4.7 Article

Targeting mitochondria-inflammation circle by renal denervation reduces atheroprone endothelial phenotypes and atherosclerosis

Journal

REDOX BIOLOGY
Volume 47, Issue -, Pages -

Publisher

ELSEVIER
DOI: 10.1016/j.redox.2021.102156

Keywords

Renal denervation; Endothelial dysfunction; Mitochondrial dysfunction; Inflammation; Atherosclerosis

Funding

  1. National Natural Science Founda-tion of China (NSFC) [81970303, 81973316]

Ask authors/readers for more resources

Chronic sympathetic hyperactivity can lead to oxidative stress and inflammation in endothelial cells, contributing to atherosclerosis development. Renal denervation (RDN) may protect endothelial cells by reducing oxidative stress-induced inflammation, thus reducing atherosclerosis.
Objective: The disruption of mitochondrial redox homeostasis in endothelial cells (ECs) can cause chronic inflammation, a substantial contributor to the development of atherosclerosis. Chronic sympathetic hyperactivity can enhance oxidative stress to induce endothelial dysfunction. We determined if renal denervation (RDN), the strategy reducing sympathetic tone, can protect ECs by ameliorating mitochondrial reactive oxygen species (ROS)-induced inflammation to reduce atherosclerosis. Methods and results: ApoE deficient (ApoE(-/-)) mice were conducted RDN or sham operation before 20-week high-fat diet feeding. Atherosclerosis, EC phenotype and mitochondrial morphology were determined. In vitro, human arterial ECs were treated with norepinephrine to determine the mechanisms for RDN-inhibited endothelial inflammation. RDN reduced atherosclerosis, EC mitochondrial oxidative stress and inflammation. Mechanistically, the chronic sympathetic hyperactivity increased circulating norepinephrine and mitochondrial monoamine oxidase A (MAO-A) activity. MAO-A activation-impaired mitochondrial homeostasis resulted in ROS accumulation and NF-kappa B activation, thereby enhancing expression of atherogenic and proinflammatory molecules in ECs. It also suppressed mitochondrial function regulator PGC-1 alpha, with involvement of NF-kappa B and oxidative stress. Inactivation of MAO-A by RDN disrupted the positive-feedback regulation between mitochondrial dysfunction and inflammation, thereby inhibiting EC atheroprone phenotypic alterations and atherosclerosis. Conclusions: The interplay between MAO-A-induced mitochondrial oxidative stress and inflammation in ECs is a key driver in atherogenesis, and it can be reduced by RDN.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available