International Journal of Molecular Sciences (Mar 2021)

Implication of EZH2 in the Pro-Proliferative and Apoptosis-Resistant Phenotype of Pulmonary Artery Smooth Muscle Cells in PAH: A Transcriptomic and Proteomic Approach

  • Karima Habbout,
  • Junichi Omura,
  • Charifa Awada,
  • Alice Bourgeois,
  • Yann Grobs,
  • Vinod Krishna,
  • Sandra Breuils-Bonnet,
  • Eve Tremblay,
  • Ghada Mkannez,
  • Sandra Martineau,
  • Valérie Nadeau,
  • Florence Roux-Dalvai,
  • Mark Orcholski,
  • Jey Jeyaseelan,
  • David Gutstein,
  • François Potus,
  • Steeve Provencher,
  • Sébastien Bonnet,
  • Roxane Paulin,
  • Olivier Boucherat

DOI
https://doi.org/10.3390/ijms22062957
Journal volume & issue
Vol. 22, no. 6
p. 2957

Abstract

Read online

Pulmonary arterial hypertension (PAH) is a progressive disorder characterized by a sustained elevation of pulmonary artery (PA) pressure, right ventricular failure, and premature death. Enhanced proliferation and resistance to apoptosis (as seen in cancer cells) of PA smooth muscle cells (PASMCs) is a major pathological hallmark contributing to pulmonary vascular remodeling in PAH, for which current therapies have only limited effects. Emerging evidence points toward a critical role for Enhancer of Zeste Homolog 2 (EZH2) in cancer cell proliferation and survival. However, its role in PAH remains largely unknown. The aim of this study was to determine whether EZH2 represents a new factor critically involved in the abnormal phenotype of PAH-PASMCs. We found that EZH2 is overexpressed in human lung tissues and isolated PASMCs from PAH patients compared to controls as well as in two animal models mimicking the disease. Through loss- and gain-of-function approaches, we showed that EZH2 promotes PAH-PASMC proliferation and survival. By combining quantitative transcriptomic and proteomic approaches in PAH-PASMCs subjected or not to EZH2 knockdown, we found that inhibition of EZH2 downregulates many factors involved in cell-cycle progression, including E2F targets, and contributes to maintain energy production. Notably, we found that EZH2 promotes expression of several nuclear-encoded components of the mitochondrial translation machinery and tricarboxylic acid cycle genes. Overall, this study provides evidence that, by overexpressing EZH2, PAH-PASMCs remove the physiological breaks that normally restrain their proliferation and susceptibility to apoptosis and suggests that EZH2 or downstream factors may serve as therapeutic targets to combat pulmonary vascular remodeling.

Keywords