Liver International - 2022 - Huang - Liver Regeneration Cellular Origin and Molecular Mechanisms

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Received: 2 September 2021 | Revised: 16 December 2021 | Accepted: 12 January 2022

DOI: 10.1111/liv.15174

REVIEW

Liver regeneration: Cellular origin and molecular mechanisms

Ru Huang1 | Xin Zhang2 | Jordi Gracia-­Sancho3 | Wei-­Fen Xie2

1
Department of Gastroenterology,
Shanghai East Hospital, Tongji University Abstract
School of Medicine, Shanghai, China
The liver is known as an organ with high proliferation potential. Clarifying the cel-
2
Department of Gastroenterology,
Changzheng Hospital, Naval Medical
lular origin and deepening the understanding of liver regeneration mechanisms will
University, Shanghai, China help provide new directions for the treatment of liver disease. With the development
3
Liver Vascular Biology Research Group, and application of lineage tracing technology, the specific distribution and dynamic
Barcelona Hepatic Hemodynamic Unit,
IDIBAPS, CIBEREHD, Barcelona, Spain changes of hepatocyte subpopulations in homeostasis and liver injury have been il-
lustrated. Self-­replication of hepatocytes is responsible for the maintenance of liver
Correspondence
Wei-­Fen Xie, Department of function and mass under homeostasis. The compensatory proliferation of remaining
Gastroenterology, Changzheng Hospital, hepatocytes is the main mechanism of liver regeneration following acute and chronic
415 Fengyang Road, Shanghai, 200003,
China. liver injury. Transdifferentiation between hepatocytes and cholangiocytes has been
Email: weifenxie@medmail.com.cn recognized upon severe chronic liver injury. Wnt/β-­catenin, Hippo/YAP and Notch sig-
Jordi Gracia-­Sancho, IDIBAPS Biomedical nalling play essential roles in the maintenance of homeostatic liver and hepatocyte-­to-­
Research Institute. Rosselló 149, 08036, cholangiocyte conversion under liver injury. In this review, we summarized the recent
Barcelona, Spain.
Email: jordi.gracia@idibaps.org studies on cell origin of newly generated hepatocytes and the underlying mechanisms
of liver regeneration in homeostasis and liver injury.
Funding information
This study is supported by National
Natural Science Foundation of China KEYWORDS
(82030021, 82072641). cholangiocytes, compensatory proliferation, hepatocyte regeneration, hepatocyte
subpopulation, lineage tracing, liver progenitor cells, metabolic zonation, ScRNA-­seq,
Handling Editor: Luca Valenti. transdifferentiation

The liver is the largest organ in the body and plays critical roles in 1 | G E N E TI C LI N E AG E TR AC I N G : S TATE-­
nutrient metabolism, detoxification, regulation of blood clotting fac- O F-­T H E-­A RT TEC H N I Q U E FO R TH E S T U DY
tors and bile synthesis. The liver lobule, as the basic functional and O F LI V E R R EG E N E R ATI O N
anatomical unit of the liver, is composed of hepatic cords or hepatic
plates, with central veins and portal veins at each end. Hepatocytes Most studies in the field of liver regeneration have been performed
and cholangiocytes are the two epithelial cell types constituting the in mice during the last decade. Lineage tracing technology has been
hepatic parenchyma. For centuries, the liver has been known as an widely employed to closely monitor the migration, expansion and
organ with high regenerative potential. Hepatocyte regeneration transdifferentiation of specific cell populations and their progeny
is pivotal to the maintenance of liver function and size during ho- during homeostasis and liver injury.1 The application of genetic la-
meostasis and after liver injury. The cell origin of newly generated belling technology provides a solid basis for the cellular source of
hepatocytes and the underlying mechanisms have been extensively newly-­produced hepatocytes in liver regeneration.
explored in recent decades. This review focuses on the cellular ori- Currently, the Cre-­loxP system is the standard lineage tracing
gin and the underlying molecular mechanisms in liver regeneration strategy, which works by placing Cre recombinase under the control
under distinct circumstances. of the transcriptional regulatory sequence of certain marker genes

Ru Huang and Xin Zhang contributed equally.

© 2022 John Wiley & Sons A/S . Published by John Wiley & Sons Ltd.

1486 | 
wileyonlinelibrary.com/journal/liv Liver International. 2022;42:1486–1495.
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
HUANG et al. 1487

and inserting a two-­loxP-­flanked stop cassette in an open gene locus


with a downstream reporter gene in mice. 2,3 Once the expression
Key points
of the Cre recombinase is activated, the stop cassette between the
• Lineage tracing technology provides a solid basis for
loxP sites is excised, and the reporter genes (GFP, tdTomato, etc.)
identifying the role of hepatocyte subpopulations and
start expressing to label certain cell populations. Since Cre-­mediated
cholangiocytes during liver regeneration.
site-­specific recombination is passed on to their progeny, the cell
• Hepatocytes self-­replication is the major contributor to
lineage could be labelled permanently from the timepoint of Cre ex-
liver regeneration.
pression. While constitutive Cre restricts reporter gene expression
• The conversion between hepatocytes and cholangio-
spatially, an inducible Cre strategy further controls gene expression
cytes during liver injury demonstrates the high plasticity
temporally by fusing Cre with a T2 mutant of the oestrogen recep-
of liver cells.
tor, which is activated through tamoxifen induction.4 However, com-
• Further characterization of cellular and molecular mech-
pared to constitutive Cre, inducible Cre activity is difficult to fully
anisms underlying liver regeneration is in great demand
activate. In addition, high dose of tamoxifen administration has been
to help provide reliable approaches towards the man-
reported to lead to liver injury with ectopic stem cell marker ex-
agement of liver diseases.
pression.5,6 Thus, it is important to determine tamoxifen quantities
and efficacy before induction of liver injury. Due to the difference
in Cre reporter gene recombination sensitivities, the efficacy of cell
labelling differs, with divergent results obtained from different lab-
oratories. Therefore, interpretation of lineage tracing results should subpopulations have been defined functionally through in vitro cul-
be conducted carefully, with immunostaining of liver cell markers tivation and cell transplantation, it remains to be determined how
incorporated. they function in vivo under homeostasis and liver injury.7 Further
characterization of those cell subpopulations through combination
of spatial transcriptomes and scRNA-­seq or by lineage-­tracing stud-
2 | CO NTROV E R S Y OV E R TH E E X I S TE N C E ies are in demand.
O F LP C s I N LI V E R R EG E N E R ATI O N Multiple groups have performed fate tracing using cholangio-
cytes markers (including SOX9, HNF1β, CK19 or OPN) to label LPCs
Unlike skin tissue and intestinal epithelial tissue where stem cells (Table 1). Intriguingly, Furuyama et al. found Sox9-­expressing cells
or progenitor cells exist, there is no consensus about the exist- contributed significantly to the hepatocytes pool under homeostasis
ence of liver progenitor cells (LPCs) during liver regeneration.7 Oval and liver injury induced by CCl4, BDL or MCDE in SOX9IRES-­CreERT
cells emerging from canals of Hering, which located at the termi- knockin mouse strain, 22 whereas Tarlow et al showed hepatocytes
8
nal branches of bile ducts, were identified by Farber and Popper rarely came from Sox9+ cells in various liver injury using BAC trans-
9
et al. in 2-­acetylaminofluorene (2-­A AF, a hepatotoxic carcinogen) genic SOX9CreERT2 mice strain. 23 The discrepancy between these
treated rats in mid-­1950s. Following studies characterized oval cells studies could be due to the different promotor specificity and sen-
as hyperplastic epithelial cells with small cellular size, a high nu- sitivity in the SOX9 genetic labelling mouse tools. HNF1β, CK19 or
clear/plasma ratio and expression of the hepatoblast marker alpha-­ OPN labelled cholangiocytes also show limited contributions to liver
fetoprotein (AFP).8-­11 Oval cells were considered to be bipotential regeneration under physiological conditions and during liver inju-
progenitor cells as they showed the capacity to differentiate into ry. 24-­27 One explanation underlying the limited role of those cells is
hepatocytes and bile duct cells in vitro.12 Oval cells were also re- the incomplete blockade of hepatocyte proliferation in mouse liver
ported as LPCs in human chronic liver disease, and their numbers injury model, which could not faithfully reflect the impaired prolifer-
are associated with disease severity.13 However, due to the lack of ation of hepatocytes in human chronic liver disease.16,28 This notion
specific markers for oval cells and the inapplicability of 2-­A AF/PHx is supported by the study that inhibition of hepatocyte proliferation
model in mice, there is still lack of direct in vivo evidence showing through overexpressing p21 leads to significant regenerated hepato-
that oval cells give rise to mature hepatocytes and cholangiocytes in cytes from CK19+ cholangiocytes. 29
any of the classical mouse injury models to date.
Through the years, adult LPCs have been investigated
through in vitro cultivation, liver repopulation assay and lin- 3 | D I S TR I B U TI O N O F H E PATO C Y TE
eage tracing.14-­19 Robust activation of LPCs was detected in S U B P O PU L ATI O N S U N D E R LI V E R
AhCre+Mdm2flox/flox mice model with extensive hepatocytes dam- H O M EOS TA S I S
age and transplantation of EpCAM+CD24 +CD133+ LPCs restored
liver mass in AhCre+Mdm2flox/flox mice. 20 Moreover, through em- Hepatocytes, accounting for approximately 70% of total liver
ploying scRNA-­seq and organoid cultivation, Aizarani et al. identi- cells, renew at a slow rate under physiological conditions. The life
fied EpCAM+TROP2intCK19low progenitor cell in human liver, with cycle of hepatocytes is approximately 200 to 300 days in mice.30
21
strong organoid forming capability. Although such hepatocyte Hepatocytes are highly heterogeneous and have different functions
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1488 HUANG et al.

TA B L E 1 Genetic labelling of
Targeting
cholangiocytes during liver regeneration
Mouse line strategy Circumstances References

Sox9-­CreERT2 Knockin (3’-­UTR) Homeostasis 22


Chronic liver injury
Transgenic Homeostasis 23
Chronic liver injury
Ck19-­CreERT Knockin (ATG) Homeostasis, Acute and Chronic liver 103
injury
Hnf1β-­CreERT2 Transgenic Homeostasis, 26,104
Chronic liver injury
OPN-­CreERT2 Transgenic Homeostasis, Acute and Chronic liver 27
injury

TA B L E 2 Characterization of
Hepatocytes Zone 1 Zone 3
hepatocytes in zone 1 and zone 3
Location Periportal area Pericentral area
Zonation marker Gls2, Pck1, Arg1 GS, Oat, Cyp1a2, Cyp2e1, Glul
Metabolic function Gluconeogenesis Glycolysis
Glycogen and Protein synthesis Lipid Glutamine synthesis
metabolism Xenobiotic metabolism
Ureagenesis

Abbreviations: Arg1, arginase 1; Cyp1a2, cytochrome P450 family 1 subfamily A member 2; Cyp2e1,
cytochrome P450 family 2 subfamily E member 1; Gls2, glutaminase 2; Glul, glutamate-­ammonia
ligase; GS, glutamine synthetase; Oat, ornithine aminotransferase; Pck1, phosphoenolpyruvate.

F I G U R E 1 Hepatocyte subpopulation
distribution under homeostasis and liver
injury. (A) The liver lobule is divided into
three zones, with periportal area as zone
1, pericentral area as zone 3, and the
region between them as zone 2. Mfsd2a+
hepatocytes and hybrid hepatocytes
(HybHPs) are sited in zone 1, while Axin2+
hepatocytes are mainly located in zone 3.
Terthigh hepatocytes are scattered in the
liver lobule. Hepatocytes self-­replication
contribute to the maintenance of liver
function and mass under homeostasis. (B)
In chronic periportal liver injury, HybHPs
and Mfsd2a+ hepatocytes are mostly
impaired. Midlobular and pericentral
hepatocytes, especially TERThigh
hepatocytes, contribute to hepatocytes
regeneration. (C) During chronic
pericentral injury, TERThigh, HybHPs and
Mfsd2a+ hepatocytes proliferate and
expand significantly to repopulate the
liver. Abbreviations: PV, portal vein; BD,
bile duct; CV, central vein; HA, hepatic
artery

and metabolic expression profiles along the portal-­


central axis (Figure 1A).31 Hepatocytes adjacent to the portal area belong to
31-­33
(Table 2). Based on their metabolic features, hepatocytes are zone 1, with access to blood containing high concentrations of oxy-
roughly divided into three regions, known as metabolic zonation gen and nutrients delivered by the hepatic artery and portal vein,
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
HUANG et al. 1489

respectively. Periportal hepatocytes are mainly involved in liver met- The contribution of periportal hepatocytes has also been investi-
abolic functions, including gluconeogenesis, glycogen and protein gated. Font-­Burgada et al. identified a subpopulation of hybrid peri-
synthesis, lipid metabolism and ureagenesis. Hepatocytes near the portal hepatocytes (HybHPs) coexpressing the mature hepatocyte
central vein belong to zone 3, where glycolysis, glutamine synthesis marker HNF4α and low levels of the bile duct cell-­specific gene SOX9.
and xenobiotic metabolism take place. Zone 2 is located between Although HybHPs contribute to hepatocyte regeneration upon in-
34
the above two regions. Single-­cell RNA sequencing (scRNA-­seq) jury, the population of HybHPs remains at approximately 4.5% under
combined with spatial reconstruction provides in-­depth characteri- physiological circumstances.43 In addition, the proportion of another
zation of liver gene expression patterns and cell population zonation periportal subpopulation—­Mfsd2a+ hepatocytes—­
was decreased
21,35,36
along the portal-­
central axis. Lineage tracing studies per- from 40% at 6 weeks to 20% at 20 weeks and then remained stable
formed by labelling hepatocyte subpopulations have revealed their under homeostasis.44 Furthermore, a type of hepatocyte subpopu-
specific distribution in the liver lobule during homeostasis and liver lation with high expression of telomerase (TERTHigh), which is distrib-
regeneration (Table 3). uted randomly in the liver lobules, expanded 10-­fold (from 2.8% to
Lgr5+/Axin2+ cells, which utilize WNT/β-­c atenin signalling nearly 30%) in 1 year during homeostasis.45 These findings suggest
to sustain their high proliferative capacity, are considered tissue that periportal hepatocytes play a limited role during homeostasis.
stem cells in many tissues. 37 Hepatocytes present a centro-­portal More recently, the proliferative capacity of hepatocytes in three
WNT/β-­c atenin signalling gradient, in which Lgr5 and Axin2 are co-­ zones has been thoroughly explored by two independent groups
expressed in pericentral hepatocytes. 38,39 The controversial roles employing multiple genetic lineage tracing approaches. He et al.
+ +
of Lgr5 /Axin2 cells in homeostasis have been reported. Using developed a dual recombinase-­mediated genetic system involving
Axin2-­CreERT2 mice expressing CreERT2 under control of the en- DreER, Ki67-­CrexER and R26-­GFP reporter (named ‘ProTracer’) to
dogenous Axin2 promoter, Wang et al. demonstrated that Axin2+ record cell proliferation continuously during a given time window,
hepatocytes are a self-­renewing cell population and expand to 30% and found that hepatocytes in zone 2 showed a higher proliferation
of the area of the entire liver for replacing other hepatocyte sub- rate than hepatocytes in zones 3 and 1.46 Wei et al. performed lin-
40
populations over 1 year during homeostasis. However, another eage tracing in 14 CreER knock-­in mouse models, labelling distinct
study using bacterial artificial chromosome (BAC)-­transgenic mice zonal hepatocyte subpopulations, and found that hepatocytes in
to trace Axin2+ cells showed that pericentral Axin2+ hepatocytes zone 2 were major contributors to homeostasis repopulation and
do not have superior proliferative capacity compared with other regeneration.47 These studies further demonstrated that the self-­
41
hepatocyte subpopulations. Consistently, Ang et al. showed replication of hepatocytes is responsible for the maintenance of
that the number of pericentral Lgr5+ hepatocytes remained stable liver function and mass under homeostasis. In particular, midlobular
under homeostasis.42 hepatocytes contribute the most to the homeostasis repopulation.

TA B L E 3 Lineage tracing of hepatocyte subpopulation during homeostasis and liver injury

Hepatocyte Percentage of
Zonation subpopulation Mouse line Targeting strategy Total hepatocytes Circumstances Refer-­ences

Zone 1 HybHP Sox9-­CreERT2 Transgenic 4.53 ± 0.24% Homeostasis 43


Acute and chronic liver
injury
SOX9+ Sox9-­CreER; Knockin Around 2.55% Homeostasis 74
Hnf4a-­DreER Acute and chronic liver
injury
Mfsd2a+ Mfsd2a-­CreER, Knockin (ATG) Around 40% Homeostasis 44
Acute and chronic liver
injury
Zone 3 Axin2+ Axin2-­CreERT2 Knockin (ATG) Around 5% Homeostasis 40,105
Axin2-­CreERT2 Transgenic ND Homeostasis 41
Acute liver injury
GS+ GS-­CreER Knockin (3’-­UTR) 9.4 ± 0.4% Homeostasis 47
Chronic liver injury
Lgr5+ Lgr5-­r tTA-­IRES Transgenic 2 ~ 5% Homeostasis 42
Acute liver injury
Distributed Terthigh Tert-­CreERT2 Knockin (ATG) 2.8 ± 0.4% Homeostasis 45
throughout Chronic liver injury
the liver Hamp2+ Hamp2-­CreER Knockin (3’-­UTR) 7.4 ± 0.7% Homeostasis 47
lobules Chronic liver injury
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1490 HUANG et al.

4 | CO M PE N S ATO RY PRO LI FE R ATI O N related genes. 20,29,58,59 CCl4 and thio-


interfering with cell cycle-­
O F R E M A I N I N G H E PATO C Y TE S I N ACU TE acetamide (TAA) are employed to induce pericentral liver injury with
LI V E R I N J U RY fibrosis.60,61 A 3,5-­diethoxycarbonyl-­1,4-­dihydrocollidine (DDC)-­
enriched diet is usually used to mimic chronic cholestatic liver injury
In contrast to the slow turnover of hepatocytes during homeostasis, by damaging the biliary compartment.62,63
hepatocytes show remarkable regeneration capability upon injury. Hepatocyte self-­replication remains to be the main contributor
Partial hepatectomy (PHx) is the classic acute liver injury model that to liver mass restoration during chronic liver injury, while certain
removes certain lobes of the liver.48 The most common PHx model, in hepatocyte subpopulations exhibit superior proliferative capabil-
which 70% of the liver is removed, induces hypertrophy and compen- ity. 24,64,65 Consistent with the finding of homeostasis, TERTHigh he-
satory proliferation of the remaining hepatocytes to restore liver mass patocytes have higher proliferative ability than TERTLow hepatocytes
49
within 3 weeks. Lineage tracing studies showed that hepatocyte in CCl4-­and DDC-­induced liver injury models.45 On the other hand,
subpopulation gives rise to its own lineage during the recovery from HybHP expands significantly in chronic liver injury models without
PHx.41,42 Shortly after PHx, the activity of urokinase-­type plasminogen causing hepatocellular carcinoma.43 During the recovery, the newly
activator (uPA) and metalloproteinases increases, leading to remodel- generated hepatocytes reprogram to adapt their metabolic function
ling of the extracellular matrix and formation of active heterodimeric with their zonation in the liver lobule.43,44 Meantime, the biliary tract
50,51
hepatocyte growth factor (HGF). The peak of liver regeneration is also remodelled with structural transformation in response to liver
following 70% PHx occurs within 48 hours, measured by the number injuries.66
of hepatocytes incorporating 3H-­
thymidine in S phase.52 A recent The cell origin of newly regenerated hepatocytes differs slightly
study showed that periportal hepatocytes are the first to proliferate in various liver injury models. Due to the differences in areas of
after PHx, followed by midlobular and pericentral hepatocytes, as injury inflicted through the liver injury model, hepatocyte subpop-
demonstrated by the hepatocyte-­specific ProTracer system.46 ulations located outside targeted areas are more capable of prolif-
Hepatocyte nuclear factor 4α (HNF4α) plays an indispensable role erating to restore liver mass and maintain physiological function
in hepatocyte differentiation and maintenance of hepatocyte function (Figure 1B-­C).46,57,67 The periportal subpopulation HybHP and
53,54 +
during embryonic development and homeostasis. A recent study Mfsd2a hepatocytes expand significantly to replace pericentral
showed that HNF4α is down-­regulated at the beginning of regener- hepatocytes in CCl4-­induced chronic liver injury, but they show
ation following PHx while re-­expression of HNF4α is pivotal for the limited proliferative ability under DDC-­induced periportal liver
termination of regeneration and recovery of hepatocyte function,55 injury.43,44
which indicates that hepatocyte might undergo de-­differentiation with
partial loss of its mature hepatocyte markers following acute liver in-
jury and the restoration of its profiles is essential for its full recovery. 6 | TR A N S D I FFE R E NTI ATI O N B E T W E E N
Both carbon tetrachloride (CCl4) and acetaminophen (APAP) H E PATO C Y TE S A N D C H O L A N G I O C Y TE S
have been employed to induce acute liver injury with pericentral
hepatocyte necrosis. In CCl4 model, proliferation of remaining he- Both hepatocytes and cholangiocytes originate from hepatoblasts
patocytes peaked at 2–­3 days after CCl4 injection and liver archi- during embryonic development.5,68 The conversion between hepat-
25,56
tecture was completely restored at 10 days after CCl4 injection. ocytes and cholangiocytes during adulthood has also been observed
Pericentral Lgr5+ hepatocytes depleted in acute CCl4 injury were (Figure 2). Loss of hepatocyte cellular identity was found in non-­
gradually replaced by Lgr5− hepatocytes, which regained Lgr5 ex- alcoholic steatohepatitis (NASH), DDC mouse model and alcoholic
pression to adapt their identity along the portal-­central axis during hepatitis (AH) mouse and human samples.69-­71 In both DDC and AH
42
recovery. Functionally, remaining hepatocytes upregulated the ex- liver, hepatocytes transcriptional reprogramming was also accompa-
pression level of pericentral hepatocytes genes, such as Cyp2e1, to nied by enrichment of cholangiocyte markers. Several studies have
compensate for the function of lost pericentral hepatocytes at 24 h shown that hepatocytes spontaneously reprogram and convert into
57
post-­APAP injury. Taken together, hepatocytes not only proliferate cholangiocytes in vivo during liver regeneration.72-­74 Tarlow et al.
to repopulate the liver mass but also transcriptionally regulate its demonstrated that mature hepatocytes exhibit cholangiocyte phe-
expression to maintain liver function and re-­establish liver metabolic notypes following DDC injury and revert to their original identity
zonation at the same time after acute liver injury. when the injury subsides.75 In vitro studies showed that mature
hepatocytes can be converted into bipotent cells by the addition
of small molecules, and these bipotent cells could repopulate liver
5 | TH E RO LE S O F H E PATO C Y TE S A N D when transplanted in Fah−/− and uPA/SCID liver injury models.76-­78
TH E I R S U B P O PU L ATI O N S I N C H RO N I C Furthermore, Johanna et al. found de novo formation of mature
LI V E R I N J U RY cholangiocytes with bile-­
draining function from hepatocytes in
Alb-­cre+/-­Rbpjf/fHnf6f/f transgenic mice that lack peripheral bile
Chronic liver injury models encompass repetitive administration ducts at birth, confirming the transdifferentiation of hepatocytes to
of chemicals and genetic modulation in transgenic mice through cholangiocytes.79
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
HUANG et al. 1491

F I G U R E 2 Conversion between
cholangiocytes and hepatocytes. In
chronic injury, hepatocytes spontaneously
transdifferentiate to cholangiocytes.
In liver injury with bile duct impaired,
the biliary system is formed through
hepatocytes transdifferentiation.
As the liver injury becomes severe
and prolonged or with hepatocytes
proliferation inhibited, cholangiocytes
transdifferentiate into hepatocytes to
replenish the liver parenchyma

The role of cholangiocytes in liver regeneration has been elu- cholangiocytes is required for the maintenance of liver metabolic
cidated through multiple genetic labelling mice (Table 1). Lineage function, proliferation and transdifferentiation ability.
tracing through labelling cholangiocytes with HNF1β, CK19, OPN The Wnt/β-­
catenin signalling gradient along the centropor-
or SOX9 promoters showed their limited contributions to liver tal axis controls homeostatic liver metabolic zonation.38,84,85
regeneration in classic liver injury model. 24-­27 However, when Overexpression of active β-­catenin promoted hepatocyte-­to-­
the extent of liver injury becomes severe, the outcome differs. cholangiocyte transdifferentiation in DDC liver injury model.86 A
In a metronidazole-­induced zebrafish liver injury model in which recent study showed that upregulation of Wnt/β-­catenin activity in
nearly all hepatocytes were lost, the newly regenerated hepato- hepatocytes by deletion of ZNRF3 and RNF43 increased hepatocyte
cytes mainly arose from transdifferentiation of biliary cells. 80 proliferation, impaired hepatocyte metabolic zonation and promoted
During this process, cholangiocytes first dedifferentiate into liver tumours.87 In addition, the Wnt-­producing niche provided by
hepatoblast-­like cells as they proliferate, and then differentiate the endothelium and macrophages plays an indispensable role in
into proliferative hepatocytes to promote restoration of the liver the maintenance of functional compensation and the proliferative
parenchyma. 81 Raven et al. found significant regenerated hepato- response of hepatocytes.40,57
cytes from cholangiocytes in CK19CreERTtdTomato LSL mice with Highly conserved Hippo/YAP signalling plays a prominent role
impaired hepatocyte proliferation by overexpressing p21 or ab- in regulating liver size and regeneration. 88 The activation of YAP
lating Itgb1. 29 We also demonstrated that nearly 10% of newly in hepatocytes led to loss of hepatocyte identity, conversion of
regenerated hepatocytes were derived from biliary epithelial cells hepatocytes into cholangiocytes and weakened liver regenera-
82
(BECs) in mice with prolonged chronic liver injury. Moreover, tion.70,89 Furthermore, clinical samples from alcoholic hepatitis
biphenotypic cells co-­e xpressing hepatocyte marker HNF4α and patients showed profound YAP activation in hepatocytes ac-
cholangiocyte marker CK19 or SOX9 were detected in human companied with increased expression of biliary marker (SOX9,
samples with several types of liver diseases as well as in injured HNF1β).70 Studies have shown transcriptional heterogeneity of
mouse liver, suggesting high cellular plasticity in both human and YAP signalling in homeostatic cholangiocytes and upregulation
rodents liver.72,82 Another study showed that CDE diet-­induced of YAP signalling in cholangiocytes and hepatocytes upon CCl 4
severe liver injury stimulates the formation of cholangiocytes-­ and DDC injury.90,91 Verboven et al. showed that the conditional
derived hepatocytes in hepatocyte-­s pecific β-­c atenin knockout knockout of the Hippo pathway downstream effectors YAP/TAZ
83
mice. These studies support the notion of cholangiocytes-­to-­ in mature hepatocytes did not affect hepatocyte proliferation
hepatocytes conversion in severe liver injury. Thus, both hepato- after acute liver injury, while the deletion of YAP/TAZ in chol-
cytes and cholangiocytes have the capability to convert into each angiocytes impaired liver regeneration with bile duct deteriora-
other, though the severity of liver injuries for induction of such tion.90,92 These findings highlighted the pivotal role of Hippo/YAP
conversion is quite not the same. signalling in periportal hepatocytes and cholangiocytes during
liver regeneration.
Notch signalling is required for the formation and repair of the
7 | TH E S I G N A LLI N G PATH WAYS bile duct during development and liver injury.93-­96 Activation of
I N VO LV E D I N LI V E R R EG E N E R ATI O N Notch signalling in hepatocytes by enhancing NOTCH1 intracel-
lular fragment expression induced the reprogramming of hepato-
During homeostasis and liver injury, fine-­tuning of Wnt/β-­ cytes into biliary cells during homeostasis and the formation of
catenin, Hippo/YAP and Notch signalling in hepatocytes and/or intrahepatic cholangiocarcinoma.72,97 Inhibition of Notch signalling
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1492 HUANG et al.

in hepatocytes through deletion of the Notch signalling effector the management of liver diseases would be provided through direct
RBP-­Jkappa reduced hepatocyte-­to-­biliary reprogramming in DDC reprogramming of differentiation pathways and indirect modifica-
injury.72 Since Notch signalling acts downstream of Hippo/YAP, the tion of surrounding niche in the liver.
effect of Hippo/YAP signalling in liver injury could be attributed
to the regulation of Notch signalling.89 Furthermore, activation of C O N FL I C T S O F I N T E R E S T
TGF-­β signalling through constitutively active TGFBR1 expression The author(s) declare that they have no conflicts of interest.
in hepatocytes promoted hepatocyte-­to-­cholangicyte transdiffer-
entiation and the formation of bile ducts in an Alagille syndrome E T H I C A L S TA N DA R D S
mouse model (Alb-­cre+/-­Rbpjf/fHnf6f/f ), demonstrating TGF-­β signal- This article does not contain any experiments with human or animal
ling as a key driver during transdifferentiation independent of Notch subjects conducted by any of the authors.
signalling.79
The crosstalk between parenchymal cells and non-­parenchymal DATA AVA I L A B I L I T Y S TAT E M E N T
cells (NPCs), which include liver sinusoidal endothelial cells (LSECs), Data sharing is not applicable to this article as no new data were
hepatic stellate cells (HSCs) and immune cells, play important role generated or analysed in this study.
in liver regeneration (reviewed previously).98-­100 Moreover, a recent
study showed that the dynamic ratio between mesenchymal cells ORCID
and ductal cells regulate the proliferation of epithelial cell through Ru Huang https://orcid.org/0000-0003-0236-3940
101
organoid co-­culture, which shed light on the effect of cell–­cell Wei-­Fen Xie https://orcid.org/0000-0002-7137-112X
contact during liver regeneration.
REFERENCES
1. Sauer B, Henderson N. Site-­specific DNA recombination in mam-
8 | CO N C LU D I N G R E M A R K S A N D FU T U R E malian cells by the Cre recombinase of bacteriophage P1. Proc Natl
Acad Sci USA. 1988;85:5166-­5170.
D I R EC TI O N S
2. Nagy A. Cre recombinase: the universal reagent for genome tailor-
ing. Genesis. 2000;26:99-­109.
Recent work has significantly advanced our understanding of 3. Kretzschmar K, Watt FM. Lineage tracing. Cell. 2012;148:33-­45.
the cellular origins and underlying mechanisms of liver regenera- 4. Hayashi S, McMahon AP. Efficient recombination in diverse tis-
sues by a tamoxifen-­inducible form of Cre: a tool for temporally
tion. Multiple lines of evidences suggest that liver regeneration is
regulated gene activation/inactivation in the mouse. Dev Biol.
a process of hepatocyte self-­duplication in all regions of the liver 2002;244:305-­318.
lobule during homeostasis and following injury, with midlobular 5. Carpentier R, Suñer RE, van Hul N, Kopp JL, Beaudry JB, Cordi
hepatocytes as the main contributor to the homeostatic turnover of S, Antoniou A, Raynaud P, Lepreux S, Jacquemin P, Leclercq IA,
Sander M and Lemaigre FP. Embryonic ductal plate cells give rise
hepatocytes. In the meantime, newly-­generated hepatocytes tran-
to cholangiocytes, periportal hepatocytes, and adult liver progen-
scriptionally regulate their zonally-­expressed genes to adapt their itor cells. Gastroenterology 2011;141: 1432–­8, 1438.e1-­4.
identities along the portal-­central axis. The conversion between 6. Lemaigre FP. Determining the fate of hepatic cells by lineage trac-
hepatocytes and cholangiocytes in several severe injury models ing: facts and pitfalls. Hepatology. 2015;61:2100-­2103.
demonstrated the high plasticity of liver cells. But the precise mech- 7. Miyajima A, Tanaka M, Itoh T. Stem/progenitor cells in liver devel-
opment, homeostasis, regeneration, and reprogramming. Cell Stem
anisms underlying their proliferation and high plasticity remain to be
Cell. 2014;14:561-­574.
further elucidated. Future studies should further characterize the 8. Fausto N, Campbell JS. The role of hepatocytes and oval cells
signalling pathways driving hepatocyte self-­replication and cell fate in liver regeneration and repopulation. Mech Dev. 2003;120:​
plasticity, particularly in situations of ageing. It also remains to be 117-­130.
9. Popper H, Kent G, Stein R. Ductular cell reaction in the liver in
clarified how non-­parenchymal cells contribute to the local niche,
hepatic injury. J mt Sinai Hosp NY. 1957;24:551-­556.
which helps re-­establish the metabolic zonation of newly gener- 10. Factor VM, Radaeva SA, Thorgeirsson SS. Origin and fate of oval
ated hepatocytes. Single-­cell sequencing technology has overcome cells in dipin-­induced hepatocarcinogenesis in the mouse. Am J
the shortcomings of bulk sequencing in the heterogeneous tissues Pathol. 1994;145:409-­422.
11. Evarts RP, Nagy P, Marsden E, Thorgeirsson SS. A precursor-­
with mixed transcriptomic characterization and will help to further
product relationship exists between oval cells and hepatocytes in
identify dynamic signalling pathways in liver cells. 36 With emerging rat liver. Carcinogenesis. 1987;8:1737-­1740.
technology, the field of liver generation is on the verge of making 12. Lazaro CA, Rhim JA, Yamada Y, Fausto N. Generation of he-
great progress in addressing those questions, which will ultimately patocytes from oval cell precursors in culture. Cancer Res.
1998;58:5514-­5522.
deepen our understanding of liver regeneration. Clinically, the gen-
13. Lowes KN, Brennan BA, Yeoh GC, Olynyk JK. Oval cell numbers in
eration of functional and transplantable hepatocytes presents as a human chronic liver diseases are directly related to disease sever-
promising strategy for patients with acute liver failure or metabolic ity. Am J Pathol. 1999;154:537-­541.
liver diseases.102 With further mechanisms underlying liver regener- 14. Schmelzer E, Zhang L, Bruce A, et al. Human hepatic stem cells
from fetal and postnatal donors. J Exp Med. 2007;204:1973-­1987.
ation uncovered, it is conceivable that reliable approaches towards
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
HUANG et al. 1493

15. Huch M, Gehart H, van Boxtel R, et al. Long-­term culture of interactions in liver physiology and disease biology. J Hepatol.
genome-­stable bipotent stem cells from adult human liver. Cell. 2020;73:1219-­1230.
2015;160:299-­312. 37. Barker N, van Es JH, Kuipers J, et al. Identification of stem
16. Lanzoni G, Cardinale V, Carpino G. The hepatic, biliary, and pan- cells in small intestine and colon by marker gene Lgr5. Nature.
creatic network of stem/progenitor cell niches in humans: a 2007;449:1003-­1007.
new reference frame for disease and regeneration. Hepatology. 38. Planas-­Paz L, Orsini V, Boulter L, et al. The RSPO-­LGR4/5-­ZNRF3/
2016;64:277-­286. RNF43 module controls liver zonation and size. Nat Cell Biol.
17. Huch M, Dorrell C, Boj SF, et al. In vitro expansion of single Lgr5+ 2016;18:467-­479.
liver stem cells induced by Wnt-­driven regeneration. Nature. 39. Torre C, Perret C, Colnot S. Transcription dynamics in a physiologi-
2013;494:247-­250. cal process: β-­c atenin signaling directs liver metabolic zonation. Int
18. Tanimizu N, Kobayashi S, Ichinohe N, Mitaka T. Downregulation J Biochem Cell Biol. 2011;43:271-­278.
of miR122 by grainyhead-­like 2 restricts the hepatocytic differ- 40. Wang B, Zhao L, Fish M, Logan CY, Nusse R. Self-­renewing dip-
entiation potential of adult liver progenitor cells. Development. loid Axin2(+) cells fuel homeostatic renewal of the liver. Nature.
2014;141:4448-­4 456. 2015;524:180-­185.
19. Cardinale V, Wang Y, Carpino G, Reid LM, Gaudio E, Alvaro D. 41. Sun T, Pikiolek M, Orsini V, et al. AXIN2(+) pericentral hepatocytes
Mucin-­producing cholangiocarcinoma might derive from biliary have limited contributions to liver homeostasis and regeneration.
tree stem/progenitor cells located in peribiliary glands. Hepatology. Cell Stem Cell. 2020;26:97-­107.e6.
2012;55:2041-­2042. 42. Ang CH, Hsu SH, Guo F, et al. Lgr5(+) pericentral hepatocytes
20. Lu WY, Bird TG, Boulter L, et al. Hepatic progenitor cells of are self-­maintained in normal liver regeneration and suscepti-
biliary origin with liver repopulation capacity. Nat Cell Biol. ble to hepatocarcinogenesis. Proc Natl Acad Sci U S A. 2019;116:​
2015;17:971-­983. 19530-­19540.
21. Aizarani N, Saviano A, Sagar ML, et al. A human liver cell atlas 43. Font-­Burgada J, Shalapour S, Ramaswamy S, et al. Hybrid peripor-
reveals heterogeneity and epithelial progenitors. Nature. tal hepatocytes regenerate the injured liver without giving rise to
2019;572:199-­204. cancer. Cell. 2015;162:766-­779.
22. Furuyama K, Kawaguchi Y, Akiyama H, et al. Continuous cell sup- 44. Pu W, Zhang H, Huang X, et al. Mfsd2a+ hepatocytes repop-
ply from a Sox9-­expressing progenitor zone in adult liver, exocrine ulate the liver during injury and regeneration. Nat Commun.
pancreas and intestine. Nat Genet. 2011;43:34-­41. 2016;7:13369.
23. Tarlow BD, Finegold MJ, Grompe M. Clonal tracing of Sox9+ 45. Lin S, Nascimento EM, Gajera CR, et al. Distributed hepatocytes
liver progenitors in mouse oval cell injury. Hepatology. 2014;60:​ expressing telomerase repopulate the liver in homeostasis and in-
278-­289. jury. Nature. 2018;556:244-­248.
24. Yanger K, Knigin D, Zong Y, et al. Adult hepatocytes are generated 46. He L, Pu W, Liu X, et al. Proliferation tracing reveals regional
by self-­duplication rather than stem cell differentiation. Cell Stem hepatocyte generation in liver homeostasis and repair. Science.
Cell. 2014;15:340-­3 49. 2021;371:eabc4346.
25. Malato Y, Naqvi S, Schurmann N, et al. Fate tracing of mature 47. Wei Y, Wang YG, Jia Y, et al. Liver homeostasis is maintained by
hepatocytes in mouse liver homeostasis and regeneration. J Clin midlobular zone 2 hepatocytes. Science. 2021;371:eabb1625.
Invest. 2011;121:4850-­4860. 48. Rabes HM, Wirsching R, Tuczek HV, Iseler G. Analysis of cell cycle
26. Rodrigo-­Torres D, Affo S, Coll M, et al. The biliary epithelium gives compartments of hepatocytes after partial hepatecomy. Cell Tissue
rise to liver progenitor cells. Hepatology. 2014;60:1367-­1377. Kinet. 1976;9:517-­532.
27. Español-­Suñer R, Carpentier R, Van Hul N, et al. Liver progenitor 49. Miyaoka Y, Ebato K, Kato H, Arakawa S, Shimizu S, Miyajima A.
cells yield functional hepatocytes in response to chronic liver in- Hypertrophy and unconventional cell division of hepatocytes un-
jury in mice. Gastroenterology. 2012;143:1564-­1575.e7. derlie liver regeneration. Curr Biol. 2012;22:1166-­1175.
28. Reid LM. Stem/progenitor cells and reprogramming (plasticity) 50. Kim TH, Mars WM, Stolz DB, Michalopoulos GK. Expression and
mechanisms in liver, biliary tree, and pancreas. Hepatology. activation of pro-­MMP-­2 and pro-­MMP-­9 during rat liver regener-
2016;64:4-­7. ation. Hepatology. 2000;31:75-­82.
29. Raven A, Lu WY, Man TY, et al. Cholangiocytes act as facultative 51. Mars WM, Liu ML, Kitson RP, Goldfarb RH, Gabauer MK,
liver stem cells during impaired hepatocyte regeneration. Nature. Michalopoulos GK. Immediate early detection of urokinase recep-
2017;547:350-­354. tor after partial hepatectomy and its implications for initiation of
30. Magami Y, Azuma T, Inokuchi H, et al. Cell proliferation and re- liver regeneration. Hepatology. 1995;21:1695-­1701.
newal of normal hepatocytes and bile duct cells in adult mouse 52. Lorup C. An autoradiographic study of the 3H-­uridine and 3H-­
liver. Liver. 2002;22:419-­425. thymidine incorporation in the regenerating mouse liver. Cell
31. Jungermann K, Kietzmann T. Zonation of parenchymal and Tissue Kinet. 1977;10:477-­485.
nonparenchymal metabolism in liver. Annu Rev Nutr. 1996;16:​ 53. Stanulović VS, Kyrmizi I, Kruithof-­de Julio M, et al. Hepatic HNF4alpha
179-­2 03. deficiency induces periportal expression of glutamine synthetase and
32. Torre C, Perret C, Colnot S. Molecular determinants of liver zona- other pericentral enzymes. Hepatology. 2007;45:433-­444.
tion. Prog Mol Biol Transl Sci. 2010;97:127-­150. 54. Parviz F, Matullo C, Garrison WD, et al. Hepatocyte nuclear factor
33. Braeuning A, Ittrich C, Köhle C, et al. Differential gene expres- 4alpha controls the development of a hepatic epithelium and liver
sion in periportal and perivenous mouse hepatocytes. FEBS J. morphogenesis. Nat Genet. 2003;34:292-­296.
2006;273:5051-­5061. 55. Huck I, Gunewardena S, Espanol-­S uner R, Willenbring H, Apte
34. Kietzmann T. Metabolic zonation of the liver: the oxygen gradient U. Hepatocyte nuclear Factor 4 alpha activation is essen-
revisited. Redox Biol. 2017;11:622-­630. tial for termination of liver regeneration in mice. Hepatology.
35. Halpern KB, Shenhav R, Matcovitch-­Natan O, et al. Single-­cell spa- 2019;70:666-­6 81.
tial reconstruction reveals global division of labour in the mamma- 56. Hoehme S, Brulport M, Bauer A, et al. Prediction and validation
lian liver. Nature. 2017;542:352-­356. of cell alignment along microvessels as order principle to restore
36. Saviano A, Henderson NC, Baumert TF. Single-­cell genomics and tissue architecture in liver regeneration. Proc Natl Acad Sci U S A.
spatial transcriptomics: discovery of novel cell states and cellular 2010;107:10371-­10376.
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
1494 HUANG et al.

57. Walesky CM, Kolb KE, Winston CL, et al. Functional compensation 78. Wu H, Zhou X, Fu GB, et al. Reversible transition between hepato-
precedes recovery of tissue mass following acute liver injury. Nat cytes and liver progenitors for in vitro hepatocyte expansion. Cell
Commun. 2020;11:5785. Res. 2017;27:709-­712.
58. Nevzorova YA, Boyer-­Diaz Z, Cubero FJ, Gracia-­Sancho J. Animal 79. Schaub JR, Huppert KA, Kurial SNT, et al. De novo formation of
models for liver disease -­a practical approach for translational re- the biliary system by TGFβ-­mediated hepatocyte transdifferentia-
search. J Hepatol. 2020;73:423-­4 40. tion. Nature. 2018;557:247-­251.
59. De Minicis S, Kisseleva T, Francis H, et al. Liver carcinogenesis: 80. He J, Lu H, Zou Q, Luo L. Regeneration of liver after extreme he-
rodent models of hepatocarcinoma and cholangiocarcinoma. Dig patocyte loss occurs mainly via biliary transdifferentiation in ze-
Liver Dis. 2013;45:450-­459. brafish. Gastroenterology. 2014;146:789-­8 00.e8.
60. Issa R, Zhou X, Trim N, et al. Mutation in collagen-­1 that confers re- 81. Choi TY, Ninov N, Stainier DY, Shin D. Extensive conversion of
sistance to the action of collagenase results in failure of recovery hepatic biliary epithelial cells to hepatocytes after near total
from CCl4-­induced liver fibrosis, persistence of activated hepatic loss of hepatocytes in zebrafish. Gastroenterology. 2014;146:​
stellate cells, and diminished hepatocyte regeneration. FASEB J. 776-­788.
2003;17:47-­49. 82. Deng X, Zhang X, Li W, et al. Chronic liver injury induces con-
61. Wallace MC, Hamesch K, Lunova M, et al. Standard operating pro- version of biliary epithelial cells into hepatocytes. Cell Stem Cell.
cedures in experimental liver research: thioacetamide model in 2018;23:114-­122.e3.
mice and rats. Lab Anim. 2015;49:21-­29. 83. Russell JO, Lu WY, Okabe H, et al. Hepatocyte-­specific β-­c atenin
62. Fickert P, Stoger U, Fuchsbichler A, et al. A new xenobiotic-­ deletion during severe liver injury provokes Cholangiocytes to dif-
induced mouse model of sclerosing cholangitis and biliary fibrosis. ferentiate into hepatocytes. Hepatology. 2019;69:742-­759.
Am J Pathol. 2007;171:525-­536. 84. Benhamouche S, Decaens T, Godard C, et al. Apc tumor sup-
63. Preisegger KH, Factor VM, Fuchsbichler A, Stumptner C, Denk H, pressor gene is the "zonation-­keeper" of mouse liver. Dev Cell.
Thorgeirsson SS. Atypical ductular proliferation and its inhibition by 2006;10:759-­770.
transforming growth factor beta1 in the 3,5-­diethoxycarbonyl-­1,4-­d 85. Yang J, Mowry LE, Nejak-­Bowen KN, et al. β-­C atenin signaling
ihydrocollidine mouse model for chronic alcoholic liver disease. Lab in murine liver zonation and regeneration: a Wnt-­Wnt situation!
Invest. 1999;79:103-­109. Hepatology. 2014;60:964-­976.
64. Schaub JR, Malato Y, Gormond C, Willenbring H. Evidence against 86. Okabe H, Yang J, Sylakowski K, et al. Wnt signaling regulates
a stem cell origin of new hepatocytes in a common mouse model hepatobiliary repair following cholestatic liver injury in mice.
of chronic liver injury. Cell Rep. 2014;8:933-­939. Hepatology. 2016;64:1652-­1666.
65. Chen F, Jimenez RJ, Sharma K, et al. Broad distribution of hepato- 87. Sun T, Annunziato S, Bergling S, et al. ZNRF3 and RNF43 cooper-
cyte proliferation in liver homeostasis and regeneration. Cell Stem ate to safeguard metabolic liver zonation and hepatocyte prolifer-
Cell. 2020;26:27-­33.e4. ation. Cell Stem Cell. 2021;28:1822-­1837.e10.
66. Kaneko K, Kamimoto K, Miyajima A, Itoh T. Adaptive remodeling 88. Camargo FD, Gokhale S, Johnnidis JB, et al. YAP1 increases organ
of the biliary architecture underlies liver homeostasis. Hepatology. size and expands undifferentiated progenitor cells. Curr Biol.
2015;61:2056-­2066. 2007;17:2054-­2060.
67. Andersson ER. In the zone for liver proliferation. Science. 89. Yimlamai D, Christodoulou C, Galli GG, et al. Hippo pathway activ-
2021;371:887-­888. ity influences liver cell fate. Cell. 2014;157:1324-­1338.
68. Gordillo M, Evans T, Gouon-­Evans V. Orchestrating liver develop- 90. Verboven E, Moya IM, Sansores-­G arcia L, et al. Regeneration
ment. Development. 2015;142:2094-­2108. defects in yap and Taz mutant mouse livers are caused
69. Loft A, Alfaro AJ, Schmidt SF, et al. Liver-­f ibrosis-­a ctivated by bile duct disruption and cholestasis. Gastroenterology.
transcriptional networks govern hepatocyte reprogramming 2021;160:847-­8 62.
and intra-­h epatic communication. Cell Metab. 2021;33:1685-­ 91. Pepe-­Mooney BJ, Dill MT, Alemany A, et al. Single-­cell analysis of
1700.e9. the liver epithelium reveals dynamic heterogeneity and an essen-
70. Bou Saleh M, Louvet A, Ntandja-­Wandji LC, et al. Loss of hepato- tial role for YAP in homeostasis and regeneration. Cell Stem Cell.
cyte identity following aberrant YAP activation: a key mechanism 2019;25:23-­38.e8.
in alcoholic hepatitis. J Hepatol. 2021;75:912-­923. 92. Planas-­Paz L, Sun T, Pikiolek M, et al. YAP, but not RSPO-­LGR4/5,
71. Merrell AJ, Peng T, Li J, et al. Dynamic transcriptional and epi- signaling in biliary epithelial cells promotes a ductular reaction in
genetic changes drive cellular plasticity in the liver. Hepatology. response to liver injury. Cell Stem Cell. 2019;25:39-­53.e10.
2021;74:444-­457. 93. Li L, Krantz ID, Deng Y, et al. Alagille syndrome is caused by mu-
72. Yanger K, Zong Y, Maggs LR, et al. Robust cellular reprogram- tations in human Jagged1, which encodes a ligand for Notch1. Nat
ming occurs spontaneously during liver regeneration. Genes Dev. Genet. 1997;16:243-­251.
2013;27:719-­724. 94. Oda T, Elkahloun AG, Pike BL, et al. Mutations in the human
73. Michalopoulos GK, Barua L, Bowen WC. Transdifferentiation of Jagged1 gene are responsible for Alagille syndrome. Nat Genet.
rat hepatocytes into biliary cells after bile duct ligation and toxic 1997;16:235-­242.
biliary injury. Hepatology. 2005;41:535-­544. 95. Fiorotto R, Raizner A, Morell CM, et al. Notch signaling regulates
74. Han X, Wang Y, Pu W, et al. Lineage tracing reveals the Bipotency tubular morphogenesis during repair from biliary damage in mice.
of SOX9(+) hepatocytes during liver regeneration. Stem Cell Rep. J Hepatol. 2013;59:124-­130.
2019;12:624-­638. 96. Minnis-­Lyons SE, Ferreira-­González S, Aleksieva N, et al. Notch-­
75. Tarlow BD, Pelz C, Naugler WE, et al. Bipotential adult liver pro- IGF1 signaling during liver regeneration drives biliary epithelial
genitors are derived from chronically injured mature hepatocytes. cell expansion and inhibits hepatocyte differentiation. Sci Signal.
Cell Stem Cell. 2014;15:605-­618. 2021;14:eaay9185.
76. Katsuda T, Kawamata M, Hagiwara K, et al. Conversion of termi- 97. Sekiya S, Suzuki A. Intrahepatic cholangiocarcinoma can arise
nally committed hepatocytes to culturable bipotent progenitor from notch-­ mediated conversion of hepatocytes. J Clin Invest.
cells with regenerative capacity. Cell Stem Cell. 2017;20:41-­55. 2012;122:3914-­3918.
77. Kim Y, Kang K, Lee SB, et al. Small molecule-­mediated repro- 98. Michalopoulos GK, Bhushan B. Liver regeneration: biological and
gramming of human hepatocytes into bipotent progenitor cells. J pathological mechanisms and implications. Nat Rev Gastroenterol
Hepatol. 2019;70:97-­107. Hepatol. 2021;18:40-­55.
|

14783231, 2022, 7, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/liv.15174 by Egyptian National Sti. Network (Enstinet), Wiley Online Library on [27/02/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
HUANG et al. 1495

99. Campana L, Esser H, Huch M, Forbes S. Liver regeneration and in- 104. Solar M, Cardalda C, Houbracken I, et al. Pancreatic exocrine duct
flammation: from fundamental science to clinical applications. Nat cells give rise to insulin-­producing beta cells during embryogenesis
Rev Mol Cell Biol. 2021;22:608-­624. but not after birth. Dev Cell. 2009;17:849-­860.
100. Rafii S, Butler JM, Ding BS. Angiocrine functions of organ-­specific 105. van Amerongen R, Bowman AN, Nusse R. Developmental
endothelial cells. Nature. 2016;529:316-­325. stage and time dictate the fate of Wnt/β-­c atenin-­r esponsive
101. Cordero-­Espinoza L, Dowbaj AM, Kohler TN, et al. Dynamic cell stem cells in the mammary gland. Cell Stem Cell. 2012;11:​
contacts between periportal mesenchyme and ductal epithe- 387-­4 00.
lium act as a rheostat for liver cell proliferation. Cell Stem Cell.
2021;28:1907-­1921.e8.
102. Dhawan A, Puppi J, Hughes RD, Mitry RR. Human hepatocyte How to cite this article: Huang R, Zhang X, Gracia-­Sancho J &
transplantation: current experience and future challenges. Nat Rev Xie W-F. Liver regeneration: Cellular origin and molecular
Gastroenterol Hepatol. 2010;7:288-­298.
mechanisms. Liver Int. 2022;42:1486–1495. doi: 10.1111/
103. Means AL, Xu Y, Zhao A, Ray KC, Gu G. A CK19(CreERT) knockin
mouse line allows for conditional DNA recombination in epithelial liv.15174
cells in multiple endodermal organs. Genesis. 2000;2008(46):318-­323.

You might also like