Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 23(3): 469-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26383972

ABSTRACT

The tumor suppressor protein promyelocytic leukemia (PML) is a key regulator of inflammatory responses and tumorigenesis and functions through the assembly of subnuclear structures known as PML nuclear bodies (NBs). The inflammation-related cytokine tumor necrosis factor-α (TNFα) is known to induce PML protein accumulation and PML NB formation that mediate TNFα-induced cell death in cancer cells and inhibition of migration and capillary tube formation in endothelial cells (ECs). In this study, we uncover a novel mechanism of PML gene regulation in which the p38 MAPK and its downstream kinase MAP kinase-activated protein kinase 1 (MNK1) mediate TNFα-induced PML protein accumulation and PML NB formation. The mechanism includes the presence of an internal ribosome entry site (IRES) found within the well-conserved 100 nucleotides upstream of the PML initiation codon. The activity of the PML IRES is induced by TNFα in a manner that involves MNK1 activation. It is proposed that the p38-MNK1-PML network regulates TNFα-induced apoptosis in breast cancer cells and TNFα-mediated inhibition of migration and capillary tube formation in ECs.


Subject(s)
Apoptosis , Human Umbilical Vein Endothelial Cells/physiology , Nuclear Proteins/genetics , Protein Biosynthesis , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/physiology , Tumor Suppressor Proteins/genetics , 5' Untranslated Regions , Breast Neoplasms , Cell Movement , Cell Proliferation , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression Regulation, Neoplastic , HL-60 Cells , Humans , Internal Ribosome Entry Sites , Intracellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Matrix Metalloproteinase 10/genetics , Matrix Metalloproteinase 10/metabolism , Neovascularization, Physiologic , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Cell Death Dis ; 5: e1340, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25032863

ABSTRACT

The promyelocytic leukemia protein (PML) is a tumor suppressor that is expressed at a low level in various cancers. Although post-translational modifications including SUMOylation, phosphorylation, and ubiquitination have been found to regulate the stability or activity of PML, little is known about the role of its acetylation in the control of cell survival. Here we demonstrate that acetylation of lysine 487 (K487) and SUMO1 conjugation of K490 at PML protein are mutually exclusive. We found that hydrogen peroxide (H2O2) promotes PML deacetylation and identified SIRT1 and SIRT5 as PML deacetylases. Both SIRT1 and SIRT5 are required for H2O2-mediated deacetylation of PML and accumulation of nuclear PML protein in HeLa cells. Knockdown of SIRT1 reduces the number of H2O2-induced PML-nuclear bodies (NBs) and increases the survival of HeLa cells. Ectopic expression of wild-type PML but not the K487R mutant rescues H2O2-induced cell death in SIRT1 knockdown cells. Furthermore, ectopic expression of wild-type SIRT5 but not a catalytic defective mutant can also restore H2O2-induced cell death in SIRT1 knockdown cells. Taken together, our findings reveal a novel regulatory mechanism in which SIRT1/SIRT5-mediated PML deacetylation plays a role in the regulation of cancer cell survival.


Subject(s)
Hydrogen Peroxide/pharmacology , Leukemia, Promyelocytic, Acute/metabolism , Nuclear Proteins/metabolism , Sirtuin 1/metabolism , Sirtuins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Acetylation , Amino Acid Motifs , HeLa Cells , Humans , Leukemia, Promyelocytic, Acute/enzymology , Leukemia, Promyelocytic, Acute/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Sirtuin 1/genetics , Sirtuins/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
4.
Oncogene ; 32(33): 3819-28, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-22945642

ABSTRACT

The promyelocytic leukemia (PML) protein is a tumor suppressor originally identified in acute promyelocytic leukemia and implicated in tumorigenesis in multiple forms of cancer. Here, we demonstrate that the PML protein undergoes ubiquitination-mediated degradation facilitated by an E3 ligase UHRF1 (ubiquitin-like with PHD and RING finger domains 1), which is commonly upregulated in various human malignancies. Furthermore, UHRF1 negatively regulates PML protein accumulation in primary human umbilical vein endothelial cells (HUVECs), HEK 293 cells and cancer cells. Knockdown of UHRF1 upregulates whereas ectopic overexpression of UHRF1 downregulates protein abundance of endogenous or exogenous PML, doing so through its binding to the N-terminus of PML. Overexpression of wild-type UHRF1 shortens PML protein half-life and promotes PML polyubiquitination, whereas deletion of the RING domain or coexpression of the dominant-negative E2 ubiquitin-conjugating enzyme, E2D2, attenuates this modification to PML. Finally, knockdown of UHRF1 prolongs PML half-life and increases PML protein accumulation, yet inhibits cell migration and in vitro capillary tube formation, whereas co-knockdown of PML compromises this inhibitory effect. These findings suggest that UHRF1 promotes the turnover of PML protein, and thus targeting UHRF1 to restore PML-mediated tumor suppression represents a promising, novel, anticancer strategy.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Epigenesis, Genetic , Neoplasms/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitination , Blotting, Western , Endothelial Cells/metabolism , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunoprecipitation , Microscopy, Fluorescence , Promyelocytic Leukemia Protein , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Ubiquitin-Protein Ligases
5.
Hum Genet ; 128(5): 501-13, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20725740

ABSTRACT

To dissect the genetic architecture of sexual dimorphism in obesity-related traits, we evaluated the sex-genotype interaction, sex-specific heritability and genome-wide linkages for seven measurements related to obesity. A total of 1,365 non-diabetic Chinese subjects from the family study of the Stanford Asia-Pacific Program of Hypertension and Insulin Resistance were used to search for quantitative trait loci (QTLs) responsible for the obesity-related traits. Pleiotropy and co-incidence effects from the QTLs were also examined using the bivariate linkage approach. We found that sex-specific differences in heritability and the genotype-sex interaction effects were substantially significant for most of these traits. Several QTLs with strong linkage evidence were identified after incorporating genotype by sex (G × S) interactions into the linkage mapping, including one QTL for hip circumference [maximum LOD score (MLS) = 4.22, empirical p = 0.000033] and two QTLs: for BMI on chromosome 12q with MLS 3.37 (empirical p = 0.0043) and 3.10 (empirical p = 0.0054). Sex-specific analyses demonstrated that these linkage signals all resulted from females rather than males. Most of these QTLs for obesity-related traits replicated the findings in other ethnic groups. Bivariate linkage analyses showed several obesity traits were influenced by a common set of QTLs. All regions with linkage signals were observed in one gender, but not in the whole sample, suggesting the genetic architecture of obesity-related traits does differ by gender. These findings are useful for further identification of the liability genes for these phenotypes through candidate genes or genome-wide association analysis.


Subject(s)
Asian People/genetics , Obesity/genetics , Sex Characteristics , Adult , Body Mass Index , Chromosome Mapping , Female , Genome-Wide Association Study , Genotype , Hawaii/epidemiology , Humans , Lod Score , Male , Middle Aged , Obesity/epidemiology , Odds Ratio , Phenotype , San Francisco/epidemiology , Sex Factors , Taiwan/epidemiology
6.
Diabetologia ; 50(8): 1631-40, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17579830

ABSTRACT

AIMS/HYPOTHESIS: Hypertension, obesity, impaired glucose tolerance and dyslipidaemia are metabolic abnormalities that often cluster together more often than expected by chance alone. Since these metabolic variables are highly heritable and are at least partially genetically determined, the clustering of defects in these traits implies that pleiotropic effects, where a common set of genes influences more than one trait simultaneously, are likely. METHODS: We conducted bivariate linkage analyses for highly correlated traits, aiming to dissect the genetic architecture affecting these traits, in 411 Chinese families participating in the Stanford Asia-Pacific Program of Hypertension and Insulin Resistance Study. RESULTS: We confirmed the pleiotropic effects of the locus at 37 cM on chromosome 20 on the following pairs: (1) fasting insulin and insulin AUC (empirical p = 0.0006); (2) fasting insulin and homeostasis model assessment of beta cell function (HOMA-beta) (empirical p = 0.0051); and (3) HOMA of insulin resistance (IR) and HOMA-beta (empirical p = 0.0044). In addition, the peak logarithm of the odds (LOD) scores of linkage between a chromosomal locus and a trait for the pair fasting insulin and HOMA-IR rose to 5.10 (equivalent LOD score in univariate analysis, LOD([1]) = 4.01, empirical p = 8.0 x 10(-5)) from 3.67 and 3.42 respectively for these two traits in univariate analysis. Additional significant linkage evidence, not shown in single-trait analysis, was identified at 45 cM on chromosome 16 for the pair 1 h insulin and the AUC for insulin, with a LOD score of 4.29 (or LOD([1]) = 3.27, empirical p = 2.0 x 10(-4)). This new locus is also likely to harbour the common genes regulating these two traits (p = 1.73 x 10(-6)). CONCLUSIONS/INTERPRETATION: These data help provide a better understanding of the genomic structure underlying the metabolic syndrome.


Subject(s)
Asian People/genetics , Genome, Human , Hypertension/genetics , Insulin Resistance/genetics , Adult , Analysis of Variance , Blood Glucose/metabolism , Body Mass Index , Cholesterol, HDL/genetics , Chromosomes, Human, Pair 20/genetics , Family Health , Fasting , Female , Genetic Linkage/genetics , Genotype , Humans , Hypertension/blood , Lod Score , Male , Metabolic Syndrome/genetics , Middle Aged , Phenotype , Quantitative Trait Loci/genetics
7.
Phytomedicine ; 13(5): 366-70, 2006 May.
Article in English | MEDLINE | ID: mdl-16635745

ABSTRACT

The water extract of Anoectochilus formosanus Hayata showed a potent tumor inhibitory activity in BALB/c mice after subcutaneous transplantation of CT-26 murine colon cancer cells. The tumor-inhibition ratios of mice pre-administered with A. formosanus for 2 days before tumor transplantation, and treated further for 12 consecutive days, were 55.4% and 58.9% at the oral dose of 50 and 10 mg/mouse per day, respectively. Even for the tumor-bearing mice, after oral administration of the water extract of A. formosanus for 12 consecutive days, the tumor inhibition ratios were still 23.8% and 40.5% at doses of 50 and 10 mg/mouse, respectively. Because the low-concentration water extract of A. formosanus does not show direct cytotoxicity in CT-26 tumor cells, we observed further that oral administration of the water extract of A. formosanus may activate murine immune responses, such as stimulating the proliferation of lymphoid tissues and activating the phagocytosis of peritoneal macrophages against Staphylococcus aureus. This study suggests that the antitumor activity of A. formosanus may be associated with its potent immunostimulating effect. It is worth further analyzing the immunomodulating component purified from A. formosanus, and evaluating its potential value for the treatment of human cancers.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Lymphocytes/drug effects , Macrophages, Peritoneal/drug effects , Orchidaceae/chemistry , Animals , Cell Line, Tumor/drug effects , Female , Lymphocyte Activation/drug effects , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Phagocytosis/drug effects , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plants, Medicinal/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Toxicity Tests/methods
8.
J Biol Chem ; 276(50): 47496-507, 2001 Dec 14.
Article in English | MEDLINE | ID: mdl-11585834

ABSTRACT

Here we show that HDAC7, a member of the class II histone deacetylases, specifically targets several members of myocyte enhancer factors, MEF2A, -2C, and -2D, and inhibits their transcriptional activity. Furthermore, we demonstrate that DNA-bound MEF2C is capable of recruiting HDAC7, demonstrating that the HDAC7-dependent repression of transcription is not due to the inhibition of the MEF2 DNA binding activity. The data also suggest that the promoter bound MEF2 is potentially capable of remodeling adjacent nucleosomes via the recruitment of HDAC7. We have also observed a nucleocytoplasmic shuttling of HDAC7 and dissected the mechanism involved. In NIH3T3 cells, HDAC7 was primarily localized in the cytoplasm, essentially due to an active CRM1-dependent export of the protein from the nucleus. Interestingly, in HeLa cells, HDAC7 was predominantly nuclear. In these cells we could restore the cytoplasmic localization of HDAC7 by expressing CaMK I. This CaMK I-induced nuclear export of HDAC7 was abolished when three critical serines, Ser-178, Ser-344, and Ser-479, of HDAC7 were mutated. We show that these serines are involved in the direct interaction of HDAC7 with 14-3-3. Mutations of these serine residues weakened the association with 14-3-3 and dramatically enhanced the repression activity of HDAC7 in NIH3T3 cells, but not in HeLa cells. Data presented in this work clearly show that the signal dependent subcellular localization of HDAC7 is essential in controlling its activities. The data also show that the cellular concentration of factors such as 14-3-3, CaMK I, and other yet unknown molecules may determine the subcellular localization of an individual HDAC member in a cell type and HDAC-specific manner.


Subject(s)
Active Transport, Cell Nucleus , Histone Deacetylases/metabolism , Saccharomyces cerevisiae Proteins , 14-3-3 Proteins , 3T3 Cells , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins , Down-Regulation , Electrophoresis, Polyacrylamide Gel , Fungal Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins , Haplorhini , HeLa Cells , Humans , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Mutation , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Serine/chemistry , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Tyrosine 3-Monooxygenase/metabolism
9.
Genes Dev ; 15(9): 1140-51, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11331609

ABSTRACT

A yeast two-hybrid screen using the conserved carboxyl terminus of the nuclear receptor corepressor SMRT as a bait led to the isolation of a novel human gene termed SHARP (SMRT/HDAC1 Associated Repressor Protein). SHARP is a potent transcriptional repressor whose repression domain (RD) interacts directly with SMRT and at least five members of the NuRD complex including HDAC1 and HDAC2. In addition, SHARP binds to the steroid receptor RNA coactivator SRA via an intrinsic RNA binding domain and suppresses SRA-potentiated steroid receptor transcription activity. Accordingly, SHARP has the capacity to modulate both liganded and nonliganded nuclear receptors. Surprisingly, the expression of SHARP is itself steroid inducible, suggesting a simple feedback mechanism for attenuation of the hormonal response.


Subject(s)
DNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Conserved Sequence , DNA-Binding Proteins/genetics , Estrogens/metabolism , Histone Deacetylase 1 , Histone Deacetylase 2 , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Mice , Molecular Sequence Data , Nuclear Receptor Co-Repressor 2 , RNA, Long Noncoding , RNA, Untranslated , Receptors, Estrogen/metabolism
10.
FEBS Lett ; 494(3): 141-4, 2001 Apr 13.
Article in English | MEDLINE | ID: mdl-11311229

ABSTRACT

Recent investigations have allowed the identification of an increasing number of distinct nuclear multi-component complexes containing several types of enzymatic activity. Many of the complexes containing histone deacetylases are believed to control transcription and chromatin remodeling. We suggest here that at least some of these abundant complexes are likely to be "molecular reservoirs" of dynamic composition that exchange factors with other less abundant and functional complexes, according to specific required activities.


Subject(s)
Histone Deacetylases/metabolism , Models, Biological , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/classification , Macromolecular Substances , Multienzyme Complexes/chemistry , Multienzyme Complexes/classification , Multienzyme Complexes/metabolism , Protein Subunits , Protein Transport
11.
Biomed Chromatogr ; 15(2): 79-82, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11268046

ABSTRACT

A technique involving rapid sampling of cephaloridine in rat blood was achieved using a combination of microdialysis and sensitive microbore liquid chromatography. A microdialysis probe was inserted into the jugular vein/right atrium of a Sprague-Dawley rat. Then after a real-time collection of the analyte by microdialysis, the dialysate was automatically injected into a liquid chromatographic system via an on-line injector. Following a 2 h stabilization period after the surgical procedure, cephaloridine (20 mg/kg, i.v.) was then administered via the femoral vein. Isocratic elution of cephaloridine was carried out with a mobile phase containing methanol-20 mM monosodium phosphate (25:75, v/v, pH 5.5), and the flow rate of the mobile phase was 0.05 mL/min within 10 min. Intra- and inter-assay accuracy and precision of the assay were each less than 10%. The in vivo recovery of the cephaloridine from the microdialysate was 49.7 +/- 8.0% and 42.4 +/- 8.4% for 0.5 and 1 microg/mL standards (n = 6), respectively. Based on the pharmacokinetic analysis, the elimination half-life was 32.2 +/- 8.6 min by cephaloridine administration (20 mg/kg, i.v., n = 6).


Subject(s)
Cephaloridine/blood , Cephalosporins/blood , Chromatography, Liquid/methods , Animals , Cephaloridine/pharmacokinetics , Cephalosporins/pharmacokinetics , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Reproducibility of Results
12.
J Chromatogr B Biomed Sci Appl ; 750(1): 93-8, 2001 Jan 05.
Article in English | MEDLINE | ID: mdl-11204227

ABSTRACT

To evaluate the biodisposition of ceftazidime in rat blood, a rapid and simple microbore liquid chromatographic technique together with a microdialysis sampling technique were developed. This method involves an on-line design for blood dialysate directly injected into a microbore liquid chromatographic system. The chromatographic conditions consisted of a mobile phase of methanol-acetonitrile-100 mM monosodium phosphoric acid (pH 3.0) (10:10:80, v/v/v) pumped through a microbore reversed-phase column at a flow-rate of 0.05 ml/min. With the detection wavelength set at 254 nm, a good linear correlation was observed between the peak area and the ceftazidime concentration at 0.1 to 50 microg/ml (r=0.999). Microdialysis probes, being custom-made, were screened for acceptable in vivo recovery while chromatographic resolution and detection were validated for response linearity, as well as intra-day and inter-day variabilities. This method was then applied to the pharmacokinetic profiling of ceftazidime in blood following intravenous 50 mg/kg administration to rats. The pharmacokinetics was calculated from the corrected data for dialysate concentrations of ceftazidime versus time. This method has been used to study ceftazidime pharmacokinetics in rats and has proven to be rapid and reproducible.


Subject(s)
Ceftazidime/pharmacokinetics , Chromatography, Liquid/methods , Animals , Brain/metabolism , Ceftazidime/blood , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Spectrophotometry, Ultraviolet
13.
Proc Natl Acad Sci U S A ; 97(19): 10330-5, 2000 Sep 12.
Article in English | MEDLINE | ID: mdl-10984530

ABSTRACT

Here, we describe the identification and characterization of a nuclear body (matrix-associated deacetylase body) whose formation and integrity depend on deacetylase activity. Typically, there are 20-40 0.5-microM bodies per nucleus, although the size and number can vary substantially. The structure appears to contain both class I and the recently described class II histone deacetylases (HDAC)5 and 7 along with the nuclear receptor corepressors SMRT (silencing mediator for retinoid and thyroid receptor) and N-CoR (nuclear receptor corepressor). Addition of the deacetylase inhibitors trichostatin A and sodium butyrate completely disrupt these nuclear bodies, providing a demonstration that the integrity of a nuclear body is enzyme dependent. We demonstrate that HDAC5 and 7 can associate with at least 12 distinct proteins, including several members of the NuRD and Sin3A repression complexes, and appear to define a new but related complex.


Subject(s)
Cell Nucleus/enzymology , Histone Deacetylases/metabolism , Amino Acid Sequence , Animals , Cell Line , Haplorhini , Histone Deacetylases/chemistry , Microscopy, Fluorescence/methods , Molecular Sequence Data , Sequence Homology, Amino Acid
14.
Genes Dev ; 14(1): 55-66, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10640276

ABSTRACT

The transcriptional corepressor SMRT functions by mediating the repressive effect of transcription factors involved in diverse signaling pathways. The mechanism by which SMRT represses basal transcription has been proposed to involve the indirect recruitment of histone deacetylase HDAC1 via the adaptor mSin3A. In contrast to this model, a two-hybrid screen on SMRT-interacting proteins resulted in the isolation of the recently described HDAC5 and a new family member termed HDAC7. Molecular and biochemical results indicate that this interaction is direct and in vivo evidence colocalizes SMRT, mHDAC5, and mHDAC7 to a distinct nuclear compartment. Surprisingly, HDAC7 can interact with mSin3A in yeast and in mammalian cells, suggesting association of multiple repression complexes. Taken together, our results provide the first evidence that SMRT-mediated repression is promoted by class I and class II histone deacetylases and that SMRT can recruit class II histone deacetylases in a mSin3A-independent fashion.


Subject(s)
DNA-Binding Proteins/physiology , Histone Deacetylases/isolation & purification , Repressor Proteins/physiology , Amino Acid Sequence , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Molecular Sequence Data , Nuclear Receptor Co-Repressor 2 , Repressor Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Sequence Homology, Amino Acid , Transcription, Genetic
15.
Mol Cell Biol ; 19(12): 8219-25, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10567547

ABSTRACT

Homeobox (hox) proteins have been shown to regulate cell fate and segment identity by promoting the expression of specific genetic programs. In contrast to their restricted biological action in vivo, however, most homeodomain factors exhibit promiscuous DNA binding properties in vitro, suggesting a requirement for additional cofactors that enhance target site selectivity. In this regard, the pbx family of homeobox genes has been found to heterodimerize with and thereby augment the DNA binding activity of certain hox proteins on a subset of potential target sites. Here we examine the transcriptional properties of a forced hox-pbx heterodimer containing the pancreas-specific orphan homeobox factor pdx fused to pbx-1a. Compared to the pdx monomer, the forced pdx-pbx1a dimer, displayed 10- to 20-fold-higher affinity for a consensus hox-pbx binding site but was completely unable to bind a hox monomer recognition site. The pdx-pbx dimer stimulated target gene expression via an N-terminal trans-activation domain in pdx that interacts with the coactivator CREB binding protein. The pdx-pbx dimer was also found to repress transcription via a C-terminal domain in pbx-1a that associates with the corepressors SMRT and NCoR. The transcriptional properties of the pdx-pbx1 complex appear to be regulated at the level of alternative splicing; a pdx-pbx polypeptide containing the pbx1b isoform, which lacks the C-terminal extension in pbx1a, was unable to repress target gene expression via NCoR-SMRT. Since pbx1a and pbx1b are differentially expressed in endocrine versus exocrine compartments of the adult pancreas, our results illustrate a novel mechanism by which pbx proteins may modulate the expression of specific genetic programs, either positively or negatively, during development.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Trans-Activators/metabolism , Binding Sites , CREB-Binding Protein , Consensus Sequence , DNA-Binding Proteins/genetics , Dimerization , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Pre-B-Cell Leukemia Transcription Factor 1 , Protein Isoforms , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation
16.
Mol Cell ; 4(2): 175-86, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10488333

ABSTRACT

The Drosophila ecdysone receptor (EcR)/ultraspiracle (USP) heterodimer is a key regulator in molting and metamorphoric processes, activating and repressing transcription in a sequence-specific manner. Here, we report the isolation of an EcR-interacting protein, SMRTER, which is structurally divergent but functionally similar to the vertebrate nuclear corepressors SMRT and N-CoR. SMRTER mediates repression by interacting with Sin3A, a repressor known to form a complex with the histone deacetylase Rpd3/HDAC. Importantly, we identify an EcR mutant allele that fails to bind SMRTER and is characterized by developmental defects and lethality. Together, these results reveal a novel nuclear receptor cofactor that exhibits evolutionary conservation in the mechanism to achieve repression and demonstrate the essential role of repression in hormone signaling.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins , Drosophila melanogaster/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Steroid/physiology , Amino Acid Sequence , Animals , Biological Evolution , Cell Line , Chromosome Mapping , Chromosomes/genetics , Chromosomes/ultrastructure , Co-Repressor Proteins , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Female , Genetic Variation , Male , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Repressor Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Vertebrates
17.
Cell ; 96(3): 393-403, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-10025405

ABSTRACT

Nucleosomal histone modification is believed to be a critical step in the activation of RNA polymerase II-dependent transcription. p300/CBP and PCAF histone acetyltransferases (HATs) are coactivators for several transcription factors, including nuclear hormone receptors, p53, and Stat1alpha, and participate in transcription by forming an activation complex and by promoting histone acetylation. The adenoviral E1A oncoprotein represses transcriptional signaling by binding to p300/CBP and displacing PCAF and p/CIP proteins from the complex. Here, we show that E1A directly represses the HAT activity of both p300/CBP and PCAF in vitro and p300-dependent transcription in vivo. Additionally, E1A inhibits nucleosomal histone modifications by the PCAF complex and blocks p53 acetylation. These results demonstrate the modulation of HAT activity as a novel mechanism of transcriptional regulation.


Subject(s)
Acetyltransferases/antagonists & inhibitors , Adenovirus E1A Proteins/physiology , Nuclear Proteins/antagonists & inhibitors , Oncogene Proteins, Viral/physiology , Saccharomyces cerevisiae Proteins , Trans-Activators/antagonists & inhibitors , Animals , Cells, Cultured , Enzyme Activation/drug effects , HeLa Cells , Histone Acetyltransferases , Humans , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Transcriptional Activation/drug effects
18.
Genes Dev ; 13(24): 3209-16, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10617570

ABSTRACT

The association of transcription corepressors SMRT and N-CoR with retinoid and thyroid receptors results in suppression of basal transcriptional activity. A key event in nuclear receptor signaling is the hormone-dependent release of corepressor and the recruitment of coactivator. Biochemical and structural studies have identified a universal motif in coactivator proteins that mediates association with receptor LBDs. We report here the identity of complementary acting signature motifs in SMRT and N-CoR that are sufficient for receptor binding and ligand-induced release. Interestingly, the motif contains a hydrophobic core (PhixxPhiPhi) similar to that found in NR coactivators. Surprisingly, mutations in the amino acids that directly participate in coactivator binding disrupt the corepressor association. These results indicate a direct mechanistic link between activation and repression via competition for a common or at least partially overlapping binding site.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cloning, Molecular , DNA Mutational Analysis , DNA-Binding Proteins , Fungal Proteins/metabolism , Molecular Sequence Data , Nuclear Receptor Co-Repressor 1 , Protein Structure, Secondary , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/chemistry , Receptors, Thyroid Hormone/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , beta-Galactosidase/metabolism
19.
Genes Dev ; 12(15): 2269-77, 1998 Aug 01.
Article in English | MEDLINE | ID: mdl-9694793

ABSTRACT

The Delta-Notch signal transduction pathway has widespread roles in animal development in which it appears to control cell fate. CBF1/RBP-Jkappa, the mammalian homolog of Drosophila Suppressor of Hairless [Su(H)], switches from a transcriptional repressor to an activator upon Notch activation. The mechanism whereby Notch regulates this switch is not clear. In this report we show that prior to induction CBF1/RBP-Jkappa interacts with a corepressor complex containing SMRT (silencing mediator of retinoid and thyroid hormone receptors) and the histone deacetylase HDAC-1. This complex binds via the CBF1 repression domain, and mutants defective in repression fail to interact with the complex. Activation by Notch disrupts the formation of the repressor complex, thus establishing a molecular basis for the Notch switch. Finally, ESR-1, a Xenopus gene activated by Notch and X-Su(H), is induced in animal caps treated with TSA, an inhibitor of HDAC-1. The functional role for the SMRT/HDAC-1 complex in CBF1/RBP-Jkappa regulation reveals a novel genetic switch in which extracellular ligands control the status of critical nuclear cofactor complexes.


Subject(s)
Histone Deacetylases/metabolism , Membrane Proteins/metabolism , Nuclear Proteins , Receptors, Cell Surface , Transcription Factors , 3T3 Cells , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins , Histone Deacetylases/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein , Membrane Proteins/genetics , Mice , Models, Biological , Morphogenesis/genetics , Morphogenesis/physiology , Mutation , Nuclear Receptor Co-Repressor 2 , Receptor, Notch1 , Receptors, Notch , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Signal Transduction , Transfection , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...