4.8 Article

Phosphorylation of human TRM9L integrates multiple stress-signaling pathways for tumor growth suppression

期刊

SCIENCE ADVANCES
卷 4, 期 7, 页码 -

出版社

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/sciadv.aas9184

关键词

-

资金

  1. David H. Koch Cancer Fellowship
  2. Howard Hughes Medical Institute International Student Research Fellowship
  3. Siebel Scholarship
  4. NSF CAREER Award [1552126]
  5. National Institute of Environmental Health Sciences [E5026856, E5024615]
  6. National Cancer Institute [CA026731]
  7. NATIONAL CANCER INSTITUTE [P01CA026731] Funding Source: NIH RePORTER
  8. NATIONAL INSTITUTE OF DIABETES AND DIGESTIVE AND KIDNEY DISEASES [K08DK090122] Funding Source: NIH RePORTER
  9. NATIONAL INSTITUTE OF ENVIRONMENTAL HEALTH SCIENCES [P30ES002109, R01ES024615, R01ES026856] Funding Source: NIH RePORTER
  10. OFFICE OF THE DIRECTOR, NATIONAL INSTITUTES OF HEALTH [S10OD025242, S10OD021486] Funding Source: NIH RePORTER
  11. Direct For Biological Sciences [1552126] Funding Source: National Science Foundation

向作者/读者索取更多资源

The human transfer RNA methyltransferase 9-like gene (TRM9L, also known as KIAA1456) encodes a negative regulator of tumor growth that is frequently silenced in many forms of cancer. While TRM9L can inhibit tumor cell growth in vivo, the molecular mechanisms underlying the tumor inhibition activity of TRM9L are unknown. We show that oxidative stress induces the rapid and dose-dependent phosphorylation of TRM9L within an intrinsically disordered domain that is necessary for tumor growth suppression. Multiple serine residues are hyperphosphorylated in response to oxidative stress. Using a chemical genetic approach, we identified a key serine residue in TRM9L that undergoes hyperphosphorylation downstream of the oxidative stress-activated MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase)-RSK (ribosomal protein S6 kinase) signaling cascade. Moreover, we found that phosphorylated TRM9L interacts with the 14-3-3 family of proteins, providing a link between oxidative stress and downstream cellular events involved in cell cycle control and proliferation. Mutation of the serine residues required for TRM9L hyperphosphorylation and 14-3-3 binding abolished the tumor inhibition activity of TRM9L. Our results uncover TRM9L as a key downstream effector of the ERK signaling pathway and elucidate a phospho-signaling regulatory mechanism underlying the tumor inhibition activity of TRM9L.

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