Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Front Immunol ; 12: 784359, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35095857

RESUMEN

Marek's disease virus (MDV), the etiologic agent for Marek's disease (MD), causes a deadly lymphoproliferative disease in chickens. Causes of the well-documented association between genetically defined lines of chicken and resistance to MD remain unknown. Here, the frequencies of IFN-gamma producing pp38 and MEQ-specific T cell responses were determined in line N (B21 haplotype; MD-resistant) and line P2a (B19 haplotype, MD-susceptible) chickens after infection with vaccine and/or virulent (RB1B) strains of MDV using both standard ex vivo and cultured chIFN-gamma ELISPOT assays. Notably, MDV infection of naïve and vaccinated MD-resistant chickens induced higher frequencies of IFN-gamma producing MDV-specific T cell responses using the cultured and ex vivo ELISPOT assay, respectively. Remarkably, vaccination did not induce or boost MEQ-specific effector T cells in the susceptible chickens, while it boosted both pp38-and MEQ-specific response in resistant line. Taken together, our results revealed that there is a direct association between the magnitude of T cell responses to pp38 and MEQ of MDV antigens and resistance to the disease.


Asunto(s)
Pollos/inmunología , Haplotipos/inmunología , Antígenos de Histocompatibilidad/inmunología , Interferón gamma/inmunología , Mardivirus/inmunología , Enfermedad de Marek/inmunología , Linfocitos T/inmunología , Animales , Pollos/virología , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/virología , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Virulencia/inmunología
2.
Front Immunol ; 10: 3131, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-32063900

RESUMEN

Duck plague virus (DPV) is a representative pathogen transmitted among aquatic animals that causes gross lesions and immune inhibition in geese and ducks. The mechanism of organ tropism and innate immune evasion of DPV has not been completely deciphered due to a lack of cell models to study the innate immune manipulation and pathogenicity of aquatic viruses. In the present study, we isolated five types of duck primary cells [duck embryo fibroblasts (DEFs), neurons, astrocytes, peripheral blood mononuclear cells (PBMCs), and monocytes/macrophages] to identify appropriate cell models for DPV, using tropism infection and innate immunologic assays. Cells responded differently to stimulation with DNA viruses or RNA virus analogs. DPV infection exhibited broad tropism, as the recombinant virulent strain (CHv-GFP) infected DEFs, neurons, astrocytes, and monocytes/macrophages, but not the PBMCs, as the expression of EGFP was negligible. The basal levels of innate immunity molecules were highest in monocytes/macrophages and lower in DEFs and astrocytes. Conversely, the titer and genomic copy number of the attenuated virus strain was higher in DEFs and astrocytes than in neurons and monocytes/macrophages. The titer and genomic copy number of the attenuated virus strain were higher compared with the virulent strain in DEFs, neurons, and astrocytes. The innate immune response was not significantly induced by either DPV strain in DEFs, neurons, or astrocytes. The virulent strain persistently infected monocytes/macrophages, but the attenuated strain did so abortively, and this was accompanied by the phenomenon of innate immune inhibition and activation by the virulent and attenuated strains, respectively. Blockage of IFNAR signaling promoted replication of the attenuated strain. Pre-activation of IFNAR signaling inhibited infection by the virulent strain. The selection assay results indicated that induction of innate immunity plays an essential role in controlling DPV infection, and monocytes/macrophages are an important cell model for further investigations. Our study provided practical methods for isolating and culturing duck primary cells, and our results will facilitate further investigations of organ tropism, innate immune responses, latent infection, and the effectiveness of antiviral drugs for treating DPV and potentially other aerial bird pathogens.


Asunto(s)
Modelos Animales de Enfermedad , Patos/inmunología , Patos/virología , Enfermedad de Marek/inmunología , Enfermedad de Marek/virología , Animales , Astrocitos/inmunología , Astrocitos/virología , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Inmunidad Innata/inmunología , Macrófagos/inmunología , Macrófagos/virología , Mardivirus/inmunología , Monocitos/inmunología , Monocitos/virología , Neuronas/inmunología , Neuronas/virología
3.
Dev Comp Immunol ; 88: 183-189, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30025984

RESUMEN

DEAD (Asp-Glu-Ala-Asp) box polypeptide 41 (DDX41), a receptor belonging to DExD/H-box helicase family, acts as an intracellular DNA sensor and induces type I IFN production in mammals and fish. However, the function of avian DDX41 in innate immune response is still unknown. In this study, the full-length duck DDX41 (duDDX41) cDNA sequence was cloned for the first time and encoded a putative protein of 618 amino acid residues which showed the high sequence similarity with both zebra finch and chicken DDX41s. The duDDX41 mRNA was widely distributed in all tested tissues, especially the cerebrum, cerebellum, and liver. Overexpression of duDDX41 triggered the activation of transcription factors IRF1 and NF-κB, as well as IFN-ß expression in DEFs. The DEADc domain of duDDX41 played an extremely vital role in duck type I IFN signaling pathway. Knockdown of duDDX41 by siRNA silencing dramatically decreased IFN-ß expression stimulated by poly(dA:dT) or duck enteritis virus (DEV). In addition, the replication of DEV was significantly inhibited in duDDX41-expressed DEFs and was enhanced in DDX41 knockdown DEFs. These results suggest that DDX41 is an important cytosolic DNA sensor and plays a crucial role in duck antiviral innate immune response.


Asunto(s)
Proteínas Aviares/fisiología , ARN Helicasas DEAD-box/fisiología , Patos/inmunología , Inmunidad Innata/fisiología , Animales , Clonación Molecular , Citosol/inmunología , Citosol/metabolismo , ADN Complementario/genética , Patos/virología , Fibroblastos , Técnicas de Silenciamiento del Gen , Células HEK293 , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón beta/genética , Interferón beta/inmunología , Mardivirus/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Filogenia , Poli dA-dT , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Dominios Proteicos/inmunología , ARN Interferente Pequeño/metabolismo , Alineación de Secuencia , Transducción de Señal/genética , Transducción de Señal/inmunología
4.
Sci Rep ; 7(1): 1478, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28469192

RESUMEN

Duck enteritis virus (DEV), duck tembusu virus (DTMUV), and highly pathogenic avian influenza virus (HPAIV) H5N1 are the most important viral pathogens in ducks, as they cause significant economic losses in the duck industry. Development of a novel vaccine simultaneously effective against these three viruses is the most economical method for reducing losses. In the present study, by utilizing a clustered regularly interspaced short palindromic repeats (CRISPR)/associated 9 (Cas9)-mediated gene editing strategy, we efficiently generated DEV recombinants (C-KCE-HA/PrM-E) that simultaneously encode the hemagglutinin (HA) gene of HPAIV H5N1 and pre-membrane proteins (PrM), as well as the envelope glycoprotein (E) gene of DTMUV, and its potential as a trivalent vaccine was also evaluated. Ducks immunized with C-KCE-HA/PrM-E enhanced both humoral and cell-mediated immune responses to H5N1 and DTMUV. Importantly, a single-dose of C-KCE-HA/PrM-E conferred solid protection against virulent H5N1, DTMUV, and DEV challenges. In conclusion, these results demonstrated for the first time that the CRISPR/Cas9 system can be applied for modification of the DEV genome rapidly and efficiently, and that recombinant C-KCE-HA/PrM-E can serve as a potential candidate trivalent vaccine to prevent H5N1, DTMUV, and DEV infections in ducks.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Sistemas CRISPR-Cas , Infecciones por Flavivirus/prevención & control , Gripe Aviar/prevención & control , Enfermedad de Marek/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Vacunas Virales/genética , Animales , Anticuerpos Neutralizantes/biosíntesis , Protección Cruzada , Patos , Flavivirus/genética , Flavivirus/inmunología , Flavivirus/patogenicidad , Infecciones por Flavivirus/inmunología , Infecciones por Flavivirus/mortalidad , Infecciones por Flavivirus/virología , Edición Génica/métodos , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Gripe Aviar/inmunología , Gripe Aviar/mortalidad , Gripe Aviar/virología , Mardivirus/genética , Mardivirus/inmunología , Mardivirus/patogenicidad , Enfermedad de Marek/inmunología , Enfermedad de Marek/mortalidad , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/mortalidad , Enfermedades de las Aves de Corral/virología , Análisis de Supervivencia , Vacunación , Vacunas Sintéticas , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/biosíntesis
5.
Viruses ; 9(4)2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28368367

RESUMEN

Marek's disease (MD) virus (MDV) has been evolving continuously, leading to increasing vaccination failure. Here, the MDV field strain BS/15 was isolated from a severely diseased Chinese chicken flock previously vaccinated with CVI988. To explore the causes of vaccination failure, specific-pathogen free (SPF) chickens vaccinated with CVI988 or 814 and unvaccinated controls were challenged with either BS/15 or the reference strain Md5. Both strains induced MD lesions in unvaccinated chickens with similar mortality rates of 85.7% and 80.0% during the experimental period, respectively. However, unvaccinated chickens inoculated with BS/15 exhibited a higher tumor development rate (64.3% vs. 40.0%), but prolonged survival and diminished immune defects compared to Md5-challenged counterparts. These results suggest that BS/15 and Md5 show a similar virulence but manifest with different pathogenic characteristics. Moreover, the protective indices of CVI988 and 814 were 33.3 and 66.7 for BS/15, and 92.9 and 100 for Md5, respectively, indicating that neither vaccine could provide efficient protection against BS/15. Taken together, these data suggest that MD vaccination failure is probably due to the existence of variant MDV strains with known virulence and unexpected vaccine resistance. Our findings should be helpful for understanding the pathogenicity and evolution of MDV strains prevalent in China.


Asunto(s)
Mardivirus/inmunología , Mardivirus/aislamiento & purificación , Enfermedad de Marek/inmunología , Enfermedad de Marek/virología , Vacunas Virales/inmunología , Animales , Pollos , China , Mardivirus/genética , Mardivirus/patogenicidad , Enfermedad de Marek/prevención & control , Insuficiencia del Tratamiento , Virulencia
6.
J Virol Methods ; 236: 111-116, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27448823

RESUMEN

Quantitative RT-PCR (qRT-PCR) is widely used in the study of relative gene expression in general, and has been used in the field of Marek's disease (MD) research to measure transcriptional responses to infection and/or vaccination. Studies in the past have either employed cellular ß-actin (BACT) or glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as internal reference genes, although the stability of their expression in the context of Marek's disease virus (MDV) infection has never been investigated. In the present study, we compared the stability of five reference genes (BACT, 28S RNA, 18S RNA, GAPDH, Peptidyl-prolyl-isomerase B [PPIB], a.k.a. cyclophilin B) as standard internal controls in chicken embryo fibroblast (CEFs) cultures infected with either MD vaccine or oncogenic MDV1 viruses. We further extend these analyses to reference gene stability in spleen lymphomas induced by infection of commercial broiler chickens with a very virulent plus MDV1 (vv+ TK-2a virus). Two excel based algorithms, (Bestkeeper and Normfinder) were employed to compare reference gene stability. Bestkeeper and Normfinder analysis of reference gene stability in virus- and mock-infected cells, showed that 28S RNA and PPIB displayed higher stability in CEF infections with either oncogenic or vaccine viruses. In addition, both Bestkeeper and Normfinder determined 28S RNA and PPIB to be the most stably-expressed reference genes in vivo in vv+ TK-2a-induced spleen lymphomas. Furthermore, Bestkeeper and Normfinder analyses both determined BACT to be the least stable reference gene during MDV infection of CEF with oncogenic viruses, vaccine viruses, as well as in vv+ TK-2a-induced spleen lymphomas.


Asunto(s)
Perfilación de la Expresión Génica/normas , Interacciones Huésped-Patógeno , Mardivirus/inmunología , Enfermedad de Marek/patología , Enfermedades de las Aves de Corral/patología , Reacción en Cadena en Tiempo Real de la Polimerasa/normas , Estándares de Referencia , Animales , Células Cultivadas , Pollos , Fibroblastos/virología , Perfilación de la Expresión Génica/métodos , Mardivirus/patogenicidad , Enfermedades de las Aves de Corral/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
7.
Vet Microbiol ; 186: 189-98, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-27016773

RESUMEN

DNA vaccines are widely used to prevent and treat infectious diseases, cancer and autoimmune diseases; however, their relatively low immunogenicity is an obstacle to their use. In this study, we constructed a novel and universal DNA vaccine vector (pSS898) that can be used to build DNA vaccines against duck enteritis virus (DEV) and other viruses that require DNA vaccines to provide protection. This vaccine vector has many advantages, including innate immunogenicity, efficient nuclear trafficking and resistance to attack from nucleases. UL24 and tgB from DEV were chosen as the antigens, and the heat labile enterotoxin B subunit (LTB) from Escherichia coli and the IL-2 gene (DuIL-2) from duck were used as adjuvants for the construction of DNA vaccine plasmids. Ducklings that were orally immunized with S739 (Salmonella Typhimurium Δasd-66 Δcrp-24 Δcya-25) and harboring these DEV DNA vaccines produced strong mucosal and systemic immune responses, and they resisted an otherwise lethal DEV challenge. More importantly, S739 (UL24-LTB) provided 90% protection after a priming-boost immunization. This study shows that our novel and universal DNA vaccine vector can be used efficiently in practical applications and may provide a promising method of orally inoculating ducks with a DEV DNA vaccine delivered by attenuated Salmonella Typhimurium for prevention of DVE.


Asunto(s)
Patos/inmunología , Infecciones por Herpesviridae/veterinaria , Enfermedades de las Aves de Corral/prevención & control , Salmonella typhimurium/inmunología , Vacunación/métodos , Vacunas de ADN , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/inmunología , Células COS , Chlorocebus aethiops , Patos/genética , Patos/virología , Escherichia coli/genética , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/mortalidad , Infecciones por Herpesviridae/prevención & control , Inmunidad Mucosa/inmunología , Mardivirus/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/mortalidad , Enfermedades de las Aves de Corral/virología , Análisis de Supervivencia
8.
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
9.
Avian Pathol ; 44(4): 254-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25968878

RESUMEN

Marek's disease virus (MDV) is an oncogenic alphaherpesvirus and the causative agent of Marek's disease (MD), characterized by immunosuppression, paralysis, nerve enlargement and induction of T-cell lymphomas in chickens. Despite widespread usage of vaccines since the 1970s to control MD, more virulent field strains of MDV have emerged that overcome vaccinal protection, necessitating the development of new and more protective MD vaccines. The ∆Meq virus, a recombinant Md5 strain MDV lacking the viral oncogene Meq, is one candidate MD vaccine with great potential but unfortunately it also causes bursal-thymic atrophy (BTA) in maternal antibody negative chickens, raising concerns that impede commercial use as a vaccine. Previously, we identified a point mutation within UL5 that reduced in vivo replication in attenuated viruses. We proposed that introduction of the UL5 point mutation into the ∆Meq virus would reduce in vivo replication and eliminate BTA yet potentially retain high protective abilities. In birds, the ∆Meq+UL5 recombinant MDV had reduced replication compared to the original ∆Meq virus, while weights of lymphoid organs indicated that ∆Meq+UL5 did not induce BTA, supporting the hypothesis that reduction of in vivo replication would also abolish BTA. Vaccine trials of the ∆Meq+UL5 virus compared to other ∆Meq-based viruses and commercial vaccines show that, while the ∆Meq+UL5 does provide vaccinal protection, this protection was also reduced compared to the original ∆Meq virus. Therefore, it appears that a very delicate balance is required between levels of replication able to induce high vaccinal protection, yet not so high as to induce BTA.


Asunto(s)
Pollos/inmunología , ADN Helicasas/inmunología , ADN Primasa/inmunología , Mardivirus/inmunología , Enfermedad de Marek/inmunología , Enfermedades de las Aves de Corral/inmunología , Animales , Atrofia/veterinaria , ADN Helicasas/genética , ADN Primasa/genética , Mardivirus/patogenicidad , Enfermedad de Marek/prevención & control , Enfermedad de Marek/virología , Vacunas contra la Enfermedad de Marek/genética , Vacunas contra la Enfermedad de Marek/inmunología , Mutación Puntual , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Proteínas Virales/genética , Proteínas Virales/inmunología
10.
Intervirology ; 58(2): 95-105, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25677615

RESUMEN

Marek's disease virus (MDV) is a cell-associated alpha-herpesvirus that causes T-cell lymphomas and nervous disorders in chickens. Different from other lymphoid organs, the thymus is the site of T-cell maturation and differentiation. However, the transcriptional response to MDV infection in the chicken thymus is still not known. In this study, we performed genome-wide expression analysis in thymus tissues of RB1B-infected chickens at different time points to investigate the molecular mechanisms of MDV pathogenesis. The number of differentially expressed genes with 2-fold or higher changes (>2) are as follows: 1,250 genes (7 dpi), 834 genes (14 dpi), 1,958 genes (21 dpi), and 2,306 genes (28 dpi). Gene ontology enrichment analysis revealed that the upregulated genes were involved in immune and inflammatory response at 7 dpi; angiogenesis, cytoskeleton organization, cell adhesion, and signal transduction showed different expressions at 21 and 28 dpi. The expression pattern of 18 randomly selected genes was confirmed by real-time RT-PCR. Several differently expressed host genes associated with tumor development are discussed. We identified the global host-gene expression pattern in the thymus of chickens that responded to MDV infection. The present data may provide groundwork for future investigation in the biology and pathogenesis of MDV.


Asunto(s)
Perfilación de la Expresión Génica , Mardivirus/inmunología , Enfermedad de Marek/genética , Enfermedad de Marek/inmunología , Timo/virología , Animales , Pollos , Biología Computacional , Enfermedad de Marek/virología , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Aves de Corral , Reacción en Cadena en Tiempo Real de la Polimerasa , Timo/inmunología , Regulación hacia Arriba
11.
Vaccine ; 33(5): 596-603, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25562789

RESUMEN

Marek's disease virus (MDV) GX0101, which is a field strain with a naturally occurring insertion of the reticuloendotheliosis virus (REV) long terminal repeat (LTR) fragment, shows distinct biological activities. Deletion of the meq gene in GX0101 contributes to its complete loss of pathogenicity and oncogenicity in SPF chickens, but this virus has a kanamycin resistance gene (kan(r)) residual at the site of meq gene. In the present study, the kan(r) was knocked out and a meq-null virus with a good replicative ability termed SC9-1 was selected. In vivo studies showed that SC9-1 had no pathogenicity or tumorigenicity to chickens. There were no obvious impacts on chicken weight, immune organ index or antibody levels induced by avian influenza virus (AIV)/newcastle disease virus (NDV) inactivated vaccines compared with the control group. The SC9-1 virus provided superior protection than CVI988/Rispens vaccine in both SPF chickens and Hy-line brown chickens when challenged with a very virulent MDV (rMd5 strain). There was no obvious change in SC9-1 protection against MDV rMd5 in SPF chickens after 20 passages in chicken embryonic fibroblast cells (CEFs). In conclusion, SC9-1 is a safe and effective vaccine candidate for the prevention of Marek's disease.


Asunto(s)
Mardivirus/genética , Mardivirus/inmunología , Vacunas contra la Enfermedad de Marek/inmunología , Enfermedad de Marek/prevención & control , Virus de la Reticuloendoteliosis/genética , Animales , Células Cultivadas , Pollos , Fibroblastos/virología , Eliminación de Gen , Genes Virales , Inestabilidad Genómica , Vacunas contra la Enfermedad de Marek/administración & dosificación , Vacunas contra la Enfermedad de Marek/genética , Vacunas contra la Enfermedad de Marek/aislamiento & purificación , Recombinación Genética , Secuencias Repetidas Terminales , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/aislamiento & purificación , Cultivo de Virus
12.
Viruses ; 6(6): 2428-43, 2014 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-24956180

RESUMEN

Duck Tembusu virus (DTMUV) is a recently emerging pathogenic flavivirus that has resulted in a huge economic loss in the duck industry. However, no vaccine is currently available to control this pathogen. Consequently, a practical strategy to construct a vaccine against this pathogen should be determined. In this study, duck enteritis virus (DEV) was examined as a candidate vaccine vector to deliver the envelope (E) of DTMUV. A modified mini-F vector was inserted into the SORF3 and US2 gene junctions of the attenuated DEV vaccine strain C-KCE genome to generate an infectious bacterial artificial chromosome (BAC) of C-KCE (vBAC-C-KCE). The envelope (E) gene of DTMUV was inserted into the C-KCE genome through the mating-assisted genetically integrated cloning (MAGIC) strategy, resulting in the recombinant vector, pBAC-C-KCE-E. A bivalent vaccine C-KCE-E was generated by eliminating the BAC backbone. Immunofluorescence and western blot analysis results indicated that the E proteins were vigorously expressed in C-KCE-E-infected chicken embryo fibroblasts (CEFs). Duck experiments demonstrated that the insertion of the E gene did not alter the protective efficacy of C-KCE. Moreover, C-KCE-E-immunized ducks induced neutralization antibodies against DTMUV. These results demonstrated, for the first time, that recombinant C-KCE-E can serve as a potential bivalent vaccine against DEV and DTMUV.


Asunto(s)
Flavivirus/inmunología , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Mardivirus/genética , Mardivirus/inmunología , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Embrión de Pollo , Patos , Flavivirus/genética , Expresión Génica , Orden Génico , Inmunización , Enfermedades de las Aves de Corral/mortalidad , Enfermedades de las Aves de Corral/prevención & control , Potencia de la Vacuna , Proteínas del Envoltorio Viral/genética , Ensayo de Placa Viral , Vacunas Virales/genética , Vacunas Virales/inmunología
13.
PLoS One ; 9(4): e95093, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24736466

RESUMEN

DNA vaccine is a promising strategy for protection against virus infection. However, little is known on the efficacy of vaccination with two plasmids for expressing the glycoprotein D (gD) and glycoprotein B (gB) of duck enteritis virus (DEV) in inducing immune response and immunoprotection against virulent virus infection in Pekin ducks. In this study, two eukaryotic expressing plasmids of pcDNA3.1-gB and pcDNA3.1-gD were constructed. Following transfection, the gB and gD expressions in DF1 cells were detected. Groups of ducks were vaccinated with pcDNA3.1-gB and/or pcDNA3.1-gD, and boosted with the same vaccine on day 14 post primary vaccination. We found that intramuscular vaccinations with pcDNA3.1-gB and/or pcDNA3.1-gD, but not control plasmid, stimulated a high frequency of CD4+ and CD8+ T cells in Pekin ducks, particularly with both plasmids. Similarly, vaccination with these plasmids, particularly with both plasmids, promoted higher levels of neutralization antibodies against DEV in Pekin ducks. More importantly, vaccination with both plasmids significantly reduced the virulent DEV-induced mortality in Pekin ducks. Our data indicated that vaccination with plasmids for expressing both gB and gD induced potent cellular and humoral immunity against DEV in Pekin ducks. Therefore, this vaccination strategy may be used for the prevention of DEV infection in Pekin ducks.


Asunto(s)
Mardivirus/genética , Mardivirus/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Animales , Anticuerpos Antivirales/inmunología , Patos , Inmunidad Celular , Inmunidad Humoral , Inmunoglobulina G/inmunología , Vacunación
14.
Acta Virol ; 57(2): 265-70, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23600883

RESUMEN

Here we would like to provide a brief overview of the modern history of Marek΄s disease (MD) research with a focus on the most recent developments in experimental work and we will try to sum up their impact on the understanding of the biological properties of Marek΄s disease type 1 (MDV-1), the only representative of the Mardivirus genus causing fatal lymphoproliferative disease in poultry. We will also compare MDV-1 with other serologically-related poultry herpesviruses, Marek΄s disease virus type 2 (MDV-2) and herpesvirus of turkeys (HVT). Although MD was first described at the beginning of the last century, proper characterization of its biological impact on poultry production and utilization of molecular biology methods for detailed characterization of causative agent MDV-1 were introduced only in recent decades. However, many characteristics of MD infection, pathogenesis and vaccine protection mechanisms remain unclarified, though novel methods bring a challenge for better understanding of these unanswered questions.


Asunto(s)
Herpesvirus Gallináceo 2/genética , Mardivirus/genética , Enfermedad de Marek/genética , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/virología , Animales , Herpesvirus Gallináceo 2/inmunología , Mardivirus/inmunología , Enfermedad de Marek/inmunología , Enfermedad de Marek/prevención & control , Vacunas contra la Enfermedad de Marek/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/prevención & control
15.
Avian Pathol ; 41(6): 589-98, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23237372

RESUMEN

Lymphoblastoid cell lines 265(L) and 990(O) are monoclonal lymphomas, derived respectively from liver and ovarian tumours, generated in inbred P-line (MHC B(19)/B(19)) chickens infected with RB-1B strain of Marek's disease virus (MDV) and pRB-1B5 BAC clone respectively. These were inoculated into inbred, MDV-susceptible, P-line chickens by intra-venous or intra-abdominal routes. Additional groups of birds were vaccinated using 1000 plaque-forming units of CVI988 vaccine 8 days prior to inoculation of the cell lines. Non-vaccinated birds developed visceral Marek's disease tumours with an increased rate 30 to 60 days post inoculation. Vaccination prevented tumour and disease development in challenged birds. TCRß repertoire analysis by spectratyping and sequencing of the inoculum was used to track tumour identity in primary tumours and tumour cell lines derived from inoculated birds. These data revealed that the tumours were a consequence of de novo virus infection and not metastasis and expansion of the inoculated tumour cells. Moreover, the data showed that the two MDV-derived cell lines were not transplantable even in syngeneic P-line birds. The data also demonstrated the application of spectratyping as a tool to track tumour identity in lymphoma transplantation studies.


Asunto(s)
Pollos , Mardivirus/inmunología , Vacunas contra la Enfermedad de Marek , Enfermedad de Marek/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Animales , Línea Celular Tumoral , Femenino , Endogamia , Linfoma/inmunología , Linfoma/prevención & control , Linfoma/veterinaria , Enfermedad de Marek/inmunología , Enfermedades de las Aves de Corral/inmunología , Organismos Libres de Patógenos Específicos , Vacunación/veterinaria
16.
Poult Sci ; 91(9): 2130-8, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22912446

RESUMEN

Marek's disease is a viral neoplastic disease of chickens caused by Marek's disease virus (MDV). Gene expression patterns have been investigated at different MDV infection stages, but there is limited research about the late tumor transformation phase. In this experiment, 44K Agilent chicken genome-wide expression microarrays were used to profile differential expression in tumorous spleens (TS) from severely morbid chickens and apparently normal spleens from survivors (SS) after MDV infection and expression in noninfected spleens (NS) from controls. There were 4,317 differentially expressed (DE) genes in TS versus NS. However, no DE genes were detected in SS versus NS, suggesting that maintenance of, or return to, homeostasis of gene activity in survivor spleens. Downregulated genes in tumorous spleens mainly enriched in the cytokine-cytokine receptor interaction pathway, and commonly investigated genes in Marek's disease study, IL6, IL18, IFNA, and IFNG were nondifferentially expressed, which indicates host inflammatory response was impaired. The IL10 and TNFRSF8 genes were upregulated in tumorous spleens. We speculated that IL10 might be exploited by MDV to escape from host immune surveillance, as reported for Epstein-Barr virus, which stimulated T cells secreting IL10 to subvert immune response. Previous study reported that transcription from TNFRSF8 promoter could be enhanced by MDV oncogene Meq. In this study, the increased expression of TNFRSF8 indicated interaction between MDV and TNFRSF8, which might facilitate pathogenesis and tumor transformation. The expression of many members in IGF system was changed in tumorous compared with noninfected spleens. The downregulation of IGFBP7 was considered to be associated with MD lymphoma transformation. Gene expression change of multiple regulatory pathways indicated their involvements in facilitating tumor transformation.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mardivirus/inmunología , Enfermedad de Marek/virología , Neoplasias/virología , Neoplasias del Bazo/metabolismo , Animales , Pollos , Citocinas/genética , Citocinas/metabolismo , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Linfoma/genética , Linfoma/metabolismo , Enfermedad de Marek/inmunología , Enfermedad de Marek/patología , Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa/veterinaria , ARN/genética , ARN/metabolismo , Organismos Libres de Patógenos Específicos
17.
Virol J ; 9: 94, 2012 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-22612856

RESUMEN

BACKGROUND: An immunoinhibitory receptor, programmed death-1 (PD-1), and its ligand, programmed death-ligand 1 (PD-L1), are involved in immune evasion mechanisms for several pathogens causing chronic infections and for neoplastic diseases. However, little has been reported for the functions of these molecules in chickens. Thus, in this study, their expressions and roles were analyzed in chickens infected with Marek's disease virus (MDV), which induces immunosuppression in infected chickens. RESULTS: A chicken T cell line, Lee1, which constitutively produces IFN-γ was co-cultured with DF-1 cells, which is a spontaneously immortalized chicken fibroblast cell line, transiently expressing PD-L1, and the IFN-γ expression level was analyzed in the cell line by real-time RT-PCR. The IFN-γ expression was significantly decreased in Lee1 cells co-cultured with DF-1 cells expressing PD-L1. The expression level of PD-1 was increased in chickens at the early cytolytic phase of the MDV infection, while the PD-L1 expression level was increased at the latent phase. In addition, the expression levels of PD-1 and PD-L1 were increased at tumor lesions found in MDV-challenged chickens. The expressions levels of PD-1 and PD-L1 were also increased in the spleens and tumors derived from MDV-infected chickens in the field. CONCLUSIONS: We demonstrated that the chicken PD-1/PD-L1 pathway has immunoinhibitory functions, and PD-1 may be involved in MD pathogenesis at the early cytolytic phase of the MDV infection, whereas PD-L1 could contribute to the establishment and maintenance of MDV latency. We also observed the increased expressions of PD-1 and PD-L1 in tumors from MDV-infected chickens, suggesting that tumor cells transformed by MDV highly express PD-1 and PD-L1 and thereby could evade from immune responses of the host.


Asunto(s)
Antígeno B7-H1/inmunología , Mardivirus/inmunología , Mardivirus/patogenicidad , Enfermedad de Marek/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Animales , Línea Celular , Pollos , Técnicas de Cocultivo , Fibroblastos/virología , Perfilación de la Expresión Génica , Evasión Inmune , Tolerancia Inmunológica , Interferón gamma/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología
18.
Avian Dis ; 55(3): 384-90, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22017035

RESUMEN

Marek's disease (MD) is a lymphoproliferative disease of domestic chickens caused by a highly infectious, oncogenic alpha-herpesvirus known as Marek's disease virus (MDV). MD is presently controlled by vaccination. Current MD vaccines include attenuated serotype 1 strains (e.g., CVI988/Rispens), avirulent serotype 2 (SB-1), and serotype 3 (HVT) MDV strains. In addition, recombinant MDV strains have been developed as potential new and more efficient vaccines to sustain the success of MD control in poultry. One of the candidate recombinant MDV strains, named rMd5deltaMeq, was derived from Md5, a very virulent strain of MDV lacking the MDV oncogene Meq. Our earlier reports suggest that rMd5deltaMeq provided protection equally well or better than commonly used MD vaccines in experimental and commercial lines of chickens challenged with very virulent plus (vv+) strains of MDV. In this study, maternal antibody-positive (trial 1) and negative (trial 2) chickens from a series of relatively MD resistant lines were either vaccinated with the rMd5deltaMeq or CVI988/Rispens followed by infection of a vv+ strain of MDV, 648A, passage 10. This report presents experimental evidence that the rMd5deltaMeq protected significantly better than the CVI988/Rispens (P < 0.01) in the relatively resistant experimental lines of chickens challenged with the vv+ strain of MDV. Together with early reports, the rMd5deltaMeq appeared to provide better protection, comparing with the most efficacious commercially available vaccine, CVI988/Rispens, for control of MD in lines of chickens regardless of their genetic background.


Asunto(s)
Pollos , Mardivirus/inmunología , Vacunas contra la Enfermedad de Marek/inmunología , Enfermedad de Marek/prevención & control , Proteínas Oncogénicas Virales/genética , Enfermedades de las Aves de Corral/prevención & control , Animales , Mardivirus/genética , Enfermedad de Marek/inmunología , Vacunas contra la Enfermedad de Marek/genética , Proteínas Oncogénicas Virales/inmunología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/inmunología , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología
19.
J Virol ; 85(21): 11146-58, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865384

RESUMEN

Marek's disease virus (MDV) is a highly contagious oncogenic alphaherpesvirus that causes disease that is both a cancer model and a continuing threat to the world's poultry industry. This comprehensive gene expression study analyzes the host response to infection in both resistant and susceptible lines of chickens and inherent expression differences between the two lines following the infection of the host. A novel pathogenicity mechanism, involving the downregulation of genes containing HIC1 transcription factor binding sites as early as 4 days postinfection, was suggested from this analysis. HIC1 drives antitumor mechanisms, suggesting that MDV infection switches off genes involved in antitumor regulation several days before the expression of the MDV oncogene meq. The comparison of the gene expression data to previous QTL data identified several genes as candidates for involvement in resistance to MD. One of these genes, IRG1, was confirmed by single nucleotide polymorphism analysis to be involved in susceptibility. Its precise mechanism remains to be elucidated, although the analysis of gene expression data suggests it has a role in apoptosis. Understanding which genes are involved in susceptibility/resistance to MD and defining the pathological mechanisms of the disease gives us a much greater ability to try to reduce the incidence of this virus, which is costly to the poultry industry in terms of both animal welfare and economics.


Asunto(s)
Predisposición Genética a la Enfermedad , Factores de Transcripción de Tipo Kruppel/metabolismo , Mardivirus/inmunología , Mardivirus/patogenicidad , Enfermedad de Marek/genética , Enfermedad de Marek/inmunología , Animales , Pollos , Perfilación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Estados Unidos
20.
Virol J ; 8: 214, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21569289

RESUMEN

BACKGROUND: A DNA vaccine expressing glycoprotein C (gC) of duck plague virus (DPV) was evaluated for inducing immunity in ducks. The plasmid encoding gC of DPV was administered via intramuscular (IM) injection and gene gun bombardment. RESULTS: After immunization by both routes virus-specific serum antibody and T-cell responses developed. Vaccination of ducks by IM injection induced a stronger humoral, but weaker cell-mediated immune response. In contrast, a better cell-mediated immune response was achieved by using a gene gun to deliver DNA-coated gold beads to the epidermis with as little as 6 µg of DNA. CONCLUSIONS: This demonstrated that both routes of DNA inoculation can be used for eliciting virus-specific immune responses. Although DNA vaccine containing DPV gC is effective in both intramuscular injection and gene gun bombardment, the latter could induce significantly higher cell-mediated responses against DPV.


Asunto(s)
Mardivirus/genética , Mardivirus/inmunología , Enfermedades de las Aves de Corral/prevención & control , Vacunas de ADN/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Biolística , Patos , Inyecciones Intramusculares , Plásmidos , Enfermedades de las Aves de Corral/inmunología , Linfocitos T/inmunología , Vacunas de ADN/administración & dosificación , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA