Endothelial PPAR-γ provides vascular protection from IL-1β-induced oxidative stress

Am J Physiol Heart Circ Physiol. 2016 Jan 1;310(1):H39-48. doi: 10.1152/ajpheart.00490.2015. Epub 2015 Nov 13.

Abstract

Loss of peroxisome proliferator-activated receptor (PPAR)-γ function in the vascular endothelium enhances atherosclerosis and NF-κB target gene expression in high-fat diet-fed apolipoprotein E-deficient mice. The mechanisms by which endothelial PPAR-γ regulates inflammatory responses and protects against atherosclerosis remain unclear. To assess functional interactions between PPAR-γ and inflammation, we used a model of IL-1β-induced aortic dysfunction in transgenic mice with endothelium-specific overexpression of either wild-type (E-WT) or dominant negative PPAR-γ (E-V290M). IL-1β dose dependently decreased IκB-α, increased phospho-p65, and increased luciferase activity in the aorta of NF-κB-LUC transgenic mice. IL-1β also dose dependently reduced endothelial-dependent relaxation by ACh. The loss of ACh responsiveness was partially improved by pretreatment of the vessels with the PPAR-γ agonist rosiglitazone or in E-WT. Conversely, IL-1β-induced endothelial dysfunction was worsened in the aorta from E-V290M mice. Although IL-1β increased the expression of NF-κB target genes, NF-κB p65 inhibitor did not alleviate endothelial dysfunction induced by IL-1β. Tempol, a SOD mimetic, partially restored ACh responsiveness in the IL-1β-treated aorta. Notably, tempol only modestly improved protection in the E-WT aorta but had an increased protective effect in the E-V290M aorta compared with the aorta from nontransgenic mice, suggesting that PPAR-γ-mediated protection involves antioxidant effects. IL-1β increased ROS and decreased the phospho-endothelial nitric oxide synthase (Ser(1177))-to-endothelial nitric oxide synthase ratio in the nontransgenic aorta. These effects were completely abolished in the aorta with endothelial overexpression of WT PPAR-γ but were worsened in the aorta with E-V290M even in the absence of IL-1β. We conclude that PPAR-γ protects against IL-1β-mediated endothelial dysfunction through a reduction of oxidative stress responses but not by blunting IL-1β-mediated NF-κB activity.

Keywords: endothelium; inflammation; interleukin-1β; peroxisome proliferator-activated receptor-γ; vascular dysfunction.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Antioxidants / pharmacology
  • Aorta / drug effects*
  • Aorta / metabolism
  • Aorta / pathology
  • Aorta / physiopathology
  • Aortic Diseases / metabolism
  • Aortic Diseases / pathology
  • Aortic Diseases / physiopathology
  • Aortic Diseases / prevention & control*
  • Dose-Response Relationship, Drug
  • Endothelial Cells / drug effects*
  • Endothelial Cells / metabolism
  • Endothelial Cells / pathology
  • Endothelium, Vascular / drug effects*
  • Endothelium, Vascular / metabolism
  • Endothelium, Vascular / pathology
  • Endothelium, Vascular / physiopathology
  • Female
  • Gene Expression Regulation
  • Genotype
  • Humans
  • I-kappa B Proteins / metabolism
  • Inflammation Mediators / metabolism
  • Interleukin-1beta / pharmacology*
  • Male
  • Mice, Inbred C57BL
  • Mice, Transgenic
  • NF-KappaB Inhibitor alpha
  • Nitric Oxide Synthase Type III / metabolism
  • Oxidative Stress / drug effects*
  • PPAR gamma / agonists
  • PPAR gamma / genetics
  • PPAR gamma / metabolism*
  • Phenotype
  • Phosphorylation
  • Reactive Oxygen Species / metabolism
  • Signal Transduction / drug effects
  • Transcription Factor RelA / metabolism
  • Vasodilation / drug effects
  • Vasodilator Agents / pharmacology

Substances

  • Antioxidants
  • I-kappa B Proteins
  • Inflammation Mediators
  • Interleukin-1beta
  • NFKBIA protein, human
  • Nfkbia protein, mouse
  • PPAR gamma
  • Reactive Oxygen Species
  • Rela protein, mouse
  • Transcription Factor RelA
  • Vasodilator Agents
  • NF-KappaB Inhibitor alpha
  • Nitric Oxide Synthase Type III
  • Nos3 protein, mouse