LncRNA Rian reduces cardiomyocyte pyroptosis and alleviates myocardial ischemia-reperfusion injury by regulating by the miR-17-5p/CCND1 axis.
Myocardial ischemia-reperfusion injury (MIRI) is a pathological process characterized by cardiomyocyte death. Long noncoding RNAs (lncRNAs) have been shown to be dysregulated in the course of MIRI. Accordingly, the current study investigated the mechanism of lncRNA Rian in MIRI-induced cardiomyocyte pyroptosis. First, a murine model of MIRI was established by using the left anterior descending (LAD) coronary artery ligation method. Cardiac function and myocardial histopathological changes were evaluated by echocardiography and hematoxylin and eosin staining. Then, a cell model of MIRI was established by oxygen-glucose deprivation/reoxygenation (OGD/R), followed by analysis of NLRP3, cleaved caspase-1, and GSDMD-N levels by western blotting. The levels of IL-1β, IL-18, TNF-α, and IL-10 were measured using ELISA. LncRNA Rian, miR-17-5p, and CCND1 expression in myocardial tissues and OGD/R cells were examined using RT-qPCR. Finally, the binding relationships between Rian and miR-17-5p and miR-17-5p and CCND1 were validated with the help of dual-luciferase and RNA pull-down assays. Rian was poorly expressed in MIRI mice and OGD/R cells. LncRNA Rian overexpression reduced cardiomyocyte pyroptosis in vivo and in vitro, as indicated by decreased NLRP3, cleaved caspase-1, GSDMD-N, IL-1β, IL-18, and TNF-α levels and increased IL-10 levels. Furthermore, Rian bound to miR-17-5p and promoted CCND1 transcription. Notably, miR-17-5p overexpression or CCND1 silencing reversed the inhibitory effect of Rian overexpression on cardiomyocyte pyroptosis. Collectively, our findings indicate that Rian overexpression reduces cardiomyocyte pyroptosis and alleviates MIRI through the miR-17-5p/CCND1 axis.
Kang H
,Yu H
,Zeng L
,Ma H
,Cao G
... -
《-》
MicroRNA-495 Ameliorates Cardiac Microvascular Endothelial Cell Injury and Inflammatory Reaction by Suppressing the NLRP3 Inflammasome Signaling Pathway.
In recent years, microRNA-495 (miR-495) has been reported to be a tumor-suppressor miR that is down-modulated in cancers. However, its potential mechanism remains unknown. Therefore, this study aimed to demonstrate the role of miR-495 in cardiac microvascular endothelial cell (CMEC) injury and inflammatory reaction by mediating the pyrin domain-containing 3 (NLRP3) inflammasome signaling pathway.
Overall, 40 mice were assigned into myocardial ischemia/reperfusion injury (MIR) and sham groups. After model establishment, the levels of troponin T (TnT), troponin I (TnI), N-terminal pro-B-type natriuretic peptide (NT-proBNP), creatine kinase isoenzyme MB (CK-MB), myoglobin (MYO), tumor necrosis factor-alpha (TNF-α), and interleukin 1beta (IL-1β) were detected by Enzyme-Linked Immunosorbent Assay (ELISA). Apoptosis was evaluated using Terminal deoxy (d)-UTP nick end labeling (TUNEL) staining, the level of NLRP3 protein was determined by immunohistochemical assay, and miR-495 was detected by in situ hybridization (ISH). The infarct size was determined using 2, 3, 5-triphenyltetrazolium chloride (TTC) staining. The expression of miR-495 and the mRNA and protein levels of NLRP3, TNF-α, IL-1β, IL-18 and caspase-1 were evaluated by RT-qPCR and western blot analysis. After transfection, the cells were treated with a miR-495 mimic, a miR-495 inhibitor, or siNLRP3. Cell proliferation was measured by the 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and cell cycle and apoptosis by flow cytometry.
Mice with myocardial I/R injury had elevated levels of TnT, TnI, NT-proBNP, CK-MB, MYO, TNF-α and IL-1β; enhanced cell apoptosis; increased expression of NLRP3, TNF-α, IL-1β, IL-18 and caspase-1; and decreased miR-495 expression. MiR-495 was confirmed to target NLRP3. Moreover, miR-495 reduced the mRNA and protein levels of NLRP3, TNF-α, IL-1β, IL-18 and caspase-1, inhibited cell apoptosis and decreased cells at the G0/G1 phase while improving cell proliferation and increasing cells at the S phase. However, the effects of NLRP4 were proved to be reciprocal.
In conclusion, the current study indicated that miR-495 improved CMEC injury and inflammation by suppressing the NLRP3 inflammasome signaling pathway.
Zhou T
,Xiang DK
,Li SN
,Yang LH
,Gao LF
,Feng C
... -
《-》
MiR-129-5p Protects H9c2 Cardiac Myoblasts From Hypoxia/Reoxygenation Injury by Targeting TRPM7 and Inhibiting NLRP3 Inflammasome Activation.
As a biomarker for heart failure, miR-129-5p is abnormally expressed during myocardial I/R, but its specific functions and mechanisms remain largely unclear. Thus, this study explored the roles and possible mechanisms of miR-129-5p in hypoxia/reoxygenation (H/R)-insulted H9c2 cardiac myoblasts. After H/R insult, miR-129-5p expression levels were decreased, along with reduced cell viability and enhanced lactate dehydrogenase release in H9c2 cells. Overexpression of miR-129-5p through transfection of miR-129-5p mimics effectively improved cell viability and reduced lactate dehydrogenase release in H9c2 cells exposed to H/R, along with decreased apoptosis and caspase-3 activities. Moreover, miR-129-5p mimics inhibited reactive oxygen species production and upsurged superoxide dismutase activity in H9c2 cells exposed to H/R, and suppressed H/R-caused massive release of proinflammatory cytokines TNF-α and IL-1β. TRPM7 was identified as the target of miR-129-5p and was negatively regulated by miR-129-5p. TRPM7 overexpression counteracted the antagonistic effect of miR-129-5p on H/R-induced increase in intracellular calcium levels. TRPM7 overexpression also abolished miR-129-5p-induced elevation on cell viability and reduction on apoptosis as well as attenuated miR-129-5p-induced inhibition on reactive oxygen species and IL-1β production. Besides, H/R-induced NLRP3 inflammasome activation was inhibited by miR-129-5p mimic but reactivated by TRPM7. In conclusion, miR-129-5p alleviates H/R injury of H9c2 cardiomyocytes by targeting TRPM7 and inhibiting NLRP3 inflammasome activation, suggesting that miR-129-5p and TRPM7 may be potential therapeutic targets for myocardial I/R injury.
Liu S
,Liao Q
,Xu W
,Zhang Z
,Yin M
,Cao X
... -
《-》