Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.589
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 151(2): 253-66, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-23063120

RESUMO

Pathogenic simian immunodeficiency virus (SIV) infection is associated with enteropathy, which likely contributes to AIDS progression. To identify candidate etiologies for AIDS enteropathy, we used next-generation sequencing to define the enteric virome during SIV infection in nonhuman primates. Pathogenic, but not nonpathogenic, SIV infection was associated with significant expansion of the enteric virome. We identified at least 32 previously undescribed enteric viruses during pathogenic SIV infection and confirmed their presence by using viral culture and PCR testing. We detected unsuspected mucosal adenovirus infection associated with enteritis as well as parvovirus viremia in animals with advanced AIDS, indicating the pathogenic potential of SIV-associated expansion of the enteric virome. No association between pathogenic SIV infection and the family-level taxonomy of enteric bacteria was detected. Thus, enteric viral infections may contribute to AIDS enteropathy and disease progression. These findings underline the importance of metagenomic analysis of the virome for understanding AIDS pathogenesis.


Assuntos
Caliciviridae/isolamento & purificação , Intestinos/virologia , Parvoviridae/isolamento & purificação , Picornaviridae/isolamento & purificação , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Caliciviridae/classificação , Caliciviridae/genética , Chlorocebus aethiops , Fezes/microbiologia , Fezes/virologia , Intestinos/microbiologia , Dados de Sequência Molecular , Parvoviridae/classificação , Parvoviridae/genética , Filogenia , Picornaviridae/classificação , Picornaviridae/genética , Reação em Cadeia da Polimerase , Síndrome de Imunodeficiência Adquirida dos Símios/microbiologia , Vírus da Imunodeficiência Símia/patogenicidade
2.
J Virol ; 97(7): e0016123, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37367301

RESUMO

Parvoviruses are among the smallest and superficially simplest animal viruses, infecting a broad range of hosts, including humans, and causing some deadly infections. In 1990, the first atomic structure of the canine parvovirus (CPV) capsid revealed a 26-nm-diameter T=1 particle made up of two or three versions of a single protein, and packaging about 5,100 nucleotides of single-stranded DNA. Our structural and functional understanding of parvovirus capsids and their ligands has increased as imaging and molecular techniques have advanced, and capsid structures for most groups within the Parvoviridae family have now been determined. Despite those advances, significant questions remain unanswered about the functioning of those viral capsids and their roles in release, transmission, or cellular infection. In addition, the interactions of capsids with host receptors, antibodies, or other biological components are also still incompletely understood. The parvovirus capsid's apparent simplicity likely conceals important functions carried out by small, transient, or asymmetric structures. Here, we highlight some remaining open questions that may need to be answered to provide a more thorough understanding of how these viruses carry out their various functions. The many different members of the family Parvoviridae share a capsid architecture, and while many functions are likely similar, others may differ in detail. Many of those parvoviruses have not been experimentally examined in detail (or at all in some cases), so we, therefore, focus this minireview on the widely studied protoparvoviruses, as well as the most thoroughly investigated examples of adeno-associated viruses.


Assuntos
Parvoviridae , Animais , Humanos , Capsídeo/ultraestrutura , Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , DNA Viral/metabolismo , Parvoviridae/genética , Parvoviridae/ultraestrutura , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Dependovirus/genética , Dependovirus/metabolismo , Dependovirus/ultraestrutura
3.
PLoS Pathog ; 16(8): e1008718, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32797103

RESUMO

APOBEC3 enzymes are innate immune effectors that introduce mutations into viral genomes. These enzymes are cytidine deaminases which transform cytosine into uracil. They preferentially mutate cytidine preceded by thymidine making the 5'TC motif their favored target. Viruses have evolved different strategies to evade APOBEC3 restriction. Certain viruses actively encode viral proteins antagonizing the APOBEC3s, others passively face the APOBEC3 selection pressure thanks to a depleted genome for APOBEC3-targeted motifs. Hence, the APOBEC3s left on the genome of certain viruses an evolutionary footprint. The aim of our study is the identification of these viruses having a genome shaped by the APOBEC3s. We analyzed the genome of 33,400 human viruses for the depletion of APOBEC3-favored motifs. We demonstrate that the APOBEC3 selection pressure impacts at least 22% of all currently annotated human viral species. The papillomaviridae and polyomaviridae are the most intensively footprinted families; evidencing a selection pressure acting genome-wide and on both strands. Members of the parvoviridae family are differentially targeted in term of both magnitude and localization of the footprint. Interestingly, a massive APOBEC3 footprint is present on both strands of the B19 erythroparvovirus; making this viral genome one of the most cleaned sequences for APOBEC3-favored motifs. We also identified the endemic coronaviridae as significantly footprinted. Interestingly, no such footprint has been detected on the zoonotic MERS-CoV, SARS-CoV-1 and SARS-CoV-2 coronaviruses. In addition to viruses that are footprinted genome-wide, certain viruses are footprinted only on very short sections of their genome. That is the case for the gamma-herpesviridae and adenoviridae where the footprint is localized on the lytic origins of replication. A mild footprint can also be detected on the negative strand of the reverse transcribing HIV-1, HIV-2, HTLV-1 and HBV viruses. Together, our data illustrate the extent of the APOBEC3 selection pressure on the human viruses and identify new putatively APOBEC3-targeted viruses.


Assuntos
Citidina Desaminase/metabolismo , Genoma Viral/genética , Interações Hospedeiro-Patógeno/genética , Seleção Genética/genética , Replicação Viral/genética , Desaminases APOBEC , Coronaviridae/genética , Humanos , Imunidade Inata/imunologia , Papillomaviridae/genética , Parvoviridae/genética , Polyomaviridae/genética , Proteínas Virais/genética
4.
Arch Virol ; 167(4): 1163-1167, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35278130

RESUMO

In this study, a novel parvovirus (zander/M5/2015/HUN, OK236393) was detected in faecal specimens from a fish - zander or pikeperch (Sander lucioperca) - and genetically characterized using viral metagenomics and PCR methods. The NS1 and VP1 proteins of zander/M5/2015/HUN share <30% aa sequence identity, respectively, with the corresponding proteins of known members of the family Parvoviridae. Out of 62 faecal specimens collected from 13 freshwater fish species, three (4.8%) samples were positive by PCR for the novel parvovirus - all from zander. This is the second parvovirus detected in fish - after the disease-causing tilapia parvovirus of the subfamily Hamaparvovirinae - and it potentially represents a novel genus in the subfamily Parvovirinae.


Assuntos
Infecções por Parvoviridae , Parvoviridae , Parvovirinae , Parvovirus , Animais , Água Doce , Infecções por Parvoviridae/veterinária , Parvovirus/genética
5.
Arch Virol ; 167(5): 1307-1310, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35355143

RESUMO

In this study, genetic counterparts of the human-stool-associated tusavirus (subfamily Parvovirinae, family Parvoviridae) with >97% and 95-100% amino acid sequence identity in the parvoviral NS1 and VP1 protein were identified in faecal specimens from domestic goats (Capra hircus) and sheep (Ovis aries) in Hungary. Eleven (17.8%) of the 62 faecal specimens from goats and 12 (25.5%) of the 47 from sheep both from less than 12 months old animals were positive for tusavirus DNA by PCR, while none of the specimens collected from cattle and swine were positive. Thus, it cannot be ruled out that tusavirus infection in humans is of zoonotic origin.


Assuntos
Parvoviridae , Parvovirinae , Parvovirus , Animais , Bovinos , Fezes , Cabras , Humanos , Ovinos , Suínos
6.
J Virol ; 94(18)2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32581107

RESUMO

Wild birds are major natural reservoirs and potential dispersers of a variety of infectious diseases. As such, it is important to determine the diversity of viruses they carry and use this information to help understand the potential risks of spillover to humans, domestic animals, and other wildlife. We investigated the potential viral causes of paresis in long-standing, but undiagnosed, disease syndromes in wild Australian birds. RNA from diseased birds was extracted and pooled based on tissue type, host species, and clinical manifestation for metagenomic sequencing. Using a bulk and unbiased metatranscriptomic approach, combined with clinical investigation and histopathology, we identified a number of novel viruses from the families Astroviridae, Adenoviridae, Picornaviridae, Polyomaviridae, Paramyxoviridae, Parvoviridae, and Circoviridae in common urban wild birds, including Australian magpies, magpie larks, pied currawongs, Australian ravens, and rainbow lorikeets. In each case, the presence of the virus was confirmed by reverse transcription (RT)-PCR. These data revealed a number of candidate viral pathogens that may contribute to coronary, skeletal muscle, vascular, and neuropathology in birds of the Corvidae and Artamidae families and neuropathology in members of the Psittaculidae The existence of such a diverse virome in urban avian species highlights the importance and challenges in elucidating the etiology and ecology of wildlife pathogens in urban environments. This information will be increasingly important for managing disease risks and conducting surveillance for potential viral threats to wildlife, livestock, and human health.IMPORTANCE Wildlife naturally harbor a diverse array of infectious microorganisms and can be a source of novel diseases in domestic animals and human populations. Using unbiased RNA sequencing, we identified highly diverse viruses in native birds from Australian urban environments presenting with paresis. This research included the clinical investigation and description of poorly understood recurring syndromes of unknown etiology: clenched claw syndrome and black and white bird disease. As well as identifying a range of potentially disease-causing viral pathogens, this study describes methods that can effectively and efficiently characterize emergent disease syndromes in free-ranging wildlife and promotes further surveillance for specific pathogens of potential conservation and zoonotic concern.


Assuntos
Animais Selvagens/virologia , Doenças das Aves/epidemiologia , Aves/virologia , Infecções por Vírus de DNA/veterinária , Metagenoma , Infecções por Vírus de RNA/veterinária , Transcriptoma , Adenoviridae/classificação , Adenoviridae/genética , Adenoviridae/isolamento & purificação , Animais , Astroviridae/classificação , Astroviridae/genética , Astroviridae/isolamento & purificação , Austrália/epidemiologia , Doenças das Aves/virologia , Circoviridae/classificação , Circoviridae/genética , Circoviridae/isolamento & purificação , Cidades , Infecções por Vírus de DNA/epidemiologia , Infecções por Vírus de DNA/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Paramyxoviridae/classificação , Paramyxoviridae/genética , Paramyxoviridae/isolamento & purificação , Parvoviridae/classificação , Parvoviridae/genética , Parvoviridae/isolamento & purificação , Filogenia , Picornaviridae/classificação , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Polyomaviridae/classificação , Polyomaviridae/genética , Polyomaviridae/isolamento & purificação , Infecções por Vírus de RNA/epidemiologia , Infecções por Vírus de RNA/virologia
7.
Hepatology ; 72(6): 2182-2196, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32654263

RESUMO

BACKGROUND AND AIMS: Alcoholic hepatitis (AH) is a severe manifestation of alcohol-associated liver disease (ALD) with high mortality. Although gut bacteria and fungi modulate disease severity, little is known about the effects of the viral microbiome (virome) in patients with ALD. APPROACH AND RESULTS: We extracted virus-like particles from 89 patients with AH who were enrolled in a multicenter observational study, 36 with alcohol use disorder (AUD), and 17 persons without AUD (controls). Virus-like particles from fecal samples were fractionated using differential filtration techniques, and metagenomic sequencing was performed to characterize intestinal viromes. We observed an increased viral diversity in fecal samples from patients with ALD, with the most significant changes in samples from patients with AH. Escherichia-, Enterobacteria-, and Enterococcus phages were over-represented in fecal samples from patients with AH, along with significant increases in mammalian viruses such as Parvoviridae and Herpesviridae. Antibiotic treatment was associated with higher viral diversity. Specific viral taxa, such as Staphylococcus phages and Herpesviridae, were associated with increased disease severity, indicated by a higher median Model for End-Stage Liver Disease score, and associated with increased 90-day mortality. CONCLUSIONS: In conclusion, intestinal viral taxa are altered in fecal samples from patients with AH and associated with disease severity and mortality. Our study describes an intestinal virome signature associated with AH.


Assuntos
Doença Hepática Terminal/virologia , Hepatite Alcoólica/virologia , Mucosa Intestinal/virologia , Cirrose Hepática/virologia , Viroma/genética , Adulto , Idoso , Animais , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Estudos de Casos e Controles , DNA Viral/isolamento & purificação , Doença Hepática Terminal/diagnóstico , Doença Hepática Terminal/mortalidade , Doença Hepática Terminal/terapia , Fezes/virologia , Feminino , Hepatite Alcoólica/diagnóstico , Hepatite Alcoólica/mortalidade , Hepatite Alcoólica/terapia , Herpesviridae/genética , Herpesviridae/isolamento & purificação , Humanos , Fígado/patologia , Cirrose Hepática/diagnóstico , Cirrose Hepática/mortalidade , Cirrose Hepática/terapia , Masculino , Metagenômica , Pessoa de Meia-Idade , Parvoviridae/genética , Parvoviridae/isolamento & purificação , RNA Viral/isolamento & purificação , Índice de Gravidade de Doença , Taxa de Sobrevida
8.
Virol J ; 18(1): 210, 2021 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-34689822

RESUMO

In line with the Latin expression "sed parva forti" meaning "small but mighty," the family Parvoviridae contains many of the smallest known viruses, some of which result in fatal or debilitating infections. In recent years, advances in metagenomic viral discovery techniques have dramatically increased the identification of novel parvoviruses in both diseased and healthy individuals. While some of these discoveries have solved etiologic mysteries of well-described diseases in animals, many of the newly discovered parvoviruses appear to cause mild or no disease, or disease associations remain to be established. With the increased use of animal parvoviruses as vectors for gene therapy and oncolytic treatments in humans, it becomes all the more important to understand the diversity, pathogenic potential, and evolution of this diverse family of viruses. In this review, we discuss parvoviruses infecting vertebrate animals, with a special focus on pathogens of veterinary significance and viruses discovered within the last four years.


Assuntos
Infecções por Parvoviridae , Parvoviridae , Parvovirus , Animais , Metagenômica , Parvoviridae/genética , Infecções por Parvoviridae/veterinária , Parvovirus/genética , Filogenia
9.
J Gen Virol ; 101(2): 226-239, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31855134

RESUMO

Diaphorina citri densovirus (DcDV) is an ambisense densovirus with a 5071 nt genome. Phylogenetic analysis places DcDV in an intermediate position between those in the Ambidensovirus and Iteradensovirus genera, a finding that is consistent with the observation that DcDV possesses an Iteradensoviris-like non-structural (NS) protein-gene cassette, but a capsid-protein (VP) gene cassette resembling those of other ambisense densoviruses. DcDV is maternally transmitted to 100 % of the progeny of infected female Diaphorina citri, and the progeny of infected females carry DcDV as a persistent infection without outward phenotypic effects. We were unable to infect naïve individuals by oral inoculation, however low levels of transient viral replication are detected following intrathoracic injection of DcDV virions into uninfected D. citri insects. Transcript mapping indicates that DcDV produces one transcript each from the NS and VP gene cassettes and that these transcripts are polyadenylated at internal sites to produce a ~2.2 kb transcript encoding the NS proteins and a ~2.4 kb transcript encoding the VP proteins. Additionally, we found that transcriptional readthrough leads to the production of longer non-canonical transcripts from both genomic strands.


Assuntos
Densovirus , Genoma Viral , Hemípteros/virologia , Viroses/transmissão , Animais , Proteínas do Capsídeo/genética , Classificação , Vírus de DNA/genética , Densovirus/classificação , Densovirus/genética , Densovirus/isolamento & purificação , Genes Virais , Transmissão Vertical de Doenças Infecciosas , Vírus de Insetos/classificação , Parvoviridae/classificação , Filogenia , Proteínas não Estruturais Virais/genética , Proteínas Virais/genética
10.
Arch Virol ; 165(9): 2133-2146, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32533329

RESUMO

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.


Assuntos
Infecções por Parvoviridae/veterinária , Infecções por Parvoviridae/virologia , Parvoviridae/classificação , Parvoviridae/fisiologia , Filogenia , Animais , Especificidade de Hospedeiro , Humanos , Parvoviridae/genética , Parvoviridae/isolamento & purificação , Proteínas Virais/genética
11.
J Gen Virol ; 100(3): 367-368, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30672729

RESUMO

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.


Assuntos
Infecções por Parvoviridae/virologia , Parvoviridae/classificação , Filogenia , Animais , Genoma Viral , Humanos , Parvoviridae/genética , Parvoviridae/isolamento & purificação , Parvoviridae/ultraestrutura , Virologia/organização & administração
12.
Mol Cell Probes ; 48: 101448, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31521579

RESUMO

Amdoparvoviruses infect carnivore species, including mink, raccoon dog, fox, skunk, and red panda. Amdoparvovirus infection is a major cause of morbidity and mortality in farmed minks. Here, we developed a direct TaqMan qPCR assay for detection and quantification of carnivore amdoparvoviruses by using three primers and one probe based on the conserved VP2 gene. The detection limit for Aleutian mink disease virus (AMDV) and Raccoon dog and arctic fox amdoparvovirus (RFAV) were 4.06 × 101 copies/µl and 2.93 × 101 copies/µl, respectively. Both intra- and inter-assay variability were less than 2%. Among 74 carnivore samples, the positive rates for amdoparvoviruses were 62.2% (46/74) by direct TaqMan qPCR, while only 40.5% (30/74) by SYBR Green I qPCR. This result suggests that the direct TaqMan qPCR was more sensitive than the SYBR Green I qPCR. Additionally, the direct TaqMan qPCR is a rapid and sensitive method for liquid samples at microliter level as the assay employed the direct alkaline lysis method to obtain viral DNA and, therefore, eliminated the cumbersome steps in extracting DNA. Overall, the direct TaqMan qPCR assay possessed high specificity, sensitivity, and reproducibility, indicating that it can be used as a powerful tool for detection and quantification of various carnivore amdoparvoviruses in epidemiological and pathogenesis studies.


Assuntos
Vírus da Doença Aleutiana do Vison/genética , Parvoviridae/genética , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , Primers do DNA/genética , DNA Viral/genética , Cães , Raposas/virologia , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Análise de Sequência de DNA/métodos
13.
Biologicals ; 61: 1-7, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31447377

RESUMO

Horses are often used as blood donors for commercial horse serum (HS) production and to manufacture biologicals. HS is an alternative for fetal bovine serum (FBS) used as a supplement for cell culture and vaccine production. Furthermore, HS is also frequently obtained in order to produce antisera toxins and pathogens. The advent of high-throughput sequencing (HTS) has promoted changes in virus detection, since previous knowledge of targets is not required. Thus, the present study aimed to describe the virome of five different batches of commercial HS from New Zealand (three batches) and Brazil and the United States (one batch each) using HTS. Each HS pool were processed and sequenced using an Illumina MiSeq platform. Sequences-related to viruses belonging to the Flaviviridae, Herpesviridae, and Parvoviridae families were detected. Particularly, equine hepacivirus (EqHV), equine pegivirus (EPgV), and Theiler's disease-associated virus (TDAV) were more frequent found in the batches analyzed. The presence of viral genomes in cell culture sera illustrates that these commercial sera can contain a mixture of different viruses and, therefore, can be regarded as potentially infectious for susceptible hosts. Moreover, the innocuity of commercial HS is important for the efficiency and security of diagnostics and the production of biological products.


Assuntos
Flaviviridae/genética , Genoma Viral , Herpesviridae/genética , Cavalos/virologia , Parvoviridae/genética , Soro/virologia , Animais , Meios de Cultura , Flaviviridae/classificação , Herpesviridae/classificação , Cavalos/sangue , Parvoviridae/classificação
14.
Mem Inst Oswaldo Cruz ; 114: e190198, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31596309

RESUMO

BACKGROUND: In Brazil the implementation of the Sentinel Surveillance System of Influenza began in 2000. Central public health laboratories use reverse transcription-quantitative polymerase chain reaction (RT-qPCR) for diagnosis of respiratory viruses, but this protocol identifies only specific targets, resulted in inconclusive diagnosis for many samples. Thus, high-throughput sequencing (HTS) would be complementary method in the identification of pathogens in inconclusive samples for RT-qPCR or other specific detection protocols. OBJECTIVES: This study aimed to detect unidentified viruses using HTS approach in negative samples of nasopharynx/tracheal secretions by the standard RT-qPCR collected in the Federal District, Brazil. METHODS: Nucleic acids were extracted from samples collected in winter period of 2016 and subjected to HTS. The results were confirmed by the multiplex PR21 RT-qPCR, which identifies 21 respiratory pathogens. FINDINGS: The main viruses identified by HTS were of families Herpesviridae, Coronaviridae, Parvoviridae and Picornaviridae, with the emphasis on rhinoviruses. The presence of respiratory viruses in the samples was confirmed by the PR21 multiplex RT-qPCR. Coronavirus, enterovirus, bocavirus and rhinovirus were found by multiplex RT-qPCR as well as by HTS analyses. MAIN CONCLUSIONS: Wide virus diversity was found by different methodologies and high frequency of rhinovirus occurrence was confirmed in population in winter, showing its relevance for public health.


Assuntos
Coronaviridae/isolamento & purificação , Herpesviridae/isolamento & purificação , Nasofaringe/virologia , Parvoviridae/isolamento & purificação , Picornaviridae/isolamento & purificação , Traqueia/virologia , Coronaviridae/classificação , Coronaviridae/genética , DNA Viral/genética , Herpesviridae/classificação , Herpesviridae/genética , Humanos , Parvoviridae/classificação , Parvoviridae/genética , Picornaviridae/classificação , Picornaviridae/genética , RNA Viral/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Virol J ; 13: 133, 2016 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-27473724

RESUMO

Chapparvovirus, a recently determined new genus in the family Parvoviridae, can infect many species of animals including bats, chickens, and pigs. Here, using viral metagenomics method, we identified a novel Chapparvovirus from feces of wild rats and designated it as rat parvovirus 2 (RPV2). The nearly complete genome of RPV2 is 4222-nt long and includes two ORFs encoding a 654-aa nonstructural protein 1 (NS1) and a 472-aa capsid protein (VP), respectively. Phylogenetic analysis over the amino acid sequence of the NS1 showed that RPV2 clustered with Eidolon helvum parvovirus 2 (EHPV2), porcine parvovirus 7 (PPV7), and turkey parvovirus 1 (TP1), forming a separate clade. Sequence analysis indicated that the NS1 protein of RPV2 shared the highest amino acid sequence identity (51 %) with that of EHPV2. According to the genetic distance-based criteria, RPV2 identified here belongs to a novel species of Chapparvovirus.


Assuntos
Infecções por Parvoviridae/veterinária , Parvoviridae/isolamento & purificação , Doenças dos Roedores/virologia , Animais , Animais Selvagens/virologia , Galinhas , China , Fezes/virologia , Genoma Viral , Fases de Leitura Aberta , Parvoviridae/classificação , Parvoviridae/genética , Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Filogenia , Ratos , Suínos , Proteínas Virais/genética
17.
Proc Natl Acad Sci U S A ; 110(25): 10264-9, 2013 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-23716702

RESUMO

Seronegative hepatitis--non-A, non-B, non-C, non-D, non-E hepatitis--is poorly characterized but strongly associated with serious complications. We collected 92 sera specimens from patients with non-A-E hepatitis in Chongqing, China between 1999 and 2007. Ten sera pools were screened by Solexa deep sequencing. We discovered a 3,780-bp contig present in all 10 pools that yielded BLASTx E scores of 7e-05-0.008 against parvoviruses. The complete sequence of the in silico-assembled 3,780-bp contig was confirmed by gene amplification of overlapping regions over almost the entire genome, and the virus was provisionally designated NIH-CQV. Further analysis revealed that the contig was composed of two major ORFs. By protein BLAST, ORF1 and ORF2 were most homologous to the replication-associated protein of bat circovirus and the capsid protein of porcine parvovirus, respectively. Phylogenetic analysis indicated that NIH-CQV is located at the interface of Parvoviridae and Circoviridae. Prevalence of NIH-CQV in patients was determined by quantitative PCR. Sixty-three of 90 patient samples (70%) were positive, but all those from 45 healthy controls were negative. Average virus titer in the patient specimens was 1.05 e4 copies/µL. Specific antibodies against NIH-CQV were sought by immunoblotting. Eighty-four percent of patients were positive for IgG, and 31% were positive for IgM; in contrast, 78% of healthy controls were positive for IgG, but all were negative for IgM. Although more work is needed to determine the etiologic role of NIH-CQV in human disease, our data indicate that a parvovirus-like virus is highly prevalent in a cohort of patients with non-A-E hepatitis.


Assuntos
Anemia Aplástica/epidemiologia , Anemia Aplástica/virologia , Povo Asiático/estatística & dados numéricos , DNA Viral/genética , Hepatite Viral Humana/epidemiologia , Hepatite Viral Humana/virologia , Adolescente , Adulto , Idoso , Criança , China/epidemiologia , Circoviridae/genética , Evolução Molecular , Feminino , Anticorpos Anti-Hepatite/sangue , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Parvoviridae/genética , Filogenia , Prevalência , Fatores de Risco , Estudos Soroepidemiológicos , Adulto Jovem
18.
Virol J ; 12: 114, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26239432

RESUMO

The parvoviruses are widely spread in many species and are among the smallest DNA animal viruses. The parvovirus is composed of a single strand molecule of DNA wrapped into an icosahedral capsid. In a viral infection, the massy capsid participates in the entire viral infection process, which is summarized in this review. The capsid protein VP1 is primarily responsible for the infectivity of the virus, and the nuclear localization signal (NLS) of the VP1 serves as a guide to assist the viral genome in locating the nucleus. The dominant protein VP2 provides an "anti-receptor", which interacts with the cellular receptor and leads to the further internalization of virus, and, the N-terminal of VP2 also cooperates with the VP1 to prompt the process of nucleus translocation. Additionally, a cleavage protein VP3 is a part of the capsid, which exists only in several members of the parvovirus family; however, the function of this cleavage protein remains to be fully determined. Parvoviruses can suffer from the extreme environmental conditions such as low pH, or even escape from the recognition of pattern recognition receptors (PRRs), due to the protection of the stable capsid, which is thought to be an immune escape mechanism. The applications of the capsid proteins to the screening and the treatment of diseases are also discussed. The processes of viral infection should be noted, because understanding the virus-host interactions will contribute to the development of therapeutic vaccines.


Assuntos
Proteínas do Capsídeo/metabolismo , Infecções por Parvoviridae/virologia , Parvoviridae/metabolismo , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Núcleo Celular/metabolismo , Genoma Viral , Humanos , Fases de Leitura Aberta , Parvoviridae/genética , Transporte Proteico
19.
Arch Virol ; 160(10): 2617-21, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26239342

RESUMO

Waterfowl parvoviruses are divided into Muscovy duck parvoviruses (MDPVs) and goose parvoviruses (GPVs). Phylogenetic analysis based on structural gene nucleotide sequences showed that the strains of three GPVs (DY, PT and D strains) and two MDPVs (GX5 and SAAH-SHNH) are closely related and formed one cluster. Recombination analysis showed that recombination between GPV-GDFsh and MDPV-89384/FRANCE strains led to five recombinant strains: GPV-DY, GPV-PT, GPV-D, MDPV-GX5 and MDPV-SAAH-SHNH. The recombinant event was confirmed using the Simplot program and phylogenetic analysis. This is the first comprehensive investigation of recombination between MDPV and GPV structural genes.


Assuntos
Infecções por Parvoviridae/veterinária , Parvoviridae/genética , Doenças das Aves Domésticas/virologia , Recombinação Genética , Proteínas Estruturais Virais/genética , Animais , Patos , França , Gansos , Dados de Sequência Molecular , Parvoviridae/classificação , Parvoviridae/isolamento & purificação , Infecções por Parvoviridae/virologia , Filogenia
20.
Arch Virol ; 160(8): 2111-5, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26085285

RESUMO

To investigate the possible role of recombination in the evolution of Muscovy duck parvovirus (MDPV) and goose parvovirus (GPV) in Taiwan, we analyzed a potentially significant recombination event that occurred only in GPV by comparing thirteen complete sequences of the capsid gene VP2 of GPV and MDPV. The recombination event occurred between GPV strain 06-0239 as the minor parent and strains 99-0808 as the major parent, which resulted in the GPV recombinant V325/TW03. GPV V325/TW03 is likely to represent a new genotype among the Taiwanese GPV strains. This represents the first evidence that intergenotype recombination within the VP2 gene cluster contributes to the genetic diversity of the VP2 genes of Taiwanese GPV field strains.


Assuntos
Proteínas do Capsídeo/genética , Gansos/virologia , Infecções por Parvoviridae/veterinária , Parvoviridae/genética , Doenças das Aves Domésticas/virologia , Recombinação Genética , Animais , Dados de Sequência Molecular , Parvoviridae/classificação , Parvoviridae/isolamento & purificação , Infecções por Parvoviridae/virologia , Filogenia , Taiwan
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA