Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Virol ; 96(18): e0123822, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36037479

RESUMO

The H7N9 subtype influenza A viruses pose a serious threat to public health, and there is still a lack of vaccines or drugs for humans against H7N9 influenza viruses. In this study, we screened two monoclonal antibodies (MAbs), 4H1E8 and 7H9A6, that specifically recognize the hemagglutinin (HA) protein of H7N9 influenza virus and display highly neutralizing activity against H7N9 virus. The epitopes recognized by two MAbs are nearly all conserved within all known H7 subtypes. Characteristic identification showed that two MAbs have high avidity for the HA protein but no hemagglutinin inhibition activity or antibody-dependent cellular cytotoxicity. Mechanistically, the 4H1E8 and 7H9A6 antibodies inhibit the pH-dependent conformational change of HA and block the HA-mediated membrane fusion. More importantly, 4H1E8 and 7H9A6 exhibit promising prophylactic and therapeutic effects against lethal challenge with H7N9 virus. Moreover, 4H1E8- and 7H9A6-treated mice displayed inhibition of pulmonary viral replication and reduced lung lesions after viral challenge. Together, these findings indicate that antibodies 4H1E8 and 7H9A6 recognize unique epitopes in the HA protein and possess the neutralizing activity and protective efficacy against the H7N9 influenza A viruses. IMPORTANCE In 2013, H7N9 influenza viruses appeared in China and other countries resulting in more than 1,500 individual infections or death. There are still limited studies on vaccines or drugs for humans against H7N9 influenza viruses. Alternative approaches against H7N9 virus infection need to be developed. Here, we identified two monoclonal antibodies (4H1E8 and 7H9A6) that possess neutralizing activity by blocking the pH-dependent HA-mediated membrane fusion. Additionally, the two monoclonal antibodies protect mice against the H7N9 virus challenge prophylactically or therapeutically. Therefore, our study demonstrates that 4H1E8 and 7H9A6 could be used for the prevention and treatment of the H7N9 influenza virus, and the conserved epitopes we identified may contribute to the development of a broad H7N9 vaccine and provide insights into unique antiviral approaches.


Assuntos
Subtipo H7N9 do Vírus da Influenza A , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antivirais , Antivirais/farmacologia , Antivirais/uso terapêutico , Epitopos/metabolismo , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Influenza Humana/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/tratamento farmacológico , Replicação Viral/efeitos dos fármacos
2.
Vet Res ; 53(1): 108, 2022 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-36517883

RESUMO

To improve understanding of the pathobiology of highly pathogenic avian influenza virus (HPAIV) infections in wild birds, pathogenicity and transmissibility of HPAIV H5N8 subtype clade 2.3.4.4b was evaluated in ~ 8-week-old herring gulls (Larus argentatus) divided into 3 groups: naïve birds (group A), birds previously exposed to low pathogenic avian influenza virus (LPAIV) H5N1 (group B) and LPAIV H13N6 (group C). The HPAIV H5N8 virus was highly virulent for naïve gulls, that showed early morbidity, high mortality, a broad spectrum of clinical signs, including violent neurological disorders, systemic distribution of the virus in organs accompanied by high level of shedding and transmission to contact birds. Pre-exposure to homologous and heterologous LPAIV subtypes conferred only partial protection: we observed increased survival rate (statistically significant only in group B), nervous signs, pantropic distribution of virus in organs, shedding (significantly reduced in gulls of group C in the early phase of disease and asymptomatic shedding in the late phase), transmission to contact gulls (more pronounced in group B) and near-complete seroconversion in survivors. Histopathological and immunohistochemical results indicate virus tropism for the neural, respiratory and myocardial tissues. In conclusion, we demonstrate that HPAIV H5N8 clade 2.3.4.4b is highly virulent and lethal for fully susceptible herring gulls and that pre-exposure to homo- and heterosubtypic LPAIV only partially modulates the disease outcome.


Assuntos
Charadriiformes , Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A Subtipo H5N8 , Vírus da Influenza A , Influenza Aviária , Animais , Virulência , Eliminação de Partículas Virais
3.
Emerg Infect Dis ; 26(7): 1557-1561, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32568059

RESUMO

We report detection of a highly pathogenic avian influenza A(H5N8) clade 2.3.4.4b virus in Europe. This virus was generated by reassortment between H5N8 subtype virus from sub-Saharan Africa and low pathogenicity avian influenza viruses from Eurasia.


Assuntos
Vírus da Influenza A Subtipo H5N8 , Influenza Aviária , África Subsaariana/epidemiologia , Animais , Europa (Continente) , Vírus da Influenza A Subtipo H5N8/genética , Influenza Aviária/epidemiologia , Filogenia , Vírus Reordenados/genética
4.
Virol J ; 17(1): 66, 2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32381003

RESUMO

BACKGROUND: Avian influenza virus infections cause significant economic losses on poultry farms and pose the threat of a possible pandemic outbreak. Routine vaccination of poultry against avian influenza is not recommended in Europe, however it has been ordered in some other countries, and more countries are considering use of the avian influenza vaccine as a component of their control strategy. Although a variety of such vaccines have been tested, most research has concentrated on specific antibodies and challenge experiments. METHODS: We monitored the transcriptomic response to a DNA vaccine encoding hemagglutinin from the highly pathogenic H5N1 avian influenza virus in the spleens of broiler and layer chickens. Moreover, in layer chickens the response to one and two doses of the vaccine was compared. RESULTS: All groups of birds immunized with two doses of the vaccine responded at the humoral level by producing specific anti-hemagglutinin antibodies. A response to the vaccine was also detected in the spleen transcriptomes. Differential expression of many genes encoding noncoding RNA and proteins functionally connected to the neuroendocrine-immune system was observed in different immunized groups. CONCLUSION: Broiler chickens showed a higher number and wider range of fold-changes in the transcriptional response than laying hens.


Assuntos
Anticorpos Antivirais/sangue , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/imunologia , Influenza Aviária/prevenção & controle , Vacinas de DNA/imunologia , Animais , Galinhas/genética , Galinhas/imunologia , Relação Dose-Resposta Imunológica , Perfilação da Expressão Gênica , Vacinas contra Influenza/administração & dosagem , Influenza Aviária/imunologia , Baço/imunologia , Vacinas de DNA/administração & dosagem
5.
Vet Res ; 51(1): 108, 2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-32859269

RESUMO

Defective interfering particles (DIPs) of influenza virus are generated through incorporation of highly truncated forms of genome segments, mostly those coding polymerase complex proteins (PB2, PB1, PA). Such particles are able to replicate only in the presence of a virus with the complete genome, thus DIPs may alter the infection outcome by suppressing production of standard virus particles, but also by stimulating the immune response. In the present study we compared the clinical outcome, mortality and transmission in chickens and turkeys infected with the same infectious doses of H7N7 low pathogenic avian influenza virus containing different levels of defective gene segments (95/95(DVG-high) and 95/95(DVG-low)). No clinical signs, mortality or transmission were noted in SPF chickens inoculated with neither virus stock. Turkeys infected with 95/95(DVG-high) showed only slight clinical signs with no mortality, and the virus was transmitted only to birds in direct contact. In contrast, more severe disease, mortality and transmission to direct and indirect contact birds was observed in turkeys infected with 95/95(DVG-low). Apathy, lower water and food intake, respiratory system disorders and a total mortality of 60% were noted. Shedding patterns in contact turkeys indicated more efficient within- and between-host spread of the virus than in 95/95(DVG-high) group. Sequencing of virus genomes showed no mutations that could account for the observed differences in pathogenicity. The results suggest that the abundance of DIPs in the inoculum was the factor responsible for the mild course of infection and disrupted virus transmission.


Assuntos
Genoma Viral , Vírus da Influenza A Subtipo H7N7/genética , Vírus da Influenza A Subtipo H7N7/patogenicidade , Influenza Aviária/transmissão , Doenças das Aves Domésticas/transmissão , Perus , Animais , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Virulência
6.
Emerg Infect Dis ; 24(12): 2270-2283, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30457528

RESUMO

We analyzed the highly pathogenic avian influenza (HPAI) H5 epizootic of 2016-17 in Europe by epidemiologic and genetic characteristics and compared it with 2 previous epizootics caused by the same H5 Guangdong lineage. The 2016-17 epizootic was the largest in Europe by number of countries and farms affected and greatest diversity of wild birds infected. We observed significant differences among the 3 epizootics regarding region affected, epidemic curve, seasonality, and outbreak duration, making it difficult to predict future HPAI epizootics. However, we know that in 2005-06 and 2016-17 the initial peak of wild bird detections preceded the peak of poultry outbreaks within Europe. Phylogenetic analysis of 2016-17 viruses indicates 2 main pathways into Europe. Our findings highlight the need for global surveillance of viral changes to inform disease preparedness, detection, and control.


Assuntos
Vírus da Influenza A/classificação , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Animais , Animais Selvagens , Aves , Surtos de Doenças , Europa (Continente)/epidemiologia , Genoma Viral , Geografia Médica , História do Século XXI , Vírus da Influenza A/patogenicidade , Influenza Aviária/história , Influenza Aviária/transmissão , Morbidade , Mortalidade , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/virologia , Análise Espaço-Temporal , Zoonoses
7.
Vet Res ; 49(1): 89, 2018 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-30208951

RESUMO

Infectious bursal disease virus (IBDV) is one of the most important immunosuppressive viral agents in poultry production. Prophylactic vaccinations of chicken flocks are the primary tool for disease control. Widely used immunoprophylaxis can, however, provide high pressure which contributes to the genetic diversification of circulating viruses, e.g. through reassortment of genome segments. We report the genetic and phenotypic characterization of a field reassortant IBDV (designated as Bpop/03) that acquired segment A from very virulent IBDV and segment B from classical attenuated D78-like IBDV. Despite the mosaic genetic make-up, the virus caused high mortality (80%) in experimentally infected SPF chickens and induced lesions typical of the acute form of IBD. The in vivo study results are in contrast with the foregoing experimental investigations in which the natural reassortants exhibited an intermediate pathotype, and underline the complex nature of IBDV virulence.


Assuntos
Infecções por Birnaviridae/veterinária , Galinhas , Genoma Viral , Vírus da Doença Infecciosa da Bursa/fisiologia , Vírus da Doença Infecciosa da Bursa/patogenicidade , Doenças das Aves Domésticas/virologia , Vírus Reordenados/fisiologia , Vírus Reordenados/patogenicidade , Sequência de Aminoácidos , Animais , Infecções por Birnaviridae/virologia , Vírus da Doença Infecciosa da Bursa/genética , Filogenia , Polônia , Vírus Reordenados/genética , Virulência
8.
Virus Genes ; 54(1): 67-76, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29052126

RESUMO

The study presents molecular characterization of H9N2 avian influenza (AI) isolates from field outbreaks in turkeys that occurred in Poland in 2013-2014. Sequences of all gene segments of one isolate from 2013 (A/turkey/Poland/14/2013(H9N2)) and two isolates from 2014 (A/turkey/Poland/08/2014(H9N2), A/turkey/Poland/09/2014(H9N2)) were obtained and analyzed in search of the phylogenetic relationship and molecular markers of zoonotic potential or increased pathogenicity. All gene segments were shown to originate from the wild bird reservoir and the close relationship of the analyzed isolates proved the link between the outbreaks in 2013 and 2014. However, remarkable molecular differences between isolates from 2013 to 2014 were identified, including mutation in the HA cleavage site (CS) leading to conversion from the PAASNR*GLF to the PAASKR*GLF motif and truncation of the PB1-F2 protein. Additionally, T97I substitution in the PA protein in A/turkey/Poland/08/2014 was detected which can be responsible for enhanced activity of viral polymerase in mammalian cells. However, experimental infection of mice with both isolates from 2014 showed their low pathogenicity, and no statistically significant differences in virus replication were observed between the viruses. Nevertheless, these findings indicate the dynamic evolution of H9N2 in the field emphasizing the need for monitoring of the situation in terms of H9N2 AI in Europe.


Assuntos
Surtos de Doenças , Genótipo , Vírus da Influenza A Subtipo H9N2/classificação , Vírus da Influenza A Subtipo H9N2/genética , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Animais , Modelos Animais de Doenças , Evolução Molecular , Vírus da Influenza A Subtipo H9N2/isolamento & purificação , Influenza Aviária/patologia , Camundongos , Epidemiologia Molecular , Filogenia , Polônia/epidemiologia , Análise de Sequência de DNA , Perus
9.
Emerg Infect Dis ; 22(7): 1201-7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27314611

RESUMO

In Poland, African swine fever (ASF) emerged in February 2014; by August 2015, the virus had been detected in >130 wild boar and in pigs in 3 backyard holdings. We evaluated ASF spread in Poland during these 18 months. Phylogenetic analysis indicated repeated incursions of genetically distinct ASF viruses of genotype II; the number of cases positively correlated wild boar density; and disease spread was very slow. More cases were reported during summer than autumn. The 18-month prevalence of ASF in areas under various animal movement restrictions was 18.6% among wild boar found dead or killed by vehicles and only 0.2% in hunted wild boar. Repeated introductions of the virus into the country, the primary role of wild boar in virus maintenance, and the slow spread of the disease indicate a need for enhanced biosecurity at pig holdings and continuous and intensive surveillance for fast detection of ASF.


Assuntos
Vírus da Febre Suína Africana/genética , Febre Suína Africana/epidemiologia , Epidemias/veterinária , Febre Suína Africana/transmissão , Febre Suína Africana/virologia , Animais , DNA Viral/genética , Filogenia , Polônia/epidemiologia , Densidade Demográfica , Prevalência , Estações do Ano , Sus scrofa , Suínos
10.
Avian Pathol ; 43(5): 427-36, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25132323

RESUMO

In April/May 2013, four outbreaks of avian influenza virus (AIV) infections caused by H9N2 subtype were diagnosed in Poland in fattening turkey flocks exhibiting a drop in feed and water intake, depression, respiratory signs and mortality. The subsequent serological survey carried out on samples collected between June 2012 and September 2013 from 92 poultry flocks detected positive sera in two additional meat turkey flocks located in the same province. The analysis of amino acids in the haemagglutinin and neuraminidase glycoproteins revealed that the detected H9N2 viruses possessed molecular profiles suggestive of low pathogenicity, avian-like SAα2,3 receptor specificity and adaptation to domestic poultry. Phylogenetic studies showed that these H9N2 AIVs grouped within the Eurasian clade of wild bird-origin AIVs and had no relationship with H9N2 AIV circulating in poultry in the Middle East and Far East Asia over the past decade. Experimentally infected SPF chickens with the index-case H9N2 virus remained healthy throughout the experiment. On the other hand, ten 3-week-old commercial turkeys infected via the oculonasal route showed respiratory signs and mortality (2/10 birds). Additional diagnostic tests demonstrated the consistent presence of DNA/RNA of Ornithobacterium rhinotracheale, Bordetella avium and, less frequently, of astro-, rota-, reo-, parvo- and adenoviruses in turkeys both from field outbreaks and laboratory experiment. Although no microbiological culture was performed, we speculate that these secondary pathogens could play a role in the pathogenicity of the current H9N2 infections.


Assuntos
Galinhas , Surtos de Doenças/veterinária , Vírus da Influenza A Subtipo H9N2/isolamento & purificação , Influenza Aviária/virologia , Perus , Animais , Influenza Aviária/epidemiologia , Filogenia , Polônia/epidemiologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testes Sorológicos
11.
Avian Dis ; 58(3): 488-90, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25518447

RESUMO

Major outbreaks of avian type C botulism have been rarely reported from Central Europe. In this paper, we report the first severe outbreak of avian type C botulism in Poland. In 2011-12, two epizootics caused by Clostridium botulinum took place at Jeziorsko dam reservoir and affected an estimated number of 5500 birds in 2011 and 1600 birds in 2012. In total, 24 species ofwaterbirds were affected, including mainly waterfowl (37.0%), shorebirds (27.0%), rallids (25.7%), and larids (9.1%). Mallards (Anas platyrhynchos) and coots (Fulica atra) were most commonly represented among all affected species (27.5% and 25.0% of all recorded carcasses, respectively). Laboratory analyses confirmed the presence of type C botulinum toxin in the internal organs of paralyzed birds. This case study from the Jeziorsko dam reservoir demonstrates that this type of shallow-water habitat is especially prone to avian botulism outbreaks in the climatic conditions of Central Europe.


Assuntos
Doenças das Aves/microbiologia , Botulismo/veterinária , Clostridium botulinum/isolamento & purificação , Animais , Animais Selvagens/microbiologia , Aves , Toxinas Botulínicas/metabolismo , Botulismo/microbiologia , Clostridium botulinum/genética , Clostridium botulinum/metabolismo , Polônia
12.
Avian Dis ; 58(4): 523-30, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25618996

RESUMO

The virulence of pigeon paramyxovirus type 1 (PPMV-1) for different species of birds was investigated in two independent sets of experiments in which groups of pigeons, chickens, turkeys, quails, and geese (10 birds per group) were inoculated with 10(6) median embryo infectious doses of PPMV-1 isolate: 1) nonpassaged (nPPMV-1, intracerebral pathogenicity [ICPI] value = 1.27) and 2) after six passages in specific-pathogen-free chickens (pPPMV-1, ICPI = 1.46) via the oculonasal route. Naive birds were placed in contact with infected birds (two birds per group) to monitor virus transmission. Clinical observation was performed daily. Additionally, cloacal swabs, oropharyngeal swabs, and selected organ samples were collected on days 2, 4, 7, 10, and 14 postinfection and tested by real-time reverse transcriptase-PCR for estimation of viral shedding and distribution in tissues. Infected pigeons exhibited nervous and digestive tract symptoms, mortality, shedding, and transmission to contact birds. Chickens, turkeys, quails, and geese did not exhibit any clinical signs regardless of the PPMV-1 strain used for inoculation. However, in contrast to quails and geese, chickens and turkeys shed the virus via the oral cavity and cloaca, and transmission to contact birds was also observed. Viral RNA was identified in tissues collected from all pPPMV-1-infected birds, whereas negative results were obtained in the case of tissues taken from nPPMV-1-infected quails and geese. We conclude that the PPMV-1 used in this study was most virulent to pigeons, followed by chickens and turkeys, while quails and geese seem to have the highest level of innate resistance to this strain. However, passaging of PPMV-1 in chickens resulted in the increase of ICPI and noticeable but sometimes contrasting changes in the replication capacities of the virus.


Assuntos
Doença de Newcastle/virologia , Vírus da Doença de Newcastle/fisiologia , Eliminação de Partículas Virais , Animais , Aves , Encéfalo/virologia , Cloaca/virologia , Duodeno/virologia , Rim/virologia , Fígado/virologia , Pulmão/virologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Especificidade da Espécie , Baço/virologia , Distribuição Tecidual
13.
Virus Evol ; 10(1): veae027, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38699215

RESUMO

Since 2016, A(H5Nx) high pathogenic avian influenza (HPAI) virus of clade 2.3.4.4b has become one of the most serious global threats not only to wild and domestic birds, but also to public health. In recent years, important changes in the ecology, epidemiology, and evolution of this virus have been reported, with an unprecedented global diffusion and variety of affected birds and mammalian species. After the two consecutive and devastating epidemic waves in Europe in 2020-2021 and 2021-2022, with the second one recognized as one of the largest epidemics recorded so far, this clade has begun to circulate endemically in European wild bird populations. This study used the complete genomes of 1,956 European HPAI A(H5Nx) viruses to investigate the virus evolution during this varying epidemiological outline. We investigated the spatiotemporal patterns of A(H5Nx) virus diffusion to/from and within Europe during the 2020-2021 and 2021-2022 epidemic waves, providing evidence of ongoing changes in transmission dynamics and disease epidemiology. We demonstrated the high genetic diversity of the circulating viruses, which have undergone frequent reassortment events, providing for the first time a complete overview and a proposed nomenclature of the multiple genotypes circulating in Europe in 2020-2022. We described the emergence of a new genotype with gull adapted genes, which offered the virus the opportunity to occupy new ecological niches, driving the disease endemicity in the European wild bird population. The high propensity of the virus for reassortment, its jumps to a progressively wider number of host species, including mammals, and the rapid acquisition of adaptive mutations make the trend of virus evolution and spread difficult to predict in this unfailing evolving scenario.

14.
Acta Vet Hung ; 61(3): 416-24, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23921353

RESUMO

In order to study the variation of low pathogenic avian influenza viruses (AIV) of H3 subtype in the natural reservoir, partial genetic characterisation of four AIV isolates of H3 subtype, recovered from wild mallards in Poland in 2006-2010, was performed. Phylogenetic analysis clearly confirms that there is a constant flow of AIV H3 between wild birds in Eurasia and Africa, and, to a limited degree, to North America (Alaska), with an occasional spill-over to poultry. The analysis of the PA gene of one isolate from 2010 suggests that it is closely related to several HPAI H5N1 viruses belonging to clade 2.3.2 and that, therefore, a reassortment event has occurred recently between low pathogenic and H5N1 highly pathogenic AIV.


Assuntos
Virus da Influenza A Subtipo H5N1 , Filogenia , Animais , Aves , Vírus da Influenza A , Influenza Aviária , Polônia , Aves Domésticas
15.
Pathogens ; 12(2)2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36839489

RESUMO

In this study, we investigated the clinical response, viral shedding, transmissibility, pathologic lesions, and tropism of HPAIV Gs/Gd H5N8 subtype (clade 2.3.4.4b), following experimental infection of three groups of captive mallards (Anas platyrhynchos): (i) fully susceptible, (ii) pre-exposed to low pathogenic avian influenza virus (LPAIV) H5N1 subtype, and (iii) pre-exposed to LPAIV H3N8 subtype. Infection of naïve mallards with HPAIV H5N8 resulted in ~60% mortality, neurological signs, abundant shedding, and transmission to contact ducks, who also became sick and died. High amounts of viral RNA were found in all collected organs, with the highest RNA load recorded in the brain. The IHC examinations performed on tissues collected at 4 and 14 days post-infection (dpi) revealed tropism to nervous tissue, myocardium, respiratory epithelium, and hepatic and pancreatic cells. The mallards pre-exposed to LPAIV H5N1 and challenged with HPAIV H5N8 were asymptomatic and showed a significant reduction of viral RNA shedding, yet still sufficient to cause infection (but no disease) in the contact ducks. The AIV antigen was not detected in organs at 4 and 14 dpi, and microscopic lesions were mild and scarce. Similarly, mallards previously inoculated with LPAIV H3N8 remained healthy after challenge with HPAIV H5N8, but viral RNA was detected in large quantities in swabs and organs, particularly in the early phase of infection. However, in contrast to mallards from group I, the IHC staining yielded negative results at the selected timepoints. The virus was transmitted to contact birds, which remained symptomless but demonstrated low levels of viral RNA shedding and mild- to moderate tissue damage despite negative IHC staining. The results indicate that naïve mallards are highly susceptible to HPAIV H5N8 clade 2.3.4.4b and that homo- and heterosubtypic immunity to LPAIV can mitigate the clinical outcomes of infection.

16.
Pathogens ; 11(1)2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-35056046

RESUMO

Bornaviruses are a diverse family of viruses infecting various hosts, including birds. Aquatic bird bornavirus 1 (ABBV-1) and aquatic bird bornavirus 2 (ABBV-2) have been found in wild waterfowl but data on their prevalence are scarce. To gain knowledge on the occurrence of ABBVs in Poland, samples originating from dead birds of the Anseriformes order collected in 2016-2021 were tested with a real time RT-PCR method targeting the ABBVs genome. A total of 514 birds were examined, including 401 swans, 96 ducks and 17 geese. The presence of ABBV-1 RNA was detected in 52 swans (10.1% of all tested birds) from 40 different locations. No positive results were obtained for ducks and geese. Sequences of about 2300 bases were generated for 18 viruses and phylogenetic analysis was performed. A relatively low genetic diversity of the examined ABBV-1 strains was observed as all were gathered in a single cluster in the phylogenetic tree and the minimum nucleotide identity was 99.14%. The Polish strains were closely related to ABBV-1 identified previously in Denmark and Germany, but a limited number of sequences from Europe hinders the drawing of conclusions about interconnections between Polish and other European ABBVs. The results of the present study provide new insights into the distribution and genetic characteristics of ABBVs in wild birds in Europe.

17.
J Vet Res ; 66(1): 1-7, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35582478

RESUMO

Introduction: Highly pathogenic avian influenza (HPAI) outbreaks caused by the Gs/Gd lineage of H5Nx viruses occur in Poland with increased frequency. The article provides an update on the HPAI situation in the 2020/2021 season and studies the possible factors that caused the exceptionally fast spread of the virus. Material and Methods: Samples from poultry and wild birds delivered for HPAI diagnosis were tested by real-time RT-PCR and a representative number of detected viruses were submitted for partial or full-genome characterisation. Information yielded by veterinary inspection was used for descriptive analysis of the epidemiological situation. Results: The scale of the epidemic in the 2020/2021 season was unprecedented in terms of duration (November 2020-August 2021), number of outbreaks in poultry (n = 357), wild bird events (n = 92) and total number of affected domestic birds (approximately ~14 million). The major drivers of the virus spread were the harsh winter conditions in February 2020 followed by the introduction of the virus to high-density poultry areas in March 2021. All tested viruses belonged to H5 clade 2.3.4.4b with significant intra-clade diversity and in some cases clearly distinguished clusters. Conclusion: The HPAI epidemic in 2020/2021 in Poland struck with unprecedented force. The conventional control measures may have limited effectiveness to break the transmission chain in areas with high concentrations of poultry.

18.
mBio ; 13(4): e0060922, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35726917

RESUMO

Phylogenetic evidence from the recent resurgence of high-pathogenicity avian influenza (HPAI) virus subtype H5N1, clade 2.3.4.4b, observed in European wild birds and poultry since October 2021, suggests at least two different and distinct reservoirs. We propose contrasting hypotheses for this emergence: (i) resident viruses have been maintained, presumably in wild birds, in northern Europe throughout the summer of 2021 to cause some of the outbreaks that are part of the most recent autumn/winter 2021 epizootic, or (ii) further virus variants were reintroduced by migratory birds, and these two sources of reintroduction have driven the HPAI resurgence. Viruses from these two principal sources can be distinguished by their hemagglutinin genes, which segregate into two distinct sublineages (termed B1 and B2) within clade 2.3.4.4b, as well as their different internal gene compositions. The evidence of enzootic HPAI virus circulation during the summer of 2021 indicates a possible paradigm shift in the epidemiology of HPAI in Europe.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Influenza Aviária , Animais , Animais Selvagens , Aves , Europa (Continente)/epidemiologia , Virus da Influenza A Subtipo H5N1/genética , Vírus da Influenza A/genética , Influenza Aviária/epidemiologia , Filogenia , Aves Domésticas
19.
Avian Dis ; 55(1): 106-12, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21500645

RESUMO

To assess the susceptibility of pigeons (Columba livia) to infection with H5N1 high pathogenicity avian influenza virus (HPAIV), four groups of 1-yr-old and 4-wk-old racing pigeons (10 birds in each group) were inoculated oculonasally with 106 50% egg infectious dose (EID50) of A/crested eagle/Belgium/01/2004 (clade 1) or A/swan/Poland/305-135V08/2006 (clade 2.2). Contact specific-pathogen-free (SPF) chickens were kept in the same isolators as young pigeons (two chickens per group). At 3, 5, 7, 10, and 14 days postinfection (PI) two pigeons from each infected group were selected randomly, and oropharyngeal and cloacal swabs (pigeons and contact chickens) as well as a number of internal organs (pigeons) were collected for viral RNA detection in real-time reverse transcription PCR (RRT-PCR) and histopathology. At the end of the experiment (14 days PI) blood samples from two pigeons in each group and from contact SPF chickens were also collected, and sera were tested using hemagglutination inhibition (HI) test and blocking enzyme-linked immunosorbent assay (bELISA). During the observation period all pigeons remained clinically healthy, and no gross lesions were observed in any of the infected groups. SPF contact chickens were also healthy and negative in RRT-PCR and HI tests. However, the clade 1 H5N1 virus produced more sustained infection manifested by the presence of histopathologic changes (consisting mainly of mild to moderate hemorrhagic and inflammatory lesions), prolonged persistence of viral RNA (detectable between 3 and 10 days PI) in a variety of tissues of both adult and juvenile birds (with highest RNA load in lungs and brain) as well as slight viral shedding from the trachea and cloaca, but without transmission to SPF contact chickens. Additionally, two clade 1-infected adult pigeons sacrificed at the end of experiment showed seroconversion in bELISA and HI test (using homologous virus as antigen). The viral RNA was found only at day 3 PI in one adult pigeon inoculated with dade 2.2 H5N1 virus, but neither microscopic lesions nor seroconversion were found in any other tested birds inoculated with A/swan/Poland/305-135V08/2006. Our results support the observations that pigeons are resistant to H5N1 HPAIV (no deaths or clinical signs), but there may be clade-dependent differences in the pathogenic potentials of H5N1 HPAIV of Asian origin.


Assuntos
Columbidae , Virus da Influenza A Subtipo H5N1 , Influenza Aviária/virologia , Animais , Encéfalo/patologia , Encéfalo/virologia , Suscetibilidade a Doenças , Influenza Aviária/imunologia , Fígado/patologia , Fígado/virologia , Pulmão/patologia , Pulmão/virologia , RNA Viral/genética , RNA Viral/isolamento & purificação , Baço/patologia , Baço/virologia , Traqueia/patologia , Traqueia/virologia
20.
J Vet Res ; 65(1): 45-52, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33817394

RESUMO

INTRODUCTION: Introduction of highly pathogenic avian influenza virus (HPAIV) into a country and its further spread may have a devastating impact on the poultry industry and lead to serious economic consequences. Various risk factors may increase the probability of HPAI outbreak occurrence but their relative influence is often difficult to determine. The study evaluates how the densities of selected poultry species and proximity to the areas inhabited by wild birds impacted HPAI outbreak occurrence during the recently reported epidemics in Poland. MATERIAL AND METHODS: The analysis was developed using these risk factors in the locations of affected and randomly chosen unaffected commercial farms. Generalised linear and non-linear models, specifically logistic regression, classification tree and random forest, were used to indicate the most relevant risk factors, to quantify their association with HPAI outbreak occurrence, and to develop a map depicting spatial risk distribution. RESULTS: The most important risk factors comprised the densities of turkeys, geese and ducks. The abundance of these species of poultry in an area increased the probability of HPAI occurrence, and their farming intensity in several areas of central, western, eastern and northern Poland put these areas at the highest risk. CONCLUSION: The results may improve the targeting of active surveillance, strengthen biosecurity in the areas at risk and contribute to early detection of HPAI in outbreak reoccurrences.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA