Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Virol ; 81(23): 12859-71, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17898060

RESUMO

Measles virus (MeV) infection is characterized by the formation of multinuclear giant cells (MGC). We report that beta interferon (IFN-beta) production is amplified in vitro by the formation of virus-induced MGC derived from human epithelial cells or mature conventional dendritic cells. Both fusion and IFN-beta response amplification were inhibited in a dose-dependent way by a fusion-inhibitory peptide after MeV infection of epithelial cells. This effect was observed at both low and high multiplicities of infection. While in the absence of virus replication, the cell-cell fusion mediated by MeV H/F glycoproteins did not activate any IFN-alpha/beta production, an amplified IFN-beta response was observed when H/F-induced MGC were infected with a nonfusogenic recombinant chimerical virus. Time lapse microscopy studies revealed that MeV-infected MGC from epithelial cells have a highly dynamic behavior and an unexpected long life span. Following cell-cell fusion, both of the RIG-I and IFN-beta gene deficiencies were trans complemented to induce IFN-beta production. Production of IFN-beta and IFN-alpha was also observed in MeV-infected immature dendritic cells (iDC) and mature dendritic cells (mDC). In contrast to iDC, MeV infection of mDC induced MGC, which produced enhanced amounts of IFN-alpha/beta. The amplification of IFN-beta production was associated with a sustained nuclear localization of IFN regulatory factor 3 (IRF-3) in MeV-induced MGC derived from both epithelial cells and mDC, while the IRF-7 up-regulation was poorly sensitive to the fusion process. Therefore, MeV-induced cell-cell fusion amplifies IFN-alpha/beta production in infected cells, and this indicates that MGC contribute to the antiviral immune response.


Assuntos
Células Dendríticas/virologia , Células Epiteliais/virologia , Células Gigantes/virologia , Interferon Tipo I/biossíntese , Vírus do Sarampo/imunologia , Vírus do Sarampo/fisiologia , Animais , Fusão Celular , Linhagem Celular , Núcleo Celular/química , Chlorocebus aethiops , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Células Gigantes/citologia , Células Gigantes/imunologia , Humanos , Fator Regulador 3 de Interferon/análise , Fator Regulador 7 de Interferon/análise , Vírus do Sarampo/genética , Microscopia de Vídeo , Proteínas Virais de Fusão/imunologia , Proteínas Virais de Fusão/fisiologia , Proteínas Virais/imunologia , Proteínas Virais/fisiologia
2.
J Virol ; 80(4): 1972-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16439553

RESUMO

Nipah virus (NiV), a member of the Paramyxoviridae family, causes a zoonotic infection in which the reservoir, the fruit bat, may pass the infection to pigs and eventually to humans. In humans, the infection leads to encephalitis with >40 to 70% mortality. We have previously shown that polyclonal antibody directed to either one of two glycoproteins, G (attachment protein) or F (fusion protein), can protect hamsters from a lethal infection. In the present study, we have developed monoclonal antibodies (MAbs) to both glycoproteins and assessed their ability to protect animals against lethal NiV infection. We show that as little as 1.2 mug of an anti-G MAb protected animals, whereas more than 1.8 mug of anti-F MAb was required to completely protect the hamsters. High levels of either anti-G or anti-F MAbs gave a sterilizing immunity, whereas lower levels could protect against a fatal infection but resulted in an increase in anti-NiV antibodies starting 18 days after the viral challenge. Using reverse transcriptase PCR, the presence of NiV in the different organs could not be observed in MAb-protected animals. When the MAbs were given after infection, partial protection (50%) was observed with the anti-G MAbs when the animals were inoculated up to 24 h after infection, but administration of the anti-F MAbs protected some animals (25 to 50%) inoculated later during the infection. Our studies suggest that immunotherapy could be used for people who are exposed to NiV infections.


