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:

High abundance of plasma cells secreting transglutaminase 2–specific IgA autoantibodies with limited somatic hypermutation in celiac disease intestinal lesions

Abstract

Celiac disease is an immune-mediated disorder in which mucosal autoantibodies to the enzyme transglutaminase 2 (TG2)1 are generated in response to the exogenous antigen gluten2 in individuals who express human leukocyte antigen HLA-DQ2 or HLA-DQ8 (ref. 3). We assessed in a comprehensive and nonbiased manner the IgA anti-TG2 response by expression cloning of the antibody repertoire of ex vivo–isolated intestinal antibody-secreting cells (ASCs). We found that TG2-specific plasma cells are markedly expanded within the duodenal mucosa in individuals with active celiac disease. TG2-specific antibodies were of high affinity yet showed little adaptation by somatic mutations. Unlike infection-induced peripheral blood plasmablasts4, the TG2-specific ASCs had not recently proliferated and were not short-lived ex vivo. Altogether, these observations demonstrate that there is a germline repertoire with high affinity for TG2 that may favor massive generation of autoreactive B cells. TG2-specific antibodies did not block enzymatic activity and served as substrates for TG2-mediated crosslinking when expressed as IgD or IgM but not as IgA1 or IgG1. This could result in preferential recruitment of plasma cells from naive IgD- and IgM-expressing B cells, thus possibly explaining why the antibody response to TG2 bears signs of a primary immune response despite the disease chronicity.

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: TG2-specific ASCs are localized in the duodenal mucosa of celiac disease patients.
Figure 2: TG2-specific ASCs identified and sorted by flow cytometry for quantification and cloning of antibody variable genes.
Figure 3: The highly restricted repertoire of TG2-specific ASCs suggests a unique origin for this autoantibody response.
Figure 4: TG2 covalently crosslinks antibodies of the IgD isotype and its enzymatic activity is not inhibited by anti-TG2 hmAbs.

Similar content being viewed by others

References

  1. Dieterich, W. et al. Identification of tissue transglutaminase as the autoantigen of celiac disease. Nat. Med. 3, 797–801 (1997).

    Article  CAS  Google Scholar 

  2. Sulkanen, S. et al. Tissue transglutaminase autoantibody enzyme-linked immunosorbent assay in detecting celiac disease. Gastroenterology 115, 1322–1328 (1998).

    Article  CAS  Google Scholar 

  3. Björck, S., Brundin, C., Lorinc, E., Lynch, K.F. & Agardh, D. Screening detects a high proportion of celiac disease in young HLA-genotyped children. J. Pediatr. Gastroenterol. Nutr. 50, 49–53 (2010).

    Article  Google Scholar 

  4. Wrammert, J. et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 453, 667–671 (2008).

    Article  CAS  Google Scholar 

  5. Sollid, L.M. Coeliac disease: dissecting a complex inflammatory disorder. Nat. Rev. Immunol. 2, 647–655 (2002).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  7. Sollid, L.M., Molberg, O., McAdam, S. & Lundin, K.E. Autoantibodies in coeliac disease: tissue transglutaminase—guilt by association? Gut 41, 851–852 (1997).

    Article  CAS  Google Scholar 

  8. Rostom, A. et al. The diagnostic accuracy of serologic tests for celiac disease: a systematic review. Gastroenterology 128, S38–S46 (2005).

    Article  Google Scholar 

  9. Salmi, T.T. et al. Endomysial antibody-negative coeliac disease: clinical characteristics and intestinal autoantibody deposits. Gut 55, 1746–1753 (2006).

    Article  CAS  Google Scholar 

  10. Marzari, R. et al. Molecular dissection of the tissue transglutaminase autoantibody response in celiac disease. J. Immunol. 166, 4170–4176 (2001).

    Article  CAS  Google Scholar 

  11. Di Niro, R. et al. Rapid generation of rotavirus-specific human monoclonal antibodies from small-intestinal mucosa. J. Immunol. 185, 5377–5383 (2010).

    Article  CAS  Google Scholar 

  12. Benckert, J. et al. The majority of intestinal IgA+ and IgG+ plasmablasts in the human gut are antigen-specific. J. Clin. Invest. 121, 1946–1955 (2011).

    Article  CAS  Google Scholar 

  13. Mei, H.E. et al. Blood-borne human plasma cells in steady state are derived from mucosal immune responses. Blood 113, 2461–2469 (2009).

    Article  CAS  Google Scholar 

  14. Meffre, E. & Wardemann, H. B-cell tolerance checkpoints in health and autoimmunity. Curr. Opin. Immunol. 20, 632–638 (2008).

    Article  CAS  Google Scholar 

  15. Tiller, T. et al. Autoreactivity in human IgG+ memory B cells. Immunity 26, 205–213 (2007).

    Article  CAS  Google Scholar 

  16. Kurppa, K. et al. Celiac disease without villous atrophy in children: a prospective study. J. Pediatr. 157, 373–380 (2010).

    Article  Google Scholar 

  17. Green, P.H. The many faces of celiac disease: clinical presentation of celiac disease in the adult population. Gastroenterology 128, S74–S78 (2005).

    Article  Google Scholar 

  18. Scheid, J.F. et al. Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 458, 636–640 (2009).

    Article  CAS  Google Scholar 

  19. MacLennan, I.C. et al. Extrafollicular antibody responses. Immunol. Rev. 194, 8–18 (2003).

    Article  CAS  Google Scholar 

  20. Chan, T.D. et al. Antigen affinity controls rapid T-dependent antibody production by driving the expansion rather than the differentiation or extrafollicular migration of early plasmablasts. J. Immunol. 183, 3139–3149 (2009).

    Article  CAS  Google Scholar 

  21. Boursier, L., Dunn-Walters, D.K. & Spencer, J. Characteristics of IgVH genes used by human intestinal plasma cells from childhood. Immunology 97, 558–564 (1999).

    Article  CAS  Google Scholar 

  22. He, B. et al. Intestinal bacteria trigger T cell-independent immunoglobulin A(2) class switching by inducing epithelial-cell secretion of the cytokine APRIL. Immunity 26, 812–826 (2007).

    Article  CAS  Google Scholar 

  23. Goodnow, C.C., Vinuesa, C.G., Randall, K.L., Mackay, F. & Brink, R. Control systems and decision making for antibody production. Nat. Immunol. 11, 681–688 (2010).

    Article  CAS  Google Scholar 

  24. Shokat, K.M. & Goodnow, C.C. Antigen-induced B-cell death and elimination during germinal-centre immune responses. Nature 375, 334–338 (1995).

    Article  CAS  Google Scholar 

  25. Anderson, S.M. et al. Taking advantage: high-affinity B cells in the germinal center have lower death rates, but similar rates of division, compared to low-affinity cells. J. Immunol. 183, 7314–7325 (2009).

    Article  CAS  Google Scholar 

  26. Batista, F.D. & Neuberger, M.S. Affinity dependence of the B cell response to antigen: a threshold, a ceiling, and the importance of off-rate. Immunity 8, 751–759 (1998).

    Article  CAS  Google Scholar 

  27. Toellner, K.M. et al. Low-level hypermutation in T cell-independent germinal centers compared with high mutation rates associated with T cell-dependent germinal centers. J. Exp. Med. 195, 383–389 (2002).

    Article  CAS  Google Scholar 

  28. Kroese, F.G., Butcher, E.C., Stall, A.M. & Herzenberg, L.A. A major peritoneal reservoir of precursors for intestinal IgA plasma cells. Immunol. Invest. 18, 47–58 (1989).

    Article  CAS  Google Scholar 

  29. Macpherson, A.J. et al. A primitive T cell–independent mechanism of intestinal mucosal IgA responses to commensal bacteria. Science 288, 2222–2226 (2000).

    Article  CAS  Google Scholar 

  30. Victora, G.D. et al. Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 143, 592–605 (2010).

    Article  CAS  Google Scholar 

  31. Hapfelmeier, S. et al. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science 328, 1705–1709 (2010).

    Article  CAS  Google Scholar 

  32. Mesin, L., Di Niro, R., Thompson, K.M., Lundin, K.E. & Sollid, L.M. Long-lived plasma cells from human small intestine biopsies secrete immunoglobulins for many weeks in vitro. J. Immunol. 187, 2867–2874 (2011).

    Article  CAS  Google Scholar 

  33. Radbruch, A. et al. Competence and competition: the challenge of becoming a long-lived plasma cell. Nat. Rev. Immunol. 6, 741–750 (2006).

    Article  CAS  Google Scholar 

  34. Halstensen, T.S., Hvatum, M., Scott, H., Fausa, O. & Brandtzaeg, P. Association of subepithelial deposition of activated complement and immunoglobulin G and M response to gluten in celiac disease. Gastroenterology 102, 751–759 (1992).

    Article  CAS  Google Scholar 

  35. Borrelli, M. et al. High density of intraepithelial gammadelta lymphocytes and deposits of immunoglobulin (Ig)M anti-tissue transglutaminase antibodies in the jejunum of coeliac patients with IgA deficiency. Clin. Exp. Immunol. 160, 199–206 (2010).

    Article  CAS  Google Scholar 

  36. Duty, J.A. et al. Functional anergy in a subpopulation of naive B cells from healthy humans that express autoreactive immunoglobulin receptors. J. Exp. Med. 206, 139–151 (2009).

    Article  Google Scholar 

  37. Stamnaes, J., Fleckenstein, B. & Sollid, L.M. The propensity for deamidation and transamidation of peptides by transglutaminase 2 is dependent on substrate affinity and reaction conditions. Biochim. Biophys. Acta 1784, 1804–1811 (2008).

    Article  CAS  Google Scholar 

  38. Smith, K. et al. Rapid generation of fully human monoclonal antibodies specific to a vaccinating antigen. Nat. Protoc. 4, 372–384 (2009).

    Article  CAS  Google Scholar 

  39. Sitia, R. et al. Developmental regulation of IgM secretion: the role of the carboxy-terminal cysteine. Cell 60, 781–790 (1990).

    Article  CAS  Google Scholar 

  40. Qiao, S.W. et al. Posttranslational modification of gluten shapes TCR usage in celiac disease. J. Immunol. 187, 3064–3071 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank E. Mummert (Phadia) for providing recombinant human TG2 produced in Sf9 insect cells, C. Khosla (Stanford University) and L. Fesus (University of Debrecen) for providing plasmids encoding His-tagged recombinant TG2 for expression in Escherichia coli, M. Ráki for help in sample collection and immunofluorescence analysis, J. Jahnsen for providing biopsy samples from two subjects, and the staffs at the LIIPAT laboratory and the flow cytometry core facility (both at Oslo University Hospital-Rikshospitalet) for technical assistance. This work was supported by the Marie Curie Research and Training Network (European Commission) contract MRTN-CT-2006-036032, the European Research Council and grants from the Research Council of Norway to L.M.S., and US National Institutes of Health National Institute of Allergy and Infectious Disease grants U19 AI082724-01, R01 AI76585-03 and HHSN266200500026C to P.C.W. We thank M. Shlomchik and B. Jabri for discussion and critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

R.D.N. conceived the study, performed experiments, analyzed data and wrote the manuscript. L.M., N.-Y.Z., J.S., M.M., J.-H.L., M.H., R.I., M.F.d.P. and S.-W.Q. performed experiments. K.E.A.L. recruited study subjects and provided materials. P.C.W. supervised the study, analyzed data and revised the manuscript. L.M.S. conceived and supervised the study and wrote the manuscript.

Corresponding authors

Correspondence to Patrick C Wilson or Ludvig M Sollid.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–13, Supplementary Tables 1 and 2 and Supplementary Methods (PDF 4662 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Di Niro, R., Mesin, L., Zheng, NY. et al. High abundance of plasma cells secreting transglutaminase 2–specific IgA autoantibodies with limited somatic hypermutation in celiac disease intestinal lesions. Nat Med 18, 441–445 (2012). https://doi.org/10.1038/nm.2656

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2656

This article is cited by

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