Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
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.
Microb Pathog ; 134: 103569, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31163247

RESUMO

Caprine parainfluenza virus type 3 (CPIV3) is one of the important viral respiratory tract agents in goats. The pathogenicity of CPIV3 has been examined in goats but it has not been explored in other laboratory animals. In the present study, an experimental infection of guinea pigs with CPIV3 was performed. The virus-inoculated guinea pigs displayed clinical signs related to the respiratory disease at 2-12 days post inoculation (dpi). Five infected guinea pigs died during 2 and 7 dpi. Apparent gross pneumonic lesions including consolidation and congestion in one or more lung lobes were observed in necropsied and dead animals. Histo-pathological changes in lungs including expansions of the alveolar interstitium, congestion, macrophage infiltration and compensatory emphysema were also observed. Virus was detectable at 2-10 dpi, 2-10 dpi and 2-7 dpi, as detected by virus isolation, real-time RT-PCR and immunohistochemistry staining, respectively. Viremia was also confirmed after CPIV3 infection during 3-7 dpi. The severe pathological lesions and highest viral load were observed before 7 dpi. Viral specific hemagglutination inhibition and neutralizing antibodies were produced from 7 dpi and 10 dpi, respectively, which related to the clearance of virus. The results present here indicated that guinea pig could be an ideal laboratory animal model for CPIV3 studies in the future.


Assuntos
Doenças das Cabras/virologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/veterinária , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Modelos Animais de Doenças , Feminino , Cabras , Cobaias , Pulmão/patologia , Pulmão/virologia , Reação em Cadeia da Polimerase em Tempo Real , Infecções por Respirovirus/imunologia , Carga Viral , Viremia , Virulência
4.
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
5.
PLoS Pathog ; 5(9): e1000587, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19806178

RESUMO

A number of paramyxoviruses are responsible for acute respiratory infections in children, elderly and immuno-compromised individuals, resulting in airway inflammation and exacerbation of chronic diseases like asthma. To understand the molecular pathogenesis of these infections, we searched for cellular targets of the virulence protein C of human parainfluenza virus type 3 (hPIV3-C). We found that hPIV3-C interacts directly through its C-terminal domain with STAT1 and GRB2, whereas C proteins from measles or Nipah viruses failed to do so. Binding to STAT1 explains the previously reported capacity of hPIV3-C to block type I interferon signaling, but the interaction with GRB2 was unexpected. This adaptor protein bridges Epidermal Growth Factor (EGF) receptor to MAPK/ERK pathway, a signaling cascade recently found to be involved in airway inflammatory response. We report that either hPIV3 infection or transient expression of hPIV3-C both increase cellular response to EGF, as assessed by Elk1 transactivation and phosphorylation levels of ERK1/2, 40S ribosomal subunit protein S6 and translation initiation factor 4E (eIF4E). Furthermore, inhibition of MAPK/ERK pathway with U0126 prevented viral protein expression in infected cells. Altogether, our data provide molecular basis to explain the role of hPIV3-C as a virulence factor and determinant of pathogenesis and demonstrate that Paramyxoviridae have evolved a single virulence factor to block type I interferon signaling and to boost simultaneous cellular response to growth factors.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Interferon Tipo I/metabolismo , Vírus da Parainfluenza 3 Humana/metabolismo , Fatores de Virulência/metabolismo , Animais , Sítios de Ligação , Contagem de Células , Linhagem Celular , Chlorocebus aethiops , Fator de Iniciação 4E em Eucariotos/metabolismo , Citometria de Fluxo , Proteína Adaptadora GRB2/metabolismo , Células HeLa , Humanos , Imuno-Histoquímica , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , Fosforilação , Mapeamento de Interação de Proteínas , Reprodutibilidade dos Testes , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Células Vero , Proteínas Virais/metabolismo
6.
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
7.
Diagn Microbiol Infect Dis ; 99(3): 115244, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33253961

RESUMO

Nosocomial virus infections cause significant morbidity and mortality. Besides influenza viruses, the disease burden of parainfluenza virus type 3 (PIV-3) is comparatively high among hospitalized patients and severe disease courses can occur. PIV-3 showed the highest rates of nosocomial infections of a panel of respiratory viruses. Therefore, a retrospective observational study was conducted among patients with either PIV-3 or influenza viruses, which served as reference pathogen. The aim was to compare the seasonal dynamics and clinical characteristics of nosocomial infections with these highly transmittable viruses. Nosocomial infection occurred in 15.8% (n = 177) of all influenza cases, mainly in the first half of a season. About 24.3% (n = 104) of the PIV-3 cases were nosocomial and occurred mainly in the second half of a season. Both nosocomial rates of influenza and nosocomial rates of PIV-3 varied between the seasons. Community acquired and nosocomial cases differed in underlying medical conditions and immunosuppression. Knowledge of the baseline rates of nosocomial infections could contribute to the implementation of appropriate infection control measures.


Assuntos
Infecção Hospitalar/epidemiologia , Infecção Hospitalar/virologia , Hospitais Universitários/estatística & dados numéricos , Influenza Humana/epidemiologia , Infecções por Respirovirus/epidemiologia , Adolescente , Adulto , Criança , Pré-Escolar , Infecção Hospitalar/transmissão , Feminino , Alemanha/epidemiologia , Humanos , Lactente , Influenza Humana/transmissão , Masculino , Pessoa de Meia-Idade , Orthomyxoviridae/patogenicidade , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções Respiratórias/virologia , Infecções por Respirovirus/transmissão , Estudos Retrospectivos , Estações do Ano , Adulto Jovem
8.
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
9.
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
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.
Zhonghua Jie He He Hu Xi Za Zhi ; 33(12): 907-11, 2010 Dec.
Artigo em Zh | MEDLINE | ID: mdl-21211410

RESUMO

OBJECTIVE: To establish a guinea pig model of cough induced by human parainfluenza virus type 3 (PIV3) infection, and to investigate the change of the cough reflex sensitivity (CRS). METHODS: Sixty male SPF guinea pigs were divided into 6 groups (n=10, each), namely, a normal control group, an asthma group and 4 groups of PIV3 inoculation which included post-infection day (PID) 6, 12, 28, and 42. Infected animals were inoculated by intranasal instillation of PIV3 suspension. Control animals were inoculated by uninfected cell culture medium. Asthma animals were sensitized and challenged by ovalbumin. The Buxco system was used to assess cough reflex sensitivity (CRS) elicited by capsaicin and airway hyper-reaction (AHR). Airway inflammation was studied by bronchoalveolar lavage (BAL) cytology and lung histopathology. RESULTS: The CRS of PID 6, 12, 28 and 42 groups was 7.50 (5.25), 7.30 (7.25), 8.40 (9.75) and 8.20 (5.50) Cough counts (CCnt). Compared with 2.50 (3.00) CCnt of the vehicle group, the CRS to capsaicin increased significantly in all the animals with PIV3 inoculation (P value were 0.024, 0.03, 0.011 and 0.008) and peaked in PID 42. There was no significant difference (P=0.18) between 3.90 (1.75) CCnt of the asthma animals and the normal control. Animals of PID 6 showed significantly greater AHR to 2 highest concentrations of methacholine than the normal controls. BAL total cell counts of both the PIV3-inoculated and the asthma animals were significantly higher than those of the normal control, with the number of lymphocytes increased significantly within first 2 weeks after PIV3 inoculation. The lung pathology of PIV3-inoculated animals showed airway inflammation without pneumonia in acute infectious phase. CONCLUSIONS: An animal model of cough induced by PIV3 was created. The CRS of infected guinea pigs increased significantly in both acute and subacute phases of cough. Elevation of CRS may be characteristic of cough caused by virus.


Assuntos
Tosse/virologia , Modelos Animais de Doenças , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Paramyxoviridae/fisiopatologia , Animais , Asma/virologia , Cobaias , Masculino
12.
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
13.
Antimicrob Agents Chemother ; 53(9): 3942-51, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19564364

RESUMO

Human parainfluenza virus type 3 (hPIV-3) is a major respiratory tract pathogen that affects young children, but no vaccines or antiviral drugs against it have yet been developed. We developed a mouse model to evaluate the efficacies of the novel parainfluenza virus hemagglutinin-neuraminidase (HN) inhibitors BCX 2798 and BCX 2855 against a recombinant Sendai virus (rSeV) in which the fusion (F) and HN surface glycoproteins (FHN) were replaced by those of hPIV-3 [rSeV(hPIV-3FHN)]. In the prophylaxis model, 129X1/SvJ mice were infected with a 90% or 20% lethal dose of the virus and were treated intranasally for 5 days with 10 mg/kg of body weight/day of either compound starting 4 h before infection. Prophylactic treatment of the mice with either compound did not prevent their death in a 90% lethality model of rSeV(hPIV-3FHN) infection. However, it significantly reduced the lung virus titers, the amount of weight lost, and the rate of mortality in mice infected with a 20% lethal virus dose. In the therapy model, mice were infected with a nonlethal dose of the virus (100 PFU/mouse) and were treated intranasally with 1 or 10 mg/kg/day of either compound for 5 days starting at 24 or 48 h postinfection. Treatment of the mice with either compound significantly reduced the virus titer in the lungs, subsequently causing a reduction in the number of immune cells and the levels of cytokines in the bronchoalveolar lavage fluid and histopathologic changes in the airways. Our results indicate that BCX 2798 and BCX 2855 are effective inhibitors of hPIV-3 HN in our mouse model and may be promising candidates for the prophylaxis and treatment of hPIV-3 infection in humans.


Assuntos
Antivirais/farmacologia , Azidas/farmacologia , Ácidos Hexurônicos/farmacologia , Vírus da Parainfluenza 3 Humana/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/patogenicidade , Vírus Sendai/efeitos dos fármacos , Sulfonamidas/farmacologia , Animais , Linhagem Celular , Cricetinae , Feminino , Haplorrinos , Humanos , Camundongos
14.
J Virol ; 82(18): 9299-302, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18614637

RESUMO

Human parainfluenza virus type 3 (HPIV3) is a major respiratory pathogen in humans. Failure to induce immunological memory associated with HPIV3 infection has been attributed to inhibition of lymphocyte proliferation. We demonstrate that the inability of mixed lymphocytes to respond to virally infected antigen-presenting cells is due to an interleukin-2-dependent, nonapoptotic mechanism involving natural killer (NK) cells and their influence is exerted in a contact-dependent manner. These results suggest a novel regulatory mechanism for NK cells during HPIV3 infection, offering an explanation for viral persistence and poor memory responses.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Linfócitos T/imunologia , Complexo CD3/metabolismo , Células Cultivadas , Técnicas de Cocultura , Humanos , Interleucina-2/metabolismo , Vírus da Parainfluenza 3 Humana/imunologia , Infecções por Respirovirus/virologia
15.
Cell Rep ; 29(8): 2229-2242.e4, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31747597

RESUMO

Many positive-strand RNA viruses remodel the endomembrane to form specialized replication organelles. However, knowledge regarding whether negative-strand RNA viruses take advantage of intracellular membranes for replication is limited. Here we show that a negative-strand RNA virus, human parainfluenza virus type 3 (HPIV3), remodels the endoplasmic reticulum (ER) membrane to form inclusion bodies (IBs), whereby the phosphoprotein (P) of HPIV3 recruits phosphatidylinositol 4-kinase beta (PI4KB) to IBs to generate PI4P, creating a PI4P-enriched microenvironment to promote HPIV3 replication. In addition, we find that human respiratory syncytial virus (HRSV) also takes advantage of the ER to form IBs and that these IBs are also enriched with PI4P. The nucleoprotein of HRSV recruits PI4KB to IBs. These results suggest that paramyxoviruses also exploit the host endomembrane to form IBs and that PI4KB is recruited by viral proteins to enrich IBs with PI4P to facilitate viral replication.


Assuntos
1-Fosfatidilinositol 4-Quinase/metabolismo , Retículo Endoplasmático/metabolismo , Corpos de Inclusão/metabolismo , Vírus da Parainfluenza 3 Humana/patogenicidade , 1-Fosfatidilinositol 4-Quinase/genética , Retículo Endoplasmático/genética , Interações Hospedeiro-Patógeno , Humanos , Corpos de Inclusão/genética , Membranas Intracelulares/metabolismo , Vírus da Parainfluenza 3 Humana/genética , Paramyxovirinae/genética , Paramyxovirinae/patogenicidade , Fosfoproteínas/metabolismo , Vírus de RNA/genética , Vírus de RNA/patogenicidade , Replicação Viral/genética , Replicação Viral/fisiologia
16.
mBio ; 10(3)2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31064833

RESUMO

Infectious viruses so precisely fit their hosts that the study of natural viral infection depends on host-specific mechanisms that affect viral infection. For human parainfluenza virus 3, a prevalent cause of lower respiratory tract disease in infants, circulating human viruses are genetically different from viruses grown in standard laboratory conditions; the surface glycoproteins that mediate host cell entry on circulating viruses are suited to the environment of the human lung and differ from those of viruses grown in cultured cells. Polarized human airway epithelium cultures have been used to represent the large, proximal airways of mature adult airways. Here we modeled respiratory virus infections that occur in children or infect the distal lung using lung organoids that represent the entire developing infant lung. These 3D lung organoids derived from human pluripotent stem cells contain mesoderm and pulmonary endoderm and develop into branching airway and alveolar structures. Whole-genome sequencing analysis of parainfluenza viruses replicating in the organoids showed maintenance of nucleotide identity, suggesting that no selective pressure is exerted on the virus in this tissue. Infection with parainfluenza virus led to viral shedding without morphological changes, while respiratory syncytial virus infection induced detachment and shedding of infected cells into the lung organoid lumens, reminiscent of parainfluenza and respiratory syncytial virus in human infant lungs. Measles virus infection, in contrast, induced syncytium formation. These human stem cell-derived lung organoids may serve as an authentic model for respiratory viral pathogenesis in the developing or infant lung, recapitulating respiratory viral infection in the host.IMPORTANCE Respiratory viruses are among the first pathogens encountered by young children, and the significant impact of these viral infections on the developing lung is poorly understood. Circulating viruses are suited to the environment of the human lung and are different from those of viruses grown in cultured cells. We modeled respiratory virus infections that occur in children or infect the distal lung using lung organoids that represent the entire developing infant lung. These 3D lung organoids, derived from human pluripotent stem cells, develop into branching airway and alveolar structures and provide a tissue environment that maintains the authentic viral genome. The lung organoids can be genetically engineered prior to differentiation, thereby generating tissues bearing or lacking specific features that may be relevant to viral infection, a feature that may have utility for the study of host-pathogen interaction for a range of lung pathogens.


Assuntos
Células Epiteliais Alveolares/virologia , Pulmão/virologia , Organoides/virologia , Vírus da Parainfluenza 3 Humana/patogenicidade , Células-Tronco Pluripotentes/virologia , Infecções por Respirovirus/patologia , Diferenciação Celular , Células Cultivadas , Genoma Viral , Humanos , Lactente , Pulmão/citologia , Pulmão/patologia , Vírus do Sarampo/patogenicidade , Vírus da Parainfluenza 3 Humana/genética , Vírus Sincicial Respiratório Humano/patogenicidade , Internalização do Vírus , Sequenciamento Completo do Genoma
17.
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
18.
Cell Host Microbe ; 21(4): 538-547.e4, 2017 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-28407488

RESUMO

Mitophagy is a form of autophagy that selectively removes damaged mitochondria. Impaired mitochondria can be tagged by the kinase PINK1, which triggers recruitment of the E3-ubiquitin ligase Parkin and subsequent mitochondrial sequestration within autophagosomes. We previously found that human parainfluenza virus type 3 (HPIV3) infection induces autophagy, but the type and mechanisms of autophagy induction remain unknown. Here, we show that matrix protein (M) of HPIV3 translocates to mitochondria and interacts with Tu translation elongation factor mitochondrial (TUFM). M-mediated mitophagy does not require the Parkin-PINK1 pathway but rather an interaction between M and the LC3 protein that mediates autophagosome formation. These interactions with both TUFM and LC3 are required for the induction of mitophagy and lead to inhibition of the type I interferon response. These results reveal that a viral protein is sufficient to induce mitophagy by bridging autophagosomes and mitochondria.


Assuntos
Interações Hospedeiro-Patógeno , Imunossupressores/metabolismo , Interferons/metabolismo , Mitofagia/efeitos dos fármacos , Vírus da Parainfluenza 3 Humana/patogenicidade , Fator Tu de Elongação de Peptídeos/metabolismo , Proteínas da Matriz Viral/metabolismo , Humanos
20.
Viral Immunol ; 18(3): 523-33, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16212531

RESUMO

Mechanisms of dendritic cells (DCs) immunomodulation by parainfluenza viruses have not been characterized. We analyzed whether the human parainfluenza 3 (HPF3) virus hemagglutinin-neuraminidase glycoprotein (HN) might influence DC maturation. HN possesses a receptor binding function and a neuraminidase or desialidating activity. To assess whether the neuraminidase activity of HN affects DC maturation, human myeloid DCs were exposed to either live or UV-inactivated HPF3 viruses containing wild type or a mutated form of HN with decreased neuraminidase activity. Exposure of human DCs to either UV-inactivated or live virus induced up-regulation of CD83 and CD86 surface markers, morphological changes, and a cytokine expression pattern consistent with maturation. However, the level of maturation was found to be lower in DCs infected with the neuraminidase deficient variant as compared to the wild type. These results suggest that during the course of viral infection, HN's neuraminidase activity may play an important role contributing to maturation and activation of DCs.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/patologia , Neuraminidase/imunologia , Vírus da Parainfluenza 3 Humana/enzimologia , Vírus da Parainfluenza 3 Humana/imunologia , Diferenciação Celular , Quimiocinas/biossíntese , Clostridium perfringens/enzimologia , Clostridium perfringens/imunologia , Citocinas/biossíntese , Genes Virais , Humanos , Técnicas In Vitro , Mutação , Neuraminidase/genética , Vírus da Parainfluenza 3 Humana/genética , Vírus da Parainfluenza 3 Humana/patogenicidade , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/patologia , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA