4.7 Article

The redox mechanism for vascular barrier dysfunction associated with metabolic disorders: Glutathionylation of Rac1 in endothelial cells

Journal

REDOX BIOLOGY
Volume 9, Issue -, Pages 306-319

Publisher

ELSEVIER SCIENCE BV
DOI: 10.1016/j.redox.2016.09.003

Keywords

Protein S-glutathionylation; Glutaredoxin-1; Actin cytoskeleton; Small Rho GTPase Rac1; Endothelial barrier function; ApoE-deficient mice

Funding

  1. AHA [SDG20140036, 1UL1TR001430, T32 HL07224, R37 HL104017]

Ask authors/readers for more resources

Background: Oxidative stress is implicated in increased vascular permeability associated with metabolic disorders, but the underlying redox mechanism is poorly defined. S-glutathionylation, a stable adduct of glutathione with protein sulfhydryl, is a reversible oxidative modification of protein and is emerging as an important redox signaling paradigm in cardiovascular physiopathology. The present study determines the role of protein S-glutathionylation in metabolic stress-induced endothelial cell permeability. Methods and results: In endothelial cells isolated from patients with type-2 diabetes mellitus, protein S-glutathionylation level was increased. This change was also observed in aortic endothelium in ApoE deficient (ApoE(-I-)) mice fed on Western diet. Metabolic stress-induced protein S-glutathionylation in human aortic endothelial cells (HAEC) was positively correlated with elevated endothelial cell permeability, as reflected by disassembly of cell-cell adherens junctions and cortical actin structures. These impairments were reversed by adenoviral overexpression of a specific de-glutathionylation enzyme, glutaredoxin-1 in cultured HAECs. Consistently, transgenic overexpression of human Glrx-1 in ApoE(-/-)mice fed the Western diet attenuated endothelial protein S-glutathionylation, actin cytoskeletal disorganization, and vascular permeability in the aorta. Mechanistically, glutathionylation and inactivation of Rac1, a small RhoGPase, were associated with endothelial hyperpermeability caused by metabolic stress. Glutathionylation of Rac1 on cysteine 81 and 157 located adjacent to guanine nucleotide binding site was required for the metabolic stress to inhibit Racl activity and promote endothelial hyperpermeability. Conclusions: Glutathionylation and inactivation of Racl in endothelial cells represent a novel redox mechanism of vascular barrier dysfunction associated with metabolic disorders. (C) 2016 The Authors. Published by Elsevier B.V.

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