Fibroblast-derived interleukin-6 exacerbates adverse cardiac remodeling after myocardial infarction

Myocardial infarction is one of the leading causes of mortality globally. Currently, the pleiotropic inflammatory cytokine interleukin-6 (IL-6) is considered to be intimately related to the severity of myocardial injury during myocardial infarction. Interventions targeting IL-6 are a promising thera...

Full description

Saved in:
Bibliographic Details
Published inThe Korean journal of physiology & pharmacology Vol. 28; no. 3; pp. 285 - 294
Main Authors Li, Hongkun, Bian, Yunfei
Format Journal Article
LanguageEnglish
Korean
Published Korea (South) The Korean Physiological Society and The Korean Society of Pharmacology 01.05.2024
대한약리학회
Subjects
Online AccessGet full text

Cover

Loading…
More Information
Summary:Myocardial infarction is one of the leading causes of mortality globally. Currently, the pleiotropic inflammatory cytokine interleukin-6 (IL-6) is considered to be intimately related to the severity of myocardial injury during myocardial infarction. Interventions targeting IL-6 are a promising therapeutic option for myocardial infarction, but the underlying molecular mechanisms are not well understood. Here, we report the novel role of IL-6 in regulating adverse cardiac remodeling mediated by fibroblasts in a mouse model of myocardial infarction. It was found that the elevated expression of IL-6 in myocardium and cardiac fibroblasts was observed after myocardial infarction. Further, fibroblast-specific knockdown of significantly attenuated cardiac fibrosis and adverse cardiac remodeling and preserved cardiac function induced by myocardial infarction. Mechanistically, the role of Il6 contributing to cardiac fibrosis depends on signal transduction and activation of transcription (STAT)3 signaling activation. Additionally, Stat3 binds to the promoter region and contributes to the increased expression of , which exacerbates cardiac fibrosis. In conclusion, these results suggest a novel role for IL-6 derived from fibroblasts in mediating Stat3 activation and substantially augmented expression in promoting cardiac fibrosis, highlighting its potential as a therapeutic target for cardiac fibrosis.
Bibliography:ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
KISTI1.1003/JNL.JAKO202413757621762
Author contributions: H.L. performed the experiments, and collected and analyzed the data. Y.B. conceived and designed the experiments.
ISSN:1226-4512
2093-3827
DOI:10.4196/kjpp.2024.28.3.285