Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 95(20): e0083721, 2021 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-34319159

RESUMO

Interferon-induced transmembrane (IFITM) proteins inhibit a broad range of enveloped viruses by blocking entry into host cells. We used an inducible overexpression system to investigate if IFITM1, IFITM2, and IFITM3 could modulate early and/or late stages of influenza A virus (IAV) or parainfluenza virus 3 (PIV-3) infection in human A549 airway epithelial cells. IAV and PIV-3 represent respiratory viruses which utilize distinct cellular entry pathways. We verify entry by endocytosis for IAV, whereas PIV-3 infection was consistent with fusion at the plasma membrane. Following induction prior to infection, all three IFITM proteins restricted the percentage of IAV-infected cells at 8 hours postinfection. In contrast, prior induction of IFITM1 and IFITM2 did not inhibit PIV-3 infection, although a modest reduction was observed with IFITM3. Small interfering RNA (siRNA)-mediated knockdown of endogenous IFITM1, IFITM2, and IFITM3 expression, in the presence or absence of pretreatment with type I interferon, resulted in increased IAV, but not PIV-3, infection. This finding suggests that while all three IFITMs display antiviral activity against IAV, they do not restrict the early stages of PIV-3 infection. IAV and PIV-3 infection culminates in viral egress through budding at the plasma membrane. Inducible expression of IFITM1, IFITM2, or IFITM3 immediately after infection did not impact titers of infectious virus released from IAV- or PIV-3-infected cells. Our findings show that IFITM proteins differentially restrict the early stages of infection of two respiratory viruses with distinct cellular entry pathways but do not influence the late stages of replication for either virus. IMPORTANCE Interferon-induced transmembrane (IFITM) proteins restrict the initial stages of infection for several respiratory viruses; however, their potential to modulate the later stages of virus replication has not been explored. In this study, we highlight the utility of an inducible overexpression system to assess the impact of IFITM proteins on either early- or late-stage replication of two respiratory viruses. We demonstrate antiviral activity by IFITM1, IFITM2, and IFITM3 against influenza A virus (IAV) but not parainfluenza virus 3 (PIV-3) during the early stages of cellular infection. Furthermore, IFITM induction following IAV or PIV-3 infection does not restrict the late stages of replication of either virus. Our findings show that IFITM proteins can differentially restrict the early stages of infection of two viruses with distinct cellular entry pathways and yet do not influence the late stages of replication for either virus.


Assuntos
Viroses/metabolismo , Replicação Viral/fisiologia , Células A549 , Antígenos de Diferenciação/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Endocitose/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Vírus da Influenza A/metabolismo , Vírus da Influenza A/patogenicidade , Interferons/metabolismo , Proteínas de Membrana/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Proteínas de Ligação a RNA/metabolismo , Internalização do Vírus
2.
J Med Virol ; 93(11): 6371-6376, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34324206

RESUMO

Human parainfluenza virus type 3 (HPIV-3) may cause lower respiratory tract infection disease (LRTI-D) after hematopoietic stem cell transplantation (HSCT). Most previous have studies focused on recipients of HSCT whereas data on characteristics and outcomes in patients with hematological malignancies (HMs) compared to non-hematological patients are limited. The prognostic value of viral load in respiratory specimens remains elusive. In a 2-year retrospective study, we determined the frequencies of LRTI-D in HM, HSCT, and in non-hematological patients, and HPIV-3 levels in respiratory tract secretions. Among 98 patients with HPIV-3 infection, including 31 HSCT and 40 HM, 36 had a diagnosis of LRTI-D. LRTI-D was significantly more frequent in patients with HM or HSCT (n = 32, 45.1%) than in non-hematological patients (n = 4, 14.8%) (p = 0.006). The median HPIV-3 loads were high in upper respiratory tract secretions regardless of the presence or absence of LRTI-D (8.3 log10 vs. 7.6 log10 TCID50 /106 cells). HPIV-3 loads in respiratory tract samples in HM were not significantly higher than those found in HSCT but significantly higher than in non-hematological patients (p = 0.007). In conclusion, LRTI-D was frequent in HM patients who were diagnosed with HPIV-3 infection.


Assuntos
Neoplasias Hematológicas/complicações , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Paramyxoviridae/virologia , Infecções Respiratórias/virologia , Adolescente , Adulto , Idoso , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Transplante Homólogo/efeitos adversos , Adulto Jovem
3.
Vet Microbiol ; 259: 109129, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34087675

RESUMO

Caprine parainfluenza virus type 3 (CPIV3) is one of the most important viral respiratory pathogens of goat. Accumulating evidence demonstrates that apoptosis is a cellular mechanism for the host response to pathogens, and it participates in regulating viral replication. However, there is little study on CPIV3-induced host cells apoptosis. In this study, primary goat tracheal epithelial (GTE) cells were established as a cellular model that is permissive to CPIV3 infection. Then, we showed that CPIV3 infection induced apoptosis in GTE cells, as determined by morphological changes, flow cytometry and TUNEL assay. Moreover, Caspase activity and the expression of pro-apoptotic genes further suggested that CPIV3 induced apoptosis by activating both the intrinsic and extrinsic pathways. Mechanistically, the ability of CPIV3 to induce apoptosis was activated by N protein, and the viral protein increased CPIV3 replication through effecting apoptosis. Overall, our findings showed that GTE cells that will enable further analysis of CPIV3 infection and offers novel insights into the mechanisms of CPIV3-induced apoptosis in host cells.


Assuntos
Apoptose/genética , Proteínas do Nucleocapsídeo/genética , Vírus da Parainfluenza 3 Humana/química , Vírus da Parainfluenza 3 Humana/genética , Infecções por Respirovirus/genética , Infecções por Respirovirus/veterinária , Replicação Viral/genética , Animais , Células Cultivadas , Células Epiteliais/virologia , Doenças das Cabras/virologia , Cabras/virologia , Proteínas do Nucleocapsídeo/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/virologia , Traqueia/citologia
4.
MAbs ; 13(1): 1912884, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33876699

RESUMO

Human parainfluenza virus type III (HPIV3) is a common respiratory pathogen that afflicts children and can be fatal in vulnerable populations, including the immunocompromised. There are currently no effective vaccines or therapeutics available, resulting in tens of thousands of hospitalizations per year. In an effort to discover a protective antibody against HPIV3, we screened the B cell repertoires from peripheral blood, tonsils, and spleen from healthy children and adults. These analyses yielded five monoclonal antibodies that potently neutralized HPIV3 in vitro. These HPIV3-neutralizing antibodies targeted two non-overlapping epitopes of the HPIV3 F protein, with most targeting the apex. Prophylactic administration of one of these antibodies, PI3-E12, resulted in potent protection against HPIV3 infection in cotton rats. Additionally, PI3-E12 could also be used therapeutically to suppress HPIV3 in immunocompromised animals. These results demonstrate the potential clinical utility of PI3-E12 for the prevention or treatment of HPIV3 in both immunocompetent and immunocompromised individuals.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Neutralizantes/farmacologia , Antivirais/farmacologia , Pulmão/virologia , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Infecções por Respirovirus/prevenção & controle , Proteínas Virais de Fusão/antagonistas & inibidores , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Especificidade de Anticorpos , Antivirais/imunologia , Linfócitos B/imunologia , Linfócitos B/virologia , Linhagem Celular , Modelos Animais de Doenças , Epitopos , Interações Hospedeiro-Patógeno , Humanos , Hospedeiro Imunocomprometido , Pulmão/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/virologia , Sigmodontinae , Proteínas Virais de Fusão/imunologia
5.
Front Immunol ; 11: 575977, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33123159

RESUMO

Human Parainfluenza Virus-3 (HPIV3) causes severe respiratory illness in immunocompromised patients and lacks approved anti-viral therapies. A phase I study of adoptively transferred virus-specific T-cells (VSTs) targeting HPIV3 following bone marrow transplantation is underway (NCT03180216). We sought to identify immunodominant epitopes within HPIV3 Matrix protein and their cross-reactivity against related viral proteins. VSTs were generated from peripheral blood of healthy donors by ex-vivo expansion after stimulation with a 15-mer peptide library encompassing HPIV3 matrix protein. Epitope mapping was performed using IFN-γ ELIspot with combinatorial peptide pools. Flow cytometry was used to characterize products with intracellular cytokine staining. In 10 VST products tested, we discovered 12 novel immunodominant epitopes. All products recognized an epitope at the C-terminus. On IFN-γ ELISpot, individual peptides eliciting activity demonstrated mean IFN-γ spot forming units per well (SFU)/1x105 cells of 115.5 (range 24.5-247.5). VST products were polyfunctional, releasing IFN-γ and TNF-α in response to identified epitopes, which were primarily HLA Class II restricted. Peptides from Human Parainfluenza Virus-1 corresponding to the HPIV3 epitopes showed cross-reactivity for HPIV1 in 11 of 12 tested epitopes (mean cross reactivity index: 1.19). Characterization of HPIV3 epitopes may enable development of third-party VSTs to treat immune suppressed patients with HPIV infection.


Assuntos
Transferência Adotiva , Epitopos Imunodominantes , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/terapia , Linfócitos T/transplante , Proteínas da Matriz Viral/imunologia , Células Cultivadas , Ensaios Clínicos Fase I como Assunto , Reações Cruzadas , ELISPOT , Mapeamento de Epitopos , Interações Hospedeiro-Patógeno , Humanos , Interferon gama/metabolismo , Testes de Liberação de Interferon-gama , Vírus da Parainfluenza 1 Humana/patogenicidade , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/metabolismo , Infecções por Respirovirus/virologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
Am J Transplant ; 17(3): 809-812, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27888562

RESUMO

Parainfluenza virus (PIV) can cause serious infections after hematopoietic stem cell or lung transplantation. Limited data exist about PIV infections after kidney transplantation. We describe an outbreak of PIV-3 in a transplant unit. During the outbreak, 45 patients were treated on the ward for postoperative care after kidney or simultaneous pancreas-kidney (SPK) transplantation. Overall, 29 patients were tested for respiratory viruses (12 patients with respiratory symptoms, 17 asymptomatic exposed patients) from nasopharyngeal swabs using polymerase chain reaction. PIV-3 infection was confirmed in 12 patients. One patient remained asymptomatic. In others, symptoms were mostly mild upper respiratory tract symptoms and subsided within a few days with symptomatic treatment. Two patients suffered from lower respiratory tract symptoms (dyspnea, hypoxemia, pulmonary infiltrates in chest computed tomography) and required supplemental oxygen. Four of six SPK patients and eight of 39 of kidney transplant patients were infected with PIV (p = 0.04). In patients with follow-up tests, PIV-3 shedding was still detected 11-16 days after diagnosis. Despite rapid isolation of symptomatic patients, PIV-3 findings were diagnosed within 24 days, and the outbreak ceased only after closing the transplant ward temporarily. In conclusion, PIV-3 infections early after kidney or SPK transplantation were mostly mild. PIV-3 easily infected immunosuppressed transplant recipients, with prolonged viral shedding.


Assuntos
Rejeição de Enxerto/etiologia , Transplante de Rim/efeitos adversos , Transplante de Pâncreas/efeitos adversos , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Paramyxoviridae/complicações , Infecções Respiratórias/complicações , Feminino , Rejeição de Enxerto/epidemiologia , Sobrevivência de Enxerto , Humanos , Masculino , Pessoa de Meia-Idade , Infecções por Paramyxoviridae/epidemiologia , Infecções por Paramyxoviridae/virologia , Complicações Pós-Operatórias , Prognóstico , RNA Viral/genética , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/virologia , Fatores de Risco
7.
Vaccine ; 31(37): 3888-93, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23831325

RESUMO

We developed a novel vaccine platform based on a paramyxoviral, genome replication-deficient Sendai virus vector that can express heterologous genes inserted into the genome. To validate the novel approach in vivo, we generated a combined vaccine candidate against human respiratory syncytial virus (RSV) and human parainfluenza virus type 3 (PIV3). The present study compares two different methods of displaying heterologous antigens: (i) the RSV fusion (F) protein, encoded as a secretable version in an additional transcription unit, serves as an antigen only after being expressed in infected cells; (ii) PIV3 fusion (F) and hemagglutinin-neuraminidase (HN) genes, replacing Sendai counterparts in the vector genome, are also expressed as structural components on the surface of vaccine particles. The efficacy of this prototype vaccine was assessed in a mouse model after mucosal administration. The vaccine candidate was able to elicit specific mucosal, humoral and T cell-mediated immune responses against RSV and PIV3. However, PIV3 antigen display on the vaccine particles' surface induced higher antibody titers than the RSV antigen, being expressed only after cell infection. Consequently, this construct induced an adequate neutralizing antibody response only to PIV3. Finally, replicating virus particles were not detected in the lungs of immunized mice, confirming the genome stability and replication deficiency of this vaccine vector in vivo. Both factors can contribute substantially to the safety profile of vaccine candidates. In conclusion, this replication-deficient Sendai vector represents an efficient platform that can be used for vaccine developments against various viral pathogens.


Assuntos
Vírus Sendai/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Animais , Anticorpos Neutralizantes/sangue , Feminino , Vetores Genéticos , Genoma Viral , Proteína HN/genética , Imunidade Humoral , Camundongos , Camundongos Endogâmicos BALB C , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Vacinas contra Vírus Sincicial Respiratório/genética , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/patogenicidade , Linfócitos T/imunologia , Vacinas Sintéticas/farmacologia , Proteínas Virais de Fusão/genética , Replicação Viral
8.
Virology ; 433(2): 320-8, 2012 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-22959894

RESUMO

Human parainfluenza viruses (PIVs) cause acute respiratory illness in children, the elderly, and immunocompromised patients. PIV3 is a common cause of bronchiolitis and pneumonia, whereas PIV1 and 2 are frequent causes of upper respiratory tract illness and croup. To assess how PIV1, 2, and 3 differ with regard to replication and induction of type I interferons, interleukin-6, and relevant chemokines, we infected primary human airway epithelium (HAE) cultures from the same tissue donors and examined replication kinetics and cytokine secretion. PIV1 replicated to high titer yet did not induce cytokine secretion until late in infection, while PIV2 replicated less efficiently but induced an early cytokine peak. PIV3 replicated to high titer but induced a slower rise in cytokine secretion. The T cell chemoattractants CXCL10 and CXCL11 were the most abundant chemokines induced. Differences in replication and cytokine secretion might explain some of the differences in PIV serotype-specific pathogenesis and epidemiology.


Assuntos
Brônquios/imunologia , Brônquios/virologia , Vírus da Parainfluenza 1 Humana/fisiologia , Vírus da Parainfluenza 2 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Traqueia/imunologia , Traqueia/virologia , Células Cultivadas , Quimiocinas/biossíntese , Citocinas/biossíntese , Citocinas/genética , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interleucina-6/biossíntese , Interleucina-6/genética , Cinética , Vírus da Parainfluenza 1 Humana/classificação , Vírus da Parainfluenza 1 Humana/imunologia , Vírus da Parainfluenza 1 Humana/patogenicidade , Vírus da Parainfluenza 2 Humana/classificação , Vírus da Parainfluenza 2 Humana/imunologia , Vírus da Parainfluenza 2 Humana/patogenicidade , Vírus da Parainfluenza 3 Humana/classificação , Vírus da Parainfluenza 3 Humana/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Polimorfismo de Nucleotídeo Único , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Sorotipagem , Especificidade da Espécie , Replicação Viral
9.
mBio ; 3(3)2012.
Artigo em Inglês | MEDLINE | ID: mdl-22669629

RESUMO

UNLABELLED: Paramyxoviruses, a family of RNA enveloped viruses that includes human parainfluenza virus type 3 (HPIV3), cause the majority of childhood croup, bronchiolitis, and pneumonia worldwide. Infection starts with host cell receptor binding and fusion of the viral envelope with the cell membrane at the cell surface. The fusion process requires interaction of the two viral surface glycoproteins, the hemagglutinin-neuraminidase (HN) and the fusion protein (F). We have previously shown that viruses with an HN/F pair that is highly fusogenic in monolayers of immortalized cells due to mutations in HN's secondary sialic acid binding site are growth impaired in differentiated human airway epithelium (HAE) cultures and in vivo. Here we have shown that adaptation of HPIV3 to growth in the lung is determined by specific features of HN and F that are different from those required for growth in cultured immortalized cells. An HPIV3 virus bearing a mutated HN (H552Q), which is fit and fusogenic in immortalized cells but unfit for growth in the lung, evolved into a less-fusogenic but viable virus in differentiated human airway epithelium. Stepwise evolution led to a progressive decrease in efficiency of fusion activation by the HN/F pair, with a mutation in F first decreasing the activation of F by HN and a mutation in HN's secondary sialic acid binding site decreasing fusion activation further and producing a stable virus. Adaptation of HPIV3 to successful growth in HAE is determined by specific features of HN and F that lead to a less easily activated fusion mechanism. IMPORTANCE: Human parainfluenza viruses (HPIVs) are paramyxoviruses that cause the majority of childhood cases of croup, bronchiolitis, and pneumonia worldwide, but there are currently no vaccines or antivirals available for treatment. Enveloped viruses must fuse their membrane with the target cell membrane in order to initiate infection. Parainfluenza virus fusion proceeds via a multistep reaction orchestrated by the two glycoproteins that make up its fusion machine. The receptor-binding hemagglutinin-neuraminidase (HN), upon receptor engagement, activates the fusion protein (F) to penetrate the target cell and mediate viral entry. In this study, we show that the precise balance of fusion activation properties of these two glycoproteins during entry is key for infection. In clinically relevant tissues, viruses evolve to acquire a set of fusion features that provide key clues about requirements for infection in human beings.


Assuntos
Adaptação Biológica , Células Epiteliais/virologia , Proteína HN/genética , Vírus da Parainfluenza 3 Humana/fisiologia , Proteínas Virais de Fusão/genética , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Evolução Molecular , Humanos , Modelos Moleculares , Proteínas Mutantes/genética , Mutação de Sentido Incorreto , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/patogenicidade , Conformação Proteica
10.
Acta Derm Venereol ; 90(6): 625-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21057748

RESUMO

Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are severe adverse cutaneous reactions to drugs. We report here the first case of severe pneumonia caused by an unusual combined infection with Pneumocystis carinii (jiroveci), parainfluenza virus type 3, cytomegalovirus and Aspergillus fumigatus in a 63-year-old female patient with allopurinol-induced SJS/TEN overlap syndrome. Following treatment with high-dose systemic corticosteroids and intravenous immunoglobulin for SJS/TEN, her mucocutaneous lesions improved and she was due to be discharged. However, 15 days after cessation of corticosteroids, she developed pneumonia. Broncho-alveolar lavage revealed that the cause of infection was Pneumocystis carinii (jiroveci), parainfluenza virus type 3, cytomegalovirus and Aspergillus. These findings indicate that patients with SJS/TEN, particularly those treated with systemic corticosteroids, may be susceptible to infection with combinations of pathological agents resulting from damage to the bronchial epithelia.


Assuntos
Aspergillus fumigatus/patogenicidade , Infecções por Citomegalovirus/complicações , Doenças Pulmonares Intersticiais/etiologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Pneumocystis carinii/patogenicidade , Pneumonia por Pneumocystis/microbiologia , Aspergilose Pulmonar/microbiologia , Infecções por Respirovirus/virologia , Síndrome de Stevens-Johnson/complicações , Corticosteroides/uso terapêutico , Alopurinol/efeitos adversos , Anti-Infecciosos/uso terapêutico , Quimioterapia Combinada , Feminino , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Doenças Pulmonares Intersticiais/diagnóstico por imagem , Doenças Pulmonares Intersticiais/microbiologia , Doenças Pulmonares Intersticiais/terapia , Doenças Pulmonares Intersticiais/virologia , Pessoa de Meia-Idade , Radiografia , Respiração Artificial , Índice de Gravidade de Doença , Síndrome de Stevens-Johnson/induzido quimicamente , Síndrome de Stevens-Johnson/tratamento farmacológico , Síndrome de Stevens-Johnson/etiologia , Síndrome de Stevens-Johnson/patologia , Resultado do Tratamento
11.
J Virol ; 83(13): 6900-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19386708

RESUMO

Three discrete activities of the paramyxovirus hemagglutinin-neuraminidase (HN) protein, receptor binding, receptor cleaving (neuraminidase), and triggering of the fusion protein, each affect the promotion of viral fusion and entry. For human parainfluenza virus type 3 (HPIV3), the effects of specific mutations that alter these functions of the receptor-binding protein have been well characterized using cultured monolayer cells, which have identified steps that are potentially relevant to pathogenesis. In the present study, proposed mechanisms that are relevant to pathogenesis were tested in natural host cell cultures, a model of the human airway epithelium (HAE) in which primary HAE cells are cultured at an air-liquid interface and retain functional properties. Infection of HAE cells with wild-type HPIV3 and variant viruses closely reflects that seen in an animal model, the cotton rat, suggesting that HAE cells provide an ideal system for assessing the interplay of host cell and viral factors in pathogenesis and for screening for inhibitory molecules that would be effective in vivo. Both HN's receptor avidity and the function and timing of F activation by HN require a critical balance for the establishment of ongoing infection in the HAE, and these HN functions independently modulate the production of active virions. Alterations in HN's F-triggering function lead to the release of noninfectious viral particles and a failure of the virus to spread. The finding that the dysregulation of F triggering prohibits successful infection in HAE cells suggests that antiviral strategies targeted to HN's F-triggering activity may have promise in vivo.


Assuntos
Proteína HN/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Paramyxoviridae/virologia , Proteínas Virais de Fusão/metabolismo , Animais , Linhagem Celular , Feminino , Regulação Viral da Expressão Gênica , Humanos , Pulmão/patologia , Pulmão/virologia , Ratos , Ratos Endogâmicos , Receptores Virais/metabolismo
12.
J Clin Invest ; 115(7): 1688-98, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16007245

RESUMO

Human parainfluenza viruses cause several serious respiratory diseases in children for which there is no effective prevention or therapy. Parainfluenza viruses initiate infection by binding to cell surface receptors and then, via coordinated action of the 2 viral surface glycoproteins, fuse directly with the cell membrane to release the viral replication machinery into the host cell's cytoplasm. During this process, the receptor-binding molecule must trigger the viral fusion protein to mediate fusion and entry of the virus into a cell. This review explores the binding and entry into cells of parainfluenza virus type 3, focusing on how the receptor-binding molecule triggers the fusion process. There are several steps during the process of binding, triggering, and fusion that are now understood at the molecular level, and each of these steps represents potential targets for interrupting infection.


Assuntos
Vírus da Parainfluenza 3 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/terapia , Infecções por Respirovirus/virologia , Antivirais/farmacologia , Sítios de Ligação , Criança , Proteína HN/química , Proteína HN/fisiologia , Humanos , Influenza Humana/etiologia , Influenza Humana/terapia , Influenza Humana/virologia , Fusão de Membrana/efeitos dos fármacos , Fusão de Membrana/fisiologia , Modelos Biológicos , Modelos Moleculares , Neuraminidase/antagonistas & inibidores , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Receptores Virais/fisiologia , Infecções por Respirovirus/etiologia , Proteínas Virais de Fusão/fisiologia , Proteínas Virais/fisiologia , Virulência
13.
J Gen Virol ; 86(Pt 3): 733-741, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15722534

RESUMO

Routine antibody surveillance for Sendai virus in a breeding colony suggested viral invasion into laboratory rats. A more specific haemagglutination-inhibition test implied that the agent was related closely to Human parainfluenza virus type 3 (hPIV3), rather than Sendai virus. To isolate this virus, Vero cells were inoculated with lung homogenates of 30 young animals from the colony. One of the cultures became positive at the second passage by RT-PCR directed to the hPIV3 NP and L genes. Cytopathic effect with cell fusion was observed at the third passage. The HN gene of this virus (KK24) had >93 % similarity to those of other hPIV3 isolates, suggesting a human origin of KK24. Experimental intranasal inoculation of KK24 into SD rats showed virus replication in the lungs at 3-5 days post-infection (p.i.). Pathological examination of the lungs at day 5 p.i. indicated a moderate detachment, degradation and apoptosis of bronchial epitheliocytes with peribronchial mononuclear infiltrations. At day 7 p.i., these changes became less prominent, and no lesions were apparent at day 10 p.i. or later. The infected rats seroconverted at day 7 p.i. On the contrary, none of the 30 experimentally infected ICR mice showed any pathological lesions in their lungs, despite seroconversion at 7 days p.i. These results suggest that hPIV3 can invade rat colonies and has a moderate and transient pathogenicity in rats. This is the first report of non-experimental hPIV3 infection in laboratory rats, unexpectedly detected by antibody screening for Sendai virus.


Assuntos
Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/veterinária , Animais , Chlorocebus aethiops , Células Epiteliais/virologia , Epitélio/microbiologia , Humanos , Pulmão/virologia , Camundongos , Dados de Sequência Molecular , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/imunologia , Filogenia , Ratos , Infecções por Respirovirus/mortalidade , Infecções por Respirovirus/patologia , Células Vero
14.
J Virol ; 78(24): 13727-42, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15564482

RESUMO

Hydrophobic fusion peptides (FPs) are the most highly conserved regions of class I viral fusion-mediating glycoproteins (vFGPs). FPs often contain conserved glycine residues thought to be critical for forming structures that destabilize target membranes. Unexpectedly, a mutation of glycine residues in the FP of the fusion (F) protein from the paramyxovirus simian parainfluenza virus 5 (SV5) resulted in mutant F proteins with hyperactive fusion phenotypes (C. M. Horvath and R. A. Lamb, J. Virol. 66:2443-2455, 1992). Here, we constructed G3A and G7A mutations into the F proteins of SV5 (W3A and WR isolates), Newcastle disease virus (NDV), and human parainfluenza virus type 3 (HPIV3). All of the mutant F proteins, except NDV G7A, caused increased cell-cell fusion despite having slight to moderate reductions in cell surface expression compared to those of wild-type F proteins. The G3A and G7A mutations cause SV5 WR F, but not NDV F or HPIV3 F, to be triggered to cause fusion in the absence of coexpression of its homotypic receptor-binding protein hemagglutinin-neuraminidase (HN), suggesting that NDV and HPIV3 F have stricter requirements for homotypic HN for fusion activation. Dye transfer assays show that the G3A and G7A mutations decrease the energy required to activate F at a step in the fusion cascade preceding prehairpin intermediate formation and hemifusion. Conserved glycine residues in the FP of paramyxovirus F appear to have a primary role in regulating the activation of the metastable native form of F. Glycine residues in the FPs of other class I vFGPs may also regulate fusion activation.


Assuntos
Sequência Conservada , Regulação Viral da Expressão Gênica , Fusão de Membrana , Paramyxovirinae/patogenicidade , Peptídeos/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Fusão Celular , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Células Gigantes , Glicina , Humanos , Dados de Sequência Molecular , Mutação , Vírus da Doença de Newcastle/patogenicidade , Vírus da Parainfluenza 3 Humana/patogenicidade , Peptídeos/metabolismo , Rubulavirus/patogenicidade , Células Vero , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/genética
15.
J Clin Microbiol ; 42(10): 4780-7, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15472341

RESUMO

Parainfluenza viruses are a common cause of seasonal respiratory disease, but in high-risk individuals (e.g., young children) these viruses can cause severe clinical manifestations that require hospitalization. Beta-defensins are a subclass of antimicrobial peptides with antiviral activity. Use of adenovirus-mediated beta-defensin gene expression has been proposed as therapy for chronic bacterial infections commonly seen in cystic fibrosis patients; however, its use during parainfluenza virus 3 (PIV3) infection has not been evaluated. The hypothesis in this experiment was that adenovirus expression of human beta-defensin 6 (HBD6) would diminish concurrent PIV3 infection in neonatal lambs. The group infected with adenovirus HBD6 and PIV3 had increased levels of pulmonary neutrophil recruitment compared to those for the group infected with PIV3 or PIV3 and adenovirus, with an increased respiration rate and body temperature late in the course of the PIV3-adenovirus HBD6 infection. Interestingly, the adenovirus-treated groups had higher levels of immunohistochemical staining for PIV3 and syncytial cell formation than the group infected with PIV3, suggesting that treatment with the adenovirus vector, regardless of whether it was carrying a target gene, exacerbated the PIV3 infection. The levels of expression of mRNA for antimicrobial surfactant proteins A and D and sheep beta-defensin 1 were increased by PIV3 and adenovirus treatment, and the increased levels of expression roughly corresponded to the degree of inflammation. While pulmonary administration of a high-dose adenovirus vector has been associated with undesirable inflammation, this is the first study to show that it can exacerbate concurrent viral infection, a concern that needs to be addressed for future studies of adenovirus in the lung. Additionally, this study showed that adenovirus-mediated HBD6 expression increases neutrophil recruitment, a recently described attribute of beta-defensins, with mild accentuation of PIV3 activity and inflammation.


Assuntos
Adenovírus Humanos/genética , Terapia Genética/efeitos adversos , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/fisiopatologia , beta-Defensinas/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Vetores Genéticos/administração & dosagem , Humanos , Inflamação , Infecções por Respirovirus/terapia , Infecções por Respirovirus/virologia , Ovinos , beta-Defensinas/genética
16.
J Virol ; 77(6): 3647-54, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12610140

RESUMO

For human parainfluenza virus type 3 and many other paramyxoviruses, membrane fusion mediated by the fusion protein (F) has a stringent requirement for the presence of the homotypic hemagglutinin-neuraminidase protein (HN). With the goal of gaining further insight into the role of HN in the fusion process, we developed a simple method for quantitative comparison of the ability of wild-type and variant HNs to activate F. In this method, HN/F-coexpressing cells with red blood cells (RBC) bound to them at 4 degrees C are transferred to 22 degrees C, and at different times after transfer 4-guanidino-neu5Ac2en (4-GU-DANA) is added; this inhibitor of the HN-receptor interaction then releases all reversibly bound RBC but not those in which F insertion in the target membrane or fusion has occurred. Thus, the amount of irreversibly bound (nonreleased) RBC provides a measure of F activation, and the use of fluorescently labeled RBC permits microscopic assessment of the extent to which F insertion has progressed to fusion. We studied two neuraminidase-deficient HN variants, C28a, which has two mutations, P111S and D216N, and C28, which possesses the D216N mutation only. C28a but not C28 exhibits a slow fusion phenotype, although determination of the HNs' receptor-binding avidity (with our sensitive method, employing RBC with different degrees of receptor depletion) showed that the receptor-binding avidity of C28a or C28 HN was not lower than that of the wild type. The F activation assay, however, revealed fusion-triggering defects in C28a HN. After 10 and also 20 min at 22 degrees C, irreversible RBC binding was significantly less for cells coexpressing wild-type F with C28a HN than for cells coexpressing wild-type F with wild-type HN. In addition, F insertion progressed to fusion more slowly in the case of C28a HN-expressing cells than of wild-type HN-expressing cells. Identical defects were found for P111S HN, whereas for C28 HN, representing the 216 mutation of C28a, F activation and fusion were as rapid as for wild-type HN. The diminished fusion promotion capacity of C28a HN is therefore attributable to P111S, a mutation in the stalk region of the molecule that causes no decrease in receptor-binding avidity. C28a HN is the first parainfluenza virus variant found so far to be specifically defective in HN's F-triggering and fusion promotion functions and may contribute to our understanding of transmission of the activating signal from HN to F.


Assuntos
Regulação Viral da Expressão Gênica , Proteína HN/metabolismo , Fusão de Membrana , Vírus da Parainfluenza 3 Humana/patogenicidade , Transdução de Sinais , Proteínas Virais de Fusão/metabolismo , Animais , Linhagem Celular , Eritrócitos , Proteína HN/genética , Humanos , Vírus da Parainfluenza 3 Humana/genética , Proteínas Virais de Fusão/genética
18.
Comp Med ; 52(6): 548-54, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12540169

RESUMO

A novel Respirovirus was isolated from nasopharyngeal swab specimens from clinically normal laboratory guinea pigs, and was characterized and named caviid parainfluenza virus 3 (CavPIV-3). The CavPIV-3 is enveloped, is 100 to 300 nm in diameter, and has a characteristic 15-nm-diameter chevron-shaped virus ribonucleocapsid protein. Sequence analysis of the fusion glycoprotein of CavPIV-3 revealed it to be 94% identical to human and guinea pig parainfluenza 3 (PIV-3) viruses and 80% identical to bovine PIV-3. To determine whether CavPIV-3 causes clinical disease in laboratory guinea pigs and to compare the serologic response of guinea pigs to CavPIV-3 and to other paramyxoviruses, an infection study was performed, in which groups of guinea pigs were inoculated with CavPIV-3, Sendai virus, simian virus 5 (SV-5), murine pneumonia virus (PVM), or bovine PIV-3 virus. During the course of the study, guinea pigs were maintained in an infectious disease suite, housed in Micro-Isolator cages, and were only manipulated under a laminar flow hood. Clinical signs of disease were not observed in any of the paramyxovirus-inoculated guinea pigs during the eight-week course of the study, and histologic signs of disease were not evident at necropsy eight weeks after inoculation. Guinea pigs inoculated with CavPIV-3, Sendai virus, PVM, and bovine PIV-3 developed robust homologous or heterologous serologic responses. In contrast, guinea pigs inoculated with SV-5 developed modest or equivocal serologic responses, as assessed by use of an enzyme-linked immunosorbent assay. Further, use of the SV-5 enzyme-linked immunosorbent assay resulted in the highest degree of non-specific reactivity among all of the paramyxovirus assays. In summary, CavPIV-3 is a novel guinea pig Respirovirus that subclinically infects laboratory guinea pigs, resulting in a robust serologic response, but no observed clinical or histologic disease. The CavPIV-3 fusion glycoprotein gene sequence is available from GenBank as accession No. AF394241, and the CavPIV-3 virus is available from the American Type Culture Collection as accession No. DR-1547.


Assuntos
Vírus da Parainfluenza 3 Humana/isolamento & purificação , Infecções por Respirovirus/veterinária , Animais , Anticorpos Antivirais/análise , Sequência de Bases , Chlorocebus aethiops , Primers do DNA/química , Eletroforese em Gel de Ágar/veterinária , Ensaio de Imunoadsorção Enzimática/veterinária , Cobaias , Dados de Sequência Molecular , Vírus da Parainfluenza 3 Humana/patogenicidade , Vírus da Parainfluenza 3 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/ultraestrutura , RNA Viral/análise , Infecções por Respirovirus/fisiopatologia , Infecções por Respirovirus/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Análise de Sequência de DNA/veterinária , Células Vero/virologia , Proteínas Virais de Fusão/análise
19.
J Virol ; 75(4): 1984-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160698

RESUMO

Human parainfluenza virus type 3 (HPIV-3) is an airborne pathogen that infects the epithelial cells of the respiratory tract. In the present study we investigated the interaction of HPIV-3 with the type II alveolar human lung polarized epithelial A549 cells. Although HPIV-3 entry and budding were bidirectional from both the apical and the basolateral domains, HPIV-3 exhibited preferential entry and release from the apical pole. While disruption of the cellular actin microfilament and microtubule by cytochalasin D and nocodazole, respectively, had no effect on virus entry, disruption of the microtubule but not the microfilament inhibited HPIV-3 release.


Assuntos
Citoesqueleto de Actina/fisiologia , Células Epiteliais/virologia , Microtúbulos/fisiologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Alvéolos Pulmonares/citologia , Polaridade Celular , Humanos , Vírus da Parainfluenza 3 Humana/fisiologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA