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:

Long-term retention of antigens in germinal centers is controlled by the spatial organization of the follicular dendritic cell network

An Author Correction to this article was published on 20 October 2023

This article has been updated

Abstract

Germinal centers (GCs) require sustained availability of antigens to promote antibody affinity maturation against pathogens and vaccines. A key source of antigens for GC B cells are immune complexes (ICs) displayed on follicular dendritic cells (FDCs). Here we show that FDC spatial organization regulates antigen dynamics in the GC. We identify heterogeneity within the FDC network. While the entire light zone (LZ) FDC network captures ICs initially, only the central cells of the network function as the antigen reservoir, where different antigens arriving from subsequent immunizations colocalize. Mechanistically, central LZ FDCs constitutively express subtly higher CR2 membrane densities than peripheral LZ FDCs, which strongly increases the IC retention half-life. Even though repeated immunizations gradually saturate central FDCs, B cell responses remain efficient because new antigens partially displace old ones. These results reveal the principles shaping antigen display on FDCs during the GC reaction.

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: Long-term dynamics of antigen localization in B cell follicles.
Fig. 2: CD40L signals induce FDC network expansion and activation but do not regulate antigen centralization or retention.
Fig. 3: Central and peripheral LZ FDCs express different levels of IC-binding receptors.
Fig. 4: Single-cell transcriptomics differentiates two LZ FDC clusters with different functional activity.
Fig. 5: Central and peripheral LZ FDCs show a similarly low ability to degrade antigens.
Fig. 6: Membrane levels of CR2 dictate the half-life of antigen retention on peripheral and central FDCs.
Fig. 7: Antigen display by FDCs regulates the B cell response while successive immunizations partially saturate central FDCs.

Similar content being viewed by others

Data availability

Ensembl GRCm38 was used as the reference genome to build the index. The mouse scRNAseq data are available in GEO under accession no GSE213254. Source data are provided with this paper.

Code availability

Matlab script for FDC segmentation is available at https://github.com/ptolar/FDCseqmentation. Code used to analyze scRNAseq is available at https://github.com/FrancisCrickInstitute/Cxcl13_project.

Change history

References

  1. Victora, G. D. & Nussenzweig, M. C. Germinal centers. Annu Rev. Immunol. 40, 413–442 (2022).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Batista, F. D. & Harwood, N. E. The who, how and where of antigen presentation to B cells. Nat. Rev. Immunol. 9, 15–27 (2009).

    Article  CAS  PubMed  Google Scholar 

  5. Mueller, S. N. & Germain, R. N. Stromal cell contributions to the homeostasis and functionality of the immune system. Nat. Rev. Immunol. 9, 618–629 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pikor, N. B. et al. Remodeling of light and dark zone follicular dendritic cells governs germinal center responses. Nat. Immunol. 21, 649–659 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rodda, L. B. et al. Single-cell RNA sequencing of lymph node stromal cells reveals niche-associated heterogeneity. Immunity 48, 1014–1028.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Alexandre, Y. O. et al. A diverse fibroblastic stromal cell landscape in the spleen directs tissue homeostasis and immunity. Sci. Immunol. 7, eabj0641 (2022).

    Article  CAS  PubMed  Google Scholar 

  9. Krautler, N. J. et al. Follicular dendritic cells emerge from ubiquitous perivascular precursors. Cell 150, 194–206 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Jarjour, M. et al. Fate mapping reveals origin and dynamics of lymph node follicular dendritic cells. J. Exp. Med. 211, 1109–1122 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pasparakis, M., Alexopoulou, L., Episkopou, V. & Kollias, G. Immune and inflammatory responses in TNF alpha-deficient mice: a critical requirement for TNF alpha in the formation of primary B cell follicles, follicular dendritic cell networks and germinal centers, and in the maturation of the humoral immune response. J. Exp. Med. 184, 1397–1411 (1996).

