Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
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.
PLoS Pathog ; 16(10): e1008942, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33035266

RESUMEN

Human metapneumovirus (hMPV) is a leading cause of viral respiratory infection in children, and can cause severe lower respiratory tract infection in infants, the elderly, and immunocompromised patients. However, there remain no licensed vaccines or specific treatments for hMPV infection. Although the hMPV fusion (F) protein is the sole target of neutralizing antibodies, the immunological properties of hMPV F remain poorly understood. To further define the humoral immune response to the hMPV F protein, we isolated two new human monoclonal antibodies (mAbs), MPV458 and MPV465. Both mAbs are neutralizing in vitro and were determined to target a unique antigenic site using competitive biolayer interferometry. We determined both MPV458 and MPV465 have higher affinity for monomeric hMPV F than trimeric hMPV F. MPV458 was co-crystallized with hMPV F, and the mAb primarily interacts with an alpha helix on the F2 region of the hMPV F protein. Surprisingly, the major epitope for MPV458 lies within the trimeric interface of the hMPV F protein, suggesting significant breathing of the hMPV F protein must occur for host immune recognition of the novel epitope. In addition, significant glycan interactions were observed with a somatically mutated light chain framework residue. The data presented identifies a novel epitope on the hMPV F protein for epitope-based vaccine design, and illustrates a new mechanism for human antibody neutralization of viral glycoproteins.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Pneumovirus/inmunología , Anticuerpos Neutralizantes/farmacología , Epítopos/inmunología , Humanos , Metapneumovirus/inmunología , Infecciones por Paramyxoviridae/virología , Virus Sincitial Respiratorio Humano/inmunología , Proteínas Virales de Fusión/inmunología
3.
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
4.
Front Immunol ; 10: 2778, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31849961

RESUMEN

The pneumoviruses respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) are two widespread human pathogens that can cause severe disease in the young, the elderly, and the immunocompromised. Despite the discovery of RSV over 60 years ago, and hMPV nearly 20 years ago, there are no approved vaccines for either virus. Antibody-mediated immunity is critical for protection from RSV and hMPV, and, until recently, knowledge of the antibody epitopes on the surface glycoproteins of RSV and hMPV was very limited. However, recent breakthroughs in the recombinant expression and stabilization of pneumovirus fusion proteins have facilitated in-depth characterization of antibody responses and structural epitopes, and have provided an enormous diversity of new monoclonal antibody candidates for therapeutic development. These new data have primarily focused on the RSV F protein, and have led to a wealth of new vaccine candidates in preclinical and clinical trials. In contrast, the major structural antibody epitopes remain unclear for the hMPV F protein. Overall, this review will cover recent advances in characterizing the antigenic sites on the RSV and hMPV F proteins.


Asunto(s)
Anticuerpos Antivirales/inmunología , Epítopos/inmunología , Infecciones por Pneumovirus/epidemiología , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Proteínas Virales de Fusión/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/química , Antígenos Virales/química , Antígenos Virales/inmunología , Costo de Enfermedad , Epítopos/química , Salud Global , Humanos , Infecciones por Pneumovirus/virología , Unión Proteica/inmunología , Vigilancia en Salud Pública , Infecciones por Virus Sincitial Respiratorio/epidemiología , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/inmunología , Relación Estructura-Actividad , Proteínas Virales de Fusión/química
5.
J Leukoc Biol ; 102(3): 905-913, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28619948

RESUMEN

Administration of immunobiotic Lactobacillus plantarum (Lp) directly to the respiratory mucosa promotes cross-protection against lethal pneumovirus infection via B-cell-independent mechanisms. In this study, we examined Lp-mediated cross protection in Rag1-/- mice which cannot clear virus from lung tissue. Although Lp was initially protective, Rag1-/- mice ultimately succumbed to a delayed lethal outcome associated with local production of the proinflammatory cytokines CCL1, -2, and -7, granulocyte recruitment, and ongoing virus replication. By contrast, CD8null mice, which are fully capable of clearing virus, are protected by Lp with no delayed lethal outcome, granulocyte recruitment to the airways, or induction of CCL7. Repeated administration of Lp to virus-infected Rag1-/- mice had no impact on delayed mortality. Moreover, administration of Lp to the respiratory mucosa resulted in no induction of IFN-α or -ß in Rag1-/- or wild-type mice, and IFN-abR gene deletion had no impact on Lp-mediated protection. Overall, our findings indicate that although Lp administered to the respiratory tract has substantial impact on lethal virus-induced inflammation in situ, endogenous virus clearance mechanisms are needed to promote sustained protection. Our results suggest that a larger understanding of the mechanisms and mediators that limit acute virus-induced inflammation may yield new and useful therapeutic modalities.


Asunto(s)
Proteínas de Homeodominio/genética , Lactobacillus plantarum , Infecciones por Pneumovirus/inmunología , Infecciones por Pneumovirus/terapia , Pneumovirus/inmunología , Animales , Quimiocinas CC/genética , Quimiocinas CC/inmunología , Ratones , Ratones Noqueados , Infecciones por Pneumovirus/genética
6.
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
7.
Clin Vaccine Immunol ; 22(5): 477-83, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25716232

RESUMEN

Acute lower respiratory tract infections (ALRTI) are the leading cause of global childhood mortality, with human respiratory syncytial virus (hRSV) being a major cause of viral ALRTI in young children worldwide. In sub-Saharan Africa, many young children experience severe illnesses due to hRSV or Plasmodium infection. Although the incidence of malaria in this region has decreased in recent years, there remains a significant opportunity for coinfection. Recent data show that febrile young children infected with Plasmodium are often concurrently infected with respiratory viral pathogens but are less likely to suffer from pneumonia than are non-Plasmodium-infected children. Here, we hypothesized that blood-stage Plasmodium infection modulates pulmonary inflammatory responses to a viral pathogen but does not aid its control in the lung. To test this, we established a novel coinfection model in which mice were simultaneously infected with pneumovirus of mice (PVM) (to model hRSV) and blood-stage Plasmodium chabaudi chabaudi AS (PcAS) parasites. We found that PcAS infection was unaffected by coinfection with PVM. In contrast, PVM-associated weight loss, pulmonary cytokine responses, and immune cell recruitment to the airways were substantially reduced by coinfection with PcAS. Importantly, PcAS coinfection facilitated greater viral dissemination throughout the lung. Although Plasmodium coinfection induced low levels of systemic interleukin-10 (IL-10), this regulatory cytokine played no role in the modulation of lung inflammation or viral dissemination. Instead, we found that Plasmodium coinfection drove an early systemic beta interferon (IFN-ß) response. Therefore, we propose that blood-stage Plasmodium coinfection may exacerbate viral dissemination and impair inflammation in the lung by dysregulating type I IFN-dependent responses to respiratory viruses.


Asunto(s)
Bronquiolitis Viral/inmunología , Coinfección , Interferón beta/inmunología , Pulmón/virología , Malaria/inmunología , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Animales , Bronquiolitis Viral/virología , Modelos Animales de Enfermedad , Femenino , Inflamación/inmunología , Inflamación/parasitología , Inflamación/virología , Interferón beta/sangre , Interleucina-10/inmunología , Pulmón/inmunología , Malaria/complicaciones , Plasmodium chabaudi , Pneumovirus/patogenicidad , Pneumovirus/fisiología , Infecciones por Pneumovirus/complicaciones , Virus Sincitial Respiratorio Humano/patogenicidad , Carga Viral , Pérdida de Peso
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 Clin Microbiol ; 51(12): 4112-9, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24088858

RESUMEN

Canine pneumovirus (CnPnV) was recently identified during a retrospective survey of kenneled dogs in the United States. In this study, archived samples from pet and kenneled dogs in the United Kingdom were screened for CnPnV to explore the relationship between exposure to CnPnV and the development of canine infectious respiratory disease (CIRD). Within the pet dog population, CnPnV-seropositive dogs were detected throughout the United Kingdom and Republic of Ireland, with an overall estimated seroprevalence of 50% (n = 314/625 dogs). In the kennel population, there was a significant increase in seroprevalence, from 26% (n = 56/215 dogs) on the day of entry to 93.5% (n = 201/215 dogs) after 21 days (P <0001). Dogs that were seronegative on entry but seroconverted while in the kennel were 4 times more likely to develop severe respiratory disease than those that did not seroconvert (P < 0.001), and dogs with preexisting antibodies to CnPnV on the day of entry were significantly less likely to develop respiratory disease than immunologically naive dogs (P < 0.001). CnPnV was detected in the tracheal tissues of 29/205 kenneled dogs. Detection was most frequent in dogs with mild to moderate respiratory signs and histopathological changes and in dogs housed for 8 to 14 days, which coincided with a significant increase in the risk of developing respiratory disease compared to the risk of those housed 1 to 7 days (P < 0.001). These findings demonstrate that CnPnV is present in the United Kingdom dog population; there is a strong association between exposure to CnPnV and CIRD in the kennel studied and a potential benefit in vaccinating against CnPnV as part of a wider disease prevention strategy.


Asunto(s)
Enfermedades de los Perros/epidemiología , Enfermedades de los Perros/virología , Infecciones por Pneumovirus/veterinaria , Pneumovirus/aislamiento & purificación , Infecciones del Sistema Respiratorio/veterinaria , Animales , Perros , Irlanda/epidemiología , Mascotas , Pneumovirus/inmunología , Infecciones por Pneumovirus/epidemiología , Infecciones por Pneumovirus/virología , Infecciones del Sistema Respiratorio/epidemiología , Infecciones del Sistema Respiratorio/virología , Estudios Retrospectivos , Estudios Seroepidemiológicos , Tráquea/virología , Reino Unido/epidemiología
10.
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
11.
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
12.
Pediatr Dev Pathol ; 12(3): 211-6, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19018665

RESUMEN

Infections of the respiratory system are responsible for the majority of hospitalizations and deaths in pediatric patients in developing countries. We selected 177 necropsies of pediatric patients who died as a result of serious respiratory infections. The histopathological findings and epidemiological data were reviewed, and lung tissue samples were separated for immunohistochemistry testing. Conventional immunohistochemistry techniques were used to detect viral antigens in formalin-fixed, paraffin-embedded (FF-PE) lung tissue samples using a pool of monoclonal antibodies against respiratory viruses (respiratory syncytial virus, influenza A and B, adenovirus, and parainfluenza 1, 2, and 3 viruses) as primary antibodies. The histopathological findings were classified into bronchopneumonia (BCP) and interstitial pneumonitis (IP) patterns. The immunohistochemistry results were compared with histopathological patterns and epidemiological data. Positive results for viruses were found in 34% and 62.5% of the BCP and IP cases, respectively. Males and infants below 1 year of age were more frequent in the group that had positive results for viruses. Acute enteritis was the main cause of hospitalization and sepsis the most frequent cause of death in this group. A clear seasonal distribution was observed, with the majority of cases occurring in the 2nd and 3rd trimesters (autumn and winter) of each year in the period studied. Immunohistochemistry is an affordable and easy-to-perform method for viral-antigen detection in FF-PE tissue samples. Although BCP is a classic histopathological pattern found in bacterial infections, it is possible that children with serious respiratory infections had concomitant viral and bacterial infections, regardless of their previous immunologic state.


Asunto(s)
Bronconeumonía/virología , Enfermedades Pulmonares Intersticiales/virología , Infecciones por Pneumovirus/virología , Pneumovirus/aislamiento & purificación , Antígenos Virales/análisis , Biomarcadores/metabolismo , Brasil/epidemiología , Bronconeumonía/epidemiología , Bronconeumonía/patología , Niño , Preescolar , Bases de Datos Factuales , Femenino , Humanos , Inmunohistoquímica/métodos , Lactante , Pulmón/virología , Enfermedades Pulmonares Intersticiales/epidemiología , Enfermedades Pulmonares Intersticiales/patología , Masculino , Pneumovirus/inmunología , Infecciones por Pneumovirus/epidemiología , Infecciones por Pneumovirus/patología
13.
Ann Clin Lab Sci ; 38(1): 41-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18316781

RESUMEN

This study evaluated the clinical usefulness of a newly introduced multiplex reverse transcription PCR assay (Seeplex RV; Seegene, Inc., Seoul, Korea) in patients with respiratory symptoms. Fifty clinical respiratory specimens (45 from children, 5 from adults) were tested for 8 viruses (influenza virus type A and B, parainfluenza virus type 1, 2, 3, respiratory syncytial virus type A and B, and adenovirus) by Seeplex RV (S-RV) and R-mix viral culture with immunofluorescence (VC-IF). Forty (80%) of the 50 samples showed concordant results between S-RV and VC-IF; 24 of these showed the same positive and 16 showed the same negative results. Among the 10 discrepant samples, 9 were S-RV-positive and VC-IF-negative. Six were obtained in patients with lower respiratory tract infection. Only 1 sample was VC-IF-positive and S-RV-negative. This patient had pneumonia. In 3 cases, more than 1 virus was identified by S-RV. The total running time of S-RV was 6 hr, which shortens the detection time for the viral presence by 2 workdays compared to VC-IF. In conclusion, S-RV is reliable, rapid, relatively easy to perform, and able to detect more than 1 virus simultaneously. Therefore, implementation of the S-RV assay in clinical laboratories will aid rapid diagnosis and treatment of major viral infections of the respiratory tract.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Pneumovirus/genética , Pneumovirus/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Adolescente , Adulto , Anciano , Niño , Preescolar , ADN Viral/análisis , ADN Viral/genética , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Pneumovirus/crecimiento & desarrollo , Pneumovirus/inmunología
14.
Vaccine ; 25(6): 1085-95, 2007 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-17052820

RESUMEN

Protective mechanisms underlying the responses to mucosal vaccination are not yet clearly defined. Using the natural mouse pneumovirus pathogen, pneumonia virus of mice (PVM), we explore responses of wild type and interferon-gamma (IFNgamma) receptor gene-deleted mice to virulent challenge after mucosal vaccination with an attenuated virus strain. Serum neutralizing antibodies develop after intranasal inoculation with 30 pfu of attenuated, replication-competent PVM strain 15, which correlate with diminished gross and microscopic pulmonary pathology and protection from weight loss in response to subsequent challenge with the virulent parent PVM strain J3666. Virus replication in response to challenge was blunted in PVM strain 15 vaccinated mice, as was local production of secretory mediators IFNgamma, TNF-alpha, MIP-1 alpha, and MIP-2. Interestingly, responses of vaccinated IFNgamma receptor gene-deleted mice were indistinguishable from those of the wild type, suggesting that IFNgamma signaling may not be crucial for the generation of adaptive responses to pneumovirus infection in vivo.


Asunto(s)
Inmunidad Mucosa/inmunología , Infecciones por Pneumovirus/prevención & control , Pneumovirus/inmunología , Receptores de Interferón/inmunología , Células TH1/inmunología , Vacunas Virales/farmacología , Animales , Quimiocina CCL4 , Quimiocina CXCL2 , Quimiocinas/inmunología , Inmunoglobulina G/inmunología , Pulmón/patología , Proteínas Inflamatorias de Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Infecciones por Pneumovirus/inmunología , Receptores de Interferón/deficiencia , Factor de Necrosis Tumoral alfa/inmunología , Vacunas Virales/inmunología , Receptor de Interferón gamma
15.
Mol Immunol ; 44(5): 993-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16650896

RESUMEN

Internalisation of the plant toxin ricin occurs by retrograde transport which delivers the toxin to the ER where it intersects with the MHC class I system for peptide antigen display. Here, we describe the generation of an inactivated, non-toxic, ricin molecule fused to a peptide which elicits a CD8+ T-cell response in mice directed against pneumonia virus of mice, a pneumovirus related to human respiratory syncytial virus. The ricin fusion elicited a significant T-cell response when delivered by intraperitoneal inoculation in the absence of adjuvent. Challenge experiments showed that the T-cell response resulting from inoculation with the ricin-peptide fusion molecule delayed the onset of virus-induced disease.


Asunto(s)
Infecciones por Pneumovirus/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Ricina/administración & dosificación , Linfocitos T Citotóxicos/inmunología , Proteínas Virales/administración & dosificación , Animales , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Femenino , Glicósidos/farmacología , Ratones , Ratones Endogámicos BALB C , Péptidos/administración & dosificación , Péptidos/genética , Péptidos/inmunología , Pneumovirus/genética , Pneumovirus/inmunología , Triterpenos/farmacología , Proteínas Virales/genética , Proteínas Virales/inmunología
16.
J Immunol ; 175(10): 6597-604, 2005 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16272314

RESUMEN

Pneumonia virus of mice (PVM) is a natural pathogen of mice and has been proposed as a tractable model for the replication of a pneumovirus in its natural host, which mimics human infection with human respiratory syncytial virus (RSV). PVM infection in mice is highly productive in terms of virus production compared with the situation seen with RSV in mice. Because RSV suppresses CD8 T cell effector function in the lungs of infected mice, we have investigated the nature of PVM-induced CD8 T cell responses to study pneumovirus-induced T cell responses in a natural virus-host setting. PVM infection was associated with a massive influx of activated CD8 T cells into the lungs. After identification of three PVM-specific CD8 T cell epitopes, pulmonary CD8 T cell responses were enumerated. The combined frequency of cytokine-secreting CD8 T cells specific for the three epitopes was much smaller than the total number of activated CD8 T cells. Furthermore, quantitation of the CD8 T cell response against one of these epitopes (residues 261-270 from the phosphoprotein) by MHC class I pentamer staining and by in vitro stimulation followed by intracellular IFN-gamma and TNF-alpha staining indicated that the majority of pulmonary CD8 specific for the P261 epitope were deficient in cytokine production. This deficient phenotype was retained up to 96 days postinfection, similar to the situation in the lungs of human RSV-infected mice. The data suggest that PVM suppresses T cell effector functions in the lungs.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por Pneumovirus/inmunología , Pneumovirus/inmunología , Secuencia de Aminoácidos , Animales , Antígenos Virales/genética , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Epítopos/genética , Femenino , Humanos , Memoria Inmunológica , Técnicas In Vitro , Interferón gamma/biosíntesis , Pulmón/inmunología , Pulmón/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Pneumovirus/genética , Infecciones por Pneumovirus/patología , Infecciones por Pneumovirus/virología , Bazo/inmunología , Bazo/patología , Factor de Necrosis Tumoral alfa/biosíntesis
17.
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
18.
Avian Dis ; 47(1): 175-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12713174

RESUMEN

Avian paramyxoviruses (PMVs) and avian pneumovirus (APV) belong to the family Paramyxoviridae. Antigenic relationships between PMVs were shown previously, hence, this study was designed to investigate possible antigenic relationships between APV and four avian PMVs (PMV-1, PMV-2, PMV-3, and PMV-7). Enzyme-linked immunosorbent assay (ELISA), hemagglutination inhibition (HI) test, and virus neutralization (VN) test in chicken embryos and in Vero cells were used. The HI test was performed with the PMVs as antigens against the APV and PMVs antisera. The ELISA and VN test in chicken embryos were performed with PMVs and APV antigens and antisera. The VN test in vero cells was performed with the APV as an antigen against the PMV antisera. All the viruses were isolated in the United States or Canada. No antigenic relationships between APV and the PMVs were detected by the described tests.


Asunto(s)
Antígenos Virales/inmunología , Avulavirus/inmunología , Pneumovirus/inmunología , Enfermedades de las Aves de Corral/virología , Animales , Antígenos Virales/análisis , Embrión de Pollo , Chlorocebus aethiops , Ensayo de Inmunoadsorción Enzimática , Pruebas de Inhibición de Hemaglutinación , Pruebas de Hemaglutinación , Pruebas de Neutralización , Pavos , Células Vero
19.
Vaccine ; 21(13-14): 1355-62, 2003 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-12615430

RESUMEN

Fertile turkey eggs after 24 days of incubation were vaccinated in ovo with a commercial live attenuated subtype A avian pneumovirus (APV) vaccine. Hatchability was not adversely affected. When a high dose (10 times maximum commercial dose) of vaccine was tested in maternal antibody negative (MA-) eggs, mild clinical signs developed in a small proportion of the poults for 1-4 days only. Post-vaccination antibody titres at 3 weeks of age were significantly higher than those seen when the same dose was administered by eyedrop or spray at day-old. A low dose (end of shelf-life titre) of vaccine given to MA- eggs did not cause disease and vaccinated poults were 100% protected against virulent APV challenge at 3 or 5 weeks of age. Post-vaccination antibody titres reached significant levels at 3 weeks of age, whereas those from MA- poults vaccinated by spray at day-old with a similar low dose did not. In a 'worst-case' scenario, maternal antibody positive (MA+) poults vaccinated in ovo with the low dose were still 77% protected against clinical disease, despite lack of seroconversion. The recommended commercial dose of vaccine given to MA- eggs in ovo induced 100% protection against virulent APV challenge for up to 14 weeks of age, even though post-vaccination antibody titres had dropped to insignificant levels at this age. In ovo vaccination with a mixture of the recommended commercial doses of live APV and Newcastle disease (ND) vaccines had no detrimental affect on the efficacy of the APV vaccine. This is the first report of the successful use of an APV vaccine being given in ovo. The results indicate that for turkeys, in ovo vaccination with a live attenuated APV vaccine is safe and effective against virulent challenge and comparable with vaccination by conventional methods.


Asunto(s)
Infecciones por Pneumovirus/veterinaria , Pneumovirus/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunación/veterinaria , Vacunas Virales/inmunología , Animales , Huevos , Virus de la Enfermedad de Newcastle/inmunología , Infecciones por Pneumovirus/prevención & control , Factores de Tiempo , Pavos , Vacunas Atenuadas
20.
Vaccine ; 21(13-14): 1371-4, 2003 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-12615432

RESUMEN

We report the development of a cold adapted strain of avian pneumovirus (APV) and its evaluation as a live vaccine candidate in 2-week-old turkey poults. A US isolate of APV (APV/MN/turkey/1-a/97) was serially passaged in Vero cells for 41 passages and then adapted to grow at sub-optimal temperatures by growing successively at 35, 33 and 31 degrees C for eight passages at each temperature. The virus thus adapted to grow at 31 degrees C was used as a candidate vaccine. The birds were vaccinated with two different doses of cold adapted virus and challenged with virulent virus 2 weeks after vaccination. No clinical signs were observed post-vaccination. Upon challenge, no clinical signs were seen in vaccinated birds but severe clinical signs were seen in non-vaccinated, challenged birds. The signs included unilateral or bilateral mucoid nasal discharge, watery eyes and swelling of infraorbital sinuses. The antibody levels in vaccinated birds were not very high. None of the vaccinated birds were found to shed virus after challenge in their choanal secretions whereas all of the non-vaccinated, challenged birds shed the virus. The absence of clinical signs and virus shedding in vaccinated birds as compared to that in non-vaccinated birds suggests that the cold adapted strain of APV is a viable candidate for use as a live, attenuated vaccine in turkeys.


Asunto(s)
Infecciones por Pneumovirus/veterinaria , Pneumovirus/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunación/veterinaria , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Frío , Infecciones por Pneumovirus/prevención & control , Pavos , Vacunas Atenuadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...