Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 14(1)2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-35062301

RESUMO

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.


Assuntos
Patrimônio Genético , Infecções por Herpesviridae/imunologia , Macrófagos Alveolares/imunologia , Infecções por Pneumovirus/imunologia , Pneumovirus/imunologia , Rhadinovirus/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Feminino , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Imunidade Inata , Inflamação/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Infecções Pneumocócicas/imunologia , Pneumovirus/fisiologia , Infecções por Pneumovirus/genética , Infecções por Pneumovirus/patologia , Infecções por Pneumovirus/virologia , Rhadinovirus/fisiologia
2.
mBio ; 12(6): e0262121, 2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34724816

RESUMO

Multiple enveloped RNA viruses of the family Paramyxoviridae and Pneumoviridae, like measles virus (MeV), Nipah virus (NiV), canine distemper virus (CDV), or respiratory syncytial virus (RSV), are of high clinical relevance. Each year a huge number of lives are lost as a result of these viral infections. Worldwide, MeV infection alone is responsible for over a hundred thousand deaths each year despite available vaccine. Therefore, there is an urgent need for treatment options to counteract these viral infections. The development of antiviral drugs in general stands as a huge challenge due to the rapid emergence of viral escape mutants. Here, we disclose the discovery of a small-molecule antiviral, compound 1 (ZHAWOC9045), active against several pneumo-/paramyxoviruses, including MeV, NiV, CDV, RSV, and parainfluenza virus type 5 (PIV-5). A series of mechanistic characterizations revealed that compound 1 targets a host factor which is indispensable for viral genome replication. Drug resistance profiling against a paramyxovirus model (CDV) demonstrated no detectable adaptation despite prolonged time of investigation, thereby mitigating the rapid emergence of escape variants. Furthermore, a thorough structure-activity relationship analysis of compound 1 led to the invention of 100-times-more potent-derivatives, e.g., compound 2 (ZHAWOC21026). Collectively, we present in this study an attractive host-directed pneumoviral/paramyxoviral replication inhibitor with potential therapeutic application. IMPORTANCE Measles virus, respiratory syncytial virus, canine distemper virus, and Nipah virus are some of the clinically significant RNA viruses that threaten substantial number of lives each year. Limited to no availability of treatment options for these viral infections makes it arduous to handle the outbreaks. This highlights the major importance of developing antivirals to fight not only ongoing infections but also potential future epidemics. Most of the discovered antivirals, in clinical trials currently, are virus targeted, which consequently poses the challenge of rapid emergence of escape variants. Here, we present compound 1 (ZHAWOC9045), discovered to target viral replication in a host-dependent manner, thereby exhibiting broad-spectrum activity against several members of the family Pneumo-/Paramyxoviridae. The inability of viruses to mutate against the inhibitor mitigated the critical issue of generation of escape variants. Importantly, compound 1 was successfully optimized to a highly potent variant, compound 2 (ZHAWOC21026), with a promising profile for pharmacological intervention.


Assuntos
Antivirais/farmacologia , Paramyxoviridae/fisiologia , Pneumovirus/fisiologia , Replicação Viral/efeitos dos fármacos , Antivirais/química , Descoberta de Drogas , Humanos , Paramyxoviridae/genética , Infecções por Paramyxoviridae/tratamento farmacológico , Infecções por Paramyxoviridae/virologia , Pneumovirus/genética , Infecções por Pneumovirus/tratamento farmacológico , Infecções por Pneumovirus/virologia
3.
Viruses ; 12(12)2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33276587

RESUMO

The paramyxo- and pneumovirus family includes a wide range of viruses that can cause respiratory and/or systemic infections in humans and animals. The significant disease burden of these viruses is further exacerbated by the limited therapeutics that are currently available. Host cellular proteins that can antagonize or limit virus replication are therefore a promising area of research to identify candidate molecules with the potential for host-targeted therapies. Host proteins known as host cell restriction factors are constitutively expressed and/or induced in response to virus infection and include proteins from interferon-stimulated genes (ISGs). Many ISG proteins have been identified but relatively few have been characterized in detail and most studies have focused on studying their antiviral activities against particular viruses, such as influenza A viruses and human immunodeficiency virus (HIV)-1. This review summarizes current literature regarding host cell restriction factors against paramyxo- and pneumoviruses, on which there is more limited data. Alongside discussion of known restriction factors, this review also considers viral countermeasures in overcoming host restriction, the strengths and limitations in different experimental approaches in studies reported to date, and the challenges in reconciling differences between in vitro and in vivo data. Furthermore, this review provides an outlook regarding the landscape of emerging technologies and tools available to study host cell restriction factors, as well as the suitability of these proteins as targets for broad-spectrum antiviral therapeutics.


Assuntos
Interações Hospedeiro-Patógeno , Infecções por Paramyxoviridae/virologia , Paramyxovirinae/fisiologia , Infecções por Pneumovirus/virologia , Pneumovirus/fisiologia , Animais , Biomarcadores , Regulação Viral da Expressão Gênica , Especificidade de Hospedeiro , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata , Infecções por Paramyxoviridae/genética , Infecções por Paramyxoviridae/metabolismo , Infecções por Pneumovirus/genética , Infecções por Pneumovirus/metabolismo , Tropismo Viral , Replicação Viral
4.
Viruses ; 12(3)2020 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-32245118

RESUMO

Paramyxoviruses and pneumoviruses infect cells through fusion (F) protein-mediated merger of the viral envelope with target membranes. Members of these families include a range of major human and animal pathogens, such as respiratory syncytial virus (RSV), measles virus (MeV), human parainfluenza viruses (HPIVs), and highly pathogenic Nipah virus (NiV). High-resolution F protein structures in both the metastable pre- and the postfusion conformation have been solved for several members of the families and a number of F-targeting entry inhibitors have progressed to advanced development or clinical testing. However, small-molecule RSV entry inhibitors have overall disappointed in clinical trials and viral resistance developed rapidly in experimental settings and patients, raising the question of whether the available structural information may provide a path to counteract viral escape through proactive inhibitor engineering. This article will summarize current mechanistic insight into F-mediated membrane fusion and examine the contribution of structural information to the development of small-molecule F inhibitors. Implications are outlined for future drug target selection and rational drug engineering strategies.


Assuntos
Antivirais/química , Antivirais/farmacologia , Descoberta de Drogas , Paramyxovirinae/fisiologia , Pneumovirus/fisiologia , Internalização do Vírus/efeitos dos fármacos , Animais , Sítios de Ligação , Descoberta de Drogas/métodos , Humanos , Modelos Moleculares , Infecções por Paramyxoviridae/tratamento farmacológico , Infecções por Paramyxoviridae/virologia , Paramyxovirinae/efeitos dos fármacos , Pneumovirus/efeitos dos fármacos , Infecções por Pneumovirus/tratamento farmacológico , Infecções por Pneumovirus/virologia , Ligação Proteica , Relação Estrutura-Atividade
5.
Virus Res ; 265: 68-73, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30844414

RESUMO

Pneumoviruses represent a major public health burden across the world. Respiratory syncytial virus (RSV) and human metapneumovirus (HMPV), two of the most recognizable pediatric infectious agents, belong to this family. These viruses are enveloped with a non-segmented negative-sense RNA genome, and their replication occurs in specialized cytosolic organelles named inclusion bodies (IB). The critical role of IBs in replication of pneumoviruses has begun to be elucidated, and our current understanding suggests they are highly dynamic structures. From IBs, newly synthesized nucleocapsids are transported to assembly sites, potentially via the actin cytoskeleton, to be incorporated into nascent virions. Released virions, which generally contain one genome, can then diffuse in the extracellular environment to target new cells and reinitiate the process of infection. This is a challenging business for virions, which must face several risks including the extracellular immune responses. In addition, several recent studies suggest that successful infection may be achieved more rapidly by multiple, rather than single, genomic copies being deposited into a target cell. Interestingly, recent data indicate that pneumoviruses have several mechanisms that permit their transmission en bloc, i.e. transmission of multiple genomes at the same time. These mechanisms include the well-studied syncytia formation as well as the newly described formation of long actin-based intercellular extensions. These not only permit en bloc viral transmission, but also bypass assembly of complete virions. In this review we describe several aspects of en bloc viral transmission and how these mechanisms are reshaping our understanding of pneumovirus replication, assembly and spread.


Assuntos
Infecções por Paramyxoviridae/transmissão , Pneumovirus/fisiologia , Montagem de Vírus , Animais , Linhagem Celular , Humanos , Metapneumovirus/genética , Metapneumovirus/fisiologia , Camundongos , Pneumovirus/genética , RNA Viral , Vírion/genética , Vírion/fisiologia , Replicação Viral
6.
Virus Res ; 234: 87-102, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28104450

RESUMO

The paramyxo- and pneumoviruses are members of the order Mononegavirales, a group of viruses with non-segmented, negative strand RNA genomes. The polymerases of these viruses are multi-functional complexes, capable of transcribing subgenomic capped and polyadenylated mRNAs and replicating the genome. Although there is no native structure available for any complete paramyxo- or pneumovirus polymerase, functional and structural studies of a fragment of a pneumovirus polymerase protein and mutation analyses and resistance profiling of small-molecule inhibitors have generated a wealth of mechanistic information. This review integrates these data with the structure of a related polymerase, identifying similarities, differences, gaps in knowledge, and avenues for antiviral drug development.


Assuntos
Paramyxoviridae/enzimologia , Pneumovirus/enzimologia , RNA Polimerase Dependente de RNA/metabolismo , Análise Mutacional de DNA , Farmacorresistência Viral , Mutação de Sentido Incorreto , Pneumovirus/fisiologia , RNA Polimerase Dependente de RNA/genética , Transcrição Gênica , Replicação Viral
7.
Immunol Lett ; 172: 106-12, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-26916143

RESUMO

The SV-40-transformed MH-S cell line maintains some, but not all, features of primary alveolar macrophages (AMs) from BALB/c mice. We show here that MH-S cells produce inflammatory cytokines IL-6 and CXCL10 in response to challenge with Gram-positive Lactobacillus reuteri, and to TLR2 and NOD2 ligands Pam3CSK4 and MDP, respectively. In contrast, although wild-type AMs are infected in vivo by pneumonia virus of mice (PVM), no virus replication was detected in MH-S cells. Interestingly, the surface immunophenotype of MH-S cells (CD11c(+)Siglec F(-)) differs from that of wild-type AMs (CD11c(+) Siglec F(+)) and is similar to that of immature AMs isolated from granulocyte macrophage-colony stimulating factor (GM-CSF) gene-deleted mice; AMs from GM-CSF(-/-) mice also support PVM replication. However, MH-S cells do not express the GM-CSF receptor alpha chain (CD116) and do not respond to GM-CSF. Due to these unusual features, MH-S cells should be used with caution as experimental models of AMs.


Assuntos
Limosilactobacillus reuteri/imunologia , Macrófagos Alveolares/virologia , Infecções por Pneumovirus/imunologia , Pneumovirus/fisiologia , Animais , Linhagem Celular Transformada , Quimiocina CXCL10/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Interleucina-6/metabolismo , Lipopeptídeos/imunologia , Macrófagos Alveolares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptor 2 Toll-Like/metabolismo , Replicação Viral
8.
PLoS One ; 10(4): e0123755, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25893441

RESUMO

This study aimed to determine the role of influenza-like illness (ILI) surveillance conducted on Leyte Island, the Philippines, including involvement of other respiratory viruses, from 2010 to 2013. ILI surveillance was conducted from January 2010 to March 2013 with 3 sentinel sites located in Tacloban city, Palo and Tanauan of Leyte Island. ILI was defined as fever ≥38°C or feverish feeling and either cough or running nose in a patient of any age. Influenza virus and other 5 respiratory viruses were searched. A total of 5,550 ILI cases visited the 3 sites and specimens were collected from 2,031 (36.6%) cases. Among the cases sampled, 1,637 (75.6%) were children aged <5 years. 874 (43.0%) cases were positive for at least one of the respiratory viruses tested. Influenza virus and respiratory syncytial virus (RSV) were predominantly detected (both were 25.7%) followed by human rhinovirus (HRV) (17.5%). The age distributions were significantly different between those who were positive for influenza, HRV, and RSV. ILI cases were reported throughout the year and influenza virus was co-detected with those viruses on approximately half of the weeks of study period (RSV in 60.5% and HRV 47.4%). In terms of clinical manifestations, only the rates of headache and sore throat were significantly higher in influenza positive cases than cases positive to other viruses. In conclusion, syndromic ILI surveillance in this area is difficult to detect the start of influenza epidemic without laboratory confirmation which requires huge resources. Age was an important factor that affected positive rates of influenza and other respiratory viruses. Involvement of older age children may be useful to detect influenza more effectively.


Assuntos
Influenza Humana/epidemiologia , Influenza Humana/virologia , Ilhas/epidemiologia , Orthomyxoviridae/fisiologia , Pneumovirus/fisiologia , Vigilância da População , Adolescente , Adulto , Criança , Pré-Escolar , Demografia , Feminino , Geografia , Humanos , Lactente , Recém-Nascido , Masculino , Filipinas/epidemiologia , Adulto Jovem
9.
Clin Vaccine Immunol ; 22(5): 477-83, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25716232

RESUMO

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.


Assuntos
Bronquiolite Viral/imunologia , Coinfecção , Interferon beta/imunologia , Pulmão/virologia , Malária/imunologia , Infecções por Pneumovirus/imunologia , Pneumovirus/imunologia , Animais , Bronquiolite Viral/virologia , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/parasitologia , Inflamação/virologia , Interferon beta/sangue , Interleucina-10/imunologia , Pulmão/imunologia , Malária/complicações , Plasmodium chabaudi , Pneumovirus/patogenicidade , Pneumovirus/fisiologia , Infecções por Pneumovirus/complicações , Vírus Sincicial Respiratório Humano/patogenicidade , Carga Viral , Redução de Peso
10.
Viruses ; 5(1): 406-22, 2013 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-23344499

RESUMO

Pneumovirus infections cause a wide spectrum of respiratory disease in humans and animals. The airway epithelium is the major site of pneumovirus replication. Apoptosis or regulated cell death, may contribute to the host anti-viral response by limiting viral replication. However, apoptosis of lung epithelial cells may also exacerbate lung injury, depending on the extent, the timing and specific location in the lungs. Differential apoptotic responses of epithelial cells versus innate immune cells (e.g., neutrophils, macrophages) during pneumovirus infection can further contribute to the complex and delicate balance between host defense and disease pathogenesis. The purpose of this manuscript is to give an overview of the role of apoptosis in pneumovirus infection. We will examine clinical and experimental data concerning the various pro-apoptotic stimuli and the roles of apoptotic epithelial and innate immune cells during pneumovirus disease. Finally, we will discuss potential therapeutic interventions targeting apoptosis in the lungs.


Assuntos
Apoptose , Infecções por Pneumovirus/fisiopatologia , Pneumovirus/fisiologia , Animais , Antivirais/farmacologia , Humanos , Pneumovirus/efeitos dos fármacos , Pneumovirus/genética , Infecções por Pneumovirus/tratamento farmacológico , Infecções por Pneumovirus/imunologia , Infecções por Pneumovirus/virologia
11.
Virology ; 416(1-2): 26-31, 2011 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-21600624

RESUMO

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.


Assuntos
Inflamação/virologia , Pulmão/virologia , Infecções por Pneumovirus/virologia , Pneumovirus/classificação , Pneumovirus/fisiologia , Replicação Viral/fisiologia , Animais , Cães , Inflamação/patologia , Camundongos , Camundongos Endogâmicos BALB C , Filogenia , Pneumovirus/genética , Infecções por Pneumovirus/patologia
12.
Blood ; 114(13): 2649-56, 2009 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-19652202

RESUMO

Eosinophils are recruited to the lung in response to infection with pneumovirus pathogens and have been associated with both the pathophysiologic sequelae of infection and, more recently, with accelerated virus clearance. Here, we demonstrate that the pneumovirus pathogens, respiratory syncytial virus (RSV) and pneumonia virus of mice (PVM), can infect human and mouse eosinophils, respectively, and that virus infection of eosinophils elicits the release of disease-related proinflammatory mediators from eosinophils. RSV replication in human eosinophils results in the release of infectious virions and in the release of the proinflammatory mediator, interleukin-6 (IL-6). PVM replication in cultured bone marrow eosinophils (bmEos) likewise results in release of infectious virions and the proinflammatory mediators IL-6, IP-10, CCL2, and CCL3. In contrast to the findings reported in lung tissue of RSV-challenged mice, PVM replication is accelerated in MyD88 gene-deleted bmEos, whereas release of cytokines is diminished. Interestingly, exogenous IL-6 suppresses virus replication in MyD88 gene-deleted bmEos, suggesting a role for a MyD88-dependent cytokine-mediated feedback circuit in modulating this response. Taken together, our findings suggest that eosinophils are targets of virus infection and may have varied and complex contributions to the pathogenesis and resolution of pneumovirus disease.


Assuntos
Quimiocinas/metabolismo , Eosinófilos/metabolismo , Interleucina-6/metabolismo , Fator 88 de Diferenciação Mieloide/fisiologia , Infecções por Pneumovirus/imunologia , Pneumovirus/fisiologia , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/virologia , Fatores Quimiotáticos/metabolismo , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/virologia , Interleucina-6/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Infecções por Pneumovirus/genética , Infecções por Pneumovirus/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética , Replicação Viral/fisiologia , Eliminação de Partículas Virais/efeitos dos fármacos , Eliminação de Partículas Virais/genética , Eliminação de Partículas Virais/fisiologia
13.
Immunol Lett ; 118(1): 6-12, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18471897

RESUMO

Pneumonia virus of mice (PVM; family Paramyxoviridae, genus Pneumovirus) is a natural mouse pathogen that is closely related to human and bovine respiratory syncytial viruses. Among the prominent features of this infection, robust replication of PVM takes place in bronchial epithelial cells in response to a minimal virus inoculum. Virus replication in situ results in local production of proinflammatory cytokines (MIP-1alpha, MIP-2, MCP-1 and IFNgamma) and granulocyte recruitment to the lung. If left unchecked, PVM infection and the ensuing inflammatory response ultimately lead to pulmonary edema, respiratory compromise and death. In this review, we consider the recent studies using the PVM model that have provided important insights into the role of the inflammatory response in the pathogenesis of severe respiratory virus infection. We also highlight several works that have elucidated acquired immune responses to this pathogen, including T cell responses and the development of humoral immunity. Finally, we consider several immunomodulatory strategies that have been used successfully to reduce morbidity and mortality when administered to PVM-infected, symptomatic mice, and thus hold promise as realistic therapeutic strategies for severe respiratory virus infections in human subjects.


Assuntos
Pneumovirus/fisiologia , Infecções Respiratórias/virologia , Animais , Genoma Viral/genética , Humanos , Hipersensibilidade/imunologia , Camundongos , Pneumovirus/patogenicidade , Infecções por Pneumovirus/tratamento farmacológico , Infecções por Pneumovirus/imunologia , Infecções por Pneumovirus/metabolismo , Infecções por Pneumovirus/virologia , Infecções Respiratórias/tratamento farmacológico , Infecções Respiratórias/imunologia , Infecções Respiratórias/metabolismo , Linfócitos T/imunologia
14.
J Immunol ; 175(7): 4735-44, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-16177121

RESUMO

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.


Assuntos
Deleção de Genes , Pulmão/patologia , Infecções por Pneumovirus/imunologia , Pneumovirus/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Animais , Quimiocina CCL3 , Quimiocina CCL4 , Quimiocina CXCL10 , Quimiocina CXCL2 , Quimiocinas/biossíntese , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Inflamação/genética , Inflamação/imunologia , Inflamação/virologia , Interferon Tipo I/biossíntese , Interferon Tipo I/genética , Leucócitos/patologia , Pulmão/imunologia , Pulmão/virologia , Proteínas Inflamatórias de Macrófagos/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quimioatraentes de Monócitos/biossíntese , Proteínas Quimioatraentes de Monócitos/genética , Pneumovirus/fisiologia , Infecções por Pneumovirus/mortalidade , Infecções por Pneumovirus/patologia , RNA Mensageiro/metabolismo , Replicação Viral/fisiologia
15.
Artigo em Inglês | MEDLINE | ID: mdl-12069273

RESUMO

The effects of testosterone, oestradiol, progesterone and cortisone on the in vitro replication of avian pneumovirus in tracheal organ cultures (TOC) were investigated. Samples of cell-associated and cell-free virus from TOC, grown in medium containing these hormones, were taken at selected intervals. Progesterone and cortisone caused a slight increase in cell-associated virus. Testosterone and oestradiol caused a slight delay and decrease in virus replication when compared with the controls. All groups shared the same time interval to reach peak cell-free virus titre, 96 h post inoculation. In comparison with the controls, only a small drop (0.25-0.50 log10) in the peak of virus titre was observed in the hormone treated groups.


Assuntos
Pneumovirus/efeitos dos fármacos , Esteroides/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Cortisona/farmacologia , Estradiol/farmacologia , Pneumovirus/fisiologia , Progesterona/farmacologia , Testosterona/farmacologia , Traqueia/microbiologia
16.
J Virol ; 75(14): 6265-72, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11413292

RESUMO

The specificity of usage of promoters for replication and transcription by the pneumoviruses human respiratory syncytial virus (HRSV) and avian pneumovirus (APV) was studied using minigenomes containing a reporter gene. When infectious HRSV or APV was used as helper virus, replication could occur only if both the leader and trailer regions (containing the replicative and transcriptional promoters) were derived from the helper virus. In contrast, when the HRSV replication complex was supplied from cDNA plasmids, a minigenome containing either the APV leader or trailer was recognized and substantial levels of replication and transcription occurred. These data suggest that in pneumovirus-infected cells, helper virus functions can discriminate between genomes on the basis of the terminal sequences and that there is an association between the leader and trailer required for productive replication. This association is required only in virus-infected cells, not when replication and transcription are mediated by plasmid-directed expression of the component proteins required for replication and transcription. The possible implications of this are discussed.


Assuntos
Pneumovirus/fisiologia , Vírus Sinciciais Respiratórios/fisiologia , Transcrição Gênica , Replicação Viral , Animais , Aves/virologia , Northern Blotting , Linhagem Celular , Genes Reporter , Genes Virais , Genoma Viral , Vírus Auxiliares/genética , Humanos , Pneumovirus/genética , RNA Viral/análise , Sequências Reguladoras de Ácido Nucleico , Vírus Sinciciais Respiratórios/genética , Especificidade da Espécie , Transfecção
17.
Cell Immunol ; 206(1): 1-6, 2000 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-11161432

RESUMO

The intent of this study was to compare the cellular and biochemical inflammatory responses of mice infected with the paramyxovirus pathogens respiratory syncytial virus (RSV) and pneumonia virus of mice (PVM). Although RSV is not a natural pathogen of mice, it has been used extensively in mouse models of the human disease, as a limited respiratory infection can be established via intranasal inoculation of virus at high titer. In earlier work, we found that acute infection with the natural rodent pathogen, PVM, elicited a rapid and sustained pulmonary inflammatory response (peak, 1.7 x 10(6) leukocytes/ml BAL fluid) that was dependent on both local production of MIP-1alpha and signaling via its receptor, CCR1. We find here that MIP-1alpha is also produced in response to RSV, although relatively few leukocytes (<200 ml BAL fluid) are recruited to the lungs in response. Further experiments with CCR1-deficient mice confirm the finding that although MIP-1alpha is produced in response to RSV infection, leukocytes do not respond via this pathway. Among the explanations for these findings, we propose that there are other, as yet to be identified proinflammatory mediators elicited in response to PVM (but not in response to RSV) that serve to prime the leukocytes in vivo, thus enabling them to respond to MIP-1alpha signaling via CCR1. Furthermore, the differences in disease pathogenesis seen in response to each of these pneumovirus infections in mice raise questions regarding the extent to which primary RSV infection in mice can be used as a model of primary RSV infection in humans.


Assuntos
Proteínas Inflamatórias de Macrófagos/fisiologia , Pneumonia Viral/fisiopatologia , Pneumovirus/fisiologia , Infecções por Vírus Respiratório Sincicial/fisiopatologia , Vírus Sinciciais Respiratórios/fisiologia , Doença Aguda , Animais , Líquido da Lavagem Broncoalveolar/citologia , Quimiocina CCL3 , Quimiocina CCL4 , Modelos Animais de Doenças , Regulação Viral da Expressão Gênica , Humanos , Contagem de Leucócitos , Proteínas Inflamatórias de Macrófagos/biossíntese , Proteínas Inflamatórias de Macrófagos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Viral/metabolismo , Receptores CCR1 , Receptores de Quimiocinas/fisiologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Especificidade da Espécie , Organismos Livres de Patógenos Específicos
18.
Res Vet Sci ; 66(2): 161-3, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10208895

RESUMO

One-day-old poults or two-week old chicks were infected oculonasally with avian pneumovirus. Cloacal swabs were collected for virus isolation as were selected tissues (Harderian gland, turbinates, trachea, lungs and kidneys) from birds killed at regular intervals up to 33 days post infection (p.i.) for poults, and up to 40 days p. i. for chicks. In an attempt to induce virus re-excretion, the T-cell-suppressor cyclosporin A (CSA) was given for 12 days starting from three weeks p.i. in poults and from four weeks p.i. in chicks. Birds were sampled for virus isolations up to day 12 post CSA treatment. Virus was recovered only up to day nine p.i. in poults, and day five p.i. in chicks during the acute phase of the infection. Despite T-cell suppression, there was no evidence of re-excretion of the virus, and hence no evidence for the persistence of virus in the tissues examined.


Assuntos
Ciclosporina/farmacologia , Imunossupressores/farmacologia , Infecções por Pneumovirus/veterinária , Pneumovirus/isolamento & purificação , Doenças das Aves Domésticas/fisiopatologia , Envelhecimento , Animais , Galinhas , Cloaca/virologia , Glândula de Harder/virologia , Rim/virologia , Pulmão/virologia , Pneumovirus/fisiologia , Infecções por Pneumovirus/imunologia , Infecções por Pneumovirus/fisiopatologia , Doenças das Aves Domésticas/imunologia , Fatores de Tempo , Traqueia/virologia , Conchas Nasais/virologia
19.
Avian Dis ; 40(4): 858-64, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8980818

RESUMO

Ornithobacterium rhinotracheale was found to cause growth retardation in both turkeys and chickens after experimental intra-air sac administration and to cause growth retardation together with airsacculitis and pneumonia after aerosol administration. Both turkey and chicken isolates of O. rhinotracheale were able to induce the same kind of respiratory inflammations and weight-gain losses in chickens as well as turkeys. Turkey rhinotracheitis virus was found to have a triggering effect on the O. rhinotracheale infection in turkeys, and Newcastle disease virus and to a lesser extent infectious bronchitis virus showed triggering effects on the O. rhinotracheale infection in chickens. Ornithobacterium rhinotracheale could be reisolated from affected organs of experimentally infected birds.


Assuntos
Galinhas/microbiologia , Bactérias Gram-Negativas/isolamento & purificação , Infecções por Bactérias Gram-Negativas/veterinária , Doenças das Aves Domésticas/patologia , Perus/microbiologia , Aerossóis , Animais , Galinhas/virologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/fisiopatologia , Infecções por Coronavirus/veterinária , Bactérias Gram-Negativas/fisiologia , Infecções por Bactérias Gram-Negativas/patologia , Infecções por Bactérias Gram-Negativas/fisiopatologia , Transtornos do Crescimento/etiologia , Transtornos do Crescimento/patologia , Transtornos do Crescimento/veterinária , Vírus da Bronquite Infecciosa/isolamento & purificação , Vírus da Bronquite Infecciosa/fisiologia , Pulmão/microbiologia , Pulmão/patologia , Pulmão/virologia , Doença de Newcastle/patologia , Doença de Newcastle/fisiopatologia , Vírus da Doença de Newcastle/isolamento & purificação , Vírus da Doença de Newcastle/fisiologia , Pneumovirus/isolamento & purificação , Pneumovirus/fisiologia , Infecções por Pneumovirus/patologia , Infecções por Pneumovirus/fisiopatologia , Infecções por Pneumovirus/veterinária , Doenças das Aves Domésticas/etiologia , Doenças das Aves Domésticas/fisiopatologia , Traqueia/microbiologia , Traqueia/patologia , Traqueia/virologia , Perus/virologia , Aumento de Peso/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...