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:

Single-cell profiling of CNS border compartment leukocytes reveals that B cells and their progenitors reside in non-diseased meninges

Abstract

The CNS is ensheathed by the meninges and cerebrospinal fluid, and recent findings suggest that these CNS-associated border tissues have complex immunological functions. Unlike myeloid lineage cells, lymphocytes in border compartments have yet to be thoroughly characterized. Based on single-cell transcriptomics, we here identified a highly location-specific composition and expression profile of tissue-resident leukocytes in CNS parenchyma, pia-enriched subdural meninges, dura mater, choroid plexus and cerebrospinal fluid. The dura layer of the meninges contained a large population of B cells under homeostatic conditions in mice and rats. Murine dura B cells exhibited slow turnover and long-term tissue residency, and they matured in experimental neuroinflammation. The dura also contained B lineage progenitors at the pro-B cell stage typically not found outside of bone marrow, without direct influx from the periphery or the skull bone marrow. This identified the dura as an unexpected site of B cell residence and potentially of development in both homeostasis and 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: Distinct leukocyte composition of each CNS-associated border compartment altered during neuroinflammation.
Fig. 2: Bcs in the dura are located within and outside of blood and lymphatic vessels and in the dura tissue.
Fig. 3: Cells transcriptionally resembling Bc precursors populate non-diseased dura.
Fig. 4: Precursor Bcs are tissue resident and proliferate locally in the dura.

Similar content being viewed by others

Data availability

All raw single-cell sequencing data are available in the Gene Expression Omnibus (GEO) repository: GSE165153. All processed, unmodified scRNA-seq data (differential expression data, cell abundances) and technical scRNA-seq information are included as supplementary tables. An interactive version of the single-cell sequencing data created with cerebroApp51 is available at https://osmzhlab.uni-muenster.de/shiny/cerebro_meninges_mouse/ and https://osmzhlab.uni-muenster.de/shiny/cerebro_meninges_rat/. Reference datasets are publicly available: Tabula Muris bone marrow scRNA-seq data (GSM2967055); cell migration Gene Ontology term (GO:0016477); murine dura scRNA-seq data from Van Hove et al. (GSE128855). Source data are provided with this paper.

References

  1. Engelhardt, B., Vajkoczy, P. & Weller, R. O. The movers and shapers in immune privilege of the CNS. Nat. Immunol. 18, 123–131 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Aspelund, A. et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 212, 991–999 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Louveau, A. et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat. Neurosci. 21, 1380–1391 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Kivisakk, P. et al. Localizing central nervous system immune surveillance: meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Ann. Neurol. 65, 457–469 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chinnery, H. R., Ruitenberg, M. J. & McMenamin, P. G. Novel characterization of monocyte-derived cell populations in the meninges and choroid plexus and their rates of replenishment in bone marrow chimeric mice. J. Neuropathol. Exp. Neurol. 69, 896–909 (2010).

    Article  PubMed  Google Scholar 

  7. Mundt, S. et al. Conventional DCs sample and present myelin antigens in the healthy CNS and allow parenchymal T cell entry to initiate neuroinflammation. Sci. Immunol. 4, eaau8380 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Van Hove, H. et al. A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat. Neurosci. 22, 1021–1035 (2019).

    Article  PubMed  CAS  Google Scholar 

  9. Jordão, M. J. C. et al. Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science 363, eaat7554 (2019).

    Article  PubMed  CAS  Google Scholar 

  10. Steinert, E. M. et al. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell 161, 737–749 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Schafflick, D. et al. Integrated single cell analysis of blood and cerebrospinal fluid leukocytes in multiple sclerosis. Nat. Commun. 11, 247 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sayed, B. A., Christy, A. L., Walker, M. E. & Brown, M. A. Meningeal mast cells affect early T cell central nervous system infiltration and blood-brain barrier integrity through TNF: a role for neutrophil recruitment? J. Immunol. 184, 6891–6900 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Sawant, K. V. et al. Chemokine CXCL1 mediated neutrophil recruitment: role of glycosaminoglycan interactions. Sci. Rep. 6, 33123 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Howell, O. W. et al. Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis. Brain 134, 2755–2771 (2011).

    Article  PubMed  Google Scholar 

  15. Pröbstel, A.-K. et al. Gut microbiota–specific IgA+ B cells traffic to the CNS in active multiple sclerosis. Sci. Immunol. 5, eabc7191 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Fitzpatrick, Z. et al. Gut-educated IgA plasma cells defend the meningeal venous sinuses. Nature 587, 472–476 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Parker Harp, C. R. et al. Neutrophils promote VLA-4-dependent B cell antigen presentation and accumulation within the meninges during neuroinflammation. Proc. Natl Acad. Sci. USA 116, 24221–24230 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wilke, G., Steinhauser, G., Grün, J. & Berek, C. In silico subtraction approach reveals a close lineage relationship between follicular dendritic cells and BP3(hi) stromal cells isolated from SCID mice. Eur. J. Immunol. 40, 2165–2173 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Heming, M. et al. Neurological manifestations of COVID-19 feature T cell exhaustion and dedifferentiated monocytes in cerebrospinal fluid. Immunity 54, 164–175.e6 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. Schaffer, S., Maul-Pavicic, A., Voll, R. E. & Chevalier, N. Optimized isolation of renal plasma cells for flow cytometric analysis. J. Immunol. Methods 474, 112628 (2019).

    Article  CAS  PubMed  Google Scholar 

  21. 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  PubMed  PubMed Central  Google Scholar 

  22. Kunis, G. et al. IFN-γ-dependent activation of the brain’s choroid plexus for CNS immune surveillance and repair. Brain 136, 3427–3440 (2013).

    Article  PubMed  Google Scholar 

  23. Louveau, A., Filiano, A. J. & Kipnis, J. Meningeal whole mount preparation and characterization of neural cells by flow cytometry. Curr. Protoc. Immunol. 121, e50 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ahn, J. H. et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 572, 62–66 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Tabula Muris Consortium et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

  26. Han, X. et al. Mapping the mouse cell atlas by Microwell-seq. Cell 172, 1091–1107.e17 (2018).

    Article  CAS  PubMed  Google Scholar 

  27. Bendall, S. C. et al. Single-cell trajectory detection uncovers progression and regulatory coordination in human B cell development. Cell 157, 714–725 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. 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 

  29. Kitamura, D., Roes, J., Kühn, R. & Rajewsky, K. A B cell-deficient mouse by targeted disruption of the membrane exon of the immunoglobulin μ chain gene. Nature 350, 423–426 (1991).

    Article  CAS  PubMed  Google Scholar 

  30. Wright, D. E., Wagers, A. J., Gulati, A. P., Johnson, F. L. & Weissman, I. L. Physiological migration of hematopoietic stem and progenitor cells. Science 294, 1933–1936 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Cai, R. et al. Panoptic imaging of transparent mice reveals whole-body neuronal projections and skull–meninges connections. Nat. Neurosci. 22, 317–327 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Herisson, F. et al. Direct vascular channels connect skull bone marrow and the brain surface enabling myeloid cell migration. Nat. Neurosci. 21, 1209–1217 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Yao, H. et al. Leukaemia hijacks a neural mechanism to invade the central nervous system. Nature 560, 55–60 (2018).

    Article  CAS  PubMed  Google Scholar 

  34. Roth, T. L. et al. Transcranial amelioration of inflammation and cell death after brain injury. Nature 505, 223–228 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Grüneboom, A. et al. A network of trans-cortical capillaries as mainstay for blood circulation in long bones. Nat. Metab. 1, 236–250 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Korin, B. et al. High-dimensional, single-cell characterization of the brain’s immune compartment. Nat. Neurosci. 20, 1300–1309 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. 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  PubMed  Google Scholar 

  38. Machado-Santos, J. et al. The compartmentalized inflammatory response in the multiple sclerosis brain is composed of tissue-resident CD8+ T lymphocytes and B cells. Brain 141, 2066–2082 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Magliozzi, R. et al. A gradient of neuronal loss and meningeal inflammation in multiple sclerosis. Ann. Neurol. 68, 477–493 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Satoh-Takayama, N. et al. Bacteria-induced group 2 innate lymphoid cells in the stomach provide immune protection through induction of IgA. Immunity 52, 635–649.e4 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Pegg, C. C., He, C., Stroink, A. R., Kattner, K. A. & Wang, C. X. Technique for collection of cerebrospinal fluid from the cisterna magna in rat. J. Neurosci. Methods 187, 8–12 (2010).

    Article  PubMed  Google Scholar 

  42. Bowyer, J. F. et al. A visual description of the dissection of the cerebral surface vasculature and associated meninges and the choroid plexus from rat brain. J. Vis. Exp. 14, e4285 (2012).

    Google Scholar 

  43. 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 

  44. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Sun, H., Zhou, Y., Fei, L., Chen, H. & Guo, G. scMCA: a tool to define mouse cell types based on single-cell digital expression. Methods Mol. Biol. 1935, 91–96 (2019).

    Article  CAS  PubMed  Google Scholar 

  48. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 9, 47 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Hillje, R., Pelicci, P. G. & Luzi, L. Cerebro: interactive visualization of scRNA-seq data. Bioinformatics 36, 2311–2313 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

A.H. was funded by grant no. RTI2018-095497-B-I00 from Ministerio de Ciencia e Innovacion (MICINN). The CNIC is supported by MICINN and the Pro-CNIC Foundation, and is a Severo Ochoa Center of Excellence (MICINN award no. SEV-2015-0505). G.M.z.H. was supported by grants from the Deutsche Forschungsgemeinschaft (DFG; grant nos. ME4050/4-1 and ME4050/12-1) and DFG grant no. ME4050/8-1, under the frame of E-Rare-3, the ERA-Net for Research on Rare Diseases G.M.z.H. was also supported by the Heisenberg program of the DFG (grant no. ME4050/13-1), by the Grant for Multiple Sclerosis Innovation (Merck) and by a grant from the Ministerium für Innovation, Wissenschaft und Forschung (MIWF) des Landes Nordrhein-Westfalen. M. Heming and G.M.z.H. were supported by the Interdisciplinary Center for Clinical Research (IZKF) of the Medical Faculty of Münster (grant no. MzH3/020/20 to G.M.z.H. and grant no. SEED/016/21 to M. Heming). A.L. received funding from the European Research Council (ERC-StG grant no. 802305) and the DFG under Germany’s Excellence Strategy (EXC 2145 SyNergy—grant ID 390857198) and grant no. LI-2534/5-1. Servier Medical Art, licensed under a Creative Common Attribution 3.0 Generic License (https://smart.servier.com/), was used for Figs. 1a and 4a,b, Extended Data Fig. 5b and Supplementary Figs. 14 and 15.

Author information

Authors and Affiliations

Authors

Contributions

D.S., J.W., M. Heming, C.T., M. Hartlehnert, X.L., S.M.-S., A.R., A.-L.B., J.-K.S., J.M. and T.S. performed experiments. M. Heming and I.-N.L. performed computational analyses. D.S. and J.W. performed data analysis and created/edited figures. A.H., A.L., S.G.M. and H.W. cosupervised the study. M.P. revised the manuscript. G.M.z.H. conceived and supervised the study, and G.M.z.H. and D.S. wrote the manuscript. All authors critically revised the manuscript and agree with its contents.

Corresponding author

Correspondence to Gerd Meyer zu Horste.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Neuroscience thanks Maiken Nedergaard and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Perfusion is not sufficient to deplete intra-vascular leukocytes in the meninges.

a, Graph depicting the percentage of remaining peripheral blood leukocytes of all leukocytes isolated from CNS-associated border tissues (CNS, SDM, Dura, choroid plexus (CP)) after intracardial perfusion as shown in Fig. 1b. Four healthy C57BL/6 mice were intravenously injected with a fluorescent coupled antibody against CD45 (CD45iv). 10 minutes later, blood was taken, mice were intracardially perfused and leukocytes were isolated from the CNS-associated border tissues and blood. Leukocytes were then stained with a second CD45 antibody and were measured on a flow cytometer. Data represented as mean ± SD, n = 4 (b) Dotplot of marker genes defining the different tissue-resident leukocyte (TRL) cell cluster in the merged dataset from Fig. 1c. Dot size encodes percentage of cells expressing the gene, color encodes the average per cell gene expression level. c, Feature plots of selected marker genes used to identify cluster identities in the merged dataset from Fig. 1c. d, Uniform Manifold Approximation and Projection (UMAP) plots of the merged dataset from Fig. 1c split by tissue of origin. Corresponding cluster frequencies are shown in Fig. 1d.

Source data

Extended Data Fig. 2 Transcriptome analysis of dura-resident leukocytes during neuroinflammation in mice.

a, Merged Uniform Manifold Approximation and Projection (UMAP) plot of tissue-resident leukocytes (TRL) isolated from the dura merged of naive and EAE mice corresponding to the split UMAP plot depicted in Fig. 1h. b, Dotplot of marker genes defining the different cell clusters identified in the dura of naive and EAE mice as shown in A and in Fig. 1h. Dot size encodes percentage of cells expressing the gene, color encodes the average per cell gene expression level. c, Gene score feature plots depicting the expression of gene sets identified in ILC subtypes previously (Vivier et al. 2016; Robinette et al. 2015). d, Dot plot depicting the expression of plasma cell related genes in the Bc, cswBc, and BcProg cluster in healthy and EAE mice merged. Dot size encodes percentage of cells expressing the gene, color encodes the average per cell gene expression level. e, Stacked bar plots depicting cell cluster proportions in the scRNA-Seq dataset from naive vs. EAE mice as shown in Fig. 1h. f, Plots corresponding to Fig. 1i quantifying the change of cluster proportions in naive (left) vs. EAE (right) depicted on a linear scale to facilitate the log to linear transformation. g, Dot plot depicting the expression of plasma cell related genes in the Bc and cswBc cluster in naive vs. EAE mice. Dot size encodes percentage of cells expressing the gene, color encodes the average per cell gene expression level.

Source data

Extended Data Fig. 3 Neuroinflammation specifically alters T helper cell subsets in the dura.

a, Dot plot of selected marker genes to identify different T helper cell subsets in the Dura of naive vs. EAE mice, corresponding to Fig. 1h. Dot size encodes percentage of cells expressing the gene, color encodes the average per cell gene expression level. b, Representative flow cytometry gating strategy to identify Th1, Th17 and Treg cells isolated from Dura of naive (top row) and EAE mice at peak of disease (bottom row). c, Quantification of the percentage of cells in the respective gates as shown in B. Data represented as mean ± SD, n = 5, two-sided Mann-Whitney U test was used to calculate statistical significance, *p < 0.05 **p < 0.01 ***p < 0.001 (d) Representative flow cytometry gating strategy showing the Ki67 staining in CD19+B220+ Bc in Dura of naive (top row) vs. EAE mice at peak of disease (bottom disease). e, Quantification of the percentage of Ki67+ cells in the representative gates as shown D. Data represented as mean ± SD, n = 5 (c), n = 4 e, two-sided Mann-Whitney U test was used to calculate statistical significance, *p < 0.05 **p < 0.01 ***p < 0.001. f, Heatmap depicting the expression of gene signature (GSE28237) from follicular vs. early germinal center (GC) vs. late GC Bc in the Bc and cswBc cluster. Higher values mean higher overlap of the respective gene signature in cells located in the individual cluster.

Source data

Extended Data Fig. 4 Characterization of CSF and IgA+ B cells from naive vs. EAE mice.

a, Proportion (%) of Bc clones expressing any Ighg (for example Ighg2b, Ighg2c, Ighg3) gene and Igha, Ighd, Ighm genes as identified by BCR reconstruction from 3′ single cell RNA-sequencing libraries from naive vs. EAE mice (Methods). Bc clones were classified based on the number of cells expressing identical clonotype sequences into categories Single (0–1 cells), Small (2–5 cells), Medium (6–10 cells), Large (11–25 cells). Related to Supplementary Table 12. b, Total number of IgA+CD19+ Bc and percentage of IgA+CD19+ Bc of tissue-resident leukocytes identified by flow cytometry in the dura from naive vs. EAE mice. Data represented as mean ± SD, n = 6, two-sided Mann-Whitney U test was used to calculate statistical significance, *p < 0.05 **p < 0.01. c, Representative flow cytometry gating strategy to identify IgA expressing Bc in the dura of naive mice (top row) and in the dura of EAE mice (bottom row). d, Representative flow cytometry gating strategy to identify lymphocytes in the CSF of naive mice (top row) and EAE mice (bottom row). e, Quantification of Bc and Tc proportions in the CSF of naive mice corresponding to D. mean ± SD, n = 5, two-sided Mann-Whitney U test was used to calculate statistical significance (f) Quantification of Bc, Tc and CD4+ Tc in CSF of naive and EAE mice corresponding to D. mean ± SD, n = 5 naive, n = 6 EAE, two-sided Mann-Whitney U test was used to calculate statistical significance, P-value not depicted = not significant.

Source data

Extended Data Fig. 5 B cell progenitors identified in Dura.

a, Additional zoomed feature plots of key B cell (Bc) progenitor markers in the dura of naive (WT) Lewis rats as shown in Fig. 3c. b, Schematic representation of marker gene expression during B cell maturation stages. Figure created using Servier Medical Art templates. c, Heatmap of cells from the ‘pre Bc’ and the ‘late pro Bc’ clusters shown in Fig. 3a submitted to the mouse cell atlas (MCA). All cell types from the MCA reference dataset that did not contain ‘B cell’ in their cell type annotation were excluded. Each column represents one cell barcode, each row one MCA reference dataset. Colors indicate Pearson correlation coefficient between the filtered MCA reference dataset and the submitted cells.

Extended Data Fig. 6 B progenitor cells populate the dura of naive mice.

a-e, 4% PFA fixed, whole mount dura of naive C57BL/6 mice stained for Cd19 (a) or Cd79a b, for Cd19 and Dntt c, for Cd19, Dntt and Igll1 d, and for Cd19, Dntt and Ezh2 e, by RNA ISH as described in the methods. Nuclei were stained with DAPI. Please note that each dot represents a single RNA molecule. An indication of where pictures were taken is depicted in each individual picture. Scale bars 20 μm (left) and 10 μm (insets). One representative staining of n = 2 mice a,b, and n = 3 mice c-e, is shown.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–16 and Supplementary Table 14.

Reporting Summary

Supplementary Table 1

Technical information on scRNA-seq samples and sequencing. STDEV, standard deviation; SEM, standard error of the mean.

Supplementary Table 2

Marker genes of merged six tissue rat dataset (corresponding to Fig. 1c,d). Marker genes were calculated in a one versus all comparison. Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. Only genes with a positive average log FC are shown. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction). Cluster names correspond to Fig. 1.

Supplementary Table 3

Cell cluster proportions of the merged six tissue rat dataset (corresponding to Fig. 1c,d). Relative cell cluster proportions are given in percentages.

Supplementary Table 4

Marker genes of individual six tissue rat dataset (corresponding to Supplementary Figs. 1a–f and 2a–f). Marker genes were calculated in a one versus all comparison. Average log FC threshold was set to 0.25 for all tissues except for ‘cns’ (average log FC set to 0.1 here due to lower differential gene expression). Genes with an adjusted P value above 0.05 were removed. Only genes with a positive average log FC are shown. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Supplementary Table 5

Cell cluster proportions of individual six tissue rat dataset (corresponding to Supplementary Figs. 1a–f and 2a–f). Relative cell cluster proportions are given in percentages. Each excel sheet shows cell cluster proportions of one tissue.

Supplementary Table 6

DE genes of merged six tissue rat dataset one versus all tissue. Differentially expressed genes of all cells in one tissue versus all cells of all remaining tissues are shown. Each sheet shows the comparison of one tissue versus all remaining tissues. A positive average log FC value demonstrates a higher expression in the respective tissue compared with the remaining tissues. Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Supplementary Table 7

DE genes of merged six tissue rat dataset one versus one tissue. Differentially expressed genes of all cells in one tissue (CNS, dura or SDM) versus all cells of another tissue (SDM, blood or CP) are shown. Each sheet shows the comparison of one tissue versus another tissue. A positive average log FC value demonstrates a higher expression in the first tissue compared with the second tissue. Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Supplementary Table 8

DE genes of merged six tissue rat dataset one versus one tissue in cluster Bc 1. Differentially expressed genes of cluster Bc 1 in CNS-associated border compartments versus blood or dura versus all remaining clusters are shown. A positive average log FC value demonstrates a higher expression in the respective first group (border compartments or dura) compared with the second group (blood or all remaining tissues). Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Supplementary Table 9

Marker genes of EAE dataset (corresponding to Fig. 1h and Extended Data Fig. 2e). Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. Only genes with a positive average log FC are shown. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Supplementary Table 10

Absolute and relative cell cluster proportions of the EAE dataset (corresponding to Fig. 1h and Extended Data Fig. 9e). Relative cell cluster proportions are given in percentages.

Supplementary Table 11

DE genes of EAE dataset EAE versus naive. Differentially expressed genes of EAE versus naive. Clusters with less than ten cells were removed. Each sheet shows the DE genes of one cluster. A positive average log FC value demonstrates a higher expression in EAE compared with control. Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Supplementary Table 12

Abundance of BCR clonotypes in naive versus EAE (corresponding to Extended Data Fig. 4a). Number of the individual BCR clonotypes identified by BCR sequencing in the naive versus EAE dataset. CTgene, BCR gene sequence; Abundance, number of B cells with the respective CTgene expressed; Condition, condition of mouse (naive versus EAE) the respective clone was found in.

Supplementary Table 13

Marker genes of a published dura dataset (corresponding to Supplementary Fig. 12a). Published dura-derived single-cell transcriptomes8 were downloaded and clustered and marker genes were calculated in one versus all comparisons. Average log FC threshold was set to 0.25; genes with an adjusted P value above 0.05 were removed. Only genes with a positive average log FC are shown. avg_log_FC, log fold change of the average expression between the cluster versus all remaining clusters; pct.1, percentage of cells with the gene detected in the cluster; pct.2, percentage of cells with the gene detected in all remaining clusters; p_val_adj, adjusted P values (based on Bonferroni correction).

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed IHC images.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed IHC images.

Source Data Fig. 4

Unprocessed IHC images.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed IHC images.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Schafflick, D., Wolbert, J., Heming, M. et al. Single-cell profiling of CNS border compartment leukocytes reveals that B cells and their progenitors reside in non-diseased meninges. Nat Neurosci 24, 1225–1234 (2021). https://doi.org/10.1038/s41593-021-00880-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-021-00880-y

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