Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Avian Pathol ; 50(6): 490-499, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34463588

RESUMEN

Md5-BAC-REV-LTR is a recombinant Marek's disease virus (MDV), with an insertion of the long terminal repeat (LTR) of reticuloendotheliosis virus (REV) into the genome of the highly virulent MDV strain rMd5. It has been shown that Md5-BAC-REV-LTR does not induce tumours and confers high protection against challenge with MDV in 15 × 7 chickens. The objective of the present study was to evaluate the protection and safety (in terms of oncogenicity and immunosuppression) of Md5-BAC-REV-LTR in commercial meat-type chickens bearing maternal antibodies against MDV. Our results show that sub-cutaneous administration of Md5-BAC-REV-LTR at 1 day of age conferred high protection (protection index PI = 84.2) against an early challenge (1 day) by contact exposure to shedder birds infected with the vv+ MDV 648A strain. In such stringent challenge conditions, Md5-BAC-REV-LTR was more protective than a commercial CVI988 (PI = 12.4) and similar to the experimental vaccine Md5-BACΔmeq (PI = 92.4). Furthermore, Md5-BAC-REV-LTR did not induce either tumours or immunosuppression in this study. Immunosuppression was evaluated by the relative lymphoid organ weights and also by the ability of the vaccine to induce late-MDV-induced immunosuppression associated with reactivation of the virus. This study shows that Md5-BAC-REV-LTR has the potential to be used as a MD vaccine and is highly protective against early challenge with vv+ MDV.RESEARCH HIGHLIGHTSMd5-BAC-REV-LTR is highly protective against early challenge with vv+ MDV in commercial meat-type chickens.Md5-BAC-REV-LTR does not cause early immunosuppression.Md5-BAC-REV-LTR does not cause late immunosuppression.Unlike other serotype 1 vaccines, Md5-BAC-REV-LTR is not detected in feather pulp at 7 days post vaccination.


Asunto(s)
Herpesvirus Gallináceo 2 , Vacunas contra la Enfermedad de Marek , Virus de la Reticuloendoteliosis , Animales , Pollos , Terapia de Inmunosupresión/veterinaria , Vacunas contra la Enfermedad de Marek/genética , Carne , Secuencias Repetidas Terminales/genética
2.
Avian Dis ; 59(2): 255-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26473676

RESUMEN

Laryngotracheitis (LT) is a highly contagious respiratory disease of chickens that produces significant economic losses to the poultry industry. Traditionally, LT has been controlled by administration of modified live vaccines. In recent years, the use of recombinant DNA-derived vaccines using turkey herpesvirus (HVT) and fowlpox virus has expanded, as they protect not only against the vector used but also against LT. However, HVT-based vaccines confer limited protection against challenge, with emergent very virulent plus Marek's disease virus (vv+MDV). Serotype 1 vaccines have been proven to be the most efficient against vv+MDV. In particular, deletion of oncogene MEQ from the oncogenic vvMDV strain Md5 (BACδMEQ) resulted in a very efficient vaccine against vv+MDV. In this work, we have developed two recombinant vaccines against MD and LT by using BACδMEQ as a vector that carries either the LT virus (LTV) gene glycoprotein B (gB; BACΔMEQ-gB) or LTV gene glycoprotein J (gJ; BACδMEQ-gJ). We have evaluated the protection that these recombinant vaccines confer against MD and LT challenge when administered alone or in combination. Our results demonstrated that both bivalent vaccines (BACΔMEQ-gB and BACδMEQ-gJ) replicated in chickens and were safe to use in commercial meat-type chickens bearing maternal antibodies against MDV. BACΔMEQ-gB protected as well as a commercial recombinant (r)HVT-LT vaccine against challenge with LTV. However, BACδMEQ-gJ did not protect adequately against LT challenge or increase protection conferred by BACΔMEQ-gB when administered in combination. On the other hand, both BACΔMEQ-gB and BACδMEQ-gJ, administered alone or in combination, protected better against an early challenge with vv+MDV strain 648A than commercial strains of rHVT-LT or CVI988. Our results open a new avenue in the development of recombinant vaccines by using serotype 1 MDV as vectors.


Asunto(s)
Pollos , Infecciones por Herpesviridae/veterinaria , Herpesvirus Gallináceo 1/inmunología , Mardivirus/clasificación , Enfermedad de Marek/prevención & control , Vacunas Virales/inmunología , Animales , Femenino , Infecciones por Herpesviridae/prevención & control , Mardivirus/inmunología , Proyectos Piloto , Vacunas de ADN , Replicación Viral
3.
Avian Dis ; 58(2): 232-43, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25055627

RESUMEN

The serotype 1 Marek's disease virus (MDV) is the causative agent for Marek's disease (MD), a lymphoproliferative disease of chickens of great concern to the poultry industry. CVI988 (Rispens vaccine), an attenuated serotype 1 MDV, is currently the most efficacious commercially available vaccine for preventing MD. However, it is difficult to detect and differentiate CVI988 when other serotype 1 MDVs are present. To facilitate the detection of CVI988, we developed two sets of primers for a mismatch amplification mutation assay (MAMA) PCR that targeted the single nulceotide polymorphism associated with the H19 epitope of the phosphorylated protein 38 gene. The PCR was very specific. One primer set (oncogenic primers) amplified DNA from 15 different serotype 1 MDVs except CVI988. The other primer set (CVI988 primers) amplified DNA from CVI988 but not from any of the other 15 serotype 1 MDVs. A real-time PCR assay was developed using MAMA primers, and specificity and sensitivity was evaluated in vitro and in vivo. Mixtures of plasmids (CVI988 plasmid and oncogenic plasmid) at various concentrations were used to evaluate the sensitivity/specificity of MAMA primers in vitro. Both primer setswere able to amplify as little as one copy of their respective plasmid. Oncogenic primers were highly specific and only amplified CVI988 plasmid when the concentration of oncogenic plasmid was very low (1 X 10(1)) and CVI988 plasmid was very high (1 X 10(6)). Specificity of CVI988 primers was not as high because they could amplify oncogenic plasmids when the concentration of CVI988 plasmid was 1 x 10(3) and the concentration of oncogenic 1 x 10(2). Validation of MAMA primers in in vivo samples demonstrated that oncogenic primers can be used for both early diagnosis of MD in feather pulp (FP) samples collected at 3 wk of age and confirmation of MD diagnosis in tumors. CVI988 primers could be used to monitor CVI988 vaccination in samples with a low load of oncogenic MDV DNA (latently infected samples or negative) but not in samples with a high load of oncogenic MDV DNA (tumors). Our results suggest that monitoring CVI988 vaccination in FP samples collected at 1 wk of age ensures the specificity of the CVI988 primers.


Asunto(s)
Pollos , Herpesvirus Gallináceo 2/inmunología , Herpesvirus Gallináceo 3/inmunología , Vacunas contra la Enfermedad de Marek/inmunología , Enfermedad de Marek/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Animales , Embrión de Pollo , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 3/genética , Enfermedad de Marek/inmunología , Vacunas contra la Enfermedad de Marek/genética , Polimorfismo de Nucleótido Simple , Enfermedades de las Aves de Corral/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
4.
Avian Pathol ; 41(3): 259-65, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22702453

RESUMEN

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.


Asunto(s)
Pollos , Cromosomas Artificiales Bacterianos/genética , Mardivirus/genética , Mardivirus/patogenicidad , Enfermedad de Marek/virología , Virus de la Reticuloendoteliosis Aviar/genética , Secuencias Repetidas Terminales/genética , Animales , Anticuerpos Antivirales/sangre , Células Cultivadas , Femenino , Masculino , Mutagénesis Insercional/métodos , Reacción en Cadena de la Polimerasa , Replicación Viral/genética
5.
Avian Pathol ; 41(3): 267-75, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22702454

RESUMEN

Previous studies have demonstrated the presence of multiple strains of Marek's disease virus simultaneously circulating within poultry flocks, leading to the assumption that individual birds are repeatedly exposed to a variety of virus strains in their lifetime. Virus competition within individual birds may be an important factor that influences the outcome of co-infection under field conditions, including the potential outcome of emergence or evolution of more virulent strains. A series of experiments was designed to evaluate virus competition within chickens following simultaneous challenge with two virulent serotype 1 Marek's disease virus strains, using either pathogenically similar (rMd5 and rMd5/pp38CVI) or dissimilar (JM/102W and rMd5/pp38CVI) virus pairs. Bursa of Fabricius, feather follicle epithelium, spleen, and tumour samples were collected at multiple time points to determine the frequency and distribution of each virus present using pyrosequencing, immunohistochemistry and virus isolation. In the similar pair, rMd5 appeared to have a competitive advantage over rMd5/pp38CVI, which in turn had a competitive advantage over the less virulent JM/102W in the dissimilar virus pair. Dominance of one strain over the other was not absolute for either virus pair, as the subordinate virus was rarely eliminated. Interestingly, competition between two viruses with either pair rarely ended in a draw. Further work is needed to identify factors that influence virus-specific dominance to better understand what characteristics favour emergence of one strain in chicken populations at the expense of other strains.


Asunto(s)
Pollos , Coinfección/virología , Herpesvirus Gallináceo 2/patogenicidad , Enfermedad de Marek/virología , Interacciones Microbianas/fisiología , Animales , Anticuerpos Monoclonales , Herpesvirus Gallináceo 2/clasificación , Inmunohistoquímica/veterinaria , Interacciones Microbianas/genética , Dinámica Poblacional , Análisis de Secuencia de ADN/veterinaria , Especificidad de la Especie , Estadísticas no Paramétricas , Virulencia
6.
Avian Dis ; 56(3): 494-500, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23050465

RESUMEN

CVI988 (Rispens) is currently the most effective vaccine used to protect against Marek's disease, a lymphoproliferative disease of chickens. A MEQ-deleted Marek's disease virus strain has shown promise as a vaccine candidate; however, unpublished results from vaccine safety trials suggest that this candidate vaccine induces unwanted lymphoid atrophy. The current study evaluated lymphoid atrophy at multiple time points between 2- and 8-wk postinoculation and attempted to correlate results with virus replication in the thymus. Results confirm reports that MEQ-deleted virus strains are able to cause thymus and bursa atrophy, which is most severe at 2-wk postinoculation. The MEQ-deleted virus strains induced lower body weights and relative thymus and bursa weights compared to uninoculated and Rispens-vaccinated chickens at multiple time points between 2- and 8-wk postinoculation. Both MEQ-deleted virus strains produced high levels of in vivo virus replication in the thymus at rates significantly greater than in Rispens-vaccinated chickens and were comparable to levels of RM1 virus, a MDV previously shown to induce severe thymus and bursa atrophy. Virus replication was highly correlated with relative thymus weights at each time point. Understanding this delicate balance between inducing maximum disease protection and preventing immunodepressive effects is critical for the development of future Marek's disease vaccines.


Asunto(s)
Bolsa de Fabricio/patología , Pollos , Vacunas contra la Enfermedad de Marek/inmunología , Enfermedad de Marek/prevención & control , Proteínas Oncogénicas Virales/genética , Timo/patología , Animales , Eliminación de Gen , Vacunas contra la Enfermedad de Marek/efectos adversos , Enfermedades de las Aves de Corral/prevención & control , Reacción en Cadena en Tiempo Real de la Polimerasa , Aumento de Peso/inmunología
7.
J Gen Virol ; 92(Pt 3): 598-607, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21123546

RESUMEN

Genetic homogeneity of a test population is essential to precisely associate a viral genome sequence and its phenotype at the nucleotide level. However, homogeneity is not easy to achieve for Marek's disease virus (MDV) due to its strictly cell-associated replication. To address this problem, two virulent infectious bacterial artificial chromosome (BAC) clones of MDV were generated from an MDV genome previously cloned as five overlapping cosmids. The Md5SN5BAC clone has the BAC vector inserted between the 3' ends of UL3 and UL4, such that no known ORFs should be disrupted. The BAC vector is flanked by loxP sites, so that it can be deleted from the viral genome by transfecting Md5SN5BAC into a newly developed chicken cell line that constitutively expresses Cre recombinase. The Md5B40BAC clone has the BAC vector replacing a portion of US2, a location similar to that used by other groups to construct MDV-BAC clones. Although both BACs were capable of producing infectious virulent MDV when inoculated into susceptible chickens, Md5B40BAC-derived viruses showed somewhat better replication in vivo and higher virulence. Removal of the BAC vector in Md5SN5BAC-derived viruses had no influence on virulence. Interestingly, when genetically homogeneous virulent MDV generated from Md5B40BAC was mixed with avirulent virus, the overall virulence of the mixed population was noticeably compromised, which emphasizes the importance of MDV population complexity in pathogenesis.


Asunto(s)
Mardivirus/genética , Mardivirus/patogenicidad , Enfermedad de Marek/virología , Animales , Línea Celular , Pollos , Cromosomas Artificiales Bacterianos , Clonación Molecular , ADN Viral/química , ADN Viral/genética , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Transfección , Carga Viral , Ensayo de Placa Viral , Virulencia
8.
Virus Genes ; 42(3): 369-76, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21340512

RESUMEN

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.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Regulación Viral de la Expresión Génica , Herpesvirus Gallináceo 2/genética , Mutagénesis Insercional , Virus de la Reticuloendoteliosis/genética , Secuencias Repetidas Terminales , Proteínas Virales/genética , Animales , Línea Celular , Pollos , Patos , Herpesvirus Gallináceo 2/metabolismo , Enfermedad de Marek/virología , Datos de Secuencia Molecular , Enfermedades de las Aves de Corral/virología , Proteínas Virales/metabolismo
9.
Avian Dis ; 55(3): 358-67, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22017031

RESUMEN

Marek's disease (MD) is a lymphoproliferative disorder of domestic chickens caused by a highly contagious and oncogenic alpha-herpesvirus, Marek's disease virus (MDV). MD is characterized by bursal-thymic atrophy and rapid onset of T-cell lymphomas that infiltrate lymphoid tissues, visceral organs, and peripheral nerves with severe clinical signs that include transient paralysis, anemia, weight loss, and neurologic disorders. Using overlapping cosmids- and BAC-cloned MDV, it has been shown that MDV-encoded vIL-8, pp38, vTR, vLIP, RLORF4, and meq are among the many essential genes that play critical roles in viral pathogenesis. Of all the genes investigated so far, only meq has been shown to be consistently expressed in all MDV-derived tumors and lymphoblastoid cell lines. Meq is a basic leucine-zipper protein that shares homology with the jun/fos family of transcriptional factors. There are two copies of meq gene within the MDV genome that are only present in the serotype-1 strains. It has been shown conclusively that deletion of meq results in loss of transformation of T cells in chickens, with no effect on the early cytolytic phase of infection in lymphoid organs, which is essential for induction of innate and adaptive immunity. The goal of this study was to investigate 1) the effect of the meq oncogene on the expression pattern of select chicken immune and nonimmune-related genes, and 2) its potential role in MDV-induced apoptosis. We used real-time reverse transcriptase-polymerase chain reaction to evaluate the expression profiling of a panel of chicken genes in rMd5- and rMd5deltameq-infected chickens at 5, 14, 21, and 35 days postinfection (dpi). Although the transcriptional activities of several immune-related genes, including IL-6, IL-10, cMGF, GM-CSF, iNOS, IFNbeta, and INFgamma, were higher in rMd5deltameq-infected chickens at 5 dpi when compared to the rMd5-infected birds, the differences in expression levels of the tested genes between the two viral constructs were not significant. In addition, a reduction in the transcriptional activity of Bdcl2 in recombinant fowlpox virus (rFPV)+meq-infected chicken embryonic fibroblasts suggested that meq alone did not impede FPV-induced apoptosis. The likely suppressive nature and anti-inflammatory function of the meq oncogene and its possible role in virus-induced cell death is discussed.


Asunto(s)
Mardivirus/genética , Enfermedad de Marek/inmunología , Proteínas Oncogénicas Virales/genética , Animales , Apoptosis , Proteínas Aviares/genética , Proteínas Aviares/inmunología , Pollos , Citocinas , Virus de la Viruela de las Aves de Corral/fisiología , Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/inmunología , Interferón beta/genética , Interferón beta/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Enfermedad de Marek/genética , Enfermedad de Marek/virología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Proteínas Oncogénicas Virales/inmunología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
10.
Avian Pathol ; 39(5): 383-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20954015

RESUMEN

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.


Asunto(s)
Galliformes , Virus de la Reticuloendoteliosis Aviar/clasificación , Reticuloendoteliosis Aviar/virología , Secuencia de Aminoácidos , Animales , Línea Celular , Embrión de Pollo , ADN Viral , Fibroblastos/virología , Regulación Viral de la Expresión Génica/fisiología , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa/veterinaria , Estados Unidos/epidemiología , Proteínas Virales/genética , Proteínas Virales/metabolismo
11.
Avian Dis ; 54(2): 862-9, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20608531

RESUMEN

The Marek's disease virus (MDV) induces T-cell tumors in susceptible chickens. Of the 80 to 100 known MDV genes, only the MDV MEQ gene was shown to have transforming properties. Further evidence that MEQ is probably the principal oncogene in MDV came when researchers used overlapping cosmid clones of MDV and demonstrated that deleting MEQ resulted in a highly protective Marek's disease (MD) vaccine. We deleted both copies of MEQ from a bacterial artificial chromosome clone (BAC) of MDV. The virus, BACdelMEQ, was completely attenuated and did not appear to have any adverse effect on chicken body weight in MDV maternal-antibody-positive chickens, as measured at 8 wk of age. In two protection studies, BACdelMEQ efficiently protected susceptible chickens from a challenge by MDV strain 686, one of the most virulent MDV strains. In both protection studies, the BACdelMEQ protected chickens significantly better than the commercial MD vaccine, CVI988/Rispens. Only the protein-coding sequences of MEQ were deleted and all upstream and downstream regulatory sequences were left intact. Thus, BACdelMEQ has the potential to be a superior MD vaccine as well as a vector to deliver various foreign genes to poultry.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Cromosomas Artificiales Bacterianos/inmunología , Mardivirus/genética , Enfermedad de Marek/prevención & control , Proteínas Oncogénicas Virales/genética , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Pollos , Clonación Molecular , Eliminación de Gen , Genoma Viral , Mardivirus/clasificación , Mardivirus/patogenicidad , Enfermedad de Marek/virología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Virulencia
12.
Avian Dis ; 54(3): 1038-49, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20945786

RESUMEN

Marek's disease virus (MDV) is ubiquitous within commercial poultry flocks because current vaccines do not prevent MDV infection or transmission. In order for newly-evolved MDV strains to become established within a flock, it seems inevitable that any new strain would need to infect and replicate in chickens previously infected with resident MDV strains. This phenomenon is difficult to detect and there is no clear evidence that it is even possible. Four experiments were performed to demonstrate superinfection and evaluate the effect of time between challenges on the effect of superinfection with the use of two pairs of fully virulent MDV strains that could be discriminated by novel technology: 1) JM/102W and rMd5//38CVI, and 2) rMd5 and rMd5//38CVI. Feather follicle epithelium (FFE), spleen, and tumor samples were collected at single or multiple time points from the same bird to determine the frequency and distribution of each virus present following superinfection, with the use of pyrosequencing and immunohistochemistry. Superinfection was observed in 82 of 149 (55%) FFE samples following short-interval challenge (24 hr) compared to only 6 of 121 (5%) samples following long-interval challenge (13 days), indicating a strong influence of challenge interval. In cases where the first inoculated virus was weak or delayed, the second inoculated virus was detected in 42 of 95 (44%) birds. In tumors from dually challenged birds, the second virus was again present much more often following short-interval challenge (68%) compared to long-interval challenge (11%). Virus mixtures in tumors were less common compared to those in FFE samples. Vaccination with turkey herpesvirus had no significant effect on the virus frequency for either virus pair or challenge time interval, suggesting these conclusions may be applicable to vaccinated chickens in the field. These studies demonstrated superinfection for the first time with two fully virulent MDV strains and suggest that short-interval challenge exposure and/or weak initial exposures may be important factors leading to superinfection--a prerequisite for the establishment of a second virus strain in the population. This model system should be useful to elucidate this important phenomenon further.


Asunto(s)
Pollos , Mardivirus/inmunología , Enfermedad de Marek/prevención & control , Sobreinfección/veterinaria , Vacunas Virales/inmunología , Animales , Femenino , Esquemas de Inmunización , Masculino , Mardivirus/patogenicidad , Enfermedad de Marek/inmunología , Organismos Libres de Patógenos Específicos , Sobreinfección/virología , Factores de Tiempo , Virulencia
13.
Avian Dis ; 64(3): 243-246, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33205163

RESUMEN

Marek's disease (MD) is an oncogenic, lymphoproliferative, and highly contagious disease of chickens. Its etiologic agent is the alphaherpesvirus Marek's disease virus (MDV, Gallid alphaherpesvirus 2), and it is a chronic and ubiquitous problem for the poultry industry with significant economic impact in the United States and worldwide. We have previously demonstrated that MDV attenuated by dicodon deoptimization of the UL54 gene results in reduced gene product accumulation in vitro, with reduced viral genome copy number upon infection and reduced atrophy of bursa and thymus in vivo as well. In this report we detail our attempts to use the same attenuation strategy on a meq-deleted MDV mutant, rMd5B40ΔMeq. Unlike the wild-type rMd5B40 virus the rMd5B40ΔMeq is no longer oncogenic, but infected birds experience an unacceptable amount of bursa and thymus atrophy (BTA). We produced two meq-deleted MDV recombinants with a dicodon-deoptimized UL54 (rMd5B40ΔMeq/UL54deop1 and -deop2) and tested their tendency to cause BTA and to serve as a protective vaccine. We found that, although dicodon deoptimization of the UL54 gene results in a virus that spares the infected animal from atrophy of the bursa and thymus, the meq-deleted UL54-deoptimized recombinant is also less protective than the meq-deleted virus without UL54 deoptimization, the HVT + SB1 combination vaccine, or the Rispens (CVI988) vaccine.


Asunto(s)
Pollos , Codón/genética , Proteínas Inmediatas-Precoces/genética , Vacunas contra la Enfermedad de Marek/genética , Enfermedad de Marek/inmunología , Proteínas Oncogénicas Virales/deficiencia , Enfermedades de las Aves de Corral/inmunología , Proteínas Virales/genética , Animales , Atrofia/veterinaria , Eliminación de Gen , Linfocitos/patología , Organismos Libres de Patógenos Específicos
14.
J Virol Methods ; 148(1-2): 125-31, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18082901

RESUMEN

Marek's disease, a T cell lymphoma, is an economically important disease of poultry caused by the Marek's disease virus (MDV), a highly cell-associated alphaherpesvirus. A greater understanding of viral gene function and the contribution of sequence variation to virulence should facilitate efforts to control Marek's disease in chickens. To characterize a naturally occurring single nucleotide polymorphism (SNP; AY510475:g.108,206C>T) in the MDV UL41 gene that results in a missense mutation (AAS01683:p.Arg377Cys), bacterial artificial chromosome (BAC)-derived MDVs that differed only in the UL41 SNP were evaluated using a head-to-head competition assay in vitro. Monitoring the frequency of each SNP by pyrosequencing during virus passage determined the ratio of each viral genome in a single monolayer, which is a very sensitive method to monitor viral fitness. MDV with the UL41*Cys allele showed enhanced fitness in vitro. To evaluate the mechanism of altered viral fitness caused by this SNP, the virion-associated host shutoff (vhs) activity of both UL41 alleles was determined. The UL41*Cys allele had no vhs activity, which suggests that enhanced fitness in vitro for MDV with inactive vhs was due to reduced degradation of viral transcripts. The in vitro competition assay should be applicable to other MDV genes and mutations.


Asunto(s)
Herpesvirus Gallináceo 2/fisiología , Polimorfismo de Nucleótido Simple , Proteínas Virales/genética , Virología/métodos , Sustitución de Aminoácidos/genética , Animales , Embrión de Pollo , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 2/crecimiento & desarrollo , Ensayo de Placa Viral , Virulencia/genética
15.
Virology ; 516: 219-226, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29407380

RESUMEN

Marek's disease virus (MDV) is an oncogenic alphaherpesvirus of Gallus gallus, the domesticated chicken. Control strategies rely upon vaccination with live attenuated viruses of antigenically similar avian herpesviruses or attenuated strains of MDV. Recent studies in other viruses have shown that recoding certain viral genes to employ synonymous but rarely-used codon pairs resulted in viral attenuation. We deoptimized two MDV proteins, UL54/ICP27 and UL49/VP22, and demonstrate that the more severely deoptimized variant of UL54 accumulates significantly less gene product in vitro. Using these UL54 deoptimized mutants, we further demonstrate that animals infected with the UL54-recoded recombinant virus exhibited decreased viral genome copy number in lymphocytes, reduced lymphoid atrophy and reduced tumor incidence. This study demonstrates that codon pair deoptimization of a single viral gene can produce attenuated strains of MDV. This approach may be useful as a rational way of making novel live attenuated virus vaccines for MDV.


Asunto(s)
Codón/genética , Herpesvirus Gallináceo 2/genética , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/virología , Proteínas Virales/genética , Animales , Pollos , Codón/metabolismo , Patos , Herpesvirus Gallináceo 2/crecimiento & desarrollo , Herpesvirus Gallináceo 2/metabolismo , Proteínas Virales/metabolismo
16.
Avian Dis ; 51(3): 663-7, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17992923

RESUMEN

Avian leukosis viruses (ALVs) are common in many poultry flocks and can be detected using an enzyme-linked immunosorbent assay or any other test designed to identify p27, the group-specific antigen located in gag. However, endogenous retroviruses expressing p27 are often present and can be confused with exogenous ALVs. A more specific and informative assay involves targeting the variable envelope glycoprotein gene (gp85) that is the basis for dividing ALVs into their different subgroups. We designed polymerase chain reaction (PCR) primers that would specifically detect and amplify viruses from each of the six ALV subgroups: A, B, C, D, E, and J. Subgroup B and D envelopes are related, and our B-specific primers also amplified subgroup D viruses. We also designed a set of common primers to amplify any ALV subgroup virus. To demonstrate the usefulness of these primers, we obtained from the Center for Veterinary Biologics in Iowa culture supernatant from chicken embryo fibroblasts infected with an ALV that was found to be a contaminant in two commercial Marek's disease vaccines. Using our PCR primers, we demonstrate that the contaminant was a subgroup A ALV. We cloned and sequenced a portion of the envelope gene and confirmed that the ALV was a subgroup A virus. Unlike typical subgroup A viruses, the contaminant ALV grew very slowly in cell culture. We also cloned and sequenced a portion of the long terminal repeat (LTR) from the contaminant virus. The LTR was found to be similar to those LTRs found in endogenous ALVs (subgroup E) and very dissimilar to LTRs normally found in subgroup A viruses. The E-like LTR probably explains why the contaminant grew so poorly in cell culture.


Asunto(s)
Virus de la Leucosis Aviar/aislamiento & purificación , Pollos/virología , Contaminación de Medicamentos , Enfermedad de Marek/prevención & control , Reacción en Cadena de la Polimerasa/veterinaria , Vacunas Virales/normas , Animales , Virus de la Leucosis Aviar/genética , Secuencia de Bases , Datos de Secuencia Molecular , Filogenia , Enfermedades de las Aves de Corral/prevención & control
17.
Avian Dis ; 51(4): 851-7, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18251393

RESUMEN

The unique open reading frame 11 (LORF11) of Marek's disease virus (MDV) is present in all three serotypes of MDV and is located in the unique long region of the MDV genome. In the serotype 1 Md5 genome, LORF11 comprises 2711 nucleotides and encodes a predicted protein of 903 amino acids. In order to study the biological function of LORF11 we deleted it from the MDV cosmid A6 by using the RecA-assisted restriction endonuclease cleavage method. The recombinant cosmid, A6DeltaLORF11, was transfected into duck embryo fibroblasts (DEF) in conjunction with parental SN5, P89, SN16, and B40 cosmid clones. Recombinant rMd5DeltaLORF11 plaques were evident at 12-13 days after transfection. Polymerase chain reaction amplification of DEF cells infected with rMd5DeltaLORF11 viruses confirmed the deletion of a 2.57-kb fragment resulting in a 296-bp fragment. Three rMd5DeltaLORF11 mutants were generated and their biological functions were studied in vitro and in vivo. In vitro growth characteristics of rMd5DeltaLORF11 viruses were similar to those of parental rMd5, indicating that LORF11 is not essential for replication in vitro. In vivo studies of rMd5DeltaLORF11 mutants showed that they were impaired in viral replication in the lymphoid organs and had 100x lower viremia than chickens infected with the parental rMd5 virus. Furthermore, rMd5-infected chickens horizontally transmitted the virus to contact controls whereas no horizontal transmission occurred in rMd5DeltaLORF11-infected chickens. Three independent deletion mutants were tested and showed the same phenotypes, so it is unlikely that the observed phenotype is because of any random mutation in the genome. Therefore the LORF11 gene of MDV is essential for normal virus replication in chickens and deletion of LORF11 renders an attenuated virus.


Asunto(s)
Genes Virales/genética , Herpesvirus Meleágrido 1/genética , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 3/genética , Animales , Pollos , Herpesvirus Meleágrido 1/patogenicidad , Herpesvirus Gallináceo 2/patogenicidad , Herpesvirus Gallináceo 3/patogenicidad , Enfermedad de Marek/virología , Mutación
18.
Chem Biol ; 12(1): 25-33, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15664512

RESUMEN

Aptamers (protein binding oligonucleotides) have potential as a new class of targeted therapeutics. For applications requiring chronic systemic administration, aptamers must achieve high-affinity target binding while simultaneously retaining high in vivo stability, tolerability, and ease of chemical synthesis. To this end, we describe a method for generating aptamers composed entirely of 2'-O-methyl nucleotides (mRmY). We present conditions under which 2'-O-methyl transcripts can be generated directly and use these conditions to select a fully 2'-O-methyl aptamer from a library of 3 x 10(15) unique 2'-O-methyl transcripts. This aptamer, ARC245, is 23 nucleotides in length, binds to vascular endothelial growth factor (VEGF) with a Kd of 2 nM, and inhibits VEGF activity in cellular assays. Notably, ARC245 is so stable that degradation cannot be detected after 96 hr in plasma at 37 degrees C or after autoclaving at 125 degrees C. We believe ARC245 has considerable potential as an antiangiogenesis therapeutic.


Asunto(s)
Oligonucleótidos/farmacología , Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Inhibidores de la Angiogénesis/química , Inhibidores de la Angiogénesis/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , ARN Polimerasas Dirigidas por ADN/metabolismo , Endotelio Vascular/efectos de los fármacos , Biblioteca de Genes , Humanos , Hidrólisis , Ratones , Oligonucleótidos/química , Oligonucleótidos/metabolismo , Factores de Tiempo , Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Virus Res ; 100(2): 243-8, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15019243

RESUMEN

We investigated whether chicken embryonic tissues are susceptible to infection with virulent Marek's disease virus (MDV). Groups of embryonic day (ED) 17 chicken embryos and 1-day-old chicks were compared for tissue sites of viral persistence of MDV and herpesvirus of turkeys (HVT) in lungs, thymuses, bursae of Fabricius and spleens. MDV DNA was detectable in the lungs and thymuses of embryos at 3 days post-inoculation (DPI) by in situ hybridization, while HVT DNA was only present in embryonic lungs. The target cells in lungs and thymuses appeared non-lymphoid and lymphoid, respectively. By 5 days post-inoculation, both viruses were detectable in all organs examined and persisted after hatch. Although MDV DNA was present in the embryo, there was little evidence of viral replication. These findings demonstrate the differences in pathogenesis of embryonic infection with MDV and HVT and provide evidence that the chicken embryo is susceptible to infection with a virulent avian herpesvirus.


Asunto(s)
Alphaherpesvirinae/aislamiento & purificación , Embrión de Pollo/virología , Infecciones por Herpesviridae/veterinaria , Animales , ADN Viral/análisis , Infecciones por Herpesviridae/transmisión , Pulmón/embriología , Pulmón/virología , Morfogénesis , Enfermedades de las Aves de Corral/transmisión , Enfermedades de las Aves de Corral/virología , Timo/embriología , Timo/virología
20.
Avian Dis ; 47(2): 425-32, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12887202

RESUMEN

We have recently described the isolation and molecular characteristics of two recombinant avian leukosis subgroup J viruses (ALV J) with an avian leukosis virus subgroup A envelope (r5701A and r6803A). In the present study, we examined the role of the subgroup A envelope in the pathogenesis of these recombinant viruses. Chickens of line 151(5) x 7(1) were inoculated at 1 day of age with r5701A, r6803A, Rous-associated virus type 1 (RAV-1), or strain ADOL-Hcl of ALV-J. At 2, 4, 10, 18, and 32 wk postinoculation (PI), chickens were tested for avian leukosis virus (ALV)-induced viremia, shedding, and neutralizing antibodies. All except one chicken inoculated with the recombinant viruses (98%) developed neutralizing antibodies by 10 wk PI compared with only 16% and 46% of the ADOL-Hcl and RAV-1-inoculated birds, respectively. ALV-induced tumors and mortality in the two groups inoculated with recombinant viruses were different. The incidence of tumors in groups inoculated with r5701A or RAV-1 was 100% compared with only 9% in the groups inoculated with r6803A or ADOL-Hcl. The data suggest that differences in pathogenicity between the two recombinant viruses might be due to differences in the sequence of the 3' untranslated region (presence or absence of the E element), and, therefore, not only the envelope but also other elements of the viral genome play an important role in the pathogenesis of ALV.


Asunto(s)
Virus de la Leucosis Aviar/genética , Virus de la Leucosis Aviar/patogenicidad , Leucosis Aviar/virología , ARN/genética , Animales , Anticuerpos Antivirales/inmunología , Leucosis Aviar/inmunología , Virus de la Leucosis Aviar/clasificación , Virus de la Leucosis Aviar/inmunología , Transformación Celular Viral , Pollos/inmunología , Pollos/virología , Femenino , Masculino , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Viremia , Virulencia , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA