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:

Class-switched memory B cells remodel BCRs within secondary germinal centers

Abstract

Effective vaccines induce high-affinity memory B cells and durable antibody responses through accelerated mechanisms of natural selection. Secondary changes in antibody repertoires after vaccine boosts suggest progressive rediversification of B cell receptors (BCRs), but the underlying mechanisms remain unresolved. Here, the integrated specificity and function of individual memory B cell progeny revealed ongoing evolution of polyclonal antibody specificities through germinal center (GC)-specific transcriptional activity. At the clonal and subclonal levels, single-cell expression of the genes encoding the costimulatory molecule CD83 and the DNA polymerase Polη segregated the secondary GC transcriptional program into four stages that regulated divergent mechanisms of memory BCR evolution. Our studies demonstrate that vaccine boosts reactivate a cyclic program of GC function in class-switched memory B cells to remodel existing antibody specificities and enhance durable immunological protection.

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

Figure 1: Class-switched memory B cells form robust secondary GCs.
Figure 2: Diversification of switched BCRs and secondary GC transcription.
Figure 3: Expression of Cd83 and Polh assorts four cyclic stages of GC activity.
Figure 4: ‘Adaptive radiation’ during subclonal BCR evolution in the GC.
Figure 5: De novo secondary GC formation.
Figure 6: Class-switched memory B cells can form secondary GCs after transfer and recall.
Figure 7: Class-switched memory B cells are dominant precursor cells for secondary GCs.

Similar content being viewed by others

References

  1. Crotty, S. Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663 (2011).

    Article  CAS  PubMed  Google Scholar 

  2. Fazilleau, N., Mark, L., McHeyzer-Williams, L.J. & McHeyzer-Williams, M.G. Follicular helper T cells: lineage and location. Immunity 30, 324–335 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Victora, G.D. & Nussenzweig, M.C. Germinal centers. Annu. Rev. Immunol. 30, 429–457 (2012).

    CAS  PubMed  Google Scholar 

  4. MacLennan, I.C. Germinal centers. Annu. Rev. Immunol. 12, 117–139 (1994).

    CAS  PubMed  Google Scholar 

  5. McHeyzer-Williams, M., Okitsu, S., Wang, N. & McHeyzer-Williams, L. Molecular programming of B cell memory. Nat. Rev. Immunol. 12, 24–34 (2012).

    CAS  Google Scholar 

  6. Allen, C.D., Okada, T., Tang, H.L. & Cyster, J.G. Imaging of germinal center selection events during affinity maturation. Science 315, 528–531 (2007).

    CAS  PubMed  Google Scholar 

  7. Hauser, A.E. et al. Definition of germinal-center B cell migration in vivo reveals predominant intrazonal circulation patterns. Immunity 26, 655–667 (2007).

    CAS  PubMed  Google Scholar 

  8. Schwickert, T.A. et al. In vivo imaging of germinal centres reveals a dynamic open structure. Nature 446, 83–87 (2007).

    CAS  PubMed  Google Scholar 

  9. Victora, G.D. et al. Germinal center dynamics revealed by multiphoton microscopy with a photoactivatable fluorescent reporter. Cell 143, 592–605 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Shulman, Z. et al. T follicular helper cell dynamics in germinal centers. Science 341, 673–677 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Bannard, O. et al. Germinal center centroblasts transition to a centrocyte phenotype according to a timed program and depend on the dark zone for effective selection. Immunity 39, 912–924 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Rajewsky, K. Clonal selection and learning in the antibody system. Nature 381, 751–758 (1996).

    CAS  PubMed  Google Scholar 

  13. Muramatsu, M. et al. Class switch recombination and hypermutation require activation-induced cytidine deaminase (AID), a potential RNA editing enzyme. Cell 102, 553–563 (2000).

    CAS  PubMed  Google Scholar 

  14. Shulman, Z. et al. Dynamic signaling by T follicular helper cells during germinal center B cell selection. Science 345, 1058–1062 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Gitlin, A.D., Shulman, Z. & Nussenzweig, M.C. Clonal selection in the germinal centre by regulated proliferation and hypermutation. Nature 509, 637–640 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Berek, C., Berger, A. & Apel, M. Maturation of the immune response in germinal centers. Cell 67, 1121–1129 (1991).

    CAS  PubMed  Google Scholar 

  17. Jacob, J., Kelsoe, G., Rajewsky, K. & Weiss, U. Intraclonal generation of antibody mutants in germinal centres. Nature 354, 389–392 (1991).

    CAS  PubMed  Google Scholar 

  18. Allen, D. et al. Timing, genetic requirements and functional consequences of somatic hypermutation during B-cell development. Immunol. Rev. 96, 5–22 (1987).

    CAS  PubMed  Google Scholar 

  19. Berek, C. & Milstein, C. Mutation drift and repertoire shift in the maturation of the immune response. Immunol. Rev. 96, 23–41 (1987).

    CAS  PubMed  Google Scholar 

  20. Siekevitz, M., Kocks, C., Rajewsky, K. & Dildrop, R. Analysis of somatic mutation and class switching in naive and memory B cells generating adoptive primary and secondary responses. Cell 48, 757–770 (1987).

    CAS  PubMed  Google Scholar 

  21. Benson, M.J. et al. Distinction of the memory B cell response to cognate antigen versus bystander inflammatory signals. J. Exp. Med. 206, 2013–2025 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  24. Zabel, F. et al. Viral particles drive rapid differentiation of memory B cells into secondary plasma cells producing increased levels of antibodies. J. Immunol. 192, 5499–5508 (2014).

    CAS  PubMed  Google Scholar 

  25. Kometani, K. et al. Repression of the transcription factor Bach2 contributes to predisposition of IgG1 memory B cells toward plasma cell differentiation. Immunity 39, 136–147 (2013).

    CAS  PubMed  Google Scholar 

  26. Corti, D. & Lanzavecchia, A. Broadly neutralizing antiviral antibodies. Annu. Rev. Immunol. 31, 705–742 (2013).

    CAS  PubMed  Google Scholar 

  27. Gao, F. et al. Cooperation of B cell lineages in induction of HIV-1-broadly neutralizing antibodies. Cell 158, 481–491 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Jiang, N. et al. Lineage structure of the human antibody repertoire in response to influenza vaccination. Sci. Transl. Med. 5, 171ra119 (2013).

    Google Scholar 

  29. Klein, F. et al. Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization. Cell 153, 126–138 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Liao, H.X. et al. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 496, 469–476 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Vollmers, C., Sit, R.V., Weinstein, J.A., Dekker, C.L. & Quake, S.R. Genetic measurement of memory B-cell recall using antibody repertoire sequencing. Proc. Natl. Acad. Sci. USA 110, 13463–13468 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. West, A.P. Jr. et al. Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156, 633–648 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Wrammert, J. et al. Rapid cloning of high-affinity human monoclonal antibodies against influenza virus. Nature 453, 667–671 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Kaji, T. et al. Both mutated and unmutated memory B cells accumulate mutations in the course of the secondary response and develop a new antibody repertoire optimally adapted to the secondary stimulus. Int. Immunol. 25, 683–695 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. McHeyzer-Williams, L.J., Cool, M. & McHeyzer-Williams, M.G. Antigen-specific B cell memory: expression and replenishment of a novel b220(-) memory b cell compartment. J. Exp. Med. 191, 1149–1166 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. McHeyzer-Williams, M.G., Nossal, G.J. & Lalor, P.A. Molecular characterization of single memory B cells. Nature 350, 502–505 (1991).

    CAS  PubMed  Google Scholar 

  38. Shapiro-Shelef, M. et al. Blimp-1 is required for the formation of immunoglobulin secreting plasma cells and pre-plasma memory B cells. Immunity 19, 607–620 (2003).

    CAS  PubMed  Google Scholar 

  39. Huson, D.H. & Scornavacca, C. Dendroscope 3: an interactive tool for rooted phylogenetic trees and networks. Syst. Biol. 61, 1061–1067 (2012).

    PubMed  Google Scholar 

  40. Amir el, A.D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013).

    Google Scholar 

  41. Delbos, F., Aoufouchi, S., Faili, A., Weill, J.C. & Reynaud, C.A. DNA polymerase eta is the sole contributor of A/T modifications during immunoglobulin gene hypermutation in the mouse. J. Exp. Med. 204, 17–23 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Tze, L.E. et al. CD83 increases MHC II and CD86 on dendritic cells by opposing IL-10-driven MARCH1-mediated ubiquitination and degradation. J. Exp. Med. 208, 149–165 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Kasturi, S.P. et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature 470, 543–547 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Schwickert, T.A., Alabyev, B., Manser, T. & Nussenzweig, M.C. Germinal center reutilization by newly activated B cells. J. Exp. Med. 206, 2907–2914 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  46. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Kinoshita, K., Harigai, M., Fagarasan, S., Muramatsu, M. & Honjo, T. A hallmark of active class switch recombination: transcripts directed by I promoters on looped-out circular DNAs. Proc. Natl. Acad. Sci. USA 98, 12620–12623 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Calado, D.P. et al. The cell-cycle regulator c-Myc is essential for the formation and maintenance of germinal centers. Nat. Immunol. 13, 1092–1100 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Dominguez-Sola, D. et al. The proto-oncogene MYC is required for selection in the germinal center and cyclic reentry. Nat. Immunol. 13, 1083–1091 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Aiba, Y. et al. Preferential localization of IgG memory B cells adjacent to contracted germinal centers. Proc. Natl. Acad. Sci. USA 107, 12192–12197 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Supported by the Foundation Bettencourt-Schueller (P.J.M.), the Swiss National Science Foundation (S.L.O.), the Novartis Jubliaeumsstiftung (S.L.O.), the Roche Research Foundation (S.L.O.) and the US National Institutes of Health (AI047231, AI040215 and AI071182 to M.G.M.-W.). This is The Scripps Research Institute manuscript number 26086.

Author information

Authors and Affiliations

Authors

Contributions

L.J.M.-W., P.J.M. and M.G.M.-W. designed and performed experiments, analyzed the data and wrote the paper; and S.L.O. designed and performed the experiments in Supplementary Figure 1c.

Corresponding author

Correspondence to Michael G McHeyzer-Williams.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Antigen-specific secondary GCs from mice primed with different adjuvants and different doses of antigen.

(a) Mice were primed with 400μg NP-KLH prepared in Alum (top row), CFA (middle row) or MPL (bottom row) adjuvants and rested for 70 days before boosting with soluble NP-KLH. NP-specific GC B cells (λ1+ NP+ B220+ CD138 CD38 GL7+ class-switched B cells) were detected in draining lymph nodes for all adjuvant-primed mice 4 days after boost. Numbers indicate mean ± sem percentage across 3 or more mice per group.

(b) Total class-switched NP-specific B cells (top) and NP-specific GC B cells (bottom) per draining lymph node of primed animals before (day 70) or 4 days after soluble NP-KLH boost in Alum, CFA and MPL-primed groups. Bars represent mean ± sem. Fold increases after boost are indicated.

(c) Mice were primed with 4μg (top row) or 40μg (bottom row) NP-KLH prepared in MPL adjuvant and rested for >240 days before boosting with NP-KLH in adjuvant. NP-specific GC B cells (λ1+ NP+ B220+ CD138 CD38 GL7+ class-switched B cells) were detected in draining lymph nodes for all doses of primed mice 5 days after boost. Numbers indicate mean ± sem percentage across 3 mice per group.

(d) Total class-switched NP-specific B cells (left) and NP-specific GC B cells (right) of mice primed with 4μg or 40μg NP-KLH in MPL and boosted with NP-KLH in adjuvant as in c. Bars represent mean ± sem.

Supplementary Figure 2 Phylogenetic tree of rearranged BCR-encoding genes from individual antigen-specific secondary GC cells.

Mice were primed with NP-KLH prepared in MPL adjuvant and rested for 70 days before boosting. Single NP-specific GC B cells (λ1+ NP+ B220+ CD138 CD38 GL7+ class-switched B cells were sorted from draining lymph nodes of mice 8 days after boost with NP-KLH in adjuvant. Nucleotide sequence of VH186.2, DH and JH joints amplified from the mRNA was compared to the germline VH186.2. Displayed is the BCR VH186.2 amino acid sequence for each cell. Only amino acids differing from the germline sequence (top) are presented with the identical amino acid indicated by dashes. The DH and JH regions are identified at the right of each sequence with the total number of VH nucleotide mutations. All boost day 8 germinal center single cells were near-neighbor sequence aligned using Clustal Omega and based on nucleotide sequence in BCR junctional regions assigned a clone and cell ID. A phylogenetic tree generated with Dendroscope 3 is displayed to the left of each cell sequence. n= 133 single cells from 4 mice. Boost day 8 clone G is highlighted in blue.

Supplementary Figure 3 Broad dynamic range for single-cell quantitative RT-PCR with minimal loss of sensitivity.

(a) Representative single cell qPCR analysis of antigen-specific GC B cells day 8 after boost (GR1 CD3, B cell+, IgD IgM, λ1+, NP+, CD138, B220+, CD38, GL7+). 10, 3 and 1 cell were sorted per well and assayed across 96 genes. Wells with 0 sorted cells were processed as negative controls. Specific target amplification for cDNA synthesis and PCR pre-amplification was followed by qPCR using nanofluidics technology. Heatmap of relative Ct value showing representative 10,3 and 1 cell for the indicated genes is displayed.

(b) Comparison of gene expression values (30-Ct) measured by single cell qPCR in 10-cell and 1-cell samples of day 8 GC B cells. For every gene in the analysis panel (n=87), the actual gene expression value measured from a 10-cell sample is plotted according to the value expected from single cell expression values for the same gene in the same population. To compute the expected value (30-Ct), we used the following formula: Expected = log2[10 x percentage of positive cells x mean (230-Ct observed in positive single cells)]. The resulting comparison has a strong correlation coefficient (r = 0.9287), and the equation of the corresponding linear regression is Actual = (0.9882 ± 0.04822) x Expected + (0.9440 ± 0.7388). Thus for the vast majority of assays, there is no sensitivity loss when measuring expression in single cell samples.

Supplementary Figure 4 Principal-component analysis for GC B cells emphasizing the distribution of the expression of Cd83 and Polh at the single-cell level.

(a) Single cell gene expression values (30-Ct, 87 genes) of n=372 GC B cells from days 4 and 8 after boost (soluble and adjuvant) were used for principal component analysis (PCA). Dot plots show the position of every cell on the space defined by the first two principal components (PC1 and PC2) (left). Cells positive for Cd83 (middle left) or Polh (middle right) expression are highlighted in yellow. Cells expressing distinct combinations of Cd83 and Polh are highlighted (right, Cd83 Polh black, Cd83+ Polh blue, Cd83+ Polh+ white, Cd83 Polh+ yellow).

(b) The same cells were subjected to PCA using only expression of 9 genes in the dataset previously published to be significantly different between DZ and LZ GC B cells (Victora et al, Cell 2010). PC1 from this analysis separates DZ and LZ GC B cells and tightly correlates with PC2 computed with all genes (left, correlation coefficient r = -0.7559, p < 0.0001). LZ and DZ gates were set according to the expression levels of Cd83 and Polh as shown (right).

(c) Based on the gates defined in b, relative proportions of GC B cells in the DZ or the LZ in primary GC (day 70) or secondary GC (days 4 and 8, soluble and adjuvant) were computed. Bars indicate mean ± sem (n=3 experiments).

(d) Based on the gates defined in b, the differences in gene expression between LZ and DZ GC B cells for every gene as described in Materials and Methods was computed. A “volcano” plot was generated to display genes significantly up-regulated in LZ (left side) or DZ (right side) B cells. The dotted line shows a p-value of 0.05. Most significantly up- or down-regulated genes are labeled on the plot.

Supplementary Figure 5 Distribution of the frequency and level of gene expression across the DZ and LZ of the GC.

Single cell gene expression values (30-Ct) of n=372 GC B cells from days 4 and 8 after boost (soluble and adjuvant) were used for principal component analysis (PCA). The cells were subjected to PCA using only expression of 9 genes previously published to be significantly different between DZ and LZ GC B cells (Victora et al, Cell 2010). GC B cells were distributed in 8 consecutive bins according to their PC1 (9 genes) values. Within each bin, the percentage of positive cells and the mean expression level (30-Ct) of positive cells was computed for every gene. We display the resulting graphs for 12 selected genes, with bars corresponding to percentage positive (left Y axis), and blue line corresponding to expression levels (right Y axis). Gene expression variation between DZ and LZ (low PC1 to high PC1) is displayed.

Supplementary Figure 6 Cyclic changes in the expression of Cd83 and Polh in GC transcriptional programming.

(a) GC B cells (from boost day 4 and day 8, n=372) were combined and clustered in a two-dimensional display using t-distributed stochastic neighbor embedding (t-SNE) algorithm. Using Cd83, Polh, Aicda and Mki67 expression, the major sub-groups are displayed and labeled 1 through 4 as described in Figure 3b.

(b) Using the graph-based trajectory algorithm, Wanderlust (Bendall, et al, Cell, 2014), Cd83 Polh cells were set as the user-defined starting point (Stage1). Wanderlust then computed a trajectory from these starting cells set at 0 through to the end-point at 1. The upper panel shows the trajectory expression patterns for Cd83 (white line) and Polh (grey dashed line). The lower panel displays t-SNE1 and t-SNE2 for the 372 GC cells. The Wanderlust predicted trajectory is displayed by the color code from blue through to dark red. The predicted trajectory is indicated as Cd83 Polh (Stage 1, blue), Cd83+ Polh (Stage 2, green), Cd83+ Polh+ (Stage 3, light red), Cd83 Polh+ (Stage 4, dark red).

(c) Proposed schematic for GC B cell stages and transitions in cyclic activity.

Supplementary Figure 7 Class-switched memory B cells form GCs upon adoptive transfer into Rag1–/– hosts.

(a) C57BL/6 mice were immunized 70 days prior with NP-KLH in adjuvant and cells sorted for adoptive transfer of class-switched memory B cells into B6.Rag1–/– hosts. Representative gates used for sorting donor memory B cells (CD8 Gr1 IgM IgD CD4 CD19+ GL7) and memory T cells (CD8 Gr1 IgM IgD CD19 CD4+ CD44high) are shown.

(b) Sorted memory B and T cells were transferred, along with congenically labeled spleen cells from B6.MD4 BCR transgenic mice, into Rag1–/– hosts that were subsequently immunized with NP-KLH in MPL adjuvant (n=5) or MPL alone (n=2). Representative plots of memory-derived B cells in host spleens 14 days after transfer are shown. Antigen-specific (λ1+ NP+) total B cell progenies of class-switched memory B cells in recipients boosted with adjuvant only (MPL, top left) or with antigen and adjuvant (NP-KLH in MPL, bottom left). GC B cells (CD19+ B220+ CD38 GL7+) progenies of class-switched memory B cells in NP-KLH boosted recipients for all donor-derived B cells (top right) or antigen-specific (bottom right) B cells. Numbers in gates indicate mean ± sem percentage.

(c) Total numbers of non-specific (top) and NP-specific (bottom) GC B cells in recipients are summarized in the column dot plots. Each dot represents an individual mouse and the bar is set at the mean.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1 and 2 (PDF 7706 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

McHeyzer-Williams, L., Milpied, P., Okitsu, S. et al. Class-switched memory B cells remodel BCRs within secondary germinal centers. Nat Immunol 16, 296–305 (2015). https://doi.org/10.1038/ni.3095

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3095

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