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:

An interferon-β-resistant and NLRP3 inflammasome–independent subtype of EAE with neuronal damage

Abstract

Inflammation induced by innate immunity influences the development of T cell–mediated autoimmunity in multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). We found that strong activation of innate immunity induced Nod-like receptor protein 3 (NLRP3) inflammasome–independent and interferon-β (IFNβ)-resistant EAE (termed type B EAE), whereas EAE induced by weak activation of innate immunity requires the NLRP3 inflammasome and is sensitive to IFNβ treatment. Instead, an alternative inflammatory mechanism, including membrane-bound lymphotoxin-β receptor (LTβR) and CXC chemokine receptor 2 (CXCR2), is involved in type B EAE development, and type B EAE is ameliorated by antagonizing these receptors. Relative expression of Ltbr and Cxcr2 genes was indeed enhanced in patients with IFNβ-resistant multiple sclerosis. Remission was minimal in type B EAE due to neuronal damages induced by semaphorin 6B upregulation on CD4+ T cells. Our data reveal a new inflammatory mechanism by which an IFNβ-resistant EAE subtype develops.

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: Type B EAE develops independently of the NLRP3 inflammasome and is resistant to IFNβ treatment.
Figure 2: mLT is a key initiator of type B EAE.
Figure 3: Gene expression profile of CD4+ T cells between type A and type B EAE.
Figure 4: Expression of Cxcr2, Cxcr1 and Ltbr in IFNβ-resistant EAE and MS.
Figure 5: Involvement of the CXCL1 and CXCR2 in development of type B EAE and type A EAE with MHV68 infection.
Figure 6: Persistent disease severity and spinal transected neurites in type B EAE mice.
Figure 7: Type B EAE-derived T cells cause neuronal retraction.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Haines, J.D., Inglese, M. & Casaccia, P. Axonal damage in multiple sclerosis. Mt. Sinai J. Med. 78, 231–243 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Río, J. et al. Defining the response to interferon-beta in relapsing-remitting multiple sclerosis patients. Ann. Neurol. 59, 344–352 (2006).

    Article  PubMed  CAS  Google Scholar 

  3. Rivers, T.M., Sprunt, D.H. & Berry, G.P. Observations on attempts to produce acute disseminated encephalomyelitis in monkeys. J. Exp. Med. 58, 39–53 (1933).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Arnon, R. & Sela, M. Immunomodulation by the copolymer glatiramer acetate. J. Mol. Recognit. 16, 412–421 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Steinman, L. The discovery of natalizumab, a potent therapeutic for multiple sclerosis. J. Cell Biol. 199, 413–416 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Simmons, S.B., Pierson, E.R., Lee, S.Y. & Goverman, J.M. Modeling the heterogeneity of multiple sclerosis in animals. Trends Immunol. 34, 410–422 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. 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 

  9. 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 

  10. Shaw, P.J. et al. Cutting edge: critical role for PYCARD/ASC in the development of experimental autoimmune encephalomyelitis. J. Immunol. 184, 4610–4614 (2010).

    Article  CAS  PubMed  Google Scholar 

  11. Smyk, D.S., Alexander, A.K., Walker, M. & Walker, M. Acute disseminated encephalomyelitis progressing to multiple sclerosis: are infectious triggers involved? Immunol. Res. 60, 16–22 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Meeker, R.B., Williams, K., Killebrew, D.A. & Hudson, L.C. Cell trafficking through the choroid plexus. Cell Adh. Migr. 6, 390–396 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Steinman, L. Some misconceptions about understanding autoimmunity through experiments with knockouts. J. Exp. Med. 185, 2039–2041 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lock, C., Oksenberg, J. & Steinman, L. The role of TNFalpha and lymphotoxin in demyelinating disease. Ann. Rheum. Dis. 58 (Suppl. 1), I121–I128 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Woodroofe, M.N. & Cuzner, M.L. Cytokine mRNA expression in inflammatory multiple sclerosis lesions: detection by non-radioactive in situ hybridization. Cytokine 5, 583–588 (1993).

    Article  CAS  PubMed  Google Scholar 

  16. Kraus, J. et al. Serum cytokine levels do not correlate with disease activity and severity assessed by brain MRI in multiple sclerosis. Acta Neurol. Scand. 105, 300–308 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Columba-Cabezas, S. et al. Suppression of established experimental autoimmune encephalomyelitis and formation of meningeal lymphoid follicles by lymphotoxin beta receptor-Ig fusion protein. J. Neuroimmunol. 179, 76–86 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Axtell, R.C. et al. T helper type 1 and 17 cells determine efficacy of interferon-β in multiple sclerosis and experimental encephalomyelitis. Nat. Med. 16, 406–412 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. 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  PubMed  Google Scholar 

  20. Wang, C. et al. CD5L/AIM regulates lipid biosynthesis and restrains Th17 cell pathogenicity. Cell 163, 1413–1427 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bustamante, M.F., Nurtdinov, R.N., Río, J., Montalban, X. & Comabella, M. Baseline gene expression signatures in monocytes from multiple sclerosis patients treated with interferon-beta. PLoS One 8, e60994 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Roth, G.A., Spada, V., Hamill, K. & Bornstein, M.B. Insulin-like growth factor I increases myelination and inhibits demyelination in cultured organotypic nerve tissue. Brain Res. Dev. Brain Res. 88, 102–108 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. McMorris, F.A., Smith, T.M., DeSalvo, S. & Furlanetto, R.W. Insulin-like growth factor I/somatomedin C: a potent inducer of oligodendrocyte development. Proc. Natl. Acad. Sci. USA 83, 822–826 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Magliozzi, R. et al. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain 130, 1089–1104 (2007).

    Article  PubMed  Google Scholar 

  25. Tawarayama, H., Yoshida, Y., Suto, F., Mitchell, K.J. & Fujisawa, H. Roles of semaphorin-6B and plexin-A2 in lamina-restricted projection of hippocampal mossy fibers. J. Neurosci. 30, 7049–7060 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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  PubMed  PubMed Central  Google Scholar 

  27. Liu, Y. et al. Preferential recruitment of neutrophils into the cerebellum and brainstem contributes to the atypical experimental autoimmune encephalomyelitis phenotype. J. Immunol. 195, 841–852 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Qin, H. et al. Signal transducer and activator of transcription-3/suppressor of cytokine signaling-3 (STAT3/SOCS3) axis in myeloid cells regulates neuroinflammation. Proc. Natl. Acad. Sci. USA 109, 5004–5009 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lukens, J.R. et al. The NLRP12 sensor negatively regulates autoinflammatory disease by modulating interleukin-4 production in T cells. Immunity 42, 654–664 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Siffrin, V. et al. In vivo imaging of partially reversible th17 cell-induced neuronal dysfunction in the course of encephalomyelitis. Immunity 33, 424–436 (2010).

    Article  CAS  PubMed  Google Scholar 

  31. Mo, R. et al. T cell chemokine receptor expression in aging. J. Immunol. 170, 895–904 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Liu, Y.J. et al. Peripheral T cells derived from Alzheimer's disease patients overexpress CXCR2 contributing to its transendothelial migration, which is microglial TNF-alpha-dependent. Neurobiol. Aging 31, 175–188 (2010).

    Article  CAS  PubMed  Google Scholar 

  33. Pokkali, S., Das, S.D. & Logamurthy, R. Expression of CXC and CC type of chemokines and its receptors in tuberculous and non-tuberculous effusions. Cytokine 41, 307–314 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Wang, Y. et al. Lymphotoxin beta receptor signaling in intestinal epithelial cells orchestrates innate immune responses against mucosal bacterial infection. Immunity 32, 403–413 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Revesz, T., Kidd, D., Thompson, A.J., Barnard, R.O. & McDonald, W.I. A comparison of the pathology of primary and secondary progressive multiple sclerosis. Brain 117, 759–765 (1994).

    Article  PubMed  Google Scholar 

  36. Thompson, A.J. et al. Major differences in the dynamics of primary and secondary progressive multiple sclerosis. Ann. Neurol. 29, 53–62 (1991).

    Article  CAS  PubMed  Google Scholar 

  37. Suto, F. et al. Plexin-a4 mediates axon-repulsive activities of both secreted and transmembrane semaphorins and plays roles in nerve fiber guidance. J. Neurosci. 25, 3628–3637 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhuang, B., Su, Y.S. & Sockanathan, S. FARP1 promotes the dendritic growth of spinal motor neuron subtypes through transmembrane Semaphorin6A and PlexinA4 signaling. Neuron 61, 359–372 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Suto, F. et al. Interactions between plexin-A2, plexin-A4, and semaphorin 6A control lamina-restricted projection of hippocampal mossy fibers. Neuron 53, 535–547 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Williams, A. et al. Semaphorin 3A and 3F: key players in myelin repair in multiple sclerosis? Brain 130, 2554–2565 (2007).

    Article  PubMed  Google Scholar 

  41. Liu, N.K. et al. Cytosolic phospholipase A2 protein as a novel therapeutic target for spinal cord injury. Ann. Neurol. 75, 644–658 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kolko, M., Rodriguez de Turco, E.B., Diemer, N.H. & Bazan, N.G. Neuronal damage by secretory phospholipase A2: modulation by cytosolic phospholipase A2, platelet-activating factor, and cyclooxygenase-2 in neuronal cells in culture. Neurosci. Lett. 338, 164–168 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Takemiya, T. et al. Endothelial microsomal prostaglandin E synthase-1 facilitates neurotoxicity by elevating astrocytic Ca2+ levels. Neurochem. Int. 58, 489–496 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Whitney, L.W., Ludwin, S.K., McFarland, H.F. & Biddison, W.E. Microarray analysis of gene expression in multiple sclerosis and EAE identifies 5-lipoxygenase as a component of inflammatory lesions. J. Neuroimmunol. 121, 40–48 (2001).

    Article  CAS  PubMed  Google Scholar 

  45. Neu, I., Mallinger, J., Wildfeuer, A. & Mehlber, L. Leukotrienes in the cerebrospinal fluid of multiple sclerosis patients. Acta Neurol. Scand. 86, 586–587 (1992).

    Article  CAS  PubMed  Google Scholar 

  46. Yoshikawa, K., Palumbo, S., Toscano, C.D. & Bosetti, F. Inhibition of 5-lipoxygenase activity in mice during cuprizone-induced demyelination attenuates neuroinflammation, motor dysfunction and axonal damage. Prostaglandins Leukot. Essent. Fatty Acids 85, 43–52 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Liu, L. et al. Functional defect of peripheral neutrophils in mice with induced deletion of CXCR2. Genesis 51, 587–595 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  49. Shinohara, M.L., Kim, J.H., Garcia, V.A. & Cantor, H. Engagement of the type I interferon receptor on dendritic cells inhibits T helper 17 cell development: role of intracellular osteopontin. Immunity 29, 68–78 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Shinohara, M.L. et al. Osteopontin expression is essential for interferon-α production by plasmacytoid dendritic cells. Nat. Immunol. 7, 498–506 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. Bioinform. in Action 17, 10–12 (2012).

    Google Scholar 

  52. Kersey, P.J. et al. Ensembl Genomes: an integrative resource for genome-scale data from non-vertebrate species. Nucleic Acids Res. 40, D91–D97 (2012).

    Article  CAS  PubMed  Google Scholar 

  53. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  Google Scholar 

  54. Robinson, M.D., McCarthy, D.J. & Smyth, G.K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  55. Gentleman, R.C. et al. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5, R80 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Robinson, M.D. & Smyth, G.K. Small-sample estimation of negative binomial dispersion, with applications to SAGE data. Biostatistics 9, 321–332 (2008).

    Article  PubMed  Google Scholar 

  57. Mootha, V.K. et al. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat. Genet. 34, 267–273 (2003).

    Article  CAS  PubMed  Google Scholar 

  58. Banker, G. & Goslin, K. Developments in neuronal cell culture. Nature 336, 185–186 (1988).

    Article  CAS  PubMed  Google Scholar 

  59. Eddy, N.B. & Leimbach, D. Synthetic analgesics. II. Dithienylbutenyl- and dithienylbutylamines. J. Pharmacol. Exp. Ther. 107, 385–393 (1953).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank A. Uezu for help in primary hippocampal neuron culture and Golgi staining, S. McKinnon for help in isolating optic nerves, M. Luftig for MHV68 experiments and M. Skeen for discussions on IFNβ treatment of MS patients. We are also grateful to S. Cote, S. Arvai, and all participants in the MURDOCK Study Community Registry and Biorepository and the MURDOCK Multiple Sclerosis Cohort Study. We thank MURDOCK Study leadership and staff, including principal investigator K. Newby, for providing samples and support. This study was funded by the National Multiple Sclerosis Society (RG4536 to M.L.S.; RG4723 to C.L.) and NIH (R01MH096979 and P41EB015897 to C.L.).

Author information

Authors and Affiliations

Authors

Contributions

M.I. and M.L.S. designed the study, analyzed data, and wrote the manuscript. M.I. performed most of the experiments. P.-h.C. performed methylation assay. S.G.G. selected RRMS patient samples from the MURDOCK study cohorts. S.S. performed qPCR analysis for RRMS patient samples. C.L. performed MRI analysis. E.B. assisted neuron imaging analyses. C.L., Q.-j.L., L.S., S.G.G. and E.B. contributed critical discussion for data and analysis.

Corresponding author

Correspondence to Mari L Shinohara.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 EAE induced by several immunization methods and phenotypic effects on cell migration, demyelination and nociceptive sensitivity in mice with type A and type B EAE.

(a) EAE severity in Nlrp3-/- mice was evaluated. EAE was induced with 6 different methods, as indicated in Online Methods. (b) AUC between 0 and 20-dpi. Method 1 (n=4), Method 2-6 (n=5). P(Method 1 vs 2)=0.0006, t(7)=5.98, P(Method 1 vs 3) <0.0001, t(7)=12.54, P(Method 1 vs 4)=0.0027, t(7)=4.54, P(Method 1 vs 5) <0.0001, t(7)=8.15, P(Method 1 vs 6) <0.0001, t(7)=10.63. (c) Numbers of total immune cells were evaluated in the brains and spinal cords (S.C.) of mice with Type-B EAE at 17-dpi. n=4. Brain: P(WT vs Nlrp3-/-)=0.8159, t(6)=0.2433, P(WT vs Asc-/-)=0.5685, t(6)=0.6031. Spinal cord: P(WT vs Nlrp3-/-)=0.5226, t(6)=0.6787, P(WT vs Asc-/-)=0.8984, t(6)=0.1332. (d) IFNβ treatment on Nlrp3-/- mice with Type-B EAE induced with Method 6. IFNβ (3x104 unit/mouse) were i.p. injected every other day from day 0 to 8 as previously performed. (e) Time course on body weight change in Type-A and Type-B EAE. (f) Il1b mRNA levels in splenic DC from naïve, Type-A, and Type-B EAE mice at 9 dpi. P(Naive vs Type-A)=0.0059, t(4)=5.344, P(Naive vs Type-B)<0.0001, t(4)=18.55, P(Type-A vs Type-B)=0.0004, t(4)=10.83. (g,h) Levels of extracellular IL-1β (f) and p20 caspase-1 (g) in 24h splenocyte culture supernatants (n=4 for Type-A, n=5 for Type-B). P=0.0261, t(6)=2.724 (g). P=0.0020, t(6)=4.794 (h). (i) Infiltrated cell numbers in the brain and spinal cord (n=7). Brain: P(Total)=0.0028, t(12)=3.747, P(CD4)=0.018, t(12)=2.737, P(Th17)=0.0167, t(12)=2.78, P(Th1)=0.0165, t(12)=2.784, P(CD8)=0.0329, t(12)=2.41, P(B)=0.0013, t(12)=4.165, P(DC)=0.0003, t(12)=5.008, P(PMN)=0.0004, t(12)=4.912, P(Mac)=0.0027, t(12)=3.759. Spinal cord: P(Total)=0.0035, t(12)=3.621, P(CD4)=0.0002, t(12)=5.372, P(Th17)=0.0003, t(12)=4.997, P(Th1)=0.00465, t(12)=2.221, P(CD8)=0.0104, t(12)=3.032, P(B)=0.0918, t(12)=1.833, P(DC)=0.0429, t(12)=2.263, P(PMN)=0.0011, t(12)=4.29, P(Mac)=0.089, t(12)=1.849. (j) Representative LFB-stained images of brains at 17-dpi. Red arrows indicate reduced LFB intensity, i.e., reduced myelin. Scale bars, 200 μm. (k) T2 FLAIR MRI analysis of spinal cords obtained from mice at 18-dpi. Yellow arrows indicate areas of potential myelin loss. (l) Thermal sensitivity evaluated by a hot-plate test in 9-dpi mice, which did not show any EAE symptoms and motor dysfunction. n=8. P(Naive vs Type-A)=0.1524, t(14)=1.514, P(Naive vs Type-B)=0.0013, t(14)=4.026, P(Type-A vs Type-B)=0.0217, t(14)=2.582. *; p<0.05. All statistical analyses in this figure were performed by two-tailed unpaired Student’s t-test. All the experimental data and images are representatives from at least 2 similar experiments for each.

Supplementary Figure 2 mLT expression on DCs.

(a) Flow charts showing mLT expression in DC (CD11c+ gated) obtained from naïve mice, Type-A EAE mice, and Type-B EAE mice at 9-dpi. (b) Percentages of LTα+ (i.e., mLT+) macrophages, determined by flow cytometry, in DLNs of naïve mice or mice with EAE at 9-dpi. Naïve (n=4), Type-A (n=3), Type-B (n=4). P(Naive vs Type-A)=0.1340, t(5)=1.7887, P(Naive vs Type-B)=0.0024, t(6)=45.021, P(Type-A vs Type-B)=0.0268, t(5)=3.101, by two-tailed unpaired Student’s t-test. (c) Evaluating mLT expression on DCs from mice received CFA injection alone without MOG. One group received 200 μg Mtb in CFA (Mtb dosage for Type-A EAE), and another group had 400 μg Mtb in CFA twice (Mtb dosage for Type-B EAE). (n=6) P(Naive vs Type-A)=00.7939, t(6)=0.2732, P(Naive vs Type-B)=0.0040, t(6)=4.519, P(Type-A vs Type-B)=0.0095, t(6)=3.748, by two-tailed unpaired Student’s t-test. (d) Methylation analysis was carried out by bisulfite conversion on the Lta promoter in DCs from naïve mice. Methylated and unmethylated CpG were shown with black and gray boxes, respectively

Supplementary Figure 3 RNA-seq analysis.

Heatmap for differentially expressed genes in CD4+ T cells isolated from naïve, Type-A EAE mice, and Type-B EAE mice at 9-dpi. Genes and samples have been clustered using correlation distance with complete linkage. A gene is listed on the heatmap if it had a p-value <= 0.05 and a log2FC >1 or < -1.

Supplementary Figure 4 Comparison of cell populations and gene expression in mice and RRMS patients who have diseases that do not respond to IFNβ therapies.

(a) Numbers of indicated cell types obtained from DLNs and spleen on 17-dpi were evaluated between Type-A and Type-B EAE. n=4. DLNs: P(Total)=0.2073, t(6)=1.413, P(CD3)=0.8466, t(6)=0.202, P(CD4)=0.9285, t(6)=0.09353, P(CD8)=0.7413, t(6)=0.3459, P(B)=0.4141, t(6)=0.8773, P(Th17)=0.9724, t(6)=0.03605, P(Th1)=0.9017, t(6)=0.1289, P(Treg)=0.3556, t(6)=1.001. Spleen: P(Total)=0.0072, t(6)=3.99, P(CD3)=0.2537, t(6)=1.262, P(CD4)=0.1781, t(6)=1.525, P(CD8)=0.3999, t(6)=0.906, P(B)=0.1371, t(6)=1.715. (b, c) Representative flow charts (b) and proportion (c) for GM-CSF-producing Th1 and Th17 cells from Type-A and Type-B EAE at 9-dpi. n=6. Th17: P=0.6568, t(10)=0.4579, Th1: P=0.3954, t(10)=0.8879. (d) Proportion for IFNγ-, GM-CSF-, IL-22-producing and CD5L-expressing Th17 cells in spinal cords from Type-A and Type-B EAE mice at 17-dpi. Type-A (n=4), Type-B (n=5). P(IFNγ)=0.3493, t(7)=1.0037, P(GM-CSF)=0.1760, t(7)=1.505, P(Il-22)=0.2859, t(7)=1.155, P(CD5L)=0.2641, t(7)=1.214. (e) Proportions and MFI of CXCR2+ neutrophils and macrophages obtained from naïve mice and mice with either Type-A or Type-B EAE at 9-dpi. n=4. PMN %: P(Naive vs Type-A)=0.0102, t(6)=3.695, P(Naive vs Type-B)=0.0004, t(6)= 7.001, P(Type-A vs Type-B)=0.0022, t(6)=5.131. PMN MFI: P(Naive vs Type-A)=0.0777, t(6)=2.125, P(Naive vs Type-B)=0.0014, t(6)= 5.588, P(Type-A vs Type-B)=0.0090, t(6)=3.81. Macrophage %: P(Naive vs Type-A)=0.0005, t(6)=6.774, P(Naive vs Type-B)=0.0006, t(6)= 6.48, P(Type-A vs Type-B)=0.0161, t(6)=3.314. Macrophage MFI: P(Naive vs Type-A)=0.33, t(6)=1.06, P(Naive vs Type-B)=0.0001, t(6)= 11.11, P(Type-A vs Type-B)=0.0001, t(6)=10.89. (f) Flow cytometry results showing CXCR2 protein expression on the CD4+ T surface in each group (naïve mice, Type-A, or Type-B EAE mice at 9-dpi). (g) Comparison of relative gene expression levels between LTBR and CXCR1, CXCR1 and CXCR2, LTBR and CXCR2, normalized to VCAM1 expression. Total PBMCs from RRMS patients who responded to IFNβ and those who did not respond to IFNβ were compared. All statistical analyses in this figure were performed by two-tailed unpaired Student’s t-test. All the experimental data sets, except for (g), are representatives from at least 2 similar experiments for each.

Supplementary Figure 5 CXCL1 expression in spinal cords.

Shown are representative images of typical CXCL1 staining in spinal cords from naïve mice and mice with either Type-A or Type-B EAE at 9-dpi. Scale bars, 200 μm. Images are representatives from 3 similar experiments.

Supplementary Figure 6 Histology of spinal cords in mice with type A or type B EAE.

(a-e) Shown are representative images of typical staining from multiple mice. (a) H/E staining in the spinal cord of Type-A and Type-B EAE mice at 70-dpi. (b) IGF-1 staining in spinal cord of naïve mice and mice with either Type-A or Type-B EAE at 22-dpi. (c) Bielschowsky neuron staining in spinal cords of naïve mice (e), and Type-A and Type-B EAE at 30-dpi. Red arrows indicate area showing reduced staining intensity in the spinal cord. (d,e) Golgi’s silver staining in spinal cord of naïve mice (d), and Type-A and Type-B at 22-dpi (e). All scale bars in this figure except magnified figure in (e), 200 μm. All scale bars in magnified figure in (e), 50 μm. All images are representatives from 3 similar experiments.

Supplementary Figure 7 Sema6b shRNA knockdown in T cells.

Sema6b expression levels in CD4+ T cells isolated from naïve or Type-B EAE mice at 9-dpi were evaluated by qPCR. Sema6b was knocked down by shRNA in the indicated group of CD4+ T cells. Control shRNA (scrambled shRNA sequence) was used for the control group. Values shown are obtained from three independent trials using one mouse per each. n=3. P(Naive T vs Type-B T+Control shRNA)=0.0011, t(4)=8.396, P(Type-B T+Control shRNA vs Type-B T+Sema6b shRNA)=0.0051, t(4)=5.559. *; p<0.05.

Supplementary Figure 8 Schematic diagram of distinct pathology of two EAE subtypes.

Type-A EAE is NLRP3 inflammasome-dependent and IFNβ-sensitive. Type-B EAE is induced by immunization with higher doses of Mtb than Type-A EAE induction, and is NLRP3 inflammasome-independent and IFNβ-resistant. Type-B EAE can also be induced with Type-A EAE induction methods with MHV68 infection or with rLT (rLTα2β1) injection. In Type-B EAE, Lta gene expression is epigenetically induced in DCs and the expression of membrane-bound LT (mLT) on DCs are enhanced. mLT stimulate LTβR on CD4+ T cells, resulting in the upregulation of CXCR2 on CD4+ T cells. Blockade of LTβR (with LTβR-Fc) and CXCR2 (with SB225002) selectively inhibits the Type-B EAE progression. mLT is also involved in the induction of Sema6B in T cells. Sema6B causes neural damages, and this may be a reason for the prolonged and minimal remission in Type-B EAE.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1–4 (PDF 1961 kb)

Supplementary Methods Checklist (PDF 838 kb)

Representative mouse with Type A EAE on 17-dpi.

This video shows a mouse with Type A EAE around the peak time. (MOV 31931 kb)

Representative mouse with Type B EAE on 17-dpi.

This video shows a mouse with Type B EAE around the peak time. There are no visible differences in movement and ataxia of this mouse compared to one with Type A EAE in Supp. Video 1. (MOV 42067 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Inoue, M., Chen, Ph., Siecinski, S. et al. An interferon-β-resistant and NLRP3 inflammasome–independent subtype of EAE with neuronal damage. Nat Neurosci 19, 1599–1609 (2016). https://doi.org/10.1038/nn.4421

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4421

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