Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Biochem J ; 447(1): 71-9, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22765757

RESUMEN

CCN5 (cysteine-rich 61/connective tissue growth factor/nephroblastoma overexpressed 5)/WISP-2 [WNT1 (wingless-type MMTV integration site family, member 1)-inducible signalling pathway protein 2] is an oestrogen-regulated member of the CCN family. CCN5 is a transcriptional repressor of genes associated with the EMT (epithelial-mesenchymal transition) and plays an important role in maintenance of the differentiated phenotype in ER (oestrogen receptor)-positive breast cancer cells. In contrast, CCN5 is undetectable in more aggressive ER-negative breast cancer cells. We now report that CCN5 is induced in ER-negative breast cancer cells such as MDA-MB-231 following glucocorticoid exposure, due to interaction of the endogenous glucocorticoid receptor with a functional glucocorticoid-response element in the CCN5 gene promoter. Glucocorticoid treatment of MDA-MB-231 cells is accompanied by morphological alterations, decreased invasiveness and attenuated expression of mesenchymal markers, including vimentin, cadherin 11 and ZEB1 (zinc finger E-box binding homeobox 1). Interestingly, glucocorticoid exposure did not increase CCN5 expression in ER-positive breast cancer cells, but rather down-regulated ER expression, thereby attenuating oestrogen pathway signalling. Taken together, our results indicate that glucocorticoid treatment of ER-negative breast cancer cells induces high levels of CCN5 expression and is accompanied by the appearance of a more differentiated and less invasive epithelial phenotype. These findings propose a novel therapeutic strategy for high-risk breast cancer patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proteínas CCN de Señalización Intercelular/metabolismo , Dexametasona/farmacología , Receptor alfa de Estrógeno/metabolismo , Glucocorticoides/farmacología , Proteínas Represoras/metabolismo , Secuencia de Bases , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular/genética , Línea Celular Tumoral , Cartilla de ADN/genética , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos
2.
BMC Cancer ; 12: 291, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22799881

RESUMEN

BACKGROUND: Ligand-dependent activation of the estrogen receptor (ER) as well as of the insulin-like growth factor type 1 (IGF1R) induces the proliferation of luminal breast cancer cells. These two pathways cooperate and are interdependent. We addressed the question of the mechanisms of crosstalk between the ER and IGF1R. METHODS: We evaluated the mitogenic effects of estradiol (E2; agonist ligand of ER) and of insulin (a ligand of IGF1R) in the MCF-7 cells by flow cytometry and by analyzing the cell levels of cell cycle-related proteins (immunoblotting) and mRNA (RT-QPCR). To verify the requirement for the kinase activity of Akt (a downstream target of IGF1R) in the mitogenic action of estradiol, we used shRNA strategy and shRNA-resistant expression vectors. RESULTS: The activation of the ER by E2 is unable to induce the cell cycle progression when the phosphatidyl inositol-3 kinase (PI3K)/Akt signaling is blocked by a chemical inhibitor (LY 294002) or by shRNA targeting Akt1 and Akt2. shRNA-resistant Akt wild-type constructs efficiently complemented the mitogenic signaling activity of E2 whereas constructs with inactivated kinase function did not. In growth factor-starved cells, the residual PI3K/Akt activity is sufficient to complement the mitogenic action of E2. Conversely, when ER function is blocked by the antiestrogen ICI 182780, IGF1R signaling is intact but does not lead to efficient reinitiation of the cell cycle in quiescent, growth factor-starved MCF-7 cells. The basal transcription-promoting activity of ligand-free ER in growth factor-starved cells is sufficient to complement the mitogenic action of the IGF1R-dependent signaling. CONCLUSIONS: The basal ER activity in the absence of ligand is sufficient to allow efficient mitogenic action of IGF1R agonists and needs to be blocked to prevent the cell cycle progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor IGF Tipo 1/metabolismo , Receptores de Estrógenos/metabolismo , Comunicación Autocrina , Neoplasias de la Mama/genética , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina A/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Estradiol/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Insulina/farmacología , Células MCF-7 , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal/efectos de los fármacos
3.
Drug Resist Updat ; 11(4-5): 123-51, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18718806

RESUMEN

The mechanisms involved in the epithelial to mesenchymal transition (EMT) are integrated in concert with master developmental and oncogenic pathways regulating in tumor growth, angiogenesis, metastasis, as well as the reprogrammation of specific gene repertoires ascribed to both epithelial and mesenchymal cells. Consequently, it is not unexpected that EMT has profound impacts on the neoplastic progression, patient survival, as well as the resistance of cancers to therapeutics (taxol, vincristine, oxaliplatin, EGF-R targeted therapy and radiotherapy), independent of the "classical" resistance mechanisms linked to genotoxic drugs. New therapeutic combinations using genotoxic agents and/or EMT signaling inhibitors are therefore expected to circumvent the chemotherapeutic resistance of cancers characterized by transient or sustained EMT signatures. Thus, targeting critical orchestrators at the convergence of several EMT pathways, such as the transcription pathways NF-kappaB, AKT/mTOR axis, MAPK, beta-catenin, PKC and the AP-1/SMAD factors provide a realistic strategy to control EMT and the progression of human epithelial cancers. Several inhibitors targeting these signaling platforms are already tested in preclinical and clinical oncology. In addition, upstream EMT signaling pathways induced by receptor and nonreceptor tyrosine kinases (e.g. EGF-R, IGF-R, VEGF-R, integrins/FAK, Src) and G-protein-coupled receptors (GPCR) constitute practical options under preclinical research, clinical trials or are currently used in the clinic for cancer treatment: e.g. small molecule inhibitors (Iressa: targeting selectively the EGF-R; CP-751,871, AMG479, NVP-AEW541, BMS-536924, PQIP, AG1024: IGF-R; AZD2171, ZD6474: VEGF-R; AZD0530, BMS-354825, SKI606: Src; BIM-46174: GPCR; rapamycin, CCI-779, RAD-001: mTOR) and humanized function blocking antibodies (Herceptin: ErbB2; Avastin: VEGF-A; Erbitux: EGF-R; Abegrin: alphavbeta3 integrins). We can assume that silencing RNA and adenovirus-based gene transfer of therapeutic miR and dominant interferring expression vectors targeting EMT pathways and signaling elements will bring additional ways for the treatment of epithelial cancers. Identification of the factors that initiate, modulate and effectuate EMT signatures and their underlying upstream oncogenic pathways should provide the basis of more efficient strategies to fight cancer progression as well as genetic and epigenetic forms of drug resistance. This goal can be accomplished using global screening of human clinical tumors by EMT-associated cDNA, proteome, miRome, and tissue arrays.


Asunto(s)
Transdiferenciación Celular , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Mesodermo/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Animales , Antineoplásicos/uso terapéutico , Supervivencia Celular , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Resistencia a Antineoplásicos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Humanos , Mesodermo/efectos de los fármacos , Mesodermo/patología , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
4.
Mol Cell Biol ; 25(6): 2419-30, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15743834

RESUMEN

Estrogen receptors (ER) are ligand-dependent transcription factors that regulate growth, differentiation, and maintenance of cellular functions in a wide variety of tissues. We report here that p21WAF1/CIP1, a cyclin-dependent kinase (Cdk) inhibitor, cooperates with CBP to regulate the ERalpha-mediated transcription of endogenous target genes in a promoter-specific manner. The estrogen-induced expression of the progesterone receptor and WISP-2 mRNA transcripts in MCF-7 cells was enhanced by p21WAF1/CIP1, whereas that of the cyclin D1 mRNA was reduced and the pS2 mRNA was not affected. Chromatin immunoprecipitation assays revealed that p21WAF1/CIP1 was recruited simultaneously with ERalpha and CBP to the endogenous progesterone receptor gene promoter in an estrogen-dependent manner. Experiments in which the p21WAF1/CIP1 protein was knocked down by RNA interference showed that the induction of the expression of the gene encoding the progesterone receptor required p21WAF1/CIP1, in contrast with that of the cyclin D1 and pS2 genes. p21WAF1/CIP1 induced not only cell cycle arrest in breast cancer cells but also milk fat globule protein and lipid droplets, indicators of the differentiated phenotype, as well as cell flattening and increase of the volume of the cytoplasm. These results indicate that p21WAF1/CIP1, in addition to its Cdk-regulatory role, behaves as a transcriptional coactivator in a gene-specific manner implicated in cell differentiation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/fisiología , Estradiol/fisiología , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , Neoplasias de la Mama/genética , Proteína de Unión a CREB , Ciclo Celular/genética , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Inmunoprecipitación de Cromatina , Ciclina D1/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Citoplasma/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas/genética , Proteínas/genética , Interferencia de ARN , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Transactivadores/metabolismo , Transactivadores/fisiología , Transcripción Genética/genética , Transcripción Genética/fisiología , Activación Transcripcional/genética , Activación Transcripcional/fisiología , Factor Trefoil-1 , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor
5.
Cancer Res ; 65(1): 195-202, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15665295

RESUMEN

Signal transducer and activator of transcription (STAT) 3 is overexpressed or activated in most types of human tumors and has been classified as an oncogene. In the present study, we investigated the contribution of the STAT3s to the proinvasive activity of trefoil factors (TFF) and vascular endothelial growth factor (VEGF) in human colorectal cancer cells HCT8/S11 expressing VEGF receptors. Both intestinal trefoil peptide (TFF3) and VEGF, but not pS2 (TFF1), activate STAT3 signaling through Tyr(705) phosphorylation of both STAT3alpha and STAT3beta isoforms. Blockade of STAT3 signaling by STAT3beta, depletion of the STAT3alpha/beta isoforms by RNA interference, and pharmacologic inhibition of STAT3alpha/beta phosphorylation by cucurbitacin or STAT3 inhibitory peptide abrogates TFF- and VEGF-induced cellular invasion and reduces the growth of HCT8/S11 tumor xenografts in athymic mice. Differential gene expression analysis using DNA microarrays revealed that overexpression of STAT3beta down-regulates the VEGF receptors Flt-1, neuropilins 1 and 2, and the inhibitor of DNA binding/differentiation (Id-2) gene product involved in the neoplastic transformation. Taken together, our data suggest that TFF3 and the essential tumor angiogenesis regulator VEGF(165) exert potent proinvasive activity through STAT3 signaling in human colorectal cancer cells. We also validate new therapeutic strategies targeting STAT3 signaling by pharmacologic inhibitors and RNA interference for the treatment of colorectal cancer patients.


Asunto(s)
Neoplasias del Colon/patología , Proteínas de Unión al ADN/fisiología , Mucinas/fisiología , Proteínas Musculares/fisiología , Transactivadores/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Apoptosis , Secuencia de Bases , División Celular , Línea Celular Tumoral , Cartilla de ADN , Humanos , Cinética , Invasividad Neoplásica , Péptidos , Isoformas de Proteínas/fisiología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Factor Trefoil-3
6.
Gene ; 374: 128-33, 2006 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-16516406

RESUMEN

Histone deacetylase inhibitors (HDIs) induced expression of the B-ind1 protein that is a component of Rac-1-signaling pathways leading to the modulation of gene expression. In the present study, we have determined the structure of the human B-ind1 gene promoter region. The oligocapping method revealed that the transcriptional start site of the human B-ind1 gene is located at 166 bases upstream of the first adenine residue of the translation start site that is highly homologous to an initiator (Inr) consensus sequence. In reporter assays, transactivation of the B-ind1 promoter was observed up to 300 bp of the initiation site. Deletion analysis of the promoter region revealed that histone deacetylase inhibitors (HDIs)-induced luciferase response was regulated by the core promoter elements. Mutation introduced into the proximal CG-boxes decreased most of the basal and HDIs-induced promoter activity. These results suggested a novel mechanism, which implicate minimal core promoter elements as potential mediator of HDIs.


Asunto(s)
Clonación Molecular , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas , Regiones Promotoras Genéticas , Proteínas/genética , Región de Flanqueo 5' , Secuencia de Bases , Sitios de Unión , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Genes Reporteros , Humanos , Hidroliasas , Péptidos y Proteínas de Señalización Intracelular , Luciferasas/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Proteínas/química , Proteínas/metabolismo , Eliminación de Secuencia , Factor de Transcripción Sp1/química , Sitio de Iniciación de la Transcripción , Activación Transcripcional
7.
Free Radic Biol Med ; 40(1): 75-86, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16337881

RESUMEN

Lung epithelium in cystic fibrosis (CF) patients is characterized by structural damage and altered repair due to oxidative stress. To gain insight into the oxidative stress-related damage in CF, we studied the effects of hyperoxia in CF and normal lung epithelial cell lines. In response to a 95% O2 exposure, both cell lines exhibited increased reactive oxygen species. Unexpectedly, the cyclin-dependent kinase inhibitor p21WAF1/CIP1 protein was undetectable in CF cells under hyperoxia, contrasting with increased levels of p21WAF1/CIP1 in normal cells. In both cell lines, exposure to hyperoxia led to S-phase arrest. Apoptotic features including nuclear condensation, DNA laddering, Annexin V incorporation, and elevated caspase-3 activity were not readily observed in CF cells in contrast to normal cells. Interestingly, treatment of hyperoxia-exposed CF cells with two proteasome inhibitors, MG132 and lactacystin, restored p21WAF1/CIP1 protein and was associated with an increase of caspase-3 activity. Moreover, transfection of p21WAF1/CIP1 protein in CF cells led to increased caspase-3 activity and was associated with increased apoptotic cell death, specifically under hyperoxia. Taken together, our data suggest that modulating p21WAF1/CIP1 degradation may have the therapeutic potential of reducing lung epithelial damage related to oxidative stress in CF patients.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibrosis Quística/enzimología , Pulmón/enzimología , Estrés Oxidativo , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Anexina A5/metabolismo , Apoptosis , Caspasa 3 , Caspasas/metabolismo , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidores de Cisteína Proteinasa/farmacología , Fibrosis Quística/patología , Células Epiteliales/enzimología , Células Epiteliales/patología , Humanos , Hiperoxia/enzimología , Leupeptinas/farmacología , Pulmón/patología , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Fase S
8.
J Endocrinol ; 191(3): 613-24, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17170219

RESUMEN

Wnt-1-induced signalling pathway protein-2 (WISP-2)/connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed (CCN)5 is a member of the CCN family of growth factors and was identified as an oestrogen- inducible gene in the MCF-7 cell line. However, the role of WISP-2/CCN5 in breast carcinogenesis remains unclear. In this study, we examined the mechanism by which oestrogens regulate the expression of human (h) Wnt-1 induced signalling pathway protein (WISP-2)/CCN5. Real-time RT-PCR showed that hWISP-2/CCN5 mRNA transcripts level is upregulated by oestrogens in the oestrogen receptor-positive human breast cancer cell lines MCF-7, T47D and ZR-75.1. Cloning of a 1.9 kb fragment of the hWISP-2/CCN5 5'-flanking sequence and subsequent analysis of potential transcription factor-binding sites identified a functional oestrogen response element site located between - 581 and - 569 upstream from the oestrogen-induced transcription start site. Transient transfections of MCF-7 cells with the cloned fragment showed that oestradiol caused an increase in reporter gene activity, which was inhibited by anti-oestrogens ICI 182 780 and 4-hydroxytamoxifen. Chromatin immunoprecipitation analysis revealed an oestradiol-dependent recruitment of the oestrogen receptor alpha to the oestrogen- responsive region of the hWISP-2/CCN5 gene promoter. We also showed that endogenous CREB-binding protein (CBP) and p21(WAF1/CIP1) are recruited to the chromosomal hWISP-2/CCN5 promoter in MCF-7 cells in an oestrogen-dependent manner, suggesting that CBP and p21(WAF1/CIP1) participate in the oestrogen receptor alpha-mediated transcriptional control of the hWISP-2/CCN5 gene.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Región de Flanqueo 5' , Secuencia de Bases , Western Blotting/métodos , Proteínas CCN de Señalización Intercelular , Proteína de Unión a CREB/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Clonación Molecular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Estradiol/análogos & derivados , Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Receptor alfa de Estrógeno/análisis , Receptor alfa de Estrógeno/metabolismo , Femenino , Fulvestrant , Expresión Génica/efectos de los fármacos , Genes Reporteros , Humanos , Péptidos y Proteínas de Señalización Intercelular/análisis , Datos de Secuencia Molecular , Proteínas de Neoplasias/análisis , Proteínas Represoras , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Factores de Transcripción/análisis , Transcripción Genética , Transfección/métodos
9.
Oncogene ; 21(37): 5773-82, 2002 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-12173048

