Tanshinone IIA alleviates liver fibrosis by suppressing hepatic stellate cell proliferation via ERK/cyclin D1/p-Smad3L signaling axis

Document Type : Original Article

Authors

1 School of Medicine, Jianghan University, Wuhan 430056, China

2 School of Medicine, Jingchu University of Technology, Jingmen 448000, China

Abstract

Objective(s): Liver fibrosis (LF) is a critical stage in chronic liver disease progression, and effective therapeutic drugs are currently lacking. Tanshinone IIA (Tan IIA), a monomer extracted from Salvia miltiorrhiza, shows potential in treating LF. This research aims to discuss the antifibrotic efficacy and underlying pharmacological mechanism of Tan IIA. 
Materials and Methods: The in vivo model was induced with CCl4 to form a LF model in mice, and the in vitro model was induced by TGF-β1 in LX-2 and HSC-T6 cells. Liver pathology was characterized by HE, Masson, and Sirius red staining, and serum levels of ALT, AST, LDH, and γ-GT were examined. Cell viability and proliferation were detected by Cell Counting Kit-8 and colony formation assays. Cell cycle distribution was detected by flow cytometry. The protein levels of p-ERK, cyclin D1, CDK4, and p-Smad3L were assessed through Western blot, immunohistochemistry, or immunofluorescence assays.
Results: Tan IIA markedly decreased serum levels of ALT, AST, LDH, and γ‐GT. Collagen I and α-SMA were reduced, as shown by in vitro and in vivo models. Moreover, while arresting HSCs in the G1 phase was increased, Tan II A markedly inhibited cell viability and colony formation. Mechanistically, Tan IIA decreased the expression of p-ERK, cyclin D1, CDK4, and p-Smad3L proteins in TGF-β1-activated cells and CCl4-induced mice.
Conclusion: Tan IIA may improve LF by regulating the signaling axis of ERK/cyclin D1/p-Smad3L, thereby blocking activated HSCs in the G1 phase and inhibiting their proliferation. 

Keywords

Main Subjects


1. Dhar D, Baglieri J, Kisseleva T, Brenner DA. Mechanisms of liver fibrosis and its role in liver cancer. Exp Biol Med (Maywood) 2020; 245: 96-108.
2. Pei Q, Yi Q, Tang L. Liver fibrosis resolution: From molecular mechanisms to therapeutic opportunities. Int J Mol Sci 2023; 24: 9671-9695.
3. Trivedi P, Wang S, Friedman SL. The power of plasticity-metabolic regulation of hepatic stellate cells. Cell Metab 2021; 33: 242-257.
4. Zheng Y, Xie L, Yang D, Luo K, Li X. Small-molecule natural plants for reversing liver fibrosis based on modulation of hepatic stellate cells activation: An update. Phytomedicine 2023; 113: 154721.
5. Kim J-W, Kim YJ. The evidence-based multifaceted roles of hepatic stellate cells in liver diseases: A concise review. Life Sci 2024; 344: 122547.
6. Fan WG, Liu TH, Chen W, Hammad S, Longerich T, Hausser I, et al. ECM1 prevents activation of transforming growth factor beta, hepatic stellate cells, and fibrogenesis in mice. Gastroenterology 2019; 157:1352-1381.
7. Xu XY, Geng Y, Xu HX, Ren Y, Liu DY, Mao Y. Antrodia camphorata-Derived Antrodin C Inhibits Liver Fibrosis by Blocking TGF-Beta and PDGF Signaling Pathways. Front Mol Biosci 2022; 9: 835508-835518.
8. Montazersaheb S, Farahzadi R, Fathi E, Alizadeh M, Abdolalizadeh Amir S, Khodaei Ardakan A, et al. Investigation of the apoptotic effect of silver nanoparticles (Ag-NPs) on MDA-MB 231 breast cancer epithelial cells via signaling pathways. Heliyon 2024; 10: e26959-26970.
9. Yuan Q, Shi Y, Zhang Y, Shi Y, Hussain Z, Zhao J, et al. Domperidone inhibits cell proliferation via targeting MEK and CDK4 in esophageal squamous cell carcinoma. Cancer Cell Int 2024; 24: 114-128.
10. Sun YM, Wu Y, Li GX, Liang HF, Yong TY, Li Z, et al. TGF-β downstream of Smad3 and MAPK signaling antagonistically regulate the viability and partial epithelial-mesenchymal transition of liver progenitor cells. Aging (Albany NY) 2024; 16: 6588-6612.
11. Tan Z, Sun H, Xue T, Gan C, Liu H, Xie Y, et al. Liver fibrosis: Therapeutic targets and advances in drug therapy. Front Cell Dev Biol 2021; 9: 730176-730194.
12. Xie X, Xu Y, Zhou X, Su P, Jiang X, Jin Z. The protective effect of an extract of Salvia miltiorrhiza Bunge (Danshen) on cerebral ischemic injury in animal models: A systematic review and meta-analysis. J Ethnopharmacol 2023; 317: 116772.
13. Bi Z, Wang Y, Zhang W. A comprehensive review of tanshinone IIA and its derivatives in fibrosis treatment. Biomed Pharmacother 2021; 137: 111404-111414.
14. Liu YW, Huang YT. Inhibitory effect of tanshinone IIA on rat hepatic stellate cells. PLoS One 2014; 9: e103229-103239.
15. Pan TL, Wang PW. Explore the molecular mechanism of apoptosis induced by tanshinone IIA on activated rat hepatic stellate cells. Evid Based Complement Alternat Med 2012; 2012: 734987.
16. Li Q, Huang D, Liao W, Su X, Li J, Zhang J, et al. Tanshinone IIA regulates CCl(4) induced liver fibrosis in C57BL/6J mice via the PI3K/Akt and Nrf2/HO-1 signaling pathways. J Biochem Mol Toxicol 2024; 38: e23648.
17. Wu M, Li H, Zhai R, Shan B, Guo C, Chen J. Tanshinone IIA positively regulates the Keap1-Nrf2 system to alleviate pulmonary fibrosis via the sestrin2-sqstm1 signaling axis-mediated autophagy. Phytomedicine 2024; 129: 155620.
18. Li H, Wu M, Guo C, Zhai R, Chen J. Tanshinone IIA regulates Keap1/Nrf2 signal pathway by activating Sestrin2 to restrain pulmonary fibrosis. Am J Chin Med 2022; 50: 2125-2151.
19. Shi MJ, Yan XL, Dong BS, Yang WN, Su SB, Zhang H. A network pharmacology approach to investigating the mechanism of Tanshinone IIA for the treatment of liver fibrosis. J Ethnopharmacol 2020; 253: 112689.
20. Wu R, Dong S, Cai FF, Chen XL, Yang MD, Liu P, et al. Active compounds derived from fuzheng huayu formula protect hepatic parenchymal cells from apoptosis based on network pharmacology and transcriptomic analysis. Molecules 2019; 24: 338-352.
21. Che XH, Park EJ, Zhao YZ, Kim WH, Sohn DH. Tanshinone II A induces apoptosis and S phase cell cycle arrest in activated rat hepatic stellate cells. Basic Clin Pharmacol Toxicol 2010; 106: 30-37.
22. Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol 2019; 70: 151-171.
23. Roehlen N, Crouchet E, Baumert TF. Liver fibrosis: Mechanistic concepts and therapeutic perspectives. Cells 2020; 9: 875-918.
24. Shen M, Cai LL, Qian L. Research progress of systems biology in anti-hepatic fibrosis of traditional Chinese medicine components. China Med Pharm 2024; 14: 21-25.
25. Yang LX, Luo M, Li SY. Tanshinone IIA improves Alzheimer’s disease via RNA nuclear-enriched abundant transcript 1/microRNA-291a-3p/member RAS oncogene family Rab22a axis. World J Psychiatry 2024; 14: 563-581.
26. Zhai P, Chen Q, Wang X, Ouyang X, Yang M, Dong Y, et al. The combination of Tanshinone IIA and Astragaloside IV attenuates myocardial ischemia-reperfusion injury by inhibiting the STING pathway. Chin Med 2024; 19: 34-56.
27. Zhang C, Wang Y, Zhang X, Zhang K, Chen F, Fan J, et al. Maintaining the mitochondrial quality control system was a key event of tanshinone IIA against deoxynivalenol-induced intestinal toxicity. Antioxidants (Basel) 2024; 13: 121-134.
28. Li H, Hu P, Zou Y, Yuan L, Xu Y, Zhang X, et al. Tanshinone IIA and hepatocellular carcinoma: A potential therapeutic drug. Front Oncol 2023; 13: 1071415-1071436.
29. Shi MJ, Dong BS, Yang WN, Su SB, Zhang H. Preventive and therapeutic role of Tanshinone ⅡA in hepatology. Biomed Pharmacother 2019; 112: 108676-108686.
30. Chen LF, Zhao Y, Yang YX, Chen X, Lu JJ. Progress of anti-hepatic fibrosis therapy targeting hepatic stellate cells. Systems Med 2024; 9:189-193.
31. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021; 18:151-166.
32. Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021; 18:151-166.
33. Eom YW, Shim KY, Baik SK. Mesenchymal stem cell therapy for liver fibrosis. Korean J Intern Med 2015; 30: 580-589.
34. Xu L, Zhang Y, Ji N, Du Y, Jia T, Wei S, et al. Tanshinone IIA regulates the TGF-β1/Smad signaling pathway to ameliorate non-alcoholic steatohepatitis-related fibrosis. Exp Ther Med 2022; 24: 486-497.
35. Liu N, Feng J, Lu X, Yao Z, Liu Q, Lv Y, et al. Isorhamnetin inhibits liver fibrosis by reducing autophagy and inhibiting extracellular matrix formation via the TGF-β1/Smad3 and TGF-β1/p38 MAPK pathways. Mediators Inflamm 2019; 2019: 6175091-6175105.
36. Li X, Yao Y, Wei L. Indirubin alleviates CCl(4)-induced liver fibrosis by regulation of TGF-β-mediated signaling pathways. Iran J Basic Med Sci 2023; 26: 1047-1052.
37. Xu F, Liu C, Zhou D, Zhang L. TGF-β/SMAD pathway and its regulation in hepatic fibrosis. J Histochem Cytochem 2016; 64: 157-167.
38. Li Q, Liu G, Yuan H, Wang J, Guo Y, Chen T, et al. Mucin1 shifts Smad3 signaling from the tumor-suppressive pSmad3C/p21(WAF1) pathway to the oncogenic pSmad3L/c-Myc pathway by activating JNK in human hepatocellular carcinoma cells. Oncotarget 2015; 6: 4253-4265.
39. Gong Y, Yang Y. Activation of Nrf2/AREs-mediated antioxidant signalling, and suppression of profibrotic TGF-β1/Smad3 pathway: A promising therapeutic strategy for hepatic fibrosis - A review. Life Sci 2020; 256: 117909.
40. Jiang N, Zhang J, Ping J, Xu L. Salvianolic acid B inhibits autophagy and activation of hepatic stellate cells induced by TGF-β1 by downregulating the MAPK pathway. Front Pharmacol 2022; 13: 938856-938867.
41. Ramos-Tovar E, Buendia-Montaño LD, Galindo-Gómez S, Hernández-Aquino E, Tsutsumi V, Muriel P. Stevia prevents experimental cirrhosis by reducing hepatic myofibroblasts and modulating molecular profibrotic pathways. Hepatol Res 2019; 49: 212-223.
42. Lu K, Zhang M, Wei G, Xiao G, Tong L, Chen D. Multiple cullin-associated E3 ligases regulate cyclin D1 protein stability. Elife 2023; 12: e80327-80337.
43. Wang C, Zhao N, Zheng Q, Zhang D, Liu Y. BHLHE41 promotes U87 and U251 cell proliferation via ERK/cyclinD1 signaling pathway. Cancer Manag Res 2019; 11: 7657-7672.
44. Chen CA, Chang JM, Yang YL, Chang EE, Chen HC. Macrophage migration inhibitory factor regulates integrin-β1 and cyclin D1 expression via ERK pathway in podocytes. Biomed Pharmacother 2020; 124: 109892-109898.
45. Liu F. Smad3 phosphorylation by cyclin-dependent kinases. Cytokine Growth Factor Rev 2006; 17: 9-17.
46. Rezaei HB, Kamato D, Ansari G, Osman N, Little PJ. Cell biology of Smad2/3 linker region phosphorylation in vascular smooth muscle. Clin Exp Pharmacol Physiol 2012; 39: 661-667.