Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 1.
Published in final edited form as: Nat Rev Gastroenterol Hepatol. 2020 Oct 13;18(2):131–142. doi: 10.1038/s41575-020-00365-6

Maladaptive regeneration — the reawakening of developmental pathways in NASH and fibrosis

Changyu Zhu 1,2, Ira Tabas 1, Robert F Schwabe 1, Utpal B Pajvani 1,
PMCID: PMC7854502  NIHMSID: NIHMS1655872  PMID: 33051603

Abstract

With the rapid expansion of the obesity epidemic, nonalcoholic fatty liver disease is now the most common chronic liver disease, with almost 25% global prevalence. Nonalcoholic fatty liver disease ranges in severity from simple steatosis, a benign ‘pre-disease’ state, to the liver injury and inflammation that characterize nonalcoholic steatohepatitis (NASH), which in turn predisposes individuals to liver fibrosis. Fibrosis is the major determinant of clinical outcomes in patients with NASH and is associated with increased risks of cirrhosis and hepatocellular carcinoma. NASH has no approved therapies, and liver fibrosis shows poor response to existing pharmacotherapy, in part due to an incomplete understanding of the underlying pathophysiology. Patient and mouse data have shown that NASH is associated with the activation of developmental pathways: Notch, Hedgehog and Hippo–YAP–TAZ. Although these evolutionarily conserved fundamental signals are known to determine liver morphogenesis during development, new data have shown a coordinated and causal role for these pathways in the liver injury response, which becomes maladaptive during obesity-associated chronic liver disease. In this Review, we discuss the aetiology of this reactivation of developmental pathways and review the cell-autonomous and cell-non-autonomous mechanisms by which developmental pathways influence disease progression. Finally, we discuss the potential prognostic and therapeutic implications of these data for NASH and liver fibrosis.


Obesity and its metabolic consequences are among the most pressing health problems to date, with alarming projections for the future1,2. Of these metabolic consequences, nonalcoholic fatty liver disease (NAFLD), defined by excess liver fat, has quickly become the most common chronic liver disease, with an estimated global prevalence of 25%35. NAFLD ranges widely in severity, from simple steatosis, a prevalent and reversible state, to the necroinflammatory changes that characterize nonalcoholic steatohepatitis (NASH)6. NASH has no approved pharmacotherapy. As such, it is currently the second most common, yet the fastest growing, indication for liver transplantation in the United States7. However, livers available for transplantation are already limited and will not keep pace with the expected growth in the prevalence of NASH. Thus, new pharmaceutical targets are a great unmet need for an increasingly obese population and demand a comprehensive understanding of the pathophysiology of the disease.

Unlike other chronic liver diseases, the pathogenesis of NASH can be viewed as substrate-overload liver injury induced by an excessive lipid flux and increased de novo lipogenesis in hepatocytes as a result of obesity. Hepatocyte injury in turn provokes a multicellular repair response in which immune cells are activated locally or recruited from the bloodstream, the extracellular matrix (ECM) is remodelled to provide support, and hepatocytes proliferate to compensate for the lost functional hepatic mass8. Although this response is fundamentally similar to the response to viral infections or toxin exposure, chronic overnutrition, unlike such acute insults, can induce a persistent regenerative response, causing extensive accumulation of ECM and disrupting normal hepatic structure and function9. The resultant liver fibrosis is the strongest predictor of long-term clinical outcomes in patients with NAFLD–NASH1014 and an important end point in NASH clinical trials15. Fibrosis also increases the risk of development of end-stage liver disease (cirrhosis) as well as of hepatocellular carcinoma (HCC)16, the third-leading cause of cancer-related deaths worldwide17.

Developmental pathways, including Notch18, Hedgehog19 and Hippo–YAP–TAZ2022, are evolutionarily programmed to coordinate morphogenesis, lineage specification of hepatic progenitors and zonation establishment during development23 but are thought to be quiescent in healthy liver unless ‘reactivated’ in acute liver injury to organize regeneration. However, studies over the past decade suggest that these pathways are persistently activated in obesity to cope with the chronic insult and that they contribute to the development of fibrosis. In this Review, we summarize the interwoven role of these developmental pathways in normal liver morphogenesis as well as in the wound-healing response. We also discuss how these protective mechanisms go awry in obesity-associated liver pathology and evaluate the potential therapeutic opportunities to target their reawakening.

Developmental signalling and morphogenesis

Notch

Signal transduction via Notch is dependent on juxtacrine interactions between cells expressing Notch receptors (NOTCH1–4) and cell surface-tethered Notch ligands (Jagged1 and Jagged2 or Delta-like ligands 1, 3 and 4)24. Productive ligand–receptor interactions result in receptor proteolytic cleavage and release of the Notch intracellular domain, which cooperates with recombination signal binding protein for immunoglobulin-κJ region (RBPJ) and its co-activator Mastermind (MAM) to activate canonical Notch targets25 such as Hairy and enhancer of split (HES) as well as HES-related with YRPW motif (HEY) genes, which are transcriptional repressors that regulate stem cell maintenance, boundary formation and cell-fate decisions26 (FIG. 1a).

Fig. 1 |. Overview of developmental pathways in mammalian cells.

Fig. 1 |

a | Notch signalling. Canonical Notch signalling is activated by the ligand-to-receptor interaction between two neighbouring cells. Upon ligand binding, Notch receptor undergoes sequential cleavages by a disintegrin and metalloproteinase (ADAM) protease and γ-secretase, leading to the release and nuclear translocation of the Notch intracellular domain (NICD). NICD interacts with immunoglobulin-κJ region (RBPJ) and Mastermind (MAM) to initiate the transcription of downstream targets such as the HES and HEY family of genes. Notch receptor can also induce non-canonical signalling by regulating β-catenin protein degradation. b | Hippo–YAP–TAZ signalling. In the Hippo-off state, the inactive STK3/4–LATS1/2 cascade is unable to phosphorylate YAP and TAZ. Unphosphorylated stable YAP and TAZ translocate into the nucleus and bind to the transcription factor TEAD family to regulate transcription. c | Hedgehog signalling. Hedgehog (HH) ligand binds to the cell-surface receptor Patched (PTCH), releasing its inhibition of Smoothened (SMO). Active SMO prevents the phosphorylation and cytoplasmic sequestration of GLI protein by kinases including protein kinase A (PKA), glycogen synthase kinase 3β (GSK3β) and casein kinase 1 (CK1). Stabilized GLI enters the nucleus and promotes Hedgehog target gene expression. d | WNT–β-catenin signalling. Binding of the WNT ligand to its receptor Frizzled and co-receptor low-density lipoprotein receptor-related protein 5 or 6 (LPR5/6) recruits the destruction complex, consisting of axin, adenomatous polyposis coli (APC), and the kinases GSK3β and CK1α, to the cell membrane via Dishevelled (DVL). This coordinated effort prevents the destruction complex from phosphorylating β-catenin, which facilitates its degradation. Stabilized β-catenin translocates to the nucleus, binds to the T cell factor (TCF) transcription factor family and promotes the expression of WNT target genes.

Notch is a well-established regulator of intrahepatic bile duct development in vertebrates. It is required for the commitment to the biliary lineage by the embryonic liver bipotential progenitor (hepatoblast) during the initial single-layer ductal plate formation at embryonic day 14.5 (E14.5) as well as for ductal morphogenesis from E16.5 until postnatal day 2 during mouse liver development23. These requirements are underscored by the finding of Notch signalling defects in patients with Alagille syndrome, a congenital disease characterized by bile duct paucity that results in cholestasis, with additional heart and kidney pathology27. Consistently, studies in rodent models revealed that hepatoblast NOTCH2, the most abundant Notch receptor during liver development28, and Jagged1 from portal fibroblasts29 form the essential receptor–ligand pair for biliary lineage specification and morphogenesis30,31. Bypassing this ligand requirement by expression of a constitutively active form of Notch in hepatoblasts directs cholangiocyte differentiation at the expense of hepatocytes32. Notch activation represses the expression of hepatocyte-enriched transcription factors, including hepatocyte nuclear factor 4α (HNF4α) and HNF1α, with a reciprocal upregulation of cholangiocyte-rich transcription factors such as HNF1β and sex-determining region Y-box 9 (SOX9), which in turn interact with HNF6 (REF.33) and SOX4 (REF.34). Conversely, RBPJ deletion in embryonic hepatoblasts prior to E14.5 prevents biliary fate commitment and even later deletion (after E16.5) disrupts tubulogenesis and the 3D structures of intrahepatic bile ducts32,35. The sum total of these actions is to favour a biliary cell fate36.

Hippo–YAP–TAZ

Hippo is considered a tumour suppressor pathway that responds to cell density and regulates organ size37. Upon activation, the mammalian counterparts of the Hippo kinases found in Drosophila, serine/threonine-protein kinase 3 (STK3) and STK4, together with the scaffold protein salvador homologue 1 (SAV1), phosphorylate the kinases large tumour suppressor homologue 1 (LATS1) and LATS2. Activated LATS1 or LATS2, together with their scaffold protein (MOB kinase activator 1A (MOB1A) or MOB1B), phosphorylate the transcriptional co-activators YAP and TAZ38. Phosphorylated YAP and TAZ are targeted for cytoplasmic retention and proteasomal degradation. The inhibition of Hippo signalling results in stabilization and nuclear translocation of YAP and TAZ, which bind to the TEAD family of transcription factors and regulate a myriad of cellular processes39 (FIG. 1b).

In liver development, hepatoblast deletion of Yap1 impairs bile duct development and the survival of hepatocytes40. Reciprocally, constitutive YAP–TAZ signalling in hepatoblasts promotes a biliary lineage commitment and ductal plate expansion in mice41.YAP also maintains the progenitor states of hepatoblasts by promoting HNF4α and forkhead box protein A2 (FOXA2) interactions with enhancers of embryonic genes before hepatocyte differentiation during liver development42. In adult mouse liver, YAP protein expression and activity is highly enriched in cholangiocytes43 and display a gradient in the hepatic parenchyma, with the highest activity near the portal triads and the lowest near the central vein44.

Hedgehog

Signal transduction via Hedgehog is initiated by the binding of Hedgehog ligand to the receptor Patched to release Smoothened, which stabilizes the Hedgehog-responsive transcription factor GLI45 (FIG. 1c). During mammalian embryonic development, Sonic hedgehog (SHH) is highly expressed in the ventral foregut endoderm, the origin of liver and pancreas46. Although the role of Hedgehog in liver development is not clearly defined, Shh expression disappears at the onset of liver bud formation, transiently increases in fetal hepatoblasts and then rapidly decreases again as progenitors differentiate into hepatocytes47, suggesting that Hedgehog signal activity is temporally regulated and restricted to ensure progenitor expansion and differentiation.

WNT–β-catenin

When biologically active WNT ligands bind to the cell-surface receptor Frizzled, β-catenin translocates to the nucleus, where it binds to T cell factor (TCF) family transcription factors. In the absence of WNT ligands, β-catenin is phosphorylated by its destruction complex, including scaffold proteins AXIN1 and AXIN2, leading to its degradation48 (FIG. 1d). During liver development, WNT–β-catenin signalling is crucial to the survival, proliferation and maturation of hepatoblasts. That is, early deletion of Ctnnb1 (the gene encoding β-catenin) in embryonic mouse hepatoblasts at E9.5 produces multiple defects, including intrahepatic bile duct paucity and disrupted hepatocyte maturity49. Deletion of Ctnnb1 after E16.5 in embryogenesis (by use of albumin-Cre rather than Foxa3-Cre) results in a milder phenotype, including impaired hepatocyte proliferation and postnatal liver mass50. WNT signalling also establishes liver zonation as β-catenin competes with HNF4α for binding to TCF to direct pericentral versus periportal gene expression in mouse hepatocytes51. Thus, persistent stabilization of β-catenin results in a pericentral phenotype across the liver lobule52, whereas hepatocyte-specific loss of WNT–β-catenin signalling in mice induces a wide-spread periportal signature53,54. Pericentral hepatocytes have high expression of WNT–β-catenin targets such as glutamine synthetase and cytochrome P450 enzymes55, which perform essential zonal metabolic functions.

Developmental signalling in liver maturation

It has become evident that a highly spatiotemporally regulated and interactive network of developmental pathways exists to precisely guide normal hepatocyte and biliary maturation. For example, β-catenin gain-of-function impairs cholangiocyte differentiation and duct morphogenesis in mice56, a phenotype that mirrors Notch loss-of-function mice32. These findings are consistent with data showing Notch-induced β-catenin degradation57 as well as the general concept that Notch and β-catenin favour opposing cell-fate decisions from the same progenitor. Notch and WNT–β-catenin also cooperate in a cell-non-autonomous way to regulate biliary maturation — in zebrafish, β-catenin induces hepatocyte Notch ligand expression to activate cholangiocyte Notch receptors and induce bile duct maturation58.

The literature is replete with other, similar examples of pathway crosstalk in liver development. Portal mesenchyme-derived transforming growth factor-β (TGFβ) establishes a gradient that induces the differentiation of portal hepatoblasts into cholangiocytes59 as well as the transdifferentiation of portal mesenchymal cells into portal myofibroblasts, the latter of which results in increased Jagged1, which activates hepatoblast Notch receptors adjacent to the portal vein60. Active YAP–TAZ signalling favours biliary cell fate by repression of HNF4α and upregulation of TGFβ41, which could interact with Notch in bile duct formation, as discussed earlier. YAP–TAZ and WNT–β-catenin signalling activities display opposite zonal distribution in the liver lobule. Liver-specific Yap1 deletion in mice results in WNT-active, glutamine synthetase-positive hepatocyte expansion into the periportal region, whereas constitutive YAP activation causes disruption of metabolic zonation and downregulation of pericentral genes44. The spatiotemporal regulation of developmental signalling and the coordination between these pathways underscore the precise nature of normal liver development (FIG. 2).

Fig. 2 |. A network of developmental signalling controls lineage commitment during mouse liver development.

Fig. 2 |

Liver development is regulated by a highly controlled network of pathways in a spatiotemporal manner. Near the portal vein, portal fibroblast-derived Jagged1 activates NOTCH2 on hepatoblasts to induce biliary differentiation, in conjunction with transforming growth factor-β (TGFβ) and YAP–TAZ, by transactivation of biliary lineage-defining factors, including SOX9, hepatocyte nuclear factor 1β (HNF1β) and HNF6. In the absence of these signals, increased HNF1α and HNF4α promote a hepatocyte lineage. In parallel, responding to WNT signals near the central vein, β-catenin activates pericentral genes to establish normal liver zonation.

Liver injury response and regeneration

Hepatocyte replenishment after liver injury

The liver can fully restore its mass and synthetic function after toxin-induced acute injury or surgical resection. One of the most frequently applied rodent models is partial hepatectomy, in which the massive hepatocyte loss is recovered primarily by proliferation and hypertrophy of the remaining hepatocytes61. Similar lineage tracing and pulse-chase methods have shown that hepatocytes also display a proliferative capability to repopulate themselves in mouse models of chronic liver disease62,63. Although several studies suggest that all hepatocytes proliferate modestly to participate in regeneration without zonal preference64,65, other studies suggest that specialized subsets (that is, AXIN2+ pericentral66, SOX9+ periportal67 and zone-non-specific hepatocytes with high telomerase reverse transcriptase (TERT) activity68) or leucine-rich repeat-containing G protein-coupled receptor 5-positive (LGR5+) cells69 might preferentially be involved.

By contrast, cholangiocytes and cholangiocytederived oval cells, a biliary hyperplastic response to chronic injury, rarely give rise to hepatocytes7072. However, when hepatocyte proliferation is severely compromised, such as in mice with hepatocyte-specific ablation of genes essential for this proliferative response7375, cholangiocytes can differentiate into hepatocytes as a fail-safe mechanism.

Post-partial hepatectomy

By promoting hepatocyte survival and proliferation, developmental pathways are crucial to liver regeneration after partial hepatectomy. Consistent with its role in maintaining hepatoblast proliferation during development, β-catenin rapidly accumulates in the nucleus within minutes after partial hepatectomy76 and drives the expression of cell cycle-related genes during rat liver regeneration77. Thus, the deletion of Ctnnb1 in hepatocytes renders a delay in regeneration50. Both Hedgehog and YAP–TAZ signalling also respond to partial hepatectomy. Hedgehog ligand expression increases transiently after partial hepatectomy, leading to peak activity at the time of maximal hepatocyte proliferation78. Similarly, YAP activity increases within 24 hours in response to partial hepatectomy79, with both YAP and TAZ being required for the coordination of normal regeneration80,81. Hedgehog and YAP also cooperate in cell-autonomous and cell-non-autonomous ways to facilitate post-partial hepatectomy regeneration: Hedgehog pathway induces YAP signal activity and the activation of myofibroblasts, which is required for the proliferative response in hepatocytes with nuclear YAP accumulation, while inhibition of Hedgehog pathway in myofibroblasts impairs hepatocyte proliferation following partial hepatectomy82. By contrast, administration of Indian Hedgehog (IHH) protein after parenchymal resection accelerates liver regeneration in mice83. Finally, liver Notch signalling is also increased after partial hepatectomy and small interfering RNA (siRNA) directed to Jag1 or Notch1 impairs hepatocyte proliferation during rat liver regeneration84. However, these siRNA findings are due to impaired Notch signalling in liver sinusoidal endothelial cells rather than in hepatocytes, in which fine-tuned Notch signalling is required for the regulation of endothelial identity and homeostasis85 in both normal liver and during regeneration in mice86,87.

Hepatocyte reprogramming in liver diseases

Unlike post-partial hepatectomy regeneration, emerging evidence suggests that mature hepatocytes maintain plasticity and utilize Notch-mediated reprogramming to adopt features of cholangiocytes and/or progenitors in chronic liver injury. This observation is exemplified by mouse models of forced Notch activation in mature hepatocytes, which results in ‘biphenotypic’ hepatocytes with the expression of biliary markers, such as SOX9 and osteopontin (OPN), that are normally absent in hepatocytes32,88. Consistently, endogenous Notch activity in mature hepatocytes is necessary for transdifferentiation into cytokeratin 19-positive (CK19+) cells that morphologically resemble cholangiocytes in response to cholestasis induced by bile duct ligation or by exposure to the biliary toxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine88, likely mediated by myofibroblast-borne Jagged1 and opposed by macrophage-derived WNT signals89. Similarly, methionine choline-deficient diet-feeding in mice, which causes obesity-independent steatohepatitis, induces hepatocyte Sox9 expression and biliary transdifferentiation in a subset of hepatocytes90, suggesting that Notch-mediated cell fate reprogramming is prevalent in different models of liver injury.

The biological role of cell fate conversion is intriguing. One possibility is that hepatocytes transdifferentiate to counteract the loss of cholangiocyte functions in cholestatic injury. In line with this idea, mice with hepatoblast-specific simultaneous deletion of Rbpj and Hnf6 lack peripheral bile ducts91 but are able to recover from postnatal cholestasis due to a functional biliary system formed by transdifferentiated hepatocytes in a Notch-independent process driven by TGFβ signalling92. This finding redefines the plasticity of hepatocytes as a possible path to biliary regeneration in extreme cases of intrahepatic bile duct deficiency such as in human Alagille syndrome. Similarly, in response to bile acids and cholestatic insults, signalling mediated by YAP is required for the transformation of hepatocytes into biliary progenitors in mouse models93,94. However, ductular reaction — that is, the oval cell response, characterized by the expansion of biliary cells in the parenchyma frequently seen in NASH and other chronic liver diseases95 — derives primarily from cholangiocytes, with only a minor contribution from hepatocytes7072. This observation suggests an alternative purpose for injury-induced cellular plasticity: an evasion mechanism by which cells retain their regenerative ability after insults. Evidence for this hypothesis arises from chimeric lineage-tracing models that reveal hepatocyte-derived ductal cells that resemble bipotential progenitors and are capable of re-differentiating back into hepatocytes upon the withdrawal of insults96.

These studies demonstrate the plasticity of hepatocytes and their regulation by reactivated developmental signalling pathways (FIG. 3). These findings are likely to translate to patients as biphenotypic cells are observed in human chronic liver diseases97,98, suggestive of cell identity conversion between hepatocytes and cholangiocytes or bipotential liver progenitors. Importantly, developmental pathways require precise control — for example, Notch can induce hepatocyte fate change in response to insults. Hepatocyte YAP can cooperate in dedifferentiation via the upregulation of Notch2 and Jag1; reprogrammed cells clonally expand and regenerate the hepatocyte compartment upon removal of activated YAP after in vivo engraftment into fumarylacetoacetate hydrolase-deficient (Fah−/−) mice43, a model of liver failure that can be rescued by successful repopulation of functional hepatocytes99. In addition, YAP can activate cell death in injured mouse hepatocytes and proliferation in undamaged ones, a selective mechanism required for homeostasis and appropriate regeneration in mice100. However, hepatocyte Notch and YAP must be turned off to enable the re-differentiation and repopulation upon injury cessation. It is possible that dysregulation of these signalling pathways occurs in chronic liver diseases such as NASH to transform a regenerative process into one leading to fibrosis and cancer.

Fig. 3 |. Cellular reprogramming during mouse liver regeneration.

Fig. 3 |

In response to acute injury, hepatocytes proliferate to maintain liver functional capacity. However, when the insult persists, Notch and YAP signalling pathways are activated in a subset of hepatocytes, resulting in ‘biphenotypic’ cells with the expression of biliary and progenitor markers such as SOX9 and osteopontin (OPN). Biphenotypic cells can dedifferentiate into progenitor-like cells as a strategy to evade the insult, with the potential to re-differentiate back into hepatocytes upon injury cessation. Alternatively, biphenotypic cells can also transdifferentiate into cholangiocytes to cope with injury-induced cholestasis. When injury persists, ductular reaction or oval cell response is induced, originated mainly from cholangiocytes, which can contribute to liver regeneration when hepatocyte proliferative capacity is severely compromised. In the absence of hepatoblast Notch and hepatocyte nuclear factor 6 (HNF6), transforming growth factor-β (TGFβ) can functionally substitute Notch signalling to form a functional biliary system.

NASH and fibrosis: maladaptive regeneration?

Multiple hits in NASH pathogenesis

Unlike other chronic liver diseases, NASH is primarily the result of ‘multiple hits’ induced by obesity, wherein parallel pathogenic events interact with host genetics, environmental factors and crosstalk between tissues8,101,102. Insulin resistance leads to liver steatosis — in a susceptible individual, the resultant lipotoxicity induces mitochondrial dysfunction, endoplasmic reticulum stress, and hepatocellular injury and death103,104. Damaged and necrotic hepatocytes in turn leak cytokines and damage-associated molecular patterns that drive immune cell infiltration and hepatic stellate cell (HSC) activation to coordinate the wound-healing effort that replaces dead hepatocytes105,106. Transient and reversible deposition of ECM during acute injury provides mechanical stability and serves as a scaffold to guide cells for the restoration of normal liver architecture107. In fact, the inhibition of HSC activity after partial hepatectomy blunts ECM production and paracrine signals that support hepatocyte proliferation82. NASH, however, is associated with chronic HSC activation, resulting in a continuous accumulation of ECM108 and progressive substitution of the liver parenchyma by fibrous tissue.

Developmental pathways in NASH

Several mouse models that recapitulate the full spectrum of NASH pathology have been described21,109113, enabling investigation of the potential role of developmental pathways in disease pathogenesis (FIG. 4).

Fig. 4 |. Pathogenesis of nasH and fibrosis in mouse models.

Fig. 4 |

As a result of chronic substrate overload, some steatotic hepatocytes undergo cell death due to sustained lipotoxicity and endoplasmic reticulum (ER) stress, which releases factors such as damage-associated molecular patterns (DAMPs) and Hedgehog (HH) ligands, which activate resident Kupffer cells and hepatic stellate cells (HSCs), resulting in inflammatory and fibrogenic responses. Other hepatocytes become reprogrammed by Notch, HH and YAP–TAZ to initiate a maladaptive response that induces hepatocyte secretion of fibrogenic factors such as osteopontin (OPN) and HH ligands to activate HSCs. This process creates a regenerative niche that aggravates fibrosis as well as compensatory proliferation to restore loss of mass. A by-product of this proliferative response is genomic instability, which might contribute to tumorigenesis. HCC, hepatocellular carcinoma; IHH, Indian Hedgehog; NASH, nonalcoholic steatohepatitis; NICD, Notch intracellular domain.

Hedgehog.

The first developmental pathway to be linked to the pathogenesis of NASH is Hedgehog signalling, which increases in hepatocytes, cholangiocytes and HSCs in response to injury to coordinate the wound-healing effort114, likely due to increased Hedgehog ligand expression from a near-zero basal level in normal livers115. Whereas Hedgehog signalling can be restorative after partial hepatectomy78, Hedgehog signalling contributes to various NASH pathologies, including inflammation and fibrosis in mouse models116118. Dysregulation of Hedgehog might even exacerbate obesity-associated steatosis as in vivo mouse studies have revealed an intersection between Hedgehog–GLI protein signalling with the circadian clock to regulate hepatic lipid metabolism119,120. Importantly, liver Hedgehog activity is evident in human NASH and positively correlates with disease severity and fibrosis stage121. SHH expression is increased in ballooned hepatocytes and drives HSC activation and ductular reaction122. Thus, in histological analyses of human liver biopsy samples from the PIVENS trial, which compared pioglitazone, vitamin E and placebo for the treatment of patients with NASH but without diabetes, it was observed that improved liver injury and fibrosis was associated with a reduction of SHH+ hepatocytes123. Similarly, liver IHH is increased in patients with NASH compared with normal and steatotic livers21.

Hippo–YAP–TAZ.

Aberrant activity of YAP and TAZ is implicated in the pathogenesis of chronic liver diseases. For example, one study explored the role of autophagy in the regulation of YAP and found that hepatocyte-specific deletion of Atg7 resulted in impaired autophagy, liver fibrosis and cancer development, driven by increased levels of the autophagy substrate YAP in mice124. Hepatocyte YAP and TAZ expression are also closely linked to NASH. TAZ protein is increased in hepatocytes derived from patients with NASH compared with simple steatosis and, in mouse models fed a NASH-promoting diet, hepatocyte TAZ action promotes the steatosisto-NASH transition by inducing injury, inflammation and fibrosis, largely mediated by increased secretion of IHH, a direct transcription target of TAZ21. Another study demonstrated that YAP and TAZ are activated in metabolically stressed hepatocytes in mouse models and in patients with NASH, inducing the expression of cysteine-rich angiogenic inducer 61 (CYR61), which is a chemokine that promotes inflammation and fibrosis22.

YAP is also an important regulator of non-parenchymal cell biology in liver diseases125. YAP drives HSC activation and a fibrogenic programme126,127, regulated by Hedgehog signalling82,128. YAP is also essential to cholangiocyte homeostasis and contributes to ductular proliferation upon injury93,94. Accordingly, YAP+ ductular reaction cells accumulate in human NASH, which is positively correlated with fibrosis129.

WNT–β-catenin.

Although WNT–β-catenin regulates hepatocyte proliferation after partial hepatectomy130, its role in NASH and fibrosis is less well defined. However, multiple indirect effects exist. Hepatocyte WNT–β-catenin signalling regulates lipogenesis131 and mitochondrial functions132 and interacts with FOXO1 to increase gluconeogenesis133. The effects of WNT–β-catenin in non-parenchymal cells are similarly unclear, with most evidence coming from in vitro studies that have contradictory outcomes. For example, β-catenin activation is required for HSC quiescence in cultured primary rat HSCs134 but knockdown of β-catenin can inhibit the proliferation and fibrogenic activation of a rat HSC line135.

Notch.

Hepatocyte Notch activity is absent in healthy livers but upregulated in obesity, where it contributes to maladaptive outcomes136. The post-developmental role of Notch in the liver was first discovered in genetic (leptin-deficient ob/ob mice) or diet-induced mouse models of obesity, in which Notch was found to increase hepatic glucose production by co-activating FOXO1 at gluconeogenic promoters112. Notch was subsequently shown to activate de novo lipogenesis and increase hepatic steatosis by activation of the mechanistic target of rapamycin complex 1 (mTORC1) in mice fed a high-fat diet137,138. These data suggest that Notch might uncouple these two insulin-regulated processes (de novo lipogenesis and hepatic glucose production) in liver, which is rare among mouse models of altered insulin action. For example, FOXO1-deficient mice show reduced glucose production but increased de novo lipogenesis139.

Studies suggest an additional, obesity-independent and lipid-independent effect of hepatocyte Notch signalling on liver pathology. Forced hepatocyte Notch activation induces OPN expression and secretion, which activates HSCs and provokes marked fibrosis even in lean mice despite unchanged liver triglyceride content. Conversely, mice with hepatocyte-specific Notch loss of function are protected from NASH-induced pathology18. Importantly, liver Notch activity, as assessed by downstream transcriptional target expression of the HES and HEY families, is also positively correlated with NAFLD and NASH severity in patients140. Hepatocyte HES1 staining is higher in livers with NASH and fibrosis than in healthy ones and, like SHH, is inversely correlated with treatment response in patients from the PIVENS trial18.

Notch activity in non-parenchymal cells can also contribute to NASH and other chronic liver diseases through a variety of proposed mechanisms. Notch promotes a pro-inflammatory (that is, ‘M1’) polarization of macrophages in CCl4-induced injury141 and alcohol-induced injury mouse models142. Notch also seems to have a cell-autonomous effect in HSCs as γ-secretase inhibitor treatment of CCl4-treated rats143 or a siRNA against Notch3 in a rat HSC line144 repressed fibrogenic gene expression. Similarly, HSC Notch inhibition dampens Hedgehog activity and the HSC–myofibroblast transition in cultured primary mouse HSCs and suppression of HSC Hedgehog signalling results in downregulation of Notch activity and in accumulation of myofibroblasts in a mouse model of bile duct ligation145. In addition, a study using mice with HSC-specific overexpression and deletion of hyaluronan synthase 2 found that hyaluronan activates HSC Notch via CD44 signalling, whereas HSC-specific Notch1 deletion attenuated bile duct ligation-induced fibrosis, providing a causal link between hyaluronan, HSC Notch and fibrosis development146. These data suggest that both hepatocyte and non-parenchymal cell Notch activity can activate HSCs to create a fibrotic niche for regeneration.

Future directions and therapeutics

Unanswered questions

The studies discussed enhance our understanding of the pathogenesis of NASH but there remain multiple unanswered questions. First, what is the mechanism by which developmental pathways are induced in response to injury or NASH? That is, what are the upstream signals that activate hepatocyte Notch and YAP–TAZ activity as well as WNT and Hedgehog ligand production in injured livers? Additionally, how are these signals turned off correctly in normal development and regeneration? Owing to the contextual importance of cell type and injury-induced activation, it is unlikely that these data can be derived using in vitro modelling, although data from stem cell-derived liver organoids are encouraging147.

In addition, developmental pathways show a coordinated response in cellular reprogramming in the liver43,82 but the mechanisms underlying this response are unclear. One intriguing idea is that of a ‘regulon’, in which multiple genes are controlled by the same regulatory molecule. For example, insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) regulates the expression of thousands of genes in HSCs via interactions with mRNAs involved in proliferation and transdifferentiation148. Upon acute injury, hepatocyte IGF2BP3 modulates the expression of fetal markers required for regeneration, with checks and balances provided, in part, by YAP to achieve a balanced regenerative response in both hepatocytes and HSCs149. The dysregulation of this pathway might theoretically result in acute liver failure and fibrosis but the specific role of this regulon and others in the wound-healing response in NASH requires further study.

Future directions — NASH-driven HCC?

Although a small percentage (2.4% to 12.8% depending on the cohort) of patients with NAFLD develop HCC1014, NASH is the most rapidly growing contributor to HCC in the United States150,151. Pathogenic mechanisms underlying NASH-induced HCC have been comprehensively reviewed elsewhere152, with a focus on the role of DNA damage response, inflammation, autophagy and the gut microbiome. However, the chronic activation of developmental pathways in obesity suggests another mechanistic link between NASH and HCC. In NASH, these pathways could provoke uncontrolled hepatocyte proliferation and genomic instability, while simultaneously altering the surrounding microenvironment to one that favours malignant transformation and tumour growth. In fact, several groups have documented roles for these developmental pathways in liver tumorigenesis. Notch accelerates tumour proliferation via the direct transcriptional activation of cell cycle-related genes153 and insulin-like growth factor 2 (IGF2)-mediated mitogenic signalling154 to drive liver oncogenesis in mice. YAP–TAZ rewires glutamine metabolism to support proliferation155, while inducing NUAK2 to increase tumour growth156 and the chemokine monocyte chemoattractant protein 1 (MCP1; also known as CCL2) to promote an inflammatory tumour microenvironment157,158. β-Catenin pathway mutations, frequently seen in human HCCs, induce a unique metabolism that depends on fatty acids and glutamine to sustain tumour proliferation159,160 but also help tumour cells to evade T cell surveillance and resist checkpoint inhibitors161163. Intriguingly, the interactions between these pathways that are seen in development and regeneration are also preserved in HCC — for example, Notch is required for YAP–TAZ activity in hepatocarcinogenesis, while β-catenin signalling inhibits the tumorigenic functions of the YAP–TAZ–Notch signalling loop164 (FIG. 5).

Fig. 5 |. The role of developmental pathways in liver cancer.

Fig. 5 |

In the injured and fibrotic liver, hepatocyte developmental pathway activity results in proliferation and cellular transformation. Notch activation induces a range of cell-cycle genes, the autocrine mitogen Igf2, and biliary and progenitor-associated genes (that is, Sox9 and Spp1) that regulate the tumour microenvironment (TME) and cellular plasticity. Enhanced cell–cell contact and extracellular matrix (ECM) stiffness as well as a variety of nutrient factors, such as bile acids and cholesterol, increase YAP–TAZ stability, nuclear translocation, and transcription factor TEAD activity to augment proliferation and influence cellular metabolism and the TME to favour tumorigenesis, and might drive further Notch activity. In parallel, β-catenin activation, in part due to driver mutations in Ctnnb1, induces a T cell factor (TCF)-dependent proliferative response, alters hepatocyte metabolism via regulation of glutamine synthetase (encoded by Glul) and peroxisome proliferator-activated receptor-α (encoded by Ppara), and promotes tumoural T cell exclusion. HSC, hepatic stellate cell; NICD, Notch intracellular domain; RBPJ, immunoglobulin-κJ region.

An important caveat to these data is that most of the mouse studies described earlier were performed in toxin-induced, carcinogen-induced, or oncogene-induced tumours and might not necessarily be extrapolatable to NASH-induced HCC. Although mouse models of NASH–HCC have been described, including prolonged exposure to high-fat, high-carbohydrate diets or combinations of diet and toxins with genetic alterations165, there is still no robust model of NASH-derived cirrhosis that precedes HCC nor a reliable and time-efficient NASH–HCC mouse model that strongly reproduces human disease progression and the complex features of HCC at histological, genomic and transcriptomic levels. New studies that combine genetic manipulations of these developmental pathways with mouse models of diet-induced NASH are necessary to determine their role in NASH-induced HCC.

Therapeutic possibilities

Dissecting the cell-autonomous and cell-non-autonomous effects will guide our understanding of the pathophysiology of liver diseases but might also enable the development of targeted therapies. Most developmental pathways depend on cell membrane ligand–receptor interactions, opening possibilities for therapeutic tractability. For example, hepatocyte Notch activity promotes HSC activation and fibrosis, though it is unclear by which ligand, borne by which cell or by which mechanism the ligand is regulated in NASH. Dissecting this regulation and blocking specific Notch ligand–receptor interactions might enable the inhibition of pathological Notch reactivation in hepatocytes, while leaving intact homeostatic Notch functions in other cell types such as liver sinusoidal endothelial cells.

Alternative therapeutic modalities might also be effective. Proof-of-principle studies with small-molecule γ-secretase inhibitors efficiently blocked Notch-dependent increases in hepatic glucose production137,166 and liver fibrosis18 in mouse models, although regulatory approval is unlikely owing to substantial intestinal adverse effects167. However, Notch-induced goblet cell metaplasia can be avoided by using liver-directed antisense oligonucleotides to the γ-secretase-targeting subunit nicastrin168. Mice treated with nicastrin antisense oligonucleotides show ameliorated fibrosis with established NASH without intestinal toxicity18. Similar benefits were seen with γ-secretase inhibitor-encapsulated liver-directed nanoparticles169. Other therapeutic approaches, such as hepatocyte-specific N-acetylgalactosamine (GalNAc)-modified siRNA170, might similarly maintain therapeutic efficacy with minimal off-target adverse effects. In fact, GalNAc-siRNA targeting hepatocyte TAZ reversed NASH and fibrosis in a mouse model of NASH171. Additionally, lipid nanoparticles containing siRNA against Ctnnb1 specifically silenced hepatic β-catenin172 and improved porphyria-induced liver injury173 and HCC174 in various mouse models.

Combination therapy with dual-pathway inhibitors might provide additional benefit. As alluded to earlier, Notch and YAP–TAZ share overlapping functions in cell fate specification and might form a feedforward activation loop in tumorigenesis. Similarly, Notch and TAZ activation in hepatocytes activate HSCs via different secreted factors — OPN and IHH, respectively. Thus, it might be beneficial to simultaneously inhibit two pathways to achieve maximal treatment efficacy for NASH and fibrosis.

Conclusions

Developmental pathways are crucial to the expansion of progenitors and lineage specification in the liver. In embryogenesis, these pathways are coordinated in a spatiotemporal and cell type-specific manner to ensure the precision and accuracy of development. In a healthy liver, they lie dormant or restrictedly active in a cell type-specific or liver zone-specific manner. These pathways are reawakened when responding to liver injury and assemble a coordinated wound-healing response by inducing proliferation and creating a regenerative niche. Upon cessation of the acute insult, developmental pathways return to their baseline quiescent state and the regenerative process tapers off. However, in NASH, lipotoxicity remains a persistent injury stimulus and this evolutionarily programmed regenerative response becomes maladaptive and ultimately a culprit in disease progression. Understanding the mechanisms of this response will refine our knowledge of NASH pathogenesis but might also lead to novel therapeutic opportunities for a disease that is highly prevalent and has high morbidity.

Key points.

  • Nonalcoholic steatohepatitis(NASH), defined as liver lipid accumulation accompanied by injury and inflammation, is a prevalent condition without approved pharmacotherapy, leading to huge unmet needs for an increasingly obese population.

  • Developmental pathways, including Notch, Hedgehog, Hippo–YAP–TAZ and WNT–β-catenin, are fundamental regulators of cell fate decisions and morphogenesis in liver development.

  • Developmental pathways are also activated to promote regenerative proliferation and/or cellular reprogramming in response to liver injury.

  • In the face of chronic liver disease such as NASH, persistent and maladaptive activation of developmental pathways leads to exacerbated NASH, fibrosis and liver cancer.

  • Inhibitors targeting developmental pathways can ameliorate NASH and fibrosis in preclinical models, uncovering novel therapeutic possibilities for patients with NASH.

Acknowledgements

The authors of this work were supported by the American Heart Association (17PRE33120000, to C.Z.) and the NIH (R01 DK103818 and R01 DK119767, to U.B.P.).

Footnotes

Competing interests

The authors declare no competing interests.

Peer review information

Nature Reviews Gastroenterology & Hepatology thanks U. Apte, X. Chen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

References

  • 1.Hossain P., Kawar B. & El Nahas M. Obesity and diabetes in the developing world — a growing challenge. N. Engl. J. Med. 356, 213–215 (2007). [DOI] [PubMed] [Google Scholar]
  • 2.Lazo M & Clark JM The epidemiology of nonalcoholic fatty liver disease: a global perspective. Semin. Liver Dis. 28, 339–350 (2008). [DOI] [PubMed] [Google Scholar]
  • 3.Araujo AR, Rosso N, Bedogni G, Tiribelli C & Bellentani S Global epidemiology of non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: what we need in the future. Liver Int. 38 (Suppl. 1), 47–51 (2018). [DOI] [PubMed] [Google Scholar]
  • 4.Loomba R & Sanyal AJ The global NAFLD epidemic. Nat. Rev. Gastroenterol. Hepatol. 10, 686–690 (2013). [DOI] [PubMed] [Google Scholar]
  • 5.Younossi Z et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 15, 11–20 (2018). [DOI] [PubMed] [Google Scholar]
  • 6.Caligiuri A, Gentilini A & Marra F Molecular pathogenesis of NASH. Int. J. Mol. Sci. 17, 1575 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Younossi ZM et al. Nonalcoholic steatohepatitis is the most rapidly increasing indication for liver transplantation in the United States. Clin. Gastroenterol. Hepatol. 10.1016/j.cgh.2020.05.064 (2020). [DOI] [PubMed] [Google Scholar]
  • 8.Friedman SL, Neuschwander-Tetri BA, Rinella M & Sanyal AJ Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 24, 908–922 (2018).This article comprehensively reviews the clinical features, risk factors, known pathogenic mechanisms, preclinical models and treatment possibilities of NAFLD.
  • 9.Schwabe RF, Tabas I & Pajvani UB Mechanisms of fibrosis development in nonalcoholic steatohepatitis. Gastroenterology 158, 1913–1928 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kim D, Kim WR, Kim HJ & Therneau TM Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology 57, 1357–1365 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Dulai PS et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology 65, 1557–1565 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Angulo P et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 149, 389–397.e10 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ekstedt M et al. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 61, 1547–1554 (2015). [DOI] [PubMed] [Google Scholar]
  • 14.Vilar-Gomez E et al. Fibrosis severity as a determinant of cause-specific mortality in patients with advanced nonalcoholic fatty liver disease: a multi-national cohort study. Gastroenterology 155, 443–457.e17 (2018).Together with references 1113, this study reports that liver fibrosis is the major predictor of clinical outcomes in patients with NAFLD.
  • 15.Hannah WN Jr. Torres DM & Harrison SA Nonalcoholic steatohepatitis and endpoints in clinical trials. Gastroenterol. Hepatol. 12, 756–763 (2016). [PMC free article] [PubMed] [Google Scholar]
  • 16.Affo S, Yu LX & Schwabe RF The role of cancer-associated fibroblasts and fibrosis in liver cancer. Annu. Rev. Pathol. 12, 153–186 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Bray F et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68, 394–424 (2018). [DOI] [PubMed] [Google Scholar]
  • 18.Zhu C et al. Hepatocyte Notch activation induces liver fibrosis in nonalcoholic steatohepatitis. Sci. Transl. Med 10.1126/scitranslmed.aat0344 (2018).This paper demonstrates that aberrant Notch activity specifically in hepatocytes promotes NASH-associated liver fibrosis in a paracrine fashion.
  • 19.Verdelho Machado M & Diehl AM Role of hedgehog signaling pathway in NASH. Int. J. Mol. Sci. 17, 857 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Yimlamai D., Fowl BH. & Camargo FD. Emerging evidence on the role of the Hippo/YAP pathway in liver physiology and cancer. J. Hepatol. 63, 1491–1501 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wang X et al. Hepatocyte TAZ/WWTR1 promotes inflammation and fibrosis in nonalcoholic steatohepatitis. Cell Metab. 24, 848–862 (2016).This study shows that hepatocyte TAZ is stabilized in NASH and causes liver inflammation and fibrosis by stimulating Hedgehog ligand secretion.
  • 22.Mooring M et al. Hepatocyte stress increases expression of YAP and TAZ in hepatocytes to promote parenchymal inflammation and fibrosis. Hepatology 71, 1813–1830 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zong Y & Stanger BZ Molecular mechanisms of liver and bile duct development. Wiley Interdiscip. Rev. Dev. Biol. 1, 643–655 (2012). [DOI] [PubMed] [Google Scholar]
  • 24.Chillakuri CR, Sheppard D, Lea SM & Handford PA Notch receptor-ligand binding and activation: insights from molecular studies. Semin. Cell Dev. Biol. 23, 421–428 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bray SJ Notch signalling: a simple pathway becomes complex. Nat. Rev. Mol. Cell Biol. 7, 678–689 (2006). [DOI] [PubMed] [Google Scholar]
  • 26.Kageyama R, Ohtsuka T & Kobayashi T The Hes gene family: repressors and oscillators that orchestrate embryogenesis. Development 134, 1243–1251 (2007). [DOI] [PubMed] [Google Scholar]
  • 27.Turnpenny PD & Ellard S Alagille syndrome: pathogenesis, diagnosis and management. Eur. J. Hum. Genet. 20, 251–257 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Loomes KM et al. Characterization of Notch receptor expression in the developing mammalian heart and liver. Am. J. Med. Genet. 112, 181–189 (2002). [DOI] [PubMed] [Google Scholar]
  • 29.Hofmann JJ et al. Jagged1 in the portal vein mesenchyme regulates intrahepatic bile duct development: insights into Alagille syndrome. Development 137, 4061–4072 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Geisler F et al. Liver-specific inactivation of Notch2, but not Notch1, compromises intrahepatic bile duct development in mice. Hepatology 48, 607–616 (2008). [DOI] [PubMed] [Google Scholar]
  • 31.Lozier J, McCright B & Gridley T Notch signaling regulates bile duct morphogenesis in mice. PLoS ONE 3, e1851 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zong Y et al. Notch signaling controls liver development by regulating biliary differentiation. Development 136, 1727–1739 (2009).This study shows that Notch controls multiple steps of bile duct development, including the determination of biliary fate and the formation of ductal structures.
  • 33.Antoniou A et al. Intrahepatic bile ducts develop according to a new mode of tubulogenesis regulated by the transcription factor SOX9. Gastroenterology 136, 2325–2333 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Poncy A et al. Transcription factors SOX4 and SOX9 cooperatively control development of bile ducts. Dev. Biol. 404, 136–148 (2015). [DOI] [PubMed] [Google Scholar]
  • 35.Sparks EE, Huppert KA, Brown MA, Washington MK & Huppert SS Notch signaling regulates formation of the three-dimensional architecture of intrahepatic bile ducts in mice. Hepatology 51, 1391–1400 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tanimizu N & Miyajima A Notch signaling controls hepatoblast differentiation by altering the expression of liver-enriched transcription factors. J. Cell Sci. 117 (Pt. 15), 3165–3174 (2004). [DOI] [PubMed] [Google Scholar]
  • 37.Dong J et al. Elucidation of a universal size-control mechanism in Drosophila and mammals. Cell 130, 1120–1133 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhao B, Tumaneng K & Guan KL The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat. Cell Biol. 13, 877–883 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hansen CG, Moroishi T & Guan KL YAP and TAZ: a nexus for Hippo signaling and beyond. Trends Cell Biol. 25, 499–513 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhang N et al. The Merlin/NF2 tumor suppressor functions through the YAP oncoprotein to regulate tissue homeostasis in mammals. Dev. Cell 19, 27–38 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lee DH et al. LATS-YAP/TAZ controls lineage specification by regulating TGFβ signaling and Hnf4alpha expression during liver development. Nat. Commun. 7, 11961 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Alder O et al. Hippo signaling influences HNF4A and FOXA2 enhancer switching during hepatocyte differentiation. Cell Rep. 9, 261–271 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yimlamai D et al. Hippo pathway activity influences liver cell fate. Cell 157, 1324–1338 (2014).This study demonstrates that YAP activation can reprogramme mature hepatocytes to adopt progenitor characteristics.
  • 44.Fitamant J et al. YAP inhibition restores hepatocyte differentiation in advanced HCC, leading to tumor regression. Cell Rep. 10, 1692–1707 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Niewiadomski P et al. Gli proteins: regulation in development and cancer. Cells 8, 147 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Deutsch G, Jung J, Zheng M, Lora J & Zaret KS A bipotential precursor population for pancreas and liver within the embryonic endoderm. Development 128, 871–881 (2001). [DOI] [PubMed] [Google Scholar]
  • 47.Hirose Y, Itoh T & Miyajima A Hedgehog signal activation coordinates proliferation and differentiation of fetal liver progenitor cells. Exp. Cell Res. 315, 2648–2657 (2009). [DOI] [PubMed] [Google Scholar]
  • 48.Logan CY & Nusse R The Wnt signaling pathway in development and disease. Annu. Rev. Cell Dev. Biol. 20, 781–810 (2004). [DOI] [PubMed] [Google Scholar]
  • 49.Tan X et al. Beta-catenin deletion in hepatoblasts disrupts hepatic morphogenesis and survival during mouse development. Hepatology 47, 1667–1679 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tan X, Behari J, Cieply B, Michalopoulos GK & Monga SP Conditional deletion of beta-catenin reveals its role in liver growth and regeneration. Gastroenterology 131, 1561–1572 (2006). [DOI] [PubMed] [Google Scholar]
  • 51.Gougelet A et al. T-cell factor 4 and β-catenin chromatin occupancies pattern zonal liver metabolism in mice. Hepatology 59, 2344–2357 (2014). [DOI] [PubMed] [Google Scholar]
  • 52.Benhamouche S et al. Apc tumor suppressor gene is the “zonation-keeper” of mouse liver. Dev. Cell 10, 759–770 (2006). [DOI] [PubMed] [Google Scholar]
  • 53.Yang J et al. β-catenin signaling in murine liver zonation and regeneration: a Wnt-Wnt situation! Hepatology 60, 964–976 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Planas-Paz L et al. The RSPO-LGR4/5-ZNRF3/RNF43 module controls liver zonation and size. Nat. Cell Biol. 18, 467–479 (2016). [DOI] [PubMed] [Google Scholar]
  • 55.Sekine S, Lan BY, Bedolli M, Feng S & Hebrok M Liver-specific loss of beta-catenin blocks glutamine synthesis pathway activity and cytochrome p450 expression in mice. Hepatology 43, 817–825 (2006). [DOI] [PubMed] [Google Scholar]
  • 56.Cordi S et al. Role of β-catenin in development of bile ducts. Differentiation 91, 42–49 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hayward P, Kalmar T & Martinez Arias A Wnt/Notch signalling and information processing during development. Development 135, 411–424 (2008). [DOI] [PubMed] [Google Scholar]
  • 58.So J et al. Wnt/β-catenin signaling controls intrahepatic biliary network formation in zebrafish by regulating notch activity. Hepatology 67, 2352–2366 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Clotman F et al. Control of liver cell fate decision by a gradient of TGF beta signaling modulated by Onecut transcription factors. Genes Dev. 19, 1849–1854 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wang W et al. TGFβ signaling controls intrahepatic bile duct development may through regulating the Jagged1-Notch-Sox9 signaling axis. J. Cell Physiol. 233, 5780–5791 (2018). [DOI] [PubMed] [Google Scholar]
  • 61.Miyaoka Y et al. Hypertrophy and unconventional cell division of hepatocytes underlie liver regeneration. Curr. Biol. 22, 1166–1175 (2012). [DOI] [PubMed] [Google Scholar]
  • 62.Yanger K et al. Adult hepatocytes are generated by self-duplication rather than stem cell differentiation. Cell Stem Cell 15, 340–349 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Schaub JR, Malato Y, Gormond C & Willenbring H Evidence against a stem cell origin of new hepatocytes in a common mouse model of chronic liver injury. Cell Rep. 8, 933–939 (2014).This study, with reference 62, reports that hepatocytes, rather than liver stem cells, are the sources of liver mass regeneration in mouse models.
  • 64.Chen F et al. Broad distribution of hepatocyte proliferation in liver homeostasis and regeneration. Cell Stem Cell 26, 27–33.e4 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Sun T et al. AXIN2+ pericentral hepatocytes have limited contributions to liver homeostasis and regeneration. Cell Stem Cell 26, 97–107.e6 (2020). [DOI] [PubMed] [Google Scholar]
  • 66.Wang B, Zhao L, Fish M, Logan CY & Nusse R Self-renewing diploid Axin2+ cells fuel homeostatic renewal of the liver. Nature 524, 180–185 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Font-Burgada J et al. Hybrid periportal hepatocytes regenerate the injured liver without giving rise to cancer. Cell 162, 766–779 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Lin S et al. Distributed hepatocytes expressing telomerase repopulate the liver in homeostasis and injury. Nature 556, 244–248 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Huch M et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature 494, 247–250 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Tarlow BD, Finegold MJ & Grompe M Clonal tracing of Sox9+ liver progenitors in mouse oval cell injury. Hepatology 60, 278–289 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rodrigo-Torres D et al. The biliary epithelium gives rise to liver progenitor cells. Hepatology 60, 1367–1377 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Jors S et al. Lineage fate of ductular reactions in liver injury and carcinogenesis. J. Clin. Invest. 125, 2445–2457 (2015).Together with references 70 and 71, this study shows that ductular reaction or the ‘oval cell response’ predominantly derives from cholangiocytes.
  • 73.Russell JO et al. Hepatocyte-specific beta-catenin deletion during severe liver injury provokes cholangiocytes to differentiate into hepatocytes. Hepatology 69, 742–759 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lu WY et al. Hepatic progenitor cells of biliary origin with liver repopulation capacity. Nat. Cell Biol. 17, 971–983 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Raven A et al. Cholangiocytes act as facultative liver stem cells during impaired hepatocyte regeneration. Nature 547, 350–354 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Monga SP, Pediaditakis P, Mule K, Stolz DB & Michalopoulos GK Changes in WNT/beta-catenin pathway during regulated growth in rat liver regeneration. Hepatology 33, 1098–1109 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Nelsen CJ, Rickheim DG, Timchenko NA, Stanley MW & Albrecht JH Transient expression of cyclin D1 is sufficient to promote hepatocyte replication and liver growth in vivo. Cancer Res. 61, 8564–8568 (2001). [PubMed] [Google Scholar]
  • 78.Ochoa B et al. Hedgehog signaling is critical for normal liver regeneration after partial hepatectomy in mice. Hepatology 51, 1712–1723 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Grijalva JL et al. Dynamic alterations in Hippo signaling pathway and YAP activation during liver regeneration. Am. J. Physiol. Gastrointest. Liver Physiol. 307, G196–G204 (2014). [DOI] [PubMed] [Google Scholar]
  • 80.Lu L, Finegold MJ & Johnson RL Hippo pathway coactivators Yap and Taz are required to coordinate mammalian liver regeneration. Exp. Mol. Med. 50, e423 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kim AR et al. TAZ stimulates liver regeneration through interleukin-6-induced hepatocyte proliferation and inhibition of cell death after liver injury. FASEB J. 33, 5914–5923 (2019). [DOI] [PubMed] [Google Scholar]
  • 82.Swiderska-Syn M et al. Hedgehog regulates yes-associated protein 1 in regenerating mouse liver. Hepatology 64, 232–244 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Langiewicz M et al. Hedgehog pathway mediates early acceleration of liver regeneration induced by a novel two-staged hepatectomy in mice. J. Hepatol. 66, 560–570 (2017). [DOI] [PubMed] [Google Scholar]
  • 84.Kohler C et al. Expression of Notch-1 and its ligand Jagged-1 in rat liver during liver regeneration. Hepatology 39, 1056–1065 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Wang L et al. Disruption of the transcription factor recombination signal-binding protein-Jkappa (RBP-J) leads to veno-occlusive disease and interfered liver regeneration in mice. Hepatology 49, 268–277 (2009). [DOI] [PubMed] [Google Scholar]
  • 86.Cuervo H et al. Endothelial notch signaling is essential to prevent hepatic vascular malformations in mice. Hepatology 64, 1302–1316 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Duan JL et al. Endothelial Notch activation reshapes the angiocrine of sinusoidal endothelia to aggravate liver fibrosis and blunt regeneration in mice. Hepatology 68, 677–690 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yanger K et al. Robust cellular reprogramming occurs spontaneously during liver regeneration. Genes. Dev. 27, 719–724 (2013).This study shows that Notch promotes transdifferentiation of mature hepatocytes into cholangiocytes in several mouse models of liver injury.
  • 89.Boulter L et al. Macrophage-derived Wnt opposes Notch signaling to specify hepatic progenitor cell fate in chronic liver disease. Nat. Med. 18, 572–579 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Morell CM et al. Notch signaling and progenitor/ductular reaction in steatohepatitis. PLoS ONE 12, e0187384 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Walter TJ, Vanderpool C, Cast AE & Huppert SS Intrahepatic bile duct regeneration in mice does not require Hnf6 or Notch signaling through Rbpj. Am. J. Pathol. 184, 1479–1488 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Schaub JR et al. De novo formation of the biliary system by TGFbeta-mediated hepatocyte transdifferentiation. Nature 557, 247–251 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Pepe-Moone BJ. et al. Single-cell analysis of the liver epithelium reveals dynamic heterogeneity and an essential role for YAP in homeostasis and regeneration. Cell Stem Cell 10.1016/j.stem.2019.04.004 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Planas-Paz L et al. YAP, but not RSPO-LGR4/5, signaling in biliary epithelial cells promotes a ductular reaction in response to liver injury. Cell Stem Cell 25, 39–53.e10 (2019). [DOI] [PubMed] [Google Scholar]
  • 95.Sato K et al. Ductular reaction in liver diseases: pathological mechanisms and translational significances. Hepatology 69, 420–430 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Tarlow BD et al. Bipotential adult liver progenitors are derived from chronically injured mature hepatocytes. Cell Stem Cell 15, 605–618 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Deng X et al. Chronic liver injury induces conversion of biliary epithelial cells into hepatocytes. Cell Stem Cell 23, 114–122.e3 (2018). [DOI] [PubMed] [Google Scholar]
  • 98.Limaye PB et al. Expression of specific hepatocyte and cholangiocyte transcription factors in human liver disease and embryonic development. Lab. Invest. 88, 865–872 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Grompe M et al. Pharmacological correction of neonatal lethal hepatic dysfunction in a murine model of hereditary tyrosinaemia type I. Nat. Genet. 10, 453–460 (1995). [DOI] [PubMed] [Google Scholar]
  • 100.Miyamura N et al. YAP determines the cell fate of injured mouse hepatocytes in vivo. Nat. Commun. 8, 16017 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wree A, Broderick L, Canbay A, Hoffman HM & Feldstein AE From NAFLD to NASH to cirrhosis - new insights into disease mechanisms. Nat. Rev. Gastroenterol. Hepatol. 10, 627–636 (2013). [DOI] [PubMed] [Google Scholar]
  • 102.Buzzetti E, Pinzani M & Tsochatzis EA The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 65, 1038–1048 (2016). [DOI] [PubMed] [Google Scholar]
  • 103.Cusi K Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: pathophysiology and clinical implications. Gastroenterology 142, 711–725.e6 (2012). [DOI] [PubMed] [Google Scholar]
  • 104.Neuschwander-Tetri BA Hepatic lipotoxicity and the pathogenesis of nonalcoholic steatohepatitis: the central role of nontriglyceride fatty acid metabolites. Hepatology 52, 774–788 (2010). [DOI] [PubMed] [Google Scholar]
  • 105.Hardy T, Oakley F, Anstee QM & Day CP Nonalcoholic fatty liver disease: pathogenesis and disease spectrum. Annu. Rev. Pathol. 11, 451–496 (2016). [DOI] [PubMed] [Google Scholar]
  • 106.Friedman SL Hepatic stellate cells: protean, multifunctional, and enigmatic cells of the liver. Physiol. Rev. 88, 125–172 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Cordero-Espinoza L & Huch M The balancing act of the liver: tissue regeneration versus fibrosis. J. Clin. Invest. 128, 85–96 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mederacke I et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat. Commun. 4, 2823 (2013).This study revealed HSCs as the predominant contributors of liver fibrosis in mouse models.
  • 109.Asgharpour A et al. A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J. Hepatol. 65, 579–588 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Clapper JR et al. Diet-induced mouse model of fatty liver disease and nonalcoholic steatohepatitis reflecting clinical disease progression and methods of assessment. Am. J. Physiol. Gastrointest. Liver Physiol. 305, G483–G495 (2013). [DOI] [PubMed] [Google Scholar]
  • 111.Wolf MJ et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 26, 549–564 (2014). [DOI] [PubMed] [Google Scholar]
  • 112.Nakagawa H et al. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell 26, 331–343 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tsuchida T et al. A simple dietand chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J. Hepatol. 69, 385–395 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Machado MV & Diehl AM Hedgehog signalling in liver pathophysiology. J. Hepatol. 68, 550–562 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Sicklick JK et al. Hedgehog signaling maintains resident hepatic progenitors throughout life. Am. J. Physiol. Gastrointest. Liver Physiol. 290, G859–G870 (2006). [DOI] [PubMed] [Google Scholar]
  • 116.Michelotti GA et al. Smoothened is a master regulator of adult liver repair. J. Clin. Invest. 123, 2380–2394 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Kwon H et al. Inhibition of hedgehog signaling ameliorates hepatic inflammation in mice with nonalcoholic fatty liver disease. Hepatology 63, 1155–1169 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Chung SI et al. Hepatic expression of Sonic Hedgehog induces liver fibrosis and promotes hepatocarcinogenesis in a transgenic mouse model. J. Hepatol. 64, 618–627 (2016). [DOI] [PubMed] [Google Scholar]
  • 119.Matz-Soja M et al. Hedgehog signaling is a potent regulator of liver lipid metabolism and reveals a GLI-code associated with steatosis. eLife 5, e13308 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Marbach-Breitruck E et al. Tick-Tock Hedgehog-Mutual crosstalk with liver circadian clock promotes liver steatosis. J. Hepatol. 70, 1192–1202 (2019). [DOI] [PubMed] [Google Scholar]
  • 121.Guy CD et al. Hedgehog pathway activation parallels histologic severity of injury and fibrosis in human nonalcoholic fatty liver disease. Hepatology 55, 1711–1721 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Jung Y et al. Signals from dying hepatocytes trigger growth of liver progenitors. Gut 59, 655–665 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Guy CD, Suzuki A, Abdelmalek MF, Burchette JL & Diehl AM Treatment response in the PIVENS trial is associated with decreased Hedgehog pathway activity. Hepatology 61, 98–107 (2015).This study, with reference 121, shows that Hedgehog pathway activation is associated with disease severity and treatment response in patients with NASH.
  • 124.Lee YA et al. Autophagy is a gatekeeper of hepatic differentiation and carcinogenesis by controlling the degradation of Yap. Nat. Commun. 9, 4962 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Manmadhan S & Ehmer U Hippo signaling in the liver — a long and ever-expanding story. Front. Cell Dev. Biol. 7, 33 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Mannaerts I et al. The Hippo pathway effector YAP controls mouse hepatic stellate cell activation. J. Hepatol. 63, 679–688 (2015). [DOI] [PubMed] [Google Scholar]
  • 127.Martin K et al. PAK proteins and YAP-1 signalling downstream of integrin beta-1 in myofibroblasts promote liver fibrosis. Nat. Commun. 7, 12502 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Du K et al. Hedgehog-YAP signaling pathway regulates glutaminolysis to control activation of hepatic stellate cells. Gastroenterology 154, 1465–1479.e13 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Machado MV et al. Accumulation of duct cells with activated YAP parallels fibrosis progression in non-alcoholic fatty liver disease. J. Hepatol. 63, 962–970 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Russell JO & Monga SP Wnt/β-catenin signaling in liver development, homeostasis, and pathobiology. Annu. Rev. Pathol. 13, 351–378 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Go GW et al. The combined hyperlipidemia caused by impaired Wnt-LRP6 signaling is reversed by Wnt3a rescue. Cell Metab. 19, 209–220 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Lehwald N et al. β-catenin regulates hepatic mitochondrial function and energy balance in mice. Gastroenterology 143, 754–764 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Liu H et al. Wnt signaling regulates hepatic metabolism. Sci. Signal. 4, ra6 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Kordes C, Sawitza I & Haussinger D Canonical Wnt signaling maintains the quiescent stage of hepatic stellate cells. Biochem. Biophys. Res. Commun. 367, 116–123 (2008). [DOI] [PubMed] [Google Scholar]
  • 135.Ge WS et al. β-catenin is overexpressed in hepatic fibrosis and blockage of Wnt/β-catenin signaling inhibits hepatic stellate cell activation. Mol. Med. Rep.9, 2145–2151 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Ni MM et al. Novel Insights on Notch signaling pathways in liver fibrosis. Eur. J. Pharmacol. 826, 66–74 (2018). [DOI] [PubMed] [Google Scholar]
  • 137.Pajvani UB et al. Inhibition of Notch signaling ameliorates insulin resistance in a FoxO1-dependent manner. Nat. Med. 17, 961–967 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Pajvani UB et al. Inhibition of Notch uncouples Akt activation from hepatic lipid accumulation by decreasing mTorc1 stability. Nat. Med. 19, 1055–1060 (2013).References 137 and 138 show that, in mature hepatocytes, Notch sits at the bifurcation of insulin signalling to regulate glucose and lipid metabolism.
  • 139.Kitamura T et al. A Foxo/Notch pathway controls myogenic differentiation and fiber type specification. J. Clin. Invest. 117, 2477–2485 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Valenti L et al. Hepatic notch signaling correlates with insulin resistance and nonalcoholic fatty liver disease. Diabetes 62, 4052–4062 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.He F et al. Myeloid-specific disruption of recombination signal binding protein Jkappa ameliorates hepatic fibrosis by attenuating inflammation through cylindromatosis in mice. Hepatology 61, 303–314 (2015). [DOI] [PubMed] [Google Scholar]
  • 142.Xu J et al. NOTCH reprograms mitochondrial metabolism for proinflammatory macrophage activation. J. Clin. Invest. 125, 1579–1590 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Chen Y et al. Inhibition of Notch signaling by a gamma-secretase inhibitor attenuates hepatic fibrosis in rats. PLoS ONE 7, e46512 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Chen YX, Weng ZH & Zhang SL Notch3 regulates the activation of hepatic stellate cells. World J. Gastroenterol. 18, 1397–1403 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Xie G et al. Cross-talk between Notch and Hedgehog regulates hepatic stellate cell fate in mice. Hepatology 58, 1801–1813 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Yang YM et al. Hyaluronan synthase 2-mediated hyaluronan production mediates Notch1 activation and liver fibrosis. Sci. Transl Med. 11, eaat9284 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ouchi R et al. Modeling steatohepatitis in humans with pluripotent stem cell-derived organoids. Cell Metab. 30, 374–384.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wang S et al. RNA binding proteins control transdifferentiation of hepatic stellate cells into myofibroblasts. Cell Physiol. Biochem. 48, 1215–1229 (2018). [DOI] [PubMed] [Google Scholar]
  • 149.Hyun J et al. Dysregulated activation of fetal liver programme in acute liver failure. Gut 68, 1076–1087 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Younossi Z et al. Nonalcoholic steatohepatitis is the fastest growing cause of hepatocellular carcinoma in liver transplant candidates. Clin. Gastroenterol. Hepatol. 17, 748–755.e3 (2019). [DOI] [PubMed] [Google Scholar]
  • 151.Younossi ZM et al. Association of nonalcoholic fatty liver disease (NAFLD) with hepatocellular carcinoma (HCC) in the United States from 2004 to 2009. Hepatology 62, 1723–1730 (2015). [DOI] [PubMed] [Google Scholar]
  • 152.Anstee QM, Reeves HL, Kotsiliti E, Govaere O & Heikenwalder M From NASH to HCC: current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 16, 411–428 (2019).This review systemically discusses the epidemiology, pathogenesis and clinical management and diagnosis of NASH-induced HCC.
  • 153.Zender S et al. A critical role for notch signaling in the formation of cholangiocellular carcinomas. Cancer Cell 23, 784–795 (2013). [DOI] [PubMed] [Google Scholar]
  • 154.Villanueva A et al. Notch signaling is activated in human hepatocellular carcinoma and induces tumor formation in mice. Gastroenterology 143, 1660–1669.e7 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Cox AG et al. Yap reprograms glutamine metabolism to increase nucleotide biosynthesis and enable liver growth. Nat. Cell Biol. 18, 886–896 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Yuan WC et al. NUAK2 is a critical YAP target in liver cancer. Nat. Commun. 9, 4834 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Kim W et al. Hepatic Hippo signaling inhibits protumoural microenvironment to suppress hepatocellular carcinoma. Gut 67, 1692–1703 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Hagenbeek TJ et al. The Hippo pathway effector TAZ induces TEAD-dependent liver inflammation and tumors. Sci. Signal. 11, eaaj1757 (2018). [DOI] [PubMed] [Google Scholar]
  • 159.Senni N et al. β-catenin-activated hepatocellular carcinomas are addicted to fatty acids. Gut 68, 322–334 (2019). [DOI] [PubMed] [Google Scholar]
  • 160.Adebayo Michael AO et al. Inhibiting glutamine-dependent mTORC1 activation ameliorates liver cancers driven by β-catenin mutations. Cell Metab. 29, 1135–1150.e6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Ruiz de Galarreta M et al. β-catenin activation promotes immune escape and resistance to anti-PD-1 therapy in hepatocellular carcinoma. Cancer Discov. 9, 1124–1141 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Sia D et al. Identification of an immune-specific class of hepatocellular carcinoma, based on molecular features. Gastroenterology 153, 812–826 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Harding JJ et al. Prospective genotyping of hepatocellular carcinoma: clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin. Cancer Res. 25, 2116–2126 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Kim W et al. Hippo signaling interactions with Wnt/beta-catenin and Notch signaling repress liver tumorigenesis. J. Clin. Invest. 127, 137–152 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Febbraio MA et al. Preclinical models for studying NASH-driven HCC: how useful are they? Cell Metab. 29, 18–26 (2019).This review comprehensively summarizes the current knowledge of NASH-driven HCC and existing mouse models to study this disease.
  • 166.Sparling DP et al. Adipocyte-specific blockade of gamma-secretase, but not inhibition of Notch activity, reduces adipose insulin sensitivity. Mol. Metab. 5, 113–121 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.van Es JH. et al. Notch/gamma-secretase inhibition turns proliferative cells in intestinal crypts and adenomas into goblet cells. Nature 435, 959–963 (2005). [DOI] [PubMed] [Google Scholar]
  • 168.Kim K et al. γ-secretase inhibition lowers plasma triglyceride-rich lipoproteins by stabilizing the LDL receptor. Cell Metab. 27, 816–827.e4 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Richter LR et al. Targeted delivery of notch inhibitor attenuates obesity-induced glucose intolerance and liver fibrosis. ACS Nano 14, 6878–6886 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Wittrup A & Lieberman J Knocking down disease: a progress report on siRNA therapeutics. Nat. Rev. Genet. 16, 543–552 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Wang X et al. A therapeutic silencing RNA targeting hepatocyte TAZ prevents and reverses fibrosis in nonalcoholic steatohepatitis in mice. Hepatol. Commun. 3, 1221–1234 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ganesh S et al. Direct pharmacological inhibition of beta-catenin by RNA interference in tumors of diverse origin. Mol. Cancer Ther. 15, 2143–2154 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Saggi H et al. Loss of hepatocyte β-catenin protects mice from experimental porphyria-associated liver injury. J. Hepatol. 70, 108–117 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Tao J et al. Targeting β-catenin in hepatocellular cancers induced by coexpression of mutant β-catenin and K-Ras in mice. Hepatology 65, 1581–1599 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES