Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 252
Filtrar
1.
PLoS One ; 19(5): e0303371, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38728352

RESUMO

Marek's disease (MD) is an important neoplastic disease caused by serotype 1 Marek's disease virus (MDV-1), which results in severe economic losses worldwide. Despite vaccination practices that have controlled the MD epidemic, current increasing MD-suspected cases indicate the persistent viral infections circulating among vaccinated chicken farms in many countries. However, the lack of available information about phylogeny and molecular characterization of circulating MDV-1 field strains in Taiwan reveals a potential risk in MD outbreaks. This study investigated the genetic characteristics of 18 MDV-1 strains obtained from 17 vaccinated chicken flocks in Taiwan between 2018 and 2020. Based on the sequences of the meq oncogene, the phylogenetic analysis demonstrated that the circulating Taiwanese MDV-1 field strains were predominantly in a single cluster that showed high similarity with strains from countries of the East Asian region. Because the strains were obtained from CVI988/Rispens vaccinated chicken flocks and the molecular characteristics of the Meq oncoprotein showed features like vvMDV and vv+MDV strains, the circulating Taiwanese MDV-1 field strains may have higher virulence compared with vvMDV pathotype. In conclusion, the data presented demonstrates the circulation of hypervirulent MDV-1 strains in Taiwan and highlights the importance of routine surveillance and precaution strategies in response to the emergence of enhanced virulent MDV-1.


Assuntos
Galinhas , Herpesvirus Galináceo 2 , Doença de Marek , Proteínas Oncogênicas Virais , Filogenia , Animais , Galinhas/virologia , Taiwan/epidemiologia , Doença de Marek/virologia , Doença de Marek/prevenção & controle , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Virulência/genética , Proteínas Oncogênicas Virais/genética , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas contra Doença de Marek/genética , Vacinas contra Doença de Marek/imunologia , Vacinação/veterinária
2.
Science ; 382(6676): 1276-1281, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38096384

RESUMO

The pronounced growth in livestock populations since the 1950s has altered the epidemiological and evolutionary trajectory of their associated pathogens. For example, Marek's disease virus (MDV), which causes lymphoid tumors in chickens, has experienced a marked increase in virulence over the past century. Today, MDV infections kill >90% of unvaccinated birds, and controlling it costs more than US$1 billion annually. By sequencing MDV genomes derived from archeological chickens, we demonstrate that it has been circulating for at least 1000 years. We functionally tested the Meq oncogene, one of 49 viral genes positively selected in modern strains, demonstrating that ancient MDV was likely incapable of driving tumor formation. Our results demonstrate the power of ancient DNA approaches to trace the molecular basis of virulence in economically relevant pathogens.


Assuntos
Galinhas , Herpesvirus Galináceo 2 , Doença de Marek , Animais , Galinhas/virologia , Herpesvirus Galináceo 2/classificação , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Linfoma/virologia , Doença de Marek/história , Doença de Marek/virologia , Virulência/genética , Filogenia
3.
J Virol ; 96(9): e0032122, 2022 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-35412345

RESUMO

Circular RNAs (circRNAs) are a recently rediscovered class of functional noncoding RNAs that are involved in gene regulation and cancer development. Next-generation sequencing approaches identified circRNA fragments and sequences underlying circularization events in virus-induced cancers. In the present study, we performed viral circRNA expression analysis and full-length sequencing in infections with Marek's disease virus (MDV), which serves as a model for herpesvirus-induced tumorigenesis. We established inverse PCRs to identify and characterize circRNA expression from the repeat regions of the MDV genome during viral replication, latency, and reactivation. We identified a large variety of viral circRNAs through precise mapping of full-length circular transcripts and detected matching sequences with several viral genes. Hot spots of circRNA expression included the transcriptional unit of the major viral oncogene encoding the Meq protein and the latency-associated transcripts (LATs). Moreover, we performed genome-wide bioinformatic analyses to extract back-splice junctions from lymphoma-derived samples. Using this strategy, we found that circRNAs were abundantly expressed in vivo from the same key virulence genes. Strikingly, the observed back-splice junctions do not follow a unique canonical pattern, compatible with the U2-dependent splicing machinery. Numerous noncanonical junctions were observed in viral circRNA sequences characterized from in vitro and in vivo infections. Given the importance of the genes involved in the transcription of these circRNAs, our study contributes to our understanding and complexity of this deadly pathogen. IMPORTANCE Circular RNAs (circRNAs) were rediscovered in recent years both in physiological and pathological contexts, such as in cancer. Viral circRNAs are encoded by at least two human herpesviruses, the Epstein Barr virus and the Kaposi's Sarcoma-associated herpesvirus, both associated with the development of lymphoma. Marek's disease virus (MDV) is a well-established animal model to study virus-induced lymphoma but circRNA expression has not been reported for MDV yet. Our study provided the first evidence of viral circRNAs that were expressed at key steps of the MDV lifecycle using genome-wide analyses of circRNAs. These circRNAs were primarily found in transcriptional units that corresponded to the major MDV virulence factors. In addition, we established a bioinformatics pipeline that offers a new tool to identify circular RNAs in other herpesviruses. This study on the circRNAs provided important insights into major MDV virulence genes and herpesviruses-mediated gene dysregulation.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Galináceo 2 , Doença de Marek , RNA Circular , Animais , Galinhas , Estudo de Associação Genômica Ampla , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Linfoma/virologia , Doença de Marek/virologia , Proteínas Oncogênicas Virais/genética , RNA Circular/genética , RNA não Traduzido/genética , Virulência/genética
4.
Viruses ; 14(2)2022 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-35215975

RESUMO

Marek's disease virus (MDV) causes malignant lymphoma in chickens (Marek's disease, MD). Although MD is currently controlled by vaccination, MDV strains have continuously increased in virulence over the recent decades. Polymorphisms in Meq, an MDV-encoded oncoprotein that serves as a transcription factor, have been associated with the enhanced virulence of the virus. In addition, insertions and deletions in Meq have been observed in MDV strains of higher virulence, but their contribution to said virulence remains elusive. In this study, we investigated the contribution of an insertion (L-Meq) and a deletion in the Meq gene (S-Meq) to its functions and MDV pathogenicity. Reporter assays revealed that both insertion and deletion enhanced the transactivation potential of Meq. Additionally, we generated RB-1B-based recombinant MDVs (rMDVs) encoding each Meq isoform and analyzed their pathogenic potential. rMDV encoding L-Meq indueced the highest mortality and tumor incidence in infected animals, whereas the rMDV encoding S-Meq exhibited the lowest pathogenicity. Thus, insertion enhanced the transactivation activity of Meq and MDV pathogenicity, whereas deletion reduced pathogenicity despite having increased transactivation activity. These data suggest that other functions of Meq affect MDV virulence. These data improve our understanding of the mechanisms underlying the evolution of MDV virulence.


Assuntos
Herpesvirus Galináceo 2/genética , Proteínas Oncogênicas Virais/fisiologia , Ativação Transcricional/fisiologia , Animais , Embrião de Galinha , Herpesvirus Galináceo 2/patogenicidade , Virulência
5.
Genes (Basel) ; 12(10)2021 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-34681024

RESUMO

The avian α-herpesvirus known as Marek's disease virus (MDV) linearly integrates its genomic DNA into host telomeres during infection. The resulting disease, Marek's disease (MD), is characterized by virally-induced lymphomas with high mortality. The temporal dynamics of MDV-positive (MDV+) transformed cells and expansion of MD lymphomas remain targets for further understanding. It also remains to be determined whether specific host chromosomal sites of MDV telomere integration confer an advantage to MDV-transformed cells during tumorigenesis. We applied MDV-specific fluorescence in situ hybridization (MDV FISH) to investigate virus-host cytogenomic interactions within and among a total of 37 gonad lymphomas and neoplastic splenic samples in birds infected with virulent MDV. We also determined single-cell, chromosome-specific MDV integration profiles within and among transformed tissue samples, including multiple samples from the same bird. Most mitotically-dividing cells within neoplastic samples had the cytogenomic phenotype of 'MDV telomere-integrated only', and tissue-specific, temporal changes in phenotype frequencies were detected. Transformed cell populations composing gonad lymphomas exhibited significantly lower diversity, in terms of heterogeneity of MDV integration profiles, at the latest stages of tumorigenesis (>50 days post-infection (dpi)). We further report high interindividual and lower intraindividual variation in MDV integration profiles of lymphoma cells. There was no evidence of integration hotspots into a specific host chromosome(s). Collectively, our data suggests that very few transformed MDV+ T cell populations present earlier in MDV-induced lymphomas (32-50 dpi), survive, and expand to become the dominant clonal population in more advanced MD lymphomas (51-62 dpi) and establish metastatic lymphomas.


Assuntos
Herpesvirus Galináceo 2/genética , Linfoma/genética , Doença de Marek/genética , Doenças das Aves Domésticas/genética , Animais , Carcinogênese/genética , Galinhas/genética , Galinhas/virologia , Herpesvirus Galináceo 2/patogenicidade , Interações Hospedeiro-Patógeno/genética , Hibridização in Situ Fluorescente , Linfoma/etiologia , Linfoma/patologia , Linfoma/virologia , Doença de Marek/complicações , Doença de Marek/patologia , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Neoplasias Esplênicas/etiologia , Neoplasias Esplênicas/genética , Neoplasias Esplênicas/patologia , Linfócitos T/virologia , Telômero/genética , Telômero/virologia , Integração Viral/genética
6.
Vet Microbiol ; 259: 109082, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34144834

RESUMO

MicroRNAs (miRNAs) are a class of ∼22 nucleotides non-coding RNAs that are encoded by a wide range of hosts. Viruses, especially herpesviruses, encode a variety of miRNAs that involved in disease progression. Recently, a cluster of virus-encoded miRNAs, miR-M8-M10, have been shown to restrict early cytolytic replication and pathogenesis of Marek's disease virus (MDV), an oncogenic avian alphaherpesvirus that causes lymphoproliferative disease in chickens. In this study, we specifically dissected the role of miR-M7, a member of cluster miR-M8-M10, in regulating MDV replication and pathogenesis. We found that deletion of miR-M7-5p did not affect the virus plaque size and growth in cell culture. However, compared to parental virus, infection of miR-M7-5p deletion virus significantly increased MDV genome copy number at 5 days post infection, suggesting that miR-M7 plays a role to restrict MDV replication during early cytolytic phase. In addition, our results showed that infection of miR-M7-5p deletion virus significantly enhanced the mortality of chickens, even it induced lymphoid organ atrophy similar to parental and revertant viruses. Taken together, our study revealed that the miR-M7 acts as a repressive factor of MDV replication and pathogenesis.


Assuntos
Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , MicroRNAs/genética , Proteínas Virais/genética , Replicação Viral/genética , Animais , Células Cultivadas , Galinhas/virologia , Fibroblastos/virologia , Deleção de Genes , Herpesvirus Galináceo 2/crescimento & desenvolvimento , Doença de Marek/virologia , Organismos Livres de Patógenos Específicos , Fatores de Virulência/genética
7.
Vet Immunol Immunopathol ; 237: 110277, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34090158

RESUMO

Cytokine transcripts were evaluated chronologically in the brain and in the eye of chickens infected with the very virulent plus Marek's disease virus (vv + MDV) strain 648A. Brain and eye samples were collected from chickens that were either suffering from transient paralysis (TP) (11 days post inoculation, dpi) or had completely recovered from TP but started developing clinical signs of persistent neurological disease (PND) (18-31 dpi). Results obtained from samples collected at 11 dpi are referred as EL (early lesions) and results obtained from samples collected at later times (18-31 dpi) are referred as LL (late lesions). Marked differences were found in the cytokine transcripts in brain and eye. While proinflammatory cytokines (IL-1ß, IL-8, IL-18), iNOS, IFN-α, IFN-γ, and IL-15 were upregulated in the brain during EL and LL, only IL-8 and IFN-γ were upregulated in the eye at both times (EL and LL). The two evaluated viral transcripts (gB and meq) were found in both eye and brain during EL and LL. Levels of the two viral transcripts evaluated were higher at LL than at EL in both brain and eye. No differences were found in any of the viral transcripts between eye and brain during EL. However, during the LL, the levels of meq transcripts were higher in the eye than in the brain. Our results suggest that MDV elicits different immune responses in the brain and in the eye of infected chickens. Because immune responses in the eye of chickens have been poorly studied, further studies on the pathogenesis of MDV in the eye could greatly contribute to our knowledge on the chicken eye immunity.


Assuntos
Encéfalo/imunologia , Galinhas , Citocinas/biossíntese , Olho/imunologia , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/imunologia , Doenças do Sistema Nervoso/veterinária , Animais , Encéfalo/patologia , Olho/patologia , Doença de Marek/patologia , Doenças do Sistema Nervoso/imunologia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/virologia , Transcriptoma , Virulência
8.
Sci Rep ; 11(1): 7753, 2021 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-33833367

RESUMO

We have formerly shown that glycoprotein C (gC) of Gallid alphaherpesvirus 2, better known as Marek's disease (MD) alphaherpesvirus (MDV), is required for interindividual spread in chickens. Since gC is conserved within the Alphaherpesvirinae subfamily, we hypothesized gC was important for interindividual spread of other alphaherpesviruses. To test this hypothesis, we first generated a fluorescent protein tagged clone of Gallid alphaherpesvirus 3 MD vaccine strain 301B/1 to track virus replication in cell culture and chickens using fluorescent microscopy. Following validation of this system, we removed the open reading frame of 301B/1 gC from the genome and determined whether it was required for interindividual spread using experimental and natural infection studies. Interindividual spread of MD vaccine 301B/1 was abrogated by removal of 301B/1 gC. Rescuent virus in which 301B/1 gC was inserted back into the genome efficiently spread among chickens. To further study the conserved function of gC, we replaced 301B/1 gC with MDV gC and this virus also efficiently spread in chickens. These data suggest the essential function of alphaherpesvirus gC proteins is conserved and can be exploited during the generation of future vaccines against MD that affects the poultry industry worldwide.


Assuntos
Galinhas/virologia , Herpesvirus Galináceo 2/patogenicidade , Proteínas do Envelope Viral/fisiologia , Sequência de Aminoácidos , Animais , Herpesvirus Galináceo 2/metabolismo , Herpesvirus Galináceo 2/fisiologia , Doença de Marek/transmissão , Doença de Marek/virologia , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/química , Replicação Viral
9.
PLoS Pathog ; 17(2): e1009307, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33596269

RESUMO

Marek's disease virus (MDV) is a potent oncogenic alphaherpesvirus that elicits a rapid onset of malignant T-cell lymphomas in chickens. Three MDV types, including GaHV-2 (MDV-1), GaHV-3 (MDV-2) and MeHV-1 (HVT), have been identified and all encode a US3 protein kinase. MDV-1 US3 is important for efficient virus growth in vitro. To study the role of US3 in MDV replication and pathogenicity, we generated an MDV-1 US3-null virus and chimeric viruses by replacing MDV-1 US3 with MDV-2 or HVT US3. Using MD as a natural virus-host model, we showed that both MDV-2 and HVT US3 partially rescued the growth deficiency of MDV-1 US3-null virus. In addition, deletion of MDV-1 US3 attenuated the virus resulting in higher survival rate and lower MDV specific tumor incidence, which could be partially compensated by MDV-2 and HVT US3. We also identified chicken histone deacetylase 1 (chHDAC1) as a common US3 substrate for all three MDV types while only US3 of MDV-1 and MDV-2 phosphorylate chHDAC2. We further determined that US3 of MDV-1 and HVT phosphorylate chHDAC1 at serine 406 (S406), while MDV-2 US3 phosphorylates S406, S410, and S415. In addition, MDV-1 US3 phosphorylates chHDAC2 at S407, while MDV-2 US3 targets S407 and S411. Furthermore, biochemical studies show that MDV US3 mediated phosphorylation of chHDAC1 and 2 affect their stability, transcriptional regulation activity, and interaction network. Using a class I HDAC specific inhibitor, we showed that MDV US3 mediated phosphorylation of chHDAC1 and 2 is involved in regulation of virus replication. Overall, we identified novel substrates for MDV US3 and characterized the role of MDV US3 in MDV pathogenesis.


Assuntos
Herpesvirus Galináceo 2/patogenicidade , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Doença de Marek/virologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/metabolismo , Replicação Viral , Animais , Galinhas , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Doença de Marek/metabolismo , Doença de Marek/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Virais/genética
10.
J Virol ; 95(3)2021 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-33115875

RESUMO

Marek's disease virus (MDV) is an oncogenic alphaherpesvirus of chickens. The MDV genome consists of two unique regions that are both flanked by inverted repeat regions. These repeats harbor several genes involved in virus replication and pathogenesis, but it remains unclear why MDV and other herpesviruses harbor these large sequence duplications. In this study, we set to determine if both copies of these repeat regions are required for MDV replication and pathogenesis. Our results demonstrate that MDV mutants lacking the entire internal repeat region (ΔIRLS) efficiently replicate and spread from cell-to-cell in vitro However, ΔIRLS replication was severely impaired in infected chickens and the virus caused significantly less frequent disease and tumors compared to the controls. In addition, we also generated recombinant viruses that harbor a deletion of most of the internal repeat region, leaving only short terminal sequences behind (ΔIRLS-HR). These remaining homologous sequences facilitated rapid restoration of the deleted repeat region, resulting in a virus that caused disease and tumors comparable to the wild type. Therefore, ΔIRLS-HR represents an excellent platform for rapid genetic manipulation of the virus genome in the repeat regions. Taken together, our study demonstrates that MDV requires both copies of the repeats for efficient replication and pathogenesis in its natural host.IMPORTANCE Marek's disease virus (MDV) is a highly oncogenic alphaherpesvirus that infects chickens and causes losses in the poultry industry of up to $2 billion per year. The virus is also widely used as a model to study alphaherpesvirus pathogenesis and virus-induced tumor development in a natural host. MDV and most other herpesviruses harbor direct or inverted repeats regions in their genome. However, the role of these sequence duplications in MDV remains elusive and has never been investigated in a natural virus-host model for any herpesvirus. Here, we demonstrate that both copies of the repeats are needed for efficient MDV replication and pathogenesis in vivo, while replication was not affected in cell culture. With this, we further dissect herpesvirus genome biology and the role of repeat regions in Marek's disease virus replication and pathogenesis.


Assuntos
Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Neoplasias/virologia , Sequências Repetitivas de Ácido Nucleico , Deleção de Sequência , Replicação Viral , Animais , Galinhas , Genoma , Doença de Marek/genética , Doença de Marek/patologia , Mutação , Neoplasias/genética , Neoplasias/patologia
11.
Vet Microbiol ; 251: 108911, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33212362

RESUMO

Marek's disease virus (MDV) genome contains a number of uncharacterized long open reading frames (LORF) and their role in viral pathogenesis has not been fully investigated. Among them, LORF9 (MDV069) and LORF10 (MDV071) are locate at the right terminus of the MDV genome unique long region (UL). To investigate their role in MDV pathogenesis, we generated LORF9 or LORF10 deletion and revertant viruses. In vitro growth kinetics results show that both LORF9 and LORF10 are not essential for virus growth in cell culture. However, LORF9, but not LORF10, is involved in MDV early cytolytic replication in vivo, as evidenced by limited viral antigen expression in lymphoid organs of LORF9 deletion virus inoculated chickens. MDV genome copy number data further confirmed that LORF9 is important for MDV replication in spleen during early cytolytic phase. Deletion of LORF9 also partially impairs the replication of MDV in feather follicle epithelium (FFE); however, it can still establish latency and transformation. In addition, pathogenesis studies show that deletion of LORF9, but not LORF10, result in significant reduction of MDV induced mortality and slightly reduce MDV associated tumors of inoculated chickens. Importantly, we confirmed these results with the generation of LORF9 and LORF10 revertant viruses that fully restore the phenotypes of parental MDV. In conclusion, our results show that deletion of LORF9, but not LORF10, significantly impair viral replication in lymphoid organs during early cytolytic phase and attenuate Marek's disease virus pathogenesis.


Assuntos
Deleção de Genes , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Proteínas Virais/genética , Replicação Viral/genética , Animais , Células Cultivadas , Embrião de Galinha/citologia , Galinhas/virologia , Fibroblastos/virologia , Herpesvirus Galináceo 2/crescimento & desenvolvimento , Fases de Leitura Aberta , Doenças das Aves Domésticas/virologia
12.
Viruses ; 12(11)2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33212952

RESUMO

Herpesvirus-encoded microRNAs (miRNAs) have been discovered in infected cells; however, lack of a suitable animal model has hampered functional analyses of viral miRNAs in vivo. Marek's disease virus (MDV) (Gallid alphaherpesvirus 2, GaHV-2) genome contains 14 miRNA precursors, which encode 26 mature miRNAs, grouped into three clusters. In this study, the role of MDV-encoded cluster 3 miRNAs, also known as mdv1-miR-M8-M10, in pathogenesis was evaluated in chickens, the natural host of MDV. Our results show that deletion of cluster 3 miRNAs did not affect virus replication and plaque size in cell culture, but increased early cytolytic replication of MDV in chickens. We also observed that deletion of cluster 3 miRNAs resulted in significantly higher virus reactivation from peripheral blood lymphocytes. In addition, pathogenesis studies showed that deletion of cluster 3 miRNAs resulted in more severe atrophy of lymphoid organs and reduced mean death time, but did not affect the incidence of MDV-associated visceral tumors. We confirmed these results by generating a cluster 3 miRNA revertant virus in which the parental MDV phenotype was restored. To the best of our knowledge, our study provides the first evidence that MDV cluster 3 miRNAs play an important role in modulating MDV pathogenesis.


Assuntos
Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , MicroRNAs/genética , Replicação Viral/genética , Animais , Células Cultivadas , Galinhas/virologia , Fibroblastos/patologia , Fibroblastos/virologia , Deleção de Genes , Herpesvirus Galináceo 2/fisiologia , RNA Viral/genética , Organismos Livres de Patógenos Específicos , Virulência
13.
J Virol ; 94(23)2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-32967954

RESUMO

Gallid herpesvirus type 2 (GaHV-2) is an oncogenic alphaherpesvirus that induces malignant T-cell lymphoma in chicken. GaHV-2 encodes a viral telomerase RNA subunit (vTR) that plays a crucial role in virus-induced tumorigenesis, enhances telomerase activity, and possesses functions independent of the telomerase complex. vTR is driven by a robust viral promoter, highly expressed in virus-infected cells, and regulated by two c-Myc response elements (c-Myc REs). The regulatory mechanisms involved in controlling vTR and other genes during viral replication and latency remain poorly understood but are crucial to understanding this oncogenic herpesvirus. Therefore, we investigated DNA methylation patterns of CpG dinucleotides found in the vTR promoter and measured the impact of methylation on telomerase activity. We demonstrated that telomerase activity was considerably increased following viral reactivation. Furthermore, CpG sites within c-Myc REs showed specific changes in methylation after in vitro reactivation and in infected animals over time. Promoter reporter assays indicated that one of the c-Myc REs is involved in regulating vTR transcription, and that methylation strongly influenced vTR promoter activity. To study the importance of the CpG sites found in c-Myc REs in virus-induced tumorigenesis, we generated recombinant virus containing mutations in CpG sites of c-Myc REs together with the revertant virus by two-step Red-mediated mutagenesis. Introduced mutations in the vTR promoter did not affect the replication properties of the recombinant viruses in vitro In contrast, replication of the mutant virus in infected chickens was severely impaired, and tumor formation completely abrogated. Our data provides an in-depth characterization of c-Myc oncoprotein REs and the involvement of DNA methylation in transcriptional regulation of vTR.IMPORTANCE Previous studies demonstrated that telomerase RNAs possess functions that promote tumor development independent of the telomerase complex. vTR is a herpesvirus-encoded telomerase RNA subunit that plays a crucial role in virus-induced tumorigenesis and is expressed by a robust viral promoter that is highly regulated by the c-Myc oncoprotein binding to the E-boxes. Here, we demonstrated that the DNA methylation patterns in the functional c-Myc response elements of the vTR promoter change upon reactivation from latency, and that demethylation strongly increases telomerase activity in virus-infected cells. Moreover, the introduction of mutation in the CpG dinucleotides of the c-Myc binding sites resulted in decreased vTR expression and complete abrogation of tumor formation. Our study provides further confirmation of the involvement of specific DNA methylation patterns in the regulation of vTR expression and vTR importance for virus-induced tumorigenesis.


Assuntos
Metilação de DNA/fisiologia , Herpesvirus Galináceo 2/genética , Regiões Promotoras Genéticas , RNA Viral/genética , Telomerase/genética , Animais , Carcinogênese/genética , Linhagem Celular , Galinhas , Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/enzimologia , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Mutagênese Sítio-Dirigida , Mutação , RNA , Replicação Viral
14.
Viruses ; 12(9)2020 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-32962247

RESUMO

Marek's disease (MD) is an infectious malignant T-cell lymphoma proliferative disease caused by Marek's disease virus (MDV). In recent years, the emergence of very virulent (vv) and/or very virulent plus (vv +) strains of MDV in the field has been suggested as one of the causes of vaccination failure. The pathogenicity of the MDV strain GX18NNM4, isolated from a clinical outbreak in a broiler breeder flock that was vaccinated with CVI988/Rispens, was investigated. In the vaccination-challenge test, GX18NNM4 was able to break through the protections provided by the vaccines CVI988 and 814. It also significantly reduced body weight gain and caused marked gross lesions and a large area of infiltration of neoplastic lymphocyte cells in the heart, liver, pancreas, etc. of the infected birds. In addition, the expressions of programmed death 1 (PD-1) and its ligand, programmed death ligand 1 (PD-L1), in the spleens and cecal tonsils (CTs) of the unvaccinated challenged birds were significantly increased compared to those in the vaccinated challenged birds, indicating that the PD-1/PD-L1 pathway is related to immune evasion mechanisms. The results showed that the GX18NNM4 strain could cause severe immunosuppression and significantly decrease the protections provided by the current commercial vaccines, thus showing GX18NNM4 to be a vv + MDV strain.


Assuntos
Herpesvirus Galináceo 2/patogenicidade , Vacinas contra Doença de Marek/imunologia , Doença de Marek/prevenção & controle , Animais , Antígeno B7-H1/metabolismo , Galinhas/virologia , Tolerância Imunológica , Terapia de Imunossupressão , Doença de Marek/patologia , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Receptor de Morte Celular Programada 1/metabolismo , Baço/imunologia , Vacinação/veterinária , Carga Viral
15.
Arch Virol ; 165(11): 2589-2597, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32876794

RESUMO

Marek's disease (MD) is a contagious avian viral disease that is responsible for large economic losses to farmers. The disease is caused by Marek's disease virus (species Gallid alphaherpesvirus 2), which causes neurological lesions, immune suppression, and tumor proliferation of lymphoid cells that invade a large number of organs and tissues. Despite widespread vaccination, Marek's disease virus (MDV), has shown a continuous increase in its virulence and has acquired the ability to overcome immune responses induced by vaccines. In the present study, the oncogenic serotype MDV-1 was detected by real-time PCR in DNA samples extracted from organs developing tumor infiltrations. Identification of the pathotype based on a 132-bp tandem repeat and sequencing and phylogenetic analysis of the Meq gene and its encoded protein allowed classification of the isolated viruses as "very virulent", with two new and unique mutations in the Meq gene resulting in amino acid substitutions. Sequencing of pp38, vIl-8, UL1 and UL44 genes did not reveal any new mutations that were characteristic of the Tunisian isolates or correlated with virulence. These results raised concerns about the ability of HVT and CVI988 vaccines, which are currently used in Tunisia and other countries, to protect chickens against highly virulent virus strains.


Assuntos
Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/patogenicidade , Proteínas Oncogênicas Virais/genética , Filogenia , Sequência de Aminoácidos , Animais , Galinhas/virologia , DNA Viral/química , Doença de Marek/virologia , Mutação Puntual , Polimorfismo de Fragmento de Restrição , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Tunísia , Virulência/genética
16.
BMC Vet Res ; 16(1): 303, 2020 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-32831091

RESUMO

BACKGROUND: Marek's disease (MD) is a chicken neoplastic disease, which brings huge economic losses to the global poultry industry. The wild type p53, a tumor suppressor gene, plays a key role in blocking cell cycle, promoting apoptosis, and maintaining the stability of the genome. However, the mutant p53 losses its tumor inhibitory role and become an oncogene when a mutation has happened. RESULTS: The mutation rate of p53 was 60% in the experimentally and naturally infected chickens. The mutations included point-mutations and deletions, and mostly located in the DNA-binding domain. The mutated p53 was expressed in various tumor tissues in an infected chicken. The mutant P53 proteins were notably accumulated in the cytoplasm due to the loss in the function of nuclear localization. Unlike the study on human cancer, the concentrations of P53 in the serums of MD infected chicken were significantly lower than the control group. CONCLUSIONS: The p53 mutations were apparent in the development of MD. P53 and P53 antibody level in serum could be a useful marker in the diagnosis and surveillance of MD.


Assuntos
Doença de Marek/genética , Mutação , Doenças das Aves Domésticas/genética , Proteína Supressora de Tumor p53/genética , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/sangue , Galinhas , Feminino , Herpesvirus Galináceo 2/imunologia , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Proteína Supressora de Tumor p53/sangue
17.
PLoS Biol ; 18(3): e3000619, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32134914

RESUMO

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


Assuntos
Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/transmissão , Vacinas Virais/farmacologia , Animais , Galinhas , Plumas/virologia , Interações Hospedeiro-Patógeno , Doença de Marek/etiologia , Doença de Marek/mortalidade , Doença de Marek/prevenção & controle , Vacinação , Carga Viral , Vacinas Virais/administração & dosagem , Virulência , Eliminação de Partículas Virais
18.
Vet Microbiol ; 240: 108501, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31902513

RESUMO

Marek's disease (MD) is a highly contagious lymphoproliferative poultry disease caused by the oncogenic herpesvirus, Marek's disease virus (MDV). MDV strains have shown a continued evolution of virulence leading to immune failure, and MD cases continue to occur. Co-infection of virulent MDV strains is an important factor leading to viral evolution and host immune failure. This study conducted a laboratory diagnosis and analysis of a MDV infected flock. Testing showed that all samples were MDV positive. PCR detection identified a variable 132-base pair repeat (132-bpr) sequence copy number. This indicated that two virulent strains of MDV were co-infecting the flock. Therefore, we performed homology, sequence alignment, and phylogenetic tree analysis of MDV variant genes including meq, pp38, and RLORF4. Two MDV strains had co-infected the flock; one was the 132bpr two-copy characteristic strain (AH2C) and the other was a 132bpr three-copy characteristic strain (AH3C). Specific mutations in AH3C were found, suggesting that it is a new variant strain. Furthermore, the viral load of the two strains in vivo indicated that both strains had high and similar replication ability. There was no significant difference in the proportion of positive samples of the two strains causing disease. In the whole flock, neither strain displayed an obvious advantage. However, there was a dominant strain in individual chickens, with the exception of one sample. This study reported the co-infection regularity of two virulent MDV strains in the same flock, and even in the same chicken in field conditions. In the context of overall epidemiology, this study is a useful reference.


Assuntos
Galinhas/virologia , Coinfecção/veterinária , Herpesvirus Galináceo 2/classificação , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Animais , Coinfecção/virologia , Evolução Molecular , Variação Genética , Mutação , Filogenia , Aves Domésticas/virologia , Doenças das Aves Domésticas/virologia , Carga Viral , Virulência
19.
PLoS Pathog ; 15(9): e1007999, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31539404

RESUMO

The cellular DNA sensor cGMP-AMP synthase (cGAS) detects cytosolic viral DNA via the stimulator of interferon genes (STING) to initiate innate antiviral response. Herpesviruses are known to target key immune signaling pathways to persist in an immune-competent host. Marek's disease virus (MDV), a highly pathogenic and oncogenic herpesvirus of chickens, can antagonize host innate immune responses to achieve persistent infection. With a functional screen, we identified five MDV proteins that blocked beta interferon (IFN-ß) induction downstream of the cGAS-STING pathway. Specifically, the MDV major oncoprotein Meq impeded the recruitment of TANK-binding kinase 1 and IFN regulatory factor 7 (IRF7) to the STING complex, thereby inhibiting IRF7 activation and IFN-ß induction. Meq overexpression markedly reduced antiviral responses stimulated by cytosolic DNA, whereas knockdown of Meq heightened MDV-triggered induction of IFN-ß and downstream antiviral genes. Moreover, Meq-deficient MDV induced more IFN-ß production than wild-type MDV. Meq-deficient MDV also triggered a more robust CD8+ T cell response than wild-type MDV. As such, the Meq-deficient MDV was highly attenuated in replication and lymphoma induction compared to wild-type MDV. Taken together, these results revealed that MDV evades the cGAS-STING DNA sensing pathway, which underpins the efficient replication and oncogenesis. These findings improve our understanding of the virus-host interaction in MDV-induced lymphoma and may contribute to the development of novel vaccines against MDV infection.


Assuntos
Herpesvirus Galináceo 2/imunologia , Herpesvirus Galináceo 2/patogenicidade , Evasão da Resposta Imune , Doença de Marek/imunologia , Doença de Marek/virologia , Animais , Proteínas Aviárias/metabolismo , Carcinogênese , Galinhas , DNA Viral/imunologia , Patos , Herpesvirus Galináceo 2/fisiologia , Interações entre Hospedeiro e Microrganismos/imunologia , Imunidade Inata , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/metabolismo , Doença de Marek/metabolismo , Modelos Imunológicos , Nucleotidiltransferases/metabolismo , Proteínas Oncogênicas Virais/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Virais/imunologia , Replicação Viral
20.
Nat Microbiol ; 4(12): 2175-2183, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31477893

RESUMO

RNA virus populations are composed of highly diverse individuals that form a cloud of related sequences commonly referred to as a 'quasispecies'1-3. This diversity arises as a consequence of low-fidelity genome replication4,5. By contrast, DNA virus populations contain more uniform individuals with similar fitness6. Genome diversity is often correlated with increased fitness in RNA viruses, while DNA viruses are thought to require more faithful genome replication. During DNA replication, erroneously incorporated bases are removed by a 3'-5' exonuclease, a highly conserved enzymatic function of replicative DNA but not RNA polymerases. This proofreading process enhances replication fidelity and ensures the genome integrity of DNA organisms, including large DNA viruses7. Here, we show that a herpesvirus can tolerate impaired exonucleolytic proofreading, resulting in DNA virus populations, which, as in RNA viruses8, are composed of highly diverse genotypes of variable individual fitness. This indicates that herpesvirus mutant diversity may compensate for individual fitness loss. Notably, in vivo infection with diverse virus populations results in a marked increase in virulence compared to genetically homogenous parental virus. While we cannot exclude that the increase in virulence is caused by selection of and/or interactions between individual genotypes, our findings are consistent with quasispecies dynamics. Our results contrast with traditional views of DNA virus replication and evolution, and indicate that a substantial increase in population diversity can lead to higher virulence.


Assuntos
Variação Genética , Genoma Viral , Herpesvirus Galináceo 2/genética , Quase-Espécies/genética , Replicação Viral , Animais , Galinhas/virologia , Aptidão Genética , Genótipo , Herpesvirus Galináceo 2/patogenicidade , Herpesvirus Galináceo 2/fisiologia , Doença de Marek/virologia , Mutação , Organismos Livres de Patógenos Específicos , Virulência/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...