Hepatic Stem Cells Methods and Protocols - Naoki Tanimizu - 2019 PDF
Hepatic Stem Cells Methods and Protocols - Naoki Tanimizu - 2019 PDF
Hepatic
Stem Cells
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Naoki Tanimizu
Sapporo Medical University, Sapporo, Hokkaido, Japan
Editor
Naoki Tanimizu
Sapporo Medical University
Sapporo, Hokkaido, Japan
Cover Illustration: Biliary epithelial cells (BECs) labeled with tdTomato proliferate to expand the CK19+ biliary epithelial
tissue structure upon injury, indicating that pre-existing BECs contribute to the “ductular reaction”. (The image related
to Chapter 7 has been provided by Dr. Kenji Kamimoto).
This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC, part of
Springer Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
The liver contains two types of epithelial cells, namely, hepatocytes and cholangiocytes
(biliary epithelial cells). Therefore, liver stem/progenitor cells (LPCs) are defined as bipo-
tential cells that can differentiate into hepatocytes and cholangiocytes. Hepatoblasts, fetal
LPCs, split into hepatocytes and cholangiocytes in mid-gestation. Bipotential LPCs also
have been isolated from neonatal liver. On the other hand, it is still controversial whether
bipotential LPCs contribute to maintenance of cellular homeostasis in healthy adult liver.
Recent works demonstrated that both hepatocytes and cholangiocytes are heterogeneous
cell populations and may contain committed progenitors. This book first contains methods
for isolation and expansion of bipotential LPCs from fetal and neonatal liver, for adult
clonogenic cholangiocytes and for hepatocyte progenitors.
The liver has the strong regenerative capability: it has been generally considered that
self-duplication of hepatocytes or cholangiocytes compensates the lost tissue after acute
injuries, whereas LPCs are activated and supply new hepatocytes after chronic injuries.
However, recent works demonstrated that dedifferentiation and lineage conversion of
liver epithelial cells also contribute to liver regeneration. These results highlight that liver
regeneration can be achieved by multiple modes of cellular responses. In the second part,
this book contains experimental methods for identifying and characterizing progenitor cells
involved in liver regeneration in vivo as well as those for investigating molecular mechanisms
regulating progenitor cell-driven liver regeneration.
It is crucial to expand functional hepatocytes and cholangiocytes ex vivo for implement-
ing regenerative medicine such as cell therapy and for establishing drug screening systems.
Recently, novel culture methods have been established to supply hepatocytes and/or
cholangiocytes from pluripotent stem cells as well as somatic terminally differentiated
cells. In addition, it was demonstrated that in vivo conversion of fibrogenic stellate cells
into hepatocytes could be useful to ameliorate liver fibrosis. In the third part, this book
contains culture methods for generating hepatocytes and cholangiocytes from multiple
cellular sources and a technique converting fibroblasts into hepatocytes in vivo.
Liver epithelial cells form three-dimensional (3D) tissue structures, which are indispens-
able for the liver to perform their physiological functions. Facing various types of injuries,
3D hepatic tissue structures are dynamically rearranged to avoid fatal liver damages. Thus,
this book also contains experimental methods for the reconstitution of 3D tissue structures
ex vivo as well as for the investigation of the dynamic structural changes induced after
injuries and during regeneration.
Fibrosis is the major pathology of the liver. Another issue is liver cancers. It is important
to establish a novel strategy to resolve liver fibrosis and to understand etiologies of hepato-
cellular (HCC) and cholangiocellular carcinomas (CC). Therefore, the last part of this book
includes methods resolving hepatic fibrosis by bone marrow transplantation as well as two
novel mouse models of HCCs and CCs.
v
vi Preface
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
Contributors
ix
x Contributors
JUNICHI KINO Department of Tissue Development and Regeneration, Research Institute for
Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan;
Tokushima Research Institute, Otsuka Pharmaceutical Co. Ltd., Tokushima, Japan
SUNGJIN KO Department of Developmental Biology, McGowan Institute for Regenerative
Medicine, Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA, USA
KAZUHIKO KOIKE Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
NOBUHIKO KOJIMA Department of Life and Environmental System Science, Graduate
School of Nanobioscience, Yokohama City University, Yokohama, Japan
YUTA KOUI Laboratory of Cell Growth and Differentiation, Institute of Molecular and
Cellular Biosciences, The University of Tokyo, Tokyo, Japan
BIN LI Oregon Stem Cell Center, Oregon Health and Science University, Portland, OR,
USA
HIROTAKA MIHARA Department of Life and Environmental System Science, Graduate
School of Nanobioscience, Yokohama City University, Yokohama, Japan
TOSHIHIRO MITAKA Department of Tissue Development and Regeneration, Research
Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo,
Japan
HAYATO NAKAGAWA Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
YUJI NISHIKAWA Division of Tumor Pathology, Department of Pathology, Asahikawa
Medical University, Asahikawa, Hokkaido, Japan
TAKAHIRO OCHIYA Division of Molecular and Cellular Medicine, National Cancer Center
Research Institute, Tokyo, Japan
MICHAEL OTT Department of Gastroenterology, Hepatology and Endocrinology, Hannover
Medical School, Hannover, Germany
TERESA RUBIO-TOMÁS Institut d’Investigacions Biomèdiques August Pi i Sunyer
(IDIBAPS), Barcelona, Spain
YASUYUKI SAKAI Institute of Industrial Science, The University of Tokyo, Tokyo, Japan
ISAO SAKAIDA Department of Gastroenterology and Hepatology, Yamaguchi University
Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
PAU SANCHO-BRU Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS),
Barcelona, Spain; Centro de Investigacion Biomédica en Red de Enfermedades Hepáticas y
Digestivas (CIBERehd), Barcelona, Spain
AMAR DEEP SHARMA Department of Gastroenterology, Hepatology and Endocrinology,
Hannover Medical School, Hannover, Germany
XIZHONG SHEN Department of Gastroenterology, Zhongshan Hospital, Fudan University,
Shanghai, China
DONGHUN SHIN Department of Developmental Biology, McGowan Institute for
Regenerative Medicine, Pittsburgh Liver Research Center, University of Pittsburgh,
Pittsburgh, PA, USA
GUANGQI SONG Department of Gastroenterology, Zhongshan Hospital, Fudan University,
Shanghai, China
ELEANOR STOUT Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, PA,
USA
RYO SUDO Department of System Design Engineering, Keio University, Yokohama, Japan
LULU SUN State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular
Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of
Sciences, University of Chinese Academy of Sciences, Shanghai, China
NOBUMI SUZUKI Department of Gastroenterology, The University of Tokyo, Tokyo, Japan
Contributors xi
Abstract
Mouse fetal liver includes abundant hepatic stem/progenitor cells (HSPCs). Easy expansion with passage of
HSPCs is necessary to obtain steady data. However, it is often difficult to enrich only HSPCs, and HSPCs
can die when usual trypsin is used for replating. Here, we introduce serum-free long-term culture with
passage of HSPCs using fetal mouse liver without a cell sorter.
Key words Hepatic progenitor cell, Spheroid, Serum-free culture, Colony, Mouse
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_1, © Springer Science+Business Media, LLC, part of Springer Nature 2019
3
4 Atsunori Tsuchiya and Shuji Terai
DMEM/F12 (1:1)
+B27+ITS+
20 ng/ml EGF+
Subpassage with maintaining With trypsin diluted 20 ng/ml bFGF+
colony formation by KSR and CaC12 10 ng/ml HGF
2 Materials
3 Methods
3.1 Spheroid Culture 1. Before performing 2D culture, create spheroids using an ultra-
low attachment plate.
2. Kill a pregnant C57BL/6 mouse at day 13.5 of gestation using
isoflurane, and perform cesarean delivery.
3. Remove and dissect fetal livers using tweezers in cold PBS.
4. Dissociate the dissected fetal livers using a 1 mL pipette tip, and
collect the fragments with centrifugation at 500 rpm for 3 min
(see Note 4).
5. Count and plate the collected cells (see Note 5) at a density of
5 105 cells/mL in the medium with growth factors men-
tioned in Subheading 2.2 on a 6-well ultralow attachment plate
(2 mL/well).
6. Add 2 mL of medium and growth factors at days 2 and
4. Hepatic spheroids will form and increase in size for 6 days
(Fig. 2) (see Note 6).
Fig. 2 Spheroids formed for 6 days by spheroid (floating) culture. The size of
spheroids was approximately 50–250 μm, comprising approximately 20–150
cells
Fig. 3 Images of the 2D (plating) culture and replating. Colonies are formed from each spheroid (a). Collected
colonies by diluted trypsin (b) are replated onto newly prepared collagen-coated plates (c). During passage,
cell-cell contact was maintained
4 Notes
References
1. Suzuki A, Zheng Y, Kondo R et al (2000) 6. Tanimizu N, Nishikawa M, Saito H et al (2003)
Flow-cytometric separation and enrichment of Isolation of hepatoblasts based on the expres-
hepatic progenitor cells in the developing sion of Dlk/Pref-1. J Cell Sci 116:1775–1786
mouse liver. Hepatology 32:1230–1239 7. Tanimizu N, Tsujimura T, Takahide K et al
2. Suzuki A, Zheng YW, Kaneko S et al (2002) (2004) Expression of Dlk/Pref-1 defines a sub-
Clonal identification and characterization of population in the oval cell compartment of rat
self-renewing pluripotent stem cells in the liver. Gene Expr Patterns 5:209–218
developing liver. J Cell Biol 156:173–184 8. Tsuchiya A, Heike T, Fujino H et al (2005)
3. Schmelzer E, Zhang L, Bruce A et al (2007) Long-term extensive expansion of mouse
Human hepatic stem cells from fetal and post- hepatic stem/progenitor cells in a novel
natal donors. J Exp Med 204:1973–1987 serum-free culture system. Gastroenterology
4. Kamiya A, Kakinuma S, Yamazaki Y et al 128:2089–2104
(2009) Enrichment and clonal culture of pro- 9. Tsuchiya A, Heike T, Baba S et al (2007) Long-
genitor cells during mouse postnatal liver term culture of postnatal mouse hepatic stem/
development in mice. Gastroenterology progenitor cells and their relative developmen-
137:1114–1126 1126 e1-14 tal hierarchy. Stem Cells 25:895–902
5. Kakinuma S, Ohta H, Kamiya A et al (2009) 10. Lu WY, Bird TG, Boulter L et al (2015)
Analyses of cell surface molecules on hepatic Hepatic progenitor cells of biliary origin with
stem/progenitor cells in mouse fetal liver. J liver repopulation capacity. Nat Cell Biol
Hepatol 51:127–138 17:971–983
Chapter 2
Abstract
Liver stem/progenitor cells (LPCs) are defined as bipotential cells differentiating into both hepatocytes and
cholangiocytes. The Notch, TGFβ, and Hippo pathways have been implicated in lineage determination of
LPCs during development and regeneration. However, the molecular mechanisms governing the lineage
specification have not been fully elucidated, yet. Epithelial adhesion molecule (EpCAM) is a marker of
cholangiocytes and of LPCs. We found that EpCAM+ cells isolated from neonatal liver contain LPCs that
clonally proliferate and are bipotential in vitro and in vivo. Furthermore, EpCAM+ progenies keep the
capacity of bidirectional differentiation even after long-term culture. These cells are useful to investigate the
molecular mechanisms regulating lineage commitment and epithelial differentiation of LPCs.
Key words Tissue stem cell, Liver, Neonate, Bipotential, Hepatocyte, Cholangiocyte, Liver stem/
progenitor cell, Fluorescence-activated cell sorting, Colony assay, Clonal culture
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_2, © Springer Science+Business Media, LLC, part of Springer Nature 2019
9
10 Naoki Tanimizu
2 Materials
2.3 Cell Culture 1. Growth factor-reduced Matrigel® (MG) (BD Biosciences, Bed-
ford, MA): Thaw the bottle of MG on ice, and make aliquots in
1.5 mL tubes. Freeze those tubes in liquid nitrogen, and store
Isolation of Neonatal Bipotential Liver Progenitors 11
3 Methods
3.1 Cell Isolation 1. Anesthetize a mouse by isoflurane. Open the abdomen, and
then cut the diagram and rib to expose the heart.
3.1.1 Collagenase
Perfusion 2. Attach a needle to a 10 mL syringe filled with pre-perfusion
medium that is pre-warmed at 37 C. Insert the needle into the
left ventricle, and inject pre-perfusion medium. Cut the inferior
vena cava after confirming the liver tissue becomes enlarged
due to pressure (Fig. 1). Change the syringe to one filled with
perfusion medium. Inject the perfusion solution slowly.
3. Isolate the liver and place it on a 6 cm plastic dish. Add 5 mL of
HBSS. Hold the liver with a forceps, and scrape off hepatocytes
from undigested liver tissue using another forceps. (Use undi-
gested tissue for further enzymatic digestion. See the next
paragraph.) Pass the cell suspension through a 70 μm cell
strainer.
4. Centrifuge the cell suspension at 50 g for 1 min. Collect the
supernatant and centrifuge it at 350 g for 4 min (see Note 1).
Resuspend the pellet in 1 mL of basal medium.
3.1.2 Enzymatic 1. Mince the undigested tissue after collagenase perfusion. Sus-
Digestion of the Remaining pend tissue pieces in 3 mL of liver perfusion medium, and
Tissue After Collagenase collect them in a 15 mL tube. Centrifuge at 150 g for 3 min.
Perfusion 2. Resuspend the pellet in 1 mL of Liberase TM solution (see
Note 3). Incubate the cell suspension at 37 C for 15 min.
Dissolve the tissue by pipetting (see Note 4). Add 1–2 mL of
3.1.3 Hemolysis 1. Combine the cell suspensions from Subheading 3.1.1, step 4,
and Subheading 3.1.2, step 3, into a new 15 mL tube. Centri-
fuge at 350 g for 4 min.
2. Resuspend pellet in 1 mL of hemolysis buffer. Incubate at 4 C
for 5 min. Then, add 1–2 mL of basal medium, and pass
through a 70 μm cell strainer. Centrifuge at 350 g for 4 min.
3. Resuspend the pellet in 200 μL of basal medium, and count the
number of cells.
3.2 Cell Sorting 1. Centrifuge the cell suspension at 350 g for 4 min. Resuspend
cells in 100 μL of basal medium.
2. Add 1 μL of anti-CD16/CD32 antibody, and incubate at 4 C
for 20 min to avoid non-specific binding of antibodies by
masking Fcγ receptors. Add ice-cold wash buffer and centrifuge
at 350 g for 4 min.
3. Resuspend cells in 100 μL of basal medium, and then add 1 μL
each of FITC-conjugated anti-mouse EpCAM,
APC-conjugated anti-CD31, APC-conjugated anti-CD45,
and APC-Cy7-conjugated anti-TER119 (see Note 5).
4. For an isotype control, add FITC-conjugated rat IgG,
APC-conjugated rat IgG, and APC-Cy7-conjugated rat IgG.
5. Incubate cells with antibodies at 4 C for 20 min.
6. Add 2 mL of ice-cold wash buffer and centrifuge at 350 g for
4 min. Resuspend cells in 300 μL of basal medium containing
1 μg/mL PI. Pass through 40 μm cell strainer before analysis
on FACS Aria II (BD Biosciences).
7. Select CD31 CD45 TER119 cells in PI singlet cells by
gating on FACS Aria II. Identify and collect EpCAM + cells
in CD31 CD45 fraction into a 5 mL tube (Fig. 2).
8. Transfer the collected cells into a 15 mL tube. Centrifuge at
350 g for 8 min. Resuspend cells and add 7 mL of basal
medium and centrifuge at 350 g for 8 min. Resuspend cells in
1 mL of basal medium.
14 Naoki Tanimizu
Fig. 2 Isolation of EpCAM+ cells by FACS. After gating live singlet CD31 CD45 cells, EpCAM+ cells can be
identified and isolated
3.3.2 Hepatocyte 1. Plate 5000 EpCAM+ cells in a well of 24-well plate coated with
Differentiation gelatin.
2. Replace basal medium to the differentiation medium contain-
ing OSM and incubate for 2 days. Replace the medium to one
containing 5% MG. Keep the culture for additional 3 days.
3. To examine expression of hepatocyte marker genes, dissolve
cells in a lysis solution and extract total RNA. Synthesize cDNA
and perform PCR with gene-specific primers.
4. To examine expression of hepatocyte marker proteins, treat
cells with Subheading 3.3.1, step 2. Incubate cells with rabbit
anti-hepatocyte nuclear factor 4 α (HNF4α) and goat anti-ALB
or with rabbit anti-CCAAT enhancer binding protein α
(C/EBPα) and goat anti-carbamoyl phosphate synthetase I
(CPSI) antibody. Typical images before and after the treatment
of OSM and MG are shown in Fig. 3b.
Isolation of Neonatal Bipotential Liver Progenitors 15
Fig. 3 Proliferation and hepatocyte differentiation of neonatal EpCAM+ cells. A EpCAM+ cell clonally
proliferates and forms a bipotential colony containing ALB+ hepatocytes and CK19+ cholangiocytes. (a) Part
of EpCAM+ cells expresses ALB and HNF4α before inducing hepatocyte differentiation, whereas they become
HNF4α+ALB+ hepatocytes in the presence of OSM and MG. (b) Bars represent 100 μm
4 Notes
Acknowledgments
I thank Dr. Toshihiro Mitaka and Dr. Norihisa Ichinohe for helpful
discussion and Ms. Yumiko Tsukamoto and Ms. Minako Kuwano
for technical assistance. This work is supported by the Ministry of
Education, Culture, Sports, Science and Technology, Japan,
Grants-in-Aid for Scientific Research (C) (25460271,
16 K08716), and Grants-in-Aid for Scientific Research on Innova-
tive Areas “Stem Cell Aging and Disease” (17H05653).
References
1. Miyajima A, Tanaka M, Itoh T (2014) Stem 5. Raven A, Lu WY, Man TY et al (2017) Cholan-
Progenitor cell sin liver development, homeosta- giocytes act as facultative liver stem cells during
sis, regeneration, and reprogramming. Cell Stem impaired hepatocyte regeneration. Nature
Cell 14:261–274 547:350–354
2. Espanol-Suner R, Carpentier R, Van HN et al 6. Kamimoto K, Kaneko K, Kok CY et al (2016)
(2012) Liver progenitor cells yield functional Heterogeneity and stochastic growth regulation
hepatocytes in response to chronic liver injury of biliary epithelial cells dictate dynamic epithe-
in mice. Gastroenterology 143:1564–1575 lial tissue remodeling. Elife 5:e15034
e1567 7. Li B, DorrellC CPS et al (2017) Adult mouse
3. Malato Y, Naqvi S, Schurmann N et al (2011) liver contains two distinct populations of cho-
Fate tracing of mature hepatocytes in mouse langiocytes. Stem Cell Reports 9:478–489
liver homeostasis and regeneration. J Clin Invest 8. Tanimizu N, Kobayashi S, Ichinohe N et al
121:4850–4860 (2014) Downregulation of miR122 by
4. Choi TY, Ninov N, Stainer DY et al (2014) grainyhead-like 2 restricts the hepatocytic differ-
Extensive conversion of hepatic biliary epithelial entiation potential of adult liver progenitor cells.
cells to hepatocytes after near total loss of hepa- Development 141:4448–4456
tocytes in zebrafish. Gastroenterology
146:776–788
Chapter 3
Abstract
Cholangiocytes are proliferative and are one of the sources for liver progenitor cells. Clonogenic cholan-
giocytes are defined as cells capable of clonally proliferating and differentiating cholangiocytes both in vitro
and in vivo. In this protocol, we describe the method for isolation of primary cholangiocytes from mouse.
To study the heterogeneity of cholangiocytes, we used flow cytometry-based cell sorting to isolate different
subsets of cholangiocytes. Organoid-forming efficiencies from sorted single cells are compared within
different cholangiocyte populations to identify clonogenic cholangiocytes.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_3, © Springer Science+Business Media, LLC, part of Springer Nature 2019
19
20 Bin Li et al.
any research has studied ST14 in the liver. In our previous report,
with the cell surface markers MIC1-1C3 [5] and ST14 [3], we
demonstrated that normal mouse cholangiocytes are heteroge-
neous: ST14hiMIC1-1C3+ cells are highly proliferative; they form
single-cell-derived organoid more efficiently than ST14loMIC1-
1C3+ cholangiocytes [3]. Here we will describe the method for
isolating cholangiocytes via liver perfusion. ST14hiMIC1-1C3+
cells in primary liver non-parenchymal cells (NPCs) are directly
sorted into 96-well plate by fluorescence-activated cell sorting
(FACS). In this format of colony-forming assay, clonogenic cho-
langiocytes form single-cell-derived organoids. In addition, orga-
noids cultured for ~3 months are used for the differentiation assay
examining their bidirectional differentiation potential.
2 Materials
3 Methods
3.1 Liver Perfusion 1. Sterilize pump tubing (at ~15 mL/min) with 70% ethanol and
then wash with solution 1. Prewarm all solutions to 40 C (see
Note 3).
2. Attach IV infusion set and fill with solution 1 (see Note 4).
3. Anaesthetize the mouse.
4. Immobilize all four limbs with tape on a moisture-absorbing
surface (diaper). Open the abdomen while avoiding
opening the chest. Push intestines to the side with the sterile
cotton applicator, thereby exposing the vessels. Make sure you
identify the inferior vena cava and portal vein (under the liver)
(Fig. 1a, b).
5. Carefully cannulate the portal vein (see Fig. 1c, d) (see Note 5).
Fill the catheter with solution 1 by syringe, attach the IV
tubing to the liquid-filled catheter (avoid introducing any
air), and start the flow of the pump at 4 mL/min. The liver
will fill up with solution and turn brown within a few seconds.
22 Bin Li et al.
Fig. 1 Liver perfusion. (a) Anesthetize the mouse and open the abdomen. (b) Expose the portal vein with cotton
applicator; white arrow shows the portal vein. (c) Cannulate the portal vein; blood will flow through the
catheter. (d) Connect the tubing to the catheter. (e) White arrow shows cutting vena cava. (f) Perfused liver
Quickly cut the vena cava (see Fig.1e). Tape down the catheter
and tubing securely. Start timer.
6. After about 15 mL of solution 1, run solution 2 for 1 min and
finally solution 3 for 10 min (see Note 6).
7. To gauge progress, poke the liver with a cotton applicator after
7 min or so: under-digested liver remains elastic, whereas a
completely digested liver will retain an indentation (Fig. 1f).
8. When the liver is digested, remove the catheter, excise the
gallbladder, and then carefully remove the liver without injur-
ing the capsule if possible.
9. Transfer the liver to a 60 mm petri dish containing solution
3, and proceed to the next perfusion (Fig. 2a). Disrupt the liver
capsules with forceps, and wash out the hepatocytes by swirling
the dish (Fig. 2b). Hepatocytes are collected and filtered
through a 100 μm filter unit into a sterile 50 mL tube that
contains 10 mL of wash medium (Fig. 2c).
10. For studies of non-parenchymal cells (NPC), tissue retained on
the filter is collected and digested further.
11. Hepatocytes are pelleted by spin at 50 g for 2 min at 4 C
(Fig. 2d). Supernatant is collected for NPC and the pellet
resuspended in 50 mL wash media. Centrifuge at 300 g,
5 min for NPC pellet; this is NPC fraction 1.
12. Transfer tissue fragments in step 10 to a 60 mm dish in 5 mL of
NPC digestion solution. Add a magnetic stirring bar, and stir at
37 C for 10 min. See Fig. 2e.
Isolation of Mouse Liver Progenitor Cells 23
Fig. 2 Liver digestion and FACS sort. (a) Move the liver into a petri dish. (b) Break the liver capsule to
dissociate the cells. (c) Filter the liver cells with cell strainer. (d) Pellet the hepatocytes. (e) Tissue leftover is
under further digestion with NPC solution. (f) Filter the digested NPCs. (g) FACS tube. (h) FACS machine: BD
Influx. (i) 96-well single cell sort
3.2 FACS 1. Count and resuspend the cells with FACS buffer at a density of
1 107 cells/mL. Add 100 μL cells into each control tube
(#1–7). The sample tube (#8) can have 500–1000 μL volume
(see Note 7).
24 Bin Li et al.
Table 1
Antibody labeling for FACS
Tube# 1 2 3 4 5 6 7 8
PI FITC PE PEcy7 APC FM1 FM1 Sort
-APC -PE
MIC1-1C3 (μL) 5 5 50
ST14 (μL) 1 1 10
FACS buffer (μL) 100 100 100 100 100 1000
20 min on ice, wash, pellet cells
Anti-rat PE (μL) 1 1 1 2
Anti-rabbit APC (μL) 1 1 1 2
FACS buffer (μL) 200 200 200 200 400
15 min on ice, wash, pellet cells, blocking tubes #6 and #7 with 5% rat serum for 10 min
FACS buffer (μL) 100 100 100 100 200
FITC CD26 (μL) 1 1 1 2
Pe-cy7 CD45/31/11b (μL) 1 1 1 2
6 6 6 6 6 6 6
Cell number 10 10 10 10 10 10 10 107
Fig. 3 FACS gating strategy. (a and b) Cells were sequentially gated based on cell size (forward scatter [FSC]
versus side scatter [SSC]). (a) Depletion of doublets (FSC vs. trigger pulse width) (b). (c) Dead cells and debris
were excluded by PI positivity. Concurrently a combination of CD45, CD31, and CD11b antibodies was used for
depleting blood, endothelium, and Kupffer cells. (d) CD26 (DPPIV) was used for hepatocyte staining. (e) MIC1-
1C3+ cells can be subdivided into two populations: ST14 high (ST14hiM+) and ST14 low (ST14loM+). (f)
Fluorescence minus 1 without secondary antibody APC was used to draw gate for ST14 APC positivity and
negativity (modified from Bin Li et al., 2017 [3])
11. Last wash and pellet the cells. Resuspend cells with PI at
10 μg/mL in FACS buffer. The FACS gates are drawn as
shown in Fig. 3.
3.3 Organoid Cell 1. Precool the 96-well plate, multichannel pipettes, and tips.
Culture 2. Make the 5% Matrigel in fresh organoid culture media before
sorting, using multichannel pipette to add media into 96-well
plate with 100 μL per well. Keep the plate on ice.
3. Sorted cells are directly injected into cold 96-well plate at a
density of 1 cell per well.
4. Carefully flip the plate to help the cells sink into the media. The
plate will be moved into 37 C CO2 incubator immediately.
5. Add more media into organoid culture at day 5. The organoid
should be seen clearly at day 7. The organoid-forming assay
should be done at day 14.
6. For organoid passage: move the organoid with media in each
well into 1.5 mL Eppendorf tube, add the same volume of
26 Bin Li et al.
TrypLE, pipette with P200, and look through the tube under
microscope. Matrigel dissolves in TrypLE. If organoids are still
stiff, add more TrypLE at room temperature (RT), or put the
tube in 37 C for 3 min.
7. When dissociated small clumps as well as single cell are seen,
add wash medium to wash and spin.
8. The pellet is resuspended in 50 μL Matrigel on ice.
9. Pipette Matrigel droplet on prewarmed 24-well suspension
culture plate at 37 C.
10. After 5 min, add prewarmed organoid medium into plate. The
medium will be changed twice a week. After 14 days culture,
the organoid forming efficiencies are calculated, see Fig. 4.
4 Notes
Acknowledgments
References
Abstract
This protocol showed procedures to isolate and expand small hepatocytes (SHs), as hepatocytic progenitor
cells, from a rat liver. SHs are identified as a subpopulation of mature hepatocytes in a healthy liver. SHs can
proliferate to form colonies in serum-free medium on hyaluronic acid-coated dishes, of which cells show
CD44 positivity (CD44+ SHs). CD44+ SHs can be separated and purified from colonies by using anti-
CD44 antibodies after enzymatic dissociation. CD44+ SHs can proliferate to form colonies on Engelbreth-
Holm-Swarm gel (EHS-gel)-coated dishes in the serum-free medium for a long period and subculture for
several times. Even after the second passage, the cells possess characteristics of hepatocytes such as
expression of albumin and HNF4α. In addition, when the cells are treated with EHS-gel, they can recover
highly differentiated functions of hepatocytes such as glycogen production, CYP activity, and bile secretion.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_4, © Springer Science+Business Media, LLC, part of Springer Nature 2019
29
30 Junichi Kino et al.
Fig. 1 Overview of procedures for the isolation and expansion of rat small hepatocytes (SHs). Liver cells are
isolated from a 6–10 week-old male rat. SHs form colonies on hyaluronic acid-coated dish and express CD44
on their cell surfaces. CD44-positive SHs (CD44+ SHs) is purified by magnetic separation using anti-CD44
antibody. CD44+ SHs can expand on EHS-coated dish and be passaged after trypsinization
Isolation and Expansion of Rat Hepatocytic Progenitor Cells 31
2 Materials
2.1 Isolation of Liver The 6- to 10-week-old F344 rats are used for cell isolation. The
Cells animal is maintained with standard chow and water ad libitum. All
apparatuses used for the collagenase perfusion are sterilized by
autoclave or treatment of 70% ethanol before the experiment starts.
All animal experiments must comply with national and institutional
regulations.
1. Phosphate-buffered saline without Mg2+/Ca2+ (PBS). Store at
room temperature (RT).
2. 500 μg/mL insulin stock solution (1000): Add 100 mg insu-
lin to 100 mL double-distilled water (ddH2O), and subse-
quently add 1.2 mL 1 N HCl (see Note 1). Adjust to 200 mL
with ddH2O and filtrate with a 0.2-μm filter.
3. Pre-perfusion solution: Add approximately 850 mL ddH2O
into a 1000-mL graduated cylinder 100-mL Ca2+, Mg2+-free
HANKS’ balanced salt solution (HBSS), 190 mg ethylene
glycol tetraacetic acid (EGTA), and 1 mL 1000 insulin
stock solution and stir, and then adjust pH to 7.5 with 1 M
NaHCO3. Adjust to 1000 mL with ddH2O and filtrate with a
0.2-μm filter. Store at 4 C until use.
4. Perfusion solution: Add 1 mL 1000 insulin stock solution to
200 mL HBSS. Add collagenase (Wako Pure Chemical Indus-
tries) of 26,000 U to the pre-warmed perfusion solution, and
make 130 U/mL collagenase solution just before a perfusion
(see Note 2).
5. Wash solution: Add 0.5 mL 1000 insulin stock solution,
2 mL penicillin-streptomycin solution (PC-SM, Sigma), and
0.5 mL gentamicin solution (50 mg/mL, Sigma) to 500 mL
HBSS. Store at 4 C until use.
6. Perfusion apparatus.
7. Water bath.
8. Peristatic pump (RP-1000, Tokyo Rika Instruments).
9. Silicon tube: inside diameter, 4.76 mm; outside diameter,
7.94 mm.
10. O2 gas (95% O2 and 5% CO2).
11. Isoflurane and vaporizer.
12. Butterfly needle, 18 gauge.
13. Surgical scissor.
14. Tweezers.
15. Mosquito forceps.
16. Surgical clip, bulldog type.
32 Junichi Kino et al.
3 Methods
3.1 Isolation of Liver 1. Set up the perfusion apparatus in the warmed water bath at
Cells approximately 39 C. Circulate the pre-perfusion solution in
the perfusion apparatus using the peristatic pump and silicone
tube before the experiment, and bubble in it with O2 gas at a
flow rate of 0.5 L/min.
2. Anesthetize a rat with an inhalation of isoflurane.
3. Cut the abdominal wall with surgical scissors and open the
abdominal cavity.
4. Keep opening the abdominal cavity by grasping the edge of cut
abdominal wall using mosquito forceps.
5. Move the intestine to expose the liver, portal vein, bile duct,
and splenic vein.
6. Ligate a common bile duct and splenic vein with a surgical
thread at the portion nearest the portal vein (see Note 4).
7. Stop the circulation of the pre-perfusion solution and insert a
butterfly needle filled with pre-perfusion solution into the
portal vein at 1.5–2.0 cm from the bifurcation of the portal
vein. Stop the tip of the needle at a position close to bifurcation
of the portal vein, and clamp the needle with a surgical clip.
8. Start the perfusion of the pre-perfusion solution at a flow rate
of 30 mL/min (see Note 5).
9. Cut the inferior vena cava and heart just after starting the
perfusion (see Note 6).
10. When the volume of the pre-perfusion solution becomes small,
add the collagenase to the perfusion solution and then pour it
into the perfusion apparatus.
11. Flow the solution at a flow rate of 15 mL/min. Heat around
the abdominal cavity using a heating lamp during the perfusion
(see Note 7).
12. When the volume of the perfusion solution becomes about
half, change the flow rate at 10mL/min (see Note 8).
13. Stop the flow before air bubbles move into the liver when the
solution flows out from the perfusion apparatus.
14. Cut the liver from the abdominal cavity and transfer it to a
sterilized Petri dish (see Note 9).
15. Prepare a 100-mL beaker with 80-mL wash solution.
Isolation and Expansion of Rat Hepatocytic Progenitor Cells 35
16. Peel the hepatic capsule carefully, and then, to drop the
digested cells, gently shake the liver in the beaker. Dropped
cells in the Petri dish are also poured to the beaker.
17. Filter the cell suspension through a 250-μm nylon filter net
into a new 100-ml beaker.
18. Filter the cell suspension through a cell strainer with 70-μm
filter into four 50-ml conical tubes using a 25-ml serological
pipette, and then adjust each tube to an equal volume (approx-
imately 40 mL) with wash solution.
19. Centrifuge the tubes at 50 g for 1 min at 4 C.
20. Collect the supernatants and transfer to four new 50-mL coni-
cal tubes (see Note 10), and wash solution is then added to
each tube up to 45 mL. Repeat Steps 19 and 20 twice.
21. Centrifuge the tubes at 45 g for 5 min at 4 C (see Note 11).
22. Aspirate the supernatant of 40 mL, then add new 40-mL wash
solution to each tube, and dissociate the cell pellet by pipetting.
23. Centrifuge the tubes at 150 g for 5 min at 4 C (see
Note 12).
24. Repeat Steps 22 and 23 twice. Aspirate the supernatant as
possible, and then add new 20-mL wash solution to each
tube, and dissociate the cell pellet by pipetting.
25. Gather suspension into two 50-mL conical tubes, and centri-
fuge the tubes at 150 g for 5 min at 4 C.
26. Aspirate the supernatant as possible, and then add new 20-mL
culture medium to each tube, and dissociate the cell pellet by
pipetting.
27. Gather the suspension into one 50-mL conical tube, and cen-
trifuge the tube at 50 g for 5 min at 4 C.
28. Aspirate the supernatant as possible, and then add new 10-mL
culture medium to the tube, and dissociate the cell pellet by
pipetting. Put this tube on ice.
3.2 Culture of the 1. Dilute the HA stock solution to 1 mg/mL with PBS, fill plastic
Isolated Cells on culture dishes, and incubate overnight at 37 C. On the next
HA-Coated Dishes day, aspirate the HA solution and wash once with PBS. Aspirate
PBS and dry in a clean bench for more than 30 min.
2. Mix 0.5 mL of the suspension of the isolated cells and 1.5 mL
trypan blue solution.
3. Count the number of viable and dead cells, which are smaller
than MHs and larger than nonparenchymal cells, using Neu-
bauer improved hemocytometer (see Note 13).
4. Adjust cell density by diluting the suspension with an appropri-
ate volume of the culture medium.
36 Junichi Kino et al.
Fig. 2 Formation of SH colonies on hyaluronic acid (HA)-coated dish. (a) One day after seeding, SHs adhere on
HA-coated dish. (b) SHs proliferate to form colonies at 9 days after plating, of which consist small-sized cells.
(c) All cells in colonies express CD44 on their cell surfaces. Scale bar: 200 μm
5. Wash dry HA-coated dishes with PBS just before cell seeding.
6. Seed the cells (2.0 104 cells/cm2) on the HA-coated dishes.
7. Place the dishes in 5% CO2/95% air-incubator at 37 C for 3 h.
8. Replace the medium with fresh medium (see Note 14).
9. Place the dishes in 5% CO2/95% air-incubator at 37 C for
9 days, and replace the medium every other day (see Fig. 2 and
Note 15).
3.3 Isolation of 1. Wash the SH colonies on the HA-coated dishes with PBS.
CD44-Positive Cells 2. Treat the colonies with 0.02% EDTA/PBS for 5 min (see Note
from the Cells on 16).
HA-Coated Dishes
3. After aspiration of the solution, treat the colonies with the
separation solution for 5 min at 37 C.
Isolation and Expansion of Rat Hepatocytic Progenitor Cells 37
3.4 Coating Dishes 1. Dispense each aliquot of EHS-gel in a cryogenic tube and store
with EHS-Gel it at 20 C until use.
2. Thaw EHS-gel stock solution on ice.
3. Dilute EHS-gel with HBSS on ice and adjust the concentration
to 0.2 mg protein/mL (see Note 21).
38 Junichi Kino et al.
3.5 Culture of the 1. Mix 1 volume of the suspension of CD44+ cells and 3 volumes
CD44-Positive (CD44+) of trypan blue solution.
Cells on the EHS-Gel- 2. Count the number of viable and dead cells using a Neubauer
Coated Dishes improved hemocytometer.
3. Centrifuge the cell suspension at 150 g for 5 min at 4 C.
4. Aspirate the supernatant and suspend the cell pellet in an
appropriate volume of the culture medium for adjusting cell
density (see Note 22).
5. Wash dry EHS-gel-coated dishes with PBS just before cell
seeding.
6. Seed the cells on the EHS-gel-coated dishes.
7. Place the dishes in 5% CO2/95% air-incubator at 37 C for 3 h.
8. Replace the medium with fresh culture medium.
9. Place the dishes in 5% CO2/95% air-incubator at 37 C, and
replace the medium every other day (see Fig. 3 and Note 23).
3.6 Passage of the 1. Wash the CD44+ cells on the EHS-gel-coated dishes with PBS.
CD44+ Cells on the 2. Incubate the cells with PBS containing 0.25% trypsin and
EHS-Gel-Coated 0.01% EDTA in 5% CO2/95% air-incubator at 37 C for
Dishes 10 min.
3. Dissociate the cells by pipetting gently.
4. Add the culture medium to the cell suspension to stop the
digestion by trypsin.
5. Centrifuge the cell suspension at 150 g for 5 min at 4 C.
Fig. 3 Growth of sorted CD44+ SHs on an EHS-gel-coated dish. Attached CD44+ SHs can grow to form
colonies on EHS-gel-coated dishes. The same colony was followed until 28 days after the passage. All photos
show the same magnification. Scale bars; 200 μm
Isolation and Expansion of Rat Hepatocytic Progenitor Cells 39
4 Notes
23. The longer a culture period is, the harder the connection
between cells become. The culture period should not be longer
than 4 weeks, if the cells will be passaged (see Subheading 3.6).
References
Abstract
Lineage tracing of liver cells is a powerful tool to understand liver embryonic development, healthy liver cell
homeostasis, tissue repair, and regeneration. Lineage tracing of biliary epithelial cells (BECs) in the adult
liver has been used to assess the contribution of the biliary epithelium to liver injury, regeneration, and
disease. These studies have shown the contribution of BECs to the expansion of ductular reaction (DR) and
liver progenitor cells (LPCs) and eventually the generation of new hepatocytes. Few genetic lineage-tracing
mouse models have been proved to trace BECs. This chapter is focused on lineage tracing of BECs in mouse
models of liver injury and regeneration. First, we mention different existing approaches to trace the biliary
epithelium based on proteins specifically expressed by BECs such as sex-determining region Y-box
9 (SOX9), osteopontin (OPN), and cytokeratin-19 (KRT19). Second, we describe mouse models that
can be used to evaluate cell fate during liver injury and regeneration (i.e., partial hepatectomy (PHx), acute
liver injury models, and chronic liver damage models such as 3,5-diethoxycarbonyl-1,4-dihydro-collidin
(DDC) diet, choline-deficient ethionine-supplemented (CDE) diet, or chronic carbon tetrachloride (CCl4)
administration). Third, we suggest possible readouts to assess BECs fate based on immunofluorescence
analysis.
Key words Lineage tracing, Liver regeneration, Biliary epithelial cells, Liver progenitor cells, Ductular
reaction, Chronic liver injury, Animal models
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_5, © Springer Science+Business Media, LLC, part of Springer Nature 2019
45
46 Teresa Rubio-Tomás et al.
2 Materials
2.1 Mouse Strains Mice expressing Cre recombinase under the control of a cell-
specific promoter can be used to induce site-specific recombination
2.1.1 Cre-Expressing
between two loxP sites (Fig. 1). The Cre-loxP system can be used
Lines
to generate conditional knockout mice by excising part of a
protein-coding sequence and, therefore, inhibiting its expression.
Cre recombinase sequence can be fused to the estrogen receptor
sequence (CreER), and, therefore, Cre recombinase activity
becomes tamoxifen-inducible. The use of inducible CreER is par-
ticularly interesting in the context of lineage tracing, since expres-
sion of markers may vary along embryonic development, which
48 Teresa Rubio-Tomás et al.
(-) Tamoxifen
ER
Cre
Cytoplasm
ER
Cre
Hnf1β Rosa26
Cre-ERT2 STOP YFP
promoter promoter Nucleus
(+) Tamoxifen
Tm
ER
ER + Tm Cre
Cre Cytoplasm
ER Tm
Cre YFP
Hnf1β Rosa26
Cre-ERT2 YFP
promoter promoter
Nucleus
Tm
ER ER
Cre Cre Tm
Fig. 1 Tamoxifen-inducible Cre-loxP system. Cre recombinase sequence is fused to hormone-binding domain
of the estrogen receptor (CreER). CreER expression is under the control of a biliary epithelial cell (BEC)-specific
promoter (e.g., hepatocyte nuclear factor 1-beta [Hnf1β] promoter). In the absence of tamoxifen, CreER
remains inactive in the cytoplasm. In the presence of tamoxifen, CreER activates and translocates to the
nucleus, where it recombines the region flanked by LoxP sites (red triangles). After the release of the stop
codon, yellow fluorescent protein (YFP) is permanently expressed in the cell and its progeny
2.3 Isolation of BEC- 1. EGTA solution: 137 mM NaCl, 5.4 mM KCl, 0.64 mM NaH2-
Derived Cells PO4H2O, 0.85 mM Na2HPO4, 1 mM HEPES, 4.17 mM
NaHCO3, 0.5 mM EGTA, and 5 mM glucose, pH ¼ 7.4.
2. Collagenase solution: 0.5 g/L Collagenase A in 137 mM
NaCl, 5.4 mM KCl, 0.64 mM NaH2PO4H2O, 0.85 mM
Na2HPO4, 1 mM HEPES, 4.17 mM NaHCO3, and 3.8 mM
CaCl2·2H2O, pH ¼ 7.4.
3. Collagenase A (Roche Diagnostics GmbH, Germany).
4. Hank’s Balanced Salt Solution (HBSS) (Biological Industries,
United States of America).
5. 70-μm cell strainer (Miltenyi Biotec, Germany).
6. Pronase (Roche Diagnostics GmbH, Germany).
7. DNase I (Roche Diagnostics GmbH, Germany).
8. Live/Dead cell stain kit (Life Technologies, United States of
America).
9. RLT buffer (Qiagen GmbH, Germany).
3 Methods
-1 0 1 or 4
(A) weeks
Tamoxifen 2/3
PHx
-1 0 2
(B) weeks
-1 0 1
(C) weeks
-1 0 4
(D) weeks Standard
DDC diet
diet
Tamoxifen
-1 0 3
Tamoxifen
-1 0 1 2 3 4 5 6 7 8
(F) weeks
Tamoxifen CCl4
0.5 ml/kg
Fig. 2 Experimental models of liver regeneration. (a) Partial hepatectomy (PHx), (b) acute acetaminophen
(APAP) treatment, (c) acute carbon tetrachloride (CCl4) administration, (d) 3,5-diethoxycarbonyl-1,4-dihydro-
collidin (DDC) diet, (e) choline-deficient ethionine-supplemented (CDE) diet, and (f) chronic CCl4 administration
52 Teresa Rubio-Tomás et al.
1. Anesthetize mice.
2. Perform two-thirds PHx in mice as previously described [22].
3. Sacrifice 1 week after PHx.
3.2.2 Acute APAP APAP model (Fig. 2b) induces hepatocellular death and release of
Treatment danger-associated molecular patterns (DAMPs) leading to macro-
phages and NKT cells infiltration. This model mimics drug-
induced injury. Mice treated with APAP show a mild induction of
DR expansion in the liver. In Rodrigo-Torres et al., no
BEC-derived hepatocyte generation was observed when assessed
by dual staining of HNF4α/YFP.
1. Fast mice for 8 h with free access to water.
2. Inject intraperitoneally a single dose of 500 mg/kg body
weight APAP diluted in DPBS at 35–40 C.
3. Inject control mice with equivalent amount of DPBS.
4. Sacrifice mice 2 weeks after the injection.
3.2.3 Acute CCl4 Acute treatment with CCl4 (Fig. 2c) is a widely used model of acute
Treatment liver failure caused by toxic substances. CCl4 induces inflammation,
thrichloromethyl radical formation, and production of reactive
oxygen species (ROS). This leads to lipid peroxidation and hepato-
toxicity [24]. As described in Rodrigo-Torres et al., acute CCl4-
treated mouse livers showed a mild induction of DR expansion.
However, no duct-derived hepatocyte generation was observed
based on dual staining of HNF4α/YFP (Fig. 3a).
1. Inject intraperitoneally a single dose of CCl4 at 0.5 mL/kg,
10–25% diluted in corn oil.
2. Inject control mice with equivalent amount of corn oil.
3. Sacrifice 1 week after the injection.
3.2.4 Chronic Liver DDC diet (Fig. 2d) induces liver damage and the expansion of DR
Damage Models and LPCs [25]. This model has been used to study the activation
with DDC Diet and proliferation of LPCs, but most especially it has been used as a
model of primary sclerosing cholangitis and biliary fibrosis. In
addition to DR, the phenotype observed in this model is character-
ized by fibrosis, inflammatory cells infiltration around periportal
areas, cholestasis and expression of inflammatory cytokines.
In Rodrigo-Torres et al., after 4 weeks of DDC diet an impor-
tant DR was observed, which was derived from HNF1β+ cells.
However, there was no contribution of HNF1β+ cells to newly-
formed hepatocytes. During recovery (2 weeks) with standard
chow, DR was reduced, but once again, HNF1β+ cells did not
contribute to hepatocyte generation.
Lineage Tracing of Biliary Epithelium 53
Fig. 3 Lineage tracing of HNF1β-expressing cells after liver injury. (a) In acute carbon tetrachloride (CCl4) liver
injury model, biliary epithelium cells (BECs) do not give rise to hepatocytes. Hepatocytes express nuclear
hepatocyte nuclear factor 4 alpha (HNF4α) (red) and cells derived from hepatocyte nuclear factor 1-beta
(HNF1β)+ BECs express yellow fluorescent protein (YFP) (green). (b) Image from choline-deficient ethionine-
supplemented (CDE) diet liver injury model. Hepatocytes express HNF4α (green) and cells derived from
HNF1β+ cells express YFP (red). A small number of hepatocytes express the reporter YFP indicating that they
are derived from HNF1β+ cells
3.2.5 Chronic Liver CDE diet (Fig. 2e) is used as a model of expansion of DR cells.
Damage Models Unlike DDC diet, CDE diet does not induce biliary injury, but
with CDE Diet steatosis and a significant hepatocellular damage lead to failure of
hepatocytes function [26]. CDE model also induces inflammatory
cells proliferation and the expression of pro-inflammatory cytokines
such as tumor necrosis factor alpha (TNF-α) and interleukin 1 beta
(IL-1β).
In Rodrigo-Torres et al., after 3 weeks of CDE treatment, there
was proliferation of DR cells derived from HNF1β+ cells. More-
over, there was a small population of YFP+ hepatocytes in the
periportal area (0.22%). This percentage increased to 1.86% after
2 weeks of recovery (Fig. 3b).
1. Feed mice with CDE diet ad libitum for 3 weeks. To perform
an injury-recovery model, feed the animals for 3 weeks with
CDE diet followed by 2 weeks of standard diet (see Note 4).
2. Feed control animals with standard chow.
3. Sacrifice mice when the protocol is completed.
54 Teresa Rubio-Tomás et al.
3.2.6 Chronic CCl4 Long-term treatment with CCl4 (Fig. 2f) induces hepatotoxicity
Administration leading to fibrosis, bile duct proliferation, and, eventually, hepato-
cellular carcinoma. Chronic CCl4 administration is one of the most
widely used models for fibrosis induction. CCl4 treatment does not
compromise hepatocyte replication. This model shows a limited
expansion of DR.
1. Inject intraperitoneally CCl4 at a dose of 0.5 mL/kg, twice a
week for 8 weeks (see Note 4).
2. Inject control mice with equivalent amount of corn oil.
3. Sacrifice 4 days after the last injection.
3.3 Isolation of BEC- Lineage tracing models allow isolating BEC-derived cells by flow
Derived Cells cytometry cell sorting.
1. Anesthetize mice and cannulate inferior cava vein.
2. Perfuse livers with EGTA solution at a flow rate of 5 mL/min
until liver gets pale.
3. Perfuse with collagenase solution containing Collagenase A
0.5 g/L at a flow rate of 5 mL/min for 11 min.
4. Mince digested liver in Petri dish with ice cold 1 HBSS.
5. Redigest with collagenase solution containing 0.5 g/L Col-
lagenase A, 0.5 g/L pronase and 50 mg/L DNAse I.
6. Stir for 30 min at 37 C.
7. Filter liver through 70-μm cell strainer.
8. Centrifuge at 100 g for 2 min and keep supernatant.
9. Centrifuge supernatant at 300 g for 5 min and keep the
pellet.
10. Sort YFP+ cells by flow cytometry. Use cells from wild type
(WT) mice to establish YFP+ gate. Collect YFP+ cells for fur-
ther analysis.
4 Notes
Acknowledgments
References
1. Arias IM et al (2009) The liver: biology and 12. Carpentier R et al (2011) Embryonic ductal
pathobiology, 5th edn. John Wiley & Sons, plate cells give rise to cholangiocytes, periportal
Hoboken hepatocytes, and adult liver progenitor cells.
2. Gouw ASH, Clouston AD, Theise ND (2011) Gastroenterology 141(4):1432–1438.e4
Ductular reactions in human liver: diversity at 13. Español-Suñer R et al (2012) Liver progenitor
the interface. Hepatology 54(5):1853–1863 cells yield functional hepatocytes in response to
3. Okabe M et al (2009) Potential hepatic stem chronic liver injury in mice. Gastroenterology
cells reside in EpCAM+ cells of normal and 143(6):1564–1575.e7
injured mouse liver. Development 136 14. Raven A et al (2017) Cholangiocytes act as
(11):1951–1960 facultative liver stem cells during impaired
4. Kholodenko IV, Yarygin KN (2017) Cellular hepatocyte regeneration. Nature 547
mechanisms of liver regeneration and cell- (7663):350–354
based therapies of liver diseases. Biomed Res 15. Yanger K et al (2014) Adult hepatocytes are
Int 2017:1–17 generated by self-duplication rather than stem
5. Lu W-Y et al (2015) Hepatic progenitor cells of cell differentiation. Cell Stem Cell 15
biliary origin with liver repopulation capacity. (3):340–349
Nat Cell Biol 17(8):971–983 16. Guest RV et al (2014) Cell lineage tracing
6. Pu W et al (2016) Mfsd2a+ hepatocytes repop- reveals a biliary origin of intrahepatic cholan-
ulate the liver during injury and regeneration. giocarcinoma. Cancer Res 74(4):1005–1010
Nat Commun 7:13369 17. Rodrigo-Torres D et al (2014) The biliary epi-
7. Schaub JR, Malato Y, Gormond C, Willenbr- thelium gives rise to liver progenitor cells.
ing H (2014) Evidence against a stem cell ori- Hepatology 60(4):1367–1377
gin of new hepatocytes in a common mouse 18. He L et al (2017) Enhancing the precision of
model of chronic liver injury. Cell Rep 8 genetic lineage tracing using dual recombi-
(4):933–939 nases. Nat Med 23(12):1488–1498
8. Huch M et al (2013) In vitro expansion of single 19. Solar M et al (2009) Pancreatic exocrine duct
Lgr5+ liver stem cells induced by Wnt-driven cells give rise to insulin-producing beta cells
regeneration. Nature 494(7436):247–250 during embryogenesis but not after birth.
9. Sancho-Bru P et al (2012) Liver progenitor cell Dev Cell 17(6):849–860
markers correlate with liver damage and predict 20. Kopp JL et al (2011) Sox9+ ductal cells are
short-term mortality in patients with alcoholic multipotent progenitors throughout develop-
hepatitis. Hepatology 55(6):1931–1941 ment but do not produce new endocrine cells
10. Guy CD et al (2012) Hedgehog pathway acti- in the normal or injured adult pancreas.
vation parallels histologic severity of injury and Development 138:653–665
fibrosis in human nonalcoholic fatty liver dis- 21. Means AL, Xu Y, Zhao A, Ray KC, Gu G
ease. Hepatology 55(6):1711–1721 (2008) A CK19 CreERT knockin mouse line
11. Grompe M (2017) Fah knockout animals as allows for conditional DNA recombination in
models for therapeutic liver repopulation. epithelial cells in multiple endodermal organs.
Springer, Cham, pp 215–230 Genesis 46(6):318–323
Lineage Tracing of Biliary Epithelium 57
22. Mitchell C, Willenbring H (2008) A reproduc- toxicological model. Crit Rev Toxicol 33
ible and well-tolerated method for 2/3 partial (2):105–136
hepatectomy in mice. Nat Protoc 3 25. Thompson MD, Awuah P, Singh S, Monga
(7):1167–1170 SPS (2010) Disparate cellular basis of
23. Kellendonk C, Tronche F, Casanova E, improved liver repair in β-catenin-overexpres-
Anlag K, Opherk C, Schütz G (1999) Induc- sing mice after long-term exposure to
ible site-specific recombination in the brain. J 3,5-diethoxycarbonyl-1,4-dihydrocollidine.
Mol Biol 285(1):175–182 Am J Pathol 177(4):1812–1822
24. Weber LWD, Boll M, Stampfl A (2003) 26. Itoh T, Miyajima A (2014) Liver regeneration
Hepatotoxicity and mechanism of action by stem/progenitor cells. Hepatology 59
of haloalkanes: carbon tetrachloride as a (4):1617–1626
Chapter 6
Abstract
The liver is unmatched in regenerative capacity. However, when exhausted, the liver is predisposed to
various diseases based on injury types and causal agents. Although hepatocytes have been proposed to be
the main source of new hepatocytes during regeneration, the existence of specialized liver stem cells has
been long debated. In mice, oval cells or ductal cells have been postulated as such stem/progenitor pool.
Exhaustive works from different laboratories have shown that in genetically unmodified mice, oval cells, or
by extension ductal cells, only contribute marginally in producing new hepatocytes during liver regenera-
tion, thus indicating that hepatocytes are the main regenerative cell source. In this debated context, we
identified a new population of periportal hepatocytes in the normal mouse liver. These cells we termed
hybrid hepatocytes (HybHP) express low levels of the transcription factor Sox9. Using complementary
lineage tracing tools, we demonstrated that HybHP regenerate the liver after chronic hepatocyte depleting
injuries. Here, we describe the two-step genetic recombination method that allowed us to study HybHP’s
lineage in two established models of liver injury.
Key words Hybrid periportal hepatocytes, Dual recombinase, Lineage tracing, Liver regeneration
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_6, © Springer Science+Business Media, LLC, part of Springer Nature 2019
59
60 Nicola de Prisco et al.
2 Materials
2.1 Mice Strains 1. Sox9-CreErt2: This strain was described in Kopp et al., 2011
[9]; Sox9-CreErt2 mice express CreErt2 recombinase in all Sox9-
expressing cells, which in the liver are both hybrid periportal
hepatocytes and bile duct cells.
Lineage Tracing of Hybrid Periportal Hepatocytes 61
PV Tamoxifen
PV AAV-TBG-FLPo
PV
cHP HybHP Bile duct cHP HybHP Bile duct cHP HybHP Bile duct
Cell Cell Cell
CreErt2 - + + - + + - + + +
nLacZ - - - - + + - - + +
eGFP - - - - - - - + - -
Fig. 1 Specific labeling of HybHP using NZG reporter. Schematic outlining of the two-step genetic recombina-
tion using NZG reporter. Sox9-CreErt2;NZG mice express inducible Cre in ductal cells and HybHP (left panel).
Tamoxifen treatment induces Cre-mediated recombination of PGK neo cassette with the subsequent expres-
sion of nuclear LacZ in ductal cells and HybHP (pink nuclei, middle panel). When Sox9-CreErt2;NZG mice
previously treated with tamoxifen are injected with adeno- or AAV-FLPo, which is expressed only in
hepatocytes, the recombination of nuclear LacZ and the subsequent expression of eGFP, only in HybHP,
occur (right panel)
2.2 Models of Liver 1. Sox9-Cre Ert2;NZG previously treated with tamoxifen and
Injury injected with AAV expressing FLPo were treated with carbon
tetrachloride (CCl4) to induce liver injury and determine the
clonal behavior of HybHP. Mice are treated with a single dose
(acute) of CCl4 or with 6 and 12 injections of CCl4.
2. To induce cholestatic liver injury and see if HybHP can give rise
to true ductal cells, Sox9-Cre Ert2;NZG previously treated with
tamoxifen and with the AAV were fed with diethoxycarbonyl-
1,4-dihydrocollidine (DDC) diet for 6 weeks.
62 Nicola de Prisco et al.
Fig. 2 First recombination step. (a) Sox9-CreErt2;NZG mice (4–6 weeks old) were injected with 100 mg/kg of
tamoxifen. Ductal cells and HybHP were positive for nuclear LacZ (arrowheads and arrows, respectively). (b)
LacZ+ ductal cells were CK19+ (arrowheads) and HybHP LacZ+ were CK19 (arrows). (c) LacZ+ HybHP were
positive for HNF4α (arrows), while LacZ+ ductal cells were HNF4α . Scale bar 20 μm
Fig. 3 Second recombination step. (a) Sox9-CreErt2;NZG mice were first treated with 100 mg/kg of tamoxifen
and 10 days later were given 20 mg/kg tamoxifen. Two weeks later, mice were given 109 adeno- or 5 1011
AAV-FLPo viral particles, respectively. After 10 days liver sections were obtained, stained, and imaged. Only
some HybHP were GFP+ (green arrows), while some HybHP were not recombined by FLPo recombinase,
remaining nuclear LacZ+ and GFP (white arrow). Ductal cells were GFP and nuclear LacZ+ (arrowhead).
After 10 days some GFP+ HybHP still had residual nuclear LacZ signal. (b) All GFP+ HybHP were CK19 (green
64 Nicola de Prisco et al.
2.6 1. Cryostat.
Immunofluorescence, 2. Slide Jar and rack.
Antibodies,
3. Staining tray.
and Staining Reagents
4. Citrate buffer: Dilute 10 citrate buffer pH 6.0 (Scytek
#CPL500) with MilliQ water.
5. Phosphate saline buffer (PBS): 137 mM NaCl, 10 mM
Na2HPO4, 1.8 mM KH2PO4, 2.7 mM KCl, pH 7.3–7.5.
6. Donkey serum.
7. Permeabilization solution: Triton X-100 0.1% diluted in PBS.
8. Blocking solution: Tween 0.1%, donkey serum 2% in PBS.
9. Wash solution: Tween 0.1% in PBS.
10. Mounting reagent: Mowiol or an equivalent reagent.
11. Goat anti-HNF-4α (Santa Cruz, sc-6556), 1:50.
Fig. 3 (continued) arrows). (c) All GFP+ HybHP were HNF4α+ (green arrows). Sox9-CreErt2;NZG were given
tamoxifen followed with AAV-FLPo and treated with 1 or 12 (chronic) CCl4 doses. Liver was excised and
analyzed as above. Single GFP+ HybHP were found after only one CCl4 dose (d), while multicellular GFP+
hepatocytes clones expanding from the portal tract (PT) were observed after 12 CCl4 doses (e). Bracketed and
open scale bars: 20 and 50 μm, respectively
Lineage Tracing of Hybrid Periportal Hepatocytes 65
3 Methods
5. Hold the tail in the warm water for 45 s to 1 min to dilate the
tail vein.
6. Inject in one of the two lateral tail veins.
3.3 Intracardiac Carry out all the procedure under chemical hood.
Perfusion and Staining Day 1 (see Note 7)
1. Prepare a 50 mL syringe with 45 mL of DPBS-heparin, and
load the syringe into the pump.
2. Prepare a 50 mL syringe with 45 mL of zinc formalin and load
in the second syringe pump.
3. Prepare the surgery tray with the formalin absorbent pad.
4. Prepare 50 mL conical tube with 30 mL of zinc formalin.
5. Anesthetize the mouse with isoflurane using anesthesia
chamber.
6. Pin the mouse on the formalin absorbing pad in the surgery
tray and spray the abdominal area with 70% ethanol.
7. Using the circle tip forceps and scissors, cut the skin up from
the stomach along the side around the heart, then carefully cut
the peritoneal membrane.
8. Carefully cut the diaphragm and the rib cage both sides of the
sternum. Clamp the sternum, and roll it to expose the heart.
9. Insert the pump needle into the left ventricle.
10. Once the needle is placed in the left ventricle, cut the right
auricle with scissors, and immediately start pumping DPBS-
heparin solution at 5 mL/min.
11. Pump 15–20 mL DPBS-heparin, and once the liver appears
clear, turn off the pump, and switch the valve to zinc formalin.
12. Perfuse the mouse with 20 mL of zinc formalin (see Note 8),
then stop the pump, and remove the needle.
13. Excise the liver, and put it into a 50 mL conical tube with
30 mL zinc formalin, and gently rock it overnight at 4 C.
14. Repeat the procedure for all mice by washing the tubing with
DPBS for one full flush followed by one full flush of DPBS-
heparin solution.
Day 2
15. Discard the zinc formalin, and rinse twice the liver filling the
50 mL conical with PBS, mix, and discard (see Note 9).
16. Wash with PBS for 15 min at room temperature (RT), rocking
gently and repeat this wash twice.
17. Rinse quickly with MilliQ water.
Lineage Tracing of Hybrid Periportal Hepatocytes 67
5. After 1 h, take the staining jar out of the water bath and let cool
at room temperature for 45 min.
6. Do two quick washes with PBS.
7. Prepare 100 mL of 0.1% Triton X-100 in PBS, and incubate the
slides for 20 min in a staining jar.
8. After 20 min, do five quick washes with PBS (see Note 11).
9. Process one slide a time, drawing a circle around the tissue
section with Dako pen and then incubate with 150–200 μL
of blocking buffer, carefully avoiding the tissue to dry for
30 min in a staining tray.
10. Prepare appropriate antibody solution by diluting antibodies in
blocking buffer (see Note 12).
11. After 30 min, pour the blocking buffer, and carefully add the
antibodies to each slide sample.
12. Incubate at 4 C overnight.
Day 2
13. Wash slides three times for 5 min each with wash solution using
a staining jar.
14. Incubate each slide with the corresponding secondary antibody
diluted in blocking buffer for 2 h at RT.
15. Wash three times for 5 min with wash solution using a
staining jar.
16. Wash two times for 5 min with PBS.
Optional if reduction of liver autofluorescence is desired; oth-
erwise follow directly to step 20:
17. Wash the slides further with two quick washes with MilliQ
water and one quick wash with 70% ethanol.
18. Incubate slides with 0.1% Sudan Black in 70% ethanol for
20–30 min.
19. After this incubation, thoroughly wash slides with PBS 0.02%
Tween, and using a squeeze bottle wash applying directly the
jet to the tissue being careful not to wipe it out.
20. Incubate each slide with DAPI diluted 1:1000 in PBS (1:100 in
PBS-0.02% Tween if Sudan Black is used) for 10 min for
nuclear staining.
21. Wash quickly the slides using a staining jar with MilliQ water. If
Sudan Black has been used, wash the slides three times for
5 min with PBS 0.02% Tween, followed by two quick washes
with MilliQ water.
22. Tap each slide dry on a paper towel, and then tip the slides up in
a drawer to finish drying and be protected from the light.
23. Once dried, mount slides using 10 μL of Mowiol.
Lineage Tracing of Hybrid Periportal Hepatocytes 69
4 Notes
Acknowledgments
References
Abstract
Recent technological advances have revealed the heterogeneity of cells and tissues. Existence of heteroge-
neity in hepatic progenitor cells is becoming apparent by various experimental approaches, and here we
describe a series of techniques to investigate the proliferative heterogeneity of these cells. We have
developed a new technique by combining genetic lineage tracking and three-dimensional imaging methods.
The data obtained can be used in statistical analysis to quantitatively investigate the mechanisms underlying
the heterogeneity of hepatic progenitor cells.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_7, © Springer Science+Business Media, LLC, part of Springer Nature 2019
71
72 Kenji Kamimoto
2 Materials
2.1 Mouse 1. Mouse strain: Prom1-CreERT2. This mouse strain has the
Experiment CreERT2 gene at the Prom1 locus [15] and may be purchased
from the Jackson Laboratory (Stock No: 017743).
2. Mouse strain: R26R-tdTomato. This mouse strain has a loxP-
flanked transcriptional stop fragment inserted upstream of the
red fluorescent protein (tdTomato) gene [16]. The strain is
used in combination with the Cre-expressing mouse strain for
lineage tracing experiments and may be purchased from the
Jackson Laboratory (Stock No: 007909).
3. Mouse strain: Prom1-CreERT2;R26R-tdTomato. Cross
Prom1-CreERT2 with R26R-tdTomato mouse strains to
obtain a Prom1-CreERT2;R26R-tdTomato mouse (see Note
1).
4. Tamoxifen solution: Dissolve 10 mg of tamoxifen in 10 mL
corn oil to obtain a 1 mg/mL solution. It takes several hours to
completely dissolve tamoxifen powder in corn oil. Place the
mixture on an agitator at 37 C, and incubate it until the
powder is dissolved.
5. Gavage needle.
6. Thioacetamide (TAA) solution: Dissolve TAA in water at
300 mg/L. Prepare this solution just before administration
(see Note 2).
7. 26G syringe needle.
8. 10 mL syringe.
9. CO2 cylinder.
3 Methods
3.2 Tissue Fixation The mouse liver should be harvested several weeks after TAA
administration. We recommend analyzing the liver after
1–8 weeks. The experimental period should be selected according
to the experimental design. Detailed information can be inferred
from analyzing a range of injury periods rather than a fixed time
point.
1. Place mouse in chamber for CO2 euthanasia.
2. Flow high concentrations of CO2 until vital signs are lost.
3. Immediately following euthanasia, set the mouse for surgery to
start PFA perfusion (see Note 10).
4. Remove the skin and muscle layers of the abdomen using
surgical scissors to expose the liver.
5. Cut the vena cava to let the blood flow out.
6. Insert the syringe needle into the portal vein, and inject 10 mL
of PBS gently until blood flows out from the liver. Next, inject
10 mL of 2% PFA and then 10 mL of 4% PFA. Make sure the
liver color changes into light brown and the liver becomes hard
(see Note 11).
7. Remove the liver from the mouse body.
Analysis for the Heterogeneity of Liver Progenitor Cells 75
3.3 Tissue 1. Cut the liver into ~1 cm thick slices with a razor blade.
Processing 2. Place the slices in a 5 mL tube filled with 4% PFA, and incubate
at 4 C overnight. Remove the PFA and wash the liver twice
with PBS. Incubate the liver slices in 20% sucrose at 4 C
overnight.
3. Embed the fixed liver slices in the OCT compound. Freeze the
sample with liquid nitrogen. The frozen sample can be stored at
80 C.
4. Place the frozen liver into the sample holder of the cryostat.
5. Slowly warm the frozen liver to melt the surface of the sample
by touching with the hand. Immediately after thawing the
surface of the sample, cut the frozen liver into 200–300 μm
thick sections using the cryostat (see Note 12).
6. Using forceps, place the samples into PBS in a 50 mL tube. At
least 30 liver slices should be obtained from a frozen liver block
with 1 cm in thickness (see Note 13).
7. Wash the sliced samples with PBS three times.
8. Wrap the tube in aluminum foil sheet, or incubate in the dark
to avoid photobleaching of fluorescent dye and the fluorescent
protein.
9. Place the liver samples into a 5 mL tube filled with 2 mL of
permeabilization buffer. Incubate the samples for 30 min on a
rocking device (see Note 14).
10. Aspirate the permeabilization buffer from the tube, and add
2 mL of nucleus staining solution. Incubate the tube overnight
on a rocking device (see Note 15).
11. Wash samples with PBS three times.
12. Remove the PBS completely. Mix samples with 2 mL of SeeDB
in a new 5 mL tube. Incubate the samples overnight on a
rocking device (see Notes 16 and 17).
3.5.1 Quantification 1. Load images using the software. All metadata will be imported
by Volocity automatically along with image data.
2. In Measurement mode, detect nuclei using the Find Object
module. The object identified by this step includes all nuclei in
the liver sample (Fig. 3a).
3. Detect labeled cells by applying the Find Object module to the
tdTomato signal. tdTomato is uniformly localized throughout
the cytoplasm, showing whole cells of the labeled colony
(Fig. 3b).
4. Add the Filter population module, and set minimum object
size to remove small objects resulting from noise.
5. Add the Separate touching object module into nuclei objects to
separate incorrectly connected nuclei. This process is necessary
because, in the first step of object identification, two different
nuclei might be mistakenly identified as a single object.
6. Use the Intersect module with nuclei and cell objects. Nuclei of
the labeled colony will be detected (Fig. 3c) (see Note 21).
Analysis for the Heterogeneity of Liver Progenitor Cells 77
Fig. 3 Quantification process of three-dimensional (3D) images. A 3D image is projected on the 2D plane. (a)
Nuclei. (b) Labeled colony visualized with tdTomato. (c) Identified nuclei of tdTomato-positive cells
3.5.2 Quantification by Instead of Volocity, we can use ImageJ/Fiji to quantify the images.
ImageJ (Fiji) An ImageJ plug-in, MorphoLibJ, is required here. Please install
Fiji (https://fiji.sc) [17] and MorphoLibJ (https://github.com/
ijpb/MorphoLibJ/releases) [18] following manufacturer’s
instructions.
1. Load 3D image of nuclei and tdTomato to ImageJ. Serial tiff
images can be imported directly (“File” ! “Import” ! “Image
Sequence. . .”). Alternatively, 3D image files in another format
can be loaded via plug-ins. For example, Olympus image for-
mat can be loaded by “Olympus ImageJ Plugin (https://
imagej.net/OlympusImageJPlugin).”
2. Apply the Gaussian filter to both nuclei channel and tdTomato
channels (“Process” ! “Filters” ! “Gaussian Blur”).
3. Create binary images by setting the fluorescent intensity
threshold (“Image” ! “Adjust” ! “Threshold” (method,
default; dark background, on)). Please adjust threshold value
to properly detect the objects. The optimal threshold value may
change depending on various factors.
4. Merge nuclei and tdTomato channel (“Process” ! “Image
Calculator” (operation, AND)). Set the binary images of nuclei
and tdTomato as input images.
5. Separate touching nuclei by the watershed method (“Plu-
gins” ! “MorphoLibJ” ! “Binary Images” ! “Distance
Transform Watershed 3D” (distances, quasi-Euclidean;
78 Kenji Kamimoto
Fig. 4 Colony size distribution. Liver was analyzed after 0, 2, 4, 6, and 8 weeks of thioacetamide (TAA) injury.
Data are shown as swarm plot and boxplot. Colony size distribution data were reproduced from our previous
study [9]
4 Notes
References
1. Donati G, Watt FM (2015) Stem cell hetero- 11. Clayton E, Doupé DP, Klein AM et al (2007) A
geneity and plasticity in epithelia. Cell Stem single type of progenitor cell maintains normal
Cell 16:465–476. https://doi.org/10.1016/ epidermis. Nature 446:185–189. https://doi.
j.stem.2015.04.014 org/10.1038/nature05574
2. Etzrodt M, Endele M, Schroeder T (2014) 12. Doupé DP, Klein AM, Simons BD, Jones PH
Quantitative single-cell approaches to stem (2010) The ordered architecture of murine ear
cell research. Cell Stem Cell 15:546–558. epidermis is maintained by progenitor cells
https://doi.org/10.1016/j.stem.2014.10. with random fate. Dev Cell 18:317–323.
015 https://doi.org/10.1016/j.devcel.2009.12.
3. Miyajima A, Tanaka M, Itoh T (2014) Stem/ 016
progenitor cells in liver development, homeo- 13. Driessens G, Beck B, Caauwe A et al (2012)
stasis, regeneration, and reprogramming. Cell Defining the mode of tumour growth by clonal
Stem Cell 14:561–574. https://doi.org/10. analysis. Nature 488:527–530. https://doi.
1016/j.stem.2014.04.010 org/10.1038/nature11344
4. Jelnes P, Santoni-Rugiu E, Rasmussen M et al 14. Ke M-T, Fujimoto S, Imai T (2013) SeeDB: a
(2007) Remarkable heterogeneity displayed by simple and morphology-preserving optical
oval cells in rat and mouse models of stem cell- clearing agent for neuronal circuit reconstruc-
mediated liver regeneration. Hepatology tion. Nat Neurosci 16:1154–1161. https://
45:1462–1470. https://doi.org/10.1002/ doi.org/10.1038/nn.3447
hep.21569 15. Zhu L, Gibson P, Currle DS et al (2009) Pro-
5. Suzuki A, Sekiya S, Onishi M et al (2008) Flow minin 1 marks intestinal stem cells that are
cytometric isolation and clonal identification of susceptible to neoplastic transformation.
self-renewing bipotent hepatic progenitor cells Nature 457:603–607. https://doi.org/10.
in adult mouse liver. Hepatology 1038/nature07589
48:1964–1978. https://doi.org/10.1002/ 16. Madisen L, Zwingman TA, Sunkin SM et al
hep.22558 (2010) A robust and high-throughput Cre
6. Okabe M, Tsukahara Y, Tanaka M et al (2009) reporting and characterization system for the
Potential hepatic stem cells reside in EpCAM+ whole mouse brain. Nat Neurosci
cells of normal and injured mouse liver. Devel- 13:133–140. https://doi.org/10.1038/nn.
opment 136:1951–1960. https://doi.org/10. 2467
1242/dev.031369 17. Schindelin J, Arganda-Carreras I, Frise E et al
7. Dorrell C, Erker L, Schug J et al (2011) Pro- (2012) Fiji: an open-source platform for
spective isolation of a bipotential clonogenic biological-image analysis. Nat Publ Group
liver progenitor cell in adult mice. Genes Dev 9:676–682. https://doi.org/10.1038/
25:1193–1203. https://doi.org/10.1101/ nmeth.2019
gad.2029411 18. Legland D, Arganda-Carreras I, Andrey P
8. Ema H, Morita Y, Yamazaki S et al (2006) (2016) MorphoLibJ: integrated library and
Adult mouse hematopoietic stem cells: purifi- plugins for mathematical morphology with
cation and single-cell assays. Nat Protoc ImageJ. Bioinformatics 32:3532–3534.
1:2979–2987. https://doi.org/10.1038/ https://doi.org/10.1093/bioinformatics/
nprot.2006.447 btw413
9. Kamimoto K, Kaneko K, Kok C et al (2016) 19. Forbes SJ, Newsome PN (2016) Liver regen-
Heterogeneity and stochastic growth regula- eration - mechanisms and models to clinical
tion of biliary epithelial cells dictate dynamic application. Nat Rev Gastroenterol Hepatol
epithelial tissue remodeling. elife 5:e15034. 13:473–485. https://doi.org/10.1038/
https://doi.org/10.7554/eLife.15034.001 nrgastro.2016.97
10. Lemaigre FP (2015) Determining the fate of 20. Hama H, Kurokawa H, Kawano H et al (2011)
hepatic cells by lineage tracing: Facts and pit- Scale: a chemical approach for fluorescence
falls. Hepatology 61:2100–2103. https://doi. imaging and reconstruction of transparent
org/10.1002/hep.27659 mouse brain. Nat Neurosci 14:1481–1488.
https://doi.org/10.1038/nn.2928
Chapter 8
Abstract
Following massive hepatocyte ablation in zebrafish, biliary epithelial cells can extensively give rise to
hepatocytes through liver progenitor cells (LPCs). The zebrafish liver injury model is an important system
to elucidate the molecular mechanisms underlying LPC-driven liver regeneration. Here, we describe a
chemical screening method using the zebrafish model for identifying small molecules that can modulate
LPC-driven liver regeneration.
Key words Nitroreductase, Biliary epithelial cells, Hepatocyte ablation, Chemical screen, Liver pro-
genitor cells
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_8, © Springer Science+Business Media, LLC, part of Springer Nature 2019
83
84 Sungjin Ko and Donghun Shin
2 Materials
3 Methods
Fig. 1 Cartoon illustrating the entire chemical screening process for identifying compounds that affect
LPC-driven liver regeneration. Red circles denote livers
3.2 Mtz and 1. Prepare 10-mM Mtz solution (see Subheading 2, item 13, and
Compound Treatments Note 3).
2. Prepare the mixture of each compound and Mtz in a 15-mL
conical tube: add 6 μL of each compound solution (see Note 4)
into 2-mL Mtz solution and vortex (see Note 5). For a control
group, add 6 μL of DMSO into 2-mL Mtz solution (see Note
6).
3. Dispense 10–15 larvae into each well of 12-well tissue culture
plates using a transfer pipette (see Note 5). Remove egg water
from each well and add 2 mL of Mtz/compound mixture. To
prevent the photoinactivation of Mtz and compounds, cover
the plates with aluminum foil, and incubate them at 28 C for
36 h.
4. Prepare each compound solution in egg water: add 6 μL of each
compound solution and 4 μL of DMSO into 2 mL of egg water
supplemented with 0.2-mM PTU and vortex. For a control
group, add 10 μL of DMSO into 2 mL of egg water supple-
mented with 0.2-mM PTU (see Note 5).
5. After 36 h of incubation, to immobilize the larvae, add a lower
concentration of tricaine (final concentration: 0.004%) (see
Note 7). Sort out larvae with well-ablated livers (optional, see
Note 8). Remove the Mtz/compound solution and add 2 mL
of egg water. Gently transfer the embryos into a well of a new
12-well plate using a transfer pipette and swirl the plate twice to
wash the embryos.
6. Remove the egg water and add 2 mL of the compound-
containing egg water. Cover the plates with aluminum foil
and incubate at 28 C for 24 h.
4 Notes
Acknowledgments
References
Abstract
Primary hepatocytes are widely used in regenerative medicine, drug metabolism analysis, and in vitro drug
screens. To overcome the shortage of liver donors, several strategies, such as differentiation of pluripotent
stem cells and transdifferentiation from somatic cells, were developed to generate hepatocytes from
alternative sources. Here, we describe in detail lenti-virus-based procedure for direct conversion of
human fibroblasts to hepatocytes (hiHep cells) in vitro. A detailed protocol for preparation of human
fibroblasts from scar tissues is also provided. Based on this protocol, FOXA3, HNF1A, and HNF4A are
introduced into SV40-large-T-antigen-expressing human scar fibroblasts by lenti-virus. It usually takes
about 5–7 days to get epithelial hiHep colonies. SV40-large-T-antigen-expressing hiHep (hiHepLT) cells
are proliferative and can be expanded to a large number for potential uses.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_9, © Springer Science+Business Media, LLC, part of Springer Nature 2019
93
94 Pengyu Huang et al.
Fig. 1 Representative pictures of human scar fibroblasts and hiHepLT cells. Primary human scar fibroblasts
(passage 3) were generated from human scar tissue and infected with lenti-virus expressing SV40 large T
antigen (left). HiHepLT cells were converted from SV40-large-T-antigen-expressing human scar fibroblasts by
enforced expressions of FOXA3, HNF1A, and HNF4A (right). Scale bar: 500 μm
Fig. 2 Representative pictures of hiHepLT cells stained with albumin and α-1-antitrypsin (AAT) before and after
enrichment. Scale bar: 200 μm
Fig. 3 Albumin and AAT double-positive hiHepLT cells were quantified by flow cytometry before and after
enrichment
2 Materials
2.1 Biological Human scar tissues are taken from scar tissues removed during
Material surgical operations after written consent following guidelines
approved by an Ethic Committee.
3 Methods
3.1 Coating Dish 1. Add collagen I solution to cell culture dishes (1.5-μg/cm2
surface).
2. Air-dry the collagen I-coated dishes in a biosafety cabinet for
future use.
3.2 Preparation 1. CDNA of SV40 large T antigen gene, FOXA3, HNF1A, and
of Lenti-Virus HNF4A were inserted into a pWPI expression vector.
2. Seed 1 106 293FT cells on a 100-mm dish, and add 10-mL
DMEM +10% FBS. Culture 293FT cells for 24 h.
3. One hour before transfection, exchange the culture medium
with 6 mL of prewarmed DMEM+10% FBS.
4. Mix 250-μL calcium phosphate solution containing 10-μg
pWPI expression vector and 250-μL 2 HEPES solution by
pipetting. Keep the mixture for 5 min at room temperature
then transfer the mixture to 293FT cells cultured in a
100-mm dish.
5. Incubate the cells for 6 h and exchange the medium with
DMEM +10% FBS.
6. 48 h after transfection, collect supernatant and filtered by a
0.45-μm filter. Store the virus solution in 80 C.
3.3 Preparation 1. Wash human scar tissue with 70% ethanol three times, then
of Human Scar with PBS once.
Fibroblasts 2. Cut the tissue into 2–5-mm3 pieces.
98 Pengyu Huang et al.
3. Place three tissue pieces per 60-mm collagen I-coated dish, and
incubate at 37 C for 1 h in an incubator.
4. Carefully add 5 mL of HFC medium to the dish (see Note 2).
5. Put the dish in an incubator at 37 C for 5 days without
exchange of the medium, and then exchange the medium
every 2 days.
6. Human scar fibroblasts migrate out of the tissue after
2–3 weeks (passage 1).
7. When the cells become confluent, remove HFC medium and
the tissues.
8. Wash once with PBS and trypsinize with 1 mL of 0.05% trypsin
for 15 min.
9. Suspend the cells with 2 mL of HFC medium. Centrifuge the
cells at 150 g for 5 min.
10. Discard the supernatant, resuspend the cells with 5 mL of HFC
medium, and transfer to a 60-mm collagen I-coated dish (pas-
sage 2).
11. When the cells become confluent, passage the cells to new
collagen I-coated dishes (5 105 cells per 60-mm collagen
I-coated dish, passage 3). Exchange HFC medium every
2 days.
12. For the following passages, seed 5 105 cells per 60-mm
collagen I-coated dish and exchange HFC medium every
2 days.
3.4 Immortalization 1. Seed 5 105 human scar fibroblasts (passage 3–5) on a 60-mm
of Human Scar collagen I-coated dish, and add HFC medium to 5 mL.
Fibroblasts 2. The next day, thaw lenti-virus expressing SV40 large T antigen
on ice. Mix lenti-virus (MOI ¼ 2), 5 μL of 8-mg/mL poly-
brene and HFC medium to the volume of 5 mL.
3. Remove the medium from the dish. Add 5 mL of lenti-virus,
polybrene, and HFC medium mixture.
4. After 24 h infection, remove the medium and add 5 mL of fresh
HFC medium.
5. When the cells become confluent, passage the cells to new
collagen I-coated dishes (5 105 cells per 60-mm dish).
6. For the following passages, seed 5 105 cells per 60-mm
collagen I-coated dish and exchange HFC medium every
2 days.
3.5 Conversion 1. Expand human scar fibroblasts expressing SV40 large T antigen
of HSFLT to hiHepLT (HSFLT) to 70–80% confluence.
Cells 2. Discard the medium and wash twice with 2 mL of PBS.
Hepatic Transdifferentiation 99
3.6 Enrichment 1. On day 12, exchange the hiHep cell culture medium with
of hiHepLT Cells 1% FBS.
2. On day 14, aspirate the medium, and wash twice with 2 mL
of PBS.
3. Add 1 mL of 2-mg/mL collagenase IV solution.
4. Incubate the cells for 5–10 min.
5. Carefully monitor the cells under a microscope. When nearly
50% of the cells curl up and start to detach from the dish
surface, gently tap the dish until most of the fibroblasts disso-
ciate, while most of epithelial colonies remain on the dish.
6. Aspirate the supernatant. Wash with 4 mL of PBS to remove
collagenase IV.
7. Add 1 mL of 0.05% trypsin and incubate the cells at 37 C for
3–5 min.
8. Suspend the cells with hiHep cell culture medium containing
1% FBS.
100 Pengyu Huang et al.
3.7 Passage 1. When the cells become confluent, discard the medium, and
of HiHepLT Cells wash twice with 2 mL of PBS.
2. Trypsinize the cells with 1 mL of 0.05% trypsin at 37 C for
5 min.
3. Add 2 mL of hiHep cell culture medium supplemented with
1% FBS.
4. Suspend the cells, and transfer 5 105 cells to each 60-mm
collagen I-coated dish.
5. Add 5 mL of hiHep cell culture medium with 1% FBS.
6. Exchange medium with fresh hiHep cell culture medium with-
out FBS every day.
7. For functional assays, usually use hiHepLT cells 4–8 days after
seeding (see Note 5).
4 Notes
References
1. Scarpelli DG, Rao MS (1981) Differentiation 2. Shen CN, Horb ME, Slack JM, Tosh D (2003)
of regenerating pancreatic cells into Transdifferentiation of pancreas to liver. Mech
hepatocyte-like cells. Proc Natl Acad Sci U S Dev 120(1):107–116
A 78(4):2577–2581 3. Reddy JK, Rao MS, Qureshi SA, Reddy MK,
Scarpelli DG, Lalwani ND (1984) Induction
Hepatic Transdifferentiation 101
and origin of hepatocytes in rat pancreas. J Cell Li YH, Bao XC, Tse HF, Grillari J, Grillari-
Biol 98(6):2082–2090 Voglauer R, Pei DQ, Esteban MA (2012) Gen-
4. Shen CN, Slack JM, Tosh D (2000) Molecular eration of human induced pluripotent stem
basis of transdifferentiation of pancreas to liver. cells from urine samples. Nat Protoc 7
Nat Cell Biol 2(12):879–887. https://doi. (12):2080–2089. https://doi.org/10.1038/
org/10.1038/35046522 nprot.2012.115
5. Huang P, He Z, Ji S, Sun H, Xiang D, Liu C, 9. Gao Y, Zhang X, Zhang L, Cen J, Ni X, Liao X,
Hu Y, Wang X, Hui L (2011) Induction of Yang C, Li Y, Chen X, Zhang Z, Shu Y,
functional hepatocyte-like cells from mouse Cheng X, Hay DC, Lai D, Pan G, Wei G, Hui
fibroblasts by defined factors. Nature 475 L (2017) Distinct gene expression and epige-
(7356):386–389. https://doi.org/10.1038/ netic signatures in hepatocyte-like cells pro-
nature10116 duced by different strategies from the same
6. Huang P, Zhang L, Gao Y, He Z, Yao D, Wu Z, donor. Stem Cell Reports 9(6):1813–1824.
Cen J, Chen X, Liu C, Hu Y, Lai D, Hu Z, https://doi.org/10.1016/j.stemcr.2017.10.
Chen L, Zhang Y, Cheng X, Ma X, Pan G, 019
Wang X, Hui L (2014) Direct reprogramming 10. Du Y, Wang J, Jia J, Song N, Xiang C, Xu J,
of human fibroblasts to functional and expand- Hou Z, Su X, Liu B, Jiang T, Zhao D, Sun Y,
able hepatocytes. Cell Stem Cell 14 Shu J, Guo Q, Yin M, Sun D, Lu S, Shi Y, Deng
(3):370–384. https://doi.org/10.1016/j. H (2014) Human hepatocytes with drug met-
stem.2014.01.003 abolic function induced from fibroblasts by
7. Shi XL, Gao YM, Yan YP, Ma HC, Sun LL, lineage reprogramming. Cell Stem Cell 14
Huang PY, Ni X, Zhang LD, Zhao X, Ren HZ, (3):394–403. https://doi.org/10.1016/j.
Hu D, Zhou Y, Tian F, Ji Y, Cheng X, Pan GY, stem.2014.01.008
Ding YT, Hui LJ (2016) Improved survival of 11. Ji S, Zhu L, Gao Y, Zhang X, Yan Y, Cen J,
porcine acute liver failure by a bioartificial liver Li R, Zeng R, Liao L, Hou C, Gao Y, Gao S,
device implanted with induced human func- Wei G, Hui L (2017) Baf60b-mediated
tional hepatocytes. Cell Res 26(2):206–216. ATM-p53 activation blocks cell identity con-
https://doi.org/10.1038/cr.2016.6 version by sensing chromatin opening. Cell Res
8. Zhou T, Benda C, Dunzinger S, Huang YH, 27(5):642–656. https://doi.org/10.1038/cr.
Ho JC, Yang JY, Wang Y, Zhang Y, Zhuang Q, 2017.36
Chapter 10
Abstract
In vivo conversion of fibroblasts into hepatocyte-like cells provides one potential approach for the treatment
of liver fibrosis. In our previous study, we showed in vivo conversion of myofibroblasts into induced
hepatocytes (iHeps) by forced expression of four transcription factors in genetic fate-tracing mouse
model of chronic liver disease. These in vivo-generated iHeps showed similar expression profile with
endogenous hepatocytes (eHeps) and also exhibited similar functional characteristics, such as albumin
secretion, urea synthesis, cytochrome activity, and drug responsiveness. Furthermore, the targeted expres-
sion of our reprogramming factors in myofibroblasts attenuated liver fibrosis. Our study suggests that
in vivo reprogramming may open new perspectives for the treatment of diseases such as liver fibrosis.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_10, © Springer Science+Business Media, LLC, part of Springer Nature 2019
103
104 Guangqi Song et al.
B
n ps n ps
ctio f iHe ctio f iHe
nje no nje no
T Fi tio TFi tio
CCl4 injection (twice weekly) .4 tec DDC diet .4 tec
Ad De Ad De
8 weeks 4 weeks
Fig. 1 (a) Schematic of in vivo conversion of myofibroblasts into iHeps in LratCre-mT/mG mice using
p75NTRp-tagged recombinant adenoviral vector. In this lineage-tracing model, the endogenous hepatocytes
(eHeps) would express membranous tdTomato, while myofibroblasts and induced hepatocytes (iHeps) derived
from myofibroblasts would express membranous EGFP. (b) Schematic of in vivo iHep generation in CCl4 and
DDC diet-induced liver fibrosis disease mouse model
2 Materials
2.3 Histology, 1. Picro-Sirius Red solution: 0.1% direct red 80 plus 0.1% fast
Immunohisto- green dissolved in 1.2% saturated aqueous picric acid solution.
chemistry, and 2. Antibodies for immunofluorescence staining: Albumin
Immunofluorescence (Abcam-19196), MUP (Santa Cruz-21856), FAH (Abcam-
81087), AAT (Abcam-117307), p75-NTR (Abcam-8874),
desmin (Thermo Scientific-RB-9140), HNF4A (Santa Cruz-
6556).
3. Hydroxyproline Assay Kit (Sigma).
Conversion of Fibroblasts to Hepatocyte-Like Cells In Vivo 107
Table 1
Primers and probes for detection of exogenous expression
Primers
Table 2
Primers and probes for drug responsiveness assay
Primers
3 Method
A Lrat Cre
Cre
pCA mT pA mG pA pCA mG pA
Fig. 2 (a) Cells in LratCre-mT/mG mice express membrane-targeted tdTomato (mT) before Cre-mediated
recombination. Cre expressed under the transcriptional control of the lecithin-retinol acyltransferase (Lrat)
promoter, which is specifically expressed in hepatic stellate cells in liver. Myofibroblasts derived from hepatic
stellate cells express membrane-targeted EGFP. (b) Schematic illustration shows that eHeps can be identified
by tdTomato expression, whereas iHeps are identified by EGFP expression. (c) These confocal figures show the
generation of in vivo iHeps in LratCre-mT/mG mice after injected with Ad.4TF, no reprogrammed cell was
found in control group
Fig. 3 Isolation of in vivo-generated iHeps (EGFP positive) and eHeps (tdTomato positive) by FACS sorting
3.4 Characterization 1. For analysis of the stability of conversion, total RNA of iHeps
of In Vivo Generated should be isolated. Real-time qPCR can be performed using
iHeps specific primers for four human factors to confirm exogenous
silence. Array-CGH can be performed to test the genomic
integrity of reprogrammed cells using Agilent Genome Micro-
array Kits 4 180k arrays with median overall probe spacing of
about 13 kb. Labeling and hybridization of gDNA should be
performed according to the protocol provided by Agilent.
2. For analysis of the global gene expression profiles, total RNA of
eHeps, iHeps, and myofibroblasts should be isolated from cells
by TRIzol reagent following a standardized protocol. Whole
Mouse Genome Oligo Microarray v2 (4 44K) (Agilent
Technologies) could be used to characterize global gene
Conversion of Fibroblasts to Hepatocyte-Like Cells In Vivo 111
A C D Col1a1 E Hydroxyproline
Albumin
Hydroxyproline contect
Retaive mRNA
(pg/24h/10000 cells)
expression
iHep eHep
(ug/mg)
Albumin
PAS
Control Ad. 4TF Control Ad. 4TF
B
Urea
HE
(g/24h/10000 cells)
Urea
Sirius Red
ICG
iHep eHep
Fig. 4 (a) Albumin ELISA revealed comparable levels of secreted albumin in iHeps and eHeps. (b) Urea
synthesis showed similar levels in iHeps and eHeps. (c) The iHeps show PAS staining, Oil red O staining, and
ICG uptake. (d–f) Amelioration of liver fibrosis in Ad.4TF-injected mice. (d) Reduced levels of Col1a1 mRNA in
Ad.4TF-injected mice compared to control mice. (e) Hydroxyproline assay showed decreased levels of entire
collagen content, measured in whole liver. (f) H&E and Sirius Red staining showed less fibrosis in Ad.4TF-
injected mice than respective controls
3.5 Evaluation 1. For analysis of liver fibrosis level of mice injected with Ad.4TF
of Liver Fibrosis Level and empty adenoviral vector, the expression level of Col1a1 in
After In Vivo the entire liver could be detected by real-time qPCR. The total
Conversion RNA should be isolated from liver tissues that were collected
on the 30th day after viral injection (Fig. 4d).
2. The entire collagen content of a liver could be detected by
hydroxyproline assay. Liver tissues should be collected from
mice injected with Ad.4TF and empty adenoviral vector, and
perform assay following the manual of Hydroxyproline Assay
Kit (Fig. 4e).
3. Paraffine sections of liver tissues from mice injected with
Ad.4TF and empty adenoviral vector can be produced follow-
ing a standardized protocol. For analysis of the fibrotic area,
Sirius Red staining could be performed. Following deparaffini-
zation, the sections should be stained with Picro-Sirius Red
solution and incubated for 60 min. Sections were rinsed with
water, dehydrated and mounted in xylene. Take ten random
photos from each section under a phase contrast microscope.
Fibrotic area could be counted using Image J software (Fig. 4f).
4. For counting the number of fibroblasts in liver tissues, Desmin
and p75-NTR staining can be performed following a standar-
dized immunohistochemistry staining protocol.
4 Notes
1. When you generate liver fibrosis using 10% CCl4, mice should
be monitored carefully. It is highly possible to result in mice’s
death due to overdose injection. You can inject 10% CCl4 two
times per week as shown in Table 3.
2. Conversed iHeps show similar volume to hepatocytes; you
should use 100-μm nylon filter after isolating them from livers.
And iHeps can fall down to the bottom of Falcon tubes in
10 min. Thus, when you perform FACS sorting, invert the
tube gently every 10 min, and use 100-μm nozzle to prevent
cells from blocking the machine.
3. We performed FACS sorting; tdTomato and GFP double-
positive cells could be found in the cells isolated from lineage-
tracing mice. We confirmed that all hepatocytes (and rare
double-positive cells) isolated from control mice are tdTomato
positive. Since LratCre-mTmG model allows specific labeling
of hepatic stellate cells and myofibroblasts with GFP, we believe
that the double-positive cells in a LratCre-mT/mG model may
represent doublets of hepatocytes and myofibroblasts.
4. For the cytochrome P450 enzymes’ activities assay, the
medium should be change every day with inducers. For
114 Guangqi Song et al.
Table 3
Dose of CCl4
Mouse weight 10% CCl4 vol. Mouse weight 10% CCl4 vol. Mouse weight 10% CCl4 vol.
(g) (μL) (g) (μL) (g) (μL)
14 56 23 92 32 128
15 60 24 96 33 132
16 64 25 100 34 136
17 68 26 104 35 140
18 72 27 108 36 144
19 76 28 112 37 148
20 80 29 116 38 152
21 84 30 120 39 156
22 88 31 124 40 160
References
1. Huang PY, He ZY, Ji SY, Sun HW, Xiang D, human fibroblasts to functional melanocytes by
Liu CC, Hu YP, Wang X, Hui LJ (2011) defined factors. Nat Commun 5:5807
Induction of functional hepatocyte-like cells 5. Song G, Pacher M, Balakrishnan A, Yuan Q,
from mouse fibroblasts by defined factors. Tsay HC, Yang D, Reetz J, Brandes S, Dai Z,
Nature 475:386–U142 Putzer BM et al (2016) Direct reprogramming
2. Huang P, Zhang L, Gao Y, He Z, Yao D, Wu Z, of hepatic myofibroblasts into hepatocytes
Cen J, Chen X, Liu C, Hu Y et al (2014) Direct in vivo attenuates liver fibrosis. Cell Stem Cell
reprogramming of human fibroblasts to func- 18:797–808
tional and expandable hepatocytes. Cell Stem 6. Reetz J, Genz B, Meier C, Kowtharapu BS,
Cell 14:370–384 Timm F, Vollmar B, Herchenroder O,
3. Sekiya S, Suzuki A (2011) Direct conversion of Abshagen K, Putzer BM (2013) Development
mouse fibroblasts to hepatocyte-like cells by of adenoviral delivery systems to target hepatic
defined factors. Nature 475:390–393 stellate cells in vivo. PLoS One 8:e67091
4. Yang R, Zheng Y, Li L, Liu S, Burrows M, 7. Mederacke I, Hsu CC, Troeger JS,
Wei Z, Nace A, Herlyn M, Cui R, Guo W Huebener P, Mu X, Dapito DH, Pradere JP,
et al (2014) Direct conversion of mouse and Schwabe RF (2013) Fate tracing reveals hepatic
Conversion of Fibroblasts to Hepatocyte-Like Cells In Vivo 115
Abstract
Bipotent liver progenitor cells (LPCs) are promising cell sources for cell transplantation therapy in hepatic
disorders as well as biliary dysfunctions. Using a cocktail of small molecules, we recently reported a novel
approach to generate bipotent LPCs, named chemically induced liver progenitors (CLiPs), from adult rat
hepatocytes. In this chapter, we describe a detailed protocol for the induction of rat CLiPs. We first describe
the method to isolate primary rat hepatocytes and then describe how to induce CLiPs from the hepatocytes.
In addition, we describe methods to induce the generated CLiPs to differentiate into hepatocytes and
biliary epithelial cells.
Key words Hepatocyte, Biliary epithelial cell, Liver progenitor cell, CLiP, Reprogramming,
Bipotentiality
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_11, © Springer Science+Business Media, LLC, part of Springer Nature 2019
117
118 Takeshi Katsuda and Takahiro Ochiya
2 Materials
2.1 Animal 1. Wistar rats aged at 5 weeks to 20 weeks (both male and female
and Reagents animals can be used).
2. Pre-perfusion buffer: Dissolve 40 g NaCl, 2.0 g KCl, 0.39 g
NaH2PO4·2H2O, 0.76 g Na2HPO4·12H2O, 4.5 g glucose,
1.9 g EGTA, 3.7 g EDTA, 0.0030 g phenol red, and 11.9 g
HEPES in 500 mL distilled water to prepare 10 stock solu-
tion. Dilute 50 mL of 10 stock solution with 450 mL distilled
water. After autoclave sterilization, add 2.35 mL of 7.5%
NaHCO3 solution and 2 mL of 1 N NaOH solution.
3. 7.5% NaHCO3 solution: Dissolve 7.5 g of NaHCO3 in 100 mL
dH2O. Filter with a 0.22 μm filter, and store at 4 C until use.
Use within 1 month.
4. 0.05% collagenase solution: Dissolve 9.6 g NaCl, 0.48 g KCl,
0.094 g NaH2PO4·2H2O, 0.18 g Na2HPO4·12H2O, 2.86 g
HEPES, 0.888 g CaCl2·2H2O, and 0.42 g NaHCO3 in 1.2 L
of dH2O supplemented with 14.4 mL of 0.05% phenol red
solution. Store this basal solution at 4 C until use. For perfu-
sion, pour 400 mL into a beaker and adjust pH to 7–7.4 with
5 N NaOH and/or 6 N HCl. Then, dissolve 0.02 g trypsin
inhibitor (Sigma-Aldrich) and 0.2 g collagenase (Wako). After
adjusting the pH to 7.6 with 5 N NaOH and/or 6 N HCl,
filter with a 0.22 μm filter, and store at 4 C until use. The
prepared collagenase solution should be used within 1 week.
5. Hepatocyte wash medium: E-MEM.
6. Percoll (GE Healthcare).
7. L-15-based medium for complete Percoll solution: Dissolve
the following reagents in 99 mL of Leibovitz’s L-15 medium
(Gibco, with additives: Sodium pyruvate; L-Glutamine; Phenol
red): 0.2 g bovine serum albumin (BSA) (Sigma-Aldrich) and
0.0429 g HEPES (Sigma-Aldrich). Filter with a 0.22 μm filter,
add 1 mL of antibiotic/antimycotic (Gibco), and store at 4 C
until use.
8. 0.4% trypan blue solution.
Generation of Chemically Induced Liver Progenitors 119
3 Methods
3.1 Isolation of Rat Rat primary MHs are isolated using the two-step collagenase per-
Primary MHs fusion method [10]. In the first step, blood is eliminated from the
liver by perfusing the liver with pre-perfusion buffer. In the second
step, the liver is digested by perfusing collagenase solution. Then,
the liver is extracted, minced, and further digested ex vivo with the
remaining collagenase solution. MHs are collected by sequential
low-speed centrifugation at 57 g (see Note 1). This sequential
low-speed centrifugation allows exclusion of nonparenchymal cells.
1. Before starting, place 20 mL E-MEM at room temperature,
which will be used for additional digestion of the extracted
liver.
2. Anesthetize a rat by inhalation of isoflurane vapor. Place a paper
towel cushion on a foam board or cork board, and lay the rat on
the cushion. Sterilize the animal by wiping its fur with 70%
EtOH. Open the abdomen using surgical scissors to expose the
liver.
3. Start drip of pre-perfusion buffer at 25–30 mL/min. Half-cut
the portal vein at the location approximately 1.5 cm from the
bifurcation of the portal vein, and insert the cannula.
4. Immediately after confirming that the color of the liver changes
from reddish brown to yellowish, cut the inferior vena cava.
Perfuse approximately 450 mL pre-perfusion buffer. It takes
approximately 15 min.
5. Pause the pump, transfer the inlet tube to the collagenase
bottle, and resume perfusion. Perfuse approximately
300–350 mL collagenase solution. It takes approximately
10 min.
6. Stop perfusion, and remove the cannula from the portal vein.
Cut the liver from the abdominal cavity, and transfer it to a
100 mm sterile plastic dish. Remove the attaching surrounding
tissue, such as the diaphragm (see Note 2).
7. Add 20 mL E-MEM kept at room temperature and 20 mL of
the remaining collagenase solution. Mince the liver with
122 Takeshi Katsuda and Takahiro Ochiya
3.2 Induction Rat CLiPs can be induced from primary MHs upon stimulation
of Primary CLiPs Using with a combination of three small molecules, YAC. An evident
the Small Molecule morphological change compared with YAC-free (YAC( )) culture
Cocktail YAC occurs approximately 4–5 days after plating. As previously reported,
rodent MHs can divide several times in vitro even without YAC, but
their proliferation completely stops thereafter [11, 12]. In contrast,
MHs can continuously proliferate to produce CLiPs during 2-week
culture (Fig. 1). When CLiPs reach 70–100% confluency, harvest
CLiPs to perform hepatic induction or biliary induction, establish
stable CLiPs, or prepare frozen stocks for future use.
1. One day after plating primary MHs, replace the culture
medium with fresh media. Thereafter, renew the medium
every 2–3 days. Given that proliferative YAC(+) cells are bipo-
tential (see the following sections), we designate these cells as
chemically induced liver progenitors (CLiPs).
2. Wash plates with PBS( ) twice.
3. Add 0.05% trypsin-EDTA or TrypLE Express (these two
reagents can be used equivalently for harvesting CLiPs), and
incubate at 37 C in a CO2 incubator until the cells are
detached from the plate (see Notes 8 and 9).
4. Further dissociate the cells physically by pipetting with a P1000
tip to harvest them as completely as possible, and transfer them
to a 15 mL or 50 mL collection tube (see Note 10).
5. Collect the cells remaining in the plate with 5 mL SHM + YAC
supplemented with 5% FBS or SHM + YAC for trypsin-EDTA
or TrypLE Express (TrypLE Express does not require neutrali-
zation of the enzymatic activity).
6. Count the cell number using a hemocytometer, determine the
suspension volume to be used for passage, transfer the required
volume to a new tube, and centrifuge the tube at 200 g for
5 min. Aspirate the supernatant, and loosen the cell pellet by
tapping the tubes. Resuspend the cells with medium to be used
for desired application.
124 Takeshi Katsuda and Takahiro Ochiya
Fig. 1 Morphological change during reprogramming of MHs into CLiPs. MHs actively divide in the presence of
YAC and become smaller in size and exhibit higher nucleus/cytoplasm ratio compared with MHs. This figure is
reproduced from ref. [9]
A
Hep-i(-) D6 Hep-i(-) D8
D0
Hep-i(+) D6 Hep-i(+) D8
Matrigel
100 mm
overlay
OsM
+ 10-6 M
dexamethasone
B
Alb Ttr G6pc
25 20 200
Relative expression to Actb [a.u.]
20 15 150
15
10 100
10
5 5 50
0 0 0
D0 Hep-i(-) Hep-i(+) D0 Hep-i(-) Hep-i(+) D0 Hep-i(-) Hep-i(+)
Fig. 2 Hepatic induction of CLiPs. (a) Morphological change of CLiPs during hepatic induction. Arrowheads
indicate cells that are rich in granules in their cytoplasm. Arrows indicate bile canaliculi formed at the
boundaries between cells that achieve hepatic maturation. (b) qRT-PCR of hepatic marker genes for Hep-i(+),
Hep-i( ), and D0 cells. The data are shown as the mean S.E.M. of four independent experiments. This
figure is partly reproduced from ref. [9]
126 Takeshi Katsuda and Takahiro Ochiya
3.4 Biliary Induction CLiPs can be differentiated also into BECs using a two-step proto-
of CLiPs col (Fig. 3a). In Step 1, CLiPs are co-cultured on cell cycle-arrested
mouse embryonic fibroblasts (MEFs) in mTeSR1 + YAC for 6 days.
In Step 2, CLiPs are cultured for another 6 days in 2% Matrigel-
containing mTeSR1 + YAC. CLiP-derived BEC-like cells form
ductal and cystic structures typically observed in Step 2 (Fig. 3b).
CLiP-derived ductal/cystic structures exhibit the capacity to trans-
port water into their luminal space in response to secretin stimula-
tion (Fig. 3c). Following incubation with fluorescein diacetate,
these cells also transport fluorescein to their luminal spaces
(Fig. 3d).
1. One day before harvesting CLiPs, seed cell cycle-arrested
MEFs on 12-well collagen plates at 5 104 cells/well.
2. Harvest the primary CLiPs following the steps described in
Subheading 3.2, and obtain the desired volume of the cell
suspension for biliary induction. We seed the cells at 5 105
cells/well in 12-well plates. Centrifuge the tube at 200 g for
5 min.
3. Aspirate the supernatant, resuspend the cells in 1 mL
SHM + YAC supplemented with 5% FBS, and seed onto the
pre-inoculated MEFs (see Note 12).
4. (Step 1) On the following day, start biliary induction by repla-
cing the medium with BIM-1. Culture the cells for 6 days while
replacing the medium with fresh BIM-1 every other day.
5. (Step 2) Replace the medium with BIM-2. Culture the cells for
another 6 days while replacing the medium with fresh BIM-2
every other day (see Notes 14 and 15).
3.5 Long-Term CLiPs can be cultured stably through continuous passages; how-
Culture of CLiPs ever, the proliferation rates transiently decrease from approximately
Through Continuous passage 2 (P2) to 7 (P7) (see Notes 16 and 17). During long-term
Passages culture, the cells can be frozen for backup or future use (see Sub-
heading 3.6). Primary CLiPs are typically passaged on days 14–16.
Generation of Chemically Induced Liver Progenitors 127
Fig. 3 Biliary induction of CLiPs. (a) Morphological change of CLiPs during biliary induction. Arrows indicate
ductular structures formed between adjacent cells. (b) Phase-contrast images of BEC-i(+) cells taken before
and 30 min after the addition of secretin. Arrowheads denote enlarged lumens following secretin treatment. (c)
Uptake and excretion of fluorescein diacetate (FD) by the ductular structures of BEC-i(+) cells. Phase-contrast
and fluorescence images were taken immediately after a 15-min incubation in the presence of FD (upper
panel) and after another 30-min incubation in the absence of FD. This figure is partly reproduced from ref. [9]
3.6 Freeze Stocking 1. Harvest the cells following the steps described in Subheading
of CLiPs 3.2, and obtain a desired volume of the cell suspension for
preparation of frozen stocks (typically 0.5–10 106 cells/
vial). Centrifuge the tube at 200 g for 5 min.
2. Aspirate the supernatant, and resuspend the cells in desired
volume of Cell Banker. The volume of Cell Banker used is
0.5 mL/vial.
3. Aliquot to serum tubes equipped with screw caps. Seal the caps
tightly, and store at 80 C for at least 1 day. Then, transfer
them to liquefied nitrogen.
4 Notes
Acknowledgments
References
1. Woo DH, Kim SK, Lim HJ et al (2012) Direct 494:247–250. https://doi.org/10.1038/
and indirect contribution of human embryonic nature11826
stem cell-derived hepatocyte-like cells to liver 8. Lu W-Y, Bird TG, Boulter L et al (2015)
repair in mice. Gastroenterology Hepatic progenitor cells of biliary origin with
142:602–611. https://doi.org/10.1053/j. liver repopulation capacity. Nat Cell Biol
gastro.2011.11.0301 17:971–983. https://doi.org/10.1038/
2. Oertel M, Menthena A, Chen Y-Q et al (2008) ncb3203
Purification of fetal liver stem/progenitor cells 9. Katsuda T, Kawamata M, Hagiwara K et al
containing all the repopulation potential for (2017) Conversion of terminally committed
normal adult rat liver. Gastroenterology hepatocytes to culturable bipotent progenitor
134:823–832. https://doi.org/10.1053/j. cells with regenerative capacity. Cell Stem Cell
gastro.2008.01.007 20:41–55. https://doi.org/10.1016/j.stem.
3. Liu H, Kim Y, Sharkis S et al (2011) In vivo 2016.10.007
liver regeneration potential of human induced 10. Seglen PO (1976) Preparation of isolated rat
pluripotent stem cells from diverse origins. Sci liver cells. Methods Cell Biol 13:29–83.
Transl Med 3:82ra39. https://doi.org/10. https://doi.org/10.1016/S0091-679X(08)
1126/scitranslmed.3002376 61797-5
4. Zhu S, Rezvani M, Harbell J et al (2014) 11. Loyer P, Cariou S, Glaise D et al (1996)
Mouse liver repopulation with hepatocytes Growth factor dependence of progression
generated from human fibroblasts. Nature through G1 and S phases of adult rat hepato-
508:93–97. https://doi.org/10.1038/ cytes in vitro. Evidence of a mitogen restriction
nature13020 point in mid-late G1. J Biol Chem
5. Sekiya S, Suzuki A (2011) Direct conversion of 271:11484–11492
mouse fibroblasts to hepatocyte-like cells by 12. Frémin C, Bessard A, Ezan F et al (2009) Mul-
defined factors. Nature 475:390–393. tiple division cycles and long-term survival of
https://doi.org/10.1038/nature10263 hepatocytes are distinctly regulated by extracel-
6. Huang P, He Z, Ji S et al (2011) Induction of lular signal-regulated kinases ERK1 and ERK2.
functional hepatocyte-like cells from mouse Hepatology 49:930–939. https://doi.org/10.
fibroblasts by defined factors. Nature 1002/hep.22730
475:386–389. https://doi.org/10.1038/ 13. Kamiya A, Kojima N, Kinoshita T et al (2002)
nature10116 Maturation of fetal hepatocytes in vitro by
7. Huch M, Dorrell C, Boj SF et al (2013) In extracellular matrices and oncostatin M: induc-
vitro expansion of single Lgr5+ liver stem cells tion of tryptophan oxygenase. Hepatology
induced by Wnt-driven regeneration. Nature 35:1351–1359. https://doi.org/10.1053/
jhep.2002.33331
Chapter 12
Abstract
Human-induced pluripotent stem cells (iPSCs) could be a useful source for production of hepatocytes.
Here, we develop protocols to generate iPSC-derived liver progenitor cells, liver sinusoidal endothelial cells
(LSECs), and hepatic stellate cells (HSCs). We also establish long-term two-dimensional co-culture system
to induce functional hepatocytes from iPSC-derived liver cells.
Key words Human-induced pluripotent stem cells, Liver, Liver progenitor cells, Hepatocytes, Liver
sinusoidal endothelial cells, Hepatic stellate cells
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_12, © Springer Science+Business Media, LLC, part of Springer Nature 2019
131
132 Taketomo Kido and Yuta Koui
Table 1
Cell surface specific markers on liver cells
2 Materials
3 Methods
a b c d
e f
iso CPM
Fig. 1 Generation of LPCs from human iPSCs. (a–d) Morphological changes of iPSCs at different stages of
hepatic differentiation. Scale bar, 100 μm. (e) FCM analysis of CPM expression. (f) Morphology of the CPM+
LPC colonies on MEF feeder cells after 7 days of culture. Scale bar, 100 μm
3.1.2 Isolation and 1. One day before isolation of iPSC-derived LPCs: Plate the
Expansion of iPSC- mitomycin C-treated MEF cells at a density of 5–8 104
Derived LPCs cells/cm2 onto a 0.1% gelatin-coated 12-well plate in MEF
culture medium. Incubate the cells overnight at 37 C with
ambient O2/5% CO2.
2. Remove the HGF medium from each well of 6-well plates, and
wash once with D-PBS ( ).
3. Aspirate D-PBS ( ), add 0.5 mL of Trypsin/EDTA solution.
4. Incubate cells at 37 C for 10–15 min. Harvest cells into 15 mL
tube, and add MEF culture medium for wash.
5. Centrifuge cells for 3 min at 340 g. Resuspend cells in MEF
culture medium, and pass through 70 μm Cell Strainer to
remove the debris.
6. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS.
7. Add FcR Blocking Reagent, and incubate the cells for 20 min
on ice.
8. Add anti-Carboxypeptidase M (CPM) antibody, and incubate
the cells for 30 min on ice. Use Purified Mouse IgG1 isotype
control as negative control.
9. Add appropriate volume of 0.3% BSA/PBS for wash.
10. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS.
136 Taketomo Kido and Yuta Koui
11. Add PE Goat anti-mouse IgG antibody, and incubate the cells
for 30 min on ice (see Note 3).
12. Wash as in step 9.
13. Add propidium iodide to identify dead cells, and pass through
35 μm Cell Strainer.
14. Isolate iPSC-derived CPM+ LPCs using a MoFlo XDP cell
sorter (Fig. 1e).
15. Centrifuge iPSC-derived CPM+ LPCs for 3 min at 340 g.
Resuspend cells in LPC culture medium.
16. Plate the cells at a density of 1–2 104 cells/cm2 on mitomy-
cin C-treated MEF feeder cells.
17. Incubate cells for 7–10 days at 37 C with ambient O2/5%
CO2. Change medium every day (Fig. 1f) (see Note 4).
3.2 iPSC-Derived 1. Remove the iPSC culture medium from each 6 cm culture dish,
LSECs and wash once with D-PBS ( ).
3.2.1 Induction of LSEC 2. Aspirate D-PBS ( ), add 1 mL of EDTA solution, and incubate
Progenitors from Human cells at 37 C for 5 min.
iPSCs 3. Aspirate EDTA solution, and add 3 mL of iPSC culture
medium. Dissociate cells into small clusters, harvest cells into
15 mL tube, and add iPSC culture medium for wash (see Note
5).
4. Centrifuge cells for 3 min at 340 g. Resuspend cells in
StemPro medium with 10 μM Y-27632 and 2 ng/mL BMP-4.
5. Plate the cell clusters on Ultra-Low Attachment 6-well plates,
and incubate cells overnight at 37 C with 4% O2/5% CO2
(Day 0).
6. Harvest cells into 15 mL tube and centrifuge cells for 2 min at
120 g. Resuspend cells in StemPro medium with 5 ng/mL
Recombinant Activin A, 5 ng/mL Recombinant Human
FGF-basic (154 a.a.), and 30 ng/mL BMP-4 (see Note 6).
7. Replate the cell clusters on Ultra-Low Attachment 6-well
plates, and incubate cells for 3 days at 37 C with 4% O2/5%
CO2 (Day 1–4).
8. Harvest cells into 15 mL tube and centrifuge cells for 2 min at
120 g. Resuspend cells in StemPro medium with 10 ng/mL
Recombinant Human VEGF 165, 5.4 μM SB431542, and
0.5 μM Dorsomorphin dihydrochloride (see Note 6).
9. Replate the cell clusters on Ultra-Low Attachment 6-well
plates, and incubate cells for 2 days at 37 C with 4% O2/5%
CO2 (Day 4–6).
10. Harvest cells into 15 mL tube and centrifuge cells for 2 min at
120 g. Resuspend cells in EC culture medium.
Induction of Functional Hepatocytes from Human iPSCs 137
11. Replate the cell clusters at a 1:2 split ratio onto a 0.1% gelatin-
coated 6-well plate.
12. Incubate the cells for 7 days at 37 C with 4% O2/5% CO2.
Change medium every other day (see Note 7).
3.2.2 Isolation and 1. Remove the EC culture medium from each well of 6-well
Expansion of iPSC-Derived plates, and wash once with D-PBS ( ).
LSEC Progenitors 2. Aspirate D-PBS ( ), and add 0.5 mL of Trypsin/EDTA
solution.
3. Incubate cells at 37 C for 10 min. Harvest cells into 15 mL
tube, and add MEF culture medium for wash.
4. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS, and pass through 70 μm Cell Strainer to remove the
debris.
5. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS.
6. Add FcR Blocking Reagent, and incubate the cells for 20 min
on ice.
7. Add FITC conjugated anti-CD34 antibody, PE conjugated
anti-CD31 antibody, and APC conjugated anti-CD309
(FLK1) antibody, and incubate the cells for 30 min on ice.
Use FITC mouse IgG1 isotype control, PE mouse IgG1 iso-
type control, and APC mouse IgG1 isotype control as negative
control.
8. Add appropriate volume of 0.3% BSA/PBS for wash.
9. Centrifuge cells for 3 min at 340 g. Resuspend cells in
autoMACS Running Buffer.
10. Add Anti-FITC microbeads and incubate the cells for 20 min
on ice.
11. Add appropriate volume of autoMACS Running Buffer
for wash.
12. Centrifuge cells for 3 min at 340 g. Resuspend cells in
autoMACS Running Buffer.
13. Enrich FITC+ cells using autoMACS Pro Separator (Miltenyi
Biotec), and centrifuge cells for 3 min at 340 g. Resuspend
cells in 0.3% BSA/PBS.
14. Add propidium iodide to identify dead cells, and pass through
35 μm Cell Strainer.
15. Isolate iPSC-derived CD34+CD31+FLK1+ LSEC progenitors
using a MoFlo XDP cell sorter (Fig. 2).
16. Centrifuge iPSC-derived CD34+CD31+FLK1+ LSEC progeni-
tors for 8 min at 340 g. Resuspend cells in EC culture
medium.
138 Taketomo Kido and Yuta Koui
CD34+ gated
FLK1
5 5
10 10
4 4
10 10
3 3
10 10
2 2
10 10
1 1
10 10
100 100
0 1 2 3 4 5 0 1 2 3 4 5
10 10 10 10 10 10 10 10 10 10 10 10
CD34 CD31
Fig. 2 Generation of LSEC progenitors from human iPSCs. FCM analysis of iPSC-derived LSEC progenitors after
enrichment of CD34+ cells using autoMACS Pro Separator. CD34+ cells were identified (left), and CD31+FLK1+
cells were identified in CD34+ cell fraction
3.2.3 Induction of iPSC- 1. Remove the EC culture medium from each well of 12-well
Derived Mature LSECs from plates, and wash once with D-PBS ( ).
LSEC Progenitors 2. Aspirate D-PBS ( ), and add 0.5 mL of Trypsin/EDTA
solution.
3. Incubate cells at 37 C for 10 min. Harvest cells into 15 mL
tube, and add MEF culture medium for wash.
4. Centrifuge cells for 3 min at 340 g. Resuspend cells in LSEC
culture medium.
5. Plate the cells at a density of 1.5 104 cells/cm2 on 12-well
plates coated with Fibronectin solution.
6. Incubate cells for 7 days at 37 C with 4% O2/5% CO2. Change
medium every other day.
7. Passage as in Subheading 3.2.3, steps 1–5.
8. Incubate cells for 7 days at 37 C with 4% O2/5% CO2. Change
medium every other day (see Note 9).
Induction of Functional Hepatocytes from Human iPSCs 139
3.3.2 Isolation of HSC 1. Harvest cells into 15 mL tube and centrifuge cells for 3 min at
Progenitors and Induction 340 g.
of iPSC-Derived 2. Aspirate the medium and resuspend cells in Accumax Cell
Mature HSCs Dissociation Solution.
3. Incubate cells at 37 C water bath for 10 min.
4. Add MEF culture medium to inactivate dissociation solution.
5. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS, and pass through 70 μm Cell Strainer to remove the
debris.
6. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS.
7. Add FcR Blocking Reagent, and incubate the cells for 20 min
on ice.
8. Add Biotin conjugated anti-CD166 (ALCAM) antibody, and
incubate the cells for 30 min on ice. Use REA Control (S)-
Biotin as negative control.
9. Add appropriate volume of 0.3% BSA/PBS for wash.
10. Centrifuge cells for 3 min at 340 g. Resuspend cells in 0.3%
BSA/PBS.
11. Add Streptavidin-APC, and incubate the cells for 20 min
on ice.
12. Wash as in step 9.
13. Add propidium iodide to identify dead cells, and pass through
35 μm Cell Strainer.
14. Isolate iPSC-derived ALCAMhigh HSC progenitors using a
MoFlo XDP cell sorter (Fig. 3).
15. Centrifuge iPSC-derived ALCAMhigh HSC progenitors for
3 min at 340 g. Resuspend cells in HSC culture medium.
16. Plate the cells at a density of 1.5 104 cells/cm2 on Cellmatrix
Type I-C-coated 24-well plates.
17. Incubate cells for 5 days at 37 C with ambient O2/5% CO2.
Change medium every other day (see Note 10).
140 Taketomo Kido and Yuta Koui
5 5
10 10
4 4
10 10
3 3
10 10
2 2
10 10
1 1
10 10
0 0.26 0 36.4
10 10
0 1 2 3 4 5 0 1 2 3 4 5
10 10 10 10 10 10 10 10 10 10 10 10
iso ALCAM
Fig. 3 Generation of HSC progenitors from human iPSCs. FCM analysis of ALCAM expression in iPSC-derived
mesodermal cells (right). The positive gate was defined by the isotype control (left)
3.4 High-Density 1. Remove the medium from LPC culture plates (from Subhead-
Co-culture System by ing 3.1.2, step 17), and wash once with D-PBS ( ).
iPSC-Derived LPCs, 2. Aspirate D-PBS ( ), add 0.5 mL of Trypsin/EDTA solution.
LSECs, and HSCs
3. Incubate cells at 37 C for 10–15 min. Harvest cells into 15 mL
3.4.1 Preparation of tube, and add MEF culture medium for wash.
iPSC-Derived LPCs 4. Centrifuge cells for 3 min at 340 g. Resuspend cells in MEF
culture medium, and pass through 70 μm Cell Strainer to
remove the debris.
5. Centrifuge cells for 3 min at 340 g. Resuspend cells in MEF
culture medium.
6. Plate the cells onto a 0.1% gelatin-coated 10 cm culture dish.
Incubate cells for 30 min to remove adherent MEF cells (see
Note 11).
7. Harvest non-adherent cells (iPSC-derived LPCs) into 15 mL
tube and centrifuge for 3 min at 340 g. Resuspend cells in
LPC culture medium.
3.4.2 Preparation of 1. Remove the medium from each culture plate (Subheading
iPSC-Derived LSECs 3.2.3, step 8, and Subheading 3.3.2, step 17), and wash
and HSCs once with D-PBS ( ).
2. Aspirate D-PBS ( ), and add 0.5 mL of Trypsin/EDTA
solution.
3. Incubate cells at 37 C for 5–10 min. Harvest cells into 15 mL
tube, and add MEF culture medium for wash.
4. Centrifuge cells for 3 min at 340 g. Resuspend cells in LPC
culture medium.
Induction of Functional Hepatocytes from Human iPSCs 141
3.4.3 Co-culture of iPSC- 1. Preparation of collagen gel-coated plates: Add 0.2 mL of colla-
Derived LPCs, LSECs, gen gel solution to coat each well of a 48-well plate, and
and HSCs incubate the plate for 30 min at 37 C.
2. Plate iPSC-derived LPCs (2 105 cells/well), iPSC-derived
LSECs (2 104 cells/well), and iPSC-derived HSCs (2 104
cells/well) on collagen gel-coated 48-well plates.
3. Incubate cells for 10 weeks at 37 C with ambient O2/5% CO2.
Change medium every other day (see Note 12).
4 Notes
CYP3A4 activity
70
(2 weeks = 1.0)
50
40
30
20
10
0
2 3 4 5 6 7 8 9 10 weeks
Fig. 4 CYP3A4 activity. Relative Cytochrome P450 3A4 (CYP3A4) activity in iPSC-
derived liver cells. The results are shown as the mean SEM of three
independent experiments. The measurements were performed every week
Acknowledgment
References
1. Takahashi K, Tanabe K, Ohnuki M, Narita M, signaling promote the maturation of human plu-
Ichisaka T, Tomoda K, Yamanaka S (2007) ripotent stem cell-derived hepatocytes. Develop-
Induction of pluripotent stem cells from adult ment 140:3285–3296
human fibroblasts by defined factors. Cell 5. Matsumoto K, Yoshitomi H, Rossant J, Zaret KS
131:861–872 (2001) Liver organogenesis promoted by endo-
2. Si-Tayeb K, Noto FK, Nagaoka M, Li J, Battle thelial cells prior to vascular function. Science
MA, Duris C, North PE, Dalton S, Duncan SA 294:559–563
(2010) Highly efficient generation of human 6. Yin C, Evason KJ, Asahina K, Stainier DY
hepatocyte-like cells from induced pluripotent (2013) Hepatic stellate cells in liver develop-
stem cells. Hepatology 51:297–305 ment, regeneration, and cancer. J Clin Invest
3. Takayama K, Inamura M, Kawabata K, 123:1902–1910
Sugawara M, Kikuchi K, Higuchi M, 7. Kido T, Koui Y, Suzuki K, Kobayashi A,
Nagamoto Y, Watanabe H, Tashiro K, Miura Y, Chern EY, Tanaka M, Miyajima A
Sakurai F, Hayakawa T, Furue MK, Mizguchi (2015) CPM is a useful cell surface marker to
H (2012) Generation of metabolically function- isolate expandable bi-potential liver progenitor
ing hepatocytes from human pluripotent stem cells derived from human iPS cells. Stem Cell
cells by FOXA2 and HNF1a transduction. J Rep 5:508–515
Hepatol 57:628–636 8. Koui Y, Kido T, Ito T, Oyama H, Chen SW,
4. Ogawa S, Surapisitchat J, Virtanen C, Ogawa M, Katou Y, Shirahige K, Miyajima A (2017) An
Niapour M, Sugamori KS, Wang S, Tamblyn L, in vitro human liver model by iPSC-derived
Guillemette C, Hoffmann E, Zhao B, Strom S, parenchymal and non-parenchymal cells. Stem
Laposa RR, Tyndale RF, Grant DM, Keller G Cell Rep 9:490–498
(2013) Three-dimensional culture and cAmp
Chapter 13
Abstract
Inducible pluripotent stem (iPS) cells are multipotent stem cells that are produced by gene transfer of
reprogramming factors to somatic cells. They are thought to be an important source of regenerative
medicine because of their pluripotency and self-renewal ability. Although the liver has high regeneration
ability, continuous death of hepatocytes due to chronic inflammation leads to liver cirrhosis and liver
carcinoma. With regard to such serious liver diseases, liver transplantation is used as a complete cure, but
there is a problem of donor shortage. Therefore, transplantation therapy using liver tissue generated from
stem cells in vitro is expected.
We are developing a system to induce the differentiation of cholangiocytes, one of important
non-parenchymal cells in living liver tissue, from human iPS cells. Bile duct-like cystic structures can be
induced by purifying human iPS cell-derived hepatoblasts expressing hepatic progenitor cell surface markers
and inducing differentiation under appropriate culture conditions. These cells are considered to be useful in
constructing a hepatic organoid that reproduces the liver structure of the living body.
Key words Hepatic progenitor cells, Human iPS cells, Induction of hepatic differentiation, Long-
term proliferation, Purification of progenitor cell
1 Introduction
The liver is the largest organ in the body and contains various
functions for maintaining homeostasis in the body. Hepatocytes
(liver parenchymal cells) express various genes for mature liver
function such as synthesis of serum proteins, metabolism of carbo-
hydrates and lipids, and detoxification of alcohol and drugs. In
addition, several types of non-parenchymal cells are also present in
the liver. Cholangiocytes constitute the intrahepatic bile ducts.
Stellate cells are involved in the accumulation of vitamin
A. Kupffer cells are intrahepatic macrophages, and sinusoidal endo-
thelial cells constitute liver sinusoidal vessels in the liver. It is known
that cell-cell interactions between these non-parenchymal cells and
hepatocytes are important for liver function. Cholangiocytes
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_13, © Springer Science+Business Media, LLC, part of Springer Nature 2019
143
144 Akihide Kamiya et al.
constitute the bile duct system for excretion of bile acid derived
from hepatocytes [1]. Bile acids synthesized by hepatocytes are
released from the apical side of the hepatocyte membrane to the
bile canaliculus and are excreted into the small intestine and gall-
bladder through the intrahepatic bile duct network formed by
cholangiocyte. Therefore, disorders of the intrahepatic bile ducts
such as primary biliary cholangitis lead to poor elimination of bile
acid from the liver and cause various liver injuries.
During embryonic development, the ventral foregut endoderm
begins to form early liver buds by migrating into the septum
transversum (embryonic day 8–9 in mice) [2, 3]. Subsequently,
hepatic buds grow by soluble factor signals from surrounding
tissues, and hepatic progenitor cells (hepatoblasts), which are the
cells that differentiate into the mature hepatocytes and bile duct
cells, grow inside. Hepatoblasts surrounding the portal vein form
ductal plate in the middle of embryogenesis (13–15 days of mouse
embryo) [4]. Part of ductal plate cells lose hepatocytic gene expres-
sion and form a luminal structure of bile duct. Several studies using
model mice describe that the cell-cell interaction between hepato-
blasts and mesenchymal cells around the portal vein and the signal
of soluble factors are important for differentiation from hepato-
blasts to cholangiocytes. For example, the concentration of trans-
forming growth factor β (TGFβ) in the liver during mid-fetal
embryos is high around the portal vein and low as it approaches
the inside of the liver parenchyma. The concentration gradient of
TGFβ induces differentiation of hepatoblasts [5]. In addition,
Notch-Jagged signal is important for bile duct formation process,
and Jagged-1 is the causative gene of Alagille syndrome accompa-
nied by abnormalities of the bile duct system [6, 7]. It is also known
that various transcription regulators are important for bile duct
formation and differentiation. Sal-like protein 4 is a transcription
factor that regulates the fate determination of hepatoblasts and
promotes bile duct differentiation by inducing cholangiocytic
gene expression [8]. It has also been reported that grainyhead-
like 2 regulates bile duct formation via gene expression control of
miR122 [9].
Complex three-dimensional structure of the liver is crucial for
its functions. In order to reproduce the liver functional structure
in vitro, organoid culture method has been applied to co-culture of
hepatocytes, mesenchymal cells, and endothelial cells
[10, 11]. However, these hepatic organoids do not contain the
bile duct structure, and the cytotoxicity of bile acids synthesized
by hepatocytes against cells in these organoids is a potential prob-
lem. Therefore, the construction of a novel hepatic organoid using
a co-culture system containing cholangiocytes is required. Various
groups described about the in vitro culture for induction of cho-
langiocytes. We previously established an in vitro amplification
system of hepatic progenitor cells derived from human-inducible
Induction of Cholangiocyte from Pluripotent Stem Cells 145
2 Materials
2.1 Differentiation The approval of a suitable committee is usually required for animal
of Human iPS Cells into experiments.
Hepatic Progenitor 1. Human iPS cells: HiPS-RIKEN-2F human iPS cells are estab-
Cells lished from human umbilical cord-derived fibroblasts by intro-
duction of Oct3/4, Klf4, Sox2, and c-Myc using a retroviral
system [18]. These cells are cryopreserved in liquid nitrogen
using DAP213 cryopreserve solution (see Note 1).
2. 0.1% gelatin: Gelatin derived from porcine skin is dissociated
with PBS and autoclaved.
3. Fetal bovine serum (FBS): FBS is inactivated by incubation at
55 C for 30 min.
4. Mouse embryonic fibroblast (MEF) culture medium: Dulbec-
co’s modified Eagle’s medium (DMEM) supplemented with
10% FBS and 1 penicillin–streptomycin–glutamine.
5. Phosphate-buffered saline (PBS): 10 PBS without CaCl2 and
MgCl2 is diluted with ultrapure water, autoclaved, and stored
at 4 C.
6. Mitomycin C: A 200 stock solution (2 mg/mL) is prepared
with ultrapure water.
146 Akihide Kamiya et al.
Table 1
List of antibodies used for flow cytometry experiments and immunocytochemistry for human iPS cell
differentiation and cholangiocyte differentiation
Table 2
PCR primers for detection of human gene expression
Human genes Forward primer (50 –30 ) Reverse primer (50 –30 ) Probe number
AFP tgtactgcagagataagtttagctgac tccttgtaagtggcttcttgaac 61
GRHL2 ggacagcacatacagcgaga agccccaactgaagcactc 5
HNF4α attgacaacctgttgcagga cgttggttcccatatgttcc 3
HPRT1 tgaccttgatttattttgcatacc cgagcaagacgttcagtcct 73
Keratin 7 ctgaggctgaagcctggta gggtattccggaggtcgt 64
Keratin 19 gccactactacacgaccatcc caaacttggttcggaagtcat 71
Afp α-feto protein, Grhl grainyhead like transcription factor, HNF hepatocyte nuclear factor, HPRT1 hypoxanthine
phosphoribosyltransferase 1
3 Methods
3.1 Differentiation Methods for maintaining human iPS cells and differentiation into
of Human iPS Cells into hepatic progenitor cells were already shown in “Generation and
Hepatic Progenitor in vitro expansion of hepatic progenitor cells from human iPS cells”
Cells in detail [19].
1. Embryonic day (E) 12 ICR mouse embryos were dissected,
and the head and internal organs are surgically removed. The
torso is minced and dissociated in 0.05% trypsin–EDTA. After
centrifugation and washing steps, cells were inoculated on
culture dishes with MEF culture medium. MEFs are incubated
with 0.01 mg/mL mitomycin C in MEF culture medium at
37 C for 2 h.
2. Human iPS cells are cultured and maintained on mitomycin
C-treated MEFs. The cells are cultured at 37 C in a 5% CO2
incubator. Human iPS cells are proliferated into Semi-
confluent state and used for hepatocytic differentiation.
3. Semi-confluent human iPS cells are cultured in Differentiation
medium supplemented with 100 ng/mL activin A on days
0–4 at 37 C in a 5% CO2 incubator.
4. After activin A stimulation, iPS cells were cultured in Differen-
tiation medium supplemented with 10 ng/mL basic FGF and
20 ng/mL BMP-4 on days 4–8.
5. After FGF and BMP-4 stimulation, iPS cells were cultured in
Differentiation medium supplemented with HGF on days
8–12. These cells are used for the isolation of hepatic progeni-
tor cells.
6. After 12 days of culture, cells are washed with PBS and dis-
sociated with 0.05% trypsin–EDTA (1 mL/60 mm dish). Tryp-
sinized cells are collected into a 15 mL tube and centrifuged
Induction of Cholangiocyte from Pluripotent Stem Cells 149
3.2 Culture 1. Mitomycin C-treated MEF cells (2 105 cells/well) are plated
of Hepatic Progenitor onto 0.1% gelatin-coated 12-well plates and maintained in
Cells and Induction MEF culture media the day before sorting. These cells are
of Cholangiocytic incubated for at least 12 h at 37 C in a CO2 incubator.
Differentiation Using 2. Sorted CD13highCD133+ cells (derived from Subheading 3.1)
Extracellular Matrices are cultured on MEF cells. Standard culture medium is used for
cell culture (750–1000 μL per well). Medium is replaced every
3 days.
3. Individual CD13highCD133+ cell clonally proliferates and
forms a hepatic progenitor colony on MEF for 10–14 days of
culture. The colonies are dissociated by 5 min incubation of
0.05% trypsin–EDTA at 37 C. After incubation, 1 mL of
DMEM containing 10% FBS is added, and cells are collected
by the centrifugation at 260 g for 5 min.
4. Cells are replated onto new mitomycin C-treated MEF cells.
Approximately 1/10 to 1/3 of the cells are seeded on individ-
ual well. After 7–10 days of culture, the passaged cells form new
colonies.
5. Several passaged colonies derived from CD13highCD133+ cells
are dissociated using 0.05% trypsin–EDTA and collected in
DMEM containing 10% FBS as shown above. The cell number
is counted.
6. The cells are then combined with an extracellular matrix gel
solution (1500 cells/50 μL extracellular matrix gel/well).
After mixing by pipetting, cells embedded in the gel are
cultured in 24-well culture plates (Fig. 1a, b) (see Note 4).
7. These gel solutions are incubated in 37 C CO2 incubator for
10 min to form gel. Cholangiocyte culture medium (500 μL/
well) is added, and cells are cultured for 10–12 days with
medium changes every 3 days.
8. Cystic structures are formed in the extracellular matrix gel after
14–16 days of culture (Fig. 1c).
150 Akihide Kamiya et al.
Fig. 1 Method for the culture of bile-duct like cysts in extracellular gels. (a) After mixing the cells into
extracellular matrix gel solution, place the solution in the center of the well of the culture dish. (b) After
incubation for 10 min in a CO2 incubator, the gel solution formed gel. (c) Cyst structures derived from human
iPS cells were formed
4 Notes
Fig. 2 Representative images of bile duct-like cysts derived from human iPS cells. Cysts were stained with
antibodies against AFP, Keratin 7 (CK7), β-catenin, protein kinase Cζ, and integrin α6. F-actin was stained
with phalloidin. Nuclei were counterstained with DAPI
Acknowledgments
References
1. Lemaigre FP (2009) Mechanisms of liver 12. Yanagida A, Ito K, Chikada H, Nakauchi H,
development: concepts for understanding Kamiya A (2013) An in vitro expansion system
liver disorders and design of novel therapies. for generation of human iPS cell-derived
Gastroenterology 137(1):62–79 hepatic progenitor-like cells exhibiting a bipo-
2. Douarin NM (1975) An experimental analysis tent differentiation potential. PLoS One 8(7):
of liver development. Med Biol 53(6):427–455 e67541
3. Houssaint E (1980) Differentiation of the 13. Dianat N, Dubois-Pot-Schneider H,
mouse hepatic primordium. I. An analysis of Steichen C, Desterke C, Leclerc P, Raveux A,
tissue interactions in hepatocyte differentia- Combettes L, Weber A, Corlu A, Dubart-
tion. Cell Differ 9(5):269–279 Kupperschmitt A (2014) Generation of func-
4. Lemaigre F, Zaret KS (2004) Liver develop- tional cholangiocyte-like cells from human plu-
ment update: new embryo models, cell lineage ripotent stem cells and HepaRG cells.
control, and morphogenesis. Curr Opin Genet Hepatology 60(2):700–714
Dev 14(5):582–590 14. Sampaziotis F, de Brito MC, Geti I, Bertero A,
5. Clotman F, Jacquemin P, Plumb-Rudewiez N, Hannan NR, Vallier L (2017) Directed differ-
Pierreux CE, Van der Smissen P, Dietz HC, entiation of human induced pluripotent stem
Courtoy PJ, Rousseau GG, Lemaigre FP cells into functional cholangiocyte-like cells.
(2005) Control of liver cell fate decision by a Nat Protoc 12(4):814–827
gradient of TGF beta signaling modulated by 15. Drenth JP, te Morsche RH, Smink R, Bonifa-
Onecut transcription factors. Genes Dev 19 cino JS, Jansen JB (2003) Germline mutations
(16):1849–1854 in PRKCSH are associated with autosomal
6. Lemaigre FP (2008) Notch signaling in bile dominant polycystic liver disease. Nat Genet
duct development: new insights raise new 33(3):345–347
questions. Hepatology 48(2):358–360 16. Davila S, Furu L, Gharavi AG, Tian X, Onoe T,
7. Zhang D, Gates KP, Barske L, Wang G, Lanc- Qian Q, Li A, Cai Y, Kamath PS, King BF,
man JJ, Zeng XI, Groff M, Wang K, Parsons Azurmendi PJ, Tahvanainen P, Kaariainen H,
MJ, Crump JG, Dong PDS (2017) Endoderm Hockerstedt K, Devuyst O, Pirson Y, Martin
Jagged induces liver and pancreas duct lineage RS, Lifton RP, Tahvanainen E, Torres VE,
in zebrafish. Nat Commun 8(1):769 Somlo S (2004) Mutations in SEC63 cause
autosomal dominant polycystic liver disease.
8. Oikawa T, Kamiya A, Kakinuma S, Zeniya M, Nat Genet 36(6):575–577
Nishinakamura R, Tajiri H, Nakauchi H
(2009) Sall4 regulates cell fate decision in 17. Wills ES, Te Morsche RHM, van Reeuwijk J,
fetal hepatic stem/progenitor cells. Gastroen- Horn N, Geomini I, van de Laarschot LFM,
terology 136(3):1000–1011 Mans DA, Ueffing M, Boldt K, Drenth JPH,
Roepman R (2017) Liver cyst gene knockout
9. Tanimizu N, Kobayashi S, Ichinohe N, Mitaka in cholangiocytes inhibits cilium formation and
T (2014) Downregulation of miR122 by Wnt signaling. Hum Mol Genet 26
grainyhead-like 2 restricts the hepatocytic dif- (21):4190–4202
ferentiation potential of adult liver progenitor
cells. Development 141(23):4448–4456 18. Itaba N, Wairagu PM, Aramaki N, Yasui T,
Matsumi Y, Kono Y, Phan AN, Otsu M,
10. Takebe T, Sekine K, Enomura M, Koike H, Kunisada T, Nakamura Y, Okano H, Jeong Y,
Kimura M, Ogaeri T, Zhang RR, Ueno Y, Shiota G (2014) Nuclear receptor gene alter-
Zheng YW, Koike N, Aoyama S, Adachi Y, ation in human induced pluripotent stem cells
Taniguchi H (2013) Vascularized and func- with hepatic differentiation propensity. Hepa-
tional human liver from an iPSC-derived tol Res 44(14):E408–E419
organ bud transplant. Nature 499
(7459):481–484 19. Yanagida A, Nakauchi H, Kamiya A (2016)
Generation and in vitro expansion of hepatic
11. Koui Y, Kido T, Ito T, Oyama H, Chen SW, progenitor cells from human iPS cells. Meth-
Katou Y, Shirahige K, Miyajima A (2017) An ods Mol Biol 1357:295–310
in vitro human liver model by iPSC-derived
parenchymal and non-parenchymal cells. Stem
Cell Rep 9(2):490–498
Part IV
Abstract
The hepatic functions of the hepatocytes in multicellular spheroid (MCS) are lower than those in the liver.
One of the causes is that conventional hepatic MCSs do not reproduce liver-specific microstructures such as
hepatic cord. It is necessary to design the inner structure of hepatic MCSs mimicking a structural feature of
hepatic cord to represent further hepatic functions. Here we introduce a unique method to engineer the
microarchitectures in the MCSs by formation of void spaces or filling of extracellular matrices (ECMs).
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_14, © Springer Science+Business Media, LLC, part of Springer Nature 2019
157
158 Fumiya Tao et al.
2 Materials
All liquids and plasticware used for cell culture must be sterile.
Regular culture medium is Dulbecco’s Modified Eagle’s Medium
(DMEM) supplemented with 10% fetal bovine serum (FBS) and
100 U/mL penicillin/streptomycin. Phosphate-buffered saline
(PBS) used uncontained calcium and magnesium ions.
2.1 Cell Cultures Choose the appropriated culture medium and centrifugation con-
ditions depending on the type of cells. In this chapter, we introduce
MCS culture using human hepatoma cell line Hep G2 cells (see
Note 1).
1. Polystyrene cell culture dish.
2. Trypsin-EDTA: 0.25 w/v% trypsin-1 mM ethylenediaminete-
traacetic acid (EDTA).
3. Trypan blue solution.
4. Hemocytometer.
2.2.2 Alginate Hydrogel 1. 1.5% sodium alginate solution: sodium alginate in Milli-Q.
Beads Sterilize the solution by autoclave.
2. 5% calcium chloride solution: calcium chloride in Milli-Q.
Sterilize the solution by autoclave.
3. Inkjet system: WaveBuilder, PulseInjector (25 μm-diameter
nozzle), and ink cartridge (see Note 3).
4. 1 mL syringe.
5. 40 μm cell strainer.
6. 0.22 μm syringe filter.
7. Magnetic stirrer and magnetic bar.
8. 100 mm petri dish.
9. 1000 μL micropipette.
3 Methods
3.2 Production Sodium alginate solution can turn into a gel after contacting with
of Alginate Hydrogel calcium ions. To produce alginate hydrogel beads, the inkjet system
Beads is used to discharge droplets of sodium alginate solution into
calcium chloride solution.
1. Pour 5% calcium chloride solution in a 100 mm petri dish with
a magnetic bar, and stir it at about 200 rpm.
2. Filter 1.5% sodium alginate solution using a 0.22 μm syringe
filter to remove small dusts because those may be jammed in a
PulseInjector, and fill an ink cartridge with sodium alginate
solution using a 1 mL syringe.
3. Attach the ink cartridge to a PulseInjector. Discharge sodium
alginate solution to calcium chloride solution at a voltage of
15 V and a frequency of 1000 Hz for several hours (see Note 9).
4. Collect alginate hydrogel beads with calcium chloride solution
in a 50 mL tube using a 1000 μL micropipette, and filtrate it
through a 40 μm cell strainer to remove particles with the
diameter of 40 μm or more.
5. Centrifuge for 3 min at 200 g and aspirate the supernatant.
6. Add 5 mL PBS to the tube and pipette up and down gently.
7. Repeat step 5.
8. Resuspend hydrogel beads with 5 mL regular culture medium,
and take a small aliquot to a hemocytometer.
160 Fumiya Tao et al.
3.3 Generation Figure 1 shows the schematic overview of the aggregation in the
of Normal MCS Using MC medium.
MC Medium
1. Culture Hep G2 cells in polystyrene cell culture dishes. Aspi-
rate the medium and wash in PBS.
2. Remove PBS, and add appropriate volume of trypsin-EDTA
for recovering cells to each dish. Incubate the dish at 37 C for
3 min.
3. Add 10 mL regular culture medium to stop trypsinization.
4. Collect cells into a 15 mL centrifuge tube.
Normal
medium
Cells
3% MC medium
Fig. 1 Schematic model of the cell aggregation in the 3% MC medium. Cells are shown as red color. Because
of its swelling property, the MC medium started absorbing the regular medium immediately. Finally,
suspended cells were gathered and form a MCS in the MC medium
Generation of Hepatic Tissue Structures Using Multicellular Spheroid Culture 161
3.5 Generation 1. Follow Subheading 3.3, steps 1–9 to obtain the cell
of MCS with Thin ECM suspension.
Layer (See Note 16) 2. Centrifuge for 3 min at 200 g and aspirate the supernatant.
3. Resuspend cells with medium containing diluted Matrigel
on ice.
4. Pour the MC medium into a petri dish using a positive dis-
placement pipette.
5. Inject 1 μL of cell suspension containing diluted Matrigel into
the MC medium to assemble MCSs composed of cells and
ECM (Fig. 3).
162 Fumiya Tao et al.
A B
: cells 50 µm
Fig. 2 MCSs with microchannel structures. (a) Schematic model of MCSs with
microchannel structures. (b) The hematoxylin and eosin staining with sectioning
of MCSs with microchannel structures. Red line demonstrates microchannel
structures
A B
Fig. 3 MCSs with ECM thin layer. (a) Schematic model of MCSs with ECM thin
layer. (b) Image of MCS with thin ECM layer obtained by confocal microscopy.
This figure was used FITC labeled collagen instead of Matrigel to visualize ECM.
Red color shows cells. Green color indicates FITC-labeled collagen
3.6 Isolation of MCS MCSs can be isolated from the MC medium after 1 day (see Note
from the MC Medium 17). Two isolation methods are described below. Choose an appro-
and Further Culture priate isolation method depending on the number of MCSs (see
Note 18). Isolated MCSs can be further cultured by transferring
MCSs to ultralow attachment plates (see Note 19).
3.6.1 Isolation of a Small 1. Take a small amount of the MC medium containing MCSs
Number of MCSs using a micropipette with a truncated tip, and transfer MCSs
to a 1.5 mL micro tube.
2. Add 1 mL PBS to remove the MC medium.
3. Wait for MCSs to sink to the bottom of the tube and aspirate
the supernatant.
4. Add an appropriate volume of regular culture medium.
Generation of Hepatic Tissue Structures Using Multicellular Spheroid Culture 163
3.6.2 Isolation of a Large 1. Overlay the 5 U/mL cellulase solution on the MC medium at
Number of MCSs 37 C for about 30 min to reduce the viscosity of the medium
(see Notes 20–22).
2. Transfer MCSs from the MC medium using a micropipette
with a truncated tip to an adequate tube.
3. Wash MCSs using PBS to remove the MC medium.
4. Wait until MCSs sink to the bottom of the tube and aspirate the
supernatant.
5. Add an appropriate volume of regular culture medium.
4 Notes
Acknowledgment
References
1. Laschke MW, Menger MD (2017) Life is 3D: 4. Kojima N, Takeuchi S, Sakai Y (2014) Fabrica-
boosting spheroid function for tissue engineer- tion of microchannel networks in multicellular
ing. Trends Biotechnol 35:133–144 spheroids. Sensor Actuat B Chem 198:249–254
2. Kojima N, Takeuchi S, Sakai Y (2012) Rapid 5. Motoyama W, Sayo K, Mihara H et al (2016)
aggregation of heterogeneous cells and Induction of hepatic tissues in multicellular
multiple-sized microspheres in methylcellulose spheroids composed of murine fetal hepatic
medium. Biomaterials 33:4508–4514 cells and embedded hydrogel beads. Regen
3. Kojima N, Takeuchi S, Sakai Y (2014) Engineer- Ther 3:7–10
ing of pseudoislets: effect on insulin secretion 6. Kamitori S, Ozeki Y, Kojima N (2016)
activity by cell number, cell population, and β-Galactoside-mediated tissue organization dur-
microchannel networks. Transplant Proc ing islet reconstitution. Regen Ther 3:11–14
46:1161–1165 7. Sayo K, Aoki S, Kojima N (2016) Fabrication of
bone marrow-like tissue in vitro from dispersed-
state bone marrow cells. Regen Ther 3:32–37
Chapter 15
Abstract
Construction of three-dimensional (3D) hepatic tissue structures is important for in vitro tissue engineer-
ing of the liver, because 3D culture of hepatocytes is critical for the maintenance of liver-specific functions.
Although conventional 3D culture methods are useful for constructing 3D hepatic tissue structures, the
precise control of culture microenvironments is required to construct more physiological tissues in vitro.
Recent advances in microfluidics technologies have allowed us to utilize microfluidic devices for hepatic cell
culture, which opened the door for creating more physiological 3D culture models of the liver. Here, we
describe the method for the construction of hepatic tissue structures using a microfluidic device which has a
3D gel region with adjacent microchannels. Primary rat hepatocytes are seeded into a microchannel in a
microfluidic device. The cells are then cultured in interstitial flow conditions, which leads to the construc-
tion of 3D tissue structures.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_15, © Springer Science+Business Media, LLC, part of Springer Nature 2019
167
168 Ryo Sudo
2 Materials
2.1 Fabrication 1. SYLGARD® 184 silicone elastomer base and the curing agent.
of Microfluidic Devices 2. Plastic cup.
3. Glass rod.
4. Desiccator connected with a vacuum pump.
5. Prepare an SU-8 mold for desired patterning of microchannels
using standard photolithography technique (see ref. [4]).
6. Oven.
7. Biopsy punch.
8. Scotch tape.
9. Plasma cleaner.
10. Culture dish.
3 Methods
3.1 Fabrication 1. Prepare SYLGARD® 184 silicone elastomer base and the cur-
of Microfluidic Devices ing agent in a 10:1 weight ratio in a disposable plastic cup.
2. Mix the solution, which is poly-dimethyl siloxane (PDMS)
prepolymer, by a glass rod.
3. Place the PDMS prepolymer in a desiccator connected with a
vacuum pump for removing air bubbles in the PDMS
prepolymer.
4. Pour the PDMS prepolymer onto an SU-8 mold to the desired
thickness (e.g., 7 mm), and place them in a desiccator again for
removing air bubbles completely.
5. Place the mold in an oven at 65 C for >3 h to cure the
degassed prepolymer (see Note 3).
6. Cut out and detach the cured PDMS from the mold using a
scalpel.
7. Trim the cured PDMS, and punch the trimmed PDMS devices
to form inlets and outlets of microchannels using a biopsy
punch.
8. Remove small particles from the surface of the PDMS devices
using Scotch tape (see Note 4).
9. Place the PDMS devices in a glass beaker with deionized water
and autoclave them.
10. Place the PDMS devices in a clean container (e.g., empty
micropipette tip box), and autoclave them.
11. Dry the autoclaved PDMS devices in an oven at 65 C.
12. Place the PDMS devices, and autoclaved coverslips in a plasma
cleaner for plasma treatment in air (see Note 5).
13. Bond a PDMS device and a coverslip immediately after air
plasma treatment (Fig. 1).
14. Place the bonded PDMS device and coverslip on a flat surface,
and gently press the PDMS device to enhance adhesion
between the PDMS device and a coverslip.
170 Ryo Sudo
Fig. 1 Plasma bonding. A PDMS device and a coverslip are bonded after air
plasma exposure, resulting in the fabrication of a microfluidic device
3.2 Surface 1. Fill channels of the microfluidic devices with PDL solution for
Treatment surface coating (see Note 6).
of Microchannels 2. Place the devices in a 5% CO2 incubator at 37 C for >4 h.
and Gel Formation
3. Remove the PDL solution and rinse the microchannels with
in a Microfluidic deionized water twice.
Device
4. Aspirate the microchannels completely and dry them in an oven
at 65 C overnight (see Note 7).
5. Prepare collagen solution on ice.
6. Slowly inject the collagen solution from an outlet of a gel
channel to fill out the gel region using a 10 μL micropipette
(Fig. 2a, b).
7. Place the devices in a humid chamber, and incubate them for
30 min in a 5% CO2 incubator at 37 C to allow gelation (see
Note 8).
8. Gently fill the microchannels with warmed culture medium
(Fig. 2c), and place the device in a culture dish.
Fig. 2 Schematic images of a microfluidic device. (a) A microfluidic device before the injection of collagen
solution. (b) A microfluidic device filled with collagen gel. Collagen solution is injected from an outlet of a gel
channel to fill out the gel region. Gelation of the collagen solution results in the formation of two parallel
microchannels for culture medium. (c) A microfluidic device filled with both collagen gel and culture medium.
Microchannels are filled with culture medium after collagen solution is gelled
Fig. 3 Schematic images of a microfluidic device during hepatocyte seeding. (a) Cell suspension is added to
the outlet of a microchannel. (b, c) A microchannel is filled with cell suspension, and the device is tipped on its
side and maintained in the incubator to allow cells to attach and stack on the sidewall of collagen gel
Fig. 4 Reconstruction of hepatic tissue structures in a microfluidic device. (a) A microfluidic device connected
with medium reservoirs. (b) A schematic image of the microchannels with medium reservoirs. A pressure
difference of 5 mmH2O between two microchannels generates interstitial flow across the gel region. (c)
Hepatocytes are stacked and construct multicellular aggregates under interstitial flow. (d) Corresponding
phase-contrast images during hepatocyte tissue formation. Hepatocytes reconstruct hepatic tissue structures
in interstitial flow conditions within 4 days of culture
3.4 Hepatocyte 1. Gently insert medium reservoirs into the microchannel outlets
Culture in Flow (Fig. 4a).
Conditions 2. Add culture medium to the reservoirs to make a pressure
difference of 5 mmH2O between two medium channels across
the gel region (Fig. 4b) (see Note 10).
3. Change culture medium daily (see Note 11).
4. The other cell type (e.g., endothelial cells) can be added to the
other channel for coculture (Fig. 5) (see Note 12).
4 Notes
Fig. 5 An example of coculture in a microfluidic device. After hepatocytes construct 3D tissue structures, the
other cell type (e.g., endothelial cells) can be added to the other channel
Acknowledgment
References
1. Sudo R (2014) Multiscale tissue engineering for 5. Sudo R, Chung S, Zervantonakis IK,
liver reconstruction. Organogenesis Vickerman V, Toshimitsu Y, Griffith LG,
10:216–224 Kamm RD (2009) Transport-mediated angio-
2. Chung S, Sudo R, Vickerman V, Zervantonakis genesis in 3D epithelial coculture. FASEB J
IK, Kamm RD (2010) Microfluidic platforms 23:2155–2164
for studies of angiogenesis, cell migration, and 6. Yamamura N, Sudo R, Ikeda M, Tanishita K
cell-cell interactions. Ann Biomed Eng (2007) Effects of the mechanical properties of
38:1164–1177 collagen gel on the in vitro formation of micro-
3. Zervantonakis IK, Kothapalli CR, Chung S, vessel networks by endothelial cells. Tissue Eng
Sudo R, Kamm RD (2011) Microfluidic devices 13:1443–1453
for studying heterotypic cell-cell interactions 7. Chung S, Sudo R, Mack PJ, Wan CR,
and tissue specimen cultures under controlled Vickerman V, Kamm RD (2009) Cell migration
microenvironments. Biomicrofluidics 5:13406 into scaffolds under co-culture conditions in a
4. Shin Y, Han S, Jeon JS, Yamamoto K, Zervanto- microfluidic platform. Lab Chip 9:269–275
nakis IK, Sudo R, Kamm RD, Chung S (2012) 8. Chung S, Sudo R, Zervantonakis IK,
Microfluidic assay for simultaneous culture of Rimchala T, Kamm RD (2009) Surface treat-
multiple cell types on surfaces or within hydro- ment induced three dimensional capillary mor-
gels. Nat Protoc 7:1247–1259 phogenesis in a microfluidic platform. Adv
Mater 21:4863–4867
Chapter 16
Abstract
Incorporation of bile drainage system into engineered liver tissue is an important issue to advance liver
regenerative medicine. Our group reported that three-dimensional (3D) coculture of fetal liver cells (FLCs)
and adult rat biliary epithelial cells (BECs) allows reconstruction of hepatic spheroids that possess bile
ductular structures. In this chapter, we describe the detailed protocol to isolate FLCs and BECs and to
generate the spheroids with bile drainage system using these two types of primary cells.
Key words Biliary epithelial cell, Fetal liver cell, Bile ducts, Hetero-spheroid, 3D culture, Tissue
engineering
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_16, © Springer Science+Business Media, LLC, part of Springer Nature 2019
175
176 Takeshi Katsuda et al.
2 Materials
3 Methods
3.1 Extraction of the This procedure is performed in parallel with fetal liver extraction
Biliary Trees and (see Subheading 3.2) using one pregnant rat.
Isolation of BECs 1. Anaesthetize a pregnant rat by inhalation of isoflurane vapor.
Place a paper towel cushion on a foam board or cork board, and
lay the rat on the cushion. Sterilize the animal by wiping its fur
with 70% EtOH. Open the abdomen using surgical scissors.
2. Start drip of pre-perfusion buffer at 25–30 mL/min. Half-cut
the portal vein at the location approximately 1.5 cm from the
bifurcation of the portal vein, and insert the cannula.
3. Immediately after confirming that the color of the liver changes
from reddish brown to yellowish, cut the inferior vena cava.
Perfuse approximately 450 mL pre-perfusion buffer (approxi-
mately 15 min).
4. Pause the pump, transfer the inlet tube to the collagenase
bottle, and resume perfusion. Perfuse approximately
300–350 mL collagenase (approximately 10 min).
5. Stop the pump, and remove the cannula from the portal vein.
6. Peel the liver capsule, and gently mash the parenchyma with
round-edged tweezers to release the parenchymal cells from
the tree structures. Gently flush away the digested parenchymal
tissue with E-MEM until white tree structures are visible
(Fig. 1a–e).
7. Resect the tree structures, and place them in bile duct wash
medium. At this moment, the tree structures still hold paren-
chymal cells as noticed by their brownish appearance.
8. Purify the tree structure ex vivo by further removing the
remaining parenchyma until most brownish-colored parts are
removed (Fig. 1f). Gently pressing the tissue with round-edged
tweezers helps releasing of parenchymal cells from the tree
structures.
9. Collect the purified tree fragments into multiple 1.5 mL tubes
so that each tube contain 150–300 μL of fragments (usually
divided into two to four tubes) (see Note 1), and mince them in
the 1.5 mL tubes with surgical scissors into small fragments
(approximately <0.5 mm). From this step, work aseptically in a
clean bench or a safety cabinet.
10. Suspend the minced fragments in 10 mL BEC digestion
medium, and divide it to two 50 mL tubes.
Generation of Hepatic Organoids with Biliary Structures 179
Fig. 1 Graphical schematic of extraction of intrahepatic bile duct trees. (a) Just after switching from
pre-perfusion buffer to collagenase solution and (b) after completion of collagenase perfusion. Note that the
liver is loosened and the subcapsular tissue is pulpy compared to (a). (c) By peeling the liver capsule and
gently mashing the parenchyma with round-edged tweezers, parenchymal tissue can be liberated from the
tree structure. (d) Flushing the parenchymal tissue with E-MEM by pipetting. Lifting up the rat’s front paws will
help this procedure. (e) White intrahepatic biliary trees can be visible after removal of the parenchymal tissue.
(f) The purified tree structures after mechanically removing the remaining parenchyma using tweezers ex vivo
3.2 Extraction of While perfusing the adult liver, proceed to fetal liver extraction in
Fetal Livers and parallel.
Purification of FLCs
1. Remove the uterus containing fetuses from the body of the
mother rat, and transfer them to a 10 cm dish filled with PBS.
2. Extract fetuses from the uterus using surgical scissors and
tweezers, and transfer them to a new 10 cm dish filled
with PBS.
3. Extract the livers from the fetuses using tweezers, and transfer
the fetal livers to a new 10 cm dish filled with PBS.
4. Remove the white tissue surrounding the red livers as
completely as possible using tweezers. Divide the purified livers
into 1.5 mL tubes so that each tube contains 150–300 μL (two
to five tubes, dependent on the number of fetuses) (see Note
1), and mince them with surgical scissors into small fragments
(approximately <0.5 mm).
5. Transfer the minced tissue into a 15 mL tube, add PBS to
adjust the total volume to 12 mL, and stand the tube until
the minced tissues fall to the bottom (3–5 min) (see Note 3).
From this step, work aseptically in a clean bench or a safety
cabinet. Carefully discard the supernatant using a pipette, and
add 12 mL PBS.
6. Repeat the wash process in step 5 until the color of the super-
natant changes from red to clear (usually two to three time
repeats are sufficient).
7. Suspend the minced tissue in 2.5 mL/tube PBS, and add
2.5 mL of 0.1% collagenase solution (final concentration:
0.05%).
8. Shake the tube at 37 C in a shaking incubator for 10 min.
Make sure that the content is homogeneously stirred.
9. Let the tube stand for approximately 10 s, and retrieve the
supernatant into a new tube while leaving the remaining frag-
ments (see Note 4).
10. Centrifuge the tube at 200 g for 2 min at 4 C.
Generation of Hepatic Organoids with Biliary Structures 181
Fig. 2 Morphology of FLCs and BECs. (a, b) Phase-contrast images of E17.5 FLCs on day 2 of culture. (c, d)
Phase-contrast images of BECs isolated from the adult rat liver on day 3 of culture. This figure is reproduced
from ref. [8]
4 Notes
Fig. 3 Time course of hetero-spheroid formation. Phase-contrast images of hetero-spheroids with the
composition of “FLC:BEC ¼ 1:1” and “FLC:BEC ¼ 1:3” hetero-spheroids on day 1 (a, b), day 2 (c, d), and
day 3 (e, f). This figure is reproduced from ref. [8]
3. If the minced tissue volume is larger than 500 μL, aliquot the
suspension to multiple tubes so that each tube contains less than
500 μL.
4. White tissue will be visible after digestion. Do not let the tube
(s) stand too long to avoid loss of FLCs.
5. At the cell density of 2 105 cells/well, we observed necrosis at
the central area of the spheroids, most likely because each spher-
oid contains too much cells. We suppose that a lower cell density
184 Takeshi Katsuda et al.
Fig. 4 Histological analyses of hetero-spheroids on day 3. (a, b) HE staining (nuclei, blue; cytoplasm,
red-purple). Arrows and arrowheads indicate duct- and cyst-like structures, respectively. (c, d) IHC of CK19
(brown). Nuclei were counterstained with hematoxylin (blue). Arrowheads indicate continuous ductular net-
works formed by CK19 (+) BECs. (e, f) IHC of CK18 (brown). Nuclei were counterstained with hematoxylin
(blue). Scale bar: 50 μm. CK cytokeratin, HE hematoxylin-eosin, IHC immunohistochemistry. This figure is
reproduced from ref. [8]
Acknowledgments
References
1. Ohashi K, Yokoyama T, Yamato M et al (2007) 3D bile duct tissue from human pluripotent
Engineering functional two- and three- stem cell-derived spheroids. Stem Cell Rev Rep
dimensional liver systems in vivo using hepatic 12:500–508. https://doi.org/10.1007/
tissue sheets. Nat Med 13:880–885. https:// s12015-016-9657-5
doi.org/10.1038/nm1576 6. Sampaziotis F, De Brito MC, Madrigal P et al
2. Takebe T, Sekine K, Enomura M et al (2013) (2015) Cholangiocytes derived from human
Vascularized and functional human liver from an induced pluripotent stem cells for disease mod-
iPSC-derived organ bud transplant. Nature eling and drug validation. Nat Biotechnol
499:481–484. https://doi.org/10.1038/ 33:845–852. https://doi.org/10.1038/nbt.
nature12271 3275
3. Sakai Y, Yamanouchi K, Ohashi K et al (2015) 7. Dianat N, Dubois-Pot-Schneider H, Steichen C
Vascularized subcutaneous human liver tissue et al (2014) Generation of functional
from engineered hepatocyte/fibroblast sheets cholangiocyte-like cells from human pluripotent
in mice. Biomaterials 65:66–75. https://doi. stem cells and HepaRG cells. Hepatology
org/10.1016/j.biomaterials.2015.06.046 60:700–714. https://doi.org/10.1002/hep.
4. Jiang J, Kojima N, Guo L et al (2004) Efficacy of 27165
engineered liver tissue based on poly-L-lactic 8. Katsuda T, Kojima N, Ochiya T, Sakai Y (2013)
acid scaffolds and fetal mouse liver cells cultured Biliary epithelial cells play an essential role in the
with oncostatin M, nicotinamide, and dimethyl reconstruction of hepatic tissue with a functional
sulfoxide. Tissue Eng 10:1577–1586. https:// bile ductular network. Tissue Eng Part A
doi.org/10.1089/ten.2004.10.1577 19:2402–2411. https://doi.org/10.1089/ten.
5. Tian L, Deshmukh A, Ye Z, Jang YY (2016) TEA.2013.0021
Efficient and controlled generation of 2D and
Chapter 17
Abstract
It has been demonstrated that the liver remarkably alter its tissue structures during regeneration, and
various types of liver stem or progenitor cells locating in specific areas in the liver tissue contribute to
regeneration. Therefore, it is important to analyze the dynamic rearrangement of the liver tissue structures
in 3D for better understanding the process and mechanism of liver regeneration. Here we describe a
macroscopic analysis method to visualize the whole 3D structure of the vasculatures and the biliary tree,
which are dynamically remodeled during regeneration, and a microscopic analysis method to visualize
detailed structures at the cellular level, which can compensate what cannot be detected in the macroscopic
analysis.
Key words Liver regeneration, Liver pathology, 3D tissue structure, 3D imaging, Ductular reaction,
Biliary tree
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_17, © Springer Science+Business Media, LLC, part of Springer Nature 2019
187
188 Kota Kaneko
Zonation
Fig. 1 The macroscopic and the microscopic structure of the liver. Bile ducts (green), portal vein (magenta),
central vein (blue), sinusoids (red), hepatic artery (pink), neurons (yellow), and other immune and mesenchy-
mal populations (orange, dark blue, and purple) are shown
the central veins, which are the landmarks to tell the “zonation.”
Therefore the whole liver structure can roughly be regarded as the
collections of the lobules defined by vasculatures (Fig. 1). Around
the portal vein exist bile ducts, hepatic artery, and the peripheral
nerve systems. Importantly, bile ducts dynamically change the
structure during liver injury and regeneration, which is called duct-
ular reaction [5]. Recent studies have shown that bile ducts can
extend their branches as far as to the pericentral area in response to
injury, independently of the vascular structure [6–8]. Together
with the classic view of the continuous epithelial structure of hepa-
tocytes and biliary epithelial cells [1] (Fig. 2), such remodeling of
the bile duct structure needs to be taken into consideration when
assessing any spatial relationships. We recommend using portal
veins, central veins, and bile ducts as basic landmarks to determine
the locations of objects in the liver tissue.
There are many different methods to analyze the 3D architec-
ture of the liver. When we select a particular method, it is necessary
to consider the actual convenience for using the method, technical
limitations of each method (maximum scale, detectable cell types,
compatibility with fluorescent proteins such as GFP, etc.), and the
resolution. Among them, resolution should be particularly impor-
tant to visualize the actual tissue structures. In fact, the analysis of
the biliary tree architecture with an unprecedented high-resolution
analysis revealed the actual heterogeneous structure (Fig. 3) and
the dynamic rearrangements of the bile ducts [6, 9]. In contrast, as
shown in Fig. 3, it is hard to know the correct tissue structures and
their changes by low-resolution analyses. Since only large ducts are
observed in a low-resolution analysis, bile ducts are mostly depicted
in textbooks as a single tube along the vein. Thus, high-resolution
analyses do not only lead to new discoveries but are also crucial for
getting the correct conclusions.
In this chapter, we will introduce macroscopic and microscopic
3D analysis methods, which should preferably be combined
together to cover most of our purposes for analyzing the tissue
3D Analysis of the Liver Tissue Structure 189
Portal Central
vein vein
Fig. 2 The continuous epithelial tissue structure of the liver. The epithelial tissue
of the liver consists of hepatocytes and biliary epithelial cells, which are lined up
as a continuous structure, connected at the canal of Hering. Bile juice is
produced by hepatocytes and drained into bile ducts through bile canaliculi.
Hepatocytes near the bile ducts are known to have different characteristics from
those located away from the bile duct
High-resolution Low-resolution
Fig. 3 Structure of the biliary tree (green) with the portal vein (pink). Bile ducts have heterogeneous structure
consisting of thick tubes running along with the portal vein and fine tubes branching and wrapping around the
portal vein. High-resolution analysis clearly shows the heterogeneous network (left panel), whereas the fine
tubular network is hardly visualized with low-resolution analysis (right panel). Therefore, they possibly lead to
different conclusions
PV
CV
PV
PV CV
CV
Fig. 4 Macroscopic observation of the liver tissue structure by ink injection. Bile
ducts (black) are running along and wrapping around the portal vein. Portal veins
(PV) and central veins (CV) can be seen with the red blood in them
2 Materials
Fig. 5 Microscopic 3D reconstitution of the liver tissue structure. This image shows the biliary epithelial cells
(white) in the CDE diet model liver. Veins (pink) can be reconstituted by inverting the autofluorescence of the
tissue. Since this method is based on immunostaining, it can show subcellular structures of any types of liver
cells as long as antibodies are available
Stretch
break
Connect
butterfly needle
Syringe
Cut
“Capillary”
Fig. 6 Glass capillary for injection. Ink can be loaded into the capillary and
injected into the bile duct by manipulating the syringe
3 Methods
Portal vein
intstine
Capillary
Fig. 7 Procedures of the ink injection into the biliary tree. (a) Extrahepatic bile duct (arrowheads) is running
from the liver to the intestine on the side of the portal vein. The bile duct has a bulge (arrow and dashed line) at
the junction with the intestine. (b) Cutting the bulge at the junction. (c) Insertion of the capillary into the bile
duct tube
12. Cover the section with a glass cover slip (see Note 14), and
acquire images with a confocal microscope.
4 Notes
Acknowledgments
References
1. Miyajima A, Tanaka M, Itoh T (2014) Stem/ reactions in human livers. Hepatology (Balti-
progenitor cells in liver development, homeo- more, MD) 39(6):1739–1745. https://doi.
stasis, regeneration, and reprogramming. Cell org/10.1002/hep.20130
Stem Cell 14(5):561–574. https://doi.org/ 6. Kaneko K, Kamimoto K, Miyajima A, Itoh T
10.1016/j.stem.2014.04.010 (2015) Adaptive remodeling of the biliary
2. Font-Burgada J, Shalapour S, Ramaswamy S, architecture underlies liver homeostasis. Hepa-
Hsueh B, Rossell D, Umemura A, Taniguchi K, tology (Baltimore, MD) 61(6):2056–2066.
Nakagawa H, Valasek Mark A, Ye L, Kopp https://doi.org/10.1002/hep.27685
Janel L, Sander M, Carter H, Deisseroth K, 7. Kamimoto K, Kaneko K, Kok CYY, Okada H,
Verma Inder M, Karin M (2015) Hybrid peri- Miyajima A, Itoh T (2016) Heterogeneity and
portal hepatocytes regenerate the injured liver stochastic growth regulation of biliary epithe-
without giving rise to cancer. Cell 162 lial cells dictate dynamic epithelial tissue remo-
(4):766–779. https://doi.org/10.1016/j.cell. deling. eLife 5:e15034
2015.07.026 8. Nagahama Y, Sone M, Chen X, Okada Y,
3. Tanimizu N, Ichinohe N, Yamamoto M, Yamamoto M, Xin B, Matsuo Y, Komatsu M,
Akiyama H, Nishikawa Y, Mitaka T (2017) Suzuki A, Enomoto K, Nishikawa
Progressive induction of hepatocyte progenitor Y. Contributions of hepatocytes and bile duct-
cells in chronically injured liver. Sci Rep ular cells in ductular reactions and remodeling
7:39990. https://doi.org/10.1038/ of the biliary system after chronic liver injury.
srep39990 Am J Pathol 184(11):3001–3012. https://doi.
4. Wang B, Zhao L, Fish M, Logan CY, Nusse R org/10.1016/j.ajpath.2014.07.005
(2015) Self-renewing diploid Axin2(+) cells 9. Tanimizu N, Kaneko K, Itoh T, Ichinohe N,
fuel homeostatic renewal of the liver. Nature Ishii M, Mizuguchi T, Hirata K, Miyajima A,
524(7564):180–185. https://doi.org/10. Mitaka T (2016) Intrahepatic bile ducts are
1038/nature14863 developed through formation of homogeneous
5. Roskams TA, Theise ND, Balabaud C, continuous luminal network and its dynamic
Bhagat G, Bhathal PS, Bioulac-Sage P, Brunt rearrangement in mice. Hepatology (Balti-
EM, Crawford JM, Crosby HA, Desmet V, more, MD) 64(1):175–188. https://doi.org/
Finegold MJ, Geller SA, Gouw AS, 10.1002/hep.28521
Hytiroglou P, Knisely AS, Kojiro M, Lefko- 10. Ke M-T, Fujimoto S, Imai T (2013) SeeDB: a
witch JH, Nakanuma Y, Olynyk JK, Park YN, simple and morphology-preserving optical
Portmann B, Saxena R, Scheuer PJ, Strain AJ, clearing agent for neuronal circuit reconstruc-
Thung SN, Wanless IR, West AB (2004) tion. Nat Neurosci 16:1154. https://doi.org/
Nomenclature of the finer branches of the bili- 10.1038/nn.3447
ary tree: canals, ductules, and ductular
Part V
Abstract
Liver cirrhosis is the end stage of chronic liver disease, and the only radical treatment for decompensated
liver cirrhosis is still liver transplantation. Development of effective regenerative therapy for liver cirrhosis is
an urgent task. Before human clinical trials can be considered, the safety and efficacy of any planned
protocol must be confirmed in medium-to-large animals. Therefore, we have developed a novel canine
liver fibrosis model for proof of concept (POC) studies.
Key words Liver cirrhosis, Canine model, Mesenchymal stem cells, Carbon tetrachloride
1 Introduction
Liver cirrhosis (LC) is the end stage of chronic liver disease caused
by viral hepatitis, excess consumption of alcohol, obesity, and so
on. Although the liver is an organ with a high regenerative capacity,
chronic inflammation results in the development of cirrhosis, and
dense fibrosis ultimately halts hepatocyte proliferation. Currently,
the only radical treatment available for decompensated LC is liver
transplantation. However, liver transplantation has its own issues,
including donor shortage, immunorejection, and medical cost. To
overcome the issues related to liver transplantation, we aimed to
develop a liver regeneration therapy for decompensated LC patients
using autologous bone marrow cells (BMCs) [1]. In a murine LC
model created by repeated carbon tetrachloride (CCl4) treatment
[2], we previously confirmed that syngeneic non-cultured whole
BMCs that were infused through the tail vein had repopulated the
damaged liver and produced matrix metalloproteinases (MMPs),
ultimately reducing liver fibrosis [3]. Based on these evidences, we
conducted a clinical study and reported the clinical efficacy and
safety of “autologous bone marrow cell infusion therapy (ABMi
therapy)” as liver regeneration therapy for decompensated LC
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_18, © Springer Science+Business Media, LLC, part of Springer Nature 2019
201
202 Taro Takami et al.
2 Materials
2.1 Gastric Catheter 1. Beagles (1–2 years old): These canines were housed in our
Placement animal facility and treated in accordance with the university’s
animal care guidelines. This study was approved by our Institu-
tional Ethics Committee (approval number; 21-033).
2. Infusion port: A P-U Celsite port (Toray Medical Co. Ltd.,
Tokyo) consisting of a catheter coated with a heparinized
hydrophilic material and a Celsite port.
3. 18 G Teflon IV catheter (6-French P-U catheter).
4. Anesthesia machine: For delivering positive pressure
ventilation.
5. Propofol 1% injection: The loading dose of propofol is about
7 mg/kg body weight (BW).
6. Lepetan: Intravenous Lepetan (buprenorphine) is administered
at a dose of 10 μg/kg BW to provide effective pain relief.
7. Isoflurane: Anesthesia is maintained with isoflurane in oxygen.
The end-tidal isoflurane concentration is monitored and main-
tained between 1.4 and 2.8%.
8. Endobronchial tube for general anesthesia.
9. Chlorhexidine acetate.
10. 2% chlorhexidine gluconate in 70% isopropyl alcohol.
11. #21 scalpel.
12. Right-angled forceps.
13. Babcock forceps.
Canine Liver Fibrosis Model 203
2.6 Sirius Red 1. Sirius red solution (1.3% aqueous picric acid/0.1% direct red
Staining 80).
204 Taro Takami et al.
3 Methods
3.1 Gastric Catheter 1. Under general anesthesia, the canine is restrained in the right
Placement lateral decubitus position and shaved over a wide area from the
posterior margin of the scapula to the anterior margin of the
femur and from the linea alba (midline) to around 2 cm left of
the vertebral body.
2. Wearing sterile gloves, the operator picks up a chlorhexidine
acetate scrub in a piece of gauze and uses it to scrub the
operating field, foaming it thoroughly, and disinfects the area
with cotton soaked in 2% chlorhexidine gluconate in 70%
isopropyl alcohol. The operating field is then sprayed with
povidone-iodine and allowed to dry naturally, after which it is
draped.
3. An approximately 3-cm-long skin incision is made with a #21
scalpel at a point 2 cm caudal to the last rib, blunt dissection of
the subcutaneous tissue is performed using right-angled for-
ceps, and the abdominal external oblique muscle is grasped
with Babcock forceps and exposed. Right-angled forceps are
used to divide the muscles bluntly, and the abdominal internal
oblique muscle is similarly divided to approach the abdominal
cavity.
4. An assistant operates the upper gastrointestinal endoscope and
inflates the stomach, assisting the operator by making it easier
to grasp the serosal side of the stomach with Babcock forceps.
Part of the stomach is exposed extracorporeally and the stom-
ach is fixed to the abdominal wall by placing single ligatures of
suture material (3-0 PDS) at four places on the gastric wall;
then, the abdominal external oblique muscle around the loca-
tion is grasped with the Babcock forceps.
5. The catheter is inserted into the stomach using a sheath, guide-
wire, and dilator (Fig. 1a). Endoscopy is used to confirm that
the catheter tip does not extend past the cardiac orifice into the
esophagus (Fig. 1b).
6. An approximately 3-cm-long skin incision is made slightly to
the left of the eighth to tenth thoracic vertebrae, and blunt
dissection of the subcutaneous tissues is performed to create a
bed for the port (Fig. 1c). An approximately 1-cm-long skin
incision is made at a point exactly midway between the sites of
port placement and catheter insertion, and right-angled for-
ceps are passed through this incision to grasp the catheter and
guide it under the skin to the port placement site.
Canine Liver Fibrosis Model 205
Fig. 1 Canine liver fibrosis model. (a) Infusion port: A P-U Celsite port. (b) The catheter tip in the stomach was
confirmed using endoscopy. (c) The port was placed in the back of the neck. Arrow shows the place of the port
3.3 Induction of Liver 1. CCl4 is diluted 1:1 in corn oil before its intragastric injection
Fibrosis through the catheter.
2. A winged Surecan port needle is inserted into the subcutaneous
port, and the mixture of carbon tetrachloride and corn oil is
injected into the stomach (see Note 1).
3. Injections are repeated for 10 weeks using the implanted cath-
eter (high-dose period, 1.0 mL/kg BW once a week and
0.5 mL/kg BW once a week for 6 weeks; low-dose period,
0.25 mL/kg BW twice a week for 4 weeks) to induce liver
fibrosis [9]. After the infusion of BMSCs, low-dose CCl4 is
further administrated for 4 weeks.
4. The CCl4 injections are made on the first and fourth day of
each week.
20 25
15 20
15
10
10
5
5
0 0
Fig. 2 Kinetics of liver fibrosis and half-life time of indocyanine green (ICG) testing after the infusion of cultured
autologous bone marrow derived-mesenchymal stem cells (BMSCs) in the canine liver fibrosis model. (a, c)
The amount of fibrosis, as seen with Sirius red staining, was increased in the control group (after continuous
carbon tetrachloride treatment without the autologous BMSC infusion). (b, c) A decreased level of fibrosis was
seen at 4 weeks after the autologous BMSC infusion, even though carbon tetrachloride treatment was also
continued. (d) The half-life of ICG was significantly shorter at 4 weeks in the BMSC-infused group compared
with 0 weeks in the same group and 4 weeks in the control group. Every value and bar shown indicates
mean SD. This figure was reproduced with permission from ref. [9]
4 Notes
Acknowledgments
References
1. Takami T, Terai S, Sakaida I (2012) Stem cell 4. Terai S, Ishikawa T, Omori K, Aoyama K,
therapy in chronic liver disease. Curr Opin Gas- Marumoto Y, Urata Y, Yokoyama Y, Uchida K,
troenterol 28(3):203–208. https://doi.org/10. Yamasaki T, Fujii Y, Okita K, Sakaida I (2006)
1097/MOG.0b013e3283521d6a Improved liver function in patients with liver cir-
2. Terai S, Sakaida I, Yamamoto N, Omori K, rhosis after autologous bone marrow cell infusion
Watanabe T, Ohata S, Katada T, Miyamoto K, therapy. Stem Cells 24(10):2292–2298. https://
Shinoda K, Nishina H, Okita K (2003) An doi.org/10.1634/stemcells.2005-0542. pii:
in vivo model for monitoring trans- 2005-0542
differentiation of bone marrow cells into func- 5. Kim JK, Park YN, Kim JS, Park MS, Paik YH, Seok
tional hepatocytes. J Biochem 134(4):551–558 JY, Chung YE, Kim HO, Kim KS, Ahn SH, Kim
3. Sakaida I, Terai S, Yamamoto N, Aoyama K, do Y, Kim MJ, Lee KS, Chon CY, Kim SJ, Terai S,
Ishikawa T, Nishina H, Okita K (2004) Trans- Sakaida I, Han KH (2010) Autologous bone mar-
plantation of bone marrow cells reduces CCl4- row infusion activates the progenitor cell compart-
induced liver fibrosis in mice. Hepatology 40 ment in patients with advanced liver cirrhosis. Cell
(6):1304–1311. https://doi.org/10.1002/ Transplant 19(10):1237–1246. https://doi.org/
hep.20452 10.3727/096368910X506863
Canine Liver Fibrosis Model 209
6. Kim JK, Kim SJ, Kim Y, Chung YE, Park YN, Kim Terai S, Sakaida I (2014) Canine mesenchymal
HO, Kim JS, Park MS, Sakaida I, Kim DY, Lee JI, stem cells show antioxidant properties against
Ahn SH, Lee KS, Han KH (2017) Long-term thioacetamide-induced liver injury in vitro and
follow-up of patients after autologous bone mar- in vivo. Hepatol Res 44(10):E206–E217.
row cell infusion for decompensated liver cirrho- https://doi.org/10.1111/hepr.12204
sis. Cell Transplant 26(6):1059–1066. https:// 9. Matsuda T, Takami T, Sasaki R, Nishimura T,
doi.org/10.3727/096368917X694778 Aibe Y, Paredes BD, Quintanilha LF,
7. Tanimoto H, Terai S, Taro T, Murata Y, Matsumoto T, Ishikawa T, Yamamoto N,
Fujisawa K, Yamamoto N, Sakaida I (2013) Tani K, Terai S, Taura Y, Sakaida I (2017) A
Improvement of liver fibrosis by infusion of canine liver fibrosis model to develop a therapy
cultured cells derived from human bone marrow. for liver cirrhosis using cultured bone marrow-
Cell Tissue Res 354(3):717–728. https://doi. derived cells. Hepatol Commun 1(7):691–703.
org/10.1007/s00441-013-1727-2 https://doi.org/10.1002/hep4.1071
8. Quintanilha LF, Takami T, Hirose Y, Fujisawa K,
Murata Y, Yamamoto N, Goldenberg RC,
Chapter 19
Abstract
Hepatic progenitor cells are defined as cells exhibiting potency for active proliferation and capacity for
bipotential differentiation into hepatocytes and cholangiocytes. To prove the capacity of target cells for
terminal differentiation and reconstitution of organs, cell transplantation models have been widely used in
previous studies, including those involving the liver. Here we describe a protocol for transplantation of
hepatic progenitor cells using retrorsine pretreatment and partial hepatectomy. This transplantation assay
reveals the potential for reconstitution of hepatocytes in recipient livers by primary hepatic progenitor cells.
Donor cells are detected as a colony composed of 5–10 mature hepatocytes.
Key words Hepatoblasts, Hepatocytes, Intrasplenic injection, Partial hepatectomy, Retrorsine, Trans-
plantation, Differentiation
1 Introduction
Stem cells are defined as cells exhibiting both the capacity for self-
renewal and the ability to undergo multilineage differentiation
[1]. Somatic stem cells in the liver (i.e., hepatic stem cells) are
defined as cells capable of self-renewal and terminal differentiation
into hepatocytes and cholangiocytes. Liver lobules are constituted
of a variety of cells, including hepatocytes, cholangiocytes, hepatic
stellate cells (Ito cells), Kupffer cells, sinusoidal endothelial cells,
and pit cells, among others. Both hepatocytes and cholangiocytes
are derived from hepatoblasts in the fetal liver [2]. Hepatic progen-
itor cells are also capable of bipotential differentiation and active
proliferation but lack the capacity for self-renewal [3]. Hepatic
stem/progenitor cells can be enriched in mouse mid-gestational
fetal hepatic cell fractions based on several cell surface markers,
including Dlk [4], E-cadherin [5], Liv2 [6], and CD13 [7]. Rat
Dlk+ or Thy-1+ cells isolated from mid-gestational fetal liver exhibit
characteristics expected for hepatic stem/progenitor cells [8]. In
addition to enrichment in fetal liver, hepatic stem/progenitor cells
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_19, © Springer Science+Business Media, LLC, part of Springer Nature 2019
211
212 Sei Kakinuma and Akihide Kamiya
2 Materials
2.2 Recipient Mice 1. Mice. C57BL/6 mice or nude mice (male or female) are used
and Pretreatment in this protocol. Pretreatment of recipient mice starts at 3 or
Before Transplantation 4 weeks old.
214 Sei Kakinuma and Akihide Kamiya
3 Methods
3.1 Preparation 1. Fetal livers derived from mid-gestational fetal mice (embryonic
of Donor Cells (Hepatic day 12.5–14.5) are harvested using fine small forceps and
Progenitor Cells placed in sterile PBS. The livers are then transferred to EGTA
Derived from Fetal buffer in a sterile Petri dish and minced using small scissors at
Mice) room temperature.
2. Transfer the minced livers gently using a pipette into a 15 mL
centrifuge tube. Incubate the minced livers in EGTA buffer at
37 C for 5 min.
3. Centrifuge the collection tube at 150–200 g for 3 min (see
Note 7). Remove the supernatant and tap the centrifugation
tube gently to suspend the cell pellet.
4. Suspend with 0.05% collagenase solution (see Note 8). Incu-
bate the cells at 37 C for 15 min. After incubation, tap the
centrifugation tube vigorously (see Note 9).
5. Add DMEM with 10% FCS (see Note 10), and centrifuge the
collection tube at 150–200 g for 3 min. Discard the
supernatant.
6. Suspend the pellet in 75 mM NH4Cl solution (see Note 11),
and incubate the cells at 4 C for 7 min.
7. Tap the collection tube (see Note 12), and add DMEM with
10% FCS to stop the hemolysis. Centrifuge the collection tube
at 150–200 g for 3 min.
8. Discard supernatant and suspend the cells in staining medium.
Cell suspensions are filtered using a cell strainer with a 70-μm
filter (see Note 13).
9. Incubate the cells with anti-Dlk antibody at 4 C for 30 min.
After incubation, cells are suspended in staining medium and
centrifuged at 150–200 g for 3 min (see Note 4).
10. Discard supernatant and suspend the cells in staining medium.
Incubate the cells with secondary antibody and treat the cells
according to the manufacturer’s instructions for each magnetic
selection system.
Transplantation of Hepatic Progenitor Cells 215
11. Suspend the harvested cells in DMEM with 10% FCS. Cell
suspensions are filtered using nylon mesh with 70-μm pores.
12. The volume of cell suspension should be adjusted to
0.1–0.15 mL/mouse (see Note 14). Harvested cells should
be transplanted into recipient mice as soon as possible.
3.3 Partial 1. Perform 70% hepatectomy under general anesthesia using the
Hepatectomy classic procedure described by Higgins and Anderson [17].
2. Briefly, after the confirmation of deep anesthesia, cut the skin
and peritoneum at midline of the abdomen.
3. Ligate the left medial lobe and right medial lobe (see Note 16)
of the liver proximally using surgical silk sutures. Confirm a
change of liver color, indicating effective ischemia of the ligated
lobes.
4. Remove the left medial lobe and right medial lobe (see Note
17).
5. Ligate the left lateral lobe of the liver proximally using surgical
silk sutures. Confirm a change of liver color, indicating effective
ischemia of the ligated lobes.
6. Remove the left lateral lobe.
3.4 Cell 1. Loosely ligate the inferior pole of the spleen using surgical silk
Transplantation sutures (see Note 18).
2. Inject an aliquot of cells suspended in 0.1–0.15 mL of DMEM
supplemented with 10% FCS into the splenic pulp from the
inferior pole of the spleen using a 30-gauge needle.
3. Tightly ligate the inferior pole of the spleen proximal to the
injection point (see Note 19).
4. Suture the skin and peritoneum using surgical sutures.
5. Sacrifice mice 3–12 weeks after transplantation, at which
time donor cells can be clearly detected in recipient livers
(Figs. 1 and 2).
216 Sei Kakinuma and Akihide Kamiya
Fig. 1 Transplanted E13.5 Dlk+ hepatic progenitor cells exhibit liver-repopulating activity in vivo. Dlk+ cells
derived from E13.5 fetal livers were transplanted into nude mice treated with retrorsine before partial
hepatectomy. Mice were analyzed 4 weeks after transplantation. Representative fluorescent stereoscopic
images of recipient liver are shown. The recipient liver contained many GFP+ signals. Bright positive signals
(arrows) and dull positive signals (arrow heads) were detected, with variation in brightness ascribed to
variation in proximity to the liver surface
Fig. 2 Microscopic view of GFP+ donor cells. GFP+ donor cells were detected as a colony composed of 5–10
cells (dashed circle). Donor-derived cells formed colonies producing GFP (green) and albumin (red), indicating
that such cells engrafted as parenchymal cells
Transplantation of Hepatic Progenitor Cells 217
4 Notes
Acknowledgments
References
1. Rashid T, Kobayashi T, Nakauchi H (2014) development in Mice. Gastroenterology 137
Revisiting the flight of Icarus: making human (3):1114–1126
organs from PSCs with large animal chimeras. 10. Dorrell C, Erker L, Schug J, Kopp JL, Canaday
Cell Stem Cell 15(4):406–409 PS, Fox AJ, Smirnova O, Duncan AW, Fine-
2. Shiojiri N, Lemire JM, Fausto N (1991) Cell gold MJ, Sander M, Kaestner KH, Grompe M
lineages and oval cell progenitors in rat liver (2011) Prospective isolation of a bipotential
development. Cancer Res 51(10):2611–2620 clonogenic liver progenitor cell in adult mice.
3. Kakinuma S, Nakauchi H, Watanabe M (2009) Genes Dev 25(11):1193–1203
Hepatic stem/progenitor cells and stem-cell 11. Huch M, Dorrell C, Boj SF, van Es JH, Li VS,
transplantation for the treatment of liver dis- van de Wetering M, Sato T, Hamer K, Sasaki N,
ease. J Gastroenterol 44(3):167–172 Finegold MJ, Haft A, Vries RG, Grompe M,
4. Tanimizu N, Nishikawa M, Saito H, Clevers H (2013) In vitro expansion of single
Tsujimura T, Miyajima A (2003) Isolation of Lgr5+ liver stem cells induced by Wnt-driven
hepatoblasts based on the expression of Dlk/ regeneration. Nature 494(7436):247–250
Pref-1. J Cell Sci 116. (Pt 9:1775–1786 12. Rhim JA, Sandgren EP, Palmiter RD, Brinster
5. Nitou M, Sugiyama Y, Ishikawa K, Shiojiri N RL (1995) Complete reconstitution of mouse
(2002) Purification of fetal mouse hepatoblasts liver with xenogeneic hepatocytes. Proc Natl
by magnetic beads coated with monoclonal Acad Sci USA 92(11):4942–4946
anti-e-cadherin antibodies and their in vitro 13. Overturf K, Al-Dhalimy M, Manning K, Ou
culture. Exp Cell Res 279(2):330–343 CN, Finegold M, Grompe M (1998) Ex vivo
6. Watanabe T, Nakagawa K, Ohata S, hepatic gene therapy of a mouse model of
Kitagawa D, Nishitai G, Seo J, Tanemura S, Hereditary Tyrosinemia Type I. Hum Gene
Shimizu N, Kishimoto H, Wada T, Aoki J, Ther 9(3):295–304
Arai H, Iwatsubo T, Mochita M, Watanabe T, 14. Maeda H, Shigoka M, Wang Y, Fu Y, Wesson
Satake M, Ito Y, Matsuyama T, Mak TW, Pen- RN, Lin Q, Montgomery RA, Enzan H, Sun Z
ninger JM, Nishina H, Katada T (2002) (2014) Disappearance of GFP-positive hepato-
SEK1/MKK4-mediated SAPK/JNK signaling cytes transplanted into the liver of syngeneic
participates in embryonic hepatoblast prolifer- wild-type rats pretreated with retrorsine. PLoS
ation via a pathway different from NF-kappaB- One 9(5):e95880
induced anti-apoptosis. Dev Biol 250 15. Dan YY, Riehle KJ, Lazaro C, Teoh N,
(2):332–347 Haque J, Campbell JS, Fausto N (2006) Isola-
7. Kakinuma S, Ohta H, Kamiya A, Yamazaki Y, tion of multipotent progenitor cells from
Oikawa T, Okada K, Nakauchi H (2009) Ana- human fetal liver capable of differentiating
lyses of cell surface molecules on hepatic stem/ into liver and mesenchymal lineages. Proc
progenitor cells in mouse fetal liver. J Hepatol Natl Acad Sci U S A 103(26):9912–9917
51(1):127–138 16. Kost DP, Michalopoulos GK (1991) Effect of
8. Oertel M, Menthena A, Chen YQ, Teisner B, 2% dimethyl sulfoxide on the mitogenic prop-
Jensen CH, Shafritz DA (2008) Purification of erties of epidermal growth factor and hepato-
fetal liver stem/progenitor cells containing all cyte growth factor in primary hepatocyte
the repopulation potential for normal adult rat culture. J Cell Physiol 147(2):274–280
liver. Gastroenterology 134(3):823–832 17. Higgins G, Anderson R (1931) Experimental
9. Kamiya A, Kakinuma S, Yamazaki Y, Nakauchi pathology of the liver I: restoration of the liver
H (2009) Enrichment and clonal culture of of the white ret following partial surgical
progenitor cells during mouse postnatal liver removal. Arch Pathol 12:186–202
Chapter 20
Abstract
Liver cancer consists of two main histological subtypes, hepatocellular carcinoma and cholangiocarcinoma,
both of which have poor prognosis. Therefore, in searching for new therapeutic targets, adequate mouse
models to develop and validate therapeutic strategies are urgently needed. Although there are mouse
models of liver cancer, each model has shortcomings. To overcome these shortcomings, a mouse model
using a hydrodynamic tail vein injection and the Sleeping Beauty transposon was developed. By inducing
stable expression of oncogenes in mouse hepatocytes in vivo, the model can easily induce liver cancer with
specific characteristics that depend on the oncogenes used to induce carcinogenesis. Here, we describe the
details of the methods to induce hepatocellular carcinoma or cholangiocarcinoma from mouse hepatocytes.
Key words Sleeping Beauty transposon, Hydrodynamic tail vein injection, Hepatocellular carcinoma,
Intrahepatic cholangiocarcinoma, Mouse model, Liver tumor, Liver cancer
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_20, © Springer Science+Business Media, LLC, part of Springer Nature 2019
221
222 Masahiro Yamamoto et al.
a b
1. Cut transposon content
Plasmid for expression of SB transposase
SB transposase
"
SB transposase
"
Promoter SB transposase polyA Promoter Gene of Interest polyA
ITR ITR
Fig. 1 The Sleeping Beauty (SB) transposon system. (a) Components of the Sleeping Beauty transposon
system. The SB transposase expression plasmid and a plasmid containing SB transposon cassette. ITR
inverted terminal repeat. (b) Integration of the transposon cassette into a host genome by the SB transposase.
SB transposase transfers a gene in the transposon cassette into a host genome in a cut-and-paste manner
Table 1
Sleeping Beauty transposon-mediated liver cancer models
2 Materials
3 Methods
3.1 HTVi 1. Purify the plasmids with the endotoxin-free Maxiprep kit (Qia-
gen) or equilibrium. Because endotoxin causes adverse or toxic
effects on animals, endotoxin must be removed [32]. A high
concentration of endotoxin reduces the transfection efficiency
in eukaryotic cells [33].
2. For integrating two oncogenic genes, the molar ratio of SB
transposase-expressing plasmid and two plasmids carrying
oncogenic genes is 1:2:2, and the total amount of plasmid is
25 μg (see Note 4).
3. Dissolve the plasmids in 2.5 mL of Ringer solution or saline in a
sterile manner.
4. Expose the mice to anesthesia with isoflurane.
5. Gather 2.5 mL of the plasmid solution with a 2.5-mL syringe
with a Luer Lock.
6. Attach a 25G needle to the syringe and remove any air bubbles
from the syringe.
7. Warm the tail of the mouse with hot water (at 40–50 C) to
dilate the lateral tail vein, and clean the insertion site with 70%
ethanol.
8. Insert the needle into the lateral tail vein, and rapidly infuse the
solution in a continuous manner within 8 s.
9. Press the insertion site with gauze until bleeding stops.
10. Successful injection leads to short-term gene expression in
about 30% of hepatocytes.
Fig. 2 Hepatocellular carcinoma model induced by myrAKT and HRASG12V. (a) Gross appearance of the liver at
2, 4, and 6 weeks after the HTVi. At 6 weeks, multinodular liver tumors develop. Scale bar, 10 mm. (b)
Hematoxylin and eosin (HE) staining of the liver. At 2 weeks, tumor cells with abundant lipid accumulation in
their cytoplasm appear. At 4 weeks, clusters of tumor cells devoid of lipid accumulation (arrowheads) emerge.
At 6 weeks, HCCs with scant lipid accumulation develop. The upper panels are enlarged in the lower panels.
Scale bar, 50 μm
Fig. 3 Cholangiocarcinoma model induced by myrAKT and YAPS127A. (a) Gross appearances of the liver at 2, 4,
and 6 weeks after the HTVi. Tumors diffusely infiltrate into the liver. Scale bar, 10 mm. (b) Microscopic
appearances of the AKT/YAP tumors. HE stain, Sirius red stain, and immunohistochemistry for cytokeratin
19 (CK19) and grainyhead-like 2 (Grhl2) at 2, 4, and 6 weeks after the HTVi. Scale bar, 50 μm. Reproduced
from Ref. 28 with permission from Elsevier
230 Masahiro Yamamoto et al.
Fig. 4 Biliary cystadenoma model induced by myrAKT and N2ICD. (a) Gross appearances of the liver 6 weeks
after the HTVi. Scale bar, 10 mm. (b) HE staining of the tumors. Scale bar, 50 μm
Table 2
Primary antibodies for immunohistochemical markers for hepatocellular and cholangiocellular
differentiation
3.7 Immunohisto- The antibodies and their optimal dilution for specification of hepa-
chemistry of Markers tocellular and cholangiocellular differentiation are listed in Table 2.
for Hepatocytic The staining procedure can be performed at room temperature
and Cholangiocytic unless otherwise specified.
Differentiation 1. Deparaffinize sections as described in Subheading 3.6.
2. Immerse sections in 3% H2O2 dissolved in methanol for 10 min
to block endogenous peroxidase.
3. Briefly wash sections with PBST once.
4. Heat sections in a preheated Target Retrieval solution at 98 C
for 40 min in an electric thermos.
5. Remove sections from the electric thermos and cool them to
room temperature.
6. Circle the tissues with a hydrophobic pen.
7. Block the tissues with blocking reagent for 30 min in a humidi-
fier chamber. If the source of the primary antibody is mouse,
please see Note 5 to avoid secondary antibody to binding to
mouse serum.
8. Incubate the tissues with primary antibody dissolved in the
blocking reagent for 1 h at room temperature or overnight at
4 C.
9. Wash the slide with PBST for 5 min twice.
10. Incubate the tissues with secondary antibody for 30 min.
11. Wash the slide with PBST for 5 min twice.
12. Visualize with the DAB solution. During visualization, check
the slide under a microscope. When optimal staining is
obtained, stop the reaction by putting the slides into tap water.
13. Counterstain slides in Mayer’s hematoxylin solution for 1 min.
14. Wash slides with running tap water for 10 min.
232 Masahiro Yamamoto et al.
15. Rehydrate and clear the slides and then mount a cover glass on
them as described in Subheading 3.6.
16. Dry slides until the mounting medium solidifies.
4 Notes
References
1. Ferlay J, Soerjomataram I, Dikshit R, Eser S, 5. Palmer WC, Patel T (2012) Are common fac-
Mathers C, Rebelo M, Parkin DM, Forman D, tors involved in the pathogenesis of primary
Bray F (2015) Cancer incidence and mortality liver cancers? A meta-analysis of risk factors
worldwide: sources, methods and major pat- for intrahepatic cholangiocarcinoma. J Hepatol
terns in GLOBOCAN 2012. Int J Cancer 57(1):69–76. https://doi.org/10.1016/j.
136(5):E359–E386. https://doi.org/10. jhep.2012.02.022
1002/ijc.29210 6. Forner A, Reig M, Bruix J (2018) Hepatocel-
2. Altekruse SF, Henley SJ, Cucinelli JE, lular carcinoma. Lancet 391:1301–1314.
McGlynn KA (2014) Changing hepatocellular https://doi.org/10.1016/s0140-6736(18)
carcinoma incidence and liver cancer mortality 30010-2
rates in the United States. Am J Gastroenterol 7. Llovet JM, Ricci S, Mazzaferro V, Hilgard P,
109(4):542–553. https://doi.org/10.1038/ Gane E, Blanc JF, de Oliveira AC, Santoro A,
ajg.2014.11 Raoul JL, Forner A, Schwartz M, Porta C,
3. El-Serag HB, Kanwal F (2014) Epidemiology Zeuzem S, Bolondi L, Greten TF, Galle PR,
of hepatocellular carcinoma in the United Seitz JF, Borbath I, Haussinger D, Giannaris T,
States: where are we? Where do we go? Hepa- Shan M, Moscovici M, Voliotis D, Bruix J,
tology 60(5):1767–1775. https://doi.org/10. SHARP Investigators Study Group (2008)
1002/hep.27222 Sorafenib in advanced hepatocellular carci-
4. Razumilava N, Gores GJ (2014) Cholangiocar- noma. N Engl J Med 359(4):378–390.
cinoma. Lancet 383(9935):2168–2179. https://doi.org/10.1056/NEJMoa0708857
https://doi.org/10.1016/S0140-6736(13) 8. Cheng AL, Kang YK, Chen Z, Tsao CJ, Qin S,
61903-0 Kim JS, Luo R, Feng J, Ye S, Yang TS, Xu J,
Mouse Models of Liver Cancers from Mature Hepatocytes 233
Sun Y, Liang H, Liu J, Wang J, Tak WY, Pan H, Ther 15(12):2063–2069. https://doi.org/10.
Burock K, Zou J, Voliotis D, Guan Z (2009) 1038/sj.mt.6300314
Efficacy and safety of sorafenib in patients in 18. Sawyer GJ, Rela M, Davenport M,
the Asia-Pacific region with advanced hepato- Whitehorne M, Zhang X, Fabre JW (2009)
cellular carcinoma: a phase III randomised, Hydrodynamic gene delivery to the liver: theo-
double-blind, placebo-controlled trial. Lancet retical and practical issues for clinical applica-
Oncol 10(1):25–34. https://doi.org/10. tion. Curr Gene Ther 9(2):128–135
1016/S1470-2045(08)70285-7 19. Bonamassa B, Hai L, Liu D (2011) Hydrody-
9. Inuzuka T, Nishikawa H, Sekikawa A, namic gene delivery and its applications in
Takeda H, Henmi S, Sakamoto A, Saito S, pharmaceutical research. Pharm Res 28
Kita R, Kimura T, Osaki Y, Kudo M (2011) (4):694–701. https://doi.org/10.1007/
Complete response of advanced hepatocellular s11095-010-0338-9
carcinoma with multiple lung metastases trea- 20. Lander ES, Linton LM, Birren B, Nusbaum C,
ted with sorafenib: a case report. Oncology 81 Zody MC, Baldwin J, Devon K, Dewar K,
(Suppl 1):152–157. https://doi.org/10. Doyle M, FitzHugh W, Funke R, Gage D,
1159/000333279 Harris K, Heaford A, Howland J, Kann L,
10. Valle J, Wasan H, Palmer DH, Cunningham D, Lehoczky J, LeVine R, McEwan P,
Anthoney A, Maraveyas A, Madhusudan S, McKernan K, Meldrim J, Mesirov JP,
Iveson T, Hughes S, Pereira SP, Roughton M, Miranda C, Morris W, Naylor J, Raymond C,
Bridgewater J, ABC Trial Investigators (2010) Rosetti M, Santos R, Sheridan A, Sougnez C,
Cisplatin plus gemcitabine versus gemcitabine Stange-Thomann Y, Stojanovic N,
for biliary tract cancer. N Engl J Med 362 Subramanian A, Wyman D, Rogers J,
(14):1273–1281. https://doi.org/10.1056/ Sulston J, Ainscough R, Beck S, Bentley D,
NEJMoa0908721 Burton J, Clee C, Carter N, Coulson A,
11. Heindryckx F, Colle I, Van Vlierberghe H Deadman R, Deloukas P, Dunham A,
(2009) Experimental mouse models for hepa- Dunham I, Durbin R, French L, Grafham D,
tocellular carcinoma research. Int J Exp Pathol Gregory S, Hubbard T, Humphray S, Hunt A,
90(4):367–386. https://doi.org/10.1111/j. Jones M, Lloyd C, McMurray A, Matthews L,
1365-2613.2009.00656.x Mercer S, Milne S, Mullikin JC, Mungall A,
12. Chen X, Calvisi DF (2014) Hydrodynamic Plumb R, Ross M, Shownkeen R, Sims S,
transfection for generation of novel mouse Waterston RH, Wilson RK, Hillier LW,
models for liver cancer research. Am J Pathol McPherson JD, Marra MA, Mardis ER, Fulton
184(4):912–923. https://doi.org/10.1016/j. LA, Chinwalla AT, Pepin KH, Gish WR, Chis-
ajpath.2013.12.002 soe SL, Wendl MC, Delehaunty KD, Miner
13. Landgraf M, McGovern JA, Friedl P, Hutma- TL, Delehaunty A, Kramer JB, Cook LL, Ful-
cher DW (2018) Rational design of mouse ton RS, Johnson DL, Minx PJ, Clifton SW,
models for cancer research. Trends Biotechnol Hawkins T, Branscomb E, Predki P,
36:242–251. https://doi.org/10.1016/j. Richardson P, Wenning S, Slezak T,
tibtech.2017.12.001 Doggett N, Cheng JF, Olsen A, Lucas S,
Elkin C, Uberbacher E, Frazier M, Gibbs RA,
14. Liu F, Song Y, Liu D (1999) Hydrodynamics- Muzny DM, Scherer SE, Bouck JB, Sodergren
based transfection in animals by systemic EJ, Worley KC, Rives CM, Gorrell JH, Metzker
administration of plasmid DNA. Gene Ther 6 ML, Naylor SL, Kucherlapati RS, Nelson DL,
(7):1258–1266. https://doi.org/10.1038/sj. Weinstock GM, Sakaki Y, Fujiyama A,
gt.3300947 Hattori M, Yada T, Toyoda A, Itoh T,
15. Zhang G, Budker V, Wolff JA (1999) High Kawagoe C, Watanabe H, Totoki Y, Taylor T,
levels of foreign gene expression in hepatocytes Weissenbach J, Heilig R, Saurin W,
after tail vein injections of naked plasmid DNA. Artiguenave F, Brottier P, Bruls T, Pelletier E,
Hum Gene Ther 10(10):1735–1737. https:// Robert C, Wincker P, Smith DR, Doucette-
doi.org/10.1089/10430349950017734 Stamm L, Rubenfield M, Weinstock K, Lee
16. Yokoo T, Kamimura K, Abe H, Kobayashi Y, HM, Dubois J, Rosenthal A, Platzer M,
Kanefuji T, Ogawa K, Goto R, Oda M, Suda T, Nyakatura G, Taudien S, Rump A, Yang H,
Terai S (2016) Liver-targeted hydrodynamic Yu J, Wang J, Huang G, Gu J, Hood L,
gene therapy: Recent advances in the tech- Rowen L, Madan A, Qin S, Davis RW, Feder-
nique. World J Gastroenterol 22 spiel NA, Abola AP, Proctor MJ, Myers RM,
(40):8862–8868. https://doi.org/10.3748/ Schmutz J, Dickson M, Grimwood J, Cox DR,
wjg.v22.i40.8862 Olson MV, Kaul R, Raymond C, Shimizu N,
17. Suda T, Liu D (2007) Hydrodynamic gene Kawasaki K, Minoshima S, Evans GA,
delivery: its principles and applications. Mol Athanasiou M, Schultz R, Roe BA, Chen F,
234 Masahiro Yamamoto et al.
pathways. Oncogene 36(36):5087–5097. 42. Calvisi DF, Wang C, Ho C, Ladu S, Lee SA,
https://doi.org/10.1038/onc.2017.114 Mattu S, Destefanis G, Delogu S,
35. Mendez-Lucas A, Li X, Hu J, Che L, Song X, Zimmermann A, Ericsson J, Brozzetti S,
Jia J, Wang J, Xie C, Driscoll PC, Tschaharga- Staniscia T, Chen X, Dombrowski F, Evert M
neh DF, Calvisi DF, Yuneva M, Chen X (2017) (2011) Increased lipogenesis, induced by
Glucose catabolism in liver tumors induced by AKT-mTORC1-RPS6 signaling, promotes
c-MYC can be sustained by various PKM1/ development of human hepatocellular carci-
PKM2 ratios and pyruvate kinase activities. noma. Gastroenterology 140(3):1071–1083.
Cancer Res 77(16):4355–4364. https://doi. https://doi.org/10.1053/j.gastro.2010.12.
org/10.1158/0008-5472.CAN-17-0498 006
36. Tao J, Zhang R, Singh S, Poddar M, Xu E, 43. Lee SA, Ladu S, Evert M, Dombrowski F, De
Oertel M, Chen X, Ganesh S, Abrams M, Murtas V, Chen X, Calvisi DF (2010) Synergis-
Monga SP (2017) Targeting beta-catenin in tic role of Sprouty2 inactivation and c-Met
hepatocellular cancers induced by coexpression up-regulation in mouse and human hepatocar-
of mutant beta-catenin and K-Ras in mice. cinogenesis. Hepatology 52(2):506–517.
Hepatology 65(5):1581–1599. https://doi. https://doi.org/10.1002/hep.23681
org/10.1002/hep.28975 44. Xu CR, Lee S, Ho C, Bommi P, Huang SA,
37. Tao J, Xu E, Zhao Y, Singh S, Li X, Couchy G, Cheung ST, Dimri GP, Chen X (2009) Bmi1
Chen X, Zucman-Rossi J, Chikina M, Monga functions as an oncogene independent of
SP (2016) Modeling a human hepatocellular Ink4A/Arf repression in hepatic carcinogene-
carcinoma subset in mice through coexpression sis. Mol Cancer Res 7(12):1937–1945.
of met and point-mutant beta-catenin. Hepa- https://doi.org/10.1158/1541-7786.MCR-
tology 64(5):1587–1605. https://doi.org/10. 09-0333
1002/hep.28601 45. Lee SA, Ho C, Roy R, Kosinski C, Patil MA,
38. Hu J, Che L, Li L, Pilo MG, Cigliano A, Tward AD, Fridlyand J, Chen X (2008) Inte-
Ribback S, Li X, Latte G, Mela M, Evert M, gration of genomic analysis and in vivo trans-
Dombrowski F, Zheng G, Chen X, Calvisi DF fection to identify sprouty 2 as a candidate
(2016) Co-activation of AKT and c-Met trig- tumor suppressor in liver cancer. Hepatology
gers rapid hepatocellular carcinoma develop- 47(4):1200–1210. https://doi.org/10.1002/
ment via the mTORC1/FASN pathway in hep.22169
mice. Sci Rep 6:20484. https://doi.org/10. 46. Tward AD, Jones KD, Yant S, Cheung ST, Fan
1038/srep20484 ST, Chen X, Kay MA, Wang R, Bishop JM
39. Ho C, Wang C, Mattu S, Destefanis G, Ladu S, (2007) Distinct pathways of genomic progres-
Delogu S, Armbruster J, Fan L, Lee SA, sion to benign and malignant tumors of the
Jiang L, Dombrowski F, Evert M, Chen X, liver. Proc Natl Acad Sci U S A 104
Calvisi DF (2012) AKT (v-akt murine thy- (37):14771–14776. https://doi.org/10.
moma viral oncogene homolog 1) and N-Ras 1073/pnas.0706578104
(neuroblastoma ras viral oncogene homolog) 47. Patil MA, Lee SA, Macias E, Lam ET, Xu C,
coactivation in the mouse liver promotes rapid Jones KD, Ho C, Rodriguez-Puebla M, Chen
carcinogenesis by way of mTOR (mammalian X (2009) Role of cyclin D1 as a mediator of
target of rapamycin complex 1), FOXM1 (fork- c-Met- and beta-catenin-induced hepatocarci-
head box M1)/SKP2, and c-Myc pathways. nogenesis. Cancer Res 69(1):253–261.
Hepatology 55(3):833–845. https://doi.org/ https://doi.org/10.1158/0008-5472.CAN-
10.1002/hep.24736 08-2514
40. Wang C, Delogu S, Ho C, Lee SA, Gui B, 48. Zhang S, Song X, Cao D, Xu Z, Fan B, Che L,
Jiang L, Ladu S, Cigliano A, Dombrowski F, Hu J, Chen B, Dong M, Pilo MG, Cigliano A,
Evert M, Calvisi DF, Chen X (2012) Inactiva- Evert K, Ribback S, Dombrowski F, Pascale
tion of Spry2 accelerates AKT-driven hepato- RM, Cossu A, Vidili G, Porcu A, Simile MM,
carcinogenesis via activation of MAPK and Pes GM, Giannelli G, Gordan J, Wei L,
PKM2 pathways. J Hepatol 57(3):577–583. Evert M, Cong W, Calvisi DF, Chen X (2017)
https://doi.org/10.1016/j.jhep.2012.04.026 Pan-mTOR inhibitor MLN0128 is effective
41. Stauffer JK, Scarzello AJ, Andersen JB, De against intrahepatic cholangiocarcinoma in
Kluyver RL, Back TC, Weiss JM, Thorgeirsson mice. J Hepatol 67(6):1194–1203. https://
SS, Wiltrout RH (2011) Coactivation of AKT doi.org/10.1016/j.jhep.2017.07.006
and beta-catenin in mice rapidly induces forma- 49. Carlson CM, Frandsen JL, Kirchhof N, McIvor
tion of lipogenic liver tumors. Cancer Res 71 RS, Largaespada DA (2005) Somatic integra-
(7):2718–2727. https://doi.org/10.1158/ tion of an oncogene-harboring Sleeping
0008-5472.CAN-10-2705 Beauty transposon models liver tumor
236 Masahiro Yamamoto et al.
development in the mouse. Proc Natl Acad Sci 51. Chow EK, Fan LL, Chen X, Bishop JM (2012)
U S A 102(47):17059–17064. https://doi. Oncogene-specific formation of chemoresis-
org/10.1073/pnas.0502974102 tant murine hepatic cancer stem cells. Hepatol-
50. Tao J, Calvisi DF, Ranganathan S, Cigliano A, ogy 56(4):1331–1341. https://doi.org/10.
Zhou L, Singh S, Jiang L, Fan B, 1002/hep.25776
Terracciano L, Armeanu-Ebinger S, 52. Ju HL, Ahn SH, Kim DY, Baek S, Chung SI,
Ribback S, Dombrowski F, Evert M, Chen X, Seong J, Han KH, Ro SW (2013) Investigation
Monga SPS (2014) Activation of beta-catenin of oncogenic cooperation in simple liver-
and Yap1 in human hepatoblastoma and induc- specific transgenic mouse models using nonin-
tion of hepatocarcinogenesis in mice. Gastro- vasive in vivo imaging. PLoS One 8(3):
enterology 147(3):690–701. https://doi.org/ e59869. https://doi.org/10.1371/journal.
10.1053/j.gastro.2014.05.004 pone.0059869
Chapter 21
Abstract
A good mouse model is mandatory for elucidating carcinogenic mechanisms and identifying the cellular
origin of cancer. Although the lack of an appropriate mouse model has hampered investigation of extrahe-
patic cholangiocarcinoma (ECC), we recently established a novel mouse model of biliary injury-related
ECC by ductal cell-specific activation of Kras and deletion of transforming growth factor (TGF) β receptor
type 2 and E-cadherin. Using this mouse model, we identified that peribiliary glands, which are considered
a biliary epithelial stem cell niche, are potential cellular origins of ECC. Furthermore, we established an
extrahepatic biliary organoid-derived xenograft cholangiocarcinoma (CC) model by lentiviral induction of
Cre in organoids. This organoid system recreated the in vivo conditions and facilitated analysis of carcino-
genesis. In this chapter, we describe the protocol used to establish our mouse model of ECC derived from
peribiliary glands and our extrahepatic biliary organoid-derived xenograft model of CC.
1 Introduction
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4_21, © Springer Science+Business Media, LLC, part of Springer Nature 2019
237
238 Hayato Nakagawa et al.
2 Materials
3 Methods
3.1 In Vivo Model 1. Tamoxifen (200 mg/kg) was administered by oral gavage to
of ECC KTC-K19CreERT mice (7–8 weeks old) for 3 consecutive days.
This protocol can induce gene recombination (~40%) in all
parts of the biliary tree, including the EHBD, PBGs, GB,
perihilar bile duct (BD), and intrahepatic bile duct.
2. On day 7, surface BECs started to detach from the BD wall.
240 Hayato Nakagawa et al.
Fig. 1 Histological findings of extrahepatic bile duct (EHBD) in KTC-K19CreERT mice. (a) Hematoxylin and eosin
(H&E)-stained images of EHBDs and PBGs from Cre-negative control and KTC-K19CreERT mice at 10 days after
tamoxifen administration (scale bar: upper panels, 200 μm; lower panels, 50 μm). Right panel shows enlarged
and dysplastic peribiliary glands (PBGs). (b) H&E-stained images of EHBDs from Cre-negative control and
KTC-K19CreERT mice at 3.5 weeks after tamoxifen administration (scale bar: left two panels, 500 μm; right
panel, 50 μm)
3.2 Extrahepatic 1. Isolate the EHBD tissue from Cre-negative KTC mice and
Biliary Organoid- place tissue in a 10-cm petri dish filled with PBS.
Derived Xenograft CC 2. Remove the adhered interstitial and pancreatic tissue from the
Model EHBD using forceps with gentle touching.
3.2.1 Culture of EHBD 3. Cut the EHBD tissue down the middle and divide it into two
Organoids parts.
4. Place the EHBD tissue in a 1.5-mL tube containing 1 mL
digestion buffer.
5. Incubate for 30 min at 37 C.
6. Gently disperse the EHBD tissue by pipetting.
7. Centrifuge at 300 g for 5 min at 4 C. Discard the
supernatant.
8. Place the 1.5-mL tube containing EHBD cells on ice and add
1 mL cold PBS. Vortex for 10 s.
9. Centrifuge at 300 g for 5 min at 4 C. Discard the
supernatant.
10. Place the 1.5-mL tube containing EHBD cells on ice. Add
80 μL ice-cold Matrigel and mix by pipetting using a cold
200-μL tip with a large-end diameter (cut off end of the tip)
(see Note 2). Do not generate bubbles while mixing.
11. Seed EHBD cells in a 40 μL drop in the center of each well of a
24-well plate.
12. Carefully place the 24-well plate in a 37 C incubator for
10 min.
13. Add 500 μL culture medium supplemented with ROCK inhib-
itor to each well.
14. Organoid formation was observed 1–2 days after plating.
EHBD organoids formed a ball-like structure and were com-
posed of a single layer of the biliary epithelium (Fig. 2a, b).
15. Refresh the culture medium every 2–3 days and passage the
EHBD organoids every 5–10 days.
16. When passaging organoids, remove the culture medium and
add 1 mL Cell Recovery Solution to dissolve the Matrigel.
17. Place the solution in a 1.5-mL tube and incubate 10 min on ice
to precipitate the organoids by gravity. Remove the upper
portion of the supernatant (~700 μL), which contains dead
cells and debris.
18. Add 700 μL Cell Recovery Solution and suspend the
organoids well.
19. Pass the solution through a 29-gauge insulin syringe once or
twice.
242 Hayato Nakagawa et al.
Fig. 2 Extrahepatic biliary organoid-derived xenograft CC model. (a) Micrograph of biliary organoids cultured
from EHBDs of mice (scale bar: 250 μm). (b) H&E-stained images of EHBD organoids from Cre-negative KTC
mice infected with Cre-expressing or control lentivirus (scale bar: 50 μm). (c) H&E-stained image of an
organoid-derived tumor (scale bar: 50 μm)
20. Centrifuge at 600 g for 5 min at 4 C. Discard the
supernatant.
21. Add 400 μL cold PBS and suspend well.
22. Transfer approximately 800 organoid fragments to a new 1.5-
mL tube on ice.
23. Centrifuge at 600 g for 5 min at 4 C. Discard the
supernatant.
24. Add 160 μL Matrigel and suspend well.
25. Seed 40 μL organoid solution into each well of a 24-well plate.
26. Carefully place the 24-well plate in a 37 C incubator for
10 min.
27. Add 500 μL culture medium supplemented with ROCK inhib-
itor into each well.
28. Place the plate in a 37 C incubator.
3.2.2 Induction of Gene 1. Resolve Matrigel and wash organoids in the same way with
Recombination in EHBD Subheading 3.2.1, steps 16–23.
Organoids Using a 2. Add 15 μL lentivirus solution (titer 1 107 transduction
Cre-Expressing Lentivirus units/mL) and suspend well.
3. Incubate 30 min on ice.
Mouse Model for Cholangiocarcinoma from Peribiliary Glands 243
3.2.3 Extrahepatic Biliary 1. Culture EHBD organoids from Cre-negative KTC mice and
Organoid-Derived induce gene recombination as described above.
Xenograft CC Model 2. Seed 100 organoid fragments in each well of a 24-well plate.
3. After 2 days, remove the medium.
4. Add 1 mL cold PBS to each well and mix with the Matrigel
containing organoids.
5. Place the solution in a 1.5-mL tube on ice.
6. Centrifuge at 600 g for 5 min at 4 C.
7. Discard the supernatant and leave approximately 100 μL Matri-
gel/PBS mixture containing organoids.
8. Add 100 μL Matrigel/PBS mixture (1:1) and suspend well. At
this stage, 200 ul suspension remains.
9. Inject 100 μL of the organoid suspension subcutaneously in
two sites at the back of nude mice using an ice-cold 29-gauge
insulin syringe.
10. After 2 months, approximately two-thirds of the organoids will
have formed tumors with a similar histology to that of ECC in
KTC-K19CreERT mice (Fig. 2c).
4 Notes
Acknowledgments
This study was supported by the Japan Society for the Promotion of
Science KAKENHI grant number 15K09039, Astellas Foundation
for Research on Metabolic Disorders, Nakayama Cancer Research
Institute, Okinaka Memorial Institute for Medical Research,
Research Grant of the Princess Takamatsu Cancer Research Fund,
and Foundation for Promotion of Cancer Research in Japan.
References
1. Rizvi S, Gores GJ (2013) Pathogenesis, diag- 7. Dipaola F, Shivakumar P, Pfister J, Walters S,
nosis, and management of cholangiocarci- Sabla G, Bezerra JA (2013) Identification of
noma. Gastroenterology 145(6):1215–1229. intramural epithelial networks linked to peri-
https://doi.org/10.1053/j.gastro.2013.10. biliary glands that express progenitor cell mar-
013 kers and proliferate after injury in mice.
2. Malhi H, Gores GJ (2006) Cholangiocarci- Hepatology 58(4):1486–1496. https://doi.
noma: modern advances in understanding a org/10.1002/hep.26485
deadly old disease. J Hepatol 45(6):856–867. 8. Lanzoni G, Cardinale V, Carpino G (2016)
https://doi.org/10.1016/j.jhep.2006.09.001 The hepatic, biliary, and pancreatic network of
3. Chan-On W, Nairismagi ML, Ong CK, Lim stem/progenitor cell niches in humans: a new
WK, Dima S, Pairojkul C, Lim KH, McPherson reference frame for disease and regeneration.
JR, Cutcutache I, Heng HL, Ooi L, Chung A, Hepatology 64(1):277–286. https://doi.org/
Chow P, Cheow PC, Lee SY, Choo SP, Tan IB, 10.1002/hep.28326
Duda D, Nastase A, Myint SS, Wong BH, 9. Xu X, Kobayashi S, Qiao W, Li C, Xiao C,
Gan A, Rajasegaran V, Ng CC, Nagarajan S, Radaeva S, Stiles B, Wang RH, Ohara N,
Jusakul A, Zhang S, Vohra P, Yu W, Huang D, Yoshino T, LeRoith D, Torbenson MS, Gores
Sithithaworn P, Yongvanit P, Wongkham S, GJ, Wu H, Gao B, Deng CX (2006) Induction
Khuntikeo N, Bhudhisawasdi V, Popescu I, of intrahepatic cholangiocellular carcinoma by
Rozen SG, Tan P, Teh BT (2013) Exome liver-specific disruption of Smad4 and Pten in
sequencing identifies distinct mutational pat- mice. J Clin Invest 116(7):1843–1852.
terns in liver fluke-related and non-infection- https://doi.org/10.1172/JCI27282
related bile duct cancers. Nat Genet 45 10. Ikenoue T, Terakado Y, Nakagawa H,
(12):1474–1478. https://doi.org/10.1038/ Hikiba Y, Fujii T, Matsubara D, Noguchi R,
ng.2806 Zhu C, Yamamoto K, Kudo Y, Asaoka Y,
4. Nakamura H, Arai Y, Totoki Y, Shirota T, Yamaguchi K, Ijichi H, Tateishi K,
Elzawahry A, Kato M, Hama N, Hosoda F, Fukushima N, Maeda S, Koike K, Furukawa Y
Urushidate T, Ohashi S, Hiraoka N, (2016) A novel mouse model of intrahepatic
Ojima H, Shimada K, Okusaka T, Kosuge T, cholangiocarcinoma induced by liver-specific
Miyagawa S, Shibata T (2015) Genomic spec- Kras activation and Pten deletion. Sci Rep
tra of biliary tract cancer. Nat Genet 47 6:23899. https://doi.org/10.1038/
(9):1003–1010. https://doi.org/10.1038/ srep23899
ng.3375 11. Guest RV, Boulter L, Kendall TJ, Minnis-
5. Sekiya S, Suzuki A (2012) Intrahepatic cholan- Lyons SE, Walker R, Wigmore SJ, Sansom
giocarcinoma can arise from Notch-mediated OJ, Forbes SJ (2014) Cell lineage tracing
conversion of hepatocytes. J Clin Invest 122 reveals a biliary origin of intrahepatic cholan-
(11):3914–3918. https://doi.org/10.1172/ giocarcinoma. Cancer Res 74(4):1005–1010.
JCI63065 https://doi.org/10.1158/0008-5472.CAN-
6. Fan B, Malato Y, Calvisi DF, Naqvi S, 13-1911
Razumilava N, Ribback S, Gores GJ, 12. Nakagawa H, Suzuki N, Hirata Y, Hikiba Y,
Dombrowski F, Evert M, Chen X, Willenbring Hayakawa Y, Kinoshita H, Ihara S, Uchino K,
H (2012) Cholangiocarcinomas can originate Nishikawa Y, Ijichi H, Otsuka M, Arita J,
from hepatocytes in mice. J Clin Invest 122 Sakamoto Y, Hasegawa K, Kokudo N,
(8):2911–2915. https://doi.org/10.1172/ Tateishi K, Koike K (2017) Biliary epithelial
JCI63212 injury-induced regenerative response by IL-33
Mouse Model for Cholangiocarcinoma from Peribiliary Glands 245
A Extrahepatic Cholangiocarcinoma
(ECC) ...............................................237–240, 243
Alginate hydrogel beads ...................................... 158–160
Animal models..........................46, 47, 50, 118, 222, 223 F
B Fetal liver cells (FLCs) ............................... 3, 7, 117, 124,
175, 179, 181–183
Bile ducts .....................46, 143, 144, 188–190, 223, 238
Fluorescence Activated Cell Sorting (FACS) ..13, 14, 16,
Biliary epithelial cells (BECs) ....................... v, 45–55, 83, 20–25, 109–111, 217
84, 125, 175, 176, 178, 180–182, 188, 189, 191,
237–240, 243 H
Biliary organoid...................................238–240, 242, 243
Biliary tree ................................. 178–180, 188, 189, 194, Hepatic progenitor cells .......................... 5, 8, 71, 72, 78,
196, 197, 239 80, 81, 144–146, 148–150, 211
Bipotential ..............................................v, 9–17, 122, 211 Hepatic stellate cells (HSCs) .............................. 104, 105,
Bipotentiality ................................................................. 117 108, 110, 111, 131, 139–141, 211
Hepatoblasts .............................................v, 144, 151, 211
C Hepatocellular carcinoma .....................54, 221, 224, 227
Hepatocyte ........................................ 4, 9, 22, 29, 45, 59,
Canine model ....................................................... 201–208 83, 93, 103, 117, 131, 143, 157, 167, 175, 187,
Carbon tetrachloride (CCl4)............... 47, 49, 51–53, 61,
201, 222, 237
80, 104, 105, 108, 111, 114, 201–203, 206–208 Hepatocyte ablation ..................................................84, 89
CD44 ..................................................... 30, 33, 36–38, 40 Heterogeneity....................................................... 9, 71–81
Cell proliferation .................................46, 50, 72, 80, 128
Human Induced Pluripotent Stem Cells (Human Ips
Chemically Induced Liver Progenitors Cells) ............................... 145, 146, 148, 150–152
(CLiPs)......................................... 31, 34, 117–129 Hybrid periportal hepatocytes........................................ 60
Chemical screen ........................................................83–89 Hydrodynamic tail vein injection
Cholangiocytes .............................v, 9, 10, 15, 16, 19–27,
(HTVi) .....................................222–227, 229, 232
46, 47, 143–146, 149, 151, 211, 227
Chronic liver injury ...................................................19, 80 I
Clonal culture.................................................................. 16
Colony ................................ 5, 10, 11, 14–16, 19, 20, 26, Induced hepatocytes (Iheps) .............. 103–105, 108–112
38, 65, 71, 72, 76–78, 81, 146, 149, 216 Induction of hepatic differentiation............................. 124
Colony assay .................................................................... 14 Inflammation .................................... 50, 52, 83, 201, 237
Interstitial flow .............................................................. 167
D Intrahepatic cholangiocarcinoma ................................. 221
In vivo reprogramming........................................ 104, 108
Differentiation...................................... 10, 11, 14–16, 20,
45, 71, 83, 124, 135, 139, 141, 144–146, L
148–151, 212, 231
Duct cells ................................................... 19, 59, 60, 144 Lineage tracing ..... 9, 45–55, 59–69, 104, 105, 111, 237
Ductular reaction (DR) ......................................... 46, 188 Liver3, 9, 19, 29, 45, 59, 72, 83, 93, 103, 117, 131, 143,
157, 167, 175, 187, 201, 211, 221, 237
E Liver cancer .......................................................v, 221–224
Liver cirrhosis (LC).............................................. 201, 202
E-cadherin ............................................................ 211, 238 Liver progenitor cells (LPCs) ....................................... 136
Extracellular matrix ........... 103, 145, 146, 149–151, 168
Liver regeneration .................. v, 46, 59, 83–89, 187, 201
Naoki Tanimizu (ed.), Hepatic Stem Cells: Methods and Protocols, Methods in Molecular Biology, vol. 1905,
https://doi.org/10.1007/978-1-4939-8961-4, © Springer Science+Business Media, LLC, part of Springer Nature 2019
247
HEPATIC STEM CELLS: METHODS AND PROTOCOLS
248 Index
Liver sinusoidal endothelial cells (LSECs)..................131, P
136, 138, 140, 141
Liver stem/progenitor cell (LPCs) ....................... v, 9–17, Partial hepatectomy (PHx)........................ 45, 47, 48, 50,
46, 71–81, 83, 117, 131, 134, 135, 140, 141, 52, 212, 215, 216
176, 187, 224 Peribiliary glands (PBGs) ........................... 237, 238, 240
Liver tumor .......................................................... 223, 227
Q
Long-term proliferation ........................................ 54, 125
Quantitative analysis .............................................. 71, 206
M
R
Mesenchymal stem cells ........................94, 203, 205, 207
Methylcellulose (MC).........................157–161, 163, 164 Reprogramming .................. 93, 103, 104, 118, 124, 223
Microfluidic device............................................... 167–174 Retrorsine .....................................................212, 214–218
Microscopy .....................................................74, 112, 162
Microstructures ............................................................. 157 S
Mouse .........................5, 6, 8–17, 19–27, 29, 32, 33, 37,
Sleeping Beauty transposon................................. 223, 224
40, 47, 49, 52, 55, 60, 62, 64–66, 69, 72–74, 78,
Small hepatocyte (SHs) ..................................... 30, 36, 38
84, 93, 103, 110, 114, 125, 132, 137, 144, 145,
Spheroid..................................4–6, 8, 167, 176, 181–184
148, 163, 177, 189, 192, 196, 197, 211, 212,
Suppressor of tumorigenicity 14 protein
215, 221–232, 237–243
(ST14)...............................................19, 21, 24–27
Mouse model................................v, 19, 59, 84, 103, 105,
221, 238, 239 T
Multicellular spheroid (MCS) ............................. 157–164
3D culture ............................................................ 167, 168
N Three-dimensional imaging............................................ 77
3D tissue structures ...................................v, 72, 168, 173
Nitroreductase (NTR) ...............84, 85, 88, 89, 105, 111
Tissue engineering ............................................... 167, 168
Tissue stem cell ............................................................... 59
O
Transdifferentiation................................... 46, 93–95, 100
Organoid ............................................19–21, 24, 26, 144, Transplantation .................................. v, 29, 83, 117, 175,
175–185, 242 201, 211–218, 222