Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Nature ; 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39053575

RESUMO

Highly pathogenic avian influenza (HPAI) H5N1 clade 2.3.4.4b virus has caused the death of millions of domestic birds and thousands of wild birds in the U.S. since January, 20221-4 Throughout this outbreak, spillovers to mammals have been frequently documented5-12. We report spillover of HPAI H5N1 virus in dairy cattle herds across several states in the U.S. The affected cows displayed clinical signs encompassing decreased feed intake, altered fecal consistency, respiratory distress, and decreased milk production with abnormal milk. Infectious virus and viral RNA were consistently detected in milk from affected cows. Viral distribution in tissues via immunohistochemistry and in situ hybridization revealed a distinct tropism of the virus for the epithelial cells lining the alveoli of the mammary gland in cows. Whole viral genome sequences recovered from dairy cows, birds, domestic cats, and a raccoon from affected farms indicated multidirectional interspecies transmissions. Epidemiologic and genomic data revealed efficient cow-to-cow transmission after apparently healthy cows from an affected farm were transported to a premise in a different state. These results demonstrate the transmission of HPAI H5N1 clade 2.3.4.4b virus at a non-traditional interface underscoring the ability of the virus to cross species barriers.

2.
Virol J ; 20(1): 298, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102683

RESUMO

BACKGROUND: Avian influenza is a highly contagious, agriculturally relevant disease that can severely affect the poultry industry and food supply. Eurasian-origin H5Nx highly pathogenic avian influenza viruses (HPAIV) (clade 2.3.4.4) have been circulating globally in wild birds with spill over into commercial poultry operations. The negative impact to commercial poultry renewed interest in the development of vaccines against these viruses to control outbreaks in the U.S. METHODS: The efficacy of three recombinant H5 vaccines delivered in ovo or day of age were evaluated in commercial broilers challenged with the 2015 U.S. H5N2 clade 2.3.4.4c HPAIV. The recombinant vaccines included an alphavirus RNA particle vaccine (RP-H5), an inactivated reverse genetics-derived (RG-H5) and recombinant HVT vaccine (rHVT-AI) expressing H5 hemagglutinin (HA) genes. In the first experiment, in ovo vaccination with RP-H5 or rHVT-AI was tested against HPAI challenge at 3 or 6 weeks of age. In a second experiment, broilers were vaccinated at 1 day of age with a dose of either 107 or 108 RP-H5, or RG-H5 (512 HA units (HAU) per dose). RESULTS: In experiment one, the RP-H5 provided no protection following in ovo application, and shedding titers were similar to sham vaccinated birds. However, when the RP-H5 was delivered in ovo with a boost at 3 weeks, 95% protection was demonstrated at 6 weeks of age. The rHVT-AI vaccine demonstrated 95 and 100% protection at 3 and 6 weeks of age, respectively, of challenged broilers with reduced virus shedding compared to sham vaccinated birds. Finally, when the RP-H5 and rHVT vaccines were co-administered at one day of age, 95% protection was demonstrated with challenge at either 3 or 6 weeks age. In the second experiment, the highest protection (92%) was observed in the 108 RP-H5 vaccinated group. Significant reductions (p < 0.05) in virus shedding were observed in groups of vaccinated birds that were protected from challenge. The RG-H5 provided 62% protection from challenge. In all groups of surviving birds, antibody titers increased following challenge. CONCLUSIONS: Overall, these results demonstrated several strategies that could be considered to protected broiler chickens during a H5 HPAI challenge.


Assuntos
Vírus da Influenza A Subtipo H5N2 , Vírus da Influenza A , Vacinas contra Influenza , Influenza Aviária , Animais , Galinhas , Vírus da Influenza A Subtipo H5N2/genética , Vacinas Sintéticas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética
3.
J Virol ; 90(21): 9967-9982, 2016 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-27558429

