DOI QR코드

DOI QR Code

miR-23a Regulates Cardiomyocyte Apoptosis by Targeting Manganese Superoxide Dismutase

  • Long, Bo (Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College) ;
  • Gan, Tian-Yi (State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College) ;
  • Zhang, Rong-Cheng (State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College) ;
  • Zhang, Yu-Hui (State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College)
  • Received : 2017.01.18
  • Accepted : 2017.06.25
  • Published : 2017.08.31

Abstract

Cardiomyocyte apoptosis is initiated by various cellular insults and accumulated cardiomyocyte apoptosis leads to the pathogenesis of heart failure. Excessive reactive oxygen species (ROS) provoke apoptotic cascades. Manganese superoxide dismutase (MnSOD) is an important antioxidant enzyme that converts cellular ROS into harmless products. In this study, we demonstrate that MnSOD is down-regulated upon hydrogen peroxide treatment or ischemia/reperfusion (I/R) injury. Enhanced expression of MnSOD attenuates cardiomyocyte apoptosis and myocardial infarction induced by I/R injury. Further, we show that miR-23a directly regulates the expression of MnSOD. miR-23a regulates cardiomyocyte apoptosis by suppressing the expression of MnSOD. Our study reveals a novel model regulating cardiomyocyte apoptosis which is composed of miR-23a and MnSOD. Our study provides a new method to tackling apoptosis related cardiac diseases.

Keywords

References

  1. Barringhaus, K.G., and Zamore, P.D. (2009). MicroRNAs: regulating a change of heart. Circulation 119, 2217-2224. https://doi.org/10.1161/CIRCULATIONAHA.107.715839
  2. Bartel, D.P. (2004). MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281-297. https://doi.org/10.1016/S0092-8674(04)00045-5
  3. Barwari, T., Joshi, A., and Mayr, M. (2016). MicroRNAs in cardiovascular disease. J. Am. Coll.Cardiol. 68, 2577-2584. https://doi.org/10.1016/j.jacc.2016.09.945
  4. Candas, D., and Li, J.J. (2014). MnSOD in oxidative stress responsepotential regulation via mitochondrial protein influx. Antioxid. Redox Signal. 20, 1599-1617. https://doi.org/10.1089/ars.2013.5305
  5. Chen, Y.R., and Zweier, J.L. (2014). Cardiac mitochondria and reactive oxygen species generation. Circ. Res. 114, 524-537. https://doi.org/10.1161/CIRCRESAHA.114.300559
  6. Chen, C., Ridzon, D.A., Broomer, A.J., Zhou, Z., Lee, D.H., Nguyen, J.T., Barbisin, M., Xu, N.L., Mahuvakar, V.R., Andersen, M.R., et al. (2005). Real-time quantification of microRNAs by stem-loop RT-PCR. Nucleic Acids Res. 33, e179. https://doi.org/10.1093/nar/gni178
  7. Choi, W.Y., Giraldez, A.J., and Schier, A.F. (2007). Target protectors reveal dampening and balancing of Nodal agonist and antagonist by miR-430. Science 318, 271-274. https://doi.org/10.1126/science.1147535
  8. Cramer-Morales, K., Heer, C.D., Mapuskar, K.A., and Domann, F.E. (2015). SOD2 targeted gene editing by CRISPR/Cas9 yields Human cells devoid of MnSOD. Free Radic. Biol. Med. 89, 379-386. https://doi.org/10.1016/j.freeradbiomed.2015.07.017
  9. Dixon, S.J., and Stockwell, B.R. (2014). The role of iron and reactive oxygen species in cell death. Nat. Chem. Biol. 10, 9-17. https://doi.org/10.1038/nchembio.1416
  10. Dumortier, O., Hinault, C., and Van Obberghen, E. (2013). MicroRNAs and metabolism crosstalk in energy homeostasis. Cell Metabol. 18, 312-324. https://doi.org/10.1016/j.cmet.2013.06.004
  11. Duygu, B., de Windt, L.J., and da Costa Martins, P.A. (2016). Targeting microRNAs in heart failure. Trends Cardiovasc. Med. 26, 99-110. https://doi.org/10.1016/j.tcm.2015.05.008
  12. Filipowicz, W., Bhattacharyya, S.N., and Sonenberg, N. (2008). Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat. Rev. Genet. 9, 102-114. https://doi.org/10.1038/nrg2290
  13. Fukai, T., and Ushio-Fukai, M. (2011). Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid. Redox Signal. 15, 1583-1606. https://doi.org/10.1089/ars.2011.3999
  14. Hata, A. (2013). Functions of microRNAs in cardiovascular biology and disease. Ann. Rev. Physiol. 75, 69-93. https://doi.org/10.1146/annurev-physiol-030212-183737
  15. Kalogeris, T., Bao, Y., and Korthuis, R.J. (2014). Mitochondrial reactive oxygen species: a double edged sword in ischemia/reperfusion vs preconditioning. Redox. Biol. 2, 702-714. https://doi.org/10.1016/j.redox.2014.05.006
  16. Khan, R.S., Fonseca-Kelly, Z., Callinan, C., Zuo, L., Sachdeva, M.M., and Shindler, K.S. (2012). SIRT1 activating compounds reduce oxidative stress and prevent cell death in neuronal cells. Front. Cell. Neurosci. 6, 63.
  17. Kimoto, K., Suzuki, K., Kizaki, T., Hitomi, Y., Ishida, H., Katsuta, H., Itoh, E., Ookawara, T., Honke, K., and Ohno, H. (2003). Gliclazide protects pancreatic beta-cells from damage by hydrogen peroxide. Biochem. Biophy. Res. Commun. 303, 112-119. https://doi.org/10.1016/S0006-291X(03)00310-3
  18. Kinscherf, R., Claus, R., Wagner, M., Gehrke, C., Kamencic, H., Hou, D., Nauen, O., Schmiedt, W., Kovacs, G., Pill, J., et al. (1998). Apoptosis caused by oxidized LDL is manganese superoxide dismutase and p53 dependent. FASEB J. 12, 461-467. https://doi.org/10.1096/fasebj.12.6.461
  19. Kopf, P.G., Scott, J.A., Agbor, L.N., Boberg, J.R., Elased, K.M., Huwe, J.K., and Walker, M.K. (2010). Cytochrome P4501A1 is required for vascular dysfunction and hypertension induced by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Sci. 117, 537-546. https://doi.org/10.1093/toxsci/kfq218
  20. Lambeth, J.D., and Neish, A.S. (2014). Nox enzymes and new thinking on reactive oxygen: a double-edged sword revisited. Ann. Rev. Pathol. 9, 119-145. https://doi.org/10.1146/annurev-pathol-012513-104651
  21. Landmesser, U., Dikalov, S., Price, S.R., McCann, L., Fukai, T., Holland, S.M., Mitch, W.E., and Harrison, D.G. (2003). Oxidation of tetrahydrobiopterin leads to uncoupling of endothelial cell nitric oxide synthase in hypertension. J. Clin. Invest. 111, 1201-1209. https://doi.org/10.1172/JCI200314172
  22. Latronico, M.V., and Condorelli, G. (2009). MicroRNAs and cardiac pathology. Nat. Rev. Cardiol. 6, 419-429.
  23. Li, Y., Huang, T.T., Carlson, E.J., Melov, S., Ursell, P.C., Olson, J.L., Noble, L.J., Yoshimura, M.P., Berger, C., Chan, P.H., et al. (1995). Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase. Nat. Genet. 11, 376-381. https://doi.org/10.1038/ng1295-376
  24. Li, J., Aung, L.H., Long, B., Qin, D., An, S., and Li, P. (2015). miR-23a binds to p53 and enhances its association with miR-128 promoter. Sci. Rep. 5, 16422. https://doi.org/10.1038/srep16422
  25. Lin, Z., Murtaza, I., Wang, K., Jiao, J., Gao, J., and Li, P.F. (2009). miR-23a functions downstream of NFATc3 to regulate cardiac hypertrophy. Proc. Natl. Acad. Sci. USA 106, 12103-12108. https://doi.org/10.1073/pnas.0811371106
  26. Liochev, S.I. (2013). Reactive oxygen species and the free radical theory of aging. Free Rad. Biol. Med. 60, 1-4. https://doi.org/10.1016/j.freeradbiomed.2013.02.011
  27. Long, B., Wang, K., Li, N., Murtaza, I., Xiao, J.Y., Fan, Y.Y., Liu, C.Y., Li, W.H., Cheng, Z., and Li, P. (2013). miR-761 regulates the mitochondrial network by targeting mitochondrial fission factor. Free Rad. Biol. Med. 65, 371-379. https://doi.org/10.1016/j.freeradbiomed.2013.07.009
  28. Moe, G.W., and Marin-Garcia, J. (2016). Role of cell death in the progression of heart failure. Heart Fail. Rev. 21, 157-167. https://doi.org/10.1007/s10741-016-9532-0
  29. Murphy, M.P. (2009). How mitochondria produce reactive oxygen species. Biochem. J. 417, 1-13. https://doi.org/10.1042/BJ20081386
  30. Murphy, M.P., Holmgren, A., Larsson, N.G., Halliwell, B., Chang, C.J., Kalyanaraman, B., Rhee, S.G., Thornalley, P.J., Partridge, L., Gems, D., et al. (2011). Unraveling the biological roles of reactive oxygen species. Cell Metabol. 3, 361-366.
  31. Olivetti, G., Abbi, R., Quaini, F., Kajstura, J., Cheng, W., Nitahara, J.A., Quaini, E., Di Loreto, C., Beltrami, C.A., Krajewski, S., et al. (1997). Apoptosis in the failing human heart. N Engl. J. Med. 336, 1131-1141. https://doi.org/10.1056/NEJM199704173361603
  32. Sena, L.A., and Chandel, N.S. (2012). Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 48, 158-167. https://doi.org/10.1016/j.molcel.2012.09.025
  33. Skommer, J., Rana, I., Marques, F.Z., Zhu, W., Du, Z., and Charchar, F.J. (2014). Small molecules, big effects: the role of microRNAs in regulation of cardiomyocyte death. Cell Death Dis. 5, e1325. https://doi.org/10.1038/cddis.2014.287
  34. Subramanian, S., and Steer, C.J. (2010). MicroRNAs as gatekeepers of apoptosis. J. Cell. Physiol. 223, 289-298.
  35. Takami, Y., Uto, H., Tamai, T., Sato, Y., Ishida, Y., Morinaga, H., Sakakibara, Y., Moriuchi, A., Oketani, M., Ido, A., et al. (2010). Identification of a novel biomarker for oxidative stress induced by hydrogen peroxide in primary human hepatocytes using the 2-nitrobenzenesulfenyl chloride isotope labeling method. Hepatol. Res. 40, 438-445. https://doi.org/10.1111/j.1872-034X.2009.00615.x
  36. Wang, K., Long, B., Liu, F., Wang, J.X., Liu, C.Y., Zhao, B., Zhou, L.Y., Sun, T., Wang, M., Yu, T., et al. (2016). A circular RNA protects the heart from pathological hypertrophy and heart failure by targeting miR-223. Eur. Heart J. 37, 2602-2611. https://doi.org/10.1093/eurheartj/ehv713
  37. Yang, J., Marden, J.J., Fan, C., Sanlioglu, S., Weiss, R.M., Ritchie, T.C., Davisson, R.L., and Engelhardt, J.F. (2003). Genetic redox preconditioning differentially modulates AP-1 and NF kappa B responses following cardiac ischemia/reperfusion injury and protects against necrosis and apoptosis. Mol. Therapy 7, 341-353. https://doi.org/10.1016/S1525-0016(02)00061-8
  38. Yang, B., Lin, H., Xiao, J., Lu, Y., Luo, X., Li, B., Zhang, Y., Xu, C., Bai, Y., Wang, H., et al. (2007). The muscle-specific microRNA miR-1 regulates cardiac arrhythmogenic potential by targeting GJA1 and KCNJ2. Nat. Med. 13, 486-491. https://doi.org/10.1038/nm1569
  39. Zhao, H., Tao, Z., Wang, R., Liu, P., Yan, F., Li, J., Zhang, C., Ji, X., and Luo, Y. (2014). MicroRNA-23a-3p attenuates oxidative stress injury in a mouse model of focal cerebral ischemia-reperfusion. Brain Res. 1592, 65-72. https://doi.org/10.1016/j.brainres.2014.09.055

Cited by

  1. Bulky DNA adducts, microRNA profiles, and lipid biomarkers in Norwegian tunnel finishing workers occupationally exposed to diesel exhaust vol.76, pp.1, 2019, https://doi.org/10.1136/oemed-2018-105445
  2. Apoptotic suppression of inflammatory macrophages and foam cells in vascular tissue by miR-23a pp.2190-7196, 2019, https://doi.org/10.1007/s12553-019-00301-y
  3. Postnatal Expression Profile of microRNAs Associated with Cardiovascular and Cerebrovascular Diseases in Children at the Age of 3 to 11 Years in Relation to Previous Occurrence of Pregnancy-Related Complications vol.20, pp.3, 2019, https://doi.org/10.3390/ijms20030654
  4. Reactive Oxygen Species Related Noncoding RNAs as Regulators of Cardiovascular Diseases vol.15, pp.3, 2019, https://doi.org/10.7150/ijbs.30464
  5. Integrative System Biology Analyses Identify Seven MicroRNAs to Predict Heart Failure vol.5, pp.1, 2017, https://doi.org/10.3390/ncrna5010022
  6. LncRNA XIST regulates myocardial infarction by targeting miR‐130a‐3p vol.234, pp.6, 2019, https://doi.org/10.1002/jcp.26327
  7. A circulating miRNA signature for early diagnosis of acute kidney injury following acute myocardial infarction vol.17, pp.None, 2019, https://doi.org/10.1186/s12967-019-1890-7
  8. Oxidative Stress-Responsive MicroRNAs in Heart Injury vol.21, pp.1, 2020, https://doi.org/10.3390/ijms21010358
  9. Substantially Altered Expression Profile of Diabetes/Cardiovascular/Cerebrovascular Disease Associated microRNAs in Children Descending from Pregnancy Complicated by Gestational Diabetes Mellitus vol.9, pp.6, 2017, https://doi.org/10.3390/cells9061557
  10. MicroRNA and ROS Crosstalk in Cardiac and Pulmonary Diseases vol.21, pp.12, 2020, https://doi.org/10.3390/ijms21124370
  11. Kawasaki disease: pathophysiology and insights from mouse models vol.16, pp.7, 2017, https://doi.org/10.1038/s41584-020-0426-0
  12. Long noncoding RNA PVT1 facilitates high glucose‐induced cardiomyocyte death through the miR‐23a‐3p/CASP10 axis vol.45, pp.1, 2017, https://doi.org/10.1002/cbin.11479
  13. In Silico Integrative Approach Revealed Key MicroRNAs and Associated Target Genes in Cardiorenal Syndrome vol.15, pp.None, 2017, https://doi.org/10.1177/11779322211027396
  14. Contribution of Oxidative Stress to HIF-1-Mediated Profibrotic Changes during the Kidney Damage vol.2021, pp.None, 2017, https://doi.org/10.1155/2021/6114132
  15. Intake of Natural Compounds and Circulating microRNA Expression Levels: Their Relationship Investigated in Healthy Subjects With Different Dietary Habits vol.11, pp.None, 2017, https://doi.org/10.3389/fphar.2020.619200
  16. Leonurine Alleviates Hypoxia-Induced Myocardial Damage by Regulating miRNAs vol.16, pp.4, 2021, https://doi.org/10.1177/1934578x211007274
  17. Micro RNAs in Regulation of Cellular Redox Homeostasis vol.22, pp.11, 2021, https://doi.org/10.3390/ijms22116022
  18. Current Knowledge of MicroRNAs (miRNAs) in Acute Coronary Syndrome (ACS): ST-Elevation Myocardial Infarction (STEMI) vol.11, pp.10, 2021, https://doi.org/10.3390/life11101057
  19. Impact of the Main Cardiovascular Risk Factors on Plasma Extracellular Vesicles and Their Influence on the Heart’s Vulnerability to Ischemia-Reperfusion Injury vol.10, pp.12, 2021, https://doi.org/10.3390/cells10123331
  20. Involvement of miRNAs in response to oxidative stress induced by the steroidal glycoalkaloid α‐solanine in hepatocellular carcinoma cells vol.37, pp.2, 2017, https://doi.org/10.1002/tox.23390