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

T Cell Fitness and Autologous CAR T Cell Therapy in Haematologic Malignancy

TL;DR: The fitness of an individual's T cells, driven by age, chronic infection, disease burden and cancer treatment, is a crucial limiting factor of Chimeric antigen receptor (CAR) T cell therapy.
Journal ArticleDOI

Metabolic Reprogramming Induces Immune Cell Dysfunction in the Tumor Microenvironment of Multiple Myeloma.

TL;DR: In this paper, metabolic changes including generation, depletion or accumulation of metabolites and signaling pathways, as well as their relationship with the TME in multiple myeloma (MM) cells are discussed.
Journal ArticleDOI

Metabolic Targets for Improvement of Allogeneic Hematopoietic Stem Cell Transplantation and Graft-vs.-Host Disease

TL;DR: The importance of metabolism on the function of GVHD-inducing and GVL-inducing alloreactive T cells as well as on antigen presenting cells (APC), which are required for presentation of host antigens are highlighted.
Journal ArticleDOI

Pharmacological Targeting of GLUT1 to Control Autoreactive T Cell Responses.

TL;DR: This paper will review recent literature to determine whether it is possible to design a pharmacological Glut1 blocking strategy and how to apply this to autoimmunity in T1D.
Journal ArticleDOI

Filling the Tank: Keeping Antitumor T Cells Metabolically Fit for the Long Haul.

TL;DR: The general features of the TME are reviewed, the metabolic demands of activated effector T cells are identified, the known metabolic checkpoints associated with intratumoral T cellsare discussed, and strategies for generating superior antitumor T Cells are proposed.
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)