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:

USP15 regulates type I interferon response and is required for pathogenesis of neuroinflammation

An Erratum to this article was published on 16 November 2016

This article has been updated

Abstract

Genes and pathways in which inactivation dampens tissue inflammation present new opportunities for understanding the pathogenesis of common human inflammatory diseases, including inflammatory bowel disease, rheumatoid arthritis and multiple sclerosis. We identified a mutation in the gene encoding the deubiquitination enzyme USP15 (Usp15L749R) that protected mice against both experimental cerebral malaria (ECM) induced by Plasmodium berghei and experimental autoimmune encephalomyelitis (EAE). Combining immunophenotyping and RNA sequencing in brain (ECM) and spinal cord (EAE) revealed that Usp15L749R-associated resistance to neuroinflammation was linked to dampened type I interferon responses in situ. In hematopoietic cells and in resident brain cells, USP15 was coexpressed with, and functionally acted together with the E3 ubiquitin ligase TRIM25 to positively regulate type I interferon responses and to promote pathogenesis during neuroinflammation. The USP15-TRIM25 dyad might be a potential target for intervention in acute or chronic states of neuroinflammation.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: An ENU-induced mutation in Usp15 protects mice against development of ECM.
Figure 2: Reduced protein expression and reduced stability of the USP15 L749 variant in vivo and in vitro.
Figure 3: Diminished cerebral pathogenesis of ECM and EAE in homozygous Usp15L749R mice.
Figure 4: Effect of USP15 on global gene expression during neuroinflammation of the brain and of the spinal cord.
Figure 5: Cell populations and associated molecular pathways regulated differentially in a USP15-dependent fashion.
Figure 6: USP15 modulates the type I interferon response through interaction with TRIM25.
Figure 7: Cellular compartments responsible for USP15-dependent effects in neuroinflammation and in type I interferon responses.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Change history

  • 24 October 2016

    In the version of this article initially published online, the symbol for the gene encoding granzyme B was incorrect (Gmzb) in the text in the third paragraph of the fourth subsection of Results and Figure 5d, and the symbol for the gene encoding granzyme A was incorrect (Gmza) in Figure 6h. These should be Gzmb and Gzma, respectively. The errors have been corrected for the print, PDF and HTML versions of this article.

References

  1. Hunt, N.H. & Grau, G.E. Cytokines: accelerators and brakes in the pathogenesis of cerebral malaria. Trends Immunol. 24, 491–499 (2003).

    CAS  PubMed  Google Scholar 

  2. Hansen, D.S. Inflammatory responses associated with the induction of cerebral malaria: lessons from experimental murine models. PLoS Pathog. 8, e1003045 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Brown, H. et al. Evidence of blood-brain barrier dysfunction in human cerebral malaria. Neuropathol. Appl. Neurobiol. 25, 331–340 (1999).

    CAS  PubMed  Google Scholar 

  4. de Souza, J.B. & Riley, E.M. Cerebral malaria: the contribution of studies in animal models to our understanding of immunopathogenesis. Microbes Infect. 4, 291–300 (2002).

    PubMed  Google Scholar 

  5. Ochiel, D.O. et al. Differential regulation of beta-chemokines in children with Plasmodium falciparum malaria. Infect. Immun. 73, 4190–4197 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Armah, H.B. et al. Cerebrospinal fluid and serum biomarkers of cerebral malaria mortality in Ghanaian children. Malar. J. 6, 147 (2007).

    PubMed  PubMed Central  Google Scholar 

  7. Kim, H. et al. Functional roles for C5a and C5aR but not C5L2 in the pathogenesis of human and experimental cerebral malaria. Infect. Immun. 82, 371–379 (2014).

    PubMed  PubMed Central  Google Scholar 

  8. Longley, R. et al. Host resistance to malaria: using mouse models to explore the host response. Mamm. Genome 22, 32–42 (2011).

    CAS  PubMed  Google Scholar 

  9. Senaldi, G. et al. Protection against the mortality associated with disease models mediated by TNF and IFN-γ in mice lacking IFN regulatory factor-1. J. Immunol. 163, 6820–6826 (1999).

    CAS  PubMed  Google Scholar 

  10. Berghout, J. et al. Irf8-regulated genomic responses drive pathological inflammation during cerebral malaria. PLoS Pathog. 9, e1003491 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Caignard, G. et al. Mouse ENU mutagenesis to understand immunity to infection: methods, selected examples, and perspectives. Genes (Basel) 5, 887–925 (2014).

    PubMed Central  Google Scholar 

  12. Bongfen, S.E. et al. An N-ethyl-N-nitrosourea (ENU)-induced dominant negative mutation in the JAK3 kinase protects against cerebral malaria. PLoS One 7, e31012 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Torre, S. et al. THEMIS is required for pathogenesis of cerebral malaria and protection against pulmonary tuberculosis. Infect. Immun. 83, 759–768 (2015).

    PubMed  PubMed Central  Google Scholar 

  14. Kennedy, J.M. et al. CCDC88B is a novel regulator of maturation and effector functions of T cells during pathological inflammation. J. Exp. Med. 211, 2519–2535 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Sawcer, S. et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 476, 214–219 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Dubois, P.C.A. et al. Multiple common variants for celiac disease influencing immune gene expression. Nat. Genet. 42, 295–302 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Beecham, A.H. et al. Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat. Genet. 45, 1353–1360 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Jostins, L. et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491, 119–124 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Okada, Y. et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 506, 376–381 (2014).

    CAS  PubMed  Google Scholar 

  20. Cunninghame Graham, D.S. et al. Association of NCF2, IKZF1, IRF8, IFIH1, and TYK2 with systemic lupus erythematosus. PLoS Genet. 7, e1002341 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Fodil, N., Langlais, D. & Gros, P. Primary immunodeficiencies and inflammatory disease: a growing genetic intersection. Trends Immunol. 37, 126–140 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Pauli, E.-K. et al. The ubiquitin-specific protease USP15 promotes RIG-I-mediated antiviral signaling by deubiquitylating TRIM25. Sci. Signal. 7, ra3 (2014).

    PubMed  PubMed Central  Google Scholar 

  23. Gack, M.U. et al. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature 446, 916–920 (2007).

    CAS  PubMed  Google Scholar 

  24. Torre, S. et al. Susceptibility to lethal cerebral malaria is regulated by epistatic interaction between chromosome 4 (Berr6) and chromosome 1 (Berr7) loci in mice. Genes Immun. 14, 249–257 (2013).

    CAS  PubMed  Google Scholar 

  25. de Jong, R.N. et al. Solution structure of the human ubiquitin-specific protease 15 DUSP domain. J. Biol. Chem. 281, 5026–5031 (2006).

    CAS  PubMed  Google Scholar 

  26. Hetfeld, B.K. et al. The zinc finger of the CSN-associated deubiquitinating enzyme USP15 is essential to rescue the E3 ligase Rbx1. Curr. Biol. 15, 1217–1221 (2005).

    CAS  PubMed  Google Scholar 

  27. Man, S., Ubogu, E.E. & Ransohoff, R.M. Inflammatory cell migration into the central nervous system: a few new twists on an old tale. Brain Pathol. 17, 243–250 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Rusinova, I. et al. Interferome v2.0: an updated database of annotated interferon-regulated genes. Nucleic Acids Res. 41, D1040–D1046 (2013).

    CAS  PubMed  Google Scholar 

  30. Schweitzer, K., Bozko, P.M., Dubiel, W. & Naumann, M. CSN controls NF-kappaB by deubiquitinylation of IkappaBalpha. EMBO J. 26, 1532–1541 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Cornelissen, T. et al. The deubiquitinase USP15 antagonizes Parkin-mediated mitochondrial ubiquitination and mitophagy. Hum. Mol. Genet. 23, 5227–5242 (2014).

    CAS  PubMed  Google Scholar 

  32. Hayes, S.D. et al. Direct and indirect control of mitogen-activated protein kinase pathway-associated components, BRAP/IMP E3 ubiquitin ligase and CRAF/RAF1 kinase, by the deubiquitylating enzyme USP15. J. Biol. Chem. 287, 43007–43018 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Villeneuve, N.F. et al. USP15 negatively regulates Nrf2 through deubiquitination of Keap1. Mol. Cell 51, 68–79 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Long, L. et al. The U4/U6 recycling factor SART3 has histone chaperone activity and associates with USP15 to regulate H2B deubiquitination. J. Biol. Chem. 289, 8916–8930 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Inui, M. et al. USP15 is a deubiquitylating enzyme for receptor-activated SMADs. Nat. Cell Biol. 13, 1368–1375 (2011).

    CAS  PubMed  Google Scholar 

  36. Eichhorn, P.J.A. et al. USP15 stabilizes TGF-β receptor I and promotes oncogenesis through the activation of TGF-β signaling in glioblastoma. Nat. Med. 18, 429–435 (2012).

    CAS  PubMed  Google Scholar 

  37. Iyengar, P.V. et al. USP15 regulates SMURF2 kinetics through C-lobe mediated deubiquitination. Sci. Rep. 5, 14733 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Zou, Q. et al. USP15 stabilizes MDM2 to mediate cancer-cell survival and inhibit antitumor T cell responses. Nat. Immunol. 15, 562–570 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang, H. et al. Ubiquitin-specific protease 15 negatively regulates virus-induced type I interferon signaling via catalytically-dependent and -independent mechanisms. Sci Rep. 5, 11220 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Inn, K.-S. et al. Linear ubiquitin assembly complex negatively regulates RIG-I- and TRIM25-mediated type I interferon induction. Mol. Cell 41, 354–365 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Liehl, P. et al. Host-cell sensors for Plasmodium activate innate immunity against liver-stage infection. Nat. Med. 20, 47–53 (2014).

    CAS  PubMed  Google Scholar 

  42. Gazzinelli, R.T., Kalantari, P., Fitzgerald, K.A. & Golenbock, D.T. Innate sensing of malaria parasites. Nat. Rev. Immunol. 14, 744–757 (2014).

    CAS  PubMed  Google Scholar 

  43. Miller, J.L., Sack, B.K., Baldwin, M., Vaughan, A.M. & Kappe, S.H.I. Interferon-mediated innate immune responses against malaria parasite liver stages. Cell Rep. 7, 436–447 (2014).

    CAS  PubMed  Google Scholar 

  44. Ball, E.A. et al. IFNAR1 controls progression to cerebral malaria in children and CD8+ T cell brain pathology in Plasmodium berghei-infected mice. J. Immunol. 190, 5118–5127 (2013).

    CAS  PubMed  Google Scholar 

  45. Palomo, J. et al. Type I interferons contribute to experimental cerebral malaria development in response to sporozoite or blood-stage Plasmodium berghei ANKA. Eur. J. Immunol. 43, 2683–2695 (2013).

    CAS  PubMed  Google Scholar 

  46. Sharma, S. et al. Innate immune recognition of an AT-rich stem-loop DNA motif in the Plasmodium falciparum genome. Immunity 35, 194–207 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Coban, C. et al. Pathological role of Toll-like receptor signaling in cerebral malaria. Int. Immunol. 19, 67–79 (2007).

    CAS  PubMed  Google Scholar 

  48. Togbe, D. et al. Murine cerebral malaria development is independent of toll-like receptor signaling. Am. J. Pathol. 170, 1640–1648 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Imboden, M. et al. Genome-wide association study of lung function decline in adults with and without asthma. J. Allergy Clin. Immunol. 129, 1218–1228 (2012).

    PubMed  PubMed Central  Google Scholar 

  50. Orimo, A. et al. Underdeveloped uterus and reduced estrogen responsiveness in mice with disruption of the estrogen-responsive finger protein gene, which is a direct target of estrogen receptor alpha. Proc. Natl. Acad. Sci. USA 96, 12027–12032 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Urano, T. et al. Efp targets 14-3-3 sigma for proteolysis and promotes breast tumour growth. Nature 417, 871–875 (2002).

    CAS  PubMed  Google Scholar 

  52. Tanaka, K. et al. Loss of suppressor of cytokine signaling 1 in helper T cells leads to defective Th17 differentiation by enhancing antagonistic effects of IFN-gamma on STAT3 and Smads. J. Immunol. 180, 3746–3756 (2008).

    CAS  PubMed  Google Scholar 

  53. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Quinlan, A.R. & Hall, I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Gold, R., Linington, C. & Lassmann, H. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 129, 1953–1971 (2006).

    PubMed  Google Scholar 

  56. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    PubMed  PubMed Central  Google Scholar 

  57. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome Biol. 14, R36 (2013).

    PubMed  PubMed Central  Google Scholar 

  58. Liao, Y., Smyth, G.K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  59. Robinson, M.D. & Oshlack, A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 11, R25 (2010).

    PubMed  PubMed Central  Google Scholar 

  60. Thorvaldsdóttir, H., Robinson, J.T. & Mesirov, J.P. Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration. Brief. Bioinform. 14, 178–192 (2013).

    PubMed  Google Scholar 

  61. Saeed, A.I. et al. TM4: a free, open-source system for microarray data management and analysis. Biotechniques 34, 374–378 (2003).

    CAS  PubMed  Google Scholar 

  62. Huang, W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    CAS  Google Scholar 

  63. Saikali, P. et al. NKG2D-mediated cytotoxicity toward oligodendrocytes suggests a mechanism for tissue injury in multiple sclerosis. J. Neurosci. 27, 1220–1228 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Durafourt, B.A. et al. Comparison of polarization properties of human adult microglia and blood-derived macrophages. Glia 60, 717–727 (2012).

    PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Van Bruggen, S. Gauthier, P. D'Arcy and G. Perreault for assistance in the ENU project, and C. Meunier for technical assistance. Supported by the Canadian Institutes of Health Research (MOP119342 and MOP133487 to P.G. and S.V.) and Amorchem (PT63088).

Author information

Authors and Affiliations

Authors

Contributions

S.T., M.L., J.M. and J.B. contributed to mutation identification. S.T. performed all of the PbA experiments. M.J.P. performed all of the EAE experiments. S.T. and M.J.P. performed biochemical work. I.R., J.M.K. and S.T. performed the immunophenotyping experiments. D.L. performed RNA sequencing analyses, and S.T. carried out validations by RT-qPCR. Primary cells from brain were provided and characterized by J.A., N.A., A.P., M.J.P. and L.M.H., with additional contribution from G.A.L.-T., S.I., K.M., C.L. and K.P.K. kindly provided gene knockout animals. S.T. performed Listeria experiments with guidance from C.M.K. N.F. performed BM transplant experiments. J.B., J.M. and M.L. performed analyses of exome sequences. P.G. and S.M.V. supervised the project, helped to design experiments and analyzed data. P.G., S.T., D.L., M.J.P. and N.F. wrote the first draft of the manuscript. All of the authors provided helpful comments on the manuscript.

Corresponding author

Correspondence to Philippe Gros.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Ubiquitous pattern of Usp15 mRNA expression in embryonic, post-natal and adult mice

Mouse sections were stained with cresyl violet to localize Usp15 RNA to specific organs and structures. In situ hybridization was carried out using radiolabelled antisense (as) and sense (s) probes. The results shown are from X-ray film autoradiography obtained following 5-days exposure. Non-specific localized signals (visible with sense and anti-sense probes) are indicated with an asterisk (*); in the teeth (p10) and the large intestine lumen (p10 and adult). (Magnification: Embryonic x2.4, Post-natal x3, Adult x2.4). Abbreviations: Adr–adrenal gland; At–heart atrium; Br–brain; Bro–bronchcus; Car– cartilage; Cb–cerebellum; Co–colon; Cx–cerebral cortex; Du – duodenum; E – eye; Ep – epididymis; Es – esophagus; GB – gallbladder; HV–heart ventricle; Il–ileum; Je–jejunum; Ki–kidney; Li–liver; LI–large intestine; Lu–lung; OL–olfactory lobe; Ov–ovary; Ovi–oviducts; PB–pelvis bone; Pc–pancreas; PG–pituitary gland; Pr–prostate; PTh–parathyroid gland; R–ribs; Sk–skin; Spl–spleen; St–stomach; SV–seminal vesicle; Te–testis; Th–thyroid gland; UB–urinary bladder; Ut-uterus; CA–central artery; GC-germinal center; LN–lymphatic nodule; RP–red pulp; Tr–trabeculum; V-vein; LF–lymphoid follicle; Me–medulla; MG–mammary glands; Cx–cortex.

Supplementary Figure 2 Reduced protein stability of the USP15 L720R human variant in vitro

HEK293 cells stably expressing HA-tagged WT or USP15L720R proteins were treated with cycloheximide (CHX, 20 μg/ml) for 2, 4, 8, and 16h, and equal amounts of protein were analyzed by immunoblotting. Data is from a single experiment.

Supplementary Figure 3 Immunophenotyping of Usp15L749R mutants at steady-state and following P. berghei ANKA infection

(a) The number and proportions of different spleen cell types from naïve and from day 5-PbA infected animals, were established by flow cytometry with markers for T cells (CD4, CD8), B cells (B220), NK cells (NK1.1), monocytes and neutrophils (CD11b, Ly6G). Results are pooled from 5 independent experiments. (b) The percentage of splenic CD4+ and CD8+ effector T cells (CD62LCD44+) were also assessed. Data represents a single experiment with 5 mice per group, and are expressed as a mean ± SD. (c-d) Cells were re-stimulated in vitro with either media alone (unstimulated, US), with anti-CD3 and anti-CD28 (TCR engagement), with PMA/Ionomycin, with CpG, or with Poly:IC and cytokine production was assessed by flow cytometry (C, intracellular staining), or by ELISA (D, culture supernatants). Data is a representation of two independent experiments with 5 mice per group, and is expressed as a mean ± SD. (e) The activation state of CD4+ and CD8+ T cells were assessed by analysis of CD69 cell surface expression in response to TCR engagement (anti-CD3/anti-CD28). Data represents a single experiment with 5 mice per group, and is expressed as a mean ± SD.

Supplementary Figure 4 USP15 negatively regulates CD4+ T cell activation during Listeria monocytogenes infection

Wild type B6 mice and Usp15L749R mutants infected with 1x104 CFU of Listeria monocytogenes (strain 10403s) expressing ovalbumin (OVA) were sacrificed on day 7 post-infection, and phenotyped for the activation of the T cell response in spleen cell populations. (a, b) CD44 expression (T cell activation) on CD4+ T cells (A), or CD8+ T cells (B), expressed as percentage and total cell numbers. (c, d) Cells were re-stimulated in vitro with Listeria-specific antigens, LLO or OVA, and IFN-γ production was assessed by flow cytometry (C, intracellular staining), or by ELISA (D, culture supernatants) for CD4+ and CD8+ T cells. (e) Serum IFN-γ levels were measured by ELISA, and plotted as optical density absorbance (OD) at 450 nm. (a-e) Data is a combination of two independent experiments. All data are expressed as a mean ± SD for each group, and all statistical analyses were performed using the two-tailed unpaired Student’s t-test.

Supplementary Figure 5 Cell populations and associated molecular pathways differentially regulated in a USP15-dependent fashion

(a) LEA dendogram for genes with reduced expression in Usp15L749R mutant mice compared to B6 (day 5 post-PbA infection) and that drive significant enrichment (FDR<0.01) of immunological expression signatures (GSEA). Enriched immunological signatures and functions are highlighted by color boxes: red = signatures of IFN activation, green = myeloid signatures and responses, and purple = T cell signatures. Refer to Online Methods for details on LEA analysis. (b) LEA clustering analysis as described in (A) for immunological signatures depleted in Usp15L749R mutant mice during EAE neuroinflammation progression.

Supplementary Figure 6 Mouse mutants bearing a loss of function mutation in Irf3 are protected against neuroinflammation

Survival plots for PbA-infected (a) Irf3 mutants (Irf3-/-) (n=13) and B6 controls (n=8), and (b) Mavs mutants (Mavs-/-) (n=22) and B6 (n=11). Statistical significance for survival between groups of mice was determined by the Log-rank test (* P<0.05, **** P<0.0001).

Supplementary Figure 7 The L720R mutation affects the ability of USP15 to deubiquitinate SMURF2

HEK293T cells transiently expressing Smurf2-FLAG with or without Usp15-Xpress construct plus HA-tagged ubiquitin (Ub) were lysed 48h post-transfection, SMURF2 was immunoprecipitated using anti-FLAG antibody and immunoblotting analysis was performed as indicated. Construct expression in whole cell lysates (WCL) was confirmed by western blot

Supplementary Figure 8 Full gating strategy for flow cytometry analysis of brain cellular infiltration

Five days post-PbA infection, infiltrating cells were isolated from perfused brains and stained for flow cytometry analyses. Viable cells were selected for based on their exclusion of the Zombie Aqua Dye. Leukocytes were gated as CD45hi cells. Populations of leukocytes gated from the CD45hi gate were analyzed as follows: CD4 T cells (TCRb+CD4+CD8), CD8 T cells (TCRb+CD4CD8+), macrophages (Cd11b+F4/80+) and neutrophils (Cd11b+Ly6G+).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 1128 kb)

Supplementary Table 1

List of dys-regulated genes in Usp15L749R mutant mice undergoing ECM and EAE models of neuroinflammatory diseases, related to Fig. 4b. (XLSX 120 kb)

Supplementary Table 2

Detailed matrix of leading edge analysis clustering performed on ECM and EAE depleted gene signatures in Usp15L749R mutant mice, related to Fig. 5a. (XLSX 462 kb)

Supplementary Table 3

qPCR validation primers, related to Fig. 5c-f. (XLSX 780 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Torre, S., Polyak, M., Langlais, D. et al. USP15 regulates type I interferon response and is required for pathogenesis of neuroinflammation. Nat Immunol 18, 54–63 (2017). https://doi.org/10.1038/ni.3581

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3581

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