4.8 Article

Translation-dependent unwinding of stem-loops by UPF1 licenses Regnase-1 to degrade inflammatory mRNAs

期刊

NUCLEIC ACIDS RESEARCH
卷 47, 期 16, 页码 8838-8859

出版社

OXFORD UNIV PRESS
DOI: 10.1093/nar/gkz628

关键词

-

资金

  1. Japan Society for the Promotion of Science (JSPS) KAKENHI [18H05278]
  2. AMED-CREST from Japan Agency for Medical Research and Development
  3. AMED-FORCE from Japan Agency for Medical Research and Development
  4. JSPS throughCore-to-Core Program
  5. JSPS KAKENHI [16K08832, 19H03488, 15KT0084]
  6. Takeda Science Foundation
  7. Uehara Memorial Foundation
  8. Shimizu Foundation for Immunology and Neuroscience
  9. Naito Foundation
  10. Senri Life Science Foundation
  11. Nakajima Foundation
  12. Mochida Memorial Foundation for Medical and Pharmaceutical Research
  13. Project for Cancer Research and Therapeutic Evolution (P-CREATE), AMED
  14. Cooperative Research Program of Institute for Protein Research, Osaka University [CR-17-01]
  15. RIKEN Epigenome and Single Cell Project Grants
  16. Nagase Science Technology Foundation
  17. Astellas Foundation for Research on Metabolic Disorders
  18. Daiichi Sankyo Foundation of Life Science
  19. JSPS [18H05278]
  20. [221S0002]
  21. [16H06279]
  22. Grants-in-Aid for Scientific Research [15KT0084, 18H05278, 16K08832, 19H03488] Funding Source: KAKEN

向作者/读者索取更多资源

Regnase-1-mediated mRNA decay (RMD), in which inflammatory mRNAs harboring specific stem-loop structures are degraded, is a critical part of proper immune homeostasis. Prior to initial translation, Regnase-1 associates with target stem-loops but does not carry out endoribonucleolytic cleavage. Single molecule imaging revealed that UPF1 is required to first unwind the stem-loops, thus licensing Regnase-1 to proceed with RNA degradation. Following translation, Regnase-1 physically associates with UPF1 using two distinct points of interaction: The Regnase-1 RNase domain binds to SMG1-phosphorylated residue T28 in UPF1; in addition, an intrinsically disordered segment in Regnase-1 binds to the UPF1 RecA domain, enhancing the helicase activity of UPF1. The SMG1-UPF1-Regnase-1 axis targets pioneer rounds of translation and is critical for rapid resolution of inflammation through restriction of the number of proteins translated by a given mRNA. Furthermore, small-molecule inhibition of SMG1 prevents RNA unwinding in dendritic cells, allowing post-transcriptional control of innate immune responses.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.8
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据