RESUMO

Wild aquatic birds have been associated with the intercontinental spread of H5 subtype highly pathogenic avian influenza (HPAI) viruses of the A/goose/Guangdong/1/96 (Gs/GD) lineage during 2005, 2010, and 2014, but dispersion by wild waterfowl has not been implicated with spread of other HPAI viruses. To better understand why Gs/GD H5 HPAI viruses infect and transmit more efficiently in waterfowl than other HPAI viruses, groups of mallard ducks were challenged with one of 14 different H5 and H7 HPAI viruses, including a Gs/GD lineage H5N1 (clade 2.2) virus from Mongolia, part of the 2005 dispersion, and the H5N8 and H5N2 index HPAI viruses (clade 2.3.4.4) from the United States, part of the 2014 dispersion. All virus-inoculated ducks and contact exposed ducks became infected and shed moderate to high titers of the viruses, with the exception that mallards were resistant to Ck/Pennsylvania/83 and Ck/Queretaro/95 H5N2 HPAI virus infection. Clinical signs were only observed in ducks challenged with the H5N1 2005 virus, which all died, and with the H5N8 and H5N2 2014 viruses, which had decreased weight gain and fever. These three viruses were also shed in higher titers by the ducks, which could facilitate virus transmission and spread. This study highlights the possible role of wild waterfowl in the spread of HPAI viruses. IMPORTANCE: The spread of H5 subtype highly pathogenic avian influenza (HPAI) viruses of the Gs/GD lineage by migratory waterfowl is a serious concern for animal and public health. H5 and H7 HPAI viruses are considered to be adapted to gallinaceous species (chickens, turkeys, quail, etc.) and less likely to infect and transmit in wild ducks. In order to understand why this is different with certain Gs/GD lineage H5 HPAI viruses, we compared the pathogenicity and transmission of several H5 and H7 HPAI viruses from previous poultry outbreaks to Gs/GD lineage H5 viruses, including H5N1 (clade 2.2), H5N8 and H5N2 (clade 2.3.4.4) viruses, in mallards as a representative wild duck species. Surprisingly, most HPAI viruses examined in this study replicated well and transmitted among mallards; however, the three Gs/GD lineage H5 HPAI viruses replicated to higher titers, which could explain the transmission of these viruses in susceptible wild duck populations.


Assuntos
Patos/virologia , Vírus da Influenza A/patogenicidade , Influenza Aviária/transmissão , Influenza Aviária/virologia , Animais , Animais Selvagens/virologia , Surtos de Doenças , Aves Domésticas/virologia , Doenças das Aves Domésticas/virologia
4.
BMC Vet Res ; 12(1): 260, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27876034

RESUMO

BACKGROUND: From December 2014 through June 2015, the US experienced the most costly highly pathogenic avian influenza (HPAI) outbreak to date. Most cases in commercial poultry were caused by an H5N2 strain which was a reassortant with 5 Eurasian lineage genes, including a clade 2.3.4.4 goose/Guangdong/1996 lineage hemagglutinin, and 3 genes from North American wild waterfowl low pathogenicity avian influenza viruses. The outbreak primarily affected turkeys and table-egg layer type chickens. Three isolates were selected for characterization in turkeys: the US index isolate from December 2014 (A/northern pintail/WA/40964/2014), and two poultry isolates from April 2015 (A/chicken/IA/13388/2015 and A/turkey/MN/12528/2015). RESULTS: Four week old broad-breasted white turkeys were inoculated with one of three doses (102, 104 or 106 50% egg infectious doses [EID50] per bird) of each of the isolates to evaluate infectious dose and pathogenesis. The mean bird infectious dose of A/northern pintail/WA/40964/2014 and A/turkey/MN/12528/2015 was 105 EID50 per bird, but was 103 EID50 per bird for A/chicken/IA/13388/2015, suggesting the latter had greater adaptation to gallinaceous birds. All three isolates had unusually long mean death time of 5.3-5.9 days post challenge, and the primary clinical signs were severe lethargy and neurological signs which started no more than 24 h before death (the average pre-clinical period was 4 days). Infected turkeys also shed high levels of virus by both the oropharyngeal and cloacal routes. CONCLUSIONS: The unusually long mean death times, high levels of virus in feces, and increased adaptation of the later viruses may have contributed to the rapid spread of the virus during the peak of the outbreak.


Assuntos
Vírus da Influenza A Subtipo H5N2/fisiologia , Influenza Aviária/patologia , Influenza Aviária/virologia , Perus , Animais , Interações Hospedeiro-Patógeno , Vírus da Influenza A Subtipo H5N2/patogenicidade , Fatores de Tempo
5.
Vet Res ; 46: 97, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26394750

RESUMO

Highly pathogenic avian influenza virus (HPAIV) and Newcastle disease virus (NDV) are two of the most important viruses affecting poultry worldwide and produce co-infections especially in areas of the world where both viruses are endemic; but little is known about the interactions between these two viruses. The objective of this study was to determine if co-infection with NDV affects HPAIV replication in chickens. Only infections with virulent NDV strains (mesogenic Pigeon/1984 or velogenic CA/2002), and not a lentogenic NDV strain (LaSota), interfered with the replication of HPAIV A/chicken/Queretaro/14588-19/95 (H5N2) when the H5N2 was given at a high dose (10(6.9) EID50) two days after the NDV inoculation, but despite this interference, mortality was still observed. However, chickens infected with the less virulent mesogenic NDV Pigeon/1984 strain three days prior to being infected with a lower dose (10(5.3-5.5) EID50) of the same or a different HPAIV, A/chicken/Jalisco/CPA-12283-12/2012 (H7N3), had reduced HPAIV replication and increased survival rates. In conclusion, previous infection of chickens with virulent NDV strains can reduce HPAIV replication, and consequently disease and mortality. This interference depends on the titer of the viruses used, the virulence of the NDV, and the timing of the infections. The information obtained from these studies helps to understand the possible interactions and outcomes of infection (disease and virus shedding) when HPAIV and NDV co-infect chickens in the field.


Assuntos
Galinhas , Vírus da Influenza A Subtipo H5N2/fisiologia , Vírus da Influenza A Subtipo H7N3/fisiologia , Influenza Aviária/imunologia , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/fisiologia , Doenças das Aves Domésticas/imunologia , Animais , Coinfecção/imunologia , Coinfecção/veterinária , Coinfecção/virologia , Influenza Aviária/mortalidade , Influenza Aviária/virologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/patogenicidade , Doenças das Aves Domésticas/mortalidade , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Virulência , Replicação Viral , Eliminação de Partículas Virais
6.
J Virol ; 87(16): 9086-96, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23760232

RESUMO

In June of 2012, an H7N3 highly pathogenic avian influenza (HPAI) virus was identified as the cause of a severe disease outbreak in commercial laying chicken farms in Mexico. The purpose of this study was to characterize the Mexican 2012 H7N3 HPAI virus (A/chicken/Jalisco/CPA1/2012) and determine the protection against the virus conferred by different H7 inactivated vaccines in chickens. Both adult and young chickens intranasally inoculated with the virus became infected and died at between 2 and 4 days postinoculation (p.i.). High virus titers and viral replication in many tissues were demonstrated at 2 days p.i. in infected birds. The virus from Jalisco, Mexico, had high sequence similarity of greater than 97% to the sequences of wild bird viruses from North America in all eight gene segments. The hemagglutinin gene of the virus contained a 24-nucleotide insert at the hemagglutinin cleavage site which had 100% sequence identity to chicken 28S rRNA, suggesting that the insert was the result of nonhomologous recombination with the host genome. For vaccine protection studies, both U.S. H7 low-pathogenic avian influenza (LPAI) viruses and a 2006 Mexican H7 LPAI virus were tested as antigens in experimental oil emulsion vaccines and injected into chickens 3 weeks prior to challenge. All H7 vaccines tested provided ≥90% protection against clinical disease after challenge and decreased the number of birds shedding virus and the titers of virus shed. This study demonstrates the pathological consequences of the infection of chickens with the 2012 Mexican lineage H7N3 HPAI virus and provides support for effective programs of vaccination against this virus in poultry.


Assuntos
Surtos de Doenças , Vírus da Influenza A Subtipo H7N3/patogenicidade , Vacinas contra Influenza/imunologia , Influenza Aviária/epidemiologia , Influenza Aviária/virologia , Estruturas Animais/virologia , Animais , Animais Domésticos , Aves , Galinhas , Análise por Conglomerados , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H7N3/imunologia , Vírus da Influenza A Subtipo H7N3/isolamento & purificação , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/isolamento & purificação , Influenza Aviária/patologia , Influenza Aviária/prevenção & controle , México/epidemiologia , Filogenia , RNA Ribossômico 28S/genética , RNA Viral/genética , Recombinação Genética , Homologia de Sequência , Análise de Sobrevida , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/isolamento & purificação , Carga Viral , Eliminação de Partículas Virais
7.
Vet Res ; 45: 1, 2014 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-24393488

RESUMO

Low pathogenicity avian influenza virus (LPAIV) and lentogenic Newcastle disease virus (lNDV) are commonly reported causes of respiratory disease in poultry worldwide with similar clinical and pathobiological presentation. Co-infections do occur but are not easily detected, and the impact of co-infections on pathobiology is unknown. In this study chickens and turkeys were infected with a lNDV vaccine strain (LaSota) and a H7N2 LPAIV (A/turkey/VA/SEP-67/2002) simultaneously or sequentially three days apart. No clinical signs were observed in chickens co-infected with the lNDV and LPAIV or in chickens infected with the viruses individually. However, the pattern of virus shed was different with co-infected chickens, which excreted lower titers of lNDV and LPAIV at 2 and 3 days post inoculation (dpi) and higher titers at subsequent time points. All turkeys inoculated with the LPAIV, whether or not they were exposed to lNDV, presented mild clinical signs. Co-infection effects were more pronounced in turkeys than in chickens with reduction in the number of birds shedding virus and in virus titers, especially when LPAIV was followed by lNDV. In conclusion, co-infection of chickens or turkeys with lNDV and LPAIV affected the replication dynamics of these viruses but did not affect clinical signs. The effect on virus replication was different depending on the species and on the time of infection. These results suggest that infection with a heterologous virus may result in temporary competition for cell receptors or competent cells for replication, most likely interferon-mediated, which decreases with time.


Assuntos
Coinfecção/veterinária , Vírus da Influenza A Subtipo H7N2/imunologia , Influenza Aviária/virologia , Doença de Newcastle/virologia , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/virologia , Vacinas Virais/imunologia , Animais , Galinhas , Coinfecção/patologia , Coinfecção/fisiopatologia , Coinfecção/virologia , Testes de Inibição da Hemaglutinação/veterinária , Influenza Aviária/patologia , Influenza Aviária/fisiopatologia , Doença de Newcastle/patologia , Doença de Newcastle/fisiopatologia , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Estudos Soroepidemiológicos , Organismos Livres de Patógenos Específicos , Distribuição Tecidual , Perus , Vacinas Virais/administração & dosagem
8.
Avian Dis ; 58(1): 46-52, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24758112

RESUMO

The adjuvant activity of chitosan (CS) and calcium phosphate (CAP) particles was studied following intranasal (mucosal) administration to commercial chickens with inactivated Newcastle disease virus (NDV) vaccine. After three vaccinations with inactivated NDV in combination with CS or CAP an increase in antibody titers in blood and mucosal samples in chickens was observed when compared with the administration of NDV antigen only. A lower level of humoral immunity was observed in broiler chickens compared to layer-type birds. The CS-based vaccine demonstrated higher antigenic and protective activity following lethal challenge than the vaccine containing CAP. Because CS particles efficiently changed mucosal and humoral immunity and protective activity, CS may in the future be considered for use as a potential adjuvant for production of vaccines for poultry.


Assuntos
Adjuvantes Imunológicos/metabolismo , Galinhas , Imunidade Humoral , Imunidade nas Mucosas , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/imunologia , Vacinas Virais/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Administração Intranasal/veterinária , Animais , Fosfatos de Cálcio/administração & dosagem , Fosfatos de Cálcio/metabolismo , Quitosana/administração & dosagem , Quitosana/metabolismo , Feminino , Doença de Newcastle/prevenção & controle , Doença de Newcastle/virologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia
9.
Virology ; 595: 110066, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38574415

RESUMO

Avian influenza virus (AIV) is a constant threat to animal health with recent global outbreaks resulting in the death of hundreds of millions of birds with spillover into mammals. Myxovirus-resistance (Mx) proteins are key mediators of the antiviral response that block virus replication. Mouse (Mu) Mx (Mx1) is a strong antiviral protein that interacts with the viral nucleoprotein to inhibit polymerase function. The ability of avian Mx1 to inhibit AIV is unclear. In these studies, Mu Mx1 was stably introduced into chicken DF1 cells to enhance the immune response against AIV. Following infection, titers of AIV were significantly decreased in cells expressing Mu Mx1. In addition, considerably less cytopathic effect (CPE) and matrix protein staining was observed in gene-edited cells expressing Mu Mx1, suggesting Mu Mx1 is broadly effective against multiple AIV subtypes. This work provides foundational studies for use of gene-editing to enhance innate disease resistance against AIV.


Assuntos
Galinhas , Imunidade Inata , Influenza Aviária , Proteínas de Resistência a Myxovirus , Replicação Viral , Animais , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , Linhagem Celular , Influenza Aviária/virologia , Influenza Aviária/imunologia , Influenza Aviária/genética , Camundongos , Mutagênese Insercional , Vírus da Influenza A/imunologia , Vírus da Influenza A/genética
10.
Front Microbiol ; 15: 1428248, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39035438

RESUMO

Avian metapneumovirus (aMPV) poses a significant threat to the poultry industry worldwide, primarily affecting turkeys and chickens. The recent detection of aMPV-A and -B subtypes in the United States marks a significant shift after a prolonged period free of aMPV following the eradication of the previously circulating subtype C. Hence, the demand for molecular diagnostic tests for aMPV has arisen due to their limited availability in the US market. In this study, we present the molecular characterization based on the complete genome sequence of aMPV subtype A, which was detected in the US for the first time. Four RT-qPCR positive samples were subjected to next-generation sequencing analysis, resulting in the assembly of one complete and one near-complete genome sequences. Phylogenetic analysis revealed that the isolated strains clustered within the aMPV-A subtype and were most closely related to recent Mexican strains. A detailed amino acid analysis identified unique mutations in the G gene of the US isolates compared to Mexican strains. Additionally, we compared the performance, cross-reactivity, and limit of detection of our revised aMPV subtype-specific RT-qPCR test with two commercial kits, demonstrating similar detection and subtyping capabilities. These findings highlight the importance of accurate diagnostic methods for disease management in the poultry industry, provide valuable insights into the epidemiology of aMPV, and underscore the need for continued vigilance and surveillance to mitigate its impact on poultry production.

11.
NPJ Vaccines ; 9(1): 136, 2024 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-39097573

RESUMO

Avian influenza poses a severe threat to poultry production and global food security, prompting the development of vaccination programs in numerous countries. Modified live virus (MLV) vaccines, with their potential for mass application, offer a distinct advantage over existing options. However, concerns surrounding reversion, recombination, and unintended transmission have hindered the progress of MLV development for avian influenza in poultry. To address these concerns, we engineered reassortment-impaired, non-transmissible, safe, immunogenic, and protective MLVs through the rearrangement of internal gene segments and additional modifications to the surface gene segments HA and NA. The unique peptide marker aspartic acid-arginine-proline-alanine-valine-isoleucine-alanine-asparragine (DRPAVIAN) was incorporated into HA, while NA was modified to encode the chicken interleukin-18 (ckIL18) gene (MLV-H9N2-IL). In vitro, the MLV-H9N2 and MLV-H9N2-IL candidates demonstrated stability and virus titers comparable to the wild-type H9N2 strain. In chickens, the MLV-H9N2 and MLV-H9N2-IL candidates did not transmit via direct contact. Co-infection studies with wild-type virus confirmed that the altered HA and NA segments exhibited fitness disadvantages and did not reassort. Vaccinated chickens showed no clinical signs upon vaccination, all seroconverted, and the inclusion of ckIL18 in the MLV-H9N2-IL vaccine enhanced neutralizing antibody production. A significant decrease in viral loads post-challenge underscored the protective effect of the MLVs. The MLV-H9N2-IL vaccine, administered via drinking water, proved immunogenic in chickens in a dose-dependent manner, generating protective levels of neutralizing antibodies upon aggressive homologous virus challenge. In summary, this study lays the groundwork for safe MLVs against avian influenza suitable for mass vaccination efforts.

12.
Virol J ; 10: 342, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24252391

RESUMO

BACKGROUND: Avian influenza virus (AIV) induced proinflammatory cytokine expression is believed to contribute to the disease pathogenesis following infection of poultry. However, there is limited information on the avian immune response to infection with low pathogenic avian influenza virus (LPAIV). METHODS: To gain a better understanding of the early viral-host interactions of LPAIV in chickens, primary chicken embryo hepatocytes (CEH) were infected with four different LPAIVs of U.S. origin. Kinetics of virus replication, transcription factor (c-Jun, p50 and IRF-3) activation and immune response gene (IL-6, IL-1beta, IFN-alpha and Mx) expression were studied at four different time points (6, 12, 24 and 48 hours) post infection and compared to non-infected controls. RESULTS: CEH can support growth of the tested LPAIVs when with trypsin supplementation. All four immune response genes tested were upregulated following infection as were transcription factors c-Jun, p50 and IRF-3. Amplification of these genes was dependant on virus replication (e.g. inclusion of trypsin), such that immune response genes and transcription factors were upregulated as viral titers increased. CONCLUSION: The results of these studies demonstrate the requirement of virus replication for innate immune regulation and broaden our understanding of transcription factor responses related to LPAIV infection in chickens.


Assuntos
Citocinas/metabolismo , Interações Hospedeiro-Patógeno , Imunidade Inata , Vírus da Influenza A/imunologia , Vírus da Influenza A/fisiologia , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Galinhas , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/virologia
13.
Front Vet Sci ; 10: 1250952, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37720472

RESUMO

Avian influenza viruses (AIVs) are naturally found in wild birds, primarily in migratory waterfowl. Although species barriers exist, many AIVs have demonstrated the ability to jump from bird species to mammalian species. A key contributor to this jump is the adaption of the viral RNA polymerase complex to a new host for efficient replication of its RNA genome. The AIV PB2 gene appears to be essential in this conversion, as key residues have been discovered at amino acid position 627 that interact with the host cellular protein, acidic nuclear phosphoprotein 32 family member A (ANP32A). In particular, the conversion of glutamic acid (E) to lysine (K) is frequently observed at this position following isolation in mammals. The focus of this report was to compare the distribution of PB2 627 residues from different lineages and origins of H5 AIV, determine the prevalence between historical and contemporary sequences, and investigate the ratio of amino acids in avian vs. mammalian AIV sequences. Results demonstrate a low prevalence of E627K in H5 non-Goose/Guangdong/1996-lineage (Gs/GD) AIV samples, with a low number of mammalian sequences in general. In contrast, the H5-Gs/GD lineage sequences had an increased prevalence of the E627K mutation and contained more mammalian sequences. An approximate 40% conversion of E to K was observed in human sequences of H5 AIV, suggesting a non-exclusive requirement. Taken together, these results expand our understanding of the distribution of these residues within different subtypes of AIV and aid in our knowledge of PB2 mutations in different species.

14.
Virology ; 586: 122-129, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37542819

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is believed to have a zoonotic origin with bats suspected as a natural host. In this work, we individually express the ACE2 of seven bat species including, little brown, great roundleaf, Pearson's horseshoe, greater horseshoe, Brazilian free-tailed, Egyptian rousette, and Chinese rufous horseshoe in DF1 cells and determine their ability to support attachment and replication of SARS-CoV-2 viruses. We demonstrate that the ACE2 receptor of all seven species made DF1 cells permissible to SARS-CoV-2. The level of virus replication differed between bat species and variants tested. The Wuhan lineage SARS-CoV-2 virus replicated to higher titers than either variant virus tested. All viruses tested grew to higher titers in cells expressing the human ACE2 gene compared to a bat ACE2. This study provides a practical in vitromethod for further testing of animal species for potential susceptibility to current and emerging SARS-CoV-2 viruses.


Assuntos
COVID-19 , Quirópteros , Animais , Humanos , SARS-CoV-2/genética , Enzima de Conversão de Angiotensina 2/genética , Receptores Virais/genética , Internalização do Vírus , Glicoproteína da Espícula de Coronavírus/genética
15.
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
16.
Avian Dis ; 56(3): 555-60, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23050473

RESUMO

Worldwide, Newcastle disease (ND) remains one of the most economically important diseases of poultry. Current vaccination strategies for commercial poultry include the use of inactivated and live ND vaccines that typically induce protection against virulent field viruses. Here, we tested the efficacy of an antigen-antibody complex (AAC) ND vaccine delivered in ovo. Commercial maternal antibody-positive broiler chickens (Gallus domesticus) were vaccinated in ovo with an AAC vaccine composed of live B1-LaSota Newcastle disease virus (NDV) complexed with NDV-specific antiserum, and then they were challenged at weekly intervals after hatch. Challenge viruses included three exotic ND disease (END) viruses: the neurotropic strain Texas GB NDV-92-01 (TxGB) and two viscerotropic isolates, one isolate from the 2002-2003 outbreak in California (California 2002 isolate S212676 [CA]) and the other isolate from a 1997 END outbreak in South Korea (South Korea 94-147 [SK]). Results demonstrate that maternal antibody was able to provide approximately 50% protection in either vaccinated or control chickens at 7 days of age after TxGB challenge. However, with challenge at > or = 14 days, most control birds died, whereas all AAC-vaccinated birds were protected. Challenge with the CA or SK viruses in chickens at 28 days of age resulted in 100% protection of vaccinated birds, whereas all control birds died. In addition, AAC-vaccinated birds displayed decreased incidence of viral shedding in oral and cloacal swabs than control birds. Antibody titers were significantly (P < 0.05) higher in vaccinated chickens, as determined by enzyme-linked immunosorbent assay and hemagglutinin-inhibition tests, than in nonvaccinated controls. Together, these results demonstrate the efficacy of AAC vaccines delivered in ovo to protect commercial poultry.


Assuntos
Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Galinhas , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle/patogenicidade , Óvulo/imunologia , Envelhecimento , Animais , Anticorpos Antivirais/sangue , Embrião de Galinha , Imunidade Materno-Adquirida , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/imunologia
17.
Avian Dis ; 66(4): 479-485, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36715482

RESUMO

The Reoviridae family represents the largest family of double-stranded RNA viruses, and members have been isolated from a wide range of mammals, birds, reptiles, fishes, insects, and plants. Orthoreoviruses, one of the 15 recognized genera in the Reoviridae family, can infect humans and nearly all mammals and birds. Genomic characterization of reoviruses has not been adopted on a large scale because of the complexity of obtaining sequences for all 10 segments. In this study, we develop a time-efficient and practical method to enrich reovirus sequencing reads from isolates that allows for full-genome recovery using a single-primer amplification method coupled with next-generation sequencing. We refer to this protocol as reovirus-single-primer amplification (R-SPA). Our results demonstrate that most of the genes are covered with at least 500 reads per base space. Furthermore, R-SPA covers both the 5' and 3' ends of each reovirus genes. In summary, this study presents a universal and fast amplification protocol that yields sufficient double-stranded cDNA and facilitates and expedites the whole-genome sequencing of reoviruses.


Protocolo universal de amplificación con un iniciador único para realizar la secuenciación del genoma completo de orthoreovirus aviares con ARN de doble cadena y segmentados La familia Reoviridae representa la familia más grande de virus de ARN de doble cadena y se han aislado miembros de una amplia variedad de mamíferos, aves, reptiles, peces, insectos y plantas. El género Orthoreovirus, uno de los 15 géneros reconocidos en la familia Reoviridae, pueden infectar a humanos y a casi todos los mamíferos y aves. La caracterización genómica de los reovirus no se ha adoptado a gran escala debido a la complejidad de obtener secuencias para los 10 segmentos. En este estudio, desarrollamos un método práctico y eficiente para enriquecer las lecturas de secuenciación de reovirus a partir de aislamientos que permite la recuperación del genoma completo utilizando un método de amplificación con un iniciador único junto con la secuenciación de próxima generación. Nos referimos a este protocolo como amplificación de un solo iniciador de reovirus (R-SPA). Estos resultados demuestran que la mayoría de los genes están cubiertos con al menos 500 lecturas por espacio base. Además, el método R-SPA cubre los extremos 5' y 3' de cada gene de reovirus. En resumen, este estudio presenta un protocolo de amplificación rápido y universal que produce suficiente ADN complementario de doble cadena y facilita y acelera la secuenciación del genoma completo de los reovirus.


Assuntos
Orthoreovirus Aviário , Orthoreovirus , Doenças das Aves Domésticas , Reoviridae , Humanos , Animais , Genoma Viral , RNA de Cadeia Dupla/genética , Doenças das Aves Domésticas/genética , Reoviridae/genética , Orthoreovirus/genética , Orthoreovirus Aviário/genética , Mamíferos/genética
18.
Virology ; 569: 1-12, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35217403

RESUMO

The SARS-CoV-2 (SARS-CoV-2) virus has caused a worldwide pandemic because of the virus's ability to transmit efficiently human-to-human. A key determinant of infection is the attachment of the viral spike protein to the host receptor angiotensin-converting enzyme 2 (ACE2). Because of the presumed zoonotic origin of SARS-CoV-2, there is no practical way to assess the susceptibility of every species to SARS-CoV-2 by direct challenge studies. In an effort to have a better predictive model of animal host susceptibility to SARS-CoV-2, we expressed the ACE2 and/or transmembrane serine protease 2 (TMPRSS2) genes from humans and other animal species in the avian fibroblast cell line, DF1, that is not permissive to infection. We demonstrated that expression of both human ACE2 and TMPRSS2 genes is necessary to support SARS-CoV-2 infection and replication in DF1 and a non-permissive sub-lineage of MDCK cells. Titers of SARS-CoV-2 in these cell lines were comparable to those observed in control Vero cells. To further test the model, we developed seven additional transgenic cell lines expressing the ACE2 and TMPRSS2 derived from Felis catus (cat), Equus caballus (horse), Sus domesticus (pig), Capra hircus (goat), Mesocricetus auratus (Golden hamster), Myotis lucifugus (Little Brown bat) and Hipposideros armiger (Great Roundleaf bat) in DF1 cells. Results demonstrate permissive replication of SARS-CoV-2 in cat, Golden hamster, and goat species, but not pig or horse, which correlated with the results of reported challenge studies. Cells expressing genes from either bat species tested demonstrated temporal replication of SARS-CoV-2 that peaked early and was not sustained. The development of this cell culture model allows for more efficient testing of the potential susceptibility of many different animal species for SARS-CoV-2 and emerging variant viruses.


Assuntos
COVID-19 , Quirópteros , Enzima de Conversão de Angiotensina 2/genética , Animais , Gatos , Quirópteros/metabolismo , Chlorocebus aethiops , Cavalos , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Suínos , Células Vero
19.
J Gen Virol ; 92(Pt 4): 931-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21177922

RESUMO

Newcastle disease virus (NDV) is an avian paramyxovirus that causes significant economic losses to the poultry industry worldwide. There is limited knowledge about the avian immune response to infection with virulent NDVs, and how this response may contribute to disease. In this study, pathogenesis and the transcriptional host response of chickens to a virulent NDV strain that rapidly causes 100% mortality was characterized. Using microarrays, a strong transcriptional host response was observed in spleens at early times after infection with the induction of groups of genes involved in innate antiviral and pro-inflammatory responses. There were multiple genes induced at 48 h post-infection including: type I and II interferons (IFNs), several cytokines and chemokines, IFN effectors and inducible nitric oxide synthase (iNOS). The increased transcription of nitric oxide synthase was confirmed by immunohistochemistry for iNOS in spleens and measured levels of nitric oxide in serum. In vitro experiments showed strong induction of the key host response genes, alpha IFN, beta interferon, and interleukin 1ß and interleukin 6, in splenic leukocytes at 6 h post-infection in comparison to a non-virulent NDV. The robust host response to virulent NDV, in conjunction with severe pathological damage observed, is somewhat surprising considering that all NDV encode a gene, V, which functions as a suppressor of class I IFNs. Taken together, these results suggest that the host response itself may contribute to the pathogenesis of this highly virulent strain in chickens.


Assuntos
Galinhas/imunologia , Imunidade Inata , Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/imunologia , Vírus da Doença de Newcastle/patogenicidade , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Animais , Citocinas/biossíntese , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Imuno-Histoquímica , Análise em Microsséries , Microscopia , Doença de Newcastle/patologia , Doença de Newcastle/virologia , Óxido Nítrico Sintase Tipo II/biossíntese , Doenças das Aves Domésticas/patologia , Baço/imunologia , Baço/virologia , Fatores de Tempo
20.
Immunogenetics ; 63(6): 363-75, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21286706

RESUMO

Myxovirus-resistance (Mx) proteins are produced by host cells in response to type I interferons, and some members of the Mx gene family in mammals have been shown to limit replication of influenza and other viruses. According to an early report, chicken Mx1 variants encoding Asn at position 631 have antiviral activity, whereas variants with Ser at 631 lack activity in experiments evaluating Mx1 complementary DNA (cDNA) expressed ectopically in a cell line. We evaluated whether the Mx1 631 dimorphism influenced pathogenesis of highly pathogenic avian influenza virus (HPAIV) infection in chickens of two commercial broiler lines, each segregating for Asn631 and Ser631 variants. Following intranasal infection with HPAIV strain A/Chicken/Queretaro/14588-19/1995 H5N2, chickens homozygous for Asn631 allele were significantly more resistant to disease based on early mortality, morbidity, or virus shedding than Ser631 homozygotes. Higher amounts of splenic cytokine transcripts were observed in the Ser631 birds after infection, consistent with higher viral loads seen in this group and perhaps contributing to their higher morbidity. Nucleotide sequence determination of Mx1 cDNAs demonstrated that the Asn631 variants in the two chicken lines differed at several amino acid positions outside 631. In vitro experiments with a different influenza strain (low pathogenicity) failed to demonstrate an effect of Mx1 Asn631 on viral replication suggesting that in vivo responses may differ markedly from in vitro, or that choice of virus strain may be critical in demonstrating effects of chicken Mx1. Overall, these studies provide the first evidence that Mx1 has antiviral effects in chickens infected with influenza virus.


Assuntos
Proteínas Aviárias/genética , Proteínas Aviárias/imunologia , Galinhas/genética , Galinhas/imunologia , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/imunologia , Influenza Aviária/genética , Influenza Aviária/imunologia , Alelos , Substituição de Aminoácidos , Animais , Citocinas/biossíntese , Feminino , Predisposição Genética para Doença , Variação Genética , Vírus da Influenza A/imunologia , Vírus da Influenza A/patogenicidade , Influenza Aviária/virologia , Masculino , Proteínas de Resistência a Myxovirus , Eliminação de Partículas Virais/genética , Eliminação de Partículas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA