Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 274
Filter
1.
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
2.
Front Cell Infect Microbiol ; 14: 1424212, 2024.
Article in English | MEDLINE | ID: mdl-39165916

ABSTRACT

Introduction: Goose astrovirus (GAstV) is a newly emerging pathogen that is currently widespread among geese, causing visceral gout and leading to substantial gosling mortalities, posing a severe threat to the waterfowl industry. GAstV II is the predominant epidemic strain, characterized by its high morbidity and mortality rate. Consequently, there is an urgent necessity to develop an effective diagnostic approach to control the dissemination of GAstV II, particularly in clinical farms with limited laboratory resources. Methods: In this study, a novel multi-enzyme isothermal rapid amplification (MIRA) and lateral flow dipstick (LFD) combined assay was developed. Different primers designed specific targeting a highly conserved region within the viral RdRp gene for the detection of GAstV II. Primers optimized and MIRA-LFD assay analyzed its performance regarding limits of detection, specificity, and efficiency of detection. Results: The developed MIRA amplification is conducted at a constant temperature and accomplished within 10 minutes. Subsequent naked-eye observation of the LFD strips merely takes 5 minutes. The established MIRA-LFD method exhibits high specificity, with no cross-reaction with other pathogens and attains a detection sensitivity of 1 copy/µl, which is consistent with the reverse transcription quantitative PCR (RT-qPCR) assay. Further evaluation with clinical samples indicates that the accuracy of this MIRA-LFD method correlates well with RT-qPCR for the detection of GAstV II. Conclusion: In summary, the convenience, sensitivity, and rapidity of this newly developed detection method offer a significant advantage for on-site diagnosis of GAstV II.


Subject(s)
Astroviridae Infections , Geese , Molecular Diagnostic Techniques , Nucleic Acid Amplification Techniques , Poultry Diseases , Sensitivity and Specificity , Animals , Astroviridae Infections/diagnosis , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Geese/virology , Nucleic Acid Amplification Techniques/methods , Molecular Diagnostic Techniques/methods , Poultry Diseases/diagnosis , Poultry Diseases/virology , Avastrovirus/genetics , Avastrovirus/isolation & purification , DNA Primers/genetics , RNA, Viral/genetics
3.
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
4.
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
5.
Microb Pathog ; 194: 106829, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39084310

ABSTRACT

Goose astroviruses (GAstVs) are important pathogens which can cause gout in goslings leading to huge economic losses for the goose farming industry in China. In 2023, an infectious disease characterized by visceral gout broke out in commercial goose farms in Guangxi and Guangdong provinces of China. In this study, two GAstV strains of GXNN and GDCS were successfully isolated from these two disease-ridden goose farms. The complete genomic lengths of these two strains were 7166 bp, and phylogenetic analysis showed that they were both GAstV-2 subtypes. The 3-dimensional structures of the capsid protein were predicted and six characteristic mutation sites at amino acid positions 60, 61, 228, 229, 456 and 523 were found within the strong antigenic regions. A recombination event occurred at 6833-7070 nt between the GAstV TZ03 and Turkey astrovirus CA/00 and this was detected in both the GXNN and GDCS strains. Another recombinant event occurred at 63-2747 nt between the GAstV XT1 and GAstV SDPY and this was detected in the GDCS strain. When 1-day-old goslings were infected with the novel GXNN and GDCS strains, they showed severe visceral gout. This was accompanied by enlarged spleens, liver hemorrhages and urate deposits in the kidneys and ureters and their blood urea nitrogen levels were significantly elevated. The mortality rates of the GXNN- and GDCS-infected groups were pathogenically high at 80 % and 60 %, respectively. These results will promote our understanding of the evolution and epidemic potential of GAstVs in China.


Subject(s)
Astroviridae Infections , Capsid Proteins , Geese , Genome, Viral , Gout , Phylogeny , Poultry Diseases , Animals , Geese/virology , China , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Poultry Diseases/virology , Poultry Diseases/pathology , Gout/virology , Gout/veterinary , Gout/pathology , Capsid Proteins/genetics , Avastrovirus/genetics , Avastrovirus/pathogenicity , Avastrovirus/isolation & purification , Avastrovirus/classification , Virulence , Astroviridae/genetics , Astroviridae/isolation & purification , Astroviridae/pathogenicity
6.
Poult Sci ; 103(8): 103930, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908126

ABSTRACT

Interferon-induced protein with tetratricopeptide repeats (IFITs), a family of proteins strongly induced by type I interferon (IFN-I), are deeply involved in many cellular and viral processes. IFIT5, the sole protein in this family found in birds, also plays a crucial role in regulating virus infection. In this study, goose IFIT5 (gIFIT5) was first cloned from peripheral blood lymphocyte (PBL) and phylogenetic analysis showed that it was highly homologous with duck IFIT5 (dIFIT5), sharing 94.6% identity in amino acid sequence. Subsequently, the expression kinetics of gIFIT5 during goose astrovirus (GAstV) infection and the regulatory effect of gIFIT5 on GAstV proliferation were evaluated. Results showed that the mRNA and protein expression level of gIFIT5 was greatly induced by GAstV infection, especially at 12 hpi. Importantly, gIFIT5 could conversely promote GAstV replication in GEF cells. Virus titers in gIFIT5 overexpression group were significantly higher than those in control group at 12 and 24 hpi. Western blot and quantitative real-time PCR (qRT-PCR) further demonstrated that the production of viral cap protein was significantly facilitated in gIFIT5-transfected group. Collectively, GAstV facilitates self-replication via promoting gIFIT5 expression.


Subject(s)
Astroviridae Infections , Avian Proteins , Geese , Poultry Diseases , Virus Replication , Animals , Geese/physiology , Geese/virology , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Poultry Diseases/virology , Avian Proteins/genetics , Avian Proteins/metabolism , Phylogeny , Avastrovirus/physiology , Avastrovirus/genetics , Amino Acid Sequence , Gene Expression Regulation
7.
Poult Sci ; 103(8): 103936, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38909502

ABSTRACT

Goose circovirus (GoCV) is a common pathogen that causes immunosuppression and promotes secondary infections with other infectious agents in geese worldwide. In the present study, we identified GoCV in 2 out of 93 duck flocks from China and successfully sequenced the complete genomes of 2 strains (AH22du and HN20du). The whole genome of the two strains shared a high identity of 90.5 to 98.63% with China GoCV reference, and low identity of 58.98% with DuCV reference, respectively. Phylogenetic tree constructed on the two and other genome sequences of GoCV revealed three main branches. Both strains sequenced in this study were distributed on different sub-branches with most other Chinese GoCV strains, and AH22du clustered into an independent sub-branch within the cluster. Recombination analysis predicted that HN20du might potentially recombine from the major parent of yk4 (Zhejiang Province, China, 2007) and minor parent of GD/YJ/g2 (Guangdong Province, China, 2020). To the best of our knowledge, this is the first report of GoCV in ducks from China. This broadened host spectrum of GoCVs requires attention from the waterfowl industry and researchers.


Subject(s)
Circoviridae Infections , Circovirus , Ducks , Phylogeny , Poultry Diseases , Animals , Ducks/virology , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , China , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , Poultry Diseases/virology , Poultry Diseases/epidemiology , Genome, Viral , Geese/virology
8.
Res Vet Sci ; 173: 105279, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704977

ABSTRACT

Emerging pathogens can threaten human and animal health, necessitating reliable surveillance schemes to enable preparedness. We evaluated the repeatability and reproducibility of a method developed previously during a single year at one study site. Hunter-harvested ducks and geese were sampled for avian influenza virus at three discrete locations in the UK. H5N1 highly pathogenic avian influenza (HPAIV) was detected in four species (mallard [Anas platyrhynchos], Eurasian teal [Anas crecca], Eurasian wigeon [Mareca penelope] and pink-footed goose [Anser brachyrhynchus]) across all three locations and two non-HPAIV H5N1, influenza A positive detections were made from a mallard and Eurasian wigeon at two locations. Virus was detected within 1-to-4 days of sampling at every location. Application of rapid diagnostic methods to samples collected from hunter-harvested waterfowl offers potential as an early warning system for the surveillance and monitoring of emerging and existing strains of avian influenza A viruses in key avian species.


Subject(s)
Ducks , Geese , Influenza in Birds , Animals , Influenza in Birds/virology , Influenza in Birds/epidemiology , United Kingdom/epidemiology , Ducks/virology , Reproducibility of Results , Geese/virology , Influenza A Virus, H5N1 Subtype/isolation & purification
9.
Viruses ; 16(5)2024 05 11.
Article in English | MEDLINE | ID: mdl-38793646

ABSTRACT

(1) Goose astrovirus (GAstV) is a novel emerging pathogen that causes significant economic losses in waterfowl farming. A convenient, sensitive, and specific detection method for GAstV in field samples is important in order to effectively control GAstV. Droplet digital polymerase chain reaction (ddPCR) is a novel, sensitive, good-precision, and absolute quantitation PCR technology which does not require calibration curves. (2) In this study, we developed a ddPCR system for the sensitive and accurate quantification of GAstV using the conserved region of the ORF2 gene. (3) The detection limit of ddPCR was 10 copies/µL, ~28 times greater sensitivity than quantitative real-time PCR (qPCR). The specificity of the test was determined by the failure of amplification of other avian viruses. Both ddPCR and qPCR tests showed good repeatability and linearity, and the established ddPCR method had high sensitivity and good specificity to GAstV. Clinical sample test results showed that the positive rate of ddPCR (88.89%) was higher than that of qPCR (58.33%). (4) As a result, our results suggest that the newly developed ddPCR method might offer improved analytical sensitivity and specificity in its GAstV measurements. The ddPCR could be widely applied in clinical tests for GAstV infections.


Subject(s)
Astroviridae Infections , Avastrovirus , Geese , Sensitivity and Specificity , Animals , Astroviridae Infections/veterinary , Astroviridae Infections/diagnosis , Astroviridae Infections/virology , Geese/virology , Avastrovirus/genetics , Avastrovirus/isolation & purification , Poultry Diseases/virology , Poultry Diseases/diagnosis , Real-Time Polymerase Chain Reaction/methods , Polymerase Chain Reaction/methods , Reproducibility of Results , Astroviridae/genetics , Astroviridae/isolation & purification , Limit of Detection
10.
Poult Sci ; 103(7): 103775, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38713985

ABSTRACT

Goose circovirus (GoCV), a potential immunosuppressive virus possessing a circular single-stranded DNA genome, is widely distributed in both domesticated and wild geese. This virus infection causes significant economic losses in the waterfowl industry. The codon usage patterns of viruses reflect the evolutionary history and genetic architecture, allowing them to adapt quickly to changes in the external environment, particularly to their hosts. In this study, we retrieved the coding sequences (Rep and Cap) and the genome of GoCV from GenBank, conducting comprehensive research to explore the codon usage patterns in 144 GoCV strains. The overall codon usage of the GoCV strains was relatively similar and exhibited a slight bias. The effective number of codons (ENC) indicated a low overall extent of codon usage bias (CUB) in GoCV. Combined with the base composition and relative synonymous codon usage (RSCU) analysis, the results revealed a bias toward A- and G-ending codons in the overall codon usage. Analysis of the ENC-GC3s plot and neutrality plot suggested that natural selection plays an important role in shaping the codon usage pattern of GoCV, with mutation pressure having a minor influence. Furthermore, the correlations between ENC and relative indices, as well as correspondence analysis (COA), showed that hydrophobicity and geographical distribution also contribute to codon usage variation in GoCV, suggesting the possible involvement of natural selection. In conclusion, GoCV exhibits comparatively slight CUB, with natural selection being the major factor shaping the codon usage pattern of GoCV. Our research contributes to a deeper understanding of GoCV evolution and its host adaptation, providing valuable insights for future basic studies and vaccine design related to GoCV.


Subject(s)
Circovirus , Codon Usage , Geese , Circovirus/genetics , Animals , Geese/virology , Poultry Diseases/virology , Poultry Diseases/genetics , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Selection, Genetic , Host Adaptation/genetics , Adaptation, Physiological/genetics
11.
Viruses ; 16(4)2024 03 30.
Article in English | MEDLINE | ID: mdl-38675884

ABSTRACT

Goose astrovirus genotype 1 (GAstV-1) has emerged in goose farms in some provinces of China in recent years and is considered to be one of the pathogens of gout in goslings in China. However, few studies have been conducted on the dynamic distribution, tissue tropism, and pathogenesis of GAstV-1 in goslings. In 2022, an epidemiological investigation of goose astrovirus (GAstV) in goslings was conducted in seven provinces of China. During the investigation, a GAstV-1 designated as GAstV-JSXZ was identified in the kidney of an 8-day-old gosling and was successfully isolated from a goose embryo. The full genome sequence of GAstV-JSXZ was determined using the next-generation sequencing technique. The complete genome of GAstV-JSXZ was 7299-nt-long. Interestingly, the phylogenetic analysis revealed that Chinese GAstV-1 has formed two distinct subgroups based on the ORF 2 genomes, designated GAstV-1 1a and GAstV-1 1b. The GAstV-JSXZ shared the highest identity with GAstV-1 1a strain FLX and TZ03 in nucleotides (ORF1a: 98.3-98.4%; ORF1b: 92.3-99.1%; ORF2: 95.8-98.8%) and amino acid sequences (ORF1a: 99.4-99.5%; ORF1b: 98.2-98.8%; ORF2: 97.0-99.4%). To evaluate the pathogenicity of GAstV-1, 1-day-old goslings were inoculated with the virus by oral and subcutaneous injection routes, respectively. The results revealed that the virus causes extensive pathological organ damage, especially in the kidney, liver, and thymus. Virus-specific genomic RNA could be detected in the cloacal swabs and tissues of infected goslings throughout the experiment. The viral copy numbers examined in the kidney and intestine were the highest, followed by the liver and spleen. These results are likely to provide a new understanding of the pathogenicity of GAstV-1 in geese.


Subject(s)
Astroviridae Infections , Geese , Genome, Viral , Genotype , Phylogeny , Poultry Diseases , Animals , Geese/virology , China , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Poultry Diseases/virology , Astroviridae/genetics , Astroviridae/isolation & purification , Astroviridae/classification , Astroviridae/pathogenicity , Avastrovirus/genetics , Avastrovirus/isolation & purification , Avastrovirus/classification , Avastrovirus/pathogenicity , Virulence , High-Throughput Nucleotide Sequencing
12.
Poult Sci ; 103(7): 103774, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38669820

ABSTRACT

Goose astrovirus genotype 2 (GAstV-2) mainly causes gout in goslings; therefore, it is a major pathogen threatening to goose flocks. However, the mechanisms underlying host-GAstV-2 interactions remain unclear because host cells suitable for GAstV-2 replication have been unavailable. We previously noted that GAstV-2 is primarily located in goose renal epithelial cells, where it causes kidney damage. Therefore, here, we derived goose primary renal tubular epithelial (RTE) cells (GRTE cells) from the kidneys of goose embryos after collagenase I digestion. After culture in Dulbecco's modified Eagle medium/Nutrient mixture F-12 with 10% fetal bovine serum (FBS), the isolated cells had polygonal with roadstone-like morphology; they were identified to be epithelial cells based on the presence of cytokeratin 18 expression detected through immunofluorescence assay (IFA). GAstV-2 infection in GRTE cells led to no obvious cytopathic effects; the maximum amounts of infectious virions were observed 48 h post infection through IFA and quantitative PCR. Next, RNA-seq was performed to identify and map post-GAstV-2 infection differentially expressed genes. The downregulated pathways were mainly related to metabolism, including tryptophan metabolism, drug metabolism by cytochrome P450, xenobiotic metabolism by cytochrome P450, retinol metabolism, butanoate metabolism, starch and sucrose metabolism, ascorbate and aldarate metabolism, and drug metabolism by other enzymes and peroxisome. In contrast, the upregulated pathways were mostly related to the host cell defense and proliferation, including extracellular matrix-receptor interaction, complement and coagulation cascades, phagosome, PI3K-Akt signaling pathway, human T-lymphotropic virus 1 infection, lysosome, and tumor necrosis factor signaling pathway. In conclusion, we developed a GRTE cell line for GAstV-2 replication and analyzed the potential host-GAstV-2 interactions through RNA-seq; our results may aid in further investigating the pathogenic mechanisms underlying GAstV-2 infection and provide strategies for its prevention and control.


Subject(s)
Astroviridae Infections , Epithelial Cells , Geese , Genotype , Poultry Diseases , Animals , Geese/virology , Epithelial Cells/virology , Poultry Diseases/virology , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Sequence Analysis, RNA/veterinary , Kidney Tubules/virology , Kidney Tubules/cytology , Avastrovirus/physiology , Avastrovirus/genetics , Cells, Cultured
13.
Poult Sci ; 103(5): 103585, 2024 May.
Article in English | MEDLINE | ID: mdl-38492247

ABSTRACT

Goose astrovirus (GAstV) is a newly identified viral pathogen threatening waterfowl, exhibiting a high prevalence across various regions in China. Notably, the Guanghan District of Deyang City, situated in Sichuan Province, has faced a outbreak of GAstV, resulting in significant mortality among goslings due to the induction of gout-like symptoms. In our research, we successfully isolated a GAstV strain known as GAstV SCG3. This strain exhibits efficient replication capabilities, proving virulent in goslings and goose embryos. Our study delved into the characteristics of GAstV SCG3 both in vitro and in vivo. Additionally, we examined tissue phagocytosis and the distribution of GAstV SCG3 in deceased goslings using H&E staining and IHC techniques. According to the classification established by the ICTV, GAstV SCG3 falls under the category of GAstV genotype-2. Notably, it demonstrates the highest homology with the published AHAU5 sequences, reaching an impressive 98%. Furthermore, our findings revealed that GAstV SCG3 exhibits efficient proliferation exclusively in goose embryos and in LMH cells, while not manifesting in seven other types of avian and mammalian cells. Significantly, the mortality of GAstV on goslings and goose embryos are 93.1 and 80%, respectively. Moreover, the viral load in the livers of infected goslings surpasses that in the kidneys when compared with the attenuated strain GAstV SCG2. The mortality of GAstV is usually between 20% and 50%, our study marks the first report of a virulent GAstV strain with such a high mortality.


Subject(s)
Astroviridae Infections , Avastrovirus , Geese , Genotype , Poultry Diseases , Animals , Geese/virology , Poultry Diseases/virology , Poultry Diseases/mortality , Astroviridae Infections/veterinary , Astroviridae Infections/virology , Virulence , Avastrovirus/genetics , Avastrovirus/physiology , Avastrovirus/pathogenicity , China , Phylogeny
14.
Poult Sci ; 103(5): 103611, 2024 May.
Article in English | MEDLINE | ID: mdl-38471226

ABSTRACT

The aim of this study was to develop an efficient and accurate platform for the detection of the newly identified goose megrivirus (GoMV). To achieve this goal, we developed a TaqMan real-time PCR technology for the rapid detection and identification of GoMV. Our data showed that the established TaqMan real-time PCR assay had high sensitivity, with the lowest detection limit of 67.3 copies/µL. No positive signal can be observed from other goose origin viruses (including AIV, GPV, GoCV, GHPyV, and GoAstV), with strong specificity. The coefficients of variation of repeated intragroup and intergroup tests were all less than 1.5%, with excellent repeatability. Clinical sample investigation data from domestic Minbei White geese firstly provided evidence that GoMV can be transmitted both horizontally and vertically. In conclusion, since the TaqMan real-time PCR method has high sensitivity, specificity, and reproducibility, it can be a useful candidate tool for GoMV epidemiological investigation.


Subject(s)
Geese , Poultry Diseases , Real-Time Polymerase Chain Reaction , Animals , Real-Time Polymerase Chain Reaction/veterinary , Real-Time Polymerase Chain Reaction/methods , Geese/virology , Poultry Diseases/virology , Poultry Diseases/diagnosis , Sensitivity and Specificity , RNA Virus Infections/veterinary , RNA Virus Infections/virology , RNA Virus Infections/diagnosis , Reproducibility of Results
15.
Sci Rep ; 13(1): 8410, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37225865

ABSTRACT

Surveillance of influenza A viruses (IAVs) among migratory waterfowl is a first step in understanding the ecology, biology, and pathogenicity of IAVs. As part of the nationwide surveillance effort for IAVs in fowl in South Korea, we collected environmental fecal samples in different migratory bird stopover sites in South Korea during the winter seasons within November 2014 through January 2018. We collected a total of 6758 fecal samples, 75 of which were positive for IAV (1.11% positivity). Prevalence of IAVs varied per site and per year. Based on sequencing, the most prevalent hemagglutinin (HA) subtypes were H1, H6, and H5, and the most prevalent neuraminidase (NA) subtypes were N1, N3, and N2. Phylogenetic analyses showed that the genes we isolated clustered with reported isolates collected from other locations along the East Asian-Australasian Flyway. All the H5 and H7 isolates collected in this study were of low pathogenicity. None of the N1 and N2 genes carried amino acid markers of resistance against NA inhibitors. The winter 2016-2017 subset were primarily borne by migratory geese (Anser spp.). These results suggest that majority of the IAVs circulating among migratory wild fowl in South Korea in 2014-2018 were of low pathogenicity.


Subject(s)
Anseriformes , Influenza A virus , Influenza in Birds , Animals , Antiviral Agents , Geese/virology , Influenza A virus/genetics , Influenza A virus/pathogenicity , Phylogeny , Republic of Korea/epidemiology , Influenza in Birds/diagnosis , Influenza in Birds/epidemiology , Influenza in Birds/genetics , Influenza in Birds/virology , Feces/virology , Anseriformes/virology , Biological Monitoring
16.
Viruses ; 14(8)2022 08 12.
Article in English | MEDLINE | ID: mdl-36016381

ABSTRACT

Goose astroviruses (GoAstVs) are small non-enveloped viruses with a genome consisting of a single-stranded positive-sense RNA molecule. A novel GoAstV was identified in Shandong in 2016 and quickly spread to other provinces in China, causing gout in goslings, with a mortality rate of approximately 50%. GoAstV can also cause gout in chickens and ducks, indicating its ability to cross the species barrier. GoAstV has only been reported in China, where it has caused serious losses to the goose-breeding industry. However, in view of its cross-species transmission ability and pathogenicity in chickens and ducks, GoAstV should be a concern to poultry breeding globally. As an emerging virus, there are few research reports concerning GoAstV. This review summarizes the current state of knowledge about GoAstV, including the epidemiology, evolution analysis, detection methods, pathogenicity, pathogenesis, and potential for cross-species transmission. We also discuss future outlooks and provide recommendations. This review can serve as a valuable reference for further research on GoAstV.


Subject(s)
Astroviridae Infections , Avastrovirus , Geese , Gout , Animals , Astroviridae Infections/epidemiology , Astroviridae Infections/veterinary , Avastrovirus/genetics , Bird Diseases , China/epidemiology , Ducks , Geese/virology , Gout/veterinary , Phylogeny
17.
Arch Virol ; 167(6): 1455-1459, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35469095

ABSTRACT

Despite their widespread distribution, only a single genotype variant of clade 2.3.4.4b H5N1 influenza viruses has been found so far in Europe. Here, we report the detection of a new highly pathogenic avian influenza H5N1 genotype in geese and ducks from a backyard farm in the Czech Republic. Phylogenetic analysis revealed that the Czech H5N1 virus retained the A/Eurasian_Wigeon/Netherlands/1/2020-like backbone with an altered PB2 segment obtained from co-circulating low-pathogenic avian influenza viruses.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza in Birds , Animals , Ducks/virology , Europe/epidemiology , Geese/virology , Genotype , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/epidemiology , Influenza in Birds/virology , Phylogeny
18.
Viruses ; 14(2)2022 01 28.
Article in English | MEDLINE | ID: mdl-35215873

ABSTRACT

Highly pathogenic avian influenza (HPAI) outbreaks have become increasingly frequent in wild bird populations and have caused mass mortality in many wild bird species. The 2020/2021 epizootic was the largest and most deadly ever reported in Europe, and many new bird species tested positive for HPAI virus for the first time. This study investigated the tropism of HPAI virus in wild birds. We tested the pattern of virus attachment of 2020 H5N8 virus to intestinal and respiratory tissues of key bird species; and characterized pathology of naturally infected Eurasian wigeons (Mareca penelope) and barnacle geese (Branta leucopsis). This study determined that 2020 H5N8 virus had a high level of attachment to the intestinal epithelium (enterotropism) of dabbling ducks and geese and retained attachment to airway epithelium (respirotropism). Natural HPAI 2020 H5 virus infection in Eurasian wigeons and barnacle geese also showed a high level of neurotropism, as both species presented with brain lesions that co-localized with virus antigen expression. We concluded that the combination of respirotropism, neurotropism, and possibly enterotropism, contributed to the successful adaptation of 2020/2021 HPAI H5 viruses to wild waterbird populations.


Subject(s)
Ducks/virology , Geese/virology , Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza in Birds/virology , Viral Tropism , Animals , Animals, Wild/virology , Brain/virology , Host Adaptation , Influenza A Virus, H5N8 Subtype/isolation & purification , Influenza A Virus, H5N8 Subtype/physiology , Intestinal Mucosa/virology , RNA, Viral/analysis , Respiratory Mucosa/virology , Virus Attachment
19.
PLoS One ; 17(2): e0264308, 2022.
Article in English | MEDLINE | ID: mdl-35226672

ABSTRACT

Avian astroviruses (AAstVs) have caused major problem for poultry breeding industries in China in recent years, and the goose gout caused by goose astrovirus has produced particularly great economic losses. To better understand the prevalence and genetic diversity of AAstVs in China, 1210 poultry samples collected from eight provinces were tested with reverse transcription-polymerase chain reaction (RT-PCR) to detect AAstV infections in different poultry populations. Then, Open reading frames 2 (ORF2) was amplified by specific primers, and the genetic evolution was analyzed. Our surveillance data demonstrate the diversity of AAstVs in China insofar as we detected 17 AAstVs, including seven chicken astroviruses (CAstVs), five avian nephritis viruses (ANVs), two goose astroviruses (GoAstVs), two duck astrovirus (DAstVs), and one new AAstV belonging to Avastrovirus Group 3. The positive rate of AAstV infection was 1.40%. Host analysis showed that CAstVs and ANVs were isolated from chickens, DAstVs and GoAstVs were isolated from ducks. Host-species-specific AAstVs infections were also identified in numerous samples collected at each stage of production. This study provides further evidence to better understand the epidemiology of AAstVs in different species of poultry in China.


Subject(s)
Astroviridae Infections/genetics , Avastrovirus/genetics , Chickens/virology , Ducks/virology , Geese/virology , Genetic Variation , Genome, Viral , Poultry Diseases , Animals , Phylogeny , Poultry Diseases/genetics , Poultry Diseases/virology
20.
Sci Rep ; 11(1): 23223, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853356

ABSTRACT

Low pathogenic avian influenza viruses (LPAIVs) have been widespread in poultry and wild birds throughout the world for many decades. LPAIV infections are usually asymptomatic or cause subclinical symptoms. However, the genetic reassortment of LPAIVs may generate novel viruses with increased virulence and cross-species transmission, posing potential risks to public health. To evaluate the epidemic potential and infection landscape of LPAIVs in Guangxi Province, China, we collected and analyzed throat and cloacal swab samples from chickens, ducks and geese from the live poultry markets on a regular basis from 2016 to 2019. Among the 7,567 samples, 974 (12.87%) were LPAIVs-positive, with 890 single and 84 mixed infections. Higher yearly isolation rates were observed in 2017 and 2018. Additionally, geese had the highest isolation rate, followed by ducks and chickens. Seasonally, spring had the highest isolation rate. Subtype H3, H4, H6 and H9 viruses were detected over prolonged periods, while H1 and H11 viruses were detected transiently. The predominant subtypes in chickens, ducks and geese were H9, H3, and H6, respectively. The 84 mixed infection samples contained 22 combinations. Most mixed infections involved two subtypes, with H3 + H4 as the most common combination. Our study provides important epidemiological data regarding the isolation rates, distributions of prevalent subtypes and mixed infections of LPAIVs. These results will improve our knowledge and ability to control epidemics, guide disease management strategies and provide early awareness of newly emerged AIV reassortants with pandemic potential.


Subject(s)
Influenza A virus/isolation & purification , Influenza in Birds/epidemiology , Influenza in Birds/virology , Poultry/virology , Animals , Chickens/virology , China/epidemiology , Ducks/virology , Epidemiological Monitoring , Geese/virology , Influenza A virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL