Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
FASEB J ; 23(3): 916-28, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19017743

RESUMO

We recently established the critical role of the PTEN/MAGI-1b signalosome in stabilization of cell-cell contacts and suppression of invasiveness. The PTEN tumor suppressor is recruited to E-cadherin junctional complexes through the binding to the second PDZ domain of the MAGI-1b scaffolding molecule, whereas beta-catenin interacts with the fifth PDZ domain. To identify additional effectors of this signalosome, we used yeast 2-hybrid screening. Among the clones identified, we focused on TRIP6, which belongs to the zyxin family of proteins. We demonstrated that TRIP6 interacted directly with MAGI-1b by binding to its fifth PDZ domain. Ectopic expression of TRIP6 induced invasiveness in the epithelial MDCK and MDCKts-src cells in a PI3-kinase- and a NF-kappaB-dependent manner and impaired cell-cell aggregation at least in part by uncoupling adherens junctional complexes from the cytoskeleton. The TRIP6Stop473 mutant, which lacks the PDZ binding motif, was still able to increase NF-kappaB and Akt activities but did not promote invasiveness or interfere with cell-cell aggregation. Intracellular delivery of competing peptides corresponding to TRIP6 or beta-catenin C terminus restored invasive properties in MDCKts-src TRIP6Stop473 cells, highlighting the requirement of PDZ scaffolds in junctional complexes activity. TRIP6 overexpression in colon tumors suggest its critical role in cancer progression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Fatores de Transcrição/metabolismo , ATPases Associadas a Diversas Atividades Celulares , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células CACO-2 , Caderinas/metabolismo , Adesão Celular , Moléculas de Adesão Celular , Moléculas de Adesão Celular Neuronais/genética , Linhagem Celular , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Citoesqueleto/metabolismo , Cães , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Guanilato Quinases , Células HeLa , Humanos , Proteínas com Domínio LIM , NF-kappa B/metabolismo , Complexo de Endopeptidases do Proteassoma , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/genética , Transfecção , Técnicas do Sistema de Duplo-Híbrido , Proteínas rho de Ligação ao GTP/metabolismo
2.
Int J Oncol ; 34(5): 1403-9, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19360353

RESUMO

Enhanced motility of cancer cells by remodelling of the actin cytoskeleton is crucial in the process of cancer cell invasion and metastasis. Although several studies propose a tumor suppressor role for the actin bundling protein myopodin, it was also shown previously that overexpression of mouse myopodin promotes invasion in vitro. In the present study, the role of myopodin in human cancer cell motility and invasion was explored using RNA interference with siRNA duplexes designed to down-regulate all human myopodin isoforms currently identified. We show that down-regulation of myopodin expression in human cancer cells significantly reduces the invasive properties of these cells both in collagen type I and in Matrigel. Furthermore, the motile characteristics of cancer cells are also curbed by reduced myopodin expression whereas cell-cell contacts are reinforced. These results point to a role for myopodin as tumor activator. While these findings are at variance with the suggested tumor suppressor role for myopodin, we hypothesize that the subcellular localization of the protein is involved in its suppressor or activator function in tumorigenesis.


Assuntos
Movimento Celular/genética , Proteínas dos Microfilamentos/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Masculino , Proteínas dos Microfilamentos/antagonistas & inibidores , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/fisiologia , Modelos Biológicos , Invasividade Neoplásica , RNA Interferente Pequeno/farmacologia , Distribuição Tecidual
3.
J Cell Biol ; 164(7): 1009-20, 2004 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-15037601

RESUMO

The Golgi apparatus has long been suggested to be important for directing secretion to specific sites on the plasma membrane in response to extracellular signaling events. However, the mechanisms by which signaling events are coordinated with Golgi apparatus function remain poorly understood. Here, we identify a scaffolding function for the Golgi matrix protein GM130 that sheds light on how such signaling events may be regulated. We show that the mammalian Ste20 kinases YSK1 and MST4 target to the Golgi apparatus via the Golgi matrix protein GM130. In addition, GM130 binding activates these kinases by promoting autophosphorylation of a conserved threonine within the T-loop. Interference with YSK1 function perturbs perinuclear Golgi organization, cell migration, and invasion into type I collagen. A biochemical screen identifies 14-3-3zeta as a specific substrate for YSK1 that localizes to the Golgi apparatus, and potentially links YSK1 signaling at the Golgi apparatus with protein transport events, cell adhesion, and polarity complexes important for cell migration.


Assuntos
Movimento Celular/fisiologia , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Proteínas 14-3-3 , Autoantígenos , DNA Complementar/genética , Ativação Enzimática , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Mutagênese Sítio-Dirigida , Fosforilação , Mutação Puntual , Reação em Cadeia da Polimerase , Proteínas Serina-Treonina Quinases/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Testículo/fisiologia , Testículo/ultraestrutura
4.
Int J Cancer ; 122(3): 572-82, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17955485

RESUMO

Dynamic crosstalk between cell adhesion molecules, extracellular matrix and soluble informative factors is essential for cancer cell migration and invasion. Here, we investigated the mechanisms by which the E-cadherin/catenin complex and alpha v integrin can modulate insulin-like growth factor-I (IGF-I)-induced cell migration. Human colon mucosa, human colon cancer cell lines, HT29-D4 and HCT-8 derivatives that differ in their expression of alpha-catenin, were used as models. Interactions between E-cadherin, alpha v integrin and IGF-I receptor (IGF-IR) were analyzed by coimmunoprecipitation and immunolocalization experiments. The impact of these interactions on cell mobility was determined by haptotaxis assays. We report that alpha v integrin, E-cadherin and IGF-IR form a ternary complex in both cultured cancer cells and human normal colonic mucosa. alpha-Catenin regulates the scaffolding of this complex. IGF-IR ligation by IGF-I induces the disruption of the complex and the relocalization of alpha v integrin from cell-cell contacts to focal contact sites. This perturbation is correlated with the observed increase in cell migration. These results suggest that regulation of the alpha v integrin/E-cadherin/IGF-IR scaffolding is essential for the modulation of cell mobility. Its alteration could be of major importance to sustain alterations in cell adhesion that occur during cancer cell invasion and metastasis.


Assuntos
Caderinas/metabolismo , Integrina alfaV/metabolismo , Receptor IGF Tipo 1/metabolismo , alfa Catenina/farmacologia , Adesão Celular , Movimento Celular , Citometria de Fluxo , Imunofluorescência , Células HT29/metabolismo , Humanos , Imunoprecipitação , Proteínas Substratos do Receptor de Insulina , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Interferente Pequeno/farmacologia
5.
Cell Signal ; 19(8): 1722-32, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17478078

RESUMO

Recent data indicate that the Bone Morphogenetic Protein BMP-7 exhibits mucosal protection against experimental colitis in rats, suggesting that this cytokine exerts direct actions in intestinal epithelial cells during inflammatory bowel diseases and other precancerous lesions of the colon. In this study, we investigated the functional expression of BMP-7 and its receptors in normal human colon crypts, aberrant crypt foci (ACF) in sigmoiditis and colorectal tumors, and their derived cancer cell lines. Transcripts encoding BMP-7 receptors type II (BMPRII, ActRII, ActRIIB) and type I (ALK-2) were clearly detected by RT-PCR in several premalignant and carcinoma cell lines. The cytokine was identified by immunohistochemistry in surface epithelial cells and crypts in the normal colon mucosa, ACF in sigmoiditis, sporadic high grade dysplastic adenoma, and in 9 of 16 colon carcinomas (56.2%). In addition, the conditioned medium collected from the adenoma PC/AA/C1 and carcinoma HCT8/S11 and SW48 cell lines in culture contained significant levels of BMP-7 ranging from 0.17 to 0.38 ng/ml. We found that BMP-7 induced scattering and proinvasive responses (EC50=1 ng/ml) in kidney and colon cancer cell lines through SMAD4 and src -independent pathways and signaling cascades using FAK phosphorylation at Y925 and activation of ERK1/2, Rac1 and JNK. This phosphorylation of FAK on Y925 was also induced by the proinvasive agent EGF. Taken together, our findings suggest that BMP-7 exerts divergent effects in the colon mucosa, one counteracting transient inflammatory situations and the other linked to pejorative functions during chronic ulcerative diseases and the neoplastic progression.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Neoplasias do Colo/patologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteína Smad4/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo , Proteína Morfogenética Óssea 7 , Linhagem Celular Tumoral , Neoplasias do Colo/enzimologia , Neoplasias do Colo/metabolismo , Ensaio de Imunoadsorção Enzimática , Células HT29 , Humanos , Imuno-Histoquímica , Estudos Retrospectivos
6.
Cancer Res ; 66(12): 6243-9, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778199

RESUMO

Emerging evidence supports neurotensin as a trophic and antiapoptotic factor, mediating its control via the high-affinity neurotensin receptor (NT1 receptor) in several human solid tumors. In a series of 51 patients with invasive ductal breast cancers, 34% of all tumors were positive for neurotensin and 91% positive for NT1 receptor. We found a coexpression of neurotensin and NT1 receptor in a large proportion (30%) of ductal breast tumors, suggesting a contribution of the neurotensinergic signaling cascade within breast cancer progression. Functionally expressed NT1 receptor, in the highly malignant MDA-MB-231 human breast cancer cell line, coordinated a series of transforming functions, including cellular migration, invasion, induction of the matrix metalloproteinase (MMP)-9 transcripts, and MMP-9 gelatinase activity. Disruption of NT1 receptor signaling by silencing RNA or use of a specific NT1 receptor antagonist, SR48692, caused the reversion of these transforming functions and tumor growth of MDA-MB-231 cells xenografted in nude mice. Our findings support the contribution of neurotensin in human breast cancer progression and point out the utility to develop therapeutic molecules targeting neurotensin or NT1 receptor signaling cascade. These strategies would increase the range of therapeutic approaches and be beneficial for specific patients.


Assuntos
Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Neurotensina/biossíntese , Receptores de Neurotensina/biossíntese , Animais , Carcinoma in Situ/metabolismo , Carcinoma in Situ/patologia , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Progressão da Doença , Ativação Enzimática , Feminino , Humanos , Imuno-Histoquímica , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica , Transplante Heterólogo
7.
Cancer Res ; 66(20): 10007-15, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17047063

RESUMO

Down-regulation of the epithelial cell-cell adhesion molecule E-cadherin is frequently associated with tumor formation and progression. Besides its role in physical cell-cell adhesion, E-cadherin is also thought to be involved in intracellular signaling in normal epithelial cells. In these cells, the Armadillo catenin p120ctn binds to the cytoplasmic domain of E-cadherin and stabilizes the adhesion complexes. On loss of E-cadherin, cytoplasmic p120ctn might accumulate and contribute to tumor malignancy. We used suppression subtractive hybridization to search for genes regulated by E-cadherin expression. We isolated human Nanos1 as a transcript of which levels decrease on E-cadherin reexpression in a human breast cancer cell line. The hNanos1 protein bears a COOH-terminal (CCHC)(2) zinc finger domain and belongs to an evolutionarily conserved protein family sharing functions in germ cell development in both vertebrates and invertebrates. We found an inverse correlation between E-cadherin and hNanos1 expression in various cell lines and under diverse conditions. Conditional expression of hNanos1 in human colorectal DLD1 cancer cells functionally abolished cell-cell adhesion. It induced cytoplasmic translocation of p120ctn, as well as strong migratory and invasive properties. We also found that the NH(2)-terminal domain of hNanos1, which is conserved only among mammals, interacts with p120ctn. hNanos1 counteracted the stimulatory effect of p120ctn on cell protrusion formation. Together, these findings describe a new function for hNanos1 as a downstream effector of E-cadherin loss contributing to tumor progression. Targeting hNanos1 might be a promising strategy in the treatment of E-cadherin-negative tumors in particular.


Assuntos
Caderinas/genética , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Tatus , Caderinas/biossíntese , Caderinas/metabolismo , Cateninas , Adesão Celular/fisiologia , Moléculas de Adesão Celular/biossíntese , Linhagem Celular Tumoral , Clonagem Molecular , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosfoproteínas/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Dedos de Zinco , delta Catenina
8.
Cancer Res ; 66(18): 9227-34, 2006 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16982767

RESUMO

A large number of hormones and local agonists activating guanine-binding protein-coupled receptors (GPCR) play a major role in cancer progression. Here, we characterize the new imidazo-pyrazine derivative BIM-46174, which acts as a selective inhibitor of heterotrimeric G-protein complex. BIM-46174 prevents the heterotrimeric G-protein signaling linked to several GPCRs mediating (a) cyclic AMP generation (Galphas), (b) calcium release (Galphaq), and (c) cancer cell invasion by Wnt-2 frizzled receptors and high-affinity neurotensin receptors (Galphao/i and Galphaq). BIM-46174 inhibits the growth of a large panel of human cancer cell lines, including anticancer drug-resistant cells. Exposure of cancer cells to BIM-46174 leads to caspase-3-dependent apoptosis and poly(ADP-ribose) polymerase cleavage. National Cancer Institute COMPARE analysis for BIM-46174 supports its novel pharmacologic profile compared with 12,000 anticancer agents. The growth rate of human tumor xenografts in athymic mice is significantly reduced after administration of BIM-46174 combined with either cisplatin, farnesyltransferase inhibitor, or topoisomerase inhibitors. Our data validate the feasibility of targeting heterotrimeric G-protein functions downstream the GPCRs to improve anticancer chemotherapy.


Assuntos
Cisteína/análogos & derivados , Proteínas Heterotriméricas de Ligação ao GTP/antagonistas & inibidores , Imidazóis/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisteína/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/antagonistas & inibidores , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Subunidades beta da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Células HL-60 , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Camundongos , Invasividade Neoplásica , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancer Lett ; 255(1): 57-70, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17493746

RESUMO

Gelsolin and CapG are both actin binding proteins that modulate a variety of physiological processes by interacting differently with the actin cytoskeleton. Several studies suggest that overexpression of these proteins promotes invasion in vitro. In this study we explored the contribution of these proteins in human cancer cell invasion and motility. We show that down regulation of CapG or gelsolin in several types of cancer cells, including MDA-MB 231 and PC-3 cells, significantly reduces the invasive and motile properties of cells, as well as cell aggregation. These results point to a role for CapG and gelsolin as tumor activator.


Assuntos
Gelsolina/biossíntese , Regulação Neoplásica da Expressão Gênica , Neoplasias/metabolismo , Actinas/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Colágeno/química , Colágeno Tipo I/metabolismo , Combinação de Medicamentos , Humanos , Técnicas In Vitro , Laminina/química , Proteínas dos Microfilamentos/metabolismo , Invasividade Neoplásica , Proteoglicanas/química , Interferência de RNA , Transdução de Sinais , Cicatrização
10.
Int J Oncol ; 31(6): 1501-8, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17982677

RESUMO

Increased src tyrosine kinase expression and activity has been associated with colon cancer cell invasion and survival. Several signaling pathways are involved in the oncogenic activation of src during the adenoma to carcinoma progression and cellular invasion. In the present study, the synthetic ether lipid analog ET-18-OMe was shown to promote invasion of HCT-8/S11 colon cancer cells into collagen type I through the concomitant activation of src by phosphorylation at Tyr416 (5-30 min) in alpha1-integrin immunoprecipitates containing the integrin binding proteins talin and paxillin, as well as the phoshorylated and activated forms of focal adhesion kinase (FAK) at Tyr397 (a FAK kinase activation signal), Tyr576 and Tyr861. This was associated with the lateral redistribution of alpha1-integrins in focal aggregates and persistent activation of the p130Cas/JNK pathways at 5-30 min, with the subsequent induction and activation of the matrix metalloproteinases MMP-2 and MMP-9 (2-12 h). These activated molecular scaffolds and signaling cascades were not observed in immunoprecipitates of alpha2- and beta1-integrins, and tetraspanin CD9, an invasion and metastasis suppressor linked to integrins and FAK signaling. Our data demonstrate that the lateral redistribution and clustering of alpha1-integrins results in the recruitment of the FAK/src motility-promoting signaling complex involved in cancer cell invasion. Disruption of this proinvasive pathway was accomplished by the dominant negative mutant of src (K295R, kinase dead), src pharmacological inhibitor (PP1) and alpha1-integrin function blocking antibodies. These findings support the notion that the alpha1-integrin- and src-dependent signalosome is a relevant therapeutic target against tumor progression in colon cancer patients.


Assuntos
Neoplasias do Colo/patologia , Proteína Substrato Associada a Crk/fisiologia , Quinase 1 de Adesão Focal/fisiologia , Integrina alfa1/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Quinases da Família src/fisiologia , Linhagem Celular Tumoral , Ativação Enzimática , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Fosforilação
11.
Nucleic Acids Res ; 33(20): 6566-78, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16314317

RESUMO

SIP1/ZEB2 is a member of the deltaEF-1 family of two-handed zinc finger nuclear factors. The expression of these transcription factors is associated with epithelial mesenchymal transitions (EMT) during development. SIP1 is also expressed in some breast cancer cell lines and was detected in intestinal gastric carcinomas, where its expression is inversely correlated with that of E-cadherin. Here, we show that expression of SIP1 in human epithelial cells results in a clear morphological change from an epithelial to a mesenchymal phenotype. Induction of this epithelial dedifferentiation was accompanied by repression of several cell junctional proteins, with concomitant repression of their mRNA levels. Besides E-cadherin, other genes coding for crucial proteins of tight junctions, desmosomes and gap junctions were found to be transcriptionally regulated by the transcriptional repressor SIP1. Moreover, study of the promoter regions of selected genes by luciferase reporter assays and chromatin immunoprecipitation shows that repression is directly mediated by SIP1. These data indicate that, during epithelial dedifferentiation, SIP1 represses in a coordinated manner the transcription of genes coding for junctional proteins contributing to the dedifferentiated state; this repression occurs by a general mechanism mediated by Smad Interacting Protein 1 (SIP1)-binding sites.


Assuntos
Células Epiteliais/metabolismo , Proteínas de Homeodomínio/fisiologia , Junções Intercelulares/metabolismo , Proteínas de Membrana/genética , Proteínas Repressoras/fisiologia , Sítios de Ligação , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Cromatina/metabolismo , Conexinas/genética , Conexinas/metabolismo , Regulação para Baixo , Células Epiteliais/citologia , Humanos , Proteínas de Membrana/metabolismo , Mesoderma/citologia , Mutação , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo
12.
Cancer Res ; 65(14): 6237-44, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16024625

RESUMO

Abberant activation of the process of epithelial-mesenchymal transition in cancer cells is a late event in tumor progression. A key inducer of this transition is the transcription factor Snail, which represses E-cadherin. We report that conditional expression of the human transcriptional repressor Snail in colorectal cancer cells induces an epithelial dedifferentiation program that coincides with a drastic change in cell morphology. Snail target genes control the establishment of several junctional complexes, intermediate filament networks, and the actin cytoskeleton. Modulation of the expression of these genes is associated with loss of cell aggregation and induction of invasion. Chromatin immunoprecipitation experiments showed that repression of selected target genes is associated with increased binding of Snail to their promoters, which contain consensus Snail-binding sites. Thus, Snail constitutes a master switch that directly represses the epithelial phenotype, resulting in malignant carcinoma cells.


Assuntos
Neoplasias Colorretais/patologia , Fatores de Transcrição/fisiologia , Actinas/genética , Actinas/metabolismo , Agregação Celular/fisiologia , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Células Epiteliais/patologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Transdução de Sinais , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
13.
Cancer Res ; 65(1): 195-202, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15665295

RESUMO

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.


Assuntos
Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/fisiologia , Mucinas/fisiologia , Proteínas Musculares/fisiologia , Transativadores/fisiologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Apoptose , Sequência de Bases , Divisão Celular , Linhagem Celular Tumoral , Primers do DNA , Humanos , Cinética , Invasividade Neoplásica , Peptídeos , Isoformas de Proteínas/fisiologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia , Fator Trefoil-3
14.
Mol Cancer Ther ; 5(8): 2070-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16928828

RESUMO

We recently showed by DNA microarray analysis that vascular endothelial growth factor (VEGF) receptor (VEGFR) is expressed in HCT8/S11 human colon cancer cells, suggesting that several angiogenic factors may target colon cancer cells themselves. In this study, transcripts encoding the VEGF-165 and semaphorin 3A (Sema3A) receptors and coreceptors Flt-1, KDR/Flk-1, plexin A1, and neuropilins NP-1 and NP-2 were identified by reverse transcription-PCR in the human colon cancer cell lines HCT8/S11, HT29, HCT116, and PCmsrc. Collagen invasion induced by VEGF-165 and Sema3A in HCT8/S11 cells (EC(50), 0.4-1 nmol/L) required p42/44 mitogen-activated protein kinase and signaling through RhoA/Rho-kinase-dependent and -independent pathways, respectively. As expected, the VEGFR signaling inhibitor ZD4190 selectively abrogated the proinvasive activity of VEGF in collagen gels (IC(50), 10 nmol/L) and chick heart fragments. We identify a novel function for VEGF-165 and Sema3A as proinvasive factors for human colorectal cancer cells. Interestingly, oral administration of the single drug ZD4190 to athymic mice (50 mg/kg/d, once daily) inhibited by 70% the growth of HCT8/S11 tumor cell xenografts. Combinations between the src tyrosine kinase inhibitor M475271 and ZD4190 or cisplatin resulted in additive therapeutic activity against LNM35 human lung tumor xenografts. Our data have significant implications for new therapeutic approaches and individualized treatment targeting VEGFR and src signaling pathways in combination with established clinical drugs at primary tumors and distant metastases in colon and lung cancer patients.


Assuntos
Neoplasias do Colo/tratamento farmacológico , Quinazolinas/farmacologia , Semaforina-3A/metabolismo , Triazóis/farmacologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Animais , Cisplatino/farmacologia , Neoplasias do Colo/patologia , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Piperidinas/farmacologia , Receptores de Fatores de Crescimento do Endotélio Vascular/efeitos dos fármacos , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Semaforina-3A/efeitos dos fármacos , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Oncogene ; 24(20): 3274-84, 2005 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-15735708

RESUMO

During a search to identify resveratrol (3,5,4'-trihydroxy-trans-stilbene, RV) target genes in the human erythroleukemic K562 cell line, we show here that the tensin gene and protein levels are remarkably induced by this dietary polyphenol. Tensin, a cell-matrix adhesion protein binding the integrins and cytoskeletal actin filaments also interacts with PI3-kinase and JNK signaling pathways. Tensin induction by RV is associated with increased K562 cell adhesion to fibronectin, cell spreading and actin polymerization. The same responses were observed in the tensin-deficient MCF7 human breast cancer cell line. In K562 and MCF7 cells treated by RV, tensin was found in punctate and intracytoplasmic areas. In MCF7 epithelial cells, induction of tensin is not exclusively associated with plasma membrane-bound vinculin, suggesting a dual localization of tensin in both focal and fibrillar adhesions. Pharmacological blockade of PI3-kinase and Rho GTPases/Rho-kinase resulted in selective depletion of focal adhesions, disorganization of tensin localization and disruption of stress fibers. RV increased cell motility and attachment to fibronectin in MCF7 cells submitted to mechanical laminar flow stress, and abrogated estrogen-induced MCF7 cancer cell invasion. Our data support the conclusion that induction of tensin by RV contributes to the chemopreventive and anti-invasive activity of this natural dietary compound in tensin-negative and -deficient leukemic cells or epithelioid cancers.


Assuntos
Anticarcinógenos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Proteínas dos Microfilamentos/biossíntese , Neoplasias/prevenção & controle , Estilbenos/farmacologia , Actinas/metabolismo , Adesão Celular , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Cicloeximida/farmacologia , Citoplasma/metabolismo , Citoesqueleto/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Humanos , Immunoblotting , Células K562 , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Resveratrol , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Tensinas , Fatores de Tempo
16.
Oncogene ; 24(56): 8240-51, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16091733

RESUMO

We recently reported that proteinase-activated receptors type I (PAR-1) are coupled to both negative and positive invasion pathways in colonic and kidney cancer cells cultured on collagen type I gels. Here, we found that treatments with the cell-permeant analog 8-Br-cGMP and the soluble guanylate cyclase activator BAY41-2272, and Rho kinase (ROK) inhibition by Y27632 or a dominant negative form of ROK lead to PAR-1-mediated invasion through differential Rac1 and Cdc42 signaling. Hypoxia or the counteradhesive matricellular protein SPARC/BM-40 (SPARC: secreted protein acidic rich in cysteine) overexpressed during cancer progression also commutated PAR-1 to cellular invasion through the cGMP/protein kinase G (PKG) cascade, RhoA inactivation, and Rac1-dependent or -independent signaling. Cultured primary cancer cells isolated from peritoneal and pleural effusions from patients with colon cancer or other malignant tumors harbored PAR-1, as shown by RT-PCR and FACS analyses. These malignant effusions also contained high levels of activated thrombin and fibrin, and induced a proinvasive response in HCT8/S11 human colorectal cancer cells. Our data underline the essential role of the tumor microenvironment and of several commutators targeting cGMP/PKG signaling and the RhoA-ROK axis in the control of PAR-1 proinvasive activity and metastatic potential of cancer cells in distant organs and peritoneal or pleural cavities. We also add new insights into the mechanisms linking the coagulation mediators thrombin and PAR-1 in the context of blood coagulation disorders and venous thrombosis often observed in cancer patients, as described in 1865 by Armand Trousseau.


Assuntos
Invasividade Neoplásica/patologia , Neoplasias/enzimologia , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptor PAR-1/fisiologia , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , GMP Cíclico/fisiologia , Cães , Receptor Quinase 1 Acoplada a Proteína G/antagonistas & inibidores , Receptor Quinase 1 Acoplada a Proteína G/fisiologia , Guanilato Ciclase , Células HCT116 , Células HT29 , Humanos , Hipóxia/enzimologia , Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Pessoa de Meia-Idade , Neoplasias/metabolismo , Osteonectina/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptor PAR-1/genética , Receptores de Antígenos/fisiologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/efeitos dos fármacos , Guanilil Ciclase Solúvel , Trombina/metabolismo , Quinases Associadas a rho , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Proteína rhoA de Ligação ao GTP/fisiologia
17.
FASEB J ; 19(1): 115-7, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15629897

RESUMO

We recently established the critical role of the lipid phosphatase activity of the PTEN tumor suppressor in stabilizing cell-cell contacts and suppressing invasiveness. To delineate the effector systems involved, we investigated the interaction of PTEN with E-cadherin junctional complexes in kidney and colonic epithelial cell lines. PTEN and the p85 regulatory subunit of phosphatidylinositol 3-OH kinase (PI3K) co-immunoprecipitated with E-cadherin and catenins. By using a yeast two-hybrid assay, we demonstrated that PTEN interacted indirectly with beta-catenin by binding the scaffolding protein MAGI-1b. This model was corroborated in various ways in mammalian cells. Ectopic expression of MAGI-1b potentiated the interaction of PTEN with junctional complexes, promoted E-cadherin-dependent cell-cell aggregation, and reverted the Src-induced invasiveness of kidney MDCKts-src cells. In this model, MAGI-1b slightly decreased the activity of AKT, a downstream effector of PI3K. By using dominant-negative and constitutively active AKT expression vectors, we demonstrated that this kinase was included in the pathways involved in Src-induced destabilization of junctional complexes and was necessary and sufficient to trigger invasiveness. We propose that the recruitment of PTEN at adherens junctions by MAGI-1b and the local down-regulation of phosphatidylinositol-3,4,5-trisphosphate pools and downstream effector systems at the site of cell-cell contacts are focal points for restraining both disruption of junctional complexes and induction of tumor cell invasion.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Junções Aderentes/metabolismo , Proteínas de Membrana/metabolismo , Invasividade Neoplásica/patologia , Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Aminoácidos , Animais , Proteína do Homeodomínio de Antennapedia , Células CACO-2/química , Células CACO-2/metabolismo , Caderinas/metabolismo , Carcinoma/genética , Moléculas de Adesão Celular , Linhagem Celular , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/metabolismo , Cães , Genes src , Guanilato Quinases , Células HT29/química , Células HT29/metabolismo , Proteínas de Homeodomínio/química , Humanos , Rim/citologia , Rim/embriologia , Masculino , Dados de Sequência Molecular , Invasividade Neoplásica/genética , Proteínas Nucleares/química , PTEN Fosfo-Hidrolase , Fosfatidato Fosfatase , Fosfatidilinositol 3-Quinases/metabolismo , Monoéster Fosfórico Hidrolases/deficiência , Neoplasias da Próstata/genética , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Fatores de Transcrição/química , Proteínas Supressoras de Tumor/deficiência , alfa Catenina
18.
FASEB J ; 19(1): 144-6, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15507471

RESUMO

Inappropriate activation of the Wnt/APC/beta-catenin signaling pathways plays a critical role at early stages in a variety of human cancers. However, their respective implication in tumor cell invasion is still hypothetical. Here, we show that two activators of the canonical Wnt/beta-catenin transcription pathway, namely Dvl-2, the Axin 501-560 fragment binding glycogen synthase kinase -3beta (GSK-3beta), and the negative Wnt regulator wt-Axin did not alter cell invasion into type I collagen. In addition, both Dvl-2 and Axin 501-560 exerted a permissive action on the proinvasive activity of HGF and intestinal trefoil factor. Upstream activation of Wnt signaling by the Wnt-2 and Wnt-3a ligands, stable overexpression of Wnt-2, as well as GSK-3beta inhibition by lithium, SB216763, and GSK-3beta dominant negative forms (K85R and R96E) conferred the invasive phenotype through several proinvasive pathways. Induction of the matrix metalloprotease MMP-7 (matrilysin) gene and protein by Wnt-2 was abolished by inactivation of the AP-1 binding site in the promoter. Accordingly, invasion induced by Wnt-2 was prevented by soluble FRP-3 and FRP-1, sequestration of Gbetagamma subunits, depletion of the GSK-3beta protein by RNA interference, the c-Jun dominant negative mutant TAM67 and was not reversed by wt-Axin. Thus, the proinvasive activity of Wnt-2 is mediated by a noncanonical Wnt pathway using GSK-3beta and the AP-1 oncogene. Our data provide a potential clue for our understanding of the action and crosstalk between Wnt activators and other proinvasive pathways, in relation with matrix substrates and proteases in human cancers.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Transdução de Sinais/genética , Fator de Transcrição AP-1/metabolismo , Animais , Anticorpos Monoclonais/metabolismo , Proteína Axina , Linhagem Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Cães , Células Epiteliais/química , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Vetores Genéticos/genética , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Glicoproteínas/farmacologia , Células HT29 , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Humanos , Indóis/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/agonistas , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intracelular , Rim/química , Rim/citologia , Rim/embriologia , Rim/metabolismo , Rim/virologia , Neoplasias Renais/genética , Neoplasias Renais/patologia , Ligantes , Cloreto de Lítio/farmacologia , Maleimidas/farmacologia , Metaloproteinase 7 da Matriz/biossíntese , Fragmentos de Peptídeos/farmacologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/farmacologia , Retroviridae , Transcrição Gênica/fisiologia , Proteínas Wnt , Proteína Wnt2
19.
Cancer Res ; 62(17): 4879-83, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12208734

RESUMO

The c-kit tyrosine kinase inhibitor STI571 exhibits a substantial therapeutic activity in patients with chronic myeloid leukemia and gastrointestinal stromal tumors respectively associated with constitutive activation of the BCR-ABL and c-kit tyrosine kinases. Human colorectal tumors also express the c-kit proto-oncogene. The present study focuses on the anticancer activity of STI571 in human colorectal tumor cells in vitro and in vivo. The c-kit receptor was identified as a M(r) 145,000 immunoreactive band in human colon cancer cells HT29, HCT8/S11, and HCT116. Cellular invasion induced by 10 ng/ml stem cell factor (EC(50) = 3 ng/ml) in HT29 cells was blocked by 1 micro M STI571 (IC(50) = 56 nM) and pharmacological inhibitors of several oncogenic signaling pathways, namely, phosphatidylinositol 3-kinase (LY294002), Rho GTPases (Clostridium botulinum exoenzyme C3 transferase), and Rho-kinase (Y27632). STI571 inhibited HT29 cell proliferation (IC(50) = 6 micro M) and induced apoptosis in vitro. These cellular effects were associated with a decrease in tumor growth. We also demonstrated that stem cell factor is a proangiogenic factor in vivo and in vitro. These encouraging results warrant further preclinical investigations and clinical trials on the use of the c-kit inhibitor STI571 as a chemotherapeutic agent in colon cancer prevention and in treatment of advanced colorectal cancers associated with liver metastases.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Piperazinas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/efeitos dos fármacos , Pirimidinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Benzamidas , Divisão Celular/efeitos dos fármacos , Embrião de Galinha , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/crescimento & desenvolvimento , Humanos , Mesilato de Imatinib , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neovascularização Fisiológica/efeitos dos fármacos , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-kit/biossíntese , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/antagonistas & inibidores , Fator de Células-Tronco/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Oncogene ; 23(19): 3317-27, 2004 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-15077188

RESUMO

STAT3 is frequently overexpressed and constitutively activated by tyrosine phosphorylation during malignant transformation. Despite the clear importance of STAT3 in cell proliferation and survival in diverse human cancers, its possible contribution to tumor cell adhesion, motility and invasion remains hypothetical. We therefore compared the transforming properties of STAT3wt, its constitutively activated dimeric form STAT3C, and the dominant negative mutant STAT3-Y705F in human colorectal HCT8/S11 cancer cells. Both STAT3wt and STAT3C exert a permissive action to the proinvasive activity of the scatter factor HGF in HCT8/S11 cells. In contrast, the monomeric and cytoplasmic mutant Y705F induces a constitutive invasive phenotype through Wnt/Rho-independent and EGFR/PI3-kinase-dependent pathways. Accordingly, Y705F decreases cell-cell homotypic adhesions, and increases cell motility and scattering, as well as lamellipodia-type cellular extensions. STAT3-Y705F-transfected HCT8/S11 cells display an increased tyrosine phosphorylation of the cell-cell adhesion regulator beta-catenin and its dissociation from the invasion suppressor E-cadherin at cell-cell contacts. Our data imply that both invasion promoter and repressor genes are controlled by the canonical STAT3 transcription pathways. Disruption of this cascade by Y705F reveals the proinvasive potential of altered forms of STAT3 as a persistent signaling adaptor in cytokine/transforming growth factor receptor scaffolds and oncogenic pathways.


Assuntos
Adesão Celular , Proteínas de Ligação a DNA/fisiologia , Invasividade Neoplásica , Transdução de Sinais , Transativadores/fisiologia , Caderinas/análise , Caderinas/fisiologia , Agregação Celular , Movimento Celular , Proteínas do Citoesqueleto/análise , Proteínas do Citoesqueleto/fisiologia , Receptores ErbB/fisiologia , Humanos , Fator 1 de Ligação ao Facilitador Linfoide , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Fator de Transcrição STAT3 , Transativadores/análise , Fatores de Transcrição/fisiologia , Proteínas Wnt , beta Catenina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA