DOI QR코드

DOI QR Code

Ginsenoside Re inhibits myocardial fibrosis by regulating miR-489/myd88/NF-κB pathway

  • Jinghui, Sun (National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences) ;
  • Ru, Wang (National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences) ;
  • Tiantian, Chao (National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences) ;
  • Jun, Peng (Institute of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine) ;
  • Chenglong, Wang (National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences) ;
  • Keji, Chen (National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences)
  • 투고 : 2021.08.26
  • 심사 : 2021.11.29
  • 발행 : 2023.03.02

초록

Background: Myocardial fibrosis (MF) is an advanced pathological manifestation of many cardiovascular diseases, which can induce heart failure and malignant arrhythmias. However, the current treatment of MF lacks specific drugs. Ginsenoside Re has anti-MF effect in rat, but its mechanism is still not clear. Therefore, we investigated the anti-MF effect of ginsenoside Re by constructing mouse acute myocardial infarction (AMI) model and AngII induced cardiac fibroblasts (CFs) model. Methods: The anti-MF effect of miR-489 was investigated by transfection of miR-489 mimic and inhibitor in CFs. Effect of ginsenoside Re on MF and its related mechanisms were investigated by ultrasonographic, ELISA, histopathologic staining, transwell test, immunofluorescence, Western blot and qPCR in the mouse model of AMI and the AngII-induced CFs model. Results: MiR-489 decreased the expression of α-SMA, collagenI, collagen III and myd88, and inhibited the phosphorylation of NF-κB p65 in normal CFs and CFs treated with AngII. Ginsenoside Re could improve cardiac function, inhibit collagen deposition and CFs migration, promote the transcription of miR-489, and reduce the expression of myd88 and the phosphorylation of NF-κB p65. Conclusion: MiR-489 can effectively inhibit the pathological process of MF, and the mechanism is at least partly related to the regulation of myd88/NF-κB pathway. Ginsenoside Re can ameliorate AMI and AngII induced MF, and the mechanism is at least partially related to the regulation of miR-489/myd88/NF-κB signaling pathway. Therefore, miR-489 may be a potential target of anti-MF and ginsenoside Re may be an effective drug for the treatment of MF.

키워드

과제정보

This work was supported by the National Natural Science Foundation of China (grant number 81874410); The authors thank the Institute of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine for providing technical help.

참고문헌

  1. Tschope C, Diez J. Myocardial fibrosis as a matter of cell differentiation: opportunities for new antifibrotic strategies. Eur Heart J 2019;40:979-81.  https://doi.org/10.1093/eurheartj/ehy307
  2. Thum T. Noncoding RNAs and myocardial fibrosis. Nat Rev Cardiol 2014;11:655-63.  https://doi.org/10.1038/nrcardio.2014.125
  3. Pan SC, Cui HH, Qiu CG. HOTAIR promotes myocardial fibrosis through regulating URI1 expression via Wnt pathway. Eur Rev Med Pharmacol Sci 2018;22:6983-90. 
  4. Tallquist MD, Molkentin JD. Redefining the identity of cardiac fibroblasts. Nat Rev Cardiol 2017;14:484-91.  https://doi.org/10.1038/nrcardio.2017.57
  5. Frangogiannis NG. Can myocardial fibrosis Be reversed? J Am Coll Cardiol 2019;73:2283-5.  https://doi.org/10.1016/j.jacc.2018.10.094
  6. Rodriguez P, Sassi Y, Troncone L, Benard L, Ishikawa K, Gordon RE, Lamas S, Laborda J, Hajjar RJ, Lebeche D. Deletion of delta-like 1 homologue accelerates fibroblast-myofibroblast differentiation and induces myocardial fibrosis. Eur Heart J 2019;40:967-78.  https://doi.org/10.1093/eurheartj/ehy188
  7. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res 2016;119:91-112.  https://doi.org/10.1161/CIRCRESAHA.116.303577
  8. Travers JG, Kamal FA, Robbins J, Yutzey KE, Blaxall BC. Cardiac fibrosis: the fibroblast awakens. Circ Res 2016;118:1021-40.  https://doi.org/10.1161/CIRCRESAHA.115.306565
  9. van den Borne SW, Diez J, Blankesteijn WM, Verjans J, Hofstra L, Narula J. Myocardial remodeling after infarction: the role of myofibroblasts. Nat Rev Cardiol 2010;7:30-7.  https://doi.org/10.1038/nrcardio.2009.199
  10. Daniels A, van Bilsen M, Goldschmeding R, van der Vusse GJ, van Nieuwenhoven FA. Connective tissue growth factor and cardiac fibrosis. Acta Physiol 2009;195:321-38.  https://doi.org/10.1111/j.1748-1716.2008.01936.x
  11. Martos R, Baugh J, Ledwidge M, O'Loughlin C, Conlon C, Patle A, Donnelly SC, McDonald K. Diastolic heart failure: evidence of increased myocardial collagen turnover linked to diastolic dysfunction. Circulation 2007;115:888-95.  https://doi.org/10.1161/CIRCULATIONAHA.106.638569
  12. Yong KW, Li Y, Huang G, Lu TJ, Safwani WK, Pingguan-Murphy B, Xu F. Mechanoregulation of cardiac myofibroblast differentiation: implications for cardiac fibrosis and therapy. Am J Physiol Heart Circ Physiol 2015;309:H532-42.  https://doi.org/10.1152/ajpheart.00299.2015
  13. Dean RG, Balding LC, Candido R, Burns WC, Cao Z, Twigg SM, Burrell LM. Connective tissue growth factor and cardiac fibrosis after myocardial infarction. J Histochem Cytochem 2005;53:1245-56.  https://doi.org/10.1369/jhc.4A6560.2005
  14. Schirone L, Forte M, Palmerio S, Yee D, Nocella C, Angelini F, Pagano F, Schiavon S, Bordin A, Carrizzo A, et al. A review of the molecular mechanisms underlying the development and progression of cardiac remodeling. Oxid Med Cell Longev 2017;2017:3920195. 
  15. Liu WY, Sun HH, Sun PF. MicroRNA-378 attenuates myocardial fibrosis by inhibiting MAPK/ERK pathway. Eur Rev Med Pharmacol Sci 2019;23:4398-405. 
  16. Mallory AC, Vaucheret H. MicroRNAs: something important between the genes. Curr Opin Plant Biol 2004;7:120-5.  https://doi.org/10.1016/j.pbi.2004.01.006
  17. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281-97.  https://doi.org/10.1016/S0092-8674(04)00045-5
  18. Liu X, Luo G, Bai X, Wang XJ. Bioinformatic analysis of microRNA biogenesis and function related proteins in eleven animal genomes. J Genet Genomics 2009;36:591-601.  https://doi.org/10.1016/S1673-8527(08)60151-4
  19. Yang KC, Yamada KA, Patel AY, Topkara VK, George I, Cheema FH, Ewald GA, Mann DL, Nerbonne JM. Deep RNA sequencing reveals dynamic regulation of myocardial noncoding RNAs in failing human heart and remodeling with mechanical circulatory support. Circulation 2014;129:1009-21.  https://doi.org/10.1161/CIRCULATIONAHA.113.003863
  20. Zhou C, Cui Q, Su G, Guo X, Liu X, Zhang J. MicroRNA-208b alleviates post-infarction myocardial fibrosis in a rat model by inhibiting GATA4. Med Sci Mon Int Med J Exp Clin Res 2016;22:1808-16.  https://doi.org/10.12659/MSM.896428
  21. Chen MH, Liu JC, Liu Y, Hu YC, Cai XF, Yin DC. MicroRNA-199a regulates myocardial fibrosis in rats by targeting SFRP5. Eur Rev Med Pharmacol Sci 2019;23:3976-83. 
  22. Kikkawa N, Hanazawa T, Fujimura L, Nohata N, Suzuki H, Chazono H, Sakurai D, Horiguchi S, Okamoto Y, Seki N. miR-489 is a tumour-suppressive miRNA target PTPN11 in hypopharyngeal squamous cell carcinoma (HSCC). Br J Cancer 2010;103:877-84.  https://doi.org/10.1038/sj.bjc.6605811
  23. Zhang B, Ji S, Ma F, Ma Q, Lu X, Chen X. miR-489 acts as a tumor suppressor in human gastric cancer by targeting PROX1. Am J Cancer Res 2016;6:2021-30. 
  24. Li J, Qu W, Jiang Y, Sun Y, Cheng Y, Zou T, Du S. miR-489 suppresses proliferation and invasion of human bladder cancer cells. Oncol Res 2016;24:391-8.  https://doi.org/10.3727/096504016X14666990347518
  25. Li Y, Ma X, Wang Y, Li G. miR-489 inhibits proliferation, cell cycle progression and induces apoptosis of glioma cells via targeting SPIN1-mediated PI3K/AKT pathway. Biomed Pharmacother 2017;93:435-43.  https://doi.org/10.1016/j.biopha.2017.06.058
  26. Wu Q, Han L, Yan W, Ji X, Han R, Yang J, Yuan J, Ni C. miR-489 inhibits silica-induced pulmonary fibrosis by targeting MyD88 and Smad3 and is negatively regulated by lncRNA CHRF. Sci Rep 2016;6:30921. 
  27. Feng Y, Zou L, Si R, Nagasaka Y, Chao W. Bone marrow MyD88 signaling modulates neutrophil function and ischemic myocardial injury. Am J Physiol Cell Physiol 2010;299:C760-9.  https://doi.org/10.1152/ajpcell.00155.2010
  28. Li Y, Si R, Feng Y, Chen HH, Zou L, Wang E, Zhang M, Warren HS, Sosnovik DE, Chao W. Myocardial ischemia activates an injurious innate immune signaling via cardiac heat shock protein 60 and Toll-like receptor 4. J Biol Chem 2011;286:31308-19.  https://doi.org/10.1074/jbc.M111.246124
  29. Li T, Wang Y, Liu C, Hu Y, Wu M, Li J, Guo L, Chen L, Chen Q, Ha T, et al. MyD88-dependent nuclear factor-kappaB activation is involved in fibrinogen-induced hypertrophic response of cardiomyocytes. J Hypertens 2009;27:1084-93.  https://doi.org/10.1097/HJH.0b013e3283293c93
  30. Wendlandt EB, Graff JW, Gioannini TL, McCaffrey AP, Wilson ME. The role of microRNAs miR-200b and miR-200c in TLR4 signaling and NF-kappaB activation. Innate Immun 2012;18:846-55.  https://doi.org/10.1177/1753425912443903
  31. Mack M. Inflammation and fibrosis. Matrix Biol 2018;68-69:106-21.  https://doi.org/10.1016/j.matbio.2017.11.010
  32. Wang K, Liu F, Zhou LY, Long B, Yuan SM, Wang Y, Liu CY, Sun T, Zhang XJ, Li PF. The long noncoding RNA CHRF regulates cardiac hypertrophy by targeting miR-489. Circ Res 2014;114:1377-88.  https://doi.org/10.1161/CIRCRESAHA.114.302476
  33. Zheng SD, Wu HJ, Wu DL. Roles and mechanisms of ginseng in protecting heart. Chin J Integr Med 2012;18:548-55.  https://doi.org/10.1007/s11655-012-1148-1
  34. Xie JT, Wang CZ, Wang AB, Wu J, Basila D, Yuan CS. Antihyperglycemic effects of total ginsenosides from leaves and stem of Panax ginseng. Acta Pharmacol Sin 2005;26:1104-10.  https://doi.org/10.1111/j.1745-7254.2005.00156.x
  35. Joo KM, Lee JH, Jeon HY, Park CW, Hong DK, Jeong HJ, Lee SJ, Lee SY, Lim KM. Pharmacokinetic study of ginsenoside Re with pure ginsenoside Re and ginseng berry extracts in mouse using ultra performance liquid chromatography/mass spectrometric method. J Pharmaceut Biomed Anal 2010;51:278-83.  https://doi.org/10.1016/j.jpba.2009.08.013
  36. Lee GH, Lee WJ, Hur J, Kim E, Lee HG, Seo HG. Ginsenoside Re mitigates 6-hydroxydopamine-induced oxidative stress through upregulation of GPX4. Molecules 2020;25. 
  37. Gao Y, Zhu P, Xu SF, Li YQ, Deng J, Yang DL. Ginsenoside Re inhibits PDGF-BB-induced VSMC proliferation via the eNOS/NO/cGMP pathway. Biomed Pharmacother 2019;115:108934. 
  38. Cai M, Yang EJ. Ginsenoside Re attenuates neuroinflammation in a symptomatic ALS animal model. Am J Chin Med 2016;44:401-13.  https://doi.org/10.1142/s0192415x16500233
  39. Peng L, Sun S, Xie LH, Wicks SM, Xie JT. Ginsenoside Re: pharmacological effects on cardiovascular system. Cardiovasc Ther 2012;30:e183-8.  https://doi.org/10.1111/j.1755-5922.2011.00271.x
  40. Wang QW, Yu XF, Xu HL, Zhao XZ, Sui DY. Ginsenoside Re improves isoproterenol-induced myocardial fibrosis and heart failure in rats. Evid Based Complement Alternat Med 2019;2019:3714508. 
  41. Yu Y, Sun J, Liu J, Wang P, Wang C. Ginsenoside Re preserves cardiac function and ameliorates left ventricular remodeling in a rat model of myocardial infarction. J Cardiovasc Pharmacol 2020;75:91-7.  https://doi.org/10.1097/fjc.0000000000000752
  42. Huang DD, Huang HF, Yang Q, Chen XQ. Liraglutide improves myocardial fibrosis after myocardial infarction through inhibition of CTGF by activating cAMP in mice. Eur Rev Med Pharmacol Sci 2018;22:4648-56. 
  43. V H, Titus AS, Cowling RT, Kailasam S. Collagen receptor cross-talk determines α-smooth muscle actin-dependent collagen gene expression in angiotensin II-stimulated cardiac fibroblasts. J Biol Chem 2019;294:19723-39.  https://doi.org/10.1074/jbc.RA119.009744
  44. Bowie A, O'Neill LA. Oxidative stress and nuclear factor-kappaB activation: a reassessment of the evidence in the light of recent discoveries. Biochem Pharmacol 2000;59:13-23.  https://doi.org/10.1016/S0006-2952(99)00296-8
  45. Jain M, Rivera S, Monclus EA, Synenki L, Zirk A, Eisenbart J, Feghali-Bostwick C, Mutlu GM, Budinger GR, Chandel NS. Mitochondrial reactive oxygen species regulate transforming growth factor-beta signaling. J Biol Chem 2013;288:770-7.  https://doi.org/10.1074/jbc.M112.431973
  46. Lijnen PJ, Petrov VV, Fagard RH. Induction of cardiac fibrosis by transforming growth factor-beta(1). Mol Genet Metabol 2000;71:418-35.  https://doi.org/10.1006/mgme.2000.3032
  47. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-beta: the master regulator of fibrosis. Nat Rev Nephrol 2016;12:325-38.  https://doi.org/10.1038/nrneph.2016.48
  48. Wang J, Shen W, Zhang JY, Jia CH, Xie ML. Stevioside attenuates isoproterenol-induced mouse myocardial fibrosis through inhibition of the myocardial NF-κB/TGF-β1/Smad signaling pathway. Food Funct 2019;10:1179-90.  https://doi.org/10.1039/c8fo01663a
  49. Chen RC, Wang J, Yang L, Sun GB, Sun XB. Protective effects of ginsenoside Re on lipopolysaccharide-induced cardiac dysfunction in mice. Food Funct 2016;7:2278-87.  https://doi.org/10.1039/C5FO01357G
  50. Lee IA, Hyam SR, Jang SE, Han MJ, Kim DH. Ginsenoside Re ameliorates inflammation by inhibiting the binding of lipopolysaccharide to TLR4 on macrophages. J Agric Food Chem 2012;60:9595-602. https://doi.org/10.1021/jf301372g