Elsevier

Differentiation

Volume 92, Issue 3, September 2016, Pages 84-92
Differentiation

Review article
The characteristics of activated portal fibroblasts/myofibroblasts in liver fibrosis

https://doi.org/10.1016/j.diff.2016.07.001Get rights and content

Abstract

Liver fibrosis results from chronic injury of hepatocytes and activation of Collagen Type I producing myofibroblasts that produce fibrous scar in liver fibrosis. Myofibroblasts are not present in the normal liver but rapidly appear early in experimental and clinical liver injury. The origin of the myofibroblast in liver fibrosis is still unresolved. The possibilities include activation of liver resident cells including portal fibroblasts, hepatic stellate cells, mesenchymal progenitor cells, and fibrocytes recruited from the bone marrow. It is considered that hepatic stellate cells and portal fibroblasts are the major source of hepatic myofibroblasts. In fact, the origin of myofibroblasts differs significantly for chronic liver diseases of different etiologies, such as cholestatic liver disease or hepatotoxic liver disease. Depending on etiology of hepatic injury, the fibrogenic foci might initiate within the hepatic lobule as seen in chronic hepatitis, or primarily affect the portal areas as in most biliary diseases. It has been suggested that activated portal fibroblasts/myofibroblasts work as “myofibroblasts for cholangiocytes” while hepatic stellate cells work as “myofibroblast for hepatocytes”. This review will focus on our current understanding of the activated portal fibroblasts/myofibroblasts in cholestatic liver fibrosis.

Introduction

Liver fibrosis and cirrhosis are the common outcomes of chronic liver diseases. Liver cirrhosis is characterized by the deposition of extracellular matrix proteins, composed mostly of Collagen Type I, formation of fibrous scar, and loss of liver function. There is no curative therapy for advanced liver cirrhosis, often liver transplantation is the only treatment available for these patients. Dependent on the etiology, liver fibrosis is caused by cholestatic liver injury (obstruction of biliary tract) such as primary sclerosing cholangitis, primary biliary cholangitis, secondary biliary cirrhosis and biliary atresia, or hepatotoxic injury (such as hepatitis B virus infection, hepatitis C virus infection, alcoholic liver disease and non-alcoholic steatohepatitis (NASH)). Despite the differences in etiology, development of liver fibrosis is associated with several early events that play an important role in the pathogenesis of liver fibrosis, including: 1) damage to hepatic epithelial (hepatocytes and cholangiocytes) and endothelial cells; 2) release of transforming growth factor-β1 (TGF-β1), the major fibrogenic cytokine; 3) increase of intestinal permeability and endogenous bacterial products; 4) recruitment of inflammatory cells; 5) induction of reactive oxygen species; and 6) generation of extracellular matrix producing myofibroblasts, which are not present in the normal liver. Hence, myofibroblasts represent a primary target for antifibrotic therapy.

Immunophenotypically, myofibroblasts are characterized by expression of abundant pericellular matrix proteins (vimentin, α-smooth muscle actin (α-SMA), non-muscle myosin, fibronectin) (Eyden, 2008). Ultrastructurally, myofibroblasts are identified by a rough endoplasmic reticulum, a Golgi apparatus producing collagen, peripheral myofilaments, fibronexus (no lamina), and gap junctions (Eyden, 2008). Studies of fibrogenesis conducted in different organs implicated myofibroblasts in wound healing and fibroproliferative disorders (Gabbiani et al., 1971, Majno et al., 1971, Schurch et al., 1998), suggesting that myofibroblasts are the primary source of extracellular matrix. Several sources of myofibroblasts have been identified in the liver: liver resident cells (Hepatic stellate cells, HSCs, and portal fibroblasts, PFs); cells originated by epithelial-to-mesenchymal transition (EMT) and bone marrow-derived cells (fibrocytes and mesenchymal cells) (Iwaisako et al., 2014, Kisseleva et al., 2006, Nitta et al., 2008, Scholten et al., 2011). Fibrocytes were shown to contribute to 3–6% of collagen Type I expressing cells in fibrotic liver, suggesting that fibrocytes are not a significant source of extracellular matrix (Kisseleva et al., 2006). Furthermore, the contribution of EMT to liver fibrosis is still controversial. Recently, Lua et al. reported that proliferating cholangiocytes in response to bile duct ligation express collagen, which means EMT of cholangiocytes (Lua et al., 2016). But several cell fate mapping of hepatic epithelial progenitors, hepatocytes, and cholangiocytes failed to detect the presence of EMT-derived myofibroblasts in the livers following cholestatic or hepatotoxic liver injury (Chu et al., 2011, Scholten et al., 2010, Taura et al., 2010).

Despite the extensive studies, there is still an ongoing discussion regarding which cell types can give rise to the hepatic myofibroblasts in response to chronic liver injury. Still “the primary suspects”, as the major source, are the resident mesenchymal cells of the liver (Iwaisako et al., 2014, Mederacke et al., 2013, Wells, 2014); Hepatic Stellate Cells (HSCs) which have been extensively studied and aPFs/myofibroblasts which are less well characterized due to the difficulties in isolation and culturing. HSCs and aPFs/myofibroblasts have been reported to comprise >90% of the collagen expressing cells (Iwaisako et al., 2014, Kisseleva et al., 2006), suggesting that they are the major source of collagen expressing cells in fibrotic liver. HSCs are generally accepted as major contributors to liver fibrosis that give rise to hepatic myofibroblasts in response to toxic liver injury. aPFs/myofibroblasts, on the other hand, have been implicated in pathogenesis of cholestatic liver fibrosis (Iwaisako et al., 2014). While experimental data validates that both HSCs and PFs can activate into myofibroblasts, the contribution of aPFs /myofibroblasts versus aHSCs to cholestatic liver fibrosis remains controversial and requires thorough examination, including lineage tracing experiments, identification and characterization of cell specific markers, and generation of new transgenic mice to study the functional properties of identified markers.

In the normal non-fibrotic liver, hepatic myofibroblasts become locally and transiently activated in response to bacterial infection. During wound healing, hepatic myofibroblasts apoptose upon completion of repair process (Iredale et al., 1998, Kisseleva et al., 2012). Therefore, pathogenic hepatic fibrosis could be viewed as a chronic state of hepatic myofibroblast activation and finding ways to terminate that activation or induce apoptosis in those aberrantly activated myofibroblasts may be the key to arresting hepatic fibrosis.

Recent studies provide potential experimental models of fibrosis reversal upon cessation of liver injury, or successful pharmacological treatment of underlying causative liver injury (Iredale et al., 1998). Experimental reversal of liver fibrosis has been closely associated with disappearance of hepatic myofibroblasts (Iredale, 2001, Iredale et al., 1998). The mechanism of hepatic myofibroblast disappearance during regression of liver fibrosis in these experiments has been suggested for aHSC-derived myofibroblasts, but remains unknown for myofibroblasts originated from aPFs /myofibroblasts. Thus, 50% aHSC-derived myofibroblasts undergo senescence (Schrader et al., 2009) and concomitant apoptosis (Iredale, 2001, Iredale, 2007, Iredale et al., 1998) during regression of liver fibrosis. The cell-fate mapping-based studies have demonstrated that 50% of aHSCs survive during regression of liver fibrosis, and obtain “inactivated” phenotype (iHSCs). iHSCs downregulate myofibroblast-specific genes, such as Collagen Type I, α-SMA. Spp1, TIMP1, and others, and upregulate some of genes associated with quiescent phenotype in qHSCs, therefore reverting to a quiescent-like state (Kisseleva and Brenner, 2008, Troeger et al., 2012). Clearly, aHSC-derived myofibroblasts are attractive primary targets for anti-fibrotic therapy (Friedman and Bansal, 2006, Ghiassi-Nejad et al., 2013). Like aHSCs, aPFs /myofibroblasts can give rise to activated myofibroblasts that drive hepatic fibrosis and therefore may offer another anti-fibrotic therapy target particularly in the case of cholestatic injury. Little is known about pathways of PF activation due to difficulties of isolating and characterizing aPFs /myofibroblasts. Also, it remains unknown if aPFs /myofibroblasts can “inactivate” during regression of cholestatic fibrosis. This review will summarize the most recent evidence supporting (or objecting) the contribution of aPFs /myofibroblasts to cholestatic liver fibrosis, and discuss their potential functional properties.

Section snippets

Hepatic Stellate Cells (HSCs)

In the normal liver, HSCs that reside in the space of Disse which is the space between hepatocyte cluster and sinusoidal endothelial cells, account for about 5–8% of cells (Lepreux and Desmouliere, 2015). HSCs in the healthy liver display a quiescent phenotype, and express neural markers (glial fibrillar acidic protein (GFAP), synaptophysin(Bataller and Brenner, 2005), nerve growth factor (NGF) receptor p75 (Kendall et al., 2009, Sachs et al., 2007)) and desmin. qHSCs also serve as the major

Experimental methods of cholestatic liver injury

The most commonly used mouse model for cholestatic fibrosis is bile duct ligation (BDL), a surgical procedure involving the double ligation of the common bile duct (Symeonidis and Trams, 1957). The pathology resulting from BDL is similar to that seen in human chronic cholestatic disease: initiation of a ductular reaction and activation of myofibroblasts surrounding biliary tracts. Development of advanced cholestatic fibrosis is usually observed in post-BDL mice at day 21. However, complications

Conclusions

There is a growing body of evidence that aPFs/myofibroblasts play a critical role in the pathogenesis of cholestatic (versus toxic) liver fibrosis. However, functional properties of aPFs/myofibroblasts and the mechanism by which aPFs/myofibroblasts contribute to cholestatic fibrosis, e.g. ductular reaction, angiogenesis, and extracellular matrix deposition are not well understood. Therefore, generation of new tools, such as aPFs/myofibroblasts specific transgenic and knockout mice, generation

Conflict of interests

None to declare.

Grant support

Supported by the National Institutes of Health (DK088837, GM41804, AA15055, and DK72237).

References (125)

  • S. Kivirikko et al.

    Primary structure of the alpha 1 chain of human type XV collagen and exon-intron organization in the 3′ region of the corresponding gene

    J. Biol. Chem.

    (1994)
  • T. Knittel et al.

    Rat liver myofibroblasts and hepatic stellate cells: different cell populations of the fibroblast lineage with fibrogenic potential

    Gastroenterology

    (1999)
  • T. Knittel et al.

    Expression patterns of matrix metalloproteinases and their inhibitors in parenchymal and non-parenchymal cells of rat liver: regulation by TNF-alpha and TGF-beta1

    J. Hepatol.

    (1999)
  • K. Koli et al.

    Bone morphogenetic protein-4 inhibitor gremlin is overexpressed in idiopathic pulmonary fibrosis

    Am. J. Pathol.

    (2006)
  • B. Ma et al.

    TGF-beta2 induces transdifferentiation and fibrosis in human lens epithelial cells via regulating gremlin and CTGF

    Biochem. Biophys. Res. Commun.

    (2014)
  • K.A. Mueller et al.

    Gremlin-1 identifies fibrosis and predicts adverse outcome in patients with heart failure undergoing endomyocardial biopsy

    J. Card. Fail.

    (2013)
  • Y. Muragaki et al.

    The human alpha 1(XV) collagen chain contains a large amino-terminal non-triple helical domain with a tandem repeat structure and homology to alpha 1(XVIII) collagen

    J. Biol. Chem.

    (1994)
  • G. Ramirez et al.

    Absence of Thy-1 results in TGF-beta induced MMP-9 expression and confers a profibrotic phenotype to human lung fibroblasts

    Lab. Invest.

    (2011)
  • D. Scholten et al.

    Genetic labeling does not detect epithelial-to-mesenchymal transition of cholangiocytes in liver fibrosis in mice

    Gastroenterology

    (2010)
  • D. Scholten et al.

    Migration of fibrocytes in fibrogenic liver injury

    Am. J. Pathol.

    (2011)
  • N. Sedlaczek et al.

    Proliferating bile duct epithelial cells are a major source of connective tissue growth factor in rat biliary fibrosis

    Am. J. Pathol.

    (2001)
  • S.H. Afroze et al.

    Activation of the renin-angiotensin system stimulates biliary hyperplasia during cholestasis induced by extrahepatic bile duct ligation

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2015)
  • M. Arias et al.

    Adenoviral delivery of an antisense RNA complementary to the 3′ coding sequence of transforming growth factor-beta1 inhibits fibrogenic activities of hepatic stellate cells

    Cell Growth Differ.

    (2002)
  • K. Asahina et al.

    Mesenchymal origin of hepatic stellate cells, submesothelial cells, and perivascular mesenchymal cells during mouse liver development

    Hepatology

    (2009)
  • K. Asahina et al.

    Septum transversum-derived mesothelium gives rise to hepatic stellate cells and perivascular mesenchymal cells in developing mouse liver

    Hepatology

    (2011)
  • A. Baghdasaryan et al.

    Curcumin improves sclerosing cholangitis in Mdr2−/− mice by inhibition of cholangiocyte inflammatory response and portal myofibroblast proliferation

    Gut

    (2010)
  • R. Bataller et al.

    Liver fibrosis

    J. Clin. Invest.

    (2005)
  • T.K. Bera et al.

    Mesothelin is not required for normal mouse development or reproduction

    Mol. Cell Biol.

    (2000)
  • N. Bosselut et al.

    Distinct proteomic features of two fibrogenic liver cell populations: hepatic stellate cells and portal myofibroblasts

    Proteomics

    (2010)
  • K. Chang et al.

    Molecular cloning of mesothelin, a differentiation antigen present on mesothelium, mesotheliomas, and ovarian cancers

    Proc. Natl. Acad. Sci. U S A

    (1996)
  • A.S. Chu et al.

    Lineage tracing demonstrates no evidence of cholangiocyte epithelial-to-mesenchymal transition in murine models of hepatic fibrosis

    Hepatology

    (2011)
  • R.H. Church et al.

    Gremlin1 preferentially binds to bone morphogenetic protein-2 (BMP-2) and BMP-4 over BMP-7

    Biochem J.

    (2015)
  • C. Colombo et al.

    Clinical features and genotype-phenotype correlations in children with progressive familial intrahepatic cholestasis type 3 related to ABCB4 mutations

    J. Pedia. Gastroenterol. Nutr.

    (2011)
  • J.M. de Vree et al.

    Mutations in the MDR3 gene cause progressive familial intrahepatic cholestasis

    Proc. Natl. Acad. Sci. U S A

    (1998)
  • D. Degiorgio et al.

    Molecular characterization and structural implications of 25 new ABCB4 mutations in progressive familial intrahepatic cholestasis type 3 (PFIC3)

    Eur. J. Hum. Genet.

    (2007)
  • J.L. Delaunay et al.

    A missense mutation in ABCB4 gene involved in progressive familial intrahepatic cholestasis type 3 leads to a folding defect that can be rescued by low temperature

    Hepatology

    (2009)
  • J.A. Dranoff et al.

    Ectonucleotidase NTPDase2 is selectively down-regulated in biliary cirrhosis

    J. Invest. Med.

    (2004)
  • J.A. Dranoff et al.

    Portal fibroblasts: Underappreciated mediators of biliary fibrosis

    Hepatology

    (2010)
  • A. Droguett et al.

    Tubular overexpression of gremlin induces renal damage susceptibility in mice

    PLoS One

    (2014)
  • A. Dudakovic et al.

    High-resolution molecular validation of self-renewal and spontaneous differentiation in clinical-grade adipose-tissue derived human mesenchymal stem cells

    J. Cell Biochem.

    (2014)
  • J. Dudas et al.

    Thy-1 is expressed in myofibroblasts but not found in hepatic stellate cells following liver injury

    Histochem. Cell Biol.

    (2009)
  • J. Dudas et al.

    Thy-1 is an in vivo and in vitro marker of liver myofibroblasts

    Cell Tissue Res.

    (2007)
  • V. Ellenrieder

    TGFbeta regulated gene expression by Smads and Sp1/KLF-like transcription factors in cancer

    Anticancer Res.

    (2008)
  • B. Eyden

    The myofibroblast: phenotypic characterization as a prerequisite to understanding its functions in translational medicine

    J. Cell Mol. Med.

    (2008)
  • J.A. Fallowfield et al.

    Scar-associated macrophages are a major source of hepatic matrix metalloproteinase-13 and facilitate the resolution of murine hepatic fibrosis

    J. Immunol.

    (2007)
  • M. Fausther et al.

    Establishment and characterization of rat portal myofibroblast cell lines

    PLoS One

    (2015)
  • M. Fausther et al.

    Activated hepatic stellate cells upregulate transcription of ecto-5′-nucleotidase/CD73 via specific SP1 and SMAD promoter elements

    Am. J. Physiol. Gastrointest. Liver Physiol.

    (2012)
  • S.L. Friedman et al.

    Reversal of hepatic fibrosis -- fact or fantasy?

    Hepatology

    (2006)
  • G. Gabbiani et al.

    Presence of modified fibroblasts in granulation tissue and their possible role in wound contraction

    Experientia

    (1971)
  • A. Geerts

    History, heterogeneity, developmental biology, and functions of quiescent hepatic stellate cells

    Semin. Liver Dis.

    (2001)
  • Cited by (90)

    • Cellular Interactions and Crosstalk Facilitating Biliary Fibrosis in Cholestasis

      2024, Cellular and Molecular Gastroenterology and Hepatology
    • The Origin and Fate of Liver Myofibroblasts

      2024, Cellular and Molecular Gastroenterology and Hepatology
    View all citing articles on Scopus
    1

    Authors equally contributed to the study.

    View full text