Abstract

Abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs) is a critical pathological feature in the pathogenesis of pulmonary arterial hypertension (PAH), but the regulatory mechanisms remain largely unknown. Herein, we demonstrated that interferon regulatory factor 9 (IRF9) accelerated PASMCs proliferation by regulating Prohibitin 1 (PHB1) expression and the AKT-GSK3β signaling pathway. Compared with control groups, the rats treated with chronic hypoxia (CH), monocrotaline (MCT) or sugen5416 combined with chronic hypoxia (SuHx), and mice challenged with CH had significantly thickened pulmonary arterioles and hyperproliferative PASMCs. More importantly, the protein level of IRF9 was found to be elevated in the thickened medial wall of the pulmonary arterioles in all of these PAH models. Notably, overexpression of IRF9 significantly promoted the proliferation of rat and human PASMCs, as evidenced by increased cell counts, EdU-positive cells and upregulated biomarkers of cell proliferation. In contrast, knockdown of IRF9 suppressed the proliferation of rat and human PASMCs. Mechanistically, IRF9 directly restrained PHB1 expression and interacted with AKT to inhibit the phosphorylation of AKT at thr308 site, which finally led to mitochondrial dysfunction and PASMC proliferation. Unsurprisingly, MK2206, a specific inhibitor of AKT, partially reversed the PASMC proliferation inhibited by IRF9 knockdown. Thus, our results suggested that elevation of IRF9 facilitates PASMC proliferation by regulating PHB1 expression and AKT signaling pathway to affect mitochondrial function during the development of PAH, which indicated that targeting IRF9 may serve as a novel strategy to delay the pathological progression of PAH.

Highlights

  • Pulmonary arterial hypertension (PAH) is a progressive and devastating lung disease that is precipitated by pulmonary arteriole remodeling, leading to pulmonary vascular resistance and right heart failure (RHF) (Leopold and Maron, 2016)

  • To investigate the involvement of interferon regulatory factor 9 (IRF9) in pulmonary arterial hypertension (PAH), we generated a variety of PAH models of chronic hypoxia (CH)-induced rats or mice, and monocrotaline (MCT), sugen5416 combined with chronic hypoxia (SuHx)-induced rats

  • The results showed that right ventricle systolic pressure (RVSP), Fulton’s mass index (RV/ left ventricle (LV) + S), right ventricular wall thickness at end-diastole (RVWd), right ventricular wall thickness at end-systole (RVWs), interventricular septal thickness at diastole (IVSd), and interventricular septal thickness at systole (IVSs) were increased in hypoxia-treated rats compared with normoxiatreated rats (Supplementary Figures S1A–C), while right ventricular inside diameter at end-diastole (RVIDd) and right ventricular inside diameter at end-systole (RVIDs) showed no significant difference between groups (Supplementary Figure S1C)

Read more

Summary

Introduction

Pulmonary arterial hypertension (PAH) is a progressive and devastating lung disease that is precipitated by pulmonary arteriole remodeling, leading to pulmonary vascular resistance and right heart failure (RHF) (Leopold and Maron, 2016). PAH is a vasculopathy that is histopathologically characterized by excessive pulmonary vasoconstriction, abnormal arteriole remodeling and plexiform lesions (Schermuly et al, 2011) In these pathological processes, the cellular phenotype of pulmonary artery smooth muscle cells (PASMCs) changes from a quiescent state to a hyperproliferative state (Leopold and Maron, 2016), and the cancer-like phenotype and metabolic shift are closely aligned with the “Warburg effect” (Vander Heiden et al, 2009). The cellular phenotype of pulmonary artery smooth muscle cells (PASMCs) changes from a quiescent state to a hyperproliferative state (Leopold and Maron, 2016), and the cancer-like phenotype and metabolic shift are closely aligned with the “Warburg effect” (Vander Heiden et al, 2009) In these cells, mitochondrial glucose oxidation is suppressed, whereas glycolysis is utilized as the major source of adenosine triphosphate production

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call