LncRNA XIST shuttled by adipose tissue-derived mesenchymal stem cell-derived extracellular vesicles suppresses myocardial pyroptosis in atrial fibrillation by disrupting miR-214-3p-mediated Arl2 inhibition

Lab Invest. 2021 Nov;101(11):1427-1438. doi: 10.1038/s41374-021-00635-0. Epub 2021 Aug 13.

Abstract

The mechanisms underlying atrial fibrillation (AF), a type of heart arrhythmia, have not been fully identified. Long noncoding RNAs (lncRNAs) have been implicated in the progression of AF. The current study aimed to ascertain the means by which X-inactive specific transcript (XIST), a lncRNA, contributes to the pathogenesis of AF in an animal model or in atrial myocytes. Extracellular vesicles (EVs) derived from mouse adipose tissue-derived mesenchymal stem cells (AMSCs) were isolated, transfected with XIST, and either injected into AF mouse models or incubated with atrial myocytes. The in vitro and in vivo effects of EV-derived XIST on myocardial pyroptosis were determined by Western blot analysis of pyroptosis-related protein and an ELISA for inflammatory factors. Bioinformatics analysis revealed a relationship between XIST, microRNA (miR)-214-3p, and Arl2, which was subsequently verified by a dual luciferase assay and RNA immunoprecipitation. Functional experiments were performed to elucidate whether changes in miR-214-3p or Arl2 regulated the effect of XIST on myocardial pyroptosis. Overexpressed XIST from AMSC-EVs were found to decrease myocardial pyroptosis while alleviating inflammation, which was demonstrated by reduced expression of nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3), apoptosis-associated speck-like protein containing a CARD (ASC), cleared-caspase-1/caspase-1 and gasdermin D (GSDMD), as well as the amount of interleukin (IL)-1β and IL-18 in both the cardiomyocytes and AF mouse tissues. Mechanistically, XIST is a competing endogenous RNA (ceRNA) of miR-214-3p, triggering upregulation of its target gene Arl2. Silencing of Arl2 or overexpression miR-214-3p reversed the effects of XIST on inflammation and pyroptosis. Taken together, the key findings of our study suggest that XIST may blunt myocardial pyroptosis by absorbing miR-214-3p to promote Arl2 expression, providing encouraging insight into XIST-based targeted therapy for AF.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Atrial Fibrillation / etiology*
  • Atrial Fibrillation / metabolism
  • Cell Line
  • Cell-Derived Microparticles / metabolism
  • Down-Regulation
  • GTP-Binding Proteins / metabolism*
  • Mesenchymal Stem Cells / metabolism
  • Mice
  • MicroRNAs / metabolism*
  • NLR Family, Pyrin Domain-Containing 3 Protein / metabolism
  • Pyroptosis*
  • RNA, Long Noncoding / metabolism*

Substances

  • MicroRNAs
  • Mirn214 microRNA, mouse
  • NLR Family, Pyrin Domain-Containing 3 Protein
  • Nlrp3 protein, mouse
  • RNA, Long Noncoding
  • XIST non-coding RNA
  • Arl2 protein, mouse
  • GTP-Binding Proteins