Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Vaccine ; 42(18): 3756-3767, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38724417

RESUMO

A Newcastle disease virus (NDV)-vectored vaccine expressing clade 2.3.4.4b H5 Hemagglutinin was developed and assessed for efficacy against H5N1 highly pathogenic avian influenza (HPAI) in specific pathogen-free (SPF) chickens, broilers, and domestic ducks. In SPF chickens, the live recombinant NDV-vectored vaccine, rK148/22-H5, achieved complete survival against HPAI and NDV challenges and significantly reduced viral shedding. Notably, the live rK148/22-H5 vaccine conferred good clinical protection in broilers despite the presence of maternally derived antibodies. Good clinical protection was observed in domestic ducks, with decreased viral shedding. It demonstrated complete survival and reduced cloacal viral shedding when used as an inactivated vaccine from SPF chickens. The rK148/22-H5 vaccine is potentially a viable and supportive option for biosecurity measure, effectively protecting in chickens against the deadly clade 2.3.4.4b H5 HPAI and NDV infections. Furthermore, it aligns with the strategy of Differentiating Infected from Vaccinated Animals (DIVA).


Assuntos
Anticorpos Antivirais , Galinhas , Patos , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Virus da Influenza A Subtipo H5N1 , Influenza Aviária , Vírus da Doença de Newcastle , Vacinas de Produtos Inativados , Vacinas Sintéticas , Eliminação de Partículas Virais , Animais , Galinhas/imunologia , Influenza Aviária/prevenção & controle , Influenza Aviária/imunologia , Vírus da Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/genética , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/patogenicidade , Patos/virologia , Patos/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/sangue , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/genética , Organismos Livres de Patógenos Específicos , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/imunologia , Doença de Newcastle/prevenção & controle , Doença de Newcastle/imunologia , Vacinas Virais/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
2.
Pathogens ; 13(1)2024 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-38276168

RESUMO

Diverse immune responses in different chicken lines can result in varying clinical consequences following avian influenza virus (AIV) infection. We compared two widely used layer breeds, Lohmann Brown (LB) and Lohmann White (LW), to examine virus replication and immune responses against H9N2 AIV infection. The transcription profile in the spleen of H9N2-infected chickens was compared using a microarray. Confirmatory real-time RT-PCR was used to measure the expression of C-type lectin, OASL, and MX1 genes. Additionally, to investigate the role of chicken lectin receptors in vitro, two C-type lectin receptors (CLRs) were expressed in DF-1 cells, and the early growth of the H9N2 virus was evaluated. The LB chickens shed a lower amount of virus from the cloaca compared with the LW chickens. Different expression levels of C-type lectin-like genes were observed in the transcription profile, with no significant differences in OASL or MX gene expression. Real-time RT-PCR indicated a sharp decrease in C-type lectin levels in the spleen of H9N2-infected LW chickens. In vitro studies demonstrated that cells overexpressing CLR exhibited lower virus replication, while silencing of homeostatic CLR had no effect on AIV replication. This study demonstrated distinct immune responses to H9N2 avian influenza in LB and LW chickens, particularly with differences in C-type lectin expression, potentially leading to lower virus shedding in LB chickens.

3.
Vaccine ; 41(49): 7369-7376, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37932132

RESUMO

Highly pathogenic avian influenza virus (HPAIV) has caused widespread outbreaks in poultry in the Americas. Because of the duration and extent of these outbreaks, vaccine use may be an additional tool to limit virus spread. Three vaccines were evaluated for efficacy in chickens against a current North American clade 2.3.4.4b H5 HPAIV isolate, A/turkey/Indiana/3703-003/2022 H5N1. The vaccines included: 1) a commercial inactivated reverse genetics (rg) generated H5N1 product with a clade 2.3.4.4c H5 hemagglutinin (HA) (rgH5N1); 2) a commercial alphavirus RNA particle (RP) vaccine with the TK/IN/22 HA; and 3) an in-house inactivated rg produced vaccine with the TK/IN/22 HA and a North American lineage N9 neuraminidase (NA) (SEP-22-N9). Both inactivated vaccines were produced with HA genes that were modified to be low pathogenic and with the remaining genes from the PR8 influenza strain. All vaccines provided 100% protection against mortality and morbidity and all vaccines reduced virus shed by the oropharyngeal and cloacal routes significantly compared to sham vaccinates. However, differences were observed among the vaccines in quantities of virus shed at two- and four-days post challenge (DPC). To determine if infected birds could be identified after vaccination to aid surveillance programs, serum was collected from the RP and SEP-22-N9 vaccine groups at 7, 10, and 14 DPC to detect antibody to the NA and nucleoprotein (NP) of the challenge virus by enzyme linked lectin assay (ELLA) and ELISA. As early as 7DPC ELLA detected antibody in sera from 100% of the chickens in the RP vaccinated group and 70% of the chickens in the SEP-22-N9 vaccinated group. Antibody to the NP was detected by commercial ELISA in more than 50% of the birds in the RP vaccinated group at each time point.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A , Vacinas contra Influenza , Influenza Aviária , Animais , Galinhas , Vacinas de Produtos Inativados , América do Norte , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética
4.
Viruses ; 15(11)2023 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-38005949

RESUMO

Highly pathogenic avian influenza viruses (HPAIVs) of subtype H5 of the Gs/GD/96 lineage remain a major threat to poultry due to endemicity in wild birds. H5N1 HPAIVs from this lineage were detected in 2021 in the United States (U.S.) and since then have infected many wild and domestic birds. We evaluated the pathobiology of an early U.S. H5N1 HPAIV (clade 2.3.4.4b, 2021) and two H5N8 HPAIVs from previous outbreaks in the U.S. (clade 2.3.4.4c, 2014) and Europe (clade 2.3.4.4b, 2016) in chickens and turkeys. Differences in clinical signs, mean death times (MDTs), and virus transmissibility were found between chickens and turkeys. The mean bird infective dose (BID50) of the 2021 H5N1 virus was approximately 2.6 log10 50% embryo infective dose (EID50) in chickens and 2.2 log10 EID50 in turkeys, and the virus transmitted to contact-exposed turkeys but not chickens. The BID50 for the 2016 H5N8 virus was also slightly different in chickens and turkeys (4.2 and 4.7 log10 EID50, respectively); however, the BID50 for the 2014 H5N8 virus was higher for chickens than turkeys (3.9 and ~0.9 log10 EID50, respectively). With all viruses, turkeys took longer to die (MDTs of 2.6-8.2 days for turkeys and 1-4 days for chickens), which increased the virus shedding period and facilitated transmission to contacts.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vírus da Influenza A Subtipo H5N8 , Vírus da Influenza A , Influenza Aviária , Doenças das Aves Domésticas , Animais , Estados Unidos/epidemiologia , Vírus da Influenza A Subtipo H5N8/genética , Galinhas , Virus da Influenza A Subtipo H5N1/genética , Perus , Virulência , Vírus da Influenza A/genética , Animais Selvagens
5.
Virology ; 587: 109860, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37572517

RESUMO

Highly pathogenic avian influenza viruses (HPAIVs) of the A/goose/Guangdong/1/1996 lineage H5 clade 2.3.4.4b continue to have a devastating effect on domestic and wild birds. Full genome sequence analyses using 1369 H5N1 HPAIVs detected in the United States (U.S.) in wild birds, commercial poultry, and backyard flocks from December 2021 to April 2022, showed three phylogenetically distinct H5N1 virus introductions in the U.S. by wild birds. Unreassorted Eurasian genotypes A1 and A2 entered the Northeast Atlantic states, whereas a genetically distinct A3 genotype was detected in Alaska. The A1 genotype spread westward via wild bird migration and reassorted with North American wild bird avian influenza viruses. Reassortments of up to five internal genes generated a total of 21 distinct clusters; of these, six genotypes represented 92% of the HPAIVs examined. By phylodynamic analyses, most detections in domestic birds were shown to be point-source transmissions from wild birds, with limited farm-to-farm spread.

6.
Front Vet Sci ; 10: 1207289, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37546334

RESUMO

High pathogenicity avian influenza (HPAI) is a viral disease with devastating consequences for the poultry industry worldwide. Domestic ducks are a major source of HPAI viruses in many Eurasian countries. The infectivity and pathogenicity of HPAI viruses in ducks vary depending on host and viral factors. To assess the factors influencing the infectivity and pathogenicity of HPAI viruses in ducks, we compared the pathobiology of two HPAI viruses (H5N1 clade 2.3.2.1c and H5N6 clade 2.3.4.4e) in 5- and 25-week-old ducks. Both HPAI viruses caused mortality in a dose-dependent manner (104, 106, and 108 EID50) in young ducks. By contrast, adult ducks were infected but exhibited no mortality due to either virus. Viral excretion was higher in young ducks than in adults, regardless of the HPAI strain. These findings demonstrate the age-dependent mortality of clade 2.3.2.1c and clade 2.3.4.4e H5 HPAI viruses in ducks.

7.
Front Vet Sci ; 10: 1158233, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37396994

RESUMO

Eurasian-lineage highly pathogenic avian influenza (HPAI) H5 viruses have spread throughout Asia, the Middle East, Europe, Africa, and most recently, North and South America. These viruses are independently evolving into genetically and antigenically divergent clades, and broad-spectrum vaccines protecting against these divergent clades are needed. In this study, we developed a chimeric virus-like particle (VLP) vaccine co-expressing hemagglutinins from two clades (clades 1 and 2.3.2.1) of HPAI H5 viruses and performed comparative cross-clade hemagglutination inhibition (HI) analysis in chickens and ducks. The chimeric VLP immunization induced a significantly broader spectrum of antibodies against various clades of HPAI H5 viruses than monovalent VLPs both in chickens and ducks. While the chimeric VLP led to broadened antibody responses in both species, significantly lower levels of HI antibodies were elicited in ducks than in chickens. Moreover, boost immunization failed to increase antibody responses in ducks regardless of the VLPs used, in contrast to chickens that showed significantly enhanced antibody responses upon boost immunization. These results suggest (1) the potential application of the chimeric VLP technology in poultry to help control HPAI H5 viruses by offering broader antibody responses against antigenically different strains and (2) possible obstacles in generating high levels of antibody responses against HPAI H5 viruses in ducks via vaccination, implying the need for advanced vaccination strategies for ducks.

8.
Vaccine ; 41(33): 4787-4797, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37355454

RESUMO

Coronavirus disease 2019 (Covid-19) caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) became a pandemic, causing significant burden on public health worldwide. Although the timely development and production of mRNA and adenoviral vector vaccines against SARS-CoV-2 have been successful, issues still exist in vaccine platforms for wide use and production. With the potential for proliferative capability and heat stability, the Newcastle disease virus (NDV)-vectored vaccine is a highly economical and conceivable candidate for treating emerging diseases. In this study, a recombinant NDV-vectored vaccine expressing the spike (S) protein of SARS-CoV-2, rK148/beta-S, was developed and evaluated for its efficacy against SARS-CoV-2 in K18-hACE-2 transgenic mice. Intramuscular vaccination with low dose (106.0 EID50) conferred a survival rate of 76 % after lethal challenge of a SARS-CoV-2 beta (B.1.351) variant. When administered with a high dose (107.0 EID50), vaccinated mice exhibited 100 % survival rate and reduced lung viral load against both beta and delta variants (B.1.617.2). Together with the protective immunity, rK148/beta-S is an accessible and cost-effective SARS-CoV-2 vaccine.


Assuntos
COVID-19 , Vacinas Virais , Camundongos , Animais , Humanos , COVID-19/prevenção & controle , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/genética , Vacinas contra COVID-19 , Vírus da Doença de Newcastle/genética , Camundongos Transgênicos , Vacinas Virais/genética , Anticorpos Antivirais , Anticorpos Neutralizantes
9.
J Virol ; 96(14): e0027822, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35862690

RESUMO

The incursions of H7 subtype low-pathogenicity avian influenza virus (LPAIV) from wild birds into poultry and its mutations to highly pathogenic avian influenza virus (HPAIV) have been an ongoing concern in North America. Since 2000, 10 phylogenetically distinct H7 virus outbreaks from wild birds have been detected in poultry, six of which mutated to HPAIV. To study the molecular evolution of the H7 viruses that occurs when changing hosts from wild birds to poultry, we performed analyses of the North American H7 hemagglutinin (HA) genes to identify amino acid changes as the virus circulated in wild birds from 2000 to 2019. Then, we analyzed recurring HA amino acid changes and gene constellations of the viruses that spread from wild birds to poultry. We found six HA amino acid changes occurring during wild bird circulation and 10 recurring changes after the spread to poultry. Eight of the changes were in and around the HA antigenic sites, three of which were supported by positive selection. Viruses from each H7 outbreak had a unique genotype, with no specific genetic group associated with poultry outbreaks or mutation to HPAIV. However, the genotypes of the H7 viruses in poultry outbreaks tended to contain minor genetic groups less observed in wild bird H7 viruses, suggesting either a biased sampling of wild bird AIVs or a tendency of having reassortment with minor genetic groups prior to the virus's introduction to poultry. IMPORTANCE Wild bird-origin H7 subtype avian influenza viruses are a constant threat to commercial poultry, both directly by the disease they cause and indirectly through trade restrictions that can be imposed when the virus is detected in poultry. It is important to understand the genetic basis of why the North American lineage H7 viruses have repeatedly crossed the species barrier from wild birds to poultry. We examined the amino acid changes in the H7 viruses associated with poultry outbreaks and tried to determine gene reassortment related to poultry adaptation and mutations to HPAIV. The findings in this study increase the understanding of the evolutionary pathways of wild bird AIV before infecting poultry and the HA changes associated with adaptation of the virus in poultry.


Assuntos
Evolução Molecular , Glicoproteínas de Hemaglutininação de Vírus da Influenza , Vírus da Influenza A , Influenza Aviária , Doenças das Aves Domésticas , Aminoácidos/genética , Animais , Animais Selvagens , Aves , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A/genética , América do Norte , Filogenia , Aves Domésticas , Doenças das Aves Domésticas/virologia
10.
Microbiol Resour Announc ; 11(7): e0025322, 2022 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-35770996

RESUMO

We report the near-complete genome sequence of an avian orthoavulavirus 13 (AOAV-13) strain isolated from a wild goose fecal sample collected in South Korea in early 2020. The AOAV-13 sequence had a unique 3' trailer region, including an 84-nucleotide (nt) deletion and a 24-nt insertion, compared to the most closely related Chinese genome sequence from 2015.

11.
Infect Genet Evol ; 101: 105281, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35395408

RESUMO

H5Nx highly pathogenic avian influenza viruses (HPAIVs) have caused transboundary epizootics in poultry and wild birds. In 2016, the H5N6 subtype of clade 2.3.4.4e HPAIVs caused multiple outbreaks in Asia, including China, Japan, Korea, and Vietnam. However, the geographical spread pattern of 2.3.4.4e H5N6 HPAIV has not been clearly identified. To better understand the emergence and transmission history of 2.3.4.4e H5N6 HPAIV, we investigated the underlying epidemiologic processes associated with this viral spread by performing a Bayesian phylogeography analysis. The results revealed that wild waterfowl played a central role in the transboundary spread of clade 2.3.4.4e H5N6 HPAIV into both endemic and non-endemic countries, causing multiple incursions of the 2.3.4.4e H5N6 HPAIV into South Korea, Japan, and Vietnam. In our analysis, Guangdong province, China was estimated to be the most probable site where 2.3.4.4e H5N6 HPAIVs emerged prior to the transboundary transmissions. Continued genomic surveillance in both wild birds and poultry would be necessary for monitoring of HPAIV incursions. In addition, enhanced biosecurity would be key to preventing the HPAIV spread by minimizing contact between domestic poultry and wild birds.


Assuntos
Vírus da Influenza A , Influenza Aviária , Animais , Animais Selvagens , Teorema de Bayes , Aves , Vírus da Influenza A/genética , Japão/epidemiologia , Filogenia , Aves Domésticas
12.
Viruses ; 14(3)2022 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-35336961

RESUMO

Within-host viral diversity offers a view into the early stages of viral evolution occurring after a virus infects a host. In recent years, advances in deep sequencing have allowed for routine identification of low-frequency variants, which are important sources of viral genetic diversity and can potentially emerge as a major virus population under certain conditions. We examined within-host viral diversity in turkeys and chickens experimentally infected with closely related H7N3 avian influenza viruses (AIVs), specifically one high pathogenicity AIV (HPAIV) and two low pathogenicity AIV (LPAIVs) with different neuraminidase protein stalk lengths. Consistent with the high mutation rates of AIVs, an abundance of intra-host single nucleotide variants (iSNVs) at low frequencies of 2-10% was observed in all samples collected. Furthermore, a small number of common iSNVs were observed between turkeys and chickens, and between directly inoculated and contact-exposed birds. Notably, the LPAIVs have significantly higher iSNV diversities and frequencies of nonsynonymous changes than the HPAIV in both turkeys and chickens. These findings highlight the dynamics of AIV populations within hosts and the potential impact of genetic changes, including mutations in the hemagglutinin gene that confers the high pathogenicity pathotype, on AIV virus populations and evolution.


Assuntos
Influenza Aviária , Doenças das Aves Domésticas , Animais , Galinhas , Variação Genética , Vírus da Influenza A Subtipo H7N3/genética , Perus , Virulência/genética
13.
Transbound Emerg Dis ; 69(5): e1445-e1459, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35150205

RESUMO

The Mexican lineage H5N2 low pathogenic avian influenza viruses (LPAIVs) were first detected in 1994 and mutated to highly pathogenic avian influenza viruses (HPAIVs) in 1994-1995 causing widespread outbreaks in poultry. By using vaccination and other control measures, the HPAIVs were eradicated but the LPAIVs continued circulating in Mexico and spread to several other countries. To get better resolution of the phylogenetics of this virus, the full genome sequences of 44 H5N2 LPAIVs isolated from 1994 to 2011, and 6 detected in 2017 and 2019, were analysed. Phylogenetic incongruence demonstrated genetic reassortment between two separate groups of the Mexican lineage H5N2 viruses between 2005 and 2010. Moreover, the recent H5N2 viruses reassorted with previously unidentified avian influenza viruses. Bayesian phylogeographic results suggested that mechanical transmission involving human activity is the most probable cause of the virus spillover to Central American, Caribbean, and East Asian countries. Increased infectivity and transmission of a 2011 H5N2 LPAIV in chickens compared to a 1994 virus demonstrates improved adaptation to chickens, while low virus shedding, and limited contact transmission was observed in mallards with the same 2011 virus. The sporadic increase in basic amino acids in the HA cleavage site, changes in potential N-glycosylation sites in the HA, and truncations of PB1-F2 should be further examined in relation to the increased infectivity and transmission in poultry. The genetic changes that occur as this lineage of H5N2 LPAIVs continues circulating in poultry is concerning not only because of the effect of these changes on vaccination efficacy, but also because of the potential of the viruses to mutate to the highly pathogenic form. Continued vigilance and surveillance efforts, and the pathogenic and genetic characterization of circulating viruses, are required for the effective control of this virus.


Assuntos
Vírus da Influenza A Subtipo H5N2 , Vírus da Influenza A , Influenza Aviária , Aminoácidos Básicos/genética , Animais , Teorema de Bayes , Galinhas , Humanos , Vírus da Influenza A Subtipo H5N2/genética , Vírus da Influenza A/genética , México/epidemiologia , Filogenia , Aves Domésticas
14.
Animals (Basel) ; 12(3)2022 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-35158697

RESUMO

Non-typhoidal Salmonella spp. cause persistent asymptomatic infections in poultry. The consumption of Salmonella-infected poultry products is associated with food poisoning. One of the pathogens that causes such infections is Salmonella ser. Enteritidis (SE). Therefore, alternative measures are required for better control of salmonellosis and to reduce potential antibiotic use. Here, the efficacy of a mixture of lactic acid bacteria (LAB), formulated based on competitive exclusion, was evaluated. The LAB mixture was administered to 1- to 20-day-old chickens using different schemes; the chickens were then inoculated with an SE strain, which was previously identified to be prevalent in broiler breeder farms. Even with short-term administration, the group treated with LAB exhibited lower SE isolation levels in the spleen and cecal content and greater weight gain than that in the control group. This protective efficacy of LAB was retained even after two weeks without LAB administration. According to the results of animal experiments and field tests, evidence of SE infection was absent after treatment of the animals with the LAB formulation used in this study. Thus, this LAB mixture can be used as a potential strategy for protecting poultry farms from Salmonella contamination. This will also help reduce potential antibiotic use.

15.
Transbound Emerg Dis ; 69(2): 881-885, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33523603

RESUMO

We report the first detection of Y280-lineage H9N2 avian influenza viruses in live bird markets in Korea during July 2020. The viruses were isolated from domestic ducks and chickens traded in three markets in two different provinces, indicating dispersal of the newly introduced viruses. Complete genome sequencing and comparative phylogenetic analyses of all eight gene segments of the viruses showed high nucleotide homology to a Y280-lineage H9N2 avian influenza virus isolated in a chicken farm in China, which belongs to one of the most prevalent H9N2 genotypes in China. Increasing human cases of the same genotype H9N2 infection in China and the mammalian specific markers present in the viruses isolated suggest potential implications for public health.


Assuntos
Vírus da Influenza A Subtipo H9N2 , Influenza Aviária , Doenças das Aves Domésticas , Animais , Galinhas , China/epidemiologia , Vírus da Influenza A Subtipo H9N2/genética , Influenza Aviária/epidemiologia , Mamíferos , Filogenia , Doenças das Aves Domésticas/epidemiologia , República da Coreia/epidemiologia
16.
Viruses ; 13(9)2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34578433

RESUMO

An outbreak caused by H7N3 low pathogenicity avian influenza virus (LPAIV) occurred in commercial turkey farms in the states of North Carolina (NC) and South Carolina (SC), United States in March of 2020. Subsequently, H7N3 high pathogenicity avian influenza virus (HPAIV) was detected on a turkey farm in SC. The infectivity, transmissibility, and pathogenicity of the H7N3 HPAIV and two LPAIV isolates, including one with a deletion in the neuraminidase (NA) protein stalk, were studied in turkeys and chickens. High infectivity [<2 log10 50% bird infectious dose (BID50)] and transmission to birds exposed by direct contact were observed with the HPAIV in turkeys. In contrast, the HPAIV dose to infect chickens was higher than for turkeys (3.7 log10 BID50), and no transmission was observed. Similarly, higher infectivity (<2-2.5 log10 BID50) and transmissibility were observed with the H7N3 LPAIVs in turkeys compared to chickens, which required higher virus doses to become infected (5.4-5.7 log10 BID50). The LPAIV with the NA stalk deletion was more infectious in turkeys but did not have enhanced infectivity in chickens. These results show clear differences in the pathobiology of AIVs in turkeys and chickens and corroborate the high susceptibility of turkeys to both LPAIV and HPAIV infections.


Assuntos
Galinhas/virologia , Vírus da Influenza A Subtipo H7N3/patogenicidade , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Perus/virologia , Animais , Surtos de Doenças/veterinária , Genoma Viral , Vírus da Influenza A Subtipo H7N3/genética , Vírus da Influenza A Subtipo H7N3/isolamento & purificação , Influenza Aviária/epidemiologia , Influenza Aviária/transmissão , North Carolina/epidemiologia , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/transmissão , South Carolina/epidemiologia , Carga Viral , Virulência , Eliminação de Partículas Virais
17.
Vet Microbiol ; 260: 109180, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34271303

RESUMO

H2N2 subtype low pathogenic avian influenza viruses (LPAIVs) have persisted in live bird markets (LBMs) in the Northeastern United States since 2014. Although unrelated to the 1957 pandemic H2N2 lineage, there is concern that the virus could have animal and public health consequences because of high contact with humans and numerous species in the LBM system. The pathogenicity, infectivity, and transmissibility of six LBM H2N2 viruses isolated from three avian species in LBMs were examined in chickens. Two of these isolates were also tested in Pekin ducks and guinea fowl. Full genome sequence was obtained from all 6 isolates and evaluated for genetic markers for host adaptation and pathogenicity in poultry. Clinical signs were not observed in any host with any of the isolates, however one recent isolate was shed at higher titers than the other isolates and had the lowest bird infectious dose of all the isolates tested in all three species. This isolate, A/chicken/NY/19-012787-1/2019, was also the only isolate with a deletion in the stalk region of the neuraminidase protein (NA). This supports the theory that the NA stalk deletion is evidence of adaptation to gallinaceous poultry.


Assuntos
Galinhas/virologia , Patos/virologia , Genoma Viral/genética , Vírus da Influenza A Subtipo H2N2/patogenicidade , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Animais , Vírus da Influenza A Subtipo H2N2/genética , Vírus da Influenza A Subtipo H2N2/fisiologia , Influenza Aviária/transmissão , Doenças das Aves Domésticas/transmissão , Virulência
18.
J Virol ; 95(18): e0095521, 2021 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-34232725

RESUMO

Highly pathogenic avian influenza (HPAI) viruses from the H5Nx Goose/Guangdong/96 lineage continue to cause outbreaks in domestic and wild bird populations. Two distinct genetic groups of H5N8 HPAI viruses, hemagglutinin (HA) clades 2.3.4.4A and 2.3.4.4B, caused intercontinental outbreaks in 2014 to 2015 and 2016 to 2017, respectively. Experimental infections using viruses from these outbreaks demonstrated a marked difference in virulence in mallards, with the H5N8 virus from 2014 causing mild clinical disease and the 2016 H5N8 virus causing high mortality. To assess which gene segments are associated with enhanced virulence of H5N8 HPAI viruses in mallards, we generated reassortant viruses with 2014 and 2016 viruses. For single-segment reassortants in the genetic backbone of the 2016 virus, pathogenesis experiments in mallards revealed that morbidity and mortality were reduced for all eight single-segment reassortants compared to the parental 2016 virus, with significant reductions in mortality observed with the polymerase basic protein 2 (PB2), nucleoprotein (NP), and matrix (M) reassortants. No differences in morbidity and mortality were observed with reassortants that either have the polymerase complex segments or the HA and neuraminidase (NA) segments of the 2016 virus in the genetic backbone of the 2014 virus. In vitro assays showed that the NP and polymerase acidic (PA) segments of the 2014 virus lowered polymerase activity when combined with the polymerase complex segments of the 2016 virus. Furthermore, the M segment of the 2016 H5N8 virus was linked to filamentous virion morphology. Phylogenetic analyses demonstrated that gene segments related to the more virulent 2016 H5N8 virus have persisted in the contemporary H5Nx HPAI gene pool until 2020. IMPORTANCE Outbreaks of H5Nx HPAI viruses from the goose/Guangdong/96 lineage continue to occur in many countries and have resulted in substantial impact on wild birds and poultry. Epidemiological evidence has shown that wild waterfowl play a major role in the spread of these viruses. While HPAI virus infection in gallinaceous species causes high mortality, a wide range of disease outcomes has been observed in waterfowl species. In this study, we examined which gene segments contribute to severe disease in mallards infected with H5N8 HPAI viruses. No virus gene was solely responsible for attenuating the high virulence of a 2016 H5N8 virus, but the PB2, NP, and M segments significantly reduced mortality. The findings herein advance our knowledge on the pathobiology of avian influenza viruses in waterfowl and have potential implications on the ecology and epidemiology of H5Nx HPAI in wild bird populations.


Assuntos
Patos/virologia , Vírus da Influenza A Subtipo H5N8/classificação , Vírus da Influenza A Subtipo H5N8/patogenicidade , Influenza Aviária/transmissão , Influenza Aviária/virologia , Doenças das Aves Domésticas/virologia , Proteínas Virais/genética , Animais , Vírus da Influenza A Subtipo H5N8/genética , Filogenia , Doenças das Aves Domésticas/genética , Virulência
19.
Front Vet Sci ; 8: 616755, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34113668

RESUMO

Highly pathogenic avian influenza (HPAI) is considered as one of the most devastating poultry diseases. It is imperative to immediately report any known outbreaks to the World Organization for Animal Health. Early detection of infected birds is of paramount importance to control virus spread, thus minimizing the associated economic loss. In this study, thermal imaging camera devices were used to detect change in the maximum surface temperature (MST) of chickens (n = 5) and ducks (n = 2) as an early indicator of experimental HPAI infection. The MST of both chickens and ducks increased at least 24 h before the manifestation of clinical signs of HPAI infection, depending on the severity of the infection. The basal MST was recorded for broiler chickens housed under small pen and normal farm conditions without intentional infection. A threshold cutoff of MST was established based on the circadian rhythm of normal MST. This study suggests that thermal imaging of chickens and ducks is a promising tool to screen any potential HPAI-infected flock in order to expedite HPAI diagnosis.

20.
Front Vet Sci ; 8: 773715, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35187138

RESUMO

The H5 subtype highly pathogenic avian influenza virus (HPAIV) has been introduced to South Korea every 2 or 3 years via wild migratory waterfowls, causing devastating damages to the poultry industry. Although most damages and economic losses by HPAIV are focused on chicken layers, domestic ducks are known to play a major role in the farm-to-farm transmission. However, most HPAIV vaccine studies on poultry have been performed with oil-emulsion inactivated vaccines. In this study, we developed a live recombinant Newcastle disease virus (NDV)-vectored vaccine against H5 HPAIV (rK148/ES2-HA) using a previously established NDV vaccine strain (K148/08) isolated from a wild mallard duck. The efficacy of the vaccine when administered via the oculonasal route or as a spray was evaluated against lethal H5 HPAIV infection in domestic ducks and chickens. Oculonasal inoculation of the rK148/ES2-HA in chickens and ducks elicited antibody titers against HPAIV as early as 1 or 2 week after the single dose of vaccination, whereas spray vaccination in ducks elicited antibodies against HPAIV after the booster vaccination. The chickens and ducks vaccinated with rK148/ES2-HA showed high survival rates and low viral shedding after H5N6 HPAIV challenge. Collectively, vaccination with rK148/ES2-HA prevented lethal infection and decreased viral shedding in both chickens and ducks. The vaccine developed in this study could be useful in suppressing the viral shedding in H5 HPAIV outbreaks, with the ease of vaccine application and fast onset of immunity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA