ArticlePDF AvailableLiterature Review

Design by Nature: Emerging Applications of Native Liver Extracellular Matrix for Cholangiocyte Organoid-Based Regenerative Medicine

Authors:

Abstract and Figures

Organoid technology holds great promise for regenerative medicine. Recent studies show feasibility for bile duct tissue repair in humans by successfully transplanting cholangiocyte organoids in liver grafts during perfusion. Large-scale expansion of cholangiocytes is essential for extending these regenerative medicine applications. Human cholangiocyte organoids have a high and stable proliferation capacity, making them an attractive source of cholangiocytes. Commercially available basement membrane extract (BME) is used to expand the organoids. BME allows the cells to self-organize into 3D structures and stimulates cell proliferation. However, the use of BME is limiting the clinical applications of the organoids. There is a need for alternative tissue-specific and clinically relevant culture substrates capable of supporting organoid proliferation. Hydrogels prepared from decellularized and solubilized native livers are an attractive alternative for BME. These hydrogels can be used for the culture and expansion of cholangiocyte organoids in a clinically relevant manner. Moreover, the liver-derived hydrogels retain tissue-specific aspects of the extracellular microenvironment. They are composed of a complex mixture of bioactive and biodegradable extracellular matrix (ECM) components and can support the growth of various hepatobiliary cells. In this review, we provide an overview of the clinical potential of native liver ECM-based hydrogels for applications with human cholangiocyte organoids. We discuss the current limitations of BME for the clinical applications of organoids and how native ECM hydrogels can potentially overcome these problems in an effort to unlock the full regenerative clinical potential of the organoids.
Content may be subject to copyright.


Citation: Willemse, J.; van der Laan,
L.J.W.; de Jonge, J.; Verstegen, M.M.A.
Design by Nature: Emerging
Applications of Native Liver
Extracellular Matrix for
Cholangiocyte Organoid-Based
Regenerative Medicine.
Bioengineering 2022,9, 110.
https://doi.org/10.3390/
bioengineering9030110
Academic Editor: Joaquim M.
S. Cabral
Received: 17 January 2022
Accepted: 4 March 2022
Published: 7 March 2022
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2022 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
bioengineering
Review
Design by Nature: Emerging Applications of Native Liver
Extracellular Matrix for Cholangiocyte Organoid-Based
Regenerative Medicine
Jorke Willemse , Luc J. W. van der Laan , Jeroen de Jonge and Monique M. A. Verstegen *,†
Department of Surgery, Erasmus MC Transplant Institute, University Medical Center,
3015 CN Rotterdam, The Netherlands; j.willemse@erasmusmc.nl (J.W.); l.vanderlaan@erasmusmc.nl (L.J.W.v.d.L.);
j.dejonge.1@erasmusmc.nl (J.d.J.)
*Correspondence: m.verstegen@erasmusmc.nl; Tel.: +31-10-703-5528
These authors contributed equally to this work.
Abstract:
Organoid technology holds great promise for regenerative medicine. Recent studies show
feasibility for bile duct tissue repair in humans by successfully transplanting cholangiocyte organoids
in liver grafts during perfusion. Large-scale expansion of cholangiocytes is essential for extending
these regenerative medicine applications. Human cholangiocyte organoids have a high and stable
proliferation capacity, making them an attractive source of cholangiocytes. Commercially available
basement membrane extract (BME) is used to expand the organoids. BME allows the cells to self-
organize into 3D structures and stimulates cell proliferation. However, the use of BME is limiting
the clinical applications of the organoids. There is a need for alternative tissue-specific and clinically
relevant culture substrates capable of supporting organoid proliferation. Hydrogels prepared from
decellularized and solubilized native livers are an attractive alternative for BME. These hydrogels
can be used for the culture and expansion of cholangiocyte organoids in a clinically relevant manner.
Moreover, the liver-derived hydrogels retain tissue-specific aspects of the extracellular microenviron-
ment. They are composed of a complex mixture of bioactive and biodegradable extracellular matrix
(ECM) components and can support the growth of various hepatobiliary cells. In this review, we
provide an overview of the clinical potential of native liver ECM-based hydrogels for applications
with human cholangiocyte organoids. We discuss the current limitations of BME for the clinical
applications of organoids and how native ECM hydrogels can potentially overcome these problems
in an effort to unlock the full regenerative clinical potential of the organoids.
Keywords:
extracellular matrix; cholangiocyte organoids; bile duct; liver; tissue engineering;
regenerative medicine; culture substrates
1. Introduction
The shortage of donor organs is a central theme in the field of liver transplantation,
which still is the only curative treatment option for patients suffering from end-stage liver
failure. The donor shortage leads to high waiting list mortality. However, suitable donor
livers often do not become available in time for up to 20% of the patients on the waiting
list [
1
3
]. Efforts made to increase the pool of available donor livers include the use of
extended criteria donor organs, such as the use of donation after circulatory death (DCD)
organs [
4
]. The use of DCD livers is associated with a higher incidence of ischemia-type
biliary lesions (16% vs. 3% when compared to donation after brain death) [
5
,
6
]. Cholangio-
cytes form an active barrier between the cytotoxic bile and surrounding tissue [
7
]. They
are sensitive to ischemia, and the extra period of warm ischemia in DCD transplantation
can cause deficits in the biliary epithelium, such as non-anastomotic strictures [
4
,
8
,
9
]. Ulti-
mately, 65% of patients with ischemic cholangiopathy require retransplantation, as there is
Bioengineering 2022,9, 110. https://doi.org/10.3390/bioengineering9030110 https://www.mdpi.com/journal/bioengineering
Bioengineering 2022,9, 110 2 of 17
currently no alternative treatment option available [
6
]. This does not only have far-reaching
impact on patients, but also reduces the number of available donor grafts for other patients.
Regenerative medicine strategies could repair the biliary deficits while the liver is
preserved ex vivo on an organ perfusion setup. Eshmuminov et al. recently showed
that it is feasible to maintain human livers on the pump for up to 7 days [
10
]. These
improvements in the field of organ preservation could open up a window of opportunity
for ex vivo organ repair. Stem cells or cholangiocytes can be used to repair deficits in the
biliary epithelium [11,12].
The aim of this review is to discuss the potential of native human liver extracellular
matrix (ECM) for the clinical grade applications of cholangiocyte organoids for regener-
ative medicine in patients. We discuss why the use of mouse tumor-derived basement
membrane extract (BME) is currently limiting the clinical applications of cholangiocyte
organoids. In addition, we provide an overview of requirements for clinically relevant
culture substrates. We will discuss how liver ECM hydrogels provide an alternative culture
substrate and why the use of decellularized liver tissue can unlock the full clinical potential
of cholangiocyte organoids.
2. The Potential of Organoids in Tissue Regeneration
The
in vitro
isolation and large-scale expansion of primary cholangiocytes cultured on
traditional cell culture plastic is challenging [
13
]. Alternative sources of cholangiocytes are
therefore required. Pluripotent stem cells (embryonic or induced) can be directed towards
cholangiocytes, but this requires extensive differentiation of cells [
14
16
]. Moreover, there
is the potential risk of aberrant (de)differentiation and teratoma formation [
17
19
]. The use
of adult tissue-specific progenitors to grow cholangiocyte organoids, enables cell expansion
from relatively small (0.5–1.0 cm
3
) human liver biopsies. These organoids maintain a
cholangiocyte-like phenotype in vitro [20].
The first cholangiocyte organoids derived from human liver biopsies were described
by Huch et al. in 2015 [
21
]. This research was based upon the discovery of Leucine-rich
repeat-containing G-protein coupled receptor 5 (LGR5) positive stem cells found in the
intestine [
22
], which gave rise to intestinal organoids [
23
]. A similar approach was described
for LGR5+ mouse liver-derived organoids [
24
], followed by human liver organoid cultures.
Huch et al. showed that EPCAM positive cells in human liver biopsies can give rise
to spheroid-like cultures with an efficiency of 28% (SD:
±
3%) [
21
]. These liver-derived
cholangiocyte organoids have a cholangiocyte-like phenotype and express cholangiocyte
markers, such as KRT-7/19 and EPCAM, as well as progenitor markers (e.g., SOX-9,
LGR-5
).
Moreover, they have a high and stable proliferation capacity. Relatively small tissue
biopsies can yield clinically relevant numbers of cells in a relatively short time span [
25
,
26
].
Moreover, the liver-derived organoids can express and upregulate hepatocyte markers,
such as albumin, HNF-4
α
and CYP-3A4, upon differentiation towards hepatocyte-like
cells [
21
,
26
,
27
]. This makes them a potential source of both cholangiocytes and hepatocytes.
However, the expression of hepatocyte markers and hepatocyte functionality does not yet
reach similar levels as primary human hepatocytes.
With the expansion of hepato-pancreato and biliary organoid research, a consistent
nomenclature was proposed, and the liver-derived organoids were renamed to intrahepatic
cholangiocyte organoids (ICO) to better reflect their origin [
20
] (see Figure 1for an overview
of the different cholangiocyte organoids and sources). Cholangiocyte organoids were estab-
lished from gallbladder tissue (gallbladder cholangiocyte organoids; GCO) [
28
], and from
the cholangiocytes’ inner lining the extrahepatic bile duct (EBD, extrahepatic cholangiocyte
organoids; ECO) [
27
,
29
]. Organoids were also initiated from the circulating EPCAM pos-
itive cells in fresh bile samples (bile-derived cholangiocyte organoids, BCO) [
30
]. These
organoids all share similar phenotypic features when cultured, as they all grow in similar
spherical structures with comparable proliferation rates [
27
,
30
,
31
]. They express similar
progenitor and mature cholangiocyte markers, but are uniquely related to their original
tissue of origin and show subtle differences. Rimland et al. showed that differences related
Bioengineering 2022,9, 110 3 of 17
to the regional origin of the organoids were retained between ICO, ECO and GCO [
31
]. In
addition, only ICO have the potential to upregulate hepatocyte markers when grown in
hepatocyte differentiation conditions [27].
Bioengineering 2022, 9, x FOR PEER REVIEW 3 of 17
They express similar progenitor and mature cholangiocyte markers, but are uniquely re-
lated to their original tissue of origin and show subtle differences. Rimland et al. showed
that differences related to the regional origin of the organoids were retained between ICO,
ECO and GCO [31]. In addition, only ICO have the potential to upregulate hepatocyte
markers when grown in hepatocyte differentiation conditions [27].
Figure 1. Schematic overview of the workup of cholangiocyte organoids in BME cultures and the
different sources of organoids. Cholangiocyte organoids can be initiated from liver tissue biopsies
(intrahepatic cholangiocyte organoids; ICO), extrahepatic bile duct tissue biopsies (extrahepatic
cholangiocyte organoids; ECO) and gallbladder tissue biopsies (gallbladder cholangiocyte organ-
oids; GCO). Organoids can also be initiated from bile samples (bile-derived cholangiocyte organ-
oids; BCO). Organoids are traditionally grown and expanded in BME.
3. Repairing Damaged Organs Using Cholangiocyte Organoids
The ability to generate large numbers of cells from relatively small (patient-derived)
biopsies is of interest for regenerative cell therapy applications. Therefore, cholangiocyte
organoid-derived cells could be a promising source of hepatobiliary cells for organ repair
applications [32–34]. ICO differentiated towards hepatocyte-like cells were capable of en-
grafting a damaged mouse liver. These cells showed some level of hepatocyte functional-
ity as human albumin was detected 120 days after engrafting [21]. However, engraftment
rates were extremely low (<1%). Therefore, it is not likely that ICO will be used as a cell
source for hepatocyte transplantation, as long as current hepatocyte differentiation proto-
cols and cell administration protocols are not improved.
Cholangiocyte organoids can also yield cholangiocyte-like cells without the need for
additional differentiation steps. Therefore, these cells are of interest for the repair of dam-
aged biliary epithelium. Recently, ground-breaking proof of concept was given by Sam-
paziotis et al. They showed that cholangiocyte organoids can successfully repair deficits
in the biliary epithelium of intrahepatic bile ducts after injecting cells that were derived
from the organoids into the biliary tree of a mouse model (Figure 2) [35]. They also showed
that GCO can be used to repair deficits of the biliary epithelium of human livers ex vivo
while the liver was perfused on a normothermic machine perfusion device. This shows
the enormous clinical potential of cholangiocyte organoids.
Figure 1.
Schematic overview of the workup of cholangiocyte organoids in BME cultures and the
different sources of organoids. Cholangiocyte organoids can be initiated from liver tissue biopsies
(intrahepatic cholangiocyte organoids; ICO), extrahepatic bile duct tissue biopsies (extrahepatic
cholangiocyte organoids; ECO) and gallbladder tissue biopsies (gallbladder cholangiocyte organoids;
GCO). Organoids can also be initiated from bile samples (bile-derived cholangiocyte organoids; BCO).
Organoids are traditionally grown and expanded in BME.
3. Repairing Damaged Organs Using Cholangiocyte Organoids
The ability to generate large numbers of cells from relatively small (patient-derived)
biopsies is of interest for regenerative cell therapy applications. Therefore, cholangiocyte
organoid-derived cells could be a promising source of hepatobiliary cells for organ repair
applications [
32
34
]. ICO differentiated towards hepatocyte-like cells were capable of en-
grafting a damaged mouse liver. These cells showed some level of hepatocyte functionality
as human albumin was detected 120 days after engrafting [
21
]. However, engraftment rates
were extremely low (<1%). Therefore, it is not likely that ICO will be used as a cell source
for hepatocyte transplantation, as long as current hepatocyte differentiation protocols and
cell administration protocols are not improved.
Cholangiocyte organoids can also yield cholangiocyte-like cells without the need
for additional differentiation steps. Therefore, these cells are of interest for the repair
of damaged biliary epithelium. Recently, ground-breaking proof of concept was given
by Sampaziotis et al. They showed that cholangiocyte organoids can successfully repair
deficits in the biliary epithelium of intrahepatic bile ducts after injecting cells that were
derived from the organoids into the biliary tree of a mouse model (Figure 2) [
35
]. They also
showed that GCO can be used to repair deficits of the biliary epithelium of human livers
ex vivo while the liver was perfused on a normothermic machine perfusion device. This
shows the enormous clinical potential of cholangiocyte organoids.
Bioengineering 2022,9, 110 4 of 17
Bioengineering 2022, 9, x FOR PEER REVIEW 4 of 17
Figure 2. Cholangiocyte organoids can repair deficits in the biliary epithelium. This treatment can
restore adequate drainage of bile and prevent build-up of toxic bile inside the liver. Cholangiocyte
organoids can be infused into the biliary tree to repair the damaged biliary tree.
4. Basement Membrane Extract as Culture Substrates
The clinical application of cholangiocyte organoids is currently limited by the use of
non-GMP (good manufacturing practice)-compliant basement membrane extracts (BMEs)
in which organoids are typically cultured. BME is a complex mixture of extracellular ma-
trix (ECM) components derived from the tumor mass produced by Englebreth-Holm-
Swarm (EHS) mouse cells. These cells produce an abundance of basement membrane
components, which can be extracted and processed into hydrogels [36,37]. The main con-
stituents of BME are laminin-111, collagen type IV and enactin [37–39]. Other components
include a myriad of other bioactive ECM components and growth factors [39]. The extrac-
tion of these components and subsequent reconstitution was first described in 1986 by
Kleinman et al. [36]. They described a protocol where the EHS cells were propagated by
transplantation in mice and basement membrane components were extracted from the
tumor mass by breaking up protein–protein bonds. The subsequent viscous liquid solidi-
fied into a hydrogel at 37 °C and was later commercialized under the name Matrigel [37–
40]. Nowadays, different BME formulations (e.g., growth factor reduced or collagen type
IV enriched) are commercially available from various manufacturers (e.g., Corning Mat-
rigel or Cultrex BME). Exact production methods are proprietary information and could
therefore differ from previously described protocols.
BME is typically used as an in vitro replacement of the ECM, as it creates a bioactive
and biodegradable 3D environment for the cells. Commercially available BMEs are ready-
to-use formulations as no additional chemicals are required for solidification of the pre-
gel solutions. The non-cytotoxic viscous pre-gel solution remains a liquid at 4 °C and so-
lidifies into a relatively soft hydrogel at 37 °C. Moreover, the optical clarity of the extracts
allows for day-to-day monitoring. Therefore, BMEs have long been the golden standard
for many different types of in vitro assays, such as angiogenesis assays [41,42], (tumor)
cell migration assays [43] or for maintaining (induced) pluripotent stem cells undifferen-
tiated [19,44,45] (see Kleinman et al. [46] and Benton et al. [47] for brief overviews on these
applications). BME is used for the expansion of organoids for similar reasons. The bioac-
tive components allow the epithelial cells to self-organize into their typical organoid struc-
tures.
However, there are also disadvantages of the use of BME for the culture of organoids.
The exact composition is poorly defined and large batch-to-batch differences have been
reported [38,44,48]. BME also keeps cells in an undifferentiated and proliferative state
[19,39]. This could also hamper the differentiation of cholangiocyte organoids towards
Figure 2.
Cholangiocyte organoids can repair deficits in the biliary epithelium. This treatment can
restore adequate drainage of bile and prevent build-up of toxic bile inside the liver. Cholangiocyte
organoids can be infused into the biliary tree to repair the damaged biliary tree.
4. Basement Membrane Extract as Culture Substrates
The clinical application of cholangiocyte organoids is currently limited by the use of
non-GMP (good manufacturing practice)-compliant basement membrane extracts (BMEs)
in which organoids are typically cultured. BME is a complex mixture of extracellular matrix
(ECM) components derived from the tumor mass produced by Englebreth-Holm-Swarm
(EHS) mouse cells. These cells produce an abundance of basement membrane components,
which can be extracted and processed into hydrogels [
36
,
37
]. The main constituents of BME
are laminin-111, collagen type IV and enactin [
37
39
]. Other components include a myriad
of other bioactive ECM components and growth factors [
39
]. The extraction of these com-
ponents and subsequent reconstitution was first described in 1986 by Kleinman et al. [
36
].
They described a protocol where the EHS cells were propagated by transplantation in mice
and basement membrane components were extracted from the tumor mass by breaking up
protein–protein bonds. The subsequent viscous liquid solidified into a hydrogel at 37
C
and was later commercialized under the name Matrigel [
37
40
]. Nowadays, different BME
formulations (e.g., growth factor reduced or collagen type IV enriched) are commercially
available from various manufacturers (e.g., Corning Matrigel or Cultrex BME). Exact pro-
duction methods are proprietary information and could therefore differ from previously
described protocols.
BME is typically used as an
in vitro
replacement of the ECM, as it creates a bioactive
and biodegradable 3D environment for the cells. Commercially available BMEs are ready-
to-use formulations as no additional chemicals are required for solidification of the pre-gel
solutions. The non-cytotoxic viscous pre-gel solution remains a liquid at 4
C and solidifies
into a relatively soft hydrogel at 37
C. Moreover, the optical clarity of the extracts allows
for day-to-day monitoring. Therefore, BMEs have long been the golden standard for many
different types of
in vitro
assays, such as angiogenesis assays [
41
,
42
], (tumor) cell migration
assays [
43
] or for maintaining (induced) pluripotent stem cells undifferentiated [
19
,
44
,
45
]
(see Kleinman et al. [
46
] and Benton et al. [
47
] for brief overviews on these applications).
BME is used for the expansion of organoids for similar reasons. The bioactive components
allow the epithelial cells to self-organize into their typical organoid structures.
However, there are also disadvantages of the use of BME for the culture of organoids.
The exact composition is poorly defined and large batch-to-batch differences have been re-
ported [
38
,
44
,
48
]. BME also keeps cells in an undifferentiated and proliferative
state [19,39].
This could also hamper the differentiation of cholangiocyte organoids towards mature
hepatocytes. Moreover, the constituents are not specific for liver tissue. Under normal
circumstances, the main constituent of BME (laminin-111) is not found in healthy adult liver
Bioengineering 2022,9, 110 5 of 17
parenchymal regions [
49
,
50
]. However, in situations where proliferating cells are required,
such as during embryonic development or regeneration after damage, laminin-111 can be
found here [
49
,
51
53
]. Evidence suggests that the presence of laminin can maintain the
stemness of certain cells and inhibit the differentiation of hepatic progenitor cells towards
mature hepatocytes [52,54,55].
5. Tissue-Specific Alternative Culture Substrates
The use of a culture substrate capable of closely mimicking the native liver ECM
could drive cells towards mature and functional cholangiocyte and/or improve differ-
entiation of ICO towards hepatocyte-like cells [
44
,
56
,
57
]. Tissue-specific microenviron-
ments can be built using liver ECM components and incorporation of cell signaling moi-
eties (e.g., growth factors). Gradients of growth factors play an important role during
embryonic development but are also important for maintaining tissue homeostasis or
are involved in tissue repair [
53
,
56
,
58
]. Spatiotemporal deposition of vascular endothe-
lial growth factor (VEGF) through the ECM, for example, guides vascular sprouting
during angio(neo)genesis [5962].
Growth factors, such as endothelial growth factor (EGF)
or hepatocyte growth factor (HGF), are known to maintain the hepatocyte phenotype in cells
and are involved in the differentiation of stem cells towards hepatocyte-like cells [
63
,
64
]. In
addition, biophysical components (e.g., stiffness or elasticity) can influence the behavior
of cells through complex mechanotransduction pathways [
65
67
]. This has been shown
in vitro
with the differentiation of mesenchymal stromal cells, which can be directed by
altering matrix elasticity [
67
,
68
]. Growing hepatocytes
in vitro
on rigid matrices decreases
their ability to maintain a hepatocyte phenotype [
57
,
69
71
]. Similarly, an increase in liver
ECM stiffness is associated with a reduction in hepatocyte functionality and can lead to
liver fibrosis [
49
51
,
72
]. This shows the importance of having a culture substrate capable of
mimicking the native ECM.
Alternative culture substrates have to meet additional requirements. They must,
for example, allow for the expansion of the organoids in a GMP-compliant environment.
Organoids self-organize into 3D structures through complex cell–cell and cell–matrix
interactions. Therefore, the alternative substrate should also allow for the expansion of the
organoids by being capable of either deformation or site-specific degradation. Moreover,
the growth of cells also implies that the mass-transfer of oxygen, nutrients and metabolic
waste products through the substrate is required in order to maintain cell viability while
cell numbers are increasing.
5.1. Hydrogels as an ECM Mimic
Hydrogels are a promising class of materials for use as
in vitro
culture substrates [
73
].
Their polymeric networks are capable of maintaining relatively large amounts of water and
can form hydrated environments for cells
in vitro
. These networks are typically porous
and allow for mass-transfer to a certain degree [
73
]. Hydrogels have found widespread
use in biomedical applications such as contact lenses, drug delivery systems and wound
dressings [
74
]. The polymer backbones of hydrogels are typically derived from natural
or synthetic sources. Examples of natural polymers used for creating hydrogels are al-
ginate, fibrin, chitosan, cellulose and collagen [
73
,
75
,
76
]. Poly(acrylic acid), poly(vinyl
alcohol) and poly(ethylene glycol) (PEG) are examples of synthetic polymers [
74
,
76
]. These
polymers often require chemical modifications to create cross-linkable and/or degradable
networks [73].
Synthetic hydrogels are generally well defined and can be modified with relative ease.
This allows for the adjustment of, for example, the number of physical crosslinking sites,
which can alter the stiffness of the hydrogel. However, synthetic hydrogels often lack the
complexity of the native ECM in terms of mixtures of different bioactive ECM molecules
or degradation sites for cells [
77
]. PEG-based hydrogels were tested for expansion and
differentiation of intestinal organoids, but required incorporation of ECM components (e.g.,
fibronectin or laminin), cell adhesion motifs (RGD) or matrix metalloproteinase-degradable
Bioengineering 2022,9, 110 6 of 17
sites [
77
,
78
]. Nonetheless, Gjorevski et al. showed that relatively stiff hydrogels (~1.3 kPa)
increased the proliferation of these organoids, whereas differentiation to hepatocyte-like
cells was more optimal in a softer hydrogel [
78
]. Similar synthetic hydrogels also support
expansion and differentiation of human ICO, but still required the addition of ECM compo-
nents or cell-adhesion motifs [
79
]. Ye et al. used a hydrogel based on polyisocyanopeptides
(PIC) supplemented with laminin-111 and showed that ICO have similar proliferation rates
when compared to ICO grown in BME [
80
]. Nanocellulose hydrogels appeared to promote
differentiation towards hepatocytes [
81
]. However, it remains elusive whether these hydro-
gels can fully replace BME, as initiation of ICO in these hydrogels was not tested. Moreover,
nanocellulose hydrogels also required supplementation with ECM-components and it was
suggested by authors that further optimization regarding addition of ECM-components
could enhance ICO growth [81].
In short, creating a tissue-specific microenvironment capable of mimicking the native
ECM using synthetic hydrogels requires extensive experience in biochemistry and bioengi-
neering. Fine-tuning an optimal tissue-specific culture substrate is practically not possible
without the use of high-throughput screening methods [
82
]. However, even when using
high throughput screening methods, combining and testing multiple ECM components
is challenging and time consuming. This problem of recreating tissue mimicking culture
substrates can also be circumvented using extracts derived from decellularized healthy
tissues [83].
Liver decellularization procedures have been described for livers from different ani-
mals, such as rodents [
84
87
] and pigs [
58
,
88
94
], but also for whole human livers [
94
96
].
These protocols aim to remove all cellular components from liver tissue while retaining
the architecture of the ECM, including the highly tissue-specific spatiotemporal deposition
of the ECM components [
97
99
]. Subsequently, the ECM components can be extracted by
solubilizing the matrix using the enzyme pepsin in an acidic environment [
100
102
]. The
resulting extracts consist of bioactive and biodegradable ECM components, which can form
collagen-based hydrogels without the need for complex chemical modifications [103].
5.2. ECM-Based Hydrogels
Similar protocols exist for the extraction of collagen from collagen-rich tissues, such as
rat-tail tendon, fish scales or skin tissue [
104
], for biomedical applications (see [
103
] for a
comprehensive review on clinical applications of collagen-based materials). The pepsin
cleaves the non-helical telopeptide regions of collagen fibers but does not affect the helical
parts of the collagen. Subsequently, the helical parts are released by the enzyme, which can
reassemble themselves in long collagen fibers after the pH is normalized to
7.4 [103,105,106].
Similar gelation occurs after pH of the pepsin solubilized ECM is normalized to 7.4. The
viscous pre-gel solution self assembles into a collagen-based hydrogel, which contain nu-
merous ECM components [
83
,
107
]. The presence of ECM molecules influence the gelation
kinetics (e.g., crosslinking of fibril formation) of the ECM hydrogels [
108
] and creates
complex hydrogels with varying mechanical characteristics.
ECM based hydrogels are derived from biological sources and can, therefore, be
subject to biological variations. This can be mitigated by creating large batches of ECM
extracts from decellularized tissues. ECM extracts are also less well suited for studying the
effects of certain biochemical or biophysical components. The hydrogels are collagen-based
and altering the concentration also alters the biophysical characteristics and vice versa.
This could be resolved by incorporating the ECM extracts into a synthetic hydrogel. This
hybrid hydrogel could allow for alterations in biophysical characteristics (e.g., stiffness)
without altering the presence of biological components. Skardal et al. used solubilized liver
ECM to decorate simplistic PEG hydrogels [109].
5.3. Applications of Liver ECM Extracts
Liver ECM-derived extracts have been investigated for improving
in vitro
hepa-
tocyte cultures by using them as a supplement for culture medium [
110
], creating 2D
Bioengineering 2022,9, 110 7 of 17
coatings [
107
,
111
,
112
] of cell culture plastics or by creating bioactive 3D environments
for hepatocytes [
71
,
100
,
107
,
111
114
]. Further applications of liver ECM extracts include
the culture of other liver cells, such as hepatic stellate cells or liver sinusoidal endothe-
lial cells [
88
,
115
], or to improve the differentiation capacity of adipose derived stromal
cells [
107
] or pluripotent stem cells towards hepatocytes [
116
118
]. The liver ECM extracts
are also an attractive alternative culture substrate for the culture of cholangiocyte organoids.
Giobbe et al. showed that different endodermal organoids (including ICO) can be cultured
in non-tissue-specific ECM-derived hydrogels derived from decellularized porcine small
intestinal submucosa (SIS) [
119
]. SIS mainly consist mostly of a mesh of collagen fibers
and contains few other proteins [
120
]. Therefore, it might not represent a tissue-specific
ECM environment for the ICO. Liver ECM extracts are a favorable alternative, since they
are tissue-specific and could be of clinical relevance. The use of liver ECM extracts could
unlock the full clinical potential of the cholangiocyte organoids.
6. Finding a Suitable Source of Liver ECM
In theory, the decellularization procedure removes all cellular components (including
immunogenic proteins and DNA) from liver tissue. ECM components are also highly
preserved between species, allowing for the use of animal liver-derived extracts for clinical
applications in humans. However, retention of species-specific differences in liver ECM
extracts have previously been reported by Loneker et al. [
110
]. These differences can
partially be explained by biological variances between species, but also depend on tissue
processing (e.g., decellularization and enzymatic digestion). Detergents can, for example,
have detrimental effects on ECM components. We previously showed that
triton-X-100 +
Sodium dodecyl sulfate removes more collagen and sGAG from porcine liver ECM than
when only triton-X-100 was used for decellularization [
94
]. Similar effects have also been
reported by others who compared different treatment methods [
100
,
121
123
]. Decellular-
ization of human livers required more detergent and longer exposure times compared to
decellularization of similar sized porcine livers [
94
]. Subsequently, this could create larger
differences between human and porcine livers. Therefore, it is important to consider the
source of liver tissue.
Decellularized human livers are a promising allogeneic source of scaffolds for tissue
engineering applications and for preparation of liver ECM extracts. However, healthy
livers are relatively scarce. Human research livers (donor livers deemed unsuitable for
transplantation) were used in our previous studies. Further improvements in the field of
organ preservation and development of ex vivo repair strategies could in the future allow
for the safe transplantation of these research livers [
12
]. Thereby, the number of available
livers for research purposes will likely further diminish. Cadaveric livers could be an
alternative source of healthy human livers. One of the disadvantages of the use of human
livers is the relative old age and subsequent age-related changes of the
ECM [124126].
With increased age comes increased stiffness and decreased elasticity due to scar tissue
formation and/or non-enzymatic crosslinking of the ECM [
127
]. The latter is caused by the
age-related accumulation of advanced glycation end-products (AGE) attached to the ECM.
This accumulation of AGE is influenced by different factors, such as dietary habits [
128
], and
can increase the aforementioned cross-links. These changes in ECM can prevent enzymatic
digestion of the ECM [
113
,
125
,
129
,
130
]. Subsequently, this can also lead to relatively large
differences between human livers [124].
Animal livers of similar age can be obtained with relative ease in a standardized
manner, thereby limiting the effect of age-related biological variances of the ECM. Small
animal livers (e.g., mice, rat or ferret) are typically well suited for small-scale recellular-
ization experiments. However, generating significant amounts of ECM extracts would
require sacrificing many animals. Porcine livers, on the other hand, are comparable in
size and weight to human livers. They are therefore promising alternatives for creating
tissue-engineering scaffolds, but also yield more liver ECM extracts per liver. The anatomy
of porcine livers is not similar to the anatomy of human livers. Porcine livers have
2–7 lobes,
Bioengineering 2022,9, 110 8 of 17
depending on breed [
131
]. Furthermore, the hepatic lobules of the porcine livers are sep-
arated by a clearly visible septa made of collagen-rich tissue (see Figure 3). These septa
resemble fibrotic human livers and are not present in human livers under normal healthy
circumstances [
72
,
131
]. The presence of these collagen-rich septa could shift the relative
distribution of ECM components present in the liver ECM extracts and this could influence
the behavior of primary human hepatocytes or stellate cells [124].
Bioengineering 2022, 9, x FOR PEER REVIEW 8 of 17
sacrificing many animals. Porcine livers, on the other hand, are comparable in size and
weight to human livers. They are therefore promising alternatives for creating tissue-en-
gineering scaffolds, but also yield more liver ECM extracts per liver. The anatomy of por-
cine livers is not similar to the anatomy of human livers. Porcine livers have 2–7 lobes,
depending on breed [131]. Furthermore, the hepatic lobules of the porcine livers are sep-
arated by a clearly visible septa made of collagen-rich tissue (see Figure 3). These septa
resemble fibrotic human livers and are not present in human livers under normal healthy
circumstances [72,131]. The presence of these collagen-rich septa could shift the relative
distribution of ECM components present in the liver ECM extracts and this could influ-
ence the behavior of primary human hepatocytes or stellate cells [124].
Figure 3. Porcine livers differ from human livers from an anatomical point of view. Porcine livers
have multiple lobes, but also contain septa (indicated by black arrows). These septa are not visible
in human livers. The scale bars represent 500 µm.
Biosafety Concerns of Using Decellularized Liver Tissue
The use of decellularized tissues in clinical settings is associated with biosafety con-
cerns. Inadequate processing of decellularized tissue can have detrimental effects on the
scaffold and cause adverse clinical outcomes, such as rapid degradation, loss of mechan-
ical integrity and/or inadequate tissue remodeling [132–134]. The presence of cellular rem-
nants, especially xenogeneic components such as α-gal (galactose-α-1,3-galactose) or
Figure 3.
Porcine livers differ from human livers from an anatomical point of view. Porcine livers
have multiple lobes, but also contain septa (indicated by black arrows). These septa are not visible in
human livers. The scale bars represent 500 µm.
Biosafety Concerns of Using Decellularized Liver Tissue
The use of decellularized tissues in clinical settings is associated with biosafety con-
cerns. Inadequate processing of decellularized tissue can have detrimental effects on the
scaffold and cause adverse clinical outcomes, such as rapid degradation, loss of mechan-
ical integrity and/or inadequate tissue remodeling [
132
134
]. The presence of cellular
remnants, especially xenogeneic components such as
α
-gal (galactose-
α
-1,3-galactose) or
MHC proteins [
135
,
136
], can influence the host response to decellularized ECM or solubi-
lized ECM (see [
137
,
138
] for comprehensive reviews on host response to animal-derived
decellularized ECM).
Bioengineering 2022,9, 110 9 of 17
Transmission of zoonotic diseases is another concern of using animal tissue-derived
materials for clinical applications. Porcine endogenous retrovirus (PERV) is an example of
a virus which could be transmitted [
139
]. Certain PERV subtypes can infect oversimplified
human cell cultures
in vitro
, but so far, there are no reports of humans which have been in-
fected with PERV after long-term exposure to pigs/porcine meat (e.g., farmers or butchers)
or patient who received porcine corneas or islets of Langerhans [
139
,
140
]. Moreover, others
have shown complete removal of detectable PERV provirus after complete decellularization
of porcine tissue [
140
,
141
]. Of note, allogeneic liver transplantation is not without risk of
transmitting human viruses [
1
]. Therefore, it is paramount that the decellularized livers
and subsequent extracts are screened for contaminants, immunogenic components and
(pro)viruses in order to mitigate these biosafety concerns.
7. Unlocking the Future Clinical Potential of Cholangiocyte Organoids
Tissue-specific ECM-based hydrogels are attractive culture substrates which can po-
tentially unlock the full clinical potential of cholangiocyte organoids. This potential is
not limited to use of the organoid-derived cells in cell therapy in ex vivo organ repair
strategies. Patient-derived cholangiocyte organoids can retain patient characteristics and
organoids cultures have been established for various hepatobiliary diseases, including
alpha1antitrypsin deficiency [
21
], cystic fibrosis [
27
], Alagille syndrome [
21
], primary
sclerosing cholangitis [
142
] and primary liver cancer [
143
,
144
]. Therefore, (personalized)
in vitro
disease models are an obvious application of the organoids (a comprehensive
review on the use of hepatobiliary organoids for disease modeling is published by Nucifero
and colleagues [
145
]). Cell–cell and cell–matrix interactions play important role during
the development of hepatobiliary diseases, as the onset is often associated with significant
changes in the ECM [
144
]. Decellularized liver tissue and liver ECM extracts can play im-
portant roles in creating these disease models. Moreover, the biological variances could also
be embraced as a means to study the effect of different environments (e.g., relatively young
ECM versus relatively old ECM [
125
,
127
]) on the behavior of cells or on the development
of certain diseases [
144
]. Ultimately, patient-derived organoids cultured in tissue-specific
matrices could lead to improved treatment strategies for hepatobiliary diseases.
7.1. Tissue Engineering the Biliary Tree
Tissue-engineered functional liver constructs have the potential of bridging the gap
between the demand and supply of donor livers of adequate livers [
146
148
]. Creating
liver constructs
in vitro
requires scaffolds that are capable of performing similar roles as
the native liver ECM. Current production techniques for alternative ECM or synthetic
supporting structures, such as 3D bio-printing, can recreate small constructs mimicking
the liver architecture with high fidelity, but producing clinically relevant sized scaffolds
is still challenging [
149
,
150
]. Use of the native liver ECM is an attractive alternative
to de novo creation of scaffolds with synthetic materials [
97
,
147
,
148
]. In recent years,
recellularization with primary hepatocytes or hepatocytes derived from various stem
cell sources have been investigated in effort to restore functionality of the hepatocyte
compartment [71,151153].
Simultaneously, a lot of effort has been invested in repopulation
of the vasculature network [
95
,
148
]. However, repopulation of the biliary compartment
has not yet been extensively studied, even though biliary epithelium is essential for proper
functioning of the liver [9,154].
Cholangiocyte organoid-derived cells are a promising source of cells for repopulation
of the biliary tree. We recently showed that cells derived from ECO and BCO were capable
or repopulating small discs (Ø3 mm) of decellularized extrahepatic bile duct (EBD) tis-
sue [
30
,
155
]. They self-organized into polarized monolayers resembling biliary epithelium
(Figure 4). Moreover, the repopulated scaffold showed increased trans epithelial electrical
resistance and cholangiocyte specific ion-channel activity could be measured. This could
be translated into transplantable EBD constructs, but the organoids could also be used to
repopulate the biliary tree of decellularized liver ECM (Figure 4) [
25
,
156
]. However, also
Bioengineering 2022,9, 110 10 of 17
here the use of BME as a culture substrate hampers the clinical applications of these lab-
grown biliary structures. Clinical grade alternatives are required for the
in vitro
expansion
of organoids, before repopulated biliary trees can be used in vivo.
Bioengineering 2022, 9, x FOR PEER REVIEW 10 of 17
epithelium (Figure 4). Moreover, the repopulated scaffold showed increased trans epithe-
lial electrical resistance and cholangiocyte specific ion-channel activity could be meas-
ured. This could be translated into transplantable EBD constructs, but the organoids could
also be used to repopulate the biliary tree of decellularized liver ECM (Figure 4) [25,156].
However, also here the use of BME as a culture substrate hampers the clinical applications
of these lab-grown biliary structures. Clinical grade alternatives are required for the in
vitro expansion of organoids, before repopulated biliary trees can be used in vivo.
Together with more liver-specific cell types, such as hepatic stellate cells and Kupffer
cells, a transplantable tissue-engineered liver construct can be made. Decellularized liver
tissue can be applied in different stages and in different forms for creating tissue-engi-
neered liver constructs in vitro. Solubilized liver ECM hydrogel can, for example, be used
for the clinically relevant expansion of cholangiocyte organoids in vitro, while decellular-
ized livers can be used as a bioactive, biodegradable and inductive scaffold for liver tissue
engineering purposes in vitro and in vivo.
Figure 4. Potential future tissue engineering applications of human cholangiocyte organoids. (A)
Cholangiocyte organoids can also be used to repopulate extrahepatic bile duct (EBD) scaffolds for
ductal tissue engineering purposes. Subsequently, these engineered ductal scaffolds can be used to
replace damaged tissue. (B) An example of a strategy for recellularization of decellularized liver
ECM. Cholangiocyte organoids can be used to repopulate the entire biliary tree. Endothelial cells
can recellularize the vasculature of the liver and ICO differentiated towards hepatocyte-like cells
can be used to repopulate the hepatocyte compartment. These different types of cells could restore
functionality of the liver.
Figure 4.
Potential future tissue engineering applications of human cholangiocyte organoids. (
A
)
Cholangiocyte organoids can also be used to repopulate extrahepatic bile duct (EBD) scaffolds for
ductal tissue engineering purposes. Subsequently, these engineered ductal scaffolds can be used to
replace damaged tissue. (
B
) An example of a strategy for recellularization of decellularized liver
ECM. Cholangiocyte organoids can be used to repopulate the entire biliary tree. Endothelial cells
can recellularize the vasculature of the liver and ICO differentiated towards hepatocyte-like cells
can be used to repopulate the hepatocyte compartment. These different types of cells could restore
functionality of the liver.
Together with more liver-specific cell types, such as hepatic stellate cells and Kupffer
cells, a transplantable tissue-engineered liver construct can be made. Decellularized liver
tissue can be applied in different stages and in different forms for creating tissue-engineered
liver constructs
in vitro
. Solubilized liver ECM hydrogel can, for example, be used for
the clinically relevant expansion of cholangiocyte organoids
in vitro
, while decellularized
livers can be used as a bioactive, biodegradable and inductive scaffold for liver tissue
engineering purposes in vitro and in vivo.
7.2. Summary
Mouse tumor-derived BMEs are commercially available and easy-to-use formulations
which allow for organoid growth and expansion. However, these one-size-fits-all BME
Bioengineering 2022,9, 110 11 of 17
formulations limit the direct clinical application of cholangiocyte organoids for cell therapy
or tissue engineering applications. In addition, BME lacks essential tissue-specific ECM
components and is known to keep cells in a high proliferative undifferentiated state. There
is a clear need for a tissue-specific alternative which can be produced according to GMP
guidelines. This alternative must also allow for the culture and large-scale expansion of
these organoids. Hydrogels derived from healthy decellularized and solubilized liver ECM
are promising alternative culture substrates for the large-scale expansion of cholangiocyte
organoids and could unlock the enormous clinical potential of the organoids.
Author Contributions:
Writing—original draft preparation, J.W.; writing—review and editing,
L.J.W.v.d.L., J.d.J. and M.M.A.V.; visualization, J.W.; funding acquisition, L.J.W.v.d.L., J.d.J. and
M.M.A.V. All authors have read and agreed to the published version of the manuscript.
Funding:
This research was funded by Medical Delta program grant, Regenerative Medicine 4D;
TKI-LSH grant, project number EMC-LSH19002 and NVGE Gastrostart fund, project number 2019-06.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Eurotransplant Annual Report 2019; Eurotransplant: Leiden, The Netherlands, 2020.
2.
Ng, M.; Fleming, T.; Robinson, M.; Thomson, B.; Graetz, N.; Margono, C.; Mullany, E.C.; Biryukov, S.; Abbafati, C.; Abera, S.F.
Global, regional, and national prevalence of overweight and obesity in children and adults during 1980–2013: A systematic
analysis for the Global Burden of Disease Study 2013. Lancet 2014,384, 766–781. [CrossRef]
3.
Adam, R.; Karam, V.; Cailliez, V.; Grady, J.G.O.; Mirza, D.; Cherqui, D.; Klempnauer, J.; Salizzoni, M.; Pratschke, J.; Jamieson, N.;
et al. 2018 Annual Report of the European Liver Transplant Registry (ELTR)—50-year evolution of liver transplantation. Transpl.
Int. 2018,31, 1293–1317. [CrossRef]
4.
De Vries, Y.; von Meijenfeldt, F.A.; Porte, R.J. Post-transplant cholangiopathy: Classification, pathogenesis, and preventive
strategies. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2018,1864, 1507–1515. [CrossRef] [PubMed]
5.
Blok, J.J.; Detry, O.; Putter, H.; Rogiers, X.; Porte, R.J.; van Hoek, B.; Pirenne, J.; Metselaar, H.J.; Lerut, J.P.; Ysebaert, D.K.; et al.
Longterm results of liver transplantation from donation after circulatory death. Liver Transpl. 2016,22, 1107–1114. [CrossRef]
6.
Foley, D.P.; Fernandez, L.A.; Leverson, G.; Anderson, M.; Mezrich, J.; Sollinger, H.W.; D’Alessandro, A. Biliary Complications
after Liver Transplantation from Donation after Cardiac Death Donors: An Analysis of Risk Factors and Long-term Outcomes
From a Single Center. Ann. Surg. 2011,253, 817–825. [CrossRef] [PubMed]
7.
Banales, J.-M.; Prieto, J.; Medina, J.-F. Cholangiocyte anion exchange and biliary bicarbonate excretion. World J. Gastroenterol.
2006,12, 3496–3511. [CrossRef] [PubMed]
8.
De Jong, I.E.M.; Matton, A.P.M.; van Praagh, J.B.; van Haaften, W.T.; Wiersema-Buist, J.; van Wijk, L.A.; Oosterhuis, D.; Iswandana,
R.; Suriguga, S.; Overi, D.; et al. Peribiliary Glands Are Key in Regeneration of the Human Biliary Epithelium After Severe Bile
Duct Injury. Hepatology 2019,69, 1719–1734. [CrossRef] [PubMed]
9.
Banales, J.M.; Huebert, R.C.; Karlsen, T.; Strazzabosco, M.; LaRusso, N.F.; Gores, G.J. Cholangiocyte pathobiology. Nat. Rev.
Gastroenterol. Hepatol. 2019,16, 269–281. [CrossRef] [PubMed]
10.
Eshmuminov, D.; Becker, D.; Bautista Borrego, L.; Hefti, M.; Schuler, M.J.; Hagedorn, C.; Muller, X.; Mueller, M.; Onder, C.;
Graf, R.; et al. An integrated perfusion machine preserves injured human livers for 1 week. Nat. Biotechnol.
2020
,38, 189–198.
[CrossRef] [PubMed]
11.
Verstegen, M.M.A.; Mezzanotte, L.; Ridwan, R.Y.; Wang, K.; de Haan, J.; Schurink, I.J.; Sierra Parraga, J.M.; Hoogduijn, M.; Kessler,
B.M.; Huang, H.; et al. First Report on Ex Vivo Delivery of Paracrine Active Human Mesenchymal Stromal Cells to Liver Grafts
During Machine Perfusion. Transplantation 2020,104, e5–e7. [CrossRef] [PubMed]
12.
Schurink, I.J.; Willemse, J.; Verstegen, M.M.A.; van Der Laan, L.J.W.; de Jonge, J. Long-Term Perfusion of the Liver Outside the
Body: Warming Up for Ex Vivo Therapies? Hepatology 2020,72, 1485–1487. [CrossRef] [PubMed]
13. Joplin, R. Isolation and culture of biliary epithelial cells. Gut 1994,35, 875–878. [CrossRef]
14.
De Assuncao, T.M.; Sun, Y.; Jalan-Sakrikar, N.; Drinane, M.C.; Huang, B.Q.; Li, Y.; Davila, J.I.; Wang, R.; O’Hara, S.P.; Lomberk,
G.A.; et al. Development and characterization of human-induced pluripotent stem cell-derived cholangiocytes. Lab. Investig.
2015,95, 684–696. [CrossRef]
15.
Sampaziotis, F.; Segeritz, C.P.; Vallier, L. Potential of human induced pluripotent stem cells in studies of liver disease. Hepatology
2015,62, 303–311. [CrossRef] [PubMed]
16.
Sampaziotis, F.; de Brito, M.C.; Geti, I.; Bertero, A.; Hannan, N.R.; Vallier, L. Directed differentiation of human induced pluripotent
stem cells into functional cholangiocyte-like cells. Nat. Protoc. 2017,12, 814–827. [CrossRef] [PubMed]
17.
Bedel, A.; Beliveau, F.; Lamrissi-Garcia, I.; Rousseau, B.; Moranvillier, I.; Rucheton, B.; Guyonnet-Dupérat, V.; Cardinaud, B.; de
Verneuil, H.; Moreau-Gaudry, F.; et al. Preventing Pluripotent Cell Teratoma in Regenerative Medicine Applied to Hematology
Disorders. Stem Cells Transl. Med. 2017,6, 382–393. [CrossRef] [PubMed]
Bioengineering 2022,9, 110 12 of 17
18.
Lee, M.-O.; Moon, S.H.; Jeong, H.-C.; Yi, J.-Y.; Lee, T.-H.; Shim, S.H.; Rhee, Y.-H.; Lee, S.-H.; Oh, S.-J.; Lee, M.-Y.; et al. Inhibition
of pluripotent stem cell-derived teratoma formation by small molecules. Proc. Natl. Acad. Sci. USA
2013
,110, E3281–E3290.
[CrossRef] [PubMed]
19.
Xu, C.; Inokuma, M.S.; Denham, J.; Golds, K.; Kundu, P.; Gold, J.D.; Carpenter, M.K. Feeder-free growth of undifferentiated
human embryonic stem cells. Nat. Biotechnol. 2001,19, 971–974. [CrossRef]
20.
Marsee, A.; Roos, F.J.M.; Verstegen, M.M.A.; Marsee, A.; Roos, F.; Verstegen, M.; Clevers, H.; Vallier, L.; Takebe, T.; Huch, M.; et al.
Building consensus on definition and nomenclature of hepatic, pancreatic, and biliary organoids. Cell Stem Cell
2021
,28, 816–832.
[CrossRef] [PubMed]
21.
Huch, M.; Gehart, H.; van Boxtel, R.; Hamer, K.; Blokzijl, F.; Verstegen, M.M.; Ellis, E.; van Wenum, M.; Fuchs, S.A.; de Ligt, J.;
et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell 2015,160, 299–312. [CrossRef]
22.
Barker, N.; van Es, J.H.; Kuipers, J.; Kujala, P.; van den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters,
P.J.; et al. Identification of Stem Cells in Small Intestine and Colon by Marker Gene Lgr5. Nature
2007
,449, 1003–1007. Available
online: https://www.nature.com/nature/journal/v449/n7165/suppinfo/nature06196_S1.html (accessed on 15 January 2022).
[CrossRef]
23.
Sato, T.; Vries, R.G.; Snippert, H.J.; Van de Wetering, M.; Barker, N.; Stange, D.E.; Van Es, J.H.; Abo, A.; Kujala, P.; Peters, P.J.
Single Lgr5 stem cells build crypt villus structures
in vitro
without a mesenchymal niche. Nature
2009
,459, 262–265. [CrossRef]
[PubMed]
24.
Huch, M.; Dorrell, C.; Boj, S.F.; van Es, J.H.; Li, V.S.W.; van de Wetering, M.; Sato, T.; Hamer, K.; Sasaki, N.; Finegold, M.J.; et al. In
Vitro Expansion of Single Lgr5
+
Liver Stem Cells Induced by Wnt-Driven Regeneration. Nature
2013
,494, 247–250. Available
online: https://www.nature.com/nature/journal/v494/n7436/abs/nature11826.html#supplementary-information (accessed
on 15 January 2022). [CrossRef] [PubMed]
25.
Willemse, J.; Lieshout, R.; van der Laan, L.J.W.; Verstegen, M.M.A. From organoids to organs: Bioengineering liver grafts from
hepatic stem cells and matrix. Best Pract. Res. Clin. Gastroenterol. 2017,31, 151–159. [CrossRef] [PubMed]
26.
Schneeberger, K.; Sánchez-Romero, N.; Ye, S.; van Steenbeek, F.G.; Oosterhoff, L.A.; Pla Palacin, I.; Chen, C.; van Wolferen, M.E.;
van Tienderen, G.; Lieshout, R.; et al. Large-Scale Production of LGR5-Positive Bipotential Human Liver Stem Cells. Hepatology
2019,72, 257–270. [CrossRef] [PubMed]
27.
Verstegen, M.M.A.; Roos, F.J.M.; Burka, K.; Gehart, H.; Jager, M.; de Wolf, M.; Bijvelds, M.J.C.; de Jonge, H.R.; Ardisasmita, A.I.;
van Huizen, N.A.; et al. Human extrahepatic and intrahepatic cholangiocyte organoids show region-specific differentiation
potential and model cystic fibrosis-related bile duct disease. Sci. Rep. 2020,10, 21900. [CrossRef]
28.
Lugli, N.; Kamileri, I.; Keogh, A.; Malinka, T.; Sarris, M.E.; Talianidis, I.; Schaad, O.; Candinas, D.; Stroka, D.; Halazonetis, T.D.
R-spondin 1 and noggin facilitate expansion of resident stem cells from non-damaged gallbladders. EMBO Rep.
2016
,17, 769–779.
[CrossRef]
29.
Sampaziotis, F.; Justin, A.W.; Tysoe, O.C.; Sawiak, S.; Godfrey, E.M.; Upponi, S.S.; Gieseck, R.L., 3rd; de Brito, M.C.; Berntsen,
N.L.; Gómez-Vázquez, M.J.; et al. Reconstruction of the mouse extrahepatic biliary tree using primary human extrahepatic
cholangiocyte organoids. Nat. Med. 2017,23, 954–963. [CrossRef] [PubMed]
30.
Roos, F.J.M.; Wu, H.; Willemse, J.; Lieshout, R.; Albarinos, L.A.M.; Kan, Y.-Y.; Poley, J.-W.; Bruno, M.J.; de Jonge, J.; Bártfai, R.; et al.
Cholangiocyte organoids from human bile retain a local phenotype and can repopulate bile ducts
in vitro
.Clin. Transl. Med.
2021
,
11, e566. [CrossRef] [PubMed]
31.
Rimland, C.A.; Tilson, S.G.; Morell, C.M.; Tomaz, R.A.; Lu, W.Y.; Adams, S.E.; Georgakopoulos, N.; Otaizo-Carrasquero, F.; Myers,
T.G.; Ferdinand, J.R.; et al. Regional differences in human biliary tissues and corresponding
in vitro
derived organoids. Hepatology
2020,73, 247–267. [CrossRef]
32.
Chia, S.-M.; Leong, K.W.; Li, J.; Xu, X.; Zeng, K.; Er, P.-N.; Gao, S.; Yu, H. Hepatocyte encapsulation for enhanced cellular functions.
Tissue Eng. 2000,6, 481–495. [CrossRef]
33.
Iansante, V.; Mitry, R.R.; Filippi, C.; Fitzpatrick, E.; Dhawan, A. Human hepatocyte transplantation for liver disease: Current
status and future perspectives. Pediatric Res. 2018,83, 232–240. [CrossRef] [PubMed]
34.
Dhawan, A.; Puppi, J.; Hughes, R.D.; Mitry, R.R. Human hepatocyte transplantation: Current experience and future challenges.
Nat. Rev. Gastroenterol. Hepatol. 2010,7, 288–298. [CrossRef]
35.
Sampaziotis, F.; Muraro, D.; Tysoe, O.C.; Sawiak, S.; Beach, T.E.; Godfrey, E.M.; Upponi, S.S.; Brevini, T.; Wesley, B.T.; Garcia-
Bernardo, J.; et al. Cholangiocyte organoids can repair bile ducts after transplantation in the human liver. Science
2021
,371,
839–846. [CrossRef] [PubMed]
36.
Kleinman, H.K.; McGarvey, M.L.; Hassell, J.R.; Star, V.L.; Cannon, F.B.; Laurie, G.W.; Martin, G.R. Basement membrane complexes
with biological activity. Biochemistry 1986,25, 312–318. [CrossRef]
37.
Kleinman, H.K. Preparation of Basement Membrane Components from EHS Tumors. Curr. Protoc. Cell Biol.
1998
,1. [CrossRef]
[PubMed]
38.
Hughes, C.S.; Postovit, L.M.; Lajoie, G.A. Matrigel: A complex protein mixture required for optimal growth of cell culture.
Proteomics 2010,10, 1886–1890. [CrossRef] [PubMed]
39.
Vukicevic, S.; Kleinman, H.K.; Luyten, F.P.; Roberts, A.B.; Roche, N.S.; Reddi, A.H. Identification of multiple active growth factors
in basement membrane Matrigel suggests caution in interpretation of cellular activity related to extracellular matrix components.
Exp. Cell Res. 1992,202, 1–8. [CrossRef]
Bioengineering 2022,9, 110 13 of 17
40. Kibbey, M.C. Maintenance of the EHS sarcoma and Matrigel preparation. J. Tissue Cult. Methods 1994,16, 227–230. [CrossRef]
41.
Kobayashi, S.; Ito, E.; Honma, R.; Nojima, Y.; Shibuya, M.; Watanabe, S.; Maru, Y. Dynamic regulation of gene expression by the
Flt-1 kinase and Matrigel in endothelial tubulogenesis. Genomics 2004,84, 185–192. [CrossRef] [PubMed]
42. Tahergorabi, Z.; Khazaei, M. A review on angiogenesis and its assays. Iran. J. Basic Med. Sci. 2012,15, 1110–1126. [PubMed]
43.
Zaman, M.H.; Trapani, L.M.; Sieminski, A.L.; MacKellar, D.; Gong, H.; Kamm, R.D.; Wells, A.; Lauffenburger, D.A.; Matsudaira, P.
Migration of tumor cells in 3D matrices is governed by matrix stiffness along with cell-matrix adhesion and proteolysis. Proc.
Natl. Acad. Sci. USA 2006,103, 10889–10894. [CrossRef] [PubMed]
44.
Hagbard, L.; Cameron, K.; August, P.; Penton, C.; Parmar, M.; Hay, D.C.; Kallur, T. Developing defined substrates for stem cell
culture and differentiation. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2018,373, 20170230. [CrossRef] [PubMed]
45.
Uemura, M.; Refaat, M.M.; Shinoyama, M.; Hayashi, H.; Hashimoto, N.; Takahashi, J. Matrigel supports survival and neuronal
differentiation of grafted embryonic stem cell-derived neural precursor cells. J. Neurosci. Res.
2010
,88, 542–551. [CrossRef]
[PubMed]
46.
Kleinman, H.K.; Martin, G.R. Matrigel: Basement membrane matrix with biological activity. Semin. Cancer Biol.
2005
,15, 378–386.
[CrossRef]
47.
Benton, G.; Kleinman, H.K.; George, J.; Arnaoutova, I. Multiple uses of basement membrane-like matrix (BME/Matrigel)
in vitro
and in vivo with cancer cells. Int. J. Cancer 2011,128, 1751–1757. [CrossRef] [PubMed]
48.
Li, Q.; Uygun, B.E.; Geerts, S.; Ozer, S.; Scalf, M.; Gilpin, S.E.; Ott, H.C.; Yarmush, M.L.; Smith, L.M.; Welham, N.V.; et al.
Proteomic analysis of naturally-sourced biological scaffolds. Biomaterials 2016,75, 37–46. [CrossRef] [PubMed]
49.
Mak, K.M.; Mei, R. Basement Membrane Type IV Collagen and Laminin: An Overview of Their Biology and Value as Fibrosis
Biomarkers of Liver Disease. Anat. Rec. 2017,300, 1371–1390. [CrossRef]
50.
Williams, M.J.; Clouston, A.D.; Forbes, S.J. Links between hepatic fibrosis, ductular reaction, and progenitor cell expansion.
Gastroenterology 2014,146, 349–356. [CrossRef] [PubMed]
51.
Benyon, R.C.; Arthur, M.J. Extracellular matrix degradation and the role of hepatic stellate cells. Semin Liver Dis.
2001
,21, 373–384.
[CrossRef] [PubMed]
52.
Dubuquoy, L.; Louvet, A.; Lassailly, G.; Truant, S.; Boleslawski, E.; Artru, F.; Maggiotto, F.; Gantier, E.; Buob, D.; Leteurtre, E.;
et al. Progenitor cell expansion and impaired hepatocyte regeneration in explanted livers from alcoholic hepatitis. Gut
2015
,64,
1949–1960. [CrossRef] [PubMed]
53.
Klaas, M.; Kangur, T.; Viil, J.; Mäemets-Allas, K.; Minajeva, A.; Vadi, K.; Antsov, M.; Lapidus, N.; Järvekülg, M.; Jaks, V. The
alterations in the extracellular matrix composition guide the repair of damaged liver tissue. Sci. Rep.
2016
,6, 27398. [CrossRef]
[PubMed]
54.
Lorenzini, S.; Bird, T.G.; Boulter, L.; Bellamy, C.; Samuel, K.; Aucott, R.; Clayton, E.; Andreone, P.; Bernardi, M.; Golding, M.; et al.
Characterisation of a stereotypical cellular and extracellular adult liver progenitor cell niche in rodents and diseased human liver.
Gut 2010,59, 645–654. [CrossRef] [PubMed]
55.
Zhu, C.; Coombe, D.R.; Zheng, M.H.; Yeoh, G.C.; Li, L. Liver progenitor cell interactions with the extracellular matrix. J. Tissue
Eng. Regen Med. 2013,7, 757–766. [CrossRef] [PubMed]
56. Yue, B. Biology of the extracellular matrix: An overview. J. Glaucoma 2014,23, S20–S23. [CrossRef]
57.
Bomo, J.; Ezan, F.; Tiaho, F.; Bellamri, M.; Langouët, S.; Theret, N.; Baffet, G. Increasing 3D Matrix Rigidity Strengthens
Proliferation and Spheroid Development of Human Liver Cells in a Constant Growth Factor Environment. J. Cell Biochem.
2016
,
117, 708–720. [CrossRef] [PubMed]
58. Hynes, R.O. The extracellular matrix: Not just pretty fibrils. Science 2009,326, 1216–1219. [CrossRef]
59.
Adams, R.H.; Alitalo, K. Molecular regulation of angiogenesis and lymphangiogenesis. Nat. Rev. Mol. Cell Biol.
2007
,8, 464–478.
[CrossRef] [PubMed]
60.
Gerhardt, H.; Golding, M.; Fruttiger, M.; Ruhrberg, C.; Lundkvist, A.; Abramsson, A.; Jeltsch, M.; Mitchell, C.; Alitalo, K.;
Shima, D. VEGF guides angiogenic sprouting utilizing endothelial tip cell filopodia. J. Cell Biol.
2003
,161, 1163–1177. [CrossRef]
[PubMed]
61.
Ruhrberg, C.; Gerhardt, H.; Golding, M.; Watson, R.; Ioannidou, S.; Fujisawa, H.; Betsholtz, C.; Shima, D.T. Spatially restricted
patterning cues provided by heparin-binding VEGF-A control blood vessel branching morphogenesis. Genes Dev.
2002
,16,
2684–2698. [CrossRef] [PubMed]
62.
Park, J.E.; Keller, G.A.; Ferrara, N. The vascular endothelial growth factor (VEGF) isoforms: Differential deposition into the
subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol. Biol. Cell
1993
,4, 1317–1326. [CrossRef]
[PubMed]
63.
Fausto, N.; Laird, A.D.; Webber, E.M. Liver regeneration. 2. Role of growth factors and cytokines in hepatic regeneration. FASEB
J. 1995,9, 1527–1536. [CrossRef]
64.
Greenhough, S.; Medine, C.N.; Hay, D.C. Pluripotent stem cell derived hepatocyte like cells and their potential in toxicity
screening. Toxicology 2010,278, 250–255. [CrossRef] [PubMed]
65.
Eyckmans, J.; Boudou, T.; Yu, X.; Chen, C.S. A hitchhiker’s guide to mechanobiology. Dev. Cell
2011
,21, 35–47. [CrossRef]
[PubMed]
66. Wells, R.G. The role of matrix stiffness in regulating cell behavior. Hepatology 2008,47, 1394–1400. [CrossRef] [PubMed]
Bioengineering 2022,9, 110 14 of 17
67.
Skardal, A.; Mack, D.; Atala, A.; Soker, S. Substrate elasticity controls cell proliferation, surface marker expression and motile
phenotype in amniotic fluid-derived stem cells. J. Mech. Behav. Biomed. Mater. 2013,17, 307–316. [CrossRef]
68.
Engler, A.J.; Sen, S.; Sweeney, H.L.; Discher, D.E. Matrix Elasticity Directs Stem Cell Lineage Specification. Cell
2006
,126, 677–689.
[CrossRef] [PubMed]
69.
Lozoya, O.A.; Wauthier, E.; Turner, R.A.; Barbier, C.; Prestwich, G.D.; Guilak, F.; Superfine, R.; Lubkin, S.R.; Reid, L.M. Regulation
of hepatic stem/progenitor phenotype by microenvironment stiffness in hydrogel models of the human liver stem cell niche.
Biomaterials 2011,32, 7389–7402. [CrossRef] [PubMed]
70.
Hansen, L.K.; Wilhelm, J.; Fassett, J.T. Regulation of hepatocyte cell cycle progression and differentiation by type I collagen
structure. Curr. Top. Dev. Biol. 2006,72, 205–236. [PubMed]
71.
Desai, S.S.; Tung, J.C.; Zhou, V.X.; Grenert, J.P.; Malato, Y.; Rezvani, M.; Español-Suñer, R.; Willenbring, H.; Weaver, V.M.; Chang,
T.T. Physiological ranges of matrix rigidity modulate primary mouse hepatocyte function in part through hepatocyte nuclear
factor 4 alpha. Hepatology 2016,64, 261–275. [CrossRef] [PubMed]
72.
Baiocchini, A.; Montaldo, C.; Conigliaro, A.; Grimaldi, A.; Correani, V.; Mura, F.; Ciccosanti, F.; Rotiroti, N.; Brenna, A.;
Montalbano, M.; et al. Extracellular Matrix Molecular Remodeling in Human Liver Fibrosis Evolution. PLoS ONE
2016
,11,
e0151736. [CrossRef] [PubMed]
73. Peppas, N.A. Hydrogels in Medicine and Pharmacy: Fundamentals; CRC Press: Boca Raton, FL, USA, 2019; Volume 1.
74.
Caló, E.; Khutoryanskiy, V.V. Biomedical applications of hydrogels: A review of patents and commercial products. Eur. Polym. J.
2015,65, 252–267. [CrossRef]
75.
Curvello, R.; Raghuwanshi, V.S.; Garnier, G. Engineering nanocellulose hydrogels for biomedical applications. Adv. Colloid
Interface Sci. 2019,267, 47–61. [CrossRef]
76.
Ye, S.; Boeter, J.W.B.; Penning, L.C.; Spee, B.; Schneeberger, K. Hydrogels for Liver Tissue Engineering. Bioengineering
2019
,6, 59.
[CrossRef] [PubMed]
77.
Broguiere, N.; Isenmann, L.; Hirt, C.; Ringel, T.; Placzek, S.; Cavalli, E.; Ringnalda, F.; Villiger, L.; Züllig, R.; Lehmann, R.; et al.
Growth of Epithelial Organoids in a Defined Hydrogel. Adv. Mater. 2018,30, 1801621. [CrossRef]
78.
Gjorevski, N.; Sachs, N.; Manfrin, A.; Giger, S.; Bragina, M.E.; Ordóñez-Morán, P.; Clevers, H.; Lutolf, M.P. Designer matrices for
intestinal stem cell and organoid culture. Nature 2016,539, 560–564. [CrossRef]
79.
Sorrentino, G.; Rezakhani, S.; Yildiz, E.; Nuciforo, S.; Heim, M.H.; Lutolf, M.P.; Schoonjans, K. Mechano-modulatory synthetic
niches for liver organoid derivation. Nat. Commun. 2020,11, 3416. [CrossRef] [PubMed]
80.
Ye, S.; Boeter, J.W.B.; Mihajlovic, M.; van Steenbeek, F.G.; van Wolferen, M.E.; Oosterhoff, L.A.; Marsee, A.; Caiazzo, M.; van der
Laan, L.J.W.; Penning, L.C.; et al. A Chemically Defined Hydrogel for Human Liver Organoid Culture. Adv. Funct. Mater.
2020
,
30, 2000893. [CrossRef] [PubMed]
81.
Krüger, M.; Oosterhoff, L.A.; van Wolferen, M.E.; Schiele, S.A.; Walther, A.; Geijsen, N.; De Laporte, L.; van der Laan, L.J.W.; Kock,
L.M.; Spee, B. Cellulose Nanofibril Hydrogel Promotes Hepatic Differentiation of Human Liver Organoids. Adv. Healthc. Mater.
2020,9, 1901658. [CrossRef] [PubMed]
82.
Ranga, A.; Gobaa, S.; Okawa, Y.; Mosiewicz, K.; Negro, A.; Lutolf, M.P. 3D niche microarrays for systems-level analyses of cell
fate. Nat. Commun. 2014,5, 4324. [CrossRef] [PubMed]
83.
Saldin, L.T.; Cramer, M.C.; Velankar, S.S.; White, L.J.; Badylak, S.F. Extracellular matrix hydrogels from decellularized tissues:
Structure and function. Acta Biomater. 2017,49, 1–15. [CrossRef] [PubMed]
84.
Shupe, T.; Williams, M.; Brown, A.; Willenberg, B.; Petersen, B.E. Method for the decellularization of intact rat liver. Organogenesis
2010,6, 134–136. [CrossRef] [PubMed]
85.
Ren, H.; Shi, X.; Tao, L.; Xiao, J.; Han, B.; Zhang, Y.; Yuan, X.; Ding, Y. Evaluation of two decellularization methods in the
development of a whole-organ decellularized rat liver scaffold. Liver Int. 2013,33, 448–458. [CrossRef]
86.
Struecker, B.; Butter, A.; Hillebrandt, K.; Polenz, D.; Reutzel-Selke, A.; Tang, P.; Lippert, S.; Leder, A.; Rohn, S.; Geisel, D. Improved
rat liver decellularization by arterial perfusion under oscillating pressure conditions. J. Tissue Eng. Regen. Med.
2017
,11, 531–541.
[CrossRef] [PubMed]
87.
Baptista, P.M.; Vyas, D.; Moran, E.; Wang, Z.; Soker, S. Human liver bioengineering using a whole liver decellularized bioscaffold.
In Organ Regeneration; Springer: Berlin/Heidelberg, Germany, 2013; pp. 289–298.
88.
Alevra Sarika, N.; Payen, V.L.; Fléron, M.; Ravau, J.; Brusa, D.; Najimi, M.; Pauw, E.D.; Eppe, G.; Mazzucchelli, G.; Sokal, E.M.;
et al. Human Liver-Derived Extracellular Matrix for the Culture of Distinct Human Primary Liver Cells. Cells
2020
,9, 1357.
[CrossRef] [PubMed]
89.
Moulisová, V.; Jiˇrík, M.; Schindler, C.; ˇ
Cervenková, L.; Pálek, R.; Rosendorf, J.; Arlt, J.; Bolek, L.; Š˚usová, S.; Nietzsche, S.; et al.
Novel morphological multi-scale evaluation system for quality assessment of decellularized liver scaffolds. J. Tissue Eng.
2020
,11.
[CrossRef] [PubMed]
90.
Barakat, O.; Abbasi, S.; Rodriguez, G.; Rios, J.; Wood, R.P.; Ozaki, C.; Holley, L.S.; Gauthier, P.K. Use of decellularized porcine
liver for engineering humanized liver organ. J. Surg. Res. 2012,173, e11–e25. [CrossRef] [PubMed]
91.
Lin, P.; Chan, W.C.; Badylak, S.F.; Bhatia, S.N. Assessing porcine liver-derived biomatrix for hepatic tissue engineering. Tissue
Eng. 2004,10, 1046–1053. [CrossRef] [PubMed]
Bioengineering 2022,9, 110 15 of 17
92.
Struecker, B.; Hillebrandt, K.H.; Voitl, R.; Butter, A.; Schmuck, R.B.; Reutzel-Selke, A.; Geisel, D.; Joehrens, K.; Pickerodt, P.A.;
Raschzok, N.; et al. Porcine liver decellularization under oscillating pressure conditions: A technical refinement to improve the
homogeneity of the decellularization process. Tissue Eng. Part C Methods 2015,21, 303–313. [CrossRef] [PubMed]
93.
Wu, Q.; Bao, J.; Zhou, Y.-j.; Wang, Y.-j.; Du, Z.-g.; Shi, Y.-j.; Li, L.; Bu, H. Optimizing perfusion-decellularization methods of
porcine livers for clinical-scale whole-organ bioengineering. BioMed Res. Int. 2015,2015, 785474. [CrossRef]
94.
Willemse, J.; Verstegen, M.M.A.; Vermeulen, A.; Schurink, I.J.; Roest, H.P.; van der Laan, L.J.W.; de Jonge, J. Fast, robust and
effective decellularization of whole human livers using mild detergents and pressure controlled perfusion. Mater. Sci. Eng. C
2020,108, 110200. [CrossRef] [PubMed]
95.
Verstegen, M.M.A.; Willemse, J.; van den Hoek, S.; Kremers, G.J.; Luider, T.M.; van Huizen, N.A.; Willemssen, F.; Metselaar,
H.J.; JNM, I.J.; van der Laan, L.J.W.; et al. Decellularization of Whole Human Liver Grafts Using Controlled Perfusion for
Transplantable Organ Bioscaffolds. Stem Cells Dev. 2017,26, 1304–1315. [CrossRef] [PubMed]
96.
Mazza, G.; Rombouts, K.; Hall, A.R.; Urbani, L.; Luong, T.V.; Al-Akkad, W.; Longato, L.; Brown, D.; Maghsoudlou, P.; Dhillon,
A.P. Decellularized human liver as a natural 3D-scaffold for liver bioengineering and transplantation. Sci. Rep.
2015
,5, 13079.
[CrossRef]
97.
Badylak, S.F.; Taylor, D.; Uygun, K. Whole-organ tissue engineering: Decellularization and recellularization of three-dimensional
matrix scaffolds. Annu Rev. Biomed. Eng. 2011,13, 27–53. [CrossRef]
98. Gilbert, T.W. Strategies for tissue and organ decellularization. J. Cell. Biochem. 2012,113, 2217–2222. [CrossRef] [PubMed]
99.
Gilpin, A.; Yang, Y. Decellularization Strategies for Regenerative Medicine: From Processing Techniques to Applications. BioMed
Res. Int. 2017,2017, 9831534. [CrossRef] [PubMed]
100.
Coronado, R.E.; Somaraki-Cormier, M.; Natesan, S.; Christy, R.J.; Ong, J.L.; Halff, G.A. Decellularization and Solubilization of
Porcine Liver for Use as a Substrate for Porcine Hepatocyte Culture: Method Optimization and Comparison. Cell Transpl.
2017
,
26, 1840–1854. [CrossRef]
101.
Spang, M.T.; Christman, K.L. Extracellular matrix hydrogel therapies:
In vivo
applications and development. Acta Biomater.
2018
,
68, 1–14. [CrossRef] [PubMed]
102.
Gazia, C.; Tamburrini, R.; Asthana, A.; Chaimov, D.; Muir, S.M.; Marino, D.I.; Delbono, L.; Villani, V.; Perin, L.; Di Nardo, P.; et al.
Extracellular matrix-based hydrogels obtained from human tissues: A work still in progress. Curr. Opin. Organ. Transpl.
2019
,24,
604–612. [CrossRef]
103.
Meyer, M. Processing of collagen based biomaterials and the resulting materials properties. BioMed. Eng. OnLine
2019
,18, 24.
[CrossRef]
104.
Jafari, H.; Lista, A.; Siekapen, M.M.; Ghaffari-Bohlouli, P.; Nie, L.; Alimoradi, H.; Shavandi, A. Fish collagen: Extraction,
characterization, and applications for biomaterials engineering. Polymers 2020,12, 2230. [CrossRef]
105. Hulmes, D.J.S. Collagen diversity, synthesis and assembly. In Collagen; Springer: Berlin/Heidelberg, Germany, 2008; pp. 15–47.
106.
Kadler, K.E.; Holmes, D.F.; Trotter, J.A.; Chapman, J.A. Collagen fibril formation. Biochem. J.
1996
,316 Pt 1, 1–11. [CrossRef]
[PubMed]
107.
Lee, J.S.; Shin, J.; Park, H.M.; Kim, Y.G.; Kim, B.G.; Oh, J.W.; Cho, S.W. Liver extracellular matrix providing dual functions of two-
dimensional substrate coating and three-dimensional injectable hydrogel platform for liver tissue engineering. Biomacromolecules
2014,15, 206–218. [CrossRef]
108.
Brightman, A.O.; Rajwa, B.P.; Sturgis, J.E.; McCallister, M.E.; Robinson, J.P.; Voytik-Harbin, S.L. Time-lapse confocal reflection
microscopy of collagen fibrillogenesis and extracellular matrix assembly in vitro. Biopolymers 2000,54, 222–234. [CrossRef]
109.
Skardal, A.; Devarasetty, M.; Kang, H.-W.; Mead, I.; Bishop, C.; Shupe, T.; Lee, S.J.; Jackson, J.; Yoo, J.; Soker, S.; et al. A hydrogel
bioink toolkit for mimicking native tissue biochemical and mechanical properties in bioprinted tissue constructs. Acta Biomater.
2015,25, 24–34. [CrossRef] [PubMed]
110.
Loneker, A.E.; Faulk, D.M.; Hussey, G.S.; D’Amore, A.; Badylak, S.F. Solubilized liver extracellular matrix maintains primary rat
hepatocyte phenotype in-vitro. J. Biomed. Mater. Res. Part A 2016,104, 957–965. [CrossRef]
111.
Bual, R.P.; Ijima, H. Intact extracellular matrix component promotes maintenance of liver-specific functions and larger aggregates
formation of primary rat hepatocytes. Regen. Ther. 2019,11, 258–268. [CrossRef] [PubMed]
112.
Nakamura, S.; Ijima, H. Solubilized matrix derived from decellularized liver as a growth factor-immobilizable scaffold for
hepatocyte culture. J. Biosci. Bioeng. 2013,116, 746–753. [CrossRef]
113.
Lewis, P.L.; Su, J.; Yan, M.; Meng, F.; Glaser, S.S.; Alpini, G.D.; Green, R.M.; Sosa-Pineda, B.; Shah, R.N. Complex bile duct network
formation within liver decellularized extracellular matrix hydrogels. Sci. Rep. 2018,8, 12220. [CrossRef] [PubMed]
114.
Saheli, M.; Sepantafar, M.; Pournasr, B.; Farzaneh, Z.; Vosough, M.; Piryaei, A.; Baharvand, H. Three-dimensional liver-derived
extracellular matrix hydrogel promotes liver organoids function. J. Cell Biochem. 2018,119, 4320–4333. [CrossRef] [PubMed]
115.
Sellaro, T.L.; Ravindra, A.K.; Stolz, D.B.; Badylak, S.F. Maintenance of hepatic sinusoidal endothelial cell phenotype
in vitro
using
organ-specific extracellular matrix scaffolds. Tissue Eng. 2007,13, 2301–2310. [CrossRef] [PubMed]
116.
Park, K.-M.; Hussein, K.H.; Hong, S.-H.; Ahn, C.; Yang, S.-R.; Park, S.-M.; Kweon, O.-K.; Kim, B.-M.; Woo, H.-M. Decellularized
liver extracellular matrix as promising tools for transplantable bioengineered liver promotes hepatic lineage commitments of
induced pluripotent stem cells. Tissue Eng. Part A 2016,22, 449–460. [CrossRef] [PubMed]
Bioengineering 2022,9, 110 16 of 17
117.
Wang, B.; Jakus, A.E.; Baptista, P.M.; Soker, S.; Soto-Gutierrez, A.; Abecassis, M.M.; Shah, R.N.; Wertheim, J.A. Functional
Maturation of Induced Pluripotent Stem Cell Hepatocytes in Extracellular Matrix—A Comparative Analysis of Bioartificial Liver
Microenvironments. Stem Cells Transl. Med. 2016,5, 1257–1267. [CrossRef]
118.
Jaramillo, M.; Yeh, H.; Yarmush, M.L.; Uygun, B.E. Decellularized human liver extracellular matrix (hDLM)-mediated hepatic
differentiation of human induced pluripotent stem cells (hIPSCs). J. Tissue Eng. Regen Med.
2018
,12, e1962–e1973. [CrossRef]
[PubMed]
119.
Giobbe, G.G.; Crowley, C.; Luni, C.; Campinoti, S.; Khedr, M.; Kretzschmar, K.; De Santis, M.M.; Zambaiti, E.; Michielin, F.; Meran,
L.; et al. Extracellular matrix hydrogel derived from decellularized tissues enables endodermal organoid culture. Nat. Commun.
2019,10, 5658. [CrossRef] [PubMed]
120.
Lindberg, K.; Badylak, S.F. Porcine small intestinal submucosa (SIS): A bioscaffold supporting
in vitro
primary human epidermal
cell differentiation and synthesis of basement membrane proteins. Burns 2001,27, 254–266. [CrossRef]
121.
White, L.J.; Taylor, A.J.; Faulk, D.M.; Keane, T.J.; Saldin, L.T.; Reing, J.E.; Swinehart, I.T.; Turner, N.J.; Ratner, B.D.; Badylak, S.F.
The impact of detergents on the tissue decellularization process: A ToF-SIMS study. Acta Biomater.
2017
,50, 207–219. [CrossRef]
122.
Fernández-Pérez, J.; Ahearne, M. The impact of decellularization methods on extracellular matrix derived hydrogels. Sci. Rep.
2019,9, 14933. [CrossRef] [PubMed]
123.
Seif-Naraghi, S.B.; Salvatore, M.A.; Schup-Magoffin, P.J.; Hu, D.P.; Christman, K.L. Design and characterization of an injectable
pericardial matrix gel: A potentially autologous scaffold for cardiac tissue engineering. Tissue Eng. Part A
2010
,16, 2017–2027.
[CrossRef] [PubMed]
124.
Acun, A.; Oganesyan, R.; Uygun, K.; Yeh, H.; Yarmush, M.L.; Uygun, B.E. Liver donor age affects hepatocyte function through
age-dependent changes in decellularized liver matrix. Biomaterials 2021,270, 120689. [CrossRef] [PubMed]
125.
Rolewska, P.; Al-Robaiy, S.; Santos, A.N.; Simm, A.; Silber, R.-E.; Bartling, B. Age-related expression, enzymatic solubility and
modification with advanced glycation end-products of fibrillar collagens in mouse lung. Exp. Gerontol.
2013
,48, 29–37. [CrossRef]
[PubMed]
126.
Wilson, S.L.; Guilbert, M.; Sulé-Suso, J.; Torbet, J.; Jeannesson, P.; Sockalingum, G.D.; Yang, Y. A microscopic and macroscopic
study of aging collagen on its molecular structure, mechanical properties, and cellular response. FASEB J.
2014
,28, 14–25.
[CrossRef] [PubMed]
127.
Wood, L.K.; Kayupov, E.; Gumucio, J.P.; Mendias, C.L.; Claflin, D.R.; Brooks, S.V. Intrinsic stiffness of extracellular matrix
increases with age in skeletal muscles of mice. J. Appl. Physiol. 2014,117, 363–369. [CrossRef] [PubMed]
128.
Snedeker, J.G.; Gautieri, A. The role of collagen crosslinks in ageing and diabetes—The good, the bad, and the ugly. Muscles
Ligaments Tendons J. 2014,4, 303–308. [CrossRef]
129.
Williams, C.; Sullivan, K.; Black, L.D., 3rd. Partially Digested Adult Cardiac Extracellular Matrix Promotes Cardiomyocyte
Proliferation In Vitro. Adv. Healthc. Mater. 2015,4, 1545–1554. [CrossRef] [PubMed]
130.
Tottey, S.; Johnson, S.A.; Crapo, P.M.; Reing, J.E.; Zhang, L.; Jiang, H.; Medberry, C.J.; Reines, B.; Badylak, S.F. The effect of source
animal age upon extracellular matrix scaffold properties. Biomaterials 2011,32, 128–136. [CrossRef]
131.
Lada, E.; Anna, M.; Patrik, M.; Zbynek, T.; Miroslav, J.; Hynek, M.; Richard, P.; Sarah, L.; Vaclav, L. Porcine Liver Anatomy
Applied to Biomedicine. J. Surg. Res. 2020,250, 70–79. [CrossRef] [PubMed]
132.
Morris, A.H.; Chang, J.; Kyriakides, T.R. Inadequate processing of decellularized dermal matrix reduces cell viability
in vitro
and
increases apoptosis and acute inflammation in vivo. BioResearch Open Access 2016,5, 177–187. [CrossRef] [PubMed]
133.
Gilbert, T.W.; Freund, J.M.; Badylak, S.F. Quantification of DNA in Biologic Scaffold Materials. J. Surg. Res.
2009
,152, 135–139.
[CrossRef]
134.
Keane, T.J.; Londono, R.; Turner, N.J.; Badylak, S.F. Consequences of ineffective decellularization of biologic scaffolds on the host
response. Biomaterials 2012,33, 1771–1781. [CrossRef] [PubMed]
135.
Naso, F.; Gandaglia, A.; Bottio, T.; Tarzia, V.; Nottle, M.B.; d’Apice, A.J.F.; Cowan, P.J.; Cozzi, E.; Galli, C.; Lagutina, I. First
quantification of alpha-G al epitope in current glutaraldehyde-fixed heart valve bioprostheses. Xenotransplantation
2013
,20,
252–261. [CrossRef] [PubMed]
136.
Stone, K.R.; Abdel-Motal, U.M.; Walgenbach, A.W.; Turek, T.J.; Galili, U. Replacement of human anterior cruciate ligaments with
pig ligaments: A model for anti-non-gal antibody response in long-term xenotransplantation. Transplantation
2007
,83, 211–219.
[CrossRef]
137.
Morris, A.H.; Stamer, D.K.; Kyriakides, T.R. The host response to naturally-derived extracellular matrix biomaterials. Semin.
Immunol. 2017,29, 72–91. [CrossRef] [PubMed]
138.
Galili, U. Human Anti-Gal and Anti-Non-Gal Immune Response to Porcine Tissue Implants. In Host Response to Biomaterials;
Elsevier: Amsterdam, The Netherlands, 2015; pp. 239–267.
139.
Łopata, K.; Wojdas, E.; Nowak, R.; Łopata, P.; Mazurek, U. Porcine Endogenous Retrovirus (PERV)—Molecular Structure and
Replication Strategy in the Context of Retroviral Infection Risk of Human Cells. Front. Microbiol.
2018
,9, 730. [CrossRef]
[PubMed]
140.
Kallenbach, K.; Leyh, R.G.; Lefik, E.; Walles, T.; Wilhelmi, M.; Cebotari, S.; Schmiedl, A.; Haverich, A.; Mertsching, H. Guided
tissue regeneration: Porcine matrix does not transmit PERV. Biomaterials 2004,25, 3613–3620. [CrossRef] [PubMed]
141.
Godehardt, A.W.; Ramm, R.; Gulich, B.; Tönjes, R.R.; Hilfiker, A. Decellularized pig pulmonary heart valves—Depletion of nucleic
acids measured by proviral PERV pol. Xenotransplantation 2020,27, e12565. [CrossRef]
Bioengineering 2022,9, 110 17 of 17
142.
Soroka, C.J.; Assis, D.N.; Alrabadi, L.S.; Roberts, S.; Cusack, L.; Jaffe, A.B.; Boyer, J.L. Bile-Derived Organoids From Patients
With Primary Sclerosing Cholangitis Recapitulate Their Inflammatory Immune Profile. Hepatology
2018
,70, 871–882. [CrossRef]
[PubMed]
143.
Broutier, L.; Mastrogiovanni, G.; Verstegen, M.; Francies, H.; Gavarró, L.; Bradshaw, C.; Allen, G.; Arnes-Benito, R.; Sidorova, O.;
Gaspersz, M.; et al. Human Primary Liver Cancer -derived Organoid Cultures for disease modelling and drug screening. Nat.
Med. 2017,23, nm.4438. [CrossRef] [PubMed]
144.
van Tienderen, G.; Koerkamp, G.; Ijzermans; Laan, V.; Verstegen, M. Recreating Tumour Complexity in a Dish: Organoid Models
to Study Liver Cancer Cells and their Extracellular Environment. Cancers 2019,11, 1706. [CrossRef] [PubMed]
145. Nuciforo, S.; Heim, M.H. Organoids to model liver disease. JHEP Rep. 2021,3, 100198. [CrossRef] [PubMed]
146.
Vacanti, J.P.; Langer, R. Tissue engineering: The design and fabrication of living replacement devices for surgical reconstruction
and transplantation. Lancet 1999,354, S32–S34. [CrossRef]
147. Jain, E.; Damania, A.; Kumar, A. Biomaterials for liver tissue engineering. Hepatol. Int. 2014,8, 185–197. [CrossRef] [PubMed]
148.
Mazza, G.; Al-Akkad, W.; Rombouts, K.; Pinzani, M. Liver tissue engineering: From implantable tissue to whole organ engineering.
Hepatol. Commun. 2018,2, 131–141. [CrossRef] [PubMed]
149. Murphy, S.V.; Atala, A. 3D bioprinting of tissues and organs. Nat. Biotechnol. 2014,32, 773–785. [CrossRef]
150.
Jung, J.P.; Bhuiyan, D.B.; Ogle, B.M. Solid organ fabrication: Comparison of decellularization to 3D bioprinting. Biomater. Res.
2016,20, 27. [CrossRef]
151.
Palakkan, A.A.; Hay, D.C.; Pr, A.K.; Tv, K.; Ross, J.A. Liver tissue engineering and cell sources: Issues and challenges. Liver Int.
2013,33, 666–676. [CrossRef]
152.
Uygun, B.E.; Soto-Gutierrez, A.; Yagi, H.; Izamis, M.-L.; Guzzardi, M.A.; Shulman, C.; Milwid, J.; Kobayashi, N.; Tilles, A.;
Berthiaume, F.; et al. Organ reengineering through development of a transplantable recellularized liver graft using decellularized
liver matrix. Nat. Med. 2010,16, 814–820. [CrossRef] [PubMed]
153.
Lang, R.; Stern, M.M.; Smith, L.; Liu, Y.; Bharadwaj, S.; Liu, G.; Baptista, P.M.; Bergman, C.R.; Soker, S.; Yoo, J.J.; et al.
Three-dimensional culture of hepatocytes on porcine liver tissue-derived extracellular matrix. Biomaterials
2011
,32, 7042–7052.
[CrossRef]
154. Boyer, J.L. Bile formation and secretion. Compr. Physiol. 2013,3, 1035–1078. [PubMed]
155.
Willemse, J.; Roos, F.J.M.; Voogt, I.J.; Schurink, I.J.; Bijvelds, M.; de Jonge, H.R.; van der Laan, L.J.W.; de Jonge, J.; Verstegen,
M.M.A. Scaffolds obtained from decellularized human extrahepatic bile ducts support organoids to establish functional biliary
tissue in a dish. Biotechnol. Bioeng. 2021,118, 836–851. [CrossRef]
156.
Hassanein, W.; Uluer, M.C.; Langford, J.; Woodall, J.D.; Cimeno, A.; Dhru, U.; Werdesheim, A.; Harrison, J.; Rivera-Pratt, C.;
Klepfer, S.; et al. Recellularization via the bile duct supports functional allogenic and xenogenic cell growth on a decellularized
rat liver scaffold. Organogenesis 2017,13, 16–27. [CrossRef] [PubMed]
... A new strategy would involve transplanting cholangiocyte organoids directly into intrahepatic ducts before organ transplantation, during machine perfusion (MP) [60]. Cholangiocytes play an important role in the etiopathogenesis of post-transplant cholangiopathies [42,49,61], and given the high incidence of biliary system disorders following transplantation, the use of cholangiocyte organoids has been proposed. ...
... Cholangiocyte organoids can derive from the epithelial cells of different compartments of the biliary tree (intrahepatic, extrahepatic, and bile); therefore, a nomenclature has been proposed that allows us to identify the organoids based on their origin: intrahepatic cholangiocyte organoids (ICOs), gallbladder cholangiocyte organoids (GCOs), extrahepatic cholangiocyte organoids (ECOs), and cholangiocyte organoids derived from bile (BCOs) [60]. All of these structures share similar phenotypic characteristics when grown, but they have different identities based on the location and composition of the bile [87]. ...
... For organoid generation from tissue-derived primary cells or bile (a schematic representation is shown in Figure 4A), the Scaffold technique that exploits the extracellular matrix (ECM)-based hydrogel or Matrigel is used [90,91]. It is commercially available, and, through different growth factors in the culture medium and signaling pathways, recreates a bioactive micro-environment in which the cells differentiate and generate spherical structures and finally mature into cholangiocytes or hepatocyte organoids [60,90]. ...
Article
Full-text available
Cholangiopathies include a group of chronic progressive disorders, affecting the cholangiocytes, the epithelial cells that line the biliary tree, leading to liver parenchymal fibrosis and eventually end-stage liver disease necessitating transplantation. Experimental modeling of these multifactorial cholestatic diseases faces challenges due to the lack of adequate experimental in vitro and in vivo models. A novel approach employs three-dimensional organoid systems that offer several advantages for modeling disease and testing drug response in vitro. Organoids mimic intercellular communication, replicate the architecture of organs, and maintain the cell’s original phenotype. Cholangiocyte organoids provide an in vitro model to study the pathogenesis and pharmacotherapeutic treatment of cholangiopathies and show great promise for regenerative therapies. In particular, patient-derived organoids allow personalized medicine approaches and the study of individual disease characteristics. This review highlights the significance of cholangiocyte organoid models in advancing our understanding of cholangiopathies and driving advancements in regenerative medicine strategies.
... Considering the high ability to adhere the cells, liver scaffolds are applied for the treatment of hepatobiliary diseases and liver cancer [16] . In recent years, improvement of two-and three-dimensional culture systems in laboratory conditions, as well as the use of organ-on-a-chip technology has led to advancements in the treatment of liver-related diseases [10,17,18] . ...
... A variety of somatic and stem cell sources have utilized for re-cellularization of liver scaffolds [10,[13][14][15][16][17][18][30][31][32] . For the first time in the present study, we seeded the MEF cells on the decellularized liver tissue, and our observations indicated that these cells were adhered on the surface and penetrated into the scaffold. ...
... These changes in ECM microstructure can increase resistance to enzymatic digestion of the ECM and lead to significant discrepancies between human livers [30,31,[35][36][37][38]. ...
Article
Full-text available
Liver transplantation represents the only definitive treatment for patients with end-stage liver disease. However, the shortage of liver donors provokes a dramatic gap between available grafts and patients on the waiting list. Whole liver bioengineering, an emerging field of tissue engineering, holds great potential to overcome this gap. This approach involves two main steps; the first is liver decellularization and the second is recellularization. Liver decellularization aims to remove cellular and nuclear materials from the organ, leaving behind extracellular matrices containing different structural proteins and growth factors while retaining both the vascular and biliary networks. Recellularization involves repopulating the decellularized liver with appropriate cells, theoretically from the recipient patient, to reconstruct the parenchyma, vascular tree, and biliary network. The aim of this review is to identify the major advances in decellularization and recellularization strategies and investigate obstacles for the clinical application of bioengineered liver, including immunogenicity of the designed liver extracellular matrices, the need for standardization of scaffold fabrication techniques, selection of suitable cell sources for parenchymal repopulation, vascular, and biliary tree reconstruction. In vivo transplantation models are also summarized for evaluating the functionality of bioengineered livers. Finally, the regulatory measures and future directions for confirming the safety and efficacy of bioengineered liver are also discussed. Addressing these challenges in whole liver bioengineering may offer new solutions to meet the demand for liver transplantation and improve patient outcomes.
... 20 Organoids for TE TE aims to create functional tissues from cells combined with a scaffold consisting of either biological or synthetic biomaterials, mostly hydrogels or decellularized organs. A wide variety of biomaterials has been used, and for a concise overview of suitable hydrogels for LRT, we refer to a review of Ye et al 54 and a review of Willemse et al. 55 Organoid-based TE would be the preferred option to treat diseases affecting the entire liver, like steatohepatitis and cirrhosis. Either a complete liver can be bioengineered, or a construct large enough to restore the failing liver function. ...
... Modern 3D cell culture scaffold techniques often rely on the use of various biomaterials, including hydrogels. Hydrogels are hydrophilic, cross-linked polymer chains [63,64] that can be pre-prepared to facilitate 3D cell culture due to their similarity to the extracellular matrix [65]. Although hydrogels can vary greatly in water content, chain composition, and ionic charge [66], their use in 3D cell culture is well-established. ...
Article
Full-text available
Biomimetic scaffolds imitate native tissue and can take a multidimensional form. They are biocompatible and can influence cellular metabolism, making them attractive bioengineering platforms. The use of biomimetic scaffolds adds complexity to traditional cell cultivation methods. The most commonly used technique involves cultivating cells on a flat surface in a two-dimensional format due to its simplicity. A three-dimensional (3D) format can provide a microenvironment for surrounding cells. There are two main techniques for obtaining 3D structures based on the presence of scaffolding. Scaffold-free techniques consist of spheroid technologies. Meanwhile, scaffold techniques contain organoids and all constructs that use various types of scaffolds, ranging from decellularized extracellular matrix (dECM) through hydrogels that are one of the most extensively studied forms of potential scaffolds for 3D culture up to 4D bioprinted biomaterials. 3D bioprinting is one of the most important techniques used to create biomimetic scaffolds. The versatility of this technique allows the use of many different types of inks, mainly hydrogels, as well as cells and inorganic substances. Increasing amounts of data provide evidence of vast potential of biomimetic scaffolds usage in tissue engineering and personalized medicine, with the main area of potential application being the regeneration of skin and musculoskeletal systems. Recent papers also indicate increasing amounts of in vivo tests of products based on biomimetic scaffolds, which further strengthen the importance of this branch of tissue engineering and emphasize the need for extensive research to provide safe for humansbiomimetic tissues and organs. In this review article, we provide a review of the recent advancements in the field of biomimetic scaffolds preceded by an overview of cell culture technologies that led to the development of biomimetic scaffold techniques as the most complex type of cell culture.
... A previous study demonstrated the utility of decellularized liver scaffolds as transplantable regenerative organs in long-term experiments using large animal models of chronic liver dysfunction [10]. In addition, the liver-derived extracellular matrix (ECM) is preserved in the scaffold, contributes to adhesion, morphology, and differentiation of perfused cells, and regulates the activation of progenitor or mature liver cells as a hepatocyte progenitor niche [11][12][13][14]. ...
Article
Full-text available
Reconstruction of the biliary system is indispensable for the regeneration of transplantable liver grafts. Here, we report the establishment of the first continuous three-dimensional biliary system scaffold for bile acid excretion using a novel method. We confirmed the preservation of the liver-derived extracellular matrix distribution in the scaffold. In addition, hepatocyte progenitors decellularized via the bile duct by slow-speed perfusion differentiated into hepatocyte- and cholangiocyte-like cells, mimicking hepatic cords and bile ducts, respectively. Furthermore, qRT-PCR demonstrated increased ALB , BSEP , and AQP8 expression, revealing bile canaliculi- and bile duct-specific genetic patterns. Therefore, we concluded that locally preserved extracellular matrices in the scaffold stimulated hepatic progenitors and provided efficient differentiation, as well as regeneration of a three-dimensional continuous biliary system from hepatic cords through bile ducts. These findings suggest that organ-derived scaffolds can be utilized for the efficient reconstruction of functional biliary systems.
Article
Purpose of review With changing donor characteristics (advanced age, obesity), an increase in the use of extended criteria donor (ECD) livers in liver transplantation is seen. Machine perfusion allows graft viability assessment, but still many donor livers are considered nontransplantable. Besides being used as graft viability assessment tool, ex situ machine perfusion offers a platform for therapeutic strategies to ameliorate grafts prior to transplantation. This review describes the current landscape of graft repair during machine perfusion. Recent findings Explored anti-inflammatory therapies, including inflammasome inhibitors, hemoabsorption, and cellular therapies mitigate the inflammatory response and improve hepatic function. Cholangiocyte organoids show promise in repairing the damaged biliary tree. Defatting during normothermic machine perfusion shows a reduction of steatosis and improved hepatobiliary function compared to nontreated livers. Uptake of RNA interference therapies during machine perfusion paves the way for an additional treatment modality. Summary The possibility to repair injured donor livers during ex situ machine perfusion might increase the utilization of ECD-livers. Application of defatting agents is currently explored in clinical trials, whereas other therapeutics require further research or optimization before entering clinical research.
Article
This review is devoted to the prospect of creating an artificial choledoch duct in case of its intraoperative injuries. Currently, there is no consensus on the optimal tactics of reconstructive surgical interventions, determining indications for the formation of biliodigestive anastomoses and transhepatic drainage of the biliary tract. The quality patients’ life after reconstruction in low mortality rate in this type of surgery decreases due to recurrent cholangitis, which in the future leads to liver abscesses and cirrhosis. The first attempts of prosthetics of the common bile duct were associated with the first use of stents in surgical practice. The second stage in the study of the reconstruction of the common bile duct were attempts to use stents and patches from transplants of subcutaneous veins, urethra, vagina of the rectus abdominis, and small intestine. The third stage in the development of this direction is using stents made with fibers of biodegradable polymer compounds. There several advantages of using biodegradable stents. Firstly, they are completely metabolized in the body and perform a skeleton function. Secondly, it is possible to use multilayer structures by means of cellular technologies that ensure the conformitythis of the model with the structures of the common bile duct. A stent from biodegradable material can be used with radiopaque dye and medications, for example, antibiotics to prevent cholangitis, or drugs supressing enhanced formation of fibrous tissue to prevent strictures.
Article
Full-text available
Hepatotoxicity‐related issues are poorly predicted during preclinical experimentation, as its relevance is limited by the inadequacy to screen all the non‐physiological subclasses of the population. These pitfalls can be solved by implementing complex in vitro models of hepatic physiology and pathologies in the preclinical phase. To produce these platforms, extrusion‐based bioprinting is focused on, since it allows to manufacture tridimensional cell‐laden constructs with controlled geometries, in a high‐throughput manner. Different bioinks, whose formulation is tailored to mimic the chemomechanical environment of hepatic steatosis, the most prevalent hepatic disorder worldwide, are proposed. Internally crosslinked alginate hydrogels are chosen as structural components of the inks. Their viscoelastic properties (G′ = 512–730 Pa and G″ = 94–276 Pa, depending on frequency) are tuned to mimic those of steatotic liver tissue. Porcine hepatic ECM is introduced as a relevant biochemical cue. Sodium oleate is added to recall the accumulation of lipids in the tissue. Downstream analyses on 14‐layered bioprinted structures cultured for 10 days reveal the establishment of steatotic‐like features (intracellular lipid vesicles, viability decrease up to ≈50%) without needing external conditionings. The presented bioinks are thus suitable to fabricate complex models of hepatic steatosis to be implemented in a high‐throughput experimental frame.
Article
Full-text available
The well-established 3D organoid culture method enabled efficient expansion of cholangiocyte-like cells from intrahepatic (IHBD) and extrahepatic bile duct (EHBD) tissue biopsies. The extensive expansion capacity of these organoids enables various applications, from cholangiocyte disease modelling to bile duct tissue engineering. Recent research demonstrated the feasibility of culturing cholangiocyte organoids from bile, which was minimal-invasive collected via endoscopic retrograde pancreaticography (ERCP). However, a detailed analysis of these bile cholangiocyte organoids (BCOs) and the cellular region of origin was not yet demonstrated. In this study, we characterize BCOs and mirror them to the already established organoids initiated from IHBD- and EHBD-tissue. We demonstrate successful organoid-initiation from extrahepatic bile collected from gallbladder after resection and by ERCP or percutaneous transhepatic cholangiopathy from a variety of patients. BCOs initiated from these three sources of bile all show features similar to in vivo cholangiocytes. The regional-specific characteristics of the BCOs are reflected by the exclusive expression of regional common bile duct genes (HOXB2 and HOXB3) by ERCP-derived BCOs and gallbladder-derived BCOs expressing gallbladder-specific genes. Moreover, BCOs have limited hepatocyte-fate differentiation potential compared to intrahepatic cholangiocyte organoids. These results indicate that organoid-initiating cells in bile are likely of local (extrahepatic) origin and are not of intrahepatic origin. Regarding the functionality of organoid initiating cells in bile, we demonstrate that BCOs efficiently repopulate decellularized EHBD scaffolds and restore the monolayer of cholangiocyte-like cells in vitro. Bile samples obtained through minimally invasive procedures provide a safe and effective alternative source of cholangiocyte organoids. The shedding of (organoid-initiating) cholangiocytes in bile provides a convenient source of organoids for regenerative medicine.
Article
Full-text available
Organoids regenerate human bile ducts Bile ducts carry bile from the liver and gall bladder to the small intestine, where it aids digestion. Cholangiocytes are epithelial cells that line bile ducts and modify bile as its transported through the biliary tree. Chronic liver diseases involving cholangiocytes account for a large fraction of liver failure and the need for liver transplantation. Because liver donors are in short supply, Sampaziotis et al. used organoid technology to develop a cell-based therapy using human tissue (see the Perspective by Kurial and Willenbring). Cholangiocyte organoids were transplanted into the intrahepatic ducts of deceased human donor livers undergoing ex vivo normothermic perfusion. The livers could be maintained for up to 100 hours, and the transplanted organoids engrafted, exhibited function, and could repair bile ducts. Science , this issue p. 839 ; see also p. 786
Article
Full-text available
The development, homeostasis, and repair of intrahepatic and extrahepatic bile ducts are thought to involve distinct mechanisms including proliferation and maturation of cholangiocyte and progenitor cells. This study aimed to characterize human extrahepatic cholangiocyte organoids (ECO) using canonical Wnt-stimulated culture medium previously developed for intrahepatic cholangiocyte organoids (ICO). Paired ECO and ICO were derived from common bile duct and liver tissue, respectively. Characterization showed both organoid types were highly similar, though some differences in size and gene expression were observed. Both ECO and ICO have cholangiocyte fate differentiation capacity. However, unlike ICO, ECO lack the potential for differentiation towards a hepatocyte-like fate. Importantly, ECO derived from a cystic fibrosis patient showed no CFTR channel activity but normal chloride channel and MDR1 transporter activity. In conclusion, this study shows that ECO and ICO have distinct lineage fate and that ECO provide a competent model to study extrahepatic bile duct diseases like cystic fibrosis.
Article
Full-text available
Biliary disorders can lead to life‐threatening disease and are also a challenging complication of liver transplantation. As there are limited treatment options, tissue engineered bile ducts could be employed to replace or repair damaged bile ducts. We explored how these constructs can be created by seeding hepatobiliary LGR5⁺ organoids onto tissue‐specific scaffold. For this, we decellularized discarded human extrahepatic bile ducts (EBD) that we recellularized with organoids of different origin, that is, liver biopsies, extrahepatic bile duct biopsies, and bile samples. Here, we demonstrate efficient decellularization of EBD tissue. Recellularization of the EBD extracellular matrix (ECM) with the organoids of extrahepatic origin (EBD tissue and bile derived organoids) showed more profound repopulation of the ductal ECM when compared with liver tissue (intrahepatic bile duct) derived organoids. The bile duct constructs that were repopulated with extrahepatic organoids expressed mature cholangiocyte‐markers and had increased electrical resistance, indicating restoration of the barrier function. Therefore, the organoids of extrahepatic sources are identified to be the optimal candidate for the development of personalized tissue engineered EBD constructs.
Article
Full-text available
The organoid model represents a major breakthrough in cell biology that has revolutionised biomedical research. Organoids are 3D physiological in vitro structures that recapitulate morphological and functional features of in vivo tissues and offer significant advantages over traditional cell culture methods. Liver organoids are of particular interest because of the pleiotropy of functions exerted by the human liver, their utility to model different liver diseases, and their potential application as cell-based therapies in regenerative medicine. Moreover, because they can be derived from patient tissues, organoid models offer new perspectives in personalised medicine and drug discovery. In this review, we discuss the current liver organoid models for the study of liver disease.
Article
Full-text available
The utilization of marine-based collagen is growing fast due to its unique properties in comparison with mammalian-based collagen such as no risk of transmitting diseases, a lack of religious constraints, a cost-effective process, low molecular weight, biocompatibility, and its easy absorption by the human body. This article presents an overview of the recent studies from 2014 to 2020 conducted on collagen extraction from marine-based materials, in particular fish by-products. The fish collagen structure, extraction methods, characterization, and biomedical applications are presented. More specifically, acetic acid and deep eutectic solvent (DES) extraction methods for marine collagen isolation are described and compared. In addition, the effect of the extraction parameters (temperature, acid concentration, extraction time, solid-to-liquid ratio) on the yield of collagen is investigated. Moreover, biomaterials engineering and therapeutic applications of marine collagen have been summarized.
Article
Full-text available
The terminology “game changing technology” is every so often used as a hyperbole to underline the importance of research to the outside world. The experimental work of the ETH Zurich and University Hospital Zurich, recently published in Nature Biotechnology by Eshmunov et al. “An integrated perfusion machine preserves injured human livers for 1 week” (1), is however no less than a real game changer in the field of liver transplantation. The authors achieved to increase ‐for the first time‐ stable liver function ex‐vivo from a few hours to 7 days using a normothermic machine perfusion (NMP) device. This leap forward in perfusion time, while maintaining physiological balances, is incredibly important, as this opens a window of opportunity to explore ex‐vivo organ repair therapies.
Article
Full-text available
The recent demonstration that primary cells from the liver can be expanded in vitro as organoids holds enormous promise for regenerative medicine and disease modelling. The use of three-dimensional (3D) cultures based on ill-defined and potentially immunogenic matrices, however, hampers the translation of liver organoid technology into real-life applications. We here use chemically defined hydrogels for the efficient derivation of both mouse and human hepatic organoids. Organoid growth is found to be highly stiffness-sensitive, a mechanism independent of acto-myosin contractility and requiring instead activation of the Src family of kinases (SFKs) and yes-associated protein 1 (YAP). Aberrant matrix stiffness, on the other hand, results in compromised proliferative capacity. Finally, we demonstrate the establishment of biopsy-derived human liver organoids without the use of animal components at any step of the process. Our approach thus opens up exciting perspectives for the establishment of protocols for liver organoid-based regenerative medicine.
Article
Hepatic, pancreatic, and biliary (HPB) organoids are powerful tools for studying development, disease, and regeneration. As organoid research expands, the need for clear definitions and nomenclature describing these systems also grows. To facilitate scientific communication and consistent interpretation, we revisit the concept of an organoid and introduce an intuitive classification system and nomenclature for describing these 3D structures through the consensus of experts in the field. To promote the standardization and validation of HPB organoids, we propose guidelines for establishing, characterizing, and benchmarking future systems. Finally, we address some of the major challenges to the clinical application of organoids.
Article
The only treatment available for end stage liver diseases is orthotopic liver transplantation. Although there is a big donor scarcity, many donor livers are discarded as they do not qualify for transplantation. Alternatively, decellularization of discarded livers can potentially render them transplantable upon recellularization and functional testing. The success of this approach will heavily depend on the quality of decellularized scaffolds which might show variability due to factors including age. Here we assessed the age-dependent differences in liver extracellular matrix (ECM) using rat and human livers. We show that the liver matrix has higher collagen and glycosaminoglycan content and a lower growth factor content with age. Importantly, these changes lead to deterioration in primary hepatocyte function potentially due to ECM stiffening and integrin-dependent signal transduction. Overall, we show that ECM changes with age and these changes significantly affect cell function thus donor age should be considered as an important factor for bioengineering liver substitutes.