Mst1 shuts off cytosolic antiviral defense through IRF3 phosphorylation

作者:Meng, Fansen; Zhou, Ruyuan; Wu, Shiying; Zhang, Qian; Jin, Qiuheng; Zhou, Yao; Plouffe, Steven W.; Liu, Shengduo; Song, Hai; Xia, Zongping; Zhao, Bin; Ye, Sheng; Feng, Xin-Hua; Guan, Kun-Liang; Zou, Jian; Xu, Pinglong*
来源:Genes & Development, 2016, 30(9): 1086-1100.
DOI:10.1101/gad.277533.116

摘要

Cytosolic RNA/DNA sensing elicits primary defense against viral pathogens. Interferon regulatory factor 3 (IRF3), a key signal mediator/transcriptional factor of the antiviral-sensing pathway, is indispensible for interferon production and antiviral defense. However, how the status of IRF3 activation is controlled remains elusive. Through a functional screen of the human kinome, we found that mammalian sterile 20-like kinase 1 (Mst1), but not Mst2, profoundly inhibited cytosolic nucleic acid sensing. Mst1 associated with IRF3 and directly phosphorylated IRF3 at Thr75 and Thr253. This Mst1-mediated phosphorylation abolished activated IRF3 homodimerization, its occupancy on chromatin, and subsequent IRF3-mediated transcriptional responses. In addition, Mst1 also impeded virus-induced activation of TANK-binding kinase 1 (TBK1), further attenuating IRF3 activation. As a result, Mst1 depletion or ablation enabled an enhanced antiviral response and defense in cells and mice. Therefore, the identification of Mst1 as a novel physiological negative regulator of IRF3 activation provides mechanistic insights into innate antiviral defense and potential antiviral prevention strategies.