Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.055
Filter
1.
Viruses ; 16(9)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39339856

ABSTRACT

Duck hepatitis virus 1 (DHV-1) is a major threat to the global poultry industry, causing significant economic losses due to high mortality rates in young ducklings. To better understand the evolution and host adaptation strategies of DHV-1, we conducted a comprehensive codon usage analysis of DHV-1 genomes. Our phylogenetic analysis revealed three well-supported DHV-1 phylogroups (Ia, Ib, and II) with distinct genetic diversity patterns. Comparative analyses of the codon usage bias and dinucleotide abundance uncovered a strong preference for A/U-ended codons and a biased pattern of dinucleotide usage in the DHV-1 genome, with CG dinucleotides being extremely underrepresented. Effective number of codons (ENC) analysis indicated a low codon usage bias in the DHV-1 ORF sequences, suggesting adaptation to host codon usage preferences. PR2 bias, ENC plot, and neutrality analyses revealed that both mutation pressure and natural selection influence the codon usage patterns of DHV-1. Notably, the three DHV-1 phylogroups exhibited distinct evolutionary trends, with phylogroups Ia and Ib showing evidence of neutral evolution accompanied by selective pressure, while the phylogroup II evolution was primarily driven by random genetic drift. Comparative analysis of the codon usage indices (CAI, RCDI, and SiD) among the phylogroups highlighted significant differences between subgroups Ia and Ib, suggesting distinct evolutionary pressures or adaptations influencing their codon usage. These findings contribute to our understanding of DHV-1 evolution and host adaptation, with potential implications for the development of effective control measures and vaccines.


Subject(s)
Codon Usage , Ducks , Evolution, Molecular , Genome, Viral , Hepatitis Virus, Duck , Host Adaptation , Phylogeny , Animals , Hepatitis Virus, Duck/genetics , Hepatitis Virus, Duck/classification , Ducks/virology , Host Adaptation/genetics , Selection, Genetic , Genetic Variation , Poultry Diseases/virology , Hepatitis, Viral, Animal/virology , Codon
2.
Emerg Microbes Infect ; 13(1): 2399970, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39221587

ABSTRACT

Multiple genotypes of highly pathogenic avian influenza (HPAI) H5 clade 2.3.4.4b viruses have caused epizootics in wild birds and poultry. The HPAI H5N1 genotype C virus caused a modest epizootic, whereas the occurrence of the HPAI H5N1 genotype AB virus in 2021 resulted in the largest avian influenza epizootic in Europe to date. Here we studied the pathogenicity of two HPAI H5N1 viruses by experimentally infecting chickens, Pekin ducks, Eurasian wigeons and Barnacle geese. Our study demonstrates that pathogenicity of the H5N1-2021-AB virus is lower in Pekin ducks, Eurasian wigeons and Barnacle geese compared to the H5N1-2020-C virus, whereas virus shedding was high for both viruses. After inoculation with H5N1-2021-C viral antigen expression was higher in the brain of Pekin ducks, Eurasian wigeons and Barnacle geese, which caused higher mortality compared to inoculation with H5N1-2021-AB virus. Subclinical infections occurred in Pekin ducks and Eurasian wigeons and mortality was reduced in Barnacle geese after inoculation with H5N1-2021-AB virus while H5N1-2020-C virus caused high morbidity and mortality in these species. This H5N1-2021-AB virus trait may have contributed to efficient spread of the virus in wild bird populations. Therefore, high mortality, virus shedding and long-lasting viral antigen expression found in Barnacle geese may have increased the risk for introduction into poultry.


Subject(s)
Chickens , Ducks , Influenza A Virus, H5N1 Subtype , Influenza in Birds , Virus Shedding , Animals , Influenza in Birds/virology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Ducks/virology , Chickens/virology , Geese/virology , Genotype , Poultry Diseases/virology
3.
Arch Virol ; 169(9): 191, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223363

ABSTRACT

A disease called "hydrosalpinx fluid and egg drop syndrome" (HFEDS) was observed in four flocks of Jinding ducks (Anas platyrhynchos domesticus) in Northeast China during the years 2022 to 2023. Here, we investigated the possible involvement of avian metapneumovirus (AMPV) infection. Full-length genome sequencing and sequence analysis of two AMPV strains showed that they belong to Eurasian lineage of AMPV subtype C. Based on surface glycoprotein (G) sequence comparisons, the Eurasian lineage can be divided into two sublineages (E1 and E2), and sublineage E2 is circulating in Jinding duck populations in Northeast China.


Subject(s)
Ducks , Genome, Viral , Metapneumovirus , Paramyxoviridae Infections , Phylogeny , Poultry Diseases , Animals , Ducks/virology , Metapneumovirus/genetics , Metapneumovirus/classification , Metapneumovirus/isolation & purification , Poultry Diseases/virology , Paramyxoviridae Infections/veterinary , Paramyxoviridae Infections/virology , China , Genome, Viral/genetics , Whole Genome Sequencing
4.
Front Cell Infect Microbiol ; 14: 1448480, 2024.
Article in English | MEDLINE | ID: mdl-39224701

ABSTRACT

Introduction: Muscovy duck parvovirus (MDPV), Goose parvovirus (GPV), Duck circovirus, (DuCV) and Duck adenovirus 3 (DAdV-3) are important pathogens that cause high morbidity and mortality in ducks, causing huge economic loss for the duck industry. Methods: The present study, a quadruplex one-step real time quantitative PCR method for the detection of MDPV, GPV, DuCV, and DAdV-3 was developed. Results: The results showed that assay had no cross-reactivity with other poultry pathogens [Duck plague virus (DPV), Duck tembusu virus (DTMUV), H6 avian influenza virus (H6 AIV), New duck reovirus (NDRV), Newcastle disease virus (NDV), H4 avian influenza virus (H4 AIV), Escherichia coli (E. coli), Muscovy duck reovirus (MDRV), Egg drop syndrome virus (EDSV), Pasteurella multocida (P. multocida)]. The sensitivity result showed that the limits of detection for MDPV, GPV, DuCV, and DAdV-3 were 10, 10, 1 and 10 copies/µl, respectively; The coefficients of variation intra- and inter-method was 1-2%; The range of linear (109 to 103 copies/µL) demonstrated the R2 values for MDPV, GPV, DuCV, and DAdV-3 as 0.9975, 0.998, 0.9964, and 0.996, respectively. The quadruplex real time quantitative PCR method efficiency was 90.30%, 101.10%, 90.72%, and 90.57% for MDPV, GPV, DuCV, and DAdV-3, respectively. 396 clinical specimens collected in some duck sausages from June 2022 to July 2023 were simultaneously detected using the established quadruplex real time quantitative PCR method and the reported assays. The detection rates for MDPV, GPV, DuCV, and DAdV-3 were 8.33% (33/396), 17.93% (71/396), 33.58% (133/396), and 29.04% (115/396), respectively. The agreement between these assays was greater than 99.56%. Discussion: The developed quadruplex real-time quantitative PCR assay can accurately detect these four viruses infecting ducks, providing a rapid, sensitive, specific and accurate technique for clinical testing.


Subject(s)
Ducks , Poultry Diseases , Real-Time Polymerase Chain Reaction , Animals , Ducks/virology , Real-Time Polymerase Chain Reaction/methods , Poultry Diseases/virology , Poultry Diseases/diagnosis , Sensitivity and Specificity , Parvovirinae/genetics , Parvovirinae/isolation & purification , Parvovirinae/classification , Aviadenovirus/genetics , Aviadenovirus/isolation & purification , Aviadenovirus/classification , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , Parvoviridae Infections/veterinary , Parvoviridae Infections/diagnosis , Parvoviridae Infections/virology
5.
Sci Rep ; 14(1): 22575, 2024 09 29.
Article in English | MEDLINE | ID: mdl-39343789

ABSTRACT

DHAV-1 is a highly infectious pathogen that can cause acute hepatitis in ducklings. MicroRNA (miRNA) plays an essential regulatory role in virus response. We characterized and compared miRNA and mRNA expression profiles in duck embryonic fibroblasts (DEF) and the liver of ducklings infected with DHAV-1. DHAV-1 infected DEF was divided into infection group (D group) and blank group (M group), and DHAV-1 infected duckling group was divided into infection group (H group) and blank group (N group). D vs. M have 130 differentially expressed (DE) miRNA (DEM) and 2204 differentially expressed (DE) mRNA (DEG), H vs. N have 72 DEM and 1976 DEG. By the intersection of D vs. M and H vs. N comparisons, 15 upregulated DEM, 5 downregulated DEM, 340 upregulated DEG and 50 downregulated DEG were found with both in vivo and in vitro DHAV-1 infection. In particular, we identified the same DE miRNA target genes and functional annotations of DE mRNA. We enriched with multiple gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, which may have important roles in viral virulence, host immunity, and metabolism. We selected miR-155, which is co-upregulated, and found that miR-155 targets SOCS1 to inhibit DHVA-1 replication.


Subject(s)
Ducks , Fibroblasts , MicroRNAs , Poultry Diseases , RNA, Messenger , Animals , Ducks/virology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Poultry Diseases/virology , Poultry Diseases/genetics , Poultry Diseases/metabolism , Gene Expression Profiling , Picornaviridae Infections/virology , Picornaviridae Infections/genetics , Picornaviridae Infections/veterinary , Picornaviridae Infections/metabolism , Mardivirus/genetics , Liver/metabolism , Liver/virology , Host-Pathogen Interactions/genetics , Gene Expression Regulation
6.
Proc Biol Sci ; 291(2031): 20241713, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39317317

ABSTRACT

High pathogenicity avian influenza virus (HPAIV) is a rapidly evolving virus causing significant economic and environmental harm. Wild birds are a key viral reservoir and an important source of viral incursions into animal populations, including poultry. However, we lack a thorough understanding of which species drive incursions and whether this changes over time. We explored associations between the abundances of 152 avian species and outbreaks of highly pathogenic avian influenza (HPAI) in poultry premises across Great Britain between October 2021 and January 2023. Spatial generalized additive models were used, with species abundance distributions sourced from eBird. Associations were investigated at the species-specific level and across species aggregations. During autumn/winter, associations were generally strongest with waterbirds such as ducks and geese; however, we also found significant associations in groups such as non-native gamebirds and rapid change in species-specific associations over time. Our results demonstrate the value of citizen science to rapidly explore wild species as potential facilitators of disease incursions into well-monitored populations, especially in regions where viral surveillance in wild species is limited. This can be a critical step towards prioritizing targeted surveillance that could inform species-specific biosecurity measures; particularly for HPAIV, which has undergone sudden shifts in host range and continues to rapidly evolve.


Subject(s)
Animals, Wild , Birds , Citizen Science , Disease Outbreaks , Influenza in Birds , Poultry , Animals , Influenza in Birds/epidemiology , Influenza in Birds/virology , Birds/virology , Poultry/virology , Disease Outbreaks/veterinary , United Kingdom/epidemiology , Poultry Diseases/epidemiology , Poultry Diseases/virology , Ducks/virology , Seasons
8.
Emerg Microbes Infect ; 13(1): 2398641, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39248597

ABSTRACT

The emergence of novel avian influenza reassortants in wild birds in recent years is a public health concern. However, the viruses that circulate in migratory birds are not fully understood. In this study, we summarized and categorized global H11 avian influenza viruses and reported that waterfowl and shorebirds are the major reservoirs of the identified H11 viruses. The surveillance data of the 35,749 faecal samples collected from wild bird habitats in eastern China over the past seven years revealed a low prevalence of H11 viruses in birds, with a positive rate of 0.067% (24 isolates). The phylogenetic analysis of the twenty viruses indicated that H11 viruses have undergone complex reassortment with viruses circulating in waterfowl and shorebirds. These tested viruses do not acquire mammalian adaptive mutations in their genomes and preferentially bind to avian-type receptors. Experimental infection studies demonstrated that the two tested H11N9 viruses of wild bird origin replicated and transmitted more efficiently in ducks than in chickens, whereas the pigeon H11N2 virus isolated from a live poultry market was more adapted to replicate in chickens than in ducks. In addition, some H11 isolates replicated efficiently in mice and caused body weight loss but were not lethal. Our study revealed the role of waterfowl and shorebirds in the ecology and evolution of H11 viruses and the potential risk of introducing circulating H11 viruses into ducks or chickens, further emphasizing the importance of avian influenza surveillance at the interface of migratory birds and poultry.


Subject(s)
Animal Migration , Animals, Wild , Birds , Columbidae , Influenza A virus , Influenza in Birds , Phylogeny , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Columbidae/virology , Influenza A virus/genetics , Influenza A virus/classification , Influenza A virus/isolation & purification , Influenza A virus/physiology , Birds/virology , China/epidemiology , Animals, Wild/virology , Mice , Reassortant Viruses/genetics , Reassortant Viruses/isolation & purification , Reassortant Viruses/classification , Ducks/virology , Evolution, Molecular , Feces/virology , Chickens/virology , Virus Replication
9.
Vet Microbiol ; 297: 110202, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094384

ABSTRACT

Herpesviruses-encoded microRNAs (miRNAs) have been discovered to be essential regulators in viral life cycle, participating in viral replication, latent or lytic infection, and immunological escape. However, the roles of miRNAs encoded by duck plague virus (DPV) are still unknown. Dev-miR-D28-3p is a miRNA uniquely encoded by DPV CHv strain. The aim of this study was to explore the effect of dev-miR-D28-3p on DPV replication and explore the potential mechanisms involved. Our findings demonstrated that transfection of dev-miR-D28-3p mimic into duck embryo fibroblasts (DEFs) effectively suppressed viral copies, viral titers and viral protein expressions during DPV infection, while the results above were reversed after transfection with dev-miR-D28-3p inhibitor. Subsequently, we further discovered that dev-miR-D28-3p specifically bound to DPV-encoded UL27 and inhibited its expression, suggesting that UL27 was the target gene of dev-miR-D28-3p. Finally, we investigated the role of UL27 in DPV replication and found the overexpression of UL27 increased viral copies, viral titers, and viral protein expressions; whereas the opposite results appear when knockdown of UL27. Our findings illustrated a novel mechanism that DPV regulated itself replication via dev-miR-D28-3p, paving the way for exploring the role of DPV-encoded miRNAs.


Subject(s)
Ducks , Fibroblasts , MicroRNAs , Virus Replication , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Ducks/virology , Fibroblasts/virology , Mardivirus/genetics , Mardivirus/physiology , Viral Proteins/genetics , Viral Proteins/metabolism , RNA, Viral/genetics , Poultry Diseases/virology , Gene Expression Regulation, Viral , Herpesviridae Infections/virology , Herpesviridae Infections/veterinary
10.
Poult Sci ; 103(10): 104076, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39096827

ABSTRACT

A suspected outbreak of duck astrovirus (DAstV) disease occurred in a large Muscovy duck farm in Guangdong Province, China, in June 2022, which severely affected the production performance and health of Muscovy ducks. This study aimed to investigate the prevalence of DAstV disease in Southeast China. Herein, we employed semi-nested PCR ethodto screen 5203 swab and liver samples from 11 Muscovy duck farms in 5 provinces of China for the presence of DAstV. Among them, 1356 samples (26.06%, 1356/5203) tested positive for DAstV, out of which 11 DAstV strains were isolated after 10 generations of blind transmission through Leghorn male hepatoma (LMH) cells and performed their whole-genome sequencing. The alignment results showed that the 11 DAstV isolates exhibited relatively low homology (15.4%-75%) with the astrovirus isolates from other species published in GenBank, whereas their homology (nucleotide: 90.4%-99.99%; amino acid: 94%-99.8%) with the DAstV type 1 (DAstV-1) reference strain was higher, indicating considerable homology. The results indicated that DAstV-1 was the main pathogenic factor. Herein, we successfully recreated the clinical symptoms of natural infection in 28-day-old specific-pathogen-free (SPF) ducks using the DAstV-1-GDB-2022 strain. The primary clinical manifestations included liver enlargement, hemorrhaging, and disruptions in liver function. Additionally, we confirmed the cross-species transmission potential of DAstV-1, marking the first occurrence of clinical symptoms of DAstV in 28-day-old SPF chickens. Our findings provide new perspectives on the epidemiology and pathogenicity of DAstV-1 and may help in advancing the development of DAstV vaccines.


Subject(s)
Astroviridae Infections , Avastrovirus , Chickens , Ducks , Hepatitis, Viral, Animal , Poultry Diseases , Animals , Ducks/virology , Poultry Diseases/virology , Poultry Diseases/epidemiology , China/epidemiology , Astroviridae Infections/veterinary , Astroviridae Infections/epidemiology , Astroviridae Infections/virology , Avastrovirus/pathogenicity , Avastrovirus/genetics , Avastrovirus/isolation & purification , Hepatitis, Viral, Animal/virology , Hepatitis, Viral, Animal/epidemiology , Molecular Epidemiology , Phylogeny , Virulence , Male , Prevalence
11.
Emerg Microbes Infect ; 13(1): 2399268, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39207215

ABSTRACT

High pathogenicity avian influenza (HPAI) virus H5N1 first emerged in Bangladesh in 2007. Despite the use of vaccines in chickens since 2012 to control HPAI, HPAI H5Nx viruses have continued to infect poultry, and wild birds, resulting in notable mass mortalities in house crows (Corvus splendens). The first HPAI H5Nx viruses in Bangladesh belonged to clade 2.2.2, followed by clade 2.3.4.2 and 2.3.2.1 viruses in 2011. After the implementation of chicken vaccination in 2012, these viruses were mostly replaced by clade 2.3.2.1a viruses and more recently clade 2.3.4.4b and h viruses. In this study, we reconstruct the phylogenetic history of HPAI H5Nx viruses in Bangladesh to evaluate the role of major host species in the maintenance and evolution of HPAI H5Nx virus in Bangladesh and reveal the role of heavily impacted crows in virus epidemiology. Epizootic waves caused by HPAI H5N1 and H5N6 viruses amongst house crows occurred annually in winter. Bayesian phylodynamic analysis of clade 2.3.2.1a revealed frequent bidirectional viral transitions between domestic ducks, chickens, and house crows that was markedly skewed towards ducks; domestic ducks might be the source, or reservoir, of HPAI H5Nx in Bangladesh, as the number of viral transitions from ducks to chickens and house crows was by far more numerous than the other transitions. Our results suggest viral circulation in domestic birds despite vaccination, with crow epizootics acting as a sentinel. The vaccination strategy needs to be updated to use more effective vaccinations, assess vaccine efficacy, and extension of vaccination to domestic ducks, the key reservoir.


Subject(s)
Chickens , Disease Reservoirs , Ducks , Influenza in Birds , Phylogeny , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , Influenza in Birds/prevention & control , Ducks/virology , Bangladesh/epidemiology , Disease Reservoirs/virology , Chickens/virology , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/isolation & purification , Crows/virology , Animals, Wild/virology , Influenza A virus/genetics , Influenza A virus/pathogenicity , Influenza A virus/classification , Influenza A virus/immunology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Poultry Diseases/prevention & control
12.
Curr Microbiol ; 81(9): 296, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105989

ABSTRACT

Duck enteritis virus (DEV) may lead to vascular injury, gastrointestinal mucosal erosion, lymphoid organ injury, and Polyinosinic-polycytidylic acid (Poly I:C) has an antiviral effect by inducing low levels of interferon. The purpose of this study was to explore the pathogenesis of DEV-induced intestinal injury in ducks and to verify the therapeutic effects of different concentrations of Poly I:C. In this study, duck enteritis model was established by infecting healthy Pekin ducks with DEV. Duck intestinal tissues were extracted from normal control group, model group, and treatment group with different doses of Poly I:C. In vivo, HE and TUNEL staining were used to observe the morphological changes and apoptosis. In vitro, the proliferation and apoptosis of duck intestinal epithelial cells were evaluated by MTT assay, TUNEL staining, and flow cytometry. The results showed that Poly I:C protected ducks from DEV toxicity by improving intestinal morphology and inhibiting apoptosis. In addition, the antiviral effect of Poly I:C on DEV was found in a dose-dependent manner, with a more relatively obvious effect at a high dose of Poly I:C. All in all, these results demonstrated that Poly I:C played a vital role in the apoptosis induced by DEV in ducks and modest dose of Poly I:C treatment worked well and may provide important reference for the development of new antiviral drugs in the future.


Subject(s)
Apoptosis , Ducks , Enteritis , Poly I-C , Animals , Ducks/virology , Poly I-C/pharmacology , Poly I-C/administration & dosage , Apoptosis/drug effects , Enteritis/virology , Enteritis/drug therapy , Enteritis/veterinary , Poultry Diseases/virology , Poultry Diseases/drug therapy , Intestines/virology , Intestines/pathology , Antiviral Agents/pharmacology , Mardivirus/drug effects , Intestinal Mucosa/virology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology
13.
Vet Res ; 55(1): 100, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39135123

ABSTRACT

High pathogenicity avian influenza viruses (HPAIVs) have caused major epizootics in recent years, with devastating consequences for poultry and wildlife worldwide. Domestic and wild ducks can be highly susceptible to HPAIVs, and infection leads to efficient viral replication and massive shedding (i.e., high titres for an extended time), contributing to widespread viral dissemination. Importantly, ducks are known to shed high amounts of virus in the earliest phase of infection, but the dynamics and impact of environmental contamination on the epidemiology of HPAIV outbreaks are poorly understood. In this study, we monitored mule ducks experimentally infected with two H5N8 clade 2.3.4.4b goose/Guangdong HPAIVs sampled in France in 2016-2017 and 2020-2021 epizootics. We investigated viral shedding dynamics in the oropharynx, cloaca, conjunctiva, and feathers; bird-to-bird viral transmission; and the role of the environment in viral spread and as a source of samples for early detection and surveillance. Our findings showed that viral shedding started before the onset of clinical signs, i.e., as early as 1 day post-inoculation (dpi) or post-contact exposure, peaked at 4 dpi, and lasted for up to 14 dpi. The detection of viral RNA in aerosols, dust, and water samples mirrored viral shedding dynamics, and viral isolation from these environmental samples was successful throughout the experiment. Our results confirm that mule ducks can shed high HPAIV titres through the four excretion routes tested (oropharyngeal, cloacal, conjunctival, and feather) while being asymptomatic and that environmental sampling could be a non-invasive tool for early viral RNA detection in HPAIV-infected farms.


Subject(s)
Ducks , Influenza A Virus, H5N8 Subtype , Influenza in Birds , Poultry Diseases , Virus Shedding , Animals , Ducks/virology , Influenza in Birds/virology , Influenza A Virus, H5N8 Subtype/physiology , Influenza A Virus, H5N8 Subtype/pathogenicity , Poultry Diseases/virology , France/epidemiology
14.
Genes (Basel) ; 15(8)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39202404

ABSTRACT

As a founding member of the Src family of kinases, Src has been confirmed to participate in the regulation of immune responses, integrin signaling, and motility. Ducks are usually asymptomatic carriers of RNA viruses such as Newcastle disease virus and avian influenza virus, which can be deadly to chickens. The beneficial role of Src in modulating the immune response remains largely unknown in ducks. Here, we characterized the duck Src and found that it contains a 192-base-pair 5' untranslated region, a 1602-base-pair coding region, and a 2541-base-pair 3' untranslated region, encoding 533 amino acid residues. Additionally, duSrc transcripts were significantly activated in duck tissues infected by Newcastle disease virus compared to controls. The duSrc transcripts were notably widespread in all tissues examined, and the expression level was higher in liver, blood, lung, pancreas, and thymus. Moreover, we found the expression levels of IFN-ß, NF-κB, IRF3, and Src were significantly increased in DEFs after infection with 5'ppp dsRNA, but there was no significant difference before and after treatment in DF1 cells. Furthermore, overexpression of duSrc followed by stimulation with 5'ppp dsRNA led to an elevation of IFN-ß levels. The SH3 and PTKc domains of duSrc contributed to promoting the activity of IFN-ß and NF-κB in DEFs stimulated by 5'ppp dsRNA.


Subject(s)
Cloning, Molecular , Ducks , Animals , Ducks/genetics , Ducks/immunology , Ducks/virology , src-Family Kinases/genetics , src-Family Kinases/metabolism , Newcastle disease virus/immunology , Newcastle disease virus/genetics , Avian Proteins/genetics , Avian Proteins/immunology , Avian Proteins/metabolism , Newcastle Disease/immunology , Newcastle Disease/virology , Newcastle Disease/genetics , Interferon-beta/genetics , Interferon-beta/immunology , Interferon-beta/metabolism , Tissue Distribution , Poultry Diseases/immunology , Poultry Diseases/virology , Poultry Diseases/genetics
15.
Microb Pathog ; 194: 106825, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39074517

ABSTRACT

Short beak and dwarfism syndrome (SBDS) is attributed to Novel Goose Parvovirus (NGPV), which has inflicted significant economic losses on farming in China. Despite its significant impact, limited research has been conducted on the pathogenesis of this disease. The SD strain, a parvovirus variant isolated from ducks in Shandong province, was identified and characterized in our study. Phylogenetic analysis and sequence comparisons confirmed the classification of the SD strain as a member of NGPV. Based on this information, we established an animal model of SBDS by inoculating Cherry Valley ducks with the SD strain. Our findings indicate that infection with the SD strain leads to a reduction in body weight, beak length, width, and tibia length. Notably, significant histopathological alterations were observed in the thymus, spleen, and intestine of the infected ducks. Furthermore, the SD strain induces bone disorders and inflammatory responses. To evaluate the impact of NGPV on intestinal homeostasis, we performed 16S rDNA sequencing and gas chromatography to analyze the composition of intestinal flora and levels of short-chain fatty acids (SCFAs) in the cecal contents. Our findings revealed that SD strain infection induces dysbiosis in cecal microbial and a decrease in SCFAs production. Subsequent analysis revealed a significant correlation between bacterial genera and the clinical symptoms in NGPV SD infected ducks. Our research providing novel insights into clinical pathology of NGPV in ducks and providing a foundation for the research of NGPV treatment targeting gut microbiota.


Subject(s)
Ducks , Parvoviridae Infections , Phylogeny , Poultry Diseases , Animals , Ducks/virology , Parvoviridae Infections/veterinary , Parvoviridae Infections/virology , Parvoviridae Infections/pathology , Poultry Diseases/virology , Poultry Diseases/pathology , China , Parvovirinae/genetics , Parvovirinae/isolation & purification , Parvovirinae/pathogenicity , Gastrointestinal Microbiome , Intestines/pathology , Intestines/virology , RNA, Ribosomal, 16S/genetics , Disease Models, Animal , Dysbiosis/virology , Dysbiosis/veterinary , Fatty Acids, Volatile/metabolism , Geese/virology , Spleen/pathology , Spleen/virology , Beak/virology , Beak/pathology
16.
Nat Microbiol ; 9(10): 2626-2641, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39009691

ABSTRACT

Influenza A viruses (IAV) pose substantial burden on human and animal health. Avian, swine and human IAV bind sialic acid on host glycans as receptor, whereas some bat IAV require MHC class II complexes for cell entry. It is unknown how this difference evolved and whether dual receptor specificity is possible. Here we show that human H2N2 IAV and related avian H2N2 possess dual receptor specificity in cell lines and primary human airway cultures. Using sialylation-deficient cells, we reveal that entry via MHC class II is independent of sialic acid. We find that MHC class II from humans, pigs, ducks, swans and chickens but not bats can mediate H2 IAV entry and that this is conserved in Eurasian avian H2. Our results demonstrate that IAV can possess dual receptor specificity for sialic acid and MHC class II, and suggest a role for MHC class II-dependent entry in zoonotic IAV infections.


Subject(s)
Histocompatibility Antigens Class II , Influenza A Virus, H2N2 Subtype , Influenza in Birds , Influenza, Human , N-Acetylneuraminic Acid , Receptors, Virus , Virus Internalization , Humans , Animals , N-Acetylneuraminic Acid/metabolism , Histocompatibility Antigens Class II/metabolism , Influenza, Human/virology , Influenza, Human/metabolism , Influenza, Human/immunology , Influenza A Virus, H2N2 Subtype/metabolism , Influenza A Virus, H2N2 Subtype/genetics , Influenza A Virus, H2N2 Subtype/immunology , Receptors, Virus/metabolism , Receptors, Virus/genetics , Influenza in Birds/virology , Influenza in Birds/metabolism , Influenza in Birds/immunology , Ducks/virology , Cell Line , Swine , Birds/virology , Chiroptera/virology , Chickens/virology
17.
Vet Microbiol ; 296: 110187, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39053390

ABSTRACT

Short-beak and dwarf syndrome (SBDS) is caused by novel goose parvovirus (NGPV) infection, which leads to farm economic losses. Our research aimed to investigate the potential of administering isolated lactic acid bacteria (LAB) in alleviating SBDS in ducks. Eight wild LAB strains were isolated from duck feces and their biosecurity was investigated in both duck embryo fibroblast (DEF) and live ducks. Moreover, the LAB strains exhibited no detrimental effects on bone metabolism levels and facilitated the tight junction proteins (TJPs) mRNA expression, and contributing to the mitigation of inflammation in healthy ducks. Subsequently, we conducted in vitrol and in vivo experiments to assess the impact of LAB on NGPV infection. The LAB strains significantly reduced the viral load of NGPV and downregulated the mRNA levels of pro-inflammatory factors in DEF. Additionally, LAB treatment alleviated SBDS in NGPV-infected ducks. Furthermore, LAB treatment alleviated intestinal damage, and reduced the inflammatory response, while also mitigating bone resorption in NGPV-infected ducks. In conclusion, the LAB strains isolated from duck feces have favorable biosecurity and alleviate SBDS in ducks, and the mechanism related to LAB improves intestinal barrier integrity, alleviates inflammation, and reduces bone resorption. Our study presents a novel concept for the prevention and treatment of NGPV, thereby establishing a theoretical foundation for the future development of probiotics in the prevention and treatment of NGPV.


Subject(s)
Ducks , Inflammation , Lactobacillales , Poultry Diseases , Animals , Ducks/virology , Ducks/microbiology , Poultry Diseases/microbiology , Poultry Diseases/prevention & control , Poultry Diseases/virology , Inflammation/veterinary , Inflammation/prevention & control , Lactobacillales/genetics , Parvoviridae Infections/veterinary , Parvoviridae Infections/prevention & control , Parvoviridae Infections/virology , Parvoviridae Infections/microbiology , Feces/microbiology , Feces/virology , Bone Resorption/prevention & control , Bone Resorption/microbiology , Bone Resorption/veterinary , Intestines/microbiology , Intestines/virology , Probiotics/administration & dosage , Probiotics/pharmacology , Probiotics/therapeutic use , Parvovirus/genetics , Geese/virology
18.
Methods Mol Biol ; 2837: 11-22, 2024.
Article in English | MEDLINE | ID: mdl-39044071

ABSTRACT

Duck hepatitis B virus (DHBV) is an avian member of the hepatotropic DNA viruses, or hepadnaviridae. It shares with the human hepatitis B virus (HBV) a similar genomic organization and replication strategy via reverse transcription, but is simpler than HBV in lacking the X gene and in expressing just two coterminal envelope proteins: Large (L) and small (S). DHBV has been extensively used as a convenient and valuable animal model for study of the hepadnaviral life cycle, and for drug screening in vitro but also in vivo. Ducks and primary duck hepatocytes (PDHs) are inexpensive, easily accessible, and readily infected with DHBV. The high levels of genome replication and protein expression in duck liver and PDHs also facilitate monitoring of viral life cycle using conventional molecular biology techniques such as Southern blot for replicative DNA and covalently closed circular DNA (cccDNA), Northern blot for viral RNAs, and Western blot for viral proteins.


Subject(s)
Ducks , Hepadnaviridae Infections , Hepatitis B Virus, Duck , Hepatocytes , Virus Replication , Animals , Ducks/virology , Hepatocytes/virology , Hepatocytes/metabolism , Hepatitis B Virus, Duck/genetics , Hepadnaviridae Infections/virology , Hepadnaviridae Infections/veterinary , Disease Models, Animal , Hepatitis, Viral, Animal/virology , DNA, Viral/genetics , Cells, Cultured , Primary Cell Culture/methods , Cell Culture Techniques/methods
19.
Cell Host Microbe ; 32(7): 1089-1102.e10, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38889725

ABSTRACT

Avian influenza A virus (IAV) surveillance in Northern California, USA, revealed unique IAV hemagglutinin (HA) genome sequences in cloacal swabs from lesser scaups. We found two closely related HA sequences in the same duck species in 2010 and 2013. Phylogenetic analyses suggest that both sequences belong to the recently discovered H19 subtype, which thus far has remained uncharacterized. We demonstrate that H19 does not bind the canonical IAV receptor sialic acid (Sia). Instead, H19 binds to the major histocompatibility complex class II (MHC class II), which facilitates viral entry. Unlike the broad MHC class II specificity of H17 and H18 from bat IAV, H19 exhibits a species-specific MHC class II usage that suggests a limited host range and zoonotic potential. Using cell lines overexpressing MHC class II, we rescued recombinant H19 IAV. We solved the H19 crystal structure and identified residues within the putative Sia receptor binding site (RBS) that impede Sia-dependent entry.


Subject(s)
Ducks , Hemagglutinin Glycoproteins, Influenza Virus , Histocompatibility Antigens Class II , Influenza A virus , Phylogeny , Receptors, Virus , Animals , Influenza A virus/genetics , Influenza A virus/immunology , Receptors, Virus/metabolism , Receptors, Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/genetics , Ducks/virology , Humans , Virus Internalization , Influenza in Birds/virology , Binding Sites , Protein Binding , Crystallography, X-Ray , Cell Line , N-Acetylneuraminic Acid/metabolism , Host Specificity , Species Specificity
20.
Microb Pathog ; 195: 106751, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38880314

ABSTRACT

Short-beak and dwarfism syndrome (SBDS) is a new disease caused by a genetic variant of goose parvovirus in ducks that results in enormous economic losses for the waterfowl industry. Currently, there is no commercial vaccine for this disease, so it is urgent to develop a safer and more effective vaccine to prevent this disease. In this study, we optimized the production conditions to enhance the expression of the recombinant VP2 protein and identified the optimal conditions for subsequent large-scale expression. Furthermore, the protein underwent purification via nickel column affinity chromatography, followed by concentration using ultrafiltration tube. Subsequently, it was observed by transmission electron microscopy (TEM) that the NGPV recombinant VP2 protein assembled into virus-like particles (VLPs) resembling those of the original virus. Finally, the ISA 78-VG adjuvant was mixed with the NGPV-VP2 VLPs to be prepared as a subunit vaccine. Furthermore, both agar gel precipitation test (AGP) and serum neutralization test demonstrated that NGPV VLP subunit vaccine could induce the increase of NGPV antibody in breeding ducks. The ducklings were also challenged with the NGPV, and the results showed that the maternal antibody level could provide sufficient protection to the ducklings. These results indicated that the use of the NGPV VLP subunit vaccine based on the baculovirus expression system could facilitate the large-scale development of a reliable vaccine in the future.


Subject(s)
Antibodies, Viral , Baculoviridae , Capsid Proteins , Ducks , Parvoviridae Infections , Parvovirinae , Poultry Diseases , Recombinant Proteins , Viral Vaccines , Animals , Baculoviridae/genetics , Antibodies, Viral/blood , Antibodies, Viral/immunology , Parvoviridae Infections/veterinary , Parvoviridae Infections/immunology , Parvoviridae Infections/prevention & control , Parvoviridae Infections/virology , Ducks/virology , Poultry Diseases/virology , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Viral Vaccines/immunology , Viral Vaccines/genetics , Capsid Proteins/genetics , Capsid Proteins/immunology , Parvovirinae/genetics , Parvovirinae/immunology , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Vaccines, Subunit/immunology , Vaccines, Subunit/genetics , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Adjuvants, Immunologic
SELECTION OF CITATIONS
SEARCH DETAIL