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.

  • Resource
  • Published:

Surface phenotypes of naive and memory B cells in mouse and human tissues

Abstract

Memory B cells (MBCs) protect the body from recurring infections. MBCs differ from their naive counterparts (NBCs) in many ways, but functional and surface marker differences are poorly characterized. In addition, although mice are the prevalent model for human immunology, information is limited concerning the nature of homology in B cell compartments. To address this, we undertook an unbiased, large-scale screening of both human and mouse MBCs for their differential expression of surface markers. By correlating the expression of such markers with extensive panels of known markers in high-dimensional flow cytometry, we comprehensively identified numerous surface proteins that are differentially expressed between MBCs and NBCs. The combination of these markers allows for the identification of MBCs in humans and mice and provides insight into their functional differences. These results will greatly enhance understanding of humoral immunity and can be used to improve immune monitoring.

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: Discovery of surface markers differentially regulated between MBCs and NBCs by mouse LEGENDScreen analysis.
Fig. 2: Discovery of differentially regulated surface markers between MBCs and NBCs by human LEGENDScreen analysis.
Fig. 3: Venn diagram of highly expressed surface markers on human and mouse NBCs and MBCs.
Fig. 4: Summary of marker expression revealed by human and mouse LEGENDScreen analysis.
Fig. 5: Combinations of surface markers that allow for the identification of mouse MBCs.
Fig. 6: Surface markers differentially regulated on NBCs and MBCs across human tissues and donors.
Fig. 7: Combinations of surface markers that allow for the identification of human MBCs across tissues.
Fig. 8: Pathway enrichment analysis of markers differentially regulated between MBCs and NBCs, as revealed by LEGENDScreen analysis.

Similar content being viewed by others

Data availability

All data are available upon request. Flow cytometry files of LEGENDScreen assays were deposited and are publicly available at http://flowrepository.org with the accession numbers FR-FCM-Z4LQ (mouse LEGENDScreen) and FR-FCM-Z4LS (human LEGENDScreen). Source data are provided with this paper.

References

  1. Gry, M. et al. Correlations between RNA and protein expression profiles in 23 human cell lines. BMC Genomics 10, 365 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Chan, J. K., Ng, C. S. & Hui, P. K. A simple guide to the terminology and application of leucocyte monoclonal antibodies. Histopathology 12, 461–480 (1988).

    Article  CAS  PubMed  Google Scholar 

  3. Weisel, F. & Shlomchik, M. Memory B cells of mice and humans. Annu Rev. Immunol. 35, 255–284 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Akkaya, M., Kwak, K. & Pierce, S. K. B cell memory: building two walls of protection against pathogens. Nat. Rev. Immunol. 20, 229–238 (2020).

    Article  CAS  PubMed  Google Scholar 

  5. Cyster, J. G. & Allen, C. D. C. B cell responses: cell interaction dynamics and decisions. Cell 177, 524–540 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Wong, R. & Bhattacharya, D. Basics of memory B-cell responses: lessons from and for the real world. Immunology 156, 120–129 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Nicholas, M. W. et al. A novel subset of memory B cells is enriched in autoreactivity and correlates with adverse outcomes in SLE. Clin. Immunol. 126, 189–201 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Sweet, R. A., Cullen, J. L. & Shlomchik, M. J. Rheumatoid factor B cell memory leads to rapid, switched antibody-forming cell responses. J. Immunol. 190, 1974–1981 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Jacobi, A. M. et al. Activated memory B cell subsets correlate with disease activity in systemic lupus erythematosus: delineation by expression of CD27, IgD, and CD95. Arthritis Rheum. 58, 1762–1773 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Pugh-Bernard, A. E. et al. Regulation of inherently autoreactive VH4-34 B cells in the maintenance of human B cell tolerance. J. Clin. Invest. 108, 1061–1070 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wong, R. et al. Affinity-restricted memory B cells dominate recall responses to heterologous Flaviviruses. Immunity 53, 1078–1094 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Ellebedy, A. H. et al. Defining antigen-specific plasmablast and memory B cell subsets in human blood after viral infection or vaccination. Nat. Immunol. 17, 1226–1234 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tiller, T. et al. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J. Immunol. Methods 329, 112–124 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Weisel, N. M. et al. Comprehensive analyses of B cell compartments across the human body reveal novel subsets and a gut resident memory phenotype. Blood 136, 2774–2785 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mahanonda, R. et al. Human memory B cells in healthy gingiva, gingivitis, and periodontitis. J. Immunol. 197, 715–725 (2016).

    Article  CAS  PubMed  Google Scholar 

  16. Tangye, S. G., Liu, Y. J., Aversa, G., Phillips, J. H. & de Vries, J. E. Identification of functional human splenic memory B cells by expression of CD148 and CD27. J. Exp. Med. 188, 1691–1703 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhao, Y. et al. Spatiotemporal segregation of human marginal zone and memory B cell populations in lymphoid tissue. Nat. Commun. 9, 3857 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Nair, N. et al. High-dimensional immune profiling of total and rotavirus VP6-specific intestinal and circulating B cells by mass cytometry. Mucosal Immunol. 9, 68–82 (2016).

    Article  CAS  PubMed  Google Scholar 

  19. Klein, U., Kuppers, R. & Rajewsky, K. Evidence for a large compartment of IgM-expressing memory B cells in humans. Blood 89, 1288–1298 (1997).

    Article  CAS  PubMed  Google Scholar 

  20. Wu, Y. C., Kipling, D. & Dunn-Walters, D. K. The relationship between CD27 negative and positive B cell populations in human peripheral blood. Front Immunol. 2, 81 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Wei, C. et al. A new population of cells lacking expression of CD27 represents a notable component of the B cell memory compartment in systemic lupus erythematosus. J. Immunol. 178, 6624–6633 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Fecteau, J. F., Cote, G. & Neron, S. A new memory CD27IgG+ B cell population in peripheral blood expressing VH genes with low frequency of somatic mutation. J. Immunol. 177, 3728–3736 (2006).

    Article  CAS  PubMed  Google Scholar 

  23. Grimsholm, O. et al. The interplay between CD27dull and CD27bright B cells ensures the flexibility, stability, and resilience of human B cell memory. Cell Rep. 30, 2963–2977 (2020).

    Article  CAS  PubMed  Google Scholar 

  24. 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 

  25. 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 

  26. Magri, G. et al. Human secretory IgM emerges from plasma cells clonally related to gut memory B cells and targets highly diverse commensals. Immunity 47, 118–134 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. 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 

  29. Schenkel, JasonM. & Masopust, D. Tissue-resident memory T cells. Immunity 41, 886–897 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Shiow, L. R. et al. CD69 acts downstream of interferon-alpha/beta to inhibit S1P1 and lymphocyte egress from lymphoid organs. Nature 440, 540–544 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Palm, A. E. & Henry, C. Remembrance of things past: long-term B cell memory after infection and vaccination. Front. Immunol. 10, 1787 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Llinàs, L. et al. Expression profiles of novel cell surface molecules on B-cell subsets and plasma cells as analyzed by flow cytometry. Immunol. Lett. 134, 113–121 (2011).

    Article  PubMed  Google Scholar 

  33. Sanz, I., Wei, C., Lee, F. E. & Anolik, J. Phenotypic and functional heterogeneity of human memory B cells. Semin. Immunol. 20, 67–82 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Glass, D. R. et al. An integrated multi-omic single-cell atlas of human B cell identity. Immunity 53, 217–232 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Good, K. L., Avery, D. T. & Tangye, S. G. Resting human memory B cells are intrinsically programmed for enhanced survival and responsiveness to diverse stimuli compared to naive B cells. J. Immunol. 182, 890–901 (2009).

    Article  CAS  PubMed  Google Scholar 

  36. Weisel, F. J. et al. Unique requirements for reactivation of virus-specific memory B lymphocytes. J. Immunol. 185, 4011–4021 (2010).

    Article  CAS  PubMed  Google Scholar 

  37. Good, K. L. & Tangye, S. G. Decreased expression of Kruppel-like factors in memory B cells induces the rapid response typical of secondary antibody responses. Proc. Natl Acad. Sci. USA 104, 13420–13425 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bhattacharya, D. et al. Transcriptional profiling of antigen-dependent murine B cell differentiation and memory formation. J. Immunol. 179, 6808–6819 (2007).

    Article  CAS  PubMed  Google Scholar 

  39. Tomayko, M. M. et al. Systematic comparison of gene expression between murine memory and naive B cells demonstrates that memory B cells have unique signaling capabilities. J. Immunol. 181, 27–38 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Tangye, S. G. & Tarlinton, D. M. Memory B cells: effectors of long-lived immune responses. Eur. J. Immunol. 39, 2065–2075 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Oh, J. E. et al. Migrant memory B cells secrete luminal antibody in the vagina. Nature 571, 122–126 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Joo, H. M., He, Y. & Sangster, M. Y. Broad dispersion and lung localization of virus-specific memory B cells induced by influenza pneumonia. Proc. Natl Acad. Sci. USA 105, 3485–3490 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ehrhardt, G. R. et al. Expression of the immunoregulatory molecule FcRH4 defines a distinctive tissue-based population of memory B cells. J. Exp. Med. 202, 783–791 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Allie, S. R. et al. The establishment of resident memory B cells in the lung requires local antigen encounter. Nat. Immunol. 20, 97–108 (2019).

    Article  CAS  PubMed  Google Scholar 

  45. Onodera, T. et al. Memory B cells in the lung participate in protective humoral immune responses to pulmonary influenza virus reinfection. Proc. Natl Acad. Sci. USA 109, 2485–2490 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Koethe, S. et al. Pivotal advance: CD45RB glycosylation is specifically regulated during human peripheral B cell differentiation. J. Leukoc. Biol. 90, 5–19 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Conter, L. J., Song, E., Shlomchik, M. J. & Tomayko, M. M. CD73 expression is dynamically regulated in the germinal center and bone marrow plasma cells are diminished in its absence. PLoS ONE 9, e92009 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Lycke, N., Bemark, M., Komban, R. & Stensson, A. Intricate properties of memory B cell development after oral immunization (MUC4P.836). J. Immunol. 192, 133.112 (2014).

    Article  Google Scholar 

  49. Krishnamurty, A. T. et al. Somatically hypermutated plasmodium-specific IgM(+) memory B Cells are rapid, plastic, early responders upon malaria rechallenge. Immunity 45, 402–414 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Bemark, M. et al. Limited clonal relatedness between gut IgA plasma cells and memory B cells after oral immunization. Nat. Commun. 7, 12698 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Yates, J. L., Racine, R., McBride, K. M. & Winslow, G. M. T cell-dependent IgM memory B cells generated during bacterial infection are required for IgG responses to antigen challenge. J. Immunol. 191, 1240–1249 (2013).

    Article  CAS  PubMed  Google Scholar 

  52. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gosselin, D. et al. Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 160, 351–352 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Barnden, M. J., Allison, J., Heath, W. R. & Carbone, F. R. Defective TCR expression in transgenic mice constructed using cDNA-based alpha- and beta-chain genes under the control of heterologous regulatory elements. Immunol. Cell Biol. 76, 34–40 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Thome, J. J. C. et al. Spatial map of human T cell compartmentalization and maintenance over decades of life. Cell 159, 814–828 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Carpenter, D. J. et al. Human immunology studies using organ donors: Impact of clinical variations on immune parameters in tissues and circulation. Am. J. Transpl. 18, 74–88 (2018).

    Article  CAS  Google Scholar 

  59. Weisel, F. J. et al. Germinal center B cells selectively oxidize fatty acids for energy while conducting minimal glycolysis. Nat. Immunol. 21, 331–342 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We thank D. Carpenter for organ acquisition, the transplant coordinators at LiveOnNY for tissues from organ donors and members of the laboratory of D. Farber at Columbia University for providing access to human organ donor tissue samples. We thank M. Berkey for supporting experimental procedures. We thank BioLegend, in particular A. Cornett and N. Lucas, for providing reagents. This work was funded by National Institutes of Health, National Institute of Allergy and Infectious Diseases (NIH/NIAID) grants R01 AI043603 (M.J.S.) and P01 AI106697 (M.J.S. and D.L.F.).

Author information

Authors and Affiliations

Authors

Contributions

N.M.W. and S.M.J. contributed equally to this work. M.J.S. and F.J.W. designed research and are equal senior authors. F.J.W., N.M.W., S.M.J. and L.J.C. performed experiments and analyzed data. D.L.F. and R.A.E. gave conceptual advice. S.S., D.J.C. and M.M.C. performed computational analysis. F.J.W. and M.J.S. wrote the manuscript.

Corresponding author

Correspondence to Mark J. Shlomchik.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Surface markers differentially expressed on human and mouse NBCs and MBCs, in support of Fig. 3.

Differentially regulated surface markers as in Fig. 3 are sorted based on the ratio of the MFI of MBCs to NBCs. a, MFI ratio of MBCs to NBCs in humans (mean of three donors (D182, D185 and D186)). b, MFI ratio MBCs to NBCs in mice. Red and blue dots depict higher expression on MBCs or NBCs, respectively. Plot was built using ggplot2 (version 3.2.1) in R (version 3.6.1).

Extended Data Fig. 2 Validation of surface markers differentially regulated on mouse NBCs and MBCs, in support of Figs. 3 and 4.

Histograms of flow cytometric expression of 24 depicted surface markers on MBCs (red) and NBCs (blue). MBCs were identified as CD45.2+NIP+CD19+ live singlets on splenocytes of transfer recipients (seven females 39 weeks old 31 weeks after immunization plus three males 34 weeks old 23 weeks after immunization) and splenic B cells of CD45.1 naive B1-8i+/− BALB/cJ mice (three males 11 weeks old plus two males, 9 weeks old) were mixed into the staining to serve as NBC, identified by their CD45 allotype mark. Cells were stained with Murine Stain 3 (Supplementary Tables 5 and 6). The FMO control for the PE channel is shown in the bottom right histogram.

Extended Data Fig. 3 Validation of surface markers differentially regulated on human NBCs and MBCs, in support of Figs. 3 and 4.

Cryopreserved human splenocytes were stained with Stain 2, Stain 3, Stain 4 or Stain 5 (Supplementary Tables 3 and 4) for flow cytometric analysis. Shown are histograms of the expression of 16 depicted surface markers on MBCs (CD19+CD27+, red) and NBCs (CD19+CD27, blue). All markers were validated on three individual donor spleens listed in the tables below the histograms.

Extended Data Fig. 4 Combinations of surface markers that allow for the identification of mouse MBCs and NBCs in the C57BL/6 background, in support of Fig. 5.

Direct immunization (left): single-cell suspensions of indicated tissues from three male C57BL/6 WT (CD45.2, 12 weeks old, 4 weeks after immunization) mice were analyzed at day 28 after i.p. immunization with 100 µg NP-KLH. 5 × 106 cells were mixed with 1 × 106 cells of corresponding tissues of one naive male C57BL/6 CD45.1 congenic mouse (8 weeks old) to allow for the simultaneous identification of MBCs and their comparable naive counterparts in a single staining tube. MBCs and NBCs were identified as described in Supplementary Fig. 8a, and displayed data are concatenated of three individual samples. OT-II adoptive transfer system (right): single-cell suspensions of indicated tissues from five individual OT-II adoptive transfer recipients (males, 12 weeks old, 4 weeks after immunization) were analyzed at day 28 after i.p. immunization with 50 µg NP-CGG. 5 × 106 cells were mixed with 1 × 106 cells of corresponding tissues of one male naive C57BL/6 WT (CD45.2) mouse (7 weeks old) to allow for the simultaneous identification of MBCs and their comparable naive counterparts in a single staining tube. Cells were stained with Murine Stain 5 (Supplementary Tables 5 and 6). MBCs (red) and NBCs (blue) were identified as described in Supplementary Fig. 8b, and displayed data are concatenated of five individual samples. Shown are contour plots of pairwise combinations of CD205/CD274, CD81/CD11a and CD267/CD180 as in Fig. 5, which can be used to distinguish MBCs and NBCs across tissues.

Extended Data Fig. 5 Surface markers differentially regulated on NBCs versus MBCs across human tissues, in support of Fig. 6.

Single-cell suspensions of spleen (SP), blood (B), BM, LN, intestinal tissue (gut) and tonsil (T) were stained for flow-cytometric analysis using Stain 2 and Stain 4 (Supplementary Tables 3 and 4). The left panel shows the summary of the differences in MFI between CD19+CD27+ B cells and CD19+CD27 B cells (ΔMFI) for the depicted surface markers. For CD74 and CD119, spleen (n = 23 in red), blood (n = 7 in blue), BM (n = 9 in magenta), LN (n = 6 in green), gut (n = 3 in brown) and tonsil (n = 2 in black) samples were analyzed. For CD218a, spleen (n = 21 in red), blood (n = 10 in blue), BM (n = 10 in magenta), LN (n = 8 in green), gut (n = 8 in brown) and tonsil (n = 2 in black) samples were analyzed. For CD370, spleen (n = 21 in red), blood (n = 10 in blue), BM (n = 10 in magenta), LN (n = 8 in green), gut (n = 8 in brown) and tonsil (n = 2 in black) samples were analyzed. Bars are mean and error bars are ± standard deviation. The right panel shows example histograms for depicted surface markers of CD19+CD27 B cells (blue) and CD19+CD27+ B cells (red) across tissues of D260. Stars indicate significant differences in ΔMFI of indicated tissues compared to spleen using the unpaired two-tailed t test with Welch’s correction. ***P < 0.001, ****P < 0.0001. Exact significant P values for comparison between spleen and the indicated tissue for each marker are for CD74: all tissues, P < 0.0001; for CD119, blood, BM and gut,P < 0.0001, LN P = 0.0006 and tonsil P = 0.1112; for CD218a, blood and BM P < 0.0001, gut P = 0.0001 and tonsil P = 0.1047; and for CD370, blood, LN, gut and tonsil P = 0.0002 and BM P = 0.0003.

Source data

Extended Data Fig. 6 Expression of surface markers CD11a and CD200 on human splenic MBCs and NBCs separated by Ig isotype, in support of Fig. 7.

Splenic single-cell suspensions were stained for depicted markers (Stain 5, Supplementary Tables 3 and 4). Overlayed histograms for expression of CD11a (top) or CD200 (bottom) of either total CD27 and CD27+ (first row), or specific Ig isotypes for CD27 overlayed with total CD27+ (row 2, CD27 IgM/D; row 3, CD27 IgG; and row 4, CD27 IgA) are shown for six individual donors (D192, D215, D228, D333, D365 and D388). CD19+CD27+ MBCs are in red, and CD19+CD27 NBCs are in blue. The last rows of each panel show a summary of the CD27 isotypes analyzed (IgM/D, green; IgG, blue; and IgA, orange) in direct comparison with the total CD27 B cells (red).

Supplementary information

Supplementary Information

Supplementary Figures 1–10 and Supplementary Tables 1–6.

Reporting Summary

Source data

Source Data Fig. 6

Delta MFI for different tissues for depicted markers..

Source Data Extended Data Fig. 5

Delta MFI for different tissues for depicted markers

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Weisel, N.M., Joachim, S.M., Smita, S. et al. Surface phenotypes of naive and memory B cells in mouse and human tissues. Nat Immunol 23, 135–145 (2022). https://doi.org/10.1038/s41590-021-01078-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01078-x

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research