Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Virol ; 84(1): 516-31, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19864388

RESUMO

Parvovirus minute virus of mice (MVMp) is endowed with oncotropic properties so far ascribed only to the dependency of the virus life cycle on cellular factors expressed during S phase and/or modulated by malignant transformation. For other viruses oncotropism relies on their inability to circumvent type I interferon (IFN)-induced innate antiviral mechanisms, the first line of defense triggered by normal cells against viral infections. These agents propagate, therefore, preferentially in transformed/tumor cells, which often lack functional antiviral mechanisms. The present study aimed at investigating whether antiviral processes also contribute to MVMp oncotropism. Our results demonstrate that in contrast to MVMp-permissive transformed mouse A9 fibroblasts, freshly isolated normal counterparts (mouse embryonic fibroblasts [MEFs]) mount, through production and release of type I IFNs upon their infection, an antiviral response against MVMp lytic multiplication. Pretreatment of MEFs with a type I IFN-beta-neutralizing antibody, prior to MVMp infection, inhibits the virus-triggered antiviral response and improves the fulfillment of the MVMp life cycle. Our results also show that part of the A9 permissiveness to MVMp relies on the inability to produce type I IFNs upon parvovirus infection, a feature related either to an A9 intrinsic deficiency of this process or to an MVMp-triggered inhibitory mechanism, since stimulation of these cells by exogenous IFN-beta strongly inhibits the parvovirus life cycle. Taken together, our results demonstrate for the first time that parvovirus infection triggers an innate antiviral response in normal cells and suggest that the MVMp oncotropism depends at least in part on the failure of infected transformed cells to mount such a response.


Assuntos
Imunidade Inata , Vírus Miúdo do Camundongo/imunologia , Animais , Linhagem Celular Transformada , Células Cultivadas , Fibroblastos/imunologia , Fibroblastos/virologia , Humanos , Interferon Tipo I/biossíntese , Interferon beta/farmacologia , Camundongos , Replicação Viral/efeitos dos fármacos
2.
J Neurosci Res ; 86(12): 2753-62, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18478553

RESUMO

Prion diseases are fatal neurodegenerative disorders characterized by long incubation periods. To investigate whether concurrent diseases can modify the clinical outcome of prion-affected subjects, we tested the effect of viral infection on the binding and internalization of PrP(Sc), essential steps of prion propagation. To this effect, we added scrapie brain homogenate or purified PrP(Sc) to fibroblasts previously infected with minute virus of mice (MVM), a mouse parvovirus. We show here that the rate of incorporation of PrP(Sc) into MVM-infected cells was significantly higher than that observed for naïve cells. Immunostaining of cells and immunoblotting of subcellular fractions using antibodies recognizing PrP and LysoTracker, a lysosomal marker, revealed that in both control and MVM-infected cells the incorporated PrP(Sc) was associated mostly with lysosomes. Interestingly, flotation gradient analysis revealed that the majority of the PrP(Sc) internalized into MVM-infected cells shifted toward raft-containing low-density fractions. Concomitantly, the MVM-infected cells demonstrated increased levels of the glycosphingolipid GM1 (an essential raft lipid component) throughout the gradient and a shift in caveolin 1 (a raft protein marker) toward lighter membrane fractions compared with noninfected cells. Our results suggest that the effect of viral infection on membrane lipid composition may promote the incorporation of exogenous PrP(Sc) into rafts. Importantly, membrane rafts are believed to be the conversion site of PrP(C) to PrP(Sc); therefore, the association of exogenous PrP(Sc) with such membrane microdomains may facilitate prion infection.


Assuntos
Membrana Celular/metabolismo , Membrana Celular/virologia , Lipídeos de Membrana/metabolismo , Vírus Miúdo do Camundongo/fisiologia , Proteínas PrPSc/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/virologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Cricetinae , Lipídeos de Membrana/fisiologia , Mesocricetus , Camundongos , Proteínas PrPSc/administração & dosagem , Doenças Priônicas/metabolismo , Doenças Priônicas/virologia
3.
Virology ; 506: 141-151, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28391161

RESUMO

The protoparvovirus early promoters, e.g. P4 of Minute Virus of Mice (MVM), play a critical role during infection. Initial P4 activity depends on the host transcription machinery only. Since this is cell-type dependent, it is hypothesized that P4 is a host cell-type range determinant. Yet host range determinants have mapped mostly to capsid, never P4. Here we test the hypothesis using the mouse embryo as a model system. Disruption of the CRE element of P4 drastically decreased infection levels without altering range. However, when we swapped promoter elements of MVM P4 with those from equivalent regions of the closely related H1 virus, we observed elimination of infection in fibroblasts and chondrocytes and the acquisition of infection in skeletal muscle. We conclude that P4 is a host range determinant and a target for modifying the productive infection potential of the virus - an important consideration in adapting these viruses for oncotherapy.


Assuntos
Vírus Miúdo do Camundongo/fisiologia , Infecções por Parvoviridae/virologia , Regiões Promotoras Genéticas , Doenças dos Roedores/virologia , Proteínas não Estruturais Virais/genética , Animais , Regulação Viral da Expressão Gênica , Especificidade de Hospedeiro , Camundongos , Vírus Miúdo do Camundongo/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral
4.
Virology ; 468-470: 631-636, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25310499

RESUMO

The P4 promoter of the autonomous parvovirus Minute Virus of Mice (MVM) drives the production of its non-structural proteins, NS1 and NS2. The NS2 isoforms are without enzymatic activity but interact with cellular proteins. While NS2 is crucial to the viral life cycle in cultured murine cells, NS2-null mutant virus productively infects transformed host cells of other species. In the mouse, sensitivity to MVM infection is age dependent, exhibiting limited subclinical infections in adults, but sustained and potentially lethal infection in embryos. We therefore questioned whether the species-dependent requirement for NS2 function in vitro would be retained in utero. We report here that it is not. NS2-null mutant MVMp is capable of mounting a productive, albeit much reduced, infection of normal embryonic mouse cells in vivo. Based on the data, we hypothesize that NS2 may bear an as-yet undescribed immunosuppressive function.


Assuntos
Embrião de Mamíferos/virologia , Regulação Viral da Expressão Gênica/fisiologia , Vírus Miúdo do Camundongo/fisiologia , Infecções por Parvoviridae/virologia , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Feminino , Deleção de Genes , Humanos , Camundongos , Vírus Miúdo do Camundongo/genética , Gravidez , Proteínas não Estruturais Virais/genética , Replicação Viral
5.
Virology ; 412(1): 233-43, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21295324

RESUMO

Parvoviruses infect and kill tumor cells in vivo and in vitro more efficiently than normal cells. Infection of transformed cells by the parvovirus minute virus of mice (MVM) results in high expression of the major non-structural cytolytic viral protein NS1, which induces a cell death modulated by cellular factors. In this work, we show that MVMp infection and/or NS1 protein expression in permissive transformed rat fibroblast cells leads to apoptosis in wild type and p53(-/-) cells. Apoptotic cell morphology correlates with mitochondrial membrane permeabilization and activation of caspases 9 and 3 but not caspase 8. Thus, further characterization of the antitumor activity of MVMp and its NS1 protein may contribute to the eradication of tumors, including those lacking p53.


Assuntos
Apoptose , Fibroblastos/fisiologia , Fibroblastos/virologia , Vírus Miúdo do Camundongo/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Caspase 3/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Camundongos , Camundongos Knockout , Ratos , Proteína Supressora de Tumor p53/deficiência
6.
Virology ; 379(2): 245-55, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18684479

RESUMO

Well-defined tissue tropism makes Autonomous Parvoviruses a valuable model for studies of virus-cell interactions and gene therapy research. We developed a new Minute Virus of Mice variant, different from the known prototype (MVMp) and immunosuppressive (MVMi) strains. The new virus variant, designated F1, was isolated from the culture of semi-permissive Fisher Rat Fibroblasts, F111, infected with MVMp. The F1 genome carried point mutations in regions known to determine the mutually restricted host ranges of MVMp and MVMi. In F111 cells, F1 cytotoxicity, gene expression and multiplication were significantly higher compared to MVMp. Conversely the wild-type virus propagated in MVMp-permissive cells more efficiently than the F1. Reversion of the F1-specific mutations to wild-type MVMp sequence, following reverse-passaging of the mutant virus in MVMp-permissive cells, confirmed a specific adaptation of the F1 virus to F111 cells. Considerable divergence in tissue specificities between the wild-type and mutant viruses was demonstrated in vivo.


Assuntos
Vírus Miúdo do Camundongo/patogenicidade , Adaptação Fisiológica , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Linhagem Celular , Primers do DNA/genética , DNA Viral/genética , Genes Virais , Variação Genética , Interações Hospedeiro-Patógeno/genética , Humanos , Camundongos , Vírus Miúdo do Camundongo/genética , Vírus Miúdo do Camundongo/fisiologia , Modelos Moleculares , Especificidade de Órgãos , Mutação Puntual , Regiões Promotoras Genéticas , Ratos , Virulência/genética
7.
Virology ; 306(2): 268-79, 2003 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-12642100

RESUMO

Activation of the minute virus of mice (MVM) P4 promoter is a key step in the life cycle of the virus and is completely dependent on host transcription factors. Since transcription-factor composition varies widely in different cell types, there is the possibility that only some cell types in the host organism have the capacity to initiate expression from the P4 promoter and therefore that the promoter may be a factor in determining the tropism of MVM. In this study, the ability of various cell types to activate P4, independent of the other virus-host interactions, was examined in transgenic mouse lines bearing a beta-galactosidase reporter sequence driven by the P4 promoter. It was found that lacZ was expressed during embryogenesis and in the adult in a cell-type-specific and differentiation-dependent pattern. The data are consistent with cell-type and stage-specific activation of the P4 promoter having a role in determining the host cell-type range of MVM. The ability of some parvoviruses to replicate in, and kill oncogenically transformed cells, and to destroy induced tumors in laboratory animals is the basis of recent approaches to use MVM-based vectors in cancer gene therapy. Since these vectors rely on the activation of the P4 promoter by the target tissues, understanding the promoter dependence on cell-type and differentiation status is important for their design and potential use.


Assuntos
Genes Virais , Vírus Miúdo do Camundongo/genética , Regiões Promotoras Genéticas , Animais , Linhagem Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Viral da Expressão Gênica , Genes Reporter , Vetores Genéticos , Óperon Lac , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Vírus Miúdo do Camundongo/crescimento & desenvolvimento , Vírus Miúdo do Camundongo/patogenicidade , Vírus Miúdo do Camundongo/fisiologia , Infecções por Parvoviridae/virologia , Gravidez , Virulência , Replicação Viral , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA