Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
Vet Q ; 44(1): 1-12, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38726839

RESUMO

Duck plague (DP) is an acute, contagious and fatal disease, caused by duck enteritis virus (DEV), with worldwide distribution causing several outbreaks and posing severe economic losses. The present study was carried out with a goal of development of a live attenuated cell culture based DP vaccine using an Indian strain of DEV and evaluation of its safety, efficacy along with complete genome analysis. The live attenuated DP vaccine (DPvac/IVRI-19) was developed by serial propagation of a virulent isolate of DEV (DEV/India/IVRI-2016) in the chicken embryo fibroblast (CEF) primary cell culture. Adaptation of DEV in CEF cell culture was indicated by more rapid appearance of cytopathic effects (CPE) and gradual increase of virus titre, which reached up to 107.5 TCID50/mL after 41 passages. The safety, immunogenicity and efficacy of the vaccine were determined by immunization trials in ducklings. The DPvac/IVRI-19 was found to be avirulent and completely safe in the ducklings. Further, the vaccine induced both humoral and cell mediated immune responses and afforded 100% protection against the virulent DEV challenge. A comparison of the whole genome of DPvac/IVRI-19 (MZ911871) and DEV/India/IVRI-2016 (MZ824102) revealed significant number of mutations, which might be associated with viral attenuation. Phylogenetic tree of DEV/India/IVRI-2016 revealed its evolutionary relationship with other DEV isolates, but it formed a separate cluster with certain unique mutations. Thus, with the proven safety and 100% efficacy, the DPvac/IVRI-19 is suitable for large scale production with precisely pure form of vaccine and has potential utility at national and global levels.


Assuntos
Patos , Fibroblastos , Mardivirus , Doenças das Aves Domésticas , Vacinas Atenuadas , Vacinas Virais , Animais , Vacinas Atenuadas/imunologia , Patos/virologia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Fibroblastos/virologia , Embrião de Galinha , Vacinas Virais/imunologia , Mardivirus/imunologia , Mardivirus/patogenicidade , Infecções por Herpesviridae/veterinária , Infecções por Herpesviridae/prevenção & controle , Infecções por Herpesviridae/virologia , Índia
2.
Science ; 382(6676): 1245-1246, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38096277
3.
Genes (Basel) ; 12(12)2021 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-34946806

RESUMO

Marek's disease (MD) was an immunosuppression disease induced by Marek's disease virus (MDV). MD caused huge economic loss to the global poultry industry, but it also provided an ideal model for studying diseases induced by the oncogenic virus. Alternative splicing (AS) simultaneously produced different isoform transcripts, which are involved in various diseases and individual development. To investigate AS events in MD, RNA-Seq was performed in tumorous spleens (TS), spleens from the survivors (SS) without any lesion after MDV infection, and non-infected chicken spleens (NS). In this study, 32,703 and 25,217 AS events were identified in TS and SS groups with NS group as the control group, and 1198, 1204, and 348 differently expressed (DE) AS events (p-value < 0.05 and FDR < 0.05) were identified in TS vs. NS, TS vs. SS, SS vs. NS, respectively. Additionally, Function enrichment analysis showed that ubiquitin-mediated proteolysis, p53 signaling pathway, and phosphatidylinositol signaling system were significantly enriched (p-value < 0.05). Small structural variations including SNP and indel were analyzed based on RNA-Seq data, and it showed that the TS group possessed more variants on the splice site region than those in SS and NS groups, which might cause more AS events in the TS group. Combined with previous circRNA data, we found that 287 genes could produce both circular and linear RNAs, which suggested these genes were more active in MD lymphoma transformation. This study has expanded the understanding of the MDV infection process and provided new insights for further analysis of resistance/susceptibility mechanisms.


Assuntos
Processamento Alternativo/genética , Galinhas/genética , Galinhas/virologia , Doença de Marek/genética , Baço/virologia , Animais , Perfilação da Expressão Gênica/métodos , Mardivirus/patogenicidade , Doença de Marek/virologia , Polimorfismo de Nucleotídeo Único/genética , RNA/genética , Sítios de Splice de RNA/genética , RNA Circular/genética , Transdução de Sinais/genética
4.
Virus Res ; 295: 198320, 2021 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-33549641

RESUMO

To better understand the pathogenicity of duck plague virus (DPV). The DPV Chinese standard challenge strain (DPV CSC) was continuously passaged 20 times in duck embryo fibroblasts (DEFs). DPV F1 was lethal for 2-week-ducks, but DPV F10 and F20 were not lethal for 2-week ducks, the 528 bp in UL2 region of DPV F1-F20 was deleted, which suggested that the deletion in UL2 region was not related with the virulence of DPV. Compared with DPV F20 infected ducks, IL-8 in DPV F1 infected ducks was significantly upregulated, but IL-1, IL-2,IFNγ and MHC-II were significantly downregulated. ISKNV copies in DPV F10 and F20 infected ducks were lower than the DPV F1 infected ducks. These results showed that massive viruses replication, upregulation of IL-8 expresssion, repression of IL-1, IL-2, IFNγ and MHC-II expression resulted in serious lesions and high mortality. This study provided a in-depth understanding of the immune-related genes expression in the different virulence of DPV.


Assuntos
Citocinas , Patos , Mardivirus , Animais , Citocinas/genética , Patos/virologia , Interleucina-1 , Interleucina-2 , Interleucina-8/genética , Mardivirus/patogenicidade
5.
Poult Sci ; 99(12): 6647-6652, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33248580

RESUMO

To determine the role of glycoprotein I (gI) in duck plague virus (DPV), a gI-deleted mutant (BAC-CHv-ΔgI) and a gI-revertant virus (BAC-CHv-ΔgI Rev) were constructed by using a markerless two-step Red recombination system implemented on the DPV genome cloned into a bacterial artificial chromosome (BAC). Mutants were characterized on duck embryo fibroblast (DEF) cells compared with wild-type virus. BAC-CHv-ΔgI produced viral plaques on DEF cells that were on average approximately 57.2% smaller than those produced by BAC-CHv-ΔgI Rev and wild-type virus. Electron microscopy confirmed that deleting of gI resulted in nucleocapsids accumulated around the cytoplasm vesicles and few of them could complete the final envelopment process. These results clearly indicated that DPV gI plays significant roles in viral cell-cell spread and viral final envelopment process.


Assuntos
Patos , Glicoproteínas , Mardivirus , Doença de Marek , Animais , Células Cultivadas , Cromossomos Artificiais Bacterianos/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Mardivirus/genética , Mardivirus/patogenicidade , Doença de Marek/transmissão , Doença de Marek/virologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
6.
Virol J ; 17(1): 186, 2020 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-33228722

RESUMO

BACKGROUND: Marek's disease virus (MDV) causes malignant lymphomas in chickens (Marek's disease, MD). MD is currently controlled by vaccination; however, MDV strains have a tendency to develop increased virulence. Distinct diversity and point mutations are present in the Meq proteins, the oncoproteins of MDV, suggesting that changes in protein function induced by amino acid substitutions might affect MDV virulence. We previously reported that recent MDV isolates in Japan display distinct mutations in Meq proteins from those observed in traditional MDV isolates in Japan, but similar to those in MDV strains isolated from other countries. METHODS: To further investigate the genetic characteristics in Japanese field strains, we sequenced the whole genome of an MDV strain that was successfully isolated from a chicken with MD in Japan. A phylogenetic analysis of the meq gene was also performed. RESULTS: Phylogenetic analysis revealed that the Meq proteins in most of the Japanese isolates were similar to those of Chinese and European strains, and the genomic sequence of the Japanese strain was classified into the Eurasian cluster. Comparison of coding region sequences among the Japanese strain and MDV strains from other countries revealed that the genetic characteristics of the Japanese strain were similar to those of Chinese and European strains. CONCLUSIONS: The MDV strains distributed in Asian and European countries including Japan seem to be genetically closer to each other than to MDV strains from North America. These findings indicate that the genetic diversities of MDV strains that emerged may have been dependent on the different vaccination-based control approaches.


Assuntos
Galinhas/virologia , Mardivirus/genética , Mardivirus/isolamento & purificação , Doença de Marek/virologia , Filogenia , Doenças das Aves Domésticas/virologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , China , Europa (Continente) , Variação Genética , Genoma Viral , Japão , Mardivirus/classificação , Mardivirus/patogenicidade , Mutação , Proteínas Oncogênicas Virais/genética , Virulência , Sequenciamento Completo do Genoma
7.
Sci Rep ; 9(1): 14182, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31578366

RESUMO

Very virulent plus Marek's disease (MD) virus (vv + MDV) induces tumors in relatively resistant lines of chickens and early mortality in highly susceptible lines of chickens. The vv + MDV also triggers a series of cellular responses in both types of chickens. We challenged birds sampled from a highly inbred chicken line (line 63) that is relatively resistant to MD and from another inbred line (line 72) that is highly susceptible to MD with a vv + MDV. RNA-sequencing analysis was performed with samples extracted from spleen tissues taken at 10-day and 21-day post infection (dpi). A total of 64 and 106 differentially expressed genes was identified in response to the vv + MDV challenge at latent phase in the resistant and susceptible lines of chickens, respectively. Direct comparisons between samples of the two lines identified 90 and 126 differentially expressed genes for control and MDV challenged groups, respectively. The differentially expressed gene profiles illustrated that intensive defense responses were significantly induced by vv + MDV at 10 dpi and 21 dpi but with slight changes in the resistant line. In contrast, vv + MDV induced a measurable suppression of gene expression associated with host defense at 10 dpi but followed by an apparent activation of the defense response at 21 dpi in the susceptible line of chickens. The observed difference in gene expression between the two genetic lines of chickens in response to MDV challenge during the latent phase provided a piece of indirect evidence that time points for MDV reactivation differ between the genetic lines of chickens with different levels of genetic resistance to MD. Early MDV reactivation might be necessary and potent to host defense system readiness for damage control of tumorigenesis and disease progression, which consequently results in measurable differences in phenotypic characteristics including early mortality (8 to 20 dpi) and tumor incidence between the resistant and susceptible lines of chickens. Combining differential gene expression patterns with reported GO function terms and quantitative trait loci, a total of 27 top genes was selected as highly promising candidate genes for genetic resistance to MD. These genes are functionally involved with virus process (F13A1 and HSP90AB1), immunity (ABCB1LB, RGS5, C10ORF58, OSF-2, MMP7, CXCL12, GAL1, GAL2, GAL7, HVCN1, PDE4D, IL4I1, PARP9, EOMES, MPEG1, PDK4, CCLI10, K60 and FST), and tumor suppression (ADAMTS2, LXN, ARRDC3, WNT7A, CLDN1 and HPGD). It is anticipated that these findings will facilitate advancement in the fundamental understanding on mechanisms of genetic resistance to MD. In addition, such advancement may also provide insights on tumor virus-induced tumorigenesis in general and help the research community recognize MD study may serve as a good model for oncology study involving tumor viruses.


Assuntos
Proteínas Aviárias/genética , Citocinas/genética , Mardivirus/patogenicidade , Doença de Marek/genética , Transcriptoma , Proteínas Supressoras de Tumor/genética , Animais , Proteínas Aviárias/metabolismo , Galinhas , Citocinas/metabolismo , Doença de Marek/imunologia , Doença de Marek/virologia , Baço/metabolismo , Baço/virologia , Proteínas Supressoras de Tumor/metabolismo
8.
J Virol ; 93(17)2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31189706

RESUMO

MicroRNAs (miRNAs) are small noncoding RNAs with profound regulatory roles in many areas of biology, including cancer. MicroRNA 155 (miR-155), one of the extensively studied multifunctional miRNAs, is important in several human malignancies such as diffuse large B cell lymphoma and chronic lymphocytic leukemia. Moreover, miR-155 orthologs KSHV-miR-K12-11 and MDV-miR-M4, encoded by Kaposi's sarcoma-associated herpesvirus (KSHV) and Marek's disease virus (MDV), respectively, are also involved in oncogenesis. In MDV-induced T-cell lymphomas and in lymphoblastoid cell lines derived from them, MDV-miR-M4 is highly expressed. Using excellent disease models of infection in natural avian hosts, we showed previously that MDV-miR-M4 is critical for the induction of T-cell lymphomas as mutant viruses with precise deletions were significantly compromised in their oncogenicity. However, those studies did not elucidate whether continued expression of MDV-miR-M4 is essential for maintaining the transformed phenotype of tumor cells. Here using an in situ CRISPR/Cas9 editing approach, we deleted MDV-miR-M4 from the MDV-induced lymphoma-derived lymphoblastoid cell line MDCC-HP8. Precise deletion of MDV-miR-M4 was confirmed by PCR, sequencing, quantitative reverse transcription-PCR (qRT-PCR), and functional analysis. Continued proliferation of the MDV-miR-M4-deleted cell lines demonstrated that MDV-miR-M4 expression is not essential for maintaining the transformed phenotype, despite its initial critical role in the induction of lymphomas. Ability to examine the direct role of oncogenic miRNAs in situ in tumor cell lines is valuable in delineating distinct determinants and pathways associated with the induction or maintenance of transformation in cancer cells and will also contribute significantly to gaining further insights into the biology of oncogenic herpesviruses.IMPORTANCE Marek's disease virus (MDV) is an alphaherpesvirus associated with Marek's disease (MD), a highly contagious neoplastic disease of chickens. MD serves as an excellent model for studying virus-induced T-cell lymphomas in the natural chicken hosts. Among the limited set of genes associated with MD oncogenicity, MDV-miR-M4, a highly expressed viral ortholog of the oncogenic miR-155, has received extensive attention due to its direct role in the induction of lymphomas. Using a targeted CRISPR-Cas9-based gene editing approach in MDV-transformed lymphoblastoid cell lines, we show that MDV-miR-M4, despite its critical role in the induction of tumors, is not essential for maintaining the transformed phenotype and continuous proliferation. As far as we know, this was the first study in which precise editing of an oncogenic miRNA was carried out in situ in MD lymphoma-derived cell lines to demonstrate that it is not essential in maintaining the transformed phenotype.


Assuntos
Transformação Celular Viral/genética , Linfoma/virologia , Mardivirus/patogenicidade , MicroRNAs/genética , Animais , Sistemas CRISPR-Cas , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Mardivirus/genética , RNA Viral/genética
9.
BMC Genomics ; 20(1): 245, 2019 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-30922224

RESUMO

BACKGROUND: Marek's disease virus (MDV) is an oncogenic herpesvirus that can cause T-cell lymphomas in chicken. Long noncoding RNA (lncRNA) is strongly associated with various cancers and many other diseases. In chickens, lncRNAs have not been comprehensively identified. Here, we profiled mRNA and lncRNA repertoires in three groups of spleens from MDV-infected and non-infected chickens, including seven tumorous spleens (TS) from MDV-infected chickens, five spleens from the survivors (SS) without lesions after MDV infection, and five spleens from noninfected chickens (NS), to explore the underlying mechanism of host resistance in Marek's disease (MD). RESULTS: By using a precise lncRNA identification pipeline, we identified 1315 putative lncRNAs and 1166 known lncRNAs in spleen tissue. Genomic features of putative lncRNAs were characterized. Differentially expressed (DE) mRNAs, putative lncRNAs, and known lncRNAs were profiled among three groups. We found that several specific intergroup differentially expressed genes were involved in important biological processes and pathways, including B cell activation and the Wnt signaling pathway; some of these genes were also found to be the hub genes in the co-expression network analyzed by WGCNA. Network analysis depicted both intergenic correlation and correlation between genes and MD traits. Five DE lncRNAs including MSTRG.360.1, MSTRG.6725.1, MSTRG.6754.1, MSTRG.15539.1, and MSTRG.7747.5 strongly correlated with MD-resistant candidate genes, such as IGF-I, CTLA4, HDAC9, SWAP70, CD72, JCHAIN, CXCL12, and CD8B, suggesting that lncRNAs may affect MD resistance and tumorigenesis in chicken spleens through their target genes. CONCLUSIONS: Our results provide both transcriptomic and epigenetic insights on MD resistance and its pathological mechanism. The comprehensive lncRNA and mRNA transcriptomes in MDV-infected chicken spleens were profiled. Co-expression analysis identified integrated lncRNA-mRNA and gene-gene interaction networks, implying that hub genes or lncRNAs exert critical influence on MD resistance and tumorigenesis.


Assuntos
Galinhas/genética , Resistência à Doença , Perfilação da Expressão Gênica/veterinária , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Baço/virologia , Animais , Epigenômica , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Masculino , Mardivirus/patogenicidade , Análise de Sequência de RNA , Baço/química , Via de Sinalização Wnt
10.
Viruses ; 11(2)2019 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-30813500

RESUMO

BACKGROUND: Duck plague virus (DPV) can induce apoptosis in duck embryo fibroblasts (DEFs) and in infected ducks, but the molecular mechanism of DPV-induced apoptosis remains unknown. METHODS: We first used qRT-PCR and a Caspase-Glo assay to determine whether the caspase protein family plays an important role in DPV-induced apoptosis. Then, we used an intracellular ROS detection kit and the mitochondrial probe JC-1 to respectively detect ROS levels and mitochondrial membrane potential (MMP). Finally, flow cytometry was used to detect apoptosis and cell cycle progression. RESULTS: In this study, the mRNA levels and enzymatic activities of caspase-3, caspase-7, caspase-8, and caspase-9 were significantly increased during DPV-induced apoptosis. The caspase inhibitors Z-DEVD-FMK, Z-LEHD-FMK, and Q-VD-OphA could inhibit DPV-induced apoptosis and promote viral replication. Subsequently, a significant decrease in MMP and an increase in the intracellular ROS levels were observed. Further study showed that pretreating infected cells with NAC (a ROS scavenger) decreased the intracellular ROS levels, increased the MMP, inhibited apoptosis, and promoted viral replication. Finally, we showed that DPV infection can cause cell cycle S-phase arrest. CONCLUSIONS: This study shows that DPV causes cell cycle S-phase arrest and leads to apoptosis through caspase activation and increased intracellular ROS levels. These findings may be useful for gaining an understanding of the pathogenesis of DPV and the apoptotic pathways induced by α-herpesviruses.


Assuntos
Apoptose , Caspases/metabolismo , Pontos de Checagem do Ciclo Celular , Mardivirus/patogenicidade , Espécies Reativas de Oxigênio/metabolismo , Animais , Caspases/genética , Células Cultivadas , Patos , Embrião não Mamífero/citologia , Fibroblastos/virologia , Potencial da Membrana Mitocondrial , Mitocôndrias/metabolismo , Reação em Cadeia da Polimerase
11.
Sci Rep ; 8(1): 16510, 2018 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-30405139

RESUMO

To investigate the function of the duck enteritis virus (DEV) tegument protein US10, we generated US10 deletion and revertant mutants (ΔUS10 and US10FRT) via two-step RED recombination based on an infectious BAC clone of DEV CHv-BAC-G (BAC-G). In multistep growth kinetic analyses, ΔUS10 showed an approximately 100-fold reduction in viral titer, while the genome copies decreased only 4-fold compared to those of BAC-G. In one-step growth kinetic analyses, there were no significant differences in genome copies among BAC-G, ΔUS10 and US10FRT, but ΔUS10 still showed a 5- to 20-fold reduction in viral titer, and the replication defect of ΔUS10 was partially reversed by infection of US10-expressing cells. The transcription levels of Mx, OASL, IL-4, IL-6 and IL-10 in ΔUS10-infected duck embryo fibroblasts (DEFs) were significantly upregulated, while TLR3 was downregulated compared with those in BAC-G-infected DEFs. Taken together, these data indicated that US10 is vital for DEV replication and is associated with transcription of some immunity genes.


Assuntos
Doenças das Aves/virologia , Patos/virologia , Enterite/veterinária , Mardivirus/genética , Proteínas Virais/genética , Animais , Doenças das Aves/imunologia , Linhagem Celular , Deleção de Genes , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno/imunologia , Mardivirus/imunologia , Mardivirus/metabolismo , Mardivirus/patogenicidade , Fases de Leitura Aberta , Proteínas Virais/metabolismo , Replicação Viral/genética
12.
Viruses ; 10(5)2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29783672

RESUMO

Superinfection of Marek's disease virus (MDV) and avian leukosis virus subgroup J (ALV-J) causes lethal neoplasia and death in chickens. However, whether there is synergism between the two viruses in viral replication and pathogenicity has remained elusive. In this study, we found that the superinfection of MDV and ALV-J increased the viral replication of the two viruses in RNA and protein level, and synergistically promoted the expression of IL-10, IL-6, and TGF-ß in chicken embryo fibroblasts (CEF). Moreover, MDV and ALV-J protein expression in dual-infected cells detected by confocal laser scanning microscope appeared earlier in the cytoplasm and the nucleus, and caused more severe cytopathy than single infection, suggesting that synergistically increased MDV and ALV-J viral-protein biosynthesis is responsible for the severe cytopathy. In vivo, compared to the single virus infected chickens, the mortality and tumor formation rates increased significantly in MDV and ALV-J dual-infected chickens. Viral loads of MDV and ALV-J in tissues of dual-infected chickens were significantly higher than those of single-infected chickens. Histopathology observation showed that more severe inflammation and tumor cells metastases were present in dual-infected chickens. In the present study, we concluded that synergistic viral replication of MDV and ALV-J is responsible for the enhanced pathogenicity in superinfection of chickens.


Assuntos
Vírus da Leucose Aviária/patogenicidade , Mardivirus/patogenicidade , Superinfecção/virologia , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/fisiologia , Galinhas/virologia , Mediadores da Inflamação/metabolismo , Mardivirus/fisiologia , Doença de Marek/virologia , Carga Viral , Virulência , Replicação Viral
13.
Avian Pathol ; 47(1): 47-57, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28762757

RESUMO

Marek's disease is a lymphoproliferative disease causing a serious threat in poultry production. Field strains of Marek's disease virus (MDVs) are continuously re-emerging, causing great economical losses to the poultry industry worldwide in spite of the intensive vaccination and restrictive management policy used. Histopathological and molecular characterizations of MDVs are essential for monitoring the changes of viruses and evaluating the effectiveness of existing vaccines. During 2016, 190 visceral tumour tissues representing 30 vaccinated chicken flocks from the Gifu prefecture, Japan, were analysed. A pathological examination revealed the presence of lymphoproliferative lesions in the visceral organs. Polymerase chain reaction screening of tissue specimens using specific primers for avian leucosis virus, reticuloendotheliosis virus, and MDV was positive only for MDV. The polymerase chain reaction products of meq, pp38, virus-induced IL-8 homology, and glycoprotein MDV genes were sequenced and used for homology, phylogenetic, and similarity level analysis with the published reference of MDVs in the database. The results revealed high similarity between the field isolates, vv and vv+ strains of MDV from the USA and China. Several point mutations in the nucleotide sequence of the field isolates and their deduced amino acid sequences were detected in those genes. The present molecular analyses indicated that nucleotide and amino acid changes could be valuable criteria for differentiation and determination of the pathogenicity and oncogenicity of MDVs according to the Avian Disease and Oncology Laboratory pathotyping in vivo studies. Furthermore, the results suggest that development of a new vaccine must be considered to overcome this devastating avian oncogenic viral disease.


Assuntos
Mardivirus/genética , Doença de Marek/virologia , Filogenia , Doenças das Aves Domésticas/virologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Galinhas , DNA Viral/genética , Regulação Viral da Expressão Gênica/fisiologia , Japão/epidemiologia , Mardivirus/patogenicidade , Doença de Marek/epidemiologia , Reação em Cadeia da Polimerase/métodos , Reação em Cadeia da Polimerase/veterinária , Doenças das Aves Domésticas/epidemiologia , RNA Viral/genética , Proteínas Virais/genética , Proteínas Virais/metabolismo , Virulência
14.
Cytokine ; 102: 191-199, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28969942

RESUMO

The human stimulator of interferon gene (STING) is an important molecule in innate immunity that stimulates type I interferon (IFN) production. However, the role of duck STING (duSTING) in innate immunity has yet to be explained. In this study, the full length of the duSTING cDNA sequence (1149bp), which encodes 382 amino acid (aa) residues, was reported and showed the highest sequence similarity with chicken STINGs. The phylogenetic analysis based on STING aa showed that duSTING was grouped onto the birds clade. According to the tissue distribution spectrum analysis, duSTING was highly present in the bursa of Fabricius, glandular stomach, liver, pancreas, and small intestine of ducklings, as well as in the blood and pancreas of the adult duck. DuSTING mainly colocalized with the endoplasmic reticulum (ER) and mitochondria in transfected Baby Hamster Syrian Kidney (BHK21) and duck embryo fibroblasts (DEF) cells by an indirect immunofluorescence assay. The transfection of the DEFs with duSTING activated NF-κB, which induced the transcription of IFN-ß, and the activated IFN induced the interferon-stimulated response element (ISRE). Furthermore, the overexpression of duSTING significantly upregulated the mRNA level of duck IFN-ß and IFN-stimulated genes (ISGs), such as duMx and duOASL and inhibited the replication of the double-stranded DNA duck plague virus (DPV) in vitro. In addition, the knockdown of endogenous duSTING by shRNA significantly reduced the poly (I:C) (pIC), poly (dA:dT), and Tembusu virus (TMUV), induced IFN-ß production and significantly promoted DPV replication in vitro. In general, these data demonstrate that duSTING is vital for duck type I interferon induction and plays an important role in the host defence of DPV infection.


Assuntos
Doenças das Aves/genética , Doenças das Aves/imunologia , Patos/genética , Patos/imunologia , Mardivirus/imunologia , Mardivirus/patogenicidade , Doença de Marek/genética , Doença de Marek/imunologia , Sequência de Aminoácidos , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/imunologia , Doenças das Aves/virologia , Patos/virologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , Interferon beta/genética , Mardivirus/fisiologia , Doença de Marek/virologia , NF-kappa B/metabolismo , Filogenia , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Replicação Viral
15.
Artigo em Inglês | MEDLINE | ID: mdl-29018776

RESUMO

Duck enteritis virus (DEV) is a large, complex double-stranded DNA virus that induces duck embryo fibroblast (DEF) cells autophagy, which is beneficial to its own replication, but the mechanism has not been described. In this study, we showed that impaired cell energy metabolism is involved in DEV-induced autophagy, whereby ATP synthesis is disrupted in cells after DEV infection, which causes metabolic stress and activation of autophagy. Methyl pyruvate (MP) inhibited conversion of LC3I to LC3II and accumulation of GFP-LC3, which could reverse the energy loss caused by DEV infection. Inhibition of DEV replication by MP confirmed the above view. We found that infection with DEV activated the metabolic regulator 5' AMP-activated kinase (AMPK) and inhibited activity of mechanistic target of rapamycin (mTOR). In the cases where AMPK expression was inhibited, the LC3-I conversion to LC3-II ratio was decreased, as was GFP-LC3 point and DEV replication; in addition, inhibition of p-mTOR showed a reverse trend. We also found that tuberous sclerosis (TSC) 2 was a key mediator between AMPK and mTOR through activation by phosphorylation. siRNA targeting TSC2 was transfected to reverse the inhibition of mTOR, and down-regulate autophagy level and DEV replication, but AMPK expression was not changed, while siRNA targeting AMPK inhibited activation of TSC2. In conclusion, our findings indicate that energy metabolism in cell damage induced by DEV contributes to autophagy via the AMPK-TSC2-MTOR signaling pathway, which provides a new perspective for DEV and host interactions.


Assuntos
Autofagia/genética , Metabolismo Energético/fisiologia , Enterite/virologia , Mardivirus/fisiologia , Proteínas Quinases/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular , Patos , Fibroblastos/virologia , Mardivirus/patogenicidade , Fosforilação , Piruvatos/metabolismo , RNA Interferente Pequeno/metabolismo , Replicação Viral
16.
J Virol Methods ; 249: 66-68, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28860100

RESUMO

Sequence analysis of duck plague virus (DPV) revealed that there was a 528bp (B fragment) deletion within the UL2 gene of DPV attenuated vaccine strain in comparison with field virulent strains. The finding of gene deletion provides a potential differentiation test between DPV virulent strain and attenuated strain based on their UL2 gene sizes. Thus we developed a polymerase chain reaction (PCR) assay targeting to the DPV UL2 gene for simultaneous detection of DPV virulent strain and attenuated strain, 827bp for virulent strain and 299bp for attenuated strain. This newly developed PCR for DPV was highly sensitive and specific. It detected as low as 100fg of DNA on both DPV virulent and attenuated strains, no same size bands were amplified from other duck viruses including duck paramyxovirus, duck tembusu virus, duck circovirus, Muscovy duck parvovirus, duck hepatitis virus type I, avian influenza virus and gosling plague virus. Therefore, this PCR assay can be used for the rapid, sensitive and specific detection of DPV virulent and attenuated strains affecting ducks.


Assuntos
Doenças das Aves/virologia , Patos/virologia , Infecções por Herpesviridae/veterinária , Mardivirus/isolamento & purificação , Mardivirus/patogenicidade , Reação em Cadeia da Polimerase/métodos , Animais , Doenças das Aves/diagnóstico , Infecções por Herpesviridae/diagnóstico , Infecções por Herpesviridae/virologia , Sensibilidade e Especificidade , Vacinas Atenuadas , Proteínas Virais/genética
17.
Arch Virol ; 162(11): 3549-3550, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28730521

RESUMO

Here, we present the complete genomic sequence of an attenuated duck enteritis virus (DEV). The Chinese standard challenge strain of DEV (DEV CSC) was serially passaged 20 times in chick embryo fibroblasts and then 85 times in chick embryos. The virus was attenuated and was avirulent to 2-month-old ducks. The attenuated DEV genome is 162,131 base pairs (bp) in length and as long as the parental genomic sequence. There are only 22 nucleotide substitutions, resulting in single amino acid changes in open reading frames LORF5, LORF4, UL41, UL39, UL32, UL13, UL10, UL3, US3, US4 and US7. The genome sequence has been deposited in the GenBank database under accession number KU216226. This study provides genetic information about DEV attenuation and further advances our understanding of the molecular basis of DEV pathogenesis.


Assuntos
Patos/virologia , Genoma Viral , Mardivirus/fisiologia , Animais , Células Cultivadas , Embrião de Galinha , DNA Viral/genética , Enterite/veterinária , Enterite/virologia , Fibroblastos/fisiologia , Mardivirus/patogenicidade , Doenças das Aves Domésticas/virologia , Virulência
18.
Avian Pathol ; 46(6): 652-657, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28609137

RESUMO

A comparison of the unique long region 2 (UL2) gene sequences between 10 virulent and 11 attenuated duck plague virus (DPV) strains (including all DPV UL2 gene sequences registered in GenBank) showed that the UL2 genes in the attenuated DPV strains had a 528 bp deletion in the B fragment. Primers were designed based on the B fragment sequence of the UL2 gene in an attempt to establish a fluorescence quantitative polymerase chain reaction (PCR) and a conventional PCR detection method that could specifically detect virulent DPV strains (i.e. positive detection for virulent DPV strains and negative detection for attenuated DPV strains). Additionally, PCR products were cloned for sequence analysis. These two methods detected five attenuated DPV strains in addition to the virulent DPV strains. Sequence analysis of the PCR products showed that the amplified products were the B fragments of the UL2 gene. These results indicated that detection methods specific for virulent DPV strains could not be established using primers designed based on the UL2 gene B fragment.


Assuntos
Patos/virologia , Enterite/veterinária , Mardivirus/isolamento & purificação , Doença de Marek/diagnóstico , Reação em Cadeia da Polimerase/veterinária , Uracila-DNA Glicosidase/genética , Animais , Enterite/diagnóstico , Enterite/virologia , Mardivirus/genética , Mardivirus/patogenicidade , Doença de Marek/virologia , Sensibilidade e Especificidade , Alinhamento de Sequência/veterinária , Análise de Sequência de DNA/veterinária , Proteínas Virais/genética
19.
Sci Rep ; 7(1): 1478, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28469192

RESUMO

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.


Assuntos
Anticorpos Antivirais/biossíntese , Sistemas CRISPR-Cas , Infecções por Flavivirus/prevenção & controle , Influenza Aviária/prevenção & controle , Doença de Marek/prevenção & controle , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais/genética , Animais , Anticorpos Neutralizantes/biossíntese , Proteção Cruzada , Patos , Flavivirus/genética , Flavivirus/imunologia , Flavivirus/patogenicidade , Infecções por Flavivirus/imunologia , Infecções por Flavivirus/mortalidade , Infecções por Flavivirus/virologia , Edição de Genes/métodos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/imunologia , Influenza Aviária/mortalidade , Influenza Aviária/virologia , Mardivirus/genética , Mardivirus/imunologia , Mardivirus/patogenicidade , Doença de Marek/imunologia , Doença de Marek/mortalidade , Doença de Marek/virologia , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/mortalidade , Doenças das Aves Domésticas/virologia , Análise de Sobrevida , Vacinação , Vacinas Sintéticas , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/biossíntese
20.
Viruses ; 9(4)2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28368367

RESUMO

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.


Assuntos
Mardivirus/imunologia , Mardivirus/isolamento & purificação , Doença de Marek/imunologia , Doença de Marek/virologia , Vacinas Virais/imunologia , Animais , Galinhas , China , Mardivirus/genética , Mardivirus/patogenicidade , Doença de Marek/prevenção & controle , Falha de Tratamento , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA