Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Biol Cell ; 33(14): ar135, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36222847

RESUMEN

The coatomer protein complex 1 (COPI) is a multisubunit complex that coats intracellular vesicles and is involved in intracellular protein trafficking. Recently we and others found that depletion of COPI complex subunits zeta (COPZ1) and delta (ARCN1) preferentially kills tumor cells relative to normal cells. Here we delineate the specific cellular effects and sequence of events of COPI complex depletion in tumor cells. We find that this depletion leads to the inhibition of mitochondrial oxidative phosphorylation and the elevation of reactive oxygen species (ROS) production, followed by accumulation of lipid droplets (LDs) and autophagy-associated proteins LC3-II and SQSTM1/p62 and, finally, apoptosis of the tumor cells. Inactivation of ROS in COPI-depleted cells with the mitochondrial-specific quencher, mitoquinone mesylate, attenuated apoptosis and markedly decreased both the size and the number of LDs. COPI depletion caused ROS-dependent accumulation of LC3-II and SQSTM1 which colocalizes with LDs. Lack of double-membrane autophagosomes and insensitivity to Atg5 deletion suggested an accumulation of a microlipophagy complex on the surface of LDs induced by depletion of the COPI complex. Our findings suggest a sequence of cellular events triggered by COPI depletion, starting with inhibition of oxidative phosphorylation, followed by ROS activation and accumulation of LDs and apoptosis.


Asunto(s)
Autofagia , Neoplasias , Especies Reactivas de Oxígeno , Apoptosis , Proteína Coat de Complejo I/metabolismo , Lípidos , Neoplasias/metabolismo
2.
Oncogene ; 35(31): 4058-68, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-26686093

RESUMEN

Arginylation is an emerging post-translational modification mediated by arginyltransferase (ATE1) that is essential for mammalian embryogenesis and regulation of the cytoskeleton. Here, we discovered that Ate1-knockout (KO) embryonic fibroblasts exhibit tumorigenic properties, including abnormally rapid contact-independent growth, reduced ability to form cell-cell contacts and chromosomal aberrations. Ate1-KO fibroblasts can form large colonies in Matrigel and exhibit invasive behavior, unlike wild-type fibroblasts. Furthermore, Ate1-KO cells form tumors in subcutaneous xenograft assays in immunocompromised mice. Abnormal growth in these cells can be partially rescued by reintroduction of stably expressed specific Ate1 isoforms, which also reduce the ability of these cells to form tumors. Tumor array studies and bioinformatics analysis show that Ate1 is downregulated in several types of human cancer samples at the protein level, and that its transcription level inversely correlates with metastatic progression and patient survival. We conclude that Ate1-KO results in carcinogenic transformation of cultured fibroblasts, suggesting that in addition to its previously known activities Ate1 gene is essential for tumor suppression and also likely participates in suppression of metastatic growth.


Asunto(s)
Aminoaciltransferasas/fisiología , Neoplasias/enzimología , Proteínas Supresoras de Tumor/fisiología , Aminoaciltransferasas/análisis , Animales , Células Cultivadas , Aberraciones Cromosómicas , Humanos , Ratones , Metástasis de la Neoplasia
3.
Mol Cell Biol ; 24(19): 8649-61, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15367683

RESUMEN

Both beta-catenin and plakoglobin can stimulate the expression of Lef/Tcf target genes in vitro. beta-Catenin is known to associate with Lef/Tcf factors and to participate directly in transactivation in vivo, whereas the role of plakoglobin in transcriptional regulation has been less studied. To analyze the functions of plakoglobin in vivo, we generated transgenic mice expressing in the epidermis N-terminally truncated plakoglobin (DeltaN122-PG) lacking the glycogen synthase kinase 3beta phosphorylation sites and therefore protected against degradation (transgenic line K5-DeltaN122-PG). The expression of DeltaN122-PG led to the formation of additional hair germs, hyperplastic hair follicles, and noninvasive hair follicle tumors, a phenotype reminiscent of that induced by expression of N-terminally truncated beta-catenin. However, if expressed in beta-catenin-null epidermis, DeltaN122-PG did not induce new hair follicle germs and follicular tumors. Thus, DeltaN122-PG cannot substitute for beta-catenin in its signaling functions in vivo and the phenotype observed in K5-DeltaN122-PG mouse skin must be due to the aberrant activation of beta-catenin signaling. On the other hand, the expression of DeltaN122-PG in beta-catenin-null skin significantly increased the survival rate of mutant mice, rescued differentiation, and limited excessive proliferation in the interfollicular epidermis, suggesting that plakoglobin may be involved in the intracellular signaling events essential for epidermal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Epidermis/crecimiento & desarrollo , Transactivadores/metabolismo , Animales , Cadherinas/metabolismo , Quistes/metabolismo , Proteínas del Citoesqueleto/genética , Desmoplaquinas , Epidermis/fisiología , Genes Reporteros , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/metabolismo , beta Catenina , gamma Catenina
4.
Oncogene ; 20(46): 6794-804, 2001 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-11709714

RESUMEN

Protein kinase C encodes a family of enzymes implicated in cellular differentiation, growth control and tumor promotion. However, not much is known with respect to the molecular mechanisms that link protein kinase C to cell cycle control. Here we report that the expression of PKCeta in MCF-7 cells, under the control of a tetracycline-responsive inducible promoter, enhanced cell growth and affected the cell cycle at several points. The induced expression of another PKC isoform, PKCdelta, in MCF-7 cells had opposite effects and inhibited their growth. PKCeta expression activated cellular pathways in these cells that resulted in the increased expression of the G1 phase cyclins, cyclin D and cyclin E. Expression of the cyclin-dependent kinase inhibitor p21(WAF1) was also specifically elevated in PKCeta expressing cells, but its overall effects were not inhibitory. Although, the protein levels of the cyclin-dependent kinase inhibitor p27(KIP1) were not altered by the induced expression of PKCeta, the cyclin E associated Cdk2 kinase activity was in correlation with the p27(KIP1) bound to the cyclin E complex and not by p21(WAF1) binding. PKCeta expression enhanced the removal of p27(KIP1) from this complex, and its re-association with the cyclin D/Cdk4 complex. Reduced binding of p27(KIP1) to the cyclin D/Cdk4 complex at early time points of the cell cycle also enhanced the activity of this complex, while at later time points the decrease in bound p21(WAF1) correlated with its increased activity in PKCeta-expressing cells. Thus, PKCeta induces altered expression of several cell cycle functions, which may contribute to its ability to affect cell growth.


Asunto(s)
Ciclinas/biosíntesis , Ciclinas/metabolismo , Isoenzimas/metabolismo , Isoenzimas/fisiología , Proteína Quinasa C/metabolismo , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas , Western Blotting , Ciclo Celular , División Celular , Ciclina D , Ciclina E/biosíntesis , Ciclina G , Ciclina G1 , Quinasa 4 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Isoenzimas/genética , Plásmidos/metabolismo , Pruebas de Precipitina , Regiones Promotoras Genéticas , Unión Proteica , Isoformas de Proteínas , Proteína Quinasa C/genética , Proteína Quinasa C-delta , Factores de Tiempo , Células Tumorales Cultivadas
5.
EMBO J ; 20(17): 4912-22, 2001 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-11532955

RESUMEN

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.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas de Ciclo Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN , Genes ras , Transactivadores , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Factores de Ribosilacion-ADP/deficiencia , Factores de Ribosilacion-ADP/genética , Animales , Transformación Celular Neoplásica/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas del Citoesqueleto/metabolismo , Factores de Transcripción E2F , Factor de Transcripción E2F1 , Embrión de Mamíferos , Fibroblastos/citología , Fibroblastos/fisiología , Regulación de la Expresión Génica , Genes Reporteros , Genes Supresores de Tumor , Genes p53 , Luciferasas/análisis , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación , Neoplasias/genética , Proteínas Recombinantes/análisis , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Transfección , Proteína p53 Supresora de Tumor/deficiencia , beta Catenina
6.
Mol Biol Cell ; 12(4): 1177-88, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11294915

RESUMEN

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.


Asunto(s)
Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila , Proteínas de Insectos/metabolismo , Transducción de Señal/fisiología , Transactivadores , Secuencia de Aminoácidos , Animales , Proteínas del Dominio Armadillo , Sitios de Unión , Células CHO , Cadherinas/genética , Línea Celular , Línea Celular Transformada , Cricetinae , Proteínas del Citoesqueleto/genética , Perros , Drosophila , Humanos , Proteínas de Insectos/genética , Mamíferos , Ratones , Datos de Secuencia Molecular , Saccharomyces cerevisiae , Factores de Transcripción , beta Catenina
7.
J Cell Sci ; 114(Pt 7): 1309-19, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11256997

RESUMEN

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.


Asunto(s)
Uniones Adherentes/metabolismo , Cadherinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Músculo Esquelético/fisiología , Transactivadores , Animales , Cadherinas/genética , Adhesión Celular/fisiología , Diferenciación Celular , Línea Celular , Medios de Cultivo , Proteínas del Citoesqueleto/genética , Expresión Génica , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miogenina/genética , Ratas , beta Catenina
8.
J Cell Sci ; 114(Pt 4): 695-707, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11171375

RESUMEN

The molecular basis for contact inhibition of cell locomotion is still largely unknown. Cadherins, the major receptors mediating cell-cell adhesion, associate in the cytoplasm with armadillo family proteins, including beta- and gamma-catenin and p120 catenin (p120ctn). E-cadherin-mediated contact formation was shown to inhibit cellular motility. We examine whether p120ctn may have a role in this regulation. We show here that overexpression of p120ctn in fibroblasts and epithelial cells induces pronounced changes in cell shape, motility and adhesion to the extracellular matrix. p120ctn-transfected cells display increased filopodial/lamellipodial activity, decreased contractility and focal adhesion formation, and augmented migratory ability. These effects of p120ctn are mediated by small GTPases of the Rho family. Direct assessment of the activity of these GTPases in cells expressing a 5-fold higher level of p120ctn as compared to non-transfected control cells revealed significant augmentation of Cdc42 and Rac activity. Moreover, co-transfection of p120ctn with dominant-negative Cdc42 and Rac, or constitutively active Rho suppressed morphological effects of p120ctn. Confocal immunofluorescence visualization of the distribution of endogenous p120ctn in dense cultures showed that formation of cadherin-mediated cell-cell contacts is accompanied by sequestering of p120ctn to the junction regions. In sparse cultures p120ctn is distributed over the cytoplasm. Co-transfection with an excess of E-cadherin leads to sequestration of exogenous p120ctn to cell-cell junctions or to small cadherin-containing vesicles, and abolishes p120ctn effects on cell morphology. Thus, p120ctn may couple the formation and disruption of cadherin-mediated contacts with regulation of cell motility by triggering pathway(s) affecting Rho family GTPases.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , GTP Fosfohidrolasas/fisiología , Fosfoproteínas/fisiología , Animales , Cateninas , Línea Celular , Humanos , Microscopía Fluorescente , Catenina delta
11.
J Biol Chem ; 275(42): 32649-57, 2000 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-10915780

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclina D1/genética , Regulación de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Pez Cebra , Factor de Transcripción Activador 2 , Animales , Neoplasias de la Mama , Antígenos CD18/fisiología , Línea Celular , Células Epiteliales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 , Glucógeno Sintasa Quinasas , Humanos , Integrina beta1/fisiología , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Subunidades de Proteína , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transfección , Células Tumorales Cultivadas , Proteínas Wnt , Proteína Wnt1
12.
J Cell Sci ; 113 ( Pt 18): 3127-39, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10954412

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Proteínas del Citoesqueleto/fisiología , Transactivadores , Proteínas de Pez Cebra , Animales , Sitios de Unión , Moléculas de Adhesión Celular/química , Moléculas de Adhesión Celular/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Desmoplaquinas , Humanos , Neoplasias , Conformación Proteica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt , beta Catenina , gamma Catenina
13.
Mol Cell Biol ; 20(12): 4238-52, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10825188

RESUMEN

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.


Asunto(s)
Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Transactivadores , Factores de Transcripción/genética , Activación Transcripcional , Transporte Biológico/genética , Línea Celular , Desmoplaquinas , Humanos , Mutación , beta Catenina , gamma Catenina
14.
Vopr Virusol ; 45(2): 20-5, 2000.
Artículo en Ruso | MEDLINE | ID: mdl-10765546

RESUMEN

Three HTLV-I-infected partially interleukin-2-dependent lymphoid cell lines were derived from a patient with T-cellular leukemia (ATL) from Georgia and a carrier of HTLV-I from Sakhalin. The strains cultured in the presence of 3-5% interleukin-2 were designated as NBK-1, NBK-2, and YE-1, respectively. Immunoblotting analysis showed typical HTLV-I proteins in them except NBK-2 which expressed nontypical proteins p40K and p28-29K. Unexpectedly, leukemic cell fraction ATL/NBK from a patient contained gag proteins p19 (CA), p24 (MA), Pr53, and unidentified protein p29. Southern blot analysis of primary leukemic cells NBK showed one full-length non-defective provirus with restriction sites Sacl in both LTRs. Limited restriction map of the provirus virtually did not differ from previously described HTLV-I prototypes. Although the mechanism of abnormal protein expression remains to be determined, this event can be explained by defective provirus formation in NBK-2 cell line during coculturing of leukemic cells with human umbilical cord blood lymphocytes.


Asunto(s)
Genoma Viral , Infecciones por HTLV-I/sangre , Leucemia de Células T/sangre , Southern Blotting , Infecciones por HTLV-I/virología , Humanos , Interleucina-2/metabolismo , Leucemia de Células T/virología , Mapeo Restrictivo , Proteínas Virales/genética
15.
J Biol Chem ; 275(28): 21203-9, 2000 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-10747899

RESUMEN

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.


Asunto(s)
Caveolinas , Ciclina D1/genética , Regulación de la Expresión Génica , Proteínas de la Membrana/fisiología , Regiones Promotoras Genéticas , Transcripción Genética , Secuencia de Aminoácidos , Animales , Células CHO , Caveolina 1 , Membrana Celular/fisiología , Cricetinae , Medio de Cultivo Libre de Suero , Proteínas de Unión al ADN/metabolismo , Humanos , Factor de Unión 1 al Potenciador Linfoide , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Proteínas Recombinantes de Fusión/biosíntesis , Factores de Transcripción/metabolismo , Transfección
16.
Mol Biol Cell ; 10(10): 3097-112, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10512853

RESUMEN

Caldesmon is known to inhibit the ATPase activity of actomyosin in a Ca(2+)-calmodulin-regulated manner. Although a nonmuscle isoform of caldesmon is widely expressed, its functional role has not yet been elucidated. We studied the effects of nonmuscle caldesmon on cellular contractility, actin cytoskeletal organization, and the formation of focal adhesions in fibroblasts. Transient transfection of nonmuscle caldesmon prevents myosin II-dependent cell contractility and induces a decrease in the number and size of tyrosine-phosphorylated focal adhesions. Expression of caldesmon interferes with Rho A-V14-mediated formation of focal adhesions and stress fibers as well as with formation of focal adhesions induced by microtubule disruption. This inhibitory effect depends on the actin- and myosin-binding regions of caldesmon, because a truncated variant lacking both of these regions is inactive. The effects of caldesmon are blocked by the ionophore A23187, thapsigargin, and membrane depolarization, presumably because of the ability of Ca(2+)-calmodulin or Ca(2+)-S100 proteins to antagonize the inhibitory function of caldesmon on actomyosin contraction. These results indicate a role for nonmuscle caldesmon in the physiological regulation of actomyosin contractility and adhesion-dependent signaling and further demonstrate the involvement of contractility in focal adhesion formation.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Actomiosina/metabolismo , Animales , Toxinas Botulínicas/farmacología , Calcimicina/farmacología , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión a Calmodulina/genética , Línea Celular Transformada , Citoesqueleto/metabolismo , Inhibidores Enzimáticos/farmacología , Expresión Génica , Proteínas Fluorescentes Verdes , Humanos , Ionóforos/farmacología , Proteínas Luminiscentes , Microscopía Fluorescente , Microtúbulos/metabolismo , Mutación , Nocodazol/farmacología , Ratas , Tapsigargina/farmacología , Transfección
17.
EMBO J ; 18(11): 3054-63, 1999 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10357817

RESUMEN

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.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Expresión Génica , Proteínas Nucleares , Transactivadores , Transcripción Genética/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Núcleo Celular/metabolismo , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/genética , Inhibidores de Cisteína Proteinasa/farmacología , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Desmoplaquinas , Proteínas Dishevelled , Humanos , Leupeptinas/farmacología , Ratones , Mutación , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-mdm2 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Solubilidad , Transfección , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , beta Catenina
18.
Proc Natl Acad Sci U S A ; 96(10): 5522-7, 1999 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-10318916

RESUMEN

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.


Asunto(s)
Ciclina D1/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras , Transactivadores , Factores de Transcripción/metabolismo , Proteína de la Poliposis Adenomatosa del Colon , Proteína Axina , Sitios de Unión , Cadherinas/metabolismo , Neoplasias del Colon/genética , Ciclina D1/metabolismo , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Factor de Unión 1 al Potenciador Linfoide , Regiones Promotoras Genéticas , Proteínas , Transducción de Señal , Activación Transcripcional , Transfección , Células Tumorales Cultivadas , beta Catenina
19.
Biochem Soc Symp ; 65: 147-72, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10320938

RESUMEN

The dynamic shape of an isolated cell results from an interplay between protrusion, adhesion and contraction activities. These are most closely associated with the actin cytoskeleton. In many cell types, microtubules have been shown to be involved in the development of morphological polarity required for directional migration. This suggests a role for the microtubule system in regulating both the actin cytoskeleton and the formation of cell-substrate adhesions. The most prominent role of microtubules in the cell is in transport of vesicles and organelles. Disruption of the microtubules, on the other hand, leads to a significant increase in actomyosin-driven contractility. This suggests the involvement of microtubules in the control of forces produced by the cell against the points at which it contacts the substrate or extracellular matrix. We show that microtubule disruption also activates an adhesion-dependent signal transduction cascade and promotes the formation of focal adhesions and associated actin microfilament bundles. Using overexpression of caldesmon, a regulatory protein which inhibits the interaction between actin and myosin, we show that these effects of microtubule disruption depend on the activation of contractility. Formation of focal adhesions induced by the small GTPase Rho is also blocked by the caldesmon inhibition of contractility. We infer that there is a step in the adhesion-dependent signalling pathway that requires mechanical tension applied to cell-substrate contacts. Although the experimental data are based on complete microtubule disruption, we suggest that a similar effect occurs locally following depolymerization of individual microtubules. We speculate that the interplay among microtubule dynamics, actomyosin contractility and adhesion-dependent signalling can produce a mechanism for the determination of cell polarity and direction of migration. In essence, microtubule depolymerization would create a local increase in contractile force, testing and promoting the maturation of nearby cell-substrate adhesions.


Asunto(s)
Adhesión Celular/fisiología , Movimiento Celular/fisiología , Microtúbulos/fisiología , Transducción de Señal , Actinas/fisiología , Citoesqueleto/fisiología
20.
Proc Natl Acad Sci U S A ; 95(26): 15339-44, 1998 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-9860970

RESUMEN

We studied the effect of N-cadherin, and its free or membrane-anchored cytoplasmic domain, on the level and localization of beta-catenin and on its ability to induce lymphocyte enhancer-binding factor 1 (LEF-1)-responsive transactivation. These cadherin derivatives formed complexes with beta-catenin and protected it from degradation. N-cadherin directed beta-catenin into adherens junctions, and the chimeric protein induced diffuse distribution of beta-catenin along the membrane whereas the cytoplasmic domain of N-cadherin colocalized with beta-catenin in the nucleus. Cotransfection of beta-catenin and LEF-1 into Chinese hamster ovary cells induced transactivation of a LEF-1 reporter, which was blocked by the N-cadherin-derived molecules. Expression of N-cadherin and an interleukin 2 receptor/cadherin chimera in SW480 cells relocated beta-catenin from the nucleus to the plasma membrane and reduced transactivation. The cytoplasmic tails of N- or E-cadherin colocalized with beta-catenin in the nucleus, and suppressed the constitutive LEF-1-mediated transactivation, by blocking beta-catenin-LEF-1 interaction. Moreover, the 72 C-terminal amino acids of N-cadherin stabilized beta-catenin and reduced its transactivation potential. These results indicate that beta-catenin binding to the cadherin cytoplasmic tail either in the membrane, or in the nucleus, can inhibit beta-catenin degradation and efficiently block its transactivation capacity.


Asunto(s)
Cadherinas/fisiología , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/metabolismo , Receptores de Interleucina-2/fisiología , Transactivadores , Factores de Transcripción/metabolismo , Activación Transcripcional , Animales , Células CHO , Cadherinas/química , Línea Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Pollos , Cricetinae , Proteínas del Citoesqueleto/genética , Cinética , Factor de Unión 1 al Potenciador Linfoide , Plásmidos , Receptores de Interleucina-2/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección , beta Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA