Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 210(3): 348-355, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36480273

RESUMO

Respiratory diseases are a major public health burden and a leading cause of death and disability in the world. Understanding antiviral immune responses is crucial to alleviate morbidity and mortality associated with these respiratory viral infections. Previous data from human and animal studies suggested that pre-existing atopy may provide some protection against severe disease from a respiratory viral infection. However, the mechanism(s) of protection is not understood. Low-dose LPS has been shown to drive an atopic phenotype in mice. In addition, LPS has been shown in vitro to have an antiviral effect. We examined the effect of LPS treatment on mortality to the murine parainfluenza virus Sendai virus. Low-dose LPS treatment 24 h before inoculation with a normally lethal dose of Sendai virus greatly reduced death. This protection was associated with a reduced viral titer and reduced inflammatory cytokine production in the airways. The administration of LPS was associated with a marked increase in lung neutrophils and macrophages. Depletion of neutrophils failed to reverse the protective effect of LPS; however, depletion of macrophages reversed the protective effect of LPS. Further, we demonstrate that the protective effect of LPS depends on type I IFN and TLR4-MyD88 signaling. Together, these studies demonstrate pretreatment with low-dose LPS provides a survival advantage against a severe respiratory viral infection through a macrophage-, TLR4-, and MyD88-dependent pathway.


Assuntos
Infecções por Paramyxoviridae , Viroses , Camundongos , Humanos , Animais , Lipopolissacarídeos/metabolismo , Receptor 4 Toll-Like/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Macrófagos/metabolismo , Viroses/metabolismo
2.
Methods Mol Biol ; 2506: 57-65, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35771463

RESUMO

Sendai virus (SeV), also known as the murine parainfluenza virus 1, is an enveloped negative-sense RNA paramyxovirus from the family Paramyxoviridae and genus Respirovirus. The virus was named after Sendai, city in Japan, where it was first isolated (Kuroya, Ishida, Yokohama Med Bull 4:217-233, 1953). Antigenically, SeV is closely related to human parainfluenza viruses 1 and 3. SeV is pneumotropic and naturally infects the respiratory tract of rodents. At the proper inoculum (2 × 105 pfu), SeV causes infection that is limited to the airway mucosa and inflammation mainly restricted to bronchiolar tissues as seen in asthma pathogenesis models using C57BL/6 wild-type mice (Walter et al, J Clin Invest 110:165-175, 2002). We utilize SeV to explore the mechanism(s) by which a respiratory viral infection translates into postviral airway disease in mice. This chapter primarily describes the protocols we use to infect mice in vivo, assay viral replication, and assess outcomes in the lungs of the host.


Assuntos
Asma , Infecções por Respirovirus , Animais , Asma/patologia , Bovinos , Modelos Animais de Doenças , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Respirovirus/patologia , Vírus Sendai/genética , Replicação Viral
3.
J Immunol ; 207(10): 2589-2597, 2021 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-34625522

RESUMO

Respiratory syncytial virus (RSV) infection in infancy is associated with increased risk of asthma, except in those with allergic disease at the time of infection. Using house dust mite allergen, we examined the effect of pre-existing atopy on postviral airway disease using Sendai virus in mice, which models RSV infection in humans. Sendai virus drives postviral airway disease in nonatopic mice; however, pre-existing atopy protected against the development of airway disease. This protection depended upon neutrophils, as depletion of neutrophils at the time of infection restored the susceptibility of atopic mice to postviral airway disease. Associated with development of atopy was an increase in polymorphonuclear neutrophil-dendritic cell hybrid cells that develop in Th2 conditions and demonstrated increased viral uptake. Systemic inhibition of IL-4 reversed atopic protection against postviral airway disease, suggesting that increased virus uptake by neutrophils was IL-4 dependent. Finally, human neutrophils from atopic donors were able to reduce RSV infection of human airway epithelial cells in vitro, suggesting these findings could apply to the human. Collectively our data support the idea that pre-existing atopy derives a protective neutrophil response via potential interaction with IL-4, preventing development of postviral airway disease.


Assuntos
Hipersensibilidade Imediata/imunologia , Neutrófilos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Respirovirus/imunologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vírus Sinciciais Respiratórios/imunologia , Vírus Sendai/imunologia
5.
Front Immunol ; 9: 1587, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30042764

RESUMO

Alterations in gastrointestinal microbiota indirectly modulate the risk of atopic disease, but effects on respiratory viral infections are less clear. Using the murine paramyxoviral virus type 1, Sendai virus (SeV), we examined the effect of altering gastrointestinal microbiota on the pulmonary antiviral immune response. C57BL6 mice were treated with streptomycin before or during infection with SeV and resulting immune response studied. Ingestion of the non-absorbable antibiotic streptomycin led to a marked reduction in intestinal microbial diversity without a significant effect on lung microbiota. Reduction in diversity in the gastrointestinal tract was followed by greatly increased mortality to respiratory viral infection (p < 0.0001). This increase in mortality was associated with a dysregulated immune response characterized by decreased lung (p = 0.01) and intestinal (p = 0.03) regulatory T cells (Tregs), and increased lung IFNγ (p = 0.049), IL-6 (p = 0.015), and CCL2 (p = 0.037). Adoptive transfer of Treg cells or neutralization of IFNγ prevented increased mortality. Furthermore, Lin-CD4+ cells appeared to be a potential source of the increased IFNγ. Together, these results demonstrate gastrointestinal microbiota modulate immune responses at distant mucosal sites and have the ability to significantly impact mortality in response to a respiratory viral infection.

6.
J Allergy Clin Immunol ; 142(4): 1206-1217.e5, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29269317

RESUMO

BACKGROUND: Viral respiratory tract infections increase the risk of development and exacerbation of atopic disease. Previously, we demonstrated the requirement for a neutrophil (PMN) subset expressing CD49d to drive development of postviral atopic airway disease in mice. OBJECTIVE: We sought to determine whether human CD49d+ PMNs are present in the nasal mucosa during acute viral respiratory tract infections and further characterize this PMN subset in human subjects and mice. METHODS: Sixty subjects (5-50 years old) were enrolled within 4 days of acute onset of upper respiratory symptoms. Nasal lavage for flow cytometry and nasal swabs for viral PCR were performed at enrollment and during convalescence. The Sendai virus mouse model was used to investigate the phenotype and functional relevance of CD49d+ PMNs. RESULTS: CD49d+ PMN frequency was significantly higher in nasal lavage fluid during acute respiratory symptoms in all subjects (2.9% vs 1.0%, n = 42, P < .001). In mice CD49d+ PMNs represented a "proatopic" neutrophil subset that expressed cysteinyl leukotriene receptor 1 (CysLTR1) and produced TNF, CCL2, and CCL5. Inhibition of CysLTR1 signaling in the first days of a viral respiratory tract infection was sufficient to reduce accumulation of CD49d+ PMNs in the lungs and development of postviral atopic airway disease. Similar to the mouse, human CD49d+ PMNs isolated from nasal lavage fluid during a viral respiratory tract infection expressed CysLTR1. CONCLUSION: CD49d and CysLTR1-coexpressing PMNs are present during symptoms of an acute viral respiratory tract infection in human subjects. Further study is needed to examine selective targeting of proatopic neutrophils as a potential therapeutic strategy to prevent development of postviral atopic airway disease.


Assuntos
Integrina alfa4/imunologia , Mucosa Nasal/imunologia , Neutrófilos/imunologia , Receptores de Leucotrienos/imunologia , Hipersensibilidade Respiratória/imunologia , Infecções Respiratórias/imunologia , Infecções por Respirovirus/imunologia , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mucosa Nasal/citologia , Mucosa Nasal/virologia , Hipersensibilidade Respiratória/virologia , Infecções Respiratórias/virologia , Infecções por Respirovirus/virologia , Vírus Sendai , Adulto Jovem
7.
Immun Inflamm Dis ; 5(2): 98-108, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28474501

RESUMO

INTRODUCTION: Asthma is major health burden throughout the world, and there are no therapies that have been shown to be able to prevent the development of disease. A severe respiratory paramyxoviral infection early in life has been demonstrated to greatly increase the risk of developing asthma. We have a mouse model of a severe respiratory paramyxoviral infection (Sendai virus, SeV) that mimics human disease, and requires early expression of the cytokine CCL28 to drive the development of post-viral airway disease. The known receptors for CCL28 are CCR3 and CCR10. However, it is not known if blockade of these receptors will prevent the development of post-viral airway disease. The objective of this study was to determine if treatment with a protein epitope mimetic antagonist of CCR10, POL7085, will provide sufficient protection against the development of post-viral airway disease. METHODS: C57BL6 mice were inoculated with SeV or UV inactivated SeV. From day 3-19 post inoculation (PI), mice were subcutaneously administered daily POL7085 or saline, or every other day anti-CCL28 mAb. On days 8, 10, and 12 PI bronchoalveolar cytokines, serum IgE, and lung cellular constituents were measured. At day 21 PI airway hyper-reactivity to methacholine and mucous cell metaplasia was measured. RESULTS: Treatment with either anti-CCL28 or POL7085 significantly reduced development of airway hyper-reactivity and mucous cell metaplasia following SeV infection. The prevention of post-viral airway disease was associated with early reductions in innate immune cells, but did not appear to be due to a reduction in IL-13 or IgE. CONCLUSIONS: Blockade of CCL28 or CCR10 during an acute severe respiratory paramyxoviral infection is sufficient to prevent the development of post-viral airway disease. However, the mechanism of action is unclear and requires further exploration.


Assuntos
Asma/tratamento farmacológico , Quimiocinas CC/antagonistas & inibidores , Receptores CCR10/antagonistas & inibidores , Infecções por Respirovirus/tratamento farmacológico , Vírus Sendai/imunologia , Animais , Asma/etiologia , Asma/imunologia , Asma/virologia , Quimiocinas CC/imunologia , Humanos , Camundongos , Receptores CCR10/imunologia , Infecções por Respirovirus/complicações , Infecções por Respirovirus/imunologia
8.
J Exp Med ; 204(11): 2759-69, 2007 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-17954569

RESUMO

Respiratory viral infections are associated with an increased risk of asthma, but how acute Th1 antiviral immune responses lead to chronic inflammatory Th2 disease remains undefined. We define a novel pathway that links transient viral infection to chronic lung disease with dendritic cell (DC) expression of the high-affinity IgE receptor (FcepsilonRIalpha). In a mouse model of virus-induced chronic lung disease, in which Sendai virus triggered a switch to persistent mucous cell metaplasia and airway hyperreactivity after clearance of replicating virus, we found that FceRIa(-/-) mice no longer developed mucous cell metaplasia. Viral infection induced IgE-independent, type I IFN receptor-dependent expression of FcepsilonRIalpha on mouse lung DCs. Cross-linking DC FcepsilonRIalpha resulted in the production of the T cell chemoattractant CCL28. FceRIa(-/-) mice had decreased CCL28 and recruitment of IL-13-producing CD4(+) T cells to the lung after viral infection. Transfer of wild-type DCs to FceRIa(-/-) mice restored these events, whereas blockade of CCL28 inhibited mucous cell metaplasia. Therefore, lung DC expression of FcepsilonRIalpha is part of the antiviral response that recruits CD4(+) T cells and drives mucous cell metaplasia, thus linking antiviral responses to allergic/asthmatic Th2 responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Pulmão/imunologia , Receptores de IgE/imunologia , Mucosa Respiratória/patologia , Carga Viral , Viroses/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Técnicas de Cultura de Células , Células Dendríticas/patologia , Células Dendríticas/virologia , Pulmão/patologia , Pulmão/virologia , Metaplasia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Mucosa Respiratória/virologia
9.
J Immunol ; 179(3): 1438-48, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17641009

RESUMO

Dendritic cells are ideally suited to orchestrate the innate and adaptive immune responses to infection, but we know little about how these cells respond to infection with common respiratory viruses. Paramyxoviral infections are the most frequent cause of serious respiratory illness in childhood and are associated with an increased risk of asthma. We therefore used a high-fidelity mouse model of paramyxoviral respiratory infection triggered by Sendai virus to examine the response of conventional and plasmacytoid dendritic cells (cDCs and pDCs, respectively) in the lung. We found that pDCs are scarce at baseline but become the predominant population of lung dendritic cells during infection. This recruitment allows for a source of IFN-alpha locally at the site of infection. In contrast, cDCs rapidly differentiate into myeloid cDCs and begin to migrate from the lung to draining lymph nodes within 2 h after viral inoculation. These events cause the number of lung cDCs to decrease rapidly and remain decreased at the site of viral infection. Maturation and migration of lung cDCs depends on Ccl5 and Ccr5 signals because these events are significantly impaired in Ccl5(-/-) and Ccr5(-/-) mice. cDCs failure to migrate to draining lymph nodes in Ccl5(-/-) or Ccr5(-/-) mice is associated with impaired up-regulation of CCR7 that would normally direct this process. Our results indicate that pDCs and cDCs respond distinctly to respiratory paramyxoviral infection with patterns of movement that should serve to coordinate the innate and adaptive immune responses, respectively.


Assuntos
Diferenciação Celular/imunologia , Movimento Celular/imunologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Pulmão/imunologia , Pulmão/patologia , Pneumonia Viral/imunologia , Infecções por Respirovirus/imunologia , Animais , Quimiocina CCL5 , Quimiocinas CC/deficiência , Quimiocinas CC/genética , Quimiocinas CC/fisiologia , Células Dendríticas/virologia , Terapia de Imunossupressão , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Viral/patologia , Pneumonia Viral/virologia , Receptores CCR5/deficiência , Receptores CCR5/genética , Receptores CCR5/fisiologia , Infecções por Respirovirus/patologia , Infecções por Respirovirus/virologia , Vírus Sendai/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...