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.

  • Article
  • Published:

Extrathymic expression of Aire controls the induction of effective TH17 cell-mediated immune response to Candida albicans

Abstract

Patients with loss of function in the gene encoding the master regulator of central tolerance AIRE suffer from a devastating disorder called autoimmune polyendocrine syndrome type 1 (APS-1), characterized by a spectrum of autoimmune diseases and severe mucocutaneous candidiasis. Although the key mechanisms underlying the development of autoimmunity in patients with APS-1 are well established, the underlying cause of the increased susceptibility to Candida albicans infection remains less understood. Here, we show that Aire+MHCII+ type 3 innate lymphoid cells (ILC3s) could sense, internalize and present C. albicans and had a critical role in the induction of Candida-specific T helper 17 (TH17) cell clones. Extrathymic Rorc-Cre-mediated deletion of Aire resulted in impaired generation of Candida-specific TH17 cells and subsequent overgrowth of C. albicans in the mucosal tissues. Collectively, our observations identify a previously unrecognized regulatory mechanism for effective defense responses against fungal infections.

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

Fig. 1: Aire+ ILC3s express C. albicans-sensing receptors.
Fig. 2: C. albicans induces transcriptional changes in ILC3 subsets.
Fig. 3: Aire+ ILC3s internalize C. albicans for antigen presentation.
Fig. 4: Aire+ ILC3s are essential for the generation of C. albicans-specific TH17 response.
Fig. 5: Mice lacking extrathymic expression of Aire have reduced survival after systemic challenge with live C. albicans.
Fig. 6: Extrathymic expression of Aire is critical for an effective TH17 response to C. albicans at mucosal sites.
Fig. 7: Aire+ ILC3s induce a prosurvival program in Candida-specific TH17 clones.

Similar content being viewed by others

References

  1. 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 

  2. Klein, L., Kyewski, B., Allen, P. M. & Hogquist, K. A. Positive and negative selection of the T cell repertoire: what thymocytes see (and don’t see). Nat. Rev. Immunol. 14, 377–391 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

  4. 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 

  5. Malchow, S. et al. Aire enforces immune tolerance by directing autoreactive T cells into the regulatory T cell lineage. Immunity 44, 1102–1113 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Abramson, J. & Husebye, E. S. Autoimmune regulator and self-tolerance: molecular and clinical aspects. Immunol. Rev. 271, 127–140 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Husebye, E. S., Anderson, M. S. & Kämpe, O. Autoimmune polyendocrine syndromes. N. Engl. J. Med. 378, 2543–2544 (2018).

    Article  PubMed  Google Scholar 

  8. Bruserud, Ø. et al. A longitudinal follow-up of autoimmune polyendocrine syndrome type 1. J. Clin. Endocrinol. Metab. 101, 2975–2983 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Perheentupa, J. Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy. J. Clin. Endocrinol. Metab. 91, 2843–2850 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Okada, S. et al. IMMUNODEFICIENCIES. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science 349, 606–613 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Puel, A. et al. Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin-17 immunity. Science 332, 65–68 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Milner, J. D. et al. Impaired T(H)17 cell differentiation in subjects with autosomal dominant hyper-IgE syndrome. Nature 452, 773–776 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ferwerda, B. et al. Human dectin-1 deficiency and mucocutaneous fungal infections. N. Engl. J. Med. 361, 1760–1767 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Glocker, E. O. et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N. Engl. J. Med. 361, 1727–1735 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liu, L. et al. Gain-of-function human STAT1 mutations impair IL-17 immunity and underlie chronic mucocutaneous candidiasis. J. Exp. Med. 208, 1635–1648 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Conti, H. R. & Gaffen, S. L. IL-17-mediated immunity to the opportunistic fungal pathogen Candida albicans. J. Immunol. 195, 780–788 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Kisand, K. et al. Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17-associated cytokines. J. Exp. Med. 207, 299–308 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Puel, A. et al. Autoantibodies against IL-17A, IL-17F, and IL-22 in patients with chronic mucocutaneous candidiasis and autoimmune polyendocrine syndrome type I. J. Exp. Med. 207, 291–297 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yamano, T. et al. Aire-expressing ILC3-like cells in the lymph node display potent APC features. J. Exp. Med. 216, 1027–1037 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Schlitzer, A. et al. IRF4 transcription factor-dependent CD11b+ dendritic cells in human and mouse control mucosal IL-17 cytokine responses. Immunity 38, 970–983 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jouault, T. et al. Candida albicans phospholipomannan is sensed through toll-like receptors. J. Infect. Dis. 188, 165–172 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Blasi, E. et al. Biological importance of the two Toll-like receptors, TLR2 and TLR4, in macrophage response to infection with Candida albicans. FEMS Immunol. Med. Microbiol. 44, 69–79 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Brown, G. D. et al. Dectin-1 mediates the biological effects of beta-glucans. J. Exp. Med. 197, 1119–1124 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Gantner, B. N., Simmons, R. M. & Underhill, D. M. Dectin-1 mediates macrophage recognition of Candida albicans yeast but not filaments. EMBO J. 24, 1277–1286 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kohatsu, L., Hsu, D. K., Jegalian, A. G., Liu, F. T. & Baum, L. G. Galectin-3 induces death of Candida species expressing specific beta-1,2-linked mannans. J. Immunol. 177, 4718–4726 (2006).

    Article  CAS  PubMed  Google Scholar 

  26. Jouault, T. et al. Specific recognition of Candida albicans by macrophages requires galectin-3 to discriminate Saccharomyces cerevisiae and needs association with TLR2 for signaling. J. Immunol. 177, 4679–4687 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Veldhoen, M., Hocking, R. J., Atkins, C. J., Locksley, R. M. & Stockinger, B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 24, 179–189 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Bettelli, E. et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235–238 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Mangan, P. R. et al. Transforming growth factor-beta induces development of the TH17 lineage. Nature 441, 231–234 (2006).

    Article  CAS  PubMed  Google Scholar 

  30. Dobeš, J. et al. A novel conditional Aire allele enables cell-specific ablation of the immune tolerance regulator Aire. Eur. J. Immunol. 48, 546–548 (2018).

    Article  PubMed  Google Scholar 

  31. Eberl, G. & Littman, D. R. Thymic origin of intestinal alphabeta T cells revealed by fate mapping of RORgammat+ cells. Science 305, 248–251 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Jiang, T. T. et al. Commensal fungi recapitulate the protective benefits of intestinal bacteria. Cell Host Microbe 22, 809–816.e804 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Shao, T. Y. et al. Commensal Candida albicans positively calibrates systemic Th17 immunological responses. Cell Host Microbe 25, 404–417.e406 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., Fouser, L. A. & Artis, D. CD4+ lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, 122–134 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Solis, N. V. & Filler, S. G. Mouse model of oropharyngeal candidiasis. Nat. Protoc. 7, 637–642 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Altieri, D. C. Survivin: the inconvenient IAP. Semin. Cell Dev. Biol. 39, 91–96 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. DiToro, D. et al. Insulin-like growth factors are key regulators of T helper 17 regulatory T cell balance in autoimmunity. Immunity 52, 650–667 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Sonnenberg, G. F. & Hepworth, M. R. Functional interactions between innate lymphoid cells and adaptive immunity. Nat. Rev. Immunol. 19, 599–613 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hepworth, M. R. et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal commensal bacteria. Nature 498, 113–117 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hepworth, M. R. et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal selection of commensal bacteria-specific CD4+ T cells. Science 348, 1031–1035 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Melo-Gonzalez, F. et al. Antigen-presenting ILC3 regulate T cell-dependent IgA responses to colonic mucosal bacteria. J. Exp. Med. 216, 728–742 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Oliphant, C. J. et al. MHCII-mediated dialog between group 2 innate lymphoid cells and CD4+ T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity 41, 283–295 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. von Burg, N. et al. Activated group 3 innate lymphoid cells promote T-cell-mediated immune responses. Proc. Natl Acad. Sci. U S A 111, 12835–12840 (2014).

    Article  Google Scholar 

  44. Kärner, J. et al. Anti-cytokine autoantibodies suggest pathogenetic links with autoimmune regulator deficiency in humans and mice. Clin. Exp. Immunol. 171, 263–272 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Dobeš, J. et al. Gastrointestinal autoimmunity associated with loss of central tolerance to enteric α-defensins. Gastroenterology 149, 139–150 (2015).

    Article  PubMed  Google Scholar 

  46. Gavanescu, I., Kessler, B., Ploegh, H., Benoist, C. & Mathis, D. Loss of Aire-dependent thymic expression of a peripheral tissue antigen renders it a target of autoimmunity. Proc. Natl Acad. Sci. U S A 104, 4583–4587 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ma, C. S. et al. Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. J. Exp. Med. 205, 1551–1557 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Minegishi, Y. et al. Human tyrosine kinase 2 deficiency reveals its requisite roles in multiple cytokine signals involved in innate and acquired immunity. Immunity 25, 745–755 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Break, T. J. et al. Aberrant type 1 immunity drives susceptibility to mucosal fungal infections. Science 371, eaay5731 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wang, J. et al. Single-cell multiomics defines tolerogenic extrathymic Aire-expressing populations with unique homology to thymic epithelium. Sci. Immunol. 6, eabl5053 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lyu, M. et al. ILC3s select for RORγt+ Tregs and establish tolerance to intestinal microbiota. Preprint at bioRxiv, https://doi.org/10.1101/2022.04.25.489463 (2022).

  52. Jiang, W., Anderson, M. S., Bronson, R., Mathis, D. & Benoist, C. Modifier loci condition autoimmunity provoked by Aire deficiency. J. Exp. Med. 202, 805–815 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gardner, J. M. et al. Deletional tolerance mediated by extrathymic Aire-expressing cells. Science 321, 843–847 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Taylor, P. R. et al. Dectin-1 is required for beta-glucan recognition and control of fungal infection. Nat. Immunol. 8, 31–38 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Gordon, J. et al. Specific expression of lacZ and cre recombinase in fetal thymic epithelial cells by multiplex gene targeting at the Foxn1 locus. BMC Dev. Biol. 7, 69 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Colnot, C., Fowlis, D., Ripoche, M. A., Bouchaert, I. & Poirier, F. Embryonic implantation in galectin 1/galectin 3 double mutant mice. Dev. Dyn. 211, 306–313 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Barnden, M. J., Allison, J., Heath, W. R. & Carbone, F. R. Defective TCR expression in transgenic mice constructed using cDNA-based alpha- and beta-chain genes under the control of heterologous regulatory elements. Immunol. Cell Biol. 76, 34–40 (1998).

    Article  CAS  PubMed  Google Scholar 

  58. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    Article  CAS  PubMed  Google Scholar 

  59. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  60. Fonzi, W. A. & Irwin, M. Y. Isogenic strain construction and gene mapping in Candida albicans. Genetics 134, 717–728 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Igyártó, B. Z. et al. Skin-resident murine dendritic cell subsets promote distinct and opposing antigen-specific T helper cell responses. Immunity 35, 260–272 (2011).

    Article  PubMed  Google Scholar 

  62. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Kohen, R. et al. UTAP: user-friendly transcriptome analysis pipeline. BMC Bioinf. 20, 154 (2019).

    Article  Google Scholar 

  64. Moon, J. J. et al. Naive CD4+ T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 27, 203–213 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Pfaffl, M. W. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 29, e45 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Research in the Abramson laboratory is kindly supported by the European Research Council (ERC-2016-CoG-724821); Israel Science Foundation (1796/16 and 1819/21); Chan Zuckerberg Initiative; Bill and Marika Glied and Family Fund, Binational Science Foundation; Pasteur-Weizmann Delegation; Enoch Foundation, Ruth and Samuel David Gameroff Family Foundation; and Erica Drake Fund and Lilly Fulop Fund for Multiple Sclerosis Research. J.D. was supported by the Dean of Faculty Fellowship by Weizmann Institute of Science and by the Weizmann Institute of Science – Czech Academy of Sciences Bilateral Fellowship by Czech Academy of Sciences. J.D., K.K., H.B. and E.V. are also supported by Charles University PRIMUS grant (Primus/21/MED/003) and Czech Science Foundation JUNIOR STAR grant (21-22435M). B.E.O. and E.S.H. are supported by the K.G. Jebsen Center for autoimmune disorders, the Norwegian Research Council, the Novo Nordisk Foundation and the Regional Health Authorities of Western Norway. D.F. is supported by the Grant Agency of the Czech Republic (17-25365S).

Author information

Authors and Affiliations

Authors

Contributions

J.A. and J.D. conceived the project, designed the experiments and wrote the manuscript; J.D. performed the experiments and analyzed the data; J.A. supervised the study; A.B., Y. Goldfarb, O.B.-N., N.K., Y. Gruper, T.G., I.Z., H.B., K.K., E.V., B.E.O. and E.S.H. assisted at different aspects of the study, including data or sample acquisition and analysis. D.F. contributed by the essential reagent. L.S.B. and Z.S. performed the two-photon microscopy and analyzed the data.

Corresponding author

Correspondence to Jakub Abramson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1

ac, GO enrichment analysis of upregulated differentially expressed genes from unstimulated vs. C. albicans- stimulated ILC3 subsets including cILC3s (related to Fig. 2a) (a); MHCII+ ILC3s (related to Fig. 2b) (b); Aire+ ILC3s (related to Fig. 2c) (c). d, Heatmap of Pearson correlation according to gene expression values between individual samples analyzed in Fig. 2a-c.

Extended Data Fig. 2

a, Imaging flow cytometry analyzing the physical interaction between HKCA with DCs or with Aire+ ILC3s. CD11c+ DCs or Lineage negative cells were isolated from mouse pLNs using MACS-beads depletion and then incubated with CPD-stained HKCA for 90 minutes. Samples were stained for Aire, MHCII, CD11c, Lamp1 (lysosomal marker) and DAPI and analyzed by Imaging flow cytometry. In both, Aire+ ILC3 cells (Aire+) and DCs (CD11c+) HKCA associate with lysosomes (Lamp1). Representative images out of five independent repetitions of the experiment are shown. b, Comparison of the capacity of different immune subsets (B cells, Ly6C+ MNPs, CD11b+ MNPs, CD11c+ MNPs and Aire+ ILC3s) to endocytose CPD-labeled HKCA in an in vitro endocytosis assay. Subsets were FACS-sorted and incubated with HKCA-CPD for three hours. The frequency of cells that have successfully internalized HKCA-CPD was then assessed by flow cytometry (mean ± SD; n = 3). c,d, Comparison of the capacity of either wild-type or Galectin-3/Dectin-1-double knockout (dKO) Aire+ ILC3s (c) or DCs (d) to endocytose CPD-labeled HKCA at 37 °C or at 4 °C. Corresponding populations were isolated from WT or dKO mice and kept either at 37 °C or at 4 °C. The internalization of HKCA-CPD by the cells was measured by FACS (mean ± SD; n = 3). e, Representative gating strategy of APC populations related to Fig. 3c–g. f, FACS analysis of in vitro assay related to Fig. 3c. g, Graphical summary of an experimental setting relevant for data shown in h. Wild-type mice were intravenously stimulated by HKCA in specified timepoints and APC population were isolated using gating strategy depicted in e. h, FACS analysis of in vitro assay related to Fig. 3d–f assessing the antigen presentation of the corresponding cell subsets.

Extended Data Fig. 3

a, FACS analysis validating that Aire protein expression in the entire LN-resident cell compartment is exclusively restricted to a CD45 positive, lineage negative (Lin 1: TCR-β, CD19, Gr1, F4/80, CD11b, CD11c, NK1.1; Lin 2: CD3, B220), NKp46-negative, MHCII positive, Rorγt positive cell subset – previously identified as Aire+ ILC3s. b, Back-gating FACS analysis of Aire-expressing cells identified in a. c, Representative flow cytometry dot plots showing the frequencies of transferred CD45.1+ OT-II T cell (red gate) vs. CD45.1/CD45.2 double positive control T cell (blue gate) populations 2 days after the transfer. d, Statistical analysis of ratios related to b (n = 5 per group, mean ± SD, two-tailed Student’s t-test) of OT-II vs. control T two days after the transfer. e,f, Survival curves of WT (Aire+/+) and knockout (Aire–/–) mice (n ≥ 10 mice per group) on either on NOD (e) and C57Bl/6 (f) genetic background after systemic challenge with live C. albicans. Mice were i.v. injected every second day by heat-killed C. albicans (HKCA) for the duration of three weeks. Subsequently, mice were infected by alive C. albicans and monitored for survival. Long-rank (Mantel-Cox) test was used to calculate the indicated P-value. g,h, Quantitative PCR analysis assessing the presence of C. albicans-specific DNA in the liver (g) and small intestine (h) from Rorc-Cre Airefl/fl (WT) and Rorc-Cre+Airefl/fl (ILC3ΔAire) mice (n = 6, mean ± SD, two-tailed Student’s t-test). i,j, ELISA assessing the amount of IL-17 (i) or (IL-22) (j) autoantibodies in the sera of 8-week old untreated Aire+/+ vs. Aire–/– mice on NOD and C57Bl/6 (B6) genetic background (n = 6 per group, two-tailed Student’s t-test). Data are shown as mean of optical density ± SD. Data are shown as mean of optical density ± SD. P-value indicators: *** = P-value < 0.0001, ** = P-value < 0.001, ns = not significant.

Extended Data Fig. 4

a, Experimental outline relevant to data shown in Fig. 6a, b: Wild-type mice were orally colonized by C. albicans and analyzed in indicated time points. b, Representative FACS gating strategy of Als1-Tet+ T cells. c, Experimental outline relevant to data shown in (d) and (e) WT (Aire+/+) and Aire−/− were orally colonized by C. albicans and analyzed after two weeks. d, Representative FACS plot of Als1-Tet+ T cells. Cells were isolated from pLNs and spleens of mice described in (c). Counts of Als1-Tet+ cells are highlighted in red rectangles (left panel). Statistical analysis of the same representative experiment showing the total counts (mean ± SD, two-tailed Student’s t-test, n = 6). Representative experiment is shown. e, Quantitative PCR analysis assessing the presence of C. albicans-specific DNA in the ileal part of small intestine from WT and Aire-deficient mice (n = 5, mean ± SD, two-tailed Student’s t-test). f, Experimental outline relevant to data shown in (g) and (h). Bone marrow (BM) chimeras restricting Aire expression either to hematopoietic (Aire+/+ BM→Aire–/–) or stromal compartment (Aire–/– BM→Aire+/+) were generated by reciprocal BM transfer to recipient mice after 900 rad whole-body irradiation. Six weeks after the BM transfer, the mice were orally colonized by C. albicans and analyzed after two weeks. g, Representative FACS plot of Als1-Tet+ T cells. Counts of Als1-Tet+ cells are highlighted in red rectangles (left panel). Statistical analysis of the same representative experiment showing the total counts (n = 6, mean ± SD, two-tailed Student’s t-test). Representative experiment is shown. h, Quantitative PCR analysis assessing the presence of C. albicans-specific DNA in the ileal part of small intestine from reciprocal bone marrow chimeras (n = 6, mean ± SD, two-tailed Student’s t-test). i, Experimental outline relevant to data shown in j and k. CD90-disperate chimeras were created by adoptive transfer of T cells and B-lymphocytes from CD90.1 mice to Rag1−/− (CD90.2) recipients and let to proliferate for 2 months. Mice were treated by anti-CD90.2 or isotype control antibody prior the C. albicans oral colonization and then each third day and analyzed after two weeks. j, Representative FACS plot of Als1-Tet+ T cells. Counts of tetramer positive cells are highlighted in red rectangles (left panel). Statistical analysis of the same representative experiment showing the total counts (n = 6, mean ± SD, two-tailed Student’s t-test). Representative experiment is shown. k, Quantitative PCR analysis assessing the presence of C. albicans-specific DNA in the ileal part of small intestine from CD90-disparate chimeras (n = 6, mean ± SD, two-tailed Student’s t-test). P-value indicators: *** = P-value < 0.0001, ** = P-value < 0.001, * = P-value < 0.05, ns = not significant.

Extended Data Fig. 5

ae, Flow cytometry analysis of C albicans-specific T cells (using Als1-Tet) after oral colonization. Aire whole-body knockout mice (Aire−/−), their wild-type littermates (Aire+/+), WT (Rorc-CreAirefl/fl) and ILC3ΔAire (Rorc-Cre+Airefl/fl) were orally colonized by C. albicans and analyzed after 24 hours (left panel) or two weeks (right panel). a, Representative FACS plot of Als1-Tet+ T cells. Cells were isolated from pLNs and spleens (SLO), oral mucosa, esophagus and intestine of mice described above 24 hours (left panel) or 2 weeks (right panel) post C. albicans colonization. Counts of Als1-Tet+ cells are highlighted in red rectangles. be, Statistical analysis of the Als1-Tet+ cells counts (n = 6, mean ± SD, two-tailed Student’s t-test in SLO (b), oral mucosa (c), esophagus (d) and small intestine (e) 24 hours (left panel) or 2 weeks (right panel) post C. albicans colonization (n = 6, mean ± SD, two-tailed Student’s t-test). P-value indicators: *** = P-value < 0.0001, ** = P-value < 0.001, * = P-value < 0.05, ns = not significant.

Extended Data Fig. 6

a, Representative FACS gating strategy of Roryt+ CD4+ TCR-β+ TH17 cells in pLNs two weeks after C. albicans colonization of Rorc-Cre Airefl/fl (WT) and Rorc-Cre+Airefl/fl (ILC3ΔAire) mice. b, Statistical analysis of related to a). The plot is showing the frequency from parent gate (n = 6, mean ± SD, two-tailed Student’s t-test). c, Representative FACS gating strategy of Roryt+ CD4+ TCR-β+ TH17 cells in mesenteric lymph nodes (mLN) two weeks after C. albicans colonization of Rorc-Cre Airefl/fl (WT) and Rorc-Cre+Airefl/fl (ILC3ΔAire) mice. d, Statistical analysis of related to c). The plot is showing the frequency from parent gate (n = 6, mean ± SD, two-tailed Student’s t-test). e, Representative FACS gating strategy of Roryt+ CD4+ TCR-β+ TH17 cells in lamina propria two weeks after C. albicans colonization of Rorc-Cre Airefl/fl (WT) and Rorc-Cre+Airefl/fl (ILC3ΔAire) mice. f), Statistical analysis of related to e). The plot is showing the frequency from parent gate (n = 6, mean ± SD, two-tailed Student’s t-test). P-value indicators: ** =P-value < 0.001, * = P-value < 0.05, ns = not significant.

Extended Data Fig. 7

ad, Colony forming units (CFU)-based assay determining the overgrowth of C albicans 1 day (left panel) and 14 days (right panel) after the oral colonization. The tissues were isolated from Aire whole-body knockout mice (Aire−/−), their wild-type littermates (Aire+/+), Rorc-Cre Airefl/fl (WT) and Rorc-Cre+Airefl/fl (ILC3ΔAire) mice. CFU was determined by plating the lysates obtained from the kidney (a), oral cavity (b), esophagus (c), small intestine (d) (n = 6, mean ± SD, two-tailed Student’s t-test). P-value indicators: *** = P-value < 0.0001, ** = P-value < 0.001, * = P-value < 0.05, ns = not significant.

Extended Data Fig. 8

a, Experimental outline relevant to data shown in bc. Rorc-Cre Airefl/fl (WT) and Rorc-Cre+Airefl/fl (ILC3ΔAire) were first primed by repeated injections of heat-killed C. albicans (HKCA) for two weeks, then exposed to prolonged protocol of oropharyngeal candidiasis and analyzed five days later. b, Representative FACS plot of Als1-Tet+ T cells. Cells were isolated from pLNs and spleens of mice described in a. Counts of Als1-Tet+ cells are highlighted in red rectangles (left panel). Statistical analysis of the same representative experiment showing the total counts of Als1-Tet+ cells (n = 6, mean ± SD, two-tailed Student’s t-test). Representative experiment is shown. c, Representative FACS plot of Als1-Tet+ T cells. Cells were isolated from tongue mucosae of mice described in a. Counts of Als1-Tet+ cells are highlighted in red rectangles (left panel). Statistical analysis of the same representative experiment showing the total counts of Als1-Tet+ cells (n = 6, mean ± SD, two-tailed Student’s t-test). Representative experiment is shown.

Extended Data Fig. 9

a, Representative FACS-sorting strategy of Aire+ ILC3s from reporter AireGFP mice. The plots are showing the frequency from parent gates. Lineage: CD3, CD19, CD11c, CD11b, N.K.1, F4/80, Gr1, B220. b, Back-gating of AireGFP+ cells.

Extended Data Fig. 10

a, Experimental outline relevant to data shown in b and c. WT (Rorc-CreAirefl/fl) and ILC3ΔAire (Rorc-Cre+Airefl/fl) mice were transferred with naïve OT-II CD4+ T cells and subsequently injected with HKCA-OVA four times during a single week. b, Statistical analysis of the frequencies of OT-II T cells subtypes (n = 4, mean ± SD, two-tailed Student’s t-test). c, GO enrichment analysis of upregulated differentially expressed genes from RorcGFP+ OT-II T cells vs Non-proliferating OT-II T cells comparison. d, Graphical model summarizing the role of Aire+ ILC3s in the induction of Candida-specific TH17 response. While the immediate immune response to C. albicans infection is dominated by neutrophils, monocytes and macrophages, Aire+ ILC3s become essential in the later phase, as they facilitate clonal expansion of the primed TH17 cell clones in the LN. The expanded candida-specific TH17 clones subsequently limit C. albicans overgrowth at mucosal surfaces and its dissemination into epithelial tissues; e, Scheme illustrating the putative mechanism through which Aire+ ILC3 cells induce the expansion of candida-specific TH17 clones.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dobeš, J., Ben-Nun, O., Binyamin, A. et al. Extrathymic expression of Aire controls the induction of effective TH17 cell-mediated immune response to Candida albicans. Nat Immunol 23, 1098–1108 (2022). https://doi.org/10.1038/s41590-022-01247-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01247-6

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