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.

  • Resource
  • Published:

Molecular signatures of antibody responses derived from a systems biology study of five human vaccines

Subjects

Abstract

Many vaccines induce protective immunity via antibodies. Systems biology approaches have been used to determine signatures that can be used to predict vaccine-induced immunity in humans, but whether there is a 'universal signature' that can be used to predict antibody responses to any vaccine is unknown. Here we did systems analyses of immune responses to the polysaccharide and conjugate vaccines against meningococcus in healthy adults, in the broader context of published studies of vaccines against yellow fever virus and influenza virus. To achieve this, we did a large-scale network integration of publicly available human blood transcriptomes and systems-scale databases in specific biological contexts and deduced a set of transcription modules in blood. Those modules revealed distinct transcriptional signatures of antibody responses to different classes of vaccines, which provided key insights into primary viral, protein recall and anti-polysaccharide responses. Our results elucidate the early transcriptional programs that orchestrate vaccine immunity in humans and demonstrate the power of integrative network modeling.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Purchase on Springer Link

Instant access to full article PDF

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Antibody responses of subjects who received vaccines against meningococcus.
Figure 2: Analysis of data on the blood transcriptome for five human vaccines.
Figure 3: Analysis of differences in gene expression for five vaccines.
Figure 4: BTMs provide a sensitive and robust statistical framework.
Figure 5: BTM analysis reveals distinct mechanisms of antibody response.
Figure 6: The polysaccharide-containing vaccine against meningococcus activates myeloid DCs.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Pulendran, B. Learning immunology from the yellow fever vaccine: innate immunity to systems vaccinology. Nat. Rev. Immunol. 9, 741–747 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Pulendran, B., Li, S. & Nakaya, H.I. Systems vaccinology. Immunity 33, 516–529 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Nakaya, H.I. & Pulendran, B. Systems vaccinology: its promise and challenge for HIV vaccine development. Curr. Opin. HIV AIDS 7, 24 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Querec, T.D. et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nat. Immunol. 10, 116–125 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Gaucher, D. et al. Yellow fever vaccine induces integrated multilineage and polyfunctional immune responses. J. Exp. Med. 205, 3119–3131 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Nakaya, H.I. et al. Systems biology of vaccination for seasonal influenza in humans. Nat. Immunol. 12, 786–795 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Vahey, M.T. et al. Expression of genes associated with immunoproteasome processing of major histocompatibility complex peptides is indicative of protection with adjuvanted RTS,S malaria vaccine. J. Infect. Dis. 201, 580–589 (2010).

    Article  CAS  PubMed  Google Scholar 

  8. Bucasas, K.L. et al. Early patterns of gene expression correlate with the humoral immune response to influenza vaccination in humans. J. Infect. Dis. 203, 921–929 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zak, D.E. et al. Merck Ad5/HIV induces broad innate immune activation that predicts CD8+ T-cell responses but is attenuated by preexisting Ad5 immunity. Proc. Natl. Acad. Sci. USA 109, E3503–E3512 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Obermoser, G. et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity 38, 831–844 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Furman, D. et al. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol. Syst. Biol. 9, 659 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Oberg, A.L., Kennedy, R.B., Li, P., Ovsyannikova, I.G. & Poland, G.A. Systems biology approaches to new vaccine development. Curr. Opin. Immunol. 23, 436–443 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Trautmann, L. & Sekaly, R. Solving vaccine mysteries: a systems biology perspective. Nat. Immunol. 12, 729 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Geeraedts, F. et al. Superior immunogenicity of inactivated whole virus H5N1 influenza vaccine is primarily controlled by Toll-like receptor signalling. PLoS Pathog. 4, e1000138 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Koyama, S. et al. Plasmacytoid dendritic cells delineate immunogenicity of influenza vaccine subtypes. Sci. Transl. Med. 2, 25ra24 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Rouphael, N.G. & Stephens, D.S. Neisseria meningitidis: biology, microbiology, and epidemiology. Methods Mol. Biol. 799, 1–20 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Goldschneider, I., Gotschlich, E.C. & Artenstein, M.S. Human immunity to the meningococcus. I. The role of humoral antibodies. J. Exp. Med. 129, 1307–1326 (1969).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Käyhty, H., Karanko, V., Peltola, H., Sarna, S. & Makela, P.H. Serum antibodies to capsular polysaccharide vaccine of group A Neissera meningitidis followed for three years in infants and children. J. Infect. Dis. 142, 861–868 (1980).

    Article  PubMed  Google Scholar 

  20. Jokhdar, H. et al. Immunologic hyporesponsiveness to serogroup C but not serogroup A following repeated meningococcal A/C polysaccharide vaccination in Saudi Arabia. Clin. Diagn. Lab. Immunol. 11, 83–88 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Findlow, H. et al. Immunoglobulin G subclass response to a meningococcal quadrivalent polysaccharide-diphtheria toxoid conjugate vaccine. Clin. Vaccine Immunol. 13, 507–510 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Borrow, R., Balmer, P. & Miller, E. Meningococcal surrogates of protection–serum bactericidal antibody activity. Vaccine 23, 2222–2227 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cerami, E., Demir, E., Schultz, N., Taylor, B.S. & Sander, C. Automated network analysis identifies core pathways in glioblastoma. PLoS ONE 5, e8918 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Nibbe, R.K., Koyuturk, M. & Chance, M.R. An integrative -omics approach to identify functional sub-networks in human colorectal cancer. PLOS Comput. Biol. 6, e1000639 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Khatri, P., Sirota, M. & Butte, A.J. Ten years of pathway analysis: current approaches and outstanding challenges. PLoS Comput. Biol. 8, e1002375 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, S., Nakaya, H.I., Kazmin, D.A., Oh, J.Z. & Pulendran, B. Systems biological approaches to measure and understand vaccine immunity in humans. Semin. Immunol. 25, 209–218 (2013).

    Article  CAS  PubMed  Google Scholar 

  28. Schaefer, C.F. et al. PID: the Pathway Interaction Database. Nucleic Acids Res. 37, D674–D679 (2009).

    Article  CAS  PubMed  Google Scholar 

  29. Lau, E. & Ronai, Z.A. ATF2-at the crossroad of nuclear and cytosolic functions. J. Cell Sci. 125, 2815–2824 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Chaveroux, C. et al. Identification of a novel amino acid response pathway triggering ATF2 phosphorylation in mammals. Mol. Cell Biol. 29, 6515–6526 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Ravindran, R. et al. Vaccine activation of the nutrient sensor GCN2 in dendritic cells enhances antigen presentation. Science (in the press).

  32. Haining, W.N. & Pulendran, B. Identifying gnostic predictors of the vaccine response. Curr. Opin. Immunol. 24, 332–336 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chaussabel, D., Pascual, V. & Banchereau, J. Assessing the human immune system through blood transcriptomics. BMC Biol. 8, 84 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Fraser, I.D.C. & Germain, R.N. Navigating the network: signaling cross-talk in hematopoietic cells. Nat. Immunol. 10, 327–331 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Friedman, N. Inferring cellular networks using probabilistic graphical models. Science 303, 799–805 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. Segal, E. et al. Module networks: identifying regulatory modules and their condition-specific regulators from gene expression data. Nat. Genet. 34, 166–176 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Basso, K. et al. Reverse engineering of regulatory networks in human B cells. Nat. Genet. 37, 382–390 (2005).

    Article  CAS  PubMed  Google Scholar 

  38. Lee, I. et al. A single gene network accurately predicts phenotypic effects of gene perturbation in Caenorhabditis elegans. Nat. Genet. 40, 181–188 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Chaussabel, D. et al. A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity 29, 150–164 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Margolin, A.A. et al. ARACNE: an algorithm for the reconstruction of gene regulatory networks in a mammalian cellular context. BMC Bioinformatics 7 (suppl. 1), S7 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Lefebvre, C. et al. A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers. Mol. Syst. Biol. 6, 377 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Lefebvre, C., Rieckhof, G. & Califano, A. Reverse-engineering human regulatory networks. Wiley Interdiscip. Rev. Syst. Biol. Med. 4, 311–325 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Sales, G. & Romualdi, C. Parmigene–a parallel R package for mutual information estimation and gene network reconstruction. Bioinformatics 27, 1876–1877 (2011).

    Article  CAS  PubMed  Google Scholar 

  44. Chuang, H.Y., Lee, E., Liu, Y.T., Lee, D. & Ideker, T. Network-based classification of breast cancer metastasis. Mol. Syst. Biol. 3, 140 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Fredlund, E., Ringner, M., Maris, J.M. & Pahlman, S. High Myc pathway activity and low stage of neuronal differentiation associate with poor outcome in neuroblastoma. Proc. Natl. Acad. Sci. USA 105, 14094–14099 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Vitour, D. et al. Polo-like kinase 1 (PLK1) regulates interferon (IFN) induction by MAVS. J. Biol. Chem. 284, 21797–21809 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Zhang, W. et al. The scaffold protein TANK/I-TRAF inhibits NF-κB activation by recruiting polo-like kinase 1. Mol. Biol. Cell 21, 2500–2513 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zughaier, S.M. Neisseria meningitidis capsular polysaccharides induce inflammatory responses via TLR2 and TLR4-MD-2. J. Leukoc. Biol. 89, 469–480 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Klein, S.L. & Poland, G.A. Personalized vaccinology: one size and dose might not fit both sexes. Vaccine 31, 2599–2600 (2013).

    Article  PubMed  Google Scholar 

  50. Nakaya, H.I., Li, S.Z. & Pulendran, B. Systems vaccinology: learning to compute the behavior of vaccine induced immunity. Wiley Interdiscip. Rev. Syst. Biol. Med. 4, 193–205 (2012).

    Article  CAS  PubMed  Google Scholar 

  51. Avci, F.Y., Li, X.M., Tsuji, M. & Kasper, D.L. A mechanism for glycoconjugate vaccine activation of the adaptive immune system and its implications for vaccine design. Nat. Med. 17, 1602–1609 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Holder, P.K. et al. Assignment of Neisseria meningitidis serogroup A and C class-specific anticapsular antibody concentrations to the new standard reference serum CDC1992. Clin. Diagn. Lab. Immunol. 2, 132–137 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gheesling, L.L. et al. Multicenter comparison of Neisseria meningitidis serogroup C anti-capsular polysaccharide antibody levels measured by a standardized enzyme-linked immunosorbent assay. J. Clin. Microbiol. 32, 1475–1482 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. van Gageldonk, P.G., van Schaijk, F.G., van der Klis, F.R. & Berbers, G.A. Development and validation of a multiplex immunoassay for the simultaneous determination of serum antibodies to Bordetella pertussis, diphtheria and tetanus. J. Immunol. Methods 335, 79–89 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Maslanka, S.E. et al. Standardization and a multilaboratory comparison of Neisseria meningitidis serogroup A and C serum bactericidal assays. The Multilaboratory Study Group. Clin. Diagn. Lab. Immunol. 4, 156–167 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Poolman, J.T. et al. Measurement of functional anti-meningococcal serogroup a activity using strain 3125 as the target strain for serum bactericidal assay. Clin. Vaccine Immunol. 18, 1108–1117 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Moreno, R.L. et al. A murine model for the study of immune memory in response to pneumococcal conjugate vaccination. Vaccine 22, 3069–3079 (2004).

    Article  CAS  PubMed  Google Scholar 

  59. Cerami, E.G. et al. Pathway Commons, a web resource for biological pathway data. Nucleic Acids Res. 39, D685–D690 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Keshava Prasad, T.S. et al. Human Protein Reference Database–2009 update. Nucleic Acids Res. 37, D767–D772 (2009).

    Article  CAS  PubMed  Google Scholar 

  61. Breitkreutz, B.J. et al. The BioGRID Interaction Database: 2008 update. Nucleic Acids Res. 36, D637–D640 (2008).

    Article  CAS  PubMed  Google Scholar 

  62. Aranda, B. et al. The IntAct molecular interaction database in 2010. Nucleic Acids Res. 38, D525–D531 (2010).

    Article  CAS  PubMed  Google Scholar 

  63. Ceol, A. et al. MINT, the molecular interaction database: 2009 update. Nucleic Acids Res. 38, D532–D539 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Matthews, L. et al. Reactome knowledgebase of human biological pathways and processes. Nucleic Acids Res. 37, D619–D622 (2009).

    Article  CAS  PubMed  Google Scholar 

  65. Romero, P. et al. Computational prediction of human metabolic pathways from the complete human genome. Genome Biol. 6, R2 (2005).

    Article  PubMed  Google Scholar 

  66. Huang, D.W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank B. Weaver and the Hope Clinic staff for assistance with the clinical study. Supported by the US National Institutes of Health (U19AI090023, U54AI057157, R37AI48638, R37DK057665, U19AI057266 and AI100663-02 to the B.P. laboratory), the Georgia Research Alliance GRA and Emory University Research Committee and the Clinical and Translational Science Award Program (National Center for Research Resources of the US National Institutes of Health; to N.R.). The findings and conclusions in this report are those of the author(s) and do not necessarily represent the views of the Centers for Disease Control and Prevention.

Author information

Authors and Affiliations

Authors

Contributions

S.L. did analyses in Figures 4 and 5 and Supplementary Figures 4,5,6,7,8,9,10,11,12,13,14 and 18,19,20; S.L. and H.I.N. did analyses in Figures 2 and 3 and Supplementary Figure 2 and helped with study design and presentation; N.R. organized the clinical study; S.D. did experiments and analyses in Figure 6 and Supplementary Figures 15,16,17; S.L. and S.D. did analysis in Supplementary Figure 3; S.R.-S., D.S.S., S.E.J., A.M., G.R. and G.M.C. did experiments in Figure 1; S.P., C.Q., D.C. and A.K.P. prepared the online data portal and Figure 3a; C.D. did experiments in Supplementary Fig. 1a,b; S.K. assisted with experiments in Figure 6 and Supplementary Figure 16; M.J.M. supervised the clinical study; R.A. supervised the study in Supplementary Figure 1a,b; D.S.S. helped conceive of and design the study; B.P. conceived of the study and designed and supervised the experiments and analyses; and S.L., H.I.N. and B.P. wrote the paper.

Corresponding author

Correspondence to Bali Pulendran.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Antibody responses induced by meningococcal vaccines.

(a) ELISPOT captured day 7 IgG antibody-secreting cells (ASCs) in a subset of vaccinees. Plates were coated with goat anti human IgG antibody, MCV4, DT or MPSV4 and detected with anti human IgG biotin and streptavidin HRP. (b) Two representative subjects are shown. On the left, 1X corresponds to 0.17 million PBMCs. DT = Diphtheria Toxoid. (c) Production of IgG is a robust indicator of antibody response. Day 30 IgG correlates well with data from day 180 and 2 years. Data are shown for both vaccines. The day 30/0 IgG data (serogroups A plus C) were thus used for the later analysis of antibody correlation.

Supplementary Figure 2 The XBP-1 network is upregulated in the MCV4 transcriptomic response.

Targets of XBP-1 include genes associated with antibody-secreting cell differentiation (tan shading) and the unfolded protein response (purple shading). MCV4 DEGs are colored in red. Enrichment was assessed by Fisher's exact test (p = 5 x 10-14).

Supplementary Figure 3 Cytokines and chemokines measured by Luminex assay for meningococcal vaccines.

Left: Day 3 and 7 levels (log2, pg/ml) compared to day 0. Each row represents a cytokine and each column represents a vaccinee. Right: Serum protein levels (log2, pg/ml) that are significantly different after vaccination. * p < 0.05, paired t-test.

Supplementary Figure 4 Integration of DEGs with interactome and bibliome data.

(a) Illustration of the network integration method. DEGs from a vaccine study are connected by their links in the Interactome or Bibliome. Additional “linker” genes are then included based on their significant association to the DEGs (see Method for details). (b) Numbers of upregulated DEGs in common to 1 to 5 vaccine datasets. (c) Numbers of genes in the integrated network, and numbers of genes in common to 1 to 5 vaccine datasets. (d) The genes in common between MCV4 network and TIV network are highly expressed in plasma cells. As all genes are ranked by their expression level in sorted plasma cells (the color bar, according to data in Abbas et al 2009, PLoS ONE, 4:e6098), the ranks of MCV4/TIV common genes are indicated by the horizontal bars. (e) A subnetwork that is common between YF-17D and LAIV, enriched for TCR signaling pathway and interferon response genes. Genes in dark gray: DEGs as in (b), light gray: “linker” genes that have p < 0.05 in YF-17D transcriptomic data. (f) Genes enriched in the “immune systems development” GO category from Figure 3c.

Supplementary Figure 5 Molecular pathways induced by different vaccines.

Selected pathways from NCI-Nature Pathway Interaction Database whose expression were significantly (p < 0.01) changing at (a) day 3 or (b) day 7 compared to pre-vaccination data (day 0). Genes are ranked by t-score and the significance of pathways is tested by GSEA (see online method for details).

Supplementary Figure 6 Pathways associated with antibody response.

Selected pathways from NCI-Nature Pathway Interaction Database whose expression at (a) day 3 or (b) day 7 post-vaccination was significantly (p < 0.01) correlated to antibody response. Genes are ranked according to Pearson correlation to antibody response, and the significance of pathways is tested by GSEA (see Method for details). Influenza LAIV is excluded here because this vaccine does not induce serum/plasma antibody responses. MCV4 has two sets of antibody correlation data: one against meningococcal polysaccharide (MCV4-PS) and another against diphtheria toxoid (MCV4-DT).

Supplementary Figure 7 Examples of protein complexes detected during BTM construction.

(a) Ribosome complex extracted from the master network. (b) Nuclear pore complex and (c) Splicesome complexes extracted from BTM modules. Each edge in the pictures represents a coexpression relationship detected in at least 3 publicly available microarray blood studies.

Supplementary Figure 8 Examples of BTM modules.

(a) “T cell differentiation via ITK and PKC” BTM module which contains genes whose functions are supported by literature. This set of genes, however was not found in pathway databases. (b) “Chromosome Y linked” BTM module contains many genes transcribed in Y chromosome.

Supplementary Figure 9 Discriminative power of BTM modules compared with canonical pathways.

Discriminative power is shown as t-scores in two-class comparison. The horizontal gray lines show the threshold of confidence level of α = 0.001. By this cutoff, the numbers of significant modules/pathways can be looked up on the X-axis (top two numbers inserted by curves). YF-17D data were from Querec et al 2009, Nature Immunology 10:116; MCV4 this study; Turberculosis from Berry et al 2010, Nature 466: 973; RTS,S from Vahey et al 2010 J Infect Dis 201:580. All testing data sets were never part of the BTM construction process. KEGG, BioCarta and NCI_PID are respective pathway databases. Chaussabel modules were version 2 based on Chaussabel et al 2008 Immunity 29:150. “Random” modules contain randomly selected genes, matched to the sizes of KEGG pathways.

Supplementary Figure 10 Day 7 activity of plasma cells–immunoglobulin module (M156.1; Figure 4d) in blood transcriptome correlated to day 30 antibody response to TIV.

The TIV 2007 (n=9) and 2008 (n=24) data sets were described in Nakaya et al 2011, Nature Immunology 12:786.

Supplementary Figure 11 Hierarchical clustering of antibody correlations of BTM modules.

Heat map of BTMs (rows) and vaccines (columns) whose baseline-normalized expression at day 3 correlated with baseline-normalized antibody response after vaccination (colors in map indicate Pearson correlation values). Clustering was performed (a) using all 346 BTM modules and (b) using the top 50% modules with highest variance across these 6 datasets.

Supplementary Figure 12 Distinctive early transcription programs correlated to vaccine antibody response.

(a) Clustering analysis from Supplementary Figure 10b, marked by the three programs: Primary viral response (b), Polysaccharide response (c) and Protein recall response (d). From each data set in (a), the top 20 modules with highest positive correlations and the top 20 modules with highest negative correlations were selected. Among these selected modules, the modules concordant between two datasets (Pearson correlation coefficient above 0.3 or below -0.3 in both datasets) are shown in (b, c, d) and the rest are shown in (e, f, g).

Supplementary Figure 13 DC-related BTM modules and genes.

A filled unit in the center grid indicates the membership of the gene (top axis) in the corresponding module (left axis). The heat map on the right shows the Pearson correlation between module activity and antibody response in each study. The bottom heat map shows correlation between module member genes and antibody response.

Supplementary Figure 14 The DC surface signature module is positively correlated to polysaccharide response.

(a) Network of “DC surface” module member genes. (b) Correlation between D3/0 module activity and D30/0 anti-polysaccharide IgG response in MCV4 and MPSV4.

Supplementary Figure 15 Polysaccharide vaccine stimulates DCs.

MPSV4 stimulates the phenotypic maturation of human myeloid DC, while MCV4 to a much lesser extent. FACS data were obtained after 24 h (n = 4–6).

Supplementary Figure 16 MPSV4 stimulation of DCs is dependent on MyD88, TRIF, TLR4 and ASC in vitro.

Expanded version of Figure 6 showing genes that were required for DC in vitro stimulation with MPSV4. CD11c+ DCs were isolated by MACS from spleens of C57BL/6 mice (WT) or various knock-out mice and stimulated for 24 h; *p<0.05, ***p<0.0001, unpaired t-test was used to compare WT vs. KO (n = 4–6).

Supplementary Figure 17 Antigen-specific serum antibody and SBA titers induced by MCV4 and MPSV4 in mice in vivo.

C57BL/6 mice were immunized sub–cutaneously with one human dose (a-c, n=4-9) or two doses (d, n=5) of MPSV4 (blue) or MCV4 (red) or left untreated. (a) Mice were bled after 7, 14, and 28 days and serum IgM, IgG and IgA levels against meningococcal serogroup C were measured. (b) Serum IgG against serogroups A, Y and W-135 were measured at day 28 and (c) Serum Bactericidal Activity (SBA) titers against serogroups A and/or C were measured after 28 days. (d) Mice were boosted with a 2nd dose at day 30. Each gray line represents an individual animal. * p<0.05, ** p<0.01, Mann-Whitney test.

Supplementary Figure 18 Cell-type expression patterns in BTM-antibody analysis.

BTM activities of cell surface signatures (see details in Suppl. Text), and their correlation to antibody response. X-axis shows module activity at D3/0 or D7/0, Y-axis the D30/0 antibody response.

Supplementary Figure 19 BTM modules whose activity on day 7 is correlated to later antibody response in two or more data sets.

Each segment on the circle corresponds to one vaccine dataset. In each segment, the inner circular bands show an ordered list of all BTM modules, layered by histograms of significantly correlated modules, red for positive correlation and blue for negative correlation. Each link inside the circles connects a significant module (top 50 in one dataset by p-value) that is common between different data sets. Modules common to three vaccines are linked in black, and their labels are marked by a black dot. Plasma and immunoglobulin modules are linked in green. An interactive version of this figure is available at online data portal (Interactive Figure 3).

Supplementary Figure 20 B cell–related BTM modules and genes.

A filled unit in the center grid indicates the membership of the gene (top axis) in the corresponding module (left axis). The heat map on the right shows the Pearson correlation between module activity and antibody response in each study. The bottom heat map shows correlation between module member genes and antibody response.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–20 and Supplementary Note (PDF 5649 kb)

Supplementary Table 1: Differences in gene expression in five vaccine studies.

Numbers of differentially expressed genes compared to day 0 in five vaccine studies, at varying statistical cutoff by FDR or p-value, without (A) or with (B) gene filtering. Gene filtering is based on Nakaya et al. (2011) Nature immunology 12:786, using probesets with at least 1.25 fold change in > 20% subjects. p-values were based paired t-test. FDR was based on Storey and Tibshirani (2003), PNAS, 100:9440. (XLSX 11 kb)

Supplementary Table 2: Correlation of BTM activity to vaccine antibody responses.

Pearson correlation coefficients are reported. For clarity, only absolute values greater than 0.25 are shown. (XLSX 62 kb)

Supplementary Table 3: GSEA analysis using the BTM modules.

Genes are ranked according to Pearson correlation to antibody response, and the significance of BTM modules is tested by GSEA (see online method for details). Normalized Enrichment Score values are shown for p-values < 0.05. (XLSX 34 kb)

Data and tutorial package of Blood Transcription Modules.

This package provides BTMs in reusable data formats, and a step-by-step demonstration of their applications. [Order of files within the zip package:] BTM_application_tutorial.pdf README_package_content_description.pdf BTM_for_GSEA_20131008.gmt monocytes_vs_bcells.txt gene_ab_correlation.rnk MCV4_D3v0_probesets.txt btm_tool.py btm_example_data.py btm_annotation_table.xls (ZIP 4031 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Cite this article

Li, S., Rouphael, N., Duraisingham, S. et al. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat Immunol 15, 195–204 (2014). https://doi.org/10.1038/ni.2789

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2789

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