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:

Defining B cell immunodominance to viruses

Abstract

Immunodominance (ID) defines the hierarchical immune response to competing antigens in complex immunogens. Little is known regarding B cell and antibody ID despite its importance in immunity to viruses and other pathogens. We show that B cells and serum antibodies from inbred mice demonstrate a reproducible ID hierarchy to the five major antigenic sites in the influenza A virus hemagglutinin globular domain. The hierarchy changed as the immune response progressed, and it was dependent on antigen formulation and delivery. Passive antibody transfer and sequential infection experiments demonstrated 'original antigenic suppression', a phenomenon in which antibodies suppress memory responses to the priming antigenic site. Our study provides a template for attaining deeper understanding of antibody ID to viruses and other complex immunogens.

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: Characterization of Δ4 viruses.
Figure 2: B cell kinetics and Ab immunodominance after IAV infection.
Figure 3: Intraperitoneal and intramuscular immunizations elicit similar Ab immunodominance patterns distinct from that in response to infection.
Figure 4: CD4+ T cells positively modulate the magnitude and duration of Ab responses but not their immunodominance.
Figure 5: Ab immunodominance varies between mouse strains.
Figure 6: Abs directed to different antigenic sites show distinct functionalities.
Figure 7: Pre-existing Abs influence immunodominance in recall responses.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Yewdell, J.W. Confronting complexity: real-world immunodominance in antiviral CD8+ T cell responses. Immunity 25, 533–543 (2006).

    Article  CAS  Google Scholar 

  2. Krammer, F. & Palese, P. Advances in the development of influenza virus vaccines. Nat. Rev. Drug Discov. 14, 167–182 (2015).

    Article  CAS  Google Scholar 

  3. Burton, D.R., Poignard, P., Stanfield, R.L. & Wilson, I.A. Broadly neutralizing antibodies present new prospects to counter highly antigenically diverse viruses. Science 337, 183–186 (2012).

    Article  CAS  Google Scholar 

  4. Wheatley, A.K. & Kent, S.J. Prospects for antibody-based universal influenza vaccines in the context of widespread pre-existing immunity. Expert Rev. Vaccines 14, 1227–1239 (2015).

    Article  CAS  Google Scholar 

  5. Victora, G.D. & Wilson, P.C. Germinal center selection and the antibody response to influenza. Cell 163, 545–548 (2015).

    Article  CAS  Google Scholar 

  6. Hutchinson, E.C. et al. Conserved and host-specific features of influenza virion architecture. Nat. Commun. 5, 4816 (2014).

    Article  CAS  Google Scholar 

  7. Reading, S.A. & Dimmock, N.J. Neutralization of animal virus infectivity by antibody. Arch. Virol. 152, 1047–1059 (2007).

    Article  CAS  Google Scholar 

  8. Altman, M.O., Bennink, J.R., Yewdell, J.W. & Herrin, B.R. Lamprey VLRB response to influenza virus supports universal rules of immunogenicity and antigenicity. eLife 4, e07467 (2015).

    Article  Google Scholar 

  9. Caton, A.J., Brownlee, G.G., Yewdell, J.W. & Gerhard, W. The antigenic structure of the influenza virus A/PR/8/34 hemagglutinin (H1 subtype). Cell 31, 417–427 (1982).

    Article  CAS  Google Scholar 

  10. Gerhard, W., Yewdell, J., Frankel, M.E. & Webster, R. Antigenic structure of influenza virus hemagglutinin defined by hybridoma antibodies. Nature 290, 713–717 (1981).

    Article  CAS  Google Scholar 

  11. Yewdell, J.W., Webster, R.G. & Gerhard, W.U. Antigenic variation in three distinct determinants of an influenza type A hemagglutinin molecule. Nature 279, 246–248 (1979).

    Article  CAS  Google Scholar 

  12. Kavaler, J., Caton, A.J., Staudt, L.M., Schwartz, D. & Gerhard, W. A set of closely related antibodies dominates the primary antibody response to the antigenic site CB of the A/PR/8/34 influenza virus hemagglutinin. J. Immunol. 145, 2312–2321 (1990).

    CAS  PubMed  Google Scholar 

  13. McKean, D. et al. Generation of antibody diversity in the immune response of BALB/c mice to influenza virus hemagglutinin. Proc. Natl. Acad. Sci. USA 81, 3180–3184 (1984).

    Article  CAS  Google Scholar 

  14. Staudt, L.M. & Gerhard, W. Generation of antibody diversity in the immune response of BALB/c mice to influenza virus hemagglutinin. I. Significant variation in repertoire expression between individual mice. J. Exp. Med. 157, 687–704 (1983).

    Article  CAS  Google Scholar 

  15. Das, S.R. et al. Defining influenza A virus hemagglutinin antigenic drift by sequential monoclonal antibody selection. Cell Host Microbe 13, 314–323 (2013).

    Article  CAS  Google Scholar 

  16. Lubeck, M. & Gerhard, W. Conformational changes at topologically distinct antigenic sites on the influenza A/PR/8/34 virus HA molecule are induced by the binding of monoclonal antibodies. Virology 118, 1–7 (1982).

    Article  CAS  Google Scholar 

  17. Marshall, D., Sealy, R., Sangster, M. & Coleclough, C. TH cells primed during influenza virus infection provide help for qualitatively distinct antibody responses to subsequent immunization. J. Immunol. 163, 4673–4682 (1999).

    CAS  PubMed  Google Scholar 

  18. Rothaeusler, K. & Baumgarth, N. B cell fate decisions following influenza virus infection. Eur. J. Immunol. 40, 366–377 (2010).

    Article  CAS  Google Scholar 

  19. Sealy, R., Surman, S., Hurwitz, J.L. & Coleclough, C. Antibody response to influenza infection of mice: different patterns for glycoprotein and nucleocapsid antigens. Immunology 108, 431–439 (2003).

    Article  CAS  Google Scholar 

  20. Baumgarth, N. How specific is too specific? B cell responses to viral infections reveal the importance of breadth over depth. Immunol. Rev. 255, 82–94 (2013).

    Article  Google Scholar 

  21. Whittle, J.R. et al. Flow cytometry reveals that H5N1 vaccination elicits cross-reactive stem-directed antibodies from multiple Ig heavy-chain lineages. J. Virol. 88, 4047–4057 (2014).

    Article  Google Scholar 

  22. Frank, G.M. et al. A simple flow-cytometric method measuring B cell surface immunoglobulin avidity enables characterization of affinity maturation to influenza A virus. MBio 6, e01156 (2015).

    Article  CAS  Google Scholar 

  23. Tsilibary, E.C. & Wissig, S.L. Light and electron microscope observations of the lymphatic drainage units of the peritoneal cavity of rodents. Am. J. Anat. 180, 195–207 (1987).

    Article  CAS  Google Scholar 

  24. Lee, B.O. et al. CD4 T cell–independent antibody response promotes resolution of primary influenza infection and helps to prevent reinfection. J. Immunol. 175, 5827–5838 (2005).

    Article  CAS  Google Scholar 

  25. Mozdzanowska, K., Furchner, M., Zharikova, D., Feng, J. & Gerhard, W. Roles of CD4+ T cell–independent and T cell–dependent antibody responses in the control of influenza virus infection: evidence for noncognate CD4+ T cell activities that enhance the therapeutic activity of antiviral antibodies. J. Virol. 79, 5943–5951 (2005).

    Article  CAS  Google Scholar 

  26. Hai, R. et al. Influenza viruses expressing chimeric hemagglutinins: globular head and stalk domains derived from different subtypes. J. Virol. 86, 5774–5781 (2012).

    Article  CAS  Google Scholar 

  27. Bennink, J.R. & Doherty, P.C. The response to H-2-different virus-infected cells is mediated by long-lived T lymphocytes and is diminished by prior virus priming in a syngeneic environment. Cell. Immunol. 61, 220–224 (1981).

    Article  CAS  Google Scholar 

  28. Jamieson, B.D. & Ahmed, R. T cell memory. Long-term persistence of virus-specific cytotoxic T cells. J. Exp. Med. 169, 1993–2005 (1989).

    Article  CAS  Google Scholar 

  29. Kim, J.H., Skountzou, I., Compans, R. & Jacob, J. Original antigenic sin responses to influenza viruses. J. Immunol. 183, 3294–3301 (2009).

    Article  CAS  Google Scholar 

  30. Kavaler, J., Caton, A.J., Staudt, L.M. & Gerhard, W. A B cell population that dominates the primary response to influenza virus hemagglutinin does not participate in the memory response. Eur. J. Immunol. 21, 2687–2695 (1991).

    Article  CAS  Google Scholar 

  31. Churchill, G.A. et al. The Collaborative Cross, a community resource for the genetic analysis of complex traits. Nat. Genet. 36, 1133–1137 (2004).

    Article  CAS  Google Scholar 

  32. Linderman, S.L. et al. Potential antigenic explanation for atypical H1N1 infections among middle-aged adults during the 2013–2014 influenza season. Proc. Natl. Acad. Sci. USA 111, 15798–15803 (2014).

    Article  CAS  Google Scholar 

  33. Jelley-Gibbs, D.M. et al. Unexpected prolonged presentation of influenza antigens promotes CD4 T cell memory generation. J. Exp. Med. 202, 697–706 (2005).

    Article  CAS  Google Scholar 

  34. Waffarn, E.E. & Baumgarth, N. Protective B cell responses to flu—no fluke! J. Immunol. 186, 3823–3829 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  36. Hollowood, K. & Macartney, J. Cell kinetics of the germinal center reaction—a stathmokinetic study. Eur. J. Immunol. 22, 261–266 (1992).

    Article  CAS  Google Scholar 

  37. Zammit, D.J., Turner, D.L., Klonowski, K.D., Lefrançois, L. & Cauley, L.S. Residual antigen presentation after influenza virus infection affects CD8 T cell activation and migration. Immunity 24, 439–449 (2006).

    Article  CAS  Google Scholar 

  38. Webster, R.G., Fazekas de St.Groth, S. & Webster, R.G. Disquisition on original antigenic sin. I. Evidence in man. J. Exp. Med. 124, 331–345 (1966).

    Article  Google Scholar 

  39. Kim, J.H., Davis, W.G., Sambhara, S. & Jacob, J. Strategies to alleviate original antigenic sin responses to influenza viruses. Proc. Natl. Acad. Sci. USA 109, 13751–13756 (2012).

    Article  CAS  Google Scholar 

  40. Andrews, S.F. et al. High pre-existing serological antibody levels correlate with diversification of the influenza vaccine response. J. Virol. 89, 3308–3317 (2015).

    Article  CAS  Google Scholar 

  41. Sasaki, S. et al. Influence of prior influenza vaccination on antibody and B cell responses. PLoS One 3, e2975 (2008).

    Article  Google Scholar 

  42. Fleishman, S.J. et al. Computational design of proteins targeting the conserved stem region of influenza hemagglutinin. Science 332, 816–821 (2011).

    Article  CAS  Google Scholar 

  43. Gamblin, S.J. et al. The structure and receptor binding properties of the 1918 influenza hemagglutinin. Science 303, 1838–1842 (2004).

    Article  CAS  Google Scholar 

  44. Yewdell, J.W. & Gerhard, W. Antigenic characterization of viruses by monoclonal antibodies. Annu. Rev. Microbiol. 35, 185–206 (1981).

    Article  CAS  Google Scholar 

  45. Pieri, L., Sassoli, C., Romagnoli, P. & Domenici, L. Use of periodate–lysine–paraformaldehyde for the fixation of multiple antigens in human skin biopsies. Eur. J. Histochem. 46, 365–375 (2002).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the NIAID Comparative Medicine Branch for maintaining the mice used in this study, and P. Palese and F. Krammer (Icahn School of Medicine at Mount Sinai) for the chimeric HA virus construct. Supported by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases (J.W.Y. and A.B.M.).

Author information

Authors and Affiliations

Authors

Contributions

D.A. designed and performed experiments, analyzed data and wrote the paper; J.S.G., M.A., I.K., H.D.H., G.M.F. and S.R.D. designed and performed experiments and analyzed data; A.K.W., M.P., D.J.L. and A.B.M. designed and generated critical reagents for the study; and J.W.Y. designed experiments, analyzed data and wrote the paper. All authors provided useful comments on the manuscript.

Corresponding author

Correspondence to Jonathan W Yewdell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quantification of the virus-purified HAs and their specificities.

(a) Sera from two individual i.n. infected guinea pigs were absorbed on the viruses indicated on the X axis. The depleted sera was tested for its capacity to block binding of site-specific Fabs. The results are presented as relative binding inhibition, where binding of Fab to PR8 virus pre incubated with neat serum equals 100% and binding inhibition to virus with no competing serum is zero. Results are from three technical replicates of two individual guinea pigs. Columns represent means and SEM (bars). (b) ELISA comparison between the virus-purified HAs and rHA showing accurate quantification of the purified glycoproteins. Results are normalized to PR8 AUC set as 100%. Columns represent mean and SEM (bars) of 5 distinct sera. We also used total protein amount, Coomassie staining, western blotting, head-specific mAbs and anti-stem mAbs (not shown) to confirm that similar amounts of antigen were used in ELISA assays for all PR8 derived HAs.

Supplementary Figure 2 Gating strategy and specificity of rHA probes.

We infected mice i.n. with 50 TCID50 PR8. (a) Flow plot depicting gating strategy to identify GC B cells based on live/dead EMA- and CD3ɛ- B220+ CD38- GL7+ surface expression. Only GC B cells specifically bind rHA. (b) The dot plot depicts one representative experiment showing the % of GC B cells that are stained by the recombinant probes. Shown is one representative example of three independent experiments with 5 pooled mice each (for both a and b). (c) We infected mice were i.n. with 50 TCID50 of either PR8 or J1, a reassortant virus with all the gene segments from PR8 except for segment 4 encoding H3 HA. At 28 d p.i, we pooled MLN from 5 infected mice and stained as above. Shown are results at 28 d.p.i. of three independent experiments with 5 mice pooled each (PR8) or one experiment with 5 mice pooled (J1).Columns represent means and SEM (bars). (d) Bar graph showing cumulative MLN GC B cell responses. After S12 AUC subtraction, the sum of the B cell frequency to the 5 antigenic sites is similar to the PR8 response. Columns represent means and SEM (bars).

Supplementary Figure 3 Heavy-chain composition of immune sera.

We infected mice infected i.n. with 50 TCID50 PR8. The sera analyzed in Figure 2C were subjected to heavy chain ELISA to determine the proportion of antigenic site-specific IgG (filled bars) vs. IgM (striped bars). Columns represent means and SEM (bars).

Supplementary Figure 4 Splenic B cell kinetics and immunodominance in IAV-infected and immunized mice.

(a) Bar graph showing the frequency of antigenic site-specific germinal center B cells at different d.p.i. S12 frequencies were considered baseline and subtracted from PR8 and Δ4 values. Column represent mean and SEM (bars). Shown are three independent experiments with three pooled spleens each. (b) Scatter plot showing the correlation between ELISA recognition of the different HAs (Fig. 2c) and frequency of splenic GC B cells P=0.4319 r=0.1976. Dashed lines are 95% confidence intervals. Scatter plots showing the correlation between ELISA recognition of different HAs (Fig. 2c) and the different cell numbers. Data is the same presented in Fig. 2b and Supplementary Fig. 3a but expressed as total number of cells in the MLN P<0.0001 r=0.8095 (c), spleen P=0.0838 r=0.4186 (d) or total cell number (MLN+spleen) P=0.0001 r=0.7865 (e). Circles represent 14 d p.i., squares, 21 d p.i. and triangles, 28 d p.i.. Dashed lines represent 95% confidence intervals. (f) We immunized mice i.p. with 2500 HAU of UV-inactivated PR8. Scatter plot shows correlation between the frequency of MLN versus splenic GC B cells. P<0.0001 r=0.9941. Shown is the average from two independent experiments with five pooled mice each.

Supplementary Figure 5 CD4+ T cell depletion.

We depleted CD4+ T cells as described in Fig. 4. Shown are representative flow cytometry plots showing CD4+ and CD8+ T cells (gated on live, CD3+, B220-) (a) and GC B cells (gated on live, CD3-, B220+) (b) in untreated versus CD4-depleted animal at 14 d.p.i.. (c) LN sectioning and immunofluorescent staining comparing GC formation in untreated versus CD4-depleted animals in LN following i.n. infection and spleen following i.p. immunization. Representative flow cytometry of one spleen from two independent experiments with five and three mice per group (a), one spleen from one experiment with three mice per group (b). Representative microscopy of one spleen from one experiment with three mice per group (c)

Supplementary Figure 6 Δ4 virus infection generates Abs specific to the intact antigenic site.

(a) ELISA results showing the reactivity of Δ4 infected animals (from Fig. 6) on PR8 HA (Δ infected) and the reactivity of PR8 infected animals on Δ4 and S12 (PR8 infected). Column represent mean and bars SEM (n=4 for PR8, Sa, Ca1, Ca2; n=5 for Cb and n=8 for Sb). (b) Correlation between the AUC values presented in A. P=0.0105 r=0.9573. Dashed lines are 95% confidence intervals.

Supplementary Figure 7 Pre-existing Abs influence recall responses.

We infected mice i.n. with 50 TCID50 Δ4Sb or Δ4Cb virus and challenged at 28 d.p.i. i.p. with 2000 HAU of PR8. We collected sera 7 d post challenge and tested by ELISA for binding to PR8, Δ4 and S12 HAs. The data are the same as Fig. 7c, but shown is the serum response to all antigenic sites 28 d after infection and 7 d after i.p. challenge. Arrows indicate the site corresponding to the primary virus. Data on graph represent mean and SEM (bars) of two independent experiments with 3 mice each.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Tables 1–3 (PDF 1616 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Angeletti, D., Gibbs, J., Angel, M. et al. Defining B cell immunodominance to viruses. Nat Immunol 18, 456–463 (2017). https://doi.org/10.1038/ni.3680

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3680

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