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

Banco de datos
Tipo de estudio
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
J Immunol ; 186(2): 1151-61, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21169550

RESUMEN

The inflammatory response to respiratory virus infection can be complex and refractory to standard therapy. Lactobacilli, when targeted to the respiratory epithelium, are highly effective at suppressing virus-induced inflammation and protecting against lethal disease. Specifically, wild-type mice primed via intranasal inoculation with live or heat-inactivated Lactobacillus plantarum or Lactobacillus reuteri were completely protected against lethal infection with the virulent rodent pathogen, pneumonia virus of mice; significant protection (60% survival) persisted for at least 13 wk. Protection was not unique to Lactobacillus species, and it was also observed in response to priming with nonpathogenic Gram-positive Listeria innocua. Priming with live lactobacilli resulted in diminished granulocyte recruitment, diminished expression of multiple proinflammatory cytokines (CXCL10, CXCL1, CCL2, and TNF), and reduced virus recovery, although we have demonstrated clearly that absolute virus titer does not predict clinical outcome. Lactobacillus priming also resulted in prolonged survival and protection against the lethal sequelae of pneumonia virus of mice infection in MyD88 gene-deleted (MyD88(-/-)) mice, suggesting that the protective mechanisms may be TLR-independent. Most intriguing, virus recovery and cytokine expression patterns in Lactobacillus-primed MyD88(-/-) mice were indistinguishable from those observed in control-primed MyD88(-/-) counterparts. In summary, we have identified and characterized an effective Lactobacillus-mediated innate immune shield, which may ultimately serve as critical and long-term protection against infection in the absence of specific antiviral vaccines.


Asunto(s)
Lactobacillus plantarum/inmunología , Limosilactobacillus reuteri/inmunología , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/prevención & control , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Administración Intranasal , Animales , Antígenos Virales/metabolismo , Citocinas/antagonistas & inhibidores , Citocinas/biosíntesis , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Neumonía Murina/patogenicidad , Infecciones por Pneumovirus/inmunología , Mucosa Respiratoria/virología , Replicación Viral/inmunología
2.
Antiviral Res ; 171: 104594, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31470041

RESUMEN

Respiratory syncytial virus (RSV) is responsible for a large proportion of acute lower respiratory tract infections, specifically in children. Pneumonia virus of mice (PVM) causes similar lung pathology and clinical disease in rodents, and is therefore an appropriate model of RSV infection. Previously, we demonstrated that a single intranasal dose of P-I-P, a novel immunomodulator composed of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene, confers protection in Balb/c mice for up to 3 days from lethal PVM-15 infection. In the present study a dual intranasal treatment with P-I-P was shown to extend the duration of the protection conferred by P-I-P from PVM-15 challenge. Balb/c mice treated twice with P-I-P showed higher survival rates and milder clinical signs when compared to animals that received a single P-I-P dose. While the mice treated with two consecutive doses of P-I-P experienced some weight loss, they all recovered. The dual P-I-P treatment mediated infiltration of several innate immune cells into the BALF and lung, including alveolar macrophages, neutrophils, and γδ T cells. Partial depletion of alveolar macrophages decreased survival rates and exacerbated clinical signs of mice subjected to the P-I-P dual treatment regime followed by PVM-15 challenge. This suggests that the alveolar macrophage is at least partially responsible for the protection elicited by this novel prophylactic treatment strategy.


Asunto(s)
Inmunidad Innata , Factores Inmunológicos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Virus de la Neumonía Murina/efectos de los fármacos , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/virología , Animales , Línea Celular , Citocinas/biosíntesis , Citocinas/sangre , Femenino , Interacciones Huésped-Patógeno , Factores Inmunológicos/administración & dosificación , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Infecciones por Pneumovirus/tratamiento farmacológico , Infecciones por Pneumovirus/mortalidad
3.
Antiviral Res ; 69(2): 53-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16337014

RESUMEN

In this work we have evaluated the clinical responses of pneumovirus-infected mice to combination therapy with the antiviral agent, ribavirin, and the CysLT1 cysteinyl leukotriene receptor antagonist, montelukast. We observed substantial virus replication in our mouse model of pneumovirus infection and significant accumulation of cysteinyl leukotrienes in lung tissue, the latter detected at levels that correlate directly with granulocyte recruitment to the airways. While administration of the nucleoside analog, ribavirin, reduced virus replication approximately 2,000-fold, the clinical outcomes as measured by morbidity and mortality, in response to ribavirin monotherapy were indistinguishable from those of the no-treatment controls. Similarly, montelukast therapy alone did not reduce granulocyte recruitment nor did it improve the clinical outcome. However, combined therapy with ribavirin and montelukast resulted in a significant reduction in morbidity and a substantial reduction in mortality (50% survival at t = 14 days and onward, compared to 10-20% survival in response to montelukast alone or to ribavirin alone, respectively, p < 0.01). These findings define further the independent contributions made by virus replication and by the ensuing inflammatory response to the detrimental sequelae of pneumovirus infection in vivo.


Asunto(s)
Acetatos/uso terapéutico , Antivirales/uso terapéutico , Bronquiolitis Viral/tratamiento farmacológico , Antagonistas de Leucotrieno/uso terapéutico , Virus de la Neumonía Murina/patogenicidad , Quinolinas/uso terapéutico , Ribavirina/uso terapéutico , Acetatos/administración & dosificación , Animales , Antivirales/administración & dosificación , Bronquiolitis Viral/mortalidad , Bronquiolitis Viral/virología , Ciclopropanos , Cisteína/antagonistas & inhibidores , Modelos Animales de Enfermedad , Quimioterapia Combinada , Humanos , Antagonistas de Leucotrieno/administración & dosificación , Leucotrienos , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Virus de la Neumonía Murina/efectos de los fármacos , Virus de la Neumonía Murina/fisiología , Infecciones por Pneumovirus/tratamiento farmacológico , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/virología , Quinolinas/administración & dosificación , Ribavirina/administración & dosificación , Sulfuros , Resultado del Tratamiento , Replicación Viral
4.
Antiviral Res ; 132: 131-40, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27312104

RESUMEN

Pattern recognition receptors (PRRs) engage microbial components in the lung, although their role in providing primary host defense against respiratory virus infection is not fully understood. We have previously shown that Gram-positive Lactobacillus plantarum (Lp) administered to the respiratory tract promotes full and sustained protection in response to an otherwise lethal mouse pneumovirus (PVM) infection, a robust example of heterologous immunity. While Lp engages PRRs TLR2 and NOD2 in ex vivo signaling assays, we found that Lp-mediated protection was unimpaired in single gene-deleted TLR2(-/-) and NOD2(-/-) mice. Here we demonstrate substantial loss of Lp-mediated protection in a double gene-deleted NOD2(-/-)TLR2(-/-) strain. Furthermore, we demonstrate protection against PVM infection by administration of the bi-functional NOD2-TLR2 agonist, CL-429. The bi-functional NOD2-TLR2 ligand CL-429 not only suppresses virus-induced inflammation, it is significantly more effective at preventing lethal infection than equivalent amounts of mono-molecular TLR2 and NOD2 agonists. Interestingly, and in contrast to biochemical NOD2 and/or TLR2 agonists, Lp remained capable of eliciting primary proinflammatory responses from NOD2(-/-)TLR2(-/-) mice in vivo and from alveolar macrophages challenged ex vivo. Taken together, we conclude that coordinate engagement of NOD2 and TLR2 constitutes a key step in the genesis of Lp-mediated protection from a lethal respiratory virus infection, and represents a critical target for modulation of virus-induced inflammatory pathology.


Asunto(s)
Inmunomodulación , Proteína Adaptadora de Señalización NOD2/metabolismo , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/metabolismo , Pneumovirus/inmunología , Transducción de Señal , Receptor Toll-Like 2/metabolismo , Animales , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Lactobacillus plantarum/inmunología , Ligandos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Noqueados , Virus de la Neumonía Murina/inmunología , Proteína Adaptadora de Señalización NOD2/genética , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/virología , Receptores de Reconocimiento de Patrones/metabolismo , Receptor Toll-Like 2/genética , Carga Viral
5.
J Interferon Cytokine Res ; 32(7): 332-7, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22385204

RESUMEN

The respiratory syncytial virus (RSV) is a major pathogen of the human species. This pneumovirus is a prominent cause of airway morbidity in children and maintains an excessive hospitalization rate despite decades of research. As involvement of a genetic vulnerability is a possibility supported by recent data, we addressed the question of whether the Mx gene products, the typical target of which consists in single-stranded negative-polarity RNA viruses, could alter the course of pneumovirus-associated disease in vivo. Wild-type and Bos taurus Mx1-expressing transgenic FVB/J mice were inoculated with the mouse counterpart and closest phylogenetic relative of RSV, pneumonia virus of mice. Survival data and follow-up of body weight, histological scores, lung virus spread, and lung viral load unequivocally showed that the viral infection was severely repressed in Mx-transgenic mice, thus suggesting that pneumoviruses belong to the antiviral spectrum of mammalian Mx GTPases. Elucidating the underlying mechanisms at the molecular level could reveal critical information for the development of new anti-RSV molecules.


Asunto(s)
Proteínas de Unión al GTP/inmunología , Inmunidad Innata , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Administración Intranasal , Animales , Animales Modificados Genéticamente , Bovinos , Femenino , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Humanos , Ratones , Proteínas de Resistencia a Mixovirus , Pneumovirus/genética , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/patología , Análisis de Supervivencia
6.
J Immunol ; 175(7): 4735-44, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16177121

RESUMEN

Pneumonia virus of mice (PVM; family Paramyxoviridae) is a natural pathogen of rodents that reproduces important clinical features of severe respiratory syncytial virus infection in humans. As anticipated, PVM infection induces transcription of IFN antiviral response genes preferentially in wild-type over IFN-alphabetaR gene-deleted (IFN-alphabetaR-/-) mice. However, we demonstrate that PVM infection results in enhanced expression of eotaxin-2 (CCL24), thymus and activation-regulated chemokine (CCL17), and the proinflammatory RNase mouse eosinophil-associated RNase (mEar) 11, and decreased expression of monocyte chemotactic protein-5, IFN-gamma-inducible protein-10, and TLR-3 in lung tissue of IFN-alphabetaR-/- mice when compared with wild type. No differential expression of chemokines MIP-1alpha or MIP-2 or Th2 cytokines IL-4 or IL-5 was observed. Differential expression of proinflammatory mediators was associated with distinct patterns of lung pathology. The widespread granulocytic infiltration and intra-alveolar edema observed in PVM-infected, wild-type mice are replaced with patchy, dense inflammatory foci localized to the periphery of the larger blood vessels. Bronchoalveolar lavage fluid from IFN-alphabetaR-/- mice yielded 7- to 8-fold fewer leukocytes overall, with increased percentages of eosinophils, monocytes, and CD4+ T cells, and decreased percentage of CD8+ T cells. Differential pathology is associated with prolonged survival of the IFN-alphabetaR-/- mice (50% survival at 10.8 +/- 0.6 days vs the wild type at 9.0 +/- 0.3 days; p < 0.02) despite increased virus titers. Overall, our findings serve to identify novel transcripts that are differentially expressed in the presence or absence of IFN-alphabetaR-mediated signaling, further elucidating interactions between the IFN and antiviral inflammatory responses in vivo.


Asunto(s)
Eliminación de Gen , Pulmón/patología , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Animales , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CXCL10 , Quimiocina CXCL2 , Quimiocinas/biosíntesis , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/genética , Inflamación/genética , Inflamación/inmunología , Inflamación/virología , Interferón Tipo I/biosíntesis , Interferón Tipo I/genética , Leucocitos/patología , Pulmón/inmunología , Pulmón/virología , Proteínas Inflamatorias de Macrófagos/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quimioatrayentes de Monocitos/biosíntesis , Proteínas Quimioatrayentes de Monocitos/genética , Pneumovirus/fisiología , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/patología , ARN Mensajero/metabolismo , Replicación Viral/fisiología
7.
J Virol ; 78(15): 7984-9, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254170

RESUMEN

We present an antiviral-immunomodulatory therapeutic strategy involving the chemokine receptor antagonist Met-RANTES, which yields significant survival in the setting of an otherwise fatal respiratory virus infection. In previous work, we demonstrated that infection with the natural rodent pathogen pneumonia virus of mice involves robust virus replication accompanied by cellular inflammation modulated by the CC chemokine macrophage inflammatory protein 1alpha (MIP-1alpha). We found that the antiviral agent ribavirin limited virus replication in vivo but had no impact on morbidity and mortality associated with this disease in the absence of immunomodulatory control. We show here that ribavirin reduces mortality, from 100% to 10 and 30%, respectively, in gene-deleted CCR1(-/-) mice and in wild-type mice treated with the small-molecule chemokine receptor antagonist, Met-RANTES. As MIP-1alpha-mediated inflammation is a common response to several distantly related respiratory virus pathogens, specific antiviral therapy in conjunction with blockade of the MIP-1alpha/CCR1 inflammatory cascade may ultimately prove to be a useful, generalized approach to severe respiratory virus infection and its pathological sequelae in human subjects.


Asunto(s)
Quimiocina CCL5/análogos & derivados , Quimiocina CCL5/uso terapéutico , Infecciones por Pneumovirus/tratamiento farmacológico , Receptores de Quimiocina/antagonistas & inhibidores , Enfermedad Aguda , Animales , Quimiocina CCL3 , Quimiocina CCL4 , Leucocitos/fisiología , Pulmón/patología , Proteínas Inflamatorias de Macrófagos/biosíntesis , Ratones , Ratones Endogámicos C57BL , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/patología , Receptores CCR1
8.
J Infect Dis ; 184(12): 1518-23, 2001 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11740726

RESUMEN

The use of glucocorticoids for the treatment of symptoms associated with respiratory syncytial virus (RSV) infection has been questioned. To evaluate the sequelae of glucocorticoid administration in the setting of pneumovirus infection in vivo, hydrocortisone was administered to mice infected with pneumonia virus of mice (PVM), a pneumovirus and natural rodent pathogen that is closely related to RSV and replicates the signs and symptoms of severe human RSV infection. Results showed that hydrocortisone spared the pulmonary neutrophilia but resulted in ablation of the pulmonary eosinophilia, despite continued production of the relevant chemoattractant, macrophage inflammatory protein-1alpha. Hydrocortisone also led to diminished production of inducible nitric oxide synthase and accumulation of reactive nitrogen species in lung tissue and bronchoalveolar lavage fluid and diminished lymphocyte recruitment. PVM-infected mice responded to hydrocortisone with enhanced viral replication and accelerated mortality. These results suggest several mechanisms to explain why glucocorticoid therapy may be of limited benefit in the overall picture of pneumovirus infection.


Asunto(s)
Antiinflamatorios/administración & dosificación , Hidrocortisona/administración & dosificación , Virus de la Neumonía Murina/fisiología , Neumonía Viral/inmunología , Infecciones por Pneumovirus/inmunología , Animales , Quimiocina CCL2/metabolismo , Quimiocina CCL4 , Modelos Animales de Enfermedad , Humanos , Pulmón/inmunología , Pulmón/virología , Proteínas Inflamatorias de Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Virus de la Neumonía Murina/aislamiento & purificación , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/virología , Infecciones por Pneumovirus/tratamiento farmacológico , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/virología , Resultado del Tratamiento , Replicación Viral/efectos de los fármacos
9.
J Immunol ; 165(5): 2677-82, 2000 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-10946298

RESUMEN

In this work, we explore the responses of specific gene-deleted mice to infection with the paramyxovirus pneumonia virus of mice (PVM). We have shown previously that infection of wild type mice with PVM results in pulmonary neutrophilia and eosinophilia accompanied by local production of macrophage-inflammatory protein-1 alpha (MIP-1 alpha). Here we examine the role of MIP-1 alpha in the pathogenesis of this disease using mice deficient in MIP-1 alpha or its receptor, CCR1. The inflammatory response to PVM in MIP-1 alpha-deficient mice was minimal, with approximately 10-60 neutrophils/ml and no eosinophils detected in bronchoalveolar lavage fluid. Higher levels of infectious virus were recovered from lung tissue excised from MIP-1 alpha-deficient than from fully competent mice, suggesting that the inflammatory response limits the rate of virus replication in vivo. PVM infection of CCR1-deficient mice was also associated with attenuated inflammation, with enhanced recovery of infectious virus, and with accelerated mortality. These results suggest that the MIP-1 alpha/CCR1-mediated acute inflammatory response protects mice by delaying the lethal sequelae of infection.


Asunto(s)
Pulmón/inmunología , Pulmón/patología , Proteínas Inflamatorias de Macrófagos/fisiología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Pneumovirus/inmunología , Receptores de Quimiocina/fisiología , Animales , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Movimiento Celular/inmunología , Quimiocina CCL4 , Eosinófilos/inmunología , Eosinófilos/patología , Eosinófilos/virología , Femenino , Pulmón/metabolismo , Pulmón/virología , Linfocitos/inmunología , Linfocitos/patología , Linfocitos/virología , Proteínas Inflamatorias de Macrófagos/deficiencia , Proteínas Inflamatorias de Macrófagos/genética , Proteínas Inflamatorias de Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Neutrófilos/patología , Neutrófilos/virología , Pneumovirus/aislamiento & purificación , Infecciones por Pneumovirus/mortalidad , Infecciones por Pneumovirus/virología , Eosinofilia Pulmonar/inmunología , Eosinofilia Pulmonar/mortalidad , Eosinofilia Pulmonar/patología , Receptores CCR1 , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA