Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 11(9)2019 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-31450681

RESUMO

Newcastle disease (ND) is responsible for significant economic losses in the poultry industry. The disease is caused by virulent strains of Avian avulavirus 1 (AAvV-1), a species within the family Paramyxoviridae. We developed a recombinant construct based on the herpesvirus of turkeys (HVT) as a vector expressing two genes: F and HN (HVT-NDV-F-HN) derived from the AAvV-1 genotype VI ("pigeon variant" of AAvV-1). This recombinant viral vaccine candidate was used to subcutaneously immunize one group of specific pathogen-free (SPF) chickens and two groups of broiler chickens (20 one-day-old birds/group). Humoral immune response was evaluated by hemagglutination-inhibition test and enzyme-linked immunosorbent assay (ELISA). The efficacy of the immunization was assessed in two separate challenge studies performed at 6 weeks of age with the use of virulent AAvV-1 strains representing heterologous genotypes IV and VII. The developed vaccine candidate elicited complete protection in SPF chickens since none of the birds became sick or died during the 2-week observation period. In the broiler groups, 90% and 100% clinical protection were achieved after challenges with AAvV-1 of IV and VII genotypes, respectively. We found no obvious relationship between antibody levels and protection assessed in broilers in the challenge study. The developed recombinant HVT-NDV-F-HN construct containing genes from a genotype VI AAvV-1 offers promising results as a potential vaccine candidate against ND in chickens.


Assuntos
Proteína HN/imunologia , Imunização/veterinária , Vírus da Doença de Newcastle , Vacinas Sintéticas/imunologia , Proteínas Virais de Fusão/imunologia , Animais , Antígenos Virais/biossíntese , Antígenos Virais/genética , Galinhas/virologia , Proteção Cruzada , Genes Virais , Proteína HN/biossíntese , Proteína HN/genética , Testes de Inibição da Hemaglutinação , Herpesvirus Meleagrídeo 1/genética , Herpesvirus Meleagrídeo 1/imunologia , Herpesvirus Meleagrídeo 1/metabolismo , Imunidade Heteróloga , Doença de Newcastle/imunologia , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/virologia , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Vacinas Sintéticas/virologia , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/genética , Vacinas Virais/biossíntese , Vacinas Virais/imunologia
2.
Drug Discov Ther ; 11(5): 246-252, 2017 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-29070744

RESUMO

The effect of glycyrrhizin on the replication of human parainfluenza virus type 2 (hPIV-2) was examined. Cell fusion induced by hPIV-2 was inhibited by glycyrrhizin, and glycyrrhizin reduced the number of viruses released from the cells. Glycyrrhizin did not change cell morphology at 1 day of culture, but caused some damage at 4 days, as determined by the effect on actin microfilaments. However, it affected the cell viability at 1 day: about 20% of the cells were not alive by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay at 1 day of culture. Real-time polymerase chain reaction (PCR) and PCR showed that virus genome synthesis was largely inhibited. mRNA synthesis was also inhibited by glycyrrhizin. Viral protein synthesis was largely inhibited as observed by an indirect immunofluorescence study. Multinucleated giant cell formation was studied using a recombinant green fluorescence protein (GFP)-expressing hPIV-2 without matrix protein (rhPIV-2ΔMGFP). A few single cells with fluorescence were observed, but the formation of giant cells was completely blocked. Taken together, it was shown that viral genome, mRNA and protein syntheses, including F and HN proteins, were inhibited by glycyrrhizin, and consequently multinucleated giant cell formation was not observed and the infectious virus was not detected in the culture medium.


Assuntos
Ácido Glicirrízico/farmacologia , Vírus da Parainfluenza 2 Humana/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Citoesqueleto de Actina/efeitos dos fármacos , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Genoma Viral/efeitos dos fármacos , Células Gigantes/efeitos dos fármacos , Proteína HN/biossíntese , Proteína HN/efeitos dos fármacos , Macaca mulatta , Vírus da Parainfluenza 2 Humana/genética , Vírus da Parainfluenza 2 Humana/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , RNA Viral/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/efeitos dos fármacos , Proteínas Virais/biossíntese , Proteínas Virais/efeitos dos fármacos , Replicação Viral/genética
3.
Protein Expr Purif ; 128: 1-7, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27496728

RESUMO

Blue eye disease caused by Porcine rubulavirus (PorPV) is an endemic viral infection of swine causing neurological and respiratory disease in piglets, and reproductive failure in sows and boars. The hemagglutinin-neuraminidase (HN) glycoprotein of PorPV is the most abundant component in the viral envelope and the main target of the immune response in infected animals. In this study, we expressed the HN-PorPV-recombinant (rHN-PorPV) protein in an Escherichia coli system and analyzed the immune responses in mice. The HN gene was cloned from the reference strain PorPV-La Piedad Michoacan Virus (GenBank accession number BK005918), into the pDual expression vector. The expressed protein was identified at a molecular weight of 61.7 kDa. Three-dimensional modeling showed that the main conformational and functional domains of the rHN-PorPV protein were preserved. The antigenicity of the expressed protein was confirmed by Western blot with a monoclonal antibody recognizing the HN, and by testing against serum samples from pigs experimentally infected with PorPV. The immunogenicity of the rHN-PorPV protein was tested by inoculation of BALB/c mice with AbISCO-100(®) as adjuvant. Analysis of the humoral immune responses in mice showed an increased level of specific antibodies 14 days after the first immunization, compared to the control group (P < 0.0005). The results show the ability of the rHN-PorPV protein to induce an antibody response in mice. Due to its immunogenic potential, the rHN-PorPV protein will be further evaluated in pig trials for its suitability for prevention and control of blue eye disease.


Assuntos
Clonagem Molecular , Expressão Gênica , Proteína HN , Imunogenicidade da Vacina , Rubulavirus , Vacinas Virais , Animais , Escherichia coli , Feminino , Proteína HN/biossíntese , Proteína HN/imunologia , Proteína HN/isolamento & purificação , Proteína HN/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Rubulavirus/enzimologia , Rubulavirus/imunologia , Suínos , Vacinas Virais/biossíntese , Vacinas Virais/imunologia
4.
J Vet Diagn Invest ; 23(3): 519-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21908282

RESUMO

The hemagglutinin-neuraminidase (HN) glycoprotein of Newcastle disease virus (NDV) constitutes, together with the fusion glycoprotein, the main surface antigen of this avian pathogen, which causes a highly contagious disease, relevant economically worldwide. The purpose of this work was to obtain the HN glycoprotein as a soluble antigen in culture supernatants of recombinant baculovirus-infected Spodoptera frugiperda (Sf9) cells and to evaluate its application to the development of a recombinant enzyme-linked immunosorbent assay (rELISA) for the analysis of chicken sera. A transfer vector for baculovirus containing the sequence of a melittin signal peptide was constructed and the sequence coding for HN protein without its own signal peptide was cloned. The recombinant protein was secreted and recovered easily from the culture medium of Sf9-infected cells. The recombinant protein was evaluated as antigen for ELISA coating the plates with the recovered HN using 79 positive and 142 negative samples. The Cohen kappa value resulted 0.91, indicating excellent agreement between the rELISA and the hemagglutinin inhibition tests. The rELISA was also compared with a commercial ELISA, finding high levels of agreement between both assays. The present results show that the cloning strategy developed yielded the HN protein free in the cell culture supernatant and that the recombinant protein retained its reactivity with anti-NDV HN antibodies in chicken sera.


Assuntos
Ensaio de Imunoadsorção Enzimática/veterinária , Proteína HN/biossíntese , Doença de Newcastle/diagnóstico , Vírus da Doença de Newcastle , Animais , Baculoviridae/genética , Células Cultivadas/virologia , Galinhas/virologia , Ensaio de Imunoadsorção Enzimática/métodos , Vetores Genéticos/genética , Testes de Inibição da Hemaglutinação/veterinária , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/metabolismo , Proteínas Recombinantes , Reprodutibilidade dos Testes , Spodoptera/virologia
5.
Cancer Gene Ther ; 16(4): 310-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18949016

RESUMO

The delivery of stimulatory signals to dendritic cells (DCs) in the tumor microenvironment could be an effective means to break tumor-induced tolerance. The work presented here evaluates the immunostimulatory properties of pathogen-associated molecular patterns (PAMPs), microbial molecules which bind Toll-like receptors and deliver activating signals to immune cells, when expressed in tumor cells using adenoviral (Ad) vectors. In vitro, transduction of A549 tumor cells with Ad vectors expressing either flagellin from Listeria monocytogenes or P40 protein from Klebsiella pneumoniae induced the maturation of human monocyte-derived DCs in co-cultures. In mixed lymphocyte reactions (MLRs), Ad-flagellin and Ad-P40 transduction of tumor cells stimulated lymphocyte proliferation and the secretion of IFN-gamma. In vivo, these vectors were used either as stand-alone immunoadjuvants injected intratumorally or as vaccine adjuvants combined with a tumor antigen-expressing vector. When Ad-PAMPs were administered intratumorally to mice bearing subcutaneous syngeneic B16F0-CAR (cocksackie-adenovirus receptor) melanomas, tumor progression was transiently inhibited by Ad-P40. In a therapeutic vaccine setting, the combination of Ad-MUC1 and Ad-PAMP vectors injected subcutaneously delayed the growth of implanted RenCa-MUC1 tumors and improved tumor rejection when compared with vaccination with Ad-MUC1 alone. These results suggest that Ad-PAMPs could be effective immunoadjuvants for cancer immunotherapy.


Assuntos
Adenoviridae/genética , Proteínas da Membrana Bacteriana Externa/imunologia , Terapia Genética , Proteína HN/imunologia , Imunoterapia , Neoplasias/terapia , Animais , Proteínas da Membrana Bacteriana Externa/biossíntese , Proteínas da Membrana Bacteriana Externa/genética , Linhagem Celular Tumoral , Técnicas de Cocultura , Citocinas/biossíntese , Células Dendríticas/patologia , Células Dendríticas/fisiologia , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos , Proteína HN/biossíntese , Proteína HN/genética , Humanos , Ativação Linfocitária , Camundongos , Neoplasias/genética , Neoplasias/imunologia , Vírus da Doença de Newcastle/genética
6.
J Virol Methods ; 98(1): 53-61, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11543884

RESUMO

Human parainfluenza virus types 1 (hPIV-1), 2, and 3 represent significant respiratory pathogens for which no antiviral treatment is currently available. To characterize the biochemical functions of the hPIV-1 hemagglutinin-neuraminidase (HN) glycoprotein, a potential target for antiviral therapy, we cloned and expressed a soluble portion of hPIV-1 HN (amino acid residues 137-575), lacking the N-terminal hydrophobic membrane anchorage region, in insect cells using the baculovirus secretion expression system. The expressed HN protein was purified through cation-exchange chromatography followed by metal affinity chromatography, using the 6xHis epitope introduced at the carboxyl terminus of the recombinant protein. N-terminal amino acid sequence analysis of purified HN indicated that the honeybee melittin secretion signal peptide was correctly removed during post-translational processing. Further characterization revealed that the purified HN protein was N-glycosylated and exhibited neuraminidase activity whose characteristics resembled those of the native HN protein of hPIV-1 virions. The establishment of this expression and purification system has allowed us to further explore the biochemical characteristics of paramyxovirus HN and to obtain material that could be suitable for X-ray crystallography studies.


Assuntos
Proteína HN/biossíntese , Vírus da Parainfluenza 1 Humana/metabolismo , Sequência de Aminoácidos , Animais , Baculoviridae/genética , Linhagem Celular , Vetores Genéticos , Proteína HN/química , Proteína HN/genética , Humanos , Concentração de Íons de Hidrogênio , Insetos , Dados de Sequência Molecular , Neuraminidase/metabolismo , Níquel , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Cloreto de Sódio
7.
Virus Res ; 75(2): 169-77, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11325471

RESUMO

The genes coding for the surface glycoproteins hemagglutinin-neuraminidase (HN) of the peste des petits ruminants virus (PPRV) and hemagglutinin (H) of rinderpest virus (RPV) were cloned in a cytomagalovirus promoter driven expression vector and expressed transiently in mammalian cells. The protein expression was apparent 24 h after transfection and the expressed proteins were detected at the cell surface. The transiently expressed PPRV HN protein was found to be biologically active in possessing hemadsorption and neuraminidase activities. On the other hand, RPV H protein exhibited neuraminidase activity but was deficient in hemadsorption activity. The substrate specificity of the neuraminidase activity of these two proteins differed distinctly. The presence of neuraminidase activity in both PPRV HN and RPV H proteins is unusual among members of the morbillivirus genus.


Assuntos
Proteína HN/biossíntese , Proteína HN/genética , Vírus da Peste dos Pequenos Ruminantes/genética , Transfecção , Animais , Linhagem Celular , Chlorocebus aethiops , Citomegalovirus/genética , Regulação Enzimológica da Expressão Gênica/genética , Regulação Viral da Expressão Gênica/genética , Genes Virais/genética , Hemadsorção/genética , Humanos , Neuraminidase/genética , Vírus da Peste dos Pequenos Ruminantes/enzimologia , Proteínas Recombinantes/biossíntese , Células Vero , Proteínas Estruturais Virais/genética
8.
J Virol ; 74(12): 5619-28, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10823869

RESUMO

The P/C mRNA of Sendai virus (SeV) encodes a nested set of accessory proteins, C', C, Y1, and Y2, referred to collectively as C proteins, using the +1 frame relative to the open reading frame of phospho (P) protein and initiation codons at different positions. The C proteins appear to be basically nonstructural proteins as they are found abundantly in infected cells but greatly underrepresented in the virions. We previously created a 4C(-) SeV, which expresses none of the four C proteins, and concluded that the C proteins are categorically nonessential gene products but greatly contribute to viral full replication and infectivity (A. Kurotani et al., Genes Cells 3:111-124, 1998). Here, we further characterized the 4C(-) virus multiplication in cultured cells. The viral protein and mRNA synthesis was enhanced with the mutant virus relative to the parental wild-type (WT) SeV. However, the viral yields were greatly reduced. In addition, the 4C(-) virions appeared to be highly anomalous in size, shape, and sedimentation profile in a sucrose gradient and exhibited the ratios of infectivity to hemagglutination units significantly lower than those of the WT. In the WT infected cells, C proteins appeared to colocalize almost perfectly with the matrix (M) proteins, pretty well with an external envelope glycoprotein (hemagglutinin-neuraminidase [HN]), and very poorly with the internal P protein. In the absence of C proteins, there was a significant delay of the incorporation of M protein and both of the envelope proteins, HN and fusion (F) proteins, into progeny virions. These results strongly suggest that the accessory and basically nonstructural C proteins are critically required in the SeV assembly process. This role of C proteins was further found to be independent of their recently discovered function to counteract the antiviral action of interferon-alpha/beta. SeV C proteins thus appear to be quite versatile.


Assuntos
Respirovirus/fisiologia , Proteínas Virais/metabolismo , Montagem de Vírus , Animais , Western Blotting , Linhagem Celular , Centrifugação com Gradiente de Concentração , Chlorocebus aethiops , Imunofluorescência , Deleção de Genes , Proteína HN/biossíntese , Proteína HN/metabolismo , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Microscopia Imunoeletrônica , Fosfoproteínas/biossíntese , Fosfoproteínas/metabolismo , RNA Viral/biossíntese , RNA Viral/genética , Respirovirus/efeitos dos fármacos , Respirovirus/genética , Respirovirus/metabolismo , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/metabolismo , Proteínas da Matriz Viral/biossíntese , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/biossíntese , Proteínas Virais/genética , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
9.
Arch Virol ; 144(4): 781-6, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10365168

RESUMO

In order to evaluate the efficiency of the removal of sialic acid residues from the influenza virus hemagglutinin by the viral neuraminidase in the course of the virus replication cycle, CV-1 cells expressing the hemagglutinin of H7 subtype from an SV40-based vector were superinfected with influenza virus strain A/Duck/Ukraine/63 (H3N8) or A/USSR/90/77 (H1N1). Vector-expressed hemagglutinin was immunoprecipitated from cell lysates and analyzed by polyacrylamide gel electrophoresis. The data indicate that the removal of sialic acid residues from the vector-expressed H7 hemagglutinin by N1 neuraminidase of A/USSR/90/77 virus in the course of the virus replication cycle is incomplete. The results are discussed in connection with previously published data showing that the low activity of NA in wild-type influenza virus results in incomplete removal of sialic acid residues from virion components.


Assuntos
Proteína HN/genética , Vírus da Influenza A/fisiologia , Neuraminidase/metabolismo , Replicação Viral , Animais , Linhagem Celular , Chlorocebus aethiops , Patos , Vetores Genéticos , Proteína HN/biossíntese , Vírus da Influenza A/enzimologia , Vírus da Influenza A/genética , Vírus 40 dos Símios , Especificidade da Espécie
10.
Vaccine ; 16(16): 1496-503, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9711795

RESUMO

We have constructed recombinant (r) fowl pox viruses (FPVs) coexpressing chicken type I interferon (IFN) and/or hemagglutinin-neuraminidase (HN) and fusion (F) proteins of Newcastle disease virus (NDV). We administered rFPVs and FPV into embryonated chicken eggs at 17 days of embryonation or in chickens after hatch. Administration of FPV or rFPVs did not influence hatchability and survival of hatched chicks. In ovo or after hatch vaccination of chickens with the recombinant viruses resulted in protection against challenge with virulent FPV and NDV. Chickens vaccinated with FPV or FPV-NDV recombinant had significantly lower body weight 2 weeks following vaccination. This loss in body weight was not detected in chickens receiving FPV-IFN and FPV-NDV-IFN recombinants. Chickens vaccinated with FPV coexpressing IFN and NDV genes produced less antibodies against NDV in comparison with chickens vaccinated with FPV expressing NDV genes.


Assuntos
Vírus da Varíola das Aves Domésticas/genética , Vírus da Varíola das Aves Domésticas/imunologia , Varíola Aviária/prevenção & controle , Proteína HN/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/uso terapêutico , Animais , Formação de Anticorpos , Peso Corporal , Embrião de Galinha , Galinhas , Vírus da Varíola das Aves Domésticas/metabolismo , Expressão Gênica , Proteína HN/biossíntese , Proteína HN/genética , Interferon Tipo I/biossíntese , Vírus da Doença de Newcastle/metabolismo , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
11.
Wei Sheng Wu Xue Bao ; 38(5): 359-64, 1998 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-12549399

RESUMO

For the construction of transfer vector pFGHN1175-1, the gene encoding haemagglutinin-neuraminidase(HN) glycoprotein of newcastle disease virus(NDV) strain F48E8 was removed from plasmid pGEMHN, and inserted into the HindIII site of insertion vector pFG1175-1, downstream of P7.5 promotor. Chicken embryo fibroblast (CEF) cell cultures which had been infected with fowlpox virus (FPV) Chinese vaccine strain 282E4 for 3-4 hours were transfected with pFGHN1175-1 plasmid DNA by liposomal transfection. Recombinant FPV with blue plaques were selected and purified 3 times in CEF cell cultures overlaid agarose containing X-gal. PCR analysis and DNA dot-blotting hybridization assay indicated that the HN gene had inserted into the FPV genomic DNA. The expression of the NDV HN gene in recombinant FPV was confirmed by indirect immunofluorescence assay with specific monoclonal antibody.


Assuntos
Vírus da Varíola das Aves Domésticas/genética , Proteína HN/genética , Vírus da Doença de Newcastle/genética , Animais , Células Cultivadas , Embrião de Galinha , Fibroblastos/virologia , Vetores Genéticos , Proteína HN/biossíntese , Regiões Promotoras Genéticas , Recombinação Genética , Transfecção
12.
Virology ; 224(2): 465-76, 1996 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8874507

RESUMO

The role of cotranslational disulfide bond formation in the folding pathway of the hemagglutinin-neuraminidase (HN) glycoprotein of Newcastle disease virus was explored. Electrophoresis of pulse-labeled HN protein in the presence or absence of reducing agent showed that, characteristic of many glycoproteins, the nascent HN protein contains intramolecular disulfide bonds. As reported by Braakman et al. (EMBO J. 11, 1717-1722, 1992), incubation of cells in dithiothreitol (DTT) blocked the formation of these bonds. Removal of DTT after a pulse-label allowed for the subsequent formation of intramolecular disulfide bonds and folding of the molecule as assayed by the appearance of conformationally sensitive antigenic sites and by the formation of disulfide-linked dimers. However, the t1/2 for the formation of a conformationally sensitive antigenic site after synthesis in the presence of DTT was over twice that of the control. Furthermore, the order of appearance of the antigenic sites was different from the control, suggesting that inhibition of cotranslational disulfide bond formation altered the folding pathway of the protein. Similar results were obtained in a cell-free system containing membranes. The HN protein forced to form intramolecular disulfide bonds posttranslationally had no detectable neuraminidase or cell attachment activity, suggesting that the protein had an abnormal conformation.


Assuntos
Dissulfetos , Proteína HN/metabolismo , Vírus da Doença de Newcastle/metabolismo , Dobramento de Proteína , Animais , Antígenos Virais/biossíntese , Sítios de Ligação , Transporte Biológico , Células COS , Membrana Celular/metabolismo , Sistema Livre de Células , Ditiotreitol/farmacologia , Proteína HN/biossíntese , Cinética , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional
13.
J Virol ; 70(9): 6418-24, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8709274

RESUMO

Mice immunized with two intragastrically administered doses of a replication-deficient recombinant vaccinia virus containing the hemagglutinin and nucleoprotein genes from H1N1 influenza virus developed serum anti-H1 immunoglobulin G (IgG) antibody that completely protected the lungs from challenge with H1N1. Almost all of the mice given two intragastric doses also developed mucosal anti-H1 IgA antibody, and those with high anti-H1 IgA titers had completely protected noses. Intramuscular injection of the vaccine protected the lungs but not the noses from challenge. We also found that the vaccine enhanced recovery from infection caused by a shifted (H3N2) influenza virus, probably through the induction of nucleoprotein-specific cytotoxic T-lymphocyte activity. A replication-deficient, orally administered, enteric-coated, vaccinia virus-vectored vaccine might safely protect humans against influenza.


Assuntos
Proteína HN/imunologia , Hemaglutininas Virais/imunologia , Vírus da Influenza A/imunologia , Vacinas contra Influenza , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas Sintéticas , Vaccinia virus/imunologia , Administração Oral , Animais , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Feminino , Genes Virais , Proteína HN/biossíntese , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Hemaglutininas Virais/biossíntese , Imunoglobulina A/biossíntese , Imunoglobulina A/sangue , Vírus da Influenza A/isolamento & purificação , Vacinas contra Influenza/administração & dosagem , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Vacinas Sintéticas/administração & dosagem , Vaccinia virus/fisiologia , Replicação Viral
14.
J Virol ; 70(9): 6112-8, 1996 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8709235

RESUMO

The cell fusion activity of most paramyxoviruses requires coexpression of a fusion protein (F) and a hemagglutinin-neuraminidase protein (HN) which are derived from the same virus type. To define the domain of the HN protein which interacts with the F protein in a type-specific manner a series of chimeric HN proteins between two different paramyxoviruses, Sendai virus (SN) and human parainfluenza virus type 3 (PI3), was constructed and coexpressed with the SN-F protein by using the vaccinia virus T7 RNA polymerase transient-expression system. Quantitative assays were used to evaluate cell surface expression as well as fusion-promoting activities of the chimeric HN molecules. A chimeric HN protein [SN(140)] containing 140 N-terminal amino acids derived from SN-HN and the remainder (432 amino acids) derived from PI3-HN was found to promote cell fusion with the SN-F protein. In contrast, a second chimeric HN with 137 amino acids from SN-HN at the N terminus could not promote fusion with SN-F, even though the protein was expressed on the cell surface. A construct in which the PI3-HN cytoplasmic tail and transmembrane domain were substituted for those of SN in the SN(140) chimera still maintained the ability to promote cell fusion. These results indicate that a region including only 82 amino acids in the extracellular domain, adjacent to the transmembrane domain of the SN-HN protein, is important for interaction with the SN-F protein and promotion of cell fusion.


Assuntos
Fusão Celular , Glicoproteínas/fisiologia , Proteína HN/fisiologia , Vírus da Parainfluenza 1 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Proteínas Virais de Fusão/fisiologia , Animais , Linhagem Celular , Membrana Celular/fisiologia , Membrana Celular/virologia , Chlorocebus aethiops , Expressão Gênica , Glicoproteínas/biossíntese , Glicoproteínas/isolamento & purificação , Proteína HN/biossíntese , Proteína HN/isolamento & purificação , Células HeLa , Humanos , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/isolamento & purificação
15.
J Virol ; 70(8): 5005-15, 1996 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-8764007

RESUMO

The human parainfluenza virus type 3 (HPIV3) fusion (F) and hemagglutinin-neuraminidase (HN) glycoproteins are the principal components involved in virion receptor binding, membrane penetration, and ultimately, syncytium formation. While the requirement for both F and HN in this process has been determined from recombinant expression studies, stable physical association of these proteins in coimmunoprecipitation studies has not been observed. In addition, coexpression of other heterologous paramyxovirus F or HN glycoproteins with either HPIV3 F or HN does not result in the formation of syncytia, suggesting serotype-specific protein differences. In this study, we report that simian virus 5 and Sendai virus heterologous HN proteins and measles virus hemagglutinin (H) were found to be down-regulated when coexpressed with HPIV3 F. As an alternative to detecting physical associations of these proteins by coimmunoprecipitation, further studies were performed with a mutant HPIV3 F protein (F-KDEL) lacking a transmembrane anchor and cytoplasmic tail and containing a carboxyl-terminal retention signal for the endoplasmic reticulum (ER). F-KDEL was defective for transport to the cell surface and could down-regulate surface expression of HPIV3 HN and heterologous HN/H proteins from simian virus 5, Sendai virus, and measles virus in coexpression experiments. HN/H down-regulation appeared to result, in part, from an early block to HPIV3 HN synthesis, as well as an instability of the heterologous HN/H proteins within the ER. In contrast, coexpression of F-KDEL with HPIV3 wild-type F or the heterologous receptor-binding proteins, respiratory syncytial virus glycoprotein (G) and vesicular stomatitis virus glycoprotein (G), were not affected in transport to the cell surface. Together, these results support the notion that the reported serotype-specific restriction of syncytium formation may involve, in part, down-regulation of heterologous HN expression.


Assuntos
Retículo Endoplasmático/metabolismo , Proteína HN/biossíntese , Respirovirus/metabolismo , Proteínas Virais/biossíntese , Regulação para Baixo , Retículo Endoplasmático/efeitos dos fármacos , Proteína HN/efeitos dos fármacos , Células HeLa , Humanos , Mutação , Plasmídeos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Virais/efeitos dos fármacos
16.
Biochem Biophys Res Commun ; 220(2): 459-66, 1996 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-8645327

RESUMO

Nectrisine, discovered as an immunomodulator, was found to inhibit alpha-glucosidase, alpha- and beta-mannosidases, beta-glucosidase and beta-N-acetylglucosaminidase, in that order of inhibition strength. Beta-Galactosidase, alpha-fucosidase, and neuraminidase were insensitive to this antibiotic. Also sensitive was the trimming glucosidase I which participates in the first step of modifying N-glycosidic oligosaccharide. Nectrisine demonstrated an inhibitory effect at the cellular level as strong as expected based on its action at enzyme levels; castanospermine and 1-deoxynojirimycin did not. Nectrisine and castanospermine suppressed syncytium formation and hemolytic activity in Newcastle disease virus (NDV)-infected BHK cells, without blocking the synthesis and cell-surface expression of HANA glycoprotein of NDV.


Assuntos
Inibidores Enzimáticos/farmacologia , Inibidores de Glicosídeo Hidrolases , Pirrolidinas/farmacologia , 1-Desoxinojirimicina/farmacologia , Acetilglucosaminidase/antagonistas & inibidores , Adjuvantes Imunológicos , Animais , Linhagem Celular , Cricetinae , Células Gigantes/efeitos dos fármacos , Glicoproteínas/metabolismo , Proteína HN/biossíntese , Imino Furanoses , Indolizinas/farmacologia , Manosidases/antagonistas & inibidores , Ratos , alfa-Manosidase , beta-Glucosidase/antagonistas & inibidores , beta-Manosidase
17.
Jpn J Med Sci Biol ; 49(1): 29-38, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8799805

RESUMO

We established two cell lines that stably express hemagglutinin-neuraminidase (HeLa-HN) and fusion proteins (HeLa-F) of a fusogenic strain of mumps virus. Infection of HeLa-F cells with a nonfusogenic strain resulted in induction of extensive cell fusion. On the other hand, HeLa-HN cells appeared resistant to cell fusion induced by mumps virus infection.


Assuntos
Vírus da Caxumba/metabolismo , Proteínas Virais de Fusão/biossíntese , Animais , Linhagem Celular , Chlorocebus aethiops , Citometria de Fluxo , Cobaias , Proteína HN/biossíntese , Células HeLa , Humanos , Transfecção , Células Vero
18.
J Virol ; 69(11): 6712-9, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7474081

RESUMO

To compare the requirements for paramyxovirus-mediated cell fusion, the fusion (F) and hemagglutinin-neuraminidase (HN) glycoproteins of simian virus 5 (SV5), human parainfluenza virus 3 (HPIV-3), and Newcastle disease virus (NDV) were expressed individually or coexpressed in either homologous or heterologous combinations in CV-1 or HeLa-T4 cells, using the vaccinia virus-T7 polymerase transient expression system. The contribution of individual glycoproteins in virus-induced membrane fusion was examined by using a quantitative assay for lipid mixing based on the relief of self-quenching (dequenching) of fluorescence of the lipid probe octadecyl rhodamine (R18) and a quantitative assay for content mixing based on the cytoplasmic activation of a reporter gene, beta-galactosidase. In these assays, expression of the individual F glycoproteins did not induce significant levels of cell fusion and no cell fusion was observed in experiments when cells individually expressing homologous F or HN proteins were mixed. However, coexpression of homologous F and HN glycoproteins resulted in extensive cell fusion. The kinetics of fusion were found to be very similar for all three paramyxoviruses studied. With NDV and HPIV-3, no cell fusion was detected when F proteins were coexpressed with heterologous HN proteins or influenza virus hemagglutinin (HA). In contrast, SV5 F protein exhibited a considerable degree of fusion activity when coexpressed with either NDV or HPIV-3 HN or with influenza virus HA, although the kinetics of fusion were two- to threefold higher when the homologous SV5 F and HN proteins were coexpressed. Thus, these data indicate that among the paramyxoviruses tested, SV5 has different requirements for cell fusion.


Assuntos
Proteína HN/metabolismo , Fusão de Membrana , Vírus da Doença de Newcastle/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Respirovirus/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Anticorpos Monoclonais , Fusão Celular , Linhagem Celular , Eritrócitos/fisiologia , Proteína HN/biossíntese , Células HeLa , Humanos , Cinética , Especificidade da Espécie , Transfecção , Vaccinia virus , Proteínas do Envelope Viral/biossíntese , beta-Galactosidase/biossíntese
19.
J Virol ; 69(11): 7045-53, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7474124

RESUMO

We have investigated the roles of the cytoplasmic domains of the human parainfluenza virus type 2 (PI2) and type 3 (PI3) fusion (F) proteins in protein transport and cell fusion activity. By using the vaccinia virus-T7 transient expression system, a series of F protein cytoplasmic tail truncation mutants was studied with respect to intracellular and surface expression and the ability to induce cell fusion when coexpressed with the corresponding hemagglutinin-neuraminidase (HN) proteins. All of the cytoplasmic tail truncation mutants of PI2F were expressed at high levels intracellularly or on cell surfaces as measured by immunoprecipitation and cell surface biotinylation assays. In addition, when coexpressed with PI2HN, these truncation mutants of PI2F were all found to be essentially unimpaired in the ability to induce cell fusion as measured by a quantitative cell fusion assay. In contrast, surface expression and cell fusion activity were found to be eliminated by a mutant of PI3F in which the entire cytoplasmic tail was deleted, and the mutant protein appeared to be unable to assemble into a high-molecular-weight oligomeric structure. To further investigate whether there is a specific sequence requirement in the cytoplasmic tail of PI3F, a chimeric protein consisting of the PI3F extracellular and transmembrane domains and the PI2F cytoplasmic tail was constructed. This chimeric protein was detected on the surface, and it was capable of inducing cell fusion when expressed together with PI3HN, although the fusogenic activity was reduced compared with that of wild-type PI3F. These results demonstrate that although PI2 and PI3 viruses belong to the same parainfluenza virus genus, these viruses show marked differences with respect to functional requirements for the cytoplasmic tail of the F glycoprotein.


Assuntos
Fusão Celular , Proteína HN/metabolismo , Vírus da Parainfluenza 2 Humana/fisiologia , Vírus da Parainfluenza 3 Humana/fisiologia , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Clonagem Molecular , Primers do DNA , Proteína HN/biossíntese , Proteína HN/química , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese , Reação em Cadeia da Polimerase , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Especificidade da Espécie , Transfecção , Proteínas Virais de Fusão/biossíntese , Proteínas Virais de Fusão/química
20.
Virology ; 209(2): 457-69, 1995 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-7778280

RESUMO

The promotion of membrane fusion by the paramyxovirus hemagglutinin-neuraminidase (HN) and fusion (F) proteins requires that they be derived from homologous viruses, suggesting the possibility that the promotion of fusion requires a virus-specific communication between the two glycoprotein spikes. We have evaluated the ability of chimeric HN proteins, composed of domains from the HN proteins of two heterologous members of the group, human parainfluenza virus 3 (hPIV3) and Newcastle disease virus (NDV), to complement the F protein of each virus in the promotion of fusion. Specificity for the F protein of hPIV3 segregates with a segment composed of the transmembrane anchor and the first 82 residues of the ectodomain of its HN protein. Specificity of NDV HN for its homologous F protein is determined by a similar domain. These findings suggest that determinants specific to this segment of the attachment protein spike may be involved in the triggering of the fusion process.


Assuntos
Fusão Celular , Proteína HN/metabolismo , Respirovirus/fisiologia , Proteínas Virais de Fusão/metabolismo , Animais , Sequência de Bases , Linhagem Celular , Cricetinae , Primers do DNA , Proteína HN/biossíntese , Rim , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos , Receptores Virais/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Mapeamento por Restrição , Especificidade por Substrato , Proteínas Virais de Fusão/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...