Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

The role of HLA-DQ8 β57 polymorphism in the anti-gluten T-cell response in coeliac disease

Abstract

Major histocompatibility complex (MHC) class II alleles HLA-DQ8 and the mouse homologue I-Ag7 lacking a canonical aspartic acid residue at position β57 are associated with coeliac disease1,2 and type I diabetes3,4. However, the role of this single polymorphism in disease initiation and progression remains poorly understood. The lack of Asp 57 creates a positively charged P9 pocket, which confers a preference for negatively charged peptides. Gluten lacks such peptides, but tissue transglutaminase (TG2) introduces negatively charged residues at defined positions into gluten T-cell epitopes by deamidating specific glutamine residues5,6 on the basis of their spacing to proline residues7. The commonly accepted model, proposing that HLA-DQ8 simply favours binding of negatively charged peptides, does not take into account the fact that TG2 requires inflammation for activation8 and that T-cell responses against native gluten peptides are found9,10, particularly in children11. Here we show that β57 polymorphism promotes the recruitment of T-cell receptors bearing a negative signature charge in the complementary determining region 3β (CDR3β) during the response against native gluten peptides presented by HLA-DQ8 in coeliac disease. These T cells showed a crossreactive and heteroclitic (stronger) response to deamidated gluten peptides. Furthermore, gluten peptide deamidation extended the T-cell-receptor repertoire by relieving the requirement for a charged residue in CDR3β. Thus, the lack of a negative charge at position β57 in MHC class II was met by negatively charged residues in the T-cell receptor or in the peptide, the combination of which might explain the role of HLA-DQ8 in amplifying the T-cell response against dietary gluten.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Native and deamidated gluten α2-219–242 peptides recruit distinct, yet overlapping TCR repertoires.
Figure 2: Negative charge at position 3 of the TCR CDR3β loop is critical for native gluten peptide reactivity.
Figure 3: Coimmunization with native and deamidated peptides amplifies the T-cell response to deamidated peptide.

Similar content being viewed by others

References

  1. Sollid, L. M. et al. Evidence for a primary association of celiac disease to a particular HLA-DQ α/β heterodimer. J. Exp. Med. 169, 345–350 (1989)

    Article  CAS  PubMed  Google Scholar 

  2. Spurkland, A., Sollid, L. M., Polanco, I., Vartdal, F. & Thorsby, E. HLA-DR and -DQ genotypes of celiac disease patients serologically typed to be non-DR3 or non-DR5/7. Hum. Immunol. 35, 188–192 (1992)

    Article  CAS  PubMed  Google Scholar 

  3. Acha-Orbea, H. & McDevitt, H. O. The first domain of the nonobese diabetic mouse class II I-A β chain is unique. Proc. Natl Acad. Sci. USA 84, 2435–2439 (1987)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  4. Hattori, M. et al. The NOD mouse: recessive diabetogenic gene in the major histocompatibility complex. Science 231, 733–735 (1986)

    Article  CAS  ADS  PubMed  Google Scholar 

  5. Molberg, O. et al. Tissue transglutaminase selectively modifies gliadin peptides that are recognized by gut-derived T cells in celiac disease. Nature Med. 4, 713–717 (1998)

    Article  CAS  PubMed  Google Scholar 

  6. van de Wal, Y. et al. Selective deamidation by tissue transglutaminase strongly enhances gliadin-specific T cell reactivity. J. Immunol. 161, 1585–1588 (1998)

    CAS  PubMed  Google Scholar 

  7. Vader, L. W. et al. Specificity of tissue transglutaminase explains cereal toxicity in celiac disease. J. Exp. Med. 195, 643–649 (2002)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Siegel, M. et al. Extracellular transglutaminase 2 is catalytically inactive, but is transiently activated upon tissue injury. PLoS ONE 3, e1861 (2008)

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  9. Henderson, K. N. et al. A structural and immunological basis for the role of human leukocyte antigen DQ8 in celiac disease. Immunity 27, 23–34 (2007)

    Article  CAS  PubMed  Google Scholar 

  10. van de Wal, Y. et al. Small intestinal T cells of celiac disease patients recognize a natural pepsin fragment of gliadin. Proc. Natl Acad. Sci. USA 95, 10050–10054 (1998)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  11. Vader, W. et al. The gluten response in children with celiac disease is directed toward multiple gliadin and glutenin peptides. Gastroenterology 122, 1729–1737 (2002)

    Article  CAS  PubMed  Google Scholar 

  12. Jabri, B. & Sollid, L. M. Mechanisms of disease: immunopathogenesis of celiac disease. Nature Clin. Pract. Gastroenterol. Hepatol. 3, 516–525 (2006)

    Article  CAS  Google Scholar 

  13. Lundin, K. E. et al. Gliadin-specific, HLA-DQ (α1*0501, β1*0201) restricted T cells isolated from the small intestinal mucosa of celiac disease patients. J. Exp. Med. 178, 187–196 (1993)

    Article  CAS  PubMed  Google Scholar 

  14. Lundin, K. E., Scott, H., Fausa, O., Thorsby, E. & Sollid, L. M. T cells from the small intestinal mucosa of a FR4, DQ7/DR4, DQ8 celiac disease patient preferentially recognize gliadin when presented by DQ8. Hum. Immunol. 41, 285–291 (1994)

    Article  CAS  PubMed  Google Scholar 

  15. Black, K. E., Murray, J. A. & David, C. S. HLA-DQ determines the response to exogenous wheat proteins: a model of gluten sensitivity in transgenic knockout mice. J. Immunol. 169, 5595–5600 (2002)

    Article  CAS  PubMed  Google Scholar 

  16. Moustakas, A. K. et al. Structure of celiac disease-associated HLA-DQ8 and non-associated HLA-DQ9 alleles in complex with two disease-specific epitopes. Int. Immunol. 12, 1157–1166 (2000)

    Article  CAS  PubMed  Google Scholar 

  17. Solinger, A. M., Ultee, M. E., Margoliash, E. & Schwartz, R. H. T-lymphocyte response to cytochrome c. I. Demonstration of a T-cell heteroclitic proliferative response and identification of a topographic antigenic determinant on pigeon cytochrome c whose immune recognition requires two complementing major histocompatibility complex-linked immune response genes. J. Exp. Med. 150, 830–848 (1979)

    Article  CAS  PubMed  Google Scholar 

  18. Bankovich, A. J., Girvin, A. T., Moesta, A. K. & Garcia, K. C. Peptide register shifting within the MHC groove: theory becomes reality. Mol. Immunol. 40, 1033–1039 (2004)

    Article  CAS  PubMed  Google Scholar 

  19. Baker, F. J., Lee, M., Chien, Y. H. & Davis, M. M. Restricted islet-cell reactive T cell repertoire of early pancreatic islet infiltrates in NOD mice. Proc. Natl Acad. Sci. USA 99, 9374–9379 (2002)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  20. Yang, Y., Charlton, B., Shimada, A., Dal Canto, R. & Fathman, C. G. Monoclonal T cells identified in early NOD islet infiltrates. Immunity 4, 189–194 (1996)

    Article  CAS  PubMed  Google Scholar 

  21. Chao, C. C., Sytwu, H. K., Chen, E. L., Toma, J. & McDevitt, H. O. The role of MHC class II molecules in susceptibility to type I diabetes: identification of peptide epitopes and characterization of the T cell repertoire. Proc. Natl Acad. Sci. USA 96, 9299–9304 (1999)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  22. Yu, B., Gauthier, L., Hausmann, D. H. & Wucherpfennig, K. W. Binding of conserved islet peptides by human and murine MHC class II molecules associated with susceptibility to type I diabetes. Eur. J. Immunol. 30, 2497–2506 (2000)

    Article  CAS  PubMed  Google Scholar 

  23. Ploski, R., Ek, J., Thorsby, E. & Sollid, L. M. On the HLA-DQ (α1*0501, β1*0201)-associated susceptibility in celiac disease: a possible gene dosage effect of DQB1*0201. Tissue Antigens 41, 173–177 (1993)

    Article  CAS  PubMed  Google Scholar 

  24. Vader, W. et al. The HLA-DQ2 gene dose effect in celiac disease is directly related to the magnitude and breadth of gluten-specific T cell responses. Proc. Natl Acad. Sci. USA 100, 12390–12395 (2003)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  25. Quinn, A. et al. T cells to a dominant epitope of GAD65 express a public CDR3 motif. Int. Immunol. 18, 967–979 (2006)

    Article  CAS  PubMed  Google Scholar 

  26. Bendelac, A. et al. CD1 recognition by mouse NK1+ T lymphocytes. Science 268, 863–865 (1995)

    Article  CAS  ADS  PubMed  Google Scholar 

  27. Casanova, J. L., Romero, P., Widmann, C., Kourilsky, P. & Maryanski, J. L. T cell receptor genes in a series of class I major histocompatibility complex-restricted cytotoxic T lymphocyte clones specific for a Plasmodium berghei nonapeptide: implications for T cell allelic exclusion and antigen-specific repertoire. J. Exp. Med. 174, 1371–1383 (1991)

    Article  CAS  PubMed  Google Scholar 

  28. Arden, B., Clark, S. P., Kabelitz, D. & Mak, T. W. Mouse T cell receptor variable gene segment families. Immunogenetics 42, 501–530 (1995)

    CAS  PubMed  Google Scholar 

  29. Blank, U., Boitel, B., Mege, D., Ermonval, M. & Acuto, O. Analysis of tetanus toxin peptide/DR recognition by human T cell receptors reconstituted into a murine T cell hybridoma. Eur. J. Immunol. 23, 3057–3065 (1993)

    Article  CAS  PubMed  Google Scholar 

  30. Scott, C. A., Garcia, K. C., Carbone, F. R., Wilson, I. A. & Teyton, L. Role of chain pairing for the production of functional soluble IA major histocompatibility complex class II molecules. J. Exp. Med. 183, 2087–2095 (1996)

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. Sadegh-Nasseri for advice and discussion, and A. Bendelac and M. Mush for critical reading of the manuscript. This work was supported by the Digestive Disease Research Core Center of the University of Chicago (DK42086), NIH DK67180, DK55037, the Celiac Disease Consortium (F.K.), EU MC-RTN 512385 (M.W.), the Research Council of Norway (L.S.) and the University of Oslo (S.T.).

Author Contributions Z.H. executed most of the studies, participated in conceptual development and in the preparation of the manuscript; A.W., A.M., C.C., S.A.C., K.Y. provided technical assistance and input into data analysis. M.W., S.T., L.M.S. and F.K. generated and characterized human T-cell clones. C.S.D. and J.A.M. provided humanized HLA-DQ8 transgenic mice. F.K. and L.M.S. reviewed the manuscript. L.T. provided input into conceptual development and experimental design. B.J. developed the concept, supervised all investigations and wrote the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bana Jabri.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-3 with Legends, Supplementary Tables 1-3 and Supplementary Methods. (PDF 995 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hovhannisyan, Z., Weiss, A., Martin, A. et al. The role of HLA-DQ8 β57 polymorphism in the anti-gluten T-cell response in coeliac disease. Nature 456, 534–538 (2008). https://doi.org/10.1038/nature07524

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature07524

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing