RESUMO
Coronavirus Disease 2019 (COVID-19) is an ongoing public health emergency and new knowledge about its immunopathogenic mechanisms is deemed necessary in the attempt to reduce the death burden, globally. For the first time in worldwide literature, we provide scientific evidence that in COVID-19 vasculitis a life-threatening escalation from type 2 T-helper immune response (humoral immunity) to type 3 hypersensitivity (immune complex disease) takes place. The subsequent deposition of immune complexes inside the vascular walls is supposed to induce a severe inflammatory state and a cytokine release syndrome, whose interleukin-6 is the key myokine, from the smooth muscle cells of blood vessels.
Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Síndrome da Liberação de Citocina/imunologia , Doenças do Complexo Imune/imunologia , Pneumonia Viral/imunologia , Síndrome Respiratória Aguda Grave/imunologia , Células Th2/imunologia , Vasculite/imunologia , Idoso , Anticorpos Antivirais/biossíntese , Complexo Antígeno-Anticorpo/biossíntese , Betacoronavirus/imunologia , Vasos Sanguíneos/imunologia , Vasos Sanguíneos/patologia , Vasos Sanguíneos/virologia , COVID-19 , Complemento C3/biossíntese , Infecções por Coronavirus/complicações , Infecções por Coronavirus/virologia , Síndrome da Liberação de Citocina/complicações , Síndrome da Liberação de Citocina/virologia , Progressão da Doença , Células Endoteliais/imunologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Humanos , Doenças do Complexo Imune/complicações , Doenças do Complexo Imune/virologia , Imunidade Humoral , Imunoglobulina G/biossíntese , Imunoglobulina M/biossíntese , Interleucina-6/biossíntese , Masculino , Pandemias , Pneumonia Viral/complicações , Pneumonia Viral/virologia , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/complicações , Síndrome Respiratória Aguda Grave/virologia , Células Th2/patologia , Células Th2/virologia , Vasculite/complicações , Vasculite/virologiaRESUMO
Severe drug eruption (SDE), a common skin disease, becomes dangerous when it occurs in patients with human immunodeficiency virus (HIV). However, the molecular mechanisms are poorly understood. Forty patients including HIV+ SDE+ (n = 15), HIV- SDE+ (n = 15) and HIV+ SDE- (n = 10) subjects were enrolled in our study. All HIV+ patients were at acquired immune deficiency syndrome (AIDS) stage. Serum levels of TNF-α, IFN-γ, IL-4, IL-13, IL-6, CXCL9, and CCL17 were quantified by ELISA. Epstein-Barr virus (EBV) and cytomegalovirus (CMV) loads were quantified by RT-qPCR. CD4, CD8, Th1, Th2, TNF-α-CD8, and IFN-γ-CD8 T cell populations were measured by flow cytometry. Levels of biochemical indexes in HIV+ SDE+ patients were significantly different from in HIV- SDE+ patients (P < .05). EBV and CMV viral loads were significantly higher in HIV+ SDE+ patients, but not in HIV- SDE+ patients (P < .05). Inflammatory cytokines TNF-α and IFN-γ were significantly elevated in HIV+ SDE+ patients (P < .05). Th2/Th1 populations and TNF-α secreting or IFN-γ secreting CD8+ T cells, were significantly up-regulated in HIV+ SDE+ patients compared to HIV- SDE+ patients (P < .05). Conversely, the CD4/CD8 ratio was significantly down-regulated in HIV+ SDE+ patients compared to HIV- SDE+ patients (P < .05). HIV infection confers distinct clinical phenotypes and immune inflammatory mechanisms in SDE. Sustained EBV and CMV activation, unbalanced Th2/Th1 and overactive CD8+ T cells mediating a pro-inflammatory response could act as distinct mechanisms in the aggravation of SDE in HIV+ SDE+ patients.
Assuntos
Linfócitos T CD8-Positivos/virologia , Citomegalovirus/patogenicidade , Toxidermias/virologia , Infecções por HIV/virologia , Herpesvirus Humano 4/patogenicidade , Células Th1/virologia , Células Th2/virologia , Ativação Viral , Adulto , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Estudos de Casos e Controles , Citocinas/sangue , Citomegalovirus/imunologia , Toxidermias/sangue , Toxidermias/imunologia , Feminino , HIV/imunologia , HIV/patogenicidade , Infecções por HIV/sangue , Infecções por HIV/imunologia , Herpesvirus Humano 4/imunologia , Interações Hospedeiro-Patógeno , Humanos , Hospedeiro Imunocomprometido , Mediadores da Inflamação/sangue , Masculino , Pessoa de Meia-Idade , Fenótipo , Índice de Gravidade de Doença , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismoRESUMO
To investigate the effect of Tanshinone IIA (TSA) on viral myocarditis (VMC). VMC animal model was established using BALB/c mice by intraperitoneally injecting Coxsackie virus B3 (CVB3). The mice were randomly divided into control group, model group, and TSA group. We detected the survival rate, the heart weight to body weight (HW/BW) ratio and hemodynamic and cardiac function parameters. The pathological features of VMC were measured through H&E staining. The expression of serum enzyme, inflammatory cytokines, and T helper (Th)1/Th2 markers was also investigated. TSA remarkably alleviated CVB3-caused myocardial injury, decreased the HW/BW ratio, and improved survival rate. TSA obviously improved hemodynamic parameters and reversed the damage to the heart pump function. Furthermore, the serum levels of lactate dehydrogenase, creatine kinase, and Th1 cytokines in the TSA group were significantly lower than those in the VMC group, and TSA treatment significantly improved the pathological condition. The interferon-gamma (IFN-γ) and interleukin-2 (IL-2) levels in VMC model group was higher than control group, and lower levels of IL-4 and IL-10 were identified. However, TSA treatment elevated IL-4 and IL-10 levels and decreased IFN-γ and IL-2 levels. TSA could effectively protect the myocardium against CVB3-induced myocarditis by the inhibition of inflammation and modulation Th1/Th2 balance in mice.
Assuntos
Abietanos/farmacologia , Anti-Inflamatórios/farmacologia , Infecções por Coxsackievirus/prevenção & controle , Enterovirus/patogenicidade , Miocardite/prevenção & controle , Miocárdio , Células Th1/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Animais , Infecções por Coxsackievirus/sangue , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/virologia , Citocinas/sangue , Modelos Animais de Doenças , Enterovirus/imunologia , Mediadores da Inflamação/sangue , Masculino , Camundongos Endogâmicos BALB C , Miocardite/sangue , Miocardite/imunologia , Miocardite/virologia , Miocárdio/imunologia , Miocárdio/metabolismo , Miocárdio/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Equilíbrio Th1-Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologiaRESUMO
UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.
Assuntos
Glutationa/deficiência , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/complicações , Síndrome de Imunodeficiência Adquirida Murina/etiologia , Infecções por Retroviridae/complicações , Células Th2/imunologia , Infecções Tumorais por Vírus/complicações , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Ativação Linfocitária , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Síndrome de Imunodeficiência Adquirida Murina/patologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Baço/imunologia , Baço/metabolismo , Baço/virologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/metabolismo , Células Th2/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologiaRESUMO
Much of our understanding of CNS immunity has been gained from models involving pathological inflammation. Attenuated rabies viruses (RABV) are unique tools to study CNS immunity in the absence of conventional inflammatory mechanisms, as they spread from the site of inoculation to the CNS transaxonally, thereby bypassing the blood-brain barrier (BBB), and are cleared without neutrophil or monocyte infiltration. To better understand the role of CD4 T cell subsets in the clearance of the virus from CNS tissues, we examined the development of antiviral immunity in wild-type (WT) and T-bet knockout mice (T-bet(-/-)), which lack Th1 cells. Early control of RABV replication in the CNS tissues of WT mice is associated with the production of IFN-γ, with antiviral effects likely mediated through the enhanced expression of type I IFNs. Of interest, IFN-α and -γ are overexpressed in the infected T-bet(-/-) by comparison with WT CNS tissues, and the initial control of RABV infection is similar. Ultimately, attenuated RABV are cleared from the CNS tissues of WT mice by Ab locally produced by the activities of infiltrating T and B cells. Although T and B cell infiltration into the CNS of infected T-bet(-/-) mice is comparable, their activities are not, the consequence being delayed, low-level Ab production and prolonged RABV replication. More importantly, neither T-bet(-/-) mice immunized with an attenuated virus, nor WT mice with Th2 RABV-specific immunity induced by immunization with inactivated virus, are protected in the long term against challenge with a pathogenic RABV.
Assuntos
Sistema Nervoso Central/imunologia , Vírus da Raiva/imunologia , Raiva/imunologia , Proteínas com Domínio T/imunologia , Animais , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/metabolismo , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos B/virologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/virologia , Citometria de Fluxo , Expressão Gênica/imunologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Interferon beta/genética , Interferon beta/imunologia , Interferon beta/metabolismo , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Raiva/metabolismo , Raiva/virologia , Vacina Antirrábica/imunologia , Vacina Antirrábica/metabolismo , Vírus da Raiva/metabolismo , Vírus da Raiva/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/deficiência , Proteínas com Domínio T/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Fatores de Tempo , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/metabolismoRESUMO
CCR5 and CXCR4, the respective cell surface coreceptors of R5 and X4 HIV-1 strains, both form heterodimers with CD4, the principal HIV-1 receptor. Using several resonance energy transfer techniques, we determined that CD4, CXCR4, and CCR5 formed heterotrimers, and that CCR5 coexpression altered the conformation of both CXCR4/CXCR4 homodimers and CD4/CXCR4 heterodimers. As a result, binding of the HIV-1 envelope protein gp120IIIB to the CD4/CXCR4/CCR5 heterooligomer was negligible, and the gp120-induced cytoskeletal rearrangements necessary for HIV-1 entry were prevented. CCR5 reduced HIV-1 envelope-induced CD4/CXCR4-mediated cell-cell fusion. In nucleofected Jurkat CD4 cells and primary human CD4(+) T cells, CCR5 expression led to a reduction in X4 HIV-1 infectivity. These findings can help to understand why X4 HIV-1 strains infection affect T-cell types differently during AIDS development and indicate that receptor oligomerization might be a target for previously unidentified therapeutic approaches for AIDS intervention.
Assuntos
Antígenos CD4/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Infecções por HIV/metabolismo , HIV-1/metabolismo , Receptores CCR5/metabolismo , Receptores CXCR4/metabolismo , Fatores de Despolimerização de Actina/metabolismo , Antígenos CD4/química , Fusão Celular , Dimerização , Citometria de Fluxo , Transferência Ressonante de Energia de Fluorescência , Células HEK293 , Humanos , Células Jurkat , Quinases Lim/metabolismo , Ligação Proteica/fisiologia , Estrutura Quaternária de Proteína , Receptores CCR5/química , Receptores CXCR4/química , Células Th1/metabolismo , Células Th1/virologia , Células Th2/metabolismo , Células Th2/virologiaRESUMO
Aspergillus fumigatus (AF) infection and sensitization are common and promote Th2 disease in individuals with asthma. Innate immune responses of bronchial epithelial cells are now known to play a key role in determination of T cell responses upon encounter with inhaled pathogens. We have recently shown that extracts of AF suppress JAK-STAT signaling in epithelial cells and thus may promote Th2 bias. To elucidate the impact of AF on human bronchial epithelial cells, we tested the hypothesis that AF can modulate the response of airway epithelial cells to favor a Th2 response and explored the molecular mechanism of the effect. Primary normal human bronchial epithelial (NHBE) cells were treated with AF extract or fractionated AF extract before stimulation with poly I:C or infection with human rhinovirus serotype 16 (HRV16). Expression of CXCL10 mRNA (real-time RT-PCR) and protein (ELISA) were measured as markers of IFN-mediated epithelial Th1-biased responses. Western blot was performed to evaluate expression of IFN regulatory factor-3 (IRF-3), NF-κB, and tyrosine-protein phosphatase nonreceptor type 11 (PTPN11), which are other markers of Th1 skewing. Knockdown experiments for protease-activated receptor-2 (PAR-2) and PTPN11 were performed to analyze the role of PAR-2 in the mechanism of suppression by AF. AF and a high-molecular-weight fraction of AF extract (HMW-AF; > 50 kD) profoundly suppressed poly I:C- and HRV16-induced expression of both CXCL10 mRNA and protein from NHBE cells via a mechanism that relied upon PAR-2 activation. Both AF extract and a specific PAR-2 activator (AC-55541) suppressed the poly I:C activation of phospho-IRF-3 without affecting activation of NF-κB. Furthermore, HMW-AF extract enhanced the expression of PTPN11, a phosphatase known to inhibit IFN signaling, and concurrently suppressed poly I:C-induced expression of both CXCL10 mRNA and protein from NHBE cells. These results show that exposure of bronchial epithelial cells to AF extract suppressed poly I:C and HRV16 signaling via a mechanism shown to involve activation of PAR-2 and PTPN11. This action of AF may promote viral disease exacerbations and may skew epithelial cells to promote Th2 inflammation in allergic airway disorders mediated or exacerbated by AF, such as asthma and chronic rhinosinusitis.
Assuntos
Aspergillus fumigatus/imunologia , Células Epiteliais/imunologia , Receptor PAR-2/imunologia , Mucosa Respiratória/imunologia , Células Th2/imunologia , Aspergillus fumigatus/metabolismo , Aspergillus fumigatus/patogenicidade , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Quimiocina CXCL10/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 3 de Interferon/metabolismo , Poli I-C/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Rhinovirus/imunologia , Rhinovirus/patogenicidade , Transdução de Sinais , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/efeitos dos fármacos , Células Th2/metabolismo , Células Th2/microbiologia , Células Th2/virologia , Fatores de Tempo , TransfecçãoRESUMO
UNLABELLED: The ability to persist long term in latently infected CD4 T cells represents a characteristic feature of HIV-1 infection and the predominant barrier to efforts aiming at viral eradication and cure. Yet, increasing evidence suggests that only small subsets of CD4 T cells with specific developmental and maturational profiles are able to effectively support HIV-1 long-term persistence. Here, we analyzed how the functional polarization of CD4 T cells shapes and structures the reservoirs of HIV-1-infected cells. We found that CD4 T cells enriched for a Th1/17 polarization had elevated susceptibilities to HIV-1 infection in ex vivo assays, harbored high levels of HIV-1 DNA in persons treated with antiretroviral therapy, and made a disproportionately increased contribution to the viral reservoir relative to their contribution to the CD4 T memory cell pool. Moreover, HIV-1 DNA levels in Th1/17 cells remained stable over many years of antiretroviral therapy, resulting in a progressively increasing contribution of these cells to the viral reservoir, and phylogenetic studies suggested preferential long-term persistence of identical viral sequences during prolonged antiretroviral treatment in this cell compartment. Together, these data suggest that Th1/17 CD4 T cells represent a preferred site for HIV-1 DNA long-term persistence in patients receiving antiretroviral therapy. IMPORTANCE: Current antiretroviral therapy is very effective in suppressing active HIV-1 replication but does not fully eliminate virally infected cells. The ability of HIV-1 to persist long term despite suppressive antiretroviral combination therapy represents a perplexing aspect of HIV-1 disease pathogenesis, since most HIV-1 target cells are activated, short-lived CD4 T cells. This study suggests that CD4 T helper cells with Th1/17 polarization have a preferential role as a long-term reservoir for HIV-1 infection during antiretroviral therapy, possibly because these cells may imitate some of the functional properties traditionally attributed to stem cells, such as the ability to persist for extremely long periods of time and to repopulate their own pool size through homeostatic self-renewal. These observations support the hypothesis that HIV-1 persistence is driven by small subsets of long-lasting stem cell-like CD4 T cells that may represent particularly promising targets for clinical strategies aiming at HIV-1 eradication and cure.
Assuntos
Fármacos Anti-HIV/uso terapêutico , Polaridade Celular/imunologia , Infecções por HIV/tratamento farmacológico , Células Th1/imunologia , Células Th17/imunologia , Latência Viral/genética , Adulto , Sequência de Bases , Contagem de Linfócito CD4 , Células Cultivadas , DNA Viral/genética , Feminino , Infecções por HIV/virologia , HIV-1/genética , HIV-1/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Sequência de DNA , Células Th1/virologia , Células Th17/virologia , Células Th2/imunologia , Células Th2/virologia , Replicação Viral/genéticaRESUMO
Noroviruses (NoV) are the most common cause of epidemic gastroenteritis world-wide. NoV infections are often asymptomatic, although individuals still shed large amounts of NoV in their stool. Understanding the differences between asymptomatic and symptomatic individuals would help in elucidating mechanisms of NoV pathogenesis. Our goal was to compare the serum cytokine responses and faecal viral RNA titres of asymptomatic and symptomatic NoV-infected individuals. We tested serum samples from infected subjects (n = 26; 19 symptomatic, seven asymptomatic) from two human challenge studies of GI.1 NoV for 16 cytokines. Samples from prechallenge and days 1-4 post-challenge were tested for these cytokines. Cytokine levels were compared to stool NoV RNA titres quantified previously by reverse transcription-polymerase chain reaction (RT-qPCR). While both symptomatic and asymptomatic groups had similar patterns of cytokine responses, the symptomatic group generally exhibited a greater elevation of T helper type 1 (Th1) and Th2 cytokines and IL-8 post-challenge compared to the asymptomatic group (all P < 0·01). Daily viral RNA titre was associated positively with daily IL-6 concentration and negatively with daily IL-12p40 concentration (all P < 0·05). Symptoms were not associated significantly with daily viral RNA titre, duration of viral shedding or cumulative shedding. Symptomatic individuals, compared to asymptomatic, have greater immune system activation, as measured by serum cytokines, but they do not have greater viral burden, as measured by titre and shedding, suggesting that symptoms may be immune-mediated in NoV infection.
Assuntos
Gastroenterite/diagnóstico , Subunidade p40 da Interleucina-12/sangue , Interleucina-6/sangue , Interleucina-8/sangue , Norovirus/imunologia , Eliminação de Partículas Virais/imunologia , Adolescente , Adulto , Doenças Assintomáticas , Fezes/química , Fezes/virologia , Feminino , Gastroenterite/imunologia , Gastroenterite/patologia , Gastroenterite/virologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Masculino , Norovirus/genética , Norovirus/crescimento & desenvolvimento , RNA Viral/genética , RNA Viral/imunologia , Índice de Gravidade de Doença , Células Th1/imunologia , Células Th1/patologia , Células Th1/virologia , Equilíbrio Th1-Th2 , Células Th2/imunologia , Células Th2/patologia , Células Th2/virologia , Carga Viral/imunologiaRESUMO
UNLABELLED: Respiratory syncytial virus (RSV) infection is the number one cause of bronchiolitis in infants, yet no vaccines are available because of a lack of knowledge of the infant immune system. Using a neonatal mouse model, we previously revealed that mice initially infected with RSV as neonates develop Th2-biased immunopathophysiologies during reinfection, and we demonstrated a role for enhanced interleukin-4 receptor α (IL-4Rα) expression on T helper cells in these responses. Here we show that RSV infection in neonates induced limited type I interferon (IFN) and plasmacytoid dendritic cell (pDC) responses. IFN alpha (IFN-α) treatment or adoptive transfer of adult pDCs capable of inducing IFN-α prior to neonatal RSV infection decreased Th2-biased immunopathogenesis during reinfection. A reduced viral load and downregulation of IL-4Rα on Th2 cells were observed in IFN-α-treated neonatal mice, suggesting dual mechanisms of action. IMPORTANCE: Respiratory syncytial virus (RSV) is the most significant cause of lower respiratory tract infection in infancy worldwide. Despite the dire need, we have failed to produce efficacious RSV vaccines or therapeutics. Part of the reason for this failure is our lack of understanding of how RSV interacts with the infant immune system to suppress the development of protective immunity. In the study described in the present paper, we used a neonatal mouse model, which more closely mimics human infants, to study the role of the innate immune system, particularly type I interferons (IFNs) and plasmacytoid dendritic cells (pDCs), in the pathogenesis of RSV infection. RSV infection in neonates induced limited type I IFN and pDC responses. IFN-α treatment or adoptive transfer of adult pDCs capable of producing IFN-α prior to neonatal RSV infection decreased Th2-biased immunopathogenesis during reinfection. These data suggest that IFN-α is a promising target for future RSV vaccine design.
Assuntos
Células Dendríticas/imunologia , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Animais , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Humanos , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Células Vero/imunologia , Células Vero/metabolismo , Células Vero/virologia , Carga Viral/imunologiaRESUMO
BACKGROUND: It is unknown why human metapneumovirus (HMPV) and respiratory syncytial virus (RSV) cause severe respiratory infection in children, particularly in premature infants. Our aim was to investigate if there are defective airway antiviral responses to these viruses in young children with history of prematurity. METHODS: Nasal airway secretions were collected from 140 children ≤ 3 y old without detectable virus (n = 80) or with PCR-confirmed HMPV or RSV infection (n = 60). Nasal protein levels of IFNγ, CCL5/RANTES, IL-10, IL-4, and IL-17 were determined using a multiplex magnetic bead immunoassay. RESULTS: Full-term children with HMPV and RSV infection had increased levels of nasal airway IFNγ, CCL5, and IL-10 along with an elevation in Th1 (IFNγ)/Th2 (IL-4) ratios, which is expected during antiviral responses. In contrast, HMPV-infected premature children (< 32 wk gestation) did not exhibit increased Th1/Th2 ratios or elevated nasal airway secretion of IFNγ, CCL5, and IL-10 relative to uninfected controls. CONCLUSION: Our study is the first to demonstrate that premature infants have defective IFNγ, CCL5/RANTES, and IL-10 airway responses during HMPV infection and provides novel insights about the potential reason why HMPV causes severe respiratory disease in children with history of prematurity.
Assuntos
Recém-Nascido Prematuro , Interferon gama/imunologia , Pulmão/imunologia , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Quimiocina CCL5/imunologia , Quimiocina CCL5/metabolismo , Pré-Escolar , Estudos Transversais , DNA Viral/genética , Feminino , Idade Gestacional , Interações Hospedeiro-Patógeno , Humanos , Lactente , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-4/imunologia , Interleucina-4/metabolismo , Pulmão/metabolismo , Pulmão/virologia , Masculino , Metapneumovirus/genética , Metapneumovirus/isolamento & purificação , Infecções por Paramyxoviridae/diagnóstico , Infecções por Paramyxoviridae/metabolismo , Infecções por Paramyxoviridae/virologia , Estudos Prospectivos , Infecções por Vírus Respiratório Sincicial/diagnóstico , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/isolamento & purificação , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th17/imunologia , Células Th17/metabolismo , Células Th17/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Regulação para CimaRESUMO
Human rhinoviruses (RVs) are a major cause of exacerbations in asthma and other chronic airway diseases. A characteristic feature of asthmatic epithelium is goblet cell metaplasia and mucus hypersecretion. Bronchial epithelium is also an important source of lipid mediators, including pro- and antiinflammatory eicosanoids. By using air-liquid interface cultures of airway epithelium from patients with asthma and nonasthmatic control subjects, we compared RV16 replication-induced changes in mRNA expression of asthma candidate genes and eicosanoid production in the epithelium with or without IL-13-induced mucus metaplasia. Mucus metaplastic epithelium was characterized by a 20-fold less effective replication of RV16 and blunted changes in gene expression; this effect was seen to the same extent in patients with asthma and control subjects. We identified ciliary cells as the main target for RV16 by immunofluorescence imaging and demonstrated that the numbers of ciliary cells decreased in RV16-infected epithelium. RV16 infection of mucociliary epithelium resulted in overexpression of genes associated with bronchial remodeling (e.g., MUC5AC, FGF2, and HBEGF), induction of cyclooxygenase-2, and increased secretion of prostaglandins. These responses were similar in both studied groups. These data indicate that structural changes associated with mucus metaplasia renders airway epithelium less susceptible to RV infection. Thus, exacerbations of the lung disease caused by RV may result from severe impairment in mucociliary clearance or activation of immune defense rather than from preferential infection of mucus metaplastic epithelium. Repeated rhinoviral infections of compromised epithelium may contribute to the remodeling of the airways.
Assuntos
Asma/imunologia , Brônquios/imunologia , Citocinas/metabolismo , Células Epiteliais/imunologia , Muco/metabolismo , Infecções por Picornaviridae/prevenção & controle , Rhinovirus/imunologia , Células Th2/imunologia , Adulto , Remodelação das Vias Aéreas , Asma/genética , Asma/patologia , Brônquios/patologia , Brônquios/virologia , Estudos de Casos e Controles , Células Cultivadas , Suscetibilidade a Doenças , Células Epiteliais/patologia , Células Epiteliais/virologia , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Masculino , Metaplasia , Pessoa de Meia-Idade , Depuração Mucociliar , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Prostaglandinas/metabolismo , RNA Mensageiro/metabolismo , Rhinovirus/crescimento & desenvolvimento , Rhinovirus/patogenicidade , Células Th2/virologia , Fatores de Tempo , Replicação ViralRESUMO
Epstein-Barr Virus (EBV) is present in the neoplastic cells of around 20-30% of patients with Hodgkin Lymphoma (HL). Although, an immunosuppressive environment is currently described in HL patients, little is known concerning the regulatory mechanism induced by EBV proteins expression in tumour cells. This study aimed to investigate an association between regulatory Type 1 cells (Tr1) and EBV tissue positivity in HL patients. Transcriptomic analysis of both EBV-positive and EBV-negative tumours showed that EBV infection increased gene expression of Tr1-related markers (ITGA2, ITGB2, LAG3) and associated-immunosuppressive cytokines (IL10). This up-regulation was associated with an over-expression of several chemokine markers known to attract T-helper type 2 (Th2) and regulatory T cells thus contributing to immune suppression. This Tr1 cells recruitment in EBV-positive HL was confirmed by immunohistochemical analysis of frozen nodes biopsies and by flow cytometric analysis of peripheral blood mononuclear cells of EBV-positive patients. Additionally, we showed that IL10 production was significantly enhanced in tumours and blood of EBV-positive HL patients. Our results propose a new model in which EBV can recruit Tr1 cells to the nodes' microenvironment, suggesting that the expression of EBV proteins in tumour cells could enable the escape of EBV-infected tumour cells from the virus-specific CTL response.
Assuntos
Infecções por Vírus Epstein-Barr/imunologia , Doença de Hodgkin/imunologia , Linfócitos T Reguladores/imunologia , Adolescente , Adulto , Idoso , Biomarcadores/metabolismo , Quimiocinas/metabolismo , Criança , Citocinas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Doença de Hodgkin/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Fenótipo , Linfócitos T Reguladores/virologia , Células Th2/imunologia , Células Th2/virologia , Regulação para Cima , Adulto JovemRESUMO
The effect of a seven-day antibiotic therapy with doxycycline was investigated on the postvaccinal humoral and cellular immune response in pigs. The selected parameters of non-specific immunity were also studied. Fifty pigs were used (control not vaccinated (C, n=10), control vaccinated (CV, n=20), and experimental - received doxycycline (DOXY, n=20)). For vaccination live-attenuated vaccine against pseudorabies (PR) was used. From day -1 to day 5 pigs from DOXY group received doxycycline orally with drinking water, at the recommended dose. Pigs from DOXY and CV groups were vaccinated at 8 and 10 weeks of age. The results of the present study showed that cell-mediated postvaccinal immune response can be modulated by oral treatment with doxycycline. Significantly lower values of stimulation index were observed after PRV restimulation in doxycycline-treated pigs. Moreover, in the DOXY group a significant decrease in IFN-γ production after PRV restimulation was noted. The significantly lower number of CD4+CD8+ cells was also observed in doxy-treated, vaccinated pigs, 2 weeks after final vaccination. Simultaneously, specific humoral response was not disturbed. This study demonstrated the importance of defining the immune modulatory activity of doxycycline because it may alter the immune responses to vaccines. The exact mechanism of T-cell response suppression by doxycycline remains to be elucidated, however the influence of doxycycline on the secretion of various cytokines, including IFN-γ, may be considered as a possible cause. The present observations should prompt further studies on the practical significance of such phenomena in terms of clinical implications.
Assuntos
Antibacterianos/farmacologia , Doxiciclina/farmacologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Vacinas contra Pseudorraiva/imunologia , Proteínas de Fase Aguda/metabolismo , Animais , Antibacterianos/administração & dosagem , Anticorpos Antivirais/sangue , Contagem de Linfócito CD4 , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Doxiciclina/administração & dosagem , Esquema de Medicação , Interações Medicamentosas , Feminino , Interferon gama/metabolismo , Interleucina-4/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Masculino , Modelos Animais , Vacinas contra Pseudorraiva/administração & dosagem , Sus scrofa , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Células Th1/virologia , Células Th2/efeitos dos fármacos , Células Th2/imunologia , Células Th2/virologia , Fatores de Tempo , Vacinação , Vacinas Atenuadas/imunologiaRESUMO
The aim of this study was to investigate the role of T-helper cell (Th)1/Th2 cytokines in the chronicity of hepatitis C virus (HCV) infection and the outcome of interferon (IFN) alpha therapy. A total of 30 patients with chronic hepatitis C were enrolled in the study. The levels of Th1/Th2 cytokines were determined. The differentiation of HCV genotypes was determined by direct sequencing. HCV RNA loads were detected by fluorescence quantitative polymerase chain reaction (qPCR). In chronic hepatitis C, the levels of interleukin (IL)-2 and transforming growth factor (TGF)-ß significantly decreased, and IL-5 and IL-18 levels increased compared with normal controls. The IL-6 serum levels were directly proportional to the serum levels of alanine aminotransferase, and were inversely proportional to the HCV RNA loading levels. Patients with severe hepatitis C had higher levels of IL-4, IL-6, and IL-1ß compared to milder cases. Patients with genotype 1 showed higher serum levels of IL-6 than those with genotype 2. The levels of IL-2 and IL-18 showed a decreasing tendency, whereas TGF-ß, IL-6, and IL-1ß showed an increasing tendency over time. There was no difference in any cytokines detected between the response and nonresponse groups before IFN therapy. However, the IFN-y level increased after IFN therapy in the response group. There was no correlation between the Th1/Th2 cytokine levels in the serum before IFN treatment and in the outcome of IFN therapy. Increasing IFN-y levels in the serum induced by IFN treatment is associated with systemic vascular resistance.
Assuntos
Antivirais/uso terapêutico , Hepacivirus/efeitos dos fármacos , Hepatite C Crônica/tratamento farmacológico , Hepatite C Crônica/patologia , Interferon-alfa/uso terapêutico , Polietilenoglicóis/uso terapêutico , Equilíbrio Th1-Th2/efeitos dos fármacos , Adulto , Alanina Transaminase/sangue , Alanina Transaminase/genética , Feminino , Expressão Gênica/efeitos dos fármacos , Genótipo , Hepacivirus/fisiologia , Hepatite C Crônica/sangue , Hepatite C Crônica/virologia , Humanos , Interferon alfa-2 , Interferon gama/sangue , Interferon gama/genética , Interleucina-18/sangue , Interleucina-18/genética , Interleucina-1beta/sangue , Interleucina-1beta/genética , Interleucina-2/sangue , Interleucina-2/genética , Interleucina-4/sangue , Interleucina-4/genética , Interleucina-5/sangue , Interleucina-5/genética , Interleucina-6/sangue , Interleucina-6/genética , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes/uso terapêutico , Células Th1/efeitos dos fármacos , Células Th1/virologia , Células Th2/efeitos dos fármacos , Células Th2/virologia , Fator de Crescimento Transformador beta/sangue , Fator de Crescimento Transformador beta/genética , Resultado do Tratamento , Carga Viral/efeitos dos fármacosRESUMO
Allergen exposure and rhinovirus infections that propagate from the upper to the lower airways are the most frequent causes of asthma exacerbation. In patients at increased risk of disease exacerbations, chronic airway inflammation is associated with the airway recruitment of circulating fibrocytes, bone marrow-derived CD34(+)CD45RO(+)CD11b(+)CD13(+)HLA-DR(+) progenitors that have antigen-presenting function and fibroblast-like properties. This study demonstrates that allergen-pulsed circulating fibrocytes from patients with allergic asthma are potent inducer of the predominant release of the T helper type (Th)2 cytokines IL-4 and IL-5 from autologous naïve and memory CD4(+) T cells. This study also provides evidence that circulating fibrocytes from allergic asthmatics are susceptible to rhinovirus infection. Infected cells release high amounts of pro-inflammatory cytokines with minimal production of IFN-α/ß. Moreover, allergen-pulsed fibrocytes support prolonged rhinovirus replication and release larger quantities of pro-inflammatory cytokines upon rhinovirus infection than unpulsed fibrocytes. Thus, fibrocytes may amplify allergen-induced, Th2 cell-driven inflammatory responses and promote further inflammation by functioning as a reservoir for rhinovirus replication in asthmatic airways. Through these mechanisms, fibrocytes may play an important role in the provocation of disease exacerbations.
Assuntos
Asma/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Infecções por Picornaviridae/imunologia , Rhinovirus/imunologia , Células Th2/imunologia , Adulto , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/patologia , Células Apresentadoras de Antígenos/virologia , Antígenos de Dermatophagoides/fisiologia , Asma/patologia , Asma/virologia , Células da Medula Óssea/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Doença Crônica , Citocinas/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Células Dendríticas/virologia , Células HeLa , Humanos , Memória Imunológica , Inflamação/imunologia , Inflamação/patologia , Inflamação/virologia , Ativação Linfocitária/imunologia , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/virologia , Infecções por Picornaviridae/patologia , Infecções por Picornaviridae/virologia , Células-Tronco/imunologia , Células-Tronco/patologia , Células-Tronco/virologia , Células Th2/patologia , Células Th2/virologiaRESUMO
Vaccines that activate humoral and cell-mediated immune responses are urgently needed for many infectious agents, including the flaviviruses dengue and West Nile (WN) virus. Vaccine development would be greatly facilitated by a new approach, in which nanoscale modules (Ag, adjuvant, and carrier) are assembled into units that are optimized for stimulating immune responses to a specific pathogen. Toward that goal, we formulated biodegradable nanoparticles loaded with Ag and surface modified with the pathogen-associated molecular pattern CpG oligodeoxynucleotides. We chose to evaluate our construct using a recombinant envelope protein Ag from the WN virus and tested the efficiency of this system in eliciting humoral and cellular responses and providing protection against the live virus. Animals immunized with this system showed robust humoral responses polarized toward Th1 immune responses compared with predominately Th2-biased responses with the adjuvant aluminum hydroxide. Immunization with CpG oligodeoxynucleotide-modified nanoparticles resulted in a greater number of circulating effector T cells and greater activity of Ag-specific lymphocytes than unmodified nanoparticles or aluminum hydroxide. Ultimately, compared with alum, this system offered superior protection in a mouse model of WN virus encephalitis.
Assuntos
Materiais Biocompatíveis/administração & dosagem , Vetores Genéticos/imunologia , Nanopartículas/administração & dosagem , Receptor Toll-Like 9/metabolismo , Febre do Nilo Ocidental/prevenção & controle , Vacinas contra o Vírus do Nilo Ocidental/administração & dosagem , Vírus do Nilo Ocidental/imunologia , Animais , Avidina/administração & dosagem , Avidina/metabolismo , Biotina/administração & dosagem , Biotina/metabolismo , Células Cultivadas , Drosophila , Marcação de Genes , Vetores Genéticos/administração & dosagem , Camundongos , Oligodesoxirribonucleotídeos/administração & dosagem , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Febre do Nilo Ocidental/imunologia , Vacinas contra o Vírus do Nilo Ocidental/imunologia , Vírus do Nilo Ocidental/genéticaRESUMO
There is limited knowledge on the identity of primary CD4(+) T cell subsets selectively targeted by HIV-1 in vivo. In this study, we established a link between HIV permissiveness, phenotype/homing potential, and lineage commitment in primary CD4(+) T cells. CCR4(+)CCR6(+), CCR4(+)CCR6(-), CXCR3(+)CCR6(+), and CXCR3(+)CCR6(-) T cells expressed cytokines and transcription factors specific for Th17, Th2, Th1Th17, and Th1 lineages, respectively. CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells expressed the HIV coreceptors CCR5 and CXCR4 and were permissive to R5 and X4 HIV replication. CCR4(+)CCR6(-) T cells expressed CXCR4 but not CCR5 and were permissive to X4 HIV only. CXCR3(+)CCR6(-) T cells expressed CCR5 and CXCR4 but were relatively resistant to R5 and X4 HIV in vitro. Total CCR6(+) T cells compared with CCR6(-) T cells harbored higher levels of integrated HIV DNA in treatment-naive HIV-infected subjects. The frequency of total CCR6(+) T cells and those of CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells were diminished in chronically infected HIV-positive subjects, despite viral-suppressive therapy. A high-throughput analysis of cytokine profiles identified CXCR3(+)CCR6(+) T cells as a major source of TNF-alpha and CCL20 and demonstrated a decreased TNF-alpha/IL-10 ratio in CXCR3(+)CCR6(-) T cells. Finally, CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells exhibited gut- and lymph node-homing potential. Thus, we identified CCR4(+)CCR6(+) and CXCR3(+)CCR6(+) T cells as highly permissive to HIV replication, with potential to infiltrate and recruit more CCR6(+) T cells into anatomic sites of viral replication. It is necessary that new therapeutic strategies against HIV interfere with viral replication/persistence in discrete CCR6(+) T cell subsets.
Assuntos
Infecções por HIV/imunologia , HIV-1/patogenicidade , Imunofenotipagem , Receptores CCR4/sangue , Receptores CCR6/sangue , Receptores CXCR3/sangue , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/virologia , Antirretrovirais/uso terapêutico , Contagem de Linfócito CD4 , Suscetibilidade a Doenças/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , HIV-1/crescimento & desenvolvimento , Imunidade Inata , Interleucina-17/biossíntese , Subpopulações de Linfócitos T/efeitos dos fármacos , Células Th1/imunologia , Células Th1/virologia , Células Th2/imunologia , Células Th2/virologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologiaRESUMO
Viral infections are able to induce autoimmune inflammation in the heart. Here, we investigated the role of virus-activated Toll-like receptor (TLR)3 and its adaptor TRIF on the development of autoimmune coxsackievirus B3 (CVB3) myocarditis in mice. Although TLR3- or TRIF-deficient mice developed similarly worse acute CVB3 myocarditis and viral replication compared to control mice, disease was significantly worse in TRIF compared to TLR3-deficient mice. Interestingly, TLR3-deficient mice developed an interleukin (IL)-4-dominant T helper (Th)2 response during acute CVB3 myocarditis with elevated markers of alternative activation, while TRIF-deficient mice elevated the Th2-associated cytokine IL-33. Treatment of TLR3-deficient mice with recombinant IL-33 improved heart function indicating that elevated IL-33 in the context of a classic Th2-driven response protects against autoimmune heart disease. We show for the first time that TLR3 versus TRIF deficiency results in different Th2 responses that uniquely influence the progression to chronic myocarditis.
Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Infecções por Coxsackievirus/imunologia , Enterovirus/fisiologia , Miocardite/imunologia , Receptor 3 Toll-Like/metabolismo , Doença Aguda , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Animais , Autoimunidade/genética , Doença Crônica , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/genética , Infecções por Coxsackievirus/fisiopatologia , Progressão da Doença , Enterovirus/patogenicidade , Regulação da Expressão Gênica/imunologia , Interleucina-33 , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-4/metabolismo , Interleucinas/genética , Interleucinas/imunologia , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/etiologia , Miocardite/genética , Miocardite/fisiopatologia , Células Th2/imunologia , Células Th2/virologia , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/imunologia , Replicação ViralRESUMO
Epidemiologic evidence suggests that cigarette smoking is a risk factor for infection with influenza, but the mechanisms underlying this susceptibility remain unknown. To ascertain if airway epithelial cells from smokers demonstrate a decreased ability to orchestrate an influenza-induced immune response, we established a model using differentiated nasal epithelial cells (NECs) from nonsmokers and smokers, co-cultured with peripheral blood monocyte-derived dendritic cells (mono-DCs) from nonsmokers. NEC/mono-DC co-cultures were infected with influenza A virus and analyzed for influenza-induced immune responses 24 hours after infection. We observed that NECs from smokers, as well as mono-DCs co-cultured with NECs from smokers, exhibited suppressed influenza-induced, interferon-related proteins interferon regulatory factor-7, Toll-like receptor-3, and retinoic acid inducible gene-1, likely because of the suppressed production of IFNα from the NECs of smokers. Furthermore, NEC/mono-DC co-cultures using NECs from smokers exhibited suppressed concentrations of T-cell/natural killer cell chemokine interferon gamma-induced protein 10 (IP-10) after infection with influenza, indicating that NECs from smokers may skew early influenza-induced Th1 responses. In contrast, NEC/mono-DC co-cultures using NEC from smokers contained increased influenza-induced concentrations of the Th2 chemokine thymic stromal lymphopoeitin (TSLP). In addition, NECs from smokers cultured alone had increased influenza-induced concentrations of the Th2 chemokine thymus and activation-regulated chemokine (TARC). Using this model, we demonstrated that in the context of infection with influenza, NECs obtained from smokers create an overall cytokine microenvironment that suppresses the interferon-mediated Th1 response and enhances the TSLP-TARC-mediated Th2 response, with the potential to modify the responses of DCs. Smoking-induced alterations in the Th1/Th2 balance may play a role in developing underlying susceptibilities to respiratory viral infections, and may also promote the likelihood of acquiring Th2 proallergic diseases.