Korean Circ J.  2023 Jun;53(6):387-403. 10.4070/kcj.2022.0301.

LOXL1-AS1 Aggravates Myocardial Ischemia/Reperfusion Injury Through the miR-761/PTEN Axis

Affiliations
  • 1Department of Cardiovascular Medicine, The First People’s Hospital of Chenzhou, Chenzhou, China
  • 2Department of Intensive Care Unit (ICU), Hunan Chest Hospital, Changsha, China
  • 3Translational Medicine Institute, The First People’s Hospital of Chenzhou, Chenzhou, China

Abstract

Background and Objectives
Myocardial ischemia and reperfusion injury (MIRI) has high morbidity and mortality worldwide. We aimed to explore the role of long noncoding RNA lysyl oxidase like 1 antisense RNA 1 (LOXL1-AS1) in cardiomyocyte pyroptosis.
Methods
Hypoxia/reoxygenation (H/R) injury was constructed in human cardiomyocyte (HCM). The level of LOXL1-AS1, miR-761, phosphatase and tensin homolog (PTEN) and pyroptosis-related proteins was monitored by quantitative real-time polymerase chain reaction or western blot. Flow cytometry examined the pyroptosis level. Lactate dehydrogenase (LDH), creatine kinase-MB and cardiac troponin I levels were detected by test kits. Enzyme-linked immunosorbent assay measured the release of inflammatory cytokines. Dual-luciferase assay validated the binding relationship among LOXL1-AS1, miR-761, and PTEN. Finally, ischemia/reperfusion (I/R) animal model was constructed. Hematoxylin and eosin staining assessed morphological changes of myocardial tissue. NOD-like receptor pyrin domain-containing protein 3 (NLRP3) and casepase-1 expression was determined by immunohistochemistry.
Results
After H/R treatment, LOXL1-AS1 and PTEN were highly expressed but miR-761 level was suppressed. LOXL1-AS1 inhibition or miR-761 overexpression increased cell viability, blocked the release of LDH and inflammatory cytokines (interleukin [IL]-1β, IL-18), inhibited pyroptosis level, and downregulated pyroptosis-related proteins (ASC, cleaved caspase-1, gasdermin D-N, NLRP3, IL-1β, and IL-18) levels in HCMs. LOXL1-AS1 sponged miR-761 to upregulate PTEN. Knockdown of miR-761 reversed the effect of LOXL1-AS1 down regulation on H/R induced HCM pyroptosis. LOXL1-AS1 aggravated the MIRI by regulating miR-761/PTEN axis in vivo.
Conclusions
LOXL1-AS1 targeted miR-761 to regulate PTEN expression, then enhance cardiomyocyte pyroptosis, providing a new alternative target for the treatment of MIRI.

Keyword

Pyroptosis; Myocardial ischemia; Reperfusion injury

Figure

  • Figure 1 Knocking down LOXL1-AS1 improved H/R-induced human cardiomyocyte pyroptosis. (A) Using qRT-PCR to detect the LOXL1-AS1 level. (B) Using qRT-PCR to examine the LOXL1-AS1 knockdown effect. (C) Using qRT-PCR to test the LOXL1-AS1 level. (D) Using MTT to determine cell viability. (E) Using the Release Assay Kit to monitor the LDH level. (F) Using ELISA to detect IL-1β and IL-18 secretion. (G) Using Western blot to measure ASC, cleaved caspase-1, GSDMD-N, NLRP3, IL-1β, and IL-18 levels. (H) Using flow cytometry to monitor the pyroptosis level. (I) Using immunofluorescence to detect NLRP3 expression. Data were presented as mean±standard deviation, and 3 separate experiments were performed in triplicate.ELISA = enzyme-linked immunosorbent assay; GAPDH = glyceraldehyde-3-phosphate dehydrogenase; GSDMD = gasdermin D; H/R = hypoxia/reoxygenation; IL = interleukin; LDH = lactate dehydrogenase; LOXL1-AS1 = lysyl oxidase like 1 antisense RNA 1; NLRP3 = NOD-like receptor pyrin domain-containing protein 3; qRT-PCR = quantitative real-time polymerase chain reaction.*p<0.05; **p<0.01; ***p<0.001.

  • Figure 2 LOXL1-AS1 negatively regulated the miR-761 level as a molecular sponge. (A) Using bioinformatics to predict LOXL1-AS1 and miR-761 binding sites. (B) Using qRT-PCR to examine miR-761 knockdown and overexpression effects. (C) Using dual-luciferase to test LOXL1-AS1 and miR-761 interaction. (D) After interfering with LOXL1-AS1 in HCMs, qRT-PCR is used to monitor the miR-761 level. (E) Under H/R conditions, LOXL1-AS1 was knocked down in HCMs, and qRT-PCR was used to examine the miR-761 level. Data were presented as mean±standard deviation, and 3 separate experiments were performed in triplicate.H/R = hypoxia/reoxygenation; HCM = human cardiomyocyte; LOXL1-AS1 = lysyl oxidase like 1 antisense RNA 1; MUT = mutant type; qRT-PCR = quantitative real-time polymerase chain reaction; WT = wild-type.**p<0.01; ***p<0.001.

  • Figure 3 MiR-761 overexpression improved H/R-induced human cardiomyocyte pyroptosis. (A) Using quantitative real-time polymerase chain reaction to examine the miR-761 level. (B) Using MTT to examine cell viability. (C) Using the Release Assay Kit to detect the LDH level. (D) Using enzyme-linked immunosorbent assay to determine IL-1β and IL-18 levels. (E) Using Western blot to monitor ASC, cleaved caspase-1, GSDMD-N, NLRP3, IL-1β, and IL-18 levels. (F) Using flow cytometry to detect caspase-1 activity. (G) Using immunofluorescence to detect NLRP3 expression. Data were presented as mean±standard deviation, and 3 separate experiments were performed in triplicate.GAPDH = glyceraldehyde-3-phosphate dehydrogenase; GSDMD = gasdermin D; HR = hypoxia/reoxygenation; IL = interleukin; LDH = lactate dehydrogenase; NLRP3 = NOD-like receptor pyrin domain-containing protein 3.*p<0.05; **p<0.01; ***p<0.001.

  • Figure 4 LOXL1-AS1 induced human cardiomyocyte pyroptosis by targeting miR-761 under H/R conditions. (A) Using quantitative real-time polymerase chain reaction to measure the miR-761 level. (B) Using MTT to detect cell viability. (C) Using the Release Assay Kit to test the LDH level. (D) Using enzyme-linked immunosorbent assay to monitor IL-1β and IL-18 levels. (E) Using Western blot to determine ASC, cleaved caspase-1, GSDMD-N, NLRP3, IL-1β, and IL-18 levels. (F) Using flow cytometry to examine caspase-1 activity. (G) Using immunofluorescence to detect NLRP3 expression. Data were presented as mean±standard deviation, and 3 separate experiments were performed in triplicate.GAPDH = glyceraldehyde-3-phosphate dehydrogenase; GSDMD = gasdermin D; H/R = hypoxia/reoxygenation; IL = interleukin; LDH = lactate dehydrogenase; LOXL1-AS1 = lysyl oxidase like 1 antisense RNA 1; NLRP3 = NOD-like receptor pyrin domain-containing protein 3.*p<0.05; **p<0.01; ***p<0.001.

  • Figure 5 MiR-761 targeted PTEN. (A) Using bioinformatics to predict miR-761 and PTEN binding sites. (B) Using dual-luciferase to verify the miR-761 and PTEN mutually binding relationship. (C, D) Using qRT-PCR and Western blot to determine the PTEN level. (E, F) Using qRT-PCR and Western blot to monitor the PTEN level. Data were presented as mean±standard deviation, and 3 separate experiments were performed in triplicate.GAPDH = glyceraldehyde-3-phosphate dehydrogenase; H/R = hypoxia/reoxygenation; MUT = mutant type; PTEN = phosphatase and tensin homolog; qRT-PCR = quantitative real-time polymerase chain reaction; WT = wild-type.***p<0.001.

  • Figure 6 LOXL1-AS1 enhanced myocardial ischemia and reperfusion injury by regulating miR-761 in mice. (A, B) Using qRT-PCR to monitor LOXL1-AS1 and miR-761 levels. (C) Using HE staining to measure changes in the myocardial tissue morphology (scale bar: 50 μm). (D-F) Using the Release Assay Kit to detect LDH, CK-MB, and cTnI levels in serum. (G, H) Using qRT-PCR to determine LOXL1-AS1 and miR-761 levels. (I, J) Using qRT-PCR and Western blot to monitor the PTEN level. (K) Using ELISA to measure IL-1β and IL-18 levels. (L) Using Western blot to examine ASC, cleaved caspase-1, GSDMD-N, NLRP3, IL-1β, and IL-18 levels. (M) Immunohistochemistry detection of NLRP3 and caspase-1 expression in the myocardial tissue (scale bar: 50 μm). Data are presented as mean±standard deviation (n=5).CK-ME = creatine kinase-MB; cTnI = cardiac troponin I; GAPDH = glyceraldehyde-3-phosphate dehydrogenase; GSDMD = gasdermin D; HE = hematoxylin and eosin; IL = interleukin; I/R = ischemia/reperfusion; LDH = lactate dehydrogenase; LOXL1-AS1 = lysyl oxidase like 1 antisense RNA 1; NLRP3 = NOD-like receptor pyrin domain-containing protein 3; PTEN = phosphatase and tensin homolog; qRT-PCR = quantitative real-time polymerase chain reaction.***p<0.001.


Reference

1. Casin KM, Calvert JW. Dynamic regulation of cysteine oxidation and phosphorylation in myocardial ischemia-reperfusion injury. Cells. 2021; 10:2388. PMID: 34572037.
2. Toldo S, Mauro AG, Cutter Z, Abbate A. Inflammasome, pyroptosis, and cytokines in myocardial ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol. 2018; 315:H1553–H1568. PMID: 30168729.
3. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009; 7:99–109. PMID: 19148178.
4. Fang Y, Tian S, Pan Y, et al. Pyroptosis: a new frontier in cancer. Biomed Pharmacother. 2020; 121:109595. PMID: 31710896.
5. Zhang J, Huang L, Shi X, et al. Metformin protects against myocardial ischemia-reperfusion injury and cell pyroptosis via AMPK/NLRP3 inflammasome pathway. Aging (Albany NY). 2020; 12:24270–24287. PMID: 33232283.
6. Ye B, Chen X, Dai S, et al. Emodin alleviates myocardial ischemia/reperfusion injury by inhibiting gasdermin D-mediated pyroptosis in cardiomyocytes. Drug Des Devel Ther. 2019; 13:975–990.
7. Joshi M, Rajender S. Long non-coding RNAs (lncRNAs) in spermatogenesis and male infertility. Reprod Biol Endocrinol. 2020; 18:103. PMID: 33126901.
8. Sun Q, Li J, Li F, et al. LncRNA LOXL1-AS1 facilitates the tumorigenesis and stemness of gastric carcinoma via regulation of miR-708-5p/USF1 pathway. Cell Prolif. 2019; 52:e12687. PMID: 31468594.
9. Song B, Dang H, Dong R. Differential expression of LOXL1-AS1 in coronary heart disease and its regulatory mechanism in ox-LDL-induced human coronary artery endothelial cell pyroptosis. Cardiovasc Drugs Ther. 2023; 37:75–87. PMID: 34633594.
10. Bushati N, Cohen SM. MicroRNA functions. Annu Rev Cell Dev Biol. 2007; 23:175–205. PMID: 17506695.
11. Ding S, Liu D, Wang L, Wang G, Zhu Y. Inhibiting microRNA-29a protects myocardial ischemia-reperfusion injury by targeting sirt1 and suppressing oxidative stress and NLRP3-mediated pyroptosis pathway. J Pharmacol Exp Ther. 2020; 372:128–135. PMID: 31481517.
12. Zhou Y, Li KS, Liu L, Li SL. MicroRNA-132 promotes oxidative stress-induced pyroptosis by targeting sirtuin 1 in myocardial ischaemia-reperfusion injury. Int J Mol Med. 2020; 45:1942–1950. PMID: 32236570.
13. Wang C, Yang W, Liang X, et al. MicroRNA-761 modulates foam cell formation and inflammation through autophagy in the progression of atherosclerosis. Mol Cell Biochem. 2020; 474:135–146. PMID: 32772311.
14. Xiang X, Zheng L, Li X. Silencing of long noncoding RNA zinc finger antisense 1 protects against hypoxia/reoxygenation-induced injury in HL-1 cells through targeting the miR-761/cell death inducing p53 target 1 axis. J Cardiovasc Pharmacol. 2020; 76:564–573. PMID: 32833901.
15. Long B, Wang K, Li N, et al. MiR-761 regulates the mitochondrial network by targeting mitochondrial fission factor. Free Radic Biol Med. 2013; 65:371–379. PMID: 23867156.
16. Frank A, Bonney M, Bonney S, Weitzel L, Koeppen M, Eckle T. Myocardial ischemia reperfusion injury: from basic science to clinical bedside. Semin Cardiothorac Vasc Anesth. 2012; 16:123–132. PMID: 22368166.
17. Bian Y, Li X, Pang P, et al. Kanglexin, a novel anthraquinone compound, protects against myocardial ischemic injury in mice by suppressing NLRP3 and pyroptosis. Acta Pharmacol Sin. 2020; 41:319–326. PMID: 31645662.
18. Liang YP, Liu Q, Xu GH, et al. The lncRNA ROR/miR-124-3p/TRAF6 axis regulated the ischaemia reperfusion injury-induced inflammatory response in human cardiac myocytes. J Bioenerg Biomembr. 2019; 51:381–392. PMID: 31768721.
19. Tan JK, Ma XF, Wang GN, Jiang CR, Gong HQ, Liu H. LncRNA MIAT knockdown alleviates oxygen-glucose deprivation-induced cardiomyocyte injury by regulating JAK2/STAT3 pathway via miR-181a-5p. J Cardiol. 2021; 78:586–597. PMID: 34489160.
20. Sun J, Zhu YM, Liu Q, et al. LncRNA ROR modulates myocardial ischemia-reperfusion injury mediated by the miR-185-5p/CDK6 axis. Lab Invest. 2022; 102:505–514. PMID: 35066566.
21. Xie Q, Li F, Shen K, Luo C, Song G. LOXL1-AS1/miR-515-5p/STAT3 positive feedback loop facilitates cell proliferation and migration in atherosclerosis. J Cardiovasc Pharmacol. 2020; 76:151–158. PMID: 32453072.
22. Li S, Sun Y, Song M, et al. NLRP3/caspase-1/GSDMD-mediated pyroptosis exerts a crucial role in astrocyte pathological injury in mouse model of depression. JCI Insight. 2021; 6:e146852. PMID: 34877938.
23. Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015; 526:660–665. PMID: 26375003.
24. Wang Y, Liu Y, Fei A, Yu Z. LncRNA XIST facilitates hypoxia-induced myocardial cell injury through targeting miR-191-5p/TRAF3 axis. Mol Cell Biochem. 2022; 477:1697–1707. PMID: 35257270.
25. Mo L, Jiang HB, Tian GR, Lu GJ. The proliferation and migration of atherosclerosis-related HVSMCs were inhibited by downregulation of lncRNA XIST via regulation of the miR-761/BMP9 axis. Kaohsiung J Med Sci. 2022; 38:18–29. PMID: 34595819.
26. Ruan H, Li J, Ren S, et al. Inducible and cardiac specific PTEN inactivation protects ischemia/reperfusion injury. J Mol Cell Cardiol. 2009; 46:193–200. PMID: 19038262.
27. Li G, Yang J, Yang C, et al. PTENα regulates mitophagy and maintains mitochondrial quality control. Autophagy. 2018; 14:1742–1760. PMID: 29969932.
28. Cui Q, Wang J, Liu X, Wang X, Su G. Knockout of PTEN improves cardiac function and inhibits NLRP3-mediated cardiomyocyte pyroptosis in rats with myocardial ischemia-reperfusion. Xibao Yu Fenzi Mianyixue Zazhi. 2020; 36:205–211. PMID: 32389167.
29. Dai ZH, Jiang ZM, Tu H, et al. MiR-129 attenuates myocardial ischemia reperfusion injury by regulating the expression of PTEN in rats. BioMed Res Int. 2021; 2021:5535788. PMID: 34435045.
Full Text Links
  • KCJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr