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:

Memory B cell subsets have divergent developmental origins that are coupled to distinct imprinted epigenetic states

Abstract

Memory B cells (MBCs) are phenotypically and functionally diverse, but their developmental origins remain undefined. Murine MBCs can be divided into subsets by expression of CD80 and PD-L2. Upon re-immunization, CD80/PD-L2 double-negative (DN) MBCs spawn germinal center B cells (GCBCs), whereas CD80/PD-L2 double-positive (DP) MBCs generate plasmablasts but not GCBCs. Using multiple approaches, including generation of an inducible GCBC-lineage reporter mouse, we demonstrate in a T cell-dependent response that DN cells formed independently of the germinal center (GC), whereas DP cells exhibited either extrafollicular (DPEX) or GCBC (DPGC) origins. Chromatin and transcriptional profiling revealed similarity of DN cells with an early memory precursor. Reciprocally, GCBC-derived DP cells shared distinct genomic features with GCBCs, while DPEX cells had hybrid features. Upon restimulation, DPEX cells were more prone to divide, while DPGC cells differentiated toward IgG1+ plasmablasts. Thus, MBC functional diversity is generated through distinct developmental histories, which imprint characteristic epigenetic patterns onto their progeny, thereby programming them for divergent functional responses.

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 transcriptomic and epigenetic profiles of FO NBCs and MBC subsets.
Fig. 2: Transcriptomic and epigenetic comparison of proliferating precursors and MBCs.
Fig. 3: GCs disproportionately produce DP MBCs.
Fig. 4: scRNA-seq identifies heterogeneity among DP MBCs, reflective of origin.
Fig. 5: GCBC-derived DP cells are less proliferative and more prone to AFC differentiation than extra-GCBC-derived DP cells in vitro.
Fig. 6: NP-Ficoll immunization produces DP MBCs independently of a GC reaction.
Fig. 7: CD40 stimulation in the absence of GCBCs produces DP MBCs.
Fig. 8: Proliferation and CD40 signals are not sufficient to establish the epigenetic identity of DPGC.

Similar content being viewed by others

Data availability

All bulk RNA-seq data have been deposited in the NCBI’s Gene Expression Omnibus (GEO) database and are publicly available under accession number GSE225748.

All bulk ATAC-seq data have been deposited in the NCBI GEO database and are publicly available under accession number GSE225672.

All scRNA-seq data have been deposited in the NCBI GEO database and are publicly available under accession number GSE225673.

The mm10 genome database (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/) was used to align sequences for the RNA-seq analysis and to align sequencing reads for the ATAC-seq analysis.

All raw data and materials will be made available to investigators upon request. Source data are provided with this paper.

References

  1. Anderson, S. M., Tomayko, M. M., Ahuja, A., Haberman, A. M. & Shlomchik, M. J. New markers for murine memory B cells that define mutated and unmutated subsets. J. Exp. Med. 204, 2103–2114 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tomayko, M. M., Steinel, N. C., Anderson, S. M. & Shlomchik, M. J. Cutting edge: hierarchy of maturity of murine memory B cell subsets. J. Immunol. 185, 7146–7150 (2010).

    Article  CAS  PubMed  Google Scholar 

  3. Zuccarino-Catania, G. V. et al. CD80 and PD-L2 define functionally distinct memory B cell subsets that are independent of antibody isotype. Nat. Immunol. 15, 631–637 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Weisel, F. J., Zuccarino-Catania, G. V., Chikina, M. & Shlomchik, M. J. A temporal switch in the germinal center determines differential output of memory B and plasma cells. Immunity 44, 116–130 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Taylor, J. J., Pape, K. A. & Jenkins, M. K. A germinal center-independent pathway generates unswitched memory B cells early in the primary response. J. Exp. Med. 209, 597–606 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Viant, C. et al. Germinal center-dependent and -independent memory B cells produced throughout the immune response. J. Exp. Med. 218, e20202489 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Shinnakasu, R. et al. Regulated selection of germinal-center cells into the memory B cell compartment. Nat. Immunol. 17, 861–869 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Glaros, V. et al. Limited access to antigen drives generation of early B cell memory while restraining the plasmablast response. Immunity 54, 2005–2023 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Inamine, A. et al. Two waves of memory B-cell generation in the primary immune response. Int Immunol. 17, 581–589 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Takemori, T., Kaji, T., Takahashi, Y., Shimoda, M. & Rajewsky, K. Generation of memory B cells inside and outside germinal centers. Eur. J. Immunol. 44, 1258–1264 (2014).

    Article  CAS  PubMed  Google Scholar 

  11. Yu, B. et al. Epigenetic landscapes reveal transcription factors that regulate CD8+ T cell differentiation. Nat. Immunol. 18, 573–582 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, K., Wang, M., Zhao, Y. & Wang, W. Taiji: system-level identification of key transcription factors reveals transcriptional waves in mouse embryonic development. Sci. Adv. 5, eaav3262 (2019).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Bories, J. C. et al. Increased T-cell apoptosis and terminal B-cell differentiation induced by inactivation of the Ets-1 proto-oncogene. Nature 377, 635–638 (1995).

    Article  ADS  CAS  PubMed  Google Scholar 

  14. John, S. A., Clements, J. L., Russell, L. M. & Garrett-Sinha, L. A. Ets-1 regulates plasma cell differentiation by interfering with the activity of the transcription factor Blimp-1. J. Biol. Chem. 283, 951–962 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Angelin-Duclos, C., Cattoretti, G., Lin, K. I. & Calame, K. Commitment of B lymphocytes to a plasma cell fate is associated with Blimp-1 expression in vivo. J. Immunol. 165, 5462–5471 (2000).

    Article  CAS  PubMed  Google Scholar 

  16. Fukuda, T. et al. Disruption of the Bcl6 gene results in an impaired germinal center formation. J. Exp. Med. 186, 439–448 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Vasanwala, F. H., Kusam, S., Toney, L. M. & Dent, A. L. Repression of AP-1 function: a mechanism for the regulation of Blimp-1 expression and B lymphocyte differentiation by the B cell lymphoma-6 protooncogene. J. Immunol. 169, 1922–1929 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Tooze, R. M. A replicative self-renewal model for long-lived plasma cells: questioning irreversible cell cycle exit. Front. Immunol. 4, 460 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Scharer, C. D., Barwick, B. G., Guo, M., Bally, A. P. R. & Boss, J. M. Plasma cell differentiation is controlled by multiple cell division-coupled epigenetic programs. Nat. Commun. 9, 1698 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. Dogan, I. et al. Multiple layers of B cell memory with different effector functions. Nat. Immunol. 10, 1292–1299 (2009).

    Article  CAS  PubMed  Google Scholar 

  22. Schenten, D., Egert, A., Pasparakis, M. & Rajewsky, K. M17, a gene specific for germinal center (GC) B cells and a prognostic marker for GC B-cell lymphomas, is dispensable for the GC reaction in mice. Blood 107, 4849–4856 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Weisel, N. M. et al. Surface phenotypes of naive and memory B cells in mouse and human tissues. Nat. Immunol. 23, 135–145 (2022).

    Article  CAS  PubMed  Google Scholar 

  24. Kuraoka, M. et al. Complex antigens drive permissive clonal selection in germinal centers. Immunity 44, 542–552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. de Vinuesa, C. G. et al. Germinal centers without T cells. J. Exp. Med. 191, 485–494 (2000).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Obukhanych, T. V. & Nussenzweig, M. C. T-independent type II immune responses generate memory B cells. J. Exp. Med. 203, 305–310 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Liu, X., Zhao, Y. & Qi, H. T-independent antigen induces humoral memory through germinal centers. J. Exp. Med. 219, e20210527 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cancro, M. P. Age-associated B cells. Annu. Rev. Immunol. 38, 315–340 (2020).

    Article  CAS  PubMed  Google Scholar 

  29. Johnson, J. L. et al. The transcription factor T-bet resolves memory B cell subsets with distinct tissue distributions and antibody specificities in mice and humans. Immunity 52, 842–855 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kim, C. C., Baccarella, A. M., Bayat, A., Pepper, M. & Fontana, M. F. FCRL5+ memory B cells exhibit robust recall responses. Cell Rep. 27, 1446–1460 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li, J. et al. CCR5+T-bet+FoxP3+ effector CD4 T cells drive atherosclerosis. Circ. Res. 118, 1540–1552 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Evrard, M. et al. Sphingosine 1-phosphate receptor 5 (S1PR5) regulates the peripheral retention of tissue-resident lymphocytes. J. Exp. Med. 219, e20210116 (2022).

    Article  CAS  PubMed  Google Scholar 

  33. Reis, B. S., Hoytema van Konijnenburg, D. P., Grivennikov, S. I. & Mucida, D. Transcription factor T-bet regulates intraepithelial lymphocyte functional maturation. Immunity 41, 244–256 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Nickerson, K. M. et al. Age-associated B cells are heterogeneous and dynamic drivers of autoimmunity in mice. J. Exp. Med. 220, e20221346 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kaji, T. et al. Distinct cellular pathways select germline-encoded and somatically mutated antibodies into immunological memory. J. Exp. Med. 209, 2079–2097 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Price, M. J., Scharer, C. D., Kania, A. K., Randall, T. D. & Boss, J. M. Conserved epigenetic programming and enhanced heme metabolism drive memory B cell reactivation. J. Immunol. 206, 1493–1504 (2021).

    Article  CAS  PubMed  Google Scholar 

  37. Lai, A. Y. et al. DNA methylation profiling in human B cells reveals immune regulatory elements and epigenetic plasticity at Alu elements during B-cell activation. Genome Res. 23, 2030–2041 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jadhav, U. et al. Extensive recovery of embryonic enhancer and gene memory stored in hypomethylated enhancer DNA. Mol. Cell 74, 542–554 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Schauder, D. M. et al. E2A-regulated epigenetic landscape promotes memory CD8 T cell differentiation. Proc. Natl Acad. Sci. USA 118, e2013452118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ruckert, T., Lareau, C. A., Mashreghi, M. F., Ludwig, L. S. & Romagnani, C. Clonal expansion and epigenetic inheritance of long-lasting NK cell memory. Nat. Immunol. 23, 1551–1563 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Naik, S. et al. Inflammatory memory sensitizes skin epithelial stem cells to tissue damage. Nature 550, 475–480 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lau, C. M. et al. Epigenetic control of innate and adaptive immune memory. Nat. Immunol. 19, 963–972 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hsu, J. & Sage, J. Novel functions for the transcription factor E2F4 in development and disease. Cell Cycle 15, 3183–3190 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. 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 

  45. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  46. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Law, C. W., Chen, Y., Shi, W. & Smyth, G. K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Danecek, P. et al. Twelve years of SAMtools and BCFtools. Gigascience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank H. Singh for extensive discussions and critical reading of the manuscript; S. Naik (New York University), J. Sun (Sloan Kettering Institute) and C. Lau (Cornell University) for critical reading of the manuscript. We thank T. Marinov, M. Berkey, L. Conter and M. Price for technical support; University of Pittsburgh DLAR for excellent animal husbandry; members of the Shlomchik Lab Memory Group for suggestions and discussion throughout the development of this work. We thank the University of Pittsburgh Innovative Technology Development Core, led by S. Gingras, and the Mouse Embryo Services Core, led by C. Bi, for generating the GCET-TamCre mice. This research was supported in part by the University of Pittsburgh Center for Research Computing, RRID:SCR_022735, through the resources provided. Specifically, this work used the HTC cluster, which is supported by National Institutes of Health (NIH) award number S10OD028483. This research was also supported by NIH grants R01-AI43603 and R01-AI105018 to M.S. D.C. was supported by NIH T32 AI060525 (J. Flynn, principal investigator).

Author information

Authors and Affiliations

Authors

Contributions

D.C. performed most experiments and analyzed all experiments. S.J. performed some GCET-TamCre experiments, and F.W. performed the scRNA-seq experiment. F.W. drove the development of the GCET-TamCre mouse. K.H., under the supervision of S.K., performed Vh186.2 immunoglobulin somatic hypermutation analysis. S.S. completed quality checks and processed bioinformatic data and was advised by M.C. Bioinformatic analyses were performed by D.C. and S.S. The project was conceived by M.S. and D.C., who both designed experiments and interpreted data. M.S. and D.C. wrote the manuscript with input from all the authors.

Corresponding author

Correspondence to Mark Shlomchik.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

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

Additional information

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

Extended data

Extended Data Fig. 1 Distinct transcriptomic and epigenetic profiles of FO NBCs and MBC subsets.

a, PCA plot of genes with expression of >= 0 log2 normalized reads in at least one cell type (n=3 per cell type). b, PCA plot of OCRs with accessibility of >= 1 log2 normalized reads in at least one cell type (n=2 per cell type). c,d, Boxplots showing relative PageRank score for TFs with higher (c) or lower (d) scores in all MBC subsets compared to FO naïve. Boxplots display median values and lower and upper quartiles, the ranges display min and maximum values. n= 2 per cell type.

Extended Data Fig. 2 DAR groups are enriched for distinct TF motifs.

a, Schematic of BALB/c transfer system using VPD-labeled donor cells. Mice were euthanized at days 2.5 and 11.5. b,c Representative flow plots depicting VPD-dilution of listed cell types compared to total transferred B cells in PBS control (unimmunized) at days 2.5 (b) and 11.5 (c). d, PCA plot of genes with expression of >= 0 log2 normalized reads in at least one cell type. e, PCA plot of OCRs with accessibility of >= 1 log2 normalized reads in at least one cell type. f, Ratio of Euclidean distance between DPs and EMPs to Euclidean distance between DNs and EMPs for given log2 FC RNA-seq differential expression thresholds, where Fig. 2c is the reference at abs(log2 FC DP vs DN) >=1. For Euclidean distance, a higher number indicates less similarity; if Euclidean distance ratio is > 1, numerator is less similar than denominator. g, Ratio of Euclidean distance between DNs and LZ GCs to Euclidean distance between DPs and LZ GCs for given log2 FC RNA-seq differential expression thresholds, where Fig. 2c is the reference at abs(log2 FC DP vs DN) >=1. h, Table of top TF motifs enriched in various DAR groups from Fig. 2d compared to background (all called peaks), computed via HOMER. Examples were manually chosen. p = p.value (calculated using cumulative binomial distribution), Motif Score = motif enrichment score above background, Known Motif = motif taken from HOMER database, TF Motif Name = specific TF and/or TF family, along with source if provided, % Coverage in DAR group = % of OCRs in a DAR group that have at least one motif sequence, % Coverage in all OCRs = % of all called OCRs that have at least one motif sequence. i, Heatmap depicting log2 normalized RNA expression of select TFs from Fig. 2f. j, Heatmap depicting all TF motifs with enrichment scores of log2 FC > = 1.4 above background, p. value <= 0.001 for any DAR group. Values shown are motif enrichment scores above background.

Extended Data Fig. 3 The GCET-TamCre is GCBC-specific.

a, Schematic to assess efficiency of GCBC-labeling and non-specific labeling in GCET-TamCre lineage tracing mice. Mice were given 1 mg tamoxifen on days 9, 10, and 11, post-immunization with NP-KLH and euthanized on day 13. b, Representative flow plots depicting YFP expression in NIP+ GCBCs and NIP+ non-GCBCs of corn oil control and tamoxifen-treated mice at day 13. c, Representative flow plots depicting YFP expression in plasmablasts of corn oil control and tamoxifen-treated mice at day 13. d, Representative flow plots depicting YFP expression in non-B cells of corn oil control and tamoxifen-treated mice at day 13. Left two columns depict a total T cell stain, while right 3 columns depict a myeloid lineage stain. e, Schematic to assess efficiency of B cell labeling GCET-TamCre lineage tracing mice. Naïve mice were given 1 mg tamoxifen every day for 3 consecutive days, whereas NP-KLH immunized mice were given 1 mg tamoxifen on days −0.5, 0.5, and 1.5 post-immunization. Mice were euthanized 1 day after final tamoxifen dose or 12 weeks later. f, Representative flow plots depicting YFP expression in naïve mice after 3 tamoxifen doses (top row) and YFP expression in early activated B cells at day 2.5 after 3 tamoxifen doses (bottom row). g, Representative flow plots depicting YFP expression in B cells of naïve mice after 3 tamoxifen doses and chasing out to 12 weeks (top row), YFP expression at week 12 post-immunization in mice not given tamoxifen (middle row), and YFP expression at 12 weeks post-immunization in mice given tamoxifen at days −0.5, 0.5, and 1.5 post-immunization.

Extended Data Fig. 4 Ly6D, PlexinB2, and CD73 can be used to identify GCBC-derived MBCs in WT BALB/c mice.

a, Flow cytometry plots depicting markers expressed by YFP+ MBCs in week 6 KP-KLH-immunized GCET-TamCre+/−R26-LSL-YFP+/− mice (from Fig. 3g; tamoxifen given on days 3–13). b-d, NIP+ Memory cells from directly immunized WT BALB/c mice 8 weeks post-NP-CGG immunization. b, Flow cytometry gating strategy to determine GC-derived MBCs (PlexinB2hiLy6DloCD73+) frequencies within CD80/PD-L2 MBC subsets. c, Flow cytometry gating strategy to determine frequencies of CD80/PD-L2 MBC subsets within total GC-derived MBCs (PlexinB2hiLy6DloCD73+). d, top: percent of GC-derived cells within each CD80/PD-L2 MBC subset; bottom: percent of each CD80/PD-L2 subset within total GC-derived MBCs (n = 5). Bars display mean ± s.d.

Source data

Extended Data Fig. 5 DPEX and DPGC have distinct functions in vivo.

a, Schematic for in vivo transfer and re-activation of 5,000 sorted IgG1 NIP+ MBC subsets. b, Day 4 in vivo data; left: NIP+CD19+CD45.1 cell counts per spleen, counted by flow cytometry (n = 14 for DPEX, n = 10 for DPGC, 4 independent experiments); right: Number of IgG1+ NIP+ spots per 1000 NIP+ cells (derived from left panel, (n = 11 for DPEX, n = 7 for DPGC, 3 independent experiments). c, Day 14 in vivo data; left: number of NIP+CD19+CD138CD45.1CD45.2+CD38CD95+ GCBCs per spleen; right: number of NIP+CD19+CD138CD45.1CD45.2+CD38+ GCBCs per spleen. Both panels counted via flow cytometry (n = 5 for DN, n = 8 for DPEX, n = 6 for DPGC, 3 independent experiments). Bars display mean ± s.d. P-values were calculated using two-tailed Welch’s t-test (*P < = 0.05, **P < 0.01, ***P < 0.001). Actual p-values are listed in source data.

Source data

Extended Data Fig. 6 NP-Ficoll does not produce GCBC-derived MBCs.

a, Number of total MBCs at Day 5 (left) and Day 28 (right) between NP-CGG + PBS (day 5; n = 4, day 28, n = 5), NP-Ficoll + PBS (day 5; n = 6, day 28, n = 4), and NP-Ficoll + GK1.5 (day 5; n = 5, day 28, n = 7) treated mice from Fig. 5. b, Left/middle: Representative flow plots of pre-gated NIP+ B cells from Day 5 post-immunization for NP-KLH + PBS, NP-Ficoll + PBS, NP-KLH + GK1.5, and NP-Ficoll + GK1.5 treated GCET-TamCre+/−R26-LSL-YFP+/− mice. Mice were given one dose of tamoxifen on Day 3; right: Representative flow plots of pre-gated NIP + B cells from Day 28 post-immunization for NP-KLH + PBS, NP-Ficoll + PBS, NP-KLH + GK1.5, and NP-Ficoll + GK1.5 treated GCET-TamCre+/−R26-LSL-YFP+/− mice. Mice were given one dose of tamoxifen every other day from day 3–13. c, Number of YFP+ GCBCs (NIP+CD19+CD138CD38GL7+) per spleen at day 5 (n = 3 per group). d, left: Percent YFP+ of DP MBCs (NIP+CD19+CD138CD38+GL7), middle: Number of total DP MBCs per spleen, and right: Number of YFP+ DPs per spleen at day 28 (n = 3 per group). Bars display mean ± s.d. P-values were calculated using two-tailed Welch’s t-test (*P < = 0.05, **P < 0.01, ***P < 0.001). Actual p-values are listed in source data.

Source data

Extended Data Fig. 7 TI-derived MBCs are not GCBC-derived.

a, Number of total MBCs at Day 5 (left) and Day 28 (right) between NP-CGG + PBS control (day 5; n = 4, day 28, n = 6) and NP-CGG + GK1.5 + FGK (day 5; n = 6, day 28, n = 5) treated mice. b, Representative flow plots of NIP+ MBCs from BALBc transfer system under indicated immunizations at week 4. c, Number of MBC subsets per spleen for each indicated immunization. d, Representative flow plots of NIP+ MBCs from BALB/c transfer system under indicated immunizations at week 4, showing gating to determine GC-derived MBCs. e, left: percent of; right: number of; total MBCs displaying a GC-derived phenotype in the 4 different immunizations. For b-e, n = 3, except NP-CGG + FGK + GK1.5, where n = 4. Bars display mean ± s.d. P-values were calculated using two-tailed Welch’s t-test (*P < = 0.05). Actual p-values are listed in source data.

Source data

Extended Data Fig. 8 A shared CD80SP epigenetic signature can be found in TD and TI systems.

a, Examples of OCRs at Cd80 and Pdcd1lg2 that display a consistent pattern of accessibility in CD80SP MBCs within each system (NP-CGG + PBS, NP-Ficoll + GK1.5, NP-CGG + FGK + GK1.5, MBCs from day 28. n = 2 per cell types). b, GSEA plots using DARs between CD80SP MBCs and DN MBCs as genesets, showing all possible comparisons across all different systems. c, GSEA plots using DARs between CD80SP MBCs and DP MBCs as genesets, showing all possible comparisons across all different systems. Cutoff of log2FC >= 1, p-value <= 0.05 (calculated via two-tailed permutations), accessibility >= 2 log2 normalized counts was used for all genesets; the top 300 DARs, ordered by log2 FC, were used for the geneset unless total DARs were fewer than 300. Number of total DARs (in parentheses): NP-CGG; CD80SP > DN (884), CD80SP < DN (383), DP > CD80SP (93), DP < CD80SP (652). NP-Ficoll + GK1.5; CD80SP > DN (395), CD80SP < DN (83), DP > CD80SP (184), DP < CD80SP (37). NP-CGG + FGK + GK1.5; CD80SP > DN (1,332), CD80SP < DN (503), DP > CD80SP (499), DP < CD80SP (665).

Extended Data Fig. 9 TD-derived CD80SPs exhibit an inflammatory signature.

a, Bar graphs depicting OCRs that differ between DN MBCs and each other MBC subset. Top plot depicts OCRs lower in DNs than each MBC subset (x-axis); bottom plot depicts OCRs higher in DNs. Differential accessibility cutoff for each MBC subset compared to DNs is log2FC >= 1, FDR < = 0.05, and log2 accessibility >=1 normalized counts. b, Venn diagrams plotting the intersection of DARs that were more accessible in any MBC subset compared to DNs on the top and less accessible compared to DNs on the bottom, as enumerated in a. c, Bar graphs depicting OCRs that differ between DP MBCs and each other MBC subset. Top plot depicts OCRs lower in DPs than each MBC subset (x-axis); bottom plot depicts OCRs higher in DPs. Differential accessibility cutoff for each MBC subset compared to DPs is log2FC >= 1, FDR < = 0.05, and log2 accessibility >=1 normalized counts. d, Venn diagrams plotting the intersection of DARs that were more accessible in any MBC subset compared to DPs on the top and less accessible compared to DPs on the bottom, as enumerated in c. e, Select DARs near S1pr5 and Tbx21 showing specificity to CD80SP MBCs from the NP-CGG system. f, RNA expression heatmap (row z-scored) of select Tbet/ABC-associated genes among different systems and cell types (n = 3 for NP-CGG, n = 2 for other systems). g, PageRank score (z-scored) of Tbx21 from NP-CGG derived cell types.

Supplementary information

Supplementary Information

Supplementary Figs. 1–7, protocols and references

Reporting Summary

Peer Review File

Supplementary Data

Statistical source data for Supplementary Fig. 3.

Supplementary Table 1

Data for Fig. 1c–f and Extended Data Fig. 1a.

Supplementary Table 2

Data for Fig. 1g–j and Extended Data Fig. 1b.

Supplementary Table 3

Data for Fig. 1k and Extended Data Fig. 1c,d.

Supplementary Table 4

Data for Fig. 2c, Extended Data Fig. 2d,f,g and Supplementary Fig. 2c.

Supplementary Table 5

Data for Fig. 2d, Extended Data Fig. 2e and Supplementary Fig. 2d.

Supplementary Table 6

Data for Fig. 2f and Extended Data Fig. h–j.

Supplementary Table 7

Data for Fig. 4a–f.

Supplementary Table 8

Data for Fig. 8.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

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

Callahan, D., Smita, S., Joachim, S. et al. Memory B cell subsets have divergent developmental origins that are coupled to distinct imprinted epigenetic states. Nat Immunol 25, 562–575 (2024). https://doi.org/10.1038/s41590-023-01721-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01721-9

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