Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 260
Filtrar
1.
Sci Rep ; 13(1): 8410, 2023 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-37225865

RESUMO

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.


Assuntos
Anseriformes , Vírus da Influenza A , Influenza Aviária , Animais , Antivirais , Gansos/virologia , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Filogenia , República da Coreia/epidemiologia , Influenza Aviária/diagnóstico , Influenza Aviária/epidemiologia , Influenza Aviária/genética , Influenza Aviária/virologia , Fezes/virologia , Anseriformes/virologia , Monitoramento Biológico
2.
Viruses ; 14(8)2022 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-36016381

RESUMO

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.


Assuntos
Infecções por Astroviridae , Avastrovirus , Gansos , Gota , Animais , Infecções por Astroviridae/epidemiologia , Infecções por Astroviridae/veterinária , Avastrovirus/genética , Doenças das Aves , China/epidemiologia , Patos , Gansos/virologia , Gota/veterinária , Filogenia
3.
Arch Virol ; 167(6): 1455-1459, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35469095

RESUMO

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.


Assuntos
Virus da Influenza A Subtipo H5N1 , Influenza Aviária , Animais , Patos/virologia , Europa (Continente)/epidemiologia , Gansos/virologia , Genótipo , Virus da Influenza A Subtipo H5N1/genética , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Filogenia
4.
Viruses ; 14(2)2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35215873

RESUMO

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.


Assuntos
Patos/virologia , Gansos/virologia , Vírus da Influenza A Subtipo H5N8/patogenicidade , Influenza Aviária/virologia , Tropismo Viral , Animais , Animais Selvagens/virologia , Encéfalo/virologia , Adaptação ao Hospedeiro , Vírus da Influenza A Subtipo H5N8/isolamento & purificação , Vírus da Influenza A Subtipo H5N8/fisiologia , Mucosa Intestinal/virologia , RNA Viral/análise , Mucosa Respiratória/virologia , Ligação Viral
5.
PLoS One ; 17(2): e0264308, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35226672

RESUMO

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.


Assuntos
Infecções por Astroviridae/genética , Avastrovirus/genética , Galinhas/virologia , Patos/virologia , Gansos/virologia , Variação Genética , Genoma Viral , Doenças das Aves Domésticas , Animais , Filogenia , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/virologia
6.
Sci Rep ; 11(1): 23223, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34853356

RESUMO

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.


Assuntos
Vírus da Influenza A/isolamento & purificação , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Aves Domésticas/virologia , Animais , Galinhas/virologia , China/epidemiologia , Patos/virologia , Monitoramento Epidemiológico , Gansos/virologia , Vírus da Influenza A/genética
7.
Viruses ; 13(11)2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34834997

RESUMO

The outbreaks of H5N2 avian influenza viruses have occasionally caused the death of thousands of birds in poultry farms. Surveillance during the 2018 winter season in South Korea revealed three H5N2 isolates in feces samples collected from wild birds (KNU18-28: A/Wild duck/South Korea/KNU18-28/2018, KNU18-86: A/Bean Goose/South Korea/KNU18-86/2018, and KNU18-93: A/Wild duck/South Korea/KNU18-93/2018). Phylogenetic tree analysis revealed that these viruses arose from reassortment events among various virus subtypes circulating in South Korea and other countries in the East Asia-Australasian Flyway. The NS gene of the KNU18-28 and KNU18-86 isolates was closely related to that of China's H10N3 strain, whereas the KNU18-93 strain originated from the H12N2 strain in Japan, showing two different reassortment events and different from a low pathogenic H5N3 (KNU18-91) virus which was isolated at the same day and same place with KNU18-86 and KNU18-93. These H5N2 isolates were characterized as low pathogenic avian influenza viruses. However, many amino acid changes in eight gene segments were identified to enhance polymerase activity and increase adaptation and virulence in mice and mammals. Experiments reveal that viral replication in MDCK cells was quite high after 12 hpi, showing the ability to replicate in mouse lungs. The hematoxylin and eosin-stained (H&E) lung sections indicated different degrees of pathogenicity of the three H5N2 isolates in mice compared with that of the control H1N1 strain. The continuing circulation of these H5N2 viruses may represent a potential threat to mammals and humans. Our findings highlight the need for intensive surveillance of avian influenza virus circulation in South Korea to prevent the risks posed by these reassortment viruses to animal and public health.


