Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 21(2): 321-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24270405

ABSTRACT

The Groucho/transducin-like Enhancer of split 1 (Gro/TLE1):Hes1 transcriptional repression complex acts in cerebral cortical neural progenitor cells to inhibit neuronal differentiation. The molecular mechanisms that regulate the anti-neurogenic function of the Gro/TLE1:Hes1 complex during cortical neurogenesis remain to be defined. Here we show that prolyl isomerase Pin1 (peptidyl-prolyl cis-trans isomerase NIMA-interacting 1) and homeodomain-interacting protein kinase 2 (HIPK2) are expressed in cortical neural progenitor cells and form a complex that interacts with the Gro/TLE1:Hes1 complex. This association depends on the enzymatic activities of both HIPK2 and Pin1, as well as on the association of Gro/TLE1 with Hes1, but is independent of the previously described Hes1-activated phosphorylation of Gro/TLE1. Interaction with the Pin1:HIPK2 complex results in Gro/TLE1 hyperphosphorylation and weakens both the transcriptional repression activity and the anti-neurogenic function of the Gro/TLE1:Hes1 complex. These results provide evidence that HIPK2 and Pin1 work together to promote cortical neurogenesis, at least in part, by suppressing Gro/TLE1:Hes1-mediated inhibition of neuronal differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carrier Proteins/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Peptidylprolyl Isomerase/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Mice , NIMA-Interacting Peptidylprolyl Isomerase , Naphthoquinones/pharmacology , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Peptidylprolyl Isomerase/antagonists & inhibitors , Transcription Factor HES-1 , Tretinoin/pharmacology
2.
Oncogene ; 33(32): 4173-84, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-24213577

ABSTRACT

The Polycomb group (PcG) proteins regulate stem cell differentiation via the repression of gene transcription, and their deregulation has been widely implicated in cancer development. The PcG protein Enhancer of Zeste Homolog 2 (EZH2) works as a catalytic subunit of the Polycomb Repressive Complex 2 (PRC2) by methylating lysine 27 on histone H3 (H3K27me3), a hallmark of PRC2-mediated gene repression. In skeletal muscle progenitors, EZH2 prevents an unscheduled differentiation by repressing muscle-specific gene expression and is downregulated during the course of differentiation. In rhabdomyosarcoma (RMS), a pediatric soft-tissue sarcoma thought to arise from myogenic precursors, EZH2 is abnormally expressed and its downregulation in vitro leads to muscle-like differentiation of RMS cells of the embryonal variant. However, the role of EZH2 in the clinically aggressive subgroup of alveolar RMS, characterized by the expression of PAX3-FOXO1 oncoprotein, remains unknown. We show here that EZH2 depletion in these cells leads to programmed cell death. Transcriptional derepression of F-box protein 32 (FBXO32) (Atrogin1/MAFbx), a gene associated with muscle homeostasis, was evidenced in PAX3-FOXO1 RMS cells silenced for EZH2. This phenomenon was associated with reduced EZH2 occupancy and H3K27me3 levels at the FBXO32 promoter. Simultaneous knockdown of FBXO32 and EZH2 in PAX3-FOXO1 RMS cells impaired the pro-apoptotic response, whereas the overexpression of FBXO32 facilitated programmed cell death in EZH2-depleted cells. Pharmacological inhibition of EZH2 by either 3-Deazaneplanocin A or a catalytic EZH2 inhibitor mirrored the phenotypic and molecular effects of EZH2 knockdown in vitro and prevented tumor growth in vivo. Collectively, these results indicate that EZH2 is a key factor in the proliferation and survival of PAX3-FOXO1 alveolar RMS cells working, at least in part, by repressing FBXO32. They also suggest that the reducing activity of EZH2 could represent a novel adjuvant strategy to eradicate high-risk PAX3-FOXO1 alveolar RMS.


Subject(s)
Forkhead Transcription Factors/metabolism , Muscle Proteins/antagonists & inhibitors , Paired Box Transcription Factors/metabolism , Polycomb Repressive Complex 2/physiology , Rhabdomyosarcoma, Alveolar/metabolism , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Adolescent , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Child , Enhancer of Zeste Homolog 2 Protein , Female , Forkhead Box Protein O1 , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Homeostasis , Humans , Male , Muscle Proteins/physiology , PAX3 Transcription Factor , SKP Cullin F-Box Protein Ligases/physiology
3.
Cell Death Differ ; 19(5): 871-81, 2012 May.
Article in English | MEDLINE | ID: mdl-22117196

ABSTRACT

Rhabdomyosarcoma (RMS) is a paediatric soft-tissue sarcoma arising from skeletal muscle precursors coexpressing markers of proliferation and differentiation. Inducers of myogenic differentiation suppress RMS tumourigenic phenotype. The Notch target gene HES1 is upregulated in RMS and prevents tumour cell differentiation in a Notch-dependent manner. However, Notch receptors regulating this phenomenon are unknown. In agreement with data in RMS primary tumours, we show here that the Notch3 receptor is overexpressed in RMS cell lines versus normal myoblasts. Notch3-targeted downregulation in RMS cells induces hyper-phosphorylation of p38 and Akt essential for myogenesis, resulting in the differentiation of tumour cells into multinucleated myotubes expressing Myosin Heavy Chain. These phenomena are associated to a marked decrease in HES1 expression, an increase in p21(Cip1) level and the accumulation of RMS cells in the G1 phase. HES1-forced overexpression in RMS cells reverses, at least in part, the pro-differentiative effects of Notch3 downregulation. Notch3 depletion also reduces the tumourigenic potential of RMS cells both in vitro and in vivo. These results indicate that downregulation of Notch3 is sufficient to force RMS cells into completing a correct full myogenic program providing evidence that it contributes, partially through HES1 sustained expression, to their malignant phenotype. Moreover, they suggest Notch3 as a novel potential target in human RMS.


Subject(s)
Cell Differentiation/physiology , Receptors, Notch/metabolism , Rhabdomyosarcoma/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21 , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/genetics , Phosphorylation/physiology , RNA Interference , Real-Time Polymerase Chain Reaction , Receptor, Notch3 , Receptors, Notch/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/therapy , Signal Transduction , Xenograft Model Antitumor Assays
4.
Neuroscience ; 146(1): 236-47, 2007 Apr 25.
Article in English | MEDLINE | ID: mdl-17331656

ABSTRACT

Aristaless-related homeobox gene (ARX) is an important paired-type homeobox gene involved in the development of human brain. The ARX gene mutations are a significant contributor to various forms of X-chromosome-linked mental retardation with and without additional features including epilepsy, lissencephaly with abnormal genitalia, hand dystonia or autism. Here we demonstrate that the human ARX protein is a potent transcriptional repressor, which binds to Groucho/transducin-like enhancer of split (TLE) co-factor proteins and the TLE1 in particular through its octapeptide (Engrailed homology repressor domain (eh-1) homology) domain. We show that the transcription repression activity of ARX is modulated by two strong repression domains, one located within the octapeptide domain and the second in the region of the polyalanine tract 4, and one activator domain, the aristaless domain. Importantly, we show that the transcription repression activity of ARX is affected by various naturally occurring mutations. The introduction of the c.98T>C (p.L33P) mutation results in the lack of binding to TLE1 protein and relaxed transcription repression. The introduction of the two most frequent ARX polyalanine tract expansion mutations increases the repression activity in a manner dependent on the number of extra alanines. Interestingly, deletions of alanine residues within polyalanine tracts 1 and 2 show low or no effect. In summary we demonstrate that the ARX protein is a strong transcription repressor, we identify novel ARX interacting proteins (TLE) and offer an explanation of a molecular pathogenesis of some ARX mutations, including the most frequent ARX mutations, the polyalanine tract expansion mutations, c.304ins(GCG)7 and c.428_451dup.


Subject(s)
Enhancer Elements, Genetic/physiology , Homeodomain Proteins/genetics , Mutation , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic/physiology , Age Factors , Alanine/genetics , Animals , Brain/cytology , Cells, Cultured , Embryo, Mammalian , Gene Expression Regulation, Developmental/physiology , Humans , Immunoprecipitation/methods , In Situ Hybridization/methods , Mice , Neurons/metabolism , Transducin/metabolism , Transfection/methods
5.
Development ; 128(21): 4361-70, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11684670

ABSTRACT

Groucho (Gro) is the founding member of a family of transcriptional co-repressors that are recruited by a number of different transcription factors. Drosophila has a single gro gene, whose loss of function affects processes ranging from sex determination to embryonic patterning and neuroblast specification. We have characterized a function of Gro in imaginal development, namely the repression of hedgehog (hh) in anterior wing pouch cells. hh encodes a secreted morphogen with potent patterning activities. In Drosophila thoracic appendages (legs, wings, halteres), hh is expressed in posterior compartments and induces the anteroposterior (AP) pattern organizer in the cells across the AP boundary. hh is repressed in anterior compartments at least partly via Ci[rep], a form of the multifunctional transcription factor Cubitus interruptus (Ci). We show that cells in the wing primordium close to the AP boundary need gro activity to maintain repression of hh transcription, whereas in more anterior cells gro is dispensable. This repressive function of Gro does not appear to be mediated by Ci[rep]. Analysis of mutant gro transgenes has revealed that the Q and WD40 domains are both necessary for hh repression. Yet, deletion of the WD40 repeats does not always abolish Gro activity. Our findings provide new insights both into the mechanisms of AP patterning of the wing and into the function of Gro.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila/genetics , Insect Proteins/genetics , Repressor Proteins/genetics , Wings, Animal/growth & development , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors , DNA-Binding Proteins/metabolism , Drosophila/growth & development , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins , Insect Proteins/metabolism , Larva , Mutation , Repressor Proteins/metabolism , Transcription Factors , Wings, Animal/anatomy & histology
6.
J Cell Biol ; 154(6): 1161-71, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11551980

ABSTRACT

HES6 is a novel member of the family of basic helix-loop-helix mammalian homologues of Drosophila Hairy and Enhancer of split. We have analyzed the biochemical and functional roles of HES6 in myoblasts. HES6 interacted with the corepressor transducin-like Enhancer of split 1 in yeast and mammalian cells through its WRPW COOH-terminal motif. HES6 repressed transcription from an N box-containing template and also when tethered to DNA through the GAL4 DNA binding domain. On N box-containing promoters, HES6 cooperated with HES1 to achieve maximal repression. An HES6-VP16 activation domain fusion protein activated the N box-containing reporter, confirming that HES6 bound the N box in muscle cells. The expression of HES6 was induced when myoblasts fused to become differentiated myotubes. Constitutive expression of HES6 in myoblasts inhibited expression of MyoR, a repressor of myogenesis, and induced differentiation, as evidenced by fusion into myotubes and expression of the muscle marker myosin heavy chain. Reciprocally, blocking endogenous HES6 function by using a WRPW-deleted dominant negative HES6 mutant led to increased expression of MyoR and completely blocked the muscle development program. Our results show that HES6 is an important regulator of myogenesis and suggest that MyoR is a target for HES6-dependent transcriptional repression.


Subject(s)
Homeodomain Proteins , Muscle, Skeletal/cytology , Repressor Proteins/pharmacology , Transcription Factors/pharmacology , Active Transport, Cell Nucleus , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation/drug effects , Cell Line , Co-Repressor Proteins , Humans , Mice , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/drug effects , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/growth & development , Nuclear Proteins/metabolism , Protein Binding , RNA, Messenger/metabolism , Repressor Proteins/physiology , Transcription Factor HES-1 , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Two-Hybrid System Techniques , Yeasts
7.
Mol Cell Biol ; 21(17): 5935-45, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11486032

ABSTRACT

The E2A-HLF fusion gene, formed by the t(17;19)(q22;p13) chromosomal translocation in leukemic pro-B cells, encodes a chimeric transcription factor consisting of the transactivation domain of E2A linked to the bZIP DNA-binding and protein dimerization domain of hepatic leukemia factor (HLF). This oncoprotein blocks apoptosis induced by growth factor deprivation or irradiation, but the mechanism for this effect remains unclear. We therefore performed representational difference analysis (RDA) to identify downstream genetic targets of E2A-HLF, using a murine FL5.12 pro-B cell line that had been stably transfected with E2A-HLF cDNA under the control of a zinc-regulated metallothionein promoter. Two RDA clones, designated RDA1 and RDA3, were differentially upregulated in E2A-HLF-positive cells after zinc induction. The corresponding cDNAs encoded two WD40 repeat-containing proteins, Grg2 and Grg6. Both are related to the Drosophila protein Groucho, a transcriptional corepressor that lacks DNA-binding activity on its own but can act in concert with other proteins to regulate embryologic development of the fly. Expression of both Grg2 and Grg6 was upregulated 10- to 50-fold by E2A-HLF. Immunoblot analysis detected increased amounts of two additional Groucho-related proteins, Grg1 and Grg4, in cells expressing E2A-HLF. A mutant E2A-HLF protein with a disabled DNA-binding region also mediated pro-B cell survival and activated Groucho-related genes. Among the transcription factors known to interact with Groucho-related protein, only RUNX1 was appreciably downregulated by E2A-HLF. Our results identify a highly conserved family of transcriptional corepressors that are activated by E2A-HLF, and they suggest that downregulation of RUNX1 may contribute to E2A-HLF-mediated leukemogenesis.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Leucine Zippers , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Amino Acid Sequence , Animals , Apoptosis , Base Sequence , Basic-Leucine Zipper Transcription Factors , Cell Line , Cell Lineage , Co-Repressor Proteins , Core Binding Factor Alpha 2 Subunit , DNA, Complementary , Drosophila melanogaster , Humans , Leukemia, B-Cell , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Proteins/genetics , Trans-Activators/genetics , Transcriptional Activation , Up-Regulation
8.
Mol Cell Biol ; 21(6): 1962-72, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238932

ABSTRACT

Brain factor 1 (BF-1) is a winged-helix transcriptional repressor that plays important roles in both progenitor cell differentiation and regional patterning in the mammalian telencephalon. The aim of this study was to elucidate the molecular mechanisms underlying BF-1 functions. It is shown here that BF-1 interacts in vivo with global transcriptional corepressors of the Groucho family and also associates with the histone deacetylase 1 protein. The ability of BF-1 to mediate transcriptional repression is promoted by Groucho and inhibited by the histone deacetylase inhibitor trichostatin A, suggesting that BF-1 recruits Groucho and histone deacetylase activities to repress transcription. Our studies also provide the first demonstration that Groucho mediates a specific interaction between BF-1 and the basic helix-loop-helix protein Hes1 and that BF-1 potentiates transcriptional repression by Hes1 in a Groucho-dependent manner. These findings suggest that Groucho participates in the transcriptional functions of BF-1 by acting as both a corepressor and an adapter between BF-1 and Hes1. Taken together with the demonstration that these proteins are coexpressed in telencephalic neural progenitor cells, these results also suggest that complexes of BF-1, Groucho, and Hes factors may be involved in the regulation of progenitor cell differentiation in the telencephalon.


Subject(s)
DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/genetics , Forkhead Transcription Factors , Helix-Loop-Helix Motifs , Homeodomain Proteins/genetics , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Rats , Repressor Proteins/genetics , Telencephalon/cytology , Telencephalon/metabolism , Transcription Factor HES-1 , Transcription Factors/genetics
9.
J Biol Chem ; 276(2): 1578-84, 2001 Jan 12.
Article in English | MEDLINE | ID: mdl-11035023

ABSTRACT

Hairy/Enhancer of split 1 (Hes1) is a mammalian transcriptional repressor that plays crucial roles in the regulation of several developmental processes, including neuronal differentiation. The aim of this study was to elucidate the molecular mechanisms that regulate the transcription repression activity of Hes1. It is shown here that Hes1 associates with the nuclear matrix, the ribonucleoprotein network of the nucleus that plays important roles in transcriptional regulation. Nuclear matrix binding is mediated by the same Hes1 C-terminal domain that is also required for transcriptional repression. This domain contains the WRPW motif that acts as a binding site for the transcriptional corepressor Groucho, which also localizes to the nuclear matrix. Both the nuclear matrix association and transcription repression activity of Hes1 are inhibited by deletion of the WRPW motif, indicating that Groucho acts as a transcriptional corepressor for Hes1. This corepressor role is not modulated by the Groucho-related gene product Grg5. In contrast, the Runt-related protein RUNX2, which localizes to the nuclear matrix and interacts with Groucho and Hes1, can inhibit both the Groucho.Hes1 interaction and the transcription repression ability of Hes1. Together, these observations suggest that transcriptional repression by Hes1 requires interactions with Groucho at the nuclear matrix and that RUNX proteins act as negative regulators of the repressive activity of Groucho.Hes1 complexes.


Subject(s)
DNA-Binding Proteins/metabolism , Fungal Proteins/metabolism , Gene Expression Regulation , Neoplasm Proteins , Nuclear Matrix/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Binding Sites , Cell Line , Core Binding Factor Alpha 1 Subunit , Core Binding Factor alpha Subunits , Genes, Reporter , Glutathione Transferase/genetics , Helix-Loop-Helix Motifs , Humans , Rats , Recombinant Proteins/metabolism
10.
J Cell Sci ; 113 ( Pt 12): 2221-31, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10825294

ABSTRACT

The Runt related transcription factors RUNX (AML/CBF(alpha)/PEBP2(alpha)) are key regulators of hematopoiesis and osteogenesis. Using co-transfection experiments with four natural promoters, including those of the osteocalcin (OC), multi drug resistance (MDR), Rous Sarcoma Virus long terminal repeat (LTR), and bone sialoprotein (BSP) genes, we show that each of these promoters responds differently to the forced expression of RUNX proteins. However, the three RUNX subtypes (i.e. AML1, AML2, and AML3) regulate each promoter in a similar manner. Although the OC promoter is activated in a C terminus dependent manner, the MDR, LTR and BSP promoters are repressed by three distinct mechanisms, either independent of or involving the AML C terminus, or requiring only the conserved C-terminal pentapeptide VWRPY. Using yeast two hybrid assays we find that the C terminus of AML1 interacts with a Groucho/TLE/R-esp repressor protein. Co-expression assays reveal that TLE proteins repress AML dependent activation of OC gene transcription. Western and northern blot analyses suggest that TLE expression is regulated reciprocally with the levels of OC gene expression during osteoblast differentiation. Digital immunofluorescence microscopy results show that TLE1 and TLE2 are both associated with the nuclear matrix, and that a significant subset of each colocalizes with AML transcription factors. This co-localization of TLE and AML proteins is lost upon removing the C terminus of AML family members. Our findings indicate that suppression of AML-dependent gene activation by TLE proteins involves functional interactions with the C terminus of AML at the nuclear matrix in situ. Our data are consistent with the concept that the C termini of AML proteins support activation or repression of cell-type specific genes depending on the regulatory organization of the target promoter and subnuclear localization.


Subject(s)
DNA-Binding Proteins/genetics , Nuclear Matrix/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors , Gene Expression Regulation , HeLa Cells , Humans , Mice , Organ Specificity/genetics , Promoter Regions, Genetic , Transcriptional Activation
11.
J Biol Chem ; 275(24): 18418-23, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10748198

ABSTRACT

Members of the hepatic nuclear factor 3 (HNF3) family, including HNF3alpha, HNF3beta, and HNF3gamma, play important roles in embryonic development, the establishment of tissue-specific gene expression, and the regulation of gene expression in differentiated tissues. We found, using the glutathione S-transferase pull-down method, that the transducin-like Enhancer of split (TLE) proteins, which are the human homologs of Drosophila Groucho, directly associate with HNF3beta. Conserved region II of HNF3beta (amino acids 361-388) is responsible for the interaction with TLE1. A mammalian two-hybrid assay was used to confirm that this interaction occurs in vivo. Overexpression of TLE1 in HepG2 and HeLa cells decreases transactivation mediated through the C-terminal domain of HNF3beta, and Grg5, a naturally occurring dominant negative form of Groucho/TLE, also increases the transcriptional activity of this region of HNF3. These results lead us to suggest that TLE proteins could influence the expression of mammalian genes regulated by HNF3.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors , Co-Repressor Proteins , DNA-Binding Proteins/genetics , HeLa Cells , Hepatocyte Nuclear Factor 3-beta , Humans , Molecular Sequence Data , Molecular Weight , Nuclear Proteins/genetics , Protein Binding , Recombinant Fusion Proteins/genetics , Transcription Factors/genetics , Transcriptional Activation
12.
Mech Dev ; 93(1-2): 105-15, 2000 May.
Article in English | MEDLINE | ID: mdl-10781944

ABSTRACT

Transducin-like Enhancer of split (TLE) 1 is a mammalian transcriptional corepressor homologous to Drosophila Groucho. In Drosophila, Groucho acts together with bHLH proteins of the Hairy/Enhancer of split (HES) family to negatively regulate neuronal differentiation. Loss of the functions of Groucho or HES proteins results in supernumerary central and peripheral neurons. This suggests that mammalian TLE/Groucho family members may also be involved in the regulation of neuronal differentiation. Consistent with this possibility, TLE1 is expressed in proliferating neural progenitor cells of the central nervous system, but its expression is transiently down-regulated in newly generated postmitotic neurons. Based on these observations, we investigated whether persistent TLE1 expression in postmitotic neurons would perturb the normal course of neuronal development. Transgenic mice were derived in which the human TLE1 gene is regulated by the promoter of the Talpha1 alpha-tubulin gene, which is exclusively expressed in postmitotic neurons. In these mice, constitutive expression of TLE1 inhibits neuronal development in the embryonic forebrain leading to increased apoptosis and neuronal loss in the ventral and dorsal telencephalon. These results provide the first direct evidence that TLE1 is an important negative regulator of postmitotic neuronal differentiation in the mammalian central nervous system.


Subject(s)
Nuclear Proteins/biosynthesis , Prosencephalon/embryology , Telencephalon/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors , Co-Repressor Proteins , DNA-Binding Proteins , Female , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons , Nuclear Proteins/genetics , Repressor Proteins , Tubulin/genetics
13.
J Biol Chem ; 275(1): 530-8, 2000 Jan 07.
Article in English | MEDLINE | ID: mdl-10617648

ABSTRACT

Drosophila Runt is the founding member of a family of related transcription factors involved in the regulation of a variety of cell-differentiation events in invertebrates and vertebrates. Runt-related proteins act as both transactivators and transcriptional repressors, suggesting that context-dependent mechanisms modulate their transcriptional properties. The aim of this study was to elucidate the molecular mechanisms that contribute to the regulation of the functions of the mammalian Runt-related protein, Cbfa1. Here we provide the first demonstration that Cbfa1 (as well as the related protein, Cbfa2/AML1) physically interacts with the basic helix loop helix transcription factor, HES-1, a mammalian counterpart of the Drosophila Hairy and Enhancer of split proteins. This interaction is mediated by the carboxyl-terminal domains of Cbfa1 and HES-1, but does not require their respective tetrapeptide motifs, WRPY and WRPW. Our studies also show that HES-1 can antagonize the binding of Cbfa1 to mammalian transcriptional corepressors of the Groucho family. Moreover, HES-1 can potentiate Cbfa1-mediated transactivation in transfected cells. Taken together, these findings implicate HES-1 in the transcriptional functions of Cbfa1 and suggest that the concerted activities of Groucho and HES proteins modulate the functions of mammalian Runt-related proteins.


Subject(s)
Helix-Loop-Helix Motifs , Homeodomain Proteins/metabolism , Neoplasm Proteins , Repressor Proteins , Transcription Factors/metabolism , Transcriptional Activation , Basic Helix-Loop-Helix Transcription Factors , Core Binding Factor Alpha 1 Subunit , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Recombinant Proteins/metabolism , Transcription Factor HES-1 , Transcription Factors/genetics
14.
Biochem J ; 337 ( Pt 1): 13-7, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-9854018

ABSTRACT

The yeast proteins TUP1 and SSN6 form a transcription repressor complex that is recruited to different promoters via pathway-specific DNA-binding proteins and regulates the expression of a variety of genes. TUP1 is functionally related to invertebrate and vertebrate transcriptional repressors of the Groucho/transducin-like Enhancer of split (TLE) family. The aim was to examine whether similar mechanisms underlie the transcription repression functions of TUP1 and Groucho/TLEs by determining whether TLE family members can interact with yeast SSN6 and mammalian SSN6-like proteins. It is shown in the present work that SSN6 binds to TLE1 and mediates transcriptional repression when expressed in mammalian cells. Moreover, TLE1 and TLE2 interact with two mammalian proteins related to SSN6, designated as the products of the ubiquitously transcribed tetratricopeptide-repeat genes on the Y (or X) chromosomes (UTY/X). These findings suggest that mammalian TLE and UTY/X proteins may mediate repression mechanisms similar to those performed by TUP1-SSN6 in yeast.


Subject(s)
Biological Evolution , DNA-Binding Proteins , Fungal Proteins/metabolism , Nuclear Proteins , Proteins/metabolism , Repressor Proteins/metabolism , Saccharomyces cerevisiae Proteins , Xenopus Proteins , Basic Helix-Loop-Helix Transcription Factors , Cell Line , Fungal Proteins/genetics , Humans , Protein Binding , Repressor Proteins/genetics , Saccharomyces cerevisiae/genetics
15.
Proc Natl Acad Sci U S A ; 95(20): 11590-5, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751710

ABSTRACT

The mammalian AML/CBFalpha runt domain (RD) transcription factors regulate hematopoiesis and osteoblast differentiation. Like their Drosophila counterparts, most mammalian RD proteins terminate in a common pentapeptide, VWRPY, which serves to recruit the corepressor Groucho (Gro). Using a yeast two-hybrid assay, in vitro association and pull-down experiments, we demonstrate that Gro and its mammalian homolog TLE1 specifically interact with AML1 and AML2. In addition to the VWRPY motif, other C-terminal sequences are required for these interactions with Gro/TLE1. TLE1 inhibits AML1-dependent transactivation of the T cell receptor (TCR) enhancers alpha and beta, which contain functional AML binding sites, in transfected Jurkat T cells. LEF-1 is an additional transcription factor that mediates transactivation of TCR enhancers. LEF-1 and its Drosophila homolog Pangolin (Pan) are involved in the Wnt/Wg signaling pathway through interactions with the coactivator beta-catenin and its highly conserved fly homolog Armadillo (Arm). We show that TLE/Gro interacts with LEF-1 and Pan, and inhibits LEF-1:beta-catenin-dependent transcription. These data indicate that, in addition to their activity as transcriptional activators, AML1 and LEF-1 can act, through recruitment of the corepressor TLE1, as transcriptional repressors in TCR regulation and Wnt/Wg signaling.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins , Neoplasm Proteins , Nuclear Proteins/metabolism , Proto-Oncogene Proteins , Repressor Proteins/metabolism , Trans-Activators , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Armadillo Domain Proteins , Basic Helix-Loop-Helix Transcription Factors , Binding Sites , Cell Line , Co-Repressor Proteins , Core Binding Factor Alpha 2 Subunit , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila/genetics , Drosophila/metabolism , Genes, Reporter , Humans , Insect Proteins/genetics , Insect Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1 , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Protein Binding , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transfection , beta Catenin
16.
Mol Cell Biol ; 18(7): 4197-208, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9632804

ABSTRACT

Osf2/Cbfa1, hereafter called Osf2, is a member of the Runt-related family of transcription factors that plays a critical role during osteoblast differentiation. Like all Runt-related proteins, it contains a runt domain, which is the DNA-binding domain, and a C-terminal proline-serine-threonine-rich (PST) domain thought to be the transcription activation domain. Additionally, Osf2 has two amino-terminal domains distinct from any other Runt-related protein. To understand the mechanisms of osteoblast gene regulation by Osf2, we performed an extensive structure-function analysis. After defining a short Myc-related nuclear localization signal, a deletion analysis revealed the existence of three transcription activation domains and one repression domain. AD1 (for activation domain 1) comprises the first 19 amino acids of the molecule, which form the first domain unique to Osf2, AD2 is formed by the glutamine-alanine (QA) domain, the second domain unique to Osf2, and AD3 is located in the N-terminal half of the PST domain and also contains sequences unique to Osf2. The transcription repression domain comprises the C-terminal 154 amino acids of Osf2. DNA-binding, domain-swapping, and protein interaction experiments demonstrated that full-length Osf2 does not interact with Cbfbeta, a known partner of Runt-related proteins, whereas a deletion mutant of Osf2 containing only the runt and PST domains does. The QA domain appears to be responsible for preventing this heterodimerization. Thus, our results uncover the unique functional organization of Osf2 by identifying functional domains not shared with other Runt-related proteins that largely control its transactivation and heterodimerization abilities.


Subject(s)
DNA-Binding Proteins/metabolism , Neoplasm Proteins , Transcription Factors/metabolism , Transcriptional Activation , 3T3 Cells , Alanine , Amino Acid Sequence , Amino Acids , Animals , Binding Sites , COS Cells , Core Binding Factor Alpha 1 Subunit , DNA-Binding Proteins/genetics , Dimerization , Glutamine , Mice , Molecular Sequence Data , Nuclear Localization Signals , Proline , Proto-Oncogene Proteins c-myc/chemistry , Sequence Homology, Amino Acid , Serine , Structure-Activity Relationship , Threonine , Transcription Factor AP-2 , Transcription Factors/chemistry , Transcription Factors/genetics
17.
Dev Growth Differ ; 40(2): 133-46, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9572356

ABSTRACT

The Drosophila protein Groucho is involved in the regulation of cell-determination events during insect neurogenesis and segmentation. A group of mammalian proteins, referred to as transducin-like Enhancer of split (TLE) 1 through 4, share with Groucho identical structures and molecular properties. The aim was to determine whether individual TLE proteins participate in the regulation of cell determination in mammals like their Drosophila counterpart. It is here reported that TLE family members are expressed in combinatorial ways during the in vitro differentiation of mouse P19 embryonic carcinoma cells (a model for neural determination) and rat CFK2 cells (a model for chondrocytic determination). TLE1 is up-regulated and TLE2 and TLE4 are down-regulated to different extents during early stages of differentiation. In contrast, later stages correlate with up-regulation of TLE2 and TLE4, and decreased expression of TLE1. Individual TLE proteins are also expressed in combinatorial as well as complementary patterns during the development of the cerebral cortex and spinal cord of mouse embryos. In particular, TLE1 is robustly expressed in both neural progenitor cells and postmitotic neurons of the outer layers of the cortical plate, whereas TLE4 expression marks preferentially postmitotic neurons of the inner layers. Taken together, these results strongly suggest non-redundant roles for individual TLE proteins during both cell-determination and cell-differentiation events.


Subject(s)
DNA-Binding Proteins/physiology , Fetal Proteins/physiology , Gene Expression Regulation, Developmental , Mice/embryology , Nervous System/embryology , Neuropeptides/physiology , Repressor Proteins/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Embryonal/pathology , Cartilage/cytology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Drosophila melanogaster/genetics , Fetal Proteins/genetics , Glutathione Transferase/genetics , HeLa Cells , Humans , Jurkat Cells , Mice/genetics , Neuropeptides/genetics , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , Tretinoin/pharmacology , Tumor Cells, Cultured/drug effects
18.
Eur J Biochem ; 258(2): 339-49, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9874198

ABSTRACT

Groucho is a Drosophila transcriptional repressor involved in neurogenesis, segmentation, and sex determination together with basic helix-loop-helix proteins of the Hairy/Enhancer of split (HES) family. Several mammalian Groucho homologues, the Transducin-like Enhancer of split (TLE) 1 through 4 proteins, share similar properties with their Drosophila counterpart, suggesting that TLE proteins perform functions analogous to the roles of Groucho in Drosophila. The aim of this study was to examine this possibility by characterizing the properties of TLE2 and extending the analysis of TLE1. It is shown here that TLE2 and TLE1 are transcriptional repressors that contain two separate repression domains, located either within a Gln-rich amino terminal region or within an internal domain characterized by an abundance of Ser, Thr, and Pro residues. In addition, both TLE2 and TLE1 can homo- and heterodimerize through a short region that is part of their amino-terminal transcription repression domains. Finally, TLE2 interacts and is co-expressed with mammalian HES proteins in both neural and non-neural tissues. Taken together, these findings implicate TLE2 in transcriptional repression and define the structural elements that mediate transcriptional and protein-protein interaction functions of Groucho/TLE proteins.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila/metabolism , Nuclear Proteins/metabolism , Proteins/metabolism , Repressor Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Line , Co-Repressor Proteins , Dimerization , Embryo, Mammalian/cytology , Embryo, Nonmammalian , Gene Expression Regulation/genetics , Genes, Reporter/genetics , Helix-Loop-Helix Motifs/genetics , Immunohistochemistry , Mice , Nerve Tissue Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transcription, Genetic/genetics
19.
Mol Immunol ; 35(17): 1121-33, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10395201

ABSTRACT

In this study we investigated recognition by ovarian tumor associated lymphocyte (OVTAL), and breast tumor associated lymphocytes (BRTAL), of peptides corresponding to the sequence 125-135 of the Aminoenhancer of split (AES) protein. Three of these peptides designated as G75:AES1/2 (128-135), G60: AES1/2 (127-137) and G61: AES1/2 (125-133) correspond to the wildtype AES sequence, while the fourth G76:GPLTPLPV, AES1/2 (128-135) corresponds to a variant sequence of the peptide G75 with the N-terminal Leu substituted to glycine. These sequences were chosen for study because mass-spectrometric analysis (MS) of a CTL active HPLC peptide fraction eluted from immunoaffinity precipitated HLA-A2 molecule, revealed: (a) the presence of an ion with a mass-to-charge ratio (m/z) of 793 which was more abundant than other ions of similar masses; (b) the tentatively reconstituted sequence of the ion 793 matched the sequence of peptide G76. We found that AES peptides G75 (128-135) and G76 (128-135) (L128G) reconstituted CTL recognition at concentrations ranging between 200-500 nM. These concentrations are lower than concentrations reported to activate effector function of CTL recognizing other epithelial tumor Ag. Furthermore, analysis with cloned CD8+ T cells indicated that G75 and G76 were not cross-reactive specificities, suggesting a key role for the N-terminal residues of the variant peptide in dictating specificities. Since the AES proteins are part of a set of transcriptional repressors encoded by the Enhancer of split [E(spl)] genes, and since these repressors are activated to suppress cell differentiation in response to Notch receptors signalling, the AES peptides may represent a novel class of self-antigens that deserve further consideration as tumor Ag in epithelial cancers.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Membrane Proteins/immunology , Ovarian Neoplasms/immunology , Proteins , Repressor Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Co-Repressor Proteins , Epitopes , Female , Humans , Mass Spectrometry , Peptide Fragments/immunology , Receptors, Notch , Sequence Analysis
20.
J Biol Chem ; 272(42): 26604-10, 1997 Oct 17.
Article in English | MEDLINE | ID: mdl-9334241

ABSTRACT

Groucho is a transcriptional repressor implicated in Notch signaling and involved in neural development and segmentation in Drosophila. We are investigating the molecular mechanisms underlying the functions of Groucho and its mammalian homologs, the transducin-like Enhancer of split (TLE) proteins. We report that Groucho/TLEs are associated with chromatin in live cells and that they co-purify with isolated histones. Affinity chromatography and far Western blotting studies show further that native Groucho/TLE proteins interact specifically with histone H3 and not with other core histones. This interaction is mediated by the H3 amino-terminal domain previously shown by genetic analysis in yeast to be essential for the role of H3 in transcriptional silencing. We also demonstrate that Groucho/TLEs form oligomeric structures in vivo. These combined findings suggest that transcription complexes containing Groucho/TLEs may associate with chromatin through interactions with the amino terminus of histone H3 and that these interactions may be propagated along the chromosome due to the ability of Groucho/TLEs to participate in higher order structures.


Subject(s)
Histones/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Animals , Biopolymers , Cell Line , Chromatin/metabolism , Co-Repressor Proteins , Drosophila , HeLa Cells , Histones/genetics , Humans , Jurkat Cells , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...