    Article  CAS  PubMed  Google Scholar 

  12. Hir, M. L. et al. Differentiation of follicular dendritic cells and full antibody responses require tumor necrosis factor receptor-1 signaling. J. Exp. Med. 183, 2367–2372 (1996).

    Article  PubMed  Google Scholar 

  13. Wang, Y., Wang, J., Sun, Y., Wu, Q. & Fu, Y.-X. Complementary effects of TNF and lymphotoxin on the formation of germinal center and follicular dendritic cells. J. Immunol. 166, 330–337 (2001).

    Article  CAS  PubMed  Google Scholar 

  14. Tumanov, A. V., Kuprash, D. V., Mach, J. A., Nedospasov, S. A. & Chervonsky, A. V. Lymphotoxin and TNF produced by B Cells are dispensable for maintenance of the follicle-associated epithelium but are required for development of lymphoid follicles in the Peyer’s patches. J. Immunol. 173, 86–91 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Wang, X. et al. Follicular dendritic cells help establish follicle identity and promote B cell retention in germinal centers. J. Exp. Med. 208, 2497–2510 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lu, E., Wolfreys, F. D., Muppidi, J. R., Xu, Y. & Cyster, J. G. S-Geranylgeranyl-l-glutathione is a ligand for human B cell-confinement receptor P2RY8. Nature 567, 244–248 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Suzuki, K. et al. The sensing of environmental stimuli by follicular dendritic cells promotes immunoglobulin a generation in the gut. Immunity 33, 71–83 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Kranich, J. et al. Follicular dendritic cells control engulfment of apoptotic bodies by secreting Mfge8. J. Exp. Med. 205, 1293–1302 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Phan, T. G., Grigorova, I., Okada, T. & Cyster, J. G. Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells. Nat. Immunol. 8, 992–1000 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Ferguson, A. R., Youd, M. E. & Corley, R. B. Marginal zone B cells transport and deposit IgM-containing immune complexes onto follicular dendritic cells. Int. Immunol. 16, 1411–1422 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Rodda, L. B., Bannard, O., Ludewig, B., Nagasawa, T. & Cyster, J. G. Phenotypic and morphological properties of germinal center dark zone Cxcl12-expressing reticular cells. J. Immunol. 195, 4781–4791 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Allen, C. D. C. & Cyster, J. G. Follicular dendritic cell networks of primary follicles and germinal centers: phenotype and function. Semin. Immunol. 20, 14–25 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Shikh, M. E. E., Sayed, R. E., Szakal, A. K. & Tew, J. G. Follicular dendritic cell (FDC)-FcgammaRIIB engagement via immune complexes induces the activated FDC phenotype associated with secondary follicle development. Eur. J. Immunol. 36, 2715–2724 (2006).

    Article  PubMed  Google Scholar 

  24. Tokatlian, T. et al. Innate immune recognition of glycans targets HIV nanoparticle immunogens to germinal centers. Science 363, 649–654 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. van der Poel, C. E. et al. Follicular dendritic cells modulate germinal center B cell diversity through FcγRIIB. Cell Rep. 29, 2745–2755.e4 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Gonzalez, S. F. et al. Complement-dependent transport of antigen into B cell follicles. J. Immunol. 185, 2659–2664 (2010).

    Article  CAS  PubMed  Google Scholar 

  27. Fang, Y., Xu, C., Fu, Y. X., Holers, V. M. & Molina, H. Expression of complement receptors 1 and 2 on follicular dendritic cells is necessary for the generation of a strong antigen-specific IgG response. J. Immunol. 160, 5273–5279 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Brockman, M. A., Verschoor, A., Zhu, J., Carroll, M. C. & Knipe, DM. Optimal long-term humoral responses to replication-defective herpes simplex virus require CD21/35 complement receptor expression on stromal cells. J. Virol. 80, 7111–7117 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hannum, L. G., Haberman, A. M., Anderson, S. M. & Shlomchik, M. J. Germinal center initiation, variable gene region hypermutation, and mutant B cell selection without detectable immune complexes on follicular dendritic cells. J. Exp. Med. 192, 931–942 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kato, Y. et al. Multifaceted effects of antigen valency on B cell response composition and differentiation in vivo. Immunity 53, 548–563.e8 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Martin, J. T. et al. Targeting HIV Env immunogens to B cell follicles in nonhuman primates through immune complex or protein nanoparticle formulations. npj Vaccines 5, 72 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Lee, J. H. et al. Long-primed germinal centres with enduring affinity maturation and clonal migration. Nature 609, 998–1004 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Foy, T. M. et al. gp39-CD40 interactions are essential for germinal center formation and the development of B cell memory. J. Exp. Med. 180, 157–163 (1994).

    Article  CAS  PubMed  Google Scholar 

  34. Szklarczyk, D. et al. STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 47, D607–D613 (2019).

    Article  CAS  PubMed  Google Scholar 

  35. Martínez‐Riaño, A. et al. Antigen phagocytosis by B cells is required for a potent humoral response. EMBO Rep. 19, e46016 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Endres, R. et al. Mature follicular dendritic cell networks depend on expression of lymphotoxin β receptor by radioresistant stromal cells and of lymphotoxin β and tumor necrosis factor by B cells. J. Exp. Med. 189, 159–168 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kapasi, Z. F., Kosco-Vilbois, M. H., Shultz, L. D., Tew, J. G. & Szakal, A. K. Cellular origin of follicular dendritic cells. Adv. Exp. Med Biol. 355, 231–235 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Gordon, L. et al. Foot-and-mouth disease virus localisation on follicular dendritic cells and sustained induction of neutralising antibodies is dependent on binding to complement receptors (CR2/CR1). PLoS Pathog. 18, e1009942 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cirelli, K. M. et al. Slow delivery immunization enhances HIV neutralizing antibody and germinal center responses via modulation of immunodominance. Cell 177, 1153–1171.e28 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ansel, K. M. et al. A chemokine-driven positive feedback loop organizes lymphoid follicles. Nature 406, 309–314 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Prados, A. et al. Fibroblastic reticular cell lineage convergence in Peyer’s patches governs intestinal immunity. Nat. Immunol. 22, 510–519 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zikherman, J., Parameswaran, R. & Weiss, A. Endogenous antigen tunes the responsiveness of naive B cells but not T cells. Nature 489, 160–164 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Noviski, M. et al. Optimal development of mature B cells requires recognition of endogenous antigens. J. Immunol. 203, 418–428 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Spillane, K. M. & Tolar, P. B cell antigen extraction is regulated by physical properties of antigen-presenting cells. J. Cell Biol. 216, 217–230 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Heesters, B. A. et al. Characterization of human FDCs reveals regulation of T cells and antigen presentation to B cells. J. Exp. Med. 218, e20210790 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Arulraj, T., Binder, S. C. & Meyer-Hermann, M. Rate of immune complex cycling in follicular dendritic cells determines the extent of protecting antigen integrity and availability to germinal center B cells. J. Immunol. 206, ji2001355 (2021).

    Article  Google Scholar 

  47. Heesters, B. A. et al. Endocytosis and recycling of immune complexes by follicular dendritic cells enhances B cell antigen binding and activation. Immunity 38, 1164–1175 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Aung, A. et al. Low protease activity in B cell follicles promotes retention of intact antigens after immunization. Science 379, eabn8934 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Suzuki, K., Grigorova, I., Phan, T. G., Kelly, L. M. & Cyster, J. G. Visualizing B cell capture of cognate antigen from follicular dendritic cells. J. Exp. Med. 206, 1485–1493 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Barrington, R. A., Pozdnyakova, O., Zafari, M. R., Benjamin, C. D. & Carroll, M. C. B lymphocyte memory: role of stromal cell complement and FcgammaRIIB receptors. J. Exp. Med. 196, 1189–1199 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Qin, D. et al. Fc gamma receptor IIB on follicular dendritic cells regulates the B cell recall response. J. Immunol. 164, 6268–6275 (2000).

    Article  CAS  PubMed  Google Scholar 

  52. Ahearn, J. M. et al. Disruption of the Cr2 locus results in a reduction in B-1a cells and in an impaired B cell response to T-dependent antigen. Immunity 4, 251–262 (1996).

    Article  CAS  PubMed  Google Scholar 

  53. Viant, C. et al. Antibody affinity shapes the choice between memory and germinal center B cell fates. Cell 183, 1298–1311.e11 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Glaros, V. et al. Limited access to antigen drives generation of early B cell memory while restraining the plasmablast response. Immunity 54, 2005–2023.e10 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Avancena, P. et al. The magnitude of germinal center reactions is restricted by a fixed number of preexisting niches. Proc. Natl Acad. Sci. USA 118, e2100576118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pappas, L. et al. Rapid development of broadly influenza neutralizing antibodies through redundant mutations. Nature 516, 418–422 (2014).

    Article  CAS  PubMed  Google Scholar 

  58. Messal, H. A. et al. Antigen retrieval and clearing for whole-organ immunofluorescence by FLASH. Nat. Protoc. 16, 239–262 (2021).

    Article  CAS  PubMed  Google Scholar 

  59. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Li, Y., Liu, P. C., Shen, Y., Snavely, M. D. & Hiraga, K. A cell-based internalization and degradation assay with an activatable fluorescence–quencher probe as a tool for functional antibody screening. J. Biomol. Screen. 20, 869–875 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Bruun, T. U. J., Andersson, A.-M. C., Draper, S. J. & Howarth, M. Engineering a rugged nanoscaffold to enhance plug-and-display vaccination. ACS Nano 12, 8855–8866 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Nagar, B., Jones, R. G., Diefenbach, R. J., Isenman, D. E. & Rini, J. M. X-ray crystal structure of C3d: a C3 fragment and ligand for complement receptor 2. Science 280, 1277–1281 (1998).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank D. Calado for the Cr2-KO (Cr2tm1Hmo) mice and B. Haynes for the YU-gp120 pcDNA3.1 plasmid. We thank M. Howarth for the SpyTag plasmids and J. Eisenman for the C3dg pET13b plasmid. We thank L. Wasim and S. Hernández for critical reading of the paper. This work was supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (CC2006), the UK Medical Research Council (CC2006) and the Wellcome Trust (CC2006), and by the UK Medical Research Council (grant MR/X009254/1). S.W. is supported by the National Science Foundation (NSF) Grant MCB-2225947 and an NSF CAREER Award PHY-2146581. For the purpose of open access, the author has applied a CC BY public copyright license to any author accepted manuscript version arising from this submission. We thank the Francis Crick Institute Animal facility, Light Microscopy, Flow Cytometry and Advanced Sequencing Technology Platforms.

Author information

Authors and Affiliations

Authors

Contributions

A.M.-R. designed and performed the experiments and analyzed the data. S.W. developed and analyzed the mathematical model of IC-FDC dissociation. S.B. analyzed the scRNAseq data. S.M. measured the CR2-C3dg binding rates. A.C. produced the YU-gp120-SpyTag protein. K.M.S. provided advice. B.L. provided the CXCL13-TdTomato mice and advice. P.T. designed the experiments and supervised the research. A.M.-R. and P.T. prepared the paper.

Corresponding author

Correspondence to Pavel Tolar.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Michael Carroll and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer review reports are available. 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 Antigens centralize on the FDC network.

a) Maximum intensity projection of confocal images of clarified LNs of mice immunized with IC-PE (magenta). FDC networks are in cyan (anti-CD21/35) (n = 6 LNs; 3 experiments). b) Image analysis to quantify antigen distribution within each B cell follicle. c) Confocal image of an FDC network (cyan) after immunization with two subsequent ICs in PBS analyzed 7 days after the first immunization (IC-PE; magenta) and 24 hours after the second (IC-488; yellow). Single-color images of IC-488 (left) and IC-PE (right) are shown below. Cyan line demarcates FDC network boundary based on anti-CD21/35 staining. Right, quantification of the distribution of both antigens on the FDC network. (n = 8 LNs; 2 experiments). d) Confocal image of an FDC network (cyan) after immunization with two subsequent ICs as in C for 14 and 7 days. Single-color images of IC-488 (left) and IC-PE (right) are shown below. Cyan line demarcates the FDC network boundary based on anti-CD21/35 staining. Right, quantification of the distribution of both antigens on the FDC network (n = 12 LNs; 2 experiments). e) Image of an LN FDC network (cyan) 56 days after immunization with IC-PE (magenta). Right, single-color image of IC-PE (gray) with cyan line demarcating the FDC network boundary based on anti-CD21/35 staining (n = 4 LNs; 1 experiment). f) Image of a draining LN 21 days after immunization with IC-PE (red). Naïve B cells are shown in gray (anti-IgD). (n = 4 LNs; 2 experiments). g) Image of a draining LN 7 days after immunization with AF555-labeled mi3-Spycatcher nanoparticles coated with YU-gp120-Spytag HIV envelope protein (magenta). FDC networks are shown in cyan (anti-CD21/35). White square indicates the region magnified. Cyan line demarcating the FDC network boundary based on anti-CD21/35 staining (n = 3 LNS; 2 experiments). h) Flow cytometry gating strategy to analyze FDCs. Quantitative data show the mean ± SD analysis by two-tailed t-test or one-way ANOVA with multiple comparisons.

Source data

Extended Data Fig. 2 B cell activation is required for FDC expansion.

a) Quantification of the FDC network volume per LN based on anti-CD21/35 staining in clarified LNs from non-immunized (n = 8 mice) and 13 days post-immunized (n = 9) mice with IC-PE. b) Representative immunofluorescence images of LN B cell GCs from mice immunized for 14 days with IC-PE. Upper row shows the merged image of GL7 (yellow), IC-PE (magenta), and anti-CD21/35 (cyan) and the corresponding single-color images (gray). Yellow line demarcates GL7 staining and magenta line IC-PE localization (n = 4 mice). Lower row shows the merged image of anti-PD1 (yellow), IC-PE (magenta) and anti-CD21/35 (cyan) and the corresponding single-color images (gray). Yellow line demarcates PD1 staining and magenta line IC-PE localization (n = 4 mice). c) FDC numbers in non-transgenic C57BL/6 (WT; n = 5) and BCR-transgenic B1-8f (B1-8flox Igκ−/−; n = 6) and MD4 mice (n = 2) 24 hours after immunization with IC-PE. d) Representative confocal images of LNs from non-tg (WT), B1-8f and MD4 mice 24 hours after immunization with IC-PE (magenta). FDC networks are shown in cyan (anti-CD21/35). The white line delimits the edges of the organs. e) Percentage of GC B cells in non-immunized (light gray; n = 4 mice) and IC-immunized mice treated with anti-CD40L (orange; n = 8 mice) or isotype control antibody (black; n = 8 mice) as described in Fig. 2b. Quantitative data show means ± SD and analysis by two-tailed one-way ANOVA with multiple comparisons.

Source data

Extended Data Fig. 3 IC-binding receptor expression on FDCs.

a) CR2, FCGR2B and FCER2A membrane expression on IC+ and IC FDCs 7 days after immunization (n = 7 mice; 2 experiments). b) Histogram showing Myosin heavy chain 11 (MYH11) expression in IC-PE+ and IC-PE FDCs 7 days after immunization. Quantitative data show means ± SD and analysis by two-tailed paired t-test.

Source data

Extended Data Fig. 4 ScRNAseq of follicular stromal cells.

a) Experimental workflow for scRNAseq of Cxcl13-TdTomato+ LN cells. Cxcl13-TdTomato mice were immunized consecutively with two ICs separated by 7 days or only with one IC. 24 h after the last immunization, draining LNs were dissociated into a single-cell suspension, stained and live cells were flow-sorted based on PDPN and TdTomato positivity. Single-sorted cells were used for 10x RNA sequencing. b) Feature plots showing expression of markers for hematopoietic cells (H2-Aa) and cytokines important for LN organization, Cxcl12 and Cxcl13. c) Violin plots showing the expression of Myh11 and Fcer2a on the three FDC clusters from Fig. 4c (LZ 1 in green, LZ 2 in red, and DZ in blue). One-tail adjusted P for multiple comparisons. d) Confocal image of a LN from a mouse after 7 days postimmunization with IC-PE (yellow). CR2 staining is shown in cyan and CD16/32 in magenta (n = 4 LNs; 2 experiments).

Extended Data Fig. 5 Antigen degradation by FDCs.

a) Quantification of Atto488 intensity after treating the control sensor lacking the BHQ-1 quencher or antigen-degradation sensor (1:9 antigen:quencher molar ratio) with protease for 30 min at 37 °C. b) Naïve B cells were incubated with beads coated with anti-IgM and the antigen-degradation sensor (green) or control sensor (orange) at indicated times at 37 °C. Plots illustrate Atto488 and AF647 intensity on B cells containing degradation sensor beads or control sensor beads. Graphs show the percentage of B cells containing quenched antigen (% of Atto488-low) and the levels of antigen degradation on B cells containing beads (measured as Atto488/AF647 intensity ratio) (2 experiments). c) Contour plots show Atto488 and AF647 levels on B cells (blue) and FDCs (gray) containing IC-antigen-degradation sensor at different time points post-immunization. d) Quantification of the antigen degradation levels in antigen+ B cells and FDCs at different time points post-immunization. (n = 7 mice; 2 experiments)). All quantitative data show means ± SD analyzed by two-tailed unpaired t-test.

Source data

Extended Data Fig. 6 In vivo IC deposition on FDCs requires CR2 expression.

a) CR2, FCER2A and FCGR2B membrane expression on FDCs from mice described in Fig. 2c. b) Experimental workflow. Lethally irradiated CD45.2 WT (n = 6) and Cr2-KO (n = 4) mice reconstituted with bone marrow cells from WT CD45.1/CD45.2 mice and immunized with IC-PE and IC-488 6 days later. c) Gating strategy based on FCGR2B and VCAM1 expression to analyze FDCs in CD45.2 WT and Cr2-KO mice reconstituted with WT CD45.1/CD45.2 BM. In gray, FCGR2B and VCAM1 expression on WT FDCs (PDPN+ CD31 Madcam1+ CD21/35hi); in red, on PDPN+ CD31 stromal cells. d) CR2 expression on WT and Cr2-KO FDCs from mice as described in (B). e) Percentage of FDCs loaded with ICPE and IC-488 from mice described in (B). f) Quantity of IC loaded in the FDC network from mice described in (B). g) Surface CR2 density on IC+ (red) or IC (gray) FDCs 7 days after immunization. Plots show the median (line), the 25th and 75th percentiles (box) and 1.5x the interquartile range (whiskers) of the number of CR2 molecules/μm2 (n = 8 mice; 2 experiments). h) Schematic of the binding of a C3d-coated IC to the surface of an FDC illustrating the mathematical modelling parameters. i) Percentage of FDCs loaded with IC-PE and amount of IC-PE 24 h after IC-PE immunization and injection of anti-CD21/35 blocking antibody (n = 2 mice). j) Immunization workflow to analyze the effect of blocking CR2-C3d binding using an anti-CD21/35 antibody (7G6). k) Percentage of FDCs within the stromal PDPN+ cells in LNs from mice untreated (black) or treated (blue) for 7 days with anti-CD21/35 (7G6) blocking antibody as shown in J (n = 4 mice). l) Number of FDCs per LN and expression of FCGR2B and VCAM1 in WT (black; n = 5), Cr2-HET (dark blue; n = 5) and Cr2-KO mice (light blue; n = 4) bone-marrow reconstituted with WT CD45.1 cells. m) Representative images of a FDC network from mice described in L. Cyan line demarcates FDC network based on FDCM1 stain (n = 5 mice; 2 experiments). Quantitative data show means ± SD and analysis by two-tailed unpaired t-test or One-way ANOVA with multiple comparisons.

Source data

Extended Data Fig. 7 Central FDCs can get partially saturated.

a) Gating strategy to analyze IgG1+ cells within GC CD45.1+ donor B cells in WT, Cr2-HET and Cr2-KO mice bone-marrow reconstituted with CD45.1+ WT cells and immunized for 21 days with IC-NP. Non-immunized mice shown as control. b) Gating strategy to analyze NP-specific B cells within GC CD45.1+ donor B cells from mice described in A. c) Gating strategy to analyze plasmablasts (CD138 + NP+ within CD45.1+/−) from mice described in A. d) Gating strategy to analyze memory B cells (CD38hi PDL-2+ within the NP-specific CD45.1+ donor cells) from mice described in A. e) Representative confocal images of FDC networks from LNs of WT, Cr2-HET and Cr2-KO mice bone-marrow reconstituted with WT CD45.1 cells and immunized for 21 days with IC NP-PE (n = 3 mice). Upper panel shows FDCM1 staining. Lower panel shows NP-PE. f) Gating strategy to analyze FDCs containing different combinations of ICs from consecutive immunizations with three (lower panel) or four (upper panel) different fluorescent antigen-ICs as indicated in Fig. 7a, b. g) Workflow to analyze antigen displacement by subsequent immunizations. Mice were immunized with IC-405 alone or followed by four subsequent immunizations with different ICs. Graphs show the percentage of FDCs loaded with the first antigen and the quantity (MFI) of the loaded antigen in the two groups of mice (n = 7 mice). h) Immunization workflow to analyze the antigen-specific antibody response generated to NP under non-saturating (2-IC) or saturating conditions (4-IC). i) Titers of high-affinity (NP(7)-BSA) and total (NP(25)-BSA) NP-specific IgG1 in sera of mice 21 and 56 days after immunization with either two antigen-ICs (No saturation condition; black; n = 8 mice) or four antigen-ICs (Saturation condition; green; n = 7 mice) as in D. j) Ratio of binding to NP(7) and NP(25), as measured by ELISA in mice immunized as in I. Quantitative data show means ± SD and analysis by two-tailed unpaired t-test with multiple comparisons.

Source data

Supplementary information

Source data

Source Data Fig. 1

Source data for Fig. 1.

Source Data Fig. 2

Source data for Fig. 2.

Source Data Fig. 3

Source data for Fig. 3.

Source Data Fig. 4

Source data for Fig. 4.

Source Data Fig. 5

Source data for Fig. 5.

Source Data Fig. 6

Source data for Fig. 6.

Source Data Fig. 7

Source data for Fig. 7.

Source Data Extended Data Fig. 1

Source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Source data for Extended Data Fig. 3.

Source Data Extended Data Fig. 5

Source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Source data for Extended Data Fig. 7.

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

Martínez-Riaño, A., Wang, S., Boeing, S. et al. Long-term retention of antigens in germinal centers is controlled by the spatial organization of the follicular dendritic cell network. Nat Immunol 24, 1281–1294 (2023). https://doi.org/10.1038/s41590-023-01559-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01559-1

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