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:

Skin and gut imprinted helper T cell subsets exhibit distinct functional phenotypes in central nervous system autoimmunity

Abstract

Multidimensional single-cell analyses of T cells have fueled the debate about whether there is extensive plasticity or ‘mixed’ priming of helper T cell subsets in vivo. Here, we developed an experimental framework to probe the idea that the site of priming in the systemic immune compartment is a determinant of helper T cell–induced immunopathology in remote organs. By site-specific in vivo labeling of antigen-specific T cells in inguinal (i) or gut draining mesenteric (m) lymph nodes, we show that i-T cells and m-T cells isolated from the inflamed central nervous system (CNS) in a model of multiple sclerosis (MS) are distinct. i-T cells were Cxcr6+, and m-T cells expressed P2rx7. Notably, m-T cells infiltrated white matter, while i-T cells were also recruited to gray matter. Therefore, we propose that the definition of helper T cell subsets by their site of priming may guide an advanced understanding of helper T cell biology in health and disease.

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

Fig. 1: Provenance mapping of CNS T cells to distinct peripheral priming sites.
Fig. 2: Clonal expansion of i-T cells versus m-T cells.
Fig. 3: Imprinting of helper T cell signatures in i-T cells versus m-T cells.
Fig. 4: Core signatures of i-T cells versus m-T cells.
Fig. 5: Transcriptional modules of i-T cells versus m-T cells.
Fig. 6: The i- and m-signatures of antigen-specific T cells are robust under various priming conditions.
Fig. 7: Distinct functional phenotypes of i-T cells versus m-T cells in the CNS.

Similar content being viewed by others

Data availability

Next-generation sequencing raw data and processed gene expression data have been deposited into the GEO repository under the accession numbers GSE156718 (scRNA-seq mouse), GSE172003 (scRNA-seq human), GSE172513 (ATAC-seq mouse) and GSE171122 (bulk RNA-seq mouse). Clinical data for human samples can be found in Supplementary Table 2. All other data generated or analyzed during this study are included in the published article or are available from the corresponding author upon reasonable request.

Code availability

No custom code or algorithms were used in this study.

References

  1. Bielekova, B. et al. Encephalitogenic potential of the myelin basic protein peptide (amino acids 83–99) in multiple sclerosis: results of a phase II clinical trial with an altered peptide ligand. Nat. Med. 6, 1167–1175 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Sospedra, M. & Martin, R. Immunology of multiple sclerosis. Annu. Rev. Immunol. 23, 683–747 (2005).

    Article  CAS  PubMed  Google Scholar 

  3. Roosendaal, S. D. & Barkhof, F. Imaging phenotypes in multiple sclerosis. Neuroimaging Clin. N. Am. 25, 83–96 (2015).

    Article  PubMed  Google Scholar 

  4. Berer, K. et al. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Kadowaki, A. & Quintana, F. J. The gut–CNS axis in multiple sclerosis. Trends Neurosci. 43, 622–634 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Mosmann, T. R. & Coffman, R. L. TH1 and TH2 cells: different patterns of lymphokine secretion lead to different functional properties. Annu. Rev. Immunol. 7, 145–173 (1989).

    Article  CAS  PubMed  Google Scholar 

  7. Korn, T., Bettelli, E., Oukka, M. & Kuchroo, V. K. IL-17 and TH17 cells. Annu. Rev. Immunol. 27, 485–517 (2009).

    Article  CAS  PubMed  Google Scholar 

  8. Zhou, L., Chong, M. M. W. & Littman, D. R. Plasticity of CD4+ T cell lineage differentiation. Immunity 30, 646–655 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Sallusto, F. Heterogeneity of human CD4+ T cells against microbes. Annu. Rev. Immunol. 34, 317–334 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Kiner, E. et al. Gut CD4+ T cell phenotypes are a continuum molded by microbes, not by TH archetypes. Nat. Immunol. 22, 216–228 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Becattini, S. et al. Functional heterogeneity of human memory CD4+ T cell clones primed by pathogens or vaccines. Science 347, 400–406 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Kremers, G.-J., Hazelwood, K. L., Murphy, C. S., Davidson, M. W. & Piston, D. W. Photoconversion in orange and red fluorescent proteins. Nat. Methods 6, 355–358 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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  PubMed  Google Scholar 

  14. Wu, H.-J. et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 32, 815–827 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kyratsous, N. I. et al. Visualizing context-dependent calcium signaling in encephalitogenic T cells in vivo by two-photon microscopy. Proc. Natl Acad. Sci. USA 114, E6381–E6389 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Alli, R., Nguyen, P. & Geiger, T. L. Retrogenic modeling of experimental allergic encephalomyelitis associates T cell frequency but not TCR functional affinity with pathogenicity. J. Immunol. 181, 136–145 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Nguyen, P. et al. Discrete TCR repertoires and CDR3 features distinguish effector and Foxp3+ regulatory T lymphocytes in myelin oligodendrocyte glycoprotein-induced experimental allergic encephalomyelitis. J. Immunol. 185, 3895–3904 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Pavlidis, N. G., Hofmeyr, D. P. & Tasoulis, S. K. Minimum density hyperplanes. J. Mach. Learn. Res. 17, 1–33 (2018).

    Google Scholar 

  19. Hartigan, J. A. & Hartigan, P. M. The dip test of unimodality. Ann. Stat. 13, 70–84 (1985).

    Article  Google Scholar 

  20. Lee, Y. et al. Induction and molecular signature of pathogenic TH17 cells. Nat. Immunol. 13, 991–999 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Spitz, F. & Furlong, E. E. M. Transcription factors: from enhancer binding to developmental control. Nat. Rev. Genet. 13, 613–626 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Li, Z. et al. Identification of transcription factor binding sites using ATAC-seq. Genome Biol. 20, 45–21 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Bettelli, E., Baeten, D., Jäger, A., Sobel, R. A. & Kuchroo, V. K. Myelin oligodendrocyte glycoprotein-specific T and B cells cooperate to induce a Devic-like disease in mice. J. Clin. Invest. 116, 2393–2402 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Krishnamoorthy, G., Lassmann, H., Wekerle, H. & Holz, A. Spontaneous opticospinal encephalomyelitis in a double-transgenic mouse model of autoimmune T cell/B cell cooperation. J. Clin. Invest. 116, 2385–2392 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Foulkes, N. S., Borjigin, J., Snyder, S. H. & Sassone-Corsi, P. Rhythmic transcription: the molecular basis of circadian melatonin synthesis. Trends Neurosci. 20, 487–492 (1997).

    Article  CAS  PubMed  Google Scholar 

  27. Rothhammer, V. & Quintana, F. J. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat. Rev. Immunol. 19, 184–197 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Sigmundsdottir, H. & Butcher, E. C. Environmental cues, dendritic cells and the programming of tissue-selective lymphocyte trafficking. Nat. Immunol. 9, 981–987 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Iwata, M. et al. Retinoic acid imprints gut-homing specificity on T cells. Immunity 21, 527–538 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Sigmundsdottir, H. et al. DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat. Immunol. 8, 285–293 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Biedermann, T. et al. IL-12 instructs skin homing of human TH2 cells. J. Immunol. 177, 3763–3770 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Delacher, M. et al. Precursors for nonlymphoid-tissue Treg cells reside in secondary lymphoid organs and are programmed by the transcription factor BATF. Immunity 52, 295–312 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Akdis, C. A. Does the epithelial barrier hypothesis explain the increase in allergy, autoimmunity and other chronic conditions? Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00538-7 (2021).

  34. Beltrán, E. et al. Early adaptive immune activation detected in monozygotic twins with prodromal multiple sclerosis. J. Clin. Invest. 129, 4758–4768 (2019).

  35. Smolders, J. et al. Tissue-resident memory T cells populate the human brain. Nat. Commun. 9, 4593 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Herich, S. et al. Human CCR5high effector memory cells perform CNS parenchymal immune surveillance via GZMK-mediated transendothelial diapedesis. Brain 142, 3411–3427 (2019).

  37. Kumar, B. V. et al. Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep. 20, 2921–2934 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hashimoto-Hill, S., Friesen, L., Kim, M. & Kim, C. H. Contraction of intestinal effector T cells by retinoic acid-induced purinergic receptor P2X7. Mucosal Immunol. 10, 912–923 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Rissiek, B., Haag, F., Boyer, O., Koch-Nolte, F. & Adriouch, S. P2X7 on mouse T cells: one channel, many functions. Front. Immun. 6, 204 (2015).

    Article  Google Scholar 

  40. Borges da Silva, H. et al. The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8+ T cells. Nature 559, 264–268 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Borges da Silva, H. et al. Sensing of ATP via the purinergic receptor P2RX7 promotes CD8+ Trm cell generation by enhancing their sensitivity to the cytokine TGF-β. Immunity 53, 158–171 (2020).

    Article  CAS  PubMed  Google Scholar 

  42. Cano-Gamez, E. et al. Single-cell transcriptomics identifies an effectorness gradient shaping the response of CD4+ T cells to cytokines. Nat. Commun. 11, 1801–1815 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Thompson, A. J. et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 17, 162–173 (2018).

    Article  PubMed  Google Scholar 

  44. Pham, A. H., McCaffery, J. M. & Chan, D. C. Mouse lines with photo-activatable mitochondria to study mitochondrial dynamics. Genesis 50, 833–843 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Lee, P. P. et al. A critical role for Dnmt1 and DNA methylation in T cell development, function, and survival. Immunity 15, 763–774 (2001).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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  PubMed  PubMed Central  Google Scholar 

  48. Mombaerts, P. et al. RAG-1-deficient mice have no mature B and T lymphocytes. Cell 68, 869–877 (1992).

    Article  CAS  PubMed  Google Scholar 

  49. 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  PubMed  PubMed Central  Google Scholar 

  50. Unutmaz, D. et al. The primate lentiviral receptor Bonzo/STRL33 is coordinately regulated with CCR5 and its expression pattern is conserved between human and mouse. J. Immunol. 165, 3284–3292 (2000).

    Article  CAS  PubMed  Google Scholar 

  51. 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  PubMed  PubMed Central  Google Scholar 

  52. Stoeckius, M. et al. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  56. 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  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Langmead, B. Aligning short sequencing reads with Bowtie. Curr. Protoc. Bioinformatics 32, 11.7.1–11.7.14 (2010).

    Article  Google Scholar 

  61. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Heinemann, C. et al. IL-27 and IL-12 oppose pro-inflammatory IL-23 in CD4+ T cells by inducing Blimp1. Nat. Commun. 5, 3770 (2014).

    Article  CAS  PubMed  Google Scholar 

  64. Kulakovskiy, I. V. et al. HOCOMOCO: towards a complete collection of transcription factor binding models for human and mouse via large-scale ChIP-seq analysis. Nucleic Acids Res. 46, D252–D259 (2018).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank V. Husterer and M. Pfaller for skillful technical assistance as well as A. Domadenik, V. Kavaka and M. Hammel for help with library preparation. This study was supported by funds from the European Research Council (ERC; CoG 647215 to T. Korn); the Deutsche Forschungsgemeinschaft (SFB1054-B06 (T. Korn), SFB1054-B05 (D.H.B., V.R.B. and S.G.), SFB1371-P04 (D.H.B., K.S. and S.J.), GA 2913/1-1 (C.G.), DO 420/7-1 (K.D.), EXC 2145 (SyNergy) ID 390857198 (T. Korn and K.D.), TRR128-A07 and TRR128-A12 (T. Korn), TRR274-A01 (T. Korn), TRR274-B03 (T.M.), TRR274-C02 (T.M.) and TRR274-Z02 (T. Korn and T.M.)); the Gemeinnützige Hertie-Stiftung (Hertie Network of Excellence in Clinical Neuroscience to C.G. and T. Korn); and the Langmatz-Foundation (to C.G.).

Author information

Authors and Affiliations

Authors

Contributions

M.H. designed experiments, performed most of the experiments, analyzed data and drafted the manuscript. E.B. performed the human scRNA-seq experiments and analyzed the mouse and human scRNA-seq data. R.K. performed histology and analyzed data. G.L. analyzed the mouse scRNA-seq and bulk RNA-seq experiments. H.D.M. performed and analyzed the ATAC-seq experiments. S.T., S.K., A.M., C.S., L.A., B.K., G.G., A.M.A., L.A.G. and S.F. performed experiments and analyzed data. I.J.B. performed and analyzed the Ca2+ imaging experiments. R.Ö. performed the mouse bulk RNA-seq experiments and analyzed data. S.G., S.J. and K.S. performed the mouse scRNA-seq experiments and analyzed data. C.G. analyzed the human scRNA experiments. V.R.B., R.R., T. Kümpfel, N.K., B.H., D.H.B., T.M. and K.D. designed experiments and analyzed data. T. Korn 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

Peer review information Nature Immunology thanks Britta Engelhardt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Site-specific labeling of T cells by photoconversion in inguinal and mesenteric lymph nodes in vivo.

a, Schematic of photoconversion of T cells in the iLN and mLN of PhAMT mice. b, Flow cytometric gating strategy for mD2GREEN or mD2RED CD4+ T cells. The DUMP channel comprised LIVE/DEADTM-Near-IR and CD8α-APC-Cy7. c, Flow cytometric assessment in the indicated LN of iLN-labeled (top) and mLN-labeled (bottom) PhAMT mice immediately after photoconversion (photo) or without photoconversion (dark). Representative plots of n = 3 mice per group. d, e, Flow cytometric proliferation readout of in vitro-activated PhAMT CD4+ T cells 4 days after photoconversion (d) and mD2RED signal over time (e). Representative plots from two independent experiments. f, Experimental design of iLN (top) or mLN (bottom) irradiation in MOG(35-55)/CFA immunized PhamT mice. g, Flow cytometric assessment of mD2REDCD44high frequencies (top) and absolute numbers (bottom) in different tissues in iLN-labeled PhAMT EAE mice 2 days after photoconversion at indicated time points. Representative plots from two mice per time point. h, i, Flow cytometric assessment in the spleen of iLN-labeled (left) and mLN-labeled (right) PhAMT EAE mice 2 days after photoconversion at disease onset (h) and of the CNS in non-photoconverted (dark) PhAMT EAE mice (i). Representative plots from spleen n = 15 mice per group, CNS n = 7 mice. j, Frequencies of regulatory and activated conventional mD2GREENCD4+ T cells in PhAMT EAE mice for different tissues. n = 2 mice per group, representative plot from three independent experiments for Foxp3 and CD44 and two for CD69. k, Frequency and duration of Ca2+ signaling in iLN (top) or mLN (bottom) of mice immunized on days 3 or 4 post transfer, that is days 13 or 14 post immunization. Left panels: individual Ca2+ signaling durations of TCRMOG Twitch-2B T cells. The dotted line indicates the cut-off (2 minutes) to distinguish between short- and long-term Ca2+ signaling. Cumulative results from iLN(PBS) and mLN(PBS) n = 2, iLN(MOG) and mLN(MOG) n = 3 mice. Right panels: fractions of T cells presenting short- and long-term Ca2+ signaling or no Ca2+ spikes. Data shown as mean. l, Representative images of TCRMOG Twitch-2B T cells from intravital time-lapse two-photon microscopy for Ca2+ imaging depicted by a fluorescence overlay of T cells (left) and a pseudocolor Ca2+ ratio image (right) from iLN(PBS) and mLN(PBS) n = 2, iLN(MOG) and mLN(MOG) n = 3 mice (see Supplementary Videos 14). Scale bars 25 μm.

Extended Data Fig. 2 TCR repertoire analysis of i-T cells and m-T cells.

a, Schematic of single cell sequencing of sorted iLN- or mLN-derived photoconverted T cells from MOG(35-55)/CFA immunized PhAMT mice in combination with TotalSeq hashtag barcoding antibodies. b, Unsupervised clustering t-distributed stochastic neighbour embedding (t-SNE) plot of all single mD2RED CD4+CD44high T cells analyzed. In the upper plot only single T cells from i-stream are colored (purple). In the lower plot only single T cells from m-stream are colored (orange). n = 5 PhAMT EAE mice per group, n = 14621 cells, i-T cells n = 7228 cells, m-T cells n = 7393 cells. c, Repertoire overlap analysis using hierarchical clustering. Dendrogram shows weighted clonal overlaps for TRB-CDR3 sequences among mice, analyzed using F pairwise similarity metric in VDJtools. Branch length shows the distance between repertoires. n = 3 PhAMT EAE mice per group. d, Average gene expression of all single mD2RED CD4+CD44high T cells analyzed for the TOP10 differentially expressed genes of cluster 0 to 7.

Extended Data Fig. 3 Single cell transcriptome analysis in i-T cells and m-T cells.

a, Average gene expression of all single mD2RED CD4+CD44high T cells analyzed from i- and m-stream in spleen and CNS grouped into T cell subsets based on key signature genes (see Fig. 3). b, Flow cytometric assessment in iLN-labeled (left) and mLN-labeled (right) PhAMIL17 EAE mice 2 days after photoconversion at disease onset. LN (top row), spleen (middle row), and CNS (bottom row). Representative plots of n = 3 mice per group. c, Unsupervised clustering t-SNE plot, colored according to i- and m-stream cell group and key gene marker (Tbx21, Rorc, Ccr6, Csf2, Ifng, Il17a, Il10) expression. n = 2 PhAMT EAE mice per group, n = 4169 cells.

Extended Data Fig. 4 Core signatures and transcription factor regulons of i-T cells and m-T cells.

a, b, Gene expression (violin plots) of i-stream signature genes Cxcr6 and Itgb1 and m-stream signature genes P2rx7 and Itga4 in the spleen (a) and CNS (b). c-e, Transcriptional module analysis of scRNAseq data from Fig. 4 based on SCENIC algorithm in i- and m-T cells. All genes controlled by a given transcription factor build a regulon. The regulon specificity score (RSS) was calculated using Jensen-Shannon divergence. c, Genome-wide regulons with RSS > 0.2 in at least one condition are displayed for i-T cells and m-T cells (i-stream and m-stream across all organs). d, e, Regulons that control Cxcr6 (d) and Itga4 (e) with no RSS threshold requirement.

Extended Data Fig. 5 ATACseq of i-T cells and m-T cells isolated from the spleen.

a, Genome browser view of key gene markers (Tbx21, Rorc, Ifng, Csf2, Il21, and Ccr6). Displayed tracks correspond to ATACseq for i-T cells (purple) and m-T cells (orange). In all regions, differential peaks (if existing) are highlighted with arrows. b, c, Ranked list of the top transcription factor motifs predicted by HOMER based on cumulative hypergeometric distribution testing for differential ATAC peaks associated to signature genes for splenic i-T cells (b) and m-T cells (c). For i-T cells 48 transcription factor sequences out of 154 are shown and for m-T cells, 16 sequences out of 30 are shown.

Extended Data Fig. 6 Robust i-T cell and m-T cell signatures in various immunization protocols.

a, Flow cytometric assessment of mD2REDCD4+CD44high T cells in the spleen of iLN-labeled (purple) and mLN-labeled (orange) PhamT mice, for Cxcr6 (top), P2rx7 (middle), and CD49d (Itga4) vs CD29 (Itgb1) (bottom). PhamT mice were subjected to different s.c. immunization regimens at the base of tail plus pertussis toxin i.v. on day 0 and day 2 after immunization (as indicated) and analyzed 2 days after photoconversion on day 11 after immunization. Representative plots from n = 3 PhamT mice per group (iLN- and mLN-labeled) and immunization condition. Numbers indicate mean fluorescent intensities. PGN, peptidoglycan. b, i-T cells and m-T cells do not cross-traffic after i.v. immunization. 2D2 PhamT mice were injected with 40 μg MOG(35-55) i.v., photoconverted at iLN (left) or mLN (right) on day 2, and analyzed for the fraction of CD44high mD2RED T cells in iLN and mLN on day 4 after injection. Representative plots from n = 4 2D2 PhamT mice per group.

Extended Data Fig. 7 Distinct functional phenotypes of i-T cells and m-T cells in the CNS compartment both in mice and humans.

a, EAE progression in iLN-labeled and mLN-labeled PhAMT EAE mice. Photoconversion at disease onset and analysis 2 days later. iLN-labeled n = 5 mice, mLN-labeled n = 3 mice, representative plot from five independent experiments. Data shown as mean ± s.d. b, Flow cytometric assessment of CD49d (Itga4) vs CD44 in transferred TCRMOG mD2REDCD4+CD44high i- and m-T cells isolated from the spleen of secondary recipient Rag1–/– mice (approximately 20 days after transfer). n = 3 mice per group. c, EAE progression in wild-type (WT) and Cxcr6–/– mice. WT n = 5 mice, Cxcr6–/– n = 6 mice, representative plot from three independent experiments. Data shown as mean ± s.d. d, Unsupervised clustering t-distributed stochastic neighbour embedding (t-SNE) plot of cerebrospinal fluid (CSF) single CD4+ T cells isolated from untreated MS patients, colored according to T helper cell and Treg cell signature gene expression. Th1 (CSF1, CXCR6, IL12B2, IL18R1, KLRC1, HOPX), Th2 (IL4, TNFSF13B, BATF, NFIL3, ATF5), Th17 (IL17A, IL21, IL2, TNFRSF13B, PTGFRN, AHR, IRF4, RORA, PLAGL2), and Treg (FOXP3, IKZF2, NRP1, FOSB, TNFSF11, IRF8). e, Gene marker expression of a selection of i-stream core signature genes (IL2RB, IL18RAP, SYTL2) (top row), and m-stream core signature genes (NT5E, DST, AIG1) (bottom row). d, e, n = 6 CSF samples from untreated MS patients, n = 14339 cells.

Supplementary information

Reporting Summary

Peer Review Information

Supplementary Tables

Supplementary Tables 1–3.

Supplementary Video 1

Intravital imaging of Ca2+ signaling in iLN of a PBS/CFA-immunized mouse after transfer of TCRMOG Twitch-2B T cells. A fluorescence overlay (left) and a pseudocolor ratio image (right) are depicted. Blue/green, TCRMOG Twitch-2B T cells; red, phagocytes visualized by detecting autofluorescence and i.v. infusion of fluorescent dextran; yellow, high Ca2+; purple, low Ca2+.

Supplementary Video 2

Intravital imaging of Ca2+ signaling in iLN of a MOG35–55/CFA-immunized mouse after transfer of TCRMOG Twitch-2B T cells. A fluorescence overlay (left) and a pseudocolor ratio image (right) are depicted. Blue/green, TCRMOG Twitch-2B T cells; red, phagocytes visualized by detecting autofluorescence and i.v. infusion of fluorescent dextran; yellow, high Ca2+; purple, low Ca2+.

Supplementary Video 3

Intravital imaging of Ca2+ signaling in mLN of a PBS/CFA-immunized mouse after transfer of TCRMOG Twitch-2B T cells. A fluorescence overlay (left) and a pseudocolor ratio image (right) are depicted. Blue/green, TCRMOG Twitch-2B T cells; red, phagocytes visualized by detecting autofluorescence and i.v. infusion of fluorescent dextran; yellow, high Ca2+; purple, low Ca2+.

Supplementary Video 4

Intravital imaging of Ca2+ signaling in mLN of a MOG35–55/CFA-immunized mouse after transfer of TCRMOG Twitch-2B T cells. A fluorescence overlay (left) and a pseudocolor ratio image (right) are depicted. Blue/green, TCRMOG Twitch-2B T cells; red, phagocytes visualized by detecting autofluorescence and i.v. infusion of fluorescent dextran; yellow, high Ca2+; purple, low Ca2+.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hiltensperger, M., Beltrán, E., Kant, R. et al. Skin and gut imprinted helper T cell subsets exhibit distinct functional phenotypes in central nervous system autoimmunity. Nat Immunol 22, 880–892 (2021). https://doi.org/10.1038/s41590-021-00948-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00948-8

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