Artigo Acesso aberto Revisado por pares

IgE cross-linking impairs monocyte antiviral responses and inhibits influenza-driven TH1 differentiation

2017; Elsevier BV; Volume: 140; Issue: 1 Linguagem: Inglês

10.1016/j.jaci.2016.11.035

ISSN

1097-6825

Autores

Regina K. Rowe, David M. Pyle, Andrew R. Tomlinson, Tinghong Lv, Zheng Hu, Michelle A. Gill,

Tópico(s)

Respiratory and Cough-Related Research

Resumo

Mechanisms underlying exacerbations of allergic asthma, although poorly defined, are clearly linked with viral infections.1Rowe R.K. Gill M.A. Asthma: the interplay between viral infections and allergic diseases.Immunol Allergy Clin North Am. 2015; 35: 115-127Abstract Full Text Full Text PDF PubMed Scopus (25) Google Scholar Rhinoviruses are the most common viruses associated with asthma exacerbations.E1Johnston S.L. Pattemore P.K. Sanderson G. Smith S. Lampe F. Josephs L. et al.Community study of role of viral infections in exacerbations of asthma in 9-11 year old children.BMJ. 1995; 310: 1225-1229Crossref PubMed Scopus (1680) Google Scholar, E2Khetsuriani N. Kazerouni N.N. Erdman D.D. Lu X. Redd S.C. Anderson L.J. et al.Prevalence of viral respiratory tract infections in children with asthma.J Allergy Clin Immunol. 2007; 119: 314-321Abstract Full Text Full Text PDF PubMed Scopus (235) Google Scholar, E3Prazma C.M. Gern J.E. Weinstein S.F. Prillaman B.A. Stempel D.A. The association between seasonal asthma exacerbations and viral respiratory infections in a pediatric population receiving inhaled corticosteroid therapy with or without long-acting beta-adrenoceptor agonist: a randomized study.Respir Med. 2015; 109: 1280-1286Abstract Full Text Full Text PDF PubMed Scopus (5) Google Scholar Influenza virus infections have a similar association,E4Johnston S.L. Ferrero F. Garcia M.L. Dutkowski R. Oral oseltamivir improves pulmonary function and reduces exacerbation frequency for influenza-infected children with asthma.Pediatr Infect Dis J. 2005; 24: 225-232Crossref PubMed Scopus (79) Google Scholar, E5Mandelcwajg A. Moulin F. Menager C. Rozenberg F. Lebon P. Gendrel D. Underestimation of influenza viral infection in childhood asthma exacerbations.J Pediatr. 2010; 157: 505-506Abstract Full Text Full Text PDF PubMed Scopus (19) Google Scholar, E6Papadopoulos N.G. Christodoulou I. Rohde G. Agache I. Almqvist C. Bruno A. et al.Viruses and bacteria in acute asthma exacerbations–a GA(2) LEN-DARE systematic review.Allergy. 2011; 66: 458-468Crossref PubMed Scopus (223) Google Scholar resulting in increased disease severity in this populationE7Dawood F.S. Kamimoto L. D'Mello T.A. Reingold A. Gershman K. Meek J. et al.Children with asthma hospitalized with seasonal or pandemic influenza, 2003-2009.Pediatrics. 2011; 128: e27-e32Crossref PubMed Scopus (62) Google Scholar, E8Jain S. Kamimoto L. Bramley A.M. Schmitz A.M. Benoit S.R. Louie J. et al.Hospitalized patients with 2009 H1N1 influenza in the United States, April-June 2009.N Engl J Med. 2009; 361: 1935-1944Crossref PubMed Scopus (1496) Google Scholar (see Table E1 in this article's Online Repository at www.jacionline.org). Defects in innate cell antiviral responses represent one potential mechanism underlying the link between viral infections and atopic disease exacerbations. For example, high-affinity IgE receptor (FcεRI) stimulation on plasmacytoid dendritic cells inhibits influenza-induced IFN-α secretion2Gill M.A. Bajwa G. George T.A. Dong C.C. Dougherty II, Jiang N. et al.Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells.J Immunol. 2010; 184: 5999-6006Crossref PubMed Scopus (238) Google Scholar and suppresses Toll-like receptor 72Gill M.A. Bajwa G. George T.A. Dong C.C. Dougherty II, Jiang N. et al.Counterregulation between the FcepsilonRI pathway and antiviral responses in human plasmacytoid dendritic cells.J Immunol. 2010; 184: 5999-6006Crossref PubMed Scopus (238) Google Scholar and Toll-like receptor 9 expression.3Schroeder J.T. Bieneman A.P. Xiao H. Chichester K.L. Vasagar K. Saini S. et al.TLR9- and FcepsilonRI-mediated responses oppose one another in plasmacytoid dendritic cells by down-regulating receptor expression.J Immunol. 2005; 175: 5724-5731Crossref PubMed Scopus (125) Google Scholar Decreasing serum IgE with omalizumab in allergic patients with asthma restores ex vivo antiviral IFN-α responses, and correlates with improved clinical outcomes.4Teach S.J. Gill M.A. Togias A. Sorkness C.A. Arbes Jr., S.J. Calatroni A. et al.Preseasonal treatment with either omalizumab or an inhaled corticosteroid boost to prevent fall asthma exacerbations.J Allergy Clin Immunol. 2015; 136: 1476-1485Abstract Full Text Full Text PDF PubMed Scopus (390) Google Scholar Monocytes are also recruited to respiratory sites during viral infectionsE17Gill M.A. Long K. Kwon T. Muniz L. Mejias A. Connolly J. et al.Differential recruitment of dendritic cells and monocytes to respiratory mucosal sites in children with influenza virus or respiratory syncytial virus infection.J Infect Dis. 2008; 198: 1667-1676Crossref PubMed Scopus (75) Google Scholar and allergen exposure,5Lee Y.G. Jeong J.J. Nyenhuis S. Berdyshev E. Chung S. Ranjan R. et al.Recruited alveolar macrophages, in response to airway epithelial-derived monocyte chemoattractant protein 1/CCl2, regulate airway inflammation and remodeling in allergic asthma.Am J Respir Cell Mol Biol. 2015; 52: 772-784Crossref PubMed Scopus (114) Google Scholar, E18Hammad H. Plantinga M. Deswarte K. Pouliot P. Willart M.A. Kool M. et al.Inflammatory dendritic cells–not basophils–are necessary and sufficient for induction of Th2 immunity to inhaled house dust mite allergen.J Exp Med. 2010; 207: 2097-2111Crossref PubMed Scopus (476) Google Scholar and implicated in food allergy development.6Zhang Y. Collier F. Naselli G. Saffery R. Tang M.L. Allen K.J. et al.Cord blood monocyte-derived inflammatory cytokines suppress IL-2 and induce nonclassic “TH2-type” immunity associated with development of food allergy.Sci Transl Med. 2016; 8: 321ra8Crossref PubMed Scopus (69) Google Scholar In addition, allergic stimulation via IgE cross-linking impacts monocyte functions including apoptosis,E19Katoh N. Kraft S. Wessendorf J.H. Bieber T. The high-affinity IgE receptor (FcepsilonRI) blocks apoptosis in normal human monocytes.J Clin Invest. 2000; 105: 183-190Crossref PubMed Scopus (89) Google Scholar cytokine secretion, and phagocytosis.7Pyle D.M. Yang V.S. Gruchalla R.S. Farrar J.D. Gill M.A. IgE cross-linking critically impairs human monocyte function by blocking phagocytosis.J Allergy Clin Immunol. 2013; 131: 491-500.e5Abstract Full Text Full Text PDF PubMed Scopus (20) Google Scholar Defining IgE-mediated effects on monocyte antiviral responses could reveal additional pathways involved in atopic disease pathogenesis. Antigen presentation, which drives T-cell activation and differentiation, represents a critical monocyte antiviral function. Influenza exposure increased monocyte-driven TH1 priming of naive CD4+ lymphocytes in primary human monocyte-T-cell cocultures, but did not significantly affect TH2 differentiation (Fig 1, A; see Fig E1 in this article's Online Repository at www.jacionline.org). Influenza-driven TH1 differentiation inversely correlated with monocyte surface FcεRI expression (Fig 1, B), a surrogate for serum IgE,E20Cheng Y.X. Foster B. Holland S.M. Klion A.D. Nutman T.B. Casale T.B. et al.CD2 identifies a monocyte subpopulation with immunoglobulin E-dependent, high-level expression of Fc epsilon RI.Clin Exp Allergy. 2006; 36: 1436-1445Crossref PubMed Scopus (25) Google Scholar suggesting that IgE-mediated stimulation may impact monocyte-directed TH1 priming during viral infection. Confirming this concept, IgE cross-linking of influenza-exposed monocytes significantly disrupted TH1 differentiation (Fig 1, C, and Fig E1), a finding consistent across all donors (Fig 1, D). Similarly, IgE cross-linking decreased IFN-γ secretion (Fig 1, E) and T-cell proliferation (Fig 1, F, and Fig E1), but had no effect on TH2 development (see Fig E2 in this article's Online Repository at www.jacionline.org). The decrease in TH1 cell stimulation was not due to IgE-mediated inhibition of influenza virus infection of monocytes because IgE cross-linking did not affect percentages of monocytes expressing the influenza proteins hemagglutinin and nucleoprotein (Fig 1, G and H). These data suggest that allergen-mediated IgE cross-linking on monocytes during concomitant viral infection could result in impaired development of virus-specific TH1 cells in sensitized individuals in vivo. We evaluated potential roles for multiple cytokines in IgE-mediated inhibition of monocyte CD4+ T lymphocyte development, including IL-12, a critical factor in TH1 differentiation (see Fig E3, Fig E4 in this article's Online Repository at www.jacionline.org). Our results indicate that IL-12, TNF-α, IL-6, and IL-10 do not regulate IgE-mediated suppression of monocyte-driven antiviral TH1 priming. Upregulation of MHC and costimulatory molecules is required for engagement and activation of the T-cell receptor complex. These signals direct CD4+ T-cell maturation; stronger antigen signal strength favors TH1 differentiation, whereas weaker signals promote TH2 or regulatory phenotypes.E21Nakayama T. Yamashita M. The TCR-mediated signaling pathways that control the direction of helper T cell differentiation.Semin Immunol. 2010; 22: 303-309Crossref PubMed Scopus (77) Google Scholar We evaluated the effect of IgE cross-linking on influenza-induced monocyte maturation. Influenza exposure significantly upregulated surface CD80, CD86, and MHC class I (HLA-ABC) and II (HLA-DR) expression, with levels significantly diminished by IgE-mediated stimulation (Fig 2, A-C). We hypothesized that IgE-mediated downregulation of HLA-DR and CD86 may disrupt monocyte-T-cell interactions. Visualization and quantitation of monocyte-T-cell interactions revealed that monocyte-T-cell clusters were significantly increased by influenza exposure, but impaired by IgE cross-linking (Fig 2, D and E). Because cell proliferation directly reflects T-cell activation, the IgE-mediated decrease in cell division (Fig 1, F) provides additional evidence that IgE cross-linking prevents monocytes from forming high-affinity interactions needed to activate multiple T cells simultaneously. These data support the model that allergic stimulation significantly impairs upregulation of proteins essential for antigen presentation, resulting in weaker T lymphocyte stimulation and diminished antiviral TH1 responses (see Fig E5 in this article's Online Repository at www.jacionline.org). IgE-mediated inhibition of virus-induced TH1 priming was remarkably conserved across all donors, and may reflect an evolutionarily maintained mechanism. Parasite-produced proteins have effects on antigen-presenting cells similar to IgE cross-linking on monocytes—downregulation of antigen presenting molecules, altered CD4 T-cell activation, and decreased TH1 responses.E22Nutman T.B. Looking beyond the induction of Th2 responses to explain immunomodulation by helminths.Parasite Immunol. 2015; 37: 304-313Crossref PubMed Scopus (62) Google Scholar, E23Everts B. Adegnika A.A. Kruize Y.C. Smits H.H. Kremsner P.G. Yazdanbakhsh M. Functional impairment of human myeloid dendritic cells during Schistosoma haematobium infection.PLoS Negl Trop Dis. 2010; 4: e667Crossref PubMed Scopus (35) Google Scholar Parasite infection can even alter viral immune responses in coinfection models.E24Actor J.K. Marshall M.A. Eltoum I.A. Buller R.M. Berzofsky J.A. Sher A. Increased susceptibility of mice infected with Schistosoma mansoni to recombinant vaccinia virus: association of viral persistence with egg granuloma formation.Eur J Immunol. 1994; 24: 3050-3056Crossref PubMed Scopus (47) Google Scholar, E25Actor J.K. Shirai M. Kullberg M.C. Buller R.M. Sher A. Berzofsky J.A. Helminth infection results in decreased virus-specific CD8+ cytotoxic T-cell and Th1 cytokine responses as well as delayed virus clearance.Proc Natl Acad Sci U S A. 1993; 90: 948-952Crossref PubMed Scopus (317) Google Scholar, E26Reese T.A. Wakeman B.S. Choi H.S. Hufford M.M. Huang S.C. Zhang X. et al.Helminth infection reactivates latent gamma-herpesvirus via cytokine competition at a viral promoter.Science. 2014; 345: 573-577Crossref PubMed Scopus (52) Google Scholar, E27Osborne L.C. Monticelli L.A. Nice T.J. Sutherland T.E. Siracusa M.C. Hepworth M.R. et al.Coinfection. Virus-helminth coinfection reveals a microbiota-independent mechanism of immunomodulation.Science. 2014; 345: 578-582Crossref PubMed Scopus (192) Google Scholar Parasite-specific IgE is protective during parasitic infectionsE28King C.L. Xianli J. Malhotra I. Liu S. Mahmoud A.A. Oettgen H.C. Mice with a targeted deletion of the IgE gene have increased worm burdens and reduced granulomatous inflammation following primary infection with Schistosoma mansoni.J Immunol. 1997; 158: 294-300PubMed Google Scholar, E29Duarte J. Deshpande P. Guiyedi V. Mecheri S. Fesel C. Cazenave P.A. et al.Total and functional parasite specific IgE responses in Plasmodium falciparum-infected patients exhibiting different clinical status.Malar J. 2007; 6: 1Crossref PubMed Scopus (56) Google Scholar; diminished TH1 responses induced by IgE cross-linking could thus be immunomodulatory in the setting of either parasitic or atopic diseases. Along these lines, many epidemiological studies reveal that children with parasitic infections have decreased atopy.E30Alcantara-Neves N.M. de S.G.B.G. Veiga R.V. Figueiredo C.A. Fiaccone R.L. da Conceicao J.S. et al.Effects of helminth co-infections on atopy, asthma and cytokine production in children living in a poor urban area in Latin America.BMC Res Notes. 2014; 7: 817Crossref PubMed Scopus (51) Google Scholar, E31Mendonca L.R. Veiga R.V. Dattoli V.C. Figueiredo C.A. Fiaccone R. Santos J. et al.Toxocara seropositivity, atopy and wheezing in children living in poor neighbourhoods in urban Latin American.PLoS Negl Trop Dis. 2012; 6: e1886Crossref PubMed Scopus (63) Google Scholar, E32Feary J. Britton J. Leonardi-Bee J. Atopy and current intestinal parasite infection: a systematic review and meta-analysis.Allergy. 2011; 66: 569-578Crossref PubMed Scopus (99) Google Scholar Parasitic infections are characterized by TH2-mediated immunity. Activation of IgE-mediated pathways by parasites, or allergen-specific IgE, could thus negatively regulate TH1 development, creating a TH2-predominant, more optimal antiparasitic response. Decreased TH1 differentiation could then dampen proinflammatory responses during parasitic (or even viral) infections, resulting in decreased inflammation and tissue damage,E33Herbert D.R. Holscher C. Mohrs M. Arendse B. Schwegmann A. Radwanska M. et al.Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology.Immunity. 2004; 20: 623-635Abstract Full Text Full Text PDF PubMed Scopus (575) Google Scholar, E34Faz-Lopez B. Morales-Montor J. Terrazas L.I. Role of macrophages in the repair process during the tissue migrating and resident helminth infections.Biomed Res Int. 2016; 2016: 8634603Crossref PubMed Scopus (26) Google Scholar, E35Baumgarth N. Kelso A. In vivo blockade of gamma interferon affects the influenza virus-induced humoral and the local cellular immune response in lung tissue.J Virol. 1996; 70: 4411-4418PubMed Google Scholar, E36Stephens R. Randolph D.A. Huang G. Holtzman M.J. Chaplin D.D. Antigen-nonspecific recruitment of Th2 cells to the lung as a mechanism for viral infection-induced allergic asthma.J Immunol. 2002; 169: 5458-5467Crossref PubMed Scopus (67) Google Scholar and providing potential benefit in allergic airway disease. Alternatively, IgE-mediated disruption of antiviral TH1 responses could promote more severe viral disease via dysregulation of normal TH1/TH2 balance and contribute to exacerbations of allergic disease. The samples in our study were predominantly from unknown donors obtained via a local blood bank; no information regarding atopic status or serum IgE levels was available. We can thus only extrapolate our findings to those with defined atopic disease. However, the range of surface FcεRI expression suggests a spectrum of IgE levels in our study participants, and the inverse correlation with influenza-induced TH1 differentiation supports the concept that elevated IgE may contribute to defective monocyte-directed antiviral TH1 responses in vivo. Consistent with our findings, others have reported blunted TH1 responses to viral infections associated with atopy. Following rhinovirus infection of patients with asthma, blood and airway CD4 T cells secreted decreased IFN-γ.8Message S.D. Laza-Stanca V. Mallia P. Parker H.L. Zhu J. Kebadze T. et al.Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production.Proc Natl Acad Sci U S A. 2008; 105: 13562-13567Crossref PubMed Scopus (391) Google Scholar Similarly, patients with atopic dermatitis had decreased virus-specific TH1 cells after transcutaneous live virus vaccination, which inversely correlated with serum IgE levels.9Slifka M.K. Leung D.Y. Hammarlund E. Raue H.P. Simpson E.L. Tofte S. et al.Transcutaneous yellow fever vaccination of subjects with or without atopic dermatitis.J Allergy Clin Immunol. 2014; 133: 439-447Abstract Full Text Full Text PDF PubMed Scopus (17) Google Scholar In future studies, it will be important to evaluate effects of IgE cross-linking on T-cell priming by monocytes exposed to other viruses, including rhinoviruses, and compare responses in atopic (high IgE) versus nonatopic individuals. To our knowledge, this is the first study demonstrating IgE-mediated impairment of human monocyte antiviral responses, via suppression of virus-induced monocyte maturation and TH1 differentiation. This extends our understanding of how allergen-mediated pathways alter host antiviral responses, highlighting a potential role for monocytes in virus-associated exacerbations of allergic disease. Further studies investigating the mechanisms underlying IgE-mediated suppression of virus-induced monocyte responses will enhance our comprehension of allergic disease pathogenesis and potentially uncover novel targets for therapeutic development. We thank the participants for donating blood, Dr Angela Mobley and the UT Southwestern Flow Cytometry Facility for assistance with flow cytometry–based assays, Dr David Farrar for providing select reagents and assistance with experimental design, and the following funding sources for supporting our research: the National Institutes of Health: the National Institute of Allergy and Infectious Diseases (grant no. R01-AI098077 to M.A.G.), Ruth L. Kirschstein National Research Service Award (grant no. T32-AI005284 to D.M.P.), the National Institute of General Medical Sciences (grant no. T32-GM008014 to D.M.P.), and the National Heart, Lung, and Blood Institute (grant no. T32-HL098040 to R.K.R. and A.R.T.); Children's Clinical Research Advisory Council (grant award to M.A.G.); and the UT Southwestern William A. and Joyce M. Sellars Distinguished Chair in Allergy and Immunology. Leukocyte-enriched blood samples were acquired predominantly from unknown donors from a local blood bank, or peripheral blood samples were obtained from known volunteers. Unknown donors were deemed healthy enough for donation by the local blood bank; neither atopic status nor serum IgE levels were obtained. Studies were approved by the UT Southwestern institutional review board (study no. STU 122010-139). Written informed consent and assent were obtained. PBS with FBS and EDTA (complete PBS [cPBS]) and complete RPMI Medium 1640 were prepared as previously described.7Pyle D.M. Yang V.S. Gruchalla R.S. Farrar J.D. Gill M.A. IgE cross-linking critically impairs human monocyte function by blocking phagocytosis.J Allergy Clin Immunol. 2013; 131: 491-500.e5Abstract Full Text Full Text PDF PubMed Scopus (20) Google Scholar Rabbit anti-human IgE (αIgE) was purchased from Bethyl Laboratories (Montgomery, Tex). Rabbit whole IgG control antibody (IgG) was purchased from Jackson ImmunoResearch (Westgrove, Pa). Unless specified, recombinant human (rh) cytokines, antihuman cytokine, and receptor antibodies were purchased from R&D Systems (Minneapolis, Minn). Monocytes were isolated from blood as previously described.7Pyle D.M. Yang V.S. Gruchalla R.S. Farrar J.D. Gill M.A. IgE cross-linking critically impairs human monocyte function by blocking phagocytosis.J Allergy Clin Immunol. 2013; 131: 491-500.e5Abstract Full Text Full Text PDF PubMed Scopus (20) Google Scholar Naive CD4+ T cells were purified using the EasySep Negative Selection Human Naive CD4+ T Cell Enrichment Kit (STEMCELL Technologies Inc, Vancouver, British Columbia, Canada) according to manufacturer recommendations. Purity of isolated cells was assessed by flow cytometry; monocytes were identified as CD14+ and enriched naive CD4+ T cells as CD3+ CD4+ CD45RA+. Samples with less than 90% purity were excluded. Monocytes were cultured in complete RPMI Medium 1640 in the presence of rh M-CSF (1 ng/mL) at a concentration of 1 × 106 to 2 × 106 cells/mL. αIgE (10 μg/mL), control rabbit IgG (10 μg/mL), or IFN-γ (20 ng/mL) was added to monocyte cultures as indicated. Influenza (Influenza A/PR/8/34 H1N1; 0.1 multiplicity of infection, Charles River Laboratories International, Inc, Wilmington, Mass) or LPS (200 ng/mL, Sigma-Aldrich, St Louis, Mo) was added to cultures. Cultures were incubated at 37°C for indicated times and cells and supernatants were harvested for analysis. Monocytes were cultured as described above in the presence or absence of αIgE, control IgG, or influenza at 37°C for 18 to 24 hours. Pretreated monocytes were mixed at a 1:1 ratio with allogeneic naive CD4+ T cells preloaded with VPD450 Proliferation Dye (BD Biosciences, San Jose, Calif) before coculture. Cells were cultured at 2 × 106 cells/mL in media containing 50 units/mL rh IL-2. For select experiments, mouse anti-human antibodies against IL-10, IL-6, IL-12, TNF-α, IL-10Rα, IL-6R, or TNFRI and IgG1 or IgG2b isotype controls were added to monocyte cultures in various combinations at 5 to 10 μg/mL each for a total mouse IgG concentration of 10 μg/mL before coculture with T cells. As indicated, rh IL-12 (1 ng/mL) was added at the initiation of the coculture. After 3 days of coculture, T lymphocytes were diluted 1:10 and cultured for an additional 4 days. T cells were then rested overnight at 37°C in media without IL-2, and subsequently stimulated with either (1) 6 hours of phorbol 12-myristate 13-acetate (50 ng/mL, Sigma, St Louis, Mo), ionomycin (1 μM, ThermoFisher, Grand Island, NY), and monensin (1×, BioLegend, San Diego, Calif) followed by cell harvesting and staining for intracellular cytokines, or (2) 24 hours on anti–CD3-coated plate (OKT3 clone, provided by David Farrar, UT Southwestern) followed by harvesting of supernatants for analysis of secreted cytokines by ELISA.E37Huber J.P. Ramos H.J. Gill M.A. Farrar J.D. Cutting edge: type I IFN reverses human Th2 commitment and stability by suppressing GATA3.J Immunol. 2010; 185: 813-817Crossref PubMed Scopus (110) Google Scholar, E38Ramos H.J. Davis A.M. George T.C. Farrar J.D. IFN-alpha is not sufficient to drive Th1 development due to lack of stable T-bet expression.J Immunol. 2007; 179: 3792-3803Crossref PubMed Scopus (36) Google Scholar The following fluorochrome-conjugated antihuman antibodies or molecules were used: CD3-AlexaFluor700, CD3-fluorescein isothiocyanate (FITC), CD4-allophycocyanin (APC), CD14-V450, CD45RA-phycoethrin (PE), CD80-FITC, CD86-PE-Cy5, FcεRI-PE, HLA-A,B,C-APC, HLA-DR-APC-Cy7, and HLA-DR-PE, CD86-biotin, and streptavidin-FITC (BD Pharmingen, San Diego, Calif). Staining and flow cytometry were performed as previously described.7Pyle D.M. Yang V.S. Gruchalla R.S. Farrar J.D. Gill M.A. IgE cross-linking critically impairs human monocyte function by blocking phagocytosis.J Allergy Clin Immunol. 2013; 131: 491-500.e5Abstract Full Text Full Text PDF PubMed Scopus (20) Google Scholar The following antibodies were used: IFN-γ-PE-Cy7 and IL-4-PE (BD Biosciences). Cells were fixed in 2% paraformaldehyde and permeabilized with 0.1% saponin in PBS to detect intracellular antigens. VPD450 Proliferation Dye was used per manufacturer's instructions to measure T-cell proliferation. Proliferated cells were defined as cells with lower VPD450 intensity than nonstimulated control T cells. Samples were acquired on a BD LSR II flow cytometer (BD Biosciences) and analyzed with the data analysis software FlowJo (FLOWJO, LLC, Ashland, Ore). Gating of cytokine-expressing cells was guided by staining negative control (unstimulated cells) and positive control T cells differentiated under TH1- or TH2-inducing conditions.E37Huber J.P. Ramos H.J. Gill M.A. Farrar J.D. Cutting edge: type I IFN reverses human Th2 commitment and stability by suppressing GATA3.J Immunol. 2010; 185: 813-817Crossref PubMed Scopus (110) Google Scholar, E38Ramos H.J. Davis A.M. George T.C. Farrar J.D. IFN-alpha is not sufficient to drive Th1 development due to lack of stable T-bet expression.J Immunol. 2007; 179: 3792-3803Crossref PubMed Scopus (36) Google Scholar Percentages of total populations that were cytokine-positive were then determined. Monocytes were isolated as above, and exposed to influenza A virus at a multiplicity of infection of 0.1 with or without IgE cross-linking or IgG control. Cells were infected for 2 hours at 37°C in complete RPMI media. Cells were washed twice with complete PBS to remove nonabsorbed virus and resuspended in complete RPMI with or without IgE cross-linking or IgG control antibody as above. At various times postinfection, cells were washed, fixed in 2% paraformaldehyde, and stained for total cell (permeablized with 0.1% saponin to detect intracellular fraction) influenza nucleoprotein (mouse antiinfluenza A virus nucleoprotein FITC, clone 431, Abcam [Cambridge, Mass]) and hemagglutinin (mouse antiinfluenza hemagglutinin H1N1, Alexa Fluor 647, clone 102, Novus Biologicals [Littleton, Colo]). Cells were analyzed by either flow cytometry on the BD LSR II (BD Biosciences), or flow cytometry imaging with the ImageStream flow cytometer (Amnis, Millipore/Sigma). Supernatants were stored at −80°C until use. The following ELISA kits were used according to manufacturer recommendations: READY-SET-GO! Human ELISA kits (eBioscience, Inc, San Diego, Calif) for IL-12p70, IFN-γ, and IL-4; ELISA Max human ELISAs for IL-10, TNF-α, and IL-6 were obtained from BioLegend. Absorbance was measured on a Biorad iMark microplate reader (Biorad, Hercules, Calif). TNF-α was also measured with the V-plex human cytokine 30-plex assay per manufacturer's instructions for increased sensitivity (Meso Scale Diagnostics, Rockville, Md). Flow cytometry imaging was performed using the Amnis ImageStream flow cytometer (Amnis). Briefly, monocytes and naive CD4 T cells were cocultured as above described. At 24-hour after coculture, cells were gently removed from the plate to preserve cell-cell interactions, pelleted at 200g, and fixed in 4% paraformaldehyde for 10 minutes. Cells were stained with anti–CD3-FITC and anti–HLA-DR-PE antibodies. Nuclei were counterstained with DRAQ5 (1:2000 dilution; Invitrogen, ThermoFisher, Grand Island, NY) per manufacturer's recommendations. Images were analyzed using the IDEAS analysis software (Amnis). CD3-HLA-DR double-positive T-cell-monocyte cell clusters were identified using a multistep gating strategy. Events were analyzed on the basis of nuclear intensity to obtain the number of nuclei per cluster. Monocyte-only staining was performed as above for surface expression with anti–HLA-DR-PE and anti–CD86-Biotin antibodies, followed by incubation with a streptavidin-FITC conjugate. Data are presented as means ± SEM. In experiments containing 3 or more conditions, 1-way or 2-way (for experiments with multiple donors) repeated-measures ANOVA and pairwise Tukey's post hoc comparisons were performed. Two monocyte-T-cell experiments (of 17 total) were eliminated from analysis because of a lack of influenza-induced TH1 response. For experiments comparing 2 conditions, paired t tests were performed with Holm-Sidak correction where appropriate. P value of less than .05 was considered significant; pertinent P values are noted in figures, significant as less than .05, less than .01, and less than .001, or more than .05 for nonsignificant values. All statistical analyses were performed using GraphPad Prism version 6.Fig E2IgE cross-linking does not alter TH2 polarization. Naive CD4 T cells polarized with monocytes exposed to no virus (mock) or influenza A virus (Flu) ± IgE cross-linking antibody (αIgE) or isotype IgG control. Graphs shown are (A) mean percentages of IL-4+ IFN-γ− cells for the indicated conditions. N = 15 donor pairs. B, Mean IL-4 concentration in monocyte-T-cell coculture supernatants. N = 5 donor pairs; error bars represent SEM, and P values represent results of 2-way ANOVA.View Large Image Figure ViewerDownload Hi-res image Download (PPT)Fig E3IL-12 is not involved in TH1 polarization by influenza-exposed monocytes. IL-12 is a critical factor in TH1 development.E21Nakayama T. Yamashita M. The TCR-mediated signaling pathways that control the direction of helper T cell differentiation.Semin Immunol. 2010; 22: 303-309Crossref PubMed Scopus (77) Google Scholar Neither blocking antibodies nor exogenous addition of IL-12 restored the IgE-mediated defect in TH1 differentiation. A, IL-12p70 production was measured in supernatants from monocytes treated ± IFN-γ for 18 hours followed by 24-hour incubation with media alone (mock), influenza A virus (Flu), or LPS. IL-12p70 was not detectable after influenza exposure, despite robust production under positive control conditions. Dashed line denotes the limit of detection. B, Monocyte-induced TH1 priming; mean % IFN-γ+ T cells in depicted conditions treated ± isotype control (black bars) or neutralizing IL-12 antibody (white bars). N = 3. C, % IFN-γ+ T cells ± rh IL-12. Representative data (of N = 3 experiments) is shown; error bars represent SEM and P values represent results of 1-way ANOVA.View Large Image Figure ViewerDownload Hi-res image Download (PPT)Fig E4IgE cross-linking suppresses TH1 priming independently of TNF-α, IL-6, and IL-10. Cytokine secretion from monocyte supernatants at 18 hours after influenza virus (Flu) ± IgE cross-linking was measured for (A) TNF-α, (B) IL-6, or (C) IL-10. These were secreted by monocytes upon IgE cross-linking,7Pyle D.M. Yang V.S. Gruchalla R.S. Farrar J.D. Gill M.A. IgE cross-linking critically impairs human monocyte function by blocking phagocytosis.J Allergy Clin Immunol. 2013; 131: 491-500.e5Abstract Full Text Full Text PDF PubMed Scopus (20) Google Scholar and in the presence of both IgE cross-linking and influenza. D-F, Neutralization of these cytokines did not impact influenza-induced TH1 priming or restore inhibition by IgE cross-linking. Mean percentages of IFN-γ+ T cells in monocyte-T-cell cocultures treated with isotype controls (black bars) or neutralizing antibodies (white bars) to (Fig E4, D) TNF-α, (Fig E4, E) IL-6, or (Fig E4, F) IL-10. Representative data from N = 3 experiments are shown; error bars represent SEM, and P values represent results of 1-way ANOVA.View Large Image Figure ViewerDownload Hi-res image Download (PPT)Fig E5Model of IgE-mediated inhibition of monocyte-driven TH1 priming. Upon monocyte influenza exposure, the normal antiviral response results in phenotypic maturation and upregulation of monocyte cell surface molecules, downstream antigen presentation, and strong TCR signal culminating in TH1 differentiation. In the setting of allergic stimulation, via IgE receptor (FcεRI) engagement, virus-induced monocyte maturation is inhibited, resulting in weak TCR signal and poor TH1 differentiation. Impaired TH1 production would have significant downstream effects, including decreased antiviral cytokine secretion (eg, IFN-γ), cytotoxic CD8+ T-cell development, and antigen-specific antibody production.View Large Image Figure ViewerDownload Hi-res image Download (PPT)Table E1Influenza virus associations with allergic disease•Influenza viruses are associated with severe disease in individuals with allergic asthma, and have been linked to exacerbations.E4Johnston S.L. Ferrero F. Garcia M.L. Dutkowski R. Oral oseltamivir improves pulmonary function and reduces exacerbation frequency for influenza-infected children with asthma.Pediatr Infect Dis J. 2005; 24: 225-232Crossref PubMed Scopus (79) Google Scholar, E5Mandelcwajg A. Moulin F. Menager C. Rozenberg F. Lebon P. Gendrel D. Underestimation of influenza viral infection in childhood asthma exacerbations.J Pediatr. 2010; 157: 505-506Abstract Full Text Full Text PDF PubMed Scopus (19) Google Scholar, E6Papadopoulos N.G. Christodoulou I. Rohde G. Agache I. Almqvist C. Bruno A. et al.Viruses and bacteria in acute asthma exacerbations–a GA(2) LEN-DARE systematic review.Allergy. 2011; 66: 458-468Crossref PubMed Scopus (223) Google Scholar•During the 2009 influenza pandemic, increased disease severity was observed in patients with asthma, with many requiring intensive level care.E7Dawood F.S. Kamimoto L. D'Mello T.A. Reingold A. Gershman K. Meek J. et al.Children with asthma hospitalized with seasonal or pandemic influenza, 2003-2009.Pediatrics. 2011; 128: e27-e32Crossref PubMed Scopus (62) Google Scholar, E8Jain S. Kamimoto L. Bramley A.M. Schmitz A.M. Benoit S.R. Louie J. et al.Hospitalized patients with 2009 H1N1 influenza in the United States, April-June 2009.N Engl J Med. 2009; 361: 1935-1944Crossref PubMed Scopus (1496) Google Scholar•However, other comorbidities besides asthma have higher rates of severe outcomes, including death, among hospitalized individuals with influenza.E10Nguyen-Van-Tam J.S. Openshaw P.J. Hashim A. Gadd E.M. Lim W.S. Semple M.G. et al.Risk factors for hospitalisation and poor outcome with pandemic A/H1N1 influenza: United Kingdom first wave (May-September 2009).Thorax. 2010; 65: 645-651Crossref PubMed Scopus (213) Google Scholar, E11Louie J.K. Acosta M. Winter K. Jean C. Gavali S. Schechter R. et al.Factors associated with death or hospitalization due to pandemic 2009 influenza A(H1N1) infection in California.JAMA. 2009; 302: 1896-1902Crossref PubMed Scopus (864) Google Scholar, E9Myles P. Nguyen-Van-Tam J.S. Semple M.G. Brett S.J. Bannister B. Read R.C. et al.Differences between asthmatics and nonasthmatics hospitalised with influenza A infection.Eur Respir J. 2013; 41: 824-831Crossref PubMed Scopus (39) Google Scholar•The Centers for Disease Control and Prevention reports that asthma is the most common underlying comorbidity in children and in the top 5 for adults hospitalized with influenza.E12Centers for Disease Control and Prevention. CDC FluNet. Available at: http://gis.cdc.gov/grasp/fluview/FluHospChars.html. September 2016. Accessed September 9, 2016.Google Scholar•More severe infections have been noted in atopic vs nonatopic children in general.E13Kim Y.J. Ryu S.L. Jung S.H. Shim J.W. Kim D.S. Jung H.L. et al.Increased prevalence of H1N1-induced severe lower respiratory tract diseases in children with atopic sensitization.Allergy Asthma Immunol Res. 2012; 4: 277-283Crossref PubMed Scopus (9) Google Scholar•Influenza is also isolated more frequently from adults with allergic rhinitis compared with healthy controls.E14Kim J.H. Moon B.J. Gong C.H. Kim N.H. Jang Y.J. Detection of respiratory viruses in adult patients with perennial allergic rhinitis.Ann Allergy Asthma Immunol. 2013; 111: 508-511Abstract Full Text Full Text PDF PubMed Scopus (14) Google Scholar•Murine models of allergic sensitization demonstrate enhanced allergic phenotypes after influenza infection.E15Suzuki S. Suzuki Y. Yamamoto N. Matsumoto Y. Shirai A. Okubo T. Influenza A virus infection increases IgE production and airway responsiveness in aerosolized antigen-exposed mice.J Allergy Clin Immunol. 1998; 102: 732-740Abstract Full Text Full Text PDF PubMed Scopus (50) Google Scholar, E16Yamamoto N. Suzuki S. Shirai A. Suzuki M. Nakazawa M. Nagashima Y. et al.Dendritic cells are associated with augmentation of antigen sensitization by influenza A virus infection in mice.Eur J Immunol. 2000; 30: 316-326Crossref PubMed Scopus (53) Google ScholarInvestigating IgE-mediated effects on antiviral immune responses to influenza virus may shed light on key mechanisms modulating allergen-virus interactions. Open table in a new tab

Referência(s)