Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Handb Exp Pharmacol ; (186): 261-84, 2008.
Article in English | MEDLINE | ID: mdl-18491056

ABSTRACT

The Wnt/beta-catenin signaling pathway plays diverse roles in embryonic development and in maintenance of organs and tissues in adults. Activation of this signaling cascade inhibits degradation of the pivotal component beta-catenin, which in turn stimulates transcription of downstream target genes. Over the past two decades, intensive worldwide investigations have yielded considerable progress toward understanding the cellular and molecular mechanisms of Wnt signaling and its involvement in the pathogenesis of a range of human diseases. Remarkably, beta-catenin signaling is aberrantly activated in greater than 70% of colorectal cancers and to a lesser extent in other tumor types, promoting cancer cell proliferation, survival and migration. Accordingly, beta-catenin has gained recognition as an enticing molecular target for cancer therapeutics. Disruption of protein-protein interactions essential for beta-catenin activity holds immense promise for the development of novel anti-cancer drugs. In this review, we focus on the regulation of beta-catenin-dependent transcriptional activation and discuss potential therapeutic opportunities to block this signaling pathway in cancer.


Subject(s)
Drug Delivery Systems , Wnt Proteins/drug effects , beta Catenin/drug effects , Animals , Antineoplastic Agents/pharmacology , Cell Nucleus/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/physiopathology , Signal Transduction , Transcriptional Activation/physiology , Wnt Proteins/metabolism , beta Catenin/metabolism
2.
J Cell Biol ; 149(2): 249-54, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10769018

ABSTRACT

Beta-catenin plays a pivotal role in the transcriptional activation of Wnt-responsive genes by binding to TCF/LEF transcription factors. Although it has been suggested that the COOH-terminal region of beta-catenin functions as an activation domain, the mechanisms of activation remain unclear. To screen for potential transcriptional coactivators that bind to the COOH-terminal region of beta-catenin, we used a novel yeast two-hybrid system, the Ras recruitment system (RRS) that detects protein-protein interactions at the inner surface of the plasma membrane. Using this system, we isolated the CREB-binding protein (CBP). Armadillo (Arm) repeat 10 to the COOH terminus of beta-catenin is involved in binding to CBP, whereas beta-catenin interacts directly with the CREB-binding domain of CBP. Beta-catenin synergizes with CBP to stimulate the activity of a synthetic reporter in vivo. Conversely, beta-catenin-dependent transcriptional activation is repressed by E1A, an antagonist of CBP function, but not by an E1A mutant that does not bind to CBP. The activation of Wnt target genes such as siamois and Xnr3 in Xenopus embryos is also sensitive to E1A. These findings suggest that CBP provides a link between beta-catenin and the transcriptional machinery, and possibly mediates the oncogenic function of beta-catenin.


Subject(s)
Cytoskeletal Proteins/metabolism , Gene Expression Regulation , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Xenopus Proteins , Animals , Binding Sites , COS Cells , CREB-Binding Protein , Cadherins/physiology , Cell Line , Cloning, Molecular , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/physiology , HeLa Cells , Homeodomain Proteins/genetics , Humans , Luciferases/analysis , Saccharomyces cerevisiae , Transcriptional Activation , Transfection , Transforming Growth Factor beta/genetics , Xenopus laevis/embryology , beta Catenin
3.
Genes Cells ; 4(7): 415-24, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10469174

ABSTRACT

BACKGROUND: Multiprotein bridging factor 1 (MBF1) is a transcriptional coactivator necessary for transcriptional activation caused by DNA binding activators, such as FTZ-F1 and GCN4. MBF1 bridges the DNA-binding regions of these activators and the TATA-box binding protein (TBP), suggesting that MBF1 functions by recruiting TBP to promoters where the activators are bound. In addition, MBF1 stimulates DNA binding activities of the activators to their recognition sites. To date, little is known about structures of coactivators that bind to TBP. RESULTS: The two-dimensional (2D) 1H-15N correlation spectrum of 15N labeled MBF1 indicated that MBF1 consists of both flexible and well structured parts. Limited digestion of MBF1 by alpha-chymotrypsin yielded a approximately 9 kDa fragment. N-terminal sequence analysis and NMR measurements revealed that this fragment originates from the C-terminal 80 residues of MBF1 and form a well structured C-terminal domain of MBF1, MBF1CTD. As previous deletion analyses have shown that MBF1CTD is capable of binding to TBP, it is suggested that MBF1CTD is the TBP binding domain of MBF1. Sequential assignments have been obtained by means of three-dimensional (3D) and four dimensional (4D) heteronuclear correlation spectroscopies, and then the secondary structure of MBF1CTD was determined. As a result, MBF1CTD was shown to contain four amphipathic helices and a conserved C-terminal region. Asp106 which is assumed to be responsible for the binding to TBP is located at the hydrophilic side of the third helix. CONCLUSIONS: Structural analyses revealed that MBF1 consists of two structurally different domains. A N-terminal region is indispensable for the binding to activators, and does not form a well defined structure. In contrast, the C-terminal 80 residues, which is capable of binding to TBP by itself, form a well-structured domain, MBF1CTD. MBF1CTD is made up of four amphipathic helices and a conserved C-terminal tail. A putative TBP binding residue is located on the hydrophilic surface of the third helix.


Subject(s)
Bombyx/chemistry , Peptide Fragments/chemistry , Trans-Activators/chemistry , Amino Acid Sequence , Animals , Binding Sites , Bombyx/genetics , Chymotrypsin/metabolism , DNA-Binding Proteins/metabolism , Escherichia coli/genetics , Hydrogen Bonding , Hydrogen-Ion Concentration , Molecular Sequence Data , Molecular Weight , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Structure, Secondary , Protons , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Analysis , Structure-Activity Relationship , TATA-Box Binding Protein , Trans-Activators/genetics , Trans-Activators/isolation & purification , Trans-Activators/metabolism , Transcription Factors/metabolism
4.
Dev Biol ; 213(1): 170-9, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10452854

ABSTRACT

Cells in the presumptive neural ectoderm of Xenopus are committed to neural fate through a process called neural induction, which may involve proteins that antagonize BMP signaling pathways. To identify genes that are induced by the BMP antagonists and that may be involved in subsequent neural patterning, we used a suppression PCR-based subtraction screen. Here we investigate the prospective activities and functions of one of the genes, a nuclear orphan receptor previously described as xGCNF. In animal cap assays, xGCNF synergizes with ectopic chordin to induce the midbrain-hindbrain marker engrailed-2 (En-2). In Keller explants, which rely on endogenous factors for neural induction, similar increases in En-2 are observed. Expression in embryos of a dominant interfering form of xGCNF reduces the expression of endogenous En-2 and Krox-20. These gain-of-function and prospective loss-of-function experiments, taken with the observation that xGCNF is expressed in the early neural plate and is elevated in the prospective midbrain-hindbrain region, which subsequently expresses En-2, suggest that xGCNF may play a role in regulating En-2 and thus midbrain-hindbrain identity.


Subject(s)
DNA-Binding Proteins/physiology , Intercellular Signaling Peptides and Proteins , Mesencephalon/embryology , Receptors, Cytoplasmic and Nuclear/physiology , Rhombencephalon/embryology , Xenopus laevis/embryology , Animals , Base Sequence , Body Patterning/genetics , Body Patterning/physiology , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/physiology , DNA Primers/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Glycoproteins/genetics , Glycoproteins/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , In Situ Hybridization , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Nuclear Receptor Subfamily 6, Group A, Member 1 , RNA/administration & dosage , RNA/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Xenopus Proteins , Xenopus laevis/genetics
5.
Proc Natl Acad Sci U S A ; 94(14): 7251-6, 1997 Jul 08.
Article in English | MEDLINE | ID: mdl-9207077

ABSTRACT

Multiprotein bridging factor 1 (MBF1) is a transcriptional cofactor that bridges between the TATA box-binding protein (TBP) and the Drosophila melanogaster nuclear hormone receptor FTZ-F1 or its silkworm counterpart BmFTZ-F1. A cDNA clone encoding MBF1 was isolated from the silkworm Bombyx mori whose sequence predicts a basic protein consisting of 146 amino acids. Bacterially expressed recombinant MBF1 is functional in interactions with TBP and a positive cofactor MBF2. The recombinant MBF1 also makes a direct contact with FTZ-F1 through the C-terminal region of the FTZ-F1 DNA-binding domain and stimulates the FTZ-F1 binding to its recognition site. The central region of MBF1 (residues 35-113) is essential for the binding of FTZ-F1, MBF2, and TBP. When the recombinant MBF1 was added to a HeLa cell nuclear extract in the presence of MBF2 and FTZ622 bearing the FTZ-F1 DNA-binding domain, it supported selective transcriptional activation of the fushi tarazu gene as natural MBF1 did. Mutations disrupting the binding of FTZ622 to DNA or MBF1, or a MBF2 mutation disrupting the binding to MBF1, all abolished the selective activation of transcription. These results suggest that tethering of the positive cofactor MBF2 to a FTZ-F1-binding site through FTZ-F1 and MBF1 is essential for the binding site-dependent activation of transcription. A homology search in the databases revealed that the deduced amino acid sequence of MBF1 is conserved across species from yeast to human.


Subject(s)
Calmodulin-Binding Proteins , DNA-Binding Proteins/genetics , Drosophila Proteins , Saccharomyces cerevisiae Proteins , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics , Amino Acid Sequence , Animals , Base Sequence , Bombyx , Conserved Sequence , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Drosophila melanogaster , Evolution, Molecular , Fushi Tarazu Transcription Factors , HeLa Cells , Homeodomain Proteins , Humans , Insect Proteins , Molecular Sequence Data , Mutation , Receptors, Cytoplasmic and Nuclear , Steroidogenic Factor 1 , TATA-Box Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...