The role of FOXO4/NFAT2 signaling pathway in dysfunction of human coronary endothelial cells and inflammatory infiltration of vasculitis in Kawasaki disease

Front Immunol. 2023 Jan 9:13:1090056. doi: 10.3389/fimmu.2022.1090056. eCollection 2022.

Abstract

Aims: The Ca+/NFAT (Nuclear factor of activated T cells) signaling pathway activation is implicated in the pathogenesis of Kawasaki disease (KD); however, we lack detailed information regarding the regulatory network involved in the human coronary endothelial cell dysfunction and cardiovascular lesion development. Herein, we aimed to use mouse and endothelial cell models of KD vasculitis in vivo and in vitro to characterize the regulatory network of NFAT pathway in KD.

Methods and results: Among the NFAT gene family, NFAT2 showed the strongest transcriptional activity in peripheral blood mononuclear cells (PBMCs) from patients with KD. Then, NFAT2 overexpression and knockdown experiments in Human coronary artery endothelial cells (HCAECs) indicated that NFAT2 overexpression disrupted endothelial cell homeostasis by regulation of adherens junctions, whereas its knockdown protected HCAECs from such dysfunction. Combined analysis using RNA-sequencing and transcription factor (TF) binding site analysis in the NFAT2 promoter region predicted regulation by Forkhead box O4 (FOXO4). Western blotting, chromatin immunoprecipitation, and luciferase assays validated that FOXO4 binds to the promoter and transcriptionally represses NFAT2. Moreover, Foxo4 knockout increased the extent of inflamed vascular tissues in a mouse model of KD vasculitis. Functional experiments showed that inhibition NFAT2 relieved Foxo4 knockout exaggerated vasculitis in vivo.

Conclusions: Our findings revealed the FOXO4/NFAT2 axis as a vital pathway in the progression of KD that is associated with endothelial cell homeostasis and cardiovascular inflammation development.

Keywords: Ca+/NFAT pathway; FOXO4; Kawasaki disease; transcription factor; vasculitis.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Animals
  • Cell Cycle Proteins / metabolism
  • Endothelial Cells / metabolism
  • Forkhead Transcription Factors* / genetics
  • Forkhead Transcription Factors* / metabolism
  • Humans
  • Leukocytes, Mononuclear / metabolism
  • Mice
  • Mucocutaneous Lymph Node Syndrome* / pathology
  • NFATC Transcription Factors* / genetics
  • NFATC Transcription Factors* / metabolism
  • Signal Transduction

Substances

  • Cell Cycle Proteins
  • Forkhead Transcription Factors
  • FOXO4 protein, human
  • FoxO4 protein, mouse
  • NFATC Transcription Factors
  • NFATC1 protein, human

Grants and funding

This work was supported by the National Natural Science Foundation of China [grant numbers 81870365, 81970436, 82270512, and 82070529] and the Project of Education and Scientific Research for Young and Middle aged Teachers in Fujian Province, China [grant number JAT200124] HH received support from the China Scholarship Council for 1 year study at the Max-Planck Institute for Heart and Lung Research. LG was supported by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation), EXC 2026, Cardio-Pulmonary Institute (CPI), Project ID 390649896..