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:

Cognate microglia–T cell interactions shape the functional regulatory T cell pool in experimental autoimmune encephalomyelitis pathology

Abstract

Microglia, the parenchymal brain macrophages of the central nervous system, have emerged as critical players in brain development and homeostasis. The immune functions of these cells, however, remain less well defined. We investigated contributions of microglia in a relapsing–remitting multiple sclerosis paradigm, experimental autoimmune encephalitis in C57BL/6 x SJL F1 mice. Fate mapping-assisted translatome profiling during the relapsing–remitting disease course revealed the potential of microglia to interact with T cells through antigen presentation, costimulation and coinhibition. Abundant microglia–T cell aggregates, as observed by histology and flow cytometry, supported the idea of functional interactions of microglia and T cells during remission, with a bias towards regulatory T cells. Finally, microglia-restricted interferon-γ receptor and major histocompatibility complex mutagenesis significantly affected the functionality of the regulatory T cell compartment in the diseased central nervous system and remission. Collectively, our data establish critical non-redundant cognate and cytokine-mediated interactions of microglia with CD4+ T cells during autoimmune neuroinflammation.

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: Longitudinal fate map-based translatome profiling of microglia during the RR-EAE course.
Fig. 2: Translatome analysis of microglia during the course of RR-EAE.
Fig. 3: Absence of overt microglia activation during the REL stage.
Fig. 4: Interactions of microglia with T cells in RR-EAE.
Fig. 5: Analysis of EAE pathology in animals harboring a microglial Ifn-γR deficiency.
Fig. 6: Microglial Ifn-γR deficiency affects brain T cells in RR-EAE.
Fig. 7: Transcriptome analysis of the CNS T cell compartment during RR-EAE.
Fig. 8: Microglial MHC class II deficiency affects Treg cells in RR-EAE.

Similar content being viewed by others

Data availability

The RNA-seq datasets reported in this study can be found at Gene Expression Omnibus under the accession code GSE214709. Other data and reagents are available from the corresponding author on request. Source data are provided with this paper.

References

  1. Lassmann, H., Brück, W. & Lucchinetti, C. F. The immunopathology of multiple sclerosis: an overview. Brain Pathol. 17, 210–218 (2007).

    Article  Google Scholar 

  2. Filippi, M. et al. Multiple sclerosis. Nat. Rev. Dis. Primers 4, 43 (2018).

    Article  Google Scholar 

  3. Martin, R., Sospedra, M., Eiermann, T. & Olsson, T. Multiple sclerosis: doubling down on MHC. Trends Genet. https://doi.org/10.1016/j.tig.2021.04.012 (2021).

  4. Korn, T. et al. Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation. Nat. Med. 13, 423–431 (2007).

    Article  CAS  Google Scholar 

  5. Garg, G. et al. Blimp1 prevents methylation of Foxp3 and loss of regulatory T cell identity at sites of inflammation. Cell Rep. 26, 1854–1868 (2019).

    Article  CAS  Google Scholar 

  6. McGeachy, M. J., Stephens, L. A. & Anderton, S. M. Natural recovery and protection from autoimmune encephalomyelitis: contribution of CD4+CD25+ regulatory cells within the central nervous system. J. Immunol. 175, 3025–3032 (2005).

    Article  CAS  Google Scholar 

  7. Wagner, C. A., Roqué, P. J. & Goverman, J. M. Pathogenic T cell cytokines in multiple sclerosis. J. Exp. Med. 217, e20190460 (2020).

    Article  Google Scholar 

  8. 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  Google Scholar 

  9. Stromnes, I. M. & Goverman, J. M. Active induction of experimental allergic encephalomyelitis. Nat. Protoc. 1, 1810–1819 (2006).

    Article  CAS  Google Scholar 

  10. Mendel, I., Rosbo, N. Kde & Ben-Nun, A. A myelin oligodendrocyte glycoprotein peptide induces typical chronic experimental autoimmune encephalomyelitis in H-2b mice: fine specificity and T cell receptor Vβ expression of encephalitogenic T cells. Eur. J. Immunol. 25, 1951–1959 (1995).

    Article  CAS  Google Scholar 

  11. Kurschus, F. C. et al. Genetic proof for the transient nature of the TH17 phenotype. Eur. J. Immunol. 40, 3336–3346 (2010).

    Article  CAS  Google Scholar 

  12. Hirota, K. et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263 (2011).

    Article  CAS  Google Scholar 

  13. Goverman, J. Autoimmune T cell responses in the central nervous system. Nat. Rev. Immunol. 9, 393–407 (2009).

    Article  CAS  Google Scholar 

  14. Mundt, S., Greter, M., Flügel, A. & Becher, B. The CNS immune landscape from the viewpoint of a T cell. Trends Neurosci. 42, 667–679 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  16. Mildner, A. et al. CCR2+Ly-6Chi monocytes are crucial for the effector phase of autoimmunity in the central nervous system. Brain 132, 2487–2500 (2009).

    Article  Google Scholar 

  17. Wolf, Y. et al. Microglial MHC class II is dispensable for experimental autoimmune encephalomyelitis and cuprizone-induced demyelination. Eur. J. Immunol. 48, 1308–1318 (2018).

    Article  CAS  Google Scholar 

  18. Bartholomäus, I. et al. Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions. Nature 462, 94–98 (2009).

    Article  Google Scholar 

  19. O’Connor, R. A., Malpass, K. H. & Anderton, S. M. The inflamed central nervous system drives the activation and rapid proliferation of Foxp3+ regulatory T cells. J. Immunol. 179, 958–966 (2007).

    Article  Google Scholar 

  20. Do, J. et al. Treg-specific IL-27Rα deletion uncovers a key role for IL-27 in Treg function to control autoimmunity. Proc. Natl Acad. Sci. USA 114, 10190–10195 (2017).

    Article  CAS  Google Scholar 

  21. McRae, B. L. et al. Induction of active and adoptive relapsing experimental autoimmune encephalomyelitis (EAE) using an encephalitogenic epitope of proteolipid protein. J. Neuroimmunol. 38, 229–240 (1992).

    Article  CAS  Google Scholar 

  22. Ajami, B., Bennett, J. L., Krieger, C., Tetzlaff, W. & Rossi, F. M. V. Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat. Neurosci. 10, 1538–1543 (2007).

    Article  CAS  Google Scholar 

  23. Ajami, B., Bennett, J. L., Krieger, C., Mcnagny, K. M. & Rossi, F. M. V. Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat. Neurosci. 14, 1142–1149 (2011).

    Article  CAS  Google Scholar 

  24. Shemer, A. et al. Engrafted parenchymal brain macrophages differ from microglia in transcriptome, chromatin landscape and response to challenge. Nat. Commun. 9, 5206 (2018).

    Article  CAS  Google Scholar 

  25. Yamasaki, R. et al. Differential roles of microglia and monocytes in the inflamed central nervous system. J. Exp. Med. 211, 1533–1549 (2014).

    Article  CAS  Google Scholar 

  26. Yona, S. et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 38, 79–91 (2013).

    Article  CAS  Google Scholar 

  27. Sanz, E. et al. Cell-type-specific isolation of ribosome-associated mRNA from complex tissues. Proc. Natl Acad. Sci. USA 106, 13939–13944 (2009).

    Article  CAS  Google Scholar 

  28. Haimon, Z. et al. Re-evaluating microglia expression profiles using RiboTag and cell isolation strategies. Nat. Immunol. 19, 636–644 (2018).

    Article  CAS  Google Scholar 

  29. Chappell‐Maor, L. et al. Comparative analysis of CreER transgenic mice for the study of brain macrophages: a case study. Eur. J. Immunol. 50, 353–362 (2020).

    Article  Google Scholar 

  30. Goldmann, T. et al. A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat. Immunol. 16, 1618–1626 (2013).

    CAS  Google Scholar 

  31. Butovsky, O. & Weiner, H. L. Microglial signatures and their role in health and disease. Nat. Rev. Neurosci. 19, 622 (2018).

    Article  CAS  Google Scholar 

  32. Shemer, A. et al. Interleukin-10 prevents pathological microglia hyperactivation following peripheral endotoxin challenge. Immunity 53, 1033–1049 (2020).

    Article  CAS  Google Scholar 

  33. De, A., Dainichi, T., Rathinam, C. V. & Ghosh, S. The deubiquitinase activity of A20 is dispensable for NF-κB signaling. EMBO Rep. 15, 775–783 (2014).

    Article  CAS  Google Scholar 

  34. Song, W. M. & Colonna, M. The identity and function of microglia in neurodegeneration. Nat. Immunol. https://doi.org/10.1038/s41590-018-0212-1 (2018).

  35. Kim, J.-S. et al. A binary Cre transgenic approach dissects microglia and CNS border-associated macrophages. Immunity 54, 176–190.e7 (2021).

    Article  CAS  Google Scholar 

  36. Schetters, S. T. T., Gomez-Nicola, D., Garcia-Vallejo, J. J. & Kooyk, Y. V. Neuroinflammation: microglia and T cells get ready to tango. Front. Immunol. 8, 1905 (2018).

    Article  Google Scholar 

  37. Hommel, M. & Kyewski, B. Dynamic changes during the immune response in T cell-antigen-presenting cell clusters isolated from lymph nodes. J. Exp. Med. 197, 269–280 (2003).

    Article  CAS  Google Scholar 

  38. Burel, J. G. et al. Circulating T cell-monocyte complexes are markers of immune perturbations. eLife 8, e46045 (2019).

    Article  Google Scholar 

  39. Eberl, G. et al. An essential function for the nuclear receptor RORγt in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 5, 64–73 (2004).

    Article  CAS  Google Scholar 

  40. Casella, G. et al. IL-27, but not IL-35, inhibits neuroinflammation through modulating GM-CSF expression. Sci. Rep. 7, 16547 (2017).

    Article  Google Scholar 

  41. 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 

  42. Ronchetti, S. et al. Glucocorticoid-induced tumour necrosis factor receptor-related protein: a key marker of functional regulatory T cells. J. Immunol. Res. 2015, 171520 (2015).

    Article  Google Scholar 

  43. Vahl, J. C. et al. Continuous T cell receptor signals maintain a functional regulatory T cell pool. Immunity 41, 722–736 (2014).

    Article  CAS  Google Scholar 

  44. Long, S. A. & Buckner, J. H. CD4+FOXP3+ T regulatory cells in human autoimmunity: more than a numbers game. J. Immunol. 187, 2061–2066 (2011).

    Article  CAS  Google Scholar 

  45. Sénécal, V. et al. Production of IL‐27 in multiple sclerosis lesions by astrocytes and myeloid cells: modulation of local immune responses. Glia 64, 553–569 (2016).

    Article  Google Scholar 

  46. Steimle, V., Siegrist, C., Mottet, A., Lisowska-Grospierre, B. & Mach, B. Regulation of MHC class II expression by interferon-γ mediated by the transactivator gene CIITA. Science 265, 106–109 (1994).

    Article  CAS  Google Scholar 

  47. Sosa, R. A., Murphey, C., Robinson, R. R. & Forsthuber, T. G. IFN-γ ameliorates autoimmune encephalomyelitis by limiting myelin lipid peroxidation. Proc. Natl Acad. Sci. USA 112, E5038–E5047 (2015).

    Article  CAS  Google Scholar 

  48. Lassmann, H. & Bradl, M. Multiple sclerosis: experimental models and reality. Acta Neuropathol. 133, 223–244 (2017).

    Article  CAS  Google Scholar 

  49. Lee, S. H. et al. Identifying the initiating events of anti-listeria responses using mice with conditional loss of IFN-γ receptor subunit 1 (IFNGR1). J. Immunol. 191, 4223–4234 (2013).

    Article  CAS  Google Scholar 

  50. Hashimoto, K., Joshi, S. K. & Koni, P. A. A conditional null allele of the major histocompatibility IA-β chain gene. Genesis 32, 152–153 (2002).

    Article  CAS  Google Scholar 

  51. Wan, Y. Y. & Flavell, R. A. Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc. Natl Acad. Sci. USA 102, 5126–5131 (2005).

    Article  CAS  Google Scholar 

  52. Hsu, P. D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).

    Article  CAS  Google Scholar 

  53. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  Google Scholar 

  54. Xu, H. et al. Sequence determinants of improved CRISPR sgRNA design. Genome Res. 25, 1147–1157 (2015).

    Article  CAS  Google Scholar 

  55. Chari, R., Mali, P., Moosburner, M. & Church, G. M. Unraveling CRISPR–Cas9 genome engineering parameters via a library-on-library approach. Nat. Methods 12, 823–826 (2015).

    Article  CAS  Google Scholar 

  56. Cossarizza, A. et al. Guidelines for the use of flow cytometry and cell sorting in immunological studies. Eur. J. Immunol. 49, 1457–1973 (2019).

    Article  CAS  Google Scholar 

  57. Lavin, Y. et al. Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159, 1312–1326 (2014).

    Article  CAS  Google Scholar 

  58. Keren-Shaul, H. et al. MARS-seq2.0: an experimental and analytical pipeline for indexed sorting combined with single-cell RNA sequencing. Nat. Protoc. https://doi.org/10.1038/s41596-019-0164-4 (2019).

  59. Kohen, R. et al. UTAP: user-friendly transcriptome analysis pipeline. BMC Bioinform. 20, 154–157 (2019).

    Article  Google Scholar 

Download references

Acknowledgements

The Jung Laboratory was supported by a collaborative network grant of the International Progressive MS Alliance (PMSA), the Minerva Foundation with funding from the Federal German Ministry for Education, the American Brain Foundation, the Roland N. Karlen Foundation, the Blythe Brenden–Mann Foundation, the Estate of David Levinson and the Deutsche Forschungsgemeinschaft (DFG) (CRC/TRR167 ‘NeuroMac’). T.K. is supported by the DFG (SFB1054-B06 (ID 210592381), TRR128-A07 (ID 213904703), TRR128-A12 (ID 213904703), TRR128-Z02 (ID 213904703), TRR274-A01 (ID 408885537) and EXC 2145 (SyNergy, ID 390857198) and the Hertie Foundation. We thank all members of the Jung Laboratory, the staff of the Weizmann FACS facility as well as the Weizmann Veterinary Services for expert technical help. We thank B. Dassa and A. Sarusi-Portuguez a for advice with bioinformatics, and I. Milenkovic (Institute of Neurology, Medical University of Vienna) for valuable comments on the manuscript. S. Jung is the incumbent of the Henry. H. Drake Professional Chair of Immunology.

Author information

Authors and Affiliations

Authors

Contributions

Z.H. and S.J. conceived the project and designed the experiments. Z.H., G.R.F., J.-S.K., S.T. and S.B.-H. performed experiments. Z.P. helped with the ImageStream analysis. L.C.-M. performed the RNA-seq. S.B.-D and R.H.-K. critically helped with the CRISPR–Cas9 mutagenesis design and generation of the mutant animals. A.M. and T.K. critically advised on the T cell analysis. Z.H. and S.J. wrote the manuscript. S.J. supervised the project and acquired funding.

Corresponding author

Correspondence to Steffen Jung.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the Nature Immunology team. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Gating strategies for flow cytometric analysis.

Representative analysis and gating strategy for Fig. 1d, upper panel, isotype control; lower panel, anti-HA staining, both at MAX stage.

Extended Data Fig. 2 Translatome analysis of microglia during the RR-EAE course.

(a) Representative genes from each cluster. Light gray circles, input, dark gray triangles, IP IgG, light blue squares, IP HA. Each dot represents individual mice, lines represent mean. NI, non-immunized, INIT, initiation, MAX, maximal score, REM, remission, RCOV, clinical recovery, REL, relapse. N = 6 NI, 4 INIT, 6 MAX, 8 REM, 5 RCOV and 4 REL individual mice per group. (b) Expression of selected genes encoding proposed BAM markers during the RR-EAE course. Heatmap represents log2 values of average HA samples in each stage, alongside graphs of expression of individual genes. N = 6 NI, 4 INIT, 6 MAX, 8 REM, 5 RCOV and 4 REL individual mice per group, lines represent mean.

Extended Data Fig. 3 Total brain transcriptome analysis during the RR-EAE course.

(a) Heatmap of 1378 genes retrieved as total brain transcriptomes. Differential expression was calculated using DESeq2 package. Each column is an average of input samples. Genes were clustered using K-means clustering. NI, non-immunized, INIT, initiation, MAX, maximal score, REM, remission, RCOV, clinical recovery, REL, relapse. N = 6 NI, 4 INIT, 6 MAX, 8 REM, 5 RCOV and 4 REL individual mice per group. (b) Heatmap of genes selected according to biological relevant categories. (c) Normalized reads of selected genes that show dynamic expression in brain transcriptome (input samples) during EAE course. N = 6 NI, 4 INIT, 6 MAX, 8 REM, 5 RCOV and 4 REL individual mice per group, lines represent mean. (d)Number of DEG (abs fold change > 2, p < 0.05) detected in the total brain transcriptomes at different stages.

Extended Data Fig. 4 Probing the extent of ex-vivo Microglia-T cell doublet formation.

(a) Scheme describing the mixing experiment where brains from a whole body GFP or whole body TdTomato mice were mixed together or isolated separately. Mixed brains went through enzymatic digestion and cell isolation process together. Gating strategy showing the extent of GFP/TdTomato cells from each population. (b) Calculation of the probability to obtain spurious vs. real doublets, showing that most of the doublets are not spurious (with kind help of Shalev Itzkovitz, WIS).

Extended Data Fig. 5 ImageStream and Immunohistochemistry analysis of microglia / T cell interactions in the brain.

(a) Image stream analysis of Mg–T doublets with Foxp3+ or RORγt+ T cells, by intracellular staining. Microglia were identified as CD11b+ Ly6C/G- and CD45 low cells, T cells were identified as CD45hi CD3+ CD4+ and Foxp3+ or RORγt+ cells. (b) Immunofluorescence staining of brain sections of TAM-treated Cx3cr1creER:Rpl22HA mice at the MAX stage, staining for IBA1 (cyan), HA (green), CD3 (red), Foxp3 (blue) and MHC-II (magenta). Scale bar 20uM. The image was acquired using linear unmixing in Zeiss LSM880 confocal microscope. Representative of 3 independent experiments. (c) Enlargement of the selected window of (b).

Extended Data Fig. 6 Generation of Ifnγr receptor deficient SJL mice.

(a) Schematic of CRISPR/Cas9based Ifnγr1 targeting strategy to delete 203 bp spanning the promoter and first exon of the Ifnγr1 gene. (b) Schematic showing the WT and mutant Ifnγr1 locus with details of the respective 203 bp deletion. (c) Genomic PCR analysis of Ifnγr1 −/− SJL mouse (~400 bp) and WT control (~600 bp). Representative out of infinite number of experiments. (d) Flow cytometric analysis of non-classical CD115+ Ly6c- blood monocytes for expression of CD119 (Ifnγr) in WT, Ifnγr1−/+ or Ifnγr1 −/− SJL mouse. e, f. Flow cytometric analysis of IFNγR (E) or MHC-II (F) expression of MoMF (gated as live CD45hi CD11b+ TMEM119-) in control or MgΔIfnγr1 mice at REM. MFI, mean fluorescent intensity. N = 5 individual mice per group, lines represent mean. Significance was calculated using two tailed t-test. Dashed line/empty circles - FMO; black histogram/black dots - control; red histogram/red triangles - MgΔIfnγr1 mice. g. Normalized MFI of Treg cells markers on Foxp3+ T cells of MgΔIfnγr1 and control brains. Data combined from 5 independent experiments n = 27 individual mice per group, lines represent mean. Significance was calculated using two tailed t-test. *** p < 0.001. h. Overlay of microglia (light blue; gated on live Ly6c/g1 CD11b+ CD45int TMEM119+), singlet T cells (red, gated on live Ly6c/g1- CD11b CD45hi CD4+ TCRβ+) and total T cells (gray; gated on live Ly6c/g1 CD45hi CD4+ TCRβ+), related to Fig. 5g. i. Normalized MFI of non-Treg cells (CD4+TCRβ+ Foxp3) of MgΔIfnγr1 and control brains. Data combined from 5 independent experiments n = 27 per group, lines represent mean. Significance was calculated using two tailed t-test. *p < 0.05, *** p < 0.001.

Extended Data Fig. 7 Bulk RNAseq analysis of T cells isolated from brains of MgΔIfnγr mice and littermate controls.

(a) Sorting strategy for isolation of CD25hi (Treg) and CD25 (Teff) cells from brains of MgΔIfnγr mice and littermate controls. (b) Normalized reads of selected genes from sorted CD25 (Teff) and CD25hi (Treg) cells extracted from B6/SJL MgΔIfnγr and control mice at REM stage of RR-EAE, representing Treg genes not changed between MgΔIfnγr and control. n = 6 individual mice per group, lines represent mean. (c) Normalized reads of selected genes from sorted CD25- (Teff) and CD25hi (Treg) cells extracted from B6/SJL MgΔIfnγr and control mice at REM stage of RR-EAE, representing genes significantly changed in Treg cells between MgΔIfnγr and control. n = 6 individual mice per group, lines represent mean. (d) GSEA comparison WT and MgΔIfnγr T reg cell transcriptomes with list of genes upregulated in WT CNS Treg cells vs. splenic Treg cells6. Normalized enrichment score (NES) and the false discovery rate (FDR) are shown in the enrichment plot. Normalized enrichment score = −1.04, FDR q-value = 0.34. This indicates that Treg cells from MgΔIfnγr brains do not acquire features of a CNS Treg signature.

Extended Data Fig. 8 Generation of MHC II deficient SJL mice and analysis of T cell compartment following RR-EAE challenge.

(a) Schematic of CRISPR/Cas9-based H2-Ab1s targeting strategy. (b) Schematic showing the WT and mutant H2-Ab1s locus with details of the respective 247 bp deletion. (c) Genomic PCR analysis of H2-Ab1s −/− SJL mouse (631 bp) and WT control (~900 bp). Representative out of infinite number of experiments. (d) Flow cytometric analysis of I-As on B220+ B cells in blood of WT C57BL6, WT SJL and H2-Ab1s hetero- or homozygote SJL mice establishing MHC-II deficiency of H2-Ab1s −/− SJL mouse. (e) Flow cytometric analysis of MHCII surface expression (I-Ab) on microglia of MgΔMHCII mice and controls (diseased brains). N = 20 individual mice per group, pooled from 3 independent experiments. Lines represent mean. Significance was calculated using two tailed t-test. ****p < 0.0001. (f) Flow cytometric analysis of FoRBY2 CD4+ T cells extracted from spleen and lymph nodes of 2D2 donor animals post MACS isolation, prior to adoptive transfer. (g) Flow cytometric analysis of blood of MgΔMHCII mice and control recipient mice at day 6 post immunization showing detection of engrafted 2D2 cells as CD45.1- TCRβ11+ cells, gated from GR1CD45hi CD11b CD4+ TCRb+ T cells. N = 11 individual mice per group, line represent mean, data are pooled from 3 independent experiments. Significance was calculated using two tailed t-test. (h) Flow cytometric analysis of Gitr, Foxp3 and CD25 expression on CD4+ T cells, showing that Foxp3+ or CD25+ Tregs can be defined by Gitr. (i) Gating strategy for detection of grafted cells and GITR Tregs. Related to Fig. 7d, e. j Normalized MFI of Gitr and Klrg1 on Gitrhi Treg population in control and MgΔMHCII mice. n = 20 individual mouse per group, lines represent mean, data pooled from 3 independent experiments. Significance was calculated using two tailed t-test.

Source data

Extended Data Fig. 9 Analysis of engrafted MOG-specific TCR transgenic cells (2D2).

(a) Workflow and gating strategy for concatenation of 2D2 cells. Related to Fig. 8h. (b) Flow cytometric analysis of brains of three representative individual control and MgΔMHCII mice to show the reporter gene expression in the 2D2 graft. (c) Gates used for concatenation shown in Fig. 8h. Upper plots represent entire 2D2 concatenated population, lower plots represent only Foxp3+ cells gated from 2D2+ cells, in control (left) or MgΔMHCII (right). Related to Fig. 8h.

Supplementary information

Supplementary Information

Supplementary note showing gene sequences for the SJL Ifngr1 locus, chromosome 10, and the SJL H2-Ab1 locus, chromosome 17.

Reporting Summary

Peer Review File

Source data

Source Data Fig. 1

Flow cytometry quantification.

Source Data Fig. 2

Gene lists, scores and DPI.

Source Data Fig. 4

Quantifications (for details see the figure legends).

Source Data Fig. 5

DPI and gene lists.

Source Data Fig. 7

Gene lists.

Source Data Fig. 8

Gene lists.

Source Data Extended Data Fig. 8

Clinical disease scores.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Haimon, Z., Frumer, G.R., Kim, JS. et al. Cognate microglia–T cell interactions shape the functional regulatory T cell pool in experimental autoimmune encephalomyelitis pathology. Nat Immunol 23, 1749–1762 (2022). https://doi.org/10.1038/s41590-022-01360-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01360-6

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