Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 364
Filtrar
1.
Viruses ; 16(5)2024 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-38793587

RESUMO

A massive mortality event concerning farmed Chinese tongue soles occurred in Tianjin, China, and the causative agent remains unknown. Here, a novel Cynoglossus semilaevis papillomavirus (CsPaV) and parvovirus (CsPV) were simultaneously isolated and identified from diseased fish via electron microscopy, virus isolation, genome sequencing, experimental challenges, and fluorescence in situ hybridization (FISH). Electron microscopy showed large numbers of virus particles present in the tissues of diseased fish. Viruses that were isolated and propagated in flounder gill cells (FG) induced typical cytopathic effects (CPE). The cumulative mortality of fish given intraperitoneal injections reached 100% at 7 dpi. The complete genomes of CsPaV and CsPV comprised 5939 bp and 3663 bp, respectively, and the genomes shared no nucleotide sequence similarities with other viruses. Phylogenetic analysis based on the L1 and NS1 protein sequences revealed that CsPaV and CsPV were novel members of the Papillomaviridae and Parvoviridae families. The FISH results showed positive signals in the spleen tissues of infected fish, and both viruses could co-infect single cells. This study represents the first report where novel papillomavirus and parvovirus are identified in farmed marine cultured fish, and it provides a basis for further studies on the prevention and treatment of emerging viral diseases.


Assuntos
Doenças dos Peixes , Linguados , Genoma Viral , Papillomaviridae , Infecções por Parvoviridae , Parvovirus , Filogenia , Animais , Doenças dos Peixes/virologia , Doenças dos Peixes/mortalidade , China , Linguados/virologia , Infecções por Parvoviridae/veterinária , Infecções por Parvoviridae/virologia , Parvovirus/genética , Parvovirus/isolamento & purificação , Parvovirus/classificação , Papillomaviridae/genética , Papillomaviridae/isolamento & purificação , Papillomaviridae/classificação , Infecções por Papillomavirus/virologia , Infecções por Papillomavirus/veterinária , Hibridização in Situ Fluorescente
2.
PLoS One ; 19(2): e0298742, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38412152

RESUMO

Subclinical infection of laboratory animals with one or more of several pathogens affects the results of experiments on animals. Monitoring the health of laboratory animals encompasses routine surveillance for pathogens, including several viruses. This study aimed to explore the development of an alternative assay to the existing ones for detecting infection of mice and rats with the parvoviruses minute virus of mice (MVM) and Kilham rat virus (KRV), respectively. Full-length VP2 and NS1 proteins of these parvoviruses, besides fragments containing multiple predicted epitopes stitched together, were studied for serological detection. The optimal dilution of full-length proteins and antigenic regions containing predicted epitopes for coating, test sera, and conjugate was determined using a checkerboard titration at each step. The assays were evaluated vis-à-vis commercially available ELISA kits. The results showed that an engineered fusion of fragments containing multiple predicted MVM VP2 and NS1 epitopes was better than either of the full-length proteins for detecting antibodies in 90% of the tested sera samples. For KRV ELISA, full-length VP2 was better compared to other individual recombinant protein fragments or combinations thereof for the detection of antibodies in sera. This report is the first description of an ELISA for KRV and an improved assay for MVM. Importantly, our assays could be exploited with small volumes of sera. The results also demonstrate the utility of immunoinformatics-driven polypeptide engineering in the development of diagnostic assays and the potential to develop better tests for monitoring the health status of laboratory animals.


Assuntos
Vírus Miúdo do Camundongo , Parvovirus , Camundongos , Animais , Ratos , Imunoinformática , Ensaio de Imunoadsorção Enzimática/métodos , Animais de Laboratório , Anticorpos Antivirais , Peptídeos , Epitopos
3.
Biotechnol Prog ; 40(1): e3398, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-37985214

RESUMO

The capacity to remove viruses by Planova filters produced by Asahi Kasei, primarily by small virus-retentive filters, were compiled from data in peer-reviewed publications and, partly, publicly available data from presentations at conferences (Planova workshops). Data from more than 100 publications and presentations at conferences covering Planova filters were assessed. The data were grouped according to the different virus filters regarding mean pore sizes and viruses of different sizes for plasma and cell culture derived products. Planova 15N and 20N filters removed parvoviruses below the limit of detection of viruses in the filtrate in approx. 50% of all studies and mean LRFs (log reduction factors) for viruses detected in the filtrate were above 4, demonstrating effective parvovirus reduction. Parvovirus removal capacity increased for Planova BioEX filters as well as for 2 Planova 20N in series. Large viruses as retroviruses (e.g., HIV and MuLV), herpesviruses, flaviviruses and togaviruses were removed effectively by Planova 15N, 20N and BioEX filters and also by Planova 35N filters. Flow interruption, transmembrane pressure, volume and protein concentration per filter area had had no substantial impact on virus removal capacity at manufacturing specification. In conclusion, the incorporation of Planova filters in manufacturing processes of biologicals remove, depending on the filter pore size, small and large viruses from the feed stream reliably. This virus reduction step with an orthogonal mechanism integrated in the manufacturing processes of biologicals, based primarily on size exclusion of viruses, improves the virus safety of these biopharmaceutical products considerably.


Assuntos
Produtos Biológicos , Parvovirus , Vírus , Filtração , Vírus da Leucemia Murina
4.
Exp Clin Transplant ; 21(11): 868-871, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-38140930

RESUMO

OBJECTIVES: Parvovirus testing is not done in routine clinical practice; thus, it is possible that reported parvovirus cases are just the tip of the iceberg of total prevalence. We present a single-center retrospective analysis of 22 events of parvovirus B19 anemia in 20 renal transplant recipients, among which 2 patients had recurrence. MATERIALS AND METHODS: For this descriptive analytical study, parvovirus B19 disease was defined as parvovirus infection (detection by real-time polymerase chain reaction) in the presence of anemia with clinical symptoms or bone marrow biopsy findings consistent with the diagnosis. Study duration was 18 months, from June 2021 through December 2022, and patients were enrolled from a single center. RESULTS: All patients detected with the virus had received induction with thymocyte globulin and were on standard triple drug immunosuppression. Mean age was 32 ± 12 years with median time to diagnosis of 2 months after transplant. Anemia was observed in all patients with mean hemoglobin level at presentation of 6.02 ± 1.28 g/dL. Creatinine at presentation was 1.49 mg/dL (interquartile range, 0.92-2.69 mg/dL). The most common presentation was asymptomatic patient with evaluation for anemia. During therapy, the highest median creatinine level was 2.0 mg/dL (interquartile range, 1.38-3.2 mg/dL), which was significantly higher than that at presentation (P < .018). After therapy, median creatinine level was 1.3 mg/dL, which was not significantly higher than the baseline level, demonstrating a mostly transient graft dysfunction. CONCLUSIONS: Parvovirus B19 is a relatively underreported disease in renal transplant recipients, with patients presenting with anemia and the disease causing transient graft dysfunction. Parvovirus B19 infection responds well to a decrease in immunosuppression and intravenous immunoglobulin therapy.


Assuntos
Anemia , Transplante de Rim , Infecções por Parvoviridae , Parvovirus B19 Humano , Parvovirus , Humanos , Adulto Jovem , Adulto , Transplante de Rim/efeitos adversos , Creatinina , Estudos Retrospectivos , Infecções por Parvoviridae/diagnóstico , Infecções por Parvoviridae/epidemiologia , Anemia/diagnóstico , Anemia/epidemiologia , Anemia/etiologia , Parvovirus B19 Humano/genética
5.
Viruses ; 15(11)2023 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-38005856

RESUMO

Viral disinfection is important for medical facilities, the food industry, and the veterinary field, especially in terms of controlling virus outbreaks. Therefore, standardized methods and activity levels are available for these areas. Usually, disinfectants used in these areas are characterized by their activity against test organisms (i.e., viruses, bacteria, and/or yeasts). This activity is usually determined using a suspension test in which the test organism is incubated with the respective disinfectant in solution to assess its bactericidal, yeasticidal, or virucidal activity. In addition, carrier methods that more closely reflect real-world applications have been developed, in which microorganisms are applied to the surface of a carrier (e.g., stainless steel frosted glass, or polyvinyl chloride (PVC)) and then dried. However, to date, no standardized methods have become available for addressing genetically modified vectors or disinfection-resistant oncolytic viruses such as the H1-parvovirus. Particularly, such non-enveloped viruses, which are highly resistant to disinfectants, are not taken into account in European standards. This article proposes a new activity claim known as "virucidal activity PLUS", summarizes the available methods for evaluating the virucidal activity of chemical disinfectants against genetically modified organisms (GMOs) using current European standards, including the activity against highly resistant parvoviridae such as the adeno-associated virus (AAV), and provides guidance on the selection of disinfectants for pharmaceutical manufacturers, laboratories, and clinical users.


Assuntos
Desinfetantes , Infecções por Parvoviridae , Parvovirus , Vírus , Humanos , Desinfetantes/farmacologia , Desinfecção/métodos , Vírus/genética
6.
Viruses ; 15(10)2023 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-37896906

RESUMO

The XVIII International Parvovirus Workshop took place in Rimini, Italy, from 14 to 17 June 2022 as an on-site event, continuing the series of meetings started in 1985 and continuously held every two years. The communications dealt with all aspects of research in the field, from evolution and structure to receptors, from replication to trafficking, from virus-host interactions to clinical and veterinarian virology, including translational issues related to viral vectors, gene therapy and oncolytic parvoviruses. The oral communications were complemented by a poster exhibition available for view and discussion during the whole meeting. The XVIII International Parvovirus Workshop was dedicated to the memory of our dearest colleague Mavis Agbandje-McKenna (1963-2021).


Assuntos
Infecções por Parvoviridae , Parvovirus , Humanos , Parvovirus/genética , Vetores Genéticos , Itália
7.
Microbiol Spectr ; 11(4): e0457022, 2023 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-37347193

RESUMO

DNA replication is a standard and essential function among DNA viruses; however, this functional domain's common ancestor, origin, and evolutionary path in invertebrate- and vertebrate-infecting viruses are not yet fully understood. Here, we present evidence, using a combination of phylogenetic relationships, coevolution, and CLANS (cluster analysis of sequences) analysis, that the parvo-NS1 domain (nonstructural protein NS1, DNA helicase domain) of these DNA viruses that infect vertebrates potentially originated from the invertebrate (Platyhelminthes) parvo-NS1 domain of parvovirus-related sequences (PRSs). Our results suggest that papillomaviruses and the parvovirus subfamilies Densovirinae and Hamaparvovirinae DNA helicase evolved directly from the Platyhelminthes NS1 domain (PRSs). Similarly, the parvovirus subfamily Parvovirinae NS1 domain displayed evolutionary heritage from the PRSs through Hamaparvovirinae. Further, our analysis also clarified that herpesviruses and adenoviruses independently obtained the parvo-NS1 domain from Dependoparvovirus (Parvovirinae). Furthermore, virus-host coevolution analysis revealed that the parvovirus NS1 domain has coevolved with hosts, from flatworms to humans, and it appears that the papillomavirus may have obtained the DNA helicase during the early stages of parvovirus evolution and later led to the development of the DNA helicase of adomavirus and polyomavirus. Finally, herpesviruses and adenoviruses likely inherited the parvo-NS1 domain from Dependoparvovirus in the later stages of evolution. To the best of our knowledge, this is the first evolutionary evidence to suggest that the DNA helicase of viruses that infect vertebrates originated from the invertebrate PRSs. IMPORTANCE DNA replication of DNA viruses is an essential function. This allows DNA replication of viruses to form virus particles. The DNA helicase domain is responsible for this primary function. This domain is present in parvoviruses, papillomaviruses, polyomaviruses, herpesviruses, and adenoviruses. But little is known about the common ancestor, origin, and evolutionary path of DNA helicase in invertebrate- and vertebrate-infecting viruses. Here, we report the possibility of the origin of DNA viruses (DNA helicase) infecting vertebrates from Platyhelminthes (invertebrate) PRSs. Our study established that the parvovirus subfamily Parvovirinae NS1 domain displayed evolutionary heritage from the Platyhelminthes PRSs through Hamaparvovirinae. Furthermore, our study suggests that the papillomavirus DNA helicase may have evolved in the early stages of parvovirus evolution and then led to the development of the adomavirus and polyomavirus. Our study suggests that the herpesviruses and adenoviruses likely inherited the parvo-NS1 domain through gene capture from Dependoparvovirus in the later stages of parvovirus evolution in their hosts.


Assuntos
Infecções por Parvoviridae , Parvovirus , Animais , Humanos , Filogenia , Vírus de DNA/genética , Invertebrados , Parvovirus/genética , Vertebrados , Adenoviridae , DNA Helicases/genética
8.
PLoS Pathog ; 19(5): e1011203, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37253065

RESUMO

The oncolytic autonomous parvovirus Minute Virus of Mice (MVM) establishes infection in the nuclear environment by usurping host DNA damage signaling proteins in the vicinity of cellular DNA break sites. MVM replication induces a global cellular DNA Damage Response (DDR) that is dependent on signaling by the ATM kinase and inactivates the cellular ATR-kinase pathway. However, the mechanism of how MVM generates cellular DNA breaks remains unknown. Using single molecule DNA Fiber Analysis, we have discovered that MVM infection leads to a shortening of host replication forks as infection progresses, as well as induction of replication stress prior to the initiation of virus replication. Ectopically expressed viral non-structural proteins NS1 and NS2 are sufficient to cause host-cell replication stress, as is the presence of UV-inactivated non-replicative MVM genomes. The host single-stranded DNA binding protein Replication Protein A (RPA) associates with the UV-inactivated MVM genomes, suggesting MVM genomes might serve as a sink for cellular stores of RPA. Overexpressing RPA in host cells prior to UV-MVM infection rescues DNA fiber lengths and increases MVM replication, confirming that MVM genomes deplete RPA stores to cause replication stress. Together, these results indicate that parvovirus genomes induce replication stress through RPA exhaustion, rendering the host genome vulnerable to additional DNA breaks.


Assuntos
Vírus Miúdo do Camundongo , Infecções por Parvoviridae , Parvovirus , Animais , Camundongos , Vírus Miúdo do Camundongo/genética , Proteína de Replicação A/genética , Parvovirus/genética , Replicação Viral/genética , Infecções por Parvoviridae/genética , Replicação do DNA/genética
9.
Viruses ; 15(4)2023 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-37112831

RESUMO

The introduction of invasive birds into new ecosystems frequently has negative consequences for the resident populations. Accordingly, the increasing population of monk parakeets (Myiopsitta monachus) in Europe may pose a threat because we have little knowledge of the viruses they can transmit to native naïve species. In this study, we describe a new dependoparvovirus detected by metagenomic analysis of cloacal samples from 28 apparently healthy individuals captured in urban areas of Madrid, Spain. The genomic characterization revealed that the genome encoded the NS and VP proteins typical of parvoviruses and was flanked by inverted terminal repeats. No recombination signal was detected. The phylogenetic analysis showed that it was closely related to a parvovirus isolated in a wild psittacid in China. Both viruses share 80% Rep protein sequence identity and only 64% with other dependoparvoviruses identified in Passeriformes, Anseriformes, and Piciformes and are included in a highly supported clade, which could be considered a new species. The prevalence was very low, and none of the additional 73 individuals tested positive by PCR. These results highlight the importance of exploring the viral genome in invasive species to prevent the emergence of novel viral pathogenic species.


Assuntos
Papagaios , Parvovirus , Humanos , Animais , Periquitos/genética , Dependovirus , Espanha , Filogenia , Ecossistema
10.
Virol J ; 20(1): 61, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-37016419

RESUMO

A group of DNA viruses called parvoviruses that have significant effects on cancer therapy and genetic engineering applications. After passing through the cell membrane to reach the cytosol, it moves along the microtubule toward the nuclear membrane. The nuclear localization signal (NLS) is recognized by importin-beta (impß) and other proteins from the complex outside the nuclear membrane and binds to enter the nucleus via the nuclear pore complex (NPC). There are two main pathways for viruses to enter the nucleus. The classical pathway is through the interaction of imp α and impß with NLS via NPC. The other is the NPC mediated by the combination of impß and it. While the capsid is introduced into the nucleus through classical nuclear transduction, there is also a transient nuclear membrane dissolution leading to passive transport into the nucleus, which has been proposed in recent years. This article mainly discusses several nuclear entry pathways and related proteins, providing a reference for subsequent research on viral entry pathways.


Assuntos
Infecções por Parvoviridae , Parvovirus , Humanos , Sinais de Localização Nuclear/genética , Núcleo Celular/metabolismo , Membrana Nuclear/metabolismo , beta Carioferinas/genética , beta Carioferinas/metabolismo , Transporte Ativo do Núcleo Celular , alfa Carioferinas/metabolismo
11.
J Neurovirol ; 29(2): 226-231, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36857017

RESUMO

Etiology of vestibular schwannoma (VS) is unknown. Viruses can infect and reside in neural tissues for decades, and new viruses with unknown tumorigenic potential have been discovered. The presence of herpesvirus, polyomavirus, parvovirus, and anellovirus DNA was analyzed by quantitative PCR in 46 formalin-fixed paraffin-embedded VS samples. Five samples were analyzed by targeted next-generation sequencing. Viral DNA was detected altogether in 24/46 (52%) tumor samples, mostly representing anelloviruses (46%). Our findings show frequent persistence of anelloviruses, considered normal virome, in VS. None of the other viruses showed an extensive presence, thereby suggesting insignificant role in VS.


Assuntos
Anelloviridae , Herpesviridae , Neuroma Acústico , Parvovirus , Polyomavirus , Humanos , Polyomavirus/genética , Anelloviridae/genética , Neuroma Acústico/genética , Herpesviridae/genética , Parvovirus/genética , DNA Viral/genética
12.
Viruses ; 15(1)2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36680249

RESUMO

Oncotoxic proteins such as the non-structural protein 1 (NS1), a constituent of the rodent parvovirus H1 (H1-PV), offer a novel approach for treatment of tumors that are refractory to other treatments. In the present study, mutated NS1 variants were designed and tested with respect to their oncotoxic potential in human hepatocellular carcinoma cell lines. We introduced single point mutations of previously described important residues of the wild-type NS1 protein and a deletion of 114 base pairs localized within the N-terminal domain of NS1. Cell-viability screening with HepG2 and Hep3B hepatocarcinoma cells transfected with the constructed NS1-mutants led to identification of the single-amino acid NS1-mutant NS1-T585E, which led to a 30% decrease in cell viability as compared to NS1 wildtype. Using proteomics analysis, we could identify new interaction partners and signaling pathways of NS1. We could thus identify new oncotoxic NS1 variants and gain insight into the modes of action of NS1, which is exclusively toxic to human cancer cells. Our in-vitro studies provide mechanistic explanations for the observed oncolytic effects. Expression of NS1 variants had no effect on cell viability in NS1 unresponsive control HepG2 cells or primary mouse hepatocytes. The availability of new NS1 variants in combination with a better understanding of their modes of action offers new possibilities for the design of innovative cancer treatment strategies.


Assuntos
Parvovirus , Proteínas não Estruturais Virais , Animais , Humanos , Camundongos , Linhagem Celular , Neoplasias Hepáticas/genética , Infecções por Parvoviridae , Parvovirus/genética , Proteínas não Estruturais Virais/metabolismo
13.
PLoS One ; 17(11): e0276164, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36350876

RESUMO

BACKGROUND: Viral diseases are a major problem in shrimp aquaculture facilities as these diseases reduce growth rates, which inevitably lead to production and profit losses. Hepatopancreatic parvoviruses (HPV) are common diseases in shrimp that appear to be associated with high or low levels of replication in specific genetic lineages. Selective breeding may result in resistance to HPV and improved body traits such as body weight, meat yield and shrimp colour, facilitating shrimp farming. HPV virus titre is commonly determined by quantitative PCR (qPCR), which is a time-consuming method requiring laboratory equipment unsuitable for field implementation. The aim of this study was to develop a simple, robust, rapid and reliable method to detect HPV in low-resource environments. METHODS: We developed a rapid shrimp HPV test that uses (1) a simple three-step sample preparation protocol, followed by (2) isothermal recombinase polymerase amplification (RPA) and lateral flow strip detection (LFD). Analytical sensitivity testing was performed in a background banana shrimp sample matrix, and retrospective testing of Fenneropenaeus merguiensis hepatopancreas tissues (n = 33) with known qPCR viral titres was used to determine diagnostic sensitivity and specificity. RESULTS: The rapid shrimp HPV test could detect as little as 35 genome-equivalent copies per reaction in homogenized F. merguiensis banana shrimp. Retrospective testing of stored tissues (n = 33) indicated 100% diagnostic sensitivity (95% confidence interval, CI: 86-100%) and 100% specificity (95% CI: 66-100%) for detection of HPV. CONCLUSION: The rapid shrimp HPV test could be completed in only 40 minutes, and required only homogenization pestles, some pipettors, and a small heating block for single temperature incubation at 39°C. Critically, our procedure eliminated the time-consuming purification of nucleic acids from samples and when combined with RPA-LFD offers a user-friendly HPV detection format that can potentially be performed on-site. Our approach represents a major step forward in the development of a simple and sensitive end-point method for quick determination of unfavourable HPV virus numbers in shrimp, and has great potential to advance on-site management of shrimps in aquaculture.


Assuntos
Infecções por Papillomavirus , Parvovirus , Penaeidae , Animais , Recombinases , Estudos Retrospectivos , Penaeidae/genética , Sensibilidade e Especificidade , Parvovirus/genética , Técnicas de Amplificação de Ácido Nucleico/métodos
14.
PLoS Biol ; 20(11): e3001867, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36445931

RESUMO

Parvoviruses (family Parvoviridae) are small DNA viruses that cause numerous diseases of medical, veterinary, and agricultural significance and have important applications in gene and anticancer therapy. DNA sequences derived from ancient parvoviruses are common in animal genomes and analysis of these endogenous parvoviral elements (EPVs) has demonstrated that the family, which includes twelve vertebrate-specific genera, arose in the distant evolutionary past. So far, however, such "paleovirological" analysis has only provided glimpses into the biology of ancient parvoviruses and their long-term evolutionary interactions with hosts. Here, we comprehensively map EPV diversity in 752 published vertebrate genomes, revealing defining aspects of ecology and evolution within individual parvovirus genera. We identify 364 distinct EPV sequences and show these represent approximately 200 unique germline incorporation events, involving at least five distinct parvovirus genera, which took place at points throughout the Cenozoic Era. We use the spatiotemporal and host range calibrations provided by these sequences to infer defining aspects of long-term evolution within individual parvovirus genera, including mammalian vicariance for genus Protoparvovirus, and interclass transmission for genus Dependoparvovirus. Moreover, our findings support a model of virus evolution in which the long-term cocirculation of multiple parvovirus genera in vertebrates reflects the adaptation of each viral genus to fill a distinct ecological niche. Our findings show that efforts to develop parvoviruses as therapeutic tools can be approached from a rational foundation based on comparative evolutionary analysis. To support this, we published our data in the form of an open, extensible, and cross-platform database designed to facilitate the wider utilisation of evolution-related domain knowledge in parvovirus research.


Assuntos
Parvovirus , Vertebrados , Animais , Vertebrados/genética , Ecologia , Aclimatação , Agricultura , Parvovirus/genética , Mamíferos
15.
BMC Genomics ; 23(1): 565, 2022 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-35933380

RESUMO

BACKGROUND: Shrimp have the ability to accommodate viruses in long term, persistent infections without signs of disease. Endogenous viral elements (EVE) play a role in this process probably via production of negative-sense Piwi-interacting RNA (piRNA)-like fragments. These bind with Piwi proteins to dampen viral replication via the RNA interference (RNAi) pathway. We searched a genome sequence (GenBank record JABERT000000000) of the giant tiger shrimp (Penaeus monodon for the presence of EVE related to a shrimp parvovirus originally named infectious hypodermal and hematopoietic necrosis virus (IHHNV). RESULTS: The shrimp genome sequence contained three piRNA-like gene clusters containing scrambled IHHNV EVE. Two clusters were located distant from one another in pseudochromosome 35 (PC35). Both PC35 clusters contained multiple sequences with high homology (99%) to GenBank records DQ228358 and EU675312 that were both called "non-infectious IHHNV Type A" (IHHNV-A) when originally discovered. However, our results and those from a recent Australian P. monodon genome assembly indicate that the relevant GenBank records for IHHNV-A are sequence-assembly artifacts derived from scrambled and fragmental IHHNV-EVE. Although the EVE in the two PC35 clusters showed high homology only to IHHNV-A, the clusters were separate and distinct with respect to the arrangement (i.e., order and reading direction) and proportional content of the IHHNV-A GenBank records. We conjecture that these 2 clusters may constitute independent allele-like clusters on a pair of homologous chromosomes. The third EVE cluster was found in pseudochromosome 7 (PC7). It contained EVE with high homology (99%) only to GenBank record AF218266 with the potential to protect shrimp against current types of infectious IHHNV. One disadvantage was that some EVE in PC7 can give false positive PCR test results for infectious IHHNV. CONCLUSIONS: Our results suggested the possibility of viral-type specificity in EVE clusters. Specificity is important because whole EVE clusters for one viral type would be transmitted to offspring as collective hereditary units. This would be advantageous if one or more of the EVE within the cluster were protective against the disease caused by the cognate virus. It would also facilitate gene editing for removal of non-protective EVE clusters or for transfer of protective EVE clusters to genetically improve existing shrimp breeding stocks that might lack them.


Assuntos
Densovirinae , Parvovirus , Penaeidae , Animais , Austrália , DNA Viral/genética , Densovirinae/genética , Genoma Viral , Parvovirus/genética , Penaeidae/genética , RNA Interferente Pequeno
16.
J Fish Dis ; 45(8): 1201-1210, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35612903

RESUMO

Hepatopancreatic parvovirus (HPV) and Enterocytozoon hepatopenaei (EHP) are emerging and reemerging pathogens in shrimps. In the present study, a novel genotype of HPV concurrently infected with EHP in Penaeus vannamei in Taiwan leading to severe atrophy and damage of hepatopancreas were confirmed by histopathology, in situ hybridization, and PCR. The novel genotype of HPV exhibited 66%-69.5% sequence identities with all known HPVs and carried unique amino acid deletions and insertions in the VP gene. According to phylogenetic analysis, the Taiwan HPV isolates were classified as the genotype IV. The present study not only provided the histopathological and molecular proof of HPV and EHP co-infection in Taiwan, but also revealed the importance of investigating the geographical expansion of novel HPV genotypes.


Assuntos
Densovirinae , Enterocytozoon , Doenças dos Peixes , Infecções por Papillomavirus , Parvovirus , Penaeidae , Animais , Enterocytozoon/genética , Genótipo , Filogenia , Taiwan/epidemiologia
17.
J Fish Dis ; 45(9): 1323-1331, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35638102

RESUMO

Tilapia parvovirus (TiPV) is an emerging virus reportedly associated with disease and mortality in farmed tilapia. Although previous descriptions of histopathological changes are available, the lesions reported in these are not pathognomonic. Here, we report Cowdry type A inclusion bodies (CAIB) in the pancreas as a diagnostic histopathological feature found in adult Nile tilapia naturally infected with TiPV. This type of inclusion body has been well-known as a histopathological landmark for the diagnosis of other parvoviral infections in shrimp and terrestrial species. Interestingly, this lesion could be exclusively observed in pancreatic acinar cells, both in the hepatopancreas and pancreatic tissue along the intestine. In situ hybridization (ISH) using a TiPV-specific probe revealed the intranuclear presence of TiPV DNA in multiple tissues, including the liver, pancreas, kidney, spleen, gills and the membrane of oocytes in the ovary. These findings suggest that although TiPV can replicate in several tissue types, CAIB manifest exclusively in pancreatic tissues. In addition to TiPV, most diseased fish were co-infected with Streptococcus agalactiae, and presented with multifocal granulomas secondary to this bacterial infection. Partial genome amplification of TiPV was successful and revealed high nucleotide identity (>99%) to previously reported isolates. In summary, this study highlights the usefulness of pancreatic tissue as a prime target for histopathological diagnosis of TiPV in diseased Nile tilapia. This pattern may be critical when determining the presence of TiPV infection in new geographic areas, where ancillary testing may not be available. TiPV pathogenesis in this landmark organ warrants further investigation.


Assuntos
Ciclídeos , Doenças dos Peixes , Parvovirus , Infecções Estreptocócicas , Tilápia , Animais , Ciclídeos/microbiologia , Doenças dos Peixes/microbiologia , Pâncreas/patologia , Parvovirus/genética , Streptococcus agalactiae/genética
18.
Transbound Emerg Dis ; 69(5): e1417-e1433, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35150091

RESUMO

Several viruses can infect wild carnivores but their impact on wildlife health is poorly understood. We investigated the presence, diversity and distribution of various DNA viruses in 303 wolves inhabiting a vast area of the Northwest Territories, Canada, over a period of 13 years. We found evidence for the presence of canine bufavirus (CBuV, 42.6%), canine parvovirus 2 (CPV-2, 34.0%), canine bocavirus 2 (CBoV-2, 5.0%), cachavirus (CachaV-1, 2.6%), canine adenovirus 1 (CAdV-1, 1%) and minute virus of canines (MVC, 0.3%). To our knowledge, this is the first detection of CBoV-2, MVC and CachV-1 in wild animals. We also demonstrate that CBuV and CachaV-1 were already circulating among wild animals at least 11 and 10 years, respectively, before their discoveries. Although CBuV prevalence was higher, CPV-2 was the most prevalent virus among juveniles, while CBuV infection was associated with poor nutrition conditions. Even if its prevalence was low, CachaV-1 had the highest multiple infection rate (87.5%). CadV-1 and MVC sequences were highly identical to reference strains, but we observed a high diversity among the other viruses and detected three new variants. One CPV-2 variant and one CBuV variant were endemic since the beginning of the 2000s in the entire investigated region, whereas one CBuV variant and two CBoV-2 variants were found in a more restricted area over multiple years and CachaV-1 was found only in one region. Two CPV-2 variants and one CachaV-1 variant were observed only once, indicating sporadic introductions or limited circulation. Different patterns of endemicity might indicate that viruses were introduced in the wolf population at different timepoints and that mixing between wolf packs may not be constant. Different epidemiological behaviors depend on viral factors like infectivity, transmission routes, pathogenicity and tissue-tropism, and on host factors like proximity to densely populated areas, carnivory and pack density and mixing.


Assuntos
Adenovirus Caninos , Carnívoros , Doenças do Cão , Infecções por Parvoviridae , Parvovirus Canino , Parvovirus , Lobos , Adenovirus Caninos/genética , Animais , Animais Selvagens , Canadá/epidemiologia , Doenças do Cão/epidemiologia , Cães , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/veterinária , Parvovirus/genética , Parvovirus Canino/genética , Filogenia
19.
Transbound Emerg Dis ; 69(5): 2952-2962, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35018730

RESUMO

Outbreaks of short beak dwarf syndrome caused by novel goose parvovirus (NGPV) have been prevalent in China since 2015, resulting in a high mortality rate of ducks. Herein we evaluated differences between two NGPV strains: Muscovy duck-origin (AH190917-RP: MD17) and Cherry Valley duck-origin (JS191021-RP: CVD21) NGPV. Both of them showed certain level of pathogenicity to primary duck embryo fibroblasts, Cherry Valley duck embryos and ducklings. CVD21 showed comparatively stronger pathogenicity than MD17. Only CVD21 caused obvious cytopathic effect (CPE), characterized by cell shedding; further, the virus titer of MD17 and CVD21 was 102.571 ELD50 (i.e. median embryo lethal dose)/0.2 ml and 106.156 ELD50 /0.2 ml, respectively, and the mortality rate of CVD21- and MD17-infected Cherry Valley ducklings was 100% and 80%, respectively. In addition, CVD21 had a greater influence on the growth and development of ducklings. Furthermore, we found that MD17 could infect Muscovy duck embryos and produce lesions similar to Cherry Valley duck embryos, but it could not infect Muscovy duck embryo fibroblasts (MDEFs,) and Muscovy ducklings. MDV21 had no infection to MDEFs, Muscovy duck embryo and Muscovy ducklings. We then sequenced the complete genome of the two isolates to enable genomic characterization. The complete genome of MD17 and CVD21 was 5046 and 5050 nucleotides in length, respectively. Nucleotide alignment, amino acid analysis and phylogenetic tree analysis revealed that MD17 showed higher homology to goose parvovirus (GPV), while CVD21 demonstrated stronger similarity with NGPV. Moreover, the two isolates shared 95.8% homology, with encoded proteins showing multiple amino acid variations. Our findings indicate that Muscovy ducks seem to have played a crucial role in the evolution of GPV to NGPV. We believe that our data should serve as a foundation for further studying the genetic evolution of waterfowl parvoviruses and their pathogenic mechanisms.


Assuntos
Infecções por Parvoviridae , Parvovirus , Doenças das Aves Domésticas , Aminoácidos/genética , Animais , Patos , Nucleotídeos , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/veterinária , Parvovirinae , Parvovirus/genética , Filogenia
20.
Intervirology ; 65(3): 160-166, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34695823

RESUMO

Cross-species transmission of viral diseases alarms our global community for its potential of novel pandemic events. Of various viral pathogens noted recently, parvoviruses have posed public health threats not only to humans but also to wild animals. To investigate the prevalence of parvoviruses in wild Manchurian chipmunks, here we detected genetic fragments of the nonstructural protein of parvovirus by polymerase chain reaction in wild Manchurian chipmunk specimens captured in the central and southern regions of South Korea and compared their sequence homology with references. Of a total of 348 specimens examined, chipmunk parvovirus (ChpPV)-specific gene fragments were detected with a 31.32% rate (109 chipmunks of 348) in their kidney, liver, lung, and spleen samples, and the chipmunks captured in Gangwon Province exhibited the highest positive rate (45.37%), followed by Gyeongsang (35.29%), Gyeonggi (31.03%), Chungcheong (20.00%), and Jeolla (19.70%). When compared with the reference sequences, a partial ChpPV sequence showed 97.70% identity to the previously reported Korean strain at the nucleic acid level. In the phylogenetic analysis, ChpPV exhibited closer relationship to primate parvoviruses, erythroviruses, and bovine parvovirus than to adeno-associated viruses. Despite limited sample size and genetic sequences examined in this study, our results underline the prevalence of ChpPV in Korea and emphasize the need of close surveillance of parvoviruses in wild animals.


Assuntos
Infecções por Parvoviridae , Parvovirus , Animais , Animais Selvagens , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/veterinária , Parvovirus/genética , Filogenia , Sciuridae
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA