Abstract

N-acetylglucosamine (GlcNAc) branching of Asn (N)–linked glycans inhibits pro-inflammatory T cell responses and models of autoimmune diseases such as Multiple Sclerosis (MS). Metabolism controls N-glycan branching in T cells by regulating de novo hexosamine pathway biosynthesis of UDP-GlcNAc, the donor substrate for the Golgi branching enzymes. Activated T cells switch metabolism from oxidative phosphorylation to aerobic glycolysis and glutaminolysis. This reduces flux of glucose and glutamine into the hexosamine pathway, thereby inhibiting de novo UDP-GlcNAc synthesis and N-glycan branching. Salvage of GlcNAc into the hexosamine pathway overcomes this metabolic suppression to restore UDP-GlcNAc synthesis and N-glycan branching, thereby promoting anti-inflammatory T regulatory (Treg) over pro-inflammatory T helper (TH) 17 and TH1 differentiation to suppress autoimmunity. However, GlcNAc activity is limited by the lack of a cell surface transporter and requires high doses to enter cells via macropinocytosis. Here we report that GlcNAc-6-acetate is a superior pro-drug form of GlcNAc. Acetylation of amino-sugars improves cell membrane permeability, with subsequent de-acetylation by cytoplasmic esterases allowing salvage into the hexosamine pathway. Per- and bi-acetylation of GlcNAc led to toxicity in T cells, whereas mono-acetylation at only the 6 > 3 position raised N-glycan branching greater than GlcNAc without inducing significant toxicity. GlcNAc-6-acetate inhibited T cell activation/proliferation, TH1/TH17 responses and disease progression in Experimental Autoimmune Encephalomyelitis (EAE), a mouse model of MS. Thus, GlcNAc-6-Acetate may provide an improved therapeutic approach to raise N-glycan branching, inhibit pro-inflammatory T cell responses and treat autoimmune diseases such as MS.

Highlights

  • Cell surface and secreted proteins are co- and post-translationally modified on Asn (N) by the addition of carbohydrates (N-glycans) in the endoplasmic reticulum

  • To increase hydrophobicity and membrane permeability, we examined GlcNAc analogs modified using one or more acetyl substituents at various positions (Fig 1B) and tested their ability to raise N-glycan branching in human CD4+ T cells

  • Cells were activated with PMA/ionomycin or plate-bound anti-CD3ε plus soluble anti-CD28 and assessed by flow cytometry with Phaseolus vulgaris leukoagglutinin (L-PHA), a plant lectin that binds β1,6GlcNAc-branched N-glycans produced by the Mgat5 enzyme (Fig 1A) and serves as an overall marker of N-glycan branching [2,3,9,10,31]

Read more

Summary

Introduction

Cell surface and secreted proteins are co- and post-translationally modified on Asn (N) by the addition of carbohydrates (N-glycans) in the endoplasmic reticulum. Nglycan branching drives Treg differentiation by promoting IL-2Rα (CD25) surface expression and signaling in T cell blasts, with loss of branching virtually eliminating induction of Tregs [10]. Consistent with these phenotypes, reductions in N-glycan branching promote proinflammatory T cell responses and autoimmunity in both mice and humans [3,12,13,14,15,16,17]

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