Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters










Publication year range
1.
Oncogene ; 36(11): 1597-1606, 2017 03.
Article in English | MEDLINE | ID: mdl-27641335

ABSTRACT

The neural L1 transmembrane cell adhesion receptor of the immunoglobulin-like family is a target gene of Wnt-ß-catenin signaling in human colorectal cancer (CRC) cells and is expressed at the invasive edge of the tumor tissue. L1 overexpression in cultured CRC cells confers enhanced proliferation, motility and liver metastasis. We have analyzed the mechanisms of L1-mediated signaling in CRC cells by using various point mutations in the L1 ectodomain that are known to cause severe genetically inherited mental retardation disorders in patients. We found that all such L1 ectodomain mutations abolish the ability of L1 to confer metastatic properties in CRC cells. Using gene array analysis, we identified L1-mutation-specific gene expression signatures for the L1/H210Q and L1/D598N mutations. We identified CD10, a metalloprotease (neprilysin, neutral endopeptidase) and a gene that is specifically induced in CRC cells by L1 in an L1/H210Q mutation-specific manner. CD10 expression was required for the L1-mediated induction of cell proliferation, motility and metastasis, as suppression of CD10 levels in L1-expressing CRC cells abolished the L1 effects on CRC progression. The signaling from L1 to CD10 was mediated through the L1-ezrin-NF-κB pathway. In human CRC tissue L1 and CD10 were localized in partially overlapping regions in the more invasive areas of the tumor tissue. The results suggest that CD10 is a necessary component conferring the L1 effects in CRC cells. The identification of gene expression patterns of L1-domain-specific point mutations may provide novel markers and targets for interfering with L1-mediated CRC progression.


Subject(s)
Colorectal Neoplasms/genetics , Neprilysin/genetics , Neural Cell Adhesion Molecule L1/genetics , Point Mutation , Protein Interaction Domains and Motifs/genetics , Amino Acid Substitution , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Liver Neoplasms/secondary , Male , Mice , NF-kappa B/metabolism , Neoplasm Metastasis , Neprilysin/metabolism , Neural Cell Adhesion Molecule L1/chemistry , Neural Cell Adhesion Molecule L1/metabolism , Signal Transduction
2.
Oncogene ; 35(5): 549-57, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-25915847

ABSTRACT

Overactivation of Wnt-ß-catenin signaling, including ß-catenin-TCF target gene expression, is a hallmark of colorectal cancer (CRC) development. We identified the immunoglobulin family of cell-adhesion receptors member L1 as a ß-catenin-TCF target gene preferentially expressed at the invasive edge of human CRC tissue. L1 can confer enhanced motility and liver metastasis when expressed in CRC cells. This ability of L1-mediated metastasis is exerted by a mechanism involving ezrin and the activation of NF-κB target genes. In this study, we identified the secreted modular calcium-binding matricellular protein-2 (SMOC-2) as a gene activated by L1-ezrin-NF-κB signaling. SMOC-2 is also known as an intestinal stem cell signature gene in mice expressing Lgr5 in cells at the bottom of intestinal crypts. The induction of SMOC-2 expression in L1-expressing CRC cells was necessary for the increase in cell motility, proliferation under stress and liver metastasis conferred by L1. SMOC-2 expression induced a more mesenchymal like phenotype in CRC cells, a decrease in E-cadherin and an increase in Snail by signaling that involves integrin-linked kinase (ILK). SMOC-2 was localized at the bottom of normal human colonic crypts and at increased levels in CRC tissue with preferential expression in invasive areas of the tumor. We found an increase in Lgr5 levels in CRC cells overexpressing L1, p65 or SMOC-2, suggesting that L1-mediated CRC progression involves the acquisition of a stem cell-like phenotype, and that SMOC-2 elevation is necessary for L1-mediated induction of more aggressive/invasive CRC properties.


Subject(s)
Calcium-Binding Proteins/genetics , Colonic Neoplasms/genetics , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Animals , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/metabolism , Cell Growth Processes/genetics , Cell Line, Tumor , Cell Movement/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Heterografts , Humans , Intestines/pathology , Mice , Mice, Nude , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Stem Cells/pathology , Transcriptional Activation , Wnt Signaling Pathway
3.
Oncogene ; 32(27): 3220-30, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-22869145

ABSTRACT

L1, a neuronal cell adhesion receptor of the immunoglobulin-like protein family is expressed in invading colorectal cancer (CRC) cells as a target gene of Wnt/ß-catenin signaling. Overexpression of L1 in CRC cells enhances cell motility and proliferation, and confers liver metastasis. We recently identified ezrin and the IκB-NF-κB pathway as essential for the biological properties conferred by L1 in CRC cells. Here, we studied the underlying molecular mechanisms and found that L1 enhances ezrin phosphorylation, via Rho-associated protein kinase (ROCK), and is required for L1-ezrin co-localization at the juxtamembrane domain and for enhancing cell motility. Global transcriptomes from L1-expressing CRC cells were compared with transcriptomes from the same cells expressing small hairpin RNA (shRNA) to ezrin. Among the genes whose expression was elevated by L1 and ezrin we identified insulin-like growth factor-binding protein 2 (IGFBP-2) and showed that its increased expression is mediated by an NF-κB-mediated transactivation of the IGFBP-2 gene promoter. Expression of a constitutively activated mutant ezrin (Ezrin567D) could also increase IGFBP-2 levels in CRC cells. Overexpression of IGFBP-2 in CRC cells lacking L1-enhanced cell proliferation (in the absence of serum), cell motility, tumorigenesis and induced liver metastasis, similar to L1 overexpression. Suppression of endogenous IGFBP-2 in L1-transfected cells inhibited these properties conferred by L1. We detected IGFBP-2 in a unique organization at the bottom of human colonic crypts in normal mucosa and at increased levels throughout human CRC tissue samples co-localizing with the phosphorylated p65 subunit of NF-κB. Finally, we found that IGFBP-2 and L1 can form a molecular complex suggesting that L1-mediated signaling by the L1-ezrin-NF-κB pathway, that induces IGFBP-2 expression, has an important role in CRC progression.


Subject(s)
Colonic Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , NF-kappa B/metabolism , Neural Cell Adhesion Molecule L1/biosynthesis , Signal Transduction/physiology , Cell Line, Tumor , Chromatin Immunoprecipitation , Colonic Neoplasms/pathology , Disease Progression , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunoblotting , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 2/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neural Cell Adhesion Molecule L1/genetics , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
Mol Microbiol ; 41(3): 561-73, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11532125

ABSTRACT

Group A streptococcus (GAS) induces its own entry into eukaryotic cells in vitro and in vivo. Fibronectin (Fn) bound to protein F1, a GAS surface protein, acts as a bridge connecting the bacterium to host cell integrins. This triggers clustering of integrins, which acquire a polar pattern of distribution similar to that of protein F1 on the GAS surface. A unique and transient adhesion complex is formed at the site of GAS entry, which does not contain alpha-actinin. Vinculin is recruited to the site of GAS entry but is not required for uptake. The invading GAS recruits focal adhesion kinase (FAK), which is required for uptake and is tyrosine phosphorylated. The Src kinases, Src, Yes and Fyn, enhance the efficiency of GAS uptake but are not absolutely required for GAS entry. In addition, Rac and Cdc42, but not Rho, are required for the entry process. We suggest a model in which integrin engagement by Fn-occupied protein F1 triggers two independent signalling pathways. One is initiated by FAK recruitment and tyrosine phosphorylation, whereas the other is initiated by the recruitment and activation of Rac. The two pathways subsequently converge to trigger actin rearrangement leading to bacterial uptake.


Subject(s)
Adhesins, Bacterial/metabolism , Integrins/metabolism , Streptococcus/metabolism , Actinin/metabolism , Actins/metabolism , Animals , Bacterial Adhesion , Cell Adhesion Molecules/metabolism , Cell Line , Dogs , Endocytosis , Enzyme Activation , Fibronectins/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , HeLa Cells , Humans , Models, Biological , Monomeric GTP-Binding Proteins/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Protein Binding , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , Vinculin/metabolism
5.
EMBO J ; 20(17): 4912-22, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11532955

ABSTRACT

Aberrant activation of beta-catenin contributes to the onset of a variety of tumors. We report that a tumor-derived beta-catenin mutant induces accumulation and activation of the p53 tumor suppressor protein. Induction is mediated through ARF, an alternative reading frame product of the INK4A tumor suppressor locus, in a manner partially dependent on the transcription factor E2F1. In wild-type mouse embryo fibroblasts, mutant beta-catenin inhibits cell proliferation and imposes a senescence-like phenotype. This does not occur in cells lacking either ARF or p53, where deregulated beta-catenin actually overrides density-dependent growth inhibition and cooperates with activated Ras in transformation. Thus, the oncogenic activity of deregulated beta-catenin is curtailed by concurrent activation of the p53 pathway, thereby providing a protective mechanism against cancer. When the p53 pathway is impaired, deregulated beta-catenin is free to manifest its oncogenic features. This can occur not only by p53 mutations, but also by ablation of ARF expression, as observed frequently in early stages of colorectal carcinogenesis.


Subject(s)
ADP-Ribosylation Factors/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cytoskeletal Proteins/genetics , DNA-Binding Proteins , Genes, ras , Trans-Activators , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , ADP-Ribosylation Factors/deficiency , ADP-Ribosylation Factors/genetics , Animals , Cell Transformation, Neoplastic/genetics , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cytoskeletal Proteins/metabolism , E2F Transcription Factors , E2F1 Transcription Factor , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Genes, Reporter , Genes, Tumor Suppressor , Genes, p53 , Luciferases/analysis , Mice , Mice, Knockout , Models, Biological , Mutation , Neoplasms/genetics , Recombinant Proteins/analysis , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection , Tumor Suppressor Protein p53/deficiency , beta Catenin
6.
Mol Cell Biol ; 21(20): 6768-81, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11564862

ABSTRACT

beta-Catenin is a cytoplasmic protein that participates in the assembly of cell-cell adherens junctions by binding cadherins to the actin cytoskeleton. In addition, it is a key component of the Wnt signaling pathway. Activation of this pathway triggers the accumulation of beta-catenin in the nucleus, where it activates the transcription of target genes. Abnormal accumulation of beta-catenin is characteristic of various types of cancer and is caused by mutations either in the adenomatous polyposis coli protein, which regulates beta-catenin degradation, or in the beta-catenin molecule itself. Aberrant accumulation of beta-catenin in tumors is often associated with mutational inactivation of the p53 tumor suppressor. Here we show that overexpression of wild-type p53, by either transfection or DNA damage, down-regulates beta-catenin in human and mouse cells. This effect was not obtained with transcriptionally inactive p53, including a common tumor-associated p53 mutant. The reduction in beta-catenin level was accompanied by inhibition of its transactivation potential. The inhibitory effect of p53 on beta-catenin is apparently mediated by the ubiquitin-proteasome system and requires an active glycogen synthase kinase 3beta (GSK3beta). Mutations in the N terminus of beta-catenin which compromise its degradation by the proteasomes, overexpression of dominant-negative DeltaF-beta-TrCP, or inhibition of GSKbeta activity all rendered beta-catenin resistant to down-regulation by p53. These findings support the notion that there will be a selective pressure for the loss of wild-type p53 expression in cancers that are driven by excessive accumulation of beta-catenin.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Down-Regulation , Trans-Activators , Tumor Suppressor Protein p53/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line , Cell Nucleus/metabolism , Cisplatin/pharmacology , Cysteine Endopeptidases/metabolism , Cytoskeletal Proteins/genetics , Cytoskeleton/metabolism , Doxorubicin/pharmacology , Fibroblasts/metabolism , Genes, Dominant , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Luciferases/metabolism , Mice , Microscopy, Fluorescence , Models, Genetic , Multienzyme Complexes/metabolism , Mutation , Plasmids/metabolism , Proteasome Endopeptidase Complex , Time Factors , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Ubiquitins/metabolism , beta Catenin
7.
Am J Pathol ; 159(1): 43-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11438452

ABSTRACT

Plakoglobin and its homologue beta-catenin are cytoplasmic proteins that mediate adhesive functions by interacting with cadherin receptors and signaling activities by interacting with transcription factors. It has been suggested that plakoglobin can suppress tumorigenicity whereas beta-catenin can act as an oncogene. We investigated the correlation between the expression pattern of N-cadherin, beta-catenin, and plakoglobin and tumor behavior in primary tumors of 20 neuroblastoma patients of all stages and in 11 human neuroblastoma cell lines. N-cadherin and beta-catenin were detected in 9 of 11 and 11 of 11 cell lines, respectively, whereas plakoglobin was undetectable or severely reduced in 6 of 11 cell lines. Tumor cells from 16 of 20 patients expressed N-cadherin and 20 of 20 patients expressed beta-catenin at levels similar to those of normal ganglion cells. Plakoglobin was undetectable in 9 of 20 tumors. Plakoglobin deficiency in the primary tumors was significantly associated with adverse clinical outcome. Five of the patients with plakoglobin-negative tumors died whereas four patients are alive without evident disease. In contrast, all patients with plakoglobin-positive tumors are alive; 2 of 11 are alive with the disease and 9 of 11 are alive without evident disease. These results suggest that down-regulation of plakoglobin may be of prognostic value for neuroblastoma patients as predictor of poor outcome.


Subject(s)
Cytoskeletal Proteins/metabolism , Neuroblastoma/metabolism , Trans-Activators , Blotting, Western , Cadherins/metabolism , Child , Child, Preschool , Cytoskeletal Proteins/deficiency , Desmoplakins , Humans , Immunohistochemistry , Infant , Infant, Newborn , Neoplasm Staging , Neuroblastoma/mortality , Neuroblastoma/pathology , Survival Analysis , Tumor Cells, Cultured , beta Catenin , gamma Catenin
8.
Mol Biol Cell ; 12(4): 1177-88, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11294915

ABSTRACT

Drosophila Armadillo and its mammalian homologue beta-catenin are scaffolding proteins involved in the assembly of multiprotein complexes with diverse biological roles. They mediate adherens junction assembly, thus determining tissue architecture, and also transduce Wnt/Wingless intercellular signals, which regulate embryonic cell fates and, if inappropriately activated, contribute to tumorigenesis. To learn more about Armadillo/beta-catenin's scaffolding function, we examined in detail its interaction with one of its protein targets, cadherin. We utilized two assay systems: the yeast two-hybrid system to study cadherin binding in the absence of Armadillo/beta-catenin's other protein partners, and mammalian cells where interactions were assessed in their presence. We found that segments of the cadherin cytoplasmic tail as small as 23 amino acids bind Armadillo or beta-catenin in yeast, whereas a slightly longer region is required for binding in mammalian cells. We used mutagenesis to identify critical amino acids required for cadherin interaction with Armadillo/beta-catenin. Expression of such short cadherin sequences in mammalian cells did not affect adherens junctions but effectively inhibited beta-catenin-mediated signaling. This suggests that the interaction between beta-catenin and T cell factor family transcription factors is a sensitive target for disruption, making the use of analogues of these cadherin derivatives a potentially useful means to suppress tumor progression.


Subject(s)
Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Drosophila Proteins , Insect Proteins/metabolism , Signal Transduction/physiology , Trans-Activators , Amino Acid Sequence , Animals , Armadillo Domain Proteins , Binding Sites , CHO Cells , Cadherins/genetics , Cell Line , Cell Line, Transformed , Cricetinae , Cytoskeletal Proteins/genetics , Dogs , Drosophila , Humans , Insect Proteins/genetics , Mammals , Mice , Molecular Sequence Data , Saccharomyces cerevisiae , Transcription Factors , beta Catenin
9.
J Cell Sci ; 114(Pt 7): 1309-19, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11256997

ABSTRACT

Cadherin-mediated cell adhesion is involved in muscle differentiation from early stages of myogenic induction to late stages of myoblast interaction and fusion. beta-Catenin is a major constituent of cadherin-based adherens junctions and also serves as a signal transduction molecule that regulates gene expression during development. In this study, we explored the involvement of beta-catenin in myogenic differentiation. We show here that shortly after a switch from growth to differentiation medium, beta-catenin translocates to cell-cell junctions and its levels increase. We further show that elevation of beta-catenin levels, induced either by inhibition of its breakdown, using LiCl, or by its overexpression, suppresses the formation of adherens junctions, resulting in a sharp decline in myogenin expression and an arrest of myogenic progression. Recruitment of beta-catenin to adherens junctions after transfection with N-cadherin restores myogenin expression in the transfected cells. These results suggest that increased cadherin-mediated adhesion and translocation of beta-catenin to adherens junctions are involved in activating the early steps of myogenic differentiation.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Muscle, Skeletal/physiology , Trans-Activators , Animals , Cadherins/genetics , Cell Adhesion/physiology , Cell Differentiation , Cell Line , Culture Media , Cytoskeletal Proteins/genetics , Gene Expression , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myogenin/genetics , Rats , beta Catenin
10.
Int J Surg Pathol ; 9(4): 273-9, 2001 Oct.
Article in English | MEDLINE | ID: mdl-12574842

ABSTRACT

Beta-catenin is a cytoskeleton-associated signaling molecule shown to be elevated in various carcinomas but mostly in colon cancer owing to its impaired degradation. In contrast, its close homologue plakoglobin was shown to suppress the tumorigenicity of certain tumor cells. In the present study, we have used a semiquantitative immunohistochemical approach to evaluate the extent of nuclear localization of beta-catenin in human colonic adenocarcinomas and adenomas and compared it to the distribution of plakoglobin in the same tissues. We show that beta-catenin accumulates in the nuclei of the epithelium of primary and metastatic colonic adenocarcinoma as well as in colonic adenomas. In contrast, nuclear plakoglobin levels in these tissues were low, even compared to those found in epithelial cells of normal colon. These results support the view that the increase in beta-catenin levels in colon cancer cells occurs early in the tumorigenic process, leading to its nuclear localization, not only in invasive adenocarcinoma, but also in colonic adenoma with mild dysplasia.


Subject(s)
Adenocarcinoma/metabolism , Adenoma/metabolism , Colonic Neoplasms/metabolism , Cytoskeletal Proteins/metabolism , Trans-Activators/metabolism , Cell Nucleus/metabolism , Colon/metabolism , Cytoplasm/metabolism , Desmoplakins , Epithelium/metabolism , Humans , Immunohistochemistry , Liver Neoplasms/secondary , Lymphatic Metastasis , Protein Transport/physiology , beta Catenin , gamma Catenin
12.
J Cell Sci ; 113 ( Pt 18): 3127-39, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954412

ABSTRACT

Beta-catenin can play different roles in the cell, including one as a structural protein at cell-cell adherens junctions and another as a transcriptional activator mediating Wnt signal transduction. Plakoglobin (gamma)-catenin), a close homolog of beta-catenin, shares with beta-catenin common protein partners and can fulfill some of the same functions. The complexing of catenins with various protein partners is regulated by phosphorylation and by intramolecular interactions. The competition between different catenin partners for binding to catenins mediates the cross-talk between cadherin-based adhesion, catenin-dependent transcription and Wnt signaling. Although plakoglobin differs from beta-catenin in its functions and is unable to compensate for defects in Wnt signaling resulting from lack of beta-catenin, recent evidence suggests that plakoglobin plays a unique role in Wnt signaling that is different from that of beta-catenin. The functional difference between catenins is reflected in their differential involvement in embryonic development and cancer progression.


Subject(s)
Cell Adhesion Molecules/physiology , Cytoskeletal Proteins/physiology , Trans-Activators , Zebrafish Proteins , Animals , Binding Sites , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/metabolism , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/metabolism , Desmoplakins , Humans , Neoplasms , Protein Conformation , Proto-Oncogene Proteins/metabolism , Wnt Proteins , beta Catenin , gamma Catenin
13.
J Biol Chem ; 275(42): 32649-57, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10915780

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that phosphorylates and inactivates the pRB tumor suppressor protein. Cyclin D1 is overexpressed in 20-30% of human breast tumors and is induced both by oncogenes including those for Ras, Neu, and Src, and by the beta-catenin/lymphoid enhancer factor (LEF)/T cell factor (TCF) pathway. The ankyrin repeat containing serine-threonine protein kinase, integrin-linked kinase (ILK), binds to the cytoplasmic domain of beta(1) and beta(3) integrin subunits and promotes anchorage-independent growth. We show here that ILK overexpression elevates cyclin D1 protein levels and directly induces the cyclin D1 gene in mammary epithelial cells. ILK activation of the cyclin D1 promoter was abolished by point mutation of a cAMP-responsive element-binding protein (CREB)/ATF-2 binding site at nucleotide -54 in the cyclin D1 promoter, and by overexpression of either glycogen synthase kinase-3beta (GSK-3beta) or dominant negative mutants of CREB or ATF-2. Inhibition of the PI 3-kinase and AKT/protein kinase B, but not of the p38, ERK, or JNK signaling pathways, reduced ILK induction of cyclin D1 expression. ILK induced CREB transactivation and CREB binding to the cyclin D1 promoter CRE. Wnt-1 overexpression in mammary epithelial cells induced cyclin D1 mRNA and targeted overexpression of Wnt-1 in the mammary gland of transgenic mice increased both ILK activity and cyclin D1 levels. We conclude that the cyclin D1 gene is regulated by the Wnt-1 and ILK signaling pathways and that ILK induction of cyclin D1 involves the CREB signaling pathway in mammary epithelial cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin D1/genetics , Gene Expression Regulation , Protein Serine-Threonine Kinases/metabolism , Zebrafish Proteins , Activating Transcription Factor 2 , Animals , Breast Neoplasms , CD18 Antigens/physiology , Cell Line , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Integrin beta1/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Subunits , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Wnt Proteins , Wnt1 Protein
14.
Mol Cell Biol ; 20(12): 4238-52, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10825188

ABSTRACT

beta-Catenin and plakoglobin are highly homologous components of cell-cell adherens junctions linking cadherin receptors to the actin cytoskeleton. beta-Catenin, in addition, activates transcription by forming a complex with LEF/TCF family transcription factors in the nucleus. Plakoglobin can also bind to LEF-1 and, when overexpressed in mammalian cells, enhances LEF-1-directed transcription. Plakoglobin overexpression, however, results in the elevation and nuclear translocation of endogenous beta-catenin. We show here, by DNA mobility shift analysis, that the formation of a plakoglobin-LEF/TCF-DNA complex in vitro is very inefficient compared to a complex containing beta-catenin-LEF-DNA. Moreover, in plakoglobin-transfected cells plakoglobin-LEF/TCF-DNA complexes were not formed; rather, the endogenous beta-catenin, whose level is elevated by plakoglobin transfection, formed a beta-catenin-LEF-DNA complex. Removal of the N- and C-terminal domains of both beta-catenin and plakoglobin (leaving the armadillo repeat domain intact) induced plakoglobin-LEF-DNA complex formation and also enhanced beta-catenin-LEF-DNA complexing, both with in vitro-translated components and in transfected cells. Transfection with these truncated catenins increased endogenous beta-catenin levels, but the truncated catenins acted as dominant-negative inhibitors of beta-catenin-driven transcription by forming transcriptionally inactive complexes with LEF-1. When these catenin mutants were prevented from entering the nucleus, by their fusion to the connexin transmembrane domain, they indirectly activated transcription by increasing endogenous beta-catenin levels. These results suggest that overexpression of plakoglobin does not directly activate transcription and that formation of catenin-LEF-DNA complexes is negatively regulated by the catenin N- and C-terminal domains.


Subject(s)
Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Trans-Activators , Transcription Factors/genetics , Transcriptional Activation , Biological Transport/genetics , Cell Line , Desmoplakins , Humans , Mutation , beta Catenin , gamma Catenin
15.
Oncogene ; 19(16): 1992-2001, 2000 Apr 13.
Article in English | MEDLINE | ID: mdl-10803460

ABSTRACT

Beta-catenin and plakoglobin are closely related armadillo family proteins with shared and distinct properties; Both are associated with cadherins in actin-containing adherens junctions. Plakoglobin is also found in desmosomes where it anchors intermediate filaments to the desmosomal plaques. Beta-catenin, on the other hand, is a component of the Wnt signaling pathway, which is involved in embryonic morphogenesis and tumorigenesis. A key step in the regulation of this pathway involves modulation of beta-catenin stability. A multiprotein complex, regulated by Wnt, directs the phosphorylation of beta-catenin and its degradation by the ubiquitin-proteasome system. Plakoglobin can also associate with members of this complex, but inhibition of proteasomal degradation has little effect on its levels while dramatically increasing the levels of beta-catenin. Beta-TrCP, an F-box protein of the SCF E3 ubiquitin ligase complex, was recently shown to play a role in the turnover of beta-catenin. To elucidate the basis for the apparent differences in the turnover of beta-catenin and plakoglobin we compared the handling of these two proteins by the ubiquitin-proteasome system. We show here that a deletion mutant of beta-TrCP, lacking the F-box, can stabilize the endogenous beta-catenin leading to its nuclear translocation and induction of beta-catenin/LEF-1-directed transcription, without affecting the levels of plakoglobin. However, when plakoglobin was overexpressed, it readily associated with beta-TrCP, efficiently competed with beta-catenin for binding to beta-TrCP and became polyubiquitinated. Fractionation studies revealed that about 85% of plakoglobin in 293 cells, is Triton X-100-insoluble compared to 50% of beta-catenin. These results suggest that while both plakoglobin and beta-catenin can comparably interact with beta-TrCP and the ubiquitination system, the sequestration of plakoglobin by the membrane-cytoskeleton system renders it inaccessible to the proteolytic machinery and stabilizes it.


Subject(s)
Cysteine Endopeptidases/metabolism , Cytoskeletal Proteins/metabolism , Multienzyme Complexes/metabolism , Trans-Activators , Ubiquitins/metabolism , Animals , Biological Transport , CHO Cells/metabolism , Cell Compartmentation , Cricetinae , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Desmoplakins , Dexamethasone/pharmacology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1 , Octoxynol/chemistry , Proteasome Endopeptidase Complex , Recombinant Proteins/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin , beta-Transducin Repeat-Containing Proteins , gamma Catenin
16.
J Biol Chem ; 275(30): 23368-77, 2000 Jul 28.
Article in English | MEDLINE | ID: mdl-10816572

ABSTRACT

Caveolin-1 is a principal component of caveolae membranes. In NIH 3T3 cells, caveolin-1 expression is dramatically up-regulated in confluent cells and localizes at areas of cell-cell contact. However, it remains unknown whether caveolin-1 is involved in cell-cell signaling. Here, we examine the potential role of caveolin-1 in regulating beta-catenin signaling. beta-Catenin plays a dual role in the cell, linking E-cadherin to the actin cytoskeleton and in Wnt signaling by forming a complex with members of the lymphoid enhancing factor (Lef-1) family of transcription factors. We show that E-cadherin, beta-catenin, and gamma-catenin (plakoglobin) are all concentrated in caveolae membranes. Moreover, we demonstrate that activation of beta-catenin/Lef-1 signaling by Wnt-1 or by overexpression of beta-catenin itself is inhibited by caveolin-1 expression. We also show that recombinant expression of caveolin-1 in caveolin-1 negative cells is sufficient to recruit beta-catenin to caveolae membranes, thereby blocking beta-catenin-mediated transactivation. These results suggest that caveolin-1 expression can modulate Wnt/beta-catenin/Lef-1 signaling by regulating the intracellular localization of beta-catenin.


Subject(s)
Caveolins , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators , Transcription Factors/metabolism , Zebrafish Proteins , 3T3 Cells , Animals , Caveolin 1 , Cell Membrane/metabolism , Lymphoid Enhancer-Binding Factor 1 , Mice , Transcriptional Activation , Wnt Proteins , Wnt1 Protein , beta Catenin
17.
J Biol Chem ; 275(28): 21203-9, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10747899

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of the holoenzyme that phosphorylates and inactivates the retinoblastoma pRB protein. Cyclin D1 protein levels are elevated by mitogenic and oncogenic signaling pathways, and antisense mRNA to cyclin D1 inhibits transformation by the ras, neu, and src oncogenes, thus linking cyclin D1 regulation to cellular transformation. Caveolins are the principal protein components of caveolae, vesicular plasma membrane invaginations that also function in signal transduction. We show here that caveolin-1 expression levels inversely correlate with cyclin D1 abundance levels in transformed cells. Expression of antisense caveolin-1 increased cyclin D1 levels, whereas caveolin-1 overexpression inhibited expression of the cyclin D1 gene. Cyclin D1 promoter activity was selectively repressed by caveolin-1, but not by caveolin-3, and this repression required the caveolin-1 N terminus. Maximal inhibition of the cyclin D1 gene promoter by caveolin-1 was dependent on the cyclin D1 promoter T-cell factor/lymphoid enhancer factor-1-binding site between -81 to -73. The T-cell factor/lymphoid enhancer factor sequence was sufficient for repression by caveolin-1. We suggest that transcriptional repression of the cyclin D1 gene may contribute to the inhibition of transformation by caveolin-1.


Subject(s)
Caveolins , Cyclin D1/genetics , Gene Expression Regulation , Membrane Proteins/physiology , Promoter Regions, Genetic , Transcription, Genetic , Amino Acid Sequence , Animals , CHO Cells , Caveolin 1 , Cell Membrane/physiology , Cricetinae , Culture Media, Serum-Free , DNA-Binding Proteins/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1 , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Transcription Factors/metabolism , Transfection
18.
J Cell Biochem ; 76(1): 1-12, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10580995

ABSTRACT

Monomeric (G) actin was shown to be involved in inhibiting its own synthesis by an autoregulatory mechanism that includes enhanced degradation of the actin mRNA [Bershadsky et al., 1995; Lyubimova et al., 1997]. We show that the 3'-untranslated region (3'-UTR) of beta-actin mRNA, but not its 5'-untranslated region, is important for this regulation. The level of full-length beta-actin mRNA in cells was reduced when actin filaments were depolymerized by treatment with latrunculin A and elevated when actin polymerization was induced by jasplakinolide. By contrast, the level of actin mRNA lacking the 3'-UTR remained unchanged when these drugs modulated the dynamics of actin assembly in the cell. Moreover, the transfection of cells with a construct encoding the autoregulation-deficient form of beta-actin mRNA led to very high levels of actin expression compared with transfection with the control actin construct and was accompanied by characteristic changes in cell morphology and the structure of the actin cytoskeleton. These results suggest that the autoregulatory mechanism working via the 3'-UTR of actin mRNA is involved in controlling the maintenance of a defined pool of actin monomers that could be necessary for the proper organization of the microfilament system and the cytoskeleton-mediated signaling.


Subject(s)
3' Untranslated Regions , Actins/genetics , Depsipeptides , Gene Expression Regulation , RNA, Messenger/genetics , 3T3 Cells , Actins/biosynthesis , Animals , Base Sequence , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line , DNA Primers , Gene Expression Regulation/drug effects , Mice , Mice, Inbred BALB C , Peptides, Cyclic/pharmacology , Sequence Deletion , Thiazoles/pharmacology , Thiazolidines
19.
EMBO J ; 18(11): 3054-63, 1999 Jun 01.
Article in English | MEDLINE | ID: mdl-10357817

ABSTRACT

beta-catenin is a multifunctional protein, acting both as a structural component of the cell adhesion machinery and as a transducer of extracellular signals. Deregulated beta-catenin protein expression, due to mutations in the beta-catenin gene itself or in its upstream regulator, the adenomatous polyposis coli (APC) gene, is prevalent in colorectal cancer and in several other tumor types, and attests to the potential oncogenic activity of this protein. Increased expression of beta-catenin is an early event in colorectal carcinogenesis, and is usually followed by a later mutational inactivation of the p53 tumor suppressor. To examine whether these two key steps in carcinogenesis are interrelated, we studied the effect of excess beta-catenin on p53. We report here that overexpression of beta-catenin results in accumulation of p53, apparently through interference with its proteolytic degradation. This effect involves both Mdm2-dependent and -independent p53 degradation pathways, and is accompanied by augmented transcriptional activity of p53 in the affected cells. Increased p53 activity may provide a safeguard against oncogenic deregulation of beta-catenin, and thus impose a pressure for mutational inactivation of p53 during the later stages of tumor progression.


Subject(s)
Cytoskeletal Proteins/metabolism , Gene Expression , Nuclear Proteins , Trans-Activators , Transcription, Genetic/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cell Nucleus/metabolism , Colorectal Neoplasms/etiology , Colorectal Neoplasms/genetics , Cysteine Proteinase Inhibitors/pharmacology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , Desmoplakins , Dishevelled Proteins , Humans , Leupeptins/pharmacology , Mice , Mutation , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-mdm2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Solubility , Transfection , Tumor Suppressor Protein p53/genetics , Up-Regulation/drug effects , beta Catenin
20.
Proc Natl Acad Sci U S A ; 96(10): 5522-7, 1999 May 11.
Article in English | MEDLINE | ID: mdl-10318916

ABSTRACT

beta-Catenin plays a dual role in the cell: one in linking the cytoplasmic side of cadherin-mediated cell-cell contacts to the actin cytoskeleton and an additional role in signaling that involves transactivation in complex with transcription factors of the lymphoid enhancing factor (LEF-1) family. Elevated beta-catenin levels in colorectal cancer caused by mutations in beta-catenin or by the adenomatous polyposis coli molecule, which regulates beta-catenin degradation, result in the binding of beta-catenin to LEF-1 and increased transcriptional activation of mostly unknown target genes. Here, we show that the cyclin D1 gene is a direct target for transactivation by the beta-catenin/LEF-1 pathway through a LEF-1 binding site in the cyclin D1 promoter. Inhibitors of beta-catenin activation, wild-type adenomatous polyposis coli, axin, and the cytoplasmic tail of cadherin suppressed cyclin D1 promoter activity in colon cancer cells. Cyclin D1 protein levels were induced by beta-catenin overexpression and reduced in cells overexpressing the cadherin cytoplasmic domain. Increased beta-catenin levels may thus promote neoplastic conversion by triggering cyclin D1 gene expression and, consequently, uncontrolled progression into the cell cycle.


Subject(s)
Cyclin D1/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Repressor Proteins , Trans-Activators , Transcription Factors/metabolism , Adenomatous Polyposis Coli Protein , Axin Protein , Binding Sites , Cadherins/metabolism , Colonic Neoplasms/genetics , Cyclin D1/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lymphoid Enhancer-Binding Factor 1 , Promoter Regions, Genetic , Proteins , Signal Transduction , Transcriptional Activation , Transfection , Tumor Cells, Cultured , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...