RESUMEN

We have investigated the interaction between the expression of p21(WAF1/CIP1/SDI1), a stoichiometric inhibitor of Cdk, and the transcriptional activity of the oestrogen receptor alpha (ER(alpha). Transient transfection experiments demonstrated that the expression of p21(WAF1/CIP1/SDI1) amplified the transcriptional activation by ER(alpha). A dominant negative mutant of Cdk2 also enhanced the ER(alpha) transcriptional activity, indicating that the underlying mechanism relies on the inhibition of Cdk2 activity and cell cycle arrest. In agreement with this conclusion, experiments with p21(WAF1/CIP1/SDI1) mutants demonstrated that the domain involved in the binding of p21(WAF1/CIP1/SDI1) to Cdks was indispensable for the modulation of ER(alpha) activity. In addition, we show that expression of p21(WAF1/CIP1/SDI1) alleviates the block on CBP function mediated by Cdk2 and in turn stimulates transcriptional activation by ER(alpha) in a CBP-histone acetyltransferase (HAT)-dependent manner. These results suggest a novel mechanism by which p21(WAF1/CIP1/SDI1) functions as an enhancer of ER(alpha) activity through the modulation of CBP function.


Asunto(s)
Quinasas CDC2-CDC28 , Ciclinas/fisiología , Receptores de Estrógenos/fisiología , Proteínas de Saccharomyces cerevisiae , Activación Transcripcional , Acetiltransferasas/metabolismo , Animales , Células COS , Proteína de Unión a CREB , Quinasa 2 Dependiente de la Ciclina , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/fisiología , Receptor alfa de Estrógeno , Histona Acetiltransferasas , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Coactivador 1 de Receptor Nuclear , Fosforilación , Proteínas Serina-Treonina Quinasas/fisiología , Transactivadores/metabolismo , Transactivadores/fisiología , Factores de Transcripción/fisiología , Células Tumorales Cultivadas
10.
FASEB J ; 17(12): 1721-3, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12958188

RESUMEN

The matrix metalloprotease matrilysin is expressed in premalignant polyps and plays a key role in local invasion during the progression of digestive tumors. In the present work, we investigated the possible relationships between the activity of the mouse and human matrilysin promoters (Mp), endogenous matrilysin protein expression, and two early oncogenetic defects frequently observed in human colonic cancers, namely activation of the src oncogene and impairment of the Wnt/APC/beta-catenin pathway. Using transient transfection assays, we report here that src signaling and the HMG-box transcription factor LEF-1 act synergistically with the proximal (-61 to -67) AP-1 binding site to transactivate the Mp in premalignant and tumorigenic kidney and colonic epithelial cells, through beta-catenin- and axin-independent signaling pathways. This synergism involves the -109 and -194 Tcf/LEF-1 binding sites in the Mp and a physical interaction between LEF-1 and c-Jun. Furthermore, src coordinates accumulation of the c-Jun factor and matrilysin transcripts. Conversely, the c-Jun dominant negative mutant TAM67 and the src tyrosine kinase inhibitor M475271 impaired src-induced Mp activation, matrilysin protein accumulation, and invasion of type I collagen gels. This mechanism may thereby contribute to cellular invasion during the early-stage adenoma/adenocarcinoma conversion and the metastatic process of digestive tumors.


Asunto(s)
Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Proteínas de Unión al ADN/metabolismo , Metaloproteinasa 7 de la Matriz/genética , Proteína Oncogénica pp60(v-src)/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Neoplasias del Colon/patología , Inducción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Unión 1 al Potenciador Linfoide , Modelos Biológicos , Invasividad Neoplásica , Elementos de Respuesta , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas
11.
Mol Endocrinol ; 18(11): 2700-13, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15297603

RESUMEN

We have addressed the question of rapid, nongenomic mechanisms that may be involved in the mitogenic action of estrogens in hormone-dependent breast cancer cells. In quiescent, estrogen-deprived MCF-7 cells, estradiol did not induce a rapid activation of either the MAPK/ERK or phosphatidylinositol-3 kinase (PI-3K)/Akt pathway, whereas the entry into the cell cycle was documented by the successive inductions of cyclin D1 expression, hyperphosphorylation of the retinoblastoma protein (Rb), activity of the promoter of the cyclin A gene, and DNA synthesis. However, pharmacological inhibitors of the src family kinases, 4-amino-5-(4-methylphenyl)-7-(t-butyl) pyrazolo[3,4-d] pyrimidine (PP1) or of the PI-3K (LY294002) did prevent the entry of the cells into the cell cycle and inhibited the late G1 phase progression, whereas the inhibitor of MAPK/ERK activation (U0126) had only a partial inhibitory effect in the early G1 phase. In agreement with these results, small interfering RNA targeting Akt strongly inhibited the estradiolinduced cell cycle progression monitored by the activation of the promoter of the cyclin A gene. The expression of small interfering RNA targeting MAPK 1 and 2 also had a clear inhibitory effect on the estradiol-induced activation of the cyclin A promoter and also antagonized the estradiol-induced transcription directed by the estrogen response element. Finally, transfection of the estrogen receptor into NIH3T3 fibroblasts did not confer to the cells sensitivity to a mitogenic action of estradiol. We conclude that the induction of the cell cycle by estradiol does not require a direct activation of MAPK/ERK or PI-3K signaling protein kinase cascades, but that these kinases appear to have a permissive role in the cell cycle progression.


Asunto(s)
Neoplasias de la Mama/enzimología , Estrógenos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Butadienos/farmacología , Línea Celular Tumoral , Cromonas/farmacología , Ciclina A/genética , Estradiol/farmacología , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Fibroblastos/química , Fibroblastos/metabolismo , Fase G1/efectos de los fármacos , Humanos , Ratones , Mitógenos/farmacología , Morfolinas/farmacología , Nitrilos/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Regiones Promotoras Genéticas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptores de Estrógenos/análisis , Receptores de Estrógenos/genética
12.
PLoS One ; 9(2): e87878, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498388

RESUMEN

It has been proposed that the epithelial-mesenchymal transition (EMT) in mammary epithelial cells and breast cancer cells generates stem cell features. WISP2 (Wnt-1-induced signaling protein-2) plays an important role in maintenance of the differentiated phenotype of estrogen receptor-positive breast cancer cells and loss of WISP2 is associated with EMT. We now report that loss of WISP2 in MCF7 breast cancer cells can also promote the emergence of a cancer stem-like cell phenotype characterized by high expression of CD44, increased aldehyde dehydrogenase activity and mammosphere formation. Higher levels of the stem cell markers Nanog and Oct3/4 were observed in those mammospheres. In addition we show that low-cell inoculums are capable of tumor formation in the mammary fat pad of immunodeficient mice. Gene expression analysis show an enrichment of markers linked to stem cell function such as SOX9 and IGFBP7 which is linked to TGF-ß inducible, SMAD3-dependent transcription. Taken together, our data demonstrate that WISP2 loss promotes both EMT and the stem-like cell phenotype.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Proteínas CCN de Señalización Intercelular/metabolismo , Transición Epitelial-Mesenquimal , Estrógenos/farmacología , Células Madre Neoplásicas/patología , Proteínas Represoras/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Proteínas CCN de Señalización Intercelular/genética , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Ratones , Ratones Desnudos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Fenotipo , Proteínas Represoras/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
13.
Mol Cell Biol ; 31(7): 1459-69, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21262769

RESUMEN

CCN5 is a member of the CCN (connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed) family and was identified as an estrogen-inducible gene in estrogen receptor-positive cell lines. However, the role of CCN5 in breast carcinogenesis remains unclear. We report here that the CCN5 protein is localized mostly in the cytoplasm and in part in the nucleus of human tumor breast tissue. Using a heterologous transcription assay, we demonstrate that CCN5 can act as a transcriptional repressor presumably through association with histone deacetylase 1 (HDAC1). Microarray gene expression analysis showed that CCN5 represses expression of genes associated with epithelial-mesenchymal transition (EMT) as well as expression of key components of the transforming growth factor ß (TGF-ß) signaling pathway, prominent among them TGF-ßRII receptor. We show that CCN5 is recruited to the TGF-ßRII promoter, thereby providing a mechanism by which CCN5 restricts transcription of the TGF-ßRII gene. Consistent with this finding, CCN5, we found, functions to suppress TGF-ß-induced transcriptional responses and invasion that is concomitant with EMT. Thus, our data uncovered CCN5 as a novel transcriptional repressor that plays an important role in regulating tumor progression functioning, at least in part, by inhibiting the expression of genes involved in the TGF-ß signaling cascade that is known to promote EMT.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas CCN de Señalización Intercelular , Cadherinas/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Histona Desacetilasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Invasividad Neoplásica , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Proteínas Represoras/química , Fracciones Subcelulares/metabolismo , Factores de Transcripción/química , Factor de Crecimiento Transformador beta/genética
14.
Mol Cell Biol ; 28(3): 1114-23, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18070926

RESUMEN

WISP-2/CCN5 is an estrogen-regulated member of the "connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed" (CCN) family of the cell growth and differentiation regulators. The WISP-2/CCN5 mRNA transcript is undetectable in normal human mammary cells, as well as in highly aggressive breast cancer cell lines, in contrast with its higher level in the breast cancer cell lines characterized by a more differentiated phenotype. We report here that knockdown of WISP-2/CCN5 by RNA interference in estrogen receptor alpha (ERalpha)-positive MCF-7 breast cancer cells induced an estradiol-independent growth linked to a loss of ERalpha expression and promoted epithelial-to-mesenchymal transdifferentiation. In contrast, forced expression of WISP-2/CCN5 directed MCF-7 cells toward a more differentiated phenotype. When introduced into the poorly differentiated, estrogen-independent, and invasive MDA-MB-231 breast cancer cells, WISP-2/CCN5 was able to reduce their proliferative and invasive phenotypes. In a series of ERalpha-positive tumor biopsies, we found a positive correlation between the expression of WISP-2/CCN5 and ID2, a transcriptional regulator of differentiation in normal and transformed breast cells. We propose that WISP-2/CCN5 is an important regulator involved in the maintenance of a differentiated phenotype in breast tumor epithelial cells and may play a role in tumor cell invasion and metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Péptidos y Proteínas de Señalización Intercelular/fisiología , Factores de Transcripción/fisiología , Neoplasias de la Mama/etiología , Proteínas CCN de Señalización Intercelular , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Represoras , Factores de Transcripción/genética
15.
Histochem Cell Biol ; 130(3): 481-94, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18648847

RESUMEN

Reduction of epithelial cell-cell adhesion via the transcriptional repression of cadherins in combination with the acquisition of mesenchymal properties are key determinants of epithelial-mesenchymal transition (EMT). EMT is associated with early stages of carcinogenesis, cancer invasion and recurrence. Furthermore, the tumor stroma dictates EMT through intensive bidirectional communication. The pathological analysis of EMT signatures is critically, especially to determine the presence of cancer cells at the resection margins of a tumor. When diffusion barriers disappear, EMT markers may be detected in sera from cancer patients. The detection of EMT signatures is not only important for diagnosis but can also be exploited to enhance classical chemotherapy treatments. In conclusion, further detailed understanding of the contextual cues and molecular mediators that control EMT will be required in order to develop diagnostic tools and small molecule inhibitors with potential clinical implications.


Asunto(s)
Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Invasividad Neoplásica/patología , Animales , Líquidos Corporales , Espacio Extracelular/metabolismo , Humanos
16.
Int J Cancer ; 97(6): 761-9, 2002 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-11857351

RESUMEN

We have studied the effects of purvalanol A on the cell cycle progression, proliferation and viability. In synchronized cells, purvalanol A induced a reversible arrest the progression in G1 and G2 phase of the cell cycle, but did not prevent the completion of DNA synthesis in S-phase cells. The specificity of action of the drug was supported by the selective inhibition of the phosphorylation of cyclin-dependent kinase (cdk) substrates such as Rb and cyclin E. The cell contents of cyclins D1 and E were lower in cells incubated with purvalanol A compared to controls, but the level of the cdk inhibitory protein p21(WAF1/CIP1) was increased, indicating that the drug did not cause a general inhibition of gene expression. Purvalanol A did not inhibit transcription under cell-free conditions. This compound, however, caused an inhibition of the estradiol-induced expression of an integrated luciferase gene, suggesting that cdk or related enzymes may participate in the regulation of the activity of certain promoters. When exponentially growing cells, both mouse fibroblasts and human cancer cell lines, were incubated with purvalanol A for prolonged periods of time (24 hr), a lasting inhibition of cell proliferation as well as cell death were observed. In contrast, a 24 hr incubation of quiescent (non-transformed) cells with purvalanol A did not prevent their resumption of cell cycle after removal of the drug.


Asunto(s)
Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/metabolismo , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteínas Musculares , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclina D1/metabolismo , Ciclina E/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Femenino , Células HT29/efectos de los fármacos , Células HeLa/efectos de los fármacos , Humanos , Metionina/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosforilación , Proteína de Retinoblastoma/metabolismo , Transcripción Genética , Células Tumorales Cultivadas/efectos de los fármacos , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/patología
17.
J Virol ; 78(22): 12198-206, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15507606

RESUMEN

In human immunodeficiency virus type 1 (HIV-1)-infected cells, a cell cycle arrest in G(2) increases viral expression and may represent a strategy for the virus to optimize its expression. In latently infected cells, balance between viral silencing and reactivation relies on the nucleosomal organization of the integrated long terminal repeat (LTR). It is shown here that nucleosome nuc-1, which is located downstream of the TATA box, is specifically modified when latently infected cells are arrested in G(2) by chemical inducers. Notably, histones H3 and H4 are hyperacetylated, and this modification is associated with an increased LTR-driven transcription. nuc-1 hyperacetylation is also associated with the recruitment of histone acetyltransferase CBP and transcription factors NF-kappaB and c-Jun. NF-kappaB and/or c-Jun binding to the LTR in G(2)-arrested cells appears to be required for CBP recruitment as well as for nuc-1 remodeling and viral reactivation.


Asunto(s)
Proteínas Portadoras/fisiología , Fase G2 , VIH-1/genética , Histonas/metabolismo , FN-kappa B/fisiología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/fisiología , Secuencias Repetidas Terminales , Activación Transcripcional , Acetilación , Línea Celular , Corticosterona , VIH-1/fisiología , Humanos , Activación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA