DOI QR코드

DOI QR Code

Cardioprotective effect of ginsenoside Rb1 via regulating metabolomics profiling and AMP-activated protein kinase-dependent mitophagy

  • Hu, Jingui (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Zhang, Ling (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Fu, Fei (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Lai, Qiong (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Zhang, Lu (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Liu, Tao (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Yu, Boyang (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Kou, Junping (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University) ;
  • Li, Fang (Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University)
  • Received : 2021.04.05
  • Accepted : 2021.06.21
  • Published : 2022.03.01

Abstract

Background: Ginsenoside Rb1, a bioactive component isolated from the Panax ginseng, acts as a remedy to prevent myocardial injury. However, it is obscure whether the cardioprotective functions of Rb1 are related to the regulation of endogenous metabolites, and its potential molecular mechanism still needs further clarification, especially from a comprehensive metabolomics profiling perspective. Methods: The mice model of acute myocardial ischemia (AMI) and oxygen glucose deprivation (OGD)-induced cardiomyocytes injury were applied to explore the protective effect and mechanism of Rb1. Meanwhile, the comprehensive metabolomics profiling was conducted by high-performance liquid chromatography and quadrupole time-of-flight mass spectrometry (HPLC-Q/TOF-MS) and a tandem liquid chromatography and mass spectrometry (LC-MS). Results: Rb1 treatment profoundly reduced the infarct size and attenuated myocardial injury. The metabolic network map of 65 differential endogenous metabolites was constructed and provided a new inspiration for the treatment of AMI by Rb1, which was mainly associated with mitophagy. In vivo and in vitro experiments, Rb1 was found to improve mitochondrial morphology, mitochondrial function and promote mitophagy. Interestingly, the mitophagy inhibitor partly attenuated the cardioprotective effect of Rb1. Additionally, Rb1 markedly facilitated the phosphorylation of AMP-activated protein kinase α (AMPKα), and AMPK inhibition partially weakened the role of Rb1 in promoting mitophagy. Conclusions: Ginsenoside Rb1 protects acute myocardial ischemia injury through promoting mitophagy via AMPKα phosphorylation, which might lay the foundation for the further application of Rb1 in cardiovascular diseases.

Keywords

Acknowledgement

This research work was supported by the National Science Foundation of China (No. 81973506, No. 81603328, No. 81774150, No. 81573719), Natural Science Foundation of Jiangsu Province (BK20160761). Project funded by China Postdoctoral Science Foundation (2016M600456, 2017T100425), and supported by Double First-Class University project (CPU2018GF06, CPU2018GF07).

References

  1. Shimokawa H, Yasuda S. Myocardial ischemia: current concepts and future perspectives. J Cardiol 2008;52:67-78. https://doi.org/10.1016/j.jjcc.2008.07.016
  2. Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nature Reviews. Molecular Cell Biology 2018;19:121-35. https://doi.org/10.1038/nrm.2017.95
  3. Hernandez-Resendiz S, Prunier F, Girao H, Dorn G, Hausenloy DJ. Targeting mitochondrial fusion and fission proteins for cardioprotection. J Cell Mol Med 2020;24:6571-85. https://doi.org/10.1111/jcmm.15384
  4. Chistiakov DA, Shkurat TP, Melnichenko AA, Grechko AV, Orekhov AN. The role of mitochondrial dysfunction in cardiovascular disease: a brief review. Ann Med 2018;50:121-7. https://doi.org/10.1080/07853890.2017.1417631
  5. Xu Y, Tang Y, Lu J, Zhang W, Zhu Y, Zhang S, Ma G, Jiang P, Zhang W. PINK1-mediated mitophagy protects against hepatic ischemia/reperfusion injury by restraining NLRP3 inflammasome activation. Free Radic Biol Med 2020;160:871-86. https://doi.org/10.1016/j.freeradbiomed.2020.09.015
  6. Lan R, Wu JT, Wu T, Ma YZ, Wang BQ, Zheng HZ, Li YN, Wang Y, Gu CQ, Zhang Y. Mitophagy is activated in brain damage induced by cerebral ischemia and reperfusion via the PINK1/Parkin/p62 signalling pathway. Brain Res Bull 2018;142:63-77. https://doi.org/10.1016/j.brainresbull.2018.06.018
  7. Tang C, Han H, Yan M, Zhu S, Liu J, Liu Z, He L, Tan J, Liu Y, Liu H, et al. PINK1-PRKN/PARK2 pathway of mitophagy is activated to protect against renal ischemia-reperfusion injury. Autophagy 2018;14:880-97. https://doi.org/10.1080/15548627.2017.1405880
  8. Zhang W, Siraj S, Zhang R, Chen Q. Mitophagy receptor FUNDC1 regulates mitochondrial homeostasis and protects the heart from I/R injury. Autophagy 2017;13:1080-1. https://doi.org/10.1080/15548627.2017.1300224
  9. Seabright AP, Lai Y-C. Regulatory roles of PINK1-Parkin and AMPK in ubiquitin-dependent skeletal muscle mitophagy. Frontiers in Physiology 2020;11:608474. https://doi.org/10.3389/fphys.2020.608474
  10. Zheng M, Xin Y, Li Y, Xu F, Xi X, Guo H, Cui X, Cao H, Zhang X, Han C. Ginsenosides: a potential neuroprotective agent. Biomed Res Int 2018:8174345. https://doi.org/10.1155/2018/8174345
  11. Zheng Q, Bao XY, Zhu PC, Tong Q, Zheng GQ, Wang Y. Ginsenoside Rb1 for myocardial ischemia/reperfusion injury: preclinical evidence and possible mechanisms. Oxid. Med. Cell. Longev. 2017:6313625.
  12. Li G, Qian W, Zhao C. Analyzing the anti-ischemia-reperfusion injury effects of ginsenoside Rb1 mediated through the inhibition of p38α MAPK. Can. J. Physiol. Pharmacol. 2016;94:97-103. https://doi.org/10.1139/cjpp-2014-0164
  13. Cui YC, Pan CS, Yan L, Li L, Hu BH, Chang X, Liu YY, Fan JY, Sun K, Li Q, et al. Ginsenoside Rb1 protects against ischemia/reperfusion-induced myocardial injury via energy metabolism regulation mediated by RhoA signaling pathway. Sci. Rep. 2017;7:44579. https://doi.org/10.1038/srep44579
  14. Newgard CB. Metabolomics and Metabolic Diseases: where do we stand? Cell Metab 2017;25:43-56. https://doi.org/10.1016/j.cmet.2016.09.018
  15. Zhang L, Wei TT, Li Y, Li J, Fan Y, Huang FQ, Cai YY, Ma G, Liu JF, Chen QQ, et al. Functional metabolomics characterizes a key role for N-acetylneuraminic acid in coronary artery diseases. Circulation 2018;137:1374-90. https://doi.org/10.1161/CIRCULATIONAHA.117.031139
  16. Lai Q, Yuan GY, Wang H, Liu ZL, Kou JP, Yu BY, Li F. Exploring the protective effects of schizandrol A in acute myocardial ischemia mice by comprehensive metabolomics profiling integrated with molecular mechanism studies. Acta Pharmacol. Sin. 2020;41:1058-72. https://doi.org/10.1038/s41401-020-0377-7
  17. Li F, Fan X, Zhang Y, Pang L, Ma X, Song M, Kou J, Yu B. Cardioprotection by combination of three compounds from ShengMai preparations in mice with myocardial ischemia/reperfusion injury through AMPK activation-mediated mitochondrial fission. Sci. Rep. 2016;6:37114. https://doi.org/10.1038/srep37114
  18. Fan HJ, Tan ZB, Wu YT, Feng XR, Bi YM, Xie LP, Zhang WT, Ming Z, Liu B, Zhou YC. The role of ginsenoside Rb1, a potential natural glutathione reductase agonist, in preventing oxidative stress-induced apoptosis of H9C2 cells. J Ginseng Res 2020;44:258-66. https://doi.org/10.1016/j.jgr.2018.12.004
  19. Jiang L, Yin X, Chen YH, Chen Y, Jiang W, Zheng H, Huang FQ, Liu B, Zhou W, Qi LW, et al. Proteomic analysis reveals ginsenoside Rb1 attenuates myocardial ischemia/reperfusion injury through inhibiting ROS production from mitochondrial complex I. Theranostics 2021;11:1703-20. https://doi.org/10.7150/thno.43895
  20. Qin L, Wang J, Zhao R, Zhang X, Mei Y. Ginsenoside-Rb1 improved diabetic cardiomyopathy through regulating calcium signaling by alleviating protein O-GlcNAcylation. J Agric Food Chem 2019;67:14074-85. https://doi.org/10.1021/acs.jafc.9b05706
  21. Zheng Q, Bao XY, Zhu PC, Tong Q, Zheng GQ, Wang Y. Ginsenoside Rb1 for myocardial ischemia/reperfusion injury: preclinical evidence and possible mechanisms. Oxid Med Cell Longev 2017:6313625.
  22. Omori K, Katakami N, Yamamoto Y, Ninomiya H, Takahara M, Matsuoka TA, Bamba T, Fukusaki E, Shimomura I. Identification of metabolites associated with onset of CAD in diabetic patients using CE-MS analysis: a pilot study. J Atheroscler Thromb 2019;26:233-45. https://doi.org/10.5551/jat.42945
  23. Khan A, Choi Y, Back JH, Lee S, Jee SH, Park YH. High-resolution metabolomics study revealing l-homocysteine sulfinic acid, cysteic acid, and carnitine as novel biomarkers for high acute myocardial infarction risk. Metabolism 2020;104:154051. https://doi.org/10.1016/j.metabol.2019.154051
  24. Lee Y, Khan A, Hong S, Jee SH, Park YH. A metabolomic study on high-risk stroke patients determines low levels of serum lysine metabolites: a retrospective cohort study. Mol Biosyst 2017;13:1109-20. https://doi.org/10.1039/c6mb00732e
  25. Soares MSP, da Silveira de Mattos B, Avila AA, Spohr L, Pedra NS, Teixeira FC, Bona NP, Oliveira PS, Stefanello FM, Spanevello RM. High levels of methionine and methionine sulfoxide: impact on adenine nucleotide hydrolysis and redox status in platelets and serum of young rats. J Cell Biochem 2018;120:2289-303. https://doi.org/10.1002/jcb.27554
  26. Sibilitz KL, Benn M, Nordestgaard BG. Creatinine, eGFR and association with myocardial infarction, ischemic heart disease and early death in the general population. Atherosclerosis 2014;237:67-75. https://doi.org/10.1016/j.atherosclerosis.2014.08.040
  27. Salabei JK, Lorkiewicz PK, Holden CR, Li Q, Hong KU, Bolli R, Bhatnagar A, Hill BG. Glutamine regulates cardiac progenitor cell metabolism and proliferation. Stem Cells 2015;33:2613-27. https://doi.org/10.1002/stem.2047
  28. Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pharmacology of adenosine receptors: the state of the art. Physiol Rev 2018;98:1591-625. https://doi.org/10.1152/physrev.00049.2017
  29. Schaffer SW, Shimada-Takaura K, Jong CJ, Ito T, Takahashi K. Impaired energy metabolism of the taurine-deficient heart. Amino Acids 2016;48:549-58. https://doi.org/10.1007/s00726-015-2110-2
  30. Jong CJ, Ito T, Schaffer SW. The ubiquitin-proteasome system and autophagy are defective in the taurine-deficient heart. Amino Acids 2015;47:2609-22. https://doi.org/10.1007/s00726-015-2053-7
  31. Oudman I, Clark JF, Brewster LM. The effect of the creatine analogue beta-guanidinopropionic acid on energy metabolism: a systematic review. PLoS One 2013;8:e52879. https://doi.org/10.1371/journal.pone.0052879
  32. Crocker CL, Baumgarner BL, Kinsey ST. β-guanidinopropionic acid and metformin differentially impact autophagy, mitochondria and cellular morphology in developing C2C12 muscle cells. J Muscle Res Cell Motil 2020;41:221-37. https://doi.org/10.1007/s10974-019-09568-0
  33. Saha AK, Xu XJ, Lawson E, Deoliveira R, Brandon AE, Kraegen EW, Ruderman NB. Downregulation of AMPK accompanies leucine- and glucose-induced increases in protein synthesis and insulin resistance in rat skeletal muscle. Diabetes 2010;59:2426-34. https://doi.org/10.2337/db09-1870
  34. Bizzarri M, Fuso A, Dinicola S, Cucina A, Bevilacqua A. Pharmacodynamics and pharmacokinetics of Inositol(s) in health and disease. Expert Opin Drug Metab Toxicol 2016;12:1181-96. https://doi.org/10.1080/17425255.2016.1206887
  35. Cai CC, Zhu JH, Ye LX, Dai YY, Fang MC, Hu YY, Pan SL, Chen S, Li PJ, Fu XQ, et al. Glycine protects against hypoxic-ischemic brain injury by regulating mitochondria-mediated autophagy via the AMPK pathway. Oxid Med Cell Longev 2019:4248529. https://doi.org/10.1155/2019/4248529
  36. Zhang Y, Su W, Zhang Q, Xu J, Liu H, Luo J, Zhan L, Xia Z, Lei S. Glycine protects H9C2 cardiomyocytes from high glucose- and hypoxia/reoxygenation-induced injury via inhibiting PKCβ2 activation and improving mitochondrial quality. J Diabetes Res 2018:9502895. https://doi.org/10.1155/2018/9502895
  37. Dhar I, Lysne V, Seifert R, Svingen GFT, Ueland PM, Nygard OK. Plasma methionine and risk of acute myocardial infarction: effect modification by established risk factors. Atherosclerosis 2018;272:175-81. https://doi.org/10.1016/j.atherosclerosis.2018.03.038
  38. Plummer JD, Johnson JE. Extension of cellular lifespan by methionine restriction involves alterations in central carbon metabolism and is mitophagy-dependent. Front Cell Dev Biol 2019;7:301. https://doi.org/10.3389/fcell.2019.00301
  39. Kulek AR, Anzell A, Wider JM, Sanderson TH, Przyklenk K. Mitochondrial quality control: role in cardiac models of lethal ischemia-reperfusion injury. Cells 2020;9:214. https://doi.org/10.3390/cells9010214
  40. Johansen T, Lamark T. Selective autophagy mediated by autophagic adapter proteins. Autophagy 2011;7:279-96. https://doi.org/10.4161/auto.7.3.14487
  41. Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ 2013;20:31-42. https://doi.org/10.1038/cdd.2012.81
  42. Kubli DA, Zhang X, Lee Y, Hanna RA, Quinsay MN, Nguyen CK, Jimenez R, Petrosyan S, Murphy AN, Gustafsson AB. Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J Biol Chem 2013;288:915-26. https://doi.org/10.1074/jbc.M112.411363
  43. Lee Y, Lee HY, Hanna RA, Gustafsson AB. Mitochondrial autophagy by Bnip3 involves Drp1-mediated mitochondrial fission and recruitment of Parkin in cardiac myocytes. Am J Physiol Heart Circ Physiol 2011;301:1924-31.
  44. Billia F, Hauck L, Konecny F, Rao V, Shen J, Mak TW. PTEN-inducible kinase 1 (PINK1)/Park6 is indispensable for normal heart function. Proc Natl Acad Sci U S A 2011;108:9572-7. https://doi.org/10.1073/pnas.1106291108
  45. Herzig S, Shaw RJ. AMPK: guardian of metabolism and mitochondrial homeostasis. Nat. Rev. Mol. Cell Biol. 2018;19:121-35. https://doi.org/10.1038/nrm.2017.95
  46. Zhang H, Liu B, Li T, Zhu Y, Luo G, Jiang Y, Tang F, Jian Z, Xiao Y. AMPK activation serves a critical role in mitochondria quality control via modulating mitophagy in the heart under chronic hypoxia. Int J Mol Med 2018;41:69-76.
  47. Tamargo-Gomez I, Marino G. AMPK: regulation of metabolic dynamics in the context of autophagy. Int J Mol Sci 2018;19:3812. https://doi.org/10.3390/ijms19123812