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:

Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity

Abstract

Polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) have been characterized in the context of malignancies. Here we show that PMN-MDSCs can restrain B cell accumulation during central nervous system (CNS) autoimmunity. Ly6G+ cells were recruited to the CNS during experimental autoimmune encephalomyelitis (EAE), interacted with B cells that produced the cytokines GM-CSF and interleukin-6 (IL-6), and acquired properties of PMN-MDSCs in the CNS in a manner dependent on the signal transducer STAT3. Depletion of Ly6G+ cells or dysfunction of Ly6G+ cells through conditional ablation of STAT3 led to the selective accumulation of GM-CSF-producing B cells in the CNS compartment, which in turn promoted an activated microglial phenotype and lack of recovery from EAE. The frequency of CD138+ B cells in the cerebrospinal fluid (CSF) of human subjects with multiple sclerosis was negatively correlated with the frequency of PMN-MDSCs in the CSF. Thus PMN-MDSCs might selectively control the accumulation and cytokine secretion of B cells in the inflamed CNS.

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

Fig. 1: LOX1+ PMN-MDSCs in the CSF are negatively correlated with intrathecal CD138+ B cells in subjects with MS and indicate stable disease.
Fig. 2: Population dynamics and phenotype of Ly6G+ neutrophils in different compartments in the course of EAE.
Fig. 3: In the EAE model, the generation of PMN-MDSCs is restricted to the inflamed CNS.
Fig. 4: PMN-MDSCs contribute to inducing recovery from EAE.
Fig. 5: Ly6G+ cells differentiate into MDSCs in the CNS in a STAT3-dependent manner.
Fig. 6: Ly6G+ cells interact with B cells in the CNS.
Fig. 7: B cells determine disease progression in the absence of functional MDSCs.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited in the European Nucleotide Archive under accession code PRJEB28339. All data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Lappat, E. J. & Cawein, M. A study of the leukemoid response to transplantable A-280 tumor in mice. Cancer Res. 24, 302–311 (1964).

    CAS  PubMed  Google Scholar 

  2. Bronte, V. et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat. Commun. 7, 12150 (2016).

    Article  CAS  Google Scholar 

  3. Condamine, T. et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci. Immunol. 1, aaf8943 (2016).

    Article  Google Scholar 

  4. Dufait, I. et al. Signal transducer and activator of transcription 3 in myeloid-derived suppressor cells: an opportunity for cancer therapy. Oncotarget 7, 42698–42715 (2016).

    Article  Google Scholar 

  5. Lelis, F. J. N. et al. Myeloid-derived suppressor cells modulate B-cell responses. Immunol. Lett. 188, 108–115 (2017).

    Article  CAS  Google Scholar 

  6. Li, Y. et al. Myeloid-derived suppressor cells as a potential therapy for experimental autoimmune myasthenia gravis. J. Immunol. 193, 2127–2134 (2014).

    Article  CAS  Google Scholar 

  7. Park, M.-J. et al. Myeloid-derived suppressor cells induce the expansion of regulatory B cells and ameliorate autoimmunity in the sanroque mouse model of systemic lupus erythematosus. Arthritis Rheumatol. 68, 2717–2727 (2016).

    Article  CAS  Google Scholar 

  8. Xu, X. et al. Myeloid-derived suppressor cells promote B-cell production of IgA in a TNFR2-dependent manner. Cell. Mol. Immunol. 14, 597–606 (2017).

    Article  CAS  Google Scholar 

  9. Wang, C. et al. Myeloid-derived suppressor cells inhibit T follicular helper cell immune response in Japanese encephalitis virus infection. J. Immunol. 199, 3094–3105 (2017).

    Article  CAS  Google Scholar 

  10. Flach, A.-C. et al. Autoantibody-boosted T-cell reactivation in the target organ triggers manifestation of autoimmune CNS disease. Proc. Natl Acad. Sci. USA 113, 3323–3328 (2016).

    Article  CAS  Google Scholar 

  11. Molnarfi, N. et al. MHC class II-dependent B cell APC function is required for induction of CNS autoimmunity independent of myelin-specific antibodies. J. Exp. Med. 210, 2921–2937 (2013).

    Article  CAS  Google Scholar 

  12. Hauser, S. L. et al. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N. Engl. J. Med. 358, 676–688 (2008).

    Article  CAS  Google Scholar 

  13. Li, R. et al. Proinflammatory GM-CSF-producing B cells in multiple sclerosis and B cell depletion therapy. Sci. Transl. Med. 7, 310ra166 (2015).

    Article  Google Scholar 

  14. 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  Google Scholar 

  15. Gotot, J. et al. Regulatory T cells use programmed death 1 ligands to directly suppress autoreactive B cells in vivo. Proc. Natl Acad. Sci. USA 109, 10468–10473 (2012).

    Article  CAS  Google Scholar 

  16. Nakamura, K., Kitani, A. & Strober, W. Cell contact-dependent immunosuppression by CD4( + )CD25( + ) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J. Exp. Med. 194, 629–644 (2001).

    Article  CAS  Google Scholar 

  17. Jang, E. et al. Foxp3 + regulatory T cells control humoral autoimmunity by suppressing the development of long-lived plasma cells. J. Immunol. 186, 1546–1553 (2011).

    Article  CAS  Google Scholar 

  18. Koutrolos, M., Berer, K., Kawakami, N., Wekerle, H. & Krishnamoorthy, G. Treg cells mediate recovery from EAE by controlling effector T cell proliferation and motility in the CNS. Acta Neuropathol. Commun. 2, 163 (2014).

    Article  Google Scholar 

  19. Bennett, J. L. et al. B lymphocytes in neuromyelitis optica. Neurol. Neuroimmunol. Neuroinflamm. 2, e104–e104 (2015).

    Article  Google Scholar 

  20. Cepok, S. et al. Short-lived plasma blasts are the main B cell effector subset during the course of multiple sclerosis. Brain 128, 1667–1676 (2005).

    Article  Google Scholar 

  21. 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).

    Article  Google Scholar 

  22. Hasenberg, A. et al. Catchup: a mouse model for imaging-based tracking and modulation of neutrophil granulocytes. Nat. Methods 12, 445–452 (2015).

    Article  CAS  Google Scholar 

  23. Marigo, I. et al. Tumor-induced tolerance and immune suppression depend on the C/EBPbeta transcription factor. Immunity 32, 790–802 (2010).

    Article  CAS  Google Scholar 

  24. Durant, L. et al. Diverse targets of the transcription factor STAT3 contribute to T cell pathogenicity and homeostasis. Immunity 32, 605–615 (2010).

    Article  CAS  Google Scholar 

  25. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9, 162–174 (2009).

    Article  CAS  Google Scholar 

  26. Veglia, F., Perego, M. & Gabrilovich, D. Myeloid-derived suppressor cells coming of age. Nat. Immunol. 19, 108–119 (2018).

    Article  CAS  Google Scholar 

  27. Yanaba, K. et al. A regulatory B cell subset with a unique CD1dhiCD5 + phenotype controls T cell-dependent inflammatory responses. Immunity 28, 639–650 (2008).

    Article  CAS  Google Scholar 

  28. Rauch, P. J. et al. Innate response activator B cells protect against microbial sepsis. Science 335, 597–601 (2012).

    Article  CAS  Google Scholar 

  29. Litzenburger, T. et al. B lymphocytes producing demyelinating autoantibodies: development and function in gene-targeted transgenic mice. J. Exp. Med. 188, 169–180 (1998).

    Article  CAS  Google Scholar 

  30. Bettelli, E. et al. Myelin oligodendrocyte glycoprotein-specific T cell receptor transgenic mice develop spontaneous autoimmune optic neuritis. J. Exp. Med. 197, 1073–1081 (2003).

    Article  CAS  Google Scholar 

  31. Weber, M. S. et al. B-cell activation influences T-cell polarization and outcome of anti-CD20 B-cell depletion in central nervous system autoimmunity. Ann. Neurol. 68, 369–383 (2010).

    Article  CAS  Google Scholar 

  32. Krasemann, S. et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neurodegenerative diseases. Immunity 47, 566–581.e9 (2017).

    Article  CAS  Google Scholar 

  33. Tak, T., Tesselaar, K., Pillay, J., Borghans, J. A. M. & Koenderman, L. What’s your age again? Determination of human neutrophil half-lives revisited. J. Leukoc. Biol. 94, 595–601 (2013).

    Article  CAS  Google Scholar 

  34. Pierson, E. R., Wagner, C. A. & Goverman, J. M. The contribution of neutrophils to CNS autoimmunity. Clin. Immunol. 189, 23–28 (2018).

    Article  CAS  Google Scholar 

  35. Lévesque, S. A. et al. Myeloid cell transmigration across the CNS vasculature triggers IL-1β-driven neuroinflammation during autoimmune encephalomyelitis in mice. J. Exp. Med. 213, 929–949 (2016).

    Article  Google Scholar 

  36. Steinbach, K., Piedavent, M., Bauer, S., Neumann, J. T. & Friese, M. A. Neutrophils amplify autoimmune central nervous system infiltrates by maturing local APCs. J. Immunol. 191, 4531–4539 (2013).

    Article  CAS  Google Scholar 

  37. Soulika, A. M. et al. Initiation and progression of axonopathy in experimental autoimmune encephalomyelitis. J. Neurosci. 29, 14965–14979 (2009).

    Article  CAS  Google Scholar 

  38. Cheretakis, C., Leung, R., Sun, C. X., Dror, Y. & Glogauer, M. Timing of neutrophil tissue repopulation predicts restoration of innate immune protection in a murine bone marrow transplantation model. Blood 108, 2821–2826 (2006).

    Article  CAS  Google Scholar 

  39. Carlson, T., Kroenke, M., Rao, P., Lane, T. E. & Segal, B. The Th17-ELR + CXC chemokine pathway is essential for the development of central nervous system autoimmune disease. J. Exp. Med. 205, 811–823 (2008).

    Article  CAS  Google Scholar 

  40. Bartosik-Psujek, H. & Stelmasiak, Z. The levels of chemokines CXCL8, CCL2 and CCL5 in multiple sclerosis patients are linked to the activity of the disease. Eur. J. Neurol. 12, 49–54 (2005).

    Article  CAS  Google Scholar 

  41. Rumble, J. M. et al. Neutrophil-related factors as biomarkers in EAE and MS. J. Exp. Med. 212, 23–35 (2015).

    Article  CAS  Google Scholar 

  42. Däbritz, J., Judd, L. M., Chalinor, H. V., Menheniott, T. R. & Giraud, A. S. Altered gp130 signalling ameliorates experimental colitis via myeloid cell-specific STAT3 activation and myeloid-derived suppressor cells. Sci. Rep. 6, 20584 (2016).

    Article  Google Scholar 

  43. Arocena, A. R. et al. Myeloid-derived suppressor cells are key players in the resolution of inflammation during a model of acute infection. Eur. J. Immunol. 44, 184–194 (2014).

    Article  CAS  Google Scholar 

  44. Heink, S. et al. Trans-presentation of IL-6 by dendritic cells is required for the priming of pathogenic TH17 cells. Nat. Immunol. 18, 74–85 (2017).

    Article  CAS  Google Scholar 

  45. Zehntner, S. P. et al. Neutrophils that infiltrate the central nervous system regulate T cell responses. J. Immunol. 174, 5124–5131 (2005).

    Article  CAS  Google Scholar 

  46. Hjelmström, P., Juedes, A. E., Fjell, J. & Ruddle, N. H. B-cell-deficient mice develop experimental allergic encephalomyelitis with demyelination after myelin oligodendrocyte glycoprotein sensitization. J. Immunol. 161, 4480–4483 (1998).

    PubMed  Google Scholar 

  47. Magliozzi, R., Columba-Cabezas, S., Serafini, B. & Aloisi, F. Intracerebral expression of CXCL13 and BAFF is accompanied by formation of lymphoid follicle-like structures in the meninges of mice with relapsing experimental autoimmune encephalomyelitis. J. Neuroimmunol. 148, 11–23 (2004).

    Article  CAS  Google Scholar 

  48. Matsushita, T., Horikawa, M., Iwata, Y. & Tedder, T. F. Regulatory B cells (B10 cells) and regulatory T cells have independent roles in controlling experimental autoimmune encephalomyelitis initiation and late-phase immunopathogenesis. J. Immunol. 185, 2240–2252 (2010).

    Article  CAS  Google Scholar 

  49. Shen, P. et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 507, 366–370 (2014).

    Article  CAS  Google Scholar 

  50. Pöllinger, B. et al. Spontaneous relapsing-remitting EAE in the SJL/J mouse: MOG-reactive transgenic T cells recruit endogenous MOG-specific B cells. J. Exp. Med. 206, 1303–1316 (2009).

    Article  Google Scholar 

  51. Polman, C. H. et al. Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann. Neurol. 69, 292–302 (2011).

    Article  Google Scholar 

  52. Shen, F. W. et al. Cloning of Ly-5 cDNA. Proc. Natl Acad. Sci. USA 82, 7360–7363 (1985).

    Article  CAS  Google Scholar 

  53. Betz, U. A. et al. Postnatally induced inactivation of gp130 in mice results in neurological, cardiac, hematopoietic, immunological, hepatic, and pulmonary defects. J. Exp. Med. 188, 1955–1965 (1998).

    Article  CAS  Google Scholar 

  54. Takeda, K. et al. Stat3 activation is responsible for IL-6-dependent T cell proliferation through preventing apoptosis: generation and characterization of T cell-specific Stat3-deficient mice. J. Immunol. 161, 4652–4660 (1998).

    CAS  PubMed  Google Scholar 

  55. Wunderlich, F. T. et al. Interleukin-6 signaling in liver-parenchymal cells suppresses hepatic inflammation and improves systemic insulin action. Cell. Metab. 12, 237–249 (2010).

    Article  CAS  Google Scholar 

  56. Anderson, K. G. et al. Intravascular staining for discrimination of vascular and tissue leukocytes. Nat. Protoc. 9, 209–222 (2014).

    Article  CAS  Google Scholar 

  57. Parekh, S., Ziegenhain, C., Vieth, B., Enard, W. & Hellmann, I. The impact of amplification on differential expression analyses by RNA-seq. Sci. Rep. 6, 25533 (2016).

    Article  CAS  Google Scholar 

  58. Macosko, E. Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell 161, 1202–1214 (2015).

    Article  CAS  Google Scholar 

  59. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome. Biol. 15, 550 (2014).

    Article  Google Scholar 

  60. Eden, E., Navon, R., Steinfeld, I., Lipson, D. & Yakhini, Z. GOrilla: a tool for discovery and visualization of enriched GO terms in ranked gene lists. BMC Bioinformatics 10, 48 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

We thank all members of the Korn Group and especially V. Husterer for her skillful technical assistance. This work was supported by the Deutsche Forschungsgemeinschaft (SFB1054-B06 to T.K., TRR128 to T.K., SyNergy to T.K. and Kompetenznetz Multiple Sklerose KKNMS to B.K.), the German Ministry of Education and Research (BMBF, T-B in NMO to T.K.,) and by the ERC (CoG 647215 to T.K.). B.K. received intramural funding from the Technical University of Munich. L.A. is supported by a clinical scientist program provided by the Deutsche Forschungsgemeinschaft (Synergy). D.M. is supported by the Swiss National Science Foundation.

Author information

Authors and Affiliations

Authors

Contributions

B.K. conceptualized parts of the study, performed most of the experiments, analyzed data and wrote the manuscript. M.H., C.S., L.A., G.G., A.M. and M.M. performed experiments and analyzed data. G.L., T.E. and R.R. performed and analyzed the RNA-seq experiments. U.K. developed the CSF drainage technique and performed certain experiments. D.M. performed histology and analyzed data. B.Höchst, P.K., M.G. and B.Hemmer designed experiments and analyzed data. T.K. conceptualized and directed the study, supervised the experiments, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Thomas Korn.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 LOX1+ MDSCs and disease activity in MS.

a, Correlation of CD15+CD11bhiCD33loLOX1lo myeloid derived suppressor cells (MDSCs) and total CD15+ neutrophils with CD138+HLA-DR+ B cells in CSF of therapy-naive subjects with relapsing-remitting MS (RRMS) or clinically isolated syndrome (CIS); not significant (ns); Spearman’s r analysis. b, Demographic and disease specific features of subjects with RRMS or CIS who served as donors for PBMCs (PBMC cohort, n = 70), CSF (CSF cohort, n = 25) and healthy controls (n = 31); NA, data not available; mean ± s.d. c–e, Individual subject information of the RRMS/CIS subject cohort used for paired analysis of the frequency of PMN-MDSCs in blood samples during relapse and after recovery from relapse in a state of either active disease showing evidence of disease activity either by relapse and/or disease activity in MRI and/or worsening of EDSS score ≥ 1 point (no NEDA-3) (d) or in subjects with no signs of disease activity defined by the absence of relapse, absence of disease activity in MRI and stable EDSS values (no evidence of disease activity, NEDA-3) (e); fingolimod (Fingoli.), ocrelizumab (Ocreli.), natalizumab (Natali.), pegylated interferon β-1a (peg-IGN-β), teriflunomide (Teriflu), glatiramer acetate (GA); Wilcoxon matched-pairs signed rank test, *P < 0.05.

Supplementary Figure 2 Transcriptome (RNA-seq) of CNS-onset Ly6G+ cells and CNS-recovery Ly6G+ cells.

a,b, Waterfall plot of log2 fold change in gene expression comparing CNS-onset Ly6GtdTomato+ cells versus spleen-onset Ly6G-tdTomato+ cells (a) and CNS-recovery Ly6G-tdTomato+ cells versus spleen-onset Ly6G-tdTomato+ cells (b). The positions of Nos2 (inducible NO synthetase) and Arg1 (arginase 1) are indicated. c,d, Heatmaps of gene expression in Ly6G-tdTomato+ subsets for top three significantly enriched gene ontology terms (GOrilla) in universally downregulated (c) and upregulated (d) genes in CNS-onset Ly6G-tdTomato+ cells versus all other Ly6G-tdTomato+ cell subsets (see Fig. 3a).

Supplementary Figure 3 Depletion efficiency of Ly6G-tdTomato+ cells using a monoclonal antibody to Ly6G.

a, Fraction of SSChi Ly6G-tdTomato+ cells in peripheral blood of naive Ly6gCre/WT mice one day after i.p. application of 400 μg monoclonal antibody to Ly6G (clone 1A8) or 400 μg of a rat-IgG2a control antibody (2A3); gate on peripheral blood cells after erythrocyte lysis. b, Fraction of SSChi Ly6G-tdTomato+ cells in peripheral blood of Ly6gCre/WT mice during EAE; mice were treated with 400 μg anti-Ly6G (Ly6G Ab, n = 5) or 400 μg rat-IgG2a control antibody (rat IgG2a, n = 5) every other day starting on day 5 after immunization; symbols depict mean ± s.d. of biological replicates; two-way ANOVA with Bonferroni’s multiple comparison test; ***P < 0.001. c, Fraction of CD45.2intermedLy6G-tdTomato+ cells purified from brain of Ly6gCre/WT mice at early EAE recovery (day 22) treated with 400 μg monoclonal antibody to Ly6G i.p. (clone 1A8, Bio X Cell) or 400 μg of a rat-IgG2a control antibody (clone 2A3) every other day starting at disease onset; representative plot of ten biological replicates from each group, repeated in three independent experiments; gate on live CD45.2+CD11b+ cells.

Supplementary Figure 4 The T cell and myeloid compartments are unaltered in the absence of Ly6G-tdTomato+ cells during recovery from EAE.

a, Total cell count of CD45+ cells, CD4+ T cells, Ly6G-tdTomato+ cells, CD45hiCD11bhiLy6G monocytes and CD45dimCD11bhi microglia, purified from the spinal cord of Ly6gCre/WT mice at early EAE recovery (day 21) treated with control antibody (rat IgG2a, clone 2A3, n = 5) or anti-Ly6G (Ly6G Ab, clone 1A8, n = 5) starting on day 12 after immunization; for gating see s. b, Cell counts of immune cell subsets purified from the spinal cord of Ly6gCre/WT mice at early EAE recovery (day 21) treated with 200 μg kg–1 G-CSF (n = 5) or 5% glucose (control, n = 5) i.p. every other day, starting on day 12 after immunization; for gating see d. c, Analysis of cytokine production by intracellular cytokine staining in CD4+ T cells purified from spinal cord of Ly6gCre/WT mice treated with control antibody (rat IgG2a, n = 4) or anti-Ly6G (Ly6G Ab, n = 4) at early EAE recovery (day 23) and stimulated ex vivo with PMA/ionomycin. Symbols depict mean ± s.d. of biological replicates; Mann-Whitney U-test; **P < 0.01 (a–c). d, Gating strategy to identify different cell subsets purified from spinal cord of Ly6gCre/WT mice at early EAE recovery (day 21); representative plot of >30 mice, gate on analyzed single cells.

Supplementary Figure 5 Effect of Ly6G conditional ablation of Il6ra and Il6st (gp130) on the disease course of EAE.

a, Course of EAE in Ly6gCre/WT control (n = 11) and Il6raΔLy6G (n = 11) mice; symbols depict mean ± s.d. of EAE scores in individual mice; two-way ANOVA with Bonferroni’s multiple comparison test; ns, not significant; representative disease course. The experiment was repeated four times. b, Course of EAE in Ly6gCre/WT (n = 13) and Il6stΔLy6G (n = 19) mice; symbols depict mean ± s.d. of EAE scores in individual mice; two-way ANOVA with Bonferroni’s multiple comparison test; *P < 0.05; representative disease course; the experiment was repeated two times.

Supplementary Figure 6 STAT3 deficiency in Ly6G+ neutrophils does not alter the T cell and myeloid compartments during recovery from EAE.

a, Total cell count of CD45+ cells, CD4+ T cells, Ly6G-tdTomato+ cells, CD45hiCD11bhiLy6G monocytes and CD45dimCD11bhi microglia, purified from the spinal cord of Ly6gCre/WT (n = 6) or Stat3ΔLy6G mice (n = 10) at EAE recovery (day 25); for gating strategy see Supplementary Fig. 4. b, Analysis of cytokine production by intracellular cytokine staining in CD4+ T cells, purified from spinal cord of Ly6gCre/WT (n = 4) or Stat3ΔLy6G mice (n = 5) at EAE recovery (day 24) and stimulated ex vivo with PMA/ionomycin. Symbols depict mean ± s.d. of biological replicates; Mann-Whitney U-test (a,b).

Supplementary Figure 7 Characterization of B cells in spleen and CNS of mice lacking functional Ly6G+ MDSCs during recovery from EAE.

a, Histologic analysis showing co-staining of CD3, CD19 and B220 in control mice (Ly6gCre/WT) and Stat3ΔLy6G mice during early recovery (day 21). Scale bar, 20 μm. b, Flow cytometric analysis of live CD19+ B cell compartment in spleen and CNS of control mice (Ly6gCre/WT), Stat3ΔLy6G mice and Ly6G-tdTomato+ MDSC-depleted mice (Ly6G Ab) during early recovery from EAE. CD1+CD5+ regulatory B cells (Breg cells) are not increased upon MDSC loss of function; representative plots of n = 6 individuals per group; gate on CD19+B220+ cells. c, Analysis of cytokine production by intracellular cytokine staining in CD19+B220+ B cells purified from CNS of Ly6gCre/WT (n = 6), Stat3ΔLy6G mice (n = 6) and Ly6gCre/WT mice treated with anti-Ly6G depleting antibody (Ly6G Ab, n = 6) at EAE recovery (day 21) and stimulated ex vivo with PMA/ionomycin in the presence of brefeldin A; symbols depict mean ± s.d. of biological replicates; Kruskal-Wallis test with Dunn’s post test; *P < 0.05, ***P < 0.001. d, Representative cytograms of live CD19+ B cells isolated from the CNS of Ly6gCre/WT control mice or Stat3ΔLy6G mice (with dysfunctional MDSCs) and stimulated ex vivo with PMA/ionomycin in the presence of brefeldin A for intracellular cytokine staining (double staining for IL-6 and IL-10).

Supplementary Figure 8 Microglial cells adopt a ‘neurotoxic’ phenotype during EAE in the absence of functional Ly6G+ MDSCs.

a,b, CD11bintCD45intLy6G microglia was sorted from Ly6gCre/WT mice (n = 4) and Stat3ΔLy6G mice (n = 4) at early disease recovery (day 22). Relative RNA abundance (RQ) of Clec7a, Gpnmb, Trem2 and ApoE, which are signature genes of a ‘neurotoxic’ microglial state (a) (Krasemann et al. 2017) and Tmem119, P2ry12, Sall1 and Spi1, which are signature genes of a ‘homeostatic’ microglial state (b) (Krasemann et al. 2017); symbols depict biological replicates (bars, mean ± s.d.); Mann-Whitney U-test; *P < 0.05. Ref: Krasemann, S. et al. The TREM2-APOE pathway drives the transcriptional phenotype of dysfunctional microglia in neuro-degenerative diseases. Immunity 47, 566–581.e9 (2017).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8

Reporting Summary

Supplementary Table 1

Genes differentially expressed in CNS onset-Ly6G+ cells when compared to all other Ly6G+ cell subsets

Supplementary Table 2

Gene ontology (GO) terms significantly enriched in genes universally downregulated in CNS onset-Ly6G+ cells

Supplementary Table 3

Gene ontology (GO) terms significantly enriched in genes universally upregulated in CNS onset-Ly6G+ cells

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Knier, B., Hiltensperger, M., Sie, C. et al. Myeloid-derived suppressor cells control B cell accumulation in the central nervous system during autoimmunity. Nat Immunol 19, 1341–1351 (2018). https://doi.org/10.1038/s41590-018-0237-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0237-5

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