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:

Acquisition of a multifunctional IgA+ plasma cell phenotype in the gut

Abstract

The largest mucosal surface in the body is in the gastrointestinal tract, a location that is heavily colonized by microbes that are normally harmless. A key mechanism required for maintaining a homeostatic balance between this microbial burden and the lymphocytes that densely populate the gastrointestinal tract is the production and transepithelial transport of poly-reactive IgA (ref. 1). Within the mucosal tissues, B cells respond to cytokines, sometimes in the absence of T-cell help, undergo class switch recombination of their immunoglobulin receptor to IgA, and differentiate to become plasma cells2. However, IgA-secreting plasma cells probably have additional attributes that are needed for coping with the tremendous bacterial load in the gastrointestinal tract. Here we report that mouse IgA+ plasma cells also produce the antimicrobial mediators tumour-necrosis factor-α (TNF-α) and inducible nitric oxide synthase (iNOS), and express many molecules that are commonly associated with monocyte/granulocytic cell types. The development of iNOS-producing IgA+ plasma cells can be recapitulated in vitro in the presence of gut stroma, and the acquisition of this multifunctional phenotype in vivo and in vitro relies on microbial co-stimulation. Deletion of TNF-α and iNOS in B-lineage cells resulted in a reduction in IgA production, altered diversification of the gut microbiota and poor clearance of a gut-tropic pathogen. These findings reveal a novel adaptation to maintaining homeostasis in the gut, and extend the repertoire of protective responses exhibited by some B-lineage cells.

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: IgA + plasma cells in the small intestinal lamina propria can produce iNOS and TNF-α.
Figure 2: TNF-α and iNOS expression in IgA + plasma cells requires microbial exposure.
Figure 3: Reduced IgA production and altered commensal flora composition in iNOS/TNF-α double-deficient mixed chimaeric mice.
Figure 4: iNOS/TNF-α double-deficient mixed chimaeras are more susceptible to infection with C. rodentium.

Similar content being viewed by others

References

  1. Hooper, L. V. & Macpherson, A. J. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nature Rev. Immunol. 10, 159–169 (2010)

    Article  CAS  Google Scholar 

  2. Fagarasan, S., Kawamoto, S., Kanagawa, O. & Suzuki, K. Adaptive immune regulation in the gut: T cell-dependent and T cell-independent IgA synthesis. Annu. Rev. Immunol. 28, 243–273 (2010)

    Article  CAS  Google Scholar 

  3. Lee, M. R., Seo, G. Y., Kim, Y. M. & Kim, P. H. iNOS potentiates mouse Ig isotype switching through AID expression. Biochem. Biophys. Res. Commun. 410, 602–607 (2011)

    Article  CAS  Google Scholar 

  4. Tezuka, H. et al. Regulation of IgA production by naturally occurring TNF/iNOS-producing dendritic cells. Nature 448, 929–933 (2007)

    Article  ADS  CAS  Google Scholar 

  5. Kang, H. S. et al. Signaling via LTβR on the lamina propria stromal cells of the gut is required for IgA production. Nature Immunol. 3, 576–582 (2002)

    Article  CAS  Google Scholar 

  6. Serbina, N. V., Salazar-Mather, T. P., Biron, C. A., Kuziel, W. A. & Pamer, E. G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 19, 59–70 (2003)

    Article  CAS  Google Scholar 

  7. Muramatsu, M. et al. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell 102, 553–563 (2000)

    Article  CAS  Google Scholar 

  8. Crouch, E. E. et al. Regulation of AID expression in the immune response. J. Exp. Med. 204, 1145–1156 (2007)

    Article  CAS  Google Scholar 

  9. Serbina, N. V. & Pamer, E. G. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nature Immunol. 7, 311–317 (2006)

    Article  CAS  Google Scholar 

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

    Article  ADS  CAS  Google Scholar 

  11. Cumano, A., Dorshkind, K., Gillis, S. & Paige, C. J. The influence of S17 stromal cells and interleukin 7 on B cell development. Eur. J. Immunol. 20, 2183–2189 (1990)

    Article  CAS  Google Scholar 

  12. Tumanov, A. V. et al. Cellular source and molecular form of TNF specify its distinct functions in organization of secondary lymphoid organs. Blood 116, 3456–3464 (2010)

    Article  CAS  Google Scholar 

  13. Ivanov, I. I. & Littman, D. R. Modulation of immune homeostasis by commensal bacteria. Curr. Opin. Microbiol. 14, 106–114 (2011)

    Article  CAS  Google Scholar 

  14. Suzuki, K. et al. Aberrant expansion of segmented filamentous bacteria in IgA-deficient gut. Proc. Natl Acad. Sci. USA 101, 1981–1986 (2004)

    Article  ADS  CAS  Google Scholar 

  15. Maaser, C. et al. Clearance of Citrobacter rodentium requires B cells but not secretory immunoglobulin A (IgA) or IgM antibodies. Infect. Immun. 72, 3315–3324 (2004)

    Article  CAS  Google Scholar 

  16. Delogu, A. et al. Gene repression by Pax5 in B cells is essential for blood cell homeostasis and is reversed in plasma cells. Immunity 24, 269–281 (2006)

    Article  CAS  Google Scholar 

  17. Li, J. et al. B lymphocytes from early vertebrates have potent phagocytic and microbicidal abilities. Nature Immunol. 7, 1116–1124 (2006)

    Article  CAS  Google Scholar 

  18. Johnson, B. A. et al. B-lymphoid cells with attributes of dendritic cells regulate T cells via indoleamine 2,3-dioxygenase. Proc. Natl Acad. Sci. USA 107, 10644–10648 (2010)

    Article  ADS  CAS  Google Scholar 

  19. Kelly-Scumpia, K. M. et al. B cells enhance early innate immune responses during bacterial sepsis. J. Exp. Med. 208, 1673–1682 (2011)

    Article  CAS  Google Scholar 

  20. Neves, P. et al. Signaling via the MyD88 adaptor protein in B cells suppresses protective immunity during Salmonella typhimurium infection. Immunity 33, 777–790 (2010)

    Article  CAS  Google Scholar 

  21. Cumano, A., Paige, C. J., Iscove, N. N. & Brady, G. Bipotential precursors of B cells and macrophages in murine fetal liver. Nature 356, 612–615 (1992)

    Article  ADS  CAS  Google Scholar 

  22. Montecino-Rodriguez, E., Leathers, H. & Dorshkind, K. Bipotential B-macrophage progenitors are present in adult bone marrow. Nature Immunol. 2, 83–88 (2001)

    Article  CAS  Google Scholar 

  23. Pelletier, N. et al. Plasma cells negatively regulate the follicular helper T cell program. Nature Immunol. 11, 1110–1118 (2010)

    Article  CAS  Google Scholar 

  24. Peterson, D. A., McNulty, N. P., Guruge, J. L. & Gordon, J. I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2, 328–339 (2007)

    Article  CAS  Google Scholar 

  25. Marshall, A. J., Paige, C. J. & Wu, G. E. V. (H) repertoire maturation during B cell development in vitro: differential selection of Ig heavy chains by fetal and adult B cell progenitors. J. Immunol. 158, 4282–4291 (1997)

    CAS  PubMed  Google Scholar 

  26. Barman, M. et al. Enteric salmonellosis disrupts the microbial ecology of the murine gastrointestinal tract. Infect. Immun. 76, 907–915 (2008)

    Article  CAS  Google Scholar 

  27. Petnicki-Ocwieja, T. et al. Nod2 is required for the regulation of commensal microbiota in the intestine. Proc. Natl Acad. Sci. USA 106, 15813–15818 (2009)

    Article  ADS  CAS  Google Scholar 

  28. Furet, J. P. et al. Comparative assessment of human and farm animal faecal microbiota using real-time quantitative PCR. FEMS Microbiol. Ecol. 68, 351–362 (2009)

    Article  CAS  Google Scholar 

  29. Milne, C. D., Fleming, H. E. & Paige, C. J. IL-7 does not prevent pro-B/pre-B cell maturation to the immature/sIgM+ stage. Eur. J. Immunol. 34, 2647–2655 (2004)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank D. White in the Faculty of Medicine Flow Cytometry core facility and H. Singh for critical reading of the manuscript. We thank E. Verdu for providing additional germ-free mice at short notice, and we also thank C. Guidos for numerous Rag2−/− mice for mixed bone marrow chimeras. C.J.P. is supported by a CIHR operating grant MOP number 9862. R.C. is supported in part by the Intramural Research Program of the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health. I.I.I. is supported by NIH (R00 DK085329-02) and CCFA (CDA #2388). A.M. is supported by a CIHR operating grant MOP number 89783. J.H.F. acknowledges support by an APART-fellowship of the Austrian Academy of Sciences, McGill start-up funds and a CIHR operating grant MOP number 114972. N.S. acknowledges the support of a CIHR Doctoral Award. J.L.G. is funded by the Canadian Institutes of Health Research (CIHR) and acknowledges the support of CIHR operating grant MOP number 67157 as well as infrastructure support from the Ontario Research Fund and that Canadian Foundation for Innovation. All authors have reviewed and agree with the content of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

J.H.F. generated data in Figs 1d–f, 3a–d, 4d–f and Supplementary Figs 2–4, 7 and 8. O.L.R. contributed data in Figs 1a–d, 3a, b, 4, Supplementary Figs 7, 8 and Supplementary Tables 1–3. N.S. and C.J.P. contributed data in Fig. 2b and Supplementary Fig. 6. D.D.M. contributed data in Figs 1a, b, 2a and Supplementary Figs 1–3. S.H. contributed data in Fig. 2a. A.M. and M.L. contributed Supplementary Fig. 1b (D.M. did the sort). R.C. provided AID–YFP mice. I.I.I. originally suggested that we examine IgA+ plasma cells as putative TNF-α/iNOS-producing cells and urged us to do the initial experiments. D.J.P and S.J.R. contributed data in Fig. 3e and provided critical insights. S.E.G. and S.R. helped us set up the Citrobacter rodentium experiments and provided critical insights. A.J.M., S.H. and K.D.M. provided intestinal tissues from germ-free and re-colonized mice. J.G. provided gene-deficient mice. J.L.G. wrote the manuscript and obtained funding for the work from the Canadian Institutes of Health Research.

Corresponding author

Correspondence to Jennifer L. Gommerman.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-8, Supplementary Tables 1-4 and an additional reference. (PDF 3061 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Fritz, J., Rojas, O., Simard, N. et al. Acquisition of a multifunctional IgA+ plasma cell phenotype in the gut. Nature 481, 199–203 (2012). https://doi.org/10.1038/nature10698

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature10698

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