1. Academic Validation
  2. SARS-CoV-2 nucleocapsid protein triggers hyperinflammation via protein-protein interaction-mediated intracellular Cl- accumulation in respiratory epithelium

SARS-CoV-2 nucleocapsid protein triggers hyperinflammation via protein-protein interaction-mediated intracellular Cl- accumulation in respiratory epithelium

  • Signal Transduct Target Ther. 2022 Jul 27;7(1):255. doi: 10.1038/s41392-022-01048-1.
Lei Chen  # 1 Wei-Jie Guan  # 2 3 4 Zhuo-Er Qiu  # 1 Jian-Bang Xu  # 2 Xu Bai 1 Xiao-Chun Hou 1 Jing Sun 2 Su Qu 1 Ze-Xin Huang 1 Tian-Lun Lei 1 Zi-Yang Huang 1 Jincun Zhao 2 Yun-Xin Zhu 1 Ke-Nan Ye 1 Zhao-Rong Lun 1 Wen-Liang Zhou 5 Nan-Shan Zhong 6 7 Yi-Lin Zhang 8
Affiliations

Affiliations

  • 1 School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
  • 2 State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
  • 3 Department of Thoracic Surgery, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China.
  • 4 Guangzhou Laboratory, Guangzhou, China.
  • 5 School of Life Sciences, Sun Yat-sen University, Guangzhou, China. [email protected].
  • 6 State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China. [email protected].
  • 7 Guangzhou Laboratory, Guangzhou, China. [email protected].
  • 8 School of Life Sciences, Sun Yat-sen University, Guangzhou, China. [email protected].
  • # Contributed equally.
Abstract

SARS-CoV-2, the culprit pathogen of COVID-19, elicits prominent immune responses and cytokine storms. Intracellular Cl- is a crucial regulator of host defense, whereas the role of Cl- signaling pathway in modulating pulmonary inflammation associated with SARS-CoV-2 Infection remains unclear. By using human respiratory epithelial cell lines, primary cultured human airway epithelial cells, and murine models of viral structural protein stimulation and SARS-CoV-2 direct challenge, we demonstrated that SARS-CoV-2 nucleocapsid (N) protein could interact with SMAD3, which downregulated cystic fibrosis transmembrane conductance regulator (CFTR) expression via microRNA-145. The intracellular Cl- concentration ([Cl-]i) was raised, resulting in phosphorylation of serum glucocorticoid regulated kinase 1 (SGK1) and robust inflammatory responses. Inhibition or knockout of SGK1 abrogated the N protein-elicited airway inflammation. Moreover, N protein promoted a sustained elevation of [Cl-]i by depleting intracellular cAMP via upregulation of phosphodiesterase 4 (PDE4). Rolipram, a selective PDE4 Inhibitor, countered airway inflammation by reducing [Cl-]i. Our findings suggested that Cl- acted as the crucial pathological second messenger mediating the inflammatory responses after SARS-CoV-2 Infection. Targeting the Cl- signaling pathway might be a novel therapeutic strategy for COVID-19.

Figures
Products