The expression of B & T cell activation markers in children's tonsils following live attenuated influenza vaccine

Jack A Panapasa, Rebecca J Cox, Kristin G I Mohn, Lara A Aqrawi, Karl A Brokstad, Jack A Panapasa, Rebecca J Cox, Kristin G I Mohn, Lara A Aqrawi, Karl A Brokstad

Abstract

Live attenuated influenza vaccines (LAIV) can prevent influenza illness and death in children. The absence of known correlates of protection induced by LAIV requires human studies of underlying mechanisms of vaccine-induced immunity, to further elucidate the immunological processes occurring. In this study, children scheduled for elective tonsillectomy were enrolled in a clinical trial to evaluate the immune response to LAIV, in order to compare T and B cell gene expression profiles. Twenty-three children (aged 3-17 years) were divided into 4 groups; unvaccinated controls, or vaccinated intranasally with LAIV at days 3-4, 6-7, and 12-15 before tonsillectomy. Total RNA extraction was performed on tonsillar tissue and high RNA quality was assured. The samples were then analyzed using a validated RT2 Profiler PCR Array containing 84 gene-specific primers involved in B and T cell activation, proliferation, differentiation, regulation and polarization. The gene expression after LAIV vaccination was subsequently compared to the controls. We observed that at d 3-4 post vaccination, 6 genes were down-regulated, namely APC, CD3G, FASLG, IL7, CD8A and TLR1. Meanwhile at 6-7 days post vaccination, 9 genes were significantly up-regulated, including RIPK2, TGFB1, MICB, SOCS1, IL2RA, MS4A1, PTPRC, IL2 and IL8. By days 12-15 the genes RIPK2, IL4, IL12B and TLR2 were overexpressed. RIPK2 was upregulated at all 3 time points. Our data suggests an overall proliferation, differentiation and regulation of B and T cells in the tonsils following LAIV, where the majority of genes were up-regulated at days 6-7 and normalized by days 12-15. These findings may provide a first step into defining future biomarkers or correlates of protection after LAIV immunization.

Keywords: ASC, Antibody secreting cells; B cells; HI, haemagglutination inhibition; LAIV, Live Attenuated Influenza Virus; T cells; TIV, Trivalent Influenza Vaccine; WHO, World Health Organization; gene expression; influenza virus; live attenuated influenza vaccines (LAIV); tonsils.

Figures

Figure 1.
Figure 1.
Serum hemagglutination inhibition (HI) antibody against the H1N1 and H3N2 vaccine viruses.
Figure 2.
Figure 2.
Identifying gene clusters that are involved in B and T cell activation. The colour gradient green-black-red represents relative level of gene expression, indicating under-even-over expression, respectively. The cluster gram shows a 2-way non-supervised hierarchical clustering of the entire data set, while displaying a heat map with a horizontal and vertical dendrogram indicating co-regulated genes across groups.
Figure 3.
Figure 3.
T cell specific gene expression. T cell specific genes were classified into 6 different categories (A) regulators of T cell activation, (B) T cell proliferation, (C) T cell differentiation, (D) T cell polarization, (E) regulators of Th1 and Th2 development, and (F) Th1 and Th2 differentiation, as indicated above each plot. Each symbol represents the mean expression level (+/− range) of T cell specific genes compared to controls. The groups 1 to 3 are described in the text. The vertical scale indicates whether these genes are up- or downregulated, where positive values on the y-axis represent an up-regulation of genes, and negative values represent a down-regulation.
Figure 4.
Figure 4.
B cell specific gene expression. Genes involved in B cell activation were classified into 4 main groups, specifically (A) antigen-dependent B cell activation, (B) B cell proliferation, (C) B cell differentiation and (D) other genes involved in B cell activation. Fold change values for all the genes combined at each time point were plotted and compared to the controls. Each symbol represents the mean level (+/− range) of B cell specific gene expression. The vertical scale indicates fold change of gene expression.
Figure 5.
Figure 5.
The expression of other immune cell related genes. Genes involved in the activation of other immune cells include (A) macrophage activation, (B) neutrophil activation, (C) natural killer cell activation, and (D) leukocyte activation. Each plot represents the relative expression of the respective combined cell specific gene in the test groups compared to controls. The symbols indicate the mean gene expression level for the 3 groups (x-axis), and the range is indicated with vertical lines. The y-axis represents the fold change in gene expression levels.
Figure 6.
Figure 6.
For figure legend, see page 1669Figure 6 (See previous page). Volcano plots of individual genes (dots) that are under and overexpressed compared to baseline (Controls). The horizontal axis indicates the relative level of expression with a vertical line dividing the under expressed genes to the left (green) and over expresses to the right (red). The y-axis indicates the statistical level of the change in gene expression with a horizontal line indicating the threshold (P-value < 0.05), and genes situated above the line are denoted significant using the student t-test. (A) gene expression in Group 1 (3–4 days post vaccination) compared to controls. The green (left, n = 45) and red (right, n = 39) dots represent the under and overexpressed genes respectively. (B) gene expression in Group 2 (6–7 days post-vaccination). The red (left, n = 62) and green (right, n = 22) dots represent genes that are over and under expressed respectively. (C) The genes that are over expressed (red, n = 52) and under expressed (green, n = 32) 12–15 days following vaccination compared to controls.
Figure 7.
Figure 7.
A suggested working model for LAIV. Upon vaccination with LAIV by intranasal droplets (1), limited virus replication of the vaccine strains may occur in the nasal epithelial lining (2). Since we were not able to detect viral RNA in the tonsillar tissue, this indicates that viral antigens are transported to the tonsils by activated DCs (3) and not through local vaccine virus replication. We observed in this study that B and T cells are activated by changes in gene expression profiles, which are most pronounced at days 6–7 post vaccination (4, 5). An efflux of activated lymphocytes that may migrate to the site of infection/vaccination (6) as is supported by an earlier study. Systemic and mucosal responses (7) were detected in these subjects after LAIV vaccination [21 and unpublished data].

References

    1. Puig-Barbera J, Natividad-Sancho A, Launay O, Burtseva E, Ciblak MA, Tormos A, Buigues-Vila A, Martinez-Ubeda S, Sominina A, Group G. 2012–2013 Seasonal influenza vaccine effectiveness against influenza hospitalizations: results from the global influenza hospital surveillance network. PLoS One 2014; 9:e100497; PMID:24945510;
    1. Sokolow LZ, Naleway AL, Li DK, Shifflett P, Reynolds S, Henninger ML, Ferber JR, Odouli R, Irving SA, Thompson MG, et al.. Severity of influenza and non-influenza acute respiratory illness among pregnant women, 2010–12. Am J Obstet Gynecol 2014; 212(2):e1-11; PMID:25111585
    1. Atkinson MP, Wein LM. Quantifying the routes of transmission for pandemic influenza. Bull Math Biol 2008; 70:820–67; PMID:18278533;
    1. Strategic Advisory Group of Experts on Immunization - report of the extraordinary meeting on the influenza A (H1N1) 2009 pandemic, 7 July 2009. Wkly Epidemiol Rec 2009; 84:301–4; PMID:19630186
    1. Palache A, Oriol-Mathieu V, Abelin A, Music T, Influenza Vaccine Supply task f . Seasonal influenza vaccine dose distribution in 157 countries (2004–2011). Vaccine 2014; 32:6369–76; PMID:25442403;
    1. Krammer F, Jul-Larsen A, Margine I, Hirsh A, Sjursen H, Zambon M, Cox RJ. An H7N1 Influenza Virus Vaccine Induces Broadly Reactive Antibody Responses against H7N9 in Humans. Clin Vaccine Immunol 2014; 21:1153–63; PMID:24943383;
    1. Kang HM, Lee EK, Song BM, Jeong J, Kim HR, Choi EJ, Shin YK, Lee HS, Lee YJ. Genetic and pathogenic characteristics of H1 avian and swine influenza A viruses. J Gen Virol 2014; 95(Pt 10):2118–26; PMID:24973238;
    1. Sugaya N, Takeuchi Y. Mass vaccination of schoolchildren against influenza and its impact on the influenza-associated mortality rate among children in Japan. Clin Infect Dis 2005; 41:939–47; PMID:16142657;
    1. Finch C, Li W, Perez DR. Design of alternative live attenuated influenza virus vaccines. Curr Top Microbiol Immunol 2015; 386:205–235; PMID:25005928
    1. Coelingh KL, Luke CJ, Jin H, Talaat KR. Development of live attenuated influenza vaccines against pandemic influenza strains. Expert Rev Vaccines 2014; 13:855–71; PMID:24867587;
    1. Harvey R, Johnson RE, MacLellan-Gibson K, Robertson JS, Engelhardt OG. A promoter mutation in the haemagglutinin segment of influenza A virus generates an effective candidate live attenuated vaccine. Influenza Other Respir Viruses 2014; 8(6):605–12; PMID:25087607;
    1. Brandtzaeg P. Potential of nasopharynx-associated lymphoid tissue for vaccine responses in the airways. Am J Respir Crit Care Med 2011; 183:1595–604; PMID:21471092;
    1. Perry M, Whyte A. Immunology of the tonsils. Immunol Today 1998; 19:414–21; PMID:9745205;
    1. Palomares O, Ruckert B, Jartti T, Kucuksezer UC, Puhakka T, Gomez E, Fahrner HB, Speiser A, Jung A, Kwok WW, et al.. Induction and maintenance of allergen-specific FOXP3+ Treg cells in human tonsils as potential first-line organs of oral tolerance. J Allergy Clin Immunol 2012; 129:510–20, 520,: e511–519; PMID:22051696;
    1. Keijzer C, Haijema BJ, Meijerhof T, Voorn P, de Haan A, Leenhouts K, van Roosmalen ML, van Eden W, Broere F. Inactivated influenza vaccine adjuvanted with bacterium-like particles induce systemic and mucosal influenza A virus specific T-cell and B-cell responses after nasal administration in a TLR2 dependent fashion. Vaccine 2014; 32:2904–10; PMID:24598720;
    1. Carter NJ, Curran MP. Live attenuated influenza vaccine (FluMist(R); Fluenz): a review of its use in the prevention of seasonal influenza in children and adults. Drugs 2011; 71:1591–622; PMID:21861544;
    1. Belshe RB, Toback SL, Yi T, Ambrose CS. Efficacy of live attenuated influenza vaccine in children 6 months to 17 years of age. Influenza Other Respir Viruses 2010; 4:141–5; PMID:20409210;
    1. Jefferson TO, Rivetti D, Di Pietrantonj C, Rivetti A, Demicheli V. Vaccines for preventing influenza in healthy adults. Cochrane Database Syst Rev 2007:CD001269; PMID:17443504
    1. Jefferson T, Rivetti A, Di Pietrantonj C, Demicheli V, Ferroni E. Vaccines for preventing influenza in healthy children. Cochrane Database Syst Rev 2012; 8:CD004879; PMID:22895945
    1. Pidelaserra Marti G, Isdahl Mohn KG, Cox RJ, Brokstad KA. The influence of tonsillectomy on total serum antibody levels. Scand J Immunol 2014; 80:377–9; PMID:25039393;
    1. Mohn KG, Bredholt G, Brokstad KA, Pathirana RD, Aarstad HJ, Tøndel C, Cox RJ. Longevity of B-cell and T-cell responses after live attenuated influenza vaccination in children. J Infect Dis. 2014. November 25. 211(10):1541-9 PMID:25425696
    1. Hobson D, Curry RL, Beare AS, Ward-Gardner A. The role of serum heamagglutination inhibition antibody in protection against challenge infection with influenza A and B viruses. J Hyg 1972; 70:767–77; PMID:4509641;
    1. Bustin SA, Nolan T. Pitfalls of quantitative real-time reverse-transcription polymerase chain reaction. J Biomol Tech 2004; 15:155–66; PMID:15331581
    1. Fleige S, Pfaffl MW. RNA integrity and the effect on the real-time qRT-PCR performance. Mol Aspects Med 2006; 27:126–39; PMID:16469371;
    1. Imbeaud S, Graudens E, Boulanger V, Barlet X, Zaborski P, Eveno E, Mueller O, Schroeder A, Auffray C. Towards standardization of RNA quality assessment using user-independent classifiers of microcapillary electrophoresis traces. Nucleic Acids Res 2005; 33:e56; PMID:15800207;
    1. Halbroth BR, Heil A, Distler E, Dass M, Wagner EM, Plachter B, Probst HC, Strand D, Hartwig UF, Karner A, et al.. Superior in vitro stimulation of human CD8+ T-cells by whole virus versus split virus influenza vaccines. PLoS One 2014; 9:e103392; PMID:25072749;
    1. Wilkinson TM, Li CK, Chui CS, Huang AK, Perkins M, Liebner JC, Lambkin-Williams R, Gilbert A, Oxford J, Nicholas B, et al.. Preexisting influenza-specific CD4+ T cells correlate with disease protection against influenza challenge in humans. Nat Med 2012; 18:274–80; PMID:22286307;
    1. Sridhar S, Begom S, Bermingham A, Hoschler K, Adamson W, Carman W, Bean T, Barclay W, Deeks JJ, Lalvani A. Cellular immune correlates of protection against symptomatic pandemic influenza. Nat Med 2013; 19:1305–12; PMID:24056771;
    1. McMichael AJ, Gotch FM, Noble GR, Beare PA. Cytotoxic T-cell immunity to influenza. N Engl J Med 1983; 309:13–7; PMID:6602294;
    1. Gaur P, Singh AK, Shukla NK, Das SN. Inter-relation of Th1, Th2, Th17 and Treg cytokines in oral cancer patients and their clinical significance. Hum Immunol 2014; 75:330–7; PMID:24486578;
    1. Weaver JM, Yang H, Roumanes D, Lee FE, Wu H, Treanor JJ, Mosmann TR. Increase in IFNgamma(-)IL-2(+) cells in recent human CD4 T cell responses to 2009 pandemic H1N1 influenza. PLoS One 2013; 8:e57275; PMID:23526940;
    1. Falchetti R, Lanzilli G, Casalinuovo IA, Gaziano R, Palamara AT, Di Francesco P, Ravagnan G, Garaci E. Splenic CD4+ and CD8+ T cells from influenza immune mice concurrently produce in vitro IL2, IL4, and IFN-gamma. Cell Immunol 1996; 170:222–9; PMID:8660821;
    1. Litjens NH, Huisman M, Hijdra D, Lambrecht BM, Stittelaar KJ, Betjes MG. IL-2 producing memory CD4+ T lymphocytes are closely associated with the generation of IgG-secreting plasma cells. J Immunol 2008; 181:3665–73; PMID:18714042;
    1. Cox RJ, Madhun AS, Hauge S, Sjursen H, Major D, Kuhne M, Hoschler K, Saville M, Vogel FR, Barclay W, et al.. A phase I clinical trial of a PER.C6 cell grown influenza H7 virus vaccine. Vaccine 2009; 27:1889–97; PMID:19368768;
    1. Lessard CJ, Li H, Adrianto I, Ice JA, Rasmussen A, Grundahl KM, Kelly JA, Dozmorov MG, Miceli-Richard C, Bowman S, et al.. Variants at multiple loci implicated in both innate and adaptive immune responses are associated with Sjogren's syndrome. Nat Genet 2013; 45:1284–92; PMID:24097067;
    1. Karpala AJ, Bingham J, Schat KA, Chen LM, Donis RO, Lowenthal JW, Bean AG. Highly pathogenic (H5N1) avian influenza induces an inflammatory T helper type 1 cytokine response in the chicken. J Interferon Cytokine Res 2011; 31:393–400; PMID:21194349;
    1. Woods CW, McClain MT, Chen M, Zaas AK, Nicholson BP, Varkey J, Veldman T, Kingsmore SF, Huang Y, et al.. A host transcriptional signature for presymptomatic detection of infection in humans exposed to influenza H1N1 or H3N2. PLoS One 2013; 8(1):e52198; PMID:23326326;
    1. Zhu HT, Ru L, Guo YF. [Clinical significance of TGF-beta1 in children with primary IgA nephropathy[. Zhongguo Dang Dai Er Ke Za Zhi 2014; 16:749–53; PMID:25008886
    1. Shlomchik MJ, Weisel F. Germinal center selection and the development of memory B and plasma cells. Immunol Rev 2012; 247:52–63; PMID:22500831;
    1. Cox RJ, Brokstad KA, Zuckerman MA, Wood JM, Haaheim LR, Oxford JS. An early humoral immune response in peripheral blood following parenteral inactivated influenza vaccination. Vaccine 1994. August; 12(11):993–9; PMID:7975853;
    1. Brokstad KA, Cox RJ, Olofsson J, Jonsson R, Haaheim LR. Parenteral influenza vaccination induces a rapid systemic and local immune response. J Infect Dis 1995. Jan; 171(1):198–203; PMID:7798664;
    1. Brokstad KA, Cox RJ, Oxford JS, Haaheim LR. IgA, IgA subclasses, and secretory component levels in oral fluid collected from subjects after parental influenza vaccination.J Infect Dis 1995. April; 171(4):1072–4; PMID:7706797;
    1. Brokstad KA, Cox RJ, Eriksson JC, Olofsson J, Jonsson R, Davidsson A. High prevalence of influenza specific antibody secreting cells in nasal mucosa. Scand J Immunol 2001. Jul-Aug; 54(1–2):243–7; PMID:11439173;
    1. Brokstad KA, Eriksson JC, Cox RJ, Tynning T, Olofsson J, Jonsson R, Davidsson A. Parenteral vaccination against influenza does not induce a local antigen-specific immune response in the nasal mucosa. J Infect Dis 2002. April 1; 185(7):878–84; PMID:11920311;
    1. Eriksson JC, Davidsson A, Garberg H, Brokstad KA. Lymphocyte distribution in the tonsils prior to and after influenza vaccination. Vaccine 2003. December 8; 22(1):57–63; PMID:14604571;
    1. McCarthy JV, Ni J, Dixit VM. RIP2 is a novel NF-kappaB-activating and cell death-inducing kinase. J Biol Chem 1998; 273:16968–75; PMID:9642260;
    1. Furman D, Jojic V, Kidd B, Shen-Orr S, Price J, Jarrell J, Tse T, Huang H, Lund P, Maecker HT, et al.. Apoptosis and other immune biomarkers predict influenza vaccine responsiveness. Mol Syst Biol 2013; 9:659; PMID:23591775;
    1. Lupfer C, Thomas PG, Anand PK, Vogel P, Milasta S, Martinez J, Huang G, Green M, Kundu M, Chi H, et al.. Receptor interacting protein kinase 2-mediated mitophagy regulates inflammasome activation during virus infection. Nat Immunol 2013; 14:480–8; PMID:23525089;
    1. Mizushima N, Levine B, Cuervo AM, Klionsky DJ: Autophagy fights disease through cellular self-digestion. Nature 2008; 451:1069–75; PMID:18305538;

Source: PubMed

3
Suscribir