Pathophysiology and Treatment Options for Hepatic Fibrosis: Can It Be Completely Cured?
Abstract
:1. Introduction
2. Triggers of Hepatic Fibrosis
2.1. Viral Hepatitis
2.2. Alcoholic Liver Disease (ALD)
2.3. NAFLD/NASH
2.4. Autoimmune Liver Disease
3. Role of HSCs in Liver Fibrosis
4. Role of LSECs in Hepatic Fibrosis
5. Role of Liver-Resident Macrophages in Hepatic Fibrosis
6. Role of Liver-Resident Lymphocytes in Hepatic Fibrosis
7. Role of Exosomes in Hepatic Fibrosis
8. Role of Apoptotic Bodies in Hepatic Fibrosis
9. Role of Inflammasomes in Hepatic Fibrosis
10. Role of MicroRNAs (miRNAs) in Hepatic Fibrosis
11. Therapeutic Approaches to Target Liver Fibrosis
11.1. Targeted Therapies against Nuclear Receptors
11.2. Targeted Therapies against HSC Activation
11.3. Targeting Therapies against Inflammation and Oxidative Stress
11.4. Targeted Therapies against Renin-Angiotensin System (RAS)
12. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Bataller, R.; Brenner, D.A. Liver fibrosis. J. Clin. Investig. 2005, 115, 209–218. [Google Scholar] [CrossRef] [PubMed]
- Marrone, G.; Shah, V.H.; Gracia-Sancho, J. Sinusoidal communication in liver fibrosis and regeneration. J. Hepatol. 2016, 65, 608–617. [Google Scholar] [CrossRef] [Green Version]
- Huang, E.; Peng, N.; Xiao, F.; Hu, D.; Wang, X.; Lu, L. The Roles of Immune Cells in the Pathogenesis of Fibrosis. Int. J. Mol. Sci. 2020, 21, 5203. [Google Scholar] [CrossRef] [PubMed]
- He, W.; Dai, C. Key Fibrogenic Signaling. Curr. Pathobiol. Rep. 2015, 3, 183–192. [Google Scholar] [CrossRef] [Green Version]
- Sziksz, E.; Pap, D.; Lippai, R.; Beres, N.J.; Fekete, A.; Szabo, A.J.; Vannay, A. Fibrosis Related Inflammatory Mediators: Role of the IL-10 Cytokine Family. Mediators. Inflamm. 2015, 2015, 764641. [Google Scholar] [CrossRef] [PubMed]
- Seki, E.; Schwabe, R.F. Hepatic inflammation and fibrosis: Functional links and key pathways. Hepatology 2015, 61, 1066–1079. [Google Scholar] [CrossRef]
- Lee, U.E.; Friedman, S.L. Mechanisms of hepatic fibrogenesis. Best Pract. Res. Clin. Gastroenterol. 2011, 25, 195–206. [Google Scholar] [CrossRef]
- McQuitty, C.E.; Williams, R.; Chokshi, S.; Urbani, L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front. Immunol. 2020, 11, 574276. [Google Scholar] [CrossRef] [PubMed]
- Alkhouri, N.; McCullough, A.J. Noninvasive Diagnosis of NASH and Liver Fibrosis Within the Spectrum of NAFLD. Gastroenterol. Hepatol. 2012, 8, 661–668. [Google Scholar]
- Sun, C.; Hu, J.J.; Pan, Q.; Cao, Y.; Fan, J.G.; Li, G.M. Hepatic differentiation of rat induced pluripotent stem cells in vitro. World J. Gastroenterol. 2015, 21, 11118–11126. [Google Scholar] [CrossRef]
- Osna, N.A.; Donohue, T.M., Jr.; Kharbanda, K.K. Alcoholic Liver Disease: Pathogenesis and Current Management. Alcohol Res. 2017, 38, 147–161. [Google Scholar] [PubMed]
- Yuan, X.; Duan, S.Z.; Cao, J.; Gao, N.; Xu, J.; Zhang, L. Noninvasive inflammatory markers for assessing liver fibrosis stage in autoimmune hepatitis patients. Eur. J. Gastroenterol. Hepatol. 2019, 31, 1467–1474. [Google Scholar] [CrossRef] [PubMed]
- David, S.; Hamilton, J.P. Drug-induced Liver Injury. US Gastroenterol. Hepatol. Rev. 2010, 6, 73–80. [Google Scholar]
- Li, X.; Jin, Q.; Xu, H.; Zhang, Z.; Zhou, H.; Yan, D.; Li, D.; Gao, P.; Niu, J. Chronic hepatitis B patients with high liver fibrosis levels should receive antiviral treatment. Exp. Ther. Med. 2017, 13, 3624–3630. [Google Scholar] [CrossRef] [Green Version]
- Udompap, P.; Kim, W.R. Development of Hepatocellular Carcinoma in Patients With Suppressed Viral Replication: Changes in Risk Over Time. Clin. Liver Dis. 2020, 15, 85–90. [Google Scholar] [CrossRef] [PubMed]
- Cavalli, M.; Pan, G.; Nord, H.; Wallen Arzt, E.; Wallerman, O.; Wadelius, C. Genetic prevention of hepatitis C virus-induced liver fibrosis by allele-specific downregulation of MERTK. Hepatol. Res. 2017, 47, 826–830. [Google Scholar] [CrossRef]
- Chiang, D.J.; Pritchard, M.T.; Nagy, L.E. Obesity, diabetes mellitus, and liver fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G697–G702. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ratziu, V.; Giral, P.; Charlotte, F.; Bruckert, E.; Thibault, V.; Theodorou, I.; Khalil, L.; Turpin, G.; Opolon, P.; Poynard, T. Liver fibrosis in overweight patients. Gastroenterology 2000, 118, 1117–1123. [Google Scholar] [CrossRef]
- Baiocchini, A.; Montaldo, C.; Conigliaro, A.; Grimaldi, A.; Correani, V.; Mura, F.; Ciccosanti, F.; Rotiroti, N.; Brenna, A.; Montalbano, M.; et al. Extracellular Matrix Molecular Remodeling in Human Liver Fibrosis Evolution. PLoS ONE 2016, 11, e0151736. [Google Scholar] [CrossRef] [Green Version]
- Seo, W.; Jeong, W.I. Hepatic non-parenchymal cells: Master regulators of alcoholic liver disease? World J. Gastroenterol. 2016, 22, 1348–1356. [Google Scholar] [CrossRef]
- Canbay, A.; Friedman, S.; Gores, G.J. Apoptosis: The nexus of liver injury and fibrosis. Hepatology 2004, 39, 273–278. [Google Scholar] [CrossRef]
- Dixon, L.J.; Barnes, M.; Tang, H.; Pritchard, M.T.; Nagy, L.E. Kupffer cells in the liver. Compr. Physiol. 2013, 3, 785–797. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- van der Heide, D.; Weiskirchen, R.; Bansal, R. Therapeutic Targeting of Hepatic Macrophages for the Treatment of Liver Diseases. Front. Immunol. 2019, 10, 2852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ying, H.Z.; Chen, Q.; Zhang, W.Y.; Zhang, H.H.; Ma, Y.; Zhang, S.Z.; Fang, J.; Yu, C.H. PDGF signaling pathway in hepatic fibrosis pathogenesis and therapeutics (Review). Mol. Med. Rep. 2017, 16, 7879–7889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dewidar, B.; Meyer, C.; Dooley, S.; Meindl-Beinker, A.N. TGF-beta in Hepatic Stellate Cell Activation and Liver Fibrogenesis-Updated 2019. Cells 2019, 8, 1419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marcher, A.B.; Bendixen, S.M.; Terkelsen, M.K.; Hohmann, S.S.; Hansen, M.H.; Larsen, B.D.; Mandrup, S.; Dimke, H.; Detlefsen, S.; Ravnskjaer, K. Transcriptional regulation of Hepatic Stellate Cell activation in NASH. Sci. Rep. 2019, 9, 2324. [Google Scholar] [CrossRef] [Green Version]
- Holt, A.P.; Salmon, M.; Buckley, C.D.; Adams, D.H. Immune interactions in hepatic fibrosis. Clin. Liver Dis. 2008, 12, 861–882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robinson, M.W.; Harmon, C.; O’Farrelly, C. Liver immunology and its role in inflammation and homeostasis. Cell Mol. Immunol. 2016, 13, 267–276. [Google Scholar] [CrossRef] [Green Version]
- Tanwar, S.; Rhodes, F.; Srivastava, A.; Trembling, P.M.; Rosenberg, W.M. Inflammation and fibrosis in chronic liver diseases including non-alcoholic fatty liver disease and hepatitis C. World J. Gastroenterol. 2020, 26, 109–133. [Google Scholar] [CrossRef]
- Wells, R.G. Cellular sources of extracellular matrix in hepatic fibrosis. Clin. Liver Dis. 2008, 12, 759–768. [Google Scholar] [CrossRef] [Green Version]
- Tan, A.; Koh, S.; Bertoletti, A. Immune Response in Hepatitis B Virus Infection. Cold Spring Harb. Perspect. Med. 2015, 5, a021428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luan, J.; Ju, D. Inflammasome: A Double-Edged Sword in Liver Diseases. Front. Immunol. 2018, 9, 2201. [Google Scholar] [CrossRef] [PubMed]
- Tao, Y.; Wang, N.; Qiu, T.; Sun, X. The Role of Autophagy and NLRP3 Inflammasome in Liver Fibrosis. Biomed. Res. Int. 2020, 2020, 7269150. [Google Scholar] [CrossRef] [PubMed]
- Inzaugarat, M.E.; Johnson, C.D.; Holtmann, T.M.; McGeough, M.D.; Trautwein, C.; Papouchado, B.G.; Schwabe, R.; Hoffman, H.M.; Wree, A.; Feldstein, A.E. NLR Family Pyrin Domain-Containing 3 Inflammasome Activation in Hepatic Stellate Cells Induces Liver Fibrosis in Mice. Hepatology 2019, 69, 845–859. [Google Scholar] [CrossRef] [Green Version]
- Wree, A.; Eguchi, A.; McGeough, M.D.; Pena, C.A.; Johnson, C.D.; Canbay, A.; Hoffman, H.M.; Feldstein, A.E. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014, 59, 898–910. [Google Scholar] [CrossRef] [Green Version]
- Shay, J.E.S.; Hamilton, J.P. Hepatic fibrosis: Avenues of investigation and clinical implications. Clin. Liver Dis. 2018, 11, 111–114. [Google Scholar] [CrossRef]
- Wang, D.; Zhang, P.; Zhang, M. Predictors for advanced liver fibrosis in chronic hepatitis B virus infection with persistently normal or mildly elevated alanine aminotransferase. Exp. Ther. Med. 2017, 14, 5363–5370. [Google Scholar] [CrossRef] [Green Version]
- Zeng, D.W.; Dong, J.; Liu, Y.R.; Jiang, J.J.; Zhu, Y.Y. Noninvasive models for assessment of liver fibrosis in patients with chronic hepatitis B virus infection. World J. Gastroenterol. 2016, 22, 6663–6672. [Google Scholar] [CrossRef]
- Mello, T.; Ceni, E.; Surrenti, C.; Galli, A. Alcohol induced hepatic fibrosis: Role of acetaldehyde. Mol. Aspects Med. 2008, 29, 17–21. [Google Scholar] [CrossRef]
- Liu, Y.; Brymora, J.; Zhang, H.; Smith, B.; Ramezani-Moghadam, M.; George, J.; Wang, J. Leptin and acetaldehyde synergistically promotes alphaSMA expression in hepatic stellate cells by an interleukin 6-dependent mechanism. Alcohol Clin. Exp. Res. 2011, 35, 921–928. [Google Scholar] [CrossRef]
- Seth, D.; Haber, P.S.; Syn, W.K.; Diehl, A.M.; Day, C.P. Pathogenesis of alcohol-induced liver disease: Classical concepts and recent advances. J. Gastroenterol. Hepatol. 2011, 26, 1089–1105. [Google Scholar] [CrossRef]
- Suh, Y.G.; Jeong, W.I. Hepatic stellate cells and innate immunity in alcoholic liver disease. World J. Gastroenterol. 2011, 17, 2543–2551. [Google Scholar] [CrossRef] [PubMed]
- Gao, B.; Seki, E.; Brenner, D.A.; Friedman, S.; Cohen, J.I.; Nagy, L.; Szabo, G.; Zakhari, S. Innate immunity in alcoholic liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 300, G516–G525. [Google Scholar] [CrossRef]
- Jagavelu, K.; Routray, C.; Shergill, U.; O’Hara, S.P.; Faubion, W.; Shah, V.H. Endothelial cell toll-like receptor 4 regulates fibrosis-associated angiogenesis in the liver. Hepatology 2010, 52, 590–601. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- M, I.; Singh, C.; Ganie, M.A.; Alsayari, K. NASH: The Hepatic injury of Metabolic syndrome: A brief update. Int. J. Health Sci. 2009, 3, 265–270. [Google Scholar]
- Ghaffari, S. Oxidative stress in the regulation of normal and neoplastic hematopoiesis. Antioxid. Redox. Signal. 2008, 10, 1923–1940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sircana, A.; Paschetta, E.; Saba, F.; Molinaro, F.; Musso, G. Recent Insight into the Role of Fibrosis in Nonalcoholic Steatohepatitis-Related Hepatocellular Carcinoma. Int. J. Mol. Sci. 2019, 20, 1745. [Google Scholar] [CrossRef] [Green Version]
- Yu, K.; Li, Q.; Shi, G.; Li, N. Involvement of epithelial-mesenchymal transition in liver fibrosis. Saudi J. Gastroenterol. 2018, 24, 5–11. [Google Scholar] [CrossRef]
- Liberal, R.; Grant, C.R. Cirrhosis and autoimmune liver disease: Current understanding. World J. Hepatol. 2016, 8, 1157–1168. [Google Scholar] [CrossRef]
- Chiang, J.Y.L. Linking long noncoding RNA to control bile acid signaling and cholestatic liver fibrosis. Hepatology 2017, 66, 1032–1035. [Google Scholar] [CrossRef]
- Glaser, S.S.; Gaudio, E.; Miller, T.; Alvaro, D.; Alpini, G. Cholangiocyte proliferation and liver fibrosis. Expert Rev. Mol. Med. 2009, 11, e7. [Google Scholar] [CrossRef] [PubMed]
- Friedman, S.L. Mechanisms of hepatic fibrogenesis. Gastroenterology 2008, 134, 1655–1669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cong, M.; Iwaisako, K.; Jiang, C.; Kisseleva, T. Cell signals influencing hepatic fibrosis. Int. J. Hepatol. 2012, 2012, 158547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arriazu, E.; Ruiz de Galarreta, M.; Cubero, F.J.; Varela-Rey, M.; Perez de Obanos, M.P.; Leung, T.M.; Lopategi, A.; Benedicto, A.; Abraham-Enachescu, I.; Nieto, N. Extracellular matrix and liver disease. Antioxid. Redox. Signal. 2014, 21, 1078–1097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Friedman, S.L. Hepatic stellate cells: Protean, multifunctional, and enigmatic cells of the liver. Physiol. Rev. 2008, 88, 125–172. [Google Scholar] [CrossRef] [PubMed]
- Canbay, A.; Taimr, P.; Torok, N.; Higuchi, H.; Friedman, S.; Gores, G.J. Apoptotic body engulfment by a human stellate cell line is profibrogenic. Lab. Investig. 2003, 83, 655–663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gandhi, C.R. Oxidative Stress and Hepatic Stellate Cells: A Paradoxical Relationship. Trends Cell Mol. Biol. 2012, 7, 1–10. [Google Scholar] [PubMed]
- Yang, C.; Zeisberg, M.; Mosterman, B.; Sudhakar, A.; Yerramalla, U.; Holthaus, K.; Xu, L.; Eng, F.; Afdhal, N.; Kalluri, R. Liver fibrosis: Insights into migration of hepatic stellate cells in response to extracellular matrix and growth factors. Gastroenterology 2003, 124, 147–159. [Google Scholar] [CrossRef] [PubMed]
- Weiskirchen, R.; Tacke, F. Cellular and molecular functions of hepatic stellate cells in inflammatory responses and liver immunology. Hepatobiliary Surg. Nutr. 2014, 3, 344–363. [Google Scholar] [CrossRef]
- Nagaraja, T.; Chen, L.; Balasubramanian, A.; Groopman, J.E.; Ghoshal, K.; Jacob, S.T.; Leask, A.; Brigstock, D.R.; Anand, A.R.; Ganju, R.K. Activation of the connective tissue growth factor (CTGF)-transforming growth factor beta 1 (TGF-beta 1) axis in hepatitis C virus-expressing hepatocytes. PLoS ONE 2012, 7, e46526. [Google Scholar] [CrossRef] [Green Version]
- Shin, J.Y.; Hur, W.; Wang, J.S.; Jang, J.W.; Kim, C.W.; Bae, S.H.; Jang, S.K.; Yang, S.H.; Sung, Y.C.; Kwon, O.J.; et al. HCV core protein promotes liver fibrogenesis via up-regulation of CTGF with TGF-beta1. Exp. Mol. Med. 2005, 37, 138–145. [Google Scholar] [CrossRef]
- Luedde, T.; Schwabe, R.F. NF-kappaB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 108–118. [Google Scholar] [CrossRef] [Green Version]
- Oakley, F.; Trim, N.; Constandinou, C.M.; Ye, W.; Gray, A.M.; Frantz, G.; Hillan, K.; Kendall, T.; Benyon, R.C.; Mann, D.A.; et al. Hepatocytes express nerve growth factor during liver injury: Evidence for paracrine regulation of hepatic stellate cell apoptosis. Am. J. Pathol. 2003, 163, 1849–1858. [Google Scholar] [CrossRef]
- Verrecchia, F.; Mauviel, A. Transforming growth factor-beta and fibrosis. World J. Gastroenterol. 2007, 13, 3056–3062. [Google Scholar] [CrossRef] [PubMed]
- Xu, F.; Liu, C.; Zhou, D.; Zhang, L. TGF-beta/SMAD Pathway and Its Regulation in Hepatic Fibrosis. J. Histochem. Cytochem. 2016, 64, 157–167. [Google Scholar] [CrossRef] [PubMed]
- DeLeve, L.D.; Maretti-Mira, A.C. Liver Sinusoidal Endothelial Cell: An Update. Semin. Liver Dis. 2017, 37, 377–387. [Google Scholar] [CrossRef]
- Shetty, S.; Lalor, P.F.; Adams, D.H. Liver sinusoidal endothelial cells - gatekeepers of hepatic immunity. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 555–567. [Google Scholar] [CrossRef] [PubMed]
- Xu, B.; Broome, U.; Uzunel, M.; Nava, S.; Ge, X.; Kumagai-Braesch, M.; Hultenby, K.; Christensson, B.; Ericzon, B.G.; Holgersson, J.; et al. Capillarization of hepatic sinusoid by liver endothelial cell-reactive autoantibodies in patients with cirrhosis and chronic hepatitis. Am. J. Pathol. 2003, 163, 1275–1289. [Google Scholar] [CrossRef] [Green Version]
- Miyao, M.; Kotani, H.; Ishida, T.; Kawai, C.; Manabe, S.; Abiru, H.; Tamaki, K. Pivotal role of liver sinusoidal endothelial cells in NAFLD/NASH progression. Lab. Investig. 2015, 95, 1130–1144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deleve, L.D.; Wang, X.; Guo, Y. Sinusoidal endothelial cells prevent rat stellate cell activation and promote reversion to quiescence. Hepatology 2008, 48, 920–930. [Google Scholar] [CrossRef] [PubMed]
- Bocca, C.; Novo, E.; Miglietta, A.; Parola, M. Angiogenesis and Fibrogenesis in Chronic Liver Diseases. Cell Mol. Gastroenterol. Hepatol. 2015, 1, 477–488. [Google Scholar] [CrossRef] [Green Version]
- Ding, B.S.; Cao, Z.; Lis, R.; Nolan, D.J.; Guo, P.; Simons, M.; Penfold, M.E.; Shido, K.; Rabbany, S.Y.; Rafii, S. Divergent angiocrine signals from vascular niche balance liver regeneration and fibrosis. Nature 2014, 505, 97–102. [Google Scholar] [CrossRef] [Green Version]
- Kolios, G.; Valatas, V.; Kouroumalis, E. Role of Kupffer cells in the pathogenesis of liver disease. World J. Gastroenterol. 2006, 12, 7413–7420. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; He, K.; Li, J.; Liu, Z.; Gong, J. The role of Kupffer cells in hepatic diseases. Mol. Immunol. 2017, 85, 222–229. [Google Scholar] [CrossRef]
- Liu, C.; Tao, Q.; Sun, M.; Wu, J.Z.; Yang, W.; Jian, P.; Peng, J.; Hu, Y.; Liu, P. Kupffer cells are associated with apoptosis, inflammation and fibrotic effects in hepatic fibrosis in rats. Lab. Investig. 2010, 90, 1805–1816. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tacke, F.; Zimmermann, H.W. Macrophage heterogeneity in liver injury and fibrosis. J. Hepatol. 2014, 60, 1090–1096. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, L.; He, S.; Mao, X.; Zhang, Y.; Cai, Y.; Li, S. Effect of Hepatic Macrophage Polarization and Apoptosis on Liver Ischemia and Reperfusion Injury During Liver Transplantation. Front. Immunol. 2020, 11, 1193. [Google Scholar] [CrossRef] [PubMed]
- Guillot, A.; Tacke, F. Liver Macrophages: Old Dogmas and New Insights. Hepatol. Commun. 2019, 3, 730–743. [Google Scholar] [CrossRef] [Green Version]
- Fabregat, I.; Caballero-Diaz, D. Transforming Growth Factor-beta-Induced Cell Plasticity in Liver Fibrosis and Hepatocarcinogenesis. Front. Oncol. 2018, 8, 357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kany, S.; Vollrath, J.T.; Relja, B. Cytokines in Inflammatory Disease. Int. J. Mol. Sci. 2019, 20, 6008. [Google Scholar] [CrossRef] [Green Version]
- Nguyen-Lefebvre, A.T.; Horuzsko, A. Kupffer Cell Metabolism and Function. J. Enzymol. Metab. 2015, 1, 101. [Google Scholar]
- Koyama, Y.; Brenner, D.A. Liver inflammation and fibrosis. J. Clin. Investig. 2017, 127, 55–64. [Google Scholar] [CrossRef] [PubMed]
- Heymann, F.; Tacke, F. Immunology in the liver--from homeostasis to disease. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 88–110. [Google Scholar] [CrossRef] [PubMed]
- Parola, M.; Pinzani, M. Liver fibrosis: Pathophysiology, pathogenetic targets and clinical issues. Mol. Aspects Med. 2019, 65, 37–55. [Google Scholar] [CrossRef] [PubMed]
- Geervliet, E.; Bansal, R. Matrix Metalloproteinases as Potential Biomarkers and Therapeutic Targets in Liver Diseases. Cells 2020, 9, 1212. [Google Scholar] [CrossRef]
- Bartneck, M.; Warzecha, K.T.; Tacke, F. Therapeutic targeting of liver inflammation and fibrosis by nanomedicine. Hepatobiliary Surg. Nutr. 2014, 3, 364–376. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Huang, X.; Werner, M.; Broering, R.; Ge, J.; Li, Y.; Liao, B.; Sun, J.; Peng, J.; Lu, M.; et al. Elevated Expression of Chemokine CXCL13 in Chronic Hepatitis B Patients Links to Immune Control during Antiviral Therapy. Front. Immunol. 2017, 8, 323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, B.M.; Liu, J.D.; Li, Y.H.; Li, J. Margatoxin mitigates CCl4induced hepatic fibrosis in mice via macrophage polarization, cytokine secretion and STAT signaling. Int. J. Mol. Med. 2020, 45, 103–114. [Google Scholar] [CrossRef] [Green Version]
- Wu, H.; Chen, G.; Wang, J.; Deng, M.; Yuan, F.; Gong, J. TIM-4 interference in Kupffer cells against CCL4-induced liver fibrosis by mediating Akt1/Mitophagy signalling pathway. Cell Prolif. 2020, 53, e12731. [Google Scholar] [CrossRef] [Green Version]
- Ji, H.; Liu, Y.; Zhang, Y.; Shen, X.D.; Gao, F.; Busuttil, R.W.; Kuchroo, V.K.; Kupiec-Weglinski, J.W. T-cell immunoglobulin and mucin domain 4 (TIM-4) signaling in innate immune-mediated liver ischemia-reperfusion injury. Hepatology 2014, 60, 2052–2064. [Google Scholar] [CrossRef] [Green Version]
- Chen, L.; Yao, X.; Yao, H.; Ji, Q.; Ding, G.; Liu, X. Exosomal miR-103-3p from LPS-activated THP-1 macrophage contributes to the activation of hepatic stellate cells. FASEB J. 2020, 34, 5178–5192. [Google Scholar] [CrossRef]
- Alzaid, F.; Lagadec, F.; Albuquerque, M.; Ballaire, R.; Orliaguet, L.; Hainault, I.; Blugeon, C.; Lemoine, S.; Lehuen, A.; Saliba, D.G.; et al. IRF5 governs liver macrophage activation that promotes hepatic fibrosis in mice and humans. JCI Insight. 2016, 1, e88689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Akcora, B.O.; Dathathri, E.; Ortiz-Perez, A.; Gabriel, A.V.; Storm, G.; Prakash, J.; Bansal, R. TG101348, a selective JAK2 antagonist, ameliorates hepatic fibrogenesis in vivo. FASEB J. 2019, 33, 9466–9475. [Google Scholar] [CrossRef] [PubMed]
- Kong, X.; Horiguchi, N.; Mori, M.; Gao, B. Cytokines and STATs in Liver Fibrosis. Front. Physiol. 2012, 3, 69. [Google Scholar] [CrossRef] [Green Version]
- Wang, Y.; Zhang, C. The Roles of Liver-Resident Lymphocytes in Liver Diseases. Front. Immunol. 2019, 10, 1582. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Park, O.; Gao, B. NKT cells in liver fibrosis: Controversies or complexities. J. Hepatol. 2011, 55, 1166. [Google Scholar] [CrossRef] [Green Version]
- Marrero, I.; Maricic, I.; Feldstein, A.E.; Loomba, R.; Schnabl, B.; Rivera-Nieves, J.; Eckmann, L.; Kumar, V. Complex Network of NKT Cell Subsets Controls Immune Homeostasis in Liver and Gut. Front. Immunol. 2018, 9, 2082. [Google Scholar] [CrossRef]
- Syn, W.K.; Oo, Y.H.; Pereira, T.A.; Karaca, G.F.; Jung, Y.; Omenetti, A.; Witek, R.P.; Choi, S.S.; Guy, C.D.; Fearing, C.M.; et al. Accumulation of natural killer T cells in progressive nonalcoholic fatty liver disease. Hepatology 2010, 51, 1998–2007. [Google Scholar] [CrossRef] [Green Version]
- Gonzalez-Polo, V.; Pucci-Molineris, M.; Cervera, V.; Gambaro, S.; Yantorno, S.E.; Descalzi, V.; Tiribelli, C.; Gondolesi, G.E.; Meier, D. Group 2 innate lymphoid cells exhibit progressively higher levels of activation during worsening of liver fibrosis. Ann. Hepatol. 2019, 18, 366–372. [Google Scholar] [CrossRef]
- McHedlidze, T.; Waldner, M.; Zopf, S.; Walker, J.; Rankin, A.L.; Schuchmann, M.; Voehringer, D.; McKenzie, A.N.; Neurath, M.F.; Pflanz, S.; et al. Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis. Immunity 2013, 39, 357–371. [Google Scholar] [CrossRef] [Green Version]
- Weiskirchen, R.; Tacke, F. Interleukin-33 in the pathogenesis of liver fibrosis: Alarming ILC2 and hepatic stellate cells. Cell Mol. Immunol. 2017, 14, 143–145. [Google Scholar] [CrossRef]
- Ochel, A.; Tiegs, G.; Neumann, K. Type 2 Innate Lymphoid Cells in Liver and Gut: From Current Knowledge to Future Perspectives. Int. J. Mol. Sci. 2019, 20. [Google Scholar] [CrossRef] [Green Version]
- Rak, G.D.; Osborne, L.C.; Siracusa, M.C.; Kim, B.S.; Wang, K.; Bayat, A.; Artis, D.; Volk, S.W. IL-33-Dependent Group 2 Innate Lymphoid Cells Promote Cutaneous Wound Healing. J. Investig. Dermatol. 2016, 136, 487–496. [Google Scholar] [CrossRef] [Green Version]
- Volarevic, V.; Mitrovic, M.; Milovanovic, M.; Zelen, I.; Nikolic, I.; Mitrovic, S.; Pejnovic, N.; Arsenijevic, N.; Lukic, M.L. Protective role of IL-33/ST2 axis in Con A-induced hepatitis. J. Hepatol. 2012, 56, 26–33. [Google Scholar] [CrossRef] [PubMed]
- Zenewicz, L.A.; Yancopoulos, G.D.; Valenzuela, D.M.; Murphy, A.J.; Karow, M.; Flavell, R.A. Interleukin-22 but not interleukin-17 provides protection to hepatocytes during acute liver inflammation. Immunity 2007, 27, 647–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Paquissi, F.C. Immunity and Fibrogenesis: The Role of Th17/IL-17 Axis in HBV and HCV-induced Chronic Hepatitis and Progression to Cirrhosis. Front. Immunol. 2017, 8, 1195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.W.; Mi, S.; Li, Z.; Zhou, J.C.; Xie, J.; Hua, F.; Li, K.; Cui, B.; Lv, X.X.; Yu, J.J.; et al. Antagonism of Interleukin-17A ameliorates experimental hepatic fibrosis by restoring the IL-10/STAT3-suppressed autophagy in hepatocytes. Oncotarget 2017, 8, 9922–9934. [Google Scholar] [CrossRef] [PubMed]
- Liu, M.; Hu, Y.; Yuan, Y.; Tian, Z.; Zhang, C. gammadeltaT Cells Suppress Liver Fibrosis via Strong Cytolysis and Enhanced NK Cell-Mediated Cytotoxicity against Hepatic Stellate Cells. Front. Immunol. 2019, 10, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meng, F.; Wang, K.; Aoyama, T.; Grivennikov, S.I.; Paik, Y.; Scholten, D.; Cong, M.; Iwaisako, K.; Liu, X.; Zhang, M.; et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 2012, 143, 765–776.e763. [Google Scholar] [CrossRef] [Green Version]
- Deng, Z.B.; Liu, Y.; Liu, C.; Xiang, X.; Wang, J.; Cheng, Z.; Shah, S.V.; Zhang, S.; Zhang, L.; Zhuang, X.; et al. Immature myeloid cells induced by a high-fat diet contribute to liver inflammation. Hepatology 2009, 50, 1412–1420. [Google Scholar] [CrossRef] [Green Version]
- Nojima, H.; Freeman, C.M.; Schuster, R.M.; Japtok, L.; Kleuser, B.; Edwards, M.J.; Gulbins, E.; Lentsch, A.B. Hepatocyte exosomes mediate liver repair and regeneration via sphingosine-1-phosphate. J. Hepatol. 2016, 64, 60–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wan, L.; Xia, T.; Du, Y.; Liu, J.; Xie, Y.; Zhang, Y.; Guan, F.; Wu, J.; Wang, X.; Shi, C. Exosomes from activated hepatic stellate cells contain GLUT1 and PKM2: A role for exosomes in metabolic switch of liver nonparenchymal cells. FASEB J. 2019, 33, 8530–8542. [Google Scholar] [CrossRef] [PubMed]
- Seo, W.; Eun, H.S.; Kim, S.Y.; Yi, H.S.; Lee, Y.S.; Park, S.H.; Jang, M.J.; Jo, E.; Kim, S.C.; Han, Y.M.; et al. Exosome-mediated activation of toll-like receptor 3 in stellate cells stimulates interleukin-17 production by gammadelta T cells in liver fibrosis. Hepatology 2016, 64, 616–631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.S.; Kim, S.Y.; Ko, E.; Lee, J.H.; Yi, H.S.; Yoo, Y.J.; Je, J.; Suh, S.J.; Jung, Y.K.; Kim, J.H.; et al. Exosomes derived from palmitic acid-treated hepatocytes induce fibrotic activation of hepatic stellate cells. Sci. Rep. 2017, 7, 3710. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Yan, Y.; Wang, B.; Qian, H.; Zhang, X.; Shen, L.; Wang, M.; Zhou, Y.; Zhu, W.; Li, W.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate liver fibrosis. Stem Cells Dev. 2013, 22, 845–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, L.; Chen, R.; Velazquez, V.M.; Brigstock, D.R. Fibrogenic Signaling Is Suppressed in Hepatic Stellate Cells through Targeting of Connective Tissue Growth Factor (CCN2) by Cellular or Exosomal MicroRNA-199a-5p. Am. J. Pathol. 2016, 186, 2921–2933. [Google Scholar] [CrossRef] [Green Version]
- Giugliano, S.; Kriss, M.; Golden-Mason, L.; Dobrinskikh, E.; Stone, A.E.; Soto-Gutierrez, A.; Mitchell, A.; Khetani, S.R.; Yamane, D.; Stoddard, M.; et al. Hepatitis C virus infection induces autocrine interferon signaling by human liver endothelial cells and release of exosomes, which inhibits viral replication. Gastroenterology 2015, 148, 392–402. [Google Scholar] [CrossRef] [Green Version]
- Gregory, C.D.; Devitt, A. The macrophage and the apoptotic cell: An innate immune interaction viewed simplistically? Immunology 2004, 113, 1–14. [Google Scholar] [CrossRef]
- Canbay, A.; Feldstein, A.E.; Higuchi, H.; Werneburg, N.; Grambihler, A.; Bronk, S.F.; Gores, G.J. Kupffer cell engulfment of apoptotic bodies stimulates death ligand and cytokine expression. Hepatology 2003, 38, 1188–1198. [Google Scholar] [CrossRef]
- Canbay, A.; Higuchi, H.; Bronk, S.F.; Taniai, M.; Sebo, T.J.; Gores, G.J. Fas enhances fibrogenesis in the bile duct ligated mouse: A link between apoptosis and fibrosis. Gastroenterology 2002, 123, 1323–1330. [Google Scholar] [CrossRef]
- Canbay, A.; Guicciardi, M.E.; Higuchi, H.; Feldstein, A.; Bronk, S.F.; Rydzewski, R.; Taniai, M.; Gores, G.J. Cathepsin B inactivation attenuates hepatic injury and fibrosis during cholestasis. J. Clin. Investig. 2003, 112, 152–159. [Google Scholar] [CrossRef] [Green Version]
- Takehara, T.; Tatsumi, T.; Suzuki, T.; Rucker, E.B., 3rd; Hennighausen, L.; Jinushi, M.; Miyagi, T.; Kanazawa, Y.; Hayashi, N. Hepatocyte-specific disruption of Bcl-xL leads to continuous hepatocyte apoptosis and liver fibrotic responses. Gastroenterology 2004, 127, 1189–1197. [Google Scholar] [CrossRef] [PubMed]
- Halder, L.D.; Jo, E.A.H.; Hasan, M.Z.; Ferreira-Gomes, M.; Kruger, T.; Westermann, M.; Palme, D.I.; Rambach, G.; Beyersdorf, N.; Speth, C.; et al. Immune modulation by complement receptor 3-dependent human monocyte TGF-beta1-transporting vesicles. Nat. Commun. 2020, 11, 2331. [Google Scholar] [CrossRef] [PubMed]
- Zhan, S.S.; Jiang, J.X.; Wu, J.; Halsted, C.; Friedman, S.L.; Zern, M.A.; Torok, N.J. Phagocytosis of apoptotic bodies by hepatic stellate cells induces NADPH oxidase and is associated with liver fibrosis in vivo. Hepatology 2006, 43, 435–443. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Inflammasome activation and regulation: Toward a better understanding of complex mechanisms. Cell Discov. 2020, 6, 36. [Google Scholar] [CrossRef] [PubMed]
- Mridha, A.R.; Wree, A.; Robertson, A.A.B.; Yeh, M.M.; Johnson, C.D.; Van Rooyen, D.M.; Haczeyni, F.; Teoh, N.C.; Savard, C.; Ioannou, G.N.; et al. NLRP3 inflammasome blockade reduces liver inflammation and fibrosis in experimental NASH in mice. J. Hepatol. 2017, 66, 1037–1046. [Google Scholar] [CrossRef]
- Zhang, W.J.; Fang, Z.M.; Liu, W.Q. NLRP3 inflammasome activation from Kupffer cells is involved in liver fibrosis of Schistosoma japonicum-infected mice via NF-kappaB. Parasit Vectors 2019, 12, 29. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Shan, S.; Kang, K.; Zhang, C.; Kou, R.; Song, F. The cross-talk of NLRP3 inflammasome activation and necroptotic hepatocyte death in acetaminophen-induced mice acute liver injury. Hum. Exp. Toxicol. 2021, 40, 673–684. [Google Scholar] [CrossRef]
- Wu, J.; Lin, S.; Wan, B.; Velani, B.; Zhu, Y. Pyroptosis in Liver Disease: New Insights into Disease Mechanisms. Aging Dis. 2019, 10, 1094–1108. [Google Scholar] [CrossRef] [Green Version]
- Wan, X.; Xu, C.; Yu, C.; Li, Y. Role of NLRP3 Inflammasome in the Progression of NAFLD to NASH. Can. J. Gastroenterol. Hepatol. 2016, 2016, 6489012. [Google Scholar] [CrossRef] [Green Version]
- Latz, E.; Xiao, T.S.; Stutz, A. Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 2013, 13, 397–411. [Google Scholar] [CrossRef]
- Dixon, L.J.; Flask, C.A.; Papouchado, B.G.; Feldstein, A.E.; Nagy, L.E. Caspase-1 as a central regulator of high fat diet-induced non-alcoholic steatohepatitis. PLoS ONE 2013, 8, e56100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, X.; Dong, L.; Lin, X.; Li, J. Relevance of the NLRP3 Inflammasome in the Pathogenesis of Chronic Liver Disease. Front. Immunol. 2017, 8, 1728. [Google Scholar] [CrossRef] [Green Version]
- Lan, T.; Kisseleva, T.; Brenner, D.A. Deficiency of NOX1 or NOX4 Prevents Liver Inflammation and Fibrosis in Mice through Inhibition of Hepatic Stellate Cell Activation. PLoS ONE 2015, 10, e0129743. [Google Scholar] [CrossRef]
- Ma, L.; Yang, X.; Wei, R.; Ye, T.; Zhou, J.K.; Wen, M.; Men, R.; Li, P.; Dong, B.; Liu, L.; et al. MicroRNA-214 promotes hepatic stellate cell activation and liver fibrosis by suppressing Sufu expression. Cell Death Dis. 2018, 9, 718. [Google Scholar] [CrossRef] [PubMed]
- Jiang, X.; Tsitsiou, E.; Herrick, S.E.; Lindsay, M.A. MicroRNAs and the regulation of fibrosis. FEBS J. 2010, 277, 2015–2021. [Google Scholar] [CrossRef]
- Jiang, X.P.; Ai, W.B.; Wan, L.Y.; Zhang, Y.Q.; Wu, J.F. The roles of microRNA families in hepatic fibrosis. Cell Biosci. 2017, 7, 34. [Google Scholar] [CrossRef] [Green Version]
- Zhu, B.; Wei, X.X.; Wang, T.B.; Zhou, Y.C.; Liu, A.M.; Zhang, G.W. Increased miR-16 expression induced by hepatitis C virus infection promotes liver fibrosis through downregulation of hepatocyte growth factor and Smad7. Arch. Virol. 2015, 160, 2043–2050. [Google Scholar] [CrossRef]
- Hyun, J.; Choi, S.S.; Diehl, A.M.; Jung, Y. Potential role of Hedgehog signaling and microRNA-29 in liver fibrosis of IKKbeta-deficient mouse. J. Mol. Histol. 2014, 45, 103–112. [Google Scholar] [CrossRef]
- Wan, Y.; McDaniel, K.; Wu, N.; Ramos-Lorenzo, S.; Glaser, T.; Venter, J.; Francis, H.; Kennedy, L.; Sato, K.; Zhou, T.; et al. Regulation of Cellular Senescence by miR-34a in Alcoholic Liver Injury. Am. J. Pathol. 2017, 187, 2788–2798. [Google Scholar] [CrossRef] [Green Version]
- Tian, X.F.; Ji, F.J.; Zang, H.L.; Cao, H. Activation of the miR-34a/SIRT1/p53 Signaling Pathway Contributes to the Progress of Liver Fibrosis via Inducing Apoptosis in Hepatocytes but Not in HSCs. PLoS ONE 2016, 11, e0158657. [Google Scholar] [CrossRef]
- Zhao, J.; Tang, N.; Wu, K.; Dai, W.; Ye, C.; Shi, J.; Zhang, J.; Ning, B.; Zeng, X.; Lin, Y. MiR-21 simultaneously regulates ERK1 signaling in HSC activation and hepatocyte EMT in hepatic fibrosis. PLoS ONE 2014, 9, e108005. [Google Scholar] [CrossRef] [Green Version]
- Rodrigues, P.M.; Rodrigues, C.M.P.; Castro, R.E. Modulation of liver steatosis by miR-21/PPARalpha. Cell Death Discov. 2018, 4, 9. [Google Scholar] [CrossRef]
- Kogure, T.; Costinean, S.; Yan, I.; Braconi, C.; Croce, C.; Patel, T. Hepatic miR-29ab1 expression modulates chronic hepatic injury. J. Cell Mol. Med. 2012, 16, 2647–2654. [Google Scholar] [CrossRef]
- Zhang, T.; Yang, Z.; Kusumanchi, P.; Han, S.; Liangpunsakul, S. Critical Role of microRNA-21 in the Pathogenesis of Liver Diseases. Front. Med. 2020, 7, 7. [Google Scholar] [CrossRef] [Green Version]
- Dong, Z.; Li, S.; Wang, X.; Si, L.; Ma, R.; Bao, L.; Bo, A. lncRNA GAS5 restrains CCl4-induced hepatic fibrosis by targeting miR-23a through the PTEN/PI3K/Akt signaling pathway. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 316, G539–G550. [Google Scholar] [CrossRef]
- Li, Q.; Li, Z.; Lin, Y.; Che, H.; Hu, Y.; Kang, X.; Zhang, Y.; Wang, L. High glucose promotes hepatic fibrosis via miR32/MTA3mediated epithelialtomesenchymal transition. Mol. Med. Rep. 2019, 19, 3190–3200. [Google Scholar] [CrossRef]
- Meng, F.; Glaser, S.S.; Francis, H.; Yang, F.; Han, Y.; Stokes, A.; Staloch, D.; McCarra, J.; Liu, J.; Venter, J.; et al. Epigenetic regulation of miR-34a expression in alcoholic liver injury. Am. J. Pathol. 2012, 181, 804–817. [Google Scholar] [CrossRef] [Green Version]
- Zheng, J.; Wu, C.; Xu, Z.; Xia, P.; Dong, P.; Chen, B.; Yu, F. Hepatic stellate cell is activated by microRNA-181b via PTEN/Akt pathway. Mol. Cell Biochem. 2015, 398, 1–9. [Google Scholar] [CrossRef]
- Gupta, P.; Sata, T.N.; Yadav, A.K.; Mishra, A.; Vats, N.; Hossain, M.M.; Sanal, M.G.; Venugopal, S.K. TGF-beta induces liver fibrosis via miRNA-181a-mediated down regulation of augmenter of liver regeneration in hepatic stellate cells. PLoS ONE 2019, 14, e0214534. [Google Scholar] [CrossRef]
- Wu, J.C.; Chen, R.; Luo, X.; Li, Z.H.; Luo, S.Z.; Xu, M.Y. MicroRNA-194 inactivates hepatic stellate cells and alleviates liver fibrosis by inhibiting AKT2. World J. Gastroenterol. 2019, 25, 4468–4480. [Google Scholar] [CrossRef]
- Venugopal, S.K.; Jiang, J.; Kim, T.H.; Li, Y.; Wang, S.S.; Torok, N.J.; Wu, J.; Zern, M.A. Liver fibrosis causes downregulation of miRNA-150 and miRNA-194 in hepatic stellate cells, and their overexpression causes decreased stellate cell activation. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G101–G106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murakami, Y.; Toyoda, H.; Tanaka, M.; Kuroda, M.; Harada, Y.; Matsuda, F.; Tajima, A.; Kosaka, N.; Ochiya, T.; Shimotohno, K. The progression of liver fibrosis is related with overexpression of the miR-199 and 200 families. PLoS ONE 2011, 6, e16081. [Google Scholar] [CrossRef]
- Okada, H.; Honda, M.; Campbell, J.S.; Takegoshi, K.; Sakai, Y.; Yamashita, T.; Shirasaki, T.; Takabatake, R.; Nakamura, M.; Tanaka, T.; et al. Inhibition of microRNA-214 ameliorates hepatic fibrosis and tumor incidence in platelet-derived growth factor C transgenic mice. Cancer Sci. 2015, 106, 1143–1152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iizuka, M.; Ogawa, T.; Enomoto, M.; Motoyama, H.; Yoshizato, K.; Ikeda, K.; Kawada, N. Induction of microRNA-214-5p in human and rodent liver fibrosis. Fibrogenesis Tissue Repair 2012, 5, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ji, D.; Chen, G.F.; Wang, J.C.; Ji, S.H.; Wu, X.W.; Lu, X.J.; Chen, J.L.; Li, J.T. Hsa_circ_0070963 inhibits liver fibrosis via regulation of miR-223-3p and LEMD3. Aging 2020, 12, 1643–1655. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Wang, Y.; Quan, J. Exosomal miR-223 derived from natural killer cells inhibits hepatic stellate cell activation by suppressing autophagy. Mol. Med. 2020, 26, 81. [Google Scholar] [CrossRef]
- Zhang, T.; Hu, J.; Wang, X.; Zhao, X.; Li, Z.; Niu, J.; Steer, C.J.; Zheng, G.; Song, G. MicroRNA-378 promotes hepatic inflammation and fibrosis via modulation of the NF-kappaB-TNFalpha pathway. J. Hepatol. 2019, 70, 87–96. [Google Scholar] [CrossRef]
- Hyun, J.; Wang, S.; Kim, J.; Rao, K.M.; Park, S.Y.; Chung, I.; Ha, C.S.; Kim, S.W.; Yun, Y.H.; Jung, Y. MicroRNA-378 limits activation of hepatic stellate cells and liver fibrosis by suppressing Gli3 expression. Nat. Commun. 2016, 7, 10993. [Google Scholar] [CrossRef] [Green Version]
- Ji, F.; Wang, K.; Zhang, Y.; Mao, X.L.; Huang, Q.; Wang, J.; Ye, L.; Li, Y. MiR-542-3p controls hepatic stellate cell activation and fibrosis via targeting BMP-7. J. Cell Biochem. 2019, 120, 4573–4581. [Google Scholar] [CrossRef]
- Ezhilarasan, D. MicroRNA interplay between hepatic stellate cell quiescence and activation. Eur. J. Pharmacol. 2020, 885, 173507. [Google Scholar] [CrossRef] [PubMed]
- Calvaruso, V.; Craxi, A. Regression of fibrosis after HBV antiviral therapy. Is cirrhosis reversible? Liver Int. 2014, 34 Suppl 1, 85–90. [Google Scholar] [CrossRef] [Green Version]
- Damiris, K.; Tafesh, Z.H.; Pyrsopoulos, N. Efficacy and safety of anti-hepatic fibrosis drugs. World J. Gastroenterol. 2020, 26, 6304–6321. [Google Scholar] [CrossRef] [PubMed]
- Glass, L.M.; Dickson, R.C.; Anderson, J.C.; Suriawinata, A.A.; Putra, J.; Berk, B.S.; Toor, A. Total body weight loss of >/= 10% is associated with improved hepatic fibrosis in patients with nonalcoholic steatohepatitis. Dig. Dis. Sci. 2015, 60, 1024–1030. [Google Scholar] [CrossRef]
- Yamada, K.; Mizukoshi, E.; Seike, T.; Horii, R.; Kitahara, M.; Sunagozaka, H.; Arai, K.; Yamashita, T.; Honda, M.; Kaneko, S. Light alcohol consumption has the potential to suppress hepatocellular injury and liver fibrosis in non-alcoholic fatty liver disease. PLoS ONE 2018, 13, e0191026. [Google Scholar] [CrossRef] [Green Version]
- Czaja, A.J. Hepatic inflammation and progressive liver fibrosis in chronic liver disease. World J. Gastroenterol. 2014, 20, 2515–2532. [Google Scholar] [CrossRef]
- Cariello, M.; Piccinin, E.; Moschetta, A. Transcriptional Regulation of Metabolic Pathways via Lipid-Sensing Nuclear Receptors PPARs, FXR, and LXR in NASH. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 1519–1539. [Google Scholar] [CrossRef]
- Li, J.; Kuruba, R.; Wilson, A.; Gao, X.; Zhang, Y.; Li, S. Inhibition of endothelin-1-mediated contraction of hepatic stellate cells by FXR ligand. PLoS ONE 2010, 5, e13955. [Google Scholar] [CrossRef] [Green Version]
- Younossi, Z.M.; Ratziu, V.; Loomba, R.; Rinella, M.; Anstee, Q.M.; Goodman, Z.; Bedossa, P.; Geier, A.; Beckebaum, S.; Newsome, P.N.; et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: Interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2019, 394, 2184–2196. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Q.; Xiang, S.; Liu, Q.; Gu, T.; Yao, Y.; Lu, X. PPARgamma Antagonizes Hypoxia-Induced Activation of Hepatic Stellate Cell through Cross Mediating PI3K/AKT and cGMP/PKG Signaling. PPAR Res. 2018, 2018, 6970407. [Google Scholar] [CrossRef] [Green Version]
- Sven, M.F.; Pierre, B.; Manal, F.A.; Quentin, M.A.; Elisabetta, B.; Vlad, R.; Philippe, H.M.; Bruno, S.; Jean-Louis, J.; Jean-Louis, A. A randomised, double-blind, placebo-controlled, multi-centre, dose-range, proof-of-concept, 24-week treatment study of lanifibranor in adult subjects with non-alcoholic steatohepatitis: Design of the NATIVE study. Contemp. Clin. Trials. 2020, 98, 106170. [Google Scholar] [CrossRef] [PubMed]
- Poilil Surendran, S.; George Thomas, R.; Moon, M.J.; Jeong, Y.Y. Nanoparticles for the treatment of liver fibrosis. Int. J. Nanomed. 2017, 12, 6997–7006. [Google Scholar] [CrossRef] [Green Version]
- Torchilin, V.P. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat. Rev. Drug Discov. 2014, 13, 813–827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, X.; Murphy, F.R.; Gehdu, N.; Zhang, J.; Iredale, J.P.; Benyon, R.C. Engagement of alphavbeta3 integrin regulates proliferation and apoptosis of hepatic stellate cells. J. Biol. Chem. 2004, 279, 23996–24006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Senoo, H. Structure and function of hepatic stellate cells. Med. Electron. Microsc. 2004, 37, 3–15. [Google Scholar] [CrossRef] [PubMed]
- Kinnman, N.; Francoz, C.; Barbu, V.; Wendum, D.; Rey, C.; Hultcrantz, R.; Poupon, R.; Housset, C. The myofibroblastic conversion of peribiliary fibrogenic cells distinct from hepatic stellate cells is stimulated by platelet-derived growth factor during liver fibrogenesis. Lab. Investig. 2003, 83, 163–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, F.; Tuyama, A.; Lee, T.F.; Loke, J.; Agarwal, R.; Cheng, X.; Garg, A.; Fiel, M.I.; Schwartz, M.; Walewski, J.; et al. Hepatic stellate cells express functional CXCR4: Role in stromal cell-derived factor-1alpha-mediated stellate cell activation. Hepatology 2009, 49, 2055–2067. [Google Scholar] [CrossRef] [Green Version]
- Kikuchi, S.; Griffin, C.T.; Wang, S.S.; Bissell, D.M. Role of CD44 in epithelial wound repair: Migration of rat hepatic stellate cells utilizes hyaluronic acid and CD44v6. J. Biol. Chem. 2005, 280, 15398–15404. [Google Scholar] [CrossRef] [Green Version]
- Ullah, A.; Wang, K.; Wu, P.; Oupicky, D.; Sun, M. CXCR4-targeted liposomal mediated co-delivery of pirfenidone and AMD3100 for the treatment of TGFbeta-induced HSC-T6 cells activation. Int. J. Nanomed. 2019, 14, 2927–2944. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.N.; Hsu, S.L.; Liao, M.Y.; Liu, Y.T.; Lai, C.H.; Chen, J.F.; Nguyen, M.T.; Su, Y.H.; Chen, S.T.; Wu, L.C. Ameliorative Effect of Curcumin-Encapsulated Hyaluronic Acid-PLA Nanoparticles on Thioacetamide-Induced Murine Hepatic Fibrosis. Int. J. Environ. Res. Public Health 2016, 14, 11. [Google Scholar] [CrossRef] [Green Version]
- Sato, Y.; Murase, K.; Kato, J.; Kobune, M.; Sato, T.; Kawano, Y.; Takimoto, R.; Takada, K.; Miyanishi, K.; Matsunaga, T.; et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat. Biotechnol. 2008, 26, 431–442. [Google Scholar] [CrossRef]
- Chang, Y.; Li, H. Hepatic Antifibrotic Pharmacotherapy: Are We Approaching Success? J. Clin. Transl Hepatol. 2020, 8, 222–229. [Google Scholar] [CrossRef] [PubMed]
- Bansal, R.; Nagorniewicz, B.; Prakash, J. Clinical Advancements in the Targeted Therapies against Liver Fibrosis. Mediators. Inflamm. 2016, 2016, 7629724. [Google Scholar] [CrossRef]
- Satapathy, S.K.; Sakhuja, P.; Malhotra, V.; Sharma, B.C.; Sarin, S.K. Beneficial effects of pentoxifylline on hepatic steatosis, fibrosis and necroinflammation in patients with non-alcoholic steatohepatitis. J. Gastroenterol. Hepatol. 2007, 22, 634–638. [Google Scholar] [CrossRef]
- Wen, W.X.; Lee, S.Y.; Siang, R.; Koh, R.Y. Repurposing Pentoxifylline for the Treatment of Fibrosis: An Overview. Adv. Ther. 2017, 34, 1245–1269. [Google Scholar] [CrossRef] [PubMed]
- Simoes, E.S.A.C.; Miranda, A.S.; Rocha, N.P.; Teixeira, A.L. Renin angiotensin system in liver diseases: Friend or foe? World J. Gastroenterol. 2017, 23, 3396–3406. [Google Scholar] [CrossRef]
- Shim, K.Y.; Eom, Y.W.; Kim, M.Y.; Kang, S.H.; Baik, S.K. Role of the renin-angiotensin system in hepatic fibrosis and portal hypertension. Korean J. Intern. Med. 2018, 33, 453–461. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Gong, H.; Zhang, Z.T.; Wang, Y. Effect of angiotensin II and angiotensin II type 1 receptor antagonist on the proliferation, contraction and collagen synthesis in rat hepatic stellate cells. Chin. Med. J. 2008, 121, 161–165. [Google Scholar] [CrossRef]
- Bataller, R.; Gines, P.; Nicolas, J.M.; Gorbig, M.N.; Garcia-Ramallo, E.; Gasull, X.; Bosch, J.; Arroyo, V.; Rodes, J. Angiotensin II induces contraction and proliferation of human hepatic stellate cells. Gastroenterology 2000, 118, 1149–1156. [Google Scholar] [CrossRef]
- Salama, Z.A.; Sadek, A.; Abdelhady, A.M.; Darweesh, S.K.; Morsy, S.A.; Esmat, G. Losartan may inhibit the progression of liver fibrosis in chronic HCV patients. Hepatobiliary Surg. Nutr. 2016, 5, 249–255. [Google Scholar] [CrossRef] [Green Version]
- Marcellin, P.; Gane, E.; Buti, M.; Afdhal, N.; Sievert, W.; Jacobson, I.M.; Washington, M.K.; Germanidis, G.; Flaherty, J.F.; Aguilar Schall, R.; et al. Regression of cirrhosis during treatment with tenofovir disoproxil fumarate for chronic hepatitis B: A 5-year open-label follow-up study. Lancet 2013, 381, 468–475. [Google Scholar] [CrossRef]
miRNAs | Role in Hepatic Fibrosis | References |
---|---|---|
miR-15 family | Cell proliferation, apoptosis, suppression of hepatocyte growth factor, an inhibitor of TGF-β | [136,144] |
miR-21 | Collagen synthesis and deposition, induction of TGF-β and α-SMA, HSC activation | [142,145] |
miR-23a | Activation of PTEN/PI3K/Akt signaling pathway | [146] |
miR-29 family | Activation of fibrosis-inducing pathways including TGF-β, NF-κB, PI3K/AKT signaling, induction of ECM related genes, inhibit HSC activation | [138,139] |
miR-32 | Promote epithelial to mesenchymal transition | [147] |
miR-34 family | HSC activation, deposition of ECM proteins, upregulation of MMPs | [140,148] |
miR-181 | Inhibit Augmenter of liver regeneration, promote epithelial mesenchymal transition, HSC activation | [149,150] |
miR-194 | Inactivate HSCs, inhibit α-SMA and type 1 collagen | [151,152] |
miR-199 and miR-200 | ECM deposition, production of pro-fibrotic cytokines | [137,153] |
miR-214 | HSC activation, ECM accumulation, induction of pro-fibrotic genes | [154,155] |
miR-223-3p | HSC activation | [156,157] |
miR-378 | Induction of NF-κB and TNF-α, inflammation, inhibition of HSC activation | [158,159] |
miR-542-3p | Inhibit HSC activation | [160,161] |
Drug | Target | Phase | Trial Number |
---|---|---|---|
NASH | |||
Tropifexor | FXR agonist | II | NCT03517540 |
Tropifexor | FXR agonist | II | NCT04065841 |
Cilofexor | FXR agonist | II | NCT02854605 |
Obeticholic acid | FXR agonist | III | NCT02548351 |
Cenicriviroc | Antagonist for CCR2 and 5 | II | NCT02217475 |
GR-MD-02 | Galectin-3 inhibitor | II | NCT024662967 |
GR-MD-02 | Galectin-3 inhibitor | I | NCT01899859 |
BMS986036 | FGF21 analogs | II | NCT02413372 |
BMS986036 | FGF21 analogs | II | NCT03486912 |
BMS986036 | FGF21 analogs | II | NCT03486899 |
NGM282 | FGF19 analogs | II | NCT02443116 |
JKB-122 | TLR4 antagonist | II | NCT04255069 |
Lanifibranor | PPAR agonist | III | NCT04849728 |
GS-4997 | Apoptosis signal-regulating kinase | II | NCT02466516 |
Emricasan | Caspase inhibitor | II | NCT02686762 |
MGL-3196 | Thyroid hormone receptor agonist | III | NCT03900429 |
CC-90001 | Mitogen activated protein kinase-8 | II | NCT04048876 |
Nitazoxanide | Collagen turnover | II | NCT03656068 |
Selonsertib Firsocostat Cilofexor and combinations | Apoptosis signal-regulating kinase Liver-directed acetyl-CoA carboxylase inhibitor, FXR target | II | NCT03449446 |
HCV and HCV/HIV | |||
Candesartan and ramipril | Angiotensin receptor blocker and angiotensin converting enzyme inhibitor | III | NCT03770936 |
Pirfenidone | Inhibitor of TGF-β | II | NCT02161952 |
Simtuzumab | LOXL2 antibody | II | NCT01707472 |
Ursodeoxycholic acid Silymarin, antioxidants and colchicine | Bile duct, Inhibition of lipid peroxidation, oxidative stress, immunomodulatory effect | N/A | NCT03568578 |
Raltegravir | Integrase inhibitor | II | NCT01231685 |
Prazosin | Alpha-adrenergic antagonist | II | NCT00148837 |
Rifaximin | Endotoxin | NCT01603108 | |
Warfarin | Anticoagulation | II | NCT00180674 |
Losartan | Angiotensin II type 1 (AT1) receptors antagonists | IV | NCT002298714 |
CHB | |||
Hydronidone | Inhibitor of TGF-β | II | NCT02499562 |
Nitazoxanide | Collagen turnover | II | NCT03905655 |
ALD | |||
Profermin | Dysbiotic microbiota | N/A | NCT03863730 |
Ciprofloxacin | Bacterial DNA topoisomerase and DNA-gyrase | I | NCT02326103 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Khanam, A.; Saleeb, P.G.; Kottilil, S. Pathophysiology and Treatment Options for Hepatic Fibrosis: Can It Be Completely Cured? Cells 2021, 10, 1097. https://doi.org/10.3390/cells10051097
Khanam A, Saleeb PG, Kottilil S. Pathophysiology and Treatment Options for Hepatic Fibrosis: Can It Be Completely Cured? Cells. 2021; 10(5):1097. https://doi.org/10.3390/cells10051097
Chicago/Turabian StyleKhanam, Arshi, Paul G. Saleeb, and Shyam Kottilil. 2021. "Pathophysiology and Treatment Options for Hepatic Fibrosis: Can It Be Completely Cured?" Cells 10, no. 5: 1097. https://doi.org/10.3390/cells10051097