Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros











Intervalo de año de publicación
1.
Virology ; 412(1): 233-43, 2011 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-21295324

RESUMEN

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.


Asunto(s)
Apoptosis , Fibroblastos/fisiología , Fibroblastos/virología , Virus Diminuto del Ratón/patogenicidad , Proteínas no Estructurales Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Ratones , Ratones Noqueados , Ratas , Proteína p53 Supresora de Tumor/deficiencia
2.
J Virol ; 84(10): 5043-51, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20219905

RESUMEN

The relevance of translational control in the gene expression and oncotropism of the autonomous parvoviruses was investigated with MVMp, the prototype strain of minute virus of mice (MVM), infecting normal and transformed rodent and human cells of different tissue origins. Mouse embryo fibroblasts (MEFs) and NIH 3T3 fibroblasts were resistant to MVMp infection, but 3T3 fibroblasts derived from double-stranded RNA (dsRNA)-dependent protein kinase R (PKR) knockout mice (PKR(o/o)) behaved in a manner that was highly permissive to productive MVMp replication. NIH 3T3 resistance correlated with significant phosphorylation of eukaryotic translation initiation factor 2 (eIF2) occurring at early time points after infection. Permissive PKR(o/o) cells were converted to MVMp-restrictive cells after reintroduction of the PKR gene by transfection. Conversely, regulated expression of the vaccinia virus E3 protein, a PKR inhibitor, in MEFs prevented eIF2alpha phosphorylation and increased MVMp protein synthesis. In vitro-synthesized genome-length R1 mRNA of MVMp was a potent activator of PKR. Virus-resistant primary MEFs and NIH 3T3 cells responded to MVMp infection with significant increases in eIF2alpha phosphorylation. In contrast, virus-permissive mouse (PKR(o/o), BHK21, and A9) and human transformed (NB324K fibroblast, U373 glioma, and HepG2 hepatoma) cells consistently showed no significant increase in the level of eIF2alpha phosphorylation following MVMp infection. The synthesis of the viral NS1 protein was inversely correlated with the steady-state PKR levels. Our results show that the PKR-mediated antiviral response is an important mechanism for control of productive MVMp infection, and its impairment in human transformed cells allowed efficient MVMp gene expression. PKR translational control may therefore contribute to the oncolysis of MVMp and other autonomous parvoviruses.


Asunto(s)
Virus Diminuto del Ratón/inmunología , Virus Diminuto del Ratón/patogenicidad , Biosíntesis de Proteínas , Proteínas Virales/biosíntesis , Replicación Viral , eIF-2 Quinasa/inmunología , eIF-2 Quinasa/metabolismo , Animales , Línea Celular , Factor 2 Eucariótico de Iniciación/metabolismo , Fibroblastos/virología , Prueba de Complementación Genética , Hepatocitos/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/virología , Tropismo Viral , eIF-2 Quinasa/deficiencia
3.
Adv Virus Res ; 70: 183-232, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17765706

RESUMEN

Parvoviruses elaborate rugged nonenveloped icosahedral capsids of approximately 260 A in diameter that comprise just 60 copies of a common core structural polypeptide. While serving as exceptionally durable shells, capable of protecting the single-stranded DNA genome from environmental extremes, the capsid also undergoes sequential conformational changes that allow it to translocate the genome from its initial host cell nucleus all the way into the nucleus of its subsequent host. Lacking a duplex transcription template, the virus must then wait for its host to enter S-phase before it can initiate transcription and usurp the cell's synthetic pathways. Here we review cell entry mechanisms used by parvoviruses. We explore two apparently distinct modes of host cell specificity, first that used by Minute virus of mice, where subtle glycan-specific interactions between host receptors and residues surrounding twofold symmetry axes on the virion surface mediate differentiated cell type target specificity, while the second involves novel protein interactions with the canine transferrin receptor that allow a mutant of the feline leukopenia serotype, Canine parvovirus, to bind to and infect dog cells. We then discuss conformational shifts in the virion that accompany cell entry, causing exposure of a capsid-tethered phospholipase A2 enzymatic core that acts as an endosomolytic agent to mediate virion translocation across the lipid bilayer into the cell cytoplasm. Finally, we discuss virion delivery into the nucleus, and consider the nature of transcriptionally silent DNA species that, escaping detection by the cell, might allow unhampered progress into S-phase and hence unleash the parvoviral Trojan horse.


Asunto(s)
Infecciones por Parvoviridae/fisiopatología , Infecciones por Parvoviridae/virología , Parvovirus , Secuencia de Aminoácidos , Animales , Gatos , Línea Celular , Perros , Humanos , Ratones , Virus Diminuto del Ratón/química , Virus Diminuto del Ratón/patogenicidad , Virus Diminuto del Ratón/ultraestructura , Modelos Moleculares , Datos de Secuencia Molecular , Parvovirus/química , Parvovirus/patogenicidad , Parvovirus/ultraestructura , Parvovirus Canino/química , Parvovirus Canino/patogenicidad , Parvovirus Canino/ultraestructura , Ratas , Especificidad de la Especie , Virión/química , Virión/ultraestructura
4.
J Biol Chem ; 281(35): 25670-7, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16822863

RESUMEN

Sialic acid binding is required for infectious cell surface receptor recognition by parvovirus minute virus of mice (MVM). We have utilized a glycan array consisting of approximately 180 different carbohydrate structures to identify the specific sialosides recognized by the prototype (MVMp) and immunosuppressive (MVMi) strains of MVM plus three virulent mutants of MVMp, MVMp-I362S, MVMp-K368R, and MVMp-I362S/K368R. All of the MVM capsids specifically bound to three structures with a terminal sialic acid-linked alpha2-3 to a common Galbeta1-4GlcNAc motif: Neu5Acalpha2-3Galbeta1-4GlcNAcbeta1-4Galbeta1-4GlcNAc (3'SiaLN-LN), Neu5Acalpha2-3Galbeta1-4GlcNAcbeta1-3Galbeta1-4GlcNAcbeta1-3Galbeta1-4GlcNAc (3'SiaLN-LN-LN), and Neu5Acalpha2-3Galbeta1-4(Fucalpha1-3)-GlcNAcbeta1-3Galbeta1-4(Fucalpha1-3)GlcNAcbeta1-3Galbeta1-4(Fucalpha1-3)GlcNAc (sLe(x)-Le(x)-Le(x)). In addition, MVMi also recognized four multisialylated glycans with terminal alpha2-8 linkages: Neu5Acalpha2-8Neu5Acalpha2-8Neu5Acalpha ((Sia)(3)), Neu5Acalpha2-8Neu5Acalpha2-3Galbeta1-4Glc (GD3), Neu5Acalpha2-8Neu5Acalpha2-8Neu5Acalpha2-3Galbeta1-4Glc (GT3), and Neu5Acalpha2-8Neu5Acalpha2-3(GalNAcbeta1-4)Galbeta1-4Glc (GD2). Interestingly, the virulent MVMp-K368R mutant also recognized GT3. Analysis of the relative binding affinities using a surface plasmon resonance biospecific interaction (BIAcore) assay showed the wild-type MVMp and MVMi capsids binding with higher affinity to selected glycans compared with the virulent MVMp mutants. The reduced affinity of the virulent MVMp mutants are consistent with previous in vitro cell binding assays that had shown weaker binding to permissive cells compared with wild-type MVMp. This study identifies the sialic acid structures recognized by MVM. It also provides rationale for the tropism of MVM for malignant transformed cells that contain sLe(x) motifs and the neurotropism of MVMi, which is likely mediated via interactions with multisialylated glycans known to be tumor cell markers. Finally, the observations further implicate a decreased binding affinity for sialic acid in the in vivo adaptation of MVMp to a virulent phenotype.


Asunto(s)
Virus Diminuto del Ratón/metabolismo , Virus Diminuto del Ratón/patogenicidad , Ácido N-Acetilneuramínico/química , Sitios de Unión , Cápside/química , Modelos Moleculares , Mutación , Fenotipo , Polisacáridos/química , Unión Proteica , Virulencia
5.
Proc Natl Acad Sci U S A ; 102(47): 17148-53, 2005 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-16284249

RESUMEN

Enveloped viruses deliver their virions into the host cell by fusion with the cellular plasma or endosomal membrane, thus creating topological continuity between the cytosol and the inside of the viral envelope. Nonenveloped viruses are, by their very nature, denied this strategy and must employ alternative methods to breach their host cell's delimiting membrane. We show here that the compact icosahedral parvoviral virion gains entry by deploying a lipolytic enzyme, phospholipase A(2) (PLA(2)), that is expressed at the N terminus of VP1, the minor coat protein. This region of VP1 is normally sequestered within the viral shell but is extruded during the entry process as a capsid-tethered domain. A single amino acid substitution in the active site of the VP1 PLA(2) inactivates enzymatic activity and abrogates infectivity. We have used transencapsidation of a vector expressing green fluorescent protein to show that infection by this PLA(2)-defective mutant can be complemented by coinfection with wild-type or mutant full virions, provided they can express a functional PLA(2). Even though wild-type empty capsids contain an active form of the enzyme, it is not externalized under physiological conditions, and such capsids are not able to complement the PLA(2) mutant. Significantly, highly efficient rescue can be achieved by polyethyleneimine-induced endosome rupture or by coinfection with adenovirus as long as uptake of the two viruses is simultaneous and the adenovirus is capable of deploying pVI, a capsid protein with endosomolytic activity. Together, these results demonstrate a previously unrecognized enzymatic mechanism for nonenveloped virus penetration.


Asunto(s)
Proteínas de la Cápside/fisiología , Endosomas/virología , Membranas Intracelulares/virología , Lipólisis/fisiología , Virus Diminuto del Ratón/enzimología , Virus Diminuto del Ratón/patogenicidad , Fosfolipasas A/fisiología , Virión/enzimología , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Secuencia de Bases , Proteínas de la Cápside/genética , Línea Celular , Endosomas/enzimología , Histidina/genética , Histidina/metabolismo , Membranas Intracelulares/enzimología , Lipólisis/genética , Ratones , Virus Diminuto del Ratón/genética , Datos de Secuencia Molecular , Fosfolipasas A/genética , Virión/genética
6.
J Gen Virol ; 86(Pt 4): 1009-1014, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15784894

RESUMEN

The non-structural proteins NS1 and NS2 of the parvovirus minute virus of mice (MVM) are required for efficient virus replication. It has previously been shown that NS1 and NS2 interact and colocalize with the survival motor neuron (Smn) gene product in novel nuclear structures that are formed late in infection, termed Smn-associated APAR (autonomous parvovirus-associated replication) bodies (SAABs). It is not clear what molecular viral intermediate(s) contribute to SAAB formation. The current results address the role of NS2 in SAAB formation. In highly synchronized wild-type MVM infection of murine A9(2L) cells, NS2 colocalizes with Smn and other SAAB constituents. An MVM mutant that does not produce NS2 still generates SAABS, albeit with a temporal delay. The lag in SAAB formation seen in the absence of NS2 is probably related to the temporal delay in virus replication, suggesting that, whilst NS2 is required for efficient viral infection, it is dispensable for SAAB formation.


Asunto(s)
Cuerpos Enrollados/metabolismo , Virus Diminuto del Ratón/fisiología , Ribonucleoproteínas Nucleares Pequeñas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Animales , Autoantígenos , Línea Celular , Regulación Viral de la Expresión Génica , Ratones , Virus Diminuto del Ratón/patogenicidad , Replicación Viral , Proteínas Nucleares snRNP
7.
J Virol ; 79(1): 289-98, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15596824

RESUMEN

Gene therapy vectors have been developed from autonomous rodent parvoviruses that carry a therapeutic gene or a marker gene in place of the genes encoding the capsid proteins. These vectors are currently evaluated in preclinical experiments. The infectivity of the vector particles deriving from the fibroblastic strain of minute virus of mice (MVMp) (produced by transfection in human cells) was found to be far less (approximately 50-fold-less) infectious than that of wild-type virus particles routinely produced by infection of A9 mouse fibroblasts. Similarly, wild-type MVMp produced by transfection also had a low infectivity in mouse cells, indicating that the method and producer cells influence the infectivity of the virus produced. Interestingly, producer cells made as many full vector particles as wild-type particles, arguing against deficient packaging being responsible for the low infectivity of viruses recovered from transfected cells. The hurdle to infection with full particles produced through transfection was found to take place at an early step following entry and limiting viral DNA replication and gene expression. Infections with transfection or infection-derived virus stocks normalized for their replication ability yielded similar monomer and dimer DNA amplification and gene expression levels. Surprisingly, at equivalent replication units, the capacity of parvovirus vectors to kill tumor cells was lower than that of the parental wild-type virus produced under the same transfection conditions, suggesting that beside the viral nonstructural proteins, the capsid proteins, assembled capsids, or the corresponding coding region contribute to the lytic activity of these viruses.


Asunto(s)
Vectores Genéticos , Virus Diminuto del Ratón/patogenicidad , Recombinación Genética , Virión/patogenicidad , Ensamble de Virus , Animales , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Línea Celular , Humanos , Ratones , Virus Diminuto del Ratón/genética , Virus Diminuto del Ratón/fisiología , Plásmidos , Transfección , Ensayo de Placa Viral , Virión/genética , Virión/metabolismo , Replicación Viral
8.
J Virol ; 77(15): 8495-503, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12857918

RESUMEN

The functional disturbance of self-renewing and multipotent hematopoietic stem cells (HSCs) in viral diseases is poorly understood. In this report, we have assessed the susceptibility of mouse HSCs to strain i of the autonomous parvovirus minute virus of mice (MVMi) in vitro and during persistent infection of an immunodeficient host. Purified 5FU(r) Lin(-) Sca-1(+) primitive hematopoietic precursors were permissive for MVMi genome replication and the expression of viral gene products. The lymphoid and myeloid repopulating capacity of bone marrow (BM) cells was significantly impaired after in vitro infection, although the degree of functional effect proportionally decreased with the posttransplantation time. This indicated that MVMi targets the heterogeneous compartment of repopulating cells with differential affinity and suggests that the virus may persist in some primitive HSCs in the quiescent stage, killing those eventually recruited for proliferative activity. Immunodeficient SCID mice oronasally infected with MVMi were cured of the characteristic virus-induced lethal leukopenia by transplantation of immunocompetent BM grafts. However, two double-stranded viral DNA species, probably uncommon replicative intermediates, remained in the marrow of every transplanted mouse months after infectious virus clearance. Genetic analysis of the rescued mice showed that the infection ensured a stable engraftment of donor hematopoiesis by markedly depleting the pool of endogenous HSCs. The MVMi-induced suppression of HSC functions illustrates the accessibility of this compartment to infection during a natural viral hematological disease. These results may provide clues to understanding delayed hematopoietic syndromes associated with persistent viral infections and to prospective gene delivery to HSCs in vivo.


Asunto(s)
Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/virología , Virus Diminuto del Ratón/patogenicidad , Infecciones por Parvoviridae/patología , Animales , Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Ensayo de Unidades Formadoras de Colonias , Hematopoyesis , Células Madre Hematopoyéticas/patología , Leucopenia/etiología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Infecciones por Parvoviridae/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
9.
Virology ; 306(2): 268-79, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12642100

RESUMEN

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.


Asunto(s)
Genes Virales , Virus Diminuto del Ratón/genética , Regiones Promotoras Genéticas , Animales , Línea Celular , Femenino , Regulación del Desarrollo de la Expresión Génica , Regulación Viral de la Expresión Génica , Genes Reporteros , Vectores Genéticos , Operón Lac , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Virus Diminuto del Ratón/crecimiento & desarrollo , Virus Diminuto del Ratón/patogenicidad , Virus Diminuto del Ratón/fisiología , Infecciones por Parvoviridae/virología , Embarazo , Virulencia , Replicación Viral , beta-Galactosidasa/genética
10.
J Virol ; 76(14): 7049-59, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12072505

RESUMEN

This report describes the distribution of conventional nuclear localization sequences (NLS) and of a beta-stranded so-called nuclear localization motif (NLM) in the two proteins (VP1, 82 kDa; VP2, 63 kDa) forming the T=1 icosahedral capsid of the parvovirus minute virus of mice (MVM) and their functions in viral biogenesis and the onset of infection. The approximately 10 VP1 molecules assembled in the MVM particle harbor in its 142-amino-acid (aa) N-terminal-specific region four clusters of basic amino acids, here called BC1 (aa 6 to 10), BC2 (aa 87 to 90), BC3 (aa 109 to 115), and BC4 (aa 126 to 130), that fit consensus NLS and an NLM placed toward the opposite end of the polypeptide (aa 670 to 680) found to be necessary for VP2 nuclear uptake. Deletions and site-directed mutations constructed in an infectious MVM plasmid showed that BC1, BC2, and NLM are cooperative nuclear transport sequences in singly expressed VP1 subunits and that they conferred nuclear targeting competence on the VP1/VP2 oligomers arising in normal infection, while BC3 and BC4 did not display nuclear transport activity. Notably, VP1 proteins mutated at BC1 and -2, and particularly with BC1 to -4 sequences deleted, induced nuclear and cytoplasmic foci of colocalizing conjugated ubiquitin that could be rescued from the ubiquitin-proteasome degradation pathway by the coexpression of VP2 and NS2 isoforms. These results suggest a role for VP2 in viral morphogenesis by assisting cytoplasmic folding of VP1/VP2 subviral complexes, which is further supported by the capacity of NLM-bearing transport-competent VP2 subunits to recruit VP1 into the nuclear capsid assembly pathway regardless of the BC composition. Instead, all four BC sequences, which are located in the interior of the capsid, were absolutely required by the incoming infectious MVM particle for the onset of infection, suggesting either an important conformational change or a disassembly of the coat for nuclear entry of a VP1-associated viral genome. Therefore, the evolutionarily conserved BC sequences and NLM domains provide complementary nuclear transport functions to distinct supramolecular complexes of capsid proteins during the autonomous parvovirus life cycle.


Asunto(s)
Cápside/metabolismo , Virus Diminuto del Ratón/patogenicidad , Señales de Localización Nuclear/fisiología , Ensamble de Virus , Animales , Cápside/química , Proteínas de la Cápside , Línea Celular Transformada , Núcleo Celular/metabolismo , Humanos , Ratones , Virus Diminuto del Ratón/genética , Virus Diminuto del Ratón/metabolismo , Infecciones por Parvoviridae/virología , Virión/metabolismo , Virión/patogenicidad
11.
J Virol ; 76(8): 3892-904, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11907229

RESUMEN

The human survival motor neuron (SMN) gene is the spinal muscular atrophy-determining gene, and a knockout of the murine Smn gene results in preembryonic lethality. Here we show that SMN can directly interact in vitro and in vivo with the large nonstructural protein NS1 of the autonomous parvovirus minute virus of mice (MVM), a protein essential for viral replication and a potent transcriptional activator. Typically, SMN localizes within nuclear Cajal bodies and diffusely in the cytoplasm. Following transient NS1expression, SMN and NS1 colocalize within Cajal bodies. At early time points following parvovirus infection, NS1 fails to colocalize with SMN within Cajal bodies; however, during the course of MVM infection, dramatic nuclear alterations occur. Formerly distinct nuclear bodies such as Cajal bodies, promyelocytic leukemia gene product (PML) oncogenic domains (PODs), speckles, and autonomous parvovirus-associated replication (APAR) bodies are seen aggregating at later points in infection. These newly formed large nuclear bodies (termed SMN-associated APAR bodies) are active sites of viral replication and viral capsid assembly. These results highlight the transient nature of nuclear bodies and their contents and identify a novel nuclear body formed during infection. Furthermore, simple transient expression of the viral nonstructural proteins is insufficient to induce this nuclear reorganization, suggesting that this event is induced specifically by a step in the viral infection process.


Asunto(s)
Cuerpos Enrollados/metabolismo , Virus Diminuto del Ratón/patogenicidad , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Parvoviridae/virología , Proteínas no Estructurales Virales/metabolismo , Animales , Técnicas Biosensibles , Línea Celular , Cuerpos Enrollados/virología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Humanos , Inmunohistoquímica , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN , Proteínas del Complejo SMN , Proteínas no Estructurales Virales/genética
12.
J Virol ; 75(23): 11573-82, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11689639

RESUMEN

The infection outcome of the Parvoviridae largely relies on poorly characterized intracellular factors modulated by proliferation, differentiation, and transformation of host cells. We have studied the interactions displayed by the highly homologous p and i strains of the murine parvovirus minute virus of mice (MVM), with a series of transformed cells of rat (C6) and human (U373, U87, SW1088, SK-N-SH) nervous system origin, seeking for molecular mechanisms governing parvovirus host range. The MVMp infection of C6 and U373 cells was cytotoxic and productive, whereas the other nervous cells behaved essentially as resistant to this virus. In contrast, MVMi did not complete its life cycle in any of the human nervous cells, though it efficiently killed the astrocytic tumor cells by two types of nonproductive infections: (i) normal synthesis of all viral macromolecules with a late defect in infectious virion maturation and release to the medium in U373; and (ii) high levels of accumulation of the full set of viral messenger RNAs and of both nonstructural (NS-1) and structural (VP-1 and VP-2) proteins, under a very low viral DNA amplification, in U87 and SW1088 cells. Further analyses showed that U87 was permissive for nuclear transport of MVMi proteins, leading to efficient assembly of empty viral capsids with a normal phosphorylation and VP1-to-VP2 ratio. The DNA amplification blockade in U87 occurred after conversion of the incoming MVMi genome to the monomeric replicative form, and it operated independently of the delivery pathway used by the viral particle, since it could not be overcome by transfection with cloned infectious viral DNA. Significantly, a chimeric MVMi virus harboring the coding region of the nonstructural (NS) gene replaced with that of MVMp showed a similar pattern of restriction in U87 cells as the parental MVMi virus, and it attained in U373 cultures an infectious titer above 100-fold higher under equal levels of DNA amplification and genome encapsidation. The results suggest that the activity of complexes formed by the NS polypeptides and recruited cellular factors restrict parvovirus DNA amplification in a cell type-dependent manner and that NS functions may in addition determine MVM host range acting at postencapsidation steps of viral maturation. These data are relevant for understanding the increased multiplication of autonomous parvovirus in some transformed cells and the transduction efficacy of nonreplicative parvoviral vectors, as well as a general remark on the mechanisms by which NS genes may regulate viral tropism and pathogenesis.


Asunto(s)
Replicación del ADN , Genoma Viral , Virus Diminuto del Ratón/genética , Proteínas no Estructurales Virales/genética , Animales , Glioblastoma/virología , Humanos , Ratones , Virus Diminuto del Ratón/patogenicidad , Virus Diminuto del Ratón/fisiología , Fosforilación , Ratas , Células Tumorales Cultivadas , Proteínas Estructurales Virales/genética , Virulencia , Replicación Viral
13.
J Gen Virol ; 79 ( Pt 3): 581-6, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9519837

RESUMEN

Two strains of minute virus of mice (MVM) show different host-cell specificities. MVM(i) grows in T lymphocytes whereas MVM(p) is fibroblast-specific. By constructing recombinant viral DNAs between the genomes of the two strains, we have shown that two segments of the MVM(i) genome are required for lytic viral growth in T lymphocytic EL4 cells. One segment (iE) was found between nucleotides 1084 and 2070, in a region encoding the early viral proteins and containing mRNA splice signals and the late P39 promoter. The other (iL) was between nucleotides 3523 and 4339 in the region coding for capsid protein. The P39 promoters within the E segment from MVM(i) or MVM(p) were equally active in transfected EL4 cells. However, pE-containing MVM DNA produced more NS2 mRNA than iE-containing DNA, apparently the result of virus-strain-specific differences in the regulation of splicing.


Asunto(s)
Replicación del ADN/genética , Fibroblastos/virología , Virus Diminuto del Ratón/genética , ARN Viral/metabolismo , Linfocitos T/virología , Secuencia de Aminoácidos , Animales , Ratones , Virus Diminuto del Ratón/patogenicidad , Datos de Secuencia Molecular , Recombinación Genética , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
14.
J Virol ; 70(11): 8109-16, 1996 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8892936

RESUMEN

Newborn BALB/c mice intranasally inoculated at birth with a lethal dose of the immunosuppressive strain of the parvovirus minute virus of mice (MVMi) developed motor disabilities and intention tremors with a high incidence by the day 6 postinfection (dpi). These neurological syndromes paralleled the synthesis of virus intermediate DNA replicative forms and yield of infectious particles in the brain, with kinetics that peaked by this time. The preferred virus replicative sites in the brain were established early in the infection (2 dpi) and at the onset of clinical symptoms (6 dpi) and were compared with major regions of cellular proliferative activity found after intraperitoneal injection of bromodeoxyuridine 24 h before encephalons were subjected to immunohistochemistry detection. At 2 dpi, viral capsid antigen was located in the laterodorsal thalamic and the pontine nuclei but not in the extensive proliferative regions of the mouse brain at this postnatal day. At 6 dpi, however, the neurotropism of the MVMi was highlighted by its ability to target the subventricular zone of the ventricles, the subependymal zone of the olfactory bulb, and the dentate gyrus of the hippocampus, which are the three main germinal centers of the cerebrum in mouse postbirth neurogenesis. Unexpectedly, in the cerebellum, the MVMi capsid antigen was confined exclusively to cells that have undergone mitosis and have migrated to the internal granular layer (IGL) and not to the proliferative external granular layer (EGL), which was stained with antiproliferative cell nuclear antigen antibody and is the main target in other parvovirus infections. This result implies temporal or differentiation coupling between MVMi cycle and neuroblast morphogenesis, since proliferative granules of the EGL should primarily be infected but must migrate in a virus carrier state into the IGL in order to express the capsid proteins. During migration, many cells undergo destruction, accounting for the marked hypocellularity specifically found in the IGL and the irregular alignment of Purkinje cell bodies, both consistent histopathological hallmarks of animals developing cerebellar symptoms. We conclude that MVMi impairs postmitotic neuronal migration occurring in the first postnatal week, when, through the natural respiratory route of infection, the virus titer peaks in the encephalon. The results illustrate the intimate connection between MVMi neuropathogenesis and mouse brain morphogenetic stage, underscoring the potential of parvoviruses as markers of host developmental programs.


Asunto(s)
Encéfalo/virología , Virus Diminuto del Ratón/metabolismo , Neuronas/virología , Infecciones por Parvoviridae/virología , Animales , Animales Recién Nacidos , Encéfalo/patología , Cápside/metabolismo , División Celular , Línea Celular Transformada , Sistema Nervioso Central/virología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Virus Diminuto del Ratón/crecimiento & desarrollo , Virus Diminuto del Ratón/patogenicidad , Infecciones por Parvoviridae/patología , Infecciones por Parvoviridae/fisiopatología , Células Tumorales Cultivadas
15.
J Virol ; 69(5): 3229-32, 1995 May.
Artículo en Inglés | MEDLINE | ID: mdl-7707557

RESUMEN

The in vivo myelosuppressive capacity of strain i of the parovirus minute virus of mice (MVMi) was investigated in newborn BALB/c mice inoculated with a lethal intranasal dose. MVMi infection reached maximum levels of DNA synthesis and infectious titers in lymphohemopoietic organs at 4 to 6 days postinoculation and was restricted by an early neutralizing humoral immune response. After viral control (by 10 days postinoculation), a significant decrease in femoral and splenic cellularity, as well as in granulocyte-macrophage colony-forming unit and erythroid burst-forming unit hemopoietic progenitors, was observed in most inoculated animals. This delayed myeloid depression, although it may be not a major cause of the lethality of the infection, implies indirect pathogenic mechanisms induced by MVMi infection in a susceptible host.


Asunto(s)
Enfermedades de la Médula Ósea/etiología , Virus Diminuto del Ratón/patogenicidad , Infecciones por Parvoviridae/etiología , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/biosíntesis , Enfermedades de la Médula Ósea/inmunología , Enfermedades de la Médula Ósea/virología , Hematopoyesis , Sistema Hematopoyético/virología , Ratones , Ratones Endogámicos BALB C , Virus Diminuto del Ratón/inmunología , Virus Diminuto del Ratón/fisiología , Infecciones por Parvoviridae/inmunología , Infecciones por Parvoviridae/virología , Factores de Tiempo , Replicación Viral
16.
J Virol ; 68(10): 6446-53, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8083981

RESUMEN

The nonstructural (NS) proteins of the autonomous parvovirus minute virus of mice are involved in viral DNA replication and in the regulation of homologous and heterologous promoters. Moreover, NS products have proved to be cytotoxic, especially for transformed cells. We show here that intracellular accumulation of NS products is not sufficient to kill rat fibroblasts from the established cell line FR3T3, which is phenotypically normal in several respects. FRNS cell lines were obtained by stable transfection of FR3T3 cells by a vector carrying the NS genes under the control of the hormone-inducible long terminal repeat promoter of the mouse mammary tumor virus. In the presence of dexamethasone, the NS proteins were synthesized without associated cell death. Transformation of FRNS cells with the c-Ha-ras oncogene or polyomavirus oncogenes had little effect on their capacity for NS induction, as measured at both concentration and transactivating activity levels, yet the transformants were now dying within a few days in the presence of the inducer. The same results were obtained with cells stably transfected by a vector expressing the NS1 product alone, suggesting that in this system there is no cooperation between NS1 and NS2 for maximal cytopathic effect. Cell mortality after NS protein induction was quantitatively related to the yield of oncogene expression, while NS-1 was not limiting in this respect. Our results show that the NS1 protein is not lethal unless cellular factors that may depend on oncogene expression trigger its cytotoxicity.


Asunto(s)
Supervivencia Celular , Expresión Génica , Genes ras , Virus Diminuto del Ratón/genética , Virus Diminuto del Ratón/patogenicidad , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Proteínas no Estructurales Virales/biosíntesis , Animales , Antígenos Transformadores de Poliomavirus/biosíntesis , División Celular/efectos de los fármacos , Línea Celular , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Cloranfenicol O-Acetiltransferasa/biosíntesis , Dexametasona/farmacología , Humanos , Recién Nacido , Riñón , Cinética , Virus Diminuto del Ratón/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/análisis , Ratas , Virus 40 de los Simios/genética , Virus 40 de los Simios/metabolismo , Activación Transcripcional , Transfección , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria , Proteínas no Estructurales Virales/análisis
17.
Res Virol ; 144(6): 465-70, 1993.
Artículo en Inglés | MEDLINE | ID: mdl-8140289

RESUMEN

Stable Psi-2 cell transformants were selected for their resistance to neomycin after transfection with a retroviral pZipNeo-SVX vector carrying sequences encoding for the non-structural proteins of parvovirus minute virus of mice (prototype strain, MVMp). Cells producing both NS-1 and NS-2 proteins (PsiNS) or only the NS-2 polypeptide (PsiNS2) were obtained. PsiNS cells exhibited morphological abnormalities and had a reduced clone-forming ability, whereas PsiNS2 cells were indistinguishable from the parental line. These cellular systems produced recombinant retroviral particles which transduced the NS gene(s) into mouse A9 cells. As in the case of Psi-2 cells, A9 transformants expressing both NS-1 and NS-2 proteins were impaired in their cloning efficiency. These results provided a direct confirmation of the predominant role of protein NS-1 in the cytopathic effect of parvoviruses.


Asunto(s)
Virus Diminuto del Ratón/patogenicidad , Proteínas no Estructurales Virales/biosíntesis , Animales , Células Cultivadas , Ratones , Virus Diminuto del Ratón/genética , Proteínas Recombinantes/biosíntesis , Retroviridae , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/fisiología
18.
Virology ; 195(1): 149-55, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8317090

RESUMEN

The parvovirus-encoded nonstructural (NS) proteins have been implicated in the cytopathogenicity of these agents. Although protein NS-1 of minute virus of mice (MVM) has been shown to be toxic, little is known about the role of NS-2 in this process. In order to determine the contribution of NS-1 and NS-2 to cytotoxicity, we took advantage of an expression system controlled by the mouse mammary tumor virus promoter which responds to glucocorticoid stimulation and which controls the expression of both MVM(p) NS proteins. Different mutations were introduced in NS genes so as to affect the NS-1 or NS-2 protein. Neoplastic human cell lines expressing only NS-1 protein after induction by dexamethasone undergo a smaller lethality compared to lines expressing both wild-type proteins. Mutations that were introduced in NS-1 coding sequence and did not affect NS-2 were found to drastically suppress the cytotoxic effect. It is concluded that the NS-2 protein has little cytotoxic activity by itself but is required for the full expression of the viral cytopathic effect on transformed human cells. Furthermore these results lead us to suggest that the NS-2 cytotoxic domain is localized in the amino-terminal portion of the protein.


Asunto(s)
Virus Diminuto del Ratón/patogenicidad , Proteínas no Estructurales Virales/biosíntesis , Secuencia de Bases , División Celular , Línea Celular Transformada , Supervivencia Celular , Clonación Molecular , Efecto Citopatogénico Viral , ADN Viral , Dexametasona , Humanos , Immunoblotting , Virus Diminuto del Ratón/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Plásmidos , Regiones Promotoras Genéticas , Proteínas no Estructurales Virales/toxicidad
19.
J Virol ; 66(5): 3118-24, 1992 May.
Artículo en Inglés | MEDLINE | ID: mdl-1373202

RESUMEN

Neonatal C3H/He mice were oronasally inoculated with similar doses of four genotypes of minute virus of mice (MVM). MVMp, a fibroblast-specific variant, caused an asymptomatic infection. MVM(1035), a chimera which had the allotropic determinant of virulent MVMi inserted onto an MVMp background, caused a lethal infection and renal papillary infarcts, the hallmark of MVMi infection. MVMi(NS2-1990), the virulent lymphocyte-specific variant mutated to eliminate NS2 synthesis, was infectious but caused an asymptomatic infection. Sequential virus titration, histology, in situ hybridization with a full-length MVMi genomic probe, and immunohistochemistry for viral capsid antigen were used to compare the pathogenesis of infection with the four MVM genotypes. Infectious virus was recovered from multiple organs of mice infected with MVMi, MVMp, and MVM(1035) but not from mice infected with MVMi(NS2-1990). MVMp titers were lower than MVMi titers in all organs except the intestine. MVM(1035) titers were higher than MVMi titers in all organs except the blood. MVMp was localized to connective tissue elements of the intestine, to cells in mesenteric lymph nodes, and rarely to cells in other organs. MVM(1035) was localized to multiple organs and shared the same target cells, endothelium, lymphoid cells, and hematopoietic cells, as MVMi. MVM(1035) also replicated in external germinal cells of the cerebellum and smooth muscle cells of the stomach and colon, which were not targets of MVMi or MVMp infection. MVMi(NS2-1990) replicated to a limited degree in some MVMi target organs.


Asunto(s)
Cápside/biosíntesis , Virus Diminuto del Ratón/patogenicidad , Infecciones por Parvoviridae/genética , Proteínas del Núcleo Viral/biosíntesis , Animales , Animales Recién Nacidos , Cápside/genética , Cápside/aislamiento & purificación , ADN Viral/análisis , Epítopos , Variación Genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C3H , Virus Diminuto del Ratón/genética , Virus Diminuto del Ratón/aislamiento & purificación , Mutagénesis , Hibridación de Ácido Nucleico , Infecciones por Parvoviridae/patología , Relación Estructura-Actividad , Distribución Tisular , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/aislamiento & purificación , Proteínas no Estructurales Virales , Virulencia/genética
20.
J Virol ; 64(6): 2537-44, 1990 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-2139892

RESUMEN

Human fibroblasts and epithelial cells differing in their susceptibility to killing by the autonomous parvoviruses H-1 and minute virus of mice were compared for their capacity to express viral mRNAs and proteins. The transition from a parvovirus-resistant to a parvovirus-sensitive phenotype correlated with a proportional increase in the production of the three major viral transcripts and of structural and nonstructural proteins. In contrast, cell sensitization to parvovirus could not be correlated with detectable changes in virus uptake, intracellular localization of gene products, stability of viral mRNAs, or phosphorylation of viral nonstructural polypeptides. Moreover, the H-1 virus-sensitive keratinocyte line studied did not sustain a greater level of viral DNA amplification than its resistant derivative. Therefore, the differential susceptibility of the human cells tested to parvovirus infection appears to be mainly controlled at the level of transcription of the viral genome. Parvoviral gene expression could not be elevated by increasing the input multiplicity of infection in either of the cell systems analyzed. Together, these data suggest that a cellular factor(s) regulating parvoviral transcription may be modulated by oncogenic transformation or by differentiation, as both features have been shown to affect cell susceptibility to parvoviruses.


Asunto(s)
Virus Diminuto del Ratón/genética , Parvoviridae/genética , Transcripción Genética , Cápside/genética , División Celular , Línea Celular , Supervivencia Celular , ADN Viral/genética , Células Epiteliales , Fibroblastos/citología , Amplificación de Genes , Expresión Génica , Genes Virales , Humanos , Queratinocitos/citología , Virus Diminuto del Ratón/patogenicidad , Hibridación de Ácido Nucleico , Parvoviridae/patogenicidad , ARN Viral/genética , ARN Viral/aislamiento & purificación , Proteínas del Núcleo Viral/genética , Proteínas no Estructurales Virales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA