Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Viruses ; 15(11)2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-38005939

RESUMO

Marek's disease (MD) is a highly infectious lymphoproliferative disease in chickens with a significant economic impact. Mardivirus gallidalpha 2, also known as Marek's disease virus (MDV), is the causative pathogen and has been categorized based on its virulence rank into four pathotypes: mild (m), virulent (v), very virulent (vv), and very virulent plus (vv+). A prior comparative genomics study suggested that several single-nucleotide polymorphisms (SNPs) and genes in the MDV genome are associated with virulence, including nonsynonymous (ns) SNPs in eight open reading frames (ORF): UL22, UL36, UL37, UL41, UL43, R-LORF8, R-LORF7, and ICP4. To validate the contribution of these nsSNPs to virulence, the vv+MDV strain 686 genome was modified by replacing nucleotides with those observed in the vMDV strains. Pathogenicity studies indicated that these substitutions reduced the MD incidence and increased the survival of challenged birds. Furthermore, using the best-fit pathotyping method to rank the virulence, the modified vv+MDV 686 viruses resulted in a pathotype similar to the vvMDV Md5 strain. Thus, these results support our hypothesis that SNPs in one or more of these ORFs are associated with virulence but, as a group, are not sufficient to result in a vMDV pathotype, suggesting that there are additional variants in the MDV genome associated with virulence, which is not surprising given this complex phenotype and our previous finding of additional variants and SNPs associated with virulence.


Assuntos
Herpesvirus Galináceo 2 , Mardivirus , Doença de Marek , Animais , Virulência/genética , Galinhas , Herpesvirus Galináceo 2/genética , Mardivirus/genética
2.
J Zoo Wildl Med ; 54(3): 473-483, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37817613

RESUMO

Acquiring baseline physiologic data for animals from a free-ranging wildlife species is an elusive objective. Between 1990 and 2020, a monitoring program on the last population of ocelot (Leopardus pardalis) to inhabit public land in the United States yielded 139 blood samples from 67 individual animals. Ocelots were live trapped and anesthetized for census and radiotelemetric studies. The protocol included morphometrics, photographs, electronic identification, and blood collection. Complete blood count and serum chemistry were performed, and after sorting of the data to remove unhealthy individuals and occasional outliers, the dataset provided sufficient information to compute reliable reference intervals (RI). According to the American Society of Veterinary Clinical Pathology consensus guidelines, RI should be elaborated by using data from each reference individual only once. RI by random selection was determined when several measurements were available over time from one same animal. Second, RI were also computed allowing repeat measurements for reference individuals, exclusively to characterize and quantify the effect on the data distribution and on the generated RI. A summary of published RI for various species of wild felids is also presented. The variations observed between species is due not only to species differences but also to variation in measurement methods and RI study design. Overall, accurate blood work interpretation requires RI generated from a healthy population, with defined measurement methods and state-of-the-art RI study design. Of note, calcium is typically tightly regulated in all mammals, as illustrated by the narrow RI (8.5-10.8 mg/dl); conversely, finding a narrow RI in calcium across as many as 49 healthy individuals suggests a high-quality design study.


Assuntos
Felidae , Hematologia , Animais , Cálcio , Testes Hematológicos/veterinária , Valores de Referência
3.
Avian Dis ; 67(1): 102-107, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-37140118

RESUMO

The U.S. Department of Agriculture Avian Disease and Oncology Laboratory currently relies on live birds of specific genetic backgrounds for producing chicken-embryo fibroblasts that are used for the diagnosis and subtyping of field isolates associated with avian leukosis virus (ALV) outbreaks. As an alternative to maintaining live animals for this purpose, we are currently developing cell lines capable of achieving the same result by ablation of the entry receptors utilized by ALV strains. We used CRISPR-Cas9 on the cell fibroblast-derived cell line DF-1 to disrupt the tva gene, which encodes the receptor required for binding and entry of ALV-A into cells. We ultimately identified seven DF-1 clones that had biallelic and homozygous indels at the Cas9 target site, exon 2 of tva. When tested in vitro for their ability to host ALV-A, the five clones that had frameshift mutations that disrupted the Tva protein were unable to support ALV-A replication. This result clearly demonstrates that modified cell lines can be used as part of a battery of tests to determine ALV subtype for isolate characterization, thus eliminating the need for live birds.


Nota de investigación- La ablación dirigida del exón 2 del gene del receptor del virus de la leucosis aviar A (ALV-A) en una línea celular de fibroblastos de pollo mediante CRISPR anula la infección por ALV-A. El Laboratorio de Oncología y Enfermedades Aviares del Departamento de Agricultura de los Estados Unidos. actualmente depende de aves vivas con antecedentes genéticos específicos para producir fibroblastos de embrión de pollo que se utilizan para el diagnóstico y la subtipificación de aislamientos de campo asociados con brotes del virus de la leucosis aviar (ALV). Como alternativa al mantenimiento de animales vivos para este propósito, actualmente se están desarrollando líneas celulares capaces de lograr el mismo resultado mediante la ablación de los receptores de entrada utilizados por las cepas ALV. Se utilizó el método repeticiones palindrómicas cortas agrupadas y regularmente interespaciadas o CRISPR-Cas9 en la línea celular DF-1 derivada de fibroblastos para interrumpir el gene Tva, que codifica el receptor requerido para la unión y entrada de ALV-A en las células. Finalmente, se identificaron siete clones de DF-1 que tenían inserciones y deleciones (indeles) bialélicos y homocigóticos en el sitio blanco Cas9, exón 2 del gene tva. Cuando se probó in vitro su capacidad para albergar ALV-A, los cinco clones que tenían mutaciones que involucraban al marco de lectura y que interrumpieron la proteína Tva no pudieron admitir la replicación de ALVA. Este resultado demuestra claramente que las líneas celulares modificadas se pueden utilizar como parte de una batería de pruebas para determinar el subtipo de ALV para la caracterización de los aislamientos, eliminando así la necesidad de aves vivas.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Doenças das Aves Domésticas , Animais , Galinhas , Vírus da Leucose Aviária/genética , Linhagem Celular , Éxons , Fibroblastos
4.
PLoS One ; 17(8): e0272557, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35939448

RESUMO

Avian lymphoid leukosis-like (LL-like) lymphoma has been observed in some experimental and commercial lines of chickens that are free of exogenous avian leukosis virus. Reported cases of avian lymphoid leukosis-like lymphoma incidences in the susceptible chickens are relatively low, but the apathogenic subgroup E avian leukosis virus (ALV-E) and the Marek's disease vaccine, SB-1, significantly escalate the disease incidence in the susceptible chickens. However, the underlying mechanism of tumorigenesis is poorly understood. In this study, we bioinformatically analyzed the deep RNA sequences of 6 lymphoid leukosis-like lymphoma samples, collected from susceptible chickens post both ALV-E and SB-1 inoculation, and identified a total of 1,692 novel long non-coding RNAs (lncRNAs). Thirty-nine of those novel lncRNAs were detected with altered expression in the LL-like tumors. In addition, 13 lncRNAs whose neighboring genes also showed differentially expression and 2 conserved novel lncRNAs, XLOC_001407 and XLOC_022595, may have previously un-appreciated roles in tumor development in human. Furthermore, 14 lncRNAs, especially XLOC_004542, exhibited strong potential as competing endogenous RNAs via sponging miRNAs. The analysis also showed that ALV subgroup E viral gene Gag/Gag-pol and the MD vaccine SB-1 viral gene R-LORF1 and ORF413 were particularly detectable in the LL-like tumor samples. In addition, we discovered 982 novel lncRNAs that were absent in the current annotation of chicken genome and 39 of them were aberrantly expressed in the tumors. This is the first time that lncRNA signature is identified in avian lymphoid leukosis-like lymphoma and suggests the epigenetic factor, lncRNA, is involved with the avian lymphoid leukosis-like lymphoma formation and development in susceptible chickens. Further studies to elucidate the genetic and epigenetic mechanisms underlying the avian lymphoid leukosis-like lymphoma is indeed warranted.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Linfoma , Neoplasias , Doenças das Aves Domésticas , RNA Longo não Codificante , Animais , Leucose Aviária/genética , Vírus da Leucose Aviária/genética , Galinhas/genética , Suscetibilidade a Doenças , Humanos , Linfoma/genética , Linfoma/veterinária , RNA Longo não Codificante/genética , Transcriptoma
5.
Avian Dis ; 65(2): 241-249, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34412454

RESUMO

Marek's disease virus (MDV) is an important poultry pathogen that is controlled through widespread vaccination with avirulent and attenuated strains. However, continued evolution of field viruses to higher virulence has required ongoing improvement of available vaccine strains, and these vaccine strains offer an attractive platform for designing recombinant vector vaccines with cross-protection against MDV and additional pathogens. Recent reports of failures in vaccine licensing trials of positive controls to reach appropriately high levels of Marek's disease incidence prompted us to evaluate possible combinations of outbred specific-pathogen-free layer lines and alternative virulent challenge strains that could provide more consistent models for serotype 3 vectored vaccine development. Choice of layer line and virulent MDV challenge strain each contributed to the ability of a challenge model to reach 80% virulence in unvaccinated positive control groups in the majority of trials, without overwhelming serotype 3 vectored vaccine protection in vaccinated groups. Conversely, reducing challenge virus dose by a factor of four, or vaccine dose by half, had no consistent effect across these models. Although MDV strain 617A had the most potential as an alternative to strains that are currently approved for licensing trials, no combination of layer line and challenge virus consistently met the goals for a successful challenge model in all study replicates, indicating that high variability is an inherent difficulty in MDV challenge studies, at least when outbred birds are used.


Artículo regular­Comparación de las cepas de desafío del virus de la enfermedad de Marek y los tipos de aves para la obtención de licencias de vacunas. El virus de la enfermedad de Marek (MDV) es un patógeno importante en la avicultura que se controla mediante la vacunación generalizada con cepas avirulentas y atenuadas. Sin embargo, la evolución continua de los virus de campo hacia una mayor virulencia ha requerido una mejora continua de las cepas vacunales disponibles y estas cepas vacunales ofrecen una plataforma atractiva para diseñar vacunas con vectores recombinantes que induzcan protección cruzada contra el virus de la enfermedad de Marek y patógenos adicionales. Los reportes recientes de fallas en los controles positivos para alcanzar niveles apropiadamente altos de incidencia de la enfermedad de Marek en los ensayos para obtener la licencia de vacunas llevaron a evaluar posibles combinaciones de líneas de postura híbridas libres de patógenos específicos y cepas de desafío virulentas alternativas que podrían proporcionar modelos más consistentes para el desarrollo de vacunas con vectores de serotipo 3. Tanto la elección de la línea de postura como de la cepa de desafío virulenta de Marek contribuyeron a obtener un modelo de desafío con capacidad para alcanzar el 80% de virulencia en grupos controles positivo no vacunados en la mayoría de los ensayos, sin una protección abrumadora de la vacuna con vector de serotipo 3 en los grupos vacunados. Por el contrario, la reducción de la dosis del virus de desafío en un factor de cuatro, o la dosis de vacuna a la mitad, no tuvieron un efecto constante en estos modelos. Aunque la cepa 617A de Marek mostró el mayor potencial como alternativa a las cepas que actualmente están aprobadas para ensayos de licenciar vacunas, ninguna combinación de línea de postura y virus de desafío cumplió consistentemente los objetivos de un modelo de desafío exitoso en todas las réplicas del estudio, lo que indica que la alta variabilidad es una dificultad inherente en los estudios de desafío para la enfermedad de Marek, al menos cuando se utilizan aves híbridas.


Assuntos
Galinhas/classificação , Herpesvirus Galináceo 3/classificação , Herpesvirus Galináceo 3/imunologia , Vacinas Virais/classificação , Animais , Galinhas/imunologia , Herpesvirus Galináceo 3/patogenicidade , Complexo Principal de Histocompatibilidade/genética , Organismos Livres de Patógenos Específicos , Vacinas Virais/normas , Virulência
6.
Microorganisms ; 10(1)2021 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-35056456

RESUMO

Marek's disease (MD) is a ubiquitous disease of domesticated chickens and its etiologic agent is the Gallid alphaherpesvirus 2 (GaHV-2), also known as Marek's disease virus (MDV). MD is currently controlled by vaccination using live attenuated strains of MDV (e.g., CVI988/Rispens), non-pathogenic serotypes of MDV (GaHV-3), or non-pathogenic strains of the related Melagrid alphaherpesvirus 1 (MeHV-1). One attractive strategy for the production of new vaccine strains is a recombinant MDV attenuated by the deletion of the major viral oncogene meq. However, meq-deleted variants of MDV cause atrophy of the bursa and thymus in maternal antibody-negative chickens, and the resulting immunosuppression makes them unsuitable. Herein we detail our attempt to mitigate the lymphoid atrophy caused by meq-deleted MDV by further attenuation of the virus through ablation of the viral thymidine kinase (tk) gene. We demonstrate that ablation of the viral tk from the meq-deleted virus rMd5B40/Δmeq resulted in a virus attenuated for replication in vitro and which spared chickens from atrophy of the lymphoid organs in vivo. When the rMd5B40/Δmeq/Δtk/GFP was used as a vaccine it was protective against challenge with the vv+MDV strain 686, but the protection was less than that provided by the CVI988/Rispens vaccine.

7.
Avian Dis ; 64(3): 347-351, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-33205183

RESUMO

In two independent submissions, a 3-yr-old, dead Bourbon Red turkey tom from a zoo and a Royal Palm turkey hen from a backyard flock were submitted for necropsy. Both birds had been kept together with chickens. Findings of the necropsy of the first turkey were an enlarged and dark liver with many pale white foci and a few small white nodules, pale and enlarged spleen, prominent thymus, mottled and pale kidneys, and pale and enlarged testes. Findings of the necropsy of the second turkey were a dark and mildly enlarged liver and severely enlarged, firm, and pale kidneys. Histopathology revealed infiltration of most organs of both birds with neoplastic lymphocytes, which were uniform in the first turkey and pleomorphic in the second turkey. Immunohistochemistry with a CD3 marker identified the neoplastic lymphocytes as T cells. Marek's disease virus serotype 1 was detected with PCR in the livers of both birds, whereas PCRs for reticuloendotheliosis virus and lymphoproliferative disease virus were negative. Based on these findings, Marek's disease was diagnosed in both turkeys, which is very rare and were the first definitive cases reported in the United States. It is likely that the chickens were the source of infection.


Assuntos
Doença de Marek/diagnóstico , Doenças das Aves Domésticas/diagnóstico , Perus , Animais , California , Feminino , Herpesvirus Galináceo 2 , Linfócitos/virologia , Masculino , Doença de Marek/virologia , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/virologia , Linfócitos T/virologia
8.
PLoS Biol ; 18(3): e3000619, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32134914

RESUMO

Many livestock and human vaccines are leaky because they block symptoms but do not prevent infection or onward transmission. This leakiness is concerning because it increases vaccination coverage required to prevent disease spread and can promote evolution of increased pathogen virulence. Despite leakiness, vaccination may reduce pathogen load, affecting disease transmission dynamics. However, the impacts on post-transmission disease development and infectiousness in contact individuals are unknown. Here, we use transmission experiments involving Marek disease virus (MDV) in chickens to show that vaccination with a leaky vaccine substantially reduces viral load in both vaccinated individuals and unvaccinated contact individuals they infect. Consequently, contact birds are less likely to develop disease symptoms or die, show less severe symptoms, and shed less infectious virus themselves, when infected by vaccinated birds. These results highlight that even partial vaccination with a leaky vaccine can have unforeseen positive consequences in controlling the spread and symptoms of disease.


Assuntos
Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/transmissão , Vacinas Virais/farmacologia , Animais , Galinhas , Plumas/virologia , Interações Hospedeiro-Patógeno , Doença de Marek/etiologia , Doença de Marek/mortalidade , Doença de Marek/prevenção & controle , Vacinação , Carga Viral , Vacinas Virais/administração & dosagem , Virulência , Eliminação de Partículas Virais
9.
J Virol ; 93(23)2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31554689

RESUMO

In 2010, sporadic cases of avian leukosis virus (ALV)-like bursal lymphoma, also known as spontaneous lymphoid leukosis (LL)-like tumors, were identified in two commercial broiler breeder flocks in the absence of exogenous ALV infection. Two individual ALV subgroup E (ALV-E) field strains, designated AF227 and AF229, were isolated from two different breeder farms. The role of these ALV-E field isolates in development of and the potential joint impact in conjunction with a Marek's disease virus (MDV) vaccine (SB-1) were further characterized in chickens of an experimental line and commercial broiler breeders. The experimental line 0.TVB*S1, commonly known as the rapid feathering-susceptible (RFS) line, of chickens lacks all endogenous ALV and is fully susceptible to all subgroups of ALV, including ALV-E. Spontaneous LL-like tumors occurred following infection with AF227, AF229, and a reference ALV-E strain, RAV60, in RFS chickens. Vaccination with serotype 2 MDV, SB-1, in addition to AF227 or AF229 inoculation, significantly enhanced the spontaneous LL-like tumor incidence in the RFS chickens. The spontaneous LL-like tumor incidence jumped from 14% by AF227 alone to 42 to 43% by AF227 in combination with SB-1 in the RFS chickens under controlled conditions. RNA-sequencing analysis of the LL-like lymphomas and nonmalignant bursa tissues of the RFS line of birds identified hundreds of differentially expressed genes that are reportedly involved in key biological processes and pathways, including signaling and signal transduction pathways. The data from this study suggested that both ALV-E and MDV-2 play an important role in enhancement of the spontaneous LL-like tumors in susceptible chickens. The underlying mechanism may be complex and involved in many chicken genes and pathways, including signal transduction pathways and immune system processes, in addition to reported viral genes.IMPORTANCE Lymphoid leukosis (LL)-like lymphoma is a low-incidence yet costly and poorly understood disease of domestic chickens. The observed unique characteristics of LL-like lymphomas are that the incidence of the disease is chicken line dependent; pathologically, it appeared to mimic avian leukosis but is free of exogenous ALV infection; inoculation of the nonpathogenic ALV-E or MDV-2 (SB-1) boosts the incidence of the disease; and inoculation of both the nonpathogenic ALV-E and SB-1 escalates it to much higher levels. This study was designed to test the impact of two new ALV-E isolates, recently derived from commercial broiler breeder flocks, in combination with the nonpathogenic SB-1 on LL-like lymphoma incidences in both an experimental egg layer line of chickens and a commercial broiler breeder line of chickens under a controlled condition. Data from this study provided an additional piece of experimental evidence on the potency of nonpathogenic ALV-E, MDV-2, and ALV-E plus MDV-2 in boosting the incidence of LL-like lymphomas in susceptible chickens. This study also generated the first piece of genomic evidence that suggests host transcriptomic variation plays an important role in modulating LL-like lymphoma formation.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/complicações , Leucose Aviária/virologia , Coinfecção/virologia , Linfoma/complicações , Linfoma/virologia , Doença de Marek/complicações , Doenças das Aves Domésticas/virologia , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/genética , Galinhas/virologia , Suscetibilidade a Doenças , Regulação Viral da Expressão Gênica , Genótipo , Herpesvirus Galináceo 3 , Incidência , Doença de Marek/virologia , Vacinas contra Doença de Marek , Análise de Sequência de DNA , Transdução de Sinais , Transcriptoma , Vacinação , Vacinas Virais
10.
Avian Pathol ; 47(4): 364-374, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29533078

RESUMO

A variety of techniques have been developed as diagnostic tools for the differential diagnosis of tumours produced by Marek's disease virus from those induced by avian leukosis virus and reticuloendotheliosis virus. However, most current techniques are unreliable when used in formalin-fixed paraffin-embedded (FFPE) tissues, which often is the only sample type available for definitive diagnosis. A collection of tumours was generated by the inoculation of different strains of Marek's disease virus, reticuloendotheliosis virus or avian leukosis virus singularly or in combination. FFPE tissue sections from tumour and non-tumour tissues were analysed by optimized immunohistochemistry (IHC) techniques and traditional as well as quantitative polymerase chain reaction (PCR) with newly designed primers ideal for DNA fragmented by fixation. IHC and PCR results were highly sensitive and specific in tissues from single-infected birds. Virus quantity was higher in tumours compared to non-tumour spleens from Marek's disease (MD) virus-infected birds. Thus, using FFPE sections alone may be sufficient for the diagnosis of MD by demonstration of high quantities of viral antigens or genome in tumour cells, along with the absence of other tumour viruses by traditional PCR, and if standard criteria are met based on clinical history and histology. IHC furthermore allowed detection of the specific cells that were infected with different viruses in tumours from birds that had been inoculated simultaneously with multiple viruses. Following validation with field samples, these new protocols can be applied for both diagnostic and research purposes to help accurately identify avian tumour viruses in routine FFPE tissue sections.


Assuntos
Galinhas/virologia , Imuno-Histoquímica/veterinária , Doença de Marek/virologia , Vírus Oncogênicos/isolamento & purificação , Doenças das Aves Domésticas/virologia , Infecções por Retroviridae/veterinária , Infecções Tumorais por Vírus/virologia , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/isolamento & purificação , Primers do DNA/genética , Diagnóstico Diferencial , Formaldeído , Mardivirus/genética , Mardivirus/isolamento & purificação , Vírus Oncogênicos/genética , Inclusão em Parafina/veterinária , Reação em Cadeia da Polimerase/veterinária , Vírus da Reticuloendoteliose/genética , Vírus da Reticuloendoteliose/isolamento & purificação , Infecções por Retroviridae/virologia
11.
Avian Pathol ; 45(6): 657-666, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27258614

RESUMO

Marek's disease virus (MDV), an alphaherpesvirus, causes Marek's disease (MD), a lymphoproliferative disease in poultry characterized by T-cell lymphomas, nerve lesions, and mortality. Vaccination is used worldwide to control MD, but increasingly virulent field strains can overcome this protection, driving a need to create new vaccines. Previous studies revealed that insertion of reticuloendotheliosis virus (REV) long terminal repeat (LTR) into a bacterial artificial chromosome (BAC) clone of a very virulent strain of MDV, Md5, rendered the resultant recombinant virus, rMd5 REV-LTR BAC, fully attenuated in maternal antibody positive (Mab+) chickens at passage 40. In the current study, the protective efficacy of rMd5 REV-LTR BAC was evaluated. First, passage 70 was identified as being fully attenuated in maternal antibody negative chickens and chosen as the optimal passage level for use in protective efficacy studies. Second, three protective efficacy trials were conducted comparing the rMd5 REV-LTR p70 BAC to the CVI988/Rispens vaccine. Groups of Mab+ and Mab- 15I5 × 71 chickens were vaccinated in ovo at 18 days of embryonation or intra-abdominally at day of hatch, and challenged at 5 days post-hatch with the vv+MDV strain 686. Vaccination at day of hatch and in ovo with rMd5 REV-LTR p70 BAC protected chickens against MDV-induced bursa and thymic atrophy, but did not provide the same level of protection against MD tumours as that afforded by the commercial vaccine, CVI988/Rispens.


Assuntos
Galinhas/imunologia , Herpesvirus Galináceo 2/imunologia , Vacinas contra Doença de Marek/imunologia , Doença de Marek/prevenção & controle , Doenças das Aves Domésticas/prevenção & controle , Vírus da Reticuloendoteliose/genética , Animais , Anticorpos Antivirais/sangue , Células Cultivadas , Galinhas/virologia , Cromossomos Artificiais Bacterianos/genética , DNA Recombinante , Patos , Feminino , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Sequenciamento de Nucleotídeos em Larga Escala/veterinária , Masculino , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Análise de Sequência de DNA/veterinária , Sequências Repetidas Terminais/genética , Vacinação/veterinária
12.
Avian Pathol ; 44(1): 23-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25407937

RESUMO

Breeders of the 2009 generation of Avian Disease and Oncology Laboratory transgenic chicken line ALVA6, known to be resistant to infection with subgroups A and E avian leukosis virus (ALV), were vaccinated at hatch with a trivalent Marek's disease (MD) vaccine containing serotypes 1, 2, and 3 Marek's disease virus (MDV) and were maintained under pathogen-free conditions from the day of hatch until 75 weeks of age. Spontaneous ALV-like bursal lymphomas, also termed lymphoid leukosis (LL)-like lymphomas, were detected in 7% of the ALVA6 breeders. There was no evidence of infection with exogenous and endogenous ALV as determined by virus isolation tests of plasma and tumour tissue homogenates. For the next three generations, serotype 2 MDV was eliminated from the trivalent MD vaccine used. Results show, for the first time, that removal of serotype 2 MDV from MD vaccines eliminated spontaneous LL-like lymphomas within 50 to 72 weeks of age for at least three consecutive generations. Two experiments were also conducted to determine the influence of in ovo vaccination with serotype 2 MD vaccines on enhancement of spontaneous LL-like lymphomas in ALVA6 chickens. Chickens from the 2012 generation were each inoculated in ovo or at hatch with 5000 plaque-forming units of serotype 2 MDV. Results indicate that by 50 weeks of age the incidence of spontaneous LL-like lymphomas in chickens inoculated in ovo with serotype 2 MDV was comparable with that in chickens inoculated with virus at hatch, suggesting that the augmentation effect of serotype 2 MDV is independent of age of vaccination.


Assuntos
Animais Geneticamente Modificados/genética , Bolsa de Fabricius/patologia , Galinhas , Herpesvirus Galináceo 3/patogenicidade , Linfoma/veterinária , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Animais , Vírus da Leucose Aviária/imunologia , Bolsa de Fabricius/virologia , Herpesvirus Galináceo 3/genética , Herpesvirus Galináceo 3/imunologia , Linfoma/patologia , Linfoma/virologia , Reação em Cadeia da Polimerase , Especificidade da Espécie , Vacinas Virais/genética , Vacinas Virais/uso terapêutico
13.
Avian Dis ; 57(4): 785-9, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24597122

RESUMO

A simple PCR method was developed for the detection of Marek's disease (MD) and reticuloendotheliosis (RE) in formalin-fixed paraffin-embedded (FFPE) tissues, and for the detection of MD in tissues only preserved in 10% neutral buffered formalin. MD virus (MDV) and RE virus proviral DNA were detected in FFPE tissues stored for over 20 yr. MDV was also detected in tissues only preserved in formalin for up to 6 mo. The data indicate that PCR of formalin-fixed and FFPE tissues is a simple and valuable tool that can be used to identify MD and RE infection. The method described in this paper is a good alternative to any biologic or immunohistochemical assay to confirm the detection of MD and RE, as it does not require shipping frozen tissues to the diagnostic laboratory.


Assuntos
Galinhas , DNA Viral/genética , Herpesvirus Meleagrídeo 1/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Doenças das Aves Domésticas/diagnóstico , Provírus/genética , Vírus da Reticuloendoteliose/isolamento & purificação , Animais , DNA Viral/metabolismo , Formaldeído/química , Doença de Marek/diagnóstico , Doença de Marek/virologia , Neoplasias/diagnóstico , Neoplasias/veterinária , Parafina/química , Inclusão em Parafina/veterinária , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/virologia , Provírus/metabolismo , Reticuloendoteliose Aviária/diagnóstico , Reticuloendoteliose Aviária/virologia
14.
Avian Pathol ; 41(3): 259-65, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22702453

RESUMO

Co-cultivation of the JM/102W strain of Marek's disease virus (MDV) with reticuloendotheliosis virus (REV) resulted in the generation of a recombinant MDV containing the REV long terminal repeat (LTR) named the RM1 strain of MDV, a strain that was highly attenuated for oncogenicity but induced severe bursal and thymic atrophy. We hypothesize that the phenotypic changes were solely due to the LTR insertion. Furthermore, we hypothesize that insertion of REV LTR into an analogous location in a different MDV would result in a similar phenotypic change. To test these hypotheses, we inserted the REV LTR into a bacterial artificial chromosome (BAC) clone of a very virulent strain of MDV, Md5, and designated the virus rMd5-RM1-LTR. The rMd5-RM1-LTR virus and the rMd5 virus were passaged in duck embryo fibroblast cells for up to 40 passages before pathogenicity studies. Susceptible chickens were inoculated intra-abdominally at hatch with the viruses rMd5-RM1-LTR, rMd5 BAC parental virus, wild-type strain Md5, or strain RM1 of MDV. The rMd5-RM1-LTR virus was attenuated at cell culture passage 40, whereas the rMd5 BAC without RM1 LTR retained its pathogenicity at cell culture passage 40. Using polymerase chain analysis, the RM1 LTR insert was detected in MDV isolated from buffy coat cells collected from chickens inoculated with rMd5-RM1-LTR, but only at 1 week post inoculation. The data suggest that the presence of the RM1 LTR insert within MDV genome for 1 week post inoculation with virus at hatch is sufficient to cause a reduction in pathogenicity of strain Md5 of MDV.


Assuntos
Galinhas , Cromossomos Artificiais Bacterianos/genética , Mardivirus/genética , Mardivirus/patogenicidade , Doença de Marek/virologia , Vírus da Reticuloendoteliose Aviária/genética , Sequências Repetidas Terminais/genética , Animais , Anticorpos Antivirais/sangue , Células Cultivadas , Feminino , Masculino , Mutagênese Insercional/métodos , Reação em Cadeia da Polimerase , Replicação Viral/genética
15.
Virus Genes ; 42(3): 369-76, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21340512

RESUMO

Researchers reported that co-cultivating the JM/102W strain of Marek's disease virus (MDV) with reticuloendotheliosis virus (REV) resulted in an REV long terminal repeat (LTR) being inserted into the internal repeat short (IRS) region of JM/102W. When the resulting recombinant virus was serially passed in cell culture, the initial LTR was duplicated and a second LTR spontaneously appeared in the terminal repeat short (TRS) region of the MDV genome. The virus, designated RM1, was significantly attenuated but still induced severe bursal and thymic atrophy (Isfort et al. PNAS 89:991-995). To determine whether the altered phenotype was due solely to the LTR, we cloned the LTR from the RM1 IRS region and inserted it into the IRS region of a very virulent bacterial artificial clone (BAC) of the Md5 strain of MDV, which we designated rMd5-RM1-LTR. During blind passage in duck embryo fibroblast cultures, the initial LTR in the rMd5-RM1-LTR was also duplicated, with LTRs appearing in both IRS and TRS regions of the MDV genome. The inserted LTR sequences and transcripts associated with the MDV open reading frames MDV085, MDV086, SORF2, US1, and US10 were molecularly characterized. The parental Md5 BAC contains a family of transcripts of 3, 2, and 1 kb that all terminate at the end of the US10 gene. The rMd5-RM1-LTR and RM1 viruses both express an additional 4 kb transcript that originates in the LTR and also terminates after US10. Collectively, the data suggest that our engineered rMd5-RM1-LTR virus very closely resembles the RM1 virus in its structure and transcription patterns.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/genética , Mutagênese Insercional , Vírus da Reticuloendoteliose/genética , Sequências Repetidas Terminais , Proteínas Virais/genética , Animais , Linhagem Celular , Galinhas , Patos , Herpesvirus Galináceo 2/metabolismo , Doença de Marek/virologia , Dados de Sequência Molecular , Doenças das Aves Domésticas/virologia , Proteínas Virais/metabolismo
16.
Avian Pathol ; 39(5): 383-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20954015

RESUMO

Nine reticuloendotheliosis virus (REV) isolates obtained from broiler breeders, turkeys, and prairie chickens located in three different geographical regions in the USA, and three isolates obtained from known contaminated live-virus vaccines were characterized using polymerase chain reaction (PCR) and indirect immunofluorescence (IFA) assays. All isolates were propagated in chicken embryo fibroblasts obtained from a specific pathogen free breeder flock. PCR analysis of all 12 isolates resulted in the amplification of the 291-bp REV long-terminal repeat region (LTR); none of the isolates exhibited a different pattern or shift from the expected PCR product of REV LTR. The subtype of the REV isolates was determined by IFA using REV-specific monoclonal antibodies, 11B118.22, 11C237.8, and 11D182. Results from sub-typing indicated that all nine isolates from broiler breeders, turkeys, and prairie chickens belonged to subtype 3, and are antigenically related to the chick syncytial virus (CSV) strain of REV, the prototype of subtype 3 REV. In contrast, the three isolates from contaminated vaccines were classified as subtype 2, and were antigenically related to spleen necrosis virus (SNV) strain of REV, the prototype of subtype 2 REV. Three isolates representing REV isolated from broiler breeders, turkeys, and prairie chickens were cloned and further evaluated by DNA sequence analysis of the envelope gene. Results from DNA sequence analysis confirmed those from sub-typing and indicated that the three REV isolates representing those from broiler breeders, turkeys, and prairie chickens are closely related to CSV of REV, with an amino acid homology of 98% or greater as compared with SNV with an amino acid homology of 95% or less. Data from this study clearly indicate that subtype 3 is the most common subtype of REV circulating in three different avian species, namely broiler breeders, turkeys and prairie chickens, located in three different geographical regions in the United States.


Assuntos
Galliformes , Vírus da Reticuloendoteliose Aviária/classificação , Reticuloendoteliose Aviária/virologia , Sequência de Aminoácidos , Animais , Linhagem Celular , Embrião de Galinha , DNA Viral , Fibroblastos/virologia , Regulação Viral da Expressão Gênica/fisiologia , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase/veterinária , Estados Unidos/epidemiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
17.
Avian Dis ; 51(3): 725-32, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17992933

RESUMO

The effects of viral strain, viral dose, and age of bird at inoculation on subgroup J avian leukosis virus (ALV J) persistence, neutralizing antibody (VNAb) response, and tumors were studied in commercial meat-type chickens. Chickens were inoculated on the fifth day of embryonation (5 ED) or on day of hatch (DOH) with either 100 or 10,000 50% tissue-culture infective dose (TCID50) of one of three ALV J strains, namely ADOL Hcl, ADOL 6803, or ADOL 4817. At 1, 3, 7, 11, 15, 19, 23, 27, and 32 wk posthatch, chickens were examined for ALV J viremia and VNAb against the inoculated strain of ALV J. A high incidence (83%-100%) of ALV J persistence was observed in all treatment groups. Development of VNAb did not always lead to viremia-free status; even though 18% of the chickens developed VNAb, only 4% were able to clear viremia. The viral strain, dose, and age of bird at inoculation seemed to have an effect on the incidence of VNAb; however, the differences were statistically significant in only some treatment groups. Chickens infected with ADOL 6803 had higher incidence of VNAb than chickens infected with ADOL Hc1 and ADOL 4817 (P < 0.05 in groups 5 ED at 100 TCID50 and DOH at 10,000 TCID50). There was a trend in all groups inoculated with 100 TCID50 to have higher incidence of VNAb than that of groups inoculated with 10,000 TCID50 (ADOL 6803 at 5 ED and ADOL 4817 at DOH [P < 0.05]; ADOL Hc1 at DOH [P < 0.08]). In most treatment groups (ADOL Hc1 at 100 and 10,000 TCID50, ADOL 6803 at 10,000 TCID50, and ADOL 4817 at 100 TCID50), chickens inoculated at DOH had higher incidence of VNAb than that of chickens inoculated at 5 ED (ADOL 6803 at 10,000 TCID50 [P < 0.05], ADOL Hc1 at 100 TCID50 [P < 0.08]). Incidence of ALV J-induced tumors and tumor spectrum were influenced by viral strain, age at inoculation, and VNAb response.


Assuntos
Envelhecimento , Vírus da Leucose Aviária/classificação , Leucose Aviária/virologia , Galinhas , Neoplasias/veterinária , Doenças das Aves Domésticas/virologia , Animais , Anticorpos Antivirais/sangue , Carne , Neoplasias/virologia
18.
Avian Dis ; 50(3): 342-7, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17039832

RESUMO

Chickens from seven different parental lines of commercial White Leghorn layer flocks from three independent breeders were inoculated with a naturally occurring avian leukosis virus (ALV) containing an ALV-B envelope and an ALV-J long terminal repeat (LTR) termed ALV-B/J. Additional groups of chickens from the same seven parental lines were inoculated with ALV-B. Chickens were tested for ALV viremia and antibody at 0, 4, 8, 16, and 32 wk postinfection. Chickens from all parental lines studied were susceptible to infection with ALV-B with 40%-100% of inoculated chickens positive for ALV at hatch following embryo infection. Similarly, infection of egg layer flocks with the ALV-B/J recombinant virus at 8 days of embryonation induced tolerance to ALV with 86%-100% of the chickens viremic, 40%-75% of the chickens shedding virus, and only 2/125 (2%) of the chickens producing serum-neutralizing antibodies against homologous ALV-B/J recombinant virus at 32 wk postinfection. In contrast, when infected with the ALV-B/J recombinant virus at hatch, 33%-82% of the chickens were viremic, 28%-47% shed virus, and 0%-56% produced serum-neutralizing antibodies against homologous ALV-B/J recombinant virus at 32 wk postinfection. Infection with the ALV-B/J recombinant virus at embryonation and at hatch induced predominately lymphoid leukosis (LL), along with other common ALV neoplasms, including erythroblastosis, osteopetrosis, nephroblastomas, and rhabdosarcomas. No incidence of myeloid leukosis (ML) was observed in any of the commercial White Leghorn egg layer flocks infected with ALV-B/J in the present study. Data suggest that the parental line of commercial layers may influence development of ALV-B/J-induced viremia and antibody, but not tumor type. Differences in type of tumors noted in the present study and those noted in the field case where the ALV-B/J was first isolated may be attributed to differences in the genetics of the commercial layer flock in which ML was first diagnosed and the present commercial layer flocks tested in the present study.


Assuntos
Vírus da Leucose Aviária/genética , Leucose Aviária/virologia , Galinhas/virologia , Predisposição Genética para Doença , Sequências Repetidas Terminais/genética , Proteínas do Envelope Viral/genética , Animais , Neoplasias/genética , Neoplasias/veterinária , Neoplasias/virologia , Vírus Reordenados/genética , Viremia , Eliminação de Partículas Virais
19.
Avian Dis ; 50(4): 572-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17274296

RESUMO

Infection of broiler chickens with subgroup J avian leukosis virus (ALV) results in the induction of myeloid tumors. However, although egg-type chickens are susceptible to infection with ALV-J, the tumor incidence is very low, and on rare occasions the tumors observed are of the myeloid lineage. We recently described the isolation of an ALV (AF115-4) from commercial egg-type chickens suffering from myeloid leukosis. AF115-4 was initially identified as an ALV-J isolate based on PCR analysis of the long terminal repeat (LTR). However, further characterization of the viral envelope indicated that the virus is recombinant with subgroups B envelope and J LTR. Here we further characterize this recombinant virus at both the molecular and biological levels. We show that the AF115-4 isolate expresses a recombinant envelope glycoprotein encoded by a subgroup B gp85 region and a subgroup E gp37 region. The host range ofAF115-4 was analyzed using cells resistant to infection by subgroups A/B, J, or E; this shows that no ALV-J was present in the isolates obtained from the affected chickens. Additional antigenic characterization of AF115-4 using chicken sera specific for subgroups B or J indicated that no ALV-J was present in the samples examined. Inoculation of AF 115-4 into ALV-susceptible 1515 X 71 chickens resulted in the induction of lymphoid leukosis but not the expected myeloid leukosis affecting the commercial chickens. These results suggest that differences in the genetic makeup of the chickens from which AF115-4 was isolated and the line 1515 X 71 used in the present experiments may be responsible for the observed differences in pathogenicity. In addition, the results suggest that ALV-J continues to evolve by recombination, generating new viruses with different pathological properties.


Assuntos
Vírus da Leucose Aviária/genética , Sequências Repetidas Terminais/genética , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Leucose Aviária/virologia , Sequência de Bases , Linhagem Celular , Galinhas/virologia , Dados de Sequência Molecular , Doenças das Aves Domésticas/virologia , Proteínas do Envelope Viral/química
20.
Avian Dis ; 49(2): 214-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16094825

RESUMO

White leghorn chickens from seven 15.B congenic lines (genetically similar except for genes linked to the major histocompatibility complex [MHC] B haplotype) and two Line 0.B semicongenic lines were infected at hatch with strain ADOL Hc-1 of subgroup J avian leukosis virus (ALV-J). At 5, 8, 16, and 36 wk of age, chickens were tested for viremia, serum-neutralizing antibody, and cloacal shedding. Chickens were also monitored for development of neoplasia. In the 15.B congenic lines (B*2, B*5, B*12, B*13, B*15, B*19, and B*21) there were no significant differences in the incidence of viremia between B haplotypes. In fact, infection at hatch in all of the 15.B congenic lines induced tolerance to ALV-J because 100% of these chickens were viremic and transient circulating serum-neutralizing antibody was detected in only a few chickens throughout the 36 wk experiment. However, at 16 wk of age more B*15 chickens had antibody and fewer B*15 chickens shed virus than did the 16-wk-old B*2, B*5, or B*13 chickens. Moreover, compared with B*15 chickens, a higher percentage of B*13 chickens consistently shed virus from 8 wk postinfection to termination at 36 wk postinfection. The B haplotype had a transient effect on viral clearance in Line 0.B semicongenics, as more B*13 than B*21 chickens remained viremic through 5 wk of age. Very few (0%-18%) of the Line 0.B semicongenic chickens shed virus. By 36 wk of age, all Line 0 B*13 and B*21 chickens produced serum-neutralizing antibodies and cleared the virus. These results show that following ALV-J infection at hatch the immune response is influenced transiently by the B haplotype and strongly by the line of chicken. Although this study was not designed to study the effect of endogenous virus on ALV-J infection, the data suggest that endogenous virus expression reduced immunity to ALV-J in Line 15I5, compared with Line 0, a line known to lack endogenous virus genes.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/genética , Galinhas/genética , Doenças das Aves Domésticas/virologia , Animais , Leucose Aviária/imunologia , Tipagem e Reações Cruzadas Sanguíneas/veterinária , Haplótipos/genética , Testes de Hemaglutinação/veterinária , Complexo Principal de Histocompatibilidade/genética , Testes de Neutralização/veterinária , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/imunologia , Viremia/veterinária , Eliminação de Partículas Virais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...