Clinical and Translational Medicine (Apr 2022)

Extracellular vesicle‐packaged mitochondrial disturbing miRNA exacerbates cardiac injury during acute myocardial infarction

  • Ping Sun,
  • Chao Wang,
  • Ge Mang,
  • Xiangli Xu,
  • Shuai Fu,
  • Jianfeng Chen,
  • Xiaoqi Wang,
  • Weiwei Wang,
  • Hairu Li,
  • Peng Zhao,
  • Yifei Li,
  • Qi Chen,
  • Naixin Wang,
  • Zhonghua Tong,
  • Xin Fu,
  • Ying Lang,
  • Shasha Duan,
  • Dongmei Liu,
  • Maomao Zhang,
  • Jiawei Tian

DOI
https://doi.org/10.1002/ctm2.779
Journal volume & issue
Vol. 12, no. 4
pp. n/a – n/a

Abstract

Read online

Abstract Mounting evidence suggests that extracellular vesicles (EVs) are effective communicators in biological signalling in cardiac physiology and pathology. However, the role of EVs in cardiac injury, particularly in ischemic myocardial scenarios, has not been fully elucidated. Here, we report that acute myocardial infarction (AMI)‐induced EVs can impair cardiomyocyte survival and exacerbate cardiac injury. EV‐encapsulated miR‐503, which is enriched during the early phase of AMI, is a critical molecule that mediates myocardial injury. Functional studies revealed that miR‐503 promoted cardiomyocyte death by directly binding to peroxisome proliferator‐activated receptor gamma coactivator‐1β (PGC‐1β) and a mitochondrial deacetylase, sirtuin 3 (SIRT3), thereby triggering mitochondrial metabolic dysfunction and cardiomyocyte death. Mechanistically, we identified endothelial cells as the primary source of miR‐503 in EVs after AMI. Hypoxia induced rapid H3K4 methylation of the promoter of the methyltransferase‐like 3 gene (METTL3) and resulted in its overexpression. METTL3 overexpression evokes N6‐methyladenosine (m6A)‐dependent miR‐503 biogenesis in endothelial cells. In summary, this study highlights a novel endogenous mechanism wherein EVs aggravate myocardial injury during the onset of AMI via endothelial cell‐secreted miR‐503 shuttling.

Keywords