Assuntos
Vírus da Influenza A Subtipo H5N2/classificação , Vírus da Influenza A Subtipo H5N2/genética , Vírus Reordenados/classificação , Vírus Reordenados/genética , Animais , Animais Selvagens/virologia , Aves/virologia , Modelos Animais de Doenças , Cães , Patos/virologia , Fezes/virologia , Gansos/virologia , Vírus da Influenza A Subtipo H5N2/isolamento & purificação , Vírus da Influenza A Subtipo H5N2/patogenicidade , Vírus da Influenza A/genética , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Japão , Células Madin Darby de Rim Canino , Mamíferos , Camundongos , Epidemiologia Molecular , Filogenia , Aves Domésticas/virologia , Vírus Reordenados/isolamento & purificação , Vírus Reordenados/patogenicidade , República da Coreia/epidemiologia , Virulência , Replicação Viral
8.
Virus Res ; 306: 198566, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34582833

RESUMO

Coronavirus disease 2019 (COVID-19), caused by Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was first reported in Wuhan, China, and rapidly spread throughout the world. This newly emerging pathogen is highly transmittable and can cause fatal disease. More than 35 million cases have been confirmed, with a fatality rate of about 2.9% to October 9, 2020. However, the original and intermediate hosts of SARS-CoV-2 remain unknown. Here, 3160 poultry samples collected from 14 provinces of China between September and December 2019 were tested for SARS-CoV-2 infection. All the samples were SARS-CoV-2 negative, but 593 avian coronaviruses were detected, including 485 avian infectious bronchitis viruses, 72 duck coronaviruses, and 36 pigeon coronaviruses, with positivity rates of 15.35%, 2.28%, and 1.14%, respectively. Our surveillance demonstrates the diversity of avian coronaviruses in China, with higher prevalence rates in some regions. Furthermore, the possibility that SARS-CoV-2 originated from a known avian-origin coronavirus can be preliminarily ruled out. More surveillance of and research into avian coronaviruses are required to better understand the diversity, distribution, cross-species transmission, and clinical significance of these viruses.


Assuntos
Doenças das Aves/virologia , Infecções por Coronavirus/veterinária , Coronavirus/genética , Coronavirus/isolamento & purificação , Variação Genética , Animais , Doenças das Aves/epidemiologia , Galinhas/virologia , China/epidemiologia , Columbidae/virologia , Coronavirus/classificação , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/virologia , Patos/virologia , Monitoramento Epidemiológico , Gansos/virologia , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/virologia , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação
9.
Microbiol Spectr ; 9(2): e0130921, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34585974

RESUMO

Highly pathogenic avian influenza (HPAI) H5 viruses have posed a substantial pandemic threat through repeated human infection since their emergence in China in 1996. Nationwide control measures, including vaccination of poultry, were implemented in 2005, leading to a sharp reduction in H5N1 virus outbreaks. In 2008, novel non-N1 subtype (H5Nx) viruses emerged, gradually replacing the dominant H5N1 subtype and causing global outbreaks. The cause of this major shift in the ecology of HPAI H5 viruses remains unknown. Here, we show that major H5N1 virus lineages underwent population bottlenecks in 2006, followed by a recovery in virus populations between 2007 and 2009. Our analyses indicate that control measures, not competition from H5Nx viruses, were responsible for the H5N1 decline, with an H5N1 lineage capable of infecting poultry and wild birds experiencing a less severe population bottleneck due to circulation in unaffected wild birds. We show that H5Nx viruses emerged during the successful suppression of H5N1 virus populations in poultry, providing an opportunity for antigenically distinct H5Nx viruses to propagate. Avian influenza vaccination programs would benefit from universal vaccines targeting a wider diversity of influenza viruses to prevent the emergence of novel subtypes. IMPORTANCE A major shift in the ecology of highly pathogenic avian influenza (HPAI) H5 viruses occurred from 2008 to 2014, when viruses with non-N1 neuraminidase genes (termed H5Nx viruses) emerged and caused global H5 virus outbreaks. Here, we demonstrate that nationwide control measures, including vaccination in China, successfully suppressed H5N1 populations in poultry, providing an opportunity for antigenically distinct H5Nx viruses to emerge. In particular, we show that the widespread use of H5N1 vaccines likely conferred a fitness advantage to H5Nx viruses due to the antigenic mismatch of the neuraminidase genes. These results indicate that avian influenza vaccination programs would benefit from universal vaccines that target a wider diversity of influenza viruses to prevent potential emergence of novel subtypes.


Assuntos
Vírus da Influenza A/isolamento & purificação , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Animais , Animais Selvagens/virologia , Aves/virologia , Galinhas/virologia , China , Patos/virologia , Gansos/virologia , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/fisiologia , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Vírus da Influenza A/fisiologia , Influenza Aviária/epidemiologia , Filogenia , Doenças das Aves Domésticas/epidemiologia
10.
Avian Pathol ; 50(5): 447-452, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34545745

RESUMO

Routine culturing of goose haemorrhagic polyomavirus (GHPV) is cumbersome, and limited data are available about its replication and gene expression profile. In this study, goose embryo fibroblast cells were infected with GHPV for temporal measurement of the viral genome copy number and mRNA levels with quantitative PCR. Accumulation of small and large tumour antigen-encoding mRNAs was detected as early as 9 hours post-infection (hpi), while high level expression of the capsid protein encoding VP1-VP3, and ORF-X mRNAs was first detected at 24 hpi. Elevation of GHPV genome copy number was noted at 48 hpi. The results indicate that the gene expression profile of GHPV is similar to that described for mammalian polyomaviruses.RESEARCH HIGHLIGHTS GHPV was propagated in culture of primary goose embryo fibroblast cells.The transcription commenced before the onset of viral DNA replication.The transcription patterns of GHPV and mammalian polyomaviruses were comparable.


Assuntos
Doenças das Aves/virologia , Gansos/virologia , Infecções por Polyomavirus/veterinária , Polyomavirus , Animais , Replicação do DNA , DNA Viral , Polyomavirus/genética , RNA Mensageiro/genética , Transcriptoma , Replicação Viral
11.
Viruses ; 13(6)2021 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-34207913

RESUMO

The emergence of a novel goose nephritic astrovirus (GNAstV) has caused economic losses to the Chinese goose industry. High viral load is found in the spleen of goslings infected with GNAstV, but pathological injuries to the spleen due to GNAstV are largely unknown. In this study, 50 two-day-old goslings were infected orally with GNAstV, and 50 goslings were treated with PBS as control. Spleens were collected at different times following infection to assess damage. GNAstV infection caused visceral gout and urate deposition in joints, and resulted in 16% mortality. GNAstV was found in the lymphocytes and macrophages within the spleen. Lymphocyte loss, especially around the white pulp, and destruction and decline in the number of reticular fibers was observed in GNAstV-infected goslings. Moreover, in GNAstV-infected goslings, ultrahistopathological examination found that splenic lymphocytes exhibited condensed chromatin and apoptotic bodies, and reticular cells displayed damage to plasma membrane integrity and swollen mitochondria. Furthermore, TUNEL staining confirmed apoptosis of lymphocytes, and the mRNA levels of Fas and FasL were significantly increased in the GNAstV-infected goslings. In addition, GNAstV infection reduced the number and protein expression of CD8. In conclusion, GNAstV infection causes lymphocyte depletion, reticular cell necrosis, reticular fiber destruction, lymphocyte apoptosis, and reduction in CD8 levels, which contribute to spleen injury.


Assuntos
Apoptose , Avastrovirus/fisiologia , Gansos/virologia , Linfócitos/metabolismo , Doenças das Aves Domésticas/etiologia , Doenças das Aves Domésticas/metabolismo , Animais , Avastrovirus/classificação , Avastrovirus/genética , Biópsia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos , Imuno-Histoquímica , Contagem de Linfócitos , Linfócitos/imunologia , Linfócitos/patologia , Doenças das Aves Domésticas/diagnóstico , Baço/imunologia , Baço/metabolismo , Baço/patologia , Baço/virologia , Carga Viral
12.
Arch Virol ; 166(9): 2495-2504, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34232400

RESUMO

Short beak and dwarfism syndrome (SBDS) emerged in Cherry Valley duck flocks in China in 2015, and novel goose parvovirus (NGPV) was shown to be the etiological agent of SBDS. To date, it is not known whether SBDS-related NGPV isolates possess common molecular characteristics. In this study, three new NGPV strains (namely, SDHT16, SDJN19, and SDLC19) were isolated from diseased ducks showing typical signs of SBDS and successfully passaged in embryonated goose or Cherry Valley duck eggs. The complete genome sequences of these NGPV strains were 98.9%-99.7% identical to each other but showed slightly less similarity (95.2%-96.1% identity) to classical GPV strains. A total of 16 common amino acid substitutions were present in the VP1 proteins of six NGPV strains (SDHT16, SDJN19, SDLC19, QH, JS1, and SDLC01) compared with the classical Chinese GPV strains, nine of which were identical to those found in European GPV strain B. The non-structural protein Rep1 of the six NGPV strains had 12 common amino acid substitutions compared with the classical GPV strains. Phylogenetic analysis indicated that the Chinese NGPV strains clustered with the European SBDS-related NGPV strains, forming a separate branch that was distinct from the group formed by the classical GPV strains. The present study shows the common molecular characteristics of NGPV isolates and suggests that the Chinese NGPV isolates probably share a common ancestor with European SBDS-related NGPV strains.


Assuntos
Nanismo/veterinária , Nanismo/virologia , Parvovirinae/classificação , Parvovirinae/genética , Filogenia , Doenças das Aves Domésticas/virologia , Animais , China , Patos/virologia , Gansos/virologia , Genoma Viral , Infecções por Parvoviridae/virologia , Parvovirus/genética , Alinhamento de Sequência , Sequenciamento Completo do Genoma
13.
Arch Virol ; 166(7): 1931-1942, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33934195

RESUMO

Since its first recognition in the early 1960s, Derzsy's disease has caused significant economic losses in the goose meat industry through the world. Today, Derzsy's disease still maintains its importance for small-scale waterfowl farming, despite not having a significant impact on public health. In the present study, we investigated the distribution of goose parvovirus (GPV) and its potential variants from a 2019 outbreak in Turkey. Tissue samples were obtained from infected eggs and goslings that were raised in distinct farming areas of the various provinces. For this purpose, a novel primer set for amplification of a 630-bp region of VP3 was designed to confirm GPV infection by conventional PCR method. A 4709-base nucleotide sequence including the structural, non-structural, and 5' inverted terminal repeat regions was obtained from three samples from the Central Anatolian region. Multiple sequence comparisons and phylogenetic analysis demonstrated that the field strains clustered with European group 2 and contained a series of unique amino acid substitutions that might affect the virulence of the virus. These results confirmed that European-related field strains caused the outbreak in Asia Minor, and this might assist in understanding the circulation of GPV in Asia and Europe.


Assuntos
Gansos/virologia , Parvovirinae/genética , Parvovirus/genética , Virulência/genética , Substituição de Aminoácidos/genética , Animais , Ásia , Surtos de Doenças , Europa (Continente) , Infecções por Parvoviridae/virologia , Filogenia , Doenças das Aves Domésticas/virologia
14.
BMC Vet Res ; 17(1): 124, 2021 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-33740981

RESUMO

BACKGROUND: Influenza viruses are a continuous threat to avian and mammalian species, causing epidemics and pandemics. After the circulation of H5N1 in 2006, 2015, and 2016 in Iraq, an H5N8 influenza virus emerged in domestic geese in Sulaymaniyah Province, Iraq. This study analyzed the genetic characteristics of the Iraqi H5N8 viruses. RESULTS: An HPAI virus subtype H5N8 was identified from domestic backyard geese in the Kurdistan Region, north Iraq. Phylogenic analyses of the hemagglutinin (HA) and neuraminidase (NA) genes indicated that Iraq H5N8 viruses belonged to clade 2.3.4.4 group B and clustered with isolates from Iran, Israel, and Belgium. Genetic analysis of the HA gene indicated molecular markers for avian-type receptors. Characterization of the NA gene showed that the virus had sensitive molecular markers for antiviral drugs. CONCLUSIONS: This is the first study ever on H5N8 in Iraq, and it is crucial to understand the epidemiology of the viruses in Iraq and the Middle East. The results suggest a possible role of migratory birds in the introduction of HPAI subtype H5N8 into Iraq.


Assuntos
Gansos/virologia , Vírus da Influenza A Subtipo H5N8/genética , Vírus da Influenza A Subtipo H5N8/isolamento & purificação , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Animais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H5N8/patogenicidade , Neuraminidase/genética , Filogenia , Proteínas Virais/genética
15.
Arch Virol ; 166(2): 559-569, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33409548

RESUMO

Marek's disease (MD) is an important disease of avian species and a potential threat to the poultry industry worldwide. In this study, 16 dead commercial chickens from flocks with suspected MD were necropsied immediately after death. Pathological findings were compatible with MD, and gallid alphaherpesvirus 2 was identified in PCR of spleen samples. Virus isolation was performed in primary cell culture, and partial sequencing of the meq gene of the isolate revealed >99% nucleotide sequence identity to virulent and very virulent plus strains from a number of European countries, placing it in the same subclade of clade III as two virulent Italian strains and a very virulent plus Polish strain as well as virulent strains of geese and ducks. The data reported here indicate that a virulent strain of Marek's disease virus is circulating in Turkey and has not been stopped by the current national vaccination programme.


Assuntos
Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/isolamento & purificação , Doença de Marek/virologia , Aves Domésticas/virologia , Animais , Sequência de Bases/genética , Células Cultivadas , Galinhas/virologia , Patos/virologia , Gansos/virologia , Itália , Filogenia , Polônia , Doenças das Aves Domésticas/virologia , Turquia , Virulência/genética
16.
Avian Pathol ; 50(1): 41-51, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33021105

RESUMO

Goose haemorrhagic polyomavirus (GHPV) is the aetiological agent of haemorrhagic nephritis enteritis of geese (HNEG), a fatal disease that impacts geese and has been recorded only in Europe. The present study describes the first clinical cases of HNEG in Taiwan and the phylogenesis of Taiwanese GHPV, and it elucidates the pathogenesis of GHPV infection using in situ hybridization (ISH). The genomes of Taiwanese GHPV were highly similar to the previously reported strains. The diseased geese showed various degrees of vascular damage, especially in the digestive tract. The affected geese in the early stage showed transmural haemorrhagic enteritis in the intestine. In the middle to late stages, the most obvious lesion was hypoxic necrosis of renal tubules around intralobular central veins. Mineralization deposited in the kidney and systemic gout were also found. ISH revealed GHPV DNA in the vascular endothelial cells throughout the body, but not in the parenchymal cells of organs. Accordingly, the pathogenesis of GHPV infection was consistent with viral tropism in the endothelial cells. Specific attack of vascular endothelium by GHPV resulted in endothelial cell necrosis and subsequent increases of blood vessel permeability, as well as secondary circulation disorders, such as oedema, haemorrhage, and ischaemic necrosis in the adjacent parenchyma. RESEARCH HIGHLIGHTS Cell tropism of GHPV is determined by in situ hybridization. The tropism results in vascular dysfunction and subsequent pathobiology. Haemorrhagic nephritis and enteritis of geese described outside Europe for the first time.


Assuntos
Gansos/virologia , Infecções por Polyomavirus/veterinária , Polyomavirus/fisiologia , Doenças das Aves Domésticas/virologia , Animais , Células Endoteliais/patologia , Células Endoteliais/virologia , Enterite/veterinária , Hemorragia/veterinária , Hibridização In Situ/veterinária , Intestinos/patologia , Intestinos/virologia , Rim/patologia , Rim/virologia , Nefrite/veterinária , Filogenia , Polyomavirus/genética , Infecções por Polyomavirus/epidemiologia , Infecções por Polyomavirus/patologia , Infecções por Polyomavirus/virologia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/patologia , Taiwan/epidemiologia , Tropismo Viral
17.
Viruses ; 12(11)2020 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-33171813

RESUMO

Aquatic bird bornavirus 1 (ABBV-1) is associated with chronic meningoencephalitis and ganglioneuritis. Although waterfowl species act as the natural host of ABBV-1, the virus has been sporadically isolated from other avian species, showing the potential for a broad host range. To evaluate the host restriction of ABBV-1, and its potential to infect commercial poultry species, we assessed the ability of ABBV-1 to replicate in cells and embryos of different avian species. ABBV-1 replication was measured using multi- and single-step growth curves in primary embryo fibroblasts of chicken, duck, and goose. Embryonated chicken and duck eggs were infected through either the yolk sac or chorioallantoic cavity, and virus replication was assessed by immunohistochemistry and RT-qPCR in embryonic tissues harvested at two time points after infection. Multi-step growth curves showed that ABBV-1 replicated and spread in goose and duck embryo fibroblasts, establishing a population of persistently infected cells, while it was unable to do so in chicken fibroblasts. Single-step growth curves showed that cells from all three species could be infected; however, persistence was only established in goose and duck fibroblasts. In ovo inoculation yielded no detectable viral replication or lesion in tissues. Data indicate that although chicken, duck, and goose embryo fibroblasts can be infected with ABBV-1, a persistent infection is more easily established in duck and goose cells. Therefore, ABBV-1 may be able to infect chickens in vivo, albeit inefficiently. Additionally, our data indicate that an in ovo model is inadequate to investigating ABBV-1 host restriction and pathogenesis.


Assuntos
Aves/virologia , Bornaviridae/fisiologia , Óvulo/virologia , Doenças das Aves Domésticas/virologia , Replicação Viral , Animais , Bornaviridae/patogenicidade , Células Cultivadas , Galinhas/virologia , Patos/virologia , Fibroblastos/virologia , Gansos/virologia , Cinética
18.
Trop Anim Health Prod ; 53(1): 36, 2020 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-33230580

RESUMO

Derzsy's disease, which is seen in goslings (Anser anser domestica) and Muscovy ducks (Cairina moschata), progresses to high mortality and causes significant yield losses. The disease agent is goose parvovirus (GPV), which is common in countries with waterfowl production. It has not previously been reported in Turkey. Using qPCR and sequencing of the VP3 protein-encoding gene, GPV is identified as the causative agent of high mortality among geese between 2018 and 2019. The VP3 sequences were also compared with the similar GenBank sequences phylogenetically. All the sequences were found to be most similar (98.90%) with Polish and Taiwan GPV strains. Phylogenetic analysis of the VP3 gene in strains in Turkey and comparison with strains from other countries demonstrated that the Turkish strains are native to the geography and circulated locally. This study detected the presence of the GPV gene for the first time in Turkey and demonstrated the importance of comparing the vaccine strain and wild type.


Assuntos
Patos/virologia , Gansos/virologia , Infecções por Parvoviridae/veterinária , Parvovirinae , Doenças das Aves Domésticas/virologia , Animais , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/virologia , Filogenia , Doenças das Aves Domésticas/epidemiologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Turquia/epidemiologia
19.
Parasit Vectors ; 13(1): 528, 2020 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-33092614

RESUMO

BACKGROUND: In this report we describe the molecular and pathological characteristics of West Nile virus (WNV) infection that occurred during the summer and fall of 2018 in avian species and equines. WNV is reported in Israel since the 1950s, with occasional outbreaks leading to significant morbidity and mortality in birds, high infection in horses and humans, and sporadic fatalities in humans. METHODS: Animal and avian carcasses in a suitable condition were examined by post-mortem analysis. Tissue samples were examined for WNV by RT-qPCR and the viral load was quantified. Samples with sufficient material quality were further analyzed by Endpoint PCR and sequencing, which was used for phylogenetic analysis. Tissue samples from positive animals were used for culturing the virus in Vero and C6/36 cells. RESULTS: WNV RNA was detected in one yellow-legged gull (Larus michahellis), two long-eared owls (Asio otus), two domesticated geese (Anser anser), one pheasant (Phasianus colchicus), four hooded crows (Corvus cornix), three horses and one donkey. Pathological and histopathological findings were characteristic of viral infection. Molecular analysis and viral load quantification showed varying degrees of infection, ranging between 70-1.4 × 106 target copies per sample. Phylogenetic analysis of a 906-bp genomic segment showed that all samples belonged to Lineage 1 clade 1a, with the following partition: five samples from 2018 and one sample detected in 2016 were of Cluster 2 Eastern European, two of Cluster 2 Mediterranean and four of Cluster 4. Four of the positive samples was successfully propagated in C6/36 and Vero cell lines for further work. CONCLUSIONS: WNV is constantly circulating in wild and domesticated birds and animals in Israel, necessitating constant surveillance in birds and equines. At least three WNV strains were circulating in the suspected birds and animals examined. Quantitative analysis showed that the viral load varies significantly between different organs and tissues of the infected animals.


Assuntos
Aves/virologia , Equidae/virologia , Febre do Nilo Ocidental , Vírus do Nilo Ocidental , Animais , Animais Selvagens/virologia , Autopsia , Charadriiformes/virologia , Corvos/virologia , Gansos/virologia , Genes Virais , Cavalos/virologia , Israel/epidemiologia , Gado/virologia , Filogenia , Carga Viral , Febre do Nilo Ocidental/patologia , Febre do Nilo Ocidental/transmissão , Febre do Nilo Ocidental/veterinária , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/isolamento & purificação
20.
J Virol ; 94(24)2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-32999029

RESUMO

The highly pathogenic avian influenza virus (HPAIV) H5N1 A/goose/Guangdong/1996 lineage (Gs/GD) is endemic in poultry across several countries in the world and has caused sporadic lethal infections in humans. Vaccines are important in HPAIV control both for poultry and in prepandemic preparedness for humans. This study assessed inactivated prepandemic vaccine strains in a One Health framework across human and agricultural and wildlife animal health, focusing on the genetic and antigenic diversity of field H5N1 Gs/GD viruses from the agricultural sector and assessing cross-protection in a chicken challenge model. Nearly half (47.92%) of the 48 combinations of vaccine and challenge viruses examined had bird protection of 80% or above. Most vaccinated groups had prolonged mean death times (MDT), and the virus-shedding titers were significantly lower than those of the sham-vaccinated group (P ≤ 0.05). The antibody titers in the prechallenge sera were not predictive of protection. Although vaccinated birds had higher titers of hemagglutination-inhibiting (HI) antibodies against the homologous vaccine antigen, most of them also had lower or no antibody titer against the challenge antigen. The comparison of all parameters and homologous or closely related vaccine and challenge viruses gave the best prediction of protection. Through additional analysis, we identified a pattern of epitope substitutions in the hemagglutinin (HA) of each challenge virus that impacted protection, regardless of the vaccine used. These changes were situated in the antigenic sites and/or reported epitopes associated with virus escape from antibody neutralization. As a result, this study highlights virus diversity, immune response complexity, and the importance of strain selection for vaccine development to control H5N1 HPAIV in the agricultural sector and for human prepandemic preparedness. We suggest that the engineering of specific antigenic sites can improve the immunogenicity of H5 vaccines.IMPORTANCE The sustained circulation of highly pathogenic avian influenza virus (HPAIV) H5N1 A/goose/Guangdong/1996 (Gs/GD) lineage in the agricultural sector and some wild birds has led to the evolution and selection of distinct viral lineages involved in escape from vaccine protection. Our results using inactivated vaccine candidates from the human pandemic preparedness program in a chicken challenge model identified critical antigenic conformational epitopes on H5 hemagglutinin (HA) from different clades that were associated with antibody recognition and escape. Even though other investigators have reported epitope mapping in the H5 HA, much of this information pertains to epitopes reactive to mouse antibodies. Our findings validate changes in antigenic epitopes of HA associated with virus escape from antibody neutralization in chickens, which has direct relevance to field protection and virus evolution. Therefore, knowledge of these immunodominant regions is essential to proactively develop diagnostic tests, improve surveillance platforms to monitor AIV outbreaks, and design more efficient and broad-spectrum agricultural and human prepandemic vaccines.


Assuntos
Proteção Cruzada/imunologia , Gansos/imunologia , Vacinas contra Influenza/imunologia , Influenza Aviária/imunologia , Influenza Aviária/prevenção & controle , Vacinas de Produtos Inativados/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Variação Antigênica , Galinhas/imunologia , Epitopos , Gansos/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/genética , Vacinação/veterinária , Eliminação de Partículas Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...