2021
DOI: 10.3389/fimmu.2021.669456
|View full text |Cite
|
Sign up to set email alerts
|

Glycolysis Inhibition Induces Functional and Metabolic Exhaustion of CD4+ T Cells in Type 1 Diabetes

Abstract: In Type 1 Diabetes (T1D), CD4+ T cells initiate autoimmune attack of pancreatic islet β cells. Importantly, bioenergetic programs dictate T cell function, with specific pathways required for progression through the T cell lifecycle. During activation, CD4+ T cells undergo metabolic reprogramming to the less efficient aerobic glycolysis, similarly to highly proliferative cancer cells. In an effort to limit tumor growth in cancer, use of glycolytic inhibitors have been successfully employed in preclinical and cl… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

0
22
0

Year Published

2021
2021
2024
2024

Publication Types

Select...
8
1

Relationship

0
9

Authors

Journals

citations
Cited by 39 publications
(22 citation statements)
references
References 60 publications
0
22
0
Order By: Relevance
“…Enhanced T cell metabolism was also detected in NOD mice and when prediabetic NOD mice were treated with 2-deoxyglucose to block aerobic glycolysis, islet antigen-specific T cell frequency and islet infiltrating lymphocytes decreased, beta-cell degranulation was improved ( 21 ). Similarly, in the adoptive transfer model of T1D, a competitive inhibitor of the glycolytic rate-limiting enzyme of CD4 + T cells reduced T cell responses to β cell antigen in vitro , thus reducing the immunopathological parameters associated with disease onset as 57% of animals remaining euglycemic at the end of the study period ( 22 ). Other studies also found that there are some potential immunotherapy drugs for T1D by inhibiting the T cell glucose metabolism pathway ( 51 53 ).…”
Section: Discussionmentioning
confidence: 99%
“…Enhanced T cell metabolism was also detected in NOD mice and when prediabetic NOD mice were treated with 2-deoxyglucose to block aerobic glycolysis, islet antigen-specific T cell frequency and islet infiltrating lymphocytes decreased, beta-cell degranulation was improved ( 21 ). Similarly, in the adoptive transfer model of T1D, a competitive inhibitor of the glycolytic rate-limiting enzyme of CD4 + T cells reduced T cell responses to β cell antigen in vitro , thus reducing the immunopathological parameters associated with disease onset as 57% of animals remaining euglycemic at the end of the study period ( 22 ). Other studies also found that there are some potential immunotherapy drugs for T1D by inhibiting the T cell glucose metabolism pathway ( 51 53 ).…”
Section: Discussionmentioning
confidence: 99%
“…The change in metabolic features of T-cells during chronic infections has been explored in recent years ( Bengsch et al, 2016 ). The consensus among researchers is that metabolism can affect T-cell development ( Sun et al, 2021 ), proliferation ( Sabharwal et al, 2018 ; Fultang et al, 2020 ), effector function ( Barili et al, 2021 ), and exhaustion ( Martins et al, 2021 ). Moreover, as the energy centers of cell activities, mitochondria are also critical for maintaining T-cell function ( Vardhana et al, 2020 ; Yu et al, 2020 ).…”
Section: Discussionmentioning
confidence: 99%
“…In contrast, activation of mTOR signaling facilitates glycolysis, fatty acid production, and mitochondrial biogenesis (42,45,46). As mentioned above, autoreactive CD4+/CD8+ T cells exhibit a higher level of glycolysis and depend less on OXPHOS, thus suggesting that glycolysis could be used as an attractive therapeutic target (47). Inhibiting mTOR signaling with rapamycin or enhancing the AMPK signaling pathway with metformin are known to reduce glycolysis (19).…”
Section: Metabolic Interventions: a New Opportunity For T1d Treatmentmentioning
confidence: 99%
“…PFK15, a competitive inhibitor of the rate-limiting glycolysis enzyme, has been found to suppress glycolysis utilization of CD4+ T cells and decrease the response of CD4+ T cells to b-cell antigens. Additionally, treatment of PFK15 in NOD mouse models delayed T1D onset due to metabolic and functional exhaustion of T cells (47). In addition, peroxisome proliferator-activated receptors (PPARs) are transcription factors that control genes involved in glucose and lipid metabolism and FAO (90)(91)(92).…”
Section: Metabolic Interventions: a New Opportunity For T1d Treatmentmentioning
confidence: 99%