Elsevier

Cellular Signalling

Volume 23, Issue 4, April 2011, Pages 683-699
Cellular Signalling

Expression and functional analysis of endoglin in isolated liver cells and its involvement in fibrogenic Smad signalling

https://doi.org/10.1016/j.cellsig.2010.12.002Get rights and content

Abstract

Endoglin is an accessory component of the TGF-β-binding receptor complex that differentially modulates TGF-β and BMP responses. The existence of two splice variants L- and S-endoglin which differ in their cytoplasmic domain has already been shown in human and mice. Endoglin is located on the cell surfaces of cultured hepatic stellate cells and transdifferentiated myofibroblasts suggesting that this receptor might be associated with the profibrogenic attributes of these liver cell subpopulations. We now show that endoglin expression is increased in transdifferentiating hepatic stellate cells and in two models of liver fibrosis (i.e. bile duct ligation and carbon tetrachloride model) and further detectable in cultured portal fibroblasts representing another important fibrogenic cell type but not in hepatocytes. In respect to TGF-β1-signalling, we demonstrate that endoglin interacts with and is phosphorylated by TβRII. In hepatic stellate cells, TGF-β1 upregulates endoglin expression most likely via the ALK5 pathway and requires the SP1 transcription factor. We further identified a novel rat splice variant that is structurally and functionally different from that identified in human and mouse. Transient overexpression of endoglin resulted in a strong increase of TGF-β1-driven Smad1/5 phosphorylation and α-smooth muscle actin expression in a hepatic stellate cell line. In supernatants of respective cultures, we could detect the ectodomain of endoglin suggesting that shedding is a further key process involved in the regulation of this surface receptor.

Introduction

Endoglin (CD105) is a type III TGF-β co-receptor that is expressed as a disulfide-linked 180-kD homodimer in the plasma membrane of diverse cells. Of fundamental interest is the expression of endoglin in profibrogenic cells including mesangial cells, cardiac fibroblasts, scleroderma fibroblasts and hepatic stellate cells (HSC) [1], [2], [3], [4]. The glycosylated extracellular domain contains a putative protease cleavage site in human, mouse and rat [5], [6], which upon proteolysis by matrix metalloproteinase-14 (MT1-MMP) gives rise to a soluble, shedded form of endoglin [7]. This process is triggered under pathological conditions [8], [9], [10]. The single membrane spanning domain is connected to a short cytoplasmic domain (CD) that lacks catalytic activity [5], [6] but comprises several serine/threonine residues and a PDZ-domain [11] both of which regulate protein–protein interactions [12]. The retention of an intron in the CD results in a longer transcript, which is translated into a shorter protein, i.e. S-endoglin. This variant misses most of the CD domain including phosphorylation sites and protein–protein interaction domains [13]. Due to this modification, S-endoglin has a different role in cellular responses in comparison to the full-length receptor [14]. The occurrence of S-endoglin has been described in human and mouse but not in rat [13], [15].

The expression of endoglin is up-regulated under conditions of cellular activation, e.g. in endothelial cells (angiogenesis) and monocytes/macrophages [16], [17], and under hypoxic conditions [18]. In addition, angiotensin II and TGF-β1 are capable of inducing endoglin expression [2], [19]. Under basal conditions, endoglin expression is mainly influenced by the transcription factor specificity protein 1 (SP1) [20], whereas a network of SP1, KLF-6, Smad3 and Hif-1α is involved under hypoxic or TGF-β1-stimulated conditions [18], [21], [22]. The fact that TGF-β/Smad3 is involved in regulating endoglin promoter activity implies a regulatory feedback loop due to the ligand-binding spectrum of endoglin encompassing TGF-β1 and TGF-β3 in addition to the BMP-ligands BMP-2, BMP-7, and BMP-9 [23], [24], [25].

In contrast to betaglycan, binding of TGF-β1 to endoglin requires the presence of the corresponding type I and II receptors [23]. As a consequence, endoglin interacts with the TGF-β signalling receptor type II (TβRII) and the two type I receptors ALK5 and ALK1 [26], [27], [28]. Interaction with the signalling receptors involves specific N- and C-terminal domains of endoglin and results in the phosphorylation of its CD [27], [28], [29], [30]. TβRII phosphorylates distinct serine residues in endoglin [11], a modification which has been postulated to be lost upon internalization in endothelial cells [31]. Interestingly, ALK1 and ALK5 are known to act on endoglin-CD serine and threonine residues [11], [32] but since endoglin binds BMP-ligands (e.g. BMP-2 and BMP-9) it is also substrate for BMP-type I receptors (e.g. ALK2) [32], [33]. In turn, phosphorylation of the CD of endoglin influences the association with ALK1 and ALK5 and interaction with other signalling intermediates [11], [34], [35].

In the context of the afore-mentioned signalling pathways, the role of endoglin is extremely versatile involving both Smad-independent and -dependent mechanisms [34], [36], [37], [38]. In HSC, TGF-β1 activates two different Smad pathways involving Smad2/3 or Smad1/5. The Smad3 branch is clearly linked to fibrogenic signalling [39]. On the other hand it has been shown that activation and transcriptional activity of Smad1 increases during in vitro activation of HSC and in HSC isolated from carbon tetrachloride-intoxicated rats [40], [41]. The pivotal role of Smad1 in the process of cellular activation is implied by the finding that BMP-2 which is a genuine activator of Smad1/5 forces α-SMA expression in activated HSC [40]. Nevertheless, α-SMA expression is preserved on a Smad3 background [42], pointing on a vital role for Smad1/5, which are prominently activated by TGF-β1 [43]. In addition, the TGF-β1/ALK1/Smad1-mediated Id1 expression was shown to promote HSC activation via re-organization of α-SMA-fibres [43].

It has been shown that endoglin is up-regulated by and negatively regulates profibrogenic responses of TGF-β1 in several cell types [1], [2], [3], [44]. Since endoglin is highly expressed in HSC [4] representing the major profibrogenic cell type in the liver, we sought to get a more comprehensive view on the expression of endoglin in liver cells and to get a first glimpse on endoglin function and involvement in TGF-β1-signalling in culture-activated, transdifferentiating HSC.

We show here that endoglin is expressed in all liver cell types except hepatocytes and that endoglin is transiently up-regulated during transdifferentiation. Likewise we found an increased expression of endoglin in two animal models of liver fibrogenesis. In line, we could show an increased expression of endoglin mediated by TGF-β1 that involves SP1 and activated Smad3. Moreover, we identified a splice variant of L-endoglin which differs from the previously identified short form of mouse and rat. Both variants interact with and are phosphorylated by TβRII. In CFSC, which represents an immortalized activated HSC cell line that show a low endogenous endoglin expression, we could demonstrate that overexpression of endoglin results in an overall stronger TGF-β1-mediated activation of Smad1/Smad5 and increased expression of α-SMA. Finally we could demonstrate that endoglin is shedded from the surface of CFSC giving rise to a soluble receptor.

Section snippets

Animal treatment

To induce liver fibrosis in male Sprague–Dawley rats, the common bile duct was double ligated (BDL) or CCl4 was repeatedly administrated as described in detail elsewhere [45], [46].

Cell culture

HSC, Kupffer cells (KC) and liver sinusoidal endothelial cells (LSEC) were isolated from male Sprague–Dawley rats by the pronase–collagenase method followed by a density centrifugation on a Nycodenz gradient as described before [47], [48]. HSC were passaged once to obtain myofibroblast-like cells (MFB). Primary

Endoglin expression in isolated liver cells

HSC and MFB express the two TβRIII receptors betaglycan and endoglin (CD105), both promiscuously detected by the antibody sc-6199 directed against an epitope of the betaglycan C-terminus [4]. To analyze endoglin in the liver in more detail, we generated two independent antibodies, i.e. PPabE1 and PPabE2 that are directed against different epitopes within the extracellular domain of rat endoglin (Fig. 2A) and are not cross-reactive with other TGF-β-receptors (Fig. 2B) and are highly specific for

Discussion

In the present study we demonstrate that endoglin is expressed in all isolated liver cells except hepatocytes. Endoglin expression is transiently up-regulated during HSC transdifferentiation and as a mutual consequence in two models of liver fibrogenesis, i.e. BDL and CCl4 administration. The increase in receptor expression in HSC is at least partly regulated by TGF-β1 itself, since this ligand induces endoglin expression in a time- and concentration-dependent manner. This regulation involves

Conclusions

Our study shows that endoglin is expressed in all profibrogenic liver cells and that its expression and surface exposure is increased during activation and transdifferentiation of HSC. TGF-β1 upregulates endoglin expression most likely via the ALK5 pathway and requires the SP1 transcription factor. Endoglin interacts with TβRII and enhances Smad1/5 phosphorylation and strongly increases the expression of α-SMA representing a marker of HSC activation. We further identified a novel rat splice

Acknowledgements

The authors thank Andreas Ludwig (Institute of Pharmacology and Toxicology, RWTH University Hospital Aachen, Germany) for kindly providing HUVEC cells, M. Rojkind (Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, New York, USA) for CFSC-2G, Scott L. Friedman (Division of Liver Diseases, Mount Sinai School of Medicine, New York, NY 10029, USA) for LX-2 cells, M. Cristina Cardoso (Max Delbrück Center for Molecular Medicine, Berlin, Germany) for L6E9 cells, and

References (88)

  • L. Diez-Marques et al.

    Biochim. Biophys. Acta

    (2002)
  • S.K. Meurer et al.

    J. Biol. Chem.

    (2005)
  • A. Gougos et al.

    J. Biol. Chem.

    (1990)
  • O. Llorca et al.

    J. Mol. Biol.

    (2007)
  • R.I. Koleva et al.

    J. Biol. Chem.

    (2006)
  • B.A. Conley et al.

    J. Biol. Chem.

    (2004)
  • J.R. Westphal et al.

    J. Invest. Dermatol.

    (1993)
  • T. Sánchez-Elsner et al.

    J. Biol. Chem.

    (2002)
  • L.M. Botella et al.

    J. Biol. Chem.

    (2001)
  • L.M. Botella et al.

    Blood

    (2002)
  • N.P. Barbara et al.

    J. Biol. Chem.

    (1999)
  • A. Letamendía et al.

    J. Biol. Chem.

    (1998)
  • S. Cheifetz et al.

    J. Biol. Chem.

    (1992)
  • M. Guerrero-Esteo et al.

    J. Biol. Chem.

    (2002)
  • H. Yamashita et al.

    J. Biol. Chem.

    (1994)
  • H. Tang et al.

    Blood

    (2005)
  • N.Y. Lee et al.

    J. Biol. Chem.

    (2007)
  • N.Y. Lee et al.

    J. Biol. Chem.

    (2008)
  • O. Scherner et al.

    J. Biol. Chem.

    (2007)
  • B. Schnabl et al.

    Hepatology

    (2001)
  • E. Borkham-Kamphorst et al.

    Lab. Invest.

    (2004)
  • E. Borkham-Kamphorst et al.

    Lab. Invest.

    (2008)
  • H. Fehrenbach et al.

    Hepatology

    (2001)
  • P.O. Seglen

    Meth. Cell Biol.

    (1976)
  • S. Vogel et al.

    J. Lipid Res.

    (2000)
  • B. Nadal-Ginard

    Cell

    (1978)
  • H.Y. Lin et al.

    Cell

    (1992)
  • R. Weiskirchen et al.

    Biochem. Biophys. Res. Commun.

    (2000)
  • S.L. Friedman

    Gastroenterology

    (2008)
  • Y.Q. Wang et al.

    Biochem. Biophys. Res. Commun.

    (2004)
  • K. Miao et al.

    Arch. Biochem. Biophys.

    (1997)
  • S. Gaidarova et al.

    J. Biol. Chem.

    (2002)
  • C. Ríus et al.

    Blood

    (1998)
  • F. Sanz-Rodriguez et al.

    J. Biol. Chem.

    (2004)
  • Q. Meng et al.

    J. Biol. Chem.

    (2006)
  • S. Dooley et al.

    FEBS Lett.

    (2001)
  • K.M. Koli et al.

    J. Biol. Chem.

    (1997)
  • R.G. Wells et al.

    J. Biol. Chem.

    (1997)
  • R.G. Wells et al.

    J. Biol. Chem.

    (1999)
  • J. Pannu et al.

    J. Biol. Chem.

    (2007)
  • K.W. Finnson et al.

    Osteoarthritis Cartilage

    (2010)
  • K. Chen et al.

    Circ. Res.

    (2004)
  • A. Leask et al.

    Arthritis Rheum.

    (2002)
  • L.J. Hawinkels et al.

    Cancer Res.

    (2010)
  • Cited by (53)

    • Endoglin and soluble endoglin in liver sinusoidal endothelial dysfunction in vivo

      2024, Biochimica et Biophysica Acta - Molecular Basis of Disease
    • High soluble endoglin levels regulate cholesterol homeostasis and bile acids turnover in the liver of transgenic mice

      2019, Life Sciences
      Citation Excerpt :

      Soluble endoglin (sEng) is free form of membrane bound Endoglin (CD 105) that is a transmembrane glycoprotein acting as coreceptor of the transforming growth factor-β (TGF-β) superfamily, which is expressed in the plasma membrane of diverse cells (e.g in endothelial cells, macrophages, fibroblasts or hepatic stellate cells) [1,2].

    • Identification of bone morphogenetic protein 9 (BMP9) as a novel profibrotic factor in vitro

      2016, Cellular Signalling
      Citation Excerpt :

      This pathway plays an anti-fibrotic role in different experimental models of renal fibrosis [8,10,21,22] or in in vitro experiments [23,24]. However, in other biological contexts, Smad1 activation promotes ECM protein synthesis in liver cells [25,26], glomerular mesangial cells [27,28] or dermal fibroblasts [29–31]. BMP9 also inhibits angiogenesis in different conditions [18,19,32].

    • Overexpression of the short endoglin isoform reduces renal fibrosis and inflammation after unilateral ureteral obstruction

      2016, Biochimica et Biophysica Acta - Molecular Basis of Disease
      Citation Excerpt :

      Surprisingly, total Smad1 levels were also lower in O than in NO kidneys in both S-ENG+ and WT mice (Fig. 8b). Eng is expressed in many profibrogenic cells as mesangial cells and renal fibroblasts of the kidney, as well as in skin, intestinal and cardiac fibroblasts, and in cultured portal fibroblasts and hepatic stellate cells of the liver [14,31]. Eng upregulation has been observed in several experimental models of renal fibrosis [12,32].

    • PDGF-D signaling in portal myofibroblasts and hepatic stellate cells proves identical to PDGF-B via both PDGF receptor type α and β

      2015, Cellular Signalling
      Citation Excerpt :

      Thereafter, the medium was supplemented to 10% FCS. Upon culturing, pF differentiated into pMF that had spindle morphology and expressed fibulin-2 [15], α-SMA but were negative for desmin [16]. Primary HSC were isolated from male Sprague–Dawley rats through density gradient centrifugation technique and cultured as described [17,18].

    View all citing articles on Scopus
    View full text