Abstract

Regulatory T cells (Tregs) are required for the maintenance of immune tolerance and adoptive Treg infusion therapy has become a promising approach to suppress immune responses in diseases such as autoimmunity and transplant rejection. However, one critical challenge of Treg therapy is the requirement of in vitro expansion of functionally stable Tregs while preventing either the contamination of T effector and/or emergence of unstable pathogenic Tregs. Recent studies showing distinct metabolic requirements of T effectors and Tregs suggest that manipulation of cell metabolism may be an attractive strategy to achieve this goal. Here we show that human thymically derived Tregs (tTregs) and in vitro induced Tregs (iTregs) from naive T cells engage glycolysis equivalently upon activation. However, inhibiting glucose metabolism via 2-deoxy-D-glucose (2DG) has distinct effects on each of these subsets. While 2DG treatment at the onset of activation significantly reduced the proliferation and expression of suppressive molecules such as ICOS and CTLA-4 in tTregs, its effect on FOXP3 expression was small. In contrast, 2DG treatment during iTreg induction modestly decreased their proliferation but strongly reduced both ICOS and FOXP3 expression. Importantly, both Treg subsets became insensitive to 2DG after day 3 post activation with little effect on either proliferation or FOXP3 expression while T conventional Th0 cells showed reduced proliferation under the same conditions. Moreover, 2DG treatment at day 3 did not impair the suppressive capabilities of Treg subsets. Collectively, these findings suggest that there is a distinct temporal requirement of glycolysis in each of the activated human Treg subsets and T conventional cells. Furthermore, 2DG treatment at the onset as a strategy to impair contaminating T effector cell proliferation is unfavorable for optimal Treg generation as well.

Highlights

  • Regulatory T cells (Tregs) are critical for the prevention of autoimmunity [1] and for transplantation tolerance [2]

  • One is thymically derived Tregs while the other is comprised of peripherally induced Tregs, that originate from naïve T cells and acquire forkhead box P3 (FOXP3) protein following stimulation [6]

  • In order to understand the glycolytic requirements of human Tregs subsets, here we directly compared the metabolic requirements of pre-existing human ex vivo Tregs and induced Tregs (iTregs) at different stages of in vitro activation. We show that both human Treg subsets engage in glycolysis to a similar degree at day 3 post activation. 2-deoxyD-glucose (2DG) treatment, that inhibits glycolysis and pentose phosphate pathway, glycolytic oxidation and protein glycosylation, at either the onset or day 3 post activation revealed that glycolysis is required for cell growth and proliferation in both subsets, it is predominantly required for optimal expression of inhibitory molecules such as CTLA4 and ICOS in thymically derived Tregs (tTregs) and critical for the FOXP3 induction in iTregs during early activation

Read more

Summary

Introduction

Regulatory T cells (Tregs) are critical for the prevention of autoimmunity [1] and for transplantation tolerance [2] Given their therapeutic potential, clinical trials of Treg immunotherapy are already underway in patients with autoimmune diseases and recipients of stem cell and solid organ transplants[3]. Clinical trials of Treg immunotherapy are already underway in patients with autoimmune diseases and recipients of stem cell and solid organ transplants[3] Despite these advances, it remains challenging to manufacture sufficient numbers of functionally stable Tregs and optimize in vivo conditioning treatments, which highlight the issue of feasibility and safety with current approaches [4, 5]. TTregs are considered by many as the more desirable candidates for Treg expansion protocols

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call