AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
View PDF
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Downregulation of miR-181a-5p alleviates oxidative stress and inflammation in coronary microembolization-induced myocardial damage by directly targeting XIAP

You ZHOU1,*Man-Yun LONG1,*Zhi-Qing CHEN1Jun-Wen HUANG1Zhen-Bai QIN1Lang LI1,2( )
Department of Cardiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Nanning, China

*The authors contributed equally to this manuscript

Show Author Information

Abstract

BACKGROUND

Coronary microembolization (CME) is a complicated problem that commonly arises in the context of coronary angioplasty. MicroRNAs play crucial roles in cardiovascular diseases. However, the role and mechanism of miR-181a-5p in CME-induced myocardial injury remains unclear.

METHODS

We established CME rat models. Cardiac function was detected by echocardiography. Haematoxylin-basic fuchsin-picric acid staining was used to measure micro-infarction size. Serum samples and cell culture supernatants were evaluated via enzyme-linked immunosorbent assay. Cellular reactive oxygen species were determined by dichloro-dihydro-fluorescein diacetate assay, and the other oxidative stress related parameters were assayed by spectrophotometry. The dual-luciferase reporter (DLR) assay and RNA pulldown were conducted to validate the association between miR-181a-5p and X-linked inhibitor of apoptosis protein (XIAP). The expression of miR-181a-5p and XIAP mRNA were determined by quantitative reverse transcription polymerase chain reaction. Proteins were evaluated via immunoblotting. The viability of the cell was evaluated via cell counting kit-8 assay.

RESULTS

The miR-181a-5p level was significantly increased in CME myocardial tissues. Downregulation of miR-181a-5p improved CME-induced cardiac dysfunction and alleviated myocardial oxidative stress and inflammatory injury, whereas miR-181a-5p exhibited the opposite effects. Then, the DLR assay and RNA pulldown results revealed that miR-181a-5p directly targeting on XIAP. The XIAP level was found to be remarkably decreased after CME. XIAP overexpression attenuated CME-induced myocardial oxidative stress and inflammatory injury. Finally, in vitro rescue experiments revealed that knockdown of XIAP could abolish the protective effects of miR-181a-5p knockdown on hypoxia-induced cardiomyocyte oxidative stress and inflammatory injury.

CONCLUSIONS

Downregulation of miR-181a-5p alleviates CME-induced myocardial damage by suppressing myocardial oxidative stress and inflammation through directly targeting XIAP.

References

[1]

Erbel R, Heusch G. Coronary microembolization. J Am Coll Cardiol 2000; 36: 22−24.

[2]

Salem SA, Haji S, Garg N, et al. Occlusion of right coronary artery by microembolization caused by excessive diagnostic catheter manipulation. Ann Transl Med 2018; 6: 20.

[3]

Heusch G, Skyschally A, Kleinbongard P. Coronary microembolization and microvascular dysfunction. Int J Cardiol 2018; 258: 17−23.

[4]

Wang Y, Zhao HW, Wang CF, et al. Incidence, predictors, and prognosis of coronary slow-flow and no-reflow phenomenon in patients with chronic total occlusion who underwent percutaneous coronary intervention. Ther Clin Risk Manag 2020; 16: 95−101.

[5]

Kalyoncuoglu M, Biter Hİ, Ozturk S, et al. Predictive accuracy of lymphocyte-to-monocyte ratio and monocyte-to-high-density-lipoprotein-cholesterol ratio in determining the slow flow/no-reflow phenomenon in patients with non-ST-elevated myocardial infarction. Coron Artery Dis 2020; 31: 518−526.

[6]

Camici PG, Crea F. Coronary microvascular dysfunction. N Engl J Med 2007; 356: 830−840.

[7]

Nakanishi K, Daimon M. Coronary slow flow and subclinical left ventricular dysfunction. Int Heart J 2019; 60: 495−496.

[8]

Heusch G. The regional myocardial flow-function relationship: a framework for an understanding of acute ischemia, hibernation, stunning and coronary microembolization. 1980. Circ Res 2013; 112: 1535−1537.

[9]

Chen ZW, Qian JY, Ma JY, et al. TNF-α-induced cardiomyocyte apoptosis contributes to cardiac dysfunction after coronary microembolization in mini-pigs. J Cell Mol Med 2014; 18: 1953−1963.

[10]

Chen A, Chen Z, Zhou Y, et al. Rosuvastatin protects against coronary microembolization-induced cardiac injury via inhibiting NLRP3 inflammasome activation. Cell Death Dis 2021; 12: 78.

[11]

Su Q, Lv X, Ye Z. Ligustrazine attenuates myocardial injury induced by coronary microembolization in rats by activating the PI3K/Akt pathway. Oxid Med Cell Longev 2019; 2019: 6791457.

[12]

Zhu L, Li N, Sun L, et al. Non-coding RNAs: the key detectors and regulators in cardiovascular disease. Genomics 2021; 113: 1233−1246.

[13]

Arabian M, Mirzadeh Azad F, Maleki M, et al. Insights into role of microRNAs in cardiac development, cardiac diseases, and developing novel therapies. Iran J Basic Med Sci 2020; 23: 961−969.

[14]

Zhou H, Ni WJ, Meng XM, et al. MicroRNAs as regulators of immune and inflammatory responses: potential therapeutic targets in diabetic nephropathy. Front Cell Dev Biol 2021; 8: 618536.

[15]

Colpaert RMW, Calore M. Epigenetics and microRNAs in cardiovascular diseases. Genomics 2021; 113: 540−551.

[16]

Ye H, Xu G, Zhang D, et al. The protective effects of the miR-129-5p/keap-1/Nrf2 axis on Ang II-induced cardiomyocyte hypertrophy. Ann Transl Med 2021; 9: 154.

[17]
Qu Y, Zhang J, Zhang J, et al. MiR-708-3p alleviates inflammation and myocardial injury after myocardial infarction by suppressing ADAM17 expression. Inflammation. Published Online First: 25 January 2021. DOI: 10.1007/s10753-020-01404-9.
[18]

Su Q, Lv X, Ye Z, et al. The mechanism of miR-142-3p in coronary microembolization-induced myocardiac injury via regulating target gene IRAK-1. Cell Death Dis 2019; 10: 61.

[19]

Zhu HH, Wang XT, Sun YH, et al. MicroRNA-486-5p targeting PTEN protects against coronary microembolization-induced cardiomyocyte apoptosis in rats by activating the PI3K/AKT pathway. Eur J Pharmacol 2019; 855: 244−251.

[20]

Sun X, Sit A, Feinberg MW. Role of miR-181 family in regulating vascular inflammation and immunity. Trends Cardiovasc Med 2014; 24: 105−112.

[21]

Sun YH, Su Q, Li L, et al. Expression of p53 in myocardium following coronary microembolization in rats and its significance. J Geriatr Cardiol 2017; 14: 292−300.

[22]

Heusch G. The coronary circulation as a target of cardioprotection. Circ Res 2016; 118: 1643−1658.

[23]

Malyar NM, Lerman LO, Gössl M, et al. Relation of nonperfused myocardial volume and surface area to left ventricular performance in coronary microembolization. Circulation 2004; 110: 1946−1952.

[24]

Dörge H, Schulz R, Belosjorow S, et al. Coronary microembolization: the role of TNF-alpha in contractile dysfunction. J Mol Cell Cardiol 2002; 34: 51−62.

[25]

Li S, Zhong S, Zeng K, et al. Blockade of NF-kappaB by pyrrolidine dithiocarbamate attenuates myocardial inflammatory response and ventricular dysfunction following coronary microembolization induced by homologous microthrombi in rats. Basic Res Cardiol 2010; 105: 139−150.

[26]

Su Q, Li L, Sun Y, et al. Effects of the TLR4/Myd88/NF-kappaB signaling pathway on NLRP3 inflammasome in coronary microembolization-induced myocardial injury. Cell Physiol Biochem 2018; 47: 1497−1508.

[27]

Ambros V. MicroRNAs: tiny regulators with great potential. Cell 2001; 107: 823−826.

[28]

Ke XS, Liu CM, Liu DP, et al. MicroRNAs: key participants in gene regulatory networks. Curr Opin Chem Biol 2003; 7: 516−523.

[29]

Kong B, Qin Z, Ye Z, et al. MicroRNA-26a-5p affects myocardial injury induced by coronary microembolization by modulating HMGA1. J Cell Biochem 2019; 120: 10756−10766.

[30]

Qin S, Yang C, Zhang B, et al. XIAP inhibits mature Smac-induced apoptosis by degrading it through ubiquitination in NSCLC. Int J Oncol 2016; 49: 1289−1296.

[31]

Estornes Y, Bertrand MJM. IAPs, regulators of innate immunity and inflammation. Semin Cell Dev Biol 2015; 39: 106−114.

[32]

Lu M, Qin X, Yao J, et al. MiR-134-5p targeting XIAP modulates oxidative stress and apoptosis in cardiomyocytes under hypoxia/reperfusion-induced injury. IUBMB Life 2020; 72: 2154−2166.

[33]

Zhao R, Yu Q, Hou L, et al. Cadmium induces mitochondrial ROS inactivation of XIAP pathway leading to apoptosis in neuronal cells. Int J Biochem Cell Biol 2020; 121: 105715.

[34]

Zilu S, Qian H, Haibin W, et al. Effects of XIAP on high fat diet-induced hepatic steatosis: a mechanism involving NLRP3 inflammasome and oxidative stress. Aging 2019; 11: 12177−12201.

[35]

Zheng J, Long M, Qin Z, et al. Nicorandil inhibits cardiomyocyte apoptosis and improves cardiac function by suppressing the HtrA2/XIAP/PARP signaling after coronary microembolization in rats. Pharmacol Res Perspect 2021; 9: e00699.

Journal of Geriatric Cardiology
Pages 426-439
Cite this article:
ZHOU Y, LONG M-Y, CHEN Z-Q, et al. Downregulation of miR-181a-5p alleviates oxidative stress and inflammation in coronary microembolization-induced myocardial damage by directly targeting XIAP. Journal of Geriatric Cardiology, 2021, 18(6): 426-439. https://doi.org/10.11909/j.issn.1671-5411.2021.06.007

531

Views

25

Downloads

0

Crossref

16

Web of Science

12

Scopus

0

CSCD

Published: 28 June 2021
© 2021 JGC All rights reserved
Return