Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 90(4): 1964-72, 2016 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-26656681

RESUMO

UNLABELLED: Studies evaluating the immunogenicity of two pediatric tick-borne encephalitis virus (TBEV) vaccines have reported contradictory results. These vaccines are based on two different strains of the European TBEV subtype: FSME-Immun Junior is based on the Neudörfl (Nd) strain, whereas Encepur Children is based on the Karlsruhe (K23) strain. The antibody (Ab) response induced by these two vaccines might be influenced by antigenic differences in the envelope (E) protein, which is the major target of neutralizing antibodies. We used an established hybrid virus assay platform to compare the levels of induction of neutralizing antibodies against the two vaccine virus strains in children aged 1 to 11 years who received two immunizations with FSME-Immun Junior or Encepur Children. The influence of amino acid differences between the E proteins of the Nd and K23 vaccine strains was investigated by mutational analyses and three-dimensional computer modeling. FSME-Immun Junior induced 100% seropositivity and similar neutralizing antibody titers against hybrid viruses containing the TBEV E protein of the two vaccine strains. Encepur Children induced 100% seropositivity only against the hybrid virus containing the E protein of the homologous K23 vaccine strain. Antibody responses induced by Encepur Children to the hybrid virus containing the E protein of the heterologous Nd strain were substantially and significantly (P < 0.001) lower than those to the K23 vaccine strain hybrid virus. Structure-based mutational analyses of the TBEV E protein indicated that this is due to a mutation in the DI-DII hinge region of the K23 vaccine strain E protein which may have occurred during production of the vaccine seed virus and which is not present in any wild-type TBE viruses. IMPORTANCE: Our data suggest that there are major differences in the abilities of two European subtype pediatric TBEV vaccines to induce antibodies capable of neutralizing heterologous TBEV strains. This is a result of a mutation in the DI-DII hinge region of the E protein of the K23 vaccine virus strain used to manufacture Encepur Children which is not present in the Nd strain used to manufacture FSME-Immun Junior or in any other known naturally occurring TBEVs.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Vacinas Virais/imunologia , Criança , Pré-Escolar , Análise Mutacional de DNA , Estabilidade de Medicamentos , Feminino , Instabilidade Genômica , Humanos , Lactente , Masculino , Modelos Moleculares , Mutação de Sentido Incorreto , Conformação Proteica , Tecnologia Farmacêutica , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem
2.
J Virol ; 87(17): 9933-8, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23804648

RESUMO

The flavivirus fusion protein E contains a "stem" region which is hypothesized to be crucial for driving fusion. This sequence element connects the ectodomain to the membrane anchor, and its structure in the trimeric postfusion conformation is still poorly defined. Using E trimers of tick-borne encephalitis virus with stem truncations of different lengths, we show that the N-terminal part of the stem increases trimer stability and also modulates the trimer structure outside the stem interaction site.


Assuntos
Flavivirus/química , Proteínas Virais de Fusão/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Vírus da Encefalite Transmitidos por Carrapatos/química , Vírus da Encefalite Transmitidos por Carrapatos/genética , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia , Flavivirus/genética , Flavivirus/fisiologia , Modelos Moleculares , Dados de Sequência Molecular , Domínios e Motivos de Interação entre Proteínas , Multimerização Proteica , Estabilidade Proteica , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos , Proteínas Virais de Fusão/genética , Proteínas Virais de Fusão/fisiologia , Internalização do Vírus
3.
Vaccine ; 30(31): 4625-31, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22580355

RESUMO

BACKGROUND: Influenza viruses of subtype A/H9N2 are enzootic in poultry across Asia and the Middle East and are considered to have pandemic potential. The development of new vaccine manufacturing technologies is a cornerstone of influenza pandemic preparedness. METHODS: A non-adjuvanted whole-virus H9N2 vaccine was developed using Vero cell culture manufacturing technology. The induction of hemagglutination inhibition (HI) and virus-neutralizing antibodies was assessed in CD1 mice and guinea pigs. A highly sensitive enzyme-linked lectin assay was used to investigate the induction of antibodies capable of inhibiting the enzymatic activity of the H9N2 neuraminidase. Protective efficacy against virus replication in the lung after challenge with the homologous virus was evaluated in BALB/c mice by a TCID(50) assay, and prevention of virus replication in the lung and associated pathology were evaluated by histology and immunohistochemistry. To investigate the ability of the vaccine to prevent severe disease, BALB/c mice were challenged with a highly virulent mouse-adapted H9N2 isolate which was generated by multiple lung-to-lung passage of wild-type virus. RESULTS: The vaccine elicited high titers of functional H9N2-specific HA antibodies in both mice and guinea pigs, as determined by HI and virus neutralization assays. High titer H9N2-specific neuraminidase inhibiting (NAi) antibodies were also induced in both species. Vaccinated mice were protected from lung virus replication in a dose-dependent manner after challenge with the homologous H9N2 virus. Immunohistochemical analyses confirmed the lack of virus replication in the lung and an associated substantial reduction in lung pathology. Dose-dependent protection from severe weight loss was also provided after challenge with the highly virulent mouse-adapted H9N2 virus. CONCLUSIONS: The induction of high titers of H9N2-specific HI, virus-neutralizing and NAi antibodies and dose-dependent protection from virus replication and severe disease in animal models suggest that the Vero cell culture-derived whole-virus vaccine will provide an effective intervention in the event of a H9N2 pandemic situation.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H9N2/imunologia , Vacinas contra Influenza/imunologia , Neuraminidase/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Relação Dose-Resposta Imunológica , Feminino , Cobaias , Testes de Inibição da Hemaglutinação , Pulmão/patologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Infecções por Orthomyxoviridae/imunologia , Redução de Peso
4.
J Infect Dis ; 205(1): 28-34, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22090447

RESUMO

A Vero cell-derived whole-virus H5N1 influenza vaccine has been shown to induce neutralizing antibodies directed against the hemagglutinin (HA) protein of diverse H5N1 strains in animal studies and clinical trials. However, neuraminidase-inhibiting (NAi) antibodies can reduce viral spread and may be of particular importance in the event of an H5N1 pandemic, where immunity due to HA antibodies is likely absent in the general population. Here we demonstrate the effective induction of NAi antibody titers after H5N1 vaccination in humans. In contrast to the immune response directed toward HA, a single vaccine dose induced a strong NAi response that was not significantly boosted by a second dose, most probably due to priming by previous vaccination or infection with seasonal influenza viruses. After 2 immunizations, seroconversion rates based on antibody titers against HA and NA were similar, indicating the induction of equally strong immune responses against both proteins by this H5N1 vaccine.


Assuntos
Anticorpos Antivirais/sangue , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Neuraminidase/antagonistas & inibidores , Adolescente , Adulto , Animais , Chlorocebus aethiops , Humanos , Vacinas contra Influenza/administração & dosagem , Influenza Humana/sangue , Pessoa de Meia-Idade , Células Vero , Adulto Jovem
5.
J Virol ; 83(17): 8482-91, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19553320

RESUMO

The envelope protein E of flaviviruses mediates both receptor-binding and membrane fusion. At the virion surface, 180 copies of E are tightly packed and organized in a herringbone-like icosahedral structure, whereas in noninfectious subviral particles, 60 copies are arranged in a T=1 icosahedral symmetry. In both cases, the basic building block is an E dimer which exposes the binding sites for neutralizing antibodies at its surface. It was the objective of our study to assess the dependence of the antigenic structure of E on its quaternary arrangement, i.e., as part of virions, recombinant subviral particles, or soluble dimers. For this purpose, we used a panel of 11 E protein-specific neutralizing monoclonal antibodies, mapped to distinct epitopes in each of the three E protein domains, and studied their reactivity with the different soluble and particulate forms of tick-borne encephalitis virus E protein under nondenaturing immunoassay conditions. Significant differences in the reactivities with these forms were observed that could be related to (i) limited access of certain epitopes at the virion surface; (ii) limited occupancy of epitopes in virions due to steric hindrance between antibodies; (iii) differences in the avidity to soluble forms compared to the virion, presumably related to the flexibility of E at its domain junctions; and (iv) modulations of the external E protein surface through interactions with its stem-anchor structure. We have thus identified several important factors that influence the antigenicity of the flavivirus E protein and have an impact on the interaction with neutralizing antibodies.


Assuntos
Antígenos Virais/química , Antígenos Virais/imunologia , Vírus da Encefalite Transmitidos por Carrapatos/química , Vírus da Encefalite Transmitidos por Carrapatos/imunologia , Estrutura Quaternária de Proteína , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Afinidade de Anticorpos , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Modelos Biológicos , Modelos Moleculares , Testes de Neutralização/métodos , Ligação Proteica
6.
J Virol ; 80(19): 9557-68, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973559

RESUMO

A number of flaviviruses are important human pathogens, including yellow fever, dengue, West Nile, Japanese encephalitis, and tick-borne encephalitis (TBE) viruses. Infection with or immunization against any of these viruses induces a subset of antibodies that are broadly flavivirus cross-reactive but do not exhibit significant cross-neutralization. Nevertheless, these antibodies can efficiently bind to the major envelope protein (E), which is the main target of neutralizing and protective antibodies because of its receptor-binding and membrane fusion functions. The structural basis for this phenomenon is still unclear. In our studies with TBE virus, we have provided evidence that such cross-reactive antibodies are specific for a cluster of epitopes that are partially occluded in the cage-like assembly of E proteins at the surfaces of infectious virions and involve-but are not restricted to-amino acids of the highly conserved internal fusion peptide loop. Virus disintegration leads to increased accessibility of these epitopes, allowing the cross-reactive antibodies to bind with strongly increased avidity. The cryptic properties of these sites in the context of infectious virions can thus provide an explanation for the observed lack of efficient neutralizing activity of broadly cross-reactive antibodies, despite their specificity for a functionally important structural element in the E protein.


Assuntos
Antígenos Virais/imunologia , Flavivirus/imunologia , Afinidade de Anticorpos/imunologia , Sítios de Ligação , Linhagem Celular , Reações Cruzadas/imunologia , Ensaio de Imunoadsorção Enzimática , Flavivirus/química , Modelos Moleculares , Conformação Molecular , Mapeamento de Peptídeos , Filogenia , Titulometria , Vírion/química , Vírion/imunologia
7.
Novartis Found Symp ; 277: 57-65; discussion 65-73, 251-3, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17319154

RESUMO

The envelope proteins (E) of flaviviruses form an icosahedral cage-like structure of homodimers that cover completely the surface of mature virions and are responsible for receptor-binding and membrane fusion. Fusion is triggered by the acidic pH in endosomes which induces dramatic conformational changes of E that drive the merger of the membranes. We have identified an alternative trigger that induces the first phase of the fusion process only, but then leads to an arrest at an intermediate stage. These data suggest that the early and late stages of flavivirus fusion are differentially controlled by intersubunit and intrasubunit constraints of the fusion protein, respectively. Details of the molecular antigenic structure of the flavivirus E protein were revealed by the use of neutralization escape mutants as well as recombinant expression systems for the generation of virus-like particles. The experimental data provide evidence that each of the three domains contributing to the external face of the E protein can induce and bind neutralizing antibodies. Broadly flavivirus cross-reactive antibodies, however, primarily recognize a site involving residues of the highly conserved fusion peptide loop which is cryptic and largely inaccessible on the surface of native infectious virions.


Assuntos
Antígenos Virais/imunologia , Flavivirus/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Infecções por Flavivirus/imunologia , Humanos , Proteínas do Envelope Viral/química , Vírion/química , Vírion/imunologia
8.
J Virol ; 78(15): 8078-84, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15254179

RESUMO

Flaviviruses have a spherical capsid that is composed of multiple copies of a single capsid protein and, in contrast to the viral envelope, apparently does not have an icosahedral structure. So far, attempts to isolate distinct particulate capsids and soluble forms of the capsid protein from purified virions as well as to assemble capsid-like particles in vitro have been largely unsuccessful. Here we describe the isolation of nucleocapsids from tick-borne encephalitis (TBE) virus and their disintegration into a capsid protein dimer by high-salt treatment. Purified capsid protein dimers could be assembled in vitro into capsid-like particles when combined with in vitro transcribed viral RNA. Particulate structures could also be obtained when single-stranded DNA oligonucleotides were used. These data suggest that the dimeric capsid protein functions as a basic building block in the assembly process of flaviviruses.


Assuntos
Proteínas do Capsídeo/isolamento & purificação , Vírus da Encefalite Transmitidos por Carrapatos/química , Montagem de Vírus , Proteínas do Capsídeo/química , Dimerização , Vírus da Encefalite Transmitidos por Carrapatos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA