particular, may have even more profound effects on the saphenous vein. Circ 1989; 79:I-24-I-29.
development of vascular graft disease through its stabilizing 3. Cox JL, Chaisson DA, Gotleib AI. Stranger in a strange
influence on vascular cell phenotype and tissue behavior. land: The pathogenesis of saphenous vein graft stenosis.
Current studies are probing the molecular links be- Prog Cardiovasc Dis 1991; 34:45-68.
tween cell cycle entry and other aspects of cellular activa- 4. Clowes AW, Reidy MA. Prevention of stenosis after vas-
tion, such as adhesion molecule expression and cytokine cular reconstruction: Pharmocologic control of intimal
secretion. Even as our understanding of vascular cell cycle hyperplasia—A review. J Vasc Surg 1991; 13:885-91.
biology continues to grow, clinical studies are already un-
248 Tissue Engineering of Prosthetic Vascular Grafts
5. Hirsch GM, Karnovsky MJ. Inhibition of vein graft inti- 20. Reidy MA, Fingerle J, Lindner V. Factors controlling the
mal proliferative lesions in the rat by heparin. Am J Path development of arterial lesions after injury. Circulation
1991; 139:581-7. 1992; 86:III43-46.
6. Chen C, Mattar SG, Hughes JD, Pierce GF, Cook JE, Ku 21. Sterpetti AV, Cucina A, Randone B, Palumbo R, Stipa F,
DN, Hanson SR, Lumsden AB. Recombinant mitotoxin Proietti P, Saragosa MT, Santoro-D’Angelo L, Cavallaro
basic fibroblast growth factor-saporin reduces venous A. Growth factor production by arterial and vein grafts:
anastomotic intimal hyperplasia in the arteriovenous graft. relevance to coronary artery bypass grafting. Surgery 1996;
Circ 1996; 94:1989-95. 120:460-7.
7. Davies MG, Dalen H, Kim JH, Barber L, Svendsen E, 22. Clowes AW, Schwartz SM. Significance of quiescent
Hagen PO. Control of accelerated vein graft atheroma smooth muscle migration in the injured rat carotid ar-
with the nitric oxide precursor: L-arginine. J Surg Res tery. Circ Res 1985; 56:139-145.
1995; 59:35-42. 23. Clowes AW, Reidy MA, Clowes MM. Mechanisms of
8. Mann MJ, Gibbons GH, Kernoff RS, Diet FP, Tsao P, stenosis after arterial injury. Lab Invest 1983; 49:208-215.
CookeJP, Kaneda Y, Dzau VJ. Genetic engineering of vein 24. Majeski MW, Lindner V, Twardzik DR, Reidy MA. Pro-
grafts resistant to atherosclerosis. Proc Natl Acad Sci USA. duction of transforming growth factor beta-1 during re-
1995; 92:4502-4506. pair of arterial injury. J Clin Invest 1991; 88:904-910.
9. Mann MJ, Gibbons GH, Tsao PS, von der Leyen HE. 25. Grant MB, Wargovich TJ, Ellis EA, Caballero S, Mansour
Buitrago R, Kernoff R, Cooke JP, Dzau VJ. Cell cycle in- M, Pepine CJ. Localization of insulin-like growth factor I
hibition preserves endothelial function in genetically en- and inhibition of coronary smooth muscle cell growth by
gineered vein grafts. J Clin Invest 1997; 99:1295-1301. somatostatin analogues in human coronary smooth
10. Zwolak RM, Adams MC, Clowes AW. Kinetics of vein muscle cells. A potential treatment for restenosis? Circ
graft hyperplasia: Association with tangential stress. J Vasc 1994; 89:1511-1517.
Surg 1987; 5:126-136. 26. Rakugi H, Wang DS, Dzau VJ, Pratt RE. Potential im-
11. Moggio RA, Ding JZ, Smith CJ, Tota RR, Stemerman MB, portance of tissue angiotensin-converting enzyme inhibi-
Reed GE Immediate-early gene expression in human tion in preventing neointima formation. Circ 1994;
saphenous veins harvested during coronary artery bypass 90:449-455.
graft operations. J Thorac Cardiovasc Surg 1995; 27. Francis SE, Hunter S, Holt CM, Gadsdon PA, Rogers S,
110:209-13. Duff GW, Newby AC, Angelini GD. Release of platelet-
12. Hoch JR, Stark VK, Turnipseed WD. The temporal rela- drived growth factor activity from pig venous arterial
tionship between the development of vein graft intimal grafts. J Thor Cardiovasc Surg 1994; 108:540-548.
hyperplasia and growth factor gene expression. J Vasc Surg 28. Stark VK, Hoch JR, Warner TF, Hullett DA. Monocyte
1995; 22:51-58. chemotactic protein-1 expression is associated with the
13. Tanaka H, Sukhova GK, Swanson SJ, Clinton, SK, Ganz development of vein graft intimal hyperplasia. Arterioscler
P, Cybulsky MI, Libby P. Sustained activation of vascular Thromb Vasc Biol 1997; 17:1614-21.
cells and leukocytes in the rabbit aorta after balloon in- 29. Zacharias RK, Kirkman TR, Clowes AW. Mechanisms of
jury. Circ 1993; 88:1788-1803. healing in synthetic grafts. J Vasc Surg 1987; 6:429-36.
14. Faries PL, Marin ML, Veith FJ, Ramirez JA, Suggs WD, 30. Clowes AW, Kirkman TR, Clowes MM. Mechanisms of
Parsons RE, Sanchez LA, Lyon RT. Immunolocalization arterial graft failure. II. Chronic endothelial and smooth
and temporal distribution of cytokine expression during muscle cell proliferation in healing polytetrafluoroethylene
the development of vein graft intimal hyperplasia in an prostheses. J Vasc Surg 1986; 3:877-84.
experimental model. J Vasc Surg 1996; 24:463-71. 31. Golden MA, Hanson SR, Kirkman TR, Schneider PA,
15. Yu X, Dluz S, Graves DT, Zhang L, Antoniades HN, Hol- Clowes AW. Healing of polytetrafluoroethylene arterial
lander W, Prusty S, Valente AJ, Schwartz CJ, Sonenshein grafts is influenced by graft porosity. J Vasc Surg 1990;
GE. Elevated expression of monocyte chemoattractant 11:838-44.
protein 1 by vascularsmooth muscle cells in hyper- 32. Zacharias RK, Kirkman TR, Kenagy RD, Bowen-Pope DF,
cholesterolemic primates. Proc Natl Acad Sci USA 1992; Clowes AW. Growth factor production by
89:6953-7. polytetrafluoroethylene vascular grafts. J Vasc Surg 1988;
16. Hamdan AD, Misare B, Contreras M, LoGerfo FW, Quist 7(4):606-10.
WC. Evaluation of anastomotic hyperplasia progression 33. Karn J, Watson JV, Lowre AD, Green SM, Vedeckis W:
using the cyclin specific antibody MIB-1. Am J Surg 1996; Regulation of cell cycle duration by c-myc levels.
172:168-70. Oncogene 1989; 4:773-787.
17. Clowes AW, Reidy MA, Clowes MM. Kinetics of cellular 34. Pai SR, Bird RC. c-fos expression is required during all
proliferation after arterial injury. I. Smooth muscle growth phases of the cell cycle during exponential cell prolifera-
in the absence of endothelium. Lab Invest 1983; tion. Anticancer Res 1994; 14:985-994.
49:327-333. 35. Avruch J, Zhang XF, Kyriakis JM. Raf meets Ras: Com-
18. Gospodarowics D, Ferrara N, Haaparanta T, Neufeld G. pleting the framework of a signal transduction pathway.
Basic fibroblast growth factor: Expression in cultured bo- Trends Biochem Sci 1994; 19:279-283.
vine vascular smooth muscle cells. Eur J Cell Biol 1988; 36. Pardee AB. G1 events and regulation of cell proliferation.
46:144-151. Science 1989; 246:603-608.
19. Walker LN, Bowen-Pope DF, Ross R, Reidy MA. Produc- 37. Morgan DO. Principles of CDK regulation. Nature 1995;
tion of platelet-derived growth factor-like molecules by 374:131-134.
cultured arterial smooth muscle cells accompanies prolif- 38. Sherr CJ. Mammalian G1 cyclins. Cell 1993; 73:1059-1065.
eration after arterial injury. Proc Natl Acad Sci USA 1986; 39. Weinberg RA. The retinoblastoma protein and cell cycle
83:7311-7315. control. Cell 1995; 81:323-330.
Cell Cycle Interruption to Inhibit Neoinitmal Hyperplasia 249
40. DeGregori J, Kowalik T, Nevins JR. Cellular targets for intimal arterial smooth muscle cell accumulation in vivo.
activation by the E2F1 transcription factor include DNA Nature 1992; 359:67-70.
synthesis- and G1/S-regulatory genes. Mol Cell Biol 1995; 57. Shi Y, Fard A, Galeo A, Hutchinson HG, Vermani P,
15:4215-4224. Dodge GR, Hall DJ, Shaheen F, Zalewski A. Transcatheter
41. King RW, Jackson PK, Kirschner MW. Mitosis in transi- delivery of c-myc antisense oligomers reduce neointimal
tion. Cell 1994; 79:563-571. formation in a porcine model of coronary artery balloon
42. Coleman TR, Dunphy WG. cdc2 regulatory factors. Curr injury. Circ. 1994; 90:944-951.
Opin Cell Biol 1994; 6:877-882. 58. Morishita R, Gibbons GH, Kaneda Y et al. Pharmacoki-
43. Galaktionov K, Chen X, Beach D. cdc25 cell-cycle phos- netics of antisense oligodeoxynucleotides (cyclin B1 and
phatase as a target of c-myc. Nature 1996; 382:511-517. cdc2 kinase) in the vessel wall in vivo: Enhanced thera-
44. King RW, Deshaies RJ, Peters JM, Kirschner MW. How peutic utility for restenosis by HVJ-liposome delivery.
proteolysis drives the cell cycle. Science 1996; Gene 1994; 149:13-9.
274:1652-1658. 59. Dev V, Parikh A, Ren M, Goldenberg T, Fishbein MC,
45. Sherr CJ, Roberts JM. Inhibitors of mamalian G1 cyclin- Eigler N, Rossi J, Litvack F. Ribozymes to cell division
dependent kinases. Gen Dev 1995; 9:1149-1163. cycle (CDC-2) kinase and proliferating cell nuclear anti-
46. Levine AJ. p53, the cellular gatekeeper for growth and gen (PCNA) prevent intimal hyperplasia in rat carotide
division. Cell 1997; 88:323-331. artery (abstract). Circulation (suppl I) 1995; 92:I-634.
47. el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons 60. Morishita R, Gibbons GH, Horiuchi M et al. A novel
R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein molecular strategy using cis element “decoy” of E2F bind-
B. WAF1, a potential mediator of p53 tumor suppression. ing site inhibits smooth muscle proliferation in vivo. Proc
Cell 1993; 75:817-825. Natl Acad Sci USA 1995; 92:5855-9.
48. Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, 61. Chang MW, Barr E, Lu MM, Barton K, Leiden JM. Ad-
Craig RW. Participation of p53 protein in the cellular enovirus-mediated over-expression of the cyclin/cyclin-
response to DNA damage. Cancer Res 1991; 51:6304-6311. dependent kinase inhibitor, p21 inhibits vascular smooth
49. Gorski DH, Patel CV, Walsh K. Homeobox transcription muscle cell proliferation and neointima formation in the
factor regulation in the cardiovascular system. Trends rat carotid artery model of balloon angioplasty. J Clin
Cardiovasc Med 1993; 3:184-190. Invest 1995; 96:2260-2268.
50. Miano JM, Vlasic N, Tota RR, Stemerman MB. Smooth 62. Maillard L, Belle EV, Perlman H, Branellec D, Barry J,
muscle cell immediate-early gene and growth factor acti- Steg G, Walsh K. GAX adenovirus percutaneous gene
vation follows vascular injury. A putative in vivo mecha- transfer inhibits neointima formation in a rabbit model
nism for autocrine growth. Arterioscler Thromb 1993; of balloon angioplasty (abstract). Circulation (suppl I)
13:211-219. 1996; 94:I-637.
51. Weir L, Chen D, Pastore C, Isner JM, Walsh K. Expres- 63. Chang MW, Barr E, Seltzer J, Jiang YQ, Nabel GJ, Nabel
sion of gax, a growth arrest homeobox gene, is rapidly EG, Parmacek MS, Leiden JM. Cytostatic gene therapy for
down-regulated in the rat carotid artery during the pro- vascular proliferative disorders with a constitutively ac-
liferative response to balloon injury. J Biol Chem 1995; tive form of the retinoblastoma gene product. Science
270:5457-5461. 1995; 267:518-522.
52. Braun-Dullaeus RC, von der Leyen HE, Mann MJ, Zhang 64. Indolfi C, Avvedimento EV, Rapacciuolo A, Di Lorenzo
L, Morris RE, Dzau VJ. Loss of p27Kip1 and induction E, Esposito G, Stabile E, Feliciello A, Mele E, Giuliano P,
of Cdk1 in the rat carotid artery following balloon cath- Condorelli G et al. Inhibition of cellular ras prevents
eter injury. In vivo and in vitro influence of rapamycin smooth muscle cell proliferation after vascular injury in
(abstract). FASEB J 1997; 11:A153. vivo. Nat Med 1995; 1:541-545.
53. Sterpetti AV, Cucina A, Randone B, Palumbo R, Stipa F, 65. Schulick AH Dong G Newman KD Virmani R Dichek DA.
Proietti P, Saragosa MT, Santoro-D’Angelo, Cavallaro A. Endothelium-specific in vivo gene transfer. Circ Res 1995;
Growth factor production by arterial and vein grafts: Rel- 77:475-85.
evance to coronary artery bypass grafting. Surgery 1996; 66. Ku DD, Caulfield JB, Kirklin JK. Endothelium-dependent
120:460-7. responses in long-term human coronary artery bypass
54. Kaneda Y, Ogihara T, Dzau VJ. Intimal hyperplasia after grafts. Circ 1991; 83:402-411.
vascular injury is inhibited by antisense cdk 2 kinase oli- 67. Mann MJ, Kernoff R, Dzau VJ. Intraoperative transfec-
gonucleotides. J Clin Invest 1994; 93:1458-1464. tion with E2F decoy oligonucleotide yields long term re-
55. Abe J, Zhou W, Taguchi J, Takuwa N, Miki K, Okazaki sistance to vein graft atherosclerosis. Journal of Investi-
H, Kurokawa K, Kumada M, Takuwa Y. Suppression of gative Medicine, 1997; 45:224A.
neointimal smooth muscle cell accumulation in vivo by 68. Mann MJ, Whittemore AD, Donaldson MC, Belkin MA,
antisense cdc2 and cdk2 oligonucleotides in rat carotid Orav EJ, Polak J, Dzau VJ. Preliminary clinical experi-
artery. Biochem Biophys Res Commun 1994; 198:16-24. ence with genetic engineering of human vein grafts: Evi-
56. Simons M, Edelman ER, DeKeyser JL, Langer R, dence for target gene inhibition. Circ. 1997; 96:I4.
Rosenberg RD. Antisense c-myb oligonucleotides inhibit
Facilitation of Healing
Chemotaxis
CHAPTER 24
Signaling Mechanisms for Vascular Cell Migration
Ian Zachary
Introduction
I t is increasingly recognized that migration of vascular smooth muscle cells (VSMC) from
the media is a key event in progressive intimal thickening leading to atherosclerosis and
other occlusive vasculoproliferative complications.1,2 Transendothelial migration into the
tunica intima of other cell types, particularly monocytes and T lymphocytes, is thought to
be central both to the formation of the mature atherosclerotic lesion and to the pathogen-
esis of the disease.1,2 Arterial bypass graft surgery is one of the commonest treatments for
atherosclerotic cardiovascular disease, and a major cause of failure of bypass graft surgery is
stenosis of the graft, which occurs predominantly at, or contiguous to, the sites of anasto-
mosis.3-6 Stenosis of the grafted vessel is primarily due to the proliferation and migration of
VSMC.7 The use of prosthetic grafts in place of autogenous vessels has been an important
development in vascular surgery, but prosthetic materials have lower patency rates com-
pared to veins. The lower patency of prosthetic grafts is widely attributed to their inability to
form an endothelial lining, thus contributing to enhanced thrombogenicity and VSMC pro-
liferation/migration. Animal studies indicate that seeding prosthetic grafts with endothelial
cells (EC) prior to implantation can significantly reduce neointimal hyperplasia and
thrombogenicity.8,9 As these brief introductory remarks suggest, vascular cell migration may
be relevant for cardiovascular disease from more than one perspective. Thus, while VSMC
and monocyte migration contribute to progression of the primary disease and—in the case
of VSMC—complications consequent upon treatment, the promotion of EC migration and
proliferation may be of equal importance in clinical situations, either where prosthetic grafts
are used or where the endothelium is damaged (e.g., angioplasty).
The aim of this chapter is to provide an overview of the extracellular factors and intra-
cellular mechanisms implicated in the regulation of vascular cell migration. Emphasis will
be placed on the migration of VSMC and EC, though the role of the migration of other cell
types in the arterial wall and the atherosclerotic microenvironment (e.g., monocytes) will
also be considered. Emphasis will be placed on recent developments in our understanding
of the molecular basis of cell migration, such as the role of focal adhesion kinase (FAK) and
Rho proteins. Inevitably, many of the findings that will be discussed have come from studies
in other nonvascular cell types, but it is anticipated that these findings will have relevance
for the migration of vascular cells. The first section of the chapter (Migration Factors) will
describe the major factors known to modulate vascular cell locomotion in cell culture. The
second section (Chemotactic Signal Transduction) will go on to discuss intracellular mecha-
nisms implicated in cell migration and linked cellular processes.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler
©1999 R.G. Landes Company.
252 Tissue Engineering of Prosthetic Vascular Grafts
tional # receptors.38 Low # receptor expression cannot en- is a distant relative of PDGF and is a member of a new fam-
tirely account for the lack of chemotactic responsiveness to ily of growth factors, which also includes VEGF-B, VEGF-C
PDGF-AA, however. For example, though human foreskin and placenta growth factor (PlGF).51-55 VEGF expression is
fibroblasts express 20,000-30,000 PDGF-AA binding sites per upregulated by hypoxia and by cytokines and growth fac-
cell, and rat and baboon VSMC possess PDGF # receptors tors in several cell types, including arterial VSMC56-58 and
albeit at relatively low levels, the AA isoform failed to stimu- EC.59,60 It has been established that VEGF plays a central
late migration of all these cells and inhibited their migra- role in angiogenesis in a variety of biological processes and
tory response to the BB isoform.34-36 In addition, TGF-!1 disease states, including embryogenesis, wound healing,
decreased migration of rat VSMC as a consequence of tumor growth, and collateral blood vessel formation in
PDGF-AA secretion.36 Koyama et al34 showed that the myocardial ischemia.61,62
PDGF-AA inhibition of baboon VSMC migration stimu- Most of the biological effects of VEGF are attributed
lated either by PDGF-BB or by fibronectin was abolished by to its ability to stimulate EC mitogenesis and recently this
specific antibodies to the # receptor and is therefore medi- has focused interest on the role of VEGF in accelerating re-
ated through this specific receptor isotype. These findings endothelialization following balloon angioplasty. Initial stud-
suggest that the stimulation of directed migration of VSMC ies in animal models of balloon injury have proved promis-
is mediated by the ! receptor, while the # receptor may play ing, and administration of VEGF protein and VEGF gene
a specific role in the negative regulation of chemotaxis. It is transfer have been shown to inhibit intimal thickening fol-
at present unclear, however, whether these conclusions can lowing balloon angioplasty and improve blood flow in is-
be extended either to human VSMC or to VSMC migration chemic limbs, effects mediated through stimulation of EC
in vivo. Human VSMC express # receptors37,38 and respond regrowth and angiogenesis respectively.63-66
mitogenically to PDGF-AA.39 Relatively few studies of the In addition to its mitogenic effects in EC, VEGF also
effects of PDGF-AA on the migration of these cells have been promotes the migration of EC and it is increasingly recog-
published, but it was reported that PDGF-AA stimulates nized that EC migration plays an essential role in angiogen-
chemotaxis in human VSMC and that this response esis and vascular modelling.61,62,67,68 VEGF also has a num-
correlated with the ability of human VSMC to respond ber of biological activities which may contribute to vessel
mitogenically to the AA isoform.39 It is unknown whether wall pathology. VEGF is a permeability-increasing factor,51,69
the # receptor has an inhibitory effect on migration of and also stimulates monocyte chemotaxis and tissue factor
human VSMC. production.70,71 It has also been proposed that VEGF may
Of the other polypeptide ligands for receptor protein play a role in neovascularization of the advanced ather-
tyrosine kinases (PTKs), heparin-binding epidermal growth osclerotic plaque.57,72-74 Induction of VEGF expression in
factor-like growth factor (HB-EGF) has been reported to the arterial wall may therefore be pro-atherogenic both
stimulate migration of bovine aortic smooth muscle cells in through its effects on the permeability of the endothelium,
a heparin-dependent manner.40 Insulin-like growth factor-I thereby facilitating the infiltration of monocytes, platelets
(IGF-1) is present at high levels in platelet #-granules, is se- and T cells, and by a direct effect on monocyte migration.
creted by cultured VSMC and EC41,42 and has increased ex- At later stages of atherogenesis, neo-vascularization induced
pression in rat aorta following balloon injury.43 IGF-1 has by hypoxia and mediated by VEGF may also contribute to
been reported to be a migration factor for human VSMC,37 disease progression.
and results from our laboratory show that IGF-1 promotes
chemotaxis in rabbit aortic VSMC (Abedi H, Zachary I, un- Angiotensin II
published observations). In both cell types, IGF-1 produces The vasoconstrictor angiotensin II is implicated in the
a weaker migratory response compared to PDGF-BB. regulation of VSMC proliferation, but it is unclear whether
Basic fibroblast growth factor (bFGF) is a mitogen for it contributes significantly to migration of these cells. Data
VSMC1,42 but there is little evidence that this factor has from in vitro studies are conflicting. Thus, it was reported
stimulatory effects on VSMC migration in vitro. This factor that angiotensin II stimulates migration of bovine aortic
may play a role in VSMC migration in vivo, however. Ad- VSMC,75 while MII and co-workers found no effect of an-
ministration of bFGF and neutralizing antibodies to bFGF giotensin II on migration of human VSMC.76 It is possible,
respectively stimulated and inhibited VSMC migration fol- however, that angiotensin II may play a role in migration of
lowing balloon injury of the rat carotid artery.44 There is VSMC in vivo. Consistent with this notion, infusion of an-
much stronger evidence that bFGF is a potent migration giotensin II enhanced neointima formation following bal-
factor for EC. Both basic and acidic FGF stimulate move- loon injury of the rat carotid artery77 and, using the same
ment of EC in vitro45-47and have been reported to promote model, it was shown that both inhibitors of angiotensin-
re-endothelialization following balloon injury.48,49 It is likely converting enzyme and antagonists of the angiotensin type 1
that the repertory of polypeptide migration factors for (AT1) receptor inhibited the accumulation of intimal
VSMC will be extended in the future. For example, a 58 kDa VSMC.78 It is unclear from these studies, however, whether
autocrine migration factor which appears to be different the effects of angiotensin II are direct or indirectly medi-
from PDGF was recently purified from the conditioned ated through increased expression of another migration
medium of rat aortic VSMC.50 factor(s) and/or receptor(s).
The secreted polypeptide vascular endothelial growth
factor (VEGF) (also known as vascular permeability factor)
254 Tissue Engineering of Prosthetic Vascular Grafts
ment/attachment to the cellular substrate is also a crucial as yet showing unambiguous PDGF-BB stimulation of stress
parameter determining the rate and direction of cell loco- fiber or focal contact formation in vascular cells, suggesting
motion. In addition to dynamic adhesive interactions be- that regulation of actin cytoskeleton organization is complex.
tween the cell and its substrate, eukaryotic cell locomotion Given their pivotal cellular location at a point of close
requires the generation of force by the cytoskeleton and as- juxtaposition between the ECM and the actin cytoskeleton,
sociated components.87,88 attention has naturally focused on the role of components
Actin filaments organized in stress fibers and their sites associated with actin and with focal adhesions in regulating
of attachment at focal adhesions are both thought to play a signal transmission between the matrix and the
critical role in the regulation of cell adhesion and locomo- actin cytoskeleton.
tion.107,108 Focal adhesions are specialized juxtamembrane
regions which form at the termini of stress fibers and at sites Actin-Binding Proteins
of attachment of cells to the ECM. Integrins, the adhesive A large number of actin-binding proteins have been
receptors for ECM components, aggregate at focal contacts identified, some of which are thought to play an important
and many nonstructural cytoskeletal-associated components role in regulating the dynamics of actin polymerization. The
are localized to these subcellular structures (Table 24.2). scope of this review precludes a comprehensive discussion,
Interactions between integrins and the cytoskeleton and brief consideration will be directed to some of the find-
are themselves very dynamic. 109,110 For example, in ings most directly relevant to chemotactic signaling (see ref.
nonmotile cells, !1 integrins are localized in focal adhesions 108 for a recent review).
at the ends of stress fibers while in motile cells, integrins are Profilin is a 12-17 kDa protein which is associated with
found in organized, but dynamic substructures termed actin filaments in dynamic areas of the cell such as the lead-
macroaggregates that are in less intimate association with ing lamellae and at the termini of newly-formed stress fi-
focal contacts.109 bers. 108,119 In vitro profilin binds to actin, and
The formation of stress fibers and the assembly of fo- phosphatidylinositol 4,5-bisphosphate (PIP2). Earlier find-
cal contacts is stimulated in adherent cells including VSMC ings suggested that profilin had a strong inhibitory effect on
and EC by a variety of extracellular factors including PDGF, actin polymerization, but it is now believed that the effect
VEGF, bombesin, angiotensin II, the serum component of this protein on actin polymerization may be consider-
lysophosphatidic acid (LPA) and the ceramide sphin- ably more complex. Profilin binds and sequesters G-actin,
gosine.68,111-115 In serum-starved Swiss 3T3 cells and at rela- thus reducing the concentration of free actin monomers and
tively low concentrations, PDGF has been reported to in- lowering the rate of polymerization. On the other hand,
duce a slow increase in focal adhesion assembly and stress profilin also stimulates ATP/ADP exchange on G-actin,
fiber formation as well as intense membrane ruffling.115 The thereby promoting actin polymerization.120 Gelsolins are a
function of membrane ruffling is unclear but it appears to large family of proteins, the most dominant form of which
be closely associated with pinocytosis.115-117 In Swiss 3T3 is 80 kDa and contains multiple actin binding sites.108,121
fibroblasts, PDGF appears to have bimodal effects on actin Gelsolins are associated with focal adhesions and can both
cytoskeleton organization and, at higher concentrations, sever actin filaments and also cap the barbed (fast growing)
induces a marked disruption of the actin cytoskeleton.118 The ends of actin filaments.121 Like profilin, gelsolin binds to PIP2,
relevance of findings in Swiss 3T3 cells for the potent chemo- and a role for this protein in modulating agonist-induced
tactic effects of PDGF-BB in VSMC are unclear, however. actin filament organization through PIP2 hydrolysis has been
PDGF-BB causes no disruption of the actin cytoskeleton in proposed (see below).
cultured human and rabbit VSMC.38 There are no reports
Table 24.2. Cellular components implicated in cytoskeletal reorganization and cell migration
Several lines of evidence suggest that the level of ex- Focal Adhesion Kinase
pression of actin-binding proteins may influence vascular FAK is a member of a growing family of protein ty-
cell motility. Gelsolin has been implicated in EGF-stimu- rosine kinases (PTKs) which also includes the recently iden-
lated cell motility and in histamine-induced changes in ac- tified FAKB and PYK-2.131-133,138,139 FAK was originally iden-
tin filament organization in HUVEC.122 Overexpression of tified as one of a group of proteins that are highly tyrosine
both gelsolin and CapG did not have significant effects on phosphorylated in chicken embryo fibroblasts transformed
actin filament content or organization, but did increase the with the v-src oncogene.140 The cDNA for FAK encodes a
rates of wound healing and chemotaxis. On the other hand, protein with a predicted molecular weight of 119-121 kDa
in rat aortic VSMC, it was reported that gelsolin protein and depending on the species, though on the basis of its migra-
mRNA are reduced during intimal thickening induced by tion in gels it is often designated p125FAK. FAK is widely ex-
endothelial injury of rat aortas.123 pressed in tissues, and is present in fibroblasts, VSMC, EC,
platelets, lymphocytes, monocytes and neuronal cells. A
Integrins 41-43 kDa protein corresponding to the noncatalytic car-
VSMC express !1 and !3 integrins as well as a variety boxyl-terminal FAK domain, termed FRNK (FAK-related
of # subunits including #v and #1.94,124-129 The !1 integrin is nonkinase) is autonomously expressed in some cells.139 A
constitutively expressed in most cells in the body106 and this salient feature of FAK is its subcellular localization to focal
also appears to be true for VSMC.127,128 In contrast, expres- adhesions and this has stimulated intense interest in the role
sion of !3 is upregulated in VSMC by TGF- ! and by of this kinase in the regulation of cell adhesion, cytoskeletal
PDGF-BB, and TGF-! was also reported to induce expres- organization and cell locomotion.131-133
sion of #1 subunits.127,128 Recent results suggest that !1 and The structure of FAK is unique, comprising a central
!3 integrins may have different functions in VSMC adhe- catalytic region flanked by two large noncatalytic domains
sion and migration. Thus, antibodies to !1 inhibit attach- with no homologies to other PTKs (Fig. 24.1). Tyrosine phos-
ment and spreading of rabbit arterial VSMC on collagen and phorylation of FAK is stimulated through the signal trans-
fibronectin substrates126 while osteopontin-stimulated mi- duction pathways initiated by v-src transformation,140,141
gration of VSMC was inhibited by antibodies to !3 but not activation of !1 integrins in fibroblasts142-144 and #IIb/!3
anti-!1 antibody.94 Interestingly, it has been reported that in integrins in platelets,145,146 by aggregation of high-affinity
the early stages of VSMC migration !1 integrins are orga- IgE receptors in mast cells,147 and in Swiss 3T3 fibroblasts
nized into focal adhesions which gradually cover the basal by regulatory peptides including bombesin and
surface of the cells, while !3 integrins remain at the leading endothelin148,149 and by LPA111 and sphingosine.112 It was
edges of cells.129 These findings suggest that !1 integrins are recently reported that tyrosine phosphorylation of FAK is
both necessary and probably sufficient for cell adhesion, also induced by hepatocyte growth factor in oral squamous
while !3 integrins may play a more specific role during cell carcinoma cells.150 The integrity of the actin cytoskeleton
chemotaxis or migration. is essential for FAK tyrosine phosphorylation.68,111,112,118,149
Most integrins have small cytoplasmic domains which Much evidence suggests that the noncatalytic amino-
lack any intrinsic enzymatic activity, and this suggests that and carboxyl-terminal domains mediate associations with
the mechanisms involved in integrin-mediated signaling are integrins, other focal adhesion components, and signaling
fundamentally distinct from those of receptor PTKs or molecules.132,133 Little is known about the function of the
G-protein linked receptors. In the past few years it has be- amino-terminal domain of FAK, but as mentioned above it
come clear that integrin-mediated cell adhesion stimulates may associate directly with the cytoplasmic domains of
several signaling pathways.106,130 In particular, it is now well integrins.151 The COOH-terminal noncatalytic domain is
established that activation of !1 integrins in adherent cells sufficient for targeting of FAK to focal adhesions152 and is
induces tyrosine phosphorylation of focal adhesion kinase also implicated in the association of FAK with paxillin, a
(FAK) and the focal adhesion-associated protein 68 kDa protein which colocalizes to focal contacts. 153
paxillin.107,131-133 Attachment of fibroblasts, EC and lympho- Bombesin, PDGF-BB, VEGF and cell adhesion to fibronectin
cytes to fibronectin also causes an increase in intracellular all induce paxillin tyrosine phosphorylation in fibroblasts
pH,106 and this response is implicated in cell proliferation.130 concomitantly with FAK phosphorylation38,68,143,154 and
Attachment of Swiss 3T3 fibroblasts to fibronectin and paxillin is a putative substrate for FAK.155,156 Paxillin has
laminin has also been reported to activate mitogen-activated recently been molecularly cloned and its sequence reveals a
protein (MAP) kinases,134-6 but, as will be discussed below, number of interesting features, including a proline-rich re-
the role of this pathway in cell migration is a matter of de- gion which is a potential binding site for SH3 domains, a
bate. Recent findings suggest that integrins may act syner- putative binding region for the focal adhesion proteins talin
gistically with receptors for extracellular growth factors.130 and vinculin and several consensus binding sites for v-Crk.157
In particular, attachment of some cells to fibronectin and Another component called p130Cas (Cas stands for Crk-as-
antibody-induced aggregation of the #5!1 integrin enhances sociated substrate) is also tyrosine phosphorylated in re-
the increase in cytoplasmic pH stimulated by PDGF.137 Such sponse to bombesin concomitantly with FAK in Swiss 3T3
cooperative interactions may underlie the well known but cells.148 P130Cas contains an SH3 domain and multiple bind-
poorly understood phenomenon of adhesion-dependent ing sites for v-Crk,158 and has been shown to associate with
proliferation. FAK.159,160
Several factors which potently stimulate stress fiber
and focal adhesion formation, including LPA, sphingosine
Signaling Mechanisms for Vascular Cell Migration 257
and bombesin, also increase tyrosine phosphorylation of receptor coupled to two G-protein pathways, one pertussis-
FAK. Recent findings have implicated FAK in the migration toxin sensitive which is implicated in activation of p21Ras,163
of other cell types.150,161 In oral squamous cell carcinoma the other pertussis toxin-insensitive leading to activation of
cells the protein tyrosine kinase inhibitor herbimycin blocked phospholipase C and tyrosine phosphorylation of FAK.164,165
the stimulation of FAK tyrosine phosphorylation and cell Consistent with the involvement of this second PLC-medi-
migration by hepatocyte growth factor.150 Intriguingly, the ated pathway, LPA-induced stress fiber formation was per-
ECM glycosaminoglycan hyaluronan has been shown to tussis toxin-insensitive.113 Though FAK lacks both src ho-
promote both the rapid assembly and disassembly of focal mology-2 (SH2) and SH3 domains,166 several recent reports
adhesions and a rapid phosphorylation-dephosphorylation indicate that FAK can associate with SH2-domain proteins.
of FAK in a c-H-ras-transformed fibroblast cell line.161 In FAK was originally identified as a prominently tyrosine phos-
VSMC and in EC, effects of several factors on FAK tyrosine phorylated protein in v-src-transformed fibroblasts,140 and
phosphorylation correlate with a chemotactic response.38,68 it has subsequently been shown that FAK associates with
Results from our laboratory show that LPA induces FAK ty- pp60c-src and pp59c-Fyn via an interaction between the SH2
rosine phosphorylation and directed migration in rabbit domain of Src-family PTKs and the major autophos-
aortic VSMC (Abedi H, Zachary I, unpublished observa- phorylation site of FAK, Y397.167 Several other SH2 domains
tions). Angiotensin II stimulates FAK and paxillin tyrosine (listed in Table 24.2) including those of the PDGF receptor
phosphorylation in rabbit VSMC (Abedi H, Zachary I, un- substrates p85#,168 GRB-2, NCK and phospholipase C-&169
published observations),89 but as already indicated it is un- have also been found to associate with FAK. Moreover, in
clear whether this factor can induce a direct migratory re- the case of the GRB-2 SH2 domain, association appears to
sponse in VSMC. PDGF-BB stimulates FAK and paxillin ty- result from the tyrosine phosphorylation of FAK at a spe-
rosine phosphorylation in VSMC through the ! receptor38,118 cific residue, Y925, induced by cell attachment to
and VEGF stimulates FAK and paxillin tyrosine phospho- fibronectin.170 Two other SH2 domain-containing proteins
rylation in human EC.68 IGF-1 has been reported to stimu- have recently been implicated in the regulation of focal ad-
late FAK and paxillin tyrosine phosphorylation concomi- hesion-associated components. The adapter protein p47Crk
tantly with lammellipodial advance in neuroblastoma associates with paxillin171 and the carboxy-terminal src ki-
cells,162 and our recent findings show that in VSMC, IGF-1 nase (Csk), which phosphorylates and negatively regulates
promotes tyrosine phosphorylation of FAK, directed migra- pp60 Src, has been shown to bind to both paxillin and
tion and an increase in immunolocalization of FAK to focal FAK.169,172 FAK also associates with SH3 domain-contain-
adhesions (Abedi H, Lobo M, Zachary I, unpublished ob- ing proteins. In platelets, FAK associates with the SH3 rather
servations). Taken together, these findings suggest that ty- than the SH2 domain of the p85 # subunit of
rosine phosphorylation of FAK might represent a conver- phosphatidylinositol 3' kinase (PI3 kinase) via a proline-rich
gent point in the signaling pathways stimulated by several sequence (residues 706-711) in the noncatalytic carboxy-
migration factors for vascular cells and other cell types. The terminal domain of FAK128.170 The same FAK motif also
role of FAK as a common target in the action of diverse fac- mediates association with p130Cas, and FAK-p130Cas com-
tors and cell surface receptors is highlighted in Figure 24.2. plexes are enriched in cytoskeleton-associated fractions in
FAK tyrosine phosphorylation may not be a common path- an adhesion-dependent manner.159,160 The associations of
way for all migration factors, however. There are, for example, FAK with other cellular components are summarized in Fig-
no reports to date that bFGF, a potent chemoattractant for ure 24.1.
EC, can regulate this pathway.
The signal transduction pathways which mediate ty- Rho-Related GTP-binding Proteins
rosine phosphorylation and presumably increased activity One of the most exciting developments in chemotac-
of FAK are unclear. LPA appears to act through a membrane tic signal transduction has been the discovery that
258 Tissue Engineering of Prosthetic Vascular Grafts
Ras-related small GTP-binding proteins of the Rho and Rac mation and cdc42 induces peripheral actin microspikes and
family are implicated in regulation of cytoskeletal organiza- filipodia.175,176
tion.115,117,173 The first evidence for the role of Rho proteins Despite the interest in Rho and other members of the
in actin filament organization came from studies of the ex- Rho/Rac family in regulating actin cytoskeleton organiza-
oenzyme C3 transferase, which is produced by the bacte- tion, there are few studies of these proteins in vascular cells,
rium Clostridium botulinum and which ADP-ribosylates Rho and consequently their role in the migration (or other func-
at residue Asn 41 and thereby inactivates it. Introduction of tions) of vascular cells remains poorly understood. A few
C3 transferase into cells causes a loss of actin stress fibers.173 studies have, however, implicated Rho in some biological
Microinjection into fibroblasts of either wild type recombi- responses in smooth muscle, including contractility and
nant Rho proteins174 or constitutively activated Rho pro- myosin light chain phosphorylation.107
teins with a valine residue instead of glycine at residue 14115 Recent findings have implicated Rho in mediating
causes a rapid formation of stress fibers and an increase in phosphorylation of FAK (Fig. 24.2). C3 transferase inhibits
the localization of the focal adhesion associated proteins FAK tyrosine phosphorylation induced either by LPA,
vinculin and talin to focal adhesions. Importantly, introduc- bombesin or by the nonhydrolyzable GTP analog
tion of either C3 transferase or of ADP-ribosylated and in- GTP&S.165,177,178 Introduction of Rho into cells has also been
activated rhoA into cells blocked the stimulation of stress shown to stimulate tyrosine phosphorylation of FAK,
fiber formation and focal adhesion assembly caused by paxillin and p130Cas.179 These findings argue strongly that
bombesin, LPA or serum.115 Recent evidence suggests that Rho is involved in the activation of FAK through some
Rho-related proteins perform specialized roles in the regu- G-protein-coupled seven transmembrane domain receptors.
lation of actin filament organization. While Rho appears to Interestingly, it was reported that the SH2/SH3 domain pro-
be important for stress fiber and focal adhesion formation, tein crk, which as mentioned above binds to paxillin, asso-
Rac promotes membrane ruffling and lammellipodia for- ciates via its SH3 domains with the guanine nucleotide ex-
Signaling Mechanisms for Vascular Cell Migration 259
change factor C3G.180 Since C3G may regulate the activity with Rho, called Rho-kinase and PKN.107 Rho-kinase has
of Ras proteins, this finding suggests that other small GTP- been reported to phosphorylate MLC in vitro.182 The pri-
binding proteins may participate in the regulation of FAK mary phosphorylation site for Rho-kinase, Ser19, is identi-
and associated proteins in the focal adhesion complex. cal to the major phosphorylation site utilized by MLC ki-
Very little is known about the role of Rho family pro- nase,183 and phosphorylation of this residue is required to
teins in the activation of the FAK pathway through receptor enable actin activation of the myosin ATPase.184 Evidence
tyrosine kinases. The recent finding that PDGF-BB stimu- supporting a direct role for Rho-kinase in stress fiber and
lates an increase in the GTP-bound form of Rac, and that focal adhesion formation has come from microinjection
Rac activation is inhibited by the PI3 kinase inhibitor studies. 185 Rho has also been reported to stimulate
wortmannin,181 suggests that this small GTP-binding pro- phosphatidylinositol 4-phosphate 5-kinase (PI4P5K) which
tein may play a role in mediating PDGF stimulation of the catalyzes formation of PIP2 and promotes actin polymer-
FAK pathway (Fig. 24.2). Regardless of the precise sequence ization (see below). The central role of Rho in signaling path-
of interactions involved, it is likely that versatile proteins ways regulating actin cytoskeletal organization is illustrated
containing SH2 and SH3 modules will provide the crucial in Figure 24.3.
mediating links between the PDGF receptor, GTP-binding
proteins of the Rho family and FAK. Receptor Protein Tyrosine Kinase Signaling
How Rho is itself activated by these (or indeed any Given the importance of ligands for receptor PTKs in
other) receptors is unclear, but recent progress has been made the regulation of cell migration, it will be helpful to give a
towards the identification of candidate downstream effec- brief general discussion of some of the principal themes
tors which mediate the actions of Rho. Two serine/threo- which have emerged in studies of receptor PTK activation
nine protein kinases have been identified which associate and signal transmission. The PDGF receptor will serve as a
paradigm. The receptor(s) for PDGF consists of an extra- through the tyrosine phosphorylation and activation of
cellular ligand-binding domain, a single membrane-span- PLC-&. PIP2 binds to the actin-binding proteins profilin and
ning region and a cytoplasmic domain with intrinsic PTK gelsolin. and it has been proposed that PIP2 can modulate
activity. Binding of PDGF in the form of homo- or actin filament formation through the sequestration of ac-
heterodimers of A and B chains induces dimerization of the tin-binding proteins like profilin and gelsolin. Anti-sense
receptor and subsequent receptor autophosphorylation. oligonucleotides to gelsolin and expression of a peptide
Autophosphorylation is thought to occur mainly by which competes for the PIP2-binding site on gelsolin both
transphosphorylation of each dimerized receptor molecule reduced epidermal growth factor-induced cell movement in
by its partner. Homodimers of # and ! receptors as well as NR6 cells.195 In platelets, gelsolin-derived PIP2-binding pep-
#/! heterodimers have all been identified, but as indicated tides also inhibited uncapping of F-actin induced by throm-
above the relative abundance of a given receptor dimer is bin receptor activation.196
highly dependent on cell-type and species. Dimerization and PIP2 has also been reported to bind to vinculin and
receptor autophosphorylation are essential for the activation unmask its talin and actin binding sites, thereby promoting
of the receptor.186 Subsequently, the receptor associates with focal adhesion and stress fiber formation.197 Profilin bind-
and tyrosine phosphorylates its intracellular targets via SH2 ing to PIP2, the substrate of PLC-&, inhibits PLC-& activity
domain interactions with specific tyrosine phosphorylated and this inhibition is overcome by tyrosine phosphoryla-
residues in the cytoplasmic domain of the receptor.186,187 tion. By effectively lowering the concentration of PIP2, acti-
Though an increasing number of the cellular compo- vation of PLC-& can potentially promote actin filament dis-
nents which are phosphorylated by the PDGF ! receptor have assembly both by raising the level of free profilin and gelsolin
been identified,186,187 it remains unclear which components and by reducing its vinculin unmasking activity (Fig. 24.4).
or intracellular signaling pathways specifically mediate the As indicated above, however, the role of profilin may be more
effects of PDGF-BB on cell migration and locomotion. In complex. Another consequence of PIP2 hydrolysis is the for-
addition, most studies of PDGF Receptor signal transduc- mation of inositol 1,4,5 trisphosphate, which in turn leads
tion have been performed on the ! receptor, which as dis- to the mobilization of intracellular Ca2+. Ca2+ also affects
cussed above is able to transduce mitogenic and chemotac- the equilibrium between actin polymerization and depoly-
tic signals in a wide variety of cells. In contrast, # receptor merization and also activates calcium-dependent enzymes
signaling has not been intensively examined. As a result, little such as Ca2+/calmodulin-dependent protein kinase II and
is known concerning either cell type-specific or receptor the Ca 2+ /calmodulin-activated protein phosphatase
isotype-specific differences in PDGF signal transduction.187 calcineurin, both of which have been implicated in the regu-
The main features of the activation mechanism that lation of VSMC migration (see Fig. 24.4).198,199
have been identified for the PDGF receptor have also been PIP2 levels can be increased in cells through the acti-
demonstrated for other receptor PTKs. In some cases, how- vation of phosphatidylinositol 4-phosphate 5-kinase
ever, the signal transduction mechanisms are not yet par- (PI4P5K). Overexpression of PI4P5K in COS-7 cells causes
ticularly well understood.188-190 The VEGF receptor will serve a striking increase in actin polymerization.200 This kinase
as an example. The diverse biological effects of VEGF are has been shown to be regulated by Rho,201 thus providing a
mediated through two specific PTK receptors, KDR/Flk-1 link with the pathway thought to be responsible for activa-
and Flt-1. Both receptor isotypes are expressed in ECs67 and tion of FAK by bombesin and LPA and potentially of other
Flt-1 is expressed in monocytes.71 Expression of VEGF re- factors which act through seven transmembrane domain,
ceptors has to date only been reported in these two cell types. G-protein coupled receptors. It is not known which extra-
KDR/Flk-1 is thought to be the receptor largely responsible cellular factors are responsible for activating PI4P5K, but it
for the angiogenic effects of VEGF and its mitogenic effects may be speculated that activation of this kinase and the con-
in EC.191,192 The signal transduction pathways which medi- sequent increase in cellular levels of PIP2 may contribute to
ate the diverse biological effects of VEGF either in EC or in the striking increase in actin filament and focal adhesion
monocytes remain unclear. Studies of VEGF signal trans- formation caused by bombesin, LPA and other factors which
duction have so far produced varying results.67,68,193,194 In activate FAK through a Rho-dependent pathway. Integrin
porcine aortic EC which have been transfected either with engagement has also been reported to activate PI4P5K.202
KDR or Flt-1, VEGF stimulated migration and proliferation
but had little effect on tyrosine phosphorylation of phos- PI3 Kinase
pholipase C-& (PLC-&), p120 GTPase-activating protein Activation of PI3 kinase is an early event in the action
(GAP) or on phosphatidylinositol 3 kinase (PI3 kinase) ac- of several receptor PTK ligands, including PDGF-BB and
tivity.67 In contrast, VEGF has been reported to stimulate IGF-1. Recent findings have implicated PI3 kinase in PDGF-
tyrosine phosphorylation of p120GAP, PLC-& and p85# sub- induced chemotaxis.203-205 PDGF-BB failed to stimulate
unit of PI3 kinase in bovine aortic EC and NIH 3T3.193,194 migration in Chinese hamster ovary cells expressing mu-
Our recent findings show, however, that in HUVEC, VEGF tant forms of the PDGFR! which are unable to associate with
activates MAP kinase and stimulates tyrosine phosphoryla- the p85# subunit of PI3 kinase.203 In addition, p85#/PI3 ki-
tion of PLC-&, FAK and paxillin.68 nase has also been implicated in membrane ruffling induced
by PDGF204 and IGF-1.205 PI3 kinase has also been impli-
Phosphatidylinositol Lipids and Calcium cated in mediating PDGF-stimulated FAK tyrosine phos-
Many chemoattractants for vascular cells, including phorylation. In Swiss 3T3 cells, inhibitors of PI3 kinase
PDGF, IGF-1, bFGF and VEGF, stimulate hydrolysis of PIP2 blocked PDGF-induced FAK and paxillin tyrosine phospho-
Signaling Mechanisms for Vascular Cell Migration 261
rylation without affecting stimulation by bombesin. The sig- The mechanisms by which products of arachidonic acid
nificance of these findings for cell migration are unclear, metabolism stimulate chemotaxis in vascular cells are at
especially since PDGF causes a profound inhibition of FAK present not well understood.
tyrosine phosphorylation, migration and actin cytoskeleton
organization at concentrations higher than 10 ng/ml in the Nitric Oxide
same cells. Our own studies in rabbit aortic VSMC indicate Recent evidence suggests that NO may play a role in
that the PI3 kinase inhibitors wortmannin and LY294002 at angiogenesis and in the migration of both epithelial cells
concentrations which inhibit PDGF-BB-stimulation of PI3 and EC.211-213 In particular, NO production has been shown
kinase activity do not impair either the migratory response to be permissive for endothelin-induced migration of
to PDGF-BB or PDGF-BB-induced FAK tyrosine HUVECs.213 These findings indicate that NO production is
phosphorylation (Abedi H, Cospedal R, Zachary I, manu- at least permissive for EC migration in response to factors
script submitted). such as endothelin, but the mechanism through which the
NO pathway could act is unknown. Other EC migration fac-
Arachidonic Acid Metabolism tors may also act through the NO pathway. Our recent find-
Studies in several cell types implicate arachidonic acid ings show that VEGF is able to induce NO production in EC
and products of its metabolism in chemotactic signaling. (Zachary I, unpublished findings).
The best evidence so far is for a role of the products of the
lipoxygenase pathway in the migration of monocytes and The MAP Kinase Pathway
eosinophils. 5-oxo-15-HETE is a potent chemoattractant for Recent findings have established that the engagement
eosinophils,206,207 and other lipoxygenase products, lipoxin of integrins can activate the MAP kinase pathway.134-136 The
A4 and lipoxin B4, have been shown to stimulate chemot- role of integrin-mediated stimulation of this pathway in
axis of human monocytes.208 Other studies have implicated cytoskeletal changes is unclear, however. Thus, Clark and
the cytosolic phospholipase A2, thought to catalyze the ago- Hynes showed that while adhesion-dependent activation of
nist-driven release of arachidonic acid, in migration of Erk-2 is dependent upon Ras, inhibition of Ras signaling by
monocytes in response to the chemokines RANTES and expression of a dominant negative Ras mutant (N17Ras)
MCP-1,209 and in migration of EC stimulated by bFGF.210 blocked Erk-2 activation but had no effect either on FAK
262 Tissue Engineering of Prosthetic Vascular Grafts
tyrosine phosphorylation or on cell adhesion, spreading, of a common signaling network relaying information be-
focal adhesion formation or stress fiber assembly.136 Other tween the cell surface and the cytoskeleton. More precise
investigators have provided evidence that integrin activa- delineation of the interactions between the FAK and Rho
tion of Erks is mediated through Rho rather than Ras.202 pathways and of both with pathways involving proteins con-
Integrins failed to activate Erk-1/2 in the absence of Rho/ taining SH2 and SH3 domains, is likely to yield important
Rac activity, and integrin-dependent MAP kinase activity new insights into the mechanisms underlying the regula-
was blocked by C3 exoenzyme and was enhanced by expres- tion of vascular cell migration.
sion of a constitutively active Rho mutant (RhoQ63L).214 In the case of the most potent known VSMC migra-
Though some chemoattractants for VSMC and EC, includ- tion factor, PDGF-BB, the migratory response appears to be
ing PDGF-BB, activate the MAP kinase cascade, other fac- determined by a complex interplay between surface expres-
tors such as IGF-1 stimulate chemotaxis of human VSMC sion of receptor isotypes, the bioavailability of ligand
without activation of MAP kinase.37 These findings suggest isoforms and other as yet unidentified cell type specific de-
that the MAP kinase pathway is at least not obligatory for terminants. Identification of the differences in signaling in
VSMC migration, and it has been proposed that mitogenic different cells via # and ! receptor is likely to be a crucial
and chemotactic signaling pathways can be fairly sharply prerequisite for understanding the mechanisms underlying
dissociated.37,215 A degree of functional segregation of sig- the PDGF chemotactic response. It is plausible that alter-
nal transduction mechanisms underlying these processes is ations in the VSMC signaling pathways induced by PDGF
attractive, especially since the same factor (e.g., PDGF-BB) and associated changes in biological responsiveness, medi-
is often able to stimulate both mitogenesis and migration. It ated in part by changes either in autocrine/paracrine ligand
is possible, however, that MAP kinases may cooperate with production and/or receptor isotype expression, could also
other signaling pathways to augment a migratory response. have important implications for our understanding of the
Unpublished results from our laboratory show that a selec- mechanisms underlying the long term pathogenesis of ath-
tive inhibitor, PD98059, of MAP kinase kinase, the enzyme erosclerosis and other vascular pathologies. Whether a simi-
which specifically phosphorylates and activates Erk 1 and 2, lar complexity underlies the chemotactic effects of other fac-
caused partial inhibition of PDGF-BB-stimulated migration tors such as bFGF, IGF-1 and VEGF awaits further study.
without reducing FAK tyrosine phosphorylation. Stimula- A further challenge, with particular relevance to the
tion of rat VSMC migration by PDGF-BB has also been vessel wall, is posed by the diversity of extracellular factors
shown to be partially inhibitable by PD98059.216 Since IGF-1 which may regulate vascular cell migration. It is probable
is a less potent chemoattractant for VSMC than PDGF-BB, that this diversity will grow and that the repertory of VSMC
it might be speculated that activation of MAP kinase by the migration factors will be extended in the future both by the
latter factor could help to explain this difference. This pos- discovery of novel regulatory molecules and by identifying
sibility remains to be tested. new functions for existing components of the hemostatic
system. The dependence of VSMC migration on the com-
Future Perspectives plex interplay between diverse environmental factors has
Elucidation of the signal transduction mechanisms important ramifications for chemotactic signal transduction.
underlying the movement of cells remains a major unre- The finding that FAK is a point of convergence in signaling
solved problem in molecular cell biology. In the past few mediated both by integrins and by several receptor PTKs
years, however, our understanding of these mechanisms has raises the possibility that, in part at least, chemotactic sig-
been transformed by the identification of a variety of com- naling mechanisms through distinct receptors may share
ponents which are implicated in key cellular processes linked common features. It is equally likely that important differ-
to cell locomotion, including assembly of focal adhesions ences also exist creating scope for complementary, crosstalk
and the formation of stress fibers. Neither the precise role and synergistic interactions between different receptor-me-
played by these components in the molecular and cellular diated pathways. Establishing the relationships between the
sequelae leading to cell migration nor the relationships and pathways regulated by integrins, receptor PTKs, and other
interactions between them have been defined as yet. Future receptor types will be crucial for understanding the molecu-
effort will also need to address the relevance of putative lar basis of the migratory response in the vessel wall. The
chemotactic signaling pathways both for the action of spe- roles of metalloproteinases and the plasminogen activator
cific migration factors and for the migration of vascular cells system are likely to prove of particular importance for vas-
such as VSMC and EC. cular cell migration in vivo.
This chapter has highlighted recent work suggesting Paracrine interactions between VSMC, EC, monocytes
that FAK and other focal adhesion-associated molecules may and other cells in the atherosclerotic microenvironment adds
play a role in vascular cell migration in response to growth a further layer of complexity to the regulation of migration
factor ligands for receptor PTKs. The latter notion is espe- of vascular cells. The production of PDGF by EC in response
cially attractive in the light of recent findings that FAK has to monocyte-, platelet- and T lymphocyte-derived cytokines,
the ability to associate both with components of focal adhe- and the induction in VSMC of VEGF, an angiogenic factor
sions and with certain SH2 domain-containing substrates whose target cells are monocytes and EC, are two striking
of the PDGF receptor. The role of small GTP-binding pro- examples of such interactions. The production of VEGF by
teins of the Rho family has also been emphasized, as has the VSMC and the role of this factor in promoting re-
possibility that FAK and Rho proteins may be constituents endothelialization may have a special relevance for tissue
Signaling Mechanisms for Vascular Cell Migration 263
engineering of prosthetic grafts. The use of VEGF gene trans- 10. Wilcox JN, Smith KM, Williams LT et al. Platelet derived
fer for accelerating endothelial regrowth following balloon growth factor mRNA detection in human atherosclerotic
injury raises the possibility of using a similar approach to plaques by in situ hybridisation. J Clin Invest 1988;
promote endothelial growth over prosthetic grafts. 82:1134-1143.
11. Majesky MW, Reidy MA, Bowen-Pope DF et al. Role of
Studies of the mechanisms regulating vascular cell
PDGF-A expression in the control of vascular smooth
migration have considerable implications for the clinical muscle cell growth by TGF- ! . J Cell Biol 1990;
treatment of cardiovascular disease. One potentially impor- 111:2149-2158.
tant inference to be drawn from the experience of bypass 12. Golden MA, Au YPT, Kirkman TR et al. Platelet derived
graft surgery and the use of prosthetic grafts is that combi- growth factor activity and mRNA expression in healing
natorial therapeutic strategies aimed at either suppressing vascular grafts in baboons. J Clin Invest 1991; 87:406-414.
the migration of VSMC leading to stenosis, and/or at en- 13. Jawien A, Bowen-Pope DF, Lindner V et al. Platelet de-
hancing the migration of EC and thus promoting more ef- rived growth factor promotes smooth muscle migration
fective re-endothelialization, may both constitute rational and intimal thickening in a rat model of balloon
approaches for the treatment of the vasculoproliferative angioplasty. J Clin Invest 1992; 89:507-511.
complications of arterial bypass grafting. Elucidation of the 14. Ferns GAA, Raines EW, Sprugel KH et al. Inhibition of
neointimal smooth muscle accumulation after angioplasty
molecular mechanisms underlying vascular cell migration
by an antibody to PDGF. Science 1991; 253:1129-1132.
and the identification of cell type-specific migration factors 15. Stroobant P, Waterfield MD. Purification and properties
will be of crucial importance in effectively targeting such of platelet-derived growth factor. EMBO J 1984;
therapies. The rapidly growing knowledge of signaling net- 3:2963-2967.
works, together with the ability to isolate specific compo- 16. Hart CE, Bailey M, Curtis DA et al. Purification of PDGF-
nents and model component-specific agents, make the tar- AB and PDGF-BB from human platelet extracts and iden-
geting of specific signal transduction components or path- tification of all three platelet derived growth factor dimers
ways for the treatment of human disease increasingly an at- in human platelets. Biochemistry 1990; 29:166-172.
tainable goal. Understanding the mechanisms involved in 17. Hammacher A, Hellman U, Johnsson A et al. A major
vascular cell migration will therefore be of considerable value part of the platelet derived growth factor purified from
human platelets is one A and one B chain. J Biol Chem
both in identifying novel targets for the design of new use-
1988; 263:16493-16498.
ful drugs, and for opening up novel perspectives for treat- 18. Kavanaugh WM, Harsh GR, Starksen NF et al.
ment of vasculoproliferative disorders. Transcriptiocgulation of the A and B chain genes of plate-
let-derived growth factor in microvascular endothelial
Acknowledgments cells. J Biol Chem 1988; 263:8470-8472.
The work on the regulation of FAK tyrosine phospho- 19. Daniel TO, Gibbs VC, Milfay DF et al. Agents that in-
rylation in VSMC is supported by the British Heart Foun- crease cAMP accumulation block endothelial c-sis induc-
dation. Dr. Ian Zachary is a BHF (Basic Science) Lecturer. tion by thrombin and transformtin growth factor-!. J Biol
Chem 1987; 262:11893-11896
References 20. Starksen NF, Harsh GR, Gibbs VC et al. Regulated ex-
1. Ross R. The pathogenesis of atherosclerosis: A perspec- pression of the PDGF A chain gene in microvascular en-
tive for the 1990’s. Nature 1993; 362:801-809. dothelial cells. J Biol Chem 1987; 262:14381-14384.
2. Ross R. Cell biology of atherosclerosis. Annu Rev Physiol 21. Newby AC, Henderson AH. Stimulus-secretion coupling
1995; 57:791-804. in vascular endothelial cells. Annu Rev Physiol 1990;
3. Wells N. Coronary heart Disease: The need for action. 52:661-674.
London: Office of Health Economics, 1987. 22. Sejersen T, Betsholtz C, Sjolund M et al. Rat skeletal myo-
4. Wolfe JHN, Harris PL, Vaughan Ruckley C. Trust Hospi- blasts and arterial smooth muscle cells express the gene
tals and vascular Services. Br Med J 1994; 309:414-419. for the A chain but not the gene for the B chain (c-sis)
5. Hillegass WB, Ohman EM, Califf RM. Restenosis: The of platelet-derived growth factor (PDGF) and produce a
clinical issues. In: Topol EJ, ed. Textbook of Interventional PDGF-like protein. Proc Natl Acad Sci USA 1986;
Cardiology. Philadelphia USA: W.B. Saunders Co., 83:6844-6848.
1994:415-435. 23. Majesky MW, Benditt EP, Schwartz SM. Expression and
6. Hertzer NR, Bevan EG, Young JR et al. Coronary disease developmental control of PDGF-A chain and B chain/sis
in vascular patients. A Classification of 1000 coronary genes in rat aortic smooth muscle cells. Proc Natl Acad
angiograms and the results of surgical management. Ann Sci USA 1988; 85:1524-1528.
Surg 1984; 199:223-233. 24. Majack RA, Majesky MW, Goodman LV. Role of PDGF-
7. Schwartz SM, Campbell GR, Campbell JH. Replication of A expression in the control of vascular smooth muscle
Smooth Muscle Cells in Vascular Disease. Circ Res 1986; cell growth by TGF-!. J Cell Biol 1990; 111:239-247.
58:427-444. 25. Battegay EJ, Raines EW, Seifert RA et al. TGF-! induces
8. Pasic M, Muller-Glauser W, Odermatt B et al. Seeding bimodal proliferation of connective tissue cells via com-
with omental cells prevents late neointimal hyperplasia in plex control of an autocrine PDGF loop. Cell 1990;
small-diameter Dacron grafts. Circulation 1995; 63:515-524.
92:2605-2616. 26. Raines E, Dower S, Ross R. Interleukin-1 mitogenic ac-
9. Zilla P, Preiss P, Groscurth P et al. In vitro-lined endot- tivity for fibroblasts and smooth muscle cells is due to
helium: Initial integrity and ultrastructural events. Sur- PDGF-AA. Science 1989; 243:393-396.
gery 1994; 116:524-534. 27. Nilsson J. Cytokines and smooth muscle cells in athero-
sclerosis. Cardiovasc Res 1993; 27:1184-1190.
264 Tissue Engineering of Prosthetic Vascular Grafts
28. Ross R, Masuda J, Raines EW et al. Localization of PDGF- 45. Terranova VP, DiFloria R, Lyall R et al. Human EC are
B protein in macrophages in all phases of atherogenesis. chemotactic to EC growth factor and heparin. J Cell Biol
Science 1990; 248:1009-1012. 1985; 101:2330-2334.
29. Shimokado K, Raines EW, Madtes DK et al Significant 46. Sato Y, Rifkin DB. Inhibition of EC movement by
part of macrophage-derived growth factor consists of at pericytes and smooth muscle cells: Activation of a latent
least two forms of PDGF. Cell 1985; 43:277-286. transformtin growth factor-!-like molecule by plasmin
30. Bowen-Pope DF, Hart CE, Seifert RA. Sera and condi- during coculture. J Cell Biol 1988; 107:1199-1205.
tioned media contain different isoforms of platelet-derived 47. Tsuboi R, Sato Y, Rifkin DB. Correlation of cell migra-
growth factor which bind to different classes of PDGF tion, cell invasion, receptor number, proteinase produc-
receptor. J Biol Chem 1989; 264:2502-2508. tion, and basic fibroblast growth factor levels in EC. J Cell
31. Hart CE, Forstrom JW, Kelly JD et al. Two classes of Biol 1990; 110:511-517.
PDGF receptor recognize different isoforms of platelet 48. Lindner V, Majack RA, Reidy MA. Basic fibroblast growth
derived growth factor. Science 1988; 240:1529-1531. factor stimulates endothelial regrowth and proliferation
32. Gronwald RGK, Grant FJ, Haldeman BA et al. Cloning in denuded arteries. J Clin Invest 1990; 85:2004-2008.
and expression of a cDNA coding for the human plate- 49. Bjornsson TD, Dryjski M, Tluczek J et al. Acidic fibro-
let-derived growth factor receptor. Evidence for more than blast growth factor promotes vascular repair. Proc Natl
one receptor class. Proc Natl Acad Sci USA 1988; Acad Sci USA 1991; 88:8651-8655.
85:3435-3439. 50. Koyama N, Harada K, Yamamoto A et al. Purification and
33. Seifert RA, Hart CE, Phillips PE et al. Two different sub- characterisation of an autocrine migration factor for vas-
units associate to create isoform-specific PDGF receptors. cular smooth muscle cells (SMC), SMC-derived migra-
J Biol Chem 1989; 264:8771-8778. tion factor. J Biol Chem 1993; 268:13301-13308.
34. Koyama N, Hart CE, Clowes, AW. Different functions of 51. Ferrara N, Houck KA, Jakeman LB et al. Molecular and
the PDGF # and ! receptors for the migration and pro- biological properties of the vascular endothelial growth
liferation of cultured baboon smooth muscle cells. Circ factor family of proteins. Endocrine Rev 1992; 13:18-32.
Res 1994; 75:682-691. 52. Plate KH, Breier G, Weich HA et al. Vascular endothelial
35. Siegbahn A, Hammacher A, Westermark B et al. Differ- growth factor is a potential tumor angiogenesis factor in
ential effects of the various isoforms of PDGF in chemo- human gliomas in vivo. Nature 1992; 359:845-848.
taxis of fibroblasts, monocytes and granulocytes. J Clin 53. Park JE, Chen HH, Winer J et al. Placenta growth factor:
Invest 1990; 85:916-920. Potentiation of vascular endothelial growth factor bioac-
36. Koyama N, Morisaki N, Saito Y et al. Regulatory effects tivity, in vitro and in vivo, and high-affinity binding to
of PDGF-AA homodimer on migration of vascular smooth Flt-1 but not to Flk-1/KDR. J Biol Chem 1994;
muscle cells. J Biol Chem 1992; 267:22806-22812. 269:25646-25654.
37. Bornfeldt KE, Raines EW, Nakano T et al. Insulin-like 54. Olofsson B, Pajusola K, Kaipainen A et al. Vascular en-
growth factor-I and platelet-derived growth factor-BB in- dothelial growth factor B, a novel growth factor for EC.
duce directed directed migration of human arterial smooth Proc Natl Acad Sci USA 1996; 93:2576-2581.
muscle cells via signalling pathways that are distinct from 55. Joukov V, Pajusola K, Kaipainen A et al. A novel vascu-
those of proliferation. J Clin Invest 1994; 93:1266-1274. lar endothelial growth factor, VEGF-C, is a ligand for the
38. Abedi H Dawes KE, Zachary I. Differential effects of plate- Flt-4 (VEGFR-3) and KDR (VEGFR-2) receptor tyr ki-
let-derived growth factor BB on p125 focal adhesion ki- nases. EMBO J 1996; 15:290-298.
nase and paxillin tyrosine phosphorylation and on cell 56. Stavri G, Zachary I, Baskerville P et al. Basic fibroblast
migration in rabbit aortic vascular smooth muscle cells growth factor upregulates the expression of vascular en-
and Swiss 3T3 fibroblasts. J Biol Chem 1995; dothelial growth factor in vascular smooth muscle cells.
270:11367-11376. Circulation 1995; 91:5-8.
39. Ferns GA, Sprugel KH, Seifert RA et al. Relative platelet- 57. Stavri G, Hong Y, Zachary I et al. Hypoxia and platelet-
derived growth factor receptor subunit expression deter- derived growth factor-BB synergistically upregulate the
mines migration to different dimeric forms of PDGF expression of vascular endothelial growth factor in vas-
growth factors. 1990; 3:315-324. cular smooth muscle cells. FEBS Lett 1995; 358:311-315.
40. Higashiyama S, Abraham JA, Klagsbrun M. Heparin-bind- 58. Brogi E, Wu T, Namiki A et al. Indirect angiogenic
ing EGF-like growth factor stimulation of smooth muscle cytokines upregulate VEGF and bFGF gene expression in
cell migration: Dependence on interactions with cell sur- vascular smooth muscle cells, whereas hypoxia upregulates
face heparan sulfate. J Cell Biol 1993; 122:933-940. VEGF expression only. Circulation 1994; 90:649-652.
41. Delafontaine P, Bernstein KE, Alexander RW. Insulin-like 59. Liu Y, Cox SR, Morita T et al. Hypoxia regulates vascular
growth factor I gene expression in vascular cells. Hyper- endothelial growth factor gene expression in EC. Identi-
tension 1991; 17:693-699. fication of a 5' enhancer. Circ Res 1995; 77:638-643.
42. Newby AC, George SJ. Proposed roles for growth factors 60. Kourembanas S, Bernfield M. Hypoxia and endothelial-
in mediating smooth muscle proliferation in vascular pa- smooth muscle cell interactions in the lung. Am J Respir
thologies. Cardiovasc Res 1993; 27:1173-1183. Cell Mol Biol 1994; 11:373-374.
43. Cercek B, Fishbein MC, Forrester JS et al. Induction of 61. Folkman J. Angiogenesis in cancer, vascular, rheumatoid
insulin-like growth factor I messenger RNA in rat aorta and other disease. Nature Med 1995; 1:27-31.
after balloon denudation. Circ Res 1990; 66:1755-1760. 62. Risau W. Mechanisms of angiogenesis. Nature 1997;
44. Jackson CL, Reidy MA. Basic fibroblast growth factor: Its 386:671-674.
role in the control of smooth muscle cell migration. Amer 63. Asahara T, Bauters C, Pastore C. et al. Local delivery of
J Pathol 1993; 143:1024-1031. vascular endothelial growth factor accelerates
reendothelialization and attenuates intimal hyperplasia in
Signaling Mechanisms for Vascular Cell Migration 265
balloon-injured rat carotid artery. Circulation 1995; 81. Assoian RK, Sporn MB. Type beta transforming growth
91:2793-2801. factor in human platelets: release during platelet degranu-
64. Bauters C, Asahara T, Zheng LP et al. Recovery of dis- lation and action on vascular smooth muscle cells. J Cell
turbed endothelium-dependent flow in the collateral-per- Biol 1986; 102:1217-1223.
fused rabbit ischemic hindlimb after administration of 82. Koyama N, Koshikawa T, Morisaki N et al. Bifunctional
vascular endothelial growth factor. Circulation 1995; effects of transformtin growth factor-! on migration of
91:2802-2809. cultured rat aortic smooth muscle cells. Biochem Biophys
65. Isner JM, Pieczek A, Schainfeld R, Blair R, Haley L, Res Commun 1990; 169:725-729.
Asahara T, Rosenfield K, Razvi S, Walsh K, Symes JF. 83. Nomoto A, Mutoh S, Hagihara H et al. Smooth muscle
Clinical evidence of angiogenesis after arterial gene trans- cell-migration induced by inflammatory cell products and
fer of phVEGF165 in patient with ischemic limb. Lancet its inhibition by a potent calcium antagonist, Nilvadipine.
1996; 348:370-374. Atherosclerosis 1988; 72:213-219.
66. Asahara T, Chen D, Tsurum, Y et al. Accelerated restitu- 84. Yu X, Dluz S, Graves DT, Zhang L, Antoniades HN, Hol-
tion of endothelial integrity and endothelial-dependent lander W, Prusty S, Valente AJ, Schwartz CJ, Sonenshein
function after phVEGF165 gene transfer. Circulation 1996; GE. Elevated expression of monocyte chemoattractant
94:3291-3302. protein 1 by vascular smooth muscle cells in hyper-
67. Waltenberger J, Claesson-Welsh L, Siegbahn A et al. Dif- cholesterolemic primates. Proc Natl Acad Sci USA 1992;
ferent signal transduction properties of KDR and Flt1, two 89:6953-6957.
receptors for vascular endothelial growth factor. J Biol 85. Massague J. The transformtin growth factor-! family.
Chem 1994; 269:26988-26995. Annu Rev Cell Biol 1990; 6:597-641.
68. Abedi H, Zachary I. Vascular endothelial growth factor 86. Sato Y, Rifkin DB. Inhibition of endothelial cell move-
stimulates tyrosine phosphorylation and recruitment to ment by pericytes and smooth muscle cells: Activation of
new focal adhesions of focal adhesion kinase and paxillin a latent transformtin growth factor-!1-like molecule by
in EC. J Biol Chem 1997; 272:15442-15451. plasmin during coculture. J Cell Biol 1989; 109:309-315.
69. Connolly DT, Olander JV, Heuvelman D et al. Human 87. Singer SJ, Kupfer A. The directed migration of eukary-
vascular permeability factor. Isolation from U937 cells. J otic cells. Annu Rev Cell Biol 1986; 2:337-365.
Biol Chem 1989; 264:20017-20024. 88. Devroetes PN, Zigmund SH. Chemotaxis in eukaryotic
70. Clauss M, Gerlach H, Brett J et al. Vascular permeability cells: A focus on leukocytes and Dictyostelium. Annu Rev
factor: A tumor-derived poypeptide that induces endot- Cell Biol 1988; 4:649-686.
helial cell and monocyte procoagulant activity and mono- 89. Koyama N, Koshikawa T, Morisaki N et al. Secretion of a
cyte migration. J Exp Med 1990; 172:1535-1545. potent new migration factor for smooth muscle cells
71. Clauss M, Weich H, Breier G et al. The vascular endot- (SMC) by cultured SMC. Atherosclerosis 1991;
helial growth factor receptor Flt-1 mediates biological ac- 86:219-226.
tivities. J Biol Chem 1996; 271:17629-17634. 90. Naito M, Hayasi T, Funaki C et al. Vitronectin-induced
72. Barger AC, Beeuwkes R, Lainey LL et al. Hypothesis: Vasa haptotaxis of vascular smooth muscle cells in vitro. Exp
vasorum and neo-vascularisation of human coronary ar- Cell Res 1991; 194:154-156.
teries. Possible role in the pathophysiology of atheroscle- 91. Giachelli CM, Bae N, Almeida M et al. Osteopontin is
rosis. N Eng J Med 1984; 310:175-178. elevated during neointima formation in rat arteries and
73. Geiringer E. Intimal vascularisation and atherosclerosis. J is a novel component of human atherosclerotic plaques.
Path Bacteriol 1951; 63:201-211. J Clin Invest 1993; 92:1686-1696.
74. O’Brien ER, Garvin MR, Dev R et al. Angiogenesis in hu- 92. Shanahan CM, Weissberg PL, Metcalfe JC. Isolation of
man coronary atherosclerotic plaques. Amer J Pathol 1994; gene markers of differentiated and proliferating vascular
145:883-894. smooth muscle cells. Circ Res 1991; 73:193-204.
75. Bell L, Madri, JA. Influence of the angiotensin system on 93. Liaw L, Almeida M, Hart CE et al. Osteopontin promotes
endothelial and smooth muscle cell migration. Amer J vascular cell adhesion and spreading and is chemotactic
Pathol 1990; 137:7-12. for smooth muscle cells in vitro. Circ Res 1994;
76. Mii S, Ware JA, Mallette SA et al. Effect of angiotensin II 74:214-224.
on human vascular smooth muscle cell growth. J Surg Res 94. Yue TL, Mckenna RJ, Ohlstein EH et al. Osteopontin-
1994; 57:174-178. stimulated vascular smooth muscle cell migration is me-
77. Daemen MJAP, Lombardi DM, Bosman FT et al. An- diated by !3 integrin. Exp Cell Res 1994; 214:459-464.
giotensin II induces smooth muscle cell proliferation in 95. Liau G, Chan LM. Regulation of extracellular matrix RNA
the normal and injured rat arterial wall. Circ Res.1991; levels in cultured smooth muscle cells. Relationship to
68:450-456. cellular quiescence. J Biol Chem 1989; 264:10315-10320.
78. Prescott MF, Webb RL, Reidy MA. Angiotensin-convert- 96. Nugent MA, Lane EA, Keski-Oja J et al. Growth stimula-
ing enzyme inhibitor versus angiotensin II, AT1 receptor tion, altered regulation of epidermal growth factor recep-
antagonist: Effects on smooth muscle cell migration and tors and autocrine transformation of spontaneously trans-
proliferation after balloon catheter injury. Amer Pathol formed normal rat kidney cells by transformtin growth
1991; 139:1291-1296. factor-beta. Cancer Res 1989; 49:3884-3890.
79. Assoian RK, Fleurdelys BE, Stevenson HC et al. Expres- 97. Lawrence R, Hartmann DJ, Sonenshein GE. Transform-
sion and secretion of type beta transforming growth fac- ing growth factor !1 stimulates type V collagen expres-
tor by activated human macrophages. Proc Natl Acad Sci sion in bovine vascular smooth muscle cells. J Biol Chem
USA 1987; 84:6020-6024. 1994; 269:9603-9609.
80. Majesky MW, Lindner V, Twardzik DR et al. Production 98. DiMilla PA, Stone JA, Quinn JA et al. Maximal migra-
of transforming growth factor beta 1 during repair of ar- tion of human smooth muscle cells on fibronectin and
terial injury. J Clin Invest 1991; 88:904-910.
266 Tissue Engineering of Prosthetic Vascular Grafts
type IV collagen occurs at an intermediate attachment 115. Ridley. A, Hall A. The small GTP-binding protein rho
strength. J Cell Biol 1993; 122:729-737. regulates the assembly of focal adhesions and actin stress
99. Southgate KM, Davies M, Booth RFG et al. Involvement fibers. Cell 1992; 70:389-399.
of extracellular-matrix-degrading metalloproteinases in 116. Bar-Sagi D, Feramisco JR. Induction of membrane ruf-
rabbit aortic smooth muscle cell proliferation. Biochem J fling and fluid-phase pinocytosis in quiescent fibroblasts
1992; 288:93-99. by ras proteins. Science 1986; 233:1061-1068.
100. Southgate KM, Fisher M, Banning AP et al. Upregulation 117. Ridley A, Paterson HF, Johnston C L et al. The small
of basement membrane-degrading metalloproteinase se- GTP-binding protein rac regulates growth factor-induced
cretion after balloon injury of pig carotid arteries. Circ membrane ruffling. Cell 1992; 70:401-410.
Res 1996; 79:1177-1187. 118. Rankin S, Rozengurt E. Platelet-derived growth factor
101. Kenagy RD, Vergel S, Mattsson E et al The role of plas- modulation of focal adhesion kinase (FAK) and paxillin
minogen, plasminogen activators, and matrix tyrosine phosphorylation in Swiss 3T3 cells. Bell shaped
metalloproteinases in primate arterial smooth muscle cell dose response and crosstalk with bombesin. J Biol Chem
migration. Arterioscler Thromb Vasc Biol 1996; 1994; 269:704-710.
16:1373-1382. 119. Haarer BK, Brown SS. Structure and function of profilin.
102. Okada SS, Grobmyer SR, Barnathan ES. Contrasting ef- Cell Motil Cytoskelet 1990; 17:71-74.
fects of plasminogen activators, urokinase receptor, and 120. Theriot JA, Mitchieson TJ. The three faces of profilin. Cell
LDL receptor-related protein on smooth muscle cell mi- 1993; 75:835-838.
gration and invasion. Arterioscler Thromb Vasc Biol 1996; 121. Yin HL. Gelsolin: Calcium- and polyphosphoinositide-
16:1269-1276. regulated actin-modulating protein. Bioessays 1987;
103. Wang W, Chen HJ, Giedd KN et al. T-cell lymphokines, 7:176-179.
interleukin-4 and gamma-interferon modulate the induc- 122. Carson MR, Shasby SS, Lind SE, Shasby DM. Histamine,
tion of vascular smooth muscle cell tissue plasminogen actin-gelsolin binding, and polyphosphoinositides in hu-
activator and migration by serum and platelet-derived man umbilical vein EC. Am J Physiol 1992; 263:
growth factor. Circ Res 1995; 77:1095-1106. L664-L669.
104. Noda-Heiny H, Sobel BE. Vascular smooth muscle cells 123. Chaponnier C, Kocher O, Gabbiani G. Modulation of
migration mediated by thrombin and urokinase receptor. gelsolin content in rat aortic smooth muscle cells during
Am J Physiol 1995; 268: C1195-C1201. development, experimental intimal thickening and culture.
105. Forough R, Koyama N, Hasenstab D et al. Overexpression Eur J Biochem 1990; 190:559-565.
of tissue inhibitor of matrix metalloproteinase-1 inhibits 124. Clyman RI, McDonald KA, Kramer RH. Integrin recep-
vascular smooth muscle cell functions in vitro and in vivo tors on aortic smooth muscle cells mediate adhesion to
Circ Res 1996; 79:812-820. fibronectin, laminin and collagen. Circ Res 1990;
106. Hynes RO. Integrins: Versatility, modulation and signal- 67:175-186.
ling in cell adhesion. Cell 1992; 69:11-25. 125. Clyman RI, Turner DC, Kramer RH. An # 1 / ! 1 -like
107. Burridge K, Chrzanowska-Wodnicka M. Focal adhesions, integrin receptor on rat aortic smooth muscle cells medi-
contractility, and signalling. Annu Rev Cell Dev Biol 1996; ates adhesion to laminin and collagen types I and IV.
12:463-519. Arteriosclerosis 1990; 10:402-9.
108. Jockusch BM, Bubeck P, Giehl K et al. The molecular 126. Yamamoto K, Yamamoto M. Cell Adhesion receptors for
architecture of focal adhesions. Annu Rev Cell Dev Biol native and denatured type I collagens and fibronectin in
1995; 11:379-416. rabbit arterial smooth muscle cells in culture. Exp Cell
109. Regan CM, Horvitz AF. Dynamics of !1 integrin-medi- Res 1994; 214:258-263.
ated adhesive contacts in motile fibroblasts. J Cell Biol 127. Janat MF, Argraves WS, Liau G. Regulation of vascular
1992; 117:1347-1359. smooth muscle cell integrin expression by transformtin
110. Schmidt CE, Horwitz AF, Lauffenburger DA et al. growth factor-! and platelet-derived growth factor-BB. J
Integrin-cytoskeletal interactions in migrating fibroblasts Cell Physiol 1992; 151:588-595.
are dynamic, asymmetric and regulated. J Cell Biol 1993; 128. Basson CT, Kocher O, Basson MD et al. Differential
123:977-991. modulation of vasuclar cell integrin and extracellular
111. Seufferlein T, Rozengurt E. Lysophosphatidic acid stimu- matrix expression in vitro by transformtin growth factor-
lates tyrosine phosphorylation of focal adhesion kinase, b1 correlates with reciprocal effects on cell migration. J
paxillin and p130. Signalling pathways and crosstalk with Cell Physiol 1992; 153:118-128.
platelet-derived growth factor. J Biol Chem 1994; 129. Clyman RI, Mauray F, Kramer RH. !1 and !3 integrins
269:9345-9351. have different roles in the adhesion and migration of vas-
112. Seufferlein T, Rozengurt E. Sphingosine induces focal cular smooth muscle cells on extracellular matrix. Exp Cell
adhesion kinase (FAK), and paxillin tyrosine phosphory- Res 1992; 200:272-284.
lation, actin stress fiber formation and focal contact as- 130. Damsky CH, Werb Z. Signal transduction by integrin re-
sembly in Swiss 3T3 cells. J Biol Chem 1994; ceptors for extracellular matrix: Cooperative processing of
269:27610-27617. extracellular information. Curr Opin Cell Biol 1992; 4:772-
113. Chrzanowska-Wodnicka M, Burridge K. Tyrosine phos- 781.
phorylation is involved in reorganization of the actin cy- 131. Zachary I, Rozengurt E. Focal adhesion kinase (FAK): A
toskeleton in response to serum or LPA stimulation. J Cell point of convergence in the action of neuropeptide,
Sci 1994; 107:3643-3654. integrins and oncogenes. Cell 1992; 71:891-894.
114. Turner CE, Pietras KM, Taylor DS et al. Angiotensin II 132. Schaller MD, Parsons JT. Focal adhesion kinase and as-
stimulation of rapid paxillin tyrosine phosphorylation sociated proteins. Curr Opin Cell Biol 1994; 6:705-710.
correlates with the formation of focal adhesions in rat
aortic smooth muscle cells. J Cell Sci 1995; 108:333-342.
Signaling Mechanisms for Vascular Cell Migration 267
133. Abedi H, Zachary I. Signalling mechanisms in the regula- phorylation. Role of Protein kinase c, Ca2+ mobilisation,
tion of vascular cell migration. Cardiovasc Res 1995; and the actin cytoskeleton. J Biol Chem 1993; 268:14261-
30:544-556. 14268.
134. Chen Q, Kinch MS, Lin TH et al. Integrin-mediated cell 150. Matsumoto K, Matsumoto K, Nakamura T et al. Hepato-
adhesion activates mitogen-activated protein kinases. J cyte growth factor/ scatter factor induces tyosine phos-
Biol Chem 1994; 269:26602-26605. phorylation of focal adhesion kinase (FAK) and promotes
135. Morino N, Mimura T, Hamasaki K et al. Matrix-integrin migration and invasion by oral squamous cell carcinoma
interaction activates the mitogen-activated protein kinase, cells. J Biol Chem 1994; 269:31807-31813.
pp44erk-1 and pp42erk-2. J Biol Chem 1995; 270:269-273. 151. Schaller MD, Otey CA, Hildebrand JD et al. Focal adhe-
136. Clark EA, Hynes RO. Ras activation is necessary for sion kinase and paxillin binds to peptides mimicking !
integrin-mediated activation of extracellular signal-regu- integrin cytoplasmic domains J Cell Biol 1995; 130:1181-
lated kinase 2 and cytosolic phospholipase A2 but not for 1187.
cytoskeletal organization. J Biol Chem 1996; 152. Hildebrand JD, Schaller MD, Parsons JT. Identification
271:14814-14818. of sequences required for the efficient localization of the
137. Schwartz MA, Lechene C. Adhesion is required for pro- focal adhesion kinase, p125 focal adhesion kinase (FAK),
tein kinase c-dependent activation of the Na + /H + to cellular focal adhesions. J Cell Biol 1993; 123:993-1005.
antiporter by PDGF. Proc Natl Acad Sci USA 1992; 153. Turner CE, Glenny JR Jr, Burridge K. Paxillin: A new
89:6138-6141. vinculin binding protein present in focal adhesions. J Cell
138. Lev S, Moreno H, Martinez R et al. Protein tyrosine ki- Biol 1990; 111:1059-1068.
nase PYK-2 is involved in Ca2+-induced regulation of ion 154. Zachary I, Sinnett-Smith J, Turner C et al. Bombesin,
channel function and MAP kinase functions. Nature 1995; vasopressin and endothelin stimulate tyrosine phospho-
376:737-745. rylation of the focal adhesion-associated protein paxillin
139. Richardson A, Parsons JT. A mechanism for regulation in Swiss 3T3 cells. J Biol Chem 1993; 268:22060-22065.
of the adhesion-associated tyrosine kinase p125 focal ad- 155. Turner CE, Schaller MD, Parsons JT. Tyrosine phospho-
hesion kinase. Nature 1996; 380:538-540. rylation of the focal adhesion kinase p125 focal adhesion
140. Kanner SB, Reynolds AB, Vines RR et al. Monoclonal kinase (FAK) during development: Relation to paxillin. J
antibodies to individual tyrosine phosphorylated protein Cell Sci 1993; 105:637-645.
subtrates of oncogene encoded tyrosine kinases. Proc Natl 156. Tachibana K, Sato T, D’Avirro N et al. Direct association
Acad Sci USA 1990; 87:3328-3332. of pp125FAK with paxillin, the focal adhesion-targetting
141. Guan J-L, Shalloway D. Regulation of focal adhesion as- mechanism of pp125FAK. J Exp Med 1995; 182:1089-1100.
sociated protein tyrosine kinase by both cellular adhesion 157. Salgia R, Li J-L, Lo SH et al. Molecular cloning of human
and oncogenic transformation. Nature 1992; 358:690-692. paxillin, a focal adhesion protein phosphorylated by
142. Hanks SK, Calalb MB, Harper MC et al. Focal adhesion p210BCR/ABL. J Biol Chem 1995; 270:5039-5047.
protein tyrosine kinase phosphorylated in response to cell 158. Sakai R, Iwamatsu A, Hirano N et al. A novel signalling
attachment to fibronectin. Proc Natl Acad Sci USA 1992; molecule, p130, forms stable complexes in vivo with v-
89:8487-8491. Crk and v-Src in a tyrosine phosphorylation-dependent
143. Burridge K, Turner CE, Romer LH. Tyrosine phosphory- manner. EMBO J 1994; 13:3748-3756.
lation of paxillin and p125 focal adhesion kinase accom- 159. Polte TR, Hanks SK. Interaction between focal adhesion
panies cell adhesion to extracellular matrix: A role in kinase and Crk-associated tyrosine kinase substrate
cytoskeletal assembly. J Cell Biol 1992; 119:893-903. p130Cas. Proc Natl Acad Sci USA 1995; 92:10678-10682.
144. Kornberg L, Earp HS, Parsons JT et al. Cell adhesion or 160. Polte TR, Hanks SK. Complexes of focal adhesion kinase
integrin clustering increases phosphorylation of a focal (FAK) and Crk-associated substrate (p130Cas) are elevated
adhesion associated tyrosine kinase. J Biol Chem 1992; in cytoskeleton-associated fractions following adhesion
267:23439-23442. and src transformation J Biol Chem 1997; 272:5501-5509.
145. Lipfert L, Haimovich B, Schaller MD et al. Integrin-de- 161. Hall CL, Wang C, Lange LA et al. Hyaluronan and the
pendent phosphorylation and activation of the protein ty- hyaluronan receptor RHAMM promote focal adhesion
rosine kinase p125 focal adhesion kinase (FAK) in plate- kinase turnover and transient tyrosine kinase activity J Cell
lets. J Cell Biol 1992; 119:905-912. Biol 1994; 126:575-588.
146. Huang M-M, Lipfert L, Cunningham M et al. Adhesive 162. Leventhal PS, Shelden EA, Kim B et al. Tyrosine phos-
ligand binding to integrin alpha (IIb) beta 3 stimulates phorylation of paxillin and focal adhesion kinase during
tyrosine phosphorylation of novel protein substrates be- insulin-like growth factor-I-stimulated lamellipodial ad-
fore phosphorylation of p125 focal adhesion kinase (FAK). vance J Biol Chem 1997; 272:5214-5218.
J Cell Biol 1993; 122:473-483. 163. van Corven EJ, Hordijk PL, Medema RH et al. Pertussis
147. Hamawy MM, Mergenhagen SE, Siraganian RP. Tyrosine toxin-sensitive activation of p21ras by G protein-coupled
phosphorylation of p125 focal adhesion kinase (FAK) by receptor agonists in fibroblasts. Proc Natl Acad Sci USA
the aggregation of high affinity immunoglobin E recep- 1993; 86:1259-1261.
tors requires cell adherence. J Biol Chem 1993; 164. Hordijk RJ, Verlaan L, van Corven PL et al. Protein ty-
268:6851-6854. rosine phosphorylation induced by lysophosphatidic acid
148. Zachary I, Sinnett-Smith J, Rozengurt E. Bombesin, va- in Rat-1 fibroblasts. Evidence that phosphorylation of
sopressin and endothelin stimulation of tyrosine phospho- MAP kinase is mediated by the Gi-p21ras pathway. J Biol
rylation in Swiss 3T3 cells. Identification of a novel ty- Chem 1994; 269:645-651.
rosine kinase as a major substrate. J Biol Chem 1992; 165. Kumagai N, Morii N, Fujisawa K et al. ADP-ribosylation
267:19031-19034. of Rho p21 inhibits lysophosphatidic acid -induced pro-
149. Sinnett-Smith J, Zachary I, Valverde A et al. Bombesin tein tyrosine phosphorylation and phosphatidylinositol
stimulation of FAK focal adhesion kinase tyrosine phos-
268 Tissue Engineering of Prosthetic Vascular Grafts
3-kinase activation in cultured Swiss 3T3 cells. J Biol 183. Ikebe M, Hartshorne DJ. Effects of Ca2+ on the confor-
Chem 1993; 268:24535-24538. mation and enzymatic activation of smooth muscle myo-
166. Schaller MD, Borgman CA, Cobb BS et al. P125 focal sin. J Biol Chem 1985; 260:10027-10031.
adhesion kinase, a structurally distinctive protein-tyrosine 184. Kamisoyama H, Araki Y, Ikebe M. Mutagenesis of the
kinase associated with focal adhesions. Proc Natl Acad Sci phosphorylation site (serine 19) of smooth muscle myo-
USA 1992; 89:5192-5196. sin regulatory light chain and its effects on the properties
167. Cobb BS, Schaller MD, Leu T-H et al. Stable association of myosin. Biochemistry 1994; 33:840-847.
of pp60Src and pp59fyn with the focal adhesion-associated 185. Amano M, Chihara K, Kimura K et al. Formation of ac-
protein tyrosine kinase, p125 focal adhesion kinase. Mol tin stress fibers and focal adhesions enhanced by rho-ki-
Cell Biol 1994; 14:147-155. nase. Science 1997; 275:1308-1311.
168. Chen H-C, Guan J-L. Association of focal adhesion ki- 186. Cantley LC, Auger KR, Carpenter C et al. Oncogenes and
nase with its potential substrate phosphatidylinositol 3- signal transduction. Cell 1991; 64:281-302.
kinase. Proc Natl Acad Sci USA 1994; 91:10148-10152. 187. Claesson-Welsh L. Platelet-derived growth factor recep-
169. Schlaepfer DD, Hanks SK, Hunter T et al. Integrin-medi- tor signals. J Biol Chem 1994; 269:32023-32026.
ated signal transduction linked to Ras pathway by GRB2 188. de Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N,
binding to focal adhesion kinase. Nature 1994; Williams LT. The fms-like tyrosine kinase, a receptor for
372:786-791. vascular endothelial growth factor. Science 1992;
170. Guinebault C, Payrastre B, Racaud-Sultan C et al. 255:989-991.
Integrin-dependent translocation of phosphoinositide 3- 189. Matthews W, Jordan CT, Gavin M, Jenkins NA, Copeland
kinase to the cytoskeleton of thrombin-activated platelets NG, Lemischka IR. A receptor tyrosine kinase cDNA iso-
involves specific interactions of p85# with actin filaments lated from a population of enriched primitive hemato-
and focal adhesion kinase. J Cell Biol 1995; 129:831-842. poietic cells and exhibiting close genetic linkage to c-kit.
171. Birge RB, Fajardo JE, Reichman C et al. Identification and Proc Natl Acad Sci USA 1991; 88:9026-9030.
characterisation of a high-affinity interaction between v- 190. Quinn TP, Peters KG, De Vries C, Ferrara N, Williams
Crk and tyrosine-phosphorylated paxillin in CT 10-trans- LT. Fetal liver kinase-1 is a receptor for vascular endot-
formed fibroblasts. Mol Cell Biol 1993; 13:4648-4656. helial growth factor and is selectively expressed in vascu-
172. Sabe H, Hata A, Okada M et al. Analysis of the binding lar endothelium. Proc. Natl. Acad. Sci. USA 1993;
of the Src homology 2 domain of Csk to tyrosine-phos- 90:7533-7537.
phorylated proteins in the suppression and mitotic acti- 191. Millauer B, Wizigmann-Voos S, Schnuch H, Martinez R,
vation of C-src. Proc Natl Acad Sci USA 1994; Moller NPH, Risau W, Ullrich A. High affinity VEGF
91:3984-3988. binding and developmental expression suggest Flk-1 as a
173. Ridley A. Rho-related proteins: Actin cytoskeleton and cell major regulator of vasculogenesis and angiogenesis. Cell
cycle. Curr opin Gen and Develop 1995; 5:24-30. 1993; 72:835-846.
174. Paterson HF, Self AJ, Garrett MD et al. Microinjection of 192. Shweiki D, Itin A, Neufeld G, Gitay-Goren H, Keshet E.
recombinant p21rho induces rapid changes in cell mor- Patterns of expression of vascular endothelial growth fac-
phology. J Cell Biol 1990; 111:1001-1007. tor (VEGF) and VEGF receptors in mice suggest a role in
175. Kozma R, Ahmed S, Best A, Lim L. The ras-related pro- hormonally regulated angiogenesis. J. Clin. Invest. 1993;
tein cdc42Hs and bradykinin promote formation of pe- 91:2235-2243.
ripheral actin microspikes and filipodia in Swiss 3T3 fi- 193. Guo D, Jia Q, Song H-Y, Warren RS, Donner DB. Vas-
broblasts. Mol Cell Biol 1995; 15:1942-1952. cular endothelial cell growth factor promotes tyrosine
176. Nobes CD, Hall A. Rho, rac and cdc42 GTPases regulate phosphorylation of mediators of signal transduction that
the assembly of multimolecular focal complexes associ- contain SH2 domains. J Biol Chem 1995; 270:6729-6733.
ated with actin stress fibers, lamellipodia and filopodia. 194. Seetharam L, Gotoh N, Maru Y, Neufeld G, Yamaguchi
Cell 1995; 81:53-62. S, Shibuya M. A unique signal transduction from Flt ty-
177. Rankin S, Morii N, Narumiya S et al. Botulinum C3 ex- rosine kinase, a receptor for vascular endothelial growth
oenzyme blocks the tyrosine phosphorylation of FAK and factor VEGF. Oncogene 1995; 10:135-147.
paxillin induced by bombesin and endothelin. FEBS Letts 195. Chen P, Murphy-Ullrich JE, Wells A. A role for gelsolin
1994; 354:315-319. in actuating epidermal growth factor receptor-mediated
178. Seckl M, Morii N, Narumiya S et al. Guanine 5'-3-O- cell motility. J Cell Biol 1996; 134:689-698.
(thio)triphosphate stimulates tyrosine phosphorylation of 196. Hartwig JH, Bokoch GM, Carpenter CL et al. Thrombin
FAK and paxillin in permeabilized Swiss 3T3 cells. J Biol receptor ligation and activated Rac uncap actin filament
Chem 1995; 270:6984-6990. barbed ends through phosphoinositide synthesis in
179. Flinn HM, Ridley AJ. Rho stimulates tyrosine phospho- permeabilized human platelets. Cell 1995; 82:643-653.
rylation of focal adhesion kinase, p130 and paxillin. J Cell 197. Gilmore AP, Burridge K. Regulation of vinculin binding
sci 1996; 109:1133-1141. to talin and actin by phosphatidylinositol-4-5-
180. Tanaka S, Morishita T, Hashimoto Y et al. C3G, a gua- bisphosphate. Nature 1996; 381:531-535.
nine nucleotide-releasing protein expressed ubiquitously, 198. Pauly RR, Bilato C, Sollott SJ et al. Role of calcium/
binds to the Src homology 3 domains of CRK and GRB2/ calmodulin dependent protein kinase II in the regulation
ASH. Proc Natl Acad Sci USA 1994; 91:3443-3447. of vascular smooth muscle cell migration. Circulation
181. Hawkins PT, Eguinoa A, Qui R-G et al. PDGF stimulates 1995; 91:1107-1115.
an increase in GTP-Rac via activation of phosphoinositide 199. Lawson MA, Maxfield FR. Ca2+-and calcineurin-depen-
3-kinase. Curr Biol 1995; 5:393-403. dent recycling of an integrin to the front of migrating
182. Amano M, Ito M, Kimura K et al. Phosphorylation and neutrophils. Nature 1995; 377:75-79.
activation of myosin by Rho-associated kinase (Rho-ki- 200. Shibasaki Y, Ishihara H, Kizuki N et al. Massive actin
nase) J Biol Chem 1996; 271:20246-20249. polymerization induced by phosphatidylinositol-4-phos-
phate 5-kinase in vivo J. Biol Chem 1997; 272:7578-7581.
Signaling Mechanisms for Vascular Cell Migration 269
201. Chong LD, Traynor-Kaplan A, Bokoch GM et al. The taxis to C-C chemokines by antisense oilgonucleotide for
small GTP-binding protein Rho regulates a cytosolic phospholipase A 2 . J Biol Chem 1996;
phosphatidylinositol 4-phosphate 5-kinase in mammalian 271:6010-6016.
cells. Cell 1994; 79:507-513. 210. Sa G, Fox PL. Basic fibroblast growth factor-stimulated
202. Renshaw MW, Toksoz D, Schwartz MA. Involvement of EC movement is mediated by a pertussis toxin-sensitive
the small GTPase Rho in integrin-mediated activation of pathway regulating phospholipase A2 activity. J Biol Chem
mitogen-activated protein kinase. J Biol Chem 1996; 1994; 269:3219-3225.
271:21691-21694. 211. Leibovich SJ, Polverini PJ, Fong TW et al. Production of
203. Kundra V, Escobedo JA, Kazlauskas A et al. Regulation angiogenic activity by human monocytes requires an L-
of chemotaxis by the platelet-derived growth factor re- arginine/nitric oxide-synthase-dependent effector mecha-
ceptor-beta. Nature 1994; 367:474-476. nism. Proc Natl Acad Sci USA 1994; 91:4190-4194.
204. Wennstrom S, Siegbahn A, Yokote K et al. Membrane 212. Noiri E, Peresleni T, Srivastava N et al. Nirtric oxide is
ruffling and chemotaxis transduced by the PDGF beta necessary for a switch from stationary to locomoting phe-
receptor require the binding site for phosphatidylinositol notype in epithelial cells. Am J Physiol 1996;
3' kinase. Oncogene 1994; 9:651-660. 270:C794-C802.
205. Kotani K, Yonezawa K, Hara K et al. Involvement of 213. Noiri E, Hu Y, Bahou WF et al. Permissive role of nitric
phosphoinositide 3-kinase in insulin- or IGF-1-induced oxide in endothelin-induced migration of endothelial cells.
membrane ruffling. EMBO J 1994; 13:2313-2321. J Biol Chem 1997; 272:in press.
206. Powell WS, Chung D, Gravel S. 5-oxo-6,8,11,14- 214. Hotchin NA, Hall A. The assembly of inyegrin adhesion
eicosatetraenoic acid is a potent stimulator of human eosi- complexes requires both extracellular matrix and intrac-
nophil migration. J Immunol 1995; 154:4123-4132. ellular rho/rac GTPases. J Cell Biol 1995; 131:1857-1865.
207. Schwenk U, Schroder JM. 5-oxo-eicosanoids are potent 215. Bornfeldt KE. Intracellular signalling in arterial smooth
eosinophil chemotactic factors. J Biol Chem 1995; muscle: Migration versus proliferation. Trends Cardiovasc
270:15029-15036. Med 1996; 6:143-151.
208. Maddox JE, Serhan CN. Lipoxin A4 and B4 are potent 216. Graf K, Xi XP, Yang D et al. Mitogen-activated protein
stimuli for human monocyte migration and adhesion: kinasde activation is involved in platelet-derived growth
Selective inactivation by dehydrogenation and reduction. factor-directed migration by vascular smooth muscle cells.
J Exp Med 1996; 183:137-146. Hypertension 1997; 29:334-339.
209. Locati M, Lamorte G, Luini W, Introna M, Bernasconi S,
Mantovani A, Sozzani S. Inhibition of monocyte chemo-
Facilitation of Healing
Chemotaxis
CHAPTER 25
Adhesion Molecules: Potent Inducers
of Endothelial Cell Chemotaxis
Zoltan Szekanecz, Alisa E. Koch
Introduction
A ngiogenesis, the formation of new blood vessels, plays an important role in a number of
physiological processes including development and tissue repair. Thus, it may also be
involved in the integration of newly implanted vascular grafts. Neovascularization is also
essential in a number of pathological situations, such as atherosclerosis, diabetic angiopa-
thy, rheumatoid arthritis, psoriasis and tumor growth.1-3
During angiogenesis, endothelial cells produce proteolytic enzymes, which leads to
the degradation of the basement membrane, followed by the emigration of endothelial cells.
Although there is another chapter on angiogenesis in this volume, here we will discuss the
role of cellular adhesion molecules and adhesive mechanisms in the induction of endothe-
lial cell migration and chemotaxis, which are essential steps in capillary formation. There is
evidence that certain adhesion molecules and extracellular matrix molecules may be chemo-
tactic for endothelial cells and thus may stimulate the migration of these cells into sites of
angiogenesis. It is possible that the induction of endothelial cell migration and
neovascularization around the vascular graft may facilitate its healing.
In this chapter, we will summarize our current understanding of the role of adhesion
factors in endothelial cell migration and chemotaxis. We will discuss the involvement of
extracellular matrix components and cellular adhesion molecules in these processes, also
highlighting our recently published data on recombinant, soluble adhesion molecule-in-
duced endothelial cell chemotaxis and neovascularization. We will also discuss the interac-
tions of other soluble mediators, such as growth factors and cytokines, with extracellular
matrix molecules and cellular adhesion molecules during endothelial cell migration and
angiogenesis. With regard to vascular surgery, we will present information on the role of
cellular adhesion molecules in the formation of human abdominal atherosclerotic aortic
aneurysms, as well as in wound healing. These data may support our theory that, after vascular
graft implantation, extracellular matrix macromolecules and cellular adhesion molecules
are involved in attracting endothelial cells and in the formation of new capillaries. Thus, the
induction of adhesion molecule expression may stimulate the revascularization of the sur-
rounding extracellular matrix, enhance blood supply and enable the survival of the graft.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler
©1999 R.G. Landes Company.
272 Tissue Engineering of Prosthetic Vascular Grafts
Table 25.1. Endothelial cellular adhesion molecules involved in endothelial cell migration and neovascularization.
receptor, however, is #6!1. There are two important recep- We have recently shown the role of certain soluble,
tors for fibronectin, the RGD (arginine-glycine-aspartic recombinant endothelial cellular adhesion molecules in en-
acid)-dependent #5!1, as well as the RGD-independent dothelial cell chemotaxis, as well as in vivo
20-22,24,25
#4!1. The former is present on most endothelial neovascularization. Soluble vascular cell adhesion mol-
cells,25 while we and others have confirmed that #4!1 is also ecule-1 and soluble E-selectin have been tested in endothe-
expressed by some endothelial cells.27,28 Another fibronectin, lial cell chemotaxis assays in vitro and the rat corneal
laminin and collagen receptor, #3!1, is also present on en- neovascularization model in vivo.27 Recombinant E-selectin
dothelial cells.25,26 Integrins containing the !3 subunit are and vascular cell adhesion molecule-1 as chemotactic fac-
involved in endothelial cell adhesion to fibronectin, tors dose-dependently increased the chemotaxis of human
vitronectin, thrombospondin, von willebrand factor and fi- umbilical vein endothelial cells and dermal microvascular
brinogen. The #v integrin subunit can be associated with endothelial cells above the basal level. The chemotactic ac-
several ! chains (!1, !3, !5, !6, !8), and mediates endothelial tion of these soluble cellular adhesion molecules was com-
cell adhesion to a variety of extracellular matrix components, parable to that of basic fibroblast growth factor, a “classical”
depending on the ! subunit.20-22,25,26 angiogenic mediator. We have also shown in checkerboard
Extracellular matrix binding integrins can be classi- analyses that both soluble E-selectin and soluble vascular
fied into subgroups of “basement membrane” (collagen- cell adhesion molecule-1 were chemotactic rather than
laminin)-binding integrins (#1!1, #2!1, #3!1 and #6!1) and chemokinetic for endothelial cells. These soluble cellular
“inflammatory matrix” integrins (fibronectin-fibrinogen adhesion molecules seem to be mediating a direct effect, as
receptors: #4!1, #5!1, #v and !3). While microvessels express they did not stimulate umbilical endothelial cells to produce
the former, but not the latter type of integrins in situ, cellu- the angiogenic basic fibroblast growth factor, tumor necro-
lar adhesion molecules belonging to both subgroups are sis factor-# or interleukin-8 in vitro.27
present on capillary endothelial cells in vitro. These data In addition, monoclonal antibodies to the E-selectin
suggest that endothelial cells have a potential to alter their ligand sialyl Lewis-X and to the vascular cell adhesion mol-
cellular adhesion molecule profile during vascular morpho- ecule-1 counterreceptor #4!1 integrin on endothelial cells,
genesis.25 Additional studies have shown that the #2!1 mol- significantly reduced soluble cellular adhesion molecule-
ecule is involved in type I collagen-induced endothelial cell mediated human umbilical vein endothelial cell chemotaxis.
migration and neovascularization.10,24 The #v!3 molecule was We and others have detected both sialyl Lewis-X and #4!1
found to be involved in endothelial cell migration on on umbilical vein endothelial cells by immunohistochemis-
vitronectin.24 #v!3 is necessary for capillary formation in try.27,30 Although sialyl Lewis-X has been implicated in bo-
tumors and wound granulation tissue and promotes endot- vine capillary morphogenesis in vitro,34 our novel results
helial cell cord formation in vitro.29,30 It has recently been suggest a link between the soluble E-selectin-sialyl Lewis-X,
shown that the #v!3 integrin is required for the survival and as well as soluble vascular cell adhesion molecule-1-#4!1,
maturation of new blood vessels.30 As described above, !3 adhesion pathways, which mediate leukocyte-endothelial cell
integrins may also be important in thrombospondin-1-me- interactions, endothelial cell recruitment and angiogenesis.27
diated endothelial cell chemotaxis.18 The #6!1 integrin stimu- These data are supported by those of other investigators
lates angiogenesis on Matrigel.31 showing that sugars, which are also major constituents of
Adhesion molecules mediating endothelial cell adhe- selectins and sialyl Lewis-X, are chemotactic for bovine cor-
sion to other cells include vascular cell adhesion molecule-1, neal endothelial cells.36 In the rat corneal neovascularization
intercellular adhesion molecules-1 and -2, CD31, E- and assay both soluble cellular adhesion molecules induced in
P-selectin, CD34 and other carbohydrate ligands for vivo capillary formation at low concentrations in most ex-
L-selectin, and the transformtin growth factor-!1 receptor periments. We have also shown in neutralization studies that
endoglin.20-23,32,33 Antibodies to E-selectin, as well as to its soluble E-selectin and soluble vascular cell adhesion mol-
ligands sialyl Lewis-X and sialyl Lewis-A, inhibit the migra- ecule-1 account for a portion of rheumatoid arthritic syn-
tion and tube formation of capillary endothelial cells.24,34 ovial fluid-mediated chemotactic activity for human um-
CD31 has been implicated in tumor-associated bilical vein endothelial cells.27
neovascularization.35 The L-selectin ligand CD34 shows high In summary, we have shown that soluble E-selectin
endothelial cell expression in developing tissues and heal- and soluble vascular cell adhesion molecule-1 may act as
ing wounds.32 proangiogenic factors. They may directly trigger the migra-
Endothelial adherens junctions, which contain clus- tion and chemotaxis of endothelial cells by binding to their
tered cellular adhesion molecules and other membrane pro- respective endothelial cell ligands.27
teins, were also implicated in endothelial cell migration, ad-
hesion and angiogenesis. VE-cadherin, a major constituent Interactions of Soluble Mediators with Cellular
of these junctions, mediates homophilic binding between Adhesion Molecules and Extracellular Matrix
endothelial cells. Many other adhesive proteins, such as Components During Endothelial Cell
CD31, CD34, endoglin, occludin and connexins show Recruitment and Neovascularization
colocalization with VE-cadherin.23 As endothelial cells bind Cellular adhesion molecules and extracellular matrix
to each other through these cellular adhesion molecules, macromolecules may interact with other soluble mediators
these cells can migrate in organized clusters during capil- during endothelial cell migration and neovascularization.
lary formation. In fact, basic fibroblast growth factor treatment of
274 Tissue Engineering of Prosthetic Vascular Grafts
endothelial cells in vitro resulted in an increased expression RANTES, macrophage inflammatory protein-1# and mono-
of the integrin subunits #2, #3, #5, #6, !1, !3, !4 and !5, as well cyte chemoattractant protein-1 stimulate the expression and
as in better adherence to type I collagen, fibronectin, laminin avidity of both !1 and !2 integrins on T cells and mono-
and vitronectin compared to untreated cells.37,38 Interactions cytes.45,46 These chemokines also promote the adhesion of
between basic fibroblast growth factor, thrombospondin and mononuclear cells to extracellular matrix components, as
cellular adhesion molecules have also been demonstrated. well as recombinant human intercellular adhesion molecule-
Thrombospondin inhibits angiogenesis by blocking the ef- 1 and vascular cell adhesion molecule-1.45,46 However, most
fects of basic fibroblast growth factor on endothelial cell of these data are available on leukocytes, and not on endot-
chemotaxis.16,17,39 In addition, thrombospondin receptors, helial cells. Therefore, the possibility that either C-X-C or
such as !3 integrins, have been implicated in endothelial cell C-C chemokines may also influence endothelial cell migra-
migration and neovascularization.18,26,30 tion and chemotaxis via the modulation of endothelial cell
Transformtin growth factor-! and basic fibroblast adhesion molecule expression needs to be elucidated.
growth factor differentially modulate the integrin profile on
human microvascular endothelial cells, as transformtin The Relevance of Angiogenesis Studies
growth factor-! downregulates integrin expression on hu- in Vascular Surgery: Aortic Aneurysms
man dermal microvascular endothelial cells, both at the and Wound Healing
mRNA and protein level.38,40 This growth factor also inhib- We have performed a number of studies on the pos-
its fibronectin-mediated endothelial cell chemotaxis.40 One sible role of angiogenic mediators and adhesion molecules
receptor for transformtin growth factor-!1, endoglin, con- in the pathogenesis of human abdominal aortic aneurysms.
tains an RGD motif and thus serves as a cellular adhesion In this disease, atherosclerotic and inflammatory mecha-
molecule for endothelial cells.23,33 We found increased, well nisms occurring in the aneurysm wall are accompanied by
correlated expression of endoglin and transformtin growth the escalation of neovascularization. Small vessels of the vasa
factor-!1 in the highly vascularized synovium of rheuma- vasorum in the adventitial layer proliferate and the migra-
toid arthritis patients.33 tion of endothelial cells into the aortic wall plays an impor-
Treatment of endothelial cells with tumor necrosis tant role in the formation of new vessels and the growth of
factor-# stimulates #1!1 expression and adhesion to laminin, aneurysm.47-50
while the combination of tumor necrosis factor-# and in- Our immunohistochemical analysis carried out on
terferon-g downregulates #v!3 expression on these cells, sug- aortic aneurysm explants revealed that there was an increased
gesting the modulating effect of angiogenic and angiostatic expression of intercellular adhesion molecule-1 on endot-
mediators in endothelium-extracellular matrix interac- helial cells found in aortic aneurysm tissues compared to
tions.26 Tumor necrosis factor-# can also induce the expres- normal control aortas.50 Moreover, aortic aneurysm explants
sion of intercellular adhesion molecule-1, vascular cell ad- released significantly more soluble intercellular adhesion
hesion molecule-1, E-selectin and some integrins on syn- molecule-1 into their culture supernatants than did normal
ovial and other types of endothelial cells.3,20,21,25,26 The in- aortic tissues.47 We have used human aortic endothelial cell
creasing production of endothelial cellular adhesion mol- culture as an in vitro model for studying the interactions of
ecules may result in the perpetuated shedding of these cel- angiogenic cytokines and cellular adhesion molecules. The
lular adhesion molecules from the cell surface. The serum angiogenic cytokine tumor necrosis factor-# stimulated in-
and synovial fluid concentrations of soluble E-selectin and tercellular adhesion molecule-1 expression of, and soluble
soluble vascular cell adhesion molecule-1 are high in rheu- intercellular adhesion molecule-1 production by, these en-
matoid arthritis, a well-known “angiogenic disease”.41,42 As dothelial cells compared to control, unstimulated aortic en-
described above, these soluble cellular adhesion molecules, dothelial cells.47 We have also determined that aortic aneu-
in turn, may stimulate endothelial cell chemotaxis and an- rysm explants produce more tumor necrosis factor-#,
giogenesis.27 interleukin-6 and interleukin-8 into their conditioned me-
Chemokines, which are chemotactic for endothelial dia, than do normal aortic tissues.48,49,51,52 These cytokines
cells, as well as for leukocytes, may also be involved in this induced neovascularization in both in vitro and in vivo
regulatory network. Some C-X-C chemokines containing the models for angiogenesis.2,3,53-55 Moreover, we have shown,
ELR motif, such as interleukin-8, have high angiogenic ac- that tumor necrosis factor-# and interleukin-8 account for
tivity and they promote endothelial cell chemotaxis. In con- a portion of aortic aneurysm-associated endothelial cell
trast, C-X-C chemokines lacking the ELR sequence, such as chemotaxis.48
interferon-inducible protein-10 and platelet factor IV, in- Thus, soluble angiogenic and chemotactic mediators
hibit endothelial cell migration and angiogenesis.43 Although and certain cellular adhesion molecules, at least intercellu-
very little is known about the effects of C-X-C chemokines lar adhesion molecule-1, can be detected in aortic aneurysm
on cellular adhesion molecules, interleukin-8 stimulates !2 tissues. The increased production of these mediators results
integrin expression on neutrophils,44 while interferon-in- in increased endothelial intercellular adhesion molecule-1
ducible protein-10 induces integrin-dependent T cell adhe- expression. In conclusion, the interactions of angiogenic and
sion to fibronectin, laminin and collagen, as well as to re- chemotactic cytokines with cellular adhesion molecules may
combinant human intercellular adhesion molecule-1 and be important in the perpetuation of endothelial cell recruit-
vascular cell adhesion molecule-1.45 There are a number of ment and capillary formation underlying the growth of aor-
data available showing that C-C chemokines such as tic aneurysms.
Adhesion Molecules: Potent Inducers of Endothelial Cell Chemotaxis 275
Regarding wound healing, in the normal skin cellular diators of endothelial cell migration and angiogenesis. In
adhesion molecule expression is restricted to the basal layer addition, as one of the possible ligands for E-selectin is closely
of the epidermis. These cellular adhesion molecules medi- related to basic fibroblast growth factor, these cellular adhe-
ate the adhesion of the basal cells to the basement mem- sion molecules may also participate in neovascularization-
brane.56,57 During injury and wound healing, cellular adhe- associated signaling triggered by other mediators. Thus, the
sion molecules, mostly !1 and !3 integrins, appear in the complex interactions between endothelial cells, extracellu-
suprabasal layers of the skin, accompanied by the influx of lar matrix macromolecules, cellular adhesion molecules and
fibroblasts and endothelial cells, their adhesion to other angiogenic factors may lead to the perpetuation of
fibronectin, vitronectin and fibrinogen, as well as angiogen- angiogenesis. There are antagonistic factors, such as
esis and scarring.56,58 Similarly, we have found strong vas- transformtin growth factor-b and thrombospondin-1, which
cular cell adhesion molecule-1, P-selectin and CD44 expres- inhibit endothelial cell chemotaxis, as well as
sion in the injured, desquamating skin of scleroderma pa- neovascularization. The net balance of these factors result
tients.57 in the perpetuation or downregulation of endothelial cell
migration in the sites of angiogenesis. The detection of these
A Regulatory Network in Sites of Endothelial factors, as well as functional studies using in vitro chemot-
Cell Migration: Potential Target Promoting axis assays or in vivo animal models, can lead us to the bet-
Graft Healing ter understanding of the role of adhesive mechanisms around
In conclusion, in “angiogenic states”, such as rheuma- the healing vascular graft (Fig. 25.1).
toid arthritis, aortic aneurysms, wound healing or vascular In addition, targeting cellular adhesion molecules, ex-
graft implantation, cellular adhesion molecules such as !1 tracellular matrix components or other angiogenic media-
and ! 3 integrins, vascular cell adhesion molecule-1, tors by local injections of any mediators described above
E-selectin, sialyl Lewis-X and others, as well as extracellular may result in the local enhancement of endothelial cell
matrix molecules including collagen, fibronectin, laminin, chemotaxis and neovascularization, and thus may be useful
tenascin, fibrinogen and heparin may be involved in endo- for the healing and the prolonged survival of implanted vas-
thelial cell chemotaxis, migration and adhesion to extracel- cular grafts. Recently, putative progenitor endothelial cells
lular matrix components. A number of soluble mediators for angiogenesis were isolated from human peripheral blood.
secreted mainly by macrophages and endothelial cells in the These originally hematopoietic stem cells carry the CD34
extracellular matrix may interact with these cellular adhe- antigen, as well as Flk-1, a receptor for the angiogenic vas-
sion molecules and extracellular matrix components. Some cular endothelial growth factor. While mononuclear cells lose
of these factors, such as basic fibroblast growth factor, tu- these two antigens during in vivo differentiation, progeni-
mor necrosis factor-#, and possibly chemokines may induce tor cells plated onto fibronectin attach and become spindle-
cellular adhesion molecule expression on and shedding from shaped within days, and continue expressing CD34 and
the endothelial cell surface. Increased cellular adhesion mol- Flk-1, as well as endothelial cell marker cellular adhesion
ecule expression may result in stronger endothelial cell ad- molecules, such as E-selectin and CD31. In in vivo animal
hesion to extracellular matrix components. As we and oth- models of hindlimb ischemia, where one femoral artery is
ers have suggested, soluble, and possibly surface-bound en- excised, the progenitor endothelial cells isolated by magnetic
dothelial cellular adhesion molecules can act as direct me- bead separation using anti-CD34 or anti-Flk-1 monoclonal
antibodies migrated into sites of ischemia and angiogenesis. 17. DiPietro LA, Nebgen DR, Polverini PJ. Downregulation
Preliminary data suggest that these endothelial cells, possi- of endothelial cell thrombospondin 1 enhances in vitro
bly with the administration of angiogenic cytokines that angiogenesis. J Vasc Res 1994; 31:178-185.
enhance endothelial cell migration, could be used in autolo- 18. Taraboletti G, Roberts D, Liotta LA et al. Platelet
thrombospondin modulates endothelial cell adhesion,
gous endothelial cell transplantation in order to promote
motility, and growth: A potential angiogenesis regulatory
neovascularization.59 Similar techniques may be applied in factor. J Cell Biol 1990; 111:765-772.
vascular surgery as well. 19. Grant DS, Lelkes PI, Fukuda K. et al. Intracellular mecha-
nisms involved in basement membrane induced blood
Acknowledgments vessel differentiation in vitro. In Vitro Cell Dev Biol 1991;
This work was supported by NIH grants AR30692 and 27A: 327-336.
AR41492 (A.E.K.), funds from the Veterans’ Administration 20. Albelda SM, Buck CA. Integrins and other cell adhesion
Research Service (A.E.K.), Hungarian Scientific Foundation molecules. FASEB J 1990; 4:2868-2880.
grant No. T013239 (Z.S.) and Hungarian Medical Research 21. Springer TA. Adhesion receptors of the immune system.
Council grant No. 156/93 (Z.S.). Nature 1990; 346:425-433.
22. Szekanecz Z, Szegedi G. Cell surface adhesion molecules:
References Structure, function, clinical importance. Orv Hetil 1992;
1. Colville-Nash PR, Scott DL. Angiogenesis in rheumatoid 133:135-142.
arthritis: Pathogenic and therapeutic implications. Ann 23. Dejana E. Endothelial adherens junctions: Implications in
Rheum Dis 1992; 51:919-925. the control of vascular permeability and angiogenesis. J
2. Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992; Clin Invest 1996; 98:1949-1953.
267:10931-10934. 24. Bischoff J. Approaches to studying cell adhesion molecules
3. Klagsbrun M, D’Amore PA. Regulators of angiogenesis. in angiogenesis. Trends Cell Biol 1995; 5:69-74.
Annu Rev Physiol 1991; 53:217-239. 25. Albelda SM. Differential expression of integrin cell-sub-
4. Diaz-Flores L, Gutierrez R, Varela H. Angiogenesis: An stratum adhesion receptors on endothelium. EXS 1991;
update. Histol Histopathol 1994; 9:807-843. 59:188-192.
5. Sage EH, Vernon, RB. Regulation of angiogenesis by ex- 26. Defilippi P, Bozzo C, Geuna M et al. Modulation of ex-
tracellular matrix: The growth and the glue. J Hypertens tracellular matrix receptors (integrins) on human endot-
1994; 12(Suppl 10):S145-S152. helial cells by cytokines. EXS 1991; 59:193-197.
6. Montesano R, Pepper MS, Vassalli J-D et al. Modulation 27. Koch AE, Halloran MM, Haskell CJ et al. Angiogenesis
of angiogenesis in vitro. EXS 1991; 59:129-136. mediated by soluble forms of E-selectin and vascular cell
7. Madri JA, Williams KS. Capillary endothelial cell cultures: adhesion molecule-1. Nature 1995; 376:517-519.
Phenotypic modulation by matrix components. J Cell Biol 28. Massia SP, Hubbell JA. Vascular endothelial cell adhesion
1983; 97:153-165. and spreading promoted by the peptide REDV of the
8. Terranova VP, DiFlorio R, Lyall RM et al. Human endot- IIICS region of plasma fibronectin is mediated by integrin
helial cells are chemotactic to endothelial cell growth fac- alpha 4 beta 1. J. Biol Chem. 1992; 267:14019-14026.
tor and heparin. J Cell Biol 1985; 101:2330-2334. 29. Brooks PC, Clark RA, Cheresh DA. Requirement of vas-
9. Garrido T, Riese HH, Aracil M et al. Endothelial cell dif- cular integrin alpha v beta 3 for angiogenesis. Science
ferentiation into capillary-like structures in response to 1994; 264:569-571.
tumour cell conditioned medium: A modified chemotaxis 30. Brooks PC, Montgomery AM, Rosenfeld M et al. Integrin
chamber assay. Br J Cancer 1995; 71:770-775. alpha v beta 3 antagonists promote tumor regression by
10. Jackson CJ, Jenkins K, Schrieber L. Possible mechanisms inducing apoptosis of angiogenic blood vessels. Cell 1994;
of type I collagen-induced vascular tube formation. EXS 79:1157-1164.
1991; 59:198-204. 31. Bauer J, Margolis M, Schreiner C et al. In vitro model of
11. Nicosia RF, Bonanno E, Smith M. Fibronectin promotes angiogenesis using a human endothelium-derived perma-
the elongation of microvessels during angiogenesis in nent cell line: Contributions of induced gene expression,
vitro. J Cell Physiol 1993; 154:654-661. G-proteins, and integrins. J Cell Physiol 1992;
12. Clark RA. Potential roles of fibronectin in cutaneous 153:437-449.
wound repair. Arch Dermatol 1988; 124:201-206. 32. Ito A, Nomura S, Hirota S et al. Enhanced expression of
13. Schnaper HW, Kleinman HK, Grant DS. Role of laminin CD34 messenger RNA by developing endothelial cells of
in endothelial cell recognition and differentiation. Kid- mice. Lab Invest 1995; 72:532-538.
ney Int 1993; 43:20-25. 33. Szekanecz Z, Haines GK, Harlow LA et al. Increased syn-
14. Canfield AE, Schor AM. Evidence that tenascin and ovial expression of transforming growth factor (TGF)-!
thrombospondin-1 modulate sprouting of endothelial receptor endoglin and TGF-!1 in rheumatoid arthritis:
cells. J Cell Sci 1995; 108:797-809. Possible interactions in the pathogenesis of the disease.
15. Dejana E, Languino LR, Polentarutti N et al. Interaction Clin Immunol Immunopathol 1995; 76:187-194.
between fibrinogen and cultured endothelial cells. Induc- 34. Nguyen M, Strubel NA, Bischoff J. A role for sialyl Lewis-
tion of migration and specific binding. J Clin Invest 1985; X/A glycoconjugates in capillary morphogenesis. Nature
75:11-18. 1993; 365:267-269.
16. Vogel T, Guo NH, Krutzsch HC et al. Modulation of en- 35. Horak ER, Leek R, Klenk N et al. Angiogenesis, assessed
dothelial cell proliferation, adhesion, and motility by re- by platelet/endothelial cell adhesion molecule antibodies,
combinant heparin-binding domain and synthetic peptides as indicator of node metastases and survival in breast can-
from the type I repeats of thrombospondin. J Cell cer. Lancet 1992; 340:1120-1124.
Biochem 1993; 53:74-84.
Adhesion Molecules: Potent Inducers of Endothelial Cell Chemotaxis 277
36. Vogel T, Blake DA, Whikehart DR et al. Specific simple cytokine-activated human aortic endothelial cells: A pos-
sugars promote chemotaxis and chemokinesis of corneal sible role for ICAM-1 and sICAM-1 in atherosclerotic
endothelial cells. J Cell Physiol 1993; 157:359-366. aortic aneurysms. Clin Exp Immunol 1994; 98:337-343.
37. Klein S, Giancotti FG, Presta M et al. Basic fibroblast 48. Szekanecz Z, Shah MR, Harlow LA et al. Interleukin-8
growth factor modulates integrin expression in microvas- and tumor necrosis factor-alpha are involved in human
cular endothelial cells. Mol Biol Cell 1993; 4:973-982. aortic endothelial cell migration. Pathobiology 1994;
38. Enenstein J, Walehm NS, Kramer RH. Basic FGF and 62:134-139.
TGF-beta differentially modulate integrin expression of 49. Szekanecz Z, Shah MR, Pearce WH et al. Human athero-
human microvascular endothelial cells. Exp Cell Res 1992; sclerotic abdominal aortic aneurysms produce interleukin
203:499-503. (IL)-6 and interferon-gamma but not IL-2 and IL-4.
39. Taraboletti G, Belotti D, Giavazzi R. Thrombospondin Agents Actions 1994; 42:159-162.
modulates basic fibroblast growth factor activities on en- 50. Davis CA III, Pearce WH, Haines GK et al. Increased
dothelial cells. EXS 1991; 59:210-213. ICAM-1 expression in aortic disease. J Vasc Surg 1993;
40. Frank R, Adelmann-Grill BC, Herrmann K et al. 18:875-880.
Transformtin growth factor-beta controls cell-matrix in- 51. Pearce WH, Sweis I, Yao JST et al. Interleukin-1! and
teraction of microvascular dermal endothelial cells by tumor necrosis factor-# release in normal and diseased
downregulation of integrin expression. J Invest Dermatol human infrarenal aortas. J Vasc Surg 1992; 16:784-789.
1996; 106:36-41. 52. Koch AE, Kunkel SL, Pearce WH et al. Enhanced pro-
41. Mason JC, Kapahi P, Haskard DO. Detection of increased duction of the chemotactic cytokines interleukin-8 and
levels of circulating intercellular adhesion molecule 1 in monocyte chemoattractant protein-1 in human abdomi-
some patients with rheumatoid arthritis but not in pa- nal aortic aneurysms. Am J Pathol 1993; 142:1423-1428.
tients with systemic lupus erythematosus. Lack of corre- 53. Leibovich SJ, Polverini PJ, Shepard HM et al. Macrophage-
lation with levels of circulating vascular cell adhesion induced angiogenesis is mediated by tumour necrosis fac-
molecule 1. Arthritis Rheum 1993; 36:519-527. tor-#. Nature 1987; 329:630-632.
42. Koch AE, Turkiewicz W, Harlow LA et al. Soluble E- 54. Koch AE, Polverini PJ, Kunkel SL et al. Interleukin-8 as a
selectin in arthritis. Clin Immunol Immunopathol 1993; macrophage-derived mediator of angiogenesis. Science
69:29-35. 1992; 258:1798-1801.
43. Strieter RM, Polverini PJ, Kunkel SL et al. The functional 55. Rosen EM, Liu D, Setter E et al. Interleukin-6 stimulates
role of the ELR motif in CXC chemokine-mediated an- motility of vascular endothelium. EXS 1991; 59:194-205.
giogenesis. J Biol Chem 1995; 270:27348-27357. 56. Van Waes C. Cell adhesion and regulatory molecules in-
44. Strieter RM, Kunkel SL. Acute lung injury: The role of volved in tumor formation, hemostasis, and wound heal-
cytokines in the elicitation of neutrophils. J Investig Med ing. Head Neck 1995; 17:140-147.
1994; 42:640-651. 57. Koch AE, Kronfeld-Harrington LB, Szekanecz Z et al. In
45. Lloyd AR, Oppenheim JJ, Kelvin DJ et al. Chemokines situ expression of cytokines and cellular adhesion mol-
regulate T cell adherence to recombinant adhesion mol- ecules in the skin of patients with systemic sclerosis.
ecules and extracellular matrix proteins. J Immunol 1996; Pathobiology 1993; 61:239-246.
156:932-938. 58. Gailit J, Clark RA. Studies in vitro on the role of alpha v
46. Weber C, Alon R, Moser B et al. Sequential regulation of and beta 1 integrins in the adhesion of human dermal
alpha 4 beta 1 and alpha 5 beta 1 integrin avidity by CC fibroblasts to provisional matrix proteins fibronectin,
chemokines in monocytes: implications for vitronectin, and fibrinogen. J Invest Dermatol 1996;
transendothelial chemotaxis. J Cell Biol 1996; 106:102-108.
134:1063-1073. 59. Asahara T, Murohara T, Sullivan A et al. Isolation of
47. Szekanecz Z, Shah MR, Pearce WH et al. Intercellular putative progenitor endothelial cells for angiogenesis. Sci-
adhesion molecule-1 (ICAM-1) expression and soluble ence 1997; 275:964-967.
ICAM-1 (sIcellular adhesion molecule-1) production by
Facilitation of Healing
Angiogenesis
CHAPTER 26
Overview of Angiogenesis
A ngiogenesis is a cellular process that starts during embryogenesis and continues through
out the life of the organism. It is defined as the formation of new blood vessels by a
process of sprouting from preexisting vessels, while vasculogenesis is the differentiation of
endothelial cells from mesodermal precursors and their subsequent organization into a pri-
mary capillary plexus. Unlike angiogenesis, vasculogenesis is limited to the embryonic pe-
riod of life.1
Angiogenesis is a highly regulated process that is predominantly inhibited except for
brief periods of time in which the normally quiescent vasculature is stimulated to form new
blood vessels. Many disease states such as arthritis, ocular neovascularization, and tumor
growth and metastasis, occur due to unregulated angiogenesis.2 Although the factors that
induce angiogenesis are poorly understood, hypoxia is thought to be a potent stimulus.
Hypoxia induces various cell lines to secrete angiogenic molecules. It is not understood how
cells sense hypoxia or how this triggers secretion of angiogenic molecules.3
Stimulating factors for angiogenesis are often categorized as direct or indirect. This
often leads to confusing, and sometimes conflicting, data in the literature due to the various
systems in which the molecules are tested. Target cell specificity for an angiogenic molecule
is usually determined using in vitro methods such as endothelial cell proliferation and mi-
gration assays. Angiogenic activity, however, is analyzed in vivo using bioassays such as the
chicken chorioallantoic membrane assay. When the two methods correlate, the angiogenic
factor is classified as being “direct” because it stimulates endothelial cell proliferation or
migration directly. When the angiogenic factor fails to stimulate activity in vitro, it is classified
as “indirect” because it is assumed that the endothelial cell activity observed in vivo must be
induced by some other factor or cell type that the indirect angiogenic molecule elicits.2
The formation of new vessels occurs by activation of endothelial cells from pre-exist-
ing capillaries or venules. Pericytes are cells that closely resemble smooth muscle cells. Pericytes
closely envelop endothelial cells in quiescent capillaries, and they become sparse or absent
in growing vessels. They reappear in mature, nongrowing capillaries where they downregulate
endothelial cell proliferation.3
Angiogenesis is thought to occur in four basic steps. Endothelial cells of a capillary
become activated by a variety of factors including hypoxia, acidosis and cytokines. This is
followed by local vasodilation and increased vascular permeability, both of which are caused,
at least in part, by vascular endothelial growth factor (VEGF). The initial step of angiogen-
esis involves degradation of the basement membrane of the parent vessel. This allows the
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
280 Tissue Engineering of Prosthetic Vascular Grafts
endothelial cells to migrate toward the angiogenic stimulus, of FGF-2 and TGF-! allow extracellular matrix degradation
which is the second step of angiogenesis. Migrating endo- to occur in a controlled fashion, thereby allowing endothe-
thelial cells elongate and align with one another to form a lial cell migration for angiogenesis. A feedback mechanism
capillary sprout. Thirdly, endothelial cells proliferate, which has been described for these two angiogenic molecules. In-
occurs proximal to the migrating tip, further increasing the creased plasminogen activation by FGF-2 results in high
length of the sprout. The solid sprout gradually develops a pericellular plasmin activity. Plasmin activates TGF-! by re-
lumen proximal to the region of proliferation. Contiguous leasing it from its latent complex. Activated TGF-! decreases
sprouts anastomose at their tips to form a functional capil- the overall proteolytic capacity by increasing synthesis of
lary loop in which blood flow can be established. The fourth PAI-1 and decreasing synthesis of uPA. The resulting de-
step is vessel maturation, which is accomplished by recon- crease in plasmin activity blocks the activation of further
stitution of the basement membrane and the reappearance TGF-!. When active TGF-! is no longer present, the effects
of surrounding pericytes (see Fig. 26.1).1,4 of FGF-2 again prevail, thereby increasing plasmin and uPA
New capillaries, as well as blood vessels that are stimu- levels. Plasmin also releases FGF-2 that is bound to the ex-
lated by VEGF, tend to be “leaky” and thus have an increased tracellular matrix by heparan sulfate proteoglycans. The
vascular permeability. This leads to the accumulation of ex- cleavage of the proteoglycan core releases FGF-2 in an ac-
travascular fibrin, which acts as a provisional matrix that is tive form that is protected from proteolytic degradation and
progressively removed and replaced by other matrix com- can diffuse to affect target cells.8
ponents.1 Fibrin itself is chemotactic for inflammatory cells Degradation of the extracellular matrix and the re-
and has been shown to regulate endothelial cell and fibro- lease of the growth factors stored there are also enhanced by
blast migration.5 macrophages. Macrophages contribute to angiogenesis in
Degradation of the extracellular matrix is a highly several ways. Macrophages release proteolytic enzymes, in-
regulated process that occurs secondary to the interplay of cluding plasminogen activators and matrix metallo-
proteolytic enzymes, their inhibitors, and angiogenic mol- proteinases, that assist in extracellular matrix breakdown.
ecules that induce the formation of these enzymes. Endo- Macrophages also degrade heparan sulfate, which binds
thelial cells produce both plasminogen activators, tissue-type many growth factors to the extracellular matrix, thus releas-
plasminogen activator (tPA) and urokinase-type plasmino- ing active cytokines to affect endothelial cells. Finally, mac-
gen activator (uPA), as well as the plasminogen activator rophages themselves secrete many angiogenic factors such
inhibitor (PAI-1).6 The plasminogen activators cleave the as FGF-2, TGF-!, VEGF, and angiotropin as well as factors
inactive plasminogen to form plasmin, a proteolytic enzyme that inhibit angiogenesis. Macrophages are important regu-
with broad specificity for fibrin, laminin, gelatin, and other lators of inflammation and tissue healing, and angiogenesis
extracellular components. Plasmin also activates the matrix is an integral part of both of these processes.9
metalloproteinases, a family of proteolytic enzymes that also Many different cytokines and growth factors have been
degrade extracellular matrix components. 7 Fibroblast implicated in either promoting or inhibiting angiogenesis,
growth factor-2 (FGF-2), also known as basic FGF, is an- and in some cases the same factor may have both stimulat-
other important angiogenic molecule that enhances endo- ing and inhibitory properties depending upon local condi-
thelial growth, proliferation, and migration. It also enhances tions. We will review the functions of several of these major
endothelial cell expression of uPA and collagenase. In con- cytokines: FGF, VEGF, TGF-!, and angiostatin.
trast, transformtin growth factor-! (TGF-!) dramatically The FGF family of growth factors plays an important
increases PAI-1 synthesis in endothelial cells. The interplay role in angiogenesis. FGF-2 stimulates endothelial cell pro-
liferation, migration, and promotes formation of differen- meability occurs rapidly and is transient and reversible, per-
tiated capillary tubes in vitro.9 Endothelial cell production sisting for less than 30 minutes; it is not associated with any
of FGF-2 leads to its sequestration in the extracellular ma- detectable cell damage.12 This increased permeability leads
trix where it can be released to participate in angiogenesis to the accumulation of extravascular fibrin, which acts as a
during the degradation of the extracellular matrix. FGF-2 provisional matrix for migrating endothelial cells during
also induces the formation of plasminogen activators and angiogenesis.
collagenase, thereby promoting extracellular degradation VEGF induces endothelial cells to invade collagen gels
and its own activation.10 Both FGF-1, also known as acidic in vitro and also incites endothelial cell sprouting. Syner-
FGF, and FGF-2 are synthesized by a broad range of cell types gism between VEGF and FGF-2 was shown in a microvas-
including endothelial cells, fibroblasts, macrophages, and cular endothelial cell assay which demonstrated endothelial
smooth muscle cells. Both forms of FGF lack a signal se- invasion of a three dimensional collagen gel with formation
quence for secretion, and it is not entirely clear how FGF of capillary-like tubules. Both FGF-2 and VEGF were
release is regulated. As previously discussed, FGF binds to angiogenic in this model, with VEGF being about half as
heparan sulfate and can be released from the extracellular potent as FGF-2 in equimolar concentrations. However,
matrix. Furthermore, FGF is released from damaged and when both cytokines were added simultaneously, the angio-
dying cells and may therefore be important in inducing an- genic response was far greater than the additive effect of each
giogenesis in ischemic and injured tissues.3 For a more de- cytokine separately and occurred at a more rapid rate.13
tailed review of the functions of the FGF family of growth VEGF also promotes expression of tPA and matrix
factors in angiogenesis, see chapter 28. metalloproteinases in endothelial cells, which as already
VEGF, also known as vascular permeability factor, is discussed, induces degradation of the extracellular matrix
another important angiogenic molecule that is expressed in as well as the provisional fibrin matrix, releasing angiogenic
many different cell types including macrophages, fibroblasts, factors. PAI-1 is also increased by VEGF and may provide a
smooth muscle cells and endothelial cells. Hypoxia may, in negative feedback mechanism to balance the proteolytic
part, control the expression of VEGF.11 Unlike FGF, VEGF process.8,14
has a peptide leader sequence which probably controls cel- There are 4 major isoforms of VEGF and the relative
lular secretion of VEGF.10 Endothelial cells exhibit VEGF affinity to heparin binding may affect the bioavailability of
receptors, which explains the exquisite specificity of VEGF the various isoforms. VEGF-121 fails to bind heparin and is
for endothelial cells. Unlike FGF and other cytokines that therefore a freely diffusible protein that is secreted from cells.
induce endothelial proliferation as well as proliferation of VEGF-165, the major isoform, has an intermediate affinity
other cell lines, VEGF is generally considered to be a specific for heparin and is secreted from cells, although a significant
endothelial cell mitogen. Binding of VEGF to its receptor portion remains bound to the cell surface and the extracel-
induces a sequence of protein phosphorylation. VEGF in- lular matrix. Both VEGF-189 and VEGF-206 have a high af-
creases intracellular calcium levels and induces the release finity for heparin and these isoforms are almost completely
of von Willebrand factor from endothelial cells. sequestered in the extracellular matrix (see Fig. 26.2.).15,16
VEGF was originally discovered because of its ability The important role that VEGF plays in angiogenesis
to increase the permeability of blood vessels, primarily and vasculogenesis in embryo development is demonstrated
postcapillary venules and small veins. This increased per- by mouse VEGF knockout models. A heterozygous VEGF
knockout resulted in embryonic lethality between days 11 came confluent at 4 weeks. Transmural capillaries were ob-
and 12. Homozygous knockouts of the VEGF receptor re- served to connect the graft lumen to extravascular granula-
sults in embryonic lethality. tion tissue. At 2 weeks, multiple small orifices 100-500 ∝m
Another growth factor that exhibits both angiogenic apart were located between the ridges of the graft. At 4 and
and antiangiogenic properties is TGF-!. TGF-! is angiogenic 12 weeks, the entire graft surface was covered with endothe-
in vivo but inhibits macrovascular proliferation of endot- lial cells and the number of small vessel orifices had dimin-
helial cells and the proliferation of endothelial cells not en- ished. Smooth muscle cells were seen underneath the en-
gaged in angiogenesis in vitro. TGF-! promotes prolifera- dothelial cells in the network of microvessels within the graft
tion of endothelial cells engaged in angiogenesis in vitro and as early as 2 weeks. A progressive thickening of the intima,
induces the formation of capillary-like tubes of microvas- which was the greatest at 12 weeks, was seen throughout the
cular endothelial cells. TGF-! potentiates FGF-2 and VEGF graft. Smooth muscle cells, which were thought to be de-
induced angiogenesis in vitro at low, but not high, doses.3 rived from pericytes, accompanied the capillary endothelial
As previously discussed, TGF-! modulates extracellular deg- cells into the graft lumen. In contrast, intimal thickening
radation by inducing PAI-1 synthesis and thus downregulates occurred only at the anastomotic ends in the 30 ∝m intern-
plasmin-mediated proteolysis. In view of its capacity to di- odal grafts (see Fig. 26.3.). These results indicate that capil-
rectly inhibit endothelial cell proliferation and migration, lary endothelium and smooth muscle cells can function in
reduce extracellular proteolysis, and promote matrix depo- the same manner as their large vessel counterparts and could
sition in vitro, TGF-! has been proposed to be a potential provide coverage of synthetic graft surfaces in contact with
mediator of the resolution phase of angiogenesis.1 the arterial circulation.24
Although many factors promote angiogenesis, uncon- The 60 ∝m internodal graft appeared to provide the
trolled angiogenesis is associated with many disease states, “optimal” porosity in the baboon model. In low porosity
including tumor formation and metastasis. It is therefore grafts, 10 and 30 ∝m internodal distance, endothelial cover-
the delicate balance of angiogenic and antiangiogenic mol- age was incomplete and was limited to the graft surface near
ecules that leads to controlled, appropriate angiogenesis. the anastomoses. In contrast, high porosity grafts, 60 and
Folkman and associates17 have recently isolated angiostatin, 90 ∝m internodal distance, revealed complete endothelial
an antiangiogenic molecule that inhibits the formation of coverage with a uniformly distributed intimal thickening
tumor metastases in the Lewis mouse lung model. throughout the graft. However, the 90 ∝m graft developed
Angiostatin is a 38 kDa fragment of plasminogen that in- areas of focal endothelial loss at 3 months, suggesting that
hibits endothelial cell proliferation in vitro. The primary the larger pore size resulted in intimal instability and en-
tumor secretes angiostatin, which inhibits the growth of dothelial cell loss in the ePTFE.25
metastases. Removing the primary tumor decreases Dacron, another porous graft material, was compared
angiostatin levels and this allows the metastases to reestab- to the 60 ∝m internodal ePTFE grafts in the baboon model.
lish their blood supply and grow. The Dacron grafts showed incomplete endothelialization at
12 weeks while all the ePTFE grafts revealed a confluent en-
Angiogenesis in Vascular Grafts dothelial layer. Although both types of grafts healed by pro-
Synthetic, small diameter vascular grafts fail prima- liferation of cells derived from invading capillaries, the au-
rily because of interactions at the blood-material and tis- thors hypothesized that the Dacron material itself may some-
sue-material interfaces.18 Currently available prostheses elicit how inhibit graft endothelialization.26
inflammatory and regenerative reactions, yielding graft Dacron may induce various cell types to secrete fac-
thrombosis and intimal hyperplasia.19 In humans, clinically tors that could inhibit angiogenesis and graft
utilized grafts of Dacron and 30 ∝m internodal distance ex- endothelialization. Evidence supporting this hypothesis is
panded polytetrafluorethylene (ePTFE) do not allow capil- provided by Greisler et al.27 Rabbit peritoneal macrophages,
lary infiltration despite the small wall thickness. Many re- grown in the presence of Dacron particles in vitro, released
searchers have attempted to improve synthetic grafts by us- more TGF-! than control macrophages. The effects of TGF-!
ing various techniques such as endothelial seeding20,21 and on angiogenesis and endothelial proliferation are complex.
bioresorbable grafts.19 Clowes and associates studied the ef- In vitro, TGF-! inhibits endothelial cell proliferation.28
fect of porosity on ePTFE grafts and demonstrated that these TGF-! instillation into newborn mice29 and into the chorio-
grafts could be lined by endothelial cells that were derived allantoic membrane assay30 induces angiogenesis, while
from graft transmural capillaries. This induction of “graft higher doses of TGF-! resulted in inhibition of angiogen-
angiogenesis” occurred in ePTFE grafts with 60 ∝m intern- esis in these models.31 The higher levels of TGF-! released
odal distance. Previous studies had shown that ePTFE grafts by macrophages in the presence of Dacron may contribute
with a 30 ∝m internodal distance were covered by an in- to the inhibition of endothelialization seen in Dacron grafts.
growth of endothelium from the adjacent artery as a con- In an attempt to apply the above research to the clini-
tinuous sheet across the anastomosis and along the graft.22 cal setting, 10 above-knee femoropopliteal composite grafts
This process was relatively slow with only 60% of grafts were placed into 8 patients. Each of these grafts consisted of
6-9 cm in length exhibiting complete coverage by 12 months equal lengths of 30 and 60 ∝m internodal distance ePTFE
in an aortoiliac baboon model.23 In contrast, when 60 ∝m with the 60 ∝m portion being randomly placed in either the
internodal grafts were used, endothelial cells began to ap- proximal or distal position. In contrast to the baboon ex-
pear on the graft surface at 1-2 weeks after surgery and be- periments which used unwrapped ePTFE, these grafts were
Angiogenesis in Tissues and Vascular Grafts 283
reinforced with a thin, nonexpanded wrap for human use. to the death of the host. On the other hand, tissue ischemia
Noninvasive assessment of endothelialization was performed caused by occlusive arteriosclerosis represents a relative de-
using 111Indium-labeled platelet imaging at 1 week and 3 ficiency of angiogenesis. Various cytokines have been used
months after surgery. There was no difference in indium to induce angiogenesis in animal models of tissue ischemia.
uptake between the 60 and 30 ∝m segments at either time Perhaps one of the most commonly used is the rabbit
point. Histologic sections were available from 60 ∝m seg- hindlimb ischemia model. The hindlimb is made ischemic
ments from 2 patients who underwent operations for graft by tying off the external iliac artery and removing the com-
thrombosis. Capillary ingrowth into the graft was observed, mon femoral and superficial femoral arteries. Growth fac-
but it rarely extended more than half the distance from the tors, such as FGF (reviewed in chapter 28) and VEGF, are
outside of the graft to the lumen. Smooth muscle cells were then introduced into the ischemic limb and their angiogenic
not seen on the luminal surface, indicating that a neointima effect is measured by counting the new vessels seen on an-
had not formed. The finding that capillary ingrowth can giography. VEGF has been extensively studied by Isner’s
occur in the 60 ∝m internodal graft, but does not lead to an group using this model. They found that a single bolus in-
endothelial lining in humans, could possibly be attributed jection of VEGF via the ipsilateral internal iliac artery caused
to an inadequacy of angiogenesis in adult humans or retar- an increase in flow at rest, in maximum flow velocity, and in
dation of capillary ingrowth by the reinforcing wrap.32 maximum blood flow at 30 days as compared to saline con-
Other attempts at inducing graft angiogenesis by coat- trols.35 VEGF treated rabbits also re-established an increased
ing the luminal surface of synthetic grafts with FGF-1 endothelium-dependent blood flow in response to seroto-
incorporated into a fibrin glue have been made by Greisler nin and acetylcholine injection as compared to saline treated
and associates. FGF-1 is a potent endothelial cell mitogen animals which did not respond to the serotonin or acetyl-
and the greater adhesivity of the FG surface for seeded choline injections, suggesting that VEGF facilitated endot-
endothelial cells plus the slow release of the suspended FGF-1 helial recovery after ischemic injury.36 VEGF also increased
suggest the tissue engineering applicability of this sub- endothelial cell proliferation 2.8-fold at day 5, followed by a
strate 33,34 For a more thorough review of this work, decrease to baseline by day 7 as compared to controls, while
see chapter 28. smooth muscle cell proliferation increased by 2.7-fold.37
Endogenous VEGF production in the ischemic hindlimb
Therapeutic Angiogenesis in Ischemic Tissues model was found to be decreased in older (4-5 years) as com-
Unregulated angiogenesis can be seen in many can- pared to younger (6-8 months) rabbits and the angiogenic
cers and is a biologically undesirable situation that often leads response was also decreased in the older rabbits. Exogenous
administration of VEGF induced a similar angiogenic re- Another human trial with transfection of phVEGF-165
sponse in both older and younger rabbits, suggesting that a by intramuscular injection or intraarterially revealed in-
relative VEGF deficiency may occur with aging and that re- creased serum levels of VEGF. Furthermore, 63% of the pa-
placement of VEGF may overcome this defect.38 This rela- tients had edema and the response seemed to correlate with
tive VEGF deficiency in response to ischemia was also seen disease severity, with no edema occurring in patients with
in diabetic mice. The diabetic mice exhibited less angiogen- claudication, variable amounts of edema occurring in pa-
esis and produced less VEGF than their normal mice coun- tients with rest pain, and profound edema occurring in pa-
terparts.39 Another model used by this group involved bal- tients with ischemic tissue loss.45 Isner’s group also reported
loon injury to the rat carotid artery with a 30 minute inocu- on a trial of 10 limbs in 9 patients with ulcers and rest pain
lation with either saline or VEGF. The VEGF treated ani- that received IM gene transfer two times at 4 week intervals.
mals re-endothelialized more quickly and exhibited less The clinical status was improved in 8, unchanged in 1, and
neointimal thickening.40 worse in 1 limb after a 3.7 month follow up. The patients
After establishing the angiogenic effect of the VEGF overall demonstrated an increased ankle-brachial index,
protein in ischemia, Isner’s group then proceeded to study improved blood flow by magnetic resonance angiography,
the effect of naked plasmid DNA encoding for human and angiographic evidence of new collaterals in 6/10 limbs.46
VEGF-165 (phVEGF-165). Using the rabbit ischemic hindlimb Further clinical trials are currently being performed using
model, they injected the phVEGF-165 intramuscularly; 30 this gene transfer technique.
days later, there were increased collateral vessels and histo- Yet another route for delivering VEGF to promote graft
logically identifiable capillaries were also seen. In addition, angiogenesis was used by Weatherford and associates.47 They
there was a superior calf blood pressure ratio in the incorporated VEGF and heparin into fibrin glue and found
phVEGF-165 treated group. They felt that ischemic skeletal that human aortic endothelial cell proliferation on this glue
muscle was a good target for gene therapy and that VEGF was increased as compared to fibrin glue alone. The addi-
was a good gene to use.41 Hypoxia upregulates VEGF recep- tion of heparin to the VEGF increased endothelial cell pro-
tor gene expression making ischemic tissues more likely to liferation while inhibiting smooth muscle cell proliferation.
respond to VEGF than normal tissues. VEGF also promotes The application of VEGF and heparin in fibrin glue to a pros-
physiological maturation of newly formed collateral chan- thetic vascular graft may be used in the future to promote
nels and restores appropriate endothelium-derived relaxing graft patency and endothelialization.
factor as well as the nitric oxide-dependent vasorelaxation
response initiated by the endothelium, thereby increasing Future Directions
the blood flow through the expanding vascular bed.42 Research into the mechanisms of angiogenesis is a rela-
An alternate method to deliver the phVEGF-165 in- tively new scientific endeavor and much progress has been
volved transfecting endothelial cells by balloon catheter made in a relatively short period of time. Control of angio-
angioplasty in the rabbit femoral artery. The phVEGF-165 genesis could provide enormous clinical benefits. Inhibition
treated animals showed enhanced endothelialization in the of angiogenesis could result in new treatment strategies for
transfected side as well as the nontransfected, control side, cancer, while promoting angiogenesis could relieve myocar-
decreased intimal thickening, decreased thrombotic occlu- dial and peripheral ischemia from atherosclerotic occlusive
sion, and accelerated recovery of endothelial-dependent va- disease. Other emerging technologies will probably utilize
somotor reactivity as compared to transfection with the LacZ our increasing knowledge of angiogenesis regulation to in-
gene in control animals.43 duce new vessel formation in ischemic tissues, perhaps some-
Recently, this work with the phVEGF-165 has been per- day making bypass grafting for chronic arterial occlusive
formed in humans. A patient with nonreconstructable oc- disease obsolete.
clusive disease received an angioplasty in a normal-appear-
ing section of her distal popliteal artery to transfect the sur- References
rounding endothelial cells with phVEGF-165. A normal sec- 1. Pepper MS. Manipulating angiogenesis, from basic science
tion of artery was chosen to enhance cellular transfection to the bedside. Arteriosclerosis Thromb Vasc Biol 1997;
and to avoid the confounding factor of increased blood flow 17:605-619.
2. Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992;
that may result from treating a stenotic segment of artery.
267:10931-10934.
The patient exhibited increased collaterals at 4 and 12 weeks 3. Battegay EJ. Angiogenesis: Mechanistic insights,
as demonstrated by angiography and increased resting and neovascular diseases, and therapeutic prospects. J Mol
maximal blood flows. Three sporadic angiomas developed Med 1995; 73:333-346.
on the foot and ankle of the treated leg. One of the angio- 4. Cotran RS. Inflammation and repair. In: Cotran RS,
mas was excised, revealing a proliferative endothelium and Robbins SL, Kumar V, eds. Robbins Pathologic Basis of
the other 2 regressed spontaneously. The patient also devel- Disease, 5th Edition. Philadelphia: WB Saunders Co.,
oped leg edema 1 week after gene transfer which resolved in 1994:51-92.
4 weeks, corresponding to the time course of VEGF 5. Brown LF, Dvorak AM, Dvorak HF. Leaky vessels, fibrin
production observed in animal models after transfection. deposition, and fibrosis: A sequence of events common
The treatment, however, was ultimately unsuccessful and the to solid tumors and to many other types of diseases. Am
Rev Respir Dis 1989; 140:1104-1107.
patient required a below-knee amputation 5 months after
the transfection.44
Angiogenesis in Tissues and Vascular Grafts 285
6. Loskutoff DJ, Curriden SA. The fibrinolytic system of the 23. Clowes AW, Kirkman TR, Clowes MM. Mechanism of
vessel wall and its role in the control of thrombosis. Ann arterial graft failure. II. Chronic endothelial and smooth
NY Acad Sci 1990; 598:238-247. muscle cell proliferation in healing poytetrafluoroethylene
7. Murphy G, Atkinson S, Ward R, Gavrilovic J, Reynolds prostheses. J Vasc Surg 1986; 3:877-884.
JJ. The role of plasminogen activators in the regulation 24. Clowes AW, Kirkman TR, Reidy MA. Mechanisms of ar-
of connective tissue metalloproteinases. Ann of NY Acad terial graft healing, rapid transmural capillary ingrowth
Sci 1992; 667:1-12. provides a source of intimal endothelium and smooth
8. Pintucci G, Bikfalvi A, Klein S, Rifkin DB. Angiogenesis muscle in porous PTFE prostheses. Am J Pathol 1986;
and the fibrinolytic system. Seminars in Thrombosis and 123:220-230.
Hemostasis. 1996; 22:517-524. 25. Golden MA, Hanson SR, Kirkman TR, Schneider PA,
9. Sunderkotter C, Steinbrink K, Goebeler M, Bhardwaj R, Clowes AW. Healing of polytetrafluoroethylene arterial
Sorg C. Macrophages and angiogenesis. J Leukoc Biol grafts is influenced by graft porosity. J Vasc Surg 1990;
1994; 55:410-422. 11:838-845.
10. Schott RJ, Morrow LA. Growth factors and angiogenesis. 26. Zacharias RK, Kirkman TR, Clowes AW. Mechanisms of
Cardio Res 1993; 27:1155-1161. healing in synthetic grafts. J Vasc Surg 1987; 6:429-436.
11. Namiki A, Brogi E, Kearney M, Kim EA, Wu T, 27. Greisler HP, Petsikas D, Cziperle DJ, Murchan PM,
Couffinhal T, Varticovski L, Isner JM. Hypoxia induces Henderson SC, Lam TM. Dacron stimulation of macroph-
vascular endothelial growth factor in cultured human en- age transformtin growth factor-! release. Cardiovasc Surg
dothelial cells. J Biol Chem 1995; 52:31189-31195. 1996; 4:169-173.
12. Brown LF, Detmar M, Claffey K, Nagy JA, Feng D, Dvorak 28. Frater-Schroeder M, Risau W, Hallmann R et al. Tumor
AM, Dvorak HF. Vascular permeability factor/vascular necrosis factor type #, a potent inhibitor of endothelial
endothelial growth factor: A multifunctional angiogenic cell growth in vitro, is angiogenic in vivo. Proc Natl Acad
cytokine. EXS 1997; 79:233-269. Sci USA. 1987; 84:5277-5281.
13. Pepper MS, Ferrara N, Orci L, Montesano R. Potent syn- 29. Roberts AB, Sporn MB, Assoian RK et al. Transforming
ergism between vascular endothelial growth factor and ba- growth factor type !: Rapid induction of fibrosis and an-
sic fibroblast growth factor in the induction of angiogen- giogenesis in vivo and stimulation of collagen formation
esis in vitro. Biochem and Biophys Res Comm 1992; in vitro. Proc Natl Acad Sci USA. 1986; 83:4167-4171.
189:824-831. 30. Yang EY, Moses HL. Transforming growth factor beta 1-
14. Pepper MS, Ferrara N, Orci L, Montesano R. Vascular induced changes in cell migration, proliferation, and an-
endothelial growth factor (VEGF) induces plasminogen giogenesis in the chicken chorioallantoic membrane. J Cell
activators and plasminogen activator inhibitor-1 in mi- Biol 1990; 111:731-741.
crovascular endothelial cells. Biochem and Biophys Res 31. Moses HL, Yang EY, Pietenpol JA. TGF-! stimulation and
Comm 1991; 181:902-906. inhibition of cell proliferation: New mechanistic insights.
15. Ferrara N, Davis-Smyth T. The biology of vascular en- Cell 1990; 63:245-247.
dothelial growth factor. Endocrine Rev 1997; 18:4-23. 32. Kohler TR, Stratton JR, Kirkman TR, Johansen KH,
16. Park JE, Keller G-A, Ferrara N. The vascular endothelial Zierler BK, Clowes AW. Conventional versus high-poros-
growth factor (VEGF) isoforms: Differential deposition ity polytetrafluoroethylene grafts: Clinical evaluation. Sur-
into the subepithelial extracellular matrix and bioactivity gery 1992; 112:901-907.
of extracellular matrix-bound VEGF. Mol Biol Cell 1993; 33. Greisler HP, Cziperle DJ, Petsikas D, Murchan PM,
4:1317-1326. Appelgren EO, Drohan W, Burgess WH, Kim DU. En-
17. O’Reilly MS, Holmgren L, Shing Y, Chen C, Rosenthal hanced endothelialization of expanded PTFE grafts by
RA, Moses M, Lane WS, Cao Y, Sage EH, Folkman J. heparin binding growth factor-type 1 (HBGF-1) pretreat-
Angiostatin: A novel angiogenesis inhibitor that mediates ment. Surgery 1992; 112:244-255.
the suppression of metastases by a Lewis lung carcinoma. 34. Zarge JI, Huang P, Husak V, Kim DU, Haudenschild CC,
Cell 1994; 79:315-328. Nord RM, Greisler HP. Fibrin glue containing fibroblast
18. Greisler HP, Ellinger J, Henderson SC, Shaheen AM, Bur- growth factor type 1 and heparin with autologous endot-
gess WH, Kim DU, Lam TM. The effects of an athero- helial cells reduces intimal hyperplasia in a canine carotid
genic diet on macrophage/biomaterial interactions. J Vasc artery balloon injury model. J Vasc Surg 1997; 25:840-849.
Surg 1991; 14:10-23. 35. Bauters C, Asahara T, Zheng LP, Takeshita S, Bunting S,
19. Greisler HP, Schwarcz TH, Ellinger J, Kim DU. Dacron Ferrara N, Symes JF, Isner JM. Physiological assessment
inhibition of arterial regenerative activities. J Vasc Surg of augmented vascularity induced by VEGF in ischemic
1986; 3:747-756. rabbit hindlimb. AM J Physiol. 1994; 267 (Heart Circ
20. Zilla P, Deutsch M, Meinhart J, Puschmann R, Eberl T, Physiol. 36): H1263-H1271.
Minar E, Dudczak R, Lugmaier H, Schmidt P, Noszian I, 36. Bauters C, Asahara T, Zheng LP, Takeshita S, Bunting S,
Fischlein T. Clinical in vitro endothelialization of Ferrara N, Symes JF, Isner JM. Recovery of disturbed en-
femoropopliteal bypass grafts: An actuarial follow-up over dothelium-dependent flow in the collateral-perfused rab-
three years. J Vasc Sug. 1994; 19:540-548. bit ischemic hindlimb after administration of vascular
21. Zilla P, Preiss P, Groscurth P, Rosemeier F, Deutsch M, endothelial growth factor. Circulation 1995; 91:2802-2809.
Odell J, Heidinger C, Fasol R, von Oppell U. In vitro- 37. Takeshita S, Rossow ST, Kearney M, Zheng LP, Bauters
lined endothelium: Initial integrity and ultrastructural C, Bunting S, Ferrara N, Symes JF, Isner JM. Time course
events. Surgery 1994; 116:524-534. of increased cellular proliferation in collateral arteries af-
22. Clowes AW, Gown AM, Hanson ST, Reidy MA. Mecha- ter administration of vascular endothelial growth factor
nisms of graft failure. I. Role of cellular proliferation in in a rabbit model of lower limb vascular insufficiency. Am
early healing of PTFE prosthesis. Am J Pathol 1985; J Pathol 1995; 147:1649-1660.
118:43-54.
286 Tissue Engineering of Prosthetic Vascular Grafts
38. Rivard A, Fabre J-E, Silver M, Murohara T, Chen D, endothelial integrity and endothelium-dependent function
Asahara T, Isner JM. Age-dependent impairment of an- after phVEGF- 165 gene transfer. Circulation 1996;
giogenesis. Circulation (Suppl) 1997; 96:I-18. 94:3291-3302.
39. Rivard A, Silver M, Fabre J-E, Magner M, Kearney M, 44. Isner JM, Pieczek A, Schainfeld R, Blair R, Haley L,
Isner JM. Diabetes impairs angiogenesis in limb ischemia. Asahara T, Rosenfield K, Razvi S, Walsh K, Symes JF.
Circulation (Suppl) 1997; 96:I-175. Clinical evidence of angiogenesis after arterial gene trans-
40. Asahara T, Bauters C, Pastore C, Kearney M, Rossow S, fer of phVEGF-165 in patient with ischaemic limb. Lan-
Bunting S, Ferrara N, Symes JF, Isner JM. Local delivery cet 1996; 348:370-374.
of vascular endothelial growth factor accelerates 45. Baumgartner I, Pieczek AM, Blair R, Manor O, Walsh K,
reendothelialization and attenuates intimal hyperplasia in Isner JM. Evidence of therapeutic angiogenesis in patients
balloon-injured rat carotid artery. Circulation 1995; with critical limb ischemia after intramuscular phVEGF-
91:2793-2801. 165 gene transfer. Circulation (Suppl) 1997; 96:I-32.
41. Tsurumi Y, Takeshita S, Chen D, Kearney M, Rossow ST, 46. Baumgartner I, Magner M, Pieczek AM, Isner JM. Clini-
Passeri J, Horowitz JR, Symes JF, Isner JM. Direct intra- cal evidence that vascular endothelial growth factor en-
muscular gene transfer of naked DNA encoding vascular hances vascular permeability. Circulation (Suppl)
endothelial growth factor augments collateral development 1997;-96:I-4.
and tissue perfusion. Circulation 1996; 94:3281-3290. 47. Weatherford DA, Sackman JE, Reddick TT, Freeman MB,
42. Majesky MW. A little VEGF goes a long way, therapeutic Stevens SL, Goldman MH. Vascular endothelial growth
angiogenesis by direct injection of vascular endothelial factor and heparin in a biologic glue promotes human
growth factor-encoding plasmid DNA. Circulation 1996; aortic endothelial cell proliferation with aortic smooth
94:3062-3064. muscle cell inhibition. Surgery 1996; 120:433-439.
43. Asahara T, Chen D, Tsurumi Y, Kearney M, Rossow S,
Passeri J, Symes JF, Isner JM. Accelerated restitution of
Facilitation of Healing
Angiogenesis
CHAPTER 27
Polypeptide Growth Factors with a Collagen Binding Domain:
Their Potential for Tissue Repair and Organ Regeneration
Bo Han, Lynn L.H. Huang, David Cheung, Fabiola Cordoba, Marcel Nimni
Introduction
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler
©1999 R.G. Landes Company.
288 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 27.2. Modified reconstituted or engineered biosynthetic extracellular matrices containing various portions of native components
and thiolated gelatin.
3. Glycosaminoglycan are also used to control the me- 2. Fibronectin molecules can assemble into fibrils as
chanical behavior, e.g., as a modulator of the ‘cables and connectors’ for linking cells to collagen
elasticity. and fibrin;11
It has been reported that certain collagen-glycosami- 3. Through binding to cell surface receptors, fibronectin
noglycan polymers have delayed or arrested the contraction can transduce signals internally to modulate cellu-
of full-thickness skin wounds. At least three conditions must lar function.14
be met to prevent wound contraction: Yaffe and Shoshan15 studied the behavior of gingival
1. The enzymatic degradation rate of the implant must epithelial cells cultured in dishes with gingival epithelial ex-
be below a certain threshold value; tracellular matrix. It was found that the movement of epi-
2. The average pore size must within the range of thelial cells stops at the contact point with extracellular ma-
20-500 ∝m in order to allow proper distribution of trix, and that this results in the piling of cells into several
mesenchymal cells; layers. This suggested that the movement of epithelial cells
3. Epidermal (basal) cells must be seeded into the po- may be influenced by collagen fibers in vivo. This also im-
rous implant before grafting. plies a role for collagen in preventing epithelial cells from
penetrating into the lower layer.
Interactions of Biosynthetic Matrices The studies of Donaldson et al16 failed to demonstrate
with Cells collagen type specificity for supporting the migration of
epidermal cells. Denatured collagen and glutaraldehyde
Cell Migration on Biosynthetic Matrices crosslinked collagen gels are also able to support the migra-
Cell migration requires a solid support such as an ex- tion of epidermal cells. It is found that the tertiary structure
tracellular matrix or a basement membrane. In the process of the collagen molecule is unimportant for the binding of
of wound healing, certain cells migrate extensively in a highly epidermal cells. It is also reported that the #1 chain of type I
organized manner.11 Evidence for the mechanisms of orga- collagen has at least three, and most likely more, epidermal
nized cell movement are: binding sites.
1. Cells are bound to the extracellular matrix through Epidermal growth factor (EGF) and transformtin
a variety of large glycoproteins;11-13 growth factor-! (TGF-!) have been shown to play a role in
290 Tissue Engineering of Prosthetic Vascular Grafts
stimulating the growth of epithelial cells.17,18 The structures mis-like appearance. It was suggested that both physical
of EGF and TGF-! are similar and both bind to the same forces and secreted factors from fibroblasts such as
cell surface receptor, tyrosine kinase.19 The interactions fibronectin or other proteoglycans contribute to the reor-
between the receptor and growth factors lead to intracellu- ganization of the collagen lattices.
lar changes in preparing cells for DNA synthesis and
proliferation. Barrandon and Green18 found that the effects Influence of Cells on the Contraction
of EGF and TGF-! in promoting cell proliferation depend of Collagen Matrices
on their abilities to increase the rate of cell migration, which Allen and Schor25 quantitatively measured the con-
is essential in the process of wound healing. Therefore, traction of collagen by fibroblasts. The spherical fibroblasts
appropriate application of EGF or TGF-! during tissue were initially incubated in the attached collagen lattice. Over
regeneration should be able to promote the re-epithelial- 90% of the cells change the morphology to stellate cells af-
ization process. ter incubation for two hours, but most of them subsequently
retract to a spherical morphology when the matrices are
Cell Infiltration into Biosynthetic Matrices carefully detached to produce a floating culture. Collagen
When cells are cultured on a hydrated collagen gel, matrices reduce to about one half of the original size within
the shape and motility of cells are very similar to cells in the first two hours in a floating culture. This initial con-
vivo.13 In addition to the biological effects of hyaluronan traction rate of collagen is proportional to the cell number
demonstrated by Huang et al5,6 Doillon et al20 have shown within the range of 105-106 cells/gel. The contraction of the
that the presence of either fibronectin or hyaluronic acid or collagen matrix appears to be the result of cell-collagen in-
both in a collagen sponge can enhance wound healing in teraction and collagen reorganization caused by fibroblasts
vivo. It has been found that fibroblasts can infiltrate and that exert a tension upon the surrounding collagen fibers. It
proliferate through the entire sponge, and subsequently in- is also suggested that cell-cell interaction does not contrib-
crease collagen biosynthesis. The effect of hyaluronic acid is ute significantly to the contraction process, since stellate fi-
to promote cellular infiltration and that of fibronectin is to broblasts and spherical fibroblasts are homogeneously dis-
mediate cell attachment to the collagen fibrils. Therefore, tributed within the collagen matrix in a floating culture.
incorporation of hyaluronic acid or fibronectin into collagen Bell et al also reported that human fibroblasts of dif-
sponges appears to improve the properties of the regener- ferent proliferative potential contract collagen lattices with
ated tissue.21 equal efficiency.26 They further found that the contraction
rate of collagen lattices can be regulated by varying the pro-
Influence of Cells on Matrix Reorganization tein content of the lattice, the cell number, or the concen-
Denefle et al22,23 used unstriated fibrils of purified tration of inhibitors such as colcemid. Inhibition of gel con-
type I collagen gels to study the anchoring process of traction by cytochalasin B and colchicine indicate that the
keratinocytes and their influences on collagen organization. integrity of the cytoskeletons inside the fibroblasts is im-
Two types of collagen striated fiber assemblies seem to ap- portant for the mechanism of gel contraction.
pear when isolated epithelium is superimposed on a col-
lagen gel. Numerous extended filopodia of cells arise at the Influence of Collagen Derived Matrices on Cells
contact sides with one type of collagen fiber assembling To improve the hematocompatibility of cardiovascu-
around and along them. In the inner side of the gel matrix, lar materials, Lee et al27,28 chemically crosslinked collagen
collagen fibers assume different shapes and widths depend- to polyurethane and investigated the growth of endothelial
ing on the concentration of acid soluble collagen.22 To fur- cells on the surface. A flossy-like collagen formed on the
ther distinguish the effect of preexisting fibronectin from polyurethane surface when the crosslinking reaction was
that of keratinocytes on the reorganization of collagen gels, carried out with (N-ethyl-N’-3-dimethyl aminopropyl)car-
frog skin fragments opposed to collagen coatings were stud- bodiimide at pH 5. A fibrillated collagen network was fur-
ied either in the presence or absence of fetal calf serum. The ther reconstituted on the polyurethane surface under physi-
results indicate that keratinocytes in the absence of serum ological conditions, using either the flossy-like collagen as a
appear to influence fibril organization of purified type I col- nucleus or an epoxy crosslinking reaction. When cultured
lagen gel. Pre-existing fibronectin appears to enhance the with endothelial cells, the collagen fibrillar networks better
capability of keratinocytes to modify collagen fibers, since supported the cell growth. The amounts as well as the den-
the presence of serum causes the collagen morphology to sity and diameter of collagen fibrils grafted on the polyure-
closely resemble that of the native wound healing process.23 thane surfaces correlated with the proliferation of endothe-
Grinnell and Lamke24 studied the reorganization of lial cells.
hydrated collagen lattices by human skin fibroblasts. They
found collagen reorganization to be similar whether the cells Endothelial cells in Tissue Engineering
were cultured on the top or at the bottom of the lattices. The main function of endothelial cells is to maintain
Initially, proximal collagen fibrils were aligned along the surface integrity, such as in the lining of heart, blood vessels
plane of cell spreading. Subsequently, the collagen fibrils and lymphatic vessels. They reduce the friction at the fluid
distal to the cells underwent reorganization. If the collagen to lumen interface, as well as control the flow of blood and
lattice is detached from the underlying substratum, the col- passage of cells in the circulation out of the vascular system
lagen fibrils roll up into a compact form which has a der- into the surrounding tissues.29 Resting endothelium ex-
Polypeptide Growth Factors with a Collagen Binding Domain: Their Potential for Tissue Repair and Organ Regeneration 291
presses low levels of adhesion molecules that prevent the branching tubular networks occurred. Based on these re-
binding of leukocytes and inhibit coagulation. Infection or sults they proposed that FGF-stimulated endothelial cells
injury stimulates the endothelium to express cell surface may be “switched” between growth and differentiation as a
molecules and adhesion proteins that promote cell interac- response to the mechanical integrity of their extracellular
tions.30 There has been a tremendous resurgence of interest matrices. The influence of the extracelluar matrix on en-
concerning the modulation of endothelial cell function by dothelial behavior is likely to be mediated through integrins,
factors, especially cytokines, and by the composition of the a family of heterodimeric cell surface receptors which trans-
extracellular matrix; this has encouraged the development mit signals from the extracellular matrix by organizing the
of endothelial cell culture techniques.31,32 Endothelial cells cytoskeleton, thus regulating cell shape, internal cellular ar-
cultured from the umbilical vein can be induced to switch chitecture and other cell functions.42 While Matrigel sup-
to a capillary-like morphology on interaction with base- ports the attachment, proliferation and differentiation of
ment.33-35 This morphologenesis results in formation of cel- endothelial cells, type I collagen containing alpha-elastin was
lular cords containing a central lumen surrounded by one found to inhibit the proliferation and migration of vascular
to three endothelial cells, and is thought to parallel capillary endothelial cells. While others found that type I collagen
formation in later stages of angiogenesis in vivo.33-35 Since inhibits the growth of endothelial cells, Lee et al found that
endothelial cells in vivo are organized in monolayers cover- endothelial cell growth is supported by large reconstituted
ing the basement membrane of the vessels, much work has type I collagen fibrils formed from collagen dialyzed in phos-
been done using in vitro models with endothelial cells seeded phate buffered saline.28 Chen et al studied human and bo-
on basement membrane equivalents such as Matrigel, re- vine capillary cells grown on surfaces coated with
constituted from solubilized basement membrane compo- micropatterned extracellular matrix substrates.43 As the ex-
nents such as type IV collagen, laminin and fibronectin ex- tracellular matrix-coated adhesive islands decreased in size,
tracted from Engelbreth-Holm-Swam mouse sarcoma.36-39 cell extension became more restricted and the endothelial
However, endothelial cells can also be successfully cultivated cells began to switch from growth to apoptosis. Spacing be-
on gelatin or poly-L-lysine-coated surfaces. The cultivation tween different adhesive islands also affected cell spreading.
of microvascular endothelial cells in Matrigel can result in Changes in the size and spacing of the micropattern of the
the formation of vessel-like cell assembly.40 Ingber and adhesive islands caused alteration in cell shape, which gov-
Folkman studied the effects of coating density of fibronectin, erned whether individual cells grow or die. The effect of the
type IV collagen or gelatin on culture dishes (adhesiveness) extracellular adhesiveness on cell growth and viability was
on the behavior of FGF-stimulated endothelial cells.41 They independent of the type of matrix protein used to mediate
observed extensive cell spreading and growth in dishes coated adhesion in the islands (Fig. 27.3).
with a high density of fibronectin or type IV collagen, while
cell rounding, detachment and loss of viability occurred in Modified Growth Factors: TGF-!
dishes with low density coating. Intermediate coating den- with a Collagen Binding Domain
sity promoted cell extension and partial retraction of multi- TGF-! appears to be an important regulator of the
cellular aggregates; cessation of growth and formation of cardiovascular system. The TGF- ! proteins are highly
expressed during embryonic development of the heart, and control the biological activity of this growth factor during
can be elevated after hemodynamic stress. Hence, they pos- its use as regulatory signal for tissue engineering.
sess the potential to significantly influence the progression A collagen targeted TGF-! recombinant fusion pro-
of a number of vascular disorders, in particular those in- tein has been designed, expressed, and renatured into active
volving vessel remodeling and repair, such as hypertension forms.45,49 In these fusion proteins, a collagen targeting
following percutaneous balloon catheter angioplasty, ath- decapeptide derived from von willebrand factor50 was modi-
erosclerosis and under some circumstances, angiogenesis. fied49 and fused at the N-terminus of the active TGF-! frag-
These and many other effects of TGF-! proteins in cardio- ment by molecular manipulation. A protease sensitive site
vascular development, physiology and pathophysiology are was also inserted in the sequence between the collagen bind-
dependent upon processes that regulate their expression and ing domain and the active TGF-! fragment, in order to re-
release from latent complexes, the receptor types available lease the growth factor at appropriate times and sites. Bac-
for binding and the interactions between receptor-mediated terial expression systems provided a good source for this
intracellular signaling events, which ultimately lead to tis- recombinant protein and appear to guarantee sufficient
sue responses.44 amounts for tissue engineering applications. BL21 (DE3)
Major problems that hamper the use of growth fac- E. coli strain was transformed with a pET 28b expression
tors in tissue engineering are: vector which carried recombinant DNA. Under IPTG in-
1. The activity is quickly lost due to the protease duction, this fusion protein can be induced to express at high
digestion; yield. The induced fusion proteins were found in insoluble
2. They are not retained at the application site; inclusion bodies. After being solubilized by a denaturant and
3. They are carried away by the circulation. purified by Ni-chelating column, the purified TGF-! mono-
Therefore, targeting growth factors to the application mer was refolded in an in vitro optimized system, in which
site and controlling their release and activity becomes cru- reduced and oxidized forms of glutathione assist TGF-! in-
cial when applying growth factors as regulatory signals for ter- and intradisulfide bond exchange.45
tissue engineering. In addition to approaches that rely on
adsorption of ionically bound, physically entrapped or co- Binding Characteristics
valently bound growth factors to resorbable matrices, a novel Active TGF-!, a highly hydrophobic basic protein, is
approach developed in our laboratory relies on the construc- rapidly lost from the medium or extracellular fluid by pro-
tion of fusion proteins which include a matrix or cell bind- tease digestion. Many extracellular matrix components bind
ing domain for targeted delivery of such growth factors45 to TGF-! in a reversible manner and may serve as a reser-
(Fig. 27.4). voir for the growth factors.51,52 TGF-! binding proteins could
Transformtin growth factor-! (TGF-!) was used as an sequester TGF-! to the extracellular matrix and act as a buffer
example for studying a ECM targeted growth factor. TGF-! for its activities.53,54 Since collagen is a major component of
is a secreted multifunctional protein that regulates many the extracellular matrix, and the structural protein of most
aspects of cellular functions, including cell proliferation, tissues, it was chosen as functional carrier of soluble growth
differentiation, and extracellular matrix metabolism.46 Based factors in this study. In the fusion of TGF-! proteins, a col-
on the different physiological effects of TGF-!, a variety of lagen binding domain derived from von willebrand factor
potential clinical applications for this growth factor have was incorporated at the N-terminal of the active growth fac-
been suggested.47,48 Since TGF-! is a pleotropic agent which tor fragment by a bridging Gly. Takagi et al50 have reported
can stimulate, inhibit, and modulate cellular events in a time that the primary structure of the von willebrand factor con-
and concentration dependent manner, it is important to tains a high-affinity collagen binding domain. They further
Fig. 27.4. A schematic representation of the genetically engineered growth factor(s). The constructs contain: a collagen
binding sequence derived from von willebrand factor; a purification tag (e.g., (His)6 or GST); and a protease-sensitive
site for control release.
Polypeptide Growth Factors with a Collagen Binding Domain: Their Potential for Tissue Repair and Organ Regeneration 293
delimited the collagen binding site to a decapeptide with collagen binding domain in F2 afforded this high affinity
the amino acid sequence WREPSFCALS. The collagen tar- interaction with collagen.
geting characteristics were assayed as described below.
Collagen targeted TGF-! fusion proteins from bacte- Biological Activity
rial inclusion bodies were isolated, dehydrated in DMF, and
labeled by [3H]NaBH4 (100 mCi, Amersham) DMF solu- Released Collagen Targeted TGF-!
tion.55 Labeled recombinant proteins were purified and rena- Retains Biological Activity
tured as described and the bioactivity was also checked. Dif- TGF-! activity was assessed by transfected mink lung
ferent doses of labeled TGF-!1-F1 or -F2 fusion proteins were cell with PAI-1 promoter/luciferase reporter genes.45 The
then added to the collagen precoated wells. After incuba- specificity and sensitivity are the result of using a truncated
tion and extensive washes in PBS, the labeled TGF-! remain- PAI-1 promoter which retains the two regions responsible
ing bound to the collagen was counted after collagenase for maximal response to TGF-! (56). Collagen bound TGF-
digestion. The results are shown in Figure 27.5. TGF-!1-F2, !1-F1/F2 fusion proteins were released from the collagen gels
which has a collagen binding domain, binds to collagen in a with collagenase. The released solubilized TGF-!1-F1 and
dose dependent manner. TGF- ! 1-F1, which lacks the TGF-!1-F2 proteins were assayed for their biological activ-
collagen binding domain, binds to collagen with less ity. It was found that collagen bound TGF-!1-F2 was still
affinity through nonspecific interactions. The only dif- active when released with collagenase. These experiments
ference in F1 and F2 constructs is the auxiliary collagen demonstrate that the collagen binding domain enables TGF-
binding decapeptide; this suggests that with a collagen !1-F2 to be sequestered by the collagen matrix and that it
binding domain, TGF-! can be specifically targeted to can be released by collagenase digestion to display its bio-
collagen matrices. logical activity.
To determine the affinity of such binding, F2 fusion
protein was first immobilized on a Ni-NTA column and then Collagen Bound TGF-!1 Activity
exposed to biosynthetically labeled [3H]collagen, which was Even though we had proven that collagen-targeted
subsequently loaded onto the column. Under these condi- TGF-! can bind selectively to the collagen matrix, the bio-
tions a large portion of the radioactivity was found to bind logical activity of bound TGF-!2-F2 was unclear. When we
to the column. Washing the column with a linear gradient precoated culture wells with type I collagen, and then added
of NaCl from 0.15 to 2.0 did not release the [3H]collagen. TGF-!2-F1 and TGF-!2-F2 on the collagen gels, then washed
However, application of a urea gradient (0-5.0 M) was able extensively with PBS, only bound TGF-!2-F1/F2 remained
to quantitatively elute all bound radioactivity (Fig. 27.6). By on the gel. Subsequently, osteoblastic osteosarcoma cells were
contrast, when the F1 fusion protein containing only a (His)6 plated on top of the collagen gel. After 72 hrs, alkaline phos-
tag and the TGF-!1 active fragment was applied to the Ni- phatase activity was tested in the cell layer. Results (Fig. 27.7)
NTA column under the same conditions, most [3H]collagen showed that when TGF-!2-F1/F2 was added to the medium,
was eluted in the void volume, suggesting that the auxiliary the cellular responses to these two factors were similar; for
the collagen-bound TGF-!2, cells could still ‘recognize’ sistent with their playing a role during the rapid changes
bound TGF-!2 and display increased alkaline phosphatase seen in cardiovascular tissues in during development and
activity. On the other hand, TGF-!2-F1, washed away by PBS induced anomalies.
from the collagen gel because it lacks a collagen binding
domain, exhibited no effect on induction of alkaline phos- bFGF
phatase activity above control. These results not only con- bFGF is present in many tissues, intracellularly or
firmed that TGF-!2-F2 could specifically target collagen, but bound to heparin-like molecules of the extracellular ma-
also demonstrated that extracellular matrix-associated trix. It is thought to exert its physiological functions in tis-
TGF-! still performed its biological function. sue repair and neovascularizatoion after being released from
the extracellular matrix.58,59 bFGF causes mesenchyme for-
Other Growth Factors Which May Individually mation and stimulates skin wound healing by increasing cell
or Synergistically Contribute to Organogenesis recruitment, mitosis, and extracellular protein production.
Growth factor targeting to responsive cells and main- Though very potent, bFGF is rapidly degraded when injected
tenance of adequate pharmacological levels becomes essen- or ingested and applications of bFGF are hampered by the
tial, particularly in view of their different effects on various instability of the compound. Additionally, a bFGF dose-re-
cells and the dose dependence of their response. Using the sponse curve on bone growth study exhibits biphasic effects,
same recombinant strategy, other growth factors besides reflected by an exaggerated inhibited cell ingrowth.60 Con-
TGF-! have also been engineered with an ECM targeting trolling bFGF activity therefore becomes meaningful for
domain. These growth factors include basic fibroblast growth proper use and clinical application of this growth factor.
factor (bFGF), epidermal growth factor (EGF), vascular en- Preservation and stabilization of bFGF was previously ac-
dothelial growth factor (VEGF), #-ECGF and bone morpho- complished by binding the factor to heparin-Sepharose
genic protein-3 (BMP-3). The targeting sites of growth fac- beads. This permitted its prolonged storage, repeated hand-
tors are not limited to the collagen matrix, since fibronectin ing and encapsulation within a microspherical controlled-
and heparin targeting domains can also be incorporated into released device.61 Recently our laboratory has engineered
the growth factor molecules. The tissue distribution and collagen targeting bFGF recombinant fusion proteins to
cellular effects of platelet-derived growth factor (PDGF), ba- enable this fusion protein to be targeted to collagen matri-
sic fibroblast growth factor (bFGF), transformtin growth ces. Its in vitro activities have been evaluated and their ap-
factor-b1 (TGF-!1) and insulin-like growth factor 1 (IGF-1) plication is under investigation.
suggest a potential role for these factors in cardiovascular
matrix regeneration. Fibroblast replication is stimulated by VEGF
PDGF and by bFGF. IGF-1 and TGF-!1 have no effect on Recent studies show that exogenously administered
fibroblast replication. Collagen production is stimulated by angiogenic factors can induce the formation of new blood
all of the growth factors tested: in order of potency, TGF-!1. vessels in adult animals and enhance collateral blood flow
PDGF, IGF, bFGF. None of the growth factors affected the to ischemic tissues. It has been suggested that this approach
proportion of rapidly degraded newly synthesized collagen.57 may have a therapeutic potential.62 VEGF (vascular endo-
The sensitivity of cardiac fibroblasts to these factors is con- thelial growth factor) is a specific mitogen for endothelial
Polypeptide Growth Factors with a Collagen Binding Domain: Their Potential for Tissue Repair and Organ Regeneration 295
cells in general and for vascular endothelial cells in particu- several BMP family members which belongs to the
lar. It has been shown to promote angiogensis, accelerate transformtin growth factor-beta superfamily. BMP-3 has
endothelial repaving, and to attenuate intimal hyperplasia been characterized by its ability to stimulate osteoblastic dif-
after arterial injury. The active forms of VEGF are disulfide- ferentiation and bone formation.64,65 It was first identified
linked, dimeric glycoproteins. The presence of two tyrosine and then purified from bovine bone by Reddi and col-
kinase receptors for VEGF have been identified in vascular leagues.65 Although in vivo tests support the role of BMP-3
endothelial cells. in bone formation, its mechanism of action is poorly un-
derstood. In our laboratory, we bioengineered human re-
EGF and ECGF combinant BMP-3 fusion proteins from E. coli and
Epidermal growth factor (EGF), a 53 amino acid mi- renaturated them into their native dimer conformation. The
togenic polypeptide present in many mammalian species, is large quantities of BMP-3 obtained enabled us to do exten-
one of a number of growth factors being investigated for sive studies on this growth factor in animal models. When
their potential to expedite the healing process. EGF has been combined with collagen sponge matrices or porous hy-
shown to stimulate keratinocyte division in vitro and epi- droxyapatite ceramics, it appears to generate a potential bone
dermal regeneration in vivo. It had been shown to have an substitute for fracture repair and regeneration.66
effect on mesenchymal cells by producing marked prolif-
eration of the dermis in partial thickness wounds and by Applications of the Growth Factor Matrix Technology
increasing the tensile strength of surgical incisions. In ex- to the Design of Bioprostheses
perimental animals a modified fibrin glue containing 1 ∝g One of the significant advances in medicine in this
of #-endothelial cell growth factor (ECGF) was implanted century is associated with the ability of the surgeon to re-
between the aorta and the myocardium of the left ventricle.63 place defective heart valves, veins and arteries with
After 9 weeks of implantation, angiography and histologic bioprosthetic devices. Nevertheless, these implants have as-
investigation showed a newly grown vascular structure be- sociated disadvantages since they may require anticoagula-
tween the aorta and the myocardium in all experimental tion, periodic monitoring for possible failure and in some
animals, but none in the controls. These studies proved the cases have a limited durability or only be usable in selected
feasibility of initiating site-directed formation of new blood cases or locations. It is obvious that if we can provide a scaf-
vessel structures to the heart by a modified fibrin glue im- folding which after being implanted would allow a tissue or
plant containing angiogenic growth factor #-ECGF. organ to regenerate, that we would be achieving a very meri-
torious objective. Our current understanding of cell biol-
BMP-3 ogy, that is, how cells differentiate from stem cells into dif-
One of the few tissues that can regenerate in mam- ferentiated cells, how cells interact with the extracellular
mals is bone. To a great extent this is due to the activity of matrix and the consequences of controlled cell-cell interac-
specific growth factors which stimulate its formation. Bone tions, should be able to lead to the ordered repair of tissues
morphogenetic protein-3 (BMP-3, osteogenin) is one of the and even organ regeneration in the near future.
296 Tissue Engineering of Prosthetic Vascular Grafts
Collagen is an ideal biomatrix, as it is the major com- lar interactions involving their cell membranes, as a result
ponent of the extracellular matrix. It can be rendered rela- of the movement of molecules such as peptides or steroids,
tively nonantigenic, even if obtained from another species, that can act locally or systematically to modulate cell func-
after removal of the nonhelical antigenic determinants and tions. Growth factors are included in the latter group.1
may soon be available in a recombinant form from human Growth factors can either stimulate or inhibit cell di-
DNA. Reconstituting soluble collagen into fibrous matrices, vision, differentiation, migration or gene expression, de-
alone or in combination with the other major component pending on the cells involved. Depending on the concentra-
of the extracellular space, namely the proteoglycans, is a rela- tion present in the cellular environment, growth factors can
tively simple procedure. Specific collagen types, including act in an opposing manner and up or downregulate the syn-
those which give rise to basement membranes, are also be- thesis of receptors. In general, both growth factors and
coming available in the design of biomatrices. mRNAs that code for them turn over very rapidly. They usu-
Growth factors are part of a large number of polypep- ally exist as inactive or partially active precursors that require
tides that transmit signals affecting cellular activities. Cells proteolytic activation, and may need to bind to matrix mol-
may communicate with each other through direct molecu- ecules for activity or stabilization. They may perform differ-
Fig. 27.9. Schematic diagram of vascular graft substitute. A TGF-! impregnated crosslinked collagen sheet supports the growth of
fibroblast. VEGF impregnated modified collagen gel supports the growth of vascular endothelial cell growth.
ent functions on different cell types: For instance, TGF-! is ible with the surrounding tissue and should be able to anas-
stimulatory for fibroblasts and inhibitory for keratinocytes. tomose readily to the existing viable tissue.
With the increasing availability of growth factors de- Our increased understanding of the extracellular ma-
rived from cultured human cells and their expansion through trix, and of the role of agents such as cytokines and growth
recombinant technologies, coupled with an increasing un- factors in the various aspects of cellular activity associated
derstanding of their functions and clinical applications, the with tissue repair and remodeling, can now provide us with
need for useful pharmaceutical forms is becoming more and sufficient insight into the design of tissue compatible scaf-
more apparent. Growth factor targeting to responsive cells foldings. These scaffoldings can be populated with cells and
and maintenance of adequate pharmacological levels be- via processes of cell-matrix and cell-cell interactions
comes essential, particularly in view of their different effects modulated to acquire shapes and functions compatible with
on various cells and the dose dependence of their response. their endothelial and mesenchymal cells. There should have
An important issue is the short biological half life of a decreased tendency to attract platelets and be able to retain
growth factors. For example, platelet-derived growth factor growth modulating polypeptides which encourage
(PDGF), an important growth factor first isolated from plate- systematic repopulation by selected cells and yield functional
lets, cannot be detected in the circulation, and when injected vascular prostheses. We hope that the data present as well as
intravenously its half life is less than 2 min.67 the suggestions made during the course of this review
Our ability to target growth factors to the extracellu- continue to assist and encourage others to move in
lar matrix components and to couple their release to native this direction.
ability or inducible enzymatic activity, and our understand-
ing of stem cell biology, as well as our ability to cause such References
cells to differentiate under the influence of specific growth 1. Nimni ME. Polypeptide growth factors: Targeted delivery
factors, should allow us to generate functional tissues as re- systems. Biomaterials 1997; 18:1201-1225.
placement for damaged ones. In Figures 27.8 and 27.9 we 2. Huang-Lee LLH, Nimni ME. Preparation of type I col-
have provided models for the design of such a matrix, with lagen fibrillar matrices and the effects of collagen con-
centration on fibroblast contraction. Biomed Eng Appl
an emphasis on the construction of a vascular graft. Further
Basis Comm 1993; 5:664-675.
experimental work along these lines is very likely to gener- 3. Huang-Lee LLH, Nimni ME. Fibroblast contraction of
ate functional tissues which, starting with vascular grafts, collagen matrices with and without covalently bound
will continue to increase in complexity until we eventually hyaluronan. J Biomater Sci Polymer Edn 1993; 5:99-109.
are able to generate more complex functional organs. 4. Huang-Lee LLH, Nimni ME. Crosslinked CNBr-activated
hyaluronan-collagen matrices: Effects on fibroblast con-
Summary and Conclusion traction. Matrix Biology, 1994; 14:147-157.
The design of artificial vascular prostheses and recon- 5. Huang-Lee LLH, Wu JH, Nimni ME. Effects of
struction with collagen scaffoldings of damaged vessels rep- hyaluronan on collagen fibrillar matrix contraction by fi-
resents a very important field of endeavor. Vascular grafts broblasts. J Biomed Mater Res, 1994; 28:123-132.
should be soft, pliable, flexible, puncturable, and compat-
298 Tissue Engineering of Prosthetic Vascular Grafts
6. Huang LLH, Chung CH. Superior wound healing of 29. Smith CW Endothelial adhesion molecules and their role
hyaluronan-collagen fibrillar matrix. In Preparation. 1997. in inflammation. Can J Physiol Pharmacol 1993; 71:76-86.
7. Yannas IV, Burke JF. Design of an artificial skin. I. Basic 30. Bevilacqua MP, Nelson RM, Mannori G, Cecconi O. En-
design principles. J Biomed Mater Res 1980; 14:65-81. dothelial-leukocyte adhesion molecules in human disease.
8. Yannas IV, Burke JF, Gordon PL, Huang C, Rubenstein Ann Rev Med 1994; 45:361-378.
RH. Design of an artificial skin. II. Control of chemical 31. Gimbrone MA Jr, Cotran RS, Folkman J. Human vascu-
composition. J Biomed Mater Res 1980; 14:107-131. lar endothelial cells in culture. J Cell Biol 1974;
9. Yannas IV. What criteria should be used for designing 60:675-684.
artificial skin replacements and how do the current graft- 32. Moyer CF, Dennis PA, Majno G, Joris I. Vascular endot-
ing materials meet these criteria? J.Trauma 1984; helium in vitro, isolation and characterization. In vitro
24(9):s29-s39. Cell Dev Biol 1988; 24:359-368.
10. Yannas IV. Regeneration of skin and nerve by use of col- 33. Bauer J, Margolis M, Schreiner C, Edgell CJ, Azizkhan J,
lagen templates. In: Nimni ME, ed. Collagen Biotechnol- Lazarowski E, Juliano RL. In vivo model of angiogenesis
ogy, Vol. 3. CRC Press, Inc. 1988:87-115. using a human endothelium-derived permanent cell line:
11. Hynes RO. Fibronectins. Scientific American. 1986; Contributions of induced gene expression, G-protein, and
256(6):42-51. integrins. J Cell Physiol 1992; 153:427-449.
12. Kleinman HK, Klebe RJ, Martin GR. Role of collagenous 34. Grant DS, Tashiro KI, Segui-Real B, Yamada Y, Martin
matrices in the adhesion and growth of cells. J Cell Biol HK. Intracellular mechanisms involved in basement mem-
1981; 88:473-485. brane induced blood vessel differentiation in vitro. In
13. Grinnell F. Cell-collagen interactions: Overview. Meth Vitro cell Dev Biol 1991; 27A:327-336.
Enzym 1982; 82:499-503. 35. Kubota Y, Kleinmam HK, Martin GR, Lawley TJ. Role of
14. Bretscher MS. How animal cells move. 1987; 257(6):72-90. laminin and basement membrane in the morphological
15. Yaffe A, Shoshan S. Cessation of epithelial cell movement differentiation of human endothelial cells into capillary-
at native type I collagen-epithelial interface in vitro. Col- like structures. J Cell Biol 1988; 107:1589-1598.
lagen Res Rel 1985; 5:533-540. 36. Schiffrin EL. The endothelium and control of blood ves-
16. Donaldson DJ, Smith GN, Kang AH. Epidermal cell mi- sel function in health and disease. Clin Invest Med 1994;
gration on collagen and collagen-derived peptides. J Cell 17:602-620.
Sci 1982; 57:15-23. 37. Yao J, Bone RC, Sawnhey RS. Different effects of tumor
17. Brunt JV, Klausner A. Growth factors speed wound heal- necrosis factor-alpha on the expression of fibronectin and
ing. Biotechnology 1988; 6(1): 25-30. collagen genes in cultured bovine endothelial cells. Cell
18. Barrandon Y, Green H. Cell migration is essential for sus- Mol Biol Res 1995; 41:17-28.
tained growth of keratinocyte colonies: The roles of 38. Fournier N, Doillon CJ. In vitro angiogenesis in fibrin
transformtin growth factor-b and epidermal growth fac- matrices containing fibronectin or hyaluronic acid. Cell
tor. Cell 1987; 50:1131-1137. Biol Int Rep 1992; 16:1251-1263.
19. Berridge MJ. The molecular basis of communication 39. Schnaper HW, Kleinman HK, Grant DS. Role of laminin
within the cell. Scientific American 1985; 253(4):142-152. in endothelial cell recognition and differentiation. Kid-
20. Doillon CJ, Silver FH. Collagen wound dressing: Effect of ney Int. 1993; 43:20-25.
hyaluronic acid and fibronectin. Biomaterials 1986; 7:3-8. 40. Passaniti A, Taylor RM, Pili R, Guo Y, Long PV, Haney
21. Doillon CJ, Silver FH. Fibroblast growth on a porous col- JA, Pauly RR, Grant DS, Martin GR. Methods in labora-
lagen spone containing hyaluronic acid and fibronectin. tory investigation. A simple, quantitative method for as-
Biomaterials 1987; 8:195-200. sessing angiogenesis and antiangiogenic agents using re-
22. Denefle JP, Lechaire JP, Miskulin M. Frog skin epithe- constituted basement membrane, heparin, and fibroblast
lium interactions with a bovine purified type I collagen growth factor. Lab Invest 1992; 67:519-528.
gel in culture. J Submicrosc Cytol 1985; 17(1):41-47. 41. Ingber DE, Folkman J. Mechanochemical switching be-
23. Denefle JP, Lechaire JP, Zhu QL. Cultured epidermis in- tween Growth and differentiation during fibroblast growth
fluences the fibril organization of purified type I collagen factor-stimulated angiogenesis in vitro: Role of extracel-
gels. Tissue & Cell 1987; 19(4):469-478. lular matrix. J Cell Biol 1989; 109:317-330.
24. Grinnell F, Lamke CR. Reorganization of hydrated col- 42. Jullano RL, Haskill S. Signal transduction from the extra-
lagen lattices by human skin fibroblasts. J Cell Sci 1984; cellular matrix. J Cell Biol 1993; 120:577-585.
66:51-63. 43. Chen CC, Mrksich M, Huang S, Whitesides GM, Ingber
25. Allen TD, Schor SL. The contraction of collagen matrices DE. Geometric control of cell life and death. Science 1997;
by dermal fibroblasts. J Ultrastructure Res 1983; 276:1425-1428.
83:205-219. 44. Saltis J, Agrotis A, Bobik A. Regulation and interactions
26. Bell E, Ivarsson B, Merrill C. Production of a tissue-like of transformtin growth factor-! with cardiovascular cells:
structure by contraction of collagen lattices by human fi- Implications for development and disease. Clin Exp Pharm
broblasts of different proliferative potential in vitro. Proc Phys 1996; 23:193-200.
Natl Acad Sci USA 1979; 76(3):1274-1278. 45. Han B, Hall FL, Nimni ME. Purification of a recombi-
27. Lee PC, Chen LW, Huang LLH. Comparison of nant collagen-targeted TGF-!2 fusion protein expressed
carbodiimide and epoxy reactions to graft collagen on in E. coli. Protein Expression and Purification. 1997;
polyurethane. Biomed Eng Appl Basis Comm 1996; 11:169-1778.
8:22-29. 46. Massague J. The transformtin growth factor-beta family.
28. Lee PC, Huang LL, Chjen LW, Hsieh KH, Tsai CL. Effect Annu Rev Cell Biol 1997; 6:597-641.
of forms of collagen linked to polyurethane on endothe- 47. Miyazono K, Heldin CH. Latent forms of TGF-!: Mo-
lial cell growth. J Biomed Mater Res 1996; 32:645-653. lecular structure and mechanisms of activation. In: Clini-
Polypeptide Growth Factors with a Collagen Binding Domain: Their Potential for Tissue Repair and Organ Regeneration 299
cal Applications of TGF-!. CIBA Foundation Symposium to diverse classes of growth factors. Eurp J Cell Biol 1995;
157. Chichester: John Wiley and Sons, 1991:81-90. 68:330-335.
48. Roberts AB, Sporn MB. Physiological actions and clinical 58. Baird A, Klagsbrun M. Annals of the New York Academy
applications of transformtin growth factor-! (TGF-!). of Science: the fibroblast growth factor family, Volume
Growth Factor 1993; 8:1-9. 638. New York Academy of Science. 1991.
49. Tuan T, Cheung DT, Wu L, Yee A, Gabriel S, Han B, 59. Voldavsky I, Fuks Z, Ishai-Michaeli R, Bashkin P, Korner
Nimni EM, Hall F. Engineering, expression and renatur- G, Bar-Shavit R, Klagsbrun M. Extracellular matrix-resi-
ation of targeted TGF-! fusion protein. Connect Tissue dent basic fibroblast growth factor: Implication for the
Res 1996; 34:1-9. control of angiogenesis. J. Cell Biochem 1991; 45:167-76.
50. Takagi J, Asai H, Satio Y. A collagen/gelatin binding 60. Wang JS, Aspenberg P. Basic fibroblast growth factor in-
decapeptide derived from bovine peopolypeptide of von creases allograft incorporation. Acta Orthop Scand 1994;
willebrand factor. Biochemistry 1992; 31:8530-4. 65:27-31.
51. Mooradian DL, Lucas RC, Weatherbee JA, Furcht LT. 61. Edelman ER Mathiowitz E, Langer R, Klagsbrun. Con-
Transformtin growth factor-beta 1 binds to immobilized trolled and modulated release of basic fibroblast growth
fibronectin. J Cell Biochem 1989; 41:189-200. factor. Biomaterial 1991; 12:619-626.
52. Butzow R, Fukushima D, Twardzik DR, Ruoslahti E. A 62. Safi J Jr, Gloe TR, Riccioni TR, Riccioni T, Kovesdi I,
60 kD protein mediates the binding of transformtin Capogrossi MC. Gene therapy with angiogenic factors: A
growth factor-! to cell surface and extracellular matrix new potential approach to the treatment of ischemic dis-
proteoglycans. J Cell Biol 1993; 256:5575-727. eases. J Mol Cell Cardiol 1997; 29:2311-2325.
53. Omitted in proof. 64. Sampath TK, Reddi AH. Dissociative extraction and re-
54. Massague J. Betaglycan can act as a dual modulator of constitution of extracellular matrix components involved
TGF-! access to signaling receptor: Mapping of ligand in local bone differentiation. Proc Natl Acad Sci USA
bonding and GAG attachment sites. J Cell Biol 1994; 1981; 78:7599-603.
124:557-68. 65. Luyten FP, Cunnungham NS, Ma S, Muthukumaran N,
55. Cheung DT, Benya PD, Gorn A, Nimni ME. An efficient Hammonds RG, Nevin WB, Wood WI, Reddi AH. Puri-
method for in vitro labeling proteins to high specific ra- fication and partial amino acid sequence of osteogenin, a
dioactivity using [3H]-NaBH4 in dimethylformamide. Anal protein initiating bone differentiation. J Biol Chem 1989;
Biochem 1981; 116:69-74. 264:13377-13380.
56. Abe M, Harpel JG, Metz CN, Nunes I, Loskutoff DJ, 66. Han B, Perelman N, Hall F, Shors E, Nimni M. Bone in-
Rifkin DB. An assay for transformtin growth factor-! us- duction by a recombinant collagen targeted BMP-3 fu-
ing cells transfected with a plasminogen activator inhibi- sion protein composite, (in prepration).
tor-1 promoter-luciferase construct. Analy Biol 1994; 67. Bowen-Pope DF, Malpass TW, Foster DM, Rodd R. Plate-
216:276-284. let-derived growth fctor in vivo: Levels, activity, and rate
57. Butt RP, Laurent GJ, Bishop JE. Collagen production and of clearance. Blood 1984; 64:458-469.
replication by cardiacfibroblasts is enhanced in response
Facilitation of Healing
Angiogenesis
CHAPTER 28
Fibroblast Growth Factors in Angiogenesis
and Tissue Engineering
Karin A. Blumofe, Timothy J. Heilizer, Paula K. Shireman, Howard P. Greisler
Introduction
T he main treatment for arterial occlusive disease has become reconstruction. This involves
utilizing a graft, either in the form of a vein or synthetic material. These treatments have
been found to be limited in their long term therapeutic value due to stenosis formation, a
process in which there is luminal narrowing of the vessel due to smooth muscle cell
proliferation and connective tissue deposition in the intima of the vessel wall or at the vessel-
synthetic graft interface. The smooth muscle cells can continue to proliferate for up to one
year after the initial injury. This response is in part modulated by a variety of growth factors
such as fibroblast growth factor (FGF), platelet derived growth factor, angiotensin ii, insulin-
like growth factor I, and so forth. This course of events results in intimal hyperplasia,
ultimately leading to a decrease in blood flow, thrombosis and failure of the
interventional reconstruction.
A major focus of vascular research has been to investigate ways in which to decrease
the smooth muscle cell migration and proliferation, ultimately decreasing the intimal hy-
perplasia response. Fibroblast growth factor, as mentioned above, is one of the growth fac-
tors that stimulates smooth muscle cell migration and proliferation. Additionally, it serves
as a potent stimulator of endothelial cells, inducing angiogenesis, the formation of new blood
vessels. The process of neovascularization has also been studied extensively as a therapeutic
treatment for ischemic disease. With this in mind, the goal of this chapter is to discuss prop-
erties of FGF and its role in tissue engineering.
History
In the mid-1900s the brain was found to be a rich source of growth factors that stimu-
lated fibroblast proliferation. However, it was not until the 1970s that partially purified pitu-
itary extracts were shown to contain potent mitogens, particularly for 3T3 fibroblasts.1 In
1974 a mitogenic polypeptide from the pituitary was identified2-4 which was named fibro-
blast growth factor. Attempts to further characterize the growth factor were hampered by
high levels of contaminating fragments of myelin basic protein.5 Thomas in 1980 used iso-
electric points to isolate acidic FGF.6 The acidic form did not interact with antibodies against
myelin basic protein. It was also found that the acidic form possessed mitogenic activity for
endothelial cells.7 Bovine brain contained another type of FGF which had different proper-
ties from the acidic FGF, as it was a basic mitogen. Thus, FGF was separated into two forms:
FGF-1 and FGF-2. The structural characterization of acidic FGF 8 and basic FGF 9
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
302 Tissue Engineering of Prosthetic Vascular Grafts
demonstrated that these growth factors are related polypep- wards the perivascular space, forming parallel processes.
tides and established the basis for a larger family of polypep- With linear movement, a single endothelial cell projection
tide growth factors. Over the next 13 years eight more or pseudopod migrates from the parent capillary to the sur-
isoforms of FGF would be identified, most recently FGF-9 rounding connective tissue.11,17
in 1993.10 The next phase in this process is that of endothelial
cell proliferation. Mature endothelial cells normally have a
Angiogenesis slow turnover rate of approximately 2 months. However,
Angiogenesis is the formation of new blood vessels when necessary, as in response to angiogenic stimuli, the
from the microcirculation, by a process of cellular outgrowth. endothelial cell can quickly convert to a proliferative state.
In contrast, the development of new vessels from stem cells, The induction of endothelial cell proliferation is associated
as found in an embryo, is termed vasculogenesis. Angiogen- with disruption of cell-cell contacts18 and alterations in the
esis plays a major role in a variety of natural and disease cytoskeletal organization, such as microtubule destabiliza-
processes significant in the adult life, such as reproduction tion.19 Development of the new vessel lumen involves the
(placenta, corpus luteum formation, etc.), wound healing, endothelial cell forming tubular channels. It is thought that
bone repair, inflammation, neoplasia and collateralization the capillary lumen is generated by adjacent endothelial pro-
in response to ischemia (heart disease and peripheral vas- cesses, causing intercellular canalization of adjacent endo-
cular disease). thelial cells.20 A new basal lamina is generated consisting of
The phenomenon of angiogenesis begins with vascu- fibronectin, laminin and type IV collagen.13
lar sprouts that originate from the walls of preexisting cap- Another important cell in the angiogenic process is
illaries and small venules.11 It is possible that endothelial the pericyte. Initially the pericytes undergo intense prolif-
cells are recruited from venular segments, because these cells eration. During the stages of capillary sprouting, the exact
are less constrained by a well formed basal lamina. More role of the pericytes is unclear. These cells appear to bridge
recently, larger vessels have been shown to play a role in the the gaps between opposing endothelial sprouts by utilizing
formation of new blood vessels. For example, it has been the pericytic processes. Capillary sprouts are then able to
demonstrated that the rat femoral vein, with a discontinu- fuse, forming capillary loops and eventually a plexus net-
ous layer of internal elastic lamina and smooth muscle cells, work. The pericytes also play a role in the regulation of
can contribute to angiogenesis. When the vein is treated with angiogenesis. There is an absence of pericytes at the tips
prostaglandins E1 and E2, vascular sprouts arise from the of migrating endothelial cells while, in contrast, their
endothelial cells in the intima layer of the vessel.12 Angio- presence in the older regions of capillaries may inhibit
genesis does not seem to originate from the arterial side of endothelial cell proliferation and migration, leading to
the circulation.13 vessel maturation.21
Scanning electron microscopy has been used to study Many cells, including pericytes, macrophages, mast
the early changes in vasculature and sprouts appear rapidly, cells, lymphocytes, connective tissue cells, endothelial cells
as early as 27 hours after exposure to angiogenic stimuli.14 and tumor cells influence the formation of new vessels by
From three to five days these sprouts continue to flourish, secreting soluble angiogenic molecules. The balance of posi-
ultimately forming a rich anastomosing plexus. tive and negative regulators of angiogenesis control when a
On a cellular level, there are multiple processes in- vessel will proceed to neovascularization. A multitude of
volved in angiogenesis. Early on there is an increase in the molecules have been characterized as influencing this pro-
vascular permeability. There is an association of inflamma- cess. Initial studies were done with the chorioallantoic mem-
tory cells with intravascular accumulation of platelets and brane of the chick embryo (CAM) and the corneal pocket
polymorphonuclear leukocytes. The new vessels arise fol- of the rabbit. In the CAM system, a quantity of an angio-
lowing the margination and diapedesis of the leukocytes.13 genic substance was placed on exposed chick chorioallan-
Then the endothelial cells become activated. They display a toic membrane. Similarly with the rabbit experiments, an-
number of morphological changes, including hypertrophy giogenic substances were introduced into intracorneal pock-
with bulging into the vascular lumen, nuclear enlargement, ets. These two models were useful in examining the angio-
increase in the number of organelles and formation of pro- genic potential of a substance. However, it was not until the
jections from their surfaces.13 The fragmentation and disin- development of cultured endothelial cells and biological as-
tegration of the basal lamina is a necessary step for the mi- says that it became possible to directly examine the mecha-
gration of endothelial cells to occur.15 This degradation is nisms which regulate cell behavior in angiogenesis. The first
due to proteolytic enzymes synthesized and secreted by the purified angiogenic factor was FGF-2. This was followed by
activated endothelial cells.16 numerous other molecules such as vascular endothelial
Angiogenesis then proceeds with the migration of growth factor (VEGF), transformtin growth factor-beta,
endothelial cells, moving into the interstitial space. Two tumor necrosis factor, interleukin-8, platelet derived endot-
different types of endothelial cell migration have helial cell growth factor (PD-ECGF), angiogenin and
been described: angiotropin, to name a few.
1. Bicellular or telescoping formation; and FGF serves as a chemotactic stimulator for endothe-
2. Linear formation. lial cells. If a monolayer of cultured endothelial cells is dam-
In the bicellular type of migration, two or more en- aged, FGF stimulates cellular migration into the denuded
dothelial cells move from the wall of the parent vessel to- area.22 FGF-2 has been shown to increase the secretion of
Fibroblast Growth Factors in Angiogenesis and Tissue Engineering 303
plasminogen activator and collagenase from bovine aortic to the FGF family, with 30-40% homology. FGF-9, the most
endothelial cells. These substances are thought to digest the recently discovered, was purified from a human glial cell
basement membrane preceding migration of endothelial line.10
cells. Antibodies directed against FGF-2 cause impairment Of these nine FGF genes that have been identified,
of endothelial cell migration and inhibit the release of plas- different isoforms can be created through a variety of meth-
minogen activator.22 Additional experiments have demon- ods. For instance, varying isoforms are generated by using
strated that FGF-2 treatments can induce capillary endot- alternative initiation codons for translation.32,33 Novel FGF
helial cells to invade a collagen matrix. These cells then or- isoforms can also be generated by alternative splicing such
ganize themselves to form characteristic tubules that re- as with FGF-234 and FGF-8.30 Finally, posttranslational
semble blood capillaries.23 In animal studies, when FGF is modification techniques provide additional isoforms.
applied to the rabbit or mouse cornea, directed ingrowth of
new vessels is stimulated.24,25 Increased levels of FGF has Structure
not only been implicated in vessel growth but becomes lim- The gene for FGF-1 is located on chromosome 5 be-
ited when neovascularization ceases.23 tween bands 5q31.3 and 5q33.2, while the FGF-2 gene is on
chromosome 4.35 These two genes are similar in their orga-
FGF Characterization nization, as they both have three exons separated by two large
introns. The major difference in these two genes is the loca-
Isoforms tion of the amino-terminus of the two proteins. Nucleotide
Currently the FGF gene family comprises nine mem- sequence analysis of FGF-1 reveals that the open reading
bers (FGF-1 through FGF-9) in mammals. All are structur- frame is flanked by termination codons. The primary se-
ally related, with a homology from 35-55% and generally quence of FGF-2, however, has revealed an amino-terminal
encode proteins with a molecular mass of 20-30 kDa. Over sequence that extends 5' to a proposed initiator, methion-
the course of time and as research has developed, so has the ine.36 FGFs also possess a distinct structural feature known
nomenclature and characterization of FGF (Table 28.1). as a nuclear localization sequence, located near the NH2 end
In 1980, FGF-1 was discovered by detecting the pres- of the protein. This sequence in FGF-1 plays a role in mov-
ence of a polypeptide with a distinct acidic isoelectric point ing extracellular FGF towards the nucleus in a receptor-de-
from extracts of bovine brain.6 FGF-2 was purified by pendent manner. FGF-2 has similar features but has been
Abraham in 1986 by using cDNA clones from kidney, heart, found to be more complicated. FGF-2 mRNA contains mul-
liver, placenta and breast carcinoma.26 FGF-3 was identified tiple translational start sites and the resulting proteins are
as the site of insertion of the mouse mammary tumor virus. transferred to either a cytosolic or nuclear locale.37
Insertion of viral DNA within genomic int-2 led to gene ac- FGF-1 and FGF-2 are single chain polypeptides which
tivation and transformation of infected cells.27 FGF-4 and share 55% sequence homology. The complete structure of
FGF-5 were identified by screening neoplastic cells for the bovine FGF-1 was first described in 1985.38 The final struc-
presence of genes capable of transforming 3T3 fibro- ture is the result of multiple proteolytic cleavages of a more
blasts.28,29 Delli Bovi transfected DNA from Kaposi’s sar- primitive form. Sequence analysis of human FGF-1 demon-
coma into fibroblasts and two new mRNAs were produced. strates similar amino-terminal truncations, with a 92% se-
The protein product of one was termed FGF-4. It was 206 quence identity with the bovine protein.39 FGF-1 is an an-
amino acids in length had significant homology to both ionic mitogen with a molecular weight ranging from 15,000-
FGF-1 and FGF-2. Subsequently, Zahn discovered FGF-5 17,000 kDa. The X-ray crystal structure of human FGF-1
after transfecting fibroblasts with human tumor DNA. Us- has been identified.40 It has been shown to contain four in-
ing a human hst specific probe to screen a mouse cosmid dependent molecules arranged in an asymmetric unit. Each
library, Marics found FGF-6. Tanaka showed that a mouse molecule contains a sulfate ion which stabilizes the other
mammary carcinoma cell line was stimulated by a growth molecules through a hydrogen bond interaction, a potential
factor which was FGF-7.30 FGF-7 is a keratinocyte cell mi- heparin binding site.
togen31 which possesses a DNA sequence complementary FGF-2 is a cationic mitogen with an isoelectric point
of 9.6. Its molecular weight is 18,000 kDa. FGF-2 has been
found to contain 146 amino acids. Within this structure are
Table 28.1. Nomenclature of the FGF family two sequences similar to known heparin binding sites.9
FGF Secretion
Historical Name (23)
The FGF prototypes regulate biological activities as
FGF-1 acidic FGF, HBGF-1, ECGF
an extracellular protein; therefore it is important to exam-
FGF-2 basic FGF, HBGF-2 ine how these proteins are released (or secreted) from cells.
FGF-3 Int-3 FGF-1, FGF-2 and FGF-9 have been found to lack a “tradi-
FGF-4 Kaposi’s FGF, KS3, Hst-1 tional” leader sequence (similar to the interleukin family)
FGF-5 and consequently secretion via the endoplasmic reticulum
FGF-6 Hst-2 does not occur. When the endoplasmic reticulum-Golgi ap-
FGF-7 Keratinocyte growth factor (KGF) paratus path is disrupted by chemical agents, the release of
FGF-8 Androgen inducible growth factor
FGF-9
FGF-1 is not inhibited.41 It is thought that FGF is released in
304 Tissue Engineering of Prosthetic Vascular Grafts
the form of a dimer which is then separated by a reducing potentiates the biological activity of FGF-1 but does not have
agent. The monomer form of FGF is able to interact with a profound effect on the mitogenic activity of FGF-2. Both
the FGF receptors37,42 (Fig. 28.1). Various methods of FGF forms of FGF are protected from degradation when associ-
release have been proposed. For example, it has been dem- ated with heparin. It is suggested that this is due to the sta-
onstrated that the release of FGF-1 is regulated by tempera- bilization of FGF’s tertiary structure and prevention of pro-
ture. NIH 3T3 cells secrete FGF-1 in response to heat shock,43 teolytic modification. Heparin has additionally been found
and terminate release following treatment with either acti- to protect FGF from heat50,51 and acid51 inactivation. FGF-1
nomycin D or cyclohexamide. Recent studies of the kinetics is further protected from trypsin, plasmin50 and thrombin.52
of FGF-1 release suggest that the FGF-1 secretion pathway As previously mentioned, FGF is found in association with
may be limited to newly translated FGF-1.22 Inflammation heparin in the extracellular matrix and basement membranes
has also been proposed to serve as the initial stressor releas- of blood vessels. FGF is ultimately released from its interac-
ing FGF. FGF-2 has also been found to be secreted by an tions with heparin by an enzyme called heparinase, which is
independent pathway.44 FGF-9 also lacks a typical N-termi- expressed by platelets, neutrophils and lymphoma cells.46
nal signal sequence, but has been found to be secreted fol-
lowing transfection into COS cells.45 Biological Activity
In contrast, the remaining members of the FGF fam-
ily possess a functional signal sequence. Some of the FGF Tissue Expression
genes have been identified as oncogenes, reflecting an abil- High levels of FGF-1 are found in neural tissue.54 The
ity to function in an autocrine manner. For example, FGF-4 structural characterization of pituitary FGF was described
(hst-1) is a frequently identified oncogene in transforma- in 1985. In 1987 it was demonstrated that FGF is located in
tion assays of NIH 3T3 cells. the subendothelial extracellular matrix53 and basement
Storage of FGF has been localized to the extracellular membrane, particularly in arterial and capillary walls. FGF-1
matrix and basement membranes of medium and large is not as widely distributed as FGF-2, which is located in
blood vessels, as well as the anastomosis site of branching brain, kidney, adrenal, corpus luteum and the macrophage
capillaries. Heaprin sulfate, which is known to prevent pro- cell lines.23 Using radiolabeled immunoassays, circulating
teolytic degradation of FGF, is also found in high concen- FGF was established in bovine, rat and human serum. Since
trations in these areas. FGF is released from the extracellu- FGF is involved in cell growth, it stands to reason that its
lar matrix in a biologically active form.46 distribution would be widespread.
RSH FGFR-1
HSPG
FGF-1 FGF-1
dimer monomer
Fibroblast Growth Factors in Angiogenesis and Tissue Engineering 305
granulation tissue. FGF is chemotactic for fibroblasts. It also Since angiogenesis plays such an important role in
acts as a negative regulator of collagen metabolism as dem- tumor biology, it has been postulated that the inhibition of
onstrated by a decrease in collagen production in cultured angiogenic growth factors may ultimately affect tumor
fibroblasts. growth and overall outcome. A number of different tech-
niques have been utilized to inhibit the effects of FGF. One
Smooth Muscle Cells particular model uses a monoclonal antibody against FGF-2
FGF-2 has been found to stimulate vascular smooth and successfully suppressed solid tumor growth.62 A differ-
muscle cell proliferation. Amplification of DNA synthesis ent approach to inhibit FGF is based on molecular targeting
occurs within the smooth muscle cell due to autocrine ef- of the gene transcripts. For instance, the use of antisense
fects of the FGF.55 Smooth muscle cell proliferation is also oligodeoxynucleotides will inhibit translation and reduce the
stimulated by FGF-1, but only in the presence of heparin (at stability of targeted RNA. For FGF-2 this method has been
physiologically relevant concentrations of FGF-1). shown to inhibit primary melanoma proliferation in tissue
culture.63
Neuroendocrine Cells
Both FGF-1 and FGF-2 are widely distributed Wound Healing
throughout the central nervous system. It has been found Wound healing consists of a series of complex pro-
that FGF has an effect on glial cells, playing a role in the cesses comprising inflammation, neovascularization, granu-
modulation of cell proliferation.56 lation tissue formation and matrix remodeling, which pro-
ceed in an overlapping manner. Exogenously applied FGF-2
In Vivo has been known to induce potent angiogenesis and granu-
The biological effects of FGF in vivo have been stud- lation tissue formation, and to result in stimulation of wound
ied in both natural and pathological processes such as an- healing in animal models. Localized immunostaining with
giogenesis (as previously described), intimal hyperplasia, antibodies shows increased FGF-2 levels in burn wounds.64
neoplasia, and wound healing. Using a variety of “healing-impaired” mice (such as obese,
diabetic, steroid treated, malnourished, etc.) the effects of
Intimal Hyperplasia FGF were observed. Each mouse sustained an injury (such
FGF has been found to be a smooth muscle cell mito- as a burn wound, decubitus ulcer, excisional wound, and so
gen and may contribute to the initial smooth muscle cell forth) and each wound was covered with an occlusive dress-
proliferation, playing a critical role in the development of ing, some with the application of FGF-2. The wound heal-
intimal hyperplasia. When rat arteries are injured to induce ing rate was monitored. It was found that the FGF-2 treat-
intimal hyperplasia, the response is limited when antibod- ment resulted in an acceleration of wound healing in every
ies to FGF-2 are applied, causing a significant reduction in condition.65 FGF has been found to have two major contri-
the smooth muscle cell response.57 By encoding porcine ar- butions to wound healing: It facilitates neovascularization
terial endothelial cells with a FGF-1 gene, increased intimal and causes a decrease in collagen production, ultimately re-
thickening was demonstrated after 3 weeks as compared to sulting in tissue with a reduced tensile strength.66
a control group.58 Finding ways to inhibit the smooth muscle
cell response to FGF while concomitantly stimulating en- Receptors
dothelial cell progression has been a major area of vascular Early studies suggested that FGF-1 and FGF-2 were
research and tissue engineering. bound by the same receptor.67 But later experiments revealed
that both high and low affinity binding sites exist.
Neoplasia
Tumor angiogenesis has properties somewhat differ- High Affinity Receptors
ent from normal angiogenesis. The fast growth of tumor The high affinity receptors for FGF-1 and FGF-2 have
cells requires that angiogenesis takes place at a high rate in been located on a wide range of cell types such as endothe-
order to keep up with the needs of an expanding tumor. lial cells, hepatoma cells, smooth muscle cells, fibroblasts,
FGF-1 and FGF-2 are found to be associated with the ma- smooth muscle cells, retinal cells, chondrocytes, hepatocytes,
jority of adult tumors (for instance, breast, lung, prostate myoblasts, and various tumor cells.68 The number of sites
cancer, renal cell and bladder carcinoma, brain tumors, hepa- per cell varies from 2,000 to over 100,000 and the molecular
tocellular carcinoma, melanoma, Kaposi’s sarcoma and oth- weight ranges from 105-165 kDa, depending on the type of
ers). A study of FGF localization in gastrointestinal tumors cell studied. The majority of FGF receptors are utilized by
found FGF-2 in the extracellular matrix. Cellular staining both FGF-1 and FGF-2. However, there is some heterogene-
also revealed FGF to be present in fibroblasts and endothe- ity among receptors, dependent on varying degrees of
lial cells of the host.59 There is a reported correlation be- glycosylation.
tween blood vessel density versus metastatic lesions in breast, The varying FGF receptor isoforms are produced by
lung, prostate, and head and neck cancers.60 This correla- the same gene. Since 1989 there have been four FGF recep-
tion was tested by injecting mice with anti-FGF antibodies, tor genes identified. The nomenclature is varied: FGFR1 (flg,
and a decrease in tumor blood vessel density was observed. bFGR, Cek1, N-bFGFR, h2, h3, h4, h5), FGFR2 (bek, Cek3,
FGF is even detectable at significant levels in the serum and K-sam, TK 14, TK 25, KGFR), FGFR3 (Cek2), and FGFR4.
urine of cancer patients.61 The FGF receptor genes share a large sequence homology
306 Tissue Engineering of Prosthetic Vascular Grafts
with each other. The gene structure reveals two mechanisms The low affinity receptor present on the cell surface
for the formation of the receptor isoforms: alternative mRNA shows many similarities to heparin. In fact, when cells are
splicing, resulting in deletions or alternate exon usage; and incubated with heparin, the interaction of FGF-2 and this
internal polyadenylation, resulting in truncated products.23 receptor is inhibited.69 Cloning of this receptor demonstrated
Thus the FGF receptor system is redundantly specific. That a cell-surface heparan proteoglycan. Heparan sulfates are
is, one receptor may bind with several different FGFs or, N-sulfated polysaccharide components of proteoglycans. It
alternatively, one FGF may interact with a number of is synthesized by most vertebrate cells, as compared to hep-
varying receptors. arin which is an exclusive product of connective tissue mast
The structure of the high affinity receptor, a trans- cells. Heparan sulfate and heparin synthesis occur within
membrane glycoprotein, is divided into an extracelluar and the Golgi complex, beginning with the nonsulfated precur-
intracellular component. The extracellular portion contains sor heparan. This precursor is transformed to heparan sul-
three Ig-like domains which vary in expression by alterna- fate or heparin by a series of polymer modifications. This
tive splicing; this area of the receptor is the least conserved results in heparin containing a significant higher concen-
region. The first Ig domain is not essential for the high af- tration of sulfate groups as compared to heparan sulfate.
finity binding of FGF-1 or FGF-2. There is also a signal pep- Heparan sulfates are mainly present on cell surfaces and in
tide and acid box region in the extracellular portion of the the extracellular matrix as proteoglycans. It is well estab-
receptor. The intracellular component has two tyrosine ki- lished that the heparan sulfates bind to a wide variety of
nase domains which are the most highly conserved regions cytokines and specifically to FGF-1 and FGF-2.70
(Fig. 28.2). The low affinity receptor is now known to be syndecan.
Syndecan is a proteoglycan initially cloned from mouse. It
Low Affinity Receptors is a polymorphic molecule with heparan and chondoitin
The low affinity receptors have been located on sulfate chains associated with a 31 kDa transmembrane
fibroblasts, endothelial cells, epithelial cells, neurons, tumor polypeptide. It is mainly found on the surface of epithelial
cells and in the basement membrane of a variety of tissues. cells from mature tissues such as skin, liver, and breast.68
These binding sites range from 0.5 x 106 to several million Syndecan is a family of proteoglycans (syndecan 1, 2, 3 or 4)
in number. based on four specific core proteins. They can interact with
acid box
lg loop II
lg loop III
transmembrane domain
varying components of the matrix like fibronectin and col- Signal Transduction
lagen as well as FGF-2.72 FGF signaling immediately follows the binding of FGF
The amino acid sequence of the core protein contains to its receptor. Heparin is required for this interaction to
a transmembrane domain with an extracellular region con- occur. Although the exact mechanism is unclear, it is thought
taining six potential attachment sites for glycosaminogly- that heparin helps stabilize the FGF-FGF receptor complex.
can side chains.71 The receptor has been characterized to The binding leads to FGF receptor dimerization,
two major components: a proteoglycan of 250 kDa which autophosphorylation and an increase in the tyrosine kinase
appears in the plasma membrane and an 800 kDa which is activity. This initiates a responding cascade of events with
located in the extracellular matrix. It is the extracellular com- activation of a variety of signaling molecules, ultimately re-
ponent that can vary, allowing for diversity.72 sulting in transcription of genes necessary for the FGF re-
It is suggested that the low affinity receptor is an ac- sponse76 (Fig. 28.3).
cessory molecule required for the binding of FGF to the high
affinity receptor. Using cells deficient in heparan sulfates, it FGF and Vascular Grafts
was found that FGF-2 could not bind to its high affinity re- The major cause of failure of vascular grafts is 2-fold:
ceptor.73 Similarly, treatment of skeletal muscle or adrenal Early on it is due to thrombosis and later the primary cause
cells with heparinase demonstrated FGF’s inability to inter- is myointimal hyperplasia. The interface between the blood
act with its high affinity receptor, with a decrease in cell pro- and graft wall is where these pathological processes occur.
liferation.74,75 All of this data implicates the low affinity FGF In 1978 Herring developed the concept that endothe-
receptor as an accessory molecule, required for the lial cells transplanted onto a graft surface could prolong the
complexing of FGF in a biologically active form to its high graft’s survival.77 The presence of a confluent monolayer of
affinity receptor. endothelial cells could theoretically improve graft thrombo-
Fig. 28.3. Schematic presentation of intracellular signaling mediated by the FGF receptor (FGFR). The FGFR is activated upon
binding FGF and associated heparan sulfate proteoglycan (HSPG). The FGFR undergoes autophosphorylation on tyrosine resi-
dues. A signal transduction cascade results in the activation of various proteins, including the F-actin-binding protein cortactin.
Further propagation of the signaling cascade induces the activity of MAP kinase (MAPK), which phosphorylates transcription
factors (TF), activating the genes necessary for the FGF response. (Reproduced with permission from ref. 37)
308 Tissue Engineering of Prosthetic Vascular Grafts
resistance and prevent the development of intimal hyper- inhibit smooth muscle cells.89,90 Additional in vitro studies
plasia. Three sources of endothelial cells seen on a prosthetic have shown that at an optimized FGF-1 to heparin ratio,
vascular lumen have been observed. Endothelial cells may using a high dose of heparin, endothelial cell proliferation
infiltrate from a native artery over the anastomosis, are ob- could selectively be stimulated while smooth muscle cells
served as ingrowth through the interstices originating from are inhibited.91
host perigraft tissue, and may occasionally be circulating. In Others have studied the release of FGF from prosthetic
humans it is rare to see complete endothelialization on a grafts. For instance, Yamamura applied FGF-2 to 0.75 mm
prosthetic graft, even after it has been well incorporated into disks made of ePTFE. Biodegradable hydroxypropylchitosan
the host tissue. Direct seeding of endothelial cells onto pros- acetate (HPCHA) was applied to control the rate of release
thetic grafts has been extensively investigated, but in clinical of FGF from these disks. The disks were implanted into rab-
trials the benefits have been modest.78-81 One of the diffi- bit skin pockets and harvested over 24 hours. The disks
culties with this technique is the relatively low cell density treated with HPCHA and FGF released only 60% of the
initially applied to the graft and inadequate cell attachment. FGF-2, while disks treated with FGF-2 alone released all of
The use of specific endothelial cell chemoattractants and the cytokine in a 24 hour period. It was concluded that
mitogens, such as FGF, can theoretically stimulate HPCHA allowed the slow, reliable release of FGF from pros-
transinterstitial capillary ingrowth, resulting in enhanced thetic material.92 In another experiment, bone marrow cells
endothelialization of the graft surface. were applied to ePTFE vascular grafts and implanted into
Work by Greisler’s group has evaluated the applica- the abdominal aorta of dogs. At six months time the grafts
tion of FGF to synthetic surfaces as a means to either en- were still immunohistochemically reactive to FGF-2.93
hance spontaneous endothelialization of that surface or to The following experiments examined the FGF reten-
stimulate the proliferation of seeded endothelial cells. In early tion within injured vessels. Using a balloon injury model,
studies, FGF-1 was applied to both Dacron and [125I]FGF-1 in FG was applied to the carotid arteries of dogs.
polydioxanone (PDS) grafts by means of sequential appli- The result revealed a 41% and 37% retention of FGF-1 after
cation of fibronectin, heparin, FGF-1 and a second layer of 10 and 60 minutes of circulation respectively.94,95 In a simi-
heparin. Using [125I]FGF-1, the retention of the growth fac- lar fashion, the retention of 111Indium-labelled platelets was
tor in vivo was quantitated. One week after the application, quantitated in FG versus untreated identically injured
the retention was 44% in the Dacron grafts and 23% in the carotids. Platelet deposition was a significant 45% less in
PDS grafts.82,83 After the graft was explanted, the FGF-1 was the FG group.95 These studies seem to indicate that the ap-
eluted and shown to have retained its mitogenic activity on plication of FG to arterial surfaces following injury has po-
quiescent lung endothelial cells.83 A study by Tomizawa used tential clinical utility. The incorporation of FGF-1 plus hep-
minced canine adipose tissue that was applied to vascular arin within FG onto synthetic grafts may promote
grafts. These were implanted into the abdominal aorta of neovascularization and prevent pseudointimal hyperplasia,
dogs. When the grafts were removed, endothelial-like cells but this is still under investigation.
extending onto luminal thrombus was observed. These cells One technique that is now being utilized is the use of
were immunohistologically positive for FGF-2.84 “mutant” forms of FGF-1. In other words, by making spe-
Further studies have utilized fibrin glue (FG), a com- cific mutations in the structure of FGF, perhaps a growth
posite of thrombin and fibrinogen, as a controlled local de- factor with a more specific function can be developed. For
livery system for FGF-1 and heparin. Three dimensional example, an extensive characterization of FGF-1 mutants of
analysis of the FGF distribution has revealed that it was even lysine 132 to a glutamic acid or a glycine residue has been
throughout the graft.85 Studies comparing the fibronectin completed.96,97 This altered form of FGF-1 in bone assays
treatment to the FG illustrated greater endothelial cell re- has been shown to induce angiogenesis with a decease in
tention with the latter.86 Expanded polytetrafluoroethylene fibroblast proliferation. These functional characteristics, se-
(ePTFE—60 micron internodal distance) grafts were treated lective stimulation of endothelial cells and inhibiting fibro-
with the FG-FGF-1-heparin combination and implanted in blasts, are qualities that are likely to be beneficial when ap-
dogs as 5 cm aortoiliac grafts and 30 cm thoracoabdominal plied to models of vascular healing.
aortic grafts. When compared to control groups, the pre- To determine the relative percent coverage of seeded
treated grafts resulted in a significant increase in endothe- versus spontaneously ingrowing endothelial cells on FG
lial cell proliferation as assayed by en face autoradiography. treated surfaces, a method of fluorescent labeling with
There was also extensive transinterstitial capillary ingrowth PKH-26 has been developed. Proliferation assays in vitro
observed throughout the graft wall.87,88 Cross-sectional au- documented virtually identical growth rates for labeled ver-
toradiography showed a similar increase in subendothelial sus unlabeled canine jugular vein endothelial cells. There is
myofibroblast proliferation in the pretreated grafts at one nearly a linear relationship between the concentration of
month. Analysis at 140 days revealed the FG-FGF-1-hep- applied PKH-26 (from 0-10 ∝M of PKH-26) and the mean
arin grafts had developed a significantly thicker inner cap- intensity of fluorescence of labeled endothelial cells. Addi-
sule (139 microns versus <95 microns in the control groups) tionally, no PKH-26 induced toxicity or alterations in growth
consisting of myofibroblasts and collagen.88 kinetics were observed using concentrations of PKH-26 up
Although heparin and FGF-1 act synergistically to to 10 ∝M (although cellular toxicity was detected at 20 ∝M
stimulate smooth muscle cell and fibroblast proliferation, concentrations). Labeled endothelial cells are visualized on
in the absence of FGF-1, heparin alone has been shown to both Dacron and ePTFE surfaces under fluorescent illumi-
Fibroblast Growth Factors in Angiogenesis and Tissue Engineering 309
nation.98 This cell labeling technique is now being applied collateral vessels were found to be augmented.107 In a simi-
to study the effects on balloon injured carotid arteries treated lar fashion, FGF-1 was administered after the ischemia was
with FG plus FGF-1 and heparin and PKH-26 labeled au- allowed to subside for 10 days. Even after 30 days there was
tologous endothelial cells. evidence of angiogenesis and increased collateral blood flow
in the rabbit hindlimb.108 This method of investigation has
FGF and Ischemia been applied to other angiogenic growth factors such as
Compromised vascular circulation is an important VEGF109 (see chapter 26 for a full review).
component in many disease processes such as coronary is- The interest in FGF and ischemic disease has broad-
chemia, congestive heart failure, and peripheral vascular in- ened, and research is now being done on brain infarcts
sufficiency. In these conditions there is insufficient capillary (strokes)110,111 and trauma injuries.112
flow, which contributes to the extent of the damage to the The therapeutic potential of angiogenic cytokines re-
tissue. It is thought that increasing the number of capillar- mains to be fully defined, but clearly FGF plays an impor-
ies, and ultimately the oxygen supply, in these underperfused tant role in the development of collateral neovascularization
tissue beds may provide a palliative effect, thus limiting ne- in ischemic disease states.
crosis or loss of function. Angiogenesis has been investigated
in terms of its relation to the development of collateral blood Conclusions
flow. Studies examining canine and swine coronary circula- Over the past 15 years our knowledge and understand-
tions have established that cellular proliferation is a feature ing of angiogenic cytokines, FGF in particular, has made
of collateral vessel development that occurs as a result of great strides. Current vascular research is focused on more
arterial occlusion.99,100 By using angiogenic growth factors, bio-interactive grafts that optimize the microenvironment
such as FGF, collateral artery development has been of the tissue-graft-blood interfaces. The application of en-
augmented. dothelial cells and growth factors to synthetic grafts to in-
Since cardiac disease is a major cause of morbidity duce angiogenesis and limit the smooth muscle cell response
and mortality, the effects of FGF on myocardial are some of the most promising areas of research.
neovascularization have been studied extensively. When rab-
bits with chronic myocardial ischemia were subjected to References
pericardial injections of FGF-2, there was a significant in- 1. Armelin, HA. Pituitary extracts and steroid hormones in
crease in the growth of small vessels.101 To study the effect the control of 3T3 cell growth. Proc Natl Acad Sci USA
of FGF on acute myocardial damage, dog right ventricles 1973; 70:2702-6.
were injected with FGF and heparin after an induced myo- 2. Gospodarowicz D, Jones KL, Sato G. Purification of a
growth factor for ovarian cells from bovine pituitary
cardial event. Examination after one month revealed less area
glands. Proc Natl Acad Sci USA 1974; 71:2295-99.
of infarction in the treated cardiac tissue as compared to the 3. Gospodarowicz D. Localisation of a fibroblast growth fac-
right ventricle.102 Sellke studied chronic myocardial is- tor and its effect alone and with hydrocortisone on T cell
chemia, showing that chronic perfusion through collateral growth. Nature 1974; 249:123-7.
vessels in the heart impairs endothelium dependent relax- 4. Gospodarowicz D. Purification of a fibroblast growth fac-
ation and promotes vasoconstriction in the microcircula- tor from bovine pituitary. J Biol Chem 1975; 250:2519-19.
tion.103 In the pig model Selke administered FGF to coro- 5. Westfall FC, Lenon VA, Gospodarowicz D. Brain derived
nary arteries and preserved endothelial relaxation in newly fibroblast growth factor: Identity with a fragment of the
formed collaterals.104 By using endothelial cells cultured from basic protein myelin. Proc Natl Acad Sci USA 1978;
human heart transplant patients, FGF-2 was applied to cells 75:4675-78.
6. Thomas KA, Riley MC, Lemmon SK, Baglan NC,
from tissue with either coronary artery disease or idiopathic
Bradshaw RAS. Brain derived fibroblast growth factor:
myopathy (noncoronary artery disease). The number of Non identity with myelin basic protein. J Biol Chem 1980;
capillary-like microtubules, a reflection of angiogenesis, was 255:5517-20.
increased in the cells from the coronary artery disease group. 7. Lemmon SK, Riley MC, Thomas KA, Hoover GA, Maciag
This demonstrated that diseased vessels, i.e., coronary artery T, Bradshaw RA. Bovine fibroblast growth factor: Com-
disease, respond better to FGF with neovascularization.105 parison of brain and piuitary preparations. J Cell Biol
It has been reported that FGF-2 is a circulating peptide and 1982; 95:162-9.
gene expression is increased after myocardial ischemia. Us- 8. Gimenez-Gallego G, Rodkey J, Bennett C, Rios-Candelore
ing a FGF-2 enzyme-linked immunoassay, FGF-2 levels in M, DiSalvo J, Thomas KA. Brain-derived acidic fibroblast
the blood were determined in patients experiencing an acute growth factr: Complete amino acid sequence and homolo-
myocardial infarction. It was found that there was an in- gies. Science 1985; 230:1385-1388.
9. Baird A, Esch F, Mormede P, Ueno N, Ling N et al. Mo-
creased serum level of FGF-2 10 days after the ischemic event,
lecular characterization of FGF: Distribution and biologi-
suggesting that FGF-2 at this time mediates the development cal activities in various tissues. Recent Prog Horm Res
of collateral coronary circulation.106 1986; 42:143-205.
The development of collateral vessel development in 10. Miyamoto M, Naruo KI, Seko C, Matsumoto S, Kondo
the setting of lower extremity ischemia has also been inves- T, Kurokawa T. Molecular cloning of a novel cytokine
tigated. Lower limb ischemia is generated by surgically ex- cDNA encoding the ninth member of the fibroblast
cising the femoral artery in rabbits. FGF-2 is applied through growth factor family which has a unique secretion prop-
intramuscular injections; after 14 days capillary density and erty. Mol Cell Biol 1993; 13:4251-59.
310 Tissue Engineering of Prosthetic Vascular Grafts
11. Sholley MM, Ferguson GP, Seibel HR, Montuor JL., Wil- androgen-dependent growth of mouse mammary carci-
son JD. Mechanisms of neovascularization. Vascular noma cells. Proc Natl Acad Sci USA 1992; 89:8928-32.
sprouting can occur without proliferation of endothelial 31. Finch P, Rubin JS, Miki T, Ron D, Aaronson SA. Human
cells. Lab Invest 1984; 51:624-34. KGF is FGF related with properties of a paracrine effec-
12. Diaz-Flores L, Guiterrez R, Valladares F, Varela H, Perez tor of epithelial cell growth. Science. 1989; 245:752-55.
M. Intense vascular sprouting from rat femoral vein in- 32. Acland P, Dixon M, Peters G, Dickson C. Subcellular fate
duced by prostaglandins E1 and E2. Anat Rec 1994; of the Int-2 oncoprotein is determined by choice of
238:68-76. intiation codon. Nature 1990; 343:662-65.
13. Diaz-Flores L, Gutierrez R, Varela H. Angiogenesis: An 33. Powell PP, Klagsbrun M. Three forms of rat basic fibro-
update Histol Histopath 1994; 9:807-843. blast growth factor are made from a single mRNA and
14. Burger PC, Chandler DB, Klintworth GK. Corneal localise to the nucleus. J Cell Physiol. 1991; 148:202-10.
neovascularization as studied by scanning electron micros- 34. Zuniga AZM, Meijers C, Zeller R. Expression of alterna-
copy of vascular casts. Lab Invest 1983; 48:169-80. tively spliced bFGF first coding exons and antisense RNAs
15. Ausprunk DH, Folkman J Migration and proliferation of during chicken embryogenesis. Dev Biol 1992; 157:110-8.
endothelial cells in preformed and newly formed blood 35. Mergia A, Eddy R, Abraham JA, Fiddes JC, Showes TB.
vessels during tumor angiogenesis. Microvasc Res 1977; The genes for basic and acidic fibroblast growth factor
14:53-65. are on different chromosomes. BioChem Biophys. Res
16. Rifkin DE, Gross JL, Moscatelli D, Jaffe EA. Proteases and Commun 1986; 136:644-51.
angiogenesis: Production of plasminogen activation and 36. Jaye M, Howk R, Burgess WH, Ricca G, Chiu IM, Ravera
collagenase by endothelial cells. In: Nossel HL, Vogel ID, MW, O’Brien SJ, Modi WS, Maciag T, Drohan WN. Hu-
eds. Pathobiology of the endothelial cell. New York: Aca- man endothelial cell growth factor: Cloning, nucleotide
demic Press Inc., 1982:191-197. sequence, and chromosome localization. Science 1986;
17. Folkman J. How is blood vessel growth regulated in nor- 233:543-45.
mal and neoplastic tissue? Cancer Res 1986; 46:467-73. 37. Freisel R, Maciag T. Molecular mechanisms of angiogen-
18. Bavisotto LM, Schwartz SM, Heimark RL. Modulation of esis: FGF signal transduction. FASEB J 1995; 920
Ca-dependent intercellular adhesion in bovine aortic and (9):919-925.
human umbilical vein endothelial cells by heparin-bind- 38. Gimenez-Gallego G, Rodkey J, Bennett C, Rios-Candelore
ing growth factors. J Cell Physiol 1990; 143:39-51. M, DiSalvo J, Thomas KA. Brain-derived acidic fibroblast
19. Liaw L, Schwatrz SM. Microtubule disruption stimulates growth factor: Complete amino acid sequence and
DNA synthesis in bovine endothelial cells and potentiates homolgies. Science. 1985; 230:1385-88.
cellular response to basic fibroblast growth factor. Am J 39. Burgess WH, Maciag T. The heparin-binding (fibroblast)
Pathol 1990; 143:937-48. growth factor family of proteins. Annu. Rev BioChem
20. Wakui S. Two and three dimensional ultrastructural ob- 1989; 58:575-606.
servation of two cell type angiogenesis in human granu- 40. Blaber M, DiSalvo J, Thomas K. X-ray crystal structure
lation tissue. Virchows Arch (B) 1988; 56:127-39. of human acidic fibroblast growth factor. BioChem 1996;
21. Orlidge A, D’Amore PA. Inhibition of capillary endothe- 35:2086-2094.
lial cell growth by pericytes and smooth muscle cells. J 41. Jackson A, Tarantini F, Gamble S, Friedman S, Maciag T.
Cell Biol 1987; 105:1455-62. Release of fibroblast growth factor-1 from NIH 3T3 cells
22. Sato Y, Rifkin DB. Autocrine activities of basic fibroblast in response to temperature involves the function of cys-
growth factor: regulation of endothelial cell movement, teine residues. J Biol Chem 1995; 270:33-6.
plasminogen activator synthesis, and DNA synthesis. J Cell 42. Mason I. The ins and outs of fibroblast growth factors.
Biol 1988; 107:1199-205. Cell; 1994; 78:547-552.
23. Slavin J. Fibroblast growth factor. At the heart of angio- 43. Jackson A, Freidman S, Zhan X, Engleka KA, Forough R,
genesis. Cell Biol International 1995; 19:431-44. Maciag T. Heat shock induces the release of fibroblast
24. Folkman J. Angiogenesis factors. Science 1987; 235:442-7. growth factor-1 from NIH-3T3 cells. Proc Natl Acad Sci
25. Kenyon BM, Voest E, Chen C, Flynn E, Folkman J, USA 1992; 89:10691-5.
D’Amato R. A model of angiogenesis in the mouse cor- 44. Mignatti P, Morimoto T, Rifkin DB. Basic fibroblast
nea. Invest Opth. & Visual Science 1996; 37(8):1625-32. growth factor, a protein devoid of secretory signal se-
26. Abraham JA, Whang JL, Tumolo A, Mergia A, Friedman quence, is released by cells via a pathway independent of
J, Gospodarowicz D, Fiddes JC. Human basic fibroblast the endoplasmic reticulum-Golgi complex. J Cell Physiol
growth factor: Nucleotide sequence and genomic organi- 1992; 151:81-93.
zation. EMBO J 1986; 5:2523-8. 45. Miyamoto M, Naruo K, Seko C, Matsumoto S, Kondo T,
27. Dickson C, Peters G. Potential oncogene product related Kurokawa T. Molecular cloning of a novel cytokine cDNA
to growth factors. Nature 1987; 326:830-33. encoding the ninth member of the fibroblast growth fac-
28. Deli Bovi P, Curatola AM, Kern FG, Greco A, Ittman M, tor family which has a unique secretory property. Mol
Bascilico C. An oncogene isolated by transfection of Cell Biol 1993; 13:4251-4259.
Kaposi’s sarcoma DNA encodes a growth factor that is a 46. Vlodavsky I, Bashkin P, Ishai-Michaeli R, Chajek-Shaul
member of the FGF family. Cell 1987; 50:729-37. T, Bar-Shavit R, Haimovitz-Friedman A, Klagsburn M,
29. Zahn X, Bates B, Hu X, Goldfarb M. The human FGF-5 Fuks Z. Sequestration and release of basic FGF. Annals
gene encodes a novel protein related to fibroblast growth NY Acad Sci 1991; 638:207-220.
factors. Mol Cell Biol 1988; 8:3487-97. 47. Thornton SC, Mueller SN, Levine EM. Human endothe-
30. Tanaka A, Miyamoto K, Minamino M, Takeda M, Sato lial cells: Use of heparin in cloning and long term serial
B, Matsuo H, Matsumoto K. Cloning and characterisation cultivation. Science 1983; 222:623-5.
of an androgen-induced growth factor essential for the 48. Maciag T, Mehlman T, Freisel R, Schriber AB. Heparin
binds endothelial cell growth factor, the principal endo-
Fibroblast Growth Factors in Angiogenesis and Tissue Engineering 311
thelial cell mitogen in bovine brain. Science 1984; cell ghosts accelerates incisional wound healing. Br J Surg
225:932-35. 1992; 79:918-21.
49. Shing Y, Folkman J, Sullivan R, Butterfield C, Murray J, 67. Neufeld G, Gospodarowicz D. Basic and acidic growth
Klagsburg M. Heparin affinity: Purification of a tumor factors interact with the same cell surface receptors. J Biol
derived capillary endothelial cell growth factor. Science Chem 1986; 261:5631-7.
1984; 223:1296-99. 68. Ledoux D, Gannoun-Zaki L, Barritault D. Interactions of
50. Rosengart TK, Johnson W, Friesel R, Clark R, Maciag T. FGFs with target cells. Progress in Growth Factor Res
Heparin protects heparin-binding-growth-factor-I from 1992; 4:107-20.
proteolytic inactivation in vitro. BioChem Biophys. Res 69. Moscatelli D. High and low affinity binding sites for ba-
Commun. 1988; 152:432-40. sic fibroblast growth factor on cultured cells: Absence of
51. Gospodarowicz D, Cheng J. Heparin protects basic and a role for low affinity binding in the stimulation of plas-
acidic FGF from inactivation. J Cell Physiol 1986; minogen activator production by bovine capillary endot-
128:475-84. helial cells. J Cell Physiol. 1987; 131:123-30.
52. Lobb R. Thrombin inactivates acidic fibroblast growth 70. Gallagher JT, Turnbull JE. Heparan sulfate in the bind-
factor but not basic fibroblast growth factor. BioChem ing and activation of basic fibroblast growth factor.
1988; 27:2572-78. Glycobiology 1992; 2(6):523-8.
53. Vlodavsky I, Folkman J, Sullivan R, Fridman R, Ishai- 71. Kiefer MC, Stephans JC, Crawford K, Okino K, Barr PJ.
Michaeli R, Sasse J, Klagsbrun M. Endothelial cell-derived Ligand-affinity cloning and structure of a cell surface
basic fibroblast growth factor: Synthesis and deposition heparan sulfate proteoglycan that binds basic fibrfoblast
into subendothelial extracellular matrix. Proc Natl Acad growth factor. Proc Natl Acad Sci USA 1990; 87:6985-9.
Sci USA 1987; 84: 2292-6. 72. Bernfield M, Sanderson RD. Syndecan, a developmentally
54. Lobb R, Sasse J, Sullivan R, Shing Y, D’Amore P, Jacobs regulated cell surface proteoglycan that binds extracellu-
J, Klagsbrun M. Purification and characterization of hep- lar matrix and growth factors. Phil Trans R Soc Lond
arin-binding endothelial cell growth factors. J Biol Chem 1990; 327:171-86.
1986; 261:1924-8. 73. Yayon A, Klagsburn M, Esko JD, Leder P, Ornitz DM.
55. Davis MG, Zhou M, Ali S, Coffin JD, Doetschman T, Cell surface, heparin-like molecules are required for bind-
Dorn GW. Intracrine and autocrine effects of basic fibro- ing of basic fibroblast growth factor to its high affinity
blast growth factor in vascular smooth muscle cells. J Mol receptor. Cell. 1991; 64:841-8.
Cell Cardiol 1997; 29(4):1061-72. 74. Rapraeger AC, Krufka A, Olwin BB. Requirement of
56. Engele J, Bohn MC. Effects of acidic and basic fibroblast heparan sulfate for bFGF-mediated fibroblast growth fac-
growth factors on glial precursor cell proliferation: Age tor and myoblast differentiation. Science 1991; 252:1705-8.
dependency and brain region specificity. Dev Biol 1992; 75. Savona C, Chambaz EM, Feige JJ. Proteoheparan sulfates
152:363-72. contribute to the binding of basic FGF to its high affinity
57. Reidy M. Factors contolling the development of arterial receptors on bovine adrenocortical cells. Growth Factors.
lesions after injury. Circ Supl 1992; 86(6):43-6. 1991; 5:273-82.
58. Nabel E. Recombinant FGF-1 promotes intimal hyperpla- 76. Friesel R, Maciag T. Molecular mechanisms of angiogen-
sia and angiogenesis in arteries in vivo. Nature 1993; esis: Fibroblast growth factor signal transduction. FASEB
363:844-6. 1995; 9:919-25.
59. Ohtani H, Nakamura S, Watanabe Y, Mizoi T, Saku T, 77. Herring MB, Gardner AL, Glover J. A single staged tech-
Nagura H. Immunocytochemical localization of basic fi- nique for the seeding of vascular grafts with autogenous
broblast growth factor in carcinomas and inflammatory endothelium. Surgery 1978; 84:498.
lesions of the human digestive tract. Lab Invest 1993; 78. Ortenwall P, Wadenvik H, Risberg B. Reduced platelet
68:520-7. deposition on seeded versus unseeded segments of ex-
60. Wellstein A. Inhibition of FGFs. Breast Ca Res and Trmt panded polytetrafluoroethylene grafts: Clinical observa-
1996; 38: 09-19. tions after a 6-month follow-up. J Vasc Surg 1989;
61. Nguyen M, Watanabe H, Budson AE, Richie JP, Hayes 10:374-80.
DF, Folkman J. Elevated levels of an angiogenic peptide, 79. Ortenwall P, Wadenvik H, Kutti J, Risberg B. Endothelial
basic fibroblast growth factor, in the urine of patients with seeding reduces thrombogenicity of Dacron grafts in hu-
a wide spectrum of cancers. J Natl Cancer Inst 1994; mans. J Vasc Surg 1990; 11:403-10.
86:356-61. 80. Park PK, Jarrell BE, Williams SK, Carter TL, Rose DG,
62. Hori A, Sasada R, Matsutani E, Naito K, Sakura Y, Fujita Martinez-Hernandez A, Carabasi RA. Thrombus-free,
T, Kozai Y. Suppression of solid tumor growth by human endothelial surface in the midregion of a Dacron
immunoneutralizing monoclonal antibody against basic vascular graft in the splanchnic venous circuit- Observa-
fibroblast growth factor. Cancer Res 1991; 51:6180-84. tions after nine months of implantation. J Vasc Surg 1990;
63. Becker D, Meier CB, Herlyn M. Proliferation of human 11:468-75.
malignant melanomas is inhibited by antisense 81. Baitella-Eberle G, Grosscurth P, Zilla P, Lachat M, Muller-
oligodeoxynucleotides targeted against basic fibroblast Glauser W, Schneider J, Neudecker A, von Segesser LK,
growth factor. EMBO J 1989; 8:3685-91. Dardel EE, Turina M. Long-term results of tissue devel-
64. Girban N. Basic FGF in the early human burn wound. J opment and cell differentiation on Dacron prostheses
Surg Res 1994; 56:226-34. seeded with microvascular cells in dogs. J Vasc Surg 1993;
65. Okamura M. Effect of bFGF on wound healing in heal- 18:1019-28.
ing-impaired animal models. Drug Res 1996; 82. Greisler HP, Klosak JJ, Dennis JW, Ellinger J et al. En-
46(1):547-551. dothelial cell growth factor attachment to biomaterials.
66. Slavin J, Hunt JA, Nash JR, Williams DF, Kingsnorth AN. Trans ASAIO 1986; 32:346-9.
Recombinant basic fibroblast growth factor in red blood
312 Tissue Engineering of Prosthetic Vascular Grafts
83. Greisler HP, Klosak JJ, Dennis JW, Karesh SM et al. for tracking during long term studies of autologous trans-
Biomaterials pretreatment with ECGF to augment endo- plantation. Chicago: Presented at the 21st Annual Meet-
thelial cell proliferation. J Vasc Surg 1987; 5:393-402. ing of the Midwestern Vascular Surgical Society. 1997:Sept
84. Tomizaw Y, Noishiki Y, Okoshi T, Nishida H, Endo M, 12-13.
Koyanagi H. Endogenous basic fibroblast growth factor 99. Cowan DF, Hollenberg NK, Connelly CM, Williams DH,
for endothelialization due to angiogenesis in fabric vas- Abrams HL. Increased collateral arterial and venous en-
cular prostheses. ASAIO J 1996; 42(5):M698-702. dothelial cell turnover after renal artery stenosis in the
85. Greisler HP. Dacron inhibition of arterial regenerative dog. Invest Radiol 1978; 13:143-9.
activity. J Vasc Surg 1986; 3:747-56. 100. White FC, Carroll SM, Magnet A, Bloor CM. Coronary
86. Gosselin C, Vorp DA, Warty V, Severyn DA, Dick EK, collateral development in swine after coronary artery oc-
Borovetz HS, Greisler HP. ePTFE coating with FG, FGF-1, clusion. Circ Res 1992; 71:1490-1500.
and heparin: Effect on retention of seeded endothelial 101. Landau C, Jacobs AK, Haudenschild CC. Intrapericardial
cells. J Surg Res 1996; 60:327-32. FGF2 induces myocardial angiogenesis in a rabbit model
87. Greisler HP, Cziperle DJ, Kim DU et al. Enhanced of chronic ischemia. Am Heart J 1995; 129(5):924-31.
endothelialization of expanded polytetrafluoroethylene 102. Uchida Y, Yanagjsawa-Miwa A, Nakamura F, Yamada K,
grafts by fibroblast growth factor type 1 pretreatment. Tomaru T, Kimura K, Morita T. Angiogenic therapy of
Surgery 1992; 112:244-55. acute myocardial infarction by intrapericardial injection
88. Gray JL, Kang SS, Zenni GC et al. FGF-1 affixation stimu- of basic fibroblast growth factor and heaprin sulfate: An
lates ePTFE endothelialization without intimal hyperpla- experimental study. Am Heart J 1995; 130(6):1182-8.
sia. J Surg Res 1994; 57:596-612. 103. Sellke FW, Quillen JE, Brooks LA, Harrison DG. Endot-
89. Reilly CF, Kindy MS, Brown KE et al. Heparin prevents helial modulation of the coronary vasculature in vessels
vascular smooth muscle cell progression through the G1 perfused via mature collaterals. Circulation 1990;
phase of the cell cycle. J Biol Chem 1989; 264:6990-6995. 81:1938-47.
90. Au TYP, Kenagy RD, Clowes MM, Clowes AW. Mecha- 104. Selke F. Basic FGF preserves the endothelial function in
nisms of inhibition by heparin of vascular smooth muscle the microcirculation perfused by collaterals. Abstracts of
cell proliferation and migration. Haemostasis 1993; the Scientific Conference on the Molecular Cellular Biol-
23:177-82. ogy of the Vascular Wall. Boston; Vol. 49; 1993:Oct 15-17.
91. Kang SS, Gosselin C, Ren D, Greisler HP. Selective stimu- 105. Chen CH, Nguyen HH, Weilbaecher D, Luo S, Gotto AM
lation of endothelial cell proliferation with inhibition of Jr, Henry PD. FGF-2 reverses the atherosclerotic impair-
smooth muscle cell proliferation by fibroblast growth fac- ment of human coronary angiogenesis-like responses in
tor-1 plus heparin delivered from fibrin glue suspensions. vitro. Atherosclerosis 1995; 116: 261-8.
Surgery 1995; 118:280-287. 106. Cuevas P, Barrios V, Gimenez-Gallego G, Martinez-Coso
92. Yamamura K, Sakuraj T, Yano K, Nabeshima T, V, Cuevas B, Benavides J, Garcia-Segovia J, Asin-Cardiel
Yotsuyanagi T. Sustained release of bFGF from synthetic E. Serum levels of basic fibroblast growth factor in acute
vascular prosthesis using hydroxypropylchitosan acetate. myocardial infarction. Eur J Med Res 1997; 2(7):282-4.
J BioChem Mat. Res 1995; 29:203-6. 107. Baffour R, Berman J, Garb JL, Rhee SW, Kaufman J,
93. Noishiki Y, Tomizawa Y, Yamane Y, Matsumoto A. Friedmann P. Enhanced angiogenesis and growth of
Autocrine angiogenic vascular prosthesis with bone mar- collaterals by in vivo administration of recombinant FGF-
row transplantation. Nature Medicine. 1996; 2(1):90-3. 2 in a rabbit model of acute lower limb ischemia: Dose-
94. Zarge J, Huang P, Husak V, Kim D, Haudenschild C, response effect of FGF-2. J Vasc Surg 1992; 16:181-91.
Nord RM, Greisler HP. Fibrin glue containing fibroblast 108. Pu L, Sniderman AD, Brassard R, Lachapelle KJ, Graham
growth factor type 1 and heparin with autologous endot- AM, Lisbona R, Symes JF. Enhanced revascularization of
helial cells reduces intimal hyperplasia in a canine carotid the ischemic limb by means of angiogenic therapy. Cir-
artery balloon injury model. J Vasc Surg 1997; culation 1993; 88:208-15.
25(5):840-8. 109. Takeshita S, Rossow S, Kearney M, Zheng L, Bauters C,
95. Greisler HP, Kang SS, Lin P, Hirko MK. In vivo and in Bunting S, Ferrara N, Symes J, Isner J. Time course of
vitro release kinetics of FGF-1 when impregnated into increased cellular proliferation in collateral arteries after
ePTFE grafts using fibrin glue suspensions. Society for administration of vascular endothelilial growth factor in
Biomaterials Transactions 1995; 18:104. a rabbit model of lower limb vascular insufficiency. Am J
96. Burgess WH, Shaheen AM, Ravera M, Jaye M, Donohue Path. 1995; 147(6):1649-60.
PJ, Winkles JA. Possible dissociation of the heparin-bind- 110. Chen HH, Chien CH, Liu HM. Correlation between an-
ing and mitogenic activities of heparin-binding (acidic fi- giogenesis and basic fibroblast growth factor expression
broblast) growth factor-1 from its receptor-binding ac- in experimental brain infarct. Stroke 1994; 25:1651-7.
tivities by site directed mutagenesis of a single lysine resi- 111. Ren JM, Finklestein SP. Time window of infarct reduc-
due. J Cell Biol 1990; 111:2129-38. tion by intravenous basic fibroblast growth factor in fo-
97. Burgess WH, Shaheen AM, Hampton B, Donohue PJ, cal cerebral ischemia. Eur J Pharmacol 1997; 327(1):11-16.
Winkles JA. Structure-function studies of heparin-bind- 112. Fu X, Sheng Z, Wang Y, Ye Y, Xu M, Sun T, Zhou B.
ing (acidic fibroblast) growth factor-1 using site-directed Basic fibroblast growth factor reduces the gut and liver
mutagenesis. J Cell. BioChem 1991; 45:131-38. morphologic and functional injuries after ischemia and
98. Fox D, Kouris G, Blumofe K, Heilizer T, Husak V, Greisler reperfusion. J Trauma 1997; 42(6):1080-5.
HP. Optimizing fluorescent labeling of endothelial cells
Facilitation of Healing
Matrix Degradation
CHAPTER 29
Role of Urokinase-Type Plasminogen Activator (uPA)
in In Vitro Angiogenesis in Fibrin Matrices
Pieter Koolwijk, Victor W.M. van Hinsbergh
Summary
T issue repair-associated angiogenesis, the formation of new blood vessels from existing
ones, usually involves cell invasion into a fibrin structure and the presence of inflamma-
tory cells. In this chapter the role of plasminogen activators and their receptors in the inva-
sion of endothelial cells into a fibrin matrix is described. At the basolateral side of the endo-
thelial cell, the urokinase-type plasminogen activator (uPA) bound to a specific cellular
receptor (uPA receptor) is involved in the proteolytic modulation of matrix proteins and
cell-matrix interaction. Interference in the activity of uPA and the binding of uPA to the
uPA receptor inhibits the in vitro invasion of human microvascular endothelial cells
(HMVEC) and the formation of capillary-like tubular structures of HMVEC in three di-
mensional fibrin matrices, induced by a combination of the cytokine tumor necrosis factor-#
(TNF-#) and the angiogenic factors basic fibroblast growth factor (bFGF) and vascular en-
dothelial growth factor (VEGF).
Introduction
Angiogenesis, the outgrowth of new blood vessels from existing ones, is an essential
process during development, but normally stops when the body becomes adult. In the ab-
sence of injury, overt angiogenesis in adults is limited to the reproductive system of females
(formation of corpus luteum and placenta).1 However, the formation of new blood vessels is
an essential factor in tissue repair (formation and regression of granulation tissue) which is
necessary to restore healthy tissue after wounding and/or inflammation. Furthermore, an-
giogenesis is associated with many pathological conditions, such as chronic inflammation
including rheumatoid arthritis,2 malignancies,3 and retinopathy caused by metabolic
dysregulation in particular diabetes.4 Common in these conditions is that angiogenesis is
accompanied by vascular leakage,5 the occurrence of inflammatory cells,6 and the presence
of fibrin.7,8 These latter factors are absent in angiogenesis during embryonic development.
Therefore, the possibility exists that “developmental angiogenesis” and adult “repair-associ-
ated or pathologic angiogenesis” are two processes with many identical features, but with
different properties with respect to their regulation.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
314 Tissue Engineering of Prosthetic Vascular Grafts
acts as a barrier preventing further blood loss, but also pro- Inhibitors
vides a structure in which new microvessels can infiltrate The activities of the proteases of the fibrinolytic sys-
during wound healing. Proper timing of the outgrowth of tem are controlled by potent inhibitors which are members
microvessels as well as the subsequent (partial) disappear- of the serine protease inhibitor (serpin) superfamily. Plas-
ance of these vessels is essential to ensure adequate wound min, if not bound to fibrin, is instantaneously inhibited by
12
healing and to prevent the formation of scar tissue. An im- #2-antiplasmin. Because this interaction is facilitated by
portant role in the invasion of a fibrin matrix by endothelial the lysine binding domain of plasmin, it is attenuated when
cells is played by the plasminogen activator/plasmin system, plasmin is bound to fibrin. The predominant regulators of
which lyses fibrin at the basolateral side of the cell. How- tPA and uPA activities are PAI-1 and PAI-2.13,14 PAI-1 is a
ever, the endothelium must respond differently to fibrin 50 kDa glycoprotein present in blood platelets and synthe-
depending on whether the fibrin is present at its luminal or sized by endothelial cells, smooth muscle cells and many
abluminal side. If fibrin is generated at the luminal side, the other cell types in culture.15,16 PAI activity in human plasma
vessel may be occluded, which will cause serious damage of is normally exclusively PAI-1. PAI-1 binds to vitronectin,
the distal tissues. To prevent this the endothelium is able to which stabilizes its inhibitory activity. PAI-1 is the main if
instantaneously increase the fibrinolytic activity in the blood not the sole inhibitor of PAs synthesized by endothelial cells,
compartment. If, however, this would lead to strong systemic vascular smooth muscle cells and hepatocytes, whereas PAI-2
fibrinolytic activity, recurrent bleeding might occur. There- is produced by monocytes/macrophages, placental tropho-
fore, fibrinolysis must be restricted to a limited distance of blasts and certain tumor cell lines.14 It can be found as a
the endothelium. To fulfill the roles of fibrinolysis in both glycosylated secreted molecule and as an nonglycosylated
the prevention of local intravascular fibrin accumulation, molecule intracellularly.17
and a differently timed contribution in neovascularization
and tissue repair, endothelial cells are equipped with a com- Receptors
plex regulatory system which involves inhibitors, cell polar- Regulation of fibrinolytic activity also occurs by cel-
ity and cellular receptors. After discussing the components lular receptors. These receptors direct the action of PAs and
of the plasminogen activator/plasmin system, we will focus plasmin to focal areas on the cell surface, or are involved in
on the contribution of the urokinase-type plasminogen ac- the clearance of the PAs. High affinity binding sites for plas-
tivator (uPA) in pericellular proteolytic events associated minogen,18-21 tPA21,22 and uPA23-25 are found on various
with cell migration and the invasion of capillary-like tubu- types of cells, including endothelial cells.
lar structures into fibrin matrices. Endothelial cells in vitro bind plasminogen with a
moderate affinity (120-340 nM depending on whether the
Components of the Plasmin/Plasminogen Lys or Glu form of plasminogen is used), but with a high
Activator System capacity (3.9-14 x 105 molecules per cell).19,26 This binding,
which is also observed with many other cell types, is medi-
Proteases ated by the lysine binding sites of kringles 1-3 of the
Plasmin is formed from its zymogen plasminogen by plasmin(ogen) molecule. Because these lysine binding sites
proteolytic activation by plasminogen activators (PAs) are also involved in the interaction of plasmin with
(Fig. 29.1). Two types of mammalian PAs are presently #2-antiplasmin, occuPAtion of lysine binding sites protects
known: tissue-type plasminogen activator (tPA) and uroki- plasmin from instantaneous inhibition by #2-antiplasmin
nase-type plasminogen activator (uPA).9,10 The three serine not only when plasmin is bound to fibrin (see above), but
proteases, plasminogen, tPA and uPA, are synthesized as also when it is bound to the cellular receptors. The nature of
single polypeptide chains, and each of them is converted by the plasminogen receptors is not fully resolved. In addition
specific proteolytic cleavage to a molecule with two polypep- to gangliosides, which directly or indirectly contribute to
tide chains connected by a disulphide bond. The C-termi- the plasminogen binding,27 at least eight proteins have been
nal part of the molecule (the so-called B chain) contains the reported to be involved in plasminogen binding. Among
proteolytically active site, whereas the amino-terminal part them are members of the low density lipoprotein receptor
of the molecule (the A-chain) is built up of domains that family, such as gp330 and LRP; annexin II; a not yet identi-
determine the interaction of the proteases with matrix pro- fied 45 kDa protein; GbIIb/IIIa and #-enolase (see ref. 21
teins and cellular receptors. The proteolytic cleavage of plas- for review). In neural cells plasminogen binding to
minogen and single chain uPA to their respective two chain amphoterin was found. Lipoprotein LpA, which has strong
forms is necessary to disclose the proteolytically active site structural homology with a large part of the plasminogen
and to activate the molecule. The interaction of plasmino- molecule, can compete for plasminogen binding to endo-
gen with fibrin or the cell surface occurs predominantly via thelial cells.20,28 This competition also involves lysine bind-
binding sites in the kringle structures, which recognize lysine ing sites.
residues of proteins, in particular C-terminal lysines. Because Specific binding of tPA to human endothelial cells has
B-type carboxypeptidases remove C-terminal lysine residues been reported.29-31 tPA binds via its growth factor domain
from potential binding sites for plasminogen in fibrin or on with high affinity to annexin II on human endothelial cells
the cell surface, they can act as negative regulators of the in culture.32 Different epitopes of the annexin II molecule
fibrinolytic system.11 are involved in plasminogen and tPA binding. It is conceiv-
able that annexin II, like fibrin, forms a ternary complex
Role of Urokinase-Type Plasminogen Activator (uPA) in In Vitro Angiogenesis in Fibrin Matrices 315
Fig. 29.1. Scheme of the plasminogen activation system. +: activation; -: inhibition. Inhibitors are indicated in italics. PA: plasmi-
nogen activator; tPA: tissue-type PA; tcuPA: two chain urokinase-type PA; scuPA: single chain urokinase-type PA; PAI: PA inhibitor.
with tPA and plasminogen on the endothelial cell surface.21 binds to the uPA receptor and is subsequently converted to
In addition, tPA interacts with matrix-bound PAI-1.31 the proteolytically active two chain uPA. Since the endothe-
Binding of uPA with the cell surface limits plasmino- lial cell also contains plasmin(ogen) receptors, an interplay
gen activation to focal areas such as the focal attachment between receptor-bound uPA and receptor-bound
sites and cellular protrusions involved in cell migration and plasmin(ogen) and plasmin formation is likely to happen.
invasion. Furthermore, uPA interaction with the cell evokes The generated plasmin can degrade a number of matrix pro-
signal transduction and phosphorylation of several pro- teins. In addition, a direct plasmin-independent proteolytic
teins.33,34 A specific uPA receptor has been identified and action of uPA on matrix proteins may also occur.41 Like free
cloned. It is present on many cell types, including endothe- uPA activity, receptor-bound two chain uPA is subject to
lial cells.35 It is a glycosyl phosphatidyl inositol (GPI)-an- inhibition by PAI-1. As a consequence, uPA is only active
chored glycoprotein,25 which binds both single chain uPA over a short period of time. In contrast to receptor-bound
and two chain uPA via their growth factor domains.36 The single chain or noninhibited two chain uPA, the uPA-PAI-1
uPA receptor is heavily glycosylated and belongs to the cys- complex is rapidly internalized together with the uPA re-
teine-rich cell surface proteins. After synthesis it is proteolyti- ceptor,42 followed by the degradation of the uPA-PAI-1 com-
cally processed at its carboxyl-terminus and subsequently plex and the return of the empty uPA receptor to the plasma
anchored in the plasma membrane by a GPI group.37 It com- membrane. Internalization of the GPI-linked uPA receptor
prises three domains, which are structurally homologous to probably occurs after interaction with another receptor, such
snake venom #-toxins.25 The uPA receptor has been found as the #2-macroglobulin/low density lipoprotein receptor-
in focal attachment sites, where integrin-matrix interactions related protein (LRP)43 or the VLDL receptor.44 VLDL re-
occur, and in cell-cell contact areas.38,39 Human endothelial ceptors were demonstrated on capillary and arteriolar en-
cells in vitro contain about 140,000 uPA receptors per cell.40 dothelial cells in vivo.45 Recently it has been found that the
The uPA receptor both acts as a site for focal pericel- uPA receptor may have an additional role. The uPA receptor
lular proteolysis by uPA and is involved in the clearance of occupied by uPA interacts avidly with vitronectin.46 Hence,
the uPA-PAI-1 complex. Upon secretion, single chain uPA cell adhesion may represent an additional function of the
uPA receptor.
316 Tissue Engineering of Prosthetic Vascular Grafts
The LRP and LRP-like proteins on liver hepatocytes blast growth factor (bFGF, aFGF) and vascular endothelial
are involved in the clearance of plasmin-#2-antiplasmin, PA- growth factor (VEGF).65-67 The number of uPA receptors
PAI-1 complexes47-49 and probably free PAs from the circu- on human and bovine endothelial cells is also enhanced by
lation. In addition, tPA can also be cleared by mannose re- the activation of protein kinase c and by the elevation of the
ceptors present on macrophages and liver endothelial cells50 cellular cAMP concentration.68,69 Preliminary experiments
and by #-fucose receptors on hepatocytes.32 in our laboratory have demonstrated that the induction of
uPA receptor by VEGF in human endothelial cells is inhib-
The Regulation of Plasminogen Activation ited by protein kinase c inhibition. The effects of bFGF and
and Pericellular Proteolysis by Inflammatory VEGF on the induction of uPA receptor in human endothe-
and Angiogenic Mediators lial cells are regulated independently of their effects on cell
proliferation. Similarly, Presta et al70 have shown that the
Regulation of Proteases And Inhibitors induction of uPA by bFGF in bovine endothelial cells pro-
The primary cytokines interleukin-1 (IL-1) and tu- ceeds independently from the stimulation of mitogenesis by
mor necrosis factor-# (TNF-#) exert many effects on the this growth factor.
vascular endothelium. Their most prominent feature is the In addition to FGFs and VEGF, which induce mitoge-
induction or increase of the transcription of many genes, nesis, TNF-# also can induce angiogenesis, but this occurs
such as the leukocyte adhesion molecules E-selectin, without stimulation of cell proliferation.71,72 TNF-# increases
VCAM-1 and ICAM-1, cyclooxygenase-2, and a number of uPA receptor levels in human microvascular endothelial
proteases and protease inhibitors. The inflammatory me- cells65 and in monocytes,73 but not in endothelial cells from
diators TNF-#, IL-1 and the bacterial lipopolysaccharide human umbilical vein or aorta.65 However, simultaneous
(LPS) induce the synthesis of uPA in human endothelial cells exposure of the latter cells to TNF-# (which induces uPA
in vitro.51 Whereas uPA is normally not found in endothe- synthesis) to bFGF and VEGF (which enhance the expres-
lial cells in vivo, association of uPA with the endothelium sion of uPA receptors) potently increases cell-bound
was observed in acute appendicitis52 and in rheumatoid ar- uPA activity.
thritis.2 Induction of endothelial uPA by TNF-# in vitro is
associated by an increased degradation of matrix proteins.53 Interaction Between the uPA/Plasmin System
The enhanced secretion of uPA occurs entirely towards the and Matrix-Degrading Metalloproteases
basolateral side of the cell, whereas the secretion of tPA and The observation that TNF-# also increases the pro-
PAI-1 proceeds equally to the luminal and basolateral sides duction of matrix-degrading metalloproteinases (MMPs) by
of the cell.51 The polar secretion of uPA suggests that uPA endothelial cells is consistent with a putative role for TNF-#
may be involved in local remodeling of the basal membrane and IL-1 in inflammation-induced local pericellular pro-
of the cell. uPA activity is controlled in space by interaction teolysis. In human microvascular and vein endothelial cells,
of uPA with its cellular receptor and by the inhibitor PAI-1. TNF-# increases the mRNA levels and the synthesis of in-
TNF-# and IL-1, as well as LPS, also elicit another ef- terstitial collagenase (MMP-1), stromelysin-1 (MMP-3)
fect on the regulation of plasminogen activator production and—if protein kinase c is also activated—gelatinase-B
in endothelial cells. Simultaneous with the increase in uPA, (MMP-9), whereas the mRNAs levels and synthesis of their
these inflammatory mediators markedly increase the pro- physiological inhibitors TIMP-1 and TIMP-2 are not
duction of PAI-1 in endothelial cells in vitro.54-57 This in- changed. 74,75 Furthermore, activation of gelatinase a
duction was also demonstrated at the transcriptional level, (MMP-2) was observed after exposure of the cells to
and was largely inhibited by the isoflavone compound TNF-#.74 Recently, it has been shown that activation of
genistein.58 In vivo, administration of TNF-#, IL-1 or LPS gelatinase a depends on the activity of membrane-type MMP
causes an increase in PAI-1 concentration in the circulation. (MT-MMP), 76,77 and that MMP-2 interacts with #v!3
After infusion of LPS in animals, PAI-1 mRNA increased in integrin.78 Interestingly, secretion of gelatinases by bovine
vascularized tissues and PAI-1 mRNA was elevated in the endothelial cells occurs predominantly towards the
endothelium of various organs.59,60 The increase of PAI-1 basolateral side of the cells,79 similarly to the TNF-#-induced
induced by inflammatory mediators may represent a pro- production of uPA.51 A role of MMPs in endothelial cell-
tective mechanism of the cell against uncontrolled uPA matrix remodeling has indeed been shown in a three dimen-
activity. sional collagen matrix in vitro.80 Furthermore, MMPs have
The effect of the angiogenic growth factors basic and been detected in vivo in proliferating endothelial cells dur-
acidic fibroblast growth factor (bFGF, aFGF) and vascular ing development,81 and in endothelial cells present in ath-
endothelial growth factor (VEGF) on the production of uPA erosclerotic plaques and growing tumors.82-84
by endothelial cells is species dependent. Whereas bFGF and The plasmin-plasminogen activator system and the
VEGF are potent inducers of both uPA in bovine cells,61,62,63 matrix metalloproteinases cooperate in the degradation of
bFGF and VEGF do not enhance uPA production in human extracellular matrix proteins.85 Figure 29.2 depicts the in-
endothelial cells.64,65 PAI-1 production by human endothelial teraction between the two systems. It should be noted, how-
cells is slightly decreased by the addition of bFGF or VEGF.65 ever, that this schematic picture is based on in vitro data,
and that it is still uncertain whether all the depicted steps
Regulation of the uPA Receptor also act in vivo. Nevertheless, uPA and MMP expression
The expression of the uPA receptor is enhanced by frequently coincide in time and location in pathological
angiogenic growth factors including basic and acidic fibro- tissues. If these data are taken together, it will be clear that
Role of Urokinase-Type Plasminogen Activator (uPA) in In Vitro Angiogenesis in Fibrin Matrices 317
activation of endothelial cells by TNF-# affects multiple sites tPA.94,95 These data suggest that uPA and plasminogen have
in the proteolytic cascades involved in the degradation of a role in cell recruitment.
matrix proteins to such an extent that it markedly enhances An involvement of uPA and the uPA receptor in the
the breakdown and remodeling of the endothelial cell migration of bovine endothelial cells has been demonstrated
basal membrane. by several investigators.63,96,97 After the wounding of a mono-
layer of these endothelial cells, the cells that migrate into the
Involvement of uPA and uPA Receptor wounded area express uPA activity96 bound to the uPA re-
in Cell Migration and Realignment ceptor.63 The migration and expression of uPA depends on
of Endothelial Cells the release of bFGF from the wounded area.97 bFGF is a
Concentration of uPA activity at the cellular protru- potent inducer of plasminogen activator activity, in particu-
sions of migrating or invading cells has been frequently ob- lar uPA, in bovine endothelial cells,61,98 but not in human
served.86,87 Blasi88 suggested that a continuous activation and endothelial cells.65 In both species bFGF increases the num-
removal of uPA bound to the receptor could contribute to ber of uPA receptors on endothelial cells.63,65,66
the formation and detachment of focal attachment sites and Receptor-bound uPA was demonstrated in focal ad-
hence to locomotion of the cell. Indeed, migrating and in- hesion sites of fibroblasts89 and endothelial cells.39 It may
vading cells, such as monocytes and tumor cells, express uPA act proteolytically on these structures and hence influence
activity bound to uPA receptors on their cellular protru- cell-matrix interactions and cell migration. After exposure
sions87 and on focal attachment sites.38,89 Receptor-bound of endothelial cells to shear forces induced by fluid flow, the
uPA activity is also thought to be involved in smooth muscle cells realign according to the direction of flow.99 This pro-
and endothelial cell migration and in the formation of new cess is paralleled by a movement of the cellular focal contact
blood vessels (angiogenesis). Inhibition of plasminogen ac- sites of endothelial cells.100 Ponfoort et al101 observed that
tivation interferes with smooth muscle cell migration in the production of uPA by human iliac vein endothelial cells
vitro90,91 and affects smooth muscle migration and prolif- was elevated several fold after exposure of the cells to fluid
eration in vivo.92 In mice lacking uPA, intimal hyperplasia flow. Interestingly, the realignment of iliac vein endothelial
of injured arteries is less pronounced than in wild type or cells according to the direction of flow appeared to be re-
tPA-deficient mice.93 Moreover, PAI-1-deficient mice show lated to cell-bound uPA activity. Anti-uPA antibodies, which
an exacerbated intimal proliferation.93 Animals made defi- inhibited uPA activity, reduced the cellular realignment in-
cient for plasminogen show comparable pathological fea- duced by fluid shear forces.101
tures to those with a combined deficiency of uPA and
Role of the Plasmin/Plasminogen Activator cells, and thus inhibits PA activity.61 In addition to bFGF,
System in the Formation of Endothelial Tubes VEGF can also stimulate bovine endothelial cells to form
in a Fibrin Matrix tubular structures. It acts cooperatively with bFGF in
The outgrowth of new blood vessels from existing this induction.113
ones, angiogenesis, is an essential process during develop- Studies in human endothelial cells showed that no
ment, but normally stops when the body becomes adult. The tubular structures are formed when a quiescent monolayer
half life of endothelial cells in the adult body varies between of human microvascular endothelial cells grown on a fibrin
100-10,000 days in normal tissues, whereas it is reduced to matrix is exposed to bFGF or VEGF. However, when bFGF
several days in placenta and tumors.102 With the exception or VEGF are added simultaneously with TNF-#, a large num-
of the female reproductive system, angiogenesis in the adult ber of capillary-like tubular structures are formed
is associated with tissue repair after injury by wounding or (Fig. 29.3).65 The outgrowth of tubular structures requires
inflammation. Repair-associated angiogenesis in the adult uPA activity and is completely reduced by anti-uPA immu-
is usually accompanied by the presence of fibrin and inflam- noglobulins but not by anti-tPA antibodies (Fig. 29.4). It is
matory cells or mediators, in contrast to developmental an- also reduced by inhibiting the interaction of uPA with its
giogenesis in the embryo. Fibrin is a temporary matrix, which receptor. Furthermore, proteolytic activation of plasmino-
is formed after the wounding of a blood vessel and when gen appears to be involved, because the plasmin inhibitor
plasma leaks from blood vessels forming a fibrous exudate, aprotinin largely inhibits the formation of tubular struc-
often seen in areas of inflammation and in tumors.7,103 The tures.65 Recent immunohistochemical studies performed on
fibrin matrix acts as a barrier preventing further blood loss, cross sections of these invading capillary-like tubular struc-
and provides a structure in which new microvessels can in- tures showed an enhanced expression of both uPA and the
filtrate during wound healing. A proper timing of the out- uPAR antigen by invading endothelial cells compared with
growth of microvessels as well as the subsequent (partial) noninvading endothelial cells (Kroon et al; manuscript sub-
disappearance of these vessels is essential to ensure adequate mitted for publication). These data agree with the data on
wound healing and to prevent the formation of scar tissue. bovine endothelial cells, except that in human endothelial
Although essential for the formation of granulation tissue cells a second mediator is required to induce uPA synthesis.
and tissue repair, angiogenesis, once under the control of In animals bFGF and aFGF have been shown to induce
pathological stimuli, can contribute to a number of patho- neovascularization.114,115 Because it is difficult to rule out
logical conditions, such as tumor neovascularization, pan- the involvement of a limited number of leukocytes in these
nus formation in rheumatoid arthritis, and diabetic retin- experiments in vivo, it remains to be elucidated whether these
opathy. Understanding the mechanisms involved in angio- growth factors act in vivo in conjunction with inflamma-
genesis may provide clues to preventing pathological angio- tory mediators or independently of the latter mediators. Min
genesis without seriously impairing tissue repair. A number et al116 have recently shown that prevention of the binding
of studies and reviews have focused on angiogenic factors of uPA to the uPAR by a fusion product of the epidermal
and the formation of capillary-like structures.4,5,108-109 growth factor-like domain of murine uPA and the Fc por-
Among them, Dvorak7 and Colvin8 have pointed to the im- tion of human IgG reduced bFGF-induced angiogenesis in
portance of fibrin in angiogenesis. Furthermore, the work vivo, and the growth of a B16 melanoma in syngeneic mice,
of Polverini and colleagues have demonstrated the involve- indicating that uPA-mediated angiogenesis also occurs in
ment of monocytes and their products in the induction of these in vivo systems.
angiogenesis.6,71,72,110 Because of the specific roles of fibrin Proteolysis of the basement membrane of endothelial
and inflammatory cells and mediators in pathological an- cells and invasion of endothelial cells into the underlying
giogenesis in the adult, we have focused our studies on the matrix are prerequisites for angiogenesis.117 However, not
invasion of human endothelial cells into three dimensional only proteolysis, but also the formation of new cell attach-
fibrin matrices and the role of endothelial plasminogen ac- ment sites, is important for the formation of tubular struc-
tivators in this process. This model resembles recanalization tures. It should be noted that fibrin contains cell binding
of a fibrin clot by invading endothelial cells. This invasion is domains for endothelial cells: a RGD sequence in its #-chains
usually preceded by the infiltration of inflammatory cells, which binds to the vitronectin receptor, i.e., the #v!3
which interact with the vascular structures from which en- integrin;118,119 and another site in the !-chain (residues
dothelial cells subsequently migrate into the fibrin clot.111 5-42),120 which binds to an 130 kDa receptor.121 Brooks et
A three dimensional fibrin matrix model was used by al122,123 and Hammes et al124 have shown that inhibition of
Pepper and Montesano to demonstrate a direct correlation the #v!3 integrin reduces angiogenesis in several in vivo
between the expression of PA activity and the formation of models. Friedlander et al125 recently reported that both #v!3
capillary sprouts by bovine microvascular endothelial cells and #v!5 integrin are involved in growth factor-stimulated
in vitro.106,112 The outgrowth of tubular structures was in- angiogenesis in the rabbit cornea, but via distinct mecha-
creased by bFGF, which increases both uPA activity and uPA nisms. It is of interest to note that bFGF- and TNF-#-in-
receptor in bovine endothelial cells. Interestingly, the extent duced angiogenesis is inhibited by an antibody against the
of tube formation and the diameter of the formed tubes were #v!3 integrin, whereas angiogenesis induced by VEGF or by
reduced by the simultaneous presence of TGF-!.112 The latter a protein kinase c activating phorbol ester required #v!5
is a growth factor which, amongst others, exerts a strong integrin. It remains to be established whether both #v!3- and
enhancement of PAI-1 synthesis in cultured endothelial #v!5-dependent mechanisms are active in the invasion of
endothelial cells into a fibrin matrix. The involvement of
Role of Urokinase-Type Plasminogen Activator (uPA) in In Vitro Angiogenesis in Fibrin Matrices 319
Fig. 29.4. Effect of various inhibitors of the uPA/plasmin system on the bFGF/VEGF/TNF-#-induced formation of tubular struc-
tures of human microvascular endothelial cells in a three dimensional fibrin matrix in vitro.
#v!3 integrin interaction with the fibrin matrix is likely,126 as fibronectin, vitronectin, hyaluronic acid and
while the role of #v!5 integrin, which more selectively inter- thrombospondin, and by the environmental conditions like
acts with vitronectin, has to be evaluated. Irrespective of the the pH and the ionic strength, determines whether new cap-
exact role of fibrin in stimulating angiogenesis, the fibrin illaries are formed or not. The invasion of endothelial cells
structure has important consequences for wound healing, into a fibrin matrix and the formation of capillary-like struc-
and proteolytic modification of fibrin, e.g., by leukocyte tures in the fibrin matrix in vitro is an attractive model to
elastase, or interaction of fibrin with other matrix proteins, study this essence of the angiogenesis process associated with
such as vitronectin and fibronectin, may affect cell invasion tissue repair. A recent paper of Nehls and Herrmann128
and angiogenesis and the success of wound healing. Fur- points to the importance of the fibrin structure in endothe-
thermore, the recent observation that MMP-2 (gelatinase-A) lial cell migration and the formation of tubular structures.
binds to #v!378 further points to the complexity of interac- These authors show that the rigidity of the fibrin gel has a
tions that the #v!3 integrin may have in fibrinous exudates, strong impact on tube formation by bovine endothelial cells
in which the matrix consists of a mixture of fibrin, collagens in response to bFGF and VEGF. In addition, we have found
and other extracellular matrix components. that this was also the case when human microvascular en-
dothelial cells were used (Collen et al, manuscript submit-
Perspective ted for publication). It remains to be determined whether
Delineation of the various cellular pathways that are this effect of the fibrin structure reflects a mechanical bar-
involved in angiogenesis is essential to selective interference rier to movement of cells by a dense fibrin network, or is
with unwanted angiogenesis, such as in rheumatoid arthri- due to an inadequate spacing of cell-binding epitopes in the
tis and tumors, and to stimulation of neovascularization at fibrin network.
sites where it is needed, such as in normal wound healing Nevertheless, these data obtained using in vitro ex-
and collateral formation. periments may be of great value when fibrin gels are used in
The structure of fibrin clot, determined not only by vivo as “temporary” matrix with regard to the initiation of
the concentration of fibrinogen and thrombin present in site-directed formation of new blood vessel structures or
the tissues but also by the presence of matrix molecules such collaterals, as shown by Dvorak et al129 and Fasol et al.130
Role of Urokinase-Type Plasminogen Activator (uPA) in In Vitro Angiogenesis in Fibrin Matrices 321
39. Conforti G, Dominguez-Jimenez C, Rønne E, Høyer- activator mediated proteolysis of extracellular matrix and
Hansen G, Dejana E. Cell-surface plasminogen activation its antagonism by &-interferon. Blood 1992; 79:678-687.
causes a retraction of in vitro cultured human umbilical 54. Colucci M, Paramo JA, Collen D. Generation in plasma
vein endothelial cell monolayer. Blood 1994; 83:994-1005. of a fast-acting inhibitor of plasminogen activator in re-
40. Haddock RC, Spell ML, Baker CD III, Grammer JR, Parks sponse to endotoxin stimulation. J Clin Invest 1985;
JM, Speidel M, Booyse FM. Urokinase binding and re- 75:818-824.
ceptor identification in cultured endothelial cells. J Biol 55. Emeis JJ, Kooistra T. Interleukin-1 and lipopolysaccha-
Chem 1991; 266:21466-21473. ride induce a fast-acting inhibitor of tissue-type plasmi-
41. Quigley JP, Gold LI, Schwimmer R, Sullivan LM. Limited nogen activator in vivo and in cultured endothelial cells.
cleavage of cellular fibronectin by plasmin activator puri- J Exp Med 1986; 163:1260-1266.
fied from transformed cells. Proc Natl Acad Sci USA 1987; 56. Schleef RR, Bevilacqua MJ, Sawdey M, Gimbrone MA,
84:2776-2780. Loskutoff DJ. Cytokine activation of vascular endothelium.
42. Olson D, Pöllänen J, Høyer-Hansen G, Rønne E, Effect on tissue-type plasminogen activator and type 1
Sakaguchi K, Wun T-C, Appella E, Danø K, Blasi F. In- plasminogen activator inhibitor. J Biol Chem 1988;
ternalization of the urokinase-plasminogen activator in- 263:5797-5803.
hibitor type-1 complex is mediated by the urokinase re- 57. Van Hinsbergh VWM, Kooistra T, Van den Berg EA,
ceptor. J Biol Chem 1992; 267:9129-9133. Princen HMG, Fiers W, Emeis JJ. Tumor necrosis factor
43. Nykjœr A, Petersen CM, Møller B, Jensen PH, Moestrup increases the production of plasminogen activator inhibi-
SK, Holtet TL, Etzerodt M, Thogersen HC, Munch M, tor in human endothelial cells in vitro and in rats in vivo.
Andreasen PA, Gliemann J. Purified #2-macroglobulin Blood 1988; 72:1467-1473.
receptor/LDL receptor-related protein binds urokinase 58. Van Hinsbergh VWM, Vermeer M, Koolwijk P,
activator inhibitor type-1 complex—Evidence that the #2- Grimbergen J, Kooistra T. Genistein reduces tumor ne-
macroglobulin receptor mediates cellular degradation of crosis factor #-induced plasminogen activator inhibitor-1
urokinase receptor-bound complexes. J Biol Chem 1992; transcription but not urokinase expression in human en-
267:14543-14546. dothelial cells. Blood 1994; 84:2984-2991.
44. Heegaard CW, Wiborg Simonsen AC, Oka K, Kjøller L, 59. Quax PHA, Van den Hoogen CR, Verheijen JH, Padro T,
Christensen A, Madsen B, Ellgaard L, Chan L, Andreasen Zeheb R, Gelehrter TD, van Berkel TJC, Kuiper J, Emeis
PA. Very low density lipoprotein receptor binds and medi- JJ. Endotoxin induction of plasminogen activator and plas-
ates endocytosis of urokinase-type plasminogen activator- minogen activator inhibitor type 1 mRNA in rat tissues
type-1 plasminogen activator inhibitor complex. J Biol in vivo. J Biol Chem 1990; 265:15560-15563.
Chem 1995; 270:20855-20861. 60. Keeton M, Eguchi Y, Swadey M, Ahn C, Loskutoff D.
45. Wyne KL, Pathak RK, Seabra MC, Hobbs HH. Expres- Cellular localization of type 1 plasminogen activator in-
sion of the VLDL receptor in endothelial cells. Arterioscl hibitor messenger RNA and protein in murine renal tis-
Thromb Vasc Biol 1996; 16:407-415. sue. Am J Pathol 1993; 142:59-70.
46. Wei Y, Waltz D, Rao N, Drummond R, Rosenberg S, 61. Saksela OD, Moscatelli D, Rifkin D. The opposing effects
Chapman H. Identification of the urokinase receptor as of basic fibroblast growth factor and transforming growth
cell adhesion receptor for vitronectin. J Biol Chem 1994; factor beta on the regulation of plasminogen activator
269:32380-32388. activity in capillary endothelial cells. J Cell Biol 1987;
47. Orth K, Madison EL, Gething M-J, Sambrook JF, Herz J. 105:957-963.
Complexes of tissue-type plasminogen activator and its 62. Pepper MS, Ferrara N, Orci L, Montesano R. Vascular
serpin inhibitor plasminogen-activator inhibitor type-1 are endothelial growth factor (VEGF) induces plasminogen
internalized by means of the low density lipoprotein re- activators and plasminogen activator inhibitor-1 in mi-
ceptor-related protein/#2-macroglobulin receptor. Proc crovascular endothelial cells. Biochem Biophys Res
Natl Acad Sci USA 1992; 89:7422-7426. Commun 1991; 181:902-906.
48. Bu G, Williams S, Strickland DK, Schwartz AL. Low den- 63. Pepper MS, Sappino A-P, Stocklin R, Montesano R, Orci
sity lipoprotein receptor-related protein/#2-macroglobu- L, Vassalli J-D. Upregulation of urokinase receptor ex-
lin receptor is an hepatic receptor for tissue-type plasmi- pression on migrating endothelial cells. J Cell Biol 1993;
nogen activator. Proc Natl Acad Sci USA 1992; 122:673-684.
89:7427-7431. 64. Bikfalvi A, Sauzeau C, Moukadiri H, Maclouf J, Busso N,
49. Bu G, Warshawsky I, Schwartz AL. Cellular receptors for Bryckaert N, Plouet J, Tobelem G. Interaction of
the plasminogen activators. Blood 1994; 83:3427-3436. vasculotropin/vascular endothelial cell growth factor with
50. Otter M, Barrett-Bergshoef MM, Rijken DC. Binding of human umbilical vein endothelial cells—Binding, inter-
tissue-type plasminogen activator by the mannose recep- nalization, degradation, and biological effects. J Cell
tor. J Biol Chem 1991; 266:13931-13935. Physiol 1991; 149:50-59.
51. Van Hinsbergh VWM, van den Berg EA, Fiers W, 65. Koolwijk P, van Erck MGM, de Vree WJA, Vermeer MA,
Dooijewaard G. Tumor necrosis factor induces the pro- Weich HA, Hanemaaijer R, van Hinsbergh VWM. Coop-
duction of urokinase-type plasminogen activator by hu- erative effect of TNF#, bFGF and VEGF on the forma-
man endothelial cells. Blood 1990; 75:1991-1998. tion of tubular structures of human microvascular endot-
52. Grøndahl-Hansen J, Kirkeby L, Ralfkiœr E, Kristensen P, helial cells in a fibrin matrix. Role of urokinase activity. J
Lund LR, Danø K. Urokinase-type plasminogen activator Cell Biol 1996; 6:1177-1188.
in endothelial cells during acute inflammtion of the ap- 66. Mignatti P, Mazzieri R, Rifkin DB. Expression of the
pendix. Am J Pathol 1989; 135:631-636. urokinase receptor in vascular endothelial cells is stimu-
53. Niedbala MJ, Stein-Picarella M. Tumor necrosis factor lated by basic fibroblast growth factor. J Cell Biol 1991;
induction of endothelial cell urokinase-type plasminogen 113:1193-1201.
Role of Urokinase-Type Plasminogen Activator (uPA) in In Vitro Angiogenesis in Fibrin Matrices 323
67. Mandriota S, Seghezzi G, Vassalli J-D, Ferrara N, Wasi S, 81. Karelina TV, Goldberg GI, Eisen AZ. Matrix
Mazzieri R, Mignatti P, Pepper M. Vascular endothelial metalloproteinases in blood vessel development in human
growth factor increases urokinase receptor expression in fetal skin and in cutaneous tumors. J Invest Dermatol
vascular endothelial cells. J Biol Chem 1995; 1995; 105:411-417.
270:9709-9716. 82. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased ex-
68. Langer DJ, Kuo A, Kariko K, Ahuja M, Klugherz BD, pression of matrix metalloproteinases and matrix degrad-
Ivanics KM, Hoxie JA, Williams WV, Liang BT, Cines DB, ing activity in vulnerable regions of human atheroscle-
Barnathan ES. Regulation of the endothelial cell uroki- rotic plaques. J Clin Invest 1994; 94:2493-2503.
nase-type plasminogen activator receptor—Evidence for 83. Nikkari ST, O’Brien KD, Ferguson M, Hatsukami T,
cyclic AMP-dependent and protein kinase-C dependent Welgus HG, Alpers CE, Clowes AW. Interstitial collage-
pathways. Circ Res 1993; 72:330-340. nase (MMP-1) expression in human carotid atheroscle-
69. van Hinsbergh VWM. Impact of endothelial activation on rosis. Circulation 1995; 92:1393-1398.
fibrinolysis and local proteolysis in tissue repair. Ann NY 84. Rao JS, Yamamoto M, Mohaman S, Gokaslan ZL, Stetler-
Acad Sci 1992; 667:151-162. Stevenson WG, Roa VH, Liotta LA, Nicolson Gl, Sawaya
70. Presta M, Maier JAM, Ragnotti G. The mitogenic signal- RE. Expression and localization of 92 kDa type IV colla-
ling pathway but not the plasminogen activator-inducing genase genatinase B (MMP-9) in human gliomas. Clin Exp
pathway of basic fibroblast growth factor is mediated Metastasis 1996; 14:12-18.
through protein kinase c in fetal bovine aortic endothe- 85. Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer me-
lial cells. J Cell Biol 1989; 109:1877-1884. tastasis and angiogenesis—An imbalance of positive and
71. Leibovich SJ, Polverini PJ, Shepard HM, Wiseman DM, negative regulation. Cell 1991; 64:327-336.
Shively V, Nuseir N. Macrophage-induced angiogenesis is 86. Danø K, Andreasen PA, Grøndahl-Hansen J, Kristensen
mediated by tumour necrosis factor- # . Nature 1987; P, Nielsen LS, Skriver L. Plasminogen activators in tissue
329:630-632. degradation and cancer. Adv Cancer Res 1985; 44:139-264.
72. Fràter-Schröder M, Risau W, Hallman R, Gautschi P, 87. Estreicher A, Mühlhauser J, Carpentier J-L, Orci L, Vassalli
Böhlen P. Tumor necrosis factor type #, a potent inhibi- J-D. The receptor for urokinase type plasminogen polar-
tor of endothelial cell growth in vitro, is angiogenic in izes expression of the protease to the leading edge of mi-
vivo. Proc Natl Acad Sci USA 1987; 84:5277-5281. grating monocytes and promotes degradation of enzyme
73. Kirchheimer JC, Nong Y, Remold HG. IFN-&, tumor ne- inhibitor complexes. J Cell Biol 1990; 111:783-792.
crosis factor-#, and urokinase regulate the expression of 88. Blasi F. Urokinase and urokinase receptor—A paracrine/
urokinase receptors on human monocytes. J Immunol autocrine system regulating cell migration and invasive-
1998; 141:4229-4234. ness. Bioessays 1993; 15:105-111.
74. Hanemaaijer R, Koolwijk P, Leclercq L, De Vree WJA, 89. Hébert CA, Baker JB. Linkage of extracellular plasmino-
Van Hinsbergh VWM. Regulation of matrix gen activator to fibroblast cytoskeleton: Colocalization of
metalloproteinase expression in human vein and mi- cell surface urokinase with vinculin. J Cell Biol 1988;
crovascular endothelial cells—Effects of tumour necrosis 105:1241-1247.
factor-#, interleukin-1 and phorbol ester. Biochem J 1993; 90. Schleef RR, Birdwell CR. The effect of proteases on en-
296:803-809. dothelial cell migration in vitro. Exp Cell Res 1982;
75. Cornelius LA, Nehring LC, Roby JD, Parks WC, Welgus 141:503-508.
HG. Human dermal microvascular endothelial cells pro- 91. Quax PHA, Koolwijk P, Verheijen JHM, Van Hinsbergh
duce matrix metalloproteinases in response to angiogenic VWM. The role of plasminogen activators in vascular
factors and migration. J Invest Dermatol 1995; pericellular proteolysis. In: van Hinsbergh VWM, ed. Vas-
105:170-176. cular Control of Hemostasis. Amsterdam: Harwood Aca-
76. Sato H, Takin T, Okada Y, Cao J, Shinagawa A, demic Publishers, 1996:227-245.
Yamamoto E, Seiki M. A matrix metalloproteinase ex- 92. Clowes AW, Clowes MM, Au YPT, Reidy MA, Belin D.
pressed in the surface of invasive tumour cells. Nature Smooth muscle cells express urokinase during mitogen-
1994; 370:61-65. esis and tissue-type plasminogen activator during migra-
77. Foda HD, George S, Conner C, Drews M, Tomkins DC, tion in injured rat carotid artery. Circ Res 1990; 67:61-67.
Zucker S. Activation of human umbilical vein endothelial 93. Carmeliet P, Collen D. Evaluation of the plasminogen/
cell progelatinase a by phorbol myristate: A protein ki- plasmin system in transgenic mice. Fibrinolysis 1994;
nase c-dependent mechanism involving a membrane-type 8:269-276.
matrix metalloproteinase. Lab Invest 1996; 74:538-545. 94. Ploplis VA, Carmeliet P, Vazirzadeh S, Van Vlaenderen I,
78. Brooks PC, Strömblad S, Sanders LC, von Schalscha TL, Moons L, Plow EF, Collen D. Effects of disruption of the
Aimes RT, Stetler-Stevenson WG, Quigley JP, Cheresh DA. plasminogen gene on thrombosis, growth, and health in
Localization of matrix metalloproteinase MMP-2 to the mice. Circulation 1995; 92:2585-2593.
surface of invasive cells by interaction with integrin #v!3. 95. Bugge TH, Suh TT, Flick MJ, Daugherty CC, Rømer J,
Cell 1996; 85:683-693. Solberg H, Ellis V, Danø K, Degen JJ. The receptor for
79. Unemori EN, Bouhana KS, Werb Z. Vectorial secretion urokinase-type plasminogen activator is not essential for
of extracellular matrix proteins, matrix-degrading protein- mouse development or fertility. JBiol Chem 1995;
ases, and tissue inhibitor of metalloproteinases by endot- 270:16886-16894.
helial cells. J Biol Chem 1990; 265:445-451. 96. Pepper MS, Vassalli J-D, Montesano R, Orci L. Uroki-
80. Fisher C, Gilbertson-Beadling S, Powers EA, Petzold G, nase-type plasminogen activator is induced in migrating
Poorman R, Mitchell MA. Interstitial collagenase is re- capillary endothelial cells. J Cell Biol 1987; 105:2535-2541.
quired for angiogenesis in vitro. Dev Biol 1994; 97. Sato Y, Rifkin DB. Autocrine activities of basic fibroblast
162:499-510. growth factor: Regulation of endothelial cell movement,
324 Tissue Engineering of Prosthetic Vascular Grafts
plasminogen activator synthesis, and DNA synthesis. J Cell 114. Thompson JA, Anderson KD, DiPietro JM, Zwiebel JA,
Biol 1988; 107:1199-1205. Zametta M, Anderson WF, Maciag T. Site-directed
98. Gualandris A, Presta M. Transcriptional and posttranscrip- neovessel formation in vivo. Science 1988; 241:1349-1352.
tional regulation of urokinase-type plasminogen activator 115. Broadley KN, Aquino AM, Woodward SC, Buckley-
expression in endothelial cells by basic fibroblast growth Sturrock A, Sato Y, Rifkin D, Davidson JM. Monospe-
factor. J Cell Physiol 1995; 162:400-409. cific antibodies implicate basic fibroblast growth factor in
99. Franke RP, Gräfe M, Schnittler H, Seiffge D, Mittermayer normal wound repair. Lab Invest 1989; 61:571-575.
C, Drenckhahn D. Induction of human vascular endot- 116. Min HY, Doyle LV, Vitt CR, Zandonella CL, Stratton-
helial stress fibres by fluid shear stress. Nature 1984; Thomas JR, Shuman MA, Rosenberg S. Urokinase recep-
307:648-649. tor antagonists inhibit angiogenesis and primary tumor
100. Davies PF, Robotewsky A., Griem ML. Quantitative stud- growth in syngeneic mice. Cancer Res 1996; 56:2428-2433
ies of endothelial adhesions: Directional modeling of fo- 117. Ausprunk D, Folkman J. Migration and proliferation of
cal adhesion sites in response to flow forces. J Clin Invest endothelial cells in preformed and newly formed blood
1994; 93:2031-2038. vessels during tumor angiogenesis. Microvasc Res 1977;
101. Ponfoort ED, van Bockel JH, van Hinsbergh VWM. Shear 14:52-65.
forces induce the synthesis of urokinase-type plasmino- 118. Dejana E, Lampugnani MG, Giorgi M, Gaboli M,
gen activator in human iliac vein endothelial cells in vitro. Marchisio PC. Fibrinogen induces endothelial cell adhe-
Circulation 1995; 92(suppl.):3016. sion and spreading via the release of endogenous matrix
102. Hobson B, Denekamp J. Endothelial proliferation in proteins and the recruitment of more than one integrin
tumours and normal tissues: Continuous labelling stud- receptor. Blood 1990; 75:1509-1517.
ies. Br J Cancer 1984; 49:405-413. 119. Thiagarajan P, Rippon AJ, Farrell DH. Alternative adhe-
103. Dvorak HF, Nagy JA, Berse B, Brown LF, Yeo K-T, Yeo sion sites in human fibrinogen for vascular endothelial
T-K, Dvorak AM, Van De Water L, Sioussat TM, Senger cells. Biochemistry 1996; 35:4169-4175.
DR. Vascular permeability factor, fibrin, and the patho- 120. Bunce LA, Sporn LA, Francis CW. Endothelial cell spread-
genesis of tumor stroma formation. Ann NY Acad Sci ing on fibrin requires fibrinopeptide B cleavage and amino
1992; 667:101-111. acid residues 15-42 of the !-chain. J Clin Invest 1992;
104. Folkman J. Tumor angiogenesis. In: Mendelsohn J, 89:842-850.
Howley PM, Israel MA, Liotta LA, eds. The Molecular 121. Erban JK, Wagner DD. A 130-kDa protein on endothe-
Basis of Cancer. Philadelphia: WB Saunders Comp., lial cells binds to amino acids 15-42 of the B! chain of
1995:206-232. fibrinogen. J Biol Chem 1992; 267:2451-2458.
105. Burgess WH, Maciag T. The heparin-binding (fibroblast) 122. Brooks PC, Clark RA, Cheresh DA. Requirement of vas-
growth factor family of proteins. Annu Rev Biochem 1989; cular integrin # v ! 3 for angiogenesis. Science 1994;
58:575-606. 264:569-571.
106. Montesano R. Regulation of angiogenesis in vitro. Euro- 123. Brooks PC, Montgomery AM, Rosenfeld M., Reisfeld RA,
pean J Clin Invest 1992; 22:504-515. Hu T, Klier G, Cheresh DA. Intergrin #v!3 antagonists
107. Fong G-H, Rossant J, Gertsenstein M, Breitman ML. Role promote tumor regression by inducing apoptosis of an-
of the flt-1 receptor tyrosine kinase in regulating the as- giogenic blood vessels. Cell 1994; 79:1157-1164.
sembly of vascular endothelium. Nature 1995; 376:66-70. 124. Hammes HP, Brownlee M, Jonczyk A, Sutter A, Preissner
108. Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu K. Subcutaneous injection of a cyclic peptide antagonist
X-F, Breitman ML, Schuh AC. Failure of blood-island of vitronectin receptor-type integrins inhibits retinal
formation and vasculogenesis in Flk-1-deficient mice. Na- neovascularization. Nature Medicine 1996; 2:529-533.
ture 1995; 376:62-66. 125. Friedlander M, Brooks PC, Shaffer RW, Kincais CM,
109. Sato TN, Tozawa Y, Deutsch U, Wolburg-Buchholz K, Varner JA, Cheresh DA. Definition of two angiogenic
Fujiwara Y, Gendron-Maguire M, Gridley T, Wolburg H, pathways by distinct #v integrins. Science 1995; 270:1500-
Risau W, Qin Y. Distinct roles of the receptor tyrosine 1502.
kinases tie-1 and tie-2 in blood vessel formation. Nature 126. Chang M-C, Wang B-R, Huang T-F. Characterization of
1995; 376:70-74. endothelial cell differential attachment to fibrin and fi-
110. Koch AE, Polverini PJ, Kunkel SL, Harlow LA, DiPietro brinogen and its inhibition by Arg-Gly-Asp-containing
LA, Elner VM, Elner SG, Strieter RM. Interleukin-8 as a peptides. Thromb Haemostas 1995; 74:764-769.
macrophage-derived mediator of angiogenesis. Science 127. Pepper MS, Vassalli J-D, Wilks JW, Schweigerer L, Orci
1991; 258:1798-1801. L, Montesano R. Modulation of bovine microvascular
111. Kwaan HC. Tissue fibrinolytic activity studied by a his- endothelial cell proteolytic properties of inhibitors of an-
tochemical method, Fed Proc 1966; 25:52-56. giogenesis. J Cell Biochem 1994; 55:419-434.
112. Pepper MS, Belin D, Montesano R, Orci L, Vassalli J. 128. Nehls V, Herrmann R. The configuration of fibrin clots
Transformtin growth factor-!-1 modulates basic fibroblast determines capillary morphogenesis and endothelial cell
growth factor induced proteolytic and angiogenic prop- migration. Microvascular Res 1996; 51:347-364.
erties of endothelial cells in vitro. J Cell Biol 1990; 129. Dvorak HF, Harvey S, Estrella P, Brown LF, McDonagh
111:743-755. J, Dvorak AM. Fibrin containing gels induce angiogen-
113. Pepper MS, Ferrara N, Orci L, Montesano R. Potent syn- esis. Implications for tumor stroma generation and wound
ergism between vascular endothelial growth factor and ba- healing. Lab Invest 1987; 57:673-686.
sic fibroblast growth factor in the induction of angiogen- 130. Fasol R, Schumacher B, Schlaudraff K, Hauenstein KH,
esis in vitro. Biochem Biophys. Res Commun 1992; Seitelberger R. Experimental use of a modified fibrin glue
189:824-831. to induce site-directed angiogenesis from the aorta to the
heart. J Thoracic Cardiovasc Surg 1994; 107:1432-1439.
Facilitation of Healing
Matrix Modulation
CHAPTER 30
How Does Extracellular Matrix Control
Capillary Morphogenesis?
Robert B. Vernon, E. Helene Sage
Angiogenesis: An Introduction
T he blood vascular system has evolved a significant capacity for change. During embry-
onic and fetal life, the vasculature increases in quantity and complexity to serve develop-
ing tissues and organs. Vasculature of the adult is, in general, quiescent, yet it retains a capac-
ity for growth, remodeling, and regression in response to the menstrual cycle, placentation,
changes in adiposity, wound repair, and inflammation. During these processes, growth of
new blood vessels is regulated and eventually ceases. In contrast, certain conditions such as
tumor growth, diabetic retinopathies, arthritis, and psoriasis involve excessive proliferation
of blood vessels that contributes directly to the pathological state.1 Therefore, an under-
standing of the principles and mechanisms that direct the assembly new blood vessels, and
the processes that start and stop vascular growth, are central to the development of agents
and strategies to control vascularization in disease.2
Tissue engineering involves the fabrication of tissue or organ-like implants comprised
of living cells in contact with a supportive matrix. Like cells of natural tissues and organs,
cells associated with an implant must interface with the circulatory system for reception of
oxygen and nutrients and removal of waste products. When implants are membranous, the
metabolic needs of the resident cells can be met by simple diffusive exchanges with sur-
rounding tissue fluids. However, a direct connection between the implant and the vascular
system may be necessary when diffusion distances are long as a consequence of thickness or
bulk of the implant. Moreover, in certain cases, resident cell types might require access to
circulating blood so that they can sense and respond rapidly to changes in body chemistry
and distribute their biosynthetic products (e.g., hormones) rapidly throughout the body.
Exchanges between cells and blood in vivo are most efficient across blood vessels with small
diameters and thin walls (e.g., capillaries); therefore, it would be desirable for implants to be
invested by a microvasculature that is connected to the blood supply of the host via large-
caliber arteries and veins. Artificial capillaries made from nonliving, semipermeable materi-
als are likely to be occluded by thrombi and rendered impermeable by precipitated plasma
proteins within a short period of time after exposure to flowing blood. Thus, it is likely that
long term function of implants will require a microvasculature derived from the host. This
microvasculature would be derived from the growth of extant blood vessels into the im-
plant, a process termed neovascularization, that is typical of the vascularization of tumors.
However, unlike the vasculature of a tumor, which grows and remodels continuously as the
tumor grows, the vascular supply of an implant of finite mass must stop growing once a
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
326 Tissue Engineering of Prosthetic Vascular Grafts
level of neovascularization sufficient to sustain the resident 1. Migration—ECs must penetrate and move through
cell population has been achieved. Excessive and unceasing interstitial ECM;
vascularization of the implant must be prevented, since an 2. Cellular alignment—Unlike a migrating epithelium
overexpression of some of the processes that mediate vas- in which the cells move as a sheet, migrating ECs
cular growth (e.g., proteolysis or forces of cellular traction, must follow one another in a “head-to-tail” orienta-
which are discussed below) could compromise the function tion for nascent vessels to have a tubular form;
of the implant or destroy it altogether. Thus, the develop- 3. Cell division—ECs must increase in number to
ment of methods to control vascular growth will be an im- lengthen the nascent vessel;
portant goal for tissue engineers. 4. Cell-cell adhesion—Migratory ECs must at some
The sprouting of new blood vessels from extant vas- point make firm connections with one another to
culature, termed angiogenesis, is the principal form of vas- form a mechanically-integrated structure;
cular growth in the adult. Sprouts arise from endothelial cells 5. Formation of a lumen—Individual ECs must assume
(ECs) that line the lumens of capillaries and postcapillary a curved shape to enclose a tubular space. Moreover,
venules—the smallest, most delicate branches of the vascu- adjacent cells must join their individual tubular
lar system where ECs are not confined by sheaths of smooth spaces to form a continuous lumen;
muscle or fibrous adventitia. The smallest capillaries are 6. Branching and anastomosis—ECs must send off
comprised of ECs alone and are capable of generating new branches from the main sprout. These branches must
capillary networks in the absence of other cell types. Al- find branches from other sprouts and fuse with them
though ECs contain all of the genetic information required to form anastomoses (also referred to as vascular
for morphogenesis, their potential to organize into capillar- loops or arcades).
ies is realized only by interaction with a noncellular entity— It is generally appreciated that genes regulate the ac-
the extracellular matrix (ECM). This review will discuss ways tivities of cells. Therefore, it has been proposed that the be-
in which ECM mediates and modulates the behaviors of ECs haviors listed above are mediated and regulated by a spe-
that lead to the development of a capillary bed. Much of cific “angiogenic program” or cascade that orchestrates the
what we will relate is speculative, because mechanistic stud- expression of EC gene products at specific times and in spe-
ies of the effects of ECM on vascular morphogenesis are in cific places.5 This program is, in turn, controlled by a vari-
their infancy. ety of soluble extracellular molecules (“growth factors”) se-
creted by target tissues, organs, and tumors.6,7 The role of
The Process of Angiogenesis growth factors in angiogenesis has been intensively studied:
Angiogenesis is a complex, multistep process initiated Much is known about the sources and structure of growth
in response to specific signals (e.g., diffusible, angiogenic factors, receptors for growth factors on the surfaces of ECs,
proteins) that emanate from sites of potential vasculariza- and signaling pathways within EC cytoplasm that are acti-
tion. The stages of angiogenesis induced by solid tumors have vated by growth factors. Moreover, the chemotaxis of EC
been described3,4 and can be summarized as follows: sprouts in response to gradients of growth factors has been
1. Upon receipt of an angiogenic stimulus, ECs of the analyzed in detail.8 What is not understood, however, is how
parent vessel degrade and penetrate the basement regulation of expression of EC genes by growth factors speci-
membrane that invests them, and subsequently mi- fies the actual construction of a capillary. Somehow, geneti-
grate into the surrounding interstitial ECM as a cally programmed, quantitative changes in the synthesis of
knoblike or cone-shaped vascular sprout; specific proteins by individual ECs are translated into spa-
2. The invading ECs assume bipolar shapes and align tial and vectorial information that directs the cooperative
behind one another in a follow-the-leader fashion; assembly of ECs into complex, three dimensional structures
3. ECs at the base or the middle of the sprout begin to (Fig. 30.1). Studies of vascular development indicate that
divide and add to the population of migrating cells; ECM plays a critical role in this translation. We will illus-
4. ECs organize into solid cords, which subsequently trate this concept by reviewing the development and char-
develop lumens; acteristics of the most important models of angiogenesis—
5. Neighboring sprouts move toward one another and an exercise that will reveal the special relationship between
fuse to form vascular anastomoses; ECs and ECM.
6. Circulatory flow begins in the vascular anastomoses;
7. Pericytes emerge along the length of capillary Models of Angiogenesis:
sprouts, and a new basement membrane is The Essential Role of ECM
synthesized.
It is likely that the above mentioned steps are typical Windows In vivo
of many forms of angiogenesis; however, solid cords of ECs Angiogenesis is difficult to observe and manipulate in
may be absent from advancing sprouts in wounds and cer- vivo because of the thickness and opacity of most tissues.
tain developing systems, such as the retina. Instead, nascent Tissue sections offer only a static view and provide little in-
lumens form immediately behind the tips, which are com- formation about the three dimensional structure of sprout-
prised of one or two spindle-shaped ECs. Although specific ing vasculature. Indeed, it is nearly impossible to identify
elements of angiogenesis might vary, the molecular processes the advancing fronts of sprouts (the critical areas of inva-
of capillary morphogenesis must mediate one or more of sion and morphogenesis) in sectioned tissue. To circumvent
the following six individual or cooperative behaviors of ECs: these problems, investigators have attempted to find or make
How Does Extracellular Matrix Control Capillary Morphogenesis? 327
windows into the living animal. In the late nineteenth and details of vascular morphogenesis toward clinical investiga-
early twentieth centuries, angiogenesis had been observed tions of the role of angiogenesis in the growth of tumors.
directly in the thin, transparent tails of amphibian larvae From these largely descriptive studies came validation of the
(for references and description of early work, see ref. 10). In central paradigm of tumor-induced vascular chemotaxis and
the 1930s, Clark and Clark10 viewed the revascularization of the corresponding discovery of diffusible, tumor-derived
wounds in rabbit ears through transparent chambers and molecules that promoted vascular growth. An increasing
made elegant drawings of the time course of angiogenic in- interest in an understanding of angiogenesis at the molecu-
vasion, anastomosis, formation of lumens, and interaction lar level led to the development of models in vitro, in which
of ECs with the ECM of the fibrin clot. Subsequent studies angiogenesis-related behaviors of ECs were studied under
with the rabbit ear chamber and other window-type mod- highly simplified, defined conditions. It was from such mod-
els (e.g., transparent chambers in mouse skin11 and hamster els that ECM was identified as a key element of vascular
cheek pouches,12 rabbit corneal pockets,13 and chick chorio- morphogenesis.
allantoic membranes)14 shifted the focus away from specific
328 Tissue Engineering of Prosthetic Vascular Grafts
medium, lateral surfaces that contacted adjacent ECs, and however, relatively little is known about the means by which
“abluminal” surfaces that faced the collagen substrate. This ECs move through ECM. Indeed, the basic mechanism of
topology resulted in an angiogenesis-like response to the ameboid movement by animal cells remains unclear, despite
tumor-promoting ester phorbol myristate acetate: ECs pen- intensive study of selected cell types such as fibroblasts.56
etrated the collagen as invaginations of the cellular sheet Moreover, almost all detailed mechanistic studies of cell
(much like the outpocketing that occurs during early stages movement have been conducted in vitro with cells on top of
of sprout formation in vivo) and formed branched, multi- planar substrates that are rigid (i.e., glass or plastic), or
cellular tubes with abluminal basal lamina-like material and semideformable (e.g., silicone rubber), and hence unlike
patent, fluid-filled lumens similar to those of capillary malleable ECM in vivo. On such artificial surfaces, fibro-
sprouts in vivo10 (Fig. 30.3). The induction of invasive blasts, ECs, and many other cell types exhibit a “crawling”
sprouting by both micro- and macrovascular ECs53-55 rein- form of motility characterized by a repeated cycle of:
forces the notion that the structure of the parent vessel (i.e., 1. Protrusion of lamellipodia or filopodia at the lead-
a tube of mutually adherent cells with fluid plasma inside ing edge of the cell;
and ECM outside) provides vital cues and constraints that 2. Adhesion of the protrusive structures to the support-
position ECs for correct orientative behaviors once their ive substrate;
angiogenic programs are initiated. 3. Movement of the cell body and nucleus forward by
the process of traction, during which the cell pulls
Interactions Between ECs and ECM strongly on the supportive substrate;
That Mediate Angiogenesis 4. De-adhesion of the rear, pointed “tail” of the cell from
As discussed above, differently-conceived models of the substrate; and
angiogenesis all agree on the central role played by ECM in 5. Retraction of the tail toward the cell center 56
vascular morphogenesis. In the following section, we will (Fig. 30.4).
move beyond descriptive studies to explore ways in which Classic time-lapse observations by Bard and Hay show
the behaviors of ECs and the structure of ECM interact chick corneal fibroblasts exhibiting this cycle of motility as
mechanistically to promote the morphogenesis of capillaries. they migrate through corneal stroma and through a gel of
fibrillar type I collagen in vitro.57 It is likely that ECs move
Characteristics of EC Motility through malleable three dimensional ECM in a simil-
Locomotion of ECs is a critical element of angiogen- ar manner.
esis. A significant number of studies have examined the mi-
gration of ECs in response to various angiogenic molecules;
Malleability of ECM as a Function of Its Composition trusions of ECs in ECM usually consist of one or more very
The migration of ECs and other cell types can be af- thin, elongate filopodia that sometimes exhibit filamentous
fected by the physical properties of the supportive ECM, branches, i.e., structures with minimal frontal area that are
notably malleability. In the context of angiogenesis, mallea- thermodynamically favored in a viscous environment. The
bility is defined as the degree to which ECM deforms in re- possibility that protrusive activity governs the rate of cell
sponse to forces generated by sprouting ECs. ECM is com- locomotion65 might explain why movement of ECs into
prised of a hydrated gel of proteoglycans, glycoproteins, and gelled collagen66 or fibrin67 is inhibited as the density of the
hyaluronan in which fibrillar proteins (e.g., collagens and ECM fibrils increases.
fibronectin) are embedded. The malleability of this mixture
of macromolecules is significantly affected by the concen- Traction of ECs as a Spatial Organizer of ECM
tration and relative proportion of the major fibrillar pro- In crawling cells, actin filaments in the cytoplasmic
teins, such as type I and type III collagens that predominate cortex are driven rearward by association with other motive
in interstitial ECM. An additional element of physical het- proteins, such as myosin.56,68,69 Movement of actin is trans-
erogeneity involves the stiffness of individual protein fibrils, duced into propulsive traction force by focal adhesions
which depends in part on interactions with other compo- (Fig. 30.4), which are spot or bar-shaped specializations of
nents of ECM. For example, the polymerization of type I the plasma membrane that adhere to the substrate (i.e.,
collagen in vitro yields thinner, more pliable fibrils in the ECM) via their extracellular faces and connect to the actin
presence of type III collagen,58 the ECM protein TRAMP,59 via their cytoplasmic faces.68-70 Traction is applied vectorially,
and the ECM proteoglycan decorin.60 The malleability of and generates tension in malleable substrates in vitro (e.g.,
ECM is decreased by increases in the physical entanglement silicone rubber) that is directed parallel to the cellular axis
of fibers (shear-drag) and by the presence of covalent of motion.64,71 Remarkably, fibroblasts exert traction that is
crosslinks, such as the lysyl oxidase-catalyzed crosslinking at least six orders of magnitude greater than is needed to
of type I collagen fibers,61 and the transglutaminase-cata- propel the cell at a normal speed of 1 ∝m/sec against the
lyzed generation of fibronectin multimers.62 The rigidify- viscous drag of aqueous medium.64 It is important to real-
ing effects of shear-drag and crosslinks are counteracted by ize that this magnitude of force and its vectorial application
specific molecular interactions (e.g., shear-drag between type to the substrate are specialized features of crawling cells that
I collagen fibers is reduced by the binding of type XII and are not intrinsic to ameboid locomotion. Neuronal growth
type XIV collagens)63 and by more general influences such cones and fish keratocytes that exhibit an alternative “glid-
as the reduction in fiber length and number by cell-medi- ing” form of ameboid motility64 move 10-60 times faster
ated proteolysis. ECs within a vascular sprout influence the than fibroblasts in vitro,56 generate only 3-12% of the trac-
structural properties of ECM in their immediate vicinity by tion force,64 and apply tension to the substrate at right angles
their synthesis of new forms of ECM and by chemical modi- to the direction of motion.64,72
fication and/or degradation of extant ECM. These processes It is thought that the “excessive” levels of traction de-
can be modulated to facilitate or inhibit vascular invasion, veloped by fibroblasts allow these cells to compress and align
contingent upon the type of signals received by the ECs. ECM in healing wounds73 and within developing tendons,
Additional control over the rate, extent, and locale of vascu- ligaments, and organ capsules.74 The traction of ECs in vitro
lar invasion resides with non-EC types (e.g., fibroblasts) that is similar in magnitude to that of dermal fibroblasts75 and
can control the composition of surrounding ECM via spe- therefore is likely to serve a morphogenetic function. Trac-
cific synthetic and degradative activities. tion at the advancing front of sprouts would tend to align
ECM in the direction of invasion and thus would facilitate
Malleability of ECM Influences the Motility of ECs protrusion of filopodia from leading ECs. Moreover, elastic
It is likely that the malleability of ECM directly influ- interactions among ECM fibrils allow them to be aligned
ences the generation of protrusive structures by cells. Pro- over great distances (as much as one centimeter in vitro),32,74
trusion of filopodia and lamellipodia by crawling cells in- and to form pathways on which invading ECs would move
volves the polymerization of cytoplasmic actin that either by contact guidance.76 Such matrical pathways mediate mi-
pushes the plasma membrane forward or stabilizes bulges gration of ECs in planar angiogenesis in vitro and have been
in the plasma membrane made by other means (e.g., via ther- associated with the movement of endocardial cells into the
mal movements or the local influx of water).56 Counterforces endocardial cushions of the developing chicken heart.77
exerted by ECM on the front of a cell (generated, for ex- Moreover, the existence of matrical pathways is consistent
ample, by the resistance of stiff, fibrillar ECM to bending or with the follow-the-leader behavior of ECs that migrate
the resistance of charged, hydrated glycans to compression) through three dimensional ECM in vitro78 and in vivo.3
would slow the rate or extent of protrusion by shifting the Matrical pathways could also mediate the development of
thermodynamic equilibrium in favor of actin depolymer- anastomoses between vascular sprouts. ECs at the tips of
ization. Another consequence of ECM counterforces would adjacent sprouts would align ECM between them by a trac-
be their effect on the shape of protrusions. For example, tion-mediated two-center effect, approach one another via
during progressive movement over rigid, planar substrates the matrical pathway, and fuse to establish a common lu-
in vitro, during which viscous drag from the culture me- men (Fig. 30.5).
dium is negligible,64 fibroblasts and ECs frequently project Beyond the role of traction to facilitate vascular inva-
blunt or fan-shaped lamellipodia. In contrast, forward pro- sion and anastomosis of sprouts is the potential for traction
How Does Extracellular Matrix Control Capillary Morphogenesis? 333
ECs could control their adhesion directly by secretion of hesive receptors (e.g., integrins) to their cognate ECM
specific types of ECM in their immediate vicinity (referred ligands. Alternatively, the cell-binding domains could inter-
to as pericellular matrix). For example, when stationary bo- act with specific cell surface receptors to trigger
vine aortic ECs are induced to migrate in vitro, they switch counteradhesive responses through second messenger sig-
from synthesis of an ECM rich in heparan sulfate naling pathways. In addition to matricellular proteins, in-
proteoglycan (which stabilizes the binding of focal adhesions vestigators have identified other important classes and fami-
to fibronectin and inhibits cell migration) to an ECM en- lies of counteradhesive molecules, including some novel
riched in chondroitin/dermatan sulfate proteoglycans which members.119 It is likely that further study of counteradhesion
destabilize adhesion sites and promote cell motility.87,88 will improve our understanding of how angiogenesis and
other types of morphogenesis are regulated.
Matricellular Proteins and Counteradhesion
In the context of EC adhesion, of particular relevance Integrins Mediate Adhesion to ECM
are matricellular proteins which reside (often transiently) and Signaling in Focal Adhesions
in the ECM and influence adhesion between cells and their Integrins are a major class of transmembrane recep-
ECM ligands.89,90 Unlike proteoglycans and other adhesion- tors that mediate the attachment of cells to ECM proteins
modulating components of ECM, matricellular proteins do via focal adhesions. They comprise a family of at least 16 #
not contribute significantly to ECM structure. Matricellular and 8 ! subunits that associate noncovalently in over 20 dif-
proteins are secreted by many cell types and consist of three ferent #/! combinations on the cell surface. Both # and !
families exemplified by SPARC (secreted protein, acidic and subunits are required for high-affinity binding to an ECM
rich in cysteine), 91 thrombospondin-1 (TSP),90,92 and ligand: The specific ligand is determined by the combina-
tenascin-C (TN).93 Although SPARC, TSP, and TN are struc- tion of distinct # and ! subunits. Some integrins recognize a
turally unrelated, a commonality is their enhanced expres- single ECM protein (e.g., integrin #5!1 recognizes only
sion by migrating, dividing cells of subconfluent cultures fibronectin); however, it is more common for an integrin to
and in tissues undergoing embryogenesis and wound re- bind to two or more ligands. An ECM protein may be rec-
pair.94 Moreover, these proteins have similar effects on cells ognized by more than one integrin; for example, fibronectin
in culture: They inhibit attachment and spreading, and cause and laminin are bound by the !1 subunit in combination
partial detachment and rounding of spread cells. In accor- with one of four and five different # subunits, respectively.
dance with these counteradhesive properties, SPARC, TSP, Different integrins that share an ECM ligand may bind to
and TN reduce the number of focal adhesions made by at- the ligand at the same site, as do #5!1 and #3!1, or they can
tached, subconfluent ECs.94-96 Although these proteins are recognize different regions of the ligand, as do the #1!1 and
associated with capillaries in vivo,97-99 their influence on #6!1 laminin receptors, or the #5!1 and #4!1 fibronectin
angiogenesis remains unclear. Whereas SPARC itself has receptors.120 ECs and other cell types typically express sev-
minimal effects on angiogenesis in vivo, specific peptide frag- eral different integrins on their surfaces concurrently and
ments of SPARC have angiogenic properties.100 TSP is de- can alter the expression of one integrin or more in response
posited in a trail behind migrating ECs101 and promotes their to stimuli.
chemotaxis in vitro.102 It is also reported to inhibit the mi- Unbound integrins are freely diffusive in the plane of
gration and sprouting of ECs in vitro.103,104 TSP is generally the plasma membrane; however, binding to ECM or other
believed to be anti-angiogenic in vivo;98,103,105 however, it is ligands causes adjacent integrins to cluster, an event medi-
proposed that TSP stimulates angiogenesis by indirect ef- ated by the cytoplasmic domains of the ! subunits.121 Clus-
fects on non-EC types.106 The association of TN with pro- tering of unligated #/! heterodimers is prevented by the #
liferating vessels in the vicinity of tumors99,107,108 could in- subunit; however, this inhibition is removed following en-
dicate that it promotes angiogenesis. In accordance with this gagement of the ligand. Integrin clusters increase in size by
hypothesis, planar angiogenesis in vitro is inhibited by anti- further aggregation and eventually become large enough to
bodies against TN.104 be recognized as focal adhesions. As a focal adhesion grows,
Matricellular proteins bind both to cells and to ECM. the clustering of the short cytoplasmic domains of the # and
SPARC binds TSP109 and type I through V collagens,110,111 ! integrin subunits nucleates the formation of large molecu-
and also interacts with ECs via as yet uncharacterized bind- lar complexes comprised of cytoskeletal linking proteins and
ing proteins or receptors.112 TSP binds collagen, fibronectin, a variety of catalytic signaling molecules. The cytoskeletal
laminin, fibrinogen,89 and heparan sulfate proteoglycans of linking proteins, which include #-actinin, talin, vinculin,
ECM or the cell surface.92 Moreover, TSP binds to the cell paxillin, and tensin, connect the clustered integrins to the
surface integrin #v!3,92 an ECM receptor that is expressed actin cytoskeleton and thereby strengthen the focal adhe-
on ECs and plays an important role in angiogenesis. TN has sion mechanically and couple it to the motility apparatus.
a low affinity for ECM proteins such as collagens, fibronectin, The catalytic signaling proteins (notably tyrosine kinases and
and laminin113 but binds to several cell surface receptors, their substrates) initiate cascades of molecular association
including integrins #2!1 and #v!3 on ECs,114,115 integrin and enzymatic activity in both the cytoplasm and the nucleus
116
#9!1, proteoglycans,117 and annexin II.118 The cell and that regulate the cell cycle, responses to growth factors, and
ECM-binding domains of matricellular proteins, either sepa- expression of specific gene products via several convergent
rately or in combination, could mediate counteradhesion pathways (see refs. 122-125 for general reviews of integrin-
by physical interference with the binding of cell surface ad- mediated signal transduction). The sequestration of signal-
How Does Extracellular Matrix Control Capillary Morphogenesis? 335
ing proteins in focal adhesions of ECs is rapid enough to vitronectin, fibronectin, fibrinogen, laminin, von willebrand
play a role in integrin-mediated responses to cytokines. Fif- factor, and denatured type I collagen. Thus, expression of
teen to thirty minutes after bovine capillary ECs bind to this integrin enables a cell to adhere to, migrate on, or re-
fibronectin or the immobilized peptide Arg-Gly-Asp (the spond to almost any ECM protein it might encounter.138
recognition site in fibronectin for integrin #5!1) in vitro, their Integrin #v!3 is expressed preferentially in angiogenic ves-
nascent focal adhesions are enriched in Flg (a high-affinity sels135,139 and is necessary for angiogenesis, since a mono-
receptor for the angiogenic protein basic fibroblast growth clonal antibody (LM609) against this integrin blocked
factor [bFGF]), the tyrosine kinases pp60c-Src and pp125FAK, cytokine-induced angiogenesis in chick chorioallantoic
and other signaling molecules including phospha- membranes,135 tumor-induced angiogenesis in human skin
tidylinositol-3-kinase, phospholipase C-&, and the Na+/H+ transplanted on severe combined immunodeficiency (SCID)
antiporter.126 mice,140 and bFGF-induced angiogenesis in the rabbit cor-
nea.141 LM609 also disrupted vascular morphogenesis and
Functional Characteristics of Integrins Expressed by ECs lumen formation within vasculature of early quail em-
Human umbilical vein ECs in vitro express integrin # bryos.142 In the chick chorioallantoic membrane, antago-
subunits 1-6127,128 and ! subunits 1 and 3,127 with #2!1 as the nists of #v!3 induced programmed cell death (apoptosis) of
predominant combination. Integrin #2!1 is present on cap- ECs in sprouting blood vessels. Engagement of #v!3 with an
illaries in vivo,129 and this integrin is responsible for most of ECM ligand might therefore provide a “survival signal” to
the binding of ECs to type I and IV collagens in vitro.128 ECs during angiogenesis.143
Integrin #2!1 promotes the migration of ECs on type I col-
lagen130 and mediates the reorganization of type I collagen Integrins, Cell Shape, and Gene Expression
by EC traction in vitro.131 Although #2!1 contributes signifi- The genetic programs that mediate proliferation and
cantly to the binding of human umbilical vein ECs to differentiation of ECs and other cell types tend to be mutu-
laminin-coated plastic,127,128 it plays little or no role in the ally exclusive in vitro; e.g., synthesis of cell-specific gene
traction-mediated organization of ECs into networks on products diminishes when cells are stimulated to divide.144
Matrigel in vitro132—a process dependent on the laminin- Since both programs are triggered by the binding of cell sur-
binding integrin #6!1.132,133 Despite the involvement of !1 face integrins to ECM, additional elements of control must
integrins in adhesion and traction-related processes in vitro, exist to determine which program is selected. A variety of
the role played by this integrin subfamily in vascular mor- studies point to cell shape as an important element in the
phogenesis is unclear. A monoclonal antibody (CSAT) switch between proliferation and differentiation. In general,
against the avian !1 integrin subunit induced abnormalities cells that assume rounded shapes exhibit a differentiated
of aortic vasculogenesis in the quail embryo that included phenotype. For example, levels of synthesis of the milk pro-
inhibition of luminal development.134 CSAT, however, does tein !-casein by mouse mammary cells (induced by the bind-
not inhibit angiogenesis in the chick chorioallantoic mem- ing of laminin to cell surface !1 integrins) are high in cells
brane despite the presence of !1 integrins in nascent blood made round by culture on nonadhesive substrates, but are
vessels.135 In accordance with the effects of CSAT in the quail low in flattened, spread cells grown on surfaces that permit
embryo, antibodies against the #2 and !1 integrin subunits cell adhesion.145 Spreading of cells facilitates proliferation:
inhibited development of lumen-like intracellular vacuoles Ligand-induced clustering of integrins, in the presence of
within human ECs cultured in three dimensional type I col- mitogenic factors, is sufficient to induce ECs and other cell
lagen gels.37 It is not known how the engagement of type I types to exit from G0 and progress into the late G1-phase of
collagen by #2!1 promotes the formation of lumens. Pos- the cell cycle in vitro;146-149 however, the cells will not enter
sible mechanisms include: S-phase unless they are allowed to spread.148,149 The path-
1. Induction of a “program for lumen formation” via ways by which cell shape is coupled to proliferation and gene
intracellular signaling pathways; expression are not understood, although evidence points to
2. Application of traction force to the collagen that involvement of the cytoskeleton. It is known that the net-
causes distortion of cell structure (cavitation) rather work of actin filaments generates tension in the cytoplasm
than cell migration or reorganization of collagen; and that promotes cell retraction and rounding. This tension is
3. Clustering of #2!1 at selected sites to provide cell- opposed both intracellularly by stiff, compression-resistant
matrix anchor points for the development of pinocy- microtubules and extracellularly by the counteracting ten-
totic membrane invaginations.37 sion of ECM, which is coupled directly to the actin network
Exposure of capillary ECs to bFGF augments their through integrins within focal adhesions. It is hypothesized
expression of integrin subunits #2, #3, #5, #6, !1, !3, !4, and !5 that the equilibrium between these forces places the cytosk-
and results in a corresponding increase in adhesion of ECs eleton at a “setpoint” of stress which defines a specific pat-
to collagen, fibronectin, laminin, and vitronectin.136,137 The tern of gene expression via mechanically sensitive biochemi-
anti-angiogenic transformtin growth factor-! (TGF-!) cal processes in the plasma membrane, cytoplasm, and
stimulates #2, #5, and !1 in these cells;137 the pattern of nucleus.150,151 It follows that a change in the setpoint of stress
integrin expression by ECs is thus altered selectively by dif- caused by alterations in:
ferent cytokines. 1. Actin-generated forces;
Integrin #v!3 binds to a variety of ECM proteins with 2. Cytoskeletal structure;
an exposed tripeptide Arg-Gly-Asp moiety, such as
336 Tissue Engineering of Prosthetic Vascular Grafts
3. Strength, number, or spatial arrangement of adhe- by ECs include the interstitial collagenase MMP-1, which
sive contacts to ECM; or cleaves native type I, II, and III collagens at a single site;158,159
4. Malleability of ECM gelatinases A (MMP-2) and B (MMP-9), which degrade de-
would alter the biochemical equilibrium to favor ex- natured collagens (gelatins), type IV and V collagens, and
pression of a different set of genes. Electron microscopic elastin,160,161 and stromelysin (MMP-3), which has a broad
studies of resinless sections152 reveal that tension applied to range of substrates that include laminin, fibronectin, the pro-
the cell surface could be transmitted first to the nuclear en- tein core of proteoglycans, and gelatin.161,162 Recently, it has
velope, via bridges of intermediate filaments, and then into been demonstrated that MMP-2 is an interstitial collagenase
the nuclear interior, via nuclear fibrils that insert into the with a specificity similar if not identical to those of MMPs-1
inner side of the nuclear envelope. In support of these ob- and -8.163 Type I and type III collagens are highly resistant
servations, Maniotis et al have shown recently that a me- to proteolysis by trypsin, plasmin, and other members of
chanical pull applied to integrins on the EC surface causes a the serine and sulfhydryl proteinase families. These collagens,
physical distortion of the nucleus and a reorganization of however, are degraded by MMPs via a two-stage process in-
nucleoli along the axis of applied tension.153 The functional volving initial cleavage by interstitial collagenases and sub-
consequence of this chain of mechanical events might in- sequent digestion of the larger fragments by gelatinases.
clude control of DNA synthesis, since the degree to which Studies in vitro indicate a role for MMPs in angiogen-
ECs spread on coatings of ECM in vitro, the physical expan- esis. Exposure of ECs to phorbol esters and angiogenic
sion of the nucleus, and the rate of DNA synthesis are closely growth factors in vitro induces production of MMPs-1, -2,
correlated.149 -3, and -9.164-167 MMPs are regulated in vivo by tissue in-
During angiogenesis in vitro, ECs are likely to hibitors of metalloproteinases—a family of low molecular
encounter a shifting pattern of external forces that are modu- weight proteins (TIMPs-1, -2, and -3) that specifically block
lated by: the catalytic activity of MMPs.161 Inhibition of endogenous
1. Cell-cell adhesion; MMP activity of ECs in vitro, either by metal ion chelators
2. Enzymatic modification of extant vicinal ECM; or by TIMPs, suppresses traction-mediated reorganization
3. Synthesis of new ECM; of type I collagen gels75 and the corresponding angiogen-
4. Changes in patterns of integrins or other cell-sur- esis-like invasion of these gels.52,55,168
face receptors that mediate adhesion to ECM; and The inherent complexity of proteolysis and its regula-
5. Restructuring of ECM by the traction of ECs and tion are well illustrated by the plasmin/PA system. PAs are
non-EC types. serine proteinases that cleave the zymogen plasminogen (a
Asymmetrical or focal application of forces to specific plasma protein) to plasmin. Plasmin is itself a proteinase
areas of a sprout could cause specific ECs to proliferate and/ that degrades several components of ECM, such as
or express distinct gene products. Such localized behaviors fibronectin, laminin, the protein core of proteoglycans, and
could alter the morphology of the sprout—for example, the gelatins.169 The tissue-type (tPA) form of PA is expressed by
sprout could bend or branch. Clearly, much remains to be ECs of quiescent microvasculature and is neither modulated
discovered regarding the complex role of biomechanical during angiogenesis nor expressed by sprouting ECs in vivo.
processes in vascular morphogenesis. In contrast, the urokinase (uPA) form of PA is expressed by
angiogenic ECs associated with tumors, inflammation, he-
Angiogenesis Requires Proteolysis of ECM mangiomas, ovarian follicles, corpora lutea, and maternal
At the site of a new vascular sprout, ECs of the parent decidua. Little or no uPA is expressed in quiescent vessels.168
vessel secrete proteases that effect a focal degradation of the Plasminogen is present in most tissues at high levels;
basement membrane.154 Proteolysis continues as the sprout therefore, its activation by PAs results in high local concen-
invades the interstitial ECM: Clark and Clark observed that trations of plasmin. Moreover, plasmin-mediated, limited
invasive capillaries within rabbit ear wounds dissolved fi- proteolysis activates latent elastase (a serine protease) and
brin in their immediate vicinity and thus created a “clear MMPs-1, -2, -3, and -9. Thus, small quantities of PA can
space” around each sprout.10 Experimental studies have generate significant levels of proteolytic activity with differ-
shown proteolysis is necessary for angiogenesis within the ent substrate specificities.169
developing chorioallantoic membrane of the chicken155 and The position of uPA as the initiator of a proteolytic
for invasion of ECs into three dimensional ECM comprised cascade dictates that its activity be strictly controlled during
of fibrin,156 type I collagen,55 and the amniotic basement angiogenesis. Expression of uPA by ECs is modulated at the
membrane.157 The basis of the permissive effect of proteoly- transcriptional level by a number of angiogenic growth fac-
sis on angiogenesis is not fully understood. It is known that tors. Moreover, angiogenic ECs in vitro and in vivo express
various angiogenic factors (e.g., bFGF) bound to the ECM high levels of PA inhibitor-1 (PAI-1),170,171 a protein which
are made available to ECs after the matrix is degraded. It is blocks the formation of plasmin by PA and thereby inhibits
likely, however, that the most important effect of proteoly- the activation of elastase and MMPs. An additional element
sis is to alter the malleability and adhesivity of the ECM to of control is the confinement of active uPA to the surface of
facilitate the movement of ECs through it. ECs. uPA is secreted as a proenzyme (pro-uPA) that binds
Two major groups of proteinases that regulate angio- to uPA receptor (uPAR), a glycoprotein which is anchored
genesis are the matrix metalloproteinases (MMPs) and the to the surface of ECs at the site of focal adhesions.172 The
plasmin/plasminogen activator (PA) system. MMPs secreted bound pro-uPA is subsequently activated by limited pro-
How Does Extracellular Matrix Control Capillary Morphogenesis? 337
teolysis.173 The proteolytic function of uPA at the cell sur- of neovascularization for efficient healing of wounds, bio-
face is facilitated by the binding of its substrate, plasmino- integration of prosthetic devices, functionality of the repro-
gen, to the plasma membrane by #-enolase-related proteins ductive system, and regulation of tumor growth and me-
and cell-surface chondroitin-sulfate proteoglycans.174,175 tastasis, we could not overemphasize the need for a more
After a period of time (possibly several hours), active uPA is complete and mechanistic understanding of angiogenesis
inactivated by binding to PAI-1, internalized, and rapidly as a therapeutic means for the prevention and treatment of
degraded.176 The importance of PAI-1 to the control of uPA human disease. In the field of tumor biology, for example,
activity is indicated by the effects of angiogenic growth fac- few would disagree that some form of anti-angiogenic
tors on the expression of the two proteins. bFGF and vascu- therapy might be beneficial for the treatment of solid tu-
lar endothelial growth factor (VEGF) increase the ratio of mors. However, there is considerable debate regarding the
uPA mRNA to PAI-1 mRNA in ECs, for a net increase in type of intervention (i.e., which stage of blood vessel growth
proteolysis. In contrast, TGF-!1 reduces the uPA:PAI-1 should be targeted), as well as the duration and route of
mRNA ratio for a net reduction in proteolysis—an effect administration of angiostatic drugs or compounds.
consonant with its inhibition of angiogenesis and invasion We have chosen to focus on questions and models that
of ECM by ECs in vitro.168 address the angiogenic process itself and have recapitulated
Migratory ECs exhibit increased expression of uPA and the acknowledged stages of vascular sprout formation and
uPAR relative to nonmigratory ECs in confluent monolay- progression—EC migration, alignment, proliferation, cohe-
ers. 168 Nanomolar concentrations of uPA or of its sion, lumen formation, and branching/anastomosis—in the
noncatalytic A-chain stimulate motility of ECs.177 Blockage context of cell adhesion, movement, and shape, molecular
of uPAR with monoclonal antibodies or a specific antago- biology, receptor-ligand interactions, and extracellular pro-
nist inhibits the migration of ECs on serum-coated substrates teolysis. To date, there are no “perfect” models in which to
in vitro to a greater degree than has been observed after the test angiogenic regulation, although much can be learned
inhibition of endogenous plasmin activity.178 Moreover, from culture systems in which ECs form capillary-like struc-
uPAR binds to the ECM component vitronectin and inhib- tures. At best, each of the models in vitro that were covered
its !1 integrin-dependent adhesion to fibronectin.179 Col- in this chapter appears optimal for one or more of the stages
lectively, these observations point to a direct involvement of requisite to blood vessel formation. In contrast, tissues of
the uPA/uPAR complex in the modulation of cell adhesion living animals provide excellent examples of angiogenesis,
during motility by a mechanism independent from proteoly- but their disadvantages include the complex interactions
sis. Since PAI-1 is also increased in migratory ECs,168 it is among several types of cells and the limitations in visualiza-
possible that this inhibitor could influence adhesive inter- tion and quantification of blood vessel growth.
actions between uPA/uPAR and ECM ligands. A synergistic We have also stressed the importance of the ECM and
role for PAI-1 in counteradhesion would explain the find- its capacity for the regulation of EC behavior and vascular
ing that synthesis of PAI-1 by subconfluent, migratory ECs morphogenesis. Clearly, alteration of the properties of ECM
is increased in the presence of the counteradhesive, through its molecular composition and/or proteolysis has
matricellular protein SPARC.180 profound effects on EC migration, proliferation, and bio-
The degree to which ECM must be degraded to facili- synthetic capacity, all of which can be affected, in part, by
tate angiogenesis in vivo is unclear; however, it appears that changes in cell shape and adhesion. Both adhesive (e.g., col-
the range of optimal proteolysis has an upper limit. For ex- lagen, fibronectin, laminin) and counteradhesive proteins
ample, the increased production of endogenous proteases (e.g., SPARC, TSP), as well as the integrins and other cell
by murine ECs that express the polyoma virus middle T surface receptors for ECM macromolecules, are important
oncogene is correlated with their organization into large, in the regulation of EC shape and function.
hollow cysts within fibrin clots in vitro.181 In the presence There remain several unanswered questions that are
of inhibitors of serine proteases, however, the cyst currently receiving considerable attention in the field of vas-
morphotype is replaced by capillary-like tubes that are gen- cular biology:
erated by the invasion of ECs into the fibrin matrix. The 1. Which are the macromolecules that are truly critical
fact that proteolysis of ECM inhibits the formation of en- for angiogenesis?
dothelial tubes indicates that angiogenic sprouts are not elon- 2. Is all angiogenesis the same (e.g., in a normal tissue
gated by forces derived solely from the parent vessel (e.g., by versus a tumor, and in the endometrium versus the
a hydraulic push). Alternatively, the sprouts pull themselves brain)?
outward by a process (e.g., traction) that requires mechani- 3. What is the initiating stimulus for sprouting?
cal support from the ECM. Accordingly, excessive proteoly- 4. How is selective or focal proteolysis controlled?
sis of the ECM would disrupt its mechanical continuity to 5. What stage of angiogenesis provides the most rea-
an extent that ECs would lack a fixed substrate against which sonable site for intervention to effect either stimula-
they could pull. tion or stasis?
Reagents and methodology are largely in place to ad-
Concluding Remarks dress these problems, although there is a continuing need
In this chapter we have introduced the fundamental for better quantitative models in vivo and in vitro. Within
characteristics of angiogenesis and have discussed the cell the next decade we should expect major and significant
biology of this highly complex process. Given the necessity progress in basic research that will allow clinicians,
338 Tissue Engineering of Prosthetic Vascular Grafts
bioengineers, and molecular/cellular biologists to develop tochemical localization and role in cell attachment. Lab
and test novel strategies for the control of vascular growth. Invest 1983; 49:362-370.
22. Sage H. Collagen synthesis by endothelial cells in culture.
References In:Jaffe EA, ed. Biology of Endothelial Cells. The
1. Folkman J. Angiogenesis in cancer, vascular, rheumatoid Hague:Martinus Nijhoff, 1984:161-177.
and other disease. Nature Med 1995; 1:27-31. 23. Pratt BM, Harris AS, Morrow JS et al. Mechanisms of
2. Sage EH. Angiogenesis inhibition in the context of en- cytoskeletal regulation. Modulation of aortic endothelial
dothelial cell biology. Adv Oncol 1997; 12:17-29. cell spectrin by the extracellular matrix. Am J Pathol 1984;
3. Folkman J. Tumor angiogenesis. Adv Cancer Res 1985; 117:349-354.
43:175-203. 24. Folkman J, Haudenschild CC, Zetter BR. Long-term cul-
4. Paweletz N, Knierim M. Tumor-related angiogenesis. Crit ture of capillary endothelial cells. Proc Natl Acad Sci USA
Rev Oncol Hematol 1989; 9:197-242. 1979; 76:5217-5221.
5. Folkman J. Angiogenesis: Retrospect and outlook. In: 25. Folkman J, Haudenschild C. Angiogenesis in vitro. Na-
Steiner R, Weisz PB, Langer R, ed(s). Angiogenesis. Basel: ture 1980; 288:551-556.
Birkhäuser Verlag, 1992:4-13. 26. Maciag T, Kadish J, Wilkins L et al. Organizational be-
6. Zagzag D. Angiogenic growth factors in neural embryo- havior of human umbilical vein endothelial cells. J Cell
genesis and neoplasia. Am J Pathol 1995; 146:293-309. Biol 1982; 94:511-520.
7. Fox SB, Gatter KC, Harris AL. Tumor angiogenesis. J 27. Feder J, Marasa JC, Olander JV. The formation of capil-
Pathol 1996; 179:232-237. lary-like tubes by calf aortic endothelial cells grown in
8. Chaplain MAJ, Stuart AM. A model mechanism for the vitro. J Cell Phys 1983; 116:1-6.
chemotactic response of endothelial cells to tumor angio- 28. Madri JA. Endothelial cell-matrix interactions in hemo-
genesis factor. IMA J Math Appl Med Biol 1993; stasis. In:Spaet TH, ed. Progress in Hemostasis and
10:149-168. Thrombosis. Vol 6. New York:Grune and Stratton,
9. Omitted in proof. 1982:1-24.
10. Clark ER, Clark EL. Microscopic observations on the 29. Ingber DE, Folkman J. Mechanochemical switching be-
growth of blood capillaries in the living mammal. Am J tween growth and differentiation during fibroblast growth
Anat 1939; 64: 251-301. factor-stimulated angiogenesis in vitro: Role of extracel-
11. Algire GH, Chalkley HW, Legallais FY et al. Vascular re- lular matrix. J Cell Biol 1989; 109:317-330.
actions of normal and malignant tissues in vivo. I. Vas- 30. Kubota Y, Kleinman HK, Martin GR et al. Role of laminin
cular reactions of mice to wounds and to normal and and basement membrane in the morphological differen-
neoplastic transplants. J Natl Cancer Inst 1945; 6:73-85. tiation of human endothelial cells into capillary-like struc-
12. Greenblatt M, Shubik P. Tumor angiogenesis:Transfilter tures. J Cell Biol 1988; 107:1589-1598.
diffusion studies in the hamster by the transparent cham- 31. Vernon RB, Angello JC, Iruela-Arispe ML et al. Reorga-
ber technique. J Natl Cancer Inst. 1968; 41:111-124. nization of basement membrane matrices by cellular trac-
13. Gimbrone MA, Cotran RS, Leapman S et al. Tumor tion promotes the formation of cellular networks in vitro.
growth and neovascularization: An experimental model Lab Invest 1992; 66:536-547.
using the rabbit cornea. J Natl Cancer Inst 1974; 32. Harris AK, Stopak D, Wild P. Fibroblast traction as a
52:413-427. mechanism for collagen morphogenesis. Nature (London)
14. Ausprunk DH, Knighton DR, Folkman J. Differentiation 1981; 290:249-251.
of vascular endothelium in the chick chorioallantois:A 33. Vernon RB, Lara SL, Drake CJ et al. Organized type I
structural and autioradiographic study. Dev Biol 1974; collagen influences endothelial patterns during “sponta-
38:237-248. neous angiogenesis in vitro”:Planar cultures as models of
15. Jaffe EA, Nachman RL, Becker CG et al. Culture of hu- vascular development. In Vitro Cell Dev Biol 1995;
man endothelial cells derived from umbilical veins. Iden- 31:120-131.
tification by morphologic and immunologic criteria. J Clin 34. Sage EH, Vernon RB. Regulation of angiogenesis by ex-
Invest 1973; 52:2745-2756. tracellular matrix: The growth and the glue. J Hyperten-
16. Gimbrone MA. Culture of vascular endothelium. In:Spaet sion 1994; 12 (suppl 10):S145-S152.
TH, ed. Progress in Hemostasis and Thrombosis. Vol 3. 35. Montesano R, Orci L, Vassalli P. In vitro rapid organiza-
New York:Grune and Stratton, 1976:1-28. tion of endothelial cells into capillary-like networks is
17. Fishman JA, Ryan GB, Karnovsky MJ. Endothelial regen- promoted by collagen matrices. J Cell Biol 1983;
eration in the rat carotid artery and the significance of 97:1648-1652.
endothelial denudation in the pathogenesis of myointimal 36. Elsdale T, Bard J. Collagen substrata for studies on cell
thickening. Lab Invest 1975; 32:339-351. behavior. J Cell Biol 1972; 54:626-637.
18. Madri JA, Pratt BM. Endothelial cell-matrix interactions: 37. Davis GE, Camarillo CW. An #2!1 integrin-dependent
In vitro models of angiogenesis. J Histochem Cytochem pinocytotic mechanism involving intracellular vacuole for-
1986; 34:85-91. mation and coalescence regulates capillary lumen and tube
19. Gimbrone MA, Cotran RS, Folkman J. Human vascular formation in three-dimensional collagen matrix. Exp Cell
endothelial cells in culture. Growth and DNA synthesis. J Res 1996; 224:39-51.
Cell Biol 1974; 60:673-684. 38. Madri JA, Pratt BM, Tucker AM. Phenotypic modulation
20. Madri JA, Stenn KS. Aortic endothelial cell migration I. of endothelial cells by transformtin growth factor-! de-
Matrix requirements and composition. Am J Pathol 1982; pends on the composition and organization of the extra-
106:180-186. cellular matrix. J Cell Biol 1988; 106:1375-1384.
21. Palotie A, Tryggvason K, Peltonen L et al. Components 39. Merwin JR, Anderson JM, Kocher O et al. Transforming
of subendothelial aorta basement membrane. Immunohis- growth factor beta1 modulates extracellular matrix orga-
How Does Extracellular Matrix Control Capillary Morphogenesis? 339
nization and cell-cell junctional complex formation dur- 60. Vogel KG, Trotter JA. The effect of proteoglycans on the
ing in vitro angiogenesis. J Cell Phys 1990; 142:117-128. morphology of collagen fibrils formed in vitro. Coll Relat
40. Marx M, Perlmutter RA, Madri JA. Modulation of plate- Res 1987; 7:105-114.
let-derived growth factor receptor expression in microvas- 61. Siegel R. Lysyl oxidase. Int Rev Connect Tissue Res 1979;
cular endothelial cells during in vitro angiogenesis. J Clin 8:73-118.
Invest 1994; 93:131-139. 62. Fogerty FJ, Mosher DF. Mechanisms for organization of
41. Kuzuya M, Kinsella JL. Induction of endothelial cell dif- fibronectin matrix. Cell Differ Dev 1990; 32:439-450.
ferentiation in vitro by fibroblast-derived soluble factors. 63. Nishiyama T, McDonough AM, Bruns RR et al. Type XII
Exp Cell Res 1994; 215:310-318. and XIV collagens mediate interactions between banded
42. Nicosia RF, Tchao R, Leighton J. Histotypic angiogenesis collagen fibers in vitro and may modulate extracellular
in vitro:Light microscopic, ultrastructural, and radioau- matrix deformability. J Biol Chem 1994; 269:28193-28199.
tographic studies. In Vitro 1982; 18:538-549. 64. Oliver T, Lee J, Jacobson K. Forces exerted by locomoting
43. Nicosia RF, Tchao R, Leighton J. Angiogenesis-dependent cells. Semin Cell Biol 1994; 5:139-147.
tumor spread in reinforced fibrin clot culture. Cancer Res 65. Keller HU, Bebie H. Protrusive activity quantitatively de-
1983; 43:2159-2166. termines the rate and direction of cell locomotion. Cell
44. Villaschi S, Nicosia RF. Angiogenic role of endogenous Motil Cytoskel 1996; 33:241-251.
basic fibroblast growth factor released by rat aorta after 66. Vernon RB, Sage EH, unpublished observations.
injury. Am J Pathol 1993; 143:181-190. 67. Nehls V, Herrmann R. The configuration of fibrin clots
45. Nicosia RF, Nicosia SV, Smith M. Vascular endothelial determines capillary morphogenesis and endothelial cell
growth factor, platelet-derived growth factor, and insu- migration. Microvasc Res 1996; 51:347-364.
lin-like growth factor-1 promote rat aortic angiogenesis 68. Schmidt CE, Horwitz AF, Lauffenburger DA et al.
in vitro. Am J Pathol 1994; 145:1023-1029. Integrin-cytoskeletal interactions in migrating fibroblasts
46. Mori M, Sadahira Y, Kawasaki S et al. Capillary growth are dynamic, asymmetric, and regulated. J Cell Biol 1993;
from reversed rat aortic segments cultured in collagen gel. 123:977-991.
Acta Pathol Jpn 1988; 38:1503-1512. 69. Felsenfeld DP, Choquet DP, Sheetz MP. Ligand binding
47. Nicosia RF, Ottinetti A. Modulation of microvascular regulates the directed movement of !1 integrins on fibro-
growth and morphogenesis by reconstituted basement blasts. Nature 1996; 383:438-440.
membrane gel in three-dimensional cultures of rat aorta:A 70. Lauffenburger DA, Horwitz AF. Cell migration:A physi-
comparative study of angiogenesis in matrigel, collagen, cally integrated molecular process. Cell 1996; 84:359-369.
fibrin, and plasma clot. In Vitro Cell Dev Biol 1990; 71. Harris AK, Wild P, Stopak D. A new wrinkle in the study
26:119-128. of locomotion. Science 1980; 208:177-179.
48. Nicosia RF, Bonanno E, Villaschi S. Large-vessel endot- 72. Oliver T, Dembo M, Jacobson K. Traction forces in
helium switches to a microvascular phenotype during locomoting cells. Cell Motil Cytoskel 1995; 31:225-240.
angiogenesis in collagen gel culture of rat aorta. Athero- 73. Reed MJ, Vernon RB, Abrass IB et al. TGF-!1 induces
sclerosis 1992; 95:191-199. the expession of type I collagen and SPARC, and enhances
49. Nicosia RF, Tuszynski GP. Matrix-bound thrombospondin contraction of collagen gels, by fibroblasts from young and
promotes angiogenesis in vitro. J Cell Biol 1994; aged donors. J Cell Physiol 1994; 158:169-179.
124:183-193. 74. Stopak D, Harris AK. Connective tissue morphogenesis
50. Nicosia RF, Bonanno E, Smith M et al. Modulation of by fibroblast traction. Dev Biol 1982; 90:383-398.
angiogenesis in vitro by laminin-entactin complex. Dev 75. Vernon RB, Sage EH. Contraction of fibrillar type I col-
Biol 1994; 164:197-206. lagen by endothelial cells:A study in vitro. J Cell Biochem
51. Diglio CA, Grammas P, Giacomelli F et al. Angiogenesis 1996; 60:185-197.
in rat aorta ring explant cultures. Lab Invest 1989; 76. Guido S, Tranquillo RT. A methodology for the system-
60:523-531. atic and quantitative study of cell contact guidance in
52. Montesano R, Orci L. Tumor-promoting phorbol esters oriented collagen gels. J Cell Sci 1993; 105:317-331.
induce angiogenesis in vitro. Cell 1985; 42:469-477. 77. Markwald RR, Fitzharris TP, Bolender DL et al. Struc-
53. Montesano R, Orci L. Phorbol esters induce angiogenesis tural analysis of cell:matrix association during the mor-
in vitro from large-vessel endothelial cells. J Cell Physiol phogenesis of atrioventricular cushion tissue. Dev Biol
1987; 130:284-291. 1979; 69:634-654.
54. Montesano R, Pepper MS, Orci L. Paracrine induction of 78. Fournier N, Doillon CJ. In vitro angiogenesis in fibrin
angiogenesis in vitro by Swiss 3T3 fibroblasts. J Cell Sci matrices containing fibronectin or hyaluronic acid. Cell
1993; 105:1013-1024. Biol Int Rep 1992; 16:1251-1263.
55. Fisher C, Gilbertson-Beadling S, Powers EA et al. 79. Manoussaki D, Lubkin SR, Vernon RB et al. A mechani-
Interstital collagenase is required for angiogenesis in vitro. cal model for the formation of vascular networks in vitro.
Dev Biol 1994; 162:499-510. Acta Biotheoret 1996; 44:271-282.
56. Mitchison TJ, Cramer LP. Actin-based cell motility and 80. Izzard CS, Lochner LR. Formation of cell-to-substrate
cell locomotion. Cell 1996; 84:371-379. contacts during fibroblast motility: An interference
57. Bard JBL, Hay ED. The behavior of fibroblasts from the reflexion study. J Cell Sci 1980; 42:81-116.
developing avian cornea. J Cell Biol 1975; 67:400-418. 81. Regen CM, Horwitz AF. Dynamics of !1 integrin-medi-
58. Notbohm H, Mosler S, Müller PK. In vitro formation and ated adhesive contacts in motile fibroblasts. J Cell Biol
aggregation of heterotypic collagen i and III fibrils. Int J 1992; 119:1347-1359.
Biol Macromol 1993; 15:299-304. 82. DiMilla PA, Barbee K, Lauffenburger DA. Mathematical
59. MacBeath JRE, Shackelton DR, Hulmes DJS. Tyrosine-rich model for the effects of adhesion and mechanics on cell
acidic matrix protein (TRAMP) accelerates collagen fibril migration speed. Biophys J 1991; 60:15-37.
formation in vitro. J Biol Chem 1993; 268:19826-19832.
340 Tissue Engineering of Prosthetic Vascular Grafts
83. Goodman SL, Risse G, von der Mark K. The E8 102. Taraboletti G, Roberts D, Liotta LA et al. Platelet
subfragment of laminin promotes locomotion of myo- thrombospondin modulates endothelial cell adhesion,
blasts over extracellular matrix. J Cell Biol 1989; motility, and growth:A potential angiogenesis regulatory
109:799-809. factor. J Cell Biol 1990; 111 765-772.
84. DiMilla P, Stone JA, Quinn JA et al. Maximal migration 103. Tolsma SS, Volpert OV, Good DJ et al. Peptides derived
of human smooth muscle cells on fibronectin and type from two separate domains of the matrix protein
IV collagen occurs at an intermediate attachment strength. thrombospondin-1 have antiangiogenic activity. J Cell Biol
J Cell Biol 1993; 122:729-737. 1993; 122:497-511.
85. Macarak EJ, Howard PS. Adhesion of endothelial cells to 104. Canfield AE, Schor AAM. Evidence that tenascin and
extracellular matrix proteins. J Cell Physiol 1983; thrombospondin-1 modulate sprouting of endothelial
116:76-86. cells. J Cell Sci 1995; 108:797-809.
86. Herbst TJ, McCarthy JB, Tsilibary EC et al. Differential 105. Hsu SC, Volpert OV, Steck PA et al. Inhibition of angio-
effects of laminin, intact type IV collagen, and specific genesis in human glioblastomas by chromosome 10 in-
domains of type IV collagen on endothelial cell adhesion duction of thrombospondin-1. Cancer Res 1996;
and migration. J Cell Biol 1988; 106:1365-73. 56:5684-5691.
87. Kinsella MG, Wight TN. Modulation of sulfate 106. Tuszynski GP, Nicosia RF. The role of thrombospondin-
proteoglycan synthesis by bovine aortic endothelial cells 1 in tumor progression and angiogenesis. BioEssays 1996;
during migration. J Cell Biol 1986; 102:679-687. 18:71-76.
88. Wight TN, Kinsella MG, Quarnström EE. The role of 107. Higuchi M, Ohnishi T, Arita N et al. Expression of
proteoglycans in cell adhesion, migration, and prolifera- tenascin in human gliomas:Its relation to histological
tion. Curr Opin Cell Biol 1992; 4:793-801. malignancy, tumor dedifferentiation and angiogenesis.
89. Sage EH, Bornstein P. Extracellular proteins that modu- Acta Neuropathol 1993; 85:481-487.
late cell-matrix interactions. SPARC, tenascin, and 108. Zagzag D, Friedlander DR, Miller DC et al. Tenascin ex-
thrombospondin. J Biol Chem 1991; 266:14831-14834. pression in astrocytomas correlates with angiogenesis.
90. Bornstein P. Diversity of function is inherent in Cancer Res 1995; 55:907-914.
matricellular proteins:An appraisal of thrombospondin 1. 109. Clezardin P, Malaval L, Ehrensperger A et al. Complex
J Cell Biol 1995; 130:503-506. formation of human thrombospondin with osteonectin.
91. Lane TF, Sage EH. The biology of SPARC, a protein that Eur J Biochem 1988; 175:275-284.
modulates cell-matrix interactions. FASEB J 1994; 110. Sage H, Vernon RB, Funk SE et al. SPARC, a secreted
8:163-173. protein associated with cellular proliferation, inhibits cell
92. Roberts DD. Regulation of tumor growth and metastasis spreading in vitro and exhibits Ca+2-dependent binding
by thrombospondin. FASEB J 1996; 10:1183-1191. to the extracellular matrix. J Cell Biol 1989; 109:341-356.
93. Erickson HP. Tenascin-C, tenascin-R, and tenascin-X: A 111. Maurer P, Hohenadl C, Hohenester E et al. The C-termi-
family of talented proteins in search of functions. Curr nal portion of BM-40 (SPARC/osteonectin) is an autono-
Opin Cell Biol 1993; 5:869-876. mously folding and crystallisable domain that binds cal-
94. Murphy-Ullrich JE, Lane TF, Pallero MA et al. SPARC cium and collagen iv. J Mol Biol 1995; 253:347-357.
mediates focal adhesion disassembly in endothelial cells 112. Yost JC, Sage EH. Specific interaction of SPARC with
through a follistatin-like region of the Ca2+-binding EF- endothelial cells is mediated through a carboxyl-terminal
hand. J Cell Biochem 1995; 57:341-350. sequence containing a calcium-binding EF hand. J Biol
95. Murphy-Ullrich JE, Höök M. Thrombospondin modulates Chem 1993; 268:25790-25796.
focal adhesions in endothelial cells. J Cell Biol 1989; 113. Lightner VA, Erickson HP. Binding of hexabrachion
109:1309-1319. (tenascin) to the extracellular matrix and substratum and
96. Murphy-Ullrich JE, Lightner VA, Aukhil I et al. Focal its effect on cell adhesion. J Cell Sci 1990; 95:263-277.
adhesion integrity is downregulated by the alternatively 114. Joshi P, Chung C-Y, Aukhil I et al. Endothelial cells ad-
spliced domain of human tenascin. J Cell Biol 1991; here to the RGD domain and the fibrinogen-like termi-
115:1127-1136. nal knob of tenascin. J Cell Sci 1993; 106:389-400.
97. Iruela-Arispe ML, Lane TF, Redmond D et al. Expression 115. Sriramarao P, Mendler M, Bourdon MA. Endothelial cell
of SPARC during development of the chicken chorioal- attachment and spreading on human tenascin is medi-
lantoic membrane:Evidence for regulated proteolysis in ated by #2 - ! 1 and # v- ! 3 integrins. J Cell Sci 1993;
vivo. Mol Biol Cell 1995; 6:327-343. 105:1001-1012.
98. Iruela-Arispe ML, Porter P, Bornstein P et al. 116. Yokosaki Y, Palmer EL, Prieto AL et al. The integrin #9!1
Thrombospondin-1, an inhibitor of angiogenesis, is regu- mediates cell attachment to a non-RGD site in the third
lated by progesterone in the human endometrium. J Clin fibronectin type III repeat of tenascin. J Biol Chem 1994;
Invest 1996; 97:403-412. 269:26691-26696.
99. Zagzag D, Friedlander DR, Dosik J et al. Tenascin-C ex- 117. Aukhil I, Joshi P, Yan Y et al. Cell- and heparin-binding
pression by angiogenic vessels in human astrocytomas and domains of the hexabrachion arm identified by tenascin
by human brain endothelial cells in vitro. Cancer Res expression proteins. J Biol Chem 1993; 268:2542-2553.
1996; 56:182-189. 118. Chung C-Y, Erickson HP. Cell surface annexin II is a high
100. Jendraschak E, Sage EH. Regulation of angiogenesis by affinity receptor for the alternatively spliced segment of
SPARC and angiostatin:Implications for tumor cell biol- tenascin-C. J Cell Biol 1994; 126:539-548.
ogy. Semin Cancer Biol 1996; 7:139-146. 119. Chiquet-Ehrismann R. Inhibition of cell adhesion by anti-
101. Vischer P, Volker W, Schmidt A et al. Association of adhesive molecules. Curr Opin Cell Biol 1995; 7:715-719.
thrombospondin of endothelial cells with other matrix 120. Juliano RL, Haskill S. Signal transduction from the extra-
proteins, attachment sites and migration tracks. Eur J Cell cellular matrix. J Cell Biol 1993; 120:577-585.
Biol 1988; 47:36-46.
How Does Extracellular Matrix Control Capillary Morphogenesis? 341
121. Sastry SK, Horwitz AF. Integrin cytoplasmic 140. Brooks PC, Stromblad S, Klemke R et al. Antiintegrin
domains:Mediators of cytoskeletal linkages and extra- and #v!3 blocks human breast cancer growth and angiogen-
intracellular initiated transmembrane signaling. Curr Opin esis in human skin. J Clin Invest 1995; 96:1815-1822.
Cell Biol 1993; 5:819-831. 141. Friedlander M, Brooks PC, Shaffer RW et al. Definition
122. Schwartz MA, Schaller MD, Ginsberg MH. of two angiogenic pathways by distinct #v integrins. Sci-
Integrins:Emerging paradigms of signal transduction. ence 1995; 270:1500-1502.
Annu Rev Cell Dev Biol 1995; 11:549-599. 142. Drake CJ, Cheresh DA, Little CD. An antagonist of
123. Clark EA, Brugge JS. Integrins and signal transduction integrin av!3 prevents maturation of blood vessels dur-
pathways:The road taken. Science 1995; 268:233-239. ing embryonic neovascularization. J Cell Sci 1995;
124. Sastry SK, Horwitz AF. Adhesion-growth factor interac- 108:2655-2661.
tions during differentiation:An integrated biological re- 143. Brooks PC, Montgomery AMP, Rosenfeld M et al. Integrin
sponse. Dev Biol 1996; 180:455-467. #v!3 antagonists promote tumor regression by inducing
125. Dedhar S, Hannigan GE. Integrin cytoplasmic interactions apoptosis of angiogenic blood vessels. Cell 1994;
and bidirectional transmembrane signalling. Curr Opin 79:1157-1164.
Cell Biol 1996; 8:657-669. 144. Vernon RB, Lane TF, Angello JC et al. Adhesion, shape,
126. Plopper GE, McNamee HP, Dike LE et al. Convergence proliferation, and gene expression of mouse Leydig cells
of integrin and growth factor receptor signaling pathways are influenced by extracellular matrix in vitro. Biol Reprod
within the focal adhesion complex. Mol Biol Cell 1995; 1991; 44:157-170.
6:1349-1365. 145. Roskelley CD, Bissell MJ. Dynamic reciprocity revisited:A
127. Languino LR, Gehlsen KR, Wayner E et al. Endothelial continuous, bidirectional flow of information between
cells use #2!1 integrin as a laminin receptor. J Cell Biol cells and the extracellular matrix regulates mammary epi-
1989; 109:2455-2462. thelial cell function. Biochem Cell Biol 1995; 73:391-397.
128. Underwood PA, Bennett FA, Kirkpatrick A et al. Evidence 146. Schwartz MA, Lechene C, Ingber DE. Insoluble fibronectin
for the location of a binding sequence for the # 2 ! 1 activates the Na+/H+ antiporter by clustering and immo-
integrin of endothelial cells, in the !1 subunit of laminin. bilizing integrin #5!1, independent of cell shape. Proc
Biochem J 1995; 309:765-771. Natl Acad Sci USA 1991; 88:7849-7853.
129. Zutter MM, Santoro SA. Widespread histologic distribu- 147. McNamee HP, Ingber DE, Schwartz MA. Adhesion to
tion of the #2!1 integrin cell-surface collagen receptor. fibronectin stimulates inositol lipid synthesis and enhances
Am J Pathol 1990; 137:113-120. PDGF-induced inositol lipid breakdown. J Cell Biol 1993;
130. Leavesley DI, Schwartz MA, Rosenfeld M et al. Integrin 121:673-678.
!1- and !3-mediated endothelial cell migration is triggered 148. Hansen LK, Mooney DJ, Vacanti JP et al. Integrin bind-
through distinct signaling mechanisms. J Cell Biol 1993; ing and cell spreading on extracellular matrix act at dif-
121:163-170. ferent points in the cell cycle to promote hepatocyte
131. Jackson CJ, Knop A, Giles I et al. VLA-2 mediates the growth. Mol Biol Cell 1994; 5:967-975.
interaction of collagen with endothelium during in vitro 149. Ingber DE, Prusty D, Sun Z et al. Cell shape, cytoskeletal
vascular tube formation. Cell Biol Internat 1994; mechanics, and cell cycle control in angiogenesis. J Bio-
18:859-867. mechanics 1995; 28:1471-1484.
132. Davis GE, Camarillo CW. Regulation of endothelial cell 150. Ingber DE, Dike L, Hansen L et al. Cellular
morphogenesis by integrins, mechanical forces, and ma- tensigrity:Exploring how mechanical changes in the cy-
trix guidance pathways. Exp Cell Res 1995; 216:113-23. toskeleton regulate cell growth, migration, and tissue pat-
133. Bauer J, Margolis M, Schreiner C et al. In vitro model of tern during morphogenesis. Int Rev Cytol 1994;
angiogenesis using a human endothelium-derived perma- 150:173-224.
nent cell line:Contributions of induced gene expression, 151. Banes AJ, Tsuzaki M, Yamamoto J et al. Mechanorecep-
G-proteins, and integrins. J Cell Physiol 1992; tion at the cellular level: The detection, interpretation, and
153:437-449. diversity of responses to mechanical signals. Biochem Cell
134. Drake CJ, Davis LA, Little CD. Antibodies to !1-integrins Biol 1995; 73:349-365.
cause alterations of aortic vasculogenesis, in vivo. Dev 152. Penman S. Rethinking cell structure. Proc Natl Acad Sci
Dynam 1992; 193:83-91. USA 1995; 92:5251-5257.
135. Brooks PC, Clark RAF, Cherish DA. Requirement of vas- 153. Maniotis AJ, Chen CS, Ingber DE. Demonstration of
cular integrin # v ! 3 for angiogenesis. Science 1994; mechanical connections between integrins, cytoskeletal
264:569-571. filaments, and nucleoplasm that stabilize nuclear struc-
136. Klein S, Giancotti FG, Presta M et al. Basic fibroblast ture. Proc Natl Acad Sci USA 1997; 94:849-854.
growth factor modulates integrin expression in microvas- 154. Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer me-
cular endothelial cells. Mol Biol Cell 1993; 4:973-982. tastasis and angiogenesis: An imbalance of positive and
137. Enenstein J, Walehm NS, Kramer RH. Basic FGF and negative regulation. Cell 1991; 64:327-336.
TGF-! differently modulate integrin expression of human 155. Moses MA, Sudhalter J, Langer R. Identification of an
microvascular endothelial cells. Exp Cell Res 1992; inhibitor of neovascularization from cartilage. Science
203:499-503. 1990; 248:1408-1410.
138. Varner JA, Brooks PC, Cheresh DA. The integrin av!3: 156. Montesano R, Pepper MS, Vassalli J-D et al. Phorbol es-
Angiogenesis and apoptosis. Cell Adhes Commun 1995; ter induces cultured endothelial cells to invade a fibrin
3:367-374. matrix in the presence of fibrinolytic inhibitors. J Cell
139. Enenstein J, Kramer RH. Confocal microscopic analysis Physiol 1987; 132:509-516.
of integrin expression on the microvasculature and its 157. Mignatti P, Tsuboi R, Robbins E et al. In vitro angiogen-
sprouts in the neonatal foreskin. J Invest Dermatol 1994; esis on the human amniotic membrane: Requirement of
103:381-386.
342 Tissue Engineering of Prosthetic Vascular Grafts
basic fibroblast growth factor-induced proteinases. J Cell 170. Hekman CM, Loskutoff DJ. Endothelial cells produce a
Biol 1989; 108:671-682. latent inhibitor of plasminogen activators that can be ac-
158. Miller EJ, Harris ED, Chung E et al. Cleavage of type II tivated by denaturants. J Biol Chem 1985;
and III collagens with mammalian collagenase:Site of 260:11581-11587.
cleavage and primary structure at the NH2-terminal por- 171. Pyke C, Kristensen P, Ralfkiaer E et al. The plasminogen
tion of the smaller fragment released from both collagens. activation system in human colon cancer: Messenger RNA
Biochem 1976; 15:787-792. for the inhibitor PAI-1 is located in endothelial cells in
159. Dixit SN, Mainardi CL, Seyer JM et al. Covalent struc- the tumor stroma. Cancer Res 1991; 51:4067-4071.
ture of collagen:Amino acid sequence of #2-CB5 of chick 172. Conforti G, Dominguez-Jimenez C, Ronne E et al. Cell-
skin collagen containing the animal collagenase cleavage surface plasminogen activation causes a retraction of in
site. Biochem 1979; 18:5416-5422. vitro cultured human umbilical vein endothelial cell
160. Stetler-Stevenson WG. Type IV collagenases in tumor in- monolayer. Blood 1994; 83:994-1005.
vasion and metastasis. Cancer Metast Rev 1990; 9:289-303. 173. Vassalli J-D, Baccino D, Belin D. A cellular binding site
161. Mignatti P, Rifkin DB, Welgus HG et al. Proteinases and for the Mr 55,000 form of the human plasminogen acti-
tissue remodeling. In:Clark RAF, ed. Molecular and Cel- vator, urokinase. J Cell Biol 1985; 100:86-92.
lular Biology of Wound Repair. Second Edition. New 174. Miles LA, Dahlberg CM, Plescia J et al. Role of cell-sur-
York:Plenum Press, 1996:427-474. face lysines in plasminogen binding to cells: Identifica-
162. Wilhelm SM, Collier IE, Kronberger A et al. Human skin tion of alpha-enolase as a candidate plasminogen recep-
fibroblast stromelysin:Structure, glycosylation, substrate tor. Biochem 1991; 30:1682-1691.
specificity, and differential expression in normal and tu- 175. Plow EF, Miles LA. Plasminogen receptors in the media-
morigenic cells. Proc Natl Acad Sci USA 1987; tion of pericellular proteolysis. Cell Differ Dev 1990;
84:6725-6729. 32:293-298.
163. Aimes RT, Quigley JP. Matrix metalloproteinase-2 is an 176. Cubellis MV, Wun TC, Blasi F. Receptor-mediated inter-
interstitial collagenase. J Biol Chem 1995; 270:5872-5876. nalization and degradation of urokinase is caused by its
164. Moscatelli D, Jaffe E, Rifkin DB. Tetradecanoyl phorbol specific inhibitor PAI-1. EMBO J 1990; 9:1079-1085.
acetate stimulates latent collagenase production by cul- 177. Fibbi G, Ziche M, Morbidelli L et al. Interaction of uroki-
tured human endothelial cells. Cell 1980; 20:343-351. nase with specific receptors stimulates mobilization of
165. Moscatelli D, Presta M, Rifkin DB. Purification of a fac- bovine adrenal capillary endothelial cells. Exp Cell Res
tor from human placenta that stimulates capillary endot- 1988; 179:385-395.
helial cell protease production, DNA synthesis, and mi- 178. Lu H, Mabilat C, Yeh P et al. Blockage of urokinase re-
gration. Proc Natl Acad Sci USA 1986; 83:2091-2095. ceptor reduces in vitro the motility and the deformability
166. Herron GS, Werb Z, Dwyer K et al. Secretion of of endothelial cells. FEBS Lett 1996; 380:21-24.
metalloproteinases by stimulated capillary endothelial cells. 179. Wei Y, Lukashev M, Simon DI et al. Regulation of integrin
I. Production of procollagenase and prostromelysin ex- function by the urokinase receptor. Science 1996;
ceeds expression of proteolytic activity. J Biol Chem 1986; 273:1551-1555.
261:2810-2813. 180. Hasselaar P, Loskutoff DJ, Sawdey M et al. SPARC in-
167. Lewalle JM, Munaut C, Pichot B et al. Plasma membrane- duces the expression of type I plasminogen activator in-
dependent activation of gelatinase a in human vascular hibitor in cultured bovine aortic endothelial cells. J Biol
endothelial cells. J Cell Physiol 1995; 165:475-483. Chem 1991; 266:13178-13184.
168. Pepper MS, Montesano R, Mandriota SJ et al. 181. Montesano R, Pepper MS, Möhle-Steinlein U et al. In-
Angiogenesis:A paradigm for balanced extracellular pro- creased proteolytic activity is responsible for the aberrant
teolysis during cell migration and morphogenesis. Enzyme morphogenetic behavior of endothelial cells expressing the
Prot 1996; 49:138-162. middle T oncogene. Cell 1990; 62:435-445.
169. Mignatti R, Rifkin DB. Plasminogen activators and an-
giogenesis. Curr Topics Microbiol Immunol 1996;
213:33-50.
Facilitation of Healing
Matrix Modulation
CHAPTER 31
Collagen Matrices Attenuate Fibroblast Response to TGF-!
Richard R. Clark, John M. McPherson
Introduction
F ollowing loss of soft tissue, fibroblasts proliferate and produce an initially loose-weave
provisional matrix which is heavily vascularized and which contains fibronectin, hyalur-
onate, and relatively little collagen.1 Gradually this cell-rich granulation tissue becomes a
predominantly collagen-rich tissue. The ultimate outcome of this biological process is an
imperfect, yet expedient repair called scar formation. The rate and extent of repair depends,
in part, on the net balance of negative and positive modulatory signals that impinge on
fibroblasts at such sites.2
Fibroblasts’ responses to various soluble mediators, e.g., growth factors and cytokines,
have for the most part been studied with cells grown on tissue culture plastic.2-5 However, in
vivo fibroblasts are surrounded by an extracellular matrix (ECM), and ECM can have pro-
found effects on cell function.6-12 Thus the responsiveness of fibroblasts to growth factors
and cytokines has been evaluated when the cells were cultured on or in various ECM com-
ponents, especially collagen.13 Such studies provide important insights into the integration
of mediator and ECM signals on fibroblasts.11-13
Transformtin growth factor-! (TGF-!), a potent multifunctional cell regulatory pro-
tein factor,5 induces increased fibroblast synthesis of collagen and fibronectin when the fi-
broblasts are grown on tissue culture plastic.14-16 Since fibroblasts are usually surrounded by
a collagen matrix in vivo, determination of the response of fibroblasts cultured in collagen
gels to TGF-! would help elucidate the action of this factor in vivo.
It has become evident that the fibroblast phenotype that develops as a consequence of
collagen matrix differs dramatically depending on whether the matrix is relaxed, as occurs
in a floating collagen gel, or under tension, as occurs in an anchored collagen gel.13 Cells in
a relaxed collagen gel become stellate, mitogenically quiescent, and produce little col-
lagen.9,17-20 These are the morphological, proliferative and synthetic characteristics of nor-
mal dermal fibroblasts. In contrast, fibroblasts in an anchored collagen gel develop an elon-
gated fusiform morphology as they attempt to contract the attached gel; furthermore, they
actively proliferate and synthesize collagen.19-21 These are the morphological, proliferative
and synthetic characteristics of wound fibroblasts which have deposited substantial amounts
of collagen in the granulation tissue, and which have begun to compact the collagen matrix
and contract the wound.1
In this paper, we review evidence that type I procollagen and TGF-! are coexpressed in
fibroblasts of early, collagen-poor granulation tissue, while fibroblasts in collagen-rich granu-
lation tissue lack type I procollagen despite the persistence of TGF-!. Furthermore, human
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
344 Tissue Engineering of Prosthetic Vascular Grafts
dermal fibroblasts cultured in stressed collagen matrices, an B, respectively); however, type 1 procollagen was not appar-
environment that simulates late collagen-rich granulation ent in day 10 fibroblasts despite the persistence of TGF-!
tissue, synthesize little collagen in response to TGF- ! (panels C and D, respectively). Collagen matrix density in-
compared to fibroblasts cultured on tissue culture plastic, creased rapidly from day 5 to day 10 as judged by trichrome-
an environment that simulates early collagen-poor granu- stained histologic sections1 and photometric analysis of
lation tissue. Sirius red stained sections (Fig. 31.4). This led to the hy-
pothesis that the rapidly accumulating collagen matrix mark-
Results edly diminishes the ability of fibroblasts to produce addi-
tional collagen despite the presence of TGF-!.
Synchronous Expression of Type I Procollagen To test this hypothesis, human dermal fibroblasts were
and TGF-! in Wound Fibroblasts cultured in conditions to simulate early, collagen-poor
of Early Collagen-Poor Granulation Tissue granulation tissue or late, collagen-rich granulation tissue.
Fibroblasts in 5 day full thickness porcine wounds were Fibroblasts cultured in plastic wells are closely juxtaposed
examined as noted in Figure 31.1 for expression of type I to one another and produce abundant amounts of
procollagen and TGF-! using a murine monoclonal anti- fibronectin, similarly to fibroblasts in early granulation tis-
type I procollagen22 or a polyclonal rabbit anti-TGF-!,23 re- sue.24 Fibroblasts cultured in a stressed collagen gel, i.e., the
spectively. The two proteins demonstrated coordinate in- gel remains attached to a plastic well, simulate 10 day, col-
creasing expression from the wound edge to the wound cen- lagen-rich granulation tissue, since in both situations fibro-
ter (Fig. 31.2). No type I procollagen or TGF-! appeared in blasts form tension across a dense collagen matrix.1,20
cells near the wound edge (panels A and D, respectively),
expression occurred in some fibroblasts between the wound Collagen Matrix Attenuates the Ability of TGF-!
edge and wound center (panels B and E, respectively), and Stimulated Fibroblasts to Make Collagen
in many fibroblasts at the wound center (panels C and F, TGF-! stimulation of collagen synthesis was attenu-
respectively). ated when neonatal human dermal fibroblasts were cultured
in attached collagen gels compared to tissue culture plastic
Discordant Expression of Type I Procollagen and TGF-! (Fig. 31.5A). In contrast, TGF-! stimulation of noncollagen
in Wound Fibroblasts of Late Collagen-Rich synthesis was not substantially affected by the collagen ma-
Granulation tissue trix environment (Fig. 31.5B). The apparent TGF-! stimu-
When day 7 and day 10 wounds were probed for type I lation of noncollagen protein synthesis was largely attribut-
procollagen and TGF-! (Fig. 31.3), both proteins appeared able to a 2-fold increase in the specific activity of intracellu-
in most, if not all, wound fibroblasts at day 7 (panels A and lar proline pools (control 19,900 dpm/nmole; 100 pM TGF-!,
Fig. 31.1. A schematic of a 5 day old cutaneous wound in the flank of a Yorkshire pig. The boxes with
capital letters represent the areas of the wound shown in the photomicrographs illustrated in Fig. 31.2.
e = epidermis, d = dermis, g = granulation tissue, ft = fat, f = fibrous septa.
Collagen Matrices Attenuate Fibroblast Response to TGF-! 345
38,900 dpm/nmole) as previously reported.15 Parenthetically, Five lines of evidence suggest that attenuation was not
TGF-! did not increase the total intracellular proline pool a trivial event. First, [14C]-labeled mixed amino acids and
(control, 1.62 nmole/∝g DNA; 100 pM TGF-!, 1.68 nmole/∝g albumin freely diffused out of collagen gels, obtaining an
DNA). Attached collagen matrix culture environment also equilibrium between the gels and the medium within 30
consistently attenuated the collagen synthetic response of minutes. Second, total DNA of cells cultured for 3 days in
adult human dermal fibroblasts to TGF- !, while the collagen gels was not appreciably different than the DNA of
noncollagen synthetic response was not affected (Table 31.1). cells cultured on plastic (Table 31.3). Third, LDH release
Four strains of fibroblasts were examined, two in duplicate from cells cultured 3 days on plastic or in collagen was not
experiments. Although marked differences were noted in appreciably different and was always less than 5% of the to-
TGF-! response among these strains, all strains demon- tal LDH. Fourth, basal collagen synthesis of fibroblasts cul-
strated attenuation of their TGF-! response when grown in tured in collagen gels was usually similar to basal collagen
collagen gels. In contrast, attached fibrin gels did not alter synthesis of fibroblasts cultured on tissue culture plastic
the collagen synthetic response to TGF-! (Table 31.2). (Table 31.4). Fifth, TGF-! equally stimulated noncollagen
synthesis of fibroblasts cultured in collagen gels or on tissue
346 Tissue Engineering of Prosthetic Vascular Grafts
Table 31.1. Collagen matrices attenuate the collagen Table 31.2. Fibrin matrices do not attenuate the
synthetic response of adult human dermal fibroblasts to collagen synthetic response of adult human dermal
TGF-!1. fibroblasts to 100pM TGF-!.
Strain # Condition Collagen Noncollagen Collagen Fold Increase Noncollagen Fold Increase
!
synthesis with TGF-! synthesis with TGF-!!
1 TC Plastic 2.1a 1.2 dpm/ug dpm/ug
Collagen gel 1.5 1.4 DNAx10-3 DNAx10-3
2 TC Plastic 4.6 3.0
Collagen gel 2.7 2.5 TC Plastic 16.24±3.03a 87.60±27.13
3 TC Plastic 4.5 3.1 TC Plastic 69.07±30.09 4.3x 314.37±76.75 3.6x
Collagen gel 3.0 1.9 +TGF-!
3 TC Plastic 5.0 3.0 TC Plastic 15.78±9.78 157.14±11.35
Collagen gel 3.8 2.1 Fibrin gel 69.63±15.95 4.4x 539.30±96.25 3.4x
4 Tc Plastic 9.1 3.4 +TGF-!
Collage Gel 1.8 3.6 aMean of 3 replicates ± standard deviations
4 TC Plastic 9.9 3.3
Collagen gel 2.8 2.9
p < 0.004b NS
aFold increase of fibroblasts stimulated with 100pM TGF-b over Beta-aminoproprionitrile did not alter the collagen gel ef-
control
fect (data not shown).
bStatistics were performed on the ranks by the Wilcoxon test
Autoradiographic Analysis of TGF-!1-Stimulated
Proteins
When equal aliquots of medium from each experi-
mental condition shown in Figure 31.5 were analyzed on
culture plastic (Table 31.1). In addition, nonspecific adsorp- SDS-PAGE autoradiographs (Fig. 31.7A), most protein
tion of TGF-! to the collagen matrix was not observed. TGF-! bands were notably increased by 100, 300, or 1000 pM TGF-!
preincubated for 24 hours in collagen gels or in TC dishes (lanes 2-4, respectively and lanes 6-8, respectively) compared
stimulated collagen synthesis to the same extent when added to no TGF-!1 (lanes 1 and 5). Type I procollagen #1-and
subsequently to fibroblasts cultured on plastic. #2-chains and fibronectin (arrows) were increased to a
TGF-! had no consistent effect on the proportion of greater degree than most other proteins. When fibroblasts
collagen retained in the cell layers of neonatal or adult der- were cultured in collagen gels, the TGF-!-induced increase
mal fibroblasts whether the cells were cultured on plastic or of some protein bands was clearly attenuated (lanes 5-8)
in collagen gels (Fig. 31.6). For reasons that may be biologi- compared to cells cultured in tissue culture plastic (lanes
cally important but are presently unclear, more collagen was 1-4). To resolve whether the attenuated TGF-! response in-
retained in the cell layers of adult fibroblasts cultured in col- volved mostly collagenous or noncollagen proteins, the newly
lagen gels regardless of whether TGF-! was added (Fig. 31.6). synthesized proteins were digested with either pepsin or col-
lagenase, respectively, prior to SDS-PAGE.
Collagen Matrices Attenuate Fibroblast Response to TGF-! 347
Increased collagen synthesis induced by TGF-!1 was 300, 1000 pM TGF-!1 (lanes 2-4 and lanes 6-8) compared
best demonstrated by SDS autoradiography of medium to no TGF-!1 (lanes 1 and 5); however, the TGF-! response
samples that had been pepsin digested to remove was markedly attenuated when fibroblasts were cultured in
noncollagen protein.25 The major bands appearing on SDS- collagen gels (lanes 6-8) compared to fibroblasts in tissue
PAGE autoradiography after pepsin digestion were #1(I) and culture plastic wells (lanes 2-4).
#1(III) chains (Fig. 31.7B, upper arrow), and #2(I) chains Using interrupted gel electrophoresis26 and autorad-
(Fig. 31.7B, lower arrow). These bands of collagen proteins iography on pepsin-digested samples, the TGF-! stimula-
were increased when fibroblasts were incubated with 100, tion of #1(I) and #1(III) collagen chains was equally attenu-
348 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 31.6. Effect of TGF-! and collagen matrix on compartmentalization of newly synthesized collagen
between the media and cell layers of neonatal (A) and adult (B) human dermal fibroblasts. Fibroblasts
were cultured on TC plastic or in collagen gels and metabolically labeled with [14C]proline as described
in Fig. 31.5. Collagen production in the culture medium and cell layer was determined using the modi-
fied Peterkofsky method. The amount of [14C]proline incorporated into newly synthesized collagen
was normalized per ∝g DNA. The percent shown in each bar represents the percent collagen retained in
the cell layer.
350 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 31.7. SDS-PAGE autoradiography confirms that collagen matrices attenuate the collagen synthetic response of neonatal
human dermal fibroblasts to TGF-! 1. (A) Total newly synthesized protein in the medium. (B) Newly synthesized pepsin-
resistant protein in the medium. (C,D) Newly synthesized collagenase-resistant protein in the medium and in the cell layer,
respectively. Medium samples from equal cell numbers based on DNA were analyzed by SDS-PAGE autoradiography. Samples
were taken from fibroblasts cultured in the absence of TGF-!1 (lanes 1 and 5), and in the presence of TGF-!1 (100 pM, lanes
2 and 6; 300 pM, lanes 3 and 7; 1000 pM, lanes 4 and 8). Arrows indicate fibronectin (FN), type I procollagen #1 (pro #1) and
#2 (pro #2) and type I collagen #1 and #2.
late (e.g., day 10), granulation tissue in which fibroblasts are environment did not appear to be a trivial effect of the col-
under tension as they reorganize and compact the collagen lagen gels on TGF-! or fibroblasts, as TGF-! was not bound
matrix.1,20 As shown in Figs. 31.4-31.7 and Table 31.1, such by the collagen gels, and collagen gels did not cause nutrient
collagen gels consistently and significantly attenuated the deprivation or cell injury. Furthermore, TGF-! stimulation
collagen synthetic response of fibroblasts to TGF-! compared of noncollagen synthesis was usually not altered by the at-
to fibroblasts cultured on plastic. When cultured on plastic, tached collagen gels used in these experiments (Table 31.1,
fibroblasts grow in close juxtaposition and deposit an abun- Figs. 31.4 and 31.6). Specifically TGF-! stimulation of
dance of fibronectin in the pericellular matrix, a situation fibronectin synthesis appeared unaffected by stressed col-
that simulates early, fibronectin-rich and collagen-poor, lagen gels. Biochemical results were confirmed by immun-
granulation tissue. The attenuation by a collagen matrix ofluorescence studies of fibroblasts (Fig. 31.8). Most if not
Collagen Matrices Attenuate Fibroblast Response to TGF-! 351
all fibroblasts cultured on plastic appeared to respond to if any, when fibroblasts are cultured in attached collagen gels
TGF-! by expressing perinuclear granular staining for type confirm the studies from the Grinnell laboratory.19-21 Al-
I procollagen (Fig. 31.8B), while most cells in collagen were though the attached collagen gel paradigm predicts that fi-
negative even after TGF-! stimulation (Fig. 31.8D). Never- broblasts under tension as observed in 10 day, collagen-rich
theless, an occasional cell in collagen stained at least as in- wounds would not show a decrease in basal collagen syn-
tense for type I procollagen as the TGF-! stimulated fibro- thesis1 ( Fig. 31.1), our studies clearly demonstrate that fi-
blasts cultured on plastic (Fig. 31.8D). Thus, there appeared broblasts in these conditions do lose their responsiveness to
to be a marked heterogeneity of cultured fibroblasts in their TGF-!. Recently we have found that while PKC-3 and Nf-∀b
ability to respond to collagen matrix. are activated by both stressed and relaxed collagen matrices,
The mechanism by which collagen matrix alters the other signal transduction pathways are, however, affected
responsiveness of fibroblasts to TGF-! is not known. One differentially by these two physical forms of collagen matrix
possibility is that the collagenous environment (unpublished data).
downregulates one or more of the three known cell surface The concept of collagen matrix attenuation of fibro-
receptors for TGF-!.37,38 In fact, given that we have observed blast collagen synthesis to TFG-! is appealing and is consis-
a differential effect of collagen gels on the biosynthesis of tent with previous results which showed that collagen ma-
collagen and fibronectin in response to TGF-! (Fig. 31.6), trices downregulated the responsiveness of fibroblasts to
collagen matrix may differentially affect these receptors. As PDGF and other growth factors.42,45 These data provide in
a first step toward investigating these possibilities, we probed some measure an explanation for the orderly progression of
RNA extracts of human adult dermal fibroblasts for type II the wound repair process by a means of positive signals that
and type III TGF-! receptor expression when fibroblasts were stimulate cells to generate an environment that both pro-
grown in plastic tissue culture dishes verses collagen matri- vides for repair and establishes feed-back regulation of the
ces. Using cDNA probes for the types II and III receptors process.
obtained from Drs. Herbert Lin, Harvey Lodish, Robert In contrast to the suppression effects of collagen gels
Weinberg, and Xiao-Fan Wang,39,40 we found no detectable on collagen synthesis, the permissive effects of fibrin gels
differences in mRNA expression among these conditions. are consistent with the role of this molecule in providing a
Nevertheless, at least one study has shown that the extracel- provisional matrix during the early phases of the wound
lular matrix environment can modulate TGF-! receptor ex- repair to support cell invasion, proliferation and matrix
pression with remarkable effects on cell responses to TGF-!.41 deposition. Thus the normal process of wound repair ap-
Alternatively, collagen matrix may influence signal pears to be under the control of both soluble factors that
transduction42 and this may influence, perhaps differentially, stimulate certain events and the extracellular matrix, which
the biosynthesis of ECM molecules. The pathway(s) through can modulate the responsiveness of cells to these signals. It
which collagen matrix transmits its effect on fibroblasts are is likely that certain pathological fibrotic conditions develop
currently a focus of study in our laboratory. It is clear that when these feedback regulation systems become ineffective.
collagen matrix activates protein kinase-3 (PKC-3) and
Nf-∀b, which leads to upregulation of #2!1 collagen recep- Acknowledgments
tors;43,44 however, the relationship of these pathways to col- Funding for this work was provided by NIH grants
lagen matrix attenuation of TGF-!-stimulated collagen syn- HL27353 and AM31514 to Richard A. F. Clark.
thesis is not known.
An interesting byproduct of this study was the failure References
of attached collagen gels to suppress basal collagen synthe- 1. Welch MP, Odland GF et al. Temporal relationships of
sis (Table 31.4). Previously, many investigators reported that F-actin bundle formation, collagen and fibronectin ma-
basal collagen synthesis was suppressed by collagen trix assembly, and fibronectin receptor expression to
wound contraction. J Cell Biol 1990; 110:133-145.
gels.17,18,20 However, in the studies reporting suppression of
2. Clark RAF. Overview and general considerations of wound
basal collagen synthesis, collagen gels were detached from repair. In: Clark RAF, Henson PM, eds. The Molecular
the tissue culture dish, allowing the fibroblasts to and Cellular Biology of Wound Repair. New York: Ple-
isotropically contract the collagen matrix under relatively num Press, 1996:3-50.
little tension. Ultimately the fibroblasts in floating collagen 3. Rifkin DB, Moscatelli D. Recent developments in the cell
gels assume a stellate phenotype similar to fibroblasts in biology of basic fibroblast growth factor. J Cell Biol 1989;
normal dermis.20 Thus the released collagen gel model pre- 109:1-6.
dicts that fibroblasts in normal skin may be maximally in- 4. Ross RR, Raines EW. Platelet-derived growth factor and
hibited by their surrounding collagen matrix. In contrast, as cell proliferation. In: Sara VR et al, eds. Growth Factors:
fibroblasts in attached collagen gels attempt to contract the From Genes to Clinical Application. New York: Raven
matrix under these anisotropic conditions, they generate Press, 1990; 193-199.
5. Sporn MB, Roberts AM. Transformtin growth factor-!:
increasing tension and assume a bipolar phenotype along
Recent progress and new challenges. J Cell Biol 1992;
the lines of maximum resistance.20 A similar phenomenon 119:1017-1021.
occurs in 10 day collagen-rich, granulation tissue as wound 6. Gospodarowicz D, Delgado D, et al. Permissive effect of
contraction commences.1 Thus, the attached collagen gels the extracellular matrix on cell proliferation in vitro. Proc
in our experiments kept the cells and collagen matrix under Nat Acad Sci USA 1980; 77:4094-4098.
tension similar to 10 day, collagen-rich granulation tissue.1
Our findings that basal collagen synthesis is depressed little,
352 Tissue Engineering of Prosthetic Vascular Grafts
7. Sugrue SP, Hay ED. Response of basal epithelial cell sur- 26. Sykes B, Puddle B et al. The estimation of two collagens
face and cytoskeleton to solubilized extracellular matrix from human dermis by interrupted gel electrophoresis.
molecules. J Cell Biol 1981; 91:45-54. Biochem Biophys Res Comm 1976; 72:1472-1480.
8. Wicha MS, Lowrie G et al. Extracellular matrix promotes 27. Hynes RO, Destree AT. Relationships between fibronectin
mammary epithelial growth and differentiation in vitro. (LETS) protein and actin. Cell 1978; 15:875-886.
Proc Nat Acad Sci USA 1982; 79:3213-3217. 28. Chen W-T, Wang J et al. Regulation of fibronectin re-
9. Mauch C, Adelmann-Grill B et al. Collagenase gene ex- ceptor distribution by transformation, exogenous
pression in fibroblasts is regulated by a three-dimensional fibronectin, and synthetic peptides. J Cell Biol 1986;
contact with collagen. FEBS Lett 1989; 250:301-305. 103:1649-1661.
10. Klein CE, Dressel D et al. Integrin #2!1 is upregulated in 29. Roman J, LaChance RM et al. The fibronectin receptor is
fibroblasts and highly aggressive melanoma cells in three organized by extracellular matrix fibronectin: Implications
demensional collagen lattices and mediates the reorgani- for oncogenic transformation and for cell recognition of
zation of collagen i fibrils. J Cell Biol 1991; 115:1427-1436. fibronectin matrices. J Cell Biol 1989; 108:2529-2543.
11. Damsky CH, Werb Z. Signal transduction by integrin re- 30. Sporn MB, Roberts AB et al. Polypeptide transforming
ceptors for extracellular matrix: Cooperative processing of growth factor isolated from bovine sources and used for
extracellular information. Curr Opin Cell Biol 1992; wound healing in vitro. Science 1983; 219:1329-1331.
4:772-781. 31. Mustoe TA, Pierce GF et al. Accelerated healing of
12. Juliano RL, Haskill S. Signal transduction from the extra- incisional wounds in rats induced by transformtin growth
cellular matrix. J Cell Biol 1992; 120:577-585. factor-beta. Science. 1987; 237:1333-1336.
13. Grinnell F. Fibroblasts, myofibroblasts, and wound con- 32. Sprugel KH, McPherson JM et al. Effects of growth fac-
traction. J Cell Biol 1994; 124:401-404. tors in vivo. Am J Path 1987; 129:601-613.
14. Ignotz RA, Massague J. Transformtin growth factor-! 33. Flanders KC, Thompson NL et al. Transformtin growth
stimulates the expression of fibronectin and collagen and factor-!1: Histochemical localization with antibodies to
their incorporation into extracellular matrix. J Biol Chem different epitopes. J Cell Biol 1989; 108:653-666.
1986; 261:4337-4340. 34. O’Connor-McCourt MD, Wakefield LM. Latent
15. Roberts AB, Sporn MB et al. Transforming growth factor transformtin growth factor-b in serum. J Biol Chem 1987;
beta: Rapid induction of fibrosis and angiogenesis in vivo 262:14090-14099.
and stimulation of collagen formation. Proc Natl Acad Sci 35. Lyons RM, Keski-Oja J et al. Proteolytic activation of la-
USA 1986; 83:4167-4171. tent transformtin growth factor-! from fibroblast-condi-
16. Fine A, Goldstein RH. The effect of transformtin growth tioned medium. J Cell Biol 1988; 106:1659-1665.
factor-! on cell proliferation and collagen formation by 36. Miyazono K, Heldin CH. Role for carbohydrate structures
lung fibroblasts. J Biol Chem 1987; 262:3897-3902. in TGF-!1 latency. Nature 1989; 338:158-160.
17. Nusgens B, Merrill C et al. Collagen biosynthesis by cells 37. Chen R-H, Ebner R et al. Inactivation of the type II re-
in a tissue equivalent matrix in vitro. Coll Relat Res 1984; ceptor reveals two receptor pathways for the diverse TGF-
4:351-363. ! activities. Science 1993; 260:1335-1338.
18. Mauch C, Hatamochi A et al. Regulation of collagen syn- 38. Lopez-Casillas F, Wrana JL et al. Betaglycan presents
thesis in fibroblasts within a three-dimensional collagen ligand to the TGF- ! signaling receptor. Cell 1993;
gel. Exp Cell Res 1988; 178: 493-530. 73:1435-1444.
19. Nakagawa S, Pawelek P et al. Extracellular matrix organi- 39. Wang XF, Lin HY et al. Expression cloning and charac-
zation modulates fibroblast growth and growth factor re- terization of the TGF-beta type III receptor. Cell 1991;
sponsiveness. Exp Cell Res 1989; 182:572-582. 67(4):797-805.
20. Nakagawa S, Pawelek P et al. Long term culture of fibro- 40. Lin HY, Wang XF et al. Expression cloning of the TGF-
blasts in contracted collagen gels: Effects on cell growth beta type II receptor, a functional transmembrane serine/
and biosynthetic activity. J Invest Dermatol 1989; threonine kinase [published erratum appears in Cell Sep
93:792-798. 18;70(6):following 1068].” Cell 68(4): 775-85.
21. Fukamizu H, Grinnell F. Spatial organization of extracel- 41. Sankar S, Mahooti-Brooks N et al. Modulation of trans-
lular matrix and fibroblast activity: Effects of serum, trans- forming growth factor ! receptor levels on microvascular
forming growth factor !, and fibronectin. Exp Cell Res endothelial cells during in vitro angiogenesis. J Clin In-
1990; 190:276-282. vest 1996; 97(6):1436-46.
22. McDonald JA, Broekelmann TJ et al. A monoclonal anti- 42. Lin Y-C, Grinnell F. Decreased level of PDGF-stimulated
body to the carboxyterminal domain of procollagen type receptor autophosphorylation by fibroblasts in mechani-
I visualizes collagen-synthesizing fibroblasts. J Clin Invest cally relaxed collagen matrice. J Cell Biol 1993;
1986; 78:1237-1244. 122:663-672.
23. Ellingsworth LR, Brennan JE et al. Antibodies to the N- 43. Xu J, Clark RAF. Collagen lattices stimulate fibroblast
terminal portion of cartilage-inducing factor A and PKC-3 activation: Role in #2 integrin subunit and colla-
transformtin growth factor-b. Immunohistochemical lo- genase-1 mRNA expression. J Cell Biol 1997; 136:473-483.
calization and association with differentiating cells. J Biol 44. Xu J, Zutter MM et al. A three-dimensional collagen lat-
Chem 1986; 261:12362-12367. tice activates Nf-∀b in human fibroblasts: Role in integrin
24. Xu J, ClarkRAF. Extracellular matrix alters PDGF regula- #2 gene expression and tissue remodeling. J Cell Biol in
tion of fibroblast integrins. J Cell Biol 1996; 132:239-249. press. 1998; 140:709-719.
25. Leibovich SJ, Weiss JB. Electron microscope studies of the 45. McPherson JM, Rhudy RW. Influence of the extracellular
effects of endo and exopeptidase digestion on tropocol- matrix on the proliferative response of human skin fibro-
lagen: A novel concept of role of terminal regions in blasts to serum and purified platelet-derived growth fac-
fibrillogenesis . Biochim Biophys Acta 1970; 214:445-454. tor. J Cell Physiol. 1988; 137:185-191.
Facilitation of Healing
Matrix Modulation
CHAPTER 32
Extracellular Matrix Proteins Are Potent Agonists
of Human Smooth Muscle Cell Migration
Terry L. Kaiura, K. Craig Kent
Introduction
T he current treatments of atherosclerotic occlusive disease are multiple and include by-
pass, endarterectomy, and angioplasty. Unfortunately, the long term success of these
interventions is significantly jeopardized by a process known as intimal hyperplasia.1,2 Tis-
sue engineering and subsequent creation of a prosthetic graft that is resistant to the forma-
tion of an intimal plaque would be instrumental in improving the long term outcome of
vascular reconstructions. In order to synthesize the “ideal” vascular graft, however, it is im-
perative that the critical cellular processes that precede the formation of intimal hyperplasia
be understood.
The three essential components of intimal hyperplasia are smooth muscle cell (SMC)
migration, proliferation, and extracellular matrix (ECM) production.3 Although all three of
these events are necessary for restenosis, their relative importance is not well understood. In
normal human vasculature, the intima is almost completely devoid of SMCs. Thus, migration
of SMC from the vessel media into the subintimal space is a critical initial step in the initiation
of the restenotic process. Confirming the importance of SMC migration are studies that
demonstrate that over 50% of the neointimal plaque is composed of nonproliferating SMC.4
Following vascular injury, SMC are exposed to a variety of growth factors, cytokines,
and ECM proteins, all of which can potentially affect SMC migration. The role of growth
factors in SMC chemotaxis has been studied in great detail and the platelet-derived growth
factor BB homodimer (PDGF-BB) has been identified as an extremely potent agonist of
SMC migration.5 Moreover, a plethora of other growth factors including fibroblast growth
factor (FGF), epidermal growth factor (EGF), and transformtin growth factor-! (TGF-!)
have been found to affect SMC chemotaxis.6,7 ECM proteins are also abundant in the arterial
wall, and SMC are exposed to these proteins following vascular injury as well.8 The role of
ECM proteins in SMC migration, however, is less well understood, and is the focus of
this chapter.
Cell Migration
Cell migration is a dynamic, complex process, the molecular biologic basis of which
has not been fully described. The central component of the cellular apparatus that is neces-
sary for migration is the actin cytoskeleton.9 Actin is organized into stress fibers that create
the framework of a cell. These filaments are anchored by actin binding proteins such as
#-actinin10 to the cell membrane via a series of proteins termed the focal adhesion complex.11
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
354 Tissue Engineering of Prosthetic Vascular Grafts
The focal adhesion complex itself is composed of clusters of Therefore, cellular secretion of proteolytic agents such as
proteins such as tensin, p125 focal adhesion kinase (FAK), matrix metalloproteinase (MMP)-2, MMP-9, tissue type
paxillin, talin, and vinculin, that bind to transmembrane plasminogen activator, and urokinase plasminogen activa-
receptors called integrins (Fig. 32.1).12 Integrin receptors in tor appear to be necessary for cell locomotion.18,19 It has
turn bind via their extracellular domains to surrounding been demonstrated that PDGF increases the expression of
matrix.13 several proteases found in the ECM;20 therefore PDGF may
Cellular migration is a cyclic process that initially re- contribute to cellular migration by acting directly on the
quires detachment of the leading edge of a cell, reorganiza- cytoskeleton or also by modifying the environment that sur-
tion of the actin filaments to allow forward movement, fol- rounds a cell. Heparin, for example, which is a known de-
lowed by reattachment of this “pseudopod” in a new loca- terrent of intimal hyperplasia inhibits expression of tissue
tion.14 The body of the cell is then pulled forward to “catch type plasminogen activator and collagenase.21
up” with the leading edge, resulting in forward movement The migratory response of SMC to an agonist can as-
of the entire cell. This complex process requires that both sume three different patterns, depending upon the state and
focal adhesions and actin filaments be cyclically dissolved solubility of the stimulatory protein. Chemokinesis describes
and then reformed. random cellular migration in response to a soluble factor,
The integrin receptor is central to the interaction be- chemotaxis refers to directed migration toward a positive
tween the cytoskeleton and the external cellular environ- gradient of soluble attractant, and haptotaxis defines directed
ment.15 Integrin receptors have large extracellular domains, cellular migration toward a positive gradient of an insoluble
a single membrane-spanning domain and a short cytoplas- agonist such as a substrate bound matrix protein.21 Whether
mic domain. The extracellular domain attaches to matrix or not matrix proteins are in a soluble or insoluble form at
proteins that are specific for each integrin receptor and the the time of vascular injury is not clear. This may be an im-
cytoplasmic domain interacts with focal adhesions and ac- portant distinction, since the magnitude of the migratory
tin filaments. It is increasingly recognized that integrin- response may vary depending upon whether the stimulus is
matrix interactions are not purely structural or biomechani- that of haptotaxis or chemotaxis. Moreover, it has been
cal, but that integrins transduce signals into cells which may shown that the signaling pathways that mediate these two
be important in the regulation of multiple processes includ- responses are distinct.22 Possibly all three stimuli have the
ing cell motility.16 Thus, integrins are similar to growth fac- potential to influence vascular SMCs during the repair and
tor receptors in that ECM proteins can act as ligands that remodeling process that follows vascular injury.3
stimulate signaling events that ultimately influence cellular
behavior. In the case of migration, integrins not only pro- Extracellular Matrix Proteins
vide the structural mechanism that allows a cell to attach ECM proteins are ubiquitous in the normal vessel wall.
and migrate across ECM, but these same integrin receptors Endothelial cells manufacture an underlying basement mem-
may transduce messages via ECM proteins into the cell that brane that is superficial to the internal elastic lamina. This
influence the propensity of a cell to migrate.17 layer is composed primarily of type IV collagen, laminin,
Lysis of ECM may also be an important component and heparan sulfate proteoglycan.23 The media is composed
of the process of migration. Lysis of these proteins allows a of smooth muscle cells that are surrounded by a plethora of
pathway to be created through which the cell can migrate. ECM proteins that include fibronectin,24 types I, III, and V
collagen,25 as well as elastin,26 and proteoglycans.27 The ad- following vascular injury, its expression rapidly increases.45
ventitia, which consists mainly of collagen, is bound on its Yabkowitz et al have found that TSP is a potent mediator of
outer aspect by loose connective tissue that includes collagen calf pulmonary artery SMC migration.47 Either inhibition
type I and proteoglycans.23 of TSP synthesis or TSP-SMC interactions decreased in re-
ECM comprises as much as 60-80% of a mature inti- sponse to SMC migration. Interestingly, SMC migration to
mal plaque.28,29 The predominant proteins are collagen types the combination of TSP and PDGF was greater than the
I and III,30 elastin,31 fibronectin,32,33 and proteoglycans such maximal response observed with PDGF alone.47
as versican-like chondroitin sulfate.34 Two weeks following The glycosaminoglycan hyaluronan (HA) is
vessel injury and after the period of maximal SMC prolif- synthesized and deposited in the neointima soon after vas-
eration and migration, cell numbers in the intima reach a cular injury.48 Riessen et al found through histological analy-
maximum. The continued expansion of intimal plaque is ses of both rat and human vascular tissues that HA is a
then due to the synthesis of the ECM proteins previously predominant component of the early vascular lesion.48
mentioned.28 A variety of less predominant proteins have Savani et al demonstrated that HA stimulated the migra-
also been identified in intimal plaque including osteo- tion of arterial bovine SMCs, and that an antibody to the
pontin,35 vitronectin,36 thrombospondin,37 and tenascin.38 HA-binding RHAMM receptor blocked migration.49 CD44
These proteins are synthesized and secreted primarily by is another HA binding receptor that is upregulated in the
SMC. The prevalence of ECM in the normal and pathologi- neointima and may also play a role in SMC migration.20
cal vessel has spurred our investigation into the effect of these Moreover, the ability of HA to bind to large amounts of water
proteins on SMC migration. and to create a gel-like environment may also potentially
facilitate SMC migration.48
Extracellular Matrix Protein and Migration Vitronectin, otherwise known as the serum-spread-
In recent years, the effect of a number of ECM pro- ing factor or S-protein is present as an ECM protein in the
teins on SMC migration has been investigated. In the fol- circulation, as well as in the vessel wall.36 This ECM protein
lowing paragraphs, the results of these investigations will be is also increased in atherosclerotic tissues.50 Vitronectin is
reviewed. involved in the adhesion of cells to the ECM and in the regu-
Osteopontin has been widely studied because of its lation of complement and coagulation pathways.51 Naito et
expression in tissues that undergo remodeling, such as the al found that vitronectin invoked SMC haptotaxis and pos-
smooth muscle layer of the intestinal tract and the normal tulated that vitronectin, which is found in the subendothe-
intima of the ductus arteriosus.20 Liaw et al have demon- lial matrix, may stimulate migration of SMCs from the me-
strated that osteopontin is dramatically elevated in the rat dia to the intima.52 Moreover, Brown et al have shown that
carotid artery and aorta following balloon injury.39 Studies vitronectin induces human aortic SMC migration via the
of human vascular tissue have revealed focal expression of #v!3 integrin to an extent that is comparable to that of the
osteopontin in atherosclerotic plaques but not in normal growth factor PDGF-BB.53
human arteries.40 Osteopontin, in vitro, has a strong stimu- Although all of the above described proteins are ex-
latory effect on SMC chemotaxis.41 Moreover, antibodies pressed following arterial injury and promote SMC migra-
directed against osteopontin inhibit SMC migration and also tion, these proteins constitute a relatively small portion of
neointima thickening in the injured rat carotid artery.42 It is the total ECM within both normal and diseased vessels. More
likely that osteopontin works in conjunction with other ECM predominant components of normal artery and vein and
proteins, integrins, and matrix proteases to promote adhe- intimal plaque include proteins such as collagen types I and
sion and migration of SMCs. IV, fibronectin, and laminin.24 The effect of these ECM pro-
Chen et al has shown that the large ECM glycopro- teins on SMC migration has been less well investigated. As a
tein, tenascin, is distributed throughout the neointima of result, our group has recently studied the effect of these four
hyperplastic lesions derived from human arteriovenous proteins on all three mechanisms of migration (namely
polytetrafluoroethylene grafts that have been removed for chemokinesis, chemotaxis, and haptotaxis) using human
revision.43 Tenascin has also been found in the intimal layer smooth muscle cells derived from saphenous veins.
of injured rat aorta and carotid arteries.38 This glycopro- We found that collagen types I and IV, fibronectin,
tein, which has been reported to have both adhesive and anti- and laminin were all very potent agonists of SMC migra-
adhesive domains, is a mediator of focal adhesion disassem- tion.54 In our initial studies, we determined the concentra-
bly as well.44 As such, Majesky has speculated that this dis- tion of each matrix protein that produced maximal cell
bursement of focal adhesions, or sites of cellular attachment movement. Maximal migration was achieved with interme-
to the ECM, may facilitate the transition of a cell from a diate concentrations of fibronectin and laminin. Higher con-
nonmotile to a migrating state.45 Accordingly, there is centrations of these two proteins did not produce a further
evidence that tenascin supports the migration of rat aortic increase in the migratory response. Type I and IV collagen
smooth muscle cells, although the effect of this ECM on similarly produced maximal migration at intermediate con-
human SMC migration has not been investigated.46 centrations of protein; however, higher concentrations of
Similarly to tenascin, thrombospondin (TSP) pos- both collagens led to a decrease in SMC migration. Corrobo-
sesses both adhesive and counteradhesive properties and is rating our findings, DiMilla et al found that high concen-
a prominent facilitator of focal adhesion disassembly.44 TSP trations of type IV collagen increased SMC attachment, but
is present in the ECM of the normal arterial wall; however, decreased SMC migration.55 These authors concluded that
356 Tissue Engineering of Prosthetic Vascular Grafts
cell migration on a matrix protein may be inversely related We found that simultaneous stimulation of SMC with
to the ability of a cell to attach this protein. PDGF-AB and collagen type I or IV, fibronectin, or laminin
To determine the relative potency of these proteins as synergistically enhanced SMC migration to a level that was
stimulants of migration, we studied SMC chemotaxis, greater than the addition of their individual effects.60 At both
haptotaxis, and chemokinesis in response to optimal con- low and high concentrations of the collagens, the chemo-
centrations of each matrix protein.54 We also compared the tactic response was enhanced 20-60% by the simultaneous
relative effect of these matrix proteins to the prototypical stimulation of SMC with PDGF and collagen. Alternatively,
migratory stimulus, PDGF. Collagen types I and IV produced with low concentrations of laminin or fibronectin the re-
the most significant increase in chemotaxis, with a respec- sponse with PDGF was additive, whereas, at higher concen-
tive increase of approximately 17- and 21-fold in the rate of trations, laminin and fibronectin produced a 40-70% en-
migration compared to control. Providing less of a stimula- hancement of PDGF-induced migration compared to what
tory effect was fibronectin (5-fold increase), followed by would be anticipated by the addition of these two proteins.
PDGF-AB (3-fold increase) and finally laminin (2-fold in- Surprisingly, we did not find synergy between the growth
crease). This same hierarchy was demonstrated for both factors EGF or bFGF and any of the ECM proteins.60
chemokinesis and haptotaxis as well. Synergism between growth factors and ECM proteins
Thus, despite the previous focus on growth factors as has been previously observed in other cell types. As previ-
stimulants of SMC migration, our findings suggest that ously mentioned, Yabkowitz et al, who studied the effect of
matrix proteins, which are plentiful in the vessel wall after TSP on PDGF-dependent migration of calf pulmonary
arterial injury, are as a group more potent stimuli of SMC artery SMCs, noted a 60% increase in PDGF-induced mi-
migration.54 Although PDGF-BB is a stronger stimulant for gration with concentrations of TSP that alone had no effect
migration than PDGF-AB, neither of these growth factors on SMC migration.47 Vitronectin and type IV collagen have
could produce a migratory response that equaled that of also been noted to synergistically increase chemotaxis
collagen type I or type IV.54 We and others have also previ- induced by insulin-like growth factor I in a human breast
ously demonstrated that soluble growth factors such as EGF cancer cell line.61
and bFGF are less potent agonists of SMC migration than
PDGF-AB.56 These findings imply that matrix proteins may Integrins
be the predominant “in vivo” stimulus for SMC migration The above outlined effect of matrix proteins on SMC
following arterial injury. locomotion are mediated largely through activation of
In these studies, we consistently found that haptotaxis integrin receptors.62 It is through these receptors that ma-
(SMC migration toward a gradient of insoluble matrix pro- trix proteins are able to influence the intracellular machin-
tein) was the most profound stimulus of human SMC mi- ery that is necessary to initiate the migratory response. Iden-
gration.54 These findings were reproducible for all four ma- tification of the integrin receptors that are required for ma-
trix proteins; the SMC response to haptotaxis was on aver- trix protein-driven SMC migration is critical to the under-
age 33% greater than the response of SMC to chemotaxis standing of this complex process.
for the same agonist. Haptotaxis of neoplastic cells has been Integrins are a family of heterodimeric cell surface
previously studied in some detail and is thought to play a receptors composed of # and ! subunits.63 Approximately
significant role in the invasion and spread of metastatic tu- 17 different # subunits and 8 ! subunits and over 20 differ-
mor cells.57 It appears that haptotaxis and chemotaxis are in ent #! pairings have been identified. Three families of
fact very distinct processes rather than an extension of the integrins have been designated according to their ! subunits,
same process.58 In studies with thrombospondin, haptotaxis which include !1, !2, or !3. Beta 2 is localized largely in plate-
and chemotaxis were found to be mediated by completely lets; however, !1 and !3 have been detected in vascular
separate peptide domains of the thrombospondin molecule SMCs.15 A single ECM protein is able to bind multiple
and the signaling pathways activated by these two processes integrin receptors and a single integrin receptor is able to
were also very different.59 bind multiple ligands.64 In this way, the same ECM protein
After investigating the influence of individual growth can mediate different cellular responses via a variety of
factors and ECM proteins on SMC migration, we next stud- integrin receptors. Therefore, past associations of a single
ied the chemotactic response of SMC to combinations of integrin and receptor such as the designation of #5!1 as the
these proteins.60 At the time of arterial injury, SMCs are ex- “fibronectin” receptor and #v!3 as the “vitronectin” receptor
posed to a plethora of growth factors and ECM proteins. are too restrictive, since both #5!1 and #v!3 are able to be
Presumably these proteins can act in concert to influence affected by multiple ECM proteins. Thus, the control over
SMC migration. Possible interactions include: cellular function exerted via integrin receptors is complex
1. One factor may mask the other, with the combined and critically important to normal and abnormal cellular
response being equivalent to that of the more po- function. Although there has been extensive investigation
tent agonist; of integrins and their importance in vascular biology, here
2. The two factors may have an additive response; we will discuss the different integrins involved in matrix
3. The two factors may act in a synergistic manner in protein driven SMC migration.
which the sum of both factors is greater than the sum Choi et al explored the role of the #v!3 integrin in hu-
of their individual effects.60 man and rabbit PDGF-AB stimulated SMC migration.65
These investigators chose to study #v!3 because of its ability
Extracellular Matrix Proteins Are Potent Agonists of Human Smooth Muscle Cell Migration 357
to bind to several ECM proteins by its RGD-containing bind- haptotaxis to type I and IV collagen, fibronectin, and
ing site, making it a possible candidate as a mediator of cell laminin.68 This effect was consistently greater for type I col-
motility. Antibodies to #v!3, as well as an RGD peptide an- lagen and laminin versus type IV collagen and fibronectin.
tagonist of this same integrin, both inhibited PDGF-AB Moreover, a blocking antibody to the #2 integrin subunit
stimulated SMC migration in vitro, albeit incompletely, inhibited chemotaxis and haptotaxis to type I and IV col-
implicating involvement of other integrins in this process. lagen and laminin, though these effects were less than those
They did not assess the in vitro role of #v!3 in matrix pro- observed with the !1 antibody.68 In similar experiments,
tein driven SMC migration. In a rabbit carotid injury model, Skinner et al found that a blocking antibody to the #2!1
these same investigators found that a monoclonal antibody integrin also inhibited migration of cultured human SMCs
to #v!3 did not inhibit intimal hyperplasia, although a spe- on type I collagen.64
cific RGD peptide antagonist did. Further supporting the There is some evidence that the #1!1 integrin might
role of #v!3 in SMC migration, Clyman et al studied migra- also be involved in SMC migration. Clyman et al detected
69
tion in fetal lamb ductus arteriosus SMC and found that #1!1 in cultured rat vascular SMC. Moreover, Gotwals et
SMC migration on type I and IV collagen, fibronectin, and al, again using rat SMC, found that #1!1 is the primary fa-
laminin was mediated predominantly by the #v!3 integrin.66 cilitator of SMC migration on collagen.70 It is possible that
The importance of the #v!3 integrin in SMC migration, how- the findings of the latter two studies were related to the use
ever, remains controversial. Recent immunocytochemical of rat SMC, and the critical integrin necessary for SMC mi-
studies by Hosiga et al revealed that the #v!3 was localized gration may differ between human and rat.
only in the media and not the neointima of rat aortic tissue In our immunohistochemical studies, the #5 subunit
following injury.67 Skinner et al were not able to identify the was found homogeneously distributed throughout normal
64
#v!3 integrin in an arterial SMC line. In immunohis- human saphenous vein vascular SMCs and in hyperplastic
tochemical studies from this laboratory, cultured human plaques from saphenous vein grafts; however, a blocking
saphenous vein SMC stained only weakly positive for the antibody to the #5 integrin subunit had no effect on chemo-
68
#v!3 integrin and only after permeabilization. We were not taxis or haptotaxis of saphenous vein SMC following their
able to identify the #v!3 integrin in the intima or media of stimulation with any of the ECM proteins.68 Though the
hyperplastic lesions excised from patients following saphe- #5!1 has been reported by other investigators to be the pro-
nous vein bypass grafting (despite positive staining for this totypical integrin for fibronectin,21 our results suggest that
integrin in the microvessels of the adventitia of these same this integrin does not mediate fibronectin migration of hu-
lesions), nor was #v!3 present in atherosclerotic plaque re- man saphenous vein SMC.
moved at the time of carotid endarterectomy. Using SMC It is important to note that there is tremendous
derived from human saphenous vein and blocking antibod- variation in the distribution and function of integrins from
ies, we found that #v!3 was not required for ECM protein one tissue to another (animal versus human, primary versus
(collagen type I or IV, laminin, fibronectin, or vitronectin) cultured cells, artery versus vein). Therefore, it is necessary
stimulated SMC migration; however #v!3 did play a role in to critically analyze data from the appropriate cell type before
PDGF mediated chemotaxis.68 making conclusions about the importance of integrins in
Our studies and those of others suggest that the !1 remodeling after vascular injury. In studies of SMC derived
integrin subunit has a more dominant role in SMC migra- from human saphenous vein, we and Skinner et al have found
tion.68 Skinner et al studied several !1 integrin combinations the #2 and !1 subunits to be the most critical receptors
and demonstrated that #1!1, but not #2!1, was expressed in required for ECM protein stimulated chemotaxis and
vivo in normal intact human arterial tissue; however in cell haptotaxis.64,68 Thus, modulating the #2!1 integrin may be a
culture the expression of #1 was downregulated and #2!1 was potentially effetive method for controlling intimal hyper-
upregulated.64 These authors postulated that #1!1 may be plasia.
present in normal venous tissue, but that hyperplastic plaque
might then preferentially express #2!1, and that this conver- Cell Signaling
sion might contribute to the pathophysiology of intimal We and others have extensively evaluated the intrac-
hyperplasia. In immunohistochemical studies of normal ellular signaling pathways associated with PDGF-induced
human saphenous vein and specimens of intimal hyperpla- cell migration. A variety of signaling proteins and ions ap-
sia derived from excised vein grafts, we found the !1 subunit pear to be involved, including phosphatidylinositol-3-OH
to be homogeneously distributed throughout both normal kinase (PI3K),71 phospholipase C (PLC),72 MAPK,73 Src,74
and abnormal tissues.68 Similarly, the #1 integrin was found calcium, 75 calmodulin-dependent protein kinase II
in normal human saphenous vein as well as hyperplastic tis- (CamKII),76 and the small G-protein rho.77 The intracellu-
sue from saphenous vein grafts. Interestingly, the #2 integrin lar signaling events that mediate matrix protein-induced
was only identified in specimens of intimal hyperplasia, cor- SMC migration have been less well studied. Our early inves-
roborating the hypothesis proposed by Skinner et al that the tigations suggest that at least some of these pathways may
#2!1 integrin is expressed only following arterial injury. be distinct from those used by growth factor receptors.
We conducted further studies which verified the physi- Matrix protein-driven signaling events are initiated
ologic importance of the #2!1 integrin in human SMC mi- through activation of integrin receptors.62 As previously
gration. We found that a blocking antibody to the !1 integrin mentioned, matrix proteins attach to the extracellular do-
subunit produced an inhibitory effect on chemotaxis and main of integrin receptors, and the short cytoplasmic
358 Tissue Engineering of Prosthetic Vascular Grafts
domains of both the # and ! integrin subunits allow trans- We have also studied the role of large G-proteins (Gs)
fer of messages to the intracellular environment.15 These in the intracellular signaling pathways involved in matrix
cytoplasmic domains have direct access to the signaling ma- protein-driven SMC migration.54 Cholera toxin, which leads
chinery in the cell, and studies have demonstrated that mu- to constitutive activation of Gs,21 profoundly inhibited SMC
tations or truncations of the distal end of both the # and the migration in response to collagen types I and IV, fibronectin,
! cytoplasmic tail can interfere with integrin signaling and and laminin and even eliminated baseline migration in
cytoskeletal organization.78 Further evidence of the impor- unstimulated cells.54 Activation of Gs increases levels of
tance of the ! subunit in intracellular signaling can be de- cAMP through stimulation of adenylate cyclase, and ago-
rived from studies where overexpression of a single ! cyto- nists of cAMP such as forskolin and 8-bromo-cAMP have
plasmic tail leads to inhibition of tyrosine phosphorylation been found to inhibit SMC migration.86 Thus, the inhibi-
of intracellular proteins such as FAK, 79 whereas tory effect of Gs activation with cholera toxin may be due to
overexpression of multiple ! subunits configured in clusters elevated levels of cAMP.87 Surprisingly, pertussis toxin, which
results in an increase in tyrosine phosphorylation.80 Through inhibits the Gi subtypes, including Gi1, Gi2, and Gi3, and
these and similar studies, the concept has evolved that thereby releases the inhibitory effect that Gi has on adeny-
integrin clustering is a prerequisite for the initiation of in- late cyclase,21 had no effect on chemotaxis or haptotaxis to
tracellular signalling.81 In in vitro studies, the # and ! cyto- fibronectin, type I or type IV collagen.54 However, pertussis
plasmic domains have been found to couple with a variety toxin did partially inhibit haptotaxis and completely elimi-
of cytoplasmic proteins such as p125FAK, #-actinin, talin, nated chemotaxis to laminin.54 Similarly, Aznavoorian et al
vinculin and paxillin.17 It is activation of these proteins that found that pertussis toxin decreased chemotactic migration
allows for the induction of intracellular signaling pathways of A2058 human melanoma cells when stimulated with
that lead to migration. laminin; however, with these same cells there was no inhibi-
Focal adhesion kinase (FAK) is a nonreceptor tyrosine tion of fibronectin-induced chemotaxis.88 Although Gi pro-
kinase that has been found to be activated by both ECM teins can influence adenylate cyclase, the multiple Gi sub-
and soluble signaling factors.81 Ligand attachment leads to types can also interact with enzymes such as phospholipase
direct phosphorylation of FAK by the ! subunit of the A2, C, and D, and ion transporters such as potassium and
integrin receptor.16 FAK can then interact with Src, another calcium channels.89 Thus, the varying role that pertussis
nonreceptor tyrosine kinase, which is localized to focal ad- toxin has on SMC migration may be due to either inhibi-
hesions by its SH2 domain.82 Src in turn can activate the tion of the inhibitory influence of Gi on cAMP or perhaps
Ras/MAPK signaling pathways,83 both which have been pre- the lack of interaction of Gi with one or more of these other
viously implicated in SMC migration.73,84 MAPK may pro- signaling substrates.
mote migration either through the phosphorylation and ECM proteins are known to play a role in the turn-
activation of transcription factors which regulate integrin over of phospholipids. In fibroblasts, fibronectin increased
gene expression, or by phosphorylating and activating cyto- the production of phosphatidylinositol 4,5 bisphosphate
plasmic phospholipase A2 (cPLA2), which hydrolyzes (PIP2) and dissociation of these same cells from fibronectin
glycerophospholipids to produce arachidonic acid.16 It has resulted in a rapid decrease in cellular PIP2.90 PIP2, in turn,
been previously demonstrated that inhibition of cPLA2 pre- has been implicated in cellular migration91 and this effect
vents cell migration, an effect that can be reversed by the may be mediated by several pathways. It has been specu-
readdition of arachidonic acid.16 lated that the upregulation of the production of PIP2 may
We and others have shown a relationship between lev- facilitate the polymerization of actin because of a regula-
els of intracellular calcium and PDGF-induced SMC migra- tory effect of PIP 2 on actin binding proteins such as
tion.75,76 We postulated that a rise in intracellular calcium profilin.16 PIP2 is also an important substrate for the en-
might also be required for SMC migration in response to zyme PLC.63 PLC hydrolyzes PIP2 into the active second
matrix proteins.60 Despite the potent stimulatory effect of messengers diacylglycerol (DAG) and inositol trisphosphate
collagen types I and IV, fibronectin, and laminin on SMC (IP3), which then respectively activate protein kinase C
migration, none of the matrix proteins produced a sustained (PKC) and the release of intracellular calcium. Both PKC
or prolonged rise in intracellular calcium.60 This was true and calcium have been implicated in the signaling pathways
even if calcium measurements were carried out for more for cell migration.75,92
than 30 minutes after stimulation. This finding was surpris- We have previously demonstrated that growth factors
ing, since activation of the #v!1 integrin by fibronectin has and matrix proteins act synergistically to promote SMC
been shown to produce a persistent rise in intracellular cal- migration.60 The mechanism through which these two ago-
cium in endothelial cells.85 These findings imply that cal- nists might interact is not clear; however, the point of inter-
cium plays little if any role in matrix protein stimulated SMC section may be either at the level of the receptor or at a mu-
locomotion. We did find that fibronectin, laminin, and type tual downstream signaling pathway. There is some evidence
I and IV collagen augmented the early peak in intracellular that ECM activation of integrins can up or downregulate
calcium produced by PDGF.60 Although it is possible that growth factor receptors. The term “crosstalk” has been used
this augmentation provides at least a partial explanation for to describe such interactions between growth factor recep-
the synergistic enhancement by matrix proteins of PDGF- tors and integrins.93 We have recently reported that type I
induced SMC migration, the physiologic relevance of this collagen enhances clustering and activation of the PDGF-!
finding is still unclear. receptor.94 Tyrosine phosphorylation by growth factor re-
Extracellular Matrix Proteins Are Potent Agonists of Human Smooth Muscle Cell Migration 359
ceptors of the cytoplasmic domains of integrins, or of pro- 7. Chen P, Gupta K, Wells A. Cell movement elicted by epi-
teins associated with the focal adhesion complex is an addi- dermal growth factor receptor requires kinase and
tional mechanism by which these two ligands/receptors autophosphorylation but is separable from mitogenesis. J
might interact.60 Other potential sites of interaction are at Cell Biol 1994; 124:547-555.
8. Madri JA, Bell L, Marx M et al. Effects of soluble factors
the level of the proteins, MAPK, PI3K and/or PIP2. It is im-
and extracellular matrix components on vascular behav-
portant that we investigate the synergistic interactions be- ior in vitro and in vivo: Models of de-endothelialization
tween matrix proteins and growth factors, since these points and repair. J Cell Biochem 1991; 45(2):123-30.
of interaction may represent exceedingly effective targets for 9. Stossel TP. From signal to pseudopod. J Biol Chem 1989;
inhibitors of intimal hyperplasia. 264:18261-18264.
10. Vandekerckhove J. Actin-binding proteins. Curr Opin in
Conclusion Cell Biol 1990; 2:41-50.
In this chapter, we have reviewed the effect of the nu- 11. Burridge K, Fath K, Kelly T et al. Focal adhesions: Trans-
merous matrix components of the vessel wall on SMC mi- membrane junctions between the extracellular matrix and
gration. It is clear that many of these proteins produce a the cytoskeleton. Ann Rev Cell Biol 1988; 4:487-525.
strong stimulus for migration. Moreover, the effect of these 12. Luna EJ, Hitt AL. Cytoskeleton-plasma membrane inter-
actions. Science 1992; 258:955-964.
matrix proteins on SMC migration may be more profound
13. Huttenlocher A, Ginsberg MH, Horwitz AF. Modulation
than that of many of the growth factors that have been tra- of cell migration by integrin-mediated cytoskeletal link-
ditionally studied. We have also shown that these matrix ages and ligand-binding affinity. J Cell Biol 1996;
proteins can interact with growth factors to provide a syn- 134(6);1551-62.
ergistic stimulus for SMC locomotion. The chemotaxic ef- 14. Stossel TP. On the crawling of animal cells. Science 1993;
fect of matrix proteins is mediated through integrin recep- 260(5111):1086-94.
tors of which, at least for human venous SMC, #2!1 appears 15. Hynes RO. Intergrins: Versatility, modulation and signal-
to be the most critical. The signaling pathways that mediate ling in cell adhesion. Cell 1992; 68:11-25.
matrix-induced migration have not been fully elucidated but 16. Clark EA, Brugge JS. Integrins and signal transduction
appear to be distinct from those that mediate migration of pathways: The road taken. Science 1995; 268:233-238.
SMC in response to growth factors. 17. Shattil SJ, Ginsberg MH. Integrin signaling in vascular
biology. J Clin Invest 1997; 100(1):1-5.
Vascular disease remains a major cause of morbidity
18. Bendeck MP, Irvin C, Reidy MA. Inhibition of matrix
and mortality in the United States. Treatment is often pal- metalloproteinase activity inhibits smooth muscle cell
liative because of the frequent occurrence of recurrent dis- migration but not neointimal thickening after arterial in-
ease at sites of vascular reconstruction. Engineering of an jury. Circ Res 1996; 78::38-43.
“ideal” vascular graft that is not affected by intimal hyper- 19. Kenagy RD, Vergel S, Mattsson E et al. The role of plas-
plasia would prolong life and reduce morbidity. Matrix pro- minogen, plasminogen activators, and matrix
teins must be an essential component of this engineered con- metalloproteinases in primate arterial smooth muscle cell
duit, and the proteins that are included should be chosen migration. Arterioscler Throm Vasc Biol 1996;
based not only on their structural characteristics but also 16(11):1373-82.
on their effect on SMC physiology and pathophysiology. 20. Schwartz SM. Smooth muscle migration in atherosclero-
sis and restenosis. J Clin Invest 1997; 99(12):2814-2817.
Moreover, with the realization that ECM proteins and
21. Zetter BR, Brightman SE. Cell motility and the extracel-
integrins play an important role in SMC migration, phar- lular matrix. Curr Opin Cell Biol 1990, 2:850-856.
macological agents such as integrin inhibitors may be de- 22. Taraboletti G, Roberts DD, Liotta LA. Thrombospondin-
veloped and integrated into vascular grafts to prevent the induced tumor cell migration: Haptotaxis and chemot-
formation of intimal hyperplasia. The development of an axis are mediated by different molecular domains. J Cell
ideal prosthesis is an onerous task but before this goal can Biol 1987; 105:2409-15.
be realized, there must be a thorough understanding of the 23. Ruoslahti E, Engvall E. Integrins and vascular extracellu-
pathophysiology of restenosis in vascular reconstructions. lar matrix assembly. J Clin Invest 1997; 99(6):1149-52.
24. Carey DJ. Control of growth and differentiation of vas-
References cular cells by extracellular matrix proteins. Annu Rev
1. Chervu A, Moore WS. An overview of intimal hyperpla- Physiol 1991; 53:161-77.
sia. Surg Gynecol Obstet 1990; 171:433-40. 25. Murata K, Motayama T, Kotake C. Collagen types in vari-
2. Donaldson MC, Mannick JA, Whittemore AD. Causes of ous layers of the human aorta and their changes with the
primary graft failure after in situ saphenous vein bypass atherosclerotic process. Athero 1986; 60:251-62.
grafting. J Vasc Surg 1992; 15:113-20. 26. Ross R, Klebanoff SJ. The smooth muscle cell: In vivo
3. Casscells W. Migration of smooth muscle and endothe- synthesis of connective tissue proteins. J Cell Biol 1971;
lial cells. Critical events in restenosis. Circ 1992; 86:723-9. 50:159-171.
4. Clowes AW, Schwartz SM. Significance of quiescent 27. Wight TN, Ross R. Proteoglycans in primate arteries: Syn-
smooth muscle cell migration in the injured rat carotid thesis and secretion of glycosaminoglycans by arterial
artery. Circ Res 1985; 56:139-45. smooth muscle cells in culture. J Cell Biol 1975;
5. Ross R. The pathogenesis of atherosclerosis: A perspec- 67:675-686.
tive for the 1990s. Nature 1993; 362:801-9. 28. Kraiss LW, Kirkman TR, Kohler TR et al. Shear stress
6. Mii S, Ware JA, Kent KC. Transformtin growth factor-! regulates smooth muscle proliferation and neointimal
inhibits human vascular smooth muscle cell growth and thickening in porous polytetrafluoroethylene grafts.
migration. Surgery 1993; 114:464-70. Arterio and throm 1991; 11:1844-1852.
360 Tissue Engineering of Prosthetic Vascular Grafts
29. Geary RL, Kohler TR, Vergel S et al. Time course of flow- pulmonary artery smooth muscle cells. Jour Cell Physio
induced smooth muscle cell proliferation and intimal 1993; 157:24-32.
thickening in endothelialized baboon vascular grafts. Circ 48. Riessen R, Wight TN, Pastore C et. al. Distribution of
Res 1993; 74:14-23. hyaluron during extracellular matrix remodeling in hu-
30. Barnes M. Collagens in atherosclerosis. Collagen Rel Res man restenotic arteries and balloon-injured rat carotid
1985; 5:65-97. arterties. Circ 1996; 93:1141-1147.
31. Snow AD, Bolender RP, Wight TN, Clowes AW. Heparin 49. Savani RC, Wang C, Yang B et al. Migration of bovine
modulates the composition of the extracellular matrix aortic smooth muscle cells after wounding injury: The role
domain surrounding arterial smooth muscle cells. Am J of hyaluronan and RHAMM. J Clin Invest 1995;
Pathol 1990; 137:313-330. 95:1158-1168.
32. Bjorkerud S. Cultivated human arterial smooth muscle 50. Niculescu F, Rus HG, Porutiu D et al. Immunoelectron-
displays heterogeneous pattern of growth and phenotypic microscopic localization of S-protein/vitronectin in hu-
variation. Lab Invest 1985; 53(3):303-310. man atherosclerotic wall. Athero 1989; 78(2-3)197-203.
33. Van Zanten GH, de Graaf S, Slootweg PJ et al. Increased 51. Preissner KT. Structure and biological role of vitronectin.
platelet deposition on atherosclerotic coronary arteries. J Annu Rev Cell Biol 1991; 7:275-310.
Clin Invest 1994; 93(2):615-32. 52. Naito M et al. Vitronectin-induced haptotaxis of vascular
34. Wolf YG, Rasmussen LM, Ruoslahti E. Antibodies against smooth muscle cells in vitro. Exp Cell Research 1991;
transforming growth factor !-1 suppresses intimal hyper- 194:154-156.
plasia in a rat model. J Clin Invest 1994; 93:1172-1178. 53. Brown SL, Lundgren CH, Nordt T et al. Stimulation of
35. O’Brien ER, Garvin MR, Stewart DK et al. Osteopontin migration of human aortic smooth muscle cells by
is synthesized by macrophage, smooth muscle, and en- vitronectin: Implications for atherosclerosis. Cardiovas
dothelial cells in primary and restenotic human coronary Research 1994; 24:1815-1820.
atherosclerotic plaques. Arterioscler Throm 1994; 54. Nelson PR, Yamamura S, Kent CK. Extracellular matrix
14(10):1648-56. proteins are potent agonist of human smooth muscle cell
36. Van Aken BE, Seiffert D, Thinnes T et al. Localization of migration. J Vasc Surg 1996; 24:25-33.
vitronectin in the normal and atherosclerotic human ves- 55. DiMilla PA, Stone JA, Quinn JA et al. Maximal migra-
sel wall. Histochem Cell Biol 1997; 107(4):313-20. tion of human smooth muscle cells on fibronectin and
37. Patel MK, Clunn GF, Lymn JS et al. Thrombospondin-1 collagen iv occurs at an intermediate attachment strength.
induces DNA synthesis and migration in human vascular J Cell Biol 1993; 122:729-37.
smooth muscle cells. Biochem Soc Trans 1996; 24(3):446 56. Mii S, Ware JA, Kent KC. Transformtin growth factor-!
38. Hedin U, Holm J, Hansson GK. Induction of tenascin in inhibits human vascular smooth muscle cell growth and
rat arterial injury. Relationship to altered smooth muscle migration. Surgery 1993; 114:464-470.
cell phenotype. Am J Path 1991; 139(3):649-56. 57. Tsuruoka T, Azetaka M, Iizuka Y et al. Inhibition of tu-
39. Liaw L, Almeida M, Hart CE et al. Osteopontin promotes mor cell haptotaxis by sodium D-glucaro-delta-lactam. Jpn
vascular cell adhesion and spreading and is chemotactic J Cancer Res 1995; 86(11):1080-5.
for smooth muscle cells in vitro. Circ Res 1994; 58. Keller UH, Wissler JH, Ploem J. Chemotaxis is not a spe-
74:214-224. cial case of haptotaxis. Experienta 1979; 35:1669-71.
40. Fitzpatrick LA, Severson A, Edwards WD et al. Diffuse 59. Taraboletti G, Roberts DD, Liotta LA. Thrombospondin-
calcification in human coronary arteries: Association of induced tumor cell migration: Haptotaxis and chemot-
osteopontin with atherosclerosis. J Clin Invest 1994; axis are mediated by different molecular domains. J Cell
94:1597-1604. Biol 1987; 105:2409-15.
41. Yue T, McKenna PJ, Ohlstein EH et al. Osteopontin- 60. Nelson PR, Yamamura S, Kent CK. Platelet-derived
stimulated vascular smooth muscle cell migration is me- growth factor and extracellular matrix proteins provide a
diated by !3 integrin. Exp Cell Res 1994; 214:459-464. synergistic stimulus for human vascular smooth muscle
42. Liaw L, Lombardi DM, Almeida SM et al. Neutralizing cell migration. J Vasc Surg 1997; 26:104-12.
antibodies directed against osteopontin inhibit rat carotid 61. Doerr ME, Jones JI. The roles of integrins and extracellu-
neointimal thickening following endothelial denudation. lar matrix proteins in the insulin-like growth factor I-
Arterioscler Thromb Vasc Biol 1997; 17:188-193. stimulated chemotaxis of human breast cancer cells. J Biol
43. Chen C, Ku DN, Kikeri D et al. Tenascin: A potential Chem 1996; 271:2443-47
role in human arteriovenous PTFE graft failure. J Surg 62. Clark EA, Brugge JS. Integrins and signal transduction
Res 1996; 60(2):409-16. pathways: The road taken. Science 1995; 268:233-238.
44. Murphy-Ullrich JE, Pallero MA, Boerth N et al. Cyclic 63. Schwartz MA. Transmembrane signaling by integrins.
GMP-dependent protein kinase is required for Trends Cell Biol 1992; 2:304-8.
thrombospondin and tenascin mediated focal adhesion 64. Skinner MP, Raines EW, and Ross R. Dynamic expres-
disassembly. J Cell Sci 1996; 109(10):2499-508. sion of #1!1 and #2!1 integrin receptors by human vascu-
45. Majesky MW. Neointima formation after acute vascular lar smooth muscle cells. Am J Pathol 1994; 145:1070-1081.
injury. Role of counteradhesive extracellular matrix pro- 65. Choi ET, Engel L, Callow A et al. Inhibition of neointimal
teins. Tex Heart Inst J 1994; 21(1):78-85. hyperplasia by blocking #v!3 integrin with a small pep-
46. Hahn AW, Jonas U, John M et al. Functional aspects of tide antagonist GpenGRDSPCA. J Vasc Surg 1994;
vascular tenascin-c expression. J Vasc Res 1995; 19:125-34.
32(3):162-74. 66. Clyman RI, Mauray F, Kramer RH. !1 and !3 integrins
47. Yabkowitz R, Mansfield PJ, Ryan US et al. have different roles in the adhesion and migration of vas-
Thrombospondin mediates migration and potentiates cular smooth muscle cells on extracellular matrix. Exp Cell
platelet-derived growth factor-dependent migration of calf Res 1992; 200:272-284.
Extracellular Matrix Proteins Are Potent Agonists of Human Smooth Muscle Cell Migration 361
67. Hosiga M, Alpers CE, Smith LL. #v!3 integrin expression 81. Illic D, Damsky CH, Yamamoto T. Focal adhesion kinase:
in normal and atherosclerotic artery. Circ Res 1995; At the crossroads of signal transduction. J Cell Sci 1997;
77:1129-1135. 110:401-407.
68. Itoh H, Nelson PR, Mureebe L et al. The role of integrins 82. Calalb MB, Polte TR, Hanks SK. Tyrosine phosphoryla-
in saphenous vein vascular smooth muscle cell migration. tion of focal adhesion kinase at sites in the catalytic do-
J Vasc Surg 1997; 25:1061-9. main regulates kinase activity: A role for Src family ki-
69. Clyman RI, Turner DC, Kramer RH. An #1!1-like integrin nases. Mol Cell Biol 1995; 15:954-63.
receptor on rat aortic smooth muscle cells mediates ad- 83. Schlaepfer DD, Hanks SK, Hunter T et al. Integrin-medi-
hesion to laminin and collagen types I and IV. Arterio ated signal transduction linked to Ras pathway by GRB2
1990; 10:402-409. binding to focal adhesion kinase. Nature 1995;
70. Gotwals PJ, Chi Rosso G, Lindner V et al. The #1! 1 372:786-791.
integrin is expressed during neointima formation in rat 84. Irani K, Herzlinger S, Finkel T. Ras proteins regulate
arteries and mediates collagen matrix reorganization. J multiple mitogenic pathways in A10 vascular smooth
Clin Invest 1996; 97:2469-2477. muscle cells. Biochem Biophys Res Commun 1994;
71. Yamamura S, Derman MP, Cantly LG et al. The role of 202(3):1252-8.
phosphatidylinositol 3-kinase in proliferation and migra- 85. Schwartz MA, Denninghoff K. Alpha v integrins mediate
tion of human vascular smooth muscle cells. Book of the rise in intracellular calcium in endothelial cells on
Abstracts, Annual Meeting of the Society for Vascular fibronectin even though they play a minor role in adhe-
Surgery, 1995, p.53 sion. J Biol Chem 1994; 269(15):11133-7.
72. Yamamura S, Nelson PR, Mureebe L, Kent KC. Prolifera- 86. Horio T, Kohno M, Kano H et al. Arenomedulin as a
tion and migration of vascular smooth muscle cells are novel antimigration factor of vascular smooth muscle cells.
mediated by distince signaling pathways involving phos- Circ Res 1995; 77:660-4.
pholipase C. Surgical Forum Volume 1996; 47:362-64. 87. Gilman AG. G-proteins: Transducers of receptor-gener-
73. Nelson PR, Mureebe L, Itoh H, Kent KC. Smooth muscle ated signals. Annu Rev Biochem 1987; 56:615-49.
cell migration and proliferation are mediated by distinct 88. Aznavoorian S, Stracke ML, Krutzch H et al. Signal trans-
phases of activation of the intracellular messenger mito- duction for chemotaxis and haptotaxis by matrix mol-
gen-activated protein kinase. Submitted J Vasc Surg. 1997. ecules in tumor cells. J Cell Biol 1990; 110:1427-1438.
74. Mureebe L, Nelson PR, Yamamura S, Kent KC. Activa- 89. Lester BR, McCarthy JB, Sun Z. G-protein involvement
tion of pp60c-src is necessary for human smooth muscle in matrix-mediated motility and invation of high and low
cell migration. In Press, Surgery. 1997. experimental metastatic B16 melanoma clones. Can Res
75. Yamamura S, Nelson PR, Kent KC. The role of intracel- 1989; 49:5940-5948.
lular calcium in migration of human vascular smooth 90. McNamee HP, Ingber DE, Schwartz MA. Adhesion to
muscle cells. Surgical Forum Volume 1995; 46:386-389. fibronectin stimulates inositol lipid synthesis and enhances
76. Pauly RR, Bilato C, Sollott SJ. Role of calcium/calmodulin- PDGF-induced inositol lipid breakdown. J Cell Biol 1993;
dependent protein kinase II in the regulation of vascular 121:673-8.
smooth muscle cell migration. Circ 1995; 91(4):1107-1115. 91. McNamee HP, Liley HG, Ingber DE. Integrin-dependent
77. Jiang B, Ware JA, Mallette SA, Kent KC. Migration and control of inositol lipid synthesis in vascular endothelial
proliferation of vascular smooth muscle cells are medi- cells and smooth muscle cells. Exp Cell Res 1996;
ated by distinct signal transduction pathways. Circ 1994; 224(1):116-22.
90:1190. 92. Vuori K and Ruoslahti E. Activation of protein kinase C
78. Ylanne J, Huuskonen J, O’Toole TE et al. Mutation of precedes alpha 5 beta 1 integrin-mediated cell spreading
the cytoplasmic domain of the integrin !3 subunit: Dif- on fibronectin. J Biol Chem 1993; 268(29):21459-2.
ferential effects on cell spreading and migration, and 93. Seki J, Koyama N, Kovach NL et al. Regulation of !1
matrix assembly. J Cell Biol 1994; 270:9550-9557. integrin function in cultured human vascular smooth
79. Chen YP, O’Toole TE, Shipley T et al. “Inside-out” sig- muscle cells. Circ Res 1996; 78:596-605.
nal transduction inhibited by isolated integrin cytoplas- 94. Itoh H, Mureebe L, Kubaska S, Kent KC. Type I collagen
mic domians. J Biol Chem 1994; 269:18307-18310. enhances phosphorylation of the PDGF receptor and acts
80. Lukashev ME, Sheppard D, Pytela R. Disruption of synergistically to augment PDGF-induced vascular smooth
integrin function and induction of tyrosine phosphoryla- muscle cell proliferation. Surgical Forum Volume XLVIII
tion by the autonomously expressed !1 integrin cytoplas- 1997:406-408.
mic domain. J Biol Chem 1994; 269:18311-18314.
Facilitation of Healing
Matrix Modulation
CHAPTER 33
Extracellular Matrix Effect on Endothelial Control
of Smooth Muscle Cell Migration and Matrix Synthesis
Richard J. Powell
Introduction
I ntimal hyperplasia remains the most common cause of early failure following angioplasty
and bypass surgery.1,2 Intimal hyperplasia is a particularly prevalent problem in small di-
ameter synthetic grafts used for extremity bypasses performed to tibial and peroneal tar-
gets.3 The development of intimal hyperplasia following smooth muscle cell (SMC) injury
is characterized by SMC dedifferentiation from a contractile to synthetic phenotype.4,5 Fol-
lowing phenotypic modulation, SMCs migrate into the subintimal space, proliferate and
secrete extracellular matrix. For the first several weeks following injury the intimal hyper-
plastic lesion is predominantly cellular. However, as the lesion continues to enlarge, extra-
cellular matrix gradually becomes the predominant component.1,4-7 Several months follow-
ing injury, extracellular matrix comprises 80% of the mature intimal hyperplastic lesion.8
In several recent studies, endothelial cell (EC) seeding of injured arterial wall seg-
ments and synthetic grafts has been shown to limit the development of intimal hyperplasia.
Bush and co-workers have shown that EC seeding of endarterectomized canine arteries de-
creased the intimal hyperplastic response.8,9 Conte has shown that EC seeding of injured
hypercholesterolemic rabbit femoral arteries also limits the intimal hyperplastic response.10
In addition, several in vivo human studies have shown improved patency of endothelial
sodded synthetic grafts when compared to unsodded grafts.11-14 The improved patency has
been attributed to decreased thrombogenicity and increased resistance to intimal hyperpla-
sia in the sodded grafts. Zilla and co-workers have recently shown that EC lined synthetic
grafts have a 73%, 5 year primary patency rate in the below knee position compared to 31%
for unlined grafts.12 They now have follow up data out to 6.5 years with a patency rate of
71% in the EC lined grafts and 0% in the controls.11 However, not all studies of EC sodded
grafts have shown improved patency rates. A report by Herring and co-workers failed to
show any improvement in EC sodded graft patency when compared to control grafts.3 One
explanation for the lack of effect of EC sodding was thought to be the low EC seeding den-
sity used to line the grafts, which resulted in an incomplete coverage of the graft lumen.
Extracellular matrix profoundly affects both endothelial and smooth muscle cell func-
tion. Following arterial wall injury, not only is extracellular matrix quantity increased but
alterations in matrix composition occurs as well. Matrix composition can affect SMC func-
tions such as migration and proliferation, and may also affect matrix synthesis.15 For in-
stance, following vascular wall injury there is an increase in type I collagen present within
the restenotic lesion. Type I collagen has been shown to stimulate smooth muscle cell
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
364 Tissue Engineering of Prosthetic Vascular Grafts
dedifferentiation and increase SMC proliferation.16 In ad- are established by plating ECs on the outer side of the mem-
dition, type I collagen has been shown to upregulate the ex- brane and grown to confluence over 3-5 days. Once the ECs
pression of the growth factor TGF-!1.17 TGF-!1 is a potent are confluent (2-3 days, as determined by phase contrast
stimulant of extracellular matrix synthesis and inhibits ma- microscopy), SMCs are plated on the opposite side of the
trix degradation.18 TGF-!1 has been shown to potentiate the PET membrane (see Fig. 33.1). After 24 h both cell types are
development of intimal hyperplasia in animal models fol- placed in DMEM/2.5% CS for the duration of each
lowing arterial injury.19 In similar studies, neutralizing an- experiment.
tibodies to TGF-!1 have been shown to inhibit the develop- This coculture model allows for the diffusion of soluble
ment of intimal hyperplasia following arterial injury. Tran- factors across the membrane, yet the two cell types remain
scripts to TGF-!1 are upregulated in intimal hyperplastic separated for examination. Previous work has shown that
human coronary lesions retrieved by coronary ECs maintain SMCs in a more differentiated contractile
atherectomy.20 TGF-!1 appears to be an important media- phenotype when compared to SMCs cultured alone, which
tor of the increased extracellular matrix deposition which exhibit a synthetic phenotype.21,22 There is limited physical
occurs during vascular wall remodeling. contact across the membrane during the first 4-6 days
The effects of various extracellular matrix proteins on following coculture; after which, however, SMC cytoplas-
endothelial cell control of smooth muscle cell function is mic projections begin to cross the membrane and contact
unknown. This information is important in the study of tis- the ECs.23
sue engineering of prosthetic grafts. To which particular
matrix protein endothelial cells and smooth muscle cells are Effect of Matrix on Endothelial Cell Control
exposed will determine what effect ECs have on such SMC of Smooth Muscle Cell Migration
functions as phenotype expression, proliferation, migration Migration of SMCs into the anastomotic site follow-
and matrix synthesis. This could obviously affect the patency ing bypass grafting is an early and important event in the
of EC sodded prosthetic grafts. The purpose of the work development of intimal hyperplasia.24 SMC migration oc-
performed in our laboratory is to define the effect of matrix curs following phenotypic modulation to a synthetic phe-
composition on the ability of ECs to regulate smooth muscle notype and prior to SMC proliferation and extracellular ma-
cell functions. In particular, we are interested in the matrix trix synthesis. The process of migration is distinct from cel-
effect on EC control of SMC migration and extracellular lular proliferation. Soluble factors such as bFGF, PDGF and
matrix synthesis. To study this we have developed a coculture extracellular matrix are involved in the control of SMC mi-
model in which ECs are cultured opposite SMCs on a semi- gration and appear to play a role in the intimal hyperplastic
permeable membrane. response.24,25
The role of ECs in modulating the SMC response to
Model injury is unclear. Previous work evaluating the effect of ECs
Bovine aortic ECs and SMCs are harvested using the on SMC proliferation have yielded controversial results and
collagenase method for ECs and the explant method for have not directly addressed the effect of ECs on SMC mi-
SMCs. Cells are grown for 3-5 passages from primary cul- gration. In vivo studies by Fingerle and co-workers have
tures in Dulbecco’s Modified Eagles Media/10% calf serum demonstrated that the loss of ECs from the arterial intima
(DMEM/10% CS). ECs are identified by their cobblestone results in the development of an intimal hyperplastic lesion
morphology and positive uptake of di-I-acetylated ldl. SMCs and that this lesion formation can be limited by rapid re-
are identified using an anti-#-actin antibody (Sigma A-2547, endothelialization.26 Although this is suggestive of the im-
Sigma Chemical, St. Louis, MO). For each component of portant role of ECs in limiting this process, these authors
these studies, SMC cultures are established by plating cells have not identified an EC released substance which might
on a 13 ∝m thick polyethylene terapthalate (PET) membrane account for this phenomenon. Conte and co-workers failed
with 0.45 ∝m pores configured at a density of 1.6 million to show any attenuation of the intimal hyperplastic response
pores/cm2 (Cyclopore membrane, Falcon cell culture insert, following early luminal EC seeding of balloon catheter in-
Becton Dickinson, Lincoln Park, NJ). EC/SMC cocultures jured arterial wall segments in rabbits with normal choles-
terol levels, but did show a reduction in intimal hyperplasia Previous investigators have shown that SMC migration
in a hypercholesterolemic model.10 In addition, Reidy and can be modulated by the extracellular matrix substrate
Silver failed to show any increase in SMC replication fol- composition. Our laboratory has shown that shown that
lowing gentle denudation of ECs from the intimal surface fibronectin decreases the migration of SMCs cultured alone
as long as there was no injury to the underlying SMCs.27 when compared to cells cultured on plastic (see Fig. 33.2).15
Several reports describing the increased resistance of EC The presence of ECs stimulated SMC migration on both
sodded grafts to the development of intimal hyperplasia have plastic and fibronectin coated membranes. This finding was
been previously mentioned. More recently, synthetic graft different when SMCs were cultured on type I collagen which
seeding studies performed by Lewis and co-workers have stimulated migration in SMCs cultured alone compared to
shown that EC sodded grafts produce EC-derived vasoac- SMCs cultured on plastic or fibronectin. In addition, the
tive factors which could limit SMC proliferation and mi- presence of type I collagen significantly inhibited the ability
gration.28 Thus it is unclear how important the role of the of ECs to stimulate SMC migration such as that observed
EC is in controlling SMC, migration and proliferation fol- on plastic and fibronectin (see Fig. 33.2). Thus, matrix
lowing vascular wall injury in-vivo. composition affected the EC ability to modulate
In vitro cell culture studies by Castellot and co-work- SMC migration.
ers have shown that ECs can inhibit SMC proliferation as a Cell matrix interactions which allow the cell to attach
result of secreted heaprin sulfates from the abluminal en- and detach from the extracellular matrix are also important
dothelial cell surface.29 These results conflict with studies in the migration process.31 It has been shown that there is
performed by Fillinger and co-workers as well as from our an intermediate cell attachment strength at which cell mi-
own laboratory which have shown that ECs stimulate SMC gration rate is optimized. Altering either the expression or
proliferation when these two cell types were cocultured on organization of cell surface integrin and nonintegrin attach-
opposite sides of a semipermeable membrane.22,30 Since pro- ment molecules will subsequently affect cell migration, as
liferation and migration are unassociated processes, these will altering extracellular matrix composition. In support
above studies cannot be used to address what effect ECs have of this hypothesis are the recent reports which demonstrate
on SMC migration. Using a steel fence assay our laboratory that following dedifferentiation SMCs alter their expression
has previously demonstrated that ECs stimulate SMC mi- of cell surface integrins, upregulating #2!1 integrin and de-
gration in vitro and that this process can be mediated by a creasing #1!1 integrin expression.32
secreted soluble factor.15 Possible factors involved include Basson and co-workers have shown that the growth
PDGF and endothelin-1, growth factors which have been factor TGF-!1 alters integrin expression, whereas matrix
shown to be secreted by cultured ECs. In addition, we have composition can change the cell surface integrin organiza-
previously shown that ECs may regulate TGF-!1 activation tion.33 Our laboratory has shown that adhesion molecule
in coculture.22 Changes in TGF-!1 levels could as a result expression and/or organization as determined by cell adhe-
alter SMC migration. Finally, ECs may secrete extracellular sion is altered by the presence of ECs.15 This is true regard-
matrix proteins which could deposit on the opposite side of less of whether SMCs are cultured on fibronectin, type I
the membrane and subsequently affect SMC migration. collagen or plastic. The mechanism by which this occurs is
unclear, but is likely related to the EC release of a soluble EC stimulation of SMC migration is counterintuitive
factor. At present antibodies specific for various bovine to what one would expect. However migration is just one
integrins are not available; however, future studies need to process in the restenotic process. As we will see in the
be performed to identify the specific integrin compositional upcoming section of this monograph, ECs may limit the
and organizational changes in integrin expression that oc- development of intimal hyperplasia by affecting other
cur in SMCs as a result of ECs. mechanisms in the process, namely by inhibiting SMC
Cell spreading has been shown to correlate more matrix production.
closely with migration rate than cell attachment.34 Whereas
cell attachment is likely to be primarily integrin mediated, Matrix Effect on Endothelial Cell Control
cell spreading appears to involve nonintegrin matrix bind- of Smooth Muscle Cell Matrix synthesis
ing proteins as well.34 Differences in cell spreading on vari- The continued accumulation of extracellular matrix
ous matrix types were similar to those seen in SMC migra- forms the bulk of the mature intimal hyperplastic lesion.1,7
tion (plastic<fibronectin<type I collagen).15 We have shown For the most part, the origin of extracellular matrix is dif-
that ECs increase SMC cell spreading. SMC cell spreading ferentiated SMCs which have migrated into the subintimal
was increased in SMCs cocultured as a bilayer with ECs com- space following vessel wall injury. In addition to increased
pared to SMCs cultured alone. Thus, ECs may alter SMC matrix deposition, the matrix composition of the
matrix adhesion molecule expression or organization to ac- fibroproliferative vascular lesion is altered, containing in-
count for the increased migration in cocultured SMCs. How- creased amounts of the fibrillar collagens such as type I col-
ever, this is indirect evidence and does not exclude the pos- lagen (8). We have shown that ECs can inhibit both SMC
sibility that ECs may stimulate SMC migration by other collagen protein synthesis and type I collagen gene expres-
mechanisms as well. For example, cultured ECs have been sion in bilayer coculture (see Fig. 33.3). It is interesting to
shown to secrete PDGF, a factor which stimulates SMC mi- note that in these studies, ECs inhibited SMC type I col-
gration. This may be an alternative or additional mecha- lagen RNA levels by 50%, which is similar to the 40% EC
nism by which ECs stimulate SMC migration in vitro. inhibition of SMC collagen protein synthesis. The ability of
ECs to inhibit SMC collagen synthesis in vitro suggests that organization and cytoskeletal structure.36 In order to deter-
this may be one mechanism by which ECs may alter vascu- mine the effect of extracellular matrix composition on EC
lar wall remodeling and smooth muscle cell function fol- inhibition of SMC collagen synthesis coculture, we repeated
lowing vascular wall injury in vivo. experiments on various matrix proteins. The extracellular
The mechanism by which ECs inhibit SMC collagen matrix proteins studied were types I and IV collagen,
synthesis in bilayer coculture is unclear. TGF- ! 1 is Matrigel and fibronectin. Type I collagen is a fibrillar col-
upregulated in the injured arterial wall.19,20 It is a potent lagen which is increased in intimal hyperplasia and has been
stimulant for extracellular matrix synthesis, and antibodies shown to increase SMC proliferation. Fibronectin is a high
against TGF- ! 1 have been shown to limit the fibro- molecular weight glycoprotein which is also present in the
proliferative response following balloon angioplasty in ani- extracellular matrix of restenotic lesions and affects SMC
mal models.19 However, in our coculture studies the addi- proliferation and migration. Type IV collagen is a constitu-
tion of TGF-!1 antibody and aprotinin (a plasmin inhibitor ent of the normal EC basement membrane and has been
which prevents the activation of TGF-!1) did not inhibit shown to maintain ECs in a more differentiated state.
SMC extracellular matrix in SMCs cultured alone. This sug- Matrigel is a reconstituted composition of basement mem-
gests that, at least in this in vitro coculture model, EC effects brane proteins laminin and type IV collagen. Laminin is a
on TGF-!1 activation do not play a major role in this pro- component of the EC basement membrane and has been
cess (see Fig. 33.4). Though we did not perform a dose re- shown to maintain ECs in a differentiated state. We have
sponse curve using the TGF-!1 antibody, our studies indi- found that type IV collagen and Matrigel decreased collagen
cate that the antibody did at the least partially inhibit TGF-!1 synthesis in SMCs cultured alone (see Fig. 33.5). These data
based on the diminished SMC hill and valley growth seen in demonstrate that matrix proteins which normally reside in
these cultures. Hill and valley growth has been shown to the EC basement membrane can inhibit SMC matrix syn-
occur as a result of active TGF-!1.35 The plasmin inhibitor thesis. In addition, the data indirectly support the concept
aprotinin completely suppressed hill and valley growth, sug- that EC synthesized matrix proteins could be a possible
gesting complete inhibition of TGF-!1 activation. mechanism by which ECs inhibit SMC matrix synthesis.
Endothelial cells have been shown to secrete various In contrast to the effects of the EC basement mem-
extracellular matrix proteins which could cross the semi- brane matrix proteins, type I collagen, a fibrillar collagen,
permeable membrane and as a result affect SMC function. actually increased collagen synthesis of SMCs cultured alone
Endothelial cells are typically seeded on the membrane for to the greatest degree. In other studies, type I collagen has
4-6 days ( to allow the ECs to grow to confluence), prior to been shown to potentiate SMC dedifferentiation to a syn-
adding SMCs. This allows ample opportunity for ECs to coat thetic phenotype and is abundant in fibroproliferative vas-
the opposite surface of the membrane with matrix proteins. cular lesions.16
Previous investigators have shown that extracellular matrix To determine if differences in cell proliferation and
composition strongly influences SMC integrin expression, thus cell density could account for the differences in collagen
synthesis observed in this study, we examined cell density at Matrix substrate composition can also affect the abil-
the completion of each experiment. As shown in Figure 33.4, ity of ECs to inhibit SMC collagen synthesis.37 We have
the cell density (cell number after three days in culture) of shown that ECs decrease smooth muscle cell collagen syn-
SMCs cultured alone was greatest in cells cultured on thesis when these cells are cocultured together on type I and
Matrigel. There was no difference in cell density when IV collagen and Matrigel (see Fig. 33.6). Fibronectin pre-
fibronectin, type I collagen, and type IV collagen were com- vented the EC inhibition of SMC collagen synthesis, and in
pared; however, collagen synthesis was significantly less in fact actually increased collagen synthesis. Thus, the ability
the type IV collagen group. This suggests that the matrix for grafts sodded with ECs to resist the development of inti-
protein effect on SMC collagen synthesis is due not only to mal hyperplasia may depend upon the matrix substrate the
changes in cell proliferation and cell density, but is extracel- ECs are cultured on.
lular matrix specific. We have not as yet explored the mecha- In conclusion, using an in vitro coculture model, we
nisms by which matrix proteins effect SMC collagen syn- have shown that ECs stimulate SMC migration but inhibit
thesis. As is the case with the effect of matrix proteins on SMC collagen synthesis and transcripts for type I collagen
proliferation and migration, it is likely that specific extra- gene expression. The effect of ECs does not appear to be
cellular matrix proteins interact with cell membrane adhe- mediated through changes in active TGF-!1 levels but may
sion molecules and as a result activate various intracellular be due to secreted matrix proteins. Extracellular matrix pro-
signaling pathways. teins have a profound effect on SMC collagen synthesis and
on the ability of ECs to modulate SMC functions such as
migration and matrix synthesis.
Fig. 33.5. Collagen synthesis (tritiated proline counts per ∝g DNA in conditioned media;
black bars) of SMCs cultured alone on Matrigel (MG) and type IV collagen (CIV) was
decreased when compared to plastic (Control). Fibronectin (FN) and type I collagen (CI)
both significantly increased SMC collagen synthesis when compared to SMCs cultured
on plastic (*p < 0.01 vs. other groups). SMC density is shown in the hatched bars. Cell
density (cell number after three days in culture) was greatest in the SMCs cultured on
MG. There was no difference in cell density when FN, CI and CIV were compared; how-
ever, collagen synthesis was significantly less in the CIV group. This suggests that matrix
effects on SMC collagen synthesis are due not only to changes in cell proliferation and cell
density but are extracellular matrix specific.
Extracellular Matrix Effect on Endothelial Control of Smooth Muscle Cell Migration and Matrix Synthesis 369
12. Zilla P, Deutsch M, Meinhart J et al. Clinical in vitro 26. Fingerle J, Au YP, Clowes AW et al. Intimal lesion for-
endothelialization of femoropopliteal bypass grafts: An mation in rat carotid arteries after endothelial denuda-
actuarial follow-up over three years. J Vasc Surg 1994; tion in absence of medial injury. Arteriosclerosis 1990;
19:540-548. 10(6):1082-87.
13. Zilla P, Fasol R, Callow A. Endothelialization of vascular 27. Reidy MA, Silver M. Endothelial regeneration: Lack of
grafts. Applied Cardiovascular Biology 1990-1991 intimal proliferation after defined injury to rat aorta. AJP
1992;:78-131. 1985; 118(2):173-177.
14. Magometschnigg H, Kadletz M, Vodrazka M et al. Pro- 28. Lewis DA, Lowell RC, Cambria RA et al.. Production of
spective clinical study with in vitro endothelial cell lining endothelium-derived factors from sodded expanded
of expanded polytetratfluoroethylene grafts in crural re- polytetrafluoroethylene grafts. J Vasc Surg 1997;
peat reconstruction. J Vasc Surg 1992; 15:527-535. 25:187-197.
15. Powell R, Carruth J, Basson M et al. Matrix specific ef- 29. Castellot J, Addonizio ML, Rosenberg R et al. Cultured
fect of endothelial cell control of smooth muscle cell mi- endothelial cells produce a heparin like inhibitor of
gration. J Vasc Surg 1996. smooth muscle cell growth. J Cell Biol 1981; 90:372-379.
16. Yamamoto M, Yamamoto K, Noumura T. Type I collagen 30. Fillinger MF, O’Conner S, Wagner RJ et al. The effect of
promotes modulation of cultured rabbit arterial smooth endothelial cell coculture on smooth muscle cell prolif-
muscle cells from a contractile to a synthetic phenotype. eration. J Vasc Surg 1993; 17(6):1058-1067.
Exp Cell Res 1993; 204:121-129. 31. DiMilla P, Stone J, Quinn J et al.. Maximal migration of
17. Streuli CH, Schmidhauser C, Kobrin M et al. Extracellu- human smooth muscle cells on fibronectin and type IV
lar matrix regulates expression of the TGF-!1 gene. J Cell collagen occurs at an intermediate attachment strength. J
Biol 1993; 120(1):253-260. Cell Biol 1993; 122:729-737.
18. Penttinen RP, Kobayashi S, Bornstein P. Transformtin 32. Skinnere M, Raines E, Ross R. Dynamic expression of al-
growth factor-! increases mRNA for matrix proteins both pha 1 beta 1 and alpha 2 beta 2 integrin receptors by
in the presence and in the absence of changes in mRNA human vascular smooth muscle cells. Am J Path 1994;
stability. Proc Natl Acad Sci 1988; 85:1105-1108. 145:1070-1081.
19. Majesky MW, Lindner V, Twardzik DR et al. Production 33. Basson C, Kocher O, Basson M et al.. Differential modu-
of transforming growth factor !1 during repair of arterial lation of vascular cell integrin and extracellular matrix
injury. J Clin Invest 1991; 88:904-10. expression in vitro by TGF-!1 correlates with reciprocal
20. Nikol S, Isner JM, Pickering JG et al. Expression of effects on cell migration. J Cell Phys 1992; 153:118-128.
transformtin growth factor-b1 is increased in human vas- 34. Basson C, Knowles W, Bell L et al.. Spatiotemporal seg-
cular restenosis lesions. J Clin Invest 1992; 90:1582-92. regation of endothelial cell integrin and nonintegrin ex-
21. Powell RJ, Cronenwett JL, Wagner RJ et al. Endothelial tracellular matrix-binding proteins during adhesion events.
modulation of smooth muscle cell phenotype and mac- J Cell Biol 1990; 110:789-801.
roscopic growth pattern. Surg Forum 1993; 44:380-384. 35. Majack RA. Beta-type transforming growth factor speci-
22. Powell RJ, Cronenwett JL, Fillinger MF et al. Effect of fies organizational behavior in vascular smooth muscle cell
endothelial cells and TGF-!1 on cultured vascular smooth cultures. J Cell Biol 1987; 105:465-71.
muscle cell growth patterns. J Vasc Surg 1994; in press. 36. Madri JA, Bell L, Marx M et al. Effects of soluable factors
23. Fillinger MF, Sampson LN, Cronenwett JL, Powell RJ, and extracellular matrix components on vascular cell be-
Wagner RJ. Coculture of endothelial cells and smooth havior in-vitro and in-vivo: Models of de-
muscle cells in bilayer and conditioned media models. J endothelialization and repair. J Cell Biochem 1991;
Surg Res 1997; 67:169-178. 45:123-130.
24. Casscells W. Migration of smooth muscle and endothe- 37. Powell RJ, Hydowski J, Frank O et al.. Endothelial cell
lial cells. Circulation 1992; 89:723-729. effect on smooth muscle cell collagen synthesis. Accepted
25. Bell L, Madri J. Effect of platelets on migration of cul- for publication.
tured bovine aortic endothelial and smooth muscle cells.
Circ Res 1989; 65:1057-1065.
Facilitation of Healing
Cell Entrapment from the Circulation
CHAPTER 34
Circulating Stem Cells: A Fourth Source for the
Endothelialization of Cardiovascular Implants
Willie R. Koen
Introduction
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
372 Tissue Engineering of Prosthetic Vascular Grafts
Later on, spontaneous endothelialization was also Properties of Hematopoietic Stem Cells
observed on other cardiovascular implants such as ventricu- and Progenitor Cells
lar assist devices. In 1982 Bossart13 described a pseudo- A hematopoietic stem cell can be defined as a cell with
neointoma (PNI) consisting of mainly myofibroblasts and self-renewal proliferative potential, coupled with the poten-
endothelial cells on a textured surface of an assist device af- tial to differentiate into progenitor cells of all blood lineages.
ter two years of implantation. This PNI consisted of a ma- Furthermore, a stem cell is typically not terminally differ-
trix layer of collagen bundles embedded in proteinaceous entiated and can divide without limit. However, when it di-
material and interspersed with myofibroblasts. Furthermore, vides, each daughter cell has a choice either to remain a stem
this layer was covered by a monolayer of cells with electron cell, or embark on a course leading irreversibly to terminal
microscopic characteristics of endothelial cells. Based on differentiation. Therefore, the pluripotent stem cell normally
these findings as well as literature evidence that similar fi- generates either more pluripotent stem cells (with self-re-
brous plaques can arise from cultured guinea pig bone mar- newal potential), or committed progenitor cells (also referred
row, Bossart postulated that the source of PNI cells could be to as colony forming cells—CFC). Committed progenitor
multipotential bone marrow stem cells.13 This hypothesis cells are irreversibly determined to produce only one or a
of circulating cells as a source for spontaneously endo- few types of blood cells.20 Moreover, committed progenitor
thelializing grafts was once again brought up by Feigel in cells divide rapidly (amplification divisions) for only a lim-
198514 after a series of experiments investigating the pro- ited number of times, and at the end of this series of divi-
cess of intravascular thrombus organization. He summarized sions they develop into terminally differentiated cells. These
clot organization as a chronological sequence of events which terminally differentiated cells usually have a limited num-
starts off with activation of the mononuclear/macrophage ber of divisions and eventually die after several days. Other
system. Large numbers of early monocytic cells are noted in properties frequently ascribed to stem cells include the abil-
the thrombus, which he subscribed to chemotactic mecha- ity to undergo asymmetric cell divisions as well as the abil-
nisms as well as the ability of monocytic cells to adhere to ity to exist in a mitotically quiescent form.21 During asym-
surfaces. This event of macrophage activation is then fol- metrical division, the stem cells divide into two daughter
lowed by the elongation of monocyte nuclei, the appear- cells, of which one remains a stem cell and the other be-
ance of myofibroblastic cells and eventually endothelial cell comes a differentiated (progenitor) daughter cell. However,
formation, suggesting a transformation of mononuclear cells during symmetrical division of stem cells, the stem cell di-
into endothelial cells.14,16 In 1994 distinct evidence in favor vides into two daughter cells, both of which either remain
of a circulating endothelial cell source was brought forward stem cells or become differentiated progenitor cells. There-
by Shi et al, 15 which he referred to as fallout endo- fore, symmetrical divisions allow the size of the stem cell
thelialization. By using impervious Dacron grafts implanted pool to be regulated.22 Yet, stem cells can also enter a mi-
into different anatomic positions in dogs, a healed surface totic quiescent stage where the divisions take place very
could be demonstrated as early as four weeks after implan- slowly or rarely. This feature of mitotic quiescence is thought
tation. Furthermore, endothelial cell presence was for the to be true for stem cells in the skin and bone marrow. In
first time confirmed by using immunohistochemistry, which contrast, stem cells in the mammalian intestinal crypts have
included von Willebrandt factor staining. been estimated to divide every 12 h.22
With enough evidence at hand that a synthetic sur-
face does have the ability to spontaneously endothelialize The Location of Hematopoietic
simply by being exposed to the circulating blood, the ques- Progenitor Cells
tion remains concerning the origin of these cells. Over the The main source of pluripotential hematopoietic stem
years two basic theories evolved: The first theory is that the cells is the bone marrow microenvironment. This complex
source of fallout endothelialization is cells that detached from space is occupied by hematopoietic cells at all different stages
the vascular wall in areas of high shear stress. This explana- of differentiation. In addition to the hematopoietic cells,
tion has also been supported by the observation that in dy- stroma cells account for the rest of the bone marrow con-
namic tissue culture studies endothelial cells are most prone tent. Stroma cells consist of fibroblasts, endothelial cells,
to detach at the time of mitotic division.17 The second theory adipocytes, monocytes and osteoclasts and can be regarded
is that the fallout endothelial cells arise from circulating pre- as the backbone of the marrow microenvironment. The func-
cursor cells which eventually differentiate into mature en- tion of the stroma is 3-fold namely:
dothelial cells.13-15,18 Although this source has been specu- 1. The production of extracellular matrix;
lated on for many years, it was only recently that published 2. The secretion of cytokines, and
data confirmed that isolated putative progenitor endothe- 3. The mediation of direct cellular contacts regulating
lial cells from the human circulation were able to differenti- hematopoiesis.23
ate in vitro into endothelial cells.19 If this differentiation From the bone marrow, hematopoietic cells enter the
phenomenon is to be the link to a nonthrombogenic tissue blood stream via the bone marrow endothelium (BMEC),
engineered synthetic cardiovascular implant, one needs to which also acts as a gatekeeper in regulating this passage.24
understand stem cell physiology to identify aspects which Furthermore, it has been hypothesized that BMEC is a
could be exploited for facilitated surface endothelialization. unique endothelium that may regulate hematopoiesis by
direct cellular contact and/or expression and secretion of
specific cytokines.25
Circulating Stem Cells: A Fourth Source for the Endothelialization of Cardiovascular Implants 373
Apart from the bone marrow, a second source of he- increase in CD34 cell concentration, suggesting surface colo-
matopoietic stem cells is the peripheral blood. Mature pe- nization with hematopoietic precursor cells.16 Therefore,
ripheral blood cells are derived from a small pool (1-3%) of should these CD34 hematopoietic precursor cells possess the
primitive precursor cells in the bone marrow that bear a ability to mature and differentiate into endothelial cells, then
unique surface glycoprotein, CD34 (Fig. 34.1). Furthermore, the answer to the origin of fall-out endothelialization be-
CD34+ cells are also found at lower concentrations in pe- comes more unclouded. The speculation that circulating
ripheral blood, where they are capable of reconstituting he- hematopoietic precursor (CD34+) cells possess the ability
matopoiesis.26 As a result of this feature, transplantation of to differentiate into endothelial cells was eventually answered
allogeneic peripheral blood stem cells has become a success- by Asahara et al (1997).19 In his study, mononuclear cells
ful and common modality of treatment in clinical practice.27 were isolated from human peripheral blood by magnetic
bead selection on the basis of cell surface antigen expres-
Recognition of Stem Cells sion. In vitro, cells isolated with anti-CD34 or anti-Flk-1
The direct microscopic recognition of stem cells is very antibodies differentiated into endothelial cells. This was con-
intricate and unreliable for various reasons, namely: firmed by the expression of ecNOS, Flk-1/KDR and CD31
1. In bone marrow samples, it is basically impossible mRNA by using the reverse transcription-polymerase chain
to distinguish between distinct precursor cells in their reaction (RT-PCR).
different stages of differentiation; and also
2. In peripheral blood, the concentration of stem cells Control of Stem Cells
is simply too low. The ligand-receptor mechanism remains the most
However, this dilemma has been overcome by the de- commonly researched topic in the control of stem cell dif-
tection of specific surface markers on hematopoietic stem ferentiation. In general, a receptor can be defined as a struc-
cells that can be harnessed for cell recognition. Probably the ture (generally a protein) located on or in a cell, which spe-
most prevalent surface antigen in stem cell isolation at cifically recognizes a binding molecule, a ligand, and thereby
present is cluster derivative 34 (CD34). This glycosylated initiates either a specific biological response (the wide sense
protein has a molecular weight of 115 kDa and is expressed of receptor) or the transduction of a signal (the narrow sense
on human hematopoietic stem and progenitor cells as well of receptor). Furthermore, receptors can be situated in the
as on vascular endothelial cells.26 During vasculogenesis, plasma membrane (receptors for neurotransmitters, growth
hematopoietic stem cells form clusters with angioblasts and factors, sensory stimulants and most circulating hormones),
share common antigenic determinants such as CD34 and in an organelle membrane (e.g., Ca2+ release transduction)
fetal liver kinase-1 receptor (Flk-1).28 Furthermore, the CD34 or in the cytosolic solution (as for the receptors for steroid
surface antigen is also detected in peripheral blood cells, but and thyroid hormones). A receptor of particular interest in
only on 0.03-0.09% of circulating hematopoietic cells.16 the field of tissue engineering as well as stem cell biology
Compared to peripheral blood, the fallout cells noticed on involves the growth factor receptor. By definition, a growth
explanted ventricular assist device surfaces express a 100-fold factor receptor is a cell surface protein whose purpose is to
receive information from outside cells and convey it across nies in soft agar culture. Currently, four major colony-stimu-
the cell membrane. It binds to a receptor and converts the lating molecules are known, namely:
receptor to an active state which then interacts with pro- 1. Granulocyte/macrophage colony-stimulating factor
teins on the inner surface of the membrane. This interac- (GM-CSF);
tion consequently stimulates a program of events leading, 2. Granulocyte colony-stimulatin factor (G-CSF);
amongst other effects, to cell division.29-31 3. Colony-stimulating factor-1 (CSF-1) or macrophage
Growth factor receptors can be classified into three colony stimulating factor (M-CSF); and
groups, namely: 4. Multi-colony stimulating factor (multi-CSF) or
1. The tyrosine kinase receptors. interleukin-3 (IL-3).
2. Seven transmembrane domain receptors; and Interleukin-3 (IL-3) is primarily an activated T cell-
3. The nontyrosine kinase receptors. derived pleiotropic factor (Fig. 34.2) that can stimulate the
The tyrosine kinase receptors have been grouped into proliferation and differentiation of pleuripotent hematopoi-
families on the basis of kinase domains, general structure etic stem cells as well as various lineage-committed progeni-
and ligand structure similarities. Among tyrosine kinase re- tors. Following the purification and cloning of IL-3, it be-
ceptors, the type I receptor family includes the receptors for came apparent that this same protein had been studied un-
epidermal growth factor as well as transformtin growth fac- der different names, including mast cell growth factor, P cell
tor-! (TGF-!). The type II receptors include insulin-like re- stimulating factor, burst promoting activity, multi-colony
ceptors and the type III receptors consist of the platelet-de- stimulating factor, Thy-1 inducing factor and WEHI-3
rived growth factor (PDGF) receptors A and B, c-Kit as well growth factor.32-34
as c-Fms (also known as colony-stimulating factor). Finally, However, more specific growth factor receptors are
the type IV family is a group of receptors whose ligands are now known to be present on stem cells. For example, the
not yet known (Eph, Elk, Eck, Eek and Erk).29-31 fetal liver kinase-1 receptors (Flk-1 in mouse, also known as
The most commonly used ligands in stem cell research KDR in human) are expressed by both the vasculogenic
as well as in clinical practice are the colony stimulating fac- angioblasts and the hematopoietic stem cells as well as vas-
tors (CSF). In bone marrow transplantation, CSF have been cular endothelial cells. Yet, although this receptor is specific
used to mobilize hematopoietic stem cells to optimize the for vascular endothelial growth factor (VEGF) (Fig. 34.3), it
harvest of peripheral blood stem cells. CSF were first de- ceases to be expressed during hematopoietic differentia-
fined by the stimulation of granulocyte/macrophage colo- tion. 28,35 Furthermore, it has been demonstrated in
Flk-1/KDR knockout mice that few, if any, vascular endot- Do Precursor Endothelial Cells Have Self-Renewal
helial cells are present.36 Moreover, Asahara et al (1997) dem- Potential?
onstrated the differentiation of Flk-1 positive precursor cells Per definition, a stem cell has the potential to renew
into VEGF producing vascular endothelial cells, thereby itself. From experiments carried out in baboons it could be
underlining once again the theory of a circulating endothe- demonstrated that highly enriched populations of autolo-
lial cell source. In contrast to the stimulation of stem cells, gous CD34+ marrow cells can restore hematopoiesis and lead
there are also known factors that inhibit the differentiation to a long term recovery of bone marrow function in lethally
and proliferation of stem cells. So far, such inhibiting fac- irradiated baboons.37 Furthermore, it has been known for
tors include nitric oxide (NO) and transformtin growth fac- more than 20 years that, after chemotherapy, circulating
tor-! (TGF-!). myeloid progenitor cells are recruited from the bone mar-
row to the periphery. This recruitment can be further en-
Speculative Application hanced by using colony stimulating factors to produce a more
In summary, our knowledge of specific surface mark- profound mobilization of CD34+ cells. Today, peripheral
ers for circulating progenitor endothelial cells is at this stage blood stem cell (CD34+) transplantation is regarded as
limited to the CD34 antigen as well as the Flk-1/KDR recep- equivalently successful, even superior in certain cases, to bone
tor. Furthermore, VEGF is a specific ligand for the Flk-1/KDR marrow transplantation.38-42 Yet, can we assume that the self-
receptor, as opposed to the less specific ligands for the CD34 renewal of CD34+ stem cells also applies to the precursor
antigen (including !-selectins) (Fig. 34.3). Therefore, circu- endothelial cell? This question was addressed by Hammond
lating progenitor endothelial cells could be addressed by tar- et al: Pigs were lethally irradiated and subsequently trans-
geting the Flk-1 receptor. planted with sibling bone marrow cells. After 6 weeks of
In summary, it seems as if in our striving toward a implantation, hematopoiesis was restored and the explanted
spontaneously healing, long lasting cardiovascular implant, vascular grafts showed the presence of donor related endo-
we could rely on circulating endothelial progenitor cells. Our thelial cells staining positive for factor VIII. Therefore, this
optimism is based on two key facts, namely: study suggests that peripheral blood stem cells exert self-
1. The proof of the presence of circulating progenitor renewal potential to eventually restore hematopoiesis as well
endothelial cells which have the potential to differ- as graft endothelialization.43
entiate into functioning endothelial cells19; and
2. Available receptors on these precursor cells making To What Extent Are Circulating Endothelial Precursor
manipulation with specific ligands possible. Cells Committed?
However, a number of questions still remain open Using fluorescence-activated cell sorting (FACS) the
before one can exploit circulating stem cells for the sponta- CD45 leukocyte common antigen could be demonstrated
neous endothelialization of synthetic impermeable implants. on 94% of freshly isolated CD34+ mononuclear blood cells.
However, attached CD34+ cells, after being in culture for a the further differentiation into CD34+CD45– cells and even-
week on a fibronectin-coated surface, expressed the CD45 tually endothelial cells could be more specifically addressed,
antigen on only 27% of cells.19 Therefore, it seems that en- for instance with VEGF.
dothelial precursor cells circulate as CD34 and CD45 posi-
tive without being finally committed. After attachment, these At What Stage of Differentiation Do Endothelial
noncomitted cells become CD45 negative (after a week) and Precursor Cells Attach to a Surface?
are then committed to further differentiate into endothelial Freshly isolated CD34+ cells attach to a fibronectin
cells. This CD34+CD45+ circulating cell stage reminds us of coated culture flask as early as on day 3 and express at that
the burst forming cell unit (BFU) in erythropoiesis that dif- stage the CD45 antigen in 94% of cells (Fig. 34.4). After a
ferentiates into colony forming cell units (CFU) or, in our week in culture, the CD45 antigen practically disapears.19
case, CD34+CD45– cells. The colony forming cells in turn Therefore, these cells attach at the CD34+CD45+ stage, the
differentiate into erythrocytes, or in our scenario, endothe- burst-forming stage. From here, they could then further dif-
lial cells. Furthermore, it is well described that erythropoi- ferentiate into CD34+CD45– cells, the colony-forming stage,
etin does not act on burst forming unit-erythroid (BFU-E) and eventually endothelial cells, or they can remain
alone, but in concert with other growth factors. The forma- CD34+CD45+ cells and further differentiate into myeloid
tion of BFU-E necessitates the presence of other bioactive leukocytes. This decision could well be influenced by
molecules, originally termed burst-promoting activities, of fibronectin and/or VEGF.
which IL-3 was the first to be well characterized.
Interleukin-3 acts synergistically with erythropoietin to in- A Practical Vision for Future
duce proliferation of BFU-E,44 but does not appear to aug- Cardiovascular Implants
ment the effect of erythropoietin on CFU-E.45 Therefore, Tissue engineering can be defined as the application
with IL-3 also stimulating the formation of circulating he- of engineering disciplines to either maintain existing tissue
matopoietic stem cells (CD34+CD45+), it might well be that structures or to facilitate tissue growth. From the materials
Fig. 34.4. A summary of the proposed endothelial cell pathway. Hematopoietic stem cell in the bone marrow enters the blood
stream and changes its surface status after a) surface attachment as well as b) the presence of IL3, to form a committed endothelial
precursor cell. This precursor differentiates into an endothelial cell in the presence of VEGF.
Circulating Stem Cells: A Fourth Source for the Endothelialization of Cardiovascular Implants 377
engineering point of view, tissues are considered to be cel- 10. Zilla P, Grimm M, Fasol R, Eberl T, Fischlein T, Preiss P,
lular composites representing multiphase systems.46 In or- Krupicka O, Deutsch M, vonOppell U. Growth proper-
der to support tissue engineered tissue, researchers used scaf- ties of cultured human endothelial cells on differently
folds made of various absorbable as well as materials in- coated artificial heart materials. J Thorac Cardiovasc Surg
1991; 101:671-680.
cluding polidioxanone, polyglycolic acid, polyesther
11. Stump MM, Jordan GJ, De Bakey, Halpert B. Endothe-
(Dacron), ePTFE, polyurethanes and collagen.47-49 Today, lium grown from circulating blood on isolated intravas-
scaffold research plays a major role in the overall drive to- cular dacron hub. Am J Path 1963; 43:361-367.
wards tissue engineered vascular implants. Furthermore, the 12. Mackenzie JR, Hackett M, Topuzlu C, Tibbs DJ. Origin
technology of covalent bonding of certain peptides, includ- of arterial prosthesis lining from circulating blood cells.
ing cytokines, to polymeric surfaces holds the promise that Arch Surg 1968; 97:879-886.
scaffolds may play a much more bioactive role in tissue en- 13. Bossart MI, Turner SA, Milam JD, Connor DJ, Urrutia
gineering in the future. It may well be possible to produce a CO, Frazier OH. Multipotential cells in the circulating
synthetic polymeric scaffold for a cardiovascular implant blood: Ultrastructural evidence in the calf. Trans Am Sac
with specific ligands within or on the surface of the struc- Artif Intern Organs 1982; 28:185-189.
ture which elicits a specific biological response. Such a re- 14. Feigl W, Susani M, Ulrich W, Matejka M, Losert U,
Sinzinger H. Organization of experimental thrombosis by
sponse, in our vision, will be the spontaneous endotheli-
blood cells. Virchows Arch (Path Anat) 1985; 406:133-148.
alization of the blood-contacting surface from recruited cir- 15. Shi Q, Wu M, Hayashida N, Werchezac AR, Clowes
culating precursor cells. Therefore, one could hypothesize AW,Sauvage LR. Proof of fallout endothelialization of
that the immobilization of VEGF onto a synthetic surface impervious Dacron grafts in the aorta and inferior vena
could be one possible step towards spontaneous cava of the dog. J Vasc Surg 1994; 20:546-547.
endothelialization, as it may promote stem cell attachment 16. Rafii S, Oz MC, Seldomridge JA, Ferris B, Asch A,
and differentiation, as well as maintenance. Nachman RL, Shapiro F, Rose EA, Levin HR. Character-
However, since surface endothelialization will even- ization of haematopoietic cells arising on the textured
tually need a mature underlying connective tissue matrix, surface of ventricular assist devices. Ann Thorac Surg
the wide open question regarding the existence of circulat- 1995; 60:1627-32.
17. Werchezac AR, Viggers RF, Coan DE, Savauge LR. Mito-
ing cells promises the next exciting chapter in this field.
sis and cytokinesis in subconfluent endothelial cells ex-
posed to increasing levels of shear stress. J Cell Physiol
References 1994; 159:83-91.
1. Berger K, Sauvage LR, Rao AM, Wood SJ. Healing of ar- 18. Dasse KA, Chipman SD, Sherman CN, Levine AH, Frazier
terial prostheses in man: Its incompleteness. Ann Surg OH. Clinical experience with textured blood contacting
1972; 175:118-127. surfaces in ventricular assist devices. Trans Am Sac Artif
2. Greisler HP. Arterial regeneration over absorbable pros- Intern Organs 1987; 33:428-425.
theses. Arch Surg 1985; 120:315-323. 19. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee
3. Greisler HP, Ellinger J, Schwartz TH, Golan J, Raymond R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isola-
RM, Kim DU. Arterial regenration over polydioxanone tion of putative progenitor endothelial cells for angiogen-
prosthesis in the rabbit. Arch Surg 1987;122:715-721. esis. Science 1997; 275:964967.
4. Greisler HP, Schwartz TH, Ellinger J, Kim DU. Dacron 20. The Cell differentiated cells and the mainteninace of tissue.
inhibition of arterial regenerative activity. J Vasc Surgery 21. Hall PA, Watt FM. Stem cells: The generationand main-
1986; 3:747-756. tenance of cellular diversity. Development 1989;
5. Zilla P, Fasol R, Dudeck U, Siedler S, Preiss P, Fischlein 106:619-633.
T, Muller-Glauser W, Baitella G, Sanan D, Odell J, 22. Potten CS, Loeffler M. Stem cells: Attributes, cycles, spi-
Reichart B. In-situ cannulation, microgrid follow up and rals, pitfalls and uncertainties. Lessons for and from the
low density plating provide first passage endothelial cell crypt. Development 1990; 110:1001-1020.
mass-cultures for in-vitro lining. J Vasc Surg 1990; 23. Wichkramasinghe. Observations on the ultrastructure of
12:180-89. sinusoids and reticular cells in human bone marrow. Clin
6. Kadletz M, Moser R, Preiss P, Deutsch M, Zilla P, Fasol Lab Haematol 1991; 13:263-269.
R. In vitro lining of fibronectin coated PTFE grafts with 24. Weiss L, Chen LT. The organization of haematopoietic
cryopreserved saphenous vein endothelial cells. Thorac cords and vascular sinusses in bone marrow. Blood Cells
Cardiovasc Surgeon 1987; 35:143-147. 1975; 1:617.
7. Fasol R, Zilla P, Deutsch M, Grimm M, Fischlein T, Laufer 25. Rafii S, Shapiro F, Rimarachin J, Nachman RL, Ferris B,
G. Human endothelial cell seeding: Evaluation of its ef- Weksler B, Moore M, Asch A. Isolation and characteriza-
fectiveness by platelet parameters after one year. J Vasc tion of human bone marrow microvascular endothelial
Surg 1989; 9:432-436. cells: Hematopoietic progenitor cell adhesion. Blood 1994;
8. Zilla P, Deutsch M, Meinhart J, Eberl T, Puschmann R, 84(1):10-19.
Minar E, Dudczak R, Lugmaier H, Schmidt P, Fischlein 26. Sutherland DR, Stewart AK, Keating A. CD34 antigen:
T. In vitro endothelialization of femoro-popliteal bypass Molecular features and potential clinical applications.
grafts: An actuarial follow up after 3-years J Vasc Surg Stem Cells; 1993; 11(suppl 3):50-57.
1994; 19:540-548. 27. Besinger WI, Appelbaum FA, Demirer T, Torok-Storb B,
9. Fasol R, Zilla P, Deutsch M, Fischlein T, Kadletz M, Storb R, Buckner CD. Transplantation of allogeneic
Griesmacher A, Mller MM. Endothelialization of artifi- peripheral blood stem cells. Stem Cells 1995; Dec 13 suppl
cial surfaces: Does surface tension determine in vitro 3:63-70.
growth of HSVEC? J Tex Heart Inst 1987; 14:119-126.
378 Tissue Engineering of Prosthetic Vascular Grafts
28. Matthews W, Jordan CT, Gavin M, Jenkins NA, Copeland patients following high-dose chemotherapy with autolo-
NG, Lemischka IR. A receptor tyrosine kinase cDNA iso- gous hematopoietic progenitor cell support. Blood 1992;
lated from a population of enriched primitive 80(suppl 1):24.
hemaropoietic cells and exhibiting close genetic linkage 39. Shpall EJ, Jones RB, Franklin W et al. Transplantation of
to c-kit. Proc Natl Acad Sci USA 1991; 88:9026. CD34+ marrow and/or peripheral blood progenitor cells
29. Albert B, Bray D, Lewis J, Raff M, Roberts K, Watson JD. (PBPC) into breast cancer patients following high dose
Cell signaling In: Albert B, Bray D, Lewis J, Raff M, Rob- chemotherapy. Blood 1994; 84(suppl 1):396.
erts K, Watson JD, eds. Molecular Biology of the Cell, 40. Shpall EJ, Jones RB, Franklin W et al. Transplantation of
2nd edition. New York & London: Garland Publishing, enrichedCD34+ autologousmarrow into breast cancer
Inc., 1989:681. patients following high-dose chemotherapy: Influence of
30. Lodish H, Baltimore D, Berk A, Zipursky SL, Matsudaira CD34+ peripheral blood progenitors and growth factors
P, Darnell J. Cell-to-cell signalling: Hormones and recep- on engraftment. J Clin Oncol 1994; 12:28.
tors In: Lodish H, Baltimore D, Berk A, Zipursky SL, 41. Jones HM, Jones SA, Watts MJ et al. Development of a
Matsudaira P, Darnell J. Molecular cell biology, 3rd edi- simplified single apheresis approach for peripheral blood
tion. New York: Scientific American Books, Inc., 1995:853. progenitor cell transplantation in previously treated pa-
31. Kendrew J. Receptor In: The Encyclopedia of Molecular tients with lymphoma. J Clin Oncol 1994; 12:661.
Biology Oxford,: Blackwell Science Ltd., 1994:939. 42. Pettengell R, Morgenstern GR, Woll PJ et al. Peripheral-
32. Quesenberry PJ. Hemopoietic stem cells, progenitor cells, blood progenitor cell transplantation in lymphoma and
and cytokines. 211. leukemia using a single apheresis. Blood 1993; 82:3770.
33. Bagley CJ, Woodcock JM, Stomski FC, Lopez AF. The 43. Hammond W. NAVBO Satelite Symposium on fallout
structural and functional basis of cytokine receptor acti- healing. Seattle, WA Sept 1996 [ABS].
vation: Lessons from the common beta subunit of the 44. Migliaccio G, Migliaccio AR, Visser JW. Synergism be-
granulocyte-macrophage-colony-stimulating factor, tween erythropoietin and IL-3 in the induction of hemato-
interleukin-3 and interleukin-5 receptors. Blood 1997; poietic stem cell proliferation and erythroid burst colony
89(5):1471. formation. Blood 1988; 72:944.
34. Hara T, Miyajima A. Function and signal transduction 45. Umemura T, Papayannopoulou T, Stamatoyannopoulos
mediated by interleukin-3 receptor system in hematopoie- G. The mechanism of expantion of late eryhtroid progeni-
sis. Stem Cells 1996; 14(6):605-618. tors during erythroid generation: Target cells and the ef-
35. Millauer B, Longhi MP, Plate KH, Shawver LK, Risau W, fect of IL-3 and erythropoietin. Blood 1989; 73:1993.
Ullrich A, Strawn LM. Dominant-negative inhibition of 46. Wintermantel E, Mayer J, Blum J, Eckert KL, Lusher P,
Flk-1 suppresses the growth of many tumor types in vivo. Mathey M. Tissue engineering scaffolds using superstruc-
Cancer Research 1996; 56(7):1615-20. tures. Biomaterials 1996; 17(2):83-91.
36. Fong GH, Rossant J, Gertsenstein M, Breitman ML. Role 47. Greisler HP. Arterial regeneration over absorbable pros-
of Flt-1 receptor tyrosine kinase in regulating the assem- theses. Arch Surg 1985; 120:315-323.
bly of vascular endothelium. Nature 1995; 367:66-70. 48. Greisler HP, Ellinger J, Schwartz TH, Golan J, Raymond
37. Berenson RJ, Andrews RG, Bensinger WI et al. Antigen RM, Kim DU. Arterial regeneration over polydioxanone
CD34+ marrow cells engraft lethally irradiated baboons. prosthesis in the rabbit. Arch Surg 1987; 122:715-721.
J Clin Invest 1988; 81:951. 49. Greisler HP, Schwartz TH, Ellinger J, Kim DU. Dacron
38. Shpall EJ, Jones RB, Franklin W et al. CD 34+ marrow inhibition of arterial regenerative activity. J Vasc Surg
and/or peripheral blood progenitor cells (PBPCs) provide 1986; 3:747-756.
effective hematopoietic reconstitution of breast cancer
Facilitation of Healing
Cell Entrapment from the Circulation
CHAPTER 35
Surface Population with Blood-Borne Cells
William P. Hammond
Background
T he developmental biology of blood vessels has long been believed to consist of two phases.
The first, vasculogenesis, is the development during embryologic life of the original pre-
cursors to all vessels.1 Our understanding of this phenomenon has been built around detailed
studies of the development of chick and quail embryos, and the elegant tracing of cellular
movement during early development. Angiogenesis, on the other hand, is the term applied
to development of new blood vessels during adult life, which largely serves a reparative func-
tion.2 In addition to wound healing, this form of new blood vessel formation by sprouting
from existing blood vessels is believed to explain the development of vessels to feed growing
tumor cell masses. The understanding of regulatory factors for this form of new vessel growth
in the adult has been a principal theoretical model for the last few decades.
The introduction of vascular prostheses prompted study of the basic biology through
which these grafts maintain patency and integrity as blood conduits. As early as 1955, Sauvage
and Wesolowski had noted the apparent importance of tissue ingrowth to prosthesis perfor-
mance.3 Wesolowski’s experimental studies convinced him of the importance of porosity to
tissue ingrowth and prompted him to develop a high porosity knitted prosthesis.4,5 In 1962,
Florey et al observed islands of endothelium around mouths of vascular channels seen on
the flow surface of knitted Dacron grafts implanted in the baboon.6 Although they could
not determine the origin of these channels, they noted that they appeared to connect with
perigraft vessels. In 1985 Greisler et al demonstrated arterial regenerative activity in
polyglycolic acid absorbable aorta replacements in the rabbit,7 and in 1986, Clowes and
colleagues demonstrated in the baboon the capacity of a special porous form of PTFE, with
an internodal distance of 90 ∝m, to admit the development of transmural microvessels from
the perigraft tissue;8 this suggested that the usual tissue response to the placement of vascu-
lar prostheses involves a local variant of angiogenesis, which subsequently leads to endothe-
lial resurfacing of the graft, thereby contributing to its patency. In 1989 Kogel et al studied
transprosthetic vascularization in the dog, using a polyester-based resin to make three di-
mensional casts, and concluded that midgraft endothelialization in PTFE grafts with 60 ∝m
internodal distances was attributable to ingrowth of microvessels from the perigraft tissue
through the graft and into the lumen; this phenomenon was not observed in microporous
polyurethane grafts.9 In 1990 Golden and Clowes et al reported the importance of graft
porosity to healing of PTFE prostheses in the baboon; they found that a 60 ∝m internodal
distance produced optimal endothelial coverage.10 In 1995, Bull et al also reported midgraft
endothelialization of 60 ∝m internodal distance PTFE grafts implanted in the dog carotid,
and attributed it to microvessel ingrowth to the flow surface; they also noted that an external
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
380 Tissue Engineering of Prosthetic Vascular Grafts
jugular vein wrap around the grafts produced endo- dothelial precursor cell and suggest several implications for
thelialization as early as 7 days.11 In 1996 our center reported the engineering of vascular prostheses in the future.
histological demonstration of the continuity of a microvessel
between the perigraft vessels and flow surface microostia in Impervious Grafts
a porous Dacron prosthesis; however, we were unable to In 1994, Shi and Sauvage et al reported the results of a
determine whether its origin was in the outer wall or on the study designed to demonstrate unequivocally that perigraft
inner flow surface of the graft.12 tissue ingrowth is the sole source of healing beyond the pan-
In 1963, Stump et al described the presence of endot- nus areas.19 Dacron grafts were made impervious by a coat-
helium on Dacron hubs suspended in the descending tho- ing of silicone rubber and shielded from pannus ingrowth
racic aorta of young pigs, and cited this as evidence for for- (which is very limited in humans and dogs)20 by 2.5-6 cm
mation of endothelium from circulating cells in the blood.13 lengths of microporous (30 ∝m internodal distance) PTFE
More recently, Scott et al found endothelium on the sur- at each end. Suitable lengths of this composite graft were
faces of pledgets of vascular graft material implanted in dog implanted in the descending thoracic aorta, abdominal aorta
aortas.14 A direct demonstration of endothelium in the blood and inferior vena cava of the same dog. At the end of the
stream was developed by Sbarbati et al using an endothe- implant periods, light microscopy study confirmed that there
lial-specific monoclonal antibody to analyze patients’ arte- was no perigraft tissue ingrowth (Fig. 35.1). Instead of the
rial and venous blood before and after heart catheteriza- expected unhealed flow surface, by 4 weeks all these imper-
tion,15 and in the following year George et al identified vious grafts had scattered islands of endothelialization
postangioplasty circulating human endothelial cells with (Fig. 35.2). We named this phenomenon “fall-out” healing,
S-Endo1 coupled to immunomagnetic beads.16 Further evi- because the only possible source appeared to be the blood
dence of circulating endothelium has been supplied by flowing through the graft.
Frazier’s17 and Rafii’s18 studies of human endothelial cells The experimental study was extended to include 70 cm
found on the lining of ventricular assist devices. long impervious grafts implanted in the carotid-femoral
position in dogs.21 At 3 months, each of the 5 extra-ana-
Previous Studies tomic grafts that remained patent had results similar to the
The following studies have produced data that, taken aorta and inferior vena cava grafts, with scattered endothe-
together, strongly support the concept of a circulating en- lial islands in the midgraft area despite a lack of ingrowth
from the outer wall.
Fig. 35.1. NOTE: See color insert for color representation. Blockage of pannus and transmural ingrowth for study of
fallout endothelialization of isolated central Dacron limb of a prosthesis made impervious by a coating of silicone rubber
and implanted for 8 weeks in the descending thoracic aorta of a dog. (A) No transmural ingrowth from perigraft tissues
and limited pannus from native aorta onto PTFE (LM, x50). (B) PTFE graft, beyond the pannus zone, showing no perigraft
tissue growth into the graft (LM, x25). (C) No perigraft tissue ingrowth into the impervious Dacron graft, but presence of
endothelial-like cells on flow surface, with #-actin positive smooth muscle cells beneath (H&E; LM, x50). Reprinted with
permission from Shi et al. Proof of fallout endothelialization of impervious Dacron grafts in the aorta and inferior vena
cava of the dog. J Vasc Surg 1994; 20:549.
Surface Population with Blood-Borne Cells 381
Fig. 35.2. NOTE: See color insert for color representation. Findings for specimens taken from areas beyond the pannus zone of
Dacron grafts made impervious to perigraft tissue ingrowth by a silicone rubber coating and implanted in the dog. (A)-(L)
Endothelial-like cells on flow surfaces of Dacron prostheses implanted in the descending thoracic aorta, abdominal aorta and
inferior vena cava of the dog: (A)-(C) LM, AgNO3, x70; (D)-(F) LM, x500; (G) SEM, x1000; (H) SEM, x500; (I) SEM, x3000;
(J)-(L) TEM, x4000. (M)-(O) Proof of endothelium by FVIII/vWF positivity on flow surface of isolated Dacron grafts: (M)-
(N) x500; (O) x1000. Reprinted with permission from Shi et al. Proof of fallout endothelialization of impervious Dacron
grafts in the aorta and inferior vena cava of the dog. J Vasc Surg 1994; 20:550.
382 Tissue Engineering of Prosthetic Vascular Grafts
woven grafts had endothelium in areas with no tissue in- area. Similar results were obtained in rabbits. Although they
growth.29 The endothelium in all 4 instances was located in refer to this process as “therapeutic angiogenesis”, it suggests
the midgraft area, far from any possible extension of pan- a form of vasculogenesis, as do our experimental and clinical
nus ingrowth from the native vessel. findings of endothelialization of synthetic flow surfaces in
The occurrence rate for endothelialization of human the absence of blood vessel ingrowth. If hematopoietic
vascular grafts is, of course, impossible to estimate. How- progenitor cells can be shown to differentiate into endothelial
ever, the usual failure to find it in explanted specimens may cells, the case for an adult form of vasculogenesis will
be due more to the amount of elapsed time after death be- be strengthened.
fore removal and preservation than to the actual frequency:
The 3 specimens we have reported were all fixed either im- Implications
mediately or within 5 hours after death. The implications of these findings are potentially far
The possibility of clinical graft endothelialization raises reaching. In basic science our understanding of the devel-
the question of whether it would be possible to encourage opment of new blood vessels, and in particular their endo-
this phenomenon, either through treatment of the graft or thelial cell linings, would be transformed by a new under-
the patient. standing of the origins of these cells. The existence of a cir-
Recently, Asahara et al reported in vitro magnetic bead culating “angioblast” would significantly alter how one ap-
studies in which they isolated putative endothelial cell proaches disorders of new blood vessel formation, includ-
progenitors from human peripheral blood.30 They then in- ing the management of tumor metastases. Should we dis-
jected DiI-labeled CD34 positive mononuclear blood cells cover the existence of a true multipotential “hemangioblast”,
into the tail veins of mice with unilateral ischemia of a hind it would be necessary to dramatically reshape our concepts
limb; later examination demonstrated proliferative DiI- of angiogenesis and vasculogenesis. In fact, definitive proof
labeled cells in the reparative vascularization of the ischemic of the presence of a mesenchymal precursor for both he-
Surface Population with Blood-Borne Cells 385
matopoietic cells and vascular cells would suggest an adult grafts is influenced by graft porosity. J Vasc Surg 1990;
version of vasculogenesis which would profoundly alter the 11:838-845.
view of both normal physiology of the vasculature as well as 11. Bull DA, Hunter GC, Holubec H, Aguirre ML, Rappaport
pathologic changes in blood vessels. WD, Putnam CW. Cellular origin and rate of endothelial
cell coverage of PTFE grafts. J Surg Res 1995; 58:58-68.
In the clinical realm the application of these concepts
12. Wu MH-D, Shi Q, Onuki Y et al. Histologic observation
could be very important. Many efforts over the past decade of continuity of transmural microvessels between the
or so employing techniques for “seeding” of vascular grafts perigraft vessels and flow surface microostia in a porous
would have to be rethought in light of new concepts in stem vascular prosthesis. Ann Vasc Surg 1996; 10:11-15.
cell biology applied to endothelium, and possibly smooth 13. Stump MM, Jordan GL Jr, De Bakey ME, Halpert B. En-
muscle cells as well. The potential for specific identification dothelium grown from circulating blood on isolated in-
of cells giving rise to new endothelial surfaces could signifi- travascular Dacron hub. Am J Pathol 1963; 43:361-367.
cantly alter the basic approaches to design of experiments 14. Scott SM, Barth MG, Gaddy LR, Ahl ET Jr. The role of
using seeding. A second clinical implication is that the evo- circulating cells in the healing of vascular prostheses. J
lution of design concepts in vascular prostheses has moved Vasc Surg 1994; 19:585-593.
from “bioinert” to “biocompatible” materials,31 and will al- 15. Sbarbati R, deBoer M, Marzilli M, Scarlattini M, Rossi G,
van Mourik JA. Immunologic detection of endothelial cells
most surely need to evolve further to a concept of
in human whole blood. Blood 1991; 4:764-769.
“biospecific” materials. For example, where a goal in the past 16. George F, Brisson C, Poncelet P et al. Rapid isolation of
may have been the synthesis of biopolymers that failed to human endothelial cells from whole blood using S-Endo1
activate coagulation processes of blood and subsequently monoclonal antibody coupled to immuno-magnetic beads:
moved toward materials that are capable of passive, Demonstration of endothelial injury after angioplasty.
nonactivating interactions with their surroundings, the Thromb Haemost 1992; 67:147-53.
present studies suggest a need to develop materials with spe- 17. Frazier OH, Baldwin RT, Eskin SG, Duncan JM. Immu-
cific affinities for the circulating cells, which would also pro- nochemical identification of human endothelial cells on
mote both their landing and growth in situ on the appro- the lining of a ventricular assist device. Texas Heart Inst
priate graft materials. If circulating endothelial progenitor J 1993; 2:78-82.
18. Rafii S, Oz MC, Seldomridge JA, Ferris B, Asch AS,
cells truly exist, the clinical approach to accelerated healing
Nachman RL, Shapiro F, Rose EA, Levin HR. Character-
of prostheses will be totally transformed, and the tissue en- ization of hematopoietic cells arising on the textured sur-
gineering required for vascular graft prostheses will be en- face of left ventricular assist devices. Ann Thorac Surg
tirely different in the future. 1995; 60:1627-1632.
19. Shi Q, Wu MH-D, Hayashida N et al. Proof of fallout
References endothelialization of impervious Dacron grafts in the aorta
1. Risau W, Flamme I. Vasculogenesis. Ann Rev Cell Dev and inferior vena cava of the dog. J Vasc Surg 1994;
Biol 1995; 11:73-91. 20:546-57.
2. Rolokman J, Shing Y. Angiogenesis. J Biol Chem 1992; 20. Sauvage LR, Berger KE, Wood SJ et al. Interspecies heal-
267:10931-10934. ing of porous arterial prostheses. Observations, 1960 to
3. Sauvage LR, Wesolowski SA. The healing and fate of ar- 1974. Arch Surg 1974; 109:698-705.
terial grafts. Surgery 1955; 38:1090-1131. 21. Kouchi Y, Onuki Y, Wu MH-D et al. Apparent blood
4. Wesolowski SA, Fries CC, McMahon JD, Martinez A. stream origin of endothelial and smooth muscle cells in
Evaluation of a new vascular prosthesis with optimal the neointima of long, impervious carotid-femoral grafts
specifications. Surgery 1966; 59:40-56. in the dog. Ann Vasc Surg, in press.
5. Wesolowski SA, Sauvage LR, Golaski WM, Komoto Y. 22. Onuki Y, Hayashida N, Wu MH-D et al. Accelerated
Rationale for the development of the gossamer small ar- endothelialization model for the study of Dacron graft
terial prosthesis. Arch Surg 1968; 97:864-871. healing. Ann Vasc Surg 1997; 11:141-48.
6. Florey HW, Greer SJ, Kiser J, Poole JCF, Telander R, 23. Onuki Y, Kouchi Y, Yoshida H et al. Early flow surface
Werthessen NT. The development of the pseudointima endothelialization before microvessel ingrowth in accel-
lining fabric grafts of the aorta. Br J Exp Pathol 1962; erated graft healing, with BrdU identification of cellular
43:655-60. proliferation. Ann Vasc Surg, 1998; 12:207-215.
7. Greisler HP, Kim DU, Price JB, Voorhees AB Jr. Arterial 24. Onuki Y, Kouchi Y, Yoshida H et al. Early presence of
regenerative activity after prosthetic implantation. Arch endothelial-like cells on the flow surface of porous arte-
Surg 1985; 120:315-323. rial prostheses implanted in the descending thoracic aorta
8. Clowes AW, Kirkman TR, Reidy MA. Mechanisms of ar- of the dog. Ann Vasc Surg, 1997; 11:604-11.
terial graft healing. Rapid transmural capillary ingrowth 25. Sauvage LR, Berger K, Beilin LB, Smith JC, Wood SJ,
provides a source of intimal endothelium and smooth Mansfield PB. Presence of endothelium in an axillary-
muscle in porous PTFE prostheses. Am J Pathol 1986; femoral graft of knitted Dacron with an external velour
123:220-230. surface. Ann Surg 1975; 182:749-753.
9. Kogel H, Amselgruber W, Frosch D, Mohr W, Cyba- 26. Wu MH-D, Shi Q, Wechezak AR et al. Definitive proof
Altunbay S. New techniques of analyzing the healing pro- of endothelialization of a Dacron arterial prosthesis in a
cess of artificial vascular grafts, transmural vascularization, human being. J Vasc Surg 1995; 21:862-67.
and endothelialization. Res Exp Med 1989; 189:61-68. 27. Montesano R, Orci L, Vassalli P. In vitro rapid organiza-
10. Golden M, Hanson SR, Kirkman TR, Schneider PA, tion of endothelial cells into capillary-like networks is
Clowes AW. Healing of polytetrafluoroethylene arterial promoted by collagen matrices. J Cell Biol 1983;
97:1648-52.
386 Tissue Engineering of Prosthetic Vascular Grafts
28. Deutsch M, Meinhart J, Vesely M et al. In vitro 30. Asahara T, Murohara T, Sullivan et al. Isolation of puta-
endothelialization of expanded polytetrafluoroethylene tive endothelial cells for angiogenesis. Science 1997;
grafts: A clinical case report after 41 months of implanta- 275:964-67.
tion. J Vasc Surg 1997; 25:757-63. 31. Hubbell JA. Engineering the cellular-synthetic substrate
29. Shi Q, Wu MH-D, Onuki Y et al. Endothelium on the interface. How to build a blood vessel; Lifeline Founda-
flow surface of human aortic Dacron vascular grafts. J tion Research Initiatives in Vascular Disease Conference,
Vasc Surg 1997; 25:736-42. Bethesda, February 27, 1997, Program Book, abstract 94.
Facilitation of Healing
Cell Entrapment from the Circulation
CHAPTER 36
Cellular Population of the Textured-Surface Left Ventricular
Assist Devices Leads to Sustained Activation of a
Procoagulant and Proinflammatory Systemic Response
Talia B. Spanier, Ann Marie Schmidt, Mehmet C. Oz
Overview
T he use of LVAD technology has a potentially critical role in the management of patients
with end-stage cardiac failure.1-6 The ability of this device to enhance left ventricular
function as a bridge to transplantation or for longer periods is well established. However, as
with any foreign material implanted into a host, the issue of host-graft interaction becomes
an integral part of LVAD biology, beyond its mechanical function as a pump. Compared
with early design LVADs, whose surface was mainly of smooth contour, the
Thermocardiosystems HeartMate LVADTM was designed with a textured surface polyure-
thane diaphragm, intended to minimize the potential for thromboembolic complications
by facilitating the formation of a tightly adherent “pseudoneointima” on the blood-contact-
ing surface7-12 (Fig.36.1). The goal of this strategy was to reduce the risk of development of
thromboemboli by eliminating direct contact between the device and circulating blood.11
The apparent success of this modification in LVAD design was suggested by studies which
reported a thromboembolic rate of < 2% associated with use of this modified surface.12
Furthermore, patients receiving this device were apparently safely maintained with no or
minimal forms of systemic anticoagulation.
The implantation of this device with its subsequent cellular population and creation
of a biological lining, however, has significant impact on its host, with both procoagulant
and proinflammatory consequences. Of considerable interest, we believe that so few in-
stances of clinically apparent thromboembolic phenomena are seen because of the creation
of a delicate balance between the generation of thrombin and activation of fibrinolytic path-
ways which occurs as a result of device implantation. Furthermore, it is tempting to specu-
late that perturbations of vascular homeostasis associated with infection, inflammation or
surgery, for example, might easily tip the balance of hemostatic mechanisms to formation of
thrombi or consumptive coagulopathy. Thus, dissecting the mechanisms by which
prothrombotic tendencies, anticoagulant defenses and fibrinolysis are balanced in this LVAD
environment become essential for understanding clinically relevant prothrombotic disor-
ders in these patients.
The development of clinically important immune alterations in patients related to the
implantation of the textured surface LVADs also is of significant interest. It has been ob-
served that LVAD patients develop increased levels of anti HLA antibodies13-15 in a time-
dependent manner over the course of LVAD implantation.15 In our own experience, the
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
388 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 36.1. LVAD. Left Panel. Schematic illustration of the LVAD implanted in a patient. The pump is positioned intraperitoneally in
the upper left quadrant of the abdomen. Dacron conduits are used to connect the pump between the apex of the left ventricle and
the ascending thoracic aorta. The LVAD is either pneumatically or electrically driven via a percutaneous drive line powered by an
external console or battery pack, respectively. Porcine valves placed in the inflow and outflow conduits ensure the unidirectional
flow of blood. Right Panel. The blood-contacting surfaces of both the titanium housing component and the flexible polyurethane
diaphragm are textured to encourage the formation and adherence of a biological lining. The flexible diaphragm consists of a
fibrillar surface integral with the base material to eliminate detaching of fibrils. The individual fibrils measure approximately
18 ∝m in diameter to 300 ∝m in length. The nonflexible housing surface is fabricated from titanium microspheres sintered together
to form a porous nonpermeable topography. The diameter of the spheres ranges from 50-75 ∝m, with a resultant pore size ranging
from 25-50 ∝m.
clinical consequences of these antibodies are significant and Thrombin Generation and Fibrinolysis
unique to each class of antibody produced. Anti-MHC class In order to better understand the hematologic profile
I antibodies, measured in the panel reactive antibody assay which underlies the low thromboembolic risk associated
(PRA), are associated with a prolonged waiting time to trans- with the textured surface LVAD, and to more fully under-
plantation because of difficulty in finding a negative stand the long term physiologic implications of its use, we
crossmatched donor organ. Anti-MHC class II antibodies analyzed measurements of thrombin generation and fibrin-
are associated with increased severity and frequency of cel- olysis in patients supported by the TCI LVAD. Our data in-
lular rejection episodes after transplantation. Of note, our dicate that despite both apparent clinical stability and “nor-
data do not suggest that these findings are related to such mal” screening values of routine hemostatic parameters such
factors as number of blood transfusions, use of filtered blood, as platelet count, prothrombin and activated partial throm-
age or gender. However, we propose that one potential boplastin times, patients with textured surface LVADs nev-
perturbant in this setting may be the presence of CD34 posi- ertheless demonstrate significant activation of coagulation
tive cells and other activated inflammatory cells populating with secondary fibrinolysis.16 The etiology (ies) underlying
the LVAD. We believe, in fact, that interactions between the these hemostatic and immune abnormalities are of consid-
LVAD surface and host mononuclear cells leads to a Th2 erable interest. Such LVAD-specific etiologies as the textured
pattern of cytokine expression by activated T cells, which polyurethane surface, the titanium housing, and the dacron
contributes to B cell hyperreactivity in vivo (Fig.36.2). These inflow / outflow lines need to be considered. Other etiolo-
interactions ultimately culminate in immune alterations in gies, such as altered flow characteristics and the presence of
the host, further emphasizing the necessity of understand- preexisting clots in the failed ventricles, must also be inves-
ing the identity and functional significance of the cellular tigated (Fig. 36.3). Our data do not suggest that these find-
populations of the LVAD surface. ings are a direct result of the surgical implantation proce-
Sustained Activation of a Procoagulant and Proinflammatory Systemic Response 389
dure of the LVAD, as hemostatic findings remain abnormal macrophage procoagulant activity (Fig. 36.5B), is consistent
at least up to the 335th day of LVAD therapy. Jeevanandam with the hypothesis that the microenvironment created by
and colleagues have shown that in the perioperative period, the LVAD was indeed reflective of a systemic pro-
compared with coronary artery bypass graft (CABG) pa- thrombotic effect.
tients, LVAD patients consistently demonstrate significantly
higher markers of thrombin generation (TAT and F1+2) and LVAD Surface Cellularization
fibrinolysis (D-dimers and FDPs).17 Our studies examining Indeed, these data suggest that a specific characteris-
LVAD patients in this regard do not, however, demonstrate tic of the LVAD textured surface itself is, at least in part, re-
that these perioperative findings are sustained, as the overt sponsible for these observations. The blood contacting sur-
indices of thrombin generation and fibrinolysis are acute faces of both the titanium housing component and the flex-
and procedure-related. In fact, prospective analysis of 20 ible polyurethane diaphragm are textured to encourage the
LVAD patients from the time of LVAD insertion for up to formation and adherence of a biological lining (Fig. 36.1,
335 days revealed an initial rise in markers of thrombin gen- right panel). The flexible diaphragm consists of a fibrillar
eration as measured by elevated levels of prothrombin frag- surface integral with the base material to eliminate detach-
ment 1+2 (F1+2) and thrombin-antithrombin III complex ing of fibrils. The individual fibrils measure approximately
(TAT). The data demonstrated an increase in each of these 18 ∝m in diameter to 300 ∝m in length. The nonflexible hous-
parameters in the immediate perioperative period which ing surface is fabricated from titanium microspheres sin-
declined progressively by days 5-7. Subsequently, indices of tered together to form a porous nonpermeable topography.
thrombin generation and fibrinolysis rose in a time-depen- The diameter of the spheres ranges from 50-75 ∝m, with a
dent manner, reaching an apparent maximum by day 35 resultant pore size ranging from 25-50 ∝m.9
which was sustained through at least day 335 (Fig 36.4A,B). Although either the sintered titanium surface or the
Similar results were observed for levels of D-dimers polyurethane diaphragm should in theory be capable of cel-
(Fig. 36.4C). lular entrapment, we and others have demonstrated in stud-
These data suggest that activation of coagulation in ies of explanted LVADs considerable cellular entrapment
LVADs is biphasic. Initial / immediate activation of coagu- specifically by the polyurethane diap-Bposits on the sintered
lation and fibrinolysis in the perioperative period is likely titanium housing.18-22 Flow cytometry studies have demon-
secondary to acute contact activation of the blood with the strated that the majority of these diaphragm cells are my-
extensive foreign surfaces of the cardiopulmonary bypass eloid/monocytic in origin. In addition, a smaller percentage
and with the LVAD itself, as well as perioperative surgical of the cells are pluripotential hematopoietic cells which can
response. Later, a second, sustained phase of activation of be induced to differentiate in culture to mature hematopoi-
coagulation/fibrinolysis is created in these LVAD patients, etic cells.22 Menconi and colleagues21 found that the surface
which is caused by the progressive cellular population of the of explanted LVADs contained adherent mononuclear cells,
LVAD surface with cellular entrapment, activation and en- platelets, and myofibroblasts intermingled with areas of com-
hanced procoagulant activity. Furthermore, although we pact fibrinous material, as well as areas of collagenous tis-
found no evidence of increased IL-1 ! or TNF-# in the pe- sue. Similarly, Salih and colleagues18 found the surface of
ripheral plasma of LVAD patients over the time course of the explanted LVAD polyurethane membrane to consist of
LVAD implantation, sustained elevations in levels of tissue fibrinous and cellular layers. The immediate layer atop the
factor antigen were present in LVAD patients compared to membrane was composed of a compact fibrin coating as well
normal controls (Fig 36.5A). This, together with the finding as numerous mononuclear cells and spindle-shaped cells.
of increased tissue factor activity as reflected by enhanced This was followed by an intermediate middle layer consisting
390 Tissue Engineering of Prosthetic Vascular Grafts
of cells resembling fibroblasts and fewer mononuclear cells of a stimulus releasing increased numbers of dendritic cells
than in the immediate inner layer, and the outermost layer, into the blood of patients with LVAD and/or the presence of
at the biomaterial/tissue interface, contained a foreign body highly efficient mechanisms capable of trapping them on
type reaction with numerous multinucleated giant cells. Sec- the LVAD surface. In the first context, the presence of sig-
tions of tissue islands removed from the titanium surface nificant hemolysis in LVAD patients (as defined by elevated
revealed organized fibrous and collagenous tissue with few plasma free hemoglobin and absent haptoglobin) suggests a
cellular areas, except for occasional mononuclear cells. While possible mechanism underlying bone marrow stress. The
Salih and colleagues found no evidence of endothelial cells presence of activated macrophages on the LVAD surface pro-
on the surface of the LVAD, other studies have disputed this. vides an environment potentially enhancing recruitment of
Specifically, Frazier and colleagues19 found evidence for the CD34 positive and other cells, such as lymphocytes. In this
presence of endothelial cells on the pseudointimal lining of context, the work of Browning, Gallo and colleagues24 con-
the textured LVAD surface. Using antibodies to von cerning Kaposi’s sarcoma-like spindle cells (derived from the
Willebrand factor, they demonstrated positive immunore- peripheral blood of HIV-1-infected individuals) is especially
activity in certain cells on the lining, suggestive of the pres- of interest. They found that such cells expressed markers
ence of endothelial cells. (Of course, platelet material may found on both activated cells of monocyte/macrophage and
also be responsible for this positive immunostaining for von endothelial lineage, similar to what we have observed in cells
Willebrand factor). The significance of defining the pres- populating the LVAD. The authors speculated that a likely
ence of endothelial cells in this setting cannot be overstated, origin of Kaposi’s spindle cells would be a circulating bone
since their presence may suggest the potential for restora- marrow progenitor cell. The finding that culturing of these
tion of normal control of the vascular pro- and anticoagu- spindle cells required the presence of conditioned medium
lant pathways. from activated lymphocytes suggested the requirement for
In an attempt to more clearly delineate the cellular an additional stimulus to promote differentiation and pro-
content of the LVAD surface, we began to try to more de- liferation of Kaposi’s cells. By analogy with the situation in
finitively define the phenotypes of cells which are adsorbed LVAD, we propose that the initial recruitment of immune/
to the LVAD surface. inflammatory cells to the LVAD surface establishes a local
Our demonstration of CD34 positive cells on the milieu facilitating recruitment of CD34 positive cells from
LVAD surface (Fig. 36.6) confirms the results of Rafii and the blood. This further promotes the establishment of the
colleagues.22 Such cells, likely to be derived from the bone LVAD as an organ with impact on diverse host response
marrow, are progenitor cells which under normal conditions mechanisms.
differentiate into red blood cells, myeloid cells and platelets. Until future studies identify whether there are elevated
Under homeostatic conditions, their numbers in the periph- levels of CD34 positive cells in the periphery and whether
ery are quite low (0.01-0.1%).23 This suggests the presence there are specific stimuli recruiting them to the LVAD in
these patients, an alternate explanation must be considered.
Sustained Activation of a Procoagulant and Proinflammatory Systemic Response 391
Previous studies have suggested that bone marrow trans- left panel). Suggestive of enhanced macrophage procoagulant
planted onto vascular prostheses (composed of activity of these cells was the presence of increased
polytetrafluoroethylene, PTFE), results in complete immunostaining for tissue factor (Fig. 36.8). And, the cells
endothelialization by three weeks. Such graft areas popu- which were phenotypically similar to the population of CD34
lated with a confluent layer of cells were completely patent positive cells appeared to bear markers suggestive of endo-
and without evidence of thrombotic occlusion. In contrast, thelium, such as thrombomodulin (Fig. 36.9, left panel) and
control grafts (without bone marrow transplantation) were von Willebrand factor (Fig. 36.9, right panel).
covered by thrombi, as expected. Despite the fact that these Since CD34 positive cells are known to express adhe-
authors did not immunotype the cells transplanted onto the sion molecules, we postulated that such cells on the LVAD
prostheses, it is likely that the complex mixture contained surface might express vascular cell adhesion molecule-1
multiple hematopoietic precursor CD34 positive-like cells. (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1),
While it is possible that the LVAD surface-derived cells have thereby favoring the recruitment and adherence of further
the capacity to form a confluent monolayer, potentially ex- mononuclear/inflammatory cells. Immunocytochemistry
plaining why frank thromboses are not often observed, there revealed immunoreactivity for both of these adhesion mol-
is a critical distinction between the experimental previous ecules (Fig. 36.10, right and left panels, respectively). In the
results on PTFE and our data on the LVAD surface. Over- context of the LVAD surface, this is likely to be significant,
whelming evidence of angiogenesis was evident on the vas- since the presence of adhesion molecules provides a mecha-
cular PTFE prostheses, manifested by new capillary forma- nism for triggering the further recruitment of inflamma-
tion.25 In contrast, our study of multiple explanted LVAD tory/immune effector cells, thus propagating cellular acti-
surfaces has not shown angiogenesis, evidence of crude cap- vation on the LVAD. In contrast to the presence of cells bear-
illary formation or continuous cell monolayers covering the ing dendritic and monocytic markers, there was no staining
LVAD surface. Rather, we consistently observe that there are with antibody to smooth muscle actin or nonimmune IgG
patches of discrete cells (data not shown). Therefore, at this (Fig. 36.11 right and left panels, respectively).
time, we conclude that it is less likely that the cells on this Finally, the presence of proinflammatory leukocytes
surface are directed toward the formation of discrete vascu- was confirmed in the abundance of T lymphocytes which
lar, confluent, nonthrombogenic structures or monolayers. expressed strong immunoreactivity for CD3/CD4 and CD25
Further immunophenotypic characterization of cel- (Fig. 36.12), consistent with activated helper T cells.
lular populations on the LVAD surface suggested that inter- There was also a distinct B cell population immunore-
actions among different lineages of cells was likely occur- active for CD20 (Fig. 36.13).
ring. Quiescent round monocyte lineage cells expressing The presence of dendritic type CD34 positive cells with
CD14 were present (Fig. 36.7, right panel). In addition, nu- a procoagulant phenotype, as well as activated macrophages
merous larger cells with prominent cytoplasmic processes surrounded by proinflammatory leukocytes, certainly set the
and multinucleated morphology were present which express stage to explain the proinflammatory/procoagulant pheno-
the macrophage marker CD68, not CD14, and are therefore type created as a result of device implantation.
most likely activated macrophage lineage cells. (Fig. 36.7,
found to be absent. In contrast, prominent expression of vated macrophages on the LVAD surface activate CD4 posi-
IL-10 mRNA was detected. Importantly, the production of tive T cells by presenting antigen in the context of MHC
IL-10 is known to have a stimulatory effect on B cells and class II molecules together with IL-1 production. The sub-
has been reported to be increased in SLE and implicated in sequent activation of a Th2 subset of T cells leads to pro-
the pathogenic mechanism of autoantibody production in duction of IL-10 and CD40-CD40L interactions, which act
this disease. Evidence for T cell / B cell interactions involved on B cells to induce antibody production (Fig. 36.2).
in B cell hyperreactivity was shown by the presence of mRNA
for CD40 ligand. (CD40 on B cells and CD40 ligand on T LVAD as Inflammatory/Immune Organ
cells are the principal molecules involved in T cell contact- Taken together, these data suggest that considerable
mediated B cell stimulation.)(Fig. 36.16) Together these find- interactions between the LVAD surface cells and the host
ings led us to our supposition, as described earlier, that acti- are occurring throughout the time course of LVAD
396 Tissue Engineering of Prosthetic Vascular Grafts
implantation. We postulate that the mononuclear cells, plate- a cell-associated and released form, suggests a direct mecha-
lets and pluripotential stem cells initially trapped by the poly- nism for activation of coagulation. This is consistent with
urethane surface, not the titanium housing or Dacron grafts, the presence of tissue factor antigen in the plasma of pa-
become activated, thus leading to the generation of a local tients with LVADs,16 and suggests a mechanism for dissemi-
proinflammatory/procoagulant state, and that this mecha- nating the locally intense inflammatory and procoagulant
nism is, at least in part, responsible for triggering and sub- stimuli in the LVAD milieu. Furthermore, when monocytes
sequently sustaining the systemic activation of the coagula- were placed onto the LVAD surface in tissue culture, their
tion and fibrinolytic cascades. Furthermore, inflammatory capacity to respond to exogenously added LPS was enhanced
cells present on the LVAD surface appear to demonstrate (unpublished observation). This supports the concept that
production of proinflammatory cytokines and expression the LVAD primes adherent cells for heightened responsive-
of adherence molecules which promote cell activation, as ness to superimposed inflammatory stimuli.
well as facilitate recruitment of other cell types from the cir- We believe, therefore, that the biology of cells in the
culating blood, thereby sustaining the immune alterations LVAD resembles a two-hit model in which the initial place-
(Fig. 36.17). Our observation that monocyte-derived mac- ment of the device within the circulation results in recruit-
rophages seeded on the LVAD surface generate tissue factor, ment of adherent cells, initially likely to be CD34 positive
pluripotent hematopoietic cells and monocytes, which then NF-∀B is involved in the transcriptional activation of in-
undergo subsequent differentiation and activation with the flammatory cytokines such as IL-1, IL-2, IL-6 and IL-8 in
capacity to recruit other cells, such as dendritic-type cells response to LPS or TNF, thereby facilitating the further
and lymphocytes, consequently sustaining and expanding propagation of the inflammatory response. Electrophoretic
the local host response. From such a view, the LVAD emerges mobility shift assays using nuclear extracts prepared from
as an immune/inflammatory organ which redirects the host cells removed from the LVAD surface26-27 as well as a
response, modulating multiple effector systems such as co- [32P]-radiolabeled consensus probe for NF-∀B demonstrated
agulation and immune mechanisms. a marked gel band shift, confirming NF-∀B activation in this
setting (Fig. 36.18). This finding, in addition to lending
further support to the concept that the cells colonizing the
NF-∀∀B Is a Marker of Cellular Activation in the surface of the LVAD are activated with respect to the
LVAD Surface Milieu and a Target for Anti- generation of a proinflammatory/procoagulant environ-
Inflammatory Intervention ment, also provided a potential target for therapeutic anti-
An important goal of our studies in attempting to inflammatory intervention in LVAD patients. In this context,
understand the intricate interactions between the populated a number of studies have suggested that NF-∀B’s role as a
LVAD surface and the host was to determine if therapeutic central regulator of the inflammatory response can be
strategies might be designed to limit the generation of downregulated by anti-inflammatory agents such as high
proinflammatory and prothrombotic mediators that appear dose salicylates (ASA).28-33 The specific cellular mechanism
to be central to the activation of coagulation/immune path- of ASA seems to be dependent on its ability to act as an an-
ways observed in the setting of the textured surface LVAD. tioxidant and free radical scavenger, thereby preventing the
We therefore sought evidence that the generation of inducible decay of I-∀B and therefore the subsequent
cytokines and tissue factor reflected ongoing cellular stimu- activation of NF-∀B. This effect has been shown to be
lation and activation of cell signaling pathways in the cells independent of the cycloxygenase pathway, as agents such
populating the diaphragm. A potential culprit in this set- as indomethacin are ineffective in blocking
ting is the nuclear transcription factor NF-∀B, an inducible I-∀B-NF-∀B signaling.
transcription factor of the Rel family which is known to have To test this hypothesis, we began a prospective evalu-
a central role in the inflammatory response. Specifically, in ation of ASA (325 mg PO/PR QD) started in LVADs within
endothelial cells, NF-∀B is required for the transcriptional three days of implantation. Markers of thrombin genera-
activation of VCAM-1, ICAM-1 and E-selectin in response tion (TAT and F1+2) and fibrinolysis (D-dimers) were sig-
to TNF-#, thus contributing to the expression of cell adhe- nificantly reduced compared to untreated LVADs (n = 20),
sion molecules involved in the recruitment of leukocytes to as were peripheral TF antigen and macrophage procoagulant
an inflammatory nidus. And, in proinflammatory leukocytes activity (p < 0.05). Inflammatory cell activation was also
abated, as none of the treated LVADs developed elevated all consistent with a primary decrease in inflammation at
PRAs up to 90 days post implantation. Analysis of cells on the LVAD surface as well as diminished ability to further
the LVAD surface at explant (n = 3) compared to untreated recruit inflammatory cells. Taken together, these data sug-
LVADs (n = 6) confirmed marked downregulation of the gest that targeted anti-inflammatory intervention with ASA
inflammatory response: may be an effective means to diminish cellular adsorption/
1.Markers of macrophage activation (CD68) were distinctly activation by the textured surface LVAD, thereby attenuat-
absent; ing the associated inflammatory response and improving
2.Cells did not express procoagulant TF or vascular cell ad- long term tolerance and eventual clinical outcome
hesion molecule-1; and (Fig. 36.19).
3.There was a marked absence of B cells (CD20) and T cells
(CD3/4/25)—
12. Slater JP, Rose EA, Levin HR et al. Low thromboembolic face of left ventricular devices. Ann Thorac Surg 1995;
risk without anticoagulation using advanced design left 60:1627-1632.
ventricular assist devices. Ann Thorac Surg 1996; 23. Stella CC, Cazzola M, De Fabritiis P et al. CD34-positive
62:1321-8. cells: Biology and clinical relevance. Haematologica 1995;
13. Lavee J, Kormos RL, Duquesnoy RJ et al. Influence of 80:367-387.
panel-reactive antibody and lymphocytoxic crossmatch on 24. Browning PJ, Sechler JMG, Kaplan M et al. Identification
survival after heart transplantation. J. Heart Lung Trans- and culture of Kaposi’s sarcoma-like spindle cells from
plant 1991; 10:921-930. the peripheral blood of human immunodeficiency virus-
14. Smith JD, Danskine AJ, Laylor RM et al. The effect of 1 infected individuals and normal controls. Blood 1994;
panel reactive antibodies and the donor specific 84:2711-2720.
crossmatch on graft survival after heart and heart-lung 25. Noishiki Y, Tomizawa Y, Yamane Y et al. Autocrine an-
transplantation. Transplant Immunol 1993; 1:60-65. giogenic vascular prosthesis with bone marrow transplan-
15. Massad MG, Cook DJ, Vargo R et al. Factors influencing tation. Nat Med 1996; 2:90-92.
HLA-sensitization in implantable LVAD recipients. Intl 26. Leonardo MJ, Fen CM, Baltimore D. The involvement of
Soc Heart & Lung Transplantation 1996; Abstract #109. Nf-∀b in interferon gene regulation reveals its role as a
16. Spanier TB, Oz MC, Levin HR et al. Activation of coagu- widely-inducible mediator of signal transduction. Cell
lation and fibrinolytic pathways in patients with left ven- 1989; 57:287-294.
tricular assist devices. J Thorac Cardiovasc Surg 1996; 27. Baeuerle PA, Henkel T. Function and activation of NF-∀B
112:1090-7. in the immune system. Ann Rev Immunol 1994;
17. Bibiokis EJ, Livingston AS, Pethak CD et al. The bio- 12:141-179.
chemical basis of bleeding during left ventricular assis- 28. Nolan G, Ghosh S, Liou HC et al. DNA binding and IkB
tance. Intl Soc for Heart & Lung Transplantation. 1996; inhibition of the cloned p65 subunit of Nf-∀b: A rel-re-
Abstract #110. lated polypeptide. Cell 1991; 64:961-969.
18. Salih V, Graham TR, Berry CL et al. The lining of tex- 29. Manning AM, Anderson DC. Transcription factor Nf-∀b:
tured surfaces in implantable left ventricular assist devices: An emerging regulator of inflammation. Annu Rep Med
An immunocytochemical and electronmicroscopic study. Chem 1994; 29:241.
Am J Cardiovasc Pathol 1993; 4:317-325. 30. Weber C, Wolfgang E, Pietsch A et al. Aspirin inhibits
19. Frazier OH, Baldwin RT, Eskin SG et al. Immunochemi- Nf-∀b mobilization and monocyte adhesion on stimulated
cal identification of human endothelial cells on the lining human endothelial cells. Circulation 1995; 91:1914-1917.
of a ventricular assist device. Texas Heart Inst J 1993; 31. Chen CC, Rosenbloom CL, Anderson DC et al.Selective
20:78-82. inhibition of E-selectin, vascular cell adhesion molecule-
20. Menconi MJ, Owen T, Dasse KA et al. Molecular ap- 1 and intercellular adhesion molecule—1 expression by
proaches to the characterization of cell and blood/bioma- inhibitors of IkB-# phosphorylation. J Immunol 1995;
terial interactions. J Cardiac Surg 1992; 7(2):177-187. 155:3538-3545.
21. Menconi MJ, Prockwinse S, Owen TA et al. Properties of 32. Koop E, Ghosh S. Inhibition of Nf-∀b by sodium salicy-
blood-contacting surfaces of clinically implanted cardiac late and aspirin. Science 1994; 265:956-958.
assist devices: Gene expression, matrix composition, and 33. Pierce JW, Read MA, Ding H et al. Salicylates inhibit IkB-
ultrastructural characterization of cellular linings. J Cell a phosphorylation, endothelial leucocyte adhesion mol-
Biochem 1995; 57:557-573. ecule expression, and neurtrophil transmigration. J
22. Rafii S, Oz MC, Seldomridge JA et al. Characterization of Immun 1996; 156:3961-3969.
human hematopoietic cells arising on the textured sur-
Facilitation of Healing
Transdifferentiation
CHAPTER 37
Transdifferentiation and the Vascular Wall
William A. Beresford
T he solidity of the idea of a continent hindered accepting that continents move and change.
Likewise, the long-accumulated robust bases for defining tissue and cell identities obliged
one to doubt that the occasional clearcut switches in character by mature, differentiated
cells—transdifferentiations1-3—were typical of cells in general and had far-reaching signifi-
cance. But today, such ‘plasticity’, although a vogue word, demands the attention of surgeons
and bioengineers if they are to understand their live working materials. For instance, has
what one grows in culture for grafting the phenotype one wants? Will this introduced tissue
be stable, and how will it interact with other tissues to maintain or alter their characters?
Will reactions to foreign materials or viral vectors for gene transfer4 encourage cells to take
on undesirable phenotypes? Can new and beneficial phenotypes be created in vitro or in
animals, for example, cells resistant to rejection or lacking dangerous cytokines? The de
novo generation of cardiac myocytes by transdifferentiation is a recent proposal.5 Certain
smooth muscle cells and fibroblasts can become epithelial and glandular. Hence, can one
graft to a vascular site cells transfected for long term prolific liberation of a therapeutic
substance, but which still have a sufficiently ‘vascular’ nature to be properly integrated? Or
which might also be regulated or transfected to express adhesion molecules to hold the cells
in place? Answers lie in seeing how each cell type of the many vascular walls can change its
phenotype (Figs. 37.1 and 37.2), uncovering the stimuli and their sources, and tracking the
regulatory pathways and interactions.
Endothelial Cells
Endothelial cells (ECs) have multiple personalities, recognized as their many respon-
sibilities at any particular site,6 and their different properties and responses at various loca-
tions7,8 and in the regions of a vascular bed.9 Examples are urea transport in the renal me-
dulla,10 and directing T lymphocyte homing to superficial rather than deep vessels of the
dermis.11 ECs’ biochemical profiles must reflect this diversity, but the transdifferentiations
detected thus far are mainly morphological.
To further aid lymphocyte homing and emigration, ECs in venules of nodes, tonsils,
and mucosal lymphoid organs become cuboidal, quite rich in organelles, and express
discernible antigens—high endothelial venules (HEVs).12 Coagulating and severing the
afferent lymphatic vessels to rat popliteal nodes causes the high endothelium to flatten and
lose its secretory ultrastructure by three weeks. Injecting sheep erythrocyte antigen into
such a node rapidly restores some of the high ECs and the lymphocyte traffic; thus the
conversion is reversible.13
HEVs develop from normal endothelium in chronically inflamed synovium, both
human and experimental animal.14,15 Other experiments demonstrate the role of an
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
404 Tissue Engineering of Prosthetic Vascular Grafts
immunoreactive environment and the generality of the microfilaments with focal densities as to lead some authors
transdifferentiation. An immune mediator—gamma-inter- to term them ‘myoendothelial’.23
feron—induces expression of an HEV antigen in cultured The contrary phenomenon—an induction of fenes-
nonlymphoid ECs from mouse lung and marrow.16 Inter- trae in continuous ECs—also occurs in vivo:
feron-!, IL-2, and mitogens for ECs did not result in HEV- 1. During regrowth of capillaries in wounded guinea-
marker expression. The HEV marker and plumper ECs ap- pig cremaster muscle;24
peared in dermal venules where sheep red blood cells in 2. In vessels growing into the rat retinal pigment epi-
Freund’s adjuvant had been injected. Subcutaneous implants thelium after urethane-induced loss of the photore-
of sponges loaded with splenic lymphocytes acquire vessels ceptors;25 and
which display a high endothelium, if the lymphocytes are 3. In human dermal papillary vessels affected by pso-
allogeneic.17 riasis.26
The second and also reversible EC transdifferentiation Experimental instances are in:
is between fenestrated and continuous endothelium. The 1. The new capillaries of hypertrophied guinea pig gut
change may come about rapidly, within 10 minutes,18 on a muscle;27
physiological time scale (Fig. 37.3), or be slow and endur- 2. Rabbit lingual microvessels a few hours after destruc-
ing. Cirrhosis of the liver in man19 and in rats injected with tion of the animal’s platelets by anti-serum;28 and
CCl4 20 is accompanied by the fenestrated sinusoids acquir- 3. Rat cremasteric and dermal capillaries treated locally
ing a basement membrane and a continuous endothelium: with vascular endothelial growth factor (VEGF).29
a capillarization. If the cirrhotic liver progresses to hepato- A month of continuous infusion of retinoic acid or
cellular carcinoma, the tumor cells appear to cause the de- phorbol acetate into the rat cortex30 results in fenestrated
velopment of a basement membrane and endothelial CD34 vessels in cerebral cysts. After cessation of the infusion and
expression in the sinusoids of the tumor.21,22 ECs of these removal of the cannula, the vessels in the cystic cavity slowly
capillary-like tumor vessels are peculiar in having so many become continuous, despite the absence of astrocytes. There
Transdifferentiation and the Vascular Wall 405
is other evidence in vivo31 that coculture work may have A differentiative role for basement membrane and
been misleading as to the importance of astrocytes in regu- neighboring tissue39 is clear in the conversion of embryonic
lating how brain ECs create the blood-brain barrier. How- cardiac endothelium of the atrioventricular canal to the mes-
ever, factors in the neural environment are significant, since enchymal cells that will form the cardiac cushion, from which
local chick vessels growing into intracoelomically grafted the AV valves and septa develop (Fig. 37.4).40 This trans-
embryonic quail brain acquire morphological and his- differentiation, in keeping with certain others of embryonic,
tochemical features of brain capillaries, with their special but partly differentiated, cells is termed an ‘epithelial-mes-
variety of continuous endothelium.32 enchymal transition/transformation’. It appears to require
For an analysis of factors controlling trans- specific matrix glycoproteins41,42 and, in the chick, TGF-!3,43
differentiations, we note that ECs are a form of epithelium, but in mouse the TGF isoform responsible and whether it
and epithelial-mesenchymal interactions are well investi- comes from the myocardium are uncertain.44 Preliminary
gated, and slowly making sense, in a variety of organs. Simi- evidence suggests that, in the earliest development of the
lar work in vitro on ECs indicates that fenestrated adrenal avian dorsal aorta, the first SMCs may be derived from the
capillaries lose their pores unless cultured on basement endothelium in chick45 and quail,46 from the sharing of en-
membrane, newly formed by MDCK cells,33 or reconstituted dothelial and smooth muscle markers. The endothelial-mes-
as Matrigel. However, the same MDCK-derived matrix was enchymal conversion may not be restricted to embryonic
unable to switch continuous capillary ECs to fenestrated.34 times. Human foreskin microvessel ECs grown in cyclic
Two specific differentiative agents, having opposite effects AMP-deficient medium lose their epithelial and EC charac-
on fenestrae of adrenal ECs, were retinoic acid, increasing teristics and become mesenchyme-like,47 as do dermal ECs
their number, and TGF-!, causing a marked reduction.35 exposed to interleukin-1!.48 That this was more than an in
A different and more comprehensive approach in vivo vitro dedifferentiation needs evidence that the resulting cells
is to examine markers of continuous endothelium and of have true mesenchymal potential, but there are other in-
sinusoids, along with changes in the molecular composition stances of possible EC to SMC conversions.49
of the adjacent matrix, as human liver sinusoids develop.36 The localized cardiac loss of endothelial character early
After an early phase of matrix and cellular structural change in development raises the issue of homeobox phenotype
(and the loss of capillary markers), the sinusoidal EC markers determining genes and their products. Receptors for the vas-
(CD4, ICAM-1, CD32, and CD14) appeared. Unexpectedly, cular endothelial growth factor (VEGF) family are the ty-
tenascin, although present, seemed not to regulate EC rosine kinases Flk-1 and Flt-1. Two related receptors, also
development, but laminin probably does.37 The influence almost restricted to endothelial cells, are Tie and Tek, but
of adjacent tissue stands out in the rabbit’s nose, where the their ligand is unknown. Working upstream of the genes for
endothelium of the capillaries and venous sinuses is these receptors is the product of a gene altered in the cloche
fenestrated only on the side of the vessel facing the mutation in zebrafish.50 This mutation causes the develop-
nasal epithelium.38 ing heart to lack endothelial cells. Here one can ask a question
406 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 37.4. Diagrammatic summary of endocardial cytodifferentiation. In (A) the endocardium of an 8 somite embryo is shown as
consisting of uniformly similar cell types in all areas of the simple tubular heart. In (B) the two pathways of endocardial develop-
ment are shown as they first occur in 16-18 somite embryos: In the atrium and ventricle, the endocardium becomes attenuated
with reduced secretory potential and acquires an “endothelial look”. Trabeculation brings the myocardium in close proximity to
the endocardium (B) and (C) and appears to be temporally related to “endothelization” of atrial and ventricular endocardium.
Conversely, in sites where prospective valvular and septal primordia (cushion tissue) will form, the endocardium hypertrophies
and acquires and amplifies secretory potential. The initial formation of cushion tissue cells (C) beneath the endocardium, and
their morphologic similarity to the latter, suggest that valvular and septal primordia are endocardial derivatives. Reprinted from
Markwald G et al. Devel Biol 1975; 42:160-180, with kind permission from Academic Press Inc., Orlando, Florida.
basic to any transdifferentiation, of whether in the endothe- can develop in vascular walls.61 Phagocytic cells removing
lial to mesenchymal switch the cell type specific cloche, Flk-1, surplus collagen from the tadpole’s degenerating tail are
flk-2, tie and tek genes stop expression. If so, one asks, ‘When?’ adapted local fibroblasts.62
since there is a sequence to gene activation during embry- When the bladder outlet is partially obstructed,
onic endothelial differentiation. Secondly, are the Hox and submesothelial fibroblasts of the rabbit’s urinary bladder
other genes needed to generate a cell type in normal devel- proliferate and thicken the serosa. The proliferating cells
opment (better understood in blood cell lineages51,52) again progress from expressing keratin-18 to become myo-
required when that type derives later from a fibroblasts, and finally resemble fetal smooth muscle cells,63
transdifferentiation? If so, this would offer an opportunity based on their immunoreactive cytoskeletal profiles. The
for experimental influences on mature cells. authors suggest that the keratin positivity indicates that some
of the proliferating fibroblasts become mesothelial cells to
Fibroblasts replace lost mesothelium. A dual potency of submesothelial
The tantalizing variety of site-related fibroblastic phe- fibroblasts is further indicated by the mixture of smooth
notypes53 extends to several kinds of transdifferentiation— muscle cells (SMCs) and decidua-like cells in the guinea pig
glandular, skeletal, and muscular, for example. Stromal fi- peritoneum, resulting from giving progesterone after three
broblasts of the rodent endometrium become glandular months of estrogen priming.64
epithelial decidual cells in normal physiology, and in re- In vitro studies mirror most of the above
sponse to various experimental provocations.54 Stromal cells transdifferentiations in vivo, and add more. Notable is the
of the human uterus, tube, and ovary behave similarly. Re- conversion of the fibroblast-like 3T3 cell line to adipocytes.65
nal medullary interstitial fibroblasts are laden with lipid Blocking mitosis with relaxin does not prevent the
droplets and may be endocrine,55 but in anemia the cortical adipogenic conversion, 66 giving further evidence that
fibroblasts also acquire significant lipid56 and are recruited transdifferentiation can be direct,67 i.e., not dependent on
to produce erythropoietin (Fig. 37.5).57,58 Fibroblasts from cell division. A mesenchymal to epithelial switch is demon-
somatic rather more than visceral sources become strated by 3T3 cells transfected with hepatocyte growth fac-
chondroblasts and/or osteoblasts, depending on the stimu- tor (HGF/SF) and the human Met receptor.68 The cells be-
lus and the animal species,59,60 but ectopic cartilage and bone come epithelial-like in vitro and tumorigenic in nude mice.
Transdifferentiation and the Vascular Wall 407
Taxol treatment of mouse embryonic fibroblasts makes them 4. Myofibroblasts occur as subtypes based on how far
adopt an epithelial morphology.69 they share other cytoskeletal components, such as
Hypoxic pulmonary vasoconstriction and hyperten- desmin with smooth muscle,79 and there are species
sion result from keeping newborn calves at simulated high differences;80
altitude. Among the structural remodelings is the switch of 5. Some myofibroblasts may be caught part way though
adventitial fibroblasts of the pulmonary arteries to making a fibroblast to smooth muscle conversion;81 many
elastin.70 Medium from culturing SMCs from hypoxic ar- others seem to be a stable endpoint of a
teries brings about an elastogenic response in cultured ad- transdifferentiation, although still able to revert to
ventitial fibroblasts from normal pulmonary arteries. In gen- fibroblasts;82
eral, the role of adventitial fibroblasts in vessel-wall remod- 6. Study of the production of extracellular matrix by
eling may have been underestimated.71 myofibroblasts has focused on collagen until re-
The ‘myofibroblast’72—the cell combining features of cently;83
fibroblast and SMC—is a practical and theoretical challenge. 7. The organ diversity of fibroblastic forms brings va-
Uncovering and understanding transdifferentiations of fi- riety to the starting cell: How fibroblastic are hepatic
broblasts to or towards muscle cells stumble on six issues: stellate cells,84 renal mesangial cells,85 and renal in-
1. How does one know that the starting cell was a fi- terstitial fibroblasts?86
broblast, since many organs normally contain The bulky literature on myofibroblasts concentrates
myofibroblasts, including aortic valves?73 on visceral fibrosis and dermal wound healing, but some of
2. Somatic connective tissues by and large lack the experiments are exciting, with new stimuli acting on the
myofibroblasts, unless disturbed or diseased,74,75 and usual cells, and old stimuli provoking newly examined cells.
help with this problem; The general phenotype-altering agent 5-azacytidine confers
3. How easily are SMCs, fibroblasts, and myofibroblasts an SMC-like phenotype on mouse mesangial cells.87 Expos-
to tell apart? They are distinguished by certain de- ing mouse dermal fibroblasts continuously to culture me-
finitive ultrastructural features, such as microfila- dium from growing C2C12 skeletal myoblasts causes the fi-
ment bundles and dense bodies which got the broblasts to express a skeletal muscle-specific transcription
myofibroblast idea started, but mostly by quantita- factor—MyoD—and desmin.88 And the differentiation-per-
tively different profiles76 rather than by specific turbing factor TGF-!1 similarly has cardiac fibroblasts in
markers, although #-smooth muscle actin has been culture becoming like cardiac myocytes.89 Intriguingly, stri-
the main standby, signifying a shift towards muscle. ated muscle, smooth muscle, and fibroblasts are already
(Specific cell type markers now exist only as linked in that all three, and endothelium, synthesize a spe-
smoothelin for smooth muscle77 and certain mAbs cial collagen—type XV.90
for fibroblast surface epitopes.78)
408 Tissue Engineering of Prosthetic Vascular Grafts
The cells involved in colonizing and incorporating a cytokines in myofibroblast activity.84 Lastly, as rat cardiac
Dacron graft to the canine aorta imply a similar connec- fibroblasts convert to myofibroblasts in response to injury,
tion. The graft becomes lined with collagen-synthesizing they express angiotensin-converting enzyme (ACE) on their
myofibroblasts, with a gradation of phenotype from fibro- surface. The resulting angiotensin ii may stimulate the re-
blasts in the pseudointima. Filaments in the early central lease of TGF-!l by autocrine and paracrine modes from the
endothelioid cells indicated that some of the endothelial cells myofibrolasts themselves and macrophages, respectively
might derive from the adjacent myofibroblasts,91 a counter- (Fig. 37.6).95
point to embryonic aortic endothelial cells being a source
of VSMCs. However, fibroblasts and endothelial cells are not Smooth Muscle Cells
directly tied to muscular phenotypes in that it is only the The diversity of smooth muscle phenotypes expresses
three muscle varieties that share use of the Gax different embryological germ layer origins, locations,
homeodomain protein during mouse embryogenesis.92 changed physiological demands, pathological effects, and
Lastly, angioplastic balloon injury elicits proliferation and a species. Ectodermal neural crest gives rise to smooth muscle
myofibroblastic reaction from adventitial fibroblasts in por- of great arteries, but, if ablated, is replaced inadequately by
cine coronary arteries.93 mesodermal SMCs.96 Lineage maintains its influence on the
Analyzing how cytokines influence the phenotypic thoracic versus abdominal aortic SMCs in their adult be-
conversions is of general interest. Four experiments point havior. Vascular smooth muscle cells (VSMCs) are distinct
the way. Fibrosis is an end result requiring adequate num- from gastrointestinal, uterine and tracheal smooth muscles,
bers of myofibroblasts, so which cytokines promote prolif- and experience most of the known transdifferentiations. In
eration? And which cause transdifferentiation? When the kidney, the afferent arterioles become glandular and se-
TGF-!1 induces quiescent normal human breast fibroblasts crete renin in response to a variety of stimuli,67 including
to express #-smooth muscle actin in culture without divid- an inhibitor of angiotensin I converting enzyme.97 SMCs of
ing, this shows the differentiative role of TGF-!, and that some muscular arteries become fibroblastic in situ, produc-
the phenotype does not arise by selection of a subpopula- ing excessive collagen and other ECM materials in medial
tion.74 This is, incidentally, another example of a direct fibromuscular dysplasia,98,99 a condition reproduced in dogs
transdifferentiation. Overexpression of GM-CSF in the rat after several months by blocking the vasa vasorum with a
lung in vivo expands and activates the macrophage popula- mixture of thrombin and gelatin.100 A more physiological
tion, which then provides the TGF-! causing the myo- phenomenon is the remodeling and increase in medial col-
fibroblasts to develop and make collagen.94 Detection of lev- lagen in mesenteric arteries of spontaneously hypertensive
els of, and relations between, cytosolic signal-transduction rats,101 although the changes precede the increase in blood
molecules, such as Ras and MAPK, in cells responding to pressure,102 so that what prompts the response is unclear.
TGF- ! can rule out the involvement of certain other
Knowing the ontogeny of the cells, migratory ties, qualifies as a transdifferentiation. An eclectic lumping
possibilities, and the actual state of the tissue believed to of all such phenomena is justified, since so little is known of
undergo conversion is essential for accurate interpretation. what aspects of phenotype are significant, and any rules of
For example, veins grafted in place of arteries are thought to regulation learned in a suspect transdifferentiation illumi-
undergo ‘arterialization’. But the smooth muscle of the nate differentiation mechanisms in general. The contractile
neointima may have migrated from host arterial wall,103 to synthetic shift is reflected in abundantly altered gene ex-
or come in as myofibroblasts from the remodeling venous pressions, with activation or reactivation for cytokeratins 8
perivascular tissue, 104 or even have been present and 18,121 tenascin,122 collagen type VIII,123 #3 integrin,124
before grafting. 105 nonmuscle myosin heavy chain,93 platelet-derived growth
Intense clinical interest focuses on atherosclerosis- factor A chain, 125 and the related transmembrane
mimicking fibroblastic behaviors of VSMCs, which include proteoglycans, syndecans 1 and 4,126 with changed balances
proliferation and migration—striking changes in the nor- of alternative RNA-splicing,127 a marked downregulation of
mally quiescent contractile cells. Circumstances evoking a ‘contractile’ genes, and upregulation for collagen,128 c-myc,129
contractile to synthetic arterial VSMC transition (Fig. 37.7) sulfated proteoglycans, 130 and matrix
include: culturing in serum; 106 luminal injury; 107 metalloproteinases.131,132 Caveolin is not reduced, but is re-
intraperitoneal injections of benzo[a]pyrene;108 uremia from distributed internally.133
subtotal nephrectomy;109 adjacent mesenteric fibrosis;110 Analysis of phenotypic regulation has penetrated to
giant-cell arteritis; 111 immune injury from allogeneic the coordination of gene transcriptions and the search for
transplantation;112 and, perhaps closer to etiological mecha- shared type-specific transcription factors.134 The control of
nisms, topical application of cytokines.113 The examples key cytokines occurs both by the sequestration by binding
show that VSMCs from both muscular and elastic arteries in the ECM135 and by localization to cell surface proteoglycan
execute the switch. modulators.136 Signals come via cell-matrix binding from
How much of a change is it for VSMCs to become the ECM made by the cells themselves137 and by adjacent
fibroblastically ‘synthetic’?106 First, the cells in development cells.124 The confirmation and stabilization of phenotype also
are substantial synthesizers of ECM, while acquiring con- derive from the linking of cells by cell-cell junctions, includ-
tractility. Secondly, normal human aortic VSMCs contain ing the innervation.138 For the earliest establishment of vas-
the synthetic organelles—granular endoplasmic reticulum cular smooth muscle, tissue factor (used later to help initiate
and a Golgi complex, in addition to the contractile machin- clotting) is needed, as shown by a gene inactivation experi-
ery,114 whereas those of the usual experimental animals are ment in mice,139 but homeotic gene expression patterns spe-
far fewer. In some species the media includes ‘nonmuscle’ cific for VSMC precursors at this stage are not yet known.
cells.115-117 Thirdly, VSMCs becoming synthetic in culture Other smooth muscles than vascular resemble VSMCs
can be returned to a contractile state.118-120 Some would in their heterogeneity140 and plasticity: Tracheal SMCs
question whether the acquisition of a phenotype by a rever- become ‘synthetic’ in culture;141 gut SMCs make more ECM
sion to embryonic behavior, or by boosting existing activi- material, if the ileum is partially obstructed in guinea-pigs;27
Fig. 37.7. (A) shows a contractile smooth muscle cell from the media of
the rat carotid artery, and (B) a synthetic smooth muscle cell from the
neointima two weeks after balloon injury. E, elastic fibers; ER, endoplas-
mic reticulum; F, myofilaments; M, mitochondria; N, nucleus. Magnifi-
cation x15,000; bar = 1 ∝m. Reprinted from Thyberg J et al. Cell Tissue
Res 1995; 281:421-433, with kind permission from Springer-Verlag
GmbH, Heidelberg, Germany.
410 Tissue Engineering of Prosthetic Vascular Grafts
implicitly, and often overtly, an intermediate zone—reflected provocation. To be useful a new phenotype must integrate
in the myofibroblast, epithelioid SMC, and such others. harmoniously over the longer term. The invasive growth
However, as the investigation probes phenotype more deeply, within the brain of fibroblasts expressing a transgene for
the extent of molecular change involved in, say, a contractile tyrosine hydrolase184 illustrates the danger of modifying only
SMC becoming a synthetic SMC may amount to a one aspect of phenotype. Fortunately, cell proliferation is
transdifferentiation. Differential gene and protein analysis not a prerequisite for a switch of phenotype, making it easier
and display will help define exactly how related cell to aim for a new cell type while taking steps to prevent run-
types differ.178 away growth in vivo.
Progress requires that the differentiated state and its Another route to sufficient cells of a desired type is to
conversions be understood in terms of cell structure, be- steer stem cells to differentiate while they proliferate, but
havior (including polarity, proliferation, migration, adhe- again the problem arises of matching in vitro cell type to
sion,179 and inclination to apoptosis180) the distinctive work- host organ locale. Can one achieve sufficient refinement in
ing molecules, the transcriptional and translational regula- this before grafting? Detection of the critical environmental
tors producing those molecules, the receptors and transduc- factors, along with coculture or transfer of medium, may be
tion mechanisms controlling the production and deploy- an avenue.
ment of the principal molecules, the external relations of The paucity of known controlling factors constitutes
the cell with signals, cells, and matrix, and what the cell ‘re- another obstacle—TGF-! and retinoic acid influence the
members’ of its lineage, i.e., its precursor states. Relative to differentiation of several different, but in vivo adjacent, cell
what should be known, transdifferentiations are testified to types. For selectivity, much more needs to be known of how
by a very few structural and chemical markers, expressing such factors are released, bind, are activated, work in se-
what available technique, curiosity, current concerns, and quence and combination, and affect the transduction sys-
often chance happened upon in regeneration and disease. tems. Beyond the signals are the transcriptional controls,
Routes to a more systematic study include such questions where the complexity multiplies almost by the month for
as: Can cells return along lineage pathways that they have any given cell type specific gene, including sometimes the
traversed? (Osteoblasts and mast cells may be more termi- discovery that the gene is expressed in cell types not con-
nal than fibroblasts and pericytes.) And, if reversion occurs, ventionally thought to be related, such as prostate-specific
what is the story of homeodomain genes and ‘master’ tran- antigen in the female breast. Nevertheless, cures can be ef-
scriptional factors, and of signaling mechanisms? Answers fective without one knowing how, and induced bone for
learned from the classic transdifferentiations in amphibia, skeletal repair60,185 shows that transdifferentiations186 can
worms, hydra, plants, and other mammalian systems2,3 be applied to healing before they are satisfactorily under-
remain pertinent. stood at cellular and molecular levels.
The question of what restrains adult cell phenotype
received new impetus from the cloning of a lamb from a References
ewe’s mammary epithelial cell nucleus.181 The lineage of the 1. Beresford WA. Chondroid Bone, Secondary Cartilage, and
mammary cells of the lamb is extraordinary, but highlights Metaplasia. Munich & Baltimore: Urban & Schwarzenberg,
what was already evident from experiments with hybrid 1981:1-454.
fused cells, transplanted differentiated embryonic cells, and 2. Okada TS, Kondoh H. Transdifferentiation and Instabil-
ity in Cell Commitment. Kyoto: Yamada Science Foun-
gene transfections,182 namely that mature cells remain able
dation, 1986:1-433.
to become vastly different cell types. This potential lies fal- 3. Reyer RW, Liou W, Pinkstaff CA. Ultrastructure and
low for the bioengineer’s cultivation, but within many con- glycoconjugate histochemistry of the lens capsule during
straints, some of which have emerged during the look at lens regeneration from the iris in the newt. Exp Eye Res
vascular transdifferentiations. 1994; 58:315-329.
If any cell can transdifferentiate, this clouds cell re- 4. Arnold TE, Gnatenko D, Bahou WE. In vivo gene trans-
sponsibility. For example, the matrix-depositing athero- fer into rat arterial walls with novel adeno-associated vi-
sclerotic intimal cell could derive from SMCs—the custom- rus vectors. J Vasc Surg 1997; 25:347-355.
ary view—or endothelial cells,46 or adventitial fibroblasts,93 5. Olson EN. Things are developing in cardiology. Circ Res
or from more than one source. Which choice is correct in- 1997; 80:604-606.
6. Shireman PK, Pearce WH. Endothelial cell function: Bio-
forms the steps to therapy and prevention. Lineage has to
logic and physiologic functions in health and disease. AJR
do not only with specific expressions, e.g., of major deter- 1996; 166:7-13.
minants of phenotype, such as NeuroD,183 but with their 7. Fajardo LF. The complexity of endothelial cells. Am J Clin
timing, to which controlled useful transdifferentiations will Pathol 1989; 92:241-250.
have to pay heed. 8. Garlanda C, Dejana E. Heterogeneity of endothelial cells.
There is an enduring consistency of cell behavior and Specific markers. Arterioscler Thromb Vasc Biol 1997;
matching phenotype, but experimental and pathological 17:1193-1202.
perturbation shows this calm harmony is precarious and the 9. Spanel-Borowski K. Diversity of ultrastructure in differ-
product of ceaseless communication between cells and their ent phenotypes of cultured microvessel endothelial cells
milieu. Transdifferentiations often happen because of dis- isolated from bovine corpus luteum. Cell Tissue Res 1991;
ruption. The inherent contradiction is in seeking what con- 266:37-49.
firms and stabilizes cell phenotype from studies of brief
412 Tissue Engineering of Prosthetic Vascular Grafts
10. Xu Y, Olives B, Bailly P et al. Endothelial cells of the kid- 30. Kaya M, Chang L, Truong A et al. Chemical induction of
ney vasa recta express the urea transporter HUT11. Kid- fenestrae in vessels of the blood-brain barrier. Exp Neurol
ney Int 1997; 51:138-146. 1996; 142:6-13.
11. Kunstfeld R, Lechleitner S, Groger M et al. HECA-452+ 31. Krum JM. Effect of astroglial degeneration on neonatal
T cells migrate through superficial vascular plexus but not blood-brain barrier marker expression. Exp Neurol 1996;
through deep vascular plexus endothelium. J Invest 142:29-35.
Dermatol 1997; 108:343-348. 32. Stewart PA, Wiley MJ. Developing nervous tissue induces
12. Kraal G, Duijvestijn AM, Hendriks HH. The endothelium formation of blood-brain barrier characteristics in invad-
of the high endothelial venule: A specialized endothelium ing endothelial cells: A study using quail-chick chimeras.
with unique properties. Exp Cell Biol 1987; 55:1-10. Devel Biol 1981; 84:183-192.
13. Hendriks HR, Eestermans IL. Disappearance and reap- 33. Milici AJ, Furie MB, Carley WW. The formation of fen-
pearance of high endothelial venules and immigrating estrations and channels by capillary endothelium in vitro.
lymphocytes in lymph nodes deprived of afferent lym- Proc Natl Acad Sci USA 1985; 82:6181-6185.
phatic vessels: A possible regulatory role of macrophages 34. Carley W, Milici AJ, Madri JA. Extracellular matrix speci-
in lymphocyte migration. Eur J Immunol 1983; ficity for the differentiation of capillary endothelial cells.
13:663-669. Exp Cell Res 1988; 178:426-434.
14. Miller JJ III. Studies of the phylogeny and ontogeny of 35. Lombardi T, Montesano R, Furie MB et al. In vitro modu-
the specialized lymphatic vessels. Lab Invest 1969; lation of endothelial fenestrae: Opposing effects of retinoic
21:484-490. acid and transforming growth factor !. J Cell Sci 1988;
15. Freemont AJ, Jones CJP, Bromley M et al. Changes in 91:313-318.
vascular endothelium related to lymphocyte collections in 36. Couvelard A, Scoazec J-Y, Dauge M-C et al. Structural
diseased synovia. Arthr Rheum 1983; 26:1427-1433. and functional differentiation of sinusoidal endothelial
16. Duijvestijn AM, Schreider AB, Butcher EC. Interferon- cells during liver organogenesis in humans. Blood 1996;
gamma regulates an antigen specific for endothelial cells 87:4568-4580.
involved in lymphocyte trafficking. Proc Natl Acad Sci 37. Couvelard A, Scoazec J-Y, Feldmann G. Expression of cell-
USA 1986; 83:9114-9118. cell and cell-matrix adhesion proteins by sinusoidal en-
17. Bishop DK, Sedmak DD, Leppink DM et al. Vascular en- dothelial cells in the normal and cirrhotic liver. Am J
dothelial differentiation in sponge matrix allografts. Hum Pathol 1993; 143:738-752.
Immunol 1990; 28:128-133. 38. Grevers G. The role of fenestrated vessels for the secre-
18. Roberts WG, Palade GE. Increased microvascular perme- tory process in the nasal mucosa: A histological and trans-
ability and endothelial fenestration induced by vascular mission electron microscopic study in the rabbit. Laryn-
endothelial growth factor. J Cell Sci 1995; 108:2369-2379. goscope 1993; 103:1255-1258.
19. Schaffner F, Popper H. Capillarization of hepatic sinuso- 39. Beresford WA. A stromal role in epithelial metaplasia and
ids. Gastroenterology 1963; 44:239-242. cancer. Cancer J 1988; 2:145-147.
20. Martinez-Hernandez A, Martinez J. The role of 40. Markwald RR, Fitzharris TP, Adams Smith WN. Struc-
capillarization in hepatic failure: Studies in carbon tetra- tural analysis of endocardial differentiation. Devel Biol
chloride-induced cirrhosis. Hepatology 1991; 14:864-874 1975; 42:160-180.
21. Cui S, Hano H, Sakata A et al. Enhanced CD34 expres- 41. Sinning AR, Krug EL, Markwald RR. Multiple glycopro-
sion of sinusoid-like vascular endothelial cells in hepato- teins localize to a particulate form of extracellular matrix
cellular carcinoma. Pathol Int 1996; 46:751-756. in regions of the embryonic heart where endothelial cells
22. Nakamura S, Muro H, Suzuki S et al. Immunohistochemi- transform into mesenchyme. Anat Rec 1992; 232:285-292.
cal studies on endothelial cell phenotype in hepatocellu- 42. Zhang H-J, Kluge M, Timpl R et al. The extracellular
lar carcinoma. Hepatology 1997; 26:407-415. matrix glycoproteins BM-90 and tenascin are expressed
23. Haratake J, Scheuer PJ. An immunohistochemical and in the mesenchyme at sites of endothelial-mesenchymal
ultrastructural study of the sinusoids of hepatocellular conversion in the embryonic mouse heart. Differentiation
carcinoma. Cancer 1990; 65:1985-1993. 1993; 52:211-220.
24. McKinney RY, Singh BB, Brewer PD. Fenestrations in 43. Nakajima Y, Mironov V, Yamagishi T et al. Expression
regenerating skeletal muscle capillaries. Am J Anat 1977; of smooth muscle alpha-actin in mesenchymal cells dur-
150:213-218. ing formation of avian endocardial cushion tissue: A role
25. Burns MS, Hartz MJ. The retinal pigment epithelium in- for transforming growth factor !3. Devel Dynam 1997;
duces fenestration of endothelial cells in vivo. Current Eye 209:296-309.
Res 1992; 11:863-873. 44. Nakajima Y, Miyazono K, Kato M et al. Extracellular
26. Braverman IM, Yen A. Ultrastructure of the capillary loops fibrillar structure of latent TGF! binding protein-1: Role
in the dermal papillae of psoriasis. J Invest Dermatol 1977; in TGF!-dpendent endothelial-mesenchymal transforma-
68:53-60. tion during endocardial cushion formation in mouse
27. Gabella G. Hypertrophic smooth muscle. V. Collagen and embryonic heart. J Cell Biol 1997; 136:193-204.
other extracellular materials. Vascularization. Cell Tissue 45. Bergwerff M, DeRuiter MC, Poelman RE et al. Onset of
Res 1984; 235:275-283. elastogenesis and downregulation of smooth muscle actin
28. Kitchens CS, Weiss L. Ultrastructural changes of endot- as distinguishing phenomena in artery differentiation in
helium associated with thrombocytopenia. Blood 1975; the chick embryo. Anat Embryol 1996; 194:545-557.
46:567-578. 46. DeRuiter MC, Poelmann RE, VanMunsteren JC et al.
29. Roberts WG, Palade GE. Neovasculature induced by vas- Embryonic endothelial cells transdifferentiate into mes-
cular endothelial growth factor is fenestrated. Cancer Res enchymal cells expressing smooth muscle actins in vivo
1997; 57:765-772. and in vitro. Circ Res 1997; 80:444-451.
Transdifferentiation and the Vascular Wall 413
47. Lipton BH, Bensch KG, Karasek MA. Microvessel endot- 68. Tsarfaty I, Rong S, Resau JH et al. The Met proto-
helial cell transdifferentiation: Phenotypic characterization. oncogene mesenchymal to epithelial cell conversion. Sci-
Differentiation 1991; 46:117-133. ence 1994; 263:98-101.
48. Romero LI, Zhang D-N, Herron GS et al. Interleukin-1 69. Pletjushkina OJ, Ivanova OJ, Kaverina IN et al. Taxol-
induces major phenotypic changes in human skin mi- treated fibroblasts acquire an epithelioid shape and a cir-
crovascular endothelial cells. J Cell Physiol 1997 173:84-92. cular pattern of actin bundles. Exp Cell Res 1994;
49. Majesky MW, Schwartz SM. An origin for smooth muscle 212:201-208.
cells from endothelium. Circ Res 1997; 80:601-603. 70. Mecham RP, Whitehouse LA, Wrenn DS et al. Smooth
50. Liao W, Bisgrove BW, Sawyer H et al. The zebrafish gene muscle-mediated connective tissue remodeling in pulmo-
cloche acts upstream of a Flk-1 homologue to regulate nary hypertension. Science 1987; 237:423-426.
endothelial cell differentiation. Development 1997; 71. Arribas SM, Hillier C, Gonzalez C et al. Cellular aspects
124:381-389. of vascular remodeling in hypertension revealed by con-
51. Shivdasani RA, Orkin SH. The transcriptional control of focal microscopy. Hypertension 1997; 30:1455-1464.
hematopoiesis. Blood 1996; 87:4025-4039. 72. Gabbiani G, Majno G. Dupuytren’s contracture: Fibro-
52. Bijl J, van Oostveen JW, Kreike M et al. Expression of blast contraction? Am J Pathol 1972; 66:131-146.
HOXC4, HOXC5, and HOXC6 in human lymphoid cell 73. Messier RH Jr, Bass BL, Aly HM et al. Dual structural
lines, leukemias, and benign and malignant lymphoid tis- and functional phenotypes of the porcine aortic valve in-
sue. Blood 1996; 87:1737-1745. terstitial population: Characteristics of the leaflet
53. Komuro T. Re-evaluation of fibroblasts and fibroblast-like myofibroblast. J Surg Res 1994; 57:1-21.
cells. Anat Embryol 1990; 182:103-112. 74. Ronnov-Jessen L, Petersen OW. Induction of #-smooth
54. Abrahamsohn PA, Zorn TMT. Implantation and muscle actin by transformtin growth factor-!1 in quies-
decidualization in rodents. J Exp Zool 1993; 266:603-628. cent human breast gland fibroblasts. Lab Invest 1993;
55. Kaissling B, Hegyi I, Loffing J et al. Morphology of inter- 68:696-707.
stitial cells in the healthy kidney. Anat Embryol 1996; 75. Nakanishi I, Kajikawa K, Okada Y et al. Myofibroblasts
193:303-318. in fibrous tumors and fibrosis in various organs. Acta
56. Kaissling B, Spiess S, Rinne B et al. Effects of anemia on Pathol Jpn 1981; 31:423-437.
morphology of rat renal cortex. Am J Physiol 1993; 76. Hebda PA, Collins MA, Tharp MD. Mast cell and
264:F608-F617. myofibroblast in wound healing. Dermatologic Clinics
57. Koury ST, Koury MJ, Bondurant MC et al. Quantitation 1993; 11:685-696.
of erythropoietin-producing cells in kidneys of mice by 77. Wehrens XHT, Mies B, Gimona M et al. Localization of
in situ hybridization: correlation with hematocrit, renal smoothelin in avian smooth muscle and identification of
erythropoietin mRNA, and serum erythropoietin concen- a vascular-specific isoform. FEBS Lett 1997; 405:315-320.
tration. Blood 1989; 74:645-651. 78. Saalbach A, Anderegg U, Bruns M et al. Novel fibroblast-
58. Maxwell PH, Ferguson DJP, Nicholls LG et al. Sites of specific monoclonal antibodies: Properties and specifici-
erythropoietin production. Kidney Int 1997; 51:393-401. ties. J Invest Dermatol 1996; 106:1314-1319.
59. Mizuno S, Glowacki J. Chondroinduction of human der- 79. Gabbiani G. The biology of the myofibroblast. Kidney Int
mal fibroblasts by demineralized bone in three-dimen- 1992; 41:530-532.
sional culture. Exp Cell Res 1996; 227:89-97. 80. Kapanci Y, Ribaux C, Chaponnier C et al. Cytoskeletal
60. Herr G, Hartwig C-H, Boll C et al. Ectopic bone forma- features of alveolar myofibroblasts and pericytes in nor-
tion by composites of BMP and metal implants in rats. mal human and rat lung. J Histochem Cytochem 1992;
Acta Orthop Scand 1996; 67:606-610. 40:1955-1963.
61. Beresford WA. Ectopic cartilage, neoplasia, and metapla- 81. Buoro S, Ferrarese, Chiavegato A et al. Myofibroblast-de-
sia. In: Hall BK, ed. Cartilage, vol 3. New York: Academic rived smooth muscle cells during remodelling of rabbit
Press, 1983:1-48. urinary bladder wall induced by partial outflow obstruc-
62. Kinoshita T, Takahama H, Sasaki F. Changes in the func- tion. Lab Invest 1993; 69:589-602.
tion of dermal fibroblasts in the tadpole tail during anu- 82. Oda D, Gown AM, Vande Berg JS et al. Instability of the
ran metamorphosis. J Exp Zool 1991; 257:166-177. myofibroblast phenotype in culture. Exp Mol Pathol 1990;
63. Pampinella F, Roelofs M, Castellucci E et al. Proliferation 52:221-234.
of submesothelial mesenchymal cells during early phase 83. Berndt A, Kosmehl H, Katenkamp D et al. Appearance of
of serosal thickening in the rabbit bladder is accompa- the myofibroblastic phenotype in Dupuytren’s disease is
nied by transient keratin 18 expression. Exp Cell Res 1996; associated with a fibronectin, laminin, collagen type IV
223:327-339. and tenascin extracellular matrix. Pathobiology 1994;
64. Fuji S, Nakashima N, Okamura H et al. Progesterone-in- 62:55-58.
duced smooth-muscle-like cells in subperitoneal nodules 84. Reimann T, Hempel U, Krautwald S et al. Transformtin
produced by estrogen. Am J Obstet Gynecol 1981; growth factor-!1 induces activation of Ras, Raf-1, MEK
139:164-172. and MAPK in rat hepatic stellate cells. FEBS Lett 1997;
65. Green H, Kehinde O. Spontaneous heritable changes lead- 403:57-60.
ing to increased adipose conversion in 3T3 cells. Cell 1976; 85. Iehara N, Takeoka H, Tsuji H et al. Differentiation of
7:105-113. smooth muscle phenotypes in mouse mesangial cells. Kid-
66. Pawlina W, Larkin LH, Ogilvie S et al. Human relaxin ney Int 1996; 49:1330-1341.
inhibits division but not differentiation of 3T3-L1 cells. 86. Hewitson TD, Becker GJ. Interstitial myofibroblasts in IgA
Mol Cell Endocrinol 1990; 72:55-61. glomerulonephritis. Am J Nephrol 1995; 15:111-117.
67. Beresford WA. Direct transdifferentiation: Can cells 87. Grupp C, Lottermoser J, Cohen DI et al. Transformation
change their phenotype without dividing? Cell Different of rat inner medullary fibroblasts to myofibroblasts in
Devel 1990; 29:81-93. vitro. Kidney Int 1997; 52:1279-1290.
414 Tissue Engineering of Prosthetic Vascular Grafts
88. Wise CJ, Watt DJ, Jones GE. Conversion of dermal fibro- in coronary arteries of patients with unstable angina pec-
blasts to a myogenic lineage is induced by a soluble fac- toris and postangioplasty restenosis. Circulation
tor derived from myoblasts. J Cell Biochem 1996; 1997:95:1169-1175.
61:363-374. 108. Zhang Y, Ramos KS. The induction of proliferative vas-
89. Eghbali M, Tomek R, Woods C et al. Cardiac fibroblasts cular smooth muscle cell phenotypes by benzo[a]pyrene
are predisposed to convert into myocyte phenotype: Spe- does not involve mutational activation of ras genes. Mu-
cific effect of transforming growth factor !. Proc Natl tation Res 1997; 373:285-292.
Acad Sci USA 1991; 88:795-799. 109. Amann K, Wolf B, Nichols C et al. Aortic changes in ex-
90. Kivirikko S, Saarela J, Myers JC et al. Distribution of type perimental renal failure. Hyperplasia or hypertrophy of
XV collagen transcripts in human tissue and their pro- smooth muscle cells? Hypertension 1997; 29:770-775.
duction by muscle cells and fibroblasts. Am J Pathol 1995; 110. Wagenaar SS, Wagenvoort CA. Experimental production
147:1500-1509. of longitudinal smooth muscle in the intima of muscular
91. Sottiurai VS, Batson RC. Role of myofibroblasts in arteries. Lab Invest 1978; 39:370-374.
pseudointima formation. Surgery 1983; 94:792-801. 111. Shiiki H, Shimokama T, Watanabe T. Temporal arteritis:
92. Skopicki HA, Lyons GE, Schattemann G et al. Embryonic Cell composition and possible pathogenetic role of cell-
expression of the Gax homeodomain protein in cardiac, mediated immunity. Hum Pathol 1989; 20:1057-1064.
smooth, and skeletal muscle. Circ Res 1997; 80:452-462. 112. Amano J, Ishiyama S, Nishikawa T et al. Proliferation of
93. De Leon H, Scott NA, Martin F et al. Expression of smooth muscle cells in acute allograft vascular rejection.
nonmuscle myosin heavy chain-B isoform in the vessel J Thorac Cardiovasc Surg 1997:113:19-25.
wall of porcine coronary arteries after balloon angioplasty. 113. Fukumoto Y, Shimokawa H, Ito A et al. Inflammatory
Circ Res 1997; 80:514-519. cytokines cause coronary arteriosclerosis-like changes and
94. Xing Z, Tremblay GM, Sime PJ et al. Overexpression of alterations in the smooth-muscle phenotypes in pigs. J
granulocyte-macrophage colony-stimulating factor induces Cardiovasc Pharmacol 1997; 29:222-231.
pulmonary granulation tissue formation and fibrosis by 114. Dingemans KP, Jansen N, Becker AE. Ultrastructure of
induction of transformtin growth factor- ! 1 and the normal human aortic media. Virchows Arch Pathol
myofibroblast accumulation. Am J Pathol 1997; 150:59-66. Anat 1981:392:199-216.
95. Weber KT, Sun Y, Katwa LC. Myofibroblasts and local 115. Bergwerff M, DeRuiter MC, Poelmann RE et al. Onset of
angiotensin ii in rat cardiac tissue repair. Int J Biochem elastogenesis and downregulation of smooth muscle actin
Cell Biol 1997; 29:31-42. as distinguishing phenomena in artery differentiation in
96. Thieszen SL, Dalton M, Gadson PF et al. Embryonic lin- the chick embryo. Anat Embryol 1996; 194:545-557.
eage of vascular smooth muscle cells determines responses 116. Frid MG, Dempsey EC, Durmowicz AG et al. Smooth
to collagen matrices and integrin receptor expression. Exp muscle heterogeneity in pulmonary and systemic vessels.
Cell Res 1996; 227:135-145. Arterioscler Thromb Vasc Biol 1997; 17:1203-1209.
97. Gomez RA, Chevalier RL, Everett AD et al. Recruitment 117. Seidel CL. Cellular heterogeneity of the vascular tunica
of renin-expressing cells in adult rat kidneys. Am J Physiol media. Implications for vessel wall repair. Arterioscler
1990; 259:F660-F665. Thromb Vasc Biol 1997; 17:1868-1871.
98. Lin WW, McGee GS, Patterson BK et al. Fibromuscular 118. Campbell GR, Chamley-Campbell JH. Invited review: The
dysplasia of the brachial artery: A case report and review cellular pathobiology of atherosclerosis. Pathology 1981;
of the literature. J Vasc Surg 1992; 16:66-70. 13:423-440.
99. Stanley JC, Gewertz BL, Bove EL et al. Arterial 119. Pauly RR, Passaniti A, Crow M et al. Experimental mod-
fibrodysplasia. Histopathologic character and current etio- els that mimic the differentiation and dedifferentiation of
logic concepts. Arch Surg 1975; 110:561-566. vascular cells. Circulation 1992; 86[suppl III]:III68-III73.
100. Sottiurai V, Fry WJ, Stanley JC. Ultrastructural charac- 120. Aikawa M, Sakomura Y, Ueda M et al. Redifferentiation
teristics of experimental arterial medial fibroplasia induced of smooth muscle cells after coronary angioplasty deter-
by vasa vasorum occlusion. J Surg Res 1978; 24:169-177. mined via myosin heavy chain expression. Circulation
101. McGuffee LJ, Little SA. Tunica media remodeling in me- 1997; 96:82-90.
senteric arteries of hypertensive rats. Anat Rec 1996; 121. Jahn L, Franke WW. High frequency of cytokeratin-pro-
246:279-292. ducing smooth muscle cells in human atherosclerotic
102. Dickhout JG, Lee RMKW. Structural and functional analy- plaques. Differentiation 1989; 40:55-62.
sis of small arteries from young spontaneously hyperten- 122 Hedin U, Holm J, Hansson GK. Induction of tenascin in
sive rats. Hypertension 1997; 29:781-789. rat arterial injury. Relationship to altered smooth muscle
103. Tennant M, McGeachie JK. Adaptive remodelling of cell phenotype. Am J Pathol 1991; 139:649-656.
smooth muscle in the neo-intima of vein-to-artery grafts 123. MacBeath JR, Kielty CM, Shuttleworth CA. Type VIII col-
in rats: A detailed morphometric analysis. Anat Embryol lagen is a product of vascular smooth-muscle cells in de-
1993; 187:161-166. velopment and disease. Biochem J 1996; 319:993-998.
104. Shi Y, O’Brien JE, Mannion JD et al. Remodeling of au- 124. Yamamoto M, Nakamura H, Yamato M et al. Retarda-
tologous saphenous vein grafts. The role of perivascular tion of phenotypic transition of rabbit arterial smooth
myofibroblasts. Circulation 1997; 96:2684-2693. muscle cells in three-dimensional primary culture. Exp
105. Milroy CM, Scott DJA, Beard JD et al. Histological ap- Cell Res 1996; 225:12-21.
pearances of the long saphenous vein. J Pathol 1989; 125. Sjolund M, Hedin U, Sejersen T et al. Arterial smooth
159:311-316. muscle cells express platelet-derived growth factor (PDGF)
106. Chamley-Campbell J, Campbell GR, Ross R. The smooth A chain mRNA, secrete a PDGF-like mitogen, and bind
muscle cell in culture. Physiol Rev 1979; 59:1-61. exogenous PDGF in a phenotype- and growth state-de-
107. Chen Y-H, Chen Y-L, Lin S-J et al. Electron microscopic pendent manner. J Cell Biol 1988; 106:403-413.
studies of phenotypic modulation of smooth muscle cells
Transdifferentiation and the Vascular Wall 415
126. Cizmeci-Smith G, Langan E, Yuokey J et al. Syndecan-4 142. Graham MF, Bryson GR, Diegelmann RF. Transforming
is a primary-response gene induced by basic fibroblast growth factor !1 selectively augments collagen synthesis
growth factor and arterial injury in vascular smooth by human intestinal smooth muscle cells. Gastroenterol-
muscle cells. Arterioscler Thromb Vasc Biol 1997; ogy 1990; 99:447-453.
17:172-180. 143. McHugh KM. Molecular analysis of gastrointestinal
127. Nakasaki Y, Iwamoto T, Hanada H et al. Cloning of the smooth muscle development. J Ped Gastroenterol Nutrit
rat aortic smooth muscle Na+/Ca2+ exchanger and tissue- 1996; 23:379-394.
specific expression of isoforms. J Biochem 1993; 144. Anderson DR, Davis EB. Glaucoma, capillaries, and
114:528-534. pericytes. 2. Identification and characterization of retinal
128. Kato S, Stanley JR, Fox JC. Serum stimulation, cell-cell pericytes in culture. Ophthalmologica 1996; 210:263-268.
interactions, and extracellular matrix independently influ- 145. Schor AM, Canfield AE, Sutton AB et al. Pericyte differ-
ence smooth muscle cell phenotype in vitro. Am J Pathol entiation. Clin Orthop Rel Res 1995:313:81-91.
1996; 149:687-697. 146. Schulze C, Firth JA. Junctions between pericytes and the
129. Miano JM, Vlasic N, Tota RR et al. Smooth muscle im- endothelium in rat myocardial capillaries: A morphomet-
mediate-early gene and growth factor activation follows ric and immunogold study. Cell Tissue Res 1993;
vascular injury. Arterioscler Thromb Vasc Biol 1993; 271:145-154.
13:211-219. 147. Christ GJ, Spray DC, El-Sabban M et al. Gap junctions
130. Merrilees MJ, Campbell JH, Spanidis E et al. Glycosami- in vascular tissues. Circ Res 1996; 79:631-646.
noglycan synthesis by smooth muscle cells of differing 148. Meyrick B, Reid L, The effect of continued hypoxia on
phenotype and their response to conditioned medium. rat pulmonary arterial circulation. Lab Invest 1978;
Atherosclerosis 1990; 81:245-254. 38:188-200.
131. Bendeck MP, Zempo N, Clowes AW et al. Smooth muscle 149. Nehls V, Drenckhahn D. The versatility of microvascular
cell migration and matrix metalloproteinase expression pericytes: From mesenchyme to smooth muscle? His-
after arterial injury in the rat. Circ Res 1994; 75:539-545. tochemistry 1993; 99:1-12.
132. Shofuda K-i, Yasumitsu H, Nishihashi A et al. Expression 150. Canfield AE, Sutton AB, Hoyland JA et al. Association of
of three membrane-type matrix metalloproteinases (MT- thrombospondin-1 with osteogenic differentiation of reti-
MMPs) in rat vascular smooth muscle cells and charac- nal pericytes in vitro. J Cell Sci 1996; 109:343-353.
terization of MT3-MMPs with and without transmem- 151. Doherty MJ, Ashton BA, Grant ME et al. Vascular
brane domain. J Biol Chem 1997; 272:9749-9754. pericytes express osteogenic markers in vitro and form
133. Thyberg J, Roy J, Tran PK et al. Expression of caveolae bone in vivo. Int J Exp Pathol 1996; 77:32A.
on the surface of rat arterial smooth muscle cells is de- 152. Sundberg C, Ivarsson M, Gerdin B et al. Pericytes as col-
pendent on the phenotypic state of the cells. Lab Invest lagen-producing cells in excessive scarring. Lab Invest
1997; 77:93-101. 1996:74:452-466.
134. Gadson PF Jr, Dalton ML, Patterson et al. Differential 153. Galli SJ. New insights into “The Riddle of the Mast cells”:
response of mesoderm- and neural crest-derived smooth Microenvironmental regulation of mast cell development
muscle to TGF- ! 1: regulation of c-myb and # 1(I) and phenotypic heterogeneity. Lab Invest 1990; 62:5-33.
procollagen genes. Exp Cell Res 1997; 230:169-180. 154. Church MK, Levi-Schaffer F. The human mast cell. J Al-
135. Lee SH, Yan H, Reeser JC et al. Proteoglycan biosyntheis lergy Clin Immunol 1997; 99:155-160.
is required in BC3H1 myogenic cells for modulation of 155. Jeziorska M, McCollum C, Wooley DE. Mast cell distri-
vascular smooth muscle #-actin gene expression in re- bution, activation, and phenotype in atherosclerotic le-
sponse to microenvironmental signals. J Cell Physiol 1995; sions of human carotid arteries. J Pathol 1997;
164:172-186. 182:115-122.
136. Sklaletz-Rorowski A, Schmidt A, Breithardt G et al. Hep- 156. Weidner N, Austen KF. Evidence for morphologic diver-
arin-induced overexpression of basic fibroblast growth sity of human mast cells. An ultrastructural study of mast
factor, basic fibroblast growth factor receptor, and cell- cells from multiple body sites. Lab Invest 1990; 63:63-72.
associated proteoheparan sulfate in cultured coronary 157. Otsu K, Nakano T, Kanakura Y et al. Phenotypic changes
smooth muscle cells. Arterioscler Thromb Vasc Biol 1996; of bone marrow-derived mast cells after intraperitoneal
16:1063-1069. transfer into W/Wv mice that are genetically deficient in
137. Sibinga NES, Foster LC, Hsieh C-M et al. Collagen VIII mast cells. J Exp Med 1987; 165:615-627.
is expressed by vascular smooth muscle cells in response 158. Yamamura T, Nakano T, Fukuzumi T et al. Electron mi-
to vascular injury. Circ Res 1997; 80:532-541. croscopic changes of bone-marrow derived cultured mast
138. Kacem K, Bonvento G, Seylaz J. Effect of sympathectomy cells after injection into the skin of genetically mast cell-
on the phenotype of smooth muscle cells of middle cere- deficient W/Wv mice. J Invest Dermatol 1988:91:269-273.
bral and ear arteries of hyperlipidaemic rabbits. Histochem 159. Sonoda S, Sonoda T, Nakano T et al. Development of
J 1997; 29:279-286. mucosal mast cells after injection of a single connective
139. Carmeliet P, Mackman N, Moons L et al. Role of tissue tissue-type mast cell in the stomach mucosa of geneti-
factor in embryonic blood vessel development. Nature cally mast cell-deficient W/Wv mice. J Immunol 1986;
1996; 383:73-75. 137:1319-1322.
140. Halyko AJ, Rector E, Stephens NL. Characterization of 160. Kanakura Y, Kuriu A, Waki N et al. Multiple bidirectional
molecular determinants of smooth muscle heterogeneity. alterations of phenotype and changes in proliferative po-
Can J Physiol Pharmacol 1997; 75:917-929. tential during the in vitro and in vivo passage of clonal
141. Tom-Moy M, Madison JM, Jones CA et al. Morphologic mast cell populations derived from mouse peritoneal mast
characterization of cultures of smooth muscle cells iso- cells. Blood 1988; 72:877-885.
lated from the tracheas of adult dogs. Anat Rec 1987; 161. Dayton ET, Pharr P, Ogawa M et al. 3T3 fibroblasts in-
218:313-328. duce cloned interleukin 3-dependent mouse mast cells to
416 Tissue Engineering of Prosthetic Vascular Grafts
resemble connective tissue mast cells in granular constitu- chorionic villi of mouse placenta with special reference
ency. Proc Natl Acad Sci USA 1988; 85:569-572. to the origin of Hofbauer cells. J Leukocyte Biol 1991;
162. Brugger W, Reinhardt D, Galanos C et al. Inhibition of 50:57-68.
in vitro differentiation of human monocytes to macro- 175. Borello MA, Phipps RP. Fibroblasts support outgrowth of
phages by lipopolysaccharides (LPS): Phenotypic and func- splenocytes simultaneously expressing B lymphocyte and
tional analysis. Int Immunol 1991; 3:221-227. macrophage characteristics. J Immunol 1995;
163. Ring WL, Riddick CA, Baker JR et al. Human monocytes 155:4155-4161.
lose 5-lipoxygenase and FLAP as they mature into mono- 176. Orikasa M, Iwanaga Y, Takahashi-Iwanaga H et al. Mac-
cyte-derived macrophages in vitro. Am J Physiol 1996; rophagic cells outgrowth from normal rat glomerular cul-
271:C372-C377. ture: Possible metaplastic changes from podocytes. Lab
164. Polverini PJ. How the extracellular matrix and macro- Invest 1996; 75:719-733.
phages contribute to angiogenesis-dependent diseases. Eur 177. Takahashi K, Naito M, Takeya M. Development and het-
J Cancer 1996; 32A:2430-2437. erogeneity of macrophages and their related cells through
165. Anrather J, Csizmadia V, Brostjan C et al. Inhibition of their differentiation pathways. Pathol Int 1997; 46:473-485.
bovine endothelial cell activation in vitro by regulated 178. Niwa H, Harrison LC, DeAizpurua HJ et al. Identifica-
expression of a transdominant inhibitor of NF-kappaB. J tion of pancreatic ! cell-related genes by representational
Clin Invest 1997; 99:763-772. difference analysis. Endocrinology 1997; 138:1419-1426.
166. De Meyer GRY, Herman AG. Vascular endothelial dys- 179. Yeh H-I, Dupont E, Coppen S et al. Gap junction local-
function. Progr Cardiovasc Dis 1997; 39:325-342. ization and connexin expression in cytochemically iden-
167. Fowler SD, Mayer EP, Greenspan P. Foam cells and tified endothelial cells of arterial tissue. J Histochem
atherogenesis. Ann NY Acad Sci 1985; 454:79-90. Cytochem 1997; 45:539-550.
168. Greenspan P, Yu H, Mao F et al. Cholesterol deposition 180. Desmouliere A, Badid C, Bochaton-Piallat M-L et al.
in macrophages: Foam cell formation mediated by cho- Apoptosis during wound healing, fibrocontractive diseases
lesterol-enriched oxidized low density lipoprotein. J Lipid and vascular wall injury. Int J Biochem Cell Biol 1997;
Res 1997; 38:101-109. 29:19-30.
169. Wang N, Winchester R, Ravalli S et al. Interleukin 8 is 181. Wilmut I, Schnieke AE, McWhir J et al. Viable offspring
induced by cholesterol loading of macrophages and ex- derived from fetal and adult mammalian cells. Nature
pressed by macrophage foam cells in human atheroma. J 1997; 385:810-813.
Biol Chem 1996; 271:8837-8842. 182. van Neck JW, Medina JJ, Onnekink C et al. Basic fibro-
170. Feigl W, Susani M, Ulrich W et al. Organisation of ex- blast growth factor has a differential effect on MyoD con-
perimental thrombosis by blood cells. Virchows Arch version of cultured aortic smooth muscle cells from new-
Pathol Anat 1985; 406:133-148. born and adult rats. Am J Pathol 1993; 143:269-282.
171. Campbell GR, Ryan GB. Origin of myofibroblasts in the 183. Lee JE. NeuroD and neurogenesis. Dev Neuroscience 1997;
avascular capsule around free-floating intraperitoneal 19:27-32.
blood clots. Pathology 1983; 15:253-261. 184. Fisher LJ, Jinnah HA, Kale LC et al. Survival and func-
172. Vaage J, Harlos JP. Collagen production by macrophages tion of intrastriatally grafted primary fibroblasts geneti-
in tumour encapsulation and dormancy. Br J Cancer 1991; cally modified to produce l-DOPA. Neuron 1991;
63:758-762. 6:371-380.
173. Moujahid A, Navascues J, Marin-Teva JL et al. Macro- 185. Urist M. Bone morphogenetic protein: The
phages during avian optic nerve development: Relation- molecularization of skeletal system development. J Bone
ship to cell death and differentiation into microglia. Anat Mineral Res 1997; 12:343-346.
Embryol 1996; 193:131-144. 186. Okada TS. Transdifferentiation. Oxford: Clarendon Press,
174. Takahashi K, Naito M, Katabuchi H et al. Development, 1991:1-233.
differentiation, and maturation of macrophages in the
Facilitation of Healing
Transdifferentiation
CHAPTER 38
Endothelial Cells Transformed
from Fibroblasts During Angiogenesis
Takashi Fujiwara, Kazunori Kon
Introduction
A ngiogenesis, the formation of new blood vessels, is of fundamental importance for sev
eral physiological and pathological processes. It occurs, as is well known, during organ
development, wound healing and tumor growth.1-6 Endothelial cells of newly formed blood
vessels are widely believed to derive from preexisting blood vessels. According to Furcht,7
and Rhodin and Fujita,8 the process of angiogenesis can be broken down into a number of
discrete yet overlapping steps:
1. Increase of vessel permeability;
2. Dissolution of basal lamina caused by a variety of enzymes and penetration of basal
lamina by endothelial projections;
3. Matrix ‘channel’ formation in the interstitial connective tissue and migration of en-
dothelial cells from the preexisting vessel;
4. Proliferation of endothelial cells;
5. Sprout and loop formation;
6. Canalization of the loops; and
7. Association of pericytes and fibroblasts with the newly formed blood vessels.
In such an angiogenic process, endothelial cell migration from preexisting vessels and
its proliferation is requisite. However, questions have been addressed to this current angio-
genic mechanism9 because of several puzzling phenomena outlined as follows:
1. Nonendothelial cells are integrated into the linings of newly formed blood vessels;10-12
2. Angiogenesis takes place without apparent endothelial cell proliferation13 and with-
out endothelial cell migration from preexisting blood vessels;10 and
3. Factors inhibiting in vitro proliferation of endothelial cells, tumor necrosis factor-#
(TNF-#)14 and transformtin growth factor-! (TGF-!),15 stimulate in vivo angiogen-
esis.16
Therefore, we aimed to examine whether endothelial cells of preexisting blood vessels
are the only origin of endothelial cells of newly formed blood vessels.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
418 Tissue Engineering of Prosthetic Vascular Grafts
(40 mg/kg body weight).17 Newly formed blood vessels ap- was confirmed by their spindle shape and by the inhibitory
peared in the chamber space about one week after the cham- effect of cis-hydroxyl proline on fibroblast proliferation.20
ber implantation. The newly formed vessels invaded the When 100 mg/ml of cis-hydroxyl proline was added to the
space which extravasated fibrin and red blood cells filled. cultures, the cellular number decreased as shown previ-
For mapping the network of newly formed blood vessels, ously.20 Furthermore, no contamination of endothelial cells
invaginated red cells prepared by treating with trifluopera- in culture was ascertained by the negative uptake to fluores-
zine were perfused through the ear artery, enabling us to cence labeled acetyl low density lipoprotein by the cultured
detect tips of blood vessels growing among numerous ex- cells.21 During the fifth passage, fibroblasts were cultured in
travasated red cells in the REC. After fixation, newly formed MEM containing India ink particles for 10 h to mark fibro-
blood vessels were processed for electron microscopy in usual blasts by their ingestion of India ink particles.22 Fibroblasts
way and observed with an electron microscope. contain India ink particles in the cytoplasm for as long as 9
Some newly formed blood vessels containing invagi- months, enabling us to keep fibroblasts marked with India
nated red cells were lined with partially discontinuous en- ink particles for a long time. The marked fibroblasts, about
dothelium (Fig. 38.1). Endothelial cells of the blood vessel 1 x 104 cells, were washed to remove free carbon particles
were provided with well developed rough ER and many mi- from the suspension, with MEM solution two times and with
tochondria, morphologically resembling fibroblasts in the Hanks’ solution three times, and then transplanted
interstitium. This phenomenon suggests that endothelial autologously into the space between protective covers of the
cells may derive from fibroblasts. Then we paid particular chamber with a fine needle at 2 days after the chamber im-
attention to the possible role of fibroblasts as progenitors of plantation. On 8 days after the autologous transplantation
endothelial cells, since mesenchymal cells are known to be of the marked fibroblasts into REC, growing vessels in the
the origin of vascular endothelial cells in fetal tissue, and REC were fixed and cut into sections for light microscopy.
fibroblasts function as embryonic mesenchymal cells in some Newly formed blood vessels of various diameters were
circumstances in adult tissue.18 found in the connective tissue, identified as venules, capil-
To give an insight into the transformation of fibro- laries and arterioles by their morphological characteris-
blasts into endothelial cells, transplantation of cultured fi- tics.23-25 Endothelial cells containing carbon particles in the
broblasts into the space of REC was designed. For this pur- cytoplasm were, though only occasionally, found to line
pose, the fibroblasts were marked so as to trace them. We newly formed blood vessels (Fig. 38.2), indicating that the
used two methods for marking fibroblasts, carbon particle transplanted fibroblasts transformed into the endothelial
ingestion and [3H]thymidine uptake of fibroblasts. cells. Extravasation of red cells from the openings of newly
formed blood vessels was sometimes observed in the speci-
Transformation of Fibroblasts Marked with Carbon mens prepared after autologous transplantation of carbon-
Particles into Endothelial Cells marked fibroblasts (Fig. 38.2). This is consistent with the
Fibroblasts were separated from rabbit ear skin as re- well known high permeability and fragility of newly formed
ported previously19 and were cultured in Eagle’s minimal blood vessels.26
essential medium (MEM). The identification of fibroblasts
Transformation of Fibroblasts Labeled with 66.8 ± 8.1 for endothelial cells and 55.9 ± 4.5 for fibroblasts
[3H]Thymidine into Endothelial Cells in 10 preparations examined, indicating that the radioactive
We labeled fibroblasts with [ 3 H]thymidine to labels are not transmitted to the endothelial cells by uncon-
confirm the result obtained by fibroblasts marked with trolled uptake from degenerating transplanted fibroblasts.
carbon particles. In electron microscope autoradiography, a few silver
Fibroblasts were separated from rabbit ear skin and grains were found over the nuclei of the endothelial cells of
cultured as mentioned above. During the fifth passage, capillaries, arterioles (Fig. 38.4A) and venules (Fig. 38.4B).
the fibroblasts were cultured in MEM containing The labeled cells were joined together by intercellular junc-
[3H]thymidine (185 Kbq/ml) for 6 days for labeling. To re- tions and were surrounded by a basal lamina on their
move free [3H]thymidine, the fibroblasts were washed twice abluminal surface, provided with some of the morphologi-
with MEM solution, then with Hanks’ solution three times. cal characteristics of endothelial cell. These data also indi-
Two days after the chamber implantation, the labeled fibro- cate that fibroblasts transform into endothelial cells of newly
blasts, about 1 x 104 cells, were autologously transplanted formed blood vessels.
into the space between protective covers of the chamber with Silver grains were observed over the nuclei of the
a fine needle. Tissues of the proximal part of the vascular pericytes of capillaries (Fig.38.3A) and venules (Fig. 38.3F),
bed were prepared for light microscopic autoradiography and the nuclei of arteriolar smooth muscle cells (Figs.
on days 7, 9, 12 and 23 and for electron microscopic autora- 38.3C,D, 38.4A), suggesting that both pericytes and vascu-
diography on day 12 after the chamber implantation. lar smooth muscle cells are derived from fibroblasts.
Capillaries (Figs. 38.3A,B), arterioles (Figs. 38.3C,D)
and venules (Figs. 38.3E,F) were found in autoradiographic Discussion
preparations. Numerous autoradiographic silver grains were
frequently observed over the endothelial cell nuclei of these Endothelial Cell Origin and Formation
newly formed blood vessels.27 There were also many fibro- of New Blood Vessels
blasts labeled with silver grains in the connective tissue Endothelial cells of newly formed blood vessels are
(Figs. 38.3A,D). In our experiments, it could not be excluded generally believed to derive from preexisting blood vessels.
that endothelial cells took up radioactive compounds which Pioneering studies on transformation of nonendothelial
might be released from degenerated fibroblasts and diffused cells, such as mesenchymal cells10 and tumor cells,11 into
toward the endothelial cells. If so, however, the concentra- endothelial cells have been carried out. These studies, how-
tion of [3H]thymidine in the interstitium surrounding en- ever, have not attracted general notice, probably because their
dothelial cells of newly formed blood vessels must be lower evidence is indirect. Here, we clearly showed by tracing cul-
than that in our culture medium, predicting that silver grain tured fibroblasts marked with carbon particles that fibro-
density on endothelial cell nucleus must be lower than that blasts transform into endothelial cells of newly formed blood
of fibroblasts. Furthermore, if diffusion of [3H]thymidine vessels, challenging the general agreement on angiogenic
from degenerated fibroblasts occurred in the interstitium, mechanism. The transformation of fibroblasts into endot-
almost all of the endothelial cells and fibroblasts should be helial cells has been supported by an experiment with quail
more or less labeled. In our experiment, the silver grain chick chimeras. Stein et al28 transferred limb buds of three
density on each nucleus of endothelial cells was very similar day old quail embryos to the chorioallantoic membrane of
to that of fibroblasts, and not all of the endothelial cells and 10 to 14 day old chick embryos. Six days after grafting, sev-
fibroblasts were labeled: The percentage of labeled cells was eral quail-derived capillaries were found in each of the
Fig. 38.3. Light microscopic autoradiography. Autoradiographic silver grains are found over the nuclei of the endothelial cells of
capillaries (A, B), arterioles (C,D) and venules (E,F). Silver grains are also observed over the nuclei of the pericytes of capillaries (A)
and venules (F) and over the nuclei of the smooth muscle cells of arterioles (C, D). Fibroblasts labeled with silver grains are
distributed in the connective tissue (A, D). Tissues were prepared on days 7 (E), 9 (A), 12 (C,D and F) and 23 (B), after chamber
implantation. e: endothelial cell; p: pericyte; s: smooth muscle cell. x930: Bar = 10 ∝m.
Endothelial Cells Transformed from Fibroblasts During Angiogenesis 421
Fig. 38.4. Electron microscopic autoradiography. The endothelial cells of an arteriole (A) and a venule
(B) and the smooth muscle cell of an arteriole are labeled with silver grains over their nuclei. The
endothelial cells are joined together with intercellular junctions and surrounded by a basal lamina,
exhibiting typical features of endothelial cells. The specimens were prepared on day 12 after cham-
ber implantation. e: endothelial cell; s: smooth muscle cell (A) x7600: Bar = 1 ∝m; (B) x5400: Bar =
1 ∝m.
transplanted limb buds, suggesting that quail fibroblasts, ing a new blood vessel. Such an incorporation of cells from
better to say mesenchymal cells, participate in new blood the abluminal side into newly formed blood vessels has been
vessel formation. Similar findings that some mesenchymal drawn schematically.29
cells have an ability to transform into endothelial cells have
been reported by several studies on quail chick transplanta- 0rigin of Pericytes and Vascular Smooth Muscle Cells
tion in the embryonic stage,29-31 and on mammalian em- from Fibroblasts
bryos.32 These studies indicate that the endothelial cells from Fibroblasts have also been clearly demonstrated here
preexisting blood vessels are not the only origin of endothe- to be the origin of both pericytes and smooth muscle cells
lial cells of newly formed blood vessels. Recently Asahara et of blood vessels, as shown previously.8,28,34,35 Accordingly,
al12 have clearly demonstrated that putative endothelial cell these results indicate that fibroblasts can be the common
progenitors in circulating blood differentiated into endot- origin for all cellular elements of blood vessels, i.e., endo-
helial cells in vitro and incorporated into sites of active an- thelial cells, pericytes and smooth muscle cells. A similar phe-
giogenesis in vivo. nomenon of cell origin is known in fetal vasculogenesis, in
We observed that endothelium of newly formed ves- which mesenchymal cells are the common origin for these
sels was occasionally discontinuous and that blood cells cir- three cellular elements.36 This is consistent with the view
culating in the vessels extravasated into interstitial space that fibroblasts function in adult tissue as embryonic mes-
through the gaps of endothelium. This extravasated blood enchymal cells in some aspects.18
may flow back and forth in the interstitium and show only
oscillatory movements as described by Hadlicka,33 forming Possible Contribution of Fibroblasts to Angiogenesis
a matrix channel in the interstitial space.8 Fibrin deposits in From our experiments, an increase in fibroblast den-
the intersititial space are supposed to play a critical role in sity can be expected to promote angiogenesis. This view is
providing a matrix channel, from our experimental finding supported by the experimental evidences that activated fi-
that fibrinosis of accumulated fibrin in REC by plasmino- broblasts are associated with angiogenesis in electrically
gen activator causes complete inhibition of new vessel for- stimulated skeletal muscle37 and that inhibition of fibroblast
mation. Fibroblasts in the interstitium may find an oppor- proliferation by proline analogs,20 on the contrary, inhibits
tunity to face and directly contact flowing blood in the ma- angiogenesis.38 The puzzling phenomenon of TNF-# and
trix channel. Then they may come to line the luminal sur- TGF-! promoting in vivo angiogenesis, in spite of their in-
face of the channel in place of normal endothelial cells, form- hibitory effect on endothelial cell proliferation, has
422 Tissue Engineering of Prosthetic Vascular Grafts
previously been rationalized by the suggestion that stimu- ever, we need further studies to reveal whether transforma-
lated secondary cells produce angiogenic factors.4,16 Based tion of fibroblasts into endothelial cells generally takes place
on our data, however, we suggest that TNF-# and TGF-! in angiogenesis.
contribute to promoting angiogenesis by the increasing
population of fibroblasts.39,40 Both TNF-#39 and TGF-!41 are References
mitogens for fibroblasts, and TGF- ! is also a 1. Blood CH, Zetter BR. Tumor interactions with the vas-
chemoattractant40,42 for fibroblasts. Fibrin also stimulates culature: Angiognesis and tumor metastasis. Biochem
angiogenesis43,44 through its proliferative43 and chemotac- Biophys Acta 1990; 1032:89-118.
tic44 effects on fibroblasts. Fibroblast-dependent angiogen- 2. Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer me-
tastasis and angiogenesis: An imblance of positive and
esis is quite favorable for wound healing tissues and tumor
negative regulation. Cell 1991; 64:327-336.
tissues where rapid blood vessel growth is needed, because 3. Risau W. Vasculogenesis, angiogenesis and endothelial dif-
the proliferation rate of endothelial cells has been reported ferentiation during embryonic development. In: Feinberg
to be too slow to account for rapid growth of the vascular RN, ed. The Development of the Vascular System, Issues
network.45 Our data suggest that angiogenesis can be modu- Biomed Volume 14. Basel: Karger, 1991:58-68.
lated by manipulating fibroblast density and activity9 and 4. Folkman J, Brem H. Angiogenesis and inflammation. In:
that fibroblast transformation into endothelial cells can pro- Gallin JI, ed. Inflammation: Basic Principles and Clinical
vide a benefit on clinical applications, in prosthetic vascular Correlates, 2nd edition. New York: Raven Press,
grafts and gene therapy. 1992:821-839.
When arterial reconstruction is required in cases of 5. Folkman J, Shing Y. Angiogenesis. J Biol Chem 1992;
267:10931-10934.
arterial stenosis or occlusion, such as ischemic heart disease,
6. Fidler IJ, Ellis LM. The implications of angiogenesis for
autologous veins or arteries or prosthetic vascular grafts are the biology and thereapy of cancer metastasis. Cell 1994;
used for the replacement of stenotic or occluded arteries. 79:185-188.
Graft failures due to thrombotic occlusion, however, are still 7. Furcht LT. Critical factors controlling angiogenesis: Cell
serious complications.46,47 One of the various methods to products, cell matrix and growth factors. Lab Invest 1986;
reduce the thrombogenicity of prosthetic grafts is to line the 55:505-509.
luminal surface with endothelial cells.48,49 As an attempt at 8. Rhodin JAG, Fujita H. Capillary growth in the mesentery
lining the lumen of synthetic vascular prostheses, microvas- of normal young rats. Intravital video and electron mi-
cular endothelial cells50 and large vessel endothelial cells51 croscope analyses. J Submicrosc Cytol Pathol 1989;
have been seeded and cultured on them.50,51 Recently 21:1-34.
Noishiki et al52 have completely endothelialized the luminal 9. Arnold F, West DC. Angiogenesis in wound healing.
Pharmac Ther 1991; 52:407-422.
surface of a vascular prosthesis by infiltration of bone mar-
10. Aloisi M, Giacomin C, Tessari R. Growth of elementary
row cells and kept patency of the vascular prosthesis for blood vessels in diffusion chambers. I. Process of forma-
longer than three weeks after the implantation of the vascu- tion and conditioning factors. Virchows Arch B Cell
lar prosthesis. Pathol 1970; 6:350-364.
It has also been considered whether the performance 11. Hammersen F, Endrich B, Messmer K. The fine structure
and versatility of prosthetic vascular grafts might be im- of tumor blood vessels. Int J Microcirc Clin Exper 1985;
proved by lining the luminal surface with endothelial cells 4:31-43.
genetically modified to prevent thromboses.49 Furthermore, 12. Asahara T, Murohara T, Sullivan A et al. Isolation of
the endothelial cells have recently been paid attention, since putative progenitor endothelial cells for angiogenesis. Sci-
endothelial cells may serve as vehicles for the introduction ence 1997; 257:964-967.
13. Sholley MM, Ferguson GP, Seibel HR et al. Mechanisms
in vivo of functioning recombinant genes because of their
of neovascularization. Vascular sprouting can occur with-
proximity to the blood stream.53,54 Endothelial cells express- out proliferation of endothelial cells. Lab Invest 1984;
ing recombinant genes have been introduced into denuded 51:624-634.
arteries of syngeneic animals.55 Because fibroblasts, in gen- 14. Fajardo LF, Kwan HH, Kowalski J et al. Dual role of tu-
eral, can more feasibly be isolated from patients, are more mor necresis factor-# in angiogenesis. Amer J Pathol 1992;
easily cultured and proliferate more quickly than endothe- 140:539-544.
lial cells, they may be more appropriate for lining the lumi- 15. Heimark RL, Twardzik DR, Schwartz SM. Inhibition of
nal surface of prosthetic vascular grafts or for transferring endothelial regeneration by type-beta transforming growth
genes into arteries than endothelial cells. factor from platelets. Science 1986; 233:1078-1080.
16. Klagsbrun M, D’Amore PA. Regulators of angiogenesis.
Concluding Remarks Annu Rev Physiol 1991; 53:217-239.
17. Asano M, Yoshida K, Tatai K. Observations of the behav-
We here described transformation of fibroblasts into
ior of microcirculation by rabbit ear chanmber technique.
endothelial cells of newly formed blood vessels based on our I. On a modification of rabbit ear chamber, development
study on angiogenesis during wound healing in the rabbit of microphotoelectric plethysmography, and rythmic fluc-
ear chamber. This gives us an another idea of the origin of tuation of microcirculation. Bull Inst Publ Health 1963;
endothelial cells of newly formed blood vessels and of the 12:34-44.
mechanism of angiogenesis than the idea which is widely 18. Alberts B, Bray D, Lewis J et al. Fibroblasts and their
believed. The transformation of fibroblasts into endothelial transformations: The connective-tissue cell family. In:
cells accounts for some puzzling phenomena on angiogen- Molecular Biology of The Cell, 3rd edition. New York and
esis and provides a benefit on clinical applications. How- London: Garland Publishing, 1994:1179-1187.
Endothelial Cells Transformed from Fibroblasts During Angiogenesis 423
19. Schneider E, Mitsui Y. The relationship between in vitro 38. Ingber D, Folkman J. Inhibition of angiogenesis through
cellular aging and in vivo human age. Proc Natl Acad Sci modulation of collagen metabolism. Lab Invest 1988;
USA 1976; 73:3584-3588. 59:44-51.
20. Kao WW-Y, Prockop DJ. Proline analogue removes fibro- 39. Piguet PF, Grau GE, Vassalli P. Subcutaneous perfusion
blasts from cultured mixed cell populations. Nature 1977; of tumor necrosis factor induces local proliferation of fi-
266:63-64. broblasts, capillaries, and epidermal cells, or massive tis-
21. McGuire PG, Orkin RW. Methods in laboratory investi- sue necrosis. Amer J Pathol 1990; 136:103-110.
gation. Isolation of rat aortic endothelial cells by primary 40. Yang EY, Moses HL. Transforming growth factor !1-in-
explant techniques and their phenotypic modulation by duced changes in cell migration, proliferation, and
defined substrata. Lab Invest 1987; 57:94-105. agiogenesis in the chicken chorioallantoic membrane. J
22. Fujita H, Nishii Y, Yamashita K et al. The uptake and Cell Biol 1990; 111:731-741.
long-term storage of India ink particles and latex beads 41. Roberts AB, Sporn MB, Assoian RK et al. Transformtin
by fibroblasts in the dermis and subcutis of mice, with growth factor type b: Rapid induction of fibrosis and an-
special regard to the non-inflammatory defense reaction giogenesis in vivo and stimulation of collagen formation
by fibroblasts. Arch Histol Cytol 1988; 51:285-294. in vitro. Proc Natl Acad Sci USA 1986; 83:4167-4171.
23. Rhodin JAG. The ultrastructure of mammalian arterioles 42. Pierce GF, Mustoe TA, Lingelbach J et al. Platelet-derived
and precapillary sphincters. J Ultrastruc Res 1967; growth factor and transformtin growth factor-b enhance
18:181-223. tissue repair activities by unique mechanisms. J Cell Biol
24. Rhodin JAG. Ultrastructure of mammalian venous capil- 1989; 109:429-440.
laries, venules and small collecting veins. J Ultrastruc Res 43. Dvorak HF, Harvey VS, Estrella P et al. Fibrin containing
1968; 25:452-500. gels indue angiogenesis. Implications for tumor stroma
25. Fujiwara T, Uehara Y. The cytoarchitecture of the wall generation and woulnd healing. Lab Invest 1987;
and the innervation pattern of the microvessels in the rat 57:673-686.
mammary gland: A scanning electron microscopic obser- 44. Brown LF, Dvorak AM, Dvarak HF. Leaky vessels, fibrin
vation. Amer J Anat 1984; 170:39-54. deposition, and fibrosis: A sequence of events common
26. Majno G. Ultrastructure of the vascular membrane. In: to solid tumors and to many other types of disease. Amer
Hamilton WF, Dow P, eds. Handbook of Physiology. Cir- Rev Respir Dis 1989; 140:1104-1107.
culation III. Washington, DC: Amer Physiol Soc, 45. Fujimoto S, Yamamoto K, Kagawa H et al.
1965:2293-2375 Neovascularization in the pre- and postnatal rabbit cor-
27. Kon K, Fujiwara T. Transformation of fibroblasts into pora cavernosa penis: Light and elctron microscopy and
endothelial cells during angiogenesis. Cell Tissue Res 1994; autoradiography. Anat Rec 1987; 218:30-39.
278:625-628. 46. Rutherford RB, Jones DN, Bergentz S-E et al. Factors af-
28. Stein J, Drenckhahn D, Nehls V. Development of peri- fecting the patency of infrainguinal bypass. J Vasc Surg
cyte-like cells during anbgiogenesis in quail chick chime- 1988; 8:236-246.
ras as detected by combined Feulgen reaction and immu- 47. Schuman ES, Gross GF, Hayes J F et al. Long-term pa-
nohistochemistry. Ann Anat 1996; 178:153-158. tency of polytetrafluoroethylene graft fistulas. Amer J Surg
29. Noden DM. Embryonic origins and assembly of blood 1988; 155:644-646.
vessels. Amer Rev Respir Dis 1989; 140:1097-1103. 48. Williams SK, Jarrell BE, Friend L et al. Adult human en-
30. Pardanaud L, Yassine F, Dieterlen-Lievre F. Relatioship dothelial cell compatibility with prosthetic graft material.
between vasculogenesis, angiogenesis and heamopoiesis J Surg Res 1985; 38:618-629.
during avian ontogeny. Development 1989; 105:473-485. 49. Wilson JM, Birinyi LK, Salomon RN et al. Implantation
31. Wilms P, Christ B, Wilting J et al. Distribution and mi- of vascular grafts lined with genetically modified endo-
gration of angiogenic cells from grafted avascular thelial cells. Science 1989; 244:1344-1346.
intraembryonic mesoderm. Anat Embryol 1991; 50. Park PK, Jarrell BE, Williams SK et al. Thrombus-free,
183:371-377. human endothelial surface in the midregion of a Dacron
32. Hara K Doi Y Nagata N et al. Role of mesenchymal cells vascular graft in the splanchnic venous circuit—Observa-
in the neovascularization of the rabbit phallus. Anat Rec tions after nin months of implantation. J Vasc Surg 1990;
1994; 238:15-22. 11:468-475.
33. Hudlická O, Tyler KR. Growth of vessels under patho- 51. Radomski JS, Jarrell BE, Pratt KJ et al. Effects of in vitro
logical conditions. In: Angiogenesis The growth of the vas- aging on human endothelial cell adherence to Dacron
cular system. London: Harcourt Brace Jovanovich, vascular graft material. J Surg Res 1989; 47:173-177.
1986:115-149. 52. Noishiki Y, Tomizawa Y, Yamane Y et al. Autocrine an-
34. Cliff WJ. Observations on healing tissue: A combined light giogenic vascular prosthesis with bone marrow transplan-
and elctron microscopic investigation. Phil Trans Roy Soc tation. Nature Medicine 1996; 2:90-93.
London Ser B 1963; 246:305-325. 53. Zwiebel JA, Freeman SM, Kantoff PW et al. High-level
35. Nehls V, Denzer K, Drenckhahn D. Pericyte involvement recombinant gene expression in rabbit endothelial cells
in capillary sprouting during angiogenesis in situ. Cell Tis- transduced by retroviral vectors. Science 1989;
sue Res 1992; 270:469-474. 243:220-222.
36. Demir R, Kaufmann P, Castellucci M et al. Fetal 54. Yao S-N, Wilson JM, Nabel EG et al. Expression of hu-
vasculogenesis and angiogenesis in human placental villi. man factor IX in rat capillary endothelial cells: Toward
Acta Anat 1989; 136:190-203. somatic gene therapy for hemophilia B. Proc Natl Acad
37. Hansen-Smith FM, Hudlicka O, Egginton S. In vivo an- Sci USA 1991; 88:8101-8105.
giogenesis in adult rat skeletal muscle: Early changes in 55. Nabel EG, Plautz G, Boyce FM et al. Recombinant gene
cappillary network architecture and ultrastructure. Cell expression in vivo within endothelial cells of the arterial
Tissue Res 1996; 286:123-136. wall. Science 1989; 244:1342-1344.
Facilitation of Healing
Transdifferentiation
CHAPTER 39
Mechanical Forces and Cell Differentiation
Ira Mills, Bauer E. Sumpio
Introduction
O ur laboratory and others have been rigorously studying the influence of mechanical forces
on vascular cell biology. It has been our contention that static conditions commonly
utilized to study vascular cells in culture may not reflect their in vivo milieu. Vascular cells in
vivo are subjected to a variety of flow-related forces including shear stress, hydrostatic pressure,
and cyclic strain1,2 (see Fig. 39.1, ref. 3). Shear stress is the tangential stress applied across the
endothelial cell surface due to the bulk flow of blood. Hydrostatic pressure is the normal
stress acting radially on the vessel wall due to the propagation of the pressure wave. Cyclic
strain represents the stress acting along the vessel wall due to circumferential deformation.
Endothelial cells are exposed to all three forces, whereas the underlying smooth muscle cells
are exposed to hydrostatic pressure and cyclic strain. There are a number of recent reviews
of the effects of shear stress on endothelial cells.3-5 Therefore, we will mention but not pro-
vide details for shear studies. We will not consider the effect of shear stress on smooth muscle
cells, since this force is considered insignificant as compared to cyclic strain.3
Cyclic strain, along with the other forces listed above, plays an important role in the
regulation of vascular tone, remodeling, and the genesis of atherosclerosis in vivo.6,7 The
mechanism by which cultured vascular endothelial cells and smooth muscle cells perceive
cyclic strain, utilizing in vitro devices to model this force, is the subject of this chapter.1,2 The
strains described for the in vitro studies herein (~10% strain) are functionally significant
based on in vivo models that replicate the major geometric features of blood vessels. These
studies report a 5-6% wall excursion at peak systole under normal conditions, which can
increase to 10% in the hypertensive state.8-10
Several recent studies in the literature suggest that the study of mechanical forces on
vascular cell biology may have fruitful applications in the field of tissue engineering of
prosthetic vascular grafts.11 In one such study, Ott and Ballerman12 found that shear stress
preconditioning of endothelial cells led to an improved tolerance (i.e., cell adherence) of
endothelialized grafts to acute shear stress. Assessment of endothelial cell tolerance was
determined by counting the number of dislodged cells, examining the grafts by light and
scanning electron microscopy and by measurement of whole blood clotting time. Shear
stress preconditioned grafts, as compared to control grafts, showed fewer numbers of
dislodged cells, the presence of an intact monolayer, and a prolonged clotting time. Thus,
this study provides evidence to suggest that preconditioning by shear stress leads to
amelioriation of the poor retention of endothelial cells, as well as the thrombogenicity of
prosthetic vascular grafts.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
426 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 39.1. Schematic showing mechanical forces of shear stress, cyclic strain, and hydrostatic pressure and associated signalling
pathways that occur in vascular cells. Adapted from ref. 3.
The mechanism responsible for the improved status In a recent study, Kraiss et al15 have provided mecha-
of the shear stress-preconditioned prosthetic grafts remains nistic data to explain the smooth muscle cell proliferation
unclear, but Ballermann and Ott11 suggest that a shift of associated with abrupt reductions in fluid shear stress. They
endothelial cells towards a more differentiated state may be found that PDGF-A gene expression in both subluminal
involved. Endothelial cells cultured under normal conditions endothelium and subadjacent smooth muscle cells is in-
tend to become dedifferentiated. In the presence of shear creased in low flow grafts. This was convincingly shown by
stress, they observed the appearance of more differentiated Northern blot analysis, in situ hybridization studies and
features, including a tighter adherence to the substratum that immunohistochemical studies. Increases in the PDGF-A
resembled in vivo endothelial cells displaying an organized transcript of 2.9-fold were noted in neointimal areas in the
cytoskeleton, Weibel-palade bodies and basal stress fibers presence of reduced shear stress. Even greater changes were
with focal adhesion plaques. indicated by in situ studies, presumably due to the predomi-
Clowes and colleagues13-15 have characterized the ef- nance of upregulated message in the luminal surface of the
fects of blood flow and shear stress on subendothelial smooth neointima. This study emphasizes the need to delineate the
muscle cell proliferation in baboon prosthetic grafts. They responsiveness of vascular cells to mechanical perturbation
have found that neointimal hyperplasia in endothelialized to better understand their behavior in situations such as tis-
grafts is inhibited by increased blood flow. In the presence sue engineering of vascular grafts.
of a distal arteriovenous fistula that caused nearly 3-fold The complexity of the nature of the forces perceived
elevations in flow and shear stress, smooth muscle cell by grafts was illustrated by Dobrin et al,16 who subjected
number was reduced with a consequent reduction in the autogenous vein grafts to a variety of conditions of pressure
cross-sectional area of the neointima at 3 months. More- and flow including three static deformations, three static
over, ligation of the fistula after 2 months caused a rapid stresses, increased pulsatile deformations, pulsatile stresses,
increase in neointimal thickness,13 showing a strong asso- and altered shear stress at the blood-intima interface. Three
ciation between blood flow and the smooth muscle cell re- sequential experiments were performed to sequentially
sponse in vascular grafts. determine the role of nine factors. In agreement with Clowes’
work,13-15 low flow velocity was associated with intimal hy-
Mechanical Forces and Cell Differentiation 427
perplasia. Moreover, medial thickening of autogenous vein tive of a dedifferentiated state occur, such as elevated prolif-
grafts was observed with increased deformation of the vein eration.38 For example, recent work by Reusch et al39 sug-
wall in the circumferential direction, which mimics gests that smooth muscle cells become more differentiated
clinical findings. upon cyclic strain as evidenced by the upregulation of myo-
Over the years, we have acquired a large body of data sin heavy chain expression. As described in greater detail
to suggest that the study of vascular cells challenged with below, other responses of smooth muscle cells to cyclic strain
mechanical stimuli is indeed a more appropriate method portend a dedifferentiated state such as enhanced prolifera-
for studying their biology.17-32 This chapter represents an tion.24,40 It is clear that much work needs to be done in char-
update of an earlier review of the literature describing the acterizing the modulation of cyclic strain on vascular cell
effect of cyclic strain on vascular cell biology.22 Data obtained biology before any mechanistic significance can be ascribed
from our laboratory and that of others have begun to delin- to its role in advancing tissue engineering of prosthetic grafts.
eate the signaling pathways that may be involving in affect- As recently reviewed by Zilla et al,41 “manipulation of
ing vascular cell phenotype and is commented on in great the endothelium might provide the next major advancement
detail in this review. We also discuss the recent discovery of for therapeutic and preventive measures for cardiovascular
strain-sensitive genes that appear to be finely tuned in their disease”. It is with this objective that the following studies
ability to distinguish mechanical perturbations based on are outlined that characterize the mechanical manipulation
unique strain sensitive or shear sensitive cis-elements in their of both endothelial cells and smooth muscle cells. It is hoped
promoter regions that respond to unique strain-activated that a greater delineation of the mechanisms involved in the
or shear-activated transcriptional factors that either induce strain-induced modulation of vascular cell phenotype will
or repress gene induction. lead to better diagnosis and treatment of vascular disease
Recent studies suggest that the state of differentiation and the implementation of improvements in tissue engineer-
of vascular cells in culture is fluid,33-35 and a novel stimulus ing of prosthetic grafts.
for controlling the phenotypic state of these cells is mechani-
cal strain.36 As described below, recent studies suggest that Effect of Cyclic Strain
cyclic strain may direct endothelial cells to a more differen- on Endothelial Cell Biology
tiated state as evidenced by phosphorylation of focal adhe-
sion proteins.37 However, the state of differentiation of en- General Phenotype
dothelial cells as well as smooth muscle cells in response to Previous studies in our and others’ laboratories have
cyclic strain is complex, since many other responses indica- shown that cyclic strain can alter the general phenotype of
Mechanical
Stress
Ion Channels
Adenylyl/Guanylyl
Cyclase
ECM & Surface Proteins
Phospholipids Cell
Response
Gene
Cell-cell Expression
Proteins
Linker Proteins
alpha-actinin, talin,
vinculin, etc.
Cytoskeleton
Fig. 39.2. Schematic showing mechanical force transmission and transduction pathways in vascular cells. Adapted from ref. 47.
428 Tissue Engineering of Prosthetic Vascular Grafts
Table 39.1. Effect of cyclic strain and shear stress on endothelial cell phenotype. (Adapted from ref. 3)
endothelial cells, including their proliferative rate,27,38 mor- analysis, they found a remodeling of focal adhesion sites in
phology18,42,43 and the secretion of macromolecules such as the direction of flow. In addition to this response, shear stress
prostacyclin,29 endothelin,30 nitric oxide,44,45 tissue plasmi- caused a redistribution of intracellular stress fibers, align-
nogen activator20 and plasminogen activator inhibitor-1 ment of individual focal adhesion sites, and the coalescing
(PAI-1)46 (see Fig. 39.2, see ref. 47). of smaller sites to larger, and fewer, focal adhesions per cell.
Changes in endothelial cell phenotype are also modu- Yano et al50 recently studied the involvement of dif-
lated by shear stress (see Table 39.1). Changes include alter- ferent integrins in signaling induced by cyclic strain, since
ations in proliferation, morphology and the synthesis and integrins have been localized to focal adhesion sites. Fur-
secretion of many of the same proteins influenced by cyclic thermore, these focal adhesion sites are known to exhibit
strain.3-5 However, the nature of the changed phenotype can pp125FAK phosphorylation by cyclic strain as well as by
be markedly different, whether the perturbation is either integrins. Cyclic strain of 4 h led to a redistribution of # and
50
shear stress or cyclic strain. For example, the orientation of ! integrins in human umbilical vein endothelial cells. In
endothelial cells in response to shear stress is parallel to the addition, !1 integrin reorganized in a linear pattern parallel
direction of flow.48 In contrast, cyclic strain causes a per- with the long axis of the elongated cells, creating a fusion of
pendicular alignment of endothelial cells in relationship to focal adhesion plaques in cells plated on fibronectin (a ligand
the strain vector.18 for #5!1) or collagen (a ligand for #2!1) coated plates. In con-
More recent studies performed in our laboratory sup- trast, the vitronectin receptor, !3 integrin, failed to redis-
port the involvement of focal adhesion plaques in the trans- tribute in endothelial cells subjected to cyclic strain. Cyclic
mission of cyclic strain to the cell. Yano et al37 demonstrated strain also caused a reorganization of #5 and #2 integrins in
phosphorylation of cytoskeletal proteins that reside at the a linear pattern on cells seeded on fibronectin and collagen-
cytoplasmic face of the focal adhesion plaques, namely coated plates, respectively. However, the expression of #5,
pp125FAK and paxillin. The strain-induced phosphorylation #2, and !1 was not influenced by 24 h of cyclic strain as as-
of these proteins occurs on tyrosine residues and can be in- sessed by immunoprecipitation of these integrins. Strain-
hibited by specific tyrosine kinase inhibitors. The tyrosine induced phosphorylation of pp125FAK occurred concomi-
phosphorylation of pp125FAK occurs earlier and is more ro- tantly with the reorganization of !1 integrin. Thus, #5!1 and
bust than that observed with paxillin. However, after a four #2!1 integrins may play an important role in transducing
hour exposure to strain, both cytoskeletal proteins were mechanical stimuli into intracellular signals.50
found to align as shown by confocal microscopy.37 This ob- Thoumine et al51 demonstrated effects on the organi-
servation was in sharp contrast to their random distribu- zation and composition of the extracellular matrix upon
tion found in stationary controls. Tyrphostin A25, a tyrosine subjecting endothelial cells to shear stress. They examined
kinase inhibitor, is blocked by the strain-induced phospho- the pattern and levels of various extracellular matrix pro-
rylation of pp125FAK and paxillin as well as by their strain- teins including fibronectin, laminin, type IV collagen, and
induced alignment. Moreover, the importance of focal ad- vitronectin. Of these, fibronectin was found to be the most
hesion proteins on the mechanically induced gross morpho- affected by shear stress, with a thickening and alignment of
logical changes of endothelial cells was implied by the abil- its fibril tracts. Moreover, the level of fibronectin increased
ity of tyrphostin A25 to reverse both cellular alignment and after 1 or 2 days of shear stress after an earlier reduction at
migration caused by strain. 12 h. The extracellular matrix response to shear was found
Davies et al49 have studied the effect of shear stress on to be specific for fibronectin, since both laminin and type
focal adhesion sites in bovine aortic endothelial cells. By tan- IV collagen showed thickening of fibrils, but no alignment.
dem scanning confocal microscopy and digitized image Vitronectin was unaffected in any respect.
Mechanical Forces and Cell Differentiation 429
Table 39.2A. Effect of cyclic strain on second messenger pathways in endothelial cells. (Adapted from ref. 3)
(average 10% strain) vacuum at 60 cycles per minute (0.5 sec lished a role of G proteins in the responsiveness of endothe-
strain; 0.5 sec relaxation) for 15 minutes, there was a 1.5- to lial cells to shear stress. Both shear stress mediated
2.2-fold increase in adenylyl cyclase, cAMP and PKA activ- prostacyclin and nitric oxide production were found to be
ity as compared to unstretched controls. The strain-induced mediated by pertussis-toxin sensitive and insensitive G pro-
activation of this pathway appears to occur by exceeding a teins, respectively.70,71
strain threshold, since no change in adenylyl cyclase, cyclic
AMP or PKA was observed in cells subjected to 37.5 mm Hg Protein Phosphatase Activity
(average 6% strain). Further studies demonstrated an in- As described above, we have reported previously that
crease in cAMP response element activity as shown by gel PKC in the membrane fraction of endothelial cell lysates is
shift analysis. Thus, cyclic strain may stimulate the expres- activated in response to cyclic strain.72 To better understand
sion of genes containing cAMP-responsive elements. cellular responses that are dependent on the phosphorylated
We next tested the hypothesis that cyclic strain modu- state of proteins, it is also important to study the role of
lates G protein function, since G proteins are intimately in- protein phosphatases. In a recent study,73 we examined the
volved in the activation of adenylyl cyclase (Mills, unpub- effect of cyclic strain on protein phosphatase-1 and -2A ac-
lished observations). To test this hypothesis, we examined tivity in bovine aortic endothelial cells. Protein phosphatase-
the effect of acute cyclic strain on the immunoreactivity of 2A activity in the cytosol was decreased by 36.1% in response
the alpha subunits of the heterotrimeric G proteins, Gs and to 60 minutes of cyclic strain, whereas activity in the mem-
Gi, that promote stimulation and inhibition of AC activity, brane was unchanged. Furthermore, treatment with a low
respectively. We observed a transient decrease in the immu- concentration of okadaic acid (0.1 nM) enhanced prolifera-
noreactivity of the inhibitory G protein alpha subunits tion of both static and stretched endothelial cells in 10%
(Gi#1,2).In contrast, immunoreactivity of Gs in bovine aor- fetal bovine serum. These data suggest that protein phos-
tic endothelial cells and Gi#1,2 and Gs in bovine aortic phatase-2A acts as a growth suppresser, and cyclic strain may
smooth muscle cells were unaffected by cyclic strain. enhance cellular proliferation by inhibiting protein phos-
Frangos and colleagues have shown shear stress acti- phatase-2A.73
vation of G proteins, particularly G#q/#11 and G#i3/#o.69
This was determined by labelling of flow-stimulated G pro- Transcriptional Factor Activation (See Table 39.3B)
teins with a nonhydrolyzable GTP photoreactive analog. The In a recent study, Cheng et al46 demonstrated strain-
rapidity by which this response is obtained (1 sec) suggests induced activation of PAI-1 secretion. Although this effect
a key role of G proteins in the mechanotransduction of shear could be accomplished directly on the secretory process, the
stimuli. Previous studies by the same group further estab- authors postulate the upregulation of the PAI-1 gene via a
Table 39.3A. Effect of cyclic strain on transcription factor activation in endothelial cells. (Adapted from ref. 3)
Table 39.3B. Effect of shear stress on transcription factor activation in endothelial cells. (Adapted from ref. 3)
functional AP-1 binding site in its promoter regions. Inter- Selective seeding of the endothelial cells to the high strain
estingly, the strain-induced secretion of PAI-1 was shown region (7-24% strain) led to a greater response than found
to involve reactive oxygen species, a known activator of both in those cells grown in the central, low strain region (0-7%).
AP-1 and NF-∀B transcription factors. Further studies will An increase in CRE binding activity was not observed in
be required to test this hypothesis. However, studies per- cells subjected to an average 6% strain. The reasons for ob-
formed in our laboratory support the hypothesis of increased servation of a strain response in stimulation of CRE bind-
activity of transcriptional activators in endothelial cells sub- ing activity of bovine aortic endothelial cells in this study,68
jected to cyclic strain.78 but not the earlier one,78 is unclear and may suggest other
Since previous studies demonstrated that cyclic strain variables aside from species and vascular bed diversity that
stimulates protein kinase c activity in endothelial cells,72 we remain to be uncovered. The nature of the mechanical force
tested the hypothesis that downstream induction of the fos may also be critical in activating a particular transcription
and jun genes and the transcription factor activator protein-1 factor. For example, Lan et al79 and Resnick et al80 have both
(AP-1) were also activated by strain. Sumpio et al31 showed shown NF-∀B activation in bovine aortic endothelial cells
that human umbilical vein endothelial cells subjected to cy- subjected to shear stress; a finding we have not been able to
clic strain for as short a duration as 30 minutes leads to the replicate in response to cyclic strain.
induction of c-fos, fosB and c-jun genes. In contrast, junB
and junD were not altered by cyclic strain under identical Gene Expression
conditions. We did detect an increase in AP-1 binding activ-
ity as measured by EMSA conducted after 2 h of cyclic strain. Endothelin
It may be concluded that cyclic strain may be coupled to the Cyclic strain stimulates endothelin-1 secretion and
endothelial cell response via activation of protein kinase c mRNA levels in human umbilical vein endothelial cells.82
and elevated steady state levels of different Fos and Jun prod- Elevated expression of endothelin-1 mRNA was detected in
ucts, which may enhance the activity of the transcriptional cells subjected to 2 h or longer duration of cyclic strain. This
activator AP-1.31 was abolished by treatment with actinomycin D. The strain-
In more rigorous studies performed to characterize induced elevation of ET-1 mRNA synthesis was found to be
the induction of transcriptional factor activation, AP-1, CRE mediated by the activation of the PKC pathway and was
binding protein and NF-∀B were examined in endothelial shown to require extracellular Ca2+. Involvement of the PKC
cells obtained from a variety of beds and species.78 These pathway was determined since the PKC inhibitor calphostin
include endothelial cells obtained from human umbilical C prevented strain-induced entothelin-1 expression. In con-
vein, human aorta and bovine aorta. In human umbilical trast, the cAMP-dependent protein kinase inhibitors KT5720
vein endothelial cells subjected to cyclic strain, AP-1 activity or KT5823 only partially blocked endothelin-1 expression
was elevated as compared to unstretched controls.31 The stimulation by cyclic strain. A requirement of extracellular
onset of strain-induced AP-1 activity was at 2 h, peaked by Ca 2+ was determined by blockade of strain-induced
4 h, and returned to baseline levels by 24 h. Similar findings endothelin-1 mRNA by EGTA as well as a lesser effect by
were obtained in human aortic endothelial cells but not in BAPTA/AM, an intracellular calcium chelator.82 In our
bovine aortic endothelial cells.78 A more pronounced retar- hands, we have observed strain-dependent reduction in ET-1
dation indicative of a supershift was noted in the presence (Koo J et al, unpublished data).
of antibodies to c-Jun in HUVECs. Similar contradictory effects of shear stress have also
CRE binding activity was also influenced by cyclic been detected in measurements of ET-1.61,62,83 Malek et al83
strain in a species and vascular bed-dependent manner.78 have shown shear stress-mediated downregulation of ET-1
CRE levels were significantly stimulated in human umbili- expression in bovine aortic endothelial cells conferred by a
cal vein endothelial cells in a biphasic manner with a 2-fold cis-element between -2.5 kb and -2.9 kb of the 5'-upstream
increase at 15 minutes and a nearly 5-fold increase at 24 h. promoter region that does not involve the AP-1 or GATA-2
Similar trends and kinetics were found in endothelial cells factor binding site. In contrast, Morita et al61,84 have shown
derived from bovine aorta. Similar to what is described above shear stress-induced ET-1 expression in porcine aortic en-
for AP-1, we failed to detect a change in CRE binding activ- dothelial cells that appears to be related to cytoskeletal
ity in this study.78 disruption.
NF-∀B binding activity in response to cyclic strain was
also dependent on the vascular bed and species under study. Prostacyclin
NF-∀B binding activity was stimulated in a monophasic Previous studies indicate that cyclic strain29 and shear
58,85
manner in endothelial cells obtained from umbilical vein stress can increase PGI2 secretion by endothelial cells,
with a 4.6-fold increase at 4 h. As observed for AP-1, NF-∀B but the effect of these forces on prostacyclin synthase (PGIS)
was stimulated earlier by 2 h in human aortic endothelial gene expression remains unclear. In a recent study, we ex-
cells, with a peak observed at 4 h. NF-∀B binding activity, as amined this question by studying PGIS gene expression by
shown above for AP-1 and CRE, was not altered by cyclic Northern blot analysis and protein level by Western blot
strain in bovine aortic endothelial cells in this study.78 analysis.57 In addition, the effect of cyclic strain on the PGIS
However, in a later study we did show evidence for promoter was determined by the transfection of a 1 kb hu-
strain-induced stimulation of CRE binding activity in bo- man PGIS gene promoter construct coupled to a luciferase
vine aortic endothelial cells.68 In this study, CRE binding reporter gene into EC, followed by determination of lu-
activity was found by 30 minutes and diminished by 4 h. ciferase activity.
432 Tissue Engineering of Prosthetic Vascular Grafts
PGIS gene expression increased 1.7-fold in EC sub- Awolesi et al 44 showed that the strain-induced
jected to cyclic strain for 24 h.57 Likewise, EC transfected upregulation of eNOS expression in bovine aortic endothe-
with a pGL3B-PGIS(-1070/-10) construct showed an ap- lial cells translates into an increase in functional activity. In
proximate 1.3-fold elevation in luciferase activity in EC sub- this study, 10% cyclic strain for 24 h led to an increase in
jected to cyclic strain for 2, 4, 8 and 12 h. The weak stimula- both citrulline production and accumulated nitrite (Greiss
tion of PGIS gene expression by cyclic strain was reflected reaction) as indices of eNOS activity. Specificity of this re-
in an inability to detect alterations in PGIS protein levels in sponse was confirmed by blockade with EDTA and L-NAME.
EC subjected to cyclic strain for as long as 5 days. These data As is the case for strain-induced expression, strain-induced
suggest that strain-induced stimulation of PGIS gene expres- eNOS functional activity was more pronounced at 10% av-
sion plays only a minor role in the ability of cyclic strain to erage strain as compared to 6% strain. The effect of 10%
stimulate PGI2 release in EC. These findings, coupled with strain was as potent as that observed with the calcium iono-
our earlier demonstration of a requisite addition of exog- phore, A23187.44
enous arachidonate in order to observe strain-induced PGI2 Shear stress has also been shown to increase nitric
release,29 implicates a mechanism that more likely involves oxide gene expression in endothelial cells.59,86,87 In a recent
strain-induced stimulation of PGIS activity. study, Uematsu et al59 reported 2 to 3-fold elevations in eNOS
by shear stress of 15 dynes/cm2 for up to 1 day. The involve-
Tissue Plasminogen Activator (tPA) ment of K+ channels was suggested, since shear stress in-
Previous studies showed an increase in tPA mRNA, duction of nitric oxide mRNA was blocked by tetraethylam-
immunoreactive tPA protein and tPA activity in the medium monium chloride, a K+ channel antagonist.
of cultured bovine aortic endothelial cells exposed to 10%
average strain.19,20 Previous studies documented that tPA MCP-1
expression is upregulated by shear stress.60 We have more Wang et al88 studied the effect of cyclic strain on mono-
recently examined the regulation of tPA gene expression in cyte chemotactic protein-1 (MCP-1) expression in cultured
endothelial cells by cyclic strain.32 A functional analysis of human umbilical vein endothelial cells (HUVECs). They
the tPA promoter was performed by transfecting bovine found a two-fold enhancement in MCP-1 expression as early
aortic endothelial cells with a 1.4 kb construct of the hu- as one hour after the initiation of strain. This response was
man tPA promoter coupled to CAT. After 4 h of cyclic strain, maintained for at least 24 h in the presence of strain, but
a nearly 3-fold increase in the activity of the 1.4 kb tPA pro- was restored to baseline levels 3 h after its release. Calphostin
moter was detected.32 A 60% drop off in activity was found C was able to abolish strain-induced MCP-1 expression,
between position -145 and -105 by analysis of deletion mu- implicating a role of PKC as a mediator of this response. In
tants. DNase I protection analysis of the segment down- addition, a strong Ca2+ requirement for strain-induced, as
stream of position -196 suggested involvement of AP-2 or well as basal, expression of MCP-1 was determined by block-
CRE-like regions, which was confirmed by EMSA analysis. ade with either EGTA pretreatment, BAPTA/AM chelation,
Site directed mutants of either the AP-2 or CRE-like regions or verapamil addition to HUVECs.88
resulted in a 65% decrease in activity compared to the wild Shear stress-induced expression of MCP-1 has been
type. In addition, double mutations abolished basal tran- shown to involve a cis-acting TRE-responsive element in its
scription and any strain-induced activity. A SSRE binding promotor.89 A construct of multiple copies of the TRE-re-
site was found to be present at -945, but site directed mu- sponsive element coupled to a prolactin minimal reporter
tants failed to show any drop in strain-induced activity as and luciferase gene was found to be sufficient to confer shear
compared to the wild type. Overall, our studies demonstrate sensitivity in transfected bovine aortic endothelial cells.
that cyclic strain regulates tPA gene transcription in bovine
aortic endothelial cells by a mechanism similar to that shown ICAM/VCAM
previously for phorbol ester. In recent studies conducted in our laboratory, we have
obtained evidence to suggest that ICAM and VCAM expres-
Nitric Oxide sion are downregulated by cyclic strain in human umbilical
Awolesi et al45 demonstrated that cyclic strain can vein endothelial cells (Lee et al, unpublished observations).
upregulate the expression of endothelial nitric oxide syn- In contrast ICAM-1 expression is upregulated in the same
thase (eNOS) in bovine aortic endothelial cells. At an aver- cell type under conditions of shear stress.87,90
age strain of 10%, eNOS expression in these cells was shown
to be increased as compared to unstretched controls as de- Effect of Cyclic Strain on Vascular Smooth
termined by Northern blot analysis. A milder elevation in Muscle Cell Biology
eNOS expression was measured in cells exposed to a lesser
degree of strain of 6%. The strain-induced activation of General Phenotype
eNOS expression was attributed to stimulation of transcrip- Smooth muscle cells are known to exist in two classic
tional activity as shown by nuclear runoff assays. Western phenotypic states.26 In vivo, under normal conditions,
blot analysis and immunochemistry confirmed that eNOS smooth muscle cell proliferation is slow and the contractile
protein levels were similarly increased in response to strain; or differentiated phenotype predominates. The differenti-
again 10% strain led to a heightened response as compared ated phenotype is characterized by a heterochromatic
to 6% strain. nucleus, abundant actin and myosin filaments. The contrac-
Mechanical Forces and Cell Differentiation 433
tile smooth muscle cells contract in response to both chemi- is mediating opposing activities; PDGF secreted in response
cal and mechanical stimulation. In vivo counterparts of the to cyclic strain opposes the effect of strain to upregulate
contractile phenotype are smooth muscle cells from media smooth muscle heavy chain isoforms.
of vessels of normal adults. The findings of Reusch et al39 also confirm the earlier
However, the smooth muscle cell is not terminally dif- work of Birukov et al95 who showed a dual effect of cyclic
ferentiated and is able to undergo phenotypic modulation strain in modulating rabbit aortic smooth muscle cell phe-
under a variety of circumstances. This phenotypic modula- notype. Cyclic strain was found to both increase prolifera-
tion occurs upon vessel injury and is characterized by a re- tion and also increase expression of markers of smooth
duction in smooth muscle contractile proteins such as #- muscle cell differentiation, in this case h-caldesmon. Com-
actin and smooth muscle myosin heavy chain, and by a shift mon attributes of these studies included a dependence on
toward a synthetic state accompanied by the laying down of quiescent conditions to observe many of their effects (i.e.,
extracellular matrix molecules.35 Such changes can be mim- strain-induced myosin heavy chain expression and prolif-
icked in vitro by growing smooth muscle cells in a serum- eration ), as well as modulation by extracellular matrix pro-
fed preconfluent condition. Morphological characteristics teins that served as a substratum for the smooth muscle cells.
of the synthetic smooth muscle cell phenotype include a In the study by Birukov et al,95 smooth muscle myosin heavy
euchromatic nucleus as well as a prominent endoplasmic chain was increased in the presence of 0.5% serum, but not
reticulum and Golgi complex. These smooth muscle cells observed at concentrations of 2, 5 and 10%. However, strain-
resemble those found in diffuse intimal thickenings of ath- induced h-caldesmon expression was less dependent on se-
erosclerotic lesions.91 rum levels. Laminin was a preferred substrate for detection
However, the identification of smooth muscle cells as of strain-induced h-caldesmon levels95 and was also a pref-
belonging to either a contractile or synthetic phenotype may erable substrate, along with type I collagen, in promoting
be a gross simplification.92,93 Rather, the behavior of smooth differentiation of smooth muscle cells in the study by Reusch
muscle cells is not locked into a particular phenotype such et al.39
that those classified as belonging to the “synthetic” pheno- The authors also studied the effect of extracellular
type are not able to contract and vice versa. For example, matrix molecules on strain-induced SM-1 expression,39 since
Chamley and Campbell94 found that single smooth muscle earlier studies indicated its modulation of strain responsive-
cells from newborn guinea pig vas deferens underwent mi- ness.96 In this study, matrix was again found to be an im-
tosis (as observed by time-lapse photography) despite the portant factor in how SMCs transduce the strain stimuli.
fact that the cells were in a morphologically-defined differ- The ability of cyclic strain to increase SM-1 was 2-fold in
entiated state. This finding emphasized the need to examine SMCs grown on type I collagen, 3-fold on laminin, and ab-
biochemical parameters, in addition to morphology, in or- sent in fibronectin-coated wells. It was concluded that me-
der to better define the phenotypic state of smooth muscle chanical strain can alter myosin isoform expression to a
cells in culture.93 more differentiated state in a matrix-dependent manner.96
As described recently by Owens,36 the factors that nor- The findings of Reusch et al39 confirmed earlier ob-
mally control the differentiation of the smooth muscle cell servations of Kanda and Matsuda97 that indicated stress-in-
are not understood. However, a recent paper by Reusch et duced differentiation of bovine aortic smooth muscle cells.
al39 is lauded as providing the most definitive study to date In their study, Kanda examined smooth muscle cells cul-
identifying a positive differentiation influence/factor in tured in a three dimensional type I collagen gel. Smooth
smooth muscle cells. The work by Reusch et al39 describes muscle cells were exposed to 3 conditions including isotonic
cyclic strain as the factor. Notably, the story as described control, isometric stress or static stress and dynamic stress
below is most complex, as strain has been shown to not only consisting of periodic stretching and recoiling of 5% above
upregulate markers of differentiation (i.e., SM-1, SM-2) but and below the resting tissue length at 60 cycles per minute.
also to stimulate smooth muscle cell growth. Whether this After 4 weeks of this regimen, smooth muscle cell morphol-
represents an artifact of the system due to its heterogeneous ogy was examined by light microscopy and transmission
strain gradient, is due to distinct subpopulations of smooth electron microscopy. Smooth muscle cells subjected to the
muscle cells, or reflects the fact that growth and differentia- dynamic regimen were unique in exhibiting a more differ-
tion are not necessarily exclusive properties remains to be entiated state as shown by increased fractions of contractile
established.36 apparatus. Over time on stress, the content of myofilaments,
Since previous studies had shown that PDGF secre- dense bodies and extracellular filamentous basement mem-
tion is elevated by cyclic strain,40 the authors examined brane-like materials increased. This was an indication that
whether secreted PDGF affects the strain-induced changes dynamic stress in cells laid down in a 3-dimensional collagen
in contractile protein expression. Interestingly, PDGF-AB matrix inclines them toward a more differentiated state.97
was shown to cause a qualitatively opposite change in myo- The effect of cyclic strain on smooth muscle cell pro-
sin isoform expression, decreasing smooth muscle myosin liferation is predominantly stimulatory40,95,98,99 Davis99 ob-
by greater than half. Moreover, neutralization of PDGF-AB served strain-induced cell proliferation of 33% in serum-
with neutralizing antibodies led to a strain-induced increase starved rat aortic smooth muscle cells supplemented with
in SM-1 that was enhanced 10-fold. Thus, PDGF fails to insulin and transferrin. Although significant, this response
mediate the effect of cyclic strain on smooth muscle myosin was only one fifth that observed in the presence of repleted
isoform distribution. Instead, it is concluded that cyclic strain calf serum of 10%. Likewise, Yang et al98 showed a significant
434 Tissue Engineering of Prosthetic Vascular Grafts
increase, albeit slight (13%), by 6 days of cyclic strain expo- muscle cell proliferation, depending on the exact source of
sure in cultured smooth muscle cells obtained from the tissue and the regimen under study. For example, Sumpio
media of human left descending coronary artery. In neona- and Banes28 showed that the proliferative response of por-
tal rat smooth muscle cells grown to quiescence, Wilson et cine aortic smooth muscle cells is inhibited by cyclic strain.
al40 observed a similar 40% increase in the proliferative re- In another study using early passaged smooth muscle cells
sponse to cyclic strain as compared to static controls. In rab- from lamb pulmonary artery, Kulik and Alvarado100 failed
bit aortic smooth muscle cells, cyclic strain also caused an to detect any increase in cell number after 24 h of 20% stretch.
increase in cell number.95 Interestingly, the ability to detect Similarly, DNA synthesis as measured by [3H]thymidine
an increase in cell proliferation induced by cyclic strain was incorporation was unaffected by stretch. In the same study,
dependent on the cells being serum activated. In quiescent a passaged clonal line of rat cells (PAC1) did show a modest
cultures, smooth muscle cells failed to respond to cyclic elevation (17% increase) in the percentage of
strain. In this study, Birukov et al95 also showed that the [3H]thymidine-labeled cells. However, TCA-precipitated
number of detached cells (i.e., an index of cell attachment [3H]thymidine was unaffected by cyclic strain.
to membranes) was unaffected by cyclic strain. In another In a follow up study to the earlier work of Kulik and
control experiment, motion effects of circulating medium Alvarado100 failing to demonstrate an effect of mechanical
in response to cyclic strain was ruled out as a contributory forces on the growth of smooth muscle cells from pulmo-
factor to the stretch-induced increase in smooth muscle cell nary artery, Kolpakov et al101 reexamined this finding in a
proliferation, since no difference was noted in control ver- whole vessel model. Interestingly, they found that subject-
sus nutational conditions. ing rabbit pulmonary arteries to varying magnitudes of
The effect of cyclic strain on DNA synthesis in smooth stretch led to an increased rate of protein synthesis (as mea-
muscle cells is more pronounced than for proliferation. In sured by quantitative autoradiography) and replication of
coronary artery smooth muscle cells obtained from human smooth muscle cells. Moreover, synthesis of extracellular
left descending coronary arteries, Yang et al98 found that matrix proteins, elastin and collagen, were elevated in 4 day
cyclic strain stimulates DNA synthesis greater than 2-fold stretched segments as compared to static controls. The au-
after 24 h. In rat aortic smooth muscle cells, Davis et al99 thors also showed that these effects were independent of
showed a 2-fold increase in [3H]thymidine incorporation endothelium interaction, as its removal did not prevent the
by cyclic strain. In smooth muscle cells obtained from new- aforementioned changes.
born rat, Wilson et al40 demonstrated a nearly 3-fold increase This study illustrates the caution that should be exer-
in [3H]thymidine incorporation by cyclic strain of -20 kPa cised in interpretation of in vitro experiments that may not
at 60 cycles/min for 48 h. This elevation afforded by strain be analogous to the condition in vivo or even in an ex vivo
was less potent as compared to that observed with 1 U/ml model. The authors rightly assert that their earlier negative
thrombin or 5 ng/ml platelet derived growth factor (PDGF). findings of in vitro studies could be attributed to cell shape,
Further study showed that combinatorial addition of strain its orientation relative to the applied strain, matrix substra-
and thrombin led to even greater stimulation of DNA syn- tum, and of course, the cell culture milieu.101
thesis. However, strain and PDGF did not exhibit signifi-
cantly greater stimulation of [3H]thymidine as compared Cell Signaling
to that observed with PDGF alone. The authors suggest that
this observation may be explained by strain itself acting via Second Messenger Pathways
autocrine production of PDGF.40
Wilson et al96 further studied the matrix dependence Adenylyl Cyclase
of the strain-induced mitogenic response in rat vascular In bovine aortic smooth muscle cells, we have dem-
smooth muscle cells. They found that cyclic strain increased onstrated strain-induced activation of the adenylyl cyclase/
DNA synthesis in SMCs grown not only on type I collagen, cyclic AMP/protein kinase a pathway.24 Basal adenylyl cy-
but also fibronectin or vitronectin. However, on plates coated clase was elevated nearly 2-fold in SMC subjected to
with elastin or laminin, strain induction of DNA synthesis 150 mm Hg vacuum deformation for 30 min and returned
was not observed. The integrin binding peptide GRGDTP to basal levels by 60 min. Likewise, cyclic AMP accumula-
was shown to block the mitogenic response of SMCs to strain tion was also stimulated by cyclic strain at 30 min and re-
in cells seeded on type I collagen. Specificity of this response turned to basal values by 60 min. Protein kinase a activity
was noted by the inability of the inactive peptide GRGESP was also stimulated by cyclic strain. The activation of this
to also block strain-induced mitogenesis. In addition, pathway was strain specific, such that cells subjected to low
strain-induced expression of a PDGF-A chain promoter- strain (0-7%) in the center of the well were found to signifi-
CAT construct transiently transfected into SMCs was blocked cantly stimulate PKA activity, as compared to cells in the
by the integrin binding protein GRGDTP. A role for specific peripheral, high strain (7-24%) region of the well. Similar
integrins involved in sensing mechanical strain in SMCs was findings of an elevated response from cells obtained from
shown by studies with antibodies to both !3 and #v!5 the center region were observed for CRE binding protein
integrins, which also blocked the mitogenic response levels as measured by gel shift analysis.
to strain. The relationship between strain-activated signaling
However, there are conflicting reports in the litera- events and downstream responses is complex. This is exem-
ture regarding the effect of mechanical strain on smooth plified by our inability to demonstrate any effect of
Mechanical Forces and Cell Differentiation 435
Rp-cAMP, a PKA inhibitor, on strain-induced proliferation Our data suggested that cyclic strain causes dephosphoryla-
or alignment.24 Thus, although strain activated the adenylyl tion of MLC20 in SMCs, which may be partially due to the
cyclase signaling pathway, the lack of an effect of PKA inhi- activation of MLC20 phosphatase and/or inhibition of
bition suggests either the lack of involvement of this path- MLC20 phosphorylation.
way or the multifactorial nature of these responses.
Gene Expression
Tyrosine Kinase
Davis et al99 showed that cyclic strain stimulates the PDGF
phosphotyrosine accumulation in smooth muscle cells ob- PDGF-AA and -BB secretion and PDGF-A expression
tained from rat aorta. Proteins with upregulated were stimulated in smooth muscle cells subjected to cyclic
phosphotyrosine content in response to cyclic strain included strain.40 Moreover, neutralizing antibodies to both PDGF-
species of 110-130 kDa and 70-80 kDa as determined by AA and -AB were found to block strain-induced stimula-
Western blot analysis. The identity of these proteins was not tion of DNA synthesis. In contrast, bFGF antibody failed to
ascertained, but ascribed to proteins of corresponding mo- prevent the mitogenic response to cyclic strain.40
lecular weight, including GTPase and p74Raf, respectively.
Specificity of tyrosine kinase activity induced by cyclic strain Myosin Heavy and Light Chain
was shown by blockade with the phosphotyrosine kinase Reusch et al39 recently examined the effect of mechani-
inhibitor herbimycin A. Moreover, both herbimycin A and cal strain on the expression of myosin heavy chain isoform
genistein, another tyrosine kinase inhibitor, were found to expression in neonatal rat vascular smooth muscle cells. As
have cyclic strain-induced DNA synthesis as measured by a marker of differentiation, they studied expression of the
incorporation of [3H]thymidine. Thus, it was concluded that smooth muscle myosin heavy chain isoforms, SM-1 and
cyclic strain stimulates vascular smooth muscle cell prolif- SM-2 (see Fig. 39.7 in ref. 39). These isoforms are well char-
eration through activation of tyrosine kinases.99 acterized markers of differentiation expressed at high levels
in postconfluent, quiescent SMC.102 In quiescent smooth
Phosphatases muscle cells grown in 0.5% fetal bovine serum, SM-1 and
Phosphorylation of the 20 kDa regulatory myosin light SM-2 protein content was elevated by 4.5- and 3.5-fold,
chain (MLC20) is the seminal event in the initiation of vas- respectively, after 72 h of cyclic strain. In addition, nonmuscle
cular smooth muscle cell contraction. We sought to deter- myosin A (NM-A) protein content decreased to 30% of con-
mine whether cyclic strain affects MLC20 phosphorylation trol levels. Cyclic strain was also found to increase gene ex-
in bovine aortic smooth muscle cells. We studied the effect pression of SM-1 and decrease that of NM-A, with peak
of cyclic strain on serum-fed SMCs already displaying a high changes occurring at 12 h and 3 h, respectively.
level of MLC20 phosphorylation. Confluent SMCs were sub-
jected to 10% average strain at 60 cycles per minute for 30 Summary
and 60 minutes. Basal MLC20 phosphorylation (N=non, In summary, recent data support the hypothesis that
M=mono, D=di) of serum-fed SMC was as follows: N=34%: vascular endothelial and smooth muscle cells are keenly sen-
M=27%: D=39%. After 60 minutes of cyclic strain, both the sitive to mechanical perturbation such as cyclic strain. Our
mono and diphosphorylated species of MLC20 were de- group and others have characterized changes in the pheno-
creased by 21% and 15%, respectively. The strain-induced type of both cell types and have begun to delineate the rel-
dephosphorylation of MLC20 was partially inhibited by the evant signaling pathways and strain-sensitive genes. Con-
protein phosphatase 1/2A inhibitor calyculin A. However, tinued advances in this field may prove beneficial in devel-
phosphorylase A phosphatase activities in Triton-soluble and oping novel approaches to improve tissue engineering of
insoluble fractions of SMCs were unaffected by cyclic strain. prosthetic grafts.
Table 39.4. Effect of cyclic strain on smooth muscle cell signaling pathways
References 21. Iba T, Mills I, Sumpio BE. Intracellular cyclic AMP levels
1. Panaro NJ, McIntire LV. Flow and shear stress effects on in endothelial cells subjected to cyclic strain in vitro. J
endothelial cell function. In: Sumpio BE, ed. Hemody- Surg Res 1992; 52(6):625-30.
namic Forces and Vascular Cell Biology. Austin: R.G. 22. Mills I, Cohen CR, Sumpio BE. Cyclic strain and vascu-
Landes Co., 1993:47-65. lar cell biology. In: Sumpio BE, ed. Hemodynamic Forces
2. Oluwole BO, Du W, Mills I, Sumpio BE. Gene regulation and Vascular Cell Biology. Austin: R.G. Landes Co.,
by mechanical forces. Endothelium 1997; 5:85-93. 1993:66-89.
3. Davies PF. Flow-mediated endothelial mechano- 23. Mills I, Murata K, Packer CS, Sumpio BE. Cyclic strain
transduction. Physiol Rev 1995; 75:519-560. stimulates dephosphorylation of the 20kDa regulatory
4. Nerem RM, Harrison DG, Taylor WR, Alexander RW. myosin light chain in vascular smooth muscle cells [pub-
Hemodynamics and vascular endothelial biology. J lished erratum appears in Biochem Biophys Res Commun
Cardiovasc Pharm 1993; 21(S1):S6-S10. 1995 Feb 27; 207(3):1058]. Biochem Biophys Res
5. Resnick N, Gimbrone MA Jr. Hemodynamic forces are Commun 1994; 205(1):79-84.
complex regulators of endothelial gene expression. FASEB 24. Mills I, Cohen CR, Kamal K, Li G, Shin T, Du W, Sumpio
J 1995; 9:874-882. BE. Strain activation of bovine aortic smooth muscle cell
6. Thubrikar MJ, Robicsek F. Pressure-induced arterial wall proliferation and alignment: Study of strain dependency
stress and atherosclerosis. Ann Thorac Surg 1995; and the role of protein kinase a and C signaling path-
59:1594-1603. ways. J Cell Physiol 1997; 170:228-234.
7. Langille BL. Chronic effects of blood flow on the artery 25. Mills I, Cohen CR, Kamal K, Sumpio BE. Cyclic strain
wall. In: Frangos JA, ed. Physical forces and the mamma- stimulates ADP-ribosylation of the inhibitory G protein,
lian cell. San Diego: Academic Press, 1993:249-274. Gi#, in bovine aortic endothelial cells. 1998; in press.
8. Patel DJ, Greenfield WG, Austen WG, Fry DL. Pressure 26. Mills I, Sumpio BE. Vascular smooth muscle cells. In:
flow relationships in the ascending aorta and femoral ar- Sidawy AN, Sumpio BE, DePalma RG, eds. The Basic Sci-
tery of man. J Appl Physiol 1965; 20:459-463. ence of Vascular Disease. Armonk: Futura Publishing
9. Bergel DH. The static elastic properties of the arterial wall. Company, Inc., 1997:187-226.
J Physiol 1961; 156:445-457. 27. Sumpio BE, Banes AJ, Levin LG, Johnson G. Mechanical
10. Steinman DA, Ethier CR. The effect of wall distensibility stress stimulates aortic endothelial cells to proliferate. J
on flow in a two-dimensional end-to-side anastomosis. J Vasc Surg 1987; 6:252-256.
Biomech Eng 1994; 116:294-301. 28. Sumpio BE, Banes AJ. Response of procine aortic smooth
11. Ballermann BJ, Ott MJ. Adhesion and differentiation of muscle cells to cyclic tensional deformation in culture. J
endothelial cells by exposure to chronic shear stress: A Surg Res 1988; 44:696-701.
vascular graft model. Blood Purification 1995; 13:125-134. 29. Sumpio BE, Banes AJ. Prostacyclin synthetic activity in
12. Ott MJ, Ballermann BJ. Shear stress-conditioned, endot- cultured aortic endothelial cells undergoing cyclic me-
helial cell-seeded vascular grafts: Improved cell adherence chanical deformation. Surgery 1988; 104:383-389.
in response to in vitro shear stress. Surgery 1995; 30. Sumpio BE, Widmann MD. Enhanced production of an
117:334-339. endothelium derived contracting factor by endothelial cells
13. Kohler TR, Kirkman TR, Kraiss LW, Zierler BK, Clowes subjected to pulsatile stretch. Surgery 1990; 108:277-282.
AW. Increased blood flow inhibits neointimal hyperpla- 31. Sumpio BE, Du W, Xu W-J. Exposure of endothelial cells
sia in endothelialized vascular grafts. Circ Res 1991; to cyclic strain induces c-fos, fosB and c-jun but not jun
69:1557-1565. B or jun D and increases the transcription factor AP-1.
14. Geary RL, Kohler TR, Vergel S, Kirkman TR, Clowes AW. Endothelium 1994; 2:149-156.
Time course of flow-induced smooth muscle cell prolif- 32. Sumpio BE, Chang R, Xu W-J, Wang X-J, Du W. Regu-
eration and intimal thickening in endothelialized baboon lation of tissue plasminogen activator in bovine endothe-
vascular grafts. Circ Res 1993; 74:14-23. lial cells exposed to cyclic strain: The functional signifi-
15. Kraiss LW, Geary RL, Mattsson EJR, Vergel S, Au YPT, cance of the CRE, AP-2 and SSRE sites. Am J Physiol
Clowes AW. Acute reductions in blood flow and shear 1997; 42:C1441-C1448.
stress induce platelet-derived growth factor-A expression 33. Augustin HG, Kozian DH, Johnson RC. Differentiation
in baboon prosthetic grafts. Circ Res 1996; 79:45-53. of endothelial cells: Analysis of the constitutive and acti-
16. Dobrin PB, Littooy FN, Endean ED. Mechanical factors vated endothelial cell phenotypes. Bioessays 1994;
predisposing to intimal hyperplasia and medial thicken- 16(12):901-6.
ing in autogenous vein grafts. Surgery 1989; 105:393-400. 34. Risau W. Differentiation of endothelium. FASEB Journal
17. Mills I, Cohen CR, Sumpio BE. In: Sumpio BE, ed. He- 1995; 9(10):926-33.
modynamic Forces and Vascular Cell Biology. Austin: R.G. 35. Thyberg J, Hedin U, Sjolund M, Palmberg L, Bottger BA.
Landes Co., 1993; 5:66-89. Regulation of differentiated properties and proliferation
18. Iba T, Sumpio BE. Morphological response of human of arterial smooth muscle cells. Arteriosclerosis 1990;
endothelial cells subjected to cyclic strain in vitro. 10:966-990.
Microvasc Res 1991; 42:245-254. 36. Owens GK. Role of mechanical strain in regulation of
19. Iba T, Shin T, Sonoda T, Rosales OR, Sumpio BE. Stimu- differentiation of vascular smooth muscle cells. Circ Res
lation of endothelial secretion of tissue-type plasminogen 1996; 79:1054-1055.
activator by repetitive stretch. J Surg Res 1991; 50:457-460. 37. Yano Y, Geibel J, Sumpio BE. Tyrosine phosphorylation
20. Iba T, Sumpio BE. Tissue plasminogen activator expres- of pp125FAK and paxillin in aortic endothelial cells induced
sion in endothelial cells exposed to cyclic strain in vitro. by mechanical strain. Am J Physiol 1996; 271:C635-C649.
Cell Transplantation 1991; (1):43-50. 38. Li G, Mills I, Sumpio BE. Cyclic strain stimulates endot-
helial cell proliferation. Characterization of the strain re-
quirements. Endothelium. 1994; 2:177-181.
Mechanical Forces and Cell Differentiation 437
39. Reusch P, Wagdy H, Reusch R, Wilson E, Ives HE. Me- inducer of prostacyclin synthase expression in bovine aor-
chanical strain increases smooth muscle and decreases tic endothelial cells. J Vasc Res 1997; 69:135-138.
nonmuscle expression in rat vascular smooth muscle cells. 58. Frangos JA, Eskin SG, McIntire LV, Ives CL. Flow effects
Circ Res 1996; 79:1046-1053. on prostacyclin production by cultured endothelial cells.
40. Wilson E, Mai Q, Sudhir K, Weiss RH, Ives HE. Mechani- Science 1985; 27:1477-1479.
cal strain induces growth of vascular smooth muscle cells 59. Uematsu M, Ohara Y, Navas JP, Nishida K, Murphy TJ,
via autocrine action of PDGF. Journal of Cell Biology Alexander RW, Nerem RM, Harrison DG. Regulation of
1993; 123(3):741-7. endothelial cell nitric oxide synthase mRNA expression
41. Zilla P, von Oppell U, Deutsch M. The endothelium: A by shear stress. Am J Physiol 1995; 269:C1371-C1378.
key to the future. J Card Surg 1993; 8:32-60. 60. Diamond SL, Sharefkin JB, Diefenbach C, Frazier-Scott
42. Ives CL, Eskin SG, McIntire LV. Mechanical effects on K, McIntire LV. Tissue plasminogen activator mRNA lev-
endothelial cell morphology: In vitro assessment. In Vitro els increase in cultured human endothelial cells exposed
Cell Dev Biol 1986; 22:500-507. to laminar shear stress. J Cell Physiol 1990; 143:364-371.
43. Shirinsky VP, Antonov AS, Birukov KG. Mechanochemi- 61. Morita T, Kurihara H, Maemura K, Yoshizumi M, Yazaki
cal control of human endothelium orientation and size. J Y. Disruption of cytoskeletal structures mediates shear
Cell Biol 1989; 109:331-339. stress-induced endothelin-1 gene expression in cultured
44. Awolesi MA, Widmann MD, Sessa WC, Sumpio BE. Cy- porcine aortic endothelial cells. J Clin Inv 1993;
clic strain increases endothelial nitric oxide synthase ac- 92:1706-1712.
tivity. Surgery 1994; 116(2):439-44; discussion 444-5. 62. Malek A, Izumo S. Physiological fluid shear stress causes
45. Awolesi MA, Sessa WC, Sumpio BE. Cyclic strain down-regulation of endothelin-1 mRNA in bovine aortic
upregulates nitric oxide synthase in cultured bovine aor- endothelium. Am J Physiol 1992; 263:C389-C396.
tic endothelial cells. J Clin Invest 1995; 96(3):1449-54. 63. Rosales OR, Isales CM, Barrett PQ, Brophy C, Sumpio
46. Cheng JJ, Chao YJ, Wung BS, Wang DL. Cyclic strain- BE. Exposure of endothelial cells to cyclic strain induces
induced plasminogen activator inhibitor-1 (PAI-1) release elevations of cytosolic calcium through mobilization of
from endothelial cells involves reactive oxygen species. intracellular and extracellular pools. Biochem J 1997;
Biochem Biophys Res Commun 1996; 225(1):100-5. 326:385-392.
47. Davies PF, Tripathi SC. Mechanical stress mechanisms and 64. Geiger RV, Berk BC, Alexander RW, Nerem RM. Flow-
the cell. An endothelial paradigm. Circ Res 1993; induced calcium transients in single endothelial cells: Spa-
72:239-245. tial and temporal analysis. Am J Physiol 1992;
48. Dewey CF Jr, Bussolari SR, Gimbrone MA Jr, Davies PF. 262:C1411-C1417.
The dynamic response of the vascular endothelial cells to 65. Evans L, Frenkel L, Brophy CM, Rosales O, Sudhaker S,
fluid shear stress. J Biomech Eng 1981; 103:177-188. Li G, Du W, Sumpio BE. Activation of diacylglycerol in
49. Davies PF, Robotewskyj A, Griem ML. Quantitative stud- cultured endothelial cells exposed to cyclic strain. Am J
ies of endothelial cell adhesion. Directional remodeling Physiol 1997; 41:C650-C659.
of focal adhesion sites in response to flow forces. J Clin 66. Bhagyalakshmi A, Berthiaume F, Reich KM, Frangos JA.
Inv 1994; 93:2031-2038. Fluid shear stress stimulates membrane phospholipid
50. Yano Y, Geibel J, Sumpio BE. Cyclic strain induces reor- metabolism in cultured human endothelial cells. J Vasc
ganization of integrin #2!1 in human umbilical vein en- Res 1992; 29:443-449.
dothelial cells. J Cell Biochem 1997; 64:505-513. 67. Kroll MH, Hellums JD, Guo Z, Durante W, Razdan K,
51. Thoumine O, Nerem RM, Girard PR. Changes in organi- Hrbolich JK, Schafer AI. Protein kinase c is activated in
zation and composition of the extracellular matrix un- platelets subjected to pathological shear stress. J Biol Chem
derlying cultured endothelial cells exposed to laminar 1993; 268:3520-3524.
steady shear stress. Lab Invest 1995; 73:565-576. 68. Cohen CR, Mills I, Du W, Kamal K, Sumpio BE. Activa-
52. Davies PF. Flow-mediated signal transduction in endo- tion of the adenylyl cyclase/cyclic AMP/protein kinase a
thelial cells. In: Bevan J, Kaley G, Rubanyi G, eds. Flow pathway in endothelial cells exposed to cyclic strain. Expt
dependent regulation of vascular function in health and Cell Res 1997; 231:184-189.
disease. New York: Academic Press, 1984:46-61. 69. Gudi SRP, Clark CB, Frangos JA. Fluid flow rapidly acti-
53. Eskin SG, Ives CL, McIntire LV, Navarro LT. Response vates G proteinse in human endothelial cells. Involvement
of cultured endothelial cells to steady flow. Microvasc Res of G proteins in mechanochemical signal transduction.
1984; 28:87-93. Circ Res 1996; 79:834-839.
54. Levesque MJ, Nerem RM. The study of rheological effects 70. Kuchan MJ, Jo H, Frangos JA. Role of G proteins in shear
on vascular endothelial cells in culture. Biorheology 1989; stress-mediated nitric oxide production by endothelial
26:345-357. cells. Am J Physiol 1994; 267:C753-C758.
55. Girard P, Nerem RM. Shear stress modulates endothelial 71. Berthiaume F, Frangos JA. Flow-induced prostacyclin pro-
cell morphology and F-actin organization through the duction is mediated by a pertussis toxin-sensitive G pro-
regulation of focal adhesion-associated proteins. J Cell tein. FEBS Lett 1992; 308:277-279.
Physiol 1995; 163:179-193. 72. Rosales OR, Sumpio BE. Protein kinase c is a mediator
56. Yano Y, Saito Y, Narumiya S, Sumpio BE. Involvement of the adaptation of vascular endothelial cells to cyclic
of rho p21 in cyclic strain-induced tyrosine phosphoryla- strain in vitro. Surgery 1992; 112(2):459-66.
tion of focal adhesion kinase (pp125FAK), morphological 73. Murata K, Mills I, Sumpio BE. Protein phosphatase 2A
changes and migration of endothelial cells. Biochem in stretch-induced endothelial cell proliferation. J Cell
Biophys Res Commun 1996; 224(2):508-15. Biochem 1996; 62:1-9.
57. Segurola RJ, Oluwole B, Mills I, Yokoyama C, Tanabe T, 74. Rosales OR, Sumpio BE. Changes in cyclic strain increase
Kito H, Nakajima N, Sumpio BE. Cyclic strain is a weak inositol trisphosphate and diacylglycerol in endothelial
cells. Am J Physiol 1992; 262(4 Pt 1):C956-62.
438 Tissue Engineering of Prosthetic Vascular Grafts
75. Nollert MU, Eskin SG, McIntire LV. Shear stress increases of mechanical strain on monocyte adhesion to endothe-
inositol trispohosphate levels in human endothelial cells. lial cells. Circulation Research. 1995; 77(2):294-302.
Biochem Biophys Res Comm 1990; 170:281-290. 89. Shyy YJ, Lin MC, Han J, Lu Y, Petrime M, Chien S. The
76. Ohno M, Cooke JP, Dzau VJ, Gibbons GH. Fluid shear cis-acting phorbol ester “12-O-tetradecanoylphorbol 13-
stress induces endothelial TGF-!1 transcription and pro- acetate”-responsive element is involved in shear stress-
duction. Modulation by potassium channel blockade. J induced monocyte chemotactic protein 1 gene expression.
Clin Inv 1995; 95:1363-1369. Proc Natl Acad Sci 1995; 92:8069-8073.
77. Tseng H, Peterson TE, Berk BC. Fluid shear stress stimu- 90. Nagel T, Resnick N, Atkinson WJ, Dewey CF Jr, Gimbrone
lates mitogen-activated protein kinase in endothelial cells. MA Jr. Shear stress upregulates intercellular adhesion
Circ Res 1995; 77:869-878. molecule-1 expression in cultured human vascular endot-
78. Du W, Mills I, Sumpio BE. Cyclic strain causes heteroge- helial cells. J Clin Inv 1994; 94:885-891.
neous induction of transcription factors, AP-1, CRE bind- 91. Moose PRL, Campbell GR, Wang ZL, Campbell JH.
ing protein and Nf-∀b, in endothelial cells: Species and Smooth muscle phenotypic expression in human carotid
vascular bed diversity. J Biomech 1995; 28(12):1485-91. arteries. I. Comparison of cells from diffuse intimal thick-
79. Lan Q, Mercurius KO, Davies PF. Stimulation of tran- enings adjacent to atheromatous plaque, and cultured rat
scription factors Nf-∀b and AP1 in endothelial cells sub- aortic media. Lab Inv 1985; 53:556-562.
jected to shear stress. Biochem Biophys Res Comm 1994; 92. Campbell JH, Campbell GR. Methods of growing vascu-
201:950-956. lar smooth muscle cells in culture. In: Campbell JH,
80. Resnick N, Collins T, Atkinson W, Bonthron DT, Dewey Campbell GR, eds. Vascular smooth muscle in culture.
CF Jr, Gimbrone MA. Platelet-derived growth factor B Boca Raton: CRC Press, 1987:15-21.
chain promoter contains a cis-acting fluid shear-stress- 93. Gordon D, Schwartz SM. Arterial smooth muscle differ-
responsive element. Proc Natl Acad Sci 1993; entiation. In: Campbell JH, Campbell GR, eds. Vascular
90:4591-4595. smooth muscle in culture. Boca Raton: CRC Press,
81. Khachigian LM, Resnick N, Gimbrone MA Jr, Collins T. 1987:1-14.
Nuclear factor-∀B interacts functionally with the platelet- 94. Chamley JH, Campbell GR. Mitosis of contractile smooth
derived growth factor B-chain shear-stress response ele- muscle cells in tissue culture. Expt Cell Res 1974;
ment in vascular endothelial cells exposed to fluid shear 84:105-110.
stress. J Clin Inv 1995; 96:1169-1175. 95. Birukov KG, Shirinsky VP, Stepanova OV, Tkachuk VA,
82. Wang DL, Wung BS, Peng YC, Wang JJ. Mechanical strain Hahn AWA, Resink TJ, Smirnov VN. Stretch affects phe-
increases endothelin-1 gene expression via protein kinase notype and proliferation of vascular smooth muscle cells.
c pathway in human endothelial cells. J Cell Physiol 1995; Mol Cell Biochem 1995; 144:131-139.
163(2):400-6. 96. Wilson E, Sudhir K, Ives HE. Mechanical strain of rat
83. Malek AM, Greene AL, Izumo S. Regulation of endothelin vascular smooth muscle cells is sensed by specific extra-
1 gene by fluid shear stress is transcriptionally mediated cellular matrix/integrin interactins. J Clin Inv 1995;
and independent of protein kinase c and cAMP. Proc Natl 96:2364-2372.
Acad Sci USA 1993; 90:5999-6003. 97. Kanda K, Matsuda T. Mechanical stress-induced orienta-
84. Morita T, Kurihara H, Maemura K, Yoshizumi M, Nagai tion and ultrastructural change of smooth muscle cells
R, Yazaki Y. Role of Ca2+ and protein kinase c in shear cultured in three-dimensional collagen lattices. Cell Trans-
stress-induced actin depolymerization and endothelin 1 plant 1994; 3(6):481-92.
gene expression. Circ Res 1994; 75:630-636. 98. Yang Z, Noll G, Luscher TF. Calcium antagonists differ-
85. Grabowski EF, Jaffe EA, Weksler BB. Prostacyclin produc- ently inhibit proliferation of human coronary smooth
tion by cultured endothelial cell monolayers exposed to muscle cells in response to pulsatile stretch and platelet-
step increases in shear stress. J Lab Clin Med 1985; derived growth factor. Circ 1993; 88:832-836.
105:36-43. 99. Davis MG, Ali S, Leikauf GD, Dorn. GWI Tyrosine ki-
86. Norris M, Morigi M, Donadelli R, Aiello S, Foppolo M, nase inhibition prevents deformation-stimulated vascular
Todeschini M, Orisio S, Remuzzi G, Remuzzi A. Nitric smooth muscle growth. Hypertension 1994; 24:706-713.
oxide synthesis by cultured endothelial cells is modulated 100. Kulik TJ, Alvarado AP. Effect of stretch on growth and
by flow conditions. Circ Res 1995; 76:536-543. collagen synthesis in cultured rat and lamb pulmonary
87. Topper JN, Cai J, Falb D, Gimbrone MA Jr Identification arterial smooth muscle cells. J Cell Physiol 1993;
of vascular endothelial genes differentially responsive to 157:615-624.
fluid mechanical stimuli: Cyclooxygenase-2, manganese 101. Kolpakov V, Rekhter MD, Gordon D, Wang WH, Kulik
superoxide dismutase, and endothelial cell nitric oxide TJ. Effect of mechanical forces on growth and matrix pro-
synthase are selectively up-regulated by steady laminar tein synthesis in the in vitro pulmonary artery. Analysis
shear stress. Proc Natl Acad Sci USA 1996; of the role of individual cell types. Circ Res 1995;
93:10417-10422. 77:823-831.
88. Wang DL, Wung BS, Shyy YJ, Lin CF, Chao YJ, Usami S, 102. Rovner AS, Murphy RA, Owens GK. Expression of smooth
Chien S. Mechanical strain induces monocyte chemotac- muscle and nonmuscle myosin heavy chains in cultured
tic protein-1 gene expression in endothelial cells. Effects vascular smooth muscle cells. J Biol Chem 1986;
261:14740-14745.
PART III
Biointeractive Prostheses: Complete Healing
Engineering Components
Scaffold Engineering
Structural Designs
CHAPTER 40
An Integral Mathematical Approach to Tissue Engineering
of Vascular Grafts
Greg R. Starke, A.S. Douglas, D.J. Conway
Introduction
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
442 Tissue Engineering of Prosthetic Vascular Grafts
graft and the host artery sizes was approximately 1.45. How- ing the design process by allowing the developer to test de-
ever, in a polymeric tissue engineered graft the implant sign concepts and parameters without the time consuming,
should not be assumed to be rigid. Therefore the optimal and costly, process of prototyping. Furthermore, it is often
size of the graft in relation to the host needs to be investi- difficult—if not impossible—to measure quantities such as
gated in the light of graft compliance. In addition, the pres- stress, strain or displacement. These quantities can, how-
ence of soft tissue within the graft structure will alter the ever, be readily calculated from mathematical models of the
compliance, and will also alter the suture line stresses at the loaded implant. Accurate predictions require sound consti-
anastomosis. Therefore the compliance matching and graft tutive (material) laws describing the behavior of the intact
size selection needs to consider the evolutionary nature of artery as well as the proposed graft material. Analytical so-
the implant compliance. Reproducing the stress/strain be- lutions of arterial systems are rarely possible because of the
havior of the native artery also means that diameter com- complexity of the material formulations, structures, bound-
patibility can be examined. If diameter matching is achieved ary conditions and initial loading conditions. However,
in the long term, then stress concentrations at the anasto- making use of sophisticated numerical techniques such as
mosis will be kept to a minimum and thus progressive re- the finite element method can approximate solutions.
modelling will not be necessary to reach a homeostatic state. Many of the mechanical design requirements can,
A tissue engineered vascular graft is intended to pro- however, be derived from the mechanics of the intact ar-
vide a scaffold for the development of tissue; therefore, the tery—as the success of the graft will depend in part on rec-
structure has to be sufficiently porous, with interconnectivity, reating the mechanics of the natural vessel. However, the
so as to allow for vascularization and the subsequent infil- mechanics of the native vessel needs to be reproduced in a
tration of cells. However, in the early post-operative period, synthetic material with a highly porous structure. In addi-
before tissue ingrowth has taken place, the scaffold needs to tion, the process of tissue infiltration and development will
have sufficient structural integrity to withstand the applied alter the mechanical properties of the graft. The remainder
loads, without leading to excessive deformation or rupture. of this chapter is therefore concerned with the development
Greer et al14 investigated the material properties of a col- of a numerical formulation for mechanics of the native ar-
lagen/smooth muscle cell gel and found that the stress/strain tery as well as a polymer graft.
characteristics, as well as the stress relaxation, were both
strongly dependent on the extent of cell seeding. This pre- The Mechanics of Arteries
liminary study shows how the biological features of a tis- The flow of blood can be related to the elasticity of an
sue-engineered graft can alter the mechanics of the graft. artery by considering the feedback loop shown in Figure 40.1.
Furthermore, the development of tissue may depend on the The bottom block shows the artery as a rigid conduit allow-
loading environment. Therefore, the graft structure needs ing blood flow. A pulsating pressure results from this flow.
to create optimal strain conditions, during both diastole and If the artery is treated as an elastic body, as shown in the top
systole, that will facilitate the development and maintenance block, the pressure will then act to deform the conduit, which
of smooth muscle cells within the porous structure. in turn will affect the nature of the flow. The flow character-
Mathematical modelling provides a means for char- istics can then be analyzed assuming that the vessel is rigid,
acterizing the mechanical response to loading of the native and has a diameter that has been altered by the flow pres-
artery, as well as the structured polymer from which a tissue sure. The deformation of the artery can then be determined
engineered graft might be constructed. Such models allow using the laws of elasticity assuming a constant pressure and
for the investigation of compliance, remodelling, sizing and determining the nature of the deformation. In this way the
failure prediction. This approach offers the potential for aid- flow of blood is coupled to the elasticity of the vessels.
Structural Morphology ure 40.2, which shows experimentally determined and nu-
The structural characteristics of arteries arise from the merically predicted stress/strain relationships for the arte-
tissue composition of the media and the adventitia. The rial walls of four normal rabbit arteries.17 The effective
media is made up of smooth muscle cells and bundles of Young’s modulus of the material can be viewed as the slope
collagen fibrils which are layered between a number of elas- of the stress/strain curve. Since the modulus increases with
tic sheets. The structure of the adventitia results from col- strain, arterial stiffness is a non-linear function of strain.
lagen fibres, fibroblasts and ground substance. A relatively The stiffening of the arterial wall, which occurs when the
thick layer of elastin separates the media and adventitia. The collagen bundles become straightened and go into tension,
endothelial cells of the intima do not have any significant is observed in the region of 150 mm Hg pressure. This be-
influence on the artery mechanics, and therefore the intima havior protects the arteries from excessive dilation and there-
and media can essentially be grouped together in so far as fore offers protection against the development of aneurysms.
the elastic response is concerned. In the low strain region The complex composition and structure of arteries
(near zero stress) the stiffness of the intima-media layer is means that the material properties of the wall are not the
almost an order of magnitude greater than that of the ad- same in all directions (the material is therefore anisotropic).
ventitia. However, at larger strains the collagen bundles be- Several research studies have shown that the stiffness of the
gin to straighten and then stretch, which increases the stiff- arterial wall in the circumferential direction is greater than
ness contribution from the adventitia. The relative thick- in the axial direction.19,20 However, this result is by no means
ness of the intima-media/adventitia varies as a function of conclusive, as previously Tanaka and Fung21 found the op-
the position in the body and therefore so too does the elas- posite result. The are many factors, including the specimen,
tic response.15 The walls of the artery are structurally aniso- species and testing methodology, to which the difference in
tropic, incompressible and highly deformable, exhibiting results may be attributed.
non-linear stress/strain characteristics. As a result of the high fluid content in the arterial wall
tissue, the material is often assumed to be incompressible—
Non-Linear Elastic Response hence volume is preserved during deformation. Such an as-
The most striking aspect of the mechanical response sumption considerably simplifies the mechanics of the walls,
of arteries is the non-linear relation between the pressure as the deformation in the radial direction can be directly
acting within an artery and the resulting change in diam- related to the deformation in the longitudinal and circum-
eter. Experimental evidence16-18 has shown that as the pres- ferential directions. The assumption of incompressibility has
sure increases, and hence the stress within the arterial walls, been validated by Choung and Fung23 who found that the
the change in diameter is initially rapid, but as the stress percentage of fluid extrusion of the undeformed tissue was
increases the change in strain becomes smaller until increases between 0.5 and 1.26% as a result of radial compression of
in stress are accompanied by only very small increases in rabbit aortic tissue. This result showed that, while the arterial
strain. This high degree of non-linearity is illustrated in Fig- tissue is slightly compressible, it would suffice to assume
incompressibility.
In spite of this evidence, most research in this field radially along the longitudinal axis. Pre-stressing has been
still assumes quasi-linear viscoelasticity (i.e., non-linear in investigated by several researchers,41,42 resulting in the “uni-
the elastic deformation response but linear in the history of form strain” hypothesis.
deformation). This approach avoids difficulties associated Fung et al42 examined the no load and no stress state
with implementing non-linear viscoelasticity numerically, of the pulmonary and ileac arteries of rats. Figure 40.4a
and the more complex materials testing that would be re- shows the cross section of the pulmonary artery at a blood
quired for the constitutive characterisation. Huyghe et al39 pressure of 15 mm Hg. In the no load state (Fig. 40.4b) the
implemented a quasi-linear viscoelastic formulation for pas- artery has reduced considerably in diameter and has there-
sive heart muscle tissue. They found that all the mechanical fore increased in wall thickness. Figure 40.4c shows the sec-
characteristics of tissue—which are essentially the same for tion of the artery following the radial cut that has removed
arterial wall tissue (including the weak frequency depen- the pre-stress. In order to quantify the extent of pre-stress-
dency of the dissipation energy during cyclic loading (i.e., ing within the wall, the angle to which the artery opens is
lag time))—were accommodated by their model. However, measured. This angle determines the amount of displace-
when they set the relaxation parameters in order to correctly ment caused by the pre-stress and can therefore be related
predict the dissipation energy, they found that the model to the magnitude of pre-stress.41
overpredicted the amount of stress relaxation by over 100%. In a further study, Greenwald et al43 measured the
This discrepancy they attributed to the non-linear viscoelas- opening angle of rat aortic arteries following the removal of
ticity of true cardiac tissue. various of the structural components. They found that the
Young et al,16 Decraemer et al,32 Wu and Lee33 and removal of smooth muscle and collagen had no effect on
Johnson et al35 have all incorporated non-linear viscoelas- the opening angle, while the removal of elastin had a sig-
ticity in their constitutive models for soft biological tissues. nificant effect, resulting in a decrease in the opening angle.
These laws were reported to predict results that correlate This study concluded that the residual stresses are stored in
fairly well with existing experimental test data for tissues the elastic component of the media.
such as human patellar tendon, canine collateral ligament The opening of the artery shows that the pre-stress-
and canine arterial wall tissue. Such models do have poten- ing puts the internal surface of the wall into a state of com-
tial to make more accurate predictions in the design process pression and the external surface into tension. Subsequently,
than the simpler quasi-linear laws. However, the experimen- the tensile stresses that are experienced during normal load-
tal determination of the constitutive parameters is neces- ing are reduced by the initial compressive component, re-
sarily a far more complex task with these more realistic sulting in a more uniform distribution of stress, as is shown
models. Until accurate and accessible experimental meth- schematically in Figure 40.5.
ods are developed which enable such parameterization, it
can be argued that the simpler, numerically more robust Material Laws for the Native Artery
quasi-linear laws are sufficient. Myers et al40 discuss the limi- and Graft Materials
tations of quasi-linear theory and suggest methods for re- The extent of the porosity of a polymeric graft, as well
ducing the effects of this simplifying assumption. as the nature of the structure, requires careful engineering
design which will consider not only the mechanical aspects,
Pre-Stress but also the coupling with the biological design. In design-
In a thick walled cylinder under internal pressure the ing an artery graft, many of the design objectives can be
distribution of the circumferential strain through the wall learned from studying the mechanics of the intact vessel.
varies considerably, with a peak at the inner surface which Also, failures of grafts typically occur in the region of the
decays exponentially towards the outer surface. It has been anastomosis, and a mechanical model of the interaction
shown that several tissue types, including bone, will remodel between the graft and the host is therefore needed for graft
so as to reach a homeostatic strain condition. In order to design. Since any mechanical model will require the consti-
moderate the peak strain, and produce a more uniform dis- tutive response of both the graft and the natural artery, the
tribution through the thickness, the artery walls are pre- fundamentals of the mechanical response of compliant
stressed. This pre-stressing is evident by the way an artery materials are reviewed.
will spring open if a transverse section is removed and cut
Compliant engineering materials (such as polymers) 5(X), to the displacements of each material point, u(X), if
and soft tissues deform appreciably under load and care must we note that the deformed position is merely the sum of the
be taken to describe their behavior. Most soft tissues sus- reference position and the displacement of that point, viz.
tain large strains, are generally non-linear in their con- x = X + u(X). To characterize the deformation of this mate-
stitutive response (see Fig. 40.2), exhibit either transverse rial we need to examine how the deformed position or the
isotropy or anisotropy and are either incompressible or displacements change spatially. Both the deformation gra-
nearly so. Compliant materials, or rubber-elastic materials, dient tensor, F, the components of which are given by
also sustain large strains, may be anisotropic and are often
incompressible. (1)
To describe the mechanical behavior of both soft
tissues and compliant engineering materials, researchers where i, j=1,2,3, and the displacement gradient ten-
usually use a hyperelastic (Green elastic) strain energy sor, H, with components given by
function of the local deformation or strain, W. The existence
of a strain energy function for a material implies that it is (2)
hyperelastic and that, regardless of deformation path, equal
work is done in going from one deformation (or strain) state characterize the deformation at the point X. The deforma-
to another. In order to characterize compliant graft materials tion gradient tensor F also contains information about the
and the natural artery, we need to formulate appropriate local rotation. However, since we would like to relate the
strain energy functions W. To do this we must first develop strain energy function, W, to deformation alone, we define
a framework with which to describe the deformations, strains the right and left Cauchy-Green deformation tensors, C=FTF
and stresses required. Note that, since soft tissue is not strictly and B=FFT, respectively, which depend only on the defor-
hyperelastic, it is characterized by a pseudostrain mation (not the rotation) at any point in the material. Strain
energy function. is also a measure of deformation, independent of rotation,
and used to characterize the strain energy function. The
Characterizing Finite Deformation Lagrangian strain, E, is defined by
The motion of a compliant material can be described
as a mapping,5, which defines the deformed position, x, of E = 12 F T F % I =
[ ] [H + H
1
2
T
+ H TH ] (3)
a particle which occupied the position, X, in the reference
configuration; thus x = 5(X). We can also relate the motion, where I is the identity tensor (Note that F = H + I).
An Integral Mathematical Approach to Tissue Engineering of Vascular Grafts 447
Spring and dashpot models are a good analogy for s(t) = ket=0e–t/t for the
modelling material viscoelastic behavior, since they allow ( Maxwell Model.
the characterisation of the material both qualitatively and The relaxation time,
quantitatively. The simplest model for the elastic behavior t, is the time taken for
of a material is the linear spring where stress, (, is simply the stress to
proportional to the strain, >, relax by a factor
e -1Χ0.37 of the initial
( ( = k>) spring , (14) t value.
where k is the spring constant. The spring element A true viscoelastic material will exhibit a delayed elas-
stores elastic energy. The lagging component of a material tic response, stress relaxation and strain recovery (i.e., a
response to an applied load can be modelled with a dashpot gradual return to the original undeformed configuration on
(which dissipates energy), where the stress is proportional the removal of all applied loads). Neither the Kelvin-Voigt
to the strain rate >: nor the Maxwell model adequately represents all these char-
acteristics. However, a combination of the two models, giv-
( ( = ?>& ) dashpot, (15) ing the 3-element or standard linear models shown in Fig-
ure 40.7, will accommodate all the required characteristics.
where ? is the viscosity constant. The two simplest vis-
In order to capture the full range of time scales (i.e., a
coelastic models are the Kelvin-Voigt and Maxwell models,
continuous spectrum) that typically exist in a true viscoelas-
illustrated in Figure 40.6.
tic material, one would need to assemble an infinite num-
In the Kelvin-Voigt model, the strain is the same in
ber of these 3-element units in series or parallel. However,
both the spring and the dashpot, and the stresses in each
the major viscoelastic trends can usually be satisfactorily
element add to balance the applied load. Thus the equilib-
characterized with only a few such units. Langewouters et
rium equation is:
al29 found good correlation to human aortic creep curves
( = k > + ?>&
for their 5-element model (i.e., with only two time scales).
(16) van Dijk46 also characterized smooth muscle cell stress re-
laxation with two time constants. These models are param-
In the Maxwell model, the strains are additive, while
eterized by curve fitting to delayed elastic or stress relax-
the stress is constant throughout, thus:
ation experimental data.
1 1 For a multi-element Maxwell model (with N units),
>& = (
& + ( (17)
k ? the stress relaxation under a constant strain is:
N
((t ) = > 0 G(t ) where G ( t ) = G≅ + Α Gn e %t /4
(The Maxwell equilibrium is written in terms of strain
rate since the dashpot equilibrium is defined in terms of
strain rate). Solving these two differential equations yields: n=1
k G( t ) = S (4 )e %t 4 d4
t for the Kelvin-Voigt
Β0
(19)
2. The relaxation stress at time, t, for a constant strain where S (4 ) is the relaxation spectrum. Experimen-
applied at time, t = 0 model where t = h/k;
tally, the determination of this S(4) is perhaps easier than
finding discrete time scales and moduli from creep test data.
The methodology for finding such a relaxation spectrum is
described in the section “Materials Testing”.
Thus far we have only considered the time-varying fore, in this case it would be advantageous to include infor-
response to a constant load applied at some time in the past. mation about the microstructure in the description of the
Assuming linear viscoelastic behavior, we can use the prin- material behavior. The porous microstructure is then mod-
ciple of linear superposition to calculate the response to a eled as a continuum (solid) region that has the apparent (or
time-varying load. Casting the problem into an incremen- effective) properties of the porous material. This idea can
tal form, an increment in strain for the function, >(t), over a be taken one step further by including structural properties,
small time increment )t is written as such as porosity, as variables in the material formulation.
d> 4 Obviously the relation between the material behavior and
)> (4 ) = ) t at time, t = 4 . (20) the structural property needs to be determined. But once
dt this has been achieved, it becomes a simple matter to exam-
Thus from Equation (18) the stress response at time t ine the influence of changes in material microstructure on
(where t > 4) to this strain increment (held constant after its the structural response of the component without having
application) is to undertake complex geometrical modeling.
d> 4 In most porous materials the single most important
((t ))> (4) = G (t % 4 ) )t . (21) parameter in determining the mechanical properties is the
dt relative density ∋/∋s, where ∋ is the apparent density while
Now, the principle of linear superposition allows us ∋s is the density of the structural members. The porosity is
to sum the elastic responses to all the past increments in therefore defined as 1-∋/∋s. It turns out that cell size is gen-
load from time -≅ to t. By taking infinitesimal increments in erally far less important in the determination of material
time we can write Equation (21) in integral form as properties; however, cell shape will have a notable influence.
t
d> The distinction needs to be made between open and closed
(( t) = G ( t % 4) d4 .
Β %≅
Integrating by part yields
dt
(22) cells: In an open configuration, the cells are interconnected,
whereas in a closed cell arrangement each cell is isolated from
its neighbor. As interconnectivity is vital in a tissue-engi-
t
dG(t % 4) neered graft, only the mechanics of open celled structures
(( t) = G(0 ) > (t ) % > (4 )d4
Β -≅ dt
where G(0) is the so called glassy or instantaneous
(23) will be considered. Most porous structures are anisotropic
to a greater or lesser extent. Anisotropy arises from the shape
of the cells, and is characterized by the ratio of the largest to
modulus. the smallest cell dimension (R). Typically in a foam that is
Similarly, the delayed strain response to a smooth stress considered to be isotropic, R Χ 1.3; however, values in the
function of time is range from one to ten are not uncommon.
t
d J (t % 4) For most linear-elastic, isotropic, cellular structures
> (t) = J ( 0) ((t ) % (( 4) d4
Β %≅ d4
≅
(24) the apparent material modulus E can be related to the ap-
parent density by
J (t ) = T(4)(1 % e %t 4 )d4 ,
where
Β 0
and E E s ∆ (∋ ∋ s )
M
(25)
Based on analytical and experimental studies,47 pa- amount of work, and therefore a strong dependence on strain
rameters such as the yield strength and the compressive col- rate exists. In the early postoperative stages of a tissue-engi-
lapse load of a porous material can also be determined as a neered graft, the graft material will contain fluid. In addi-
function of relative density and material structure. How- tion, the normal loading rate of approximately one cycle per
ever, these investigations are limited to the case where the second will likely be high enough to make the influence of
material comprising the structure of a foam is operating in loading rate noticeable. For these reasons the fluid phase of
the linear elastic regime. In a highly porous material sub- the porous structure needs to be given consideration. The
jected to large forces this would not be the case, and there- foam can be treated as a porous material with an absolute
fore other means of characterizing the material response are permeability K. The velocity of the fluid v within the porous
required. One approach for determining the apparent prop- material can then be determined from Darcy’s law
K dp
erties of a material is to develop numerical models of the
microstructure under investigation. If the behavior of the v= % , (26)
material from which the structure is made has been com- ∝ dx
pletely characterized, and if the material has a uniform struc- where ∝ is the dynamic viscosity of the fluid and dp/dx
ture, then a single structural unit can be tested in order to is the pressure gradient within the porous structure. The per-
obtain the apparent properties of the material. meability can be determined from the relative density and
An example of this approach is shown in Figure 40.8. the pore structure, while the pressure gradient can be re-
The polyurethane microstructure (Fig. 40.8a) has been ide- lated to the applied stress. After some mathematical manipu-
alized and modeled using the finite element method lation the strength of the porous material can be related to
(Fig. 40.8b). This model is then subjected to a range of nu- the viscosity of the fluid, the strain rate and some geometri-
merical tests, from which the apparent material behavior can cal information.
be determined. This approach has great flexibility, as it makes
possible the investigation of the influence of relative den- Material Testing
sity, cell shape and size. It also becomes a relatively simple Material testing forms the basis of the characteriza-
matter to distort the cells so as to create an anisotropic struc- tion of the mechanical properties of the native artery and
ture and then quantify the structural response. Once the the graft material. The material morphology and the extent
nature of the apparent material has been determined this to which the material needs to be characterized determine
information can be used to model a complete implant the complexity of the testing. For isotropic materials, simple
(Fig. 40.8c) without having to consider the geometry of the uniaxial extension tests may be sufficient to obtain the nec-
internal microstructure. essary information for characterizing the material. However,
The presence of a viscous fluid within a porous mate- for transversely isotropic and orthotropic materials, biaxial
rial will have a considerable influence on the strength. When testing is required.
the material is compressed the fluid will be squeezed out, Hayashi et al9 developed a highly elastic, blood com-
while it will be drawn in when the material is extended. As patible, small diameter vascular graft fabricated from seg-
the fluid has viscosity, work is done during material defor- mented polyurethane. Introducing porosity into the mate-
mation. The higher the loading rate the greater will be the rial, which also allowed for possible tissue ingrowth, created
a physiologically realistic compliance for the graft. Electron more, the time dependent behavior (viscoelasticity) of the
microscopy revealed the graft material to be approximately material will influence the sizing of the graft, and therefore
isotropic, and therefore the mechanical properties were de- needs to be considered in the design of the implant.
termined using simple uniaxial tests. Their strength tests
revealed the importance of pretreatment of the material prior Hyperelastic Properties
to mechanical testing. They found that the material became In order to develop the constitutive models for soft
stable, in terms of mechanical properties, after having been tissues and compliant engineering materials, the general
immersed in a saline solution for 30 minutes. form of the strain energy function must be postulated. This
They compared the stress/strain characteristics of their should be as simple and convenient as possible, as labora-
graft material to those of traditional grafts as well as the tory tests will be required to set the relevant parameters in
natural artery (Fig. 40.9). In order to present excessive dila- the strain energy function. These energy functions are based
tion, the graft was coated with a woolly polyester net. How- on qualitative observations of the material behavior and on
ever, such a net will reduce the compliance of the graft. The prior testing of similar materials. The functional form of
mechanical test results showed that the porous polyurethane the strain energy function is subject to much debate and
exhibited an almost linear stress/strain response (KP), with both polynomial and exponential forms are in current use.
a slight tendency to strain more at higher stresses. This re- The development of sound constitutive laws to de-
sult is in contrast to the highly non-linear behavior of the scribe the mechanical behavior of arterial vessels has been
calf ’s thoracic aorta (Ao) which shows a very strong decline the focus of numerous research efforts.17,48-50 Since these
in the amount of strain at elevated stresses. However, in the constitutive laws are set out in terms of large strain
low strain region (up to approximately 30%) the stiffness of hyperelasticity, experiments are required to determine both
the polyurethane graft is very similar to the thoracic aorta. the functional form and the coefficients defining the strain
This result further shows the considerable difference in stiff- energy function, W.
ness between the compliant polyurethane graft and the more Essentially three forms of strain energy functions are
stiff traditional Dacron grafts (CW, GT, CV and DB). used to describe arterial walls: polynomial, exponential and
Although the material developed for this graft is very logarithmic. The polynomial function of Vaishnav et al,51
compliant and exhibits strain of over 200% before failure, it which was used to describe a cylindrical vessel subject to
does not show the stiffening behavior normally associated internal pressure, is the simplest; in general it has seven co-
with polymeric materials. The slight softening at high strains efficients and is given as follows:
may be a result of the failure of the woolly coating. Further-
(27)
where a, b, c, d, e, f, g are material constants, and E∗∗ Dynamic testing with sinusoidal loading over a large
and Ezz are the longitudinal strain components in the cir- range of frequencies will provide data for the determina-
cumferential and axial directions, respectively. tion of the continuous spectrum functions S(4) and T(4).
Fung et al17 proposed an exponential function of the The viscoelastic strain response to a sinusoidal stress,
form
( ( t ) = ( 0 sin Ε t , (30)
W = C exp a1 E∗∗
2
(+ a 2 E 2zz + a4 E∗∗ E zz ) (28)
at frequency Ε, and amplitude (0 is
where c, #1, #2 and #4 are the material constants.
Experimental data was determined from the cardiac, left iliac, >( t ) = > 0 sin(Ε t - Φ ) (31)
lower aorta and upper aortic arteries of rabbits, which was
used to determine the coefficients defining the strain energy where the peak strain >0 lags the peak stress by Φ—the
functions. The comparison of the experimental and theo- loss angle. These relations can be written in complex form
retical results, using Equation (10), are shown in Figure 40.2. as
Lastly, Takamizawa and Hayashi18 proposed a loga-
( ( t) = ( *0 e iΕ t and >(t ) = >*0e
i (Ε t % Φ )
rithmic function that again has four coefficients (32)
subjected to mechanical loading. The finite element proce- tor {u}represents these displacements. The finite element
dure essentially involves discretizing a component into a method seeks to minimize the potential energy by selecting
number of small (finite size) regions called the elements, the values of {u} which minimize
For time independent linear problems, the potential ume of the body and {f} is the vector of nodal loads.
energy of the entire system can be minimized in one step
that produces a large number of algebraic simultaneous Element Behavior
equations in the nodal displacements, which are solved us- To calculate elastic strain energy in the body, the ma-
ing a computer. The results obtained using this method are terial domain is discretized into small regions called elements
not exact but with increased model refinement, and subse- (see, for example, Fig. 40.10). The geometry of each element
quently more computational effort, solutions to the approxi- is described by the location of a set of nodes (different ele-
mated displacement field of the desired resolution may ment types have different numbers of nodes) which also
be obtained. serve as inter-element connections.
For the non-linear and time-dependent problems pre- Within each element the displacement field is approxi-
sented in the design of artery grafts, the problem is broken mated by low-order piecewise continuous polynomials, spe-
down into a number of linearized increments. The method cially chosen such that their coefficients are the nodal dis-
therefore requires repeated solution of simultaneous equa- placement components for that element, {#e}, which is a sub-
tions to solve for increments in nodal displacements. How- set of the global displacements, {u}. In each element, the el-
ever, to illustrate the finite element method here, we limit ement strains, {E}, are obtained from the displacements of
the discussion to the linear, time independent problem. the nodes in that element {#e}, by writing Equation (3) as
The finite element method requires: the compatibility requirement
1. That the behavior of the component material be pre-
cisely defined; {E} = B({a e }) {a e }.
[ ] (36)
2. That the geometry of the component in the unloaded
state be known; and The Piola-Kirchhoff stress components in each ele-
3. That the boundary conditions, which include the ment are written as a vector {S}, so that the constitutive re-
mechanical loading, the supports or restraints hold- lation, Equation (10), which accounts for all of the interac-
ing the structure and, for time dependent problems, tions between the stress and strain components in a linear
the initial velocities, are fully characterized. elastic body, can now be written as
From this problem definition the finite element pro-
cedure will be able to analyze the structure and calculate the {S }= [D]{E} (37)
design parameters, such as the deformations and stresses over
the entire component domain. These results will allow deci- where [D] is the square matrix of coefficients
sions on the appropriate material choice and component representing the tensor Dijkl in Equation (12). Essentially,
geometry to be made. The flexibility and predictive power the matrix [D] describes the stresses required to produce
of the finite element method allows engineers the ability to the given strains in the material, and is determined by the
test various design parameters. In the case of a porous graft, strain energy function, W, using Equation (12). The strain
for example, the finite element method is an efficient way to energy W is determined by laboratory testing on the mate-
examine the effect on overall component compliance of rial of interest.
changes in the graft diameter, thickness, material stiffness, Materials such as soft tissue exhibit non-linear time
porosity, pre-stress, etc. dependent behavior and Equation (13) must be written in
an incremental form
Minimizing the Potential Energy
In the continuum description of hyperelastic mate- {dS }= [D({E}, {E& })]{dE} . (38)
rial behavior, it was most convenient to use tensor notation
to describe the field quantities (stresses, strains and displace- Therefore the components of [D] depend on the strain
ments) in a deformable body. However, since the finite ele- {E}and the strain rate {E}. Materials of this type can cause
ment method describes these fields with their values at dis- significant computational difficulties, but the general pro-
crete points (for example, at the nodes for the displacements) cedures are the same as in the linear case.
it is most effective to use matrix notation for the finite ele- For time-independent hyperelastic materials, the elas-
ment equations. tic strain energy in each element, We, is computed by inte-
Since the finite element method computes the dis- grating the hyperelastic strain energy potential W over the
placement field that minimizes the potential energy, the pri- volume of the element, Ve, viz.,
mary variables are the displacements of the nodes. The vec-
454 Tissue Engineering of Prosthetic Vascular Grafts
Ve
e
(40)
design process, a few examples of artery graft modeling are
presented. Figure 40.10 shows the displaced shape of a simple
so that the elastic potential energy (or strain energy) finite element model of the anastomosis region of the natu-
for a single element is given by ral artery and a porous polyurethane graft. The artery
(shaded) is modeled as a single layer, using the simple
e T
W e ae({ })= {a } [k ]{a } 1 e e hyperelastic material model proposed by Fung et al,17 and
2 (41)
does not include any pre-stress. The graft is modeled as a
hyperelastic and isotropic material, using the experimental
Element Assembly data from the uniaxial tensile test data of the porous poly-
To compute the strain-energy for the entire structure, urethane. The graft is analyzed for a range of relative po-
WΗ({u}), the element strain energies, We({#e}), are summed. rosities. The different porosities were created by altering the
This is done by summing the element stiffness matrices, [ke], ratio of the salt to polymer concentration in the manufac-
for each element. Since each set of element displacements, turing process (values of 1:1, 3:1, 5:1 and 7:1 have been in-
{#e}, is a different subset of the global displacements, {u}, vestigated). Both the native artery and the graft are assumed
we must relate the local to the global displacements, which to have an internal diameter (D) of 6 mm and a wall thick-
can be done by defining a locator matrix for each element, ness of 1 mm. The model was loaded with an internal pres-
from which {#e}=[Pe]{u}. This allows us to compute a glo- sure, which is initially set to 70 mm Hg, and then increased
bal stiffness matrix, which represents the structural stiffness linearly to 120 mm Hg—thus )P is 50 mm Hg.
of the entire component. Mathematically, The resultant compliance of the artery and the graft,
as a function of the axial distance, is shown in Figure 40.11.
A simple measure of vascular compliance58
6 2 96 ) D 9
Cv = 8 ; 8 ;, (45)
7 D :7 ) P :
(42)
which relates the change in graft diameter )D to the
Problem Solution change in pressure, is used to compare the compliance of
To compute the total energy of the system, U({u}), we the native artery to the porous graft. This relation is chosen,
require the work done by the loads on the component, which as it does not contain any information about the properties
is merely the inner product of the loads at each node, {f}, of the two materials, which are non-linear and therefore
and the nodal displacements, {u}. Thus, the total energy of cannot be characterized with a single constant parameter.
the system The compliance of the native artery was predicted to be
0.273%/mm Hg (or 13.65% for a pulse pressure of
U ({u}) = 1
2 {u}T [K ]{u}% {u}T {f }. (43) 50 mm Hg), which was greater than any of the four grafts
analyzed. In the region of the anastomosis the compliance
Minimizing the total energy requires the solution of of the artery is affected by the presence of the graft and there-
the (usually large) system of simultaneous linear equations fore reduces towards the junction, which is halfway (5 mm)
along the length. The lowest porosity graft (1:1) results in
[K ]{u}% {f } = {0}, (44) the smallest compliance, which is 0.05%/mm Hg. As the
porosity increases to 7:1, the graft compliance increases to
for which modern computers are highly effective. The approximately 80% that of the host. It is interesting to see
displacements, {u}, which are an approximation to the true that with this more compliant graft material a hyper-com-
An Integral Mathematical Approach to Tissue Engineering of Vascular Grafts 455
pliant zone of about 2 mm, on the arterial side of the anas- The results of the proceeding example are based on a
tomosis, is predicted. In this region the compliance of the static analysis and do not show the influence of the time
artery is increased by approximately 0.4%. A similar zone of dependent behavior of the material. The viscoelasticity of
hyper-compliance was predicted by Chandan et al59 and the polyurethane will result in a gradual increase in the graft
observed in vivo by Hasson et al.60 However, in the in vivo diameter, as a result of strain accumulation during cyclic
study the extent of hyper-compliance was more dramatic, loading. The increase in diameter will reach a steady state
with a localized increase in compliance of up to 50%. after some time. Therefore the inclusion of viscoelasticity is
The graft compliance as a function of the porosity is important in determining the final size of the graft after
shown in Figure 40.12. The result shows that the percentage implantation.
compliance increases exponentially as the porosity increases. The flexibility of the modeling approach means that
However, increase in porosity will also be accompanied by a complex geometrical features, with a host of different
decrease in yield strength of the material. interacting materials, can be simulated. Figure 40.13 shows
Fig. 40.12. Graft compliance as a function of the material porosity (salt:polymer ratio).
An Integral Mathematical Approach to Tissue Engineering of Vascular Grafts 457
9. Hayashi K. Elastic properties and strength of a novel 15. Fung YC, Biomechanics: Circulation 2 nd Edition,
small-diameter compliant polyurethane vascular graft. J Springer,1996.
Biomed Mater Res 1989; 23:229-244. 16. Young JT, Vaishnav RN, Patel DJ. Non-linear anisotro-
10. Deweese JA. Anastomotic neointimal fibrous hyperplasia. pic viscoelastic properties of canine arterial segments. J
In: Complications in vascular surgery, 2nd edition. Or- Biomech 1977; 10:549-559.
lando: Grune and Stratton, 1985:157-170. 17. Fung YC, Fronek K, Patitucci P. Pseudoelasticity of ar-
11. Ballyk PD, Ojha M, Walsh C, Butany J. Suture-induced teries and the choice of its mathematical expression. Am
intramural stresses and intimal hyperplasia. Advances in J Physiol 1979; 237:H620-H631.
Bioengineering ASME BED- 1996; 33:213-214. 18. Takamizawa K, Hayashi K. Strain energy density function
12. Ballyk PD, Walsh C, Ojha M. Effect of intimal thickening and uniform strain hypothesis for arterial mechanics. J
on the stress distribution of an end-to-side graft-artery Biomech 1987; 9:293-300.
junction. Advances in Bioengineering ASME BED- 1995; 19. Cox RH. Anisotropic properties of the canine carotid ar-
31:327-328. tery in vitro. J Biomech 1975; 8:293-300.
13. Paasche PE, Kinley CE, Dolan FG, Gonza ER, Marble AE. 20. Dobrin PB. Biaxial anisotropy of dog carotid artery: Esti-
Consideration of suture line stresses in the selection of mation of circumferential elastic modulus. J Biomech
synthetic grafts for implantation. J Biomech 1973; 1986; 19:351-358.
6:253-259. 21. Tanaka TT, Fung YC. Elastic and inelastic properties of
14. Greer LS, Vito RP, Nerem RM. Material property testing the canine aorta and the variation along the aortic tree. J
of a collagen-smooth muscle cell lattice for the construc- Biomech 1974; 7:357-370.
tion of a bioartificial vascular graft. Advances in Bioengi- 22. Fung YC. Biomechanics: Mechanical properties of living
neering ASME BED 1994; 28:69-70. tissue 2nd Edition, Springer,1993.
458 Tissue Engineering of Prosthetic Vascular Grafts
23. Chuong CJ, Fung YC. Compressibility and constitutive 42. Fung YC, Liu SQ. Strain distribution in small blood ves-
equations of arterial wall in radial compression experi- sels with zero stress state taken into consideration. Am J
ments. J Biomech 1984; 17:35-50. Physiol 1992; 264:H544-H552.
24. Fung YC. Stress-strain history relations of soft tissues in 43. Greenwald SE, Rachev A, Moore JE, Meister JJ. The con-
simple elongation. In: Fung YC, Perrone N, Anliker M, tribution of the structural components of the arterial wall
eds. Biomechanics: Its foundations and objectives to residual strain. Advances in Bioengineering ASME BED
Englewood Cliffs: Prentice-Hall, 1972. 1994; 28:63-64.
25. Hutchinson KJ. Effect of variation of transmural pressure 44. Humphrey JD, Strumpf RK, Yin FCP. Determination of
on the frequency response of isolated segments of canine a constitutive relation for passive myocardium: I. A new
arotid arteries, Cirulation Res 1974; 35:742-751. functional form. J Biomech Eng 1990; 112:333-339.
26. Newman DL, Bowden NLR, Gosling RG. The dynamic 45. Humphrey JD, Strumpf RK, Yin FCP. Determination of
and static elastic properties of the aorta of the dog. a constitutive relation for passive myocardium: II.—
Cardiovasc Res 1975; 9:679-684. Parameter estimation. J Biomech Eng 1990; 112:340-345.
27. Greenwald SE, Newman DL, Denyer HT. Effect of smooth 46. Van Dijk AM. Vascular single smooth muscle cells and
muscle activity on the static and dynamic elastic whole tissue. Mechanical properties and response of
propertries of the rabbit carotid artery. Cardiovasc Res stimuli. PhD Thesis University of Leiden, Netherlands:
1982; 16:86-94. 1983.
28. Langewouters GJ, Wesseling KH, Goedhard WJA. The 47. Gibson LJ, Ashby MF. Cellular solids: Structure and prop-
static elastic properties of 45 human thoracic and 20 ab- erties, Pergamon Press, 1988.
dominal aortas in vitro and the parameters of a new 48. Carmines DV, McElhaney JH, Stack R. A piece-wise non-
model. J Biomech 1984; 17:425-435. linear elastic stress expression of human and pig coro-
29. Langewouters GJ, Wesseling KH, Goedhard WJA. The nary arteries tested in vitro. J Biomech 1991; 24:899-906.
pressure dependent dynamic elasticity of 35 thoracic and 49. Hayashi K. Experimental approaches on measuring the
16 abdominal human aortas in vitro described by a five mechanical properties and constitutive laws of arterial
component model. J Biomech 1985; 18:613-620. walls. J Biomech Eng 1993; 115:481-487.
30. Haut RC, Little RW. A constitutive equation for collagen 50. McAfee MA, Kaufmann MV, Simon BR, Baldwin AL.
fibres. J Biomech 1972; 5:423-430. Experimental/numerical approach to the determination of
31. Dehoff PH. On the nonlinear viscoelastic behavior of soft material properties of large arteries. Advances in Bioengi-
biological tissues. J Biomech 1978; 11:35-40. neering ASME BED 1994; 28:111-112.
32. Decraemer WF, Maes MA, Van Huyes VJ, Van 51. Vaishnav RN, Young JT, Patel DJ. Distribution of stresses
Peperstraete P. A non-linear viscoelastic constitutive equa- and of strain-energy-density through the wall thickness
tion for soft biological tissues. J Biomech 1980; in a canine aortic segment. Circ Res 1973; 32:577-583.
13:559-564. 52. von Maltzahn WW, Warriyar RG, Kietzer WF. Experimen-
33. Wu SG, Lee GC. On nonlinear viscoelastic properties of tal determination of the elastic properties of media and
arterial tissue. ASME J Biomech Eng 1984; 106:42-47. adventitia of bovine carotid arteries. J Biomech 1984;
34. Demiray H. A quasi-linear constitutive relation for arte- 17:839-847.
rial wall materials. J Biomech 1996; 29:1011-1014. 53. von Maltzahn WW, Besdo D, Wiemer W. Elastic proper-
35. Johnson GA, Livesay GA, Woo SL-Y, Rajagopal KR. A ties of arteries: A non-linear two-layer cylindrical model.
single integral finite strain viscoelastic model of ligaments J Biomech 1981; 14:389-397.
and tendons. J Biomech Eng 1996; 118:221-226. 54. Ferry JD. Viscoelastic properties of polymers. New York:
36. Lanir Y. A microstructural model for the rheology of Wiley and Sons, 1961.
mammalian tendon. J Biomech Eng 1980; 102:332-339. 55. Lockett FJ. Nonlinear Viscoelastic Solids. New York: Aca-
37. Lanir Y. Constitutive equations for fibrous connective tis- demic Press, 1972.
sue. J Biomech 1983; 16:1-12. 56. Christensen RM. Theory of Viscoelasticity: An Introduc-
38. Minns RJ, Soden PD, Jackson DS. The role of the fibrous tion, New York: Academic Press, 1971.
components and ground substance in the mechanical 57. Sternstein SS. Mechanical properties of glassy polymers:
properties of biological tissues: A preliminary investiga- Treatise on Materials Science and Technology. In: Schultz
tion. J Biomech 1973; 6:153-165. JM, ed. Volume 10, part B, New York: Academic Press
39. Huyghe JM, Van Campen DH, Arts T, Heethaar RM. The 58. Gow BS, Taylor MG. Measurement of viscoelastic prop-
constitutive behavior of passive heart muscle tissue: A erties of arteries in the living dog. Circ Res 1968;
quasi-linear viscoelastic formulation. J Biomech 1991; 23:111-122.
24:841-849. 59. Chanran KB, Gao D, Han G, Baraniewski H, Corson JD.
40. Myers BS, McElhaney JH, Doherty BJ. The viscoelastic Finite element analysis of arterial anastomoses with vein,
responses of the human cervical spine in torsion: Experi- Dacron and PTFE grafts. Med Biol Eng Comp 1992;
mental limitations of quasi-linear theory, and a method 30:413-418.
for reducing these effects. J Biomech 1991; 24:811-817. 60. Hasson JE, Megerman J, Abbott WA. Increased compli-
41. Chuong CJ, Fung YC. On residual stresses in arteries. J ance near vascular anastomosis. J Vasc Surg 1985;
Biomech Eng 1986; 108:189-192. 2:419-423.
Scaffold Engineering
Material Aspects
CHAPTER 41
Bioinertness: An Outdated Principle
David F. Williams
B iomaterials have been with us for the majority of the twentieth century. Their nature has
evolved during this time, and the applications for which they have been used have in-
creased in complexity and diversity. However, for much of this time, the functions required
of these materials and the performance parameters of the medical devices in which they
have been used have been relatively straightforward and largely confined to mechanical and
physical characteristics. The selection and design of biomaterials have therefore been simi-
larly constrained by the conventional engineering concepts underlying these applications.
During the last few years the situation has changed quite radically as new concepts and new
treatment modalities have required a fundamental reappraisal of the scientific principles
upon which biomaterials science are based. Nowhere is this seen more vividly than in the
use of biomaterials for implantable devices within the cardiovascular system, and specifi-
cally in relation to the application of tissue engineering concepts in reconstructive vascular
surgery. In this chapter we attempt to provide a rationale for this fundamental change and
to demonstrate the pivotal role of surface reactivity in the products and devices of the future.
If we consider the historical role of biomaterials in implantable devices we can iden-
tify several clinical conditions that underlie their use, including congenital and develop-
mental defects and trauma, but must recognize that it is in the treatment of diseases which
have caused irreversible changes in tissues and organs that biomaterials play their biggest
part. Thus, we see the widespread use of total joint replacements associated with the very
widespread incidence of osteoarthritis, of intraocular lenses necessitated by cataracts, of
prosthetic heart valves in the treatment of valvular disease, of dental implants following
tooth and periodontal destruction, and of vascular prostheses and devices used for the pros-
thetic reconstruction of arteries.
In all of these situations the functional requirements of the materials are simple and
essentially nonbiological. The major implantable devices of the twentieth century either
transmit loads, facilitate sliding motion, passively direct fluid flow, control fluid flow or
simply fill a space. The materials that permit the devices to perform these functions need to
have the appropriate mechanical properties and little else. Indeed, in the majority of these
cases, the mechanical and physical requirements are not themselves very onerous, and a
wide range of materials is theoretically available for the construction of these devices.
In reality, of course, the situation is not quite so straightforward, since the devices
usually have to perform their function for a long period of time within the confines of the
human body. This implies that the materials have to supply these mechanical or physical
characteristics without interference from the hostile physiological environment and without
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
460 Tissue Engineering of Prosthetic Vascular Grafts
causing any untoward or adverse effects in the patient. The There are three fundamental reasons why bioinertness
latter two aspects are subsumed within the phenomenon of may not necessarily be the sensible choice. The first is that
biocompatibility, and it has been a combination of the me- complete inertness is unachievable, so that any strategy
chanical and physical properties together with predicated on this quality is ultimately doomed to fail. Sec-
biocompatibility considerations that have determined the ondly, if a device is made from materials which are inert
selection of materials available for implantable medical de- and which do not interact with the body in any way, then it
vices at the present time. is unlikely that it can be truly incorporated into the body.
The term biocompatibility sounds simple to interpret For effective, long term performance in the dynamic tissue
since, as alluded to above, it implies compatibility or har- environment, it is usually preferable for there to be func-
mony with living systems. This concept, however, is too tional incorporation, which implies that the device should
simple to be useful and the meaning of compatibility has to be stimulating the tissues to react to it positively rather than
be explored further, both to understand the real logic of permitting them to ignore it. Thirdly, the emergence of tis-
today’s material selection and the opportunities for the fu- sue engineering principles, in which biomaterials are usu-
ture. It is intuitively obvious that a biomaterial or an im- ally seen to act in a supporting role to biological compo-
planted medical device should cause no harm to the recipi- nents, has placed a very considerable emphasis on the utili-
ent. This is the underlying principle of biological safety, and zation of intentionally degradable materials. These three fea-
it is this rather than the totality of biocompatibility which tures, which are determining the changing emphasis away
has shaped the historical evolution of biomaterials. Most from bioinertness, are explained below.
crucially, the requirements of biomaterials have been domi-
nated by the perceived necessity to be safe, which has usu- The Impossibility of Inertness
ally been interpreted as a requirement that a biomaterial The convention described above is based on the as-
should be totally inert in the physiological environment and sumption that a prosthetic device replaces, augments or
should itself exert no effect on that environment. In other modifies tissues or organs and is intended to remain within
words, there should be no interaction between biomaterials the patient for the length of time that the function is re-
and their host, implying that the material should be non- quired, typically for the natural lifetime of that patient. The
toxic, nonirritant, nonallergenic, noncarcinogenic, non- inertness was considered to be of crucial importance, both
thrombogenic and so on. to allow the device to retain its structural integrity and
This concept of biocompatibility which equates “bio- thereby maintain its performance, and to prevent or at least
logical performance” to inertness and biological indifference limit the release of any substance or component from the
has resulted in the selection of a portfolio of acceptable or material which would have some toxicological or other ad-
standard biomaterials that have widespread usage in appli- verse biological consequence. In the vast majority of circum-
cations such as those mentioned above. These range from stances, sufficient control has been exercised over the chemi-
the passive alloys such as stainless steel and titanium alloys, cal stability of biomaterials to ensure that no catastrophic
the noble metals of gold and platinum, some oxide ceram- loss of that integrity occurs as a result of compromised
ics such as alumina and zirconia, various forms of carbon bioinertness. There have been some exceptions and there
and a range of putatively stable polymeric materials includ- are no grounds for undue complacency in this respect, but
ing silicone elastomers, polyolefins, fluorocarbon polymers this is generally not the most important of considerations.
and some acrylics and polyesters. The precise material cho- On the other hand, the potential to initiate harmful biologi-
sen for an application from this type of list would depend cal processes in the host will always be finite as long as there
on the precise mechanical or physical property specifications. are structures capable of degradation or components free to
It cannot be denied that this approach has led to the be released.
successful deployment of many types of prostheses in wide- Several features of the inherent structure of materials
ranging clinical areas, and, apart from a few notable and and of the complex nature of the tissue environment aggra-
controversial situations, such implantable devices rarely vate this situation. In particular, inertness in the physiologi-
impact adversely on the health of the patient. The concept cal sense requires a great deal more than resisting degrada-
of bioinertness has therefore served a useful purpose. Even tion at the atomic or molecular level and, furthermore, even
when we look at these generally successful areas, however, it if it were that straightforward it would be extremely diffi-
can be seen that there are significant limitations to their per- cult to achieve. Indeed, it is now recognized that no material
formance and to the type of patient in which they can be is totally inert in the body. Even those very stable materials
used with confidence. Total joint replacements are generally mentioned earlier will interact to some extent with tissues.
contra-indicated in the younger, more active patient; the Titanium, although one of the most corrosion-resistant en-
performance of vascular prostheses decreases as the devices gineering alloys, corrodes in the body, judging by the pres-
are used more distally; bioprosthetic heart valves give greater ence of the metal in surrounding tissues. Gold and plati-
cause for concern in very young patients; and intraocular num will interact electrochemically with the saline-based
lenses may not give satisfactory results in patients with un- extracellular fluid. For reasons not entirely understood, the
derlying inflammatory conditions. Questions may be raised, most inert of the oxide ceramics suffer some long term
therefore, as to whether bioinertness is the most acceptable changes within the body, and almost all known polymers
principle underpinning material selection or whether alter- will undergo oxidation, hydrolysis or other changes upon
native concepts would be preferable. implantation.
Bioinertness: An Outdated Principle 461
With many materials, while the main component it- that there is no such thing as an inert biomaterial. It is never
self may be exceptionally inert, there are often minor com- a question of whether a biomaterial will interact with the
ponents, perhaps impurities or additives, which can be re- body but rather when and how. Under these circumstances,
leased under some circumstances. The leaching of plasticiz- the preferred alternative strategy is to accept that such in-
ers and other additives from plastics provide good examples teractions take place and to attempt to control these inter-
of interactions which are not necessarily related to the mo- actions proactively and to incorporate these interactions into
lecular breakdown of the material, but which nevertheless the design specifications.
confer a degree of instability to the product.
It should also be noted that descriptions of material The Requirement for Controlled Reactivity
degradation mechanisms have to take into account the spe- The above arguments indicate that complete inertness
cial and indeed, unique, features of the tissue environment. may not be possible, but still do not indicate that it is unde-
Whatever its location, a biomaterial will continuously en- sirable. The possibility that the products of any interaction
counter an aqueous environment during its use. This is not between a biomaterial and its physiological environment
simply a saline solution, however, but a complex solution could be released into the host has generally been consid-
containing a variety of anions and cations, a variety of large ered a sufficient deterrent to utilizing any material that was
molecules, some of which are very reactive chemically, and significantly reactive in that environment. This logic is clearly
a variety of cells. There are occasions when a degradation only valid if those products were going to initiate an unde-
process can be explained, mechanistically and qualitatively, sirable response from the host, either locally or systemically.
by the presence of electrolyte. This is the situation with most If interactions took place whereby the products were totally
metals when they suffer from corrosion in a physiological harmless, then there would be less cause for concern over
environment. Even here, however, it is known that the ki- the inability to achieve inertness. More importantly, if the
netics of corrosion may be influenced by the organic species nature of the interaction were one which produced a more
present, especially the proteins, and it is indeed possible for appropriate response from a tissue, then a positive virtue
the corrosion mechanism to be somewhat different from that could be made of this lack of inertness.
found in nonbiological situations. This quite different thinking is now enshrined in the
This phenomenon is even more pronounced with current concepts and definition of biocompatibility. Instead
other groups of materials and it is clear that with polymers of biocompatibility being equated with inertness, it is now
the kinetics and mechanism of degradation are fundamen- recognized that it should encompass a wide range of reac-
tally related to the precise details of the environment. Al- tivity, with the caveat that any reactivity is beneficial rather
though hydrolysis remains the substantive mechanism for than harmful. On the basis of these ideas biocompatibility
degradation of most heterochain polymers, including polya- was redefined a few years ago as “the ability to perform with
mides and polyesters, this hydrolysis may be profoundly in- an appropriate host response in a specific application”. It is
fluenced by the active species present in the tissue. In par- apparent that this definition still encompasses the situation
ticular, the lysosomal enzymes synthesized and released from where inertness is required, since the most appropriate re-
cells of the inflammatory response to biomaterials may in- sponse in some situations is indeed no response. A tradi-
fluence the degradation process. Moreover, the hydrolysis tional bone fracture plate is most effective when it is attached
may be supplemented by oxidative degradation, again oc- mechanically to the bone and does not corrode. No response
curring not only by virtue of passively dissolved oxygen in of the tissue to the material is required under these
body fluids, but by active oxidative species such as superox- circumstances.
ides, peroxides and free radicals generated by activated in- In the type of device mentioned earlier in this chap-
flammatory cells such as macrophages. It is thus possible ter, in which the performance is dependent upon its physi-
for homochain polymers not particularly susceptible to hy- cal replacement of diseased tissues and its incorporation into
drolysis and not normally oxidized at room temperature to the structure of the body, inertness of all of the components
undergo oxidative degradation upon implantation. may prevent optimal performance from being achieved. In
Polyolefins such as polyethylene and polypropylene come particular, if a material is inert and unreactive within tis-
into this category. sues, the long term host response will be associated with a
On the basis of a vast amount of experimental work lack of recognition and a lack of functional incorporation.
and clinical experience, it is now clear that all biomaterials An inert polymer such as polyethelyne or PTFE will induce
are inherently susceptible to some degradation process the formation around it of a thin layer of collagenous fi-
within the physiological environment. It is equally clear that brous tissue which can neither facilitate incorporation of
although a variety of surface treatment methodologies are the device into the tissue nor assist the device in achieving
available to reduce or ameliorate the degradation, none of any of its functions. Moreover, this fibrous layer is unlikely
these are entirely effective and their availability does not to be stable and may alter its characteristics over time, this
negate the now accepted principle that complete inertness often being the ultimate cause of the device failure. In such
cannot be achieved. Moreover, in the context of interactions circumstances, the biocompatibility characteristics of the
which affect the overall performance of the material in the materials in contact with their host tissues should be those
physiological environment, it is important to note that an which favor a positive interaction between the molecules of
interfacial reaction involving a physicochemical process such the material and the relevant molecules of that tissue, such
as protein adsorption will inevitably take place, further em- that there is a functional attachment between the two. A to-
phasizing the fact that inertness is a very relative term and tal joint replacement which has the nonarticulating surfaces
462 Tissue Engineering of Prosthetic Vascular Grafts
composed of materials or substances that are able to inter- with the delivery of active tissue repair promoting molecules
act with the cellular components that promote bone regen- and with the physical support necessary for the developing
eration at the interface should yield a more appropriate host tissue structure. It goes without saying of course that such a
response in the context of the long term mechanical stabil- material should not degrade to produce harmful side effects,
ity of the joint. Similar arguments could be put forward for which could be associated with either the chemical charac-
many of today’s implantable devices that are used in recon- teristics of any degradation products or their physical mor-
structive surgery. phology. Although this is not a trivial matter, such that in-
This concept may be extended to many other types of flammatory reactions associated with the degradation can-
implantable device, including those which function through not be ruled out, there are several biodegradable polymers
an intervention in healing processes. For example, whilst it which may now be used with reasonable confidence in these
was noted before that conventional fracture plates need to situations.
be inert, on the basis that degradation or corrosion prod- Tissue engineering products should, however, involve
ucts could be harmful to the local tissue, it is not impossible more than a degradable structural polymer. The real essence
for such devices to be made with surfaces that interact with of such a product is the biological activity which it imparts
osteoblasts, thus actively promoting or accelerating the heal- to the host site. A simple, empty, porous scaffold of a de-
ing process. Similarly, while many intravascular stents are gradable polymer implanted within connective tissue may
fabricated from alloys chosen on the basis of their corrosion become infiltrated with repair tissue and may ultimately
resistance and general inertness, the application of substances degrade to leave an area of reconstituted tissue derived from
to the surfaces of these alloys which potentially have an ability that infiltration. This, however, is not tissue engineering,
to interact at the molecular level with those agents that are since the tissue will not have been directed or controlled in
responsible for restenosis provides a new concept of terms of its structure and nature. In order to be really use-
bioreactivity rather than bioinertness for these devices. ful, this product would have to incorporate the appropriate
Almost every type of implantable device which has cellular components, normally previously derived from the
hitherto been associated with materials chosen for their ap- hosts, or certain molecules that are able to signal to the host
parent inertness may now be reconsidered in this light. This cells the appropriate instructions to produce the desired tis-
applies equally to devices where active surface molecules can sue morphology and function. Under these circumstances,
control thrombogenicity, to devices which are able to en- the biodegradable polymer has to be chosen very carefully,
courage tissue repair and to devices which are able to mini- such that at the very least it will be compatible with the de-
mize the undesirable consequences of inflammation. sired cell function in its vicinity or, preferably, such that it is
capable of delivering these cell signaling molecules with the
The Essential Requirement of Reactivity desired activity and in the desired manner. Inherent inert-
in Tissue Engineering ness is therefore unacceptable in these tissue-engineered
The desirability of some degree of reactivity in con- products. The challenge is to introduce the desired level of
ventional implantable devices can be seen as a precursor to reactivity without compromising the biological safety of the
the essentiality of reactivity associated with the concepts and material.
products of tissue engineering. It is important to remember
here that the underlying principle of tissue engineering in- Conclusions
volves the combination of biological principles and sub- For all the reasons outlined above, bioinertness can
stances with medical engineering principles and devices in be considered as an outdated principle. Although there are
order to provide products that are able to persuade the body some types of implantable device which are still better served
to heal itself. In this rapidly emerging field, the major prod- by inert materials, the majority should preferably incorpo-
ucts utilize some material structure as a support, matrix or rate intentionally interactive materials, whilst others, espe-
vehicle for the delivery of active biological molecules or cel- cially in this new area of tissue engineering, positively de-
lular species to a target site in the host where repair or re- mand this characteristic.
construction is to be effected. Important as these material It has to be said, however, that academic arguments in
structures are, they do not normally constitute a compo- favor of replacing a concept that is perceived to be out of
nent that is intended to remain in the body for very long, date with a new principle cannot be sustained unless this
and certainly their function is incompatible with the char- new principle can be verified and reduced to practice. In
acteristic of total inertness. view of the fact that bioinertness has never produced any
There are several different levels at which reactivity is harm, it is indeed a brave new world which is predicated on
desirable or essential. As alluded to above, the most obvious the availability of biomaterials that are both functionally
of these is the desirability for intentional biodegradation in interactive with their host and intrinsically safe. Many might
the polymeric support structures that form the basis of tis- argue that these in fact are contradictory requirements.
sue-engineered reconstruction devices. Whether this is con- Clearly that need not be the case, but the whole future of
cerned with the repair of cartilage defects, of nerve tissue, of tissue engineering products that are able to demonstrate ef-
damaged skin or diseased tissue within the vascular system, ficacy and safety is dependent on the development of the
the template of a device usually involves a polymer that can appropriate materials. However outdated bioinertness may
degrade over a period of time ranging from a few weeks to a be, it should not be replaced by materials based upon hope
few years and where the degradation profile is consistent rather than scientific reality.
Scaffold Engineering
Material Aspects
CHAPTER 42
Bioinert Biomaterials: Are Their Properties Irreplaceable?
Patrick T. Cahalan
n 1955 Sewell and colleagues1 performed a study comparing ovine and bovine sources of
I catgut sutures in three animal models. The objective of the study was to quantitatively
compare the tissue response to the implanted material, and this event is often cited as the
beginning of biocompatibility testing. Coincident to this was the rapidly expanding postwar
technology in high performance plastics, with mounting evidence that biological grafts, even
homografts, were not going to survive the challenges in vascular graft applications.2 Voorhees’
observations in 1952 of a neointima free of thrombi on a silk thread, and his later experi-
ments with Vinyon “N” cloth in dog aortas, clearly showed that a synthetic material could
serve as a conduit, and be accepted by the tissue.3 Studies would follow showing the merits
of other materials, particularly Teflon and Dacron.4 The paradigm was set, and materials
were classified as very reactive to inert.
What followed was once described as “20 years of frustration”.5 During this period
commercial efforts were driven by the promise of a very large market, estimated between 0.6
to 1 billion dollars annually. Scientific efforts were focused on comparing properties of ma-
terials with limited physiological markers, primarily the occurrence of thrombi on the ma-
terial surface. An example of surface property concepts that were hypothesized to be benefi-
cial are seen in Figure 42.1.6 The majority of the surfaces listed are in essence attempts to
create surfaces with minimal effect, whether it be termed low protein or platelet adhesion.
In the same 1972 article, Baier refers to much of the individual surface property character-
ization towards blood compatibility as people working in a maze within a maze.
Hydrophilic surfaces, for example, while showing low platelet adhesion and therefore
thought to be blood compatible, when tried in a vascular graft position did not prove to
provide patency. Thus, researchers thinking they had found a way out of the blood compat-
ibility maze found themselves in another maze with new barriers to solve. Baier’s represen-
tation of the surface properties compared to the living blood vessel intima serves to high-
light the focus of blood compatibility to the vascular application. This narrow focus, plus
the lack of clearer understanding of cellular and molecular biology, helped continue the
quest for the “holy grail” of inert materials.
Not until it was shown in the late 1970s that endothelial cells were more active in
maintaining blood compatibility7 did the paradigm begin to change. Until this time it was
even possible to hypothesize that endothelial cells were the perfect inert surface. The emer-
gence of RIA and ELISA in the mid-70s opened up the possibilities of looking at the mo-
lecular and cellular aspects involved at the surface. The common use of these tools did not
make its way into blood compatibility research until well into the 1980s, and today a signifi-
cant number of papers are still offered that rely largely on platelet adhesion alone to deter-
mine blood compatibility of surfaces. The increase in understanding of reactions between
coagulation factors and formed elements of the blood has led to a change in thinking that
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
464 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 42.1. General approaches to improve blood compatibility: early years (70s).
previously focused on the bulk reactions in blood to the re- explanation for this failure may be the nondiscriminatory
alization that these critical reactions “occur almost exclu- nature of some of the cell adhesion receptors to adhesion
sively on surfaces”.8 The formerly thought to be inert endot- proteins. Some new approaches to deal with the nondiscrimi-
helial cell has been shown to have as many procoagulant natory cell receptor binding to adhesion proteins attempt
properties as anticoagulant.9 The pat answer for years to the to attach peptide adhesion ligands to surfaces.12 A critical
question, “What is the perfect surface?” was: “An requirement stated by Hubbell in such attachment is to ren-
endothelialized surface.” The fact that an endothelialized der the surfaces resistant to protein adsorption by first graft-
surface can be thrombogenic opens up a new maze. It also ing with a hydrophilic nonionic polyethylene oxide (PEO)
begs the question, “Bioinert biomaterials: Are their proper- chain to which the peptide ligand could be tethered.
ties irreplaceable?” The latter stated requirement could serve to finally
“The long sought after biologically inert material has propose a more pragmatic application of the “inert” con-
not as yet been developed, and the realization of the inher- cept. Inertness with respect to a nonionic hydrophilic spacer
ent ‘biointeractivity’ of all implanted polymers has led many that is resistant to protein adhesion permits a selectivity or
investigators to pursue strategies aimed at optimizing the control of molecular and cellular events at surfaces. The chal-
biological interactions with the synthetic polymer rather lenges left with this approach are to achieve the proper den-
than to minimize all biological interaction.”10 This statement sity of ligands on the surface, their orientation, and the abil-
would seem to put an end to the question, “Is there a need ity to accomplish this on synthetic substrates in the geomet-
for bioinert materials?” Unfortunately, vascular grafts and ric configuration of a biomedical device such as a vascular
other biomedical devices still use synthetic materials of con- graft, artificial heart, heart valve, or ventricular assist de-
struction, and their replacement with other biological ma- vice. While this elegant concept represents a major advance
terials or repair therapies is not on the very near horizon. in the effort to make an endothelialized and hopefully long
The great promise of biodegradable materials has not been term blood-compatible surface on a vascular graft, it is not
realized, and attempts at endothelialization of synthetic certain whether another maze will not open up, such as con-
materials, while showing promise, have not consistently ex- tinued hyperplasia. Due to noncompliance of synthetic
ceeded the performance of autologous tissue. Preadsorption structures, some might argue that a truly new totally func-
of synthetic vascular grafts with cellular adhesive proteins tioning vascular tissue might not be possible, and that even
tends to enhance the short term attachment of endothelial though there is an endothelial layer on the lumen, the struc-
cells, but platelet adhesion, and therefore increased ture will succumb to other mechanisms of failure. More
thrombogenicity, typically increases with time.11 A possible optimistically, it could be hoped that compliant structures
Bioinert Biomaterials: Are Their Properties Irreplaceable? 465
could be engineered that also could be modified to promote as optimal pretreatments followed by confirming XPS and
spatially correct cellularization of smooth muscle cells, and TofSIMS data is required to assure optimal presentation of
even include neovascularization. The progress made by per- PC moieties on the surface before testing. There is support-
sistent researchers10,12 in the vascular graft field suggests that ing evidence for the inertness of a pure PC surface.19 Stud-
the latter possibility is not out of the question. ies performed at the University of Maastricht have demon-
As mentioned earlier, the vascular graft focus may have strated that a phospholipid bilayer of pure dioleoyl-phospha-
actually been in part responsible for a lack of more rapid tidylcholine (DOPC) on an ellipsometry plate is indeed an
progress in understanding the cellular and molecular events inert surface, and assembly of the prothrombinase complex
at the surface, particularly in development of more blood- requires the addition of dioleoyl-phosphatidylserine (DOPS)
compatible surfaces. Other application areas such as dialy- in significant amounts.28 DOPS is present on the internal
sis and cardiopulmonary bypass (CPB) have created a strong surface of the platelet membrane, and platelet activation
research effort in providing surfaces with enhanced blood causes a flip-flop of the DOPS to the external surface of the
compatibility, and in this effort industry, academia, and cli- membrane. When DOPS is on the surface, the
nicians have invested large amounts of time and capital in prothrombinase complex can be assembled and thrombin
investigating improved biocompatible surfaces on devices generation can take place. Unfortunately, the stability of a
not necessarily designed for permanent implant. From a DOPC bilayer is very poor, and at this time has not been
strictly clinical perspective, the ability to monitor coagula- attained with consistency on a polymeric surface. The MPC
tion and inflammatory responses to foreign surfaces during polymer approach would be to assure uniformity and den-
CPB, and evaluate devices after use, has created a plethora sity of the PC moiety on the surface, and thus impart through
of scientific publications involving measurement of molecu- mimicry an inertness to the surface.
lar and cellular events on blood-contacting surfaces. This While the DOPC surface appears to be the closest thing
has also created a commercial stimulus for funding research to an inert surface, the actual surface of the plasma mem-
on blood materials interactions and research in surface brane is in fact made of more than just DOPC. Transmem-
modification of biomedical devices. brane proteins assure that platelets in the end are very reac-
In our laboratory, surfaces deemed “inert” have been tive with their environment. It is perhaps appropriate to re-
studied using multiple ELISA markers such as TAT, F1.2, flect back to the comments of Spaet8 with respect to all im-
elastase, and SC5b-9 (terminal complement complex) in portant reactions in the body taking place on surfaces, and
addition to measuring platelet adhesion and activation. The that these surfaces we have learned to be anything but inert.
most often cited inert molecule is poly(ethylene glycol) or In the end, one of the roles or properties of inert
(PEG). It has been suggested by some that a PEG surface has biomaterials that is needed is their ability to enhance the
little or no protein adsorption or interaction.13,14 This expression of another molecule, such as a peptide as sug-
premise has also been challenged by some researchers.15,16 gested by Hubbell. This may be accomplished by minimiz-
It has further been suggested that PEG surfaces may serve to ing competing phenomena such as protein adsorption, as
delay rather than reduce protein adsorption and activation.17 well as presenting a molecule in a conformation more ac-
With respect to blood compatibility, we found that while a cessible to its target receptor. This property is indeed possi-
PEG surface does in fact show lower protein adsorption and bly irreplaceable. Our studies specific to the attachment of
platelet adhesion, it does not show any improvement in TAT, heparin to surfaces clearly indicates that the bioactivity of
complement (SC5b-9), or elastase. These studies used a an attached molecule is dependent on its ability to interact
blood loop system that contained a valve to effect physiologi- with target molecules, and the ability to control the loading
cal flow, and human blood containing 1 U/ml of heparin. and the reactivity of the attached molecules is critical to
While PEG surfaces have been studied and widely reported achieve optimal functioning on the surface.20 Heparin on
to approach inertness, it is obvious that there is yet much to these surfaces was tethered to a nonionic hydrogel that was
be learned before this technology is optimized. Neverthe- derivatized to achieve different loadings of heparin. Here
less, the suggested use of this molecule to tether a peptide, the surface is certainly not inert, but it is designed to opti-
or possibly a specific adhesion molecule, may in the end mally compete with the undesirable physiological mecha-
prove to be the best application of PEG. nisms of coagulation and platelet adhesion. These surfaces
Another molecule that is receiving considerable atten- do not prevent protein adhesion, but the proteins that have
tion for its low protein and platelet adhesion is phosphatidyl- adhered show different relative concentrations to control
choline (PC). Methacrylate copolymers with pendant PC surfaces, and the performance of the surfaces correlates with
groups (MPC) have been coated with a facile process to poly- these adhesion profiles.21 In nature, a denuded endothelial
mers, and the PC moiety is expressed to a high degree on lining exposes a procoagulant surface, yet this surface can
the uppermost surface.18 Conceptually, the MPC coating is rapidly passivate without excessive thrombin formation. Fi-
said to mimic the natural phosphatidylcholine present on brinolysis, controlled inflammation and, finally, remodel-
the surface of nonactivated platelets. In testing in our labo- ing give a normal healing response that restores the tissue to
ratory, this surface does show reduced platelet adhesion and a functional state, barring the introduction of complications
less TAT formation. Additional testing needs to be completed of a biomaterial, ongoing disease state or flow restrictions.
to evaluate the complement and white cell response to this A normal healing response replaces inertness with spatially
surface. In our hands this polymer has shown material-de- correct signals via the subendothelial extracellular matrix
pendent coating characteristics, and evaluation is tedious, and thus controls the first step of repair, which is platelet
466 Tissue Engineering of Prosthetic Vascular Grafts
adhesion and coagulation. A naturally formed and controlled of these materials can easily be controlled by coupling the
hemostatic response (clot) presumably is resolved without release of anti-inflammatory drugs; this may possibly open
the excessive inflammatory reactions associated with the up yet another maze.
presence of foreign materials. If those foreign materials pos- In summary, the 20 years of frustration27 to which
sess an inert surface, the resultant histology should resemble Andrade referred have been replaced by numerous
that of a injury without the presence of a foreign material. multidisciplined efforts, aided by a much better understand-
Since injury exposes the blood to nonendothelialized sur- ing of molecular and cellular events at surfaces. The knowl-
faces, and these surfaces do bind plasma proteins, it might edge gained has opened up many approaches to finding the
be a better approach to attempt to modify a surface to have holy grail, and also created a number of positive spin-offs in
a similar protein adsorption profile. Implants of biomate- therapies apart from vascular grafts. The concept of bioinert
rial surfaces modified with collagen (data to be published) materials still persists, not as an end in itself, but rather as a
after 10 weeks show minimal capsule formation with mi- tool to enhance positive mechanisms or to retard undesir-
crovascular structures within two fibroblast layers of the bio- able competing mechanisms. In the quest for new
material surface. The control biomaterial surface (polyure- biomaterials, and for tissue engineering to replace diseased
thane) has macrophage and foreign body giant cells (two tissue with new functioning tissue, we will surely progress
layers) followed by 12-15 layers of densely packed fibroblasts in an iterative manner. Mundane issues such as sterilization
adjacent to the material surface (a typical fibrotic response). may render new concepts nonfunctional, but may stimulate
The attachment of the collagen to the above mentioned sur- new sterilization technology in order to bring them to mar-
face was not simple adsorption, but rather a covalent at- ket. Mechanical property limitations may require creative
tachment with isolation of the collagen layer to the upper- processing or fabrication advances. Promising surface modi-
most layer (70 Å).22 XPS data showed a pure surface of col- fications may first produce composite hybrid structures with
lagen, and this was attainable based on the inherent proper- traditional materials that can later be replaced by resorbable
ties of the grafted surface beneath. This should be a target materials. What in the end is probably irreplaceable is the
for modification of surfaces whether inert or interactive, and learning process involved in trying to find inert biomaterials.
that being complete and uniform coverage without influ-
ence from intermediate chemistries used to couple the mol- References
ecules to the surface. 1. Sewell WR, Wiland J, Craver BN. A new method of com-
In addition to the property of inertness being used to paring suture of ovine catgut with sutures of bovine cat-
enhance the presentation of molecular species to the physi- gut in three species. Surg Gynecol Obstet 1955; 100:483.
ological environment, there is another possibly irreplace- 2. Szilagyi DD, McDonald RT, Smith RF et al. Biological fate
of human arterial homografts. Arch Surg 1957; 75:506.
able property of inertness. That is the relative chemical in-
3. Voorhees AB, Jaretski A, Blakemore AH. The use of tubes
ertness of biomaterials. In addition to finding the optimal constructed from Vinyon “N” cloth in bridging arterial
protein adhesion mosaic on a surface, the biomaterial of defects. Ann Surg 1952; 135:332.
construction of a device must withstand the degradative 4. Deterling RA, Bhonslay SB. An evaluation of synthetic
environment of the body. Numerous attempts have been materials and fabrics suitable for blood vessel replacement.
made, for example, to fabricate vascular prostheses using Surgery 1955; 38:71.
polyurethane, when several instances of urethane degrada- 5. Andrade JD, Nagaoka S, Cooper S, Okano T, Kim SW.
tion in vivo have been documented.23,24 Attempts to mimic Surfaces and blood compatibility: Current hypotheses. Vol
the in vivo response in an in vitro system have shown that XXXIII Trans Am Soc Artif Organs 1987.
adsorption of the plasma protein #2-macroglobulin could 6. Baier RE. The role of surface energy in thrombogenesis,
Bulletin of The NY Academy of Medicine 1972;
play a role in biodegradation of the urethane.25 It might be
48(2):257-272.
hypothesized that a surface modification that could isolate 7. Weskler BB, Marcus AJ, Jaffe EA. Synthesis of prostag-
the biomaterial surface from protein and cell attachment landin 12 (prostacyclin) by cultured human and bovine
could serve to protect its stability. Those who would choose endothelial cells. Proc Natl Acad Sci USA 1977; 74:
to continue the use of polyurethanes for vascular grafts will S.3922-3926.
need to deal with the potential degradation mechanisms 8. Spaet TH. Blood in contact with artificial surfaces: Where
demonstrated. An alternative approach to elastomeric ma- have we been and where are we going. Annals of The New
terials may be fabricated grafts using very stable materials York Academy of Sciences, 1987; 516.
such as PTFE and PET that have mechanical properties cre- 9. Gimbrone Jr MA. Vascular endothelium: Nature’s blood
ated by the bulk design of the device. container. Vascular Endothelium in Hemostasis and
Biodegradable materials have boasted the promise of Thrombosis. Gimbrone Jr MA, Ed. Edinburgh: Churchill
Livingstone, 1986; 1-13.
ultimate inertness in that they are not left behind for reac-
10. Greisler HP, Gosselin C, Ren D, Kang SS, Kim DU.
tion with the body. While very impressive advances are be- Biointeractive polymers and tissue engineered blood ves-
ing made in resorbable polymers,26 their application at this sels. Biomaterials 1996; 17:329-336.
time may still be limited by the need for mechanical proper- 11. Seeger JM, Klingman N. Improved endothelial cell seed-
ties that are best met with traditional biomaterials. An addi- ing with cultured cells in fribronection-coated grafts. J
tional issue with resorbable materials is the control of the Surg Res 1985; 38:641-647.
rate and type of resorption, ablative or eroding. It is also 12. Massia SP, Hubbell JA. Tissue engineering in the vascu-
somewhat taken for granted that the inflammatory responses lar graft. Cytotechnology 1992; 10:189-204.
Bioinert Biomaterials: Are Their Properties Irreplaceable? 467
13. Merrill EW, Salzman EW. Polyethylene oxide as a bio- with enhanced infection resistance. University of
material. Am Soc Artif Intern Organs 1993; 6:60. Eindhoven, The Netherlands. 1996:127-148.
14. Harris JM. Poly(ethylene glycol) Chemistry: Biotechnical 23. Stokes K. Environmental stress cracking in implanted
and Biomedical Applications. New York: Plenum Press, polyether-polyurethanes. Polyurethanes in Biomedical
1992:15. Engineering. Plank H, Egbeers G, Syre I, eds. Amsterdam:
15. Llanos G, Sefton MV. J Biomater Sci Polymer Edn, 1993; Elsevier, 1984:243.
4:381-400. 24. Takahara A, Hergenrother RW, Coury AJ, Cooper SL.
16. Sheth SR, Leckband D. Measurements of attractive forces Effect of soft segment chemistry on the biostability of seg-
between proteins and end-grafted poly(ethylene glycol) mented polyurethanes. II. In vitro hydrolytic degradation
chains. and lipid adsorption. J Biomed Mater Res 1992;
17. Kulik E et al. Poly(ethylene glycurface: Reduced vs de- 26:801-818.
layed protein adsorption and activation. Abstract from 25. Schubert MA, Wiggins MJ, Schaefer MP, Hiltner A,
Firth World Biomaterials Congress, Toronto, Canada. Anderson J. Oxidative biodegradation mechanisms of bi-
1996; 29 May-2 June. axially strained poly(etherurethane urea) elastomers. J
18. Ishihar K, Nakabayashi N. Part A: Polymer Chemistry: Biomed Mater Res 1995; 29:337-347.
Specific interaction between water-soluble phospholipid 26. Sawhney AS, Pathak CP, Hubbell JA. Bioerodible
polymer and liposome. J Poly Sci 1991; 29:831-835. hydrogels based on photopolymerized poly(thylene gly-
19. Andree HAM. Phospholipid binding and anticoagulant col)-co-poly(#-hydroxy acid) diacrylate macromers. Mac-
action of annexin V. Ph.D. thesis at the University of romolecules 1993; 26:581-587.
Maastricht, The Netherlands 1992. 27. Andrade JD, Coleman DL, Didisheim P, Hanson SR,
20. Lindhout T et al. Antithrombin activity of surface-bound Mason R, Merrill E. Blood-materials interactions: 20 years
heparin studied under flow conditions. J Biomed Mater of frustration. Trans Am Soc Artif Intern Organs 1981;
Res 1995; 29:1255-1266. 27:659-662.
21. Sapatnekar et al. Blood-biomaterial interactin in a flow 28. Smeets E. Scrambling of Membrane Phospholipids in
system in the presence of bacteria: Effect of protein ad- Platelets and Erythrocytes. Ph.D. Thesis, University of
sorption. J Biomed Mater Res, 1995; 29:247-256. Maastricht, The Netherlands, 1996.
22. HendriksM. A study on the covalent surface-immobiliza-
tion of collagen. Ph.D. thesis, Development of biomaterials
Scaffold Engineering
Material Aspects
CHAPTER 43
Biostable Polymers as Durable Scaffolds
for Tissue Engineered Vascular Prostheses
Arthur J. Coury
Introduction
Design Concept
The product is a vascular prosthesis intended to address the unmet medical need for
functional replacement of small (, 6 mm) and medium (6-12 mm) diameter arterial seg-
ments.3-4 The device consists of a structural matrix (scaffold) of available synthetic
biomaterials and possibly other components assembled prior to implantation. The scaffold
promotes and retains attachment and/or ingrowth of biological tissues. The synthetic scaf-
fold is biostable, that is, resistant to excessive structural deterioration for the projected life-
time of the device, which is considered to be a “permanent” implant. The synthetic compo-
nents of the device elicit an acceptable host response alone or through pharmacologic inter-
vention. The device can be produced by available techniques into configurations which pro-
vide adequate hemodynamic flow. It may be packaged, sterilized, stored and implanted by
accepted processes. It is amenable to any special techniques (e.g., in vitro cell seeding or
surface modification) which are required for its proper function.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
470 Tissue Engineering of Prosthetic Vascular Grafts
Hydrolytically susceptible bonds generally consist of Inside the body, biomaterials are subject to oxidative
carbonyl groups with a heteroatom (O,N,S) on one or both insult as a consequence of inflammation due to the foreign
sides of it. These structures and several other hydrolyzable body reaction (phagocytic attack).9,17 Powerful oxidative
groups are listed in Figure 43.1. products result from phagocyte activation (e.g., hydroxyl
Hydrolysis rates differ among the susceptible groups. radical, peroxynitrite, hypochlorite), which can attack oxi-
For example, carbonyl group reactivity decreases in the order: dizable bonds. Susceptible sites generally are those that can
anhydride > ester > urethane > amide. Ethers, ketals and stabilize a free radical (Figure 43.2).9
acetals are labile to acidic pH but stable to basic conditions. Oxidatively stable structures include straight-chain
Backbone structure and intermolecular interactions hydrocarbons, halocarbons and fully-oxidized groups (e.g.,
of hydrolyzable polymers also affect degradation rate. Fac- ketones, sulfones, carbonates, esters, urethanes). We have,
tors such as crystallinity, hydrophobicity and crosslink den- however, produced some evidence that the carbon atom next
sity tend to decrease hydrolysis rate9 and render polymers to the heteroatom of the latter carbonyl groups may be ame-
based on hydrolytically labile groups potentially viable can- nable to oxidation.17
didates for long term implant. Evidence for direct (receptor-ligand) enzymatic
Polymer structures that resist hydrolysis include hy- catalysis of oxidation is limited and inidcative of minor con-
drocarbons, silicones, sulfones, halocarbons and isolated sequences.12,18
carbonyl-containing molecules (i.e., ketones). In vivo oxidation of polymers has been most exten-
sively studied for polyurethanes and polyethylenes.9,15-24
Oxidation Structural degradation due to oxidation is usually manifest
Oxidative degradation of polymers involves destruc- as surface fissuring, deep crack formation, fragmentation
tion or modification of molecular structure via electron or wear, often in zones of applied stress. Radiation steriliza-
transfer reactions.9 The polymers to be considered have vary- tion (e.g., of polyethylene) can produce long-lived radicals
ing degrees of susceptibility to oxidation. Prior to implanta- with autooxidation and embrittlement leading to increased
tion, specific operations such as melt processing, radiation wear and fragmentation in use.
sterilization or exposure to light containing ultraviolet wave- Mitigation of structural degradation due to oxidation
lengths can promote oxidative degradation. Autooxidation can be accomplished by minimizing residual and applied
involves reaction with molecular oxygen and is a likely stress, controlling exposure to destructive radiation, use of
mechanism of many oxidative events.15 Other oxidants (e.g., antioxidants, isolating susceptible polymers in a device from
metal ions, hypochlorite, hydroperoxides)16 are also effec- direct attack by phagocytes or soluble oxidants and, finally,
tive in causing degradation. by the use of oxidation resistant materials.9,10,15,16
X=O,N,S,H
Y=O,N,S,H Z=O,N,S
X or YΙH
Hydrolyzable carbonyl structures
O CH2 (ether)
O
CH CH
(cyanoacrylate)
O P O (phosphate)
C N C OR
O O
O
S NH (sulfonamide)
R=alkyl
O
Other hydrolyzable groups
472 Tissue Engineering of Prosthetic Vascular Grafts
*
OH
*
C O
H
phenol aldehyde
* = susceptible site
cation has been reported to occur with synthetic vascular Poly(Ethylene Terephthalate)(PET) OC COCH2CH2 n
prostheses, causing stiffening and obstruction.25 The most PET vascular prostheses have dominated the large di-
prominent effects of calcification occur, however, in devices ameter field since their introduction in 1957.27 PET displays
that undergo extensive cyclical deformations in vivo (e.g., several advantages in its “standard” or “micro-denier” fiber
prosthetic heart valves, heart assist devices).25 Generation form. It is a strong material having a tensile strength in ori-
of microscopic defects in the polymer may produce nucle- ented form of 170-180 MPa and a tensile modulus of about
ation sites for the initiation of crystal formation, which can 14,000 MPa.28 It is readily fabricable into woven or knitted
lead to extensive mineralization and, ultimately, mechani- textile or mesh form. It can be crimped to enhance kink re-
cal failure of the device. Chemical degradation of the sistance. It is relatively stable. Although it is based on the
biomaterials has not yet been implicated in initiating or pro- hydrolytically susceptible ester group, it is highly crystalline
moting calcification. Anticalcification additives have been (MP ~ 265°C) because of its ordered chain structure and
applied to synthetic biomaterials with some success.26 Vas- deep drawing during fiber formation.9 Implant durability
cular implants that undergo moderate deformation should studies have shown or estimated progressive deterioration
present a relatively low risk for calcification. of physical properties over decades as a consequence of hy-
drolysis, and, possibly, some oxidation induced by activated
Polymers with Potential to Serve phagocytes.29-31 In vivo studies have projected approximately
as Biostable Scaffolds 30 years to full resorption in humans.29,30
This section describes polymers with potential for use PET is relatively stable to sterilization by ethylene ox-
as scaffolds for permanent tissue engineered vascular pros- ide and ionizing radiation;32,33 however, techniques involv-
theses. Commercial polymers used in existing prostheses will ing steam, dry heat or chemically reactive sterilants32-34 may
be described first. Polymers that may not have been com- cause deformation or degradation.
mercialized but have been described in the literature for use Mitigating against the use of PET for small diameter
in medical devices will be briefly described. Finally, com- prostheses is the high reactivity of blood and vascular tissue
mercial polymers that have not been reported in vascular toward the implant with consequent high inflammation,
prosthesis literature, but may satisfy the design requirements neointimal proliferation and inhibition of cellular regenera-
for scaffolds listed above, will be mentioned. tion. This response to PET has been demonstrated with tex-
All of the polymers to be described are assumed to tile vascular prostheses35-39 and vascular stents in open mesh
contain additives such as processing aids and stabilizers. configuration.40,41 The blood and vascular tissue reactivity
Different grades, lots or brands of polymers with the same of PET can vary with the proprietary additives and treat-
generic structure may be more or less stable as a result of ments applied to the individual manufactured devices.42
additives, processing conditions, molecular weight differ- PET has successfully served as the structural matrix
ences, crystallinity differences and other variables. for composite vascular devices. Albumin, collagen or gela-
Biostable Polymers as Durable Scaffolds for Tissue Engineered Vascular Prostheses 473
CH3
tin impregnation of PET textile prostheses is performed to
seal them against blood leakage.38-43 Healing responses of Polypropylene(PP) CH CH2 n
the composite are comparable to the unsealed PET.44 The PP has considerable appeal as a biostable scaffold. It is
Omniflow Vascular Prosthesis (Bio Nova, International) is a relatively strong (tensile strength = 400 MPa), crystalline,
formed from PET mesh placed on a silicone mandrel and high modulus (tensile modulus = 2.6 GPa) thermoplastic in
implanted in sheep to produce a collagen-PET composite its isotactic form.56 Its hydrocarbon structure renders PP
device. It has been used successfully for peripheral vascular insensitive to hydrolytic attack. However, since every other
replacement.45 Other modifications, such as incorporation atom on the polymeric chain is a tertiary carbon, it is sus-
of bioactive molecules46,47 and surface modification for ceptible to oxidation during processing, storage and implan-
blood compatibility48 have shown promise. tation.57 This type of degradation can be effectively coun-
In sum, PET should be considered a highly credible tered by the use of antioxidants, which are universally used
candidate for biostable vascular scaffolds. in PP products.57,58 Consequently, stabilized PP is consid-
ered a “permanent” implant material.
Polytetrafluoroethylene(PTFE) CF2CF2 n Biostability, strength, and durability combined with a
PTFE is a member of the fluorocarbon class of poly- relatively low inflammatory tendency59-61 make stabilized PP
mers. It is, by far, the most commonly used fluorocarbon49 the material of choice for vascular anastomotic sutures,59-61
in implants because of its demonstration of long term certain ligament augmentation devices58,62 and mesh for
biostability and biocompatibility in vivo. PTFE vascular pros- surgical repair.63
theses are prominent in the medium diameter (~7-9 mm) PP yarns have been woven into multifilament tubes
and dialysis access markets.27 for investigation as single component vascular prostheses59
The polymer is prepared in powdered form from or as the reinforcing matrix for partially absorbable com-
tetrafluoroethylene monomer. The product is a highly crys- posite devices.64 Chronic animal implant studies indicated
talline material (> 90% crystallinity) which is not completely that PP offered potential advantages in efficacy over ex-
fusible, but is formed into shapes by sintering.49 A densely panded PTFE and PET in small diameter vascular applica-
sintered configuration approaches the intrinsic properties tions.59 Biomechanical behavior, which could be readily
of the material and gives a moderate stiffness (tensile modu- modulated by varying fiber diameter and weaving condi-
lus of elasticity = 0.5 GPa) and tensile strength (14 MPa).49 tions, was shown to have a significant effect on tissue in-
Implantable forms of the material of interest for vas- growth and the resulting hybrid bio-artificial composite.64,65
cular prostheses include the “expanded” and textile prod- PP is one of the least resistant structural polymers to
ucts.50 Expanded PTFE (ePTFE) is made by an extrusion, sterilization by ionizing radiation.32,33 However, it is safely
drawing and sintering process50 to produce a tube with a sterilized by chemical agents and autoclaving.32-34
porous wall consisting of fibrils and nodules which is con- Recent innovations in the technology of PP include a
trollable to different pore sizes (e.g., 30 and 60 ∝m). The syndiotactic product and an ionomer (ion-containing poly-
porosity can be used advantageously to promote ingrowth ethylene)-modified isotactic PP which produce lower moduli
of tissue and formation and retention of an endothelial layer (0.5 GPa) than isotactic PP (2.6 GPa).56 Finally, a formula-
in vascular prostheses.51,52 tion of polypropylene rendered radiation resistant by the
Knitted PTFE cloth is effectively used as sewing rings incorporation of hindered amine light stabilizers and a
for heart valve prostheses,53,54 and should be considered plastomeric ethylene polymer has been described.66
when a textile configuration for a biostable scaffold is pre- PP is normally considered a low surface energy, hy-
ferred. It should be noted here that the preparation of PTFE drophobic material to which surface bonding is difficult.
fibers requires the use of sizing agents which must be re- However, certain highly hydrophobic cellular strains adhere
moved by vigorous (and proprietary) cleaning processes. strongly to PP.67 This suggests that the adherence of tissue
PTFE has a notable shortcoming—its susceptibility to generated on PP scaffolds should be studied on a case by
degradation by ionizing radiation as experienced during case basis with consideration of the possibility surface modi-
gamma sterilization.32,33 It is completely stable to other forms fication to control delamination.
of sterilization, however. Polypropylene should be considered in the top tier of
Another potential issue with PTFE concerns the ad- candidates for biostable vascular matrices.
herence of other materials to its surface. The polymer is noted O
for its release properties, and this is an advantage for
declotting vascular access prostheses.27 However, if a hybrid Polyurethanes(PUR) NHCO
bio-artificial device requires the bonding of a substantial PURs comprise a large family of polymers which is
amount of tissue to the PTFE surface (i.e., without com- notable for its diversity. The only required attribute that in-
plete mechanical interlock), a potential for delamination may dividual members have in common is the presence of the
exist. This factor has been recognized and promising ap- urethane [-NH(CO)O-] group in some repeating sequence
proaches to enhance the binding of, for example, endothe- on the main chain, from the reaction of an isocyanate group
lial cells are being developed. These include denucleation of with an alcohol group. Most likely, however, there are other
ePTFE by saturating its pores with water, use of surfactants functional groups which make up the soft segment of the
and treatment with adhesion molecules (e.g., fibronectin) PUR, which is generally a copolymer consisting of hard and
or adhesion peptides.52,55 soft segments. The hard segment generally consists of the
474 Tissue Engineering of Prosthetic Vascular Grafts
reaction product of a diisocyanate and a diol or diamine. biodegradation is the soft segment (ester, ether, carbonate).
The soft segment is derived from a macromonomer rang- While hydrolysis of the urethane (or urea) hard segment link-
ing from several hundred to several thousand in molecular ages is possible, those groups are relatively stable and do not
weight.10 Typical monomers for polyurethanes which have comprise the primary mode of biodegradation.
been used as “permanent” implants are listed in Figure 43.3.10 Polyester soft segments have generally degraded
Most of the polyurethanes used in medical devices are based quickly and severely by hydrolysis.16 Polether urethanes are
on difunctional monomers. susceptible to oxidative degradation as described in the
Commercial PURs usually contain additives such as “Modes of Polymer Degradation” section, above.71,72 Since
catalyst residues, processing aids and stabilizers. These ad- oxidative susceptibility is generally proportional to ether
ditives often migrate to the surface of the PUR part and serve content,9,10,15,16 relatively hard (low ether content) PURs
as the material in contact with blood or soft tissue.10 Cer- have performed with minimal degradation for periods as
tain PURs have demonstrated relatively low long as the lifetime of pacemakers (8-10 yr). Even soft
thrombogenicity.10 polether urethanes are capable of performing as intended
PURs range in physical properties from soft, tough for years in the absence of high stress or strong oxidative
elastomers to strong, rigid structural polymers (e.g., tensile attack.15,16
strength = 20-50 MPa, tensile modulus = 5-1150 MPa).10 As Over the past few years, several industrial concerns
a result, PURs have been considered for soft, flexible im- have reported on the development of polyurethanes based
plantable device components such as pacemaker lead insu- on polycarbonate soft segments.69,73-75 These have shown
lation15 tubing and compliant vascular prostheses68-70 as well very high resistance to degradation in short term studies,
as hard, rigid components such as pacemaker lead connec- although minor hydrolytic effects have been noted on the
tors.10 The polyurethanes used in medical devices are usu- implant surfaces. 69,76 However, in vascular implants
ally formed into shapes by solution or melt processes10 and approaching one year, structural degradation of the
can be solution or melt spun into fibers, cast into porous or poly(carbonate urethane) fibers was detectable.69 This is
solid-wall structures, or extruded into solid-wall tubing. predictive, in my opinion, of a progressive hydrolytic degra-
The record for PURs in “permanent” implants is dation that should make a device designer very wary of
mixed, because of hydrolytic and oxidative degradative choosing poly(carbonate urethanes) as “permanent”
mechanisms that may come into play. Generally, the site of
Biostable Polymers as Durable Scaffolds for Tissue Engineered Vascular Prostheses 475
scaffolds until long term (> 5 years) studies have confirmed ecules” in the zones between crystalline regions in these semi-
their stability. crystalline polymers.84 ESC is reduced in PE with lower crys-
An approach to polyurethane design that uses soft seg- tallinity (LLDPE) or higher molecular weight (UHMWPE)
ments composed entirely of hydrocarbon molecules (i.e., a because of higher concentrations of tie molecules relative
36 carbon structure derived from dimerized fatty acids) with to crystalline regions.84 Therefore, two forms of PE, LLDPE85
no ether, ester or carbonate groups to degrade77,78 has theo- and UHMWPE, have theoretical appeal for ESC resistant
retical appeal. However, this concept must also pass the test fiber-based prostheses. The latter has been used as fibers for
of long term implant stability before it may be used with sutures.86 The LLDPE is reported to be resistant to crack-
confidence. ing.85 A previously unmentioned form of PE deserves men-
Polyurethanes are generally resistant to sterilization tion—gel-spun UHMWPE. These fibers achieve levels of
of all types except steam and, possibly, dry heat.10 They are tensile strength (2-4 GPa) and tensile modulus (125-175
susceptible to calcification, especially in highly dynamic ap- GPa) that are multiples of the PE levels reported above.87
plications such as leaflet heart valves.25 Their high stability and strength at very low fiber diameters
Although polyurethanes display some of the most fa- are potentially useful design features for scaffolds.
vorable mechanical properties for compliant grafts, no com- PE is resistant to chemical sterilization and suscep-
position has yet demonstrated dependable long term stabil- tible to degradation by ionizing radiation.22 Most forms of
ity in vivo, especially as a fibrous vascular implant. thermal sterilization would warp the relatively low melting
PE (MP 104-135°C).83
The PE family, in my opinion, has not received
Polyethylene(PE) CH2 CH2
n adequate consideration for use in filament-based
PE is produced in several forms including very low vascular prostheses.
density (VLDPE), low density (LDPE), medium density CH3
(MDPE), high density (HDPE), linear low density (LLDPE) O Si
and ultra high molecular weight (UHMWPE), each with n
Polydimethylsiloxane (PDMS)
different thermal and mechanical characteristics.49 The CH3
variations in density and physical properties result from dif- The PDMSs comprise a family of thermoset elastomers
ferences in polymerization conditions or the use of other with a long history of successful use in implantable de-
olefinic hydrocarbon comonomers.49 All of these molecular vices. 79,88 Structural devices are usually produced by
structures are amenable to fabrication into fibers or cellular crosslinking 2-part systems at room temperature or higher.
structures, which are the forms most likely to meet the re- The crosslinked products are highly elastic and extensible.
quirements of the “design concept.” The tensile modulus (2-9 MPa) and ultimate tensile strength
Although PE has been reported as being used in vas- (2-10 MPa) are relatively low compared to other polymers
cular prostheses40 and is regularly used in several forms of considered in this chapter.10 However, excellent flex fatigue
permanent implants,79 recent literature describing its use in resistance and tear resistance of some compositions,10 com-
vascular prostheses is sparse. More commonly reported is bined with high biostability,88,89 make PDMS a worthy con-
the use of solid-wall polyethylene tubing with or without tender for compliant vascular prosthesis scaffolds.
coatings for thrombogenicity studies.80-82 The studies show PDMS has been studied in vascular prostheses. Be-
that PE can be modified to improve surface wetability for cause of its low modulus, porous-wall structures have been
binding of coatings (e.g., heparin)81 and, potentially, other favored over filamentous configurations. The
components for tissue engineered prostheses. Structurally, “Replamineform” process uses sea urchin spine machined
PE compositions range from relatively low to medium into tubular shape as template for fabrication of porous
modulus when thermally processed (Table 43.1).83 PDMS vascular devices. 90,91 The spine, consisting of
PE in its various structural forms is stable to hydro- microporous calcite, is dissolved in acid, leaving behind an
lytic media and resistant to oxidation in proportion to its open-cell PDMS structure with adequate strength and the
ratio of linear to branched chain structure and crystallinity. potential for tissue ingrowth.90
PE is susceptible to the phenomenon of environmental stress PDMS derives much of its strength from silica filler.
cracking (ESC) produced by exposure of stressed specimens The filler is actively thrombogenic; however, PDMS with-
to aggressive media such as detergents (and, possibly, blood). out filler can be used as a less thrombogenic coating.82 PDMS
Cracking occurs because of stress relaxation of “tie mol- has been used as a surface for endothelial cell growth;82 how-
ever, it is a relatively low energy surface and the authors cau-
tion that resistance to cell detachment under conditions of
blood flow must be verified before a hybrid device based on
PDMS is deemed suitable for long term function.82
Table 43.1. Properties of PE compositions83
PDMS can be sterilized under relatively mild ionizing
radiation conditions and by heat or chemical sterilants.32
2 Tensile Strength (MPa) Tensile modulus (MPa) A final note on PDMS is in order. The PDMS class of
elastomers has come under severe scrutiny in recent years
LDPE 15-80 55-170
because of its use in mammary prostheses. Reports of
MDPE 10-20 170-380
HDPE 15-35 415-1035 autoimmune responses to implanted devices have focused
476 Tissue Engineering of Prosthetic Vascular Grafts
on PDMS oil-filled PDMS shell designs. Leakage of the oil mers (LCPs). They are so named because of their high de-
is alleged to cause systemic responses to PDMS.92 However, gree of order in the liquid phase. Upon cooling, these poly-
decades of successful use of PDMS in solid form79,93 provide mers solidify to structures with anisotropic skin-core mor-
a massive body of evidence that these materials are phologies and tensile properties among the highest of known
chemically stable and safe as implants. The list of suppliers polymers (i.e., tensile strengths to 240 MPa, tensile moduli
of PDMS for “permanent” implants is currently short, but to 32 GPa).98 Chemical degradation resistance is also excep-
PDMS raw materials are available93 and these elastomers tionally high. The materials can be readily extruded into
merit serious consideration as scaffolds for “permanent” tubing and fibers.99 If the fibers are not too stiff, LCPs may
hybrids. be worth considering as structural matrices for hybrid
vascular prostheses.
Other Polymer Scaffold Materials A group of rigid thermoplastics having amorphous
This subsection speculates briefly on polymers that or semicrystalline morphologies and superior resistance to
have not received much reported consideration for vascular chemical degradation includes the polysulfones (polysulfone,
prostheses, but are judged to have the potential to meet the polyethersulfone, polyarylsulfone) the polyketones (PEK,
design criteria for a tissue engineered device. The selection PEEK, PAEK, PEKK, where P=poly; A=aryl; E=ether;
is based on my experience in working with or considering K=ketone), poly(ether imide), polyimide, poly(methyl meth-
the materials for use in “permanent” implants. The sparse- acrylate) and others.98,100-104 The stated polymers normally
ness of available information offers the opportunity for would comprise the nonfibrous component of a fiber-rein-
technical advancement associated with several risks. At best, forced composite used, for example, in orthopedic de-
there will be substantial development expense to assure vices.100-104 Investigation of fibrous forms of these materials
successful implementation. There will likely be increased is a worthwhile undertaking, in my opinion. This is being
regulatory complexity for new vascular materials. A new done with poly(methyl methacrylate) currently, and the re-
vascular material is riskier, because an established clinical sults are impressive.103 For example, the ultimate tensile
history would have revealed characteristics of a material that strength of PMMA can be increased from 25-50 MPa to as
might not be anticipated in the development plan. Finally, high as 220 MPa while enhancing ultimate elongation from
there is the risk that some of these materials have been stud- 5-35% with fiber drawdown ratios of ~20.103,104 Tensile
ied by others and rejected for good cause without the results moduli increase from ~2 GPa in castings to ~8 GPa in the
being published. fibers, which would suggest the use of very thin fiber diam-
The subsection is now separated into two parts, the eters in vascular scaffolds.
first listing high-modulus linear polymers which are best Poly(acrylonitrile-co-vinyl chloride) has been fabri-
utilized in fiber form, the second listing low-modulus elas- cated into hollow fiber form as a cell encapsulation mem-
tomers which may be fabricated into fiber-based, solid-wall brane for hybrid bio-artificial organs and hemo-filtration
or porous devices. membranes. It has been shown to be stable in long term
implants and can likely be extruded or solution spun into
High Modulus Polymers fibers suitable for consideration for vascular scaffolds.105
PTFE is just one of several polymers which belong to
the fluoropolymer category. It also includes: poly(ethylene Low Modulus Elastomers
tetrafluoroethylene), poly(ethylene chlorotrifluoroethylene), Elastomers meriting consideration in this subsection,
poly(vinylidine fluoride), fluorinated ethylene propylene, in addition to the ones already considered, may be either
perfluoroalkoxy resin, polychlorotrifluoroethylene and oth- thermoplastic or thermoset. Both types may be fabricated
ers.49,94 They all offer exceptional resistance to hydrolysis and into fibrous, porous or solid wall form. Thermoset rubbers
oxidation. Just as with PTFE, they are sterilizable by all com- are often prepared from prepolymers (“millable gums”) by
mon means except, possibly, ionizing radiation.32 The “other” hot molding processes. In such cases, residues from the “vul-
fluoropolymers are worth considering as an alternative to canization” (crosslinking) process may need to be removed
PTFE because most are more readily proccessable by melt by extraction to make the scaffold nontoxic. The elastomers
or solution techniques. They also offer a broad range of are often compounded with fillers, stabilizers, processing aids
modulus characteristics to choose from.94 and other ingredients to control physical and chemical prop-
Several rigid polyamides should have the chemical erties of the final product.
resistance to provide long term service in vivo. These in- Certain of the compositions to be described may not
clude Nylon 11, Nylon 1295 and aromatic polyamides (Ara- be available in prepolymer form for fabrication into desired
mids).96,97 Their stability arises either from hydrophobicity shapes, but are sold in finished form by the materials manu-
of the backbone (Nylon 11, 12) or high crystallinity (Ara- facturer. Therefore, any evaluation of a scaffold configura-
mid). Braided Aramid fiber prostheses have been studied tion would require the approval and fabrication by the ma-
for artificial ligaments.96,97 While mechanical degradation terial supplier. In these times of high liability risk, some elas-
was observed under dynamic conditions,96 long term chemi- tomer suppliers may not approve the use of their product as
cal stability in human plasma was confirmed. “permanent” implants.
Certain aromatic polyesters and poly(ester amides) Nonetheless, elastomers offer the potential advantage
belong to a class of polymers called liquid crystalline poly- of radial compliance and deserve consideration. This sec-
Biostable Polymers as Durable Scaffolds for Tissue Engineered Vascular Prostheses 477
tion provides a very brief overview, but recent reviews of indeed, a worthy one which I believe will ultimately be
elastomers are more detailed and comprehensive.106,107 successful.
Polyolefins include rubbers synthesized from olefinic
hydrocarbons, generally crosslinked with dienes. Rubbers of Acknowledgments
ethylene, propylene, isobutylene and higher olefins are in- The literature provided by Drs. Giovani Galletti, Rob-
cluded here. They may be compounded to various ranges of ert Guidoin, Howard Greisler, Vince Medenhall, Colin Pitt
strength, hardness and elongation. They are highly resistant and J. Paul Santerre is gratefully acknowledged. I sincerely
to hydrolysis, and have low residual olefin content for oxi- thank Mrs. Sandra Brigham for her expert preparation of
dation stability (in contrast to rubbers from dienes such as this manuscript.
butadiene). However, they have branched-chain sites which
may be susceptible to irradiation and thermooxidative deg- References
radation, similar to polypropylene. One polyolefin, 1. Stoeger KJ. Implementing the new quality system require-
poly(1-hexene) crosslinked with methyl hexadiene, has en- ments. Design controls. Biomed Instrum Technol 1997;
joyed special success in implantable devices (finger joints, 31(2):119-127.
2. AAMI Standards and Recommended Practices. Biological
artificial heart pumps, compliance chambers, intervetrebral
evaluation of medical devices. Washington, D.C.: Assoc
discs).106 It has excellent biocompatibility (after solvent ex- for the Advancement of Med Instrumentation, 1996:4.
traction), fatigue resistance and biostability. 3. Graham L, Whittlesey D, Bevacqua B. Cardiovascular
Fluoroelastomers and fluorosilicone elastomers should implantation: Vascular grafts. In: Ratner B et al, eds.
have excellent resistance to processing, storage and biologi- Biomaterials Science, An Introduction to Materials in
cal media while retaining low modulus and resilience of true Medicine. San Diego: Academic Press, 1996:420-422.
elastomers.106,107 4. Kantor C. Biological small diameter vascular grafts. AAMI
Polychloroprene, a diene rubber, will not hydrolyze Medical Device Research Report 1996; 3(1):8-11.
and has much greater resistance to oxidation than other diene 5. Galletti PM. Biomaterials availability in the U.S. J Biomed
rubbers (e.g., natural rubber, butadiene rubber). It should Mater Res 1996; 32(3):289-291.
be quite biocompatible if extracted after vulcanization. 6. Citron P. Medical devices: Factors adversely affecting in-
novation. J Biomed Mater Res 1996; 32(1):1-2.
Many other elastomers are available commercially
7. O’Connor KW. Biomaterial bill passes Congress, signed
which may be worth considering as scaffolds.106,107 The ones into law. The AIMBE News, Fall, 1998; 6(3):1,2.
selected for discussion are the ones I might select in a first 8. Coury AJ. Preparation of specimens for blood compat-
round of testing. All of those listed in this subsection would ibility testing. Cardiovasc Pathol 1993; 2(3)
require substantial process development and validation be- (Suppl):1015-1105.
fore clinical studies would be justified. 9. Coury AJ. Chemical and biochemical degradation of poly-
mers. In: Ratner B et al, eds. Biomaterials Science, An
Concluding Comments Introduction to Materials in Medicine. San Diego: Aca-
The extensive list of polymers suggested as potential demic Press, 1996; 243-260.
scaffolds for hybrid bio-artificial vascular prostheses was 10. Coury AJ, Slaikeu P, Cahalan P et al. Factors and inter-
feasible because the “design concept” was rather broad and actions affecting the performance of polyurethane elas-
tomers in medical devices. J Biomater Appl 1988;
limited in detail. With a more stringent set of design criteria
3:130-179.
in place, some of the polymer candidates should be elimi- 11. Zhu KJ, Hendren RW, Jensen K et al. Synthesis, proper-
nated. The strategy of this chapter was to be inclusive, so ties and biodegradation of poly(1,3-trimethylene carbon-
that the major categories of relatively stable polymers were ate). Macromolecules (1991); 24:1736-1740.
captured. One category and one polymer within that cat- 12. Santerre JP, Labow RS, Adams GA: Enzyme-biomaterial
egory will be the best one. The search for that biomaterial interactions: Effect of biosystems on degradation of poly-
offers certain challenge but great potential reward. urethanes. J Biomed Mater Res 1993; 27:97-109.
Finally, I offer a biased opinion about the hybrid de- 13. Ratner BD, Gladhill KW, Horbett TA. In vitro studies of
vice discussed in this chapter. Ideally, the permanent scaf- the enzymatic biodegradation of polether urethanes. Trans
fold is an interim solution on the road to a resorbable scaf- 12th Ann Mtg Soc Biomater 1986; 9:190.
14. Ratner BA, Tyler BJ. Variations between biomer lots 2:
fold which would produce a completely biological, fully func-
The effect of differences between lots on in vitro enzy-
tional neo-artery. My personal experience with a degrad- matic and oxidative degradation of a commercial poly-
able cellular polyurethane vascular prosthesis was that a bio- urethane. J Biomed Mater Res 1993; 27:327-334.
logical conduit indeed was generated. It had many of the 15. Stokes K, Coury A, Urbanski P. Autooxidative degrada-
structural elements of natural artery, but they were not or- tion of implanted polyether polyurethane devices. J
ganized in the same way. The conduit dilated to unaccept- Biomater Appl 1987; 1:412-448.
able levels over several months. Dilatation was also observed 16. Coury AJ, Stokes KB, Cahalan PT et al. Biostability con-
by other investigators using this type of material.108 siderations for implantable polyurethanes. Life Support
A successful result requires guided tissue regeneration Systems 1987; 5:25-39.
which withstands the pressures of the arterial system for the 17. Sutherland K, Mahoney JR, Coury AJ et al. Degradation
long term. The answer will likely lie in the structural design of materials by phagocyte-derived oxidants. J Clin Invest
1993; 92:2360-2367.
of the temporary scaffold, its mechanical properties and its
18. Santerre JP, Labow RS, Duguay DG et al. Biodegradation
degradation characteristics. The pursuit of this design is, evaluation of polyether- and polyester-urethanes with
478 Tissue Engineering of Prosthetic Vascular Grafts
oxidative and hydrolytic enzymes. J Biomed Mater Res cells induced by different graft materials in vitro are af-
1994; 28:1187-1199. fected by pentoxifylline and iloprost. J Biomed Mater Res
19. Taylor G, Gsell R, King R et al. Stability of N2 packaged 1977; 36: 400-406.
gamma irradiated UHMWPE. Trans 23rd Ann Mtg Soc 39. Greisler HP, Schwarcz TH, Ellinger J et al. Dacron inhi-
Biomater 1997; 20:421. bition of arterial regenerative activities. J Vasc Surg 1986;
20. Furman BD, Reish TG, Li S. The effect of implantation 3(5):747-756.
on the oxidation of ultra high molecular weight polyeth- 40. Peng T, Gibula P, Yao K et al. Role of polymers in im-
ylene. Trans 23rd Ann Mtg Soc Biomater 1997; 20:427. proving the results of stenting in coronary arteries.
21. Wang A, Polineni VK, Essner A et al. Effect of radiation Biomaterials 1996; 17(7):685-694.
dosage on the wear of stabilized UHMWPE evaluated by 41. Murphy JG, Schwartz RS, Edwards WD et al. Percutane-
hip and knee joint simulators. Trans 23rd Ann Mtg Soc ous polymeric stents in porcine coronary arteries. Circ
Biomater 1997; 20:394. 1992; 86:1596-1604.
22. Premnath V, Harris WH, Jasty M et al. Gamma steriliza- 42. Marois Y, Guidoin R, Roy R et al. Selecting valid in vitro
tion of UHMWPE articular implants: An analysis of the biocompatibility tests that predict in vivo healing response
oxidation problem. Biomaterials 1996; 17:1741-1753. of synthetic vascular prostheses. Biomaterials 1996;
23. del Prever EB, Crova M, Costa L et al. Unacceptable bio- 17:1835-1842.
degradation of polyethylene in vivo. Biomaterials 1996; 43. Marois Y, Chakfe N, Guidoin R et al. An albumin-coated
17:873-878. polyester arterial graft: In vivo assessment of
24. Lewis G. Polyethylene wear in total hip and knee arthro- biocompatibility and healing characteristics. Biomaterials
plasties. J Biomed Mater Res 1997; 38(1):55-75. 1996; 17:3-14.
25. Pathak Y, Schoen FJ, Levy RJ. Pathologic calcification of 44. Cziperle DJ, Joyce KA, Tattersall CW et al. Albumin im-
biomaterials. In: Ratner B et al, eds. Biomaterials Science, pregnated vascular grafts: Albumin resorption and tissue
An Introduction to Materials in Medicine. San Diego: reactions. J Cardiovasc Surg 1992; 33:407-414.
Academic Press, 1996; 272-281. 45. White JE, Werkmeister JA, Edwards GA et al. Structural
26. Joshi RR, Frautschi JR, Phillips RE. Immobilized heparin analysis of a collagen-polyester composite vascular pros-
and heparin-bisphosphonate prevent polyurethane calci- thesis. Clin Mater 1993; 14:271-276.
fication and thrombosis: In vitro and in vivo studies. 5th 46. Fournier N, Doillon CJ. Biological molecule-impregnated
World Biomater Congress 1996; May 29-June 2: 610. polyester: An in vivo angiogenesis study. Biomaterials
27. Ku DN, Allen RC. Vascular grafts. In: Bronzino JD, ed. 1996; 17:1659-1665.
The Biomedical Engineering Handbook. Boca Raton: CRC 47. Greisler HP, Klosak I, Dennis JW et al. Endothelial cell
Press, Inc 1995; 1871-1878. growth factor attachment to biomaterials. Trans Am Soc
28. Lawton EL, Ringwald EL. Physical constants of Artif Intern Organs 1986; XXXII:346-349.
Poly(oxyethylene oxyterephthaloyl)[poly(ethylene tereph- 48. Phaneuf MD, Quist WC, Bide MJ et al. Modification of
thalate)]. In: Brandrup J., Immergut EH, eds, Polymer polyethylene terephthalate (Dacron) via denier reduction:
Handbook. 3rd Ed, New York: John Wiley & Sons, Effects on material tensile strength, weight, and protein
1989:V101-V105. binding capabilities. J Appl Biomater 1995; 6:289-299.
29. Williams DF. Review: Biodegradation of surgical polymers. 49. Lee HB, Kim SS, Khang G. Polymers used as biomaterials.
J Mater Sci 1982; 17:1239-1240. In: Bronzino JD, ed. The Biomedical Engineering Hand-
30. Kopecek J, Ulbrich K. Biodegradation of biomedical poly- book. Boca Raton: CRC Press, Inc 1995; 580-591.
mers. Prog Polym Sci 1983; 9:31. 50. Shalaby S. Classes of materials used in medicine: Fabrics.
31. Ambrosio L, Apicella A, Mensitieri M et al. Physical and In: Ratner B et al, eds. Biomaterials Science, An Intro-
chemical decay of prosthetic ACL after in vivo implanta- duction to Materials in Medicine. San Diego: Academic
tion. Clin Mater 1994; 15:29-36. Press 1996; 118-124.
32. Lee HB, Kim SS, Khang G. Polymeric biomaterials: Ster- 51. Greisler HP. Growth factor release from vascular grafts. J
ilization. In: Bronzino JD, ed. The Biomedical Engineer- Controlled Release 1996; 39:267-280.
ing Handbook. Boca Raton: CRC Press, Inc 1995; 581-592. 52. Greisler HP, Johnson S, Joyce K et al. The effects of shear
33. Kowalski JB, Morrissey RF. Sterilization of implants. In: stress on endothelial cell retention and function on ex-
Ratner, B et al, eds. Biomaterials Science, An Introduc- panded polytetrafluoroethylene. Arch Surg 1990;
tion to Materials in Medicine. San Diego: Academic Press 125:1622-1625.
1996; 415-420. 53. Changdran KB. Blood-interfacing implants. In: Bronzino
34. Booth AE. Industrial sterilization technologies: New and JD, ed. The Biomedical Engineering Handbook. Boca
old trends shape manufacturer choices. Med Device Diagn Raton; CRC Press, Inc 1995; 648-655.
Industry; 1995; Feb: 64-72. Author: Please provide vol- 54. Yoganathan AP. A brief history of heart valve prostheses.
ume number rather than month. In: Bronzino JD, ed. The Biomedical Engineering Hand-
35. Zenni GC, Ellinger J, Lam T et al. Biomaterial-induced book. Boca Raton: CRC Press, Inc 1995; 1848-1853.
macrophage activation and monokine release. J Invest 55. Wigod MD, Klitzman B. Quantification of in vitro en-
Surg 1994; 7:135-141. dothelial cell adhesion to vascular graft material. J Biomed
36. Greisler HP, Petsikas D, Cziperle DJ et al. Dacron stimu- Mater Res 1993; 27:1057-1062.
lation of macrophage transformtin growth factor-! release. 56. Liu CK. Medical fibers spun from polypropylene. Proc
Cardiovasc Surg 1996; 4(2):169-173. 13th Southern Biomed Eng Conf 1994; 748-751.
37. Greisler HP, Dennis JW, Endean ED et al. Derivation of 57. Williams DF. Review: Biodegradation of surgical polymers.
neointima in vascular grafts. Circ 1988; 78(Suppl 1):I-6 J Mater Sci 1982; 17:1233-1237.
to I-12. 58. Gibbons DF, Mendenhall HV, Van Kampen CL et al. The
38. Swartbol P, Truedsson L, Parsson H et al. Tumor necro- effect of motion on the tissue response to polymeric fiber
sis factor-# and interleukin-6 release from white blood implants. Clin Mater 1994; 15:37-41.
Biostable Polymers as Durable Scaffolds for Tissue Engineered Vascular Prostheses 479
59. Greisler HP, Tattersall CW, Henderson SC et al. Polypro- 77. Coury AJ, Hobot CM, Iverson VB. Novel soft segment
pylene small-diameter vascular grafts. J Biomed Mater Res approaches to implantable biostable polyurethanes. Trans
1992; 26:1383-1394. 4th World Biomater Cong 1992; 661.
60. Bakkum EA, Dalmeijer RAJ, Verdel MJC et al. Quantita- 79. Visser SA, Hergenrother RA, Cooper SL. Classes of mate-
tive analysis of the inflammatory reaction surrounding rials used in medicine: Polymers. In: Ratner B et al, eds.
sutures commonly used in operative procedures and the Biomaterials Science, An Introduction of Materials in
relation of postsurgical adhesion formation. Biomaterials Medicine. San Diego: Academic Press 1996; 50-60.
1995; 16:1283-1289. 80. Rubens FD, Weitz JI, Brash JL et al. The effect of anti-
61. Faulkner BC, Tribble CG, Thacker JG et al. Knot perfor- thrombin III-independent thrombin inhibitors and hep-
mance of polypropylene sutures. J Biomed Mater Res arin on fibrin accretion onto fibrin-coated polyethylene.
(Appl Biomater ) 1996; 33:187-192. Thromb Hemost 1993; 69(2):130-134.
62. McPherson GK, Mendenhall HV, Gibbons DF et al. Ex- 81. Evangelista RA, Sefton MV. Coating of two polyether-
perimental mechanical and histologic evaluation of the polyurethanes and polyethylene with a heparin-poly(vinyl
Kennedy ligament augmentation device. Clin Orthop alcohol) hydrogel. Biomaterials 1986; 7:206-211.
1985; 196:186-195. 82. Keough EM, Mackey WC, Connolly R et al. The interac-
63. Bellon JM, Contreras LA, Bujan J et al. Effect of phos- tion of blood components with PDMS
phatidyl choline on the process of peritoneal adhesion [polydimethylsiloxane and LDPE (low-density polyethyl-
following implantation of a polypropylene mesh prosthe- ene)] in a baboon ex vivo arteriovenous shunt model. J
sis. Biomaterials 1996; 17:1369-1372. Biomed Mater Res 1985; 19:577-587.
64. Greisler HP. Effects of polypropylene’s mechanical prop- 83. Quirk RP, Alsamarraie MAA. Physical constants of
erties on histological and functional reactions to poly(ethylene). In: Brandrup J and Immergut EH, eds.
polyglactin 910/polypropylene vascular prostheses. Am Polymer Handbook. 3rd Ed. New York: John Wiley &
Coll Surg Surg Forum 1987; XXXVIII:323-326. Sons, 1989:V15-V26.
65. Zenni GC, Gray JL, Appelgren EO et al. Modulation of 84. Lustiger A. Understanding environmental stress cracking
myofibroblast proliferation by vascular prosthesis biome- in polyethylene. Med Plast Biomaterials 1996; 3(4):12-18.
chanics. ASAIO J (1993); Vol. 39, No. 3:M496-M500. 85. Thermoplastic Processes, Inc. LLDPE tubing resists crack-
66. Portnoy R. Clear, radiation-tolerant, autoclavable polypro- ing. Med Plast Biomater 1996; 3(3):61.
pylene. Med Plast Biomater 1997; 4(1):40-48. 86. Tomita N, Tamai S, Morihara T et al. Handling charac-
67. Kiremitci-Gumusderelioglu M, Pesmen A. Microbial ad- teristics of braided structure materials for tight tying. J
hesion to ionogenic PHEMA, PU and PP implants. Appl Biomater 1993; 4:61-65.
Biomaterials 1996; 17:443-449. 87. Prevorsek DC. Preparation, structure, properties and ap-
68. DeCossart L, Annis D. An assessment of rigidly controlled plications of gel-spun ultrastrong polyethylene fibers.
technique for the implantation in dogs of a new Trends Pol Sci 1995; 3(1):4-11.
microfibrous 3.8 mm I.D. polyurethane arterial prosthe- 88. McMillin CR. An assessment of elastomers for biomedi-
sis. Proc 2nd Ann Scientific Session, Acad Surg Res 1986; cal applications. In: Szycher, M, ed. High Performance
Oct. 31-Nov. 1, Clemson SC: 36. Biomaterials. Lancaster: Technomic Publishing Co, Inc
69. Zhang Z, Marois Y, Guidoin RG et al. Vascugraft® poly- 1991; 37-49.
urethane arterial prosthesis as femoro-popliteal and 89. Chawala AS, Hinber I. Laboratory evaluation of explanted
femoro-peroneal bypasses in humans: Pathological, struc- gel filled silicone breast implants. Trans World Biomater
tural and chemical analyses of four excised grafts. Cong 1996:300.
Biomaterials 1997; 18:113-124. 90. Tizian C, Salyer KE. Production process of a microvascu-
70. Doi K, Matsuda T. Enhanced vascularization in a lar prosthesis using the “replamineform” principle. Int J
microporous polyurethane graft impregnated with basic Artif Organs 1980; 6:364-465.
fibroblast growth factor and heparin. J Biomed Mater Res 91. Hiratzka LF, Goeken JA, White RA et al. In vivo com-
1997; 34:361-370. parison of replamineform silastic and bioelectric polyure-
71. Zhao QH, McNally AK, Rubin KR et al. Human plasma thane arterial grafts. Arch Surg 1979; 114(6):698-702.
#2-macroglobulin promotes in vitro oxidative stress crack- 92. Nicholson J III, Hill SL, Frondoza CG et al. Silicone gel
ing of Pellethane 2363-80A: In vivo and in vitro correla- and octamethylcyclotetrasiloxane [D4] enhances antibody
tions. J Biomed Mater Res 1993; 27:379-389. production to bovine serum albumin in mice. Trans 5th
72. Schubert MA, Wiggins MJ, Anderson JA. Role of oxygen World Biomater Cong 1996; 304.
in biodegradation of poly(etherurethane urea) elastomers. 93. Winn A. Factors in selecting medical silicones. Med Plast
J Biomed Mater Res 1997; 34:519-530. Biomater 1996; 3(2):16-19.
73. Szycher M, Edwards A, Carson RJ. In vivo testing of a 94. Resins and compounds: Fluoroplastics. In: Kaplan WA,
biodurable polyurethane. Trans 23rd Ann Mtg Soc managing ed. Modern Plast Encyclopedia ’97. New York:
Biomater 1997; 296. McGraw-Hill Co’s., Inc., 1996:B158-B160.
74. Ward RS, White KA, Gill RS. Development of biostable 95. Resins and compounds: polyamides. In: Kaplan WA, ed.
thermoplastic polyurethanes with oligomeric Modern Plast Encyclopedia, ‘97. New York: McGraw-Hill
polydimethylsiloxane end groups. Trans 21st Ann Mtg Soc Co., Inc., 1996:B175.
Biomater 1995; 18:268. 96. Durselen L, Claes L, Ignatius A et al. Comparative ani-
75. Kato YP, Dereume JP, Kontges H et al. Preliminary me- mal study of three ligament prostheses for the replace-
chanical evaluation of a novel endoluminal graft. Trans ment of the anterior cruciate and medical collateral liga-
21st Ann Mtg Soc Biomater 1995; 18:81. ment. Biomaterials 1996; 17(10):977-982.
76. Mathur AB, Collier TO, Kao W et al. In vivo 97. Wening JV, Lorke DE. A scanning electron microscope
biocompatibility and biostability of modified polyure- (SEM) investigation of Aramid (Kevlar) fibers after incu-
thanes. J Biomed Mater Res 1997; 36:246-257. bation in plasma. Clin Mater 1992; 9:1-5.
480 Tissue Engineering of Prosthetic Vascular Grafts
98. Canale B, Hanley S, Braeckel M. New possibiliities for liq- 104. Lewis G. Properties of acrylic bone cement: state of the
uid crystal polymers. Med Plast Biomater 1995; 2(3):24-31. art review. J Biomed Mater Res (Appl Biomater) 1997;
99. ACT Medical, Inc. Liquid crystal polymer tubing. Med 38:155-182.
Device Diagn Industry 1997; 19:211. 105. Shoichet MS, Rein DH. In vivo biostability of a polymeric
100. Moore R, Beredjiklian P Rhoad R et al. A comparison of hollow fiber membrane for cell encapsulation.
the inflammatory potential of particulated derived from Biomaterials 1996; 17(3):285-290.
two composite materials. J Biomed Mater Res 1997; 106. McMillin CR. An assessment of elastomers for biomedi-
34:137-147. cal applications. In: Szycher M, ed. High Performance
101. Zhang G, Latour Jr RA, Kennedy JM et al. Long-term Biometerals. Lancaster: Technomic Publishing Co., Inc.,
compressive property durability of fiber reinforced 1991:37-49.
polyetheretherketone composite in physiological saline. 107. Courtney PJ, Sevenson JA, Verosky C. Bonding elastomers
Biomaterials 1996; 17(8):781-789. with adhesives. Med Plast Biomater 1997; 4(3):60-68.
102. Barton AJ, Sagers RD, Pitt WG. Bacterial adhesion to or- 108. Galletti G, Farruggia F, Baccarini E et al. Prevention of
thopedic implant polymers. J Biomed Mater Res 1996; platelet aggregation by dietary polyunsaturated fatty ac-
30:403-410. ids in the biodegradable polyurethane vascular prosthe-
103. Gilbert JL, Ney DS, Lautenschlager EP. Self-reinforced sis: An experimental model in pigs. Ital J Surg Sci 1989;
composite poly(methyl methacrylate): static and fatigue 19(2):121-130.
properties. Biometerials 1995; 16:1043-1055.
Scaffold Engineering
Material Aspects
CHAPTER 44
Biophilic Polymers: What’s on the Horizon?
Patrick T. Cahalan
T his chapter was outlined for a section of this book entitled ‘Bio-Interactive’ Prostheses,
and was further subdivided to a section including biostable polymers/materials. The other
chapter in this subsection, titled “Biostable Polymers as Durable Scaffolds for Tissue Engi-
neered Vascular Prostheses” is being written by a dear colleague, Art Coury, who shares this
author’s sense of trepidation as to achieving substance in the context of the remaining chap-
ters of the overall section. It is hoped that, although this chapter will be written from an
industrial perspective, it will have some practical value for the reader.
The word biophilic cannot be found in the dictionary, but then neither can the word
biocompatible. In the early 1980s materials were placed in four basic groups:
1. Bioinert;
2. Biocompatible;
3. Bioactive; and
4. Biointeractive.
Bioinert surfaces were hypothesized to be “invisible” to the body, and to have little or
no interaction. Proposed early examples were negative surface charge to repel cell adhe-
sion,1 high surface energy materials2 such as pyrolytic carbon and low critical surface ten-
sion materials3 such as fluoropolymers, which also claimed low protein and platelet adhe-
sion. Also in the early 1980s, hydrophilic materials were suggested to be low protein adsorbing
and platelet adhering,4 and in particular PEG-like surfaces were claimed to have increased
surface motion (flagella-like activity) that served to decrease protein adsorption and dena-
turation.5 The term biocompatible could be applied to any material that when implanted
showed an equal or better tissue response compared to a control material, such as polypro-
pylene, that was accepted as biocompatible. Bioactive surfaces were surfaces designed to
promote an advantageous effect such as preferential adsorption of albumin by C-18 alky-
lated surfaces.6 Finally, biointeractive surfaces were those designed to interact with a specific
physiological mechanism such as coagulation; an early example is ionically bound heparin
for local release to decrease coagulation.7,8 Heparin was finally covalently immobilized and
shown to have bioactivity without releasing by Larm;9 this surface has been shown in vitro
to bind ATIII to produce a catalytic rate of thrombin deactivation. These categories were
arrived at largely from the perspective of the empirical surface response, and arguably need
some revaluation with more instrument-intensive analytical methods now available to prop-
erly order them by rank in a new era of tissue engineered materials.
The editors have chosen new labels, and have done so with the healing response in
mind. Thus they have set the stage for the conceptual advantages of tissue engineering. In
labeling bio-inert materials as those exhibiting insufficient healing, biolized (endothelialized
surfaces) as surface healing, and bio-interactive as complete healing, they have set the goal
to incorporate all that has been learned to date towards designing materials and structures
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
482 Tissue Engineering of Prosthetic Vascular Grafts
that should more closely approximate naturally function- plete loss of heparin activity, it is highly optimistic to hope
ing tissue. In the early years of vascular graft implantation, for a synthetic anionic polymer with heparin properties.22
Voorhees’ observations with Vinyon “N” cloth in dog aortas Studies in our laboratory with compounds reported as
clearly showed that a synthetic material could serve as a con- heparinoid or anticoagulant fail to produce heparin-like
duit, and be accepted by the tissue.10 Tissue engineering, or activity when measured in solution with ATIII and throm-
the integration of biology and biomaterials, holds out the bin. Surfaces with immobilized cationic or anionic charac-
promise of producing more than an accepted conduit, and ter can adsorb ATIII and thrombin respectively, and give false
obviating all the problems associated with the current com- positives for heparin activity. An amine functional surface
mercial products such as thrombotic occlusion, anastomotic can show binding of ATIII that is resistant to extensive 0.15
hyperplasia, aneurysmal dilatation, and infection. Presum- ionic strength rinsings, and if given time will deactivate
ably, bio-interactive prostheses are the desired approach to thrombin, though not in a catalytic fashion. An anionic sur-
achieving this goal. face can bind thrombin that does not rinse off in the nor-
During the period between 1982 and 1988, this au- mal rinse steps used before difference measurement using
thor was involved with research to find a small diameter chromogenic substrate, giving a false measure of thrombin
(4 mm or less) vascular graft that could exhibit patency at deactivated. Active thrombin can be witnessed by adding
least equivalent to that of autologous saphenous vein. In re- substrate back to the surface and obtaining a color change
ality the effort was more a development, or an evaluation in the substrate. Since it is well known that anionic surfaces
exercise, that included animal implantation of numerous are contact activating, it is of concern to have an anionic
technology platforms to include: surface present that does not have high heparin activity. Since
1. Biodegradable polyurethane-PLLA grafts; coagulation is so dependent on platelets, it is hypothesized
2. Biostable polyurethane grafts made by phase inversion, that by using a polymer that mimics the nonactivated plate-
electrostatic spinning, and spray techniques; let membrane, namely phosphatidylcholine, it would be
3. Alternatively (non glutaraldehyde) fixed heterografts; possible to prevent platelet interaction with the polymer
4. Plasma TFE coated Dacron prostheses; surface (Fig. 44.1).
5. Silicone replamarinaform porous conduits; and Phosphatidylcholine in the form of commercially
6. Dacron and PTFE control grafts from commercial sources. available lecithin can coat surfaces of hydrophobic polymers,
Predominant modes of failure were: aneurysmal dila- but is easily removed with flowing plasma. In attempt to
tation (1 and 2); in vivo degradation across species (3); re- remedy this problem, commercial efforts have given rise to
jection by surgeons at the need to cut the prostheses with an a network polymer with strong adhesive properties to poly-
electrical device (the flaming graft), and undesirable suture mers, and containing pendant phosphorylcholine moieties
characteristics (4); poor crush resistance leading to throm- as seen in Figure 44.2.23
botic occlusions, and poor suture pull out properties (5). The most common network polymer is an acrylate
The Dacron and PTFE grafts had patency rates not com- backbone that has good adhesive properties for PVC and
petitive with autologous vein, and in general showed a fi- polyurethanes. The polymer gives excellent wetting proper-
brotic nonintegrated histology. Fabricating prostheses for ties to the surfaces it coats, and has very low platelet adhe-
implantation gives one an appreciation for necessary me- sion. Human blood testing in our laboratories show this
chanical properties such as suture strength, ease of suture, polymer to be promising, but material-dependent.
burst strength, porosity, and wall thickness for matching Perhaps the most innovative biophilic polymer syn-
vessel anastomosis. Since these properties have been achieved thesis to date is the work of David Tirrell in recombinant
for the most part in commercially available materials, it was artificial structural proteins. These polymers hold out the
felt that applying surface modification technologies to ex- potential of engineering proteins with controlled crystallin-
isting materials might be able to control problems such as ity and expression of specific peptide sequences at surfaces.
thrombosis and anastomotic hyperplasia, and possibly If desirable mechanical properties can be engineered into
achieve a healing response that would result in the protein polymers, and if processing methods are devised
neovascularization, endothelialization, and a tissue response to make fibers, extrusion, or coating possible, then these
that was not primarily a fibrotic or a chronic inflammatory polymers can make their way into devices.
response. The latter could help a graft towards long term There has been somewhat of a rebirth in surface modi-
patency and infection resistance. fying additives (SMAs). If a commercial group could blend
If one defines biophilic polymers as those specifically in additives to polymers that would express themselves on
synthesized to interact in an advantageous manner with the surfaces of devices that their customers manufacture, and
body, there are many claims, but few real candidates are on demonstrate enhanced biological response, then a premium
the near horizon for application. There are numerous claims price could be charged for such materials. To date, because
for heparinoid polymers.21 These polymers often have car- of the lack of tonnage of polymers used in biomaterials, it
boxyl and sulfate functionality approximating the ratio con- has been less than attractive for larger chemical companies
tained in heparin. Studies showing less adhered platelets, or to specifically manufacture biomaterials. That, together with
changing clotting times, are generally used as proof of hep- the liability issues surrounding medical devices, makes it
arin-like activity. In light of studies demonstrating the unique difficult to see new formulations coming from the larger
pentasaccharide sequence required for activation of ATIII, chemical companies. SMA technology is particularly being
and slight changes in this structure resulting in 90% to com-
Biophilic Polymers: What’s on the Horizon? 483
applied for blood-contacting surfaces for short term use, attachment, then, with some license in nomenclature, we
such as cardiopulmonary bypass circuits. have created new polymers. In light of later chapters on sur-
For the most part, biophilic polymers on the near ho- face modification for specific surfaces, we hope that it is
rizon are going to be polymers that have been surface modi- appropriate in this chapter to discuss the more general meth-
fied. Simple adsorbed coatings will be problematic from the ods of surface modification that can be used to get to the
standpoint of assuring stability for permanent implants, and biophilic surface, and point to critical concerns and oppor-
there continues to be a quest for the universal biophilic poly- tunities that may be afforded from our experiences.
mer that can coat all materials. It was the choice of our group In 1989 we created a biomaterials laboratory in
to attempt to comprehensively evaluate methods to co- Maastricht, The Netherlands. The main objective was to fo-
valently couple molecules to the surfaces of common poly- cus on surface modification technologies to improve the
meric biomaterials in hopes of creating new modified biocompatibility of materials used to construct biomedical
biophilic polymers that are stable and have controlled load- devices. The industrial laboratory was ideally situated
ing of bioactive molecules. If we achieve covalent and stable 100 meters from the Biomedical Research Institute of The
484 Tissue Engineering of Prosthetic Vascular Grafts
University of Maastricht, and the group of Professor Coen boxyl groups. This can also be accompanied by crosslinking
Hemker, an experienced and respected expert in thrombo- or chain scission, the former leading to increase in tensile
sis and hemostasis. A partnership was established between properties and the latter to polymer degradation. The first
the University of Maastricht (RL), The Bakken Research consideration on the use of irradiation should be if the
Center (BRC) and The Center for Surface and Materials polymer to be modified is predominantly a crosslinking or
Analysis (CSMA), in Manchester, England. The BRC was to chain scission polymer. Some common chain scission poly-
provide modified biomaterials for characterization by mers are polymethylmethacrylate (PMMA), polytetra-
CSMA, using predominantly XPS and TofSIMS, and fluoroethylene (PTFE), poly(vinylidene chloride), polyiso-
biocompatibility testing by RL. The partnership received a butylene (and copolymers) and polypropylene. Attempting
three year research subsidy from the EC in the form of a to modify these materials with irradiation may give a sur-
Brite EuRam grant (BE 5972). In this project a broad survey face modification, but on a degraded material. Recent high
of physical and chemical methods to modify biomaterials voltage accelerators with high penetration potential have
was performed (Table 44.1), and these methods were evalu- been suggested as an alternative, because relatively low total
ated on seven common biomaterials for feasibility. 11 dosage is required. It has also been suggested that total de-
Equipped in our laboratory with a plasma reactor, corona vices can be modified.12 The listed materials that are claimed
treater, ozone generator, UV photopolymerization equip- to have been successfully modified include in particular the
ment, and a nearby gamma-beta commercial irradiation scission polymer PMMA. These irradiation techniques, par-
source, we were prepared to generate many of the surfaces ticularly when grafting is performed in situ, require near
proposed to be more biocompatible for head to head com- oxygen free environments and thus create some problems
parisons with our partners’ assistance. What will be presented for ease of manufacture. Nevertheless, irradiation is a very
in this chapter are our somewhat narrow perspectives on effective method for covalently grafting molecules to bio-
making polymers biophilic; our experience with synthesis material surfaces.
is limited to modifying the surface of biomaterials, and does
not include creating new bulk polymers. With respect to the Plasma
latter, we maintain surveillance of the literature and sample Plasma polymerization has been touted as the most
new materials through our testing schemes when available exact method to modify biomaterial surfaces; this is in a large
from the developers. part due to the control over the plasma gases, and the thick-
The processes mentioned in Table 44.1, although not ness and uniformity of the modified surface. Some draw-
complete, represent treatments that were performed on at backs to plasma are:
least 3-7 common materials for each method. Some brief 1. Requirement for reactor specific design based on ge-
comments will be given here that are felt to be important ometry and materials of construction;
concerning these surface modification methods and their 2. In almost all cases the process will be a batch opera-
applicability to biomaterials and biomedical devices. tion; and
3. Contamination of the reactor and thus the require-
Physical Methods ment for cleaning cycles and for single process dedi-
cation of reactors. Several investigators have pro-
Irradiation posed simple plasma treatment to make materials
Early work by Ratner and Hoffman4 showed the po- more hydrophilic and thus more blood compatible.13
tential of grafting biomaterials with irradiation sources. Our Plasma treatments may show initial lowering of con-
efforts included irradiation using electron beam sources to tact angle, and will follow with an inversion within
successfully graft to polyolefins. E-beam or gamma radia- hours back to a hydrophobic surface. The latter is
tion can introduce several functionalities to polymeric ma- particularly true of elastomers.
terials, such as free radicals, vinyl bonds and, in the presence Plasma polymerized surfaces can be directly depos-
of oxygen, hydroperoxides, carbonyls, aldehydes and car- ited on a surface, or plasma can be used to activate a surface
for subsequent grafting of molecules. The latter is difficult,
as the surface generally requires a short plasma discharge
time, or pulsed plasma discharge. It has been hypothesized
Table 44.1. Methods to functionalize/activate surfaces that free radicals are short lived on the surface of plasma
(Brite EuRam) treated materials, due largely to the rapid termination by
vicinal radicals.14 A final observation on plasma-modified
Physical Chemical surfaces is that the reactivity in terms of coupling further
biomolecules has been less than expected. Evaluation of cou-
Irradiation Oxidation pling to plasma functionalized surfaces based on the results
Plasma Reduction of XPS data suggests that a significant portion of the func-
Corona Hydrolysis tional groups do not react. One possible explanation is that
CVD and PVD Ozonization organic chemistry, and particularly biomolecules, react in
“Simple” Coating Silanization three dimensions, and the plasma surface is highly ordered
Entanglement Grafting
and rather two dimensional in its ability to enter into reac-
Textruing Photocuping
Ion Beam Implant Add’n/Subst’n tions.15 If one sees new biophilic surfaces as containing high
Biophilic Polymers: What’s on the Horizon? 485
density biomolecules such as heparin, proteins and/or cations where the device was simply coated with the poly-
growth factors, they will have to be coupled postplasma treat- urethane polymer. This was best effected by finding a sol-
ment and the ability to control the quantity and density of vent that could attack the surface of the material to be coated
coupling may prove to be a challenge. and result in a mixing of polymer chains that would give a
mechanical bond upon solvent removal. The same method
Corona has been used to imbrue drugs into polymers using solvents
Corona treatment has been used extensively in indus- that could swell the polymer, taking in the drug and leaving
try for improvement of adhesion. It is also the subject of it upon solvent evaporation. The latter method has been used
several papers on surface activation to effect grafting to sur- for introducing antimicrobial agents to polymers. The same
faces.16-18 Several affinity schemes can be used to attach principle applies for SMAs or blooming agents. Addition of
biomolecules to the grafted surfaces. Corona treatment is a amphipathic molecules to polymers can result in the more
rough process compared to plasma discharge, and takes place hydrophilic portion being expressed on the polymer sur-
in an air atmosphere. This results in a highly oxidized sur- face, anchored by the more hydrophobic end entangled in
face, to include sufficient free radicals to effect grafting of the polymer surface. In the case of polyurethanes, most con-
vinyl monomers. It can be used to graft to most polymers, tain processing waxes that migrate to the surface and pre-
but does not work for most fluoropolymers. Corona and vent the polymer from being tacky during extrusion. One
plasma can be used to make surfaces more wettable for such additive, ethylene bis-stearamide wax, has been reported
simple adsorptive coating approaches. There are several sug- to be in part responsible for improved blood compatibility.
gested universal adsorptive coating schemes that often re- Studies in our laboratory of such additives reveal that per-
quire a pretreatment such as corona to improve the unifor- formance in blood is optimal immediately after thermal pro-
mity of coating as well as the adhesive strength of the coat- cessing, but changes with time. XPS shows considerable in-
ing to the base material. crease over time in amide functionality, which may be less
effective than the long fatty chain in improving blood com-
CVD and PVD patibility. It appears that perhaps entanglement or SMAs may
Chemical vapor deposition (CVD) and physical va- be difficult to control at the surface, and thus present some
por deposition (PVD) have long been used in industry as challenges to be solved.
barrier coatings. In the European food industry, PVD has
been used to apply a layer of SiO to packaging material to Texturing
prevent moisture transmission through wrapping. CVD has Professor Andreas von Recum has been one of the
been used in electronics to provide moisture barriers and leading researchers in the field of tissue response to textured
insulative coatings. It was hoped that these methods could surfaces. He has demonstrated that textured implants with
be used as a base for engineering biologically active surfaces. internodal distances of approximately 1-3 microns appear
The ability to deposit a metallic surface such as gold would to have much thinner capsules, indicating less fibrotic re-
give a base for self assembled surfaces.19 It has also been sug- sponse.38,39 One of the hypotheses offered in explanation is
gested that a ceramic based PVD surface could be activated that the textured surface allows for more stable anchorage
to produce functionality (carboxyl groups) to which of fibroblasts, and less destruction of these cells due to
biomolecules could be coupled.20 microvibration that can lead to further inflammatory re-
sponse. A possible further explanation is that the
Simple Coatings microtextrue allows for an improved production and attach-
We define simple coatings as those that are designed ment of extracellular matrix on the material surface. Re-
to be applied by simple dipping, spraying, or by pumping search on immunoisolation membranes by Becton
solutions through devices. There is a plethora of coatings of Dickinson presented at an ACS workshop indicated that
this nature offered for short term blood compatibility. All optimal surface texture could result in minimal capsule for-
of these coatings have performance that is material depen- mation and vascular structures present very close to the
dent; this problem is often overcome by a pretreatment such material surface. The latter was independent of the material
as corona. Special attention should be paid to devices made of construction for the membrane. Applying microtextrue
from materials that undergo bending and flexing, as these to devices is a developing technology, and one promising
coatings may have a tendency to crack. Dislodgement of the approach is the use of photopolymerization. This technique
coating may cause a problem, depending on the application. holds out the promise of introducing texture and chemistry
Most manufacturing engineers of biomedical devices will to surfaces, plus the ability to use photo resists or screening
require a battery of tests to assure integrity of the coating, techniques to pattern the sights for cellular attachment to
especially if the device is to be a permanent implant such as surfaces. Application of this technology to vascular grafts
a vascular graft. While the list of short term coatings is long, will be more complicated than simply applying this tech-
the list for permanent implants is very limited. nique to the inside of a polymeric tube that has laser drilled
holes for porosity if one hopes to maintain other important
Entanglement features for grafts such as porosity for ingrowth, kink resis-
In the late 70s and early 80s the wide acceptance of tance, compliance, and suture characteristics.
Biomer as a blood compatible material led to several appli-
486 Tissue Engineering of Prosthetic Vascular Grafts
Ion Beam Implantation can be done fairly easy in aqueous solutions. The ozone pen-
Ion beam has been used to impart surface texture in etrates materials at different rates and this can be measured
addition to surface chemistry. In the orthopedic area it is using colorimetric methods.29 Materials modified by ozone
useful in improving fixation of prostheses to tissue. The Spire may need final reduction to assure that radicals are not
Corporation offers ion beam treated surfaces for improved present, and should undergo rigorous mechanical testing to
slip properties and antimicrobial surfaces, and has made assure no loss in durability of the native polymer substrate.
claims to improved blood compatibility.24 As with PVD, a
metallic surface could provide advantages for further sur- Silanization
face modification using self assembling techniques. Ion beam Silanization is most frequently used for coupling to
implantation can result in crosslinking the surface, and im- metal surfaces. The use of organofunctional silanes allows
part mechanical properties to the material. The tendency the addition of reactive organofunctional groups to the metal
for cracking of the surface on elastomers and materials that oxide surface. These groups can the be used to directly couple
will see chronic flexing should be considered. For elastomeric biomolecules. The most common mistake made with
materials, and in particular silicone, the resultant surface may silanization treatments is improper application and cure
still present the elastomer chemistry primarily. methods. Excessive coating with silanes results in weak sur-
faces that will delaminate, and failure to properly hydrolyze
Chemical Methods the silane before drying will fail to create enough reactive
silanol groups to couple during the dehydration and final
Oxidation, Reduction, Hydrolysis formation of the polysiloxane network. Covalently attached
Most synthetic polymers do not possess functionality heparin via silane activation has been released for commer-
to perform coupling reactions. Several of the physical meth- cial application to intravascular stents in Europe.30
ods mentioned can oxidize the surface and produce radi-
cals, hydroxyl, carboxyl, and carbonyl groups. This oxida- Photocoupling
tion can also be induced by chemical means such as acid In 1969 Knowles demonstrated the effectiveness of
etching or treatment with peroxides.25 For fluoropolymers 2-nitro-4-azidobenzene in forming a nitrene radical upon
that are oxidation resistant, reduction by exposure to so- exposure to UV that could extract a proton directly from a
dium metal in napthalene can produce unsaturation in the polymeric backbone and substitute the organo group to
polymer, and subsequent direct grafting of hydrogels can be which it was attached directly to the polymer.31 Application
effected.26 Hydrolysis, particularly of esters, can introduce of this method to the biological field was pioneered by
functional groups such as carboxyls to surfaces. All these Patrick Guire,32 leading eventually to commercialization by
methods involve degradation of the polymer, and it is nec- the BSI (Biometric Systems Inc.) Corporation.33 Numerous
essary to control or limit the degradation to have stable sur- applications are envisioned by BSI to include enhancement
faces, as well as to prevent degradation of the mechanical of endothelial cell attachment, blood compatible surfaces,
properties of the base material. and infection resistant surfaces. While photocoupling ap-
pears to be a facile process of direct coupling to polymers, it
Addition/Substitution is also material dependent, and often requires a pretreat-
Some polymers such as polyurethane have sites that ment of the surface to remove contaminants or promote
can be attacked by proton extracting bases such as sodium better wetting of the coupling reagents. It also often requires
hydride. The reduced urethane group can then undergo alkyl repeated coatings and light exposures to effect a uniform
substitution using alkyl halides such as octadecly iodide.27 and dense coupling. This latter process can result in
An impressive body of work has been documented by the crosslinking of photobiomolecules within a matrix and
group of Cooper, showing improvement in blood compat- somewhat diminish their activity. Careful cleaning of these
ibility and the ability to control the loading of alkyl groups surfaces is required to remove any unreacted or leachable
on the surface. This method of introducing alkyl groups to species, as is the case with almost all surface modification
the surface may provide better and more stable performance approaches. Nevertheless, this method is receiving very fa-
than previously mentioned methods using SMAs. vorable attention as a commercially feasible method to
modify biomaterials, especially for short term use.
Ozonization
Treatment of materials by exposure to ozone has Grafting
proven effective in activating surfaces for subsequent graft- Direct grafting of hydrogels to surfaces is only pos-
ing reactions.28 Ozone can be applied as a gas or dissolved sible on a limited number of materials. The best know ma-
in water, which gives very attractive manufacturing features terial is polyurethane, where the carbamate group can be
for devices with complex geometry and multiple materials, activated using ceric ion initiation.34 Another material also
as ozone is effective on almost all polymers save shown many years ago to undergo direct grafting is dialysis
fluoropolymers. Ozone is primarily an oxidative approach, membrane.35 Early studies on hydrogel grafted surfaces
and can produce peroxy radicals, but also can introduce func- showed promises of being cell friendly.36 Ratner and
tionality from oxidative degradation. Because it can result Hoffman, using a baboon shunt model, later showed that
in degradation, care needs to be taken to control the expo- hydrogel surfaces may produce emboli and, while not being
sure time, concentration of ozone, and temperature. These thromboadherent, were in fact capable of being thrombo-
Biophilic Polymers: What’s on the Horizon? 487
genic. Attempts to utilize the positive aspects of grafted 4. Ratner BD, Weathersby P, Hoffman AS, Kelly MA,
hydrogels by coupling bioactive agents is now commonplace. Scharpen LH. Radiation-grafted hydrogels for biomaterial
A possible advantage of this approach is lower protein ad- applications as studied by the ESCA technique. J Appl
sorption and nonspecific cell adhesion. Grafted hydrogels Polym Sci 1978; 22:643-664.
5. Merrill EW, Salzman EW. Polyethylene oxide as a bio-
do not require crosslinking and when coupled with
material. ASAIO Journal 1983; 6:60-64.
biomolecules can express more bioactivity of the attached 6. Munro MS, Eberhart RC, Make NJ, Brink BE, Fry WJ.
molecule. For most grafted hydrogels some sort of activa- Thromboresistant alkyl-derivatized polyurethanes. J Am
tion of the base polymer is required, such as plasma, co- Soc Artif Int Organs 1982; 6:65-75.
rona, irradiation, or ozonization. Depending on the grafted 7. Leininger RI. Polymers as surgical implants. CRC Critical
species, additional functionalization may be required to pro- Reviews in Bioengineering 1972; 1:333-381.
vide optimal control of coupling with respect to loading and 8. Gott VL, Koepke DE, Daggett RL, Zarnstorff W, Young
spacing chemistry. A good stable grafted hydrogel allows for WP. The coating of intravascular plastic prostheses with
numerous coupling strategies available from affinity chro- colloidal graphite. Surgery 1961; 50:382.
matography techniques. Some affinity schemes have unstable 9. Larm O, Larsson R, Olsonn P. A new nonthrombogenic
bonds that are allowable for chromatographic application, surface prepared by selective covalent binding of heparin
via a modified reducing terminal residue. Biomat Med Dev
but not desirable for biological application. The coupling
Art Org 1983; 11:161-173.
scheme should be tested rigorously to assure that there is no 10. Voorhees AB, Jaretski A, Blakemore AH. The use of tubes
leaching. In our laboratory we have found that grafting fol- constructed from Vinyon “N” cloth in bridging arterial
lowed by intermediate functionalization allows for easier defects. Ann Surg 1952; 135:332.
control and clean up of the grafted surface, as well as opti- 11. Cahalan PT. Brite EuRam Final Technical Report, Con-
mal loading of biomolecules, even to the point of present- tract Number: BREu-336, Project Number BE-5972-
ing the biomolecule in high purity at the uppermost layers 92.Title: Surface modification of Biomaterials for Biomedi-
of the hydrogel.37 cal Devices. European research subsidy granted in Nove.
1992 running until Nov. 1995. Report available through
Summary European Commission, Director General XII, Science, Re-
The horizon for biophilic polymers was discussed in search, and Development, Rue de la Loi 200, B-1049 Brus-
sels, Wetstraat 200, Brussels, Belgium Office: M075 1/5.
this chapter with an obvious bias toward surface modifica-
12. Goldberg et al. Surface modified surgical instruments, de-
tion of polymers to make them biointeractive. Our efforts vices, implants, contact lenses and the like. U.S. Patent
have led to covalent grafted surfaces on almost all common 5,100,689, Assignee: University of Florida, Gainesville
biomaterials including metals, and stable covalent March 31, 1992.
biomolecules coupled to these surfaces. In our opinion, the 13. Andrade JD, Triolo PM. Surface modification and evalu-
closest thing to a biophilic commercially available polymer ation of some commonly used catheter materials. I. J
in the near future would be one that maintains high me- Biomed Mater Res 1983; 17:129-247.
chanical performance characteristics of existing polymers 14. Suzuke M, Kishida A, Iwata H, Ikada Y. Graft copoly-
and contains pendant reactive groups that can have merization of acrylamide onto a polyethylene surface pre-
biomolecules coupled to these functionalities in an economic treated with a glow discharge. Macromolecules 1986;
19:1804-1808.
process. It is doubtful that a biomolecule could be reacted
15. Dias AJ, McCarthy TJ. Synthesis of a two-dimensional ar-
into a material and still maintain its activity through pro- ray of organic functional groups: Surface-selective modi-
cessing. It would be also difficult to expect optimal presen- fications of poly(vinylidene). Macromolecules 1984;
tation of the biomolecule to the blood or tissue. It may be 17:2529-2531.
possible for certain molecules or drugs that are targeted for 16. Okada T, Ikada Y. Tissue reactions to subcutaneously im-
release, but probably not for scaffolding applications. planted, surface modified silicones. J Biomed Mater Res
It is hoped that this chapter gives some useful infor- 1993; 27:1509-1518.
mation regarding several of the methods employed to make 17. Okada T, Ikada Y. In vitro and in vivo digestion of col-
surfaces more biocompatible, and some appreciation for lagen covalently immobilized onto the silicone surface. J
problems and opportunities that are attendant upon these Biomed Mater Res 1992; 26:1569-1581.
methods. Knowledge of several techniques can allow for a 18. Okada T, Tamada Y, Ikada Y. Surface modification of sili-
cone for tissue adhesion. Biomaterials and Clinical Ap-
number of combined processes that can bring optimal per-
plications, 1987.
formance as well as economic reality to creating biocom- 19. Whiteside et al. Langmuir, 1988; 4:365.
patible surfaces. 20. Gauckler L. Personal communication at The Monte Verita
Conference on Biocompatible Materials Systems. Ascona,
References Switzerland, 1993: October 11.
1. Sawyer PN. Surface charge and thrombosis. Ann NY Acad 21. Csomor K, Karpati E, Nagy M, Gyorgyi-Edeleny J,
Sci 1984; 416: 561-584. Machovich R. Blood coagulation is inhibited by sulphated
2. Chin TH, Nyilas E, Turcotte LR. Microcalorimetric and copolymers of vinyl alcohol and acrylic acid under in vitro
electrophoretic studies of protein sorption. Trans Am as well as in vivo conditions. Thrombosis Research, 1994;
SocArtif Intern Organs 1978; 24:389-402. 74(4):389-398.
3. Andrade JD. Interfacial phenomena and biomaterials. Med
Instrum 1976; 7:110-120.
488 Tissue Engineering of Prosthetic Vascular Grafts
22. van Boeckel CAA. From heparin to a synthetic drug: A 30. Cahalan L et al. Biocompatible medical article and
multi-disciplinary approach. Trends in Receptor Research. method. U.S. Patent 5,607,475. Assignee: Medtronic Inc.
Elsevier Publishers B.V. 1993. 1997:March 4.
23. Ishihara K, Nakabayashi N, Nishida K, Sakakida M, 31. Knowles JR. Accounts of Chemical Research 1972;
Shchiri M. Designing biocompatible materials. Chemtech, 5:90-119.
1993. 32. Guire P, Fliger D, Hodgson J. Photochemical coupling of
24. Sioshansi P. Ion beam modification of materials for bio- enzymes to mammalian cells. Pharmacological Research
medical application. Seminar: Biomaterials: Medical and Communications, 1977; 9(2).
Pharmaceutical Applications, sponsored by Technomic 33. Guire PE. Binding reagents and methods. U.S. Patent
Publishing Company, Inc, 851 New Holland Ave., 4,722,906, Assignee: Bio-Metric Systems Inc. 1988:Feb. 2.
Lancaster, PA: 1990. 34. Annual Report (July 1, 1971-June 30, 1972), Medical De-
25. Larsson N, Senius P, Eriksson JC, Maripuu R, Lindberg vices Applications Program of the National Heart & Lung
B. J Colloid Interface Sci, 1982; 90:127-136. Institute, Bethesda, MD.
26. Yun JK, DeFife K, Colton E, Stack S, Azeez A, Cahalan L, 35. Luttinger M, Cooper CW. J Biomed Mater Res 1967; 1:67.
Verhoeven M, Cahalan PL, Anderson JM. Human mono- 36. Ratner BD, Horbett T, Hoffman AS. Cell adhesion to
cyte/macrophage adhesion and cytokine production on polymeric materials: Implications with respect to
surface-modified poly(tetrafluoroethylene/ hexafluoro- biocompatibility. J Biomed Mater Res, 1975; 9:407-422.
propylene) polymers with and without protein 37. Hendriks M. A study on the covalent surface-immobili-
preadsorption. J Biomed Mater Res, 1995; 29:257-268. zation of collagen. Development of biomaterials with en-
27. Pitt WG, Cooper SL. Albumin adsorption on alkyl chain hanced infection resistance; a surface modification ap-
derivatized polyurethanes: I. The effect of C-18 alkylation. proach, Ph.D. Thesis from University of Eindhoven, 1966.
J Biomed Mater Res 1988; 22:359-382. 38. von Recum AF, Park JDB. Permanent percutaneous de-
28. Yamauchi J, Yamaoka A, Ikemoto K, Matsui T. J Appl vices, CRC Crit Rev Bioeng 1981; 5:37-77.
Polym Sci 1991; 43:1197-1203. 39. von Recum AF, van Kooten TG. The influence of micro-
29. Kulik E et al. Poly(ethylene glycol) enriched surface: Re- topography on cellular response and the implications for
duced vs delayed protein adsorption and activation. Ab- silicone implants. J Biomater Sci Polymer Edn 1995;
stract from Firth World Biomaterials Congress. Toronto, 7:181-198.
Canada: 1996:May 29-June 2.
Scaffold Engineering
Material Axpects
CHAPTER 45
Bioresorbable Grafts: A Counterintuitive Approach
David Fox, David A. Vorp, Howard P. Greisler
Overview
T his chapter reviews the use of bioresorbable materials in vascular grafting. First, the theo
retical basis for the use of bioresorbable materials is presented. Next, the various materi-
als and the results of experimental work with them are discussed. Bioresorbable materials
have been incorporated into vascular prostheses in a number of novel ways. Accordingly, the
chapter is organized primarily by the manner in which bioresorbable materials have been
incorporated. These include the bioresorbable material as a stand-alone graft, in combina-
tion with nonresorbable materials as a partially resorbable bi-component, or compound
graft, and finally as a supportive scaffold over a biological graft. The chapter also discusses
the use of biological grafts that have been chemically modified so as to be at least partially
resorbable.
Definitions
The terms biodegradable, bioresorbable and bioabsorbable have been used relatively
interchangeably in the literature reporting this heterogeneous class of biomaterials. An at-
tempt was made to reach a consensus on precise definitions at The First International Scien-
tific Consensus Workshop on Degradable Materials held in Toronto in 1989. Biodegradation
was defined as “loss of a property (of a biomaterial) caused by a biological agent.” The Work-
shop was unable, however, to agree on definitions of degradation, bioabsorption and
bioresorption.1 Therefore, in this chapter the terms biodegradable, bioresorbable and
bioabsorbable are in general applied in accordance with the original authors, without impli-
cation of specific mechanisms of degradation.
Introduction
Despite the successful application of prosthetic grafts in the replacement of large ar-
teries, the performance of prosthetics as medium and small caliber replacements has been
less than satisfactory, resulting in both early and late graft failures, increasing the need for
reoperation and limb loss. Factors contributing to these failures include thrombogenicity,
inadequate tissue ingrowth and hyperproliferation of tissue with deposition of extracellular
matrix resulting in myointimal hyperplasia.2
Biodegradable grafts have been investigated as a potential alternative to conventional
biostable prosthetic grafts. The concept of a biodegradable graft emerged from the work of
Arthur Voorhees’ group at Columbia University. In 1954 Voorhees reported the first truly
successful clinical application of prosthetic arterial prostheses, replacing 17 abdominal aor-
tas and 1 popliteal aneurysm with grafts composed of Vinyon-N cloth tubes.3 The success of
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
490 Tissue Engineering of Prosthetic Vascular Grafts
the Vinyon-N fabric prostheses was in large part a function Single Component Resorbable Grafts
of its porosity, which allowed the ingress of capillaries and The materials most commonly used in the fabrication
fibroblasts. This granulation tissue served as the nidus for of resorbable grafts have been synthetic polymers based upon
the formation of an organized inner surface of flattened naturally occurring hydroxy acids such as lactic and glycolic
cells.4 Thus, it became apparent that the graft material itself acid.11 The rate of resorption of these copolymers is deter-
may be needed only transiently, to function as a scaffolding mined to a degree by the ratio of lactide to glycolide rings
for regeneration of the vascular wall rather than as a perma- and decreases as the percentage of lactide component in-
nent conduit. creases.12 The primary mechanism of resorption is by the
After Voorhees’ success, many other materials were hydrolytic cleavage of ester bonds. Inflammatory cells such
proposed. Sigmund Wesolowski and his co-workers, work- as macrophages, lymphocytes and neutrophils perform the
ing at Walter Reed, evaluated 45 prospective materials and final degradation and resorption.11
confirmed the concept that the porosity of a graft material
and not its inertness determined the success of a prosthetic Polyglactin 910 (Vicryl)
vascular graft.5 Prior to this time, the prevailing concept was Bowald’s group from Sweden is credited with the first
that the biological inertness of a prosthetic material was the report of the use of a totally biodegradable prosthesis as a
critical determinant of its clinical success as a vascular graft.6 stand-alone replacement for large caliber vessels.7 Knitted
Wesolowski determined that grafts constructed of materials mesh composed of Polyglactin 910 (PG910), “Vicryl,” manu-
of relatively high porosity were resistant to late calcification factured by Ethicon Inc., was preclotted and interposed as
and occlusion. The problem of hemorrhage during implan- tube and patch grafts into the thoracic aorta of growing pigs.
tation, however, remained an important obstacle. To over- PG910 is a polymer composed of glycolic and lactic acids
come this problem, the concept of constructing a compos- and thus is absorbed by hydrolysis within 70 days.13 The
ite graft emerged. The ideal composite graft would exhibit mesh had a pore size of 400 x 400 microns.14
the property of low porosity at implantation, by virtue of a The grafts were explanted at intervals up to 6 weeks.
degradable component combined with a permanent com- All grafts remained patent and no instances of graft rupture
ponent of high porosity. Over time, the degradable compo- or aneurysmal dilatation occurred. At 40 days only small
nent would be depleted, leaving a high porosity graft ca- fragments of the PG910 remained and the grafted areas dis-
pable of being incorporated by tissue.7 played some similarities to normal aortic walls. The lumi-
The feasibility of incorporating absorbable collagen nal surface was covered with a monolayer of mature endo-
and gelatin components into Dacron grafts had been re- thelial-like cells in a mosaic pattern. Beneath this was a layer
ported by Humphries and Bascon in 1961.8,9 Weselowski of longitudinally directed smooth muscle-like cells. No in-
took a different approach, suggesting the use of a tempo- ternal elastic lamina was seen, although scattered fibrillar
rary scaffold fabricated from a slowly absorbable polymeric elastic material was found interspersed amongst the smooth
material as a vascular graft.5 This scaffold would enable the muscle-like cells. An outer “adventitial” layer was reconsti-
arterial wall to reorganize by virtue of natural repair pro- tuted as well.13 With respect to the source of the repopulat-
cesses.7 The American physiologist Claude Guthrie has been ing cells, Bowald observed that the endothelium and smooth
credited with the origin of this concept. In 1919 he wrote, muscle-like cells seemed to be derived from the cut edges of
“To restore and maintain mechanical function an implanted the native vessel. Occasional inflammatory and giant cells
segment only temporarily restores mechanical continuity were noted around residual PG910 filaments in the 20 day
and serves as a scaffolding or bridge for the laying down of explants.14
an ingrowth of tissue derived from the host.”10 Late results after 2 years were reported for one pig that
Since Wesolowski’s report, many additional biode- received a 10 mm by 4 cm tube graft in the thoracic aorta.
gradable materials have been proposed for use as compo- The graft was patent angiographically. On gross examina-
nents in the fabrication of vascular prostheses. These mate- tion it was free of thrombus, calcification or fatty infiltra-
rials have been incorporated into grafts in a number of in- tion. Of note, the diameter of the grafted area had increased
teresting and novel ways that will be described in this chapter. proportionally in size with the nongrafted aorta and was
The majority of investigators have used the biodegradable not aneurysmal. They also reported reconstitution of a coarse
material as a stand-alone graft. Others have constructed com- internal elastic lamina.15
pound grafts wherein the degradable material is combined Based on these observations, Bowald sought to deter-
with a nonabsorbable prosthetic or biological graft. The mine the maximal regenerative capacity of arterial tissue.
degradable materials have been applied as a lining or wrap To this end, the descending thoracic aorta of 20 pigs was
and in some cases have been interweaved with the prosthetic. bypassed with 8-10 mm diameter double layer Vicryl seg-
The biomaterials used in the fabrication of ments ranging from 7-20 cm in length. All animals receiv-
bioresorbable vascular grafts have a unique dual function. ing a graft greater than 15 cm in length died from graft rup-
They serve as temporary vascular conduits while simulta- ture. There were no ruptures in the animals receiving grafts
neously inducing the regeneration of the arterial wall. As less than 15 cm and the histologic appearance of these grafts
will be elaborated, complex interactions between the was equivalent to those in the previous reports.16
bioresorbable material and host macrophages are a critical Extensive implant and in vitro work with lactic and
step in the process of arterial regeneration. glycolide copolymers have also been done by our group in
the research laboratories of Loyola University. We have in-
Bioresorbable Grafts: A Counterintuitive Approach 491
vestigated PG910 and other copolymers including turally primitive mesenchymal cells that differentiated into
polyglycolic acid (PGA) and polydioxonone (PDS), as well smooth muscle-like myofibroblasts and the establishment
as combinations of these polymers. PG910 grafts were im- of a confluent endothelial-like luminal lining cells was dem-
planted into the abdominal aorta of rabbits. Likewise, the onstrated, thus confirming Bowald’s earlier observations
transinterstitial migration and proliferation of ultrastruc- (Figs. 45.1A-C).14 This process was found to parallel the time
course of macrophage-mediated prosthetic dissolution. Peak ated aorta was unknown, as was the atherogenicity of the
inner capsule (IC) thickness was achieved at 2 months. Again, vessels.19
there was no graft related morbidity or mortality. Twenty Stimulated by these early results, our group compared
percent of the PG910 grafts become aneurysmal.17 Dacron aortic prostheses to PGA. The Dacron grafts were
In order to further assess the potential contribution fabricated to specifications that closely matched the physi-
of transanastomotic pannus ingrowth to the arterial regen- cal characteristics of woven PGA, most notably in terms of
eration seen, prostheses were constructed of three 10 mm pore diameter, pore area and wall thickness. Over the 12
segments with a PG910 segment interposed between two month observation period both the PGA and Dacron grafts
Dacron segments. Grafts were again implanted in rabbit remained free of thrombosis, infection or hemorrhagic com-
aortas and harvested at intervals up to 4 months. All grafts plications. Again, 10% of PGA grafts showed moderate an-
remained patent with no aneurysms or stenoses. Inner cap- eurysmal dilatation and 15% demonstrated significant inti-
sule thickness in the PG910 segments increased only during mal hyperplasia.20
the interval from 2 weeks to 2 months and was statistically After three months the PGA, as before, was found to
greater than either Dacron segment at 1 and 2 months. In- have been resorbed and in its place a thick, dense neointima
ner capsules of PG910 segments at 1 month were composed lined with a confluent layer of endothelial cells was seen.
predominantly of myofibroblasts. Pannus ingrowth was lim- Beneath this layer were numerous smooth muscle like
ited to the first 2 mm of the inner capsules of the Dacron myofibroblasts in a circumferential and longitudinal orien-
segments, the remainder consisting of a fibrin coagulum. tation. The inner capsules of the Dacron grafts, however,
These findings essentially ruled out transanastomotic pan- exhibited only matrix and small numbers of myofibroblasts
nus ingrowth as the primary source of cells replacing ab- in a radial orientation. The flow surface was thin, fibrinous
sorbable vascular prostheses.18 and acellular.
The Dacron grafts explanted at 1 year contained ex-
Polyglycolic Acid (Dexon) tensive calcium deposits and some advanced atherosclerotic
In 1982, we reported the regeneration of major com- plaques. The PGA explants at 1 year remained remarkably
ponents of the rabbit aorta over a scaffold of absorbable stable with the exception of an occasional lipid-laden his-
porous polyglycolic acid (PGA). PGA has the simplest struc- tiocyte, as had been previously observed.
ture of the linear aliphatic polyesters. It is hydrophilic and Despite their similarities in mechanical porosity, these
loses its strength rapidly over 2-4 weeks.11 Woven PGA, two graft materials achieved remarkably different histologic
manufactured by Davis & Geck, was fabricated into 3.5 mm outcomes, suggesting that porosity was not the only factor
diameter grafts and implanted into the abdominal aorta of active in graft healing. Insight into the pathophysiology of
rabbits and explanted over a seven and a half month period. these differences was gained by studying the grafts explanted
Seventy-six percent (76%) of the animals were found to have after two weeks.
maintained a lumen of 3-4 mm in the region of implanta- As in the previous experiment, an inflammatory re-
tion. Eleven percent (11%) of the animals demonstrated an- action containing macrophages, polymorphonuclear leuko-
eurysmal dilatation in this region; however, no animal died cytes and giant cells was observed in the tissues surround-
of graft rupture. In addition, there was no evidence of ing the PGA grafts. Additionally, numerous macrophages
perigraft hematomas or of false aneurysm formation. There were found within the mesh of the PGA grafts. The Dacron
were no instances of graft thrombosis or infection. Experi- grafts as well were surrounded by an inflammatory reac-
mental regenerated aortas were subjected to bursting tion; however, cellular infiltration was limited to the outer
strength determinations and withstood hemodynamic forces capsule with little or no permeation of the woven Dacron. It
at three to five times systolic pressure. was hypothesized that phagocytosis of PGA by macrophages
Histologic analysis revealed some marked similarities inducing their phenotypic alteration and synthesis of growth
to native aorta. The regenerated aortas were composed of a factors chemotactic and mitogenic for endothelial cells and
luminal surface containing factor VIII positive endothelial myofibroblasts, as well as subtle differences in the surface
cells, a subendothelial zone containing smooth muscle-like electronegativity of the materials, could have accounted for
myofibroblasts amidst dense fibrous tissue and an outer layer differences in transinterstitial migration of inflammatory
of vascularized connective tissue. Elastin regeneration, how- cells.20
ever, was minimal. Some lipid-laden macrophages and his- Additional speculations were made regarding the role
tiocytes were observed in the six month explants, suggest- of growth factors in inducing myofibroblast and endothe-
ing possible early atherogenesis. Intimal hyperplasia result- lial cell ingrowth and deposition. Macrophages were known
ing in severe graft stenosis was observed in 13% of animals. to stimulate the proliferation of endothelial cells, smooth
The PGA graft material underwent progressive disso- muscle and fibroblasts through the elucidation of “macro-
lution. At two weeks the PGA was densely invaded by mes- phage-derived growth factors (MDGFs).”21-23 The degree of
enchymal cells, histiocytes and giant cells. Complete resorp- macrophage infiltration corresponded temporally with
tion occurred between three weeks and three months. myofibroblast proliferation as quantitated by autoradiogra-
Important questions emerged from this study. Prin- phy with tritiated thymidine and suggested the possibility
cipally, it remained to be defined what were the factors re- that a macrophage derived factor played an important role
sponsible for the initiation and modulation of the repair in the modulation of myofibroblast response. The possibil-
process. Additionally, the origin of the cells in the regener- ity that the degradable materials had directly resulted in
Bioresorbable Grafts: A Counterintuitive Approach 493
macrophage stimulation was suggested by the demonstra- to have real potential for improved efficacy over prosthetics
tion of PGA remnants within the cytoplasm of macrophages in current use.
by TEM. Additional speculations surrounded the potential
roles of PDGF, ECGF (now FGF-1), and FGF-2 and of me- Characteristics of the Healing Response
chanical forces on the tissue bed, which were found to in- In addition to rate of resorption and kinetics of IC
crease as the PGA degrades and the integrity and mechani- formation, additional studies were performed in order to
cal strength of the graft material was lost.20 further characterize differences between the healing re-
sponses induced by the various materials. The prostaglan-
Polydioxanone (Pds) din metabolite contents of inner capsules formed in the pres-
The ability of lactide/glycolide polymers to induce ence of PDS, PG910 and Dacron containing grafts were com-
arterial regeneration was also demonstrated with grafts fab- pared. PDS inner capsules were found to have greater ratios
ricated from PDS, a more slowly resorbed polymer.24 Grafts, of 6-keto-PGF1#:TxB2 as compared to PG910 and Dacron
24 x 4 mm in size and composed of woven PDS were again implants. Notably, the ratio of 6-keto-PGF1#:TxB2 for PDS
interposed into the abdominal aorta of rabbits. The animals was nearly identical to its normal aortic control. This favor-
were studied at intervals up to one year. Again, no aortic- able ratio suggested that grafts containing PDS may be less
related deaths or hemorrhages occurred. Aneurysmal dila- thrombogenic and again suggests the potential for clinical
tation developed in 10% of specimens, whereas stenosis from efficacy of resorbable grafts.31
neointimal hyperplasia occurred in 15%. As with PGA The kinetics of collagen deposition induced by PDS,
(Fig. 45.2), a vessel wall composed of myofibroblasts and PG910 and Dacron containing grafts was determined by
matrix covered by a confluent endothelial-like flow surface examining the inner capsules of grafts explanted at 1, 3 and
resulted. The PDS implants (Figs. 45.3 and 45.4) yielded an 12 months. In the Dacron grafts, collagen formation peaked
inner capsule (IC) thickness as well as compliance and at 1 month at approximately 12 ∝g/mg. In contrast, the col-
strength characteristics that were comparable to the PGA lagen content of the inner capsules of the PDS and PG910
grafts. Not unexpectedly, tissue formation with the PDS grafts at 1 month was around 27 ∝g/mg. This amount of
grafts occurred more gradually. These grafts reached maxi- collagen is essentially equivalent to that found in normal
mal IC thickness between 3 and 6 months as compared to 2 infrarenal rabbit aorta. The level of collagen in the Dacron
months for PGA. The difference in the rates paralleled the grafts at one year remained the same as at 1 month. The
differing dissolution rates of the two materials.25 These ob- level of collagen in the PDS and PG910 grafts increased to
servations reinforced the concept that the process of arte- approximately 38 ∝g/mg at 3 months, after which it stabi-
rial regeneration was regulated by some factor or factors re- lized.32
lated to the absorption of the material by macrophages. The kinetics of myofibroblast proliferation in the in-
ner capsules of PDS, PG910 and Dacron implants was also
Mechanical Stimuli of Arterial Regeneration analyzed, again in the rabbit infrarenal aorta model. Grafts
A study by Vorp et al to evaluate the stress history were explanted at intervals up to 52 weeks. Mitotic indices
within the inner capsule demonstrated that, early in the re- were determined by autoradiography using tritiated thymi-
sorption phase, the neointima is under a state of circumfer- dine. Of interest was the finding that the mitotic index asso-
ential compression.26 Later in the resorption phase, as the ciated with each graft material was parallel to that material’s
graft material weakens upon dissolution and as the rate of resorption. The peak mitotic index for PG910 was
neointimal tissue strengthens upon reorganization, the in- 28.34 in a 24 h period and occurred at 3 weeks postimplant,
ner capsule stress becomes tensile. The occurrence of com- whereas the peak for PDS was 7.50; this occurred at 4 weeks
pressive stresses parallels the time point and location of and was prolonged. There was little evidence of inner cap-
maximal mitotic index for the neointimal tissue. We showed sule formation in the Dacron grafts until 12 weeks
using the well established rabbit model that the mitotic in- postimplant and the mitotic indices for Dacron grafts never
dex of inner capsule myofibroblasts in PG910 grafts was sig- exceeded 1.22.27
nificantly higher during the early period of resorption than
in the later period.27 Macrophage/Biomaterial Interactions
Using a pulse duplicator apparatus developed at the In order to further characterize the contribution of
University of Pittsburgh,28 changes in the dynamic compli- the macrophage/biomaterial interaction in the elaboration
ance of PG910 grafts occurring over 0 to 36 weeks were as- of factors regulating the process of arterial regeneration, a
sessed.29 Using the rabbit model, explanted PG910 grafts series of in vitro experiments were performed.33-36 In each
were exposed to realistic pulsatile hemodynamics in vitro. A series of experiments, rabbit peritoneal macrophages were
rapid increase in compliance was noted after 3 weeks, level- harvested and cultured in Minimum Essential Medium
ing off at 13 weeks. In a companion study, compliance in- (MEM) with platelet-poor serum and containing particles
creased over time in the proximal and distal graft segments of Dacron, polyglactin 910 or no biomaterial (Fig. 45.5A,B).
compared to baseline. Loss of compliance has been cited as MEM conditioned in this way was collected and mitogenic-
an important mechanism in the failure of small and me- ity assays were performed with quiescent fibroblasts
dium sized Dacron and ePTFE implants.30 Thus, by achiev- (BALB/c3T3), rabbit aortic smooth muscle cells, and mu-
ing greater compliance over time, resorbable grafts appear rine capillary lung (LE-II) endothelial cells. Mitogenic ac-
Bioresorbable Grafts: A Counterintuitive Approach 495
tivity was assayed by scintillation counting of tritiated thy- rophages exposed either to Dacron or to no biomaterial. In
midine incorporation into deoxyribonucleic acid (DNA). In fact, it could be demonstrated that macrophages harvested
order to examine these processes in a model more accurately from rabbits fed a cholesterol rich atherogenic diet exposed
reflecting the clinical setting, macrophages were also har- in culture to Dacron produced conditioned media that ex-
vested from rabbits that had been rendered hyperlipidemic erted an inhibitory effect on endothelial cell thymidine in-
through an atherosclerotic diet.33-35 corporation. In order to isolate the factor or factors respon-
All cell types grown in conditioned media from mac- sible for these alterations in mitogenic activity, Western blot
rophages exposed to PG910 demonstrated significantly analysis was performed using antibodies raised against
greater increases in tritiated thymidine incorporation as PDGF-B chain, acidic fibroblast growth factor (FGF-1) and
compared to those grown in media conditioned from mac- basic fibroblast growth factor (FGF-2). Only anti-bFGF
496 Tissue Engineering of Prosthetic Vascular Grafts
antibody immunoreacted with samples of the conditioned smooth muscle cells grown in conditioned media from mac-
media.33 rophages exposed to Dacron (Fig. 45.6A). When macro-
In order to further define the contribution of bFGF phages were obtained from atherosclerotic rabbits a gener-
to the mitogenic response, conditioned media was alized decrease in the incorporation of tritiated thymidine
preincubated with neutralizing anti-basic-FGF antibody. In by smooth muscle cells occurred in both the Dacron and
rabbits fed normal diets, Dacron induced mitogen release PG910 groups. The macrophages from atherosclerotic rab-
was completely inhibited by anti-basic-FGF antibody. PG910 bits cultured with Dacron, however, showed a particular in-
mitogen release was diminished, but to a significantly less crease in the mitogenic factors most active on the smooth
degree (36%). Thus it appeared that much of the mitogenic muscle cells (Fig. 45.6B). To further characterize this effect,
effect on smooth muscle cell was mediated via basic-FGF.35 Western blot and immunoprecipitation studies were per-
In another series of experiments, the effect of condi- formed, and as before, the presence of FGF-2 was demon-
tioned media on rabbit aortic smooth muscle cells was as- strated, whereas PDGF and FGF-1 were not. TGF-! inhibi-
sessed. Smooth muscle cells grown in conditioned media tory activity was demonstrated to be increased for the ath-
from macrophages exposed to PG910 again resulted in sig- erosclerotic macrophage containing media. These findings
nificantly higher tritiated thymidine incorporation than. support the hypothesis that interactions between Dacron,
Bioresorbable Grafts: A Counterintuitive Approach 497
macrophage and smooth muscle cells play a role in the patho- all groups; however, only the PU/PLLA grafts reconstituted
genesis of graft failure.34 an elastic lamina. They concluded that both biodegradation
and compliance were important stimuli of arterial
Polyurethane/Poly-L-Lactide Copolymers (Pu-Plla) regeneration.40
A different approach has been taken by the polymer To characterize the functional characteristics of the
chemists at the University of Groningen in The Netherlands. regenerated arteries, endothelialization and prostacyclin syn-
They have worked extensively with copolymers of polyure- thesis were determined using a bioassay and RIA for the
thane and poly-L-lactide (PU-PLLA) with the goal of com- prostacyclin metabolite 6-oxo-PGF1#. Comparisons were
bining the properties of the two materials to produce an anti- made between PU/PLLA and PTFE grafts and normal en-
thrombogenic, flexible as well as resorbable material. PU- dothelium. Looking at prostacyclin production per unit graft
PLLA was described by Gogolewski in 1982.37 area covered with neoendothelium, there were no signifi-
Lommen et al interposed 1.5 x 10 mm PU-PLLA grafts cant differences between the two graft types and native ar-
into the abdominal aorta of rats.2 These grafts were of tery. Endothelialization and healing, however, as determined
40-50 ∝m porosity and were explanted at 3, 6 and 12 weeks. by light and electron microscopy were incomplete in the
These grafts were compared to a control group that received PTFE grafts, even after 12 weeks.41 Additional ultrastruc-
PTFE grafts of the same dimensions and of 30 ∝m porosity. tural analyses were performed and, as was observed by our
At explant all grafts were patent and there was no evi- group, epithelioid cells and multinucleated giant cells were
dence of aneurysm formation, dilatation or other compli- seen to engulf polymer particles of the disintegrating grafts.42
cations in either group. By 6 weeks the PTFE grafts had de- The long term fate of PU/PLLA grafts was assessed in
veloped a complete “endothelial” lining that was continu- a group of 8 implants. After 1 year all grafts were found to
ous with that of the adjacent vessel. However, this lining be patent. Three of the grafts were of normal shape and were
detached easily during processing, suggesting that there was visibly pulsatile. Two grafts were mildly dilated, 2 were an-
poor attachment with the prosthesis. The internodal spaces eurysmal and 1 had sustained a dissection. These grafts were
of the graft appeared to contain fibrin. Similar findings were not visibly pulsatile. Histologic analysis of the “neomedia”
noted in the 12 week PTFE explants. revealed circularly arranged smooth muscle cells (as in nor-
The PU-PLLA grafts demonstrated arterial regenera- mal artery) in the normally shaped implants. However, in
tion. A complete “endothelial” lining was observed at three the nonpulsatile implants, the neomedia had a predomi-
weeks. Likewise, the outer surface of the grafts was covered nantly longitudinal arrangement. They concluded that the
with vascularized connective tissue that was morphologi- pattern of arrangement of smooth muscle cells in the
cally indistinct from native adventitia. At 6 weeks giant cell neomedia affects the fate of the regenerated arteries.43
activity was observed within the graft walls, most promi- There was uncertainty as to the origin of the cells in
nently at the periphery. By 6-12 weeks a subintimal layer the regenerated arteries. Hypothesized origins have included
containing “smooth muscle cells,” collagen and elastin (dem- migration from the cut ends of the adjacent vessel and the
onstrated by Orcein staining) had developed. Multiple fe- granulation tissue surrounding the graft, as well as deposi-
nestrated elastic laminae were observed as well. The body of tion of circulating cells onto the luminal surface.44 To gain
the PU-PLLA graft gradually became infiltrated with cells, insight into the source origin of the cells in the arteries re-
and this process was associated with progressive degrada- generated from PU/PLLA, sequential explants at intervals
tion of the graft substance.2 of 1 h to twelve weeks were performed. They observed that
Having demonstrated that biodegradable PU-PLLA the endothelium originated from adjacent intima. The
stimulates the regeneration of an arterial wall they next smooth muscle cells seemed to originate from the media of
sought to optimize some of the properties of the grafts re- the adjacent aorta; however, they could not exclude the other
sponsible for this process. They postulated that the compli- postulated mechanisms which would be required for clini-
ance of the grafts was an important property and they per- cal applicability, and proposed experiments with longer
formed in vitro stress/strain measurements of grafts com- grafts to decrease the likelihood of trans-anastomotic cell
posed of several combinations of PU and PLLA and com- migration.44
pared them to native artery. They determined that grafts Other work with lactide polymers has been done by
composed of 5-20% PLLA possessed compliance before Hanson’s group at the University of Texas. They fabricated
implantation comparable to rat aorta. 38 They subsequently grafts with copolymers of L-lactide, DL-lactide, and
investigated the effect of varying the pore size of grafts com- >-caprolactone in various compositions. Lactide polymers
posed of 95%/5% PU/PLLA in the rat model. They deter- were chosen for their strength, >-caprolactone for their flex-
mined that there was no distinct advantage of a pore size of ibility. The grafts were 3 mm by 8 cm, nonwoven, nonpo-
40 ∝m as compared to pores in a gradient of 10-100 ∝m from rous and completely resorbable. The mechanical properties
the inner to the outer regions of the graft in terms of arterial and degradation rates of the grafts were evaluated in vitro.
regeneration. The advocated the use of a pore gradient, how- Circumferential tension, compliance, and kink angle were
ever, as the smaller pores on the luminal surface were thought determined. Tensile strength and modulus of elasticity were
to decrease thrombogenicity.39 found to be in the range of normal artery; however, the grafts
They compared these compliant grafts to less compli- were significantly less compliant than artery. It was antici-
ant grafts composed of biostable PU and of PU/PLLA sur- pated that the addition of porosity, a necessity for in vivo
rounded by PTFE. Partial arterial regeneration was seen in
498 Tissue Engineering of Prosthetic Vascular Grafts
studies, would improve compliance. Further investigations which more closely approximate the situation of clinical
were planned.45 vascular grafting, resorbable grafts have had an increased
incidence of graft dilation, aneurysm formation and rup-
Polyurethane Elastomers (PU) ture.47-49 Investigators have taken several approaches to
Extensive work has been done in the evaluation of counteracting this problem, including incorporating
small diameter vascular grafts composed of polyurethane resorbables with enhanced resistance to degradation,7,50-56
elastomers. Polyurethanes hold promise by virtue of their increasing the rapidity of tissue regeneration with growth
superior compliance as compared to conventional prosthet- factors57,58 and cell seeding.59,60 Others have taken an inter-
ics. By and large, these polyurethanes are nonresorbable mediate approach and constructed compound or partially
(biostable).46 Gogolewski fabricated vascular grafts from a resorbable vascular prostheses combining resorbable and
bioresorbable segmented aliphatic polyurethane (PU). Like permanent materials.
the PU/PLLA grafts, these were degradable, microporous and
compliant. Grafts of 1.5 mm inner diameter and 0.7 mm Enhancing the Resistance to Degradation
wall thickness were interposed into 13 mm defects in rat of Resorbable Materials
abdominal aorta and explanted at intervals between 3 and Our group evaluated woven bicomponent totally
16 weeks. The luminal surface of the graft consisted of in- bioresorbable grafts fabricated from compound yarns con-
terconnected pores of 5-10 ∝m. The outer surface of the graft taining 74% PG910 with 26% polydioxanone (PDS). The
had a porosity of 20-50 ∝m. The tissue response was reported resorption kinetics of PDS are slower than PG910. Thirty-
as being comparable to that previously observed with seven grafts were implanted into the infrarenal aorta of rab-
polylactide and polyglycolide. Again, prosthetic debris were bits and explanted at intervals between 2 weeks to 12 months.
surrounded by giant cells and macrophages. A “glistening Complete resorbtion of the PG910 occurred by 2 months
neointima” was observed at 3 weeks, with “no tendency” to- and the PDS by 6 months. All grafts were patent at explant.
wards intimal thickening. Smooth muscle cells and elastin One graft developed a 50% stenosis that appeared to be
fibers were observed in the intima. By four months almost thrombotic in origin. No aneurysmal dilatation was seen in
complete degradation of the PU had occurred. Stress/strain any graft. Inner capsule thickness was intermediate to that
analysis revealed mechanical characteristics similar to na- seen with PDS and PG910.51
tive aorta.46 The Groningen group hypothesized that the dilata-
Encouraged by these results, 8 mm by 6 cm grafts were tion observed with their PU/PLLA grafts was related to tech-
implanted into the infrarenal aorta of 50 young pigs. The nical problems with the dip-coating process of graft prepa-
grafts were explanted at intervals of 1 month to 1 year. Eigh- ration.52 They produced a modified 2-ply graft with a more
teen pigs died as a result of prosthetic rupture, infection or biostable inner layer. The inner layer was made highly anti-
thrombosis. Ten pigs received no aspirin and a regular diet. thrombogenic by crosslinking of a mixture of linoleic acid
These grafts were all thrombosed by 90 days. Twenty-five and an aliphatic polyetherurethane with dicumyl peroxide.
pigs received aspirin; 55% of these grafts remained patent The outer ply was a (95/5) mixture of polyesterurethane and
at 90 days. Only 3 were patent at 1 year. Of 15 pigs receiving poly(L-lactide).
a diet high in lipid, 7 remained patent at 1 year. Three of Twenty-two two-ply grafts were implanted in the ab-
these animals were noted to have dilation of the prosthesis dominal aorta of rats. All grafts remained patent at least up
similar to that observed by our group in PG910 grafts placed to 1 year and did not exhibit any aneurysm formation. As in
in the abdominal aorta of rabbits fed an atherogenic diet. 47 the earlier formulations, the inner layer was covered with
It was postulated that the enhanced patency of these grafts endothelial cells and several layers of smooth muscle cells.53
was related to an anti-thrombotic effect of the lipid-rich Galletti’s group at Brown University approached the
diet.48 problem of aneurysmal degeneration of Vicryl grafts by coat-
Histologic analysis again revealed macrophage and ing the yarns used in the fabrication process with slowly re-
giant cell infiltration of the prosthesis and phagocytosis of sorbed polyesters, thus extending their functional life. They
prosthetic debris. A neointima and neomedia containing produced tubular conduits, 1 mm by 3 mm, composed of
longitudinal and transverse smooth muscle cells developed triple ply Vicryl coated with a 1:1 composite of poly-DL-
by 180 days. A fibrous neoadventitia was observed at early lactic acid and poly-2,3-butylene malate or poly-2,3-buty-
time points; however, at 1 year this was comprised of fatty lene fumarate. Coated or uncoated Vicryl grafts were im-
tissue. Elastic fibers dispersed throughout the muscle layer planted subcutaneously in mice. Samples were retrieved at
were observed. The prostheses were completely resorbed by intervals between 2 and 12 weeks and the remaining mass
1 year. Prostacycline synthesis was found to be comparable of polymer implant was analyzed for the presence of Vicryl
to normal aorta with no apparent differences between using gel permeation chromatography.
groups.48,49 After 8 weeks Vicryl was undetectable in the uncoated
implants. Grafts coated with poly-2,3-butylene malate or
Preventing Aneurysmal Dilatation poly-2,3-butylene fumarate, however, retained 48 and 88%
The chief obstacle in the development of a clinically of their Vicryl content respectively.54
useful resorbable graft has been the potential for loss of Galletti’s group next constructed 8 mm by 13 cm grafts
mechanical and structural integrity in the graft prior to the of triple ply Vicryl coated with a 1:1 composite of poly-DL-
regeneration of an arterial wall. Adding to this concern is lactic acid and poly-2,3-butylene fumarate. To minimize
the finding that when evaluated in hyperlipidemic models, oozing through the graft interstices, they added a fourth
Bioresorbable Grafts: A Counterintuitive Approach 499
dilatation was seen when 20% Dacron was used. Cellularity finding to the decreased compliance associated with the
of the regenerated tissue in general was decreased in the pres- PTFE wrap.40
ence of Dacron, suggesting an inhibitory effect of the bio-
material.17 Subsequent work demonstrated decreased pros- Compound Grafts: Polyethylene Oxide
taglandin content of the inner capsules of Dacron/Vicryl and Polylactic Acid Copolymers (PELA)
composite grafts as compared to those of Vicryl grafts.31 The Biomaterials Research Laboratory at The Hebrew
In order to avoid the deleterious effects of Dacron, University of Jerusalem has developed and evaluated com-
compound grafts were constructed using polypropylene, pound vascular prostheses fabricated from resorbable block
which possesses greater biocompatibility and a incites only copolymers of polyethylene oxide and polylactic acid (PELA)
a relatively low-grade inflammatory response.65 Grafts fab- applied as a coating to knitted Dacron and polether ure-
ricated from yarns containing 69% Vicryl (PG910) and 31% thane urea (Lycra) grafts. PELA is an elastomer that fully
polypropylene were implanted into rabbit infrarenal aortas. degrades in 3 weeks after implantation.68-71 The in vivo per-
Over 2 weeks to 12 months all grafts remained patent with- formance of the Dacron grafts was evaluated in dogs as right
out aneurysm formation. Only residual polypropylene re- ventricle to pulmonary artery conduits. Unlike control wo-
mained in the prostheses after 2 months. Production of ven grafts, the PELA coated grafts showed complete trans-
6-keto-PGF1# was in the normal range.64 mural tissue ingrowth after 14 months.68,69
PG910/polypropylene prostheses were also implanted In a separate study, the Hebrew University group com-
into the aorto-iliac positions of dogs. Again, all grafts were pared ePTFE grafts to grafts comprised of Lycra fibers coated
without aneurysm formation over one year. Patency was with PELA. 6 mm by 6 cm grafts were implanted in the ca-
enhanced as compared to Dacron and ePTFE control grafts. nine carotid artery for 90 days. Unlike the ePTFE grafts, the
Arterial regeneration was demonstrated.65 PELA coated grafts remained compliant and demonstrated
successful healing and incorporation. Further work with
Compound Grafts: PGA small caliber prostheses was anticipated.70
Our group also evaluated grafts made of polyglycolic
acid (PGA) fabric reinforced by an outer wrap of Dacron. Resorbable Outer Wraps
In this series, the incidence of aneurysmal dilatation was zero A novel application of resorbable materials in vascu-
with or without the outer wrap up to nine months after lar grafting has been reported by Moritz and the Groningen
implant. Again, Dacron appeared to have an inhibitory ef- group.72-75 Moritz applied a constrictive mesh tube of
fect on the cellularity of the luminal surface and inner cap- resorbable mesh to dilated, otherwise unusable veins with
sule.17 Evaluation of an advanced PGA based compound the goal of inducing neoarterial wall growth. He found that
prosthesis is currently underway in our lab. the meshes can effectively reduce the diameter of a venous
Chu’s group at Cornell also investigated the combi- graft by providing external support to the vessel wall, thus
nation of PGA and Dacron. They produced knitted fabric limiting distension.72 The Groningen group has also inves-
grafts composed of PGA and Dacron fibers blended at vari- tigated biodegradable prostheses as external supports for vein
ous compositional ratios. They studied the properties of grafts. They sought to prevent vein graft wall damage due to
these bicomponent fabrics in vitro. They felt that their most the higher arterial pressures encountered after implantation.
important finding was the achievement of increasing water They found that the prostheses can function as a protective
porosity over time without significant losses in the struc- scaffold for vein grafts in the arterial circulation, reducing
tural integrity and strength of the specimens.67 damage to the vein graft wall and allowing gradual arterial-
Having demonstrated the effectiveness of partially ization.73-75
resorbable arterial prostheses in rabbits, we sought to evalu-
ate longer grafts in a higher order species. 4 mm by 5 cm Biopolymers: Resorbable Prostheses
conduits were woven from composite yarns containing 70% of Biologic Origin
PDS/30% polypropylene and implanted into the aorto-iliac Another novel approach has been taken by several
positions of dogs for periods of up to one year. No aneu- groups in Europe and the United States who have sought to
rysms or perigraft hematomas developed. The patency of develop resorbable prostheses through the chemical modi-
the PDS/polypropylene grafts was significantly greater than fication of arterial xenografts.76-81 Vascular prostheses of
that of Dacron or ePTFE control grafts. Inner capsules were biological origin have been shown to develop aneurysms
completely endothelialized by 1 month. IC cellularity and within several years of implantation. It has been hypoth-
thickness were greater than those within Dacron or ePTFE.65 esized that, if properly processed, xenografts can function
as a bioresorbable scaffold. Should this occur, an orderly re-
Compound Grafts: PU/PLLA generation of structural elements during healing could take
The Groningen group investigated compound grafts place, the result being continued mechanical integrity of the
of PTFE fitted around PU/PLLA. They found that these graft.78
grafts, as compared to grafts composed entirely of The biopolymer that has received the most attention
bioresorbable PU/PLLA or biostable but compliant PU, had is aldehyde crosslinked collagen.76 Moczar’s group in France
diminished regeneration of all layers of the arterial wall, most developed a biodegradable microarterial graft from rat aorta.
strikingly in terms of elastic lamina. They attributed this Trypsin treated arterial segments were coated with heparin
or chondroitin sulfate to reduce thrombogenicity. Grafts
Bioresorbable Grafts: A Counterintuitive Approach 501
were implanted into the infrarenal aorta of rats for 3-8 weeks. 15. Audell L, Bowald S, Busch C, Eriksson I. Polyglactin mesh
The grafts were found to undergo resorption in parallel with grafting of the pig aorta. The two-year follow-up in an
re-endothelialization and scar tissue formation. Patency was experimental animal. Acta Chir Scand 1980; 146:97-9.
around 50%.76 Macromolecular repair of the host aortic wall 16. Bowald S, Busch C, Eriksson I. Absorbable material in
vascular prostheses: A new device. Acta Chir Scand 1980;
was documented in one year explants by the incorporation
146:391-5.
pattern of [3H]valine into proteins and the demonstration 17. Greisler HP, Schwarcz TH, Ellinger J, Kim DU. Dacron
of de novo elastin synthesis in the scar replacing the pros- inhibition of arterial regenerative activities. J Vasc Surg
thesis.77 1986; 3:747-56.
18. Greisler HP, Dennis JW, Endean ED, Ellinger J, Buttle
Conclusion KF, Kim DU. Derivation of neointima in vascular grafts.
The resorbable graft represents a paradigm shift in Circulation. 1988; 78:I6-12.
vascular prostheses. Unlike conventional prostheses whose 19. Greisler HP. Arterial regeneration over absorbable pros-
effectiveness depends by and large upon an inherent resis- theses. Arch Surg 1982; 117:1425-31.
tance to a natural healing process, resorbable grafts work 20. Greisler HP, Kim DU, Price JB, Voorhees AB Jr. Arterial
with the body, providing a framework for well established regenerative activity after prosthetic implantation. Arch
Surg 1985; 120:315-23.
healing patterns. It seems intuitive that by augmenting rather
21. Leibovich SJ, Ross R. The role of the macrophage in
than opposing these patterns a more efficacious vascular wound repair. A study with hydrocortisone and
graft should result. Great strides have been made toward this antimacrophage serum. Am J Pathol 1975; 78:71-100.
end. Further research will yield grafts for clinical evaluation 22. Martin BM, Gimbrone MA Jr, Unanue ER, Cotran RS.
in the near future. Stimulation of nonlymphoid mesenchymal cell prolifera-
tion by a macrophage-derived growth factor. J Immunol
References 1981; 126:1510-5.
1. Shalaby S. Degradable materials: Perspectives, issues and 23. Greenburg GB, Hunt TK. The proliferative response in
opportunities. In: Barenberg S, Brash J, Narayan R, vitro of vascular endothelial and smooth muscle cells ex-
Redpath A, eds. The First International Scientific Con- posed to wound fluids and macrophages. J Cell Physiol
sensus Workshop on Degradable Materials. Toronto: CRC 1978; 97:353-60.
Press, 1989:678. 24. Zarge J, Huang P, Greisler H. Blood vessels. In: Lanza R,
2. Lommen E, Gogolewski S, Pennings AJ, Wildevuur CR, Langer R, Chick W, eds. Principles of Tissue Engineer-
Nieuwenhuis P. Development of a neo-artery induced by ing. Georgetown, Texas: RG Landes Company; 1997.
a biodegradable polymeric vascular prosthesis. Trans Am 25. Greisler HP, Ellinger J, Schwarcz TH, Golan J, Raymond
Soc Artif Intern Organs 1983; 29:255-9. RM, Kim DU. Arterial regeneration over polydioxanone
3. Friedman S. A History of Vascular Surgery. Mount Kisco, prostheses in the rabbit. Archives of Surgery 1987;
New York: Futura Publishing Company, Inc., 1989. 122:715-21.
4. Dennis C. Brief history of development of vascular grafts. 26. Vorp DA, Raghavan ML, Borovetz HS, Greisler HP,
In: Sawyer P, ed. Modern Vascular Grafts. New York: Webster MW. Modeling the transmural stress distribution
McGraw-Hill, 1987. during healing of bioresorbable vascular prostheses. Ann
5. Wesolowski SA et al. The compound prosthetic vascular Biomed Eng 1995; 23:178-88.
graft. A pathologic survey. Surgery 1963; 53:19. 27. Greisler HP, Petsikas D, Lam TM et al. Kinetics of cell
6. Fries CC, Wesolowski SA. The polyester-oxidized cellu- proliferation as a function of vascular graft material. J
lose compound vascular prostheses: A preliminary report. Biomed Mater Res 1993; 27:955-61.
Trans Am Soc Artif Intern Organs 1964;X:227-30. 28. Brant AM, Chmielewski JF, Hung T-K, Borovetz HS.
7. Robinson PH, van der Lei B, Knol KE, Pennings AJ. Pa- Simulation in-vitro of pulsatile vascular hemodynamics
tency and long-term biological fate of a two-ply biode- using a CAD/CAM designed cam disk and roller follower.
gradable microarterial prosthesis in the rat. Br J Plast Surg Artif Organs 1986; 10:419-421.
1989; 42:544-9. 29. Pham S, S.J. D, R. J et al. Compliance changes in
8. Humphries A et al. Arterial prosthesis of collagen-impreg- bioresorbable vascular prostheses following implantation.
nated dacron. Surgery 1961; 50:947. Surg Forum 1988; 39:330-332.
9. Bascon J. Gelatin sealing to prevent blood loss from knit- 30. Greisler HP, Joyce KA, Kim DU, Pham SM, Berceli SA,
ted arterial grafts. Surgery 1961; 50:504. Borovetz HS. Spatial and temporal changes in compliance
10. Guthrie C. End-results of arterial restitution with devital- following implantation of bioresorbable vascular grafts. J
ized tissue. J Am Med Assoc 1919; 73:186. Biomed Mater Res 1992; 26:1449-61.
11. Pachence J, Kohn J. Biodegradable polymers for tissue 31. Schwarcz TH, Nussbaum ML, Ellinger J, Kim DU, Greisler
engineering. In: Lanza R, Langer R, Chick W, eds. Prin- HP. Prostaglandin content of tissue lining vascular pros-
ciples of Tissue Engineering. Georgetown, Texas: RG theses. Curr Surg 1987; 44:18-21.
Landes Company, 1997. 32. Greisler HP, Cabusao EB, Lam TM, Murchan PM, Ellinger
12. Greisler HP. Bioresorbable materials and macrophage in- J, Kim DU. Kinetics of collagen deposition within
teractions. J Vasc Surg 1991; 13:748-50. bioresorbable and nonresorbable vascular prostheses.
13. Bowald S, Busch C, Eriksson I. Arterial grafting with ASAIO Trans 1991; 37:M472-5.
polyglactin mesh in pigs [letter]. Lancet. 1978; 1:153. 33. Greisler HP, Ellinger J, Henderson SC et al. The effects
14. Bowald S, Busch C, Eriksson I. Arterial regeneration fol- of an atherogenic diet on macrophage/biomaterial inter-
lowing polyglactin 910 suture mesh grafting. Surgery. actions. J Vasc Surg 1991; 14:10-23.
1979; 86:722-9. 34. Lam TM, Whereat NE, Henderson SC, Burgess WH,
Shaheen A, Greisler HP. Effects of hypercholesterolemia
502 Tissue Engineering of Prosthetic Vascular Grafts
on monokine-induced smooth muscle cell proliferation. 51. Greisler HP, Endean ED, Klosak JJ et al. Polyglactin 910/
Exs 1992; 61:346-56. polydioxanone bicomponent totally resorbable vascular
35. Greisler HP, Henderson SC, Lam TM. Basic fibroblast prostheses. J Vasc Surg 1988; 7:697-705.
growth factor production in vitro by macrophages exposed 52. Leenslag JWK, Michel T, Pennings AJ, Van der Lei B.A
to Dacron and polyglactin 910. J Biomater Sci Polym Ed. complaint, biodegradable vascular graft: basic aspects of
1993; 4:415-30. its construction and biological performance. New Polym
36. Zenni GC, Ellinger J, Lam TM, Greisler HP. Biomaterial- Mater 1988; 1:111-26.
induced macrophage activation and monokine release. J 53. Pennings AJK, K.E., Hoppen HJ, Leenslag JW, Van der
Invest Surg 1994; 7:135-41. Lei B. A two-ply artificial blood vessel of polyurethane
37. Gogolewski S, Pennings A. Porous biomedical materials and poly(L-lactide). Colloid Polym Sci 1990; 268:2-11.
based on mixtures of polylactides and polyurethanes. 54. Galletti PM, Ip TK, Chiu TH, Nyilas E, Trudeli LA, Sasken
Makromol Chem Rapid Commun 1982; 3:839-845. H. Extending the functional life of bioresorbable yarns for
38. Gogolewski S, Pennings AJ, Lommen E, Wildevuur CR. vascular grafts. Trans Am Soc Artif Intern Organs 1984;
Small-caliber biodegradable vascular grafts from 30:399-400.
Groningen. Life Support Syst 1983; 1:382-5. 55. Galletti PM, Trudell LA, Chiu TH et al. Coated
39. van der Lei B, Bartels HL, Nieuwenhuis P, Wildevuur CR. bioresorbable mesh as vascular graft material. Trans Am
Microporous, complaint, biodegradable vascular grafts for Soc Artif Intern Organs 1985; 31:257-63.
the regeneration of the arterial wall in rat abdominal aorta. 56. Galletti PM, Aebischer P, Sasken HF, Goddard MB, Chiu
Surgery 1985; 98:955-63. TH. Experience with fully bioresorbable aortic grafts in
40. van der Lei B, Wildevuur CR, Nieuwenhuis P. Compli- the dog. Surgery 1988; 103:231-41.
ance and biodegradation of vascular grafts stimulate the 57. Greisler HP, Kim DU. Vascular grafting in the manage-
regeneration of elastic laminae in neoarterial tissue: an ment of thrombotic disorders. Semin Thromb Hemost
experimental study in rats. Surgery 1986; 99:45-52. 1989; 15:206-14.
41. van der Lei B, Darius H, Schror K, Nieuwenhuis P, 58. Greisler HP, Klosak J, Dennis JW et al. Endothelial cell
Molenaar I, Wildevuur CR. Arterial wall regeneration in growth factor attachment to biomaterials. ASAIO Trans
small-caliber vascular grafts in rats. Neoendothelial heal- 1986; 32:346-9.
ing and prostacyclin production. J Thorac Cardiovasc Surg 59. Wildevuur CR, van der Lei B, Schakenraad JM. Basic as-
1985; 90:378-86. pects of the regeneration of small-calibre neoarteries in
42. van der Lei B, Wildevuur CR, Nieuwenhuis P et al. Re- biodegradable vascular grafts in rats. Biomaterials 1987;
generation of the arterial wall in microporous, compli- 8:418-22.
ant, biodegradable vascular grafts after implantation into 60. Yue X, van der Lei B, Schakenraad JM et al. Smooth
the rat abdominal aorta. Ultrastructural observations. Cell muscle cell seeding in biodegradable grafts in rats: A new
Tissue Res 1985; 242:569-78. method to enhance the process of arterial wall regenera-
43. van der Lei B, Nieuwenhuis P, Molenaar I, Wildevuur CR. tion. Surgery 1988; 103:206-12.
Long-term biologic fate of neoarteries regenerated in 61. Greisler HP, Klosak JJ, Dennis JW, Karesh SM, Ellinger J,
microporous, compliant, biodegradable, small-caliber vas- Kim DU. Biomaterial pretreatment with ECGF to augment
cular grafts in rats. Surgery 1987; 101:459-67. endothelial cell proliferation. J Vasc Surg 1987; 5:393-9.
44. van der Lei B, Wildevuur CR, Dijk F, Blaauw EH, 62. Gray JL, Kang SS, Zenni GC et al. FGF-1 affixation stimu-
Molenaar I, Nieuwenhuis P. Sequential studies of arterial lates ePTFE endothelialization without intimal hyperpla-
wall regeneration in microporous, compliant, biodegrad- sia. J Surg Res 1994; 57:596-612.
able small-caliber vascular grafts in rats. J Thorac 63. Ruderman RJH, Andrew F, Hattler BG, Leonard F. Par-
Cardiovasc Surg. 1987; 93:695-707. tially biodegradable vascular prosthesis. Trans Am Soc
45. Hanson SJ, Jamshidi K, Eberhart RC. Mechanical evalua- Artif Intern Organs 1972; 18:30-7.
tion of resorbable copolymers for end use as vascular 64. Greisler HP, Kim DU, Dennis JW et al. Compound
grafts. ASAIO Trans 1988; 34:789-93. polyglactin 910/polypropylene small vessel prostheses.
46. Gogolewski SG, G. Degradable, microporous vascular Journal of Vascular Surgery 1987; 5:572-83.
prosthesis from segmented polyurethane. Colloid Polym 65. Greisler HP, Tattersall CW, Klosak JJ, Cabusao EA,
Sci 1986; 264:854-8. Garfield JD, Kim DU. Partially bioresorbable vascular
47. Greisler HP, Klosak JJ, Endean ED, McGurrin JF, Garfield grafts in dogs. Surgery 1991; 110:645-54.
JD, Kim DU. Effects of hypercholesterolemia on healing 66. Zenni GC, Gray JL, Appelgren EO et al. Modulation of
of vascular grafts. J Invest Surg 1991; 4:299-312. myofibroblast proliferation by vascular prosthesis biome-
48. Galletti G, Gogolewski S, Ussia G, Farruggia F. Long-term chanics. Asaio J 1993; 39:M496-500.
patency of regenerated neoaortic wall following the im- 67. Chu CCL, L.E. Design and in vitro testing of newly made
plant of a fully biodegradable polyurethane prosthesis: bicomponent knitted fabrics for vascular surgery. Polym
Experimental lipid diet model in pigs. Ann Vasc Surg Sci Technol 1987; 35:185-213.
1989; 3:236-43. 68. Cohn DY, Hani, Appelbaum Y, Uretzky G. A selectively
49. Galletti G, Ussia G, Farruggia F, Baccarini E, Biagi G, biodegradable vascular graft. Prog Biomed Eng 1988;
Gogolewski S. Prevention of platelet aggregation by di- 5:73-9.
etary polyunsaturated fatty acids in the biodegradable 69. Uretzky G, Appelbaum Y, Younes H et al. Long-term
polyurethane vascular prosthesis: An experimental model evaluation of a new selectively biodegradable vascular graft
in pigs. Ital J Surg Sci 1989; 19:121-30. coated with polyethylene oxide-polylactic acid for right
50. van der Lei B, Wildevuur CR. From a synthetic, ventricular conduit. An experimental study. J Thorac
microporous, compliant, biodegradable small-caliber vas- Cardiovasc Surg 1990; 100:769-76.
cular graft to a new artery. Thorac Cardiovasc Surg 1989; 70. Cohn D, Elchai Z, Gershon B et al. Introducing a selec-
37:337-47. tively biodegradable filament wound arterial prosthesis: A
Bioresorbable Grafts: A Counterintuitive Approach 503
short-term implantation study. J Biomed Mater Res 1992; 76. Moczar M, Godeau G, Robert AM, Moczar E, Loisance
26:1184-204. D, Bessous JP. Biodegradable arterial prosthesis from rat
71. Hellener G, Cohn D, Marom G. Elastic response of fila- aorta. Pathol Biol (Paris) 1980; 28:517-24.
ment wound arterial prostheses under internal pressure. 77. Moczar M, Bessous JP, Loisance D. Healing of biodegrad-
Biomaterials 1994; 15:1115-21. able vascular prostheses. Incorporation of 3H-Valine into
72. Moritz A, Grabenwoger F, Windisch A et al. A method proteins in the subendothelial scar and host intima-media
for constricting large veins for use in arterial vascular re- of rat aorta. Connective Tissue Research 1983; 12:33-42.
construction. Artif Organs 1990; 14:394-8. 78. Schmitz-Rixen T, Megerman J, Anderson JM et al.
73. Zweep HP, Satoh S, van der Lei B et al. Autologous vein Longterm study of a compliant biological vascular graft.
supported with a biodegradable prosthesis for arterial Eur J Vasc Surg 1991; 5:149-58.
grafting. Ann Thorac Surg 1993; 55:427-33. 79. Walter M, Erasmi H, Schmidt R. A new biological vascu-
74. Zweep HP, Satoh S, van der Lei B, Hinrichs WL, Feijen lar prosthesis. Vasa Suppl 1991; 33:90-1.
J, Wildevuur CR. Degradation of a supporting prosthesis 80. Walter M, Erasmi H. A new vascular prosthesis of bovine
can optimize arterialization of autologous veins. Ann origin. Thorac Cardiovasc Surgeon 1992; 40:38-41.
Thorac Surg 1993; 56:1117-22. 81. Walter M, Asoklis S, Erasmi H, Meisch JP. Bovine heter-
75. Hinrichs WL, Zweep HP, Satoh S, Feijen J, Wildevuur CR. ologous graft in replacement of a small calibre artery. Ann
Supporting, microporous, elastomeric, degradable prosthe- Chir 1994; 48:870-5.
ses to improve the arterialization of autologous vein grafts.
Biomaterials 1994; 15:83-91.
Scaffold Engineering
Material Aspects
CHAPTER 46
Biodegradable Materials
K.J.L. Burg, S.W. Shalaby
Introduction
A bsorbable materials are unique as implants, in that they are absorbed and excreted from
the body at the conclusion of their functional period, thus alleviating the expense and
potential complications of a retrieval surgery. Additionally, as these materials gradually ab-
sorb, they allow a gradual shift in mechanical strength from the biomaterial back to the
healing tissue. The absorption time and therefore loss of mechanical strength of the implant
may be modulated to best match a given application.
Synthetic absorbable materials were first developed as medical implants in the late
1960s and found use in applications such as barriers, scaffolds, and drug delivery systems.1
Prior to this time, naturally derived materials such as collagen, catgut, and silk were used;
however, they were subject to unpredictable, enzymatic degradation and elicited immune
responses due to impurities. The evaluation and application of absorbable materials gradu-
ally expanded from soft tissue devices into bone fracture fixation devices, an area of ongoing
development.2-5 A more recent area of interest is the role of absorbable materials as struc-
tural matrices or supports in tissue engineering.6,7
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
506 Tissue Engineering of Prosthetic Vascular Grafts
tissue culture. This included injectable gels and gel formers, sizes larger than 10 micron will allow invasion of fibrovas-
fibrous scaffolds, porous foams, and channeled substrates cular tissue and therefore enhance capillary network devel-
to allow vascularization. The processing techniques also de- opment and subsequent vascularization.16 The rate of the
veloped to allow more custom-made designs and provision tissue infiltration increases with pore size and porosity, and
of more complicated, site-specific shapes. The processing of its survival and integration with the host tissue depends
an absorbable material allows a reproducible end product, largely on the vascularization.
and the use of an absorbable material reduces both the cost An even distribution of cells throughout the matrix is
and the risk of donor and recipient surgery. important to the development and functionality of the tis-
There are many synthetic polymers which are used in sue, as well as the interaction of the device and transplanted
tissue engineering research, among which are polylactide, cells with the surrounding tissue. The hydrophilicity of the
polyglycolide, polyphosphazene, polycaprolactone, and vari- polymeric surface contributes to the wettability of the po-
ous copolymers. Copolymers such as lactide-colysine have rous structure and hence to the distribution of cells through-
been specifically developed in order to provide attachment out the matrix. The crystalline polylactide and polyglycolide
points for cell binding peptide groups.12-14 The synthetic materials are relatively hydrophobic; therefore, aqueous cell
polymers are advantageous in that their properties may be solutions do not adequately infiltrate the polymeric inte-
directly modulated during synthesis and processing to rior. Less polar solvents such as ethanol may be used to
achieve particular requirements. They tend to degrade in a prewet the scaffold before applying the cell solution;17 the
very predictable fashion, largely hydrolytically, which has concern then becomes the minimization of the scaffold-sol-
obvious benefits. vent contact time in order to avoid excessive ester bond cleav-
The natural materials which have found use in tissue age. Another approach to improve cellular distribution
engineering research include alginate, collagen, chitosan and throughout the scaffold without addressing the bulk sub-
coral, as well as similarly peptide modified natural materi- strate properties is to surface coat the device with an ab-
als (Table 46.1). These materials are naturally derived and sorbable, hydrophilic material prior to cell seeding. Low
therefore readily available; their absorption is generally en- molecular weight, soluble polyvinyl alcohol coatings, as well
zymatically driven and subsequently less predictable. Typi- as pluronic coatings, may be successfully applied to these
cally this group of absorbable materials may vary consider- hydrophobic substrates, thus rendering them more attrac-
ably from batch to batch. They do, however, allow gelati- tive for cellular attachment.18 These surfactants do not im-
nous, injectable structures and therefore a broader range of prove the cellular adhesion to the polymeric structure, but
applications. they do allow a much better cell distribution throughout
There are many factors to consider when designing the matrix. Furthermore, they dissolve readily in aqueous
an absorbable substrate for tissue engineering purposes. The environment and are therefore unlikely to interfere with the
amount of exposed polymeric surface area will relate to the intended implant function.
intensity of the inflammatory response; therefore, an im- The criteria for a tissue engineered absorbable vascu-
plant with greater exposed surface will induce a greater re- lar graft material are very focused. The material must be
sponse than a smaller area.7 The polymer purity, crystallin- readily sculpted into an appropriately sized tubular shape,
ity, molecular orientation, molecular weight, and polydis- matching that of surrounding target tissue and, more prob-
persity may all influence the presence and timing of an in- lematic in the case of small diameter vessels, must be able to
flammatory response. The effect of bulk mass loss of the withstand large flow forces without hindering transport. The
polymeric material on the success rate of the newly devel- construct must be a specific porosity in order to optimize
oped tissue is, as a result, an important design issue. the tissue integration and reformation. The tissue ingrowth
The pore size of a three-dimensional scaffold, as well must occur over the entire length of the prosthesis rather
as the means of dispersion and flow rate of cells into the than primarily from a transanastomotic pannus ingrowth.
scaffold, is of importance in determining both the distribu- Most importantly, it must have an anti-thrombogenic lu-
tion of cells and the integrity of the cells.15 Additionally, pore menal cellular lining and must facilitate long term viability.
Traditionally the ideal vascular graft material was as- to success, as too high a degree causes lack of anastomotic
sumed to be biologically inert. Today, however, research has endothelialization and transinterstitial tissue ingrowth.40
shifted toward creating an active material that will interact The compliance and longevity of a potential vascular
and integrate favorably with the surrounding biological en- graft material can influence the retention of mechanical in-
vironment. The absorbable materials, as dynamic implant tegrity. 41,42 Studies in the polyurethane/polylactide
systems, have this quality and appear to stimulate growth (PU/PLLA) system have shown that a decrease in the per-
factor release through macrophage induction. Both cent polyurethane causes a decrease in compliance and an
polyglycolide (PG), 90/10 glycolide/L-lactide copolymer, and increase in aneurysmal degeneration.43 These studies fur-
polydioxanone (PDS) woven grafts have successfully dem- ther showed that high mechanical compliance of the graft
onstrated, in animal studies, inner capsule development as at implantation stimulates elastin formation and an absorb-
well as capillary invasion, thus resulting in an integrated, able graft material allows maintenance of the mechanical
mechanically stable prosthesis.35-37 The inner capsule con- stability of the newly formed tissue. The PU/PLLA system is
tains layers of myofibroblasts covered by endothelial-like cells initially a compliant one; however, long term incidences of
(presumably from a capillary endothelial source). In similar aneurysmal dilation are significantly greater in this system
studies, Dacron reinforced PGA shows no such capsule de- than the polyester ones. This is possibly because the ongo-
velopment and low amounts of transinterstitial macrophage ing presence of PU leads to fibroplasia, resulting in decreased
migration, presumably due to the inhibitive nature of compliance and elastin deposition. Another important con-
Dacron.38 These types of partially absorbable vascular sideration for scaffold development, as with other applica-
patches, based on woven composite yarn comprising a tions, is that the tissues should gain adequate strength be-
24-82% polyglycolide yarn, the balance being polyethylene fore the material absorbs. If this is not possible, the absorb-
terephthalate yarn, were also evaluated in dogs in terms of able scaffold may be reinforced by an inert (not Dacron)
blood compatibility. 39 Results indicate that the patch component or developed into a composite material whereby
biocompatibility and effect on blood components increased the tissue ingrowth occurs through a series (two or more)
with the increase in the polyglycolide content. This signals of polymers. The multicomponent absorbable vascular pros-
the importance of tissue ingrowth to the construction of theses composed of compound yarns of this type appear to
successful synthetic vascular grafts. be efficacious.44 Studies have shown that, in contrast to the
Studies have also focused on the creation of an opti- absorbable component, the biomechanical properties of the
mal biological component, integral to the prosthesis, for nonabsorbable component modulate the histologic at-
example, tubular nonabsorbable polyester fabrics incorpo- tributes and longevity of the final tissue structure.
rated with bovine serum albumin and a luminal coating of Other attempts at tissue engineering blood vessels have
gelatin heparin sodium solution. These constructs are been made by constructing the vessels ex vivo directly from
crosslinked with a polyepoxy compound. The grafts, once the cellular components. This may be accomplished by
implanted, show no signs of platelet aggregation or fibrin culturing a sheet of human vascular smooth muscle cells in
formation and show development of neointima over the collagen and placing this about a lumenal support to produce
anastomotic graft site. The degree of crosslinking is critical the media.45 A fibroblast sheet is then similarly cultivated
510 Tissue Engineering of Prosthetic Vascular Grafts
and placed about the media to form the adventitia. The need for a vascular graft implies that the sur-
Endothelial cells are later seeded into the lumen of the vessel, rounding target tissue may be “abnormal” (atherosclerotic,
thus forming a mechanically sound three-dimensional vessel. for example). It has been shown in animal studies that the
quality of such tissues (for example, in the presence of ex-
Newly Tailored Materials cessive amounts of lipoprotein) may have a profound effect
for Tissue Engineering on the development of new tissue.49 It would therefore be
Two recently developed technologies are expected to useful to study the absorbable material in such suboptimal
be of value in tissue engineering applications. First, Shalaby46 conditions or in compromised tissue environments.
developed a series of injectable, absorbable liquid copoly-
mers which undergo gel formation upon contacting moist References
biological tissues. These copolymers are based on polyeth- 1. Gilding DK. Biodegradable polymers. In: Williams DF, ed.
ylene glycol segments and copolymeric segments derived Biocompatibility of Clinical Implant Materials. Boca
from glycolide, lactide, trimethylene carbonate, >-capro- Raton, FL: CRC Press, 1981:209-232.
lactone, and/or p-dioxanone. These monomers are used in 2. Bos RRM, Rozema FR, Boering G et al. In vivo and in
vitro degradation of poly(l-lactide) used for fracture fixa-
the production of commercial absorbable sutures. The in-
tion. In: Putter C de, Lange GL de, Groot K de, Lee AJC,
jectable liquid polymers were described as a suitable vehicle eds. Advances in Biomaterials. 8. Amsterdam, The Neth-
for housing living cells for injection into desired biological erlands: Elsevier Science, 1988:245-250.
sites to continue propagation into three-dimensional con- 3. Leenslag JW, Pennings AJ, Bos RRM et al. Resorbable
structs with the gradual absorption of the gel carrier. Sur- materials of poly(L-lactide). VI. Plates and screws for in-
face phosphonylation of preformed devices, including ternal fracture fixation. Biomater 1987; 8(1):70-73.
microporous open-cell foams that were described by Shalaby 4. Vainionpää S, Kilpikari J, Laiho J et al. Strength and
and co-workers,27,47,48 represents a second area of technol- strength retention in vitro, of absorbable, self-reinforced
ogy with promising potential for use in tissue engineering polyglycolide (PGA) rods for fracture fixation. Biomater
(Fig 46.5). This technology entails the formation of co- 1987; 8(1):46-48.
valently bonded surface phosphonate groups which can: 5. Vert M, Christel P, Garreau H et al. Totally bioresorbable
composite systems for internal fixation of bone fractures.
1. Introduce hydrophilicity to hydrophobic substrates;
In: Chiellini E, Giusti P, Migliaresi C, Nicolais L, eds.
2. Bind specific proteins or peptides to direct cell at- Polymers in Medicine II. New York, New York: Plenum
tachment; and Press, 1986:263-275.
3. Bind Ca2+ for directing the deposition of hydroxya- 6. Vacanti JP. Beyond transplantation. Arch Surg 1988;
patite toward bone formation. 123:545-549.
Among the substrates suitable for surface 7. Vacanti JP, Morse MA, Saltzman WM et al. Selective cell
phosphonylation are certain hydrophobic absorbable transplantation using bioabsorbable artificial polymers as
polyesters. matrices. J Pedia Surg 1988; 23(1):3-9.
8. Mikos AG, Sarakinos G, Lyman MD et al.
Future Prospectives Prevascularization of porous biodegradable polymers.
One of the drawbacks to the synthetic materials as Biotechnol Bioeng 1993; 42:716-723.
9. Boyce ST, Christianson DJ, Hansbrough JF. Structure of
potential vascular graft materials is that, as homopolymers,
a collagen-GAG dermal skin substitute optimized for cul-
they do not have the cell surface receptors required for cel- tured human epidermal keratinocytes. J Biomed Mater Res
lular recognition. The polymers must be optimized to best 1988; 22:939-957.
modulate features such as crystallinity, surface texturing, and 10. Boyce ST, Hansbrough JF. Biologic attachment, growth,
surface wettability in order to selectively attract specific bio- and differentation of cultured human epidermal
logical components while preventing spreading of blood keratinocytes on a graftable collagen and chondroitin-6-
platelets. Surface modification such as that described by sulphate substrate. Surgery 1988; 103(4):421-431.
Shalaby and co-workers may be of value in overcoming some 11. Hansbrough JF, Boyce ST, Cooper ML et al. Burn wound
of these issues. closure with cultured autologous keratinocytes and fibro-
blasts attached to a collagen-glycosaminoglycan substrate.
JAMA 1989; 262(15):2125-2130.
Biodegradable Materials 511
12. Cook AD, Hrkach JS, Gao NN et al. Characterization and 31. Galletti PM, Ip TK, Chiu T-H et al. Extending the func-
development of RGD-peptide-modified poly(lactic acid- tional life of bioresorbable yarns for vascular grafts. Trans
co-lysine) as an interactive, resorbable biomaterial. J ASAIO 1984; 30:399-400.
Biomed Mater Res 1997; 35:513-523. 32. Galletti PM, Trudell LA, Chiu T-H et al. Coated
13. Barrera DA, Zylstra E, Lansbury PT et al. Copolymeriza- bioresorbable mesh as vascular graft material. Trans
tion and degradation of poly(lactic acid-co-lysine). Mac- ASAIO 1985; 31:257-263.
romolecules 1995; 28:425-432. 33. Galletti PM, Aebischer P, Sasken HF et al. Experience with
14. Hrkach JS, Ou J, Lotan N et al. Synthesis of poly(L-lactic fully bioresorbable aortic grafts in the dog. Surgery 1988;
acid-co-L-lysine) graft copolymers. Macromolecules 1995; 103:231-241.
28:4736-4739. 34. Lei Bvd, Blaau EH, Dijk F et al. Microporous compliant
15. Wald HL, Sarakinos G, Lyman MD et al. Cell seeding in biodegradable graft materials: A new concept for mi-
porous transplantation devices. Biomater 1993; 14(4):270- crovascular surgery. In: Skotnicki SH, Buskens FGM, and
278. Reinaerts HHM, eds. Recent Advances in Vascular Graft-
16. Mooney DJ, Langer R. Engineering biomaterials for tis- ing. The Netherlands: Nijmegan, 1984:19-23.
sue engineering: The 10-100 micron size scale. In 35. Greisler HP. Arterial regeneration over absorbable pros-
Bronzino JD, ed. Biomedical Engineering Handbook. Boca theses. Arch Surg 1982; 177:1425-1431.
Raton, FL: CRC Press, 1995:1609-1618. 36. Greisler HP, Kim DU, Price JB et al. Arterial regenerative
17. Mikos AG, Lyman MD, Freed LE et al. Wetting of poly(L- activity after prosthetic implantation. Arch Surg 1985;
lactic acid) and poly(DL-lactic-co-glycolic acid) foams for 120:315-323.
tissue culture. Biomater 1994; 15(1):55-57. 37. Greisler HP, Ellinger J, Schwarcz TH et al. Arterial re-
18. Mooney DJ, Park S, Kaufmann PM et al. Biodegradable generation over polydioxanone prostheses in the rabbit.
sponges for hepatocyte transplantation. J Biomed Mater Arch Surg 1987; 122:715-721.
Res 1995; 29:959-965. 38. Greisler HP, Schwarcz TH, Ellinger J et al. Dacron inhi-
19. Schugens Ch, Grandfils Ch, Jerome R et al. Preparation bition of arterial regenerative activity. J Vasc Surg 1986;
of a macroporous biodegradable polylactide implant for 3:747-756.
neuronal transplantation. J Biomed Mater Res 1995; 39. Yu TJ, Ho DM, and Chu CC. Bicomponent vascular grafts
29:1349-1362. consisting of synthetic absorbable fibers: Part II: in vivo
20. Mikos AG, Bao Y, Cima LG et al. Preparation of healing response. J. Investig. Surg 1994; 7(3):195-211.
poly(glycolic acid) bonded fiber structures for cell attach- 40. Niu S, Kurumatani H, Satoh S et al. Small vascular pros-
ment and transplantation. J Biomed Mater Res 1993; theses with incorporated bioabsorbable matrices. A pre-
27:183-189. liminary study. ASAIO J 1993; 39(3):M750-M753.
21. Frisch KC, Saunders JH, eds. Plastic Foams. New York: 41. Greisler HP, Joyce KA, Kim DU et al. Spatial and tempo-
Marcel Dekker, 1972. ral changes in compliance following implantation of
22. Mooney DJ, Baldwin DF, Suh NP et al. Novel approach bioresorbable vascular grafts. J Biomed Mater Res 1992;
to fabricate porous sponges of poly(DL-lactic-co-glycolic 26(11):1449-61.
acid) without the use of organic solvents. Biomater 1996; 42. Greisler HP, Pham SM, Endean ED et al. Relationship
17(14):1417-1422. between changes in biomechanical properties and cellu-
23. Williams JM, Wrobleski DA. Microstructures and prop- lar ingrowth in absorbable vascular prostheses. ASAIO
erties of some microcellular foams. J Mater Sci Letters Abstracts 1987; 16:25.
1989; 24:4062-4067. 43. Lei Bvd, Bartels HL, Nieuwenhuis P et al. Microporous,
24. Mikos AG, Thorsen AJ, Czerwonka LA et al. Preparation compliant, biodegradable vascular grafts for the regenera-
and characterization of poly(L-lactic acid) foams. Poly- tion of the arterial wall in rat abdominal aorta. Surgery
mer 1994; 35:1068-1077. 1985; 98:955-963.
25. De Ponti R, Torricelli C, Martini A et al. Use of 44. Endean ED, Kim DU, Ellinger J et al. Effects of
supercritical fluids to obtain porous sponges of biodegrad- polypropylene’s mechanical properties on histological and
able polymers. International Patent WO 91/09079, June functional reactions to polyglactin 910/polypropylene vas-
1991. cular prostheses. Surg Forum 1987; 38:323-325.
26. Roweton SL. A new approach to the formation of tailored 45. Germain L, L’Heureux N, Labbé R et al. Human blood
microcellular foams and microtextrued surfaces of absorb- vessel produced in vitro by tissue engineering. Workshop
able and non-absorbable thermoplastic biomaterials. Mas- on Biomaterials and Tissue Engineering Abstracts 1997;
ter of Science Thesis. Department of Bioengineering, 23.
Clemson University. 1994. 46. Shalaby SW. Hydrogel-forming self-solvating absorbable
27. Shalaby SW, Roweton SL. Continuous open-cell polymeric polyester copolymers and methods for use thereof. U.S.
foams containing living cells. 1997: U.S. Patent #5,677,355. Patent 5,612,052 Assignee: Poly-Med, Inc. 1997.
28. Herring MB, Gardner AK, Glover JL. Seeding human ar- 47. Shalaby SW, McCaig SM. Process for phosphonylating the
terial prostheses with mechanically derived endothelium. surface of an organic polymeric preform, U.S. Patent
The detrimental effect of smoking. J Vasc Surg 1984; 5,491,198, Assignee: Clemson University 1996.
1:279-289. 48. Shalaby SW, Rogers KR. Polymeric prosthesis having a
29. Yue X, van der Lei B, Schakenraad JM et al. Smooth phosphonylated surface, U.S. Patent 5,558,517, Assignee:
muscle cell seeding in biodegradable grafts in rats: A new Clemson University 1996.
method to enhance the process of arterial wall regenera- 49. Greisler HP, Klosak JJ, Endean ED et al. Effects of hyper-
tion. Surgery 1988; 103:206-212. cholesterolemia on healing of vascular grafts. J Investiga-
30. Kempczinski RF, Rosenman JE, Pearce WH et al. Endo- tive Surg 1991; 4(3):299-312.
thelial cell seeding of a new PTFE vascular prosthesis. J
Vasc Surg 1985; 2(3):424-429.
Scaffold Engineering
Material Aspects
CHAPTER 47
The Influence of Porosity and
Surface Roughness on Biocompatibility
J.M. Schakenraad, K.H. Lam
Introduction
V ascular prostheses might solve a lot of clinical problems, if only they would “do their job”
in a functional way. Functional in this respect includes: nonthrombogenicity; compli-
ance, diameter and wall thickness similar to vessel wall; blood compatibility in all its aspects
(complement activation, red and white blood cell damage, thrombocyte activation etc.);
immunological inertness etc. Some of these parameters can be influenced in such a way that
an acceptable functionality is achieved. Especially with regard to small diameter vascular
grafts (< 3 mm), it has proven impossible to solve all problems simultaneously. A vast chal-
lenge still lies ahead for the next generation of scientists.
This chapter will discuss two material surface parameters (roughness and porosity)
having a large influence on biocompatibility and ultimate patency of vascular grafts. A pa-
rameter indicative for short term biocompatibility is the inflammatory response towards an
implanted biomaterial. Upon implantation of a biomaterial, local1,2 and systemic3 effects
can be observed. The local tissue reaction consists of an inflammatory response which serves
to eliminate the cause of an injury, minimize the damage and trigger mechanisms for repair-
ing the tissue damaged by injury. The surgical procedure itself initially determines the type
and intensity of the inflammatory response. However, in addition, implanted biomaterials
themselves provoke an inflammatory response.
Many material characteristics may influence the inflammatory response against a bio-
material (being either a natural polymer, a chemical polymer, metals, ceramics etc.): chemi-
cal composition,1 material toxicity4,5 surface free energy (wettability),6 surface charge7 sur-
face morphology such as porosity,8 roughness of the surface9 and shape,7 implantation site,10
rate of degradation,11 degradation products,12,13 and many more.
Surface morphology is one of the first factors directly influencing the tissue/inflam-
matory response. This is indicated in various studies.7,14 Matlaga et al demonstrated the role
of shape.7 The intensity of the inflammatory response increases when the number of edges
of the implanted materials increases. Also, there are studies indicating that the inflamma-
tory response against implanted porous polymers or biomaterials is more intense when com-
pared to nonporous materials.8
Wettability may influence the tissue reaction to the polymer, because there is a range
in wettability values that is optimal for cell adhesion, growth and spreading. Cells attach and
proliferate less well on polymers having a wettability which is too low15 or too high.16
Another factor is degradability. Degrading polymers provoke a more intense inflam-
matory response compared to nondegrading polymers. A possible cause for this observation
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
514 Tissue Engineering of Prosthetic Vascular Grafts
Evaluation Models
Materials
As a degradable material, the well known and widely
used polylactic acid is used, both as a solid, a porous and a
combination form. As a nondegradable reference material,
PTFE is used, both in a solid shape and as the expanded
PTFE with a pore size of 30 micrometers.
Poly(L-lactic acid) (PLLA) films were cast from PLLA
with a reported Mn of 50,000 (Purac Biochem B.V., The
Netherlands).17 Three types of films were cast: a nonporous
type, a porous type and a “combi” type (porous with a non-
porous layer on one side). The base parameters of the PLLA
films are listed in Table 47.1. All PLLA films were cut in strips
of 15 x 2 mm.
Polytetrafluoroethylene (PTFE) was obtained com-
mercially (Wientjes, The Netherlands). PTFE was cut into
strips of 15 x 2 x 1 mm. Expanded polytetrafluoroenthylene
(ePTFE) was obtained as nonsterile GORE-TEX® ePTFE cell
collector tubing (WL Gore & Associates GMBH, Germany).
The tubing was cut open along the longitudinal axis to ob-
tain films measuring 15 x 2 x 0.25 mm.
Parameter Non-porous Porous Combi Fig. 47.1. Scanning electron micrographs of a cross-section of
PLLA film PLLA film PLLA film PLLA films. (A, top.) nonporous, (B, middle.) porous,
(C, bottom.) “combi”; arrowhead indicates the nonporous side.
Mw 98,000 109,000 167,000 The thickness of the films is: nonporous, 33 ∝m; porous, 244 ∝m
Mn 42,000 41,000 53,000 and “combi”, 82 ∝m. The nonporous layer of the “combi” film
Mw/Mn 2.3 2.7 3.1
Tm 176°C 180°C 181°C
was approximately 5 ∝m. The pore size of the porous film var-
Heat of fusion 53 J/g 51 J/g 50 J/g ied from approximately 1-150 ∝m and of the “combi” film from
1-50 ∝m.
The Influence of Porosity and Surface Roughness on Biocompatibility 515
Surface Morphology porous PLLA film, a porous PLLA film, a “combi” PLLA film,
The surface morphology of the different specimens is a PTFE film and an ePTFE film were implanted. The sixth
illustrated in Figure 47.1A-C. Specimens of all films were incision and subcutaneous pocket served as a control (sham
sputter-coated with gold (Balzers 07 120B) and their sur- operation). Three samples per polymer and time interval
face morphology was examined with DS 130 scanning elec- were implanted. The rats had free access to standard rat food
tron microscope (SEM) (ISI), operated at 10 KV. and water. All national rules concerning the care and use of
The thickness of the polymers was calculated using laboratory animals have been observed.
SEM: nonporous 33 ∝ m (Fig.47.1A); porous 244 ∝ m The rats were sacrificed after 1, 3, 7, 14, 40, 90 or 180
(Fig. 47.1B); and the “combi” film 82 ∝m (Fig. 47.1C). The days and the polymer films were removed with excess sur-
nonporous layer of the “combi” film was approximately 5 ∝m. rounding tissue.
The pore size of the porous film varied from approximately
1 to 150 ∝m and of the “combi” film from 1 to 50 ∝m. PTFE Evaluation of the Inflammatory Response
was nonporous, having the same morphological appearance
of the surface as the nonporous PLLA film. The thickness of Light Microscopy
the porous ePTFE film was approximately 250 ∝m with a After harvesting, the samples were immediately fixed
fibril length of approximately 90 ∝m (Fig. 47.2). for at least 24 h at 4°C in a 0.1 M Na-cacodylate buffer,
pH 7.4, containing 2% glutaraldehyde and 0.1 M sucrose.
Wettability The samples were then dehydrated in a graded ethanol se-
Wettability, as a measure for surface free energy (hy- ries and embedded in glycolmethacrylate (Technovit®,
drophobicity) was determined by contact angle measure- Kulzer, Germany), allowing sections to be cut perpendicu-
ments using the sessile drop technique described by Busscher lar to the longitudinal axis of the polymer film. Sections for
et al.18 For PTFE and the nonporous PLLA, the contact angles light microscopy examination were cut on a microtome
were determined using H20 and #%bromonaphthalene as (Jung autocut 1140, equipped with a D knife with a tung-
wetting agents. Five measurements were made for each poly- sten carbide cutting edge), mounted on glass slides and
mer and wetting agent. stained with toluidine blue and alkaline fuchsin.19
The highest value of each set of five measurements is
maximally 5° higher than the lowest value. Therefore, it was Immunohistochemical Staining
not relevant to determine the standard deviation. The H2O After harvesting, samples were snap-frozen at -80°C
contact angles were 101° for PTFE and 72° for nonporous using liquid freon. Cryostat sections of 7 ∝m were cut,
PLLA, and the #-bromonaphthalene contact angles were 66° mounted on glass slides, air-dried and fixed in acetone for
for PTFE and 23° for nonporous PLLA. This shows that 12 minutes. The sections were then again air-dried for 1 h
nonporous PLLA has a less hydrophobic surface than PTFE. and incubated with the first-stage, cell type specific, mono-
clonal antibody (mAb) for 1 h. Subsequently, the sections
Implantation Procedure were washed 3 times in PBS, followed by incubation with
The films were disinfected prior to implantation and the second-stage antibody conjugated to peroxidase, diluted
implanted subcutaneously in 21 female (AO x BN)F1 rats 1:40 in PBS and supplemented with 5% v/v normal rat se-
obtained from our own breeding facility. In each rat a non- rum to prevent nonspecific binding. Swine anti-rabbit Ig
(Dakopatts, Denmark) was used as second-stage antibody Macroscopically, there were no signs of an infection
to detect the first-stage mAb #-Asialo GM1 (Table 47.2). demonstrated in any of the rats. The results of the semi-
Rabbit anti-mouse Ig (Dakopatts, Denmark) was used to quantitative evaluation of the immunohistochemical stain-
detect the other first-stage mAbs. After incubation with the ing to detect leukocytes (OX 1), neutrophilic granulocytes
second-stage antibody, sections were rinsed 3 times in PBS (HIS 48), macrophages (ED 1, ED 2, ED 3), T lymphocytes
for 5 minutes. Peroxidase activity was demonstrated by ap- (OX 19), vast majority of B lymphocytes (HIS 40), natural
plying 3,3-diaminobenzidine tetrahydrocloride (Sigma) at killer cells (#-Asialo GM1), cells expressing MHC class II
a concentration of 0.5 mg/ml in 0.05 M Tris-HCI buffer (pH antigen—such as activated fibroblasts—(HIS 19) are dem-
7.6) containing 0.01% H2O2 for 10 minutes. After rinsing in onstrated in Figures 47.3A-I respectively. False positive cells
fresh tap water, sections were counterstained lightly with were observed occasionally in PBS controls. This was due to
hematoxylin for 10 seconds. The sections were then dehy- endogenous peroxidase activity expressed only by neutro-
drated using a graded ethanol series and xylene and subse- philic granulocytes, since the staining pattern using HIS 48
quently covered with coverslips using DePeX (Gurr, BDH corresponds with the pattern of endogenous peroxidase ac-
Ltd, England) mounting medium. In controls, PBS was used tivity. Moreover, cells expressing endogenous peroxidase
instead of the first-stage mAb. The first-stage mAbs used, activity can be well distinguished due to their more intense
and their sources, are shown in Table 47.2. staining as compared to cells stained for mAbs.
The concentration of B lymphocytes (Fig. 47.3D) and
Quantification of the Inflammatory Response the concentration of natural killer cells (Fig. 47.3H) was to
The magnification of the light microscope was set at a large extent similar and low for both PLLA, PTFE films
x400 when examining the section stained using mAbs. The and the subcutaneous tissue under the scar of the sham op-
staining patterns of the tissue surrounding or invading the eration.
polymer film was evaluated and the number of positive cells At day3, the concentration of each cell type involved
surrounding or invading the polymer films per field of view in the inflammatory response as a measure for the intensity
counted. Four fields of view per section, two sections per was approximately the same for all the polymer films
sample and three samples for each period of implantation, (Fig. 47.3A to 47.3G). At day 7, macrophages are beginning
polymer film and monoclonal antibody respectively were to play a prominent role in the inflammatory response
examined. The tissue reactions at the edges of the polymer (Fig. 47.3E). Subsets of macrophages surrounding the poly-
films were excluded from evaluation, to avoid artifacts due mer films were found in different locations. ED 1 positive
to mechanical irritation. macrophages are found in the entire area, demonstrating
The number of cells staining positive per field of view inflammatory response against the implant, especially in the
was classified as follows: area in closest approximation to the polymer film
grade 0 = no positive cells, (Fig. 47.4A). In contrast, ED 2 and ED 3 positive macro-
1 = 1 to 5 positive cells per field of view, phages are neither found in the area in closest approxima-
2 = 5 to 10 positive cells per field of view, tion the polymer film, nor in the pores of the PLLA or PTFE
3 = 10 to 25 positive cells per field of view and film. ED 2 and ED 3 positive macrophages remain restricted
4 = more than 25 positive cells per field of view. to the surrounding tissue at some distance from the implant
The average class of the 24 fields of view for each pe- (Fig. 47.4B and 47.4C). The relative ratio between the sub-
riod of implantation, polymer film and monoclonal anti- sets was fairly constant for the respective implants.
body respectively is reported.
Table 47.2. Antibodies (mAbs) used to examine and quantify the inflammatory response against the implanted
polymer films
OX 1 CD 45 All leukocytes
HIS 48 probably surface Granulocytes
OX 19 CD 5 T-lymphocytes
HIS 40 IgM heavy chain B-lymphocyte subset likely to react first upon inflammatory response
ED 1 Lysosomal antigen Majority of macrophages. Probably associated with active phagocytosis.
ED 2 Surface antigen Subset macrophages. Probably associated with maturity.
ED 3 Surface antigen Subset macrophages. Probably associated with downregulation of
inflammatory reaction
#-Asialo GM1 Probably surface antigen Large granular lymphocytes natural killer cells
HIS 19 MHC class II Activated tissue cells (fibroblast) IDC, subset macrophage
Source of the mAbs: OX antibodies were a generous gift of the late Dr. A.F. Williams, Department of Biochemistry, University of Oxford,
Great Britain; ED antibodies were a generous gift of Dr. C.D. Dijkstra, Department of Cell Biology. The HIS and a-Asialo antibodies were
generous gifts of Dr. P. Nieuwenhiu’s of the Department of Histology, University of Groningen.
The Influence of Porosity and Surface Roughness on Biocompatibility 517
In GMA sections it is observed that the inflammatory flammatory response than PTFE films as demonstrated by
response at day 7 is becoming more intense (Fig. 47.5) for the higher concentration of neutrophils, macrophages/gi-
the porous PLLA and porous side of the “combi” PLLA films. ant cells and T lymphocytes surrounding the PLLA films.
For the nonporous PLLA and the nonporous side of the The concentration of cells against PTFE and ePTFE films is
“combi” PLLA film, the onset of encapsulation by approxi- comparable to the concentration of cells in the subcutane-
mately 3 layers of fibroblasts was observed. Only a minimal ous tissue under the scar of the sham operation. The in-
encapsulation is observed for the porous PLLA film at day flammatory response against the nonporous PLLA film was
7. In contrast to the porous PLLA film almost no cellular mainly localized at the edges of the film or at the edges of
invasion of the ePTFE film was observed. The cell layer sur- the pieces when broken. In contrast, the inflammatory re-
rounding the ePTFE consists mainly of macrophages. The sponse against porous PLLA films was localized in the pores.
PTFE film was surrounded by one or two layers of mac- The “combi” film shows a more pronounced inflammatory
rophages and the onset of encapsulation can be observed. response at the porous side.
At day 14, the inflammatory response becomes chronic At day 90, the difference in the intensity of the inflam-
with predominantly macrophages and T lymphocytes sur- matory response between PLLA and PTFE films and also
rounding the films. In the GMA sections, foreign body gi- between nonporous and porous PLLA films had become
ant cells can be observed surrounding the PLLA fims much more pronounced. This is demonstrated by the in-
(Fig. 47.6). All films are now encapsulated by continuous creased concentration of macrophages, leukocytes and cells
layers of fibrocytes and collagen. The fibrocytes are not expressing MHC class II (HIS 19) antigen, (probably acti-
stained by HIS 19 monoclonal antibody, indicating a de- vated fibroblasts) surrounding or invading the PLLA films.
crease in cell activity. The porous PLLA film and porous side The porous PLLA films provoke a more intense inflamma-
of the “combi” film provoke a more intense inflammatory tory response than the nonporous PLLA film. In contrast,
response than the nonporous PLLA film and nonporous side the inflammatory response against PTFE and ePTFE films
of the “combi” film respectively. However, this observation remained the same as at day 40.
could not be made for the ePTFE film as compared to the At day180, the difference in the intensity of the in-
PTFE film. flammatory response between the porous and the nonpo-
At day 40, in general, the intensity of the inflamma- rous PLLA film was more pronounced. In contrast, the tis-
tory response against the polymer films had decreased fur- sue reaction (“inflammatory response”) against PTFE and
ther. However, PLLA films still provoked a more intense in- ePTFE did not differ much from the tissue reaction in the
522 Tissue Engineering of Prosthetic Vascular Grafts
subcutaneous tissue under the scar of the sham operation. The relative polymer surface area in sections (RPSA)
There are almost no cells localized in the pores of ePTFE was determined by morphometrical analysis using light
(Fig. 47.7A). In contrast, the inflammatory response against microscopical sections and a Quantimet 520 (Cambridge
porous PLLA was localized mainly in the large pores Instruments) image analyzer. The RPSA was defined as the
(Fig. 47.7B). ratio between the polymer surface area and measurement
frame area. The boundaries of the measurement frame were
Relative Polymer Surface Area in Sections set at the outer boundaries of the polymer surface area. The
The relative polymer surface area (RPSA) in sections morphometrical analysis was performed on porous and
was determined after in vivo and in vitro procedures. After “combi” films only, because the nonporous film was not
harvesting, samples were immediately fixed by immersion eroded nor did it become porous, not even after an immer-
in a 0.1 M Na-cacodylate buffer, pH 7.4, containing 2% glu- sion or implantation period of 180 days. The RPSA of the
taraldehyde and 0.1 M sucrose, for at least 24 h at 4°C. After nonporous film remained 100%. For each implantation pe-
rinsing with buffer, the samples were dehydrated in graded riod or immersion period ten measurements of the remain-
ethanol series. The samples were then embedded in a posi- ing polymer surface area (of at least two sections) were per-
tion which allowed sections to be cut perpendicular to the formed. The mean value and standard deviation were cal-
longitudinal axis of the polymer films. After embedding in culated from these ten values. All data were normalized to
glycolmethacrylate (GMA) (Technovit, Kulzer, Germany), the initial value (t = 0).
sections of 3 ∝m were cut on a Jung 1140 autocut micro- The results of the morphometrical analyses are pre-
tome, using a D knife with a tungsten carbide cutting edge. sented in Figures 47.8A (in vitro) and 47.8B (in vivo). The
The sections were mounted on glass and stained with tolui- RPSA in vivo shows no significant decrease for the nonpo-
dine blue and alkaline fuchsin. rous, the porous or the “combi” film. Also, no decrease was
524 Tissue Engineering of Prosthetic Vascular Grafts
observed in vitro for the nonporous and porous film up till sample of 5-10 mg was dissolved in 10 ml chloroform and
day 180 and for the “combi” film, up till day 90. It was not filtered (Spartan 13/20 filter, 0.45 ∝l). The injection volume
possible to obtain the surface area of the “combi” film at day per measurement was 200 ∝l. Chloroform was used as elu-
180, due to fragmentation. ent at a flow rate of 2.0 ml/min. The Mw, Mn and polydis-
During the in vivo experiment with the nonporous persity ratio were calculated using the calibration data of
polymer film, no erosion or pore formation was observed. polystyrene standards of narrow molecular weight distri-
Moreover, after immersion in PBS (in vitro), it was not pos- bution dissolved in tetrahydrofuran (THF).
sible to carry the nonporous film through the embedding The Mw (Fig. 47.9A) and Mn (Fig. 47.9B) decreased
procedures for light microscopy from day 40 on, due to in- for all three films at approximately the same rate. From day
creased brittleness. The surface area of the remaining non- 7 on, the porous film generally retained the highest Mw and
porous film remained 100% over the entire test period. Mn. The initial (t = 0) Mw of the “combi” film was the high-
est (167,000) and of the nonporous film the lowest (98,000;
Mw and Mn porous was 109,000). Before day 7, the curves of the porous
Mw (molecular weight) and MN in vitro were deter- and “combi” film showed large fluctuations for Mw. There-
mined by gel permeation chromatography (GPC) at 20°C after, all values for Mw demonstrated a decreasing trend till
on a Waters Associates GPC apparatus using Waters Associ- the 170th day (Mw nonporous: 24,000, porous: 60,000,
ates columns (bead size of 105, 104, 103 Å). A precolumn “combi”:29,000). The Mn curves roughly approximate the
with a pore size of 500 Å was used. A Waters Associates R same trend as the Mw curves. The fluctuations of the values
403 differential refractometer was used as a detector. A for the “combi” film are larger.
The Influence of Porosity and Surface Roughness on Biocompatibility 525
Fig. 47.7. (A, left.) Porous e-PTFE (P) and (B, right.) porous PLLA (P) after 180 days of implantation. There are almost no inflam-
matory cells localized in the pores of the e-PTFE film. In contrast, the inflammatory response is localized mainly in the pores
F=fibrous encapsulation. Bar indicates 63∝m.
Interpretation of Results blood. SAA may then be converted into amyloid A, which is
deposited in tissues. However, the exact mechanism is yet to
Inflammatory Response and Biocompatibility be fully uncovered, and the relation between the chronic
The results demonstrate that there are two phases in inflammatory response against biomaterials and amyloido-
the inflammatory response against the films. Phase 1 is ob- sis also remains to be investigated.
served upon implantation of the film. It is mainly caused by
the injury sustained by the implantation procedure. This The Role of Macrophages and Giant Cells
uncomplicated inflammatory response, part of the wound ED 1 stains an intracellular antigen, probably associ-
healing reaction, ends after 7-10 days and has been well de- ated with the lysosomal membrane. The antigen probably
scribed.2,20 In this phase, the contribution of the implanted remains present even in the event of fusion of macrophages
polymer film to the intensity of the inflammatory response and formation of foreign body giant cells, since ED 1 posi-
is in most cases minimal, except when leakage of large quan- tive cells are observed on the same location of foreign body
tities of toxic products occurs.5 After one week, any remain- giant cells in conventional light microscopical sections. ED 2
ing inflammatory response can be considered as a tissue re- is a marker which is found on mature tissue macrophages.
action against the implanted biomaterial.11 This chronic It takes about one week for monocytes/macrophages to ex-
inflammatory response (phase 2) is often described as a for- press ED 2 on their cell surface after leaving the vascular
eign body reaction.1,2,5 It mainly consists of macrophages system. The role of ED 2 and ED 3 in the inflammatory re-
and giant cells surrounding the implant. sponse is not well understood. The different staining pat-
A minimal inflammatory response is preferred when terns of the ED antibodies indicate the possibility of either
biomaterials are implanted for a long time span, because a different populations of macrophages which play a role in
persistent (chronic) inflammatory response may predispose the tissue reaction against biomaterials, or macrophages ex-
for amyloidosis,21,22 or carcinogenesis.23 A persistent inflam- pressing different surface receptors in the course of the in-
matory response increases the concentration of both serum flammatory response against a biomaterial.
amyloid A (SAA) and amyloid enhancing factor (AEF) in
526 Tissue Engineering of Prosthetic Vascular Grafts
The exact mechanism of foreign body giant cell for- the nonporous PLLA in an aqueous environment. This in-
mations is still to be elucidated. Certain cell types, e.g., T dicates that porosity is an important factor determining the
lymphocytes, may play a pivotal role in this process. T helper intensity of the inflammatory response against implanted
lymphocytes capable of secreting interferon-&, especially, may PLLA films. The small difference in the intensity of the in-
play a role in the fusion of macrophages, forming giant flammatory response between PTFE and ePTFE as compared
cells.24,25 Activation of macrophages may be induced by vari- to porous and nonporous PLLA respectively indicates that
ous pathways. In one pathway, (limited) damage to neutro- porosity as a single factor is not enough to enhance the in-
phils and macrophages may lead to secretion of cytokines flammatory response. The relatively high wettability of PLLA
(IL-1), activating T helper lymphocytes. Another possibility compared to ePTFE allows for better ingrowth of tissue into
is change of shape when a macrophage comes in contact the pores of PLLA and probably more exposure to other fac-
with a boimaterial,26,27 especially when a single macrophage tors, determining the intensity of the inflammatory response.
is not able to phagocytose the polymer(fragment). According to many authors, the surface properties of
an implant have a large influence on the inflammatory re-
Porosity, Wettability, Degradability and Inflammatory sponse. Thus, the difference in wettability as one of the sur-
Response face properties may also be a factor, although Baier et al could
Porous PLLA provokes a more intense inflammatory not demonstrate a difference in the inflammatory response
response from day 7 on, despite a higher degradation rate of against smooth metal pieces having a different wettability.28
The Influence of Porosity and Surface Roughness on Biocompatibility 527
However, other authors demonstrated that different poly- Also, the size of the pores of the porous PLLA films has in-
mers induce a different level of IL-1 production, correlating creased, possibly with the same effect as fragmentation. In
with the intensity of the inflammatory response,29,30,31 al- the case of PLLA films, the increase in surface area during
though the relation to wettability was not investigated. How- the degradation process as a single factor may be sufficient
ever, the production of IL-1 seems to be less when using for increasing the intensity of the inflammatory response.
relatively hydrophobic materials such as silicon. Further in- It can be concluded that biodegradable PLLA films
vestigation is needed to establish the precise relation between provoke a more intense inflammatory response than non-
wettability and inflammatory response. degradable PTFE films. Also, porosity enhances the inflam-
The differences in inflammatory response against the matory response. However, porosity enhances the inflam-
PLLA (degradable) and PTFE (nondegradable) films became matory response only when the wettability of a biomaterial
apparent from day 40. One reason may be the difference in permits cellular ingrowth.
the rate of degradation and subsequently the difference in
the release of degradation products such as monomers, oli- Relative Polymer Surface Area
gomers and finally fragments. As stated previously, only a The RPSA, which was only determined for the porous
few (mostly nonporous) PLLA films were observed to be and the nonporous polymer film, did not decrease signifi-
broken into two or three pieces. However, there is probably cantly, neither in vitro nor in vivo. However, these results
also increase in surface area which could hardly be observed. must be interpreted with some caution, because RPSA
528 Tissue Engineering of Prosthetic Vascular Grafts
measurements cannot detect release of small amounts of pected to contribute very much to the degradation of poly-
degradation products from the core of the polymer film. The esters in vivo. The role of enzymes in the degradation pro-
loss of weight of the nonporous film could not be detected cess in vivo is not clear. Most enzymes in eukaryotic cells are
with RPSA measurements since there was no detectable loss substrate specific. Therefore, a specific three dimensional
of surface area in cross sections of the PLLA film. There was structure of the substrate (polymer) is required to reach the
also no indication of measurable surface erosion, such as active center of the enzyme. This is not likely at 37°C under
increasing surface roughness and/or decreasing thickness of physiologic enzyme concentrations. The probability is even
the nonporous film. However, this method might be useful smaller for the crystalline parts of the polymer (film). How-
in quantifying the later stages of the degradation and the ever, enhancement of the degradation rate in vitro by en-
resorption process in vivo, when weight measurement has zymes was observed for some polymer/enzyme systems. The
become practically impossible. The results indicate that in role of enzymes may be larger when smaller molecules have
vitro the core of the PLLA films remain largely intact till day been formed by other degradation mechanisms (hydroly-
180, and that in vivo there was no resorption of large sis). This was not investigated in the experiments described
amounts of polymer. These findings support the results of in this paper.
the weight measurements. Degradation of polyesters, such as PLLA, primarily
takes place in the amorphous part of the polymer film.34,35
Molecular Weight This may explain the increasing crystallinity, also observed
The results of the molecular weight measurements by other authors.34 However, in order to explain the differ-
indicate a higher degradation rate for the nonporous film ence in degradation rate between the nonporous and po-
compared to the “combi” and porous film, the latter having rous PLLA film, one must assume other processes taking
the lowest degradation rate. The decrease of MW and MN place during degradation. Vert et al, using different polyes-
is obvious for all three types of film, although results ob- ter “plates” of 2 x 17 x 20 mm, observed the formation of a
tained with GPC must be interpreted with caution when polymer layer around the core of the polymer film, largely
low molecular weight polymers resulting from degradation preventing degradation products (e.g., oligomers) released
processes are examined. in the core, to diffuse freely to the aqueous environment.
Therefore, the differences between the films may not The chemical reactive endgroups of the accumulated oligo-
be significant, but the higher MW and MN of the porous mers in the core then enhance the degradation (autocata-
film compared to the nonporous film, from day 7 till the lytic) process. The nonporous polymer film is more suscep-
end of the experiment at day 180, is very suggestive. The tible to this form of degradation, probably because a larger
initial MW and MN of the different films are not equal inner compartment can be formed compared to the porous
(Figs. 47.9A and 47.9B). A possible explanation may be the and “combi” film. The surface/volume ratio of the latter two
washing out of a larger part of the low molecular weight are probably larger than the cut off value of the polymer
fraction into the media, at the extra rinsing step to remove surface/volume ratio regarding this effect.
the sodium citrate, of the films with a porous component; However, other mechanisms are also possible. There
therefore higher MW and MN were measured. However, it is less flow of the aqueous media in pores of the porous poly-
can be expected that washing out of low molecular weight mer film. Therefore, the concentration of oligomers might
fractions probably also occurs during immersion of the non- be higher in the pores, enhancing degradation. Neverthe-
porous film in the buffer. As a consequence, it can be ex- less, this mechanism apparently has less effect compared to
pected that the difference in MW and MN between the films the mechanism(s) leading to a higher degradation rate of
would disappear in the course of the experiment. This was the nonporous PLLA film.
not the case, indicating another process having an effect on
MW and MN. At day 180 the MW and MN values of the Summary and Conclusions
“combi” film tend to approach the ones of the nonporous In summary, it can be stated that the parameters of
film. This trend corresponds with the erosion of the thin porosity and surface roughness do influence the ultimate
nonporous layer of the “combi” film as observed with SEM: biocompatibility of implanted biomaterials. Potential deg-
The “combi” film was becoming a porous one. Also, the MW radation of a biomaterial will enhance these reactions.
and MN show a lot of fluctuation during the first two weeks
of the experiment. These fluctuations might be caused by Porosity
two phenomena having opposite effects on MW and MN: A high porosity will, as a rule, induce a higher inflam-
washing out of low molecular weight fractions and molecu- matory response as compared to smooth and solid
lar chain fragmentation. However, as stated earlier, this re- biomaterials. The size of pores will, in turn, determine the
mains hypothetical, as the amount of polymer compared to type of foreign body reaction. Small (surface) pores (up to
the amount of buffer in which they were placed did not al- 10 microns) will give rise to more adhesion. In addition,
low for an analysis using GPC. guidance of cells may occur. Ingrowth will not take place.
Larger pores will facilitate tissue ingrowth and therewith
Degradation by Hydrolysis provide a mechanical anchorage of tissue to biomaterial.
Hydrolysis of the ester bonds is the major mechanism Optimal pore sizes have been estimated to be between 50
of PLLA degradation.32 Mechanical stress may enhance the and 200 microns.
degradation process.33 Radiation (UV, IR) or heat is not ex-
The Influence of Porosity and Surface Roughness on Biocompatibility 529
implants: A comparison of in vitro and in vivo models. J mances of biomaterials. Amsterdam: Elsevier,
Biomed Mater Res 1989; 23:1007-1026. 1986:227-233.
31. Krause TJ, Robertson FM, Liesch JB, Wasserman AJ, 34. Leenslag JW, Pennings AJ, Bos RRM, Rozema FR, Boering
Greco RS. Differential production of IL-1 on the surface G. Resorbable materials of poly-L-lactide VII: In vivo and
of biomaterials. Arch Sorg 1990; 125:1158-1160. in vitro degradation. Biomaterials 1987; 8:311-314.
32. Richardson MJ. Thermal analysis. In: Booth C, Price C, 35. Schakenraad JM, Hardonk MJ, Feijen J, Molenaar I,
eds. Comprehensive Polymer Science. Oxford: Pergamon Nieuwenhuis P. Enzymatic activity towards poly (L-lactic
Press, 1989:867-901. acid) implants. J Biomed Mater Res 1990; 24:529-545.
33. Jamshidi K, Hyon S-H, Nakamura T, Ikada Y, Teramatsu 36. Braber ET den, Ruitjer JE de, Smits HTJ, Ginsel LA,
Y. In vitro and in vivo degradation of poly-L-lactide fi- Recum AF von, Jansen JA. Quantitative analysis of cell
bers. In: Christel P, Neunier A, Lee AJC, eds. Advances proliferation and orientation on substrata with uniform
in biomaterials: Biological and biomechanical perfor- parallel surfaxe micro-grooves. Biomaterials 1996;
17:1093-1099.
Scaffold Engineering
Surface Modification
CHAPTER 48
Microgroove Driven Tissue Ingrowth
Edwin T. Den Braber, John A. Jansen
D uring the last two decades the availability and application of medical implants has in
creased dramatically. This concerns a broad variety of medical implants ranging from
knee prostheses to insulin infusion pumps, and from vascular grafts to pacemakers. Some
estimate figures were presented by Ratner1 in his Presidential Address for the Society for
Biomaterials in 1993 (Table 48.1). Although he emphasized that these figures were estimates,
it is clear that the use of implants is considerable. Long term projections even suggest that
implant applications are going to rise in the future. Factors that contribute to this increase
can be ascribed roughly to three major causes. First, the life expectancy of humans increases.
This will inevitably lead to a rise in the demand for implants like hip replacements or artifi-
cial lenses for the treatment of geriatric diseases and defects. Second, more and more medi-
cal treatments are going to include the use of implants in the future. One example of such a
development is the use of percutaneous implants in dialysis.2 Instead of treating patients
with chronic renal failure though intermittent hemodialysis, percutaneous implants enable
continuous ambulatory peritoneal dialysis (CAPD). Third, technologies are evolving that
open new ways of treating specific disorders or defects. This is demonstrated by techniques
that are being developed in the field of tissue engineering.3 Basically, tissue engineering
combines the principles and methods of the life sciences with those of engineering to eluci-
date fundamental understanding of structure-function relationships in normal and diseased
tissues, to develop materials and methods to repair damaged or diseased tissues, and to
create entire tissue replacements.4 Tissue engineering thus spans controlling cellular responses
to implant materials, manipulating the healing environment to control the structure of the
regenerated tissue, producing cells and tissues for transplantation into the body, and devel-
oping a quantitative understanding of many biological equilibrium and rate processes.5
Biomaterials play an important role in many of these activities. Originally, inertness
was thought to be one of the major contributions of the performance of an implant or
biomaterial, but later Williams adapted the definition of biocompatibility to include the
idea that a biomaterial perform with an appropriate host response in a specific application.6
Sadly, most currently used implant materials do not possess these desirable qualities. At his
Presidential Address, Ratner1 voiced this problem very vividly by saying, “For the majority
of our widely used bio-materials, no one sat down in advance and said ‘How can I engineer
the surface of this material to produce the desired biological response?’” He stressed that
most currently used biomaterials, although demonstrating generally satisfactory clinical
performance, were “ad-hoc” biomaterials, developed through a trial and error optimization,
rather than being engineered to produce, for instance, a desired interfacial interaction. The
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
532 Tissue Engineering of Prosthetic Vascular Grafts
prehensive listing of the most obvious and general accepted circular shape and size, washed, sterilized, and inspected as
alterations in cell behavior as a result of cells contacting sur- described elsewhere.18-19
faces with a pattern of parallel microgrooves. We will dem- For the evaluation of the cellular behavior on the
onstrate this by describing various studies that were per- smooth and microtextured silicone substrata, primary cul-
formed at our laboratories. In addition, we will discuss the ture rat dermal fibroblasts (RDFs) were used. These cells were
leading theories that attempt to create a model in which the harvested from ventral skin grafts taken from male Wistar
phenomenon of contact guidance is clarified. rats (100-120 g). After dissociation, these cells were incu-
bated for several days according to a specific protocol.18-19
Changed Cell Shape and Cellular Orientation Subsequently, the fifth generation was used for incubation
If cells are cultured on a surface with parallel micro- on the microtextured surfaces. Therefore, the smooth and
grooves as shown in Figure 48.1, the first thing that can be microtextured substrata were placed in the culture wells of
observed is the change of the shape of the cells. In earlier 24 well plates, after which approximately 1.0 x 104 viable
studies,18-19 we investigated several groove and ridge dimen- RDFs ml-1 were added to each substratum. The RDFs were
sions to study and quantify the influence of these groove incubated on a specific substratum up to 7 days under static
patterns on the size, shape, and orientation of the cells cul- conditions.
tured on these substrata. The effect of the surface microgeometry on the cellu-
In order to obtain a microgrooved substratum, we first lar morphology was quantified by digital image analysis.18-19
produced silicon oxide molds in a class 100 clean room us-
ing photolithography.20-21 Photolithography is a technique
that is developed for the production of microelectronic com-
ponents like, for example, computer microcircuits. In our Table 48.2.Dimensions of the micro features on the
experiments, these mold surfaces were produced by coating substrata surfaces (Gd=groove depth, Gw=groove
silicon oxide masks with high reflective chrome, after which width, Rw=ridge width, and P=pitch).
the chrome was coated with a thin (0.5 ∝m) layer of positive
photoresist. Subsequently, the photoresist was exposed and Gd Gw Rw P
developed, uncovering the underlying chrome, which was (∝
∝m) (∝
∝m) (∝
∝m) (∝
∝m)
etched. Finally, the unexposed photoresist was stripped off,
thus creating a parallel groove pattern. The substrata were 1.00 1.00 1.00 2.00
finally obtained by covering the molds with polydimethyl- 1.00 1.00 2.00 3.00
1.00 1.00 4.00 5.00
siloxane, thus producing negative surface replicas which
1.00 1.00 8.00 9.00
possessed either a smooth surface or a surface with parallel 1.00 4.00 1.00 5.00
grooves, showing a wide variety of groove and ridge widths 1.00 8.00 1.00 9.00
(Table 48.2). During our experiments, the groove depth was 0.45 2.00 2.00 4.00
either 0.5 ∝m or 1.0 ∝m. After polymerization, the silicone 0.45 5.00 5.00 10.00
rubber castings were removed, cut into their appropriate 0.45 10.00 10.00 20.00
In short, RDFs were photographed by phase contrast mi- the microtextured substratum surfaces and the overlaying
croscopy on every day of the incubation period. These pho- fibroblasts. During these additional experiments, we cultured
tographs were scanned digitally and analyzed with an image the RDFs similarly as in the earlier studies,18-19 but visualized
analysis program. This program made it possible to mea- specific cytoskeletal components and proteins interacting
sure several cell parameters, i.e., the cellular surface area, in the attachment of the RDFs with the following mono-
cellular perimeter, cellular circularity, maximum cell length, clonal and polyclonal antibodies:22
cell breadth perpendicular to the maximum length, the angle 1. The mouse monoclonal antibody hVIN-1, specific
of cellular orientation relative to the surface grooves (#), and for vinculin;23
number of pitches spanned by a single cell (Fig. 48.2). 2. The rabbit monoclonal anti-fibronectin antibody
These studies provided a lot of interesting data, but FN-3E2;24
most of all clearly showed that the RDFs on surfaces with a 3. A rabbit polyclonal antiserum raised against bovine
ridge width smaller than 4.0 ∝m were highly orientated fibronectin;25
(# < 10°) and elongated along the surface grooves (Figs. 48.3 4. The rabbit polyclonal antiserum against bovine
and 48.4). However, if the ridge had a width larger than vitronectin;26
4.0 ∝m, then the cellular orientation was random (Χ45°) and 5. A polyclonal antiserum raised against human
the shape of the RDFs became significantly more circular vitronectin in rabbits, which has been shown to have
(Fig. 48.5). The appearance and orientation of the cells on a good crossreactivity with rat vitronectin, but not
the surfaces with ridge widths larger than 4.0 ∝m in many with bovine vitronectin.27
cases proved to be not significantly different from those on After incubation, these primary antibodies were
the smooth substratum surfaces (Fig. 48.6). Furthermore, it incubated with the appropriate secondary antibody, i.e., fluo-
became apparent that the ridge width was the most impor- rescein isothiocyanate (FITC)-conjugated goat anti-mouse
tant parameter, since varying the groove width and groove IgG or FITC-conjugated goat anti-rabbit IgG. For the
depth did not affect the RDF size, shape, or the angle of staining of the RDF filamentous actin no antibodies were
cellular orientation (#) significantly. used, since F-actin was visualized with thiorhodamine
isothiocyanate (TRITC)-labeled phalloidin. Immediately
Cell Attachment on Microtextured Surfaces after performing the double stain procedures, the RDFs were
During these studies, we found after careful examina- examined with a confocal laser scanning microscope
tion of the phase contrast images that the fibroblasts showed (CLSM), which was equipped with a krypton/argon mixed
indications of attaching specifically to the ridges of the sur- gas laser. This type of laser offers separate, well spaced wave-
face patterns (Fig. 48.4). If the fibroblasts did actually attach lengths for the excitation of FITC (ϑ = 488 nm) and TRITC
solely to the surface ridges, this would mean that the cells (ϑ = 568 nm).28 Next to the fluorescence mode, the reflection
displayed a specific preference of attachment location. In- mode of the CLSM was used to visualize the underlying sub-
trigued by the phase contrast microscopy/digital image stratum surface.22 The resulting digital images were captured
analysis results, we performed experiments which were de- and overlay images were created, thus making it possible to
signed to show us more of the intricate interaction between capture the fluorescent and reflection data in one 24 bit RGB
Microgroove Driven Tissue Ingrowth 535
(Red-Green-Blue) picture. Creation of these 24 bit images vinculin relative to the grooves and ridges of the
enabled the composition of 1 digital image with 3 different microtextured surface was charted and analyzed.22
information levels, each within one color segment, and of- The results of the CLSM observations and additional
fered the possibility to investigate the stained objects in con- image and statistical analysis showed that the microfilaments
junction with each other and the surface patterns. First, the and vinculin aggregates of the RDFs on the 2.0 ∝m grooved
(acute) angle of orientation of F-actin, vinculin, fibronectin, substrata were orientated along the surface grooves, while
vitronectin, and the surface grooves relative to a virtual X-Y these proteins were significantly less orientated on the 5.0
axis were measured. Second, the relative position and the and 10.0 ∝m grooved surfaces (Fig. 48.7). In contrast, bo-
angle of the linear components of vinculin, fibronectin, and vine and endogenous fibronectin and vitronectin were ori-
vitronectin were compared with the position and angle of entated along the surface grooves on all textured surfaces.
orientation of the actin filaments. Finally, the location of These extracellular proteins did not seem to be hindered by
536 Tissue Engineering of Prosthetic Vascular Grafts
the surface grooves, since many groove spanning filamen- vealed that these focal adhesion points were occasionally
tous deposits were found on all microgrooved surfaces wrapped around the edges of the ridges. Attachment of the
(Fig. 48.8). Vinculin was located mainly on the surface ridges cells on the silicone rubber and titanium microtextured sub-
on all textured surfaces (Fig. 48.9). strata was never observed on the surfaces with 1.0 or 2.0 ∝m
These findings were later confirmed in additional stud- grooves. Only the RDFs on the 5.0 ∝m and 10.0 ∝m grooved
ies.29-30 During these investigations we made ultrathin sec- surfaces protruded into the grooves, while attachment to the
tions of RDFs cultured on silicone and titanium groove floor was observed only on the 10.0 ∝m textures. In
microgrooved surfaces with parallel grooves ranging from addition, only the RDFs on the titanium 5.0 ∝m and 10.0
1.0 ∝m up to 10.0 ∝m. The depth of these grooves varied ∝m grooved surfaces possessed focal adhesion points on the
between 0.45 ∝m and 2.2 ∝m. Transmission electron micros- walls of the grooves (Fig. 48.10). Comparison between the
copy (TEM) again showed that the focal adhesion points of observations of the cells on the microtextured silicone rub-
the RDFs on both the silicone rubber and titanium ber and titanium substrata suggested that material specific
microtextured surfaces were located mainly on the surface properties did not influence the orientational effect of the
ridges (Fig. 48.10). On the titanium surfaces, TEM also re- surface texture on the observed RDF cellular behavior.30
Microgroove Driven Tissue Ingrowth 537
Other Surface Topography Induced Cell changes in cellular differentiation, DNA/RNA transcription,
Behavior Alterations In Vitro cell metabolism, and cellular protein production.16-17,34-35
Apart from changes in cell size, shape, orientation, and For example, increased F-actin content and increased per-
attachment, investigators have suggested and studied many sistence and speed of cell movement of macrophage-like cells
other cell processes that could possibly be influenced by the on grooved surfaces were observed, and changes in the regu-
surface topography of substrata. One of these processes is lation of fibronectin mRNA levels, mRNA stability, and
cell proliferation. On the basis of the results of some of our fibronectin stability and assembly on surfaces with micro-
earlier studies,18,31 we concluded that the presence or dimen- grooves have been reported. Similar results were described
sions of the parallel surface grooves as used in our experi- by Hong and Brunette,36 who saw clear differences in the
ments did not result in a change in RDF proliferation rate. fibronectin mRNA and proteinase levels between oral epi-
This conclusion was, however, in contrast to the findings of thelial cells that were cultured on smooth and microgrooved
Green et al and Ricci et al. For example, Green et al32 re- surfaces. Consequently, they remarked that these surface
ported that especially abdomen fibroblasts (CCD-969sk) topography induced changes could be considered as “good
cultured on surfaces with 2.0 and 5.0 ∝m square pillars news/bad news”. According to these investigators, the good
showed increased proliferation rates. In addition, Ricci et news was the fact that specific surface topographies could
al33 evaluated the in vitro growth of rat tendon fibroblasts be used to enhance the production of a specific protein, while
and rat bone marrow colonies on unidirectional (grooved) the bad news might be that the production/secretion of other
surface microgeometries. They found that the overall colony proteins might also be enhanced. If the production/secre-
growth rate was changed, and concluded that surface tion of these “other proteins” would be elevated, this might
microgeometry could be used to control the growth rate at have a deleterious effect on the integration of the implant.
implant surfaces. However, this study by Ricci et al also For example, a rise in the production/release of proteinases
showed that the response to surface topography is depen- could result in a large scale degradation of the connective
dent on cell type, which could account for the discrepancy tissue. This simple example demonstrates that, at the mo-
in the results that we found between the proliferation rates lecular level, the regulation of cell function by altering the
of our rat dermal fibroblasts, the results of Ricci et al, and surface topography of an implant or substratum could be a
the data of Green et al. A point of discrepancy could be the very complex affair.
use of different surface textures. Although Ricci et al used a
pattern of parallel grooves, Green et al used a texture con- Microtextured Surfaces In Vivo
sisting of square pillars. This could mean that we not only Up to this point, we have only described the effect of
have to consider the possibility of cell type dependent dif- microtextured surfaces on cells in vitro. However, the re-
ferences in cell proliferation, but also have to contemplate sults of these in vitro studies have led to the idea that sur-
the effect that different surface textures could trigger. face microtexturing could be used deliberately to achieve
Another process that seems to be affected by the to- certain desired end results in processes like morphogenesis,
pography of the surface that the cell contacts is that of the cell invasion, repair, and regeneration.4 If this hypothesis
cell metabolism. Several publications have reported behav- proves to be true, it is obvious that surface texturing can be
ioral differences of cells on microtextured surfaces, such as a very important tool in designing successful implants.17,37
Microgroove Driven Tissue Ingrowth 539
Sadly, in vivo studies with microtextured implants challeng- gradually after cell-cell contacts are formed.18,37,43 In tissues,
ing these ideas are scarce. In addition, review of these in vivo these contacts with other cells are already present, which
studies shows that the design of the used textured implants could mean that the orientational effect of the textured sur-
is very diverse. But even with this large diversity, it is pos- faces is overruled by stronger tissue related signals or cues.
sible to perceive the possible potential of microtextured im- Three dimensional reconstruction of CLSM images
plant surfaces on several implant related processes. For ex- and normal light microscopy showed no significant differ-
ample, some studies38-39 have reported on the reduction of ences between the thickness of the capsule surrounding the
epithelial downgrowth with microgrooved skin penetrating smooth and microgrooved implants. Since differences be-
devices. Other investigators, who implanted microporous tween the 2.0, 5.0, and 10.0 ∝m grooved implants were not
or pillared surfaces subcutaneously, found tightly adherent detected, we concluded that the depth of the grooves used
fibrous capsules without inflammatory cells,40 reduced fi- was not sufficient to facilitate “mechanical interlocking”. This
brosis,41 and improved blood vessel proximity.41 interlocking would reduce the stress and movement at the
In order to investigate the effect of microtextured im- implant interface and limit the consequential “mechanical
plants on the surrounding tissue and to be able to compare irritation” of the surrounding tissues, which is supposed to
our in vitro and in vivo data, we implanted the smooth and induce tissue damage, fibrosis, and severe inflammatory re-
microtextured silicone rubber substrata with 2.0, 5.0, and sponses.16-17,40-41 Other investigators have indeed reported
10.0 ∝m grooves (Table 48.2) subcutaneously in a total of 12 reduction of the capsule size due to microtextured surfaces.
female New Zealand White rabbits for periods of 3, 7, 42, However, review of these studies40-41,44-45 shows that the sur-
and 84 days.42 Scanning electron microscopy (SEM) showed face texture of the implants in these studies differs signifi-
fibroblasts, erythrocytes, lymphocytes, macrophages, fibrin, cantly from our implants, both in terms of microfeature
and collagen on all implant surfaces after 3 and 7 days appearance (pores, pillars, tapered pits, or V-shaped grooves)
(Fig. 48.11). After 42 and 84 days only a little collagen and a and dimensions (feature depth, size, and pitch). As a result,
small number of fibroblasts, but no inflammatory cells, were the mechanical irritation of the smooth and grooved sur-
seen on the implant surfaces. In contrast with the RDFs in faces would be comparable, resulting in capsules of equal
the in vitro experiments, the fibroblasts that were observed thickness. Furthermore, our textured implants possessed one
on the microtextured implant surfaces, were not orientated smooth and one textured side. Although this opened up the
along the surface grooves. A possible explanation for these possibility for intraimplant evaluation, it did not enhance
differences between in vitro and in vivo orientational cell possible mechanical interlocking between the implant and
behavior could be that the cells that are used in in vitro stud- the surrounding tissues. Therefore, it can be questioned
ies are isolated cells, which have no contact with other cells, whether the capsule thickness would have been less if both
cell types, or ECM. Previous studies18,37,43 have shown that sides of the implant had been textured.
prolonged in vitro incubation on microtextured surfaces re- Furthermore, normal light microscopy did show a sig-
sults in the formation of cell-cell contacts, an increase of the nificantly lower number of inflammatory cells, and a sig-
spread area, and a decrease of the orientation of the cells on nificantly higher number of blood vessels, in the capsules
these surfaces. Consequently, it was supposed that the ob- surrounding the microgrooved implants. The cause of these
served guidance phenomenon is an initial response of cells differences remains unclear. However, it is possible to specu-
in vitro to certain microtextured surfaces, which is lost late that the differences in the number of blood vessels were
part of the proliferation phase of the wound healing pro- energy have an influence on the displayed cellular behavior.
cess.46 This proliferation phase is a part of the formation of The vinculin location and orientation however, pleas in fa-
granulation tissue, which is characterized by high fibroblast vor of the “ridge width” theory, although warped focal ad-
densities, the formation of new blood vessels, and a new hesion points that were found in the TEM results30 seem to
connective tissue matrix.46 After repair, the number of the contradict this theory. Finally, whether the cells orientate to
vessels decreases generally, marking the end of the wound the microtextured surfaces as a result of the force distribu-
healing process and the start of a steady state. The fact that tion that is created by the texture of these surfaces is impos-
more vessels were observed around the textured implants sible to determine since no (known) studies have been per-
during our study could indicate a higher rate of tissue re- formed to map the force distribution within cells cultured
pair around these implants. Further research is, however, on microgrooved substratum surfaces. Recognizing the fact
required to determine if this in fact is the case. that these three hypotheses can even be integrated into one
overall model contributes to the intriguing phenomena of
The Hypotheses cellular behavior on microtextured surfaces.
In the previous paragraphs we have presented several
studies with microgrooved (implant) surfaces. In spite of Future Perspectives: Vascular Grafts
the fact that several publications like these and some excel- and Microtextured Surfaces
lent reviews 3,16-19,31,37 have reported on the effects of As mentioned before, many investigators have already
microtextured surfaces, little is known about the exact speculated on the benefits of microtextured implants. For
mechanism whereby surface topography exerts its effects. example, Ratner1 speculated on the benefits of an implant
Several theories have been suggested, however. First, it has surface that would not cause the formation of a fibrous cap-
been hypothesized that wettability plays a role in these phe- sule. According to Ratner, such an implant would not “be
nomena. A microtextured surface could possess local differ- walled off ”, but the cells contacting the implant surface would
ences in surface free energy which promote a specific depo- respond “as if they are not seeing and interacting with the
sition pattern of the substratum-bound attachment pro- biomaterial”. This wound healing reaction would be prefer-
teins.16-17,47-49 In addition, the spatial arrangement of the able for the clinical success of several frequently used im-
adsorbed proteins50-52 and the conformation of these pro- plants. For example, reduction of the capsule thickness
teins53-54 would be influenced by these substratum surface around an implant would mean a reduction of the capsule
properties. Second, it has been suggested that the specific contraction that is often observed with, for example, sili-
geometrical dimensions of the focal adhesion plaques force cone breast implants.66 Furthermore, capsule reduction
a cell on a surface with small grooves and ridges to orientate would enhance the performance of many implanted
itself parallel to these ridges.55-56 This hypothesis is based biosensors, pacemakers, and infusion pumps.17 These de-
on the observation that a minimum length of 2.0 ∝m is re- vices all benefit from an optimal contact between the tissues
quired for focal contacts to establish adhesion.57 This im- and the implant for the transduction of signals. For instance,
plies that, if the ridge width increases, multiple vectors of the necessary electrical pulse of a pacemaker would be bet-
adhesion plaque orientation are possible, enabling less ori- ter conducted to the heart muscle if the capsule around the
entated cell attachment. Finally, a third hypothesis58-59 sup- electrode of this device were minimized. Another good ex-
poses that the orientation and alignment of cells on ample is the sensor of an implanted insulin infusion pump.
microtextured surfaces are a part of the cellular efforts to For optimal detection of insulin levels, maximal contact
reach a biomechanical equilibrium with the net sum of forces between the sensor of this device and the surrounding tis-
minimized. This phenomenon has, for instance, been de- sues is required. However, if a fibrous capsule shields the
scribed extensively in the so-called tensegrity models.58-60 sensor from the crucial signal, the performance of the im-
According to these models, it is possible that the anisotropic plant will be insufficient.
geometry of substratum surface grooves and ridges estab- Additional applications of microtextured biomaterials
lishes stresses and shear-free planes that influence the direc- have been reported in the discipline of tissue engineering.
tion of microtubule61 and microfilament growth62-63 in or- For example, microtextured surfaces have been used in in
der to create a force economic situation. vitro experiments to decrease hepatocyte dedifferentia-
Given the current available information, it is impos- tion67-68 or to induce guided nerve regeneration.42,69-70 Skin
sible to express an opinion on which or whether one of these autografts already have been generated out of individual
hypotheses is true. On the other hand, several separately keratinocytes by using orienting scaffolds,71 while specula-
performed studies have reported that surface microtextrues tions are voiced that guided tissue regeneration could per-
can have a profound effect on specific elements of the cy- haps reduce the formation of scarring tissue and enhance
toskeleton like, for example, the microfilament bundles,62-65 the repair of highly orientated structures like tendons.46,72-73
focal contacts,55 and microtubules,61 making it safe to say Furthermore, attempts have been made to produce large
that microtextured surfaces influence the orientation of the tubular morphologies with the help of tissue engineering
intracellular and extracellular proteins. Although results of that could function as intestine or ureter segmental replace-
our studies corroborate with all three hypotheses, they do ments.74 In addition, many publications have reported on
not justify a specific choice for one of these hypotheses. The efforts to produce blood vessels.1,3,75-82 Microtextured
differences in deposition patterns and the appearances of biomaterials could be very useful as scaffolds in this field of
the ECM proteins during the CLSM study for example, make tissue engineering research.
it possible to suggest that surface properties like surface free
Microgroove Driven Tissue Ingrowth 541
Figure 48.12 shows the basic anatomy of a blood ves- possibly be obtained by applying longitudinal or circular
sel. Large blood vessels have a thick, tough wall of connec- groove patterns. Not only would the overall orientation of
tive tissue and smooth muscle, which is lined by an exceed- the cell be influenced, but also the orientation of the cytosk-
ingly thin, single layer of endothelial cells, separated from eleton, thus contributing to the mechanical stiffness of the
the surrounding outer layers by a basal lamina. The thick- cells.84 By using microgrooved scaffolds, endothelial orien-
ness of the connective tissue component of the vessel wall tation could be achieved without exposing the cells to flow
varies with the vessel’s diameter and function, but the en- and cyclic stretch, phenomena that have a comparable ef-
dothelial lining is always present.83 Study of embryos has fect on the orientation of endothelial cells.85-86 This com-
revealed that large blood vessels have all developed from parison also concerns the level of gene expression. It has been
small simple vessels constructed solely out of endothelial reported that flow and cyclic stretch can influence the regu-
cells and a basal lamina.83 Therefore, the endothelial cells lation of messenger RNA,87-88 an effect on the cellular me-
are often referred to as the pioneers, preceding the develop- tabolism that is also supposed for cells on microtextured
ment of connective tissue and smooth muscle around the surfaces. What kind of effect the combination of the effects
vessels when required. This is why early studies have con- of factors like surface texture, flow, and cyclic stretch will
centrated on the formation of capillary tubes out of colo- have remains to be seen, since no study investigating this
nies of endothelial cells.76 However, the tissue engineering combination has been performed up to this date.
of a functional, well developed substitute blood vessel does Another interesting option concerns the extracellular
not depend solely on the presence of vascular cells and ex- matrix proteins. The endothelial cells and smooth muscle
tracellular matrix (ECM) components. Several cellular sig- cells in vivo are surrounded by an intricate mixture of pro-
nals are equally important to processes such as cell growth teins like collagens, elastin, laminin, fibronectin, and
and cellular differentiation. Ziegler and Nerem79 identified glycoaminoglycans.79 Several studies have shown that these
these signals as originating from three sources, i.e.: proteins can affect the growth, differentiation, and choles-
1. Chemical signals, derived from the fluid (blood) terol metabolism of both the endothelial and smooth muscle
flowing through the vessel; cells. Endothelial derived ECM can affect smooth muscle
2. Signals associated with the ECM—the ECM proteins cell growth, depending on its composition.89 Collagen and
not only hold the vascular wall together, but also par- fibronectin containing matrices promote the growth of
ticipate in regulating the biology of the vascular wall; smooth muscle cells, while matrices containing heparan sul-
3. The mechanical environment of the vascular wall, fate proteoglycans selectively inhibit identical smooth muscle
imposed by the hemodynamics of the vascular sys- cell populations.89 Furthermore, ECM proteins can change
tem. the response of smooth muscle cells to low density lipopro-
If we speculate on the roles that microtextured scaf- teins (LDL). Earlier study90 has shown that collagen type I,
folds could play in meeting these demands, one is evident. for example, induces a decrease in smooth muscle cell
The orientation of the endothelial cells in the direction of growth, while endothelial derived ECM induces an increase
the flow, and circularly orientated smooth muscle cells, could in smooth muscle cell growth following incubation with
LDL.90 The ability of microtextured surfaces to enhance the 13. Rovensky YA, Slavnaya IL. Spreading of fibroblast-like
production of specific proteins as mentioned earlier8 could, cells on grooved surfaces. Exp Cell Res 1974; 84:199-206.
if true, prove a powerful tool in the creation of blood vessels 14. Maroudas NG. Anchorage dependence: Correlation be-
through tissue engineering. tween amount of growth and diameter of bead, for single
cells grown on individual glass beads. Exp Cell Res 1972;
These few examples show that microtextured surfaces
74:337-342.
could contribute in the study of, and the creation of, vascu- 15. Maroudas NG. Growth of fibroblasts on linear and pla-
lar grafts. This ranges from in vitro study of smooth muscle nar anchorages of limiting dimensions. Exp Cell Res 1973;
cells, where these surfaces could stop/slow down the change 81:104-110.
from a nongrowing, contractile phenotype to a proliferat- 16. Singhvi R, Stephanopoulos G, Wang DIC. Review: Effects
ing, protein secreting mode,91-92 to the induction of endothe- of substratum morphology on cell physiology. Biotech-
lial and smooth muscle cell orientation in artificial grafts.1,75 nology and Bioengineering, 1994; 43:764-771.
Therefore, it is recommended that research of microtextured 17. von Recum AF, van Kooten TG. The influence of micro-
surfaces be exploited further in order to develop “smart” topography on cellular response and the implications for
manipulative implants with higher clinical success rates. In silicone implants. J Biomater Sci Polymer Edn 1995;
addition, these new studies could also contribute to a better 7:181-198.
18. den Braber ET, de Ruijter JE, Smits HTJ et al. Quantita-
understanding of the mechanisms that cause cellular con-
tive analysis of cell proliferation and orientation on sub-
tact guidance and guidance related processes. Such insight strata with uniform parallel surface micro grooves.
would not only enlarge the general knowledge of these Biomaterials 1996; 17:1093-1099.
processes, but also offer implant designers a tool for design- 19. den Braber ET, de Ruijter JE, Ginsel LA et al. Quantita-
ing a wider variety of more successful implants with tive analysis of fibroblast morphology on microgrooved
predicable qualities. surfaces with various groove and ridge dimensions.
Biomaterials 1996; 17:2037-2044.
Acknowledgments 20. Schmidt JA, von Recum AF. Texturing of polymer sur-
This study is supported by the Technology Founda- faces at the cellular level. Biomaterials 1991; 12:385-389.
tion (STW). 21. Schmidt JA, von Recum AF. Surface characterization of
microtextured silicone. Biomaterials 1992; 13:675-681.
22. den Braber ET, de Ruijter JE, Ginsel LA et al. Confocal
References
laser scanning microscopical study of the fibroblast cy-
1. Ratner BD. New ideas in biomaterial science—a path to
toskeleton, attachment complexes, and ECM protein depo-
engineered biomaterials. J Biomed Mater Res 1993;
sition on silicone microgrooved surfaces. J Biomed Mater
27:837-850.
Res 1997; in press.
2. von Recum AF, Park JB. Permanent percutaneous devices.
23. Benori R, Salomon D, Geiger B. Identification of two dis-
CRC Crit Rev Bioeng 1981; 5:37-77.
tinct domains on vinculin involved in its association with
3. Hubbell JA. Biomaterials in tissue engineering. Biotech-
focal contacts. J Cell Biol 1989; 108:2383-2393.
nology 1995; 13:565-576.
24. Garbarsch C, Matthiessen ME, Olsen BE et al. Immuno-
4. Langer R, Vacanti JP. Tissue engineering. Science 1993;
histochemistry of the intercellular matrix components and
260: 920-926.
the epithelio-mesenchymal junction of the tooth germ.
5. Peppas NA, Langer R. Challenges in biomaterials. Science
Histochem J 1994; 26:110-118.
1994; 263:1715-172.
25. Hayman EG, Oldberg A, Martin GR et al. Co-distribu-
6. Williams DF. Definitions in Biomedicals. Progress in Bio-
tion of heparan sulfate proteoglycan, laminin, and
medical Engineering. New York: Elsevier, 1987:Vol. 4.
fibronectin in extra cellular matrix of normal rat kidney
7. Hockberger PE, Lom B, Soekarno A, Thomas CH, Healy
cells and their coordinate absence in normal cells. J Cell
KE. Cellular engineering: Control of cell-substrate inter-
Biol 1982; 94:28-35.
actions. In: Hoch HC, Jelinski LW, Craighead HG, eds.
26. Hayman EG, Pierschbacher MD, Suzuki S et al.
Nanofabrication and Biosystems. Cambridge: Cambridge
Vitronectin; a major cell attachment-promoting protein
University Press, 1996:276-299.
in fetal bovine serum. Exp Cell Res 1985; 160:245-258.
8. Brunette DM. Effects of surface topography of implant
27. Hayman EG, Pierschbacher MD, Ohgren Y et al.
materials on cell behavior in vitro and in vivo. In: Hoch
Serumspreading factor (vitronectin) is present at the cell
HC, Jelinski LW, and Craighead HG, eds. Nanofabrication
surface and in tissues. Proc Natl Acad Sci 1983;
and Biosystems. Cambridge: Cambridge University Press,
80:4003-4007.
1996: 335-355.
28. Cox G. Trends in confocal microscopy. Micron, 1993;
9. Harrison RG. The reaction of embryonic cells to solid
24:237-247.
structures. J Exp Zool 1914; 17:521-544.
29. den Braber ET, de Ruijter JE, Croes HJE et al. Transmis-
10. Loeb L, Fleisher MS. On the factors which determine the
sion electron microscopical study of fibroblast attachment
movements of tissues in culture media. J Med Res 1917;
to microtextured silicone rubber surfaces. Cells and Ma-
37:75-99.
terials 1997; in press.
11. Weiss P. Experiments on cell and axon orientation in
30. den Braber ET, Jansen HV, de Boer MJ et al. SEM, TEM,
vitro: The role of colloidal exudates in tissue organiza-
and CLSM observation of fibroblasts cultured on
tion. J Exp Zool 1945; 100:353-386.
microgrooved surfaces of bulk titanium susbtrata. J
12. Rovensky YA, Slavnaya IL, Vasiliev JM. Behavior of fi-
Biomed Mater Res 1997; submitted.
broblast-like cells on grooved surfaces. Exp Cell Res 1971;
31. den Braber ET, de Ruijter JE, Smits HTJ et al. Effect of
65:193-201.
parallel surface micro grooves and surface energy on cell
growth. J Biomed Mater Res 1995; 29:511-518.
Microgroove Driven Tissue Ingrowth 543
32. Green AM, Jansen JA, and von Recum AF. The fibroblast 51. Rudee ML, Price TM. The initial stages of adsorption of
response to microtextured silicone surfaces: Texture ori- plasma derived proteins on artificial surfaces in a con-
entation into or out of the surface. J Biomed Mater Res trolled flow environment J Biomed Mater Res 1985;
1994; 28:647-653. 19:57-66.
33. Ricci JL, Charvet J, Chang R et al. In vitro effects of sur- 52. Uyen HMW, Schakenraad JM, Sjollema J et al. Amount
face microgeometry on colony formation by fibroblasts and surface structure of albumin adsorbed to solid sub-
and bone cells. 20th Annual Meeting of the Society for strata with different wettabilities in a parallel plate flow
Biomaterials. Boston, USA. April 5-9, 1994:401. cell. J Biomed Mater Res 1990; 24:1599-1614.
34. Chou LS, Firth JD, Uitto VJ et al. Substratum surface to- 53. Rapoza RJ, Horbett TA. Postadsorptive transitions in fi-
pography alters cell shape and regulates fibronectin mRNA brinogen: Influence of polymer properties. J Biomed
level, mRNA stability, secretion and assembly in human Mater Res 1990; 24:1263-1287.
fibroblasts. J Cell Sci 1995; 108:1563-1573. 54. Shiba E, Lindon JN, Kushner L et al. Antibody detectable
35. Wójciak-Stothard B, Madeja Z, Korohoda W et al. Acti- changes in fibrinogen adsorption affecting platelet activa-
vation of marcophage-like cells by multiple grooved sub- tion on polymer surfaces. Am J Physiol 1991;
strata. Topographical control of cell behavior. Cell Biol- 260:C965-974.
ogy International, 1995; 19:485-490. 55. Ohara PT, Buck RC. Contact guidance in vitro. Exp Cell
36. Hong HL and Brunette DM. Effect of cell shape on pro- Res 1979; 121:235-249.
teinase secretion. J Cell Sci 1987; 87:259-267. 56. Dunn GA, Brown AF. Alignment of fibroblasts on grooved
37. Curtis ASG, Clark P. The effects of topographic and me- surfaces described by a simple geometric transformation.
chanical properties of materials on cell behavior. Critical J Cell Sci 1986; 83:313-340.
Reviews in Biocompatibility 1990; 5:344-362. 57. Izzard CS, Lochner LR. Cell-to-substrate contacts in liv-
38. Chehroudi B, Gould TRL, Brunette DM. Effects of a ing fibroblasts: An interference reflection study with an
grooved epoxy substratum on epithelial cell behavior in evaluation of the technique. J Cell Sci 1976; 21:129-159.
vitro and in vivo. J Biomed Mater Res 1988; 22:459-473. 58. Ingber DE. Cellular tensegrity; defining new rules of bio-
39. Chehroudi B, Gould TRL, Brunette DM. A light and elec- logical design that govern the cytoskeleton. J Cell Sci 1993;
tron microscope study of the effects of surface topogra- 104:613-927.
phy on the behavior of cells attached to titanium-coated 59. Ward MD, Hammer DA. A theoretical analysis for the
percutaneous implants. J Biomed Mater Res 1991; effect of focal contact formation on cell-substrate attach-
25:387-405. ment strength. Biophys J 1993; 64:936-959.
40. Campbell CE, von Recum AF. Microtopography and soft 60. Wang N, Butler JP, Ingber DE. Mechanotransduction
tissue response J Invest Surg 1989; 2:51-74. across the cell surface and through the cytoskeleton. Sci-
41. Picha GJ, Drake RF. Pillared-surface microstructure and ence, 1993; 260:1124-1127.
soft-tissue implants: Effect of implant site and fixation. J 61. Oakley C, Brunette DM. The sequence of alignment of
Biomed Mater Res 1996; 30:305-312. microtubules, focal contacts and actin filaments in fibro-
42. den Braber ET, de Ruijter JE, Jansen JA. The effect of a blasts spreading on smooth and grooved titanium sub-
subcutaneous silicone rubber implant with shallow sur- strata. J Cell Sci 1993; 106:343-354.
face micro grooves on the surrounding tissues in rabbits. 62. O’Neill C, Jordan P, Riddle P et al. Narrow linear strips
J Biomed Mater Res 1996; in press. of adhesive substratum are powerful inducers of both
42a. Clark P, Connolly P, Curtis ASG et al. Cell guidance by growth and total focal contact area. J Cell Sci 1990;
ultrafine topography in vitro. J Cell Sci 1991; 99:73-77. 95:577-586.
43. Squier CA, Collins P. The relationship between soft tis- 63. Oakley C, Brunette DM. Topographic compensation:
sue attachment, epithelial downgrowth and surface po- Guidance and directed locomotion of fibroblasts on
rosity. J Perio Res 1981; 16:434-440. grooved micromachined substrata in the absence of mi-
44. Chehroudi B., Gould TRL., and Brunette DM. The role crotubules. Cell Motility and the Cytoskeleton 1995;
of connective tissue in inhibiting epithelial downgrowth 31:45-58.
on titanium-coated percutaneous devices. J. Biomed. 64. Ben-ze’ev A. The role of changes in cell shape and con-
Mater. Res., 1992; 26: 493-515. tacts in the regulation of cytoskeleton expression during
46. Ehrlich HP. Regulation der Wundheilung aus der Sicht differentiation. J Cell Sci Suppl 1987; 8:293-312.
des Bindesgewebes. Der Chirurg, 1995; 66: 165-173. 65. Dunn GA, Heath JP. A new hypothesis of contact guid-
47. Baier RE. Surface properties influencing biological adhe- ance in tissue cells. Exp Cell Res 1976; 101:1-14.
sion. Adhesion in biological systems, RS Manly (ed.) Aca- 66. Bern S, Burd A, May J Jr. The biophysical and histologic
demic Press, New York, 1970, 15-48. properties of capsules formed by smooth and textured sili-
48. Schakenraad JM, Busscher HJ, Wildevuur CRH et al. The cone implants in the rabbit. Plast Reconstr Surg 1992;
influence of substratum surface free energy on growth and 89:1037-1042.
spreading of human fibroblasts in the presence and ab- 67. Cima LG, Ingber DE, Vacanti JP et al. Hepatocyte cul-
sence of serum proteins. J Biomed Mater Res 1986; ture on biodegradable polymeric substrates. Biotech.
20:773-784. Bioengineering 1991; 38:145-158.
49. Altankov G, Groth TH. Reorganization of substratum- 68. Dunn JCY, Thomkins RG, Yarmush ML. Hepatocytes in
bound fibronectin on hydrophillic and hydrophobic ma- collagen sandwich: Evidence for transcriptional and trans-
terials is related to biocompatibility. J Mater Sci 1994; lational regulation. J Cell Biol 1992; 116:1043-1053.
5:732-737. 69. Guenard V, Kleitman N, Morrissey TK et al. Syngeneic
50. Williams RL, Williams DF. The spatial resolution of pro- Schwann cells derived from adult nerves seeded in semi-
tein adsorption on surfaces of heterogeneous metallic permeable guidance channels enhance the peripheral nerve
biomaterials. J Biomed Mater Res 1989; 23:339-350. generation. J Neurosci 1992; 12:3310-3320.
544 Tissue Engineering of Prosthetic Vascular Grafts
70. Clark P, Connolly P, Curtis ASG et al. Topographical 81. Langer R, Vacanti JP, Vacanti CA et al. Tissue engineer-
control of cell behavior: II. Multiple grooved substrata. ing: Biomedical applications. Tissue Engineering 1995;
Development 1990; 108:635-644. 1:151-161.
71. Bell E, Rosenberg M, Kemp P et al. Recipes for reconsti- 82. Bell E. Strategy for the selection of scaffolds for tissue
tuting skin. J Biomech Eng Trans ASME, 1991; engineering. Tissue Engineering 1995; 1:163-179.
113:113-119. 83. Alberts B, Bray D, Lewis J et al. Molecular biology of the
72. Chehroudi B, Gould TRL, Brunette DM. A light and elec- cell, 3rd edition. New York: Garland Publishing Inc., 1994.
tron microscope study of the effects of surface topogra- 84. Nerem RM, Girard PR. Hemodynamic influences on vas-
phy on the behavior of cells attached to titanium-coated cular endothelial biology. Toxicol Pathol 1990; 4:572-582.
percutaneous implants. J Biomed Mater Res 1991; 85. Nerem RM, Levesque MJ, Cornhill JF. Vascular endothe-
25:387-405. lial morphology as an indicator of blood flow. ASME J
73. Wòjciak B, Crossan J, Curtis ASG. Grooved substrata fa- Biomech Eng 1981; 103:172-176.
cilitate in vitro healing of completely divided flexor ten- 86. Levesque MJ, Liepsch D, Moravec S et al. Correlation of
dons. J Mat Sci Mat in Med 1995; 6:266-271. endothelial cell shape and wall shear stress in a stenosed
74. Mooney DJ, Organ G, Vacanti JP et al. Design and fabri- dog aorta. Arteriosclerosis, 1986; 6:220-229.
cation of biodegradable polymer devices to engineer tu- 87. Diamond SL, Sharefkin JB, Dieffenbach C et al. Tissue
bular tissues. Cell Transplant 1994; 3:203-210. plasminogen activator messenger RNA levels increase in
75. Spargo BJ, Testoff MA, Nielson TB et al. Spatially con- cultured human endothelial cells exposed to laminar shear
trolled adhesion, spreading, and differentiation of endo- stress. J Cell Physiol 1990; 143:364-371.
thelial cells on self-assembled molecular monolayers. Proc 88. Mitsumata M, Fishel RS, Nerem RM et al. Fluid shear
Nat Acad Sci USA, 1994; 91:11070-11074. stress stimulates platelet-derived growth factor expression
76. Folkman J, Haudenschild C. Angiogenesis in vitro. Na- in endothelial cells. Am J Physiol Heart Circ Physiol 1993;
ture 1980; 288:551-556. 265:H3-H8.
77. Weinberg CB, Bell E. A blood vessel model constructed 89. Hermann IM. Endothelial cell matrices modulate smooth
from collagen and cultured vascular cells. Science 1986; muscle cell growth, contractile phenotype and sensitivity
231:397-400. to heparin. Heamostasis, 1990; 20:166-177.
78. Leenslag JW, Kroes MT, Pennings AJ et al. A compliant, 90. Harris-Hooker S, Sanford GL, Montgomery V et al. In-
biodegradable vascular graft: Basic aspects of its construc- fluence of low density lipoproteins on vascular smooth
tion and biological performance. New Polymeric Mater muscle cell growth and motility: Modulation by extracel-
1988; 1:111- 126. lular matrix. Cell Biol Int Rep 1992; 16:433-450.
79. Ziegler T, Nerem RM. Tissue engineering of a blood ves- 91. Chamley-Campbell JH, Campbell GR. What controls
sel: Regulation of vascular biology by mechanical stresses. smooth muscle phenotype. Arthereosclerosis, 1981;
J Cell Biochem 1994; 56:204-209. 40:347-357.
80. Nerem RM, Sambanis A. Tissue engineering: From biol- 92. Campbell JH, Campbell GR. Endothelial cell influences
ogy to biological substitutes. Tissue Engineering 1995; on smooth muscle phenotype. Annu Rev Physiol 1986;
1:3-12. 48:295-306.
Scaffold Engineering
Surface Modification
CHAPTER 49
Surface Bonding of Heparin
Patrick T. Cahalan
n the early 1960s Hufnagel began experiments to form “autogenized” vascular prostheses.1
I This was accomplished by implanting a Teflon rod containing a loosely woven Dacron or
polypropylene cloth surrounding it, and allowing 6-8 weeks for the formation of a collagen
tube. Early experiments involved implantation back into the same animal to avoid immu-
nogenic response. At the start of his work, important knowledge concerning glutaraldehyde
fixation of tissue was awaiting further publications.2 “Autogenized” was replaced with the
term “xenograft” due to parallel efforts by Sparks.3 Hufnagel stayed with his studies over the
next two decades and moved on to attempt heparin incorporation into mandril formed
prostheses as well as commercially available glutaraldehyde-treated human umbilical vein.
Rigorous experimental efforts, including radioactively tagged heparin, allowed him to de-
termine concentration of heparin in solution to effect maximal heparin loading conditions,
and to his surprise he found that continued washing with saline for two hours post “imbib-
ing” into the prosthesis did not significantly diminish the retained heparin, which was about
300 mg/ml. In implant studies the rate of early occlusion was significantly reduced, and
Hufnagel reported that the half life of the heparin was limited to a few days.
While the goal was to prevent thrombotic occlusion, in retrospect the most interesting
observation is the ability of heparin to bind and release from collagen in an unpredicted
manner. The hypothetical paradigms of the time with respect to interactions of biomolecules
and surfaces were still rather narrowly focused in simple ionic charge relationships.4 Pro-
teins (collagen) and polysaccharides (heparin) were widely known in the 70s by carbohy-
drate chemists to possess unique complexing capabilities that could be used to form
viscosifying agents, stabilizers, and precipitation agents in food applications. The synergistic
effect of these biomolecules could not be explained by simple ionic charge relationships.
The tertiary and quaternary structures of biomolecules play an important role in their in-
teractions with other molecules. While this concept is well known to anyone with an intro-
ductory education in biochemistry, it appears to be often forgotten in strategies for immo-
bilizing biomolecules on surfaces. There remain very active efforts at producing a synthetic
ionic analog heparinoid surface,5,6 while at the same time studies are showing how critical is
the dependence of heparin’s bioactivity on its unique structure.7
Early attempts at heparinizing surfaces used fatty quaternary ammonium compounds
as a base coating on polymers in order to present the positive charge of the quaternary
amine for ionic coupling of heparin.8 These early studies clearly showed an impact of hep-
arinized surfaces, but the heparin released rather quickly and was material dependent, sug-
gesting that the quaternary compound (generally considered toxic) also was releasing. In
attempts to make the heparin more stable, or release more slowly at the surface, different
solvent systems were use to attack the base material and entangle the fatty chains with the
polymer surface. Attempts were made to calculate the sustained release rate required to
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
546 Tissue Engineering of Prosthetic Vascular Grafts
maintain thromboresistant surfaces.9 This clearly showed lessons learned should be useful insights into attachment of
that it was impractical to sustain thromboresistance, due to other biomolecules, as cited by Hubbell in attachment of
the large loading required to maintain effectiveness for more peptides to surfaces.23
than a few days. At the same time, Larm published his re- Our group has been studying the performance of he-
sults for “end point” attached heparin on surfaces.10 This parinized surfaces together with the University of Maastricht,
was the first time that an immobilized heparin, with no leach- the Netherlands in attempts to understand the role of cou-
ing or releasing heparin, could demonstrate significant de- pling methodology in the performance of these surfaces in
activation of thrombin. The premise of Larm’s surface was human blood using numerous blood testing systems.24 Us-
that nitrous acid degraded heparin (NAD-Hep) containing ing a rotating disc apparatus as seen in Figure 49.1, it be-
high affinity pentasaccharides for ATIII was covalently im- came evident that deactivation of thrombin at a heparin-
mobilized on the surface by a terminal aldehyde group, thus ized surface is a diffusion limited reaction.
presenting an optimal conformation of the molecule to the The reactor was designed with baffles to assure redi-
blood interface. The clearest evidence of the effectiveness of rection of the flow perpendicular to the test surface, which
Larm’s premise is seen in controlled experiments using hu- was rotating, and thus assured a uniform shear rate profile
man blood in simulated cardiopulmonary bypass circuits on the sample surface. A mathematical model can be hy-
where improvement in platelet protection, platelet release pothesized for the thrombin decay if the rate of deactiva-
products, hemolysis, less white cell activation, and less con- tion is dependent on the diffusion to the surface, and if the
tact activation is demonstrated.11 The group of Wendel has concentrations of thrombin and ATIII added to the reactor,
tested several coatings in the latter system, and nothing to and the speed of the motor, are known. The control line in
date has been reported equal in performance to the co- the graph of Figure 49.1 represents a blank control surface,
valently attached heparin surface. and the limited decay of thrombin is explained by simple
The method of Larm, commercially known as the noncatalytic deactivation of thrombin by ATIII in solution.
Carmeda biologically active surface (CBAS), is in fact a co- The heparinized surface agrees fairly well with the theoreti-
valent attachment of heparin to an adsorbed base, and thus cal decay rate for a diffusion limited reaction. All surfaces
is not completely covalent to the base material. Since Larm’s that were heparinized were tested before the experiment was
invention several additional methods of surface modifica- started to assure that no heparin was releasing.
tion have been used to couple heparin and reported in the Our method of immobilizing heparin consisted of
literature.12-18 The traditional model for the mode of action covalent attachment of hydrogels that were derivatized with
of heparin on a surface is depicted by a heparin molecule amine functionality to which aldehyde-functionalized hep-
with a high affinity pentasaccharide tethered to a surface arin (NAD-Hep, or periodate oxidized) was coupled. By con-
and free to bind circulating ATIII, which it conformation- trolling the amount of amine functionality we were able to
ally alters to make it catalytic in its ability to bind and deac- prepare surfaces with different amounts of heparin coupled.
tivate circulating thrombin. Once this action has occurred, Regardless of how much heparin was coupled per cm2 on
the thrombin-antithrombin complex (TAT) is released, and the surface, or the measured bioactivity the surface possessed
the site is capable of repeating the activity. The relatively to deactivate thrombin, the observed behavior in Figure 49.1
high circulating concentrations of ATIII assists in the dis- was only seen at low concentrations of ATIII. At physiologi-
placement of the TAT complex from the surface.19 The ques- cal concentrations of ATIII (1 U/ml), there was no differ-
tion of protein adsorption and its effect on the heparin ac- ence in thrombin decay between heparinized and uncoated
tivity is a common one. Van Delden showed that the spacer controls. The rate of diffusion to, and deactivation at, the
composition was important for the surface retaining the surface was not as great as the noncatalyzed deactivation in
ability to deactivate thrombin after exposure to plasma.20 In solution with physiological concentrations of ATIII, even
his studies he showed that heparin immobilized via albu- with the most active surfaces. In addition to the latter re-
min-heparin conjugates retain 27% of initial activity after sults, ELISA measurements of TAT and F1+2 fragments from
exposure to blood, whereas heparin immobilized via PEO blood loop experiments showed that very few of the latter
spacers can retain as much as 55% of the initial activity. Most complexes were formed after exposure of the heparinized
interesting from these experiments was the fact that the to- surface to human whole blood in 90 minutes, as compared
tal amount of protein adsorbed onto heparinized surfaces to uncoated control polymers. These results question the
did not correlate with the retention of heparin activity. It model proposed wherein thrombin in flowing blood is de-
has been reported that albumin and fibrinogen adsorbed activated by the heparin-ATIII surface to form TAT com-
onto heparinized surfaces are not conformationally altered.21 plexes that are released, and the heparin repeats this activity
Earlier studies by Sevastianov suggest that it is critical to several times. Further studies in flowing systems developed
properly attach heparin if the proteins adsorbed are to be to study thromboresistance of intravascular stents, where
non platelet adhering and activating.22 During a panel dis- platelet free and platelet rich plasma were used, helped so-
cussion at the Biomaterials conference in New Orleans, May, lidify a refinement of the model (Figs. 49.2, 49.3, and 49.4).
1997, a comment was made to the effect that after all these Figure 49.2 represents the flow through system
years of research on blood-compatible surfaces we are no developed for monitoring thrombin generation for an in-
farther than the heparinized surface. Another perspective travascular stent. Citrated PRP or PFP could be pumped
might suggest that we have finally reached a point where we through the system at controlled flow rates and samples col-
understand how to properly attach heparin to a surface. The lected over time.
Surface Bonding of Heparin 547
Fig. 49.1. Rotating disc apparatus; thrombin decay in rotating disc test.
Figures 49.3A and 49.B represent a comparison be- Table 49.1 helps to further explain the importance of
tween heparin coated and uncoated intravascular stents with the quality (ability to bind ATIII) of the attached heparin to
respect to contact activation as measured with Factor IXa, the surface. ATIII uptake to the surface directly correlates
and its comparison to subsequent thrombin generation in with the delay in the onset of thrombin generation, the
PRP or PFP. The black points represent PRP and the gray amount produced at equilibrium and, most importantly, the
represent PFP. The first thing that can be seen is that contact number of adhered platelets. The platelets on the surface
activation is independent of platelets, and that without plate- are determined by the LDH method, and therefore some
lets the even more “thrombogenic” metallic surface does not platelets could be present that are “adhered”, but not acti-
produce significant amounts of thrombin (Fig. 49.3A). Not vated. SEM photos of the surfaces of the heparinized mate-
surprisingly, the anionic heparin stent does cause some rials did not reveal any visible platelets after mild rinsing,
contact activation, but less than the noncoated stent whereas the controls were covered with adhered and spread
(Fig. 49.3B). Clearly, in the presence of platelets the hep- platelets. Much might be learned in the future by study of
arinized surface delays the onset of thrombin generation, platelet adhesion to surfaces such as heparin and collagen.
and at equilibrium is much less thrombogenic than the Exposed subendothelial layers of collagen and GAGs can be
noncoated control material. seen to adhere platelets in dense single cell layers that do not
Surface Bonding of Heparin 549
Fig. 49.4. Contribution of immobilized heparin to clot prevention at blood-material interface by antithrombin (ATIII) deactiva-
tion of platelet-produced thrombin.
Table 49.1. Relation between the AT uptake and the Thrombogenicity of Wiktor stents
pmol/cm-2 nM Min
None 0.26±0.36 8.83±0.37 5 287±34
HepamedTM 1.26±0.36 4.98±0.62 20 135±18
HepamedTM 1.99±0.57 3.40±0.25 20 197±45
HepamedTM 7.28±0.21 1.75±0.09 30 42±20
HepamedTM 10.89±0.21 1.48±0.15 34 92±33
Table 49.2 Effects of heparin bonded surface on blood clotting in polyurethane tubing
appear to be spead or fully activated. This phenomenon has Heparin has several other properties in addition to
led us to further experiments that will be described shortly, acting as an anticoagulant, such as antibacterial and antivi-
but for now suffice it to say that the natural process of wound ral activity, inhibition of several enzymes, and the stimula-
healing of damaged endothelium begs the question as to tion of lipoprotein lipase release from the surface of endo-
whether or not a natural material such as collagen in the thelial cells.26,27 It has been proposed that heparin has a role
presence of certain glycosaminoglycans might produce a in the defense against pathogens outside of the immune sys-
“controlled” and important thrombogenic response. tem.28 This hypothesis seems to have merit based on the fact
Thrombin generation and subsequent fibrin forma- that heparin is predominantly located in organs and tissues
tion is due in a large part to the inability of flow conditions that come in direct contact with the outside environment
to keep the local concentration of thrombin low. Our expe- (lung, skin, and intestine).
rience has been that rapid clot formation takes place when Heparin binding growth factors (HBGFs) represent
the local concentration of thrombin reaches about 2-5 nM. an important family of mediators for the healing response,
Once this point has been reached, the fibrin network is rap- capable of inducing mesenchymal cell proliferation and dif-
idly formed and can entrap platelets and amplify the local ferentiation, tissue regeneration, morphogenesis, and
generation of thrombin. neovascularization.29 Several investigations of the growth
Figure 49.4 is a recent attempt at refining the concep- factors that are found in wound sites use heparin columns
tual model of how heparin works when immobilized on a in separation and identification of these growth factors.30-32
surface. The critical mechanism for preventing clotting is Numerous experiments have been tried by simple addition
that thrombin produced by platelets at the surface is imme- of growth factors to wound sites or to matrices. There are
diately deactivated, thus keeping the local concentration low, also some experiments where the role that heparin might
which in turn minimizes the strongest agent (thrombin) in play has been investigated. Angiogenesis was found to be
platelet activation. When the thrombin level is low, it can- enhanced by the addition of heparin to gels made of base-
not amplify its own production by activating vicinal plate- ment membrane extract that also had added acidic and ba-
lets, and less fibrin will minimize entrapment of additional sic fibroblast growth factors.33 Heparin, along with ECGF, is
platelets. The latter is of great significance for the longer term suggested to play a role as a response modifier of human
maintenance of low thrombogenicity, as thrombin can ad- endothelial cell migration, which may be relevant to tumor
here to fibrin and in this state it is not deactivated by hep- metastasis, wound healing, and atherogenesis.34 Another
arin and ATIII, and can continue to activate platelets.25 study using heparin analogs (chemically substituted dex-
Intramuscular implant studies of immobilized trans) in a collagen plaster was able to show a remarkable
collagen on biomaterials by our group showed immediate effect both on the kinetics and on the quality of the restored
thrombogenic response followed by strong apparent neu- skin.35 This study strongly suggests that endogenous growth
trophil recruitment at the tissue-material interface. This re- factors naturally releasing during the regeneration process
sponse quickly became quiescent, and after 6 weeks the could be trapped, protected and released by the addition of
tissue-material interface had only an occasional macroph- heparin analogs or heparin.
age with mostly fibroblasts (two cell layers) followed by It may be wishful thinking, but perhaps immobilized
adjacent microvascular structures. The control material or surface bonded heparin may play a strong role in tissue
(polyurethane) had a typical foreign body fibrotic response engineering. This may be particularly true if it is properly
with macrophages and foreign body giant cells adjacent to immobilized and capable of trapping, protecting and releas-
the majority of the surface, at least 15 layers of densely packed ing growth factors at locations and rates desired. Realiza-
fibroblasts with no vascular structures, and loose connective tion of this potential probably will require some additional
tissue outside of the fibroblast layers. These results led to thinking beyond heparin by itself. Coupling to biomolecules
preliminary experiments in measuring the coagulation such as collagens and other glycosaminoglycans to polymeric
response to collagen immobilized surfaces, and the surfaces may prove fruitful. From some of the studies men-
potentiating effect that heparin coupled to this collagen layer tioned, it is apparent that gel matrices can be modified to
might have. exhibit angiogenesis and remodeling. It may be entirely pos-
Using the same flow through system developed at the sible to fill porous polymer matrices with such gels and ap-
University of Maastricht and described previously, polyure- proach the ultimate regenerated tissue-like vascular graft.
thane tubings were prepared with immobilized collagen and Still, for those concerned with optimal mechanical proper-
immobilized collagen with heparin covalently bonded to the ties, the proper polymeric scaffold with a more fibrous char-
collagen (type I collagen). acter can have the surfaces modified for covalent attachment
Apparently the fact that more platelets are attached to that may achieve a similar end. The former will require a
the collagen and collagen plus heparin surfaces than the bigger step in regulatory and commercial acceptance, while
polyurethane does not lead to a faster clotting time, and in the latter may lead to incremental improvements. It is hope-
the case of the heparinized collagen the clotting time was ful that efforts in both areas will lead to understanding that
significantly delayed (Table 49.2). These are very prelimi- will result in a vascular graft with complete healing that is
nary experiments, and only suggest additional studies to at- long term patent, and infection resistant.
tempt to look at heparin in a broader context of the total
healing response.
Surface Bonding of Heparin 551
33. Passaniti A, Taylor RM, Pili R, Guo Y, Long PV, Haney dothelial cell growth factor and heparin. J Cell Biol 1985;
JA, Paul RR, Grant DS, Martin GR. A simple, quantita- 101(6):2330-4.
tive method for assessing angiogenesis and antiangiogenic 35. Meddahi A, Blanquaert F, Saffar JL, Colombier ML,
agents using reconstituted basement membrane, heparin, Caruelle JP, Josefonvicz J, Barritault D. New approaches
and fibroblast growth factor. Lab Inves 1992; 67(4):519-28. to tissue regeneration and repair. Path Res Prac 1994;
34. Terranova VP, DiFlorio R, Lyall RM, Hic S, Friesel R, 190(9-10):923-8.
Maciag T. Human endothelial cells are chemtactic to en-
Scaffold Engineering
Surface Modification
CHAPTER 50
Covalent Grafting of RGD Peptides to Synthetic Surfaces
Nina M.K. Lamba, S.L. Cooper
Introduction
F abric materials were first used as vascular prostheses in the 1950s, when Voorhees et al
implanted a polymeric vascular graft manufactured from vinyl chloride and acryloni-
trile.1 Since then, a number of polymers have been used to fabricate vascular prostheses, and
today, polyethylene terephthalate (PET, DacronΚ) and polytetrafluoroethylene (PTFE, Gore-
TexΚ) are the most commonly used biomaterials for this application. Vascular grafts made
from either of these materials have an internal diameter usually greater than 6 mm, and are
restricted to use in high shear environments. Currently there is no clinically acceptable syn-
thetic small diameter vascular prosthesis, i.e., one less than 6 mm I.D. Thrombus and
neointima formation will readily occlude synthetic vascular grafts. The resulting loss of pa-
tency remains the greatest obstacle to the development of a small caliber vascular prosthesis.
Thus, there is a need for an engineered material that will provide the necessary mechanical
and thromboresistant properties for a successful synthetic vascular prosthesis, to facilitate
the advancements in vascular surgery. In this chapter we will discuss the motivation for the
endothelialization of luminal surfaces, and describe some of the approaches that have been
taken to achieve this. A discussion of methods to covalently graft Arg-Gly-Asp (RGD) pep-
tides to polymers follows, focusing on methods and results obtained using polymers that are
used to fabricate vascular prostheses.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
554 Tissue Engineering of Prosthetic Vascular Grafts
technology to deposit reactive groups onto polymer surfaces, sequence in a specific protein, whereas other receptors may
or to indirectly influence the proteins adsorbed to the sur- recognize the RGD sequence in more than one protein. The
face. These methods to encourage cell attachment have met specificity of the RGD protein is believed to be modulated
with limited success, owing to the lack of specificity and poor by the conformation of the sequence. This may be deter-
control over protein orientation. Physical methods to im- mined by the amino acids that are immediately adjacent to
prove the development of a pseudointima on the surface the RGD sequence.12-14 Substitution of peptides within the
have involved altering the porosity and texture of surfaces, RGD sequence has been shown to produce a large reduc-
to promote tissue ingrowth into the graft. The neointima tion in the adhesivity of the peptide.15 The presence of a
that forms in the lumen of a graft may not provide compa- peptide adjacent to the sequence has been shown to alter
rable physiological properties of a natural endothelium, lim- the activity of the RGD sequence.10,16,17
iting the success of integration of the implanted device with Covalent grafting of biological molecules to a synthetic
the host by this method.5 Grafts have been coated with ad- substrate prevents the desorption of the agent over time from
hesive proteins or extracellular matrix components in order the surface. An excellent review of the chemistry of covalent
to promote attachment and proliferation of endothelial immobilization of RGD peptides is available.18 Generally,
cells.6-8 An improvement in the extent of endothelialization either the peptide or the surface must be “activated” to pro-
has been observed in animal models. RGD-protein conju- vide suitable sites for immobilization of the ligand to the
gates have also been synthesized,9 but problems in defining substrate. Biological structures are less resistant than syn-
the precise conformation of the protein once it has adsorbed thetic polymers to harsh chemical environments. Prolonged
to the substrate impede interpretation of results. The long exposure of a ligand to a harsh environment may lead to a
term success of materials modified through adsorption of reduction in the biological activity of the peptide. Thus, the
proteins to mediate cellular adhesion is not guaranteed, due polymer substrate is often derivatized or activated prior to
to possible desorption of the protein or proteolysis. peptide coupling. Some of the more commonly used reac-
More recently, there has been an interest in the graft- tion schemes involve the coupling of the peptide sequence
ing of synthetic peptide sequences onto polymeric substrates. to a polymer substrate that contains hydroxyl, thiol, carboxy-
By covalently grafting short peptide sequences onto poly- lic acid or amine groups. Surfaces or ligands can also be
meric substrates, it is believed that cellular adhesion can be derivatized and immobilized using photochemical tech-
mediated directly through receptor-ligand interactions, niques. The efficiency of the coupling reaction between the
rather than through a conditioning layer of adsorbed pro- substrate and the peptide can influence the surface peptide
tein. The covalent grafting of the Arg-Gly-Asp (RGD) pep- density, which has been shown to influence the degree of
tide sequence to synthetic materials has also been shown to cell spreading.19,20 There are also other factors that can af-
promote cell adhesion and attachment for applications such fect the coupling yield and biological activity of the ligand.
as tissue culture substrates and biomaterials for implanta- The RGD sequence can be immobilized by covalent bond-
tion and hybrid artificial organs. Some of the approaches to ing at either end of the sequence, either through the car-
modify biomaterials with cell adhesive peptide sequences boxyl terminus or the amino terminus. The orientation of
containing the RGD sequence relevant to the development the sequence can affect the interactions of the peptide with
of synthetic vascular grafts will be discussed in more detail cell surface receptors. The immobilization of peptide se-
in this chapter. quences on surfaces may also impose steric constraints on
the peptide, affecting the affinity and specificity of the ligand.
Immobilization of RGD Peptides This has been reported with the grafting of other biological
The RGD peptide sequence has been shown to be the molecules, and may be overcome by grafting the ligand onto
minimal cell-recognizable sequence in many adhesive plasma spacer arms, reducing steric hindrance imposed by the prox-
and extracellular matrix proteins.10 The RGD sequence was imity of the ligand to a rigid surface.21 The protection of the
first found in fibronectin, and is also present in vitronectin, peptide during the coupling reaction scheme is also a con-
von willebrand factor, fibrinogen, and collagen. The RGD sideration, to ensure that the activity of the peptide is re-
sequence has been shown to play a crucial role in mediating tained. Competing side reactions such as hydrolysis, poly-
cell attachment and subsequent spreading. It has been shown merization and crosslinking can also affect the grafting yield.
to be adhesive towards platelets and other cells, including The final surface density of ligands can be determined by
fibroblasts and endothelial cells. The RGD tripeptide may radiolableling, photochrome labeling, surface analysis or
also act as a ligand for the integrin superfamily of receptors. gravimetry.18
In particular, many integrin receptors recognize the RGD A number of studies have been performed investigat-
sequence which is present in many adhesive proteins.11 It ing methods to immobilize peptide sequences containing
has been demonstrated that the synthetic RGD peptide in the RGD sequence to polymeric surfaces. With respect to
solution is able to compete with adhesive proteins adsorbed the development of synthetic vascular prostheses, RGD
onto a surface for binding receptors on the cell in solution grafted materials have been studied with the goal of improv-
and prevent cellular adhesion to an adhesive protein ing the integration of the prosthesis into the host cardiovas-
adsorbed to the surface.10 Synthetic surfaces containing im- cular system, with the ability to perform the biological func-
mobilized RGD peptides have been shown to promote cell tions of the endothelium. RGD sequences have been grafted
attachment in a manner similar to fibronectin. Some cell onto polyethylene terephthalate (PET) and polytetra-
surface receptors have been shown to bind with the RGD fluoroethylene (PTFE),22 polyvinyl alcohol (PVA),23 poly-
Covalent Grafting of RGD Peptides to Synthetic Surfaces 555
acrylamide,24 polystyrene,25 and polyurethane.26 It has been Over the years, it has become apparent that the
demonstrated that the presence of RGD peptide sequences thrombogenicity of a synthetic graft is not the only crite-
at the surface increases endothelial cell attachment in vitro. rion by which a vascular graft should be assessed. The me-
The attachment of endothelial cells through specific recep- chanical properties, particularly the compliance and the tex-
tor-ligand interactions should overcome the problems of cell ture of the graft are important factors in promoting tissue
detachment under shear conditions.22 The methods to im- ingrowth, reducing anastomotic hyperplasia and favoring
mobilize RGD peptides to polymers, described below, dem- the long term patency of the graft. Both PET and PTFE are
onstrate that successful grafting can be achieved by either much less distensible when compared with the natural ar-
bulk or surface modification. The main advantages of using tery wall. Natural arteries show an increase in diameter of
surface modification techniques are that smaller quantities about 10% when pressurized to 150 mm Hg (normal arte-
of reagents are required, and the mechanical properties of rial pressure). In comparison, PET and PTFE grafts distend
the material can be retained. However, on a commercial scale, by about 1% under these conditions. Polyurethanes are one
bulk modification of a material requires fewer processing of the strongest candidates for the development of small
steps, which can ease fabrication and reduce costs. diameter vascular prostheses, due to their high tensile
Sugawara and Matsuda have derivatized peptides with strength, compliance and blood compatibility.29,30 Polyure-
4-azidobenzoyloxysuccinimide. The peptides were then thanes have been shown to distend by about 6% under nor-
adsorbed to a polyvinylalcohol surface.27 The surface was mal arterial pressure, 31 and a blended polyurethane-
exposed to UV radiation to covalently bond the peptides to polylactide vascular prosthesis possesses mechanical prop-
the surface. Bovine aortic endothelial cell adhesion was re- erties similar to rat abdominal aorta.32 Arterial substitutes
ported to be enhanced in a biologically specific manner. need to have mechanical properties that match the natural
Ozeki and Matsuda have used a similar strategy to attach vessel wall, to avoid turbulent blood flow, which may result
photochemically peptides containing the RGD sequence to in thrombus formation or destruction of formed blood ele-
polystyrene.25 An increase in the attachment of bovine aor- ments. Lyman et al33,34 have shown that the distensibility of
tic endothelial cells was reported. The specific peptide se- the graft can influence the biocompatibility through the
quence that is grafted may also modulate the adhesivity of degree of anastomotic hyperplasia by comparing vascular
the RGD sequence. Hirano et al16 immobilized RDGS, grafts fabricated from the same material, but with different
RGDV, RGDT, and RGD onto ethylene-acrylic acid copoly- degrees of compliance. Furthermore, a mismatch in com-
mer by coupling the terminal NH2 to the carboxylic resi- pliance between the synthetic graft and the natural vessel
dues in the polymer. The surfaces were evaluated for cell may traumatize the natural vessel and disrupt the endothe-
recognition by cell adhesion activity towards 5 different cell lium, which will in turn initiate thrombosis and stimulate
lines. It was found that the fourth amino acid in the intimal hypertrophy.
tetrapeptides played an important role in determining the Polyurethanes are block copolymers consisting of al-
precise specificity of the RGD sequence. The presence of ei- ternating soft and hard segments. The incompatibility be-
ther serine (S), threonine (T), or valine (V) at the end of the tween these two components of the polymer allows
peptide enhanced cellular adhesion. They postulated that microphase separation to occur within the bulk polymer.
the modulation in the adhesive nature of the RGD sequence The material can be described as being comprised of hard
may arise from a conformational change in the RGD se- domains dispersed within a soft segment matrix. This gives
quence, exerted by the terminal peptide. rise to the superior physical properties of the polyurethanes
Massia and Hubbell have grafted RGD peptides onto and is believed to be a contributory factor to their blood
the surface of polyethylene terephthalate (PET) and and tissue compatibility. Recently, RGD peptide sequences
polytetrafluoroethylene (PTFE) films.22,28 This was achieved have been successfully grafted to polyurethane substrates.35
through the hydroxylation of the polymer followed by sub- Polyurethanes for medical applications are usually synthe-
sequent immobilization of the synthetic peptide sequence sized via a two-step ‘prepolymer’ method. Lin et al synthe-
via tresyl chloride. The RGD could only react via the N-ter- sized a polyurethane from methylene bis (p-phenyl-
minus, ensuring that the orientation of the grafted peptide isocyanate) (MDI) and butane diol (BD), with polytetra-
was the same throughout. Radiolabeling of peptides was used methyleneoxide (PTMO) as the soft segment. Hydrogen at-
to confirm the presence and concentration of the peptide oms on the urethane groups were substituted with carboxyl
sequence. Endothelial cell adhesion was evaluated on the groups, via abstraction of urethane hydrogen by sodium
unmodified, hydroxylated and peptide containing surfaces, hydride, followed by reaction of the polyurethane with
both with and without the presence of serum. In the ab- !-propiolactone. The degree of substitution could be varied
sence of serum, the RGD grafted films were able to support by the quantities of reagents used. This was performed to
the adhesion and spreading of human umbilical vein en- alter the density of the grafted peptides. Peptide sequences
dothelial cells (HUVECs). Neither the unmodified films nor were then coupled to the carboxyl group using 1-(3-
the hydroxylated intermediates showed the same degree of dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
adhesion and spreading. Stress fibers were also observed, (EDCl). The reaction scheme for the carboxylation of the
implying that the cells were attached strongly to the sub- urethane group, followed by coupling of the RGD peptide
strate, and may possess the mechanical strength to resist sequence is shown in Figure 50.1.
detachment by shear forces. Lin investigated the effect of the coupling reaction on
the endothelialization of the substrate. Two coupling
556 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 50.1. Reaction scheme for the synthesis of RGD-containing peptide grafted polyurethane. (a) deprotonation of urethane
group; (b) carboxylation of polyurethane; (c) coupling of RGD peptide.26
methods were used. The first, one step method coupled the where.25 The increased adhesion on the RGD-grafted
free hexapeptide directly to the carboxyl group through the polyurethanes that were hydrated prior to contact with
formation of an amide bond. The second method involved endothelial cells provides further evidence that reorienta-
two steps, coupling a protected peptide to the carboxyl group, tion of the RGD peptides does occur on hydration, and that
followed by deprotection of the peptide. The two step cellular adhesion to RGD grafted materials is mediated
method using the protected peptides appeared to have a through receptor interactions with the peptide. Figure 50.3
higher coupling efficiency of the peptide to the substrate of shows SEMs of endothelial cells grown in vitro, on the base
the unprotected peptide, as coupling could only occur via polyurethane (PEU), carboxylated polyurethane
the N-terminus of the peptide. Furthermore, there were (PEU-COOH), and peptide grafted polyurethanes (PEU-
fewer competing side reactions in the synthetic scheme us- GRGESY, PEU-GRGDSY, and PEU-GRGDVY). The effect
ing the protected peptides, improving the yield of grafting. of substituting one of the amino acids within the RGD se-
Previously, other researchers had reported that the quence reduces the adhesivity of the peptide (PEU-
adhesion and growth of endothelial cells on polyurethane is GRGESY). The substitution of the peptide adjacent to the
relatively poor.36,37 The immobilization of RGD peptides to RGD sequence has the ability to alter the adhesivity, as
a polyurethane as described above increased the number of adhesion was greater on GRGDVY grafted material than
adherent HUVECs after in vitro seeding. A larger number GRGDSY grafted polyurethane.
of cells attached to RGD peptides that had been protected Both Massia22 and Lin35 have investigated the role of
during synthesis than those that had not been protected. In serum proteins in mediating cell adhesion to RGD grafted
addition, greater endothelial cell adhesion to hydrated materials. Endothelial cell adhesion was evaluated on the
samples was observed compared with samples that had not unmodified, intermediate and peptide grafted materials,
been previously hydrated. Polyurethanes contain hydrophilic both with and without the presence of serum. Massia and
and hydrophobic domains that reorient upon hydration, Hubbell found that both the unmodified PET and the hy-
minimizing the interfacial free energy. Cold-stage ESCA droxylated PTFE supported cell adhesion only in the pres-
studies of hydrated and dry samples showed that reorienta- ence of serum proteins, implying that cell adhesion was
tion of the peptides occurs at the hydrated surface,38 lead- mediated by adsorbed proteins. Lin et al35 reached a similar
ing to the conclusion that the RGD peptides on the polyure- conclusion in their study of HUVEC attachment to the RGD
thane backbone do orient towards the surface on hydration. grafted polyurethane. Hubbell and co-workers have also cre-
This reorientation of the functional peptide groups is de- ated highly specific cell adhesive substrates of RGD-contain-
picted schematically in Figure 50.2.38 Reorientation of RGD ing surfaces with low protein adsorption.15,22,39 The low pro-
peptides at an aqueous interface has been reported else- tein adsorbing characteristics of these surfaces has allowed
Covalent Grafting of RGD Peptides to Synthetic Surfaces 557
Fig. 50.2. Schematic of the surface re-orientation of the peptide grafted polyurethanes in the hydrated
and dehydrated states. From: Lin H-B, Lewis KB, Leach-Scampavia D et al. Surface properties of RGD-
peptide grafted polyurethane block copolymers: Variable take-off angle and cold-stage ESCA studies.
J Biomater Sci, Polym Ed 1993; 4:183-198.
558 Tissue Engineering of Prosthetic Vascular Grafts
the study of cell attachment and spreading independently ronment, the physiological properties of the endothelial layer,
of protein adsorption. Using these substrates they have and the interaction of endothelial cells with the formed ele-
shown that the RGD and Tyr-Ile-Gly-Ser-Arg (YIGSR) pep- ments of the blood, such as platelets and white blood cells.
tide sequences promote endothelial cell adhesion indepen-
dently of adsorbed cell adhesion proteins. They have also References
demonstrated that serum proteins play a role in cell spread- 1. Voorhees AB, Jaretzki A, Blakemore AH. The use of tubes
ing, with serum proteins affecting the final extent of spread- constructed from Vinyon N cloth in bridging arterial de-
ing, but not the initial rate.40 The effect of the surface den- fects. Ann Surg 1952; 135:322-324.
sity of the grafted peptide has also been investigated using 2. Szycher M. Thrombosis, hemostasis and thrombolysis at
prosthetic interfaces. In: Szycher M, ed. Biocompatible
these surfaces. The effect of ligand surface density on cell
Polymers, Metals and Composites. Lancaster, PA:
adhesion is discussed elsewhere in this book. Ultimately, it Technomic, 1983:1-33.
is hoped that optimization of the spacing of RGD peptides 3. Brash JL. Role of plasma protein adsorption in the re-
to promote the attachment of endothelial cells will allow sponse of blood to foreign surfaces. In: Sharma CP,
the inclusion of other ligands, such as growth factors, that Szycher M, ed. Blood compatible materials and devices.
may be necessary to further modulate adhesion and spread- Lancaster, PA: Technomic, 1991:3-24.
ing, or otherwise maintain the cell layer. 4. Forbes CD, Courtney JM. Thrombosis and artificial sur-
faces. In: Bloom AL, Forbes CD, Thomas AL, Tuddenham
Summary EGD, eds. Haemostasis and Thrombosis. London:
The natural endothelium offers the only Churchill Livingstone, 1993:1301-1324.
nonthrombogenic surface known to man. This is actively 5. Zilla P, Fasol R, Grimm M, Fischlein T, Eberl T, Preiss P,
Krupicka O, Oppell U, Deutsch M. Growth properties of
achieved through regulation of transport across the endo-
cultured human endothelial cells on differently coated
thelium, and the synthesis of agents that actively prevent artificial heart materials. J Thorac Cardiovasc Surg 1991;
events such as protein adsorption, platelet deposition and 101:671-680.
thrombus formation. The development of a successful small 6. Lee Y, Park DK, Kim YB, Seo JW, Lee KB, Min B. En-
diameter vascular prosthesis is central to the advancement dothelial cell seeding onto the extracellular matrix of fi-
of peripheral vascular surgery. The issues of occlusion and broblasts for the development of a small diameter poly-
infection of vascular grafts are problematic, and remain the urethane vessel. ASAIO J 1993; 39:M740-M745.
greatest impediment to the development of small caliber 7. Miwa H, Matsuda T, Tani N, Kondo K, Iida F. An in
grafts. The immobilization of RGD peptides to the luminal vitro endothelialized compliant vascular graft minimizes
surfaces of synthetic vascular prostheses may offer the means anastomotic hyperplasia. ASAIO J 1993; 39:M501-M505.
8. Stansby G, Berwanger C, Shukla N, Schmidt-Rixen T,
to promote endothelialization, and improve the long term
Hamilton G. Endothelial seeding of compliant polyure-
patency of vascular grafts. Further studies are required to thane vascular graft material. Brit J Surg 1994; 81:1286-
establish the long term clinical performance of such devices. 1289.
Studies need to include examination of endothelial cell re-
tention once the material is exposed to a high shear envi-
Covalent Grafting of RGD Peptides to Synthetic Surfaces 559
9. Kishida A, Takatsuka M, Matsuda T. RGD-albumin con- ognizable peptidyl side chain. Trans Am Soc Artif Intern
jugate: Expression of tissue regeneration activity. Organs 1990; 36:M294-M296.
Biomaterials, 1992; 13:924-930. 26. Lin H, Zhao Z, Garcia-Echeverria C, Rich DH, Cooper
10. Pierschbacher MD, Ruoslahti E. Cell attachment activity SL. Synthesis of a novel polyurethane copolymer contain-
of fibronectin can be duplicated by small synthetic frag- ing covalently attached RGD peptide. J Biomater Sci Poly
ments of the molecule. Nature 1984; 309:30-34. Ed 1992; 3:217-227.
11. Hynes RO. Integrins: A family of cell surface receptors. 27. Sugawara T. and Matsuda T. Photochemical surface
Cell 1987; 48:549-559. derivatization of a peptide containing Arg-Gly-Asp (RGD).
12. Ruoslahti E, Pierschbacher MD. New perspectives in cell J Biomed Mater Res 1995; 29:1047-1052.
adhesion: RGD and integrins. Science 1987; 238:491-497. 28. Hubbell JA, Massia SP, Drumheller PD. Surface-grafted
13. D’Souza SE, Ginsberg MH, Plow EF. Arginyl-glycyl-aspar- cell-binding peptides in tissue engineering of the vascular
tic acid (RGD): A cell adhesion motif. Trends Biol Sci graft. In: Pedersen H, Matharasan R, DiBiasio D, eds.
1991; 16:246-250. Annals of the New York Academy of Science 1992:253-
14. Hautanen A, Gailit J, Mann M, Ruoslahti E. Effects of 258.
modification of the RGD sequence and its context on rec- 29. Zdrahala RJ. Small caliber vascular grafts. Part II. Poly-
ognition by the fibronectin receptor. J Biol Chem 1989; urethanes revisited. J Biomater Appl 1996; 11:37-61.
264:1437-1492. 30. Lamba NMK, Woodhouse KA, Cooper SL. Polyurethanes
15. Drumheller PD, Hubbell JA. Polymer networks with in Biomedical Applications. Boca Raton, FL: CRC Press
grafted cell adhesion peptides for highly specific cell ad- 1998:277.
hesive substrates. Anal Biochem 1994; 222:380-388. 31. Annis D, Bornat A, Edwards RO, Higham A, Loveday B,
16. Hirano Y, Okuna M, Hayashi T, Goto K, Nakajima A. Wilson J. An elastomeric vascular prosthesis. Trans Am
Cell-attachment activities of surface immobilized Soc Artif Intern Organs 1978; 24:209-214.
oligopeptides RGD, RGDS, RGDV, RGDT, and YIGSR 32. Gogolewski S, Pennings AJ, Lommen E, Wildevuur CRH,
towards five cell lines. J Biomater Sci Polym Ed 1993; Niewenhuis P. Growth of a neo artery induced by a bio-
4:235-243. degradable polymeric vascular prosthesis. Makromol
17. Lin H-B, Garcia-Echeverria C, Asakura S, Sun W, Mosher Chem Rapid Commun 1983; 4:213-219.
DF, Cooper SL. Endothelial cell adhesion on polyure- 33. Lyman DJ, Alb D, Jackson R, Knutson K. Development
thanes containing covalently attached RGD peptides. of small diameter vascular graft prostheses. Trans Am Soc
Biomaterials 1992; 13:905-914. Artif Intern Organs 1977; 23:253-256.
18. Drumheller PD, Hubbell JA. Surface immobilization of 34. Lyman DJ, Fazzio FJ, Voorhes H, Robinson G, Albo D.
adhesion ligands for investigations of cell-substrate inter- Compliance as a factor affecting the patency of a
actions. In: Bronzino JD, ed. The Biomedical Engineer- copolyurethane vascular graft. J Biomed Mater Res 1978;
ing Handbook. Boca Raton, FL: CRC Press and IEEE 12:337-345.
Press, 1995:1583-1596. 35. Lin H-B, Sun W, Mosher DF, Garcia-Echeverria C,
19. Massia SP, Hubbell JA. An RGD spacing of 440 nm is Schaufelberger K, Lelkes PI, Cooper SL. Synthesis, sur-
sufficient for integrin #v!3%mediated fibroblast spreading face and cell adhesion properties of polyurethanes con-
and 140 nm for focal contact and stress fiber formation. taining covalently grafted RGD-peptides. J Biomed Mater
J Cell Biol 1991; 114:1089-1100. Res 1994; 28:329-342.
20. Goodman SL, Cooper SL, Albrecht RM. Integrin recep- 36. Gospardarowicz D, Ill C. Extracellular matrix and con-
tors and platelet adhesion to synthetic surfaces. J Biomed trol of proliferation of vascular endothelial cells. J Clin
Mater Res 1993; 27:683-695. Inv 1981; 68:1351-1364.
21. Nojiri C, Okano T, Park KD, Kim SW. Suppression 37. Nichols NK, Gospardarowicz D, Kellser TR, Oslen DB.
mechanisms for thrombus formation on heparin-immo- Increased adherence of vascular endothelial cells to Biomer
bilized segmented polyurethane-ureas. Trans Am Soc Artif precoated with extracellular matrix. Trans Am Soc Artif
Intern Organs 1988; 34:386-398. Intern Organs 1981; 27:208-211.
22. Massia SP, Hubbell JA. Human endothelial cell interac- 38. Lin H-B, Lewis KB, Leach-Scampavia D, Ratner BD, Coo-
tions with surface coupled adhesion peptides on a non- per SL. Surface properties of RGD-peptide grafted poly-
adhesive glass substrate and two polymeric biomaterials. urethane block copolymers: Variable take-off angle and
J Biomed Mater Res 1991; 25:223-242. cold-stage ESCA studies. J Biomater Sci Polym Ed 1993;
23. Matsuda T, Kondo A, Makino K, Akutsu T. Development 4:183-198.
of a novel artificial matrix with cell adhesion peptide for 39. Drumheller PD, Elbert DL, Hubbell JA. Multifunctional
cell culture and artificial hybrid organs. Trans Am Soc poly(ethylene glycol) semiinterpenetrating polymer net-
Artif Intern Organs 1989; 35:677-679. works as highly selective adhesive substrates for
24. Brandley BK, Schnaar RL. Covalent attachment of an Arg- bioadhesive peptide grafting. Biotech Bioeng 1994;
Gly-Asp sequence peptide to derivatizable polyacrylamide 43:772-780.
surface: Support of fibroblast adhesion and long-term 40. Massia SP, Hubbell JA. Covalent surface immobilization
growth. Anal Biochem 1988; 172:270-278. of Arg-Gly-Asp and Tyr-Ile-Gly-Ser-Arg containing pep-
25. Ozeki E, Matsuda T. Development of an artificial extra- tides to obtain well-defined cell adhesive substrates. Anal
cellular matrix. Solution castable polymers with cell rec- Biochem 1990; 17:292-301.
Matrix Engineering
CHAPTER 51
Hydrogels in Biological Control During Graft Healing
Jeffrey A. Hubbell
Introduction—Why Hydrogels?
H ydrogels are polymeric materials that imbibe a large fraction of water and yet remain
intact, not dissolving even given an infinite period of time. These materials are formed
from polymer chains that have a high affinity for water, either such that the chains would be
individually soluble in water and are restricted from dissolving by virtue of participation in
the polymeric material as a network, or such that the chains are almost, but not quite, soluble
in water. By network, it is meant that the polymer chains in the hydrogel are somehow inter-
acting with each other so as to keep the individual chains from diffusing away into the aque-
ous milieu, either by virtue of being covalently bonded together or by interacting physi-
cally—specific examples will be provided in the section on “Hydrogel Structure and Synthesis”.
Many extracellular structures in the body can be considered as hydrogels. The extra-
cellular matrix of soft tissues and cartilage, for example, exists as a network of glycoproteins
and proteoglycans that both interact with each other biophysically (e.g., by specific biologi-
cal interactions between collagen and a variety of adhesion proteins,1 by specific biological
interactions between glycosaminoglycans in proteoglycans and a variety of adhesion pro-
teins,2 and by hydrogen bonding as in collagen due to a very high content of hydroxypro-
line3) and by covalent interactions (e.g., chemical crosslinking between glutamine residues
on one protein and lysine residues on a neighboring protein under the enzymatic action of
a transglutaminase to catalyze the formation of an amide linkage between the two).4
It is not by coincidence that hydrogels are found extensively in Nature: They possess
distinct biologically useful features, which will likewise be useful in tissue engineering of the
vascular graft and other tissues. Hydrogels are mechanically flexible, are freely permeable to
small molecules such as dissolved gases and low molecular weight nutrients and wastes, and
are controllably permeable to larger molecules such as proteins. Because the gels are highly
swollen with water, the amount of polymer mass per unit volume is relatively low, and this is
of key importance in cell migration in three dimensions. Cell migration is facilitated by
enzymatic remodeling of the three dimensional extracellular matrix, and the amount of
protein and proteoglycan mass to be degraded and moved out of the path of the cell is much
lower in the case of a hydrogel than if the tissue had consisted of less water. Indeed, in cul-
ture models of leukocyte migration through collagen-based gels, cell migration speed was
observed to be reduced by nearly an order of magnitude when the collagen concentration
was doubled.5 Also, because gels are highly swollen with water, their effective surface area is
very high, and this is used in Nature to store large quantities of biologically active molecules
that can be released in a triggerable fashion from the extracellular matrix, e.g., the release of
heparin-binding polypeptide growth factors from heparan sulfate glycosaminoglycan
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
562 Tissue Engineering of Prosthetic Vascular Grafts
components of the proteoglycans in the extracellular ma- trol on shape retention, by freezing the poly(vinyl alcohol)
trix. As engineered hydrogels are developed for specific ap- solution. When ice crystals form, the polymer chains are
plications in tissue engineering, it should be possible to excluded from the water crystal and pushed to the crystal
mimic these biological interactions to achieve these same boundaries, resulting in very highly concentrated regions of
sorts of biological interactions.6-9 polymer. At these highly concentrated domains, when all of
Hydrogels have other potential advantages that are less the water is frozen, segments of the poly(vinyl alcohol) chains
extensively used in Nature. As is addressed in more detail in crystallize due to hydrogen bonding, resulting in an insoluble
the section “Protein and Cell Interactions with Hydrogels”, (without heating far above 37°C) hydrogel in the shape of
there exists the potential to design gels without the the polymer solution that was frozen. Thus, this gel is a net-
incorporation of binding sites for proteins, with the result work polymer where the bonding between polymer chains
being a gel that is highly resistant to cell adhesion. Such is physical by hydrogen bonding.
materials may be useful in preventing the interaction of cells Hydrophobic interactions can also be important in the
with material substrates in a scaffold for tissue engineering formation of hydrogels, and block copolymers of
at sites of application of a gel coating. An additional feature poly(ethylene glycol) and poly(propylene glycol) form a
of hydrogels of potential tissue engineering advantage is that characteristic example.13 Poly(ethylene glycol) is a water-
a solvent for the gel precursors is water; as such, the soluble poly(ether), while poly(propylene glycol) is water-
possibility exists to apply a hydrogel precursor to a tissue insoluble, containing a nonpolar methyl group that the
site, dissolved in physiological saline, and to form the poly(ethylene glycol) repeat unit lacks. Block copolymers of
hydrogel in situ, as is addressed below in the section “In Situ these two compositions in the form of a domain of the hy-
Transformations”. drophobic poly(propylene glycol) flanked on both sides by
domains of poly(ethylene glycol) are water soluble polymers
that, at relatively high concentrations, can form gels by
Hydrogel Structure and Synthesis hydrophobic interactions between the poly(propylene gly-
Hydrogels consist of hydrophilic polymer chains that col) domains. This is to say, the poly(propylene glycol) do-
are held together in a single mass by virtue of either slight mains from several different polymer chains cluster to shield
insolubility or some form of bonding between chains to yield each other from interactions from water, thereby forming
a polymer network.10 The structure of the hydrogel is highly physical crosslinks (within these clusters) based on hydro-
dependent upon the nature of the synthetic procedure that phobic interactions, resulting in a hydrogel. This gel, as do
was used to obtain the gel; for this reason, different means several others based on hydrophobic interactions, has the
of hydrogel synthesis will be illustrated in the paragraphs useful feature that the aqueous mixture is a liquid at cold
below with a few examples. temperatures (below approximately 10°C) but rapidly forms
The nature of some hydrogels is controlled by the slight a hydrogel at 37°C.
insolubility of the polymer chains; a typical example is Electrostatic interactions also play important roles in
hydrogels of poly(hydroxyethylmethacrylate).11 The vinyl hydrogel formation, and gels of alginate with calcium ion14
monomer for this polymer, hydroxyethylmethacrylate, is and polyelectrolyte complexes of alginate and poly(lysine)15
soluble in water. Likewise, oligomers and low molecular form good examples of these principles. Alginate is a natu-
weight polymers of hydroxyethylmethacrylate are soluble in rally occurring polysaccharide product of kelp and several
water, but high molecular weight polymers are slightly bacterial strains. Each sugar residue in the polysaccharide
insoluble in water, leading to a gel that swells to imbibe chain bears a carboxyl group, so the polysaccharide chain is
approximately 50% water by mass. This swelling extent can highly anionic, and these charges can play multiple roles in
be readily controlled by copolymerization with more gel formation. Firstly, divalent cations, such as Ca2+, can form
hydrophobic monomers, such as ethylmethacrylate (i.e., the a bridge between the anionic groups on adjacent alginate
analogous monomer to hydroxyethylmethacrylate, only backbones, resulting in regions of adjacent chains that are
lacking the polar hydroxyl group), to achieve a series of paired, these regions being connected to other paired re-
polymers with swelling ranging from almost nil (for gions by amorphous domains. Practically, dropping the al-
poly(ethylmethacrylate)) to that of poly(hydroxyethyl- ginate solution in Ca2+-free saline into a saline solution con-
methacrylate). These gels have little internal structure, i.e., taining greater than 1 mM Ca2+ results in the rapid forma-
no permanent pores or dense regions, but rather exist as tion of a hydrogel which is stable under physiological con-
dynamic structures with small pores opening and closing in ditions. Secondly, alginate (either solution or as a Ca2+-
equilibrium. crosslinked gel) can be mixed with a polycation in solution,
Hydrogels can also be formed by hydrogen bonding, such as poly(lysine), and these two polymer chains form a
and gels formed from poly(vinyl alcohol) represent a typi- hydrogel, referred to as a polyelectrolyte complex, crosslinked
cal example.12 Poly(vinyl alcohol) is a water-soluble poly- into a network by the electrostatic interactions between the
mer at 37°C. When a solution of poly(vinyl alcohol) is dried oppositely charged polymers. Gels formed in either manner
to form a cast solid, the solid does not dissolve in water when described above can have interesting microscopic structures
it is again placed in an aqueous milieu. In the dry state, re- that are lacking in the other gels described in the paragraphs
gions of the poly(vinyl alcohol) solid crystallize due to hy- above, this microscopic structure deriving from the diffu-
drogen bonding. These regions are so strongly bonded that sive process of gel formation. For example, when the algi-
dissolution of the polymer again at 37°C is infinitesimally nate solution is dropped into a Ca2+-containing bath, Ca2+
slow. This behavior can also be obtained, with greater con- ions begin diffusing into the polymer solution, causing the
Hydrogels in Biological Control During Graft Healing 563
formation of the gel at the interface between the polymer bin cleaves the fibrinogen protein at two points, exposing a
solution and the surrounding Ca2+-containing bath. Because new terminus on two polypeptide chains. This new termi-
the gel region exists at a lower soluble polymer concentra- nus physically binds at a site on other fibrinogen (and cleaved
tion than does the alginate solution, alginate from the core fibrinogen) proteins, this process resulting in a physically
of the droplet diffuses toward the gelled interface; this poly- bonded hydrogel. The coagulation protein factor XIII is also
mer then gels, and more polymer diffuses from the core to cleaved by thrombin to form the active transglutaminase fac-
the periphery, and so on: The net result of this phenom- tor xiiia, which acts to catalyze the formation of an amide
enon is that the periphery of the Ca2+-alginate gel is more bond between glutamine residues and lysine residues on
dense than the core of the particle, owing to the diffusive opposing polypeptide domains in fibrin, resulting in a
nature of the process. The case is likewise with the incorpo- chemically crosslinked fibrin network.
ration of the polyelectrolyte counter-ion poly(lysine). The brief overview provided above should provide the
Poly(lysine) diffuses into the gel particle from the periphery reader with a background understanding of the physical
and interacts first at that periphery, forming denser mem- nature of hydrogels and the vast flexibility in the physical
brane at the periphery, which restricts further poly(lysine) and chemical processes for forming them. Based upon this
diffusion into the core in a self-limiting process. Accordingly, brief background, the reader is well positioned to consider
a thin shell of polyelectrolyte complex can be formed, a mi- the application of hydrogels for tissue engineering of the
crostructure that could be put to good use in tissue engi- vascular graft.
neering.
In addition to the physical interactions described In Situ Transformations
above, hydrogels can also be formed that depend on cova- One interesting advantage in the use of hydrogels in
lent bonding between polymer chains; these gels have a tissue engineering is that they may be delivered in the form
characteristic advantage over those described above in that of a precursor that is dissolved in a physiological saline and
they can be considerably more stable under physiological is then converted into the insoluble hydrogel upon the tar-
conditions. Three examples are presented below, two based get tissue (see Fig. 51.2), in some cases even in direct con-
on synthetic polymers with nonbiological reactions and one tact with cells.19 To perform adequately in such an environ-
based on proteins with enzymatic reactions. The simplest ment, the precursor solution should be lacking in high con-
way to form a covalently crosslinked hydrogel is to centrations of toxic species and low molecular weight mono-
polymerize an otherwise water-soluble polymer in the mers, which may be more likely to cross the cell membrane
presence of a multifunctional crosslinker.16 As an example, than a high molecular weight macromonomer, and the gel
acrylamide is a monomer that polymerizes to form a conversion should occur at physiologically acceptable tem-
water-soluble polymer. If acrylamide is polymerized in the peratures. This has been accomplished with aqueous solu-
presence of a difunctional monomer (i.e., one that has two tions of poly(ethylene glycol) diacrylate and related com-
sites for polymerization, then referred to as a crosslinker), pounds, which may be converted to a hydrogel under
that monomer will participate in two poly(acrylamide) biocompatible reaction conditions by exposure to visible
chains. Accordingly, bis-acrylamido crosslinkers are routinely light in the presence of a visible light sensitizer.20 The
used to crosslink acrylamide hydrogels. Given that each biocompatibility of this process in direct contact with cells
poly(acrylamide) chain may react with more than one can be attributed to the following features:
crosslinker molecule, this polymerization process results in 1. Physiological saline is the solvent;
the formation of a chemically crosslinked hydrogel. In this 2. A very high fraction of the polymerizable precur-
case, the entire macroscopic hydrogel object is a single sors are macromolecular and thus unable to pass the
molecule. cell membrane;
As a second example of the formation of covalently 3. The photoinitiation process does not involve short
crosslinked synthetic hydrogels, hydrogels formed from wa- wavelength ultraviolet light;
ter-soluble polymeric diacrylates form a good example.17 4. The photoinitiation process does not generate large
Poly(ethylene glycol) diacrylates (Fig. 51.1)have been formed amounts of reactive oxygen species; and
by coupling one acrylate moiety to each end of a 5. The polymerization process does not generate large
poly(ethylene glycol) chain. These acrylates are capable of amounts of heat.
polymerizing to form a poly(acrylate), and if a poly(ethylene These principles serve as a design basis for other bio-
glycol) monoacrylate were polymerized the resulting poly- material systems for the formation of hydrogels in situ by
mers would be a comb-like structure, a poly(acrylate) struc- transformation from liquid precursors.
ture forming the back of the comb and poly(ethylene gly- In situ transformation has at least two major advan-
col) chains forming the teeth. If, however, a poly(ethylene tages, for some applications, vs. the implantation of
glycol) diacrylate is polymerized, the acrylates on either end preformed hydrogels. One advantage is that a large hydro-
of the chain can enter into different poly(acrylate) chains, gel implant may be delivered through a very small surgical
resulting in a chemically crosslinked gel that can only be hole, somewhat like assembly of the model ship inside the
broken apart by the cleavage of a covalent bond. bottle. This has been put toward therapeutic development
It is also possible for form covalently crosslinked in the coating of organs in the abdominopelvic cavity
hydrogels using enzymatic crosslinking, and fibrin forms an following surgery to prevent the formation of postoperative
excellent case in point.18 Fibrin is formed by the enzymatic adhesions, engineering the healing response.21 Here is it
action of thrombin and factor xiiia on fibrinogen. Throm- possible to deliver biomaterial implants as organ coatings
564 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 51.1. As an example of designing various mechanisms of degradation and bioactivity of hydrogels, the case of gels formed by
crosslinking poly(ethylene glycol) diacrylates is considered. Gels can be made to be substantially non-degradable (a), to incorpo-
rate nonenzymatic degradation sites by hydrolysis (b), to incorporate oligopeptide domains (c) which could be sites for cleavage by
a protease, e.g. plasmin (d) or which could be sites for binding to adhesion receptors on a targeted cell (e). In each case in this
example, the central block of the macromer is a poly(ethylene glycol) chain, selected for resistance to protein adsorption and for
favorable toxicology of the degradation product (f). These diacrylated poly(ethylene glycol) chains polymerize by free radical
reaction at the acrylate to form oligomers of acrylic acid that are linked to other oligomers of acrylic acid via the poly(ethylene
glycol) chains with the relevant degradation sites (g). Thus, the acrylic acid oligomers form nodes in a three dimensional network,
each connected to other nodes by long strings of poly(ethylene glycol). The permeability of the gels to proteins, and the rate of
release of incorporated protein drugs, can be controlled via manipulation of the density of the oligomeric acrylic acid nodes, by the
number of poly(ethylene glycol) strings emanating therefrom, and by the length of the poly(ethylene glycol) strings. Degradation
products are poly(ethylene glycol) and oligomers of acrylic acid, which are eliminated by glomerular filtration, and lactic acid.
that are many centimeters in diameter, but to deliver these teries, to attempt to engineer a healing response, the ability
implants using laparoscopic instruments to convert the liq- to form gels in situ presents an attractive feature. Such trans-
uid precursor to a hydrogel. A second advantage is that hy- formations can be accomplished even in the presence of in-
drogel implants of very complex shape may be formed in dividual and aggregated cells,24 suggesting that such chemi-
situ; this has been put to advantage in the coating of arterial cal schemes can be adapted for the delivery of cellular thera-
surfaces after deendothelialization to prevent the deposition peutics either within or adjacent to the vascular system.
of platelets on the subendothelium.22,23 In this case, the
photoinitiator was separated from the other components of Protein and Cell Interactions with Hydrogels
the precursor solution and was adsorbed to the arterial sur- Hydrogels can be designed to have minimal interac-
face; when the arterial lumen was flooded with all of the tions with proteins and cells. Cell adhesion to biomaterials
remaining precursors and the artery was irradiated with vis- is mediated by an adsorbed protein overlayer.25 Protein ad-
ible light, the formation of the hydrogel was restricted to sorption to materials is dominated primarily by hydropho-
the interface between the arterial wall and its contents. Thus, bic interactions and secondarily by electrostatic interac-
a hydrogel barrier was formed, only several microns thick, tions. 26 Most insoluble polymers (e.g., poly(ester)s,
that was conformal with the complex shape of the poly(ethylene), poly(propylene), poly(urethane)s, and
deendothelialized artery. poly(tetrafluoroethylene)) achieve their water-insolubility
Given that one may want to employ hydrogels in tis- by interacting poorly with water. These hydrophobic poly-
sue engineering for the delivery of therapeutics and even mers accordingly adsorb large amounts of proteins, which
living cells to polymeric scaffolds and even preexisting ar- act as surfactants to expose more hydrophobic domains to-
Hydrogels in Biological Control During Graft Healing 565
Fig. 51.2. As an example of a transformation that can be carried out in situ, hydrogels can be formed from macromolecular
precursors dissolved in buffered saline, directly on the tissue surface. A tissue surface to be treated (a), such as the anastomotic
region of a vascular graft, is first flushed with a solution of a nontoxic photoinitiator (in this example eosin Y), to stain the tissue
surface with the photoinitiator (b). After the nonadsorbed photoinitiator is flushed away and the artery lumen is filled with the
other components of the initiation system (in this example, the nontoxic buffer triethanolamine) and the polymerizable macromer
(such as a diacrylated poly(ethylene glycol) copolymer with lactic acid, as in Fig. 51.1) (c), the artery is irradiated with light at an
appropriate wavelength (in the case of eosin Y, green light) to cause a wave of polymerization to proceed from the tissue surface out
into the lumen (d). After the nonpolymerized macromer solution is rinsed away, the final result is a crosslinked solid hydrogel,
adherent to and conformal with the tissue surface (e). In the photoinitiation scheme shown in this example, the red eosin Y adsorbs
green light and is excited to a triplet state, from which it participates in a single-electron transfer reaction with triethanolamine,
resulting in a radical on the eosin and the triethanolamine. The free radical on the triethanolamine rearranges and serves as the
actual initiator of the free radical polymerization of the diacrylated macromer.
ward the material surface and thus present a more hydro- Work on the theory of these interactions has been performed
philic new surface to the aqueous surroundings. Electrostatic by Jeon et al.27,28 These investigators determined that when
interactions are secondary (due to the high ionic strength the tethered polymer brush exists at a sufficient density of
of physiological fluids) but remain important in protein in- polymer chains of a sufficient length, compression of the
teraction with materials, especially with regard to adsorp- polymer brush by a potentially adsorbing protein results in
tion of proteins to cationic polymers (since almost all pro- a strong resistance, i.e., steric stabilization, to the adsorp-
teins bear a net anionic charge). Accordingly, one may de- tion of the protein. Thus, one may understand with these
sign a biomaterial to be relatively free of cell adhesion by theoretical analyses based on the physics of polymer chains
making it be both very hydrophilic (e.g., a hydrogel) and that the thermodynamic interactions are unfavorable for
nonionic or anionic. protein adsorption.29 Very well characterized experimenta-
Much of what is understood about protein and cell tion has also been performed with model two dimensional
interactions with hydrogels has been learned from two di- polymer brushes, e.g., by Prime and Whitesides employing
mensional hydrogel models, namely water-soluble polymers self-assembling monolayers of alkane thiols on gold sub-
(which would form hydrogels) tethered to insoluble surfaces. strates, the alkane thiols bearing grafted poly(ethylene glycol)
566 Tissue Engineering of Prosthetic Vascular Grafts
chains.30 These studies demonstrated experimentally that a biomaterials engineering aspects of incorporation of these
high density of poly(ethylene glycol) chains would suffice, signals into hydrogels are addressed.
even if the chains were very short, to dramatically limit pro- In order to be able to engineer a selective adhesion
tein adsorption. response to hydrogel materials, one must devise means by
The physical character of hydrogels is important, in which to incorporate synthetic adhesion ligands, such as
tandem with the chemical character, in determining hydro- synthetic peptides derived from adhesion proteins, into hy-
gel biocompatibility. This is to say, it is not just the chemical drogel networks. Hydrogels derived from poly(ethylene gly-
structure of the repeat unit of the polymer in the hydrogel col) diacrylate again serve as an instructive example. Hern
that determines the biocompatibility of the hydrogel. This and the author34 examined means by which to incorporate
can be considered in the context of crosslinked gels of adhesion peptides into these photopolymerizable gels, us-
poly(ethylene glycol) diacrylate.31 When gels were formed ing the adhesion peptide GRGDY as a model (the N-termi-
from poly(ethylene glycol) of molecular weight 200 or 1000, nal G serving as a spacer, and the C-terminal Y serving as a
these materials were rapidly encapsulated in a typical for- site for radioiodination). The N-terminus of the peptide was
eign body reaction and calcified after subcutaneous implan- acrylated, either with a minimal spacer or with a
tation in the rat; however, gels formed from poly(ethylene poly(ethylene glycol) chain of molecular weight 3400, and
glycol) diacrylate of higher molecular weight remained free these acrylated peptides were incorporated into the hydrogels
of fibrous encapsulation and did not calcify. The calcifica- by copolymerization (i.e., mixing the peptide acrylates and
tion response may be due to a specific interaction between the poly(ethylene glycol) diacrylates in the hydrogel precur-
Ca2+ and poly(ethylene glycol), but the induction of a for- sor solution, and then photopolymerizing the mixture).
eign body reaction almost certainly relates to the difference When the peptides were omitted from the hydrogel, the re-
in water content of these gels (from approximately 75% water sulting material was very resistant to cell adhesion. When
for the poly(ethylene glycol) 200 diacrylate to approx. 90% an acrylated RGD or control inactive RDG peptide was in-
for the higher molecular weight poly(ethylene glycol)-based corporated without a spacer arm, the resulting material sup-
hydrogels. As such, one should understand clearly that all ported cell adhesion in both cases, but only in the presence
hydrogels are not alike, and even that all hydrogels of the of serum proteins; this result can only be explained by the
same chemical composition are not necessarily alike in their peptide being sterically unavailable for biospecific binding
biological responses. to cell adhesion receptors but serving as a site for the non-
specific adsorption of serum proteins. When the RGD and
Designed Cell Adhesiveness of Hydrogels RDG peptides were incorporated with a poly(ethylene gly-
In the above section it was demonstrated that, with col) spacer, the materials with RGD supported cell adhe-
adequate attention to the details of hydrogel design, sion, while those with the inactive RDG supported no adhe-
hydrogels can be formed to effectively resist the attachment sion. Thus, it would seem that for the sequence to be steri-
of cells, even in vivo. Based upon this observation it then cally available for binding to receptors on the cell it must be
becomes attractive to incorporate into such a hydrogel bio- immobilized within the gel via a long spacer; and it would
logical adhesion ligands targeting specific receptors and even further seem that the peptide immobilized without the
perhaps specific cell types. In this manner, one might be able spacer serves as a site for protein adsorption, while the pep-
to develop a hydrogel material for tissue engineering that tide immobilized with the spacer does not. Having these re-
would reject the attachment on one cell type (e.g., the plate- sults in hand, one is then prepared to begin to incorporate
let) but that would accept the attachment of another cell cell type specific adhesion signals (such as the endothelial
type (e.g., the endothelial cell). Tissue engineering with syn- targeting REDV sequence) to engineer the tissue response
thetic adhesion ligands is based on profound advances in to a hydrogel, and thus to a vascular graft.
the molecular biology of cell adhesion over the past several
years, in which a host of extracellular matrix adhesion gly- Nonenzymatic Degradation of Hydrogels
coproteins have been identified, together with their corre- It is clear that one must have in one’s repertoire of
sponding receptors. Moreover, the domains on many of the materials for tissue engineering hydrogels that are degrad-
adhesion proteins that bind to the receptors have been iden- able. Degradation by nonenzymatic hydrolysis can be ob-
tified, and it has been demonstrated that, in many of these tain by incorporating hydrolytically labile bonds within the
cases, synthetic peptide and nonpeptide analogs can be de- gel precursor, either in a crosslinker or within the polymer
veloped with appropriate affinity for the appropriate recep- backbone. The example of poly(ethylene glycol) diacrylate
tors on the cell surfaces. It has also been demonstrated that hydrogels, in this case degradable variations, will be selected
some of these adhesion ligands have affinity for receptors for discussion.
that exist on one cell type do not exist on other cell types A host of degradable polymers are available for use in
present in the same biological environment; e.g., the se- medicine, and these are based on hydrolysis of esters,
quence REDV, which is present in some fibronectin forms, carbonates, and anhydrides, to name only a few of the im-
binds to the integrin receptor #4!1 that is present on human portant approaches (see chapter 47). These compositions
endothelial cells but not on human blood platelets.32,33 The can be incorporated into polymers that form hydrogels, to
biology of adhesion proteins, their receptors and synthetic obtain the physical and biological properties of the hydro-
analogs of the adhesion proteins is the topic of chapter 54 in gel along with the degradability inferred by the water-labile
this book and is not addressed further here; only the bond. To accomplish this in the scheme of photopolymerized
poly(ethylene glycol) diacrylate hydrogels, block copolymers
Hydrogels in Biological Control During Graft Healing 567
have been formed, with poly(ethylene glycol) forming the activity to clear the fibrin from their path. Electron
central block and oligomers of, e.g., lactic acid forming microscopic observation of the morphology of the neurite
flanking blocks; the termini of the lactic acid blocks can be tips and the fibrin near the neurite tips demonstrated that
acrylated, leading to an analog of poly(ethylene glycol) fibrils were present in the immediate vicinity of all aspects
diacrylate with a degradable segment between the of the growth cone, with less than a 50 nm gap between the
poly(ethylene glycol) segments.35 The hydrogel to result from neurite plasma membrane and the fibrin fibrils, suggesting
such a precursor consists of nodes of oligo(acrylic acid) that the neurite did not migrate by finding a pathway that
esterified to poly(ethylene glycol) chains via an oligo(lactic was enzymatically created in a spatially nonlocalized man-
acid) intermediate. Degradation thus yields a low molecular ner, but rather that local enzymatic activity associated with
weight oligo(acrylic acid), lactic acid and its oligomers, and the cell membrane was responsible for local removal of
poly(ethylene glycol) chains of the original molecular weight. material to create a pathway. Furthermore, these studies also
The rates of degradation of hydrogels behave some- demonstrated that there was no dependence in neurite mi-
what differently than those of the poly(ester), gration rate on fibrin fibril morphology, but only on den-
poly(carbonate), or poly(anhydride) parent family. In the sity; i.e., the only feature that mattered was the amount of
parent families, two features control the rate of degradation: material present to be cleared from the pathway of the cell
The intrinsic susceptibility of the water-labile bond to hy- to permit migration.
drolysis, and the rate of transport of water into the hydro- Studies as described above both emphasize the im-
phobic polymer. In hydrogels, by contrast, the entire mac- portance of biologically derived materials for tissue engi-
roscopic material is equally exposed to water, so the intrin- neering and also provide motivation to develop synthetic
sic rate of hydrolysis controls the overall degradation rate. analogs. The fact that, at least in some circumstances, the
This feature can be used to modulate the rate of resorption detailed morphology of the biological structures to be cleared
of such hydrogels: For example, poly(ethylene glycol)- by cell-associated proteolytic activity is less important than
glycolide diacrylate hydrogels degrade faster than the mass of that material encourages one to proceed with
poly(ethylene glycol)-lactide diacrylate hydrogels, which development of such synthetic analogs, in that only the
degrade faster than poly(ethylene glycol)-caprolactone chemical nature need be mimicked, not the morphological
diacrylate hydrogels, in accordance with the expected order nature as well (at least in some cases). With this in mind,
based on chemical kinetics. West et al have examined poly(ethylene glycol)-based hy-
drogel materials that degrade by proteolytic activity.38
Enzymatic Degradation of Hydrogels Poly(ethylene glycol) was used as a substrate for liquid phase
In addition to degradation based on nonenzymatic peptide synthesis, and either plasmin or collagenase-sensi-
hydrolysis, one could contemplate degradation based on tive peptide sequences were synthesized at the termini of
enzymatic processes. Enzymatic degradation may have con- the polymer chains. Upon the termini of these peptide blocks
ceptual advantages over nonenzymatic degradation in some an acrylate group was coupled, to create an analog of the
applications of tissue engineering. For example, cell migra- poly(ethylene glycol)-lactide diacrylate, namely
tion through tissues and through fibrin clots and granula- poly(ethylene glycol)-peptide diacrylate, where the identify
tion tissue in wound healing is dependent upon enzymatic of the peptide was selected for sensitivity to the protease plas-
hydrolysis of glycoproteins and glycosaminoglycans in the min or collagenase. Hydrogels were formed from such pre-
extracellular matrix. Thus, endowing a hydrogel with the cursors and were exposed to either plasmin or collagenase,
ability to be degraded by enzymes would permit the hydro- with the result that the plasmin-sensitive gel degraded in
gel to be remodeled by biological processes associated with the presence of plasmin but not collagenase, and the colla-
cell migration. genase-sensitive gel degraded in the presence of collagenase
It is instructive to consider cell migration through bio- but not plasmin, as designed. Such preliminary studies dem-
logically derived hydrogels, as a prototype for the ultimate onstrate at least the potential for the synthesis of hydrogel
design of synthetic materials that permit cell infiltration by materials that mimic the ability to be remodeled like the
proteolysis. Fibrin serves as one such good prototype, being natural biological hydrogels used in nature.
the primary matrix through which cells migrate in a healing
response. Herbert et al examined the migration of the growth Controlled Release
cones of neurites emanating from aggregated neural and glial Hydrogels have been extensively explored for the con-
cells within a three dimensional fibrin matrix in a culture trolled release of drugs, particularly macromolecular drugs
model.36,37 Fibrin was formed under controlled conditions such as proteins and oligonucleotides. One reason for uti-
around the cell aggregates, and the rate of neurite extension lizing hydrogels for drug delivery is the relative ease in con-
was measured as a function of fibrin fibril density and mor- trolling the permeability of the gel structure to the macro-
phology; additionally, the dependence of the extension pro- molecular drug. While in most gels permanent pore struc-
cess on local protease activity was examined. It was observed tures do not exist, transient pores do exist that can be de-
that either direct inhibition of plasmin activity or inhibi- scribed in terms of molecular weight between crosslinks and
tion of plasminogen activation activity completely inhib- pore dimensions.39 These parameters can be readily con-
ited cell migration, indicating that the neurites did not find trolled, and by adjusting these dimensions relative to the
preexisting pores through which to migrate, and that they radius of gyration of the macromolecular drug one can ei-
did not mechanically push fibrin fibrils out of their path ther restrict drug diffusion to a controlled degree and ob-
during migration, but rather that they expressed chemical tain release from the hydrogel material on a time scale faster
568 Tissue Engineering of Prosthetic Vascular Grafts
than that of degradation, or one can entrap the drug, using Graft Sealing
a gel with a pore dimension much less than the drug size, Knitted and woven grafts must be preclotted to pre-
and obtain drug release that is mediated by the rate of hy- vent excessive blood leakage from the graft, and this process
drogel degradation.40 It is relatively straightforward to syn- of preclotting may predispose the graft to further thrombo-
thetically manipulate the pore dimensions of a hydrogel. For sis in the short term (e.g., due to platelet interactions with
example, with gels formed by crosslinking an unsaturated fibrin in the clot or due to prothrombin activation on the
monomer (refer to the example of acrylamide with a bis- surfaces of cells in the clot) and to the development of an
acrylamido crosslinker) the pore dimensions may be altered excessive neointima in the long term (e.g., due to the growth
via control of the ratio of acrylamide to bis-acrylamide, a factors that are released by activated platelets in the clot). To
high amount of bis-acrylamido crosslinker yielding small reduce the possibility of short and long term graft failure as
pore dimensions and restricted permeability. In the example a result of preclotting, grafts that are based on collagen,44
of poly(ethylene glycol)-diacrylate based hydrogels, one may gelatin,45 and albumin46 preclotting have been developed.
alter pore dimensions via the molecular weight of the These materials are indeed hydrogels, and thus hydrogel-
poly(ethylene glycol) diacrylate, low molecular weights yield- sealed vascular grafts have already been used clinically. One
ing small pore dimensions and low permeability. As an ex- could also employ hydrogels designed more specifically for
ample, Cruise et al41 reported pore dimensions of 19 Å for graft sealing. Such a hydrogel sealing material could either
gels formed from poly(ethylene glycol) 2000 diacrylate, 22 resorb by physical erosion (e.g., with poly(ethylene glycol)-
Å from poly(ethylene glycol) 4000 diacrylate, 34 Å from poly(propylene glycol)-poly(ethylene glycol) block copoly-
poly(ethylene glycol) 8000 diacrylate and 58 Å from mers) or by nonenzymatic hydrolysis (e.g., with a
poly(ethylene glycol) 20,000 diacrylate. It was likewise pos- poly(ethylene glycol)-lactide diacrylate hydrogel). Such a
sible to control pore dimensions via manipulation of the designed hydrogel has the possible benefit that other fea-
reaction conditions with the same poly(ethylene glycol) tures could also be designed into the gel, as described below.
diacrylate precursor; e.g., pore dimensions of 70 Å were
obtained by polymerization of poly(ethylene glycol) Promotion of Endothelialization
diacrylate 20,000 from a 10% precursor solution, 58 Å from It is clear that the rapid establishment of an endothe-
a 20% precursor solution, and 42 Å from a 30% precursor lial cell monolayer atop the neointima within a graft is key
solution. Because of this high flexibility in the control of to success in the tissue engineering of a vascular graft. It is
pore dimensions and gel permeability to macromolecular also clear that thrombosis within the graft lumen is an un-
drugs, hydrogels are extremely attractive for controlled re- desirable outcome. In light of these two observations, one
lease applications in tissue engineering. could consider employing a hydrogel coating or a hydrogel
sealing to both reduce thrombosis and to enhance
Design Principles, Hydrogels for Biological endothelialization, e.g., via a gel that resists fibrinogen ad-
Control During Graft Healing sorption and accordant platelet deposition, that gel also con-
One may consider many possible applications for taining a grafted ligand that demonstrates specificity for an
hydrogels in tissue engineering, some of which have been endothelial cell adhesion receptor. In such a manner, it might
explored already and some of which remain to be experi- be possible to fail to induce platelet adhesion but to succeed
mentally addressed. In this section, some of the more im- in inducing endothelial cell adhesion.32 In the absence of
portant possible applications in vascular tissue engineering seeding of endothelial cells along the entire lumen of the
will be called into focus. These relate to altering the blood graft, one must be concerned with the rate of endothelial
response to a graft, altering the arterial response to the graft, cell migration along the coated graft surface. Lauffenburger
and directing the response of the surrounding tissue to a and colleagues have demonstrated, both based on theoreti-
graft implant. cal considerations and also by experiment, that there exists
an optimum in the surface density of adhesion ligands for
Reduction of Thrombosis fastest cell migration rates.47 At low surface density of adhe-
Because hydrogels can be designed that adsorb rela- sion ligand, cell traction is low and cell migration rates, as
tively low amounts of protein and that thus support rela- well as adhesion strength, are accordingly slow. At high sur-
tively low amounts of platelet adhesion (which has been face density of adhesion ligand, cell traction is very high,
demonstrated to be modulated principally by adsorbed fi- but the rate of binding of an adhesion receptor on the cell
brinogen),42 one obvious, yet important, application of surface with surface-bound adhesion ligand is very high,
hydrogels to vascular graft tissue engineering is the reduc- resulting in very strong adhesion but very low cell migra-
tion of platelet deposition in the hours and days following tion rates. Accordingly, one must design the display of ad-
implantation. Hydrogel coatings have been used to modu- hesion ligands in the hydrogel coating such that adhesion
late the deposition of platelets and other cells on a host of strength is sufficient to resist fluid forces within the vascular
medical devices, most of which are implanted for relatively flow but such that it is not so strong that the rate of cell
short periods of time. Indeed, heparinization of graft sur- migration is unacceptably reduced.
faces may improve graft biocompatibility by both a phar- It has been demonstrated that thrombosis at the anas-
macological effect (the specific biological activity of hep- tomoses of a graft may be well in excess of thrombosis on
arin as it interacts with antithrombin III) as well as a bio- the graft itself.48 Accordingly, efforts at reducing platelet
physical effect (the hydrophilic, anionic heparin hydrogel deposition on the biomaterial per se may be slightly mis-
coating acting to reduce the adsorption of fibrinogen).43
Hydrogels in Biological Control During Graft Healing 569
placed relative to the thrombogenic potential of the anasto- stimulate endothelial cell migration and proliferation, and
moses. One may then consider the use of a hydrogel to pas- another factor to limit perigraft inflammation or smooth
sivate the graft anastomoses. It was demonstrated above that muscle activity.
one advantage of hydrogels over other materials for tissue Stated more generally, Nature’s extracellular matrix
engineering is that they may be formed on the surface of a hydrogel serves to provide a great deal of information to
tissue by an in situ transformation. As an example, it was cells to direct their behavior and organization in space. It
discussed that hydrogel coatings have been formed on the provides information in the form of structural cues, of im-
surfaces of injured arteries to replace one function of the mobilized adhesion molecules, and of immobilized and dif-
endothelium, namely to regulate the deposition of blood fusible polypeptide growth factors. Furthermore, it receives
platelet on the arterial surface.22 Accordingly, it could be information from the cells as they respond. It does this by
envisioned to limit the extent of thrombus deposition on binding to growth factors that are released from cells and by
the anastomoses within a graft that had already been su- degrading in the face of cellularly-displayed enzymes. It
tured in place by the local, in situ deposition of a hydrogel might be possible, and indeed advantageous, in tissue engi-
coating spanning the junction of the native artery and the neering of the vascular graft, to mimic some or all of these
implanted graft. Using such an approach, it might be pos- activities, either in a biologically derived hydrogel matrix or
sible to directly impact one of the most troublesome regions in a completely synthetic biomimetic hydrogel matrix, with
of an implanted graft. the goal of harnessing some of these biological activities for
controlling tissue morphogenesis and function.
Drug Release
Hydrogels have been explored extensively in drug References
release, due to the facility with which the permeability of 1. Shimizu M, Minakuchi K, Moon M, and Koga J, Differ-
the gel material to a drug of a given size may be modulated. ence in interaction of fibronectin with type I collagen and
Because of this high flexibility in control of release type IV collagen. Biochim Biophys Acta 1997; 1339:53-61.
characteristics, it is attractive to combine some of the other 2. Jackson RL, Busch SJ, and Cardin AD, Glycosaminogly-
cans: Molecular properties, protein interactions, and role
uses described above with local release of a biologically active
in physiological processes. Physiol Rev 1991; 71:481-539.
factor, e.g., an angiogenic growth factor such as basic fibro- 3. Vanderrest M, and Garrone R, Collagen family of pro-
blast growth factor or vascular endothelial growth factor, to teins. FASEB J 1991; 5:2814-2823.
induce a specific biological response in tissue engineering 4. Mosher DF, Assembly of fibronectin into extracellular
of the vascular graft. Some experimentation has been per- matrix. Curr Opin Struct Biol 1993; 3:214-222.
formed along these lines, using hydrogels in vascular tissue 5. Parkhurst MR, and Saltzman WM, Quantification of hu-
engineering. For example, Simons et al49 employed a hydro- man neutrophil motility in 3-dimensional collagen gels:
gel, formed in situ by application of liquid solution of a co- Effect of collagen concentration. Biophys J 1992;
polymer of poly(ethylene glycol)-poly(propylene glycol)- 61:306-315.
poly(ethylene glycol) block copolymer, which solidifies to 6. Langer R, and Vacanti JP, Tissue engineering. Science
form a gel upon warming to body temperature.13 This gel 1993; 260:920-926.
7. Peppas NA, and Langer R, Challenges in biomaterials.
was used to release an antisense oligonucleotide that inhib-
Science 1994; 263:1715-1720.
ited cell proliferation; this treatment was observed to greatly 8. Hubbell JA, Biomaterials in tissue engineering. Bio/Tech-
limit the thickening of the neointima following injury by nology 1995; 13: 565-576.
balloon angioplasty of the carotid arteries of rats. As another 9. Elbert DL, and Hubbell JA, Surface treatments of poly-
example, Greisler and colleagues50 employed a fibrin-based mers for biocompatibility. Ann Rev Material Sci 1996; 26:
hydrogel formed within the pore spaces of a vascular graft 365-394.
to release basic fibroblast growth factor, and this resulted in 10. Kim SW, Bae YH, and Okano T, Hydrogels: Swelling, drug
an overall acceleration of the rate of endothelialization and loading, and release. Pharm Res 1992; 9:283-290.
a corresponding limitation of the formation of an exces- 11. Slack SM, and Horbett TA, Changes in fibrinogen
sively thickened neointima in animal models. adsorbed to segmented polyurethanes and hydroxyethyl-
methacrylate-ethylmethacrylate copolymers. J Biomed
Mater Res 1992; 26:1633-1649.
Promotion of Transmural Capillary Ingrowth
12. Mallapragada SK, Peppas NA, and Colombo P, Crystal
Studies by Golden et al provided great encouragement dissolution-controlled release systems. II. Metronidazole
for the possibility of transmural capillary infiltration and release from semicrystalline poly(vinylalcohol) systems. J
differentiation to form large vessel endothelium with large Biomed Mater Res 1997; 36:125-130.
pore graft implants in nonhuman primate models.51 One 13. Leach RE, and Henry RL. Reduction of postoperative ad-
can consider designing hydrogel treatments for tissue-engi- hesion in the rat uterine horn model with Poloxamer 407.
neered vascular grafts to attempt to achieve similar results Am J Obstet Gynecol 1990; 162:1317-1319.
in the human. In such an approach, one might integrate the 14. O’Shea GM, and Sun AM, Encapsulation of rat islets of
concepts that have been presented above: For example, one Langerhans prolongs xenograft survival in diabetic mice.
might consider sealing a graft with a hydrogel that bore cell Diabetes 1986; 35:943-946.
15. Decher G, Lvov Y, and Schmitt J, Proof of multilayer
adhesion sites for endothelial cells but not for blood plate-
structural organization in self-assembled polycation-
lets, which further bore sites for degradation by proteases polyanion molecular films. Thin Solid Films 1994;
expressed during endothelial cell migration. One might fur- 244:772-777.
ther incorporate a growth factor within such a hydrogel to
570 Tissue Engineering of Prosthetic Vascular Grafts
16. Gin H, Dupuy B, Caix J, Baquey C, and Ducassou D, In 34. Hern DL, and Hubbell JA, Incorporation of adhesion pep-
vitro diffusion in polyacrylamide embedded agarose tides into nonadhesive hydrogels useful for tissue resur-
microbeads. J Microencapsul 1990; 7:17-23. facing. J Biomed Mater Res, 1997; 39:266-276.
17. Pathak CP, Sawhney AS, and Hubbell JA, Rapid 35. Sawhney AS, Pathak CP, and Hubbell JA, Bioerodible
photopolymerization of gels in contact with cells and tis- hydrogels based on photopolymerized poly(ethylene gly-
sue. J Am Chem Soc 1992; 114:8311-8312. col)-co-poly(#-hydroxy acid) diacrylate macromers. Mac-
18. Moser D, Blood coagulation and fibrinolysis. Clin Cardiol romolecules 1993; 26:581-587.
1990; 13:5-11. 36. Herbert CB, Bittner GD, and Hubbell JA, Effects of fi-
19. Hubbell JA, In situ material transformations in tissue brinolysis on neurite growth from dorsal root ganglia cul-
engineering. MRS Bulletin 1996; 21:33-35. tured in 2-dimensional and 3-dimensional fibrin gels. J
20. Sawhney AS, Pathak CP, Van Rensberg JL, and Hubbell Comp Neurol 1996; 365:380-391.
JA, Optimization of photopolymerized bioerodible hydro- 37. Herbert CB, Nagaswami C, Bittner GD, JW Weisel, and
gel properties for adhesion prevention. J Biomed Mater Hubbell JA, Effects of fibrin micro-morphology on neu-
Res 1994; 28: 831-838. rite growth from dorsal root ganglia cultured within three-
21. Hill-West JL, Chowdhury SM, Sawhney AS, Pathak CP, dimensional fibrin gels. J Biomed Mater Res, 1997; in ress.
Dunn RC, and Hubbell JA, Prevetion of postoperative 38. West JL, Pratt A, and Hubbell JA, Proteolytically degrad-
adhesions in the rat by in situ photopolymerization of able hydrogels, Trans Soc Biomaterials 1997; 23:103.
bioresorbable hydrogel barrires. Obstet Gynecol 1994; 39. Canal T, and Peppas NA, Correlation between mesh size
83:59-64. and equilibrium degree of swelling of polymeric networks.
22. Hill-West JL, Chowdhury SM, Slepian MJ, and Hubbell J Biomed Mater Res 1989; 23:1183-1193.
JA, Inhibition of thrombosis and intimal thickening by 40. JA Hubbell, Hydrogel systems for barriers and local drug
in situ photopolymerization of thin hydrogel barriers. Proc delivery in the control of wound healing. J Contr Rel 1996;
Nat Acad Sci USA 1994; 91:5967-5971. 39:305-313.
23. West JL and Hubbell JA, Separation of the arterial wall 41. Cruise GM, Scharp DS, and Hubbell JA, Characterization
from blood contact using hydrogel barriers reduces inti- of permeability and network structure of interfacially
mal thickening after balloon injury in the rat: The roles photopolymerized poly(ethylene glycol) diacrylate
of medial and luminal factors in arterial healing. Proc Nat hydrogels. Biomaterials 1998; 19:1287-1294.
Acad Sci USA, 1996; 93:13188-13193. 42. Kiaei D, Hoffman AS, Horbett TA, and Lew KR, Platelet
24. Sawhney AS, Pathak CP, and Hubbell JA, Modification and monoclonal antibody binding to fibrinogen adsorbed
of islet of Langerhans surfaces with immunoprotective on glow-discharge-deposited polymers. J Biomed Mater
poly(ethylene glycol) coatings via interfacial Res 1995; 29:729-739.
photopolymerization. Biotechnol Bioeng 1994; 44:383-386. 43. Hubbell JA, Pharmacologic modification of materials.
25. Pettit DK, Hoffman AS, and Horbett TA, Correlation be- Cardiovasc Pathol 1993; 2:121S-128S.
tween corneal epithelial cell outgrowth and monoclonal 44. Tolan M, Wells F, Kendall S, Large S, and Wallwork J,
antibody binding to the cell-binding domain of adsorbed Clinical experience with a collagen-impregnated woven
fibronectin. J Biomed Mater Res 1994; 28:685-691. Dacron graft. J Cardiovasc Surg 1995; 36:323-327.
26. Andrade JD, and Hlady V, Protein adsorption and mate- 45. Utoh J, Goto H, Obayashi H, Hirata T, and Miyauchi Y,
rials biocompatibility: A tutorial review and suggested Dilation of gelatin-sealed knitted dacron prosthesis. J
hypotheses. Adv Polym Sci 1986; 79:1-63. Cardiovasc Surg 1996; 37:343-344.
27. Jeon Si, Lee JH, Andrade JD, and de Gennes PG, Pro- 46. Kang SS, Petsikas D, Murchan P, Cziperle DJ, Ren D, Kim
tein-surface interactions in the presence of polyethylene DU, and Greisler HP, Effects of albumin coating of knit-
oxide I, simplified theory. J Coll Interf Sci 1991; ted Dacron grafts on transinterstitial blood loss and tis-
142:149-158. sue ingrowth and incorporation. Cardiovasc Surg 1997;
28. Jeon SI, and Andrade JD, Protein-surface interactions in 5:184-189.
the presence of polyethylene oxide II, effect of protein size. 47. DiMilla PA, Stone JA, Quinn JA, Albelda SM, and
J Colloid Interface Sci 1991; 142:159-166. Lauffenberger DA, Maximal migration of human smooth
29. Norde W, Protein adsorption at solid surfaces: A ther- muscle cells on fibronectin and type IV collagen occurs
modynamic approach. Pure Appl Chem 1994; 66:491-496. at an intermediate attachment strength. J Cell Biol 1993;
30. Prime KL, and Whitesides GM, Adsorption of proteins 122:729-737.
onto surfaces containing end-attached oligo(ethylene ox- 48. Johnson PC, Dickson CS, Garrett KO, Sheppeck RA, and
ide): A model system using self-assembled monolayers. J Bentz ML, The effect of microvascular anastomosis con-
Am Chem Soc 1993; 115:10714-10721. figuration on initial platelet deposition. Plast Reconstr
31. Hossainy SFA, and Hubbell JA, Molecular weight depen- Surg 1993; 91:522-527.
dence of calcification of poly(ethylene glycol) hydrogels. 49. Simons M, Edelman ER, DeKeyser JL, Langer R, and
Biomaterials 1994; 15:921-925. Rosenberg RD, Antisense c-myb oligonucleotides inhibit
32. Hubbell JA, Massia SP, Desai NP, and Drumheller PD intimal arterial smooth muscle cell accumulation in vivo.
Endothelial cell-selective materials for tissue engineering Nature 1992; 359:67-70.
in the vascular graft via a new receptor. Bio/Technology 50. Greisler HP, Cziperle DJ, Kim DU, Garfield JD, Petsikas
1991; 9:568-572. D, Murchan PM, Applegren EO, Drohan W, and Burgess
33. Massia SP, and Hubbell JA, Vascular endothelial cell ad- WH, Enhanced endothelialization of expanded
hesion and spreading promoted by the peptide REDV of polytetrafluoroethylene grafts by fibroblast growth factor
the IIICS region of plasma fibronectin is mediated by type 1 pretreatment. Surgery 1992; 112:244-254.
integrin #4!1. J Biol Chem 1992; 267:14019-14026. 51. Golden MA, Hanson SR, Kirkman TR, and Clowes AW,
Healing of poly(tetrafluoroethylene) arterial grafts is in-
fluenced by graft porosity. J Vasc Surg 1990; 22:838-844.
Matrix Engineering
CHAPTER 52
In Vivo Synthesis of Organs Using Collagen-GAG Copolymers
Ioannis V. Yannas
Introduction
In this chapter, I will first present the evidence supporting the conclusion that organ
regeneration can indeed be reproducibly achieved both in animal models and clinically. The
methodology of induced regeneration will then be briefly summarized, followed by an
introduction to the models which are used to describe the mechanism of induced
regeneration.
Although in vivo synthesis of vascular tissue has not yet been demonstrated, I suggest
that future experimentation in this area could be very fruitful and could benefit directly by
use of the principles described briefly below for other tissues and organs.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
572 Tissue Engineering of Prosthetic Vascular Grafts
in large areas of full-thickness skin loss in the guinea pig.3-10 has an average pore diameter of 5 ∝m, an average molecular
This finding has been independently confirmed repeatedly weight between crosslinks of 30-40 kDa, a preferred orien-
in humans11-13 and recently in swine.14 The evidence for re- tation of pore channel axes in the direction of the nerve axis,
generation was based on histological and ultrastructural and a 98/2 w/w ratio of type I collagen to GAG.16,17 Clearly,
studies, the use of small-angle laser light scattering studies there are significant differences between the network struc-
of histological sections, and functional studies. It was con- ture of skin and nerve regeneration templates.
cluded that the new skin was structurally and functionally Yet a third ECM analog has been reported, capable of
competent; however, it was totally lacking in hair follicles inducing regeneration of the canine meniscus following 80%
and other skin adenexa. transection.21,22 Although the ECM analog used in these
The emphasis in early studies of in vivo synthesis of studies has been stated by the authors to be similar to that
skin was on keratinocyte seeding of the analog prior to graft- described earlier,9 its detailed structure was not reported.
ing, as this procedure led to simultaneous regeneration of There is, therefore, substantial evidence that, under
an epidermis as well as a dermis. It was later appreciated appropriate conditions, the adult mammal can be induced
that cell seeding speeds up epidermal regeneration but it to regrow certain organs which are not regenerated sponta-
appears otherwise not to be required for regeneration ei- neously. This exciting conclusion suggests new therapeutic
ther of the epidermis or of the dermis. avenues. However, there are still many unanswered ques-
The evidence cited above led to identification of a cell- tions about the detailed cell-biological mechanism by which
free macromolecular network with highly specific structure, active ECM analogs modify so spectacularly one or more of
the cell-free skin regeneration template (SRT), which alone the processes involved in conventional wound healing.
possessed this unprecedented morphogenetic activity. Only
one of several collagen-GAG matrices studied as described
above was capable of preventing scar tissue formation and Methodological Principles
promoting dermal regeneration. The active ECM analog was of Induced Regeneration
characterized by a collagen/GAG ratio of 98/2 w/w, average Over the years it has become clear that a number of
pore diameter between 20 and 120 ∝m, random orientation conditions must be in place in order to conduct a reproduc-
of pore channels, and sufficiently high density of covalent ible study of induced regeneration. One of the most impor-
crosslinks (average molecular weight between crosslinks in tant is the choice of an anatomically well-defined lesion (ex-
the template, 12 kDa) to resist degradation by collagenases perimental volume) in which to study the phenomena. This
for about 10 days following grafting. Several other very concept was pioneered in the studies of Billingham and
closely related ECM analogs showed either significantly re- Medawar1 as a means of minimizing animal to animal vari-
duced activity or no activity at all.9 In related studies it was ability in observations. The experimental volume can be iso-
observed that ECM analogs which showed high activity in lated by containing it within anatomically distinct tissues
promoting dermal regeneration also delayed significantly the belonging to a neighboring organ or by a device or (in the
onset of wound contraction.9 The available evidence com- case of skin) by the atmosphere.
pelled the conclusion that the activity of this insoluble net- Since mammals possess a very small but finite poten-
work depended critically on maintenance of a highly spe- tial for regeneration, it is necessary to “correct” the observed
cific three dimensional structure inside the wound bed over regeneration by subtracting the amount which has been con-
a period of time between about 5 and 15 days. The active tributed by spontaneous regeneration. This requirement is
network has been referred to as skin regeneration template equivalent to defining a negative control in every experi-
(SRT). The observed activity of SRT, consisting in drastic mental model, consisting of the observations obtained in
modification of the outcome of the wound healing process, the absence of the presumptive regeneraion template.
has not been duplicated by application on the wound bed Although the mechanistic interpretation of induced
of solutions of one or more growth factors or by application regeneration has not been completed, two arguments have
of suspensions of keratinocytes or fibroblasts. been advanced to support the contention that there is a re-
Regeneration of a partially functional sciatic nerve was quirement for structuring a regeneration template as an
induced across a transected gap of 15 mm in the rat sciatic ECM analog. First, studies of organ development have shown
nerve15-18 by another ECM analog, which was also found to conclusively that the early presence of specific ECM com-
possess a highly specific network structure. In this animal ponents is required for formation of physiological or-
model the nerve stumps at either side of the gap were in- gans.23-25 In contrast, the exudate which flows into a spon-
serted in a silicone tube (tubulation); in the absence of a taneously healing wound after injury does not contain com-
tube, regeneration was decidedly absent and neuroma for- ponents of the insoluble and nondiffusible ECM networks
mation was invariably reported. Although spontaneous re- (although it contains several types of cells and regulators);
generation of nerve through the tube occurred reproduc- the healing process in this case eventually leads to synthesis
ibly at a gap length of 5 mm, regeneration across a 15 mm of scar. Second, the evidence presented in the preceding sec-
gap has not been observed.19,20 The study eventually led to tion clearly supports the contention that specific ECM ana-
identification of an ECM analog which, when filling the sili- logs indeed suffice to lead to regeneration of at least two
cone tube and thereby connecting the two nerve stumps, organs. However, there is clearly insufficient evidence to
induced regeneration across a 15 mm gap. This analog has conclude definitively that regeneration can be induced only
been referred to as nerve regeneration template (NRT). It by specific ECM analogs.
In Vivo Synthesis of Organs Using Collagen-GAG Copolymers 573
Conclusive evidence that an ECM analog has induced mole/cm3/s. Picture the nutrient being transported from the
organ regeneration can only be obtained with a performance solid-like tissue, where the concentration of nutrient is as-
assay which simply compares the structure of the regener- sumed to be a constant C0 due to the presence of vascular
ate to that of a normal control. Assays of this type can be supply, over a distance L through the exudate, until it reaches
destructive (e.g., histology), in which case only one experi- the cell. Prior to the onset of angiogenesis, i.e., during the
mental time point can be obtained per animal, or first few days following template implantation, the nutrient
nondestructive, such that the complete kinetic curve for re- is transported exclusively by diffusion, characterized by a
generation can be obtained from a single animal. The fidel- diffusivity D cm2/s. Dimensional analysis readily yields the
ity of regeneration is a figure of merit which is assigned to cell lifeline number:
each candidate template. The regeneration template is then
the ECM analog which has scored a very high value of the S = RL2/DC0 (2)
fidelity index.
which can be used to compare the relative magnitude
of the rate of nutrient consumption by the cell nutrient (nu-
Models of the Mechanism of Regeneration merator) to the rate of supply of nutrient to the cell by dif-
I summarize below the major physicochemical mod- fusion (denominator). The cell dies if the rate of consump-
els26-28 which have been used to compile a mechanistic in- tion of the critical nutrient exceeds greatly the rate of sup-
terpretation of the process of regeneration. It is evident that ply, S>>1. At steady state the rate of consumption of nutri-
a regeneration template modifies drastically the kinetics and ent by the cell just equals the rate of transport by diffusion
mechanism of spontaneous wound healing which normally over the distance L. Under conditions of steady state,
lead to wound contraction and scar synthesis. However, a S = O(1); at that point, the value of L becomes the critical
view of a template as a classical catalyst is not supported by cell path length, Lc, the distance of migration beyond which
the well known fact that regeneration templates are neces- cells require the presence of a vascular supply. Another way
sarily consumed during the process which they modify. of looking at Lc is that it is the longest distance away from
the wound bed boundary along which the cell can migrate
Micrometer Scale Proximity of Host to Template Surface without requiring nutrient in excess of that supplied by dif-
Cells and cytokines present in the exudate are trans- fusion. For many cell nutrients of low molecular weight, Lc
ferred to the surface of the template following implantation is of order 100 ∝m, an estimate of the maximum template
of the porous template into the wound bed. Surface tension dimension which can support cells.
forces pull the exudate inside the capillaries (pore channels)
of the template, as described by: Template Specific Surface Is Bounded
Following its migration onto the template surface a
P = 2&/r (1) host cell is visualized interacting with binding sites on the
surface. The surface density of binding sites can be expressed
where r is the radius of the pore channel in a template as Λb, equal by definition to the number of sites Nb per unit
undergoing wetting by exudate with an air-liquid surface surface A of template. Λb can also be expressed more use-
tension of & in dyne/cm. Wetting of the opposed surfaces is fully in terms of quantities potentially measurable by opti-
promoted as the suction pressure P in Equation (1) increases, cal microscopy, i.e., in terms of the volume density of bind-
and such increase is occasioned by a decrease in average pore ing sites ∋b (number of sites per unit volume porous tem-
radius. The values reached by P are not negligible; water with plate) and the specific surface ( of the template expressed in
an air-liquid surface tension of & = 72 dyne/cm, for example, units of mm2/cm3:
is pulled inside a pore radius of 100 ∝m with a suction pres-
sure of almost one-hundredth of one atmosphere and P in- Λb = Nb/A = ∋b/( (3)
creases almost to one full atmosphere when the pore radius
decreases to a value as low as 1 ∝m. Under conditions where Assuming that each cell is bound to (an a priori un-
exudate flows inside the pore channels of a template with known number of) 5 binding sites, there will be Nb/5 bound
average pore diameter of 100 ∝m, cells and cytokines are cells per unit surface; the volume density of cells will be ∋c =
within a distance of less than 50 ∝m from the template sur- ∋b/5 and the surface density of cells will be:
face; this distance can then be covered by these components
of the exudate within a few minutes. Λc = Λb/5 = Nb/5A = ∋b/5( = ∋c/( (4)
Maximum Dimension of Template, ~100 ∝m Levels of myofibroblast density inside templates with
Cell migration from the solid-like tissue of the wound pore diameter of about 10 ∝m have been observed to reach
bed into the template requires the availability of adequate typical values of the volume density, ∋c, of order 107
nutrition. The precise nutritional requirements of any cell myofibroblasts per cm3 porous template. For a template of
are too complex to incorporate into a simple model; instead, average pore diameter 10 ∝m the specific surface ( is calcu-
these requirements will be simplified by defining a critical lated to be approximately 8 x 104 mm2/cm3 template; there-
nutrient which is required for normal cell function; such a fore, 1 cm3 porous template is characterized by a cell surface
nutrient is assumed to be metabolized by the cell at a rate R density of Λc = ∋c/( = 107/8 x 104 = 125 cells/mm2. For a
574 Tissue Engineering of Prosthetic Vascular Grafts
template of identical composition but average pore diam- bed in order not to impede cellular processes which lead to
eter as large as 300 ∝m, Λc is the same as above; however, the the emerging organ.
specific surface is calculated to be only about 3 x A steady state model is probably the simplest way to
103 mm2/cm3 template. In this case, the volume density of accommodate these two requirements. It requires synchro-
cells is, accordingly, only ∋c = Λc − ( = 125 x 3 x 103 = 3.75 x nization of the two processes: organ synthesis and template
105 per cm3 porous template. We conclude that the template degradation.5,9 This model leads directly to the hypothesis
which has the smaller average pore diameter (10 ∝m) has a of isomorphous tissue replacement:
volume density of myofibroblasts which is about 27 times
lower than with the template which has the larger pore di- td/ts = O(1) (5)
ameter (300 ∝m). The existence of a maximum pore diam-
eter requirement for the template is suggested by these cal- In Equation (5) td denotes a characteristic time con-
culations, simply to ensure a specific surface which is large stant for degradation of the template at the tissue site where
enough to bind an appropriately large number of cells. a new organ is synthesized with a time constant of ts. The
In addition, it is clear that cells originating in the degradation rate can be estimated by histological observa-
wound bed cannot migrate inside the template and eventu- tion of the decrease in mass of template fragments at vari-
ally reach binding sites on its surface unless the template ous times.10,30 A closer estimate of td has been obtained by
has an average pore diameter large enough to allow for this. measuring the kinetics of disintegration of the macromo-
We conclude that there is, therefore, a requirement for a lecular network using rubber elasticity theory.3 A third pro-
minimum pore diameter for the template, about equal to cedure consists of monitoring the kinetics of mass disap-
the characteristic diameter of the cells (of order 5 ∝m). Thus, pearance of a radioactively labeled template. A rough esti-
the pore diameter of the regeneration template is limited mate of ts can be obtained by observing th, the timescale of
both by an upper and a lower bound. This conclusion is in synthesis of new tissue during healing (in the absence of a
agreement with the experimental evidence which shows that template) at the anatomical site.31 Using the latter approach
ECM analogs, identical in chemical composition but differ- it has been estimated that ts for the regenerating dermis is of
ing only in average pore diameter, show maximum activity order 3 weeks31 and of order 6 weeks for the regenerating
(inhibition of onset of wound contraction, consistent with peripheral nerve.16 These estimates allow adjustment of td
regeneration rather than scar formation) when the average for the template, by adjustment of the crosslink density and
pore diameter lies between 20 and 120 ∝m.9 Further evi- GAG content, to levels which are approximately equal to the
dence has shown that, when other structural parameters of values of ts, as the latter are dictated by the nature of the
the template remain constant, loss of the 20-120 ∝m porous anatomical site.
structure of the template by simple evaporation at room tem- Experimental support for the isomorphous tissue re-
perature (a process which yields an ECM analog with aver- placement hypothesis has been based on observations such
age pore diameter of less than 1 ∝m) leads to synthesis of a that, when the ratio in Equation (5) was adjusted to values
scar capsule at the surface of the grafted analog, evidence of much smaller than one (by implanting a rapidly degrading
a barrier to cell migration inside an implant.29 ECM analog, for which td<<ts), the wound healing process
resulted in contraction and synthesis of scar (as would have
Critical Residence Time of Template been the case if the template was missing). Furthermore,
The residence time of a template is also bounded. A when the ratio in Equation (5) was much larger than one 1
template has to stay in place long enough to induce the ap- (by implanting an ECM analog which degraded very slowly,
propriate synthetic processes to take place; soon after that, so that td<<ts) the ECM analog was surrounded by a cap-
it must disappear in a timely fashion so as not to interfere sule of scar tissue.29,31 Clearly, the available limited evidence
with these same processes which it induces. The time pe- cannot be used to test the hypothesis of Equation (5) con-
riod necessary to induce synthesis is roughly equal to that clusively; nevertheless, such evidence is, at the least, consis-
required to complete the wound healing process at that ana- tent with a template half life which is bounded. Studies of
tomical site. (In general, the rate of wound healing is quite inhibition of wound contraction by several ECM analogs
different in tissues such as, say, the dermis and the sciatic with defined structure have provided direct experimental
nerve.) Since the template is an insoluble (and, therefore, support for this conclusion. These studies have shown that,
nondiffusible) three dimensional network it follows that cells of several ECM analogs studied, the dermal regeneration
which are bound on it become immobilized and their mi- template was the analog which degraded at a rate corre-
gration is, accordingly, arrested. Not only cells are prevented sponding to a half life of about 1.5-2 weeks; ECM analogs
from migrating to locations which are appropriate for syn- which degraded at much slower or much faster rates were
thesis of a new organ but, in addition, the laying down of not active.9
newly synthesized ECM by the cells in the space of the wound From the above it can be inferred that the simplest
bed is probably blocked physically by the presence of the template structure which can degrade and diffuse away with
template. It follows that the persisting insolubility of the tem- minimum harm to the host is one in which the template
plate will increasingly interfere with the synthesis of the new undergoes degradation by enzymes of the wound bed to
organ at that site. To avoid this eventuality, the template is nontoxic low molecular weight fragments.31
required to become diffusible (by degradation to small mo-
lecular fragments) and thereby disappear from the wound
In Vivo Synthesis of Organs Using Collagen-GAG Copolymers 575
Template Chemical Composition sity of covalent bonds between collagen chains and GAG
Developmentally significant interactions are known molecules, i.e., to form a collagen-GAG graft copolymer.34
to involve cells, growth factors and ECM components. The There is evidence that an increase in the fraction of GAG in
latter include the collagens, elastin and several proteoglycans the copolymer increases the resistance of the macromolecu-
as well as cell adhesion molecules such as fibronectin and lar network to degradation by mammalian collagenases3
laminin. Since development and induced regeneration have Such resistance also increases with the density of collagen-
a common end point we will assume quite simply, as a first collagen crosslinks and collagen-GAG crosslinks.29,35 A re-
approximation, that the required cell-matrix binding events view of the effect of each of these structural features of the
in each case are similar; if so, the identity of matrix compo- dermal regeneration template on its activity in the inhibi-
nents in each case must also be similar. This presumptive tion of the onset of wound contraction can be made based
similarity between developmental and regenerative mecha- on the published evidence.9,33 This review suggests that the
nisms has been previously referred to briefly in terms of the chemical composition and the detailed pore structure of the
hypothetical rule: Regeneration recapitulates ontogeny;8 dermal regeneration template contribute about equally to
however, we caution to the lack of detailed evidence for such its activity. A similar study of the relation between structure
an identity. In the dermis, as well as in the connective tissue and activity for the nerve regeneration template has not been
of peripheral nerves, type I collagen is present in greatest made.
abundance, whereas the most prominent glycosaminogly-
cans in the dermis are dermatan sulfate and chondroitin Conclusions
6-sulfate; in peripheral nerves, type I collagen and sulfated There is strong evidence that the adult mammal is
proteoglycans have also been prominently observed.32 capable of almost complete organ regeneration. This sur-
The early exudate of a spontaneously healing skin prising result has been documented with several animal
wound is free of ECM components; although quite richly models and in repeated clinical trials. It is clear that both
endowed with undifferentiated cells and growth factors, it the dermis and the epidermis, as well as the peripheral nerve
is, nevertheless, lacking in components which are known to and the knee meniscus, can be induced to regenerate in le-
be required for development.23-25 As pointed out above, this sions where spontaneous regeneration is well known to be
lack of ECM components is hypothetically associated with negligible. In these instances regeneration has been induced
the absence of synthetic processes which lead to a physio- by analogs of the ECM. A model of the mechanism of re-
logical organ. generation provides an explanation for the observed struc-
This article has been based on the author’s article tural features of the ECM analogs (termed regeneration tem-
“Models of Organ Regeneration Processes Induced by Tem- plates) which have shown this remarkable behavior.
plates”, in Bioartificial Organs, Prokop A, Hunkels D,
Cherrington AD, eds. Ann Ny Acad Sci 1997; 831:280-293. References
The reasoning above is consistent with the choice of 1. Billingham RE, Medawar PB. Contracture and intussus-
type I collagen and at least one of the proteoglycans or gly- ceptive growth in the healing of extensive wounds in
cosaminoglycans (GAGs) as basic structural components of mammalian skin. J Anat 1955; 89:114-123.
2. Peacock Jr EE, Van Winkle Jr W. Wound Repair, Second
regeneration templates. Several efforts have been made to
Edition. Philadelphia: W.B.Saunders 1976.
replace the use of ECM analogs in templates with synthetic 3. Yannas IV, Burke JF, Huang C, Gordon PL. Suppression
polymers; at this time, however, there is no firm evidence of in vivo degradability and of immunogencity by reac-
that synthetic polymers can induce regeneration of the der- tion with glycoaminoglycans. Polymer Repr Am Chem Soc
mis or of a peripheral nerve in lesions where the physiologi- 1975; 16:209-214.
cal structures are not regenerated spontaneously. 4. Yannas IV, Burke JF, Gordon PL, Huang C. Multilayer
Considerable experimental evidence links the biologi- membrane useful as synthetic skin. 1977; US Patent
cal activity of the dermal regeneration template to its de- 4,060,081.
tailed structural features. Two ECM analogs, one of which 5. Yannas IV, Burke JF, Umbreit M, Stasikelis P. Progress
was prepared with a GAG while the other was prepared with in design of an artificial skin. Fed Proc 1979; 38:988.
the corresponding proteoglycan, showed the same activity 6. Yannas IV, Burke JF, Warpehoski M, Stasikelis P, Skrabut
EM, Orgill D, Giard DJ. Prompt, long-term functional
in an in vivo assay (inhibition of onset of wound contraction)
replacement of skin. Trans Am Soc Artif Intern Organs
which predicts dermal regeneration.33 This result suggested 1981; 27:19-22.
that the dermal regeneration template can be constructed 7. Yannas IV, Burke JF, Orgill DP, Skrabut EM. Wound tis-
using a GAG, rather than the corresponding proteoglycan, sue can utilize a polymeric template to synthesize a func-
without loss of activity. A covalently crosslinked network of tional extension of skin. Science 1982; 215:174-176.
collagen and the sulfated GAG appears to be necessary; this 8. Yannas IV, Orgill DP, Skrabut EM, Burke JF. Skin regen-
is inferred by the observation that whereas these two mac- eration with a bioreplaceable polymeric template. In:
romolecules form an ionic complex spontaneously at acidic Gebelein CG, ed. American Chemical Symposium Series,
pH, the complex is dissociated at neutral pH, i.e., under con- Number 256 Washington, D.C. 1984:191-197.
ditions which prevail following implantation.34 To preserve 9. Yannas IV, Lee E, Orgill DP, Skrabut EM, Murphy GF.
Synthesis and characterization of a model extracellular
the chemical composition of the ECM analog in vivo over
matrix that induces partial regeneration of adult mam-
the period suggested by the residence time considerations malian skin. Proc Natl Acad Sci USA 1989; 86:933-937.
discussed above, it is necessary to introduce a certain den-
576 Tissue Engineering of Prosthetic Vascular Grafts
10. Murphy GF, Orgill DP, Yannas IV. Partial dermal regen- 22. Stone KR, Webber RJ, Rodkey WG, Steadman JR. Pros-
eration is induced by biodegradable collagen-glycosami- thetic meniscal replacement: In vitro studies of meniscal
noglycan grafts. Lab Invest 1990; 63:305-313. regeneration using copolymeric collagen prostheses.
11. Burke JF, Yannas IV, Quinby Jr WC, Bondoc CC, Jung Arthroscopy 1989; 5:152.
WK. Successful use of a physiologically acceptable artifi- 23. McPherson JM, Piez KA. In: Clark RAF, Henson PM, eds.
cial skin in the treatment of extensive skin injury. Ann The Molecular and Cellular Biology of Wound Repair.
Surg 1981; 194:413-428. New York: Plenum, 1988:471-496.
12. Heimbach D, Luterman A, Burke J, Cram A, Herndon D, 24. Hay ED, ed. Cell biology of extracellular matrix. New
Hunt J, Jordan M, McManus W, Solem L, Warden G, York: Plenum Press, 1981.
Zawacki B. A multi-center randomized clinical trial. Ar- 25. Loomis WF. Developmental biology. New York:
tificial dermis for major burns. Ann Surg 1988; Macmillan, 1986.
208:313-320. 26. Yannas IV. Models of organ regeneration processes in-
13. Stern R, McPherson M, Longaker MT. Histologic study duced by templates. In: Prokop A, Hunkeler D,
of artificial skin used in the treatment of full-thickness Cherrington AD, eds. Ann NY Acad Sci 1997; 831:
thermal injury. J Burn Care Rehabil 1990; 11:7-13. 280-293.
14. Orgill DP, Butler CE, Regan JF. Behavior of collagen-GAG 27. Yannas IV. Regeneration templates. In: Bronzino JD, ed.
matrices as dermal replacement in rodent and porcine The Biomedical Engineering Handbook. Boca Raton: CRC
models. Wounds 1996; 8:151-157. Press, 1995:1619-1635.
15. Yannas IV, Orgill DP, Silver J, Norregaard TV, Zervas NT, 28. Yannas IV. In vivo synthesis of tissues and organs. In:
Schoene WC. Regeneration of sciatic nerve across 15-mm Lanza RP, Langer RS, Chick WL, eds. Textbook of Tissue
gap by use of a polymeric template. In: Gebelein CG, ed. Engineering. New York: R.G. Landes/Academic Press,
Advances in Biomedical Polymers. New York: Plenum 1996:169-178.
Press,1987:1-9. 29. Yannas IV. Use of artificial skin in wound management.
16. Chang AS, Yannas IV, Perutz S, Loree H, Sethi RR, Krarup In: Dineen P, ed. The Surgical Wound. Philadelphia: Lea
C, Norregaard TV, Zervas NT, Silver J. Electrophysiologi- and Febiger, 1981:171-190.
cal study of recovery of peripheral nerves regenerated by 30. Yannas IV, Burke JF, Huang C, Gordon PL. Correlation
a collagen-glycosaminoglycan copolymer matrix. In: of in vivo collagen degradation rate with in vitro mea-
Gebelein CG, ed. Progress in Biomedical Polymers. New surements. J Biomed Mat Res 1975; 9:623-628.
York: Plenum Press, 1990:107-120. 31. Yannas IV, Burke JF. Design of an artificial skin.Part I.
17. Chang AS, Yannas IV. Peripheral nerve regeneration. In: Design principles. J Biomed Mat Res 1980; 14:65-68.
Smith B, Adelman G, eds. Neuroscience Year (Supplement 32. Rutka JT, Apodaca G, Stern R, Rosenblum M. The extra-
2 to the Encyclopedia of Neuroscience). Boston: cellular matrix of the central and peripheral nervous sys-
Birkhauser, 1992:125-126. tems: Structure and function J Neurosurg 1988;
18. Landstrom A, Yannas IV. Peripheral nerve regeneration. 69:155-170.
In: Smith B, Adelman G, eds. Encyclopedia of Neuro- 33. Shafritz TA, Rosenberg LC, Yannas IV. Specific effects of
science. Boston: Birkhaüser, 1996: In press. glycosaminoglycans in an analog of extracellular matrix
19. Lundborg G, Dahlin LB, Danielson N, Gelberman RH, that delays wound contraction and induces regeneration.
Longo FM, Powell HC, Varon S. Nerve regeneration in Wound Rep Reg 1994; 2:270-276.
silicone chambers: Influence of gap length and of distal 34. Yannas IV, Burke JF, Gordon PL, Huang C, Rubenstein
stump components. Exp Neurol 1982; 76:361-375. RH. Design of an artificial skin. Part II. Control of chemi-
20. Lundborg G. Nerve regeneration and repair: A review. cal composition. J Biomed Mat Res 1980; 14:107-131.
Acta Orthop Scand 1987; 58:145-169. 35. Yannas IV. Regeneration of skin and nerves by use of
21. Stone KR, Rodkey WG, Webber RJ, McKineey L, collagen templates. In: Nimni M, ed. Collagen: Biotech-
Steadman JR. Collagen-based prostheses for meniscal re- nology, Vol. III. Boca Raton: CRC Press, 1988:87-115.
generation. Clin Orthop 1990; 252:129-135.
Matrix Engineering
CHAPTER 53
Artificial Extracellular Matrix Proteins
for Graft Design
Alyssa Panitch, David A. Tirrell
Introduction
ore than 500,000 vascular grafts are implanted annually in the United States.1 Although
M large diameter grafts implanted in regions of high blood flow remain patent for many
years, small and medium caliber prostheses are plagued by unacceptable rates of failure due
to thrombosis and intimal hyperplasia.2
Despite their high failure rates, two materials—expanded polytetrafluoroethylene
(ePTFE) and poly(ethylene terephthalate) (Dacron)—have remained dominant in the tech-
nology of synthetic vascular grafts for more than thirty years. In attempts to reduce leakage,
platelet adhesion, thrombosis and intimal hyperplasia, researchers have modified the sur-
faces of ePTFE and Dacron in many ways, including impregnation with albumin,3 gelatin4
or collagen,5 preclotting with whole blood,6 pretreatment with fibrin glue with or without
growth factors or heparin,7 and preseeding with endothelial cells, either in the operating
room8 or in a preliminary procedure that allows preestablishment of a confluent endothe-
lial cell monolayer.9 Recent reports on clinical trials using endothelialized ePTFE grafts have
been particularly encouraging.10
Nevertheless, the factors leading to the development of thromboresistant,
endothelialized grafts free of intimal hyperplasia remain poorly understood. It is clear that a
confluent layer of endothelial cells is not sufficient; although the endothelial lining of the
healthy artery resists thrombus formation, a procoagulant state can be induced by mechani-
cal trauma, by inflammatory mediators such as interleukin-1,11 by bacterial endotoxin,12
and by other stimuli. Furthermore, endothelial cells are capable of releasing platelet-derived
growth factor-like proteins13-15 and other smooth muscle cell mitogens, and elevated mi-
totic activity in smooth muscle cells located beneath an apparently confluent endothelium
has been reported by Clowes and co-workers in studies of ePTFE grafts implanted in ba-
boons.16 Thus, even confluent endothelial layers can play an active role in graft failure via
thrombosis or intimal hyperplasia.
Our laboratory has adopted a new approach to the problem of preparing vascular
grafts of improved long term patency, an approach based on the synthesis of new artificial
extracellular matrix (ECM) proteins suitable as substrates for the culture of endothelial cells.
The proteins under current study are based on repetitive oligopeptide sequences analogous
to those of mammalian elastin,17 and incorporate ECM-derived cell binding domains, in-
cluding the CS5 region of fibronectin,18 which contains the REDV sequence previously shown
to support attachment and spreading of endothelial cells, but not vascular smooth muscle
cells.19 The potential endothelial cell selectivity of such polymers makes them attractive
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
578 Tissue Engineering of Prosthetic Vascular Grafts
candidates for use in vascular grafts, and it is the prospect of faces on which HUVECs attached and spread, while human
using such materials to culture thromoresistant endothelial foreskin fibroblasts, human vascular smooth muscle cells,
tissue that has attracted our attention. and human blood platelets did not. Subsequent investiga-
The choice of elastin-like sequences to constitute the tions demonstrated that endothelial cell attachment to
structural elements of the artificial ECM proteins was based REDV-grafted substrates is mediated by integrin #4!1,37 and
on several factors. First, elastin is abundant in the walls of that attachment can be inhibited specifically by soluble
small muscular arteries,20 and thus is a critical element in REDV peptides. Endothelial cells also recognize the RGD
the organization and function of the healthy artery. Second, sequence from the central cell-binding domain of fibronectin
Urry and co-workers have demonstrated a remarkably wide (via integrins #5!1 and #v!1), and fibronectin carries at least
range of mechanical properties in repetitive elastin-like poly- two additional recognition sites for #4!1, designated CS1 and
mers, reporting elastic moduli from 104 to more than 108 H1, respectively.34 The integrin binding affinities of CS5 and
dynes/cm2, depending on sequence and water content.17 H1 are comparable, while that of CS1 is approximately
Because the moduli of the arterial wall are of the same or- 20-fold higher.34
der,21 elastin-like polymers offer the prospect of reduced Thus it appears likely that endothelial cell function
compliance mismatch between artery and graft, a recurrent can be regulated by presentation of ECM-derived recogni-
cause of endothelial damage, peri-anastomotic smooth tion sequences, with control exercised by variation in the
muscle cell proliferation, and graft failure.22-25 Elastin-like density and affinity of recognition sites and in the presenta-
polymers are readily processed from aqueous solutions, and tion of multiple sites in combination. The design of artifi-
can be converted in simple operations into films, tubes, and cial ECM proteins allows this approach to be pursued in sys-
surface coatings convenient for evaluation of materials prop- tematic fashion, in that specific recognition sequences, one
erties.26 Finally, the polypentapeptide of repeating unit se- by one or in combination, can be built into structural do-
quence -GVGVP- has been subjected to extensive mains denuded of their intrinsic binding sites. We have be-
biomaterials testing, including determination of mutagenic- gun to test this idea, beginning with elastin-like structural
ity (Ames test), toxicity (mice and rabbits), antigenicity domains, and introducing initially CS5, and subsequently
(Guinea pigs), pyrogenicity (rabbits), and thrombogenicity CS1, as ligands for the #4!1 integrin receptor on the endot-
(dogs), and has performed well in each of these tests.27 helial cell surface.
Poly(GVGVP) is nonadhesive toward human umbilical vein
endothelial cells (HUVECs),28 suggesting that cell attach- Artificial Proteins
ment to such polymers ought to be mediated by specific in- Since 1990, the primary focus of our laboratory has
teractions with recognition sequences built into the chain. been the design and bacterial synthesis of artificial proteins
Urry and co-workers have reported adhesion of HUVECs that exhibit novel and potentially useful material proper-
to variants of poly(GVGVP) that contain periodic GRGDS ties. Our initial efforts addressed strategies for controlling
inserts.28 The same group reported that incorporation of chain folding and supramolecular organization in solid poly-
REDV oligopeptides into poly(GVGVP) did not yield sub- mers, and led to a family of repetitive proteins that adopt
strates capable of supporting adhesion of HUVECs. As chain-folded lamellar architectures of controlled dimensions
shown in the following section, however, insertion of the and surface functionality.38-41 A second set of experiments
longer (20 amino acid) CS5 region does yield adhesive sub- has been directed toward the engineering of novel liquid
strates. Perhaps the short REDV inserts in poly(GVGVP) crystal (LC) phases in solutions of rod-like artificial pro-
are unable to adopt the conformation(s) required for cell teins,42 and has yielded unique smectic LC structures with
surface recognition and attachment. layer spacings subject to precise control on length scales of
The cell-binding domain of primary interest in our tens of nanometers.43 An important, continuing objective
work to date has been the CS5 region of human fibronectin. of this work has been the development of methods for in-
Comprising residues 90-109 of the type III connecting seg- corporating nonnatural amino acids into artificial proteins
ment (IIICS) of fibronectin, CS5 includes the minimal ac- in vivo, and good success has been achieved for selenated,44
tive sequence REDV,18 which is recognized by the integrin fluorinated,45 electroactive,46 olefinic,47 acetylenic and con-
#4!1.29-33 Binding to IIICS appears to be considerably more formationally constrained48 amino acid analogs.
selective than is recognition of the central cell-binding do-
main of fibronectin,34 which carries the cell recognition se- Artificial ECM Proteins
quence RGDS.35,36 In our preliminary experiments, two artificial ECM
Hubbell and co-workers have exploited the selectivity proteins (1a and 1b, see Fig. 53.1) have been prepared and
of the REDV sequence to prepare surfaces that show prefer- subjected to evaluation as substrates for the culture of hu-
ential affinity for endothelial cells.19 Immobilization of the man umbilical vein endothelial cells (HUVECs).
hexapeptide GREDVY on glycophase glass produced sur-
monolayers, with typical cobblestone appearance, within 5. Freischlag JA, Moore WS. Clinical experience with a col-
48 h. lagen-impregnated knitted dacron vascular graft. Ann Vasc
Preliminary measurements of competitive inhibition Surg 1990; 4:449-454.
of EC attachment by soluble CS5 analogs have also been 6. Yates SG, Barros-D’Sa ABB, Berger K et al. The preclotting
of porous arterial prostheses. Ann Surg 1978; 188:611-622.
made. Preincubation of HUVECs with the soluble peptide
7. Gray JL, Kang SS, Zenni GC et al. FGF-1 affixation stimu-
GREDVDY (Research Genetics, Inc.) at concentrations up lates ePTFE endothelialization without intimal hyperpla-
to 2 mg/ml has no observable effect on attachment to sia. J Surg Res 1994; 57:596-612.
fibronectin, as expected. In contrast, attachment to 1b is 8. Williams SK. Endothelial cell transplantation. Cell Trans-
completely inhibited by GREDVDY at concentrations of plantation 1995; 4:401-410.
0.6-0.8 mg/ml, consistent with specific attachment of 9. Zilla P, Deutsch M, Meinhart J et al. Clinical in vitro
HUVECs to CS5 via the #4!1 receptor. endothelialization of femoropopliteal bypass graft: An ac-
tuarial follow-up over three years. J Vasc Surg 1994;
Current and Future Directions 19:540-548.
Our overall objective is the development of a new class 10. Zilla P, Deutsch M, Meinhart M, Fischlein T, Hofmann
of artificial ECM proteins to be used in small diameter vas- G. Long-term effects of clinical in vitro endothelialization
on grafts. J Vasc Surg 1997; 25:1110-1112.
cular grafts of improved long term patency. We believe the
11. Bevilacqua MP, Pober JS, Majeau GR et al. Recombinant
proposed approach to be novel in that genetic engineering, tumor necrosis factor induces procoagulant activity in
polymer chemistry, and materials processing methods can cultured human vascular endothelium. Characterization
be combined to provide control of both the mechanical and comparison with the actions of interleukin I. Proc
properties and the cell-surface interactions of the new pro- Nat Acad Sci USA 1986; 83:4533.
teins. Mechanical properties can be engineered through 12. Colucci M, Balcon GI, Lorenzet R. Cultured human en-
variation in the length of the elastin-like domains, control dothelial cells generate tissue factor in response to endo-
of the overall chain length, and introduction of a controlled toxin. J Clin Investi 1983; 71:1893.
level of interchain crosslinking. Cell-surface interactions are 13. DiCorleto PE, Bowen-Pope D. Cultured endothelial cells
to be regulated through presentation of recognition sites for produce a platelet-derived growth factor-like protein. Proc
membrane-bound receptors on the target endothelial cell Nat Acad Sci USA 1983; 80:1919-1923.
14. Fox P.L, DiCorleto PE. Regulation of production of a
surfaces, and through control of the identity and frequency
platelet-derived growth factor-like protein in cultured bo-
of protein-bound recognition sites. In concert with comple- vine aortic endothelial cells. J Cell Physiol 1984;
mentary methods described elsewhere in this volume, arti- 121:298-308.
ficial ECM proteins may provide a route to compliant, 15. Ross R, Raines E, Bowen-Pope D. Growth factors from
thromboresistant grafts subject to resorption and remodel- platelets, monocytes, and endothelium: Their role in cell
ing, and ultimately may play a role in defining strategies for proliferation. Ann NY Acad Sci 1982; 397:18-24.
regeneration of healthy vascular tissue. 16. Clowes AW, Kirkman RD, Reidy MA. Mechanism of ar-
terial graft healing: Rapid transmural capillary ingrowth
References provides a source of intimal endothelium and smooth
1. Langer RW, Vacanti J. Tissue engineering. Science 1993; muscle in porous PTFE prostheses. Am J Pathol 1986;
260:920-926. 123:220-230.
2. Greisler HP. New Biologic and Synthetic Vascular Pros- 17. Urry D.W, Luan C-H, Harris CM et al. Protein-based
theses. Austin, TX: R.G. Landes Co., 1991. materials with a profound range of properties and appli-
3. Rumisek JD, Wade CE, Brooks DE et al. Heat-denatured cations: The elastin )T t hydrophobic paradigm.
albumin-coated dacron vascular grafts; physical charac- In:McGrath, K, Kaplan, D, eds. Protein-Based Materials.
teristics and in vivo performance. J Vasc Surg 1986; Boston, MA:Birkhauser, 1997.
4:136-143. 18. Humphries JJ, Akiyama SK, Komoriya A et al. Identifica-
4. Drury JK, Ashton TR, Cunningham JD et al. Experimen- tion of an alternatively spliced site in human plasma
tal and clinical experience with a gelatin impregnated fibronectin that mediates cell type-specific adhesion. J Cell
dacron prosthesis. Ann Vasc Surg 1987; 1:542-547. Biol 1986; 103:2637-2647.
Artificial Extracellular Matrix Proteins for Graft Design 581
19. Hubbell JA, Massia SP, Desai NP et al. Endothelial cell- 36. Yamada KM, Kennedy DW. Dualistic nature of adhesive
selective materials for tissue engineering in the vascular protein function: Fibronectin and its biologically active
graft via a new receptor. Biotechnology 1991; 9:568-572. peptide fragments can autoinhibit fibronectin function. J
20. Ross MH, Romrell LJ, Kaye GI. Histology: A Text and Cell Bio 1984; 99:29-36.
Atlas. 3rd ed. Baltimore, MD:Williams & Wilkins, 1995. 37. Massia SP, Hubbell JA. Vascular endothelial cell adhesion
21. Chandran KB. Cardiovascular Biomechanics. New York: and spreading promoted by the peptide REDV of the
University Press, 1992. IIICS region of plasma fibronectin is mediated by integrin
22. Greisler HP, Joyce KA, Kim DU et al. Spatial and tempo- #4!1. J Biol Chem 1992; 267(20):14019-14026.
ral changes in compliance following implantation of 38. Krejchi MT, Atkins EDT, Waddon AJ et al. Chemical se-
bioresorbable vascular grafts. J Biomed Mat Res 1992; quence control of !-sheet assembly in macromolecular
26:1449-1461. crystals of periodic polypeptides. Science 1994;
23. Kinley CE, Marble AE. Compliance: A continuing prob- 265:1427-1432.
lem with vascular grafts. J Cardiovasc Surg 1980; 39. Parkhe A, Fournier MJ, Mason TL et al. Determination
21:163-170. of the chain folding pattern in the crystalline domains of
24. Zwolak RM, Adams MC, Clowes AW. Kinetics of vein the repetitive polypeptide {(AlaGly) 3GluGly(GlyAla) 3
graft hyperplasia: Association with tangential stress. J Vasc GluGly}10 by FTIR studies of its blends with a 13C en-
Surg 1987; 5:126-136. riched analogue. Macromolecules 1993; 26:6691-6693.
25. Clowes AW, Reidy MA, Clowes MM. Kinetics of cellular 40. McGrath KP, Fournier MJ, Mason TL et al. Genetically-
proliferation after arterial injury. Smooth muscle growth directed syntheses of new polymeric materials. Synthesis
in the absence of endothelium. Lab Invest 1983; and expression of a family of artificial genes encoding
49(3):327-333. proteins with repeating -(AlaGly)3ProGluGly- elements. J
26. Urry DW, Nicol A, McPherson DT et al. Properties, Am Chem Soc 1992; 114:727-733.
preparations and applications of bioelastic materials. 41. Creel HS, Fournier MJ, Mason TL et al. Genetically-di-
In:Handbook of Biomaterials and Applications. New rected syntheses of new polymeric materials. Efficient ex-
York:Marcel Dekker, 1995. pression of a monodisperse copolypeptide containing
27. Urry DW, Parker TM, Reid MC et al. Biocompatibility of fourteen tandemly repeated -(AlaGly)4ProGluGly- ele-
the bioelastic materials, poly(GVGVP) and its &-irradia- ments. Macromolecules 1991; 24:1213-1214.
tion cross-linked matrix: Summary of generic biological 42. Zhang G, Fournier MJ, Mason TL et al. Biosynthesis of
test results. J Bioac Comp Poly (1991); 6:263-282. monodisperse derivatives of poly( # ,L-glutamic acid):
28. Nicol A, Gowda DC, Parker TM et al. Cell adhesive prop- Model rod-like polymers. Macromolecules 1992;
erties of bioelastic materials containing cell attachment 25:3601-3603.
sequences. In:Gebelein, C & Carraher, C, eds. Biotechnol- 43. Yu SM, Conticello V, Kayser C et al. Smectic ordering in
ogy and Bioactive Polymers:Plenum Press, NY, 1994. solutions and films of a monodisperse derivative of poly(&-
29. Wayner E.A, Garcia-Pardo A, Humphries MJ et al. Iden- benzyl #,L-glutamate). Nature 1997; 389:167-170.
tification and characterization of the T lymphocyte adhe- 44. Dougherty MJ, Kothakota S, Mason TL et al. Synthesis of
sion receptor for an alternative cell attachment domain a genetically engineered repetitive polypeptide containing
(CS-1) in plasma fibronectin. J Cell Biol 1989; periodic selenomethionine residues. Macromolecules 1993;
109(3):1321-1330. 26:1779-1781.
30. Garcia-Pardo A, Wayner EA, Carter WG et al. Human B 45. Yoshikawa E, Fournier MJ, Mason TL et al. Genetically
lymphocytes define an alternative mechanism of adhesion engineered fluoropolymers. Synthesis of a repetitive
to fibronectin. The interaction of the #4!1 integrin with polypeptide containing p-fluorophenylalanine residues.
the LHGPEILDVPST sequence of the type III connecting Macromolecules 1994; 27:5471-5475.
segment is sufficient to promote cell attachment. J 46. Kothakota S, Mason TL, Fournier MJ et al. Biosynthesis
Immunol 1990; 144:3361-3366. of a periodic protein containing 3-thienylalanine: A step
31. Guan J-L, Hynes RO. Lymphoid cells recognize an alter- toward genetically engineered conducting polymers. J Am
natively spliced segment of fibronectin via the integrin Chem Soc 1995; 117:536-537.
receptor #4!1. Cell 1990; 60:53-60. 47. Deming TJ, Fournier MJ, Mason TL et al. Biosynthetic
32. Mould AP, Wheldon LA, Komoriya A et al. Affinity chro- incorporation and selective chemical modification of ole-
matographic isolation of the melanoma adhesion recep- finic functionality in genetically engineered polymers. J
tor for the IIICS region of fibronectin and its identifica- Macromol Sci-Pure Appl Chem 1997; A34(10):2143-2150.
tion as the integrin # 4 ! 1 . J Biol Chem 1990; 48. Deming TJ, Fournier MJ, Mason TL et al. Structural
265(7):4020-4024. modification of a periodic polypeptide through biosyn-
33. Mould AP, Komoriya A, Yamada KM et al. The CS5 pep- thetic replacement of proline with azetidine-2-carboxylic
tide is a second site in the IIICS region of fibronectin acid. Macromolecules 1996; 29(5):1442-1444.
recognized by the integrin #4!1. Inhibition of #4!1 func- 49. Studier FW, Rosenberg AH, Dunn, AH et al. Use of T7
tion by RGD peptide homologues. J Biol Chem 1991; RNA polymerase to direct expression of cloned genes.
266(6):3579-3585. Meth Enzymol 1991; 185:60-68.
34. Mould AP, Humphries MJ. Identification of a novel rec- 50. Urry DW, Gowda C, Parker TM et al. Hydrophobicity
ognition sequence for the integrin #4!1 in the COOH- scale for proteins based on inverse temperature transitions.
terminal heparin-binding domain of fibronectin. EMBO Biopolymers 1992; 32:1243-1250.
J 1991; 10(13):4089-4095.
35. Pierschbacher MD, Ruoslahti E. Cell attachment activity
of fibronectin can be duplicated by small synthetic frag-
ments of the molecule. Nature 1984; 309:30-33.
Matrix Engineering
CHAPTER 54
Cell-Extracellular Matrix Interactions Relevant
to Vascular Tissue Engineering
Stephen P. Massia
Introduction
T he interaction between cells and extracellular surfaces plays a major role in determining
cellular behavior in tissues and on biomaterials. These interactions modulate many aspects
of cell behavior, including adhesion, spreading, migration, proliferation, differentiation, and
metabolism. In the fields of biomaterials and tissue engineering, a fundamental understanding
of cell surface interactions is critical for developing new methods that precisely control cell
interactions at the cell-biomaterial interface. Enhancement of tissue cell adhesion to im-
planted biomaterials would facilitate tissue adhesion/integration at the tissue-biomaterial
interface. In contrast, the reduction of cell adhesion to implanted biomaterials is desirable
for limiting cell-mediated events such as thrombosis and macrophage activation on bioma-
terial surfaces, which often result in biomaterial implant failure. It is also desirable for tissue
engineered implants to promote specific cell-biomaterial interactions which facilitate tissue
regeneration/repair. Tissue engineered devices in the form of prefabricated biohybrid tissue
constructs require the appropriate interactions of transplanted cells with biomaterial scaf-
folds to initiate extracellular matrix (ECM) synthesis and deposition as well as other activi-
ties which promote biohybrid tissue formation in vitro prior to implantation in vivo.
Two major approaches for developing materials which elicit precisely controlled cel-
lular interactions have emerged. One involves fabricating implants, scaffolds, etc. from natural
biomaterials such as collagen where the base material has intrinsic and specific biological
activities which influence cellular interactions. The major disadvantages of using natural
biomaterials is that they generally have a largely unmodifiable and limited range of physico-
chemical properties and can be difficult to isolate and characterize. The second approach
for improving the control of cell-material interactions is to use synthetic materials which
have surface immobilized, biologically active molecules and biologically active surface
microtextrue. This methodology takes advantage of the inexpensive manufacturing costs
for synthetic material-based devices and exploits specific biological activities to modulate
cellular interactions for improved biocompatibility and biohybrid tissue formation. Essen-
tially, these types of surface modifications on synthetic implant materials are attempting to
create a biomimetic surface with surface physicochemical properties that more closely match
those of the ECM in the tissues where the synthetic material will be implanted. This review
will focus on the biology of cell-ECM interactions and how they are utilized in developing
biomimetic surfaces on synthetic biomaterials. The role of these technologies in developing
vascular implants with improved biocompatibility will also be discussed.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
584 Tissue Engineering of Prosthetic Vascular Grafts
integrins. Integrins are a superfamily of transmembrane gly- tein signaling complexes can form within adhesion plaques
coprotein adhesion receptors consisting of a noncovalently and are associated with signal transduction activities.
linked # and ! subunit.10-13 At least 17 # and 8 ! subunits Models of the structural architecture of cytoskeletal
have been described, with resulting combinations of over complexes, which form on the cytoplasmic side of focal ad-
21 functional integrins with varying ligand specificity.13 hesions, have been developed based on in vitro protein bind-
Integrins bind to ECM proteins via cell binding domains, ing assays (Fig. 54.2). These models show how proteins may
and rather small polypeptide sequences within these do- link with each other within cytoskeletal complexes. These
mains promote integrin binding with an affinity that is nearly structural arrangements are considered models because
as high as in the intact molecule.10-13 The tripeptide RGD is many of the protein-protein linkages have not been dem-
a ubiquitous recognition sequence that binds to most onstrated in living cells.20 Talin is a major protein of
integrins and is found within the cell binding domain of cytoskeletal complexes and was the first reported integrin-
many cell adhesion ECM proteins.10-13 Binding of integrins binding cytoplasmic protein.21 Talin also binds to actin22 and
to RGD and other related recognition sequences results in a to vinculin.23 Vinculin apparently serves as a structural hub
cascade of events that promotes cell-matrix adhesion inter- for cytoskeletal complexes, since it can bind to many other
actions that are stable in stationary cells, e.g., epithelial cells cytoskeletal complex proteins. Studies have shown that
on basement membranes, and are transient in motile, mi- vinculin can bind to #-actinin,24 paxillin,25 tensin,26 and
gratory cells. Although this review will focus on integrin- possibly actin via a cryptic binding site.27 #-actinin, which
ECM interactions, there are many nonintegrin cell-ECM crosslinks actin microfilaments to form microfilament
binding receptors including cell surface proteoglycans bundles, has been shown to directly bind to the cytoplasmic
(syndecans),14 hyaluronan receptors (CD44, RHAMM and tails of integrin ! subunits.28 With at least two integrin-bind-
ICAM-1,15 67 kDa laminin-binding proteins,16 annexin II,17 ing cytoplasmic proteins (talin and #-actinin) and other
and receptor type protein tyrosine phosphatase !.18 various interconnecting proteins, e.g., vinculin, an indirect
linkage between ECM-bound integrins and the cytoskeleton
Integrin-Mediated Cytoplasmic Activities is formed via cytoskeletal complexes.
Following ligand-binding, integrins undergo confor- Unlike many other types of transmembrane cell sur-
mational changes, cluster into aggregates, and form adhe- face receptors, the cytoplasmic domains of integrins do not
sion plaques or focal contacts. It is well documented in many have enzymatic activity. Therefore, integrin-mediated intra-
studies that integrin clustering is required for initiating in- cellular signal transduction depends on recruited, enzymati-
tracellular activities.13,19 Integrin-mediated cytoplasmic cally active signal transduction molecules. Integrin cluster-
functions within adhesion plaques can be categorized into ing has been shown to induce phosphorylation of tyrosine
two experimentally separable types: residues of a number of proteins, even though integrins do
1. Cytoskeletal organization and establishment of me- not intrinsically have tyrosine kinase activity.29 One protein
chanical linkages between ligand-bound integrins that is rapidly phosphorylated and localized to focal adhe-
and cytoskeletal structures; and sions is focal adhesion kinase (FAK or pp125FAK).30,31 The
2. Tyrosine kinase-mediated signal transduction. exact mechanism for clustered integrin-mediated FAK phos-
Associated with integrin-cytoskeleton linkages are phorylation is not known. Studies demonstrating direct
cytoskeletal complexes that consist of clustered integrins, binding of FAK to the !1 subunit cytoplasmic domain32 sug-
several cytoplasmic proteins, and actin microfilament gest that changes in integrin aggregation and/or conforma-
bundles. Structurally and biochemically distinct multi-pro- tion alters FAK to promote autophosphorylation, which
upregulates the enzymatic activity of FAK and triggers
further downstream signal transduction phosphorylation influence a broad range of cellular activities beyond cell ad-
events.33 Activated FAK phosphorylates many focal contact hesion (Fig. 54.3).
proteins, which generally induces them to form protein-pro-
tein bonds and facilitate multi-protein complex formation.34 Biomimetic Cell-Adhesive Biomaterials
Similar to models of cytoskeletal complexes described in the
previous paragraph, models for an integrin-mediated sig- Controlling Cell Adhesion on Biomaterials
nal transduction complex have been developed from in vitro For both tissue engineers and biomaterial scientists,
studies. Following integrin-mediated autophosphorylation, the precise control of cell interactions with biomaterials is
FAK phosphorylates paxillin, which activates its SH2 and desirable. The tissue engineer requires the interaction of
SH3 (Src homology 2 and 3 binding domains. The signal specific cell types with materials in implants and devices
transducing tyrosine kinase c-Csk readily binds to the designed for tissue ingrowth and regeneration. In biohybrid
paxillin SH2 domain35 and c-Src binds to SH3.36 GRB-2, tissue constructs, the tissue engineer seeds specific cells in
which promotes MAP kinase activation, has been shown to biomaterial scaffolds in vitro and requires cell-biomaterial
bind to phosphorylated FAK and to c-Src.37 This finding interactions which promote appropriate ECM deposition
suggests that GRB-2 can associate with focal adhesion sig- and assembly for tissue formation. The biomaterial scien-
nal transduction complexes to couple MAP kinase-medi- tist requires selective attachment of tissue cells to implanted
ated signal transduction to integrins. In growth factor-me- materials so that tissue adhesion and integration is optimized
diated signal transduction pathways, MAP kinase has been and inflammatory cell-mediated encapsulation is limited at
shown to translocate to the nucleus and play a direct role in the tissue-biomaterial interface. In order to achieve high
gene regulatory pathways.38 Similar signaling mechanisms precision control of cell adhesive and functional responses
may occur in integrin signal transduction complexes. Other to biomaterials, protein adsorption has to be precisely con-
studies have shown that a G protein & subunit and protein trolled as well. Protein adsorption is an exceedingly com-
kinase c type 3 (PKC type 3 ) bind and localize to focal ad- plex phenomenon, and, correspondingly, it is very difficult
hesions, suggesting that activated integrins possibly utilize to control.44-46 The main difficulty is in controlling the speci-
G protein and PKC type 3-mediated signal transduction ficity of protein adsorption, e.g., it is difficult to selectively
pathways coordinately with or independently of tyrosine promote the adsorption of cell adhesion proteins over the
kinase pathways.39,40 Integrin binding has also been shown adsorption of nonadhesive proteins. A second difficulty with
to promote signal transduction-associated events such as the adsorbed protein layer is its lack of stability to denatur-
increased cytoplasmic levels of PIP241 and Ca2+,42 as well as ation and proteolysis, which may affect the long term per-
increased cytoplasmic pH.43 Integrin signal transduction formance of implanted biomaterials and tissue engineered
complexes potentially provide a mechanism where integrin- devices. 44-46 Therefore, a material surface that promotes cell
mediated signaling could be transmitted to the nucleus and adhesion and spreading independently of protein adsorp-
invoke specific changes in gene transcription patterns that tion would be more controllable and beneficial in designing
tissue engineered biomaterials. Essentially, this material sur-
face would mimic physicochemical features of tissue ECM Surface Immobilized Biologically
adequately enough to intrinsically promote cell interactions, Active Molecules on Biomaterials
circumventing the requirement for adsorbed ECM proteins. Many methodologies for designing biomimetic, ad-
hesion-promoting materials have been developed to circum-
Mimicking the Extracellular Matrix vent the requirement for protein adsorption by covalently
on Biomaterial Surfaces supplying the substrates with stable, synthetic ligands for
The extracellular matrix (ECM) of soft tissues is a cell surface adhesion.47-82 These adhesion ligand peptide-
complex composite material consisting of dense fibrillar grafted substrates have been utilized as simplified models to
cable-like structures (e.g., type I collagen fibers) for rigid investigate molecular aspects of cell-ECM interactions. One
mechanical support and a loose, more compliant gelatinous of the earliest described methods developed polyacrylamide
interstitial matrix consisting of glycoproteins, proteoglycans, substrates with surface-immobilized RGD peptides47 and
and other components which provide a protective cushion quantitatively evaluated tumor cell haptotactic responses to
against harmful external mechanical forces. The complex surface concentration gradients of immobilized RGD pep-
three dimensional arrangement of fibrillar and interstitial tides.48 Silanized glass substrates containing surface immo-
ECM components provides topographical and biochemical bilized adhesion peptides were developed and utilized to
cues for cell orientation and guidance in tissues. Therefore, quantitatively determine the minimal density and spacing
the surfaces of synthetic biomaterials could potentially of surface immobilized RGD peptides for maximal cell
mimic the ECM if biochemical and biologically relevant to- spreading, cytoskeletal development, and focal contact for-
pographical cues were incorporated into the material sur- mation.51,55 Although complete cell spreading was observed
faces (Fig. 54.4). Biochemical cues could be mimicked using at a minimal RGD surface concentration of 1 fmol/cm2, fo-
surface immobilized biologically active molecules, and cal contacts were scarce and the cytoskeletal structures were
biomimetic topographical cues could be provided by high poorly developed. When the RGD surface concentration was
precision surface microtexturing. increased to 10 fmol/cm2, focal contacts and a fully devel-
oped cytoskeleton was observed in all spread cells.55 These
results suggested that RGD is a sufficient signal for promot- immobilized biological molecule is the predominant deter-
ing a complete and maximal cell adhesive response when minant for cellular responses to these materials.
the ligand is tethered to the material surface. Consequently,
cell surface receptor interactions with tethered RGD ligands Biomaterial Surface Microtexturing to Mimic ECM
generated the contractile force necessary to produce nor- Topography in Tissues
mal focal contact and cytoskeleton formation. The value of The cellular response to attachment substrate topog-
10 fmol/cm2 required for maximal cell spreading, focal con- raphy was first described in 1912 where the migration of
tact, and cytoskeleton formation corresponded to an aver- cultured cells on spider webs was observed to be guided by
age spacing of 140 nm between RGD ligands, or 10 integrin spider web filaments.84 More studies confirmed that surface
widths. It was also observed that covalently immobilized topography influenced cell orientation on solid substrates
RGD peptide promotes cell adhesion and spreading, pre- and the term “contact guidance” was introduced to describe
dominantly via integrin #v!3.55 It was suggested in these stud- this phenomenon.85 Surface topography became an issue in
ies that covalent immobilization of RGD peptide rendered soft tissue biomaterial implant biocompatibility studies be-
it conformationally constrained with highest affinity for cause many researchers have demonstrated a positive corre-
integrin #v!3. Other studies which demonstrated preferen- lation between capsule thickness (biocompatibilty) and tis-
tial binding of cyclic, conformationally constrained RGD sue-implant interfacial motion.86 It was realized that sur-
peptides to integrin #v!3 supported this hypothesis.83 Simi- face topography could be utilized to promote tissue inter-
lar peptide-grafted glass substrates were utilized to charac- digitation or anchorage to reduce interfacial motion. An early
terize nonintegrin interactions to the laminin-based cell study evaluated the effect of surface topography on soft tis-
adhesion ligand YIGSR.63 Cell attachment, spreading, and sue responses to synthetic materials in vivo by implanting
stress fiber formation were observed on these substrates and filter membranes with different uniform pore sizes subcu-
were mediated by the 67 kDa laminin receptor independently taneously in rats and evaluating the tissue response at 6 weeks
of integrins. The 67 kDa receptor was also observed to postimplantation. Excellent tissue attachment was observed
colocalize with vinculin and #-actinin in focal adhesions.63 without signs of inflammation when pore sizes were 1-3 ∝m.
Other studies of cell interactions with peptide-grafted glass Pore sizes above and below this range exhibited symptoms
substrates compared cell migration on glass versus peptide- of a chronic inflammatory response, e.g., diminished tissue
grafted glass substrates and demonstrated that cell motility attachment and thick granulation capsules.87 It was con-
was significantly decreased on RGD peptide-grafted sub- cluded from these studies that the surface topography cre-
strates.81 These results suggested that cell attachment on ated by the 1-3 ∝m pores in membrane filters provided cel-
RGD substrates was stronger and more inhibitory to cell lular cues to promote tissue attachment and improved
migration (more resistant to disruption of cell adhesion sites) biocompatibility. Another study, using the same in vivo
than glass without surface-coupled peptides. Another study model evaluated the effect of the pore size of PTFE mem-
using similar glass substrates quantitated and compared the branes on neovascularization at the membrane-tissue in-
attachment strength of endothelial cells on RGD peptide- terface. This study found that the extent of vascularization
grafted glass versus glass with adsorbed fibronectin and ob- at the membrane-tissue interface was 80-100 times higher
served stronger cell attachment on peptide-grafted sub- in membranes with 5 ∝m pores versus membranes with
strates.82 Overall, substrates containing surface immobilized 0.02 ∝m pores.88 This study provided further evidence of how
cell adhesion peptides have been utilized extensively as sim- surface topographical feature size can influence the
plified models to isolate and study cell adhesive interactions biocompatibility of implanted materials.
at the molecular level. Many in vitro studies have investigated how surface
RGD and other cell-adhesive peptides have been sur- microtextrue influences cellular behavior to correlate with
face immobilized on a wide variety of polymeric biomaterials in vivo studies which have evaluated surface texture effects
to enhance their biocompatibility by promoting cell adhe- on soft tissue biocompatibility. Photo-etching and high pre-
sion in a more controlled manner than relying on random cision milling techniques were developed to precisely con-
protein adsorption. These methods have been developed for trol all geometrical aspects of surface topography down to
many biomedical polymers, including polyethylene tereph- the nanometer range.86 In one report, a simplified model
thalate,54,66 poly tetrafluoroethylene and related polymeric for studying surface topography effects on cell interactions
fluorocarbons,54,77,78 polyurethanes,60,69,75 polyethylene,62 was developed utilizing materials that were fabricated using
cellulose,73 hyaluronic acid,80 poly amino acids,70,76 and high precision methods. On this simple topography, it was
crosslinked dextran.74 Cell-resistant polymeric materials observed that the response of cells in vitro to a single step
which support minimal cell adhesion have been utilized for on a material surface was cell type dependent. Neuronal cell
peptide immobilization to promote highly specific cell- and fibroblast motility were observed to decrease with in-
ligand interactions with a very low background adhesive sig- creasing step height. Neutrophil migration, however, was
nal from adsorbed serum- and cell-borne proteins. These observed to not change in response to step height changes.89
materials include polyethylene glycol,72 nonhydrogel net- Since the cellular response to surface topography is cell type
works of polyacrylate/polyethylene glycol,71 polyacryla- dependent, it is possible that specific microtextured feature
mide,47,48 and polyvinyl alcohol.53 These materials provide sizes may differentially influence tissue cell and inflamma-
maximal control of cell interactions because the surface tory cell interactions with materials to maximize tissue ad-
herence and minimize the inflammatory response. Using
Cell-Extracellular Matrix Interactions Relevant to Vascular Tissue Engineering 589
high precision microtextured surfaces with regular repeated cytoskeletal elements including microtubules, actin mi-
topographical features, the effect of feature dimensions on crofilaments, and focal contacts.96-100 Surface microtexturing
fibroblast contact guidance was assessed in vitro. It was ob- has also been shown to alter cell function, including
served that repeat spacing had a minor and inversely pro- fibronectin mRNA level, mRNA stability, secretion and as-
portional effect on cell alignment, whereas groove depth had sembly by fibroblasts .101 Another study of surface
a more significant role in determining cell alignment, which microtopography and cell function demonstrated that sur-
correlated positively with increased depth.90 In a study ex- face microtextrue promoted osteogenesis.100 These studies
amining cell type dependent responses to repeating alter- further characterize the effects of surface microtextrue on
nating 1 ∝m grooves and 1 ∝m ridges, keratinocytes and neu- cell-material interactions. Overall, surface microtextrue is a
trophils were observed to exhibit no contact guidance, a 20% fundamental surface property that influences cell behavior
response was observed with monocytes and macrophages, and can be exploited to modulate cell-material interactions.
and a 100% response was observed with fibroblasts.91 Simi-
lar studies showed that epithelial cells did not exhibit strong Cell-ECM Interactions in Vascular Biology
contact guidance on microtextured surfaces92 and that they
also responded differently than fibroblasts in vivo as well.93 ECM Modulation of Smooth Muscle Cell Function
In studies comparing fibroblast (model soft tissue cell) and The two major cell types of vascular tissues are smooth
macrophage (model inflammatory cell) responses to muscle and endothelial cells. In the normal adult arterial
microtextured features in vitro, microtextured silicone discs wall, the predominant phenotype or functional state of the
were prepared with multiple surface discontinuities of wells medial smooth muscle cell (SMC) population is predomi-
and pillars with various dimensions, repeat spacings, and nantly nonproliferative and contractile.102-104 These muscle-
90° surface contour angles. These studies revealed that me- like contractile cells contract and relax coordinately in the
tabolism, spreading, and migration of fibroblasts and mac- arterial wall in response to chemical and mechanical stimuli
rophages were altered when they were cultured on substrata to control blood pressure and flow. 102-104 Following me-
with texture feature dimensions ranging from 1-3 ∝m in chanical injury to arterial wall, a large population of medial
width.94,95 Specifically, fibroblast spreading was observed to SMCs will undergo a phenotypic modulation from the con-
increase while macrophage spreading/activation decreased tractile state to a highly proliferative synthetic state in which
in response to this size range of surface feature dimensions extracellular matrix (ECM) proteins are synthesized and
when compared to untextured surfaces. Smaller or larger secreted into the media to repair injury-induced structural
features were observed to have no change in cell responses damage in the arterial wall ECM (Fig. 54.5).105-115 In addi-
when compared to untextured surfaces. When these textured tion to medial repair activity in response to injury, synthetic
silicone materials containing 2 ∝m wide square wells with medial SMCs become motile and migrate from the media
0.5 ∝m depth were implanted subcutaneously in rats, a mini- toward the arterial lumen to form a neointimal layer on the
mal inflammatory response was observed. In contrast, luminal surface of the artery wall. 105-115 Neointima
smooth silicone materials with the same chemical composi- development and progressive neointimal thickening occurs
tion as the textured materials were observed to promote a via endoluminal accumulation of proliferating SMCs with
strong inflammatory response with encapsulation. 86 coordinate ECM deposition.105-115 In the clinical setting,
Therefore, it was concluded that increased fibroblast adhe- neointimal thickening of coronary arteries in response to
sion and decreased macrophage activation in vitro corre- mechanical injury to the artery wall via balloon angioplasty
lated well with the increased tissue attachment and decreased or other percutaneous revascularization procedures is
inflammatory response observed in vivo. Similar results were problematic when the extent of thickening is enough to se-
observed with materials having a wide variety of chemical verely reduce the initial therapeutically beneficial large di-
compositions, including metals, suggesting that surface lated lumen.105-110 To date, restenosis due to extensive
microtextrue effects on cell and tissue responses were inde- neointimal thickening following balloon angioplasty or other
pendent of surface chemistry.86 Overall, these studies indi- catheter-based nonsurgical revascularization procedures
cate a range of topographical parameters that appear to mini- occurs at a rate of 30-50%. 105-110 In vascular and
mize inflammatory responses. Optimal topographical fea- cardiovascular surgery, a synthetic small diameter vascular
tures for maximal cell and tissue attachment include ridges graft that remains patent after implantation has not been
of 1-3 ∝m width with grooves 0.5 ∝m in depth, uniform dis- developed because injury-induced thrombosis and
tribution of features, and sharp feature edges with 90° sur- neointimal hyperplasia causes decreased patency and occlu-
face contour angles. sion at anastomotic sites.111-115
At optimal surface microtextrue dimensions using al- Unlike phenotypically contractile medial SMCs in the
ternating ridge and groove patterns, fibroblast interactions normal arterial wall, synthetic neointimal SMCs in the in-
have been characterized at the subcellular level. Transmis- jured arterial wall synthesize and deposit large quantities of
sion electron microscopy studies have shown that the plasma ECM proteins including collagens,116-119 fibronectin,116,118
membrane of adherent fibroblasts in contact with elastin,119,120 proteoglycans,117,119,121,122 tenascin123,124 and
microtextured surfaces frequently conforms to the textured thrombospondin.125 Tenascin and thrombospondin expres-
feature to interlock with the material surface.96 Several stud- sion in SMCs is greatly increased during postinjury
ies have shown that microtextrue-dependent orientation/ neointima development and accumulates at high levels in
guidance of cells positively correlates with an alignment of the neointimal ECM.123-125 Unlike the other ECM proteins
590 Tissue Engineering of Prosthetic Vascular Grafts
secreted by neointimal SMCs, tenascin and thrombospondin ECM Modulation of Endothelial Cell Function
have cell adhesive and anti-adhesive properties and have been Endothelial cells (ECs) form a monolayer on the lu-
implicated as specialized ECM proteins for events such as minal surfaces of vessels and normally serve as a
tissue regeneration and wound repair.126,127 Tenascin is of nonthrombogenic and selectively permeable boundary be-
particular interest in vascular injury, since its expression in tween the bloodstream and extravascular space.136,137 Fol-
the vessel wall is induced only after mechanical injury or lowing injury and in vascular disease, ECs can become
exposure to growth factors that are released in the vessel wall thrombogenic and promote leukocyte adhesion.136,137
following injury.123,124 It has been speculated that tenascin Throughout the entire vascular system, normal uninjured
facilitates SMC migration and neointima formation in ECs abluminally synthesize, secrete and maintain a basement
vivo124 by creating an ECM of intermediate attachment membrane on their basal surfaces. The subendothelial base-
strength that maximizes SMC migration.129 In vitro studies ment membrane is a complex network of ECM glycopro-
have shown that exogenously added soluble tenascin en- teins including laminin, type IV collagen, heparan sulfate
hances injury-induced SMC migration.130 proteoglycan, nidogen/entactin, and fibronectin.138 Cultured
The ECM plays an active role in the phenotypic modu- endothelial cells have been shown to attach and spread on
lation of SMCs. In vitro studies have shown that SMCs grown various ECM proteins, including fibronectin, laminin, col-
on fibronectin- and RGD peptide-coated materials quickly lagens, vitronectin, and von willebrand factor.139 Similar to
transform from the contractile phenotype to synthetic SMCs, ECM composition has a profound effect on EC phe-
state.131,132 Conversely, similar studies on laminin- and type notype. When ECs are cultured on type I collagen, type III
IV collagen-coated substrates have shown that the contrac- collagen or fibronectin, migration and proliferation were
tile phenotype is conserved.133 observed to be enhanced.140,141 Type IV collagen, laminin,
The interaction of SMCs with the ECM occurs pre- and reconstituted basement membrane (Matrigel) were ob-
dominantly via several members of the integrin superfam- served to inhibit EC proliferation and promote aggregation
ily of extracellular matrix adhesion receptors.12 Ligand af- into capillary-like tubes.138,140,141 Type V collagen has been
finity chromatography and immunoprecipitation analyses observed to selectively inhibit EC attachment and prolifera-
of SMC extracts have identified several !1 integrins which tion without affecting these activities in SMCs and fibro-
bind to a variety of ECM proteins: #5!1, #v!1, and #3!1 bind blasts.142-143
to fibronectin; #1!1 and #7!1 bind to laminin; #v!1 binds to EC-ECM interactions are mediated by several mem-
vitronectin; #1!1 and #2!1 bind to type I collagen; #1!1 binds bers of the integrin superfamily. ECs express several !1
to type IV collagen.128 Integrin #v!3 was observed to bind to integrins, #1!1, #2!1, #3!1, #4!1, #5!1, and #6!1, which pro-
several ECM proteins including fibronectin, laminin, mote adhesion to collagens (#3!1),144,145 LN(#2!1,144-148
vitronectin, type I collagen, and type IV collagen.128 Other #6!1147) and fibronectin (#4!1,149 #5!1).144,145,150,151 Integrin
findings in this study revealed that !1 integrins play a major #v!3, which is present on endothelial cells, recognizes RGD
role in the adhesion and spreading of stationary SMCs, ligands in vitronectin, von Willebrand factor, fibrinogen,
whereas integrin #v!3 is predominant in ECM interactions thrombospondin, and fibronectin.144,145,150,152-159 Like SMCs,
of migrating SMCs (Fig. 54.6). In support of these studies, the expression of specific integrins on ECs is modulated by
TGF-!1 and PDGF, growth factors that are expressed fol- growth factors. For example, TGF-!1 has been observed to
lowing vascular injury and promote SMC migration, were increase surface levels of both !1 and !3 integrins in ECs .134
observed to promote increased surface levels of integrin #v!3 TNF-# and interferon-& have been shown to promote sig-
without subsequent increases in !1 integrins.134-135 These nificant decreases in EC surface levels of !3 integrins with-
findings suggest that growth factor-mediated increased sur- out changing !1 integrin levels.160 Overall, EC and SMC in-
face levels of #v!3 facilitate SMC migration, and further teractions with ECM components contribute significantly
emphasize the role of this integrin in SMC migration. to their overall function in the normal, injured, and diseased
vessel wall. Although not discussed in this review, platelet
and inflammatory cell interactions with the vessel wall play
Cell-Extracellular Matrix Interactions Relevant to Vascular Tissue Engineering 591
an important role in vascular healing and during the pro- evaluated in vitro for promoting EC attachment indepen-
gression of disease.137,161 dently of protein adsorption. Surface-grafted RGD- and
YIGSR-containing peptides were observed to promote en-
Modulating Cell-Extracellular Interactions dothelial cell attachment and spreading on polyethylene
for Vascular Tissue Engineering terepthalate (PET) and polytetrafluoroethylene (PTFE), the
two most widely used materials for vascular grafts.54 EC re-
Tissue Engineered Vascular Implants tention on peptide-grafted materials may generally be im-
Common to all endeavors in tissue engineering, the proved over ECM protein-coated materials, since one study
precise control of interactions between cells and extracellu- has shown that EC retention is better on RGD peptide-
lar surfaces is a fundamental goal in vascular tissue engi- grafted glass than on fibronectin-coated glass.82 EC-selec-
neering. Vascular implant materials which promote cell type tive materials containing surface immobilized GREDVY have
selective adhesion and appropriate cellular functions would been observed to promote adhesion and spreading of en-
vastly improve the long term biocompatibility of synthetic dothelial cells, but not fibroblasts, smooth muscle cells or
vascular implants. With such improvements in platelets.56 The cell adhesion ligand REDV is located on some
biocompatibility, synthetic small diameter vascular grafts types of human plasma fibronectin in alternately spliced cell
would no longer fail and would become a viable option for binding domains.163 EC-selective adhesion has been ob-
surgical procedures. The development of biomimetic sur- served on PET films containing surface immobilized UEA-I,
face modifications and controlled release modalities for vas- a lectin that binds ECs with high affinity. These materials
cular implant materials is critical for achieving controllable promoted EC adhesion without significant monocyte, SMC,
modulation of cellular interactions at the tissue-biomate- or fibroblast adhesion.66 Biomimetic, EC-selective materi-
rial interface. als may enhance the long term biocompatibility of vascular
implants, since thrombosis (mediated by platelet attach-
Modulation of EC Interactions ment) and neointimal hyperplasia (mediated by smooth
with Extracellular Surfaces muscle cell attachment and migration) are major modes of
ECs line the luminal surfaces of all components of the failure. Although extensive studies have evaluated the effect
vascular system and actively maintain homeostasis in the of surface microtextrue on cell-material interactions, few
bloodstream and at the vessel wall. Small diameter synthetic have specifically investigated surface microtextrue effects on
vascular grafts fail clinically due, in part, to the lack of a EC-material interactions. One study has shown that ECs
functional endothelium on the luminal surface. For over 20 spread more rapidly on microtextured polymers prepared
years, many research efforts have focused on transplanting from nanoscale surface replicas of subendothelial ECM than
ECs onto synthetic vascular grafts to improve graft function on untextured polymers.164 This finding suggests that EC
and performance.111-115,136,162 Among the many critical is- adhesion and function are altered on the microtextured sur-
sues in developing this technology, modulation of EC-ma- faces. Transplantation of genetically modified ECs in syn-
terial interactions on vascular grafts is very important, since thetic vascular grafts has been considered as an approach
it influences cell retention and function on the graft lumen. for enhancing EC function and improving graft patency
Biomimetic cell-adhesive biomaterials with surface rates. Unfortunately, the postgraft implantation retention
immobilized cell adhesion peptides have been developed and rates of transplanted, genetically modified ECs have been
592 Tissue Engineering of Prosthetic Vascular Grafts
observed to be significantly lower than unmodified injury-induced neointimal hyperplasia.169-171 Local release
ECs.165-167 Therefore, the benefits gained from improved EC of cyclic RGD from a delivery vehicle incorporated in vas-
function are diminished because of increased cell detach- cular grafts may prove to be effective in limiting graft hy-
ment under flow. Biomimetic surface modifications may sig- perplasia. Some ECM components are synthesized and se-
nificantly improve the retention rate of genetically modi- creted in response to vascular injury to facilitate neointimal
fied ECs by increasing their attachment strength to graft hyperplasia development. Local release of agents which spe-
surfaces. Overall, biomimetic material surfaces may be valu- cifically inhibit the synthesis of these molecules could po-
able in promoting well-controlled EC-material interactions tentially limit graft hyperplasia. One study which tested this
and improved biocompatibility of vascular implants. concept observed that local delivery of tenascin mRNA
antisense oligonucleotides to injured arteries significantly
Modulation of SMC Interactions reduces neointimal hyperplasia in vascular grafts.172 As de-
with Extracellular Surfaces scribed earlier, tenascin is synthesized and secreted in the
The SMC population that normally resides within the neointimal ECM by SMCs following injury and has been
media (inside the wall) of uninjured blood vessels is pre- shown to promote SMC migration in vitro. Other
dominantly contractile and functionally regulates blood counteradhesive ECM proteins like thrombospondin and
pressure and flow. Following mechanical injury, medial SMC type V collagen, that are selectively synthesized and depos-
populations convert to a predominantly ECM-secreting, ited following vascular injury, are also potential ECM com-
proliferative phenotype and migrate from the media to the ponents that could be selectively inhibited by local delivery
lumen, forming a thick multilayered neointima (Fig. 54.5). to limit graft hyperplasia. Systems for local delivery of bio-
In small synthetic vascular grafts, neointimal hyperplasia
(progressive neointimal thickening) is a major mode of fail-
ure due to severely reduced patency and blood flow or total
occlusion, especially at anastomoses.111-115,136,162,168 SMC
phenotype conversion and the development of neointimal
hyperplasia results from the initial mechanical injury of graft
implantation surgery and the long term generation of ab-
normal mechanical forces due to compliance mismatch be-
tween the native vessel and the synthetic graft.111-113,168 Since
SMC-ECM interactions play an important role in modulat-
ing SMC phenotype, migration, proliferation, metabolism,
etc., well-controlled, selective modulation of these interac-
tions by biomimetic material surface modifications and lo-
cal delivery of soluble mediators may limit hyperplasia and
improve the long term biocompatibility of small diameter
synthetic vascular grafts.
Since ECM components play an active role in modu-
lating SMC phenotype, biomimetic cell adhesive
biomaterials may provide a way to selectively control SMC
phenotype expression to limit graft hyperplasia. Laminin and
type IV collagen have been identified as ECM components
which promote expression of the contractile phenotype in
cultured SMCs.102,133 Therefore, surface immobilization of
these proteins or biologically active peptides from these pro-
teins on vascular graft biomaterials may promote pheno-
type reversion of injury-activated SMCs that initially con-
tact the implanted graft and limit further development of
neointimal hyperplasia. Currently, the effect of surface
microtextrue on SMC behavior has been unexplored. How- Fig. 54.7. Tissue engineered vascular grafts via modulation of
ever, it could be speculated that surface microtextrue will cell-ECM interactions. The optimal high performance vascu-
influence SMC adhesion/migration and/or phenotype. lar graft must promote complete luminal coverage with a
An alternative approach for limiting vascular graft hy- nonthrombogenic endothelium that adheres strongly to graft
perplasia is to deliver locally from the implanted graft soluble lumen. Biomimetic surface modifications may facilitate the
agents which selectively limit SMC migration. As described design of these characteristics by providing improved control
earlier in this review, in vitro studies have shown that integrin of EC-biomaterial interactions. This idealized graft must also
#v!3 is the predominant receptor in promoting SMC migra- limit neointimal hyperplasia so that long term patency and
tion (Fig. 54.6). Cyclic RGD peptides, which bind to integrin adequate blood flow is maintained. Biomimetic surface modi-
#v!3 with high affinity, have recently been shown to inhibit fications and local drug delivery systems that modulate SMC
SMC migration in vitro; continuous local or systemic deliv- phenotype and restrict SMC migration may provide means for
ery of cyclic RGD peptides has been shown to limit balloon limiting graft hyperplasia.
Cell-Extracellular Matrix Interactions Relevant to Vascular Tissue Engineering 593
logically active soluble agents in implanted vascular grafts 17. Chung CA, Erickson HP. Cell surface annexin II is a high
may improve long term graft biocompatibility. affinity receptor for the alternatively spliced segment of
tenascin-C. J Cell Biol 1994; 126:539-548.
Conclusion 18. Barnea G, Grumet M, Milev et al. Receptor tyrosine phos-
Development of biomimetic materials which contain phatase ! is expressed in the form of proteoglycan and
binds to the extracellular matrix protein tenascin. J Biol
well-defined ECM-like physicochemical features to promote
Chem 1994; 269:14349-14352.
selective cell interactions is a fundamentally important area 19. Lafrienie RM, Yamada KM. Integrin-dependent signal
of research in the fields of tissue engineering and transduction. J Cell Biochem 1996; 61:543-553.
biomaterials science. In vascular tissue engineering, it is de- 20. Otey CA. pp125FAK in the focal adhesion. Int Rev Cytol
sirable to have a synthetic vascular graft that promotes com- 1996; 167:161-183.
plete luminal coverage with a healthy, nonthrombogenic 21. Horwitz A, Duggan K, Buck C et al. Interaction of plasma
endothelium. Further, this biohybrid graft must limit membrane fibronectin receptor with talin—a transmem-
neointimal hyperplasia so that long term patency and ad- brane linkage. Nature 1986; 320:531-533.
equate blood flow is maintained (Fig. 54.7). Development 22. Muguruma M, Matsumura S, Fukazawa. Direct interac-
of appropriate biomimetic material surface modifications tions between talin and actin. Biochem Biophys Res
Commun 1990; 171:1217-1223.
in conjunction with other technologies such as genetic ma-
23. Burridge K, Mangeat P. An interaction between vinculin
nipulation of cells will provide the tissue engineer and bio- and talin. Nature 1984; 308:744-746.
material scientist with new tools for creating high-perfor- 24. Wachsstock DH, Wilkins JA, Lin S. Specific interaction
mance implants and devices, including those for cardiovas- of vinculin with #-actinin. Biochem Biophys Res Commun
cular applications. 1987; 146:554-560.
25. Turner CE, Glenney JR, Burridge K. Paxillin: A new
References vinculin-binding protein present in focal adhesions. J Cell
1. Folkman J, Moscona A. Role of cell shape in growth con- Biol 1990; 111:1059-1068.
trol. Nature 1978; 273:345-349. 26. Wilkins JA, Risinger MA, Lin S. Studies on proteins that
2. Stoker M, O’Neill C, Berryman S et al. Anchorage and copurify with smooth muscle vinculin: Identification of
growth regulation in normal and virus-transformed cells. immunologically related species in focal adhesions of
Int J Cancer 1968; 3:638-648. nonmuscle and Z-lines of muscle cells. J Cell Biol 1986;
3. O’Neill CH, Riddle PN, Jordan PW. The relation between 103:1383-1394.
surface area and anchorage dependence of growth in ham- 27. Johnson RP, Craig SW. F-actin binding site masked by
ster and mouse fibroblasts. Cell 1979; 16:909-918. the intramolecular association of vinculin head and tail
4. Ruoslahti E, Reed JC. Anchorage dependence, integrins, domains. Nature 1995; 373:261-264.
and apoptosis. Cell 1994; 77:477-478. 28. Otey CA, Pavalko FM, Burridge K. An interaction between
5. Grinnell F. Cellular adhesiveness and extracellular sub- alpha-actinin and the !1 subunit in vitro. J Cell Biol 1990;
strata. Int Rev Cytol. New York: Academic Press, 1978; 111:721-729.
53:67-149. 29. Kornberg LJ, Earp HS, Turner CE et al. Signal transduc-
6. Jauregui HO. Cell adhesion to biomaterials: The role of tion by integrins: Increased protein tyrosine phosphory-
several extracellular matrix components in the attachment lation caused by clustering of !1 integrins. Proc Natl Acad
of nontransformed fibroblasts and parenchymal cells. Sci USA 1991; 88:8392-8396.
Trans Am Soc Artif Itern Organs 1987; 33:66-74. 30. Guan J-L, Trevithick JE, Hynes RO. Fibronectin/integrin
7.. Hay ED, ed. Cell Biology of Extracellular matrix, 2nd ed., interaction induces tyrosine phosphorylation of a 120 kDa
New York: Plenum Press, 1991. protein. Cell Regul 1991; 2:951-964.
8. Paulsson M. Basement membrane proteins: Structure, as- 31. Kornberg LJ, Earp HS, Parsons JT et al. Cell adhesion or
sembly, and cellular interactions. Crit Rev Biochem Mol integrin clustering increases phosphorylation of a focal
Biol 1992; 27:93-127. adhesion-associated tyrosine kinase. J Biol Chem 1992;
9. Ratner BD. New ideas in biomaterials science—a path to 267:23439-23442.
engineered biomaterials. J Biomed Mater Res 1993; 32. Hildebrand JD, Schaller M, Parsons JT. Identification of
27:837-850. sequences required for the efficient localization of the fo-
10. Hynes RO. Integrins: A family of cell surface receptors. cal adhesion kinase, pp125FAK, to cellular focal adhesions.
Cell 1987; 48:549-554. J Cell Biol 1993; 123:993-1005.
11. Ruoslahti E. Integrins. J Clin Invest 1991; 87:1-5. 33. Parsons JT, Schaller MD, Hildebrand JD et al. Focal ad-
12. Hynes RO. Integrins: Variety, versatility, and interactions hesion kinase: Structure and signaling. J Cell Sci Suppl
in cell adhesions. Cell 1992; 69:11-25. 1994; 18:109-113.
13. Gille J, Swerlick RA. Integrins: Role in cell adhesion and 34. Cohen GB, Ren R, Baltimore D. Modular binding do-
communication. Ann NY Acad Sci 1996; 797:93-106. mains in signal transduction proteins. Cell 1995; 80:
14. Couchman JR, Woods A. Syndecans, signaling, and cell 237-248.
adhesion. J Cell Biochem 1996; 61:578-584. 35. Sabe H, Hata A, Okada M et al. Analysis of the binding
15. Entwistle J, Hall CL, Turley EA. HA receptors: Regula- of the src homology 2 domain of csk to tyrosine-phos-
tors of signaling to the cytoskeleton. J Cell Biochem 1996; phorylated proteins in the suppression and activation of
61:569-577. c-src. Proc Natl Acad Sci USA 1994; 91:3984-3988.
16. Mecham RP. Laminin receptors. Ann Rev Cell Biol 1991; 36. Weng Z, Taylor JA, Turner CE et al. Detection of src
7:71-91. homology 3-binding proteins, including paxillin, in nor-
mal and v-src transformed Balb/c 3T3 cells. J Biol Chem
1993; 268:14956-14963.
594 Tissue Engineering of Prosthetic Vascular Grafts
37. Schlaepfer DD, Hanks SK, Hunter T et al. Integrin-medi- and 140 nm for focal contact and stress fiber formation.
ated signal transduction linked to ras pathway by GRB-2 J Cell Biol 1991; 114:1089-1100.
binding to focal adhesion kinase. Nature 1994; 56. Hubbell JA, Massia SP, Desai NP et al. Endothelial cell-
372:786-791. selective materials for tissue engineering in the vascular
38. Lenormand P, Sardet C, Pages G et al. Growth factors graft via a new receptor. Bio/Technology 1991; 9:568-572.
induce nuclear translocation of MAP kinases (p42mapk and 57. Ito Y, Kajihara M, Imanishi Y. Materials for enhancing
p44mapk) but not of their activator MAP kinase kinase cell adhesion by immobilization of cell-adhesive peptide.
(p45mapkk). J Cell Biol 1993; 122:1079-1088. J Biomed Mater Res 1991; 25:1325-1337.
39. Hansen CA, Schroering AG, Carey DJ et al. Localization 58. Hirano Y, Kando Y, Hayashi T et al. Synthesis and cell
of a heterodynamic G protein gamma subunit to focal attachment activity of bioactive oligopeptides: RGD,
adhesions and stress fibers. J Cell Biol 1994; 126:811-819. RGDS, RGDV, and RGDT. J Biomed Mater Res 1991;
40. Jaken S, Leach K, Klauk T. Association of type 3 protein 25:1523-1534.
kinase c with focal contacts in rat embryo fibroblasts. J 59. Britland S, Perez-Arnaud E, Clark P et al. Micropatterning
Cell Biol 1989; 109:697-707. proteins and synthetic peptides on solid supports: A novel
41. McNamee HP, Ingber DE, Schwartz MA. Adhesion to application for microelectronics fabrication technology.
fibronectin stimulates inositol lipid synthesis and enhances Biotechnol Prog 1992; 8:155-60.
PDGF-induced inositol lipid breakdown. J Cell Biol 1993; 60. Lin HB, Garcia-Echeverria C, Asakura S et al. Endothe-
121:673-678. lial cell adhesion on polyurethanes containing covalently
42. Schwartz MA. Spreading of endothelial cells on fibronectin attached RGD-peptides. Biomaterials 1992; 13:905-914.
or vitronectin triggers elevation of intracellular free cal- 61. Fassina G. Oriented immobilization of peptide ligands on
cium. J Cell Biol 1993; 120:1003-1010. solid supports. J Chromatogr 1992; 591:99.
43. Schwarz MA, Ingber DE, Lawrence M et al. Multiple 62. Shenoy NR, Bailey JM, Shively JE. Carboxylic acid-modi-
integrins share the ability to induce elevation of intracel- fied polyethylene: A novel support for the covalent im-
lular pH. Exp Cell Res 1991; 195:533-535. mobilization of polypeptides for C-terminal sequencing.
44. Ratner BD. New ideas in biomaterials science—a path to Protein Sci 1992; 1:58-67.
engineered biomaterials. J Biomed Mater Res 1993; 63. Massia SP, Rao S, Hubbell JA. Covalently immobilized
27:837-850. laminin peptide Tyr-Ile-Gly-Ser-Arg (YIGSR) supports cell
45. Andrade JD, Hlady V. Protein adsorption and materials spreading and colocalization of the 67-kilodalton laminin
biocompatibility: A tutorial review and suggested hypoth- receptor with #-actinin and vinculin. J Biol Chem 1993;
esis. Adv Polymer Sci 1986; 79:1-63. 268:8053-8059.
46. Horbett TA, Brash JL. Proteins at interfaces: Current is- 64. Kondoh A, Makino K, Matsuda T. Two-dimensional ar-
sues and future prospects. ACS Symposium Ser 1987; tificial extracellular matrix: Bioadhesive peptide-immobi-
343:1-33. lized surface design. J Appl Polm Sci 1993; 47:1983.
47. Brandley BK, Schnaar RL, Covalent attachment of an Arg- 65. Hirano Y, Okuno M, Hayashi T et al. Cell-attachment
Gly-Asp sequence peptide to derivatizable polyacrylamide activities of surface immobilized oligopeptides RGD,
surfaces: Support of fibroblast adhesion and long-term RGDS, RGDT, and YIGSR toward five cell lines. J
growth. Anal Biochem 1988; 172:270-278. Biomater Sci Polym Edn 1993; 4:235.
48. Brandley BK, Schnaar RL. Tumor cell haptotaxis on co- 66. Ozaki CK, Phaneuf MD, Hong SL et al. Glycoconjugate
valently immobilized linear and exponential gradients of mediated endothelial cell adhesion to Dacron polyester
a cell adhesion peptide. Dev Biol 1989; 135:74-86. film. J Vasc Surg 1993; 18:486.
49. Matsuda T, Kondo A, Makino K et al. Development of a 67. Nakayama Y, Matsuda T, Irie M. A novel surface photo-
novel artificial matrix with cell adhesion peptides for cell graft polymerization method for fabricated devices. ASAIO
culture and artificial and hybrid organs. Trans Am Soc J 1993; 39:M542.
Artif Intern Organs 1989; 35:677-679. 68. Collioud A, Clemence JF, Sanger M et al. Oriented and
50. Massia SP, Hubbell JA. Covalently grafted RGD- and covalent immobilization of target molecules to solid sup-
YIGSR-containing synthetic peptides support receptor- ports: Synthesis and application of a light-activatable and
mediated adhesion of cultured fibroblasts. Analytical Bio- thiol-reactive cross-linking reagent. Bioconjug Chem 1993;
chemistry 1990; 187:292-301. 4:528-536.
51. Massia SP, Hubbell JA. Covalently attached GRGD on 69. Lin HB, Garcia-Echeverria C, Sun W et al. Synthesis of a
polymer surfaces promotes biospecific adhesion of mam- novel polyurethane copolymer containing covalently at-
malian cells. Ann NY Acad Sci 1990; 589:261-270. tached RGD peptide. J Biomater Sci Polym Edn 1993;
52. Ozeki E, Matsuda T. Development of an artificial extra- 4:235.
cellular matrix. Solution castable polymers with cell rec- 70. Barrera DA, Zystra E, Lansbury PT et al. Synthesis and
ognizable peptidyl side chain. ASAIO Trans 1990; RGD peptide modification of a new biodegradable copoly-
36:M294. mer: Poly(lactic acid-colysine). J Am Chem Soc 1993;
53. Nakajima K, Hirano Y, Iida T et al. Adsorption of plasma 115:11010.
proteins an Arg-Gly-Asp-Ser peptide-immobilized 71. Drumheller PD, Elbert DL, Hubbell JA. Multifunctional
poly(vinyl alcohol) and ethylene-acrylic copolymer films. poly(ethylene glycol) semiinterpenetrating polymer net-
Polym J 1990; 22:985. works as highly selective adhesive substrates for
54. Massia SP, Hubbell JA. Human endothelial cell interac- bioadhesive peptide grafting. Biotech Bioeng 1994; 43:772.
tions with surface-coupled adhesion peptides on a non- 72. Cima LG. Polymer substrates for controlled biological
adhesive glass substrate and two polymeric biomaterials. interactions. J Cell Biochem 1994; 56:155-161.
J Biomed Mater Res 1991; 25:223-242. 73. Englebretsen DR, Harding DR. High yield, directed im-
55. Massia SP, Hubbell JA. An RGD spacing of 440 nm is mobilization of a peptide-ligand onto a beaded cellulose
sufficient for integrin #v!3%mediated fibroblast spreading support. Pept Res 1994; 7:322-326.
Cell-Extracellular Matrix Interactions Relevant to Vascular Tissue Engineering 595
74. Besselink GA, Beugeling T, Poot AA et al. Preparation of 94. Schmidt, JA, von Recum AF. The macrophage response
cell-affinity adsorbents by immobilization of peptides to to microtextured silicone. Biomaterials 1991;
Sephadex G-10. J Biomater Sci Polym Ed 1994; 6:675-693. 13:1095-1099.
75. Lin HB, Sun W, Mosher DF et al. Synthesis, surface, and 95. Green AM, Jansen JA, von Recum AF. The fibroblast re-
cell adhesion properties of polyurethanes containing co- sponse to microtextured silicone surfaces: Texture orien-
valently grafted RGD-peptides. J Biomed Mater Res 1994; tation into or out of the surface. J Biomed Mater Res
28:1523-1534. 1994; 28:647-653.
76. Kugo K, Okuno M, Matsuda K et al. Fibroblast attach- 96. Meyle J, Wolburg H, von Recum AF. Surface micromor-
ment to Arg-Gly-Asp peptide-immobilized poly(gamma- phology and cellular interactions. J Biomat Appl 1993;
methyl L-glutamate). J Biomater Sci Polym Ed 1994; 7:362-374.
5:325-337. 97. Oakley C, Brunette DM. The sequence of alignment of
77. Vargo TG, Bekos EJ, Kim YS et al. Synthesis and charac- microtubules, focal contacts, and actin filaments in fibro-
terization of fluoropolymeric substrata with immobilized blasts spreading on smooth and grooved titanium sub-
minimal peptide sequences for cell adhesion studies. I. J strata. J Cell Sci 1993; 106:303.
Biomed Mater Res 1995; 29:767-778. 98. den Braber ET, de Ruijter JE, Smits HT et al. Quantita-
78. Ranieri JP, Bellamkonda R, Bekos EJ et al. Neuronal cell tive analysis of cell proliferation and orientation on sub-
attachment to fluorinated ethylene propylene films with strata with uniform parallel surface micro-grooves.
covalently immobilized laminin oligopeptides YIGSR and Biomaterials 1996; 17:1093-1099.
IKVAV. II. J Biomed Mater Res 1995; 29:779-785. 99. Wojciak-Stothard B, Curtis A, Monaghan W et al. Guid-
79. Sugawara T, Matsuda T. Photochemical surface ance and activation of murine macrophages by nanometric
derivatization of a peptide containing Arg-Gly-Asp (RGD). scale topography. Exp Cell Res 1996; 223:426-435.
J Biomed Mater Res 1995; 29:1047-1052. 100. Qu J, Chehroudi B, Brunette DM. The use of
80. Glass JR, Dickerson KT, Stecker K et al. Characterization micromachined surfaces to investigate the cell behavioral
of a hyaluronic acid-Arg-Gly-Asp peptide cell attachment factors essential to osseointegration. Oral Dis 1996;
matrix. Biomaterials 1996; 17:1101-1108. 2:102-115.
81. Olbrich KC, Anderson TT, Blumenstock FA et al. Sur- 101. Chou L, Firth JD, Uitto VJ et al. Substratum surface to-
faces modified with covalently-immobilized adhesive pep- pography alters cell shape and regulates fibronectin mRNA
tides affect fibroblast population motility. Biomaterials level, mRNA stability, secretion and assembly in human
1996; 17:144-153. fibroblasts. J Cell Sci 1995; 108:1563-1573.
82. Xiao Y, Truskey GA. Effect of receptor-ligand affinity on 102. Thyberg J. Differentiated properties and proliferation of
the strength of endothelial cell adhesion. Biophys J 1996; arterial smooth muscle cells in culture. Int Rev Cytol 1996;
71:2869-2894. 169:183-265.
83. Pierschbacher MD, Ruoslahti E. Influence of stereochem- 103. Somlyo AP, Somylo AV. Signal transduction and regula-
istry of the sequence Arg-Gly-Asp-Xaa on binding speci- tion in smooth muscle. Nature 1994; 17:231-236.
ficity in cell adhesion. J Biol Chem 1987; 262:17294-17298. 104. Owens GK. Regulation of differentiation of vascular
84. Harrison RG. The cultivation of tissues in extraneous smooth muscle cells. Physiol Rev 1995; 75:487-517.
media as a method of a morphogenic study. Anat Rec 105. Liu MW, Roubin GS, King SB. Restenosis after coronary
1912; 6:181-193. angioplasty: Potential biological determinants and role of
85. Weiss P. Experiments on cell and axon orientation in intimal hyperplasia. Circulation 1989; 79:1374-1387.
vitro: The role of colloidal exudates in tissue organiza- 106. Forrester JS, Fishbein M, Helfant R et al. A paradigm for
tion. J Exp Zool 1945; 100:353. restenosis based on cell biology: Clues for the develop-
86. von Recum AF, Shannon CE, Cannon CE et al. Surface ment of new preventive therapies. J Am Coll Cardiol 1991;
roughness, porosity, and texture as modifiers of cellular 17:758-769.
adhesion. Tissue Engineering 1996; 2:241-253. 107. Reidy MA, Jackson C, Lindner V. Neointimal prolifera-
87. Campbell CE, von Recum AF. Microtopography and soft tion: Control of vascular smooth muscle cell growth. Vasc
tissue repsonse. J Invest Surg 1989; 2:51-74. Med Rev 1992; 3:156-167.
88. Brauker JH, Carr-Brendel VE, Martinson LA et al. 108. Casscells W. Migration of smooth muscle and endothe-
Neovascularization of synthetic membranes directed by lial cells. Critical events in restenosis. Circulation 1992;
membrane architecture. J Biomed Mater Res 1996; 86:723-729.
29:1517-1524. 109. Ferns AA, Stewart-Lee AL, Änggård EE. Arterial response
89. Clark P, Connolly P, Curtis AS et al. Topographical con- to mechanical injury: Balloon catheter de-endo-
trol of cell behaviour. I. Simple step cues. Development thelialization. Atherosclerosis 1992; 92:89-104.
1987; 99:439-448. 110. Casscells W, Engler D, Willerson JT. Mechanisms of
90. Clark P, Connolly P, Curtis AS et al. Topographical con- restenosis. Texas Heart Inst J 1994; 21:68-77.
trol of cell behaviour. II. Mutiple grooved substrata. De- 111. Yeager A, Callow AD. New graft materials and current
velopment 1990; 108:635-644. approaches to an acceptable small diameter vascular graft.
91. Meyle J, Gultig K, Nisch W. Variation in contact guid- Trans Am Soc Artif Intern Organs 1988; 34:88-94.
ance by human cells on a microstructured surface. J 112. Callow AD. Presidential address: The microcosm of the
Biomed Mater Res 1995; 29:81-88. arterial wall—a plea for research. J Vasc Surg 1987; 5:1-17.
92. Oakley C, Brunette DM. Response of single, pairs, and 113. Burkel WE. The challenge of small diameter vascular
clusters of epithelial cells to substratum topography. grafts. Med Prog Technol 1988; 14:165-175.
Biochem Cell Biol 1995; 73:473-489. 114. Greisler HP. Interactions at the blood/material interface.
93. Cheroudi B, Gould TR, Brunette DM. Effects of grooved Ann Vasc Surg 1990; 4:98-103.
epoxy substratum on epithelial cell behavior in vitro and 115. Zilla P. The endothelium: A key to the future. J Card Surg
in vivo. J Biomed Mater Res 1995; 6:459-473. 1993; 8:32-60.
596 Tissue Engineering of Prosthetic Vascular Grafts
116. Majesky MW, Lindner V, Twardzik DR et al. Production ciprocal effects on cell migration. J Cell Physiol 1992;
of transforming growth factor beta 1 during repair of ar- 153:118-128.
terial injury. J Clin Invest 1991; 88:904-910. 135. Janat MF, Argraves WS, Liau G. Regulation of vascular
117. Snow AD, Bolender RP, Wight TN et al. Heparin modu- smooth muscle cell integrin expression by transforming
lates the composition of the extracellular matrix domain growth factor !1 and platelet-derived growth factor-BB. J
surrounding arterial smooth muscle cells. Am J Pathol Cell Physiol 1992; 151:588-595.
1990; 137:313-390. 136. Massia SP, Hubbell JA. Tissue engineering in the vascu-
118. Madri JA, Reidy MA, Kocher O et al. Endothelial cell lar graft. Cytotechnology 1992; 10:189-204.
behavior after denudation injury is modulated by 137. Luscinskas FW, Lawler J. Integrins as dynamic regulators
transformtin growth factor-beta 1 and fibronectin. Lab of vascular function. FASEB J 1994; 8:929-938.
Invest 1989; 60:755-765. 138. Grant DS, Kleinman HK, Martin GR. The role of base-
119. Burke JM, Ross R. Synthesis of connective tissue macro- ment membranes in vascular development. Ann NY Acad
molecules by smooth muscle. Int Rev Connect Tissue Res Sci 1990; 588:61-72.
1979; 8:119-157. 139. Carey DJ. Control of growth and differentiation of vas-
120. Majesky MW, Giachelli CM, Reidy MA et al. Rat carotid cular cells by extracellular matrix proteins. Ann Rev
neointimal smooth muscle cells reexpress a developmen- Physiol 1991; 53:161-177.
tally regulated mRNA phenotype during repair of arterial 140. Madri JA, Pratt BM. Endothelial-matrix interactions: In
injury. Circ Res 1992; 71:759-768. vitro models of angiogenesis. J Histochem Cytochem 1986;
121. Wight TN. Cell biology of arterial proteoglycans. Arte- 34:85-91.
riosclerosis 1989; 9:1-20. 141. Madri JA, Williams SK. Capillary endothelial cell cultures:
122. Alvai M, Moore S. Glycosaminoglycan composition and Phenotypic modulation by matrix components. J Cell Biol
biosynthesis in the endothelium-covered neointima and 1983; 97:153-165.
de-endothelialized rabbit aorta. Exp Mol Pathol 1985; 142. Fukuda K, Koshihara Y, Oda H et al. Type V collagen
42:389-400. selectively inhibits human endothelial cell proliferation.
123. Hedin U, Holm J, Hansson GK. Induction of tenascin in Biochem Biophys Res Commun 1988; 151:1060-1068.
rat arterial injury. Relationship to altered smooth muscle 143. Ziats NP, Anderson JM. Human vascular endothelial cell
cell phenotype. Am J Pathol 1989; 139:649-656. attachment and growth inhibition by type V collagen. J
124. Majesky MW. Neointima formation after acute vascular Vasc Surg 1993; 17:710-718.
injury. Role of counteradhesive extracellular matrix pro- 144. Cheng YF, Kramer RH. Integrin receptors in human mi-
teins. Texas Heart Inst J 1994; 21:78-85. crovascular endothelial cells that mediate adhesion to the
125. Raugi GL, Mullen JS, Bark DH et al. Thrombospondin extracellular matrix. J Cell Physiol 1989; 139:275-286.
deposition in rat carotid artery injury. Am J Pathol 1990; 145. Albelda SM, Daise M, Levine EM et al. Identification and
137:179-185. characterization of cell-substratum adhesion receptors on
126. Chuong CM, Chen HM. Enhanced expression of neural cultured human endothelial cells. J Clin Invest 1989;
cell adhesion molecules and tenascin (cytotactin) during 83:1992-2002.
wound healing. Am J Path 1990; 138:427-440. 146. Kramer RH, Cheng Y-F, Clyman R. Human microvascu-
127. Mackie EJ, Halfter W, Liverani D. Induction of tenascin lar endothelial cells use !1 and !3 integrin receptor com-
in healing wounds. J Cell Biol 1988; 107:2757-2767. plexes to attach to laminin. J Cell Biol 1990;
128. Clyman RI, Mauray F, Kramer RH. !1 and !3 integrins 111:1233-1243.
have different roles in the adhesion and migration of vas- 147. Languino LR, Gehlsen KR, Wayner E et al. Endothelial
cular smooth cells on extracellular matrix. Exp Cell Res cells use #2!1 integrin as a laminin receptor. J Cell Biol
1992; 200:272-284. 1989; 109:2455-2462.
129. DiMilla PA, Stone J, Quinn JA et al. Maximal migration 148. Kirchhofer D, Languino LR, Ruoslahti E et al. # 2 ! 1
of human smooth muscle cells on fibronectin and type integrins from different cell types show different binding
IV collagen occurs at an intermediate attachment strength. specificities. J Biol Chem 1990; 265:615-618.
J Cell Biol 1993; 122:729-737. 149. Massia SP, Hubbell JA. Vascular endothelial cell adhesion
130. Massia SP, Slepian MJ. Tenascin enhances injury-induced and spreading promoted by the peptide REDV of the
vascular smooth muscle cell migration partially via IIICS region of plasma fibronectin is mediated by integrin
integrin #v!3. Exp Cell Res, 1997; in revision. #4!1. J Biol Chem 1992; 267:14019-14026.
131. Hedin U, Thyberg J. Plasma fibronectin promotes modu- 150. Dejana E, Colella S, Conforti G et al. Fibronectin and
lation of arterial smooth muscle cells form contractile to vitronectin regulate the organization of their respective
synthetic phenotype. Differentiation 1987; 33:239-246. Arg-Gly-Asp receptors in cultured human endothelial
132. Hedin U, Bottger BA, Johansson S et al. A substrate of cells. J Cell Biol 1988; 107:1215-1223.
the cell attachment sequence of fibronectin (Arg-Gly-Asp- 151. Conforti G, Zanetti A, Colella S et al. Interaction of
Ser) is sufficient to promote transition of arterial smooth fibronectin with cultured human endothelial cells: Char-
muscle cells from a contractile to a synthetic phenotype. acterization of the specific receptor. Blood 1989;
Dev Biol 1989; 133:489-501. 73:1576-1585.
133. Hedin U, Bottger BA, Forsberg E et al. Diverse effects of 152. Dejana E, Colella S, Languino LR et al. Fibrinogen in-
fibronectin and laminin on phenotype properties of cul- duces adhesion, spreading, and microfilament organiza-
tured arterial smooth muscle cells. J Cell Biol 1988; tion of human endothelial cells in vitro. J Cell Biol 1987;
107:307-319. 104:1403-1411.
134. Basson CT, Kocher O, Basson MD et al. Differential 153. Dejana E, Lampugnami MG, Giorgi M et al. Von
modulation of vascular cell integrin and extracellular willebrand factor promotes endothelial cell adhesion via
matrix expression in vitro by TGF-!1 correlates with re- an Arg-Gly-Asp-dependent mechanism. J Cell Biol 1989;
109:367-375.
Cell-Extracellular Matrix Interactions Relevant to Vascular Tissue Engineering 597
154. Dejana E, Languino LR, Colella S et al. The localization 164. Goodman SL, Sims PA, Albrecht RM. Three-dimensional
of a platelet gpIIb-IIIa-related protein in endothelial cell extracellular matrix textured biomaterials. Biomaterials
adhesion structures. Blood 1988; 71:566-572. 1996; 17:2087-2095.
155. Dejana E, Languino LR, Polentarutti N et al. Interaction 165. Dunn PF, Newman KD, Jones M et al. Seeding of vascu-
between fibrinogen and cultured endothelial cells: Induc- lar grafts with genetically modified endothelial cells. Se-
tion of migration and specific binding. J Clin Invest 1985; cretion of recombinant tPA results in decreased seeded
75:11-18. cell retention in vitro and in vivo. Circulation 1996;
156. Charo IF, Bekeart LS, Phillips DR. Platelet glycoprotein 93:1439-1446.
IIb-IIIa-like proteins mediate endothelial cell attachment 166. Sackman JE, Freeman MB, Petersen MG et al. Synthetic
to adhesive proteins and the extracellular matrix. J Biol vascular grafts seeded with genetically modified endothe-
Chem 1987; 262:9935-9938. lium in the dog: Evaluation of the effect of seeding tech-
157. Cheresh DA. Human endothelial cells synthesize and ex- nique and retroviral vector on cell persistence in vivo. Cell
press an Arg-Gly-Asp directed adhesion receptor involved Transplant 1995; 4:219-235.
in attachment to fibrinogen and von willebrand factor. 167. Sackman JE, Cezeaux JL, Reddick TT et al. Evaluation of
Proc Natl Acad Sci 1987; 84:6471-6475. the effect of retroviral gene transduction on vascular en-
158. Cheng Y-F, Clyman RI, Enenstein J et al. The integrin dothelial cell adhesion. Tissue Eng 1996; 2:223-236.
complex #v!3 participates in the adhesion of microvascu- 168. Painter TA. Myointimal hyperplasia: Pathogenesis and
lar endothelial cells to fibronectin. Exp Cell Res 1991; implications. 2. Animal injury models and mechanical
194:69-77. factors. Artif Organs 1991; 15:102-118.
159. Lawler J, Weinstein R, Hynes RO. Cell attachment to 169. Slepian MJ, S Massia, Dehdashti B et al. !3 Rather than
thrombospondin: The role of Arg-Gly-Asp, calcium, and !1 integrins dominate integrin-matrix interactions involved
integrin receptors. J Cell Biol 1988; 107:2351-2361. in postinjury smooth muscle cell migration. Circulation
160. Defilippi P, Truffa G, Stefanuto G et al. Tumor necrosis 1997: in revision.
factor # and interferon & modulate the expression of the 170. Choi ET, Engel L, Callow AD et al. Inhibition of
vitronectin receptor (integrin !3 in human endothelial neointimal hyperplasia by blocking #v!3 integrin with a
cells. J Biol Chem 1991; 266:7638-7645. small peptide antagonist GpenGRGDSPCA. J Vasc Surg
161. Rubin BG, Santoro SA, Sicard GA. Platelet interactions 1994;19:125-134.
with the vessel wall and prosthetic grafts. Ann Vasc Surg 171. Matsuno H, Stassen JM, Vermylen J et al. Inhibition of
1993; 7:200-207. integrin fucntion by a cyclic RGD-containing peptide pre-
162. Williams SK. Endothelial cell transplantation. Cell Trans- vents neointima formation. Circulation 1994;
plant 1995; 4:401-410. 90:2203-2206.
163. Humphries MJ, Akiyama SK, Komoriya A et al. Identifi- 172 Hirko M, Kang SS, Denner L et al. Antisense oligonucle-
cation of an alternatively spliced site in human plasma otides against FGFR-1, tenascin, and osteopontin inhibit
fibronectin that mediates cell type-specific adhesion. J Cell canine vein graft myointimal hyperplasia. Surg Forum
Biol 1986; 103:2637-2647. 1995; 46:349-351.
Matrix Engineering
CHAPTER 55
Use of Hydroxypropylchitosan Acetate as a Carrier
for Growth Factor Release
Keiko Yamamura, Toshitaka Nabeshima, Tsunehisa Sakurai
Introduction
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
600 Tissue Engineering of Prosthetic Vascular Grafts
The subject of this article is limited to the possible use function of the initial concentrations of the factor, where
of HPCHA as a carrier controlling the release of growth fac- the increase in protein contents was used as a measure for
tor in vitro and vivo.11 the proliferation of the cells. bFGF content in a bFGF graft
was 2.05 ± 0.32 ∝g (SE, n = 5-6). bFGF content in a bFGF-
Use of HPCHA as a Carrier for bFGF HPCHA graft was 2.71 ± 0.41 ∝g (SE, n = 5). Considering
Acetylation of the amino group in hydroxy- that both Gore grafts were water loaded, this value appears
propylchitosan (HPCH, MW ~800,000; deacetylation de- reasonable. Also, nearly 100% biological activity of the bFGF
gree, 60%; 7200 cps, 5% w/v) was performed following the incorporated into the grafts was maintained. The present
procedure of Tokura et al.8 In brief, 10 g of HPCH was dis- data demonstrates that recombinant bFGF can be incorpo-
solved in 150 ml of deionized water and 100 ml of methanol rated into a graft during aseptic procedures, maintaining
was added to the solution. Then 25 ml of acetic anhydride biological activity.
was added and stirred vigorously overnight. After the re-
sulting solution was dialyzed in deionized water, HPCHA Influence of HPCHA on Release of bFGF
was obtained by lyophilizing the solution. Sixty percent
acetylation was confirmed by IR spectrometry. The chemi- In Vitro
cal structure of HPCHA is shown in Figure 55.2. The bFGF-HPCHA Gore graft was placed into a bo-
The amount of recombinant human basic fibroblast ron coated glass tube containing 1 ml of phosphate buffer
growth factor (bFGF) incorporated in an expanded (pH 5.5) maintained at 37°C, and transferred into new ones
polytetrafluoroethylene graft (Gore-Tex®, inner diameter after 0.5, 1, 2, 4, 6, 8, 16 and 24 h. Samples were stored at 5°C
3 mm, wall thickness 0.64 mm, mean pore size 30 ∝m, po- and assayed within 3 days. The bFGF Gore graft was also
rosity 80%) was predicted from the water accessible volume. studied. Fig. 55.5 shows the cumulative percent of released
Water loading was 0.00116 ml/graft (0.75 cm in diameter) bFGF from bFGF Gore grafts and bFGF-HPCHA Gore
for distilled water and 0.0147 ml/graft (0.75 cm in diam- grafts. bFGF release from the bFGF Gore graft reached 100%
eter) for the HPCHA solution (4%, w/v), respectively. at 24 h. For the bFGF-HPCHA Gore graft, bFGF was re-
bFGF was dissolved in sterilized water at a concentra- leased at about 30% at 8 h and 55% at 24 h. Incorporation
tion of 1800 ∝g/ml. Gore-Tex® loaded with bFGF solution of bFGF with HPCHA can apparently sustain its release as
was freeze-dried by the procedure of Yamamura et al.12 compared with the bFGF Gore graft.
HPCHA was dissolved in bFGF solution (180 ∝g/ml) at a
concentration of 4% (w/v). Specific gravity of the solution In Vivo
was 1.01. Figure 55.3 shows scanning electron micrographs Skin pockets in a single row were made in the left and
(SEM) of inner surfaces of bFGF-HPCHA Gore grafts. the right flanks of Japanese white rabbits under thiopental
To determine the bFGF activity in the Gore grafts, five anesthesia administered intravenously. Six bFGF Gore grafts
bFGF-HPCHA Gore grafts were soaked in 2 ml of a 20 mM were implanted in the left pockets and six bFGF-HPCHA
isotonic phosphate buffer (pH 7.4) in a boron coated screw- Gore grafts in the right pockets; these were removed at 0.5,
cap tube and stored at 5°C for 3 days. The biological activity 1, 2, 4, 6, 8, 16 and 24 h. The removed grafts were placed into
of bFGF was assayed in baby hamster kidney (BHK)-21 cells, phosphate buffer (pH 7.4) at 5°C for 3 days and assayed.
cultured according to Fukunaga et al.13 In brief, BHK-21 Figure 55.6 shows the percent of remaining bFGF in the
cells were seeded at 1 X 103 cells/well with bFGF at various bFGF and bFGF-HPCHA Gore grafts after implantation. In
concentrations. After 3 days in culture, the increase in pro- vivo release of bFGF from the bFGF Gore grafts reached al-
tein contents as a measure for the cell proliferation was de- most 100% at 24 h following implantation, whereas 60% of
termined by the bicinchoninic acid method (BCA Protein bFGF remained in the bFGF-HPCHA Gore grafts at 24 h.
Assay Kit Pierce Co., IL USA) at 595 nm using a microplate Both the animal and in vitro experiments demonstrated that
reader (BIO-RAD model 450). Figure 55.4 shows a typical almost 100% of bFGF was released from the bFGF Gore
proliferation profile of BHK-21 cells treated with bFGF as a grafts after 24 h, while bFGF release from the bFGF-HPCHA
Use of Hydroxypropylchitosan Acetate as a Carrier for Growth Factor Release 601
Gore grafts was sustained. Although the release rate from Effect of bFGF-HPCHA on the
the HPCHA-free graft in the animal experiment was closely Endothelialization of Grafts
correlated to that in the in vitro experiment, a few differ- To evaluate the effect of bFGF on the
ences in results were observed between these experiments. endothelialization of bFGF-HPCHA Gore grafts, bFGF-
In the animal experiment, the release pattern showed an ini- HPCHA and drug-free Gore (control) grafts were implanted
tial lag period of about 1 h compared with the in vitro ex- between artery and vein in dogs. Four adult mongrel dogs
periment. It is probable that a slight boundary layer will con- were anesthetized with intravenous sodium thiopental, and
trol the release. maintained with nitrous monoxide and halothane. Femoral
artery and vein were exposed on both sides and dogs were
heparinized before clamping. bFGF-HPCHA grafts were
interposed between right femoral artery and vein by double
Use of Hydroxypropylchitosan Acetate as a Carrier for Growth Factor Release 603
end to side suture. Control grafts were implanted on the left Further, bFGF-HPCHA Gore grafts are useful for vascular
sides. Specimens were harvested after 6 weeks to investigate reconstruction, because bFGF stimulates regrowth of endo-
proliferation of endothelial cells on the internal surface of thelial cells on the graft surface.
the grafts. Grafts were explanted from anesthetized living
animals, opened longitudinally for observation and fixed for References
light microscopy. Fixed specimens were embedded in paraf- 1. Berger K , Sauvage LR, Rao AM et al. Healing of arterial
fin and stained with hematoxylin and eosin. The histology graft in man; Its incompleteness. Ann Surg 1972;
of bFGF-HPCHA treated sites showed a thicker layer of con- 175:118-127.
nective tissue around the grafts compared with control 2. Schmit S, Hunter T, Falkow L et al. Effects of antiplatelet
agents in combination with endothelial cell seeding on
(Fig. 55.7). The histological examination demonstrated that
small-diameter Dacron vascular graft performance in ca-
the local delivery system of bFGF acts to stimulate re- nine carotid artery model. J Vasc Surg 1985; 2:898-906.
endothelialization. 3. Herring M, Garner A, Peigh P et al. Patency in canine
inferior vena cava grafting: Effects of graft material, size,
Concluding Remarks and endothelial seeding. J Vasc Surg 1984; 1:877-887.
Our work so far has produced evidence to support 4. Gospodarowicz D, Ferrara N, Schweigerer L et al. Struc-
the use of HPCHA as a biodegradable carrier for controlled tural characterization and biological functions of fibro-
release of bFGF. Coupling bFGF to HPCHA succeeded in a blast growth factor. Endocr Rev 1987; 8:95-114.
stable and reproducible way. 5. Saksera O, Moscatelli D, Rifkin DB. The opposing effects
bFGF-HPCHA solution absorbed into Gore grafts was of basic fibroblast growth factor and transforming growth
about 13-fold in weight greater than that of the bFGF solu- factor beta on the regulation of plasminogen activity in
capillary endothelial cells. J Cell Biol 1987; 105:957-963.
tion. It can be assumed that HPCHA solution is held in the
6. Schweigere L, Neufeld G, Friedman J et al. Basic fibro-
pore network of Gore grafts better than water. Calculations blast growth factor: Production and growth stimulation
of the porosity (~80%) of Gore-Tex® have shown that each in cultured adrenal cortex cells. Endocr 1987; 120:796-800.
Gore-Tex® graft (0.75 cm in diameter) contains pores to- 7. Lindner V, Majak RA, Reidy MA. Basic fibroblast growth
taling 0.0226 cm3; accordingly, the amount of water or factor stimulates endothelial regrowth and proliferation
HPCHA solution that could be held in the pores is estimated in denuded arteries. J Clin Invest 1990; 85:2004-2008.
to be roughly 0.0226 ml. 8. Tokura S, Azuma I. Distribution of acetamide group in
Nevertheless, actual experiments with water or partially deacetylated chitin and its biodegradability. De-
chitosan solution content have shown figures that are sub- rivatives in Life Science 1992; 1:86-92.
stantially lower: For water, roughly one-twentieth of the pro- 9. Machida Y, Banba T, Fujimoto M et al. Synthesis and
antitumor activity of chitosan carrying 5-fluorouracils.
jected volume is held; for chitosan solution, about two-thirds
Macromol Chem 1989; 190:1817-1825.
of the projected volume. 10. Nishioka Y, Kyotani S, Okamura M et al. Release charac-
The explanation for this may be that hydrophobic teristics of cisplatin chitosan microspheres and effect of
materials such as Gore-Tex® repel water, making it difficult containing chitin. Chem Pharm Bull 1990; 38:2871-2873.
for the pores in the graft to retain fluid. A high molecular 11. Yamamura K, Sakurai T, Yano K et al. Sustained release
solution like chitosan has greater inherent viscosity, how- of basic fibroblast growth factor from the synthetic vas-
ever, and is thus more likely to be retained within the Gore- cular graft using hydroxypropylchitosan acetate. J Biomed
Tex® pores than is water. Therefore, one effective way to in- Mater Res 1995; 29:203-206.
crease the amounts of drugs contained within Gore-Tex® 12. Yamamura K, Iwata H, Yotsuyanagi T. Synthesis of anti-
pores is to improve retention by increasing the viscosity of biotic-loaded hydroxyapatite beads and in vitro drug re-
fluids through use of high molecular solutions, and its fur- lease testing. J Biomed Mater Res 1992; 26:1053-1064.
13. Fukunaga K, Hijikata S, Ishimura K et al. Aluminium
ther release rate can be controlled by changing the ratio of
!-cyclodextrin sulphate as a stabilizer and sustained-re-
HPCHA. In conclusion, HPCHA could be used as a carrier lease carrier for basic fibroblast growth factor. J Pharm
to control continuous release of unstable substances such as Pharmacol 1994; 46:168-171.
peptide, hydrophilic and lipid-soluble chemicals in vivo.
Cell Engineering
CHAPTER 56
Transplantation of Transduced Smooth Muscle Cells:
A Vehicle for Local and Systemic Gene Therapy
Randolph L. Geary, Alexander W. Clowes, Monika M. Clowes
Introduction
S ince the advent of techniques to introduce recombinant DNA into mammalian tissues, a
new science of gene transfer has emerged with broad application throughout the life sci-
ences.1-3 With it has come the ability to genetically alter vascular smooth muscle cells (SMC)
and endothelial cells in vitro and in vivo, allowing the vascular biologist to ask and answer
questions previously unanswerable.4-16 Over the past decade an intense enthusiasm for the
clinical application of vascular gene transfer has matured into cautious optimism, as the
limitations of vector systems used to introduce and express recombinant genes in vascular
tissues have been better defined.3 Refinements in vector systems have improved the preci-
sion and safety of vascular gene transfer and proof-of-concept experiments have demon-
strated the potential for both local and systemic therapeutic effects.17-30 While the field is
still in its infancy, the first clinical application of vascular gene transfer has been initiated
and the realization of vascular gene therapy may well be at hand.31,32
The use of prosthetic materials in the treatment of cardiovascular diseases is expand-
ing as new devices are created and the indications for applying existing devices are broad-
ened. A significant limitation of all implantable prosthetic materials is biocompatibility.
Prosthetic vascular grafts have been widely used to reconstruct damaged and diseased arter-
ies and veins. Despite extensive research to improve graft function, thrombosis and intimal
hyperplasia continue to limit graft durability, particularly when used to reconstruct small
arteries or veins. Infections also lead to graft failure and significant morbidity and mortality
in a small but significant number of patients. Application of existing gene transfer technol-
ogy to prosthetic vascular devices has thus been directed toward inhibition of neointimal
growth and thrombus formation and toward improving the extent of graft healing to de-
crease the risk of infection.4,27,33-38 If successful, these strategies will improve graft function
and constitute an important advance in patient care and limb salvage.
In the following discussion we will review the use SMCs as hosts for the expression of
therapeutic recombinant genes. A comprehensive review of vascular gene therapy is beyond
the scope of this chapter and we wish to acknowledge the pioneering work by many investi-
gators in this field without which our studies would not have been possible.39 While our
focus has been on the SMC, others have made extraordinary advances in the application of
endothelial cell-based gene transfer and we refer the reader to those important studies for a
more complete appreciation of the potential for vascular gene therapy.
Tissue Engineering of Prosthetic Vascular Grafts, edited by Peter Zilla and Howard P. Greisler.
©1999 R.G. Landes Company.
606 Tissue Engineering of Prosthetic Vascular Grafts
Smooth Muscle Cells as Targets sertion and promoter sequences essential for integration and
for Gene Transfer expression of the recombinant gene within the host cell ge-
SMCs provide an attractive target for gene transfer, as nome remain (Fig. 56.1). As retroviral vectors integrate their
they comprise the majority of cells within the wall of blood recombinant genes directly into the host cell genome, the
vessels and play a central role in vascular diseases such as transgene is expressed throughout the life of the cell and
atherosclerosis and intimal hyperplasia following arterial can be duplicated during mitosis and passed on to daughter
reconstruction. Modifying SMC behavior by recombinant cells. This provides the potential for stable long term ex-
gene transfer then provides the opportunity to limit SMC pression in contrast to the transient expression (days) of
growth, migration and extracellular matrix elaboration, criti- recombinant genes introduced by plasmid and adenoviral
cal components of atherosclerotic plaque or neointima for- vectors.56,57 The lack of viral coding sequences prevents im-
mation. Vessel wall function could also be improved by lo- mune responses towards foreign proteins, which has been a
cal over-expression of genes encoding factors to inhibit vaso- significant problem with first generation adenoviral vectors
constriction, inflammation, and thrombosis. These and other containing a number of wild type coding sequences.58
modifications could translate into improved outcomes for A major limitation of retroviral vectors is that expres-
patients undergoing surgical or endovascular arterial and sion of the recombinant transgene requires prior integra-
venous reconstruction. tion into the host cell genome. Efficient integration in turn
SMCs have characteristics which make them an at- requires that the host cell be replicating at the time of viral
tractive target tissue in which to express recombinant genes transduction.54,55 As SMCs are generally quiescent within
for systemic therapy. Muscle cells in general have proven to the undisturbed artery wall, direct in vivo transduction is
be durable hosts for recombinant gene expression.40-43 SMCs very inefficient.45 Even at sites of vascular reconstruction,
represent a large population of cells separated from the sys- SMC replication is delayed for hours or days after the injury
temic circulation by a single endothelial cell layer. SMCs are caused by the procedure. Retroviral gene transfer directed
generally quiescent in vivo with replication rates of less than at improving the response to surgical or endovascular re-
one percent,44 and as a result SMCs are very long-lived, pro- construction would require a delayed application of virus
viding the potential for stable long term expression of thera- (24-96 h post-procedure). By that time however, the injury
peutic genes. These attributes have led our group and oth- response is well underway with local thrombus formation,
ers to explore the application of SMCs as hosts for local and release of platelet growth factors, and influx of leukocytes
systemic gene delivery. that may overcome any potential benefits of delayed gene
transfer. Vein grafts present a similar obstacle for retroviral
Vectors for SMC Gene Transfer vectors. For these reasons, alternative vectors capable of gene
As noted above, the practical application of vascular transfer into quiescent SMC in vivo are being explored for
gene therapy has been limited by the vector systems used to gene transfer to SMCs at the time of vascular reconstruc-
introduce recombinant genetic material. Virtually all vector tion (e.g., adeno-associated virus and adenovirus).46,48,56
systems have been used successfully to transduce vascular To accommodate the need for cell replication for
SMCs, including viral vectors (retrovirus,4-6,45 adenovi- retroviral vector integration, ex vivo gene transfer of target
rus,46,47 adeno-associated virus,48 and others3); plasmid cells has been performed in culture. SMCs can be readily
DNA;49 liposomal vectors;50 and hybrid systems.51,52 Each established in culture from small artery or vein biopsies and
vector has its own set of limitations, which generally fall into induced to replicate by serum growth factors.14,45,59 Early
the following categories: inefficient in vivo SMC transduc- passage cells can then be exposed to retroviral vectors
tion; inefficient gene expression following transduction; containing antibiotic resistance genes that allow for selection
transient expression due to a lack of genomic integration; of transduced cells by adding antibiotic to the culture
transient gene expression due to an immune response to medium (Fig. 56.1). Populations of transduced cells can then
products of viral coding sequences; and inability to limit be expanded further in culture for later transplantation back
gene transfer to the target site due to systemic spread of re- into the donor vasculature. This strategy requires the use of
combinant material to nontarget tissues. autologous SMCs to prevent immune rejection of trans-
Our group has employed retroviral vectors13,14,43,45,53 planted cells. It also takes time (weeks) to accumulate the
and adeno-associated virus vectors48 to transduce SMCs ex numbers of transduced cells required to achieve local or
vivo and in vivo, respectively. We will review below our ex- systemic effects.
perience with the retroviral vector system for cell-based vas- We have used this strategy successfully for ex vivo gene
cular gene transfer using transplanted SMCs. transfer and subsequent in vivo transplantation of SMCs
expressing a number of vectors (Fig. 56.1). These include
Retroviral Vectors for Ex Vivo reporter proteins (!-galactosidase (!%gal) and human pla-
Arterial Gene Transfer cental alkaline phosphatase (hAP))13,14,43,45 and proteins with
Retroviruses have been used extensively for recombi- local and systemic biological effects (adenosine deaminase,
nant gene transfer in a wide variety of cell types, both in purine nucleoside phosphorylase, erythropoietin, and oth-
vitro and in vivo.1,2,39,54 This system has a number of ad- ers).14,28,29,43,45,53 Initial experiments were performed using
vantages and disadvantages. Retroviral vectors are made in- SMCs cultured from aortas of the syngeneic Fischer 344 rat
capable of self-replication, as all viral coding sequences es- strain. The cells were transduced with the human adenos-
sential for encapsidation have been deleted.55 Only the in- ine deaminase (ADA) gene which codes for a nonsecreted
Transplantation of Transduced Smooth Muscle Cells: A Vehicle for Local and Systemic Gene Therapy 607
protein that can be identified and distinguished from rat the neointima from the underlying media. Lesion size and
ADA by protein gel electrophoresis.45 The transduced cells SMC replication were self-limited and the resulting
were then transplanted into Fischer rat carotid arteries neointima was no larger than that of sham-seeded control
following endothelial cell denudation. This strategy results arteries. Stenosis did not occur and transduced cells were
in the immediate formation of a nonocclusive neointima not found outside of the transduced artery wall or in loca-
composed entirely of transduced SMCs (Fig. 56.2). Arteries tions beyond the site of initial transplantation (Fig. 56.2).
were removed at various times following seeding and sig- These results underscore the durability and apparent safety
nificant human ADA expression was documented as late as of SMC-based retroviral vascular gene transfer.
6 months after carotid seeding.45 A follow up study in the Subsequent experiments have demonstrated that this
same model demonstrated effective retroviral vector expres- technique can be used to achieve a potentially therapeutic
sion by seeded SMCs even at one year from the time of trans- systemic response.28,29,53 SMCs were transduced with a
plantation (see below), documenting the durability of the retroviral vector encoding the rat erythropoietin gene and
retroviral approach.43 then transplanted successfully into the rat carotid artery as
While these experiments and those of others4,6 estab- described above. The small number of cells transplanted into
lished the proof-of-concept for ex vivo transduction of carotid arteries (105 cells) produced enough erythropoietin
SMCs, questions regarding the behavior of transduced SMCs to significantly increase erythrocyte production, causing an
following transplantation remained unanswered. For in- increase in reticulocyte counts followed by increases in he-
stance, retroviral transduction preferentially occurs in rep- matocrit and hemoglobin.
licating cells. If cells selected in culture for replication were We have also demonstrated that local gene expression
more prone to replicate in vivo, intimal hyperplasia could within the injured artery wall can inhibit intimal hyperpla-
occur and lead to lumen narrowing in treated vessels or sia. SMCs were transduced with a retroviral vector express-
grafts. Altered migration or matrix elaboration by trans- ing the gene encoding tissue inhibitor of matrix
duced cells would also have adverse effects in vivo. metalloproteinase-1 (TIMP-1). TIMP-1 blocks metallo-
To address these concerns, subsequent experiments proteinase activity, limiting extracellular matrix degradation
were performed using a vector encoding the hAP marker essential for cell migration and replication. TIMP-1
gene to localize transduced cells within seeded rat carotid overexpression impaired migration of transduced cells in
arteries and surrounding tissues (Figs. 56.1 and 56.2).43 culture and, when seeded into rat carotid arteries after bal-
Bromodeoxyuridine was administered to label replicating loon injury, inhibited neointima formation.28 Allaire from
cells, and arteries were removed for analysis at various times our group has used this same TIMP-1 vector to inhibit an-
following transduction. Seeding led to a nonocclusive in- eurysm formation and rupture in rat aortic xenografts
tima composed entirely of transduced SMCs expressing the (guinea pig into rat).29 All grafts transplanted without SMC
AP gene product (Fig. 56.2). Within 2 weeks the neointima seeding developed aneurysms which ruptured between 4 and
also contained nontransduced SMCs that had migrated into 21 days. In contrast, none of the grafts seeded with SMCs
608 Tissue Engineering of Prosthetic Vascular Grafts
Fig. 56.2. Composite photomicrograph of rat carotid artery cross sections removed at 1 week, 2 weeks or 12 months following
transplantation of smooth muscle cells transduced with a retroviral vector encoding the alkaline phosphatase (AP) reporter gene
(dark blue reaction product). At 1 week a nonocclusive intima has been created, with seeded cells expressing the AP gene product.
Within 2 weeks cells have migrated into the neointima from the injured media so that the final intimal lesion is composed of a
mixture of transduced (blue stain) and nontransduced smooth muscle cells. Neointimal area is maximal 4-6 weeks after seeding
and expression of the AP reporter gene is durable, with long term expression shown in an artery removed 12 months following
transplantation.
expressing TIMP-1 developed aneurysms. Grafts seeded with prior to implantation. Autologous SMCs were obtained from
SMCs transduced with a vector encoding only the neo gene baboons via saphenous vein biopsy, and early passage SMCs
(LXSN, Fig. 56.1) all developed aneurysms within 4 weeks were established in culture using standard techniques.14,59
of transplantation, and 2 of 6 ruptured. These results have Cells were then exposed to replication-deficient retroviral
prompted us to extend studies of retroviral vascular gene particles encoding the human placental AP gene coupled to
transfer to other animal models. the neomycin resistance gene (Fig. 56.1). Cells were initially
selected in neomycin, then expanded and within 4-6 weeks
Retroviral Vectors for SMC Gene Transfer of obtaining the vein biopsy, 107 to 109 transduced SMC were
in Prosthetic Vascular Grafts available for PTFE graft seeding (Fig. 56.3). Transduced
As described in the accompanying chapters, prosthetic SMCs were then suspended in a solution of type I collagen
vascular grafts present a unique set of challenges when used and the mixture injected into the lumen of 10 cm long, 4 mm
to reconstruct damaged or occluded arteries and veins. Pros- diameter, PTFE grafts (60 ∝m internodal distance, WL Gore
thetic conduits are more prone to infection than are autolo- Corp., Flagstaff, AZ). Collagen was used to facilitate cell re-
gous grafts and suffer disproportionately from thrombosis tention and adhesion to the PTFE graft material. The cell
and neointima formation at anastomoses. This is particu- suspension was gently infused into the graft lumen and ex-
larly true when grafts are used to reconstruct small arteries truded through graft wall interstices (Fig. 56.3). Seeded grafts
and veins. Prosthetic grafts heal by neointimal ingrowth from were incubated overnight and then grafted end-to-side into
the cut arterial ends at anastomoses. The neointima extends the aorto-iliac position of the original donor baboon.
a limited distance onto the graft lumen surface depending Grafts were allowed to heal for 3-6 weeks, a time pe-
on graft porosity. In more porous grafts, neointima may form riod when an unseeded PTFE graft would have formed a
along the entire lumen surface by ingrowth of granulation significant neointima along its entire length.60-62 In the first
tissue and microvessels directly through graft wall inter- experiments, grafts were more prone to thrombosis due to
stices.60-62 The resulting graft neointima is made up largely exposure of the collagen gel to flowing blood. In subsequent
of SMCs and their surrounding extracellular matrix beneath experiments autologous endothelial cells were cultured from
an endothelial cell monolayer. each vein biopsy. The endothelial cells were not transduced,
SMCs then provide a direct target for gene therapy to but were seeded onto the lumen surface of PTFE grafts fol-
reduce graft intimal hyperplasia and neointimal formation. lowing SMC seeding. This strategy eliminated the problem
Indirect targets include reducing graft thrombogenicity and of increased graft thrombosis. When grafts were removed
improving the resistance of graft material to infection. More- for analysis, a significant population of transduced SMCs
over, given that prosthetic grafts heal by accumulating SMCs, was localized within the graft wall by histochemical staining
we hypothesized that preseeding grafts with large numbers for the hAP gene product (Fig. 56.3). To our surprise, the
of genetically modified SMCs could provide a device for sys- neointima which had formed was devoid of transduced cells,
temic delivery of recombinant gene products capable of indicating that, while they survived the transplantation and
treating inherited or acquired disorders. graft implantation process, cells appeared to stay where they
Using a previously established nonhuman primate were initially seeded and did not migrate into the forming
model of PTFE bypass grafting,60-62 we sought to establish neointima. In addition, transduced SMCs were not recruited
genetically modified autologous vascular SMCs within grafts by microvessels forming within the graft wall granulation
Transplantation of Transduced Smooth Muscle Cells: A Vehicle for Local and Systemic Gene Therapy 609
tissue. Perhaps more importantly, transduced SMCs were with specific matrix molecules.63 By implanting cells in
not found outside of the graft wall within the fibrous cap- unique matrix environments, cell growth and adhesion may
sule around the graft or in the vasculature of lower extrem- be enhanced, as it is well established that SMC behavior in
ity muscle beds beyond the seeded interposition grafts.13 vitro and in vivo is altered by adhesion to various matrix
These experiments were repeated using a separate re- components.63,64
porter gene in order to establish that the healing character- Ex vivo techniques, while suitable for elective modifi-
istics of seeded grafts were not influenced by expression of cation of bioprostheses, are not applicable to the majority
the hAP gene. In the follow up study we employed a retroviral of clinical procedures. Furthermore, while retroviral vectors
vector encoding the !-gal reporter gene (Fig. 56.1).14 The have been used in a number of early human gene therapy
same seeding technique was employed and grafts were left trials,2 concerns about their safety remain. Retrovirus vec-
in place for 4-5 weeks and then removed for analysis. The tors integrate randomly into host cell DNA, leading to the
findings were essentially the same. Transduced cells were possibility of latent oncogene activation.2,54 Better vectors
located within the interstices of each seeded graft and cells are being created for direct in vivo vascular gene transfer
were not seen within the neointima or in the surrounding that are more simple to use and more durable than current
perigraft tissues (Fig. 56.3). vectors. Sophisticated systems have been designed to include
transcription initiation sequences that can be turned on and
Unanswered Questions and Future Directions off in the presence or absence of specific cofactors.65 Once
While the results of SMC-based gene transfer in these systems are available, concerns over the lack of control
experimental models are encouraging, a number of ques- of the magnitude and duration of gene expression may be
tions remain to be addressed. Long term expression of alleviated. Device technology is also helping to make clini-
recombinant genes by rat SMCs must be reproduced in cal application of vectors a reality through inventions such
models likely to better represent the human situation. This as through-lumen and spray catheters.66
is critical, as the long term biological behavior of transduced While still in its infancy the first application of vascu-
human SMCs is unknown. Strategies to improve SMC re- lar gene transfer to improve patient care is on the horizon.
tention upon transplantation are also needed in order to Use of this technology to improve the biocompatibility and
apply this strategy effectively to human prosthetic devices.38 function of prosthetic vascular devices has been initi-
This problem may be overcome as we learn more about the ated14,23,27,34,35,37,38 and if successful a revolution in vascular
role of cell surface integrins and their adhesive interactions reconstruction should follow.
610 Tissue Engineering of Prosthetic Vascular Grafts
31. Isner JM, Walsh K, Symes J, Pieczek A, Takeshita S, Lowry 46. Lemarchand P, Jaffe HA, Danel C, Cid MC, Kleinman HK,
J, Rossow S, Rosenfield K, Weir L, Brogi E, Schainfeld R. Stratford-Perricaudet LD, Perricaudet M, Pavirani A,
Arterial gene therapy for therapeutic angiogenesis in pa- Lecocq JP, Crystal RG. Adenovirus-mediated transfer of
tients with peripheral artery disease. Circulation 1995; recombinant human alpha 1-antitrypsin cDNA to human
91:2687-2692. endothelial cells. Proc Natl Acad Sci USA 1992;
32. Isner JM, Pieczek A, Schainfeld R, Blair R, Haley L, 89:6482-6486.
Asahara T, Rosenfield K, Razvi S, Walsh K, Symes JF. 47. Lee SW, Trapnell BC, Rade JJ, Virmani R, Dichek DA. In
Clinical evidence of angiogenesis after arterial gene trans- vivo adenoviral vector-mediated gene transfer into bal-
fer of phVEGF165 in patient with ischaemic lim. Lancet loon-injured rat carotid arteries. Circ Res 1993;
1996; 348:370-374. 73:797-807.
33. Dichek DA, Neville RF, Zwiebel JA, Freeman SM, Leon 48. Lynch CM, Hara PS, Leonard JC, Williams JK, Dean RH,
MB, Anderson WF. Seeding of intravascular stents with Geary RL. Adeno-associated virus vectors for vascular gene
genetically engineered endothelial cells. Circulation 1989; delivery. Circ Res 1997; 80:497-505.
80:1347-1353. 49. Riessen R, Rahimizadeh H, Blessing E, Takeshita S, Barry
34. Flugelman MY, Virmani R, Leon MB, Bowman RL, JJ, Isner JM. Arterial gene transfer using pure DNA ap-
Dichek DA. Genetically engineered endothelial cells re- plied directly to a hydrogel-coated angioplasty balloon.
main adherent and viable after stent deployment and ex- Hum Gene Ther 1993; 4:749-758.
posure to flow in vitro. Circ Res 1992; 70:348-354. 50. Leclerc G, Gal D, Takeshita S, Nikol S, Weir L, Isner JM.
35. Huber TS, Welling TH, Sarkar R, Messina LM, Stanley Percutaneous arterial gene transfer in a rabbit model. Ef-
JC. Effects of retroviral-mediated tissue plasminogen ac- ficiency in normal and balloon-dilated atherosclerotic ar-
tivator gene transfer and expression on adherence and teries. J Clin Invest 1992; 90:936-944.
proliferation of canine endothelial cells seeded onto ex- 51. Morishita R, Gibbons GH, Kaneda Y, Ogihara T, Dzau
panded polytetrafluoroethylene. J Vasc Surg 1995; VJ. Novel in vitro gene transfer method for study of local
22:795-803. modulators in vascular smooth muscle cells. Hyperten-
36. Flugelman MY: Inhibition of intravascular thrombosis and sion 1993; 21:894-899.
vascular smooth muscle cell proliferation by gene therapy. 52. Dzau VJ, Mann MJ, Morishita R, Kaneda Y. Fusigenic
Thrombosis & Haemostasis. Thromb Haemost 1995; viral liposome for gene therapy in cardiovascular diseases.
74:406-410. Proc Natl Acad Sci USA 1996; 93:11421-11425.
37. Dichek DA, Anderson J, Kelly AB, Hanson SR, Harker 53. Osborne WRA, Ramesh N, Lau S, Clowes MM, Dale DC,
LA. Enhanced in vivo antithrombotic effects of endothe- Clowes AW. Gene therapy for long-term expression of
lial cells expressing recombinant plasminogen activators erythropoietin in rats. Proc Natl Acad Sci USA 1995;
transduced with retroviral vectors. Circulation 1996; 92:8055-8058.
93:301-309. 54. Morgan JR, Tompkins RG, Yarmush ML. Advances in
38. Scott-Burden T, Tock CL, Schwarz JJ, Casscells SW, Engler recombinant retroviruses for gene delivery. Adv Drug
DA. Genetically engineered smooth muscle cells as lin- Deliv Rev 1993; 12:143-158.
ings to improve the biocompatibility of cardiovascular 55. Miller AD, Rosman GJ. Improved retroviral vectors for
prostheses. Circulation 1997; 94:II-235-II-238. gene transfer and expression. BioTechniques 1989;
39. Clowes AW. Vascular gene therapy in the 21st century. 7:980-990.
Thromb Haemost 1997; 78:605-610. 56. Schulick AH, Newman KD, Virmani R, Dichek DA. In
40. Dai Y, Roman M, Naviaux RK, Verma IM. Gene therapy vivo gene transfer into injured carotid arteries: Optimi-
via primary myoblasts: Long-term expression of factor IX zation and evaluation of acute toxicity. Circulation 1995;
protein following transplantation in vivo. Proc Natl Acad 91:2407-2414.
Sci USA 1992; 89:10892-10895. 57. Feldman LJ, Steg PG, Zheng LP, Chen D, Kearney M,
41. Isaka Y, Brees DK, Ikegaya K, Kaneda Y, Imai E, Noble McGarr SE, Barry JJ, Dedieu J-F, Perricaudet M, Isner JM.
NA, Border WA. Gene therapy by skeletal muscle expres- Low-efficiency of percutaneous adenovirus-mediated ar-
sion of decorin prevents fibrotic disease in rat kidney. terial gene transfer in the atherosclerotic rabbit. J Clin
Nature Med 1996; 2:418-423. Invest 1995; 95:2662-2671.
42. Blau HM, Springer ML. Muscle-mediated gene therapy. 58. Newman KD, Dunn PF, Owens JW, Schulick AH, Virmani
N Engl J Med 1995; 333:1554-1556. R, Sukhova G, Libby P, Dichek DA. Adenovirus-mediated
43. Clowes MM, Lynch CM, Miller AD, Miller DG, Osborne gene transfer into normal rabbit arteries results in pro-
WRA, Clowes AW. Long-term biological response of in- longed vascular cell activation, inflammation, and
jured rat carotid artery seeded with smooth muscle cells neointimal hyperplasia. J Clin Invest 1995; 96:2955-2965.
expressing retrovirally introduced human genes. J Clin 59. Libby P, Obrien KV. Culture of quiescent arterial smooth
Invest 1994; 93:644-651. muscle cells in a defined serum-free medium. J Cell
44. Clowes AW, Reidy MA, Clowes MM. Kinetics of cellular Physiol 1984; 115:217-223.
proliferation after arterial injury.I.Smooth muscle growth 60. Clowes AW, Kirkman TR, Reidy MA. Mechanisms of ar-
in the absence of endothelium. Lab Invest 1983; terial graft healing. Rapid transmural capillary ingrowth
49:327-333. provides a source of intimal endothelium and smooth
45. Lynch CM, Clowes MM, Osborne WRA, Clowes AW, muscle in porous PTFE prostheses. Am J Pathol 1986;
Miller AD. Long-term expression of human adenosine 123:220-230.
deaminase in vascular smooth muscle cells of rats: A 61. Golden MA, Hanson SR, Kirkman TR, Schneider PA,
model for gene therapy. Proc Natl Acad Sci USA 1992; Clowes AW. Healing of polytetrafluoroethylene arterial
89:1138-1142. grafts is influenced by graft porosity. J Vasc Surg 1990;
11:838-845.
612 Tissue Engineering of Prosthetic Vascular Grafts
62. Geary RL, Kohler TR, Vergel S, Kirkman TR, Clowes AW. 65. Wang Y, O’Malley BW,Jr., Tsai SY, O’Malley BW. A regu-
Time course of flow-induced smooth muscle cell prolif- latory system for use in gene transfer. Proc Natl Acad Sci
eration and intimal thickening in endothelialized baboon USA 1994; 91:8180-8184.
vascular grafts. Circ Res 1994; 74:14-23. 66. Wilensky RL, March KL, Hathaway DR. Direct intraarte-
63. Ruoslahti E, Engvall E. Integrins and vascular extracellu- rial wall injection of microparticles via a catheter: A po-
lar matrix assembly. J Clin Invest 1997; 99:1149-1152. tential drug delivery strategy following angioplasty. Am
64. Vernon RB, Sage EH. Between molecules and morphol- Heart J 1991; 122:1136-1140.
ogy—Extracellular matrix and creation of vascular form.
Am J Pathol 1995; 147:873-883.
Angioplasty, 73, 76, 93, 146, 165, 229-231, 233, 235-240,
Index 242, 249, 251-253, 263, 284, 292, 353, 363, 367, 369,
380, 383, 385, 414, 569, 589, 595, 610-612
Angiotensin converting enzyme (ACE), 158, 232, 233 408
Angiotensin II, 231-233, 238, 239, 242, 253, 255, 257, 265,
Symbols 266, 301, 408, 414
Angiotropin, 280, 302
150.95 protein, 107, 108 Antioxidants, 470, 471, 473
Antithrombin, 63, 115, 117, 389-391, 467, 479, 546, 549,
A 551, 568
#-actinin, 255, 334, 358, 427, 585, 588 Antithrombin complex (TAT), 117, 389-391, 398, 465, 546
#-granule, 28-30, 231, 232, 253, 254 AP-1, 430, 431, 436, 438
#-smooth muscle actin, 105, 407, 408 APAAP technique, 107, 109, 110
#1!1, 272-274, 334, 357, 360, 361, 365, 590 Apoptosis, 44, 122, 164-166, 168, 169, 238, 245, 276, 291,
#2-macroglobulin/low density lipoprotein receptor, 315 324, 335, 341, 411, 416, 593
#4!1, 220, 272, 273, 334, 566, 578-580, 590 Arachidonic acid derivatives, 157
#5!1, 256, 273, 334, 335, 356, 357, 428, 578, 590 Atherectomy, 93, 132, 138, 142, 230, 231, 233, 236,
#6!1, 273, 334, 335, 590 239, 364
Acetylated LDL, 110, 364 Atherosclerotic lesion, 140, 180, 229, 230, 233, 235, 237,
Acidic fibroblast growth factor (aFGF), 32, 158, 159, 162, 251, 252, 254, 410, 415, 433
202, 264, 310, 311, 316, 318, 495 Atherosclerotic plaque, 33, 183, 185, 191, 230, 233, 235,
Acidosis, 279 238, 246, 252, 253, 263, 265, 316, 323, 355, 357, 360,
Actin filaments, 104, 183, 185, 254, 255, 268, 331, 332, 335, 414, 492, 606
354, 535, 585 ATIII, 481, 482, 546, 548-551
Actin stress fiber, 254, 258 Autologous cell(s), 73, 80, 163
Acute inflammatory response, 43, 198, 207-209, 212, 220 #v!3, 233, 239, 272-274, 316, 318, 320, 323, 324, 334, 335,
#d!2, 173 341, 355-357, 360, 361, 559, 588, 590-592, 594,
Adenosine diphosphate (ADP), 116, 255, 258 596, 597
Adenosine monophosphate (AMP), 19, 25, 26, 32, 34-36, #v!5, 434
39, 40, 44, 116, 159, 162, 168, 323, 341, 405, 429, 430,
B
434
Adenovirus, 65, 68, 234, 245, 249, 606 !-galactosidase, 65, 606, 607, 609
Adenylyl cyclase, 159, 429, 430, 434, 435 Balloon angioplasty, 142, 242, 249, 252, 253, 263, 367, 414,
Adipocytes, 95, 96, 102, 103, 105, 107, 108, 110, 111, 124, 569, 589, 610
133, 372, 406 Balloon catheter, 73, 76, 142, 230, 238, 239, 249, 265, 284,
Adipose tissue, 62, 94, 100, 102, 112, 113, 115, 121-124, 292, 364, 595
144, 189, 308 Basic fibroblast growth factor (bFGF), 27, 29, 32, 34, 35,
Adventitia, 6, 9, 10, 12, 20, 22, 23, 25-27, 29, 31, 34, 36, 87, 42-44, 124, 138, 142, 159-164, 168, 190, 202, 203,
197-200, 202, 203, 230, 274, 326, 355, 357, 382, 407, 231-233, 236-239, 242, 243, 248, 252, 253, 257,
408, 411, 425, 441, 443, 452, 458, 490, 497, 498, 510 260-262, 264, 269, 272-275, 277, 285, 294, 299,
Affinity chromatography, 487, 590 310-313, 316-320, 322-324, 335-337, 339, 341, 342,
Aggregation, 40, 42, 61, 82, 116, 158, 172, 231, 233, 256, 351, 356, 364, 415, 416, 435, 479, 495, 496, 502, 550,
267, 334, 339, 480, 502, 507, 509, 585, 590 569, 599-604
Albumin, 31, 199, 207-211, 214-216, 222, 234, 345, 478, Biophilic polymers, 481-483, 487
479, 481, 488, 509, 543, 546, 551, 559, 568, 570, Bioprostheses, 88, 155, 172, 176, 287, 288, 295, 609
577, 580 Bone morphogenetic protein-3, 294, 295, 299
Alginate, 506, 562, 563 Bovine aortic endothelial cell (BAEC), 80-83, 85-89, 176,
Anastomotic hyperplasia, 39, 40, 57, 62, 65, 163, 200, 203, 272, 302, 323, 340, 342, 428-432, 436, 437, 555, 580
248, 482, 555, 558 Burst strength, 482
Aneurysm, 20, 50, 51, 53, 54, 58, 102, 271, 274, 275, 277,
443, 482, 489, 490, 492, 494, 497-500, 508, 509, C
607, 608
c-fos, 238, 243, 245, 248, 431, 436
Angioblast, 373, 374
c-myb, 244, 245, 249, 415, 570
Angiogenin, 302
c-myc, 238, 243-245, 248, 249, 409
Angiography, 47-49, 57, 58, 133, 134, 136, 180, 283,
C3b, 202
284, 295
C3bi, 32, 202, 204
C5a, 199, 208, 216
614 Tissue Engineering of Prosthetic Vascular Grafts
Calf/Calves, 11, 16, 18, 407 Collagen I, 180, 254, 287, 289-293, 296, 298, 330, 332,
Canine model, 18, 40, 41, 76, 77, 89, 96, 97, 100, 131, 338-340, 343,-345, 347, 348, 350, 352, 355, 360, 366,
140-142, 168, 186, 236, 369 367, 408, 414, 494, 545, 550, 575
Capillary sinuses, 10, 12-14, 33, 35 Collagen IV, 80, 180, 254, 340, 360
Cardiac myocytes, 403, 407 Collagen-gag copolymers, 571
Carmeda biologically active surface (CBAS), 546 Collagenase, 33, 35, 42, 62, 67, 96, 100, 102, 108, 110, 122,
Carotid artery, 47, 48, 96, 97, 133, 135, 231, 234-238, 246, 124, 133, 144, 157, 159, 163, 164, 189, 190, 219, 225,
248, 249, 252-254, 265, 284-286, 312, 323, 338, 355, 239, 280, 281, 288, 293, 296, 302, 310, 316, 323, 336,
359, 369, 409, 457, 458, 500, 596, 604, 607, 608, 339, 342, 346, 348, 350, 352, 354, 364, 567, 572, 575
610, 611 Colony stimulating factors (CSF), 202-204, 374, 375, 408
Catecholamines, 231-233 Colony stimulating factor-1 (CSF-1), 374
Cathepsin, 219, 225, 410 Complement activation, 199, 204, 206, 208, 209, 216,
Cd11a, 204, 220 513, 551
Cd11a/cd18, 173, 204 Compliance, 15, 24, 25, 36, 45-48, 50-58, 129, 132, 138,
Cd11b, 31, 32, 42, 43, 204, 205, 213, 217, 220 153, 186, 441, 442, 447, 451-456, 458, 464, 470, 476,
Cd11b/cd18, 32, 42, 204, 205, 213, 217 477, 485, 494, 497, 498, 500-502, 509, 511, 513, 555,
Cd11c/cd18, 204, 205 559, 578, 581, 592
CD14, 393, 405 Compliance mismatch, 36, 45, 46, 50, 51, 56, 58, 132, 456,
CD18, 173, 176, 177, 204, 205, 213, 217, 220, 225, 226 578, 592
CD25, 396 Contact inhibition, 157, 185
CD3, 393, 396, 399 Copolymers, 43, 215, 484, 487, 490, 491, 497, 500, 502,
CD31, 106-109, 111, 113, 124, 158, 173, 174, 180, 272, 273, 506, 507, 510, 511, 529, 551, 555, 557, 559, 562, 566,
275, 373 568, 569, 571
CD32, 405 Corona treatment, 485
CD34, 102, 104-110, 113, 124, 139, 180, 272, 273, 275, 276, Coronary artery bypass graft (CABG), 93, 98-100, 143,
373, 375-378, 384, 388, 390, 392, 393, 397, 401, 146, 180, 247-249, 389
404, 412 Creep, 444, 448, 452, 470
Cd34, 375 Crush resistance, 482
Cd35 receptor, 204 Cyclic adenosine monophosphatase (cAMP), 116, 159,
CD4, 222, 226, 227, 389, 393, 395, 396, 405 160, 162, 263, 358, 429-431, 435, 438
CD40, 389, 395, 397 Cyclic RGD peptides, 592
CD45, 375, 376 Cyclin-dependent kinases, 244, 249
CD68, 31, 182, 393, 399 Cyclooxygenase, 234, 240, 316, 438, 610
Cdk1, 244-246, 249 Cytoplasmic extensions, 101, 104, 105
Cdk2, 244, 245, 249 Cytoplasmic granules, 95, 232
Ceramide sphingosine, 255 Cytoskeletal complexes, 585, 586
Chacma baboon, 5-10, 13, 14, 16-18, 27, 35, 36 Cytoskeletal elements, 75, 101, 589
Chain scission, 484 Cytoskeleton, 65, 75, 140, 175, 233, 254-259, 261, 262,
Chemical vapor deposition, 485 266-268, 272, 290, 291, 323, 334, 335, 341, 352-354,
Chemokinesis, 201, 354 359, 426, 427, 540-543, 585, 587, 588, 593
Chemokinesis, 201, 252, 277, 355, 356
Chemotaxis, 31, 198, 201, 202, 208, 212, 213, 216, 222, 223, D
226, 251-254, 256, 260-262, 264, 265, 269, 271-277, Dacron, 4, 9-22, 24-27, 30-34, 36-41, 50, 51, 62, 67, 69-73,
326, 327, 334, 353-361 75-77, 79, 80, 88, 89, 98-100, 119, 120, 123, 125, 126,
Chloramphenicol acetyltransferase, 65 128, 130, 133, 135, 136, 138, 140-144, 154, 167, 168,
Cholera toxin, 159, 160, 358 172, 187, 193, 197-204, 207, 209, 214, 216, 240, 243,
Cholesterol, 129, 142, 165, 166, 168, 230, 235, 237, 240, 263, 282, 285, 308, 311, 312, 371, 372, 377-386, 388,
248, 254, 265, 363-365, 369, 410, 416, 495, 496, 501, 397, 408, 423, 425, 441, 451, 458, 463, 478, 482,
502, 511, 541 490-496, 499-502, 509, 511, 545, 553, 570, 577, 580,
Chondroitin-sulfate proteoglycans, 337 594, 604
CK18, 107, 110 Degradation, 27, 29-33, 36, 42, 67, 112, 116, 159, 161, 179,
CK19, 107 199-201, 207, 210, 215, 254, 271, 279-282, 287-289,
CK7, 107 302, 304, 313, 315-317, 321-323, 332, 336, 342, 364,
CK8, 107 390, 460-462, 466, 467, 470-479, 482, 484, 486, 489,
Coagulation factor(s), 27, 117, 158, 209, 463 490, 497, 498, 502, 503, 505, 508, 510, 511, 513, 514,
Collagen gel, 272, 281, 289, 290, 293-295, 297, 298, 330, 526-530, 538, 564, 566-569, 572, 574-576, 607
335, 336, 339, 343-352, 433, 569, 608, 609
Index 615
Desmin, 104, 105, 107, 108, 139, 180, 383, 407 Extracellular matrix (ECM), 3, 27-30, 34, 41, 44, 51, 52, 61,
Diabetes, 166, 169, 238, 313, 569 63, 67, 68, 71, 72, 75-77, 80, 81, 84-87, 89, 90, 101,
Diacylglycerol, 358, 429, 437 104, 122, 126, 131, 137, 158, 160, 173, 184, 191, 200,
Diapedesis, 174, 214, 220, 221, 223, 302 220-222, 231-234, 236, 239, 242, 252, 254, 255, 257,
Differential scanning calorimetry, 208, 212, 216 265-267, 269, 271-277, 280, 281, 285, 287, 289, 291,
Dog(s), 7, 10-12, 18, 39, 40, 66, 75, 89, 97, 99, 119, 120, 292, 294-299, 304-307, 311, 316, 320, 322, 323,
130, 132, 133, 140, 141, 159, 162, 168, 187, 193, 198, 326-341, 343, 348, 351-359, 360, 363-370, 372, 394,
236, 238, 240, 243, 254, 272-274, 276, 283, 308, 309, 407-409, 412, 413, 415, 416, 427, 428, 433, 434, 437,
311, 312, 324, 328, 336, 337, 339, 371, 372, 377, 438, 441, 465, 485, 489, 539-542, 544, 554, 558, 559,
379-383, 385, 408, 415, 457, 458, 463, 479, 482, 499, 561, 562, 566, 567, 569, 571-580, 583-594, 596, 597,
500, 502, 509, 511, 516, 522, 529, 535, 544, 550, 578, 606-608, 610, 612
590, 597, 602 Exudation, 198
DOT immunoassay, 110, 111
Duplex sonography, 190 F
Dynabeads, 102, 104, 106, 110, 113, 126, 161 Factor VIII, 13, 14, 68, 95, 107-111, 129, 130, 133, 140, 180,
E 199, 375, 382, 492
Factor VIII-related antigen, 95
E-selectin, 74, 172-174, 272-277, 316, 398, 401 Factor XIIIa, 563
E2F, 244-246, 249 Fallout endothelialization, 18, 40, 236, 240, 372, 377, 380,
ED 1, 33, 516, 519, 522, 525 381, 385
ED 2, 33, 516, 519, 522, 525 Fetal liver kinase-1 receptor (Flk-1), 260, 264, 268, 275,
ED 3, 516, 520, 522, 525 324, 373-375, 378, 405, 406, 413
Elastase, 219, 225, 320, 336 FGF receptor (FGFR), 142, 159, 304-307, 597
Elastic response, 443, 444, 447-449, 458, 503 Fiber density, 28
Elastin, 3, 17, 52, 55, 225, 233, 291, 336, 355, 407, 434, Fibrin, 4-7, 9-16, 18, 25-38, 40-43, 63, 67, 71, 74, 76, 77,
443-445, 492, 497, 498, 501, 541, 575, 577-580, 589 80-83, 85-87, 89, 93, 100, 112, 115, 117-120, 129, 135,
Elastomers, 44, 467, 470, 474-477, 479, 480, 484, 486, 139, 141, 154, 158, 161, 162, 168, 169, 171, 172, 175,
498, 529 176, 179-181, 186, 187, 190, 191, 193, 194, 199, 201,
Electrostatic endothelial cell seeding, 62, 64, 65-67 204-217, 232, 236, 238, 240, 272, 273, 275-277, 280,
ELISAs, 201 281, 283-285, 289, 295, 298, 308, 312-314, 318-324,
EN4, 106, 107, 110, 113, 139 327, 329, 332, 334-337, 339-341, 345, 346, 351, 382,
Endarterectomy, 73, 76, 77, 131, 138, 230, 231, 237, 353, 387-390, 397, 398, 401, 418, 421, 423, 479, 492, 493,
357, 369 497, 499, 509, 532, 539, 543, 546, 550, 551, 554, 563,
Endoglin, 272-274, 276 567-570, 577, 584, 590, 596, 597
Endothelial cell growth supplement (ECGS), 94, 158, Fibrin clots, 41, 324, 337, 339, 567
159, 161 Fibrin coagulum, 5, 26, 30, 492, 493
Endothelial cell seeding, 17, 38, 40, 51, 55, 61-67, 72, 73, Fibrin glue, 67, 71, 76, 80, 81, 89, 100, 154, 168, 176, 180,
75-77, 89, 90, 94, 99, 100, 111, 125, 126, 128-132, 141, 187, 190, 191, 193, 194, 201, 206, 240, 283-285, 295,
152, 154, 161, 167, 168, 176, 194, 369, 377, 467, 511, 308, 312, 324, 499, 577
558, 604 Fibrinogen, 26-32, 34, 36-41, 43, 74, 81-83, 89, 141, 199,
Endothelial cell transplantation, 62, 63, 66, 79, 90-100 201, 204, 207-217, 272, 273, 275-277, 308, 320, 324,
Endothelin-1, 238, 365, 428, 431, 437, 438 334, 335, 340, 390, 543, 546, 551, 554, 563, 568-570,
Endothelium derived relaxing factor (EDRF), 132, 141, 584, 590, 596, 597
159, 162 Fibrinolysis, 29, 31, 42, 169, 171, 175, 238, 314, 321, 323,
Endothelium-specific cytoplasmatic organelles, 140 387-390, 398, 465, 570
Entactin, 272, 339, 590 Fibroblast, 7, 9-13, 15, 17, 25, 27-29, 35, 42-44, 63, 64, 67,
Epidermal growth factor (EGF), 158, 164, 202, 203, 231, 68, 74-77, 86, 87, 89, 96, 102-104, 106, 113, 115, 122,
252, 253, 256, 260, 264, 265, 268, 289, 290, 294, 295, 124, 129, 138, 139, 142, 158-162, 164, 167, 168, 172,
298, 318, 353, 356, 359, 374, 551, 552 175, 183-187, 190, 199-207, 214, 219, 221, 223, 226,
Erythropoietin, 376, 378, 406, 407, 413, 606, 607, 611 231, 232, 237-239, 242, 248, 252, 253, 255-258, 263,
Ethylene oxide, 214, 464, 467, 472, 487, 500, 502, 551, 570 264, 266-269, 272-275, 277, 280, 281, 285, 288, 290,
Expanded polytetrafluoroethylene, 4-13, 15-25, 27, 30-40, 294, 297-299, 301, 303-306, 308-313, 316, 317, 322,
58, 62, 66-68, 76, 88, 90, 100, 113, 119, 122, 126, 128, 323, 324, 330, 332, 335, 338, 339, 341-353, 358, 372,
130, 141, 142, 154, 155, 160, 167, 168, 176, 187, 193, 389, 390, 403, 406-411, 413-423, 443, 466, 479, 485,
207, 208, 237, 308, 311, 312, 370, 386, 478, 525, 570, 490-495, 502, 509, 510, 516, 521, 529, 532-534,
577, 600, 611 536-540, 542, 543, 550, 552, 554, 558, 559, 569, 570,
572-574, 578, 588-591, 593-595, 599, 600, 604
616 Tissue Engineering of Prosthetic Vascular Grafts
Fibronectin, 27, 33, 40, 41, 63, 65-68, 71, 72, 74, 76, 77, 80, Granulocyte-macrophage colony-stimulating factor, 414
85, 89, 99, 104, 106-108, 122, 154, 158, 165, 168, Great saphenous vein, 154, 159, 161, 168, 186
190-193, 199-201, 204, 209, 216, 217, 220, 223, 227, GTP-binding proteins, 255, 257-259, 262
233, 253, 254, 256, 257, 265-267, 272-277, 289-291,
294, 298, 299, 302, 307, 308, 320, 322, 328, 332-337, H
339-341, 350, 354-358, 360, 361, 365-370, 376, 377, Hageman factor, 43, 199, 206
413, 428, 433, 434, 473, 534, 535, 537, 538, 541-543, Haptotaxis, 222, 265, 354-361, 594
554, 559, 566, 569, 570, 575, 577-581, 584, 588-591, Heaprin sulfate, 117, 304, 312, 365
593-597 Heart valves, 153, 155, 171, 172, 175, 176, 295, 459, 460,
Fibronexus, 103, 104 472, 475, 532
Fibrosis, 198, 199, 207, 214, 218-221, 223-225, 227, 284, Helper T cells, 393
285, 352, 407-410, 413, 414, 423, 539 Hematopoiesis, 372, 373, 375, 378, 413
Fibrous bodies, 103 Hemostasis, 31, 41, 63, 83, 119, 171, 212, 217, 238, 277,
Filopodia, 106, 254, 268, 290, 330, 332 285, 321, 323, 338, 466, 484, 558
Finite element method, 442, 449, 450, 453, 454, 456 Heparin binding growth factors (HBGFs), 550
First trimester decidua, 102 Heparin proteoglycans, 132
Flk-1/kdr knockout mice, 374 Heterografts, 482, 551
Flt-1, 260, 264, 265, 324, 378, 405 High endothelial venules, 403, 412
Fluid dynamics, 441 Hill and valley, 367
Fluid flow patterns, 441 Histamine, 77, 116, 172, 177, 213, 214, 217, 256, 266
Fluorescence-activated cell sorting (FACS), 375 Histamine receptor, 213, 214
Fluorocarbon, 460, 473, 588 HLA-DR, 173, 176
Foamy macrophages, 129, 181, 182 Homologous serum, 166
Focal adhesion kinase (FAK), 251, 255-268, 335, 354, 358, Human bone marrow, 102, 113, 377
361, 428, 436, 437, 585, 593, 594 Human placenta, 102, 113, 342, 423, 606-608
Focal adhesion plaques, 254, 426, 428, 540 Human saphenous vein endothelial cells (HSVECs),
Fourier transform, 208 75-77, 125, 162, 174, 187
G Human umbilical vein endothelial cells (HUVECs), 65, 69,
70, 118, 122, 139, 140, 157, 159, 256, 260, 261, 273,
G protein, 267, 430, 436, 437, 586, 594 310, 321, 322, 338, 428, 429-432, 437, 555, 556, 558,
G-CSF, 374 578-580, 589
GAX, 245, 249 Hyaluronan, 257, 267, 288, 290, 297, 298, 332, 355,
Gelatin, 70-73, 76, 124, 158, 165, 168, 174, 175, 233, 280, 360, 585
289, 291, 299, 316, 320, 323, 336, 342, 408, 472, 490, Hyaluronic acid, 290, 298, 320, 339, 588, 595
501, 506, 509, 568, 570, 577, 580, 587 Hydrogel, 465, 467, 479, 486, 487, 506, 511, 546, 561-570,
Gelatinase a, 233, 316, 323, 342 588, 611
Gelatinase b, 233 Hydroxyethylmethacrylate, 43
Gelsolins, 255 Hyperlipidemia, 165, 166
Gene therapy, 3, 68, 80, 89, 99, 160, 175, 239, 247, 249, 284, Hyperplasia, 15, 35, 38-40, 45, 51, 52, 57, 58, 61, 62, 65, 72,
299, 422, 423, 605, 606, 608-611 73, 76, 122, 124, 126-128, 130-138, 140-142, 163, 167,
Genetic manipulation, 75, 112, 160, 245, 593 168, 171, 175, 179, 189, 200, 203, 227, 229-243,
Genetically engineered, 65, 86, 112, 113, 162, 246, 248, 292, 245-249, 251, 254, 263, 264, 282, 285, 286, 295, 301,
581, 611 305, 307, 308, 311, 312, 317, 353, 354, 357, 359, 360,
Glucosaminoglycan, 15 363-369, 414, 426, 436, 441, 457, 464, 482, 489, 492,
Glutaraldehyde, 50, 58, 86, 103, 106, 107, 155, 172, 175, 494, 502, 555, 558, 577, 580, 581, 589-593, 595, 597,
288, 289, 482, 507, 515, 523, 545, 551 605-608, 610, 611
Glycophosphoprotein, 106 Hypoxia, 27, 32, 43, 159, 162, 279, 281, 284, 285, 415
Glycoprotein, 29, 63, 106, 110, 111, 172, 205, 217, 225, 239,
289, 295, 306, 314, 315, 332, 336, 355, 367, 373, 405, I
412, 561, 566, 567, 585, 587, 590, 597 IBMX, 159, 160
GM-CSF, 202, 203, 374, 408 ICAM-1, 52, 74, 172-174, 176, 202, 242, 277, 316, 394, 395,
Golgi apparatus, 103, 104, 303 398, 405, 432, 585
Granulation tissue, 10, 12, 34, 198, 199, 206, 226, 273, 282, ICAM-2, 74, 173, 176
283, 304, 305, 310, 313, 318, 343-346, 350, 351, 414, ICAM-3, 74, 173, 176
490, 497, 540, 567, 608, 609 IFN-&, 44, 174, 203, 220, 227, 323, 394, 397
Granulocyte colony-stimulatin factor, 374 IL-10, 223, 224, 389, 397
Index 617
IL-13, 204, 205 Laminin, 63, 71, 72, 74, 122, 158, 233, 254, 256, 266,
IL-1b, 30, 31, 33, 36, 43, 174, 202, 203, 206, 392 272-276, 280, 291, 298, 302, 328, 333-341, 354-358,
IL-1R, 202, 205 361, 405, 413, 428, 433, 434, 529, 541, 542, 575, 585,
IL-2, 277, 394, 397, 398, 404 588, 590, 594-596
IL-2R, 397 Latency-associated peptide (LAP), 29
IL-3, 374, 376, 378 LFA-1, 173, 176, 220, 529
IL-4, 33, 38, 44, 204, 205, 223, 277, 394, 397 Lipid vacuoles, 183, 185
IL-5, 394, 397 Lipopolysacharide, 172
IL-6, 202, 204, 398 Lipoprotein, 133, 139, 141, 158, 165, 166, 168, 169, 202,
IL-8, 173, 201, 202, 204, 221, 224, 227, 398 203, 254, 314, 315, 321, 322, 416, 418, 510, 541,
Image analysis, 23, 84, 112, 133, 428, 533, 534 544, 550
Immunoglobulins (IgG), 172, 174, 199, 204, 207-209, 216, Liposuction fat, 110, 147
222, 318, 395, 399, 534 Liquid crystalline polymers (LCP), 476
Immunohistochemistry, 103, 106, 108, 202-204, 273, 372, Low-density lipoprotein, 77, 139, 169, 254
423, 542 LPS, 172, 173, 316, 397, 398, 416
In situ hybridization, 33, 112, 113, 202-204, 407, 413, 426 LTB-P, 29
Inner capsule, 11, 12, 15, 16, 32, 198, 202, 485, 49-494, LVAD, 265, 387-401
498-500, 509 Lysophosphatidic acid, 255, 266, 267
Insulin-like growth factor, 158, 160, 162, 202, 203,
231-233, 238, 239, 242, 243, 248, 252, 253, 257, 260, M
262, 264, 267, 269, 294, 301, 339, 356, 360 MAC 387, 107, 108
Interferon-&, 100, 173, 176, 202, 203, 277, 412, 526, 590 Mac-1 receptor, 31, 32
Intermediate filaments, 103-105, 111, 139, 336 Macrophage chemotactic protein 1 (MCP-1), 31, 201-203,
Internal elastic lamina, 230, 242, 302, 354, 490 213, 214, 221, 224, 226, 227, 242, 243, 252, 254,
Internodal distances, 5, 19, 21, 32, 72, 379, 485 261, 432
Interstitial collagenase, 239, 281, 316, 323, 336, 342 Macrophage colony stimulating factor, 202, 203, 374
Intima, 8, 15, 17, 36, 45, 51, 52, 58, 61, 63, 66, 68, 73, 76, Macrophage inflammatory protein-1 (MIP-1), 32, 43, 204,
79, 80, 82, 87-89, 97, 99, 128, 129, 135, 137, 139, 169, 213, 214, 217, 221, 222, 226, 274
180, 181, 185, 232-237, 239, 251, 282, 301, 302, 353, Macrovascular endothelial cells, 116, 117, 122, 123, 140,
355, 357, 371, 382, 383, 414, 426, 443, 463, 497, 498, 153, 161
503, 607, 608 Magnetic particle concentrator, 102
Intimal hyperplasia, 35, 38, 39, 45, 51, 52, 58, 61, 73, 76, Mammary adipose tissue, 102, 112
122, 124, 126-128, 130-138, 140-142, 167, 168, 171, MAP kinase, 260-262, 267, 307, 429, 586, 594
175, 179, 189, 200, 229-249, 251, 254, 263, 264, 282, Mast cells, 214, 217, 218, 256, 302, 306, 410, 411, 415, 416
285, 286, 295, 301, 305, 307, 308, 311, 312, 317, 353, Matrical tracks, 328, 329
354, 357, 359, 360, 363-369, 426, 436, 441, 457, 489, Matricellular protein, 334, 337, 340
492, 494, 502, 577, 580, 589-593, 595, 597, 605-608, Matrigel, 272, 273, 291, 328, 335, 339, 367-369, 405, 590
610, 611 Matrix degradation, 29, 36, 280, 313, 364, 607
Ion beam implantation, 486 Matrix metalloproteinase, 231, 233, 236-239, 254, 266,
Irradiation, 218, 245, 477, 484, 487, 581 280, 281, 316, 317, 320, 323, 336, 342, 354, 359, 409,
Ischemia, 123, 145, 227, 230, 253, 275, 276, 283, 284, 286, 415, 607, 610
302, 309, 312, 384 Matrix synthesis, 112, 223, 363, 364, 366-369
K Mechanotransduction, 430, 436, 543
Membrane-type MMP (MT-MMP), 316, 317
Kaplan-meier analysis, 191 Mesenchymal cells, 35, 96, 289, 295, 297, 405, 412, 413,
Kink resistance, 131, 470, 472, 485 418, 419, 421, 423, 491, 492
Kinking, 24, 54, 470 Mesothelial cells, 67, 76, 95, 100, 103, 104, 107, 112, 113,
KP-1, 181 123, 126, 139, 147, 158, 161
Mesothelium, 62, 95, 102, 144, 406
L MHC class II, 43
L-fucose, 106 Microcystic lacunae, 103-106
L-selectin, 220, 223, 226, 227, 273 Microgrooves, 533, 538
Lamellipodia, 330, 332 Micropatterned surfaces, 532
Laminin, 71, 158, 272, 433, 592, 593 Micropinocytotic vesicles, 111
Microtexture, 485, 511, 532-536, 538-543, 583, 587-589,
591, 592, 595
618 Tissue Engineering of Prosthetic Vascular Grafts
Polydioxonone (PDS), 308, 491, 493, 494, 498-500, 509 RGD, 34, 80, 83, 89, 217, 254, 273, 274, 318, 340, 511,
Polyethylene (PE), 31, 41, 42, 141, 201, 207-209, 214, 264, 553-559, 578, 581, 585, 587, 588, 590-592, 594,
364, 457, 467, 464, 471, 473, 475, 478, 479, 487, 488, 595, 597
500, 502, 509, 510, 542, 553-555, 580, 588, 591, 594 RGD peptide(s), 80, 554-556, 558, 559, 581
Polyethylene terephthalate (PET), 31, 32, 41, 208-211, 214, Rho, 251, 257, 258-260, 262, 267-269
364, 472, 473, 478, 509, 553-556, 588, 591 RNA-splicing, 409
Polyglactin 910, 479, 490, 491, 494-496, 501, 502, 511 Rough endoplasmic reticulum, 101, 103, 104
Polyglycolic acid (PGA), 377, 49-494, 500, 509, 510 RT-PCR, 373, 394, 396
Polyninyl chloride, 209
Polyolefins, 477, 484 S
Polypropylene (PP), 112, 113, 133, 310, 470, 473, 477-479, Saphenous vein, 44, 69, 131, 138, 140, 141, 180, 186, 187,
481, 484, 499, 500, 502, 511 377, 482, 608
Polytetrafluoroethylene (PTFE), 4, 6-8, 15, 17, 20-22, 24, Scanning angle reflectometry, 208, 215
25, 31, 32, 38-40, 44, 56, 58, 62, 66-68, 75, 76, 88-90, Scanning electron microscopy (SEM), 20, 23-25, 42, 56,
99, 100, 113, 138, 140-142, 167-169, 176, 177, 180, 83-85, 88, 101, 106-108, 110, 112, 133, 139, 142, 180,
181, 187, 207, 208, 229, 232, 236, 237, 240, 242, 243, 181, 199, 206, 365, 480, 514, 515, 528, 539, 542
248, 285, 355, 359, 360, 369, 370, 377, 380, 385, 386, Scanning force microscopy, 208, 215
393, 423, 441, 458, 470, 473, 476, 478, 482, 484, 497, Scar formation, 3, 574
499, 500, 511, 514-516, 521, 523, 525-527, 553-556, Seeded grafts, 65, 72, 73, 132-138, 141, 499
577, 580, 588, 591, 600, 608, 609, 611 Selectins, 74, 272, 273, 375
Polyurethane (PUR), 31, 32, 39, 43, 44, 80-85, 87, 200, 201, Serine proteases, 27-29, 314, 337
205-208, 215, 377, 379, 387-389, 397, 450, 451, 454, Serotonin, 283
455, 457, 470, 471, 473-475, 477-480, 482, 485-488, Severe combined immunodeficiency (SCID), 208, 209, 335
497, 498, 502, 509, 555-559, 588, 594, 595 Shear stress, 15, 28, 38-40, 44, 45, 52, 54, 58, 63, 65, 69, 72,
Pressure sodding, 144 73, 75, 76, 89, 90, 132, 138, 140, 179, 187, 200, 206,
Procoagulant activity, 30, 42, 176, 265, 389, 393, 398 324, 441, 456, 478, 544
Profilin, 255, 260, 261, 266, 358 Sialomucin, 106
Progenitor cells, 106, 185, 186, 275, 372, 373, 375, 378, 390 Sialyl Lewis-A, 273
Prostacyclin, 38, 40, 76, 99, 110, 132, 139, 140, 141, 165, Sialyl Lewis-X, 272, 273, 275, 276
168, 216, 231, 234, 428, 430, 431, 436-438, 497, 498, Signal-transducer receptors, 138
502, 599 Silanization, 484, 486
Prostaglandin metabolite, 494 Silicone, 43, 199, 215, 471, 473, 477, 479, 533, 536, 537,
Protease(s), 27-30, 87, 219, 225, 232, 233, 314, 316, 321, 539, 540, 542, 543
323, 336, 337, 470 Sintering, 507
Protein A-gold, 107, 109, 113 Skirted cells, 106, 111
Protein C, 77, 80, 81, 89, 139, 205, 208, 215, 216, 223, 225, Smooth muscle cells, 3, 7-9, 11, 12, 15, 17, 19, 29, 33-40,
256, 258-260, 268, 336, 435, 554, 579, 581, 585, 44, 61, 62, 68, 74, 77, 86, 87, 179, 180, 185-187, 199,
586, 600 202-204, 364, 369, 370, 441-443, 458, 541, 542
Protein kinase A, 429, 434, 436, 437 Sodding, 67, 88, 96-100, 363
Protein kinase C, 267, 316, 318, 323, 431, 437, 438, Sodium dodecyl sulfate, 208, 212, 215
586, 594 SPARC, 202
Protein phosphatase, 260 Stem cells, 186
Proximal anastomoses, 15 Stereotropism, 532
PTCA, 171, 229, 233, 237 Stress, 15, 19, 24, 27, 28, 38-40, 44-47, 51, 52, 54, 57, 58, 63,
R 65, 69, 72, 73, 75, 76, 89, 90, 104, 116, 118, 122, 123,
125, 130, 132, 138, 140, 153, 154, 174, 175, 177, 179,
Rabbit ear chamber, 327, 417-419, 422 187, 189, 190, 192, 194, 200, 206, 215, 220, 234, 235,
ras, 257, 266, 268, 414, 594 240, 241, 246, 248, 254-260, 262, 266, 268, 292, 304,
Rats, 18, 38, 40, 77, 102, 215, 224, 227, 232-234, 240, 252, 324, 329, 335, 337, 344, 348, 350, 351, 353, 359, 372,
322, 352, 404, 407, 408, 410, 413, 414, 416, 445, 377, 390, 425, 426, 428-433, 436-438, 441-454,
497-499, 501, 502, 511, 515, 516, 533, 588, 589, 611 456-458, 467, 470, 471, 474, 475, 478, 479, 494,
Reactive oxygen intermediates, 33, 219 497-499, 501, 508, 528, 531, 539, 540, 544, 555, 559,
Renin-angiotensin system, 232 581, 588, 594
Reporter proteins, 606 Stress cracking, 467, 470, 475, 479
Resorbable grafts, 282, 489, 490, 494, 498, 499, 501 Stroma cell, 372
Retinoic acid, 404, 405, 411, 412 Stromelysin-1, 316
Retrovirus, 606, 609, 611 Subcutaneous adipose tissue, 62, 121, 122, 124
620 Tissue Engineering of Prosthetic Vascular Grafts
Subcutaneous fat, 95, 99, 102, 103, 105, 107-111, 113, 115, Titanium, 388-390, 397, 460, 529, 536, 538, 542-544, 595
126, 139, 144, 154, 158 TNF-#, 30-32, 172, 173, 202-204, 213, 214, 220, 224, 313,
Subdermal vein, 163, 189 316-320, 389, 392, 396, 398, 417, 421, 422, 590
Supercritical fluid, 507, 511 Topographical morphology, 532
Superoxide, 172, 173, 219, 246, 438, 461 Toxicity, 89, 164, 166, 168, 225, 308, 513, 578, 611
Surface modification, 41, 67, 89, 111, 122, 143, 154, 465, tPA, 27-29, 52, 65, 117-119, 124, 160, 231-233, 238, 280,
466, 469, 470, 473, 482, 483, 484, 486, 487, 488, 508, 281, 314-318, 336, 428, 432, 597
510, 531, 545, 546, 549, 553, 555, 583, 591, 592, 593 Traction, 34, 48, 49, 57, 68, 202, 212, 234, 254, 288-291,
Surface modifying additives, 482 297-299, 322, 326, 328-333, 335, 336, 337-339, 342,
Surface morphology, 106, 183, 513-515 351, 352, 410, 413, 435, 476, 477, 507, 540, 568, 572-
Surface roughness, 513, 528, 529, 595 576, 591
Surface thrombogenicity, 189, 191, 215 Transcription factor, 52, 227, 244, 246, 307, 358, 398, 401,
Syndecans, 409, 585, 593 407, 409, 430, 431, 436, 438
Transdifferentiation, 35, 403-411, 413, 416, 417, 425
T Transduction, 52, 89, 138, 223, 224, 240, 245, 248, 251, 254,
Talin, 255, 256, 258, 260, 261, 268, 334, 354, 358, 427, 585, 256-258, 260, 262, 263, 265, 266, 268, 298, 307, 310,
593, 604 311, 315, 334, 340, 341, 351, 352, 359-361, 373, 378,
Tek, 405 401, 408, 411, 427, 430, 436, 437, 540, 543, 585, 586,
Tenascin, 40, 233, 239, 272, 275, 276, 334, 340, 355, 360, 593-595, 597, 606, 607
405, 409, 412-414, 589, 590, 592, 593, 596, 597 Transfection, 65, 66, 68, 162, 245, 246, 249, 284, 304, 310,
Tensile modulus, 472-475 411, 431
Tensin, 140, 158, 231-233, 237-239, 242, 248, 253, 255, 257, Transformtin growth factor-!, 28-30, 39, 41, 42, 44, 52, 89,
265, 266, 301, 334, 354, 408, 414, 585, 586, 610 168, 202, 203, 219, 225, 239, 242, 252, 263-266, 273-
TGF-!, 27-31, 35-37, 41, 42, 52, 158, 162, 201-203, 219, 275, 277, 280, 285, 289, 292, 294, 295, 298, 299, 302,
220, 224, 227, 233, 238, 242, 243, 252-254, 256, 263, 324, 335, 338, 352, 353, 359, 360, 369, 370, 374, 375,
276, 277, 280, 282, 285, 289-295, 297, 298, 299, 318, 413, 414, 417, 423, 478, 596
335, 337, 339, 341, 343-353, 364, 365, 367, 368, 370, Transmural ingrowth channels, 22-24
374, 375, 405, 407, 408, 410, 411, 415, 417, 421, 422, Transmural microvessel, 19, 379, 385
438, 496, 590, 596 Triglyceride, 165, 166, 168
Th2 lymphocytes, 33 Trypsin, 62, 96, 117, 133, 159, 161, 162, 164, 304, 336,
Thermoplastic, 473, 476, 479, 511 500, 611
Thrombin, 26, 29, 42, 63, 77, 81, 115-119, 139, 155, 161, Tumor necrosis factor alpha, 172
172, 201, 202, 216, 231, 232, 260, 263, 266, 268, 304, Type 1 plasminogen activator inhibitor, 322
308, 311, 320, 387-391, 398, 408, 434, 465, 467, 479, Type I collagen, 203, 266, 272-274, 276, 288-291, 293, 297,
481, 482, 546-551, 563, 568 298, 328-330, 332, 333, 335, 336, 338, 339, 350, 357,
Thrombin generation, 388-390, 398, 465, 546-548, 550 358, 361, 363-370, 433, 434, 508, 550, 569, 571, 572,
Thrombocytes, 8, 28, 116 575, 587, 590, 608
Thromboemboli, 79, 80, 88, 172, 387, 388, 400, 401 Tyrosine kinase, 138, 142, 162, 244, 253, 256, 257, 259, 267,
Thrombomodulin, 116, 117, 139, 162, 393, 394, 599 268, 290, 295, 306, 307, 324, 334, 335, 358, 374, 378,
Thromboplastin, 26, 388 405, 428, 435, 438, 585, 586, 593
Thrombospondin (TSP1), 27-30, 42, 74, 202, 233, 272- U
277, 320, 334, 339, 340, 355, 356, 359, 360, 415, 589,
590, 592, 596, 597 Ulex europaeus Type I Lectin, 106, 107
Thrombotic occlusion, 284, 393, 422, 482, 545 Umbilical vein, 50, 52, 58, 65, 66, 69, 94, 118-120, 122, 125,
Thrombus, 5, 7-10, 16, 27, 30, 40, 62, 63, 68, 80, 82, 88, 139, 140, 157, 160, 168, 175, 266, 273, 291, 310, 316,
128, 129, 133, 135-137, 139, 154, 172, 187, 215, 229, 321-323, 328, 335, 338, 342, 428, 431, 432, 437, 545,
231, 236, 372, 423, 490, 551, 553, 555, 558, 559, 569, 555, 578, 579
577, 605, 606 uPA, 29, 31, 118, 231-233, 238, 280, 313-318, 320, 321,
Thy-1, 110, 374 336, 337
Tight junctions, 103, 104, 140 uPA receptor, 313, 315-318, 336
Tissue factor, 28, 30, 42, 162, 253, 389, 392-394, 397, 398, Urokinase, 27, 29, 42, 65, 117-119, 231-233, 238, 254, 266,
409, 415, 580 280, 313-315, 317, 321-324, 336, 342, 354
Tissue inhibitors of MMPs (TIMP), 231, 233, 254, 316, Urokinase plasminogen activator, 29, 354
317, 336, 607, 608, 610
Tissue plasminogen activator, 27, 29, 52, 65, 68, 77, 154,
161, 266, 321, 610, 611
Index 621
V W
Vascular cell adhesion molecule-1 (VCAM-1), 74, Weibel-Palade bodies, 95, 96, 103-106, 111, 140, 158, 172,
172-174, 202, 273-276, 316, 393-395, 398, 399, 401 176, 426
Vasculogenesis, 268, 279, 281, 302, 324, 335, 341, 373, 379, Wettability, 68, 506, 510, 513, 515, 526, 527, 540
384, 385, 421-423 Wound healing, 29-31, 33, 34, 131, 138, 197, 198, 200-202,
VEGF, 27, 29, 32, 34, 35, 160, 202, 203, 252, 253, 255-257, 206, 212, 220, 221, 223, 225, 253, 256, 271, 272, 274,
260-264, 268, 279-286, 294, 295, 297, 302, 313, 316, 275, 277, 288-290, 294, 298, 302, 305, 311, 314, 318,
318, 320, 322, 337, 374-377, 404, 405 320, 321, 324, 352, 379, 407, 413, 416, 417, 422, 425,
Vein wrap, 380 441, 525, 540, 567, 570, 572-574, 596
Ventricular assist devices, 79, 88, 89, 372, 377, 380, 385,
387, 400, 401, 403 Y
Vimentin, 104, 105, 107, 108, 139 Yellow baboon, 7, 9, 11, 17, 18, 35, 36
Vinculin, 255, 256, 258, 260, 261, 267, 268, 323, 334, 354, YIGSR, 558, 559, 588, 591, 594, 595
358, 427, 534, 535, 537, 540, 542, 585, 586, 588,
593, 594
Vitronectin, 27, 74, 85, 165, 168, 254, 265, 272-275, 277,
314, 315, 318, 320, 322, 324, 335, 337, 355-357, 360,
428, 434, 534, 535, 542, 554, 584, 590, 594, 596, 597
von Willebrand factor, 28, 95, 100, 110, 133-135, 139, 140,
158, 273, 281, 292, 299, 335, 382, 390, 393, 394, 554,
590, 596, 597