Assuntos
Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/uso terapêutico , Infecções por Henipavirus/tratamento farmacológico , Infecções por Henipavirus/prevenção & controle , Imunização Passiva , Vírus Nipah/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/sangue , Cricetinae , Ensaio de Imunoadsorção Enzimática , Feminino , Mesocricetus , Camundongos , Testes de Neutralização , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/imunologia
3.
J Virol ; 78(2): 834-40, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14694115

RESUMO

Nipah virus, a member of the paramyxovirus family, was first isolated and identified in 1999 when the virus crossed the species barrier from fruit bats to pigs and then infected humans, inducing an encephalitis with up to 40% mortality. At present there is no prophylaxis for Nipah virus. We investigated the possibility of vaccination and passive transfer of antibodies as interventions against this disease. We show that both of the Nipah virus glycoproteins (G and F) when expressed as vaccinia virus recombinants induced an immune response in hamsters which protected against a lethal challenge by Nipah virus. Similarly, passive transfer of antibody induced by either of the glycoproteins protected the animals. In both the active and passive immunization studies, however, the challenge virus was capable of hyperimmunizing the vaccinated animals, suggesting that although the virus replicates under these conditions, the immune system can eventually control the infection.


Assuntos
Anticorpos Antivirais/imunologia , Infecções por Henipavirus/prevenção & controle , Imunização Passiva/métodos , Vírus Nipah/imunologia , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Cricetinae , Modelos Animais de Doenças , Células HeLa , Infecções por Henipavirus/imunologia , Humanos , Mesocricetus , Vacinação/métodos , Vaccinia virus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Vacinas Virais/imunologia
4.
J Med Virol ; 72(2): 290-8, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14695672

RESUMO

The availability of new generation serological assays allowed re-evaluation of the antibody response to measles virus. IgM, IgA, total IgG, and IgG subclass responses were studied to the three major immunogenic measles virus proteins: the fusion protein (F), haemagglutinin (H), and nucleoprotein (N). Plasma samples were obtained from clinically diagnosed measles cases (n = 146) in Khartoum (Sudan) within a week after onset of the rash. Convalescent phase samples were collected from 32 of 117 laboratory-confirmed measles cases at different time points after onset of rash. Glycoprotein-specific IgM, IgG, and IgA antibody levels correlated well to the N-specific response. For IgG and IgA, responses to F were higher than to H. IgA antibody levels were undetectable in about one third of the laboratory-confirmed cases during the acute phase, but positive in all patients tested 1-4 weeks after infection. IgM levels declined rapidly and were lost 3-6 months after infection. IgA levels declined slowly during the first year but did not return to background levels during the subsequent 2 years. IgG avidity maturation was detected during a 3-6 month period after infection. The predominant IgG subclasses during the acute phase were IgG(1) and IgG(3). The latter was lost in the convalescent phase, while the IgG(4) isotype showed a slight rise afterwards. Interestingly, acute phase IgG(3) and IgA responses were associated, and were only detected in samples with high IgG. This study provides a comprehensive perspective on the antibody response to wild-type measles virus infection.


Assuntos
Anticorpos Antivirais/sangue , Especificidade de Anticorpos , Isotipos de Imunoglobulinas/sangue , Vírus do Sarampo/imunologia , Sarampo/imunologia , Proteínas Virais/imunologia , Doença Aguda , Hemaglutininas Virais/imunologia , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Lactente , Sarampo/virologia , Proteínas do Nucleocapsídeo , Nucleoproteínas/imunologia , Proteínas do Core Viral/imunologia , Proteínas Virais de Fusão/imunologia
5.
J Med Virol ; 68(3): 433-40, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12226833

RESUMO

Africa remains one of the major reservoirs of measles infection. Molecular epidemiological studies have permitted different measles virus isolates to be grouped into clades and genotypes; the major group, which has been identified as indigenous to Africa, is clade B. The viruses from epidemics in the Gambia (1993) and in the Cameroon (2001) were examined. In both studies, the homogeneity of the virus isolates within the epidemic as shown by sequence analysis revealed less than 0.2% variation of nucleotides between isolates. The measles viruses isolated in 1983 in Yaoundé, Cameroon, were designated as the B1 genotype. However, in 2001 only viruses belonging to the B3 genotype were found in this city. The viruses in the Gambia (1993) were also of the B3 genotype. However, these viruses could be distinguished from each other at the antigenic level and by comparative sequence analysis. The B3 Cameroon (2001) viruses were related to the proposed B3.1 subgroup, whereas the Gambian (1993) isolates corresponded to the B3.2 subgroup. The geographical distribution for the period 1993-2001 of these two viruses shows that B3.1 is found from the Sudan to Nigeria and Ghana extending south to the Cameroon, whereas the B3.2 genotype is found in West Africa. In Nigeria and Ghana, the viruses co-circulate. The identification of these viruses will permit more meaningful epidemiological studies after the proposed increase in measles vaccination coverage.


Assuntos
Vírus do Sarampo/classificação , Vírus do Sarampo/genética , Sarampo/epidemiologia , África Ocidental/epidemiologia , Anticorpos Antivirais/sangue , Camarões/epidemiologia , Criança , Pré-Escolar , Citometria de Fluxo , Gâmbia/epidemiologia , Genótipo , Hemaglutininas , Hemaglutininas Virais/genética , Humanos , Lactente , Recém-Nascido , Sarampo/virologia , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo , Nucleoproteínas/genética , Filogenia , Análise de Sequência de DNA , Proteínas do Core Viral/genética
6.
Virology ; 268(1): 122-31, 2000 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10683334

RESUMO

The limited induction of Th1 and cytotoxic immune responses is regarded as the main reason for the increased susceptibility to intracellular microorganisms in early life. Recently, in vitro IL-12 supplementation was shown to enhance the limited IFN-gamma release of measles-specific infant T cells. Using a series of IL-12 delivery systems, we show here that in vivo IL-12 supplementation may enhance early life murine Th1 responses to two model vaccine antigens, measles virus hemagglutinin and tetanus toxin peptide. However, this required multiple repeat injections of recombinant rIL-12, which were poorly tolerated in young mice. Local IL-12 delivery by an IL-12 expressing canarypox vector proved safe but failed to modulate vaccine responses. An IL-12 DNA plasmid or a CD40L DNA plasmid efficiently enhanced neonatal Th1 responses to measles hemagglutinin DNA vaccine. However, both plasmids only enhanced Th1 responses to DNA and not to peptide, protein, or live viral vaccines. Thus, inducing adult-like Th1 responses may be achieved in vivo by inducing (CD40L) or substituting for (IL-12 supplementation) optimal activation of neonatal APC. However, these immunomodulatory effects appear limited to certain antigen-presentation approaches and may not be broadly applicable to vaccines.


Assuntos
Adjuvantes Imunológicos , Interleucina-12/imunologia , Vacina contra Sarampo/imunologia , Toxoide Tetânico/imunologia , Células Th1/imunologia , Envelhecimento/imunologia , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antivirais/sangue , Antígenos de Bactérias/imunologia , Antígenos CD40/imunologia , Ligante de CD40 , Hemaglutininas Virais/imunologia , Imunização , Interleucina-12/genética , Linfocinas/análise , Vírus do Sarampo/imunologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Proteínas Recombinantes/imunologia
7.
J Virol ; 73(1): 787-90, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9847389

RESUMO

Although measles virus is an antigenically monotypic virus, nucleotide sequence analysis of the hemagglutinin and nucleoprotein genes has permitted the differentiation of a number of genotypes. In contrast, the fusion (F) protein is highly conserved; only three amino acid changes have been reported over a 40-year period. We have isolated a measles virus strain which did not react with an anti-F monoclonal antibody (MAb) which we had previously shown to be directed against a dominant antigenic site. This virus strain, Lys-1, had seven amino acid changes compared with the Edmonston strain. We have shown that a single amino acid at position 73 is responsible for its nonreactivity with the anti-F MAb. With the same MAb, antibody-resistant mutants were prepared from the vaccine strain. A single amino acid change at position 73 (R-->W) was observed. The possibility of selecting measles virus variants in vaccinated populations is discussed.


Assuntos
Anticorpos Monoclonais/imunologia , Antígenos Virais/imunologia , Vírus do Sarampo/imunologia , Mutação , Proteínas Virais de Fusão/imunologia , Humanos , Vacina contra Sarampo/imunologia , Vírus do Sarampo/genética , Relação Estrutura-Atividade , Proteínas Virais de Fusão/química
8.
J Virol ; 72(11): 8472-6, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9765383

RESUMO

We have studied the immune responses to the two glycoproteins of the Morbillivirus canine distemper virus (CDV) after DNA vaccination of BALB/c mice. The plasmids coding for both CDV hemagglutinin (H) and fusion protein (F) induce high levels of antibodies which persist for more than 6 months. Intramuscular inoculation of the CDV DNA induces a predominantly immunoglobulin G2a (IgG2a) response (Th1 response), whereas gene gun immunization with CDV H evokes exclusively an IgG1 response (Th2 response). In contrast, the CDV F gene elicited a mixed, IgG1 and IgG2a response. Mice vaccinated (by gene gun) with either the CDV H or F DNA showed a class I-restricted cytotoxic lymphocyte response. Immunized mice challenged intracerebrally with a lethal dose of a neurovirulent strain of CDV were protected. However, approximately 30% of the mice vaccinated with the CDV F DNA became obese in the first 2 months following the challenge. This was not correlated with the serum antibody levels.


Assuntos
Vírus da Cinomose Canina/genética , Vírus da Cinomose Canina/imunologia , Cinomose/prevenção & controle , Vacinas de DNA/farmacologia , Vacinas Virais/farmacologia , Animais , Anticorpos Antivirais/biossíntese , Cinomose/imunologia , Cães , Feminino , Glicoproteínas/genética , Glicoproteínas/imunologia , Células HeLa , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Humanos , Imunidade Celular , Células L , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Citotóxicos/imunologia , Transfecção , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
9.
J Gen Virol ; 78 ( Pt 1): 107-11, 1997 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9010292

RESUMO

Measles virus (MV) infections are characterized by the induction of syncytia, i.e. the fusion of infected cells. Two MV proteins, the haemagglutinin (HA) and fusion (F) proteins, are involved in this process. Synthetic peptides representing two alpha-helical regions of the MV F protein were studied for their ability to inhibit MV fusion. A peptide corresponding to the leucine zipper region (amino acids 455-490) inhibited MV fusion, whereas a peptide to amino acids 148-177, corresponding to the amphipathic alpha-helix region, did not. Fusion inhibition was also obtained with vaccinia virus-expressed HA and F, a recent wild-type MV isolate and the closely related canine distemper virus, but not with mumps virus. The F455-490 peptide did not affect the synthesis of MV F or its transport to the cell membrane. Virus-cell attachment was unaffected, but haemolysis and virus entry into the cell were inhibited. In one-step growth curves the virus yield was unaffected.


Assuntos
Fusão Celular , Zíper de Leucina , Vírus do Sarampo/fisiologia , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Chlorocebus aethiops , Vírus da Cinomose Canina/fisiologia , Cães , Células Gigantes , Células HeLa , Hemaglutinação , Hemólise , Humanos , Vírus do Sarampo/patogenicidade , Dados de Sequência Molecular , Vírus da Caxumba/fisiologia , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos , Células Vero
10.
J Gen Virol ; 77 ( Pt 11): 2695-9, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8922462

RESUMO

To unravel the intracellular fate of measles virus (MV) haemagglutinin (H) following fusion of the virus envelope with the cell membrane, its presentation by MHC molecules to T cells was explored. After MV infection, murine cells expressing CD46 were lysed by MHC class I-restricted CD8 CTLs specific for the ectodomain of H. In contrast, when sensitized with UV-inactivated MV, they were not lysed by these effectors, but were recognized by H-specific and class II-restricted CD4 CTLs. Thus, after MV binding and fusion, H becomes associated with plasma membrane and its ectodomain can reach the endosomal MHC-II but not the cytosolic MHC-I antigen presentation pathway. From these data and a reappraisal of previous reports, it appears that the ectodomains of both MV haemagglutinin fusion proteins, having undergone the fusion step, are not translocated into the cytosol and end up in the endosomes.


Assuntos
Apresentação de Antígeno , Hemaglutininas Virais/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Vírus do Sarampo/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Antígenos CD/análise , Citosol/virologia , Vírus do Sarampo/imunologia , Proteína Cofatora de Membrana , Fusão de Membrana , Glicoproteínas de Membrana/análise , Camundongos , Linfócitos T Citotóxicos/imunologia
11.
Virology ; 225(2): 293-9, 1996 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-8918915

RESUMO

We have evaluated the DNA vaccination strategy for measles virus (MV) hemagglutinin (HA) and nucleoprotein (NP) genes. Plasmids encoding either the MV, HA, or NP proteins inoculated intramuscularly into Balb/c mice induced both humoral and CTL class I restricted responses. Antibody responses were not increased by multiple inoculations. The major antibody isotype induced by both the HA and NP was IgG2a consistent with a Th1 response. In contrast, immunization with a plasmid which directed the synthesis of a partially secreted form of HA gave mainly IgG1 antibodies. When the amount of DNA was reduced for the HA plasmid (1 or 10 microg/animal), although the antibody was not induced, a CTL response was observed.


Assuntos
Anticorpos Antivirais/imunologia , Citotoxicidade Imunológica , Hemaglutininas Virais/genética , Vírus do Sarampo/genética , Nucleoproteínas/genética , Células Th1/imunologia , Proteínas Virais/genética , Vacinas Virais , Animais , Linhagem Celular , DNA Viral/genética , DNA Viral/imunologia , Feminino , Hemaglutininas Virais/imunologia , Humanos , Ativação Linfocitária , Vírus do Sarampo/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas do Nucleocapsídeo , Nucleoproteínas/imunologia , Plasmídeos/genética , Plasmídeos/imunologia , Células Th1/virologia , Vacinas Sintéticas , Proteínas Virais/imunologia
12.
Virology ; 219(1): 133-9, 1996 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-8623522

RESUMO

In Balb/c (H-2d) mice, the nucleoprotein (NP) of measles virus (MV) induces a MHC class I restricted-CTL response to a single 9-amino-acid epitope (aa 281--289). This L(d)-restricted epitope is also present in the NP of the closely related canine distemper virus (CDV). To investigate whether this epitope is immunologically effective when it is present within the primary sequence of a nonviral protein, we have incorporated the 281--289 motif into the human CD36 protein. When cells are infected with vaccinia virus (VV) recombinants expressing this protein, CD36NP, the MV epitope is correctly processed and the cells are lysed by MVNP-specific CTLs. In vivo, VV-CD36NP induced CTLs which protected mice from a lethal dose of CDV, but did not block virus replication. The MVNP contains four other potential L(d)-restricted motifs. To investigate if these could be utilized in the absence of the dominant epitope, a mutant NP was produced in which one of the anchor residues in the aa 281--289 motif was mutated. Cells infected with a VV recombinant expressing this protein (VV-NP F289S) were only poorly lysed by MVNP-specific CTLs. Similarly, immunization of Balb/c mice with VV-NP F289S induced a lower level of CTL activity compared to the VV-NP, but the activity was now directed to three other epitopes. When mice were vaccinated with VV-NP F289S they were only partially protected from a lethal CDV challenge. The significance of these results for MV vaccine development is discussed.


Assuntos
Epitopos/imunologia , Vírus do Sarampo/imunologia , Nucleoproteínas/imunologia , Linfócitos T Citotóxicos/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Apresentação de Antígeno/imunologia , Sequência de Bases , Sítios de Ligação , Antígenos CD36/genética , Antígenos CD36/imunologia , Chlorocebus aethiops , DNA Viral , Cinomose/imunologia , Cinomose/prevenção & controle , Vírus da Cinomose Canina/imunologia , Cães , Epitopos/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo , Nucleoproteínas/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Células Tumorais Cultivadas , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Vaccinia virus/genética , Células Vero , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/genética , Vacinas Virais/imunologia
13.
Virology ; 212(1): 255-8, 1995 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-7676641

RESUMO

To induce an MHC-restricted specific CTL or Th response, an antigen must be delivered into the appropriate cellular compartment. We explored the role of CD46 in the presentation of measles virus (MV) nucleoprotein (NP) to murine NP-specific and MHC Class I-restricted polyclonal CTLs and the effect of inactivating MV by uv or formaldehyde. CD46(-)- and CD46(+)-transfected murine cells were used as target cells. After MV infection, only the targets which expressed CD46 were lysed by NP-specific class I-restricted CTLs. When MV was uv-inactivated, NP presentation by MHC class I molecules was retained but could be blocked by fusion inhibitors which block virus cell entry. When MV was inactivated with formaldehyde, NP was no longer presented by MHC class I molecules, although it was still presented by MHC class II molecules to a NP-specific class II-restricted T cell hybridoma. These data show that MV binding to the CD46 molecule is a prerequisite for virus-to-cell fusion and that cytosolic delivery of NP is necessary for presentation by class I molecules. Moreover, formaldehyde inactivation of virus induces the loss of class I-restricted presentation of NP due to selective abrogation of fusion and cytosolic delivery of NP.


Assuntos
Antígenos Virais/imunologia , Antígenos H-2/imunologia , Vírus do Sarampo/química , Nucleoproteínas/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD/imunologia , Antígenos Virais/química , Formaldeído/química , Vírus do Sarampo/imunologia , Proteína Cofatora de Membrana , Glicoproteínas de Membrana/imunologia , Camundongos , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo , Oligopeptídeos/química , Oligopeptídeos/imunologia
14.
Vet Microbiol ; 44(2-4): 267-70, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-8588321

RESUMO

The morbilliviruses have a restricted host range. This is probably dependent on the use of specific host cell receptors. In the present article, we have reviewed our approach to identify a host cell receptor for one of the morbilliviruses, measles virus and to elucidate the interaction between viral and cellular proteins during virus entry into the host cell.


Assuntos
Antígenos CD/metabolismo , Hemaglutininas Virais/metabolismo , Vírus do Sarampo/patogenicidade , Glicoproteínas de Membrana/metabolismo , Animais , Linhagem Celular , Haplorrinos , Células HeLa , Humanos , Vírus do Sarampo/metabolismo , Proteína Cofatora de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Virais de Fusão/metabolismo
15.
Vaccine ; 13(2): 197-201, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7625116

RESUMO

We have used vaccinia-measles recombinant viruses to study vaccination in the presence of pre-existing antibody. When mice were vaccinated with recombinants expressing either the haemagglutinin (H) or fusion (F) measles virus (MV) proteins, the humoral response to the MV protein was suppressed by passively administered polyclonal antibody. However, individual monoclonal antibodies (H or F) did not affect the response. Mice whose anti-MV antibody response to H or F was initially suppressed by passive administration of anti-MV antibody were revaccinated 120 days later and gave a normal humoral response to the MV proteins. The VV-H recombinant induces a strong class I CTL response in Balb/c mice. This was not affected by the presence of levels of anti-MV antibody which inhibited the humoral response.


Assuntos
Anticorpos Antivirais/biossíntese , Vacina contra Sarampo/farmacologia , Vírus do Sarampo/imunologia , Vaccinia virus/imunologia , Animais , Anticorpos Antivirais/farmacologia , Especificidade de Anticorpos , Feminino , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Imunização Passiva , Vírus do Sarampo/genética , Camundongos , Camundongos Endogâmicos BALB C , Recombinação Genética , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia , Vacinas Sintéticas/farmacologia , Vaccinia virus/genética , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia
17.
J Virol ; 68(11): 7546-8, 1994 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7933140

RESUMO

Measles virus (MV) fusion requires the participation of both the fusion (F) and hemagglutinin (H) glycoproteins. The canine distemper virus fusion protein (CDVF) cannot substitute for the measles virus fusion protein (MVF) in this process. Introduction of restriction enzyme sites into the cDNAs of CDVF and MVF by site-directed mutagenesis facilitated the production of chimeric F proteins which were tested for their capacity to give fusion when coexpressed with MVH. Fusion resulted when the amino-terminal half of the MVF cysteine-rich region was transferred to CDVF.


Assuntos
Glicoproteínas/fisiologia , Vírus do Sarampo/fisiologia , Proteínas Virais de Fusão/fisiologia , Sequência de Aminoácidos , Cisteína/análise , Vírus da Cinomose Canina/fisiologia , Glicoproteínas/química , Células HeLa , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Relação Estrutura-Atividade
18.
J Gen Virol ; 74 ( Pt 11): 2365-72, 1993 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8245852

RESUMO

We have investigated the structure and interaction of the measles virus (MV) glycoproteins expressed at the cell membrane. Cross-linking studies with a variety of chemicals stabilized dimeric forms of the haemagglutinin (HA) or fusion (F) proteins, although by sucrose density gradient analysis, oligomers corresponding to tetramers and larger were observed for both proteins. In cells in which both HA and F were expressed at the surface, their close association was shown by cross-linking and co-immunoprecipitation.


Assuntos
Glicoproteínas/química , Hemaglutininas Virais/química , Vírus do Sarampo/química , Proteínas Virais de Fusão/química , Proteínas Virais/química , Centrifugação com Gradiente de Concentração , Precipitação Química , Reagentes de Ligações Cruzadas , Células HeLa , Humanos , Testes de Precipitina , Proteínas Recombinantes/química
19.
J Gen Virol ; 74 ( Pt 6): 1073-9, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8509760

RESUMO

A surface glycoprotein (gp57/67) was previously shown to be involved in measles virus (MV) binding and characterized in our laboratory. Here, we described down-regulation of cell surface gp57/67 after infection with MV. This effect is specific for MV since cells infected with canine distemper virus, closely related to MV, did not down-regulate gp57/67. The decrease in cell surface gp57/67 correlated with expression of MV glycoproteins and more particularly with the expression of MV haemagglutinin (MV-H). Indeed, expression of MV-H after infection with a vaccinia virus recombinant coding for MV-H was necessary and sufficient to induce down-regulation of gp57/67. Kinetics of cell surface expression of MV-H and gp57/67 showed that the degree of down-regulation was correlated with the amount of MV-H expressed by infected cells. Experiments using antibody-prelabelled gp57/67 and indirect immunofluorescence microscopy allowed us to follow the fate of gp57/67 and showed that down-regulation was occurring by rapid internalization of gp57/67 from the cell surface. These results provide additional evidence that the gp57/67 molecule is closely associated with the pathway of MV infection and also reveal a phenomenon which may be related to viral pathogenesis and persistence.


Assuntos
Regulação para Baixo , Hemaglutininas Virais/metabolismo , Vírus do Sarampo/crescimento & desenvolvimento , Glicoproteínas de Membrana/biossíntese , Receptores Virais/biossíntese , Transporte Biológico , Linhagem Celular , Células Epiteliais , Epitélio/microbiologia , Hemaglutininas Virais/genética , Humanos , Proteínas Recombinantes/metabolismo , Linfócitos T/citologia , Linfócitos T/microbiologia , Vaccinia virus/genética
20.
J Gen Virol ; 73 ( Pt 2): 359-67, 1992 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1538193

RESUMO

Measles virus genes encoding the haemagglutinin (HA), fusion protein (F) or nucleoprotein (NP) have been inserted into the vaccinia virus genome either alone or in various combinations. In each case the measles virus genes were expressed from the 7.5K promoter and were incorporated into the thymidine kinase (tk) or K1L loci of the Copenhagen strain of vaccinia virus. Cells infected by the recombinants synthesized measles virus proteins indistinguishable from those induced in measles virus-infected cells. However, in some instances the level of expression in cells infected by recombinants expressing more than one measles virus gene was reduced when compared to those encoding a single gene. The sera from mice immunized with recombinants containing either HA, HA.F, HA.NP or HA.F.NP had similar levels of measles virus neutralizing antibodies which remained constant throughout a 7 month period. Analysis of these sera by immunoprecipitation of radiolabelled measles virus confirmed the presence of specific antibody to each of the antigens where appropriate. The introduction of the measles virus genes into the K1L and the tk sites despite attenuating the virus for mice by 10-fold and 1000-fold respectively did not affect the vaccination efficiency, i.e. ability to induce measles virus antibody and protect mice. Vaccination of BALB/c (H2d) mice with HA and F, but not NP, recombinants completely protected the animals against a lethal measles virus challenge. In contrast, although the HA recombinant protected CBA (H2k) mice, the F recombinant did so poorly. However, by immunizing CBA mice with a recombinant expressing both F and NP, protection was increased to more than 75%. Our findings demonstrate the ability of three measles virus antigens expressed from the vaccinia virus genome alone or in combination to contribute to protective immunity against measles virus infection of mice. They also suggest that the association of measles virus antigens in a single recombinant DNA vaccine could be beneficial to overcome host-related restriction of the immune response to particular antigens.


Assuntos
Vacina contra Sarampo , Vírus do Sarampo/genética , Sarampo/prevenção & controle , Nucleoproteínas , Proteínas Virais/genética , Animais , Anticorpos Antivirais/biossíntese , Sequência de Bases , Linhagem Celular , Feminino , Regulação Viral da Expressão Gênica , Hemaglutininas Virais/genética , Hemaglutininas Virais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo , Testes de Precipitina , Vacinação , Vacinas Sintéticas , Vaccinia virus/genética , Proteínas do Core Viral/genética , Proteínas do Core Viral/imunologia , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/imunologia , Proteínas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA