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:

Neural stem cells sustain natural killer cells that dictate recovery from brain inflammation

Abstract

Recovery from organ-specific autoimmune diseases largely relies on the mobilization of endogenous repair mechanisms and local factors that control them. Natural killer (NK) cells are swiftly mobilized to organs targeted by autoimmunity and typically undergo numerical contraction when inflammation wanes. We report the unexpected finding that NK cells are retained in the brain subventricular zone (SVZ) during the chronic phase of multiple sclerosis in humans and its animal model in mice. These NK cells were found preferentially in close proximity to SVZ neural stem cells (NSCs) that produce interleukin-15 and sustain functionally competent NK cells. Moreover, NK cells limited the reparative capacity of NSCs following brain inflammation. These findings reveal that reciprocal interactions between NSCs and NK cells regulate neurorepair.

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: Retention of NK cells in the SVZ of MS patients.
Figure 2: Preferential accumulation of NK cells in the SVZ during progression of brain inflammation.
Figure 3: NSCs have a distinct cytokine profile during progression of brain inflammation.
Figure 4: NSC-derived IL-15 is necessary to maintain NK cell proliferation, survival and IFN-γ production.
Figure 5: Coordinated upregulation of IL-15 and IL-15Rα in NSCs during progression of brain inflammation.
Figure 6: IL-15Rα upregulation in NSCs during the late stages of brain inflammation.
Figure 7: NK cells eliminate NSCs during the late stages of brain inflammation.
Figure 8: Altered expression of MHC class I molecules in NSPCs and effects of NK cell removal on EAE.

Similar content being viewed by others

References

  1. Shi, F.D., Ljunggren, H.G., La Cava, A. & Van Kaer, L. Organ-specific features of natural killer cells. Nat. Rev. Immunol. 11, 658–671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Vivier, E. et al. Innate or adaptive immunity? The example of natural killer cells. Science 331, 44–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sun, J.C. & Lanier, L.L. NK cell development, homeostasis and function: parallels with CD8+ T cells. Nat. Rev. Immunol. 11, 645–657 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Yokoyama, W.M., Kim, S. & French, A.R. The dynamic life of natural killer cells. Annu. Rev. Immunol. 22, 405–429 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Schleinitz, N., Vély, F., Harlé, J.R. & Vivier, E. Natural killer cells in human autoimmune diseases. Immunology 131, 451–458 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Flodström-Tullberg, M., Bryceson, Y.T., Shi, F.D., Höglund, P. & Ljunggren, H.G. Natural killer cells in human autoimmunity. Curr. Opin. Immunol. 21, 634–640 (2009).

    Article  PubMed  CAS  Google Scholar 

  7. Sun, J.C., Beilke, J.N., Bezman, N.A. & Lanier, L.L. Homeostatic proliferation generates long-lived natural killer cells that respond against viral infection. J. Exp. Med. 208, 357–368 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jaeger, B.N. et al. Neutrophil depletion impairs natural killer cell maturation, function, and homeostasis. J. Exp. Med. 209, 565–580 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Sun, J.C., Beilke, J.N. & Lanier, L.L. Adaptive immune features of natural killer cells. Nature 457, 557–561 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Robbins, S.H., Tessmer, M.S., Mikayama, T. & Brossay, L. Expansion and contraction of the NK cell compartment in response to murine cytomegalovirus infection. J. Immunol. 173, 259–266 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Zhang, B., Yamamura, T., Kondo, T., Fujiwara, M. & Tabira, T. Regulation of experimental autoimmune encephalomyelitis by natural killer (NK) cells. J. Exp. Med. 186, 1677–1687 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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  PubMed  PubMed Central  Google Scholar 

  13. Hao, J. et al. Interleukin-2/interleukin-2 antibody therapy induces target organ natural killer cells that inhibit central nervous system inflammation. Ann. Neurol. 69, 721–734 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Huang, D. et al. The neuronal chemokine CX3CL1/fractalkine selectively recruits NK cells that modify experimental autoimmune encephalomyelitis within the central nervous system. FASEB J. 20, 896–905 (2006).

    Article  CAS  PubMed  Google Scholar 

  15. Alvarez-Buylla, A. & Garcia-Verdugo, J.M. Neurogenesis in adult subventricular zone. J. Neurosci. 22, 629–634 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Faiz, M. et al. Substantial migration of SVZ cells to the cortex results in the generation of new neurons in the excitotoxically damaged immature rat brain. Mol. Cell. Neurosci. 38, 170–182 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. James, R., Kim, Y., Hockberger, P.E. & Szele, F.G. Subventricular zone cell migration: lessons from quantitative two-photon microscopy. Front. Neurosci. 5, 30 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Tepavčević, V. et al. Inflammation-induced subventricular zone dysfunction leads to olfactory deficits in a targeted mouse model of multiple sclerosis. J. Clin. Invest. 121, 4722–4734 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Rasmussen, S. et al. Reversible neural stem cell niche dysfunction in a model of multiple sclerosis. Ann. Neurol. 69, 878–891 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pluchino, S. et al. Persistent inflammation alters the function of the endogenous brain stem cell compartment. Brain 131, 2564–2578 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Mosher, K.I. et al. Neural progenitor cells regulate microglia functions and activity. Nat. Neurosci. 15, 1485–1487 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yang, J. et al. Adult neural stem cells expressing IL-10 confer potent immunomodulation and remyelination in experimental autoimmune encephalitis. J. Clin. Invest. 119, 3678–3691 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kokaia, Z., Martino, G., Schwartz, M. & Lindvall, O. Cross-talk between neural stem cells and immune cells: the key to better brain repair? Nat. Neurosci. 15, 1078–1087 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. Poli, A. et al. NK cells in central nervous system disorders. J. Immunol. 190, 5355–5362 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. 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  PubMed  PubMed Central  Google Scholar 

  26. Gan, Y. et al. Ischemic neurons recruit natural killer cells that accelerate brain infarction. Proc. Natl. Acad. Sci. USA 111, 2704–2709 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Pastrana, E., Cheng, L.C. & Doetsch, F. Simultaneous prospective purification of adult subventricular zone neural stem cells and their progeny. Proc. Natl. Acad. Sci. USA 106, 6387–6392 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yirmiya, R. & Goshen, I. Immune modulation of learning, memory, neural plasticity and neurogenesis. Brain Behav. Immun. 25, 181–213 (2011).

    Article  CAS  PubMed  Google Scholar 

  29. Huntington, N.D. et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J. Exp. Med. 206, 25–34 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee, G.A. et al. Different NK cell developmental events require different levels of IL-15 trans-presentation. J. Immunol. 187, 1212–1221 (2011).

    Article  CAS  PubMed  Google Scholar 

  31. Lucas, M., Schachterle, W., Oberle, K., Aichele, P. & Diefenbach, A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 26, 503–517 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Oda, K. et al. Brefeldin A arrests the intracellular transport of a precursor of complement C3 before its conversion site in rat hepatocytes. FEBS Lett. 214, 135–138 (1987).

    Article  CAS  PubMed  Google Scholar 

  33. Broutman, G. & Baudry, M. Involvement of the secretory pathway for AMPA receptors in NMDA-induced potentiation in hippocampus. J. Neurosci. 21, 27–34 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Shi, F.D. et al. Natural killer cells determine the outcome of B cell-mediated autoimmunity. Nat. Immunol. 1, 245–251 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Lee, N. et al. Ciliary neurotrophic factor receptor regulation of adult forebrain neurogenesis. J. Neurosci. 33, 1241–1258 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Suyama, S. et al. Purinergic signaling promotes proliferation of adult mouse subventricular zone cells. J. Neurosci. 32, 9238–9247 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu, C. & Hermann, T.E. Characterization of ionomycin as a calcium ionophore. J. Biol. Chem. 253, 5892–5894 (1978).

    Article  CAS  PubMed  Google Scholar 

  38. Brilot, F., Strowig, T., Roberts, S.M., Arrey, F. & Münz, C. NK cell survival mediated through the regulatory synapse with human DCs requires IL-15Ralpha. J. Clin. Invest. 117, 3316–3329 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bergamaschi, C. et al. Intracellular interaction of interleukin-15 with its receptor alpha during production leads to mutual stabilization and increased bioactivity. J. Biol. Chem. 283, 4189–4199 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Aharoni, R., Arnon, R. & Eilam, R. Neurogenesis and neuroprotection induced by peripheral immunomodulatory treatment of experimental autoimmune encephalomyelitis. J. Neurosci. 25, 8217–8228 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sajad, M. et al. Cytokinetics of adult rat SVZ after EAE. Brain Res. 1371, 140–149 (2011).

    Article  CAS  PubMed  Google Scholar 

  42. 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 

  43. Liu, Q. et al. Interleukin-17 inhibits adult hippocampal neurogenesis. Sci. Rep. 4, 7554 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jang, H. et al. Inflammatory effects of highly pathogenic H5N1 influenza virus infection in the CNS of mice. J. Neurosci. 32, 1545–1559 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Liu, Q., Xie, X., Lukas, R.J., St John, P.A. & Wu, J. A novel nicotinic mechanism underlies β-amyloid-induced neuronal hyperexcitation. J. Neurosci. 33, 7253–7263 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kemper, K. et al. The AC133 epitope, but not the CD133 protein, is lost upon cancer stem cell differentiation. Cancer Res. 70, 719–729 (2010).

    Article  CAS  PubMed  Google Scholar 

  47. Suzuki, M. et al. Defective membrane expression of the Na+HCO3 cotransporter NBCe1 is associated with familial migraine. Proc. Natl. Acad. Sci. USA 107, 15963–15968 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Carbajal, K.S., Schaumburg, C., Strieter, R., Kane, J. & Lane, T.E. Migration of engrafted neural stem cells is mediated by CXCL12 signaling through CXCR4 in a viral model of multiple sclerosis. Proc. Natl. Acad. Sci. USA 107, 11068–11073 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Cearley, C.N. & Wolfe, J.H. A single injection of an adeno-associated virus vector into nuclei with divergent connections results in widespread vector distribution in the brain and global correction of a neurogenetic disease. J. Neurosci. 27, 9928–9940 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Y. Yan for technical help. This study was supported in part by National Basic Research Program of China grant 2013CB966900; National Science Foundation of China grants 81230028, 81301044 and 81471535; the National Key Clinical Specialty Construction Program of China; US National Institutes of Health grants R01NS082745, R01CA175391 and R01DK081536; and American Heart Association grant 16SDG27250236; and a National Multiple Sclerosis Society research grant.

Author information

Authors and Affiliations

Authors

Contributions

Q.L., N.S. and F.-D.S. designed the studies; Q.L. and W.-N.J. performed the studies; A.L.C. and L.V.K. advised on design, execution of experiments and interpretation of results; and Q.L., A.L.C., L.V.K. and F.-D.S. wrote and edited the manuscript.

Corresponding authors

Correspondence to Qiang Liu or Fu-Dong Shi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Fluorescence minus one (FMO) controls for the identification of CNS-infiltrating NK cells.

Single cell suspensions were prepared from CNS tissues of Cd1d−/− or Cd1d−/−NK1.1-tdTomato+ mice at day 30 of EAE and stained with fluorochrome-conjugated antibodies. a-b. Representative flow cytometry dot plots show the identification of gating boundaries of CD3 (FITC), td-Tomato and IFN-γ (APC) with corresponding FMO controls.

Supplementary Figure 2 Reduced expression of proinflammatory factors during the recovery phase of EAE.

CNS homogenates were prepared from EAE mice of the indicated groups at 14, 30, 40, 60 dpi. Concentrations were measured by a Multi-Analyte ELISArray kit. Average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 3.5 ± 0.7 at 14 dpi, 2.5 ± 0.7 at 30 dpi, 2.3 ± 0.8 at 40 dpi and 2.1 ± 0.8 at 60 dpi. n = 9 per group. IFN-γ: P < 0.001 (14 dpi versus 30 dpi), P < 0.001 (14 dpi versus 40 dpi), P < 0.001 (14 dpi versus 60 dpi). F (3, 32) = 8.30, one-way ANOVA. IL-17A: P < 0.001 (14 dpi versus 30 dpi), P < 0.001 (14 dpi versus 40 dpi), P < 0.001 (14 dpi versus 60 dpi). F (3, 32) = 6.11, one-way ANOVA. TNF-α: P < 0.001 (14 dpi versus 30 dpi), P < 0.001 (14 dpi versus 40 dpi), P < 0.001 (14 dpi versus 60 dpi). F (3, 32) = 6.83, one-way ANOVA. IL-1β: P < 0.001 (14 dpi versus 30 dpi), P < 0.001 (14 dpi versus 40 dpi), P < 0.001 (14 dpi versus 60 dpi). F (3, 32) = 7.16, one-way ANOVA. IL-6: P < 0.001 (14 dpi versus 30 dpi), P < 0.001 (14 dpi versus 40 dpi), P < 0.001 (14 dpi versus 60 dpi). F (3, 32) = 5.54, one-way ANOVA. MCP-1: P < 0.001 (14 dpi versus 30 dpi), P < 0.001 (14 dpi versus 40 dpi), P < 0.001 (14 dpi versus 60 dpi). F (3, 32) = 3.39, one-way ANOVA. Results are from three individual experiments. Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 3 FMO controls for the identification of SVZ type B, type C and type A cells.

Single cell suspensions were prepared from dissociated SVZ tissues of WT adult mice and stained with fluorochrome-conjugated antibodies. a. Representative flow cytometry dot plots show the identification of gating boundaries of GFAP (PE), CD24 (FITC) and EGFR (AF647) with corresponding FMO controls. b. Representative flow cytometry dot plots show SVZ type B (GFAP+EGFR+CD24), type C (GFAPEGFR+CD24) and type A (GFAPEGFRCD24low) cell types from WT mice before sorting. c. Representative flow cytometry dot plots show purity of SVZ type B, type C and type A cell types (>98%) after sorting.

Supplementary Figure 4 Representative factors produced by CNS cells.

a. ELISA for IL-15, VEGF, IL-4, and MCP-1 from lysates of sorted astrocytes, microglia, neurons and NSCs show that IL-15 is a major factor produced by NSCs as compared to other CNS cells during the recovery phase of EAE (30 dpi). b-c. Immunostaining shows that most IL-15-expressing cells in SVZ are GFAP+, whereas only a small portion (<10%) of IL-15+ cells are microglia/macrophages (Iba1+). Average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.3 ± 0.6 at 30 dpi. n = 12 per group. Data represent three individual experiments. Error bars represent s.e.m.

Supplementary Figure 5 Removal of IL-15 from NSCs prevents NK cell retention during the late stage of brain inflammation.

a. Schematic map of the nestin-specific shIL-15 lentiviral vector. b. Intraventricular injection of 1 x 107 lentivirus particles containing shIL-15 or control sequence into recipient mice. c. Intraventricular injection of a lentivirus containing a shIL-15 sequence results in loss of IL-15 in nestin-expressing cells in the SVZ of Cd1d−/− mice. n = 12 per group. P < 0.001, t (22) = 5.58, unpaired t-test. d. Intraventricular injection of a lentivirus containing shIL-15 sequence at 20 dpi prevents NK cell retention in SVZ of Cd1d−/− mice at EAE 30 dpi, but not in striatum. Absolute numbers of cells per dissected SVZ or striatum tissue are shown. Average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.5 ± 0.6 in lenti-control group and 1.7 ± 0.5 in lenti-shIL-15 group at 30 dpi. n = 12 per group. P < 0.001 (Lenti-control SVZ versus Lenti-control striatum), P < 0.001 (Lenti-control SVZ vs. Lenti-sh-IL-15 SVZ). F (1, 44) = 5.5, two-way ANOVA. e. To determine BrdU incorporation in NK cells in vivo, EAE mice were given i.p. injections of 120 mg kg−1 BrdU in PBS daily, and were sacrificed 10 days after the last injection at 30 dpi. Representative flow cytometry plots show that intraventricular injection of a lentivirus containing a shIL-15 sequence reduces BrdU+ NK cell counts in SVZ of Cd1d−/− mice at 30 dpi. Bar graphs summarize the decreased counts of BrdU+ NK cells after intraventricular injection of a lentivirus containing a shIL-15 sequence at 30 dpi. Absolute numbers of cells per dissected SVZ tissue are shown. n = 12 per group. P = 0.006, t (22) = 2.23, unpaired t-test. f. Intraventricular injection of a lentivirus containing a shIL-15 sequence at 20 dpi reduces the counts of IFN-γ-expressing NK cells in SVZ of Cd1d−/− mice at EAE 30 dpi. Bar graphs summarize the decreased counts of IFN-γ-expressing NK cells after intraventricular injection of a lentivirus containing a shIL-15 sequence at 30 dpi. Absolute numbers of cells per dissected SVZ tissue are shown. n = 12 per group. P = 0.007, t (22) = 2.11, unpaired t-test. In e-f, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.6 ± 0.5 in lenti-control group and 1.8 ± 0.5 in Lenti-shIL-15 group at 30 dpi. Data represent three independent experiments. Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 6 FMO controls and gating strategy for BrdU+, IFN-γ+ or annexin V+ NK cells.

NK cells were cultured with flow cytometry-sorted NSCs from EAE SVZ (30 dpi) in transwell plates and counted by flow cytometry in the same well (with cell contact) or spatially separated (no cell contact) by a membrane (0.4 μm pores). Representative flow cytometry dot plots show FMO controls and gating strategy for BrdU+, IFN-γ+ or annexin V+ NK cells.

Supplementary Figure 7 Full-length blots for Figure 6.

Supplementary Figure 8 Depletion of NK cells increases the number of proliferating NSCs, but does not affect the numbers of proliferating SVZ type C and A cells during EAE recovery.

a. “Long-term” BrdU injection was used to determine the number of SVZ-retaining type B cells. Anti-NK1.1 mAb was used to deplete NK cells in Cd1d−/− mice devoid of natural killer T (NKT) cells. b. Increased numbers of SVZ GFAP+BrdU+ cells in EAE Cd1d−/− mice treated with anti-NK1.1 mAb as compared to mice injected with IgG control at 30 dpi. c. Counts of GFAP+BrdU+ cells were increased in EAE Cd1d−/− mice treated with NK1.1 mAb during the late stages of EAE (30, 40, 60 dpi) but not in the peak phase (14 dpi). n = 6 mice per group at each time point. P = 0.003 (30dpi), P = 0.002 (40dpi), P = 0.005 (60dpi). F (3, 40) = 2.82, two-way ANOVA. d. Decreased SVZ GFAP+caspase-3+ cells in EAE Cd1d−/− mice treated with NK1.1 mAb as compared to those injected with IgG control at 30 dpi. e. Counts of GFAP+caspase-3+ cells were significantly decreased in EAE Cd1d−/− mice treated with NK1.1 mAb as compared to IgG-treated control during the late stages of EAE (30, 40, 60 dpi) but not in the peak phase (14 dpi). n = 6 mice per group at each time point. P = 0.005 (30dpi), P = 0.004 (40dpi), P = 0.008 (60dpi). F (3, 40) = 2.23, two-way ANOVA. In c and e, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 3.5 ± 0.7 (IgG) and 3.3 ± 0.7 (NK 1.1 mAb) at 14 dpi, 2.6 ± 0.6 (IgG) and 1.6 ± 0.5 (NK 1.1 mAb) at 30 dpi, 2.3 ± 0.6 (IgG) and 1.5 ± 0.5 (NK 1.1 mAb) at 40 dpi and 2.2 ± 0.6 (IgG) and 1.3 ± 0.5 (NK 1.1 mAb) at 60 dpi. f. Anti-NK1.1 mAbs were injected after EAE induction, and “short-term” BrdU labeling was used to birthdate rapidly dividing SVZ progenitors in Cd1d−/− mice. g. Representative images show MASH1+BrdU+ or DCX+BrdU+ cells in EAE Cd1d−/− mice at 30 dpi. h. Unchanged counts of MASH1+BrdU+ or DCX+BrdU+ cells in EAE Cd1d−/− mice as compared to IgG-injected controls after pretreatment with anti-NK1.1 mAbs. In h, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 3.2 ± 0.6 (IgG) and 3.3 ± 0.5 (NK 1.1 mAb) at 14 dpi, 2.5 ± 0.7 (IgG) and 1.7 ± 0.5 (NK 1.1 mAb) at 30 dpi, 2.5 ± 0.7 (IgG) and 1.6 ± 0.6 (NK 1.1 mAb) at 40 dpi and 2.3 ± 0.5 (IgG) and 1.5 ± 0.6 (NK 1.1 mAb) at 60 dpi. n = 6 mice per group at each time point. Data represent three independent experiments. Scale bars: 40 μm, 20 μm (inset). Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 9 Removal of NK cells is associated with proliferation of SVZ NSPCs during the late stages of EAE.

a. Protocol of anti-NK1.1mAb injection, EAE induction and BrdU injection. b. Depletion of NK cells using anti-NK1.1 mAb in EAE Cd1d−/− mice is associated with increased numbers of BrdU+ cells in SVZ at 30 dpi. Control: 0 dpi. c. Quantitation of BrdU+ cells in EAE Cd1d−/− mice pretreated with IgG (control) or anti-NK1.1 mAb. n = 6 mice per group at each time point. P = 0.008 (30dpi), P = 0.006 (40dpi), P = 0.007 (60dpi). F (3, 40) = 7.73, two-way ANOVA. d. Depletion of NK cells is associated with increased numbers of DCX+ cells in SVZ of EAE Cd1d−/− mice at 30 dpi. Control: 0 dpi. e. Quantitation of DCX+ cells in EAE Cd1d−/− mice pretreated with IgG or anti-NK1.1 mAb. n = 6 mice per group at each time point. P = 0.007 (30dpi), P = 0.012 (40dpi), P = 0.013 (60dpi). F (3, 40) = 5.35, two-way ANOVA. In b-e, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 3.5 ± 0.7 (IgG) and 3.3 ± 0.7 (NK 1.1 mAb) at 14 dpi, 2.7 ± 0.5 (IgG) and 1.8 ± 0.7 (NK 1.1 mAb) at 30 dpi, 2.6 ± 0.8 (IgG) and 1.5 ± 0.8 (NK 1.1 mAb) at 40 dpi and 2.5 ± 0.6 (IgG) and 1.3 ± 0.5 (NK 1.1 mAb) at 60 dpi. f. EAE induction and BrdU injection in Rag2−/−or Rag2−/−γc−/− Cd1d−/− mice that received 2D2 Cd1d−/− T cells (devoid of NKT cells). g. Deletion of NK cells prevents reduction of SVZ-proliferating (BrdU+) cells independently of T, B, and NKT cells at 30 dpi. Control: 0 dpi. h. Quantitation of BrdU+ cells in SVZ from Rag2−/− and Rag2−/−γc−/− mice during EAE. n = 6 mice per group at each time point. P = 0.008 (30dpi), P = 0.006 (40dpi), P = 0.008 (60dpi). F (3, 40) = 8.36, two-way ANOVA. i. Deficiency of NK cells is associated with increased numbers of DCX+ cells in Rag2−/− and Rag2−/−γc−/− mice at 30 dpi. Control: 0 dpi. j. Quantitation of DCX+ cells in SVZ from Rag2−/− and Rag2−/−γc−/− mice. n = 6 mice per group at each time point. P = 0.043 (30dpi), P = 0.036 (40dpi), P = 0.009 (60dpi). F (3, 40) = 5.65, two-way ANOVA. In g-j, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 3.2 ± 0.6 (Rag2−/−) and 3.4 ± 0.7 (Rag2−/−γc−/−) at 14 dpi, 2.5 ± 0.8 (Rag2−/−) and 1.7 ± 0.7 (Rag2−/−γc−/−) at 30 dpi, 2.3 ± 0.7 (Rag2−/−) and 1.5 ± 0.8 (Rag2−/−γc−/−) at 40 dpi and 2.2 ± 0.6 (Rag2−/−) and 1.3 ± 0.5 (Rag2−/−γc−/−) at 60 dpi. Experiments were repeated three times. Scale bars: 40 μm. Error bars represent s.e.m.; * P < 0.05; ** P < 0.01.

Supplementary Figure 10 NK cell–induced cytotoxicity against SVZ type B cells.

a. Cytotoxicity against NSCs by NK cells was measured by 51Cr-release assay. Target cells (cultured NSCs) were labeled with 51Cr. 104 NSCs/well from naïve control, EAE 14 dpi and EAE 30 dpi SVZ, were co-cultured with activated NK cells at different effector:target ratios (50:1, 10:1; 5:1; 1:1, and 1:10) for 24 h. Activated NK cells were prepared by IL-2 (10 μg ml−1), IL-15 (10 μg ml−1), and LPS (5 μg ml−1) stimulation for 96 h prior to co-culture. Cytotoxicity was measured and data shown are expressed as percentage of naïve controls from four independent experiments with each value repeated in triplicate. Average EAE disease grade for mice used for NSC isolation at the time of tissue harvesting was 3.6 ± 0.7 at 14 dpi and 2.6 ± 0.6 at 30 dpi. n = 12 per group. P < 0.001 (50:1), P < 0.001 (10:1), P < 0.001 (5:1), P < 0.001 (1:1). F (8, 165) = 6.26, two-way ANOVA. b. A representative image of Fura-2AM-loaded NSCs and selected region of interest (ROI). Scale bar, 20 µm. c. Increased Ca2+ influx signals in NSCs isolated from EAE SVZ at 30 dpi and treated with activated NK cells. Ca2+ influx signals were measured by the ratio of 340/380nm in the selected ROIs of NSCs. d. Averaged Ca2+ influx signals in ROIs selected from NSCs from the indicated groups, which were treated or untreated with activated NK cells for 12 h. Average EAE disease grade for mice used for NSC isolation at the time of tissue harvesting was 3.5 ± 0.6 at 14 dpi and 2.3 ± 0.8 at 30 dpi. n = 8 per group. P = 0.005 (30 dpi NSCs + NK cells versus 0 dpi NSCs), P = 0.008 (30 dpi NSCs + NK cells versus 14 dpi NSCs + NK cells). F (2, 21) = 2.78, one-way ANOVA. In a-d, data represent four independent experiments. e. NSCs were sorted from EAE SVZ at 30 dpi by flow cytometry and cultured in vitro for 48 h. Immunostaining shows increased counts of GFAP+caspase-3+ cells relative to total GFAP+ cells after treatment with NK cells for 24 h. n = 12 per group. P = 0.006 (30 dpi NSCs + NK cells 1:1 versus 30 dpi NSCs), P = 0.003 (30 dpi NSCs + NK cells 1:10 versus 30 dpi NSCs). F (2, 33) = 5.88, one-way ANOVA. f. Cell proliferation was determined by in vitro BrdU incorporation and co-labeling with GFAP. Counts of BrdU+GFAP+ cells relative to total GFAP+ cells in NSC cultures were unaffected by treatment with NK cells. Results in e-f are from three independent experiments. n = 12 per group. Average EAE disease grade for mice used for NSC isolation at the time of tissue harvesting was 2.5 ± 0.6 at 30 dpi. Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 11 Construction of and treatment with adenoviral vectors encoding Qa1.

a. Full-length Qa1 cDNA was amplified by RT-PCR from RNA isolated from splenocytes of C57BL/6 mice. After verification of the sequence, cDNA was ligated into the DUAL-CCM+ shuttle vector with a C-terminal Flag epitope tag under the control of a CMV promoter. The recombinant adenovirus was custom generated by Vector Biolabs. b. A representative image of SVZ type B cells (GFAP+EGFR+DAPI+) freshly isolated and sorted by flow cytometry from Cd1d−/− mice at 30 dpi. Scale: 10 μm. c−d. SVZ type B cells freshly isolated and sorted by flow cytometry from Cd1d−/− mice at 30 dpi were cultured prior to infection with 1X107 pfu adenovirus without Qa1 sequence (control, c) or 1X107 pfu adenovirus containing Qa1 sequence (Ad-Qa1, d). Cells were fixed, permeabilized, blocked with anti-Flag Ab and stained with an Alexa Fluor secondary Ab. Scale: 20 μm. e. Intraventricular injection of adenovirus containing Qa1 sequence at 20 dpi prevents loss of Qa1 expression in SVZ of Cd1d−/− mice at 30 dpi, Scale: 50 μm. f. Intraventricular injection of adenovirus containing Qa1 sequence at 20 dpi prevents loss of Qa1 expression in SVZ, but not in striatum (STM) or cortex (CTX) of Cd1d−/− mice at 30 dpi, Scale: 40 μm. n = 12 mice per group. P = 0.001 (Ad-control SVZ versus Ad-Qa1 SVZ), t (22) = 3.83, unpaired t-test. In e-f, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.6 ± 0.5 in Ad-control group and 1.5 ± 0.5 in Ad-Qa1 group at 30 dpi. Data represent three independent experiments. Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 12 Qa1 overexpression in the SVZ promotes the survival of NSCs during the late stages of brain inflammation.

a. “Long-term” BrdU injection was used to determine the number of SVZ-retaining type B cells. Representative images and bar graph show increased BrdU+ cells in EAE SVZ (30 dpi) of Cd1d−/− mice receiving intraventricular injection of adenovirus containing Qa1 sequence at 20 dpi. EAE mice receiving adenovirus containing null control sequence were used as control (Ad-control). Control: 0 dpi. Average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.4 ± 0.5 in Ad-control group and 1.5 ± 0.4 in Ad-Qa1 group at 30 dpi. Scale: 50 μm. n = 12 mice per group. P = 0.02 (EAE + Ad-control versus EAE + Ad-Qa1), t (22) = 2.42, unpaired t-test. b. Cytotoxicity against NSCs by NK cells was measured by 51Cr-release assay. Target cells (NSCs) were obtained from naïve control SVZ, EAE 30 dpi SVZ from Cd1d−/− mice receiving intraventricular injection of adenovirus containing Qa1 or null control sequence at 20 dpi. Target cells were labeled with 51Cr. 104 NSCs/well were then co-cultured with activated NK cells at different effector:target ratios (50:1, 10:1; 5:1; 1:1, and 1:10) for 24 h. Activated NK cells were prepared by IL-2 (10 μg ml−1), IL-15 (10 μg ml−1), and LPS (5 μg ml−1) stimulation for 96 h prior to co-culture. Cytotoxicity was measured and data shown are expressed as percentage of naïve controls from four independent experiments with each value repeated in triplicate. Average EAE disease grade for mice used for NSC isolation at the time of tissue harvesting was 2.6 ± 0.7 in Ad-control group and 1.4 ± 0.6 in Ad-Qa1 group at 30 dpi. n = 12 per group. P < 0.001 (50:1), P < 0.001 (10:1), P < 0.001 (5:1), P < 0.001 (1:1). F (8, 165) = 3.73, two-way ANOVA. c. Qa1 overexpression attenuates the increase of Ca2+ influx signals in NSCs treated with activated NK cells. Ca2+ influx signals were measured by the ratio of 340/380nm in the selected ROIs of NSCs. NSCs were obtained from naïve control SVZ, EAE 30 dpi SVZ of Cd1d−/− mice receiving intraventricular injection of adenovirus containing Qa1 or null control sequence at 20 dpi. Averaged Ca2+ influx signals in ROIs selected from NSCs from the indicated groups, which were treated with activated NK cells for 12 h. Average EAE disease grade for mice used for NSC isolation at the time of tissue harvesting was 2.5 ± 0.6 in Ad-control group and 1.5 ± 0.5 in Ad-Qa1 group at 30 dpi. Control: 0 dpi. n = 8 per group. P = 0.003 (30 dpi NSCs + Ad-control versus Control), P = 0.006 (30 dpi NSCs +Ad-control versus 30 dpi NSCs + Ad-Qa1). F (2, 21) = 9.37, one-way ANOVA. d. NSCs were sorted from naïve control SVZ, EAE 30 dpi SVZ of Cd1d−/− mice receiving intraventricular injection of adenovirus containing Qa1 or null control sequence at 20 dpi. Sorted NSCs were then cultured in vitro for 48 h. Immunostaining shows that Qa1 overexpression reduced the increase of GFAP+caspase-3+ cell counts relative to total GFAP+ cells after treatment with NK cells for 24 h. Control: 0 dpi. Average EAE disease grade for mice used for NSC isolation at the time of tissue harvesting was 2.3 ± 0.5 in Ad-control group and 1.4 ± 0.4 in Ad-Qa1 group at 30 dpi. n = 12 per group. P = 0.003 (30 dpi NSCs + Ad-control versus Control), P = 0.02 (30 dpi NSCs + Ad-control versus 30 dpi NSCs + Ad-Qa1). F (2, 33) = 3.12, one-way ANOVA. Data represent four independent experiments. Error bars represent s.e.m.; * P < 0.05, ** P < 0.01.

Supplementary Figure 13 Removal of NK cells or overexpression of Qa1 in the SVZ promotes oligodendrogenesis around the SVZ.

EAE was induced in Cd1d−/−, Rag2−/− and Rag2−/−γ c−/− mice. To induce EAE, Rag2−/− and Rag2−/−γc−/− mice were transferred with 5X106 2D2 T cells on day 0. To deplete NK cells, Cd1d−/− mice received anti-NK1.1 mAb injection (i.p.) at two days prior to MOG immunization and every 5 days thereafter until termination of experiments. EAE mice received intraventricular injection of adenovirus containing Qa1 or null control sequence at 20 dpi and SVZ tissues from EAE mice were harvested at 30 dpi. a. To determine the generation of neural/glial antigen 2 (NG2)/ BrdU+ oligodendrocyte progenitors, EAE mice were given i.p. injections of 120 mg kg−1 BrdU in PBS every 2 h for 10 h, and were sacrificed 1 week (7 d washout time) after the last injection at 30 dpi. Quantitation of BrdU+NG2+ cells in the SVZ of EAE mice (30 dpi) is expressed as the total number of positive cells per area measured (cells per mm2). n = 12 mice per group. P = 0.004 (IgG versus NK1.1 mAb), t (22) = 2.36. P = 0.004 (Ad-control versus Ad Qa1), t (22) = 2.28. P = 0.006 (Rag2−/− versus Rag2−/−γc−/−), t (22) = 2.03. P = 0.007 (Rag2−/− + Ad-control versus Rag2−/−+ Ad-Qa1), t (22) = 1.96, unpaired t-test. b. To determine newly generated mature cyclic nucleotide phosphohydrolase (CNPase)/ BrdU+ oligodendrocytes, mice were injected i.p. with 150 mg kg−1 BrdU twice per day for 2 days and were killed 2 weeks (14 d washout time) after the last injection. Quantitation of BrdU+CNPase+ cells in the SVZ of EAE mice (30 dpi) is expressed as the total number of positive cells per area measured (cells per mm2). In a-b, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.6 ± 0.6 in IgG group, 1.3 ± 0.5 in NK1.1-mAb group, 2.5 ± 0.5 in Ad-control group, 1.4 ± 0.6 in Ad-Qa1 group, 2.3 ± 0.5 in Rag2−/− group, 1.2 ± 0.6 in Rag2−/−γc−/− group, 2.4 ± 0.7 in Rag2−/− + Ad-control group, 1.3 ± 0.6 in Rag2−/− + Ad-Qa1 group at 30 dpi, Scale bars: 40 μm. n = 12 mice per group. P = 0.003 (IgG versus NK1.1 mAb), t (22) = 2.83. P = 0.003 (Ad-control versus Ad Qa1), t (22) = 3.01. P = 0.007 (Rag2−/− versus Rag2−/−γc−/−), t (22) = 2.05. P = 0.006 (Rag2−/− + Ad-control versus Rag2−/−+ Ad-Qa1), t (22) = 2.12, unpaired t-test. Data represent three independent experiments. Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 14 Removal of NK cells promotes oligodendrogenesis in the spinal cord.

EAE was induced in Cd1d−/−, Rag2−/− and Rag2−/−γc−/− mice. To deplete NK cells, Cd1d−/− mice received anti-NK1.1 mAb injection (i.p.) at two days prior to MOG immunization and every 5 days thereafter until termination of experiments. Spinal cord tissues were obtained at 30 dpi from EAE mice. Counts of NG2+ or CNPase+ cells were measured by flow cytometry analysis. a. Representative flow cytometry plots show the identification of gating boundaries of NG2 (FITC) with corresponding FMO control. b. Representative flow cytometry plots show the identification of gating boundaries of CNPase (FITC) with corresponding FMO control. c. Quantitation of NG2+ cells in the spinal cord of EAE mice (30 dpi) is expressed as the total number of positive cells per mouse spinal cord. n = 12 mice per group. P = 0.006 (IgG versus NK1.1 mAb), t (22) = 2.06. P = 0.008 (Rag2−/− versus Rag2−/−γc−/−), t (22) = 1.92, unpaired t-test. d. Quantitation of CNPase+ cells in the spinal cord of EAE mice (30 dpi) is expressed as the total number of positive cells per mouse spinal cord. n = 12 mice per group. P = 0.007 (IgG versus NK1.1 mAb), t (22) = 1.98. P = 0.006 (Rag2−/− versus Rag2−/−γc−/−), t (22) = 2.23, unpaired t-test. In c-d, average EAE disease grade for mice used for tissue analysis at the time of tissue harvesting was 2.7 ± 0.6 in IgG group, 1.5 ± 0.6 in NK1.1-mAb group, 2.4 ± 0.6 in Rag2−/− group and 1.3 ± 0.4 in Rag2−/−γc−/− group at 30 dpi. Data represent three independent experiments. Error bars represent s.e.m.; ** P < 0.01.

Supplementary Figure 15 Oligodendrocyte progenitors are resistant to NK cell killing.

a. Representative flow cytometry plots and immunostained image of oligodendrocyte progenitor cells show that oligodendrocyte progenitor cells express Qa1. Scale bar, 20 µm. b. Quantitation of Qa1+ oligodendrocyte progenitor cells shows unaltered expression of Qa1 in WT controls (0 dpi) or EAE mice at 14 or 30 dpi. Average EAE disease grade for mice used for cell isolation at the time of tissue harvesting was 3.3 ± 0.8 at 14 dpi and 2.6 ± 0.7 at 30 dpi. n = 6 mice per data plot. Results were obtained from three independent experiments. c. Cytotoxicity against oligodendrocyte progenitor cells by NK cells was measured by 51Cr-release assay. Target cells (oligodendrocyte progenitor cells) were obtained from naïve control, EAE 14 or 30 dpi CNS tissues from WT mice by sorting NG2+ cells from CNS tissues. Target cells were labeled with 51Cr. 1 x 104 oligodendrocyte progenitor cells s/well were then co-cultured with activated NK cells at different effector:target ratios (50:1, 10:1; 5:1; 1:1, and 1:10) for 24 h. Activated NK cells were prepared by IL-2 (10 μg ml−1), IL-15 (10 μg ml−1), and LPS (5 μg ml−1) stimulation for 96 h prior to coculture. Cytotoxicity was measured and data shown are expressed as percentage of naïve controls from four independent experiments with each value repeated in triplicate. Average EAE disease grade for mice used for cell isolation at the time of tissue harvesting was 3.5 ± 0.7 at 14 dpi and 2.5 ± 0.8 at 30 dpi. n = 12 per group. d. NSCs were sorted from SVZ of WT mice and oligodendrocyte progenitor cells (NG2+) were sorted from UBC-GFP mice respectively by flow cytometry at 30 dpi. Thereafter, oligodendrocyte progenitor cells and NSCs were co-cultured with or without IL-15 mAb in vitro for 48 h. Oligodendrocyte progenitor cells cultured alone without IL-15 mAb treatment were used as controls. Immunostaining shows unaltered counts of GFP+NG2+ cells relative to those in controls after treatment with IL-15 mAb or co-cultured with NSCs. n = 12 per group. e. NSCs were sorted from SVZ of WT mice and oligodendrocytes (CNPase+) were sorted from UBC-GFP mice respectively by flow cytometry at 30 dpi. Thereafter, oligodendrocytes and NSCs were co-cultured with or without IL-15 mAb in vitro for 48 h. Oligodendrocytes cultured alone without IL-15 mAb treatment were used as controls. Immunostaining shows unaltered counts of GFP+CNPase+ cells relative to those in controls after treatment with IL-15 mAb or co-cultured with NSCs. n = 12 per group. Average EAE disease grade for mice used for cell isolation at the time of tissue harvesting was 3.5 ± 0.8 (WT) and 3.2 ± 0.7 (UBC-GFP) at 14 dpi, 2.5 ± 0.7 (WT) and 2.3 ± 0.8 (UBC-GFP) at 30 dpi. Results were obtained from four independent experiments. Error bars represent s.e.m.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, Q., Sanai, N., Jin, WN. et al. Neural stem cells sustain natural killer cells that dictate recovery from brain inflammation. Nat Neurosci 19, 243–252 (2016). https://doi.org/10.1038/nn.4211

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4211

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