Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 584
Filtrar
1.
Vet Res ; 55(1): 40, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38532469

RESUMO

The interaction between viral components and cellular proteins plays a crucial role in viral replication. In a previous study, we showed that the 3'-untranslated region (3'-UTR) is an essential element for the replication of duck hepatitis A virus type 1 (DHAV-1). However, the underlying mechanism is still unclear. To gain a deeper understanding of this mechanism, we used an RNA pull-down and a matrix-assisted laser desorption/ionization time-of-flight mass spectrometry assay to identify new host factors that interact with the 3'-UTR. We selected interleukin-2 enhancer binding factor 2 (ILF2) for further analysis. We showed that ILF2 interacts specifically with both the 3'-UTR and the 3D polymerase (3Dpol) of DHAV-1 through in vitro RNA pull-down and co-immunoprecipitation assays, respectively. We showed that ILF2 negatively regulates viral replication in duck embryo fibroblasts (DEFs), and that its overexpression in DEFs markedly suppresses DHAV-1 replication. Conversely, ILF2 silencing resulted in a significant increase in viral replication. In addition, the RNA-dependent RNA polymerase (RdRP) activity of 3Dpol facilitated viral replication by enhancing viral RNA translation efficiency, whereas ILF2 disrupted the role of RdRP in viral RNA translation efficiency to suppress DHAV-1 replication. At last, DHAV-1 replication markedly suppressed the expression of ILF2 in DEFs, duck embryo hepatocytes, and different tissues of 1 day-old ducklings. A negative correlation was observed between ILF2 expression and the viral load in primary cells and different organs of young ducklings, suggesting that ILF2 may affect the viral load both in vitro and in vivo.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Interleucina-2/genética , RNA Polimerase Dependente de RNA/genética , Regulação da Expressão Gênica , RNA Viral/genética , Patos/genética , Infecções por Picornaviridae/veterinária
2.
Infect Genet Evol ; 120: 105585, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38508364

RESUMO

In this study, a picornavirus and a nidovirus were identified from a single available nasopharyngeal swab (NPS) sample of a freshly deceased sheep, as the only vertebrate viruses found with viral metagenomics and next-generation sequencing methods. The sample was originated from a mixed feedlot farm in Hungary where sheep and cattle were held together but in separate stalls. Most of the sheep had respiratory signs (coughing and increased respiratory effort) at the time of sampling. Other NPS were not, but additional enteric samples were collected from sheep (n = 27) and cattle (n = 11) of the same farm at that time. The complete/nearly complete genomes of the identified viruses were determined using RT-PCR and Nanopore (MinION-Flonge) / Dye-terminator sequencing techniques. The results of detailed genomic and phylogenetic analyses indicate that the identified picornavirus most likely belongs to a type 4 genotype of species Bovine rhinitis B virus (BRBV-4, OR885914) of genus Aphthovirus, family Picornaviridae while the ovine nidovirus (OvNV, OR885915) - as a novel variant - could belong to the recently created Bovine nidovirus 1 (BoNV) species of genus Bostovirus, family Tobaniviridae. None of the identified viruses were detectable in the enteric samples using RT-PCR and generic screening primer pairs. Both viruses are well-known respiratory pathogens of cattle, but their presence was not demonstrated before in other animals, like sheep. Furthermore, neither BRBV-4 nor BoNVs were investigated in European cattle and/or sheep flocks, therefore it cannot be determined whether the presence of these viruses in sheep was a result of a single host species switch/spillover event or these viruses are circulating in not just cattle but sheep populations as well. Further studies required to investigate the spread of these viruses in Hungarian and European sheep and cattle populations and to identify their pathogenic potential in sheep.


Assuntos
Filogenia , Infecções por Picornaviridae , Picornaviridae , Doenças dos Ovinos , Animais , Hungria , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Picornaviridae/classificação , Ovinos , Doenças dos Ovinos/virologia , Bovinos , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/virologia , Coinfecção/virologia , Coinfecção/veterinária , Genoma Viral , Nidovirales/genética , Nidovirales/isolamento & purificação , Nidovirales/classificação , Infecções por Nidovirales/veterinária , Infecções por Nidovirales/virologia
3.
Poult Sci ; 103(3): 103416, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38301494

RESUMO

As the most prevalent pathogen of duck viral hepatitis (DVH), duck hepatitis A virus genotype 3 (DHAV-3) has caused huge economic losses to the duck industry in China. Herein, we obtained whole-transcriptome sequencing data of susceptible (S) and resistant (R) Pekin duckling samples at 0 h, 12 h, and 24 h after DHAV-3 infection. We found that DHAV-3 infection induces 5,396 differentially expressed genes (DEGs), 85 differentially expressed miRNAs (DEMs), and 727 differentially expressed lncRNAs (DELs) at 24 hpi in S vs. R ducks, those upregulated genes were enriched in inflammation and cell communications pathways and downregulated genes were related to metabolic processes. Upregulated genes showed high connectivity with the miR-33, miR-193, and miR-11591, and downregulated genes were mainly regulated by miR-2954, miR-125, and miR-146b. With the construction of lncRNA-miRNA-mRNA axis, we further identified a few aberrantly expressed lncRNAs (e.g., MSTRG.36194.1, MSTRG.50601.1, MSTRG.34328.7, and MSTRG.29445.1) that regulate expression of hub genes (e.g., THBD, CLIC2, IL8, ACOX2, GPHN, SMLR1, and HAO1) by sponging those highly connected miRNAs. Altogether, our findings defined a dual role of ncRNAs in immune and metabolic regulation during DHAV-3 infection, suggesting potential new targets for treating DHAV-3 infected ducks.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , MicroRNAs , Infecções por Picornaviridae , RNA Longo não Codificante , Animais , Vírus da Hepatite do Pato/fisiologia , Patos/genética , Transcriptoma , RNA Longo não Codificante/genética , Infecções por Picornaviridae/veterinária , Galinhas/genética , Genótipo , MicroRNAs/genética
4.
Vet Microbiol ; 289: 109958, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38181600

RESUMO

Neonatal mortality has been increasingly reported on swine breeding farms experiencing swine idiopathic vesicular disease (SIVD) outbreaks, which can be accompanied by lethargy, diarrhea, and neurologic signs in neonates. Seneca Valley Virus (SVV), or Senecavirus A, has been detected in clinical samples taken from pigs with SIVD. Experimental SVV inoculation has caused vesicular disease in pigs, particularly during the stages from weaning to finishing. However, it remains crucial to investigate whether SVV directly contributes to the increase in neonatal mortality rates. The following study was conducted to chronicle the pathogenesis of SVV infection in sows and their offspring. Ten sows were intranasally inoculated with 4.75 × 107 plaque-forming units of the virus per sow either late in gestation (n = 5) or within fourteen days of farrowing (n = 5). Each sow replicated SVV following intranasal inoculation, but only one out of ten sows developed a vesicular lesion on the snout. Evidence of transplacental infection was observed in two litters, and an additional two litters became infected following parturition out of five litters from sows inoculated in late gestation. No clinical signs were observed in the infected neonates. Likewise, no clinical signs were observed in the other five litters inoculated after farrowing, although each piglet did replicate the challenge virus. In this study, the experimental challenge of SVV did not result in neonatal mortality in contrast to observations in the field; however, it has shed light on the pathogenesis of the virus, the transmission of SVV between sows and their offspring, and host immune response that can help shape control measures in the field.


Assuntos
Infecções por Picornaviridae , Picornaviridae , Doenças dos Suínos , Suínos , Animais , Feminino , Gravidez , Infecções por Picornaviridae/veterinária , Surtos de Doenças/veterinária
5.
Vet Microbiol ; 290: 109987, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38246107

RESUMO

The duck hepatitis A virus type 1 (DHAV-1) causes rapid death in ducklings by triggering a severe cytokine storm. Pyroptosis is an inflammatory form of programmed cell death that is directly related to an increase in pro-inflammatory cytokine levels. Only a few studies have explored the mechanisms underlying pyroptosis in virus-infected avian cells. In this study, we established an avian infection model in vitro by infecting duck embryo fibroblasts (DEFs) with the virulent DHAV-1 LY0801 strain. DHAV-1 infection induced pyroptosis in the DEFs by activating gasdermin E (GSDME) protein via caspase-3-mediated cleavage. The genes encoding the different structural and non-structural DHAV-1 proteins were cloned into eukaryotic expression plasmids, and the 2A2 protein was identified as the key protein involved in pyroptosis. The HPLC-tandem mass spectrometry (HPLC-MS/MS) and co-immunoprecipitation (Co-IP) analysis established that DHAV-1 2A2 directly interacted with the mitochondrial anti-viral signaling protein (MAVS) both intracellularly and in vitro. Furthermore, we got the results that N-terminal 1-130 aa of 2A2 was involved in the interaction with MAVS and the C-terminal TM domain of MAVS is necessary for the interaction with 2A2 by Co-IP analysis. To our knowledge, this is the first study to reveal that DHAV-1 protein interacts with host proteins to induce pyroptosis. Our findings provide new insights into the molecular pathogenesis of DHAV-1 infection, and a scientific basis for the prevention and treatment of duck viral hepatitis.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Patos , Gasderminas , Piroptose , Espectrometria de Massas em Tandem/veterinária , Fibroblastos , Infecções por Picornaviridae/veterinária
6.
Poult Sci ; 103(2): 103265, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38042039

RESUMO

Duck hepatitis A virus type 1 (DHAV-1) can cause severe liver damage in infected ducklings and is a fatal and contagious pathogen that endangers the Chinese duck industry. The objective of this study was to explore the correlation mechanism of liver metabolism-gut microbiota in DHAV-1 infection. Briefly, liquid chromatography-mass spectrometry and 16S rDNA sequencing combined with multivariate statistical analysis were used to evaluate the effects of DHAV-1 infection on liver metabolism, gut microbiota regulation, and other potential mechanisms in ducklings. In DHAV-1-infected ducklings at 72 h postinfection, changes were found in metabolites associated with key metabolic pathways such as lipid metabolism, sugar metabolism, and nucleotide metabolism, which participated in signaling networks and ultimately affecting the function of the liver. The abundance and composition of gut microbiota were also changed, and gut microbiota is significantly involved in lipid metabolism in the liver. The evident correlation between gut microbiota and liver metabolites indicates that DHAV-host gut microbiome interactions play important roles in the development of duck viral hepatitis (DVH).


Assuntos
Microbioma Gastrointestinal , Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Vírus da Hepatite do Pato/fisiologia , Patos , Infecções por Picornaviridae/veterinária , Galinhas
7.
BMC Vet Res ; 19(1): 179, 2023 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-37773135

RESUMO

BACKGROUND: Duck hepatitis A virus (DHAV) is a single-stranded, positive-strand small RNA virus that causes a very high mortality rate in ducklings. The DHAV-3 subtype incidence rate has recently increased in China, causing great economic losses to the waterfowl breeding industry. We analyzed the protection rate of DHAV vaccines used in mainland China from 2009 to 2021 and evaluated the effectiveness of vaccine prevention and control to reduce the economic losses caused by DHAV to the waterfowl breeding industry. We screened five electronic research databases and obtained 14 studies and patents on the protection efficiency of DHAV-1 and DHAV-3 vaccines. RESULTS: Meta-analysis demonstrated that immunized ducklings produced higher antibody levels and had a significantly higher survival rate than non-immunized ducklings [relative risk (RR) = 12, 95% confidence interval (CI) 6-26, P < 0.01]. The age of the ducks and vaccine valence did not affect protection efficiency. Data source analysis of the vaccine protection rate demonstrated that the vaccines conferred immune protection for ducklings in both small-scale experiments and large-scale clinical conditions. The analysis results revealed that although the vaccines conferred protection, the immune protective effect differed between small-scale experimental conditions and large-scale clinical conditions. This might have been due to non-standard vaccination and environmental factors. CONCLUSIONS: Domestic DHAV vaccines can protect ducklings effectively. The subjects immunized (breeding ducks or ducklings) and vaccine valence had no effect on the protective effect. Both small-scale experiments and large-scale clinical conditions conferred immune protection on ducklings, but vaccine immunization under small-scale experimental conditions had slightly better protective effects than large-scale clinical immunization.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Vacinas , Animais , Infecções por Picornaviridae/veterinária , Vacinação/veterinária , China/epidemiologia , Patos
8.
J Vet Sci ; 24(3): e48, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37271515

RESUMO

BACKGROUND: Senecavirus A (SVA), a member of the family Picornaviridae, is newly discovered, which causes vesicular lesions, lameness in swine, and even death in neonatal piglets. SVA has rapidly spread worldwide in recent years, especially in Asia. OBJECTIVES: We conducted a global meta-analysis and systematic review to determine the status of SVA infection in pigs. METHODS: Through PubMed, VIP Chinese Journals Database, China National Knowledge Infrastructure, and Wanfang Data search data from 2014 to July 26, 2020, a total of 34 articles were included in this analysis based on our inclusion criteria. We estimated the pooled prevalence of SVA in pigs by the random effects model. A risk of bias assessment of the studies and subgroup analysis to explain heterogeneity was undertaken. RESULTS: We estimated the SVA prevalence to be 15.90% (1,564/9,839; 95% confidence interval [CI], 44.75-65.89) globally. The prevalence decreased to 11.06% (945/8,542; 95% CI, 28.25-50.64) after 2016. The highest SVA prevalence with the VP1-based RT-PCR and immunohistochemistry assay was 58.52% (594/1,015; 95% CI, 59.90-83.96) and 85.54% (71/83; 95% CI, 76.68-100.00), respectively. Besides, the SVA prevalence in piglet herds was the highest at 71.69% (119/166; 95% CI, 68.61-98.43) (p < 0.05). Moreover, our analysis confirmed that the subgroups, including country, sampling year, sampling position, detected gene, detection method, season, age, and climate, could be the heterogeneous factors associated with SVA prevalence. CONCLUSIONS: The results indicated that SVA widely exists in various countries currently. Therefore, more prevention and control policies should be proposed to enhance the management of pig farms and improve breeding conditions and the environment to reduce the spread of SVA.


Assuntos
Infecções por Picornaviridae , Picornaviridae , Doenças dos Suínos , Animais , Suínos , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/veterinária , Prevalência , Picornaviridae/genética
9.
Res Vet Sci ; 159: 232-236, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37172452

RESUMO

Avian encephalomyelitis (AE) is a highly infectious disease caused by the avian encephalomyelitis virus (AEV), which primarily affects the central nervous system of 1- to 4-week-old chicks and causes significant economic losses in the worldwide poultry industry. Despite heavy dependency on vaccine immunization, AEV has persisted on farms for extended periods, which increases its virulence and makes quick and accurate detection crucial to preventing and controlling the disease. Classical diagnostic methods have been unable to meet the current requirements for rapid diagnosis of AE cases. To address this issue, this paper reviews the etiological and molecular biological detection techniques of AE, and it seeks to provide a reference for future research and to establish differential diagnostic techniques for AE epidemiological investigation, identification of epidemic strains, and early diagnosis of clinical cases. Through improving our understanding of AE, we can better combat the disease and protect the global poultry industry.


Assuntos
Encefalite Viral , Vírus da Encefalomielite Aviária , Encefalomielite , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Doenças das Aves Domésticas/prevenção & controle , Galinhas , Infecções por Picornaviridae/veterinária , Encefalite Viral/veterinária , Encefalomielite/veterinária
10.
Poult Sci ; 102(7): 102724, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37207573

RESUMO

The placenta-specific 8 (PLAC8) gene, also known as ONZIN or C15, codes for a cysteine-rich peptide originally identified in mouse placental tissue and subsequently identified in a variety of epithelial tissues and immune cells. PLAC8 is also expressed in birds, such as ducks, where its functional roles remain unknown. Here, we aimed to determine the mRNA and protein expression profiles and the functional role of duck PLAC8 during the infection of duck hepatitis A virus type 1 (DHAV-1). We found that the duck PLAC8 is also a cysteine-rich polypeptide composed of 114 amino acid residues, with no signal peptide. Duck PLAC8 is highly expressed in the immune organs of young cherry valley ducks, including the thymus, bursa fabricius, and spleen. However, it has negligible expression level in liver, brain, kidney, and heart. Additionally, PLAC8 expression was considerably induced after DHAV-1 infection both in vitro and in vivo, especially in the immune organs of ducklings. This tissue expression distribution and induction upon infection suggest that PLAC8 might play a critical role in innate immunity. Our data showed that PLAC8 significantly suppressed the expression of Toll-like receptor 7 (TLR7), leading to decreased expression of downstream signaling molecules including myeloid differentiation primary response gene 88 (MyD88) and nuclear factor kappa-B (NF-κB). This ultimately resulted in low levels of type I interferon and interleukin 6 (IL-6). Additionally, PLAC8 positively regulated DHAV-1 replication levels. RNAi against PLAC8 in duck embryo fibroblasts considerably inhibited DHAV-1 propagation, while PLAC8 overexpression significantly facilitated DHAV-1 replication.


Assuntos
Patos , Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Feminino , Camundongos , Gravidez , Cisteína , Patos/genética , Patos/virologia , Fator 88 de Diferenciação Mieloide/genética , Infecções por Picornaviridae/veterinária , Placenta , Transdução de Sinais , Receptor 7 Toll-Like/genética , Células HEK293 , Humanos
11.
Viruses ; 15(1)2023 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-36680281

RESUMO

The newly identified porcine Kobuvirus (PKV) has raised concerns owing to its association with diarrheal symptom in pigs worldwide. The process involving the emergence and global spread of PKV remains largely unknown. Here, the origin, genetic diversity, and geographic distribution of PKV were determined based on the available PKV sequence information. PKV might be derived from the rabbit Kobuvirus and sheep were an important intermediate host. The most recent ancestor of PKV could be traced back to 1975. Two major clades are identified, PKVa and PKVb, and recombination events increase PKV genetic diversity. Cross-species transmission of PKV might be linked to interspecies conserved amino acids at 13-17 and 25-40 residue motifs of Kobuvirus VP1 proteins. Phylogeographic analysis showed that Spain was the most likely location of PKV origin, which then spread to pig-rearing countries in Asia, Africa, and Europe. Within China, the Hubei province was identified as a primary hub of PKV, transmitting to the east, southwest, and northeast regions of the country. Taken together, our findings have important implications for understanding the evolutionary origin, genetic recombination, and geographic distribution of PKV thereby facilitating the design of preventive and containment measures to combat PKV infection.


Assuntos
Kobuvirus , Infecções por Picornaviridae , Doenças dos Suínos , Suínos , Animais , Coelhos , Ovinos , Filogeografia , Kobuvirus/genética , Filogenia , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/diagnóstico , Recombinação Genética
12.
Vet Res Commun ; 47(3): 1231-1241, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36595200

RESUMO

Sudden death of ducklings was reported in a duck farm located at Tiruvallur district in Tamil Nadu, India. Disease investigation began with post mortem findings of dead birds revealing enlarged pale-pink / pale-yellow liver with multifocal petechiae and ecchymosis. A positive amplification with duck hepatitis A virus specific primers by reverse transcription-polymerase chain reaction (RT-PCR) on the tissue samples collected from dead birds indicated infection by duck hepatitis A virus (DHAV), an avian picornavirus, known to cause acute and high-mortality in ducklings. The virus isolation was successful in 9-days old embryonated chicken eggs, in primary chicken embryo fibroblast (CEF) cells and from experimentally infected ducklings. The embryonic death on day 5 to 7 post inoculation in chicken embryos with signs of cutaneous hemorrhage, edema and greenish yellow liver together with histopathology of embryonic liver and kidney further confirmed DHAV infection. TEM analysis of the infected allantoic fluid and infected CEF cell culture supernatant showed the presence of spherical shaped, non-enveloped virion particles of ~ 20-38 nm diameter, typical for DHAV. Experimental infection of ducklings with RT-PCR positive tissue supernatant caused 40% to 50% mortality with typical petechial hemorrhages on the surface of liver. Further, histopathological analysis and RT-PCR of the inoculated duckling's tissues confirmed the presence of DHAV. Nucleotide sequencing of the 5'UTR region and VP1 region confirmed duck hepatitis A virus genotype 2 (DHAV-2). To the best of our knowledge, this is the first report of laboratory confirmation of DHAV-2 in India. This study warrants the need for the extensive epidemiological surveillance to understand the prevalence of DHAV-2 in India and to take appropriate control measures to curtail the disease spread.


Assuntos
Vírus da Hepatite do Pato , Infecções por Picornaviridae , Doenças das Aves Domésticas , Embrião de Galinha , Animais , Vírus da Hepatite do Pato/genética , Índia/epidemiologia , Doenças das Aves Domésticas/epidemiologia , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/veterinária , Patos , Genótipo
13.
BMC Vet Res ; 19(1): 2, 2023 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-36597091

RESUMO

BACKGROUND: Porcine Teschovirus (PTV), also named Teschovirus A, is prevalent in pig populations, mainly causing neurological symptoms, diarrhea, pneumonia, and reproductive failure, however the morbidity and mortality are usually low in pig farms. CASE PRESENTATION: In this study, we reported a PTV outbreak investigation in one large-scale pig farm in China with severe symptoms including diarrhea, lethargy, locomotor ataxia, nystagmus, paralysis of the hind limbs, and coma in piglets. More importantly, the mortality reached 38% in suckling pigs, which is remarkably high in PTV history. A novel PTV strain, named HeNZ1, was isolated from cerebral samples of one suckling pig and the genome sequence was obtained by NGS sequencing. Phylogenetic and evolutionary divergence analyses revealed that HeNZ1 belongs to PTV genotype 2. Surprisingly, the VP1 coding region of HeNZ1 shares the highest sequence similarity with European PTV-2 strains, instead of China domestic PTV-2 strains, implying it may not derive from China local PTV-2 strains. Multiple sequence alignment and B cell epitope prediction of PTV VP1 and VP2 protein revealed 10 B cell epitopes, 5 mutant clusters and 36 unique mutation sites, of which 19 unique mutation sites are located in B cell epitopes and exposed on the surface of VP1 or VP2, implying significant antigenic drift potential of HeNZ1. CONCLUSION: These results indicate that HeNZ1 is a highly virulent PTV-2 strain, which capable of causing severe neurological symptoms and high mortality in piglets. Bioinformatic analysis suggest that HeNZ1 is genetically and antigenically different from other Chinese PTV-2 strains. Overall, current case expanded our understanding of PTV-2 clinical spectrum and revealed the emergence of a highly virulent PTV-2 strain with substantial genetic diversity and antigenic drift potential in VP1 and VP2.


Assuntos
Encefalomielite , Infecções por Picornaviridae , Doenças dos Suínos , Teschovirus , Suínos , Animais , Filogenia , Epitopos de Linfócito B , Diarreia/veterinária , China/epidemiologia , Encefalomielite/veterinária , Infecções por Picornaviridae/veterinária
14.
Vet Microbiol ; 277: 109621, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36525908

RESUMO

Duck hepatitis A virus type 1 (DHAV-1) infection causes an acute and highly fatal disease in young ducklings. Exosomes are nano-sized small extracellular vesicles secreted by various cells, which participate in intercellular communication and play a key role in the physiological and pathological processes. However, the role of exosomes in DHAV-1 transmission remains unknown. In this study, through RT-PCR, WB analysis and TEM observation, the complete DHAV-1 genomic RNA, partial viral proteins, and virions were respectively identified in the exosomes derived from DHAV-1-infected duck embryo fibroblasts (DEFs). The productive DHAV-1 infection was transmitted by exosomes in DEFs, duck embryos, and ducklings, and high titers of neutralizing antibodies completely blocked DHAV-1 infection but did not significantly neutralize exosome-mediated DHAV-1 infection. To the best of our knowledge, this is the first report that exosome-mediated DHAV-1 infection was resistant to antibody neutralization in vivo and in vitro, which might be an immune evasion mechanism of DHAV-1.


Assuntos
Exossomos , Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Vírus da Hepatite do Pato/genética , Exossomos/patologia , Infecções por Picornaviridae/veterinária , Patos
15.
Avian Pathol ; 52(2): 89-99, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36571394

RESUMO

Duck viral hepatitis (DVH), mainly caused by duck hepatitis A virus (DHAV), is a highly fatal and rapidly spreading infectious disease of young ducklings that seriously jeopardizes the duck industry worldwide. DHAV type 1 (DHAV-1) is the main genotype responsible for disease outbreaks since 1945, and the disease situation is complicated by the emergence and dissemination of a novel genotype (DHAV-3) in some countries in Asia and Africa. Live attenuated DHAV vaccines are widely used to induce a considerable degree of protection in ducklings. Breeder ducks are immunized with inactivated or/and live DHAV vaccines to achieve satisfactory levels of passive immunity in progeny. In addition, novel characteristics of virus transmission, pathogenicity and pathogenesis of DHAV were recently characterized, necessitating the development of new vaccines and effective vaccination programmes against DVH. Therefore, a systematic dissection of the profiles, strengths and shortcomings of the available DHAV vaccines is essential. Moreover, to further increase the efficiency of vaccine production and administration, the development of next-generation DHAV vaccines using cutting-edge technologies is also required. In this review, based on a comprehensive summary of the research advances in the epidemiology, pathogenicity, and genomic features of DHAV, we focus on reviewing and analysing the features of the commercial and experimental DHAV vaccines. We also propose perspectives for disease control based on the specific disease situations in different countries. This review provides essential information for vaccine development and disease control of DVH.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Infecções por Picornaviridae/veterinária , Vacinas Atenuadas , Vacinação/veterinária , Patos
16.
Transbound Emerg Dis ; 69(6): e3462-e3468, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36327129

RESUMO

Seneca Valley virus (SVV) is a non-enveloped RNA virus and the only member of the Senecavirus A (SVA) species, in the Senecavirus genus, Picornaviridae family. SVV infection causes vesicular lesions in the oral cavity, snout and hooves of pigs. This infection is clinically indistinguishable from trade-restrictions-related diseases such as foot-and-mouth disease. Other clinical manifestations include diarrhoea, anorexia, lethargy, neurological signs and mortality in piglets during their first week of age. Before this study, Chile was considered free of vesicular diseases of swine, including SVV. In April 2022, a suspected case of vesicular disease in a swine farm was reported in Chile. The SVV was confirmed and other vesicular diseases were ruled out. An epidemiological investigation and phylogenetic analyses were performed to identify the origin and extent of the outbreak. Three hundred ninety-five samples from 44 swine farms were collected, including faeces (208), oral fluid (28), processing fluid (14), fresh semen (61), environmental samples (80) and tissue from lesions (4) for real-time RT-PCR detection. Until June 2022, the SVV has been detected in 16 out of 44 farms, all epidemiologically related to the index farm. The closest phylogenetic relationship of the Chilean SVV strain is with viruses collected from swine in California in 2017. The direct cause of the SVV introduction has not yet been identified; however, the phylogenetic analyses suggest the USA as the most likely source. Since the virus remains active in the environment, transmission by fomites such as contaminated feed cannot be discarded. Further studies are needed to determine the risk of the introduction of novel SVV and other transboundary swine pathogens to Chile.


Assuntos
Infecções por Picornaviridae , Picornaviridae , Doenças dos Suínos , Animais , Suínos , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/diagnóstico , Chile/epidemiologia , Filogenia , Picornaviridae/genética , RNA Viral
17.
Infect Genet Evol ; 104: 105362, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36084837

RESUMO

Porcine kobuvirus (PKV) infection is very common in both healthy pigs and diarrhea pigs throughout the world. However, there is no proof that it causes diarrhea, and little is known about its role in diarrhea. There are only a few reports concerning porcine kobuvirus separation at present, which makes investigating its invasion and pathogenesis mechanisms difficult. This study sequenced the entire genome of a porcine kobuvirus strain termed "Wuhan2020" after it was isolated from intestinal tissue samples of healthy piglets. The analysis results revealed that it shared the most resemblance with the WUH1 strain (89.5%) and belonged to the same evolutionary branch as the Hungarian strain S-1-SUN. The PKV was located using the in situ hybridization (ISH) approach, which revealed that it was colonized in intestinal villus epithelial cells and lymphocytes in the Peyer's patch. In general, we analyzed the genetic evolution of PKV, discovered PKV susceptible cells and determined PKV localization in the intestine of infected pigs, providing a reference for future research.


Assuntos
Kobuvirus , Infecções por Picornaviridae , Doenças dos Suínos , Animais , China , Diarreia , Fezes , Genômica , Intestinos , Kobuvirus/genética , Filogenia , Infecções por Picornaviridae/veterinária , Análise de Sequência , Suínos
18.
Virus Res ; 322: 198930, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36130655

RESUMO

BACKGROUND: Duck viral hepatitis (DVH) has a significant economic impact on duck industry, and duck hepatitis A virus genotype 3 (DHAV-3) is the most prevalent pathogen of DVH in Asian duck industry. The detailed study connecting differentially expressed genes (DEGs) and the differential resistance to DHAV-3 have not been accurately described, although a large numbers of DEGs have been identified by transcriptomic studies. RESULTS: Here, a resistant Pekin duck line (Z8R) and a susceptible Pekin duck line (Z8S) as models, high mortality and dramatically increased aspartate aminotransferase (AST), alanine aminotransferase (ALT) and the expression of immune-related genes of Z8S group were shown to be noticeable signs of cases caused by DHAV-3 infection. Compared with the control (Con) group, 1117 down-regulated DEGs and 612 up-regulated DEGs were found in the Z8S group and 37 down-regulated DEGs and 82 up-regulated DEGs were found in the Z8R group. Ultimately, the expression patterns of 10 DEGs were found to be diametrically opposite in Z8R and Z8S group. Functional analysis revealed that IFITM1 was associated with cell growth suppression, which was considered a key candidate gene. Results of flow cytometry showed that the conserved regions of IFITM1 (213-317 bp) could affected the cell cycle of duck embryo fibroblast (DEF) cells after infection with DHAV-3. Transcriptome and western blot analysis suggested that the CCND1, CCNE1 and CDK6 were significantly up-regulated in susceptible ducks by comparing with Con group. CONCLUSIONS: The hepatic injury and pathogenic outcomes caused by DHAV-3 infection were more severe in Z8S group compared to Z8R. Results of transcriptomics analysis and flow cytometry suggested that DHAV-3 infection can induce cell cycle changes that may be associated with IFITM1 expression level. These data will greatly enhance our understanding of the pathogenesis of DHAV-3 infection in ducklings and have implications for development of resistance breeding.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Patos , Genótipo , Vírus da Hepatite do Pato/genética , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/veterinária , Infecções por Picornaviridae/patologia , Transcriptoma
19.
Vet Res ; 53(1): 64, 2022 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-35978392

RESUMO

Duck hepatitis A virus type 1 (DHAV-1) is one of the main pathogens responsible for death in ducklings. Autophagy is a catabolic process that maintains cellular homeostasis, and the PI3KC3 protein plays an important role in the initiation of autophagy. DHAV-1 infection induces autophagy in duck embryo fibroblasts (DEFs) but the molecular mechanism between it and autophagy has not been reported. First, we determined that DHAV-1 infection induces autophagy in DEFs and that autophagy induction is dependent on the integrity of viral proteins by infecting DEFs with UV-inactivated or heat-inactivated DHAV-1. Then, in experiments using the pharmacological autophagy inducer rapamycin and the autophagy inhibitor chloroquine, autophagy inhibition was shown to reduce intracellular and extracellular DHAV-1 genome copies and viral titres. These results suggest that autophagy activated by DHAV-1 infection in DEFs affects DHAV-1 proliferation and extracellular release. Next, we screened the autophagy-inducing effects of the DHAV-1 structural proteins VP0, VP3, and VP1 and found that all DHAV-1 structural proteins could induce autophagy in DEFs but not the full autophagic flux. Finally, we found that VP1 promotes protein expression of PI3KC3 and Beclin1 by western blot experiments and that VP1 interacts with PI3KC3 by co-immunoprecipitation experiments; moreover, 3-MA-induced knockdown of PI3KC3 inhibited VP1 protein-induced autophagy in DEFs. In conclusion, the DHAV-1 structural protein VP1 regulates the PI3KC3 complex by interacting with PI3KC3 to induce autophagy in DEFs.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Autofagia , Proteína Beclina-1 , Patos , Vírus da Hepatite do Pato/fisiologia , Infecções por Picornaviridae/veterinária
20.
Prev Vet Med ; 208: 105730, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35964373

RESUMO

Duck hepatitis A virus (DHAV) is a single-strand positive-sense small RNA virus that causes high mortality in ducklings. In recent years, the incidence of DHAV-3 subtype has been increasing in China, leading to great economic losses to the duck-breeding industry. We investigated the incidence and mortality rates of DHAV in ducks and analysed the seroprevalence of DHAV in mainland China, by meta-analysis. Twenty-six studies published between 2009 and 2021 were retrieved, with a total of 689,549 cases from 14 provinces. Using the DerSimonian-Laird model, DHAV prevalence was estimated with the variance-stabilizing double arcsine transformation. The incidence of DHAV in mainland China was 12 % (95 % confidence interval [CI]: 3-20 %), and the mortality rate was 11 % (95 % CI: 2-19 %), suggesting that the virus was highly virulent and mortality was high. Time analysis showed that DHAV incidence decreased over time. The typing survey showed that strains of DHAV-1 serotype accounted for 38 % (95 % CI: 21-56 %) and strains of DHAV-3 serotype accounted for 49 % (95 % CI: 31-68%) of the tested samples. The decline in the detection rate of DHAV-1 may be due to the widespread use of the DHAV-1 vaccine, which has effectively controlled the DHAV-1 serotype virus. The DHAV-3 vaccine has been on the market for a short time and has no cross protection with DHAV-1, so DHAV-3 accounted for a high proportion of the tested samples. Subgroup analysis of the detection methods showed little difference between PCR and other detection methods.


Assuntos
Vírus da Hepatite do Pato , Hepatite Viral Animal , Infecções por Picornaviridae , Doenças das Aves Domésticas , Animais , Vírus da Hepatite do Pato/genética , Hepatite Viral Animal/epidemiologia , Hepatite Viral Animal/prevenção & controle , Prevalência , Estudos Soroepidemiológicos , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/veterinária , Doenças das Aves Domésticas/prevenção & controle , Patos , China/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...