期刊论文详细信息
eLife
m6A modifications regulate intestinal immunity and rotavirus infection
Wanyiin Tao1  Siyuan Ding1  Huabing Li1  Xingxing Ren1  Shu Zhu1  Juanzi Gao1  Anmin Wang2  Chen Qian2  Gaopeng Hou2  Kaiguang Zhang2  Richard A Flavell2  Wen Pan2  Decai Wang2  Runzhi Li2  Jiyu Tong2  Guorong Zhang3  Jinghao Wang4 
[1] Institute of Immunology, the Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China;Department of Digestive Disease, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China;Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, United States;Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, China;
关键词: m6A;    virus infection;    innate immune;    rotavirus;    IRF7;   
DOI  :  10.7554/eLife.73628
来源: DOAJ
【 摘 要 】

N6-methyladenosine (m6A) is an abundant mRNA modification and affects many biological processes. However, how m6A levels are regulated during physiological or pathological processes such as virus infections, and the in vivo function of m6A in the intestinal immune defense against virus infections are largely unknown. Here, we uncover a novel antiviral function of m6A modification during rotavirus (RV) infection in small bowel intestinal epithelial cells (IECs). We found that rotavirus infection induced global m6A modifications on mRNA transcripts by down-regulating the m6a eraser ALKBH5. Mice lacking the m6A writer enzymes METTL3 in IECs (Mettl3ΔIEC) were resistant to RV infection and showed increased expression of interferons (IFNs) and IFN-stimulated genes (ISGs). Using RNA-sequencing and m6A RNA immuno-precipitation (RIP)-sequencing, we identified IRF7, a master regulator of IFN responses, as one of the primary m6A targets during virus infection. In the absence of METTL3, IECs showed increased Irf7 mRNA stability and enhanced type I and III IFN expression. Deficiency in IRF7 attenuated the elevated expression of IFNs and ISGs and restored susceptibility to RV infection in Mettl3ΔIEC mice. Moreover, the global m6A modification on mRNA transcripts declined with age in mice, with a significant drop from 2 weeks to 3 weeks post birth, which likely has broad implications for the development of intestinal immune system against enteric viruses early in life. Collectively, we demonstrated a novel host m6A-IRF7-IFN antiviral signaling cascade that restricts rotavirus infection in vivo.

【 授权许可】

Unknown   

  文献评价指标  
  下载次数:0次 浏览次数:0次