Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
PLoS Negl Trop Dis ; 14(3): e0008092, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32119657

RESUMO

In August 2012, a wildlife biologist became severely ill after becoming infected with a novel paramyxovirus, termed Sosuga virus. In the weeks prior to illness, the patient worked with multiple species of bats in South Sudan and Uganda, including Egyptian rousette bats (ERBs: Rousettus aegyptiacus). A follow-up study of Ugandan bats found multiple wild-caught ERBs to test positive for SOSV in liver and spleen. To determine the competency of these bats to act as a natural reservoir host for SOSV capable of infecting humans, captive-bred ERBs were inoculated with a recombinant SOSV, representative of the patient's virus sequence. The bats were inoculated subcutaneously, sampled daily (blood, urine, fecal, oral and rectal swabs) and serially euthanized at predetermined time points. All inoculated bats became infected with SOSV in multiple tissues and blood, urine, oral, rectal and fecal swabs tested positive for SOSV RNA. No evidence of overt morbidity or mortality were observed in infected ERBs, although histopathological examination showed subclinical disease in a subset of tissues. Importantly, SOSV was isolated from oral/rectal swabs, urine and feces, demonstrating shedding of infectious virus concomitant with systemic infection. All bats euthanized at 21 days post-inoculation (DPI) seroconverted to SOSV between 16 and 21 DPI. These results are consistent with ERBs being competent reservoir hosts for SOSV with spillover potential to humans.


Assuntos
Quirópteros/virologia , Reservatórios de Doenças/virologia , Transmissão de Doença Infecciosa , Infecções por Paramyxoviridae/transmissão , Paramyxoviridae/crescimento & desenvolvimento , Tropismo Viral , Animais , Humanos , Masculino , Uganda
2.
J Virol ; 92(20)2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-30068647

RESUMO

J paramyxovirus (JPV) was first isolated from moribund mice with hemorrhagic lung lesions in Australia in 1972. It is a paramyxovirus classified under the newly proposed genus Jeilongvirus JPV has a genome of 18,954 nucleotides, consisting of eight genes in the order 3'-N-P/V/C-M-F-SH-TM-G-L-5'. JPV causes little cytopathic effect (CPE) in tissue culture cells but severe disease in mice. The small hydrophobic (SH) protein is an integral membrane protein encoded by many paramyxoviruses, such as mumps virus (MuV) and respiratory syncytial virus (RSV). However, the function of SH has not been defined in a suitable animal model. In this work, the functions of SH of JPV, MuV, and RSV have been examined by generating recombinant JPV lacking the SH protein (rJPV-ΔSH) or replacing SH of JPV with MuV SH (rJPV-MuVSH) or RSV SH (rJPV-RSVSH). rJPV-ΔSH, rJPV-MuVSH, and rJPV-RSVSH were viable and had no growth defect in tissue culture cells. However, more tumor necrosis factor alpha (TNF-α) was produced during rJPV-ΔSH infection, confirming the role of SH in inhibiting TNF-α production. rJPV-ΔSH induced more apoptosis in tissue culture cells than rJPV, rJPV-MuVSH, and rJPV-RSVSH, suggesting that SH plays a role in blocking apoptosis. Furthermore, rJPV-ΔSH was attenuated in mice compared to rJPV, rJPV-MuVSH, and rJPV-RSVSH, indicating that the SH protein plays an essential role in virulence. The results indicate that the functions of MuV SH and RSV SH are similar to that of JPV SH even though they have no sequence homology.IMPORTANCE Paramyxoviruses are associated with many devastating diseases in animals and humans. J paramyxovirus (JPV) was isolated from moribund mice in Australia in 1972. Newly isolated viruses, such as Beilong virus (BeiPV) and Tailam virus (TlmPV), have genome structures similar to that of JPV. A new paramyxovirus genus, Jeilongvirus, which contains JPV, BeiPV, and TlmPV, has been proposed. Small hydrophobic (SH) protein is present in many paramyxoviruses. Our present study investigates the role of SH protein of JPV in pathogenesis in its natural host. Understanding the pathogenic mechanism of Jeilongvirus is important to control and prevent potential diseases that may emerge from this group of viruses.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Infecções por Paramyxoviridae/patologia , Paramyxoviridae/crescimento & desenvolvimento , Proteínas Oncogênicas de Retroviridae/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Chlorocebus aethiops , Modelos Animais de Doenças , Deleção de Genes , Teste de Complementação Genética , Humanos , Camundongos , Viabilidade Microbiana , Vírus da Caxumba/genética , Vírus da Caxumba/fisiologia , Infecções por Paramyxoviridae/virologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/fisiologia , Proteínas Oncogênicas de Retroviridae/genética , Virulência , Fatores de Virulência/genética
3.
Sci Rep ; 7: 43395, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28262699

RESUMO

GS-5734 is a monophosphate prodrug of an adenosine nucleoside analog that showed therapeutic efficacy in a non-human primate model of Ebola virus infection. It has been administered under compassionate use to two Ebola patients, both of whom survived, and is currently in Phase 2 clinical development for treatment of Ebola virus disease. Here we report the antiviral activities of GS-5734 and the parent nucleoside analog across multiple virus families, providing evidence to support new indications for this compound against human viruses of significant public health concern.


Assuntos
Alanina/análogos & derivados , Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Marburgvirus/efeitos dos fármacos , Paramyxoviridae/efeitos dos fármacos , Pneumovirinae/efeitos dos fármacos , Pró-Fármacos/farmacologia , Ribonucleotídeos/farmacologia , Células A549 , Monofosfato de Adenosina/análogos & derivados , Alanina/síntese química , Alanina/metabolismo , Alanina/farmacologia , Animais , Antivirais/síntese química , Antivirais/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Ebolavirus/enzimologia , Ebolavirus/crescimento & desenvolvimento , Expressão Gênica , Células HEK293 , Células HeLa , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Marburgvirus/enzimologia , Marburgvirus/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Nucleosídeos/síntese química , Nucleosídeos/metabolismo , Nucleosídeos/farmacologia , Paramyxoviridae/enzimologia , Paramyxoviridae/crescimento & desenvolvimento , Pneumovirinae/enzimologia , Pneumovirinae/crescimento & desenvolvimento , Pró-Fármacos/síntese química , Pró-Fármacos/metabolismo , RNA Polimerase Dependente de RNA/antagonistas & inibidores , RNA Polimerase Dependente de RNA/química , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Ribonucleotídeos/síntese química , Ribonucleotídeos/metabolismo , Células Vero , Proteínas Virais/antagonistas & inibidores , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
4.
J Virol ; 88(19): 11199-214, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25031353

RESUMO

Neurotropic alphaviruses, including western, eastern, and Venezuelan equine encephalitis viruses, cause serious and potentially fatal central nervous system infections in humans for which no currently approved therapies exist. We previously identified a series of thieno[3,2-b]pyrrole derivatives as novel inhibitors of neurotropic alphavirus replication, using a cell-based phenotypic assay (W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275), and subsequently developed second- and third-generation indole-2-carboxamide derivatives with improved potency, solubility, and metabolic stability (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). In this report, we describe the antiviral activity of the most promising third-generation lead compound, CCG205432, and closely related analogs CCG206381 and CCG209023. These compounds have half-maximal inhibitory concentrations of ∼1 µM and selectivity indices of >100 in cell-based assays using western equine encephalitis virus replicons. Furthermore, CCG205432 retains similar potency against fully infectious virus in cultured human neuronal cells. These compounds show broad inhibitory activity against a range of RNA viruses in culture, including members of the Togaviridae, Bunyaviridae, Picornaviridae, and Paramyxoviridae families. Although their exact molecular target remains unknown, mechanism-of-action studies reveal that these novel indole-based compounds target a host factor that modulates cap-dependent translation. Finally, we demonstrate that both CCG205432 and CCG209023 dampen clinical disease severity and enhance survival of mice given a lethal western equine encephalitis virus challenge. These studies demonstrate that indole-2-carboxamide compounds are viable candidates for continued preclinical development as inhibitors of neurotropic alphaviruses and, potentially, of other RNA viruses. IMPORTANCE There are currently no approved drugs to treat infections with alphaviruses. We previously identified a novel series of compounds with activity against these potentially devastating pathogens (J. A. Sindac et al., J. Med. Chem. 55:3535-3545, 2012, doi:http://dx.doi.org/10.1021/jm300214e; W. Peng et al., J. Infect. Dis. 199:950-957, 2009, doi:http://dx.doi.org/10.1086/597275; J. A. Sindac et al., J. Med. Chem. 56:9222-9241, 2013, http://dx.doi.org/10.1021/jm401330r). We have now produced third-generation compounds with enhanced potency, and this manuscript provides detailed information on the antiviral activity of these advanced-generation compounds, including activity in an animal model. The results of this study represent a notable achievement in the continued development of this novel class of antiviral inhibitors.


Assuntos
Antivirais/farmacologia , Vírus da Encefalite Equina do Oeste/efeitos dos fármacos , Encefalomielite Equina/tratamento farmacológico , Indóis/farmacologia , Piridinas/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Antivirais/síntese química , Bunyaviridae/efeitos dos fármacos , Bunyaviridae/crescimento & desenvolvimento , Linhagem Celular , Vírus da Encefalite Equina do Oeste/crescimento & desenvolvimento , Vírus da Encefalite Equina do Oeste/patogenicidade , Encefalomielite Equina/mortalidade , Encefalomielite Equina/virologia , Feminino , Indóis/síntese química , Concentração Inibidora 50 , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/virologia , Paramyxoviridae/efeitos dos fármacos , Paramyxoviridae/crescimento & desenvolvimento , Picornaviridae/efeitos dos fármacos , Picornaviridae/crescimento & desenvolvimento , Biossíntese de Proteínas/efeitos dos fármacos , Piridinas/síntese química , Replicon/efeitos dos fármacos , Relação Estrutura-Atividade , Análise de Sobrevida
5.
Uirusu ; 57(1): 1-7, 2007 Jun.
Artigo em Japonês | MEDLINE | ID: mdl-18040149

RESUMO

Our knowledge about envelope virus budding has been dramatically increased, since L-domain motifs were identified within their matrix and retroviral Gag proteins which drive virus budding. These viral proteins have been shown to interact with host cellular proteins involved in endocytosis and/or multi-vesicular body (MVB) sorting via their L-domains. Since budding of many enveloped viruses have been reported to be dependent on the activity of cellular Vps4, which catalyzes the disassembly of ESCRT machinery in the final step of protein sorting, this cellular function is believed to be utilized for efficient virus budding. However, for many enveloped viruses, L-domain motifs have not yet been identified, and the involvement of MVB sorting machinery in virus budding is still unknown. In this review, we will focus on paramyxoviruses among such viruses, and discuss their budding with the latest information.


Assuntos
Paramyxoviridae/fisiologia , Animais , Membrana Celular/virologia , Citoplasma/virologia , Endocitose , Humanos , Paramyxoviridae/crescimento & desenvolvimento , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Virais/química , Proteínas Virais/fisiologia , Montagem de Vírus , Replicação Viral
6.
Intervirology ; 50(2): 115-22, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17191013

RESUMO

OBJECTIVES: To explore the effects of ectodomain sequences between HR1 and HR2 of F1 protein on the specific interaction with its homologous hemagglutinin-neuraminidase (HN) in paramyxoviruses. METHODS: Site-directed mutagenesis was used to obtain mutants containing new enzyme sites on the F genes of Newcastle disease virus (NDV) and human parainfluenza virus (hPIV), and four DNA segments located between the HR1 and HR2 (NDV F-1, hPIV F-1, NDV F-2 and hPIV F-2) were obtained by cutting mutant F genes with specific endonucleases. Gene recombination was used to get chimeric F proteins NDV-C1 and hPIV-C1 by exchanging NDV F-1 and hPIV F-1 each other, and NDV-C2 and hPIV-C2 were also obtained by the same way. All the mutants and chimeric F proteins were co-expressed with their homologous or heterologous HN proteins in eukaryocytes. The fusion functions were assayed with Giemsa staining and reporter gene method for qualitative and quantitative analyses, respectively. The cell surface expression of F proteins was assayed with fluorescence-activated cell sorter (FACS) for quantitative analysis. RESULTS: All the mutants of F proteins had the same functions as their relevant wild types. Chimeric F proteins NDV-C1 and hPIV-C1 had 76.34 and 65.82% of fusion activities, and NDV-C2 and hPIV-C2 had 96.25 and 93.78% of fusion activities, respectively, as compared with their relevant wild types. The analysis of FACS indicated that all the mutants and chimeric F proteins had almost the same expression efficiencies as their relevant wild types. CONCLUSIONS: The segments of NDV F-1 and hPIV F-1 were important for their specific membrane fusion, but NDV F-2 and hPIV F-2 were not.


Assuntos
Vírus da Doença de Newcastle/fisiologia , Paramyxoviridae/crescimento & desenvolvimento , Respirovirus/fisiologia , Proteínas Virais de Fusão/fisiologia , Internalização do Vírus , Citometria de Fluxo , Genes Reporter , Proteína HN/fisiologia , Histocitoquímica , Imuno-Histoquímica , Microscopia , Mutagênese Sítio-Dirigida , Vírus da Doença de Newcastle/genética , Paramyxoviridae/genética , Recombinação Genética , Respirovirus/genética , Proteínas Virais de Fusão/genética , Proteínas Virais/fisiologia , beta-Galactosidase/análise , beta-Galactosidase/genética
7.
Microbiol Immunol ; 49(9): 835-44, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16172538

RESUMO

Effects of proteasome inhibitors on the replication of a paramyxovirus in comparison with the effects on replication of an orthomyxovirus and rhabdovirus were investigated. Treatment of Sendai virus (SeV)-infected LLC-MK2 cells with 50 microM MG132 reduced virus growth to ca. 1/10,000, and treatment with different concentrations of MG132 reduced virus growth in a dose-dependent manner. Released amounts of viral proteins were reduced in correspondence with decrease in infectivity. The inhibition of virus maturation was confirmed by an SeV-like particle formation system. Lactacystin also impaired SeV growth and zLL impaired the growth to a lesser extent, suggesting involvement of proteasomes in the restriction of virus growth. In the presence of MG132, localizations of the M protein and viral F and HN glycoproteins on the cell membrane appeared to be partly dissociated, although the viral glycoproteins were normally transported to the cell surface. These results suggest that an early step of SeV assembly was disturbed by proteasome inhibitors. The relationship of the results with ubiquitin is also discussed. SeV maturation was less susceptible and resistant to MG132 in CV1 cells and A549 cells, respectively, indicating cell specificity of the drug effect. Release of vesicular stomatitis virus also showed high susceptibility to MG132 and release of influenza virus A/WSN/33 was only mildly susceptible to the drug in LLC-MK2 cells. Effects of proteasome inhibitors on virus maturation are thus highly cell-specific and partly virus-specific.


Assuntos
Leupeptinas/farmacologia , Orthomyxoviridae/efeitos dos fármacos , Paramyxoviridae/efeitos dos fármacos , Inibidores de Proteassoma , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Animais , Linhagem Celular , Chlorocebus aethiops , Inibidores de Cisteína Proteinase/farmacologia , Proteína HN/metabolismo , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/crescimento & desenvolvimento , Macaca mulatta , Macrófagos/virologia , Orthomyxoviridae/crescimento & desenvolvimento , Paramyxoviridae/crescimento & desenvolvimento , Rhabdoviridae/efeitos dos fármacos , Rhabdoviridae/crescimento & desenvolvimento , Vírus Sendai/efeitos dos fármacos , Vírus Sendai/crescimento & desenvolvimento , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Proteínas Virais de Fusão/metabolismo , Proteínas da Matriz Viral/metabolismo , Montagem de Vírus/efeitos dos fármacos , Montagem de Vírus/fisiologia , Replicação Viral/efeitos dos fármacos , Replicação Viral/fisiologia
8.
J Virol ; 79(13): 8591-601, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15956600

RESUMO

Some paramyxovirus V proteins induce STAT protein degradation, and the amino acids essential for this process in the human parainfluenza virus type 2 (hPIV2) V protein have been studied. Various recombinant hPIV2s and cell lines constitutively expressing various mutant V proteins were generated. We found that V proteins with replacement of Cys residues of the Cys cluster were still able to bind STATs but were unable to induce their degradation. The hPIV2 V protein binds STATs via a W-(X)3-W-(X)9-W Trp motif located just upstream of the Cys cluster. Replacements of two or more Trp residues in this motif resulted in a failure to form a V/STAT2 complex. We have also identified two Phe residues of the hPIV2 V protein that are essential for STAT degradation, namely, Phe207, lying within the Cys cluster, and Phe143, in the P/V common region of the protein. Interestingly, infection of BHK cells with hPIV2 led to the specific degradation of STAT1 and not STAT2. Other evidence for the cell species specificity of hPIV2-induced STAT degradation is presented. Finally, a V-minus hPIV2, which can express only the P protein from its P gene, was generated and partially characterized. In contrast to V-minus viruses of other paramyxovirus genera, this V-minus rubulavirus was highly debilitated, and its growth even in Vero cells was very limited. The structural rubulavirus V proteins, as expected, are thus clearly important in promoting virus growth, independent of their anti-interferon (IFN) activity. Interestingly, many of the residues that are essential for anti-IFN activity, e.g., the Cys of this cluster and Phe207 within this cluster, as well as the Trp of this motif, are also essential for promoting virus growth.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Paramyxoviridae/genética , Transativadores/metabolismo , Proteínas Virais/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Genes Reporter , Células HeLa , Humanos , Dados de Sequência Molecular , Paramyxoviridae/crescimento & desenvolvimento , Plasmídeos/genética , Ligação Proteica , Fator de Transcrição STAT2 , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transativadores/química , Transativadores/genética , Células Vero , Proteínas Virais/genética , Proteínas Virais/metabolismo
9.
Virus Res ; 106(2): 133-45, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15567493

RESUMO

Components of paramyxoviruses are assembled at the plasma membrane of infected cells, and progeny viruses are formed by the budding process. Although the molecular mechanisms that drive budding (membrane curving and "pinching-off" reaction) are not well understood, the viral matrix (M) protein is thought to play a major role in the process. The M protein forms a dense layer tightly associated with the inner leaflet of the plasma membrane of infected cells. Expression of the M protein of some paramyxoviruses results in the formation and release of virus-like particles that contain the M protein; thus, in these viruses, the M protein alone can apparently trigger all steps required for the formation and release of virus-like particles. M also interacts specifically with viral envelope glycoproteins and nucleocapsids and is involved in directed transport of viral components to the budding site at the apical surface of polarized cells. In addition, protein-protein interactions between M and the cytoplasmic tail of viral glycoproteins and between M and the nucleocapsid affect the efficiency of virus production. The structural organization of the virion and the functions of the M protein clearly indicate that this protein orchestrates the budding of paramyxovirus.


Assuntos
Proteínas do Capsídeo/genética , Paramyxoviridae/genética , Vírion/genética , Animais , Proteínas do Capsídeo/fisiologia , Humanos , Paramyxoviridae/crescimento & desenvolvimento , Paramyxoviridae/fisiologia , Vírion/química
10.
J Clin Virol ; 30(1): 100-5, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15072762

RESUMO

Respiratory viruses cause significant morbidity and mortality. The management of these infections can be improved by a rapid diagnosis and administration of available virus-specific therapy. The goal of this study was to compare R-Mix, an engineered tissue monolayer for rapid shell vial (SV) diagnosis of viral respiratory infections, with conventional tissue culture (TC) and conventional respiratory SV (primary rhesus monkey kidney (RhMK) and Hep2 monolayers). The primary outcome measure was sensitivity for detection of influenza A and B, respiratory syncytial virus, parainfluenza 1-3, and adenovirus. The study was performed in two phases: (1) the three methods were compared using 250 nasal washes from children with lower respiratory tract infections; (2) a modified R-Mix SV harvesting schedule (SV were harvested at 24 and 120 h) was compared with TC and conventional RhMK/Hep2 SV using 311 respiratory specimens. A total of 110 viruses were identified in the first and 55 in the second phase. Diagnostic accuracies of R-Mix harvested at 24, 48, and 120 h were 98%, whereas for TC varied between 99 and 100%, and for RhMK/Hep2 SV between 98 and 99%. Sensitivities of R-Mix harvested at 24, 48, and 120 h were 26, 75, and 47%, respectively, whereas for TC varied between 60 and 94%, and for RhMK/Hep2 SV between 62 and 85%. R-Mix harvested at 48 h represent a valuable substitute for RhMK/Hep2 SV because they have comparable sensitivities and diagnostic accuracies, but R-Mix offers several technical advantages. In contrast, R-Mix harvested at 24h did not seem a very useful diagnostic tool. The utility of R-Mix harvested at 120 h, which accelerated the diagnosis of 16% of positive specimens in study phase 2, needs further investigation.


Assuntos
Infecções por Orthomyxoviridae/diagnóstico , Orthomyxoviridae/isolamento & purificação , Infecções por Paramyxoviridae/diagnóstico , Paramyxoviridae/isolamento & purificação , Infecções Respiratórias/diagnóstico , Adenovírus Humanos/crescimento & desenvolvimento , Adenovírus Humanos/isolamento & purificação , Animais , Linhagem Celular , Criança , Humanos , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza A/isolamento & purificação , Vírus da Influenza B/crescimento & desenvolvimento , Vírus da Influenza B/isolamento & purificação , Líquido da Lavagem Nasal/virologia , Orthomyxoviridae/crescimento & desenvolvimento , Infecções por Orthomyxoviridae/virologia , Vírus da Parainfluenza 1 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 1 Humana/isolamento & purificação , Vírus da Parainfluenza 2 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 2 Humana/isolamento & purificação , Vírus da Parainfluenza 3 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 3 Humana/isolamento & purificação , Paramyxoviridae/crescimento & desenvolvimento , Infecções por Paramyxoviridae/virologia , Valor Preditivo dos Testes , Vírus Sincicial Respiratório Humano/crescimento & desenvolvimento , Vírus Sincicial Respiratório Humano/isolamento & purificação , Infecções Respiratórias/virologia , Sensibilidade e Especificidade , Fatores de Tempo , Cultura de Vírus
11.
J UOEH ; 25(3): 295-305, 2003 Sep 01.
Artigo em Japonês | MEDLINE | ID: mdl-14503041

RESUMO

Joyce et al. proposed the hypothesis that the sugar moiety of nucleic acid evolved from some achiral, stable and acyclic sugar into the ribose or the deoxyribose. According to their hypothesis, we designed and synthesized new pseudonucleotides having pentaerythritol moiety as a sugar moiety of nucleic acid and estimated their biological activities. Although all chemicals were not toxic to Lepidium sativum, Rhodotolula rubra and Cercospora kikuchi in the eucaryotes, three compounds having adenine, benzimidazole or 6-chloropurine residue as the base component of nucleotide exhibited the growth inhibiting activity to a prokaryote Spirulina platensis at 100 ppm. In the plaque formation test with Vero cells, a chemical with 6-chloropurine moiety inhibited 65.7% of plaque formation by Herpes simplex virus(HSV-1) at 500 ppm. Three chemicals with 6-chloropurine, 2-mercaptomethylbenzimidazole or guanine as the nucleic base moiety inhibited 62.3, 63.1 and 52.5% of plaque formation by Parainfluenza virus(PIFV) at 500 ppm, respectively. The prepared chemicals exhibited no effect on the Vero cells at the same concentration.


Assuntos
Carboidratos/química , Evolução Molecular , Ácidos Nucleicos/química , Nucleotídeos/química , Nucleotídeos/síntese química , Animais , Antivirais/síntese química , Antivirais/química , Antivirais/farmacologia , Células Cultivadas , Chlorocebus aethiops , Cianobactérias/crescimento & desenvolvimento , Depressão Química , Herpesvirus Humano 1/crescimento & desenvolvimento , Nucleotídeos/farmacologia , Paramyxoviridae/crescimento & desenvolvimento , Relação Estrutura-Atividade , Células Vero/virologia
12.
J Virol ; 76(20): 10109-21, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12239285

RESUMO

The V protein of the paramyxovirus simian virus 5 (SV5) is responsible for targeted degradation of STAT1 and the block in alpha/beta interferon (IFN-alpha/beta) signaling that occurs after SV5 infection of human cells. We have analyzed the growth properties of a recombinant SV5 that was engineered to be defective in targeting STAT1 degradation. A recombinant SV5 (rSV5-P/V-CPI-) was engineered to contain six naturally occurring P/V protein mutations, three of which have been shown in previous transfection experiments to disrupt the V-mediated block in IFN-alpha/beta signaling. In contrast to wild-type (WT) SV5, human cells infected with rSV5-P/V-CPI- had STAT1 levels similar to those in mock-infected cells. Cells infected with rSV5-P/V-CPI- were found to express higher-than-WT levels of viral proteins and mRNA, suggesting that the P/V mutations had disrupted the regulation of viral RNA synthesis. Despite the inability to target STAT1 for degradation, single-step growth assays showed that the rSV5-P/V-CPI- mutant virus grew better than WT SV5 in all cell lines tested. Unexpectedly, cells infected with rSV5-P/V-CPI- but not WT SV5 showed an activation of a reporter gene that was under control of the IFN-beta promoter. The secretion of IFN from cells infected with rSV5-P/V-CPI- but not WT SV5 was confirmed by a bioassay for IFN. The rSV5-P/V-CPI- mutant grew to higher titers than did WT rSV5 at early times in multistep growth assays. However, rSV5-P/V-CPI- growth quickly reached a final plateau while WT rSV5 continued to grow and produced a final titer higher than that of rSV5-P/V-CPI- by late times postinfection. In contrast to WT rSV5, infection of a variety of cell lines with rSV5-P/V-CPI- induced cell death pathways with characteristics of apoptosis. Our results confirm a role for the SV5 V protein in blocking IFN signaling but also suggest new roles for the P/V gene products in controlling viral gene expression, the induction of IFN-alpha/beta synthesis, and virus-induced apoptosis.


Assuntos
Apoptose , Interferon-alfa/biossíntese , Interferon beta/biossíntese , Paramyxoviridae/patogenicidade , Fosfoproteínas/fisiologia , Proteínas Virais/fisiologia , Proteínas Estruturais Virais/fisiologia , Substituição de Aminoácidos , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Efeito Citopatogênico Viral , Proteínas de Ligação a DNA/metabolismo , Genes Virais , Proteínas de Fluorescência Verde , Humanos , Interferon-alfa/genética , Interferon beta/genética , Proteínas Luminescentes/genética , Mutagênese , Paramyxoviridae/genética , Paramyxoviridae/crescimento & desenvolvimento , Fosfoproteínas/genética , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , RNA Viral/biossíntese , Proteínas de Ligação a RNA , Fator de Transcrição STAT1 , Transativadores/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , Proteínas Estruturais Virais/genética
13.
J Virol ; 75(9): 4068-79, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287556

RESUMO

In some cell types the paramyxovirus simian virus 5 (SV5) causes little cytopathic effect (CPE) and infection continues productively for long periods of time; e.g., SV5 can be produced from MDBK cells for up to 40 days with little CPE. SV5 differs from most paramyxoviruses in that it encodes a small (44-amino-acid) hydrophobic integral membrane protein (SH). When MDBK cells were infected with a recombinant SV5 containing a deletion of the SH gene (rSV5DeltaSH), the MDBK cells exhibited an increase in CPE compared to cells infected with wild-type SV5 (recovered from cDNA; rSV5). The increased CPE correlated with an increase in apoptosis in rSV5DeltaSH-infected cells over mock-infected and rSV5-infected cells when assayed for annexin V binding, DNA content (propidium iodide staining), and DNA fragmentation (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling assay). In rSV5DeltaSH-infected MDBK cells an increase in caspase-2 and caspase-3 activities was observed. By using peptide inhibitors of individual caspases it was found that caspase-2 and caspase-3 were activated separately in rSV5DeltaSH-infected cells. Expression of caspase-2 and -3 in rSV5DeltaSH-infected MDBK cells appeared not to require STAT1 protein, as STAT1 protein could not be detected in SV5-infected MDBK cells. When mutant mice homologous for a targeted disruption of STAT1 were used as a model animal system and infected with the viruses it was found that rSV5DeltaSH caused less mortality than wild-type rSV5, consistent with the notion of clearance of apoptotic cells in a host species.


Assuntos
Apoptose , Paramyxoviridae/fisiologia , Proteínas Oncogênicas de Retroviridae/metabolismo , Animais , Caspase 2 , Caspase 3 , Caspases/metabolismo , Bovinos , Linhagem Celular , Efeito Citopatogênico Viral , Proteínas de Ligação a DNA/metabolismo , Cães , Ativação Enzimática , Haplorrinos , Células HeLa , Humanos , Paramyxoviridae/genética , Paramyxoviridae/crescimento & desenvolvimento , Paramyxoviridae/metabolismo , Proteínas Oncogênicas de Retroviridae/genética , Fator de Transcrição STAT1 , Transativadores/metabolismo
15.
J Gen Virol ; 74 ( Pt 4): 631-41, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8468554

RESUMO

A previously unidentified morbillivirus was isolated from two harbour porpoises (Phocoena phocoena) that had died in the Dutch Waddensea (North Sea) in 1990. This porpoise morbillivirus (PMV) and a dolphin morbillivirus (DMV), which had recently caused a heavy mortality in Mediterranean striped dolphins (Stenella coeruleoalba), were compared antigenically with other members of the genus Morbillivirus, including the newly recognized phocine distemper virus type 1. DMV and PMV proved to be similar but distinct morbillivurses, closely related to rinderpest virus and peste-des-petitsruminants virus. Cell cultures of cetacean, pinniped, ruminant and canine origin showed a different pattern of susceptibility to DMV and PMV infection. Ruminants and dogs proved to be susceptible to experimental infection with DMV and PMV, which both caused a transient leukopenia most pronounced in the ruminants. Pre-exposure of dogs to DMV and PMV protected them from developing CDV viraemia and clinical signs upon challenge infection with virulent CDV. A serological survey among stranded animals of different cetacean species in Europe indicated that infections with DMV- and PMV-like morbilliviruses are not uncommon among these aquatic mammals.


Assuntos
Golfinhos/microbiologia , Paramyxoviridae/classificação , Animais , Antígenos Virais/imunologia , Artiodáctilos/microbiologia , Reações Cruzadas , Cães/microbiologia , Europa (Continente) , Paramyxoviridae/crescimento & desenvolvimento , Paramyxoviridae/imunologia , Infecções por Respirovirus/imunologia , Infecções por Respirovirus/fisiopatologia , Especificidade da Espécie , Replicação Viral
17.
J Virol Methods ; 39(1-2): 83-90, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1430067

RESUMO

CDC/EU.HMEC-1 is the first immortalized human microvascular endothelial cell line that retains morphologic, phenotypic, and functional characteristics of a normal human microvascular endothelial cell. This study evaluates a variety of viruses and their effects on this human endothelial cell line. The data indicate that adenoviruses, some herpesviruses, reoviruses and most picornaviruses grow well in HMEC-1, with distinctive cytopathic effects. The paramyxoviruses, however, do not appear to propagate, nor does HIV. The findings indicate that microvascular endothelial cells may act as a reservoir of these viruses; it also suggests the possibility that microvascular endothelium could be involved in the processing and presentation of antigen to immune cells.


Assuntos
Endotélio Vascular/microbiologia , Vírus/patogenicidade , Adenoviridae/crescimento & desenvolvimento , Adenoviridae/patogenicidade , Linhagem Celular , Efeito Citopatogênico Viral , Endotélio Vascular/citologia , HIV/crescimento & desenvolvimento , HIV/patogenicidade , Herpesviridae/crescimento & desenvolvimento , Herpesviridae/patogenicidade , Humanos , Paramyxoviridae/crescimento & desenvolvimento , Paramyxoviridae/patogenicidade , Picornaviridae/crescimento & desenvolvimento , Picornaviridae/patogenicidade , Reoviridae/crescimento & desenvolvimento , Reoviridae/patogenicidade , Virologia/métodos , Vírus/crescimento & desenvolvimento
18.
FEBS Lett ; 296(3): 274-8, 1992 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-1537403

RESUMO

In chick embryo, certain paramyxoviruses mainly target the chorioallantois and the allantoamnion and show no extensive further spreading in the other organs. This has been explained by the possible presence of an endoprotease activating the viral fusion glycoprotein precursor in the allantoic and the amniotic fluid, and its absence in other places or organs. We previously isolated such an endoprotease from the allantoic fluid and demonstrated its identity with the clotting factor Xa. Exactly the same endoprotease by all the criteria including the N-terminal amino acid sequence was now isolated from the amniotic fluid. Thus, the factor Xa seems to be a major host determinant of the viral tropism in chick embryo.


Assuntos
Líquido Amniótico/enzimologia , Fator Xa/fisiologia , Paramyxoviridae/crescimento & desenvolvimento , Serina Endopeptidases/fisiologia , Ativação Viral , Sequência de Aminoácidos , Animais , Embrião de Galinha , Cromatografia em Gel , Eletroforese em Gel de Poliacrilamida , Fator Xa/química , Fator Xa/isolamento & purificação , Dados de Sequência Molecular , Alinhamento de Sequência , Serina Endopeptidases/química , Serina Endopeptidases/isolamento & purificação
19.
Virology ; 175(2): 345-57, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2158179

RESUMO

Virus-induced cell fusion has been examined in a series of stable cell lines which were originally selected for resistance to the fusogenic effects of polyethylene glycol (PEG). For a wide variety of viruses, including murine hepatitis virus (a coronavirus), vesicular stomatitis virus (a rhabdovirus), and two paramyxoviruses (Sendai virus and SV5), susceptibility to virus-induced fusion was found to be inversely correlated with susceptibility to PEG-induced fusion. This phenomenon was observed both for cell fusion occurring in the course of viral infection and for fusion induced "from without" by the addition of high titers of noninfectious or inactivated virus. The fusion-altered cell lines (fusible by virus but not by PEG) are characterized by their unusual lipid composition, including marked elevation of saturated fatty acids and the presence of an unusual ether-linked neutral lipid. To test the association between lipid composition and fusion, acyl chain saturation was manipulated by supplementing the culture medium with exogenous fatty acids. In such experiments, it was possible to control the responses of these cells to both viral and chemical fusogens. Increasing the cellular content of saturated fatty acyl chains increased the susceptibility of cells to viral fusion and decreased susceptibility to PEG-induced fusion, whereas lowering fatty acid saturation had the opposite effect. Thus, parallel cultures of cells can be either driven toward the PEG-fusible/virus-fusion-resistant phenotype of the parental cells or rendered susceptible to viral fusion but resistant to PEG-induced fusion, solely by the alteration of cellular lipids. The ability of cellular lipid composition to regulate virus-induced membrane fusion suggests a possible role for lipids in viral infection and pathogenesis.


Assuntos
Fusão Celular/fisiologia , Lipídeos/análise , Vírus da Hepatite Murina/fisiologia , Paramyxoviridae/fisiologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Linhagem Celular , Embrião de Galinha , Células Clonais , Ácidos Graxos/análise , Ácidos Graxos/metabolismo , Células L , Camundongos , Vírus da Hepatite Murina/crescimento & desenvolvimento , Vírus da Parainfluenza 1 Humana/crescimento & desenvolvimento , Vírus da Parainfluenza 1 Humana/fisiologia , Paramyxoviridae/crescimento & desenvolvimento , Polietilenoglicóis/farmacologia , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento
20.
J Gen Virol ; 71 ( Pt 3): 609-13, 1990 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2313267

RESUMO

The porcine paramyxovirus is a newly identified agent of a fatal disease in piglets, endemic in Mexico since 1980, where it was seen around the town of La Piedad, Michoacan, Mexico (hence LPM virus). At least six [35S]methionine-labelled proteins could be resolved by SDS-PAGE and five of them were clearly immunoprecipitated. Selective labelling of LPMV-infected cells with [3H]glucosamine revealed two bands with an Mr of about 66K and 59K, corresponding to the two viral glycoproteins, the haemagglutinin-neuraminidase protein and the fusion protein. Labelling of virus with [32P]orthophosphate disclosed one band with an Mr of 52K, corresponding to the phosphoprotein. Analysis of nucleocapsids obtained from purified virus or from a permanently infected cell line revealed one major band with an Mr of 68K, the nucleoprotein. Two other proteins were also identified, the large protein and the matrix protein, with apparent Mr of about 200K and 40K, respectively. The protein migration pattern of LPMV was compared, by SDS-PAGE, with that of Newcastle disease virus, bovine parainfluenza 3 virus and Sendai virus. Differences in the Mr of LPMV proteins and the proteins of these paramyxoviruses were observed. We propose that LPMV should be classified as a novel member of the genus Paramyxovirus.


Assuntos
Paramyxoviridae/análise , Proteínas Estruturais Virais/isolamento & purificação , Animais , Autorradiografia , Capsídeo/isolamento & purificação , Capsídeo/ultraestrutura , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Metionina/metabolismo , Microscopia Eletrônica , Peso Molecular , Paramyxoviridae/crescimento & desenvolvimento , Radioisótopos de Enxofre , Suínos , Proteínas do Core Viral/isolamento & purificação , Proteínas do Core Viral/ultraestrutura , Proteínas Estruturais Virais/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...