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:

EBV induces CNS homing of B cells attracting inflammatory T cells

Abstract

Epidemiological data have identified Epstein–Barr virus (EBV) infection as the main environmental risk factor for multiple sclerosis, the predominant autoimmune disease of the central nervous system (CNS)1. However, how EBV infection initiates multiple sclerosis pathogenesis remains unclear. Here we demonstrate that EBV expands oligoclonal T-bet+CXCR3+ B cells that home to the CNS in humanized mice. Effector memory CD8+ T cells and CD4+ TH1 cells as well as CD4+ TH17 cells co-migrate to the brain of EBV-infected humanized mice. T-bet+CXCR3+ B cells can colonize submeningeal brain regions in the absence of other lymphocytes and attract T cells. Depletion of B cells with rituximab or blocking of CXCR3 significantly decreases lymphocyte infiltration into the CNS. Thus, we suggest that symptomatic primary EBV infection generates B cell subsets that gain access to the CNS, attract T cells and thereby initiate multiple sclerosis.

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: EBV infection drives T cell infiltration into the CNS in a new humanized mouse model.
Fig. 2: EBV infection is linked to T-bet+CXCR3+ B cell expansion.
Fig. 3: EBV infection drives oligoclonal expansion of CNS-homing T-bet+ B cells.
Fig. 4: EBV-infected B cells independently home to the brain and attract T cells.
Fig. 5: Depletion of B cells during EBV infection diminishes T cell infiltrates in the brain.

Similar content being viewed by others

Data availability

The scRNA-seq datasets analysed in this study are available from the Gene Expression Omnibus (Fig. 3; GSE299939) and Zenodo50 (Extended Data Fig. 5; https://doi.org/10.5281/zenodo.15602185). The multiplex immunofluorescence images are also available to download from Zenodo51 (https://doi.org/10.5281/zenodo.15599580).

Code availability

The code used to analyse scRNA-seq data and multiplex immunofluorescence images can be found via GitHub (https://github.com/EannaFennell/EBV-induces-CNS-homing-of-B-cells-attracting-inflammatory-T-cells). The package used for multiplex immunofluorescence analysis is also available from GitHub (https://github.com/EannaFennell/MISSILe).

References

  1. Bjornevik, K. et al. Longitudinal analysis reveals high prevalence of Epstein–Barr virus associated with multiple sclerosis. Science 375, 296–301 (2022).

    Article  ADS  CAS  PubMed  Google Scholar 

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

    Article  PubMed  Google Scholar 

  3. Oksenberg, J. R., Baranzini, S. E., Sawcer, S. & Hauser, S. L. The genetics of multiple sclerosis: SNPs to pathways to pathogenesis. Nat. Rev. Genet. 9, 516–526 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Bray, P. F., Bloomer, L. C., Salmon, V. C., Bagley, M. H. & Larsen, P. D. Epstein–Barr virus infection and antibody synthesis in patients with multiple sclerosis. Arch. Neurol. 40, 406–408 (1983).

    Article  CAS  PubMed  Google Scholar 

  5. Leibowitz, U. et al. Epidemiological study of multiple sclerosis in Israel. II. Multiple sclerosis and level of sanitation. J. Neurol. Neurosurg. Psychiatry 29, 60–68 (1966).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Angelini, D. F. et al. Increased CD8+ T cell response to Epstein–Barr virus lytic antigens in the active phase of multiple sclerosis. PLoS Pathog. 9, e1003220 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Lünemann, J. D. et al. Increased frequency and broadened specificity of latent EBV nuclear antigen-1-specific T cells in multiple sclerosis. Brain 129, 1493–1506 (2006).

    Article  PubMed  Google Scholar 

  8. Lanz, T. V. et al. Clonally expanded B cells in multiple sclerosis bind EBV EBNA1 and GlialCAM. Nature 603, 321–327 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  9. Bar-Or, A. et al. Rituximab in relapsing-remitting multiple sclerosis: a 72-week, open-label, phase I trial. Ann. Neurol. 63, 395–400 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. 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  PubMed  Google Scholar 

  11. Hauser, S. L. et al. Ocrelizumab versus interferon β-1a in relapsing multiple sclerosis. N. Engl. J. Med. 376, 221–234 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Rubtsova, K. et al. B cells expressing the transcription factor T-bet drive lupus-like autoimmunity. J. Clin. Invest. 127, 1392–1404 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Punnanitinont, A. et al. TLR7 activation of age-associated B cells mediates disease in a mouse model of primary Sjogren’s disease. J. Leukoc. Biol. 115, 497–510 (2024).

    Article  CAS  PubMed  Google Scholar 

  14. Tsubaki, T. et al. Accumulation of plasma cells expressing CXCR3 in the synovial sublining regions of early rheumatoid arthritis in association with production of Mig/CXCL9 by synovial fibroblasts. Clin. Exp. Immunol. 141, 363–371 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Fox, R. I., Luppi, M., Kang, H. I. & Pisa, P. Reactivation of Epstein–Barr virus in Sjogren’s syndrome. Springer Semin. Immunopathol. 13, 217–231 (1991).

    Article  CAS  PubMed  Google Scholar 

  16. James, J. A. et al. Systemic lupus erythematosus in adults is associated with previous Epstein–Barr virus exposure. Arthritis Rheum. 44, 1122–1126 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Alspaugh, M. A., Jensen, F. C., Rabin, H. & Tan, E. M. Lymphocytes transformed by Epstein–Barr virus. Induction of nuclear antigen reactive with antibody in rheumatoid arthritis. J. Exp. Med. 147, 1018–1027 (1978).

    Article  CAS  PubMed  Google Scholar 

  18. SoRelle, E. D. et al. Time-resolved transcriptomes reveal diverse B cell fate trajectories in the early response to Epstein–Barr virus infection. Cell Rep. 40, 111286 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mouat, I. C. et al. Gammaherpesvirus infection drives age-associated B cells toward pathogenicity in EAE and MS. Sci. Adv. 8, eade6844 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  20. Miller, G. & Lipman, M. Comparison of the yield of infectious virus from clones of human and simian lymphoblastoid lines transformed by Epstein–Barr virus. J. Exp. Med. 138, 1398–1412 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Miller, G. & Lipman, M. Release of infectious Epstein–Barr virus by transformed marmoset leukocytes. Proc. Natl Acad. Sci. USA 70, 190–194 (1973).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zdimerova, H. et al. Attenuated immune control of Epstein–Barr virus in humanized mice is associated with the multiple sclerosis risk factor HLA-DR15. Eur. J. Immunol. 51, 64–75 (2021).

    Article  CAS  PubMed  Google Scholar 

  23. van Langelaar, J. et al. Induction of brain-infiltrating T-bet-expressing B cells in multiple sclerosis. Ann. Neurol. 86, 264–278 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Jain, R. W. & Yong, V. W. B cells in central nervous system disease: diversity, locations and pathophysiology. Nat. Rev. Immunol. 22, 513–524 (2022).

    Article  CAS  PubMed  Google Scholar 

  25. SoRelle, E. D. et al. An EBV-associated atypical B cell signature in clinically isolated syndrome is implicated in progression of multiple sclerosis. Preprint at medRxiv https://doi.org/10.1101/2023.02.26.23286433 (2024).

  26. Fournier, B. et al. Inherited TNFSF9 deficiency causes broad Epstein–Barr virus infection with EBV+ smooth muscle tumors. J. Exp. Med. 219, e20211682 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Baranzini, S. E. et al. B cell repertoire diversity and clonal expansion in multiple sclerosis brain lesions. J. Immunol. 163, 5133–5144 (1999).

    Article  CAS  PubMed  Google Scholar 

  28. Yang, X. et al. Liquid–liquid phase separation of RBM33 facilitates hippocampus aging by inducing microglial senescence by activating CDKN1A. Int. J. Biol. Macromol. 310, 142986 (2025).

    Article  CAS  PubMed  Google Scholar 

  29. Evans, A. K. et al. Impact of noradrenergic inhibition on neuroinflammation and pathophysiology in mouse models of Alzheimer’s disease. J. Neuroinflammation 21, 322 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Reali, C. et al. B cell rich meningeal inflammation associates with increased spinal cord pathology in multiple sclerosis. Brain Pathol. 30, 779–793 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Choi, S. R. et al. Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis. Brain 135, 2925–2937 (2012).

    Article  PubMed  Google Scholar 

  32. McHugh, D. et al. EBV renders B cells susceptible to HIV-1 in humanized mice. Life Sci. Alliance 3, e202000640 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  33. White, R. E. et al. EBNA3B-deficient EBV promotes B cell lymphomagenesis in humanized mice and is found in human tumors. J. Clin. Invest. 122, 1487–1502 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Münz, C. Latency and lytic replication in the oncogenesis of the Epstein Barr virus. Nat. Rev. Microbiol. 17, 691–700 (2019).

    Article  PubMed  Google Scholar 

  35. Babcock, J. G., Hochberg, D. & Thorley-Lawson, A. D. The expression pattern of Epstein–Barr virus latent genes in vivo is dependent upon the differentiation stage of the infected B cell. Immunity 13, 497–506 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. SoRelle, E. D., Reinoso-Vizcaino, N. M., Horn, G. Q. & Luftig, M. A. Epstein–Barr virus perpetuates B cell germinal center dynamics and generation of autoimmune-associated phenotypes in vitro. Front. Immunol. 13, 1001145 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hochberg, D. et al. Demonstration of the Burkitt’s lymphoma Epstein–Barr virus phenotype in dividing latently infected memory cells in vivo. Proc. Natl Acad. Sci. USA 101, 239–244 (2004).

    Article  ADS  CAS  PubMed  Google Scholar 

  38. Nowag, H. et al. Macroautopphagy proteins assist Epstein Barr virus production and get incorporated into the virus particles. EBioMedicine 1, 116–125 (2014).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  39. Lünemann, J. D. et al. EBNA1-specific T cells from patients with multiple sclerosis cross react with myelin antigens and co-produce IFN-γ and IL-2. J. Exp. Med. 205, 1763–1773 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  40. Serafini, B., Rosicarelli, B., Veroni, C., Mazzola, G. A. & Aloisi, F. Epstein–Barr virus-specific CD8 T cells selectively infiltrate the brain in multiple sclerosis and interact locally with virus-infected cells: clue for a virus-driven immunopathological mechanism. J. Virol. https://doi.org/10.1128/JVI.00980-19 (2019).

  41. Gottlieb, A., Pham, H. P. T., Saltarrelli, J. G. & Lindsey, J. W. Expanded T lymphocytes in the cerebrospinal fluid of multiple sclerosis patients are specific for Epstein–Barr-virus-infected B cells. Proc. Natl Acad. Sci. USA 121, e2315857121 (2024).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Thomas, O. G. et al. Cross-reactive EBNA1 immunity targets α-crystallin B and is associated with multiple sclerosis. Sci. Adv. 9, eadg3032 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tengvall, K. et al. Molecular mimicry between anoctamin 2 and Epstein–Barr virus nuclear antigen 1 associates with multiple sclerosis risk. Proc. Natl Acad. Sci. USA 116, 16955–16960 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sahir, F., Mateo, J. M., Steinhoff, M. & Siveen, K. S. Development of a 43 color panel for the characterization of conventional and unconventional T-cell subsets, B cells, NK cells, monocytes, dendritic cells, and innate lymphoid cells using spectral flow cytometry. Cytometry A https://doi.org/10.1002/cyto.a.24288 (2020).

  45. Puccio, S. et al. CRUSTY: a versatile web platform for the rapid analysis and visualization of high-dimensional flow cytometry data. Nat. Commun. 14, 5102 (2023).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  46. Adhikary, D. et al. Immunodominance of lytic cycle antigens in Epstein–Barr virus-specific CD4+ T cell preparations for therapy. PLoS ONE 2, e583 (2007).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  47. Nuckel, J. et al. Association between IgG responses against the nucleocapsid proteins of alphacoronaviruses and COVID-19 severity. Front. Immunol. 13, 889836 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Soldan, S. S. et al. Epigenetic plasticity enables CNS-trafficking of EBV-infected B lymphocytes. PLoS Pathog. 17, e1009618 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Oner, A. & Kobold, S. Transwell migration assay to interrogate human CAR-T cell chemotaxis. STAR Protoc. 3, 101708 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Fennell, E. Single-cell RNA and B cell receptor sequencing processed data from EBV-infected and PBS humanised BRGS-A2DR2 mice. Zenodo https://doi.org/10.5281/zenodo.15602185 (2025).

  51. Fennell, E. Multiplex immunofluorescence images of spleens from EBV-infected BRGS-A2DR2 mice. Zenodo https://doi.org/10.5281/zenodo.15599580 (2025).

Download references

Acknowledgements

We thank the Functional Genomics Center Zürich of the University of Zürich and ETH Zurich, and in particular Q. Zhang, for support on the genomics analyses; K. Gehrmann for technical support with the humanized mice; J. Fullin for creating the graphical illustrations featured in the figures; and F.L. thanks L. Yuka for ongoing support. This research was in part financially supported by the Swiss National Science Foundation (310030_204470/1, 310030L_197952/1 and CRSII_222718_10000065), Cancer Research Switzerland (KFS-5896-08-2023-R), the Swiss MS Society (2023-17), the Swiss State Secretariat for Education, Research and Innovation for EU Horizon BEHIND-MS, CRPP-ImmunoCure of the University of Zürich, the Sobek Foundation, the Swiss Vaccine Research Institute, the Vontobel Foundation, Roche, Novartis and Viracta. L.R. was supported by career advancement grants from the University of Zurich (Forschungskredit, FK-21-034). É.F. is funded by HORIZON-TMA-MSCA-PF-GF (no. 101110227). G.F. is funded by Next Generation EU — NRRP M6C2 — Investment 2.1 Enhancement and strengthening of biomedical research in the NHS — PNRR-MCNT2-2023-12378122; CUP: C43C23000880006.

Author information

Authors and Affiliations

Authors

Contributions

F.L., I.P., É.F., S.S., L.R. and J. Mietz performed the in vivo experiments. F.L., I.P., E.B. and C.C. performed the in vitro EBV infection and transwell migration experiments. E.B. and A.M. performed the ChipCytometry experiments. É.F. and P.G. performed the bioinformatics analysis. I.L.-P., L.B., A.G., K.E. and J.S. performed light-sheet microscopy. M.L. and J. Mautner performed serological assays for viral and autoantigen recognition by IgM. G.P. and G.F. performed HLA typing. C.M. supervised the study. F.L., E.B. and C.M. wrote the first draft of the manuscript. All authors edited the manuscript.

Corresponding author

Correspondence to Christian Münz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Deepak Rao and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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 figures and tables

Extended Data Fig. 1 T cell infiltration into the CNS and associated NfL release.

a, Weekly blood viral loads (IU/ml blood) of EBV-infected humanized NSG and BRGS-A2DR2 mice (p-value = 0.010256; NSG = 8, BRGS-A2DR2 = 8). b, IHC staining of EBV-infected BRGS-A2DR2 tongue and spleen sections, scale bar=100μm. c, Weekly blood viral loads (IU/ml) of EBV-infected humanized BRGS-A2DR2 mice (n = 28). d, Correlation between brain EBV viral load and bioluminescent signal (n = 23). e, UMAP of marker expression by T cell populations. f, Percentage of the indicated T cell differentiation subpopulations. g, Frequency of CD19+ B cells in brains of PBS or EBV-infected mice (PBS n = 20, EBV n = 23). h-j, Total numbers of CD3+, activated HLA-DR+CD4+ (p-value = 0.0435), and activated HLA-DR+CD8+ T cells (p-value = 0.0002) in brains of PBS or EBV-infected mice (PBS n = 15, EBV n = 15). k-l, Frequencies of HLA-DR+CD8+ (p-value < 0.0001) and HLA-DR+CD4+ T cells of PBS or EBV-infected mouse spleens (PBS n = 20, EBV n = 23). m-n, Frequency of CD4+ (naïve p-value = 0.0001, effector memory p-value < 0.0001) and CD8+ (naïve p-value < 0.0001, effector memory p-value < 0.0001) T cell differentiation subtypes of PBS or EBV-infected mouse spleens. o-p, Correlation between HLA-DR+CD4+ T cells and alanine aminotransferase activity or enzymatic creatinine concentrations in blood (n = 18). q, ELISA of neurofilament light chain (NfL) in plasma before (Baseline) and 6 weeks post-EBV infection (Week 6; p-value < 0.0001). r, Fold increase of plasma NfL levels before and 6 weeks post-EBV infection or PBS injection. For all correlations, solid line represents trend line obtained by linear regression, the dotted lines indicate 95% confidence bands, two-tailed p-value by Spearman correlation. Unless otherwise indicated, p-values by two-tailed MWU test. a, was generated from one experiment using mice reconstituted from one human CD34+ hematopoietic progenitor cells donor. b-m, were generated from 3 independent experiments with 4 donors. d, was created with crusty.humanitas.it. Error bar indicates SEM; each dot represents one single animal. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 2 Characteristics and origin of EBV expanded T-bet+CXCR3+ B cells.

a, Representative flow cytometric contour plots of T-bet+CXCR3+ and T-bet+CD11c+ B cell gating of splenocytes from PBS or EBV-infected humanized mice. b, Frequencies of CD19+CD11c+ B cells in blood, spleen and brains of PBS or EBV-infected humanized mice (PBS n = 21, EBV n = 28). c, Frequencies of CD19+CD11c+T-bet+ B cells in blood (p-value = 0.0277), spleen and brains of PBS or EBV-infected humanized mice (PBS n = 21, EBV n = 28). d, Total numbers of CD19+T-bet+CD11c+ B cells in blood, spleen and brains of PBS or EBV-infected humanized mice (PBS n = 21, EBV n = 28). e, Frequency of T-bet+CXCR3+ and T-bet-CXCR3- cells among CD11c+ B cells in blood (p-value = 0.0004), spleen (p-value < 0.0001) and brains of EBV infected mice. f, Frequency of T-bet+CXCR3+ and T-bet-CXCR3- cells among CD11c+ B cells in blood (p-value < 0.0001), spleen (p-value < 0.0001) and brains of PBS mice. g, In vitro EBV infection of CXCR3+ and CXCR3- B cells monitoring T-bet/CXCR3 co-expression frequency over time (p-values from left to right: 0.0211, 0.0160, 0.0016, 0.0007, 0.0039, <0.0001). h, In vitro EBV infection of CXCR3+ and CXCR3- B cells monitoring CD11c frequency over time (p-value = 0.0461). i, Growth assay of established LCLs derived from CXCR3+ or CXCR3- B cells over time depicted as total numbers. g-i, p-values were calculated using mixed-effects model with Geisser-Greenhouse correction followed by Sidak’s multiple comparisons test. Unless otherwise indicated, p-values were calculated using the two-tailed MWU test. Data from figures b-f, was generated from 5 independent experiments with mice reconstituted from 6 different human CD34+ hematopoietic progenitor cells donors. Error bar indicates SEM; each dot represents one single animal. Data from Figure g-i was generated from 3 independent experiment using 3 B cells donors. Error bar indicates SEM; dots represent mean of all 3 donors. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 3 Spatial analysis of T-bet+CXCR3+ B cells and activated T cells.

a, ChipCytometry of DNA (Helix-NP), CD39, CD45, CD11c and Ki67 on splenic section of PBS and EBV-infected mice. Scale bar=500μm. b, Frequency of T-bet+CXCR3+ of total B cells in spleen of PBS and EBV-infected mice (PBS n = 1, EBV n = 1). c, Frequency of EBNA2+ among T-bet+CXCR3+ and non-T-bet+CXCR3+ (other) B cells in spleen of EBV infected mice (PBS n = 1, EBV n = 1). d, Cell type enrichment per calculated tissue neighbourhood using HLA-DR as activation marker and e, frequency of respective tissue neighbourhoods (N-1 to N-5) in PBS and EBV-infected spleens (PBS n = 1, EBV n = 1). f-g, Frequency of activated CD4+ and CD8+ T cells (HLA-DR+) in the immediate microenvironment (ME; 100μm) of T-bet+CXCR3+ B cells in PBS and EBV-infected spleens (PBS n = 6, EBV n = 92 microenvironments of 1 experiment). P-values were calculated using two-sided MWU test (p-values = 0.00062 & 0.000045 respectively). h-i, Frequency of proliferative CD4+ and CD8+ T cells (Ki67+) in the immediate microenvironment (100μm) of T-bet+CXCR3+ B cells in PBS and EBV-infected (PBS n = 6, EBV n = 92 microenvironments of 1 experiment). P-values were calculated using two-sided MWU test (p-values = 0.0001 & 0.00014 respectively). Box plots define the medium value (centre), the interquartile range (25th and 75th percentiles) as the boundaries of the box and whiskers which extend to 1.5 times the 25th and 75th percentiles. Outliers outside the boundaries of the whiskers are displayed as individual points. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. j, Cell type enrichment per calculated tissue neighbourhood using Ki67 as a T cell proliferation marker and k, frequency of respective tissue neighbourhoods (N*-1 to N*-6) in PBS and EBV-infected spleens. l, 3D multiplex imaging by light sheet microscopy of cleared brains of two EBV-infected mice (EBV n = 3, PBS n = 1) stained for human CD3 (magenta), human CD45 (cyan) and mouse CD31 (yellow).

Extended Data Fig. 4 Transcriptional characteristics of T-bet+CXCR3+ B cells.

a, UMAP of T-bet+CXCR3+ among splenic B cells of EBV-infected humanized mice by scRNA-seq. b-c, Stacked bar plots showing (left) the CD19+ B cell population frequency and (right) immunoglobulin isotype distribution within T-bet+CXCR3+ and non-T-bet+CXCR3+ (other) cells. d, Expression of indicated Ig isotypes and e, expression of indicated EBV transcripts. f, Heatmap of shared clones between B cell subtypes with Morisita overlap index. g-h, Violin plot of expression of MKI67 (adjusted p-value = 1.41e-260) and pro-proliferative gene module (MKI67, PCNA, TOP2A, CCNB1, CCNB2, CDK1; adjusted p-value = 2.64e-157) between infected (EBV+) and un-infected (EBV-) T-bet+CXCR3+ B cells. i-j, Violin plot of expression of MKI67 (adjusted p-value = 2.105e-86) and pro-proliferative gene module (MKI67, PCNA, TOP2A, CCNB1, CCNB2, CDK1; adjusted p-value = 2.86e-32) between clonally expanded and diverse BCR expressing T-bet+CXCR3+ B cells. k, Violin plot of expression of TBX21 between infected (EBV+) and un-infected (EBV-) CXCR3+ B cells (adjusted p value = 1.706e-29). l, IGHV gene expression among the top 50 expanded B cell clones. IGHV4-34 and IGHV4-39 expressing clones are highlighted. The black line defines the mean expression of each group from g-j. P-values for differential gene expression analysis between groups were calculated using the two-sided MWU test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 5 CNS accumulation and EBV antigen recognition of oligoclonally EBV expanded B cells.

a, tSNE of single cell RNA sequencing (scRNA-seq) of sorted CD19+ B cells from spleen and perfused brains of EBV infected humanized BRGS-A2DR2 mice (n = 10) 6 weeks after EBV infection with manually annotated B cell differentiation clusters. b, BCR receptor clonality analysis. Clones were defined by Single (0 < X < = 1), Small (1 < X < = 5), Medium (5 < X < = 20), Large (20 < X < = 50), Hyperexpanded 1 (50 < X < = 200). c, Z-scored heatmap of 20 most expanded BCRs in matched perfused brain and spleen samples of the 10 analyzed mice (mouse #9, 12, 14, 17, 2, 21, 23, 4, 13, 8). d, Z-scored heatmap of IgM responses to the indicated EBV gene products in blood plasma of the same 10 by scRNA-seq analyzed EBV infected mice and three PBS controls (mouse #1, 6, 10). Plasma of mouse 1 and 10 was pooled due to low individual blood plasma volume.

Extended Data Fig. 6 Oligoclonal B cell expansion and autoantigen recognition after EBV infection.

a, Stacked bar plot showing B cell population frequency in spleen and brain of EBV-infected mice. b, tSNE of combined expression of all EBV transcripts split by organ of origin (spleen and brain). c, Stacked bar plot showing clonality distribution of non-naïve clusters (i.e. memory B cells, cycling B cells, plasmablasts & plasma cells) in spleen and brain of EBV-infected mice only. d-e, tSNE of sorted CD19+ B cell population colored by B cell populations (d) and BCR clonality (e), split by organ of origin (spleen and brain) of PBS mice only. Clones were defined by Single (0 < X < = 1), Small (1 < X < = 5), Medium (5 < X < = 20), Large (20 < X < = 50), Hyperexpanded 1 (50 < X < = 200). f, Shared spleen and brain BCR clones heatmap of individual PBS mice (most abundant 20 BCRs are plotted). BCR frequencies were calculated from total B cells per organ per mouse and were log2 normalised. g, TNFSF9 (encodes 4-1BBL/CD137L) expression in spleen and brain memory B cells. h, Distribution of IGHV gene in spleen versus brain B cells. The linear regression line (which represents the fitted values/mean predicted response from the linear model) and 95% confidence interval (in grey) are plotted. i, Blood plasma IgM antibody specificities against known autoantigens of all mice used for the scRNA-seq analysis. j, Replicate serum antibody specificities against EBV proteins and known autoantigens from pooled mice of previous EBV infection experiments. Pool #1 consisted of 2 mice (EBV infected), pool #2 and #3 consisted of 3 mice each (EBV infected), pool #4 from 3 mice (PBS controls). Pooled mice originated from individual CD34+ hematopoietic progenitor cell donors.

Extended Data Fig. 7 CNS homing and T cell attraction by EBV infected T-bet+CXCR3+ B cells.

a, Heatmap of indicated LCL marker frequency. b, Representative T-bet staining on LCL. c, Total mouse numbers with neuroinvasion. d-e, IHC staining of EBNA2 in indicated (f) meningeal and parenchymal brain areas. Scale bar=50μm. g, EBNA2+ cell frequency (n = 3 per region). h, T-bet and CXCR3 frequency of T-betlo and T-bethi. i, Correlation between blood CXCR3 frequency and neuroinvasion (n = 23). j, Correlation between blood T-bet frequency and neuroinvasion (n = 23). k, T-bet and CXCR3 frequency of LCL used for CXCR3 inhibition (TAK-779). l, EBV viral loads (IU/brain; T-betlo n = 12, T-bethi n = 11). m-n, Brain viral loads by bioluminescence after CXCR3 inhibition (TAK-779; PBS n = 6, CXCR3 antagonist n = 7) or Natalizumab treatment (p-value = 0.0401; PBS n = 8, Natalizumab n = 7). o, Transwell migration assays with indicated LCL supernatants. p, Frequency of migrated HLA-DR+CD8+ T cells (LCL spleen p-value = 0.0015). q, Transwell migration assays with LCL supernatant (p-value = 0.0015) with CCL3/4/5 (p = 0.0366) or CXCR3/CCR5 inhibition (TAK-779; p-value = 0.0125). r, Transwell migration assays with CXCL9+ medium or wild-type vs. EBNA3B deficient LCL supernatants (p-value = 0.0292). s, ELISA on wild-type and EBNA3B deficient LCL supernatants (p-value = 0.0004). t, Transwell migration assays with T-bethi (p-value = 0.0372) or T-betlo LCL supernatants. Each dot represents one well. For all correlations, solid line represents trend line obtained by linear regression, the dotted lines indicate 95% confidence bands two-tailed p-value was calculated using Spearman correlation. Data from figure a-c was generated from 3 independent experiments and h-n from 2 independent experiments with 2 LCL donors. o-r and t were tested with =>3 T cells donors. p-values were calculated using one-way ANOVA with Dunnett’s multiple comparison test. s, was generated using supernatant of n = 3 wild-type (WT) and n = 3 EBNA3B deficient LCLs (EBNA3A KO) in 2 independent experiments, p-value was calculated with two-way ANOVA followed by Sidak’s multiple comparisons test. Error bar indicates SEM. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 8 CD20 targeted B cell depletion.

a-b, Weekly total numbers of CD45+CD19+ or CD20+ B cells in blood of EBV-infected mice with and without Rituximab treatment (PBS n = 5, EBV n = 4, Rituximab n = 7). p-values from left to right: CD20:0.0026, 0.0003,0.029,0.0120CD19: 0.0014,0.0004.0.0235,0.0110, (two-way mixed effects model with Geisser-Greenhouse correction with Tukey’s multiple comparisons test). Dots present mean and error bars SEM. c, Frequency and d, total numbers of CD19+ B cells in spleen (p-value < 0.0001) and brain with and without Rituximab treatment. e, Frequency of CD20+ B cells in spleen (p-value = 0.0007) and brain (p-value = 0.0011) with and without Rituximab treatment. f, Total numbers of CD20+ B cells in spleen (p-value = 0.0007) and brain (p-value = 0.0217) with and without Rituximab treatment. g, In vivo bioluminescence signal of perfused brains (PBS n = 5, EBV n = 4, Rituximab n = 7). h, Total numbers of CD19+CD11c+T-bet+CXCR3+ B cells in the blood (p-value < 0.0001), spleen (p-value = 0.0002), and brain (p-value = 0.0201) with and without Rituximab treatment (PBS n = 5, EBV n = 4, Rituximab n = 7). i, Total numbers of CD4+ T cells in the brain with and without Rituximab treatment (EBV n = 4, Rituximab n = 7). j, Correlation between activated HLA-DR+ CD4+ T cells and T-bet+CXCR3+ B cells in the brains of EBV infected humanized mice. Solid line represents trend line obtained by linear regression, the dotted lines indicate 95% confidence bands (n = 12). k, Total numbers of CD4+ T cell subtypes, including naïve (CD45Ra+CCR7+), effector memory (CD45Ra-CCR7-), central memory (CD45Ra-CCR7+) and Temra (CD45Ra+CCR7-) cells, with and without Rituximab treatment (EBV n = 4, Rituximab n = 7). a-k, were generated from 2 independent experiments with mice reconstituted from 2 different human CD34+ hematopoietic progenitor cell donors. Unless otherwise indicated, p-values were calculated using the two-tailed MWU test. Error bar indicates SEM; each dot represents one single animal. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Extended Data Fig. 9 Gating strategy for human T and B cell populations ex vivo.

a, Representative flow cytometric contour plots showing FACS gating strategy used for gating on lymphocytes, single cells, live cells and human CD45+ cells followed by CD3 and CD19 gating. b, Representative flow cytometric contour plots following gating of Extended Data Fig. 9a for T cell populations. Depicted gating strategy was used for following FACS data panels: Fig. 1h–p and Fig. 5k–p and Extended Data Figs. 1g–p and 8i-k. c, Representative flow cytometric contour plots for gating of Extended Data Fig. 9a for B cell populations. Depicted gating strategy was used for following FACS data panels: Figs. 2a–d4e and 5b,c,h–j and Extended Data Figs. 1f, 2b–h, 7a,h,k and 7a–f,h.

Extended Data Fig. 10 Gating strategy for human T and B cell populations for in vitro experiments.

a, Representative flow cytometric contour plots showing FACS gating strategy used for sorting CD19+ B cells for data in Fig. 3 and Extended Data Figs. 46. b, Representative flow cytometric contour plots showing FACS gating strategy used for sorting CD19+CXCR3+ B cells for Extended Data Fig. 2g,h. c, Representative flow cytometric contour plots showing FACS gating strategy used for migration assays in Fig. 4h,i,k,l and Extended Data Fig. 7o–r,t.

Supplementary information

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

Läderach, F., Piteros, I., Fennell, É. et al. EBV induces CNS homing of B cells attracting inflammatory T cells. Nature 646, 171–179 (2025). https://doi.org/10.1038/s41586-025-09378-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41586-025-09378-0

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