Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses

Denis Gaucher, René Therrien, Nadia Kettaf, Bastian R Angermann, Geneviève Boucher, Abdelali Filali-Mouhim, Janice M Moser, Riyaz S Mehta, Donald R Drake 3rd, Erika Castro, Rama Akondy, Aline Rinfret, Bader Yassine-Diab, Elias A Said, Younes Chouikh, Mark J Cameron, Robert Clum, David Kelvin, Roland Somogyi, Larry D Greller, Robert S Balderas, Peter Wilkinson, Giuseppe Pantaleo, Jim Tartaglia, Elias K Haddad, Rafick-Pierre Sékaly, Denis Gaucher, René Therrien, Nadia Kettaf, Bastian R Angermann, Geneviève Boucher, Abdelali Filali-Mouhim, Janice M Moser, Riyaz S Mehta, Donald R Drake 3rd, Erika Castro, Rama Akondy, Aline Rinfret, Bader Yassine-Diab, Elias A Said, Younes Chouikh, Mark J Cameron, Robert Clum, David Kelvin, Roland Somogyi, Larry D Greller, Robert S Balderas, Peter Wilkinson, Giuseppe Pantaleo, Jim Tartaglia, Elias K Haddad, Rafick-Pierre Sékaly

Abstract

Correlates of immune-mediated protection to most viral and cancer vaccines are still unknown. This impedes the development of novel vaccines to incurable diseases such as HIV and cancer. In this study, we have used functional genomics and polychromatic flow cytometry to define the signature of the immune response to the yellow fever (YF) vaccine 17D (YF17D) in a cohort of 40 volunteers followed for up to 1 yr after vaccination. We show that immunization with YF17D leads to an integrated immune response that includes several effector arms of innate immunity, including complement, the inflammasome, and interferons, as well as adaptive immunity as shown by an early T cell response followed by a brisk and variable B cell response. Development of these responses is preceded, as demonstrated in three independent vaccination trials and in a novel in vitro system of primary immune responses (modular immune in vitro construct [MIMIC] system), by the coordinated up-regulation of transcripts for specific transcription factors, including STAT1, IRF7, and ETS2, which are upstream of the different effector arms of the immune response. These results clearly show that the immune response to a strong vaccine is preceded by coordinated induction of master transcription factors that lead to the development of a broad, polyfunctional, and persistent immune response that integrates all effector cells of the immune system.

Figures

Figure 1.
Figure 1.
Vaccination with YF17D induces early gene modulation. Global view of gene modulation in total blood cells after YF17D vaccination, as analyzed by gene array on the Montreal cohort (n = 9–15, depending on the time point). Heat map representation (a) and PCA (b) using the significantly modulated genes, in at least one comparison versus day 0.
Figure 2.
Figure 2.
Transcriptional network of differentially expressed genes after YF17D vaccination, as inferred by gene set enrichment. Network representation of inferred transcription factors (11) and predicted target genes that are significantly modulated. Node colors indicate fold change of gene expression between day 0 and 7 in n = 11 volunteers. Rectangular nodes indicate transcription factors identified by gene enrichment; the different shapes indicate genes in the different functional categories of Fig. 3 (a–e). Genes that were subsequently evaluated by RT-PCR are identified by an asterisk. IRF1, IRF8, and genes targeted by IRF1 or IRF8, but no other transcription factors in the figure were removed to increase readability (see Fig. S1 for a complete map). Fig. S1 is available at http://www.jem.org/cgi/content/full/jem.20082292/DC1.
Figure 3.
Figure 3.
YF17D vaccination stimulates multiple arms of the innate and adaptive immunity. (a–e) Heat map representation of significantly modulated genes, P values listed with each gene indicate the largest (least significant) P value among the significant fold changes. n = 9–15, depending on the time point. The listed genes fall under different functional categories: IFN-induced and TLR-associated genes (a); complement-associated genes (b); macrophage-associated genes (c); NK cell–associated genes (d); and B cell–associated genes (e). Supplemental document 2 includes a justification for the attribution of each gene into its respective group. Supplemental document 2 is available at http://www.jem.org/cgi/content/full/jem.20082292/DC1.
Figure 4.
Figure 4.
Heat map of fold change gene expression between day 0 and 7, as measured by qPCR. For a complete heat map of all measured fold changes see Fig. S3. Fig. S3 is available at http://www.jem.org/cgi/content/full/jem.20082292/DC1.
Figure 5.
Figure 5.
YF17D induces expression of genes associated with IL-1β and activates the inflammasome. (a) Heat maps showing the up-regulation of genes encoding inflammasome components and the modulation of other, IL-1β–associated genes. For IL-1R2 and -1RN only probes targeting all transcripts were considered. (b) IL-1β production by monocyte-derived DCs incubated with live (YF), UV-inactivated (UV), or heat-inactivated (HI) YF17D, as determined by ELISA. NI, noninfected cells. This represents the results from one representative of three separate experiments. The results for the two other experiments are shown in Fig. S4. Fig. S4 is available at http://www.jem.org/cgi/content/full/jem.20082292/DC1.
Figure 6.
Figure 6.
YF17D induces a mixed Th1/Th2 response. (a) Day 60 PBMCs were stimulated with the 22 peptide pools and assayed by CFSE labeling for their proliferative response. Bar graphs show data for six selected volunteers, and the dataset for all the volunteers can be found in Fig. S6 (c and d). At 24 h of culture, supernatants were analyzed by CBA to determine the Th1/Th2 cytokine secretion profile in response to each pool. The heat maps represent the data for the same six volunteers. The Th1/Th2 profiles determined this way for all the volunteers and pools are shown in Fig. S6 e. Fig. S6 is available at http://www.jem.org/cgi/content/full/jem.20082292/DC1.
Figure 7.
Figure 7.
YF17D vaccination induces specific CD4+ T cells of mixed T helper phenotype. (a) PBMCs from YF17D-vaccinated volunteers (day 28 after vaccination) were stimulated with a group of three immunostimulatory YF17D-derived peptides pools, and then stained with antibodies against CD4 (surface) and CD154, IL-2, and IFN-g (intracellular). The numbers indicate the percentages of cells within the parent population. (b) FACS analysis of PBMCs from day 365 after vaccination. Cells from four volunteers were stimulated (S) for with immunostimulatory YF17D peptide pools, and then restimulated (RS) or not with the same peptide pools before staining. The data were analyzed according to the gating strategy shown in Fig. S7, and are expressed as the percentage of central memory CD4+ T cells (CD45RA2CCR7+) that express the marker for recent activation CD154, and that are either Th1 or Th2 in the RS samples, over background (S). Fig. S7 is available at http://www.jem.org/cgi/content/full/jem.20082292/DC1.
Figure 8.
Figure 8.
YF17D elicits robust and diverse primary T helper cell responses in DC/T cell co-cultures. (a and b) Purified T helper cells were mixed with DCs that had been incubated with live or UV-inactivated YF17D. After 14 d, the cells were harvested and evaluated by flow cytometry for cytokine and CD154 expression after a short restimulation with autologous DCs targets that had been left untouched, pulsed with killed YF17D, or infected with live YF17D. All data plots show live CD4+-gated events. These are representative data of experiments done on cells from at least 10 volunteers. (c) Alternatively, the culture supernatants were harvested and evaluated by a multiplex cytokine analysis. Data representative of experiments done on cells from two volunteers.
Figure 9.
Figure 9.
Consensus transcriptional network of genes differentially expressed on day 7 as compared with day 0. Network representation of inferred transcription factors and predicted target genes that are consistently modulated in at least two out of three datasets, with the third dataset not being contradictory.

References

    1. Sekaly, R.P. 2008. The failed HIV Merck vaccine study: a step back or a launching point for future vaccine development? J. Exp. Med. 205:7–12.
    1. Plotkin, S., and W. Orenstein. 2004. Vaccines. WB Saunders, Philadelphia.
    1. McMichael, A.J. 2006. HIV vaccines. Annu. Rev. Immunol. 24:227–255.
    1. Monath, T.P. 2004. Yellow fever vaccine. In Vaccines. S. Plotkin and W. Orenstein, editors. WB Saunders, Philadelphia. 1095–1176.
    1. Bauer, J.H. 1931. The duration of passive immunity in yellow fever. Am. J. Trop. Med. Hyg. 11:451.
    1. Brandriss, M.W., J.J. Schlesinger, E.E. Walsh, and M. Briselli. 1986. Lethal 17D yellow fever encephalitis in mice. I. Passive protection by monoclonal antibodies to the envelope proteins of 17D yellow fever and dengue 2 viruses. J. Gen. Virol. 67(Pt 2):229–234.
    1. Reinhardt, B., R. Jaspert, M. Niedrig, C. Kostner, and J. Lapos;Age-Stehr. 1998. Development of viremia and humoral and cellular parameters of immune activation after vaccination with yellow fever virus strain 17D: a model of human flavivirus infection. J. Med. Virol. 56:159–167.
    1. Santos, A.P., A.L. Bertho, D.C. Dias, J.R. Santos, and R. Marcovistz. 2005. Lymphocyte subset analyses in healthy adults vaccinated with yellow fever 17DD virus. Mem. Inst. Oswaldo Cruz. 100:331–337.
    1. Co, M.D., M. Terajima, J. Cruz, F.A. Ennis, and A.L. Rothman. 2002. Human cytotoxic T lymphocyte responses to live attenuated 17D yellow fever vaccine: identification of HLA-B35-restricted CTL epitopes on nonstructural proteins NS1, NS2b, NS3, and the structural protein E. Virology. 293:151–163.
    1. Miller, J.D., R.G. van der Most, R.S. Akondy, J.T. Glidewell, S. Albott, D. Masopust, K. Murali-Krishna, P.L. Mahar, S. Edupuganti, S. Lalor, et al. 2008. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity. 28:710–722.
    1. Xie, X., J. Lu, E.J. Kulbokas, T.R. Golub, V. Mootha, K. Lindblad-Toh, E.S. Lander, and M. Kellis. 2005. Systematic discovery of regulatory motifs in human promoters and 3′ UTRs by comparison of several mammals. Nature. 434:338–345.
    1. Monaco, J.J., S. Cho, and M. Attaya. 1990. Transport protein genes in the murine MHC: possible implications for antigen processing. Science. 250:1723–1726.
    1. Trowsdale, J., I. Hanson, I. Mockridge, S. Beck, A. Townsend, and A. Kelly. 1990. Sequences encoded in the class II region of the MHC related to the ‘ABC’ superfamily of transporters. Nature. 348:741–744.
    1. Nicholson, S.E., and D.J. Hilton. 1998. The SOCS proteins: a new family of negative regulators of signal transduction. J. Leukoc. Biol. 63:665–668.
    1. Scherer, C.A., C.L. Magness, K.V. Steiger, N.D. Poitinger, C.M. Caputo, D.G. Miner, P.L. Winokur, D. Klinzman, J. McKee, C. Pilar, et al. 2007. Distinct gene expression profiles in peripheral blood mononuclear cells from patients infected with vaccinia virus, yellow fever 17D virus, or upper respiratory infections. Vaccine. 25:6458–6473.
    1. Martinon, F., K. Burns, and J. Tschopp. 2002. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol. Cell. 10:417–426.
    1. Chattopadhyay, P.K., J. Yu, and M. Roederer. 2005. A live-cell assay to detect antigen-specific CD4+ T cells with diverse cytokine profiles. Nat. Med. 11:1113–1117.
    1. Frentsch, M., O. Arbach, D. Kirchhoff, B. Moewes, M. Worm, M. Rothe, A. Scheffold, and A. Thiel. 2005. Direct access to CD4+ T cells specific for defined antigens according to CD154 expression. Nat. Med. 11:1118–1124.
    1. Rivino, L., M. Messi, D. Jarrossay, A. Lanzavecchia, F. Sallusto, and J. Geginat. 2004. Chemokine receptor expression identifies Pre-T helper (Th)1, Pre-Th2, and nonpolarized cells among human CD4+ central memory T cells. J. Exp. Med. 200:725–735.
    1. Poland, J.D., C.H. Calisher, T.P. Monath, W.G. Downs, and K. Murphy. 1981. Persistence of neutralizing antibody 30-35 years after immunization with 17D yellow fever vaccine. Bull. World Health Organ. 59:895–900.
    1. Colina, R., M. Costa-Mattioli, R.J. Dowling, M. Jaramillo, L.H. Tai, C.J. Breitbach, Y. Martineau, O. Larsson, L. Rong, Y.V. Svitkin, et al. 2008. Translational control of the innate immune response through IRF-7. Nature. 452:323–328.
    1. Honda, K., H. Yanai, H. Negishi, M. Asagiri, M. Sato, T. Mizutani, N. Shimada, Y. Ohba, A. Takaoka, N. Yoshida, and T. Taniguchi. 2005. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature. 434:772–777.
    1. Querec, T., S. Bennouna, S. Alkan, Y. Laouar, K. Gorden, R. Flavell, S. Akira, R. Ahmed, and B. Pulendran. 2006. 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.
    1. Kawai, T., and S. Akira. 2006. TLR signaling. Cell Death Differ. 13:816–825.
    1. Gallant, S., and G. Gilkeson. 2006. ETS transcription factors and regulation of immunity. Arch. Immunol. Ther. Exp. (Warsz.). 54:149–163.
    1. Bhat, N.K., C.B. Thompson, T. Lindsten, C.H. June, S. Fujiwara, S. Koizumi, R.J. Fisher, and T.S. Papas. 1990. Reciprocal expression of human ETS1 and ETS2 genes during T-cell activation: regulatory role for the protooncogene ETS1. Proc. Natl. Acad. Sci. USA. 87:3723–3727.
    1. Sun, H.J., X. Xu, X.L. Wang, L. Wei, F. Li, J. Lu, and B.Q. Huang. 2006. Transcription factors Ets2 and Sp1 act synergistically with histone acetyltransferase p300 in activating human interleukin-12 p40 promoter. Acta Biochim. Biophys. Sin. (Shanghai). 38:194–200.
    1. Blumenthal, S.G., G. Aichele, T. Wirth, A.P. Czernilofsky, A. Nordheim, and J. Dittmer. 1999. Regulation of the human interleukin-5 promoter by Ets transcription factors. Ets1 and Ets2, but not Elf-1, cooperate with GATA3 and HTLV-I Tax1. J. Biol. Chem. 274:12910–12916.
    1. Koizumi, H., M.F. Horta, B.S. Youn, K.C. Fu, B.S. Kwon, J.D. Young, and C.C. Liu. 1993. Identification of a killer cell-specific regulatory element of the mouse perforin gene: an Ets-binding site-homologous motif that interacts with Ets-related proteins. Mol. Cell. Biol. 13:6690–6701.
    1. Harada, H., T. Taniguchi, and N. Tanaka. 1998. The role of interferon regulatory factors in the interferon system and cell growth control. Biochimie. 80:641–650.
    1. Masumi, A., S. Tamaoki, I.M. Wang, K. Ozato, and K. Komuro. 2002. IRF-8/ICSBP and IRF-1 cooperatively stimulate mouse IL-12 promoter activity in macrophages. FEBS Lett. 531:348–353.
    1. Wang, I.M., C. Contursi, A. Masumi, X. Ma, G. Trinchieri, and K. Ozato. 2000. An IFN-gamma-inducible transcription factor, IFN consensus sequence binding protein (ICSBP), stimulates IL-12 p40 expression in macrophages. J. Immunol. 165:271–279.
    1. Liu, J., X. Guan, T. Tamura, K. Ozato, and X. Ma. 2004. Synergistic activation of interleukin-12 p35 gene transcription by interferon regulatory factor-1 and interferon consensus sequence-binding protein. J. Biol. Chem. 279:55609–55617.
    1. Tsujimura, H., T. Tamura, H.J. Kong, A. Nishiyama, K.J. Ishii, D.M. Klinman, and K. Ozato. 2004. Toll-like receptor 9 signaling activates NF-kappaB through IFN regulatory factor-8/IFN consensus sequence binding protein in dendritic cells. J. Immunol. 172:6820–6827.
    1. Lee, C.H., M. Melchers, H. Wang, T.A. Torrey, R. Slota, C.F. Qi, J.Y. Kim, P. Lugar, H.J. Kong, L. Farrington, et al. 2006. Regulation of the germinal center gene program by interferon (IFN) regulatory factor 8/IFN consensus sequence-binding protein. J. Exp. Med. 203:63–72.
    1. Natkunam, Y., S. Zhao, D.Y. Mason, J. Chen, B. Taidi, M. Jones, A.S. Hammer, S. Hamilton Dutoit, I.S. Lossos, and R. Levy. 2007. The oncoprotein LMO2 is expressed in normal germinal-center B cells and in human B-cell lymphomas. Blood. 109:1636–1642.
    1. Hansson, A., C. Manetopoulos, J.I. Jonsson, and H. Axelson. 2003. The basic helix-loop-helix transcription factor TAL1/SCL inhibits the expression of the p16INK4A and pTalpha genes. Biochem. Biophys. Res. Commun. 312:1073–1081.
    1. Sardet, C., M. Vidal, D. Cobrinik, Y. Geng, C. Onufryk, A. Chen, and R.A. Weinberg. 1995. E2F-4 and E2F-5, two members of the E2F family, are expressed in the early phases of the cell cycle. Proc. Natl. Acad. Sci. USA. 92:2403–2407.
    1. van Grevenynghe, J., F.A. Procopio, Z. He, N. Chomont, C. Riou, Y. Zhang, S. Gimmig, G. Boucher, P. Wilkinson, Y. Shi, et al. 2008. Transcription factor FOXO3a controls the persistence of memory CD4(+) T cells during HIV infection. Nat. Med. 14:266–274.
    1. Furukawa, Y., N. Nishimura, M. Satoh, H. Endo, S. Iwase, H. Yamada, M. Matsuda, Y. Kano, and M. Nakamura. 2002. Apaf-1 is a mediator of E2F-1-induced apoptosis. J. Biol. Chem. 277:39760–39768.
    1. Petrilli, V., C. Dostert, D.A. Muruve, and J. Tschopp. 2007. The inflammasome: a danger sensing complex triggering innate immunity. Curr. Opin. Immunol. 19:615–622.
    1. Muruve, D.A., V. Petrilli, A.K. Zaiss, L.R. White, S.A. Clark, P.J. Ross, R.J. Parks, and J. Tschopp. 2008. The inflammasome recognizes cytosolic microbial and host DNA and triggers an innate immune response. Nature. 452:103–108.
    1. Li, H., S.B. Willingham, J.P. Ting, and F. Re. 2008. Cutting edge: inflammasome activation by alum and alum's adjuvant effect are mediated by NLRP3. J. Immunol. 181:17–21.
    1. Martinon, F., and J. Tschopp. 2004. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell. 117:561–574.
    1. Mantovani, A., M. Locati, A. Vecchi, S. Sozzani, and P. Allavena. 2001. Decoy receptors: a strategy to regulate inflammatory cytokines and chemokines. Trends Immunol. 22:328–336.
    1. Wan, L., C.W. Lin, Y.J. Lin, J.J. Sheu, B.H. Chen, C.C. Liao, Y. Tsai, W.Y. Lin, C.H. Lai, and F.J. Tsai. 2008. Type I IFN induced IL1-Ra expression in hepatocytes is mediated by activating STAT6 through the formation of STAT2: STAT6 heterodimer. J. Cell. Mol. Med. 12:876–878.
    1. Gabay, C., M. Dreyer, N. Pellegrinelli, R. Chicheportiche, and C.A. Meier. 2001. Leptin directly induces the secretion of interleukin 1 receptor antagonist in human monocytes. J. Clin. Endocrinol. Metab. 86:783–791.
    1. Stanley, S.L. Jr., S.E. Frey, P. Taillon-Miller, J. Guo, R.D. Miller, D.C. Koboldt, M. Elashoff, R. Christensen, N.L. Saccone, and R.B. Belshe. 2007. The immunogenetics of smallpox vaccination. J. Infect. Dis. 196:212–219.
    1. Csomor, E., Z. Bajtay, N. Sandor, K. Kristof, G.J. Arlaud, S. Thiel, and A. Erdei. 2007. Complement protein C1q induces maturation of human dendritic cells. Mol. Immunol. 44:3389–3397.
    1. Mehlhop, E., and M.S. Diamond. 2006. Protective immune responses against West Nile virus are primed by distinct complement activation pathways. J. Exp. Med. 203:1371–1381.
    1. Mehlhop, E., K. Whitby, T. Oliphant, A. Marri, M. Engle, and M.S. Diamond. 2005. Complement activation is required for induction of a protective antibody response against West Nile virus infection. J. Virol. 79:7466–7477.
    1. Santos, A.P., D.C. Matos, A.L. Bertho, S.C. Mendonca, and R. Marcovistz. 2008. Detection of Th1/Th2 cytokine signatures in yellow fever 17DD first-time vaccinees through ELISpot assay. Cytokine. 42:152–155.
    1. Brunner, C., A. Sindrilaru, I. Girkontaite, K.D. Fischer, C. Sunderkotter, and T. Wirth. 2007. BOB.1/OBF.1 controls the balance of TH1 and TH2 immune responses. EMBO J. 26:3191–3202.
    1. Wang, Y., and R.H. Carter. 2005. CD19 regulates B cell maturation, proliferation, and positive selection in the FDC zone of murine splenic germinal centers. Immunity. 22:749–761.
    1. Steinman, R.M., and J. Banchereau. 2007. Taking dendritic cells into medicine. Nature. 449:419–426.
    1. Redecke, V., H. Hacker, S.K. Datta, A. Fermin, P.M. Pitha, D.H. Broide, and E. Raz. 2004. Cutting edge: activation of Toll-like receptor 2 induces a Th2 immune response and promotes experimental asthma. J. Immunol. 172:2739–2743.
    1. Gorden, K.B., K.S. Gorski, S.J. Gibson, R.M. Kedl, W.C. Kieper, X. Qiu, M.A. Tomai, S.S. Alkan, and J.P. Vasilakos. 2005. Synthetic TLR agonists reveal functional differences between human TLR7 and TLR8. J. Immunol. 174:1259–1268.
    1. Van Gelder, R.N., M.E. von Zastrow, A. Yool, W.C. Dement, J.D. Barchas, and J.H. Eberwine. 1990. Amplified RNA synthesized from limited quantities of heterogeneous cDNA. Proc. Natl. Acad. Sci. USA. 87:1663–1667.
    1. Gentleman, R.C., V.J. Carey, D.M. Bates, B. Bolstad, M. Dettling, S. Dudoit, B. Ellis, L. Gautier, Y. Ge, J. Gentry, et al. 2004. Bioconductor: open software development for computational biology and bioinformatics. Genome Biol. 5:R80.
    1. Smyth, G.K. 2004. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3.
    1. Smith, G.K. 2005. Limma: linear models for microarray data. In Bioinformatics and Computational Biology Solutions using R and Bioconductor. W. Huber, editor. Springer, New York. 397-420.
    1. Baron, C., R. Somogyi, L.D. Greller, V. Rineau, P. Wilkinson, C.R. Cho, M.J. Cameron, D.J. Kelvin, P. Chagnon, D.C. Roy, et al. 2007. Prediction of graft-versus-host disease in humans by donor gene-expression profiling. PLoS Med. 4:e23.
    1. Teschendorff, A.E., M. Journee, P.A. Absil, R. Sepulchre, and C. Caldas. 2007. Elucidating the altered transcriptional programs in breast cancer using independent component analysis. PLoS Comput Biol. 3:e161.
    1. Younes, S.A., B. Yassine-Diab, A.R. Dumont, M.R. Boulassel, Z. Grossman, J.P. Routy, and R.P. Sekaly. 2003. HIV-1 viremia prevents the establishment of interleukin 2-producing HIV-specific memory CD4+ T cells endowed with proliferative capacity. J. Exp. Med. 198:1909–1922.

Source: PubMed

3
구독하다