Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Cell Biol ; 97(2): 118-129, 2019 04.
Article in English | MEDLINE | ID: mdl-30142277

ABSTRACT

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.


Subject(s)
Chromatin/metabolism , DNA/metabolism , Forkhead Box Protein O1/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Insulin/metabolism , Chromatin/genetics , DNA/genetics , Forkhead Box Protein O1/genetics , Hep G2 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Insulin/genetics , Insulin-Like Growth Factor Binding Protein 1/biosynthesis , Insulin-Like Growth Factor Binding Protein 1/genetics , Phosphorylation/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Response Elements
2.
J Biol Chem ; 291(16): 8848-61, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26929406

ABSTRACT

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.


Subject(s)
Forkhead Transcription Factors/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Insulin/metabolism , Promoter Regions, Genetic/physiology , Transcriptional Activation/physiology , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Hep G2 Cells , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Insulin/genetics , Insulin-Like Growth Factor Binding Protein 1/biosynthesis , Insulin-Like Growth Factor Binding Protein 1/genetics , Mice , Phosphorylation/physiology , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
3.
J Biol Chem ; 285(1): 464-72, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19897491

ABSTRACT

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.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Acetylation , Animals , DNA/metabolism , Enhancer Elements, Genetic/genetics , Hep G2 Cells , Hepatocytes/metabolism , Humans , Mice , Nucleosomes/metabolism , Protein Binding , p300-CBP Transcription Factors/metabolism
5.
J Biol Chem ; 282(49): 35583-93, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17923482

ABSTRACT

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.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA/metabolism , Forkhead Transcription Factors/metabolism , Histones/metabolism , Nucleosomes/metabolism , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cell Survival/physiology , Cell-Free System/metabolism , DNA/genetics , DNA Repair/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Histones/genetics , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Mice , Nucleosomes/genetics , Oxidative Stress/physiology , Promoter Regions, Genetic/physiology , Protein Binding/physiology , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Xenopus laevis
6.
J Virol ; 78(11): 5848-55, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15140982

ABSTRACT

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.


Subject(s)
Chromatin/virology , Integrases/physiology , Retroviridae/genetics , Virus Integration , Avian Sarcoma Viruses/enzymology , Binding Sites , Chromatin/chemistry , DNA-Binding Proteins/physiology , GATA4 Transcription Factor , HIV Integrase/physiology , Hepatocyte Nuclear Factor 3-alpha , Nuclear Proteins/physiology , Nucleosomes/metabolism , Transcription Factors/physiology
8.
Mol Cell Biol ; 23(8): 2633-44, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12665567

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Trans-Activators/chemistry , Trans-Activators/metabolism , Viral Proteins/metabolism , Acetylation , Acetyltransferases/metabolism , Amino Acid Sequence , Base Sequence , Binding Sites/genetics , CREB-Binding Protein , Cell Line , DNA, Viral/genetics , DNA, Viral/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Histone Acetyltransferases , Humans , Molecular Sequence Data , Nuclear Proteins/genetics , Nucleosomes/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/metabolism , Sequence Deletion , Trans-Activators/genetics , Transcriptional Activation , Transfection , Viral Proteins/chemistry , Viral Proteins/genetics
9.
Mol Cell ; 9(2): 279-89, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11864602

ABSTRACT

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.


Subject(s)
Chromatin/ultrastructure , DNA-Binding Proteins/physiology , Fetal Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Histones/physiology , Nuclear Proteins/physiology , Transcription Factors/physiology , Transcription, Genetic/physiology , Amino Acid Motifs , Animals , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/chemistry , Endoderm/metabolism , Enhancer Elements, Genetic/physiology , Fetal Proteins/biosynthesis , Fetal Proteins/genetics , GATA4 Transcription Factor , Gene Silencing , Hepatocyte Nuclear Factor 3-alpha , Liver/embryology , Liver/metabolism , Macromolecular Substances , Mice , Models, Genetic , Neurofibromin 1/metabolism , Nuclear Proteins/chemistry , Nucleosomes/physiology , Promoter Regions, Genetic/physiology , Recombinant Fusion Proteins/physiology , Serum Albumin/biosynthesis , Serum Albumin/genetics , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...