Transcriptomic profiling facilitates classification of response to influenza challenge

Emma E Davenport, Richard D Antrobus, Patrick J Lillie, Sarah Gilbert, Julian C Knight, Emma E Davenport, Richard D Antrobus, Patrick J Lillie, Sarah Gilbert, Julian C Knight

Abstract

Despite increases in vaccination coverage, reductions in influenza-related mortality have not been observed. Better vaccines are therefore required and influenza challenge studies can be used to test the efficacy of new vaccines. However, this requires the accurate post-challenge classification of subjects by outcome, which is limited in current methods that use artificial thresholds to assign 'symptomatic' and 'asymptomatic' phenotypes. We present data from an influenza challenge study in which 22 healthy adults (11 vaccinated) were inoculated with H3N2 influenza (A/Wisconsin/67/2005). We generated genome-wide gene expression data from peripheral blood taken immediately before the challenge and at 12, 24 and 48 h post-challenge. Variation in symptomatic scoring was found amongst those with laboratory confirmed influenza. By combining the dynamic transcriptomic data with the clinical parameters this variability can be reduced. We identified four subjects with severe laboratory confirmed influenza that show differential gene expression in 1103 probes 48 h post-challenge compared to the remaining subjects. We have further reduced this profile to six genes (CCL2, SEPT4, LAMP3, RTP4, MT1G and OAS3) that can be used to define these subjects. We have used this gene set to predict symptomatic infection from an independent study. This analysis gives further insight into host-pathogen interactions during influenza infection. However, the major potential value is in the clinical trial setting by providing a more quantitative method to better classify symptomatic individuals post influenza challenge.

Key message: Differential gene expression signatures are seen following influenza challenge. Expression of six predictive genes can classify response to influenza challenge. The genomic influenza response classification replicates in an independent dataset.

Figures

Fig. 1
Fig. 1
Clinical observations and classification of study subjects. Each individual is assigned a unique letter code (A-V). Vaccinated individuals are shown in red. Individuals are defined as having no symptoms, mild or moderate/severe based on the summed self-reported symptoms collected over 6 days. Positive shedding indicating viral shedding was detected on at least 1 day after challenge. Seroconversion was determined by HI titre using serum samples obtained at 26-days post-challenge (Online Resource, Table S1). LCI is defined as mild or moderate/severe symptoms and viral shedding (shown by grey shaded area)
Fig. 2
Fig. 2
Principal components and hierarchical clustering plot. a The first three principal components are plotted with the proportion of variance explained by each component. The yellow oval highlights the samples taken from the four subjects with moderate/severe LCI 48 h post-challenge. N none or mild LCI, S moderate/severe LCI, V vaccinee, C control. b The hierarchical tree illustrates the relationship between clusters of samples. The height of the branches indicates the strength of the separation. The red oval highlights the samples taken from the four subjects with moderate/severe LCI 48 h post-challenge
Fig. 3
Fig. 3
Heatmaps for differentially expressed probes. The top 100 most significant differentially expressed probes between the four moderate/severe LCI, and the remaining samples at 48 h post-challenge were used to generate heatmaps of gene expression. a Moderate/severe LCI subjects. b Mild LCI subjects. Individuals D and G were vaccinated; others are unvaccinated
Fig. 4
Fig. 4
Pathway analysis of differential expression in LCI. The differentially expressed probes between the four moderate/severe LCI samples 48 h post-challenge and the remaining samples were used to determine pathway enrichment with IPA. a Pathways with significant enrichment based on Benjamini-Hochberg (B-H) multiple testing corrected p value <0.01 are shown. The blue bars show the −log(p value). Differentially expressed genes are plotted for each enriched pathway (red upregulated in moderate/severe LCI individuals, green downregulated) with numbers of enriched genes shown in brackets below x-axis labels. b The most significantly enriched pathway, interferon signalling, is shown with enriched genes shaded in colour

References

    1. World Health Organization. Influenza. Available via: . Accessed: 11/06/2013
    1. Simonsen L, Taylor RJ, Viboud C, Miller MA, Jackson LA. Mortality benefits of influenza vaccination in elderly people: an ongoing controversy. Lancet Infect. Dis. 2007;7:658–666. doi: 10.1016/S1473-3099(07)70236-0.
    1. Kobasa D, Jones SM, Shinya K, Kash JC, Copps J, Ebihara H, Hatta Y, Kim JH, Halfmann P, Hatta M, et al. Aberrant innate immune response in lethal infection of macaques with the 1918 influenza virus. Nature. 2007;445:319–323. doi: 10.1038/nature05495.
    1. Bermejo-Martin JF, Martin-Loeches I, Rello J, Anton A, Almansa R, Xu L, Lopez-Campos G, Pumarola T, Ran L, Ramirez P, et al. Host adaptive immunity deficiency in severe pandemic influenza. Crit. Care. 2010;14:R167. doi: 10.1186/cc9259.
    1. Herberg JA, Kaforou M, Gormley S, Sumner ER, Patel S, Jones KD, Paulus S, Fink C, Martinon-Torres F, Montana G et al (2013) Transcriptomic profiling in childhood H1N1/09 influenza reveals reduced expression of protein synthesis genes. J Infect Dis 208:1664–1668
    1. Mejias A, Dimo B, Suarez NM, Garcia C, Suarez-Arrabal MC, Jartti T, Blankenship D, Jordan-Villegas A, Ardura MI, Xu Z et al (2013) Whole blood gene expression profiles to assess pathogenesis and disease severity in infants with respiratory syncytial virus infection. PLoS Med 10:e1001549
    1. Parnell GP, McLean AS, Booth DR, Armstrong NJ, Nalos M, Huang SJ, Manak J, Tang W, Tam OY, Chan S et al (2012) A distinct influenza infection signature in the blood transcriptome of patients with severe community-acquired pneumonia. Crit Care 16:R157
    1. Song H, Wang Q, Guo Y, Liu S, Song R, Gao X, Dai L, Li B, Zhang D, Cheng J. Microarray analysis of microRNA expression in peripheral blood mononuclear cells of critically ill patients with influenza A (H1N1) BMC Infect. Dis. 2013;13:257. doi: 10.1186/1471-2334-13-257.
    1. Bucasas KL, Franco LM, Shaw CA, Bray MS, Wells JM, Nino D, Arden N, Quarles JM, Couch RB, Belmont JW. Early patterns of gene expression correlate with the humoral immune response to influenza vaccination in humans. J. Infect. Dis. 2011;203:921–929. doi: 10.1093/infdis/jiq156.
    1. Woods CW, McClain MT, Chen M, Zaas AK, Nicholson BP, Varkey J, Veldman T, Kingsmore SF, Huang Y, Lambkin-Williams R, et al. A host transcriptional signature for presymptomatic detection of infection in humans exposed to influenza H1N1 or H3N2. PLoS One. 2013;8:e52198. doi: 10.1371/journal.pone.0052198.
    1. Huang Y, Zaas AK, Rao A, Dobigeon N, Woolf PJ, Veldman T, Oien NC, McClain MT, Varkey JB, Nicholson B et al (2011) Temporal dynamics of host molecular responses differentiate symptomatic and asymptomatic influenza A infection. PLoS Genet 7:e1002234
    1. Berthoud TK, Hamill M, Lillie PJ, Hwenda L, Collins KA, Ewer KJ, Milicic A, Poyntz HC, Lambe T, Fletcher HA et al (2011) Potent CD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza A vaccine, MVA-NP M1. Clin Infect Dis 52:1–7
    1. Antrobus RD, Lillie PJ, Berthoud TK, Spencer AJ, McLaren JE, Ladell K, Lambe T, Milicic A, Price DA, Hill AV et al (2012) A T cell-inducing influenza vaccine for the elderly: safety and immunogenicity of MVA-NP+M1 in adults aged over 50 years. PLoS One 7:e48322
    1. Antrobus RD, Berthoud TK, Mullarkey CE, Hoschler K, Coughlan L, Zambon M, Hill AV, Gilbert SC (2014) Coadministration of seasonal influenza vaccine and MVA-NP+M1 simultaneously achieves potent humoral and cell-mediated responses. Mol Ther 22:233–238
    1. Lillie PJ, Berthoud TK, Powell TJ, Lambe T, Mullarkey C, Spencer AJ, Hamill M, Peng Y, Blais ME, Duncan CJ et al (2012) Preliminary assessment of the efficacy of a T-cell-based influenza vaccine, MVA-NP+M1, in humans. Clin Infect Dis 55:19–25
    1. Jackson GG, Dowling HF, Spiesman IG & Boand AV. (1958) Transmission of the common cold to volunteers under controlled conditions. I. The common cold as a clinical entity. A.M.A. archives of internal medicine 101:267–278
    1. Zaas AK, Chen M, Varkey J, Veldman T, Hero AO, 3rd, Lucas J, Huang Y, Turner R, Gilbert A, Lambkin-Williams R, et al. Gene expression signatures diagnose influenza and other symptomatic respiratory viral infections in humans. Cell Host Microbe. 2009;6:207–217. doi: 10.1016/j.chom.2009.07.006.
    1. Huber W, von Heydebreck A, Sultmann H, Poustka A, Vingron M. Variance stabilization applied to microarray data calibration and to the quantification of differential expression. Bioinformatics. 2002;18(Suppl 1):S96–S104. doi: 10.1093/bioinformatics/18.suppl_1.S96.
    1. R: A language and environment for statistical computing (R Foundation for Statistical Computing, Vienna, Austria, 2013)
    1. Smyth GK. in Bioinformatics and computational biology solutions using R and Bioconductor (ed Robert Gentleman et al.) 397–420 (Springer, 2005)
    1. Conesa A, Nueda MJ, Ferrer A, Talon M. maSigPro: a method to identify significantly differential expression profiles in time-course microarray experiments. Bioinformatics. 2006;22:1096–1102. doi: 10.1093/bioinformatics/btl056.
    1. Tai YC. timecourse: Statistical Analysis for Developmental Microarray Time Course Data. Available via:. Accessed: version 1.28.0. 2007
    1. Hastie TT, Narasimhan B & Chu G. pamr: Pam: prediction analysis for microarrays. Available via:. Accessed: version 1.54.1. 2013
    1. Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–210. doi: 10.1093/nar/30.1.207.
    1. Smyth GK. (2004) Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol 3:Article3
    1. Garcia-Sastre A, Biron CA. Type 1 interferons and the virus-host relationship: a lesson in detente. Science. 2006;312:879–882. doi: 10.1126/science.1125676.
    1. Hermant P, Michiels T. Interferon-lambda in the context of viral infections: production, response and therapeutic implications. J Innate Immun. 2014;6:563–574. doi: 10.1159/000360084.
    1. Jewell NA, Cline T, Mertz SE, Smirnov SV, Flano E, Schindler C, Grieves JL, Durbin RK, Kotenko SV, Durbin JE. Lambda interferon is the predominant interferon induced by influenza A virus infection in vivo. J. Virol. 2010;84:11515–11522. doi: 10.1128/JVI.01703-09.
    1. Sadler AJ, Williams BR. Interferon-inducible antiviral effectors. Nat. Rev. Immunol. 2008;8:559–568. doi: 10.1038/nri2314.
    1. Iwai A, Shiozaki T, Miyazaki T. Relevance of signaling molecules for apoptosis induction on influenza A virus replication. Biochem. Biophys. Res. Commun. 2013;441:531–537. doi: 10.1016/j.bbrc.2013.10.100.
    1. Baccam P, Beauchemin C, Macken CA, Hayden FG, Perelson AS. Kinetics of influenza A virus infection in humans. J. Virol. 2006;80:7590–7599. doi: 10.1128/JVI.01623-05.
    1. Smith AM, Perelson AS. Influenza A virus infection kinetics: quantitative data and models. Wiley Interdiscip. Rev. Syst. Biol. Med. 2011;3:429–445. doi: 10.1002/wsbm.129.
    1. Kawada J, Kimura H, Kamachi Y, Nishikawa K, Taniguchi M, Nagaoka K, Kurahashi H, Kojima S, Morishima T. Analysis of gene-expression profiles by oligonucleotide microarray in children with influenza. J. Gen. Virol. 2006;87:1677–1683. doi: 10.1099/vir.0.81670-0.
    1. Wang J, Nikrad MP, Travanty EA, Zhou B, Phang T, Gao B, Alford T, Ito Y, Nahreini P, Hartshorn K, et al. Innate immune response of human alveolar macrophages during influenza A infection. PLoS One. 2012;7:e29879. doi: 10.1371/journal.pone.0029879.
    1. Shapira SD, Gat-Viks I, Shum BO, Dricot A, de Grace MM, Wu L, Gupta PB, Hao T, Silver SJ, Root DE, et al. A physical and regulatory map of host-influenza interactions reveals pathways in H1N1 infection. Cell. 2009;139:1255–1267. doi: 10.1016/j.cell.2009.12.018.
    1. Zhang YH, Wang S, Yan HP, Zhao DT, Zhang X, Li A, Wang ZK, Wu H, Zhao Y. Prognostic values of chemokines in different clinical outcomes of influenza A (H1N1)-infected patients. Zhonghua yi xue za zhi. 2012;92:2020–2022.
    1. Sivan G, Martin SE, Myers TG, Buehler E, Szymczyk KH, Ormanoglu P, Moss B. Human genome-wide RNAi screen reveals a role for nuclear pore proteins in poxvirus morphogenesis. Proc. Natl. Acad. Sci. U. S. A. 2013;110:3519–3524. doi: 10.1073/pnas.1300708110.
    1. Mehraj V, Textoris J, Ben Amara A, Ghigo E, Raoult D, Capo C, Mege JL. Monocyte responses in the context of Q fever: from a static polarized model to a kinetic model of activation. J. Infect. Dis. 2013;208:942–951. doi: 10.1093/infdis/jit266.
    1. Pauwels M, van Weyenbergh J, Soumillion A, Proost P, De Ley M. Induction by zinc of specific metallothionein isoforms in human monocytes. Eur. J. Biochem. 1994;220:105–110. doi: 10.1111/j.1432-1033.1994.tb18603.x.
    1. Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, Rice CM. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472:481–485. doi: 10.1038/nature09907.
    1. Balasubramaniam VR, Wai TH, Omar AR, Othman I, Hassan SS. Cellular transcripts of chicken brain tissues in response to H5N1 and Newcastle disease virus infection. Virol. J. 2012;9:53. doi: 10.1186/1743-422X-9-53.
    1. de Saint-Vis B, Vincent J, Vandenabeele S, Vanbervliet B, Pin JJ, Ait-Yahia S, Patel S, Mattei MG, Banchereau J, Zurawski S, Davoust J, Caux C, Lebecque S. A novel lysosome-associated membrane glycoprotein, DC-LAMP, induced upon DC maturation, is transiently expressed in MHC class II compartment. Immunity. 1998;9:325–336. doi: 10.1016/S1074-7613(00)80615-9.
    1. Zhou Z, Xue Q, Wan Y, Yang Y, Wang J, Hung T. Lysosome-associated membrane glycoprotein 3 is involved in influenza A virus replication in human lung epithelial (A549) cells. Virol. J. 2011;8:384. doi: 10.1186/1743-422X-8-384.
    1. Zak DE, Andersen-Nissen E, Peterson ER, Sato A, Hamilton MK, Borgerding J, Krishnamurty AT, Chang JT, Adams DJ, Hensley TR, 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. U. S. A. 2012;109:E3503–E3512. doi: 10.1073/pnas.1208972109.

Source: PubMed

3
Abonner