Abstract
Many successful vaccines induce persistent antibody responses that can last a lifetime. The mechanisms by which they do so remain unclear, but emerging evidence indicates that they activate dendritic cells via Toll-like receptors (TLRs)1,2. For example, the yellow fever vaccine YF-17D, one of the most successful empiric vaccines ever developed3, activates dendritic cells via multiple TLRs to stimulate proinflammatory cytokines4,5. Triggering specific combinations of TLRs in dendritic cells can induce synergistic production of cytokines6, which results in enhanced T-cell responses, but its impact on antibody responses remain unknown. Learning the critical parameters of innate immunity that program such antibody responses remains a major challenge in vaccinology. Here we demonstrate that immunization of mice with synthetic nanoparticles containing antigens plus ligands that signal through TLR4 and TLR7 induces synergistic increases in antigen-specific, neutralizing antibodies compared to immunization with nanoparticles containing antigens plus a single TLR ligand. Consistent with this there was enhanced persistence of germinal centres and of plasma-cell responses, which persisted in the lymph nodes for >1.5 years. Surprisingly, there was no enhancement of the early short-lived plasma-cell response relative to that observed with single TLR ligands. Molecular profiling of activated B cells, isolated 7 days after immunization, indicated that there was early programming towards B-cell memory. Antibody responses were dependent on direct triggering of both TLRs on B cells and dendritic cells, as well as on T-cell help. Immunization protected completely against lethal avian and swine influenza virus strains in mice, and induced robust immunity against pandemic H1N1 influenza in rhesus macaques.
This is a preview of subscription content, access via your institution
Relevant articles
Open Access articles citing this article.
-
A TLR7-nanoparticle adjuvant promotes a broad immune response against heterologous strains of influenza and SARS-CoV-2
Nature Materials Open Access 30 January 2023
-
Emerging role of microbiota derived outer membrane vesicles to preventive, therapeutic and diagnostic proposes
Infectious Agents and Cancer Open Access 19 January 2023
-
Ancestral reconstruction reveals catalytic inactivation of activation-induced cytidine deaminase concomitant with cold water adaption in the Gadiformes bony fish
BMC Biology Open Access 27 December 2022
Access options
Subscribe to this journal
Receive 51 print issues and online access
$199.00 per year
only $3.90 per issue
Rent or buy this article
Get just this article for as long as you need it
$39.95
Prices may be subject to local taxes which are calculated during checkout




References
Pulendran, B. & Ahmed, R. Translating innate immunity into immunological memory: implications for vaccine development. Cell 124, 849–863 (2006)
Kawai, T. & Akira, S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nature Immunol. 11, 373–384 (2010)
Pulendran, B. Learning immunology from the yellow fever vaccine: innate immunity to systems vaccinology. Nature Rev. Immunol. 9, 741–747 (2009)
Querec, T. et al. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J. Exp. Med. 203, 413–424 (2006)
Querec, T. D. et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nature Immunol. 10, 116–125 (2009)
Napolitani, G., Rinaldi, A., Bertoni, F., Sallusto, F. & Lanzavecchia, A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nature Immunol. 6, 769–776 (2005)
Peek, L. J., Middaugh, C. R. & Berkland, C. Nanotechnology in vaccine delivery. Adv. Drug Deliv. Rev. 60, 915–928 (2008)
Kazzaz, J. et al. Encapsulation of the immune potentiators MPL and RC529 in PLG microparticles enhances their potency. J. Control. Release 110, 566–573 (2006)
Singh, M., Chakrapani, A. & O’Hagan, D. Nanoparticles and microparticles as vaccine-delivery systems. Expert Rev. Vaccines 6, 797–808 (2007)
Young, J. A. & Collier, J. R. Attacking anthrax. Sci. Am. 286, 48–59 (2002)
Chen, G. L. & Subbarao, K. Live attenuated vaccines for pandemic influenza. Curr. Top. Microbiol. Immunol. 333, 109–132 (2009)
Hangartner, L., Zinkernagel, R. M. & Hengartner, H. Antiviral antibody responses: the two extremes of a wide spectrum. Nature Rev. Immunol. 6, 231–243 (2006)
Jung, S. et al. In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity 17, 211–220 (2002)
Kissenpfennig, A. et al. Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells. Immunity 22, 643–654 (2005)
Mata-Haro, V. et al. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4. Science 316, 1628–1632 (2007)
Bernasconi, N. L., Traggiai, E. & Lanzavecchia, A. Maintenance of serological memory by polyclonal activation of human memory B cells. Science 298, 2199–2202 (2002)
Pasare, C. & Medzhitov, R. Control of B-cell responses by Toll-like receptors. Nature 438, 364–368 (2005)
Mitchison, N. A. T-cell–B-cell cooperation. Nature Rev. Immunol. 4, 308–312 (2004)
van Essen, D., Dullforce, P., Brocker, T. & Gray, D. Cellular interactions involved in Th cell memory. J. Immunol. 165, 3640–3646 (2000)
McHeyzer-Williams, L. J. & McHeyzer-Williams, M. G. Antigen-specific memory B cell development. Annu. Rev. Immunol. 23, 487–513 (2005)
Slifka, M. K., Antia, R., Whitmire, J. K. & Ahmed, R. Humoral immunity due to long-lived plasma cells. Immunity 8, 363–372 (1998)
McHeyzer-Williams, M. G., McLean, M. J., Lalor, P. A. & Nossal, G. J. Antigen-driven B cell differentiation in vivo. J. Exp. Med. 178, 295–307 (1993)
Luckey, C. J. et al. Memory T and memory B cells share a transcriptional program of self-renewal with long-term hematopoietic stem cells. Proc. Natl Acad. Sci. USA 103, 3304–3309 (2006)
Wille-Reece, U. et al. HIV Gag protein conjugated to a Toll-like receptor 7/8 agonist improves the magnitude and quality of Th1 and CD8+ T cell responses in nonhuman primates. Proc. Natl Acad. Sci. USA 102, 15190–15194 (2005)
Steel, J. et al. Live attenuated influenza viruses containing NS1 truncations as vaccine candidates against H5N1 highly pathogenic avian influenza. J. Virol. 83, 1742–1753 (2009)
Jurk, M. et al. Human TLR7 or TLR8 independently confer responsiveness to the antiviral compound R-848. Nature Immunol. 3, 499 (2002)
Dorner, M. et al. Plasma cell toll-like receptor (TLR) expression differs from that of B cells, and plasma cell TLR triggering enhances immunoglobulin production. Immunology 128, 573–579 (2009)
Clark, T. W. et al. Trial of 2009 influenza A (H1N1) monovalent MF59-adjuvanted vaccine. N. Engl. J. Med. 361, 2424–2435 (2009)
Potter, C. W. & Oxford, J. S. Determinants of immunity to influenza infection in man. Br. Med. Bull. 35, 69–75 (1979)
Bachmann, M. F., Odermatt, B., Hengartner, H. & Zinkernagel, R. M. Induction of long-lived germinal centers associated with persisting antigen after viral infection. J. Exp. Med. 183, 2259–2269 (1996)
Sah, H. A new strategy to determine the actual protein content of poly(lactide-co-glycolide) microspheres. J. Pharm. Sci. 86, 1315–1318 (1997)
Henri, S. et al. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J. Exp. Med. 207, 189–206 (2010)
den Haan, J. M., Kraal, G. & Bevan, M. J. Cutting edge: lipopolysaccharide induces IL-10-producing regulatory CD4+ T cells that suppress the CD8+ T cell response. J. Immunol. 178, 5429–5433 (2007)
Zhu, Q. et al. Immunization by vaccine-coated microneedle arrays protects against lethal influenza virus challenge. Proc. Natl Acad. Sci. USA 106, 7968–7973 (2009)
Staats, H. F. et al. In vitro and in vivo characterization of anthrax anti-protective antigen and anti-lethal factor monoclonal antibodies after passive transfer in a mouse lethal toxin challenge model to define correlates of immunity. Infect. Immun. 75, 5443–5452 (2007)
Compans, R. W. Hemagglutination-inhibition: rapid assay for neuraminic acid-containing viruses. J. Virol. 14, 1307–1309 (1974)
Enioutina, E. Y., Visic, D. & Daynes, R. A. The induction of systemic and mucosal immune responses to antigen-adjuvant compositions administered into the skin: alterations in the migratory properties of dendritic cells appears to be important for stimulating mucosal immunity. Vaccine 18, 2753–2767 (2000)
Reed, L. J. & Muench, H. A simple method of estimating fifty percent endpoints. Am. J. Hyg. 27, 493–497 (1938)
Badr, G. et al. Type I interferon (IFN-α/β) rescues B-lymphocytes from apoptosis via PI3Kδ/Akt, Rho-A, NFκB and Bcl-2/Bcl(XL). Cell. Immunol. 263, 31–40 (2010)
Bekeredjian-Ding, I. B. et al. Plasmacytoid dendritic cells control TLR7 sensitivity of naive B cells via type I IFN. J. Immunol. 174, 4043–4050 (2005)
Thibault, D. L. et al. IRF9 and STAT1 are required for IgG autoantibody production and B cell expression of TLR7 in mice. J. Clin. Invest. 118, 1417–1426 (2008)
Tovey, M. G., Lallemand, C. & Thyphronitis, G. Adjuvant activity of type I interferons. Biol. Chem. 389, 541–545 (2008)
Swanson, C. L. et al. Type I IFN enhances follicular B cell contribution to the T cell-independent antibody response. J. Exp. Med. 207, 1485–1500 (2010)
Liu, H. et al. Functional studies of BCL11A: characterization of the conserved BCL11A-XL splice variant and its interaction with BCL6 in nuclear paraspeckles of germinal center B cells. Mol. Cancer 5, 18 (2006)
Smith, K. G. et al. bcl-2 transgene expression inhibits apoptosis in the germinal center and reveals differences in the selection of memory B cells and bone marrow antibody-forming cells. J. Exp. Med. 191, 475–484 (2000)
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)
Zhou, G. & Ono, S. J. Induction of BCL-6 gene expression by interferon-γ and identification of an IRE in exon I. Exp. Mol. Pathol. 78, 25–35 (2005)
Mitsdoerffer, M. et al. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc. Natl Acad. Sci. USA 107, 14292–14297 (2010)
Chin, A. I. et al. TANK potentiates tumor necrosis factor receptor-associated factor-mediated c-Jun N-terminal kinase/stress-activated protein kinase activation through the germinal center kinase pathway. Mol. Cell. Biol. 19, 6665–6672 (1999)
Basso, K. & Dalla-Favera, R. BCL6: master regulator of the germinal center reaction and key oncogene in B cell lymphomagenesis. Adv. Immunol. 105, 193–210 (2010)
Kano, G. et al. Ikaros dominant negative isoform (Ik6) induces IL-3-independent survival of murine pro-B lymphocytes by activating JAK-STAT and up-regulating Bcl-xl levels. Leuk. Lymphoma 49, 965–973 (2008)
Ke, N., Godzik, A. & Reed, J. C. Bcl-B, a novel Bcl-2 family member that differentially binds and regulates Bax and Bak. J. Biol. Chem. 276, 12481–12484 (2001)
Airoldi, I. et al. Expression and function of IL-12 and IL-18 receptors on human tonsillar B cells. J. Immunol. 165, 6880–6888 (2000)
Airoldi, I. et al. Heterogeneous expression of interleukin-18 and its receptor in B-cell lymphoproliferative disorders deriving from naive, germinal center, and memory B lymphocytes. Clin. Cancer Res. 10, 144–154 (2004)
Hikida, M. et al. PLC-γ2 is essential for formation and maintenance of memory B cells. J. Exp. Med. 206, 681–689 (2009)
Nera, K. P. & Lassila, O. Pax5—a critical inhibitor of plasma cell fate. Scand. J. Immunol. 64, 190–199 (2006)
Acknowledgements
We thank R. Ahmed and B. Rouse for discussion and comments on the manuscript. We thank B. Norris for assistance with FACS analysis, and H. Oluoch for assistance with cryostat sectioning. The work in the laboratory of B.P. was supported by grants U54AI057157, R37AI48638, R01DK057665, U19AI057266, HHSN266200700006C, NO1 AI50025 and U19AI090023 from the National Institutes of Health and a grant from the Bill & Melinda Gates Foundation. Work in A.G.-S. laboratories was partly funded by grants HHSN266200700010C, U54AI57158) and U01AI070469 from the National Institutes of Health.
Author information
Authors and Affiliations
Contributions
S.P.K. and B.P. designed the study, planned the experiments and analysed the data. B.P. and S.P.K. wrote the manuscript. S.P.K., I.S. and B.P. designed and performed the H1N1 vaccine studies in mice and primates. D.K. assisted with the H1N1 vaccine studies in mice and primates. R.A.A., A.G.-S. and J.S. designed and performed the neutralization assays and challenge experiments with H5N1 vaccine studies in mice. T.H. and R.R. assisted with experiments. H.I.N. performed the microarray analysis. S.S. and M.A. designed and carried out the SPR-based avidity experiments. M.K. assisted with design and execution of mice and non-human primate experiments. N.M. assisted with design of formulations. J.J. assisted with immunohistochemistry and design of experiments. R.J.H. expressed and purified the recombinant H5HA protein. R.C. helped plan and design the H1N1 vaccine study in mice and primates.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing financial interests.
Supplementary information
Supplementary Figures
The file contains Supplementary Figures 1-18 with legends. (PDF 3185 kb)
Rights and permissions
About this article
Cite this article
Kasturi, S., Skountzou, I., Albrecht, R. et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature 470, 543–547 (2011). https://doi.org/10.1038/nature09737
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/nature09737
This article is cited by
-
Emerging role of microbiota derived outer membrane vesicles to preventive, therapeutic and diagnostic proposes
Infectious Agents and Cancer (2023)
-
SREBP signaling is essential for effective B cell responses
Nature Immunology (2023)
-
A TLR7-nanoparticle adjuvant promotes a broad immune response against heterologous strains of influenza and SARS-CoV-2
Nature Materials (2023)
-
A nanoadjuvant that dynamically coordinates innate immune stimuli activation enhances cancer immunotherapy and reduces immune cell exhaustion
Nature Nanotechnology (2023)
-
Immunization against Zika by entrapping live virus in a subcutaneous self-adjuvanting hydrogel
Nature Biomedical Engineering (2023)
Comments
By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.