Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
PLoS Biol ; 13(7): e1002198, 2015 07.
Artigo em Inglês | MEDLINE | ID: mdl-26214839

RESUMO

Could some vaccines drive the evolution of more virulent pathogens? Conventional wisdom is that natural selection will remove highly lethal pathogens if host death greatly reduces transmission. Vaccines that keep hosts alive but still allow transmission could thus allow very virulent strains to circulate in a population. Here we show experimentally that immunization of chickens against Marek's disease virus enhances the fitness of more virulent strains, making it possible for hyperpathogenic strains to transmit. Immunity elicited by direct vaccination or by maternal vaccination prolongs host survival but does not prevent infection, viral replication or transmission, thus extending the infectious periods of strains otherwise too lethal to persist. Our data show that anti-disease vaccines that do not prevent transmission can create conditions that promote the emergence of pathogen strains that cause more severe disease in unvaccinated hosts.


Assuntos
Mardivirus/patogenicidade , Vacinas contra Doença de Marek/efeitos adversos , Doença de Marek/transmissão , Seleção Genética , Animais , Galinhas , Mardivirus/genética , Doença de Marek/imunologia , Eliminação de Partículas Virais
2.
J Gen Virol ; 98(4): 779-790, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28475033

RESUMO

Marek's disease virus (MDV), an alphaherpesvirus of poultry, causes Marek's disease and is characterized by visceral CD4+TCRαß+ T-cell lymphomas in susceptible hosts. Immortal cell lines harbouring the viral genome have been generated from ex vivo cultures of MD tumours. As readily available sources of large numbers of cells, MDV-transformed lymphoblastoid cell lines (LCLs) are extremely valuable for studies of virus-host interaction. While the viral genome in most cells is held in a latent state, minor populations of cells display spontaneous reactivation identifiable by the expression of lytic viral genes. Spontaneous reactivation in these cells presents an opportunity to investigate the biological processes involved in the virus reactivation. For detailed characterization of the molecular events associated with reactivation, we used two lymphoblastoid cell lines derived from lymphomas induced by pRB1B-UL47eGFP, a recombinant MDV engineered to express enhanced green fluorescent protein (EGFP) fused with the UL47. We used fluorescence-activated cell sorting to purify the low-frequency EGFP-positive cells with a spontaneously activating viral genome from the majority EGFP-negative cells and analysed their gene expression profiles by RNA-seq using Illumina HiSeq2500. Ingenuity pathway analysis on more than 2000 differentially expressed genes between the lytically infected (EGFP-positive) and latently infected (EGFP-negative) cell populations identified the biological pathways involved in the reactivation. Virus-reactivating cells exhibited differential expression of a significant number of viral genes, with hierarchical differences in expression levels. Downregulation of a number of host genes including those directly involved in T-cell activation, such as CD3, CD28, ICOS and phospholipase C, was also noticed in the LCL undergoing lytic switch.


Assuntos
Perfilação da Expressão Gênica , Herpesvirus Galináceo 2/genética , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Proteínas Virais/genética , Animais , Linhagem Celular Tumoral , Galinhas , Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/fisiologia , Linfoma/virologia , Proteínas Virais/metabolismo
3.
J Gen Virol ; 98(4): 810-820, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28113043

RESUMO

The v-rel oncoprotein encoded by reticuloendotheliosis virus T strain (Rev-T) is a member of the rel/NF-κB family of transcription factors capable of transformation of primary chicken spleen and bone marrow cells. Rapid transformation of avian haematopoietic cells by v-rel occurs through a process of deregulation of multiple protein-encoding genes through its direct effect on their promoters. More recently, upregulation of oncogenic miR-155 and its precursor pre-miR-155 was demonstrated in both Rev-T-infected chicken embryo fibroblast cultures and Rev-T-induced B-cell lymphomas. Through electrophoresis mobility shift assay and reporter analysis on the gga-miR-155 promoter, we showed that the v-rel-induced miR-155 overexpression occurred by the direct binding to one of the putative NF-κB binding sites. Using the v-rel-induced transformation model on chicken embryonic splenocyte cultures, we could demonstrate a dynamic increase in miR-155 levels during the transformation. Transcriptome profiles of lymphoid cells transformed by v-rel showed upregulation of miR-155 accompanied by downregulation of a number of putative miR-155 targets such as Pu.1 and CEBPß. We also showed that v-rel could rescue the suppression of miR-155 expression observed in Marek's disease virus (MDV)-transformed cell lines, where its functional viral homologue MDV-miR-M4 is overexpressed. Demonstration of gene expression changes affecting major molecular pathways, including organismal injury and cancer in avian macrophages transfected with synthetic mature miR-155, underlines its potential direct role in transformation. Our study suggests that v-rel-induced transformation involves a complex set of events mediated by the direct activation of NF-κB targets, together with inhibitory effects on microRNA targets.


Assuntos
Transformação Celular Viral , Interações Hospedeiro-Patógeno , Proteínas Oncogênicas v-rel/metabolismo , RNA Mensageiro/biossíntese , Vírus da Reticuloendoteliose/patogenicidade , Animais , Células Cultivadas , Galinhas , Perfilação da Expressão Gênica , Leucócitos Mononucleares/virologia , Regiões Promotoras Genéticas , Ligação Proteica
4.
PLoS Pathog ; 7(2): e1001305, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21383974

RESUMO

Notwithstanding the well-characterised roles of a number of oncogenes in neoplastic transformation, microRNAs (miRNAs) are increasingly implicated in several human cancers. Discovery of miRNAs in several oncogenic herpesviruses such as KSHV has further highlighted the potential of virus-encoded miRNAs to contribute to their oncogenic capabilities. Nevertheless, despite the identification of several possible cancer-related genes as their targets, the direct in vivo role of virus-encoded miRNAs in neoplastic diseases such as those induced by KSHV is difficult to demonstrate in the absence of suitable models. However, excellent natural disease models of rapid-onset Marek's disease (MD) lymphomas in chickens allow examination of the oncogenic potential of virus-encoded miRNAs. Using viruses modified by reverse genetics of the infectious BAC clone of the oncogenic RB-1B strain of MDV, we show that the deletion of the six-miRNA cluster 1 from the viral genome abolished the oncogenicity of the virus. This loss of oncogenicity appeared to be primarily due to the single miRNA within the cluster, miR-M4, the ortholog of cellular miR-155, since its deletion or a 2-nucleotide mutation within its seed region was sufficient to inhibit the induction of lymphomas. The definitive role of this miR-155 ortholog in oncogenicity was further confirmed by the rescue of oncogenic phenotype by revertant viruses that expressed either the miR-M4 or the cellular homolog gga-miR-155. This is the first demonstration of the direct in vivo role of a virus-encoded miRNA in inducing tumors in a natural infection model. Furthermore, the use of viruses deleted in miRNAs as effective vaccines against virulent MDV challenge, enables the prospects of generating genetically defined attenuated vaccines.


Assuntos
Herpesvirus Galináceo 2/genética , Linfoma/etiologia , Doença de Marek/etiologia , MicroRNAs/genética , Animais , Sequência de Bases , Células Cultivadas , Embrião de Galinha , Galinhas , Fibroblastos/citologia , Fibroblastos/metabolismo , Genoma Viral , Herpesvirus Galináceo 2/crescimento & desenvolvimento , Humanos , Linfoma/patologia , Linfoma/prevenção & controle , Doença de Marek/patologia , Doença de Marek/prevenção & controle , Dados de Sequência Molecular , Mutação/genética , RNA Viral/genética , Vacinação
5.
Avian Dis ; 57(2 Suppl): 387-94, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23901751

RESUMO

In addition to tumors, Marek's disease (MD) virus (MDV) can induce a variety of syndromes linked to the central nervous system. In fact, early descriptions of MD suggested that it was a condition affecting mainly the nervous system. Cytokines and other immune-related genes have been suggested to play a crucial role in MDV-mediated neuropathology, but the mechanisms behind the viral-induced neurologic dysfunction are still poorly understood. In the present study we have used reverse genetic strategies to show that pp14 is not involved in the oncogenic phenotype of MDV1 and is not required for viral replication; however, we provide evidence indicating that the absence of pp14 expression is correlated with increased survival of MDV1-infected chickens, and that its expression is associated with enhanced viral neurovirulence. Our data identify for the first time pp14 as a neurovirulence factor from MDV1 and open the possibility to investigate the molecular mechanisms by which pp14 mediates the damage to the avian nervous system.


Assuntos
Galinhas , Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/patogenicidade , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Proteínas Virais/genética , Fatores de Virulência/genética , Animais , Células Cultivadas , Embrião de Galinha , Deleção de Genes , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Sistema Nervoso/virologia , Capuzes de RNA/genética , Capuzes de RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Organismos Livres de Patógenos Específicos , Transcrição Gênica , Proteínas Virais/metabolismo , Fatores de Virulência/metabolismo
6.
Avian Dis ; 57(2 Suppl): 440-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23901759

RESUMO

To assess the effect of various vaccine strains on replication and shedding of virulent Marek's disease virus from experimentally infected chickens, quantitative PCR (q-PCR) methods were developed to accurately quantify viral DNA in infected chickens and in the environment in which they were housed. Four groups of 10 chickens, kept in poultry isolators, were vaccinated at 1 day old with one of four vaccines covering each of the three vaccine serotypes, then challenged with very virulent MDV strain Md5 at 8 days of age. At regular time-points, feather tips were collected from each chicken and poultry dust was collected from the air-extract prefilter of each isolator. DNA was extracted from feather and dust samples and subjected to real-time q-PCR, targeting the U(S)2 gene of MDV-1, in order to measure Md5 level per 10(4) feather tip cells or per microgram of dust. Accuracy of DNA extraction from dust and real-time q-PCR were validated by comparing either q-PCR cycle threshold values or the calculated MDV genome level; for use in q-PCR, DNA was extracted from serial dilutions of MDV-infected dust diluted with noninfected dust, or DNA from MDV-infected dust was diluted with DNA from noninfected dust. The results confirmed the accuracy and sensitivity of dust DNA extraction and subsequent q-PCR and showed that differences in virus levels between dust samples truly reflect differences in shedding. Vaccination delayed both replication of Md5 in feather tips and shedding of Md5. First detection of Md5 in feather tips always preceded or coincided with first detection in dust in each group. pCVI988 and HVT+SB-1 were the most efficient vaccines in reducing both replication and shedding of Md5. There was close correlation between mean virus level in feathers of each group and mean virus level in the dust shed by that group. This relationship was similar in each of the vaccinated groups, demonstrating that measurement of the virus in dust can be used to monitor accurately both the infection status of the chickens and environmental contamination by MDV.


Assuntos
Galinhas , Poeira/análise , Plumas/virologia , Genoma Viral , Herpesvirus Galináceo 2/genética , Doença de Marek/virologia , Doenças das Aves Domésticas/virologia , Reação em Cadeia da Polimerase em Tempo Real/métodos , Animais , Herpesvirus Galináceo 2/patogenicidade , Abrigo para Animais , Vacinas contra Doença de Marek/administração & dosagem , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Organismos Livres de Patógenos Específicos
7.
J Virol ; 83(11): 5846-53, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19297480

RESUMO

In this study, we have identified an internal ribosome entry site (IRES) from the highly infectious herpesvirus Marek's disease virus (MDV). The IRES was mapped to the intercistronic region (ICR) of a bicistronic mRNA that we cloned from the MDV-transformed CD4(+) T-cell line MSB-1. The transcript is a member of a family of mRNAs expressed as immediate-early genes with two open reading frames (ORF). The first ORF encodes a 14-kDa polypeptide with two N-terminal splice variants, whereas the second ORF is contained entirely within a single exon and encodes a 12-kDa protein also known as RLORF9. We have shown that the ICR that separates the two ORFs functions as an IRES that controls the translation of RLORF9 when cap-dependent translation is inhibited. Deletion analysis revealed that there are two potential IRES elements within the ICR. Reverse genetic experiments with the oncogenic strain of MDV type 1 indicated that deletion of IRES-controlled RLORF9 does not significantly affect viral replication or MDV-induced mortality.


Assuntos
DNA Intergênico/genética , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Mardivirus/genética , Mardivirus/metabolismo , Doença de Marek , Ribossomos/metabolismo , Animais , Linhagem Celular , Galinhas , Deleção de Genes , Regulação Viral da Expressão Gênica , Genoma Viral/genética , Ribossomos/genética , Transcrição Gênica/genética
8.
J Virol ; 83(21): 11142-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19692466

RESUMO

Marek's disease virus (MDV) is a lymphotropic alphaherpesvirus that induces fatal rapid-onset T-cell lymphomas in chickens, its natural host. The MDV-encoded nuclear oncoprotein Meq is essential for lymphomagenesis and acts as a regulator of transcription. Meq has structural features, including a basic domain adjacent to a leucine zipper motif (B-ZIP), that suggest it is related to the Jun/Fos family of transcription factors. Via the leucine zipper, Meq can form homodimers or heterodimerize with c-Jun. Meq/Meq homodimers are associated with transrepression, and Meq/Jun heterodimers can transactivate target genes carrying an AP-1-like binding site. In order to determine the role of the leucine zipper and of Meq dimerization in T lymphomagenesis, specific point mutations were engineered into the highly oncogenic RB-1B strain of MDV to produce virus completely lacking a functional Meq leucine zipper (RB-1B Meq(BZIP/BZIP)) or virus encoding Meq that cannot homodimerize but can still bind to c-Jun and an AP-1-like site on DNA (RB-1B Meq(Hom/Hom)). Both of these mutant viruses were capable of replication in cultured chicken embryo fibroblasts. However both mutations resulted in a complete loss of oncogenicity, since no lymphomas were produced up to 90 days postinfection in experimentally infected chicks. We conclude that the leucine zipper is necessary for the oncogenic activity of Meq and/or the efficient establishment of long-term MDV latency in T cells. Moreover, it appears that the ability to form homodimers is an absolute requirement and the ability to bind c-Jun alone is insufficient for the T-cell lymphomagenesis associated with virulent MDV.


Assuntos
Transformação Celular Viral , Linfoma de Células T/virologia , Mardivirus , Proteínas Oncogênicas Virais/química , Proteínas Oncogênicas Virais/metabolismo , Estrutura Quaternária de Proteína , Sequência de Aminoácidos , Animais , Galinhas/virologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Zíper de Leucina , Mardivirus/química , Mardivirus/metabolismo , Mardivirus/patogenicidade , Doença de Marek/virologia , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Proteínas Oncogênicas Virais/genética , Doenças das Aves Domésticas/virologia , Multimerização Proteica , Reprodutibilidade dos Testes , Alinhamento de Sequência , Análise de Sobrevida , Latência Viral
9.
J Virol ; 83(1): 489-92, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18945769

RESUMO

Kaposi's sarcoma-associated herpesvirus-encoded microRNA (miRNA) MiR-K12-11 was recently shown to be a functional ortholog of miR-155, a miRNA that plays a major role in lymphoid malignancies and the modulation of immune responses. Here we show that miR-M4, encoded by the highly oncogenic Marek's disease virus of chickens, shares common targets with miR-155 and thus is also a functional ortholog of miR-155, the first one identified in an alphaherpesvirus. The observation that two distinct oncogenic herpesviruses associated with distinct types of lymphomas in different species encode functional miR-155 orthologs suggested the importance of this miRNA in regulatory pathways and the biology of lymphomagenesis.


Assuntos
Mardivirus/genética , MicroRNAs/genética , RNA Viral/genética , Animais , Linhagem Celular , Galinhas , Regulação da Expressão Gênica , Genes Reporter , Luciferases/biossíntese , Luciferases/genética , Mardivirus/fisiologia , MicroRNAs/metabolismo , RNA Viral/metabolismo
10.
PLoS One ; 9(12): e114466, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25503397

RESUMO

Poly(A) binding protein 1 (PABP1) plays a central role in mRNA translation and stability and is a target by many viruses in diverse manners. We report a novel viral translational control strategy involving the recruitment of PABP1 to the 5' leader internal ribosome entry site (5L IRES) of an immediate-early (IE) bicistronic mRNA that encodes the neurovirulence protein (pp14) from the avian herpesvirus Marek's disease virus serotype 1 (MDV1). We provide evidence for the interaction between an internal poly(A) sequence within the 5L IRES and PABP1 which may occur concomitantly with the recruitment of PABP1 to the poly(A) tail. RNA interference and reverse genetic mutagenesis results show that a subset of virally encoded-microRNAs (miRNAs) targets the inhibitor of PABP1, known as paip2, and therefore plays an indirect role in PABP1 recruitment strategy by increasing the available pool of active PABP1. We propose a model that may offer a mechanistic explanation for the cap-independent enhancement of the activity of the 5L IRES by recruitment of a bona fide initiation protein to the 5' end of the message and that is, from the affinity binding data, still compatible with the formation of 'closed loop' structure of mRNA.


Assuntos
Herpesvirus Galináceo 2/genética , Iniciação Traducional da Cadeia Peptídica , Proteína I de Ligação a Poli(A)/metabolismo , Proteínas Virais/biossíntese , Fatores de Virulência/biossíntese , Animais , Sequência de Bases , Linhagem Celular , Galinhas , Regulação da Expressão Gênica , Humanos , MicroRNAs/genética , Dados de Sequência Molecular , Mutagênese , Mutação , Poli A/genética , Poli A/metabolismo , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas Virais/genética , Fatores de Virulência/genética
11.
Vaccine ; 27(2): 298-306, 2009 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-18977264

RESUMO

Live attenuated herpesvirus vaccines such as herpesvirus of turkey (HVT) have been used since 1970 for the control of Marek's disease (MD), a highly infectious lymphoproliferative disease of poultry. Despite the success of these vaccines in reducing losses from the disease, Marek's disease virus (MDV) strains have shown a continuing increase in virulence, presumably due to the inability of the current vaccines in preventing MDV replication. The highly specific and effective nature of RNA interference (RNAi) makes this technology particularly attractive for new antiviral strategies. In order to exploit the power of RNAi-mediated suppression of MDV replication in vivo delivered through existing vaccines, we engineered recombinant HVT expressing short hairpin RNA (shRNA) against MDV genes gB and UL29. The levels of protection induced by the RNAi-expressing HVT against virulent virus challenge were similar to the parent pHVT3 virus. However, chickens vaccinated with recombinant HVT expressing shRNA showed moderate reduction of challenge virus replication in blood and feather samples. Delivery of RNAi-based gene silencing through live attenuated vaccines for reducing replication of pathogenic viruses is a novel approach for the control of infectious diseases.


Assuntos
Herpesvirus Meleagrídeo 1 , Vacinas contra Doença de Marek/administração & dosagem , Doença de Marek/prevenção & controle , Interferência de RNA , RNA Interferente Pequeno/genética , Recombinação Genética , Animais , Sequência de Bases , Galinhas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Herpesvirus Meleagrídeo 1/genética , Herpesvirus Meleagrídeo 1/metabolismo , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/metabolismo , Herpesvirus Galináceo 2/fisiologia , Doença de Marek/virologia , Vacinas contra Doença de Marek/genética , Dados de Sequência Molecular , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , RNA Interferente Pequeno/química , RNA Interferente Pequeno/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA