Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39011896

RESUMO

Alu retrotransposons, which form the largest family of mobile DNA elements in the human genome, have recently come to attention as a potential source of regulatory novelties, most notably by participating in enhancer function. Even though Alu transcription by RNA polymerase III is subjected to tight epigenetic silencing, their expression has long been known to increase in response to various types of stress, including viral infection. Here we show that, in primary human fibroblasts, adenovirus small e1a triggered derepression of hundreds of individual Alus by promoting TFIIIB recruitment by Alu-bound TFIIIC. Epigenome profiling revealed an e1a-induced decrease of H3K27 acetylation and increase of H3K4 monomethylation at derepressed Alus, making them resemble poised enhancers. The enhancer nature of e1a-targeted Alus was confirmed by the enrichment, in their upstream regions, of the EP300/CBP acetyltransferase, EP400 chromatin remodeler and YAP1 and FOS transcription factors. The physical interaction of e1a with EP400 was critical for Alu derepression, which was abrogated upon EP400 ablation. Our data suggest that e1a targets a subset of enhancer Alus whose transcriptional activation, which requires EP400 and is mediated by the e1a-EP400 interaction, may participate in the manipulation of enhancer activity by adenoviruses.

2.
J Virol ; 97(12): e0099323, 2023 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-37962355

RESUMO

IMPORTANCE: Inactivation of EP300/CREBB paralogous cellular lysine acetyltransferases (KATs) during the early phase of infection is a consistent feature of DNA viruses. The cell responds by stabilizing transcription factor IRF3 which activates transcription of scores of interferon-stimulated genes (ISGs), inhibiting viral replication. Human respiratory adenoviruses counter this by assembling a CUL4-based ubiquitin ligase complex that polyubiquitinylates RUVBL1 and 2 inducing their proteasomal degradation. This inhibits accumulation of active IRF3 and the expression of anti-viral ISGs, allowing replication of the respiratory HAdVs in the face of inhibition of EP300/CBEBBP KAT activity by the N-terminal region of E1A.


Assuntos
ATPases Associadas a Diversas Atividades Celulares , Proteínas E1A de Adenovirus , Proteínas de Transporte , DNA Helicases , Imunidade Inata , Complexo de Endopeptidases do Proteassoma , Estresse Fisiológico , Humanos , Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/enzimologia , Adenovírus Humanos/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Culina/metabolismo , DNA Helicases/metabolismo , Interferons/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Quaternária de Proteína , Complexos Ubiquitina-Proteína Ligase/química , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitinação , Replicação Viral
3.
Elife ; 102021 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-33704060

RESUMO

Regulation of RNA polymerase II (Pol2) elongation in the promoter-proximal region is an important and ubiquitous control point for gene expression in metazoans. We report that transcription of the adenovirus 5 E4 region is regulated during the release of paused Pol2 into productive elongation by recruitment of the super-elongation complex, dependent on promoter H3K18/27 acetylation by CBP/p300. We also establish that this is a general transcriptional regulatory mechanism that applies to ~7% of expressed protein-coding genes in primary human airway epithelial cells. We observed that a homeostatic mechanism maintains promoter, but not enhancer, H3K18/27ac in response to extensive inhibition of CBP/p300 acetyl transferase activity by the highly specific small molecule inhibitor A-485. Further, our results suggest a function for BRD4 association at enhancers in regulating paused Pol2 release at nearby promoters. Taken together, our results uncover the processes regulating transcriptional elongation by promoter region histone H3 acetylation and homeostatic maintenance of promoter, but not enhancer, H3K18/27ac in response to inhibition of CBP/p300 acetyl transferase activity.


Assuntos
Regulação da Expressão Gênica , Histonas/metabolismo , Homeostase , Fatores de Transcrição de p300-CBP/genética , Acetilação , Linhagem Celular , Humanos , Fatores de Transcrição de p300-CBP/metabolismo
4.
Genes Dev ; 33(13-14): 828-843, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31171701

RESUMO

Adenovirus transformed cells have a dedifferentiated phenotype. Eliminating E1A in transformed human embryonic kidney cells derepressed ∼2600 genes, generating a gene expression profile closely resembling mesenchymal stem cells (MSCs). This was associated with a dramatic change in cell morphology from one with scant cytoplasm and a globular nucleus to one with increased cytoplasm, extensive actin stress fibers, and actomyosin-dependent flattening against the substratum. E1A-induced hypoacetylation at histone H3 Lys27 and Lys18 (H3K27/18) was reversed. Most of the increase in H3K27/18ac was in enhancers near TEAD transcription factors bound by Hippo signaling-regulated coactivators YAP and TAZ. E1A causes YAP/TAZ cytoplasmic sequestration. After eliminating E1A, YAP/TAZ were transported into nuclei, where they associated with poised enhancers with DNA-bound TEAD4 and H3K4me1. This activation of YAP/TAZ required RHO family GTPase signaling and caused histone acetylation by p300/CBP, chromatin remodeling, and cohesin loading to establish MSC-associated enhancers and then superenhancers. Consistent results were also observed in primary rat embryo kidney cells, human fibroblasts, and human respiratory tract epithelial cells. These results together with earlier studies suggest that YAP/TAZ function in a developmental checkpoint controlled by signaling from the actin cytoskeleton that prevents differentiation of a progenitor cell until it is in the correct cellular and tissue environment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas E1A de Adenovirus/metabolismo , Diferenciação Celular/genética , Inativação Gênica , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fosfoproteínas/genética , Citoesqueleto de Actina/metabolismo , Adenoviridae , Animais , Células Cultivadas , Células HEK293 , Humanos , Ratos , Transdução de Sinais , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
5.
J Virol ; 92(18)2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29976669

RESUMO

How histone acetylation promotes transcription is not clearly understood. Here, we confirm an interaction between p300 and the adenovirus 2 large E1A activation domain (AD) and map the interacting regions in E1A by observing colocalization at an integrated lacO array of fusions of LacI-mCherry to E1A fragments with YFP-p300. Viruses with mutations in E1A subdomains were constructed and analyzed for kinetics of early viral RNA expression and association of acetylated H3K9, K18, K27, TBP, and RNA polymerase II (Pol II) across the viral genome. The results indicate that this E1A interaction with p300 is required for H3K18 and H3K27 acetylation at the E2early, E3, and E4 promoters and is required for TBP and Pol II association with the E2early promoter. In contrast, H3K18/27 acetylation was not required for TBP and Pol II association with the E3 and E4 promoters but was required for E4 transcription at a step subsequent to Pol II preinitiation complex assembly.IMPORTANCE Despite a wealth of data associating promoter and enhancer region histone N-terminal tail lysine acetylation with transcriptional activity, there are relatively few examples of studies that establish causation between these histone posttranslational modifications and transcription. While hypoacetylation of histone H3 lysines 18 and 27 is associated with repression, the step(s) in the overall process of transcription that is blocked at a hypoacetylated promoter is not clearly established in most instances. Studies presented here confirm that the adenovirus 2 large E1A protein activation domain interacts with p300, as reported previously (P. Pelka, J. N. G. Ablack, J. Torchia, A. S. Turnell, R. J. A. Grand, J. S. Mymryk, Nucleic Acids Res 37:1095-1106, 2009, https://doi.org/10.1093/nar/gkn1057), and that the resulting acetylation of H3K18/27 affects varied steps in transcription at different viral promoters.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Histonas/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica , Acetilação , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica , Humanos , RNA Polimerase II/metabolismo , Ativação Transcricional
6.
Cell Host Microbe ; 22(6): 789-800.e5, 2017 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-29241042

RESUMO

The N-terminal half of adenovirus e1a assembles multimeric complexes with host proteins that repress innate immune responses and force host cells into S-phase. In contrast, the functions of e1a's C-terminal interactions with FOXK, DCAF7, and CtBP are unknown. We found that these interactions modulate RAS signaling, and that a single e1a molecule must bind all three of these host proteins to suppress activation of a subset of IFN-stimulated genes (ISGs). These ISGs were otherwise induced in primary respiratory epithelial cells at 12 hr p.i. This delayed activation of ISGs required IRF3 and coincided with an ∼10-fold increase in IRF3 from protein stabilization. The induced IRF3 bound to chromatin and localized to the promoters of activated ISGs. While IRF3, STAT1/2, and IRF9 all greatly increased in concentration, there were no corresponding mRNA increases, suggesting that e1a regulates the stabilities of these key activators of innate immune responses, as shown directly for IRF3.


Assuntos
Adenoviridae/imunologia , Proteínas E1A de Adenovirus/metabolismo , Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Imunidade Inata , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Oxirredutases do Álcool/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Fator Regulador 3 de Interferon/metabolismo , Ligação Proteica
8.
Cell Host Microbe ; 16(5): 663-76, 2014 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-25525796

RESUMO

Oncogenic transformation by adenovirus small e1a depends on simultaneous interactions with the host lysine acetylases p300/CBP and the tumor suppressor RB. How these interactions influence cellular gene expression remains unclear. We find that e1a displaces RBs from E2F transcription factors and promotes p300 acetylation of RB1 K873/K874 to lock it into a repressing conformation that interacts with repressive chromatin-modifying enzymes. These repressing p300-e1a-RB1 complexes specifically interact with host genes that have unusually high p300 association within the gene body. The TGF-β, TNF-, and interleukin-signaling pathway components are enriched among such p300-targeted genes. The p300-e1a-RB1 complex condenses chromatin in a manner dependent on HDAC activity, p300 lysine acetylase activity, the p300 bromodomain, and RB K873/K874 and e1a K239 acetylation to repress host genes that would otherwise inhibit productive virus infection. Thus, adenovirus employs e1a to repress host genes that interfere with viral replication.


Assuntos
Adenoviridae/genética , Proteínas E1A de Adenovirus/metabolismo , Proteína do Retinoblastoma/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Adenoviridae/fisiologia , Proteínas E1A de Adenovirus/genética , Transformação Celular Viral , Células Cultivadas , Quimiocina CXCL1/metabolismo , Cromatina/metabolismo , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Replicação Viral
9.
J Virol Methods ; 192(1-2): 28-38, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23624118

RESUMO

Adenoviruses are employed in the study of cellular processes and as expression vectors used in gene therapy. The success and reproducibility of these studies is dependent in part on having accurate and meaningful titers of replication competent and helper-dependent adenovirus stocks, which is problematic due to the use of varied and divergent titration protocols. Physical titration methods, which quantify the total number of viral particles, are used by many, but are poor at estimating activity. Biological titration methods, such as plaque assays, are more biologically relevant, but are time consuming and not applicable to helper-dependent gene therapy vectors. To address this, a protocol was developed called "infectious genome titration" in which viral DNA is isolated from the nuclei of cells ~3 h post-infection, and then quantified by Q-PCR. This approach ensures that only biologically active virions are counted as part of the titer determination. This approach is rapid, robust, sensitive, reproducible, and applicable to all forms of adenovirus. Unlike other Q-PCR-based methods, titers determined by this protocol are well correlated with biological activity.


Assuntos
Adenoviridae/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real/métodos , Carga Viral/métodos , Animais , DNA Viral/isolamento & purificação , Feminino , Vetores Genéticos/isolamento & purificação , Humanos , Camundongos Nus , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Manejo de Espécimes/métodos
10.
Proc Natl Acad Sci U S A ; 109(51): 20913-8, 2012 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-23213214

RESUMO

Although aberrant protein aggregation has been conclusively linked to dozens of devastating amyloid diseases, scientists remain puzzled about the molecular features that render amyloid fibrils or small oligomers toxic. Here, we report a previously unobserved type of amyloid fibril that tests as cytotoxic: one in which the strands of the contributing ß-sheets are out of register. In all amyloid fibrils previously characterized at the molecular level, only in-register ß-sheets have been observed, in which each strand makes its full complement of hydrogen bonds with the strands above and below it in the fibril. In out-of-register sheets, strands are sheared relative to one another, leaving dangling hydrogen bonds. Based on this finding, we designed out-of-register ß-sheet amyloid mimics, which form both cylindrin-like oligomers and fibrils, and these mimics are cytotoxic. Structural and energetic considerations suggest that out-of-register fibrils can readily convert to toxic cylindrins. We propose that out-of-register ß-sheets and their related cylindrins are part of a toxic amyloid pathway, which is distinct from the more energetically favored in-register amyloid pathway.


Assuntos
Amiloide/química , Vermelho Congo/farmacologia , Cristalografia por Raios X/métodos , Corantes Fluorescentes/farmacologia , Humanos , Ligação de Hidrogênio , Microscopia Eletrônica de Transmissão/métodos , Modelos Moleculares , Conformação Molecular , Peptídeos/química , Conformação Proteica , Estrutura Secundária de Proteína , Proteínas/química , Termodinâmica , Difração de Raios X , Microglobulina beta-2/química
12.
Genome Res ; 22(7): 1212-21, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22499665

RESUMO

Adenovirus small e1a oncoprotein causes ~70% reduction in cellular levels of histone H3 lysine 18 acetylation (H3K18ac). It is unclear, however, where this dramatic reduction occurs genome-wide. ChIP-sequencing revealed that by 24 h after expression, e1a erases 95% of H3K18ac peaks in normal, contact-inhibited fibroblasts and replaces them with one-third as many at new genomic locations. The H3K18ac peaks at promoters and intergenic regions of genes with fibroblast-related functions are eliminated after infection, and new H3K18ac peaks are established at promoters of highly induced genes that regulate cell cycling and at new putative enhancers. Strikingly, the regions bound by the retinoblastoma family of proteins in contact-inhibited fibroblasts gain new peaks of H3K18ac in the e1a-expressing cells, including 55% of RB1-bound loci. In contrast, over half of H3K9ac peaks are similarly distributed before and after infection, independently of RB1. The strategic redistribution of H3K18ac by e1a highlights the importance of this modification for transcriptional activation and cellular transformation as well as functional differences between the RB-family member proteins.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/genética , Epigênese Genética , Genoma Humano , Histonas/metabolismo , Acetilação , Proteínas E1A de Adenovirus/genética , Adenovírus Humanos/metabolismo , Adenovírus Humanos/patogenicidade , Ciclo Celular , Transformação Celular Viral , Células Cultivadas , Imunoprecipitação da Cromatina , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Histonas/genética , Humanos , Pulmão/metabolismo , Pulmão/patologia , Pulmão/virologia , Anotação de Sequência Molecular/métodos , Nucleossomos/genética , Nucleossomos/metabolismo , Regiões Promotoras Genéticas , Fatores de Tempo , Ativação Transcricional
13.
Virology ; 420(1): 1-9, 2011 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21917285

RESUMO

SV40 is comprised of the viral minichromosome and the capsid proteins VP1, VP2, and VP3. Complete reconstitution of SV40 virions in vitro remains a challenge. Here we describe in vitro reconstitution of SV40 particles that contain ~5-kb circular nucleosomal DNA with hyperacetylated histones and are encapsidated in a coat composed of VP1, VP2, and VP3, closely mimicking the characteristics of authentic SV40 virions. When inoculated into mammalian cells, VP1/2/3 particles containing nucleosomal DNA carrying a reporter gene yielded a significantly higher level of gene expression than VP1-only particles containing the corresponding naked DNA. The elevated gene expression resulted mainly from enhanced association of the particles with the cell surface and from facilitation of subsequent uptake into cells. Thus, the in vitro reconstitution system reported here should be useful for the elucidation of Polyomaviridae assembly mechanisms and for the development of novel carriers for gene delivery.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA/genética , Técnicas de Transferência de Genes/instrumentação , Nucleossomos/genética , Vírus 40 dos Símios/fisiologia , Vírion/fisiologia , Montagem de Vírus , Animais , Proteínas do Capsídeo/genética , Linhagem Celular , DNA/metabolismo , Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Nucleossomos/metabolismo , Vírus 40 dos Símios/genética , Vírion/genética
14.
J Virol ; 84(23): 12210-25, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20861261

RESUMO

Oncogenic transformation by adenovirus E1A and E1B-55K requires E1B-55K inhibition of p53 activity to prevent E1A-induced apoptosis. During viral infection, E1B-55K and E4orf6 substitute for the substrate-binding subunits of the host cell cullin 5 class of ubiquitin ligases, resulting in p53 polyubiquitinylation and proteasomal degradation. Here we show that E1B-55K alone also functions as an E3 SUMO1-p53 ligase. Fluorescence microscopy studies showed that E1B-55K alone, in the absence of other viral proteins, causes p53 to colocalize with E1B-55K in promyelocytic leukemia (PML) nuclear bodies, nuclear domains with a high concentration of sumoylated proteins. Photobleaching experiments with live cells revealed that E1B-55K tethering of p53 in PML nuclear bodies decreases the in vivo nuclear mobility of p53 nearly 2 orders of magnitude. E1B-55K-induced p53 sumoylation contributes to maximal inhibition of p53 function since mutation of the major p53 sumoylation site decreases E1B-55K-induced p53 sumoylation, tethering in PML nuclear bodies, and E1B-55K inhibition of p53 activity. Mutation of the E1B-55K sumoylation site greatly inhibits E1B-55K association with PML nuclear bodies and the p53 nuclear export to cytoplasmic aggresomes observed in E1A-E1B-transformed cells. Purified E1B-55K and p53 form high-molecular-weight complexes potentially through the formation of a network of E1B-55K dimers bound to the N termini of p53 tetramers. In support of this model, a p53 mutation that prevents tetramer formation greatly reduces E1B-55K-induced tethering in PML nuclear bodies and p53 nuclear export. These data indicate that E1B-55K's association with PML nuclear bodies inactivates p53 by first sequestering it in PML nuclear bodies and then greatly facilitating its nuclear export.


Assuntos
Proteínas E1B de Adenovirus/metabolismo , Transformação Celular Neoplásica/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteína SUMO-1/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Proteínas E1B de Adenovirus/genética , Linhagem Celular Tumoral , Dimerização , Humanos , Microscopia de Fluorescência , Modelos Biológicos , Mutação/genética , Fotodegradação , Proteína da Leucemia Promielocítica , Sumoilação
15.
Nat Rev Genet ; 10(5): 290-4, 2009 05.
Artigo em Inglês | MEDLINE | ID: mdl-19290008

RESUMO

The cancerous cellular state is associated with multiple epigenetic alterations, but elucidating the precise order of such alterations during tumorigenic progression and their contributions to the transformed phenotype remains a significant challenge in cancer biology. Here we discuss recent findings on how viral oncoproteins exploit specific epigenetic processes to coerce normal cells to replicate when they should remain quiescent - a hallmark of cancer. These findings may highlight roles of epigenetic processes in normal biology and shed light on epigenetic events occurring along the path of non-viral neoplastic transformation.


Assuntos
Transformação Celular Viral/genética , Epigênese Genética/genética , Genoma , Proteínas Oncogênicas Virais/metabolismo , Animais , Humanos , Neoplasias/genética , Neoplasias/metabolismo
16.
J Virol ; 83(7): 3249-57, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158239

RESUMO

To make a safe, long-lasting gene delivery vehicle, we developed a hybrid vector that leverages the relative strengths of adenovirus and Epstein-Barr virus (EBV). A fully gene-deleted helper-dependent adenovirus (HDAd) is used as the delivery vehicle for its scalability and high transduction efficiency. Upon delivery, a portion of the HDAd vector is recombined to form a circular plasmid. This episome includes two elements from EBV: an EBV nuclear antigen 1 (EBNA1) expression cassette and an EBNA1 binding region. Along with a human replication origin, these elements provide considerable genetic stability to the episome in replicating cells while avoiding insertional mutagenesis. Here, we demonstrate that this hybrid approach is highly efficient at delivering EBV episomes to target cells in vivo. We achieved nearly 100% transduction of hepatocytes after a single intravenous injection in mice. This is a substantial improvement over the transduction efficiency of previously available physical and viral methods. Bioluminescent imaging of vector-transduced mice demonstrated that luciferase transgene expression from the hybrid was robust and compared well to a traditional HDAd vector. Quantitative PCR analysis confirmed that the EBV episome was stable at approximately 30 copies per cell for up to 50 weeks and that it remained circular and extrachromosomal. Approaches for adapting the HDAd-EBV hybrid to a variety of disease targets and the potential benefits of this approach are discussed.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Hepatócitos/virologia , Herpesvirus Humano 4/genética , Plasmídeos , Transdução Genética , Animais , Feminino , Genes Reporter , Vetores Genéticos/administração & dosagem , Instabilidade Genômica , Injeções Intravenosas , Luciferases/genética , Luciferases/metabolismo , Luminescência , Camundongos , Camundongos Nus , Imagem Corporal Total
17.
Science ; 321(5892): 1084-5, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18719283

RESUMO

Adenovirus small early region 1a (e1a) protein drives cells into S phase by binding RB family proteins and the closely related histone acetyl transferases p300 and CBP. The interaction with RB proteins displaces them from DNA-bound E2F transcription factors, reversing their repression of cell cycle genes. However, it has been unclear how the e1a interaction with p300 and CBP promotes passage through the cell cycle. We show that this interaction causes a threefold reduction in total cellular histone H3 lysine 18 acetylation (H3K18ac). CBP and p300 are required for acetylation at this site because their knockdown causes specific hypoacetylation at H3K18. SV40 T antigen also induces H3K18 hypoacetylation. Because global hypoacetylation at this site is observed in prostate carcinomas with poor prognosis, this suggests that processes resulting in global H3K18 hypoacetylation may be linked to oncogenic transformation.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/metabolismo , Ciclo Celular , Histonas/metabolismo , Acetilação , Proteínas E1A de Adenovirus/genética , Antígenos Transformantes de Poliomavirus/metabolismo , Proteína de Ligação a CREB/metabolismo , Linhagem Celular , Transformação Celular Viral , Células Cultivadas , Células HeLa , Humanos , Lisina/metabolismo , Mutação , Fatores de Transcrição de p300-CBP/metabolismo
18.
Science ; 321(5892): 1086-8, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18719284

RESUMO

Adenovirus e1a induces quiescent human cells to replicate. We found that e1a causes global relocalization of the RB (retinoblastoma) proteins (RB, p130, and p107) and p300/CBP histone acetyltransferases on promoters, the effect of which is to restrict the acetylation of histone 3 lysine-18 (H3K18ac) to a limited set of genes, thereby stimulating cell cycling and inhibiting antiviral responses and cellular differentiation. Soon after expression, e1a binds transiently to promoters of cell cycle and growth genes, causing enrichment of p300/CBP, PCAF (p300/CBP-associated factor), and H3K18ac; depletion of RB proteins; and transcriptional activation. e1a also associates transiently with promoters of antiviral genes, causing enrichment for RB, p130, and H4K16ac; increased nucleosome density; and transcriptional repression. At later times, e1a and p107 bind mainly to promoters of development and differentiation genes, repressing transcription. The temporal order of e1a binding requires its interactions with p300/CBP and RB proteins. Our data uncover a defined epigenetic reprogramming leading to cellular transformation.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Adenovírus Humanos/metabolismo , Epigênese Genética , Proteínas E1A de Adenovirus/genética , Adenovírus Humanos/genética , Proteína de Ligação a CREB/metabolismo , Transformação Celular Viral , Proteína Substrato Associada a Crk/genética , Proteína Substrato Associada a Crk/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Histonas/metabolismo , Humanos , Mutação , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteína p107 Retinoblastoma-Like/genética , Proteína p107 Retinoblastoma-Like/metabolismo , Transcrição Gênica , Fatores de Transcrição de p300-CBP/metabolismo
19.
J Virol ; 81(2): 575-87, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17079297

RESUMO

Theadenovirus type 5 (Ad5) E1B-55K and E4orf6 proteins are required together to stimulate viral late nuclear mRNA export to the cytoplasm and to restrict host cell nuclear mRNA export during the late phase of infection. Previous studies have shown that these two viral proteins interact with the cellular proteins elongins B and C, cullin 5, RBX1, and additional cellular proteins to form an E3 ubiquitin-protein ligase that polyubiquitinates p53 and probably one or more subunits of the MRE11-RAD50-NBS1 (MRN) complex, directing their proteasomal degradation. The MRN complex is required for cellular DNA double-strand break repair and induction of the DNA damage response by adenovirus infection. To determine if the ability of E1B-55K and E4orf6 to stimulate viral late mRNA nuclear export requires the ubiquitin-protein ligase activity of this viral ubiquitin-protein ligase complex, we designed and expressed a dominant-negative mutant form of cullin 5 in HeLa cells before infection with wild-type Ad5 or the E1B-55K null mutant dl1520. The dominant-negative cullin 5 protein stabilized p53 and the MRN complex, indicating that it inhibited the viral ubiquitin-protein ligase but had no effect on viral early mRNA synthesis, early protein synthesis, or viral DNA replication. However, expression of the dominant-negative cullin 5 protein caused a decrease in viral late protein synthesis and viral nuclear mRNA export similar to the phenotype produced by mutations in E1B-55K. We conclude that the stimulation of adenovirus late mRNA nuclear export by E1B-55K and E4orf6 results from the ubiquitin-protein ligase activity of the adenovirus ubiquitin-protein ligase complex.


Assuntos
Transporte Ativo do Núcleo Celular , Adenovírus Humanos/enzimologia , RNA Mensageiro/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas E1B de Adenovirus/genética , Proteínas E1B de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/genética , Proteínas E4 de Adenovirus/metabolismo , Adenovírus Humanos/genética , Adenovírus Humanos/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Proteínas Culina/metabolismo , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Fases de Leitura Aberta , RNA Mensageiro/genética , Ubiquitina-Proteína Ligases/química , Ubiquitina-Proteína Ligases/genética
20.
FASEB J ; 20(7): 935-46, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16675851

RESUMO

We describe an inducible genetic model for degeneration of midbrain dopaminergic neurons in adults. In previous studies, knock-in mice expressing hypersensitive M2 domain Leu9'Ser (L9'S) alpha4 nicotinic receptors (nAChR) at near-normal levels displayed dominant neonatal lethality and dopaminergic deficits in embryonic midbrain, because the hypersensitive nAChR is excitotoxic. However, heterozygous L9'S mice that retain the neomycin resistance cassette (neo) in a neighboring intron express low levels of the mutant allele (approximately 25% of normal levels), and these neo-intact mice are therefore viable and fertile. The neo cassette is flanked by loxP sites. In adult animals, we locally injected helper-dependent adenovirus (HDA) expressing cre recombinase. Local excision of the neo cassette, via cre-mediated recombination, was verified by genomic analysis. In L9'S HDA-cre injected animals, locomotion was reduced both under baseline conditions and after amphetamine application. There was no effect in L9'S HDA-control treated animals or in wild-type (WT) littermates injected with either virus. Immunocytochemical analyses revealed marked losses (> 70%) of dopaminergic neurons in L9'S HDA-cre injected mice compared to controls. At 20-33 days postinjection in control animals, the coexpressed marker gene, yellow fluorescent protein (YFP), was expressed in many neurons and few glial cells near the injection, emphasizing the neurotropic utility of the HDA. Thus, HDA-mediated gene transfer into adult midbrain induced sufficient functional expression of cre in dopaminergic neurons to allow for postnatal deletion of neo. This produced increased L9'S mutant nAChR expression, which in turn led to nicotinic cholinergic excitotoxicity in dopaminergic neurons.


Assuntos
Dopamina/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Receptores Nicotínicos/genética , Receptores Nicotínicos/metabolismo , Substância Negra/patologia , Adenoviridae , Animais , Animais Geneticamente Modificados , Morte Celular , Regulação da Expressão Gênica , Locomoção/fisiologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA