Browse > Article
http://dx.doi.org/10.1016/j.jgr.2019.08.006

Ginsenoside compound-Mc1 attenuates oxidative stress and apoptosis in cardiomyocytes through an AMP-activated protein kinase-dependent mechanism  

Hong, So-hyeon (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Hwang, Hwan-Jin (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Kim, Joo Won (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Kim, Jung A. (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Lee, You Bin (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Roh, Eun (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Choi, Kyung Mook (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Baik, Sei Hyun (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Yoo, Hye Jin (Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Korea University)
Publication Information
Journal of Ginseng Research / v.44, no.4, 2020 , pp. 664-671 More about this Journal
Abstract
Background: Ginsenoside compound-Mc1 (Mc1) is a member of the deglycosylated ginsenosides obtained from ginseng extract. Although several ginsenosides have a cardioprotective effect, this has not been demonstrated in ginsenoside Mc1. Methods: We treated H9c2 cells with hydrogen peroxide (H2O2) and ginsenoside Mc1 to evaluate the antioxidant effects of Mc1. The levels of antioxidant molecules, catalase, and superoxide dismutase 2 (SOD2) were measured, and cell viability was determined using the Bcl2-associated X protein (Bax):B-cell lymphoma-extra large ratio, a cytotoxicity assay, and flow cytometry. We generated mice with high-fat diet (HFD)-induced obesity using ginsenoside Mc1 and assessed their heart tissues to evaluate the antioxidant effect and the fibrosis-reducing capability of ginsenoside Mc1. Results: Ginsenoside Mc1 significantly increased the level of phosphorylated AMP-activated protein kinase (AMPK) in the H9c2 cells. The expression levels of catalase and SOD2 increased significantly after treatment with ginsenoside Mc1, resulting in a decrease in the production of H2O2-mediated reactive oxygen species. Treatment with ginsenoside Mc1 also significantly reduced the H2O2-mediated elevation of the Bax:Bcl2 ratio and the number of DNA-damaged cells, which was significantly attenuated by treatment with an AMPK inhibitor. Consistent with the in vitro data, ginsenoside Mc1 upregulated the levels of catalase and SOD2 and decreased the Bax:B-cell lymphoma-extra large ratio and caspase-3 activity in the heart tissues of HFD-induced obese mice, resulting in reduced collagen deposition. Conclusion: Ginsenoside Mc1 decreases oxidative stress and increases cell viability in H9c2 cells and the heart tissue isolated from HFD-fed mice via an AMPK-dependent mechanism, suggesting its potential as a novel therapeutic agent for oxidative stress-related cardiac diseases.
Keywords
AMP-activated protein kinases; Antioxidant; Cardiomyocyte;
Citations & Related Records
Times Cited By KSCI : 2  (Citation Analysis)
연도 인용수 순위
1 Lee HM, Lee OH, Kim KJ, Lee BY. Ginsenoside Rg1 promotes glucose uptake through activated AMPK pathway in insulin-resistant muscle cells. Phytother Res 2012;26:1017-22.   DOI
2 Ko SR, Suzuki Y, Suzuki K, Choi KJ, Cho BG. Marked production of ginsenosides Rd, F2, Rg3, and compound K by enzymatic method. Chem Pharm Bull (Tokyo) 2007;55:1522-7.   DOI
3 Rushworth GF, Megson IL. Existing and potential therapeutic uses for Nacetylcysteine: the need for conversion to intracellular glutathione for antioxidant benefits. Pharmacol Ther 2014;141:150-9.   DOI
4 Guo M, Xiao J, Sheng X, Zhang X, Tie Y, Wang L, Zhao L, Ji X. Ginsenoside Rg3 mitigates atherosclerosis progression in diabetic apoE-/- mice by skewing macrophages to the M2 phenotype. Front Pharmacol 2018;9:464.   DOI
5 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.
6 Gao Y, Chu S, Shao Q, Zhang M, Xia C,Wang Y, Li Y, Lou Y, Huang H, Chen N. Antioxidant activities of ginsenoside Rg1 against cisplatin-induced hepatic injury through Nrf2 signaling pathway in mice. Free Radic Res 2017;51:1-13.   DOI
7 Zhang Y, Zhang Z, Wang H, Cai N, Zhou S, Zhao Y, Chen X, Zheng S, Si Q, Zhang W. Neuroprotective effect of ginsenoside Rg1 prevents cognitive impairment induced by isoflurane anesthesia in aged rats via antioxidant, anti-inflammatory and anti-apoptotic effects mediated by the PI3K/AKT/GSK-3beta pathway. Mol Med Rep 2016;14:2778-84.   DOI
8 Shin KC, Oh DK. Classification of glycosidases that hydrolyze the specific positions and types of sugar moieties in ginsenosides. Crit Rev Biotechnol 2016;36:1036-49.   DOI
9 Cleutjens JP, Verluyten MJ, Smiths JF, Daemen MJ. Collagen remodeling after myocardial infarction in the rat heart. Am J Pathol 1995;147:325-38.
10 Russo I, Frangogiannis NG. Diabetes-associated cardiac fibrosis: cellular effectors, molecular mechanisms and therapeutic opportunities. J Mol Cell Cardiol 2016;90:84-93.   DOI
11 Khan R, Sheppard R. Fibrosis in heart disease: understanding the role of transforming growth factor-beta in cardiomyopathy, valvular disease and arrhythmia. Immunology 2006;118:10-24.   DOI
12 Matsushima S, Kuroda J, Ago T, Zhai P, Park JY, Xie LH, Tian B, Sadoshima J. Increased oxidative stress in the nucleus caused by Nox4 mediates oxidation of HDAC4 and cardiac hypertrophy. Circ Res 2013;112:651-63.   DOI
13 Zhang YJ, Zhang XL, Li MH, Iqbal J, Bourantas CV, Li JJ, Su XY, Muramatsu T, Tian NL, Chen SL. The ginsenoside Rg1 prevents transverse aortic constrictioninduced left ventricular hypertrophy and cardiac dysfunction by inhibiting fibrosis and enhancing angiogenesis. J Cardiovasc Pharmacol 2013;62:50-7.   DOI
14 Li Q, Xiang Y, Chen Y, Tang Y, Zhang Y. Ginsenoside Rg1 protects cardiomyocytes against hypoxia/reoxygenation injury via activation of Nrf2/HO- 1 signaling and inhibition of JNK. Cell Physiol Biochem 2017;44:21-37.   DOI
15 Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, Wu M, Ventre J, Doebber T, Fujii N, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 2001;108:1167-74.   DOI
16 Tsutsui H, Kinugawa S, Matsushima S. Oxidative stress and heart failure. Am J Physiol Heart Circ Physiol 2011;301:H2181-90.   DOI
17 Symons JD, McMillin SL, Riehle C, Tanner J, Palionyte M, Hillas E, Jones D, Cooksey RC, Birnbaum MJ, McClain DA, et al. Contribution of insulin and Akt1 signaling to endothelial nitric oxide synthase in the regulation of endothelial function and blood pressure. Circ Res 2009;104:1085-94.   DOI
18 Wende AR, Symons JD, Abel ED. Mechanisms of lipotoxicity in the cardiovascular system. Curr Hypertens Rep 2012;14:517-31.   DOI
19 Xie JT, Shao ZH, Vanden Hoek TL, Chang WT, Li J, Mehendale S, Wang CZ, Hsu CW, Becker LB, Yin JJ, et al. Antioxidant effects of ginsenoside Re in cardiomyocytes. Eur J Pharmacol 2006;532:201-7.   DOI
20 He F, Guo R, Wu SL, Sun M, Li M. Protective effects of ginsenoside Rb1 on human umbilical vein endothelial cells in vitro. J Cardiovasc Pharmacol 2007;50:314-20.   DOI
21 Li J, Shao ZH, Xie JT, Wang CZ, Ramachandran S, Yin JJ, Aung H, Li CQ, Qin G, Vanden Hoek T, et al. The effects of ginsenoside Rb1 on JNK in oxidative injury in cardiomyocytes. Arch Pharm Res 2012;35:1259-67.   DOI
22 Noh KH, Oh DK. Production of the rare ginsenosides compound K, compound Y, and compound Mc by a thermostable beta-glycosidase from Sulfolobus acidocaldarius. Biol Pharm Bull 2009;32:1830-5.   DOI
23 Xu QF, Fang XL, Chen DF. Pharmacokinetics and bioavailability of ginsenoside Rb1 and Rg1 from Panax notoginseng in rats. J Ethnopharmacol 2003;84:187-92.   DOI
24 Ruderman NB, Carling D, Prentki M, Cacicedo JM. AMPK, insulin resistance, and the metabolic syndrome. J Clin Invest 2013;123:2764-72.   DOI
25 Liu CY, Zhou RX, Sun CK, Jin YH, Yu HS, Zhang TY, Xu LQ, Jin FX. Preparation of minor ginsenosides C-Mc, C-Y, F2, and C-K from American ginseng PPDginsenoside using special ginsenosidase type-I from Aspergillus niger g.848. J Ginseng Res 2015;39:221-9.   DOI
26 Shen L, Xiong Y, Wang DQ, Howles P, Basford JE, Wang J, Xiong YQ, Hui DY, Woods SC, Liu M. Ginsenoside Rb1 reduces fatty liver by activating AMPactivated protein kinase in obese rats. J Lipid Res 2013;54:1430-8.   DOI
27 Huang Q, Wang T, Yang L, Wang HY. Ginsenoside Rb2 alleviates hepatic lipid accumulation by restoring autophagy via induction of Sirt1 and activation of AMPK. Int J Mol Sci 2017;18:E1063.
28 Russell 3rd RR, Li J, Coven DL, Pypaert M, Zechner C, Palmeri M, Giordano FJ, Mu J, Birnbaum MJ, Young LH. AMP-activated protein kinase mediates ischemic glucose uptake and prevents postischemic cardiac dysfunction, apoptosis, and injury. J Clin Invest 2004;114:495-503.   DOI
29 Wang J, Tong C, Yan X, Yeung E, Gandavadi S, Hare AA, Du X, Chen Y, Xiong H, Ma C, et al. Limiting cardiac ischemic injury by pharmacological augmentation of macrophage migration inhibitory factor-AMP-activated protein kinase signal transduction. Circulation 2013;128:225-36.   DOI
30 Carling D. The AMP-activated protein kinase cascade-a unifying system for energy control. Trends Biochem Sci 2004;29:18-24.   DOI
31 Hardie DG, Ross FA, Hawley SA. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat Rev Mol Cell Biol 2012;13:251-62.   DOI
32 Nagendran J, Waller TJ, Dyck JR. AMPK signalling and the control of substrate use in the heart. Mol Cell Endocrinol 2013;366:180-93.   DOI
33 Lee KT, Jung TW, Lee HJ, Kim SG, Shin YS, Whang WK. The antidiabetic effect of ginsenoside Rb2 via activation of AMPK. Arch Pharm Res 2011;34:1201-8.   DOI
34 Leask A. Potential therapeutic targets for cardiac fibrosis: TGFbeta, angiotensin, endothelin, CCN2, and PDGF, partners in fibroblast activation. Circ Res 2010;106:1675-80.   DOI
35 Dzeshka MS, Lip GY, Snezhitskiy V, Shantsila E. Cardiac fibrosis in patients with atrial fibrillation: mechanisms and clinical implications. J Am Coll Cardiol 2015;66:943-59.   DOI
36 Travers JG, Kamal FA, Robbins J, Yutzey KE, Blaxall BC. Cardiac fibrosis: the fibroblast awakens. Circ Res 2016;118:1021-40.   DOI
37 Segura AM, Frazier OH, Buja LM. Fibrosis and heart failure. Heart Fail Rev 2014;19:173-85.   DOI
38 Wold LE, Ceylan-Isik AF, Ren J. Oxidative stress and stress signaling: menace of diabetic cardiomyopathy. Acta Pharmacol Sin 2005;26:908-17.   DOI
39 Sabri A, Hughie HH, Lucchesi PA. Regulation of hypertrophic and apoptotic signaling pathways by reactive oxygen species in cardiac myocytes. Antioxid Redox Signal 2003;5:731-40.   DOI
40 Cesselli D, Jakoniuk I, Barlucchi L, Beltrami AP, Hintze TH, Nadal-Ginard B, Kajstura J, Leri A, Anversa P. Oxidative stress-mediated cardiac cell death is a major determinant of ventricular dysfunction and failure in dog dilated cardiomyopathy. Circ Res 2001;89:279-86.   DOI
41 Gui QF, Xu ZR, Xu KY, Yang YM. The efficacy of ginseng-related therapies in type 2 diabetes mellitus: an updated systematic review and meta-analysis. Medicine (Baltimore) 2016;95. e2584.   DOI
42 Zhang Y, Yu L, Cai W, Fan S, Feng L, Ji G, Huang C. Protopanaxatriol, a novel $PPAR{\gamma}$ antagonist from Panax ginseng, alleviates steatosis in mice. Sci Rep 2014;4:7375.   DOI
43 Kim J-H. Cardiovascular diseases and Panax ginseng: a review on molecular mechanisms and medical applications. J Ginseng Res 2012;36:16-26.   DOI
44 Rastogi V, Santiago-Moreno J, Dore S. Ginseng: a promising neuroprotective strategy in stroke. Front Cell Neurosci 2015;8:457.   DOI
45 Christensen LP. Ginsenosides chemistry, biosynthesis, analysis, and potential health effects. Adv Food Nutr Res 2009;55:1-99.   DOI
46 Zhou W, Chai H, Lin PH, Lumsden AB, Yao Q, Chen C. Ginsenoside Rb1 blocks homocysteine-induced endothelial dysfunction in porcine coronary arteries. J Vasc Surg 2005;41:861-8.   DOI