Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(11): e81695, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24303065

RESUMEN

Innate immune responses elicited upon virus exposure are crucial for the effective eradication of viruses, the onset of adaptive immune responses and for establishing proper immune memory. Respiratory syncytial virus (RSV) is responsible for a high disease burden in neonates and immune compromised individuals, causing severe lower respiratory tract infections. During primary infections exuberant innate immune responses may contribute to disease severity. Furthermore, immune memory is often insufficient to protect during RSV re-exposure, which results in frequent symptomatic reinfections. Therefore, identifying the cell types and pattern recognition receptors (PRRs) involved in RSV-specific innate immune responses is necessary to understand incomplete immunity against RSV. We investigated the innate cellular response triggered upon infection of epithelial cells and peripheral blood mononuclear cells. We show that CD14(+) myeloid cells and epithelial cells are the major source of IL-8 and inflammatory cytokines, IL-6 and TNF-α, when exposed to live RSV Three routes of RSV-induced IFN-α production can be distinguished that depend on the cross-talk of different cell types and the presence or absence of virus specific antibodies, whereby pDC are the ultimate source of IFN-α. RSV-specific antibodies facilitate direct TLR7 access into endosomal compartments, while in the absence of antibodies, infection of monocytes or epithelial cells is necessary to provide an early source of type I interferons, required to engage the IFN-α,ß receptor (IFNAR)-mediated pathway of IFN-α production by pDC. However, at high pDC density infection with RSV causes IFN-α production without the need for a second party cell. Our study shows that cellular context and immune status are factors affecting innate immune responses to RSV. These issues should therefore be addressed during the process of vaccine development and other interventions for RSV disease.


Asunto(s)
Anticuerpos Antivirales/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interferón Tipo I/biosíntesis , Monocitos/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Virus Sincitial Respiratorio Humano/inmunología , Especificidad de Anticuerpos/inmunología , Comunicación Celular , Técnicas de Cultivo de Célula , Línea Celular , Citocinas/biosíntesis , Humanos , Inmunidad Innata , Mediadores de Inflamación , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Receptores de Lipopolisacáridos/metabolismo , Monocitos/metabolismo , Monocitos/virología , Receptores de Interferón/metabolismo , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Transducción de Señal , Receptor Toll-Like 7/metabolismo , Acoplamiento Viral
2.
J Virol ; 87(13): 7550-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23637394

RESUMEN

Infants are protected from a severe respiratory syncytial virus (RSV) infection in the first months of life by maternal antibodies or by prophylactically administered neutralizing antibodies. Efforts are under way to produce RSV-specific antibodies with increased neutralizing capacity compared to the currently licensed palivizumab. While clearly beneficial during primary infections, preexisting antibodies might affect the onset of adaptive immune responses and the ability to resist subsequent RSV infections. Therefore, we addressed the question of how virus neutralizing antibodies influence the priming of subsequent adaptive immune responses. To test a possible role of the neonatal Fc receptor (FcRn) in this process, we compared the responses in C57BL/6 wild-type (WT) and FcRn(-/-) mice. We observed substantial virus-specific T-cell priming and B-cell responses in mice primed with RSV IgG immune complexes resulting in predominantly Th1-type CD4(+) T-cell and IgG2c antibody responses upon live-virus challenge. RSV-specific CD8(+) T cells were primed as well. Activation of these adaptive immune responses was independent of FcRn. Thus, neutralizing antibodies that localize to the airways and prevent infection-related routes of antigen processing can still facilitate antigen presentation of neutralized virus particles and initiate adaptive immune responses against RSV.


Asunto(s)
Inmunidad Adaptativa/inmunología , Anticuerpos Neutralizantes/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Virión/inmunología , Administración Intranasal , Análisis de Varianza , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/genética , Citocinas/inmunología , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Antígenos de Histocompatibilidad Clase I/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Picratos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Fc/genética , Virus Sincitiales Respiratorios/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virión/genética
3.
J Virol ; 86(21): 11472-82, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22896622

RESUMEN

Breast feeding reduces the risk of developing severe respiratory syncytial virus (RSV) infections in infants. In addition to maternal antibodies, other immune-modulating factors in human milk contribute to this protection. Specific dietary prebiotic oligosaccharides, similar to oligosaccharides present in human milk, were evaluated in a C57BL/6 mouse RSV infection model. During primary RSV infection, increased numbers of RSV-specific CD4(+) T cells producing gamma interferon (IFN-γ) were found in the lungs at days 8 to 10 postinfection in mice receiving diet containing short-chain galactooligosacharides, long-chain fructooligosaccharides, and pectin-derived acidic oligosaccharides (termed scGOS/lcFOS/pAOS). In a Th2-skewed formalin-inactivated (FI)-RSV vaccination model, the prebiotic diet reduced RSV-specific Th2 cytokine (interleukin-4 [IL-4], IL-5, and IL-13)-producing CD4(+) T cells in the lung and the magnitude of airway eosinophilia at day 4 and 6 after infection. This was accompanied by a decreased influx of inflammatory dendritic cells (CD11b(+)/CD11c(+)) and increased numbers of IFN-γ-producing CD4(+) and CD8(+) T cells at day 8 after viral challenge. These findings suggest that specific dietary oligosaccharides can influence trafficking and/or effector functions of innate immune, CD4(+), and CD8(+) T cell subsets in the lungs of RSV-infected mice. In our models, scGOS/lcFOS/pAOS had no effect on weight but increased viral clearance in FI-RSV-vaccinated mice 8 days after infection. The increased systemic Th1 responses potentiated by scGOS/lcFOS/pAOS might contribute to an accelerated Th1/Th2 shift of the neonatal immune system, which might favor protective immunity against viral infections with a high attack rate in early infancy, such as RSV.


Asunto(s)
Dieta/métodos , Factores Inmunológicos/administración & dosificación , Oligosacáridos/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Células TH1/inmunología , Animales , Peso Corporal , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra Citomegalovirus/administración & dosificación , Vacunas contra Citomegalovirus/inmunología , Modelos Animales de Enfermedad , Femenino , Interferón gamma/metabolismo , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/patogenicidad
4.
Eur Respir J ; 40(6): 1508-15, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22496321

RESUMEN

Respiratory syncytial virus (RSV) bronchiolitis causes severe respiratory tract infection in infants, frequently necessitating mechanical ventilatory support. However, life-saving, mechanical ventilation aggravates lung inflammation. We set up a model to dissect the host molecular response to mechanical ventilation in RSV infection. Furthermore, the response to induced hypercapnic acidosis, reported to dampen the inflammatory response to mechanical ventilation in non-infectious models, was assessed. BALB/c mice were inoculated with RSV or mock-suspension and ventilated for 5 h on day 5 post inoculation. Mechanical ventilation of infected mice resulted in enhanced cellular influx and increased concentrations of pro-inflammatory cytokines in the bronchoalveolar space. Microarray analysis showed that enhanced inflammation was associated with a molecular signature of a stress response to mechanical ventilation with little effect on the virus-induced innate immune response. Hypercapnic acidosis during mechanical ventilation of infected mice did not change host transcript profiles. We conclude that mechanical ventilation during RSV infection adds a robust but distinct molecular stress response to virus-induced innate immunity activation, emphasising the importance of lung-protective mechanical ventilation strategies. Induced hypercapnic acidosis has no major effect on host transcription profiles during mechanical ventilation for RSV infection, suggesting that this is a safe approach to minimise ventilator-induced lung injury.


Asunto(s)
Respiración Artificial/métodos , Infecciones por Virus Sincitial Respiratorio/terapia , Acidosis/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Análisis por Conglomerados , Hemodinámica , Hipercapnia/metabolismo , Inflamación , Masculino , Ratones , Ratones Endogámicos BALB C , Virus Sincitiales Respiratorios/inmunología , Virus Sincitiales Respiratorios/fisiología , Factores de Tiempo , Lesión Pulmonar Inducida por Ventilación Mecánica/diagnóstico
5.
Vaccine ; 29(15): 2730-41, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21316502

RESUMEN

Inactivated respiratory syncytial virus (RSV) vaccines tend to predispose for immune mediated enhanced disease, characterized by Th2 responses and airway hypersensitivity reactions. We show in a C57BL/6 mouse model that the early innate response elicited by the challenge virus (RSV versus influenza virus) influences the outcome of the Th1/Th2 balance in the lung after intramuscular priming with inactivated vaccine. Priming of CD4(+)/IFN-γ(+) T cells by mature dendritic cells administered intravenously and/or priming of a virus specific CD8(+) T cell response ameliorated the Th2-mediated inflammatory response in the lung, suggesting that vaccination procedures are feasible that prevent vaccine induced immune pathology.


Asunto(s)
Pulmón/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Inmunidad Adaptativa , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Neumonía Viral/inmunología , Neumonía Viral/patología , Células Th2/inmunología , Vacunas de Productos Inactivados/inmunología
6.
J Immunol ; 185(11): 6489-98, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20971927

RESUMEN

Following infection with respiratory syncytial virus (RSV), reinfection in healthy individuals is common and presumably due to ineffective memory T cell responses. In peripheral blood of healthy adults, a higher CD4(+)/CD8(+) memory T cell ratio was observed compared with the ratio of virus-specific effector CD4(+)/CD8(+) T cells that we had found in earlier work during primary RSV infections. In mice, we show that an enhanced ratio of RSV-specific neutralizing to nonneutralizing Abs profoundly enhanced the CD4(+) T cell response during RSV infection. Moreover, FcγRs and complement factor C1q contributed to this Ab-mediated enhancement. Therefore, the increase in CD4(+) memory T cell response likely occurs through enhanced endosomal Ag processing dependent on FcγRs. The resulting shift in memory T cell response was likely amplified by suppressed T cell proliferation caused by RSV infection of APCs, a route important for Ag presentation via MHC class I molecules leading to CD8(+) T cell activation. Decreasing memory CD8(+) T cell numbers could explain the inadequate immunity during repeated RSV infections. Understanding this interplay of Ab-mediated CD4(+) memory T cell response enhancement and infection mediated CD8(+) memory T cell suppression is likely critical for development of effective RSV vaccines.


Asunto(s)
Anticuerpos Antivirales/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Virus Sincitiales Respiratorios/inmunología , Inmunidad Adaptativa , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/patología , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , Epítopos de Linfocito T/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/patología
7.
J Virol ; 83(14): 7235-43, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19420085

RESUMEN

In the respiratory tract, different dendritic cell (DC) populations guard a tight balance between tolerance and immunity to infectious or harmless materials to which the airways are continuously exposed. For infectious and noninfectious antigens administered via different routes, different subsets of DC might contribute during the induction of T-cell tolerance and immunity. We studied the impact of primary respiratory syncytial virus (RSV) infection on respiratory DC composition in C57BL/6 mice. We also tracked the migration of respiratory DC to the lymph nodes and studied antigen presentation by lung-derived and lymph node-resident DC to CD4(+) and CD8(+) T cells. We observed a massive influx of mainly CD103(-) CD11b(high) CD11c(+) conventional DC (cDC) and plasmacytoid DC during the first 7 days of RSV infection, while CD103(+) CD11b(low) CD11c(+) cDC disappeared from the lung. The two major subsets of lung tissue DC, CD103(+) CD11b(low) CD11c(+) and CD103(-) CD11b(high) CD11c(+) cDC, both transported RSV RNA to the lung-draining lymph node. Furthermore, these lung-derived cDC subsets as well as resident LN DC, which did not contain viral RNA, displayed viral antigen by major histocompatibility complex class I and class II to CD8(+) and CD4(+) T cells. Taken together, our data indicate that during RSV infections, at least three DC subsets might be involved during the activation of lymph node-homing naïve and memory CD4(+) and CD8(+) T cells.


Asunto(s)
Presentación de Antígeno , Movimiento Celular , Células Dendríticas/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/virología , Femenino , Humanos , Pulmón/virología , Ganglios Linfáticos/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Virus Sincitial Respiratorio/virología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...