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:

T cell–intrinsic ASC critically promotes TH17-mediated experimental autoimmune encephalomyelitis

Abstract

Interleukin 1β (IL-1β) is critical for the in vivo survival, expansion and effector function of IL-17–producing helper T (TH17) cells during autoimmune responses, including experimental autoimmune encephalomyelitis (EAE). However, the spatiotemporal role and cellular source of IL-1β during EAE pathogenesis are poorly defined. In the present study, we uncovered a T cell–intrinsic inflammasome that drives IL-1β production during TH17-mediated EAE pathogenesis. Activation of T cell antigen receptors induced expression of pro-IL-1β, whereas ATP stimulation triggered T cell production of IL-1β via ASC-NLRP3–dependent caspase-8 activation. IL-1R was detected on TH17 cells but not on type 1 helper T (TH1) cells, and ATP-treated TH17 cells showed enhanced survival compared with ATP-treated TH1 cells, suggesting autocrine action of TH17-derived IL-1β. Together these data reveal a critical role for IL-1β produced by a TH17 cell–intrinsic ASC–NLRP3–caspase-8 inflammasome during inflammation of the central nervous system.

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: Genetic deletion of the inflammasome adaptor ASC in T cells protects from EAE.
Figure 2: MOG35-55-reactive T effector cell priming in the secondary lymphoid organs is unaffected by ASC deficiency in T cells.
Figure 3: T cell–specific ASC deficiency is required for TH17 cell–mediated but not TH1 cell–mediated EAE.
Figure 4: TH17 cells process IL-1β in response to stimulation with extracellular ATP in an ASC–NLRP3–caspase-8–dependent manner.
Figure 5: TH17 cells express IL-1R and pro-IL-1β.
Figure 6: IL-1β is produced by CD4+ T cells in vivo.
Figure 7: T cell–intrinsic inflammasome activation is required for TH17 cell survival and proliferation in the CNS.

Similar content being viewed by others

References

  1. Frohman, E.M. Multiple sclerosis. Med. Clin. North Am. 87, 867–897 (2003).

    Article  PubMed  Google Scholar 

  2. Ransohoff, R.M. Animal models of multiple sclerosis: the good, the bad and the bottom line. Nat. Neurosci. 15, 1074–1077 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Stromnes, I.M., Cerretti, L.M., Liggitt, D., Harris, R.A. & Goverman, J.M. Differential regulation of central nervous system autoimmunity by TH1 and TH17 cells. Nat. Med. 14, 337–342 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kroenke, M.A., Carlson, T.J., Andjelkovic, A.V. & Segal, B.M. IL-12- and IL-23-modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition. J. Exp. Med. 205, 1535–1541 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lees, J.R., Golumbek, P.T., Sim, J., Dorsey, D. & Russell, J.H. Regional CNS responses to IFN-gamma determine lesion localization patterns during EAE pathogenesis. J. Exp. Med. 205, 2633–2642 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Jäger, A., Dardalhon, V., Sobel, R.A., Bettelli, E. & Kuchroo, V.K. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J. Immunol. 183, 7169–7177 (2009).

    Article  PubMed  CAS  Google Scholar 

  7. Cua, D.J. et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 421, 744–748 (2003).

    Article  CAS  PubMed  Google Scholar 

  8. Veldhoen, M., Hocking, R.J., Atkins, C.J., Locksley, R.M. & Stockinger, B. TGFβ 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 

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

    Article  CAS  PubMed  Google Scholar 

  10. Chung, Y. et al. Critical regulation of early Th17 cell differentiation by interleukin-1 signaling. Immunity 30, 576–587 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Gulen, M.F. et al. The receptor SIGIRR suppresses Th17 cell proliferation via inhibition of the interleukin-1 receptor pathway and mTOR kinase activation. Immunity 32, 54–66 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Staschke, K.A. et al. IRAK4 kinase activity is required for Th17 differentiation and Th17-mediated disease. J. Immunol. 183, 568–577 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Matsuki, T., Nakae, S., Sudo, K., Horai, R. & Iwakura, Y. Abnormal T cell activation caused by the imbalance of the IL-1/IL-1R antagonist system is responsible for the development of experimental autoimmune encephalomyelitis. Int. Immunol. 18, 399–407 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Gulen, M.F. et al. Inactivation of the enzyme GSK3α by the kinase IKKi promotes AKT-mTOR signaling pathway that mediates interleukin-1-induced Th17 cell maintenance. Immunity 37, 800–812 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chang, J. et al. MyD88 is essential to sustain mTOR activation necessary to promote T helper 17 cell proliferation by linking IL-1 and IL-23 signaling. Proc. Natl. Acad. Sci. USA 110, 2270–2275 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Gris, D. et al. NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses. J. Immunol. 185, 974–981 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Inoue, M., Williams, K.L., Gunn, M.D. & Shinohara, M.L. NLRP3 inflammasome induces chemotactic immune cell migration to the CNS in experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 109, 10480–10485 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Inoue, M. et al. Interferon-β therapy against EAE is effective only when development of the disease depends on the NLRP3 inflammasome. Sci. Signal. 5, ra38 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Hirota, K. et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Arbeloa, J., Perez-Samartin, A., Gottlieb, M. & Matute, C. P2X7 receptor blockade prevents ATP excitotoxicity in neurons and reduces brain damage after ischemia. Neurobiol. Dis. 45, 954–961 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Qu, Y. et al. Pannexin-1 is required for ATP release during apoptosis but not for inflammasome activation. J. Immunol. 186, 6553–6561 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Gordon, G.R. et al. Norepinephrine triggers release of glial ATP to increase postsynaptic efficacy. Nat. Neurosci. 8, 1078–1086 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Antonopoulos, C., El Sanadi, C., Kaiser, W.J., Mocarski, E.S. & Dubyak, G.R. Proapoptotic chemotherapeutic drugs induce noncanonical processing and release of IL-1β via caspase-8 in dendritic cells. J. Immunol. 191, 4789–4803 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Gringhuis, S.I. et al. Dectin-1 is an extracellular pathogen sensor for the induction and processing of IL-1β via a noncanonical caspase-8 inflammasome. Nat. Immunol. 13, 246–254 (2012).

    Article  CAS  PubMed  Google Scholar 

  25. Bossaller, L. et al. FAS (CD95) mediates noncanonical IL-1β and IL-18 maturation via caspase-8 in an RIP3-independent manner. J. Immunol. 189, 5508–5512 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Oberst, A. et al. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 471, 363–367 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Leverrier, S., Salvesen, G.S. & Walsh, C.M. Enzymatically active single chain caspase-8 maintains T-cell survival during clonal expansion. Cell Death Differ. 18, 90–98 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Bell, B.D. et al. FADD and caspase-8 control the outcome of autophagic signaling in proliferating T cells. Proc. Natl. Acad. Sci. USA 105, 16677–16682 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kang, T.B. et al. Caspase-8 serves both apoptotic and nonapoptotic roles. J. Immunol. 173, 2976–2984 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Salmena, L. et al. Essential role for caspase 8 in T-cell homeostasis and T-cell-mediated immunity. Genes Dev. 17, 883–895 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Chun, H.J. et al. Pleiotropic defects in lymphocyte activation caused by caspase-8 mutations lead to human immunodeficiency. Nature 419, 395–399 (2002).

    Article  CAS  PubMed  Google Scholar 

  32. Bauernfeind, F.G. et al. NF-κB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J. Immunol. 183, 787–791 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Martin, B.N. et al. IKKα negatively regulates ASC-dependent inflammasome activation. Nat. Commun. 5, 4977 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Bryan, N.B., Dorfleutner, A., Rojanasakul, Y. & Stehlik, C. Activation of inflammasomes requires intracellular redistribution of the apoptotic speck-like protein containing a caspase recruitment domain. J. Immunol. 182, 3173–3182 (2009).

    Article  CAS  PubMed  Google Scholar 

  35. Schenten, D. et al. Signaling through the adaptor molecule MyD88 in CD4+ T cells is required to overcome suppression by regulatory T cells. Immunity 40, 78–90 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Doitsh, G. et al. Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature 505, 509–514 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Shaw, P.J. et al. Critical role for PYCARD/ASC in the development of experimental autoimmune encephalomyelitis. J. Immunol. 184, 4610–4614 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. El-Behi, M. et al. The encephalitogenicity of TH17 cells is dependent on IL-1– and IL-23–induced production of the cytokine GM-CSF. Nat. Immunol. 12, 568–575 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Wei, D.G. et al. Expansion and long-range differentiation of the NKT cell lineage in mice expressing CD1d exclusively on cortical thymocytes. J. Exp. Med. 202, 239–248 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Shornick, L.P. et al. Mice deficient in IL-1β manifest impaired contact hypersensitivity to trinitrochlorobenzone. J. Exp. Med. 183, 1427–1436 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Tomalka, J. et al. A novel role for the NLRC4 inflammasome in mucosal defenses against the fungal pathogen Candida albicans. PLoS Pathog. 7, e1002379 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sarkar, A. et al. Caspase-1 regulates Escherichia coli sepsis and splenic B cell apoptosis independent of interleukin-1β and interleukin-18. Am. J. Respir. Crit. Care Med. 174, 1003–1010 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kuida, K. et al. Altered cytokine export and apoptosis in mice deficient in interleukin-1β converting enzyme. Science 267, 2000–2003 (1995).

    Article  CAS  PubMed  Google Scholar 

  44. Kaiser, W.J. et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 471, 368–372 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Qian, Y. et al. The adaptor Act1 is required for interleukin 17–dependent signaling associated with autoimmune and inflammatory disease. Nat. Immunol. 8, 247–256 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Kang, Z. et al. Astrocyte-restricted ablation of interleukin-17-induced Act1-mediated signaling ameliorates autoimmune encephalomyelitis. Immunity 32, 414–425 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Suzuki, S. et al. Shigella type III secretion protein MxiI is recognized by Naip2 to induce Nlrc4 inflammasome activation independently of Pkcδ. PLoS Pathog. 10, e1003926 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

This investigation was supported by the National Multiple Sclerosis Society (grant RG5130A2/1 to X.L.) and the US National Institutes of Health (grants 5R01NS071996-05, 1RO1AA023722 and MSTP-T32GM007250 to X.L.).

Author information

Authors and Affiliations

Authors

Contributions

B.N.M. and C.W. did the experiments and analyzed the data; C.Z., Z.K., M.F.G., J.A.Z., J.Z., C.E.-S. and G.B. contributed to the experiments; J.D. and P.G.P. helped to make the Cd3e−/− and Il1b−/− bone marrow chimera mice; A.A., Y.I., A.S., M.D.W., W.J.K., E.S.M., M.E.R., P.L.F. and A.G.H. provided reagents and participated in discussion; B.M. and G.R.D. contributed reagents, helped design experiments and helped edit the manuscript; B.N.M., C.W. and X.L. wrote the manuscript; R.M.R. and X.L. conceived the study, oversaw the experiments and analyzed the data.

Corresponding authors

Correspondence to Richard M Ransohoff or Xiaoxia Li.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation of the Pycard conditional knockout allele.

(a) The Pycard gene is localized on chromosome 7 and contains three coding exons. For generation of the conditional knockout allele, the entire open reading frame including all three exons was targeted for conditional deletion. After generation of the targeting vector (using Pcr4.0 backbone), C57BL/6 embryonic stem cells were injected for gene targeting. Southern hybridization was used to identify progeny bearing the recombinant allele. (b) CD4+ T Cells were sorted from Ascf/+Lck-Cre and Ascf/-Lck-Cre mice by flow cytometry, and mRNA expression of three different exons of ASC was measured by Real-time PCR analysis. Unpaired t test was used to analyze the data. Data are representative of three independent experiments (b). n=3 mice per group in each experiment. Error bars represent s.e.m. *P < 0.05.

Supplementary Figure 2 Numbers of T cells and B cells were similar in the lymph nodes, spleens and thymus between Ascf/+Lck-Cre and Ascf/–Lck-Cre mice.

(a) The total cell number of CD4+, CD8+ and B220+CD19+ cells were counted from lymph nodes and spleen of Ascf/+Lck-Cre and Ascf/-Lck-Cre mice. (b) Total cell number of CD4-CD8-, CD4+CD8-, CD4+ and CD8+ were counted from thymus of Ascf/+Lck-Cre and Ascf/-Lck-Cre mice. Data are representative of two independent experiments (a-b). n=5 mice per group in each experiment. Error bars represent s.e.m.

Supplementary Figure 3 The EAE phenotype of adoptive transfer of sorted CD4+ T cells from Ascf/+Lck-Cre and Ascf/–Lck-Cre mice.

(a) Absolute numbers of CD4+ IL-17A+ cells (left panel) and concentrations of IL-17A in the supernatant of cells (right panel) from the draining lymph nodes and spleens of Ascf/+Lck-Cre and Ascf/-Lck-Cre mice immunized with MOG35-55 and harvested 10 days after immunization, followed by ex vivo re-stimulation in the presence of MOG35-55 and IL-23. (b-d) Cells from lymph nodes of Ascf/+Lck-Cre and Ascf/-Lck-Cre mice 10 days after immunization with MOG35-55 were re-stimulated with MOG 35-55 in vitro in the presence of recombinant IL-23 for 5 days, sorted for CD4+ T cells by flow cytometry, and then transferred into CD45.1 congenic mice. (b) Graph represents the average clinical score after T cell transfer. (c) Absolute numbers of CNS-infiltrating CD45.2+ CD4+ cells were determined at the peak of disease. Brains and spinal cords were harvested together and CD45.2+CD4+ cells were stained with anti-CD45.2 and anti-CD4 antibodies, followed by flow cytometric analysis. (d) Absolute numbers of CNS-infiltrating cells were determined at the peak of disease. Brains and spinal cords were harvested together and mononuclear infiltrating cells were stained with anti-CD45, anti-CD4, anti-CD8, anti-F4/80, anti-Ly6C, anti-CD19 and anti-Ly6G antibodies, followed by flow cytometric analysis. Two-way ANOVA (b) and Unpaired t test (a, c and d) were used to analyzed the data. Data are representative of three independent experiments (a-d). n=5 mice per group in each experiment. Error bars represent s.e.m. *P < 0.05 (a, c and d). Error bars represent s.d. *P < 0.05 (b).

Supplementary Figure 4 Comparison of T cell and macrophage secretion of IL-1β.

Macrophages and TH17 cells were left untreated or treated with LPS for 4 hours (1ug/ul for macrophages) or LPS+ATP (5 mM, 30 mins) or ATP alone (5 mM, 8 hours). Cell pellet and supernatant were collected and subjected to western blotting for IL-1β and Actin. Data are representative of two independent experiments.

Supplementary Figure 5 Model for T cell intrinsic ASC function during CNS inflammation.

Supplementary Figure 6 The EAE phenotype of Cd3e−/−Il1b−/− chimera mice.

(a) Lethal irradiated WT mice were reconstituted with WT+Cd3e-/- bone marrow or Il1b-/-+Cd3e-/- bone marrow. 6 weeks after reconstitution, mice were immunized with MOG35-55 peptide and 200 ng pertussis toxin on days 1 and 4. Graph represents the average clinical score after active immunization. (b) Inflammatory gene expression in the lumbar spinal cord was assessed at the peak of disease. (c) Absolute numbers of CNS-infiltrating cells were determined at the peak of disease. Brains and spinal cords were harvested together and mononuclear infiltrating cells were stained with anti-CD45, anti-CD4, anti-CD8, anti-F4/80, anti-Ly6C and anti-Ly6G antibodies, followed by flow cytometric analysis. Two-way ANOVA (a) and Unpaired t test (b and c) were used to analyzed the data. Data are representative of three independent experiments (a-c). n=5 mice per group in each experiment. Error bars represent s.e.m. *P < 0.05.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Table 1 (PDF 1734 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Martin, B., Wang, C., Zhang, Cj. et al. T cell–intrinsic ASC critically promotes TH17-mediated experimental autoimmune encephalomyelitis. Nat Immunol 17, 583–592 (2016). https://doi.org/10.1038/ni.3389

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3389

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