Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Avian Pathol ; 48(3): 209-220, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30640536

RESUMO

Infectious laryngotracheitis (ILT) is an acute respiratory disease of chickens controlled through vaccination with live-modified attenuated vaccines, the chicken embryo origin (CEO) vaccines and the tissue-culture origin (TCO) vaccines. Recently, novel recombinant vaccines have been developed using herpesvirus of turkey (HVT) and fowl pox virus (FPV) as vectors to express ILTV immunogens for protection against ILT. The objective of this study was to assess the protection efficacy against ILT induced by recombinants, live-modified attenuated, and inactivated virus vaccines when administered alone or in combination. Commercial layer pullets were vaccinated with one or more vaccines and challenged at 35 (35 WCH) or 74 weeks of age (74 WCH). Protection was assessed by scoring clinical signs; and by determining the challenge viral load in the trachea at five days post-challenge. The FPV-LT vaccinated birds were not protected when challenged at 35 weeks; the HVT-LT and TCO vaccines in combination provided protection similar to that observed in chickens vaccinated with either HVT-LT or TCO vaccines when challenged at 35 weeks, whereas protection induced by vaccination with HVT-LT followed by TCO was superior in the 74 WCH group compared with the 35 WCH group. Birds given the inactivated ILT vaccine had fewer clinical signs and/or lower viral replication at 74 WCH when combined with TCO or HVT-LT, but not when given alone. Finally, the CEO-vaccinated birds had top protection as indicated by reduction of clinical signs and viral replication when challenged at 35 weeks (74 weeks not done). These results suggest that certain vaccine combinations may be successful to produce long-term protection up to 74 weeks of age against ILT.


Assuntos
Galinhas/imunologia , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinação/veterinária , Vacinas Virais/imunologia , Animais , Galinhas/virologia , Feminino , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos/genética , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Meleagrídeo 1/genética , Doenças das Aves Domésticas/virologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia , Vacinas Virais/administração & dosagem
2.
Avian Pathol ; 47(5): 497-508, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29963906

RESUMO

The objective of this study was to determine how cytokine transcription profiles correlate with patterns of infectious laryngotracheitis virus (ILTV) replication in the trachea, Harderian gland, and trigeminal ganglia during the early and late stages of infection after intratracheal inoculation. Viral genomes and transcripts were detected in the trachea and Harderian gland but not in trigeminal ganglia. The onset of viral replication in the trachea was detected at day one post-infection and peaked by day three post-infection. The peak of pro-inflammatory (CXCLi2, IL-1ß, IFN-γ) and anti-inflammatory (IL-13, IL-10) cytokine gene transcription, 5 days post-infection, coincided with the increased recruitment of inflammatory cells, extensive tissue damage, and limiting of virus replication in the trachea. In contrast, transcription of the IFN-ß gene in the trachea remained unaffected suggesting that ILTV infection blocks type I interferon responses. In the Harderian gland, the most evident transcription change was the early and transient upregulation of the IFN-γ gene at 1 day post-infection, which suggests that the Harderian gland is prepared to rapidly respond to ILTV infection. Overall, results from this study suggest that regulation of Th1 effector cells and macrophage activity by Th1/2 cytokines was pertinent to maintain a balanced immune response capable of providing an adequate Th1-mediated protective immunity, while sustaining some immune homeostasis in preparation for the regeneration of the tracheal mucosa.


Assuntos
Citocinas/metabolismo , Glândula de Harder/metabolismo , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/patogenicidade , Traqueia/metabolismo , Gânglio Trigeminal/metabolismo , Animais , Galinhas , Citocinas/genética , DNA , Regulação da Expressão Gênica/imunologia , Genoma Viral , Glândula de Harder/virologia , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/fisiologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/virologia , RNA , Organismos Livres de Patógenos Específicos , Traqueia/virologia , Transcrição Gênica , Gânglio Trigeminal/virologia , Carga Viral , Virulência , Replicação Viral
3.
Avian Pathol ; 47(5): 489-496, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29974790

RESUMO

Chicken infectious anaemia virus (CIAV) is a widely distributed immunosuppressive agent. SPF flocks and eggs used for vaccine production and diagnostics must be CIAV-free. Detection of CIAV infection in SPF flocks involves primarily serology or other invasive methods. In order to evaluate different types of samples for rapid detection of CIAV infection, a trial was conducted in serologically negative broiler breeder pullets vaccinated with a commercial live-attenuated CIAV vaccine. Controls and vaccinated groups were sampled before and after vaccination. Invasive and non-invasive samples were used for CIAV DNA detection by real-time PCR. Seroconversion occurred at 14 days post-inoculation (DPI) in the vaccinated group, whereas CIAV genome was detected by qPCR at 7 DPI in both invasive and non-invasive samples. Only invasive samples remained qPCR positive for CIAV DNA by 21 DPI despite seroconversion of the chickens.


Assuntos
Vírus da Anemia da Galinha/genética , Galinhas , Infecções por Circoviridae/veterinária , Doenças das Aves Domésticas/virologia , Animais , Infecções por Circoviridae/virologia , DNA Viral/genética , Feminino , Masculino , Reação em Cadeia da Polimerase em Tempo Real
4.
Avian Pathol ; 46(6): 585-593, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28532159

RESUMO

Infectious laryngotracheitis virus (ILTV) has a high proclivity to replicate in the larynx and trachea of chickens causing severe lesions. There is a lack of knowledge on the ability of ILTV to replicate in other respiratory associated tissues apart from in the trachea. The objective of this study was to investigate how tissues that first encounter the virus dictate further sites of viral replication during the lytic stage of infection. Replication patterns of the pathogenic strain 63140 and the chicken embryo origin (CEO) vaccine in the conjunctiva, the Harderian gland, nasal cavity and trachea were evaluated after ocular, oral, intranasal or intratracheal inoculation of specific pathogen-free chickens. Viral replication was assessed by detection of microscopic cytolytic lesions, detection of viral antigen and viral genome load. The route of viral entry greatly influenced virus replication of both strain 63140 and CEO vaccine in the conjunctiva and trachea, while replication in the nasal cavity was not affected. In the Harderian gland, independently of the route of viral entry, microscopic lesions characteristic of lytic replication were absent, whereas viral antigen and viral genomes for either virus were detected, suggesting that the Harderian gland may be a key site of antigen uptake. Findings from this study suggest that interactions of the virus with the epithelial-lymphoid tissues of the nasal cavity, conjunctiva and the Harderian gland dictate patterns of ILTV lytic replication.


Assuntos
Galinhas/imunologia , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/fisiologia , Doenças das Aves Domésticas/virologia , Vacinas Virais/imunologia , Animais , Embrião de Galinha , Galinhas/virologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/imunologia , Herpesvirus Galináceo 1/patogenicidade , Doenças das Aves Domésticas/prevenção & controle , Organismos Livres de Patógenos Específicos , Traqueia/virologia , Vacinação/veterinária , Carga Viral/veterinária , Replicação Viral
5.
J Virol ; 89(9): 4712-9, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25673726

RESUMO

UNLABELLED: Avian leukosis virus subgroup J (ALV-J) is a simple retrovirus that can cause hemangiomas and myeloid tumors in chickens and is currently a major economic problem in Asia. Here we characterize ALV-J strain PDRC-59831, a newly studied U.S. isolate of ALV-J. Five-day-old chicken embryos were infected with this virus, and the chickens developed myeloid leukosis and hemangiomas within 2 months after hatching. To investigate the mechanism of pathogenesis, we employed high-throughput sequencing to analyze proviral integration sites in these tumors. We found expanded clones with integrations in the MET gene in two of the five hemangiomas studied. This integration locus was not seen in previous work characterizing ALV-J-induced myeloid leukosis. MET is a known proto-oncogene that acts through a diverse set of signaling pathways and is involved in many neoplasms. We show that tumors harboring MET integrations exhibit strong overexpression of MET mRNA. IMPORTANCE: These data suggest that ALV-J induces oncogenesis by insertional mutagenesis, and integrations in the MET oncogene can drive the overexpression of MET and contribute to the development of hemangiomas.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/virologia , Hemangioma/virologia , Proteínas Proto-Oncogênicas c-met/genética , Integração Viral , Animais , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , Sequenciamento de Nucleotídeos em Larga Escala , Estados Unidos
6.
Avian Pathol ; 45(1): 106-13, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26926298

RESUMO

Infectious laryngotracheitis is a highly contagious disease of chickens responsible for significant economic losses for the poultry industry worldwide. The disease is caused by Gallid herpesvirus-1 (GaHV-1) commonly known as the infectious laryngotracheitis virus. Although characterized by their potential to regain virulence, chicken embryo origin (CEO) vaccines are the most effective vaccines against laryngotracheitis as they significantly reduce the replication of challenge virus in the trachea and conjunctiva. Knowledge on the nature of protective immunity elicited by CEO vaccines is very limited. Therefore, elucidating the origin of the immune responses elicited by CEO vaccination is relevant for development of safer control strategies. In this study the transcription levels of key host immune genes (IFN-γ, IFN-ß, IL-1ß, IL-6, IL-8, IL-18) and viral genes (ICP4, ICP27, UL46, UL49), as well as viral genome loads in trachea were quantified at 6 and 12 hours post-challenge of CEO vaccinated and non-vaccinated chickens. Immediately after challenge a significant increase in IFN-γ gene expression was followed by a significant reduction in viral replication. In contrast to the rapid induction of IFN-γ, expression of the pro-inflammatory cytokines (IL-1ß, IL-6, IL-8) and type I IFN ß was either slightly reduced or remained at basal levels. These suggest that the former cytokines may not play important roles during immediate early responses induced by ILTV challenge in either vaccinated or non-vaccinated chickens. Overall, these results suggest that the rapid expression of IFN-γ may induce pathways of antiviral responses necessary for blocking early virus replication.


Assuntos
Anticorpos Antivirais/imunologia , Galinhas/imunologia , Citocinas/metabolismo , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/imunologia , Animais , Galinhas/virologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/genética , Interferon gama/metabolismo , Interleucina-6/metabolismo , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Traqueia/imunologia , Traqueia/virologia , Vacinas Atenuadas , Carga Viral/veterinária
7.
Biologicals ; 44(1): 24-32, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26586283

RESUMO

Traditionally, substrates for production of viral poultry vaccines have been embryonated eggs or adherent primary cell cultures. The difficulties and cost involved in scaling up these substrates in cases of increased demand have been a limitation for vaccine production. Here, we assess the ability of a newly developed chicken-induced pluripotent cell line, BA3, to support replication and growth of Newcastle disease virus (NDV) LaSota vaccine strain. The characteristics and growth profile of the cells were also investigated. BA3 cells could grow in suspension in different media to a high density of up to 7.0 × 10(6) cells/mL and showed rapid proliferation with doubling time of 21 h. Upon infection, a high virus titer of 1.02 × 10(8) EID50/mL was obtained at 24 h post infection using a multiplicity of infection (MOI) of 5. In addition, the cell line was shown to be free of endogenous and exogenous Avian Leukosis viruses, Reticuloendotheliosis virus, Fowl Adenovirus, Marek's disease virus, and several Mycoplasma species. In conclusion, BA3 cell line is potentially an excellent candidate for vaccine production due to its highly desirable industrially friendly characteristics of growing to high cell density and capability of growth in serum free medium.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Newcastle/prevenção & controle , Vírus da Doença de Newcastle , Vacinas Virais/biossíntese , Animais , Linhagem Celular , Embrião de Galinha , Galinhas , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/virologia
8.
Avian Dis ; 59(3): 394-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26478158

RESUMO

Infectious laryngotracheitis (ILT) is a highly contagious disease of chickens and is responsible for significant economic losses in the poultry industry worldwide; it is caused by Gallid herpesvirus-1 (GaHV-1), commonly known as infectious laryngotracheitis virus (ILTV). Experimental evaluation of ILTV strains is fundamental to identify changes in virulence that can contribute to the severity and spread of outbreaks and consequently influence the efficacy of vaccination. Several criteria had been utilized to determine the degree of virulence associated with ILTV strains. The objectives of this study were to compare the levels of virulence of the standard United States Department of Agriculture (USDA) challenge strain with a contemporary outbreak-related strain (63140) and to evaluate the efficacy of individual criteria to identify changes in virulence. Broilers were inoculated with increasing infectious doses of each strain. The criteria utilized to evaluate virulence were clinical signs of the disease, mortality, microscopic tracheal lesions, trachea genome viral loads, and antibody titers. Clinical signs scores were a useful parameter to define the peak of clinical disease but did not reveal differences in virulence between strains. Similarly, trachea microscopic lesion scores or levels of serum antibody titers were parameters that did not reveal obvious differences in virulence between strains. However, mortalities and increased viral genome loads in trachea of chickens inoculated with lower (log10 1 to 2) infectious doses clearly differentiated 63140 as a more-virulent ILTV strain. This study provides the framework to compare the virulence level of emerging ILTV isolates to the now-characterized USDA and 63140 strains.


Assuntos
Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/classificação , Doenças das Aves Domésticas/virologia , Traqueia/patologia , Animais , Galinhas , Genoma Viral , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/genética , Doenças das Aves Domésticas/patologia , Traqueia/virologia , Carga Viral
9.
Clin Pract ; 14(3): 1100-1109, 2024 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-38921265

RESUMO

Acute kidney injury (AKI) is a highly prevalent and a critical complication of cardiac surgery (CS). Serum lactate (sLac) levels have consistently shown an association with morbimortality after CS. We performed a cross-sectional study including 264 adult patients that had a cardiac surgery between January and December 2020. Logistic regression analysis was performed to determine factors associated with AKI development. We measured the postoperative levels of sLac for all participants immediately after CS (T0) and at 4 h (T4) after the surgical intervention. A linear regression model was used to identify the factors influencing both sLac metrics. We identified four risk predictors of AKI; one was preoperative (atrial fibrillation), one intraoperative (cardiopulmonary bypass time), and two were postoperative (length of hospital stay and postoperative sLac). T0 and T4 sLac levels were higher among CS-AKI patients than in Non-CS-AKI patients. Postoperative sLac levels were significant independent predictors of CSA-AKI, and sLac levels are influenced by length of hospital stay, the number of transfused packed red blood cells, and the use of furosemide in CS-AKI patients. These findings may facilitate the earlier identification of patients susceptible to AKI after CS.

10.
Avian Dis ; 57(2 Suppl): 523-31, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23901771

RESUMO

Conventional live attenuated vaccines have been used as the main tool worldwide for the control of infectious laryngotracheitis. However, their suboptimal attenuation combined with poor mass administration practices allowed chicken embryo origin vaccine-derived isolates to circulate in the field, regain virulence, and be the cause of continuous outbreaks of the disease. Previous studies indicated that stable attenuation of infectious laryngotracheitis virus (ILTV) can be achieved by the deletion of individual viral genes that are not essential for viral replication in vitro. One of these genes is the glycoprotein J (gJ) gene. Its deletion provided significant attenuation to virulent ILTV strains from Europe and the United States. The objective of this study was to construct an attenuated gJ-deleted ILTV strain and evaluate its safety and efficacy for in ovo (IO) administration of commercial broilers. A novel gJ-deleted virus (N(delta)gJ) was constructed, and a 10(3) median tissue culture infective dose administered at 18 days of embryo age was considered safe because it did not affect hatchability or survivability of chickens during the first week posthatch. Broilers vaccinated IO and IO + eye drop at 14 days of age presented a significant reduction in clinical signs and reduction of virus loads after challenge, as compared with the nonvaccinated challenged group of chickens. Therefore, this study presents initial proof that the N(delta)gJ strain is a potential ILTV live-attenuated vaccine candidate suitable for IO vaccination of commercial broilers.


Assuntos
Galinhas , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/imunologia , Animais , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Óvulo/virologia , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/virologia , Organismos Livres de Patógenos Específicos , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/efeitos adversos , Vacinas Virais/genética , Virulência
11.
J Clin Med ; 12(12)2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37373759

RESUMO

The aim of this study was to analyze the risk factors and predictors of mortality in a retrospective cohort of patients with coronavirus disease (COVID-19) who presented central nervous system (CNS) manifestations and complications when admitted to hospital. Patients hospitalized from 2020 to 2022 were selected. Demographic variables; history of neurological, cardiological and pulmonary manifestations; comorbidities; prognostic severity scales; and laboratory tests were included. Univariate and adjusted analyses were performed to determine risk factors and predictors of mortality. A forest plot diagram was used to show the strength of the associated risk factors. The cohort included 991 patients; at admission, 463 patients presented CNS damage and of these, 96 hospitalized patients presented de novo CNS manifestations and complications. We estimate a general mortality of 43.7% (433/991) and 77.1% (74/96), for hospitalized patients with de novo CNS manifestations and complications, respectively. The following were identified as risks for the development of hospital CNS manifestations and complications when in hospital: an age of ≥64 years, a history of neurological disease, de novo deep vein thrombosis, D-dimer ≥ 1000 ng/dL, a SOFA ≥ 5, and a CORADS 6. In a multivariable analysis, the mortality predictors were an age of ≥64 years, a SOFA ≥ 5, D-dimer ≥ 1000 ng/mL and hospital CNS manifestations and complications when admitted to hospital. Old age, being hospitalized in critical condition, and having CNS manifestations and complications in hospital are predictors of mortality in hospitalized patients with COVID-19.

12.
Avian Pathol ; 41(1): 21-31, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22845318

RESUMO

Viral vector vaccines using fowl poxvirus (FPV) and herpesvirus of turkey (HVT) as vectors and carrying infectious laryngotracheitis virus (ILTV) genes are commercially available to the poultry industry in the USA. Different sectors of the broiler industry have used these vaccines in ovo or subcutaneously, achieving variable results. The objective of the present study was to determine the efficacy of protection induced by viral vector vaccines as compared with live-attenuated ILTV vaccines. The HVT-LT vaccine was more effective than the FPV-LT vaccine in mitigating the disease and reducing levels of challenge virus when applied in ovo or subcutaneously, particularly when the challenge was performed at 57 days rather than 35 days of age. While the FPV-LT vaccine mitigated clinical signs more effectively when administered subcutaneously than in ovo, it did not reduce the concentration of challenge virus in the trachea by either application route. Detection of antibodies against ILTV glycoproteins expressed by the viral vectors was a useful criterion to assess the immunogenicity of the vectors. The presence of glycoprotein I antibodies detected pre-challenge and post challenge in chickens vaccinated with HVT-LT indicated that the vaccine induced a robust antibody response, which was paralleled by significant reduction of clinical signs. The chicken embryo origin vaccine provided optimal protection by significantly mitigating the disease and reducing the challenge virus in chickens vaccinated via eye drop. The viral vector vaccines, applied in ovo and subcutaneously, provided partial protection, reducing to some degree clinical signs, and challenge VIRUS replication in the trachea.


Assuntos
Galinhas , Sistemas de Liberação de Medicamentos/veterinária , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/imunologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinas Atenuadas/uso terapêutico , Vacinas Virais/uso terapêutico , Animais , Técnica Indireta de Fluorescência para Anticorpo/veterinária , Vírus da Varíola das Aves Domésticas/genética , Vetores Genéticos/genética , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Meleagrídeo 1/genética , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Vacinas Atenuadas/administração & dosagem , Vacinas Virais/administração & dosagem
13.
Avian Dis ; 56(1): 7-14, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22545523

RESUMO

Myeloblastosis-associated virus type 1 (MAV-1) is an exogenous avian retrovirus with oncogenic potential. MAV-1 was detected in young chicks hatching from eggs produced by an experimental genetic line of egg-type chickens. Transmissibility of MAV-1 had not been documented previously. This investigation was intended to partially characterize the virus involved and to study its transmissibility and oncogenicity in naturally and contact-infected chickens. Commercially produced white and brown layer pullets free of exogenous avian leukosis viruses were commingled at hatch with naturally MAV-1-infected chickens. The original MAV-1-infected chickens were discarded after approximately 8 wk, and the contact-exposed chickens were maintained in isolation for 36 wk. Young specific-pathogen-free (SPF) single comb white leghorn chickens were added to the group to study possible horizontal transmission of MAV-1 in young chickens. Upon weekly virus isolation attempts, MAV-1 was readily isolated from the contact-exposed white layers but not from the brown layers between 36 and 53 wk of age (18 wk in total). Three-week-old SPF chickens were readily infected with MAV-1 by contact as early as 1 wk postexposure. Throughout 22 hatches derived from the white and brown MAV-1-contact-exposed layers (between 36 and 53 wk of age), MAV-1 was frequently detected in the white layer progeny, whereas the virus was seldom isolated from the progeny produced by the brown layers during the same 18-wk period. MAV-1 induced a persistent infection in some of the SPF chickens that were exposed by contact at 3 wk of age. Gross tumors were not detected in any of the originally infected experimental chickens at 8 wk of age, in the contact-exposed brown or white layers at the termination of the study at 53 wks of age, or in the contact-exposed SPF chickens at the end of the study at 12 wk of age. Exogenous avian leukosis-related viruses may still be detected in egg-type chickens, emphasizing the importance of thorough screening before incorporation of experimental genetic material into commercial genetic lines of egg-type chickens.


Assuntos
Leucose Aviária/transmissão , Vírus da Mieloblastose Aviária/classificação , Vírus da Mieloblastose Aviária/genética , Galinhas , Doenças das Aves Domésticas/transmissão , Animais , Anticorpos Antivirais/sangue , Leucose Aviária/patologia , Vírus da Mieloblastose Aviária/isolamento & purificação , DNA Viral/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Masculino , Ovalbumina/imunologia , Filogenia , Doenças das Aves Domésticas/patologia , Análise de Sequência de DNA , Organismos Livres de Patógenos Específicos , Viremia/sangue
14.
Avian Dis ; 56(2): 406-10, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22856202

RESUMO

Infectious laryngotracheitis is a highly contagious respiratory disease of chickens controlled by biosecurity and vaccination with live attenuated or recombinant vaccines. Infectious laryngotracheitis virus (ILTV) infections are characterized by a peak of viral replication in the trachea followed by a steady decrease in replication that results in the establishment of latency. Estimation of viral load is an important tool to determine the stage of ILTV infection. Here, a multiplex real-time PCR was optimized for the quantification of ILTV genomes. Quantification of viral genomes was based on the amplification of the ILTV UL44 gene, and sample variability was normalized using the chicken (Gallusgallus domesticus) alpha2-collagen gene as an endogenous control in a duplex reaction.


Assuntos
Galinhas , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/isolamento & purificação , Doenças das Aves Domésticas/diagnóstico , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , Colágeno/metabolismo , Genoma Viral , Infecções por Herpesviridae/diagnóstico , Herpesvirus Galináceo 1/genética , Herpesvirus Galináceo 1/fisiologia , Sensibilidade e Especificidade , Análise de Sequência de DNA , Traqueia/virologia
15.
Avian Dis ; 55(3): 516-21, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22017059

RESUMO

Peracute onset of disease was reported in a 42-wk-old broiler breeder flock that was presented by error with feed containing monensin at approximately seven times the approved level for broiler chickens. Morbidity and mortality were extremely high, and the affected chickens displayed feed refusal, decreased water consumption, and severe paralysis that ranged from abnormal gait to a complete inability to move. During the first 10 days postingestion of the suspect feed, mortality in hens reached 13.7% and 70.9% in the roosters. Hen day production decreased from 67% to 3% in the same period of time. A total of 638 g/ton of monensin was detected in suspect feed samples by one laboratory and 740 g/ton in a second laboratory. Twenty-one days after removal of the suspect feed, the mortality rate returned to normal levels in both hens and roosters, albeit feed consumption and egg production remained extremely low, which prompted the company involved to eliminate the flock.


Assuntos
Ração Animal/microbiologia , Galinhas , Monensin/toxicidade , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/epidemiologia , Animais , Desidratação/diagnóstico , Desidratação/epidemiologia , Desidratação/microbiologia , Desidratação/patologia , Feminino , Masculino , Músculo Esquelético/patologia , Miocárdio/patologia , Oviposição , Paralisia/diagnóstico , Paralisia/epidemiologia , Paralisia/microbiologia , Paralisia/patologia , Doenças das Aves Domésticas/microbiologia , Doenças das Aves Domésticas/patologia , Streptomyces/patogenicidade , Estados Unidos/epidemiologia
16.
Avian Dis ; 54(4): 1210-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21313841

RESUMO

Two types of live attenuated vaccines have been used worldwide for the control of infectious laryngotracheitis virus (ILTV): 1) chicken embryo origin (CEO) vaccines; and 2) tissue culture origin vaccines (TCO). However, the disease persists in spite of extensive use of vaccination, particularly in areas of intense broiler production. Among the factors that may influence the efficiency of ILTV live attenuated vaccines is a possible interference of Newcastle Disease virus (NDV) and infectious bronchitis virus (IBV) vaccines with the protection induced by ILTV vaccines. The protection induced by CEO and TCO vaccines was evaluated when administered at 14 days of age alone or in combination with the B1 type strain of NDV (B1) and/or the Arkansas (ARK) and Massachusetts (MASS) serotypes of IBV vaccines. Two weeks after vaccination (28 days of age), the chickens were challenged with a virulent ILTV field strain (63140 isolate, group V genotype). Protection was evaluated at 5 and 7 days postchallenge by scoring clinical signs and quantifying the challenge virus load in the trachea using real-time PCR (qPCR). In addition, the viral load of the vaccine viruses (ILTV, NDV, and IBV) was quantified 3 and 5 days postvaccination also using qPCR. The results of this study indicate that the NDV (B1) and IBV (ARK) vaccines and a multivalent vaccine constituted by NDV (B1) and IBV (ARK and MASS) did not interfere with the protection induced by the CEO ILTV vaccine. However, the NDV (BI) and the multivalent (B1/MASS/ARK) vaccines interfered with the protection induced by the TCO vaccine (P < 0.05). Either in combination or by themselves, the NDV and IBV vaccines decreased the tracheal replication of the TCO vaccine and the protection induced by this vaccine, since the ILTV-vaccinated and -challenged chickens displayed significantly more severe clinical signs and ILTV load (P < 0.05) than chickens vaccinated with the TCO vaccine alone. Although NDV and IBV challenges were not performed, the antibody responses elicited by NDV and/or the IBV vaccinations were significantly reduced (P < 0.05) when applied in combination with the CEO vaccine.


Assuntos
Herpesvirus Galináceo 1/imunologia , Vírus da Bronquite Infecciosa/imunologia , Vírus da Doença de Newcastle/imunologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/administração & dosagem , Viroses/veterinária , Animais , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/veterinária , Interações Medicamentosas , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/veterinária , Doença de Newcastle/prevenção & controle , Doenças das Aves Domésticas/virologia , Vacinas Virais/imunologia , Viroses/prevenção & controle
17.
Avian Dis ; 54(4): 1319-22, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21313858

RESUMO

Formalin-fixed suspect tumors were submitted to the Poultry Diagnostic and Research Center at the University of Georgia (Athens, GA) for diagnosis. Samples were from fancy breed chickens with a history of increased tumor prevalence in both hens and roosters. Microscopically, in all the samples, there were neoplastic proliferations of spindle-shaped cells. The matrix surrounding tumor cells stained positively with Alcian blue at pH 2.5, but neoplastic cells did not stain with periodic acid-Schiff. Immunohistochemistry stains were positive for vimentin and neuron-specific enolase and negative for desmin, smooth muscle actin, and S-100 protein. Tumors were determined to be myxosarcomas. All samples were positive for PCR targeting the gp85 avian leukosis virus (ALV) envelope protein. However, analysis of the predicted amino acid sequences in the envelope gene from three separate samples showed high similarity between them and to ALV subgroup A.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas , Mixossarcoma/veterinária , Neoplasias Cutâneas/veterinária , Substituição de Aminoácidos , Animais , Feminino , Imuno-Histoquímica , Masculino , Mixossarcoma/patologia , Mixossarcoma/virologia , Filogenia , Reação em Cadeia da Polimerase/veterinária , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
18.
Avian Dis ; 54(3): 981-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20945777

RESUMO

Avian leukosis virus (ALV) is known to cause several neoplastic conditions in chickens, such as B-cell lymphomas, myelocytomas, erythroblastosis, and other types of neoplasia including osteopetrosis. We describe herein the identification of unique ALV-related proviral DNA sequences in an archived chicken bone affected with osteopetrosis. The osteopetrotic bone was obtained from an affected 46-week-old brown layer during an outbreak of osteopetrosis in Costa Rica in 1986. Analysis of proviral DNA in the 23-year-old osteopetrotic bone revealed unique exogenous ALV-related sequences that were named CR-1986 (Costa Rica, 1986). The 5' and 3' long terminal repeats (LTR) in the proviral DNA were identical to each other. The U3 regions in the LTRs were most similar to equivalent sequences in ALV-J, while U5 was identical to known endogenous ALV-E sequences. The predicted CR-1986 envelope protein was most similar to the envelope of myeloblastosis associated virus type 1 (MAV-1), although the percentage of amino acid sequence similarity to MAV-1 was low (90.4%). The variable and hypervariable regions of gp85 displayed several mutations compared to representative strains of ALV. The gp37 (transmembrane or TM) envelope protein showed three leucine to serine mutations that may represent important changes in the conformation of this protein, a finding that is currently being investigated. Several recombination events may have contributed to the emergence of CR-1986 because each analyzed segment was similar to a different ALV. CR-1986 may represent a unique ALV based on distinctive characteristics of its predicted envelope protein in comparison to previously reported ALVs.


Assuntos
Vírus da Leucose Aviária/genética , Surtos de Doenças/veterinária , Osteopetrose/veterinária , Doenças das Aves Domésticas/virologia , Animais , Sequência de Bases , Osso e Ossos/virologia , Costa Rica/epidemiologia , DNA Viral/genética , Feminino , Regulação Viral da Expressão Gênica/fisiologia , Genoma Viral , Dados de Sequência Molecular , Osteopetrose/epidemiologia , Osteopetrose/virologia , Proteínas Virais/química , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
Avian Dis ; 54(4): 1251-9, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21313847

RESUMO

Infectious laryngotracheitis (ILT) is a highly contagious respiratory disease of chickens caused by infectious laryngotracheitis virus (ILTV). The disease is mainly controlled through biosecurity and by vaccination with live-attenuated vaccines. The chicken embryo origin (CEO) vaccines, although proven to be effective in experimental settings, have limited efficacy in controlling the disease in dense broiler production sites due to unrestricted use and poor mass vaccination coverage. These factors allowed CEO vaccines to regain virulence, causing long lasting and, consequently, severe outbreaks of the disease. A new generation of viral vector fowl poxvirus (FPV) and herpesvirus of turkey (HVT) vaccines carrying ILTV genes has been developed and such vaccines are commercially available. These vaccines are characterized by their lack of transmission, lack of ILTV-associated latent infections, and no reversion to virulence. HVT-vectored ILTV recombinant vaccines were originally approved for subcutaneous HVT or transcutaneous (pox) delivery. The increased incidence of ILTV outbreaks in broiler production sites encouraged the broiler industry to deliver the FPV-LT and HVT-LT recombinant vaccines in ovo. The objective of this study was to evaluate the protection induced by ILTV viral vector recombinant vaccines after in ovo application in 18-day-old commercial broiler embryos. The protection induced by recombinant ILTV vaccines was assessed by their ability to prevent clinical signs and mortality; to reduce challenge virus replication in the trachea; to prevent an increase in body temperature; and to prevent a decrease in body weight gain after challenge. In this study, both recombinant-vectored ILTV vaccines provided partial protection, thereby mitigating the disease, but did not reduce challenge virus loads in the trachea.


Assuntos
Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/imunologia , Vacinas Virais/administração & dosagem , Animais , Anticorpos Antivirais/sangue , Galinhas , Vírus da Varíola das Aves Domésticas , Variação Genética , Infecções por Herpesviridae/sangue , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Meleagrídeo 1 , Óvulo , Reação em Cadeia da Polimerase , Fatores de Tempo , Traqueia/virologia , Vacinas Atenuadas/imunologia , Vacinas Sintéticas/imunologia , Carga Viral , Vacinas Virais/imunologia
20.
Avian Pathol ; 38(1): 47-53, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19156579

RESUMO

In a recent study, several US infectious laryngotracheitis virus (ILTV) strains and field isolates were genotyped by polymerase chain reaction and restriction fragment length polymorphism (PCR-RFLP) into nine different genotypes. All of the commercial poultry isolates were identified within genotypes IV, V, and VI. Based on the PCR-RFLP, Group IV isolates were characterized as genetically identical to the chicken embryo origin (CEO) vaccines, Group V as genetically closely related to the CEO vaccines, and Group VI as genetically different to the vaccine strains. The objective of this study was to determine the pathogenicity and growth characteristics of six ILTV commercial poultry isolates as compared with the CEO vaccine. Two isolates representative of PCR-RFLP Groups IV, V, and VI were selected. Differences in disease severity, viral tissue distribution in chickens, and plaque formation ability in cell culture were observed among viral genotypes IV, V, and VI, and between V-A and V-B isolates. Mild respiratory clinical signs were produced by IV-A, IV-B and the CEO vaccine, while VI-A and VI-B isolates produced severe respiratory signs and severe depression, and during the peak of clinical signs both isolates were re-isolated from the conjunctiva, sinus, trachea and thymus. Similarly to Group VI isolates, V-A and V-B produced severe respiratory signs, depression, and were re-isolated from conjunctiva, sinus, and trachea; on cell culture, both isolates produced significant larger plaques than any of the other isolates analysed. Overall, differences in pathogenicity and growth characteristics were observed among genetically closely related US ILTV isolates; however, complete genomes will be necessary to identify molecular determinants linked to the pathogenic viral phenotypes.


Assuntos
Galinhas , Infecções por Herpesviridae/veterinária , Herpesvirus Galináceo 1/crescimento & desenvolvimento , Herpesvirus Galináceo 1/patogenicidade , Doenças das Aves Domésticas/virologia , Animais , Linhagem Celular , Infecções por Herpesviridae/epidemiologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Infecções por Herpesviridae/virologia , Herpesvirus Galináceo 1/classificação , Herpesvirus Galináceo 1/imunologia , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/patologia , Organismos Livres de Patógenos Específicos , Estados Unidos/epidemiologia , Ensaio de Placa Viral , Vacinas Virais/imunologia , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa