scispace - formally typeset
Open AccessJournal ArticleDOI

Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function

Reads0
Chats0
TLDR
It is indicated that augmenting glycolytic flux drives CD8+ T cells toward a terminally differentiated state, while its inhibition preserves the formation of long-lived memory CD8+, and the efficacy of T cell-based therapies against chronic infectious diseases and cancer.
Abstract
Naive CD8+ T cells rely upon oxidation of fatty acids as a primary source of energy. After antigen encounter, T cells shift to a glycolytic metabolism to sustain effector function. It is unclear, however, whether changes in glucose metabolism ultimately influence the ability of activated T cells to become long-lived memory cells. We used a fluorescent glucose analog, 2-NBDG, to quantify glucose uptake in activated CD8+ T cells. We found that cells exhibiting limited glucose incorporation had a molecular profile characteristic of memory precursor cells and an increased capacity to enter the memory pool compared with cells taking up high amounts of glucose. Accordingly, enforcing glycolytic metabolism by overexpressing the glycolytic enzyme phosphoglycerate mutase-1 severely impaired the ability of CD8+ T cells to form long-term memory. Conversely, activation of CD8+ T cells in the presence of an inhibitor of glycolysis, 2-deoxyglucose, enhanced the generation of memory cells and antitumor functionality. Our data indicate that augmenting glycolytic flux drives CD8+ T cells toward a terminally differentiated state, while its inhibition preserves the formation of long-lived memory CD8+ T cells. These results have important implications for improving the efficacy of T cell–based therapies against chronic infectious diseases and cancer.

read more

Content maybe subject to copyright    Report

Citations
More filters
Journal ArticleDOI

Metabolic Modulation of Immunity: A New Concept in Cancer Immunotherapy

TL;DR: The metabolic features of the diverse immune cell types in the context of the TME are described and how these immunomodulatory strategies could synergize with immunotherapy to circumvent its current limitations are discussed.
Journal ArticleDOI

Engineering Chimeric Antigen Receptor T-Cells for Racing in Solid Tumors: Don't Forget the Fuel.

TL;DR: CAR T-cell expansion protocols that promote a less differentiated phenotype, combined with optimal receptor design and coengineering strategies, along with immunomodulatory therapies that also promote endogenous immunity, offer great promise in surmounting immunometabolic barriers in the tumor microenvironment (TME) and curing solid tumors.
Journal ArticleDOI

Metabolic reprograming of anti-tumor immunity.

TL;DR: The key metabolic properties of anti-cancer T cells are discussed, along with potential strategies to enhance immunotherapy through the modulation of T cell metabolism.
Journal ArticleDOI

Metabolic exhaustion in infection, cancer and autoimmunity.

TL;DR: It is shown that early metabolic changes are responsible for the later emergence of exhaustion, do not simply reflect changes secondary to chronic activation and are modifiable.
Journal ArticleDOI

Metabolic regulation of immune responses: therapeutic opportunities.

TL;DR: This Review will outline the metabolic configurations adopted by different immune cell subsets, describe the emerging roles for metabolic enzymes and metabolites in the control of immune cell function, and discuss the therapeutic implications of this emerging immune regulatory axis.
References
More filters
Journal ArticleDOI

Lineage relationship and protective immunity of memory CD8 T cell subsets.

TL;DR: It is proposed that TCM and TEM do not necessarily represent distinct subsets, but are part of a continuum in a linear naive → effector → TEM → TCM differentiation pathway.
Journal ArticleDOI

The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation

TL;DR: Metabolic tracer analysis revealed a Myc-dependent metabolic pathway linking glutaminolysis to the biosynthesis of polyamines, which may represent a general mechanism for metabolic reprogramming under patho-physiological conditions.
Journal ArticleDOI

Cutting Edge: Distinct Glycolytic and Lipid Oxidative Metabolic Programs Are Essential for Effector and Regulatory CD4+ T Cell Subsets

TL;DR: Teff and Treg were selectively increased in Glut1 transgenic mice and reliant on glucose metabolism, whereas Treg had activated AMP-activated protein kinase and were dependent on lipid oxidation.
Related Papers (5)