M2 macrophage‑derived exosomes alleviate KCa3.1 channel expression in rapidly paced HL‑1 myocytes via the NF‑κB (p65)/STAT3 signaling pathway

Mol Med Rep. 2024 Apr;29(4):55. doi: 10.3892/mmr.2024.13179. Epub 2024 Feb 9.

Abstract

The present study was designed to explore the role of M2 macrophage‑derived exosomes (M2‑exos) on the KCa3.1 channel in a cellular atrial fibrillation (AF) model using rapidly paced HL‑1 myocytes. M2 macrophages and M2‑exos were isolated and identified. MicroRNA (miR)‑146a‑5p levels in M2 macrophages and M2‑exos were quantified using reverse transcription‑quantitative PCR (RT‑qPCR). HL‑1 myocytes were randomly divided into six groups: Control group, pacing group, pacing + coculture group (pacing HL‑1 cells cocultured with M2‑exos), pacing + mimic‑miR‑146a‑5p group, pacing + NC‑miR‑146a‑5p group and pacing + pyrrolidine dithiocarbamate (PDTC; a special blocker of the NF‑κB signaling pathway) group. Transmission electron microscopy, nanoparticle tracking analysis, western blotting, RT‑qPCR and immunohistochemistry were performed in the present study. A whole‑cell clamp was also applied to record the current density of KCa3.1 and action potential duration (APD) in each group. The results revealed that miR‑146a‑5p was highly expressed in both M2 macrophages and M2‑exos. Pacing HL‑1 cells led to a shorter APD, an increased KCa3.1 current density and higher protein levels of KCa3.1, phosphorylated (p‑)NF‑κB p65, p‑STAT3 and IL‑1β compared with the control group. M2‑exos, miR‑146a‑5p‑mimic and PDTC both reduced the protein expression of KCa3.1, p‑NF‑κB p65, p‑STAT3 and IL‑1β and the current density of KCa3.1, resulting in a longer APD in the pacing HL‑1 cells. In conclusion, M2‑exos and their cargo, which comprised miR‑146a‑5p, decreased KCa3.1 expression and IL‑1β secretion in pacing HL‑1 cells via the NF‑κB/STAT3 signaling pathway, limiting the shorter APD caused by rapid pacing.

Keywords: KCa3.1; M2 macrophages; atrial fibrillation; exosomes.

MeSH terms

  • Animals
  • Atrial Fibrillation* / metabolism
  • Cardiac Pacing, Artificial
  • Cell Line
  • Exosomes* / metabolism
  • Humans
  • Macrophages / metabolism
  • Mice
  • MicroRNAs* / genetics
  • MicroRNAs* / metabolism
  • Myocytes, Cardiac / metabolism
  • NF-kappa B / metabolism
  • Proline* / analogs & derivatives
  • STAT3 Transcription Factor / metabolism
  • Signal Transduction
  • Thiocarbamates*

Substances

  • MicroRNAs
  • NF-kappa B
  • Proline
  • prolinedithiocarbamate
  • STAT3 protein, human
  • STAT3 Transcription Factor
  • Thiocarbamates
  • Kcnn4 protein, mouse

Grants and funding

The present study was supported by the National Natural Science Foundation of China (grant nos. 81970277 and 82170312).