Review Article

天然产物在肝纤维化治疗中的应用:5年回顾

卷 31, 期 31, 2024

发表于: 14 February, 2024

页: [5061 - 5082] 页: 22

弟呕挨: 10.2174/0109298673288458240203064112

价格: $65

摘要

肝纤维化以肝组织内细胞外基质蛋白的过量产生为特征,引起了日益严重的全球健康问题。然而,目前还没有获得批准的抗纤维化药物,这凸显了了解肝纤维化分子机制的迫切需要。这些知识不仅可以帮助开发治疗方法,还可以进行早期干预,增强疾病预测,并提高我们对各种潜在疾病与肝脏之间相互作用的理解。值得注意的是,在世界各地的传统医学系统中使用的天然产物,显示出多种生化和药理活性,越来越多地认识到它们在治疗肝纤维化方面的潜力。本文旨在全面了解肝纤维化,重点介绍近五年来天然产物抗纤维化潜力的分子机制和研究进展。它也承认它们在发展中的挑战,并寻求强调它们在提高患者预后和减少全球肝病负担方面的潜力。

关键词: 肝纤维化,肝星状细胞,病因,抗纤维化,分子机制,天然产物,免疫细胞。

[1]
Berumen, J.; Baglieri, J.; Kisseleva, T.; Mekeel, K. Liver fibrosis: Pathophysiology and clinical implications. WIREs Mech. Dis., 2021, 13(1), e1499.
[http://dx.doi.org/10.1002/wsbm.1499] [PMID: 32713091]
[2]
Kim, D.; Li, A.A.; Perumpail, B.J.; Gadiparthi, C.; Kim, W.; Cholankeril, G.; Glenn, J.S.; Harrison, S.A.; Younossi, Z.M.; Ahmed, A. Changing trends in etiology-based and ethnicity-based annual mortality rates of cirrhosis and hepatocellular carcinoma in the United States. Hepatology, 2019, 69(3), 1064-1074.
[http://dx.doi.org/10.1002/hep.30161] [PMID: 30014489]
[3]
Ciardullo, S.; Monti, T.; Perseghin, G. High prevalence of advanced liver fibrosis assessed by transient elastography among U.S. adults with type 2 diabetes. Diabetes Care, 2021, 44(2), 519-525.
[http://dx.doi.org/10.2337/dc20-1778] [PMID: 33303638]
[4]
Seki, E.; Brenner, D.A. Recent advancement of molecular mechanisms of liver fibrosis. J. Hepatobiliary Pancreat. Sci., 2015, 22(7), 512-518.
[http://dx.doi.org/10.1002/jhbp.245] [PMID: 25869468]
[5]
Demir, Y; Ceylan, H; Turkes, C; Beydemir, S Molecular docking and inhibition studies of vulpinic, carnosic and usnic acids on polyol pathway enzymes. J. Biomol. Struct. Dyn., 2022, 40(22), 12008-12021.
[6]
Yamali, C; Gul, HI; Cakir, T; Demir, Y; Gulcin, I Aminoalkylated phenolic chalcones: Investigation of biological effects on acetylcholinesterase and carbonic anhydrase I and II as potential lead enzyme inhibitors. Lett. Drug Design Discov., 2020, 17(10), 1283-1292.
[7]
Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J. Nat. Prod., 2020, 83(3), 770-803.
[http://dx.doi.org/10.1021/acs.jnatprod.9b01285] [PMID: 32162523]
[8]
Zhang, H.W.; Lv, C.; Zhang, L.J.; Guo, X.; Shen, Y.W.; Nagle, D.G.; Zhou, Y.D.; Liu, S.H.; Zhang, W.D.; Luan, X. Application of omics- and multi-omics-based techniques for natural product target discovery. Biomed. Pharmacother., 2021, 141, 111833.
[http://dx.doi.org/10.1016/j.biopha.2021.111833] [PMID: 34175822]
[9]
Bayrak, S.; Öztürk, C.; Demir, Y.; Alım, Z.; Küfrevioglu, Ö.İ. Purification of polyphenol oxidase from potato and investigation of the inhibitory effects of phenolic acids on enzyme activity. Protein Pept. Lett., 2020, 27(3), 187-192.
[http://dx.doi.org/10.2174/0929866526666191002142301] [PMID: 31577197]
[10]
Palabıyık, E.; Sulumer, A.N.; Uguz, H.; Avcı, B.; Askın, S.; Askın, H.; Demir, Y. Assessment of hypolipidemic and anti-inflammatory properties of walnut ( Juglans regia ) seed coat extract and modulates some metabolic enzymes activity in triton WR-1339-INDUCED hyperlipidemia in rat kidney, liver, and heart. J. Mol. Recognit., 2023, 36(3), e3004.
[http://dx.doi.org/10.1002/jmr.3004] [PMID: 36537558]
[11]
Özaslan, M.S.; Sağlamtaş, R.; Demir, Y.; Genç, Y.; Saraçoğlu, İ.; Gülçin, İ. Isolation of some phenolic compounds from Plantago subulata L. and determination of their antidiabetic, anticholinesterase, antiepileptic and antioxidant activity. Chem. Biodivers., 2022, 19(8), e202200280.
[http://dx.doi.org/10.1002/cbdv.202200280] [PMID: 35796520]
[12]
Türkeş, C; Demir, Y; Beydemir, Ş. In vitro inhibitory activity and molecular docking study of selected natural phenolic compounds as ar and sdh inhibitors. ChemistrySelect, 2022, 7(48), e202204050.
[13]
Demir, Y.; Türkeş, C.; Küfrevioğlu, Ö.İ.; Beydemir, Ş. Molecular docking studies and the effect of fluorophenylthiourea derivatives on glutathione-dependent enzymes. Chem. Biodivers., 2023, 20(1), e202200656.
[http://dx.doi.org/10.1002/cbdv.202200656] [PMID: 36538730]
[14]
Yıldız, M.L.; Demir, Y.; Küfrevioğlu, Ö.I. Screening of in vitro and in silico effect of Fluorophenylthiourea compounds on glucose 6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase enzymes. J. Mol. Recognit., 2022, 35(12), e2987.
[http://dx.doi.org/10.1002/jmr.2987] [PMID: 36326002]
[15]
Malarkey, D.E.; Johnson, K.; Ryan, L.; Boorman, G.; Maronpot, R.R. New insights into functional aspects of liver morphology. Toxicol. Pathol., 2005, 33(1), 27-34.
[http://dx.doi.org/10.1080/01926230590881826] [PMID: 15805053]
[16]
Taslimi, P.; Kandemir, F.M.; Demir, Y.; İleritürk, M.; Temel, Y.; Caglayan, C.; Gulçin, İ. The antidiabetic and anticholinergic effects of chrysin on cyclophosphamide-induced multiple organ toxicity in rats: Pharmacological evaluation of some metabolic enzyme activities. J. Biochem. Mol. Toxicol., 2019, 33(6), e22313.
[http://dx.doi.org/10.1002/jbt.22313] [PMID: 30801880]
[17]
Çağlayan, C.; Taslimi, P.; Demir, Y.; Küçükler, S.; Kandemir, F.M.; Gulçin, İ. The effects of zingerone against vancomycin-induced lung, liver, kidney and testis toxicity in rats: The behavior of some metabolic enzymes. J. Biochem. Mol. Toxicol., 2019, 33(10), e22381.
[http://dx.doi.org/10.1002/jbt.22381] [PMID: 31454121]
[18]
Bouwens, L.; De Bleser, P.; Vanderkerken, K.; Geerts, B.; Wisse, E. Liver cell heterogeneity: Functions of non-parenchymal cells. Enzyme, 1992, 46(1-3), 155-168.
[http://dx.doi.org/10.1159/000468782] [PMID: 1289080]
[19]
Senoo, H. Structure and function of hepatic stellate cells. Med. Electron Microsc., 2004, 37(1), 3-15.
[http://dx.doi.org/10.1007/s00795-003-0230-3] [PMID: 15057600]
[20]
Dixon, L.J.; Barnes, M.; Tang, H.; Pritchard, M.T.; Nagy, L.E. Kupffer cells in the liver. Compr. Physiol., 2013, 3(2), 785-797.
[http://dx.doi.org/10.1002/cphy.c120026] [PMID: 23720329]
[21]
Elvevold, K.; Smedsrød, B.; Martinez, I. The liver sinusoidal endothelial cell: A cell type of controversial and confusing identity. Am. J. Physiol. Gastrointest. Liver Physiol., 2008, 294(2), G391-G400.
[http://dx.doi.org/10.1152/ajpgi.00167.2007] [PMID: 18063708]
[22]
Tabibian, J.H.; Masyuk, A.I.; Masyuk, T.V.; O’Hara, S.P.; LaRusso, N.F. Physiology of cholangiocytes. Compr. Physiol., 2013, 3(1), 541-565.
[http://dx.doi.org/10.1002/cphy.c120019] [PMID: 23720296]
[23]
Kumar, S.; Duan, Q.; Wu, R.; Harris, E.N.; Su, Q. Pathophysiological communication between hepatocytes and non-parenchymal cells in liver injury from NAFLD to liver fibrosis. Adv. Drug Deliv. Rev., 2021, 176, 113869.
[http://dx.doi.org/10.1016/j.addr.2021.113869] [PMID: 34280515]
[24]
Wang, L.; Wang, Y.; Quan, J. Exosomes derived from natural killer cells inhibit hepatic stellate cell activation and liver fibrosis. Hum. Cell, 2020, 33(3), 582-589.
[http://dx.doi.org/10.1007/s13577-020-00371-5] [PMID: 32449114]
[25]
Zhou, W.; Luo, J.; Xie, X.; Yang, S.; Zhu, D.; Huang, H.; Yang, D.; Liu, J. Gut microbiota dysbiosis strengthens kupffer cell-mediated hepatitis B virus persistence through inducing endotoxemia in mice. J. Clin. Transl. Hepatol., 2022, 10(1), 17-25.
[http://dx.doi.org/10.14218/JCTH.2020.00161] [PMID: 35233369]
[26]
Matsumoto, M.; Zhang, J.; Zhang, X.; Liu, J.; Jiang, J.X.; Yamaguchi, K.; Taruno, A.; Katsuyama, M.; Iwata, K.; Ibi, M.; Cui, W.; Matsuno, K.; Marunaka, Y.; Itoh, Y.; Torok, N.J.; Yabe-Nishimura, C. The NOX1 isoform of NADPH oxidase is involved in dysfunction of liver sinusoids in nonalcoholic fatty liver disease. Free Radic. Biol. Med., 2018, 115, 412-420.
[http://dx.doi.org/10.1016/j.freeradbiomed.2017.12.019] [PMID: 29274380]
[27]
Lim, H.K.; Jeffrey, G.P.; Ramm, G.A.; Soekmadji, C. Pathogenesis of viral hepatitis-induced chronic liver disease: Role of extracellular vesicles. Front. Cell. Infect. Microbiol., 2020, 10, 587628.
[http://dx.doi.org/10.3389/fcimb.2020.587628] [PMID: 33240824]
[28]
Mao, X.; Cheung, K.S.; Peng, C.; Mak, L.Y.; Cheng, H.M.; Fung, J. Steatosis, HBV-related HCC, cirrhosis, and HBsAg seroclearance: A systematic review and meta-analysis. Hepatology, 2022.
[PMID: 36111362]
[29]
Wang, C.C.; Cheng, P.N.; Kao, J.H. Systematic review: Chronic viral hepatitis and metabolic derangement. Aliment. Pharmacol. Ther., 2020, 51(2), 216-230.
[http://dx.doi.org/10.1111/apt.15575] [PMID: 31746482]
[30]
Stockdale, A.J.; Kreuels, B.; Henrion, M.Y.R.; Giorgi, E.; Kyomuhangi, I.; de Martel, C.; Hutin, Y.; Geretti, A.M. The global prevalence of hepatitis D virus infection: Systematic review and meta-analysis. J. Hepatol., 2020, 73(3), 523-532.
[http://dx.doi.org/10.1016/j.jhep.2020.04.008] [PMID: 32335166]
[31]
Taylor, R.S.; Taylor, R.J.; Bayliss, S.; Hagström, H.; Nasr, P.; Schattenberg, J.M.; Ishigami, M.; Toyoda, H.; Wai-Sun Wong, V.; Peleg, N.; Shlomai, A.; Sebastiani, G.; Seko, Y.; Bhala, N.; Younossi, Z.M.; Anstee, Q.M.; McPherson, S.; Newsome, P.N. Association between fibrosis stage and outcomes of patients with nonalcoholic fatty liver disease: A systematic review and meta-analysis. Gastroenterology, 2020, 158(6), 1611-1625.e12.
[http://dx.doi.org/10.1053/j.gastro.2020.01.043] [PMID: 32027911]
[32]
Hyun, J.; Han, J.; Lee, C.; Yoon, M.; Jung, Y. Pathophysiological aspects of alcohol metabolism in the liver. Int. J. Mol. Sci., 2021, 22(11), 5717.
[http://dx.doi.org/10.3390/ijms22115717] [PMID: 34071962]
[33]
Nguyen-Khac, E.; Thiele, M.; Voican, C.; Nahon, P.; Moreno, C.; Boursier, J.; Mueller, S.; de Ledinghen, V.; Stärkel, P.; Gyune Kim, S.; Fernandez, M.; Madsen, B.; Naveau, S.; Krag, A.; Perlemuter, G.; Ziol, M.; Chatelain, D.; Diouf, M. Non-invasive diagnosis of liver fibrosis in patients with alcohol-related liver disease by transient elastography: An individual patient data meta-analysis. Lancet Gastroenterol. Hepatol., 2018, 3(9), 614-625.
[http://dx.doi.org/10.1016/S2468-1253(18)30124-9] [PMID: 29983372]
[34]
Hamesch, K.; Mandorfer, M.; Pereira, V.M.; Moeller, L.S.; Pons, M.; Dolman, G.E.; Reichert, M.C.; Schneider, C.V.; Woditsch, V.; Voss, J.; Lindhauer, C.; Fromme, M.; Spivak, I.; Guldiken, N.; Zhou, B.; Arslanow, A.; Schaefer, B.; Zoller, H.; Aigner, E.; Reiberger, T.; Wetzel, M.; Siegmund, B.; Simões, C.; Gaspar, R.; Maia, L.; Costa, D.; Bento-Miranda, M.; van Helden, J.; Yagmur, E.; Bzdok, D.; Stolk, J.; Gleiber, W.; Knipel, V.; Windisch, W.; Mahadeva, R.; Bals, R.; Koczulla, R.; Barrecheguren, M.; Miravitlles, M.; Janciauskiene, S.; Stickel, F.; Lammert, F.; Liberal, R.; Genesca, J.; Griffiths, W.J.; Trauner, M.; Krag, A.; Trautwein, C.; Strnad, P. Liver fibrosis and metabolic alterations in adults with alpha-1-antitrypsin deficiency caused by the Pi*ZZ mutation. Gastroenterology, 2019, 157(3), 705-719.e18.
[http://dx.doi.org/10.1053/j.gastro.2019.05.013] [PMID: 31121167]
[35]
Strnad, P.; Mandorfer, M.; Choudhury, G.; Griffiths, W.; Trautwein, C.; Loomba, R.; Schluep, T.; Chang, T.; Yi, M.; Given, B.D.; Hamilton, J.C.; San Martin, J.; Teckman, J.H. Fazirsiran for liver disease associated with alpha 1 -antitrypsin deficiency. N. Engl. J. Med., 2022, 387(6), 514-524.
[http://dx.doi.org/10.1056/NEJMoa2205416] [PMID: 35748699]
[36]
Powell, L.W.; Dixon, J.L.; Ramm, G.A.; Purdie, D.M.; Lincoln, D.J.; Anderson, G.J.; Subramaniam, V.N.; Hewett, D.G.; Searle, J.W.; Fletcher, L.M.; Crawford, D.H.; Rodgers, H.; Allen, K.J.; Cavanaugh, J.A.; Bassett, M.L. Screening for hemochromatosis in asymptomatic subjects with or without a family history. Arch. Intern. Med., 2006, 166(3), 294-301.
[http://dx.doi.org/10.1001/archinte.166.3.294] [PMID: 16476869]
[37]
Przybylkowski, A; Szeligowska, J; Januszewicz, M; Raszeja-Wyszomirska, J; Szczepankiewicz, B; Nehring, P Evaluation of liver fibrosis in patients with Wilson's disease. Eur. J. Gastroenterol. Hepatol., 2021, 33(4), 535-540.
[38]
Huang, S.P.; Chen, S.; Ma, Y.Z.; Zhou, A.; Jiang, H.; Wu, P. Evaluation of the mechanism of jiedu huazhuo quyu formula in treating wilson’s disease-associated liver fibrosis by network pharmacology analysis and molecular dynamics simulation. Evid. Based Complement. Alternat. Med., 2022, 2022, 1-14.
[http://dx.doi.org/10.1155/2022/9363131] [PMID: 35707473]
[39]
Manns, M.P.; Czaja, A.J.; Gorham, J.D.; Krawitt, E.L.; Mieli-Vergani, G.; Vergani, D.; Vierling, J.M. Diagnosis and management of autoimmune hepatitis. Hepatology, 2010, 51(6), 2193-2213.
[http://dx.doi.org/10.1002/hep.23584] [PMID: 20513004]
[40]
Galina, P.; Alexopoulou, E.; Mentessidou, A.; Mirilas, P.; Zellos, A.; Lykopoulou, L.; Patereli, A.; Salpasaranis, K.; Kelekis, N.L.; Zarifi, M. Diagnostic accuracy of two-dimensional shear wave elastography in detecting hepatic fibrosis in children with autoimmune hepatitis, biliary atresia and other chronic liver diseases. Pediatr. Radiol., 2021, 51(8), 1358-1368.
[http://dx.doi.org/10.1007/s00247-020-04959-9] [PMID: 33755748]
[41]
Zhang, J.; Lyu, Z.; Li, B.; You, Z.; Cui, N.; Li, Y.; Li, Y.; Huang, B.; Chen, R.; Chen, Y.; Peng, Y.; Fang, J.; Wang, Q.; Miao, Q.; Tang, R.; Gershwin, M.E.; Lian, M.; Xiao, X.; Ma, X. P4HA2 induces hepatic ductular reaction and biliary fibrosis in chronic cholestatic liver diseases. Hepatology, 2023, 78(1), 10-25.
[http://dx.doi.org/10.1097/HEP.0000000000000317] [PMID: 36799463]
[42]
Biswas, A.; Santra, S.; Bishnu, D.; Dhali, G.K.; Chowdhury, A.; Santra, A. Isoniazid and rifampicin produce hepatic fibrosis through an oxidative stress-dependent mechanism. Int. J. Hepatol., 2020, 2020, 1-12.
[http://dx.doi.org/10.1155/2020/6987295] [PMID: 32373368]
[43]
Huang, C.H.; Lai, Y.Y.; Kuo, Y.J.; Yang, S.C.; Chang, Y.J.; Chang, K.K.; Chen, W.K. Amiodarone and risk of liver cirrhosis: A nationwide, population-based study. Ther. Clin. Risk Manag., 2019, 15, 103-112.
[http://dx.doi.org/10.2147/TCRM.S174868] [PMID: 30666120]
[44]
Gelfand, J.M.; Wan, J.; Zhang, H.; Shin, D.B.; Ogdie, A.; Syed, M.N.; Egeberg, A. Risk of liver disease in patients with psoriasis, psoriatic arthritis, and rheumatoid arthritis receiving methotrexate: A population-based study. J. Am. Acad. Dermatol., 2021, 84(6), 1636-1643.
[http://dx.doi.org/10.1016/j.jaad.2021.02.019] [PMID: 33607181]
[45]
Dewidar, B; Meyer, C; Dooley, S; Meindl-Beinker, AN TGF-beta in hepatic stellate cell activation and liver fibrogenesis-updated 2019. Cells, 2019, 8(11), 1419.
[46]
Massague, J. TGFbeta signalling in context. Nat. Rev. Mol. Cell Biol., 2012, 13, 616-630.
[47]
Armendáriz-Borunda, J.; Rincón, A.R.; Muñoz-Valle, J.F.; Bueno-Topete, M.; Oregón-Romero, E.; Islas-Carbajal, M.C.; Medina-Preciado, D.; González-García, I.; Bautista, C.A.; García-Rocha, S.; Godoy, J.; Vázquez-Del Mercado, M.; Troyo-SanRoman, R.; Arellano-Olivera, I.; Lucano, S.; Álvarez-Rodríguez, A.; Salazar, A. Fibrogenic polymorphisms (TGF-beta, PAI-1, AT) in Mexican patients with established liver fibrosis. Potential correlation with pirfenidone treatment. J. Investig. Med., 2008, 56(7), 944-953.
[http://dx.doi.org/10.2310/JIM.0b013e3181891512] [PMID: 18797412]
[48]
Tao, R.; Fan, X.X.; Yu, H.J.; Ai, G.; Zhang, H.Y.; Kong, H.Y.; Song, Q.Q.; Huang, Y.; Huang, J.Q.; Ning, Q. Retracted : MicroRNA-29b-3p prevents Schistosoma japonicum -induced liver fibrosis by targeting COL1A1 and COL3A1. J. Cell. Biochem., 2018, 119(4), 3199-3209.
[http://dx.doi.org/10.1002/jcb.26475] [PMID: 29091295]
[49]
Xiong, L.J.; Zhu, J.F.; Luo, D.D.; Zen, L.L.; Cai, S.Q. Effects of pentoxifylline on the hepatic content of TGF-β1 and collagen in Schistosomiasis japonica mice with liver fibrosis. World J. Gastroenterol., 2003, 9(1), 152-154.
[http://dx.doi.org/10.3748/wjg.v9.i1.152] [PMID: 12508372]
[50]
Yang, Y.; Sun, M.; Li, W.; Liu, C.; Jiang, Z.; Gu, P.; Li, J.; Wang, W.; You, R.; Ba, Q.; Li, X.; Wang, H. Rebalancing TGF-β/Smad7 signaling via Compound kushen injection in hepatic stellate cells protects against liver fibrosis and hepatocarcinogenesis. Clin. Transl. Med., 2021, 11(7), e410.
[http://dx.doi.org/10.1002/ctm2.410] [PMID: 34323416]
[51]
Schon, H.T.; Weiskirchen, R. Immunomodulatory effects of transforming growth factor-β in the liver. Hepatobiliary Surg. Nutr., 2014, 3(6), 386-406.
[PMID: 25568862]
[52]
Milani, S.; Herbst, H.; Schuppan, D.; Stein, H.; Surrenti, C. Transforming growth factors beta 1 and beta 2 are differentially expressed in fibrotic liver disease. Am. J. Pathol., 1991, 139(6), 1221-1229.
[PMID: 1750499]
[53]
Rosenfeld, M.; Keating, A.; Bowen-Pope, D.F.; Singer, J.W.; Ross, R. Responsiveness of the in vitro hematopoietic microenvironment to platelet-derived growth factor. Leuk. Res., 1985, 9(4), 427-434.
[http://dx.doi.org/10.1016/0145-2126(85)90001-3] [PMID: 2987621]
[54]
Niba, E.T.E.; Nagaya, H.; Kanno, T.; Tsuchiya, A.; Gotoh, A.; Tabata, C.; Kuribayashi, K.; Nakano, T.; Nishizaki, T. Crosstalk between PI3 kinase/PDK1/Akt/Rac1 and Ras/Raf/MEK/ERK pathways downstream PDGF receptor. Cell. Physiol. Biochem., 2013, 31(6), 905-913.
[http://dx.doi.org/10.1159/000350108] [PMID: 23817184]
[55]
Pan, T.L.; Wang, P.W.; Leu, Y.L.; Wu, T.H.; Wu, T.S. Inhibitory effects of Scutellaria baicalensis extract on hepatic stellate cells through inducing G2/M cell cycle arrest and activating ERK-dependent apoptosis via Bax and caspase pathway. J. Ethnopharmacol., 2012, 139(3), 829-837.
[http://dx.doi.org/10.1016/j.jep.2011.12.028] [PMID: 22210104]
[56]
Zvibel, I.; Bar-Zohar, D.; Kloog, Y.; Oren, R.; Reif, S. The effect of Ras inhibition on the proliferation, apoptosis and matrix metalloproteases activity in rat hepatic stellate cells. Dig. Dis. Sci., 2008, 53(4), 1048-1053.
[http://dx.doi.org/10.1007/s10620-007-9984-0] [PMID: 17934818]
[57]
Brady, L.M.; Fox, E.S.; Fimmel, C.J. Polyenylphosphatidylcholine inhibits PDGF-induced proliferation in rat hepatic stellate cells. Biochem. Biophys. Res. Commun., 1998, 248(1), 174-179.
[http://dx.doi.org/10.1006/bbrc.1998.8935] [PMID: 9675106]
[58]
Deng, W.; Meng, Z.; Sun, A.; Yang, Z. Pioglitazone suppresses inflammation and fibrosis in nonalcoholic fatty liver disease by down-regulating PDGF and TIMP-2: Evidence from in vitro study. Cancer Biomark., 2017, 20(4), 411-415.
[http://dx.doi.org/10.3233/CBM-170157] [PMID: 28946547]
[59]
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. Mol. Med. Rep., 2017, 16(6), 7879-7889.
[http://dx.doi.org/10.3892/mmr.2017.7641] [PMID: 28983598]
[60]
Sims, G.P.; Rowe, D.C.; Rietdijk, S.T.; Herbst, R.; Coyle, A.J. HMGB1 and RAGE in inflammation and cancer. Annu. Rev. Immunol., 2010, 28(1), 367-388.
[http://dx.doi.org/10.1146/annurev.immunol.021908.132603] [PMID: 20192808]
[61]
Scaffidi, P.; Misteli, T.; Bianchi, M.E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature, 2002, 418(6894), 191-195.
[http://dx.doi.org/10.1038/nature00858] [PMID: 12110890]
[62]
Mencin, A.; Kluwe, J.; Schwabe, R.F. Toll-like receptors as targets in chronic liver diseases. Gut, 2009, 58(5), 704-720.
[http://dx.doi.org/10.1136/gut.2008.156307] [PMID: 19359436]
[63]
Yu, M.; Wang, H.; Ding, A.; Golenbock, D.T.; Latz, E.; Czura, C.J.; Fenton, M.J.; Tracey, K.J.; Yang, H. HMGB1 signals through toll-like receptor (TLR) 4 and TLR2. Shock, 2006, 26(2), 174-179.
[http://dx.doi.org/10.1097/01.shk.0000225404.51320.82] [PMID: 16878026]
[64]
Luedde, T.; Schwabe, R.F. NF-κB in the liver-linking injury, fibrosis and hepatocellular carcinoma. Nat. Rev. Gastroenterol. Hepatol., 2011, 8(2), 108-118.
[http://dx.doi.org/10.1038/nrgastro.2010.213] [PMID: 21293511]
[65]
Rim, E.Y.; Clevers, H.; Nusse, R. The WnT pathway: From signaling mechanisms to synthetic modulators. Annu. Rev. Biochem., 2022, 91(1), 571-598.
[http://dx.doi.org/10.1146/annurev-biochem-040320-103615] [PMID: 35303793]
[66]
Ma, Z.G.; Lv, X.D.; Zhan, L.L.; Chen, L.; Zou, Q.Y.; Xiang, J.Q.; Qin, J.L.; Zhang, W.W.; Zeng, Z.J.; Jin, H.; Jiang, H.X.; Lv, X.P. Human urokinase-type plasminogen activator gene-modified bone marrow-derived mesenchymal stem cells attenuate liver fibrosis in rats by down-regulating the Wnt signaling pathway. World J. Gastroenterol., 2016, 22(6), 2092-2103.
[http://dx.doi.org/10.3748/wjg.v22.i6.2092] [PMID: 26877613]
[67]
Klein, D.; Demory, A.; Peyre, F.; Kroll, J.; Augustin, H.G.; Helfrich, W.; Kzhyshkowska, J.; Schledzewski, K.; Arnold, B.; Goerdt, S. Wnt2 acts as a cell type-specific, autocrine growth factor in rat hepatic sinusoidal endothelial cells cross-stimulating the VEGF pathway. Hepatology, 2008, 47(3), 1018-1031.
[http://dx.doi.org/10.1002/hep.22084] [PMID: 18302287]
[68]
Xin, P.; Xu, X.; Deng, C.; Liu, S.; Wang, Y.; Zhou, X.; Ma, H.; Wei, D.; Sun, S. The role of JAK/STAT signaling pathway and its inhibitors in diseases. Int. Immunopharmacol., 2020, 80, 106210.
[http://dx.doi.org/10.1016/j.intimp.2020.106210] [PMID: 31972425]
[69]
Xiang, D.M.; Sun, W.; Ning, B.F.; Zhou, T.F.; Li, X.F.; Zhong, W.; Cheng, Z.; Xia, M.Y.; Wang, X.; Deng, X.; Wang, W.; Li, H.Y.; Cui, X.L.; Li, S.C.; Wu, B.; Xie, W.F.; Wang, H.Y.; Ding, J. The HLF/IL-6/STAT3 feedforward circuit drives hepatic stellate cell activation to promote liver fibrosis. Gut, 2018, 67(9), 1704-1715.
[http://dx.doi.org/10.1136/gutjnl-2016-313392] [PMID: 28754776]
[70]
Martí-Rodrigo, A.; Alegre, F.; Moragrega, Á.B.; García-García, F.; Martí-Rodrigo, P.; Fernández-Iglesias, A.; Gracia-Sancho, J.; Apostolova, N.; Esplugues, J.V.; Blas-García, A. Rilpivirine attenuates liver fibrosis through selective STAT1-mediated apoptosis in hepatic stellate cells. Gut, 2020, 69(5), 920-932.
[http://dx.doi.org/10.1136/gutjnl-2019-318372] [PMID: 31530714]
[71]
Xuan, Y.; Chen, S.; Ding, X.; Wang, L.; Li, S.; Yang, G.; Lan, T. Tetrahydropalmatine attenuates liver fibrosis by suppressing endoplasmic reticulum stress in hepatic stellate cells. Chin. Med. J., 2022, 135(5), 628-630.
[http://dx.doi.org/10.1097/CM9.0000000000001883] [PMID: 34967794]
[72]
Shu, G.; Yusuf, A.; Dai, C.; Sun, H.; Deng, X. Piperine inhibits AML-12 hepatocyte EMT and LX-2 HSC activation and alleviates mouse liver fibrosis provoked by CCl 4 : Roles in the activation of the Nrf2 cascade and subsequent suppression of the TGF-β1/Smad axis. Food Funct., 2021, 12(22), 11686-11703.
[http://dx.doi.org/10.1039/D1FO02657G] [PMID: 34730139]
[73]
Abdelhamid, A.M.; Selim, A.; Zaafan, M.A. The hepatoprotective effect of piperine against thioacetamide-induced liver fibrosis in mice: The involvement of miR-17 and TGF-β/smads pathways. Front. Mol. Biosci., 2021, 8, 754098.
[http://dx.doi.org/10.3389/fmolb.2021.754098] [PMID: 34778375]
[74]
Vargas-Pozada, E.E.; Ramos-Tovar, E.; Rodriguez-Callejas, J.D.; Cardoso-Lezama, I.; Galindo-Gómez, S.; Talamás-Lara, D.; Vásquez-Garzón, V.R.; Arellanes-Robledo, J.; Tsutsumi, V.; Villa-Treviño, S.; Muriel, P. Caffeine inhibits NLRP3 inflammasome activation by downregulating TLR4/MAPK/NF-κB signaling pathway in an experimental NASH model. Int. J. Mol. Sci., 2022, 23(17), 9954.
[http://dx.doi.org/10.3390/ijms23179954] [PMID: 36077357]
[75]
Vargas-Pozada, E.E.; Ramos-Tovar, E.; Acero-Hernández, C.; Cardoso-Lezama, I.; Galindo-Gómez, S.; Tsutsumi, V.; Muriel, P. Caffeine mitigates experimental nonalcoholic steatohepatitis and the progression of thioacetamide-induced liver fibrosis by blocking the MAPK and TGF-β/Smad3 signaling pathways. Ann. Hepatol., 2022, 27(2), 100671.
[http://dx.doi.org/10.1016/j.aohep.2022.100671] [PMID: 35065262]
[76]
Alkreathy, H.M.; Esmat, A. Lycorine ameliorates thioacetamide-induced hepatic fibrosis in rats: Emphasis on antioxidant, anti-inflammatory, and STAT3 inhibition effects. Pharmaceuticals, 2022, 15(3), 369.
[http://dx.doi.org/10.3390/ph15030369] [PMID: 35337166]
[77]
Song, L.Y.; Ma, Y.T.; Fang, W.J.; He, Y.; Wu, J.L.; Zuo, S.R.; Deng, Z.Z.; Wang, S.F.; Liu, S.K. Inhibitory effects of oxymatrine on hepatic stellate cells activation through TGF-β/miR-195/Smad signaling pathway. BMC Complement. Altern. Med., 2019, 19(1), 138.
[http://dx.doi.org/10.1186/s12906-019-2560-2] [PMID: 31221141]
[78]
Yamaguchi, M.; Ohbayashi, S.; Ooka, A.; Yamashita, H.; Motohashi, N.; Kaneko, Y.K.; Kimura, T.; Saito, S.; Ishikawa, T. Harmine suppresses collagen production in hepatic stellate cells by inhibiting DYRK1B. Biochem. Biophys. Res. Commun., 2022, 600, 136-141.
[http://dx.doi.org/10.1016/j.bbrc.2022.02.054] [PMID: 35219102]
[79]
Hu, Z.; Su, H.; Zeng, Y.; Lin, C.; Guo, Z.; Zhong, F.; Jiang, K.; Yuan, G.; He, S. Tetramethylpyrazine ameliorates hepatic fibrosis through autophagy-mediated inflammation. Biochem. Cell Biol., 2020, 98(3), 327-337.
[http://dx.doi.org/10.1139/bcb-2019-0059] [PMID: 32383631]
[80]
Xiang, D.; Zou, J.; Zhu, X.; Chen, X.; Luo, J.; Kong, L.; Zhang, H. Physalin D attenuates hepatic stellate cell activation and liver fibrosis by blocking TGF-β/Smad and YAP signaling. Phytomedicine, 2020, 78, 153294.
[http://dx.doi.org/10.1016/j.phymed.2020.153294] [PMID: 32771890]
[81]
Zhu, X.; Ye, S.; Yu, D.; Zhang, Y.; Li, J.; Zhang, M.; Leng, Y.; Yang, T.; Luo, J.; Chen, X.; Zhang, H.; Kong, L. Physalin B attenuates liver fibrosis via suppressing LAP2α–HDAC1-mediated deacetylation of the transcription factor GLI1 and hepatic stellate cell activation. Br. J. Pharmacol., 2021, 178(17), 3428-3447.
[http://dx.doi.org/10.1111/bph.15490] [PMID: 33864382]
[82]
Tan, Y.; Li, C.; Zhou, J.; Deng, F.; Liu, Y. Berberine attenuates liver fibrosis by autophagy inhibition triggering apoptosis via the miR-30a-5p/ATG5 axis. Exp. Cell Res., 2023, 427(2), 113600.
[http://dx.doi.org/10.1016/j.yexcr.2023.113600] [PMID: 37062521]
[83]
Eissa, L.A.; Kenawy, H.I.; El-Karef, A.; Elsherbiny, N.M.; El-Mihi, K.A. Antioxidant and anti-inflammatory activities of berberine attenuate hepatic fibrosis induced by thioacetamide injection in rats. Chem. Biol. Interact., 2018, 294, 91-100.
[http://dx.doi.org/10.1016/j.cbi.2018.08.016] [PMID: 30138605]
[84]
Sheng, J.; Zhang, B.; Chen, Y.; Yu, F. Capsaicin attenuates liver fibrosis by targeting Notch signaling to inhibit TNF-α secretion from M1 macrophages. Immunopharmacol. Immunotoxicol., 2020, 42(6), 556-563.
[http://dx.doi.org/10.1080/08923973.2020.1811308] [PMID: 32811220]
[85]
Lv, X.T.; Wang, R.H.; Liu, X.T.; Ye, Y.J.; Liu, X.Y.; Qiao, J.D.; Wang, G.E. Theacrine ameliorates experimental liver fibrosis in rats by lowering cholesterol storage via activation of the Sirtuin 3-farnesoid X receptor signaling pathway. Chem. Biol. Interact., 2022, 364, 110051.
[http://dx.doi.org/10.1016/j.cbi.2022.110051] [PMID: 35872049]
[86]
Demir, Y. Naphthoquinones, benzoquinones, and anthraquinones: Molecular docking, ADME and inhibition studies on human serum paraoxonase-1 associated with cardiovascular diseases. Drug Dev. Res., 2020, 81(5), 628-636.
[http://dx.doi.org/10.1002/ddr.21667] [PMID: 32232985]
[87]
Demir, Y.; Durmaz, L.; Taslimi, P.; Gulçin, İ. Antidiabetic properties of dietary phenolic compounds: Inhibition effects on α-amylase, aldose reductase, and α-glycosidase. Biotechnol. Appl. Biochem., 2019, 66(5), 781-786.
[http://dx.doi.org/10.1002/bab.1781] [PMID: 31135076]
[88]
Aslan, H.E.; Demir, Y.; Özaslan, M.S.; Türkan, F.; Beydemir, Ş.; Küfrevioğlu, Ö.I. The behavior of some chalcones on acetylcholinesterase and carbonic anhydrase activity. Drug Chem. Toxicol., 2019, 42(6), 634-640.
[http://dx.doi.org/10.1080/01480545.2018.1463242] [PMID: 29860891]
[89]
Mahfouz, M.M.; Abdelsalam, R.M.; Masoud, M.A.; Mansour, H.A.; Ahmed-Farid, O.A.; kenawy, S.A. The neuroprotective effect of mesenchymal stem cells on an experimentally induced model for multiple sclerosis in mice. J. Biochem. Mol. Toxicol., 2017, 31(9), e21936.
[http://dx.doi.org/10.1002/jbt.21936] [PMID: 28557239]
[90]
Özaslan, M.S.; Demir, Y.; Aslan, H.E.; Beydemir, Ş.; Küfrevioğlu, Ö.İ. Evaluation of chalcones as inhibitors of glutathione S-transferase. J. Biochem. Mol. Toxicol., 2018, 32(5), e22047.
[http://dx.doi.org/10.1002/jbt.22047] [PMID: 29473699]
[91]
Demir, Y.; Özaslan, M.S.; Duran, H.E.; Küfrevioğlu, Ö.İ.; Beydemir, Ş. Inhibition effects of quinones on aldose reductase: Antidiabetic properties. Environ. Toxicol. Pharmacol., 2019, 70, 103195.
[http://dx.doi.org/10.1016/j.etap.2019.103195] [PMID: 31125830]
[92]
Feng, J.; Wang, C.; Liu, T.; Li, J.; Wu, L.; Yu, Q.; Li, S.; Zhou, Y.; Zhang, J.; Chen, J.; Ji, J.; Chen, K.; Mao, Y.; Wang, F.; Dai, W.; Fan, X.; Wu, J.; Guo, C. Procyanidin B2 inhibits the activation of hepatic stellate cells and angiogenesis via the Hedgehog pathway during liver fibrosis. J. Cell. Mol. Med., 2019, 23(9), 6479-6493.
[http://dx.doi.org/10.1111/jcmm.14543] [PMID: 31328391]
[93]
Xu, Y.; Zhang, D.; Yang, H.; Liu, Y.; Zhang, L.; Zhang, C.; Chen, G.; Hu, Y.; Chen, J.; Zhang, H.; Mu, Y.; Liu, P.; Liu, W. Hepatoprotective effect of genistein against dimethylnitrosamine-induced liver fibrosis in rats by regulating macrophage functional properties and inhibiting the JAK2/STAT3/SOCS3 signaling pathway. Frontiers in Bioscience-Landmark, 2021, 26(12), 1572-1584.
[http://dx.doi.org/10.52586/5050] [PMID: 34994171]
[94]
Xu, T.; Huang, S.; Huang, Q.; Ming, Z.; Wang, M.; Li, R.; Zhao, Y. Kaempferol attenuates liver fibrosis by inhibiting activin receptor–like kinase 5. J. Cell. Mol. Med., 2019, 23(9), 6403-6410.
[http://dx.doi.org/10.1111/jcmm.14528] [PMID: 31273920]
[95]
Huang, S.; Wang, Y.; Xie, S.; Lai, Y.; Mo, C.; Zeng, T.; Kuang, S.; Zhou, C.; Zeng, Z.; Chen, Y.; Huang, S.; Gao, L.; Lv, Z. Isoliquiritigenin alleviates liver fibrosis through caveolin-1-mediated hepatic stellate cells ferroptosis in zebrafish and mice. Phytomedicine, 2022, 101, 154117.
[http://dx.doi.org/10.1016/j.phymed.2022.154117] [PMID: 35489326]
[96]
Liu, N.; Feng, J.; Lu, X.; Yao, Z.; Liu, Q.; Lv, Y.; Han, Y.; Deng, J.; Zhou, Y. Isorhamnetin inhibits liver fibrosis by reducing autophagy and inhibiting extracellular matrix formation via the TGF- β 1/Smad3 and TGF- β 1/p38 MAPK pathways. Mediators Inflamm., 2019, 2019, 1-14.
[http://dx.doi.org/10.1155/2019/6175091] [PMID: 31467486]
[97]
Liu, G.; Wei, C.; Yuan, S.; Zhang, Z.; Li, J.; Zhang, L.; Wang, G.; Fang, L. Wogonoside attenuates liver fibrosis by triggering hepatic stellate cell ferroptosis through SOCS1 / P53 / SLC7A11 pathway. Phytother. Res., 2022, 36(11), 4230-4243.
[http://dx.doi.org/10.1002/ptr.7558] [PMID: 35817562]
[98]
Wang, W.; Chen, Z.; Zheng, T.; Zhang, M. Xanthohumol alleviates T2DM-induced liver steatosis and fibrosis by mediating the NRF2/RAGE/NF-κB signaling pathway. Future Med. Chem., 2021, 13(23), 2069-2081.
[http://dx.doi.org/10.4155/fmc-2021-0241] [PMID: 34551612]
[99]
Li, S; Li, X; Chen, F; Liu, M; Ning, L; Yan, Y. Nobiletin mitigates hepatocytes death, liver inflammation, and fibrosis in a murine model of NASH through modulating hepatic oxidative stress and mitochondrial dysfunction. J. Nutr. Biochem., 2022, 100, 108888.
[100]
Amer, M.A.; Othman, A.I.; EL-Missiry, M.A.; Farag, A.A.; Amer, M.E. Proanthocyanidins attenuated liver damage and suppressed fibrosis in CCl4-treated rats. Environ. Sci. Pollut. Res. Int., 2022, 29(60), 91127-91138.
[http://dx.doi.org/10.1007/s11356-022-22051-7] [PMID: 35881285]
[101]
Lee, E.H.; Park, K.I.; Kim, K.Y.; Lee, J.H.; Jang, E.J.; Ku, S.K.; Kim, S.C.; Suk, H.Y.; Park, J.Y.; Baek, S.Y.; Kim, Y.W. Liquiritigenin inhibits hepatic fibrogenesis and TGF-β1/Smad with Hippo/YAP signal. Phytomedicine, 2019, 62, 152780.
[http://dx.doi.org/10.1016/j.phymed.2018.12.003] [PMID: 31121384]
[102]
Du, X.S.; Li, H.D.; Yang, X.J.; Li, J.J.; Xu, J.J.; Chen, Y.; Xu, Q.Q.; Yang, L.; He, C.S.; Huang, C.; Meng, X.M.; Li, J. Wogonin attenuates liver fibrosis via regulating hepatic stellate cell activation and apoptosis. Int. Immunopharmacol., 2019, 75, 105671.
[http://dx.doi.org/10.1016/j.intimp.2019.05.056] [PMID: 31377590]
[103]
Casas-Grajales, S.; Reyes-Gordillo, K.; Cerda-García-Rojas, C.M.; Tsutsumi, V.; Lakshman, M.R.; Muriel, P. Rebaudioside A administration prevents experimental liver fibrosis: An in vivo and in vitro study of the mechanisms of action involved. J. Appl. Toxicol., 2019, 39(8), 1118-1131.
[http://dx.doi.org/10.1002/jat.3797] [PMID: 30883860]
[104]
Liu, D.; Qin, H.; Yang, B.; Du, B.; Yun, X. Oridonin ameliorates carbon tetrachloride-induced liver fibrosis in mice through inhibition of the NLRP3 inflammasome. Drug Dev. Res., 2020, 81(4), 526-533.
[http://dx.doi.org/10.1002/ddr.21649] [PMID: 32219880]
[105]
Yan, H.; Huang, Z.; Bai, Q.; Sheng, Y.; Hao, Z.; Wang, Z.; Ji, L. Natural product andrographolide alleviated APAP-induced liver fibrosis by activating Nrf2 antioxidant pathway. Toxicology, 2018, 396-397, 1-12.
[http://dx.doi.org/10.1016/j.tox.2018.01.007] [PMID: 29355602]
[106]
Ji, D.; Zhao, Q.; Qin, Y.; Tong, H.; Wang, Q.; Yu, M.; Mao, C.; Lu, T.; Qiu, J.; Jiang, C. Germacrone improves liver fibrosis by regulating the PI3K/AKT/mTOR signalling pathway. Cell Biol. Int., 2021, 45(9), 1866-1875.
[http://dx.doi.org/10.1002/cbin.11607] [PMID: 33835632]
[107]
Wan, S.; Luo, F.; Huang, C.; Liu, C.; Luo, Q.; Zhu, X. Ursolic acid reverses liver fibrosis by inhibiting interactive NOX4/ROS and RhoA/ROCK1 signalling pathways. Aging, 2020, 12(11), 10614-10632.
[http://dx.doi.org/10.18632/aging.103282] [PMID: 32496208]
[108]
Kim, J.K.; Han, N.R.; Park, S.M.; Jegal, K.H.; Jung, J.Y.; Jung, E.H.; Kim, E.O.; Kim, D.; Jung, D.H.; Lee, J.R.; Park, C.A.; Ku, S.K.; Cho, I.J.; Kim, S.C. Hemistepsin A alleviates liver fibrosis by inducing apoptosis of activated hepatic stellate cells via inhibition of nuclear factor-κB and Akt. Food Chem. Toxicol., 2020, 135, 111044.
[http://dx.doi.org/10.1016/j.fct.2019.111044] [PMID: 31830547]
[109]
Zhang, Y.; Cai, B.; Li, Y.; Xu, Y.; Wang, Y.; Zheng, L.; Zheng, X.; Yin, L.; Chen, G.; Wang, Y.; Liang, G.; Chen, L. Identification of linderalactone as a natural inhibitor of SHP2 to ameliorate CCl4-induced liver fibrosis. Front. Pharmacol., 2023, 14, 1098463.
[http://dx.doi.org/10.3389/fphar.2023.1098463] [PMID: 36843936]
[110]
Zhou, M.; Zhao, X.; Liao, L.; Deng, Y.; Liu, M.; Wang, J.; Xue, X.; Li, Y. Forsythiaside A regulates activation of hepatic stellate cells by inhibiting NOX4-dependent ROS. Oxid. Med. Cell. Longev., 2022, 2022, 1-17.
[http://dx.doi.org/10.1155/2022/9938392] [PMID: 35035671]
[111]
Zhu, H.; He, C.; Zhao, H.; Jiang, W.; Xu, S.; Li, J.; Ma, T.; Huang, C. Sennoside A prevents liver fibrosis by binding DNMT1 and suppressing DNMT1-mediated PTEN hypermethylation in HSC activation and proliferation. FASEB J., 2020, 34(11), 14558-14571.
[http://dx.doi.org/10.1096/fj.202000494RR] [PMID: 32946656]
[112]
Li, R.; Li, J.; Huang, Y.; Li, H.; Yan, S.; Lin, J.; Chen, Y.; Wu, L.; Liu, B.; Wang, G.; Lan, T. Polydatin attenuates diet-induced nonalcoholic steatohepatitis and fibrosis in mice. Int. J. Biol. Sci., 2018, 14(11), 1411-1425.
[http://dx.doi.org/10.7150/ijbs.26086] [PMID: 30262993]
[113]
Bao, X.; Li, J.; Ren, C.; Wei, J.; Lu, X.; Wang, X.; Du, W.; Jin, X.; Ma, B.; Zhang, Q.; Ma, B. Aucubin ameliorates liver fibrosis and hepatic stellate cells activation in diabetic mice via inhibiting ER stress-mediated IRE1α/TXNIP/NLRP3 inflammasome through NOX4/ROS pathway. Chem. Biol. Interact., 2022, 365, 110074.
[http://dx.doi.org/10.1016/j.cbi.2022.110074] [PMID: 35961541]
[114]
Lin, L.; Zhou, M.; Que, R.; Chen, Y.; Liu, X.; Zhang, K.; Shi, Z.; Li, Y. Saikosaponin-d protects against liver fibrosis by regulating the estrogen receptor-β/NLRP3 inflammasome pathway. Biochem. Cell Biol., 2021, 99(5), 666-674.
[http://dx.doi.org/10.1139/bcb-2020-0561] [PMID: 33974808]
[115]
Chen, Y.; Que, R.; Zhang, N.; Lin, L.; Zhou, M.; Li, Y. Saikosaponin-d alleviates hepatic fibrosis through regulating GPER1/autophagy signaling. Mol. Biol. Rep., 2021, 48(12), 7853-7863.
[http://dx.doi.org/10.1007/s11033-021-06807-x] [PMID: 34714484]
[116]
Xiao, Z.; Ji, Q.; Fu, Y.; Gao, S.; Hu, Y.; Liu, W.; Chen, G.; Mu, Y.; Chen, J.; Liu, P. Amygdalin ameliorates liver fibrosis through inhibiting activation of TGF-β/smad signaling. Chin. J. Integr. Med., 2023, 29(4), 316-324.
[http://dx.doi.org/10.1007/s11655-021-3304-y] [PMID: 34816365]
[117]
Liu, X.; Mi, X.; Wang, Z.; Zhang, M.; Hou, J.; Jiang, S.; Wang, Y.; Chen, C.; Li, W. Ginsenoside Rg3 promotes regression from hepatic fibrosis through reducing inflammation-mediated autophagy signaling pathway. Cell Death Dis., 2020, 11(6), 454.
[http://dx.doi.org/10.1038/s41419-020-2597-7] [PMID: 32532964]
[118]
Casas-Grajales, S.; Alvarez-Suarez, D.; Ramos-Tovar, E.; Dayana Buendía-Montaño, L.; Reyes-Gordillo, K.; Camacho, J.; Tsutsumi, V.; Lakshman, M.R.; Muriel, P. Stevioside inhibits experimental fibrosis by down-regulating profibrotic Smad pathways and blocking hepatic stellate cell activation. Basic Clin. Pharmacol. Toxicol., 2019, 124(6), 670-680.
[http://dx.doi.org/10.1111/bcpt.13194] [PMID: 30561898]
[119]
Zhang, Y.; Zhang, S.; Luo, X.; Zhao, H.; Xiang, X. Paeoniflorin mitigates PBC-induced liver fibrosis by repressing NLRP3 formation. Acta Cir. Bras., 2021, 36(11), e361106.
[http://dx.doi.org/10.1590/acb361106] [PMID: 35195182]
[120]
Ceylan, H.; Demir, Y.; Beydemir, Ş. Inhibitory effects of usnic and carnosic acid on some metabolic enzymes: An in vitro study. Protein Pept. Lett., 2019, 26(5), 364-370.
[http://dx.doi.org/10.2174/0929866526666190301115122] [PMID: 30827223]
[121]
Oztaskin, N.; Goksu, S.; Demir, Y.; Maras, A.; Gulcin, İ. Synthesis of novel bromophenol with diaryl methanes-determination of their inhibition effects on carbonic anhydrase and acetylcholinesterase. Molecules, 2022, 27(21), 7426.
[http://dx.doi.org/10.3390/molecules27217426] [PMID: 36364255]
[122]
Saadati, S.; Sadeghi, A.; Mansour, A.; Yari, Z.; Poustchi, H.; Hedayati, M.; Hatami, B.; Hekmatdoost, A. Curcumin and inflammation in non-alcoholic fatty liver disease: A randomized, placebo controlled clinical trial. BMC Gastroenterol., 2019, 19(1), 133.
[http://dx.doi.org/10.1186/s12876-019-1055-4] [PMID: 31345163]
[123]
Yang, Y.; Qu, Y.; Lv, X.; Zhao, R.; Yu, J.; Hu, S.; Kang, J.; Zhang, Y.; Gong, Y.; Cui, T.; Zhang, X.; Yan, Y. Sesamol supplementation alleviates nonalcoholic steatohepatitis and atherosclerosis in high-fat, high carbohydrate and high-cholesterol diet-fed rats. Food Funct., 2021, 12(19), 9347-9359.
[http://dx.doi.org/10.1039/D1FO01517F] [PMID: 34606548]
[124]
Wu, J.; Xue, X.; Fan, G.; Gu, Y.; Zhou, F.; Zheng, Q.; Liu, R.; Li, Y.; Ma, B.; Li, S.; Huang, G.; Ma, L.; Li, X. Ferulic acid ameliorates hepatic inflammation and fibrotic liver injury by inhibiting PTP1B activity and subsequent promoting AMPK phosphorylation. Front. Pharmacol., 2021, 12, 754976.
[http://dx.doi.org/10.3389/fphar.2021.754976] [PMID: 34566665]
[125]
Li, L.; Wang, K.; Jia, R.; xie, J.; Ma, L.; Hao, Z.; Zhang, W.; Mo, J.; Ren, F. Ferroportin-dependent ferroptosis induced by ellagic acid retards liver fibrosis by impairing the SNARE complexes formation. Redox Biol., 2022, 56, 102435.
[http://dx.doi.org/10.1016/j.redox.2022.102435] [PMID: 36029649]
[126]
El-Gendy, Z.A.; Ramadan, A.; El-Batran, S.A.; Ahmed, R.F.; El-Marasy, S.A.; Abd El-Rahman, S.S.; Youssef, S.A.H. Carvacrol hinders the progression of hepatic fibrosis via targeting autotaxin and thioredoxin in thioacetamide-induced liver fibrosis in rat. Hum. Exp. Toxicol., 2021, 40(12), 2188-2201.
[http://dx.doi.org/10.1177/09603271211026729] [PMID: 34155936]
[127]
Hu, M.; Zhang, D.; Xu, H.; Zhang, Y.; Shi, H.; Huang, X.; Wang, X.; Wu, Y.; Qi, Z. Salidroside activates the AMP-activated protein kinase pathway to suppress nonalcoholic steatohepatitis in mice. Hepatology, 2021, 74(6), 3056-3073.
[http://dx.doi.org/10.1002/hep.32066] [PMID: 34292604]
[128]
Liang, F.; Xu, X.; Tu, Y. Resveratrol inhibited hepatocyte apoptosis and alleviated liver fibrosis through miR-190a-5p /HGF axis. Bioorg. Med. Chem., 2022, 57, 116593.
[http://dx.doi.org/10.1016/j.bmc.2021.116593] [PMID: 35093804]
[129]
Wu, B.; Wang, R.; Li, S.; Wang, Y.; Song, F.; Gu, Y.; Yuan, Y. Antifibrotic effects of Fraxetin on carbon tetrachloride-induced liver fibrosis by targeting NF-κB/IκBα, MAPKs and Bcl-2/Bax pathways. Pharmacol. Rep., 2019, 71(3), 409-416.
[http://dx.doi.org/10.1016/j.pharep.2019.01.008] [PMID: 31003150]
[130]
Tong, Y.; Zhu, W.; Wen, T.; Mukhamejanova, Z.; Xu, F.; Xiang, Q.; Pang, J. Xyloketal B reverses nutritional hepatic steatosis, steatohepatitis, and liver fibrosis through activation of the PPARα/PGC1α signaling pathway. J. Nat. Prod., 2022, 85(7), 1738-1750.
[http://dx.doi.org/10.1021/acs.jnatprod.2c00259] [PMID: 35749236]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy