Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 14(1)2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-35062301

RESUMEN

Human respiratory syncytial virus (hRSV) infection brings a wide spectrum of clinical outcomes, from a mild cold to severe bronchiolitis or even acute interstitial pneumonia. Among the known factors influencing this clinical diversity, genetic background has often been mentioned. In parallel, recent evidence has also pointed out that an early infectious experience affects heterologous infections severity. Here, we analyzed the importance of these two host-related factors in shaping the immune response in pneumoviral disease. We show that a prior gammaherpesvirus infection improves, in a genetic background-dependent manner, the immune system response against a subsequent lethal dose of pneumovirus primary infection notably by inducing a systematic expansion of the CD8+ bystander cell pool and by modifying the resident alveolar macrophages (AMs) phenotype to induce immediate cyto/chemokinic responses upon pneumovirus exposure, thereby drastically attenuating the host inflammatory response without affecting viral replication. Moreover, we show that these AMs present similar rapid and increased production of neutrophil chemokines both in front of pneumoviral or bacterial challenge, confirming recent studies attributing a critical antibacterial role of primed AMs. These results corroborate other recent studies suggesting that the innate immunity cells are themselves capable of memory, a capacity hitherto reserved for acquired immunity.


Asunto(s)
Antecedentes Genéticos , Infecciones por Herpesviridae/inmunología , Macrófagos Alveolares/inmunología , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Rhadinovirus/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Citocinas/metabolismo , Femenino , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Inmunidad Innata , Inflamación/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Infecciones Neumocócicas/inmunología , Pneumovirus/fisiología , Infecciones por Pneumovirus/genética , Infecciones por Pneumovirus/patología , Infecciones por Pneumovirus/virología , Rhadinovirus/fisiología
2.
Mucosal Immunol ; 13(5): 799-813, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32424182

RESUMEN

Human respiratory syncytial virus (RSV) is a pneumovirus that causes severe infections in infants worldwide. Despite intensive research, safe and effective vaccines against RSV have remained elusive. The main reason is that RSV infection of children previously immunized with formalin-inactivated-RSV vaccines has been associated with exacerbated pathology, a phenomenon called RSV vaccine-enhanced respiratory disease. In parallel, despite the high RSV prevalence, only a minor proportion of children develop severe diseases. Interestingly, variation in the immune responses against RSV or following RSV vaccination could be linked with differences of exposure to microbes during childhood. Gammaherpesviruses (γHVs), such as the Epstein-Barr virus, are persistent viruses that deeply influence the immune system of their host and could therefore affect the development of pneumovirus-induced immunopathologies for the long term. Here, we showed that a previous ɣHV infection protects against both pneumovirus vaccine-enhanced disease and pneumovirus primary infection and that CD8 T cells are essential for this protection. These observations shed a new light on the understanding of pneumovirus-induced diseases and open new perspectives for the development of vaccine strategies.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Susceptibilidad a Enfermedades , Gammaherpesvirinae/inmunología , Interacciones Huésped-Patógeno/inmunología , Infecciones por Pneumovirus/etiología , Infecciones por Pneumovirus/metabolismo , Pneumovirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunofenotipificación , Leucocitos/inmunología , Leucocitos/metabolismo , Leucocitos/patología , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones , Interacciones Microbianas , Infecciones por Pneumovirus/patología , Infecciones por Virus Sincitial Respiratorio/etiología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitial Respiratorio Humano/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Vacunación , Vacunas Virales/inmunología
3.
PLoS One ; 11(12): e0168779, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-28005954

RESUMEN

The human pneumovirus respiratory syncytial virus (RSV) is the most common pathogen causing lower respiratory tract disease in young children worldwide. A hallmark of severe human RSV infection is the strong neutrophil recruitment to the airways and lungs. Massive neutrophil activation has been proven detrimental in numerous diseases, yet in RSV the contribution of neutrophils to disease severity, and thereby, the relevance of targeting them, is largely unknown. To determine the relevance of potential neutrophil targeting therapies, we implemented antibody-mediated neutrophil depletion in a mouse pneumonia virus of mice (PVM) model. PVM is a host specific murine pneumovirus closely related to human RSV, which reproduces many of the features of RSV infection, such as high viral replication and neutrophil recruitment. Clinical disease and markers of lung inflammation and injury were studied in PVM-infected mice treated with either depleting or isotype control antibodies. To confirm our results we performed all experiments in two mice strains: C57Bl6 and BALBc mice. Neutrophil depletion in blood and lungs was efficient throughout the disease. Remarkably, in both mouse strains we found no difference in clinical disease severity between neutrophil-depleted and control arms. In line with this observation, we found no differences between groups in histopathological lung injury and lung viral loads. In conclusion, our study shows that in mice neutrophil recruitment to the lungs does not affect disease outcome or viral clearance during severe PVM infection. As such, this model does not support the notion that neutrophils play a key role in mouse pneumovirus disease.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Neumonía Murina/inmunología , Neutrófilos/inmunología , Neumonía/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neutrófilos/patología , Neumonía/patología , Infecciones por Pneumovirus/patología , Virus Sincitiales Respiratorios/inmunología
4.
Antiviral Res ; 121: 109-19, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26145728

RESUMEN

We reported previously that priming of the respiratory tract with immunobiotic Lactobacillus prior to virus challenge protects mice against subsequent lethal infection with pneumonia virus of mice (PVM). We present here the results of gene microarray which document differential expression of proinflammatory mediators in response to PVM infection alone and those suppressed in response to Lactobacillus plantarum. We also demonstrate for the first time that intranasal inoculation with live or heat-inactivated L. plantarum or Lactobacillus reuteri promotes full survival from PVM infection when administered within 24h after virus challenge. Survival in response to L. plantarum administered after virus challenge is associated with suppression of proinflammatory cytokines, limited virus recovery, and diminished neutrophil recruitment to lung tissue and airways. Utilizing this post-virus challenge protocol, we found that protective responses elicited by L. plantarum at the respiratory tract were distinct from those at the gastrointestinal mucosa, as mice devoid of the anti-inflammatory cytokine, interleukin (IL)-10, exhibit survival and inflammatory responses that are indistinguishable from those of their wild-type counterparts. Finally, although L. plantarum interacts specifically with pattern recognition receptors TLR2 and NOD2, the respective gene-deleted mice were fully protected against lethal PVM infection by L. plantarum, as are mice devoid of type I interferon receptors. Taken together, L. plantarum is a versatile and flexible agent that is capable of averting the lethal sequelae of severe respiratory infection both prior to and post-virus challenge via complex and potentially redundant mechanisms.


Asunto(s)
Factores Inmunológicos/administración & dosificación , Lactobacillus plantarum/inmunología , Limosilactobacillus reuteri/inmunología , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/patología , Infecciones por Pneumovirus/terapia , Probióticos/administración & dosificación , Administración Intranasal , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Análisis de Supervivencia
5.
Virology ; 468-470: 140-149, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25173090

RESUMEN

Interferon gamma (IFNγ) has complex immunomodulatory and antiviral properties. While IFNγ is detected in the airways in response to infection with the pneumovirus pathogen, pneumonia virus of mice (PVM; Family Paramyxoviridae), its role in promoting disease has not been fully explored. Here, we evaluate PVM infection in IFNγ(-/-) mice. Although the IFNγ gene-deletion has no impact on weight loss, survival or virus kinetics, expression of IFNß, IFNλ2/3 and IFN-stimulated 2-5' oligoadenylate synthetases was significantly diminished compared to wild-type counterparts. Furthermore, PVM infection in IFNγ(-/-) mice promoted prominent inflammation, including eosinophil and neutrophil infiltration into the airways and lung parenchyma, observed several days after peak virus titer. Potential mechanisms include over-production of chemoattractant and eosinophil-active cytokines (CXCL1, CCL11, CCL3 and IL5) in PVM-infected IFNγ(-/-) mice; likewise, IFNγ actively antagonized IL5-dependent eosinophil survival ex vivo. Our results may have clinical implications for pneumovirus infection in individuals with IFNγ signaling defects.


Asunto(s)
Citocinas/metabolismo , Regulación Viral de la Expresión Génica/inmunología , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/patología , Animales , Citocinas/genética , Eosinófilos/citología , Eliminación de Gen , Inflamación/metabolismo , Inflamación/patología , Interferón Tipo I/genética , Interferón beta/genética , Interferón beta/metabolismo , Interferón gamma/genética , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Neumonía Murina/genética , Infecciones por Pneumovirus/metabolismo
6.
Vet Microbiol ; 173(1-2): 136-40, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25129384

RESUMEN

A pneumonia virus of mice (PVM) from an African hedgehog (Atelerix arbiventris) with suspected wobbly hedgehog syndrome (WHS) was detected and genetically characterized. The affected hedgehog had a nonsuppurative encephalitis with vacuolization of the white matter, and the brain samples yielded RNA reads highly homogeneous to PVM strain 15 (96.5% of full genomic sequence homology by analysis of next generation sequencing). PVM antigen was also detected in the brain and the lungs immunohistochemically. A PVM was strongly suggested as a causative agent of encephalitis of a hedgehog with suspected WHS. This is a first report of PVM infection in hedgehogs.


Asunto(s)
Encefalitis Viral/veterinaria , Virus de la Neumonía Murina/aislamiento & purificación , Infecciones por Pneumovirus/veterinaria , Animales , Encéfalo/patología , Encéfalo/virología , Encefalitis Viral/patología , Encefalitis Viral/virología , Femenino , Erizos , Secuenciación de Nucleótidos de Alto Rendimiento , Pulmón/patología , Pulmón/virología , Ratones , Virus de la Neumonía Murina/genética , Infecciones por Pneumovirus/patología , Infecciones por Pneumovirus/virología
7.
PLoS One ; 9(7): e102749, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25047452

RESUMEN

BACKGROUND: Pulmonary edema plays a pivotal role in the pathophysiology of respiratory syncytial virus (RSV)-induced respiratory failure. In this study we determined whether treatment with TIP (AP301), a synthetic cyclic peptide that mimics the lectin-like domain of human TNF, decreases pulmonary edema in a mouse model of severe human RSV infection. TIP is currently undergoing clinical trials as a therapy for pulmonary permeability edema and has been shown to decrease pulmonary edema in different lung injury models. METHODS: C57BL/6 mice were infected with pneumonia virus of mice (PVM) and received TIP or saline (control group) by intratracheal instillation on day five (early administration) or day seven (late administration) after infection. In a separate set of experiments the effect of multiple dose administration of TIP versus saline was tested. Pulmonary edema was determined by the lung wet-to-dry (W/D) weight ratio and was assessed at different time-points after the administration of TIP. Secondary outcomes included clinical scores and lung cellular response. RESULTS: TIP did not have an effect on pulmonary edema in different dose regimens at different time points during PVM infection. In addition, TIP administration did not affect clinical severity scores or lung cellular response. CONCLUSION: In this murine model of severe RSV infection TIP did not affect pulmonary edema nor course of disease.


Asunto(s)
Virus de la Neumonía Murina/aislamiento & purificación , Péptidos Cíclicos/uso terapéutico , Infecciones por Pneumovirus/tratamiento farmacológico , Edema Pulmonar/tratamiento farmacológico , Edema Pulmonar/virología , Animales , Humanos , Masculino , Ratones Endogámicos C57BL , Virus de la Neumonía Murina/efectos de los fármacos , Péptidos Cíclicos/química , Infecciones por Pneumovirus/complicaciones , Infecciones por Pneumovirus/patología , Edema Pulmonar/patología , Infecciones por Virus Sincitial Respiratorio/complicaciones , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/aislamiento & purificación , Factor de Necrosis Tumoral alfa/química
8.
J Immunol ; 192(11): 5265-72, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24748495

RESUMEN

We have shown previously that priming of respiratory mucosa with live Lactobacillus species promotes robust and prolonged survival from an otherwise lethal infection with pneumonia virus of mice, a property known as heterologous immunity. Lactobacillus priming results in a moderate reduction in virus recovery and a dramatic reduction in virus-induced proinflammatory cytokine production; the precise mechanisms underlying these findings remain to be elucidated. Because B cells have been shown to promote heterologous immunity against respiratory virus pathogens under similar conditions, in this study we explore the role of B cells in Lactobacillus-mediated protection against acute pneumovirus infection. We found that Lactobacillus-primed mice feature elevated levels of airway Igs IgG, IgA, and IgM and lung tissues with dense, B cell (B220(+))-enriched peribronchial and perivascular infiltrates with germinal centers consistent with descriptions of BALT. No B cells were detected in lung tissue of Lactobacillus-primed B cell deficient µMT mice or Jh mice, and Lactobacillus-primed µMT mice had no characteristic infiltrates or airway Igs. Nonetheless, we observed diminished virus recovery and profound suppression of virus-induced proinflammatory cytokines CCL2, IFN-γ, and CXCL10 in both wild-type and Lactobacillus-primed µMT mice. Furthermore, Lactobacillus plantarum-primed, B cell-deficient µMT and Jh mice were fully protected from an otherwise lethal pneumonia virus of mice infection, as were their respective wild-types. We conclude that B cells are dispensable for Lactobacillus-mediated heterologous immunity and were not crucial for promoting survival in response to an otherwise lethal pneumovirus infection.


Asunto(s)
Linfocitos B/inmunología , Lactobacillus/inmunología , Pulmón/inmunología , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Mucosa Respiratoria/inmunología , Animales , Anticuerpos Antibacterianos/genética , Anticuerpos Antibacterianos/inmunología , Citocinas/genética , Citocinas/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Pneumovirus/genética , Infecciones por Pneumovirus/genética , Infecciones por Pneumovirus/patología , Mucosa Respiratoria/patología , Mucosa Respiratoria/virología
9.
J Virol ; 87(20): 11267-75, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23946463

RESUMEN

Cytotoxic T cells (CTL) play a critical role in the clearance of respiratory viral infections, but they also contribute to disease manifestations. In this study, we infected mice with a genetically modified pneumonia virus of mice (PVM) that allowed visualization of virus-specific CTL and infected cells in situ. The first virus-specific T cells entered the lung via blood vessels in the scattered foci of PVM-infected cells, which densely clustered around the bronchi at day 7 after infection. At this time, overall pulmonary virus load was maximal, but the mice showed no overt signs of disease. On days 8 to 9, T cells gained access to the infected bronchial epithelium and to the lung interstitium, which was associated with a reduction in the number of virus-infected cells within the initial clusters but could not prevent further virus spread throughout the lung tissue. Interestingly, recruitment of virus-specific CTL throughout the parenchyma was still ongoing on day 10, when the virus infection was already largely controlled. This also represented the peak of clinical disease. Thus, disease was associated with an exuberant T cell infiltration late in the course of the infection, which may be required to completely eliminate virus at residual foci of infection. PVM-induced immunopathology may thus result from the need to generate widespread T cell infiltrates to complete the elimination of virus-infected cells in a large organ like the lung. This experimental model provides the first insights into the spatiotemporal evolution of pulmonary antiviral T cell immunity in vivo.


Asunto(s)
Pulmón/inmunología , Pulmón/patología , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Linfocitos T Citotóxicos/inmunología , Animales , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Infecciones por Pneumovirus/virología , Factores de Tiempo , Carga Viral
10.
Virology ; 443(2): 257-64, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23763766

RESUMEN

A previous report of a novel pneumovirus (PnV) isolated from the respiratory tract of a dog described its significant homology to the rodent pathogen, pneumonia virus of mice (PVM). The original PnV-Ane4 pathogen replicated in and could be re-isolated in infectious state from mouse lung but elicited minimal mortality compared to PVM strain J3666. Here we assess phylogeny and physiologic responses to 10 new PnV isolates. The G/glycoprotein sequences of all PnVs include elongated amino-termini when compared to the characterized PVMs, and suggest division into groups A and B. While we observed significant differences in cytokine production and neutrophil recruitment to the lungs of BALB/c mice in response to survival doses (50 TCID50 units) of representative group A (114378-10-29-KY-F) and group B (7968-11-OK) PnVs, we observed no evidence for positive selection (dN > dS) among the PnV/PnV, PVM/PnV or PVM/PVM G/glycoprotein or F/fusion protein sequence pairs.


Asunto(s)
Evolución Molecular , Inflamación/patología , Infecciones por Pneumovirus/patología , Pneumovirus/clasificación , Pneumovirus/patogenicidad , Secuencia de Aminoácidos , Animales , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Inflamación/inmunología , Inflamación/virología , Pulmón/inmunología , Pulmón/patología , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Virus de la Neumonía Murina/genética , Virus de la Neumonía Murina/patogenicidad , Filogenia , Pneumovirus/genética , Infecciones por Pneumovirus/virología , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
11.
J Allergy Clin Immunol ; 131(5): 1331-9.e10, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23561801

RESUMEN

BACKGROUND: Respiratory tract viruses are a major environmental risk factor for both the inception and exacerbations of asthma. Genetic defects in Toll-like receptor (TLR) 7-mediated signaling, impaired type I interferon responses, or both have been reported in asthmatic patients, although their contribution to the onset and exacerbation of asthma remains poorly understood. OBJECTIVE: We sought to determine whether Pneumovirus infection in the absence of TLR7 predisposes to bronchiolitis and the inception of asthma. METHODS: Wild-type and TLR7-deficient (TLR7(-/-)) mice were inoculated with the rodent-specific pathogen pneumonia virus of mice at 1 (primary), 7 (secondary), and 13 (tertiary) weeks of age, and pathologic features of bronchiolitis or asthma were assessed. In some experiments infected mice were exposed to low-dose cockroach antigen. RESULTS: TLR7 deficiency increased viral load in the airway epithelium, which became sloughed and necrotic, and promoted an IFN-α/ß(low), IL-12p70(low), IL-1ß(high), IL-25(high), and IL-33(high) cytokine microenvironment that was associated with the recruitment of type 2 innate lymphoid cells/nuocytes and increased TH2-type cytokine production. Viral challenge of TLR7(-/-) mice induced all of the cardinal pathophysiologic features of asthma, including tissue eosinophilia, mast cell hyperplasia, IgE production, airway smooth muscle alterations, and airways hyperreactivity in a memory CD4(+) T cell-dependent manner. Importantly, infections with pneumonia virus of mice promoted allergic sensitization to inhaled cockroach antigen in the absence but not the presence of TLR7. CONCLUSION: TLR7 gene defects and Pneumovirus infection interact to establish an aberrant adaptive response that might underlie virus-induced asthma exacerbations in later life.


Asunto(s)
Asma/inmunología , Asma/patología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Virus de la Neumonía Murina , Infecciones por Pneumovirus/complicaciones , Receptor Toll-Like 7/deficiencia , Receptor Toll-Like 7/genética , Animales , Animales Recién Nacidos , Asma/etiología , Modelos Animales de Enfermedad , Pulmón/patología , Pulmón/fisiopatología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Virus de la Neumonía Murina/patogenicidad , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Carga Viral
12.
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
13.
Curr Med Chem ; 19(10): 1424-31, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22360479

RESUMEN

Respiratory syncytial virus (RSV; Family Paramyxoviridae, Genus Pneumovirus) is a major respiratory pathogen of infants and children and an emerging pathogen of the elderly. Current management of RSV disease includes monoclonal antibody prophylaxis for infants identified as high risk and supportive care for those with active infection; there is no vaccine, although several are under study. In this manuscript, we review published findings from human autopsy studies, as well as experiments that focus on human clinical samples and mouse models of acute pneumovirus infection that elucidate basic principles of disease pathogenesis. Consideration of these data suggests that the inflammatory responses to RSV and related pneumoviral pathogens can be strong, persistent, and beyond the control of conventional antiviral and anti-inflammatory therapies, and can have profound negative consequences to the host. From this perspective, we consider the case for specific immunomodulatory strategies that may have the potential to alleviate some of the more serious sequelae of this disease.


Asunto(s)
Factores Inmunológicos/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antivirales/uso terapéutico , Citocinas/antagonistas & inhibidores , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Oligodesoxirribonucleótidos Antisentido/uso terapéutico , Pneumovirus/genética , Pneumovirus/inmunología , Infecciones por Pneumovirus/tratamiento farmacológico , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Infecciones por Virus Sincitial Respiratorio/patología , Virus Sincitial Respiratorio Humano/genética
14.
J Immunol ; 188(4): 1924-32, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22238461

RESUMEN

IL-21 is a cytokine with pleiotropic actions, promoting terminal differentiation of B cells, increased Ig production, and the development of Th17 and T follicular helper cells. IL-21 is also implicated in the development of autoimmune disease and has antitumor activity. In this study, we investigated the role of IL-21 in host defense to pneumonia virus of mice (PVM), which initiates an infection in mice resembling that of respiratory syncytial virus disease in humans. We found that PVM-infected mice expressed IL-21 in lung CD4(+) T cells. Following infection, Il21r(-/-) mice exhibited less lung infiltration by neutrophils than did wild-type (WT) mice and correspondingly had lower levels of the chemokine CXCL1 in bronchoalveolar lavage fluid and lung parenchyma. CD8(+), CD4(+), and γδ T cell numbers were also lower in the lungs of PVM-infected Il21r(-/-) mice than in infected WT mice, with normal Th17 cytokines but diminished IL-6 production in PVM-infected Il21r(-/-) mice. Strikingly, Il21r(-/-) mice had enhanced survival following PVM infection, and moreover, treatment of WT mice with soluble IL-21R-Fc fusion protein enhanced their survival. These data reveal that IL-21 promotes the pathogenic inflammatory effect of PVM and indicate that manipulating IL-21 signaling may represent an immunomodulatory strategy for controlling PVM and potentially other respiratory virus infections.


Asunto(s)
Interleucinas/inmunología , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Quimiocina CXCL1/biosíntesis , Quimiocina CXCL1/inmunología , Interleucina-6/biosíntesis , Interleucina-6/deficiencia , Interleucinas/biosíntesis , Interleucinas/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Virus de la Neumonía Murina/patogenicidad , Receptores de Interleucina-21/inmunología , Células Th17/inmunología
15.
Viruses ; 4(12): 3494-510, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23342367

RESUMEN

Pneumonia Virus of Mice (PVM) is related to the human and bovine respiratory syncytial virus (RSV) pathogens, and has been used to study respiratory virus replication and the ensuing inflammatory response as a component of a natural host­pathogen relationship. As such, PVM infection in mice reproduces many of the clinical and pathologic features of the more severe forms of RSV infection in human infants. Here we review some of the most recent findings on the basic biology of PVM infection and its use as a model of disease, most notably for explorations of virus infection and allergic airways disease, for vaccine evaluation, and for the development of immunomodulatory strategies for acute respiratory virus infection.


Asunto(s)
Modelos Animales de Enfermedad , Virus de la Neumonía Murina/patogenicidad , Infecciones por Pneumovirus/patología , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/virología , Animales , Ratones , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones del Sistema Respiratorio/inmunología
16.
Virology ; 416(1-2): 26-31, 2011 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-21600624

RESUMEN

Canine pneumovirus (CnPnV) was recently isolated from the respiratory tracts of shelter dogs and shares sequence similarity with the rodent pathogen, pneumonia virus of mice (PVM). We show here that CnPnV replicates in and can elicit local proinflammatory cytokine production and neutrophil recruitment to lung tissue and the airways. In contrast to PVM J3666 infection, fatal CnPnV infections are observed only in response to high titer intranasal inocula (>67 TCID(50) units). Sera from mice that recover from CnPnV infection contain antibodies that cross-react with PVM antigens; these mice are protected against lethal PVM infection. Given these findings, it will be intriguing to determine the relative role(s) of CnPnV and PVM in eliciting respiratory symptoms in susceptible canine species.


Asunto(s)
Inflamación/virología , Pulmón/virología , Infecciones por Pneumovirus/virología , Pneumovirus/clasificación , Pneumovirus/fisiología , Replicación Viral/fisiología , Animales , Perros , Inflamación/patología , Ratones , Ratones Endogámicos BALB C , Filogenia , Pneumovirus/genética , Infecciones por Pneumovirus/patología
17.
J Virol ; 85(9): 4071-84, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21307191

RESUMEN

Infection of mice with pneumonia virus of mice (PVM) provides a convenient experimental pathogenesis model in a natural host for a human respiratory syncytial virus-related virus. Extending our previous work showing that the PVM nonstructural (NS) proteins were pathogenicity factors in mice, we identify both the NS1 and NS2 proteins as antagonists of alpha/beta interferon (IFN-α/ß) and IFN-λ by use of recombinant PVM (rPVM) with single and combined deletions of the NS proteins (ΔNS1, ΔNS2, and ΔNS1 ΔNS2). Wild-type and NS deletion PVMs were evaluated for growth and pathogenesis by infecting knockout mice that lack functional receptors to IFN-α/ß, IFN-λ, or both. The absence of the receptor to IFN-α/ß (IFNAR) or IFN-λ (interleukin-28 receptor α chain [IL-28Rα]) individually did not reverse the attenuated virulence of the NS deletion viruses although loss of IFNAR partially restored replication efficiency. When both receptors were deleted, replication and virulence were largely rescued for rPVM ΔNS1 and were significantly but not completely rescued for rPVM ΔNS2. As for rPVM ΔNS1 ΔNS2, the effect was mostly limited to partial enhancement of replication. This indicates that both IFN-α/ß and IFN-λ contributed to restricting the NS deletion viruses, with the former playing the greater role. Interestingly, the replication and virulence of wild-type PVM were completely unaffected by the presence or absence of functional receptors to IFN-α/ß and IFN-λ, indicating that both systems are strongly suppressed during infection. However, pretreatment of mice with IFN-α/ß was protective against lethal rPVM challenge, whereas pretreatment with IFN-λ delayed but did not prevent disease and, in some cases, reduced mortality. The fact that virulence of rPVM lacking NS2 was not recovered completely when both interferon receptors were deleted suggests that NS2 may have further functions outside the IFN system.


Asunto(s)
Citocinas/antagonistas & inhibidores , Interferón Tipo I/antagonistas & inhibidores , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/veterinaria , Enfermedades de los Roedores/virología , Proteínas no Estructurales Virales/inmunología , Factores de Virulencia/inmunología , Animales , Eliminación de Gen , Histocitoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía , Virus de la Neumonía Murina/patogenicidad , Infecciones por Pneumovirus/patología , Infecciones por Pneumovirus/virología , Enfermedades de los Roedores/patología , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Replicación Viral
18.
Virol J ; 7: 320, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21078159

RESUMEN

BACKGROUND: The innate immune responses of neonates differ dramatically from those of adults. Here we examine the acute inflammatory responses of neonatal and weanling mice infected with pneumonia virus of mice (PVM), a rodent pathogen (family Paramyxoviridae, genus Pneumovirus) that replicates the sequelae of severe respiratory syncytial virus infection. RESULTS: We demonstrate that virus replication proceeds indistinguishably in all age groups (inoculated at 1, 2, 3 and 4 weeks of age), although inflammatory responses vary in extent and character. Some of the biochemical mediators detected varied minimally with age at inoculation. Most of the mediators evaluated demonstrated elevated expression over baseline correlating directly with age at the time of virus inoculation. Among the latter group are CCL2, CCL3, and IFN-γ, all cytokines previously associated with PVM-induced inflammatory pathology in mature mice. Likewise, we detect neutrophil recruitment to lung tissue in all age groups, but recruitment is most pronounced among the older (3 - 4 week old) mice. Interestingly, all mice exhibit failure to thrive, lagging in expected weight gain for given age, including the youngest mice that present little overt evidence of inflammation. CONCLUSIONS: Our findings among the youngest mice may explain in part the phenomenon of atypical or minimally symptomatic respiratory infections in human neonates, which may be explored further with this infection model.


Asunto(s)
Virus de la Neumonía Murina/inmunología , Virus de la Neumonía Murina/patogenicidad , Infecciones por Pneumovirus/veterinaria , Factores de Edad , Animales , Animales Recién Nacidos , Peso Corporal , Citocinas/biosíntesis , Expresión Génica , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/inmunología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Replicación Viral
19.
J Immunol ; 183(1): 604-12, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19542471

RESUMEN

Enhanced disease is the term used to describe the aberrant Th2-skewed responses to naturally acquired human respiratory syncytial virus (hRSV) infection observed in individuals vaccinated with formalin-inactivated viral Ags. Here we explore this paradigm with pneumonia virus of mice (PVM), a pathogen that faithfully reproduces features of severe hRSV infection in a rodent host. We demonstrate that PVM infection in mice vaccinated with formalin-inactivated Ags from PVM-infected cells (PVM Ags) yields Th2-skewed hypersensitivity, analogous to that observed in response to hRSV. Specifically, we detect elevated levels of IL-4, IL-5, IL-13, and eosinophils in bronchoalveolar lavage fluid of PVM-infected mice that were vaccinated with PVM Ags, but not among mice vaccinated with formalin-inactivated Ags from uninfected cells (control Ags). Interestingly, infection in PVM Ag-vaccinated mice was associated with a approximately 10-fold reduction in lung virus titer and protection against weight loss when compared with infected mice vaccinated with control Ags, despite the absence of serum-neutralizing Abs. Given recent findings documenting a role for eosinophils in promoting clearance of hRSV in vivo, we explored the role of eosinophils in altering the pathogenesis of disease with eosinophil-deficient mice. We found that eosinophil deficiency had no impact on virus titer in PVM Ag-vaccinated mice, nor on weight loss or levels of CCL11 (eotaxin-1), IFN-gamma, IL-5, or IL-13 in bronchoalveolar lavage fluid. However, levels of both IL-4 and CCL3 (macrophage inflammatory protein-1alpha) in bronchoalveolar lavage fluid were markedly diminished in PVM Ag-vaccinated, PVM-infected eosinophil-deficient mice when compared with wild-type controls.


Asunto(s)
Eosinófilos/inmunología , Eosinófilos/patología , Formaldehído , Pulmón/inmunología , Pulmón/patología , Virus de la Neumonía Murina/inmunología , Infecciones por Pneumovirus/prevención & control , Vacunas Virales/inmunología , Animales , Antígenos Virales/administración & dosificación , Antígenos Virales/inmunología , Línea Celular , Eosinófilos/virología , Fijadores , Hipersensibilidad/inmunología , Hipersensibilidad/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Mutantes , Virus de la Neumonía Murina/crecimiento & desarrollo , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/efectos adversos , Vacunas de Productos Inactivados/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos
20.
Am J Physiol Lung Cell Mol Physiol ; 296(1): L46-56, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18996903

RESUMEN

Severe infection with respiratory syncytial virus (RSV) in children can progress to respiratory distress and acute lung injury (ALI). Accumulating evidence suggests that mechanical ventilation (MV) is an important cofactor in the development of ALI by modulating the host immune responses to bacteria. This study investigates whether MV enhances the host response to pneumonia virus of mice (PVM), a mouse pneumovirus that has been used as a model for RSV infection in humans. BALB/c mice were inoculated intranasally with diluted clarified lung homogenates from mice infected with PVM strain J3666 or uninfected controls. Four days after inoculation, the mice were subjected to 4 h of MV (tidal volume, 10 ml/kg) or allowed to breathe spontaneously. When compared with that of mice inoculated with PVM only, the administration of MV to PVM-infected mice resulted in increased bronchoalveolar lavage fluid concentrations of the cytokines macrophage inflammatory protein (MIP)-2, MIP-1alpha (CCL3), and IL-6; increased alveolar-capillary permeability to high molecular weight proteins; and increased caspase-3 activity in lung homogenates. We conclude that MV enhances the activation of inflammatory and caspase cell death pathways in response to pneumovirus infection. We speculate that MV potentially contributes to the development of lung injury in patients with RSV infection.


Asunto(s)
Caspasas/metabolismo , Virus de la Neumonía Murina , Neumonía Viral/terapia , Neumonía/virología , Infecciones por Pneumovirus/terapia , Respiración Artificial/efectos adversos , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/virología , Animales , Apoptosis/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Leucocitos/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Permeabilidad , Neumonía/etiología , Neumonía/inmunología , Neumonía Viral/inmunología , Neumonía Viral/patología , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/patología , Respiración Artificial/métodos , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...