Skip to main content
Top
Published in: Fibrogenesis & Tissue Repair 1/2015

Open Access 01-12-2015 | Research

In vitro reversion of activated primary human hepatic stellate cells

Authors: Adil El Taghdouini, Mustapha Najimi, Pau Sancho-Bru, Etienne Sokal, Leo A. van Grunsven

Published in: Fibrogenesis & Tissue Repair | Issue 1/2015

Login to get access

Abstract

Background

Liver fibrosis is characterized by the excessive formation and accumulation of matrix proteins as a result of wound healing in the liver. A main event during fibrogenesis is the activation of the liver resident quiescent hepatic stellate cell (qHSC). Recent studies suggest that reversion of the activated HSC (aHSC) phenotype into a quiescent-like phenotype could be a major cellular mechanism underlying fibrosis regression in the liver, thereby offering new therapeutic perspectives for the treatment of liver fibrosis. Whether human HSCs have the ability to undergo a similar reversion in phenotype is currently unknown. The aim of the present study is to identify experimental conditions that can revert the in vitro activated phenotype of primary human HSCs and consequently to map the molecular events associated with this reversion process by gene expression profiling.

Results

We find that epidermal growth factor (EGF) and fibroblast growth factor 2 (FGF2) synergistically downregulate the expression of ACTA2 and LOX in primary human aHSCs. Their combination with oleic acid, palmitic acid, and retinol further potentiates a more quiescent-like phenotype as demonstrated by the abundant presence of retinyl ester-positive intra-cytoplasmic lipid droplets, low expression levels of activation markers, and a reduced basal as well as cytokine-stimulated proliferation and matrix metalloproteinase activity. Gene expression profiling experiments reveal that these in vitro reverted primary human HSCs (rHSCs) display an intermediary phenotype that is distinct from qHSCs and aHSCs. Interestingly, this intermediary phenotype is characterized by the increased expression of several previously identified signature genes of in vivo inactivated mouse HSCs such as CXCL1, CXCL2, and CTSS, suggesting also a potential role for these genes in promoting a quiescent-like phenotype in human HSCs.

Conclusions

We provide evidence for the ability of human primary aHSCs to revert in vitro to a transitional state through synergistic action of EGF, FGF2, dietary fatty acids and retinol, and provide a first phenotypic and genomic characterization of human in vitro rHSCs.
Appendix
Available only for authorised users
Literature
7.
go back to reference Geerts ABL, Wisse E. History, heterogeneity, developmental biology, and functions of quiescent hepatic stellate cells. Semin Liver Dis. 2001;21:311–35.PubMedCrossRef Geerts ABL, Wisse E. History, heterogeneity, developmental biology, and functions of quiescent hepatic stellate cells. Semin Liver Dis. 2001;21:311–35.PubMedCrossRef
9.
go back to reference Iredale JPBRC, Pickering J, McCullen M, Northrop M, Pawley S, Hovell C, et al. Mechanisms of spontaneous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998;102:538–49.PubMedCentralPubMedCrossRef Iredale JPBRC, Pickering J, McCullen M, Northrop M, Pawley S, Hovell C, et al. Mechanisms of spontaneous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998;102:538–49.PubMedCentralPubMedCrossRef
11.
go back to reference Hazra S, Xiong S, Wang J, Rippe RA, Krishna V, Chatterjee K, et al. Peroxisome proliferator-activated receptor γ induces a phenotypic switch from activated to quiescent hepatic stellate cells. J Biol Chem. 2004;279(12):11392–401. doi:10.1074/jbc.M310284200.PubMedCrossRef Hazra S, Xiong S, Wang J, Rippe RA, Krishna V, Chatterjee K, et al. Peroxisome proliferator-activated receptor γ induces a phenotypic switch from activated to quiescent hepatic stellate cells. J Biol Chem. 2004;279(12):11392–401. doi:10.​1074/​jbc.​M310284200.PubMedCrossRef
15.
go back to reference Liu XXJ, Brenner DA, Kisseleva T. Reversibility of liver fibrosis and inactivation of fibrogenic myofibroblasts. Curr Pathobiol Rep. 2013;1(3):209–14.PubMedCentralPubMedCrossRef Liu XXJ, Brenner DA, Kisseleva T. Reversibility of liver fibrosis and inactivation of fibrogenic myofibroblasts. Curr Pathobiol Rep. 2013;1(3):209–14.PubMedCentralPubMedCrossRef
17.
go back to reference Niki T, Pekny M, Hellemans K, De Bleser P, Van Den Berg K, Vaeyens F, et al. Class VI intermediate filament protein nestin is induced during activation of rat hepatic stellate cells. Hepatology. 1999;29(2):520–7. doi:10.1002/hep.510290232.PubMedCrossRef Niki T, Pekny M, Hellemans K, De Bleser P, Van Den Berg K, Vaeyens F, et al. Class VI intermediate filament protein nestin is induced during activation of rat hepatic stellate cells. Hepatology. 1999;29(2):520–7. doi:10.​1002/​hep.​510290232.PubMedCrossRef
18.
go back to reference Niki T, De Bleser PJ, Xu G, Van der Berg K, Wisse E, Geerts A. Comparison of glial fibrillary acidic protein and desmin staining in normal and CCl4-induced fibrotic rat livers. Hepatology. 1996;23(6):1538–45. doi:10.1002/hep.510230634.PubMedCrossRef Niki T, De Bleser PJ, Xu G, Van der Berg K, Wisse E, Geerts A. Comparison of glial fibrillary acidic protein and desmin staining in normal and CCl4-induced fibrotic rat livers. Hepatology. 1996;23(6):1538–45. doi:10.​1002/​hep.​510230634.PubMedCrossRef
20.
go back to reference Hammel P, Couvelard A, O’Toole D, Ratouis A, Sauvanet A, Fléjou JF, et al. Regression of liver fibrosis after biliary drainage in patients with chronic pancreatitis and stenosis of the common bile duct. New Engl J Med. 2001;344(6):418–23. doi:10.1056/NEJM200102083440604.PubMedCrossRef Hammel P, Couvelard A, O’Toole D, Ratouis A, Sauvanet A, Fléjou JF, et al. Regression of liver fibrosis after biliary drainage in patients with chronic pancreatitis and stenosis of the common bile duct. New Engl J Med. 2001;344(6):418–23. doi:10.​1056/​NEJM200102083440​604.PubMedCrossRef
23.
24.
go back to reference Abdalla A, Zalata K, Ismail A, Shiha G, Attiya M, Abo-Alyazeed A. Regression of fibrosis in paediatric autoimmune hepatitis: morphometric assessment of fibrosis versus semiquantitative methods. Fibrogenesis Tissue Repair. 2009;2(1):2.PubMedCentralPubMedCrossRef Abdalla A, Zalata K, Ismail A, Shiha G, Attiya M, Abo-Alyazeed A. Regression of fibrosis in paediatric autoimmune hepatitis: morphometric assessment of fibrosis versus semiquantitative methods. Fibrogenesis Tissue Repair. 2009;2(1):2.PubMedCentralPubMedCrossRef
26.
go back to reference Olsen PS, Boesby S, Kirkegaard P, Therkelsen K, Almdal T, Poulsen SS, et al. Influence of epidermal growth factor on liver regeneration after partial hepatectomy in rats. Hepatology. 1988;8(5):992–6. doi:10.1002/hep.1840080503.CrossRef Olsen PS, Boesby S, Kirkegaard P, Therkelsen K, Almdal T, Poulsen SS, et al. Influence of epidermal growth factor on liver regeneration after partial hepatectomy in rats. Hepatology. 1988;8(5):992–6. doi:10.​1002/​hep.​1840080503.CrossRef
29.
go back to reference Rosenbaum J, Blazejewski S, Préaux A-M, Mallat A, Dhumeaux D, Mavier P. Fibroblast growth factor 2 and transforming growth factor β1 interactions in human liver myofibroblasts. Gastroenterology. 1995;109(6):1986–96. doi:10.1016/0016-5085(95)90767-x.PubMedCrossRef Rosenbaum J, Blazejewski S, Préaux A-M, Mallat A, Dhumeaux D, Mavier P. Fibroblast growth factor 2 and transforming growth factor β1 interactions in human liver myofibroblasts. Gastroenterology. 1995;109(6):1986–96. doi:10.​1016/​0016-5085(95)90767-x.PubMedCrossRef
30.
go back to reference Ciccolini F, Svendsen CN. Fibroblast growth factor 2 (FGF-2) promotes acquisition of epidermal growth factor (EGF) responsiveness in mouse striatal precursor cells: identification of neural precursors responding to both EGF and FGF-2. J Neurosci. 1998;18(19):7869–80.PubMed Ciccolini F, Svendsen CN. Fibroblast growth factor 2 (FGF-2) promotes acquisition of epidermal growth factor (EGF) responsiveness in mouse striatal precursor cells: identification of neural precursors responding to both EGF and FGF-2. J Neurosci. 1998;18(19):7869–80.PubMed
36.
go back to reference Botella LM, Sanz-rodriguez F, Komi Y, Fernandez-l A, Varela E, Garrido-martin EM, et al. TGF-β regulates the expression of transcription factor KLF6 and its splice variants and promotes co-operative transactivation of common target genes through a Smad3–Sp1–KLF6 interaction. Biochem J. 2009;419(2):485–95. doi:10.1042/bj20081434.PubMedCentralPubMedCrossRef Botella LM, Sanz-rodriguez F, Komi Y, Fernandez-l A, Varela E, Garrido-martin EM, et al. TGF-β regulates the expression of transcription factor KLF6 and its splice variants and promotes co-operative transactivation of common target genes through a Smad3–Sp1–KLF6 interaction. Biochem J. 2009;419(2):485–95. doi:10.​1042/​bj20081434.PubMedCentralPubMedCrossRef
37.
go back to reference Kojima S, Hayashi S, Shimokado K, Suzuki Y, Shimada J, Crippa MP, et al. Transcriptional activation of urokinase by the Krüppel-like factor Zf9/COPEB activates latent TGF-β1 in vascular endothelial cells. Blood. 2000;95(4):1309–16.PubMed Kojima S, Hayashi S, Shimokado K, Suzuki Y, Shimada J, Crippa MP, et al. Transcriptional activation of urokinase by the Krüppel-like factor Zf9/COPEB activates latent TGF-β1 in vascular endothelial cells. Blood. 2000;95(4):1309–16.PubMed
38.
go back to reference Zhang W, Ou J, Inagaki Y, Greenwel P, Ramirez F. Synergistic cooperation between Sp1 and Smad3/Smad4 mediates transforming growth factor β1 stimulation of α2(I)-collagen (COL1A2) transcription. J Biol Chem. 2000;275(50):39237–45. doi:10.1074/jbc.M003339200.PubMedCrossRef Zhang W, Ou J, Inagaki Y, Greenwel P, Ramirez F. Synergistic cooperation between Sp1 and Smad3/Smad4 mediates transforming growth factor β1 stimulation of α2(I)-collagen (COL1A2) transcription. J Biol Chem. 2000;275(50):39237–45. doi:10.​1074/​jbc.​M003339200.PubMedCrossRef
40.
go back to reference Park J-H, Jo J-H, Kim K-H, Kim S-J, Lee W-R, Park K-K, et al. Antifibrotic effect through the regulation of transcription factor using ring type-Sp1 decoy oligodeoxynucleotide in carbon tetrachloride-induced liver fibrosis. J Gene Med. 2009;11(9):824–33. doi:10.1002/jgm.1355.PubMedCrossRef Park J-H, Jo J-H, Kim K-H, Kim S-J, Lee W-R, Park K-K, et al. Antifibrotic effect through the regulation of transcription factor using ring type-Sp1 decoy oligodeoxynucleotide in carbon tetrachloride-induced liver fibrosis. J Gene Med. 2009;11(9):824–33. doi:10.​1002/​jgm.​1355.PubMedCrossRef
41.
go back to reference Chen H, Zhou Y, Chen KQ, An G, Ji SY, Chen QK. Anti-fibrotic effects via regulation of transcription factor Sp1 on hepatic stellate cells. Cell Physiol Biochem. 2012;29(1–2):51–60.PubMedCrossRef Chen H, Zhou Y, Chen KQ, An G, Ji SY, Chen QK. Anti-fibrotic effects via regulation of transcription factor Sp1 on hepatic stellate cells. Cell Physiol Biochem. 2012;29(1–2):51–60.PubMedCrossRef
44.
go back to reference Galli A, Crabb D, Price D, Ceni E, Salzano R, Surrenti C, et al. Peroxisome proliferator-activated receptor γ transcriptional regulation is involved in platelet-derived growth factor–induced proliferation of human hepatic stellate cells. Hepatology. 2000;31(1):101–8. doi:10.1002/hep.510310117.PubMedCrossRef Galli A, Crabb D, Price D, Ceni E, Salzano R, Surrenti C, et al. Peroxisome proliferator-activated receptor γ transcriptional regulation is involved in platelet-derived growth factor–induced proliferation of human hepatic stellate cells. Hepatology. 2000;31(1):101–8. doi:10.​1002/​hep.​510310117.PubMedCrossRef
48.
go back to reference Wakabayashi K-i, Okamura M, Tsutsumi S, Nishikawa NS, Tanaka T, Sakakibara I, et al. The peroxisome proliferator-activated receptor γ/retinoid X receptor α heterodimer targets the histone modification enzyme PR-Set7/Setd8 gene and regulates adipogenesis through a positive feedback loop. Mol Cell Biol. 2009;29(13):3544–55. doi:10.1128/mcb.01856-08.PubMedCentralPubMedCrossRef Wakabayashi K-i, Okamura M, Tsutsumi S, Nishikawa NS, Tanaka T, Sakakibara I, et al. The peroxisome proliferator-activated receptor γ/retinoid X receptor α heterodimer targets the histone modification enzyme PR-Set7/Setd8 gene and regulates adipogenesis through a positive feedback loop. Mol Cell Biol. 2009;29(13):3544–55. doi:10.​1128/​mcb.​01856-08.PubMedCentralPubMedCrossRef
51.
go back to reference Eeckhoute J, Oger F, Staels B, Lefebvre P. Coordinated regulation of PPARγ expression and activity through control of chromatin structure in adipogenesis and obesity. PPAR Res. 2012;2012:9. doi:10.1155/2012/164140.CrossRef Eeckhoute J, Oger F, Staels B, Lefebvre P. Coordinated regulation of PPARγ expression and activity through control of chromatin structure in adipogenesis and obesity. PPAR Res. 2012;2012:9. doi:10.​1155/​2012/​164140.CrossRef
52.
53.
go back to reference Lee TF, Mak KM, Rackovsky O, Lin Y-L, Kwong AJ, Loke JC, et al. Downregulation of hepatic stellate cell activation by retinol and palmitate mediated by adipose differentiation-related protein (ADRP). J Cell Physiol. 2010;223(3):648–57. doi:10.1002/jcp.22063.PubMedCentralPubMed Lee TF, Mak KM, Rackovsky O, Lin Y-L, Kwong AJ, Loke JC, et al. Downregulation of hepatic stellate cell activation by retinol and palmitate mediated by adipose differentiation-related protein (ADRP). J Cell Physiol. 2010;223(3):648–57. doi:10.​1002/​jcp.​22063.PubMedCentralPubMed
55.
go back to reference Najimi M, Khuu DN, Lysy PA, Jazouli N, Abarca J, Sempoux C, et al. Adult-derived human liver mesenchymal-like cells as a potential progenitor reservoir of hepatocytes? Cell Transplant. 2007;16(7):717–28.PubMedCrossRef Najimi M, Khuu DN, Lysy PA, Jazouli N, Abarca J, Sempoux C, et al. Adult-derived human liver mesenchymal-like cells as a potential progenitor reservoir of hepatocytes? Cell Transplant. 2007;16(7):717–28.PubMedCrossRef
56.
go back to reference Coll M, Taghdouini AE, Perea L, Mannaerts I, Vila-Casadesús M, Blaya D et al. Integrative miRNA and gene expression profiling analysis of human quiescent hepatic stellate cells. Sci Rep. 2015;5. doi:10.1038/srep11549. Coll M, Taghdouini AE, Perea L, Mannaerts I, Vila-Casadesús M, Blaya D et al. Integrative miRNA and gene expression profiling analysis of human quiescent hepatic stellate cells. Sci Rep. 2015;5. doi:10.​1038/​srep11549.
57.
go back to reference Mannaerts I, Nuytten NR, Rogiers V, Vanderkerken K, van Grunsven LA, Geerts A. Chronic administration of valproic acid inhibits activation of mouse hepatic stellate cells in vitro and in vivo. Hepatology. 2010;51(2):603–14. doi:10.1002/hep.23334.PubMedCrossRef Mannaerts I, Nuytten NR, Rogiers V, Vanderkerken K, van Grunsven LA, Geerts A. Chronic administration of valproic acid inhibits activation of mouse hepatic stellate cells in vitro and in vivo. Hepatology. 2010;51(2):603–14. doi:10.​1002/​hep.​23334.PubMedCrossRef
59.
go back to reference Berardis S, Lombard C, Evraerts J, El Taghdouini A, Rosseels V, Sancho-Bru P, et al. Gene Expression Profiling and Secretome Analysis Differentiate Adult-Derived Human Liver Stem/Progenitor Cells and Human Hepatic Stellate Cells. PLoS ONE. 2014;9(1). doi:10.1371/journal.pone.0086137. Berardis S, Lombard C, Evraerts J, El Taghdouini A, Rosseels V, Sancho-Bru P, et al. Gene Expression Profiling and Secretome Analysis Differentiate Adult-Derived Human Liver Stem/Progenitor Cells and Human Hepatic Stellate Cells. PLoS ONE. 2014;9(1). doi:10.​1371/​journal.​pone.​0086137.
Metadata
Title
In vitro reversion of activated primary human hepatic stellate cells
Authors
Adil El Taghdouini
Mustapha Najimi
Pau Sancho-Bru
Etienne Sokal
Leo A. van Grunsven
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Fibrogenesis & Tissue Repair / Issue 1/2015
Electronic ISSN: 1755-1536
DOI
https://doi.org/10.1186/s13069-015-0031-z

Other articles of this Issue 1/2015

Fibrogenesis & Tissue Repair 1/2015 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine