Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Gen Virol ; 105(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38767609

RESUMO

Hepeviruses have been identified in a broad range of animal hosts, including mammals, birds, and fish. In this study, rodents (n=91) from seven different species and ten pikas (Ochotona curzoniae) were collected in Qinghai Province, China. Using transcriptomic sequencing and confirmatory molecular testing, hepeviruses were detected in 27 of 45 (60 %) long-tailed dwarf hamsters (Cricetulus longicaudatus) and were undetected in other rodents and pika. The complete genome sequences from 14 representative strains were subsequently obtained, and phylogenetic analyses suggested that they represent a novel species within the genus Rocahepevirus, which we tentatively designated as Cl-2018QH. The virus was successfully isolated in human hepatoma (Huh-7) and murine fibroblast (17 Cl-1) cell lines, though both exhibited limited replication as assayed by detection of negative-sense RNA intermediates. A129 immunodeficient mice were inoculated with Cl-2018QH and the virus was consistently detected in multiple organs, despite relatively low viral loads. In summary, this study has described a novel rodent hepevirus, which enhances our knowledge of the genetic diversity of rodent hepeviruses and highlights its potential for cross-species transmission.


Assuntos
Genoma Viral , Hepevirus , Filogenia , Animais , China , Cricetinae , Camundongos , Hepevirus/genética , Hepevirus/isolamento & purificação , Hepevirus/classificação , Humanos , Linhagem Celular , RNA Viral/genética
2.
Poult Sci ; 103(4): 103501, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38350386

RESUMO

Previous studies have shown that avian hepatitis E virus (HEV) decreases egg production by 10-40% in laying hens, but have not fully elucidated the mechanism of there. In this study, we evaluated the replication of avian HEV in the ovaries of laying hens and the mechanism underlying the decrease in egg production. Forty 150-days-old commercial laying hens were randomly divided into 2 groups of 20 hens each. A total of 1 mL (104GE) of avian HEV stock was inoculated intravenously into each chicken in the experimental group, with 20 chickens in the other group serving as negative controls. Five chickens from each group were necropsied weekly for histopathological examination. The pathogenicity of avian HEV has been characterized by seroconversion, viremia, fecal virus shedding, ovarian lesions, and decreased egg production. Both positive and negative-strand avian HEV RNA, and ORF2 antigens can be detected in the ovaries, suggesting that avian HEV can replicate in the ovaries and serve as an important extrahepatic replication site. The ovaries of laying hens underwent apoptosis after avian HEV infection. These results indicate that avian HEV infection and replication in ovarian tissues cause structural damage to the cells, leading to decreased egg production.


Assuntos
Vírus da Hepatite E , Hepevirus , Cistos Ovarianos , Neoplasias Ovarianas , Doenças das Aves Domésticas , Animais , Feminino , Galinhas , Cistos Ovarianos/veterinária , Neoplasias Ovarianas/veterinária , Hepevirus/genética , Apoptose
3.
Sci Rep ; 12(1): 21743, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36526693

RESUMO

In 2019, outbreaks of hepatitis-splenomegaly syndrome (HSS) were observed in six commercial layer chicken flocks, belonging to three different Polish farms, and characterized by increased mortality, hemorrhagic hepatitis with attached blood clots on the liver surface, and splenomegaly. Diseased flocks were initially investigated for the presence of avian hepatitis E virus (aHEV) - the etiological agent of HSS - by conventional reverse transcriptase polymerase chain reaction, which revealed aHEV sequences clustering separately from all known aHEV genotypes. Additionally, an aHEV genome was identified for the first time in common pheasants, from a flock in France, using Next Generation Sequencing. This genome clustered together with the Polish aHEVs here investigated. Complete genome aHEV sequences from the HSS outbreaks confirmed the divergent cluster, with a shared nucleotide sequence identity of 79.6-83.2% with other aHEVs, which we propose to comprise a novel aHEV genotype - genotype 7. Histology and immunohistochemistry investigations in the liver and spleen established an association between aHEV and the observed lesions in the affected birds, consolidating the knowledge on the pathogenesis of aHEV, which is still largely unknown. Thus, the present investigation extends the natural host range and genotypes of aHEV and strengthens knowledge on the pathogenesis of HSS.


Assuntos
Hepatite Viral Animal , Hepevirus , Doenças das Aves Domésticas , Infecções por Vírus de RNA , Animais , Hepevirus/genética , Galinhas , Esplenomegalia , Especificidade de Hospedeiro , Genótipo , Codorniz , Filogenia
4.
Virus Genes ; 58(6): 589-593, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36183048

RESUMO

Hepatitis E virus (HEV) infection has a global distribution with diverse hosts, including mammals and avians. In this study, an avian Hepatitis E virus (aHEV) strain with a high mortality rate of about 30%, designated as SDXT20, was obtained from the liver of 30-week-old Hubbard chickens with severe hepatosplenomegaly in 2020 in Eastern China and HEV was proved to be the only pathogen by next-generation sequencing. Its complete genome, which encodes three open reading frames (ORFs), is 6649 nt in length. ORF1-3 encodes three proteins with lengths of 1532 aa, 606 aa, and 82 aa, respectively, and ORF2 and ORF3 overlap with each other. BLAST-based similarity analysis of the complete viral genome demonstrated that SDXT20 had merely 80.5-92.2% similarity with avian Avihepevirus magniiecur strains and 50.4%-54.8% lower similarity with Paslahepevirus balayani, Rocahepevirus ratti, and Chirohepevirus eptesici species. Further genetic evolution analysis of the complete genome and ORF2 revealed that the isolate was genetically distinct from known aHEVs, and it belonged to a novel genetically distinct aHEV. This study provides data for further analysis of the multi-host and cross-host genetic evolution of HEVs.


Assuntos
Vírus da Hepatite E , Hepatite E , Hepevirus , Animais , Hepevirus/genética , Galinhas , Vírus da Hepatite E/genética , Hepatite E/veterinária , Genoma Viral/genética , Fases de Leitura Aberta/genética , China , Mamíferos
5.
J Gen Virol ; 103(9)2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36170152

RESUMO

The family Hepeviridae includes enterically transmitted small quasi-enveloped or non-enveloped positive-sense single-stranded RNA viruses infecting mammals and birds (subfamily Orthohepevirinae) or fish (Parahepevirinae). Hepatitis E virus (genus Paslahepevirus) is responsible for self-limiting acute hepatitis in humans; the infection may become chronic in immunocompromised individuals and extrahepatic manifestations have been described. Avian hepatitis E virus (genus Avihepevirus) causes hepatitis-splenomegaly syndrome in chickens. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Hepeviridae, which is available at www.ictv.global/report/hepeviridae.


Assuntos
Hepevirus , Vírus de RNA , Animais , Galinhas , Peixes , Genoma Viral , Hepevirus/genética , Humanos , Mamíferos , Vírus de RNA/genética , Vírion , Replicação Viral
6.
Poult Sci ; 101(10): 102103, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36041385

RESUMO

To investigate the prevalence of avian hepatitis E virus (HEV) in chickens and gather evidence of viral vertical transmission, we collected 288 cloacal swabs and 288 yolks samples from 12 farms with clinically healthy chickens in 4 different areas in Tai'an City, Shandong Province, China (i.e., Daiyue District, Xintai City, Feicheng City, and Ningyang County). We also collected 240 samples from 2 breeder farms (from each of which 30 chicks, 30 dead embryos, 30 live embryos, and 30 hatching eggs were taken). PCR detection revealed that the positive rates of cloacal swabs and yolks were 6.25% (18/288) and 4.51% (13/288), respectively. Besides, avian HEV was detected with higher positive rates in the chicks (11.67%), hatching eggs (10.00%), live embryos (13.33%), and dead embryos (26.67%) from 2 breeder farms. Sequence and genetic evolution analyses revealed that the nucleotide homology of the isolated strains was 76.4to 83.9% compared with 4 reported genotypes, but the isolated strains were located in a separate branch, indicating they were potential novel genotypes. In conclusion, those results indicate that the latent infection of avian HEV novel genotypes has been widespread in chicken farms in Tai'an City, and provide reliable evidence of the possible vertical transmission of avian HEV.


Assuntos
Hepevirus , Doenças das Aves Domésticas , Animais , Galinhas/genética , China/epidemiologia , Genótipo , Hepevirus/genética , Nucleotídeos , Óvulo/química , Filogenia , Prevalência , RNA Viral/genética
7.
Microbiol Spectr ; 10(1): e0226521, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35138149

RESUMO

Avian hepatitis E virus (HEV) causes liver diseases and multiple extrahepatic disorders in chickens. However, the mechanisms involved in avian HEV entry remain elusive. Herein, we identified the RAS-related protein 1b (Rap1b) as a potential HEV-ORF2 protein interacting candidate. Experimental infection of chickens and cells with an avian HEV isolate from China (CaHEV) led to upregulated expression and activation of Rap1b both in vivo and in vitro. By using CaHEV capsid as mimic of virion to treat cell in vitro, it appears that the interaction between the viral capsid and Rap1b promoted cell membrane recruitment of the downstream effector Rap1-interacting molecule (RIAM). In turn, RIAM further enhanced Talin-1 membrane recruitment and retention, which led to the activation of integrin α5/ß1, as well as integrin-associated membrane protein kinases, including focal adhesion kinase (FAK). Meanwhile, FAK activation triggered activation of downstream signaling molecules, such as Ras-related C3 botulinum toxin substrate 1 RAC1 cell division cycle 42 (CDC42), p21-activated kinase 1 (PAK1), and LIM domain kinase 1 (LIMK1). Finally, F-actin rearrangement induced by Cofilin led to the formation of lamellipodia, filopodia, and stress fibers, contributes to plasma membrane remodeling, and might enhance CaHEV virion internalization. In conclusion, our data suggested that Rap1b activation was triggered during CaHEV infection and appeared to require interaction between CaHEV-ORF2 and Rap1b, thereby further inducing membrane recruitment of Talin-1. Membrane-bound Talin-1 then activates key Integrin-FAK-Cofilin cascades involved in modulation of actin kinetics, and finally leads to F-actin rearrangement and membrane remodeling to potentially facilitate internalization of CaHEV virions into permissive cells. IMPORTANCE Rap1b is a multifunctional protein that is responsible for cell adhesion, growth, and differentiation. The inactive form of Rap1b is phosphorylated and distributed in the cytoplasm, while active Rap1b is prenylated and loaded with GTP to the cell membrane. In this study, the activation of Rap1b was induced during the early stage of avian HEV infection under the regulation of PKA and SmgGDS. Continuously activated Rap1b recruited its effector RIAM to the membrane, thereby inducing the membrane recruitment of Talin-1 that led to the activation of membrane α5/ß1 integrins. The triggering of the signaling pathway-associated Integrin α5/ß1-FAK-CDC42&RAC1-PAK1-LIMK1-Cofilin culminated in F-actin polymerization and membrane remodeling that might promote avian HEV virion internalization. These findings suggested a novel mechanism that is potentially utilized by avian HEV to invade susceptible cells.


Assuntos
Citoesqueleto/metabolismo , Hepatite Viral Animal/metabolismo , Hepevirus/patogenicidade , Doenças das Aves Domésticas/metabolismo , Proteínas Virais/metabolismo , Internalização do Vírus , Proteínas rap de Ligação ao GTP/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Galinhas , Citoesqueleto/genética , Citoesqueleto/virologia , Hepatite Viral Animal/genética , Hepatite Viral Animal/virologia , Hepevirus/genética , Interações Hospedeiro-Patógeno , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia , Ligação Proteica , Proteínas Virais/genética , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Proteínas rap de Ligação ao GTP/genética
8.
Vet Microbiol ; 265: 109331, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34999311

RESUMO

Hepatitis E virus (HEV), a zoonotic virus, infects many animal species, including humans. Capsid proteins of human, swine, rabbit and avian HEVs share 48 %-100 % amino acid identity. In the present study, antigenic cross-reactivity among human, swine, rabbit and avian HEV capsid proteins were analyzed in detail using indirect and blocking enzyme-linked immunosorbent assays (ELISAs). The C-terminal 268 amino acids of genotype 1 human, genotype 4 swine, genotype 3 rabbit and genotype B3 avian HEV capsid proteins served as coating antigens for ELISA. Hyperimmune rabbit antisera (against four HEV capsid proteins) and human, pig, rabbit and chicken clinical sera were as primary antibodies. Closely correlated and statistically indistinguishable results were obtained for detection of anti-HEV antibodies in human and pig sera using human, swine and rabbit HEV capsid proteins as coating antigens. Moderately correlated differences in detection of anti-HEV antibodies in rabbit sera were obtained using the three capsid proteins. Statistically significant differences with no correlations were obtained for anti-HEV antibodies detection in chicken sera between avian HEV capsid protein and human, swine and rabbit ones. Blocking ELISA results demonstrated that two common epitopes among the four species HEVs were immunodominant in avian HEV, but were non-immunodominant in human, swine and rabbit HEVs. Nevertheless, three epitopes common to human, swine and rabbit HEVs were all immunodominant epitopes for the three species HEVs. Collectively, these results demonstrate that anti-HEV antibodies in human and pig sera can be detected using human, swine and rabbit HEV capsid proteins. By contrast, for optimal detection of anti-HEV antibodies in rabbit and chicken sera, the respective rabbit and avian HEV capsid proteins should be used. These results provide insights to guide future development of serological assays for diagnosing HEV infections in various animal species.


Assuntos
Vírus da Hepatite E , Hepatite E , Hepevirus , Doenças dos Suínos , Animais , Antígenos Virais , Aves , Proteínas do Capsídeo/genética , Hepatite E/veterinária , Vírus da Hepatite E/genética , Hepevirus/genética , Humanos , Coelhos , Suínos
9.
Poult Sci ; 101(3): 101633, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34986452

RESUMO

Avian hepatitis E virus (HEV) is the major causative pathogen of the big liver and spleen disease, hepatitis-splenomegaly syndrome, and hepatic rupture hemorrhage syndrome. Until now, there are 6 different avian HEV genotypes that infect chickens have been reported worldwide. Epidemiologic investigations of the avian HEV demonstrated that avian HEV has been widely spread in China in recent years. In this study, an avian HEV named YT-aHEV was obtained from white-feathered broilers using LMH cells by virus isolation assay in Shandong province, China. The complete genome consists of 6656-nt excluding the poly(A) tail. The isolate was highly similar to the CaHEV strain and segregated into the same branch belonging to avian HEV genotype 3. Indirect immunofluorescence using capsid protein-specific polyclonal antibodies confirmed that YT-aHEV could establish productive infection and replicate stably in LMH cells. Furthermore, an in vivo avian HEV infection model was established successfully in specific pathogen-free chicken embryos by intravenous experiments. In the present study, we demonstrate an avian HEV infection associated with liver lesions of hemorrhage and swelling by LMH cells for the first time in a white-feather broiler flock in China. This research also provides a new diagnosis method for detection of avian HEV, which laid a foundation for the understanding of pathogenicity and molecular biology of this virus for further study.


Assuntos
Hepatite Viral Animal , Hepevirus , Doenças das Aves Domésticas , Animais , Embrião de Galinha , Galinhas , China/epidemiologia , Plumas/patologia , Hepatite Viral Animal/epidemiologia , Hepevirus/genética
10.
BMC Vet Res ; 18(1): 56, 2022 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-35078465

RESUMO

BACKGROUND: Avian hepatitis E virus (HEV) is the pathogenic agent of big liver and spleen disease (BLS) and of hepatitis-splenomegaly syndrome (HSS) in chickens, which have caused economic losses to the poultry industry in China. In this study, 18 samples of BLS chickens were collected to reveal the molecular epidemiological characteristics of avian HEV in the province of Shandong, China. RESULTS: Gross and microscopic lesions of clinical samples were observed; then, virology detection and genetic analysis of avian HEV were performed. The results showed that there was significant swelling and rupture in the liver and that the spleen was enlarged. Microscopic lesions demonstrated obvious hemorrhage in the liver, with infiltration of heterophilic granulocytes, lymphocytes, and macrophages, as well as the reduction of lymphocytes in the spleen. Eleven of the 18 samples were positive for avian HEV, with a positive rate of 61.11%. More importantly, all avian HEV-positive samples were mixed infections: among these, the mixed infections of avian HEV and chicken infectious anemia virus (CIAV) and avian HEV and fowl adenovirus (FAdV) were the most common. Furthermore, the genetic evolution analysis showed that all avian HEV strains obtained here did not belong to the reported 4 genotypes, thus constituting a potential novel genotype. CONCLUSIONS: These results of this study further enrich the epidemiological data on avian HEV in Shandong, prove the genetic diversity of avian HEV in China, and uncover the complex mixed infections of avian HEV clinical samples.


Assuntos
Coinfecção , Hepatite E , Hepatite Viral Animal , Doenças das Aves Domésticas , Animais , Galinhas , China/epidemiologia , Coinfecção/veterinária , Hepatite E/epidemiologia , Hepatite E/veterinária , Hepatite Viral Animal/diagnóstico , Hepatite Viral Animal/epidemiologia , Hepevirus/genética , Epidemiologia Molecular , Filogenia , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/epidemiologia
11.
Infect Genet Evol ; 96: 105095, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34597819

RESUMO

Avian hepatitis E virus (aHEV) is the causative agent of an important disease of broiler breeders and layers. aHEV cannot be readily propagated in cell culture and is characterised primarily by sequencing of amplicons generated through several RT-PCRs that target individual genes. This study aims to uncover the origin of current Australian aHEV isolates based on whole genome sequencing using clinical liver tissues. Complete genome sequences of the two aHEV isolates were assembled using Nanopore and Illumina reads. The two isolates possessed only four single nucleotide polymorphisms to each other. Comparison of the sequences with aHEV genome sequences available in the GenBank showed the highest nucleotide sequence identity of 88% with the prototype USA strain (AY535004), 82% with the European (AM943647) and genotype 1 Australian strains (AM943647). Recombination analysis suggested that aHEV isolates characterised in this study are progeny of a cross between a US and a Hungarian strain. Phylogenetic tree and phylogenetic networks constructed using complete genome and individual coding sequences revealed that Australian aHEV isolates formed a distinct clade closer to the USA strains and classified as genotype 2 whereas genotype 1 Australian strain clustered together with South Korean strains.


Assuntos
Galinhas , Genoma Viral , Hepatite Viral Animal/virologia , Hepevirus/genética , Doenças das Aves Domésticas/virologia , Infecções por Vírus de RNA/veterinária , Animais , Feminino , Fígado/virologia , Filogenia , Infecções por Vírus de RNA/virologia , Recombinação Genética , Sequenciamento Completo do Genoma
12.
Viruses ; 13(9)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34578311

RESUMO

The sole member of the Piscihepevirus genus (family Hepeviridae) is cutthroat trout virus (CTV) but recent metatranscriptomic studies have identified numerous fish hepevirus sequences including CTV-2. In the current study, viruses with sequences resembling both CTV and CTV-2 were isolated from salmonids in eastern and western Canada. Phylogenetic analysis of eight full genomes delineated the Canadian CTV isolates into two genotypes (CTV-1 and CTV-2) within the Piscihepevirus genus. Hepevirus genomes typically have three open reading frames but an ORF3 counterpart was not predicted in the Canadian CTV isolates. In vitro replication of a CTV-2 isolate produced cytopathic effects in the CHSE-214 cell line with similar amplification efficiency as CTV. Likewise, the morphology of the CTV-2 isolate resembled CTV, yet viral replication caused dilation of the endoplasmic reticulum lumen which was not previously observed. Controlled laboratory studies exposing sockeye (Oncorhynchus nerka), pink (O. gorbuscha), and chinook salmon (O. tshawytscha) to CTV-2 resulted in persistent infections without disease and mortality. Infected Atlantic salmon (Salmo salar) and chinook salmon served as hosts and potential reservoirs of CTV-2. The data presented herein provides the first in vitro and in vivo characterization of CTV-2 and reveals greater diversity of piscihepeviruses extending the known host range and geographic distribution of CTV viruses.


Assuntos
Doenças dos Peixes/virologia , Hepevirus/classificação , Hepevirus/genética , Hepevirus/isolamento & purificação , Animais , Canadá , Genótipo , Hepevirus/patogenicidade , Infecção Persistente/virologia , Filogenia , Salmo salar/virologia , Salmão/virologia , Truta , Virulência , Vírus não Classificados/classificação , Vírus não Classificados/genética , Vírus não Classificados/isolamento & purificação , Vírus não Classificados/patogenicidade
13.
Viruses ; 13(6)2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34064072

RESUMO

Avian hepatitis E virus (aHEV) is the major etiological agent of hepatitis-splenomegaly syndrome (HSS), big liver and spleen disease (BLSD), and hepatic rupture hemorrhage syndrome (HRHS) in chickens. Infections with aHEV cause a significant decrease in egg production and increased mortality in chickens worldwide. However, studies on the prevalence of aHEV in Nigeria are scarce. In this study, serum (n = 88) and fecal samples (n = 110) obtained from apparently healthy layer chickens from three states in southwestern Nigeria were analyzed by nested reverse transcription-polymerase chain reaction (nRT-PCR) targeting the helicase and capsid gene for the presence of aHEV. Avian HEV was detected in 12.5% (n = 11/88) of serum samples and 9.1% (n = 10/110) of fecal samples tested. Phylogenetic analysis showed that five of the twelve identified aHEV sequences belonged to genotype 2. The remaining seven sequences were only distantly related to other known aHEV isolates. After amplification of the near-complete ORF2 fragment (1618 bp) and part of the ORF1 (582 bp) of isolate YF40_aHEV_NG phylogenetic analysis revealed a nucleotide sequence identity between 79.0 and 82.6% and 80.1 and 83.5%, respectively, to other known aHEV strains, indicating that the Nigerian isolate YF40_aHEV_NG belongs to a novel aHEV genotype. This is the first report of co-circulation of aHEV genotypes in chickens in Nigeria.


Assuntos
Galinhas , Genoma Viral , Genótipo , Hepatite Viral Animal/virologia , Hepevirus/classificação , Doenças das Aves Domésticas/virologia , Infecções por Vírus de RNA/veterinária , Animais , Hepatite Viral Animal/epidemiologia , Hepevirus/genética , Hepevirus/isolamento & purificação , Nigéria/epidemiologia , Fases de Leitura Aberta , Filogenia , Doenças das Aves Domésticas/epidemiologia , RNA Viral
14.
PLoS One ; 16(6): e0252534, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34133435

RESUMO

Many recent disease outbreaks in humans had a zoonotic virus etiology. Bats in particular have been recognized as reservoirs to a large variety of viruses with the potential to cross-species transmission. In order to assess the risk of bats in Switzerland for such transmissions, we determined the virome of tissue and fecal samples of 14 native and 4 migrating bat species. In total, sequences belonging to 39 different virus families, 16 of which are known to infect vertebrates, were detected. Contigs of coronaviruses, adenoviruses, hepeviruses, rotaviruses A and H, and parvoviruses with potential zoonotic risk were characterized in more detail. Most interestingly, in a ground stool sample of a Vespertilio murinus colony an almost complete genome of a Middle East respiratory syndrome-related coronavirus (MERS-CoV) was detected by Next generation sequencing and confirmed by PCR. In conclusion, bats in Switzerland naturally harbour many different viruses. Metagenomic analyses of non-invasive samples like ground stool may support effective surveillance and early detection of viral zoonoses.


Assuntos
Quirópteros/virologia , Fezes/virologia , Metagenômica/métodos , Viroma/genética , Vírus/genética , Zoonoses/virologia , Adenoviridae/classificação , Adenoviridae/genética , Animais , Quirópteros/classificação , Reservatórios de Doenças/virologia , Variação Genética , Genoma Viral/genética , Hepevirus/classificação , Hepevirus/genética , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/classificação , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Filogenia , Rotavirus/classificação , Rotavirus/genética , Análise de Sequência de DNA/métodos , Suíça , Vírus/classificação
15.
Infect Genet Evol ; 93: 104942, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34044191

RESUMO

Rodents including rats are reservoir of several pathogens capable of affecting human health. In this study, faecal and different organ specimens from free-living Norway rats (Rattus norvegicus) (N = 18) and faecal samples from laboratory rodents (rats N = 21 and mice N = 20) collected from different geographic areas in Hungary between 2017 and 2020 were investigated by viral metagenomics and conventional RT-PCR methods. The complete genome of three different RNA viruses, rat astrovirus, rat norovirus and rat hepevirus were characterized and analysed in detail. Rat norovirus was detected in faecal (17.6%, 3/17) and kidney (7.1%, 1/14) samples; rat astrovirus in faecal (23.5%, 4/17) and spleen (13.3%, 2/15) samples, and rat hepevirus in 43% to 67% the faecal, liver, kidney, lung, heart, muscle, brain and blood samples from Norway rats, respectively. Rat norovirus was also identifiable in 5% (1/21) of laboratory rats and rat astrovirus in 40% (8/20) of faecal samples from laboratory mice. Co-infections were found in 28% (5/18) wild Norway rats. The highest RNA viral load of astrovirus (1.81 × 108 copy/g) and norovirus (3.49 × 107 copy/g) were measured in faecal samples; while the highest RNA viral load of hepevirus (1.16 × 109 copy/g) was found in liver samples of Norway rats, respectively. This study confirms the wide geographic distribution and high prevalence of astrovirus, norovirus and hepevirus among wild rats in Hungary with confirmation of different organ involvement of as well as the detection of norovirus and astrovirus in laboratory rats and mice, respectively. This finding further strengthens the role of rodents in the spread of viral pathogens especially infecting human.


Assuntos
Astroviridae/isolamento & purificação , Hepevirus/isolamento & purificação , Camundongos , Norovirus/isolamento & purificação , Ratos , Doenças dos Roedores/epidemiologia , Animais , Animais de Laboratório , Animais Selvagens , Astroviridae/genética , Infecções por Astroviridae/epidemiologia , Infecções por Astroviridae/veterinária , Infecções por Astroviridae/virologia , Infecções por Caliciviridae/epidemiologia , Infecções por Caliciviridae/veterinária , Infecções por Caliciviridae/virologia , Hepatite Viral Animal/epidemiologia , Hepatite Viral Animal/virologia , Hepevirus/genética , Hungria/epidemiologia , Norovirus/genética , Infecções por Vírus de RNA/epidemiologia , Infecções por Vírus de RNA/veterinária , Infecções por Vírus de RNA/virologia , Doenças dos Roedores/virologia
16.
Avian Dis ; 65(4): 572-577, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-35068100

RESUMO

Hepatitis-splenomegaly syndrome is caused by avian hepatitis E virus (aHEV), a nonenveloped, single-stranded RNA virus. The economic importance of this disease in the poultry industry is due to the decline in egg production (10%-40%) and the rise in mortality (1%-4%). In the present study, 1540 serum samples from 33 broiler breeder flocks were analyzed by an enzyme-linked immunosorbent assay for the presence of an anti-aHEV antibody. In addition, a diagnostic nested reverse transcriptase-PCR was done on all farm samples. In the serologic study, 66.7% (22/33) of the flocks and 28.5% (439/1540) of the chickens were positive. The molecular study showed that three farms were positive, and PCR products were observed for the conserved regions of the aHEV helicase and capsid virus genes as 386 bp and 242 bp, respectively. It should be noted that clinical and pathologic symptoms including decreased egg production, enlarged livers and spleens, and a slight rise in mortality rate were observed in eight farms. To our knowledge, this is the first documented study on the aHEV identification and its antibody detection in broiler breeder farms in Iran.


Evidencia serológica y molecular de una infección diseminada del virus de la hepatitis E aviar en granjas avícolas en Irán. El síndrome de hepatitis-esplenomegalia es causado por el virus de la hepatitis E aviar (aHEV), un virus de ARN de cadena simple sin envoltura. La importancia económica de esta enfermedad en la industria avícola se debe a la disminución en la producción de huevo (10%-40%) y al aumento de la mortalidad (1%-4%). En el presente estudio, se analizaron 1540 muestras de suero de 33 parvadas de reproductores pesados mediante un ensayo de immunoabsorción con enzimas ligadas para determinar la presencia de anticuerpos contra el virus de la hepatitis E aviar. Además, se realizó un método de transcripción reversa y PCR anidado de diagnóstico en todas las muestras de la granja. En el estudio serológico, el 66.7% (22/33) de las parvadas y el 28.5% (439/1540) de los pollos fueron positivos. El estudio molecular mostró que tres granjas fueron positivas, y se observaron productos de PCR para las regiones conservadas de los genes del virus de la cápside y de la helicasa del virus de la hepatitis E aviar con tamaños de 386 pb y 242 pares de bases, respectivamente. Cabe señalar que en ocho granjas se observaron signos clínicos y patológicos como disminución de la producción de huevos, agrandamiento del hígado y del bazo y un ligero aumento en la tasa de mortalidad. Hasta donde se conoce, este es el primer estudio documentado sobre la identificación del virus de la hepatitis E aviar y la detección de anticuerpos en granjas de pollos de engorde en Irán.


Assuntos
Hepatite Viral Animal , Hepevirus , Doenças das Aves Domésticas , Animais , Galinhas , Fazendas , Hepatite Viral Animal/diagnóstico , Hepatite Viral Animal/epidemiologia , Hepevirus/genética , Irã (Geográfico)/epidemiologia , Aves Domésticas , Doenças das Aves Domésticas/patologia
17.
Poult Sci ; 99(12): 6657-6663, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33248582

RESUMO

In recent years, the avian hepatitis E virus (HEV) has been widely spread in China, causing huge economic losses. Several studies have carried out detailed epidemiologic investigations of the avian HEV, but no data were from Jiangxi province. Since early April 2020, diseases similar to hepatic rupture hemorrhage syndrome caused by the avian HEV occurred in a Roman Brown layer farm in Jiangxi province, indicating this virus may also be epidemic there. To make this assumption clear, 20 liver samples were collected from the sick flock and then analyzed by detailed viral detection, which confirmed that the avian HEV should be responsible for the aforementioned disease (6 of 20). Then, the capsid gene of the virus was sequenced to show the molecular characteristics of the strain circulating in the aforementioned flock. Sequence comparison showed that it shared 80.7 to 94.7% identities with 12 published strains, while phylogenetic analysis confirmed that it belongs to a new subtype of genotype 3. Moreover, basing on a 242 bp fragment, the novel also shared high similarities to reference strains identified as genotypes before, revealing the genotype 3 maybe very popular in China and even can be divided into several subgroups. In conclusion, a novel avian HEV strain was identified in this study, which belongs to a new subtype of genotype 3. The analysis makes up for the molecular epidemiologic data of avian HEV and provides a basis for further understanding the spread of avian HEV in China.


Assuntos
Hepatite Viral Animal , Hepevirus , Doenças das Aves Domésticas , Infecções por Vírus de RNA , Animais , Galinhas , China , Genótipo , Hepatite Viral Animal/virologia , Hepevirus/classificação , Hepevirus/genética , Filogenia , Doenças das Aves Domésticas/virologia , Infecções por Vírus de RNA/veterinária , Infecções por Vírus de RNA/virologia
18.
J Virol Methods ; 283: 113907, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32502499

RESUMO

Characterisation of the entire genome of Fowl aviadenoviruses (FAdV) requires isolation and propagation of the virus in chicken embryo liver or kidney cells, a process which is not only time consuming but may occasionally fail to result in viral growth. Furthermore, in a mixed infection, isolation in cell culture may result in the loss of viral strains. In this study, we optimised a FAdV DNA extraction technique directly from affected liver tissues using kaolin hydrated aluminium silicate treatment. The whole genome of FAdV was sequenced directly from extracted DNA without any targetted PCR based enrichment. The extraction method was also tested on avian liver tissues affected with the RNA virus Avian hepatitis E virus and demonstrated to yield sequencing grade RNA. Therefore, the method described here is a simple technique which is potentially useful for the extraction of sequencing grade DNA/RNA from tissues with high fat content.


Assuntos
Aviadenovirus/genética , DNA Viral/isolamento & purificação , Fígado/virologia , RNA Viral/isolamento & purificação , Sequenciamento Completo do Genoma/métodos , Infecções por Adenoviridae/virologia , Animais , Aviadenovirus/isolamento & purificação , Galinhas/virologia , Genoma Viral , Hepatite , Hepevirus/genética , Sequenciamento de Nucleotídeos em Larga Escala
19.
Transbound Emerg Dis ; 67(6): 2403-2407, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32304264

RESUMO

Avian hepatitis E virus (HEV) is highly variable and has multiple genotypes. Previous studies showed that the current epidemic strain in China is genotype 5, but the relevant detection was only carried out in flocks with hepatic rupture haemorrhage syndrome, which does not mean that other genotypes do not exist. In this study, a broader analysis involving different chicken flocks was performed to understand the epidemic status of avian HEV in China. The results showed that the HEV-positive rate of all samples was 7.92% (78/985), and four different genotypes have been identified by analysing a truncate capsid gene fragment, while the homology between them is about 80%. Two of them are separately known as genotype 3 and genotype 5, while the other two are completely unidentified, indicating that there are multiple genotypes of avian HEV prevalent in China. At the same time, the distribution of these genotypes has no obvious geographical clustering pattern, only slightly different in commercial layers, broilers and some indigenous species. This study shows the genetic diversity of avian HEV in China and reminds us to pay more attention to its variation and evolution.


Assuntos
Galinhas/virologia , Variação Genética , Hepatite Viral Animal/virologia , Hepevirus/genética , Doenças das Aves Domésticas/virologia , Infecções por Vírus de RNA/veterinária , Animais , Proteínas do Capsídeo/genética , China/epidemiologia , Genótipo , Hepatite Viral Animal/epidemiologia , Filogenia , Doenças das Aves Domésticas/epidemiologia , Prevalência , Infecções por Vírus de RNA/epidemiologia , Infecções por Vírus de RNA/virologia
20.
Vet Microbiol ; 242: 108577, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32122587

RESUMO

Since 2016, severe outbreaks of hepatic rupture hemorrhage syndrome (HRHS) associated with infections of tentative novel avian hepatitis E virus (HEV) have emerged in chickens in China, causing increased mortality and decreased laying rate in adult hens and disturbing the hatching and breeding of chicks. To further identify the genotype and gain a better understanding of the genetic properties of the avian HEV responsible for that, a strain from Hebei province was isolated, purified and sequenced in this study. Results identified a novel avian HEV genotype, sharing 79.5-86.9% identities with other published avian HEV strains, and having higher identities with Orthohepevirus A HEV strains. More importantly, the new isolate contains various amino-acid substitutions in its functional proteins, including methyltransferase, helicase, RNA-dependent RNA polymerase. The data presented in this report will enhance the current understanding of the genetic diversity of the avian HEV and provide additional insight into the critical factors that determine the pathogenicity.


Assuntos
Genoma Viral , Hemorragia/veterinária , Hepatite Viral Animal/virologia , Hepevirus/genética , Animais , Galinhas/virologia , China , Fazendas , Variação Genética , Genótipo , Hemorragia/virologia , Hepatite Viral Animal/complicações , Hepevirus/patogenicidade , Fígado/patologia , Fígado/virologia , Mutação , Filogenia , Doenças das Aves Domésticas/virologia , Análise de Sequência de DNA , Sequenciamento Completo do Genoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...