Citation

4724 total record number 96 records this year

SRC1 promotes Th17 differentiation by overriding Foxp3 suppression to stimulate RORt activity in a PKC–dependent manner

Sen, S;Wang, F;Zhang, J;He, Z;Ma, J;Gwack, Y;Xu, J;Sun, Z;

Th17 cells are major players in multiple autoimmune diseases and are developmentally contingent on reciprocal functionality between the transcription factor Retineic acid receptor-related orphan nuclear receptor gamma (RORt) and Forkhead box protein P3 (Foxp3). Here we deciphered a previously unappreciated role of Steroid receptor coactivator 1 (SRC1) in defining the lineage decision for the development of Th17 versus induced T-regulatory (iTreg) cells. We demonstrate that SRC1 functions as a critical coactivator for RORt in vivo to promote the functional dominance of RORt over Foxp3 and thus establishing an unopposed Th17 differentiation program. In the absence of SRC1, T cell polarization resulted in decreased IL-17+ and increased Foxp3+ cells during both in vitro differentiation and in vivo development of experimental autoimmune encephalomyelitis. Mechanistically, T cell receptor (TCR) signaling molecule protein kinase C theta (PKC-)-mediated phosphorylation of SRC1 is important for inducing enhanced RORt-SRC1 interaction, stable DNA binding, and resultant IL-17A transcription. Furthermore, phospho-SRC1-mediated recruitment of CARM1 induced prominent asymmetric dimethylation of H3R17 while preventing repressive H3K9 trimethylation and hence further modifying the IL-17 locus for optimal transcription. Moreover, binding of phospho-SRC1 to RORt displaced bound Foxp3, leading to prompt degradation of the dissociated Foxp3 via a ubiquitin-proteosomal pathway and hence reversing the inhibitory action of Foxp3 on RORt activity. Thus, SRC1 acts as a crucial molecular mediator to integrate positive PKC–dependent TCR signals to induce peak RORt activity and establish phenotypic dominance of Th17 over the iTreg pathway.