Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Eur J Immunol ; 44(2): 397-408, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24222381

RESUMO

Recent evidence suggests that an individual's unique history and sequence of exposures to pathogens and antigens may dictate downstream immune responses to disparate antigens. We show that the i.n. delivery of nonreplicative virus-like particles (VLPs), which bear structural but no antigenic similarities to respiratory pathogens, acts to prime the lungs of both C56BL/6 and BALB/c mice, facilitating heightened and accelerated primary immune responses to high-dose influenza challenge, thus providing a nonpathogenic model of innate imprinting. These responses correspond closely to those observed following natural infection with the opportunistic fungus, Pneumocystis murina, and are characterized by accelerated antigen processing by DCs and alveolar macrophages, an enhanced influx of cells to the local tracheobronchial lymph node, and early upregulation of T-cell co-stimulatory/adhesion molecules. CD11c⁺ cells, which have been directly exposed to VLPs or Pneumocystis are necessary in facilitating enhanced clearance of influenza virus, and the repopulation of the lung by Ly-6C⁺ precursors relies on CCR2 expression. Thus, immune imprinting 72 h after VLP-priming, or 2 weeks after Pneumocystis-priming is CCR2-mediated and results from the enhanced antigen processing, maturation, and trafficking abilities of DCs and alveolar macrophages, which cause accelerated influenza-specific primary immune responses and result in superior viral clearance.


Assuntos
Antígenos/imunologia , Antígeno CD11c/imunologia , Imunidade Inata/imunologia , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/imunologia , Animais , Apresentação de Antígeno/imunologia , Antígenos Ly/imunologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Células Dendríticas/virologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/virologia , Macrófagos Alveolares/imunologia , Macrófagos Alveolares/microbiologia , Macrófagos Alveolares/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Pneumocystis/imunologia , Infecções por Pneumocystis/imunologia , Receptores CCR2/imunologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Linfócitos T/virologia , Regulação para Cima/imunologia , Vacinas de Partículas Semelhantes a Vírus/imunologia
2.
Eur J Immunol ; 44(11): 3263-72, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25091976

RESUMO

Superinfection in mice at day 7 postinfluenza infection exacerbates bacterial pneumonia at least in part via downstream effects of increased IFN-γ signaling. Here we show that up to 3 days postinfluenza infection, mice have reduced susceptibility to superinfection with methicillin-resistant Staphylococcus aureus (MRSA), but that superinfection during that time exacerbated influenza disease. This was due to IL-13 signaling that was advantageous for resolving MRSA infection via inhibition of IFN-γ, but was detrimental to the clearance of influenza virus. However, if superinfection did not occur until the near resolution of influenza infection (day 7), IL-13 signaling was inhibited, at least in part by upregulation of IL-13 decoy receptor (IL-13Rα2), which in turn caused increases in IFN-γ signaling and exacerbation of bacterial infection. Understanding these cytokine sequelae is critical to development of immunotherapies for influenza-MRSA coinfection since perturbations of these sequelae at the wrong time could increase susceptibility to MRSA and/or influenza.


Assuntos
Interferon gama/genética , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Interleucina-13/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções Estafilocócicas/imunologia , Superinfecção/imunologia , Animais , Carga Bacteriana , Coinfecção , Suscetibilidade a Doenças , Interleucina-13/genética , Interleucina-13/farmacologia , Subunidade alfa2 de Receptor de Interleucina-13/antagonistas & inibidores , Subunidade alfa2 de Receptor de Interleucina-13/biossíntese , Pulmão/microbiologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Bacteriana/imunologia , Proteínas Recombinantes/farmacologia , Superinfecção/microbiologia , Carga Viral
4.
mBio ; 8(6)2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29138299

RESUMO

Although viruses and viral capsids induce rapid immune responses, little is known about viral pathogen-associated molecular patterns (PAMPs) that are exhibited on their surface. Here, we demonstrate that the repeating protein subunit pattern common to most virus capsids is a molecular pattern that induces a Toll-like-receptor-2 (TLR2)-dependent antiviral immune response. This early antiviral immune response regulates the clearance of subsequent bacterial superinfections, which are a primary cause of morbidities associated with influenza virus infections. Utilizing this altered susceptibility to subsequent bacterial challenge as an outcome, we determined that multiple unrelated, empty, and replication-deficient capsids initiated early TLR2-dependent immune responses, similar to intact influenza virus or murine pneumovirus. These TLR2-mediated responses driven by the capsid were not dependent upon the capsid's shape, size, origin, or amino acid sequence. However, they were dependent upon the multisubunit arrangement of the capsid proteins, because unlike intact capsids, individual capsid subunits did not enhance bacterial clearance. Further, we demonstrated that even a linear microfilament protein built from repeating protein subunits (F-actin), but not its monomer (G-actin), induced similar kinetics of subsequent bacterial clearance as did virus capsid. However, although capsids and F-actin induced similar bacterial clearance, in macrophages they required distinct TLR2 heterodimers for this response (TLR2/6 or TLR2/1, respectively) and different phagocyte populations were involved in the execution of these responses in vivo Our results demonstrate that TLR2 responds to invading viral particles that are composed of repeating protein subunits, indicating that this common architecture of virus capsids is a previously unrecognized molecular pattern.IMPORTANCE Rapid and precise pathogen identification is critical for the initiation of pathogen-specific immune responses and pathogen clearance. Bacteria and fungi express common molecular patterns on their exteriors that are recognized by cell surface-expressed host pattern recognition receptors (PRRs) prior to infection. In contrast, viral molecular patterns are primarily nucleic acids, which are recognized after virus internalization. We found that an initial antiviral immune response is induced by the repeating subunit pattern of virus exteriors (capsids), and thus, induction of this response is independent of viral infection. This early response to viral capsids required the cell surface-expressed PRR TLR2 and allowed for improved clearance of subsequent bacterial infection that commonly complicates respiratory viral infections. Since the repeating protein subunit pattern is conserved across viral capsids, this suggests that it is not easy for a virus to change without altering fitness. Targeting this vulnerability could lead to development of a universal antiviral vaccine.


Assuntos
Bactérias/imunologia , Proteínas do Capsídeo/imunologia , Capsídeo/imunologia , Imunidade Inata , Moléculas com Motivos Associados a Patógenos , Receptor 2 Toll-Like/metabolismo , Vírus/imunologia , Animais , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Viroses/imunologia
5.
mBio ; 7(3)2016 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-27143388

RESUMO

UNLABELLED: Bacterial superinfections are a primary cause of death during influenza pandemics and epidemics. Type I interferon (IFN) signaling contributes to increased susceptibility of mice to bacterial superinfection around day 7 post-influenza A virus (IAV) infection. Here we demonstrate that the reduced susceptibility to methicillin-resistant Staphylococcus aureus (MRSA) at day 3 post-IAV infection, which we previously reported was due to interleukin-13 (IL-13)/IFN-γ responses, is also dependent on type I IFN signaling and its subsequent requirement for protective IL-13 production. We found, through utilization of blocking antibodies, that reduced susceptibility to MRSA at day 3 post-IAV infection was IFN-ß dependent, whereas the increased susceptibility at day 7 was IFN-α dependent. IFN-ß signaling early in IAV infection was required for MRSA clearance, whereas IFN-α signaling late in infection was not, though it did mediate increased susceptibility to MRSA at that time. Type I IFN receptor (IFNAR) signaling in CD11c(+) and Ly6G(+) cells was required for the observed reduced susceptibility at day 3 post-IAV infection. Depletion of Ly6G(+) cells in mice in which IFNAR signaling was either blocked or deleted indicated that Ly6G(+) cells were responsible for the IFNAR signaling-dependent susceptibility to MRSA superinfection at day 7 post-IAV infection. Thus, during IAV infection, the temporal differences in type I IFN signaling increased bactericidal activity of both CD11c(+) and Ly6G(+) cells at day 3 and reduced effector function of Ly6G(+) cells at day 7. The temporal differential outcomes induced by IFN-ß (day 3) and IFN-α (day 7) signaling through the same IFNAR resulted in differential susceptibility to MRSA at 3 and 7 days post-IAV infection. IMPORTANCE: Approximately 114,000 hospitalizations and 40,000 annual deaths in the United States are associated with influenza A virus (IAV) infections. Frequently, these deaths are due to community-acquired Gram-positive bacterial species, many of which show increasing resistance to antibiotic therapy. Severe complications, including parapneumonic empyema and necrotizing pneumonia, can arise, depending on virulence factors expressed by either the virus or bacteria. Unfortunately, we are unable to control the expression of these virulence factors, making host responses a logical target for therapeutic interventions. Moreover, interactions between virus, host, and bacteria that exacerbate IAV-related morbidities and mortalities are largely unknown. Here, we show that type I interferon (IFN) expression can modulate susceptibility to methicillin-resistant Staphylococcus aureus (MRSA) infection, with IFN-ß reducing host susceptibility to MRSA infection while IFN-α increases susceptibility. Our data indicate that treatments designed to augment IFN-ß and/or inhibit IFN-α production around day 7 post-IAV infection could reduce susceptibility to deadly superinfections.


Assuntos
Suscetibilidade a Doenças , Influenza Humana/complicações , Interferon Tipo I/metabolismo , Leucócitos/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Infecções Estafilocócicas/imunologia , Superinfecção/imunologia , Animais , Antígenos Ly/análise , Antígeno CD11c/análise , Humanos , Influenza Humana/imunologia , Interleucina-13/metabolismo , Leucócitos/química , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA