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-bet-dependent NKp46+ innate lymphoid cells regulate the onset of TH17-induced neuroinflammation

An Author Correction to this article was published on 29 June 2018

Abstract

The transcription factor T-bet has been associated with increased susceptibility to systemic and organ-specific autoimmunity, but the mechanism by which T-bet expression promotes neuroinflammation remains unknown. In this study, we demonstrate a cardinal role of T-bet-dependent NKp46+ innate lymphoid cells (ILCs) in the initiation of CD4+ TH17-mediated neuroinflammation. Loss of T-bet specifically in NKp46+ ILCs profoundly impaired the ability of myelin-reactive TH17 cells to invade central nervous system (CNS) tissue and protected the mice from autoimmunity. T-bet-dependent NKp46+ ILCs localized in the meninges and acted as chief coordinators of meningeal inflammation by inducing the expression of proinflammatory cytokines, chemokines and matrix metalloproteinases, which together facilitated T cell entry into CNS parenchyma. Our findings uncover a detrimental role of T-bet-dependent NKp46+ ILCs in the development of CNS autoimmune disease.

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: T-bet expression in T cells is insufficient to cause EAE.
Figure 2: Peripheral licensing of CD4+ TH17 cells in Tbx21−/− host.
Figure 3: T-bet-expressing cells in the CNS during EAE.
Figure 4: T-bet-dependent NKp46+ ILCs initiate TH17-mediated EAE.
Figure 5: Localization of T-bet-dependent NKp46+ ILCs during EAE.
Figure 6: Pathogenic function of T-bet maps to ILC1/NKp46+ ILC3.
Figure 7: NKp46+ ILCs regulate infiltration of CD4+ TH17 cells.
Figure 8: NKp46+ ILCs regulate the stability of CD4+ TH17 cells.

Similar content being viewed by others

References

  1. McFarland, H.F. & Martin, R. Multiple sclerosis: a complicated picture of autoimmunity. Nat. Immunol. 8, 913–919 (2007).

    Article  CAS  Google Scholar 

  2. Park, H. et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol. 6, 1133–1141 (2005).

    Article  CAS  Google Scholar 

  3. Langrish, C.L. et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med. 201, 233–240 (2005).

    Article  CAS  Google Scholar 

  4. McGeachy, M.J. et al. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat. Immunol. 10, 314–324 (2009).

    Article  CAS  Google Scholar 

  5. 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  Google Scholar 

  6. Schläger, C. et al. Effector T-cell trafficking between the leptomeninges and the cerebrospinal fluid. Nature 530, 349–353 (2016).

    Article  Google Scholar 

  7. Bettelli, E. et al. Loss of T-bet, but not STAT1, prevents the development of experimental autoimmune encephalomyelitis. J. Exp. Med. 200, 79–87 (2004).

    Article  CAS  Google Scholar 

  8. Lovett-Racke, A.E. et al. Silencing T-bet defines a critical role in the differentiation of autoreactive T lymphocytes. Immunity 21, 719–731 (2004).

    Article  CAS  Google Scholar 

  9. Lazarevic, V., Glimcher, L.H. & Lord, G.M. T-bet: a bridge between innate and adaptive immunity. Nat. Rev. Immunol. 13, 777–789 (2013).

    Article  CAS  Google Scholar 

  10. Lazarevic, V. & Glimcher, L.H. T-bet in disease. Nat. Immunol. 12, 597–606 (2011).

    Article  CAS  Google Scholar 

  11. Lee, Y. et al. Induction and molecular signature of pathogenic TH17 cells. Nat. Immunol. 13, 991–999 (2012).

    Article  CAS  Google Scholar 

  12. Wang, Y. et al. The transcription factors T-bet and Runx are required for the ontogeny of pathogenic interferon-γ-producing T helper 17 cells. Immunity 40, 355–366 (2014).

    Article  CAS  Google Scholar 

  13. Yang, Y. et al. T-bet is essential for encephalitogenicity of both Th1 and Th17 cells. J. Exp. Med. 206, 1549–1564 (2009).

    Article  CAS  Google Scholar 

  14. Ghoreschi, K. et al. Generation of pathogenic TH17 cells in the absence of TGF-β signalling. Nature 467, 967–971 (2010).

    Article  CAS  Google Scholar 

  15. Odoardi, F. et al. T cells become licensed in the lung to enter the central nervous system. Nature 488, 675–679 (2012).

    Article  CAS  Google Scholar 

  16. Flügel, A. et al. Migratory activity and functional changes of green fluorescent effector cells before and during experimental autoimmune encephalomyelitis. Immunity 14, 547–560 (2001).

    Article  Google Scholar 

  17. Zhu, J. et al. The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses. Immunity 37, 660–673 (2012).

    Article  CAS  Google Scholar 

  18. Greter, M. et al. Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat. Med. 11, 328–334 (2005).

    Article  CAS  Google Scholar 

  19. Kivisäkk, P. et al. Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann. Neurol. 65, 457–469 (2009).

    Article  Google Scholar 

  20. McMahon, E.J., Bailey, S.L., Castenada, C.V., Waldner, H. & Miller, S.D. Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat. Med. 11, 335–339 (2005).

    Article  CAS  Google Scholar 

  21. Fuchs, A. et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells. Immunity 38, 769–781 (2013).

    Article  CAS  Google Scholar 

  22. Gordon, S.M. et al. The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation. Immunity 36, 55–67 (2012).

    Article  CAS  Google Scholar 

  23. Klose, C.S.N. et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).

    Article  CAS  Google Scholar 

  24. Klose, C.S. et al. A T-bet gradient controls the fate and function of CCR6-RORγt+ innate lymphoid cells. Nature 494, 261–265 (2013).

    Article  CAS  Google Scholar 

  25. Rankin, L.C. et al. The transcription factor T-bet is essential for the development of NKp46+ innate lymphocytes via the Notch pathway. Nat. Immunol. 14, 389–395 (2013).

    Article  CAS  Google Scholar 

  26. Sciumé, G. et al. Distinct requirements for T-bet in gut innate lymphoid cells. J. Exp. Med. 209, 2331–2338 (2012).

    Article  Google Scholar 

  27. Townsend, M.J. et al. T-bet regulates the terminal maturation and homeostasis of NK and Valpha14i NKT cells. Immunity 20, 477–494 (2004).

    Article  CAS  Google Scholar 

  28. Morandi, B. et al. Role of natural killer cells in the pathogenesis and progression of multiple sclerosis. Pharmacol. Res. 57, 1–5 (2008).

    Article  CAS  Google Scholar 

  29. Winkler-Pickett, R. et al. In vivo regulation of experimental autoimmune encephalomyelitis by NK cells: alteration of primary adaptive responses. J. Immunol. 180, 4495–4506 (2008).

    Article  CAS  Google Scholar 

  30. Liu, Q. et al. Neural stem cells sustain natural killer cells that dictate recovery from brain inflammation. Nat. Neurosci. 19, 243–252 (2016).

    Article  CAS  Google Scholar 

  31. Bielekova, B. et al. Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis. Proc. Natl. Acad. Sci. USA 103, 5941–5946 (2006).

    Article  CAS  Google Scholar 

  32. Hao, J. et al. Central nervous system (CNS)-resident natural killer cells suppress Th17 responses and CNS autoimmune pathology. J. Exp. Med. 207, 1907–1921 (2010).

    Article  CAS  Google Scholar 

  33. Pikovskaya, O. et al. Cutting edge: eomesodermin is sufficient to direct type 1 innate lymphocyte development into the conventional NK lineage. J. Immunol. 196, 1449–1454 (2016).

    Article  CAS  Google Scholar 

  34. Becher, B., Spath, S. & Goverman, J. Cytokine networks in neuroinflammation. Nat. Rev. Immunol. 17, 49–59 (2017).

    Article  CAS  Google Scholar 

  35. Rothhammer, V. et al. Th17 lymphocytes traffic to the central nervous system independently of α4 integrin expression during EAE. J. Exp. Med. 208, 2465–2476 (2011).

    Article  CAS  Google Scholar 

  36. Reboldi, A. et al. C-C chemokine receptor 6–regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat. Immunol. 10, 514–523 (2009).

    Article  CAS  Google Scholar 

  37. Bauer, M. et al. Beta1 integrins differentially control extravasation of inflammatory cell subsets into the CNS during autoimmunity. Proc. Natl. Acad. Sci. USA 106, 1920–1925 (2009).

    Article  CAS  Google Scholar 

  38. Dungan, L.S., McGuinness, N.C., Boon, L., Lynch, M.A. & Mills, K.H. Innate IFN-γ promotes development of experimental autoimmune encephalomyelitis: a role for NK cells and M1 macrophages. Eur. J. Immunol. 44, 2903–2917 (2014).

    Article  CAS  Google Scholar 

  39. Kipnis, J. Multifaceted interactions between adaptive immunity and the central nervous system. Science 353, 766–771 (2016).

    Article  CAS  Google Scholar 

  40. Song, J. et al. Focal MMP-2 and MMP-9 activity at the blood-brain barrier promotes chemokine-induced leukocyte migration. Cell Rep. 10, 1040–1054 (2015).

    Article  CAS  Google Scholar 

  41. Chiang, E.Y. et al. Targeted depletion of lymphotoxin-α–expressing TH1 and TH17 cells inhibits autoimmune disease. Nat. Med. 15, 766–773 (2009).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  43. Pikor, N.B. et al. Integration of Th17- and lymphotoxin-derived signals initiates meningeal-resident stromal cell remodeling to propagate neuroinflammation. Immunity 43, 1160–1173 (2015).

    Article  CAS  Google Scholar 

  44. Codarri, L. et al. RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat. Immunol. 12, 560–567 (2011).

    Article  CAS  Google Scholar 

  45. 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  Google Scholar 

  46. Jenne, C.N. et al. T-bet-dependent S1P5 expression in NK cells promotes egress from lymph nodes and bone marrow. J. Exp. Med. 206, 2469–2481 (2009).

    Article  CAS  Google Scholar 

  47. van Helden, M.J. et al. Terminal NK cell maturation is controlled by concerted actions of T-bet and Zeb2 and is essential for melanoma rejection. J. Exp. Med. 212, 2015–2025 (2015).

    Article  CAS  Google Scholar 

  48. Narni-Mancinelli, E. et al. Fate mapping analysis of lymphoid cells expressing the NKp46 cell surface receptor. Proc. Natl. Acad. Sci. USA 108, 18324–18329 (2011).

    Article  CAS  Google Scholar 

  49. Intlekofer, A.M. et al. Anomalous type 17 response to viral infection by CD8+ T cells lacking T-bet and eomesodermin. Science 321, 408–411 (2008).

    Article  CAS  Google Scholar 

  50. Gossa, S., Nayak, D., Zinselmeyer, B.H. & McGavern, D.B. Development of an immunologically tolerated combination of fluorescent proteins for in vivo two-photon imaging. Sci. Rep. 4, 6664 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Y. Belkaid and A. Singer for critical review of the manuscript; S. Sharrow, L. Granger and A. Adams for flow cytometry and cell sorting; T. Loo for technical assistance; and the members of the laboratories of A. Singer and H. Park for discussions. S. Reiner (Columbia University) generously provided the Tbx21f/f mice. This study was supported by the Intramural Research Program of the US National Institutes of Health, National Cancer Institute, Center for Cancer Research (ZIA BC 011431 and ZIA BC 011432 to V.L.) and National Institute of Neurological Disorders and Stroke (1ZIA NS 003111-08 and 1ZIA NS 003112-08 to D.B.M.).

Author information

Authors and Affiliations

Authors

Contributions

B.K. conducted the experiments, analyzed the data and contributed to the first draft of the manuscript. R.R., Y.G., J.F. and Y.W. conducted experiments and analyzed data. D.B.M. contributed to experimental design and manuscript preparation. M.J.K. performed quantification of apoptotic cells in the CNS in a blinded manner. E.V. and J.Z. generated NKp46-Cre+ and T-bet ZsGreen reporter mice, respectively. V.L. designed and directed the study, conducted the experiments, analyzed the data and wrote the manuscript.

Corresponding author

Correspondence to Vanja Lazarevic.

Ethics declarations

Competing interests

E.V. is the cofounder of and a shareholder in Innate Pharma.

Integrated supplementary information

Supplementary Figure 1 T-bet expression in endogenous (host-derived) T cells is not required for EAE induction by 2D2 WT TH17 cells.

(a) Flow cytometric analysis of CNS-infiltrating CD4+ T cells following adoptive transfer of 7.5 × 106 2D2 WT TH17 cells into Tbx21f/f or Tbx21f/f CD4-Cre+ recipient mice. CNS-infiltrating cells were harvested at the peak of EAE disease (day 16 post-transfer) and analyzed for the fraction of 2D2 (Vβ11+) versus endogenous (Vβ11) cells among live CD4+ T cells. (b-c) Cytokine production of CNS-infiltrating endogenous or 2D2 CD4+ T cells at the peak of EAE disease was determined by intracellular cytokine staining following brief ex vivo PMA/ionomycin re-stimulation. (d) Mean clinical scores in Tbx21f/f or Tbx21f/f CD4-Cre+ mice receiving 2D2 WT TH17 cells (7.5 × 106). NS, P > 0.05; *, P < 0.01 (two-tailed Student’s t-test (b,c) or two-way ANOVA (d)). Data are combined from two independent experiments (a-c; n = 9, 10 mice per group) or three independent experiments (d; n = 15, 16 mice per group).

Supplementary Figure 2 T-bet expression in dendritic cells is not required for EAE induction by 2D2 WT TH17 cells.

(a) Intracellular cytokine staining of CNS-infiltrating 2D2 CD4+ T cells (CD4+Vβ11+) from Tbx21f/f or Tbx21f/f CD11c-Cre+ mice following adoptive transfer of 7.5 × 106 2D2 WT TH17 cells. CNS-infiltrating cells were harvested at the peak of EAE disease (day 15-17 post-transfer) and re-stimulated ex vivo with PMA/ionomycin. (b) Mean clinical scores in Tbx21f/f or Tbx21f/f CD11c-Cre+ mice which received 2D2 WT TH17 cells (7.5 × 106). NS, P > 0.05 (two-way ANOVA). Data are combined from two independent experiments (a; mean ± s.e.m. of n = 10, 12 mice per group) or three independent experiments (b; mean ± s.e.m. of n = 17, 19 mice per group).

Supplementary Figure 3 Efficiency of NK cell deletion in anti-NK1.1-treated mice.

Depletion of NK cells in WT (BL6) mice by anti-NK1.1 (clone PK136) treatment. 500 μg of αNK1.1 antibody per dose, or an equivalent volume of PBS, was injected intravenously via the tail vein into WT mice, beginning 5 days prior to the adoptive transfer of 2D2 WT TH17 cells and continuing until 16 days post-transfer (indicated by red arrows). On the day of the adoptive transfer of 2D2 WT TH17 cells (day 0), peripheral lymphoid and non-lymphoid tissues were harvested from representative mice, and NK cells were quantified to assess the efficacy of depletion. On day 21 post-transfer, CNS-infiltrating cells were harvested from representative mice to assess the efficacy and duration of NK cell depletion in the CNS. Data are representative of three independent experiments; numbers indicate the frequency of CD3NKp46+ cells among total live cells.

Supplementary Figure 4 NK cell deficiency in Eomesf/f NKp46-Cre+mice.

(a-b) Frequency and absolute numbers of conventional NK cells (TCRβNKp46+NK1.1+IL-7Rα) in lymphoid and non-lymphoid tissues of Eomesf/f or Eomesf/f NKp46-Cre+mice 12-14 days after active immunization with MOG35-55/CFA and pertussis toxin. Data are representative of two independent experiments (n = 3 mice per group).

Supplementary Figure 5 IFN-γ is required for the activation/maturation of meningeal APCs.

(a) Mean clinical scores of Ifng−/− and WT mice, which received 7.5 × 106 2D2 WT TH17 cells. (b) Absolute number of 2D2 CD4+ T cells (CD4+Vβ11+) in the spinal cord meninges and CNS parenchyma of WT or Ifng−/− mice as quantitated by flow cytometry. (c) Absolute number of CD206+ perivascular macrophages (PVMs), myeloid DCs (mDCs), inflammatory monocytes (iMO) and neutrophils in the meninges of WT or Ifng−/− mice as quantitated by flow cytometry (Supplementary Fig. 6 for gating strategy). (d) Flow cytometric analyses of the maturation markers on meningeal antigen presenting cells in WT or Ifng−/− mice following the adoptive transfer of 7.5 × 106 2D2 WT TH17 cells. Activation of spinal cord meningeal antigen presenting cells was determined by staining perivascular macrophages and myeloid-derived dendritic cells with antibodies to MHC ClassII (I-A/I-E) molecules, CD80 and ICAM1. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001 (two-way ANOVA (a) or two-tailed Student’s t-test (b-d)). Data are representative of two independent experiments (a; mean ± s.e.m. of n = 10 mice per group) or combined from two independent experiments (b-d; mean ± SEM of n = 10 mice per group).

Supplementary Figure 6 Gating strategy for resident and infiltrating meningeal APCs.

Spinal cord meningeal cells were harvested at the peak of EAE disease (day 17 - 22 post-transfer), gated on live CD45+ cells, and further identified as follows: perivascular macrophages (Thy1.2 CD11b+ Ly6C Ly6GF4/80+ CD206+), myeloid-derived dendritic cells (Thy1.2MHCII+CD11c+CD11b+), inflammatory monocytes (Thy1.2 CD11b+Ly6C+ Ly6G) and neutrophils (Thy1.2 CD11b+Ly6CLy6G+).

Supplementary Figure 7 NKp46+ ILCs are dispensable for activation of meningeal APCs.

(a) Absolute number of CD206+ perivascular macrophages (PVMs), myeloid DCs (mDCs), inflammatory monocytes (iMO) and neutrophils in the meninges of Tbx21f/f or Tbx21f/f NKp46-Cre+ mice as quantitated by flow cytometry (Supplementary Fig. 6 for gating strategy). (b) Flow cytometric analyses of the activation markers on meningeal antigen presenting cells in Tbx21f/f or Tbx21f/f NKp46-Cre+ mice following the adoptive transfer of 7.5 × 106 2D2 WT TH17 cells. Activation of spinal cord meningeal antigen presenting cells was determined by staining perivascular macrophages and myeloid-derived dendritic cells with antibodies to MHC ClassII (I-A/I-E) molecules, CD80 and ICAM1. Naïve healthy WT (BL6) mice were used to assess the steady-state expression of maturation markers. Expression levels are presented as mean fluorescence intensity (MFI) of staining (b); data are combined from 3 independent experiments with 11 mice per group.

Supplementary Figure 8 T-bet-dependent NKp46+ ILCs are not required for GM-CSF production by IL-17A-producing 2D2 CD4+ T cells.

Intracellular cytokine staining of 2D2 CD4+ T cells (CD4+Vβ11+) isolated from the meninges and CNS parenchyma of Tbx21f/f or Tbx21f/f NKp46-Cre+ mice following adoptive transfer of 7.5 × 106 2D2 WT TH17 cells. CNS-infiltrating cells were harvested at the peak of EAE disease (day 17-22 post-transfer) and re-stimulated ex vivo with PMA/ionomycin prior to staining. Bar graphs present mean ± s.e.m of cytokine producing 2D2 CD4+ T cells isolated from the spinal cord meninges and parenchyma of Tbx21f/f or Tbx21f/f NKp46-Cre+ mice. NS, P > 0.05; *, P < 0.01 (two-tailed Student’s t-test). Data are combined from 2 independent experiments (n = 6, 8 mice per group).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Table 1.

Life Sciences Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kwong, B., Rua, R., Gao, Y. et al. T-bet-dependent NKp46+ innate lymphoid cells regulate the onset of TH17-induced neuroinflammation. Nat Immunol 18, 1117–1127 (2017). https://doi.org/10.1038/ni.3816

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3816

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