Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Avian Pathol ; 49(4): 335-341, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32242456

RESUMO

Two or more different live attenuated infectious bronchitis virus (IBV) vaccine types are often given to broilers to induce homologous protection as well as to broaden protection against other IBV types in the field. However, the ability of broilers to respond to three or four different antigenic types of IBV vaccine has not been examined experimentally. In this study, we vaccinated one-day-old broiler chicks by eyedrop with three or four different IBV vaccine types simultaneously. The presence and relative amount of each vaccine was examined in all of the birds by IBV type-specific real-time RT-PCR at 5 days post-vaccination and each vaccine was detected in all of the birds given that vaccine. The birds were challenged at 28 days of age and protection was measured by clinical signs, virus detection and by ciliostasis. Birds vaccinated with three different IBV types (Ark, Mass and GA98) were protected against challenge with each of those IBV types and were partially protected against challenge with the GA08 virus. Birds vaccinated with four different IBV types (Ark, Mass, GA98 and GA08) were protected against challenge with each of those IBV types with the exception of Mass challenged birds which clearly had 3/11 birds not protected based on individual ciliostasis scores, but had an average ciliostasis score of >50% which is considered protected. The results are important for the control of IBV because they indicate that simultaneous vaccination with up to four different IBV vaccine types can provide adequate protection against challenge for each type.


Assuntos
Galinhas/virologia , Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa/imunologia , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Vacinas Atenuadas/imunologia
2.
Avian Dis ; 59(3): 368-74, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26478154

RESUMO

In this study, we examined the association among clinical signs, ciliostasis, virus detection, and histopathology for evaluating protection of vaccinated chickens against homologous and heterologous infectious bronchitis virus (IBV) challenge. At 5 days following challenge with IBV, we found a good correlation among clinical signs, ciliostasis in the trachea, challenge virus detection, and microscopic lesions in the trachea, with all four criteria being negative in fully protected birds and positive in fully susceptible birds. In partially protected birds we observed clinical signs and detected challenge virus; however, the ciliated epithelium was intact. In a second experiment, we challenged fully protected, partially protected, and fully susceptible birds with IBV, and then at 5 days postchallenge we gave the birds an opportunistic bacterium intranasally. Twenty Bordetella avium colonies were recovered from one of five fully protected birds, and only five colonies were isolated from two of five partially protected birds without ciliostasis, whereas in birds with ciliostasis, numerous colonies were isolated. Obviously, decreasing IBV infection and replication in the upper respiratory tract will decrease transmission and mutations, leading to variant viruses, and herein we demonstrate that protection of the cilia will decrease secondary bacterial infections, which have been shown to lead to condemnations and increased mortality. Thus, it appears that examining both criteria would be important when evaluating IBV vaccine efficacy.


Assuntos
Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/imunologia , Animais , Galinhas , Cílios/patologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Imunidade Materno-Adquirida , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Traqueia/patologia
3.
Avian Dis ; 59(1): 149-52, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26292549

RESUMO

studies in our laboratory showed that the Arkansas-Delmarva Poultry Industry (Ark-DPI) vaccine given to 1-day-old chickens by hatchery spray cabinet replicated poorly and failed to adequately protect broilers against homologous virus challenge, whereas the same vaccine given by eye-drop did replicate and the birds were protected following homologous virus challenge. To determine if mechanical damage following spray application plays a role in failure of the Ark-DPI vaccine, we examined the morphology of three Ark-DPI vaccines from different manufacturers using an electron microscope and included a Massachusetts (Mass) vaccine as control. One of the Ark-DPI vaccines (vaccine A) and the Mass vaccine had significantly (P < 0.005) fewer spikes than the other two Ark-DPI vaccines. We also found that the Ark-DPI and Mass vaccines had significantly (P < 0.005) fewer spike proteins per virus particle when compared to their respective challenge viruses. This observation is interesting and may provide some insight into the mechanism behind infectious bronchitis virus attenuation. No obvious differences were observed in virus morphology and no consistent trend in the number of spikes per virion was found in before- and after-spray samples. We also determined the vaccine titer before and after spray in embryonated eggs and found that both Ark-DPI and Mass vaccines had a similar drop in titer, 0.40 logi and 0.310 logi, respec10ively. Based on these data, it appears that mechanical damage to the Ark-DPI vaccine is not occurring when delivered by a hatchery spray cabinet, suggesting that some other factor is contributing to the failure of that vaccine when given by that method.


Assuntos
Galinhas , Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa/imunologia , Vacinas Virais/imunologia , Administração por Inalação , Animais , Infecções por Coronavirus/prevenção & controle , Microscopia Eletrônica , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Vacinas Virais/administração & dosagem
4.
Avian Dis ; 58(3): 398-403, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25518434

RESUMO

Real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) assays have been used to detect the presence of challenge virus when the efficacy of infectious bronchitis virus (IBV) vaccine against field viruses is being experimentally evaluated. However, federal guidelines for licensing IBV vaccines indicate that challenge-virus detection following vaccination is to be conducted in embryonated eggs. In this study, we examined qRT-PCR data with the use of universal and type-specific primers and probe sets for IBV detection and compared those data with challenge-virus detection in embryonated eggs to determine if the two methods of evaluating vaccine efficacy are comparable. In addition, we tested the qRT-PCR assays on thermocyclers from two different manufacturers. We found the universal IBV primers and probe set to be comparable to challenge-virus detection in embryonated eggs. However, for some IBV types (Mass41 and Conn on the SmartCycler II and Ark, Mass41, Conn, and GA98 on the ABI 7500) the qRT-PCR assay was more sensitive than virus detection in embryonated eggs. This may simply be due to the universal IBV qRT-PCR assay being more sensitive than virus detection in eggs or to the assay detecting nucleic acid from nonviable virus. This finding is important and needs to be considered when evaluating challenge-virus detection for vaccination and challenge studies, because qRT-PCR could potentially identify positive birds that would otherwise be negative by virus detection in embryonated eggs; thus it could lead to a more stringent measure of vaccine efficacy. We also found that the IBV type-specific primers and probe sets designed in this study were in general less sensitive than the universal IBV primers and probe set. Only the Ark-DPI-spedcific assay on the SmartCycler II and the Ark-DPI-, Mass41-, and DE072/GA98- (for detection of GA98 virus only) specific assays on the ABI 7500 were comparable in sensitivity to virus detection in eggs. We found that a number of variables, including the virus type examined, primers and probe efficiency and stability, and assay conditions, including thermocycler platform, can affect the data obtained from qRT-PCR assays. These results indicate that qRT-PCR assays can be used to detect IBV challenge virus, but each assay, including the assay conditions and thermocycler, should be individually evaluated if those data are expected to be comparable to virus detection in embryonated eggs.


Assuntos
Ovos/virologia , Vírus da Bronquite Infecciosa/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Animais , Embrião de Galinha , Primers do DNA/genética , Vírus da Bronquite Infecciosa/classificação , Vírus da Bronquite Infecciosa/genética , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinas Virais/genética , Vacinas Virais/isolamento & purificação
5.
Avian Dis ; 56(4): 634-41, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23397833

RESUMO

Infectious bronchitis virus (IBV) is a gamma coronavirus that causes a highly contagious disease in chickens. The virus can affect the upper respiratory tract and the reproductive tract, and some strains can cause a nephritis. Different serotypes and genetic types of the virus have been identified worldwide and for the most part do not cross-protect. In addition, new types of the virus continue to arise due to mutations and recombination events in the viral genome, making this virus difficult to identify and extremely difficult to control. Surveillance and identification of IBV types is extremely important for control of the disease and the advancement of molecular methods have aided in this pursuit. Genetic typing of IBV, which involves reverse transcription-PCR amplification and sequence analysis of the S1 glycoprotein gene, has revolutionized diagnosis and identification of this virus by making it possible to type and compare the relatedness of a large number of virus isolates in a short period of time. The purpose of this review is to give an update on the strains of IBV currently circulating in commercial chickens worldwide and hopefully to present a clear picture of the relationship between many of these viruses. The information on IBV types presented herein is from published manuscripts, submissions to GenBank, our own unpublished data, and personal communications with scientists and diagnosticians working with IBV worldwide.


Assuntos
Galinhas , Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa/genética , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/fisiopatologia , Animais , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/fisiopatologia , Infecções por Coronavirus/virologia , Vírus da Bronquite Infecciosa/classificação , Filogenia , Doenças das Aves Domésticas/virologia
6.
Avian Dis ; 56(1): 90-6, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22545533

RESUMO

In an outbreak of highly pathogenic H5 and H7 avian influenza, rapid analysis of a large number of clinical samples with the potential to rapidly identify the virus subtype is extremely important. Herein, we report on the development of a rapid multiplex microsphere assay for the simultaneous detection of all avian influenza viruses (AIV) as well as the differentiation of H5, H7, N1, and N2 subtypes. A reverse transcriptase-PCR (RT-PCR) reaction, followed by hybridization of the amplified product with specific oligonucleotide probe-coated microspheres, was conducted in a multiplex format. Following incubation with a reporter dye, the fluorescence intensity was measured using a suspension array system. The limit of detection of the probe-coupled microspheres ranged from 1 x 10(5) to 1 x 10(9) copies of RT-PCR amplified product and the sensitivity of the multiplex assay ranged from 1 x 10(2.5) to 1 x 10(3.2) 50% embryo infectious doses of virus. The diagnostic accuracy of the assay, compared to the standard real-time RT-PCR, was evaluated using 102 swab samples from chickens exposed to low pathogenic AIV, and 97.05% of samples gave identical results with both the assays. The calculated specificity of the assay was 97.43%. Although the assay still needs to be validated, it appears to be a suitable diagnostic tool for detection and differentiation of avian influenza virus H5, H7, N1, and N2 subtypes.


Assuntos
Galinhas , Vírus da Influenza A/classificação , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/diagnóstico , Microesferas , Doenças das Aves Domésticas/diagnóstico , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Animais , Cloaca/virologia , Vírus da Influenza A/genética , Influenza Aviária/virologia , Orofaringe/virologia , Doenças das Aves Domésticas/virologia , RNA Viral/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Sensibilidade e Especificidade
7.
Avian Dis ; 56(3): 449-55, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23050459

RESUMO

Conventional and molecular epidemiologic studies have confirmed the ability of infectious bronchitis virus (IBV) to rapidly evolve and successfully circumvent extensive vaccination programs implemented since the early 1950s. IBV evolution has often been explained as variation in gene frequencies as if evolution were driven by genetic drift alone. However, the mechanisms regulating the evolution of IBV include both the generation of genetic diversity and the selection process. IBV's generation of genetic diversity has been extensively investigated and ultimately involves mutations and recombination events occurring during viral replication. The relevance of the selection process has been further understood more recently by identifying genetic and phenotypic differences between IBV populations prior to, and during, replication in the natural host. Accumulating evidence suggests that multiple environmental forces within the host, including immune responses (or lack thereof) and affinity for cell receptors, as well as physical and biochemical conditions, are responsible for the selection process. Some scientists have used or adopted the related quasispecies frame to explain IBV evolution. The quasispecies frame, while providing a distinct explanation of the dynamics of populations in which mutation is a frequent event, exhibits relevant limitations which are discussed herein. Instead, it seems that IBV populations evolving by the generation of genetic variability and selection on replicons follow the evolutionary mechanisms originally proposed by Darwin. Understanding the mechanisms underlying the evolution of IBV is of basic relevance and, without doubt, essential to appropriately control and prevent the disease.


Assuntos
Evolução Biológica , Variação Genética , Vírus da Bronquite Infecciosa/genética , Seleção Genética
8.
Avian Dis ; 56(1): 200-7, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22545547

RESUMO

Current avian influenza (AI) virus surveillance programs involving wild birds rely on sample collection methods that require refrigeration or low temperature freezing to maintain sample integrity for virus isolation and/or reverse-transcriptase (RT) PCR. Maintaining the cold chain is critical for the success of these diagnostic assays but is not always possible under field conditions. The aim of this study was to test the utility of Finders Technology Associates (FTA) cards for reliable detection of AI virus from cloacal and oropharyngeal swabs of wild birds. The minimum detectable titer was determined, and the effect of room temperature storage was evaluated experimentally using multiple egg-propagated stock viruses (n = 6). Using real time RT-PCR, we compared results from paired cloacal swab and samples collected on FTA cards from both experimentally infected mallards (Anasplatyrhynchos) and hunter-harvested waterfowl sampled along the Texas Gulf Coast. Based on the laboratory trials, the average minimal detectable viral titer was determined to be 1 x 10(4.7) median embryo infectious dose (EID50)/ml (range: 1 x 10(4.3) to 1 x 10(5.4) EID50/ml), and viral RNA was consistently detectable on the FTA cards for a minimum of 20 days and up to 30 days for most subtypes at room temperature (23 C) storage. Real-time RT-PCR of samples collected using the FTA cards showed fair to good agreement in live birds when compared with both real-time RT-PCR and virus isolation of swabs. AI virus detection rates in samples from several wild bird species were higher when samples were collected using the FTA cards compared with cloacal swabs. These results suggest that FTA cards can be used as an alternative sample collection method when traditional surveillance methods are not possible, especially in avian populations that have historically received limited testing or situations in which field conditions limit the ability to properly store or ship swab samples.


Assuntos
Patos , Vírus da Influenza A/isolamento & purificação , Influenza Aviária/virologia , RNA Viral/análise , Manejo de Espécimes/métodos , Virologia/métodos , Animais , Cloaca/virologia , Orofaringe/virologia , Reação em Cadeia da Polimerase , Especificidade da Espécie , Manejo de Espécimes/veterinária , Texas
9.
J Vet Sci ; 23(2): e21, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35187879

RESUMO

Newcastle disease (ND), infectious laryngotracheitis (ILT) and avian metapneumovirus (aMPV) can be similar making it critical to quickly differentiate them. Herein, we adapted pre-existing molecular-based diagnostic assays for NDV and ILTV, and developed new assays for aMPV A and B, for use under synchronized thermocycling conditions. All assays performed equivalently with linearity over a 5 log10 dynamic range, a reproducible (R² > 0.99) limit of detection of ≥ 10 target copies, and amplification efficiencies between 86.8%-98.2%. Using biological specimens for NDV and ILTV showed 100% specificity. Identical amplification conditions will simplify procedures for detection in diagnostic laboratories.


Assuntos
Metapneumovirus , Doença de Newcastle , Doenças das Aves Domésticas , Animais , Galinhas , Metapneumovirus/genética , Doença de Newcastle/diagnóstico , Vírus da Doença de Newcastle/genética , Aves Domésticas , Doenças das Aves Domésticas/diagnóstico , Reação em Cadeia da Polimerase em Tempo Real/veterinária
10.
Avian Dis ; 65(4): 631-636, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-35068108

RESUMO

Infectious bronchitis virus (IBV) is a highly infectious and transmissible gammacoronavirus that is nearly impossible to control through biosecurity. Coronaviruses are RNA viruses with an enormous capacity for rapid replication and high rates of mutation, leading to a tremendous amount of genetic diversity. Viral evolution occurs when selection working on genetic diversity leads to new mutations being fixed in the population over time. For IBV, the emergence of variant viruses is likely due to a combination of selection acting on existing genetic diversity, as well as on newly created mutations as the virus replicates, or genetic drift. Immunity against IBV creates a strong selection pressure; however, immunity can also reduce the viral load, decreasing replication and the development of new mutations. Examining the balance between immunity reducing infection, replication, and genetic diversity, and immune pressure selecting for new variants, is extremely difficult at best. Nonetheless, vaccination and immunity do play a role in the emergence of new antigenic variants of IBV. To complicate the situation even more, coronaviruses can undergo recombination, and several studies in the literature report recombination between IBV vaccines and field viruses. However, to our knowledge, unlike genetic drift, recombination alone has not been shown to result in a new antigenic and pathogenic IBV type emerging to cause widespread disease in poultry. Vaccines against IBV that result in an immune population can reduce transmission (basic reproductive number R0 less than 1), making vaccines for IBV the best control strategy available. However, IBV control remains extremely challenging because of the high number of antigenic variants causing disease in poultry and a limited number of vaccines that mostly provide only partial protection against infection and replication of those variants. Currently, there is one major variant IBV circulating in all sectors of US commercial poultry production: DMV/1639/11. This virus was initially detected in 2011, but only began causing significant disease in 2014/2015. Since then, it has affected all three sectors of poultry production (layers, breeders, broilers) and continues to predominate in certain regions of the United States. Additionally, a previously classified variant IBV, which is no longer considered a variant virus, GA08, is highly prevalent. This is attributed to heavy GA08-type IBV vaccine usage because disease caused by the GA08-type virus is rare. Interestingly, the major IBV detected in poultry for several decades, ArkDPI, is no longer among the most detected viruses in the United States. This change corresponds to the shift away from ArkDPI vaccine usage in the broiler sector as GA08 vaccine usage has increased and highlights the role IBV vaccines play in influencing viral populations in commercial chickens.


Estudio recapitulativo- Evolución molecular del virus de la bronquitis infecciosa y aparición de virus variantes que circulan en los Estados Unidos. El virus de la bronquitis infecciosa es un gammacoronavirus altamente infeccioso y transmisible que es casi imposible de controlar mediante bioseguridad. Los coronavirus son virus ARN con una enorme capacidad de replicación rápida y altas tasas de mutación, lo que conduce a una gran cantidad de diversidad genética. La evolución viral ocurre cuando la selección que tiene influencia sobre la diversidad genética conduce a la fijación de nuevas mutaciones en la población a lo largo del tiempo. En el caso del virus de la bronquitis infecciosa, la aparición de variantes virales probablemente se deba a una combinación de selección que actúa sobre la diversidad genética existente, así como a mutaciones recién creadas a medida que el virus se replica o desarrolla deriva genética. La inmunidad contra el virus de la bronquitis infecciosa crea una fuerte presión de selección; sin embargo, la inmunidad también puede reducir la carga viral, disminuyendo la replicación y el desarrollo de nuevas mutaciones. La evaluación del equilibrio entre la inmunidad que reduce la infección, la replicación, la diversidad genética y la presión inmune que selecciona nuevas variantes, es extremadamente difícil en el mejor de los casos. No obstante, la vacunación y la inmunidad desempeñan un papel en la aparición de nuevas variantes antigénicas del virus de bronquitis. Para complicar aún más la situación, los coronavirus pueden someterse a recombinación y varios estudios en la literatura describen acerca de la recombinación entre las vacunas de bronquitis infecciosa y los virus de campo. Sin embargo, hasta donde se conoce, a diferencia de la deriva genética, no se ha demostrado que la recombinación por sí sola dé como resultado nuevos tipos antigénicos o patógenos del virus de la bronquitis infecciosa que causen una enfermedad generalizada en la avicultura. Las vacunas contra el virus de la bronquitis infecciosa que dan como resultado poblaciones inmune pueden reducir la transmisión (número reproductivo básico R0 menor que 1), lo que hace que las vacunas contra bronquitis infecciosa sean la mejor estrategia de control disponible. Sin embargo, el control de la bronquitis infecciosa sigue siendo un gran desafío debido a la gran cantidad de variantes antigénicas que causan enfermedades en la avicultura y a una cantidad limitada de vacunas que, en su mayoría, brindan solo una protección parcial contra la infección y la replicación de esas variantes. Actualmente, existe una variante principal del virus de la bronquitis infecciosa que circula en todos los sectores de la producción avícola comercial de los Estados Unidos: la variante DMV/1639/11. Este virus se detectó inicialmente en 2011, pero solo comenzó a causar una enfermedad significativa entre los años 2014/2015. Desde entonces, ha afectado a los tres sectores de la producción avícola (ponedoras, reproductoras, pollos de engorde) y continúa predominando en ciertas regiones de los Estados Unidos. Además, una variante de este virus previamente clasificada, que ya no se considera una variante, el virus GA08, es muy prevalente. Esto se atribuye al uso intensivo de la vacuna contra este tipo GA08 porque la enfermedad causada por el virus de tipo GA08 es poco común. Curiosamente, el principal virus de la bronquitis detectado en la avicultura durante varias décadas, ArkDPI, ya no se encuentra entre los virus más detectados en los Estados Unidos. Este cambio corresponde a la disminución en el uso de la vacuna ArkDPI en el sector de pollos de engorde a medida que el uso de la vacuna GA08 ha aumentado y se destaca el papel que desempeñan las vacunas de bronquitis infecciosa en la influencia de las poblaciones virales en los pollos comerciales.


Assuntos
Infecções por Coronavirus , Vírus da Bronquite Infecciosa , Doenças das Aves Domésticas , Vacinas Virais , Animais , Galinhas , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Evolução Molecular , Vírus da Bronquite Infecciosa/genética , Estados Unidos
11.
Avian Pathol ; 39(3): 227-33, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20544430

RESUMO

In the present study we describe the rapid development of an attenuated live vaccine for GA08, a new serotype of infectious bronchitis virus, using a heat-treatment method. Incubation of the GA08 strain of IBV at 56 degrees C and passage in embryonated eggs was used as a method to fast track the attenuation process. The virus was incubated in a 56 degrees C water bath and aliquots were removed every 5 min for up to 1 h, and then each aliquot was inoculated into 10-day-old embryonated eggs. Virus with the longest incubation time that produced lesions in the embryos was harvested, again incubated at 56 degrees C as described and passaged in embryonated eggs. Attenuation of the virus, designated GA08/GA08HSp16/08, was verified in 1-day-old specific pathogen free chicks. A 10x dose of the vaccine was found to be safe for 2-week-old broiler chicks of commercial origin. The efficacy of the heat-treated attenuated virus was determined by vaccinating broiler chicks of commercial origin at 1 and 14 days of age intraocularly/intranasally. Vaccinated birds that were challenged with 10(4.5) median embryo infectious doses of pathogenic GA08 virus/bird at 28 days of age were protected from the disease, and challenge virus was only detected in the trachea of one of 21 birds by real-time reverse transcriptase-polymerase chain reaction at 5 days post challenge. The attenuation process took 10 weeks to complete, which is a substantially shorter time than required to attenuate infectious bronchitis virus by serial passage in embryonated eggs without heat treatment (38 weeks or more).


Assuntos
Infecções por Coronavirus/imunologia , Vírus da Bronquite Infecciosa/imunologia , Doenças das Aves Domésticas/imunologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos/imunologia , Animais Recém-Nascidos/virologia , Sequência de Bases , Embrião de Galinha/imunologia , Galinhas/imunologia , Galinhas/virologia , Infecções por Coronavirus/veterinária , Primers do DNA , Temperatura Alta , Abrigo para Animais/normas , Vírus da Bronquite Infecciosa/classificação , Vírus da Bronquite Infecciosa/genética , Filogenia , RNA Viral/genética , RNA Viral/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Segurança , Vacinas Atenuadas/imunologia , Vacinas Virais/administração & dosagem
12.
Avian Dis ; 54(1): 120-5, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20408410

RESUMO

Low pathogenic avian influenza H6N2 viruses were biologically characterized by infecting chickens and ducks in order to compare adaptation of these viruses in these species. We examined the clinical signs, virus shedding, and immune response to infection in 4-wk-old white leghorn chickens and in 2-wk-old Pekin ducks. Five H6N2 viruses isolated between 2000 and 2004 from chickens in California, and one H6N2 virus isolated from chickens in New York in 1998, were given intrachoanally at a dose of 1 x 10(6) 50% embryo infectious dose per bird. Oral-pharyngeal and cloacal swabs were taken at 2, 4, and 7 days postinoculation (PI) and tested by real-time reverse-transcriptase polymerase chain reaction for presence of virus. Serum was collected at 7, 14, and 21 days PI and examined for avian influenza virus antibodies by commercial enzyme-linked immunosorbent assay (ELISA) and hemagglutination inhibition (HI) testing. Virus shedding for all of the viruses was detected in the oral-pharyngeal swabs from chickens at 2 and 4 days PI, but only three of the five viruses were detected at 7 days PI. Only two viruses were detected in the cloacal swabs from the chickens. Virus shedding for four of the five viruses was detected in the oral-pharyngeal cavity of the ducks, and fecal shedding was detected for three of the viruses (including the virus not shed by the oral-pharyngeal route) in ducks at 4 and 7 days PI. All other fecal swabs from the ducks were negative. Fewer ducks shed virus compared to chickens. Both the chickens and the ducks developed antibodies, as evidenced by HI and ELISA titers. The data indicate that the H6N2 viruses can infect both chickens and ducks, but based on the number of birds shedding virus and on histopathology, the viruses appear to be more adapted to chickens. Virus shedding, which could go unnoticed in the absence of clinical signs in commercial chickens, can lead to transmission of the virus among poultry. However, the viruses isolated in 2004 did not appear to replicate or cause more disease than earlier virus isolates.


Assuntos
Galinhas , Patos , Vírus da Influenza A/patogenicidade , Influenza Aviária/virologia , Animais , Vírus da Influenza A/classificação , Vírus da Influenza A/genética , Influenza Aviária/patologia , Filogenia , Virulência
13.
J Virol Methods ; 276: 113773, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31712094

RESUMO

Infectious bronchitis (IB) is a highly contagious upper respiratory tract disease of chickens caused by infectious bronchitis virus (IBV), which has various serotypes that do not cross-protect. Vaccine control strategies for this virus are only effective when designed around the currently circulating serotypes. It is essential to not only rapidly detect IBV but also to identify the type of virus causing disease. Six TaqMan™-based quantitative real-time RT-PCR assays (Universal, Ark, Mass, DE/GA98, GA07, GA08) were developed and examined the sensitivity and specificity for each assay. Assays were developed targeting the hypervariable region in the S1 gene subunit. The analytical sensitivity of TaqMan™-based quantitative real-time RT-PCR assays (qRT-PCR) assays was evaluated using synthetic DNA standards that were identical with the target sequence and specificity was further validated using clinical and biological specimens. All developed assays performed equivalently when using synthetic DNA templates as standard material, as it achieved linearity over a 5 log10 dynamic range with a reproducible limit of detection of ≤10 target copies per reaction, with high calculated amplification efficiencies ranging between 90%-115%. Further validation of specificity using clinical and biological specimens was also successful.


Assuntos
Aves/virologia , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/veterinária , DNA Viral/síntese química , Vírus da Bronquite Infecciosa/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , Infecções por Coronavirus/virologia , Primers do DNA/genética , Sondas de DNA/genética , DNA Viral/genética , Vírus da Bronquite Infecciosa/classificação , Vírus da Bronquite Infecciosa/genética , Limite de Detecção , Reação em Cadeia da Polimerase em Tempo Real/normas , Estudos Retrospectivos , Sensibilidade e Especificidade
14.
Avian Dis ; 53(4): 613-7, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20095165

RESUMO

In this study, the sequence of the H5 and PB1 genes of the low-pathogenic avian influenza virus (LPAI) A/Black Duck/NC/674-964/06 isolate were determined for replikin peptides and used to design and chemically synthesize a vaccine. The vaccine was used to immunize specific-pathogen-free (SPF) leghorn chickens held in Horsfall isolation units, by the upper respiratory route, at 1, 7, and 14 days of age. The birds were challenged at 28 days of age with 1 x 10(6) 50% embryo infective dose of the LPAI Black Duck/NC/674-964/06 H5N1 virus per bird. Oropharyngeal and cloacal swabs were collected at 2, 4, and 7 days postinoculation (PI) for virus detection by real-time RT-PCR. Serum was collected at 7, 14, and 21 days PI and examined for antibodies against avian influenza virus by the enzyme-linked immunosorbent assay and hemagglutination inhibition (HI) tests. Tissue samples for histopathology were collected from three birds per group at 3 days PI. The experimental design consisted of a negative control group (not vaccinated and not challenged) and a vaccinated group, a vaccinated and challenged group, and a positive control group (challenged only). None of the nonchallenged birds, the vaccinated birds, or the vaccinated and challenged birds showed overt clinical signs of disease during the study. A slight depression was observed in the nonvaccinated challenged birds on day 2 postchallenge. Although the numbers of birds per group are small, no shedding of the challenge virus was detected in the vaccinated and challenged birds, whereas oropharyngeal and cloacal shedding was detected in the nonvaccinated and challenged birds. HI antibodies were detected in the vaccinated and nonchallenged group as well as in the vaccinated and challenged group, but rising antibody titers, indicating infection with the LPAI challenge virus, were not detected. Rising HI titers were observed in the nonvaccinated and challenged group. In addition, no antibodies were detected in the nonchallenged birds. Noteworthy microscopic lesions were not observed in the vaccinated and challenged birds, whereas nonvaccinated-challenged birds had microscopic lesions consistent with infection with LPAI viruses. Taken together, these data indicate that a replikin peptide vaccine, specifically made against the H5N1 Black Duck/NC/674-964/06 isolate, and administered three times to the upper respiratory tract, was capable of protecting chickens from infection and from shedding of the homologous virus, which is extremely important because reduced virus shedding and transmission decreases the potential for H5 LPAI viruses to become HPAI viruses. The study is also important because it shows that the vaccine can be effectively mass-delivered to the upper respiratory tract.


Assuntos
Galinhas , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Influenza Aviária/prevenção & controle , Animais , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Proteínas Recombinantes/imunologia , Organismos Livres de Patógenos Específicos , Proteínas Virais/genética , Proteínas Virais/imunologia
15.
Avian Dis ; 53(2): 175-83, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19630221

RESUMO

To determine the coverage of infectious bronchitis virus (IBV) vaccine field boost in commercial broilers, estimate the relative amount of vaccine virus in the trachea, and follow the clearance of the vaccine, we collected approximately 100 tracheal swabs at various times postvaccination from 10 different flocks and used real-time reverse transcriptase-PCR (RT-PCR) to detect the virus. This allowed us to detect vaccine virus in as few as 3% of the birds in a flock of 20,000 birds with a 95% confidence level. We found that the number of birds positive for IBV vaccine following vaccination in the field resembled a parabolic-shaped curve that peaked around 14 days postvaccination, or it resembled a sinusoidal-type wave with a frequency of about 2 wk. The patterns did not appear to correlate with water or spray vaccination methods, nor did they correlate with the type of backpack sprayer used. The highest number of positive birds in a flock ranged from 66% to 100%. The viral genome copies in the tracheal swabs, as determined by real-time RT-PCR, ranged from 1 x 10(2.6)/ml to 1 x 10(5.2)/ml and, in most studies, had a positive correlation with the number of birds positive for vaccine virus in the flock. On the last sample day of each study, 21, 28, or 35 days postvaccination, from 12% to 66% of the birds were still positive for vaccine virus, and although different IBV vaccine types were used in each study, only Arkansas vaccine virus was identified in selected samples on those days. Arkansas vaccine virus was also the only virus identified in selected samples at 1, 3, and 5 days postvaccination, clearly indicating that Arkansas vaccine virus is persisting in the birds. Protection studies conducted on birds vaccinated with Arkansas- and Delaware-type vaccines and removed from the field at 21 days postvaccination showed complete protection against challenge with Delaware (except for one bird), whereas protection against Arkansas challenge was between 37.5% and 62.5%. Our findings show that introduction of IBV vaccines into a commercial broiler flock do not necessarily follow a seemingly logical pattern of a high number of birds infected followed by clearance from the trachea, but resembled either a parabolic curve or a sinusoidal-type wave. In addition, Arkansas vaccine viruses are clearly persisting in commercial broilers, which may be because of incomplete protection observed for that IBV type.


Assuntos
Galinhas , Infecções por Coronavirus/veterinária , Vírus da Bronquite Infecciosa/imunologia , Vírus da Bronquite Infecciosa/fisiologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/imunologia , Animais , Infecções por Coronavirus/virologia , Imunização Secundária , Doenças das Aves Domésticas/virologia , Vacinas Virais/administração & dosagem
16.
Avian Dis ; 53(4): 552-62, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20095156

RESUMO

The pathogenesis, virus shedding, and serologic response in specific-pathogen-free (SPF) chickens and commercial turkeys against H4, H6, and H9 type low pathogenic avian influenza viruses (LPAI) from wild birds was examined. Four-week-old chickens and three-week-old turkeys were given 1 x 10(6) EID50 of LPAI per bird, intrachoanally, and examined for clinical signs for 3 wk. Oropharyngeal and cloacal swabs, and fecal samples, were collected at 2, 4, and 7 days postinoculation (PI) for virus detection by real-time RT-PCR. Serum was collected at 7, 14, and 21 days PI and examined for antibodies against avian influenza virus (AIV) by the enzyme-linked immunosorbant assay (ELISA) and hemagglutination inhibition tests. Tissue samples for histopathology were collected from three birds per group at 3 days PI. The hemagglutinin genes of the viruses were sequenced, and phylogenetic analysis was conducted. Clinical signs ranged from no clinical signs to moderate depression, decreased activity, and decreased food and water consumption. Based on virus detection results, SPF chickens were generally found to be shedding more virus from both the oropharynx and cloaca than were commercial turkeys. Microscopic lesion results in both species showed the predominance of lesions in the respiratory and gastrointestinal tract, which is consistent with the fact that these viruses are of low pathogenicity. In chickens and turkeys, oropharyngeal shedding strongly correlated with the lesions found in the upper respiratory tract. Turkeys had fewer lesions in the respiratory tract and more lesions in the gastrointestinal tract compared to chickens. Thirteen LPAI viruses caused seroconversion in commercial turkeys, whereas only 6 LPAI viruses caused seroconversion in SPF chickens. Phylogenetic analysis of the HA genes showed that the H4, H6, and H9 viruses evaluated here represented the full genetic diversity of North American AIVs of their respective subtypes. This data is important for surveillance and control because some of the LPAI viruses (of wild bird origin and examined in this study) that can infect and be shed by chickens and turkeys would be difficult to detect in commercial poultry. Specifically, detection is difficult because these viruses did not cause overt clinical disease or mortality, but only induced mild microscopic lesions and exhibited poor seroconversion.


Assuntos
Galinhas , Hemaglutininas/classificação , Vírus da Influenza A/genética , Vírus da Influenza A/patogenicidade , Influenza Aviária/virologia , Perus , Animais , Filogenia , Testes Sorológicos/veterinária , Organismos Livres de Patógenos Específicos , Virulência , Eliminação de Partículas Virais
17.
Viruses ; 11(2)2019 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-30717342

RESUMO

Avian respiratory disease causes significant economic losses in commercial poultry. Because of the need to protect long-lived poultry against respiratory tract pathogens from an early age, vaccination programs for pullets typically involve serial administration of a variety of vaccines, including infectious bronchitis virus (IBV), Newcastle disease virus (NDV), and infectious laryngotracheitis virus (ILTV). Often the interval between vaccinations is only a matter of weeks, yet it is unknown whether the development of immunity and protection against challenge when vaccines are given in short succession occurs in these birds, something known as viral interference. Our objective was to determine whether serially administered, live attenuated vaccines against IBV, NDV, and ILTV influence the development and longevity of immunity and protection against challenge in long-lived birds. Based on a typical pullet vaccination program, specific-pathogen-free white leghorns were administered multiple live attenuated vaccines against IBV, NDV, and ILTV until 16 weeks of age (WOA), after which certain groups were challenged with IBV, NDV, or ILTV at 20, 24, 28, 32, and 36 WOA. Five days post-challenge, viral load, clinical signs, ciliostasis, tracheal histopathology, and antibody titers in serum and tears were evaluated. We demonstrate that pullets serially administered live attenuated vaccines against IBV, NDV, and ILTV were protected against homologous challenge with IBV, NDV, or ILTV for at least 36 weeks, and conclude that the interval between vaccinations used in this study (at least 2 weeks) did not interfere with protection. This information is important because it shows that a typical pullet vaccination program consisting of serially administered live attenuated vaccines against multiple respiratory pathogens can result in the development of protective immunity against each disease agent.


Assuntos
Infecções por Coronavirus/veterinária , Infecções por Herpesviridae/veterinária , Longevidade , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/uso terapêutico , Animais , Anticorpos Antivirais/sangue , Galinhas , Infecções por Coronavirus/prevenção & controle , Feminino , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Galináceo 1 , Vírus da Bronquite Infecciosa , Vírus da Doença de Newcastle , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Vacinação/veterinária , Vacinas Atenuadas/uso terapêutico
18.
J Wildl Dis ; 55(2): 363-374, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30284951

RESUMO

During summer and early fall of 2012, the US experienced the largest outbreak of hemorrhagic disease (HD) on record; deer (both Odocoileus virginianus and Odocoileus hemionus) in 35 states were affected, including many northern states where HD typically does not occur. Epizootic hemorrhagic disease virus (EHDV) was the predominant virus isolated, with serotype 2 (EHDV-2) representing 66% (135/205) of all isolated viruses. Viruses within the EHDV serogroup are genetically similar, but we hypothesized that subtle genetic distinctions between viruses would exist across the geographic range of the outbreak if multiple EHDV-2 strains were responsible. We examined viral relatedness and molecular epidemiology of the outbreak by sequencing the mammalian binding protein (VP2) gene and the insect vector binding protein (VP7) gene of 34 EHDV-2 isolates from 2012 across 21 states. Nucleotide sequences of VP2 had 99.0% pairwise identity; VP7 nucleotide sequences had 99.1% pairwise identity. Very few changes were observed in either protein at the amino acid level. Despite the high genetic similarity between isolates, subtle nucleotide differences existed. Both VP2 and VP7 gene sequences separated into two distinct clades based on patterns of single-nucleotide polymorphisms after phylogenetic analysis. The clades were divided geographically into eastern and western clades, although those divisions were not identical between VP2 and VP7. There was also an association between percent sequence identity and geographic distance between isolates. We concluded that multiple EHDV-2 strains contributed to this outbreak.


Assuntos
Cervos/virologia , Surtos de Doenças , Vírus da Doença Hemorrágica Epizoótica/genética , Infecções por Reoviridae/veterinária , Animais , Filogenia , Infecções por Reoviridae/epidemiologia , Infecções por Reoviridae/virologia , Estados Unidos/epidemiologia
19.
Avian Dis ; 52(2): 235-44, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18646452

RESUMO

Intestinal samples collected from 43 commercial broiler and 33 commercial turkey flocks from all regions of the United States during 2005 and 2006 were examined for the presence of astrovirus, rotavirus, reovirus, and coronavirus by reverse transcription-polymerase chain reaction (PCR), and for the presence of groups 1 and 2 adenovirus by PCR. Phylogenetic analysis was performed to further characterize the viruses and to evaluate species association and geographic patterns. Astroviruses were identified in samples from 86% of the chicken flocks and from 100% of the turkey flocks. Both chicken astrovirus and avian nephritis virus (ANV) were identified in chicken samples, and often both viruses were detected in the same flock. Turkey astrovirus type-2 and turkey astrovirus type-1 were found in 100% and 15.4% of the turkey flocks, respectively. In addition, 12.5% of turkey flocks were positive for ANV. Rotaviruses were present in 46.5% of the chicken flocks tested and in 69.7% of the turkey flocks tested. Based upon the rotavirus NSP4 gene sequence, the chicken and turkey origin rotaviruses assorted in a species-specific manner. The turkey origin rotaviruses also assorted based upon geographical location. Reoviruses were identified in 62.8% and 45.5% of chicken and turkey flocks, respectively. Based on the reovirus S4 gene segment, the chicken and turkey origin viruses assorted separately, and they were distinct from all previously reported avian reoviruses. Coronaviruses were detected in the intestinal contents of chickens, but not turkeys. Adenoviruses were not detected in any chicken or turkeys flocks. Of the 76 total chicken and turkey flocks tested, only three chicken flocks were negative for all viruses targeted by this study. Most flocks were positive for two or more of the viruses, and overall no clear pattern of virus geographic distribution was evident. This study provides updated enteric virus prevalence data for the United States using molecular methods, and it reinforces that enteric viruses are widespread in poultry throughout the United States, although the clinical importance of most of these viruses remains unclear.


Assuntos
Galinhas/virologia , Enterite Transmissível dos Perus/virologia , Doenças das Aves Domésticas/virologia , Perus/virologia , Animais , Avastrovirus/classificação , Avastrovirus/genética , Avastrovirus/isolamento & purificação , Sequência de Bases , Coronavirus/classificação , Coronavirus/genética , Coronavirus/isolamento & purificação , Coronavirus do Peru/classificação , Coronavirus do Peru/genética , Coronavirus do Peru/isolamento & purificação , Primers do DNA/genética , DNA Viral/genética , Dados de Sequência Molecular , Orthoreovirus Aviário/classificação , Orthoreovirus Aviário/genética , Orthoreovirus Aviário/isolamento & purificação , Filogenia , Síndrome de Mortalidade do Peruzinho por Enterite/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Rotavirus/classificação , Rotavirus/genética , Rotavirus/isolamento & purificação , Estados Unidos
20.
Vaccine ; 36(41): 6077-6086, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30197283

RESUMO

Almost all commercial poultry are vaccinated against avian coronavirus infectious bronchitis virus (IBV) using live attenuated vaccines mass administered by spray at day of hatch. Although many different types of IBV vaccines are used successfully, the ArkDPI serotype vaccine, when applied by spray, does not infect and replicate sufficiently to provide protection against homologous challenge. In this study, we examined a different Ark vaccine strain (Ark99), which is no longer used commercially due to its reactivity in one day old chicks, to determine if it could be further attenuated by passage in embryonated eggs but still provide adequate protection. Further attenuation of the Ark99 vaccine was achieved by passage in embryonated eggs but ArkGA P1, P20, and P40 (designated ArkGA after P1) were still too reactive to be suitable vaccine candidates. However, ArkGA P60 when given by spray had little or no vaccine reaction in one day old broiler chicks, and it induced protection from clinical signs and ciliostasis following homologous challenge. In addition, vaccinated and challenged birds had significantly less challenge virus, an important measure of protection, compared to non-vaccinated and challenged controls. The full-length genomes of viruses from egg passages 1, 20, 40, and 60 were sequenced using the Illumina platform and the data showed single nucleotide polymorphisms (SNPs) had accumulated in regions of the genome associated with viral replication, pathogenicity, and cell tropism. ArkGA P60 accumulated the most SNPs in key genes associated with pathogenicity (polyprotein gene 1ab) and cell tropism (spike gene), compared to previous passages, which likely resulted in its more attenuated phenotype. These results indicate that the ArkGA P60 vaccine is safe for spray vaccination of broiler chicks and induces suitable protection against challenge with pathogenic Ark-type virus.


Assuntos
Infecções por Coronavirus/imunologia , Infecções por Coronavirus/prevenção & controle , Vírus da Bronquite Infecciosa/imunologia , Vírus da Bronquite Infecciosa/patogenicidade , Animais , Galinhas , Vírus da Bronquite Infecciosa/genética , Polimorfismo de Nucleotídeo Único/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sorogrupo , Vacinação , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Replicação Viral/imunologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa