Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arch Virol ; 165(9): 2133-2146, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32533329

RESUMEN

Parvoviridae, a diverse family of small single-stranded DNA viruses was established in 1975. It was divided into two subfamilies, Parvovirinae and Densovirinae, in 1993 to accommodate parvoviruses that infect vertebrate and invertebrate animals, respectively. This relatively straightforward segregation, using host association as the prime criterion for subfamily-level classification, has recently been challenged by the discovery of divergent, vertebrate-infecting parvoviruses, dubbed "chapparvoviruses", which have proven to be more closely related to viruses in certain Densovirinae genera than to members of the Parvovirinae. Viruses belonging to these genera, namely Brevi-, Hepan- and Penstyldensovirus, are responsible for the unmatched heterogeneity of the subfamily Densovirinae when compared to the Parvovirinae in matters of genome organization, protein sequence homology, and phylogeny. Another genus of Densovirinae, Ambidensovirus, has challenged traditional parvovirus classification, as it includes all newly discovered densoviruses with an ambisense genome organization, which introduces genus-level paraphyly. Lastly, current taxon definition and virus inclusion criteria have significantly limited the classification of certain long-discovered parvoviruses and impedes the classification of some potential family members discovered using high-throughput sequencing methods. Here, we present a new and updated system for parvovirus classification, which includes the introduction of a third subfamily, Hamaparvovirinae, resolves the paraphyly within genus Ambidensovirus, and introduces new genera and species into the subfamily Parvovirinae. These proposals were accepted by the ICTV in 2020 March.


Asunto(s)
Infecciones por Parvoviridae/veterinaria , Infecciones por Parvoviridae/virología , Parvoviridae/clasificación , Parvoviridae/fisiología , Filogenia , Animales , Especificidad del Huésped , Humanos , Parvoviridae/genética , Parvoviridae/aislamiento & purificación , Proteínas Virales/genética
2.
J Gen Virol ; 100(3): 367-368, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30672729

RESUMEN

Members of the family Parvoviridae are small, resilient, non-enveloped viruses with linear, single-stranded DNA genomes of 4-6 kb. Viruses in two subfamilies, the Parvovirinae and Densovirinae, are distinguished primarily by their respective ability to infect vertebrates (including humans) versus invertebrates. Being genetically limited, most parvoviruses require actively dividing host cells and are host and/or tissue specific. Some cause diseases, which range from subclinical to lethal. A few require co-infection with helper viruses from other families. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the Parvoviridae, which is available at www.ictv.global/report/parvoviridae.


Asunto(s)
Infecciones por Parvoviridae/virología , Parvoviridae/clasificación , Filogenia , Animales , Genoma Viral , Humanos , Parvoviridae/genética , Parvoviridae/aislamiento & purificación , Parvoviridae/ultraestructura , Virología/organización & administración
3.
Bioinformatics ; 34(13): 2195-2200, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29474519

RESUMEN

Motivation: The increasing rate of submission of genetic sequences into public databases is providing a growing resource for classifying the organisms that these sequences represent. To aid viral classification, we have developed ViCTree, which automatically integrates the relevant sets of sequences in NCBI GenBank and transforms them into an interactive maximum likelihood phylogenetic tree that can be updated automatically. ViCTree incorporates ViCTreeView, which is a JavaScript-based visualization tool that enables the tree to be explored interactively in the context of pairwise distance data. Results: To demonstrate utility, ViCTree was applied to subfamily Densovirinae of family Parvoviridae. This led to the identification of six new species of insect virus. Availability and implementation: ViCTree is open-source and can be run on any Linux- or Unix-based computer or cluster. A tutorial, the documentation and the source code are available under a GPL3 license, and can be accessed at http://bioinformatics.cvr.ac.uk/victree_web/. Supplementary information: Supplementary data are available at Bioinformatics online.


Asunto(s)
Filogenia , Análisis de Secuencia de Proteína/métodos , Programas Informáticos , Proteínas Virales/metabolismo , Virus/genética , Secuencia de Aminoácidos , Bases de Datos Factuales , Proteínas Virales/química , Proteínas Virales/genética , Virus/metabolismo
4.
Viruses ; 10(2)2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29385689

RESUMEN

Combining virus-enhanced immunogenicity with direct delivery of immunomodulatory molecules would represent a novel treatment modality for melanoma, and would require development of new viral vectors capable of targeting melanoma cells preferentially. Here we explore the use of rodent protoparvoviruses targeting cells of the murine melanoma model B16F10. An uncloned stock of mouse parvovirus 1 (MPV1) showed some efficacy, which was substantially enhanced following serial passage in the target cell. Molecular cloning of the genes of both starter and selected virus pools revealed considerable sequence diversity. Chimera analysis mapped the majority of the improved infectivity to the product of the major coat protein gene, VP2, in which linked blocks of amino acid changes and one or other of two apparently spontaneous mutations were selected. Intragenic chimeras showed that these represented separable components, both contributing to enhanced infection. Comparison of biochemical parameters of infection by clonal viruses indicated that the enhancement due to changes in VP2 operates after the virus has bound to the cell surface and penetrated into the cell. Construction of an in silico homology model for MPV1 allowed placement of these changes within the capsid shell, and revealed aspects of the capsid involved in infection initiation that had not been previously recognized.


Asunto(s)
Proteínas de la Cápside/genética , Melanoma/virología , Mutación , Parvovirus/genética , Proteínas Virales/genética , Animales , Cápside/química , Proteínas de la Cápside/química , Línea Celular , Evolución Molecular , Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/aislamiento & purificación , Células HEK293 , Humanos , Ratones , Modelos Moleculares , Infecciones por Parvoviridae/virología , Parvovirus/aislamiento & purificación , Parvovirus/patogenicidad , Selección Genética , Pase Seriado , Virulencia/genética , Replicación Viral/genética
5.
Virology ; 510: 216-223, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28750325

RESUMEN

In minute virus of mice (MVM) capsids, icosahedral five-fold channels serve as portals mediating genome packaging, genome release, and the phased extrusion of viral peptides. Previous studies suggest that residues L172 and V40 are essential for channel function. The structures of MVMi wildtype, and mutant L172T and V40A virus-like particles (VLPs) were solved from cryo-EM data. Two constriction points, termed the mid-gate and inner-gate, were observed in the channels of wildtype particles, involving residues L172 and V40 respectively. While the mid-gate of V40A VLPs appeared normal, in L172T adjacent channel walls were altered, and in both mutants there was major disruption of the inner-gate, demonstrating that direct L172:V40 bonding is essential for its structural integrity. In wildtype particles, residues from the N-termini of VP2 map into claw-like densities positioned below the channel opening, which become disordered in the mutants, implicating both L172 and V40 in the organization of VP2 N-termini.


Asunto(s)
Cápside/ultraestructura , Microscopía por Crioelectrón , Virus Diminuto del Ratón/ultraestructura , Mutación , Virosomas/ultraestructura
6.
Virology ; 476: 61-71, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25528417

RESUMEN

Members of the Parvoviridae family all encode a non-structural protein 1 (NS1) that directs replication of single-stranded viral DNA, packages viral DNA into capsid, and serves as a potent transcriptional activator. Here we report the X-ray structure of the minute virus of mice (MVM) NS1 N-terminal domain at 1.45Å resolution, showing that sites for dsDNA binding, ssDNA binding and cleavage, nuclear localization, and other functions are integrated on a canonical fold of the histidine-hydrophobic-histidine superfamily of nucleases, including elements specific for this Protoparvovirus but distinct from its Bocaparvovirus or Dependoparvovirus orthologs. High resolution structural analysis reveals a nickase active site with an architecture that allows highly versatile metal ligand binding. The structures support a unified mechanism of replication origin recognition for homotelomeric and heterotelomeric parvoviruses, mediated by a basic-residue-rich hairpin and an adjacent helix in the initiator proteins and by tandem tetranucleotide motifs in the replication origins.


Asunto(s)
Roturas del ADN de Cadena Simple , ADN Helicasas/química , ADN Helicasas/metabolismo , Virus Diminuto del Ratón/enzimología , Transactivadores/química , Transactivadores/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/química , Animales , Secuencia de Bases , ADN Helicasas/genética , Replicación del ADN , Ratones , Virus Diminuto del Ratón/química , Virus Diminuto del Ratón/genética , Modelos Moleculares , Infecciones por Parvoviridae/veterinaria , Infecciones por Parvoviridae/virología , Unión Proteica , Estructura Terciaria de Proteína , Origen de Réplica , Enfermedades de los Roedores/virología , Transactivadores/genética , Proteínas no Estructurales Virales/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
7.
Virology ; 468-470: 226-237, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25194919

RESUMEN

Parvoviruses encode a small number of ancillary proteins that differ substantially between genera. Within the genus Protoparvovirus, minute virus of mice (MVM) encodes three isoforms of its ancillary protein NS2, while human bocavirus 1 (HBoV1), in the genus Bocaparvovirus, encodes an NP1 protein that is unrelated in primary sequence to MVM NS2. To search for functional overlap between NS2 and NP1, we generated murine A9 cell populations that inducibly express HBoV1 NP1. These were used to test whether NP1 expression could complement specific defects resulting from depletion of MVM NS2 isoforms. NP1 induction had little impact on cell viability or cell cycle progression in uninfected cells, and was unable to complement late defects in MVM virion production associated with low NS2 levels. However, NP1 did relocate to MVM replication centers, and supports both the normal expansion of these foci and overcomes the early paralysis of DNA replication in NS2-null infections.


Asunto(s)
Regulación Viral de la Expresión Génica/fisiología , Prueba de Complementación Genética/métodos , Parvoviridae/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Supervivencia Celular , Replicación del ADN , ADN Viral , Humanos , Ratones , Parvoviridae/clasificación , Parvoviridae/genética , Especificidad de la Especie , Proteínas no Estructurales Virales/genética , Replicación Viral/fisiología
8.
Genome Announc ; 2(4)2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25081268

RESUMEN

The orphan parvovirus tumor virus X (TVX) has potent oncolytic activity. Compared to other viruses from the species Rodent protoparvovirus 1, TVX has a 111 nucleotide deletion in its nonstructural (NS) gene, a 24 nucleotide insertion in VP1, and a 93 nucleotide repeat initiating from the C-terminus of the capsid gene.

9.
Arch Virol ; 159(5): 1239-47, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24212889

RESUMEN

A set of proposals to rationalize and extend the taxonomy of the family Parvoviridae is currently under review by the International Committee on Taxonomy of Viruses (ICTV). Viruses in this family infect a wide range of hosts, as reflected by the longstanding division into two subfamilies: the Parvovirinae, which contains viruses that infect vertebrate hosts, and the Densovirinae, encompassing viruses that infect arthropod hosts. Using a modified definition for classification into the family that no longer demands isolation as long as the biological context is strong, but does require a near-complete DNA sequence, 134 new viruses and virus variants were identified. The proposals introduce new species and genera into both subfamilies, resolve one misclassified species, and improve taxonomic clarity by employing a series of systematic changes. These include identifying a precise level of sequence similarity required for viruses to belong to the same genus and decreasing the level of sequence similarity required for viruses to belong to the same species. These steps will facilitate recognition of the major phylogenetic branches within genera and eliminate the confusion caused by the near-identity of species and viruses. Changes to taxon nomenclature will establish numbered, non-Latinized binomial names for species, indicating genus affiliation and host range rather than recapitulating virus names. Also, affixes will be included in the names of genera to clarify subfamily affiliation and reduce the ambiguity that results from the vernacular use of "parvovirus" and "densovirus" to denote multiple taxon levels.


Asunto(s)
Parvoviridae/clasificación , Parvoviridae/genética , Genoma Viral , Filogenia , Especificidad de la Especie
10.
Annu Rev Virol ; 1(1): 517-37, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26958732

RESUMEN

Parvoviruses are small, rugged, nonenveloped protein particles containing a linear, nonpermuted, single-stranded DNA genome of ∼5 kb. Their limited coding potential requires optimal adaptation to the environment of particular host cells, where entry is mediated by a variable program of capsid dynamics, ultimately leading to genome ejection from intact particles within the host nucleus. Genomes are amplified by a continuous unidirectional strand-displacement mechanism, a linear adaptation of rolling circle replication that relies on the repeated folding and unfolding of small hairpin telomeres to reorient the advancing fork. Progeny genomes are propelled by the viral helicase into the preformed capsid via a pore at one of its icosahedral fivefold axes. Here we explore how the fine-tuning of this unique replication system and the mechanics that regulate opening and closing of the capsid fivefold portals have evolved in different viral lineages to create a remarkably complex spectrum of phenotypes.

11.
J Virol ; 87(19): 10501-14, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23903839

RESUMEN

The 121-nucleotide left-end telomere of Minute Virus of Mice (MVM) can be folded into a Y-shaped hairpin with short axial ears that are highly conserved within genus Parvovirus. To explore their potential role(s) during infection, we constructed infectious plasmid clones that lacked one or other ear. Although these were nonviable when transfected into A9 cells, excision of the viral genome and DNA amplification appeared normal, and viral transcripts and proteins were expressed, but progeny virion production was minimal, supporting the idea of a potential role for the ears in genome packaging. To circumvent the absence of progeny that confounded further analysis of these mutants, plasmids were transfected into 293T cells both with and without an adenovirus helper construct, generating single bursts of progeny. These virions bound to A9 cells and were internalized but failed to initiate viral transcription, protein expression, or DNA replication. No defects in mutant virion stability or function could be detected in vitro. Significantly, mutant capsid gene expression and DNA replication could be rescued by coinfection with wild-type virions carrying a replication-competent, capsid-gene-replacement vector. To pinpoint where such complementation occurred, prior transfection of plasmids expressing only MVM nonstructural proteins was explored. NS1 alone, but not NS2, rescued transcription and protein expression from both P4 and P38 promoters, whereas NS1 molecules deleted for their C-terminal transactivation domain did not. These results suggest that the mutant virions reach the nucleus, uncoat, and are converted to duplex DNA but require an intact left-end hairpin structure to form the initiating transcription complex.


Asunto(s)
Genoma Viral , Virus Diminuto del Ratón/genética , Infecciones por Parvoviridae/virología , Parvovirus/genética , Transcripción Genética , Replicación Viral , Animales , Emparejamiento Base , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Cartilla de ADN/química , Cartilla de ADN/genética , ADN Viral/genética , Vectores Genéticos , Ratones , Infecciones por Parvoviridae/genética , Infecciones por Parvoviridae/metabolismo , Transfección , Proteínas Virales/genética , Proteínas Virales/metabolismo
12.
Virology ; 442(1): 20-7, 2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23676303

RESUMEN

Engagement of innate viral sensors elicits a robust antiviral program via the induction of type I interferons (IFNs). Innate defense mechanisms against ssDNA viruses are not well defined. Here, we examine type I IFN induction and effectiveness in controlling a ssDNA virus. Using mouse embryonic fibroblasts (MEFs), we found that a murine parvovirus, minute virus of mice (MVMp), induced a delayed but significant IFN response. MEFs deficient in mitochondrial antiviral signaling protein (MAVS) mounted a wild-type IFN response to MVMp infection, indicating that RIG-I-dependent RNA intermediate recognition is not required for innate sensing of this virus. However, MVMp-induced IFNs, as well recombinant type I IFNs, were unable to inhibit viral replication. Finally, MVMp infected cells became unresponsive to Poly (I:C) stimulation. Together, these data suggest that the MVMp efficiently evades antiviral immune mechanisms imposed by type I IFNs, which may in part explain their efficient transmission between mice.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antivirales/inmunología , Fibroblastos/virología , Interferón Tipo I/inmunología , Virus Diminuto del Ratón/inmunología , Virus Diminuto del Ratón/patogenicidad , Infecciones por Parvoviridae/inmunología , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antivirales/metabolismo , Femenino , Fibroblastos/inmunología , Inmunidad Innata , Interferón Tipo I/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Virus Diminuto del Ratón/efectos de los fármacos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Infecciones por Parvoviridae/virología , ARN Polimerasa III , Receptores de Superficie Celular , Replicación Viral/inmunología
13.
J Virol ; 87(6): 3605-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23302877

RESUMEN

Toll-like receptor 9 (TLR9) recognizes genomes of double-stranded DNA (dsDNA) viruses in the endosome to stimulate plasmacytoid dendritic cells (pDCs). However, how and if viruses with single-stranded DNA (ssDNA) genomes are detected by pDCs remain unclear. Here we have shown that despite the ability of purified genomic DNA to stimulate TLR9 and despite the ability to enter TLR9 endosomes, ssDNA viruses of the Parvoviridae family failed to elicit an interferon (IFN) response in pDCs.


Asunto(s)
Células Dendríticas/inmunología , Evasión Inmune , Parvovirus/inmunología , Receptor Toll-Like 9/inmunología , Animales , Células Cultivadas , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL
14.
Artículo en Inglés | MEDLINE | ID: mdl-23293137

RESUMEN

Parvoviruses have a linear single-stranded DNA genome, around 5 kb in length, with short imperfect terminal palindromes that fold back on themselves to form duplex hairpin telomeres. These contain most of the cis-acting information required for viral "rolling hairpin" DNA replication, an evolutionary adaptation of rolling-circle synthesis in which the hairpins create duplex replication origins, prime complementary strand synthesis, and act as hinges to reverse the direction of the unidirectional cellular fork. Genomes are packaged vectorially into small, rugged protein capsids ~260 Å in diameter, which mediate their delivery directly into the cell nucleus, where they await their host cell's entry into S phase under its own cell cycle control. Here we focus on genus-specific variations in genome structure and replication, and review host cell responses that modulate the nuclear environment.


Asunto(s)
Daño del ADN , Parvovirus/genética , Replicación del ADN , ADN Viral/química , Variación Genética , Genoma Viral , Parvovirus/patogenicidad , Parvovirus/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
15.
J Virol ; 86(22): 12187-97, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22933276

RESUMEN

Parvoviral terminal hairpins are essential for viral DNA amplification but are also implicated in multiple additional steps in the viral life cycle. The palindromes at the two ends of the minute virus of mice (MVM) genome are dissimilar and are processed by different resolution mechanisms that selectively direct encapsidation of predominantly negative-sense progeny genomes and conserve a single Flip sequence orientation at the 3' (left) end of such progeny. The sequence and predicted structure of these 3' hairpins are highly conserved within the genus Parvovirus, exemplified by the 121-nucleotide left-end sequence of MVM, which folds into a Y-shaped hairpin containing small internal palindromes that form the "ears" of the Y. To explore the potential role(s) of this hairpin in the viral life cycle, we constructed infectious clones with the ear sequences either inverted, to give the antiparallel Flop orientation, or with multiple transversions, conserving their base composition but changing their sequence. These were compared with a "bubble" mutant, designed to activate the normally silent origin in the inboard arm of the hairpin, thus potentially rendering symmetric the otherwise asymmetric junction resolution mechanism that drives maintenance of Flip. This mutant exhibited a major defect in viral duplex and single-strand DNA replication, characterized by the accumulation of covalently closed turnaround forms of the left end, and was rapidly supplanted by revertants that restored asymmetry. In contrast, both sequence and orientation changes in the hairpin ears were tolerated, suggesting that maintaining the Flip orientation of these structures is a consequence of, but not the reason for, asymmetric left-end processing.


Asunto(s)
Genoma Viral , Virus Diminuto del Ratón/genética , Parvovirus/genética , Animales , Secuencia de Bases , Línea Celular , Replicación del ADN , ADN Viral , Cinética , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Mutación , Conformación de Ácido Nucleico , Plásmidos/metabolismo , Análisis de Secuencia de ADN , Replicación Viral/genética
16.
J Virol ; 86(1): 69-80, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22013064

RESUMEN

The linear single-stranded DNA genome of minute virus of mice can be ejected, in a 3'-to-5' direction, via a cation-linked uncoating reaction that leaves the 5' end of the DNA firmly complexed with its otherwise intact protein capsid. Here we compare the phenotypes of four mutants, L172T, V40A, N149A, and N170A, which perturb the base of cylinders surrounding the icosahedral 5-fold axes of the virus, and show that these structures are strongly implicated in 3'-to-5' release. Although noninfectious at 37°C, all mutants were viable at 32°C, showed a temperature-sensitive cell entry defect, and, after proteolysis of externalized VP2 N termini, were unable to protect the VP1 domain, which is essential for bilayer penetration. Mutant virus yields from multiple-round infections were low and were characterized by the accumulation of virions containing subgenomic DNAs of specific sizes. In V40A, these derived exclusively from the 5' end of the genome, indicative of 3'-to-5' uncoating, while L172T, the most impaired mutant, had long subgenomic DNAs originating from both termini, suggesting additional packaging portal defects. Compared to the wild type, genome release in vitro following cation depletion was enhanced for all mutants, while only L172T released DNA, in both directions, without cation depletion following proteolysis at 37°C. Analysis of progeny from single-round infections showed that uncoating did not occur during virion assembly, release, or extraction. However, unlike the wild type, the V40A mutant extensively uncoated during cell entry, indicating that the V40-L172 interaction restrains an uncoating trigger mechanism within the endosomal compartment.


Asunto(s)
Genoma Viral , Virus Diminuto del Ratón/fisiología , Mutación , Infecciones por Parvoviridae/veterinaria , Enfermedades de los Roedores/virología , Internalización del Virus , Replicación Viral , Animales , Línea Celular , ADN de Cadena Simple/química , ADN de Cadena Simple/genética , ADN Viral/química , ADN Viral/genética , Ratones , Virus Diminuto del Ratón/química , Virus Diminuto del Ratón/genética , Conformación de Ácido Nucleico , Infecciones por Parvoviridae/virología , Ensamble de Virus
17.
J Virol ; 85(10): 4822-7, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21367911

RESUMEN

The parvovirus minute virus of mice (MVM) packages a single copy of its linear single-stranded DNA genome into preformed capsids, in a process that is probably driven by a virus-encoded helicase. Parvoviruses have a roughly cylindrically shaped pore that surrounds each of the 12 5-fold vertices. The pore, which penetrates the virion shell, is created by the juxtaposition of 10 antiparallel ß-strands, two from each of the 5-fold-related capsid proteins. There is a bottleneck in the channel formed by the symmetry-related side chains of the leucines at position 172. We report here the X-ray crystal structure of the particles produced by a leucine-to-tryptophan mutation at position 172 and the analysis of its biochemical properties. The mutant capsid had its 5-fold channel blocked, and the particles were unable to package DNA, strongly suggesting that the 5-fold pore is the packaging portal for genome entry.


Asunto(s)
Cápside/metabolismo , Cápside/ultraestructura , ADN Viral/metabolismo , Virus Diminuto del Ratón/fisiología , Virus Diminuto del Ratón/ultraestructura , Ensamble de Virus , Sustitución de Aminoácidos/genética , Animales , Cápside/química , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Proteínas de la Cápside/ultraestructura , Cristalografía por Rayos X , Leucina/genética , Virus Diminuto del Ratón/química , Virus Diminuto del Ratón/genética , Mutación Missense , Estructura Cuaternaria de Proteína , Triptófano/genética
18.
Virology ; 410(2): 375-84, 2011 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-21193212

RESUMEN

MVM NS2 is essential for viral DNA amplification, but its mechanism of action is unknown. A classification scheme for autonomous parvovirus-associated replication (APAR) center development, based on NS1 distribution, was used to characterize abnormal APAR body maturation in NS2null mutant infections, and their organization examined for defects in host protein recruitment. Since acquisition of known replication factors appeared normal, we looked for differences in invoked DNA damage responses. We observed widespread association of H2AX/MDC1 damage response foci with viral replication centers, and sequestration and complex hyperphosphorylation of RPA(32), which occurred in wildtype and mutant infections. Quantifying these responses by western transfer indicated that both wildtype and NS2 mutant MVM elicited ATM activation, while phosphorylation of ATR, already basally activated in asynchronous A9 cells, was downregulated. We conclude that MVM infection invokes multiple damage responses that influence the APAR environment, but that NS2 does not modify the recruitment of cellular proteins.


Asunto(s)
Replicación del ADN , Interacciones Huésped-Patógeno , Virus Diminuto del Ratón/fisiología , Proteínas/metabolismo , Proteínas no Estructurales Virales/deficiencia , Replicación Viral , Animales , Línea Celular , Reparación del ADN , ADN Viral/genética , ADN Viral/metabolismo , Ratones , Virus Diminuto del Ratón/genética
19.
J Virol ; 84(4): 1945-56, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19955311

RESUMEN

We describe a structural rearrangement that can occur in parvovirus minute virus of mice (MVMp) virions following prolonged exposure to buffers containing 0.5 mM EDTA. Such particles remain stable at 4 degrees C but undergo a conformational shift upon heating to 37 degrees C at pH 7.2 that leads to the ejection of much of the viral genome in a 3'-to-5' direction, leaving the DNA tightly associated with the otherwise intact capsid. This rearrangement can be prevented by the addition of 1 mM CaCl(2) or MgCl(2) prior to incubation at 37 degrees C, suggesting that readily accessible divalent cation binding sites in the particle are critical for genome retention. Uncoating was not seen following the incubation of virions at pH 5.5 and 37 degrees C or at pH 7.2 and 37 degrees C in particles with subgenomic DNA, suggesting that pressure exerted by the full-length genome may influence this process. Uncoated genomes support complementary-strand synthesis by T7 DNA polymerase, but synthesis aborts upstream of the right-hand end, which remains capsid associated. We conclude that viral genomes are positioned so that their 3' termini and coding sequences can be released from intact particles at physiological temperatures by a limited conformational rearrangement. In the presence of divalent cations, incremental heating between 45 degrees C and 65 degrees C induces structural transitions that first lead to the extrusion of VP1 N termini, followed by genome exposure. However, in cation-depleted virions, the sequence of these shifts is blurred. Moreover, cation-depleted particles that have been induced to eject their genomes at 37 degrees C continue to sequester their VP1 N termini within the intact capsid, suggesting that these two extrusion events represent separable processes.


Asunto(s)
Virus Diminuto del Ratón/genética , Virus Diminuto del Ratón/fisiología , Animales , Cápside/fisiología , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/fisiología , Cationes Bivalentes/metabolismo , Línea Celular , Quelantes , ADN Viral/biosíntesis , ADN Viral/genética , Genoma Viral , Concentración de Iones de Hidrógeno , Ratones , Virión/genética , Virión/fisiología , Replicación Viral/genética , Replicación Viral/fisiología
20.
J Infect Dis ; 198(1): 41-50, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18491974

RESUMEN

BACKGROUND: Human bocavirus (HBoV) is a newly identified human parvovirus for which seroepidemiology and antigenic properties remain undefined. METHODS: The HBoV VP2 gene, expressed from a baculovirus vector, produced virus-like particles (VLPs), which were used to raise rabbit anti-HBoV antisera and to develop an enzyme-linked immunosorbent assay (ELISA). The VLP-based ELISA was used to screen for HBoV-specific immunoglobulin G antibodies in a convenience sample of 270 serum specimens, mostly from children, obtained at Yale-New Haven Hospital; 208 specimens were also screened for erythrovirus B19-specific antibodies by a B19 VLP-based ELISA. RESULTS: Immunofluorescence and ELISA showed that human parvoviruses HBoV and B19 are antigenically distinct. By the HBoV VLP-based ELISA, 91.8% and 63.6% of serum specimens from infants in the first and second months of life, respectively, were found to be seropositive, as were 45.4% from 3-month-old infants and 25.0% from 4-month-old infants. The percentages of HBoV-seropositive children increased to 40.7%-60.0% for children 5-47 months of age and to >85% for individuals >or=48 months old. However, the overall percentage of B19-seropositive individuals was <40.5% for all age groups screened. CONCLUSIONS: HBoV infection is common during childhood, but a minority of children and young adults screened have evidence of B19 infection.


Asunto(s)
Bocavirus/aislamiento & purificación , Proteínas de la Cápside/genética , Infecciones por Parvoviridae/epidemiología , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Bocavirus/genética , Bocavirus/inmunología , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Línea Celular , Niño , Preescolar , Humanos , Inmunoprecipitación , Lactante , Datos de Secuencia Molecular , Infecciones por Parvoviridae/diagnóstico , Infecciones por Parvoviridae/inmunología , Parvovirus B19 Humano/inmunología , Parvovirus B19 Humano/aislamiento & purificación , Conejos , Proteínas Recombinantes , Estudios Seroepidemiológicos , Virión/genética , Virión/inmunología , Virión/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...