Dexmedetomidine postconditioning suppresses myocardial ischemia/reperfusion injury by activating the SIRT1/mTOR axis

Xiong Zhang, Yongxing Li, Yong Wang, Yuerong Zhuang, Xiaojie Ren, Kai Yang, Wuhua Ma, Ming Zhong, Xiong Zhang, Yongxing Li, Yong Wang, Yuerong Zhuang, Xiaojie Ren, Kai Yang, Wuhua Ma, Ming Zhong

Abstract

Myocardial ischemia/reperfusion (MI/R) triggers a complicated chain of inflammatory reactions. Dexmedetomidine (Dex) has been reported to be important in myocardial disorders. We evaluated the role of Dex in MI/R injury via the silent information regulator factor 2-related enzyme 1 (SIRT1)/mammalian target of rapamycin (mTOR) signaling pathway. First, Dex was immediately injected into rat models of MI/R injury during reperfusion. After Evans Blue-triphenyl tetrazolium chloride (TTC) and Hematoxylin-Eosin (H-E) staining, MI/R injury was observed. The extracted serum and myocardial tissues were used to detect oxidative stress and the inflammatory response. Western blot analysis was performed to evaluate MI/R autophagy and the levels of proteins associated with the SIRT1/mTOR axis. The effects of the combination of Dex and SIRT1 inhibitor EX527 on MI/R injury and autophagy were evaluated. Finally, the mechanism of Dex was tested, and autophagy levels and the levels of proteins associated with the SIRT1/mTOR signaling pathway were assessed in MI/R rats. The results of the present study suggested that Dex relieved MI/R injury, reduced cardiomyocyte apoptosis, oxidative stress and inflammatory reactions, up-regulated the SIRT1/mTOR axis and decreased overautophagy in MI/R rats. SIRT1 inhibitor EX527 attenuated the protective effects of Dex. Our study demonstrated that Dex alleviated MI/R injury by activating the SIRT1/mTOR axis. This investigation may offer new insight into the treatment of MI/R injury.

Keywords: Dexmedetomidine; EX527; Myocardial ischemia/reperfusion; SIRT1; mTOR.

Conflict of interest statement

The authors declare that there are no competing interests associated with the manuscript.

© 2020 The Author(s).

Figures

Figure 1. Protective functions of Dex in…
Figure 1. Protective functions of Dex in MI/R rats
(A) Representative ECG of MI/R rats. (B) Images of myocardial infarction in rats visualized by TTC staining and analysis of the infarct size, with the white part representing ischemic infarcted myocardium, red representing ischemic noninfarcted myocardium, and blue representing normal myocardium; the percentage of the infarct area relative to the total area analyzed. (C) Representative images of cardiomyocytes stained with HE. (D,E) LDH content and CK-MB content in rat serum. (F) TUNEL-stained myocardial tissues; the TUNEL-positive cells are shown in green. (G) Bcl-2 and cleaved caspase-3 expression detected by Western blot analysis. The data are expressed as the mean ± standard deviation, n=6. One-way ANOVA and Tukey’s multiple comparisons test were applied to determine statistical significance. *P<0.05, compared with the sham group, #P<0.05, compared with the MI/R group.
Figure 2. Effects of Dex on inflammatory…
Figure 2. Effects of Dex on inflammatory responses and oxidative stress in MI/R rats
(AC), (A) TNF-α, (B) IL-1β and (C) IL-6 contents in serum. (DG) (D) SOD, (E) CAT, (F) GSH-Px and (G) MDA contents in myocardial tissues. The data are expressed as the mean ± standard deviation, n=6. One-way ANOVA and Tukey’s multiple comparisons test were applied to determine statistical significance. *P<0.05, compared with the sham group, #P<0.05, compared with the MI/R group.
Figure 3. Dex reduces autophagy in MI/R…
Figure 3. Dex reduces autophagy in MI/R rats
Western blot analysis of LC3II/LC3I, p62 and Beclin-1 expression in myocardial tissues. The data are expressed as the mean ± standard deviation, n=6. One-way ANOVA and Tukey’s multiple comparisons test were applied to determine statistical significance. *P<0.05, compared with the sham group, #P<0.05, compared with the MI/R group. Abbreviations: MI/R, myocardial/ischemia reperfusion.
Figure 4. Effects of Dex on SIRT1…
Figure 4. Effects of Dex on SIRT1 and mTOR expression
(A) Representative results of SIRT1 immunohistochemistry in myocardial tissue. (B) SIRT1 expression in nuclei in myocardial tissue detected using Western blot analysis. (C) mTOR expression in myocardial tissue measured by Western blot analysis. The data are expressed as the mean ± standard deviation, n=6. One-way ANOVA and Tukey’s multiple comparisons test were applied to determine statistical significance. *P<0.05, compared with the sham group, #P<0.05, compared with the MI/R group.
Figure 5. Effects of EX527 on autophagy…
Figure 5. Effects of EX527 on autophagy and the SIRT1/mTOR axis in MI/R rats after Dex postconditioning
(A) Representative results of SIRT1 immunohistochemistry in myocardial tissues. (B) SIRT1 expression in nuclei in myocardial tissue detected by Western blot analysis. (C,D) mTOR (C) LC3II/LC3I, p62 and Beclin-1 (D) expression in myocardial tissues measured by Western blot analysis. The data are expressed as the mean ± standard deviation, n=6. The t test was employed to analyze comparisons between two groups. *P<0.05, compared with the MI/R + Dex group.
Figure 6. Effects of EX527 on rats…
Figure 6. Effects of EX527 on rats with MI/R injury after Dex postconditioning
(A) Representative images of myocardial infarction in rats visualized by TTC staining and analysis of the infarct size. (B) Rat cardiomyocytes stained with H-E. (C) Representative images and analysis of TUNEL staining. (D,E) LDH (D) and CK-MB (E) levels in serum. The data are expressed as the mean ± standard deviation, n=6. The t test was employed to analyze comparisons between two groups. *P<0.05, compared with the MI/R + Dex group.

References

    1. Powers S.K., Smuder A.J., Kavazis A.N. and Quindry J.C. (2014) Mechanisms of exercise-induced cardioprotection. Physiology 29, 27–38
    1. Guo J., Zhu J., Ma L. et al. . (2018) Chronic kidney disease exacerbates myocardial ischemia reperfusion injury: role of endoplasmic reticulum stress-mediated apoptosis. Shock 49, 712–720 10.1097/SHK.0000000000000970
    1. Halladin N.L. (2015) Oxidative and inflammatory biomarkers of ischemia and reperfusion injuries. Dan Med. J. 62, B5054.
    1. Nakamura T., Mizuno S., Matsumoto K., Sawa Y., Matsuda H. and Nakamura T. (2000) Myocardial protection from ischemia/reperfusion injury by endogenous and exogenous HGF. J. Clin. Invest. 106, 1511–1519 10.1172/JCI10226
    1. Ferrari R., Balla C., Malagu M. et al. . (2017) Reperfusion damage- a story of success, failure, and hope. Circ. J. 81, 131–141 10.1253/circj.CJ-16-1124
    1. Ndrepepa G., Colleran R. and Kastrati A. (2017) Reperfusion injury in ST-segment elevation myocardial infarction: the final frontier. Coron. Artery Dis. 28, 253–262 10.1097/MCA.0000000000000468
    1. Ibanez B., Heusch G., Ovize M. and Van de Werf F. (2015) Evolving therapies for myocardial ischemia/reperfusion injury. J. Am. Coll. Cardiol. 65, 1454–1471 10.1016/j.jacc.2015.02.032
    1. Wu D., Wang J., Li H., Xue M., Ji A. and Li Y. (2015) Role of hydrogen sulfide in ischemia-reperfusion injury. Oxid. Med. Cell. Longev. 2015, 186908 10.1155/2015/186908
    1. Xia Z., Li H. and Irwin M.G. (2016) Myocardial ischaemia reperfusion injury: the challenge of translating ischaemic and anaesthetic protection from animal models to humans. Br. J. Anaesth. 117, ii44–ii62 10.1093/bja/aew267
    1. Mokhtari-Zaer A., Marefati N., Atkin S.L., Butler A.E. and Sahebkar A. (2018) The protective role of curcumin in myocardial ischemia-reperfusion injury. J. Cell. Physiol. 234, 214–222 10.1002/jcp.26848
    1. Keating G.M. (2015) Dexmedetomidine: a review of its use for sedation in the intensive care setting. Drugs 75, 1119–1130 10.1007/s40265-015-0419-5
    1. Hu J., Gu X.Y., Meng Y. et al. . (2017) Effect of dexmedetomidine postconditioning on myocardial ischemia-reperfusion injury and inflammatory response in diabetic rats. Nan Fang Yi Ke Da Xue Xue Bao 37, 1506–1511
    1. Kida Y. and Goligorsky M.S. (2016) Sirtuins, cell senescence, and vascular aging. Can. J. Cardiol. 32, 634–641 10.1016/j.cjca.2015.11.022
    1. Meng X., Tan J., Li M., Song S., Miao Y. and Zhang Q. (2017) Sirt1: role under the condition of ischemia/hypoxia. Cell. Mol. Neurobiol. 37, 17–28 10.1007/s10571-016-0355-2
    1. Han D., Wang J., Ma S., Chen Y. and Cao F. (2017) SIRT1 as a promising novel therapeutic target for myocardial ischemia reperfusion injury and cardiometabolic disease. Curr. Drug Targets 18, 1746–1753 10.2174/1389450116666150630110529
    1. Chen L., Cao J., Cao D. et al. . (2019) Protective effect of dexmedetomidine against diabetic hyperglycemia-exacerbated cerebral ischemia/reperfusion injury: an in vivo and in vitro study. Life Sci. 235, 116553 10.1016/j.lfs.2019.116553
    1. Yoon M.S. (2017) The role of mammalian target of rapamycin (mTOR) in insulin signaling. Nutrients 9, 1176 10.3390/nu9111176
    1. Kim Y.C. and Guan K.L. (2015) mTOR: a pharmacologic target for autophagy regulation. J. Clin. Invest. 125, 25–32 10.1172/JCI73939
    1. Liang S., Ping Z. and Ge J. (2017) Coenzyme Q10 regulates antioxidative stress and autophagy in acute myocardial ischemia-reperfusion injury. Oxid. Med. Cell. Longev. 2017, 9863181 10.1155/2017/9863181
    1. Lempiainen J., Finckenberg P., Mervaala E.E. et al. . (2014) Dexmedetomidine preconditioning ameliorates kidney ischemia-reperfusion injury. Pharmacol. Res. Perspect. 2, e00045 10.1002/prp2.45
    1. Godar R.J., Ma X., Liu H. et al. . (2015) Repetitive stimulation of autophagy-lysosome machinery by intermittent fasting preconditions the myocardium to ischemia-reperfusion injury. Autophagy 11, 1537–1560 10.1080/15548627.2015.1063768
    1. Lejay A., Fang F., John R. et al. . (2016) Ischemia reperfusion injury, ischemic conditioning and diabetes mellitus. J. Mol. Cell Cardiol. 91, 11–22 10.1016/j.yjmcc.2015.12.020
    1. Afonso J. and Reis F. (2012) Dexmedetomidine: current role in anesthesia and intensive care. Rev. Bras. Anestesiol. 62, 118–133 10.1016/S0034-7094(12)70110-1
    1. Cai Y., Xu H., Yan J., Zhang L. and Lu Y. (2014) Molecular targets and mechanism of action of dexmedetomidine in treatment of ischemia/reperfusion injury. Mol. Med. Rep. 9, 1542–1550 10.3892/mmr.2014.2034
    1. Zhou H., Zhou D., Lu J., Wu C. and Zhu Z. (2019) Effects of pre-cardiopulmonary bypass administration of dexmedetomidine on cardiac injuries and the inflammatory response in valve replacement surgery with a sevoflurane postconditioning protocol: a pilot study. J. Cardiovasc. Pharmacol. 74, 91–97 10.1097/FJC.0000000000000698
    1. Zhang W. and Zhang J. (2017) Dexmedetomidine preconditioning protects against lung injury induced by ischemia-reperfusion through inhibition of autophagy. Exp. Ther. Med. 14, 973–980 10.3892/etm.2017.4623
    1. Shi J., Dai W. and Kloner R.A. (2017) Therapeutic hypothermia reduces the inflammatory response following ischemia/reperfusion injury in rat hearts. Ther. Hypotherm. Temp. Manag. 7, 162–170 10.1089/ther.2016.0042
    1. Sukegawa S., Higuchi H., Inoue M., Nagatsuka H., Maeda S. and Miyawaki T. (2014) Locally injected dexmedetomidine inhibits carrageenin-induced inflammatory responses in the injected region. Anesth. Analg. 118, 473–480 10.1213/ANE.0000000000000060
    1. Sinning C., Westermann D. and Clemmensen P. (2017) Oxidative stress in ischemia and reperfusion: current concepts, novel ideas and future perspectives. Biomark. Med. 11, 11031–11040 10.2217/bmm-2017-0110
    1. Fu C., Dai X., Yang Y., Lin M., Cai Y. and Cai S. (2017) Dexmedetomidine attenuates lipopolysaccharide-induced acute lung injury by inhibiting oxidative stress, mitochondrial dysfunction and apoptosis in rats. Mol. Med. Rep. 15, 131–138 10.3892/mmr.2016.6012
    1. Ravanan P., Srikumar I.F. and Talwar P. (2017) Autophagy: the spotlight for cellular stress responses. Life Sci. 188, 53–67 10.1016/j.lfs.2017.08.029
    1. Shao H., Yang L., Wang L., Tang B., Wang J. and Li Q. (2018) MicroRNA-34a protects myocardial cells against ischemia-reperfusion injury through inhibiting autophagy via regulating TNFalpha expression. Biochem. Cell. Biol. 96, 349–354 10.1139/bcb-2016-0158
    1. Carafa V., Rotili D., Forgione M. et al. . (2016) Sirtuin functions and modulation: from chemistry to the clinic. Clin. Epigenetics 8, 61 10.1186/s13148-016-0224-3
    1. Sato A. (2016) mTOR, a potential target to treat autism spectrum disorder. CNS Neurol. Disord. Drug Targets 15, 533–543 10.2174/1871527315666160413120638
    1. Yang H., Bi Y., Xue L. et al. . (2015) Multifaceted modulation of SIRT1 in cancer and inflammation. Crit. Rev. Oncog. 20, 49–64 10.1615/CritRevOncog.2014012374
    1. Hattori Y. and Ihara M. (2016) Sirt1. Nihon Rinsho 74, 589–594
    1. Fang S., Chen Y., Yao P., Li Y., Yang Y. and Xu G. (2018) Dexmedetomidine alleviates postoperative cognitive dysfunction in aged rats probably via silent information regulator 1 pathway. Nan Fang Yi Ke Da Xue Xue Bao 38, 1071–1075
    1. Zhao D., Yang J. and Yang L. (2017) Insights for oxidative stress and mTOR signaling in myocardial ischemia/reperfusion injury under diabetes. Oxid. Med. Cell. Longev. 2017, 6437467 10.1155/2017/6437467
    1. Gertz M., Fischer F., Nguyen G.T. et al. . (2013) Ex-527 inhibits Sirtuins by exploiting their unique NAD+-dependent deacetylation mechanism. Proc. Natl. Acad. Sci. U.S.A. 110, E2772–E2781 10.1073/pnas.1303628110
    1. Li L. and Bhatia R. (2015) Role of SIRT1 in the growth and regulation of normal hematopoietic and leukemia stem cells. Curr. Opin. Hematol. 22, 324–329 10.1097/MOH.0000000000000152
    1. Choi J., Jo M., Lee E., Kim H.J. and Choi D. (2014) Differential induction of autophagy by mTOR is associated with abnormal apoptosis in ovarian endometriotic cysts. Mol. Hum. Reprod. 20, 309–317 10.1093/molehr/gat091
    1. Zhang W.X., He B.M., Wu Y., Qiao J.F. and Peng Z.Y. (2019) Melatonin protects against sepsis-induced cardiac dysfunction by regulating apoptosis and autophagy via activation of SIRT1 in mice. Life Sci. 217, 8–15 10.1016/j.lfs.2018.11.055

Source: PubMed

3
Předplatit