Frontiers in Immunology (Oct 2022)

ATF6α contributes to rheumatoid arthritis by inducing inflammatory cytokine production and apoptosis resistance

  • Luna Ge,
  • Luna Ge,
  • Ting Wang,
  • Dandan Shi,
  • Yun Geng,
  • Huancai Fan,
  • Ruojia Zhang,
  • Yuang Zhang,
  • Yuang Zhang,
  • Jianli Zhao,
  • Shufeng Li,
  • Yi Li,
  • Haojun Shi,
  • Guanhua Song,
  • Jihong Pan,
  • Jihong Pan,
  • Lin Wang,
  • Lin Wang,
  • Jinxiang Han,
  • Jinxiang Han

DOI
https://doi.org/10.3389/fimmu.2022.965708
Journal volume & issue
Vol. 13

Abstract

Read online

ObjectiveThe contribution of activating transcription factor 6α (ATF6α) in rheumatoid arthritis (RA) pathogenesis, especially on fibroblast-like synoviocytes (FLSs), has been suggested by its sensitivity to inflammatory stimulus. However, the exact role and therapeutic potential of ATF6α in RA remains to be fully elucidated.MethodsATF6α expression was determined in joint tissues and FLS, and gain-of-function and loss-of-function analyses were applied to evaluate the biological roles of ATF6α in RA FLSs. A murine collagen-induced arthritis (CIA) model, combining both gene deletion of ATF6α and treatment with the ATF6α inhibitor Ceapin-A7, was employed. Joint inflammation, tissue destruction, circulating levels of inflammatory cytokines were assessed in CIA mice. Transcriptome sequencing analysis (RNASeq), molecular biology, and biochemical approaches were performed to identify target genes of ATF6α.ResultsATF6α expression was significantly increased in synovium of RA patients and in synovium of mice subjected to CIA. ATF6α silencing or inhibition repressed RA FLSs viability and cytokine production but induced the apoptosis. CIA-model mice with ATF6α deficiency displayed decreased arthritic progression, leading to profound reductions in clinical and proinflammatory markers in the joints. Pharmacological treatment of mice with Ceapin-A7 reduced arthritis severity in CIA models. RNA-sequencing of wild-type and knockdown of ATF6α in RA FLSs revealed a transcriptional program that promotes inflammation and suppresses apoptosis, and subsequent experiments identified Baculoviral IAP Repeat Containing 3 (BIRC3) as the direct target for ATF6α.ConclusionThis study highlights the pathogenic role of ATF6α-BIRC3 axis in RA and identifies a novel pathway for new therapies against RA.

Keywords