Progression of whole-blood transcriptional signatures from interferon-induced to neutrophil-associated patterns in severe influenza

Jake Dunning, Simon Blankley, Long T Hoang, Mike Cox, Christine M Graham, Philip L James, Chloe I Bloom, Damien Chaussabel, Jacques Banchereau, Stephen J Brett, Miriam F Moffatt, Anne O'Garra, Peter J M Openshaw, MOSAIC Investigators, Maximillian S Habibi, Sebastian L Johnston, Trevor T Hansel, Mike Levin, Ryan S Thwaites, John O Warner, William O Cookson, Brian G Gazzard, Alan Hay, John McCauley, Paul Aylin, Deborah Ashby, Wendy S Barclay, Ruth A Elderfield, Simon Nadel, Jethro A Herberg, Lydia N Drumright, Laura Garcia-Alvarez, Alison H Holmes, Onn M Kon, Stephen J Aston, Stephen B Gordon, Tracy Hussell, Catherine Thompson, Maria C Zambon, Kenneth J Baillie, David A Hume, Peter Simmonds, Andrew Hayward, Rosalind L Smyth, Paul S McNamara, Malcolm G Semple, Jonathan S Nguyen-Van-Tam, Ling-Pei Ho, Andrew J McMichael, Paul Kellam, Walt E Adamson, William F Carman, Mark J Griffiths, Jake Dunning, Simon Blankley, Long T Hoang, Mike Cox, Christine M Graham, Philip L James, Chloe I Bloom, Damien Chaussabel, Jacques Banchereau, Stephen J Brett, Miriam F Moffatt, Anne O'Garra, Peter J M Openshaw, MOSAIC Investigators, Maximillian S Habibi, Sebastian L Johnston, Trevor T Hansel, Mike Levin, Ryan S Thwaites, John O Warner, William O Cookson, Brian G Gazzard, Alan Hay, John McCauley, Paul Aylin, Deborah Ashby, Wendy S Barclay, Ruth A Elderfield, Simon Nadel, Jethro A Herberg, Lydia N Drumright, Laura Garcia-Alvarez, Alison H Holmes, Onn M Kon, Stephen J Aston, Stephen B Gordon, Tracy Hussell, Catherine Thompson, Maria C Zambon, Kenneth J Baillie, David A Hume, Peter Simmonds, Andrew Hayward, Rosalind L Smyth, Paul S McNamara, Malcolm G Semple, Jonathan S Nguyen-Van-Tam, Ling-Pei Ho, Andrew J McMichael, Paul Kellam, Walt E Adamson, William F Carman, Mark J Griffiths

Abstract

Transcriptional profiles and host-response biomarkers are used increasingly to investigate the severity, subtype and pathogenesis of disease. We now describe whole-blood mRNA signatures and concentrations of local and systemic immunological mediators in 131 adults hospitalized with influenza, from whom extensive clinical and investigational data were obtained by MOSAIC investigators. Signatures reflective of interferon-related antiviral pathways were common up to day 4 of symptoms in patients who did not require mechanical ventilator support; in those who needed mechanical ventilation, an inflammatory, activated-neutrophil and cell-stress or death ('bacterial') pattern was seen, even early in disease. Identifiable bacterial co-infection was not necessary for this 'bacterial' signature but was able to enhance its development while attenuating the early 'viral' signature. Our findings emphasize the importance of timing and severity in the interpretation of host responses to acute viral infection and identify specific patterns of immune-system activation that might enable the development of novel diagnostic and therapeutic tools for severe influenza.

Conflict of interest statement

Competing Financial Interests

The authors declare no competing financial interests.

Figures

Figure 1. Transcriptional signature of influenza compared…
Figure 1. Transcriptional signature of influenza compared to healthy controls.
(a) Principal component analysis of all transcripts significantly above background in at least 10% of samples (130 healthy controls (green squares), 97 influenza A (red circles: H1N1; green triangles, H3N2), and 12 influenza B (purple squares); all from 2010/11). (b) Modular analysis of 2010/11 influenza patients relative to healthy controls. The expression of the modules is shown on the left according to the colour intensity display; the corresponding modules are identified in the key to the right. (c) Weighted ‘molecular distance to health’ (MDTH24) of 2010/11 influenza patients compared to healthy controls, undertaken on 4526 transcripts that were significantly detected above background, filtered for low expression (transcripts retained if >2 fold-change (FC) from median normalised intensity value in more than 10% of all samples). Box whisker plot with min/max lines; statistical test: Mann-Whitney P<0.0001. (d) Heat-map of 1255 normalised intensity value transcripts, filtered for low expression then statistically filtered (Mann-Whitney with Bonferroni multiple testing correction P<0.01) followed by fold change filter between groups (transcripts retained if >2FC between any 2 groups). Listed next to the heat-map are the top five IPA canonical pathways (by significance; P<0.05, Fisher’s Exact test) for upregulated and downregulated transcripts. (e) Heat-map of normalised intensity values of the top 25 significant transcripts by mean fold-change between healthy controls and influenza groups clustered on entities and by individuals (Pearson’s uncentered (cosine) with averaged linkage).
Figure 2. Severity of disease is associated…
Figure 2. Severity of disease is associated with diminished expression of interferon-related modules and over-expression of inflammation modules.
(a) Weighted MDTH of 2010/11 influenza patients (n=109) grouped by severity of illness score (1: normoxic (n=47); 2: hypoxia requiring correction by mask oxygen (n=34); 3: mechanical ventilation (n=28)), compared to healthy controls (HC; n=130), based on 4526 transcripts that were significantly expressed above background and filtered for low expression (transcripts retained if >2FC from median normalised intensity value in more than 10% of all samples). Box whisker plots are shown with min/max lines. (b) Modular analysis of 2010/11 influenza patients (n=109) grouped by severity, relative to healthy controls (n=130). The colour intensity correlates with the percentage of genes in that module that are significantly differentially expressed.
Figure 3. Severe disease is associated with…
Figure 3. Severe disease is associated with lower expression of “viral” response genes, compared to early and less severe influenza.
(a) Heat-map of 231 normalised intensity value transcripts, obtained by filtering for low expression followed by statistical filtering (Kruskal-Wallis with Bonferroni multiple testing correction P<0.01) followed by fold change filter between groups (restricted to initial T1 samples, transcripts retained if >2FC between severity 3 and severity 1&2). Listed next to the heat-map are the top GO terms for the 3 major subdivisions of the dendrogram (clustered by Pearson’s uncentered (cosine) with average linkage rule). 2010/11 cohort: severity 1, n=47; severity 2, n=34; severity 3, n=28; HC, n=130. (b) Weighted molecular score (relative to healthy controls, n=130) of the 112 ‘bacterial response’ transcripts plotted against molecular score of the 51 ‘viral response’ transcripts for the 109 influenza individuals at the T1 time point. Severity of illness is indicated by different colours of dots: severity 1, black dots; severity 2, blue dots; severity 3, red dots. Circled dots identify patients with confirmed bacteraemia. (c) IPA significantly activated (z score >2) or (d) repressed (z score <2) biofunctions, identified by analysis of 231 transcript list; selected networks of biofunctional genes are shown.
Figure 4. Relationship between severity of illness,…
Figure 4. Relationship between severity of illness, bacterial infection, procalcitonin and molecular scores.
’Viral’ and ‘bacterial’ MDTH scores (according to GO terms, as described in Fig. 3) calculated for patients with confirmed influenza (2010/11 cohort, n=109) according to clinical categories at both the first and second sampling time-points (T1 and T2). Loess fitting was used to interpolate and estimate mean values non-parametrically from the data (solid lines); dashed lines show the estimated 95% confidence interval values of the mean; statistical significance of differences were calculated using Chi-squared tests to compare the deviance of generalized linear models. (a) Viral MDTH according to day of illness, for influenza patients stratified by severity of illness. (b) Bacterial MDTH according to day of illness, for influenza patients stratified by severity of illness. (c) Viral MDTH according to day of illness, for influenza patients stratified by presence (Bac+, n=39; 63 samples) or absence (Bac-, n=34; 52 samples) of clinically significant bacterial co-infections. (d) Bacterial MDTH according to day of illness, for influenza patients stratified by presence (Bac+, n=39; 63 samples) or absence (Bac-, n=34; 52 samples) of clinically significant bacterial co-infections. (e) Viral MDTH according to blood procalcitonin levels, for influenza patients stratified by presence (Bac+, n=39; 63 samples) or absence (Bac-, n=34; 52 samples) of clinically significant bacterial co-infections. (f) Bacterial MDTH according to blood procalcitonin levels, for influenza patients stratified by presence (Bac+, n=39; 63 samples) or absence (Bac-, n=34; 52 samples) of clinically significant bacterial co-infections.
Figure 5. Levels of selected mediators in…
Figure 5. Levels of selected mediators in different compartments according to severity of illness and clinical designation of probable bacterial co-infection status.
Serum, nasopharyngeal aspirate (NPA) and nasabsorption eluates were obtained from influenza patients at T1, and from adult healthy controls. Results are shown for IL-1β (a, b, c), IL-6 (d, e, f), CXCL8 (g, h, i), and IFN-α2a (j, k, l). For each mediator box plot, the central line shows the median mediator level (pg/mL, log scale) and the box margins show the interquartile range; outer bars show the range. Zero values and values below the lower limit of detection were assigned half the geometric mean lower limit of detection for display purposes. The upper limit of detection for all assays was 2500 pg/mL. Kruskal-Wallis test with Dunn’s post test was used to assess significance (*** p<0.001; ** p<0.01; * p<0.05; NS = not significant). Severity of illness at T1 is shown. HC = healthy controls. Serum samples for HCs and participants with severity 1, 2, and 3 illness (a, d, g, and j): n = 36, 58, 43, and 31, respectively. NPA samples for healthy controls and participants with severity 1, 2, and 3 illness (b, e, h and k): n = 35, 50, 32, and 27, respectively. Nasabsorption eluate samples for healthy controls and participants with severity 1, 2, and 3 illness (c, f, i and l): n = 36, 60, 43, and 30, respectively.
Figure 6. Relationships between severity of illness,…
Figure 6. Relationships between severity of illness, bacterial infection, and selected mediators.
Levels (pg/ml) of CXCL10 and IL-6 in serum (a-d) and NPA (e-h) according to day of illness at both the first and second sampling time-points (T1 and T2). Patients were stratified according to severity of illness (a, b, e, f) and by presence (Bac+; 39 subjects, 63 samples) or absence (Bac-; 34 subjects, 52 samples) of proven bacterial infection (c, d, g, h). Loess fitting was used to demonstrate time trends of mean values interpolated non-parametrically from the data (solid lines); dashed lines show the estimated 95% confidence interval values of the mean. Statistical significance of differences was calculated using Chi-squared tests to compare the deviance of generalized linear models.

References

    1. Stöhr K. Preventing and treating influenza. BMJ. 2003;326:1223–1224.
    1. Hayward AC, et al. Comparative community burden and severity of seasonal and pandemic influenza: results of the Flu Watch cohort study. The Lancet Respiratory medicine. 2014;2:445–454.
    1. Dawood FS, et al. Estimated global mortality associated with the first 12 months of 2009 pandemic influenza A H1N1 virus circulation: a modelling study. The Lancet infectious diseases. 2012;12:687–695.
    1. Bautista E, et al. Clinical aspects of pandemic 2009 influenza A (H1N1) virus infection. The New England journal of medicine. 2010;362:1708–1719.
    1. Hui DS, Lee N, Chan PK. Clinical management of pandemic 2009 influenza A(H1N1) infection. Chest. 2010;137:916–925.
    1. Peiris JS, Cheung CY, Leung CY, Nicholls JM. Innate immune responses to influenza A H5N1: friend or foe? Trends in immunology. 2009;30:574–584.
    1. Tisoncik JR, et al. Into the eye of the cytokine storm. Microbiology and molecular biology reviews : MMBR. 2012;76:16–32.
    1. de Jong MD, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat Med. 2006;12:1203–1207.
    1. Ioannidis I, et al. Plasticity and virus specificity of the airway epithelial cell immune response during respiratory virus infection. Journal of virology. 2012;86:5422–5436.
    1. Ramilo O, et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood. 2007;109:2066–2077.
    1. Zaas AK, et al. Gene expression signatures diagnose influenza and other symptomatic respiratory viral infections in humans. Cell Host Microbe. 2009;6:207–217.
    1. Woods CW, et al. A host transcriptional signature for presymptomatic detection of infection in humans exposed to influenza H1N1 or H3N2. PloS one. 2013;8:e52198.
    1. Nakaya HI, et al. Systems biology of vaccination for seasonal influenza in humans. Nat Immunol. 2011;12:786–795.
    1. Li S, et al. Molecular signatures of antibody responses derived from a systems biology study of five human vaccines. Nat Immunol. 2014;15:195–204.
    1. Obermoser G, et al. Systems scale interactive exploration reveals quantitative and qualitative differences in response to influenza and pneumococcal vaccines. Immunity. 2013;38:831–844.
    1. Blankley S, et al. The application of transcriptional blood signatures to enhance our understanding of the host response to infection: the example of tuberculosis. Philos Trans R Soc Lond B Biol Sci. 2014;369 20130427.
    1. Pascual V, Chaussabel D, Banchereau J. A genomic approach to human autoimmune diseases. Annual review of immunology. 2010;28:535–571.
    1. Everitt AR, et al. IFITM3 restricts the morbidity and mortality associated with influenza. Nature. 2012;484:519–523.
    1. Ruggiero T, et al. A(H1N1)pdm09 hemagglutinin D222G and D222N variants are frequently harbored by patients requiring extracorporeal membrane oxygenation and advanced respiratory assistance for severe A(H1N1)pdm09 infection. Influenza and other respiratory viruses. 2013;7:1416–1426.
    1. Rykkvin R, Kilander A, Dudman SG, Hungnes O. Within-patient emergence of the influenza A(H1N1)pdm09 HA1 222G variant and clear association with severe disease, Norway. Euro surveillance : bulletin Europeen sur les maladies transmissibles = European communicable disease bulletin. 2013;18
    1. Herfst S, et al. Introduction of virulence markers in PB2 of pandemic swine-origin influenza virus does not result in enhanced virulence or transmission. Journal of virology. 2010;84:3752–3758.
    1. Elderfield RA, et al. Accumulation of human-adapting mutations during circulation of A(H1N1)pdm09 influenza virus in humans in the United Kingdom. Journal of virology. 2014;88:13269–13283.
    1. Chaussabel D, et al. A modular analysis framework for blood genomics studies: application to systemic lupus erythematosus. Immunity. 2008;29:150–164.
    1. Pankla R, et al. Genomic transcriptional profiling identifies a candidate blood biomarker signature for the diagnosis of septicemic melioidosis. Genome Biol. 2009;10:R127.
    1. Simon L, Gauvin F, Amre DK, Saint-Louis P, Lacroix J. Serum procalcitonin and C-reactive protein levels as markers of bacterial infection: a systematic review and meta-analysis. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2004;39:206–217.
    1. Wu MH, et al. Can procalcitonin tests aid in identifying bacterial infections associated with influenza pneumonia? A systematic review and meta-analysis. Influenza and other respiratory viruses. 2013;7:349–355.
    1. Falsey AR, et al. Bacterial complications of respiratory tract viral illness: a comprehensive evaluation. The Journal of infectious diseases. 2013;208:432–441.
    1. McNab F, Mayer-Barber K, Sher A, Wack A, O'Garra A. Type I interferons in infectious disease. Nat Rev Immunol. 2015;15:87–103.
    1. Marshall JC. Iatrogenesis, inflammation and organ injury: insights from a murine model. Crit Care. 2006;10:173.
    1. Openshaw PJ, Chiu C. Protective and dysregulated T cell immunity in RSV infection. Curr Opin Virol. 2013;3:468–474.
    1. Davidson S, Crotta S, McCabe TM, Wack A. Pathogenic potential of interferon alphabeta in acute influenza infection. Nat Commun. 2014;5:3864.
    1. Gregory DJ, Kobzik L. Influenza lung injury: mechanisms and therapeutic opportunities. Am J Physiol Lung Cell Mol Physiol. 2015;309:L1041–1046.
    1. Short KR, Kroeze EJ, Fouchier RA, Kuiken T. Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect Dis. 2014;14:57–69.
    1. Sandler NG, et al. Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature. 2014;511:601–605.
    1. Biron CA. Interferons alpha and beta as immune regulators--a new look. Immunity. 2001;14:661–664.
    1. Narasaraju T, et al. Excessive neutrophils and neutrophil extracellular traps contribute to acute lung injury of influenza pneumonitis. The American journal of pathology. 2011;179:199–210.
    1. Lukens MV, et al. A systemic neutrophil response precedes robust CD8(+) T-cell activation during natural respiratory syncytial virus infection in infants. Journal of virology. 2010;84:2374–2383.
    1. Tate MD, Brooks AG, Reading PC, Mintern JD. Neutrophils sustain effective CD8(+) T-cell responses in the respiratory tract following influenza infection. Immunology and cell biology. 2012;90:197–205.
    1. Lim K, et al. Neutrophil trails guide influenza-specific CD8(+) T cells in the airways. Science (New York, N.Y.) 2015;349:aaa4352.
    1. Mayer-Barber KD, et al. Host-directed therapy of tuberculosis based on interleukin-1 and type I interferon crosstalk. Nature. 2014;511:99–103.
    1. Carr MJ, et al. Development of a real-time RT-PCR for the detection of swine-lineage influenza A (H1N1) virus infections. J Clin Virol. 2009;45:196–199.
    1. Nguyen-Van-Tam JS, et al. Risk factors for hospitalisation and poor outcome with pandemic A/H1N1 influenza: United Kingdom first wave (May-September 2009) Thorax. 2010;65:645–651.
    1. Ashburner M, et al. Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet. 2000;25:25–29.

Source: PubMed

3
Abonner