Back to Search Start Over

Glc7/PP1 dephosphorylates histone H3T11 to regulate autophagy and telomere silencing in response to nutrient availability.

Authors :
Zhang, Xinyu
Yu, Qi
Wu, Yinsheng
Zhang, Yuan
He, Yi
Wang, Rongsha
Yu, Xilan
Li, Shanshan
Source :
Cell Discovery; 7/11/2023, Vol. 9 Issue 1, p1-25, 25p
Publication Year :
2023

Abstract

How cells adapt their gene expression to nutritional changes remains poorly understood. Histone H3T11 is phosphorylated by pyruvate kinase to repress gene transcription. Here, we identify the protein phosphatase 1 (PP1), Glc7 as the enzyme that specifically dephosphorylates H3T11. We also characterize two novel Glc7-containing complexes and reveal their roles in regulating gene expression upon glucose starvation. Specifically, the Glc7–Sen1 complex dephosphorylates H3T11 to activate the transcription of autophagy-related genes. The Glc7–Rif1–Rap1 complex dephosphorylates H3T11 to derepress the transcription of telomere-proximal genes. Upon glucose starvation, Glc7 expression is up-regulated and more Glc7 translocates into the nucleus to dephosphorylate H3T11, leading to induction of autophagy and derepressed transcription of telomere-proximal genes. Furthermore, the functions of PP1/Glc7 and the two Glc7-containing complexes are conserved in mammals to regulate autophagy and telomere structure. Collectively, our results reveal a novel mechanism that regulate gene expression and chromatin structure in response to glucose availability. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
20565968
Volume :
9
Issue :
1
Database :
Complementary Index
Journal :
Cell Discovery
Publication Type :
Academic Journal
Accession number :
164817659
Full Text :
https://doi.org/10.1038/s41421-023-00551-1