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.

  • Review Article
  • Published:

AIRE expands: new roles in immune tolerance and beyond

Key Points

  • Autoimmune regulator (AIRE) has a well-known role in preventing autoimmunity through upregulation of tissue-specific antigen (TSA) expression in medullary thymic epithelial cells (mTECs). Recognition of these thymic TSAs by self-reactive T cells leads to clonal deletion and/or diversion to the regulatory T cell lineage.

  • Mutations in AIRE result in multi-organ autoimmune disease in both humans and mice. In humans, autosomal recessive mutations result in autoimmune polyendocrinopathy syndrome 1, whereas dominant mutations result in autoimmunity with a more narrow disease spectrum.

  • AIRE expression is under strict spatiotemporal control. Regulation of AIRE expression is achieved through several mechanisms, including enhancer elements that regulate the transcription and alternative splicing of AIRE, which in turn control AIRE protein levels.

  • The array of TSAs expressed by each individual mTEC is diverse. Nevertheless, clusters of TSAs are co-expressed, with distinct rules governing their co-expression.

  • AIRE interacts with dozens of proteins with various functions, including the recruitment of AIRE to TSA genes, elongation of AIRE-dependent TSA transcripts and modification of AIRE itself.

  • AIRE has important roles in conditions beyond autoimmunity, such as graft-versus-host disease and cancer. Thus, modulation of AIRE function may have potential therapeutic benefit in a wide range of diseases.

Abstract

More than 15 years ago, mutations in the autoimmune regulator (AIRE) gene were identified as the cause of autoimmune polyglandular syndrome type 1 (APS1). It is now clear that this transcription factor has a crucial role in promoting self-tolerance in the thymus by regulating the expression of a wide array of self-antigens that have the commonality of being tissue-restricted in their expression pattern in the periphery. In this Review, we highlight many of the recent advances in our understanding of the complex biology that is related to AIRE, with a particular focus on advances in genetics, molecular interactions and the effect of AIRE on thymic selection of regulatory T cells. Furthermore, we highlight new areas of biology that are potentially affected by this key regulator of immune tolerance.

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: AIRE mutations associated with autoimmune disease.
Figure 2: AIRE-regulated TSA expression is 'stochastic' and 'ordered'.
Figure 3: AIRE and its binding partners.
Figure 4: The various functions of AIRE in mTECs.
Figure 5: AIRE enforces central tolerance towards self-antigens.

Similar content being viewed by others

References

  1. Cheng, M. H. & Anderson, M. S. Monogenic autoimmunity. Annu. Rev. Immunol. 30, 393–427 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bennett, C. L. et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat. Genet. 27, 20–21 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Brunkow, M. E. et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat. Genet. 27, 68–73 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Wildin, R. S. et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat. Genet. 27, 18–20 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Kuehn, H. S. et al. Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4. Science 345, 1623–1627 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Schubert, D. et al. Autosomal dominant immune dysregulation syndrome in humans with CTLA4 mutations. Nat. Med. 20, 1410–1416 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Finnish-German APECED Consortium. An autoimmune disease, APECED, caused by mutations in a novel gene featuring two PHD-type zinc-finger domains. Nat. Genet. 17, 399–403 (1997).

  8. Nagamine, K. et al. Positional cloning of the APECED gene. Nat. Genet. 17, 393–398 (1997).

    Article  CAS  PubMed  Google Scholar 

  9. Anderson, M. S. et al. Projection of an immunological self shadow within the thymus by the AIRE protein. Science 298, 1395–1401 (2002).

    Article  CAS  PubMed  Google Scholar 

  10. DeVoss, J. et al. Spontaneous autoimmunity prevented by thymic expression of a single self-antigen. J. Exp. Med. 203, 2727–2735 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Su, M. A. et al. Defective autoimmune regulator-dependent central tolerance to myelin protein zero is linked to autoimmune peripheral neuropathy. J. Immunol. 188, 4906–4912 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Metzger, T. C. & Anderson, M. S. Control of central and peripheral tolerance by Aire. Immunol. Rev. 241, 89–103 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Cetani, F. et al. A novel mutation of the autoimmune regulator gene in an Italian kindred with autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy, acting in a dominant fashion and strongly cosegregating with hypothyroid autoimmune thyroiditis. J. Clin. Endocrinol. Metab. 86, 4747–4752 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Su, M. A. et al. Mechanisms of an autoimmunity syndrome in mice caused by a dominant mutation in Aire. J. Clin. Invest. 118, 1712–1726 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Oftedal, B. E. et al. Dominant mutations in the autoimmune regulator AIRE are associated with common organ-specific autoimmune diseases. Immunity 42, 1185–1196 (2015). This study provides additional evidence that autosomal dominant mutations in AIRE can predispose to autoimmunity and may be more widespread than initially thought.

    Article  CAS  PubMed  Google Scholar 

  16. Pitkanen, J. et al. The autoimmune regulator protein has transcriptional transactivating properties and interacts with the common coactivator CREB-binding protein. J. Biol. Chem. 275, 16802–16809 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Koh, A. S. et al. Aire employs a histone-binding module to mediate immunological tolerance, linking chromatin regulation with organ-specific autoimmunity. Proc. Natl Acad. Sci. USA 105, 15878–15883 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Org, T. et al. The autoimmune regulator PHD finger binds to non-methylated histone H3K4 to activate gene expression. EMBO Rep. 9, 370–376 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Waterfield, M. et al. The transcriptional regulator Aire coopts the repressive ATF7ip-MBD1 complex for the induction of immunotolerance. Nat. Immunol. 15, 258–265 (2014). This study provides evidence for a mechanism by which AIRE may recognize TSA target genes, involving the ATF7IP–MBD1 repressive complex.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nishikawa, Y. et al. Biphasic Aire expression in early embryos and in medullary thymic epithelial cells before end-stage terminal differentiation. J. Exp. Med. 207, 963–971 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gardner, J. M. et al. Extrathymic Aire-expressing cells are a distinct bone marrow-derived population that induce functional inactivation of CD4+ T cells. Immunity 39, 560–572 (2013). This study further clarifies a peripheral cell population that expresses AIRE and may tolerize CD4+ T cells through an anergic mechanism.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yamano, T. et al. Thymic B cells are licensed to present self antigens for central T cell tolerance induction. Immunity 42, 1048–1061 (2015). This interesting study shows that thymic B cells can acquire AIRE expression through a RANK signalling mechanism.

    Article  CAS  PubMed  Google Scholar 

  23. Metzger, T. C. et al. Lineage tracing and cell ablation identify a post-Aire-expressing thymic epithelial cell population. Cell Rep. 5, 166–179 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. LaFlam, T. N. et al. Identification of a novel cis-regulatory element essential for immune tolerance. J. Exp. Med. 212, 1993–2002 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Haljasorg, U. et al. A highly conserved NF-κB-responsive enhancer is critical for thymic expression of Aire in mice. Eur. J. Immunol. 45, 3246–3256 (2015). References 24 and 25 identify a highly conserved non-coding sequence that is crucial for the expression of thymic Aire . The sequence contains two conserved NF-κB responsive elements that may link RANK signalling to this element.

    Article  CAS  PubMed  Google Scholar 

  26. Yanagihara, T. et al. Intronic regulation of Aire expression by Jmjd6 for self-tolerance induction in the thymus. Nat. Commun. 6, 8820 (2015).

    Article  CAS  PubMed  Google Scholar 

  27. Danso-Abeam, D. et al. Aire mediates thymic expression and tolerance of pancreatic antigens via an unconventional transcriptional mechanism. Eur. J. Immunol. 43, 75–84 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Sansom, S. N. et al. Population and single-cell genomics reveal the Aire dependency, relief from Polycomb silencing, and distribution of self-antigen expression in thymic epithelia. Genome Res. 24, 1918–1931 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gray, D. H. et al. Developmental kinetics, turnover, and stimulatory capacity of thymic epithelial cells. Blood 108, 3777–3785 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Brennecke, P. et al. Single-cell transcriptome analysis reveals coordinated ectopic gene-expression patterns in medullary thymic epithelial cells. Nat. Immunol. 16, 933–941 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Meredith, M., Zemmour, D., Mathis, D. & Benoist, C. Aire controls gene expression in the thymic epithelium with ordered stochasticity. Nat. Immunol. 16, 942–949 (2015). References 30 and 31 use state-of-the-art single-cell transcript sequencing to further characterize the properties of TSA expression in mTECs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Derbinski, J., Pinto, S., Rosch, S., Hexel, K. & Kyewski, B. Promiscuous gene expression patterns in single medullary thymic epithelial cells argue for a stochastic mechanism. Proc. Natl Acad. Sci. USA 105, 657–662 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Villasenor, J., Besse, W., Benoist, C. & Mathis, D. Ectopic expression of peripheral-tissue antigens in the thymic epithelium: probabilistic, monoallelic, misinitiated. Proc. Natl Acad. Sci. USA 105, 15854–15859 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pinto, S. et al. Overlapping gene coexpression patterns in human medullary thymic epithelial cells generate self-antigen diversity. Proc. Natl Acad. Sci. USA 110, E3497–E3505 (2013). This study uses a cell-sorting approach to capture pools of distinct mTECs on the basis of their TSA expression and provides evidence that TSA expression involves coordination of distinct regions of the chromatin in each cell.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Abramson, J., Giraud, M., Benoist, C. & Mathis, D. Aire's partners in the molecular control of immunological tolerance. Cell 140, 123–135 (2010).

    Article  CAS  PubMed  Google Scholar 

  36. Gaetani, M. et al. AIRE-PHD fingers are structural hubs to maintain the integrity of chromatin-associated interactome. Nucleic Acids Res. 40, 11756–11768 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Giraud, M. et al. An RNAi screen for Aire cofactors reveals a role for Hnrnpl in polymerase release and Aire-activated ectopic transcription. Proc. Natl Acad. Sci. USA 111, 1491–1496 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Chignola, F. et al. The solution structure of the first PHD finger of autoimmune regulator in complex with non-modified histone H3 tail reveals the antagonistic role of H3R2 methylation. Nucleic Acids Res. 37, 2951–2961 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Koh, A. S., Kingston, R. E., Benoist, C. & Mathis, D. Global relevance of Aire binding to hypomethylated lysine-4 of histone-3. Proc. Natl Acad. Sci. USA 107, 13016–13021 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Zumer, K., Low, A. K., Jiang, H., Saksela, K. & Peterlin, B. M. Unmodified histone H3K4 and DNA-dependent protein kinase recruit autoimmune regulator to target genes. Mol. Cell. Biol. 32, 1354–1362 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Oven, I. et al. AIRE recruits P-TEFb for transcriptional elongation of target genes in medullary thymic epithelial cells. Mol. Cell. Biol. 27, 8815–8823 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zumer, K., Plemenitas, A., Saksela, K. & Peterlin, B. M. Patient mutation in AIRE disrupts P-TEFb binding and target gene transcription. Nucleic Acids Res. 39, 7908–7919 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Giraud, M. et al. Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells. Proc. Natl Acad. Sci. USA 109, 535–540 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Incani, F. et al. AIRE acetylation and deacetylation: effect on protein stability and transactivation activity. J. Biomed. Sci. 21, 85 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Saare, M., Rebane, A., Rajashekar, B., Vilo, J. & Peterson, P. Autoimmune regulator is acetylated by transcription coactivator CBP/p300. Exp. Cell Res. 318, 1767–1778 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Chuprin, A. et al. The deacetylase Sirt1 is an essential regulator of Aire-mediated induction of central immunological tolerance. Nat. Immunol. 16, 737–745 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Liiv, I. et al. DNA-PK contributes to the phosphorylation of AIRE: importance in transcriptional activity. Biochim. Biophys. Acta 1783, 74–83 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rattay, K. et al. Homeodomain-interacting protein kinase 2, a novel autoimmune regulator interaction partner, modulates promiscuous gene expression in medullary thymic epithelial cells. J. Immunol. 194, 921–928 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Yang, S., Bansal, K., Lopes, J., Benoist, C. & Mathis, D. Aire's plant homeodomain(PHD)-2 is critical for induction of immunological tolerance. Proc. Natl Acad. Sci. USA 110, 1833–1838 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Binda, O. On your histone mark, SET, methylate! Epigenetics 8, 457–463 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ju, B. G. et al. A topoisomerase IIβ-mediated dsDNA break required for regulated transcription. Science 312, 1798–1802 (2006).

    Article  CAS  PubMed  Google Scholar 

  52. Zumer, K., Saksela, K. & Peterlin, B. M. The mechanism of tissue-restricted antigen gene expression by AIRE. J. Immunol. 190, 2479–2482 (2013).

    Article  CAS  PubMed  Google Scholar 

  53. Yoshida, H. et al. Brd4 bridges the transcriptional regulators, Aire and P-TEFb, to promote elongation of peripheral-tissue antigen transcripts in thymic stromal cells. Proc. Natl Acad. Sci. USA 112, E4448–4457 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Liston, A., Lesage, S., Wilson, J., Peltonen, L. & Goodnow, C. C. Aire regulates negative selection of organ-specific T cells. Nat. Immunol. 4, 350–354 (2003).

    Article  CAS  PubMed  Google Scholar 

  55. Anderson, M. S. et al. The cellular mechanism of Aire control of T cell tolerance. Immunity 23, 227–239 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Taniguchi, R. T. et al. Detection of an autoreactive T-cell population within the polyclonal repertoire that undergoes distinct autoimmune regulator (Aire)-mediated selection. Proc. Natl Acad. Sci. USA 109, 7847–7852 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Aschenbrenner, K. et al. Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nat. Immunol. 8, 351–358 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Lei, Y. et al. Aire-dependent production of XCL1 mediates medullary accumulation of thymic dendritic cells and contributes to regulatory T cell development. J. Exp. Med. 208, 383–394 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Yang, S., Fujikado, N., Kolodin, D., Benoist, C. & Mathis, D. Immune tolerance. Regulatory T cells generated early in life play a distinct role in maintaining self-tolerance. Science 348, 589–594 (2015). This recent study suggests that a unique, early repertoire of T Reg cells is generated in the thymus and seeds tissues to help maintain tolerance.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Malchow, S. et al. Aire-dependent thymic development of tumor-associated regulatory T cells. Science 339, 1219–1224 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Perry, J. S. et al. Distinct contributions of Aire and antigen-presenting-cell subsets to the generation of self-tolerance in the thymus. Immunity 41, 414–426 (2014). References 28, 60 and 61 are recent studies that identify unique individual TCRs that are preferentially recruited into the T Reg cell lineage in the thymus by AIRE.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Guerau-de-Arellano, M., Martinic, M., Benoist, C. & Mathis, D. Neonatal tolerance revisited: a perinatal window for Aire control of autoimmunity. J. Exp. Med. 206, 1245–1252 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kuroda, N. et al. Development of autoimmunity against transcriptionally unrepressed target antigen in the thymus of Aire-deficient mice. J. Immunol. 174, 1862–1870 (2005).

    Article  CAS  PubMed  Google Scholar 

  64. Chen, Z., Benoist, C. & Mathis, D. How defects in central tolerance impinge on a deficiency in regulatory T cells. Proc. Natl Acad. Sci. USA 102, 14735–14740 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Hubert, F. X. et al. Aire regulates the transfer of antigen from mTECs to dendritic cells for induction of thymic tolerance. Blood 118, 2462–2472 (2011).

    Article  CAS  PubMed  Google Scholar 

  66. Niki, S. et al. Alteration of intra-pancreatic target-organ specificity by abrogation of Aire in NOD mice. J. Clin. Invest. 116, 1292–1301 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Laan, M. et al. Autoimmune regulator deficiency results in decreased expression of CCR4 and CCR7 ligands and in delayed migration of CD4+ thymocytes. J. Immunol. 183, 7682–7691 (2009).

    Article  CAS  PubMed  Google Scholar 

  68. Gray, D., Abramson, J., Benoist, C. & Mathis, D. Proliferative arrest and rapid turnover of thymic epithelial cells expressing Aire. J. Exp. Med. 204, 2521–2528 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kyewski, B. & Derbinski, J. Self-representation in the thymus: an extended view. Nat. Rev. Immunol. 4, 688–698 (2004).

    Article  CAS  PubMed  Google Scholar 

  70. Nishikawa, Y. et al. Temporal lineage tracing of Aire-expressing cells reveals a requirement for Aire in their maturation program. J. Immunol. 192, 2585–2592 (2014).

    Article  CAS  PubMed  Google Scholar 

  71. Wang, X. et al. Post-Aire maturation of thymic medullary epithelial cells involves selective expression of keratinocyte-specific autoantigens. Front. Immunol. 3, 19 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Matsumoto, M. Contrasting models for the roles of Aire in the differentiation program of epithelial cells in the thymic medulla. Eur. J. Immunol. 41, 12–17 (2011).

    Article  CAS  PubMed  Google Scholar 

  73. Yano, M. et al. Aire controls the differentiation program of thymic epithelial cells in the medulla for the establishment of self-tolerance. J. Exp. Med. 205, 2827–2838 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Trager, U. et al. The immune response to melanoma is limited by thymic selection of self-antigens. PLoS ONE 7, e35005 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhu, M. L., Nagavalli, A. & Su, M. A. Aire deficiency promotes TRP-1-specific immune rejection of melanoma. Cancer Res. 73, 2104–2116 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Gotter, J., Brors, B., Hergenhahn, M. & Kyewski, B. Medullary epithelial cells of the human thymus express a highly diverse selection of tissue-specific genes colocalized in chromosomal clusters. J. Exp. Med. 199, 155–166 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Conteduca, G. et al. The role of AIRE polymorphisms in melanoma. Clin. Immunol. 136, 96–104 (2010).

    Article  CAS  PubMed  Google Scholar 

  78. Akiyama, N. et al. Limitation of immune tolerance-inducing thymic epithelial cell development by Spi-B-mediated negative feedback regulation. J. Exp. Med. 211, 2425–2438 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Rossi, S. W. et al. RANK signals from CD4+3 inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla. J. Exp. Med. 204, 1267–1272 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Khan, I. S. et al. Enhancement of an anti-tumor immune response by transient blockade of central T cell tolerance. J. Exp. Med. 211, 761–768 (2014). This study demonstrates that in vivo blockade of RANK signalling can lead to selective depletion of AIRE-expressing mTECs and that this can be used to promote a break in tolerance to a melanoma tumour cell line.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Sharma, P. & Allison, J. P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205–214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Hobbs, R. P. et al. Keratin-dependent regulation of Aire and gene expression in skin tumor keratinocytes. Nat. Genet. 47, 933–938 (2015). In this paper, the authors demonstrate that the acquisition of AIRE expression in a skin tumour model may help to promote tumorigenesis and suggest a function for AIRE aside from thymic TSA expression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Dertschnig, S., Hauri-Hohl, M. M., Vollmer, M., Hollander, G. A. & Krenger, W. Impaired thymic expression of tissue-restricted antigens licenses the de novo generation of autoreactive CD4+ T cells in acute GVHD. Blood 125, 2720–2723 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Dertschnig, S. et al. Epithelial cytoprotection sustains ectopic expression of tissue-restricted antigens in the thymus during murine acute GVHD. Blood 122, 837–841 (2013).

    Article  CAS  PubMed  Google Scholar 

  85. Kisand, K. et al. Mucocutaneous candidiasis and autoimmunity against cytokines in APECED and thymoma patients: clinical and pathogenetic implications. Eur. J. Immunol. 41, 1517–1527 (2011).

    Article  CAS  PubMed  Google Scholar 

  86. Parent, A. V. et al. Generation of functional thymic epithelium from human embryonic stem cells that supports host T cell development. Cell Stem Cell 13, 219–229 (2013).

    Article  CAS  PubMed  Google Scholar 

  87. Sun, X. et al. Directed differentiation of human embryonic stem cells into thymic epithelial progenitor-like cells reconstitutes the thymic microenvironment in vivo. Cell Stem Cell 13, 230–236 (2013).

    Article  CAS  PubMed  Google Scholar 

  88. Doudna, J. A. & Charpentier, E. Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science 346, 1258096 (2014).

    Article  CAS  PubMed  Google Scholar 

  89. Takaba, H. et al. Fezf2 orchestrates a thymic program of self-antigen expression for immune tolerance. Cell 163, 975–987 (2015). This new study describes FEZ family zinc finger protein 2 (FEZF2) as a novel regulator of thymic TSA expression that complements the function of AIRE.

    Article  CAS  PubMed  Google Scholar 

  90. Pomie, C. et al. Autoimmune regulator (AIRE)-deficient CD8+CD28low regulatory T lymphocytes fail to control experimental colitis. Proc. Natl Acad. Sci. USA 108, 12437–12442 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Najafian, N. et al. Regulatory functions of CD8+CD28 T cells in an autoimmune disease model. J. Clin. Invest. 112, 1037–1048 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Menager-Marcq, I., Pomie, C., Romagnoli, P. & van Meerwijk, J. P. CD8+CD28 regulatory T lymphocytes prevent experimental inflammatory bowel disease in mice. Gastroenterology 131, 1775–1785 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the US National Institutes of Health to both M.S.A. and M.A.S.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Mark S. Anderson or Maureen A. Su.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Autoimmune polyglandular syndrome type 1

(APS1). A rare human autoimmune disorder that is inherited in an autosomal recessive manner and is characterized by various endocrine deficiencies, chronic mucocutaneous candidiasis and ectodermal dystrophies. It is caused by mutations in the gene that encodes autoimmune regulator (AIRE).

Co-immunoprecipitation

A protein purification experiment used to identify proteins that are in complex with each other.

Yeast two-hybrid screening

A screening system for protein–protein interactions that results in the transcription of a reporter gene when a bait protein attached to a DNA-binding domain comes into contact with a prey protein bound to a transcriptional activator.

RNA interference

(RNAi). A phenomenon in which the expression of a gene is inhibited when a double-stranded complementary RNA is introduced into the organism.

Silenced chromatin states

Regions of chromatin that are in a repressed or silenced state, such that genes in these regions are not expressed.

Morisita–Horn similarity index

A statistical algorithm that is used to determine the similarity of complex sequences, such as those seen in individual T cell receptors.

Graft-versus-host disease

(GVHD). A potentially serious complication arising when donor-derived T cells attack host tissues, typically resulting in hepatic, dermatological and gastrointestinal damage. Acute GVHD occurs within the first 100 days after transplantation, whereas chronic GVHD occurs later and has a different pathophysiology.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Anderson, M., Su, M. AIRE expands: new roles in immune tolerance and beyond. Nat Rev Immunol 16, 247–258 (2016). https://doi.org/10.1038/nri.2016.9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri.2016.9

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