Cell-Based Systems Biology Analysis of Human AS03-Adjuvanted H5N1 Avian Influenza Vaccine Responses: A Phase I Randomized Controlled Trial

Leigh M Howard, Kristen L Hoek, Johannes B Goll, Parimal Samir, Allison Galassie, Tara M Allos, Xinnan Niu, Laura E Gordy, C Buddy Creech, Nripesh Prasad, Travis L Jensen, Heather Hill, Shawn E Levy, Sebastian Joyce, Andrew J Link, Kathryn M Edwards, Leigh M Howard, Kristen L Hoek, Johannes B Goll, Parimal Samir, Allison Galassie, Tara M Allos, Xinnan Niu, Laura E Gordy, C Buddy Creech, Nripesh Prasad, Travis L Jensen, Heather Hill, Shawn E Levy, Sebastian Joyce, Andrew J Link, Kathryn M Edwards

Abstract

Background: Vaccine development for influenza A/H5N1 is an important public health priority, but H5N1 vaccines are less immunogenic than seasonal influenza vaccines. Adjuvant System 03 (AS03) markedly enhances immune responses to H5N1 vaccine antigens, but the underlying molecular mechanisms are incompletely understood.

Objective and methods: We compared the safety (primary endpoint), immunogenicity (secondary), gene expression (tertiary) and cytokine responses (exploratory) between AS03-adjuvanted and unadjuvanted inactivated split-virus H5N1 influenza vaccines. In a double-blinded clinical trial, we randomized twenty adults aged 18-49 to receive two doses of either AS03-adjuvanted (n = 10) or unadjuvanted (n = 10) H5N1 vaccine 28 days apart. We used a systems biology approach to characterize and correlate changes in serum cytokines, antibody titers, and gene expression levels in six immune cell types at 1, 3, 7, and 28 days after the first vaccination.

Results: Both vaccines were well-tolerated. Nine of 10 subjects in the adjuvanted group and 0/10 in the unadjuvanted group exhibited seroprotection (hemagglutination inhibition antibody titer > 1:40) at day 56. Within 24 hours of AS03-adjuvanted vaccination, increased serum levels of IL-6 and IP-10 were noted. Interferon signaling and antigen processing and presentation-related gene responses were induced in dendritic cells, monocytes, and neutrophils. Upregulation of MHC class II antigen presentation-related genes was seen in neutrophils. Three days after AS03-adjuvanted vaccine, upregulation of genes involved in cell cycle and division was detected in NK cells and correlated with serum levels of IP-10. Early upregulation of interferon signaling-related genes was also found to predict seroprotection 56 days after first vaccination.

Conclusions: Using this cell-based systems approach, novel mechanisms of action for AS03-adjuvanted pandemic influenza vaccination were observed.

Trial registration: ClinicalTrials.gov NCT01573312.

Conflict of interest statement

KME has received grant funding for group B streptococcal vaccine research from Novartis and has participated as a member on a Novartis Data Safety Monitoring Board on influenza vaccines. CBC has received institutional grant support for staphylococcal vaccine research from Pfizer and Novartis Vaccines. JBG, TJL, and HH are employed by The Emmes Corporation. The Emmes Corporation is an independent contract research organization NIH-NIAID-DMID contracted with for collecting and analyzing data for clinical trials and studies to evaluate the safety and efficacy of vaccines, devices, and therapeutics for infectious diseases. The Emmes Corporation does not have any patents, products in development or marketed products to declare. These affiliations do not alter our adherence to PLOS ONE policies on sharing data and materials; however, we have not released raw sequencing data to maintain accordance with subject confidentiality requirements as outlined in the study’s Informed Consent Document, as approved by the Vanderbilt University Medical Center Institutional Review Board. The other authors declare that they have no conflicts of interest.

Figures

Fig 1. CONSORT diagram outlining study group…
Fig 1. CONSORT diagram outlining study group enrollment and randomization.
Fig 2. AS03-adjuvanted H5N1 vaccination induced early…
Fig 2. AS03-adjuvanted H5N1 vaccination induced early IP-10 and IL-6 serum cytokine and later protective immune responses relative to non-adjuvanted vaccine.
Serum Antibody and cytokine responses by vaccine group (red: SV-AS03 (n = 10), blue: SV-PBS (n = 10)). (A) HAI and Nt antibody GMT and 95% CI at each time point by vaccine group; p-values are based on two-sided t-test on the log scale adjusting for unequal variances if necessary. No multiple-testing adjustment was carried out. (B) Hemagglutination inhibition (HAI) and neutralizing antibody (Nt) titers in individual subjects at days 28 and 56 by vaccine group; titer is represented by bar height (left y-axis) while fold change from baseline is shown as a connected black line (right y-axis), cut offs are indicated by grey lines (solid: 1:40 titer, dashed: 4-fold change). (C) Median fold change and 95% bootstrap CI for serum cytokines/chemokines with significantly different responses at each post-vaccination time point by vaccine group; individual subject fold changes are shown in lighter colors; p-values are based on two-sided non-parametric exact Wilcoxon rank-sum test. No multiple-testing adjustment was carried out. (D) Radar chart of cytokines/chemokine median fold changes at each time point by vaccine group.
Fig 3. AS03-adjuvanted H5N1 vaccination primarily modulated…
Fig 3. AS03-adjuvanted H5N1 vaccination primarily modulated early innate immune cell gene expression responses.
Heatmaps of baseline log2 fold changes of DE genes for each cell type at each post-vaccination time point (A: monocytes, B: neutrophils, C: DC, D: NK cells, E: B cells, F: T cells). Dendograms were obtained using complete linkage clustering of uncentered pairwise Pearson correlation distances between log2 fold changes. Co-expressed gene cluster membership based on multiscale bootstrapping is highlighted along the gene dendogram on the left side. Cells are color-coded by log2 fold change (in red: upregulated from baseline; in green: downregulated from baseline.
Fig 4. Response time trends of clusters…
Fig 4. Response time trends of clusters of co-expressed differential genes by vaccine group.
Co-expressed gene cluster time trends of baseline log2 fold change in LCPM of DE genes by vaccine group. Header indicates cell type, primary functional classification, and time point. Mean log2 fold change across cluster genes is drawn in bold. Individual mean gene log2 fold changes are plotted in lighter colors. Multiscale bootstrapping was used to determine co-expressed gene clusters. (A) Interferon-inducible clusters with increased responses for the AS03-SV group identified in monocytes, B cells and T cells at day 1. (B) Interferon-inducible clusters with similar gene composition but different responses for the AS03-SV group in monocytes and neutrophils at day 1. (C) Additional interferon-inducible cluster modulated by AS03-SV identified in neutrophils at day 3. (D) Cell proliferation-related clusters with increased responses for the AS03-SV group identified in NK cells at day 3. (E) MHC class-II-related cluster with increased responses for the PBS-SV group identified in NK cells at day 28.
Fig 5. AS03-adjuvanted H5N1 vaccination triggered immune…
Fig 5. AS03-adjuvanted H5N1 vaccination triggered immune system pathways including interferon signaling, cell proliferation and antigen processing and presentation.
Heatmap of MSigDB Reactome pathways enriched in DE genes. Gene sets significantly enriched in at least two conditions (time point or cell type) are shown. Heatmap cell counts represent the number of DE genes in a pathway. Cells with numbers in brackets indicate significantly enriched sets. Cells are color-coded by Jaccard similarity index (in dark-red: high level of similarity, in light-yellow: low level of similarity). The index was calculated by comparing DE genes that had any Reactome annotation with genes in a certain Reactome pathway. Pathways were clustered based on the Jaccard distance between binary enrichment patterns across conditions.
Fig 6. AS03-adjuvanted H5N1 vaccination resulted in…
Fig 6. AS03-adjuvanted H5N1 vaccination resulted in increased cell cycle responses in NK-cells relative to non-adjuvanted vaccine 3 days post-vaccination.
The pathway map is based on the KEGG Cell cycle pathway. Pathway node color gradient encodes log2 fold change difference (LFCD) between vaccine groups (for multi-gene nodes the median LFCD is used). In red: up-regulated for the SV-AS03 group compared to the SV-PBS group, in green: down-regulated for the SV-AS03 group, in black: fold change close to 1, in dark grey: genes filtered out due to low overall expression, light grey: gene missing database mapping, white: non-human gene. DE genes are highlighted using red and green label colors.
Fig 7. Antigen processing and presentation responses…
Fig 7. Antigen processing and presentation responses for Neutrophils and NK-cells showed increased MHC-II sub-pathway activity that differed between vaccine groups.
(A) Neutrophil responses. (B) NK-cell responses. To the right: Heatmaps of subject-specific baseline log2 fold changes for KEGG Antigen processing and presentation genes across post-vaccination days by vaccine group. Colored in red: up regulated from baseline; colored in green: down regulated from baseline. To the left: KEGG pathway maps for post-vaccination days at which this pathway was significantly perturbed (day 1 for neutrophils, day 28 for NK-cells). Pathway node color gradient encodes log2 fold change difference (LFCD) between vaccine groups (for multi-gene nodes the median LFCD was used). In red: increased log2 fold change response for the SV-AS03 group relative to the SV-PBS group, in green: decreased log2 fold change response for the SV-AS03 group relative to the SV-AS03 group, and vice versa, in black: fold change close to 1, in dark grey: genes filtered out due to low overall expression, light grey: gene missing database mapping, white: non-human gene. DE genes are highlighted using red and green label colors.
Fig 8. Differential gene overlap between immune…
Fig 8. Differential gene overlap between immune cell types.
(A) Numbers of overlapping DE genes for each cell type and post-vaccination time point combination. Cells are color-coded by number of overlapping genes (dark-red: high overlap, light-yellow: low overlap). (B) Venn diagram summarizing DE gene overlap between neutrophils, monocytes, and dendritic cells at day 1.
Fig 9. Network analysis of proteins encoded…
Fig 9. Network analysis of proteins encoded by the 80 core AS03-responsive genes shared between neutrophils, monocytes, and dendritic cells 1 day post-vaccination.
Nodes are labeled by corresponding gene name. Edges represent experimentally determined (reported in at least two publications) binary protein-protein interactions or protein complex membership. Color and size of individual nodes indicates the degree of mean log2 fold change difference (LFCD) in corresponding gene expression between vaccine groups. Further details are provided as part of the legend within the figure.
Fig 10. AS03-adjuvanted vaccine gene responses in…
Fig 10. AS03-adjuvanted vaccine gene responses in neutrophils, monocytes and dendritic cells correlated with serum IP-10 and MIP−1α serum cytokine responses 1 day post-vaccination.
Canonical correlation plots summarizing key correlation patterns between changes in serum cytokine/antibody responses and gene responses at day 1 in (A,B) neutrophils (SV-AS03 and SV-PBS, respectively), (C) monocytes (SV-AS03), and (D) dendritic cells (SV-AS03). Red: upregulated genes; Green: downregulated genes; +/- gene name prefix: DE gene (yes/no); colored-dots: KEGG pathway class (BRITE level 2); grey lines: co-expressed gene clusters. The serum-based variable set (in black) included baseline log fold changes for 12 serum cytokines as well as baseline log fold changes of antibody titers (HAI: hemagglutination inhibition, Nt: microneutralization inhibition). The strength of the correlation is encoded by the distance from the center of the circle as well as by the angle between variables when viewed as vectors originating from the center. An acute angle (<90°) between variables represents a positive correlation, an obtuse angle (>90°) indicates a negative correlation, while a right angle is observed for zero correlation. Thus, maximum correlation is achieved when variables are closely placed together in the outer circle (or directly opposed on the outer circle.
Fig 11. AS03-adjuvanted vaccine NK-cell gene responses…
Fig 11. AS03-adjuvanted vaccine NK-cell gene responses correlated with serum IP-10 cytokine responses 3 days post-vaccination.
Canonical correlation plots summarizing key correlation patterns between changes in serum cytokine/antibody responses and gene responses at day 3 in NK cells; (A) SV-AS03, (B) SV-PBS. See Fig 10legend for additional information.
Fig 12. Key molecular immune events observed…
Fig 12. Key molecular immune events observed in peripheral blood cells after AS03-adjuvanted H5N1 split-virus vaccination.
(A) Intramuscular (IM) administration of AS03-adjuvanted H5N1 vaccine results in interferon (IFN) signaling. (B) Activation of IFN-inducible genes, in particular for monocytes (Mo), dendritic cells (DC), and neutrophils (Neut). (C) Significant up-regulation of CXCL10, the gene encoding IP-10, in peripheral blood Mo and DC implies that these cells contribute to increased serum IP-10 levels (in addition to local immune cells). (D) MHC class I- related antigen presentation genes are upregulated in monocytes Mo, Neut, and DC. (E) MHC class II- related antigen presentation genes are upregulated in Neut and, to a lesser extent, in Mo. At day 3, (F) IP-10 stimulates expansion of NK cells. (G) IFN-stimulated genes remain upregulated in Neut. (H) Upregulation of MHC class I and class II molecules stimulate cytotoxic T cells (CD8+) and T helper cells (CD4+), respectively, to initiate transition to the adaptive immune response. (I) Generation of protective H5-specific antibodies after a 2-dose vaccine regimen. Solid lines indicate that direct evidence for these events was seen in our serologic or transcriptomic data, while dashed lines indicate no direct evidence was observed.

References

    1. Russell CA, Fonville JM, Brown AE, Burke DF, Smith DL, James SL, et al. The potential for respiratory droplet-transmissible A/H5N1 influenza virus to evolve in a mammalian host. Science. 2012;336(6088):1541–7. PubMed Central PMCID: PMC3426314. 10.1126/science.1222526
    1. Organization WH. Cumulative number of confirmed human cases of avian influenza A (H5N1) reported to WHO: World Health Organization; 2014 [cited 2015 January 12]. Available from: .
    1. Treanor JJ, Campbell JD, Zangwill KM, Rowe T, Wolff M. Safety and immunogenicity of an inactivated subvirion influenza A (H5N1) vaccine. The New England journal of medicine. 2006;354(13):1343–51. 10.1056/NEJMoa055778
    1. Garcon N. Preclinical development of AS04. Methods in molecular biology. 2010;626:15–27. 10.1007/978-1-60761-585-9_2
    1. Garcon N, Chomez P, Van Mechelen M. GlaxoSmithKline Adjuvant Systems in vaccines: concepts, achievements and perspectives. Expert review of vaccines. 2007;6(5):723–39. 10.1586/14760584.6.5.723
    1. Garcon N, Vaughn DW, Didierlaurent AM. Development and evaluation of AS03, an Adjuvant System containing alpha-tocopherol and squalene in an oil-in-water emulsion. Expert review of vaccines. 2012;11(3):349–66. 10.1586/erv.11.192
    1. Klucker MF, Dalencon F, Probeck P, Haensler J. AF03, an alternative squalene emulsion-based vaccine adjuvant prepared by a phase inversion temperature method. Journal of pharmaceutical sciences. 2012;101(12):4490–500. 10.1002/jps.23311
    1. O'Hagan DT, Ott GS, Van Nest G. Recent advances in vaccine adjuvants: the development of MF59 emulsion and polymeric microparticles. Molecular medicine today. 1997;3(2):69–75. 10.1016/S1357-4310(96)10058-7
    1. Atmar RL, Keitel WA. Adjuvants for pandemic influenza vaccines. Current topics in microbiology and immunology. 2009;333:323–44. 10.1007/978-3-540-92165-3_16
    1. Coffman RL, Sher A, Seder RA. Vaccine adjuvants: putting innate immunity to work. Immunity. 2010;33(4):492–503. PubMed Central PMCID: PMC3420356. 10.1016/j.immuni.2010.10.002
    1. Reed SG, Orr MT, Fox CB. Key roles of adjuvants in modern vaccines. Nature medicine. 2013;19(12):1597–608. 10.1038/nm.3409
    1. Chu DW, Hwang SJ, Lim FS, Oh HM, Thongcharoen P, Yang PC, et al. Immunogenicity and tolerability of an AS03(A)-adjuvanted prepandemic influenza vaccine: a phase III study in a large population of Asian adults. Vaccine. 2009;27(52):7428–35. 10.1016/j.vaccine.2009.07.102
    1. Hwang SJ, Chang SC, Yu CJ, Chan YJ, Chen TJ, Hsieh SL, et al. Immunogenicity and safety of an AS03(A)-adjuvanted H5N1 influenza vaccine in a Taiwanese population. Journal of the Formosan Medical Association = Taiwan yi zhi. 2011;110(12):780–6. 10.1016/j.jfma.2011.11.009
    1. Langley JM, Frenette L, Ferguson L, Riff D, Sheldon E, Risi G, et al. Safety and cross-reactive immunogenicity of candidate AS03-adjuvanted prepandemic H5N1 influenza vaccines: a randomized controlled phase 1/2 trial in adults. The Journal of infectious diseases. 2010;201(11):1644–53. 10.1086/652701
    1. Leroux-Roels I, Borkowski A, Vanwolleghem T, Drame M, Clement F, Hons E, et al. Antigen sparing and cross-reactive immunity with an adjuvanted rH5N1 prototype pandemic influenza vaccine: a randomised controlled trial. Lancet. 2007;370(9587):580–9. 10.1016/S0140-6736(07)61297-5
    1. Leroux-Roels I, Roman F, Forgus S, Maes C, De Boever F, Drame M, et al. Priming with AS03 A-adjuvanted H5N1 influenza vaccine improves the kinetics, magnitude and durability of the immune response after a heterologous booster vaccination: an open non-randomised extension of a double-blind randomised primary study. Vaccine. 2010;28(3):849–57. 10.1016/j.vaccine.2009.10.017
    1. Nicholson KG, Abrams KR, Batham S, Clark TW, Hoschler K, Lim WS, et al. Immunogenicity and safety of a two-dose schedule of whole-virion and AS03A-adjuvanted 2009 influenza A (H1N1) vaccines: a randomised, multicentre, age-stratified, head-to-head trial. The Lancet Infectious diseases. 2011;11(2):91–101. 10.1016/S1473-3099(10)70296-6
    1. Jackson LA, Campbell JD, Frey SE, Edwards KM, Keitel WA, Kotloff KL, et al. Effect of Varying Doses of a Monovalent H7N9 Influenza Vaccine With and Without AS03 and MF59 Adjuvants on Immune Response: A Randomized Clinical Trial. Jama. 2015;314(3):237–46. 10.1001/jama.2015.7916
    1. Morel S, Didierlaurent A, Bourguignon P, Delhaye S, Baras B, Jacob V, et al. Adjuvant System AS03 containing alpha-tocopherol modulates innate immune response and leads to improved adaptive immunity. Vaccine. 2011;29(13):2461–73. 10.1016/j.vaccine.2011.01.011
    1. Caproni E, Tritto E, Cortese M, Muzzi A, Mosca F, Monaci E, et al. MF59 and Pam3CSK4 boost adaptive responses to influenza subunit vaccine through an IFN type I-independent mechanism of action. Journal of immunology. 2012;188(7):3088–98.
    1. Mosca F, Tritto E, Muzzi A, Monaci E, Bagnoli F, Iavarone C, et al. Molecular and cellular signatures of human vaccine adjuvants. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(30):10501–6. PubMed Central PMCID: PMC2483233. 10.1073/pnas.0804699105
    1. Sobolev O, Binda E, O'Farrell S, Lorenc A, Pradines J, Huang Y, et al. Adjuvanted influenza-H1N1 vaccination reveals lymphoid signatures of age-dependent early responses and of clinical adverse events. Nature immunology. 2016;17(2):204–13. 10.1038/ni.3328
    1. Burel JG, Apte SH, Doolan DL. Systems Approaches towards Molecular Profiling of Human Immunity. Trends in immunology. 2016;37(1):53–67. 10.1016/j.it.2015.11.006
    1. Trautmann L, Sekaly RP. Solving vaccine mysteries: a systems biology perspective. Nature immunology. 2011;12(8):729–31. 10.1038/ni.2078
    1. Buonaguro L, Pulendran B. Immunogenomics and systems biology of vaccines. Immunological reviews. 2011;239(1):197–208. PubMed Central PMCID: PMC3253346. 10.1111/j.1600-065X.2010.00971.x
    1. Nakaya HI, Li S, Pulendran B. Systems vaccinology: learning to compute the behavior of vaccine induced immunity. Wiley interdisciplinary reviews Systems biology and medicine. 2012;4(2):193–205. PubMed Central PMCID: PMC3288517. 10.1002/wsbm.163
    1. Pulendran B, Li S, Nakaya HI. Systems vaccinology. Immunity. 2010;33(4):516–29. PubMed Central PMCID: PMC3001343. 10.1016/j.immuni.2010.10.006
    1. Oberg AL, Kennedy RB, Li P, Ovsyannikova IG, Poland GA. Systems biology approaches to new vaccine development. Current opinion in immunology. 2011;23(3):436–43. PubMed Central PMCID: PMC3129601. 10.1016/j.coi.2011.04.005
    1. Hoek KL, Samir P, Howard LM, Niu X, Prasad N, Galassie A, et al. A cell-based systems biology assessment of human blood to monitor immune responses after influenza vaccination. PloS one. 2015;10(2):e0118528 PubMed Central PMCID: PMC4338067. 10.1371/journal.pone.0118528
    1. Noah DL, Hill H, Hines D, White EL, Wolff MC. Qualification of the hemagglutination inhibition assay in support of pandemic influenza vaccine licensure. Clinical and vaccine immunology: CVI. 2009;16(4):558–66. PubMed Central PMCID: PMC2668270. 10.1128/CVI.00368-08
    1. Eichelberger M, Golding H, Hess M, Weir J, Subbarao K, Luke CJ, et al. FDA/NIH/WHO public workshop on immune correlates of protection against influenza A viruses in support of pandemic vaccine development, Bethesda, Maryland, US, December 10–11, 2007. Vaccine. 2008;26(34):4299–303. 10.1016/j.vaccine.2008.06.012
    1. Services USDoHaH. Guidance for Industry: Clinical Data Needed to Support the Licensure of Pandemic Influenza Vaccines. Research FaDACfBEa; 2007.
    1. Efron B. Bootstrap methods: another look at the jackknife. The Annals of Statistics. 1979;7(1):1–26.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–40. PubMed Central PMCID: PMC2796818. 10.1093/bioinformatics/btp616
    1. Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome biology. 2010;11(3):R25 PubMed Central PMCID: PMCPMC2864565. 10.1186/gb-2010-11-3-r25
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B (Methodological). 1995:289–300.
    1. Dohm JC, Lottaz C, Borodina T, Himmelbauer H. Substantial biases in ultra-short read data sets from high-throughput DNA sequencing. Nucleic acids research. 2008;36(16):e105 PubMed Central PMCID: PMC2532726. 10.1093/nar/gkn425
    1. Suzuki R, Shimodaira H. Pvclust: an R package for assessing the uncertainty in hierarchical clustering. Bioinformatics. 2006;22(12):1540–2. 10.1093/bioinformatics/btl117
    1. Ogata H, Goto S, Fujibuchi W, Kanehisa M. Computation with the KEGG pathway database. Bio Systems. 1998;47(1–2):119–28.
    1. Liberzon A, Subramanian A, Pinchback R, Thorvaldsdottir H, Tamayo P, Mesirov JP. Molecular signatures database (MSigDB) 3.0. Bioinformatics. 2011;27(12):1739–40. PubMed Central PMCID: PMC3106198. 10.1093/bioinformatics/btr260
    1. Young MD, Wakefield MJ, Smyth GK, Oshlack A. Gene ontology analysis for RNA-seq: accounting for selection bias. Genome biology. 2010;11(2):R14 PubMed Central PMCID: PMC2872874. 10.1186/gb-2010-11-2-r14
    1. Segal L, Wouters S, Morelle D, Gautier G, Le Gal J, Martin T, et al. Non-clinical safety and biodistribution of AS03-adjuvanted inactivated pandemic influenza vaccines. Journal of applied toxicology: JAT. 2015;35(12):1564–76. 10.1002/jat.3130
    1. Joshi-Tope G, Gillespie M, Vastrik I, D'Eustachio P, Schmidt E, de Bono B, et al. Reactome: a knowledgebase of biological pathways. Nucleic acids research. 2005;33(Database issue):D428–32. PubMed Central PMCID: PMC540026. 10.1093/nar/gki072
    1. Kobayashi KS, van den Elsen PJ. NLRC5: a key regulator of MHC class I-dependent immune responses. Nature reviews Immunology. 2012;12(12):813–20. 10.1038/nri3339
    1. Neefjes J, Jongsma ML, Paul P, Bakke O. Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nature reviews Immunology. 2011;11(12):823–36. 10.1038/nri3084
    1. Karakikes I, Morrison IE, O'Toole P, Metodieva G, Navarrete CV, Gomez J, et al. Interaction of HLA-DR and CD74 at the cell surface of antigen-presenting cells by single particle image analysis. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2012;26(12):4886–96.
    1. Daugherty MD, Young JM, Kerns JA, Malik HS. Rapid evolution of PARP genes suggests a broad role for ADP-ribosylation in host-virus conflicts. PLoS genetics. 2014;10(5):e1004403 PubMed Central PMCID: PMC4038475. 10.1371/journal.pgen.1004403
    1. Ramsauer K, Farlik M, Zupkovitz G, Seiser C, Kroger A, Hauser H, et al. Distinct modes of action applied by transcription factors STAT1 and IRF1 to initiate transcription of the IFN-gamma-inducible gbp2 gene. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(8):2849–54. PubMed Central PMCID: PMC1815270. 10.1073/pnas.0610944104
    1. Samuel CE. Antiviral actions of interferons. Clinical microbiology reviews. 2001;14(4):778–809, table of contents. PubMed Central PMCID: PMC89003. 10.1128/CMR.14.4.778-809.2001
    1. Zhu Z, Shi Z, Yan W, Wei J, Shao D, Deng X, et al. Nonstructural protein 1 of influenza A virus interacts with human guanylate-binding protein 1 to antagonize antiviral activity. PloS one. 2013;8(2):e55920 PubMed Central PMCID: PMC3566120. 10.1371/journal.pone.0055920
    1. Itsui Y, Sakamoto N, Kakinuma S, Nakagawa M, Sekine-Osajima Y, Tasaka-Fujita M, et al. Antiviral effects of the interferon-induced protein guanylate binding protein 1 and its interaction with the hepatitis C virus NS5B protein. Hepatology. 2009;50(6):1727–37. 10.1002/hep.23195
    1. Talukder AH, Bao M, Kim TW, Facchinetti V, Hanabuchi S, Bover L, et al. Phospholipid scramblase 1 regulates Toll-like receptor 9-mediated type I interferon production in plasmacytoid dendritic cells. Cell research. 2012;22(7):1129–39. PubMed Central PMCID: PMC3391020. 10.1038/cr.2012.45
    1. Vladimer GI, Gorna MW, Superti-Furga G. IFITs: Emerging Roles as Key Anti-Viral Proteins. Frontiers in immunology. 2014;5:94 PubMed Central PMCID: PMC3948006. 10.3389/fimmu.2014.00094
    1. Chen HL, Gabrilovich D, Tampe R, Girgis KR, Nadaf S, Carbone DP. A functionally defective allele of TAP1 results in loss of MHC class I antigen presentation in a human lung cancer. Nature genetics. 1996;13(2):210–3. 10.1038/ng0696-210
    1. Procko E, Raghuraman G, Wiley DC, Raghavan M, Gaudet R. Identification of domain boundaries within the N-termini of TAP1 and TAP2 and their importance in tapasin binding and tapasin-mediated increase in peptide loading of MHC class I. Immunology and cell biology. 2005;83(5):475–82. 10.1111/j.1440-1711.2005.01354.x
    1. Hayashi T, Kobayashi Y, Kohsaka S, Sano K. The mutation in the ATP-binding region of JAK1, identified in human uterine leiomyosarcomas, results in defective interferon-gamma inducibility of TAP1 and LMP2. Oncogene. 2006;25(29):4016–26. 10.1038/sj.onc.1209434
    1. Alagarasu K, Honap T, Damle IM, Mulay AP, Shah PS, Cecilia D. Polymorphisms in the oligoadenylate synthetase gene cluster and its association with clinical outcomes of dengue virus infection. Infection, genetics and evolution: journal of molecular epidemiology and evolutionary genetics in infectious diseases. 2013;14:390–5. 10.1016/j.meegid.2012.12.021
    1. Balachandran S, Venkataraman T, Fisher PB, Barber GN. Fas-associated death domain-containing protein-mediated antiviral innate immune signaling involves the regulation of Irf7. Journal of immunology. 2007;178(4):2429–39.
    1. Querec TD, Akondy RS, Lee EK, Cao W, Nakaya HI, Teuwen D, et al. Systems biology approach predicts immunogenicity of the yellow fever vaccine in humans. Nature immunology. 2009;10(1):116–25. PubMed Central PMCID: PMC4049462. 10.1038/ni.1688
    1. Zaslavsky E, Hershberg U, Seto J, Pham AM, Marquez S, Duke JL, et al. Antiviral response dictated by choreographed cascade of transcription factors. Journal of immunology. 2010;184(6):2908–17. PubMed Central PMCID: PMC2856074.
    1. Rock RB, Hu S, Deshpande A, Munir S, May BJ, Baker CA, et al. Transcriptional response of human microglial cells to interferon-gamma. Genes and immunity. 2005;6(8):712–9. 10.1038/sj.gene.6364246
    1. Ceppi M, Clavarino G, Gatti E, Schmidt EK, de Gassart A, Blankenship D, et al. Ribosomal protein mRNAs are translationally-regulated during human dendritic cells activation by LPS. Immunome research. 2009;5:5 PubMed Central PMCID: PMC2788525. 10.1186/1745-7580-5-5
    1. Zhang S, Kwan P, Baum L. The potential role of CAMSAP1L1 in symptomatic epilepsy. Neuroscience letters. 2013;556:146–51. 10.1016/j.neulet.2013.10.020
    1. Guo Y, Baum LW, Sham PC, Wong V, Ng PW, Lui CH, et al. Two-stage genome-wide association study identifies variants in CAMSAP1L1 as susceptibility loci for epilepsy in Chinese. Human molecular genetics. 2012;21(5):1184–9. 10.1093/hmg/ddr550
    1. Loo YM, Fornek J, Crochet N, Bajwa G, Perwitasari O, Martinez-Sobrido L, et al. Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity. Journal of virology. 2008;82(1):335–45. PubMed Central PMCID: PMC2224404. 10.1128/JVI.01080-07
    1. Ramilo O, Allman W, Chung W, Mejias A, Ardura M, Glaser C, et al. Gene expression patterns in blood leukocytes discriminate patients with acute infections. Blood. 2007;109(5):2066–77. PubMed Central PMCID: PMC1801073. 10.1182/blood-2006-02-002477
    1. Ioannidis I, McNally B, Willette M, Peeples ME, Chaussabel D, Durbin JE, et al. Plasticity and virus specificity of the airway epithelial cell immune response during respiratory virus infection. Journal of virology. 2012;86(10):5422–36. PubMed Central PMCID: PMC3347264. 10.1128/JVI.06757-11
    1. Nakaya HI, Wrammert J, Lee EK, Racioppi L, Marie-Kunze S, Haining WN, et al. Systems biology of vaccination for seasonal influenza in humans. Nature immunology. 2011;12(8):786–95. PubMed Central PMCID: PMC3140559. 10.1038/ni.2067
    1. Nelli RK, Dunham SP, Kuchipudi SV, White GA, Baquero-Perez B, Chang P, et al. Mammalian innate resistance to highly pathogenic avian influenza H5N1 virus infection is mediated through reduced proinflammation and infectious virus release. Journal of virology. 2012;86(17):9201–10. PubMed Central PMCID: PMC3416141. 10.1128/JVI.00244-12
    1. Pothlichet J, Chignard M, Si-Tahar M. Cutting edge: innate immune response triggered by influenza A virus is negatively regulated by SOCS1 and SOCS3 through a RIG-I/IFNAR1-dependent pathway. Journal of immunology. 2008;180(4):2034–8.
    1. Gosselin EJ, Wardwell K, Rigby WF, Guyre PM. Induction of MHC class II on human polymorphonuclear neutrophils by granulocyte/macrophage colony-stimulating factor, IFN-gamma, and IL-3. Journal of immunology. 1993;151(3):1482–90.
    1. Takashima A, Yao Y. Neutrophil plasticity: acquisition of phenotype and functionality of antigen-presenting cell. J Leukoc Biol. 2015;98(4):489–96. 10.1189/jlb.1MR1014-502R
    1. Culshaw S, Millington OR, Brewer JM, McInnes IB. Murine neutrophils present Class II restricted antigen. Immunol Lett. 2008;118(1):49–54. PubMed Central PMCID: PMCPMC2430030. 10.1016/j.imlet.2008.02.008
    1. Ostanin DV, Kurmaeva E, Furr K, Bao R, Hoffman J, Berney S, et al. Acquisition of antigen-presenting functions by neutrophils isolated from mice with chronic colitis. Journal of immunology. 2012;188(3):1491–502. PubMed Central PMCID: PMCPMC3262914.
    1. Abi Abdallah DS, Egan CE, Butcher BA, Denkers EY. Mouse neutrophils are professional antigen-presenting cells programmed to instruct Th1 and Th17 T-cell differentiation. Int Immunol. 2011;23(5):317–26. PubMed Central PMCID: PMCPMC3082529. 10.1093/intimm/dxr007
    1. Kambayashi T, Laufer TM. Atypical MHC class II-expressing antigen-presenting cells: can anything replace a dendritic cell? Nature reviews Immunology. 2014;14(11):719–30. 10.1038/nri3754
    1. Calabro S, Tortoli M, Baudner BC, Pacitto A, Cortese M, O'Hagan DT, et al. Vaccine adjuvants alum and MF59 induce rapid recruitment of neutrophils and monocytes that participate in antigen transport to draining lymph nodes. Vaccine. 2011;29(9):1812–23. 10.1016/j.vaccine.2010.12.090
    1. Yang CW, Strong BS, Miller MJ, Unanue ER. Neutrophils influence the level of antigen presentation during the immune response to protein antigens in adjuvants. Journal of immunology. 2010;185(5):2927–34. PubMed Central PMCID: PMC3509756.
    1. Yan Z, DeGregori J, Shohet R, Leone G, Stillman B, Nevins JR, et al. Cdc6 is regulated by E2F and is essential for DNA replication in mammalian cells. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(7):3603–8. PubMed Central PMCID: PMC19882.
    1. Taub DD, Sayers TJ, Carter CR, Ortaldo JR. Alpha and beta chemokines induce NK cell migration and enhance NK-mediated cytolysis. Journal of immunology. 1995;155(8):3877–88.
    1. Romagnani P, Annunziato F, Lazzeri E, Cosmi L, Beltrame C, Lasagni L, et al. Interferon-inducible protein 10, monokine induced by interferon gamma, and interferon-inducible T-cell alpha chemoattractant are produced by thymic epithelial cells and attract T-cell receptor (TCR) alphabeta+ CD8+ single-positive T cells, TCRgammadelta+ T cells, and natural killer-type cells in human thymus. Blood. 2001;97(3):601–7.
    1. Inngjerdingen M, Rolstad B, Ryan JC. Activating and inhibitory Ly49 receptors modulate NK cell chemotaxis to CXC chemokine ligand (CXCL) 10 and CXCL12. Journal of immunology. 2003;171(6):2889–95.
    1. Wuest TR, Carr DJ. Dysregulation of CXCR3 signaling due to CXCL10 deficiency impairs the antiviral response to herpes simplex virus 1 infection. Journal of immunology. 2008;181(11):7985–93. PubMed Central PMCID: PMC2596651.
    1. Wennerberg E, Kremer V, Childs R, Lundqvist A. CXCL10-induced migration of adoptively transferred human natural killer cells toward solid tumors causes regression of tumor growth in vivo. Cancer immunology, immunotherapy: CII. 2015;64(2):225–35. 10.1007/s00262-014-1629-5
    1. Roncarolo MG, Bigler M, Haanen JB, Yssel H, Bacchetta R, de Vries JE, et al. Natural killer cell clones can efficiently process and present protein antigens. Journal of immunology. 1991;147(3):781–7.
    1. Hanna J, Gonen-Gross T, Fitchett J, Rowe T, Daniels M, Arnon TI, et al. Novel APC-like properties of human NK cells directly regulate T cell activation. The Journal of clinical investigation. 2004;114(11):1612–23. PubMed Central PMCID: PMC529284. 10.1172/JCI22787
    1. Nakayama M, Takeda K, Kawano M, Takai T, Ishii N, Ogasawara K. Natural killer (NK)-dendritic cell interactions generate MHC class II-dressed NK cells that regulate CD4+ T cells. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(45):18360–5. PubMed Central PMCID: PMC3215013. 10.1073/pnas.1110584108
    1. Gillard P, Chu DW, Hwang SJ, Yang PC, Thongcharoen P, Lim FS, et al. Long-term booster schedules with AS03A-adjuvanted heterologous H5N1 vaccines induces rapid and broad immune responses in Asian adults. BMC infectious diseases. 2014;14:142 PubMed Central PMCID: PMC4008266. 10.1186/1471-2334-14-142

Source: PubMed

3
Sottoscrivi