4.5 Article

Effect of JAK Inhibition on the Induction of Proinflammatory HLA-DR+CD90+Rheumatoid Arthritis Synovial Fibroblasts by Interferon-γ

Journal

ARTHRITIS & RHEUMATOLOGY
Volume 74, Issue 3, Pages 441-452

Publisher

WILEY
DOI: 10.1002/art.41958

Keywords

-

Categories

Funding

  1. medical faculty of the University of Heidelberg
  2. Eva Luise and Horst Kohler Foundation
  3. German Society for Rheumatology
  4. Roche Pharma AG
  5. Third Affiliated Hospital of Nantong University
  6. National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH [K08-AR-072791]
  7. EQUIP-Funding for Medical Scientists, Faculty of Medicine, University of Freiburg
  8. Deutsche Forschungsgemeinschaft [DFG FOR2830 HE 2526/9-1]
  9. NaFoUniMedCovid19 [FKZ: 01KX2021-COVIM]
  10. Joint Biology Consortium - National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH [P30-AR-070253]

Ask authors/readers for more resources

Expanded HLA-DR+CD90+ synovial fibroblasts in patients with rheumatoid arthritis (RA) play a key role in inflammation. Activated NK cells induced the expression of HLA-DR on CD90+ synovial fibroblasts through IFNγ, leading to an inflammatory phenotype. JAK inhibition can target these proinflammatory synovial fibroblasts.
Objective Findings from recent transcriptome analyses of the synovium of patients with rheumatoid arthritis (RA) have revealed that 15-fold expanded HLA-DR+CD90+ synovial fibroblasts potentially act as key mediators of inflammation. The reasons for the expansion of HLA-DR+CD90+ synovial fibroblasts are unclear, but genetic signatures indicate that interferon-gamma (IFN gamma) plays a central role in the generation of this fibroblast subset. The present study was undertaken to investigate the generation, function and therapeutically intended blockage of HLA-DR+CD90+ synovial fibroblasts. Methods We combined functional assays using primary human materials and focused bioinformatic analyses of mass cytometry and transcriptomics patient data sets. Results We detected enriched and activated Fc gamma receptor type IIIa-positive (CD16+) NK cells in the synovial tissue from patients with active RA. Soluble immune complexes were recognized by CD16 in a newly described reporter cell model, a mechanism that could be contributing to the activation of natural killer (NK) cells in RA. In vitro, NK cell-derived IFN gamma induced HLA-DR on CD90+ synovial fibroblasts, leading to an inflammatory, cytokine-secreting HLA-DR+CD90+ phenotype. HLA-DR+CD90+ synovial fibroblasts consecutively activated CD4+ T cells upon receptor crosslinking via superantigens. HLA-DR+CD90+ synovial fibroblasts also activated CD4+ T cells in the absence of superantigens, an effect that was initiated by NK cell-derived IFN gamma and that was 4 times stronger in patients with RA compared to patients with osteoarthritis. Finally, JAK inhibition in synovial fibroblasts prevented HLA-DR induction and blocked proinflammatory signals to T cells. Conclusion The HLA-DR+CD90+ phenotype represents an activation state of synovial fibroblasts during the process of inflammation in RA that can be induced by IFN gamma, likely generated from infiltrating leukocytes such as activated NK cells. The induction of these proinflammatory, interleukin-6-producing, and likely antigen-presenting synovial fibroblasts can be targeted by JAK inhibition.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.5
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available