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:

The establishment of resident memory B cells in the lung requires local antigen encounter

Abstract

Memory B cells are found in lymphoid and non-lymphoid tissues, suggesting that some may be tissue-resident cells. Here we show that pulmonary influenza infection elicited lung-resident memory B cells (BRM cells) that were phenotypically and functionally distinct from their systemic counterparts. BRM cells were established in the lung early after infection, in part because their placement required local antigen encounter. Lung BRM cells, but not systemic memory B cells, contributed to early plasmablast responses following challenge infection. Following secondary infection, antigen-specific BRM cells differentiated in situ, whereas antigen-non-specific BRM cells were maintained as memory cells. These data demonstrate that BRM cells are an important component of immunity to respiratory viruses such as influenza virus and suggest that vaccines designed to elicit BRM cells must deliver antigen to the lungs.

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: Identification of influenza-specific B cells.
Fig. 2: Phenotype of memory B cells in the lung and lymphoid organs.
Fig. 3: Identification of influenza-specific, non-circulating BRM cells in the lung.
Fig. 4: BRM cells in the lung are established early after infection.
Fig. 5: BRM cells in the lung are generated from early CD40-dependent precursors.
Fig. 6: Establishment of BRM cells in the lung requires local antigen encounter.
Fig. 7: BRM cells are associated with protection from secondary infection.
Fig. 8: BRM cells are required for rapid secondary ASCs in the lung.

Similar content being viewed by others

Data availability

The data supporting the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Halliley, J. L. et al. Long-lived plasma cells are contained within the CD19CD38hiCD138+ subset in human bone marrow. Immunity 43, 132–145 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Dorner, T. & Radbruch, A. Antibodies and B cell memory in viral immunity. Immunity 27, 384–392 (2007).

    Article  PubMed  Google Scholar 

  3. Phan, T. G. & Tangye, S. G. Memory B cells: total recall. Curr. Opin. Immunol. 45, 132–140 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Bannard, O. & Cyster, J. G. Germinal centers: programmed for affinity maturation and antibody diversification. Curr. Opin. Immunol. 45, 21–30 (2017).

    Article  CAS  PubMed  Google Scholar 

  5. Shlomchik, M. J. & Weisel, F. Germinal center selection and the development of memory B and plasma cells. Immunol. Rev. 247, 52–63 (2012).

    Article  PubMed  Google Scholar 

  6. Schittek, B. & Rajewsky, K. Maintenance of B-cell memory by long-lived cells generated from proliferating precursors. Nature 346, 749–751 (1990).

    Article  CAS  PubMed  Google Scholar 

  7. 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 

  8. Jaimes, M. C. et al. Maturation and trafficking markers on rotavirus-specific B cells during acute infection and convalescence in children. J. Virol. 78, 10967–10976 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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 

  10. Joo, H. M., He, Y., Sundararajan, A., Huan, L. & Sangster, M. Y. Quantitative analysis of influenza virus-specific B cell memory generated by different routes of inactivated virus vaccination. Vaccine 28, 2186–2194 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. 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 

  13. Pape, K. A., Taylor, J. J., Maul, R. W., Gearhart, P. J. & Jenkins, M. K. Different B cell populations mediate early and late memory during an endogenous immune response. Science 331, 1203–1207 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Palladino, G., Mozdzanowska, K., Washko, G. & Gerhard, W. Virus-neutralizing antibodies of immunoglobulin G (IgG) but not of IgM or IgA isotypes can cure influenza virus pneumonia in SCID mice. J. Virol. 69, 2075–2081 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. 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 

  16. Bernasconi, N. L., Traggiai, E. & Lanzavecchia, A. Maintenance of serological memory by polyclonal activation of human memory B cells. Science 298, 2199–2202 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, F. E. et al. Circulating human antibody-secreting cells during vaccinations and respiratory viral infections are characterized by high specificity and lack of bystander effect. J. Immunol. 186, 5514–5521 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Adachi, Y. et al. Distinct germinal center selection at local sites shapes memory B cell response to viral escape. J. Exp. Med. 212, 1709–1723 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Moyron-Quiroz, J. E. et al. Role of inducible bronchus associated lymphoid tissue (iBALT) in respiratory immunity. Nat. Med. 10, 927–934 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Hwang, J. Y., Randall, T. D. & Silva-Sanchez, A. Inducible bronchus-associated lymphoid tissue: taming inflammation in the lung. Front. Immunol. 7, 258 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Zaid, A. et al. Persistence of skin-resident memory T cells within an epidermal niche. Proc. Natl. Acad. Sci. USA 111, 5307–5312 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. McMaster, S. R. et al. Pulmonary antigen encounter regulates the establishment of tissue-resident CD8 memory T cells in the lung airways and parenchyma. Mucosal Immunol. 11, 1071–1078 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Moyron-Quiroz, J. E. et al. Persistence and responsiveness of immunologic memory in the absence of secondary lymphoid organs. Immunity 25, 643–654 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Vu Van, D. et al. Local T/B cooperation in inflamed tissues is supported by T follicular helper-like cells. Nat. Commun. 7, 10875 (2016).

    Article  Google Scholar 

  26. Rao, D. A. et al. Pathologically expanded peripheral T helper cell subset drives B cells in rheumatoid arthritis. Nature 542, 110–114 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Keating, R. et al. The kinase mTOR modulates the antibody response to provide cross-protective immunity to lethal infection with influenza virus. Nat. Immunol. 14, 1266–1276 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Matloubian, M. et al. Lymphocyte egress from thymus and peripheral lymphoid organs is dependent on S1P receptor 1. Nature 427, 355–360 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Misulovin, Z., Yang, X. W., Yu, W., Heintz, N. & Meffre, E. A rapid method for targeted modification and screening of recombinant bacterial artificial chromosome. J. Immunol. Methods 257, 99–105 (2001).

    Article  CAS  PubMed  Google Scholar 

  30. Beckett, D., Kovaleva, E. & Schatz, P. J. A minimal peptide substrate in biotin holoenzyme synthetase-catalyzed biotinylation. Protein Sci. 8, 921–929 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors would like to thank U. Mudunuru and S. Simpler for animal husbandry and the Rheumatic Diseases Core Center flow cytometry facility, which is supported by AI078907. This work was supported by NIH grants HL69409, AI100127, AI097357, AI109962 to T.D.R. and AI120508 to S.R.A.

Author information

Authors and Affiliations

Authors

Contributions

S.R.A., F.E.L. and T.D.R. designed the experiments. J.E.B., B.A.G. and T.D.R. designed the recombinant influenza proteins and J.E.B. and B.A.G. expressed, purified and characterized the tetramers. U.M. and S.R.A performed the surgeries. M.D.S. performed the intratracheal infections. S.R.A performed and analyzed the experiments and generated the figures. S.R.A. and T.D.R. wrote the manuscript. All authors edited the manuscript.

Corresponding author

Correspondence to Troy D. Randall.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Integrated supplementary information

Supplementary Fig. 1 Gating strategies.

Gates are shown sequentially from left to right. Coloured gates are used in the figures listed to the right in font of the same colour.

Supplementary Fig. 2 Identification of influenza-specific memory B cells.

(af) Cells from the lungs (a,d), mLNs (b,e) and spleens (c,f) of naïve mice or PR8-infected mice were gated on live, singlet lymphocytes and then on CD19+CD38hiIgDIgM+ (IgM memory) or CD19+CD38hiIgDIgM (ISW memory) (Supplementary Fig. 1c) and analysed for NP-specific or HA-specific memory B cells on day 30 (ac) or day 70 (df) after infection. Data are representative of four experiments with five mice/timepoint.

Supplementary Fig. 3 Phenotypic characteristics of non-circulating and total memory B cells.

(a,b) Mice were infected with PR8 influenza A virus, and cells from the mLN, lung and spleen were gated on live singlet lymphocytes and then on NP-specific CD19+CD38+IgMIgD ISW memory B cells that were either B220+/– (combined circulating and non-circulating; a) or B220 (non-circulating; b) (gating strategy as in Fig. 2a–d). The phenotype within these populations was determined as in Fig. 2e–i. Data are representative of five experiments with five mice each. Graphs show individual data points (n = 5) as well as mean ± SD. Data were analysed by one-way ANOVA with Tukey’s correction for multiple comparisons: (a) CD73+PD-L2+ ***p = 0.0002, ****p = 0.0001; (a) CD80PD-L2+ ****p = 0.0001, ####p = 0.0001; (a) CD69+CD103 ns = 0.4447, *p = 0.0129; (a) CD62L+CD69+ ns = 0.0511, *p = 0.0237; (a) CXCR3+ *p = 0.0350, **p = 0.0054; (b) CD73+PD-L2+ ****p = 0.0001, ####p = 0.0001; (b) CD80PD-L2+ ****p = 0.0001, ####p = 0.0001; (b) CD69+CD103 ***p = 0.0002, *p = 0.0105; (b) CD62L+CD69+ *p = 0.0471, **p = 0.0042; (b) CXCR3+ *p = 0.0206, ***p = 0.0008; p < 0.05 is considered significant. (ce) Cells from the mLN (c), spleen (d) and lung (e) of day 44 PR8-infected mice were gated on live, singlet lymphocytes (Supplementary Fig. 1a) and then on NP-specific CD19+CD38+IgD memory B cells, and the frequency of IgM-, IgA-, IgG1-, IgG2b-, IgG2c- and IgG3-expressing cells was determined. Data are representative of three experiments with five mice.

Supplementary Fig. 4 Identification of influenza-specific, non-circulating and total memory B cells.

(al) Mice were infected on day 0, surgically paired with partner mice on day 44 and analysed on day 59. Cells from the lung (ad), mLN (eh) or spleen (il) were gated on live, singlet lymphocytes (Supplementary Fig. 1a) and subsequently gated on CD19+CD38+IgDIgM+ (IgM) or CD19+CD38+IgDIgM (ISW) NP-specific memory B cells in either the B220 (non-circulating; a,c,e,g,i,k) or B220+/– (combined circulating and non-circulating; b,d,f,h,j,l) fractions. The numbers of NP-specific memory B cells derived from host and partner mice in the lung (c,d), mLN (g,h) and spleen (i,j) are shown for both the non-circulating fraction and the total (combined circulating and non-circulating) fraction. Data are representative of three experiments combined, totalling 11 pairs of mice. Graphs show individual data as well as mean ± SD. Significance was determined using one-way ANOVA followed by the Bonferroni–Sidak method for multiple comparisons: (c) Lung, IgM HA B220 **p = 0.0031, *p = 0.0243; (c) Lung, IgM NP B220 ****p = 0.0001, **p = 0.0040; (c) Lung, ISW HA B220 ***p = 0.0003, ###p = 0.0002; (c) Lung, ISW NP B220 ****p = 0.0001, **p = 0.0022; (d) Lung, IgM HA B220+/– **p = 0.0013, *p = 0.0158; (d) Lung, IgM NP B220+/– ****p = 0.0001, **p = 0.0017; (d) Lung, ISW HA B220+/– ****p = 0.0001, ***p = 0.0001; (d) Lung, ISW NP B220+/– ****P = 0.0001, **p = 0.0024; (g) mLN, IgM HA B220 p = 0.0618, p = 0.1136; (g) mLN, IgM NP B220 p = 0.0783, *p = 0.0183; (g) mLN, ISW HA B220 **p = 0.0014, ****p = 0.0001; (g) mLN, ISW NP B220 **p = 0.0079, ****p = 0.0001; (h) mLN, IgM HA B220+/– p = 0.0718, p = 0.1631; (h) mLN, IgM NP B220+/– p = 0.0746, *p = 0.0237; (h) mLN, ISW HA B220+/– **p = 0.0022, ****p = 0.0001; (h) mLN, ISW NP B220+/– **p = 0.0081, ***p = 0.0001; (k) Spleen, IgM HA B220 p = 0.2821, **p = 0.0039; (k) Spleen, IgM NP B220 *p = 0.0160, **p = 0.0012; (k) Spleen, ISW HA B220 p = 0.6749, p = 0.2475; (k) Spleen, ISW NP B220 *p = 0.0337, **p = 0.0028; (l) Spleen, IgM HA B220+/– p = 0.0662, *p = 0.0116; (l) Spleen, IgM NP B220+/– **p = 0.0078, *p = 0.0156; (l) Spleen, ISW HA B220+/– p = 0.4171, p = 0.1469; (l) Spleen, ISW NP B220+/– **p = 0.0029, *p = 0.0108; p < 0.05 is considered significant.

Supplementary Fig. 5 HA-specific BRM cells in the lung are generated from early CD40-dependent precursors.

(ad) Mice were infected with PR8 and administered anti-CD40L (MR1) or isotype control (CT) antibody every other day for 10 days starting on day 5 (a), day 10 (b), day 20 (c) or day 30 (d). Cells from the lung were gated on live, singlet lymphocyte, B220CD19+CD38+IgM+IgD IgM BRM cells or B220CD19+CD38+IgMIgD ISW BRM cells (Supplementary Fig. 1g). Data are representative of three experiments with five mice/group per timepoint. Graphs show mean ± SD as well as individual data points. Significance was determined using an unpaired, two-tailed t-test: ***p = 0.0009, ###p = 0.0006 (a), **p = 0.0090 (b), *p = 0.0219 (c). p < 0.05 is considered significant.

Supplementary Fig. 6 Poor recruitment of antigen-non-specific IgM memory cells to inflamed lungs.

(ag), Mice were peritoneally infected with PR8 on day 0, intranasally challenged with X31 on day 30 and analysed on day 40 (ad) and day 75 (eg). Cells from the lung were gated on live, singlet lymphocytes (Supplementary Fig. 1a) and subsequently gated on CD19+B220CD38+IgDIgM+ IgM memory B cells (a). NP-specific (b,e), HA(PR8)-specific (c,f) and HA(X31)-specific (d,g) IgM BRM cells were enumerated on day 40 (bd) and day 75 (eg). Graphs show individual data points as well as mean ± SD. These data are representative of two independent experiments with five mice/timepoint. Data were analysed with a one-sided unpaired t-test: ***p = 0.001 (b), ***p = 0.0098 (d), *p = 0.0205 (e). p < 0.05 is considered significant.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Allie, S.R., Bradley, J.E., Mudunuru, U. et al. The establishment of resident memory B cells in the lung requires local antigen encounter. Nat Immunol 20, 97–108 (2019). https://doi.org/10.1038/s41590-018-0260-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0260-6

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