Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccine ; 27(22): 2930-9, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19428903

RESUMO

Recombinant vesicular stomatitis viruses (rVSVs) are being developed as potential HIV-1 vaccine candidates. To characterize the in vivo replication and dissemination of rVSV vectors in mice, high doses of a highly attenuated vector expressing HIV-1 Gag, rVSV(IN)-N4CT9-Gag1, and a prototypic reference virus, rVSV(IN)-HIVGag5, were delivered intramuscularly (IM), intranasally (IN), or intravenously (IV). We used quantitative, real-time RT-PCR (Q-PCR) and standard plaque assays to measure the temporal dissemination of these viruses to various tissues. Following IM inoculation, both viruses were detected primarily at the injection site as well as in draining lymph nodes; neither virus induced significant weight loss, pathologic signs, or evidence of neuroinvasion. In contrast, following IN inoculation, the prototypic virus was detected in all tissues tested and caused significant weight loss leading to death. IN administration of rVSV(IN)-N4CT9-Gag1 resulted in detection in numerous tissues (brain, lung, nasal turbinates, and lymph nodes) albeit in significantly reduced levels, which caused little or no weight loss nor any mortality. Following IV inoculation, both prototypic and attenuated viruses were detected by Q-PCR in all tissues tested. In contrast to the prototype, rVSV(IN)-N4CT9-Gag1 viral loads were significantly lower in all organs tested, and no infectious virus was detected in the brain following IV inoculation, despite the presence of viral RNA. These studies demonstrated significant differences in the biodistribution patterns of and the associated pathogenicity engendered by the prototypic and attenuated vectors in a highly susceptible host.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Vacinas contra a AIDS/farmacocinética , Vetores Genéticos , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Vacinas contra a AIDS/efeitos adversos , Administração Intranasal , Animais , Feminino , Injeções Intramusculares , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos BALB C , RNA Viral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/farmacocinética , Ensaio de Placa Viral
2.
J Virol ; 82(1): 207-19, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17942549

RESUMO

Recombinant vesicular stomatitis virus (rVSV) has shown great potential as a new viral vector for vaccination. However, the prototypic rVSV vector described previously was found to be insufficiently attenuated for clinical evaluation when assessed for neurovirulence in nonhuman primates. Here, we describe the attenuation, neurovirulence, and immunogenicity of rVSV vectors expressing human immunodeficiency virus type 1 Gag. These rVSV vectors were attenuated by combinations of the following manipulations: N gene translocations (N4), G gene truncations (CT1 or CT9), noncytopathic M gene mutations (Mncp), and positioning of the gag gene into the first position of the viral genome (gag1). The resulting N4CT1-gag1, N4CT9-gag1, and MncpCT1-gag1 vectors demonstrated dramatically reduced neurovirulence in mice following direct intracranial inoculation. Surprisingly, in spite of a very high level of attenuation, the N4CT1-gag1 and N4CT9-gag1 vectors generated robust Gag-specific immune responses following intramuscular immunization that were equivalent to or greater than immune responses generated by the more virulent prototypic vectors. MncpCT1-gag1 also induced Gag-specific immune responses following intramuscular immunization that were equivalent to immune responses generated by the prototypic rVSV vector. Placement of the gag gene in the first position of the VSV genome was associated with increased in vitro expression of Gag protein, in vivo expression of Gag mRNA, and enhanced immunogenicity of the vector. These findings demonstrate that through directed manipulation of the rVSV genome, vectors that have reduced neurovirulence and enhanced immunogenicity can be made.


Assuntos
Vacinas contra a AIDS/imunologia , Vetores Genéticos , HIV-1/genética , Vesiculovirus/genética , Vacinas Virais/imunologia , Vacinas contra a AIDS/genética , Animais , Citocinas/biossíntese , Anticorpos Anti-HIV/sangue , Injeções Intramusculares , Dose Letal Mediana , Camundongos , Camundongos Endogâmicos BALB C , Mutação Puntual , Recombinação Genética , Deleção de Sequência , Linfócitos T Citotóxicos/imunologia , Translocação Genética , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/genética , Virulência , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/imunologia
3.
J Virol ; 81(4): 2056-64, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17151112

RESUMO

A variety of rational approaches to attenuate growth and virulence of vesicular stomatitis virus (VSV) have been described previously. These include gene shuffling, truncation of the cytoplasmic tail of the G protein, and generation of noncytopathic M gene mutants. When separately introduced into recombinant VSV (rVSV), these mutations gave rise to viruses distinguished from their "wild-type" progenitor by diminished reproductive capacity in cell culture and/or reduced cytopathology and decreased pathogenicity in vivo. However, histopathology data from an exploratory nonhuman primate neurovirulence study indicated that some of these attenuated viruses could still cause significant levels of neurological injury. In this study, additional attenuated rVSV variants were generated by combination of the above-named three distinct classes of mutation. The resulting combination mutants were characterized by plaque size and growth kinetics in cell culture, and virulence was assessed by determination of the intracranial (IC) 50% lethal dose (LD(50)) in mice. Compared to virus having only one type of attenuating mutation, all of the mutation combinations examined gave rise to virus with smaller plaque phenotypes, delayed growth kinetics, and 10- to 500-fold-lower peak titers in cell culture. A similar pattern of attenuation was also observed following IC inoculation of mice, where differences in LD(50) of many orders of magnitude between viruses containing one and two types of attenuating mutation were sometimes seen. The results show synergistic rather than cumulative increases in attenuation and demonstrate a new approach to the attenuation of VSV and possibly other viruses.


Assuntos
Glicoproteínas de Membrana/genética , Infecções por Rhabdoviridae/virologia , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Feminino , Deleção de Genes , Genes Virais/genética , Camundongos , Proteínas do Nucleocapsídeo/genética , Células Vero , Vírus da Estomatite Vesicular Indiana/crescimento & desenvolvimento , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas da Matriz Viral/genética , Virulência , Replicação Viral
4.
J Virol ; 80(9): 4447-57, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16611905

RESUMO

Recombinant vesicular stomatitis virus (rVSV) vectors offer an attractive approach for the induction of robust cellular and humoral immune responses directed against human pathogen target antigens. We evaluated rVSV vectors expressing full-length glycoprotein D (gD) from herpes simplex virus type 2 (HSV-2) in mice and guinea pigs for immunogenicity and protective efficacy against genital challenge with wild-type HSV-2. Robust Th1-polarized anti-gD immune responses were demonstrated in the murine model as measured by induction of gD-specific cytotoxic T lymphocytes and increased gamma interferon expression. The isotype makeup of the serum anti-gD immunoglobulin G (IgG) response was consistent with the presence of a Th1-CD4+ anti-gD response, characterized by a high IgG2a/IgG1 IgG subclass ratio. Functional anti-HSV-2 neutralizing serum antibody responses were readily demonstrated in both guinea pigs and mice that had been immunized with rVSV-gD vaccines. Furthermore, guinea pigs and mice were prophylactically protected from genital challenge with high doses of wild-type HSV-2. In addition, guinea pigs were highly protected against the establishment of latent infection as evidenced by low or absent HSV-2 genome copies in dorsal root ganglia after virus challenge. In summary, rVSV-gD vectors were successfully used to elicit potent anti-gD Th1-like cellular and humoral immune responses that were protective against HSV-2 disease in guinea pigs and mice.


Assuntos
Vacinas contra o Vírus do Herpes Simples/imunologia , Herpesvirus Humano 2/imunologia , Células Th1/imunologia , Vagina/imunologia , Vagina/virologia , Vírus da Estomatite Vesicular Indiana/genética , Proteínas do Envelope Viral/imunologia , Animais , Formação de Anticorpos/imunologia , Feminino , Vetores Genéticos/genética , Glicoproteínas/genética , Glicoproteínas/imunologia , Glicoproteínas/metabolismo , Cobaias , Vacinas contra o Vírus do Herpes Simples/genética , Herpesvirus Humano 2/genética , Herpesvirus Humano 2/metabolismo , Camundongos , Modelos Animais , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...