Abstract

Cardiac fibrosis is a common pathological process in multiple cardiovascular diseases, including myocardial infarction (MI). Abnormal cardiac fibroblast (CF) activity is a key event in cardiac fibrosis. Although the Notch signaling pathway has been reported to play a vital role in protection from cardiac fibrosis, the exact mechanisms underlying cardiac fibrosis and protection from it have not yet been elucidated. Similarly, Hif1α and the RhoA/ROCK signaling pathway have been shown to participate in cardiac fibrosis. The RhoA/ROCK signaling pathway has been reported to be an upstream pathway of Hif1α in several pathophysiological processes. In the present study, we aimed to determine the effects of notch3 on CF activity and its relationship with the RhoA/ROCK/Hif1α signaling pathway. Using in vitro experiments, we demonstrated that notch3 inhibited CF proliferation and fibroblast to myofibroblast transition (FMT) and promoted CF apoptosis. A knockdown of notch3 using siRNAs had the exact opposite effect. Next, we found that notch3 regulated CF activity by negative regulation of the RhoA/ROCK/Hif1α signaling pathway. Extending CF-based studies to an in vivo rat MI model, we showed that overexpression of notch3 by the Ad-N3ICD injection attenuated the increase of RhoA, ROCK1, ROCK2, and Hif1α levels following MI and further prevented MI-induced cardiac fibrosis. On the basis of these results, we conclude that notch3 is involved in the regulation of several aspects of CF activity, including proliferation, FMT, and apoptosis, by inhibiting the RhoA/ROCK/Hif1α signaling pathway. These findings are significant to further our understanding of the pathogenesis of cardiac fibrosis and to ultimately identify new therapeutic targets for cardiac fibrosis, potentially based on the RhoA/ROCK/Hif1α signaling pathway.

Highlights

  • Heart failure (HF), secondary to ischemic or non-ischemic cardiomyopathy, is a serious health issue with high rates of associated morbidity and mortality

  • Our results showed that notch3 regulated cardiac fibroblast (CF) activity in vitro and cardiac fibrosis in vivo

  • We found that 2ME weakened the positive regulation of notch3 interference on α-SMA, Col I, Col III, and CF proliferation, whereas DMOG abolished the effect of notch3 on CF apoptosis

Read more

Summary

Introduction

Heart failure (HF), secondary to ischemic or non-ischemic cardiomyopathy, is a serious health issue with high rates of associated morbidity and mortality. CFs have a proliferative and migratory phenotype and exhibit enhanced secretion of ECM. Several proinflammatory and profibrotic factors are involved in the regulation of CF activity in cardiac fibrosis, such as NF-κB, bone morphogenetic protein, and TGF-β1 (Sun et al, 2013; Fix et al, 2019; Li A.Y. et al, 2019). These factors have been shown to be involved in similar pathological processes for several different etiologies (Harvey and Leinwand, 2011)

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call