Proinflammatory stimuli engage Brahma related gene 1 and Brahma in endothelial injury

Circ Res. 2013 Sep 27;113(8):986-96. doi: 10.1161/CIRCRESAHA.113.301296. Epub 2013 Aug 20.

Abstract

Rationale: Endothelial dysfunction inflicted by inflammation is found in a host of cardiovascular pathologies. One hallmark event in this process is the aggregation and adhesion of leukocyte to the vessel wall mediated by the upregulation of adhesion molecules (CAM) in endothelial cells at the transcriptional level. The epigenetic modulator(s) of CAM transactivation and its underlying pathophysiological relevance remain poorly defined.

Objective: Our goal was to determine the involvement of Brahma related gene 1 (Brg1) and Brahma (Brm) in CAM transactivation and its relevance in the pathogenesis of atherosclerosis.

Methods and results: In the present study, we report that proinflammatory stimuli augmented the expression of Brg1 and Brm in vitro in cultured endothelial cells and in vivo in arteries isolated from rodents. Overexpression of Brg1 and Brm promoted while knockdown of Brg1 and Brm abrogated transactivation of adhesion molecules and leukocyte adhesion induced by inflammatory signals. Brg1 and Brm interacted with and were recruited to the CAM promoters by nuclear factor κB/p65. Conversely, depletion of Brg1 and Brm disrupted the kinetics of p65 binding on CAM promoters and crippled CAM activation. Silencing of Brg1 and Brm also altered key epigenetic changes associated with CAM transactivation. Of intrigue, 17β-estradiol antagonized both the expression and activity of Brg1/Brm. Most importantly, endothelial-targeted elimination of Brg1/Brm conferred atheroprotective effects to Apoe(-/-) mice on a Western diet.

Conclusions: Our data suggest that Brg1 and Brm integrate various proinflammatory cues into CAM transactivation and endothelial malfunction and, as such, may serve as potential therapeutic targets in treating inflammation-related cardiovascular diseases.

Keywords: Brg1; atherosclerosis; endothelial dysfunction; epigenetics; inflammation; transcription.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Apolipoproteins E / deficiency
  • Apolipoproteins E / genetics
  • Binding Sites
  • Cell Adhesion Molecules / genetics
  • Cell Adhesion Molecules / metabolism
  • Cell Line, Tumor
  • DNA Helicases / genetics
  • DNA Helicases / metabolism*
  • Disease Models, Animal
  • Endothelial Cells / drug effects
  • Endothelial Cells / immunology
  • Endothelial Cells / metabolism*
  • Endothelial Cells / pathology
  • Endothelium, Vascular / drug effects
  • Endothelium, Vascular / immunology
  • Endothelium, Vascular / injuries
  • Endothelium, Vascular / metabolism*
  • Endothelium, Vascular / pathology
  • Epigenesis, Genetic
  • Estradiol / pharmacology
  • HEK293 Cells
  • Human Umbilical Vein Endothelial Cells / immunology
  • Human Umbilical Vein Endothelial Cells / metabolism
  • Human Umbilical Vein Endothelial Cells / pathology
  • Humans
  • Inflammation Mediators / metabolism*
  • Mice
  • Mice, Knockout
  • NF-kappa B / metabolism
  • Nuclear Proteins / genetics
  • Nuclear Proteins / metabolism*
  • Promoter Regions, Genetic
  • RNA Interference
  • Signal Transduction
  • Time Factors
  • Transcription Factors / genetics
  • Transcription Factors / metabolism*
  • Transcriptional Activation
  • Transfection
  • Vascular System Injuries / genetics
  • Vascular System Injuries / immunology
  • Vascular System Injuries / metabolism*
  • Vascular System Injuries / pathology

Substances

  • Apolipoproteins E
  • Cell Adhesion Molecules
  • Inflammation Mediators
  • NF-kappa B
  • Nuclear Proteins
  • SMARCA2 protein, human
  • Smarca2 protein, mouse
  • Transcription Factors
  • Estradiol
  • SMARCA4 protein, human
  • Smarca4 protein, mouse
  • DNA Helicases