4.8 Article

O-GlcNAc modification of leucyl-tRNA synthetase 1 integrates leucine and glucose availability to regulate mTORC1 and the metabolic fate of leucine

Journal

NATURE COMMUNICATIONS
Volume 13, Issue 1, Pages -

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41467-022-30696-8

Keywords

-

Funding

  1. National Research Foundation (NRF) of Korea - Ministry of Education [2018R1A6A1A03023718, 2020R1I1A1A01067423]
  2. Bio & Medical Technology Development Program of the National Research Foundation (NRF) - Ministry of Science ICT [2020M3E5E2040282]
  3. National Research Foundation of Korea (NRF) - Ministry of Science ICT (MSIT) [2020R1A2C2099586, 2021R1C1C2006283]
  4. Health Fellowship Foundation
  5. National Research Foundation of Korea [2018R1A6A1A03023718, 2021R1C1C2006283, 2020R1I1A1A01067423, 2020R1A2C2099586, 2020M3E5E2040282] Funding Source: Korea Institute of Science & Technology Information (KISTI), National Science & Technology Information Service (NTIS)

Ask authors/readers for more resources

This study reveals that under glucose starvation, intracellular leucine sensor LARS1 is O-GlcNAcylated, which regulates mTORC1 activity and leucine metabolism, impacting the utilization of glucose and leucine.
All living organisms have the ability to sense nutrient levels to coordinate cellular metabolism. Despite the importance of nutrient-sensing pathways that detect the levels of amino acids and glucose, how the availability of these two types of nutrients is integrated is unclear. Here, we show that glucose availability regulates the central nutrient effector mTORC1 through intracellular leucine sensor leucyl-tRNA synthetase 1 (LARS1). Glucose starvation results in O-GlcNAcylation of LARS1 on residue S1042. This modification inhibits the interaction of LARS1 with RagD GTPase and reduces the affinity of LARS1 for leucine by promoting phosphorylation of its leucine-binding site by the autophagy-activating kinase ULK1, decreasing mTORC1 activity. The lack of LARS1 O-GlcNAcylation constitutively activates mTORC1, supporting its ability to sense leucine, and deregulates protein synthesis and leucine catabolism under glucose starvation. This work demonstrates that LARS1 integrates leucine and glucose availability to regulate mTORC1 and the metabolic fate of leucine. Leucyl-tRNA synthetase 1 (LARS1) is a leucine sensor for mTORC1 signaling and regulates leucine utilization depending on glucose availability. Here, the author show that O-GlcNAcylation of LARS1 is crucial for its ability to regulate mTORC1 activity and leucine metabolism upon glucose starvation.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.8
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available