Acetyl-CoA Carboxylase Obstructs CD8+ T-Cell Lipid Utilization and Energy Synthesis in the Tumor Microenvironment

bioRxiv (Cold Spring Harbor Laboratory)(2023)

Cited 0|Views6
No score
Abstract
The solid tumor microenvironment (TME) imprints a compromised metabolic state in tumor infiltrating T cells (TILs) hallmarked by the inability to maintain effective energy synthesis for antitumor function and survival. T cells in the TME must catabolize lipids via mitochondrial fatty acid oxidation (FAO) to supply energy in nutrient stress, and it is established that T cells enriched in FAO are adept at cancer control. However, endogenous TILs and unmodified cellular therapy products fail to sustain bioenergetics in tumors. Using patient samples and mouse models, we reveal that the solid TME imposes perpetual acetyl-CoA carboxylase (ACC) activity, enforcing lipid biogenesis and storage in TILs that directly opposes FAO. Using metabolic, lipidomic, and confocal imaging strategies, we find that restricting ACC wholly rewires T cell metabolism, enabling energy maintenance in TME stress. Moreover, limiting ACC activity potentiates a gene and phenotypic program indicative of T cell memory, engendering TILs with increased survival and polyfunctionality, with the ability to control solid cancer. ### Competing Interest Statement The authors have declared no competing interest.
More
Translated text
Key words
Tumor Microenvironment,Cancer Cell Metabolism,Cancer Metabolism,Tumor Targeting,Metabolism
AI Read Science
Must-Reading Tree
Example
Generate MRT to find the research sequence of this paper
Chat Paper
Summary is being generated by the instructions you defined