Circulating gluten-specific FOXP3+CD39+ regulatory T cells have impaired suppressive function in patients with celiac disease

J Allergy Clin Immunol. 2017 Dec;140(6):1592-1603.e8. doi: 10.1016/j.jaci.2017.02.015. Epub 2017 Mar 8.

Abstract

Background: Celiac disease is a chronic immune-mediated inflammatory disorder of the gut triggered by dietary gluten. Although the effector T-cell response in patients with celiac disease has been well characterized, the role of regulatory T (Treg) cells in the loss of tolerance to gluten remains poorly understood.

Objective: We sought to define whether patients with celiac disease have a dysfunction or lack of gluten-specific forkhead box protein 3 (FOXP3)+ Treg cells.

Methods: Treated patients with celiac disease underwent oral wheat challenge to stimulate recirculation of gluten-specific T cells. Peripheral blood was collected before and after challenge. To comprehensively measure the gluten-specific CD4+ T-cell response, we paired traditional IFN-γ ELISpot with an assay to detect antigen-specific CD4+ T cells that does not rely on tetramers, antigen-stimulated cytokine production, or proliferation but rather on antigen-induced coexpression of CD25 and OX40 (CD134).

Results: Numbers of circulating gluten-specific Treg cells and effector T cells both increased significantly after oral wheat challenge, peaking at day 6. Surprisingly, we found that approximately 80% of the ex vivo circulating gluten-specific CD4+ T cells were FOXP3+CD39+ Treg cells, which reside within the pool of memory CD4+CD25+CD127lowCD45RO+ Treg cells. Although we observed normal suppressive function in peripheral polyclonal Treg cells from patients with celiac disease, after a short in vitro expansion, the gluten-specific FOXP3+CD39+ Treg cells exhibited significantly reduced suppressive function compared with polyclonal Treg cells.

Conclusion: This study provides the first estimation of FOXP3+CD39+ Treg cell frequency within circulating gluten-specific CD4+ T cells after oral gluten challenge of patients with celiac disease. FOXP3+CD39+ Treg cells comprised a major proportion of all circulating gluten-specific CD4+ T cells but had impaired suppressive function, indicating that Treg cell dysfunction might be a key contributor to disease pathogenesis.

Keywords: CD39; OX40; Regulatory T cells; celiac disease; forkhead box protein 3; gluten.

MeSH terms

  • Adult
  • Antigens, CD / metabolism
  • Apyrase / metabolism
  • Celiac Disease / immunology*
  • Cells, Cultured
  • Enzyme-Linked Immunospot Assay
  • Female
  • Forkhead Transcription Factors / metabolism
  • Glutens / immunology*
  • HLA-DQ Antigens / genetics
  • HLA-DQ Antigens / metabolism
  • Humans
  • Immunosuppression Therapy
  • Interferon-gamma / metabolism
  • Lymphocyte Count
  • Male
  • Polymorphism, Single Nucleotide
  • T-Cell Antigen Receptor Specificity / immunology
  • T-Lymphocytes, Regulatory / immunology*

Substances

  • Antigens, CD
  • FOXP3 protein, human
  • Forkhead Transcription Factors
  • HLA-DQ Antigens
  • HLA-DQ2 antigen
  • Glutens
  • Interferon-gamma
  • Apyrase
  • CD39 antigen