Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 18(4): 756-771, 2023 04 21.
Article in English | MEDLINE | ID: mdl-36988910

ABSTRACT

Repetitive physical exercise induces physiological adaptations in skeletal muscle that improves exercise performance and is effective for the prevention and treatment of several diseases. Genetic evidence indicates that the orphan nuclear receptors estrogen receptor-related receptors (ERRs) play an important role in skeletal muscle exercise capacity. Three ERR subtypes exist (ERRα, ß, and γ), and although ERRß/γ agonists have been designed, there have been significant difficulties in designing compounds with ERRα agonist activity. Additionally, there are limited synthetic agonists that can be used to target ERRs in vivo. Here, we report the identification of a synthetic ERR pan agonist, SLU-PP-332, that targets all three ERRs but has the highest potency for ERRα. Additionally, SLU-PP-332 has sufficient pharmacokinetic properties to be used as an in vivo chemical tool. SLU-PP-332 increases mitochondrial function and cellular respiration in a skeletal muscle cell line. When administered to mice, SLU-PP-332 increased the type IIa oxidative skeletal muscle fibers and enhanced exercise endurance. We also observed that SLU-PP-332 induced an ERRα-specific acute aerobic exercise genetic program, and the ERRα activation was critical for enhancing exercise endurance in mice. These data indicate the feasibility of targeting ERRα for the development of compounds that act as exercise mimetics that may be effective in the treatment of numerous metabolic disorders and to improve muscle function in the aging.


Subject(s)
Estrogens , Exercise Tolerance , Receptors, Estrogen , Animals , Mice , Exercise Tolerance/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Receptors, Estrogen/drug effects , Receptors, Estrogen/metabolism , Estrogens/chemistry , Estrogens/pharmacology , ERRalpha Estrogen-Related Receptor
2.
Int J Cell Biol ; 2011: 715642, 2011.
Article in English | MEDLINE | ID: mdl-21785598

ABSTRACT

The p400 and SRCAP (Snf2-related CBP activator protein) complexes remodel chromatin by catalyzing deposition of histone H2A.Z into nucleosomes. This remodeling activity has been proposed as a basis for regulation of transcription by these complexes. Transcript levels of p21 or Sp1 mRNAs after knockdown of p400 or SRCAP reveals that each regulates transcription of these promoters differently. In this study, we asked whether deposition of H2A.Z within specific nucleosomes by p400 or SRCAP dictates transcriptional activity. Our data indicates that nucleosome density at specific p21 or Sp1 promoter positions is not altered by the loss of either remodeling complex. However, knockdown of SRCAP or p400 reduces deposition of H2A.Z∼50% into all p21 and Sp1 promoter nucleosomes. Thus, H2A.Z deposition is not targeted to specific nucleosomes. These results indicate that the deposition of H2A.Z by the p400 or SRCAP complexes is not sufficient to determine how each regulates transcription. This conclusion is further supported by studies that demonstrate a SRCAP(ΔATP ) mutant unable to deposit H2A.Z has similar transcriptional activity as wild-type SRCAP.

3.
Cell Cycle ; 9(22): 4533-41, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-21088490

ABSTRACT

Cyclin E-associated CDK2 activity is required for the initiation of DNA synthesis in human cells. CDK2 activity is tightly regulated; CDK2 must be in the nucleus, bound to a cyclin, phosphorylated on T160, and dephosphorylated on T14/Y15 for complete kinase activation. Nuclear localization exposes CDK2 to activating enzymes (CAK, Cdc25A) in stimulated cells. Previous studies from our lab indicate CDK2 nuclear localization and cyclin E co-expression are insufficient to cause CDK2 activation or T160 phosphorylation in stimulated IIC9 cells; these activities still require serum stimulation and ERK kinase activity. Recent studies have implicated a role for origin of replication (ORC) licensing proteins in the activation of G1/S Cdks. In this study, we show that CDK2 associates with chromatin and Cdc6 in an ERK-dependent manner following stimulation of IIC9 CHEF cells. We show that nuclear-localized CDK2 (CDK2-NLS) ectopically expressed with cyclin E requires mitogenic stimulation and ERK activation for chromatin association, in addition to previously shown kinase activation and T160 phosphorylation in IIC9 cells. Additionally, we show that expression of Cdc6 in stimulated IIC9 cells treated with ERK inhibitor rescues CDK2-NLS chromatin association, kinase activation, and T160 phosphorylation. From the above data, we deduce ERK-dependent CDK2 activation is due in part to ERK-dependent Cdc6 expression. To examine the role of Cdc6 directly in stimulated primary human fibroblasts, we used RNA interference to attenuate the expression of Cdc6. We show that Cdc6 expression is required for CDK2 chromatin association and kinase activation in stimulated primary human fibroblasts. Additionally, we show that Cdc6 expression is required for the initiation of DNA synthesis and S phase entry in stimulated primary human fibroblasts. Ultimately, this data implicates Cdc6 expression as an important mitogen-induced mechanism in the activation of CDK2/cyclin E, the initiation of DNA synthesis, and the regulation of G1-S phase progression.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Nuclear Proteins/metabolism , Animals , Butadienes/pharmacology , Cell Cycle Proteins/genetics , Cell Line , Chromatin/metabolism , Cricetinae , Cricetulus , Cyclin E/analysis , Cyclin E/genetics , Cyclin-Dependent Kinase 2/analysis , Cyclin-Dependent Kinase 2/genetics , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , G1 Phase , Humans , Nitriles/pharmacology , Nuclear Proteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering/metabolism , S Phase
4.
J Biol Chem ; 282(36): 26132-9, 2007 Sep 07.
Article in English | MEDLINE | ID: mdl-17617668

ABSTRACT

Studies in Saccharomyces cerevisiae indicate the histone variant H2A.Z is deposited at promoters by the chromatin remodeling protein Swr1 and plays a critical role in the regulation of transcription. In higher eukaryotes, however, little is known about the distribution, method of deposition, and function of H2A.Z at promoters. Using biochemical studies, we demonstrated previously that SRCAP (SNF-2-related CREB-binding protein activator protein), the human ortholog of Swr1, could catalyze deposition of H2A.Z into nucleosomes. To address whether SRCAP directs H2A.Z deposition in vivo, promoters targeted by SRCAP were identified by a chromatin immunoprecipitation (ChIP)-on-chip assay. ChIP assays on a subset of these promoters confirmed the presence of SRCAP on inactive and active promoters. The highest levels of SRCAP were observed on the active SP-1, G3BP, and FAD synthetase promoters. Detailed analyses of these promoters indicate sites of SRCAP binding overlap or occur adjacent to the sites of H2A.Z deposition. Knockdown of SRCAP levels using siRNA resulted in loss of SRCAP at these promoters, decreased deposition of H2A.Z and acetylated H2A.Z, and a decrease in levels of SP-1, G3BP, and FAD synthetase mRNA. Thus, these studies provide the first evidence that SRCAP is recruited to promoters and is critical for the deposition of H2A.Z.


Subject(s)
Adenosine Triphosphatases/metabolism , Chromatin Assembly and Disassembly/physiology , Histones/metabolism , Nucleosomes/metabolism , Response Elements/physiology , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Cell Line, Tumor , Chromatin Assembly and Disassembly/drug effects , Chromatin Immunoprecipitation , DNA Helicases , Histones/genetics , Humans , Microarray Analysis , Nucleosomes/genetics , Nucleotidyltransferases/biosynthesis , Nucleotidyltransferases/genetics , Poly-ADP-Ribose Binding Proteins , RNA Helicases , RNA Recognition Motif Proteins , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sp1 Transcription Factor/biosynthesis , Sp1 Transcription Factor/genetics
5.
Biochemistry ; 45(17): 5671-7, 2006 May 02.
Article in English | MEDLINE | ID: mdl-16634648

ABSTRACT

The Snf-2-related CREB-binding protein activator protein (SRCAP) serves as a coactivator for a number of transcription factors known to interact with CBP. Swr1, the closest Saccharomyces cerevisiae ortholog of SRCAP, is a component of the chromatin remodeling complex SWR-C, which catalyzes exchange of the histone variant H2A.Z into nucleosomes. In this report, we use a combination of conventional chromatography and anti-SRCAP immunoaffinity chromatography to purify a native human SRCAP complex with a polypeptide composition similar to that of SWR-C, and we show for the first time that this SRCAP-containing complex supports ATP-dependent exchange of histone dimers containing H2B and H2A.Z into mononucleosomes reconstituted with recombinant H2A, H2B, H3, and H4. Our findings, together with previous evidence implicating H2A.Z in transcriptional regulation, suggest that SRCAP's coactivator function may depend on its ability to promote incorporation of H2A.Z into chromatin.


Subject(s)
Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/metabolism , Chromatin/metabolism , Histones/metabolism , Multiprotein Complexes/isolation & purification , Multiprotein Complexes/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Chromatin/drug effects , Chromatography, Ion Exchange/methods , Proteomics
6.
Mol Cell Biol ; 25(15): 6559-69, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16024792

ABSTRACT

The putative ATPase chromatin-remodeling machine SRCAP was identified in a yeast two-hybrid protein screen by interaction with the histone acetylase CBP. SRCAP is implicated in the transcriptional coactivation of cyclic AMP- and steroid-dependent promoters, but no natural chromosomal targets for SRCAP regulation have been identified. DOM is the unique SRCAP homolog in Drosophila melanogaster. The goal of this study was to test whether SRCAP is a functional homolog of DOM and to identify potential activities and targets of SRCAP in vivo. We show that human SRCAP complements recessive domino mutant phenotypes. This rescue depends on an intact ATPase homology domain. SRCAP colocalizes extensively with DOM on Drosophila polytene chromosomes and is recruited to sites of active transcription, such as steroid-regulated loci, but not to activated heat shock loci. We show that SRCAP recruits Drosophila CBP to ectopic chromosomal sites, providing the first evidence to suggest that SRCAP and CBP interact directly or indirectly on chromosomes. We show that DOM is a Notch pathway activator in Drosophila and that wild-type SRCAP-but not an ATPase domain mutant-can substitute for DOM in Notch-dependent wing development. We show that SRCAP potentiates Notch-dependent gene activation in HeLa cells. Taken together, these data implicate SRCAP and DOM in developmental gene activation.


Subject(s)
Adenosine Triphosphatases/physiology , Drosophila Proteins/physiology , Membrane Proteins/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , HeLa Cells , Heat-Shock Proteins/genetics , Humans , Infertility, Female/genetics , Infertility, Female/metabolism , Mutation , Receptors, Notch , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
7.
Mol Cell Endocrinol ; 214(1-2): 71-9, 2004 Feb 12.
Article in English | MEDLINE | ID: mdl-15062546

ABSTRACT

To directly activate specific gene expression, the estrogen receptor (ER) must bind to estrogen receptor response elements (EREs) in the context of nucleosomes. In order to investigate the interaction of the ER with mononucleosomes, we developed a mononucleosome gel shift assay. A 164 bp high specific activity [(32)P]probe DNA (32 bp consensus ERE with flanking regions separated by 23 nucleotides from an artificial nucleosome positioning sequence) was prepared. Nuclear extracts from MCF-7 cells or recombinant human ERalpha were incubated with the labeled ERE +/- excess ERE. A retarded band was seen which was completely obliterated with excess ERE, confirming the specificity of binding. This probe was then used to make reconstituted mononucleosomes by sequential dilution of a high salt histone preparation. The nucleosomes were purified by sucrose density gradients and footprinting analysis was performed to demonstrate that the mononucleosomes were rotationally phased as seen by a periodic digestion pattern (10 bp) of the nucleosomes versus ERE. Nucleosomes were incubated with nuclear extracts containing ER or recombinant ERalpha. Dose dependence in the shift of the mononucleosomes with increasing concentrations of ER was observed. Specificity was demonstrated in experiments with excess ERE and anti-ER antibody. Footprinting analysis was also performed. We also determined that addition of high mobility group protein-2 (HMGB-2, a protein closely related to HMGB-1) with the ER increased the interaction of ER with mononucleosomes. These studies will allow us to address the interactions of ER with core histones containing a multiplicity of variants and modifications in nucleosomal structure.


Subject(s)
Nucleosomes/metabolism , Protein Footprinting/methods , Receptors, Estrogen/metabolism , Cell Line, Tumor , DNA Probes , Electrophoretic Mobility Shift Assay/methods , Female , HMGB2 Protein/pharmacology , Humans , Protein Binding/drug effects , Response Elements
8.
Mol Endocrinol ; 17(12): 2519-28, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14500758

ABSTRACT

SRCAP (SNF2-related CBP activator protein) is a 350-kDa protein that shares homology with the SNF2 family of proteins whose members function in various aspects of transcriptional regulation. In various cell types, SRCAP is found in distinct multiprotein complexes that include proteins found in SWI/SNF chromatin remodeling complexes. SRCAP was identified by its ability to bind to CBP and was found to potentiate the ability of CBP to activate transcription. Studies in our laboratory have demonstrated that SRCAP functions as a coactivator for CREB-mediated transcription of a number of promoters, including that of the phosphoenolpyruvate carboxykinase gene. Our current studies demonstrate that SRCAP enhances phosphoenolpyruvate carboxykinase promoter transcription induced by glucocorticoids. SRCAP also enhances glucocorticoid receptor-mediated transcription of a simple promoter containing only two glucocorticoid response elements, indicating that SRCAP functions as a glucocorticoid receptor coactivator. In similar studies, SRCAP was also found to serve as a coactivator for the androgen receptor. SRCAP exhibits synergistic activation with nuclear receptor coactivators and functionally interacts in vivo with glucocorticoid receptor-interacting protein-1 and coactivator-associated arginine methyltransferase-1. We propose that SRCAP, by virtue of its ability to interact with CBP, functions as a coactivator to regulate transcription initiated by several signaling pathways.


Subject(s)
Adenosine Triphosphatases/physiology , Carrier Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Receptors, AMPA/metabolism , Transcription, Genetic/genetics , Animals , COS Cells , Chlorocebus aethiops , Cloning, Molecular , HeLa Cells , Humans , Plasmids/genetics , Recombinant Proteins/metabolism , Transfection
9.
Virology ; 313(2): 615-21, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12954226

ABSTRACT

The SNF2-related CBP activator protein (SrCap) is a potent activator of transcription mediated by CBP and CREB. We have previously demonstrated that the Adenovirus 2 DNA Binding Protein (DBP) binds to SrCap and inhibits the transcription mediated by the carboxyl-terminal region of SrCap (amino acids 1275-2971). We report here that DBP inhibits the ability of full-length SrCap (1-2971) to activate transcription mediated by Gal-CREB and Gal-CBP. In addition, DBP also inhibits the ability of SrCap to enhance Protein Kinase A (PKA) activated transcription of the enkaphalin promoter. DBP was found to dramatically inhibit transcription of a mammalian two-hybrid system that was dependent on the interaction of SrCap and CBP binding domains. We also found that DBP has no effect on transcription mediated by a transcriptional activator that is not related to SrCap, indicating that our reported transcriptional inhibition is specific for SrCap and not due to nonspecific effects of DBP's DNA binding activity on the CAT reporter plasmid. Taken together, these results suggest a model in which DBP inhibits cellular transcription mediated by the interaction between SrCap and CBP.


Subject(s)
Adenosine Triphosphatases/genetics , Adenoviridae/genetics , Cyclic AMP Response Element-Binding Protein/genetics , DNA-Binding Proteins/genetics , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Viral Proteins/genetics , Acetyltransferases/genetics , Adenosine Triphosphatases/metabolism , Animals , CHO Cells , CREB-Binding Protein , Cricetinae , Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation , Nuclear Proteins/metabolism , Protein Binding , Trans-Activators/metabolism , Transcription, Genetic
10.
Parasitol Res ; 90(1): 1-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12743798

ABSTRACT

We have identified and mapped a gene in Toxoplasma gondii that encodes a homologue of SRCAP (Snf2-related CBP activator protein), a member of the SNF/SWI family of chromatin remodeling factors. The genomic locus (TgSRCAP) is present as a single copy and contains 16 introns. The predicted cDNA contains an open reading frame of 8,775 bp and encodes a protein of 2,924 amino acids. We have identified additional SRCAP-like sequences in Apicomplexa for comparison by screening genomic databases. An analysis of SRCAP homologues between species reveals signature features that may be indicative of SRCAP members. Expression of mRNA encoding TgSRCAP is upregulated when tachyzoite (invasive form) parasites are induced to differentiate into bradyzoites (encysted form) in vitro. Recombinant TgSRCAP protein is functionally equivalent to the human homologue, being capable of increasing transcription mediated by CREB.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Chromatin/genetics , Genes, Protozoan , Toxoplasma/genetics , Adenosine Triphosphatases/analysis , Amino Acid Sequence , Animals , CREB-Binding Protein , Cloning, Molecular , Gene Expression Regulation , HeLa Cells , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , RNA, Messenger/genetics , Sequence Homology, Amino Acid , Toxoplasma/isolation & purification , Trans-Activators/metabolism , Transcription, Genetic
11.
J Biol Chem ; 277(4): 2517-24, 2002 Jan 25.
Article in English | MEDLINE | ID: mdl-11705990

ABSTRACT

The CBP and p300 co-activators play a key role in many aspects of gene regulation being recruited to the DNA via transcription factors that are targets for specific signaling pathways. It has previously been demonstrated that in neuronal cells the ability of CBP and p300 to activate transcription can be directly stimulated by nerve growth factor or calcium-activated signaling pathways. Here we demonstrate that, in cardiac cells, the activity of CBP and p300 is stimulated by phenylephrine (PE) treatment and that they are required for the activation of atrial naturetic factor (ANF) gene expression by PE. Activation of CBP/p300 by PE involves the p42/p44 MAPK pathway and targets primarily the N terminus of p300 and the C terminus of CBP, which are not homologous to one another. To our knowledge, this is the first report of a specific stimulus modulating the activity of CBP and p300 in cardiac cells and it suggests that these factors play an important role in the hypertrophic effect of PE.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/physiology , Phenylephrine/metabolism , Trans-Activators/physiology , Transcription, Genetic , Animals , Animals, Newborn , Blotting, Western , CREB-Binding Protein , Cells, Cultured , DNA/metabolism , E1A-Associated p300 Protein , Genes, Dominant , Luciferases/metabolism , Mitogen-Activated Protein Kinase 3 , Mutation , Myocardium/cytology , Myocardium/metabolism , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Promoter Regions, Genetic , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...