Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nucleic Acids Res ; 50(1): 490-511, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34893887

RESUMO

In infected cells, Epstein-Barr virus (EBV) alternates between latency and lytic replication. The viral bZIP transcription factor ZEBRA (Zta, BZLF1) regulates this cycle by binding to two classes of ZEBRA response elements (ZREs): CpG-free motifs resembling the consensus AP-1 site recognized by cellular bZIP proteins and CpG-containing motifs that are selectively bound by ZEBRA upon cytosine methylation. We report structural and mutational analysis of ZEBRA bound to a CpG-methylated ZRE (meZRE) from a viral lytic promoter. ZEBRA recognizes the CpG methylation marks through a ZEBRA-specific serine and a methylcytosine-arginine-guanine triad resembling that found in canonical methyl-CpG binding proteins. ZEBRA preferentially binds the meZRE over the AP-1 site but mutating the ZEBRA-specific serine to alanine inverts this selectivity and abrogates viral replication. Our findings elucidate a DNA methylation-dependent switch in ZEBRA's transactivation function that enables ZEBRA to bind AP-1 sites and promote viral latency early during infection and subsequently, under appropriate conditions, to trigger EBV lytic replication by binding meZREs.


Assuntos
DNA Viral/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , Transativadores/metabolismo , Proteínas Virais/metabolismo , Metilação de DNA , Regulação Viral da Expressão Gênica , Células HEK293 , Humanos , Ligação Proteica , Replicação Viral
2.
Mamm Genome ; 34(3): 408-417, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37468728

RESUMO

Over the last decade, INFRAFRONTIER has positioned itself as a world-class Research Infrastructure for the generation, phenotyping, archiving, and distribution of mouse models in Europe. The INFRAFRONTIER network consists of 22 partners from 15 countries, and is continuously enhancing and broadening its portfolio of resources and services that are offered to the research community on a non-profit basis. By bringing together European rodent model expertise and providing valuable disease model services to the biomedical research community, INFRAFRONTIER strives to push the accessibility of cutting-edge human disease modelling technologies across the European research landscape. This article highlights the latest INFRAFRONTIER developments and informs the research community about its extensively utilised services, resources, and technical developments, specifically the intricacies of the INFRAFRONTIER database, use of Curated Disease Models, overview of the INFRAFRONTIER Cancer and Rare Disease resources, and information about its main state-of-the-art services.


Assuntos
Pesquisa Biomédica , Camundongos , Animais , Humanos , Modelos Animais de Doenças , Europa (Continente)
3.
J Virol ; 89(14): 7248-61, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25948739

RESUMO

UNLABELLED: The human herpesviruses Epstein-Barr virus (EBV) and Kaposi's sarcoma-associated herpesvirus (KSHV) are associated with Hodgkin's lymphoma (HL) and Primary effusion lymphomas (PEL), respectively, which are B cell malignancies that originate from germinal center B cells. PEL cells but also a quarter of EBV-positive HL tumor cells do not express the genuine B cell receptor (BCR), a situation incompatible with survival of normal B cells. EBV encodes LMP2A, one of EBV's viral latent membrane proteins, which likely replaces the BCR's survival signaling in HL. Whether KSHV encodes a viral BCR mimic that contributes to oncogenesis is not known because an experimental model of KSHV-mediated B cell transformation is lacking. We addressed this uncertainty with mutant EBVs encoding the KSHV genes K1 or K15 in lieu of LMP2A and infected primary BCR-negative (BCR(-)) human B cells with them. We confirmed that the survival of BCR(-) B cells and their proliferation depended on an active LMP2A signal. Like LMP2A, the expression of K1 and K15 led to the survival of BCR(-) B cells prone to apoptosis, supported their proliferation, and regulated a similar set of cellular target genes. K1 and K15 encoded proteins appear to have noncomplementing, redundant functions in this model, but our findings suggest that both KSHV proteins can replace LMP2A's key activities contributing to the survival, activation and proliferation of BCR(-) PEL cells in vivo. IMPORTANCE: Several herpesviruses encode oncogenes that are receptor-like proteins. Often, they are constitutively active providing important functions to the latently infected cells. LMP2A of Epstein-Barr virus (EBV) is such a receptor that mimics an activated B cell receptor, BCR. K1 and K15, related receptors of Kaposi's sarcoma-associated herpesvirus (KSHV) expressed in virus-associated tumors, have less obvious functions. We found in infection experiments that both viral receptors of KSHV can replace LMP2A and deliver functions similar to the endogenous BCR. K1, K15, and LMP2A also control the expression of a related set of cellular genes in primary human B cells, the target cells of EBV and KSHV. The observed phenotypes, as well as the known characteristics of these genes, argue for their contributions to cellular survival, B cell activation, and proliferation. Our findings provide one possible explanation for the tumorigenicity of KSHV, which poses a severe problem in immunocompromised patients.


Assuntos
Linfócitos B/fisiologia , Proliferação de Células , Herpesvirus Humano 4/fisiologia , Herpesvirus Humano 8/fisiologia , Proteínas da Matriz Viral/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Humanos
4.
Nucleic Acids Res ; 42(5): 3059-72, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24371273

RESUMO

CpG methylation in mammalian DNA is known to interfere with gene expression by inhibiting the binding of transactivators to their cognate sequence motifs or recruiting proteins involved in gene repression. An Epstein-Barr virus-encoded transcription factor, Zta, was the first example of a sequence-specific transcription factor that preferentially recognizes and selectively binds DNA sequence motifs with methylated CpG residues, reverses epigenetic silencing and activates gene transcription. The DNA binding domain of Zta is homologous to c-Fos, a member of the cellular AP-1 (activator protein 1) transcription factor family, which regulates cell proliferation and survival, apoptosis, transformation and oncogenesis. We have identified a novel AP-1 binding site termed meAP-1, which contains a CpG dinucleotide. If methylated, meAP-1 sites are preferentially bound by the AP-1 heterodimer c-Jun/c-Fos in vitro and in cellular chromatin in vivo. In activated human primary B cells, c-Jun/c-Fos locates to these methylated elements in promoter regions of transcriptionally activated genes. Reminiscent of the viral Zta protein, c-Jun/c-Fos is the first identified cellular member of the AP-1 family of transactivators that can induce expression of genes with methylated, hence repressed promoters, reversing epigenetic silencing.


Assuntos
Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Elementos Reguladores de Transcrição , Fator de Transcrição AP-1/metabolismo , Ativação Transcricional , 5-Metilcitosina/metabolismo , Linfócitos B/metabolismo , Sítios de Ligação , Linhagem Celular , Ilhas de CpG , DNA/química , DNA/metabolismo , Metilação de DNA , DNA Viral/química , DNA Viral/metabolismo , Dimerização , Genoma Humano , Herpesvirus Humano 4/genética , Humanos , Motivos de Nucleotídeos , Regiões Promotoras Genéticas
5.
J Virol ; 85(4): 1732-46, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21106746

RESUMO

Human cytomegalovirus (HCMV) infection causes a rapid induction of c-Fos and c-Jun, the major subunits of activator protein 1 (AP-1), which in turn have been postulated to activate the viral immediate-early (IE) genes. Accordingly, the major IE promoter (MIEP) enhancer, a critical control region for initiating lytic HCMV infection and reactivation from the latent state, contains one well-characterized AP-1 site and a second candidate interaction site. In this study we explored the role of these AP-1 elements in the context of the infection. We first show that the distal candidate AP-1 motif binds c-Fos/c-Jun heterodimers (AP-1 complex) and confers c-Fos/c-Jun-mediated activity to a core promoter. Site-directed mutagenesis studies indicate that both AP-1 response elements are critical for 12-O-tetradecanoylphorbol-13-acetate (TPA)-enhanced MIEP activity in transient-transfection assays. In marked contrast to the results obtained with the isolated promoter, disruption of the AP-1 recognition sites of the MIEP in the context of the infectious HCMV genome has no significant influence on the expression of the MIE protein IE1 or viral replication in different cell types. Moreover, a chimeric murine CMV driven by the HCMV MIEP (hMCMV-ES) with the two AP-1 binding sites mutated is not compromised in virulence, is able to grow and disseminate to different organs of the newborn mice as efficiently as the parental virus, and is competent in reactivation. We show, however, that combined inactivation of the enhancer AP-1 and NF-κB recognition sites leads to an attenuation of the hMCMV-ES in the neonatal murine infection model, not observed when each single element is abolished. Altogether, these results underline the functional redundancy of the MIEP elements, highlighting the plasticity of this region, which probably evolved to ensure maximal transcriptional performance across many diverse environments.


Assuntos
Motivos de Aminoácidos , Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , Elementos Facilitadores Genéticos , Proteínas Imediatamente Precoces/química , NF-kappa B/metabolismo , Fator de Transcrição AP-1/metabolismo , Doença Aguda , Motivos de Aminoácidos/genética , Animais , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Citomegalovirus/genética , Citomegalovirus/metabolismo , Modelos Animais de Doenças , Genes Precoces , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/genética , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Ligação Proteica , Elementos de Resposta , Replicação Viral
6.
J Virol ; 82(20): 10302-7, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684819

RESUMO

The human cytomegalovirus (HCMV) major immediate-early enhancer has been postulated to play a pivotal role in the control of latency and reactivation. However, the absence of an animal model has obstructed a direct test of this hypothesis. Here we report on the establishment of an in vivo, experimentally tractable system for quantitatively investigating physiological functions of the HCMV enhancer. Using a neonate BALB/c mouse model, we show that a chimeric murine CMV under the control of the HCMV enhancer is competent in vivo, replicating in key organs of mice with acute CMV infection and exhibiting latency/reactivation features comparable for the most part to those of the parental and revertant viruses.


Assuntos
Infecções por Citomegalovirus/genética , Citomegalovirus/genética , Elementos Facilitadores Genéticos , Proteínas Imediatamente Precoces/genética , Muromegalovirus , Ativação Viral/fisiologia , Latência Viral/fisiologia , Animais , Animais Recém-Nascidos , Linhagem Celular , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/patologia , Modelos Animais de Doenças , Feminino , Humanos , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Muromegalovirus/genética , Muromegalovirus/metabolismo , Muromegalovirus/patogenicidade , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Replicação Viral
7.
J Virol ; 80(19): 9899-904, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16973595

RESUMO

The role of NF-kappaB in regulating human cytomegalovirus (HCMV) replication and gene transcription remains controversial. Multiple, functional NF-kappaB response elements exist in the major immediate-early promoter (MIEP) enhancer of HCMV, suggesting a possible requirement for this transcription factor in lytic viral replication. Here we demonstrate by generating and analyzing HCMVs with alterations in the MIEP-enhancer that, although this region is essential for HCMV growth, none of the four NF-kappaB response elements contained within the enhancer are required for MIE gene expression or HCMV replication in multiple cell types. These data reveal the robustness of the regulatory network controlling the MIEP enhancer.


Assuntos
Citomegalovirus/genética , Citomegalovirus/fisiologia , Elementos Facilitadores Genéticos/genética , Regulação Viral da Expressão Gênica/genética , NF-kappa B/metabolismo , Transcrição Gênica/genética , Replicação Viral , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Deleção de Genes , Humanos , Cinética , Dados de Sequência Molecular , Mutação/genética , Regiões Promotoras Genéticas/genética
8.
J Virol ; 79(11): 7182-94, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15890957

RESUMO

The major immediate-early (MIE) genes of cytomegaloviruses (CMV) are broadly thought to be decisive regulators of lytic replication and reactivation from latency. To directly assess the role of the MIE protein IE1 during the infection of murine CMV (MCMV), we constructed an MCMV with exon 4 of the ie1 gene deleted. We found that, independent of the multiplicity of infection, the resulting recombinant virus, MCMVdie1, which fails to express the IE1 protein, was fully competent for early gene expression and replicated in different cultured cell types with identical kinetics to those of parental or revertant virus. Immunofluorescence microscopy studies revealed that MCMVdie1 was greatly impaired in its capacity to disrupt promyelocytic leukemia bodies in NIH 3T3 cells early after infection, a process that has been proposed to increase viral transcription efficiency. We examined MCMVdie1 in the murine model using both immunocompetent BALB/c and severe combined immunodeficient (SCID) mice. When MCMVdie1 was inoculated into these two types of mice, significantly lower viral titers were detected in infected organs than in those of the wild-type virus-infected animals. Moreover, the ie1-deficient MCMV exhibited a markedly reduced virulence. While all animals infected with 5 x 10(4) PFU of parental virus died by 30 days postinfection, SCID mice infected with a similar dose of MCMVdie1 did not succumb before 60 days postinfection. The in vivo defective growth phenotype of MCMVdie1 was abrogated upon rescue of ie1. These results demonstrate the significance of the ie1 gene for promoting an acute MCMV infection and virulence yet indicate that MCMV is able to grow in vivo, although impaired, in the absence of the ie1 gene.


Assuntos
Genes Precoces , Muromegalovirus/genética , Muromegalovirus/patogenicidade , Animais , Linhagem Celular , Células Cultivadas , Cromossomos Artificiais Bacterianos/genética , DNA Viral/genética , Deleção de Genes , Expressão Gênica , Proteínas Imediatamente Precoces/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Muromegalovirus/fisiologia , Células NIH 3T3 , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares/fisiologia , Fenótipo , Proteína da Leucemia Promielocítica , Recombinação Genética , Fatores de Transcrição/fisiologia , Proteínas Supressoras de Tumor , Proteínas Virais/genética , Virulência/genética , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA