Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Cell Biol ; 25(10): 4034-45, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15870276

RESUMO

Gap junctions are composed of connexins and are critical for the maintenance of the differentiated state. Consistently, connexin expression is impaired in most cancer cells, and forced expression of connexins following cDNA transfection reverses the tumor phenotype. We have found that the restoration of density inhibition of human pancreatic cancer cells by the antiproliferative somatostatin receptor 2 (sst2) is due to overexpression of endogenous connexins Cx26 and Cx43 and consequent formation of functional gap junctions. Immunoblotting along with protein metabolic labeling and mRNA monitoring revealed that connexin expression is enhanced at the level of translation but is not sensitive to the inhibition of cap-dependent translation initiation. Furthermore, we identified a new internal ribosome entry site (IRES) in the Cx26 mRNA. The activity of Cx26 IRES and that of the previously described Cx43 IRES are enhanced in density-inhibited cells. These data indicate that the restoration of functional gap junctions is likely a critical event in the antiproliferative action of the sst2 receptor. We further suggest that the existence of IRESes in connexin mRNAs permits connexin expression in density-inhibited or differentiated cells, where cap-dependent translation is generally reduced.


Assuntos
Conexina 43/biossíntese , Conexinas/biossíntese , Junções Comunicantes/fisiologia , Neoplasias/metabolismo , Neoplasias/patologia , Ribossomos/metabolismo , Regiões 5' não Traduzidas/genética , Comunicação Celular , Contagem de Células , Linhagem Celular Tumoral , Proliferação de Células , Conexina 26 , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Humanos , Neoplasias/genética , Biossíntese de Proteínas/efeitos dos fármacos , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Sirolimo/farmacologia , Regulação para Cima/efeitos dos fármacos
2.
Mol Biol Cell ; 14(9): 3911-28, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12972574

RESUMO

The G protein-coupled sst2 somatostatin receptor acts as a negative cell growth regulator. Sst2 transmits antimitogenic signaling by recruiting and activating the tyrosine phosphatase SHP-1. We now identified Src and SHP-2 as sst2-associated molecules and demonstrated their role in sst2 signaling. Surface plasmon resonance and mutation analyses revealed that SHP-2 directly associated with phosphorylated tyrosine 228 and 312, which are located in sst2 ITIMs (immunoreceptor tyrosine-based inhibitory motifs). This interaction was required for somatostatin-induced SHP-1 recruitment and activation and consequent inhibition of cell proliferation. Src interacted with sst2 and somatostatin promoted a transient Gbetagamma-dependent Src activation concomitant with sst2 tyrosine hyperphosphorylation and SHP-2 activation. These steps were abrogated with catalytically inactive Src. Both catalytically inactive Src and SHP-2 mutants abolished somatostatin-induced SHP-1 activation and cell growth inhibition. Sst2-Src-SHP-2 complex formation was dynamic. Somatostatin further induced sst2 tyrosine dephosphorylation and complex dissociation accompanied by Src and SHP-2 inhibition. These steps were defective in cells expressing a catalytically inactive Src mutant. All these data suggest that Src acts upstream of SHP-2 in sst2 signaling and provide evidence for a functional role for Src and SHP-2 downstream of an inhibitory G protein-coupled receptor.


Assuntos
Proteínas Tirosina Fosfatases/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Quinases da Família src/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Células COS , Divisão Celular/fisiologia , Chlorocebus aethiops , Cricetinae , Ciclofosfamida , Análise Mutacional de DNA , Doxorrubicina , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Moleculares , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Ratos , Receptores de Somatostatina/análise , Somatostatina/farmacologia , Ressonância de Plasmônio de Superfície , Vincristina
3.
Cancer Res ; 65(7): 2770-7, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15805277

RESUMO

MTI/G-Gly mice and hGAS mice, overexpressing glycine-extended gastrin (G-Gly) and progastrin, respectively, display colonic mucosa hyperplasia, hyperproliferation, and an increased susceptibility to intestinal neoplasia. Here, we have used these transgenic mice to analyze in vivo the modulation of intracellular signaling pathways that may be responsible for the proliferative effects of gastrin precursors. The expression, activation, and localization of signaling and cell-to-cell adhesion molecules were studied using immunofluorescence and Western blot techniques on colonic tissues derived from MTI/G-Gly, hGAS, or wild-type FVB/N mice. These analyses revealed an up-regulation of Src tyrosine kinase and related signaling pathways [phosphatidyl inositol 3'-kinase (PI3K)/Akt, Janus-activated kinase (JAK) 2, signal transducer and activator of transcription (STAT) 3, and extracellular-signal regulated kinases (ERK)] in both MTI/G-Gly and hGAS mice compared with the wild-type control animals as well as an overexpression of transforming growth factor-alpha (TGF-alpha). In contrast, overexpression of the gastrin precursors did not affect the activation status of STAT1 nor the expression and the distribution of adhesion proteins (focal adhesion kinase, cadherins, and catenins). We report for the first time that the transition from a normal colonic epithelium to a hyperproliferative epithelium in MTI/G-Gly and hGAS mice may be a consequence of the up-regulation of Src, PI3K/Akt, JAK2, STAT3, ERKs, and TGF-alpha. Deregulation of cell adhesion, a late event in tumor progression, does not occur in these transgenic models.


Assuntos
Colo/patologia , Gastrinas/metabolismo , Mucosa Intestinal/patologia , Precursores de Proteínas/metabolismo , Animais , Adesão Celular/fisiologia , Proliferação de Células , Colo/enzimologia , Proteínas de Ligação a DNA/metabolismo , Feminino , Hiperplasia/enzimologia , Hiperplasia/patologia , Mucosa Intestinal/enzimologia , Janus Quinase 2 , Masculino , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia , Transativadores/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Regulação para Cima , Quinases da Família src/metabolismo
4.
World J Gastroenterol ; 12(21): 3344-51, 2006 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-16733850

RESUMO

AIM: To compare gene expression profiles of pancreatic adenocarcinoma tissue specimens, human pancreatic and colon adenocarcinoma and leukemia cell lines and normal pancreas samples in order to distinguish differentially expressed genes and to validate the differential expression of a subset of genes by quantitative real-time RT-PCR (RT-QPCR) in endoscopic ultrasound-guided fine needle aspiration (EUS-guided FNA) specimens. METHODS: Commercially dedicated cancer cDNA macroarrays (Atlas Human Cancer 1.2) containing 1176 genes were used. Different statistical approaches (hierarchical clustering, principal component analysis (PCA) and SAM) were used to analyze the expression data. RT-QPCR and immunohistochemical studies were used for validation of results. RESULTS: RT-QPCR validated the increased expression of LCN2 (lipocalin 2) and for the first time PLAT (tissue-type plasminogen activator or tPA) in malignant pancreas as compared with normal pancreas. Immunohistochemical analysis confirmed the increased expression of LCN2 protein localized in epithelial cells of ducts invaded by carcinoma. The analysis of PLAT and LCN2 transcripts in 12 samples obtained through EUS-guided FNA from patients with pancreatic adenocarcinoma showed significantly increased expression levels in comparison with those found in normal tissues, indicating that a sufficient amount of high quality RNA can be obtained with this technique. CONCLUSION: Expression profiling is a useful method to identify biomarkers and potential target genes. Molecular analysis of EUS-guided FNA samples in pancreatic cancer appears as a valuable strategy for the diagnosis of pancreatic adenocarcinomas.


Assuntos
Adenocarcinoma/genética , Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Pancreáticas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas de Fase Aguda/análise , Proteínas de Fase Aguda/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Biópsia por Agulha Fina/métodos , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Endossonografia/métodos , Genes Neoplásicos/genética , Humanos , Queratina-7 , Queratinas/análise , Queratinas/genética , Leucemia/genética , Leucemia/patologia , Lipocalina-2 , Lipocalinas , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/patologia , Prognóstico , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/genética , Reprodutibilidade dos Testes , Ativador de Plasminogênio Tecidual/análise , Ativador de Plasminogênio Tecidual/genética
5.
Cancer Res ; 62(21): 6124-31, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12414637

RESUMO

Our previous studies conducted in pancreatic cancer models established in nude mice and hamsters revealed that cloned somatostatin receptor subtype 2 (sst2) gene expression induced both antioncogenic and local antitumor bystander effects in vivo. In the present study, in vivo gene transfer of sst2 was investigated in two transplantable models of primary and metastatic pancreatic carcinoma developed in hamsters. LacZ reporter or mouse sst2 genes were expressed by means of two different delivery agents: an adenoviral vector and a synthetic polycationic carrier [linear polyethylenimine (PEI)]. sst2 was injected into either exponentially growing pancreatic primary tumors or hepatic metastases, and then transgene expression and tumor progression were investigated 5-6 days after gene transfer. Molecular mechanisms involved in the inhibition of tumor growth were also analyzed. Both adenovirus- and PEI-mediated in vivo gene transfer in primary pancreatic tumors induced an increase of beta-galactosidase activity and expression of sst2 transgene nRNA (100% and 86% of tumors for adenovirus and PEI vector, respectively). Adenoviral vector-based sst2 gene transfer resulted in significant reduction of pancreatic tumor growth (P < 0.05). Using PEI vector, both pancreatic primary tumor growth and metastatic tumor growth were also significantly slackened as compared with both LacZ-treated and untreated control groups (P < 0.02). Moreover, the proliferative index decreased significantly (P < 0.005), whereas apoptosis increased (P < 0.005) in tumors transferred with sst2 gene. The increase of apoptosis correlated with an activation of the caspase-3 and poly(ADP-ribose) polymerase pathways. We concluded that in both primary and metastatic pancreatic cancer models, the synthetic gene delivery system can achieve in vivo sst2 gene transfer and results in a significant antitumor effect characterized by an increase of apoptosis and an inhibition of cell proliferation. This new strategy of gene therapy allows the restoration of expression of an antioncogenic molecule and could be promising for the treatment of advanced pancreatic cancer.


Assuntos
Carcinoma Ductal Pancreático/terapia , Terapia Genética/métodos , Neoplasias Pancreáticas/terapia , Receptores de Somatostatina/genética , Adenoviridae/genética , Animais , Apoptose/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/secundário , Divisão Celular/genética , Cricetinae , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Neoplasias Hepáticas Experimentais/genética , Neoplasias Hepáticas Experimentais/metabolismo , Neoplasias Hepáticas Experimentais/secundário , Neoplasias Hepáticas Experimentais/terapia , Masculino , Mesocricetus , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Polietilenoimina/administração & dosagem , Receptores de Somatostatina/biossíntese , Receptores de Somatostatina/fisiologia , Transgenes
6.
Oncogene ; 21(50): 7656-70, 2002 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-12400008

RESUMO

The presence of gastrin and CCK-2/gastrin receptors in human preneoplastic and neoplastic lesions of pancreas and colon suggests a role in cancer development. Gastrin's growth-promoting action has been established, but a role in cellular morphogenetic processes promoting tumor invasion has been elusive. Our aim was (i) to investigate whether activation of the CCK-2R affects cellular morphology, intercellular adhesion and motility, as crucial parameters of epithelial differentiation, and (ii) to identify the signaling pathways and mechanisms implicated. Madin-Darby Canine Kidney (MDCK) cells were chosen to generate an epithelial non-tumorigenic model system expressing human CCK-2R. Epithelial differentiation and motility were analysed upon CCK-2R activation using immunocytochemistry and invasion assays. The functionality of adhesion complexes and activity of signaling proteins was determined with biochemical techniques. CCK-2R activation induced cell dissociation and enhanced invasion, preceded by decreased membrane localization of adherens junction molecules and nuclear accumulation of beta-catenin. Concomitantly, and requiring the activation of several signaling pathways, catenins were shifted from the cytoskeletal to the cytoplasmic fraction, suggesting the detachment of the cytoskeleton from the adherens complex. These data represent the first evidence for the CCK-2R, regulating cell-cell and cell-substrate adhesion and support a role for CCK-2R in the progression of carcinoma.


Assuntos
Adesão Celular/fisiologia , Células Epiteliais/metabolismo , Gastrinas/metabolismo , Receptores da Colecistocinina/metabolismo , Acetamidas/farmacologia , Junções Aderentes/efeitos dos fármacos , Junções Aderentes/metabolismo , Animais , Benzodiazepinonas/farmacologia , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Adesão Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Cães , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Gastrinas/farmacologia , Humanos , Rim/citologia , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Compostos de Fenilureia/farmacologia , Fosfatidilinositol 3-Quinases/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Colecistocinina B , Receptores da Colecistocinina/antagonistas & inibidores , Receptores da Colecistocinina/efeitos dos fármacos , Receptores da Colecistocinina/genética , Transfecção , Vinculina/efeitos dos fármacos , Vinculina/metabolismo , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
7.
Oncogene ; 22(38): 6081-9, 2003 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-12955087

RESUMO

The cholecystokinin 2 receptor (CCK2R) increases proliferation of normal and neoplastic gastrointestinal cells and activates various mitogenic signaling pathways when stimulated by gastrin. To study the incidence of permanent activation of this receptor in tumorigenicity, a constitutively active mutant was generated by replacing residue Glu151 in the conserved E/DRY motif by Ala. Expression of the E151A-CCK2R mutant in NIH-3T3 cells causes ligand-independent activation of phospholipase C and ornithine decarboxylase, two enzymes critical for mitogenesis. Strikingly, the constitutive activity of this mutant was associated with dramatic alteration of NIH-3T3 cell morphology, enhanced cell proliferation and invasion. Moreover, injection of cells expressing E151A-CCK2R in nude mice resulted in the development of large and rapidly growing tumors. By contrast, none of these effects was observed with cells expressing the wild-type CCK2R, indicating that the tumorigenic properties of the E151A-CCK2R mutant is the result of its constitutive activation. To date, this is the first report that provides evidence for the high tumorigenic effect of a constitutively active CCK2R mutant, thus raising a potential role of the CCK2R in human cancer.


Assuntos
Transformação Celular Neoplásica/genética , Mutação Puntual , Receptores da Colecistocinina/genética , Receptores da Colecistocinina/metabolismo , Células 3T3/patologia , Alanina/genética , Motivos de Aminoácidos , Animais , Testes de Carcinogenicidade , Divisão Celular/genética , Ativação Enzimática , Feminino , Ácido Glutâmico/genética , Humanos , Camundongos , Camundongos Nus , Ornitina Descarboxilase/metabolismo , Receptor de Colecistocinina B , Fosfolipases Tipo C/metabolismo
8.
Hum Gene Ther ; 16(10): 1175-93, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16218779

RESUMO

Pancreatic cancer is one of the most aggressive and devastating human malignancies. The present study was conducted to determine whether in vivo sst2 gene transfer into human pancreatic tumors would impair tumor progression, and to characterize sst2 antitumoral bystander mechanisms. sst2 administration, using the synthetic vector PEI, strongly inhibited tumor progression of human pancreatic adenocarcinoma, in vivo. sst2 gene transfer induced intratumoral production of its ligand somatostatin. Disruption of this autocrine loop by RNA interference completely reversed sst2 antitumoral activity. Mice depleted of natural killer (NK) cells did not hamper sst2 tumor growth inhibition. However, microvessel density and vascular endothelial growth factor (VEGF) expression were markedly reduced in sst2-transfected tumors, whereas sst3 somatostatin receptor was upregulated. Depleting somatostatin by RNA interference completely abolished the sst2 inhibitory effect on VEGF expression and tumor angiogenesis, and sst2-induced sst3 expression in peripheral tumor vessels. We conclude that in vivo sst2 gene transfer elicited intratumoral somatostatin production and strongly impaired human pancreatic tumor growth. NK cells were not involved in this antitumoral bystander effect. VEGF and tumor vascularization were identified as novel targets for sst2-mediated antitumoral bystander effect. sst3 somatostatin receptor was upregulated in sst2-transfected tumors. Therefore, in vivo gene delivery of sst2 receptor to target the angiogenic process in pancreatic ductal adenocarcinoma might be a new therapeutic approach for treatment of pancreatic cancer in patients with unresectable disease.


Assuntos
Efeito Espectador , Carcinoma , Terapia Genética , Transplante de Neoplasias , Neoplasias Pancreáticas , Receptores de Somatostatina/metabolismo , Animais , Comunicação Autócrina/genética , Efeito Espectador/genética , Carcinoma/irrigação sanguínea , Carcinoma/metabolismo , Carcinoma/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Camundongos , Transplante de Neoplasias/métodos , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Receptores de Somatostatina/genética
9.
Trends Pharmacol Sci ; 24(1): 36-40, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12498729

RESUMO

Molecular modelling is of major help to understand structure-function data on G-protein-coupled receptors (GPCRs). Since the first determination of the structure of rhodopsin, at high resolution, the view has emerged that it will be now easy to automatically obtain realistic models for any GPCR by homology modeling. Our experience on cholecystokinin CCK(1) receptor modelling together with available data on other GPCRs leads us to rule out this opinion. We believe that construction of realistic models of certain GPCRs still remains time-consuming and requires many refinements of the models in close association with experiments. This conclusion has important consequences for modelling orphan GPCRs.


Assuntos
Proteínas de Ligação ao GTP/química , Modelos Moleculares , Receptores de Superfície Celular/química , Rodopsina/química , Animais , Cristalização , Humanos
10.
J Med Chem ; 48(1): 180-91, 2005 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-15634012

RESUMO

The Cholecystokinin-1 receptor (CCK1R) mediates actions of CCK in areas of the central nervous system and of the gut. It is a potential target to treat a number of diseases. As for all G-protein-coupled receptors, docking of ligands into modeled CCK1R binding site should greatly help to understand intrinsic mechanisms of activation. Here, we describe the procedure we used to progressively build a structural model for the CCK1R, to integrated, and on the basis of site-directed mutagenesis data on its binding site. Reliability of the CCK1R model was confirmed by interaction networks that involved conserved and functionally crucial motifs in G-protein-coupled receptors, such as Glu/Asp-Arg-Tyr and Asn-Pro-Xaa-Xaa-Tyr motifs. In addition, the 3-D structure of CCK1R-bound CCK resembled that determined by NMR in a lipid environment. The derived computational model was also used for revealing binding modes of several nonpeptide ligands and for rationalizing ligand structure-activity relationships known from experiments. Our findings indeed support that our "validated CCK1R model" could be used to study the intrinsic mechanism of CCK1R activation and design new ligands.


Assuntos
Modelos Moleculares , Receptor de Colecistocinina A/química , Receptor de Colecistocinina A/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Benzodiazepinonas/química , Benzodiazepinonas/metabolismo , Sítios de Ligação , Células COS , Chlorocebus aethiops , Colecistocinina/química , Colecistocinina/metabolismo , Devazepida/química , Devazepida/metabolismo , Humanos , Ácidos Indolacéticos/química , Ácidos Indolacéticos/metabolismo , Indóis/química , Indóis/metabolismo , Ligantes , Metabolismo dos Lipídeos , Lipídeos/química , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Compostos de Fenilureia/química , Compostos de Fenilureia/metabolismo , Conformação Proteica , Receptor de Colecistocinina A/agonistas , Receptor de Colecistocinina A/genética , Rodopsina/química , Rodopsina/metabolismo , Homologia de Sequência de Aminoácidos , Tiazóis/química , Tiazóis/metabolismo
11.
Pharmacogenetics ; 12(1): 23-30, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11773861

RESUMO

Cholecystokinin (CCK) and gastrin (G) and their receptors (CCK1 and CCK2) are involved in multiple physiological functions. Notably, CCK1R plays a role in the regulation of food intake whereas both CCK1R and CCK2R play a role in the regulation of pancreatic endocrine function. CCK1R and CCK2R may therefore serve as pharmacological targets in diabetes and obesity and genes encoding these receptors may be candidate genes in the pathogenesis of the diseases. In this study, we used single nucleotide polymorphism analysis and allele specific amplification for mutation screening of the CCK2 receptor gene and family linkage study. Mutated receptors were constructed, expressed in COS-7 cells for analysis of their binding and functional properties. V125I-CCK2 receptor variant was found in 2 out of 18 type 2 diabetes mellitus families tested. V125I mutation co-segregated in those 2 initial families, but further association studies showed that this mutation was not associated with diabetes or early age at diagnosis of the disease. V125I-CCK2 receptor high affinity sites exhibited a 2-fold enhanced binding affinity for CCK which was correlated to a slightly increased potency in coupling to inositol phosphate production. Since CCK2 receptor is expressed in pancreatic glucagon-producing cells in humans and is involved in secretion of glucagon, an increase of binding affinity of the mutated CCK2 receptor could enhance glucagon secretion in patients bearing V125I mutation. We also characterized a mutant of the CCK1 receptor which was previously identified in an obese patient. This mutant, V365I-CCK1, demonstrated a decreased level of expression (26%) and efficacy (25%) to stimulate inositol phosphates. It can therefore be expected that in humans bearing V365I mutation, decreases in CCK1 receptor expression and coupling efficiency may affect CCK-induced regulation of satiety. Polymorphism or mutations in the CCK receptors may be involved in type 2 diabetes mellitus and obesity. However, further studies are necessary to precisely evaluate this role in humans.


Assuntos
Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus/genética , Ligação Genética/genética , Obesidade , Polimorfismo de Nucleotídeo Único/genética , Receptores da Colecistocinina/genética , Sequência de Aminoácidos , Sítios de Ligação , Primers do DNA/química , Diabetes Mellitus/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Gastrinas/genética , Frequência do Gene , Genótipo , Humanos , Fosfatos de Inositol/metabolismo , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Receptor de Colecistocinina A , Receptor de Colecistocinina B , Receptores da Colecistocinina/metabolismo
12.
Clin Endocrinol (Oxf) ; 64(1): 63-7, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16402930

RESUMO

OBJECTIVE: The distinction between benign and malignant well-differentiated endocrine tumours is hard to achieve. The aim of the present study was to determine whether detection of telomerase or quantification of human telomerase reverse transcriptase protein subunit (hTERT) differ between benign and malignant endocrine tumours. PATIENTS AND METHODS: This retrospective study investigated 31 well-differentiated primary endocrine tumours. Based on clinical and histopathological criteria, tumours were categorized with the most recent WHO classification as 'benign' (n = 14), 'uncertain' (n = 5) or 'malignant' (n = 12) with (n = 7) or without (n = 5) metastasis after a mean follow-up of 40.4 +/- 25.8 months (4-122 months). All these tumours were assayed for telomerase activity and hTERT mRNA expression [real-time quantitative reverse transcriptase-polymerase chain reaction (RT-PCR)]. RESULTS: Telomerase activity was detected in 7 malignant and metastatic tumours, in 1 malignant tumour without metastases, in 1 uncertain tumour and in 1 benign tumour. hTERT mRNA levels were significantly higher in malignant endocrine tumours with or without metastases (P = 0.001) when compared to benign tumours. The negative predictive value of hTERT mRNA quantification for the diagnosis of malignancy was 88.9%, whereas the positive predictive value was 68.7%. CONCLUSION: The presence of telomerase activity within the primary endocrine tumour might indicate a malignant tumour and might suggest the need for an attentive search for concomitant metastases. Quantification of hTERT mRNA could be used in clinical practice to exclude malignancy in most endocrine tumours.


Assuntos
Biomarcadores Tumorais/análise , Ensaios Enzimáticos Clínicos , Proteínas de Ligação a DNA/análise , Neoplasias das Glândulas Endócrinas/diagnóstico , Telomerase/análise , Neoplasias das Glândulas Suprarrenais/diagnóstico , Adulto , Idoso , Tumor Carcinoide/diagnóstico , Feminino , Gastrinoma/diagnóstico , Glucagonoma/diagnóstico , Humanos , Insulinoma/diagnóstico , Neoplasias Intestinais/diagnóstico , Neoplasias Pulmonares/diagnóstico , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/diagnóstico , Feocromocitoma/diagnóstico , Valor Preditivo dos Testes , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Supressora de Tumor p53/análise
13.
Mol Ther ; 14(6): 758-67, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17000136

RESUMO

Excepting surgical resection, there is no efficient treatment against pancreatic cancer. The chemotherapeutic agent gemcitabine improves the patient's clinical status but survival is not prolonged. The aim of this study was to design a new strategy to render gemcitabine more efficient in the treatment of pancreatic cancer using gene therapy. We have generated a fusion gene (DCK::UMK) combining deoxycytidine kinase (DCK) and uridine monophosphate kinase (UMK), which converts gemcitabine into its toxic phosphorylated metabolite. Antitumor effects of DCK::UMK gene expression were tested in vitro and in vivo in an orthotopic transplantable model of pancreatic cancer established in hamsters. DCK::UMK sensitizes pancreatic cancer cells to gemcitabine by reducing dramatically both in vitro cell viability and in vivo tumor volume. We found that in vivo expression of DCK::UMK resulted in an antitumor bystander effect due to apoptosis of untransduced cells. In vivo intratumoral gene transfer of DCK::UMK using the synthetic carrier PEI induced a potent tumor regression. Taken together, the results show that the fusion gene DCK::UMK sensitizes pancreatic cancer cells to gemcitabine treatment to induce cell death by apoptosis and tumor regression. Intratumoral delivery of the DCK::UMK gene in combination with gemcitabine might be of high interest for pancreatic cancer management.


Assuntos
Desoxicitidina Quinase/genética , Desoxicitidina/análogos & derivados , Terapia Genética/métodos , Núcleosídeo-Fosfato Quinase/genética , Neoplasias Pancreáticas/terapia , Animais , Antimetabólitos Antineoplásicos/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Desoxicitidina/metabolismo , Desoxicitidina/farmacologia , Desoxicitidina Quinase/metabolismo , Vetores Genéticos/genética , Masculino , Mesocricetus , Neoplasias Experimentais/terapia , Núcleosídeo-Fosfato Quinase/metabolismo , Neoplasias Pancreáticas/patologia , Gencitabina
14.
J Biol Chem ; 281(28): 19156-71, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16690617

RESUMO

Somatostatin receptor SST5 is an inhibitory G protein-coupled receptor that exerts a strong cytostatic effect on various cell types. We reported previously that the SST5 anti-proliferative effect results in the inhibition of mitogen-induced increases in intracellular cGMP levels and MAPK activity. This study was conducted to define the early molecular events accountable for the SST5-mediated anti-proliferative effect. Here, we demonstrate that, in Chinese hamster ovary cells expressing SST5 (CHO/SST5 cells), somatostatin inhibited cell proliferation induced by nitric oxide donors and overexpression of the neuronal nitric-oxide synthase (nNOS) protein isoform. Accordingly, nNOS activity and dimerization were strongly inhibited following SST5 activation by the somatostatin analog RC-160. In CHO/SST5 cells, nNOS was dynamically recruited by the SST5 receptor and phosphorylated at tyrosyl residues following RC-160 treatment. RC-160 induced SST5-p60(src) kinase complex formation and subsequent p60(src) kinase activation. Coexpression of an inactive p60(src) kinase mutant with SST5 blocked RC-160-induced nNOS phosphorylation and inactivation and prevented the SST5-mediated anti-proliferative effect. In CHO/SST5 cells, p60(src) kinase associated with nNOS to induce its inactivation by phosphorylation at tyrosyl residues following RC-160 treatment. Using recombinant proteins, we demonstrated that such phosphorylation prevented nNOS homodimerization. Next, surface plasmon resonance and mutation analysis revealed that p60(src) directly associated with nNOS phosphorylated Tyr604. SST5-mediated inhibition of nNOS activity was demonstrated to be essential to the RC-160 anti-proliferative effect on pancreatic endocrine tumor-derived cells. We therefore identified nNOS as a new p60(src) kinase substrate essential for SST5-mediated anti-proliferative action.


Assuntos
Regulação Enzimológica da Expressão Gênica , Óxido Nítrico Sintase Tipo I/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Animais , Células CHO , Linhagem Celular Tumoral , Proliferação de Células , Cricetinae , Dimerização , Humanos , Fosforilação , Isoformas de Proteínas , Proteínas Proto-Oncogênicas pp60(c-src)/química , Ratos , Proteínas Recombinantes
15.
EMBO J ; 25(17): 3943-54, 2006 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16917505

RESUMO

Phosphatidylinositol 3-kinase (PI3K) regulates many cellular functions including growth and survival, and its excessive activation is a hallmark of cancer. Somatostatin, acting through its G protein-coupled receptor (GPCR) sst2, has potent proapoptotic and anti-invasive activities on normal and cancer cells. Here, we report a novel mechanism for inhibiting PI3K activity. Somatostatin, acting through sst2, inhibits PI3K activity by disrupting a pre-existing complex comprising the sst2 receptor and the p85 PI3K regulatory subunit. Surface plasmon resonance and molecular modeling identified the phosphorylated-Y71 residue of a p85-binding pYXXM motif in the first sst2 intracellular loop, and p85 COOH-terminal SH2 as direct interacting domains. Somatostatin-mediated dissociation of this complex as well as p85 tyrosine dephosphorylation correlates with sst2 tyrosine dephosphorylation on the Y71 residue. Mutating sst2-Y71 disabled sst2 to interact with p85 and somatostatin to inhibit PI3K, consequently abrogating sst2's ability to suppress cell survival and tumor growth. These results provide the first demonstration of a physical interaction between a GPCR and p85, revealing a novel mechanism for negative regulation by ligand-activated GPCR of PI3K-dependent survival pathways, which may be an important molecular target for antineoplastic therapy.


Assuntos
Fosfatidilinositol 3-Quinases/fisiologia , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Ativação Enzimática , Feminino , Humanos , Camundongos , Camundongos Nus , Mutação , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Ligação Proteica , Receptores de Somatostatina/genética , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Transplante Heterólogo , Tirosina/metabolismo , Domínios de Homologia de src
16.
Int J Cancer ; 114(6): 863-9, 2005 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-15609298

RESUMO

Tumor cells frequently synthesize an N-terminally extended the FGF-2 isoform of 24 kDa devoid of signal peptide but that contains a functional nuclear localization sequence (NLS). Although the signaling pathways elicited by secreted FGF-2 are well described, the molecular mechanisms involved in the growth promoting action of nuclearized 24 kDa FGF-2 remain unknown. The cancer cell line AR4-2J was engineered to stably express only the 24 kDa FGF-2 isoform and cDNA microarrays were used to identify targets implicated in the intracrine-induced cell proliferation. Levels of 27 transcripts were found either upregulated or downregulated compared to control cells. Among the 18 upregulated genes was c-jun, which is often involved in cell proliferation. Real-time PCR and Western blot analyses confirmed c-jun induction at both mRNA and protein levels. The c-jun antisense oligonucleotide strategy pointed out the involvement of c-Jun in the 24 kDa FGF-2-induced cell proliferation. The mitogenic effect was found to depend on ERK pathway and not on phosphoinositide 3-kinase, p38 MAPK, c-Jun NH2-terminal kinase signal transducers. In addition, the MEK inhibitor PD98059 reduced the 24 kDa FGF-2-dependent c-Jun level. These data show that intracrine FGF-2-mediated regulation of cell growth involves ERK activation and consequent c-Jun expression. Thus, despite its incapacity to be secreted, the intracellular-localized 24 kDa FGF-2 can activate a growth-related signaling pathway normally elicited by cell surface receptors.


Assuntos
Proliferação de Células , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas Proto-Oncogênicas c-jun/farmacologia , Animais , Western Blotting , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso , Neoplasias Pancreáticas/patologia , Reação em Cadeia da Polimerase , Isoformas de Proteínas , Ratos , Retroviridae/genética , Transdução de Sinais , Transfecção , Células Tumorais Cultivadas
17.
Int J Cancer ; 115(1): 46-54, 2005 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-15688412

RESUMO

In humans, initial events of pancreatic carcinogenesis remain unknown, and the question of whether this cancer, which has a ductal phenotype, exclusively arises from duct cells has been raised. Previous studies have demonstrated that transgenic expression of the CCK2 receptor in acinar cells of ElasCCK2 mice plays a role in the development of pancreatic neoplasia. The aim of our study was to examine initial steps of carcinogenesis in ElasCCK2 mice, adding a supplementary defect by using a chemical carcinogen, azaserine. Results of posttreatment sequential immunohistochemical examinations and quantifications demonstrate that mice responded to azaserine. Transition of acinar cells into duct-like cells expressing Pdx1 and gastrin, as well as proliferation of acinar cells, were transiently observed in both transgenic and control mice. The carcinogen also induced formation of preneoplastic lesions, adenomas, exhibiting properties of autonomous growth. Importantly, expression of the CCK2 receptor increased the susceptibility of pancreas to azaserine. Indeed, treated ElasCCK2 mice exhibited larger areas of pancreatic acinar-ductal transition, increased cellular proliferation as well as larger adenomas areas vs. control mice. These amplified responses may be related to auto/paracrine stimulation of CCK2 receptor by gastrin expressed in newly formed duct-like cells. Our results demonstrate that activation of CCK2 receptor and azaserine result in cumulative effects to favor the emergence of a risk situation that is a potential site for initiation of carcinogenesis.


Assuntos
Carcinoma de Células Acinares/etiologia , Carcinoma de Células Acinares/genética , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/genética , Receptor de Colecistocinina B/genética , Receptor de Colecistocinina B/fisiologia , Transgenes , Adenoma/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Azasserina/química , Azasserina/farmacologia , Bromodesoxiuridina/farmacologia , Carcinógenos , Carcinoma de Células Acinares/induzido quimicamente , Proliferação de Células , Corantes/farmacologia , Proteínas de Homeodomínio/metabolismo , Homozigoto , Imuno-Histoquímica , Inflamação , Linfócitos/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/induzido quimicamente , Fenótipo , Lesões Pré-Cancerosas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Risco , Fatores de Tempo , Transativadores/metabolismo
18.
J Biol Chem ; 280(9): 7976-84, 2005 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15611055

RESUMO

The peptide hormone gastrin is secreted from G cells of the gastric antrum and is the main inducer of gastric acid secretion via activation of its receptor the cholecystokinin 2 (CCK2) receptor. Both gastrin and CCK2 receptors are also transiently detected in the fetal pancreas and believed to exert growth/differentiation effects during endocrine pancreatic development. We demonstrated previously that whereas gastrin expression is extinguished in adult pancreas, CCK2 receptors are present in human glucagon-producing cells where their activation stimulates glucagon secretion. Based on these findings, we investigate in the present study whether gastrin regulates glucagon gene expression. To this aim, the CCK2 receptor was stably expressed into a glucagon-producing pancreatic islet cell line, and a glucagon-reporter fusion gene was transiently transfected in this new cellular model. We report that gastrin stimulates glucagon gene expression in glucagon-producing pancreatic cells. By using progressively 5'-increased sequences of the glucagon gene, gastrin responsiveness was located within the minimal promoter. Moreover, we clearly identified early growth response protein 1 (Egr-1) as an essential transcription factor interacting with the islet cell-specific G4 element. Egr-1 was shown to be essential for basal and gastrin-dependent glucagon gene transactivation. Furthermore, our results demonstrate that the MEK1/ERK1/2 pathway couples the CCK2 receptor to nuclearization and DNA binding of Egr-1. In conclusion, our data provide new information concerning the transcriptional regulation of the glucagon gene. Moreover they open new working hypothesis with reference to a potential role of gastrin in glucagon-producing pancreatic cells.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Gastrinas/metabolismo , Glucagon/metabolismo , Proteínas Imediatamente Precoces/fisiologia , Ilhotas Pancreáticas/metabolismo , Fatores de Transcrição/fisiologia , Ativação Transcricional , Animais , Sequência de Bases , Northern Blotting , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Cloranfenicol O-Acetiltransferase/metabolismo , Imunoprecipitação da Cromatina , Cricetinae , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteína 1 de Resposta de Crescimento Precoce , Regulação da Expressão Gênica , Glucagon/química , Glucagon/genética , Humanos , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/metabolismo , Receptor de Colecistocinina B/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transfecção
19.
J Biol Chem ; 280(11): 10710-5, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15640156

RESUMO

To date very few G protein-coupled receptors (GPCRs) have been shown to be connected to the Janus kinase (JAK)/STAT pathway. Thus our understanding of the mechanisms involved in the activation of this signaling pathway by GPCRs remains limited. In addition, little is known about the role of the JAK pathway in the physiological or pathophysiological functions of GPCRs. Here, we described a new mechanism of JAK activation that involves Galpha(q) proteins. Indeed, transfection of a constitutively activated mutant of Galpha(q) (Q209L) in COS-7 cells demonstrated that Galpha(q) is able to associate and activate JAK2. In addition, we showed that this mechanism is used to activate JAK2 by a GPCR principally coupled to G(q), the CCK2 receptor (CCK2R), and involves a highly conserved sequence in GPCRs, the NPXXY motif. In a pancreatic tumor cell line expressing the endogenous CCK2R, we demonstrated the activation of the JAK2/STAT3 pathway by this receptor and the involvement of this signaling pathway in the proliferative effects of the CCK2R. In addition, we showed in vivo that the targeted CCK2R expression in pancreas of Elas-CCK2 mice leads to the activation of JAK2 and STAT3. This process may contribute to the increase of pancreas growth as well as the formation of preneoplastic lesions leading to pancreatic tumor development observed in these transgenic animals.


Assuntos
Proteínas Tirosina Quinases/química , Proteínas Proto-Oncogênicas/química , Receptor de Colecistocinina B/química , Receptor de Colecistocinina B/metabolismo , Motivos de Aminoácidos , Animais , Western Blotting , Células COS , Linhagem Celular , Proliferação de Células , Meios de Cultura Livres de Soro/farmacologia , DNA Complementar/metabolismo , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Homozigoto , Imuno-Histoquímica , Inositol 1,4,5-Trifosfato/química , Janus Quinase 2 , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Mutação , Pâncreas/metabolismo , Ligação Proteica , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Transcrição STAT3 , Transdução de Sinais , Fatores de Tempo , Transativadores/metabolismo , Transfecção
20.
J Biol Chem ; 280(11): 10664-74, 2005 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-15632187

RESUMO

The cholecystokinin-1 receptor (CCK1R) is a G protein-coupled receptor (GPCR) that regulates important physiological functions. As for other GPCRs, the molecular basis of full and partial agonism is still far from clearly understood. In the present report, using both laboratory experiments and molecular modeling approaches, we have investigated the partial agonism mechanism of JMV 180, on the human CCK1R. We first showed that efficacy of the CCK1R to activate phospholipase C is dependent on the correct orientation of the C-terminal end of peptidic ligands toward residue Phe(330) of helix VI. We have previously reported that a single mutation of Met(121) (helix III) markedly reduced the receptor-mediated inositol phosphate production upon stimulation by CCK. Computational simulations predicted that residue 121 affected orientation of the C-terminal end of CCK, thus suggesting that the molecular complex with a reduced inositol phosphate production observed with the mutated CCK1R resembles that resulting from binding of JMV 180 to the WT-CCK1R. Pharmacological, biochemical, and functional characterizations of the two receptor.ligand complexes with decreased abilities to signal were carried out in different cell types. We found that they presented the same features, such as total dependence of inositol phosphate production to Galpha(q) expression, single affinity of binding sites, insensitivity of binding to non-hydrolyzable GTP, absence of GTPgamma[S(35)] binding following agonist stimulation, similarity of dose-response curves for amylase secretion, and incapacity to induce acute pancreatitis in pancreatic acini. We concluded that helices VI and III of the CCK1R are functionally linked through the CCK1R agonist binding site and that positioning of the C-terminal ends of peptidic agonists toward Phe(330) of helix VI is responsible for extent of phospholipase C activation through Galpha(q) coupling. Given the potential therapeutic interest of partial agonists such as JMV 180, our structural data will serve for target structure-based design of new CCK1R ligands.


Assuntos
Receptor de Colecistocinina A/agonistas , Receptor de Colecistocinina A/química , Adenoviridae/genética , Amilases/química , Animais , Arginina/química , Sítios de Ligação , Células COS , Membrana Celular/metabolismo , Ativação Enzimática , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/química , Humanos , Concentração Inibidora 50 , Fosfatos de Inositol/química , Cinética , Ligantes , Lisina/química , Substâncias Macromoleculares/química , Masculino , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Pâncreas/citologia , Peptídeos/química , Fosfatos/química , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Software , Fosfolipases Tipo C/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA