Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 12386, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34117281

RESUMO

Parenteral nutrition-associated cholestasis (PNAC) significantly limits the safety of intravenous parenteral nutrition (PN). Critically ill infants are highly vulnerable to PNAC-related morbidity and mortality, however the impact of hepatic immaturity on PNAC is poorly understood. We examined developmental differences between fetal/infant and adult livers, and used human induced pluripotent stem cell-derived hepatocyte-like cells (iHLC) to gain insights into the contribution of development to altered sterol metabolism and PNAC. We used RNA-sequencing and computational techniques to compare gene expression patterns in human fetal/infant livers, adult liver, and iHLC. We identified distinct gene expression profiles between the human feta/infant livers compared to adult liver, and close resemblance of iHLC to human developing livers. Compared to adult, both developing livers and iHLC had significant downregulation of xenobiotic, bile acid, and fatty acid metabolism; and lower expression of the sterol metabolizing gene ABCG8. When challenged with stigmasterol, a plant sterol found in intravenous soy lipids, lipid accumulation was significantly higher in iHLC compared to adult-derived HepG2 cells. Our findings provide insights into altered bile acid and lipid metabolizing processes in the immature human liver, and support the use of iHLC as a relevant model system of developing liver to study lipid metabolism and PNAC.


Assuntos
Colestase/dietoterapia , Hepatócitos/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fígado/fisiopatologia , Nutrição Parenteral , Feminino , Humanos , Recém-Nascido , Masculino
2.
Biochem Cell Biol ; 97(2): 118-129, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30142277

RESUMO

We have previously shown that cooperative, interdependent binding by the pioneer factors FoxO1 and FoxA1/2 is required for recruitment of RNA polymerase II and H3K27 acetylation to the promoters of insulin-regulated genes. However, the underlying mechanisms are unknown. In this study, we demonstrate that, in HepG2 cells, FoxO1 and FoxA2 form a complex on DNA that is disrupted by insulin treatment. Insulin-mediated phosphorylation of FoxO1 and FoxA2 does not impair their cooperative binding to mononucleosome particles assembled from the IGFBP1 promoter, indicating that direct disruption of complex formation by phosphorylation is not responsible for the loss of interdependent FoxO1:FoxA1/2 binding following insulin treatment. Since FoxO1 and FoxA1/2 binding is required for the establishment and maintenance of transcriptionally active chromatin at insulin-regulated genes, we hypothesized that cooperative FoxO1 and FoxA1/2 binding dictates the chromatin remodeling events required for the initial activation of these genes. In support of this idea, we demonstrate that FoxO1 and FoxA2 cooperatively open linker histone compacted chromatin templates containing the IGFBP1 promoter. Taken together, these results provide a mechanism for how interdependent FoxO1:FoxA1/2 binding is negatively impacted by insulin and provide a developmental context for cooperative gene activation by these factors.


Assuntos
Cromatina/metabolismo , DNA/metabolismo , Proteína Forkhead Box O1/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Insulina/metabolismo , Cromatina/genética , DNA/genética , Proteína Forkhead Box O1/genética , Células Hep G2 , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/genética , Humanos , Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Fosforilação/genética , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Elementos de Resposta
3.
Genomics ; 107(6): 267-73, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27184763

RESUMO

Currently available methods for interrogating DNA-protein interactions at individual genomic loci have significant limitations, and make it difficult to work with unmodified cells or examine single-copy regions without specific antibodies. In this study, we describe a physiological application of the Hybridization Capture of Chromatin-Associated Proteins for Proteomics (HyCCAPP) methodology we have developed. Both novel and known locus-specific DNA-protein interactions were identified at the ENO2 and GAL1 promoter regions of Saccharomyces cerevisiae, and revealed subgroups of proteins present in significantly different levels at the loci in cells grown on glucose versus galactose as the carbon source. Results were validated using chromatin immunoprecipitation. Overall, our analysis demonstrates that HyCCAPP is an effective and flexible technology that does not require specific antibodies nor prior knowledge of locally occurring DNA-protein interactions and can now be used to identify changes in protein interactions at target regions in the genome in response to physiological challenges.


Assuntos
Proteínas de Ligação a DNA/genética , Galactoquinase/genética , Fosfopiruvato Hidratase/genética , Proteômica/métodos , Proteínas de Saccharomyces cerevisiae/genética , Cromatina/genética , Imunoprecipitação da Cromatina/métodos , Regiões Promotoras Genéticas , Ligação Proteica/genética , Saccharomyces cerevisiae/genética
4.
J Biol Chem ; 291(16): 8848-61, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26929406

RESUMO

FoxO1 binds to insulin response elements located in the promoters of insulin-like growth factor-binding protein 1 (IGFBP1) and glucose-6-phosphatase (G6Pase), activating their expression. Insulin-mediated phosphorylation of FoxO1 promotes cytoplasmic translocation, inhibiting FoxO1-mediated transactivation. We have previously demonstrated that FoxO1 opens and remodels chromatin assembled from the IGFBP1 promoter via a highly conserved winged helix motif. This finding, which established FoxO1 as a "pioneer" factor, suggested a model whereby FoxO1 chromatin remodeling at regulatory targets facilitates binding and recruitment of additional regulatory factors. However, the impact of FoxO1 phosphorylation on its ability to bind chromatin and the effect of FoxO1 loss on recruitment of neighboring transcription factors at its regulatory targets in liver chromatin is unknown. In this study, we demonstrate that an amino acid substitution that mimics insulin-mediated phosphorylation of a serine in the winged helix DNA binding motif curtails FoxO1 nucleosome binding. We also demonstrate that shRNA-mediated loss of FoxO1 binding to the IGFBP1 and G6Pase promoters in HepG2 cells significantly reduces binding of RNA polymerase II and the pioneer factors FoxA1/A2. Knockdown of FoxA1 similarly reduced binding of RNA polymerase II and FoxO1. Reduction in acetylation of histone H3 Lys-27 accompanies loss of FoxO1 and FoxA1/A2 binding. Interdependent binding of FoxO1 and FoxA1/A2 possibly entails cooperative binding because FoxO1 and FoxA1/A2 facilitate one another's binding to IGFPB1 promoter DNA. These results illustrate how transcription factors can nucleate transcriptional events in chromatin in response to signaling events and suggest a model for regulation of hepatic glucose metabolism through interdependent FoxO/FoxA binding.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Insulina/metabolismo , Regiões Promotoras Genéticas/fisiologia , Ativação Transcricional/fisiologia , Animais , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Células Hep G2 , Fator 3-alfa Nuclear de Hepatócito/genética , Fator 3-beta Nuclear de Hepatócito/genética , Humanos , Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/biossíntese , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Camundongos , Fosforilação/fisiologia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo
5.
PLoS One ; 6(10): e26217, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22028835

RESUMO

The regulation of gene transcription is fundamental to the existence of complex multicellular organisms such as humans. Although it is widely recognized that much of gene regulation is controlled by gene-specific protein-DNA interactions, there presently exists little in the way of tools to identify proteins that interact with the genome at locations of interest. We have developed a novel strategy to address this problem, which we refer to as GENECAPP, for Global ExoNuclease-based Enrichment of Chromatin-Associated Proteins for Proteomics. In this approach, formaldehyde cross-linking is employed to covalently link DNA to its associated proteins; subsequent fragmentation of the DNA, followed by exonuclease digestion, produces a single-stranded region of the DNA that enables sequence-specific hybridization capture of the protein-DNA complex on a solid support. Mass spectrometric (MS) analysis of the captured proteins is then used for their identification and/or quantification. We show here the development and optimization of GENECAPP for an in vitro model system, comprised of the murine insulin-like growth factor-binding protein 1 (IGFBP1) promoter region and FoxO1, a member of the forkhead rhabdomyosarcoma (FoxO) subfamily of transcription factors, which binds specifically to the IGFBP1 promoter. This novel strategy provides a powerful tool for studies of protein-DNA and protein-protein interactions.


Assuntos
DNA/genética , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/metabolismo , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/química , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Espectrometria de Massas , Proteômica/métodos , Animais , Sequência de Bases , Cromatina/metabolismo , Reagentes de Ligações Cruzadas/farmacologia , DNA/química , DNA/metabolismo , Escherichia coli/enzimologia , Exodesoxirribonucleases/metabolismo , Proteína Forkhead Box O1 , Formaldeído/farmacologia , Camundongos , Hibridização de Ácido Nucleico , Oligonucleotídeos/química , Oligonucleotídeos/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Especificidade por Substrato
6.
Virology ; 421(2): 167-72, 2011 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-22018782

RESUMO

Association of herpesvirus DNA with histones has important implications for lytic and latent infections; thus herpesviruses arbitrate interactions with histones to productively infect host cells. While regulation of alpha and betaherpesvirus chromatin during lytic infection has been actively investigated, very little is known about interaction of gammaherpesvirus DNA with histones upon de novo lytic infection. Murine gammaherpesvirus-68 (MHV68) is a rodent pathogen that offers a tractable system to study gammaherpesvirus lytic infection in primary cells. In this study we report that MHV68 promoter and orilyt sequences underwent dynamic association with histone H3 during de novo lytic infection of primary macrophages and fibroblasts. Similar to HSV-1, the degree of MHV68 DNA association with histone H3 was dependent on the multiplicity of infection and was further regulated by viral DNA synthesis. Our work sets a precedent for future studies of gammaherpesvirus chromatin during de novo lytic infection.


Assuntos
DNA Viral/metabolismo , Histonas/metabolismo , Rhadinovirus/genética , Rhadinovirus/fisiologia , Animais , Células Cultivadas , Cromatina/virologia , DNA Viral/biossíntese , DNA Viral/genética , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Histonas/genética , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Rhadinovirus/metabolismo , Proteínas Virais/genética
7.
Virology ; 420(2): 73-81, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21943826

RESUMO

Gammaherpesvirus protein kinases are an attractive therapeutic target as they support lytic replication and latency. Via an unknown mechanism these kinases enhance expression of select viral genes and DNA synthesis. Importantly, the kinase phenotypes have not been examined in primary cell types. Mouse gammaherpesvirus-68 (MHV68) protein kinase orf36 activates the DNA damage response (DDR) and facilitates lytic replication in primary macrophages. Significantly, H2AX, a DDR component and putative orf36 substrate, enhances MHV68 replication. Here we report that orf36 facilitated expression of RTA, an immediate early MHV68 gene, and DNA synthesis during de novo infection of primary macrophages. H2AX expression supported efficient RTA transcription and phosphorylated H2AX associated with RTA promoter. Furthermore, viral DNA synthesis was attenuated in H2AX-deficient macrophages, suggesting that the DDR system was exploited throughout the replication cycle. The interactions between a cancer-associated gammaherpesvirus and host tumor suppressor system have important implications for the pathogenesis of gammaherpesvirus infection.


Assuntos
Replicação do DNA/genética , Histonas/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Macrófagos/virologia , Proteínas Quinases/metabolismo , Rhadinovirus/genética , Transcrição Gênica , Proteínas Virais/metabolismo , Animais , Imunoprecipitação da Cromatina , Reparo do DNA , DNA Viral/biossíntese , Regulação Viral da Expressão Gênica , Histonas/deficiência , Histonas/genética , Proteínas Imediatamente Precoces/biossíntese , Proteínas Imediatamente Precoces/genética , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rhadinovirus/patogenicidade , Proteínas Virais/genética
8.
J Biol Chem ; 285(1): 464-72, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19897491

RESUMO

FoxA1-3 (formerly HNF3alpha, -beta, and -gamma), members of the FoxA subfamily of forkhead transcription factors, function as initial chromatin-binding and chromatin-remodeling factors in a variety of tissues, including liver and pancreas. Despite essential roles in development and metabolism, regulation of FoxA factors is not well understood. This study examines a potential role for acetylation in the regulation of FoxA chromatin binding and remodeling. Using in silico analysis, we have identified 11 putative p300 acetylation sites within FoxA1, five of which are located within wings 1 and 2 of its winged-helix DNA-binding domain. These polypeptide structures stabilize FoxA DNA and chromatin binding, and we have demonstrated that acetylation attenuates FoxA binding to DNA and diminishes its ability to remodel chromatin. FoxA acetylation is inhibited by chromatin binding. We propose a model whereby stable chromatin binding protects the FoxA DNA-binding domain from acetylation to preserve chromatin binding and remodeling by FoxA factors in the absence of extracellular cues.


Assuntos
Montagem e Desmontagem da Cromatina , Cromatina/metabolismo , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Acetilação , Animais , DNA/metabolismo , Elementos Facilitadores Genéticos/genética , Células Hep G2 , Hepatócitos/metabolismo , Humanos , Camundongos , Nucleossomos/metabolismo , Ligação Proteica , Fatores de Transcrição de p300-CBP/metabolismo
10.
Exp Eye Res ; 86(4): 586-600, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18291368

RESUMO

Maspin, a 42 kDa non-classical serpin (serine protease inhibitor) that controls cell migration and invasion, is mainly expressed by epithelial-derived cells but is also expressed in corneal stromal keratocytes. Upon culture of stromal keratocytes in the presence of FBS, maspin is down-regulated to nearly undetectable levels by passage two. DNA methylation is one of several processes that controls gene expression during cell differentiation, development, genetic imprinting, and carcinogenesis but has not been studied in corneal stromal cells. The purpose of this study was to determine whether DNA methylation of the maspin promoter and histone H3 dimethylation is involved in the mechanism of down-regulation of maspin synthesis in human corneal stromal fibroblasts and myofibroblasts. Human donor corneal stroma cells were immediately placed into serum-free defined medium or cultured in the presence of FBS and passed into serum-free medium or medium containing FBS or FGF-2 to induce the fibroblast phenotype or TGF-beta1 for the myofibroblast phenotype. These cell types are found in wounded corneas. The cells were used to prepare RNA for semi-quantitative or quantitative RT-PCR or to extract protein for Western analysis. In addition, P4 FBS cultured fibroblasts were treated with the DNA demethylating agent, 5-aza-2'-deoxycytidine (5-Aza-dC), and the histone deacetylase inhibitor, trichostatin A (TSA). Cells with and without treatment were harvested and assayed for DNA methylation using sodium bisulfite sequencing. The methylation state of histone H3 associated with the maspin gene in the P4 fibroblast cells was determined using a ChIP assay. Freshly harvested corneal stromal cells expressed maspin but upon phenotypic differentiation, maspin mRNA and protein were dramatically down-regulated. Sodium bisulfite sequencing revealed that the maspin promoter in the freshly isolated stromal keratocytes was hypomethylated while both the P0 stromal cells and the P1 cells cultured in the presence of serum-free defined medium, FGF-2 and TGF-beta1 were hypermethylated. Down-regulation of maspin synthesis was also associated with histone H3 dimethylation at lysine 9. Both maspin mRNA and protein were re-expressed at low levels with 5-Aza-dC but not TSA treatment. Addition of TSA to 5-Aza-dC treated cells did not increase maspin expression. Treatment with 5-Aza-dC did not significantly alter demethylation of the maspin promoter but did demethylate histone H3. These results show maspin promoter hypermethylation and histone methylation occur with down-regulation of maspin synthesis in corneal stromal cells and suggest regulation of genes upon conversion of keratocytes to wound healing fibroblasts can involve promoter and histone methylation.


Assuntos
Substância Própria/metabolismo , Proteínas do Olho/biossíntese , Fibroblastos/citologia , Inativação Gênica , Serpinas/biossíntese , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Sequência de Bases , Western Blotting/métodos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Substância Própria/citologia , Metilação de DNA , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Proteínas do Olho/genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Serpinas/genética
11.
J Biol Chem ; 282(49): 35583-93, 2007 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-17923482

RESUMO

FoxO1, a member of the forkhead rabdomyosarcoma (FoxO) subfamily of transcription factors, binds DNA via a highly conserved winged-helix "forkhead box" motif used by other regulatory proteins to mediate their effects through chromatin binding and remodeling. To examine how FoxO1 regulates target genes in chromatin, we studied the binding of purified recombinant FoxO1 protein to nucleosome particles and chromatin arrays containing the insulin-like growth factor-binding protein 1 promoter. We found that FoxO1 is able to bind to its cognate sites within the insulin-like growth factor-binding protein 1 promoter on a nucleosome. This binding stably perturbs core histone:DNA contacts extending up- and downstream from sites of FoxO1 binding without disrupting the underlying core particle. FoxO1 is able to harness these capabilities to bind to and de-condense linker histone-compacted chromatin arrays. Chromatin opening by FoxO1 requires both the N and C termini of the protein, which are also required for high affinity core histone binding and, in the case of the N terminus, nucleosome perturbation. We suggest that the chromatin binding and remodeling functions revealed here for FoxO1 endow all FoxO factors with the ability to initiate and dynamically modulate active chromatin states, enabling their diverse roles as gene regulatory factors in metabolism, cell survival, apoptosis, cell cycle progression, DNA repair, and protection against oxidative stress.


Assuntos
Montagem e Desmontagem da Cromatina/fisiologia , DNA/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Histonas/metabolismo , Nucleossomos/metabolismo , Animais , Apoptose/fisiologia , Ciclo Celular/fisiologia , Sobrevivência Celular/fisiologia , Sistema Livre de Células/metabolismo , DNA/genética , Reparo do DNA/fisiologia , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Histonas/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/metabolismo , Camundongos , Nucleossomos/genética , Estresse Oxidativo/fisiologia , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/fisiologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus laevis
12.
J Mol Biol ; 366(3): 720-4, 2007 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-17189638

RESUMO

FOX (forkhead box) transcription factors have diverse regulatory roles in development, signaling, and longevity, as well as being able to bind stably to target sites in silent chromatin. Crystal structure analysis showed that the FOXA DNA binding domain folds into a helix-turn-helix (HTH) motif flanked on either side by "wings" of polypeptide chain. The wings have the potential to interact with the DNA minor groove along the long axis of the DNA helix, flanking the HTH interactions with the major groove. Diverse FOX family homologs exist, and structural studies with certain DNA target sites suggest that neither of the wing regions are well ordered or provide a stable contribution to DNA target site binding. However, FOXA1 binds certain DNA target sites with high affinity, and as a monomer. To determine whether the wing domains contribute to stable DNA binding, we assessed complexes of FOXA with high and lower affinity DNA target sites by hydroxyl radical footprinting and site-directed mutagenesis. The data revealed clear protections predicted for wing interactions at the high affinity target, but less so at the lower affinity target, indicating that the wing domains stably interact with high affinity DNA sites for FOXA proteins.


Assuntos
DNA/metabolismo , Fator 3-alfa Nuclear de Hepatócito/química , Fator 3-alfa Nuclear de Hepatócito/metabolismo , Sequência de Bases , DNA/química , DNA/genética , Pegada de DNA , Radical Hidroxila , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , Ligação Proteica , Estrutura Terciária de Proteína
14.
J Virol ; 78(11): 5848-55, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15140982

RESUMO

Integration of viral DNA into the host chromosome is an obligatory step in retroviral replication and is dependent on the activity of the viral enzyme integrase. To examine the influence of chromatin structure on retroviral DNA integration in vitro, we used a model target comprising a 13-nucleosome extended array that includes binding sites for specific transcription factors and can be compacted into a higher-ordered structure. We found that the efficiency of in vitro integration catalyzed by human immunodeficiency virus type 1 (HIV-1) integrase was decreased after compaction of this target with histone H1. In contrast, integration by avian sarcoma virus (ASV) integrase was more efficient after compaction by either histone H1 or a high salt concentration, suggesting that the compacted structure enhances this reaction. Furthermore, although site-specific binding of transcription factors HNF3 and GATA4 blocked ASV DNA integration in extended nucleosome arrays, local opening of H1-compacted chromatin by HNF3 had no detectable effect on integration, underscoring the preference of ASV for compacted chromatin. Our results indicate that chromatin structure affects integration site selection of the HIV-1 and ASV integrases in opposite ways. These distinct properties of integrases may also affect target site selection in vivo, resulting in an important bias against or in favor of integration into actively transcribed host DNA.


Assuntos
Cromatina/virologia , Integrases/fisiologia , Retroviridae/genética , Integração Viral , Vírus do Sarcoma Aviário/enzimologia , Sítios de Ligação , Cromatina/química , Proteínas de Ligação a DNA/fisiologia , Fator de Transcrição GATA4 , Integrase de HIV/fisiologia , Fator 3-alfa Nuclear de Hepatócito , Proteínas Nucleares/fisiologia , Nucleossomos/metabolismo , Fatores de Transcrição/fisiologia
16.
Mol Cell Biol ; 23(8): 2633-44, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12665567

RESUMO

The Epstein-Barr virus (EBV)-encoded lytic activator Zta is a bZIP protein that can stimulate nucleosomal histone acetyltransferase (HAT) activity of the CREB binding protein (CBP) in vitro. We now show that deletion of the CBP bromo- and C/H3 domains eliminates stimulation of nucleosomal HAT activity in vitro and transcriptional coactivation by Zta in transfected cells. In contrast, acetylation of free histones was not affected by the addition of Zta or by deletions in the bromo or C/H3 domain of CBP. Zta stimulated acetylation of oligonucleosomes assembled on supercoiled DNA and dinucleosomes assembled on linear DNA, but Zta-stimulated acetylation was significantly reduced for mononucleosomes. Western blotting and amino-terminal protein sequencing indicated that all lysine residues in the H3 and H4 amino-terminal tails were acetylated by CBP and enhanced by the addition of Zta. Histone acetylation was also dependent upon the Zta basic DNA binding domain, which could not be substituted with the homologous basic region of c-Fos, indicating specificity in the bZIP domain nucleosome binding function. Finally, we show that Zta and CBP colocalize to viral immediate-early promoters in vivo and that overexpression of Zta leads to a robust increase in H3 and H4 acetylation at various regions of the EBV genome in vivo. Furthermore, deletion of the CBP bromodomain reduced stable CBP-Zta complex formation and histone acetylation at Zta-responsive viral promoters in vivo. These results suggest that activator- and bromodomain-dependent targeting to oligonucleosomal chromatin is required for stable promoter-bound complex formation and transcription activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Herpesvirus Humano 4/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Transativadores/química , Transativadores/metabolismo , Proteínas Virais/metabolismo , Acetilação , Acetiltransferases/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação/genética , Proteína de Ligação a CREB , Linhagem Celular , DNA Viral/genética , DNA Viral/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidade , Histona Acetiltransferases , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/genética , Nucleossomos/metabolismo , Estrutura Terciária de Proteína , Proteínas de Saccharomyces cerevisiae/metabolismo , Deleção de Sequência , Transativadores/genética , Ativação Transcricional , Transfecção , Proteínas Virais/química , Proteínas Virais/genética
17.
Mol Cell ; 9(2): 279-89, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11864602

RESUMO

The transcription factors HNF3 (FoxA) and GATA-4 are the earliest known to bind the albumin gene enhancer in liver precursor cells in embryos. To understand how they access sites in silent chromatin, we assembled nucleosome arrays containing albumin enhancer sequences and compacted them with linker histone. HNF3 and GATA-4, but not NF-1, C/EBP, and GAL4-AH, bound their sites in compacted chromatin and opened the local nucleosomal domain in the absence of ATP-dependent enzymes. The ability of HNF3 to open chromatin is mediated by a high affinity DNA binding site and by the C-terminal domain of the protein, which binds histones H3 and H4. Thus, factors that potentiate transcription in development are inherently capable of initiating chromatin opening events.


Assuntos
Cromatina/ultraestrutura , Proteínas de Ligação a DNA/fisiologia , Proteínas Fetais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histonas/fisiologia , Proteínas Nucleares/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Motivos de Aminoácidos , Animais , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Cromatina/genética , Cromatina/metabolismo , Proteínas de Ligação a DNA/química , Endoderma/metabolismo , Elementos Facilitadores Genéticos/fisiologia , Proteínas Fetais/biossíntese , Proteínas Fetais/genética , Fator de Transcrição GATA4 , Inativação Gênica , Fator 3-alfa Nuclear de Hepatócito , Fígado/embriologia , Fígado/metabolismo , Substâncias Macromoleculares , Camundongos , Modelos Genéticos , Neurofibromina 1/metabolismo , Proteínas Nucleares/química , Nucleossomos/fisiologia , Regiões Promotoras Genéticas/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Albumina Sérica/biossíntese , Albumina Sérica/genética , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...