1. Academic Validation
  2. Mitochondrial Damage-Induced Innate Immune Activation in Vascular Smooth Muscle Cells Promotes Chronic Kidney Disease-Associated Plaque Vulnerability

Mitochondrial Damage-Induced Innate Immune Activation in Vascular Smooth Muscle Cells Promotes Chronic Kidney Disease-Associated Plaque Vulnerability

  • Adv Sci (Weinh). 2021 Jan 6;8(5):2002738. doi: 10.1002/advs.202002738.
Xianjin Bi 1 Changhong Du 2 Xinmiao Wang 2 Xue-Yue Wang 3 Wenhao Han 1 Yue Wang 1 Yu Qiao 1 Yingguo Zhu 1 Li Ran 1 Yong Liu 1 Jiachuan Xiong 1 Yinghui Huang 1 Mingying Liu 1 Chi Liu 1 Chunyu Zeng 4 Junping Wang 2 Ke Yang 1 Jinghong Zhao 1
Affiliations

Affiliations

  • 1 Department of Nephrology the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing Kidney Center of PLA Xinqiao Hospital Army Medical University (Third Military Medical University) Chongqing 400037 China.
  • 2 State Key Laboratory of Trauma Burns and Combined Injury Institute of Combined Injury Chongqing Engineering Research Center for Nanomedicine College of Preventive Medicine Army Medical University (Third Military Medical University) Chongqing 400038 China.
  • 3 Laboratory of Stem Cell & Developmental Biology Department of Histology and Embryology College of Basic Medical Sciences Army Medical University (Third Military Medical University) Chongqing 400038 China.
  • 4 Department of Cardiology Daping Hospital Army Medical University (Third Military Medical University) Chongqing 400042 China.
Abstract

Chronic kidney disease (CKD) is associated with accelerated atherosclerosis progression and high incidence of cardiovascular events, hinting that atherosclerotic plaques in CKD may be vulnerable. However, its cause and mechanism remain obscure. Here, it is shown that apolipoprotein E-deficient (ApoE-/-) mouse with CKD (CKD/ApoE-/- mouse) is a useful model for investigating the pathogenesis of plaque vulnerability, and premature senescence and phenotypic switching of vascular smooth muscle cells (VSMCs) contributes to CKD-associated plaque vulnerability. Subsequently, VSMC phenotypes in patients with CKD and CKD/ApoE-/- mice are comprehensively investigated. Using multi-omics analysis and targeted and VSMC-specific gene knockout mice, VSMCs are identified as both type-I-interferon (IFN-I)-responsive and IFN-I-productive cells. Mechanistically, mitochondrial damage resulting from CKD-induced oxidative stress primes the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway to trigger IFN-I response in VSMCs. Enhanced IFN-I response then induces VSMC premature senescence and phenotypic switching in an autocrine/paracrine manner, resulting in the loss of fibrous cap VSMCs and fibrous cap thinning. Conversely, blocking IFN-I response remarkably attenuates CKD-associated plaque vulnerability. These findings reveal that IFN-I response in VSMCs through immune sensing of mitochondrial damage is essential for the pathogenesis of CKD-associated plaque vulnerability. Mitigating IFN-I response may hold promise for the treatment of CKD-associated cardiovascular diseases.

Keywords

atherosclerosis; chronic kidney disease; cyclic GMP‐AMP synthase‐stimulator of interferon genes pathway; plaque vulnerability; vascular smooth muscle cell.

Figures
Products