Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 4630, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29545534

ABSTRACT

The general transcription factor IID (TFIID) is the first component of the preinitiation complex (PIC) to bind the core promoter of RNA polymerase II transcribed genes. Despite its critical role in protein-encoded gene expression, how TFIID engages promoter DNA remains elusive. We have previously revealed a winged-helix DNA-binding domain in the N-terminal region of the largest TFIID subunit, TAF1. Here, we report the identification of a second DNA-binding module in the C-terminal half of human TAF1, which is encoded by a previously uncharacterized conserved zinc knuckle domain. We show that the TAF1 zinc knuckle aids in the recruit of TFIID to endogenous promoters vital for cellular proliferation. Mutation of the TAF1 zinc knuckle with defects in DNA binding compromises promoter occupancy of TFIID, which leads to a decrease in transcription and cell viability. Together, our studies provide a foundation to understand how TAF1 plays a central role in TFIID promoter binding and regulation of transcription initiation.


Subject(s)
DNA/metabolism , Histone Acetyltransferases/metabolism , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Zinc/metabolism , Amino Acid Motifs , Amino Acid Sequence , DNA/chemistry , HEK293 Cells , Histone Acetyltransferases/chemistry , Humans , Models, Molecular , Protein Conformation , Sequence Homology , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Zinc/chemistry
2.
Neuronal Signal ; 2(3): NS20180141, 2018 Sep.
Article in English | MEDLINE | ID: mdl-32714589

ABSTRACT

We investigated the genome of a 5-year-old male who presented with global developmental delay (motor, cognitive, and speech), hypotonia, possibly ataxia, and cerebellar hypoplasia of unknown origin. Whole genome sequencing (WGS) and mRNA sequencing (RNA-seq) were performed on a family having an affected proband, his unaffected parents, and maternal grandfather. To explore the molecular and functional consequences of the variant, we performed cell proliferation assays, quantitative real-time PCR (qRT-PCR) array, immunoblotting, calcium imaging, and neurite outgrowth experiments in SH-SY5Y neuroblastoma cells to compare the properties of the wild-type TATA-box-binding protein factor 1 (TAF1), deletion of TAF1, and TAF1 variant p.Ser1600Gly samples. The whole genome data identified several gene variants. However, the genome sequence data failed to implicate a candidate gene as many of the variants were of unknown significance. By combining genome sequence data with transcriptomic data, a probable candidate variant, p.Ser1600Gly, emerged in TAF1. Moreover, the RNA-seq data revealed a 90:10 extremely skewed X-chromosome inactivation (XCI) in the mother. Our results showed that neuronal ion channel genes were differentially expressed between TAF1 deletion and TAF1 variant p.Ser1600Gly cells, when compared with their respective controls, and that the TAF1 variant may impair neuronal differentiation and cell proliferation. Taken together, our data suggest that this novel variant in TAF1 plays a key role in the development of a recently described X-linked syndrome, TAF1 intellectual disability syndrome, and further extends our knowledge of a potential link between TAF1 deficiency and defects in neuronal cell function.

3.
J Biol Chem ; 291(35): 18210-21, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27382054

ABSTRACT

The α1D-adrenergic receptor (ADRA1D) is a key regulator of cardiovascular, prostate, and central nervous system functions. This clinically relevant G protein-coupled receptor has proven difficult to study, as it must form an obligate modular homodimer containing the PDZ proteins scribble and syntrophin or become retained in the endoplasmic reticulum as non-functional protein. We previously determined that targeted removal of the N-terminal (NT) 79 amino acids facilitates ADRA1D plasma membrane expression and agonist-stimulated functional responses. However, whether such an event occurs in physiological contexts was unknown. Herein, we report the ADRA1D is subjected to innate NT processing in cultured human cells. SNAP near-infrared imaging and tandem-affinity purification revealed the ADRA1D is expressed as both full-length and NT truncated forms in multiple human cell lines. Serial truncation mapping identified the cleavage site as Leu(90)/Val(91) in the 95-amino acid ADRA1D NT domain, suggesting human cells express a Δ1-91 ADRA1D species. Tandem-affinity purification MS/MS and co-immunoprecipitation analysis indicate NT processing of ADRA1D is not required to form scribble-syntrophin macromolecular complexes. Yet, label-free dynamic mass redistribution signaling assays demonstrate that Δ1-91 ADRA1D agonist responses were greater than WT ADRA1D. Mutagenesis of the cleavage site nullified the processing event, resulting in ADRA1D agonist responses less than the WT receptor. Thus, we propose that processing of the ADRA1D NT domain is a physiological mechanism employed by cells to generate a functional ADRA1D isoform with optimal pharmacodynamic properties.


Subject(s)
Neoplasm Proteins/metabolism , Proteolysis , Receptors, Adrenergic, alpha-1/metabolism , Hep G2 Cells , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , PDZ Domains , Receptors, Adrenergic, alpha-1/genetics
4.
Am J Hum Genet ; 97(6): 922-32, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26637982

ABSTRACT

We describe an X-linked genetic syndrome associated with mutations in TAF1 and manifesting with global developmental delay, intellectual disability (ID), characteristic facial dysmorphology, generalized hypotonia, and variable neurologic features, all in male individuals. Simultaneous studies using diverse strategies led to the identification of nine families with overlapping clinical presentations and affected by de novo or maternally inherited single-nucleotide changes. Two additional families harboring large duplications involving TAF1 were also found to share phenotypic overlap with the probands harboring single-nucleotide changes, but they also demonstrated a severe neurodegeneration phenotype. Functional analysis with RNA-seq for one of the families suggested that the phenotype is associated with downregulation of a set of genes notably enriched with genes regulated by E-box proteins. In addition, knockdown and mutant studies of this gene in zebrafish have shown a quantifiable, albeit small, effect on a neuronal phenotype. Our results suggest that mutations in TAF1 play a critical role in the development of this X-linked ID syndrome.


Subject(s)
Developmental Disabilities/genetics , Histone Acetyltransferases/genetics , Intellectual Disability/genetics , Neurodegenerative Diseases/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Adolescent , Animals , Child , Child, Preschool , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Disease Models, Animal , E-Box Elements , Facies , Family , Gene Expression Regulation , Histone Acetyltransferases/metabolism , Humans , Infant , Inheritance Patterns , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mutation , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Pedigree , Phenotype , Signal Transduction , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Young Adult , Zebrafish
5.
Cell Res ; 24(12): 1433-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25412659

ABSTRACT

The general transcription factor IID (TFIID) initiates RNA polymerase II-mediated eukaryotic transcription by nucleating pre-initiation complex formation at the core promoter of protein-encoding genes. TAF1, the largest integral subunit of TFIID, contains an evolutionarily conserved yet poorly characterized central core domain, whose specific mutation disrupts cell proliferation in the temperature-sensitive mutant hamster cell line ts13. Although the impaired TAF1 function in the ts13 mutant has been associated with defective transcriptional regulation of cell cycle genes, the mechanism by which TAF1 mediates transcription as part of TFIID remains unclear. Here, we present the crystal structure of the human TAF1 central core domain in complex with another conserved TFIID subunit, TAF7, which biochemically solubilizes TAF1. The TAF1-TAF7 complex displays an inter-digitated compact architecture, featuring an unexpected TAF1 winged helix (WH) domain mounted on top of a heterodimeric triple barrel. The single TAF1 residue altered in the ts13 mutant is buried at the junction of these two structural domains. We show that the TAF1 WH domain has intrinsic DNA-binding activity, which depends on characteristic residues that are commonly used by WH fold proteins for interacting with DNA. Importantly, mutations of these residues not only compromise DNA binding by TAF1, but also abrogate its ability to rescue the ts13 mutant phenotype. Together, our results resolve the structural organization of the TAF1-TAF7 module in TFIID and unveil a critical promoter-binding function of TAF1 in transcription regulation.


Subject(s)
Histone Acetyltransferases/chemistry , TATA-Binding Protein Associated Factors/chemistry , Transcription Factor TFIID/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cricetinae , Crystallography, X-Ray , DNA/metabolism , Histone Acetyltransferases/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Promoter Regions, Genetic , Protein Conformation , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism
6.
Mol Cell Biol ; 32(16): 3358-69, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22711989

ABSTRACT

The largest transcription factor IID (TFIID) subunit, TBP-associated factor 1 (TAF1), possesses protein kinase and histone acetyltransferase (HAT) activities. Both enzymatic activities are essential for transcription from a subset of genes and G(1) progression in mammalian cells. TAF7, another TFIID subunit, binds TAF1 and inhibits TAF1 HAT activity. Here we present data demonstrating that disruption of the TAF1/TAF7 interaction within TFIID by protein phosphorylation leads to activation of TAF1 HAT activity and stimulation of cyclin D1 and cyclin A gene transcription. Overexpression and small interfering RNA knockdown experiments confirmed that TAF7 functions as a transcriptional repressor at these promoters. Release of TAF7 from TFIID by TAF1 phosphorylation of TAF7 increased TAF1 HAT activity and elevated histone H3 acetylation levels at the cyclin D1 and cyclin A promoters. Serine-264 of TAF7 was identified as a substrate for TAF1 kinase activity. Using TAF7 S264A and S264D phosphomutants, we determined that the phosphorylation state of TAF7 at S264 influences the levels of cyclin D1 and cyclin A gene transcription and promoter histone H3 acetylation. Our studies have uncovered a novel function for the TFIID subunit TAF7 as a phosphorylation-dependent regulator of TAF1-catalyzed histone H3 acetylation at the cyclin D1 and cyclin A promoters.


Subject(s)
Cyclin A/metabolism , Cyclin D1/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/chemistry , Animals , Cell Cycle , Cell Nucleus/metabolism , HeLa Cells , Histone Acetyltransferases , Histones/metabolism , Humans , Insecta/cytology , Phosphorylation , Protein Interaction Mapping/methods , Serine/chemistry , Transcription Factor TFIID/metabolism , Transfection
7.
Hybridoma (Larchmt) ; 30(2): 181-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21529292

ABSTRACT

Muscleblind-like 3 (MBNL3) belongs to a family of RNA binding proteins that regulate alternative splicing. We have generated a set of monoclonal antibodies (MAbs) against mouse MBNL3, three of which do not cross-react with the other muscleblind-like (MBNL) proteins, MBNL1 and MBNL2. Epitope mapping revealed that MAbs P1C7, P1E7, SP1C2, and P2E6 recognize distinct, non-overlapping segments of the MBNL3 polypeptide sequence. Immunohistochemical staining of proliferating muscle precursor cells localized MBNL3 to the nucleus in a punctate pattern, characteristic of subcellular structures in the nucleus enriched in pre-messenger RNA splicing factors. Although MBNL3 did not co-localize with SC35 and PSP1 (widely used markers of splicing speckles and paraspeckles), the punctate localization pattern of MBNL3 within interchromatin regions of the nucleus is highly predictive of proteins involved in pre-mRNA processing. Monoclonal antibodies specific for mouse MBNL3 will facilitate further investigation of the expression pattern and unique functions of this splicing factor during development and in different adult mouse tissues.


Subject(s)
Antibodies, Monoclonal , Antibody Specificity , Carrier Proteins/metabolism , Gene Expression Regulation, Developmental , Myoblasts/metabolism , Protein Isoforms/metabolism , Recombinant Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/genetics , Chromatin/metabolism , Cloning, Molecular , Epitope Mapping , Escherichia coli , Hybridomas/immunology , Hybridomas/metabolism , Immunization , Mice , Mice, Inbred Strains , Molecular Sequence Data , Myoblasts/cytology , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA Precursors/metabolism , RNA-Binding Proteins , Recombinant Proteins/genetics , Recombinant Proteins/immunology
8.
J Biol Chem ; 285(44): 33779-87, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20709755

ABSTRACT

Mammalian MBNL (muscleblind-like) proteins are regulators of alternative splicing and have been implicated in myotonic dystrophy, the most common form of adult onset muscular dystrophy. MBNL3 functions as an inhibitor of muscle differentiation and is expressed in proliferating muscle precursor cells but not in differentiated skeletal muscle. Here we demonstrate that MBNL3 regulates the splicing pattern of the muscle transcription factor myocyte enhancer factor 2 (Mef2) by promoting exclusion of the alternatively spliced ß-exon. Expression of the transcriptionally more active (+)ß isoform of Mef2D was sufficient to overcome the inhibitory effects of MBNL3 on muscle differentiation. These data suggest that MBNL3 antagonizes muscle differentiation by disrupting Mef2 ß-exon splicing. MBNL3 regulates Mef2D splicing by directly binding to intron 7 downstream of the alternatively spliced exon in the pre-mRNA. The RNA binding activity of MBNL3 requires the CX(7)CX(4-6)CX(3)H zinc finger domains. Using a cell culture model of myotonic dystrophy and myotonic dystrophy patient tissue, we have evidence that expression of CUG expanded RNAs can lead to an increase in MBNL3 expression and a decrease in Mef2D ß-exon splicing. These studies suggest that elevating MBNL3 activity in myogenic cells could lead to muscle degeneration disorders such as myotonic dystrophy.


Subject(s)
Carrier Proteins/metabolism , Myogenic Regulatory Factors/metabolism , RNA/metabolism , Alternative Splicing , Animals , Cell Differentiation , Cell Line , Exons , Immunohistochemistry/methods , MEF2 Transcription Factors , Mice , Models, Genetic , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myotonic Dystrophy/metabolism , Myotonic Dystrophy/pathology , RNA-Binding Proteins , Retroviridae/metabolism
9.
Biochem Biophys Res Commun ; 393(4): 603-8, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20152817

ABSTRACT

Precise spatial and temporal expression of the recently identified G-protein coupled receptor GPR54 is critical for proper reproductive function and metastasis suppression. However, regulatory factors that control GPR54 expression remain unknown. Thus, the identification of these cis-acting DNA elements can provide insight into the role of GPR54 in reproduction and cancer. Using luciferase reporter, electrophoretic mobility shift, and chromatin immunoprecipitation assays, we demonstrate that three SP1 sites and a partial estrogen response element modulate mouse GPR54 (mGPR54) promoter activity. Supporting experiments show transcription factor SP1 binds directly to the mGPR54 promoter region and activates gene expression. In conclusion, these novel findings now identify factors that regulate activity of the mGPR54 promoter, and these factors are highly conserved across multiple mammalian species.


Subject(s)
Pituitary Gland/metabolism , Receptors, G-Protein-Coupled/genetics , Response Elements , Sp1 Transcription Factor/metabolism , Transcription, Genetic , Transcriptional Activation , Animals , Base Sequence , Cell Line , Estrogens/metabolism , Estrogens/pharmacology , Genes, Reporter , Genome , Luciferases/genetics , Mice , Molecular Sequence Data , Pituitary Gland/cytology , Pituitary Gland/drug effects , Receptors, Kisspeptin-1
10.
Differentiation ; 76(3): 299-309, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17697127

ABSTRACT

Muscle differentiation is controlled by positive and negative signals. While much attention has been placed on proteins that promote muscle formation, the importance of negative regulators has been underemphasized. MBNL3/CHCR belongs to the muscleblind family of Cys3His zinc finger proteins implicated in myotonic dystrophy. MBNL3 is expressed in myoblasts, muscle precursor cells, and during the early stages of myogenesis, but is detected at very low levels in terminally differentiated myotubes. Constitutive expression of MBNL3 inhibits myotube formation and antagonizes myogenin and myosin heavy chain expression. To identify MBNL3 target genes, we compared the expression profile of C2C12 mouse myoblasts that constitutively express MBNL3 with control cells. From the 15,247 genes represented on the DNA microarray, classification by biological function indicated that genes involved in muscle development/contraction and cell adhesion were down-regulated by MBNL3 expression. mRNA and protein levels for the muscle transcription factor MyoD and E-box regulated transcription were reduced in C2C12-MBNL3 expressing cells. We hypothesize that MBNL3 serves to antagonize muscle differentiation by suppressing MyoD expression levels to prevent unwanted myogenic gene transcription. These findings are the first indication that a mammalian muscleblind-like (MBNL) protein plays a regulatory role in muscle differentiation under nonpathogenic conditions.


Subject(s)
Carrier Proteins/physiology , Cell Differentiation , MyoD Protein/physiology , Transcription, Genetic , Animals , Base Sequence , Blotting, Western , Cell Line , DNA Primers , Down-Regulation , Immunohistochemistry , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA-Binding Proteins
11.
Biochem Biophys Res Commun ; 361(1): 151-5, 2007 Sep 14.
Article in English | MEDLINE | ID: mdl-17644069

ABSTRACT

Muscleblind-like (MBNL) proteins are believed to be regulators of myogenesis and are implicated in myotonic dystrophy. While Drosophila melanogaster muscleblind is required for terminal muscle differentiation, mammalian MBNL3 functions as an inhibitor of myogenesis. In this study, we analyzed the expression pattern of MBNL3 in different adult mouse tissues and tissue culture cells. MBNL3 transcript is enriched in the lung, spleen, and testis and not in heart and skeletal muscle. By Western blotting, we found that MBNL3 was expressed in C2C12 myoblasts and ts13 myofibroblasts, but was detected at significantly lower levels in fibroblasts. MBNL3 protein levels decreased when cells were shifted to muscle differentiation conditions, but the closely related MBNL1 protein was unaffected. These results suggest that myoblasts and fibroblasts respond to differentiation conditions by activating signaling pathways that repress MBNL3 but not MBNL1 expression.


Subject(s)
Carrier Proteins/metabolism , DNA-Binding Proteins/metabolism , Myoblasts/metabolism , RNA-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Differentiation , Cell Line , Cricetinae , Fibroblasts/metabolism , Gene Expression , Mice , Myoblasts/cytology , Nuclear Proteins/metabolism , Tissue Distribution
12.
Mol Cell Biol ; 25(10): 4321-32, 2005 May.
Article in English | MEDLINE | ID: mdl-15870300

ABSTRACT

A missense mutation within the histone acetyltransferase (HAT) domain of the TATA binding protein-associated factor TAF1 induces ts13 cells to undergo a late G(1) arrest and decreases cyclin D1 transcription. We have found that TAF1 mutants (Delta844-850 and Delta848-850, from which amino acids 844 through 850 and 848 through 850 have been deleted, respectively) deficient in HAT activity are unable to complement the ts13 defect in cell proliferation and cyclin D1 transcription. Chromatin immunoprecipitation assays revealed that histone H3 acetylation was reduced at the cyclin D1 promoter but not the c-fos promoter upon inactivation of TAF1 in ts13 cells. The hypoacetylation of H3 at the cyclin D1 promoter was reversed by treatment with trichostatin A (TSA), a histone deacetylase inhibitor, or by expression of TAF1 proteins that retain HAT activity. Transcription of a chimeric promoter containing the Sp1 sites of cyclin D1 and c-fos core remained TAF1 dependent in ts13 cells. Treatment with TSA restored full activity to the cyclin D1-c-fos chimera at 39.5 degrees C. In vivo genomic footprinting experiments indicate that protein-DNA interactions at the Sp1 sites of the cyclin D1 promoter were compromised at 39.5 degrees C in ts13 cells. These data have led us to hypothesize that TAF1-dependent histone acetylation facilitates transcription factor binding to the Sp1 sites, thereby activating cyclin D1 transcription and ultimately G(1)-to-S-phase progression.


Subject(s)
Acetyltransferases/metabolism , Cyclin D1/genetics , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Transcriptional Activation/genetics , Acetylation/drug effects , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/chemistry , Acetyltransferases/genetics , Amino Acid Sequence , Base Sequence , Cell Line , Genetic Complementation Test , Histone Acetyltransferases , Histones/metabolism , Humans , Molecular Sequence Data , Mutation/genetics , Phenotype , Protein Structure, Tertiary , Response Elements/genetics , TATA-Binding Protein Associated Factors/chemistry , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/chemistry , Transcription Factor TFIID/genetics , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transcriptional Activation/drug effects
13.
Proc Natl Acad Sci U S A ; 102(2): 497-502, 2005 Jan 11.
Article in English | MEDLINE | ID: mdl-15625104

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a major regulator of monocyte to macrophage differentiation. In both humans and mice, the main phenotype of decreased GM-CSF function is pulmonary proteinosis due to aberrant function of alveolar macrophages. Recently, this cytokine has been shown to up-regulate a cyclic nucleotide phosphodiesterase, PDE1B. Two PDE1B variants with unique N-terminal sequences, PDE1B1 and PDE1B2, have been identified. Here, we report that the previously uncharacterized PDE1B2 is selectively increased by GM-CSF by stimulation of transcription at a previously unknown transcriptional start site. Analysis of the exon and intron organization of the PDE1B gene reveals that PDE1B2 has a different N-terminal sequence because of a separate first exon that is located 11.5 kb downstream from the PDE1B1 first exon. By using 5'-RACE, alignment of EST sequences, and a luciferase-reporter system, we provide evidence that PDE1B2 has a separate transcriptional start site from PDE1B1 that can be activated by monocyte differentiation. Furthermore, IL-4 treatment in the presence of GM-CSF, which shifts the differentiation from a macrophage to a dendritic cell phenotype, suppresses the up-regulation of PDE1B2. Induction of PDE1B2 is also found in T cells upon activation by PHA. Therefore, PDE1B2 may have a regulatory role in multiple immune cell types. Last, characterization of the catalytic properties of recombinant PDE1B2 shows that it prefers cGMP over cAMP as a substrate and, thus, is likely to regulate cGMP in macrophages. Also, PDE1B2 has a nearly 3-fold lower EC(50) for activation by calmodulin than PDE1B1.


Subject(s)
Macrophages/cytology , Monocytes/cytology , Phosphoric Diester Hydrolases/physiology , Cell Differentiation , Cells, Cultured , Cyclic Nucleotide Phosphodiesterases, Type 1 , Dendritic Cells/physiology , Humans , Interleukin-4/pharmacology , Kinetics , Phosphoric Diester Hydrolases/genetics , Promoter Regions, Genetic , RNA, Messenger/analysis , Transcription Initiation Site , Up-Regulation
14.
Mol Cell Biol ; 24(6): 2296-307, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14993269

ABSTRACT

Histone deacetylase inhibitors (HDIs) induce cell cycle arrest, differentiation, or apoptosis in numerous cancer cell types both in vivo and in vitro. These dramatic effects are the result of a specific reprogramming of gene expression. However, the mechanism by which these agents activate the transcription of some genes, such as p21(WAF1), but repress others, such as cyclin D1, is currently unknown. We have been studying the human SRC gene as a model for HDI-mediated transcriptional repression. We found previously that both the tissue-specific and housekeeping SRC promoters were equally repressed by HDIs. Here we show that, despite an overt dissimilarity, both SRC promoters do share similar core promoter elements and transcription is TAF1 dependent. Detailed analysis of the SRC promoters suggested that both core and proximal promoter elements were responsible for HDI-mediated repression. This was confirmed in a series of promoter-swapping experiments with the HDI-inducible, TAF1-independent p21(WAF1) promoter. Remarkably, all the SRC-p21(WAF1) chimeric promoter constructs were not only repressed by HDIs but also dependent on TAF1. Together these experiments suggest that the overall promoter architecture, rather than discrete response elements, is responsible for HDI-mediated repression, and they implicate core promoter elements in particular as potential mediators of this response.


Subject(s)
Genes, src , Histone Deacetylase Inhibitors , Promoter Regions, Genetic , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIID/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , Cell Line, Tumor , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA, Complementary/genetics , Enzyme Inhibitors/pharmacology , Genes, src/drug effects , Histone Acetyltransferases , Humans , Mutagenesis, Site-Directed , Promoter Regions, Genetic/drug effects , Sequence Homology, Nucleic Acid , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Transcription, Genetic/drug effects , Transfection
15.
J Biol Chem ; 278(15): 12992-3002, 2003 Apr 11.
Article in English | MEDLINE | ID: mdl-12569092

ABSTRACT

Cyclin D1 is an oncogene that regulates progression through the G(1) phase of the cell cycle. A temperature-sensitive missense mutation in the transcription factor TAF1/TAF(II)250 induces the mutant ts13 cells to arrest in late G(1) by decreasing transcription of cell cycle regulators, including cyclin D1. Here we provide evidence that TAF1 serves two independent functions, one at the core promoter and one at the upstream activating Sp1 sites of the cyclin D1 gene. Using in vivo genomic footprinting, we have identified protein-DNA interactions within the cyclin D1 core promoter that are disrupted upon inactivation of TAF1 in ts13 cells. This 33-bp segment, which we termed the TAF1-dependent element 1 (TDE1), contains an initiation site that displays homology to the consensus motif and is sufficient to confer a requirement for TAF1 function. Electrophoretic mobility shift assays reveal that binding of ts13-TAF1-containing TFIID complexes to the cyclin D1 TDE1 occurs at 25 degrees C but not at 37 degrees C in vitro and involves the initiator element. Temperature-dependent DNA binding activity is also observed for TAF1-TAF2 heterodimers assembled with the ts13 mutant but not the wild-type TAF1 protein. These data suggest that a function of TAF is required for the interaction of TFIID with the cyclin D1 initiator. Our finding that recruitment of TFIID, by insertion of a TBP binding site upstream of the TDE1, restores basal but not activated transcription supports the model that TAF1 carries out two independent functions at the cyclin D1 promoter.


Subject(s)
Cyclin D1/genetics , Gene Expression Regulation/physiology , Podophyllin/analogs & derivatives , Podophyllin/metabolism , Transcription Factor TFIID/metabolism , 5' Untranslated Regions/genetics , Animals , Base Sequence , Binding Sites , Cell Line , Chromosomal Proteins, Non-Histone/metabolism , Cricetinae , Cyclin D1/metabolism , DNA Footprinting , DNA Primers , Kidney , Molecular Sequence Data , Podophyllotoxin/analogs & derivatives , Promoter Regions, Genetic , Protein Subunits/metabolism , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Transfection
16.
Dev Biol ; 250(1): 218-30, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12297108

ABSTRACT

Growth factor withdrawal from proliferating myoblasts induces the expression of muscle-specific genes essential for myogenesis. By suppression subtractive hybridization (SSH), we have cloned a novel human cDNA that encodes a Cys3His zinc finger protein named CHCR (Cys3His CCG1-Required). CHCR is related to Muscleblind (Mbl), a Drosophila melanogaster protein required for terminal muscle differentiation. It also displays sequence similarity to EXP/MBNL, a human Mbl protein that interacts with CUG expansions associated with the degenerative muscular disease, myotonic dystrophy (DM1). This relationship with EXP/MBNL and Mbl suggests that CHCR also functions during muscle differentiation. We have found that CHCR mRNA and protein levels decrease upon differentiation of mouse myoblast cells. Constitutive expression of CHCR in C2C12 cells inhibits the induction of sarcomeric myosin heavy chain (MyHC) upon serum deprivation. Induction of myogenin, an earlier marker of muscle differentiation, is inhibited to a lesser extent, while expression of the cell cycle inhibitor, p21, remains unaffected. Loss of CHCR function by morpholino antisense oligonucleotide treatment accelerates MyHC induction during differentiation of myoblast cells. These complementary gain- and loss-of-function results suggest that CHCR is an inhibitor of myogenesis. CHCR represents the first muscleblind-related protein that antagonizes, instead of promotes, muscle differentiation.


Subject(s)
Carrier Proteins/physiology , Muscles/cytology , Proteins/physiology , RNA-Binding Proteins , Zinc Fingers , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , Cell Differentiation , Cell Line , Cloning, Molecular , Cricetinae , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , DNA, Complementary , Drosophila Proteins , Gene Expression , Gene Targeting , HeLa Cells , Humans , Mice , Molecular Sequence Data , Myogenin/biosynthesis , Myosin Heavy Chains/biosynthesis , Nuclear Proteins , Oligodeoxyribonucleotides, Antisense , Proteins/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...