DOI QR코드

DOI QR Code

Integrin alpha 11 in the regulation of the myofibroblast phenotype: implications for fibrotic diseases

  • Bansal, Ruchi (Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente) ;
  • Nakagawa, Shigeki (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Yazdani, Saleh (Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente) ;
  • van Baarlen, Joop (Laboratorium Pathologie Oost-Nederland) ;
  • Venkatesh, Anu (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Koh, Anna P (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Song, Won-Min (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Goossens, Nicolas (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Watanabe, Hideo (Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School) ;
  • Beasley, Mary B (Department of Medicine, Icahn School of Medicine at Mount Sinai) ;
  • Powell, Charles A (Division of Pulmonary, Critical Care, and Sleep Medicine, Icahn School of Medicine at Mount Sinai) ;
  • Storm, Gert (Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente) ;
  • Kaminski, Naftali (Yale School of Medicine, Pulmonary, Critical Care and Sleep Medicine) ;
  • van Goor, Harry (Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen) ;
  • Friedman, Scott L (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Hoshida, Yujin (Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute) ;
  • Prakash, Jai (Targeted Therapeutics, Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, Faculty of Science and Technology, University of Twente)
  • Received : 2016.12.09
  • Accepted : 2017.05.14
  • Published : 2017.11.30

Abstract

Tissue fibrosis, characterized by excessive accumulation of aberrant extracellular matrix (ECM) produced by myofibroblasts, is a growing cause of mortality worldwide. Understanding the factors that induce myofibroblastic differentiation is paramount to prevent or reverse the fibrogenic process. Integrin-mediated interaction between the ECM and cytoskeleton promotes myofibroblast differentiation. In the present study, we explored the significance of integrin alpha 11 (ITGA11), the integrin alpha subunit that selectively binds to type I collagen during tissue fibrosis in the liver, lungs and kidneys. We showed that ITGA11 was co-localized with ${\alpha}$-smooth muscle actin-positive myofibroblasts and was correlatively induced with increasing fibrogenesis in mouse models and human fibrotic organs. Furthermore, transcriptome and protein expression analysis revealed that ITGA11 knockdown in hepatic stellate cells (liver-specific myofibroblasts) markedly reduced transforming growth factor ${\beta}$-induced differentiation and fibrotic parameters. Moreover, ITGA11 knockdown dramatically altered the myofibroblast phenotype, as indicated by the loss of protrusions, attenuated adhesion and migration, and impaired contractility of collagen I matrices. Furthermore, we demonstrated that ITGA11 was regulated by the hedgehog signaling pathway, and inhibition of the hedgehog pathway reduced ITGA11 expression and fibrotic parameters in human hepatic stellate cells in vitro, in liver fibrosis mouse model in vivo and in human liver slices ex vivo. Therefore, we speculated that ITGA11 might be involved in fibrogenic signaling and might act downstream of the hedgehog signaling pathway. These findings highlight the significance of the ITGA11 receptor as a highly promising therapeutic target in organ fibrosis.

Keywords

References

  1. Wynn TA. Cellular and molecular mechanisms of fibrosis. J Pathol 2008; 214: 199-210. https://doi.org/10.1002/path.2277
  2. Desmouliere A, Darby IA, Gabbiani G. Normal and pathologic soft tissue remodeling: role of the myofibroblast, with special emphasis on liver and kidney fibrosis. Lab Invest 2003; 83: 1689-1707. https://doi.org/10.1097/01.LAB.0000101911.53973.90
  3. Hinz B, Phan SH, Thannickal VJ, Prunotto M, Desmouliere A, Varga J et al. Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am J Pathol 2012; 180: 1340-1355. https://doi.org/10.1016/j.ajpath.2012.02.004
  4. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther 2006; 5: 1640-1646. https://doi.org/10.4161/cbt.5.12.3354
  5. Wynn TA. Integrating mechanisms of pulmonary fibrosis. J Exp Med 2011; 208: 1339-1350. https://doi.org/10.1084/jem.20110551
  6. Hinz B, Phan SH, Thannickal VJ, Galli A, Bochaton-Piallat ML, Gabbiani G. The myofibroblast: one function, multiple origins. Am J Pathol 2007; 170: 1807-1816. https://doi.org/10.2353/ajpath.2007.070112
  7. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 2010; 176: 85-97. https://doi.org/10.2353/ajpath.2010.090517
  8. Kalluri R, Neilson EG. Epithelial-mesenchymal transition and its implications for fibrosis. J Clin Invest 2003; 112: 1776-1784. https://doi.org/10.1172/JCI200320530
  9. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell 2009; 139: 871-890. https://doi.org/10.1016/j.cell.2009.11.007
  10. Piera-Velazquez S, Li Z, Jimenez SA. Role of endothelial-mesenchymal transition (EndoMT) in the pathogenesis of fibrotic disorders. Am J Pathol 2011; 179: 1074-1080. https://doi.org/10.1016/j.ajpath.2011.06.001
  11. Friedman SL. Mechanisms of hepatic fibrogenesis. Gastroenterology 2008; 134: 1655-1669. https://doi.org/10.1053/j.gastro.2008.03.003
  12. Gabbiani G. The myofibroblast in wound healing and fibrocontractive diseases. J Pathol 2003; 200: 500-503. https://doi.org/10.1002/path.1427
  13. Gressner OA, Weiskirchen R, Gressner AM. Evolving concepts of liver fibrogenesis provide new diagnostic and therapeutic options. Comp Hepatol 2007; 6: 7. https://doi.org/10.1186/1476-5926-6-7
  14. Trautwein C, Friedman SL, Schuppan D, Pinzani M. Hepatic fibrosis: concept to treatment. J Hepatol 2015; 62: S15-S24. https://doi.org/10.1016/j.jhep.2015.02.039
  15. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med 2012; 18: 1028-1040. https://doi.org/10.1038/nm.2807
  16. Agarwal SK. Integrins and cadherins as therapeutic targets in fibrosis. Front Pharmacol 2014; 5: 131.
  17. Henderson NC, Sheppard D. Integrin-mediated regulation of TGFbeta in fibrosis. Biochim Biophys Acta 2013; 1832: 891-896. https://doi.org/10.1016/j.bbadis.2012.10.005
  18. Humphries JD, Byron A, Humphries MJ. Integrin ligands at a glance. J Cell Sci 2006; 119: 3901-3903. https://doi.org/10.1242/jcs.03098
  19. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell 2002; 110: 673-687. https://doi.org/10.1016/S0092-8674(02)00971-6
  20. Henderson NC, Arnold TD, Katamura Y, Giacomini MM, Rodriguez JD, McCarty JH et al. Targeting of alphav integrin identifies a core molecular pathway that regulates fibrosis in several organs. Nat Med 2013; 19: 1617-1624. https://doi.org/10.1038/nm.3282
  21. Munger JS, Huang X, Kawakatsu H, Griffiths MJ, Dalton SL, Wu J et al. The integrin alpha v beta 6 binds and activates latent TGF beta 1: a mechanism for regulating pulmonary inflammation and fibrosis. Cell 1999; 96: 319-328. https://doi.org/10.1016/S0092-8674(00)80545-0
  22. Popova SN, Lundgren-Akerlund E, Wiig H, Gullberg D. Physiology and pathology of collagen receptors. Acta Physiol (Oxf) 2007; 190: 179-187. https://doi.org/10.1111/j.1748-1716.2007.01718.x
  23. Tiger CF, Fougerousse F, Grundstrom G, Velling T, Gullberg D. ${\alpha}11{\beta}1$ integrin is a receptor for interstitial collagens involved in cell migration and collagen reorganization on mesenchymal nonmuscle cells. Dev Biol 2001; 237: 116-129. https://doi.org/10.1006/dbio.2001.0363
  24. Popova SN, Rodriguez-Sanchez B, Liden A, Betsholtz C, Van Den Bos T, Gullberg D. The mesenchymal alpha11beta1 integrin attenuates PDGF-BBstimulated chemotaxis of embryonic fibroblasts on collagens. Dev Biol 2004; 270: 427-442. https://doi.org/10.1016/j.ydbio.2004.03.006
  25. Talior-Volodarsky I, Connelly KA, Arora PD, Gullberg D, McCulloch CA. ${\alpha}11$ integrin stimulates myofibroblast differentiation in diabetic cardiomyopathy. Cardiovasc Res 2012; 96: 265-275. https://doi.org/10.1093/cvr/cvs259
  26. Honda E, Yoshida K, Munakata H. Transforming growth factor-beta upregulates the expression of integrin and related proteins in MRC-5 human myofibroblasts. Tohoku J Exp Med 2010; 220: 319-327. https://doi.org/10.1620/tjem.220.319
  27. Lu N, Carracedo S, Ranta J, Heuchel R, Soininen R, Gullberg D. The human alpha11 integrin promoter drives fibroblast-restricted expression in vivo and is regulated by TGF-beta1 in a Smad- and Sp1- dependent manner. Matrix Biol 2010; 29: 166-176. https://doi.org/10.1016/j.matbio.2009.11.003
  28. Carracedo S, Lu N, Popova SN, Jonsson R, Eckes B, Gullberg D. The fibroblast integrin alpha11beta1 is induced in a mechanosensitive manner involving activin A and regulates myofibroblast differentiation. J Biol Chem 2010; 285: 10434-10445. https://doi.org/10.1074/jbc.M109.078766
  29. Navab R, Strumpf D, To C, Pasko E, Kim KS, Park CJ et al. Integrin alpha11beta1 regulates cancer stromal stiffness and promotes tumorigenicity and metastasis in non-small cell lung cancer. Oncogene 2016; 35: 1899-1908. https://doi.org/10.1038/onc.2015.254
  30. Vaira V, Fedele G, Pyne S, Fasoli E, Zadra G, Bailey D et al. Preclinical model of organotypic culture for pharmacodynamic profiling of human tumors. Proc Natl Acad Sci USA 2010; 107: 8352-8356. https://doi.org/10.1073/pnas.0907676107
  31. Intraobserver and interobserver variations in liver biopsy interpretation in patients with chronic hepatitis C. The French METAVIR Cooperative Study Group. Hepatology 1994; 20: 15-20. https://doi.org/10.1002/hep.1840200104
  32. Kramann R, Schneider RK, DiRocco DP, Machado F, Fleig S, Bondzie PA et al. Perivascular Gli1+ progenitors are key contributors to injury-induced organ fibrosis. Cell Stem Cell 2015; 16: 51-66. https://doi.org/10.1016/j.stem.2014.11.004
  33. Inagaki Y, Okazaki I. Emerging insights into transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut 2007; 56: 284-292. https://doi.org/10.1136/gut.2005.088690
  34. Margadant C, Sonnenberg A. Integrin-TGF-beta crosstalk in fibrosis, cancer and wound healing. EMBO Rep 2010; 11: 97-105. https://doi.org/10.1038/embor.2009.276
  35. Hu L, Lin X, Lu H, Chen B, Bai Y. An overview of hedgehog signaling in fibrosis. Mol Pharmacol 2015; 87: 174-182. https://doi.org/10.1124/mol.114.095141
  36. Olinga P, Schuppan D. Precision-cut liver slices: a tool to model the liver ex vivo. J Hepatol 2013; 58: 1252-1253. https://doi.org/10.1016/j.jhep.2013.01.009

Cited by

  1. Fibroblast polarization over the myocardial infarction time continuum shifts roles from inflammation to angiogenesis vol.114, pp.2, 2017, https://doi.org/10.1007/s00395-019-0715-4
  2. Integrin α11 in pancreatic stellate cells regulates tumor stroma interaction in pancreatic cancer vol.33, pp.5, 2019, https://doi.org/10.1096/fj.201802336r
  3. α11β1 Integrin is Induced in a Subset of Cancer-Associated Fibroblasts in Desmoplastic Tumor Stroma and Mediates In Vitro Cell Migration vol.11, pp.6, 2017, https://doi.org/10.3390/cancers11060765
  4. Magnetic-Assisted Treatment of Liver Fibrosis vol.8, pp.10, 2019, https://doi.org/10.3390/cells8101279
  5. The Many Roles of Cell Adhesion Molecules in Hepatic Fibrosis vol.8, pp.12, 2017, https://doi.org/10.3390/cells8121503
  6. Integrin α11 in non–small cell lung cancer is associated with tumor progression and postoperative recurrence vol.111, pp.1, 2020, https://doi.org/10.1111/cas.14257
  7. Integrin Binding Dynamics Modulate Ligand-Specific Mechanosensing in Mammary Gland Fibroblasts vol.23, pp.3, 2020, https://doi.org/10.1016/j.isci.2020.100907
  8. The Potential Application of Magnetic Nanoparticles for Liver Fibrosis Theranostics vol.9, pp.None, 2017, https://doi.org/10.3389/fchem.2021.674786
  9. Cancer‐associated fibroblast migration in non‐small cell lung cancers is modulated by increased integrin α11 expression vol.15, pp.5, 2017, https://doi.org/10.1002/1878-0261.12937
  10. Protein Signatures of Remodeled Airways in Transplanted Lungs with Bronchiolitis Obliterans Syndrome Obtained Using Laser-Capture Microdissection vol.191, pp.8, 2017, https://doi.org/10.1016/j.ajpath.2021.05.014
  11. Tumor Nonimmune-Microenvironment-Related Gene Expression Signature Predicts Brain Metastasis in Lung Adenocarcinoma Patients after Surgery: A Machine Learning Approach Using Gene Expression Profiling vol.13, pp.17, 2017, https://doi.org/10.3390/cancers13174468
  12. Lung epithelial and endothelial damage, loss of tissue repair, inhibition of fibrinolysis, and cellular senescence in fatal COVID-19 vol.13, pp.620, 2017, https://doi.org/10.1126/scitranslmed.abj7790
  13. New Therapeutic Targets for Hepatic Fibrosis in the Integrin Family, α8β1 and α11β1, Induced Specifically on Activated Stellate Cells vol.22, pp.23, 2021, https://doi.org/10.3390/ijms222312794
  14. Transcriptomic and Epigenetic Profiling of Fibroblasts in Idiopathic Pulmonary Fibrosis vol.66, pp.1, 2022, https://doi.org/10.1165/rcmb.2020-0437oc