Transcription factor NFAT5 promotes migration and invasion of rheumatoid synoviocytes via coagulation factor III and CCL2

S Lee, JS Kong, S You, HM Kwon, SA Yoo… - The Journal of …, 2018 - journals.aai.org
S Lee, JS Kong, S You, HM Kwon, SA Yoo, CS Cho, WU Kim
The Journal of Immunology, 2018journals.aai.org
Fibroblast-like synoviocytes (FLSs) play a key role in the progression of rheumatoid arthritis
(RA) as a primary component of invasive hypertrophied pannus. FLSs of RA patients (RA-
FLSs) exhibit cancer-like features, including promigratory and proinvasive activities that
largely contribute to joint cartilage and bone destruction. In this study, we hypothesized that
the NF of activated T cell 5 (NFAT5), a transcription factor involving tumor invasiveness,
would control the migration and invasion of RA-FLSs. Analyses of transcriptomes …
Abstract
Fibroblast-like synoviocytes (FLSs) play a key role in the progression of rheumatoid arthritis (RA) as a primary component of invasive hypertrophied pannus. FLSs of RA patients (RA-FLSs) exhibit cancer-like features, including promigratory and proinvasive activities that largely contribute to joint cartilage and bone destruction. In this study, we hypothesized that the NF of activated T cell 5 (NFAT5), a transcription factor involving tumor invasiveness, would control the migration and invasion of RA-FLSs. Analyses of transcriptomes demonstrated the significant involvement of NFAT5 in locomotion of RA-FLSs and that tissue factor (TF; also known as coagulation factor III) and CCL2 were the major downstream target genes of NFAT5 involving FLS migration and invasion. In cultured RA-FLSs, IL-1β and TGF-β increased TF and CCL2 expression by upregulating NFAT5 expression via p38 MAPK. Functional assays demonstrated that NFAT5-or TF-deficient RA-FLSs displayed decreased lamellipodia formation, cell migration, and invasion under IL-1β–or TGF-β–stimulated conditions. Conversely, factor VIIa, a specific activator of TF, increased migration of RA-FLSs, which was blocked by NFAT5 knockdown. Recombinant CCL2 partially restored the decrease in migration and invasion of NFAT5-deficient RA-FLSs stimulated with IL-1β. NFAT5-knockout mouse FLSs also showed decreased expressions of TF and CCL2 and reduced cell migration. Moreover, KRN2, a specific inhibitor of NFAT5, suppressed migration of FLSs stimulated with TGF-β. Conclusively, to our knowledge, this is the first study to provide evidence of a functional link between osmoprotective NFAT5 and TF in the migration and invasion of RA-FLSs and supports a role for NFAT5 blockade in the treatment of RA.
journals.aai.org