Extracellular Matrix For Tissue Engineering and Biomaterials PDF
Extracellular Matrix For Tissue Engineering and Biomaterials PDF
Anna C. Berardi Editor
Extracellular
Matrix for Tissue
Engineering and
Biomaterials
Stem Cell Biology and Regenerative Medicine
Series editor
Kursad Turksen, Ph.D.
e-mail: [email protected]
Our understanding of stem cells has grown rapidly over the last decade. While the
apparently tremendous therapeutic potential of stem cells has not yet been realized,
their routine use in regeneration and restoration of tissue and organ function is greatly
anticipated. To this end, many investigators continue to push the boundaries in areas
such as the reprogramming, the stem cell niche, nanotechnology, biomimetics and
3D bioprinting, to name just a few. The objective of the volumes in the Stem Cell
Biology and Regenerative Medicine series is to capture and consolidate these
developments in a timely way. Each volume is thought-provoking in identifying
problems, offering solutions, and providing ideas to excite further innovation in the
stem cell and regenerative medicine fields.
Extracellular Matrix
for Tissue Engineering
and Biomaterials
Editor
Anna C. Berardi
Department of Hematology, Transfusional
Medicine and Biotechnology
Hospital Santo Spirito
Pescara
Italy
This Humana Press imprint is published by the registered company Springer International Publishing
AG part of Springer Nature
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface
v
Contents
vii
viii Contents
Nicolò Nicoli Aldini Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic
Institute, Bologna, Italy
Stephen F. Badylak Surgery and Bioengineering, University of Pittsburgh, Pittsburgh,
PA, USA
Caterina Bason Department of Medicine, University of Verona, Verona, Italy
Anna C. Berardi Research Laboratory “Stem Cells” U.O.C. Immunohematology,
Transfusion Center and Laboratory of Hematology, Hospital Santo Spirito, Pescara,
Italy
Amelia Cataldi University G. d’Annunzio, Chieti-Pescara, Italy
Daniela Cigognini Network of Excellence for Functional Biomaterials, National
University of Ireland, Dangan, Galway, Ireland
Luis M. Delgado Network of Excellence for Functional Biomaterials, National
University of Ireland, Dangan, Galway, Ireland
Giovanna Della Porta Department of Medicine, Surgery and Dentistry, University
of Salerno, Baronissi, SA, Italy
Viviana di Giacomo University G. d’Annunzio, Chieti-Pescara, Italy
Anna Domogatskaya Division of Transplantation Surgery, Department of Clinical
Science, Intervention and Technology, Karolinska Institute, Huddinge, Sweden
Milena Fini Laboratory of Biocompatibility, Innovative Technologies and Advanced
Therapies, Rizzoli RIT Department, Bologna, Italy
Gianluca Fontana Network of Excellence for Functional Biomaterials, National
University of Ireland, Dangan, Galway, Ireland
Francesco Oliva Department of Orthopaedic and Traumatology, School of
Medicine, University of Rome “Tor Vergata”, Rome, Italy
ix
x Contributors
DC Dendritic cell
EAE Experimental autoimmune encephalomyelitis
ECM Extracellular matrix
GAG Glycosaminoglycan
HA Hyaluronan
LPS Lypopolysaccharide
MMP Matrix metalloproteinase
NO Nitric oxide
NOS Nitric oxide synthase
PPAR Peroxisome proliferator-activated receptor
TLR Toll-like receptor
TSP Thrombospondin
TIMP Tissue inhibitor of metalloproteinases
xi
Part I
Extracellular Matrix
Chapter 1
The Extracellular Matrix,
Growth Factors and Morphogens
in Biomaterial Design and Tissue
Engineering
Abstract Cells, morphogens, growth factors, and custom scaffolds are the critical
ingredients for successful tissue regeneration in which morphogens and growth
factors function sequentially. Extensive studies, in vitro and in vivo, have been
made to explore the mechanisms and the roles played by these molecules. As a
consequence, precise, localized control over the signaling of these factors and
appropriate strategy selection, depending on the tissue or organ to be repaired or
regenerated, is known to permit specific management of regenerative processes.
The first part of the chapter examines natural ECMs which are a set of molecules
secreted by cells that provide structural and biochemical support to the surrounding
cells. ECMs also perform many other functions, such as actively regulating cell
function through the control of biochemical gradients, cell density, spatial organi-
zation, and ligand attachment, thus influencing various types of cell processes.
Subsequently, growth factors and morphogens are examined in greater depth to
clarify to what degree progress has been made into improving methodologies and
functionality and, perhaps, to hint at what remains to be done for the future of tissue
engineering.
C. Bason
Department of Medicine, University of Verona, 37134 Verona, Italy
e-mail: [email protected]
M. Gallorini
Department of Pharmacy, University G. d’Annunzio Chieti-Pescara, Pescara, Italy
e-mail: [email protected]
A. C. Berardi (&)
Research Laboratory “Stem Cells” U.O.C. Immunohematology,
Transfusion Center and Laboratory of Hematology,
Hospital Santo Spirito, Pescara, Italy
e-mail: [email protected]; [email protected]
Introduction
Tissue and organ regeneration in the adult are complex processes that often rep-
resent a recapitulation of embryonic development and patterning processes and
which seek to repair tissue and/or to maintain integrity and function. Increasing
1 The Extracellular Matrix, Growth Factors … 5
Proteoglycans (PG) are composed of a protein axis to which one or more gly-
cosaminoglycan chains (GAGs) are covalently linked. Usually the same protein
binds to a single type of GAG. However, more than one type of GAG exists, such
as those found in cartilage-based proteoglycans, which contain similar quantities of
chondroitin sulfate and keratan sulfate [8]. GAG variety depends on the location of
the ECM, age, and gender of the individual. GAGs promote water retention and
contribute to gel-like properties of the ECM. GAGs also bind cytokines and growth
factors and retain them in the ECM [9]. Around 36 ECM PGs have been identified
in mammals with multiple, diverse functions, which also vary in different types of
tissue. There are three main families of PG: (1) small leucine-rich proteoglycans
(SLRPs), (2) modular proteoglycans, and (3) cell surface proteoglycans [10].
SLRPs, which are ubiquitously expressed in most ECMs, are the largest class of
PGs. They can function as structural components and are involved in multiple
signaling pathways. SLRPs bind with various collagens, tyrosine kinase receptors
(RTK), and innate immune receptors, and, in so doing, they participate in several
biological functions [11], being able to regulate fundamental processes like
migration, proliferation, innate immunity, apoptosis, autophagy, and angiogenesis
[11].
Furthermore, they bind to and activate epidermal growth factor receptors
(EGFR), insulin-like growth factor 1 receptor (IGF1R), and low-density lipoprotein
receptor-related protein 1 (LRP1).
Hyalectans, which are modular PGs, are key structural components of central
nervous systems, cartilage, and blood vessels and can modulate cell adhesion,
migration, and proliferation, by binding hyaluronan to form complexes of high
viscosity. The hyalectan family, found in interstitial membrane matrices, contains
aggrecan, versican, neurocan, and brevican [10]. The essential role of aggrecan, the
principal load-bearing proteoglycan in cartilage, has been confirmed by several
studies. Versican, a large chondroitin sulfate proteoglycan, forms long filaments in
the ECM and has been shown to play an important role in modulating inflammatory
responses to tissue injury. Basement membrane modular PGs (perlecan, agrin, and
collagen type XVIII) have a dual function as pro- and anti-angiogenic factors [12],
mediate ligand, cell–matrix and cell–cell interactions, and interaction with integrins
and RTK.
Heparan sulfate (HS) binds a large number of extracellular proteins. The func-
tions of heparan sulfate-binding proteins range from extracellular matrix compo-
nents to enzymes and coagulation factors, as well as most growth factors, cytokines,
chemokines, and morphogens. For example, heparin sulfate proteoglycans (HSPGs)
bind FGFs and sequester these molecules for storage. HS regulates a wide range of
biological activities, including developmental processes, coagulation, and angio-
genesis [13].
1 The Extracellular Matrix, Growth Factors … 7
Growth Factors
Growth factors (GFs) (soluble signals) are the major regulators of cell behavior;
they promote cell proliferation, migration, and differentiation through specific GF
receptor binding which stimulates cellular signal transduction pathways. GFs are
involved in several physiological and pathological processes, such as tissue repair
and maintenance [32]. GFs are secreted from the cells directly or are sequestered by
1 The Extracellular Matrix, Growth Factors … 9
ECM for presentation to cell surface receptors. Some studies demonstrate that GF
can be released from ECM by the degradation of ECM proteins, of GAGs, or of
PGs. The ability of growth factors to interact with the extracellular matrix is a
dynamic, tissue-specific property [33–35]. GFs, ECM components, and cell surface
receptors form complexes that may lead to additive or synergistic cell signaling
events. The ECM can regulate GF-mediated cell function [36] through many
insoluble (ECM-bound) and soluble (un-bound) cell secretions, through cell surface
proteins and through proteoglycans. Specifically, the ECM regulates GF activity by
sequestering soluble GFs and by cell-demanded release via enzymatic degradation
of the ECM, eliciting a variety of effects on GF signaling that are dependent on the
context and presentation of the GF to cells [36–38]. Both cytokines and growth
factors are present within the ECM in very small quantities, but act as potent
modulators of cell behavior. The list of GFs found within the ECM is substantial
and includes vascular endothelial cell growth factor (VEGF), transforming growth
factor beta (TGF-b), the fibroblast growth factor (FGF) family, insulin-like growth
factor (IGF), epithelial cell growth factor (EGF), keratinocyte growth factor (KGF),
hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), bone
morphogenetic proteins (BMPs), and many others. Multiple isoforms of these
molecules are present, each with its own specific biological activity. VEGF, which
has been the subject of a number of studies, is the major inducer of endothelial cell
homeostasis and of angiogenesis and is an example of context-dependent GF sig-
naling. Soluble and insoluble ECM components regulate VEGF activity in various
ways. Several distinct VEGF genes produce multiple VEGF isoforms which exhibit
variable binding domains for heparan sulfate (HS). Furthermore, every VEGF
isoform plays a distinct role in vascular patterning and arterial development in the
ECM [39, 40]. VEGF induces a1b1 and a2b1 integrin expression in microvascular
endothelial cells, as well as endothelial cell migration and proliferation. Although
the major role of VEGF is related to vascular tissue, several studies have shown
VEGF to also be implicated in bone cell recruitment, activity, and survival [41].
VEGF binding to the cell surface or to ECM and various MMPs and plasminogen
activators can generate diffusible, non-heparin-binding fragments. ECM component
fibronectin can be modified in the presence of heparin or HSPGs which expose a
binding site for VEGF [42–44]. The binding of VEGF to the ECM enhances
endothelial cell proliferation, probably through an increase in mitogen-activated
protein kinase (MAPK) activity [5, 45]. VEGFR-2 and VEGFR-1 tyrosine kinase
receptors mediate VEGF activity [40]. VEGFR-1 has minimal kinase activity, but a
high affinity for VEGF suggesting that it serves as a VEGF sink, preventing VEGF
binding to the active VEGFR-2 receptor [40]. While VEGFR-2 mediates most of
the endothelial growth and survival signals, VEGFR-1-mediated signaling plays an
important role in certain pathological conditions such as inflammation and cancer.
VEGF–VEGFR-2 binding on the cell surface leads to AKT activation and calcium
release, whereas internalization of the receptor complex triggers ERK signaling in
early endosomal compartments [46, 47]. Internalization and trafficking of
VEGFR-2 can be influenced by many factors, including neuropilin-1 (NRP-1),
syndecans, and hypothetically by b1-integrin [46, 47]. The activation of VEGFR-2
10 C. Bason et al.
mineralization. Two mechanisms of IGF release have been identified: the ECM
binding and the IGFBP proteolysis [52]. These two actions lower the affinity of
IGFBPs for IGFs increasing the local concentration of bioactive IGF. IGFBP-2, -3,
-5, and -6 interact with GAGs [53–55]. IGFBP-2 interacts with heparin-binding
surface-pH-dependent and with GAGs; these process may be relevant where
extracellular pH is low, like in site of wound healing. Interestingly, IGFBP-2 and -6
bind a broad range of GAGs whereas IGFBP-3 and -5 preferentially bind to hep-
arin, heparan sulfate, dermatan sulfate, and minimally to chondroitin sulfates and
hyaluronic acid [56]. These differences in GAG binding specificity confer
tissue-specific actions upon the IGFBPs. GAG binding can also alter IGFBP
interactions with other ligands, thereby modulating the IGF-independent actions of
IGFBPs. For example, activation of plasminogen to plasmin is promoted by the
interaction of IGFBP-5 and plasminogen activator inhibitor-1 (PAI-1), leading to
ECM degradation and remodeling independently of IGF-I. This process is impor-
tant in mammary gland involution. Further interaction with ECM proteins via
IGFBP C-domains has been reported [57]. For example, IGFBP-3 binds fibrin/
fibrinogen, fibronectin, vitronectin [58], and plasminogen, which then can influence
both its IGF-dependent and IGF-independent actions. Also, fibronectin binds
IGFBP-5 and inhibits its ability to promote IGF-dependent cell migration [59].
IGFBPs are well-known MMP substrates. MMPs cleave IGFBPs predominantly
in the linker domain and are not dependent on IGF binding [52, 60]. Proteolysis
provides a mechanism by which the concentration of freely bioavailable IGFs is
increased, leading to subsequent activations of IGF1R. Specifically, the processing
of IGFBP-3 and -5 by MMP-1,-2, -3,-7, and -19 and IGFBP-1 by MMP-3, -9, and -
11 releases active forms of IGFs [50]. The dual effect of IGFs on MMP expression
has also been observed: IGF-1 can upregulate MMP-2 synthesis via PI3-kinase/Akt/
mTOR signaling while concomitantly transmitting a negative regulatory signal via
the Raf/ERK pathway [50].
Remodeling enzymes, which are capable of modifying and degrading ECM pro-
teins, are central to the interactions between host immune systems and ECMs.
Migration of immune cells into areas of disease-induced tissue damage is facilitated
by localized ECM breakdown. Additionally, the innate immune system initiates
responses to infection; thanks to products of ECM degradation which can serve as
early signals. In response to injury and infection, ECM remodeling enzymes con-
tribute to modulation of inflammation and tissue repair processes. In this case, most
of these remodeling enzymes are produced by immune cells, and in particular
myeloid lineage immune cells [61].
There are a large number of molecules which play a role in protease activity and
are also involved in proteolytic processes in the ECM, which can be divided into
12 C. Bason et al.
three main groups: (1) serine proteases; (2) matrix metalloproteinases (MMPs),
which are a large family of highly conserved, Zn-dependent endopeptidases;
(3) bone morphogenetic protein 1 (BMP1), which is a member of the tolloid family
of metalloproteinases.
These proteinases have been linked to cellular differentiation and pattern for-
mation through a role in activating latent growth factors in the TGF-b superfamily.
Further, a disintegrin and metalloprotease proteins (ADAMs) are a family of
transmembrane glycoproteins with diverse roles in cell–cell adhesion and proteol-
ysis. MMPs/ADAMs are a family of zinc-dependent, ECM-degrading endopepti-
dases that share common functional domains, activation mechanisms, and,
collectively, have the capability to degrade all types of ECM protein [61]. In
addition to playing a central role in ECM macromolecule turnover, within tissue
microenvironments MMPs/ADAMs can modify extracellular, soluble, or
membrane-bound proteins and induce their rapid release or inactivation. MMPs,
which were initially recognized for collagen remodeling, have now been shown to
play a significant role in the control of immune responses. These enzymes can
proteolytically activate or degrade a variety of non-matrix substrates, including
signaling molecules such as growth factors, chemokines, and cytokines.
Moreover, the main proteolytic enzyme families, MMPs and ADAMs, are
considered to be the predominant proteases in ECM pathophysiological regulation.
Each individual MMP influences the properties of ECMs differently, generating
distinct chemical, biomechanical, and morphological features in the ECM [61]. The
effects produced by a specific MMP through selective ECM degradation uniquely
influences cell behavior, including migration, morphology, gene expression profile,
and activation of intracellular cascades. In addition to degrading ECM proteins,
MMPs modulate a variety of biological factors and non-ECM molecules, directly
influencing tissue homeostasis. Collagenolysis and elastolysis by MMPs occur
during development, wound healing, and in major inflammatory diseases. MMP-2,
MMP-7, MMP-9, MMP-12, and MT1-MMP have been suggested to be elastolytic.
Controlled proteolytic cleavage of ECM protein-release growth factors, such as
FGFs, contributes to localized cell proliferation and differentiation.
Studies have shown that certain cytokines and growth factors, including insulin
growth factors (IGFs), epidermal growth factors (EGFs), vascular endothelial
growth factor (VEGF), transforming growth factor beta (TGF-b), fibroblast growth
factors (FGF), tumor necrosis factor-a (TNF-a), and interleukin-1b (IL-1b), can
stimulate expression of MMPs and ADAMs [61].
components, while the intracellular domain interacts with the cytoskeletal proteins
like actin, affecting polarization and motility. For example, some integrins appear to
only be linked to one type of ECM protein (fibronectin or laminin), whereas others
may interact with a number of types (such as collagen, fibronectin, and laminin). It
is also possible that a single integrin may bind to several distinct ligands and that an
individual ligand may be recognized by more than one type of integrin.
A significant number of cytoskeletal and signaling proteins have been reported to
bind to b-cytoplasmic tails, and some have also been found to interact with specific
a tails. Moreover, alternative mRNA splicing, in extracellular, as well as in intra-
cellular, regions, produces a large number of variant forms of a and b subunits.
Differently spliced isoforms of both a and b subunits can differ in their effect on
signal transduction pathways, or in their specificity and affinity for ligands and
interaction with cytoskeletons. The preferential selection of various splice variants
in specific types of cells at precise differentiation stages leads to significant func-
tional differences. For example, a7b1 integrin can be detected in proliferating and
adult myofibers [67], and it is reported that a7A has a minor function in mature
muscle fibers, but a relevant role in regenerating muscle fibers [68, 69].
Furthermore, integrin-mediated adhesion modulates signaling cascades in the
control of cell survival, proliferation, differentiation, and motility. Migration of
adhesive mesenchymal cells depends on ECM proteolysis and the interaction of
integrins with ECM proteins. In addition, integrins are also involved in leukocyte
interactions with the endothelial basement membrane and the transmigration of
these cells to sites of inflammation. Some integrins even bind to receptors present
on other cell types, such as intercellular adhesion molecules (ICAMs) or vascular
cell adhesion molecules (VCAMs) which are expressed on leukocytes and
endothelial cells. There is evidence suggesting that lymphocyte motility and
retention in certain compartments is influenced by integrin–ECM interactions, in
particular, in inflamed tissues with altered ECM composition and integrin expres-
sion, for example in the context of influenza infection. Another important role of
integrins is in the activation of ECM-bound cytokines and growth factors. For
example, the a4b6 integrin activates latent TGF-b regulating the spatial bioavail-
ability of the growth factor. Integrin activation introduces another level of regula-
tion of ECM-bound molecules. The effect of a4b6 activation of TGF-b is likely to
be context dependent. An early study found that mice lacking a4b6 develop airway
hyper-responsiveness due to infiltration of inflammatory cells into the lungs and
skin. Activation of TGF-b by a4b6 may also contribute to fibrotic lung disease
following influenza infection due to increased collagen deposition. Integrins con-
tribute to development and tissue morphogenesis and also play a key role in tissue
homeostasis and tissue repair. The presence of cytokines and growth factors in the
ECM provides a means for host cells to rapidly respond to infection or injury as
these molecules are released and/or activated. In this manner, these ECM-bound
molecules may represent some of the earliest signals to the host immune system to
further rapid responses.
1 The Extracellular Matrix, Growth Factors … 15
Morphogenesis
activity [87]. However, integrin and cadherin interaction may involve not only
shared cytoskeletal linkages, cell–cell, or cell–ECM engagement, but can also
potentially interact with adaptor proteins [i.e., tetraspanin or growth factor receptors
like insulin growth factor I receptor (IGF1R)] which facilitate lateral association of
integrins and cadherin. RhoGTPases are central to cell signaling pathways both
upstream and downstream of cadherin and integrin adhesions [88, 89], making
them prime candidates for mediating integration of adhesion-dependent
signals [90].
Bone morphogenesis is induced by bone morphogenetic proteins (BMPs), which
play a role in pattern formation, cell differentiation, maintenance, and regeneration
of tissues. BMPs are pleiotropic and act on chemotaxis and mitosis and can dif-
ferentiate progenitor cells and stem cells, which may also be programmed by
BMPs. The name is slightly misleading as their range of action also extends beyond
simple bone formation in that they play a role in the development of the teeth, heart,
kidneys, eyes, skin, and brain. The ECM, the native scaffold, can be used to deliver
morphogens such as BMPs for tissue engineering [91].
Many morphogenetic processes begin with a flat or curved sheet of cells that
eventually gives rise to complex topologies such as folds. Folds can be generated by
a monolayer of cells, by stratified cell sheets, or by multiple interacting tissues.
Consequently, cell–cell adhesions must play different roles depending on the cel-
lular behaviors required. Although the precise mechanism remains unclear,
E-cadherin, for example, is required for fold formation during embryogenesis [92].
It may be involved in transmitting mechanical cell–cell cues and could play a role in
establishing appropriate cell polarity.
Tubulogenesis is generally initiated by tissue folding, usually generating tubes of
larger diameters, and is completed by the formation of new adhesions to seal the
tube [93]. Proper distribution and transmission of tension require precisely con-
trolled adhesion between cells, in blood vessel tubulogenesis; for example, cell–cell
adhesions participate in establishing polarity and may also act as mechano-sensors
linking blood flow to vascular remodeling. Vascular endothelial (VE)-cadherin and
apical–basal polarity are required for endothelial cells to form lumena [94, 95].
Polarity is established through interactions between VE-cadherin and cell polarity
complex proteins Par3 and Pals1 [95]. Without proper localization of cell–cell
junctions and establishment of cell polarity, lumen formation and, therefore, blood
flow are prevented and blood flow is, itself, a major determinant of vascular
morphogenesis, including sprouting and lumen elongation [96, 97]. Pressure and
shear stress cause changes in endothelial cell behavior. These mechanical signals
18 C. Bason et al.
In the complexity of the ECM, with the vast range of physical, biological, and
chemical interactions, signaling pathways and networks all producing a host of
specific responses to given stimuli, mimicking or recreating these processes through
tissue engineering and regeneration techniques requires a number of different
components. Vital among these are growth factors which, as described above, play
fundamental roles in a variety of tissue and organ formation mechanisms and
processes. The properties of the ECM itself, as a reservoir, have a pivotal role in the
physiological delivery of GFs, crucial to the regenerative process [32]. While the
use of GFs has had some clinical success, their potential as therapeutic agents has
generally been hindered by certain limitations. Many growth factors are intrinsically
unstable, with very brief functional activity [110], while the absence of suitable
delivery methods excludes others. For instance, some GFs like BMP-2 and VEGF
have minimal interaction in their native form with the surrounding ECM, which in
clinical situations requiring repeated dosage due to rapid outward diffusion and fast
proteolysis makes them unsuitable [111, 112]. Therefore, some form of GF engi-
neering may be required in order for regenerative applications to overcome these
limitations [32].
Similarly, given that morphogens are another essential regulatory mechanism in
the creation of improved human tissue models, researchers over the past two
1 The Extracellular Matrix, Growth Factors … 19
decades have used them to develop a variety of hydrogel compositions which can
envelop 3D cell cultures, or modified, synthetic biomatrices, in order to better
deliver both GFs and morphogens into the target tissue [113].
The major issue when a signaling delivery system has to be improved is how to best
mimic naturally occurring, inductive signal sequestering which serves as a template
for the design of synthetic molecules that can sequester GFs and morphogens [36].
“Biomimicry,” which is the underlying mechanism of GF sequestration, was first
used in studying the interaction between a2-M and TGF-b1, a2-M and PDGF-BB,
and TGF-1b and TGF-1b receptor III [114–116] and can be used to identify GF
sequestering moieties. Researchers have demonstrated that moieties engineered to
mimic known proteins, or proteoglycans such as fibronectin and fibrinogen,
exhibited GF or heparin-like sequestering [117]. In addition to soluble approaches,
GFs can be sequestered at 2D and 3D interfaces in the ECM environment. Surfaces
presenting proteoglycans and glycoproteins in a synthetic functionalized monolayer
can sequester GFs and modulate GF-related cell response. As an example, it has
been demonstrated that GFs can be sequestered by a self-assembled monolayer
modified with a HEPpep peptide derived from the heparin-binding domain of
FGF-2 and an Arg-Gly-Glu (RGD), derived from a sequence of fibronectin which
allows integrin-mediated cell adhesion [118].
A 3D interface design, however, requires the optimum choice of an appropriate
biomaterial which must retain GFs. Controlled tailoring of physical properties, like
density, porosity, viscosity, and charge, is essential for the implementation of GF
sequestration inside the scaffold [110]. Additionally, GF activity must be preserved
when incorporated within the biomaterial, which can easily be done by designing
formulations which mimic the ECM, such as hyaluronan, chitosan, collagen/gelatin,
and fibrin-based scaffolds. Moreover, natural biomaterials are cell-growth-friendly
and they avoid limitations such as pathogen transmission or ethical considerations
which can arise using animal or human sources in a clinical situation. Furthermore,
in a 3D matrix scenario, the proximity of the cells to the sequestering event and the
source of the sequestered GF (cell secreted or supplemented into the growth
medium) can influence the final cell response [36]. Hydrogels are excellent sub-
stitutes for native ECM because they can mimic its physical structure and bio-
chemical functions. They are increasingly used for GF sequestering and to control
release for a variety of purposes, such as to increase angiogenesis, and nerve and
bone regeneration [119–121]. However, most organoids are currently formed by
envelopment within Matrigel hydrogels, which are a poorly defined, heterogeneous
mixture of soluble basement membrane proteins from mouse sarcomas. Some
researchers have shown that chemically well-defined hydrogels could be used to
20 C. Bason et al.
References
4. Patterson J, Siew R, Herring SW, Lin AS, Guldberg R, Stayton PS. Hyaluronic acid
hydrogels with controlled degradation properties for oriented bone regeneration.
Biomaterials. 2010;31(26):6772–81.
5. Hynes RO. The extracellular matrix: not just pretty fibrils. Science. 2009;326(5957):1216–9.
6. Chan BP, Leong KW. Scaffolding in tissue engineering: general approaches and
tissue-specific considerations. Eur Spine J. 2008;17(Suppl 4):467–79.
7. Labouesse M. Role of the extracellular matrix in epithelial morphogenesis: a view from C.
elegans. Organogenesis. 2012;8(2):65–70.
8. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci.
2010;123(Pt 24):4195–200.
9. Sarrazin S, Lamanna WC, Esko JD. Heparan sulfate proteoglycans. Cold Spring Harb
Perspect Biol. 2011;3(7).
10. Schaefer L. Extracellular matrix molecules: endogenous danger signals as new drug targets
in kidney diseases. Curr Opin Pharmacol. 2010;10(2):185–90.
11. Iozzo RV, Schaefer L. Proteoglycan form and function: A comprehensive nomenclature of
proteoglycans. Matrix Biol. 2015;42:11–55.
12. Iozzo RV, Zoeller JJ, Nyström A. Basement membrane proteoglycans: modulators Par
Excellence of cancer growth and angiogenesis. Mol Cells. 2009;27(5):503–13.
13. Ori A, Wilkinson MC, Fernig DG. The heparanome and regulation of cell function:
structures, functions and challenges. Front Biosci. 2008;1(13):4309–38.
14. Boyd DF, Thomas PG. Towards integrating extracellular matrix and immunological
pathways. Cytokine. 2017;98:79–86.
15. Iozzo RV, San Antonio JD. Heparan sulfate proteoglycans: heavy hitters in the angiogenesis
arena. J Clin Invest. 2001;108(3):349–55.
16. Patel VN, Knox SM, Likar KM, Lathrop CA, Hossain R, Eftekhari S, Whitelock JM,
Elkin M, Vlodavsky I, Hoffman MP. Heparanase cleavage of perlecan heparan sulfate
modulates FGF10 activity during ex vivo submandibular gland branching morphogenesis.
Development. 2007;134(23):4177–86.
17. Goldoni S, Iozzo RV. Tumor microenvironment: modulation by decorin and related
molecules harboring leucine-rich tandem motifs. Int J Cancer. 2008;123(11):2473–9.
18. Schaefer L, Iozzo RV. Biological functions of the small leucine-rich proteoglycans: from
genetics to signal transduction. J Biol Chem. 2008;283(31):21305–9.
19. Brinckerhoff CE. Matrix metalloproteinases in health and disease: sculpting the human body.
1st ed. (Republic of Singapore): World Scientific Publishing Co; 2017.
20. Kozel BA, Ciliberto CH, Mecham RP. Deposition of tropoelastin into the extracellular
matrix requires a competent elastic fiber scaffold but not live cells. Matrix Biol. 2004;23
(1):23–34.
21. Gheduzzi D, Guerra D, Bochicchio B, Pepe A, Tamburro AM, Quaglino D, Mithieux S,
Weiss AS, Pasquali Ronchetti I. Heparan sulphate interacts with tropoelastin, with some
tropoelastin peptides and is present in human dermis elastic fibers. Matrix Biol. 2005;24
(1):15–25.
22. Trinh LA, Stainier DY. Fibronectin regulates epithelial organization during myocardial
migration in zebrafish. Dev Cell. 2004;6(3):371–82.
23. Matsui T, Raya A, Callol-Massot C, Kawakami Y, Oishi I, Rodriguez-Esteban C, Izpisúa
Belmonte JC. miles-apart-mediated regulation of cell-fibronectin interaction and myocardial
migration in zebrafish. Nat Clin Pract Cardiovasc Med. 2007;4(Suppl 1):S77–82.
24. Taylor-Weiner H, Schwarzbauer JE, Engler AJ. Defined extracellular matrix components are
necessary for definitive endoderm induction. Stem Cells. 2013;31(10):2084–94.
25. Takahashi S, Leiss M, Moser M, Ohashi T, Kitao T, Heckmann D, Pfeifer A, Kessler H,
Takagi J, Erickson HP, Fässler R. The RGD motif in fibronectin is essential for development
but dispensable for fibril assembly. J Cell Biol. 2007;178(1):167–78.
26. Pankov R, Yamada KM. Fibronectin at a glance. J Cell Sci. 2002;115(Pt 20):3861–3.
27. Singh P, Carraher C, Schwarzbauer JE. Assembly of fibronectin extracellular matrix. Annu
Rev Cell Dev Biol. 2010;26:397–419.
22 C. Bason et al.
28. Riederer I, Bonomo AC, Mouly V, Savino W. Laminin therapy for the promotion of muscle
regeneration. FEBS Lett. 2015;589(22):3449–53.
29. Mosher DF, Adams JC. Adhesion-modulating/matricellular ECM protein families: a
structural, functional and evolutionary appraisal. Matrix Biol. 2012;31(3):155–61.
30. Sweetwyne MT, Murphy-Ullrich JE. Thrombospondin1 in tissue repair and fibrosis:
TGF-b-dependent and independent mechanisms. Matrix Biol. 2012;31(3):178–86.
31. Sahni A, Francis CW. Vascular endothelial growth factor binds to fibrinogen and fibrin and
stimulates endothelial cell proliferation. Blood. 2000;96(12):3772–8.
32. Mitchell AC, Briquez PS, Hubbell JA, Cochran JR. Engineering growth factors for
regenerative medicine applications. Acta Biomater. 2016;30:1–12.
33. Hutchings H, Ortega N, Plouët J. Extracellular matrix-bound vascular endothelial growth
factor promotes endothelial cell adhesion, migration, and survival through integrin ligation.
FASEB J. 2003;17(11):1520–2.
34. Bissell MJ, Hines WC. Why don’t we get more cancer? A proposed role of the
microenvironment in restraining cancer progression. Nat Med. 2011;17(3):320–9.
35. Wu J, Strawn TL, Luo M, Wang L, Li R, Ren M, Xia J, Zhang Z, Ma W, Luo T,
Lawrence DA, Fay WP. Plasminogen activator inhibitor-1 inhibits angiogenic signaling by
uncoupling vascular endothelial growth factor receptor-2-aVb3 integrin cross talk.
Arterioscler Thromb Vasc Biol. 2015;35(1):111–20.
36. Belair DG, Le NN, Murphy WL. Design of growth factor sequestering biomaterials. Chem
Commun (Camb). 2014;50(99):15651–68.
37. Rundhaug JE. Matrix metalloproteinases and angiogenesis. J Cell Mol Med. 2005;9(2):267–
85.
38. Davis GE, Senger DR. Endothelial extracellular matrix: biosynthesis, remodeling, and
functions during vascular morphogenesis and neovessel stabilization. Circ Res. 2005;97
(11):1093–107.
39. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003;9
(6):669–76.
40. Olsson R, Maxhuni A, Carlsson PO. Revascularization of transplanted pancreatic islets
following culture with stimulators of angiogenesis. Transplantation. 2006;82(3):340–7.
41. Hsiong SX, Mooney DJ. Regeneration of vascularized bone. Periodontology. 2000;2006
(41):109–22.
42. Wijelath ES, Rahman S, Namekata M, Murray J, Nishimura T, Mostafavi-Pour Z, Patel Y,
Suda Y, Humphries MJ, Sobel M. Heparin-II domain of fibronectin is a vascular endothelial
growth factor-binding domain: enhancement of VEGF biological activity by a singular
growth factor/matrix protein synergism. Circ Res. 2006;99(8):853–60.
43. Mitsi M, Hong Z, Costello CE, Nugent MA. Heparin-mediated conformational changes in
fibronectin expose vascular endothelial growth factor binding sites. Biochemistry. 2006;45
(34):10319–28.
44. Mitsi M, Forsten-Williams K, Gopalakrishnan M, Nugent MA. A catalytic role of heparin
within the extracellular matrix. J Biol Chem. 2008;283(50):34796–807.
45. Chen LL, Johansson JK, Hodges RR, Zoukhri D, Ghinelli E, Rios JD, Dartt DA. Differential
effects of the EGF family of growth factors on protein secretion, MAPK activation, and
intracellular calcium concentration in rat lacrimal gland. Exp Eye Res. 2005;80(3):379–89.
46. Eichmann A, Simons M. VEGF signaling inside vascular endothelial cells and beyond. Curr
Opin Cell Biol. 2012;24(2):188–93.
47. Simons M. An inside view: VEGF receptor trafficking and signaling. Physiology (Bethesda).
2012;27(4):213–22.
48. Mettouchi A. The role of extracellular matrix in vascular branching morphogenesis. Cell
Adh Migr. 2012;6(6):528–34.
49. Dong X, Zhao B, Iacob RE, Zhu J, Koksal AC, Lu C, Engen JR, Springer TA. Force interacts
with macromolecular structure in activation of TGF-b. Nature. 2017;542(7639):55–9.
1 The Extracellular Matrix, Growth Factors … 23
50. Levin M, Udi Y, Solomonov I, Sagi I. Next generation matrix metalloproteinase inhibitors—
novel strategies bring new prospects. Biochim Biophys Acta. 2017;1864(11 Pt A):1927–
1939.
51. Parker A, Rees C, Clarke J, Busby WH Jr, Clemmons DR. Binding of insulin-like growth
factor (IGF)-binding protein-5 to smooth-muscle cell extracellular matrix is a major
determinant of the cellular response to IGF-I. Mol Biol Cell. 1998;9(9):2383–92.
52. Firth SM, Baxter RC. Cellular actions of the insulin-like growth factor binding proteins.
Endocr Rev. 2002;23(6):824–54.
53. Booth BA, Boes M, Andress DL, Dake BL, Kiefer MC, Maack C, Linhardt RJ, Bar K,
Caldwell EE, Weiler J, et al. IGFBP-3 and IGFBP-5 association with endothelial cells: role
of C-terminal heparin binding domain. Growth Regul. 1995;5(1):1–17.
54. Kuang Z, Yao S, Keizer DW, Wang CC, Bach LA, Forbes BE, Wallace JC, Norton RS.
Structure, dynamics and heparin binding of the C-terminal domain of insulin-like growth
factor-binding protein-2 (IGFBP-2). J Mol Biol. 2006;364(4):690–704.
55. Forbes BE, Hartfield PJ, McNeil KA, Surinya KH, Milner SJ, Cosgrove LJ, Wallace JC.
Characteristics of binding of insulin-like growth factor (IGF)-I and IGF-II analogues to the
type 1 IGF receptor determined by BIAcore analysis. Eur J Biochem. 2002;269(3):961–8.
56. Fowlkes JL, Serra DM. Characterization of glycosaminoglycan-binding domains present in
insulin-like growth factor-binding protein-3. J Biol Chem. 1996;271(25):14676–9.
57. Bach LA, Headey SJ, Norton RS. IGF-binding proteins–the pieces are falling into place.
Trends Endocrinol Metab. 2005;16(5):228–34.
58. Kricker JA, Hyde CE, Van Lonkhuyzen DR, Hollier BG, Shooter GK, Leavesley DI,
Herington AC, Upton Z. Mechanistic investigations into interactions between IGF-I and
IGFBPs and their impact on facilitating cell migration on vitronectin. Growth Factors.
2010;28(5):359–69.
59. Xu J, Liao K. Protein kinase B/AKT 1 plays a pivotal role in insulin-like growth factor-1
receptor signaling induced 3T3-L1 adipocyte differentiation. J Biol Chem. 2004;279
(34):35914–22.
60. Bunn RC, Fowlkes JL. Insulin-like growth factor binding protein proteolysis. Trends
Endocrinol Metab. 2003;14(4):176–81. Review. PubMed PMID: 12714278.
61. Boyd DF, Thomas PG. Towards integrating extracellular matrix and immunological
pathways. Cytokine. 2017;98:79–86.
62. Yan C, Boyd DD. Regulation of matrix metalloproteinase gene expression. J Cell Physiol.
2007;211(1):19–26.
63. Rilla K, Mustonen AM, Arasu UT, Härkönen K, Matilainen J, Nieminen P. Extracellular
vesicles are integral and functional components of the extracellular matrix. Matrix Biol.
2017.
64. Perea-Gil I, Prat-Vidal C, Bayes-Genis A. In vivo experience with natural scaffolds for
myocardial infarction: the times they are a-changin’. Stem Cell Res. 2015;6(6):248.
65. An M, Kwon K, Park J, Ryu DR, Shin JA, Lee Kang J, Choi JH, Park EM, Lee KE, Woo M,
Kim M. Extracellular matrix-derived extracellular vesicles promote cardiomyocyte growth
and electrical activity in engineered cardiac atria. Biomaterials. 2017;146:49–59.
66. Berardocco M, Radeghieri A, Busatto S, Gallorini M, Raggi C, Gissi C, D’Agnano I,
Bergese P, Felsani A, Berardi AC. RNA-seq reveals distinctive RNA profiles of small
extracellular vesicles from different human liver cancer cell lines. Oncotarget. 2017;8
(47):82920–39.
67. Belkin AM, Zhidkova NI, Balzac F, Altruda F, Tomatis D, Maier A, Tarone G,
Koteliansky VE, Burridge K. Beta 1D integrin displaces the beta 1A isoform in striated
muscles: localization at junctional structures and signaling potential in nonmuscle cells.
J Cell Biol. 1996;132(1–2):211–26.
68. Kääriäinen M, Nissinen L, Kaufman S, Sonnenberg A, Järvinen M, Heino J, Kalimo H.
Expression of alpha7beta1 integrin splicing variants during skeletal muscle regeneration.
Am J Pathol. 2002;161(3):1023–31.
24 C. Bason et al.
69. Tarone G, Hirsch E, Brancaccio M, De Acetis M, Barberis L, Balzac F, Retta SF, Botta C,
Altruda F, Silengo L. Integrin function and regulation in development. Int J Dev Biol.
2000;44(6):725–31. Review. Erratum in: Int J Dev Biol 2001 Sep;45(5–6): following 770.
70. Jansen KA, Donato DM, Balcioglu HE, Schmidt T, Danen EH, Koenderink GH. A guide to
mechanobiology: where biology and physics meet. Biochim Biophys Acta. 2015;1853(11 Pt
B):3043–52.
71. Jaalouk DE, Lammerding J. Mechanotransduction gone awry. Nat Rev Mol Cell Biol.
2009;10(1):63–73.
72. Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression.
J Cell Biol. 2012;196(4):395–406.
73. Snedeker JG, Gautieri A. The role of collagen crosslinks in ageing and diabetes—the good,
the bad, and the ugly. Muscles Ligaments Tendons J. 2014;4(3):303–8.
74. Cukierman E, Pankov R, Stevens DR, Yamada KM. Taking cell-matrix adhesions to the
third dimension. Science. 2001;294(5547):1708–12.
75. Winograd-Katz SE, Fässler R, Geiger B, Legate KR. The integrin adhesome: from genes and
proteins to human disease. Nat Rev Mol Cell Biol. 2014;15(4):273–88.
76. Sun Z, Guo SS, Fässler R. Integrin-mediated mechanotransduction. J Cell Biol. 2016;215
(4):445–56.
77. Geiger B, Tokuyasu KT, Dutton AH, Singer SJ. Vinculin, an intracellular protein localized
at specialized sites where microfilament bundles terminate at cell membranes. Proc Natl
Acad Sci USA. 1980;77(7):4127–31.
78. Atherton P, Stutchbury B, Jethwa D, Ballestrem C. Mechanosensitive components of
integrin adhesions: role of vinculin. Exp Cell Res. 2016;343(1):21–7.
79. Dupont S. Role of YAP/TAZ in cell-matrix adhesion-mediated signalling and mechan-
otransduction. Exp Cell Res. 2016;343(1):42–53.
80. Elbediwy A, Vincent-Mistiaen ZI, Thompson BJ. YAP and TAZ in epithelial stem cells: a
sensor for cell polarity, mechanical forces and tissue damage. BioEssays. 2016;38(7):644–
53.
81. Kim IG, Gil CH, Seo J, Park SJ, Subbiah R, Jung TH, Kim JS, Jeong YH, Chung HM,
Lee JH, Lee MR, Moon SH, Park K. Mechanotransduction of human pluripotent stem cells
cultivated on tunable cell-derived extracellular matrix. Biomaterials. 2018;150:100–11.
82. Clause KC, Barker TH. Extracellular matrix signaling in morphogenesis and repair. Curr
Opin Biotechnol. 2013;24(5):830–3.
83. Lubarsky B, Krasnow MA. Tube morphogenesis: making and shaping biological tubes. Cell.
2003;112(1):19–28.
84. Hoffman MP, Kidder BL, Steinberg ZL, Lakhani S, Ho S, Kleinman HK, Larsen M. Gene
expression profiles of mouse submandibular gland development: FGFR1 regulates branching
morphogenesis in vitro through BMP- and FGF-dependent mechanisms. Development.
2002;129(24):5767–78.
85. Nam JM, Onodera Y, Bissell MJ, Park CC. Breast cancer cells in three-dimensional culture
display an enhanced radioresponse after coordinate targeting of integrin alpha5beta1 and
fibronectin. Cancer Res. 2010;70(13):5238–48.
86. Avizienyte E, Wyke AW, Jones RJ, McLean GW, Westhoff MA, Brunton VG, Frame MC.
Src-induced de-regulation of E-cadherin in colon cancer cells requires integrin signalling.
Nat Cell Biol. 2002;4(8):632–8.
87. Weber GF, Bjerke MA, DeSimone DW. A mechanoresponsive cadherin-keratin complex
directs polarized protrusive behavior and collective cell migration. Dev Cell. 2012;22
(1):104–15.
88. Huveneers S, Danen EH. Adhesion signaling—crosstalk between integrins, Src and Rho.
J Cell Sci. 2009;122(Pt 8):1059–69.
89. Watanabe T, Sato K, Kaibuchi K. Cadherin-mediated intercellular adhesion and signaling
cascades involving small GTPases. Cold Spring Harb Perspect Biol. 2009;1(3):a003020.
90. Van Aelst L, Symons M. Role of Rho family GTPases in epithelial morphogenesis. Genes
Dev. 2002;16(9):1032–54.
1 The Extracellular Matrix, Growth Factors … 25
91. Reddi AH. Cartilage morphogenetic proteins: role in joint development, homoeostasis, and
regeneration. Ann Rheum Dis. 2003;62(Suppl 2):ii73–8.
92. Bondow BJ, Faber ML, Wojta KJ, Walker EM, Battle MA. E-cadherin is required for
intestinal morphogenesis in the mouse. Dev Biol. 2012;371(1):1–12.
93. Iruela-Arispe ML, Beitel GJ. Tubulogenesis. Development. 2013;140(14):2851–5.
94. Lampugnani MG. Endothelial adherens junctions and the actin cytoskeleton: an ‘infinity
net’? J Biol. 2010;9(3):16.
95. Brinkmann BF, Steinbacher T, Hartmann C, Kummer D, Pajonczyk D, Mirzapourshafiyi F,
Nakayama M, Weide T, Gerke V, Ebnet K. VE-cadherin interacts with cell polarity protein
Pals1 to regulate vascular lumen formation. Mol Biol Cell. 2016;27(18):2811–21.
96. Gebala V, Collins R, Geudens I, Phng LK, Gerhardt H. Blood flow drives lumen formation
by inverse membrane blebbing during angiogenesis in vivo. Nat Cell Biol. 2016;18(4):443–
50.
97. Galie PA, Nguyen DH, Choi CK, Cohen DM, Janmey PA, Chen CS. Fluid shear stress
threshold regulates angiogenic sprouting. Proc Natl Acad Sci USA. 2014;111(22):7968–73.
98. Barry AK, Wang N, Leckband DE. Local VE-cadherin mechanotransduction triggers
long-ranged remodeling of endothelial monolayers. J Cell Sci. 2015;128(7):1341–51.
99. Wang S, Sekiguchi R, Daley WP, Yamada KM. Patterned cell and matrix dynamics in
branching morphogenesis. J Cell Biol. 2017;216(3):559–70.
100. Schuchardt A, D’Agati V, Larsson-Blomberg L, Costantini F, Pachnis V. Defects in the
kidney and enteric nervous system of mice lacking the tyrosine kinase receptor Ret. Nature.
1994;367(6461):380–3.
101. Luetteke NC, Qiu TH, Fenton SE, Troyer KL, Riedel RF, Chang A, Lee DC. Targeted
inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor
ligands in mouse mammary gland development. Development. 1999;126(12):2739–50.
102. De Moerlooze L, Spencer-Dene B, Revest JM, Hajihosseini M, Rosewell I, Dickson C. An
important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in
mesenchymal-epithelial signalling during mouse organogenesis. Development. 2000;127
(3):483–92.
103. Hennighausen L, Robinson GW. Information networks in the mammary gland. Nat Rev Mol
Cell Biol. 2005;6(9):715–25.
104. Costantini F, Shakya R. GDNF/Ret signaling and the development of the kidney. BioEssays.
2006;28(2):117–27.
105. Djouad F, Delorme B, Maurice M, Bony C, Apparailly F, Louis-Plence P, Canovas F,
Charbord P, Noël D, Jorgensen C. Microenvironmental changes during differentiation of
mesenchymal stem cells towards chondrocytes. Arthritis Res Ther. 2007;9(2):R33.
106. Lu P, Werb Z. Patterning mechanisms of branched organs. Science. 2008;322(5907):1506–
9.
107. Affolter M, Zeller R, Caussinus E. Tissue remodelling through branching morphogenesis.
Nat Rev Mol Cell Biol. 2009;10(12):831–42.
108. Costantini F, Kopan R. Patterning a complex organ: branching morphogenesis and nephron
segmentation in kidney development. Dev Cell. 2010;18(5):698–712.
109. Blake J, Rosenblum ND. Renal branching morphogenesis: morphogenetic and signaling
mechanisms. Semin Cell Dev Biol. 2014;36:2–12.
110. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general
approaches and a review of recent developments. J R Soc Interface. 2011;8(55):153–70.
111. Ferrara N. Binding to the extracellular matrix and proteolytic processing: two key
mechanisms regulating vascular endothelial growth factor action. Mol Biol Cell. 2010;21
(5):687–90.
112. Hankenson KD, Gagne K, Shaughnessy M. Extracellular signaling molecules to promote
fracture healing and bone regeneration. Adv Drug Deliv Rev. 2015;1(94):3–12.
113. Marti-Figueroa CR, Ashton RS. The case for applying tissue engineering methodologies to
instruct human organoid morphogenesis. Acta Biomater. 2017;54:35–44.
26 C. Bason et al.
114. Webb DJ, Roadcap DW, Dhakephalkar A, Gonias SL. A 16-amino acid peptide from human
alpha2-macroglobulin binds transforming growth factor-beta and platelet-derived growth
factor-BB. Protein Sci. 2000;9(10):1986–92.
115. Ezquerro IJ, Lasarte JJ, Dotor J, Castilla-Cortázar I, Bustos M, Peñuelas I, Blanco G,
Rodríguez C, Lechuga Mdel C, Greenwel P, Rojkind M, Prieto J, Borrás-Cuesta F.
A synthetic peptide from transforming growth factor beta type III receptor inhibits liver
fibrogenesis in rats with carbon tetrachloride liver injury. Cytokine. 2003;22(1–2):12–20.
Erratum in: Cytokine. 2006;33(2):119.
116. Serratì S, Margheri F, Pucci M, Cantelmo AR, Cammarota R, Dotor J, Borràs-Cuesta F,
Fibbi G, Albini A, Del Rosso M. TGFbeta1 antagonistic peptides inhibit
TGFbeta1-dependent angiogenesis. Biochem Pharmacol. 2009;77(5):813–25.
117. Martino MM, Briquez PS, Ranga A, Lutolf MP, Hubbell JA. Heparin-binding domain of
fibrin(ogen) binds growth factors and promotes tissue repair when incorporated within a
synthetic matrix. Proc Natl Acad Sci USA. 2013;110(12):4563–8.
118. Terada T, Mizobata M, Kawakami S, Yabe Y, Yamashita F, Hashida M. Basic fibroblast
growth factor-binding peptide as a novel targeting ligand of drug carrier to tumor cells.
J Drug Target. 2006;14(8):536–45.
119. Modaresifar K, Hadjizadeh A, Niknejad H. Design and fabrication of GelMA/chitosan
nanoparticles composite hydrogel for angiogenic growth factor delivery. Artif Cells
Nanomed Biotechnol. 2017;24:1–10.
120. Berkovitch Y, Cohen T, Peled E, Schmidhammer R, Florian H, Teuschl A, Wolbank S,
Yelin D, Redl H, Seliktar D. Hydrogel composition and laser micro-patterning to regulate
sciatic nerve regeneration. J Tissue Eng Regen Med. 2017.
121. Paduano F, Marrelli M, Alom N, Amer M, White LJ, Shakesheff KM, Tatullo M.
Decellularized bone extracellular matrix and human dental pulp stem cells as a construct for
bone regeneration. J Biomater Sci Polym Ed. 2017;28(8):730–48.
122. Hughes CS, Postovit LM, Lajoie GA. Matrigel: a complex protein mixture required for
optimal growth of cell culture. Proteomics. 2010;10(9):1886–90.
123. Lindborg BA, Brekke JH, Vegoe AL, Ulrich CB, Haider KT, Subramaniam S,
Venhuizen SL, Eide CR, Orchard PJ, Chen W, Wang Q, Pelaez F, Scott CM, Kokkoli E,
Keirstead SA, Dutton JR, Tolar J, O’Brien TD. Rapid induction of cerebral organoids from
human induced pluripotent stem cells using a chemically defined hydrogel and defined cell
culture medium. Stem Cells Transl Med. 2016;5(7):970–9.
124. Ranga A, Girgin M, Meinhardt A, Eberle D, Caiazzo M, Tanaka EM, Lutolf MP. Neural
tube morphogenesis in synthetic 3D microenvironments. Proc Natl Acad Sci USA. 2016;113
(44):E6831–9.
125. Boccardo S, Gaudiello E, Melly L, Cerino G, Ricci D, Martin I, Eckstein F, Banfi A,
Marsano A. Engineered mesenchymal cell-based patches as controlled VEGF delivery
systems to induce extrinsic angiogenesis. Acta Biomater. 2016;15(42):127–35.
126. Lorentz KM, Yang L, Frey P, Hubbell JA. Engineered insulin-like growth factor-1 for
improved smooth muscle regeneration. Biomaterials. 2012;33(2):494–503.
127. Miller RE, Grodzinsky AJ, Cummings K, Plaas AH, Cole AA, Lee RT,
Patwari P. Intraarticular injection of heparin-binding insulin-like growth factor 1 sustains
delivery of insulin-like growth factor 1 to cartilage through binding to chondroitin sulfate.
Arthritis Rheum. 2010;62(12):3686–94.
128. Sun W, Lin H, Chen B, Zhao W, Zhao Y, Xiao Z, Dai J. Collagen scaffolds loaded with
collagen-binding NGF-beta accelerate ulcer healing. J Biomed Mater Res A. 2010;92(3):
887–95.
Chapter 2
ECM Hydrogels for Regenerative
Medicine
Abstract The ECM is a highly complex mix of structural and functional proteins
and other biomolecules. These molecules are secreted by the cells resident in every
tissue in the body but can also influence their behavior through a process of “dy-
namic reciprocity.” As a result, there has been significant interest in utilizing ECM
as a biologic scaffold material in tissue repair and replacement. Numerous pre-
clinical and clinical studies have demonstrated the efficacy of ECM biomaterials,
and more than 4 million patients have now been treated with these scaffold mate-
rials. The discovery that these materials could be formed into hydrogels promised to
further expand their clinical utility by offering minimally invasive delivery and the
ability to fill irregularly shaped defects. This chapter will briefly outline the history
and characterization of ECM biomaterials and their evolution from single sheet to
multisheet, powder, and ultimately hydrogel form. The first studies describing the
production of early-generation ECM hydrogels used well-characterized porcine
small intestinal submucosa and urinary bladder matrix, and these materials will be
discussed in the context of the general methods used to produce and characterize
ECM hydrogels. A detailed consideration of the many second-generation hydrogels
which have since been produced from a wide range of tissues will then be discussed
M. J. Sawkins
Department of Anatomy, Royal College of Surgeons in Ireland,
123 St. Stephen’s Green, Dublin 2, Ireland
L. T. Saldin
Department of Bioengineering, University of Pittsburgh,
Pittsburgh, PA 15219, USA
e-mail: [email protected]
S. F. Badylak
Surgery and Bioengineering, University of Pittsburgh, 450 Technology Drive,
Suite 319, Pittsburgh, PA 15219, USA
e-mail: [email protected]
L. J. White (&)
School of Pharmacy, University of Nottingham, University Park,
Nottingham NG7 2RD, UK
e-mail: [email protected]
in the context of tissue specificity. The hydrogels discussed in this chapter have
been evaluated in vitro or in small scale in vivo animal studies. More substantial
evaluation is required before these materials can be considered ready for clinical
application, but these hydrogels provide the possibility for minimally invasive
delivery, treatment of irregularly shaped defects in anatomic sites that prove
challenging for invasive surgical procedures, and may provide an ECM formation
that delivers immediate bioactivity as a consequence of its distinctive biomolecular
composition.
Keywords Adipose Brain Cartilage Central nervous system
Cornea Decellularization Demineralized bone matrix Dermis
Extracellular matrix Hydrogel Intervertebral disk Ligament
Liver Myocardium Nucleus pulposus Pericardium Regenerative medicine
Skeletal muscle Skin Spinal cord Small intestinal submucosa
Tendon Tissue engineering Tissue specificity Urinary bladder matrix
Vocal fold
Biochemical Content
The ECM consists of the structural and functional proteins secreted by the cells
resident in each tissue and organ and is a primary component of the cell’s
microenvironmental niche. ECM is comprised primarily of fibrillar collagen but
also contains glycoproteins (laminin, fibronectin), proteoglycans, glycosaminogly-
cans (GAGs), and growth factors. The 3D arrangement and biochemical compo-
sition of these proteins are specific to tissue type [59]. Ideally, an ECM-derived
material would preserve the components of native tissue ECM. However, the choice
of decellularization agents, techniques, and post-processing terminal sterilization
steps to produce ECM-derived materials will unavoidably affect the ECM bio-
chemical properties. The balance lies in achieving decellularization and sterility
criteria while best preserving ECM composition and structure [28]. Thin tissues
such as urinary bladder matrix (UBM) and small intestinal submucosa (SIS) may be
effectively decellularized with mechanical scraping, agitation in a dilute acidic
solution (0.1% peracetic acid), and subsequent rinsing with deionized water. UBM
and SIS contain growth factor and GAG levels similar to native ECM after de-
cellularization [3, 88]. Brown et al. [18] reported the urinary bladder matrix con-
tained an intact basement membrane after decellularization.
In contrast, denser tissues such as porcine dermis require a harsher decellular-
ization protocol, usually involving a combination of detergents, organic solvents,
and enzymatic solutions. Reing et al. [67] tested the effect of different processing
steps on the properties of porcine dermal matrix. While a trypsin/Triton X-100
protocol moderately affected ECM properties, the addition of a sodium dodecyl
sulfate (SDS) processing step more severely affected ECM mechanical strength,
growth factor, and GAG content, and the ability of the ECM to support cell growth
in vitro. The results may not be surprising considering Triton X-100 is a relatively
gentle nonionic detergent and trypsin is an enzyme that selectively cleaves cell
adherent proteins to detach cells from the tissue surface. In contrast, SDS is a more
harsh ionic detergent that can disrupt protein covalent bonding.
When processed appropriately, ECM-derived materials have many advantages
compared to their synthetic counterparts. ECM-derived materials recapitulate the
rich natural diversity of proteins, unlike matrices that are manufactured from
individual molecular components of the extracellular matrix (e.g., laminin or col-
lagen gels). Furthermore, the GAGs in the ECM-derived material present growth
factors to cell surface receptors in biologically relevant ratios and 3D conformations
[3]. It is well known that growth factors have an important impact on regulating cell
behavior [83]. Finally, non-chemically cross-linked ECM-derived materials degrade
and release biochemical signals at a similar rate to native tissue ECM.
30 M. J. Sawkins et al.
Bioinductive Properties
Antimicrobial Properties
Chemoattraction
Badylak et al. [7] used a mouse model to show that a population of bone marrow
cells contributes to constructive tissue remodeling. Briefly, mice deficient for the
b-galactosidase gene were lethally irradiated and received both a transplantation of
b-galactosidase+ bone marrow cells, and one of 4 scaffold implants. The mice
received ECM-derived materials (SIS or urinary bladder submucosa) or control
2 ECM Hydrogels for Regenerative Medicine 31
Macrophage Polarization
The host remodeling response to SIS and its chemically cross-linked form has
been characterized using autologous graft tissue as a control [8]. SIS elicited a
predominantly “M2” profile (CD163+) when implanted into a rat model, with
long-term constructive remodeling evidenced by vascularization, new skeletal
muscle fiber formation, and organized connective tissue at 16 weeks. Conversely,
the chemically cross-linked SIS elicited a predominantly “M1” profile (CD80+,
CCR7+) with chronic inflammation at 16 weeks evidenced by fibrosis and scarring
around the implant, presence of multinucleated giant cells, and the associated
classic foreign body response. The autologous tissue graft elicited a mixed M1/M2
profile at 16 weeks with moderately well-organized scar tissue, but no detectable
new muscle fibers. A subsequent study showed that the presence of a cellular
component, even an autologous one, polarized macrophages to an M1 profile [20].
Furthermore, the M2:M1 ratio for 14 commercially available ECM-derived mate-
rials was shown to correlate with in vivo constructive remodeling histologic scores
at 14 and 35 days post-implantation, an indication of the broad applicability of this
concept [19].
The first ECM scaffolds were intact sheets (60 lm), reflecting “skilled decellular-
ization” [3]. The fragile tissue layer of interest had to be carefully separated from
other layers and withstand the scraping and decellularization process without
tearing. These initial sheets were typically stored as lyophilized sheets and hydrated
before use [5]. ECM sheets are commonly used for wound repair (e.g., partial and
full thickness wounds, ulcers, second-degree burns, trauma wounds). A common
misconception in the use of ECM-derived materials is the requirement of the
material to replace volumetric tissue loss and provide mechanical strength at the
wound site. Instead, the ECM sheets are intended to serve as a bioinductive tem-
plate that degrades as the tissue is restored.
Single-layer ECM sheets may be insufficient for regenerative medicine appli-
cations with load-bearing requirements (e.g., esophagus repair, laryngeal recon-
struction, abdominal wall reconstruction, hernia repair, pelvic floor). ECM-derived
materials were vacuum compressed to form multilayer devices (4–8 layers) to
increase the strength of the material. Collagen fiber orientation has been used
advantageously in sheet stacking to improve the strength of the material device. For
example, a 10-layer ECM device (Restore®, DePuy Orthopedics) was stacked with
collagen fibers at 72° angles every 2 layers resulting in an isotropic construct that
matched the strength of the tendon tissue it was intended to replace [5]. In addition,
vacuum pressing allowed for the creation of a variety of 3D shapes for certain
applications (e.g., tube for trachea, funnel for esophageal anastomosis) [9].
ECM-derived materials can be powdered to apply topically to the wound site and
fill irregular defects (e.g., digit amputation) [40]. The particulates derived from
powdering are of the order of 50–200 um in diameter and retain the surface
2 ECM Hydrogels for Regenerative Medicine 33
topology of the native matrix [5, 72]. The ECM powder may also be suspended in a
pharmacologically approved carrier (e.g., saline, water). High concentrations of
ECM powder suspension (e.g., 500 mg/mL) can form a putty with complex rhe-
ological behavior which has been applied to fill tissue defects [22].
Gelation of the first SIS and UBM hydrogels was based on well-established pro-
tocols for collagen gelation. Native collagen is a main constituent of the ECM
hydrogel, and its gelation characteristics have been extensively characterized [42,
46, 47, 62]. Although ECM hydrogel preparation has been described with respect to
collagen biochemistry and fibrillogenesis, it is important to note that ECM hydrogel
formation is undoubtedly influenced by the presence of other ECM components
[42]. While bulk mechanical properties of the hydrogel can be described, further
work is required to understand microscale differences that affect collagen poly-
merization and are manifested in distinct viscoelastic properties.
The pepsin enzyme is used in both the SIS and UBM digestion protocols. While
pepsin, derived from the gastric juices, is not a physiologically relevant method for
ECM degradation, biochemists have been using pepsin since 1972 to solubilize a
substantial portion (up to 99%) of the acid-insoluble collagen [33, 57]. It has been
shown that pepsin selectively cleaves telopeptides, the protein domains external to
the triple helix-body of the tropocollagen molecule [42]. These telopeptides form
the intramolecular bonds between the collagen fibril aggregates [33, 70]. The
unraveling of the collagen fibril aggregates by pepsin digestion produces almost
entirely monomeric segments [70]. Visible changes are observed as the pepsin in
dilute acid solubilizes the ECM powder and the suspension transforms into an
opaque, viscous solution (“solubilized ECM”). The solubilized ECM is ready for
gelation when the liquid is homogenous, with no visible particles [45].
Another general method of protein solubilization which has been applied to
ECM materials is homogenization, either by pestle and mortar or high-speed shear
mixer. This is carried out in the presence of a high salt buffer and acts to physically
break down the ECM particles and disrupt the collagen fiber structure [23, 25, 37,
64, 65, 84, 85]. Prior to this physical processing, the material is often treated with
dispase, a specific protease which cleaves fibronectin, collagen IV, and collagen I
[23, 64, 65, 84, 85]. The treatment is used primarily to decellularize the tissue but
will also begin to digest the ECM protein structure. After dispase treatment and
homogenization, the ECM is then typically further processed via one or more urea
extraction steps [23, 37, 64, 65, 84, 85]. Urea in high concentrations acts to disrupt
the non-covalent bonding which is essential to protein folding and structure and
thereby denatures many proteins and increases their solubility. Centrifugation can
be used to remove any remaining insoluble ECM components leaving a viscous,
cloudy solution similar to that obtained by pepsin digestion.
The solubilized ECM is neutralized to physiologic pH and salt concentration and
is referred to as “pre-gel solution.” Gelation is chiefly determined by the poly-
merization of fibrillar collagen as the primary ECM constituent. When the tem-
perature is raised to physiologic conditions (37 °C), solubilized collagen can
self-assemble in an entropy-driven process. It is energetically favorable for collagen
monomers to lose surface water and bind to aggregates in order to bury
hydrophobic residues within the fibril [42, 62]. Self-assembly follows four
2 ECM Hydrogels for Regenerative Medicine 35
Biocompatibility
Biochemical Composition
Studies of UBM and SIS gels provided confirmation that the hydrogels possessed
similar biochemical compositions to the intact UBM and SIS scaffolds in terms of
collagen and GAG content [17, 38, 90]. Intact SIS contained 90% collagen, pri-
marily collagen I, with minor amounts of collagen III, IV, V, and VI. Intact UBM
had a similar composition to intact SIS, but with greater amounts of collagen III and
the additional presence of collagen VII [5]. SIS hydrogels were shown to contain at
least three types of collagen (predominantly collagen I and collagen III with minor
amounts of collagen IV), and sulfated GAGs [17]. Gel electrophoresis of the UBM
gel displayed similar bands to purified collagen I, with additional bands present for
other ECM proteins [38].
Recognizing the biochemical complexity of ECM hydrogels, mass spectroscopy
is beginning to be used to determine the proteomic profile of pepsin-solubilized
ECM [14, 76]. Published studies to date have used reversed-phase
high-performance liquid chromatography interfaced with tandem mass spec-
troscopy (LC-MS/MS) and compared the peptide fragments generated with a
protein data bank (Swiss Protein data bank). SIS and UBM hydrogels have yet to be
characterized by LC-MS/MS.
Gel Topology
Upon gelation, SIS, and UBM hydrogels form porous, nanofibrous structures,
which provide cells with a dynamic 3D environment. In contrast to other forms of
ECM-derived materials (sheet and powder), the ECM hydrogels lose the native 3D
structure. However, the complex nanoscale mesh of ECM fibers guides cellular
behavior via contact guidance and fibril-bound proteins that interact with cell
membrane-bound integrin receptors.
Visualization of the SIS and UBM gel topology with scanning electron micro-
scopy (SEM) showed that both hydrogels formed loosely organized, interwoven
networks of fibers [38]. D’Amore et al. [31] developed a MATLAB algorithm that
enables high-throughput, automated analysis of SEM images to more accurately
assess the characteristics of the overall fiber network. Using the image-based
algorithm, UBM hydrogel characteristics were assessed [90]. The UBM hydrogels
had a fiber diameter of 0.074 ± 0.004 lm and pore size of 0.112 ± 0.005 lm,
which is within biological criteria for hydrogel design. Fiber alignment was ran-
domly organized (isotropic) as characterized by fiber alignment close to 0% with
respect to any axis. The described metrics were concentration-independent for
UBM hydrogels, but ECM concentration has been shown to be an important
covariate of fiber diameter and porosity for other ECM hydrogels, such as dermal
matrix hydrogels [90].
Tissue-Specific Hydrogels
Heart
Among the most frequently targeted tissues is cardiac muscle, primarily with the
aim of restoring heart function after myocardial infarction (MI), which is the
leading cause of death in most industrialized countries (Rosamond et al. [69].
Hydrogels from heart tissue have been developed, characterized, and applied to
tissue repair in a number of studies published by Christman and colleagues [44, 45,
75–77, 80, 81]. A protocol was developed for the production of decellularized
matrix from porcine myocardium via a detergent-based technique using sodium
dodecyl sulfate (SDS) and Triton X-100 [80]. Pepsin digestion of this matrix
resulted in a solution which underwent thermoresponsive gelation both in vitro and
in vivo after catheter-based delivery, thus suggesting that the material had good
clinical handling properties. As with many other ECM materials, the myocardial
matrix was shown to be composed predominantly of collagen, but also to contain a
significant amount of GAGs. The most important result of this initial study, how-
ever, was that both smooth muscle cells and endothelial cells migrated toward the
matrix in vitro and in vivo to a greater degree than toward collagen or fetal bovine
serum (FBS). The cell-free material may therefore be sufficient to recruit appro-
priate cell types to defect sites and to ultimately form appropriate new tissue.
Further development of myocardial matrix was shown in a subsequent study which
demonstrated that the fiber size and structure, mechanical properties, and gelation
kinetics could be modulated by alterations of the pH, temperature, ionic strength,
and concentration used for gelation [45]. Modification of the hydrogel in this
fashion could allow the handling properties and cell response to be tailored to
specific applications.
Most recently, the treatment efficacy of the material has been evaluated in both
rat and porcine preclinical models of MI [77, 81]. In the rat model, myocardial
matrix was injected into the infarcted area 1 week after MI and led to significantly
increased numbers of cardiomyocytes and proliferative cells 1 month after injection
when compared to saline-injected controls [81]. Cardiac function was also signif-
icantly increased at this time point as measured by parameters such as the ejection
fraction, end-diastolic dimension, and end-systolic volume. In a later study in the
porcine model [77], myocardial matrix was delivered into the affected tissue
2 weeks after the induction of MI. Treatment contributed to an increased muscle
volume in the affected area when compared to saline-injected controls 3 months
post-MI, and significant increases in cardiac function were again observed. Taken
together the results of these studies suggest great promise for this material in
40 M. J. Sawkins et al.
the protein was delivered in matrix than in collagen or saline, and this resulted in
significant increases in arteriole density. Most importantly, the newly formed
vasculature was anastomosed with that preexisting in the host, allowing it to
function to restore oxygen supply to the ischemic tissue. The next stage for the
development of pericardial matrix will be to demonstrate that the observed
improvements in neo-vessel formation result in recovery of cardiac function, as
observed for myocardial matrix.
In addition to the heart-derived materials described thus far, porcine bone
marrow has also been considered as a source tissue for hydrogel biomaterials to
treat MI [66]. A series of nuclease, ethylenediaminetetraacetic acid (EDTA), vortex
and centrifuge steps were used to produce a bone marrow ECM extract which
contained a number of growth factors important in angiogenesis and cardiac repair.
Coating non-tissue culture-treated polystyrene (non-TCPS) with this extract was
found to significantly increase endothelial cell adherence relative to fibronectin.
Fibronectin is an effective single component substrate coating containing many
RGD peptide sequences which can interact with integrins on the cell surface. The
ability of the bone marrow ECM-derived material to support greater cell attachment
is therefore a highly significant result. For three-dimensional hydrogel studies, the
bone marrow extract was reinforced with methylcellulose in order to improve
material handling and retention at the injection site in vivo. When this reinforced
material was delivered into a rat MI model, the infarct area was rapidly reduced
relative to untreated controls, with associated increases in cardiac function.
Furthermore, 3 weeks post-intervention there was a significant reduction in the
numbers of apoptotic cells and macrophages present, alongside increases in stem
cell and blood vessel numbers. This bone marrow-derived ECM material may
therefore be a useful supplement to other biomaterials utilized for cardiac repair
post-MI.
As one of the most heavily targeted tissues for the development of ECM-derived
hydrogel biomaterials, it is perhaps unsurprising that proposed therapies in the area
of post-MI cardiac repair are also among the most well-developed. All three distinct
materials, developed from different source tissues, have shown, in principle at least,
that they can contribute to the generation of functional repair tissue in vivo.
Fat
Like cardiac tissue, fat has also been widely studied as a target for repair by
extracellular matrix biomaterials [23, 65, 85, 92]. If a successful therapy could be
developed, then it could be used in a large number of procedures following trauma
and tumor resection which require the reconstruction of significant volumes of
adipose tissue. The use of ECM hydrogels to support adipogenesis was first
explored in a pair of studies by Brey and coworkers [23, 84]. In the first of these
studies, a combination of dispase digestion, high salt homogenization, and urea
extraction were used to produce rat adipose tissue extracts which underwent
42 M. J. Sawkins et al.
autologous or allogeneic, rather than xenogeneic tissue as the ideal source for
scaffold material as it carries less risk of disease transmission or adverse immune
response.
Skin
In an attempt to generate biomaterials for wound healing and skin repair, hydrogels
have been derived from rat dermis by a combination of dispase treatment, high salt
homogenization, and urea extraction to produce tissue extracts to undergo thermo-
or pH-responsive gelation [64, 85]. In the first of these studies, it was shown that
both endothelial cells and fibroblasts could proliferate on these dermis-derived
biomaterials and that endothelial cells formed tubule networks as an indicator of the
potential for blood vessel formation [85]. However, it is noteworthy that fibroblasts
placed into the gels as spheroids were unable to migrate into the substrate as they
did when cultured in MatrigelTM.
The second study examined the effects of glutaraldehyde cross-linking on the
dermis-derived hydrogels [64]. Cross-linking was shown to significantly increase
the elastic moduli and yield stresses of the hydrogels and to significantly slow
enzymatic degradation in vitro and also after subcutaneous implantation in the rat.
Cross-linked gels exhibited minimal cytotoxicity provided they were adequately
washed prior to exposure to cells or tissue. Furthermore, glutaraldehyde
cross-linking led to significantly increased blood vessel density in the tissue formed
after implantation, suggesting that it has the potential to improve the performance of
these materials in wound healing.
Dermis-derived hydrogels show promise for application in wound healing as
they permit proliferation of relevant cell types and can support new blood vessel
formation both in vitro and in vivo. Nevertheless, it remains to be established that
fibroblasts can migrate into and deposit ECM proteins within these materials. This
is a necessary step if the appropriate structural matrix is to be deposited around the
neovasculature formed in these scaffolds.
Liver
Liver is another tissue in which the potential for ECM-derived hydrogels to support
tissue formation has been established in principle [52, 78]. Initially, a porcine liver-
derived gel was produced using a combined surfactant and enzyme-based approach
to decellularize the tissue, followed by pepsin digestion to solubilize the protein
mixture [78]. It was shown that primary human hepatocytes grown in a sandwich
culture between two layers of liver-derived matrix expressed relevant genes and
proteins as highly as those grown between layers of MatrigelTM, the gold standard
substrate. Subsequently, promising hydrogel materials were generated from rat liver
44 M. J. Sawkins et al.
Skeletal Muscle
The use of hydrogels for the repair and regeneration of damaged skeletal muscle has
been investigated using two distinct materials in two distinct models [32, 91].
Firstly, a hydrogel material was developed from porcine skeletal muscle and
evaluated in a rat hindlimb ischemia model [32]. The source tissue was decellu-
larized by the action of SDS, with pepsin digestion used for ECM solubilization.
The solution resulting from this process was found to significantly enhance pro-
liferation of both rat smooth muscle cells and C2C12 myoblasts relative to
pepsin-digested collagen as an in vitro culture supplement. The results of in vivo
experiments in which the muscle-derived hydrogel was injected intramuscularly
into ischemic tissue were also positive. Relative to collagen gels, those derived
from skeletal muscle matrix resulted in significant increases in arteriole density,
endothelial cell numbers, and proliferating muscle cell numbers at all time points
over a 2-week period post-injection.
The second type of hydrogel considered for skeletal muscle repair was devel-
oped from porcine dermis by a combined enzyme, surfactant, and peracetic acid
treatment, followed by pepsin digestion [91]. The performance of this material was
compared to that of a gel derived from UBM both in vitro and in a partial-thickness
rat abdominal wall defect. In vitro results clearly suggested that dermal ECM was a
superior scaffold material for this application. The C2C12 myoblast cell line more
efficiently fused to form myotubes on this substrate, and it was found to be sig-
nificantly more resistant to fibroblast-mediated contraction. The same resistance to
contraction was also found after in vivo implantation, and this is important in
ensuring scar-free wound healing. However, UBM hydrogels contained
2 ECM Hydrogels for Regenerative Medicine 45
the pre-gel solutions were tested as culture medium additives to assess the effects of
ECM-derived soluble peptides. None of the three materials adversely affected the
viability of either neural stem cells (NSCs) or perivascular stem cells at concen-
trations of up to 100 lg mL−1. Furthermore, all three were able to exert positive
chemotactic effects on both cell types relative to FBS controls, though both the
presence and size of these effects were dose- and source tissue-dependent. UBM
pre-gel solutions were also able to significantly increase the proliferation rate of
both stem cell types at all concentrations, while the CNS-derived solutions
exhibited more complex, dose-dependent effects. However, both CNS-derived
solutions were able to direct neuronal differentiation of NSCs with equivalent
efficiency to the positive control differentiation medium. The observed expression
of bIII-Tubulin and neurite extension suggested that the CNS-derived materials
promote a less proliferative, more differentiated, and functional phenotype and
could drive tissue repair by endogenous stem cells. The conclusions of this
investigation suggest that in the case of CNS repair, a match between the source and
target tissues may be important for cell maturation and tissue formation.
Cartilage
The first ECM-derived material considered for cartilage repair was an extract from
human lipoaspirate, produced by homogenization, followed by SDS and nuclease
treatment [25]. A number of relevant growth factors were identified in this tissue
extract, including transforming growth factor-b1 (TGF-b1), an important mediator
of chondrogenic differentiation. Pellet cultures of this material in combination with
ASCs showed that the cells retained high viability across a culture period of more
than 6 weeks. The inclusion of the adipose-derived material in the pellets, with or
without the addition of TGF-b1, led to significant increases in total protein, col-
lagen, and sulfated GAG content of cultures indicating greater new matrix syn-
thesis. Important cartilage markers such as SOX9, aggrecan, and collagen types II
and X were expressed by cells in all ECM-containing pellets, though more strongly
in those cultured in the presence of TGF-b1. This suggests further that the matrix
being synthesized in these constructs had an appropriate, cartilage-like composition.
Although in vivo studies are necessary for progression, the fact that this material
could direct both stem cell differentiation and cartilage-like tissue formation in vitro
is nonetheless promising. It may be instructive to consider whether the positive
results obtained here are specific to the use of ASCs in conjunction with a hydrogel
of the same tissue origin. For a number of practical or biological reasons, it may be
desirable to design therapies using cells from other sources, such as chondrocytes or
mesenchymal stem cells. In these cases, it would be necessary to assess whether the
benefits observed here were retained.
A material derived from cartilage itself has also been considered for use in
cartilage repair, though its evaluation is at an earlier stage than that for the adipose-
derived hydrogel [51]. Porcine articular cartilage was decellularized by the use of
2 ECM Hydrogels for Regenerative Medicine 47
hypotonic buffer, SDS, and nuclease and subsequently solubilized by the action of
pepsin. This study extended the “classical” ECM-derived hydrogel approach by
including lyophilization of the pre-gel solution to form a powder which was sub-
sequently soluble in PBS. Gel formation by subcutaneous injection of reconstituted
material into rats was shown in principle, as was the capability of the ECM-derived
material to bind bovine serum albumin as a model protein and subsequently release
it in vivo over 2–3 weeks. This ability can presumably be attributed to the presence
of GAGs such as heparin in the material, but the exact mechanism has yet to be
conclusively deduced. A dose-dependent inflammatory response was observed for
the cartilage-derived matrix when implanted subcutaneously in rats, but the pro-
portions of activated immune cells within implant sites were low in all cases. This
study demonstrated the fundamental safety of the approach. Future work will be
required to show that endogenous and/or exogenous cells interacting with the
cartilage-derived material can proliferate and deposit appropriate matrix.
As was the case for cartilage, the first study aiming to demonstrate ECM-derived
hydrogel mediated tendon/ligament repair relied on the use of an already
well-characterized ECM material, namely porcine SIS [36]. This study used a large
animal (goat) model of anterior cruciate ligament (ACL) repair involving the
suturing of a surgically transected ligament with no tissue removal. In an effort to
augment repair, the sutured ligament was wrapped with a bilayered sheet of SIS and
fibrin sponge, and an SIS-derived pre-gel solution, produced by pepsin digestion,
was injected into the defect area. In ECM treated groups, the ACL cross-sectional
area was restored to equivalence with the sham-operated controls 12-week
post-intervention, which was several times the area of ligaments sutured without
augmentation. Furthermore, significant recovery of mechanical strength was seen
relative to suture-only tissue, though these ligaments still only withstood around
half of the load supported by the sham-operated tissues. It is unclear what pro-
portion of the benefit derived in this study is attributable to the gel, rather than the
bilayered sheet scaffold. Additionally, it will be necessary to address more chal-
lenging models before it becomes clear what promise SIS-derived hydrogels may
hold for the repair of damaged tendons and ligaments.
A subsequent study established that human tendon tissue could also be pro-
cessed to form hydrogels [35]. Decellularization using SDS and EDTA was fol-
lowed by pepsin digestion, resulting in a thermoresponsive gelling solution
composed primarily of collagen type I. Pre-gel solutions were delivered subcuta-
neously into rats to demonstrate in vivo gel formation and the host cell response.
Histological staining of explanted gels revealed that they were infiltrated by mac-
rophages and other immune cells around the periphery. Fibroblasts were also seen
throughout the hydrogels, elongated and aligned along the material fiber structure,
indicating the ability of the scaffold to direct cell behavior. Additionally, it was
48 M. J. Sawkins et al.
shown that ASCs could remain viable during in vitro culture either on the surface of
preformed gels or after encapsulation within them. This demonstrates the potential
of this material to be used in cell-seeded form in addition to the off-the-shelf,
cell-free alternative.
A third and final study sought to demonstrate that this human tendon-derived
hydrogel material could contribute to improved functional recovery after tendon
injury [49]. Defects 5 mm in length were created in the Achilles tendons of rats, and
the injection of tendon derived pre-gel solutions compared to contralateral controls
of injected saline. Histological analysis suggested that tendon-derived gel was able
to promote accelerated wound healing, with greater amounts of collagen seen in
gel-treated tendons 2-week post-injection than those injected with saline. At
4 weeks, this difference was still apparent, with a good degree of alignment seen in
the matrix deposited in gel-treated tendons; however, by 8-week post-injection,
there was no discernable difference. Functional mechanical evaluation of the
healing tendons revealed a similar trend, with gel treatment leading to significantly
increased ultimate failure load at the 4-week time point, with no differences seen at
2 or 8 weeks.
As with many other tissues, ECM-derived materials show promise for applica-
tion in tendon and ligament repair and regeneration. They appear to be well tol-
erated in vivo and may be able to accelerate defect healing. However, more
extensive studies are required to determine whether long-term tissue repair is
significantly enhanced by the presence of these hydrogels.
Intervertebral Disk
The nucleus pulposus (NP) of the intervertebral disk (IVD) exists in a naturally
gel-like state and does not therefore require the extensive processing used to gen-
erate hydrogels from other tissues. This has been exploited by Simionescu and
colleagues who have developed decellularized porcine NP as a scaffold for the
reconstitution of the IVD [55, 56]. They showed that both the mechanical properties
and relative concentrations of protein and GAG constituents of the decellularized
NP were unchanged relative to the native tissue [55]. When ASCs were seeded on
the surface of decellularized NP, they remained highly viable over the culture
period of 7 days and were able to migrate into the scaffold. This latter result
suggests that decellularized NP scaffolds could support the infiltration of appro-
priate host cell types for tissue repair after cell-free implantation.
In a second study by the same authors, decellularized NP was further studied to
elucidate the effects of the material on ASC differentiation and tissue repair in vivo
[56]. When ASCs were cultured on decellularized NP scaffolds, they expressed a
number of markers of the NP cell phenotype at significantly higher levels than on
TCPS. This study was conducted in basal growth medium in the absence of soluble
cues, indicating some inherent capacity for the material to direct appropriate stem
cell differentiation. The cells remained predominantly viable across 2 weeks,
2 ECM Hydrogels for Regenerative Medicine 49
contracted the hydrogel scaffold, and deposited a matrix containing collagen and
GAGs, leading to significant increases in scaffold mechanical properties.
Histological staining of scaffolds after subcutaneous implantation in rats indicated
that 4-week post-implantation the material was infiltrated by immune cells and
contained newly formed vasculature and newly deposited collagen. These results
established in principle that the material was well tolerated and the next step would
be to extend the in vivo analysis to include longer-term demonstrations of appro-
priate functional tissue formation.
Others
this bone ECM hydrogel has potential as a carrier and supplement for existing bone
graft materials such as DBM and allogeneic bone.
The vocal fold (VF) is another tissue, where SIS hydrogel has been evaluated. In
an in vivo proof-of-principle study, SIS gel showed promise, primarily as a carrier
for mesenchymal stem cells rather than as a therapy in its own right [26]. This
publication details the production of an SIS hydrogel by mechanical dissociation
and saline washing of the small intestine, followed by pepsin digestion to solubilize
the resulting material. As in an earlier study targeting cartilage repair (see
Section “Cartilage”), hydrogels formed from this solubilized material were lyo-
philized and milled to form a PBS soluble powder. A rabbit model of VF scar
formation and regeneration was used to evaluate injectable therapies composed of
rabbit MSCs, SIS gel, or both components together. The combined therapy led to
increased MSC retention and engraftment at the injection site, with fluorescently
labeled cells visible 3-week post-injection by in vivo imaging. Significantly, more
cells were observed histologically in defects treated with SIS at both 3- and 8-week
time points, although by this method cells were visible in all MSC-treated groups.
The combined therapy also promoted increased accumulation of sulfated GAGs and
significantly decreased collagen deposition relative to all other groups, indicating
the propensity for more effective regeneration and reduced scar formation. Finally,
these compositional changes were reflected in the assessment of functional regen-
eration of the tissue, as combined treatment led to significantly increased vibratory
capacity relative to other groups. Longer-term large animal studies are needed to
further establish the potential of combined SIS-gel and MSCs as a therapy to
prevent vocal fold scarring, but this study is nevertheless an effective proof of
principle.
Hybrid Hydrogels
A recent evolution in the use of hydrogels derived from decellularized tissue has
been the combination of these materials with other natural or synthetic hydrogels, to
provide mechanical reinforcement and/or enhanced biological activity [41, 82].
Other approaches have utilized particulate ECM materials as additives to synthetic
hydrogels [24, 61] or applied tissue derived hydrogels as coatings for solid bio-
materials [89], but these will not be considered further in this chapter.
The first application of this next-generation concept occurred in liver repair and
utilized extracts from liver cross-linked to the natural ECM components collagen
type I and hyaluronic acid (HyA) [82]. Liver tissue was decellularized using Triton
X-100 and ammonium hydroxide; non-decellularized liver was used as a com-
parator to deduce any adverse effects of the residual cellular and genetic compo-
nent. Both materials were solubilized by the action of pepsin and combined with
collagen or HyA and the cross-linker prior to gel formation. In sandwich culture of
primary human hepatocytes, the addition of liver tissue extracts was found to result
in significantly increased cell numbers. This effect was especially pronounced at
2 ECM Hydrogels for Regenerative Medicine 51
later time points in the 4-week experiment. Decellularized liver extract had a greater
effect than that from non-decellularized tissue, which can be attributed to the higher
concentrations of both structural ECM constituents and growth factors. Although it
is promising that these materials can boost primary cell proliferation, the data in this
study show no significant effect on hepatocyte functions such as albumin and urea
secretion despite the increase in cell numbers. To fully establish their potential, it
must be shown that they can aid functional recovery in damaged tissue.
The second example of this hybrid approach again utilized chemical
cross-linking to conjugate ECM-derived peptides to supplementary polymers, based
on poly(ethylene glycol) (PEG), and intended to offer enhanced mechanical prop-
erties and delayed degradation. These materials were used to reinforce a porcine
myocardial matrix generated by SDS-based decellularization [41]. Incorporation of
PEG-based polymers in the hydrogel network provided significant mechanical
reinforcement alongside structural changes which were dependent on the incor-
poration method and the polymer molecular weight. Experiments with a murine
fibroblast cell line showed that cells seeded on the surface of preformed hydrogels
could migrate into the gels and proliferate on the surfaces regardless of the presence
of the PEG component. This is despite the fact that PEG is an inert synthetic
polymer that generally does not interact with cells as it contains no binding sites for
cell surface receptors such as integrins and cadherins. It was also shown that cells
encapsulated within gels could proliferate rapidly across all conditions, with no
inhibitory effect due to the synthetic component. The fibroblast cell line used has
broad relevance to a range of tissues and the cross-linking methods could be used
with a range of tissue extracts, containing a significant collagen component and
similar functional groups. This technique could therefore potentially be used to
overcome current limitations and tailor the mechanical and degradation properties
of naturally derived hydrogel biomaterials.
Future Directions
evaluated both in vitro and in vivo and is used clinically [11]. In addition, SIS and
UBM hydrogels have been well characterized in terms of biocompatibility, bio-
chemical composition, gelation kinetics, viscoelastic properties, and gel topology.
The choice to use a tissue-specific ECM hydrogel must therefore be driven by
significantly increased efficacy or additional functionality in specific applications.
The small-scale proof-of-principle studies undertaken to date do not sufficiently
demonstrate tissue specificity in order to justify the additional time and cost
involved in developing clinical grade products for therapeutic application. In all
cases, longer-term preclinical studies are required to fully inform this
decision-making process. It is important that researchers are able to determine the
biochemical and structural factors driving cell responses and tissue formation in
these novel ECM hydrogels. In particular, it is important to consider the effect of
the tissue source (species and anatomical location where appropriate) and the
macrophage polarization response to tissue-specific ECM hydrogels as part of the
global inflammatory and immune response. These factors have been studied for
ECM materials in powder and sheet form but not yet for hydrogels, which have
altered three-dimensional architecture and subtle differences in biochemical
composition.
The knowledge derived from these further characterization studies and pre-
clinical trials may allow the development of optimized homologous and
non-homologous ECM hydrogels for a wide range of applications. These novel
materials may then provide a new hope for the treatment of defects with complex
architecture.
References
1. Agrawal V, Johnson SA, Reing J, Zhang L, Tottey S, Wang G, Hirschi KK, Braunhut S,
Gudas LJ, Badylak SF. Epimorphic regeneration approach to tissue replacement in adult
mammals. Proceedings of the National Academy of Sciences of the USA. 2010;107(8):3351–
5. https://doi.org/10.1073/pnas.0905851106.
2. Agrawal V, Tottey S, Johnson SA, Freund JM, Siu BF, Badylak SF. Recruitment of
progenitor cells by an extracellular matrix cryptic peptide in a mouse model of digit
amputation. Tissue Eng Part A. 2011;17(19–20):2435–43. https://doi.org/10.1089/ten.TEA.
2011.0036.
3. Badylak SE. The extracellular matrix as a scaffold for tissue reconstruction. Semin Cell Dev
Biol. 2002;13(5):377–83. https://doi.org/10.1016/S1084-9521(02)00094-0.
4. Badylak SF, Coffey AC, Lantz GC, Tacker WA, Geddes LA. Comparison of the resistance to
infection of intestinal submucosa arterial autografts versus polytetrafluoroethylene arterial
prostheses in a dog model. J Vasc Surg. 1994;19(3):465–72.
5. Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold material:
structure and function. Acta Biomater. 2009;5(1):1–13. https://doi.org/10.1016/j.actbio.2008.
09.013.
6. Badylak SF, Gilbert TW. Immune response to biologic scaffold materials. Sem Immunol.
2008;20(2):109–16. https://doi.org/10.1016/j.smim.2007.11.003.
2 ECM Hydrogels for Regenerative Medicine 53
25. Choi JS, Kim BS, Kim JD, Choi YC, Lee HY, Cho YW. In vitro cartilage tissue engineering
using adipose-derived extracellular matrix scaffolds seeded with adipose-derived stem cells.
Tissue Eng Part A. 2012;18(1–2):80–92. https://doi.org/10.1089/ten.tea.2011.0103.
26. Choi JW, Park JK, Chang JW, Kim DY, Kim MS, Shin YS, Kim CH. Small intestine
submucosa and mesenchymal stem cells composite gel for scarless vocal fold regeneration.
Biomaterials. 2014;35(18):4911–8. https://doi.org/10.1016/j.biomaterials.2014.03.008.
27. Cortiella J, Niles J, Cantu A, Brettler A, Pham A, Vargas G, Winston S, Wang J, Walls S,
Nichols JE. Influence of acellular natural lung matrix on murine embryonic stem cell
differentiation and tissue formation. Tissue Eng. 2010;16(8):2565–80. https://doi.org/10.
1089/ten.tea.2009.0730.
28. Crapo PM, Gilbert TW, Badylak SF. An overview of tissue and whole organ decellularization
processes. Biomaterials. 2011;32(12):3233–43. https://doi.org/10.1016/j.biomaterials.2011.
01.057.
29. Crapo PM, Tottey S, Slivka PF, Badylak SF. Effects of biologic scaffolds on human stem cells
and implications for CNS tissue engineering. Tissue Eng. 2014;20(1–2):313–23. https://doi.
org/10.1089/ten.TEA.2013.0186.
30. Crapo PM, Wang Y. Small intestinal submucosa gel as a potential scaffolding material for
cardiac tissue engineering. Acta Biomater. 2010;6(6):2091–6. https://doi.org/10.1016/j.actbio.
2009.10.048.
31. D’Amore A, Stella JA, Wagner WR, Sacks MS. Characterization of the complete fiber
network topology of planar fibrous tissues and scaffolds. Biomaterials. 2010;31(20):5345–54.
https://doi.org/10.1016/j.biomaterials.2010.03.052.
32. DeQuach JA, Lin JE, Cam C, Hu D, Salvatore MA, Sheikh F, Christman KL. Injectable
skeletal muscle matrix hydrogel promotes neovascularization and muscle cell infiltration in a
hindlimb ischemia model. Eur Cells Mater. 2012;23:400–12.
33. Drake MP, Davison PF, Bump S, Schmitt FO. Action of proteolytic enzymes on
tropocollagen and insoluble collagen. Biochemistry. 1966;5(1):301–12.
34. Drury JL, Mooney DJ. Hydrogels for tissue engineering: scaffold design variables and
applications. Biomaterials. 2003;24(24):4337–51.
35. Farnebo SJ, Woon CY, Schmitt T, Joubert LM, Kim M, Pham H, Chang J. Design and
characterization of an injectable tendon hydrogel: a scaffold for guided tissue regeneration in
the musculoskeletal system. Tissue Eng. 2013. https://doi.org/10.1089/ten.TEA.2013.0207.
36. Fisher MB, Liang R, Jung HJ, Kim KE, Zamarra G, Almarza AJ, McMahon PJ, Woo SLY.
Potential of healing a transected anterior cruciate ligament with genetically modified
extracellular matrix bioscaffolds in a goat model. Knee Surg Sports Traumatol Arthrosc.
2012;20(7):1357–65. https://doi.org/10.1007/s00167-011-1800-x.
37. Francis D, Abberton K, Thompson E, Daniell M. Myogel supports the ex vivo amplification of
corneal epithelial cells. Exp Eye Res. 2009;88(3):339–46. https://doi.org/10.1016/j.exer.2008.
06.016.
38. Freytes DO, Martin J, Velankar SS, Lee AS, Badylak SF. Preparation and rheological
characterization of a gel form of the porcine urinary bladder matrix. Biomaterials. 2008;29
(11):1630–7. https://doi.org/10.1016/j.biomaterials.2007.12.014.
39. Ganz T. Defensins: antimicrobial peptides of innate immunity. Nat Rev Immunol. 2003;3
(9):710–20. https://doi.org/10.1038/nri1180.
40. Gilbert TW, Stolz DB, Biancaniello F, Simmons-Byrd A, Badylak SF. Production and
characterization of ECM powder: implications for tissue engineering applications.
Biomaterials. 2005;26(12):1431–5.
41. Grover GN, Rao N, Christman KL. Myocardial matrix-polyethylene glycol hybrid hydrogels
for tissue engineering. Nanotechnology. 2014;25(1). https://doi.org/10.1088/0957-4484/25/1/
014011.
42. Hulmes DJS. Collagen diversity, synthesis, and assembly. In: Fratzl P, editors. Collagen:
structure and mechanics. Berlin: Springer; 2008.
2 ECM Hydrogels for Regenerative Medicine 55
43. Jernigan TW, Croce MA, Cagiannos C, Shell DH, Handorf CR, Fabian TC. Small intestinal
submucosa for vascular reconstruction in the presence of gastrointestinal contamination. Ann
Surg. 2004;239(5):733–8.
44. Johnson TD, Dequach JA, Gaetani R, Ungerleider J, Elhag D, Nigam V, Behfar A,
Christman KL. Human versus porcine tissue sourcing for an injectable myocardial matrix
hydrogel. Biomater Sci. 2014;2(5):735–44. https://doi.org/10.1039/c3bm60283d.
45. Johnson TD, Lin SY, Christman KL. Tailoring material properties of a nanofibrous
extracellular matrix derived hydrogel. Nanotechnology. 2011;22(49):494015. https://doi.org/
10.1088/0957-4484/22/49/494015.
46. Kadler KE, Hill A, Canty-Laird EG. Collagen fibrillogenesis: fibronectin, integrins, and minor
collagens as organizers and nucleators. Curr Opin Cell Biol. 2008;20(5):495–501. https://doi.
org/10.1016/j.ceb.2008.06.008.
47. Kadler KE, Holmes DF, Trotter JA, Chapman JA. Collagen fibril formation. Biochem J.
1996;316(Pt 1):1–11.
48. Keane TJ, Londono R, Turner NJ, Badylak SF. Consequences of ineffective decellularization
of biologic scaffolds on the host response. Biomaterials. 2012;33(6):1771–81. https://doi.org/
10.1016/j.biomaterials.2011.10.054.
49. Kim MY, Farnebo S, Woon CYL, Schmitt T, Pham H, Chang J. Augmentation of tendon
healing with an injectable tendon hydrogel in a rat achilles tendon model. Plast Reconstr Surg.
2014;133(5):645e–53e. https://doi.org/10.1097/PRS.0000000000000106.
50. Kopecek J. Hydrogel biomaterials: a smart future? Biomaterials. 2007;28(34):5185–92.
https://doi.org/10.1016/j.biomaterials.2007.07.044.
51. Kwon JS, Yoon SM, Shim SW, Park JH, Min KJ, Oh HJ, Kim JH, Kim YJ, Yoon JJ,
Choi BH, Kim MS. Injectable extracellular matrix hydrogel developed using porcine articular
cartilage. Int J Pharm. 2013;454(1):183–91. https://doi.org/10.1016/j.ijpharm.2013.06.023.
52. Lee JS, Shin J, Park HM, Kim YG, Kim BG, Oh JW, Cho SW. Liver extracellular matrix
providing dual functions of two-dimensional substrate coating and three-dimensional
injectable hydrogel platform for liver tissue engineering. Biomacromol. 2013;15(1):206–18.
https://doi.org/10.1021/bm4015039.
53. Lee KY, Mooney DJ. Hydrogels for tissue engineering. Chem Rev. 2001;101(7):1869–79.
54. Medberry CJ, Crapo PM, Siu BF, Carruthers CA, Wolf MT, Nagarkar SP, Agrawal V,
Jones KE, Kelly J, Johnson SA, Velankar SS, Watkins SC, Modo M, Badylak SF. Hydrogels
derived from central nervous system extracellular matrix. Biomaterials. 2013;34(4):1033–40.
https://doi.org/10.1016/j.biomaterials.2012.10.062.
55. Mercuri JJ, Gill SS, Simionescu DT. Novel tissue-derived biomimetic scaffold for
regenerating the human nucleus pulposus. J Biomed Mater Res. 2011;96A(2):422–35.
https://doi.org/10.1002/jbm.a.33001.
56. Mercuri JJ, Patnaik S, Dion G, Gill SS, Liao J, Simionescu DT. Regenerative potential of
decellularized porcine nucleus pulposus hydrogel scaffolds: stem cell differentiation, matrix
remodeling, and biocompatibility studies. Tissue Eng. 2013;19(7–8):952–66. https://doi.org/
10.1089/ten.TEA.2012.0088.
57. Miller EJ. Structural studies on cartilage collagen employing limited cleavage and
solubilization with pepsin. Biochemistry. 1972;11(26):4903–9.
58. Moore AJ, Beazley WD, Bibby MC, Devine DA. Antimicrobial activity of cecropins.
J Antimicrob Chemother. 1996;37(6):1077–89.
59. Nelson CM, Bissell MJ. Of extracellular matrix, scaffolds, and signaling: tissue architecture
regulates development, homeostasis, and cancer. Annu Rev Cell Dev Biol. 2006;22:287–309.
https://doi.org/10.1146/annurev.cellbio.22.010305.104315.
60. Nerem R. The challenge of imitating nature. Principles of tissue engineering. 3rd ed.
Burlington: Elsevier; 2007.
61. Ni P, Ding Q, Fan M, Liao J, Qian Z, Luo J, Li X, Luo F, Yang Z, Wei Y. Injectable
thermosensitive PEG-PCL-PEG hydrogel/acellular bone matrix composite for bone regen-
eration in cranial defects. Biomaterials. 2014;35(1):236–48. https://doi.org/10.1016/j.
biomaterials.2013.10.016.
56 M. J. Sawkins et al.
62. Parkinson J, Kadler KE, Brass A. Simple physical model of collagen fibrillogenesis based on
diffusion limited aggregation. J Mol Biol. 1995;247(4):823–31. https://doi.org/10.1006/jmbi.
1994.0182.
63. Petersen TH, Calle EA, Zhao LP, Lee EJ, Gui LQ, Raredon MB, Gavrilov K, Yi T,
Zhuang ZW, Breuer C, Herzog E, Niklason LE. Tissue-engineered lungs for in vivo
implantation. Science. 2010;329(5991):538–41. https://doi.org/10.1126/science.1189345.
64. Pilipchuk SP, Vaicik MK, Larson JC, Gazyakan E, Cheng M-H, Brey EM. Influence of
crosslinking on the stiffness and degradation of dermis-derived hydrogels. J Biomed Mater
Res. 2013;101(10):2883–95. https://doi.org/10.1002/jbm.a.34602.
65. Poon CJ, Pereira E. Cotta MV, Sinha S, Palmer JA, Woods AA, Morrison WA,
Abberton KM. Preparation of an adipogenic hydrogel from subcutaneous adipose tissue.
Acta Biomaterialia. 2013;9(3):5609–20. https://doi.org/10.1016/j.actbio.2012.11.003.
66. Ravi S, Caves JM, Martinez AW, Xiao J, Wen J, Haller CA, Davis ME, Chaikof EL. Effect of
bone marrow-derived extracellular matrix on cardiac function after ischemic injury.
Biomaterials. 2012;33(31):7736–45. https://doi.org/10.1016/j.biomaterials.2012.07.010.
67. Reing JE, Brown BN, Daly KA, Freund JM, Gilbert TW, Hsiong SX, Huber A, Kullas KE,
Tottey S, Wolf MT, Badylak SF. The effects of processing methods upon mechanical and
biologic properties of porcine dermal extracellular matrix scaffolds. Biomaterials. 2010;31
(33):8626–33. https://doi.org/10.1016/j.biomaterials.2010.07.083.
68. Reing JE, Zhang L, Myers-Irvin J, Cordero KE, Freytes DO, Heber-Katz E, Bedelbaeva K,
McIntosh D, Dewilde A, Braunhut SJ, Badylak SF. Degradation products of extracellular
matrix affect cell migration and proliferation. Tissue Eng. 2009;15(3):605–14. https://doi.org/
10.1089/ten.tea.2007.0425.
69. Rosamond W, Flegal K, Friday G, Furie K, Go A, Greenlund K, Haase N, Ho M, Howard V,
Kissela B, Kittner S, Lloyd-Jones D, McDermott M, Meigs J, Moy C, Nichol G, O’Donnell
CJ, Roger V, Rumsfeld J, Sorlie P, Steinberger J, Thom T, Wasserthiel-Smoller S, Hong Y,
American Heart Association Statistics C, Stroke Statistics S. Heart disease and stroke statistics
—2007 update: a report from the American Heart Association Statistics Committee and
Stroke Statistics Subcommittee. Circulation. 2007;115(5):e69–e171. https://doi.org/10.1161/
circulationaha.106.179918.
70. Rubin AL, Drake MP, Davison PF, Pfahl D, Speakman PT, Schmitt FO. Effects of pepsin
treatment on the interaction properties of tropocollagen macromolecules. Biochemistry.
1965;4(2):181–90. https://doi.org/10.1021/bi00878a001.
71. Russell A, Bertram T. Moving into the clinic. Principles of tissue engineering. 3rd ed.
Burlington: Elsevier; 2007.
72. Sampath TK, Reddi AH. Importance of geometry of the extracellular matrix in endochondral
bone differentiation. J Cell Biol. 1984;98(6):2192–7.
73. Sarikaya A, Record R, Wu CC, Tullius B, Badylak S, Ladisch M. Antimicrobial activity
associated with extracellular matrices. Tissue Eng. 2002;8(1):63–71. https://doi.org/10.1089/
107632702753503063.
74. Sawkins MJ, Bowen W, Dhadda P, Markides H, Sidney LE, Taylor AJ, Rose FRAJ,
Badylak SF, Shakesheff KM, White LJ. Hydrogels derived from demineralized and
decellularized bone extracellular matrix. Acta Biomater. 2013;9(8):7865–73. https://doi.org/
10.1016/j.actbio.2013.04.029.
75. Seif-Naraghi SB, Horn D, Schup-Magoffin PJ, Christman KL. Injectable extracellular matrix
derived hydrogel provides a platform for enhanced retention and delivery of a heparin-binding
growth factor. Acta Biomater. 2012;8(10):3695–703. https://doi.org/10.1016/j.actbio.2012.
06.030.
76. Seif-Naraghi SB, Salvatore MA, Schup-Magoffin PJ, Hu DP, Christman KL. Design and
characterization of an injectable pericardial matrix gel: a potentially autologous scaffold for
cardiac tissue engineering. Tissue Eng. 2010;16(6):2017–27. https://doi.org/10.1089/ten.
TEA.2009.0768.
2 ECM Hydrogels for Regenerative Medicine 57
77. Seif-Naraghi SB, Singelyn JM, Salvatore MA, Osborn KG, Wang JJ, Sampat U, Kwan OL,
Strachan GM, Wong J, Schup-Magoffin PJ, Braden RL, Bartels K, DeQuach JA, Preul M,
Kinsey AM, DeMaria AN, Dib N, Christman KL. Safety and efficacy of an injectable
extracellular matrix hydrogel for treating myocardial infarction. Sci Transl Med. 2013;5(173).
78. Sellaro TL, Ranade A, Faulk DM, McCabe GP, Dorko K, Badylak SF, Strom SC.
Maintenance of human hepatocyte function in vitro by liver-derived extracellular matrix gels.
Tissue Eng. 2010;16:1075–82. https://doi.org/10.1089/ten.TEA.2008.0587.
79. Shell DH, Croce MA, Cagiannos C, Jernigan TW, Edwards N, Fabian TC. Comparison of
small-intestinal submucosa and expanded polytetrafluoroethylene as a vascular conduit in the
presence of gram-positive contamination. Ann Surg. 2005;241(6):995–1001.
80. Singelyn JM, DeQuach JA, Seif-Naraghi SB, Littlefield RB, Schup-Magoffin PJ,
Christman KL. Naturally derived myocardial matrix as an injectable scaffold for cardiac tissue
engineering. Biomaterials. 2009;30(29):5409–16. https://doi.org/10.1016/j.biomaterials.2009.
06.045.
81. Singelyn JM, Sundaramurthy P, Johnson TD, Schup-Magoffin PJ, Hu DP, Faulk DM,
Wang J, Mayle KM, Bartels K, Salvatore M, Kinsey AM, DeMaria AN, Dib N,
Christman KL. Catheter-deliverable hydrogel derived from decellularized ventricular
extracellular matrix increases endogenous cardiomyocytes and preserves cardiac function
post-myocardial infarction. J Am Coll Cardiol. 2012;59(8):751–63. https://doi.org/10.1016/j.
jacc.2011.10.888.
82. Skardal A, Smith L, Bharadwaj S, Atala A, Soker S, Zhang Y. Tissue specific synthetic ECM
hydrogels for 3-D in vitro maintenance of hepatocyte function. Biomaterials. 2012;33(18):
4565–75. https://doi.org/10.1016/j.biomaterials.2012.03.034.
83. Streuli C. Extracellular matrix remodelling and cellular differentiation. Curr Opin Cell Biol.
1999;11(5):634–40.
84. Uriel S, Huang J-J, Moya ML, Francis ME, Wang R, S-y Chang, Cheng M-H, Brey EM. The
role of adipose protein derived hydrogels in adipogenesis. Biomaterials. 2008;29(27):3712–9.
https://doi.org/10.1016/j.biomaterials.2008.05.028.
85. Uriel S, Labay E, Francis-Sedlak M, Moya ML, Weichselbaum RR, Ervin N, Cankova Z,
Brey EM. Extraction and assembly of tissue-derived gels for cell culture and tissue
engineering. Tissue Eng. 2009;15(3):309–21. https://doi.org/10.1089/ten.tec.2008.0309.
86. Uygun BE, Soto-Gutierrez A, Yagi H, Izamis M-L, Guzzardi MA, Shulman C, Milwid J,
Kobayashi N, Tilles A, Berthiaume F, Hertl M, Nahmias Y, Yarmush ML, Uygun K. Organ
reengineering through development of a transplantable recellularized liver graft using
decellularized liver matrix. Nat Med. 2010;16(7):814–20. https://doi.org/10.1038/nm.2170.
87. Voytik-Harbin SL, Brightman AO. Small intestinal submucosa: a tissue derived extracellular
matrix that promotes tissue-specific growth and differentiation of cells in vitro. Tissue Eng.
1998;4:157–74.
88. Voytik-Harbin SL, Brightman AO, Kraine MR, Waisner B, Badylak SF. Identification of
extractable growth factors from small intestinal submucosa. J Cell Biochem. 1997;67(4):
478–91.
89. Wolf MT, Carruthers CA, Dearth CL, Crapo PM, Huber A, Burnsed OA, Londono R,
Johnson SA, Daly KA, Stahl EC, Freund JM, Medberry CJ, Carey LE, Nieponice A,
Amoroso NJ, Badylak SF. Polypropylene surgical mesh coated with extracellular matrix
mitigates the host foreign body response. J Biomed Mater Res. 2014;102(1):234–46. https://
doi.org/10.1002/jbm.a.34671.
90. Wolf MT, Daly KA, Brennan-Pierce EP, Johnson SA, Carruthers CA, D’Amore A,
Nagarkar SP, Velankar SS, Badylak SF. A hydrogel derived from decellularized dermal
extracellular matrix. Biomaterials. 2012;33(29):7028–38. https://doi.org/10.1016/j.biomaterials.
2012.06.051.
58 M. J. Sawkins et al.
91. Wolf MT, Daly KA, Reing JE, Badylak SF. Biologic scaffold composed of skeletal muscle
extracellular matrix. Biomaterials. 2012;33(10):2916–25. https://doi.org/10.1016/j.biomaterials.
2011.12.055.
92. Young DA, Ibrahim DO, Hu D, Christman KL. Injectable hydrogel scaffold from
decellularized human lipoaspirate. Acta Biomater. 2011;7(3):1040–9. https://doi.org/10.
1016/j.actbio.2010.09.035.
93. Zhang L, Zhang F, Weng Z, Brown BN, Yan H, Ma XM, Vosler PS, Badylak SF, Dixon CE,
Cui XT, Chen J. Effect of an inductive hydrogel composed of urinary bladder matrix upon
functional recovery following traumatic brain injury. Tissue Eng. 2013;19(17–18):1909–18.
https://doi.org/10.1089/ten.tea.2012.0622.
Chapter 3
Biologically Relevant Laminins
in Regenerative Medicine
A. Domogatskaya (&)
Division of Transplantation Surgery, Department of Clinical Science,
Intervention and Technology, Karolinska Institute, 14152 Huddinge, Sweden
e-mail: [email protected]
S. Rodin
Department of Medical Biochemistry and Biophysics, Karolinska Institute,
Scheeles vagen 2, B1, Division of Matrix Biology, 17177 Stockholm, Sweden
e-mail: [email protected]
Keywords Laminin Embryonic stem cell Stem cell Extracellular matrix
Regenerative medicine Neuron Pancreatic islets Insulin-producing beta cell
Stem cell technologies Proliferation Survival Apoptosis Differentiation
Adhesion Safety Cell culture Signaling Integrin
Introduction
embryonic stem cells, hematopoietic stem cells, peripheral neurons, Schwann cells,
insulin-producing pancreatic b-cells, vascular endothelial cells, keratinocytes,
muscle cells [15]. Laminins can mediate behavior of associated cells in many
different ways, such as: survival, adhesion, migration or stable anchorage, prolif-
eration or quiescence, phenotype stability or dedifferentiation, undifferentiated
status in stem cells, or differentiation. Moreover, laminins are essential for mor-
phogenesis during embryonic/prenatal development and may enable selective
restriction for cell permeability between compartments and cell-guiding function in
regeneration and development. There is evidence that niche-specific laminins act in
concert with specific growth factors and cell–cell contacts to affect cell behavior.
In mammals, at least 16 laminin isoforms are known up to date, and every
isoform has a specific tissue and developmental stage-specific function [15, 16].
Laminins are large cross-shaped heterotrimeric proteins, comprising one a, one b,
and one c chain. Molecular weight of a laminin molecule varies from 400 to
1000 kDa. Laminins convey signals from surrounding extracellular matrix to the
adherent cells via binding to laminin-specific cell receptors, e.g., integrins, and are
essential part of natural mechanotransduction system [25].
Laminins first appear early in evolution, already in hydra, and are essential for
ability of hydra to regenerate [70, 72, 97]. In primitive species, such as
Caenorhabditis elegans and Drosophila melanogaster, every laminin isoform is
essential; lack of any laminin chain results in early lethality [28, 32, 46, 85, 92]. In
zebrafish, mouse and human laminin chains mutations often result in either
embryonic/postnatal lethality or diseases. Notably, knockout phenotypes are totally
different for any of a chains, or b chains, or c chains [15, 16, 51]. This indicates that
biological role of every laminin isoforms is unique. It is shown that certain laminins
isoforms, despite visible similarity in structure and amino acid sequence, exert
distinct, and sometimes even opposite influences on certain cell types.
In early mammalian embryos, laminins are the first extracellular matrix molecules
to appear: already at 2–4-cell stage, b- and c-laminin chains are detected, and at 8-cell
stage the whole laminin trimer appears in extracellular space [10, 17, 38]. At the stage
of blastula, at least two laminin isoforms appear: a5 laminins are expressed between
pluripotent cells of inner cell mass and, probably, supporting their pluripotency and
proliferation, while a1 laminins, provided by trophectoderm, cause polarization of
pluripotent cells and subsequent differentiation into ectoderm, endoderm, and
mesoderm [34, 39–41]. At the later stages of embryonic development, tissue-specific
and developmental stage-specific laminins drive morphogenesis; lack of specific
laminins often cannot be compensated by similar laminin isoforms and results in
diseases or sometimes lethality (reviewed in [15, 51]). For example, in kidney
development all the laminin a chains are expressed in a niche-specific manner; in
development of glomerular basement membrane (GBM) early LM-111 is replaced by
intermediate LM-511 and finally by LM-521. Inability to transit from LM-511 to
LM-521 results in renal failures (Pierson syndrome) [50, 76].
Use of niche-specific laminins for expanding functional cells in vitro is an
emerging trend in regenerative medicine-related research. Long-term (up to 150
doublings) and comprehensive (including in-depth analysis of random mutations
62 A. Domogatskaya and S. Rodin
and chromosomal abnormalities) studies were performed for mouse and human
embryonic stem cells [14, 67, 68]; short-term studies demonstrated obvious benefits
of biologically relevant laminins for in vitro cultures of adult stem cells and dif-
ferentiated cells, such as neurons, insulin-producing b-cells, and even whole pan-
creatic insulin-producing islets [53, 64, 82, 86]. However, for many mammalian cell
types that could be used in regenerative medicine, knowledge of biologically rel-
evant laminins in vivo and in vitro is scarce. Therefore, we shall discuss not only
existing laminin-based technologies, but rather the approaches, success criteria, and
possible pitfalls in implementing concept of biologically relevant laminins use into
generation of new cell culture systems. In conclusion, we shall discuss the existing
challenges in laminin research, regarding regenerative medicine needs, and
potential possibilities to exploit more sophisticated functions of laminins in tech-
nologies that are yet to be invented.
Human embryonic stem cells, when cultured in vitro for long passages, may
undergo not only random mutagenesis, but also non-random gains of chromosomes
or large chromosome fragments, similar to what happens during malignant trans-
formation [3, 52]. Though such cells may have typical phenotypic features and high
proliferation rate, they can be dangerous in transplantation.
Prolonged cell culture in vitro, unlike organ transplantation, requires use of
culture media, specific supplements, growth factors, and other molecular sub-
stances, often undefined chemical compositions. One should consider risks of
introducing pathogens to the cultured cells. For example, bacterial collagenase
preparations used for clinical derivation of insulin-producing pancreatic islets aimed
for transplantation were subject to bovine spongiform encephalopathy contamina-
tion risk; therefore, measures were taken to exclude it [80]. Human embryonic stem
cells, when cultured in the presence of animal-derived substances, have been shown
to acquire immunogenicity [47]. Therefore, FDA and EU regulatory agencies
suggest that all the compounds in contact with the cells aimed for medical use
would better be chemically defined and xeno-free [26, 27, 84].
Human laminins are chemically defined, xeno-free, non-immunogenic, and
biologically relevant molecules [67, 68]. Specific laminins, as essential part of
natural niches for majority of mammalian cell types, such as insulin-producing b-
cells, vascular endothelial cells, or embryonic stem cells, can contribute to gener-
ation of safe and robust in vitro culture systems that would imitate the natural cell
niches and maintain healthy phenotypes.
Cell behavior is guided by signals that cell receives from its environment.
Influenced by environmental cues, a cell may survive or undergo apoptosis, migrate
in scattered or directed fashion, form a stable anchorage, proliferate or remain
quiescent, maintain its phenotype, differentiate, dedifferentiate, or lose phenotype.
Understanding the nature of cues received by cell from its microenvironment is
essential to design a solution to safe and robust regenerative medicine protocols.
Mimicking natural niches by providing a set of appropriate molecular cues to
mammalian cells in vitro could allow to expand them in healthy and functional way
[67]. On the contrary, unnatural molecular cues may provoke loss of function and
phenotype and even genetic mutations [7].
Usually, a cell receives three types of cues from its microenvironment:
(1) growth factors, hormones, and other soluble molecules that activate cell
receptors by binding them, (2) cell–cell contacts, being essential for cells like
embryonic stem cells or insulin-producing pancreatic b-cells, and (3) niche-specific
extracellular matrix. The latter modulates cell signaling not merely by binding to
the cell receptors, but also by mechanotransduction [25].
64 A. Domogatskaya and S. Rodin
In this chapter, we shall briefly review common knowledge about the laminin
family: (1) molecular structure of laminins, (2) functional interactions with other
molecules, such as cell receptors and other extracellular matrix molecules, (3) effect
3 Biologically Relevant Laminins in Regenerative Medicine 65
Laminins are large, heterotrimeric molecules that comprise one a, one b, and one c
chain. Size of laminin trimer varies from 400 to 1000 kDa. Laminin trimers have
either cross-like, or Y-like, or rodlike shape (see Fig. 3.1). Short arms of laminins
(N-terminal parts of a, b, and c chains) can bind other laminins short arms and other
extracellular matrix proteins. Long arm consists of (1) a, b, and c chains inter-
twined together into a trimeric spiral via coil-coiled domains and (2) C-terminal
fragment of a chain, comprising of five globular LG domains (LG1-LG5) that
interact with cell receptors.
Five a (a1-a5), four b (b1-b4), and three c (c1-c3) chains are known in
mammals; a5, b1, b2, and c1 are expressed ubiquitously, and other chains are more
tissue-restricted [4]. Up to date, 16 trimeric laminin isoforms present in vivo have
been characterized in mammals; however, theoretically 60 combinations are pos-
sible. Modern nomenclature describes laminin isoforms according to their chain
composition [4]. For example, ubiquitous laminin-511 (LM-511) consists of a5, b1,
and c1 chains.
It is shown by Macdonald et al. [43] that not all the combinations of a, b, and c
chains can assemble into trimeric forms. Sixteen laminin trimers have been iden-
tified in vivo and two more have been predicted by Macdonald et al. [43].
Fig. 3.1 Schematic representation of typical laminin trimer forms. a Cross-shaped. b Y-shaped.
c Rod-shaped
66 A. Domogatskaya and S. Rodin
Associations of c1 with b1 or b2 form trimers with all the five a chains. The a1, a2,
a3B, a5 trimers are cross-shaped, while the a3A, a4 trimers are Y-shaped or
rod-shaped.
Extensive body of data has been generated on single laminin chain expression
and distribution in various mammalian tissues, and yet very little is known about
specific trimers localizations in vivo. However, information on single laminin
chains expression patterns and knockout animal models can give us some insights
into in vivo function of certain laminin trimers.
a1 laminins. Vast majority of laminin studies in twentieth century involved
either LM-111 (called “laminin”) or Matrigel, a tumor-derived gel comprising
LM-111. Lack of a1 results in early embryonic lethality, presumably, due to inner
cell mass polarization failure [49, 71]. However, expression pattern of LM-111 in
mature mammalian organisms is restricted (compared to ubiquitous LM-511/521)
and biological function clearly known for embryonic events, such as blastocyst
inner cell mass polarization [39, 41, 49] and early kidney glomerular basement
membrane development [50, 76].
a2 laminins play important roles in differentiation and maintenance of muscular
and neural cells; mutations in LAMA2 (gene encoding a2 laminin) result in
muscular dystrophy and neural system disorders [8, 31, 78, 90]. a2 laminins are the
major laminins in muscle [61, 69] and play an important role in smooth muscle
myogenesis [66]. a2 laminins are an important part of neural system and are
essential for thymocytes maturation [29, 44]. a2 laminins are expressed in testis
[37] and deficiency of those results in infertility [23].
a3 laminins are specific for epithelial basement membranes, for example
underlying skin epidermis and intestinal epithelia. Two splice variants of LAMA3
gene exist: a3A, the short chain, and a3B, the long chain.
a4 laminins, Y-shaped, are supposed to be “not very important” since no lethal
or early severe phenotype is observed. However, mutations in LAMA4 cause
microcirculation development disorders [81], cardiomyopathy [36, 87], chronic
kidney disorders [1], neurodisorders [62, 86], and impairment of leukocyte
recruitment [33]. It appears that a4 laminins attenuate fine guidance of cells, such as
tip cells Dll4/Notch signaling in angiogenesis [77], or high-precision spatial
matching of active zones of motor axon terminus with junctional folds on muscle
end plate within developing neuromuscular junctions [62].
a5 laminins, cross-shaped molecules capable of self-polymerization, are ubiq-
uitously expressed. They first appear in earliest embryonic development stages; for
example, they are expressed by blastocyst inner cell mass to provide themselves
vital cues; a5 laminins are essential part of vascular basement membranes, skin,
hair follicle niches, and many others. Detailed review of knockout phenotypes and
list of references are available in [15, 16]. Lack of a5 laminins causes late-stage
embryonic lethality in mice [48] and cannot be compensated by other isoforms.
b1 laminins are ubiquitously expressed. They are essential for organism
maintenance from the earliest stages of embryonic development to complete mat-
uration of mammalian organism. b1 laminins are present in almost every organ
3 Biologically Relevant Laminins in Regenerative Medicine 67
Macdonald et al. predicted that two additional isoforms LM-312 and LM-422
may exist since coil-coiled domains of the named chains can associate as trimers
[43].
binding the mammary epithelial receptors on the basal side of the cell attracts the
prolactin receptor to the same side, as it occurs in vivo. If prolactin is provided from
the same basal side, it meets the prolactin receptor and signaling pathway occurs
[91].
Co-signaling in vascular endothelial cells. Co-signaling from LM-411 and
VEGF to vascular endothelial tip cell allows to switch on the Notch/Dll pathway,
thus restricting the occurrence of vessel branching [77]. In the absence of LM-411,
the mechanism fails, and pathological vessel branching occurs and results in
pathologies.
Clustering calcium channels in motor nerve terminals. Laminin b2 binds and
clusters together voltage-gated calcium channels within synaptic cleft, which is
essential for neurotransmitter release from motor nerve terminals [54].
Neuromuscular junctions in mice lacking laminin b2 suffer abnormalities: synaptic
cleft being blocked by invading Schwann cell processes and reduced number of
active zones in nerve terminals [55, 60]. Apparently, b1-laminins present in the
synaptic cleft cannot compensate for the missing b2 isoforms.
Self-assembly and interaction with other laminin isoforms. Laminins are cap-
able of self-assembly via N-terminal short arms. N-terminal parts of non-truncated
a, b, and c chains can interact with each other. Cross-shaped laminins, like
LM-111, can form 3D gels, for which they require calcium ions [94]. It is important
to note that Y-shaped laminins, like LM-311/321 or 411/421, as such cannot form
3D gels without help of cross-shaped laminins or basement membranes molecules.
Possible combinations of interaction between N-terminal short arms of a1, a2, a5,
b1, b2, b3, c1, and c3 laminin chains and respective Kd values are described in
[57].
Other extracellular matrix molecules. Laminins constitute basement mem-
branes together with collagen IV. Collagen IV and laminins can self-assemble;
however, those two intertwining networks are not connected directly, but via
smaller connector molecules, such as nidogens [95, 96].
Mechanotransduction
Biological function of laminins relies on ability to interact with cell receptors and
extracellular matrix scaffold simultaneously. It is shown that cell function, such as
differentiation or activation, depends on surrounding extracellular scaffold
mechanical properties (stiffness or elasticity modulus) [13, 18–20]. Laminins are
capable of outside-in signaling.
Important implications for biotechnology are: (1) laminin peptides or single
domains, such as whole or partial LG domains, often lack an ability to couple
70 A. Domogatskaya and S. Rodin
Certain laminin isoforms are highly homological and are expressed in same tissues.
One may assume that they would exert similar influence on the cells, which are in
contact with them. Indeed, it sometimes such as laminins impose similar effects on
the cells; for example, LM-511 and LM-521 support embryonic stem cells
self-renewal in vitro [67, 68]. However, sometimes closely related laminin isoforms
exert antagonistic impact on cells.
Example 1: b1- versus b2-laminins within neuromuscular junction. Laminin
b1 isoforms, such as LM-211, 411, and 511, are enveloping surface of muscular
fibers and motor nerves that innervate them, in exception for neuromuscular
junction: the very spot where signal is transmitted from nerve terminal to the
muscle. The neuromuscular junction contains b2-analogues of the same laminins:
LM-221, 421, and 521. In LAMB2-/knockout mice, the b1 isoforms LM-211, 411,
and 511 appear within synaptic cleft, in order to compensate for the absence of
b2-laminins. The Schwann cells are repelled by b2-laminin LM-521 and thus
prevented from spreading processes into synaptic cleft in healthy mice.
3 Biologically Relevant Laminins in Regenerative Medicine 71
On contrary, the Schwann cells are attracted by b1-laminins to invade and block the
synaptic cleft in LAMB2-/knockout mice that results in severe neural system dis-
orders [55, 60].
Example 2: Processed versus unprocessed 3 chain in LM-332 in cell adhe-
sion. LM-332 affects epithelial cells behavior differently depending on whether a3
chain is processed or not. Unprocessed a3 chain binds integrin a3b1 and facilitates
cell migration, wherein cells form temporary contacts with the LM-332 surface.
However, processed a3 LM-332 binds integrin a6b4 and forms hemidesmosomes
that enable stable anchorage of the cells.
In this part, we shall present several examples where biologically relevant laminins
are used for establishing functional cell cultures in vitro.
Mouse embryonic stem cells that were cultured on a1-, a3-, a4- and a5-lami-
nins, respectively, in the absence of differentiation inhibitors underwent four dif-
ferent scenarios [14]. Cells, cultured on LM-111, differentiated within 2 weeks, and
proliferation abruptly reduced (which is consistent with natural role of LM-111
[40]). Cells cultured on LM-411 suffered lack of adhesion contacts, detached and
died. Cells cultured on LM-332 and LM-511 proliferated for over 150 doublings
with approximately same rate, while continuously expressing the pluripotency
markers: Oct-4, Sox2, and Nanog. However, the functional test—ability to form
three germ layers and give rise to whole functional organism—revealed the drastic
difference between LM-511 and LM-332. As latter gave rise to very weakly chi-
meric and/or sick chimeric animals which failed to undergo germ line transfer, the
former (LM-511) gave rise to healthy animals with strong chimerism [14] that in
turn gave rise to germ line transfer, healthy animals (Domogatskaya, Rodin,
Tryggvason: unpublished manuscript).
Due to unlimited proliferative capacity and ability to differentiate into practically
all adult cell types, human embryonic stem (ES) cells may be a valuable source of
cells for regenerative medicine. Nevertheless, only a few clinical trials have been
approved for human ES therapies. One of the major reasons of that was lack of
xeno-free (animal substance-free) and chemically defined environments that sup-
port self-renewal of human pluripotent stem (PS) cells (human ES and induced
pluripotent stem cells). Using a5-laminins, we have been able to develop such
human cell culture systems. Indeed, LM-511 and especially LM-521 allow robust
proliferation of human PS cells for more than 6 months in culture. The number of
cultured cells has been multiplied more than 1020 times, which is enough to develop
enough cells to treat hundreds of people. During the culturing, the cells express
stable levels of markers of pluripotency Oct-4, Nanog, Sox2, SSEA-4, etc. After six
months in culture, human PS cells can be differentiated into all three germ lineages
of the human embryo both in in vivo and in vitro assays. In-depth genotyping assay
has revealed that the cells cultured on LM-521 acquire little number of genetic
abnormalities and can be used in regenerative medicine. Importantly, LM-521
allows passaging of human PS cells in single-cell suspensions, which facilitates
automation of the culturing.
Individualized human PS cells die from anoikis that is a form of apoptosis
caused by unnatural milieu. That feature of human PS cells complicates manipu-
lations with their genomes, which can be used in regenerative medicine, e.g., for
correction of hereditary monogenic diseases. Since relevant integrin-mediated ex-
tracellular matrix signaling and cadherin-mediated cell–cell signaling prevent
anoikis, we sought to identify molecular cues that are sufficient to mimic natural
environment of human PS cells. E-cadherin, which is abundantly expressed on
human ES cell membranes, and LM-521 taken at 1:9 w/w ratio and used as cell
culture substratum have been able to prevent anoikis and permit survival of indi-
vidualized human PS cells. LM-521/E-cadherin substratum also allows derivation
of new hES cell lines from single blastomeres acquired through a single-cell biopsy
3 Biologically Relevant Laminins in Regenerative Medicine 73
Many early in vitro studies with hematopoietic stem cells have been performed on
Engelbreth-Holm-Swarm sarcoma-derived LM-111; however, it is not a natural
laminin for this type of cells. LM-511 and LM-411 are expressed in human and
bone marrow, and LM-211 expression is weak and restricted to arterioles and
LM-111 not expressed at all. b1-laminins are present in adult bone marrow, but not
b2-laminins [21, 73].
In vitro, human CD34+ cells adhere strongly to LM-511/-521, but not to
LM-211 or LM-111. LM-511 exerts mitogenic activity in human hematopoietic
progenitor cells. LM-511/-521, unlike LM-111, are strongly adhesive for multi-
potent hematopoietic FDCP cells. LM-521/-511, unlike other isoforms, enable
robust adhesion for variety of hematopoietic lineages [22, 65, 73].
Neurobiology
Major laminin isoforms in the peripheral nervous system are: a2, a4, and a5.
b1-laminins are ubiquitous, while b2 are restricted to specific locations, like neu-
romuscular junctions. Deficiency in either of those chains results in severe neural
pathologies (reviewed in [15]).
Different cells of peripheral nervous system have specific preferences regarding
laminin substrate in vitro. Spinal motoneurons prefer to sprout long axons on
LM-211 compared to LM-411, while Schwann cells, in the opposite, form longer
sproutings on LM-411 compared to LM-211 [86]. Adult dorsal root ganglion
neurons formed longed neuritis on LM-511 and LM-111, but not on LM-411 or
LM-211. Notably, though neurons spread axons on LM-511 as well as on LM-111,
they engage different integrin receptors [64]. It is possible that different signaling
pathways are activated and cells would differ in receptiveness from growth factor
cues. We suggest therefore that one has not only considered mere adhesiveness and
morphological phenotype as sole evaluation criteria of cell culture system, but also
whether the extracellular matrix coating is biologically relevant and whether it can
provide relevant niche cues to the cell.
Future Challenges
Summary
the worst, is malignant transformation of the cells. Such cells may exhibit
abnormally high survival, adhesion, and proliferation properties, which would
give them a competitive advantage over non-malignant, healthy cells.
7. Use of tissue-specific laminins for expanding tissue-specific cell cultures is a
new trend in regenerative medicine. Natural properties of laminins potentially
can enable not only expansion of a specific cell type, but many other advanced
technologies, like generation of complex, natural-like 3D tissues.
References
1. Abrass CK, Hansen KM, Patton BL. Laminin alpha4-null mutant mice develop chronic
kidney disease with persistent overexpression of platelet-derived growth factor. Am J Pathol.
2010;176:839–49.
2. Akhmanova M, Osidak E, Domogatsky S, Rodin S, Domogatskaya A. Physical, spatial, and
molecular aspects of extracellular matrix of in vivo niches and artificial scaffolds relevant to
stem cells research. Stem Cells Int. 2015;167025.
3. Amps K, Andrews PW, Anyfantis G, Armstrong L, Avery S, Baharvand H, Baker J, Baker D,
Munoz MB, Beil S, Benvenisty N, Ben-Yosef D, Biancotti JC, Bosman A, Brena RM,
Brison D, Caisander G, Camarasa MV, Chen J, Chiao E, Choi YM, Choo AB, Collins D,
Colman A, Crook JM, Daley GQ, Dalton A, de Sousa PA, Denning C, Downie J, Dvorak P,
Montgomery KD, Feki A, Ford A, Fox V, Fraga AM, Frumkin T, Ge L, Gokhale PJ,
Golan-Lev T, Gourabi H, Gropp M, Lu G, Hampl A, Harron K, Healy L, Herath W, Holm F,
Hovatta O, Hyllner J, Inamdar MS, Irwanto AK, Ishii T, Jaconi M, Jin Y, Kimber S,
Kiselev S, Knowles BB, Kopper O, Kukharenko V, Kuliev A, Lagarkova MA, Laird PW,
Lako M, Laslett AL, Lavon N, Lee DR, Lee JE, Li C, Lim LS, Ludwig TE, Ma Y, Maltby E,
Mateizel I, Mayshar Y, Mileikovsky M, Minger SL, Miyazaki T, Moon SY, Moore H,
Mummery C, Nagy A, Nakatsuji N, Narwani K, Oh SK, Olson C, Otonkoski T, Pan F,
Park IH, Pells S, Pera MF, Pereira LV, Qi O, Raj GS, Reubinoff B, Robins A, Robson P,
Rossant J, Salekdeh GH, Schulz TC, et al. Screening ethnically diverse human embryonic
stem cells identifies a chromosome 20 minimal amplicon conferring growth advantage. Nat
Biotechnol. 2011;29:1132–44.
4. Aumailley M, Bruckner-Tuderman L, Carter WG, Deutzmann R, Edgar D, Ekblom P,
Engel J, Engvall E, Hohenester E, Jones JC, Kleinman HK, Marinkovich MP, Martin GR,
Mayer U, Meneguzzi G, Miner JH, Miyazaki K, Patarroyo M, Paulsson M, Quaranta V,
Sanes JR, Sasaki T, Sekiguchi K, Sorokin LM, Talts JF, Tryggvason K, Uitto J, Virtanen I,
von der Mark K, Wewer UM, Yamada Y, Yurchenco PD. A simplified laminin nomenclature.
Matrix Biol. 2005;24:326–32.
5. Barak T, Kwan KY, Louvi A, Demirbilek V, Saygi S, Tuysuz B, Choi M, Boyaci H,
Doerschner K, Zhu Y, Kaymakcalan H, Yilmaz S, Bakircioglu M, Caglayan AO, Ozturk AK,
Yasuno K, Brunken WJ, Atalar E, Yalcinkaya C, Dincer A, Bronen RA, Mane S, Ozcelik T,
Lifton RP, Sestan N, Bilguvar K, Gunel M. Recessive LAMC3 mutations cause
malformations of occipital cortical development. Nat Genet. 2011;43:590–4.
6. Baudin B, Bruneel A, Bosselut N, Vaubourdolle M. A protocol for isolation and culture of
human umbilical vein endothelial cells. Nat Protoc. 2007;2:481–5.
78 A. Domogatskaya and S. Rodin
49. Miner JH, Li C, Mudd JL, Go G, Sutherland AE. Compositional and structural requirements
for laminin and basement membranes during mouse embryo implantation and gastrulation.
Development. 2004;131:2247–56.
50. Miner JH, Patton BL, Lentz SI, Gilbert DJ, Snider WD, Jenkins NA, Copeland NG, Sanes JR.
The laminin alpha chains: expression, developmental transitions, and chromosomal locations
of alpha1-5, identification of heterotrimeric laminins 8-11, and cloning of a novel alpha3
isoform. J Cell Biol. 1997;137:685–701.
51. Miner JH, Yurchenco PD. Laminin functions in tissue morphogenesis. Annu Rev Cell Dev
Biol. 2004;20:255–84.
52. Narva E, Autio R, Rahkonen N, Kong L, Harrison N, Kitsberg D, Borghese L, Itskovitz-Eldor
J, Rasool O, Dvorak P, Hovatta O, Otonkoski T, Tuuri T, Cui W, Brustle O, Baker D,
Maltby E, Moore HD, Benvenisty N, Andrews PW, Yli-Harja O, Lahesmaa R.
High-resolution DNA analysis of human embryonic stem cell lines reveals culture-induced
copy number changes and loss of heterozygosity. Nat Biotechnol. 2010;28:371–7.
53. Nikolova G, Jabs N, Konstantinova I, Domogatskaya A, Tryggvason K, Sorokin L, Fassler R,
Gu G, Gerber HP, Ferrara N, Melton DA, Lammert E. The vascular basement membrane: a
niche for insulin gene expression and Beta cell proliferation. Dev Cell. 2006;10:397–405.
54. Nishimune H, Sanes JR, Carlson SS. A synaptic laminin-calcium channel interaction
organizes active zones in motor nerve terminals. Nature. 2004;432:580–7.
55. Noakes PG, Gautam M, Mudd J, Sanes JR, Merlie JP. Aberrant differentiation of
neuromuscular junctions in mice lacking s-laminin/laminin beta 2. Nature. 1995;374:258–62.
56. Noakes PG, Miner JH, Gautam M, Cunningham JM, Sanes JR, Merlie JP. The renal
glomerulus of mice lacking s-laminin/laminin beta 2: nephrosis despite molecular compen-
sation by laminin beta 1. Nat Genet. 1995;10:400–6.
57. Odenthal U, Haehn S, Tunggal P, Merkl B, Schomburg D, Frie C, Paulsson M, Smyth N.
Molecular analysis of laminin N-terminal domains mediating self-interactions. J Biol Chem.
2004;279:44504–12.
58. Orlando G, Wood KJ, Stratta RJ, Yoo JJ, Atala A, Soker S. Regenerative medicine and organ
transplantation: past, present, and future. Transplantation. 2011;91:1310–7.
59. Patton BL. Laminins of the neuromuscular system. Microsc Res Tech. 2000;51:247–61.
60. Patton BL, Chiu AY, Sanes JR. Synaptic laminin prevents glial entry into the synaptic cleft.
Nature. 1998;393:698–701.
61. Patton BL, Connoll AM, Martin PT, Cunningham JM, Mehta S, Pestronk A, Miner JH,
Sanes JR. Distribution of ten laminin chains in dystrophic and regenerating muscles.
Neuromuscul Disord. 1999;9:423–33.
62. Patton BL, Cunningham JM, Thyboll J, Kortesmaa J, Westerblad H, Edstrom L,
Tryggvason K, Sanes JR. Properly formed but improperly localized synaptic specializations
in the absence of laminin alpha4. Nat Neurosci. 2001;4:597–604.
63. Pinzon-Duarte G, Daly G, Li YN, Koch M, Brunken WJ. Defective formation of the inner
limiting membrane in laminin beta2- and gamma3-null mice produces retinal dysplasia. Invest
Ophthalmol Vis Sci. 2010;51:1773–82.
64. Plantman S, Patarroyo M, Fried K, Domogatskaya A, Tryggvason K, Hammarberg H,
Cullheim S. Integrin-laminin interactions controlling neurite outgrowth from adult DRG
neurons in vitro. Mol Cell Neurosci. 2008;39:50–62.
65. Qian H, Georges-Labouesse E, Nystrom A, Domogatskaya A, Tryggvason K, Jacobsen SE,
Ekblom M. Distinct roles of integrins alpha6 and alpha4 in homing of fetal liver
hematopoietic stem and progenitor cells. Blood. 2007;110:2399–407.
66. Relan NK, Yang Y, Beqaj S, Miner JH, Schuger L. Cell elongation induces laminin alpha2
chain expression in mouse embryonic mesenchymal cells: role in visceral myogenesis. J Cell
Biol. 1999;147:1341–50.
67. Rodin S, Antonsson L, Niaudet C, Simonson OE, Salmela E, Hansson EM, Domogatskaya A,
Xiao Z, Damdimopoulou P, Sheikhi M, Inzunza J, Nilsson AS, Baker D, Kuiper R, Sun Y,
Blennow E, Nordenskjold M, Grinnemo KH, Kere J, Betsholtz C, Hovatta O, Tryggvason K.
3 Biologically Relevant Laminins in Regenerative Medicine 81
The extracellular matrix (ECM) is the non-cellular component of any tissue and
organ [30]. Depending on the origin and on the condition (injury, inflammation,
tumor) of the tissue, the three-dimensional (3D) matrix architecture can assume a
variety of conformations [131]. In some tissues, the ECM can be dense and stiff,
while, in others, it is soft and more porous with gap size of different diameters.
ECM fibers also display highly variable thickness, straightness, and spatial
arrangements. The fibers can be relaxed and non-oriented or, in contrast, linearized
and oriented in a specific direction. These physical characteristics determine the
architecture of the tissue but, as reported below, they are also essential for regu-
lating immune cell migration and behavior [92].
The extracellular matrix consists of a complex assembly of many proteins and
polysaccharides whose specific composition varies from tissue to tissue. The pri-
mary components include insoluble fibrous structural proteins (i.e., collagens,
laminins, fibronectin, vitronectin, and elastin) and proteoglycans. These large and
negatively charged sugars efficiently bind water and fill the space between the
fibers. The ECM primarily fulfills a structural role by maintaining an insoluble
scaffold, which ultimately defines the shape and stiffness of organs [92].
The extracellular matrix consists in a reinforced composite of collagens and
elastic fibers embedded in a viscoelastic gel of proteoglycans, hyaluronan (HA),
and water, together with a wide variety and arrangement of assorted glycoproteins
[40, 46, 54, 78]. These molecules interact by entanglement and cross-linking to
form a bioactive polymer which, in part, regulates the biomechanical properties of
tissues and the phenotype of the cells belonging to those tissues. This regulation
involves molecular interactions that govern the attachment of cells to their ECM
scaffolds through integrin and non-integrin receptors, detachment of cells from
those scaffolds, and molecular rearrangements in the matrix that allow cells to
change shape during morphogenetic and remodeling events that occur in devel-
opment and disease. The amount and composition of the ECM is controlled by the
coordinated and differential regulation of synthesis and turnover of each of the
ECM components. It is becoming increasingly evident that matrix individual
components can exert dramatic effects on cell behavior and tissue response to
endogenous and exogenous stimuli [129].
The ECM provides not only support, tensile strength, and scaffolding for tissues
and cells, but also biochemical signals and specialized proteins (i.e., growth factors,
chemokines, cytokines, small matricellular proteins and small integrin-binding
glycoproteins) [18].
The destabilization or alteration of the structural and chemical matrix compo-
sition affects cell growth, morphogenesis, differentiation, migration, communication
and survival [33], along with inflammation and immune response [82].
In addition to providing structural integrity, the extracellular matrix is recognized
to play critical roles in regulating progenitor and reparative cell behaviors such as
migration, differentiation, proliferation, and survival. The ECM dictates these
activities through its binding to adhesion receptors as well as its ability to regulate
growth factor bioavailability and signaling. More recently, a key role for mechanical
control of cell fate through their interaction with the ECM has emerged [121].
Both mechanical and biochemical molecules influence extracellular matrix
dynamics in multiple ways, by releasing small bioactive signaling molecules and
growth factors stored within the ECM, by eliciting structural changes to matrix
proteins which expose cryptic sites, and by degrading matrix proteins directly [18].
4 Extracellular Matrix: Immunity and Inflammation 85
Versican
the basis of various diseases. Hence, targeting versican as a way to control cell
phenotype offers a novel approach in the treatment of many diseases [129].
Versican is negatively charged due to its glycosaminoglycan (GAG) chains and
attracts water, contributing to the viscoelasticity of the pericellular microenviron-
ment [29]. In addition, versican interacts with a number of matrix components near
the cell surface including hyaluronan (HA), tenascin-R and -C, thrombospondin 1,
fibronectin, and fibrillin [49, 62, 74, 133] to create a mechanically active
biopolymer around cells which influences their ability to change shape, adhere,
proliferate, migrate, assemble other ECM components, and survive. Versican and
extracellular matrix-related molecules may modify the mechanical stiffness around
cells contributing to alterations in mechanotransduction influencing cell behavior
and phenotype [23, 26, 125]. Versican can also act as a reservoir for cytokines and
growth factors to be released at various times, establishing a further subtle control
over cell activity and behavior [34, 75, 126].
Versican interacts with several different ECM molecules and, in part, plays a
central role in matrix assembly. The domain structure of versican lends itself to
multiple types of interactions through either protein–protein or protein–carbohy-
drate interactions. The best known of these interactions is the binding between the
amino-terminal domain of versican (G1) and hyaluronan (HA) [64]. This interac-
tion is stabilized by another protein—link protein—which exhibits selective bind-
ing specificity for both HA and versican [20] (Fig. 4.1).
In addition to hyaluronan, versican interacts with other extracellular matrix
molecules controlling their organization: it interacts with tenascin-R through its
Fibrillin
NH 3
Versican Thrombo-
spondin-1
Chemokines,
Cytokines,
Growth
factors Fibronectin
Cell
receptors
Hyaluronan
Thrombospondins
multiple cell surface receptors. Information emerged over the past decade identifies
TSPs as important mediators of cellular homeostasis, assigning new important roles
in cardiovascular pathology to these proteins [102].
Recent studies of the functions of TSP in the cardiovascular system, diabetes,
and aging, which placed several thrombospondins in a position of critical regula-
tors, demonstrated the involvement of these proteins in practically each aspect of
cardiovascular pathophysiology related to atherosclerosis: inflammation, immunity,
leukocyte recruitment and function, function of vascular cells, angiogenesis, and
responses to hypoxia, ischemia, and hyperglycemia. TSPs play also a major role in
the development and ultimate outcome of the complications associated with
atherosclerosis–myocardial infarction, and heart hypertrophy and failure. Their
expression and significance increase with age and with the progression of diabetes,
two major contributors to the development of atherosclerosis and its complications
[102].
Thrombospondins comprise a conserved family of extracellular, oligomeric,
multidomain, calcium-binding glycoproteins. In general, basal metazoa and pro-
tostomes encode a single TSP in their genomes, and deuterostomes have multiple
TSP genes. The mammal thrombospondins have many complex tissue-specific
roles, including activities in wound healing and angiogenesis, vessel wall biology,
connective tissue organization, and synaptogenesis. These activities derive mech-
anistically from interactions with cell surfaces, growth factors, cytokines, or com-
ponents of the extracellular matrix that collectively regulate many aspects of cell
phenotype. Emerging evidence on the functions of TSPs in invertebrates suggests
that ancient functions include bridging activities in cell–cell and cell–ECM inter-
actions. Knowledge of TSP domain structures provides a rational basis for under-
standing their roles in vivo and associations with human disease and is assisting
ongoing translational applications [2].
Cells
Cells can remodel and reshape the extracellular matrix by degrading and
reassembling it, thus playing an active role in sculpting their surrounding envi-
ronment and directing their own phenotypes. The dynamic reciprocal communi-
cation between cells and the ECM plays a fundamental role in tissue development,
homeostasis, and wound healing [18].
Cells, in order to change shape during division and migration, must modify their
pericellular environment by first degrading the existing matrix and replacing it with
new components. Hyaluronan and versican drive the changes leading to expansion
of the pericellular matrix and to modifications in the mechanical properties of the
ECM that influence cell phenotype [129].
Most matrix components are produced by fibroblasts that also play a role in their
assembly into fibers and their spatial disposition. Collagen cross-linking is almost
exclusively mediated by an enzyme, the lysyl oxidase (LOX) [134]. The synthesis
4 Extracellular Matrix: Immunity and Inflammation 91
Inflammation
appears to be the most important sheddase in terms of the range of its targets. The
inflammatory condition is characterized by an increased accumulation of leukocytes
in tissue mediated by the transmembrane protein selectins. ADAM-17 also has a
role in the shedding of L-selectin, a cellular adhesion molecule, from macrophages,
and this process is promoted by leukocyte attachment to endothelial or E-selectins.
Thus, ADAM-17 was shown to shed several factors contributing to successful
recruitment of leukocytes to the inflammation site. Interestingly, there is some
evidence that ADAM17 controls not only pro- but also anti-inflammatory signals.
The enzyme was reported to down-regulate the macrophages activation cleaving
colony-stimulating factor-1 (CSF-1) from their surface [123]. In addition, a corre-
lation was reported between ADAM-17 expression and IL-15 receptor a activation,
which is a soluble form of the receptor implied in the collagen-induced arthritis and
cardiac allograph rejection. Also, different stimuli activate ADAM17-mediated IL-6
receptor-a cleavage, contributing to the decline of neutrophil infiltration and to
promotion of monocyte recruitment, essential for resolution of the inflammation.
Among the tissue inhibitors of matrix metalloproteinases, TIMP-3 is the only one
that binds to the extracellular matrix and contains an amino acid sequence
(PFG) required to inhibit ADAM-17 [66]. TIMP-3 is induced by molecules
involved in inflammation, such as the pro-inflammatory agent PMA and the
anti-inflammatory cytokine TGF-b, and has the potential to impact many different
branches of haematopoiesis, immunity, and inflammation, primarily as a regulator
of ADAM-17, but also as an inhibitor of other proteases which target growth
factors, cytokines, and adhesion molecules [70].
Phagocytosis
TSP-1 CD47
Leukocytes
NO Leukocytes infiltration
apoptosis
Angiogenesis
ABT
CD36
510
PPAR γ
Endothelial cell sapoptosis
Immunity
tissues, can play a key role in facilitating or suppressing the anti-tumor immune
surveillance. It is now well accepted that a deregulated ECM favours tumor pro-
gression and metastasis. Recent progress in imaging technologies has also high-
lighted the impact of the matrix architecture found in solid tumor on immune cells
and especially T cells. Besides the effect exerted on tumor cells, the physical and
biochemical properties of the ECM are also able to modulate a number of processes
in immune cells, especially lymphocytes, that can ultimately lead to inefficient
tumor killing. Extracellular matrix determinants can indeed promote tumor evasion
from the immune system, both by inhibiting the anti-tumor effector activity of T
cells, either directly or through the recruitment of immunosuppressive cells, and by
limiting cell contact with tumor cells [92].
Along with collagen and fibronectin, some non-structural ECM proteins are
often upregulated during tumor development and can modulate cell–cell and cell–
ECM interactions, eventually restraining T cell activation [15, 98]. For instance,
tenascin C seems to directly inhibit T cell proliferation and IFN-c production in
lung cancer [90] while the glycoprotein SPARC seems to influence the trafficking
and the function of immune cells, as SPARC-knockout mice display an increased
number of macrophages and neutrophils in tumors and higher cytotoxicity in
polymorphonuclear cells [4, 99]. In addition, several ECM fragments are able to
recruit and activate macrophages and neutrophils to sites of inflammation, where
they could regulate inflammation and adaptive responses [1, 101].
The innate immune system down-regulates effector mechanisms and restores
homoeostasis in injured tissue via cytokines from the IL-10 and TGF (transforming
growth factor) families mainly released from macrophages, preferentially the M2
subset, which have a capacity to induce regulatory T cells, inhibit the production of
pro-inflammatory cytokines, and induce healing of the tissue by regulating extra-
cellular matrix protein deposition and angiogenesis [106]. Proteoglycans are also
synthesized by leukocytes and may play a role in the inflammatory response [110].
Within the last year, the role of hyaluronan as an endogenous activator of innate
alloimmunity has been investigated [67]. Recent studies have described that the
hyaluronan receptor, CD44, modulates these innate immune responses by aug-
menting regulatory T cell function, and the expression of negative regulators of
toll-like receptor signaling. Hyaluronan activates innate alloimmune responses and
subsequently influences adaptive alloimmunity.
In regulatory T cells, HMW-HA stimulates STAT5 signaling through CD44
cross-linking, promoting their maintenance and thereby inhibiting their prolifera-
tion. Conventional T cell precursors stimulated with HMW-HA produce IL-10, and
infusion of these cells attenuates the disease course in a murine model of colitis
[12].
CD44 is a type I transmembrane glycoprotein and is widely regarded as the
major cell surface HA-binding protein. Widely studied in several contexts, CD44
interactions with HA have important roles in tumor metastasis, lymphocyte adhe-
sion, T cell signaling, angiogenesis, and inflammation. CD44 is expressed on many
cell types that contribute to inflammation including leukocytes, neutrophils, mac-
rophages, chondrocytes, fibroblasts, epithelial, and endothelial cells. While the
glycosaminoglycans (GAGs) of CD44 can bind to cytokines, growth factors, and
extracellular matrix proteins such as fibronectin, the majority of the functions of
CD44 depend upon its ability to bind to hyaluronan [93].
Toll-like receptors function as surveillance receptors, interacting with a number
of microbial-derived molecules and activating the innate immune system in
response to pathogen-associated molecular patterns. Increasingly, the TLRs are also
shown to sense damage-associated molecular patterns in response to injury as well.
The idea that endogenous matrix degradation products act as regulators of cellular
processes is not a new one, but with respect to GAG fragments, the role of HA is
the best studied. Hyaluronan is a component of the cellular coat on some pathogens
and many of them also express hyaluronidases. The presence of a HA coat likely
assists in evasion by the immune system, while the hyaluronidase enzymes may aid
in colonization of the host [76].
4 Extracellular Matrix: Immunity and Inflammation 103
References
24. Dubois B, Masure S, Hurtenbach U, et al. Resistance of young gelatinase B-deficient mice to
experimental autoimmune encephalomyelitis and necrotizing tail lesions. J Clin Invest.
1999;104:1507–15.
25. Ducharme A, Frantz S, Aikawa M, et al. Targeted deletion of matrix metalloproteinase-9
attenuates left ventricular enlargement and collagen accumulation after experimental
myocardial infarction. J Clin Invest. 2000;106:55–62.
26. DuFort CC, Paszek MJ, Weaver VM. Balancing forces: architectural control of mechan-
otransduction. Nat Rev Mol Cell Biol. 2011;12:308–19.
27. Edelstam GA, Laurent UB, Lundkvist OE, et al. Concentration and turnover of
intraperitoneal hyaluronan during inflammation. Inflammation. 1992;16:459–69.
28. Egeblad M, Werb Z. New functions for the matrix metalloproteinases in cancer progression.
Nat Rev Cancer. 2002;2:161–74.
29. Evanko SP, Tammi MI, Tammi RH, et al. Hyaluronan-dependent pericellular matrix. Adv
Drug Deliv Rev. 2007;59:1351–65.
30. Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. J Cell Sci.
2010;123:4195–200.
31. Frey H, Schroeder N, Manon-Jensen T, et al. Biological interplay between proteoglycans
and their innate immune receptors in inflammation. FEBS J. 2013;280:2165–79.
32. Gabison EE, Huet E, Baudouin C, et al. Direct epithelial-stromal interaction in corneal
wound healing: role of EMMPRIN/CD147 in MMPs induction and beyond. Prog Retin Eye
Res. 2009;28:19–33.
33. Gaudet AD, Popovich PG. Extracellular matrix regulation of inflammation in the healthy and
injured spinal cord. Exp Neurol. 2014;258:24–34.
34. Gill S, Wight TN, Frevert CW, et al. Proteoglycans: key regulators of pulmonary
inflammation and the innate immune response to lung infection. Anat Rec (Hoboken).
2010;293:968–81.
35. Gomis-Rüth FX. Structural aspects of the metzincin clan of metalloendopeptidases. Mol
Biotechnol. 2003;24:157–202.
36. Graesser D, Mahooti S, Madri JA. Distinct roles for matrix metalloproteinase-2 and a4
integrin in autoimmune T cell extravasation and residency in brain parenchyma during
experimental autoimmune encephalomyelitis. J Neuroimmunol. 2000;109:21–131.
37. Grimbert P, Bouguermouh S, Baba N, et al. Thrombospondin/CD47 interaction: a pathway
to generate regulatory T cells-from human CD4+ CD25− T cells in response to inflammation.
J Immunol. 2006;177:3534–41.
38. Gross J, Lapiere CM. Collagenolytic activity in amphibian tissues: a tissue culture assay.
Proc Natl Acad Sci U S A. 1962;48:1014–22.
39. Hadler-Olsen E, Fadnes B, Sylte I, et al. Regulation of matrix metalloproteinase activity in
health and disease. FEBS J. 2011;278:28–45.
40. Hay ED. Cell biology of extracellular matrix. New York: Plenum Press; 1991.
41. Hernandez-Barrantes S, Bernardo M, Toth M, et al. Regulation of membrane type-matrix
metalloproteinases. Semin Cancer Biol. 2002;12:131–8.
42. Hirose J, Kawashima H, Yoshie O, et al. Versican interacts with chemokines and modulates
cellular responses. J BiolChem. 2001;276:5228–34.
43. Holmbeck K, Bianco P, Caterina J, et al. MT1-MMP-deficient mice develop dwarfism,
osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell.
1999;99:81–92.
44. Huang H, Campbell SC, Bedford DF, et al. Peroxisome proliferator-activated receptor c
ligands improve the antitumor efficacy of thrombospondin peptide ABT510. Mol Cancer
Res. 2004;2:541–50.
45. Hynes RO. The extracellular matrix: not just pretty fibrils. Science. 2009;326:1216–9.
46. Hynes RO, Yamada KM. Extracellular matrix biology. Cold Spring Harbor, NY: Cold
Spring Harbor Laboratory Press; 2012.
47. Isenberg JS, Frazier WA, Roberts DD. Thrombospondin-1: a physiological regulator of
nitric oxide signaling. Cell Mol Life Sci. 2008;65:728–42.
106 A. Cataldi and V. di Giacomo
48. Isenberg JS, Martin-Manso G, Maxhimer JB, et al. Regulation of nitric oxide signalling by
thrombospondin 1: implications for anti-angiogenic therapies. Nat Rev Cancer. 2009;9:
182–94.
49. Isogai Z, Aspberg A, Keene DR, et al. Versican interacts with fibrillin-1 and links extracellular
microfibrils to other connective tissue networks. J Biol Chem. 2002;277:4565–72.
50. Jiang D, Liang J, Noble PW. Hyaluronan in tissue injury and repair. Annu Rev Cell Dev
Biol. 2007;23:435–61.
51. Jiang D, Liang J, Noble PW. Hyaluronan as an immune regulator in human diseases. Physiol
Rev. 2011;91:221–64.
52. Joronen KR, Ala-aho ML, Majuri H, et al. Adenovirus mediated intra-articular expression of
collagenase-3 (MMP-13) induces inflammatory arthritis in mice. Ann Rheum Dis.
2004;63:656–64.
53. Joronen K, Kähäri VM, Vuorio E. Temporospatial expression of matrix metalloproteinases
and tissue inhibitors of matrix metalloproteinases in mouse antigen induced arthritis.
Histochem Cell Biol. 2005;124:535–45.
54. Karamanos N. Extracellular matrix: pathobiology and signaling. Berlin, Boston: Walter de
Gruyter; 2012.
55. Kendall RT, Feghali-Bostwick CA. Fibroblasts in fibrosis: novel roles and mediators. Front
Pharmacol. 2014;5:123.
56. Kessenbrock K, Plaks V, Werb Z. Matrix metalloproteinases: regulators of the tumor
microenvironment. Cell. 2010;141:52–67.
57. Khokha R, Murthy A, Weiss A. Metalloproteinases and their natural inhibitors in
inflammation and immunity. Nat Rev Immunol. 2013;13:649–65.
58. Kieseier BC, Kiefer R, Clements JM, et al. Matrix metalloproteinase-9 and -7 are regulated
in experimental autoimmune encephalomyelitis. Brain. 1998;121:159–66.
59. Kim KH, Burkhart K, Chen P, et al. Tissue inhibitor of metalloproteinase–1 deficiency
amplifies acute lung injury in bleomycin-exposed mice. Am J Respir Cell Mol Biol.
2005;33:271–9.
60. Kirk JA, Cingolani OH. Thrombospondins in the transition from myocardial infarction to
heart failure. J Mol Cell Cardiol. 2016;90:102–10.
61. Kudo-Saito C, Shirako H, Takeuchi T, et al. Cancer metastasis is accelerated through
immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell. 2009;15:
195–206.
62. Kuznetsova SA, Issa P, Perruccio EM, et al. Versican-thrombospondin-1 binding in vitro
and colocalization in microfibrils induced by inflammation on vascular smooth muscle cells.
J Cell Sci. 2006;119:4499–509.
63. Laurent TC, Fraser JR. Hyaluronan. FASEB. 1992;J6:2397–404.
64. LeBaron RG, Zimmermann DR, Ruoslahti E. Hyaluronate binding properties of versican.
J Biol Chem. 1992;267:10003–10.
65. Lelongt B, Bengatta S, Delauche M, et al. Matrix metalloproteinase 9 protects mice from
anti-glomerular basement membrane nephritis through its fibrinolytic activity. J Exp Med.
2001;193:793–802.
66. Lee MM, Yoon BJ, Osiewicz K, et al. Tissue inhibitor of metalloproteinase 1 regulates
resistance to infection. Infect Immun. 2005;73:661–5.
67. Lee-Sayer SS, Dong Y, Arif AA, et al. The where, when, how, and why of hyaluronan
binding by immune cells. Front Immunol. 2015;6:150.
68. Li L, Li H. Role of microRNA-mediated MMP regulation in the treatment and diagnosis of
malignant tumors. Cancer Biol Ther. 2013;14:796–805.
69. Li Q, Park PW, Wilson CL, et al. Matrilysin shedding of syndecan-1 regulates chemokine
mobilization and transepithelial efflux of neutrophils in acute lung injury. Cell.
2002;111:635–46.
70. Lisi S, D’Amore M, Sisto M. ADAM17 at the interface between inflammation and
autoimmunity. Immunol Lett. 2014;S0165-2478(14):00178-3.
4 Extracellular Matrix: Immunity and Inflammation 107
71. Löffek S, Schilling O, Franzke CW. Series “matrix metalloproteinases in lung health and
disease”: Biological role of matrix metalloproteinases: a critical balance. Eur
Respir J. 2011;38:191–208.
72. Lopez-Dee Z, Pidcock K, Gutierrez LS. Thrombospondin-1: multiple paths to inflammation.
Mediat Inflamm. 2011;2011:296069.
73. Lu P, Takai K, Weaver VM, et al. Extracellular matrix degradation and remodeling in
development and disease. Cold Spring Harb Perspect Biol. 2011;3(ii):a005058.
74. Lundell A, Olin AI, Morgelin M, et al. Structural basis for interactions between tenascins
and lectican C-type lectin domains: evidence for a crosslinking role for tenascins. Struct
(Camb). 2004;12:1495–506.
75. Macri L, Silverstein D, Clark RA. Growth factor binding to the pericellularmatrix and its
importance in tissue engineering. Adv Drug Deliv Rev. 2007;59:1366–81.
76. Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol.
1994;12:991–1045.
77. McDonald JA, Camenisch TD. Hyaluronan: genetic insights into the complex biology of a
simple polysaccharide. Glycoconj J. 2002;1:331–9.
78. Mecham RP. The extracellular matrix: an overview. Berlin: Springer; 2011.
79. Mittal R, Gonzalez-Gomez I, Prasadarao NV. Escherichia coli K1 promotes the ligation of
CD47 with thrombospondin-1 to prevent the maturation of dendritic cells in the pathogenesis
of neonatal meningitis. J Immunol. 2010;185:2998–3006.
80. Moncada-Pazos A, Obaya AJ, Llamazares M, et al. ADAMTS-12 metalloprotease is
necessary for normal inflammatory response. J Biol Chem. 2012;287:39554–63.
81. Morris DG, Huang X, Kaminski N, et al. Loss of integrin avb6-mediated TGF-b activation
causes MMP12-dependent emphysema. Nature. 2003;422:169–73.
82. Morwood SR, Nicholson LB. Modulation of the immune response by extracellular matrix
proteins. Arch Immunol Ther Exp (Warsz). 2006;54:367–74.
83. Murasawa Y, Watanabe K, Yoneda M, et al. Homotypic versican G1 domain interactions
enhance hyaluronan incorporation into fibrillin microfibrils. J Biol Chem. 2013;288:29170–81.
84. Nagase H, Visse R, Murphy G. Structure and function of matrix metalloproteinases and
TIMPs. Cardiovasc Res. 2006;69:562–73.
85. Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harb
Perspect Biol. 2012;4:a006049.
86. Nissinen L, Kähäri VM. Matrix metalloproteinases in inflammation. Biochim Biophys Acta.
2014;1840:2571–80.
87. Nygårdas PT, Hinkkanen AE. Up-regulation ofMMP-8 andMMP-9 activity in the BALB/c
mouse spinal cord correlates with the severity of experimental autoimmune encephalomyeli-
tis. Clin Exp Immunol. 2002;128:245–54.
88. Olin AI, Morgelin M, Sasaki T, et al. The proteoglycans aggrecan and Versican form networks
with fibulin-2 through their lectin domain binding. J Biol Chem. 2001;276:1253–61.
89. Owen CA, Hu Z, Lopez-Otin C, et al. Membrane-bound matrix metalloproteinase-8 on
activated polymorphonuclear cells is a potent, tissue inhibitor of metalloproteinase-resistant
collagenase and serpinase. J Immunol. 2004;172:7791–803.
90. Parekh K, Ramachandran S, Cooper J, et al. Tenascin-C, over expressed in lung cancer down
regulates effector functions of tumor infiltrating lymphocytes. Lung Cancer. 2005;47:17–29.
91. Parks WC, Wilson CW, Lopez-Boado YS. Matrix metalloproteinases as modulators of
inflammation and innate immunity. Nat Rev Immunol. 2004;4:617–29.
92. Peranzoni E, Rivas-Caicedo A, Bougherara H, et al. Positive and negative influence of the
matrix architecture on antitumor immune surveillance. Cell Mol Life Sci. 2013;70:4431–48.
93. Petrey AC, de la Motte CA. Hyaluronan, a crucial regulator of inflammation. Front
Immunol. 2014;5:101.
94. Piperi C, Papavassiliou AG. Molecular mechanisms regulating matrix metalloproteinases.
Curr Top Med Chem. 2012;12:1095–112.
95. Ra HJ, Parks WC. Control of matrix metalloproteinase catalytic activity. Matrix Biol.
2007;26:587–96.
108 A. Cataldi and V. di Giacomo
96. Reinhardt DP, Sasaki T, Dzamba BJ, et al. Fibrillin-1 and fibulin-2 interact and are
colocalized in some tissues. J Biol Chem. 1996;271:19489–96.
97. Rietz A, Spiers J. The relationship between the MMP system, adrenoceptors and
phosphoprotein phosphatases. Br J Pharmacol. 2012;166:1225–43.
98. Sangaletti S, Colombo MP. Matricellular proteins at the crossroad of inflammation and
cancer. Cancer Lett. 2008;267:245–53.
99. Sangaletti S, Stoppacciaro A, Guiducci C, et al. Leukocyte, rather than tumor-produced
SPARC, determines stroma and collagen type IV deposition in mammary carcinoma. J Exp
Med. 2003;198:1475–85.
100. Silini A, Parolini O, Huppertz B, et al. Soluble factors of amnion-derived cells in treatment
of inflammatory and fibrotic pathologies. Curr Stem Cell Res Ther. 2013;8:6–14.
101. Sorokin L. The impact of the extracellular matrix on inflammation. Nat Rev Immunol.
2010;10:712–23.
102. Stenina-Adognravi O. Thrombospondins: old players, new games. Curr Opin Lipidol.
2013;24:401–9.
103. Stern R, Asari AA, Sugahara KN. Hyaluronan fragments: an information-rich system. Eur J
Cell Biol. 2006;85:699–715.
104. Stern R, Jedrzejas MJ. Hyaluronidases: their genomics, structures, and mechanisms of
action. Chem Rev. 2006;106:818–39.
105. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev
Cell Dev Biol. 2001;17:463–516.
106. Striz I, Brabcova E, Kolesar L, et al. Cytokine networking of innate immunity cells: a
potential target of therapy. Clin Sci (Lond). 2014;126:593–612.
107. Takahashi C, Sheng Z, Horan TP, et al. Regulation of matrix metalloproteinase-9 and
inhibition of tumor invasion by the membrane-anchored glycoprotein RECK. Proc Natl
Acad Sci U S A. 1998;95:13221–6.
108. Tezvergil-Mutluay A, Agee KA, Hoshika T, et al. The requirement of zinc and calcium ions
for functional MMP activity in demineralized dentin matrices. Dent Mater. 2010;26:1059–67.
109. Toba H, Cannon PL, Yabluchanskiy A, et al. Transgenic overexpression of macrophage
matrix metalloproteinase-9 exacerbates age-related cardiac hypertrophy, vessel rarefaction,
inflammation, and fibrosis. Am J Physiol Heart Circ Physiol. 2017;312:H375–83.
110. Uhlin-Hansen L, Wik T, Kjellen L, et al. Proteoglycan metabolism in normal and
inflammatory human macrophages. Blood. 1993;82:2880–9.
111. Van den Steen PE, Opdenakker G, Wormald MR, et al. Matrix remodelling enzymes, the
protease cascade and glycosylation. Biochim Biophys Acta. 2001;1528:61–73.
112. Vandenbroucke RE, Dejonckheere E, Van Hauwermeiren F, et al. Matrix metalloproteinase
13 modulates intestinal epithelial barrier integrity in inflammatory diseases by activating
TNF. EMBO Mol Med. 2013;5:932–48.
113. Vargová V, Pytliak M, Mechírová V. Matrix metalloproteinases. EXS. 2012;103:1–33.
114. Verma S, Kesh K, Ganguly N, et al. Matrix metalloproteinases and gastrointestinal cancers:
impacts of dietary antioxidants. J Biol Chem. 2014;5:355–76.
115. Vermaelen KY, Cataldo D, Tournoy K, et al. Matrix metalloproteinase-9-mediated dendritic
cell recruitment into the airways is a critical step in a mouse model of asthma. J Immunol.
2003;171:1016–22.
116. Vigetti D, Karousou E, Viola M, et al. Hyaluronan: biosynthesis and signaling. Biochim
Biophys Acta. 2014;1840:2452–9.
117. Vigetti D, Rizzi M, Moretto P, et al. Glycosaminoglycans and glucose prevent apoptosis in
4-methylumbelliferone-treated human aortic smooth muscle cells. J Biol Chem.
2011;286:34497–503.
118. Vigetti D, Genasetti A, Karousou E, et al. Proinflammatory cytokines induce hyaluronan
synthesis and monocyte adhesion in human endothelial cells through hyaluronan synthase 2
(HAS2) and the nuclear factor-kappaB (NF-kappaB) pathway. J Biol Chem.
2011;285:24639–45.
4 Extracellular Matrix: Immunity and Inflammation 109
Abstract The extracellular matrix (ECM) has unique biochemical, mechanical and
organisational properties through which it provides a physical scaffolding for cells;
a barrier that protects tissues; several signals that affect cell behaviour; and a
reservoir for biologically active molecules. Considering the importance of ECM in
regulating many fundamental cell processes, a myriad of strategies and materials
has been developed to reproduce its properties. The first part of the chapter covers
various approaches aiming to generate scaffolds whose fibre size, orientation and
stiffness could mimic the ECM nanofibrous structure. In particular, the use of
natural fibrous proteins, the application of electrospinning and freeze-drying and
examples of tissue engineering applications are presented. The second part dis-
cusses strategies aiming to address the ECM ligand-binding function and to
reproduce the dynamic, reciprocal, dialogue between cells and their microenvi-
ronment; examples of 3D scaffolds for controlled release of growth factors, drugs
and genetic material are reported. Researchers have also used native ECM com-
ponents to recapitulate the biochemical and biophysical properties of ECM. In the
third part of the chapter, the use of fibrinogen and fibrin is presented as an example
of natural scaffolds recapitulating ECM functions. Fibrinogen and fibrin can be used
as provisional matrix in regenerating tissues; moreover, by varying the fabrication
method and by blending them with other materials, it is possible to produce
biodegradable scaffolds with reasonable control of degradation rate and drug
release.
Keywords Fibrous scaffolds Natural biomaterials Electrospinning
Hydrogels Freeze-drying 3D scaffolds Controlled drug release
Growth factors Gene therapy Fibrinogen Fibrin Crosslinkers
Cell carrier
Fig. 5.1 Images obtained by scanning electron microscopy of different fibrous scaffold structure
formed by micropores (sponge, left), nano- or micro-fibres (electrospun film, right)
5 Biologically Inspired Materials in Tissue Engineering 115
because they are recognised by the host tissue as normal constituents rather than as
a foreign matter [17]. Other advantages of collagen are its abundance and the ease
of functionalisation with therapeutic moieties. However, the mechanical and
enzymatic stability of collagen produced from animal/human source is minimal
because natural crosslinking does not occur in vitro. For that reason, exogenous
crosslinking (chemical, biological or physical) is required to control stability and
durability in the body [18–21], even though crosslinking has been associated with
cytotoxic issues [22], scaffold calcification [23] and foreign body response [24, 25].
Collagen-based materials can be used in different forms—hydrogels, fibres, films
and sponges—for tissue engineering. For example, self-assembled collagen
hydrogels have been shown to be a promising candidate as cell carrier for adipose
stem cells and nucleus pulposus cells [26]. Electrospun collagen has been able to
enhance wound healing [27, 28]; however electrospinning processing can denature
collagen [29]. Although collagen is mainly extracted from porcine, bovine and
human tissues [30], these sources carry the potential risk of disease transmission
[31], allergic reactions [32], immunogenicity [33] and variability between batches
[30, 34]. In order to avoid these possible issues, fish-extracted collagen and
recombinant human collagen have been proposed as possible alternatives [35, 36].
Recombinant collagen from transgenic tobacco plant, mammalian cells, bacteria or
silkworm have been employed in research [30, 36, 37], whereas fish-extracted
collagen has been rarely used [38] because of its non-mammalian origin.
Elastin is a rubber-like protein that exhibits high elasticity, low stiffness and high
resilience [39]. Therefore, elastin is a crucial protein in tissues requiring large
stretching and shape recovery, such as blood vessels, ligaments, lungs or skin [39].
In the same way, elastin-based scaffolds are particularly interesting for tissue
engineering applications that require large elastic deformation [40, 41]. As for
collagen scaffolds, mechanical and biological stability of elastin-based scaffolds can
be increased by means of chemical crosslinking [40]. Elastin is composed of
approximately 800 amino acids, and it is synthesized from tropoelastin that is
formed by two domains: hydrophobic domains—mainly glycine, valine, alanine
and proline residues—, and hydrophilic domains—predominantly lysine and ala-
nine residues [42]. The principal sources of elastin are solubilised elastin from
mammalian tissues and recombinant elastin-like protein [43]. The main advantage
of recombinant elastin is the better control over the physical and chemical char-
acteristics of the resulting scaffold in comparison to solubilised elastin [40]. For
example, elastin-like proteins have been used to produce films for hernia repair
[44], carrier for growth factor [45] or gene therapy [46].
Elastin has good hemocompatibility, anti-thrombogenic properties and regula-
tory functions over luminal endothelial cells and vascular smooth muscle cells [47].
Consequently, elastin is an excellent biomaterial for vascular applications [48, 49].
Elastin-coated polymeric or metallic devices have been used for increasing
hemocompatibility [50] and for decreasing platelet adhesion, fibrin deposition [51,
52] and implant calcification [53]. Nevertheless, the success of elastin coating may
be affected by the chemical modifications required to covalently bond elastin to the
device surface [54]. Elastin has been also proposed to construct blood vessels;
5 Biologically Inspired Materials in Tissue Engineering 117
however, the use of elastin alone has limited applications due to the low ultimate
tensile strength of the scaffold [54]. Therefore, the combination of elastin with
polyurethane (PU), poly-lactic-co-glycolic acid (PLGA) or polycaprolactone
(PCL) has been explored to improve mechanical properties [55–57]. Finally,
industrial and clinical use of elastin-based scaffolds has been limited by the issues
related to elastin extraction process, which can compromise its biomechanical and
biological properties [54].
Keratin is one of the main constituents of skin, hair and nails [58]. Keratin
contains cell-binding motifs, such as RGD and LDV, that enhance cell attachment,
migration and proliferation [40]. In addition, waste products from the poultry
processing can be used as source of keratin. This fact ensures a cheap and con-
tinuous source, and therefore, keratin extraction has been widely investigated.
Keratin can be extracted by oxidative or reductive methods, but only reductive
extraction with DTT or mercaptoethanol ensures long in vivo stability of recon-
stituted scaffolds [40]. These keratin solutions can be employed to produce hy-
drogels, sponges, films and fibres [59].
Although keratin-based products are well known in cosmetic industry, its clinical
use is limited because of its low mechanical properties and extremely high flexibility
[60]. However, keratin hydrogels and films have been demonstrated to promote
proliferation of endothelial cells, keratinocytes and fibroblasts, and to regulate
Schwann cells activity [61]. Furthermore, keratin-based scaffolds have been suc-
cessfully used as tuneable drug delivery system [62], to improve nerve repair [63] to
accelerate epithelisation and wound remodelling [64] in preclinical models.
Silk is a lightweight, strong and elastic protein produced by several insects and
spiders [65]. The two main components of silk are sericin and fibroin; sericin is a
glue-like protein that links fibroin fibres, while fibroin is a structural protein
commonly used to produce scaffolds [66]. Silk fibroin is composed of glycine-X
sequences, where X is often alanine, serine, threonine and valine [67] and is typ-
ically extracted from silkworm chrysalis [68]. Silk fibroin can be easily processed in
water or other solvents to form hydrogels, fibres, films or sponges [69]. The
degradation rate of silk-based scaffolds can be adjusted from few months to up to
1 year by modifying processing parameters, such as the solvent, fibroin concen-
tration and pore size [70].
Silk fibroin has been used for producing blood vessels with successful attach-
ment and ingrowth of both smooth muscle and endothelial cells [71, 72]. Indeed,
electrospun silk fibroin grafts have been demonstrated to be an encouraging strategy
to repair small blood vessels [73], as they promote endothelial cell and smooth
muscle cells infiltration and the development of an elastic lamina on the internal
surface of scaffolds in a short term [74]. In addition, silk electrospun films have
been studied as potential candidates for wound healing dressings due to their
protective capacity against dehydration and microorganisms and their remarkable
mechanical properties [75–77]. Clinical data concerning silk, that is mainly used for
suturing, suggest that silk is well tolerated at short term with few cases of hyper-
sensitivity associated with sericin residues [69]. However long-term immune
response to this non-mammalian derived material is still unknown [69].
118 G. Fontana et al.
Table 5.2 Examples of strategies for controlled drug delivery from 3D scaffolds
Bio-molecule Material Type of strategy Outcome References
VEGF Type I Covalent The covalent incorporation of [152]
collagen incorporation VEGF enhanced the angiogenic
capabilities of the collagen
matrices
VEGF Fibrin Immobilization, The immobilization of VEGF [154]
use of within the scaffold allowed
enzymatically cell-demanded release with
labile sequences increased formation of new
blood vessels with normal
morphological appearance
VEGF PEG Covalent The subcutaneous implantation [156]
incorporation of VEGF-containing scaffolds
in rats resulted in remodelling
of the scaffold and formation of
native, vascularised tissue
bFGF Type I Immobilization Scaffolds in which heparin was [164]
collagen immobilized showed increased
vascularisation for periods up to
3 weeks in vivo
SDF-1a Type I Immobilization Scaffolds containing [166]
collagen immobilized heparin to trap the
stem cell chemo-attractant
SDF-1a were implanted
subcutaneously in mice. Only
few progenitor cells were
recruited early after
implantation
BMP-7 PLLA Particles Scaffolds containing [181]
reservoir system BMP-7-loaded nanospheres
allowed for a prolonged
maintenance of therapeutic
activity of BMP-7 and resulted
in improved bone formation in
rats
BMP-7 Fibrin or Embedding In vitro tests showed that [203]
expressing collagen adenoviruses maintained their
adenovirus therapeutic efficacy for longer
when embedded in fibrin gels.
In vivo, bone formation was
observed in animals implanted
with fibrin or collagen
virus-loaded gels
(continued)
122 G. Fontana et al.
Even though diseases and trauma are often associated with alterations in the
structure and properties of ECM, conventional drug delivery approaches focused on
restoring cell function without repairing the microenvironment that surrounds the
targeted cells [132–134]. This uncontrolled drug delivery often results in poor
therapeutic effect or in severe side effects. An altered microenvironment can affect
the absorption and retention of the drug into the tissue. Moreover, a hostile
microenvironment can accelerate the degradation or denaturation of the bioactive
molecule, resulting in a poor outcome of the therapy. In fact, the pharmacokinetics
in the host tissue is often characterised by an initial excess followed by a sharp
decrease [135–137]. In order to counterbalance this transient effect, biomacro-
molecules are often delivered at high doses, which can lead to severe side effects, as
reported in both animal and clinical studies [138, 139].
Due to these limitations, current research focused on developing 3D drug
releasing scaffolds which can mimic ECM and can also restore the altered
microenvironment [140, 141]. Scaffolds were found not only to preserve and
5 Biologically Inspired Materials in Tissue Engineering 123
to obtain good control over drug release profile by simply varying scaffold prop-
erties such as pore size or swelling ratio, which can be easily modulated by
changing scaffold hydrophilicity, crosslinker molecular weight or the extent of
crosslinking [170, 171]. For instance, an increase in pore size can favour cell
infiltration and proteolytic degradation by immune cells, which in turn accelerates
drug release; also, an increase of scaffold hydrophilicity can enhance drug delivery
by accelerating bulk degradation via hydrolysis [102]; for example, the degrad-
ability of PEG hydrogels can be controlled by varying the length of hydrolytically
degradable units within the polymer crosslink [172].
However, a scaffold with controlled biodegradation must degrade into non-toxic
products [173], and the method chosen to control the degradation rate should not
affect the mechanical properties, in particular the stiffness, of the scaffold. A good
example is the approach proposed by Kong and colleagues that aimed at acceler-
ating the degradation rate of alginate hydrogels without affecting their stiffness.
Alginate degradation can be accelerated by increasing its oxidation degree; how-
ever, it also results in softer gels. To overcome this issue, they adjusted the
molecular weight distribution (MWD) of oxidised alginates without varying the
number of oxidised uronic acids, thus achieving both regulation of the degradation
rate and maintenance of the elastic modulus [174].
Nonetheless, changes in the degradation rate of hydrogels can affect their ability
to retain and transport fluids. This issue is particularly important when hydrogels
are used as cell carrier, which requires controlled pore size to allow the exchange of
nutrients and the release of their paracrine products. Kang et al. [175] developed a
3D hybrid scaffold in which a 3D framework and a hydrogel were combined to
enhance the mechanical properties without chemically altering the transport prop-
erties of the hydrogel. This system consisted in the injection of a mixture of
dopamine-releasing cells and an alginate gel into the internal space of a 3D
framework of Ormocomp®. Release studies revealed that the encapsulated cells
were able to secrete dopamine for over 8 weeks in vitro and the introduction of
RGD peptides into the alginate hydrogel increased the release rate. Moreover, the
performance of the system was tested in a mouse model: the subcutaneous
implantation of the hybrid scaffold mixed with encapsulated cells showed no signs
of immune rejection after 7 days, indicating that the system shielded the exogenous
cells from the host immune system while enabling cells to fulfil their paracrine
function [175].
Another innovation in the field of 3D drug-releasing scaffolds is the develop-
ment of supramolecular hydrogels. Conventional hydrogels have a less flexible pore
size due to the presence of covalent bonds, thereby limiting the responsiveness to
the surrounding environment [176]. Supramolecular hydrogels instead are formed
by small molecule hydrogelators that self-assemble into 3D networks of nanofibers.
These hydrogels show the ability to imbibe large amounts of water, and also to
change their pore size as a result of the reassembly of the hydrogelators during the
shrinkage/swelling processes [176]. These properties favour the diffusion of
molecules from the delivery system in response to microenvironmental changes.
Liang et al. tested the use of supramolecular hydrogels for controlled drug release
5 Biologically Inspired Materials in Tissue Engineering 125
The functionalization of the scaffold with cell adhesion molecules was also
found to influence the outcome of gene delivery [187]. For this reason, the first
scaffold that functioned as gene delivery vehicle was made of collagen, a RGD-rich
biomacromolecule [188]. Such collagen scaffolds have been employed for the
delivery of both viral [189] and non-viral vectors [190, 191]. With these approa-
ches, it was shown that it is possible to induce transgene expression in vivo which
led to tissue repair in a wide range of applications such as bone regeneration [188,
192], wound healing [193–196], muscle repair [197] and optic nerve repair [198].
Other natural polymer-based hydrogels were also used as a platform for gene
therapy. For instance, alginate, which is inert to cell adhesion and protein
adsorption [199], was functionalized with RGD peptides. The spatial distribution of
these cell adhesion peptides influenced the rate of DNA uptake by pre-osteoblasts
cells, and RGD-functionalised alginate hydrogels showed the highest transgene
expression [187, 200].
3D scaffolds were also found to prolong the half-life of viral vectors and to
shield them from the host immune response [201]. Furthermore, delivery of the
vector from the scaffold has the advantage of concentrating the transgene expres-
sion in the implant site [143]. The method used to functionalise the scaffold with the
gene vectors can also determine the gene release profile and the duration of their
biological activity [143]. For example, Schek et al. compared in vitro the bioactivity
of adenoviruses embedded in fibrin hydrogels or simply resuspended in liquid form.
When in solution, the adenoviruses decreased to half-maximal activity after only
15 h, whereas when embedded in fibrin hydrogels the half-life of the adenoviruses
was extended to 45 h [201]. The bioactivity of adenoviruses was tested also in
mice. In this study, adenoviruses bearing the gene for BMP-7 were implanted
intramuscularly: the suspended form of adenoviruses induced bone formation in
50% of the muscles, whereas the efficacy of adenoviruses embedded in fibrin
hydrogels was much higher, with bone formation occurring in 80% of the muscles
treated [201]. A drawback in the use of viral vectors for gene therapy is the
potential damage they can cause when they leak into nearby tissues. Pascher et al.
compared the delivery of viral vectors through collagen/glycosaminoglycan
matrices or via bone marrow clots (BMC) in a rabbit model. Interestingly, BMC
showed superior containment of the vector within the osteochondral defects.
Moreover, high levels of transgenic expression and deposition of healthy matrix
were observed within the clots [202]. Another interesting aspect of this approach is
the use of autologous-derived materials that can overcome the issues related to the
manufacturing process and biocompatibility of scaffolds.
To immobilize gene vectors in biomaterials, a number of techniques mimicking
the binding of viruses to ECM proteins have been developed. For instance, by using
the electrostatic interactions that occur between nucleotides and calcium phosphate
(CaP), plasmids (pDNA) can be complexed into a nanocrystalline form and
immobilized onto a surface [203]. These complexes have been extensively used as
gene vectors for the functionalisation of scaffolds for bone tissue engineering [204,
205].
5 Biologically Inspired Materials in Tissue Engineering 127
There are alternatives to viral vectors for gene therapy. Cationic polymers or
lipids are often used as complexing agents to condense pDNA to form small
particles. These complexes can penetrate the cell membrane and deliver the genetic
material intracellularly. Nonetheless, this approach has lower efficacy than viral
vectors and higher concentrations of complexes are generally required to obtain
substantial biological effects. This can often result in significant toxicity in vitro and
in vivo [206]. The use of reservoir systems, such as microspheres and nanoparticles,
in collagen hydrogels was found to significantly decrease the toxicity of complexed
pDNA [191]. Generally, pDNA delivery is more effective in actively dividing cells
as during mitosis the nucleus disassembles temporarily, facilitating the nuclear
localisation of the transgene in the newly formed cells. However, tissues that most
often are the target of gene therapy are harsh environments in which cell prolif-
eration is often impaired. For this reason, recent research is shifting toward mRNA
delivery, as mRNA does not need to enter the nucleus to be expressed, and
moreover, its structure can be modified to considerably increase its stability and
therapeutic efficacy [207].
Overall, advances in stem cell biology and biomaterial science provide exciting
opportunities for tissue engineering. Biomimetic scaffolds can be rationally
designed to take advantage of the body’s inherent healing capacity and, by incor-
porating appropriate signals, to use the body as a bioreactor and guide the healing
process. However, a more in-depth knowledge of the role of ECM in growth factor
binding/release and cell signalling will lead to more effective implants. One of the
major challenges is presenting the appropriate combination of signals in the
appropriate spatial and temporal patterns. As discussed above, the release pattern
can be controlled by modulating the degradation rate and structure of the scaffold.
Other options are the use of supramolecular hydrogels, which provide a rapid and
uniform release, or the covalent bond of growth factors, which allows a gradual
release. As growth factors present a very short half-life, the incorporation into 3D
scaffolds of genetically engineered cells or gene vectors could offer a prolonged
production in situ. However, improvements in transfection efficiency and bio-
compatibility of non-viral gene vectors will likely enable a wider adoption of gene
therapy in the clinic. Furthermore, a successful tissue engineering strategy will
depend on the development of scaffolds able to influence cell behaviour. Hence, the
integration of on-demand drug delivery systems with instructive matrices is the
future direction of 3D releasing scaffolds in tissue engineering.
Fibrinogen-Based Scaffolds
Fibrinogen and fibrin are attractive polymers for tissue engineering applications
because they can provide a natural environment and they have intrinsic healing
properties.
Fibrinogen is a 340 kDa plasma glycoprotein involved in formation of blood
clots [209]. The molecule is composed of two sets of disulphide-bridged Aa-, Bb-
128 G. Fontana et al.
Table 5.3 Examples of in vitro and in vivo applications of fibrinogen- and fibrin-based scaffolds
Material Formulation Tissue Outcome References
engineering
application
Fibrin/ Hydrogel Spinal cord repair The fibrin/fibrinogen [229]
fibrinogen hydrogel integrated with
the host spinal cord tissue
and supported robust
growth of axon
Fibrin Hydrogel Spinal cord repair Implants of fibrin [236]
containing neurotrophic
factors enhanced axonal
regeneration into
transected rat spinal cord
Fibrin Hydrogel Spinal cord repair Implants of fibrin and [237]
crosslinked MSCs resulted in the
with EDC formation of
longitudinally-aligned
neurites
Platelet-rich Glue Bone repair Glue mixed with MSCs [238]
fibrin and BMP-2 induced bone
tissue formation in dental
implants in dogs
Fibrin Hydrogel Cartilage repair A significant [241]
improvement in knee
function was recorded in
four patients implanted
with fibrin glue and
autologous chondrocytes.
The graft well integrated
with host tissue
Fibrin Glue Wound healing Fibrin glue combined [259]
with fibroblasts and
growth factors enhanced
wound healing in rabbits
Fibrin Glue Wound healing In vitro, human umbilical [247]
and vein endothelial cells
vasculogenesis proliferated and
organised themselves
into capillary-like
structures within the
fibrin matrix. The skin
substitute showed similar
structure to native skin
Fibrinogen Electrospun Wound healing In an in vitro model, [249]
scaffolds fibroblasts migrated into
and remodelled the
electrospun fibrinogen
scaffold with deposition
of collagen
(continued)
130 G. Fontana et al.
strength and rapid degradation. Indeed, fibrin and fibrinogen scaffolds lose their
structure within a few days through degradation by proteases in vitro and in vivo
[209]. To overcome these limitations, fibrin and fibrinogen hydrogels can be
(1) blended with synthetic or natural polymers, (2) crosslinked with different
chemical and biological agents or (3) functionalized with plasmin inhibitors.
Examples of materials used to achieve adequate mechanical strength are poly-
urethane (PU), poly(e-caprolactone) (PCL) and polyethylene glycol (PEG). For
instance, a composite construct made from the commercially available fibrin and
PU-based scaffold exhibited increased stability in vitro and in vivo as compared to
fibrin gels alone: in vitro, the fibrin/PU scaffold was seeded with human adipose-
derived stem cells and was shown to be able to maintain its size and weight for
21 days; in vivo, the implantation of fibrin/PU construct resulted in formation of
well-vascularized adipose tissue [221]. The same research group used the fibrin/PU
scaffold also for cartilage tissue engineering applications [222].
5 Biologically Inspired Materials in Tissue Engineering 131
References
1. Lim EH, Sardinha JP, Myers S. Nanotechnology biomimetic cartilage regenerative scaffolds.
Arch plastic Surg. 2014;41:231–40.
2. Abbah SA, Delgado LM, Azeem A, Fuller K, Shologu N, Keeney M, et al. Harnessing
hierarchical nano- and micro-fabrication technologies for musculoskeletal tissue engineer-
ing. Adv Healthc Mater. 2015;4:2488–99.
3. Fuller K, Pandit A, Zeugolis DI. The multifaceted potential of electro-spinning in
regenerative medicine. Pharm Nanotechnol. 2014;2:23–34.
4. Carbone EJ, Jiang T, Nelson C, Henry N, Lo KWH. Small molecule delivery through
nanofibrous scaffolds for musculoskeletal regenerative engineering. Nanomed Nanotechnol
Biol Med. 2014;10:1691–9.
5. Woo KM, Chen VJ, Ma PX. Nano-fibrous scaffolding architecture selectively enhances
protein adsorption contributing to cell attachment. J Biomed Mater Res A. 2003;67A:531–7.
6. Ma PX. Biomimetic materials for tissue engineering. Adv Drug Deliv Rev. 2008;60:184–98.
7. Barnes CP, Sell SA, Boland ED, Simpson DG, Bowlin GL. Nanofiber technology: designing
the next generation of tissue engineering scaffolds. Adv Drug Deliv Rev. 2007;59:1413–33.
8. Shields KJ, Beckman MJ, Bowlin GL, Wayne JS. Mechanical properties and cellular
proliferation of electrospun collagen type II. Tissue Eng. 2004;10:1510–7.
9. Heydarkhan-Hagvall S, Schenke-Layland K, Dhanasopon AP, Rofail F, Smith H, Wu BM,
et al. Three-dimensional electrospun ECM-based hybrid scaffolds for cardiovascular tissue
engineering. Biomaterials. 2008;29:2907–14.
10. Fleischer S, Shapira A, Regev O, Nseir N, Zussman E, Dvir T. Albumin fiber scaffolds for
engineering functional cardiac tissues. Biotechnol Bioeng. 2014;111:1246–57.
11. Nicodemus GD, Bryant SJ. Cell encapsulation in biodegradable hydrogels for tissue
engineering applications. Tissue Eng Part B Rev. 2008;14:149–65.
136 G. Fontana et al.
12. Hong Y, Takanari K, Amoroso NJ, Hashizume R, Brennan-Pierce EP, Freund JM, et al.
An elastomeric patch electrospun from a blended solution of dermal extracellular matrix and
biodegradable polyurethane for rat abdominal wall repair. Tissue Eng Part C Methods.
2012;18:122–32.
13. Harkness RD. Biological functions of collagen. Biol Rev Camb Philos Soc. 1961;36:399–463.
14. Soderhall C, Marenholz I, Kerscher T, Ruschendorf F, Esparza-Gordillo J, Worm M, et al.
Variants in a novel epidermal collagen gene (COL29A1) are associated with atopic
dermatitis. PLoS Biol. 2007;5:e242.
15. Kielty CM, Grant ME. The collagen family: structure, assembly and organization in
the extracellular matrix. In: Royce PM, Steinmann B, editors. Connective tissue and its
heritable disorders: molecular, genetic and medical aspects. 2nd ed. New York: Wiley; 2002.
p. 159–221.
16. Piez KA. Collagen. In: Kroschwitz JI, editor. Encyclopedia of polymer science and
engineering. New York: Wiley; 1985. p. 699–727.
17. Zeugolis DI, Raghunath M. Collagen: materials analysis and implant uses. In: Ducheyne P,
Healy KE, Hutmacher DW, Grainger DW, Kirkpatrick CJ, editors. Comprehensive
biomaterials. Oxford: Elsevier; 2011. p. 261–78.
18. Friess W. Collagen—biomaterial for drug delivery. Eur J Pharm Biopharm. 1998;45:113–36.
19. Zeugolis DI, Paul GR, Attenburrow G. Cross-linking of extruded collagen fibres—a
biomimetic three-dimensional scaffold for tissue engineering applications. J Biomed Mater
Res A. 2009;89:895–908.
20. Zeugolis DI, Paul RG, Attenburrow G. The influence of a natural cross-linking agent
(Myrica rubra) on the properties of extruded collagen fibres for tissue engineering
applications. Mater Sci Eng C. 2010;30:190–5.
21. Delgado LM, Fuller K, Zeugolis DI. Collagen cross-linking: biophysical, biochemical, and
biological response analysis. Tissue Eng Part A. 2017;23:1064–77.
22. Gough JE, Scotchford CA, Downes S. Cytotoxicity of glutaraldehyde crosslinked collagen/
poly(vinyl alcohol) films is by the mechanism of apoptosis. J Biomed Mater Res.
2002;61:121–30.
23. Levy RJ, Schoen FJ, Sherman FS, Nichols J, Hawley MA, Lund SA. Calcification of
subcutaneously implanted type I collagen sponges. Effects of formaldehyde and glutaralde-
hyde pretreatments. Am J Pathol. 1986;122:71–82.
24. Brown BN, Londono R, Tottey S, Zhang L, Kukla KA, Wolf MT, et al. Macrophage
phenotype as a predictor of constructive remodeling following the implantation of
biologically derived surgical mesh materials. Acta Biomater. 2012;8:978–87.
25. Delgado LM, Bayon Y, Pandit A, Zeugolis DI. To cross-link or not to cross-link?
Cross-linking associated foreign body response of collagen-based devices. Tissue Eng Part B
Rev. 2015;21:298–313.
26. Collin EC, Grad S, Zeugolis DI, Vinatier CS, Clouet JR, Guicheux JJ, et al. An injectable
vehicle for nucleus pulposus cell-based therapy. Biomaterials. 2011;32:2862–70.
27. Rho KS, Jeong L, Lee G, Seo BM, Park YJ, Hong SD, et al. Electrospinning of collagen
nanofibers: effects on the behavior of normal human keratinocytes and early-stage wound
healing. Biomaterials. 2006;27:1452–61.
28. Li M, Mondrinos MJ, Gandhi MR, Ko FK, Weiss AS, Lelkes PI. Electrospun protein fibers
as matrices for tissue engineering. Biomaterials. 2005;26:5999–6008.
29. Zeugolis DI, Khew ST, Yew ESY, Ekaputra AK, Tong YW, Yung L-YL, et al.
Electro-spinning of pure collagen nano-fibres—just an expensive way to make gelatin?
Biomaterials. 2008;29:2293–305.
30. Browne S, Zeugolis DI, Pandit A. Collagen: finding a solution for the source. Tissue Eng
Part A. 2013;19:1491–4.
31. IAEA. Trends in radiation sterilization of health care products. Vienna: International Atomic
Energy Agency; 2008.
5 Biologically Inspired Materials in Tissue Engineering 137
32. Hori H, Hattori S, Inouye S, Kimura A, Irie S, Miyazawa H, et al. Analysis of the major
epitope of the a2 chain of bovine type I collagen in children with bovine gelatin allergy.
J Allergy Clin Immunol. 2002;110:652–7.
33. Lynn AK, Yannas IV, Bonfield W. Antigenicity and immunogenicity of collagen. J Biomed
Mater Res Part B Appl Biomater. 2004;71:343–54.
34. Delgado LM, Shologu N, Fuller K, Zeugolis D. Acetic acid and pepsin result in high yield,
high purity and low macrophage response collagen for biomedical applications. Biomed
Mater. 2017;12:065009.
35. Tang Y, Yang X, Hang B, Li J, Huang L, Huang F, et al. Efficient production of
hydroxylated human-like collagen via the co-expression of three key genes in escherichia
coli origami (DE3). Appl Biochem Biotechnol. 2016;178:1458–70.
36. Ruggiero F, Koch M. Making recombinant extracellular matrix proteins. Methods
(San Diego, Calif). 2008;45:75–85.
37. Yang C, Hillas PJ, Baez JA, Nokelainen M, Balan J, Tang J, et al. The application of
recombinant human collagen in tissue engineering. BioDrugs Clin Immunother Biopharm
Gene Ther. 2004;18:103–19.
38. Parenteau-Bareil R, Gauvin R, Berthod F. Collagen-based biomaterials for tissue engineer-
ing applications. Materials. 2010;3:1863–87.
39. Gosline J, Lillie M, Carrington E, Guerette P, Ortlepp C, Savage K. Elastic proteins:
biological roles and mechanical properties. Philos Trans R Soc Lond B Biol Sci.
2002;357:121–32.
40. Silva R, Fabry B, Boccaccini AR. Fibrous protein-based hydrogels for cell encapsulation.
Biomaterials. 2014;35:6727–38.
41. Miranda-Nieves D, Chaikof EL. Collagen and elastin biomaterials for the fabrication of
engineered living tissues. ACS Biomater Sci Eng. 2017;3:694–711.
42. Daamen WF, Veerkamp JH, van Hest JC, van Kuppevelt TH. Elastin as a biomaterial for
tissue engineering. Biomaterials. 2007;28:4378–98.
43. Werkmeister JA, Ramshaw JA. Recombinant protein scaffolds for tissue engineering.
Biomed Mater. 2012;7:012002.
44. Caves JM, Cui W, Wen J, Kumar VA, Haller CA, Chaikof EL. Elastin-like protein matrix
reinforced with collagen microfibers for soft tissue repair. Biomaterials. 2011;32:5371–9.
45. Devalliere J, Dooley K, Hu Y, Kelangi SS, Uygun BE, Yarmush ML. Co-delivery of a
growth factor and a tissue-protective molecule using elastin biopolymers accelerates wound
healing in diabetic mice. Biomaterials. 2017;141:149–60.
46. Monfort DA, Koria P. Recombinant elastin based nanoparticles for targeted gene therapy.
Gene Ther. 2017;24:610–20.
47. Waterhouse A, Wise SG, Ng MK, Weiss AS. Elastin as a nonthrombogenic biomaterial.
Tissue Eng Part B Rev. 2011;17:93–9.
48. Liu SQ, Tieche C, Alkema PK. Neointima formation on vascular elastic laminae and
collagen matrices scaffolds implanted in the rat aortae. Biomaterials. 2004;25:1869–82.
49. Henry JJD, Yu J, Wang A, Lee R, Fang J, Li S. Engineering the mechanical and biological
properties of nanofibrous vascular grafts for in situ vascular tissue engineering.
Biofabrication. 2017;9:035007.
50. Waterhouse A, Yin Y, Wise SG, Bax DV, McKenzie DR, Bilek MM, et al. The
immobilization of recombinant human tropoelastin on metals using a plasma-activated
coating to improve the biocompatibility of coronary stents. Biomaterials. 2010;31:8332–40.
51. Woodhouse KA, Klement P, Chen V, Gorbet MB, Keeley FW, Stahl R, et al. Investigation
of recombinant human elastin polypeptides as non-thrombogenic coatings. Biomaterials.
2004;25:4543–53.
52. Jordan SW, Haller CA, Sallach RE, Apkarian RP, Hanson SR, Chaikof EL. The effect of a
recombinant elastin-mimetic coating of an ePTFE prosthesis on acute thrombogenicity in a
baboon arteriovenous shunt. Biomaterials. 2007;28:1191–7.
138 G. Fontana et al.
53. Wachi H, Sugitani H, Murata H, Nakazawa J, Mecham RP, Seyama Y. Tropoelastin inhibits
vascular calcification via 67-kDa elastin binding protein in cultured bovine aortic smooth
muscle cells. J Atherosclerosis Thromb. 2004;11:159–66.
54. Almine JF, Bax DV, Mithieux SM, Nivison-Smith L, Rnjak J, Waterhouse A, et al.
Elastin-based materials. Chem Soc Rev. 2010;39:3371–9.
55. Wong CS, Liu X, Xu Z, Lin T, Wang X. Elastin and collagen enhances electrospun aligned
polyurethane as scaffolds for vascular graft. J Mater Sci Mater Med. 2013;24:1865–74.
56. McClure MJ, Sell SA, Simpson DG, Walpoth BH, Bowlin GL. A three-layered electrospun
matrix to mimic native arterial architecture using polycaprolactone, elastin, and collagen: a
preliminary study. Acta Biomater. 2010;6:2422–33.
57. Foraida ZI, Kamaldinov T, Nelson DA, Larsen M, Castracane J. Elastin-PLGA hybrid
electrospun nanofiber scaffolds for salivary epithelial cell self-organization and polarization.
Acta Biomater. 2017;62:116–27.
58. Feughelman M. Natural protein fibers. J Appl Polym Sci. 2002;83:489–507.
59. Shavandi A, Silva TH, Bekhit AA, Bekhit AEA. Keratin: dissolution, extraction and
biomedical application. Biomater Sci. 2017;5:1699–735.
60. Rouse JG, Van Dyke ME. A review of keratin-based biomaterials for biomedical
applications. Materials. 2010;3:999–1014.
61. Sierpinski P, Garrett J, Ma J, Apel P, Klorig D, Smith T, et al. The use of keratin
biomaterials derived from human hair for the promotion of rapid regeneration of peripheral
nerves. Biomaterials. 2008;29:118–28.
62. Ham TR, Lee RT, Han S, Haque S, Vodovotz Y, Gu J, et al. Tunable keratin hydrogels for
controlled erosion and growth factor delivery. Biomacromol. 2016;17:225–36.
63. Pace LA, Plate JF, Mannava S, Barnwell JC, Koman LA, Li Z, et al. A human hair keratin
hydrogel scaffold enhances median nerve regeneration in nonhuman primates: an
electrophysiological and histological study. Tissue Eng Part A. 2014;20:507–17.
64. Blanchard CR, Timmons SF, Smith RA. Keratin-based hydrogel for biomedical applications
and method of production. 1999.
65. Kaplan D, Adams WW, Farmer B, Viney C. Silk: biology, structure, properties, and
genetics. Silk Polymers: American Chemical Society; 1993. p. 2–16.
66. Kasoju N, Bora U. Silk fibroin in tissue engineering. Adv Healthc Mater. 2012;1:393–412.
67. Zhou CZ, Confalonieri F, Jacquet M, Perasso R, Li ZG, Janin J. Silk fibroin: structural
implications of a remarkable amino acid sequence. Proteins. 2001;44:119–22.
68. Rockwood DN, Preda RC, Yucel T, Wang X, Lovett ML, Kaplan DL. Materials fabrication
from Bombyx mori silk fibroin. Nat Protoc. 2011;6:1612–31.
69. Kundu B, Rajkhowa R, Kundu SC, Wang X. Silk fibroin biomaterials for tissue
regenerations. Adv Drug Deliv Rev. 2013;65:457–70.
70. Wang Y, Rudym DD, Walsh A, Abrahamsen L, Kim H-J, Kim HS, et al. In vivo degradation
of three-dimensional silk fibroin scaffolds. Biomaterials. 2008;29:3415–28.
71. Lovett ML, Cannizzaro CM, Vunjak-Novakovic G, Kaplan DL. Gel spinning of silk tubes
for tissue engineering. Biomaterials. 2008;29:4650–7.
72. Zhang X, Wang X, Keshav V, Wang X, Johanas JT, Leisk GG, et al. Dynamic culture
conditions to generate silk-based tissue-engineered vascular grafts. Biomaterials.
2009;30:3213–23.
73. Zamani M, Khafaji M, Naji M, Vossoughi M, Alemzadeh I, Haghighipour N. A biomimetic
heparinized composite silk-based vascular scaffold with sustained antithrombogenicity. Sci
Rep. 2017;7:4455.
74. Cattaneo I, Figliuzzi M, Azzollini N, Catto V, Fare S, Tanzi MC, et al. In vivo regeneration
of elastic lamina on fibroin biodegradable vascular scaffold. Int J Artif Organs. 2013;36:
166–74.
75. Zhang X, Reagan MR, Kaplan DL. Electrospun silk biomaterial scaffolds for regenerative
medicine. Adv Drug Delivery Rev. 2009;61:988–1006.
5 Biologically Inspired Materials in Tissue Engineering 139
76. Song DW, Kim SH, Kim HH, Lee KH, Ki CS, Park YH. Multi-biofunction of antimicrobial
peptide-immobilized silk fibroin nanofiber membrane: Implications for wound healing. Acta
Biomater. 2016;39:146–55.
77. Gil ES, Panilaitis B, Bellas E, Kaplan DL. Functionalized silk biomaterials for wound
healing. Adv Healthc Mater. 2013;2:206–17.
78. Liu W, Thomopoulos S, Xia Y. Electrospun nanofibers for regenerative medicine. Adv
Healthc Mater. 2012;1:10–25.
79. Binan L, Tendey C, De Crescenzo G, El Ayoubi R, Ajji A, Jolicoeur M. Differentiation of
neuronal stem cells into motor neurons using electrospun poly-L-lactic acid/gelatin scaffold.
Biomaterials. 2014;35:664–74.
80. Liu S, Qin M, Hu C, Wu F, Cui W, Jin T, et al. Tendon healing and anti-adhesion properties
of electrospun fibrous membranes containing bFGF loaded nanoparticles. Biomaterials.
2013;34:4690–701.
81. Ortega I, Ryan AJ, Deshpande P, MacNeil S, Claeyssens F. Combined microfabrication and
electrospinning to produce 3-D architectures for corneal repair. Acta Biomater.
2013;9:5511–20.
82. Hu X, Liu S, Zhou G, Huang Y, Xie Z, Jing X. Electrospinning of polymeric nanofibers for
drug delivery applications. J Controlled Release. 2014;185:12–21.
83. Katti DS, Robinson KW, Ko FK, Laurencin CT. Bioresorbable nanofiber-based systems for
wound healing and drug delivery: optimization of fabrication parameters. J Biomed Mater
Res Part B Appl Biomater. 2004;70:286–96.
84. Cramariuc B, Cramariuc R, Scarlet R, Manea LR, Lupu IG, Cramariuc O. Fiber diameter in
electrospinning process. J Electrostat. 2013;71:189–98.
85. Fuller KP, Gaspar D, Delgado LM, Pandit A, Zeugolis DI. Influence of porosity and pore
shape on structural, mechanical and biological properties of poly -caprolactone electro-spun
fibrous scaffolds. Nanomedicine (London, England). 2016;11:1031–40.
86. Yin Z, Chen X, Chen JL, Shen WL, Hieu Nguyen TM, Gao L, et al. The regulation
of tendon stem cell differentiation by the alignment of nanofibers. Biomaterials.
2010;31:2163–75.
87. Lee BK, Ju YM, Cho JG, Jackson JD, Lee SJ, Atala A, et al. End-to-side neurorrhaphy using
an electrospun PCL/collagen nerve conduit for complex peripheral motor nerve regeneration.
Biomaterials. 2012;33:9027–36.
88. Xie J, MacEwan M, Ray W, Liu W, Siewe D, Xia Y. Radially aligned, electrospun
nanofibers as dural substitutes for wound closure and tissue regeneration applications. ACS
Nano. 2010;4:5027–36.
89. Whitesides GM, Grzybowski B. Self-assembly at all scales. Science (New York, NY).
2002;295:2418–21.
90. Friess W. Collagen–biomaterial for drug delivery. Eur J Pharm Biopharm. 1998;45:113–36.
91. Zeugolis DI, Paul RG, Attenburrow G. Extruded collagen-polyethylene glycol fibers
for tissue engineering applications. J Biomed Mater Res Part B Appl Biomater. 2008;85:
343–52.
92. Daly WT, Yao L, Abu-rub MT, O’Connell C, Zeugolis DI, Windebank AJ, et al. The effect
of intraluminal contact mediated guidance signals on axonal mismatch during peripheral
nerve repair. Biomaterials. 2012;33:6660–71.
93. Abu-Rub MT, Billiar KL, van Es MH, Knight A, Rodriguez BJ, Zeugolis DI, et al.
Nano-textured self-assembled aligned collagen hydrogels promote directional neurite
guidance and overcome inhibition by myelin associated glycoprotein. Soft Matter.
2011;7:2770.
94. Li Y, Asadi A, Monroe MR, Douglas EP. pH effects on collagen fibrillogenesis in vitro:
electrostatic interactions and phosphate binding. Mater Sci Eng C. 2009;29:1643–9.
95. Olmos Buitrago J, Perez RA, El-Fiqi A, Singh RK, Kim JH, Kim HW. Core-shell fibrous
stem cell carriers incorporating osteogenic nanoparticulate cues for bone tissue engineering.
Acta Biomater. 2015;28:183–92.
140 G. Fontana et al.
96. Perez RA, Kim HW. Core-shell designed scaffolds for drug delivery and tissue engineering.
Acta Biomater. 2015;21:2–19.
97. Perez RA, Kim JH, Buitrago JO, Wall IB, Kim HW. Novel therapeutic core-shell hydrogel
scaffolds with sequential delivery of cobalt and bone morphogenetic protein-2 for synergistic
bone regeneration. Acta Biomater. 2015;23:295–308.
98. Perez RA, Kim M, Kim TH, Kim JH, Lee JH, Park JH, et al. Utilizing core-shell fibrous
collagen-alginate hydrogel cell delivery system for bone tissue engineering. Tissue Eng
Part A. 2014;20:103–14.
99. Hartgerink JD, Beniash E, Stupp SI. Peptide-amphiphile nanofibers: a versatile scaffold for
the preparation of self-assembling materials. Proc Natl Acad Sci U S A. 2002;99:5133–8.
100. Hartgerink JD, Beniash E, Stupp SI. Self-assembly and mineralization of peptide-amphiphile
nanofibers. Science (New York, NY). 2001;294:1684–8.
101. Loo Y, Zhang SG, Hauser CAE. From short peptides to nanofibers to macromolecular
assemblies in biomedicine. Biotechnol Adv. 2012;30:593–603.
102. Saracino GA, Cigognini D, Silva D, Caprini A, Gelain F. Nanomaterials design and tests for
neural tissue engineering. Chem Soc Rev. 2013;42:225–62.
103. Cunha C, Panseri S, Villa O, Silva D, Gelain F. 3D culture of adult mouse neural stem cells
within functionalized self-assembling peptide scaffolds. Int J Nanomed. 2011;6:943–55.
104. Yang H, Xie Z, Wang P, Bi J. Self-assembling nanofibers alter the processing of amyloid
precursor protein in a transgenic mouse model of Alzheimer’s disease. Neurol Res.
2015;37:84–91.
105. Cigognini D, Satta A, Colleoni B, Silva D, Donega M, Antonini S, et al. Evaluation of early
and late effects into the acute spinal cord injury of an injectable functionalized
self-assembling scaffold. PLoS ONE. 2011;6:e19782.
106. Cigognini D, Silva D, Paloppi S, Gelain F. Evaluation of mechanical properties and
therapeutic effect of injectable self-assembling hydrogels for spinal cord injury. J Biomed
Nanotechnol. 2014;10:309–23.
107. Kisiday J, Jin M, Kurz B, Hung H, Semino C, Zhang S, et al. Self-assembling peptide
hydrogel fosters chondrocyte extracellular matrix production and cell division: implications
for cartilage tissue repair. Proc Natl Acad Sci U S A. 2002;99:9996–10001.
108. Schneider A, Garlick JA, Egles C. Self-assembling peptide nanofiber scaffolds accelerate
wound healing. PLoS ONE. 2008;3:e1410.
109. Ma PX, Zhang R. Synthetic nano-scale fibrous extracellular matrix. J Biomed Mater Res.
1999;46:60–72.
110. Stokols S, Sakamoto J, Breckon C, Holt T, Weiss J, Tuszynski MH. Templated agarose
scaffolds support linear axonal regeneration. Tissue Eng. 2006;12:2777–87.
111. Stokols S, Tuszynski MH. The fabrication and characterization of linearly oriented nerve
guidance scaffolds for spinal cord injury. Biomaterials. 2004;25:5839–46.
112. Garcia Y, Wilkins B, Collighan RJ, Griffin M, Pandit A. Towards development of a dermal
rudiment for enhanced wound healing response. Biomaterials. 2008;29:857–68.
113. Stokols S, Tuszynski MH. Freeze-dried agarose scaffolds with uniaxial channels stimulate
and guide linear axonal growth following spinal cord injury. Biomaterials. 2006;27:443–51.
114. Bozkurt A, Lassner F, O’Dey D, Deumens R, Bocker A, Schwendt T, et al. The role of
microstructured and interconnected pore channels in a collagen-based nerve guide on axonal
regeneration in peripheral nerves. Biomaterials. 2012;33:1363–75.
115. Chen H, Yang X, Liao Y, Zeng X, Liang P, Kang N, et al. MRI and histologic analysis of
collagen type II sponge on repairing the cartilage defects of rabbit knee joints. J Biomed
Mater Res Part B Appl Biomater. 2011;96:267–75.
116. Shen W, Chen X, Hu Y, Yin Z, Zhu T, Hu J, et al. Long-term effects of knitted silk–collagen
sponge scaffold on anterior cruciate ligament reconstruction and osteoarthritis prevention.
Biomaterials. 2014;35:8154–63.
117. Yao L, Daly W, Newland B, Yao S, Wang W, Chen BK, et al. Improved axonal regeneration
of transected spinal cord mediated by multichannel collagen conduits functionalized with
neurotrophin-3 gene. Gene Ther. 2013;20:1149–57.
5 Biologically Inspired Materials in Tissue Engineering 141
118. Caliari SR. Harley BaC. The effect of anisotropic collagen-GAG scaffolds and growth factor
supplementation on tendon cell recruitment, alignment, and metabolic activity. Biomaterials.
2011;32:5330–40.
119. Caliari SR, Ramirez MA, Harley BAC. The development of collagen-GAG
scaffold-membrane composites for tendon tissue engineering. Biomaterials. 2011;32:8990–8.
120. Boekema BK, Vlig M, Olde Damink L, Middelkoop E, Eummelen L, Buhren AV, et al.
Effect of pore size and cross-linking of a novel collagen-elastin dermal substitute on wound
healing. J Mater Sci Mater Med. 2014;25:423–33.
121. Min BM, Jeong L, Nam YS, Kim JM, Kim JY, Park WH. Formation of silk fibroin matrices
with different texture and its cellular response to normal human keratinocytes. Int J Biol
Macromol. 2004;34:281–8.
122. Daley WP, Peters SB, Larsen M. Extracellular matrix dynamics in development and
regenerative medicine. J Cell Sci. 2008;121:255–64.
123. Geiger B, Bershadsky A, Pankov R, Yamada KM. Transmembrane crosstalk between the
extracellular matrix–cytoskeleton crosstalk. Nat Rev Mol Cell Biol. 2001;2:793–805.
124. Kong HJ, Mooney DJ. Microenvironmental regulation of biomacromolecular therapies. Nat
Rev Drug Discovery. 2007;6:455–63.
125. Aszodi A, Hunziker EB, Brakebusch C, Fassler R. Beta1 integrins regulate chondrocyte
rotation, G1 progression, and cytokinesis. Genes Dev. 2003;17:2465–79.
126. Schneiderbauer MM, Dutton CM, Scully SP. Signaling “cross-talk” between TGF-beta1 and
ECM signals in chondrocytic cells. Cell Signal. 2004;16:1133–40.
127. Fontana G, See E, Pandit A. Current trends in biologics delivery to restore intervertebral disc
anabolism. Adv Drug Deliv Rev. 2015;84:146–58.
128. Teti A. Regulation of Cellular Functions by Extracellular-Matrix. J Am Soc Nephrol.
1992;2:S83–7.
129. Kim SH, Turnbull J, Guimond S. Extracellular matrix and cell signalling: the dynamic
cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol.
2011;209:139–51.
130. Bosnakovski D, Mizuno M, Kim G, Takagi S, Okumura M, Fujinaga T. Chondrogenic
differentiation of bovine bone marrow mesenchymal stem cells (MSCs) in different
hydrogels: influence of collagen type II extracellular matrix on MSC chondrogenesis.
Biotechnol Bioeng. 2006;93:1152–63.
131. DeLise AM, Fischer L, Tuan RS. Cellular interactions and signaling in cartilage
development. Osteoarthritis Cartilage. 2000;8:309–34.
132. Masuda K, An HS. Growth factors and the intervertebral disc. Spine J. 2004;4:330S–40S.
133. Masuda K, Oegema TR, Jr., An HS. Growth factors and treatment of intervertebral disc
degeneration. Spine (Phila Pa 1976). 2004;29:2757–69.
134. Masuda K, Lotz JC. New challenges for intervertebral disc treatment using regenerative
medicine. Tissue Eng Part B Rev. 2010;16:147–58.
135. Eppler SM, Combs DL, Henry TD, Lopez JJ, Ellis SG, Yi JH, et al. A target-mediated model
to describe the pharmacokinetics and hemodynamic effects of recombinant human vascular
endothelial growth factor in humans. Clin Pharmacol Ther. 2002;72:20–32.
136. Lee K, Silva EA, Mooney DJ. Growth factor delivery-based tissue engineering: general
approaches and a review of recent developments. J R Soc Interface. 2011;8:153–70.
137. Saik JE, Gould DJ, Watkins EM, Dickinson ME, West JL. Covalently immobilized
platelet-derived growth factor-BB promotes angiogenesis in biomimetic poly(ethylene
glycol) hydrogels. Acta Biomater. 2011;7:133–43.
138. Carmeliet P. Mechanisms of angiogenesis and arteriogenesis. Nat Med. 2000;6:389–95.
139. Lee RJ, Springer ML, Blanco-Bose WE, Shaw R, Ursell PC, Blau HM. VEGF gene delivery
to myocardium—deleterious effects of unregulated expression. Circulation. 2000;102:
898–901.
140. Yang WW, Pierstorff E. Reservoir-based polymer drug delivery systems. Jala-J Lab Autom.
2012;17:50–8.
142 G. Fontana et al.
162. Markowicz M, Heitland A, Steffens GCM, Pallua N. Effects of modified collagen matrices
on human umbilical vein endothelial cells. Int J Artif Organs. 2005;28:1251–8.
163. Steffens GCM, Yao C, Prevel P, Markowicz M, Schenck P, Noah EM, et al. Modulation of
angiogenic potential of collagen matrices by covalent incorporation of heparin and loading
with vascular endothelial growth factor. Tissue Eng. 2004;10:1502–9.
164. van Wachem PB, Plantinga JA, Wissink MJB, Beernink R, Poot AA, Engbers GHM, et al.
In vivo biocompatibility of carbodiimide-crosslinked collagen matrices: Effects of crosslink
density, heparin immobilization, and bFGF loading. J Biomed Mater Res. 2001;55:368–78.
165. Wissink MJB, Beernink R, Pieper JS, Poot AA, Engbers GHM, Beugeling T, et al. Binding
and release of basic fibroblast growth factor from heparinized collagen matrices.
Biomaterials. 2001;22:2291–9.
166. Bladergroen BA, Siebum B, Siebers-Vermeulen KGC, Van Kuppevelt TH, Poot AA,
Feijen J, et al. In vivo recruitment of hematopoietic cells using stromal cell-derived factor 1
alpha-loaded heparinized three-dimensional collagen scaffolds. Tissue Eng Part A.
2009;15:1591–9.
167. Lienemann PS, Lutolf MP, Ehrbar M. Biomimetic hydrogels for controlled biomolecule
delivery to augment bone regeneration. Adv Drug Deliver Rev. 2012;64:1078–89.
168. Uebersax L, Merkle HP, Meinel L. Biopolymer-based growth factor delivery for tissue
repair: from natural concepts to engineered systems. Tissue Eng Part B Rev. 2009;15:
263–89.
169. Lin CC, Anseth KS. Controlling affinity binding with peptide-functionalized poly(ethylene
glycol) hydrogels. Adv Funct Mater. 2009;19:2325–31.
170. Wieland JA, Houchin-Ray TL, Shea LD. Non-viral vector delivery from PEG-hyaluronic
acid hydrogels. J Controlled Release. 2007;120:233–41.
171. Kasper FK, Seidlits SK, Tang A, Crowther RS, Carney DH, Barry MA, et al. In vitro release
of plasmid DNA from oligo(poly(ethylene glycol) fumarate) hydrogels. J Controlled
Release. 2005;104:521–39.
172. Bjugstad KB, Lampe K, Kern DS, Mahoney M. Biocompatibility of poly(ethylene glycol)-
based hydrogels in the brain: an analysis of the glial response across space and time.
J Biomed Mater Res Part A. 2010;95A:79–91.
173. Jeon O, Alt DS, Ahmed SM, Alsberg E. The effect of oxidation on the degradation of
photocrosslinkable alginate hydrogels. Biomaterials. 2012;33:3503–14.
174. Kong HJ, Kaigler D, Kim K, Mooney DJ. Controlling rigidity and degradation of alginate
hydrogels via molecular weight distribution. Biomacromol. 2004;5:1720–7.
175. Kang KS, Lee SI, Hong JM, Lee JW, Cho HY, Son JH, et al. Hybrid scaffold composed of
hydrogel/3D-framework and its application as a dopamine delivery system. J Controlled
Release. 2014;175:10–6.
176. Zhou SL, Matsumoto S, Tian HD, Yamane H, Ojida A, Kiyonaka S, et al. pH-Responsive
shrinkage/swelling of a supramolecular hydrogel composed of two small amphiphilic
molecules. Chem Eur J. 2005;11:1130–6.
177. Liang GL, Yang ZM, Zhang RJ, Li LH, Fan YJ, Kuang Y, et al. Supramolecular hydrogel of
a D-amino acid dipeptide for controlled drug release in vivo. Langmuir. 2009;25:8419–22.
178. Alvarez Z, Castano O, Castells AA, Mateos-Timoneda MA, Planell JA, Engel E, et al.
Neurogenesis and vascularization of the damaged brain using a lactate-releasing biomimetic
scaffold. Biomaterials. 2014;35:4769–81.
179. Polet F, Feron O. Endothelial cell metabolism and tumour angiogenesis: glucose and
glutamine as essential fuels and lactate as the driving force. J Intern Med. 2013;273:156–65.
180. Speder P, Liu J, Brand AH. Nutrient control of neural stem cells. Curr Opin Cell Biol.
2011;23:724–9.
181. Wei GB, Jin QM, Giannobile WV, Ma PX. The enhancement of osteogenesis by
nano-fibrous scaffolds incorporating rhBMP-7 nanospheres. Biomaterials. 2007;28:2087–96.
182. Saraf A, Mikos AG. Gene delivery strategies for cartilage tissue engineering. Adv Drug
Deliver Rev. 2006;58:592–603.
144 G. Fontana et al.
183. Shimer AL, Chadderdon RC, Gilbertson LG, Kang JD. Gene therapy approaches for
intervertebral disc degeneration. Spine. 2004;29:2770–8.
184. Woods BI, Vo N, Sowa G, Kang JD. Gene therapy for intervertebral disk degeneration.
Orthop Clin N Am. 2011;42:563–74.
185. Jang JH, Houchin TL, Shea LD. Gene delivery from polymer scaffolds for tissue
engineering. Expert Rev Med Devices. 2004;1:127–38.
186. Riddle KW, Kong HJ, Leach JK, Fischbach C, Cheung C, Anseth KS, et al. Modifying the
proliferative state of target cells to control DNA expression and identifying cell types
transfected in vivo. Mol Ther. 2007;15:361–8.
187. Rowley JA, Mooney DJ. Alginate type and RGD density control myoblast phenotype.
J Biomed Mater Res. 2002;60:217–23.
188. Fang JM, Zhu YY, Smiley E, Bonadio J, Rouleau JP, Goldstein SA, et al. Stimulation of
new bone formation by direct transfer of osteogenic plasmid genes. P Natl Acad Sci U S A.
1996;93:5753–8.
189. Zhang Y, Song J, Shi B, Wang Y, Chen X, Huang C, et al. Combination of scaffold and
adenovirus vectors expressing bone morphogenetic protein-7 for alveolar bone regeneration
at dental implant defects. Biomaterials. 2007;28:4635–42.
190. Fontana G, Srivastava A, Thomas D, Lalor P, Dockery P, Pandit A. Three-dimensional
microgel platform for the production of cell factories tailored for the nucleus pulposus.
Bioconjug Chem. 2014;26:1297–306.
191. Browne S, Fontana G, Rodriguez BJ, Pandit A. A protective extracellular matrix-based
gene delivery reservoir fabricated by electrostatic charge manipulation. Mol Pharm.
2012;9:3099–106.
192. Bonadio J, Smiley E, Patil P, Goldstein S. Localized, direct plasmid gene delivery in vivo:
prolonged therapy results in reproducible tissue regeneration. Nat Med. 1999;5:753–9.
193. Chandler LA, Doukas J, Gonzalez AM, Hoganson DK, Gu DL, Ma CL, et al. FGF2-targeted
adenovirus encoding platelet-derived growth factor-B enhances de novo tissue formation.
Mol Ther. 2000;2:153–60.
194. Chandler LA, Gu DL, Ma CL, Gonzalez AM, Doukas J, Nguyen T, et al. Matrix-enabled
gene transfer for cutaneous wound repair. Wound Repair Regeneration. 2000;8:473–9.
195. Doukas J, Chandler LA, Gonzalez AM, Gu DL, Hoganson DK, Ma CL, et al. Matrix
immobilization enhances the tissue repair activity of growth factor gene therapy vectors.
Hum Gene Ther. 2001;12:783–98.
196. Tyrone JW, Mogford JE, Chandler LA, Ma CL, Xia YP, Pierce GF, et al.
Collagen-embedded platelet-derived growth factor DNA plasmid promotes wound healing
in a dermal ulcer model. J Surg Res. 2000;93:230–6.
197. Doukas J, Blease K, Craig D, Ma CL, Chandler LA, Sosnowski BA, et al. Delivery of FGF
genes to wound repair cells enhances arteriogenesis and myogenesis in skeletal muscle. Mol
Ther. 2002;5:517–27.
198. Berry M, Gonzalez AM, Clarke W, Greenlees L, Barrett L, Tsang W, et al. Sustained effects
of gene-activated matrices after CNS injury. Mol Cell Neurosci. 2001;17:706–16.
199. Smetana K. Cell biology of hydrogels. Biomaterials. 1993;14:1046–50.
200. Kong HJ, Hsiong S, Mooney DJ. Nanoscale cell adhesion ligand presentation regulates
nonviral gene delivery and expression. Nano Lett. 2007;7:161–6.
201. Schek RM, Hollister SJ, Krebsbach PH. Delivery and protection of adenoviruses using
biocompatible hydrogels for localized gene therapy. Mol Ther. 2004;9:130–8.
202. Pascher A, Palmer G, Steinert A, Oligino T, Gouze E, Gouze J, et al. Gene delivery to
cartilage defects using coagulated bone marrow aspirate. Gene Ther. 2004;11:133–41.
203. Shen H, Tan J, Saltzman WM. Surface-mediated gene transfer from nanocomposites of
controlled texture. Nat Mater. 2004;3:569–74.
204. Kumta PN, Sfeir C, Lee DH, Olton D, Choi D. Nanostructured calcium phosphates for
biomedical applications: novel synthesis and characterization. Acta Biomater. 2005;1:65–83.
5 Biologically Inspired Materials in Tissue Engineering 145
205. Olton D, Li JH, Wilson ME, Rogers T, Close J, Huang L, et al. Nanostructured calcium
phosphates (NanoCaPs) for non-viral gene delivery: Influence of the synthesis parameters on
transfection efficiency. Biomaterials. 2007;28:1267–79.
206. Newland B, Moloney TC, Fontana G, Browne S, Abu-Rub MT, Dowd E, et al. The
neurotoxicity of gene vectors and its amelioration by packaging with collagen hollow
spheres. Biomaterials. 2013;34:2130–41.
207. Guan S, Rosenecker J. Nanotechnologies in delivery of mRNA therapeutics using nonviral
vector-based delivery systems. Gene Ther. 2017;24:133–43.
208. Frisch J, Rey-Rico A, Venkatesan JK, Schmitt G, Madry H, Cucchiarini M. Chondrogenic
differentiation processes in human bone marrow aspirates upon rAAV-mediated gene
transfer and overexpression of the insulin-like growth factor I. Tissue Eng Part A.
2015;21:2460–71.
209. Rajangam T, An SS. Fibrinogen and fibrin based micro and nano scaffolds incorporated with
drugs, proteins, cells and genes for therapeutic biomedical applications. Int J Nanomed.
2013;8:3641–62.
210. Ahmed TA, Dare EV, Hincke M. Fibrin: a versatile scaffold for tissue engineering
applications. Tissue Eng Part B Rev. 2008;14:199–215.
211. Dare EV, Griffith M, Poitras P, Kaupp JA, Waldman SD, Carlsson DJ, et al. Genipin
cross-linked fibrin hydrogels for in vitro human articular cartilage tissue-engineered
regeneration. Cells Tissues Organs. 2009;190:313–25.
212. Moncion A, Arlotta KJ, Kripfgans OD, Fowlkes JB, Carson PL, Putnam AJ, et al. Design
and characterization of fibrin-based acoustically responsive scaffolds for tissue engineering
applications. Ultrasound Med Biol. 2016;42:257–71.
213. Raut SD, Lei P, Padmashali RM, Andreadis ST. Fibrin-mediated lentivirus gene transfer:
implications for lentivirus microarrays. J Control Release. 2010;144:213–20.
214. Lei P, Padmashali RM, Andreadis ST. Cell-controlled and spatially arrayed gene delivery
from fibrin hydrogels. Biomaterials. 2009;30:3790–9.
215. Sahni A, Odrljin T, Francis CW. Binding of basic fibroblast growth factor to fibrinogen and
fibrin. J Biol Chem. 1998;273:7554–9.
216. Mosesson MW. Fibrinogen and fibrin structure and functions. J Thromb Haemost.
2005;3:1894–904.
217. Tabele C, Montana M, Curti C, Terme T, Rathelot P, Gensollen S, et al. Organic glues or
fibrin glues from pooled plasma: efficacy, safety and potential as scaffold delivery systems.
J Pharm Pharm Sci. 2012;15:124–40.
218. de la Puente P, Ludena D. Cell culture in autologous fibrin scaffolds for applications in tissue
engineering. Exp Cell Res. 2014;322:1–11.
219. Noori A, Ashrafi SJ, Vaez-Ghaemi R, Hatamian-Zaremi A, Webster TJ. A review of fibrin
and fibrin composites for bone tissue engineering. Int J Nanomed. 2017;12:4937–61.
220. Gasperini L, Mano JF, Reis RL. Natural polymers for the microencapsulation of cells. J R
Soc Interface. 2014;11:20140817.
221. Wittmann K, Storck K, Muhr C, Mayer H, Regn S, Staudenmaier R, et al. Development of
volume-stable adipose tissue constructs using polycaprolactone-based polyurethane scaffolds
and fibrin hydrogels. J Tissue Eng Regen Med. 2016;10:E409–18.
222. Eyrich D, Wiese H, Mailer G, Skodacek D, Appel B, Sarhan H, et al. In vitro and in vivo
cartilage engineering using a combination of chondrocyte-seeded long-term stable fibrin gels
and polycaprolactone-based polyurethane scaffolds. Tissue Eng. 2007;13:2207–18.
223. Almany L, Seliktar D. Biosynthetic hydrogel scaffolds made from fibrinogen and
polyethylene glycol for 3D cell cultures. Biomaterials. 2005;26:2467–77.
224. Oss-Ronen L, Seliktar D. Polymer-conjugated albumin and fibrinogen composite hydrogels
as cell scaffolds designed for affinity-based drug delivery. Acta Biomater. 2011;7:163–70.
225. Frisman I, Seliktar D, Bianco-Peled H. Nanostructuring PEG-fibrinogen hydrogels to control
cellular morphogenesis. Biomaterials. 2011;32:7839–46.
146 G. Fontana et al.
226. Jiang B, Waller TM, Larson JC, Appel AA, Brey EM. Fibrin-loaded porous poly(ethylene
glycol) hydrogels as scaffold materials for vascularized tissue formation. Tissue Eng Part A.
2013;19:224–34.
227. Rao RR, Peterson AW, Ceccarelli J, Putnam AJ, Stegemann JP. Matrix composition
regulates three-dimensional network formation by endothelial cells and mesenchymal stem
cells in collagen/fibrin materials. Angiogenesis. 2012;15:253–64.
228. Lee F, Kurisawa M. Formation and stability of interpenetrating polymer network hydrogels
consisting of fibrin and hyaluronic acid for tissue engineering. Acta Biomater. 2013;9:
5143–52.
229. King VR, Alovskaya A, Wei DYT, Brown RA, Priestley JV. The use of injectable forms of
fibrin and fibronectin to support axonal ingrowth after spinal cord injury. Biomaterials.
2010;31:4447–56.
230. Lorentz KM, Kontos S, Frey P, Hubbell JA. Engineered aprotinin for improved stability of
fibrin biomaterials. Biomaterials. 2011;32:430–8.
231. Roura S, Galvez-Monton C, Bayes-Genis A. Fibrin, the preferred scaffold for cell
transplantation after myocardial infarction? An old molecule with a new life. J Tissue Eng
Regenerative Med. 2017;11:2304–13.
232. Ye L, Chang YH, Xiong Q, Zhang P, Zhang L, Somasundaram P, et al. Cardiac repair in a
porcine model of acute myocardial infarction with human induced pluripotent stem
cell-derived cardiovascular cells. Cell Stem Cell. 2014;15:750–61.
233. Johnson PJ, Parker SR, Sakiyama-Elbert SE. Controlled release of neurotrophin-3 from
fibrin-based tissue engineering scaffolds enhances neural fiber sprouting following subacute
spinal cord injury. Biotechnol Bioeng. 2009;104:1207–14.
234. Wilems TS, Pardieck J, Iyer N, Sakiyama-Elbert SE. Combination therapy of stem cell
derived neural progenitors and drug delivery of anti-inhibitory molecules for spinal cord
injury. Acta Biomater. 2015;28:23–32.
235. Agbay A, Edgar JM, Robinson M, Styan T, Wilson K, Schroll J, et al. Biomaterial strategies
for delivering stem cells as a treatment for spinal cord injury. Cells Tissues Organs.
2016;202:42–51.
236. Iwaya K, Mizoi K, Tessler A, Itoh Y. Neurotrophic agents in fibrin glue mediate adult dorsal
root regeneration into spinal cord. Neurosurgery. 1999;44:589–95.
237. Hyatt AJT, Wang D, van Oterendorp C, Fawcett JW, Martin KR. Mesenchymal stromal cells
integrate and form longitudinally-aligned layers when delivered to injured spinal cord via a
novel fibrin scaffold. Neurosci Lett. 2014;569:12–7.
238. Ito K, Yamada Y, Naiki T, Ueda M. Simultaneous implant placement and bone regeneration
around dental implants using tissue-engineered bone with fibrin glue, mesenchymal stem
cells and platelet-rich plasma. Clin Oral Implan Res. 2006;17:579–86.
239. Galler KM, Brandl FP, Kirchhof S, Widbiller M, Eidt A, Buchalla W, et al. Suitability of
different natural and synthetic biomaterials for dental pulp tissue engineering. Tissue Eng
Part A. 2018;24:234–44.
240. Haleem AM, Singergy AA, Sabry D, Atta HM, Rashed LA, Chu CR, et al. The clinical use
of human culture-expanded autologous bone marrow mesenchymal stem cells transplanted
on platelet-rich fibrin glue in the treatment of articular cartilage defects: a pilot study and
preliminary results. Cartilage. 2010;1:253–61.
241. Handl M, Trc T, Hanus M, Stastny E, Fricova-Poulova M, Neuwirth J. et al [Therapy of
severe chondral defects of the patella by autologous chondrocyte implantation]. Acta Chir
Orthop Traumatol Cechoslovaca. 2006;73:373–9.
242. Horch RE, Kopp J, Kneser U, Beier J, Bach AD. Tissue engineering of cultured skin
substitutes. J Cell Mol Med. 2005;9:592–608.
243. Yamamoto M, Yanaga H, Nishina H, Watabe S, Mamba K. Fibrin stimulates the
proliferation of human keratinocytes through the autocrine mechanism of transforming
growth factor-alpha and epidermal growth factor receptor. Tohoku J Exp Med.
2005;207:33–40.
5 Biologically Inspired Materials in Tissue Engineering 147
N. N. Aldini (&)
Laboratory of Preclinical and Surgical Studies, Rizzoli Orthopedic Institute,
Bologna, Italy
e-mail: [email protected]
M. Fini
Laboratory of Biocompatibility, Innovative Technologies
and Advanced Therapies, Rizzoli RIT Department, Bologna, Italy
e-mail: milena.fi[email protected]
clinical functions. Since research into these applications is quite new, we can expect
interesting future developments, in which the nanotechnologies might play a
decisive role.
Keywords Silk fibroin Nanotechnologies Nanomaterials Biomaterials
Biocompatibility Composite materials Preclinical studies Hard tissues
In vitro study In vivo study
Silks are protein fibers produced by silkworms (Bombyx mori), but also by spiders
and others arthropods. The architecture of silk is based on two proteins: fibroin,
which is the core filament, and sericin, which is a protein that glues fibroin fibers
together.
Although silk was used extensively in surgery for making suture threads, this
application has now been replaced by synthetic materials with greater histocom-
patibility than natural ones. Benefits and drawbacks of silks for biomaterial
applications have been well detailed by Altman, Vepari, and other authors and are
summarized in Table 6.1 [1, 2]. More recently, because sericin has been recognized
as the main cause of silk’s poor performance due to its antigenicity, fibroin alone
has remained popular as a biomaterial.
Fibroin is made of highly organized crystals and semi-crystalline regions that
account for its elasticity. The primary structure is mainly composed of the amino
acids glycine, alanine, serine, valine, and tyrosine with characteristic sequences.
These structural elements produce the strength and resiliency of silk fibroin [3].
Indeed, silk has interesting mechanical properties regarding its use as biomaterial.
Other advantages of this biological product are its water-based processing,
biodegradability, and the presence of easily accessible chemical groups for func-
tional modifications [4].
Nowadays, tissue engineering procedures have become widespread in regener-
ative medicine for the treatment of diseases when standard medical procedures fail.
Regenerative strategies are based on the combination of three main tools: cells
(differentiated and not differentiated), scaffolds, and growth factors. Silk fibroin also
regeneration of the musculoskeletal system have been reviewed to focus on the use
of fibroin as a scaffold and the technologies adopted for the nanostructure
development.
Following a preclinical protocol, for example, in an in vitro and in vivo study
Fini et al. [27] evaluated the behavior of an injectable silk fibroin hydrogel through
osteoblast cultures and after implantation in critical size defects of rabbit distal
femurs, using synthetic poly (D, L-lactide–glycolide) copolymer as control material.
In vitro biocompatibility was evaluated by measuring cytotoxicity and cytocom-
patibility on human osteoblast-like cell line (MG 63), whereas in vivo the bone
defect healing rate and quality of the newly formed bone inside the defects were
determined by measuring histomorphometric parameters, such as trabecular bone
volume, trabecular thickness, trabecular number, and trabecular separation. In vitro
tests indicated that both materials significantly increased cell proliferation in
comparison with the negative control. Both materials promoted bone healing when
used to fill critical size defects in rabbit femurs, but the histomorphometry showed
better results in new-formed bone of the silk fibroin hydrogel-treated defects in
comparison with the control gel. The regrown bone of the Silk fibroin hydrogel-
treated defects appeared to be more similar to normal bone than that of the control
synthetic polymeric material-treated defects, in comparison with controls treated
with a synthetic polymeric material, thus suggesting that silk fibroin hydrogel can
accelerate remodeling processes. Like this study, which is aimed at hard tissue
repair, many others describe a range of scaffold preparation procedures and the
tissues to be replaced.
Electrospinning technology, which uses an electrical charge to draw very fine
fibers on the micro- or nanoscale, enables porous nanofibrous scaffolds to be
obtained, which are able to mimic the ECM. Considering the physical–chemical
properties and the structure of the scaffolds, micro- and nanoparticles can be
obtained from silk solutions by various procedures, such as freeze-drying and
grinding procedures, spray drying, jet breaking, self-assembly, and freeze-thawing.
Milling of silk fibers is also an option to obtain silk particles using any chemicals.
According to Kundu et al. [4], these particles can play the dual role of improving
mechanical properties of scaffolds and at the same time act as a carrier of growth
factors for rapid tissue regeneration. Indeed to improve mechanical and biological
performances, inorganic or organic fillers have been incorporated in silk 3D scaf-
folds during or after fabrication to obtain composites. The main advantage is in this
case the compatibility between the components. Consequences of a poor compat-
ibility may result in inhomogeneous mixtures, phase separation, and adverse tissue
reactions. To obtain better compatibility, silk–silk composite scaffolds are made by
incorporating milled silk particles in porous silk sponge, resulting in a significant
improvement in mechanical properties.
With respect to the material porosity, different methods of processing are pre-
sented in the literature. The importance of the processing method is highlighted by
Kuboyama et al. These authors evaluated the porosity of scaffolds prepared using
regenerated Bombyx mori silk fibroin dissolved either in water (AF) orin
hexafluoroisopropanol (HFIP). The two preparations were comparatively analyzed
6 Advances in Nanotechnologies for the Fabrication of Silk … 155
in an animal model in which the formation and growth of new bone in the
implantation site (rabbit femoral epicondyle) was examined by means of micro-CT
and histology. The AF scaffold exhibited significantly greater osteoconductivity
than that obtained by the protein dissolved in HFIP. Micro-CT analysis showed that
the morphology of the newly formed bone differed significantly in the two types of
silk fibroin scaffold. After 4 weeks of implantation, new trabecular bone was seen
inside the pores of the AF scaffold implant, whereas the HFIP scaffold only con-
tained necrotic cells. No trabecular bone was seen within the pores of the latter
scaffolds, although the pores of these did contain giant cells and granulation tissue
[28].
Lin et al. [29] evaluated silk fibroin with different nanostructures, self-assembled
in aqueous solution to improve porous structure formation. In comparison with
scaffolds derived from fresh solution, the nanofibrous silk scaffolds showed better
cell compatibility in vitro. These observations suggest that the control of silk
nanoscale can regulate matrix features including porous structure and nanostructure,
which are important in regulating cell and tissue outcomes.
In the specific field of bone tissue engineering, silk fibroin gained much interest
as a scaffold material and various strategies were developed to create a
three-dimensional (3D) structure with high porosity and osteoconductivity.
Salt-leaching or freeze-drying methods were used to create porous scaffolds.
Moreover, in attempts to mimic bone properties, the incorporation of ceramic
components into silk fibroin scaffolds has also been shown. Fibrous silk fibroin
scaffolds were obtained using the electrospinning technique and the bone regen-
eration in the scaffolds was confirmed; they are considered to be effective in
replacing collagen for many tissue engineering applications [30].
The use of non-mulberry tropical silk fibroin as a bioactive polymer in blended
nanofibrous matrices resulted in osteoconductive scaffolds. The blending of 2 wt%
silk fibroin exhibited higher cell attachment, growth, and ECM formation, when
compared to unmodified polyvinyl alcohol as control and the other blend ratio. The
mechanical robustness of constructs resembled the original bone tissue, thus indi-
cating a promising future for this blend in bone regeneration and reconstruction.
This in vitro study, therefore, lays the foundation for designing clinically relevant
orthopedic grafts in vivo [31]. Considering in particular, the behavior of scaffolds
interacting with cells, Teuschl et al. [32] found that surface modifications of fibroin
with carbohydrate-binding protein lectin enhanced the adhesion of cells, in par-
ticular adipose-derived stromal cells; the proliferation and differentiation in osteo-
genic lineages were however not influenced. Considering that silk fibroin is used to
obtain scaffolds for bone tissue engineering, this possibility may be of practical
interest. Uchida et al. [33] also showed that plasma-irradiated silk fibroin was a
regulator of bone matrix properties in an animal model based on the placement of
scaffolds in critical size defects in rabbit femurs, which increased bone matrix,
mineral concentration, cortical thickness, and trabecular bone volume.
Fibroin fibers can also be used as composites with other polymers. Yang et al.
[34] studied different types of scaffolds based on silk fibroin and poly (L-lactide-co-
caprolactone) blends intended for tendon tissue engineering. Biocompatibility
156 N. N. Aldini and M. Fini
Conclusions
The manifold investigations that span through very different fields of applications
concur that fibroin is a promising biomaterial. Further advancements will open up
new perspectives in the use of this product in tissue regeneration. Silk-based
materials are of particular interest where slow biodegradation and good mechanical
properties are required, such as in tissue engineering of bone, ligaments, and
musculoskeletal tissues.
Their versatility in processing, biocompatibility properties, ease of sterilization,
thermal stability, possibility for surface chemical modifications, and controllable
degradation therefore make silk-derived proteins promising biomaterials for many
clinical functions. Since research into these applications is quite new, we can expect
interesting future developments, in which the nanotechnologies might play a
decisive role.
References
1. Altman GH, Diaz F, Jakuba C, Calabro T, Horan RL, Chen J, Lu H, Richmond J, Kaplan DL.
Silk-based biomaterials. Biomaterials. 2003;24:401–16.
2. Vepari C, Kaplan DL. Silk as a biomaterial. Prog Polym Sci. 2007;32:991–1007.
3. Meinel L, Hofmann S, Karageorgiou V, Kirker-Heade C, McCoole J, Gronowicz G,
Zichner L, Langer R, Vunjak-Novakovica G, Kaplan DL. The inflammatory responses to silk
films in vitro and in vivo. Biomaterials. 2005;26:147–55.
4. Kundu B, Rajkhowa R, Kundu SC, Wang X. Silk fibroin biomaterials for tissue regenerations.
Advanc Drug Deliv Rev. 2013;65:457–70.
5. Chen CH, Liu JM, Chua CK, Chou SM, Shyu VBH, Chen JP. Cartilage tissue engineering
with silk fibroin scaffolds fabricated by indirect additive manufacturing technology. Materials.
2014;7:2104–19.
6. Ma Z, Kotaki M, Inai R, Ramakrishna S. Potential of nanofiber matrix as tissue-engineering
scaffolds. Tissue Eng. 2005;11(1–2):101–9.
7. Lu G, Liu S, Lin S, Kaplan DL, Lu Q. Silk porous scaffolds with nanofibrous microstructures
and tunable properties. Coll Surf B Biointerfaces. 2014;120:28–37.
8. Causin F, Pascarella R, Pavesi G, Marasco R, Zambon G, Battaglia R, Munari M. Acute
endovascular treatment (48 hours) of uncoilable ruptured aneurysms at non-branching sites
using silk flow-diverting devices. Interv Neuroradiol. 2011;17(3):357–64.
9. Leonardi M, Cirillo L, Toni F, Dall’Olio M, Princiotta C, Stafa A, Simonetti L, Agati R.
Treatment of intracranial aneurysms using flow-diverting silk stents (BALT): a single centre
experience. Interv Neuroradiol. 2011;17(3):306–15.
10. Wang CY, Zhang K-H, Fan CY, Mo XM, Ruan HJ, Li FF. Aligned natural–synthetic
polyblend nanofibers for peripheral nerve regeneration. Acta Biomater. 2011;7:634–43.
11. Yoo CR, Yeo IS, Park KE, Park JH, Lee SJ, Park WH, Min BM. Effect of chitin/silk fibroin
nanofibrous bicomponent structures on interaction with human epidermal keratinocytes. Int J
Biol Macromol. 2008;42:324–34.
12. Meinel L, Betz O, Fajardo R, Hofmann S, Nazarian A, Cory E, Hilbe M, McCool J, Langer R,
Vunjak-Novakovic G, Merkle HP, Rechenberg B, Kaplan DL, Kirker-Head C. Silk based
biomaterials to heal critical sized femur defects. Bone. 2006;39:922–31.
6 Advances in Nanotechnologies for the Fabrication of Silk … 159
13. Li C, Vepari C, Jin HJ, Kim HJ, Kaplan DL. Electrospun silk-BMP-2 scaffolds forbone tissue
engineering. Biomaterials. 2006;27:3115–24.
14. Kweon H, Lee K-G, Chae C-H, Balázsi C, Min S-K, Kim JY, Choi JY, Kim SG.
Development of nano-hydroxyapatite graft with silk fibroin scaffold as a new bone substitute.
J Oral Maxillofacc Surg. 2011;69:1578–86.
15. Wang Y, Blasioli DJ, Kim HJ, Kim HS, Kaplan DL. Cartilage tissue engineering with silk
scaffolds and human articular chondrocytes. Biomaterials. 2006;27:4434–42.
16. Gellynck K, Verdonk PCM, Nimmen EV, Almqvist KF, Gheysens T, Schokens G,
Langenhove LV, Kiekens P, Mertens J, Verbruggen G. Silkwormand spider silk scaffolds for
chondrocyte support. J Mater Sci Mater Med. 2008;19:3399–409.
17. Chen X, Qi YY, Wang LL, Yin Z, Yin GL, Zou XH, Ouyang HW. Ligament regeneration
using a knitted silk scaffold combined with collagen matrix. Biomaterials. 2008;29:3683–92.
18. Sahoo S, LokToh S, Hong JC. Goh PLGA nanofiber-coated silk microfibrous scaffold for
connective tissue engineering. J Biomed Mater Res B Appl Biomater. 2010;95B:19–28.
19. Yang MC, Wang SS, Chou NK, Chi NH, Huang YY, Chang YL, Shieh MJ, Chung TW. The
cardiomyogenic differentiation of rat mesenchymal stem cells on silk fibroin– polysaccharide
cardiac patches in vitro. Biomaterials. 2009;30:3757–65.
20. Patra C, Talukdar S, Novoyatleva T, Velagala SR, Mühlfeld C, Kundu B, Kundu SC,
Engel FB. Silk protein fibroin from Antheraea mylitta for cardiac tissue engineering.
Biomaterials. 2012;33:2673–80.
21. Yang T, Zhang M. Potential of nanofiber matrix as tissue-engineering scaffolds. Int J
Ophthalmol. 2008;8:1557–9.
22. Lawrence BD, Marchant JK, Pindrus MA, Omenetto FG, Kaplan DL. Silk film biomaterials
for cornea tissue engineering. Biomaterials. 2009;30:1299–308.
23. Cirillo B, Morra M, Catapano G. Adhesion and function of rat liver cells adherentto silk
fibroin/collagen blend films. Int J Artif Organs. 2004;27:60–8.
24. Chang G, Kim HJ, Vunjak-Novakovic G, Kaplan DL, Kandel R. Enhancing annulusfibrosus
tissue formation in porous silk scaffolds. J Biomed Mater Res A. 2010;92A:43–51.
25. Cannon GM Jr, Mauney JR, Gong EM, Yu RN, Adam RM, Estrada CR. Silk Asa novel
biomaterial in bladder tissue engineering. J Pediatr Urol. 2010;6(S1):S82.
26. Ghassemifar R, Redmond S, Chirila TV, Zainuddin. Advancing towards a tissue-engineered
tympanic membrane: silk fibroin as a substratum for growing human eardrum keratinocytes.
J Biomater Appl. 2010;24:591–606.
27. Fini M, Motta A, Torricelli P, Giavaresi G, Nicoli Aldini N, Tschon M, Giardino R,
Migliaresi C. The healing of confined critical size cancellous defects in the presence of silk
fibroin hydroge. Biomaterials. 2005;26:3527–36.
28. Kuboyama N, Kiba H, Arai K, Uchida R, Tanimoto Y, Bhawal UK, Abiko Y, Miyamoto S,
Knight D, Asakura T, Nishiyama N. Silk fibroin-based scaffolds for bone regeneration.
J Biomed Mater Res Part B Appl Biomater. 2013;2013(101B):295–302.
29. Lin S, Lu G, Liu S, Bai S, Liu X, Lu Q, Zuo B, Kaplan DL, Zhu H. Nanoscale control of silks
for nanofibrous scaffold formation with improved porous structure. J Mater Chem B Mater
Biol Med. 2014;2(17):2622–33.
30. Kim H, Che L, Ha Y, Ryu W. Mechanically-reinforced electrospun composite silk fibroin
nanofibers containing hydroxyapatite nanoparticles. Mater Sci Eng. 2014;40:324–35.
31. Bhattacharjee P, Kundu B, Naskar D, Maiti TK, Bhattacharya D, Kundu SC. Nanofibrous
Nonmulberry Silk/PVA Scaffold for Osteoinduction and Osseointegration. Biopolymers.
2015;103:271–84.
32. Teuschl AH, Neutsch L, Monforte X, Rünzler D, van Griensven M, Gabor F, Redl H.
Enhanced cell adhesion on silk fibroin via lectin surface modification. Acta Biomaterialia.
2014;10:2506–17.
33. Uchida R, Bhawal UK, Kiba H, Arai K, Tanimoto Y, Kuboyama N, Asakura T, Nishiyama N.
Effect of plasma-irradiated silk fibroin in bone regeneration. J Biosci Bioeng. 2014;118(3):
333e–40e.
160 N. N. Aldini and M. Fini
Keywords Musculoskeletal tissues healing Mechanotransduction
Scaffolds Nanoscale engineering Bone Tendon Muscle Ligament
Nanometric Architecture
Nanoscale Engineering
cues is especially important in engineering complex tissues that have multiple cell
types and require precisely defined cell–cell and cell–matrix interactions on the
nanoscale. Thus, in a regenerative engineering approach, nanoscale materials/
scaffolds play a paramount role in controlling cell fate and the consequent regen-
erative capacity. Advances in nanotechnology have generated a new toolbox for the
fabrication of tissue-specific nanostructured scaffolds. For example, biodegradable
polymers such as polyesters, polyphosphazenes, polymer blends, and composites
can be electrospun into ECM-mimicking matrices composed of nanofibers, which
provide high surface area for cell attachment, growth, and differentiation [20]. It is
also possible to add to the scaffolds different factors that improve the ECM
generation.
Tensegrity
The musculoskeletal system supports the weight of our bodies, allows us to rapidly
adjust to resist external forces, and permits us to move freely in our environment.
Selective pressures demand that the construction of such a machine minimizes mass
with flexibility and without compromising its structural integrity to handle unex-
pected forces. The musculoskeletal system has evolved to address these demands
through optimization of both material properties (how the individual support ele-
ments are designed) and architecture (how the different elements are oriented and
connected together).
The human skeleton is actually strongly influenced by the architecture of our
bodies. Their mechanical behavior depends on how the surrounding tensile
164 F. Oliva et al.
muscles, tendons, and ligaments are joined and oriented in. The internal tension
and/or prestress stabilizes the joints: Even when the bones are pulled away from
each other, the joint does not dislocate. Actually, there are multiple muscles, ten-
dons, and ligaments that contribute to the structure of each joint and the number and
position of these tensile elements play a critical role in defining the joint’s potential
strength, power, speed, and range of motion.
The local stress patterns are clearly visualized in sections or radiographs of the
human femur which demonstrate that the network of trabecular bone is organized to
approximate the principal stress directions. This is known by 1892, when Julius
Wolff and others realized that mechanical loads can affect bone architecture in
living beings, but the mechanisms responsible for this effect were unknown. By
2003, we know how this effect occurs and some of its applications. Our
load-bearing bones include tibias, femurs, humeri, vertebrae, radii, mandibles,
maxillae, wrists, hips, etc. The strength of such bones and their trabeculae would
represent their most important physiologic feature but in the special sense of rel-
ative to the size of the typical peak voluntary loads on them. The biologic “ma-
chinery” that determines whole-bone strength forms a tissue-level negative
feedback system called the mechanostat. Two thresholds make a bone’s strains
determine its strength by switching on and off the biologic mechanisms that
increase or decrease its strength. The largest voluntary loads on load-bearing bones
determine most of their strength after birth, and these loads come from muscle. This
process affects, in part, the healing of fractures, bone grafts, osteotomies, and
arthrodeses; the bone’s ability to endure load-bearing joint and dental endopros-
theses; why healthy bones are stronger than the minimum needed to keep voluntary
loads from breaking them suddenly or from fatigue; some general functions and
disorders of bone modeling and basic multicellular unit-based bone remodeling;
some limitations of in vitro data and of pharmaceutical actions; and the fact that
many bone-active humoral and local agents have permissive roles in a bone’s
adaptations and healing, instead of forcing them to occur [22].
This observation suggests that the living cells that continually remodel bone are
able to sense changes in mechanical stresses in their local environment and that they
respond by depositing new ECM where it is needed and removing it from where it
is not. This process results in deposition of bone ECM in specific patterns that
correspond precisely to engineering lines of tension and compression characteristic
for elastic structure of this size and shape with similar loading conditions. It is an
example of the importance of cellular mechanotransduction for regulation of tissue
morphogenesis. The specialized microarchitecture of cancellous bone further
optimizes its structural efficiency (strength/mass ratio). Architectural organization
on a smaller size scale (the molecular level) also contributes significantly to the
mechanical strength of biologic tissue [23]. In the bone, the matrix of each tra-
beculum consists of a composite material containing hydroxyapatite crystals
embedded within a network of collagen fibrils [24]. The collagen augments the
tensile strength of the bone, while the minerals contribute largely to its compressive
stiffness and strength. In the living organism, the stress in the bone ECM is
influenced by the shape of the entire bone, the pull of the surrounding muscles and
7 Nanoscale Architecture for Controlling Cellular … 165
tendons, and its loading conditions. Contractile fibroblasts also prestress the col-
lagen network during the process of tissue development and remodeling, before the
surrounding ECM calcifies. Prestress also plays an important role in determining
the mechanics of cartilage, tendons, and ligaments [23]. In cartilage, the loose
collagen network is stretched open and prestressed by the osmotic force of
hydration of embedded proteoglycan molecules [9, 25, 26]; however, the cellular
components (chondrocytes) and their internal support elements (cytoskeleton,
nucleus) may also bear some mechanical loads [27, 28]. In soft tissues, that are
composed mostly of parallel collagen fibers and elastin [29, 30], such as ligaments
and tendons, the prestress results from the active contraction of living cells (my-
ofibroblasts) that are embedded within its ECM. The cell contractions pull the
collagen into an undulating, buckled structure and keep the ligament under tension
at all times [31]. Thus, these soft tissues remodel and adjust their fiber orientations
to optimize their load-bearing capacity much like bone and cartilage [23].
on the cytoskeletal backbone of the focal adhesion [35, 36]. Thus, mechanical
signals may be integrated with other environmental signals and transduced into a
biochemical response through stress-dependent changes in cytoskeletal scaffold
geometry or mechanical deformation [9]. For example, one potential mechanism for
mechanochemical transduction is through stress-dependent cytoskeletal rearrange-
ments that result in changes in proximity between different immobilized enzymes
and substrates. If a protein kinase and its physiological substrate were both
immobilized on the cytoskeleton and physically separated, then no phosphorylation
would result. However, if there is a mechanical deformation of the tissue, ECM,
cytoskeletal composite results in structural rearrangements that bring these two
molecules into direct apposition and then phosphorylation might proceed causing a
downstream signaling cascade to initiate. In addition, the transfer of focused
mechanical energy to soluble molecules in the cytosol will alter their shape and
hence their electrochemical potential through mechanical distortion. Stress-induced
changes in molecular mechanics (stiffness and conformation) can then produce
direct mechanochemical transduction by altering thermodynamic (association and
dissociation constants) or kinetic (molecular motion) parameters. Regulation of
tubulin polymerization (microtubule formation) by mechanical stresses balanced
between microtubules, contractile microfilaments, and ECM provides one example
of a thermodynamic transduction mechanism [37]. Stress-dependent changes in the
frequency of opening and closing “stretch-sensitive” ion channels are an excellent
example of the kinetic form of transduction [33]. Living cells sense multiple
simultaneous inputs and are able to organize a single, concerted response. The
tensegrity structure may be used to focus mechanical energy on critical transducing
molecules and to “tune” the entire cellular response to stress by mechanically
coupling biologic structures at different size scales and in different locations within
living cells, tissues, and organs. This tuning function may be accomplished by
altering the prestress in the system (e.g., by varying cytoskeletal tension), remod-
eling architecture, or modifying the mechanics of individual structural components.
Specificity results from local changes in material properties of the structural ele-
ments (e.g., stress will rapidly dissipate in highly viscous regions), and from how
the different elements are mechanically coupled (e.g., linkage of the cytoskeleton to
the ECM by integrins and to the cytoskeleton of neighboring cells by cell–cell
adhesion molecules, such as cadherins) [13].
Also in tendon studies, it is evident the important role of ECM. Tendon ruptures
are increasingly common, repair can be difficult, and healing is poorly understood.
Tissue engineering approaches often require expansion of cell numbers to populate
a construct, and maintenance of cell phenotype is essential for tissue regeneration.
Total collagen, the ratio of collagen types I and III, and decorin can be used as
indicators of matrix formation in human tendon, and expression of the integrin beta
1 subunit as a marker of cell–matrix interactions, for example in patients receiving
surgery for rupture. During the healing, cells become more rounded and are more
widely spaced at confluence, and confluent cell density declines. No change to total
cell layer collagen is observed, but the ratio of type III to type I collagen increases at
the end. Decorin expression significantly decreases, while integrin expression does
7 Nanoscale Architecture for Controlling Cellular … 167
not change. Thus, the phenotype of tenocytes in culture rapidly drifts during the
healing [38].
This architectural model of biologic organisms may help to explain one of the
most fundamental properties of living creatures: how the parts and the whole
function as a single integrated system. Due to use of tensegrity architecture,
mechanical stress is concentrated and focused on signal-transducing molecules that
physically associate with cell surface molecules that anchor cells to extracellular
matrix, such as integrins, and with load-bearing elements within the internal
cytoskeleton and nucleus. Mechanochemical transduction may then proceed
through local stress-dependent changes in molecular mechanics, thermodynamics,
and kinetics within the cell. In this manner, the entire cellular response to stress may
be orchestrated and tuned by altering the prestress in the cell, just as changing
muscular tone can alter mechanical stability and structural coordination throughout
the whole musculoskeletal system [13].
The finding that cells are not bits of viscous cytoplasm surrounded by an elastic
membrane but instead structured as tensegrities with internal load-bearing struts and
tensed cables also has led to novel insights into developmental control and
pathobiology also in other fields, and not only for the musculoskeletal system. For
example, the finding that microtubules bear compression in living cells is extremely
relevant for heart physiology because an increased density of the microtubule
component of the extramyofilament portion of the cardiocyte cytoskeleton caused
by pressure overload can physically interfere with inward-directed shortening of the
myofibrillar bundle and hence lead to contractile dysfunction associated with car-
diac hypertrophy. The integrated nature of biological architecture also helps to
explain why cardiac diseases and developmental abnormalities can be caused by
mutations in various ostensibly unrelated molecules, including integrins,
cytoskeletal filaments, ion channels, or nuclear components. Forces channeled over
ECMs and to integrins are converted into biochemical changes by producing
changes in deformation of other load-bearing mechanotransducer molecules, such
as stress-sensitive ion channels, protein kinases, G proteins, and other signaling
molecules, inside the cell. Recent advances in mathematics, engineering, and sta-
tistical mechanical models of tensegrity structures, and in the use of nanotechnol-
ogy to create artificial cell–material control interfaces, may provide new ways to
investigate, model, manipulate, probe, and control these fundamental mechan-
otransduction mechanisms in living cells, tissues, and organs, including heart, in the
future [39].
Hormesis
responses, from adaptation to cell death. For example, physical exercises and
chronic contractile activity induce a number of physiological adaptations that
ameliorate muscle function and exercise performance. Trained muscle undergoes a
remodeling toward a more oxidative phenotype altering the ultrastructural and
subcellular organization. One of the most important effects of endurance training is
mitochondrial biogenesis with an increase in mitochondrial content. It is
well-known that physical exercise increases ROS generation in skeletal muscle
cells. During exercise, the increased ROS production is mainly due to the high
oxygen consumption that takes place during increased mitochondrial activity. There
is probably generation of ROS at multiple subcellular locations in response to a
multiplicity of mechanical and metabolic stimuli in muscle cells [41]. Thus,
contraction-induced production of ROS has been shown to cause oxidative stress to
skeletal muscle. As an adaptive response, muscle antioxidant defense systems are
upregulated after heavy exercise. Nuclear factor (NF) kappaB and
mitogen-activated protein kinases (MAPKs) are the major oxidative stress-sensitive
signal transduction pathways. Activation of NF-kappaB signaling cascade has been
shown to enhance the gene expression of important enzymes, such as mitochondrial
superoxide dismutase (MnSOD) and inducible nitric oxide synthase (iNOS).
MAPK activations are involved in a variety of cellular functions including growth,
proliferation, and adaptation. The exercise has effects on NF-kappaB and MAPK
signaling, as well as on the time course of activation, in rat skeletal muscle. In
addition, ROS have a role in the exercise-induced upregulation of MnSOD and
iNOS, and in the potential interactions of NF-kappaB and MAPK in the signaling of
these enzymes. It is clear that ROS may serve as messenger molecules to activate
adaptive responses through these redox-sensitive signaling pathways to maintain
cellular oxidant–antioxidant homeostasis during exercise [42]. Furthermore, phys-
ical exercise shares common metabolic paths with caloric restriction and glucose
restriction increasing mitohormesis and inducing a positive adaptive response that
ends with stress resistance, antioxidant defense, and prolonged life span [41]. In
addition, the mild stress-induced hormesis stimulates maintenance and repair sys-
tems and strengthens the homeodynamic space of cells and organisms. Hormesis
through mild heat shock, natural and synthetic hormetins, and other stressors brings
about several antiaging effects in human fibroblasts, keratinocytes, and
telomerase-immortalized bone-marrow stem cells. Depending on the cell type,
these antiaging hormetic effects include extension of replicative life span, enhanced
proteasomal activities, increased chaperone levels, and improved wound healing,
angiogenesis, and differentiation. The main molecular pathways for achieving such
hormetic effects are through targeting the processes for the repair and removal
of molecular damage, which can slow aging [43].
Hormesis has a role not only in the muscle but also in the bone and in its diseases
such as Alzheimer pathology and osteoporosis [40]. Epidemiological studies also
indicate that patients suffering from atherosclerosis are predisposed to develop
osteoporosis. Atherogenic determinants such as oxidized low-density lipoprotein
(oxLDL) particles have been shown both to stimulate the proliferation and promote
apoptosis of bone-forming osteoblasts. OxLDL particles cause hormesis-like
170 F. Oliva et al.
response with the stimulation of both proliferation and cellular NAD (P)
H-dependent reduction potential by low concentrations, whereas high concentra-
tions are associated with the cell death. The induction of hormesis-like response by
oxLDL in osteoblastic cells is associated with the stimulation of cell proliferation
and ROS production by low concentrations of oxLDL. It is generally accepted that
the stimulation of osteoblastic proliferation may compromise their differentiation
into competent bone-forming cells. In accordance, low concentrations of oxLDL
reduce the alkaline phosphatase activity, a marker of osteoblastic maturity. In
addition, oxLDL compromises the migration of osteoblastic cells. Both functions
have been shown to play a critical role in bone formation, remodeling, and fracture
repair. Therefore, low concentrations of oxLDL may alter the bone metabolism by
reducing osteoblastic differentiation in favor of uncontrolled cell proliferation and
by affecting cell migration. On the other hand, high concentrations of oxLDL cause
osteoblastic cell death that will result in reduced bone formation. In summary,
oxLDL particles alter osteoblastic cell proliferation, migration, and apoptosis rate
and thereby may contribute to alteration of bone metabolism equilibrium and may
be responsible for the reduction of bone mass associated with atherogenic condi-
tions [44].
Mesenchymal stem cells (MSCs) have the potential to replace or restore the
function of damaged tissues and offer much promise in the successful application of
tissue engineering and regenerative medicine strategies [45]. It has been demon-
strated that adult mesenchymal stem cells (MSCs) are capable of giving rise to
several different cell types, including myoblasts, adipocytes, fibroblasts, chondro-
cytes, and osteoblasts [46–49]. MSCs exist in a highly specialized microenviron-
ment in which their maintenance, proliferation, migration, and differentiation
involve a complex interplay of many local and systematic signals between stem
cells and adjacent cells [50–55]. Osteoblasts are derived from MSCs and exist in
close proximity to the bone marrow, which serves as a major source of MSCs and
hematopoietic stem cells. They are located on the surface of bone, express multiple
hormone receptors and various cell surface molecules, and secrete hormones and
enzymes that maintain the balance between bone formation and resorption [56, 57].
However, it remains unclear whether and how osteoblasts affect the self-renewal
and differentiation of another major stem cell population in bone-marrow MSCs.
Co-cultures with MSCs and with a higher density of osteoblasts express greater
mRNA expressions of Runx2, type I collagen, and osteocalcin [58]. Human bone-
marrow-derived MSCs, by virtue of their capacity for self-renewal and multipotent
differentiation, are considered promising candidate cells for regenerative medicine
and tissue engineering applications. The multipotent differentiation capabilities of
MSCs into various types of cell lineage, such as osteoblasts, chondrocytes, and
adipocytes, have been characterized in numerous studies [46, 47]. Osteoblasts,
7 Nanoscale Architecture for Controlling Cellular … 171
which are derived from bone-marrow MSCs, are the main cell type responsible for
bone formation and are one of the neighboring cell types close to the bone marrow.
It is believed that interaction between bone-marrow MSCs and their neighboring
cells may lead to MSC osteogenesis via cell–cell contact or osteoblast-derived
soluble factors [59]. It has been demonstrated that Runx2 is as an essential tran-
scription factor for the induction of early osteogenic differentiation. Studies of the
expression of Runx2 demonstrate that the early osteogenesis of co-cultures is highly
dependent on osteoblast numbers co-cultured in an osteoinductive environment.
Co-cultures with MSCs and osteoblasts demonstrated that direct cell–cell contact is
sufficient to induce osteogenic differentiation by enhancing the gene expressions of
Runx2, type I collagen, and osteocalcin. More significant and distinct osteogenic
differentiation is promoted by additional osteogenic supplements. The quality of
osteogenesis, as evidenced by protein expression, is proportional to the quantity of
osteoblasts in the co-cultures. A recent study has also suggested that primary bone-
derived cells promote the osteogenesis of human embryonic stem cells in a
co-culture model by releasing bone morphogenetic proteins 2 and 4 [60]. These
findings demonstrate that the osteogenesis of stem cells may be induced or pro-
moted in a co-culture system through either direct cell–cell contact or secreted
soluble factors derived from osteoblasts. These findings show that direct cell–cell
contact between MSCs and osteoblasts contributed little to MSC osteogenic dif-
ferentiation. In addition, the co-cultures required at least one osteoinductive factor
to induce or enhance the osteogenic differentiation in MSCs: either an appropriate
number of osteoblasts or treatment with osteogenic supplements. Culture conditions
that included both a large quantity of osteoblasts and osteogenic supplements
resulted in synergistic inhibition during osteogenesis by shrinking the process of
osteogenic differentiation. The direct cell–cell contact and soluble osteogenic fac-
tors might act as synergistic modulators to promote or inhibit osteogenic differ-
entiation in MSCs. Understanding the possible synergistic interactions between
MSCs and osteoblasts might be an alternative approach to manipulating the fates of
stem cells and for differentiating lineages in cell-therapeutic strategies and regen-
erative medicine [58]. Mesenchymal tissues are subjected to mechanical stimuli
in vivo, and terminally differentiated cells from the mesenchymal lineage respond
to mechanical stimulation in vivo and in vitro. MSCs have also been shown to be
highly mechanosensitive, and this may present an ideal method for controlling
MSC differentiation. External mechanical forces are able to induce or enhance MSC
differentiation into a wide variety of tissue-specific cells; however, precisely con-
trolling the timing and outcome of MSC differentiation is still a large challenge.
Characterization of the exact force MSC experience in loading systems is important
to know what mechanisms are actually inducing the observed responses and to help
simplify the design of future loading systems. While most bioreactor systems
employ a common force type (e.g., tension or compression), there are also likely to
be other mechanisms at work causing significant secondary effects. Mathematical
and computer modeling is important for characterizing the forces that are being
experienced by the cells [61].
172 F. Oliva et al.
membrane, in ligaments, and in subchondral bone of human joints [68]. CGRP also
seems to be involved in tendon and fracture healing [68, 69]. SP and CGRP have
been identified in the perichondrium and in periarticular neurons [70]. Neuronal
involvement may play an important role in the development of degenerative joint
disease, consequent to progressive age-related loss of joint innervation. Autonomic
nerve fibers may also actively participate in bone repair [71]. It is possible that the
periarticular autonomic nerve fibers, through their neuropeptides, exert a trophic
effect on joint development.
On the other hand, it is necessary to focus the attention on the soft tissues, such as
tendons. Only through the clarification of the microanatomy, pathophysiology,
genetics of ECM, we may be able to improve current therapeutic knowledge on
tendon diseases. Unfortunately, today too often the media and some areas of
medicine offer to the patients new medical therapy based on tissue regeneration of
tendons without scientific certainty of the effectiveness. The main effort of the
research at this time should be focused not only on the in vivo application of new
potential therapies but also and especially on the mechanisms that regulate the
homeostasis of the ECM during exercise and pathologic conditions [72].
Tendon structure. The structure of a tendon is an important example of com-
plexity of ECM three-dimensional organization. The extracellular matrix (ECM) is
a macromolecular network with both structural and regulatory functions. Tendons
are made by a fibrous, compact connective tissue that connects muscle to bone
designed to transmit forces and withstand tension during muscle contraction. The
extracellular matrix (ECM) is a macromolecular network with both structural and
regulatory functions. Indeed, ECM furnishes mechanical and biochemical signals
that cooperate in the integrated control of cell proliferation, survival, migration, and
differentiation. ECM components belong to four major types of macromolecules:
the collagens, elastin, proteoglycans, and noncollagenous glycoproteins (e.g.,
laminins, entactin/nidogen, fibronectin, and tenascins). The most copious proteins
in the ECM are members of the collagen family. The collagens are involved in the
formation of ECM fibrillar and microfibrillar networks and play a key role in
determining tissue-specific mechanical properties.
Tendon’s ultrastructure and ECM. The apparently simple tendonʼs structure
and composition provide both rigidity and flexibility. This ability is based on
tendon nonlinear, viscous-elastic, anisotropic, and heterogeneous mechanical
properties. Similar to other tissues, there is a clear relationship between structure
and function of the tendon [73]. Indeed, different tissue microenvironments provide
specific characteristics to the different three-dimensional organization of the ECM
since embryogenesis [74]. The organization of the ECM in the tendon is peculiar as
well as within different tendons. The dialogues between different cells (fibroblasts,
muscle, and cartilage cells) or better the dialects between different cells mediated by
174 F. Oliva et al.
various signaling cascades dependent on FGFs and TGF beta are used at decisive
stages of development to make specific the phases of induction, organization,
aggregation, or differentiation of cells [75]. The structure of the tendon is so
complex that it is very difficult to maintain its function in the healing since this
process is reparative rather than regenerative in the adult [76, 77]. A tendon consists
of 70% of water and 30% of dry mass, which is composed by 60–80% of type I
collagen and 2% of elastin. Among collagens, the most abundant component is
collagen type I (95%), while type III and type V collagens represent the remaining
5% of the total collagens [77]. There are many types of collagen, and each of these
has a specific function; every year one new type or one new function is discovered.
Tendons in addition to collagen contain proteoglycans, glycosaminoglycans, and
glycoproteins including fibronectin, thrombospondin, and tenascin-C immersed in
different compositions of ECM lying cellular elements represented (90/95%) by
tenoblasts and tenocytes. The remaining 5–10% of the cells consists of chondro-
cytes, synovial and vascular cells [73]. This information can be useful to create
appropriated nanoengineering techniques [78].
endings have been described in normal ACL: (1) fibers of large diameter which are
fast-conducting mechanoreceptive sensory afferents; (2) fibers of small diameter
slow-conducting nociceptive sensory afferents, and (3) sympathetic efferent vaso-
motor fibers. How we said before, four nerve endings have been identified in human
cruciate ligaments: Ruffini corpuscles, Pacinian corpuscles, Golgi tendon organ-like
endings, and free nerve endings [82, 85]. Ligament mechanoreceptors influence
movements via the c-muscle spindle system. They contribute to the stiffness pre-
programming around the joint and are crucial for joint stability maintenance.
Therefore, the knee ligament injury would damage the mechanoreceptors and alter
neuromuscular functions secondary to diminished somatosensory information. ACL
rupture causes considerable changes in stretch reflex excitability, so the “giving
way” of the knee is not simply related to the decrease in mechanical joint stability,
but is closely associated with altered stretch reflex excitability that most probably
takes place on the spinal level. Reduction of mechanoreceptors is not only due to a
lesion of the soft tissue. Mechanoreceptors are not immutable structures, but their
properties change with aging. Aging affects numbers and morphology of
mechanoreceptors. This phenomenon could explain why normal aging process is
associated with deficits in proprioception.
It was left to Abbott et al. [86] to attribute more importance to knee structures, as
they first described the knee ligaments as having rich sensory innervation, which
allowed them to act as the first link in the kinetic chain. Ever since, our under-
standing of this complex functioning has evolved, and it is now widely accepted
that movement or charge in knee position stimulates receptors in knee ligaments
that allow the conscious appreciation of limb position in space. Proprioception is
receptor and neural arc mediated. The stimulation of mechanoreceptors in the knee
ligaments initiates different types of reflex muscle contractions through the neural
arc involving the dorsal root ganglion sensory neurons. Many studies over the last
30 years have demonstrated significant presence of mechanoreceptors in the fibers
of an intact anterior cruciate ligament (ACL). These were first described by Schultz
et al. [87] in 1984, and it was subsequently established that the receptors included
not just Pacinian, Ruffini, or Golgi tendon-type bodies, but also numerous nerve
endings distributed all over the ACL. These receptors play an important role in the
complicated neural network of proprioception. They are capable of detecting
changes not only in tension, speed, acceleration, and direction of movement, but
also allow a subconscious determination of the position of knee joint in space. It
becomes a corollary that damaged mechanoreceptors would alter neuromuscular
functions secondary to diminished somatosensory information (proprioception and
kinesthesia). In modern orthopedics, this has become a key factor in understanding
functional instability after ACL injuries and methods to treat it. Subsequent to an
ACL injury, it has been observed that the relationship between passive stability and
the functional stability of the knee joint is often ambiguous. It is possible that the
functional instability that occurs after an injury to the ACL is due to the combined
effects of excessive tibial translation and a lack of “coordinated muscle activity” to
stabilize the knee joint. This lack of coordinated muscle stabilization of the knee
176 F. Oliva et al.
joint is thought to be due to diminished or absent sensory feedback from the ACL to
the neuromuscular system.
Mechanoreceptors in intact ACL. The first histological demonstration of
mechanoreceptors in the human ACL was done by Schultz et al. [87]. The cruciate
ligaments were obtained at the time of total knee replacement and from autopsy and
amputation specimens. The ultrastructure of nerve endings in a human knee joint
capsule was subsequently described by Halala et al. [88]. These authors found the
three types of nerve endings: free nerve endings, Ruffini corpuscles, and Pacinian
corpuscles. In the joint capsule, free nerve endings were located below the synovial
layer and within the fibrous layer near blood vessels. These nerve terminals are
derived from myelinated A-delta fibers or from unmyelinated C-fibers. Ruffini
corpuscles were present within the fibrous layer and the ligaments of the capsule in
three variations: small Ruffini corpuscles without a capsule, small corpuscles with a
connective tissue capsule, and large Ruffini corpuscles with an incomplete per-
ineural capsule. Their afferent axons were myelinated, and, inside the corpuscle,
nerve terminals were anchored in the connective tissue belonging to the fibrous
layer or to the ligaments, respectively. The presence of an incomplete perineural
capsule depended on the structure of the surrounding connective tissue. In liga-
ments with collagenous fibrils oriented in a parallel fashion, the perineural capsule
was well-developed and the Ruffini corpuscle resembled a Golgi tendon organ; in
areas where the fibrils showed no predominant orientation, Ruffini lacked a capsule.
Small Pacinian corpuscles were situated within the fibrous layer near the capsular
insertion at the meniscus articularis or at the periosteum. Larger Pacinian corpuscles
with one or several inner cores and a perineural capsule were found on the outer
surface of the fibrous layer. By the turn of the century, it was becoming clear that
the mechanoreceptors located in the ACL constitute an afferent source of infor-
mation toward the central nervous system. The ACL deficiency can cause a dis-
turbance in neuromuscular control and affects central programs and consequently
the motor response resulting in serious dysfunction of the injured limb. It was
examined brain activation by using functional magnetic resonance imaging tech-
nique (1.5-T scanner): ACL deficiency can cause reorganization of the central
nervous system, suggesting that such an injury might be regarded as a neuro-
physiologic dysfunction, not a simple peripheral musculoskeletal injury [89]. This
evidence could explain the variation of clinical symptoms that accompany this type
of injury, and the degrees of dysfunctions in different individuals with an
ACL-deficient knee.
Mechanoreceptors in the stump of an injured ACL. In untreated ACL lesions in
humans, morphologically normal mechanoreceptors persisted in the ACL remnant
for about 3 months after injury. Beyond that time, the number of receptors grad-
ually decreased. By the ninth month after injury, only a few nerve endings were
found, and they were totally absent after 1 year. These results indicate that the
proprioceptive potential of the stump may diminish with the passage of time, and
this may have a potential bearing on surgical outcomes in cases where recon-
struction is delayed. Thus, preserving the ACL remnants might improve functional
7 Nanoscale Architecture for Controlling Cellular … 177
the mechanical input to the Golgi tendon organs. Missing of the responses coincides
with an alteration of the dynamic sensitivities [91].
Muscle mechanoreceptors. Golgi tendon organs co-work with another
mechanoreceptor: the muscle spindle, which is the most innervated receptor, with
afferent and efferent fibers. These receptors are aligned parallel to the muscle fibers,
and they primarily detect changes in the length and the speed of muscles [92].
Mechanoreceptors function as transducer devices converting a signal in one form of
energy to another one (i.e., the physical stimulus of tension into a specific nerve
signal). The summation of receptor discharges forms a frequency-modulated code
that the central nervous system uses to analyze joint position, motion, and accel-
eration [93]. The transduction of mechanical stimuli into electrical responses is
essential for audition, kinesthesis, proprioception, and automatic sensation of
pressure and volume. Seems that Pacinian corpuscles and Golgi tendon organs are
more represented than Ruffini endings in ligaments, both Pacinian corpuscles and
Golgi tendon organs are physiologically reflexogenic. Pacinian corpuscles dis-
charge at the onset or cessation of movement and are involved in quick movements;
this afferent feedback to the central nervous system would have a stabilizing effect
on the joints. Golgi tendon organs are found in ligaments, myotendinous and
myoaponeurotic junctions of mammalian skeletal muscle. Their discharges are
responsible for tension and joint position; thus, they are also responsible for joint
stabilization [80, 94].
Healing of muscle mechanoreceptors. Few reports focused the healing of
mechanoreceptors. Some of these showed that muscle spindle afferents reinnervate
their original ending sites with the spindle after a nerve injury but the afferents that
had been regenerated are smaller than the control group. In contrast Haftel and
coworkers showed that not all the regenerated afferences were able to restore a
pathway to central nervous system. Moreover, there were some differences in
muscle spindle responses to stretch, showing that reinnervated afferences had an
increased length in threshold, probably due to an incomplete recovery of gamma
motoneurons drive to reinnervated muscle spindles [95].
In conclusion, mechanoreceptors in normal tissues contribute to stability of the
joints. Injuries of ligaments, tendons, and muscles contribute to the mechanical
instability and to a disturbance in neuromuscular control due to the loss of
mechanoreceptors. This field remains to be investigated more deeply in the way to
clarify the complex relationship between soft tissues and nervous system in normal
and pathological conditions. Further studies need to be conducted on mechanore-
ceptor functions in normal and pathological conditions and in the close future
maybe our surgical procedures; rehabilitation protocols for conservative or post-
surgical treatments will become modified if proprioception sense will be better
addressed [96].
7 Nanoscale Architecture for Controlling Cellular … 179
Nanoscale Biotechnology
Although further studies and new techniques are necessary, there are already dif-
ferent types of nanoscale biotechnology studied and experimented.
Electrospinning. Electrospinning has emerged to be a simple, elegant, and
scalable technique to fabricate polymeric nanofibers. Pure polymers as well as
blends and composites of both natural and synthetics have been successfully
electrospun into nanofiber matrices. Physiochemical properties of nanofiber
matrices can be controlled by manipulating electrospinning parameters to meet the
requirements of a specific application. Such efforts include the fabrication of fiber
matrices containing nanofibers, microfibers, combination of nano- and microfibers,
and also different fiber orientations/alignments. Polymeric nanofiber matrices have
been extensively investigated for diversified uses such as filtration, barrier fabrics,
wipes, personal care, biomedical and pharmaceutical applications. Recently elec-
trospun nanofiber matrices have gained a lot of attention and are being explored as
scaffolds in tissue engineering due to their properties that can modulate cellular
behavior. Electrospun nanofiber matrices show morphological similarities to the
natural extracellular matrix (ECM), characterized by ultrafine continuous fibers,
high surface-to-volume ratio, high porosity, and variable pore-size distribution.
Efforts have been made to modify nanofiber surfaces with several bioactive
molecules to provide cells with the necessary chemical cues and a more in
vivo-like environment. The current paper provides an overlook on such efforts in
designing nanofiber matrices as scaffolds in the regeneration of various soft tissues
including skin, blood vessel, tendon/ligament, cardiac patch, nerve, and skeletal
muscle [97].
Nanofiber scaffolds. Nanofiber scaffolds, produced by the electrospinning
technique, have gained widespread attention in tissue engineering due to their
morphological similarities to the native extracellular matrix. For cartilage repair,
studies have examined their feasibility; however, these studies have been limited,
excluding the influence of other scaffold design features. It evaluated the effect of
scaffold design, specifically examining a range of nano- to micron-sized fibers and
resulting pore size and mechanical properties, on human mesenchymal stem cells
(MSCs) derived from the adult bone marrow during chondrogenesis. MSC differ-
entiation was examined on these scaffolds with an emphasis on temporal gene
expression of chondrogenic markers and the pluripotent gene, Sox2, which has yet
to be explored for MSCs during chondrogenesis and in combination with tissue
engineering scaffolds. Chondrogenic markers of aggrecan, chondroadherin, sox9,
and collagen type II were highest for cells on micron-sized fibers (5 and 9 lm) with
pore sizes of 27 and 29 lm, respectively, in comparison with cells on nano-sized
fibers (300 nm and 600 to 1400 nm) having pore sizes of 2 and 3 lm, respectively.
Undifferentiated MSCs expressed high levels of the Sox2 gene but displayed
negligible levels on all scaffolds with or without the presence of inductive factors,
suggesting that the physical features of the scaffold play an important role in
differentiation. Micron-sized fibers with large pore structures and mechanical
180 F. Oliva et al.
tissue engineering research has focused on developing and utilizing scaffolds with
similar nanoscale architecture. However, the current literature lacks consensus
regarding the ideal fiber diameter, with differences in culture conditions making it
difficult to compare between studies. It was studied how to develop a more thor-
ough understanding of how cell–cell and cell–biomaterial interactions drive in vitro
chondrogenic differentiation of bone-marrow-derived mesenchymal stem
cells (MSCs). Electrospun poly(e-caprolactone) microfibers (4.3 ± 0.8 µm diam-
eter, 90 lm2 pore size) and nanofibers (440 ± 20 nm diameter, 1.2 lm2 pore size)
were seeded with MSCs at initial densities ranging from 1 105 to 4 106 cells
cm−3 -scaffold and cultured under transforming growth factor-b (TGF-b) induced
chondrogenic conditions for 3 or 6 weeks. Chondrogenic gene expression, cellular
proliferation, as well as sulfated glycosaminoglycan and collagen production were
enhanced on microfiber in comparison with nanofiber scaffolds, with high initial
seeding densities being required for significant chondrogenic differentiation and
extracellular matrix deposition. Both cell–cell and cell–material interactions appear
to play important roles in chondrogenic differentiation of MSCs in vitro, and
consideration of several variables simultaneously is essential for understanding cell
behavior in order to develop an optimal tissue engineering strategy [101].
The objective of the scientific studies is to develop a scaffold for mesenchymal
stromal cell (MSC) recruitment, proliferation, and chondrogenic differentiation. The
concept behind the design is to mimic the cartilage matrix and contain stimulatory
agents that make continuous supply of inductive factors redundant.
The use of electrospun extracellular matrix (ECM)-mimicking nanofibrous
scaffolds for tissue engineering is limited by poor cellular infiltration. Cell pene-
tration could be enhanced in scaffolds by using a hierarchical structure where
nanofibers are combined with micron-scale fibers while preserving the overall
scaffold architecture. To assess this, were fabricated electrospun porous poly(lactic
acid) (PLA) scaffolds having nanoscale, microscale, and combined micro/
nanoarchitecture and evaluated the structural characteristics and biological
response in detail. Although the bioactivity was intermediate to that for nanofiber
and microfiber scaffold, a unique result of this study was that the micro/nano
combined fibrous scaffold showed improved cell infiltration and distribution than
the nanofibrous scaffold. Although the cells were found to be lining the scaffold
periphery in the case of nanofibrous scaffold, micro/nanoscaffolds had cells dis-
persed throughout the scaffold. Further, as expected, the addition of nanoparticles of
hydroxyapatite (nHAp) improved the bioactivity, although it did not play a sig-
nificant role in cell penetration. Thus, this strategy of creating a three-dimensional
(3D) micro/nanoarchitecture that would increase the porosity of the fibrous scaffold
and thereby improving the cell penetration can be utilized for the generation of
functional tissue-engineered constructs in vitro [102].
Thus, it is possible to combine nanofibrous (N: *400 nm) and microfibrous (M:
*10 lm) poly-e-caprolactone (PCL) scaffolds; for example, they were combined
with 1% high-molecular-weight sodium hyaluronate (NHA/MHA), 1% hyaluronan
(HA) and 200 ng transforming growth factor beta 1 (TGF-b1; NTGF/MTGF), or
0.1% bovine serum albumin (N/M). Scaffolds were seeded with MSCs from bone
182 F. Oliva et al.
marrow and cultured without growth factors in vitro. Cultures with chondrogenic
medium supplemented with TGF-b1 served as controls. Proliferation, migration,
and release of TGF-b1 were investigated. Cell differentiation was evaluated by
polymerase chain reaction (PCR) and real-time PCR. NTGF and MTGF exhibited
primarily an initial release of TGF-b1. None of the factors released by the scaffolds
recruited MSCs. The expression of aggrecan was dependent on the scaffold ultra-
structure with nanofibers promoting increasing and microfibers decreasing
expression levels. Composites containing HA demonstrated elevated seeding effi-
ciency and lower type I collagen expression. Expression of type II collagen was
dependent on continuous or late supply of TGF-b1, which was not provided by the
scaffold design. The initial release of TGF-b1 induced an expression of type I
collagen and osteogenic marker genes. Thus, nanofibrous PCL scaffolds with or
without augmentation are suitable for chondrogenic initiation of MSCs. Initial
release of HA is sufficient in terms of directing the implanted MSCs toward a
chondrogenic end, whereas a late release of TGF-b1 is preferred to foster type II
and avoid type I collagen expression [103].
Hyaluronic acid (HA) is a molecule of particular interest for producing scaffold
for tissue engineering. It has been reported that bone marrow (BM) is the MSC
source an appropriate in the setting of cartilage regeneration to treat cartilage focal
lesions. Thus, can be used HA membranes reached of BM-MSCs, aspirated by the
posterior iliac crest, as scaffolds. The interaction of MSCs with the scaffolds such as
HA membranes may contribute to positively influence the differentiation process
toward chondrogenesis. Many other factors may help MSC differentiation into
chondrocytes as the spatial conformation of the culture system, additional reagents
as platelet rich plasma (PRP), PL or TGF-b1, but the influence of the structure and
composition of the scaffold is particularly relevant since it could provide favorable
microenvironment for MSC chondrogenesis [104].
PRP and MSCs for tendon regeneration. PRP and mesenchymal stem cells can
be used also for tendon injuries. Tendon tissue shows limited regeneration potential
with formation of scar tissue and inferior mechanical properties. Several growth
factors have the capacity to improve the healing response and decrease scar for-
mation. PRP and MSC work as efficient growth factor carriers. Platelets contained
in PRP produce a number of relevant cytokines participating in physiological
tendon healing (e.g., platelet-derived growth factor (PDGF), epidermal growth
factor (EGF), vascular endothelial growth factor (VEGF)). The angiogenic effect is
vital for tendon and ligament healing, and PRP may contribute to increase it. In
vitro experiments and animal studies showed promising results for the use of PRP;
however, clinical controlled studies have shown a tendency to reduce the
pain-related symptoms but no significant differences in overall clinical scores.
Unfortunately is yet unknown which dose and which type of PRP should be used to
have more significant results. On the other hand, MSCs can contribute to tendon
healing in different ways. First, they can provide tenocytes by direct differentiation
into these cells. Secondly, they can provide a number of anabolic cytokines by their
extraordinary paracrine activity. Thirdly, they show significant anti-inflammatory
activity that may contribute to the healing process. MSCs are not totally arrived in
7 Nanoscale Architecture for Controlling Cellular … 183
clinical use so that there is still a lack of randomized controlled trials. In basic
research experiments, they show an extraordinary paracrine activity,
anti-inflammatory effect, and the possibility to differentiate in tenocytes when
different activating factors are added [105].
PAM and SCF. For some specific tissue regeneration, to overcome problems
such as cell survival, lack of cell differentiation, and integration in the host tissue, a
new tool described as pharmacologically active microcarriers (PAM) has been
described. PAMs are biocompatible and biodegradable microdevices coated with
cell adhesion molecules, conveying cells on their surface and presenting a con-
trolled delivery of growth factor. For other specific tissue regeneration, micro- and
nanoporous scaffolds with a specific 3D-shape serve as temporary support for cells
to grow into a new tissue, before it is transplanted back to the host tissue. PAM
production can be performed by using an innovative continuous process that
involves the use of supercritical fluid (SCF) (mainly supercritical carbon dioxide),
for the treatment of simple or multiple emulsions at relative low temperatures.
Scaffolds with a specific 3D shape require several characteristics, such as high
regular and reproducible 3D structure, porosity exceeding 90% and an open pore
geometry, high internal surface areas, and specific mechanical properties. To pro-
duce interconnected microcells and a nanometric substructure, a new
supercritical-fluid-assisted technique for the formation of 3D scaffolds has been
proposed. It consists of three steps: formation of a polymeric gel loaded with a solid
porogen, drying of the gel using SCF, washing with water to eliminate the porogen.
When gel drying is performed by SCF, the supercritical mixture formed during the
process has no surface tension and can be easily eliminated in a single step. One of
the major problems in gel drying is the possibility of gel collapse; using SCF, the
absence of surface tension avoids this problem preserving the nanoporous structure.
“Injectable scaffold” or temporary scaffold was also tested, respectively, for the
culture of mesenchymal stem cell in order to study their growth and differentiation.
The result showed higher biocompatibility with respect to similar product obtained
by conventional technology [106–110].
Conclusions
References
1. Ingber DE. Integrins as mechanochemical tranducers. Curr Opin Cell Biol. 1991;3:841–8.
2. Ingber DE. Tensegrity: the architectural basis of cellular mechanotranduction. Ann Rev
Physiol. 1997;59:575–99.
3. Rodan GA, Bourret LA, Harvey A, Mensi T. Cyclic AMP and cyclic GMP: mediators of the
mechanical effects on bone remodeling. Science. 1975;189:467–9.
4. Davies PF, Remuzzi A, Gordon EJ, Dewey CF, Gimbrone MA. Turbulent fluid shear stress
induces vascular endothelial cell turnover in vitro. Proc Nat Acad Sci USA. 1986;83:
2114–7.
5. Jones DB, Scholubbers J-G. Evidence that phospholipase C mediates the mechanical stress
response in bone. Calc Tiss Int. 1987;41(Suppl. 2):4.
6. Brighton CT, Strafford B, Gross SB, Leatherwood DF, Williams JL, Pollack SR. The
proliferative and synthetic response of isolated calvarial bone cells of rats to cyclic biaxial
mechanical strain. J Bone Joint Surg. 1991;73A:320–31.
7. Sah RL-Y, Grodizinsky AJ, Plass AHK, Sandy JD. Effects of static and dynamic
compression on matrix metabolism in cartilage explants. In: Kuettner K, et al. editors.
Articular Cartilage and Osteoarthritis. New York: Raven Press, Ltd; 1992:373–92.
8. Bachrach NM, Valhmu WB, Stazzone E, Ratcliffe A, Lai WM, Mow VC. Changes in
proteoglycan synthesis rates of chondrocytes in articular cartilage are associated with the
time dependent changes in their mechanical environment. J Biomechanics. 1995;28:1561–9.
9. Guilak F, Sah RL-Y, Setton LA. Physical regulation of cartilage metabolism. In: Mow VC,
Hayes WC, editors. Basic orthopaedic biomechanics. Philadelphia: Lippincott-Raven; 1997.
p. 179–208.
10. Comper WD editor. Extracellular Matrix. Amsterdam, The Netherlands: Harwood Academic
Press 1996, Vols I & II.
11. Sung KLP, Sung LA, Crimmins M, Burakoff SJ, Chien S. Dynamic changes in viscoelastic
properties in cytotoxic T-lymphocytes mediating killing. J Cell Sci. 1988;91:179–89.
12. Dong C, Skalak R, Sung KP, Schmid-Schonbien GW, Chien S. Passive deformation analysis
of human leukocytes. J Biomech Eng. 1998;110:27–36.
13. Chen Christopher S, Ingber Donald E. Tensegrity and mechanoregulation: from skeleton to
cytoskeleton. Osteoarthritis Cartilage. 1999;7:81–94.
14. Viidik A. Tensile strength properties of achilles tendon systems in trained and untrained
rabbits. Acta Orthop Scand. 1969;40:261–72.
15. Tipton CM, Matthes RD, Maynard JA, et al. The influence of physical activity on ligaments
and tendons. Med Sci Sports. 1975;7:165–75.
16. Lanyon LE, Rubin CT. Static versus dynamic loads as an influence on bone remodelling.
J Biomech. 1984;17:897–905.
17. Frost HM. Vital biomechanics. General concepts for structural adaptations to mechanical
usage. Calc Tiss Int. 1987;42:145–54.
18. DeLee JC. Tissue remodeling and response to therapeutic exercise [review]. In:
Leadbetter WB, Buckwalter JA, Gordon SL, editors. Sports-Induced Inflammation.
AAOS: American Orthopaedic Society; 1989. p. 547–54.
19. Forwood MR, Turner CH. Skeletal adaptations to mechanical usage: results from tibial
loading studies in rats. Bone. 1995;4(suppl):197S–205S.
20. Deng M, James R, Laurencin CT, Kumbar SG. Nanostructured polymeric scaffolds for
orthopaedic regenerative engineering. IEEE Trans Nanobiosci. 2012;11(1):3–14. https://doi.
org/10.1109/TNB.2011.2179554 Epub 2012 Jan 23.
21. Ingber DE, Wang N, Stamenovic D. Tensegrity, cellular biophysics, and the mechanics of
living systems. Rep Prog Phys. 2014;77(4):046603.
22. Frost HM. A 2003 update of bone physiology and Wolff’s Law for clinicians. Angle Orthod.
2004;74(1):3–15.
7 Nanoscale Architecture for Controlling Cellular … 185
23. Mow VC, Ratcliffe A, Poole AR. Cartilage and diarthrodial joints as paradigms for
hierarchical materials and structures. Biomaterials. 1992;13:67–97.
24. Martin RB, Burr DB. Structure, function, and adaptation of compact bone. New York:
Raven Press; 1989.
25. Silbert JE. Advances in the biochemistry of proteoglycans. In: Uitto J, Perejda AJ, editors.
Connective tissue disease: molecular pathology of the extracellular matrix. New York:
Marcel Dekker; 1987. p. 83–98.
26. Mow VC, Ratcliffe A. Structure and function of articular cartilage. In: Mow VC, Hayes WC,
editors. Basic orthopaedic biomechanics. Philadelphia, PA: Lippincott-Raven; 1997. p. 113–
77.
27. Guilak F. Compression-induced changes in the shape and volume of the chondrocyte
nucleus. J Biomech. 1995;28:1529–41.
28. Guilak F, Ratcliffe A, Mow VC. Chondrocyte deformation and local tissue strains in
articular cartilage: a confocal microscopy study. J Orthop Res. 1995;13:410–21.
29. Nimni ME. Collagen: structure, function, and metabolism in normal and fibrotic tissues. Sem
Arth Rheum. 1983;13:1–86.
30. Woo SL-Y, Gomez MA, Woo YK, Akeson WH. Mechanical properties of tendons and
ligaments. Quasistatic and nonlinear viscoelastic properties. Biorheology 1982;19:385–96.
31. Dickinson RB, Guido S, Tranquillo RT. Biased cell migration of fibroblasts exhibiting
contact guidance in oriented collagen gels. Ann Biomed Eng. 1994;22:342–56.
32. Chen BM, Grinnell AD. Integrins and modulation of transmitter release from motor nerve
terminals by stretch. Science. 1995;269:1578–80.
33. Hamill OP, McBride DW Jr. The cloning of a mechano-gated membrane ion channel.
Trends Neurosci. 1994;17:439–45.
34. Ingber DE, Dike L, Hansen L, Karp S, Liley H, Maniotis A, McNamee H, Mooney D,
Plopper G, Sims J, Wang N. Cellular tensegrity: exploring how mechanical changes in the
cytoskeleton regulate cell growth, migration, and tissue pattern during morphogenesis. Int
Rev Cytol. 1994;150:173–224.
35. Plopper G, McNamee H, Dike L, Bojanowski K, Ingber DE. Convergence of integrin and
growth factor receptor signaling pathways within the focal adhesion complex. Mol Biol Cell.
1995;6:1349–65.
36. Miyamoto S, Akiyama S, Yamada KM. Synergistic roles for receptor occupancy and
aggregation in integrin transmembrane function. Science. 1995;267:883–5.
37. Buxbaum RE, Heidemann SR. A thermodynamic model for force integration and
microtubule assembly during axonal elongation. J Theor Biol. 1988;134:379–90.
38. Yao L, Bestwick CS, Bestwick LA, Maffulli N, Aspden RM. Phenotypic drift in human
tenocyte culture. Tissue Eng. 2006;12(7):1843–9.
39. Ingber DE. Tensegrity-based mechanosensing from macro to micro. Prog Biophys Mol Biol.
2008;97(2–3):163–79. https://doi.org/10.1016/j.pbiomolbio.2008.02.005.
40. Cornelius C, Koverech G, Crupi R, Di Paola R, Koverech A, Lodato F, Scuto M,
Salinaro AT, Cuzzocrea S, Calabrese EJ, Calabrese V. Osteoporosis and Alzheimer
pathology: role of cellular stress response and hormetic redox signaling in aging and bone
remodeling. Front Pharmacol. 2014;5.
41. Barbieri E, Sestili P, Vallorani L, Guescini M, Calcabrini C, Gioacchini AM, Annibalini G,
Lucertini F, Piccoli G, Stocchi V. Mitohormesis in muscle cells: a morphological, molecular,
and proteomic approach. MLTJ. 2013.
42. Ji LL, Gomez-Cabrera MC, Vina J. Exercise and hormesis: activation of cellular antioxidant
signaling pathway. Ann N Y Acad Sci. 2006;1067:425–35.
43. Rattan SI. Targeting the age-related occurrence, removal, and accumulation of molecu-
lar damage by hormesis. Ann N Y Acad Sci. 2010;1197:28–32. https://doi.org/10.1111/j.
1749-6632.2010.05193.x.
44. Hamel P, Abed E, Brissette L, Moreau R. Characterization of oxidized low-density
lipoprotein-induced hormesis-like effects in osteoblastic cells. Am J Physiol Cell Physiol.
2008;294:C1021–33.
186 F. Oliva et al.
45. Delaine-Smith R, Reilly GC. Mesenchymal stem cell responses to mechanical stimuli.
MLTJ. 2012.
46. Baksh D, Song L, Tuan RS. Adult mesenchymal stem cells: characterization, differentiation,
and application in cell and gene therapy. J Cell Mol Med. 2004;8(3):301–16.
47. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA,
Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal
stem cells. Science. 1999;284(5411):143–7.
48. Hao W, Dong J, Jiang M, Wu J, Cui F, Zhou D. Enhanced bone formation in large segmental
radial defects by combining adipose-derived stem cells expressing bone morphogenetic
protein 2 with nHA/RHLC/PLA scaffold. Int Orthop. 2010;34(8):1341–9.
49. Whu SW, Tsai CL, Hsu SH. Evaluation of human bone marrow mesenchymal stem cells
seeded into composite scaffolds and cultured in a dynamic culture system for neocartilage
regeneration in vitro. J Med Biol Eng. 2009;29(2):52–9.
50. Kratchmarova I, Blagoev B, Haack-Sorensen M, Kassem M, Mann M. Mechanism of
divergent growth factor effects in mesenchymal stem cell differentiation. Science. 2005;308
(5727):1472–7.
51. Watanabe T, Sakai D, Yamamoto Y, Iwashina T, Serigano K, Tamura F, Mochida J. Human
nucleus pulposus cells significantly enhanced biological properties in a co-culture system
with direct cell-to-cell contact with autologous mesenchymal stem cells. J Orthop Res.
2010;28(5):623–30.
52. Yu H, Vandevord PJ, GongW WuB, Song Z, Matthew HW, Wooley PH, Yang SY.
Promotion of osteogenesis in tissue-engineered bone by pre-seeding endothelial progenitor
cells-derived endothelial cells. J Orthop Res. 2008;26(8):1147–52.
53. Ozturk AM, Cila E, Kanatli U, Isik I, Senkoylu A, Uzunok D, Piskin E. Treatment of
segmental bone defects in rats by the stimulation of bonemarrow osteo-progenitor cells with
prostaglandin E2. Int Orthop. 2005;29(2):73–7.
54. Chen KY, Chung CM, Kuo SM, Chen YS, Yao CH. Influence of collagen I nanospheres on
the growth and osteogenic difference of rat bone marrow stromal cells. J Med Biol Eng.
2009;29(6):284–9.
55. Jung Y, Song J, Shiozawa Y, Wang J, Wang Z, Williams B, Havens A, Schneider A, Ge C,
Franceschi RT, McCauley LK, Krebsbach PH, Taichman RS. Hematopoietic stem cells
regulate mesenchymal stromal cell induction into osteoblasts thereby participating in the
formation of the stem cell niche. Stem Cells. 2008;26(8):2042–51.
56. Borovecki F, Pecina-Slaus N, Vukicevic S. Biological mechanisms of bone and cartilage
remodelling-genomic perspective. Int Orthop. 2007;31(6):799–805.
57. Li M, Thompson DD, Paralkar VM. Prostaglandin E-2 receptors in bone formation. Int
Orthop. 2007;31(6):767–72.
58. Tsai MT, Lin DJ, Huang S, Lin HT, Chang WH. Osteogenic differentiation is synergistically
influenced by osteoinductive treatment and direct cell–cell contact between murine
osteoblasts and mesenchymal stem cells. Int Orthop (SICOT). 2012;36:199–205.
59. Kim H, Lee JH, Suh H. Interaction of mesenchymal stem cells and osteoblasts for in vitro
osteogenesis. Yonsei Med J. 2003;44(2):187–97.
60. Ahn SE, Kim S, Park KH, Moon SH, Lee HJ, Kim GJ, Lee YJ, Park KH, Cha KY,
Chung HM. Primary bone-derived cells induce osteogenic differentiation without exogenous
factors in human embryonic stem cells. Biochem Biophys Res Commun. 2006;340(2):
403–8.
61. Delaine-Smith R, Reilly GC. Mesenchymal stem cell responses to mechanical stimuli.
MLTJ. 2012.
62. Ytteborg E, Torgersen JS, Pedersen ME, Helland SJ, Grisdale-Helland B, Takle H. Exercise
induced mechano-sensing and substance P mediated bone modeling in Atlantic salmon.
Bone. 2013;53(1):259–68. https://doi.org/10.1016/j.bone.2012.11.025 Epub 2012 Dec 3.
63. Oliva F, Tarantino U, Maffulli N. Immunohistochemical localization of calcitonin
gene-related peptide and substance P in the rat knee cartilage at birth. Physiol Res.
2005;54:549–56.
7 Nanoscale Architecture for Controlling Cellular … 187
108. Cardea S, Pisanti P, Reverchon E. Generation of chitosan nanoporous structures for tissue
engineering applications using a supercritical fluid assisted process. J Supercrit Fluids.
2010;54:290–5.
109. Della Porta G, Cavalcanti S, Reverchon E. Supercritical Fluid emulsions extraction: a novel
technology for the production of poly-lactic-co-glycolic nanostructured microdevices. In:
Proceedings of II Congresso Nazionale di Bioingegneria; 2010 July 8–10; Turin (Italy):
Patron, p. 637–8, ISBN 978-88-555-3082-8.
110. Reverchon E, Adami R, Cardea S, Della Porta G. Supercritical fluids processing of polymers
for pharmaceutical and medical applications. J Supercrit Fluids. 2009;47:484–92.
Chapter 8
Modular Tissue Engineering:
An Artificial Extracellular Matrix
to Address and Stimulate Regeneration/
Differentiation
populate the scaffold with the aid of dynamic cultivation, often supplemented with
growth factors, generating an appropriate extracellular matrix (ECM) [4–6]. These
3D scaffolds are still far from leading to successful tissue reconstruction in a clinical
setting because of the absence of specific microarchitecture, a limited mass trans-
port of metabolites or oxygen through the 3D structure, a reduced control over their
biodegradation. Moreover, when bio-signals or growth factors are incorporated by
simple adsorption onto bulk scaffolds, they normally leads to uncontrolled burst
release on implantation with an overdose that may give rise to undesired effects
[7, 8].
More recent tissue engineering approach described as “bottom-up” one or
modular tissue engineering aims to address the challenge of recreating bio-mimetic
structures by designing structural microfeatures that can be used to build modular
tissues such as building blocks to create larger tissues (see Fig. 8.2). These 3D
modules can be fabricated in different ways, such as microfabrication of cell-laden
hydrogels [9, 10], creation of cell sheets [11], or direct tissue printing [12]. By
mimicking native microstructural functional units, these approaches aim to create
more bio-mimetic engineered tissues because these modules can be assembled into
larger tissues-like structure through a number of methods such as random packing
[13, 14], stacking of layers [15], or ordinate assembly [16].
Bottom-up approach is aimed to provide guidance on the cellular level to direct
tissue morphogenesis by creating modular tissues with more physiological
microarchitectural features [17]. In addition, bio-plotting technology can help this
approach by designing a zonal organization into the 3D engineered artificial matric
plotted; for example, one strategy may be to spatially pattern cells with specific
additives from different zones into stratified layers to better mimic the in vivo zonal
organizations of the extracellular matrix. In this sense, bio-plotting additives such as
Fig. 8.1 Top-down approach involves monolithic scaffold on which the cells are seeded; often,
cells do not colonize the interior part of the scaffold
8 Modular Tissue Engineering … 193
Fig. 8.2 Bottom-up approach allows generating scaffolds of higher complexity and functionality
that can be designed with several advantages over conventional monolithic scaffolds
designed with several advantages over conventional monolithic bulk scaffolds [18, 19].
Moreover, by introducing microspheres, mechanically weak scaffolds can be sta-
bilized or reinforced providing an improved mechanical behaviour [20, 21].
Biopolymer microspheres with peptides immobilized on the surface have been
also reported as a mouldable scaffold for cartilage tissue engineering [22, 23],
whereas gelatin microspheres with bioactive domains including peptides resulted in
cytocompatible microcarriers suitable to deliver live cells such as chondrocytes for
in vivo chondrogenesis [24, 25]. During the dynamic cultivation, cells adhered,
proliferated and synthesized a thin layer of extracellular matrix (ECM) in and
around the macroporous beads allowing a biological sintering via cell–cell and
cell–matrix interaction after only a few days of dynamic seeding [26].
Natural polymers are of great importance for tissue engineering, basically due to
their intrinsic biocompatibility and biodegradability. This class of polymers is often
favoured more than synthetic ones due to their abundant side groups in their
molecular chains that allow for further functionalization. Further, natural polymers
such as collagen and gelatine contain motifs such as RGD (arginine–glycine–as-
partic acid) sequences, which can modulate cell adhesion and improve their
behaviour. Collagen is the most extensively investigated natural polymer since the
ECM of many mesenchymal tissues (including bone and cartilage) is mainly
composed of collagen as organic phase. Collagen is an attractive candidate material
for bone regeneration due to its excellent biocompatibility, desirable biodegrad-
ability and negligible immunogenicity [27, 28]. As a derivative from collagen,
Gelatin has been also used for biomedical applications; its characteristics include a
controllable degradation, and abundant presence of functional groups that allow for
further functionalization and modification via chemical derivatization. Specifically,
the unique electrical nature of gelatin (commercially available as both positively or
negatively charged polymers at neutral pH) enables gelatin to encapsulate bioactive
molecules by forming polyion complexes [29, 30].
Fibrin can be prepared by combining fibrinogen with thrombin, which are both
derived from the patient’s own blood, thereby fabricating an autologous scaffold
that does not induce an excessive foreign body reaction. However, the rapid
degradation rate and poor mechanical stability of fibrin have been stated as the main
limitation for bone tissue engineering. To overcome this problem, composite
microspheres (alginate/fibrin, poly-hyaluronic acid/fibrin) were developed to sta-
bilize the fibrin matrix, solving the problems of both alginate shortage of bioactive
sequence for cell attachment and fibrin poor capacity for cell encapsulation [31, 32].
Chitosan is a frequently used hydrophilic polysaccharide derived from chitin,
which exhibits favourable physicochemical and biological properties for biomedical
8 Modular Tissue Engineering … 195
suspension is cast into the mould of a desired shape; after solvent evaporation or
lyophilization, the salt particles are leached out to obtain biopolymer porous 3D
structure with porosity that can be of 90%. Closed cavities in the resulting matrix
and smooth wall are the described drawbacks of these structures as well as complex
post-treatments to remove the residual amount of solvents used [43]; moreover,
poor metabolites mass transfer within the structure during the cell cultivation and
poor cell colonization inside the structure is often described for these scaffolds,
even if cultivated in dynamic conditions [44, 45]. Electrospinning is a more
advanced fabrication technology that uses an electric field to control the formation
and deposition of biopolymer microfibres onto a target substrate (see Fig. 8.4).
A critical voltage is applied in order to overcome the surface tension of the
biopolymer solution and obtain an electrically charged jet; the jet within the electric
field is, then, directed towards the ground target, during which time the solvent
evaporates forming oriented or random fibres with diameters ranging from several
microns down to several hundred nanometres [46]. Limitations described are: low
mechanical strength and difficulty in controlling the 3D structure pore shapes and
sizes. Rapid prototyping is a technology based on the development of computer
science and manufacturing industry. The main advantage of these techniques is
their ability to produce complex products rapidly from a computer-aided design
(CAD) model. One of these rapid prototyping techniques, called 3D printing, has
been used to process biodegradable polymer scaffolds for tissue engineering
applications [47, 48]. All the described technologies lack biological agents incor-
porating within the biopolymer structure; for example, peptides such as growth
factors cannot be easily loaded into the 3D scaffolds because of the extreme con-
ditions (temperature/organic solvents) used in the fabrication process.
Technologies that replace the use of an organic solvent with dense gases or
supercritical fluids are also emerging in fabricating 3D devices for biomedical
applications. Supercritical fluids (SCFs) are defined substances at a temperature and
pressure above their critical point as described by the pressure vs. temperature
diagram for a pure compound (see Fig. 8.5). In a specific pressure and temperature
conditions, a substance becomes supercritical and can be described as an homo-
geneous fluid that can effuse through solids like a gas, but has also the ability to
Fig. 8.3 Solvent casting and particulate leaching method and SEM image of the microstructure of
the 3D scaffold obtainable
8 Modular Tissue Engineering … 197
Fig. 8.4 Schematic representation of electrospinning method and SEM image of the microstruc-
ture of the 3D scaffold obtainable
dissolve materials like a liquid. In addition, close to the critical point, small changes
in pressure or temperature result in large changes in density, allowing a fine tuning
of the supercritical fluid solvent power. As a consequence, the supercritical fluids
seemed suitable to substitute organic solvents in a wide range of industrial pro-
cesses [49]. Particularly, its reduced surface tension coupled with higher diffusivity
(that is similar to gases and of about two orders of magnitude larger than that of
liquid solvents) offers possibilities of fabrication structures not obtainable by using
the conventional liquid solvents. Carbon dioxide is the most commonly used
supercritical fluids (SC-CO2) because it is not toxic, not flammable, cheap and with
critical parameters of pressure and temperature are readily accessible on the
industrial scale (304.1 K and 73.8 bar).
The first use of SC-CO2 for temporary 3D scaffolds production has been sug-
gested with the gas-foaming technique. The process is solventless and very efficient
in producing the porous biopolymer structure because the dense gas can foam a
given biopolymer inducing the formation of bubbles within its structure (see
Fig. 8.6). It was also described that the morphology of the foams can be varied by
controlling the depressurization rate of the foamed biopolymer, such as PLA or
PLGA, and the density of the gas; particularly, a rapid depressurization locked in
large numbers of spherical pores within the biopolymer structure, whereas a slower
depressurization reduced pores elongation. Drawbacks of the gas-foaming process
are smooth surfaces and generally closed pores structures [50]. Another technology
described for scaffold production by dense gases is the supercritical assisted phase
inversion method (SAPIM); the process reproduces the traditional phase inversion
method (obtained by liquid–liquid diffusion or by varying the system temperature)
by using SC-CO2. Therefore, the dense gas is used to dry the biopolymer structure
more rapidly due to an enhanced mass transfer caused by a reduced surface tension
and the absence of liquid–vapour interface. In a specific condition of pressure and
temperature and system composition, open pores are obtained and no traces of
organic solvents are described in the resulted scaffold. Moreover, the use of
SC-CO2 by means of pressure introduces an additional variable to influence the
biopolymer/solvent solution de-mixing process and, therefore, the porous structure
morphology. Indeed, it was described that it is possible to control the average pore
size that can decrease with the increase of SC-CO2 density (either by increasing the
pressure or by decreasing the temperature), whereas the average pore diameter
decreased with the increase of the initial polymer concentration [51]. Reverchon &
Cardea [52] explained this behaviour indicating that the phase separation process
can be responsible for the porous cellular structure obtainable in depending on the
liquid/liquid de-mixing by nucleation and growth of droplets of a polymer lean
phase. The subsequent removal of the solvent produces a dry and stable structure.
The proposed process is versatile, and the scaffold characteristics can be continu-
ously modulated varying supercritical CO2 properties. Limitations are again smooth
porous walls and poorly interconnected pores within the 3D structure. To fulfil the
necessity of producing interconnected micropores, a supercritical fluid-assisted
technique named supercritical gel drying has been also proposed and described
[53]. The process involves the formation of a biopolymer alcohol gel loaded with a
solid porogen that is then dried by SC-CO2; the final washing with water eliminates
the porogen fabricating a porous structure. When the gel drying is performed by
supercritical CO2, the supercritical mixture formed during the process (ethanol plus
CO2) has no surface tension and can be easily eliminated in a single step by the
continuous flow of SC-CO2 in the drying vessel. The major problem during gel
drying is the possibility of gel collapse; however, the reduced surface tension of the
supercritical mixture used to avoid this problem preserving the nanoporous struc-
ture in the fabricated 3D aerogel [54]. These scaffolds were successfully tested for
human mesenchymal cells cultivation in a specific dynamic cultivation [55].
8 Modular Tissue Engineering … 199
Fig. 8.6 Schematic representation of supercritical gas-foaming method with SEM image of the
microstructure of the 3D scaffold obtainable
Several techniques can be used to prepare biopolymer microcarriers, but the solvent
evaporation/extraction of emulsion (single, double or multiple) is the most widely
used. The process involves single oil-in-water (o/w) or double water-in-oil-in-water
(w/o/w) emulsions to encapsulate biomolecules [56]. The biopolymer is first dis-
solved in a water-immiscible organic solvent. The bioactive molecule is then added
to the polymer solution to produce a solution or dispersion of the substance;
however, more often, a double emulsion is prepared that involves the formulation of
a water-in-oil (w/o) primary emulsion to encapsulate water-soluble molecules like
peptides or proteins, unlike the o/w formulation which is ideal for water-insoluble
compounds [56]. The primary w/o emulsion is, then, emulsified (with appropriate
stirring and temperature conditions) in a larger volume of water in presence of an
emulsifier to yield the final w/o/w emulsion. The emulsion is subjected to solvent
removal by either evaporation or extraction process to harden the oil droplets [57].
The solid microspheres obtained are, then, washed and collected by filtration or
centrifugation and then dried under appropriate conditions or lyophilized. One
of the disadvantages of the emulsification method is the poor encapsulation
efficiencies of moderately water-soluble and water-soluble compounds. Indeed,
these molecules could diffuse out or partition from the dispersed oil phase into the
aqueous continuous phase, and their microcrystalline fragments can deposit
on the microsphere surface and/or outside of the biopolymer matrix [58].
200 G. Della Porta and E. Reverchon
Moreover, the evaporation has several drawbacks because it requires relatively high
temperatures or reduced pressures and shows batch-to-batch reproducibility dis-
turbances, whereas the solvent extraction uses relatively large amounts of a second
solvent with the subsequent problem of solvent mixtures recovery. Both processes,
also, require quite long processing times (several hours) and, as a consequence,
aggregation phenomena may occur between the droplets, producing microspheres
with a larger poly-dispersity with respect to the droplets in the starting emulsion
[59].
Several supercritical technologies were proposed to produce microparticles, and
they are reviewed elsewhere [60]. Recently, the use of SC-CO2 has also been
proposed for biopolymer micro-/nanocarriers manufacturing, replacing the con-
ventional emulsions evaporation/extraction technology. Particularly, SC-CO2 was
proposed as extracting agent of the oily phase of emulsions to lead to solvent-free
microparticles [61–65]. The supercritical extraction of emulsion (SEE) process was
also proposed with a continuous operating mode that allowed greater product
uniformity, higher throughput with smaller plant volumes and elimination of
batch-to-batch repeatability problems (see Fig. 8.7). Indeed, SC-CO2 assured a
faster oily phase extraction with respect to the traditional evaporation/extraction
processes that will have a significant effect on the size distribution of the
biopolymer microcapsules which reproduce exactly the size distribution of the
originally droplets (with a given shrinkage) because any aggregation phenomena is
prevented due to the faster processing [66, 67]. The supercritical technology
achieved the fabrication of microcarriers of several biopolymers such as PLGA,
PLLA, PCL, PMMA with no solvent residues and higher encapsulation efficiencies
with respect to the conventional extraction/evaporation technology, especially when
double emulsions were processed [68, 69].
Micro-/nanocapsules entrapping magnetic iron oxide or gold nanoparticles have
been also fabricated by SEE technology in order to obtain a remote controlled or
triggered drug delivery [70, 71]. SEE-C technology can also generate multiloaded
microsystems with different molecules such as hydroxyapatite (HA), gentamicin
(GE) and teriparatide hormone (TH), for example, to be used in the bone cement
formulation in the treatment of osteoporosis [72].
Recent tissue engineering strategies suggested the use of 3D scaffold not only to
simply support the cells growth but also to interact with the cells on-board pro-
viding an additional level of cell environment regulation. In this sense, the locally
and controlled release of specific growth factors and/or bio-signals can be a good
approach in order to mimic more properly the microenvironment that the extra-
cellular matrix provides to the cells in order to address their behaviour.
8 Modular Tissue Engineering … 201
Fig. 8.7 Supercritical emulsion extraction (SEE) process layout description with optical
microscope image of an emulsion (left side) and of an SEM image of the microcapsules produced
(right side)
Current fabrication methods for GF-loaded scaffolds can be divided into two
main categories: attachment of the GF (physical and non-physical) to the scaffold or
entrapment of the GF within the scaffold. The attachment may be achieved by
adsorption or through chemical crosslinking. Fibrin-based matrices are popular
biomaterials for this purpose [73, 74], and the release then occurs passively and
through simultaneous fibrin degradation. Entrapment of GFs has been also
202 G. Della Porta and E. Reverchon
Fig. 8.8 Schematic representation of the use of micro-/nanospheres for spatio/temporal delivery
of bioactive molecules. These, pre-designed 3D structure may better reproduce the cells
microenvironment that encourage cells differentiation and ECM production
The bottom-up approaches seemed the best tissue engineering approach choice
because it is able to fabricate a better reproduction of the 3D microenvironment that
interacts with the cells to faster and better address their fate. The modular approach
also seemed to rely on the assembly of micron scale structural elements as the basis
for providing defined signals to drive the formation of tissues that can be scaled up
to create larger tissues. Moreover, the use of biopolymer microcapsules inserted
within the 3D scaffold can allow the design of a specific microenvironment able to
instruct the cell behaviour and to organize a pre-designed microenvironment with
an engineered spatio-temporal delivery of multiple bio-signals.
Biopolymer microspheres are also proposed as regulators of the chondrogenic
microenvironment within high-density cell cultures as three-dimensional structural
elements for cell expansion and delivery or vehicles for spatiotemporally controlled
growth factor delivery, to regulate cell behaviour and/or cell material interactions.
These drug-releasing microspheres have the capability not only to overcome dif-
fusion limitations posed by traditional culture techniques, but also to lend tailorable
release kinetics for temporal control of differentiation and extended maintenance of
differentiation state in vivo.
Furthermore, micro-/nanospheres can serve as reinforcement components or
crosslinking agents to provide hydrogels with additional mechanical support.
Indeed, microspheres loaded with inorganic materials of high stiffness can reinforce
the initial mechanical strength of the composites. For example, biopolymer
microspheres can be used as different “bio-ink additives” added to a 3D bioplotted
hydrogel to give specific mechanical or biochemical indications to the cells in the
different layers following the novel concept of 3D organ printing, in which scaffold/
cellularized tissues or organ can be fabricated layer by using tissue spheroids as
building blocks.
206 G. Della Porta and E. Reverchon
References
23. Kang SW, Jeon O, Kim BS. Poly(lactic-co-glycolic acid) microspheres as an injectable
scaffold for cartilage tissue engineering. Tissue Eng. 2005;11:438–47.
24. Hong Y, Gao CY, Xie Y, Gong YH, Shen JC. Collagen-coated poly-lactide microspheres as
chondrocyte microcarriers. Biomaterials. 2005;26:6305–13.
25. Tan H, Huang D, Lao L, Gao C. RGD modified PLGA/gelatin microspheres as microcarriers
for chondrocyte delivery. J Biomed Mater Res Part B Appl Biomater. 2009;91:228–38.
26. Palmiero C, Imparato G, Urciuolo F, Netti P. Engineered dermal equivalent tissue in vitro by
assembly of microtissue precursors. Acta Biomater. 2010;6(7):2548–53.
27. Lee CH, Singla A, Lee Y. Biomedical applications of collagen. Int J Pharm. 2001;221:1.
28. Li S-T. Biologic biomaterials: tissue-derived biomaterials (collagen). In: Park JB, Bronzino JD,
editors. Biomaterials: principles and applications. Boca Roton, FL: CRC Press; 2003.
29. Schrieber R, Gareis H. Gelatine handbook: theory and industrial practice. Weinheim:
Wiley-VCH Verlag; 2007.
30. Yamamoto M, Ikada Y, Tabata Y. Controlled release of growth factors based on
biodegradation of gelatin hydrogel. J Biomater Sci Polym Ed. 2001;12:77.
31. Ahmed TAE, Dare EV, Hincke M. Fibrin: a versatile scaffold for tissue engineering
applications. Tissue Eng B. 2008;14:199.
32. Perka C, Schultz O, Spitzer R-S, Lindenhayn K, Burmester G-R, Sittinger M. Segmental bone
repair by tissue-engineered periosteal cell transplants with bioresorbable fleece and fibrin
scaffolds in rabbits. Biomaterials. 2000;21:1145.
33. Di Martino A, Sittinger M, Risbud MV. Chitosan: a versatile biopolymer for orthopaedic
tissue-engineering. Biomaterials. 2005;26:5983.
34. Slaughter BV, Khurshid SS, Fisher OZ, Khademhosseini A, Peppas NA. Hydrogels in
regenerative medicine. Adv Mater. 2009;21:3307.
35. Augst AD, Kong HJ, Mooney DJ. Alginate hydrogels as biomaterials. Macromol Biosci.
2006;6:623.
36. Mundargi RC, Babu VR, Rangaswamy V, Patel P, Aminabhavi TM. Nano/micro technologies
for delivering macromolecular therapeutics using poly (D, L lactide-co-glycolide) and its
derivatives. J Control Release. 2008;125:193–209.
37. Verrijk R, Smolde IJ, Bosnie N, Begg AC. Reduction of systemic exposure and toxicity of
cisplatin by encapsulation in poly(lactide-co-glycolide). Cancer Res. 1992;52:6653–6.
38. Meinel L, Illi OE, Zapf J, Malfanti M, Merkle HP, Gander B. Stabilizing insulin-like grow
factor in poly(lactide-co glycolide) microspheres. J Control Release. 2001;70:193–202.
39. Walter E, Dreher D, Kok M, Thiele L, Kiama SG, Gehr P, Merkle HP. Hydrophylic poly
(lactide-co-glycolide) microspheres for the delivery of DNA to human derived macrophages
and dentritic cells. J Control Release. 2001;76:149–68.
40. Luten J, van Nostrum CF, De Smedt SC, Hennink WE. Biodegradable polymers as non-viral
carriers for plasmid DNA delivery. J Control Release. 2008;126:97–110.
41. Jaklenec A, Wan E, Murray ME, Mathiowitz E. Novel scaffolds fabricated from
protein-loaded microspheres for tissue engineering. Biomaterials. 2008;9:185–92.
42. Temenoff JS, Mikos AG, editors. Biomaterials. The intersection of biology and materials
science. NJ: Pearson Prentice Hall; 2008. ISBN 10-0-13-009710-1.
43. Zhang J, Zhang H, Wu L, Ding J. Fabrication of three dimensional polymeric scaffolds with
spherical pores. J Mater Sci. 2006;41:1725–30.
44. Liu X, Ma PX. Polymeric scaffold for bone tissue engineering. Ann Biomed Eng.
2003;32:477–82.
45. Volkmer E, et al. Hypoxia in static and dynamic 3D culture systems for tissue engineering of
bone. Tissue Eng Part A. 2008;14(8):1331–40.
46. Shin M, Yoshimoto H, Vacanti JP. In vivo bone tissue engineering using mesenchymal stem
cells on a novel electrospun nanofibrous scaffold. Tissue Eng. 2004;10(1–2):33–41.
47. Landers R, Pfister A, Hübner U, John H, Schmelzeisen R, Mülhaupt R. Fabrication of soft
tissue engineering scaffolds by means of rapid prototyping techniques. J Mater Sci. 2002;
37(15):3107–16.
208 G. Della Porta and E. Reverchon
48. Hutmacher DW, Sittinger M, Risbud MV. Scaffold-based tissue engineering: rationale for
computer-aided design and solid free-form fabrication systems. Trends Biotechnol. 2004;
22(7):354–62.
49. Brunner G. Gas extraction: an introduction to fundamentals of supercritical fluids and the
application to separation processes. In: Topics in physical chemistry, vol. 4. Ed. Techniques
Ingénieur; 1994. ISBN 3798509441.
50. Mathieu LM, Montjovent MO, Bourban PE, Pioletti DP, Manson JAE. Bioresorbable
composites prepared by supercritical fluid foaming. J Biomed Mater Res A. 2005;75:89–94.
51. Tsivintzelis I, Marras SI, Zuburtikudis I, Panayiotou C. Porous poly(lactic acid) nanocom-
posite scaffolds prepared by phase inversion using supercritical CO2 as antisolvent. Polymer.
2007;48:6311–3.
52. Reverchon E, Cardea S. Formation of cellulose acetate membranes using a supercritical fluid
assisted process. J Membr Sci. 2004;240:187–95.
53. Reverchon E, Cardea S, Rapuano C. A new supercritical fluid-based process to produce
scaffolds for tissue replacement. J Supercrit Fluids. 2008;45:365–70.
54. Della Porta G, Del Gaudio P, De Cicco F, Aquino RP, Reverchon E. Supercritical drying of
alginate beads for the development of aerogel biomaterials: optimization of process
parameters and exchange solvents. Ind Eng Chem Res. 2013;52:12003–9.
55. Pisanti P, Yeatts AB, Cardea S, Fisher JP, Reverchon E. Tubular perfusion system culture of
human mesenchymal stem cells on poly-L-lactic acid scaffolds produced using a supercritical
carbon dioxide-assisted process. J Biomed Mater Res A. 2012;100(10):2563–72.
56. Li WI, Anderson KW, Deluca PP. Kinetic and thermodynamic modelling of the formation of
polymeric microspheres using solvent extraction/evaporation method. J Contr Rel. 1995;37:
187–98.
57. Li M, Rouand O, Poncelet D. Microencapsulation by solvent evaporation: state of the art for
process engineering approaches. Int J Pharm. 2008;363:26–39.
58. Mao S, Shi Y, Li L, Xu J, Schaper A, Kissel T. Effects of process and formulation parameters
on characteristics and internal morphology of poly(D, L-lactide-co-glycolide) microspheres
formed by the solvent evaporation method. Eur J Pharm Biopharm. 2008;68:214–23.
59. Yang YY, Chia HH, Chung TS. Effect of preparation temperature on the characteristics and
release profiles of PLGA microspheres containing protein fabricated by double-emulsion
solvent extraction/evaporation method. J Control Release. 2000;69:81–96.
60. Della Porta G, Reverchon E. Supercritical fluid based technologies for particulate drug
delivery. In: Ravi Kumar MNV, editor. Handbook of particulate drug delivery. American
Scientific Publishers; 2008. p. 35–59. ISBN 978-1-58883-124-8 (Chapter 3).
61. Della Porta G, Reverchon E. Nanostructured microspheres produced by supercritical fluid
extraction of emulsions. Biotechnol Bioeng J. 2008;100(5):1020–33.
62. Kluge J, Fusaro F, Casas N, Mazzotti M, Muhrer G. Production of PLGA micro- and
nanocomposites by supercritical fluid extraction of emulsions: I. Encapsulation of lysozime.
J Supercrit Fluids. 2009;50:327–35.
63. Della Porta G, Falco N, Reverchon E. Antiflammatory drugs release from injectable
microspheres produced by supercritical fluid emulsion extraction. J Pharm Sci. 2010;99(3):
1484–99.
64. Della Porta G, Castaldo F, Scognamiglio M, Paciello L, Parascandola P, Reverchon E.
Bacteria microencapsulation in PLGA microdevices by supercritical emulsion extraction.
J Supercrit Fluids. 2012;63:1–7.
65. Della Porta G, Campardelli R, Falco N, Reverchon E. PLGA microdevices for retinoids
sustained release produced by supercritical emulsion extraction: continuous versus batch
operation layouts. J Pharm Sci. 2011;100(10):4357–67.
66. Della Porta G, Reverchon E. Continuous supercritical emulsions extraction: a new technology
for biopolymer microparticles production. Biotechnol Bioeng J. 2011;108(3):676–86.
67. Falco N, Reverchon E, Della Porta G. Continuous supercritical emulsions extraction: packed
tower characterization and application to poly(lactic-co-glycolic acid) + insulin microspheres
production. Ind Eng Chem Res. 2012;51:8616–23.
8 Modular Tissue Engineering … 209
86. Roth EA, Xu T, Das M, Gregory C, Hickman JJ, Boland T. Inkjet printing for
high-throughput cell patterning. Biomaterials. 2004;25:3707–15.
87. Adam EJ, Alexandra LR, Ramille NS. Advancing the field of 3D biomaterial printing.
Biomed Mater. 2016;11:014102. https://doi.org/10.1088/1748-6041/11/1/014102.
88. Della Porta G, Nguyen BN, Campardelli R, Reverchon E, Fisher JP. Synergistic effect of
sustained release of growth factors and dynamic culture on osteoblastic differentiation of
mesenchymal stem cells. J Biomed Mater Res Part A. 2014. https://doi.org/10.1002/jbm.a.
35354.
89. Chan BP, Hui TY, Wong MY, Yip KHK, Chan GCF. Mesenchymal stem cell-encapsulated
collagen microspheres for bone tissue engineering. Tissue Eng. 2010;16(2):225–35.
90. Kuroda Y, Akiyama H, Kawanabe K, Tabata Y, Nakamura T. Treatment of experimental
osteonecrosis of the hip in adult rabbits with a single local injection of recombinant human
FGF-2 microspheres. J Bone Miner Metab. 2010;28:608–16.
91. Ben-Ari A, Rivkin R, Frishman M, Gaberman E, Levdansky L, Gorodetsky R. Isolation and
implantation of bone marrow-derived mesenchymal stem cells with fibrin micro beads to
repair a critical-size bone defect in mice. Tissue Eng A. 2009;15(9):2537–46.
92. Jayasuriya AC, Bhat A. Fabrication and characterization of novel hybrid organic/inorganic
microparticles to apply in bone regeneration. J Biomed Mater Res A. 2010;93(4):1280–8.
93. Liu X, Jin X, Ma PX. Nanofibrous hollow microspheres self-assembled from star-shaped
polymers as injectable cell carriers for knee repair. Nat Mater. 2011;10:398–406.
94. Kang SW, La WG, Kim BS. Open macroporous poly(lactic-co-glycolic Acid) microspheres
as an injectable scaffold for cartilage tissue engineering. J Biomater Sci Polym Ed.
2009;20:399–409.
95. Solorio LD, Fu AS, Hernandez-Irizarry R, Alsberg E. Chondrogenic differentiation of human
mesenchymal stem cell aggregates via controlled release of TGF-beta1 from incorporated
polymer microspheres. J Biomed Mater Res A. 2010;92:1139–44.
96. Solorio LD, Dhami CD, Dang PN, Vieregge EL, Alsberg E. Spatiotemporal regulation of
chondrogenic differentiation with controlled delivery of transforming growth factor-b1 from
gelatin microspheres in mesenchymal stem cell aggregates. Stem Cells Translational Med.
2012;1(8):632–9.
97. Han Y, Wei Y, Wang S, Song Y. Cartilage regeneration using adipose-derived stem cells and
the controlled released hybrid microspheres. Joint Bone Spine. 2010;77:27–31.
98. Park JS, Na K, Woo DG, Yang HN, Park KH. Determination of dual delivery for stem cell
differentiation using dexamethasone and TGF-beta 3 in/on polymeric microspheres.
Biomaterials. 2009;30:4796–805.
99. Park JS, Yang HN, Woo DG, Jeon SY, Park KH. The promotion of chondrogenesis,
osteogenesis, and adipogenesis of human mesenchymal stem cells by multiple growth factors
incorporated into nanosphere-coated microspheres. Biomaterials. 2011;32:28–38.
Index
A C
Adhesion, 4–8, 12, 14–17, 19, 35, 44, 59, 61, Cartilage, 4, 6, 7, 13, 20, 46, 47, 50, 119, 120,
64, 71–74, 84, 85, 91, 93–95, 97, 129–133, 153, 156, 161, 165, 172, 173,
101–103, 115, 116, 119, 126, 133, 155, 179–182, 194, 204
157, 166, 168, 183, 194 Cell carrier, 116, 124
Adhesive glycoproteins, 7 Cell-cell contacts, 15, 60, 61, 63
Adipose, 41, 42, 46, 116, 130, 132, 133, 155 Cell culture, 49, 59, 60, 62, 63, 72, 74, 75, 131,
Antimicrobial properties, 30 133, 156, 195, 205
Apoptosis, 6, 59, 60, 63, 64, 72, 74, 89, 97, 98, Cell migration, 9, 11, 15, 16, 67, 71, 84, 92, 95,
101, 103, 168–170 132, 134, 170
Architecture and prestress, 163 Cell niche and extracellular matrix, 62
Cell polarity, 16, 17, 75
B Cell proliferation, 8, 9, 12, 15, 35, 49, 51, 73,
Biochemical composition, 5, 29, 36, 52 99, 115, 125, 127, 128, 154, 157, 170,
Biochemical content, 29 173
Biocompatibility, 8, 35, 52, 126, 127, 131, 135, Cells, 3–5, 7–19, 27–32, 35–51, 59–64, 66–68,
153–155, 158, 183, 194, 195 70–76, 83–85, 87–92, 94, 95, 97–99,
Bioinductive properties, 30 101–103, 113–135, 152, 153, 155–157,
Biomaterials, 15, 20, 27, 33, 38, 41, 43, 49–51, 162–174, 179–183, 191, 192, 194, 198,
114, 120, 126, 153, 157, 158, 201 200, 202–205
Biomaterials for scaffold fabrication, 194 Central nervous system, 45, 67, 176, 178
Bone, 4, 7, 9, 10, 12, 13, 17, 19, 30, 31, 41, 49, Chains and trimers, 65
50, 70, 73, 99, 119, 121, 122, 125, 126, Chemoattraction, 30, 31
129, 132, 133, 135, 153–156, 158, 161, Clustering calcium channels in motor nerve
162, 164, 165, 168–173, 177, 179, 181, terminals, 69
182, 194, 200, 202, 203 Controlled drug release, 123, 124, 131
Bone and osteoinduction, 170 Cornea, 153
Bone marrow-derived hematopoietic stem Co-signaling in mammary epithelial cells, 68
cells, 73 Co-signaling in vascular endothelial cells, 69
Brain, 17, 45, 67, 76, 122, 125, 132, 176 Crosslinkers, 131
Muscle, 14, 32, 35, 36, 39, 40, 44, 45, 49, 51, Signaling, 3, 4, 6–19, 59, 60, 63, 65–69, 72,
61, 66, 67, 70, 115–117, 126, 130, 132, 74, 75, 84, 86, 93–95, 102, 162,
162, 164, 168, 169, 173–175, 178, 180 165–169, 174, 180
Musculoskeletal tissues healing, 161 Silk fibroin, 115, 117, 152–157
Skeletal muscle, 32, 44, 45, 49, 168, 169, 178,
N 179
Nanomaterials, 153 Skin, 4, 7, 14, 17, 43, 66, 67, 89, 93, 116, 117,
Nanometric architecture, 162 119, 129, 132, 133, 153, 157, 179
Nanoscale biotechonology, 179 Small intestinal submucosa, 27, 29, 33
Nanoscale engineering, 162 Spinal cord, 45, 119, 120, 129, 131, 132
Nanotechnologies, 158 Stem cell, 15, 40, 41, 46, 48, 49, 59, 60, 119,
Natural biomaterials, 4, 19 121, 127, 130, 170, 183
Neurobiology, 74 Stem cell technologies, 60
Niche-specific extracellular matrix, 59, 63, 64 Survival, 9, 10, 14–16, 59, 61, 62, 72, 73, 76,
Nucleus pulposus, 48, 116 83, 84, 89, 92, 162, 173, 183
O T
Other extracellular matrix molecules, 64, 69, Tendon, 32, 47, 48, 118–120, 132, 153,
96 155–157, 166, 173–179, 182
Tensegrity, 163, 165–167
P Tensegrity and mechanochemical transduction,
Pancreatic islets, 63 165
Pericardium, 40 3D drug releasing scaffolds, 120, 122, 124
Possible antagonistic functions of related Three pillars on cell niche, 63
laminins, 70 3D scaffolds, 113, 123, 125–127, 183, 192,
Preclinical studies, 30, 132 195–197, 202
Proliferation, 4–6, 8–10, 14, 15, 31, 35, 42–44, Thrombospondins and immunity, 103
46, 59, 61, 63, 64, 71, 72, 74–76, 84, 91, Thrombospondins and inflammation, 96
92, 96, 102, 117, 125, 130, 133, 134, Thrombospondins, 89, 90
155–157, 168–170, 180–182 Tissue engineering, 3–5, 13, 15–18, 28,
Prominent role of the proteoglycans, 6 113–116, 119, 120, 126–130, 132, 133,
Protease activity, 11 135, 152, 153, 155–158, 162, 166, 170,
179, 181–183, 191, 192, 194–196, 200
R Tissue-specific hydrogels, 38
Regenerative medicine, 16, 32, 59, 61–63, 71, Tissue specificity, 28, 38, 42, 45, 52
72, 75, 119, 152, 170, 171 Types of ECM-derived materials, 32
Role of proteolic enzyme in ECM, 11
U
S Urinary bladder matrix, 27, 29, 33
Safety, 28, 40, 47, 75
Scaffolds, 3–5, 13, 19, 20, 28, 31, 32, 36, 42, V
43, 48, 49, 51, 75, 84, 113–123, Versican, 6, 83, 85, 87–90, 94–96, 101, 102
125–130, 132–135, 152–157, 161–163, Versican and immunity, 101
165, 179–183, 191, 193–196, 198, Versican and inflammation, 94
201–203 Vocal fold, 50
Self-assembly and interaction with other
laminin isoforms, 69