[en] Adamalysins, a family of metalloproteinases containing a disintegrin and metalloproteinases (ADAMs) and ADAM with thrombospondin motifs (ADAMTSs), belong to the matrisome and play important roles in various biological and pathological processes, such as development, immunity and cancer. Using a liver cancer dataset from the International Cancer Genome Consortium, we developed an extensive in silico screening that identified a cluster of adamalysins co-expressed in livers from patients with hepatocellular carcinoma (HCC). Within this cluster, ADAMTS12 expression was highly associated with recurrence risk and poorly differentiated HCC signatures. We showed that ADAMTS12 was expressed in the stromal cells of the tumor and adjacent fibrotic tissues of HCC patients, and more specifically in activated stellate cells. Using a mouse model of carbon tetrachloride-induced liver injury, we showed that Adamts12 was strongly and transiently expressed after a 24 h acute treatment, and that fibrosis was exacerbated in Adamts12-null mice submitted to carbon tetrachloride-induced chronic liver injury. Using the HSC-derived LX-2 cell line, we showed that silencing of ADAMTS12 resulted in profound changes of the gene expression program. In particular, genes previously reported to be induced upon HSC activation, such as PAI-1, were mostly down-regulated following ADAMTS12 knock-down. The phenotype of these cells was changed to a less differentiated state, showing an altered actin network and decreased nuclear spreading. These phenotypic changes, together with the down-regulation of PAI-1, were offset by TGF-β treatment. The present study thus identifies ADAMTS12 as a modulator of HSC differentiation, and a new player in chronic liver disease.
Disciplines :
Oncology
Author, co-author :
Dekky, Bassil ✱; University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
Azar, Fida ✱; University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
Bonnier, Dominique; University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
Kalebić, Chiara; University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
Arpigny, Esther ; Université de Liège - ULiège > Département des sciences biomédicales et précliniques
Colige, Alain ; Université de Liège - ULiège > GIGA > GIGA Cancer - Connective Tissue Biology
Legagneux, Vincent ; University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
Théret, Nathalie ; University of Rennes, INSERM, EHESP, IRSET (Institut de Recherche en Santé, Environnement et Travail)-UMR_S 1085, Rennes, France
✱ These authors have contributed equally to this work.
Language :
English
Title :
ADAMTS12 is a stromal modulator in chronic liver disease.
Publication date :
November 2023
Journal title :
FASEB Journal
ISSN :
0892-6638
eISSN :
1530-6860
Publisher :
John Wiley and Sons Inc, United States
Volume :
37
Issue :
11
Pages :
e23237
Peer reviewed :
Peer Reviewed verified by ORBi
Funders :
Fonds De La Recherche Scientifique - FNRS Ligue Contre le Cancer
Funding text :
This work was supported by the Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Rennes 1, the Ligue Contre le Cancer and the Région Bretagne. BD was recipient of PhD fellowships from the Ligue Contre le Cancer and Région Bretagne. A. C. and C. M are supported by the “Fonds de la Recherche Scientifique–FNRS” (Grant 7.6536.18), the “Fonds Léon Frédéricq” and ULiège. The authors thank the Rennes Biological Resources Center (CHRU Pontchaillou, IFR 140) for its contribution to human tissue sampling, and the Biosit H2P2 facility for histological studies. We acknowledge the excellent support of the GIGA center at University of Liege, Belgium, in which the animal experimentations were carried out. RNA-sequencing (libraries, runs and primary analyses) was performed by the GenoBiRD facility homed by the Structure Fédérative de Recherche en Santé François Bonamy, University of Nantes (France). Histopathological analyses were performed at the HistoPathology of High Precision facility (H2P2) hosted at UMS Biosit (Inserm UMS 018, CNRS UMS3480). Computing resources were provided by the GenOuest Bioinformatics, a BioGenOuest facility hosted at IRISA/INRIA Rennes Bretagne Atlantique. The authors thank Dr. C. Lucas (Service Biochimie, CHU Rennes) for enzyme measurements, Dr Vincent Guen (CNRS, University of Rennes, France) and Bruno Turlin (Univ Rennes, CHU Rennes, F-35000 Rennes, France) for fruitful discussions, Frederic Ezan and Michel Rauch for technical assistance, Olivier Collin for the excellent support of the GenOuest bioinformatics core facility, Servane Le Page and Laurence Huc for providing anti-GPR91 antibodies, and Dr Catherine Lavau (Inserm U1085, University of Rennes 1) for critical reading of the manuscript. Fida Azar's thesis was co-financed by the municipality of Hayata (Lebanon) and supervised within the frame of a partnership between le Lebanese University (Beirut, Lebanon) and the University of Rennes (France).This work was supported by the Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Rennes 1, the Ligue Contre le Cancer and the Région Bretagne. BD was recipient of PhD fellowships from the Ligue Contre le Cancer and Région Bretagne. A. C. and C. M are supported by the “Fonds de la Recherche Scientifique–FNRS” (Grant 7.6536.18), the “Fonds Léon Frédéricq” and ULiège. The authors thank the Rennes Biological Resources Center (CHRU Pontchaillou, IFR 140) for its contribution to human tissue sampling, and the Biosit H2P2 facility for histological studies. We acknowledge the excellent support of the GIGA center at University of Liege, Belgium, in which the animal experimentations were carried out. RNA‐sequencing (libraries, runs and primary analyses) was performed by the GenoBiRD facility homed by the Structure Fédérative de Recherche en Santé François Bonamy, University of Nantes (France). Histopathological analyses were performed at the HistoPathology of High Precision facility (H2P2) hosted at UMS Biosit (Inserm UMS 018, CNRS UMS3480). Computing resources were provided by the GenOuest Bioinformatics, a BioGenOuest facility hosted at IRISA/INRIA Rennes Bretagne Atlantique. The authors thank Dr. C. Lucas (Service Biochimie, CHU Rennes) for enzyme measurements, Dr Vincent Guen (CNRS, University of Rennes, France) and Bruno Turlin (Univ Rennes, CHU Rennes, F‐35000 Rennes, France) for fruitful discussions, Frederic Ezan and Michel Rauch for technical assistance, Olivier Collin for the excellent support of the GenOuest bioinformatics core facility, Servane Le Page and Laurence Huc for providing anti‐GPR91 antibodies, and Dr Catherine Lavau (Inserm U1085, University of Rennes 1) for critical reading of the manuscript. Fida Azar's thesis was co‐financed by the municipality of Hayata (Lebanon) and supervised within the frame of a partnership between le Lebanese University (Beirut, Lebanon) and the University of Rennes (France).
Théret N, Musso O, Turlin B, et al. Increased extracellular matrix remodeling is associated with tumor progression in human hepatocellular carcinomas. Hepatology. 2001;34:82-88.
Cordero-Espinoza L, Huch M. The balancing act of the liver: tissue regeneration versus fibrosis. J Clin Invest. 2018;128:85-96.
Naba A, Clauser KR, Hoersch S, Liu H, Carr SA, Hynes RO. The matrisome: in silico definition and in vivo characterization by proteomics of normal and tumor extracellular matrices. Mol Cell Proteomics. 2012;11:M111.014647.
Massey VL, Dolin CE, Poole LG, et al. The hepatic “matrisome” responds dynamically to injury: characterization of transitional changes to the extracellular matrix in mice. Hepatology. 2017;65:969-982.
Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 2017;14:397-411.
Théret N, Bouezzedine F, Azar F, Diab-Assaf M, Legagneux V. ADAM and ADAMTS proteins, new players in the regulation of hepatocellular carcinoma microenvironment. Cancers (Basel). 2021;13:1563.
Edwards DR, Handsley MM, Pennington CJ. The ADAM metalloproteinases. Mol Aspects Med. 2008;29:258-289.
Apte SS. A disintegrin-like and metalloprotease (reprolysin-type) with thrombospondin type 1 motif (ADAMTS) superfamily: functions and mechanisms. J Biol Chem. 2009;284:31493-31497.
Le Pabic H, Bonnier D, Wewer UM, et al. ADAM12 in human liver cancers: TGF-beta-regulated expression in stellate cells is associated with matrix remodeling. Hepatology. 2003;37:1056-1066.
Atfi A, Dumont E, Colland F, et al. The disintegrin and metalloproteinase ADAM12 contributes to TGF-beta signaling through interaction with the type II receptor. J Cell Biol. 2007;178:201-208.
Leyme A, Bourd-Boittin K, Bonnier D, Falconer A, Arlot-Bonnemains Y, Théret N. Identification of ILK as a new partner of the ADAM12 disintegrin and metalloprotease in cell adhesion and survival. Mol Biol Cell. 2012;23:3461-3472.
Mazzocca A, Coppari R, De Franco R, et al. A secreted form of ADAM9 promotes carcinoma invasion through tumor-stromal interactions. Cancer Res. 2005;65:4728-4738.
Schwettmann L, Wehmeier M, Jokovic D, et al. Hepatic expression of A disintegrin and metalloproteinase (ADAM) and ADAMs with thrombospondin motives (ADAM-TS) enzymes in patients with chronic liver diseases. J Hepatol. 2008;49:243-250.
Bourd-Boittin K, Basset L, Bonnier D, L'helgoualc'h A, Samson M, Théret N. CX3CL1/fractalkine shedding by human hepatic stellate cells: contribution to chronic inflammation in the liver. J Cell Mol Med. 2009;13:1526-1535.
Fujita T, Maesawa C, Oikawa K, Nitta H, Wakabayashi G, Masuda T. Interferon-gamma down-regulates expression of tumor necrosis factor-alpha converting enzyme/a disintegrin and metalloproteinase 17 in activated hepatic stellate cells of rats. Int J Mol Med. 2006;17:605-616.
Oikawa H, Maesawa C, Tatemichi Y, et al. A disintegrin and metalloproteinase 17 (ADAM17) mediates epidermal growth factor receptor transactivation by angiotensin II on hepatic stellate cells. Life Sci. 2014;97:137-144.
Xia Y, Chen R, Song Z, et al. Gene expression profiles during activation of cultured rat hepatic stellate cells by tumoral hepatocytes and fetal bovine serum. J Cancer Res Clin Oncol. 2010;136:309-321.
Mazzocca A, Giannelli G, Antonaci S. Involvement of ADAMs in tumorigenesis and progression of hepatocellular carcinoma: is it merely fortuitous or a real pathogenic link? Biochim Biophys Acta. 2010;1806:74-81.
Goto K, Arai J, Stephanou A, Kato N. Novel therapeutic features of disulfiram against hepatocellular carcinoma cells with inhibitory effects on a disintegrin and metalloproteinase 10. Oncotarget. 2018;9:18821-18831.
Diamantis I, Lüthi M, Hösli M, Reichen J. Cloning of the rat ADAMTS-1 gene and its down regulation in endothelial cells in cirrhotic rats. Liver. 2000;20:165-172.
Bourd-Boittin K, Bonnier D, Leyme A, et al. Protease profiling of liver fibrosis reveals the ADAM metallopeptidase with thrombospondin type 1 motif, 1 as a central activator of transforming growth factor beta. Hepatology. 2011;54:2173-2184.
Kesteloot F, Desmoulière A, Leclercq I, et al. ADAM metallopeptidase with thrombospondin type 1 motif 2 inactivation reduces the extent and stability of carbon tetrachloride-induced hepatic fibrosis in mice. Hepatology. 2007;46:1620-1631.
Bekhouche M, Leduc C, Dupont L, et al. Determination of the substrate repertoire of ADAMTS2, 3, and 14 significantly broadens their functions and identifies extracellular matrix organization and TGF-β signaling as primary targets. FASEB J. 2016;30:1741-1756.
Bauters D, Spincemaille P, Geys L, et al. ADAMTS5 deficiency protects against non-alcoholic steatohepatitis in obesity. Liver Int. 2016;36:1848-1859.
Pi L, Jorgensen M, Oh S-H, et al. A disintegrin and metalloprotease with thrombospondin type I motif 7: a new protease for connective tissue growth factor in hepatic progenitor/oval cell niche. Am J Pathol. 2015;185:1552-1563.
Uemura M, Fujimura Y, Ko S, Matsumoto M, Nakajima Y, Fukui H. Pivotal role of ADAMTS13 function in liver diseases. Int J Hematol. 2010;91:20-29.
El Hour M, Moncada-Pazos A, Blacher S, et al. Higher sensitivity of Adamts12-deficient mice to tumor growth and angiogenesis. Oncogene. 2010;29:3025-3032.
Cancer Genome Atlas Research Network. Comprehensive and integrative genomic characterization of hepatocellular carcinoma. Cell. 2017;169:1327-1341.e23.
Bekhouche M, Colige A. The procollagen N-proteinases ADAMTS2, 3 and 14 in pathophysiology. Matrix Biol. 2015;44–46:46-53.
Bukong TN, Maurice SB, Chahal B, Schaeffer DF, Winwood PJ. Versican: a novel modulator of hepatic fibrosis. Lab Invest. 2016;96:361-374.
Guilliams M, Bonnardel J, Haest B, et al. Spatial proteogenomics reveals distinct and evolutionarily conserved hepatic macrophage niches. Cell. 2022;185:379-396.e38.
Azar F, Courtet K, Dekky B, et al. Integration of miRNA-regulatory networks in hepatic stellate cells identifies TIMP3 as a key factor in chronic liver disease. Liver Int. 2020;40:2021-2033.
Xu L, Hui AY, Albanis E, et al. Human hepatic stellate cell lines, LX-1 and LX-2: new tools for analysis of hepatic fibrosis. Gut. 2005;54:142-151.
Cantagrel V, Silhavy JL, Bielas SL, et al. Mutations in the cilia gene ARL13B lead to the classical form of Joubert syndrome. Am J Hum Genet. 2008;83:170-179.
Takahara T, Zhang LP, Yata Y, et al. Modulation of matrix metalloproteinase-9 in hepatic stellate cells by three-dimensional type I collagen: its activation and signaling pathway. Hepatol Res. 2003;26:318-326.
Robert S, Gicquel T, Bodin A, Lagente V, Boichot E. Characterization of the MMP/TIMP imbalance and collagen production induced by IL-1β or TNF-α release from human hepatic stellate cells. PLoS ONE. 2016;11:e0153118.
Tahashi Y, Matsuzaki K, Date M, et al. Differential regulation of TGF-beta signal in hepatic stellate cells between acute and chronic rat liver injury. Hepatology. 2002;35:49-61.
Maniotis AJ, Chen CS, Ingber DE. Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure. Proc Natl Acad Sci U S A. 1997;94:849-854.
Wang N, Tytell JD, Ingber DE. Mechanotransduction at a distance: mechanically coupling the extracellular matrix with the nucleus. Nat Rev Mol Cell Biol. 2009;10:75-82.
Thomas CH, Collier JH, Sfeir CS, Healy KE. Engineering gene expression and protein synthesis by modulation of nuclear shape. Proc Natl Acad Sci U S A. 2002;99:1972-1977.
De Smet V, Eysackers N, Merens V, et al. Initiation of hepatic stellate cell activation extends into chronic liver disease. Cell Death Dis. 2021;12:1110.
De Minicis S, Seki E, Uchinami H, et al. Gene expression profiles during hepatic stellate cell activation in culture and in vivo. Gastroenterology. 2007;132:1937-1946.
El Taghdouini A, Sørensen AL, Reiner AH, et al. Genome-wide analysis of DNA methylation and gene expression patterns in purified, uncultured human liver cells and activated hepatic stellate cells. Oncotarget. 2015;6:26729-26745.
Rosenthal SB, Liu X, Ganguly S, et al. Heterogeneity of HSCs in a mouse model of NASH. Hepatology. 2021;74:667-685.
El Taghdouini A, Najimi M, Sancho-Bru P, Sokal E, van Grunsven LA. In vitro reversion of activated primary human hepatic stellate cells. Fibrogenesis Tissue Repair. 2015;8:14.
Rabadán R, Mohamedi Y, Rubin U, et al. Identification of relevant genetic alterations in cancer using topological data analysis. Nat Commun. 2020;11:3808.
Wang D, Zhu T, Zhang F-B, He C. Expression of ADAMTS12 in colorectal cancer-associated stroma prevents cancer development and is a good prognostic indicator of colorectal cancer. Dig Dis Sci. 2011;56:3281-3287.
He R-Z, Zheng J-H, Yao H-F, et al. ADAMTS12 promotes migration and epithelial-mesenchymal transition and predicts poor prognosis for pancreatic cancer. Hepatobiliary Pancreat Dis Int. 2023;22(2):169-178.
Hu G, Sun N, Jiang J, Chen X. Establishment of a 5-gene risk model related to regulatory T cells for predicting gastric cancer prognosis. Cancer Cell Int. 2020;20:433.
Kunadirek P, Chuaypen N, Jenjaroenpun P, et al. Cell-free DNA analysis by whole-exome sequencing for hepatocellular carcinoma: a pilot study in Thailand. Cancers (Basel). 2021;13:2229.
Kordes C, Sawitza I, Götze S, Häussinger D. Hepatic stellate cells support hematopoiesis and are liver-resident mesenchymal stem cells. Cell Physiol Biochem. 2013;31:290-304.
Amann T, Bataille F, Spruss T, et al. Activated hepatic stellate cells promote tumorigenicity of hepatocellular carcinoma. Cancer Sci. 2009;100:646-653.
Carloni V, Luong TV, Rombouts K. Hepatic stellate cells and extracellular matrix in hepatocellular carcinoma: more complicated than ever. Liver Int. 2014;34:834-843.
Fregni G, Quinodoz M, Möller E, et al. Reciprocal modulation of mesenchymal stem cells and tumor cells promotes lung cancer metastasis. EBioMedicine. 2018;29:128-145.
Corbit KC, Aanstad P, Singla V, Norman AR, Stainier DYR, Reiter JF. Vertebrate smoothened functions at the primary cilium. Nature. 2005;437:1018-1021.
Michelotti GA, Xie G, Swiderska M, et al. Smoothened is a master regulator of adult liver repair. J Clin Invest. 2013;123:2380-2394.
Pitaval A, Tseng Q, Bornens M, Théry M. Cell shape and contractility regulate ciliogenesis in cell cycle-arrested cells. J Cell Biol. 2010;191:303-312.
Drummond ML, Li M, Tarapore E, et al. Actin polymerization controls cilia-mediated signaling. J Cell Biol. 2018;217:3255-3266.
Bai XH, Wang DW, Luan Y, Yu XP, Liu CJ. Regulation of chondrocyte differentiation by ADAMTS-12 metalloproteinase depends on its enzymatic activity. Cell Mol Life Sci. 2009;66:667-680.
Olsen AL, Bloomer SA, Chan EP, et al. Hepatic stellate cells require a stiff environment for myofibroblastic differentiation. Am J Physiol Gastrointest Liver Physiol. 2011;301:G110-G118.
Weiskirchen R, Weimer J, Meurer SK, et al. Genetic characteristics of the human hepatic stellate cell line LX-2. PLoS One. 2013;8:e75692.
Dou C, Liu Z, Tu K, et al. P300 acetyltransferase mediates stiffness-induced activation of hepatic stellate cells into tumor-promoting myofibroblasts. Gastroenterology. 2018;154:2209-2221.e14.
Kisseleva T, Cong M, Paik Y, et al. Myofibroblasts revert to an inactive phenotype during regression of liver fibrosis. Proc Natl Acad Sci U S A. 2012;109:9448-9453.
Correa PRAV, Kruglov EA, Thompson M, Leite MF, Dranoff JA, Nathanson MH. Succinate is a paracrine signal for liver damage. J Hepatol. 2007;47:262-269.
Krizhanovsky V, Yon M, Dickins RA, et al. Senescence of activated stellate cells limits liver fibrosis. Cell. 2008;134:657-667.
Paulissen G, El Hour M, Rocks N, et al. Control of allergen-induced inflammation and hyperresponsiveness by the metalloproteinase ADAMTS-12. J Immunol. 2012;189:4135-4143.
Moncada-Pazos A, Obaya AJ, Llamazares M, et al. ADAMTS-12 metalloprotease is necessary for normal inflammatory response. J Biol Chem. 2012;287:39554-39563.