4.8 Article

Cancer SLC43A2 alters T cell methionine metabolism and histone methylation

Journal

NATURE
Volume 585, Issue 7824, Pages 277-+

Publisher

NATURE PORTFOLIO
DOI: 10.1038/s41586-020-2682-1

Keywords

-

Funding

  1. Advanced Genomics Core and Bioinformatics Core of the University of Michigan Medical School's Biomedical Research Core Facilities
  2. US NIH/NCI [CA217648, CA123088, CA099985, CA193136, CA152470]
  3. NCI Cooperative Human Tissue Network (CHTN)
  4. 2017 AACR NextGen Grant for Transformative Cancer Research [17-20-01-LYSS]
  5. ACS Research Scholar Grant [RSG-18-186-01]
  6. NIH grant [DK097153]
  7. Charles Woodson Research Fund
  8. University of Michigan Pediatric Brain Tumor Initiative
  9. NIH through the University of Michigan Rogel Cancer Center Grant [P30 CA046592]

Ask authors/readers for more resources

Expression of the transporter SLC43A2 by tumour cells allows them to outcompete T cells for methionine and thereby disrupt the survival and function of tumour-infiltrating T cells. Abnormal epigenetic patterns correlate with effector T cell malfunction in tumours(1-4), but the cause of this link is unknown. Here we show that tumour cells disrupt methionine metabolism in CD8(+)T cells, thereby lowering intracellular levels of methionine and the methyl donorS-adenosylmethionine (SAM) and resulting in loss of dimethylation at lysine 79 of histone H3 (H3K79me2). Loss of H3K79me2 led to low expression of STAT5 and impaired T cell immunity. Mechanistically, tumour cells avidly consumed methionine and outcompeted T cells for methionine by expressing high levels of the methionine transporter SLC43A2. Genetic and biochemical inhibition of tumour SLC43A2 restored H3K79me2 in T cells, thereby boosting spontaneous and checkpoint-induced tumour immunity. Moreover, methionine supplementation improved the expression of H3K79me2 and STAT5 in T cells, and this was accompanied by increased T cell immunity in tumour-bearing mice and patients with colon cancer. Clinically, tumour SLC43A2 correlated negatively with T cell histone methylation and functional gene signatures. Our results identify a mechanistic connection between methionine metabolism, histone patterns, and T cell immunity in the tumour microenvironment. Thus, cancer methionine consumption is an immune evasion mechanism, and targeting cancer methionine signalling may provide an immunotherapeutic approach.

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