Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Cancer ; 106(2): 348-57, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22146521

RESUMO

BACKGROUND: Pancreatic cancer is a deadly disease characterised by high incidence of TP53 mutations. Restoration of TP53 function is perceived as a highly attractive therapeutic strategy, whose effects are not well characterised. METHODS: The current work adapted an inducible strategy of stage-specific reexpression of wild-type (wt) TP53 in an in vivo orthotopic mouse model of pancreatic cancer. RESULTS: The reconstitution of wt TP53 function in TP53-mutant DanG and MiaPaCa-2 cells caused G1 cell cycle arrest but no evidence of apoptosis induction. Consistent with subcutaneous xenograft models, we found that wt TP53 reduced primary tumour growth. Wt TP53 reexpression during early tumour growth led to significant increase in vascularisation. This correlated with an unexpectedly high rate of micro-metastases in lymph nodes of animals with wt TP53 induction, despite the 90% decrease in median primary tumour weight. Reexpression of wt TP53 later in tumour development did not significantly affect the number of CD31-reactive vessels, but increased lymphatic vessel density. CONCLUSION: The increased number of lymphatic vessels and micro-metastases suggests that wt TP53 induction complexly affected the biology of different tumour constituents of pancreatic cancer. Our observation suggests that combination of the inducible system with an orthotopic model can yield important insights into in vivo pancreatic cancer biology.


Assuntos
Genes p53 , Linfangiogênese/genética , Neoplasias Pancreáticas/patologia , Prolina/genética , Animais , Sequência de Bases , Western Blotting , Linhagem Celular Tumoral , Primers do DNA , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica , Neoplasias Pancreáticas/irrigação sanguínea , Neoplasias Pancreáticas/genética , Reação em Cadeia da Polimerase
2.
Cell Death Differ ; 18(5): 806-16, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21113146

RESUMO

Anoikis resistance is a hallmark of transformed epithelial cells. Here, we show that treatment of anoikis-resistant carcinoma cell lines with the endogenous lectin galectin-1 (Gal-1) promoted apoptosis via interaction with the unligated fibronectin receptor α(5)ß(1)-integrin. Gal-1 efficiency correlated with expression of α(5)ß(1)-integrin, and transfection of the α(5)-subunit into deficient cell lines conferred Gal-1 binding and anoikis stimulation. Furthermore, Gal-1 and the α(5)- and ß(1)-integrin subunits co-precipitated in Gal-1-stimulated cells undergoing anoikis. Other members of the galectin family failed to be active. The functional interaction between Gal-1 and α(5)ß(1)-integrin was glycan dependent with α2,6-sialylation representing a switch-off signal. Desialylation of cell surface glycans resulted in increased electrophoretic mobility of α(5)ß(1)-integrin and facilitated Gal-1 binding and anoikis stimulation. On the level of signaling, Gal-1-stimulated anoikis was prevented by filipin, which impaired the internalization of α(5)ß(1)-integrin via cholesterol-enriched microdomains, and by pretreatment with a caspase-8 inhibitor. We propose that Gal-1/α(5)ß(1)-integrin interaction participates in the control of epithelial integrity and integrin sialylation may enable carcinoma cells to evade this Gal-1-dependent control mechanism.


Assuntos
Anoikis , Caspase 8/metabolismo , Galectina 1/fisiologia , Integrina alfa5beta1/metabolismo , Neoplasias/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Galectina 1/farmacologia , Galectinas/farmacologia , Humanos , Imunoprecipitação , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Neoplasias/patologia , Neuraminidase/metabolismo , Oligossacarídeos/metabolismo , Ligação Proteica , Receptores de Fibronectina/metabolismo
3.
Gut ; 58(2): 261-70, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18829975

RESUMO

BACKGROUND AND AIMS: Current systemic therapies for neuroendocrine tumours (NETs) do not provide sufficient control of tumour growth. However, efficient evaluation of novel drugs is hindered by the lack of a suitable preclinical animal model. Here an orthotopic mouse model of pancreatic NET is established and used to study the action of ZK 304709, a first in class, oral multitarget tumour growth inhibitor. ZK 304709 is an inhibitor of cyclin-dependent kinases (Cdks) 1, 2, 4, 7 and 9, vascular endothelial growth factor receptor-type kinases (VEGF-RTKs) 1-3 and platelet-derived growth factor receptor-type kinase beta (PDGF-RTKss). METHODS: BON and QGP-1 human NET cells were used to study proliferation, survival and cell cycle distribution in vitro. For induction of orthotopic NETs, BON cells were injected into the pancreas of NMRI(nu/nu) mice. Primary tumour growth and metastatic spread were recorded after 9 weeks, and apoptosis, microvessel density and lymphatic vessel density were determined. RESULTS: ZK 304709 dose-dependently suppressed proliferation and colony formation of NET cells. Direct effects on NET cells were consistent with Cdk inhibition and involved G(2) cell cycle arrest and apoptosis induction, which was associated with reduced expression of MCL1 (myeloid cell leukaemia sequence 1), survivin and hypoxia-inducible factor 1alpha (HIF1alpha). Apoptosis similarly occurred in vivo in ZK 304709-treated orthotopic BON tumours, resulting in a 80% reduction of primary tumour growth. In contrast, treatment with lanreotide or 5-fluorouracil and streptozotocin failed to inhibit tumour gowth. ZK 304709 also reduced tumour microvessel density, implicating antiangiogenic mechanisms. CONCLUSION: BON orthotopic tumours provide an informative model for preclinical drug evaluation in NETs. In this model, ZK 304709 achieved efficacious tumour growth control via induction of apoptosis and inhibition of tumour-induced angiogenesis.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Tumores Neuroendócrinos/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Núcleo Celular/química , Relação Dose-Resposta a Droga , Feminino , Citometria de Fluxo , Imunofluorescência , Histocitoquímica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Proteínas Inibidoras de Apoptose , Metástase Linfática , Camundongos , Camundongos Nus , Proteínas Associadas aos Microtúbulos/análise , Neovascularização Patológica/tratamento farmacológico , Tumores Neuroendócrinos/irrigação sanguínea , Neoplasias Pancreáticas/irrigação sanguínea , Survivina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
4.
Br J Cancer ; 99(1): 110-7, 2008 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-18577984

RESUMO

Functional inactivation of the tumour suppressor protein p16(INK4a) constitutes a key event in the multistep process of pancreatic ductal cell transformation. However, the significance of p16 inactivation for complex and tissue-specific aspects of pancreatic cancer progression, such as angiogenesis and metastasis, is less understood. Here, we inducibly re-expressed p16 in vivo in an orthotopic model of pancreatic cancer and examined the impact on these clinically relevant aspects of pancreatic cancer tumour biology. Consistent with previous work in subcutaneous xenograft models, we found p16 capable of reducing primary tumour growth. In addition, p16 restitution resulted in a marked reduction of tumour angiogenesis, largely accounted for by a p16-dependent inhibition of lymphangiogenesis. In excellent agreement with the antilymphangiogenic effect, re-expression of p16 almost completely prevented lymph node metastases of MiaPaca-2 pancreatic tumours. To our knowledge, this is the first report that experimentally links the tumour suppressor p16 to the process of lymphangiogenesis.


Assuntos
Carcinoma Ductal Pancreático/genética , Genes p16/fisiologia , Linfangiogênese/genética , Metástase Linfática/genética , Neoplasias Pancreáticas/genética , Animais , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Humanos , Camundongos , Camundongos Nus
5.
Biomed Pharmacother ; 60(6): 269-72, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16887322

RESUMO

Loss of cell cycle control and tumor-induced neovascularization are major drivers of human tumor growth. The multi-target tumor growth inhibitor ZK 304709 is a nanomolar inhibitor of cyclin-dependent kinases 1, 2, 4, 7 and 9, as well as vascular endothelial growth factor receptor tyrosine kinase 1-3 and of platelet-derived growth factor receptor beta tyrosine kinase. The multi-targeted mode of action of ZK 304709 acting on cell cycle and angiogenesis resulted in superior efficacy compared to standard chemotherapeutic compounds both in s.c. human tumor xenografts as well as orthotopic human pancreatic carcinoma models.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Pirimidinas/farmacologia , Sulfonamidas/farmacologia , Inibidores da Angiogênese/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores do Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptores de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores , Transplante Heterólogo
6.
Int J Oncol ; 21(5): 1133-40, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12370765

RESUMO

Although biotherapy of gastroenteropancreatic neuroendocrine tumors (NET) provides excellent control for the hypersecretion syndrome, tumor regression is rarely observed, implying the need for novel antiproliferative strategies. Here, we demonstrate that human pancreatic QGP-1 NET cells express functionally intact interferon-gamma (IFN-gamma) receptors and downstream effectors, including the putative tumor suppressor interferon regulatory factor-1 (IRF-1). IFN-gamma treatment profoundly inhibited anchorage-dependent and anchorage-independent growth of QGP-1 cells. Concomitant with the onset of growth inhibition, apoptotic cells were detected in cell cycle analyses of IFN-gamma treated cultures. Apoptosis was confirmed by evaluation of DNA fragmentation and PARP cleavage. Immunoblots of IFN-gamma treated QGP-1 cells revealed a substantial upregulation of caspase-1, followed by the appearance of active proteolytic fragments of caspase-3, suggesting that autocatalytic activation of caspase-1 might initiate the caspase cascade. Apoptosis induction by IFN-gamma was also observed in two of four primary cultures established from tumors of patients with for- and midgut NETs, respectively. Taken together our results characterize IFN-gamma as a potent proapoptotic stimulus in a subset of gastrointestinal NETs and suggest an IRF-1 mediated induction of caspase-1 as a relevant underlying mechanism. Based on these results, the potential of IFN-gamma in experimental biotherapeutic treatment of NETs can be further explored.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Neuroendócrino/tratamento farmacológico , Interferon gama/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Carcinoma Neuroendócrino/patologia , Caspase 1/metabolismo , Caspase 3 , Caspases/metabolismo , Proteínas de Ligação a DNA/biossíntese , Ativação Enzimática , Humanos , Fator Regulador 1 de Interferon , Neoplasias Pancreáticas/patologia , Fosfoproteínas/biossíntese , Poli(ADP-Ribose) Polimerases/metabolismo , Células Tumorais Cultivadas , Regulação para Cima
7.
Gut ; 49(2): 251-62, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11454803

RESUMO

BACKGROUND AND AIMS: The poor prognosis of pancreatic cancer is partly due to resistance to a broad spectrum of apoptotic stimuli. To identify intact proapoptotic pathways of potential clinical relevance, we characterised the effects of interferon gamma (IFN-gamma) on growth and survival in human pancreatic cancer cells. METHODS: IFN-gamma receptor expression and signal transduction were examined by reverse transcriptase-polymerase chain reaction (RT-PCR), immunoprecipitation, western blot analysis, and transactivation assays. Effects on cell growth and survival were evaluated in terms of cell numbers, colony formation, cell cycle analysis, DNA fragmentation, and poly(ADP ribose) polymerase (PARP) cleavage. RESULTS: All four pancreatic cancer cell lines examined expressed functional IFN-gamma receptors and downstream effectors, including the putative tumour suppressor interferon regulatory factor 1 (IRF-1). IFN-gamma treatment profoundly inhibited anchorage dependent and independent growth of pancreatic cancer cells. Cell cycle analyses revealed subdiploid cells suggesting apoptosis, which was confirmed by demonstration of DNA fragmentation and PARP cleavage. Time and dose dependency of apoptosis induction and growth inhibition correlated closely, identifying apoptosis as the main, if not exclusive, mechanism responsible for growth inhibition. Apoptosis was preceded by upregulation of procaspase-1 and accompanied by proteolytic activation. Furthermore, the caspase inhibitor z-vad-fmk completely prevented IFN-gamma mediated apoptosis. CONCLUSIONS: These results identify an intact proapoptotic pathway in pancreatic cancer cells and suggest that IRF-1 and/or procaspase-1 may represent potential therapeutic targets to be further explored.


Assuntos
Apoptose/fisiologia , Caspase 1/fisiologia , Interferon gama/uso terapêutico , Neoplasias Pancreáticas/tratamento farmacológico , Análise de Variância , Western Blotting , Contagem de Células , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fragmentação do DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Humanos , Neoplasias Pancreáticas/patologia , Testes de Precipitina , Receptores de Interferon/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia , Ativação Transcricional/fisiologia , Células Tumorais Cultivadas/efeitos dos fármacos , Regulação para Cima
8.
Hepatology ; 33(2): 346-56, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11172336

RESUMO

The potential antiproliferative effects of interferon-alpha (IFN-alpha) in the treatment of hepatocellular carcinoma (HCC) are controversial, and the growth inhibitory mechanisms remain poorly understood. Therefore, the current study was designed to delineate the molecular mechanisms responsible for direct antiproliferative actions of IFN-alpha in HCC cells. IFN-alpha receptor expression and signal transduction were examined by RT-PCR, immunoprecipitation, Western analysis, and transient transactivation assays. Effects of IFN-alpha on cell growth and cell-cycle distribution were evaluated based on cell numbers and flow cytometry. Composition and activity of cyclin-dependent kinase complexes were determined by immunoblotting and histone-H1-kinase assays. Expression of IFN-alpha receptors was found in all 3 HCC cell lines. IFN-alpha binding initiated phosphorylation of Jak1 and Tyk2 kinases leading to Stat1/Stat2 activation, nuclear translocation, and transactivation of an ISRE-luciferase reporter gene construct. IFN-alpha treatment resulted in a time- and dose-dependent reduction of proliferation. Cell cycle analysis of G1-synchronized, IFN-alpha-treated HCC cells revealed a substantial delay in S-phase progression but no alteration of G1/S-phase transition or evidence of apoptotic cell death. Reflecting the time course of S-phase accumulation, cell cycle-dependent induction of Cyclin A and Cyclin B was impaired, resulting in reduced activity of Cdk2 and Cdc2 kinases. Furthermore, Cdc25C was selectively down-regulated. IFN-alpha treatment inhibits growth of HCC cells by specifically delaying S-phase progression, most likely because of inhibition of Cyclin A induction, resulting in decreased activity of the associated Cdk2 and Cdc2 kinases.


Assuntos
Antineoplásicos/farmacologia , Quinases relacionadas a CDC2 e CDC28 , Carcinoma Hepatocelular/patologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Interferon-alfa/farmacologia , Neoplasias Hepáticas/patologia , Fase S/efeitos dos fármacos , Proteína Quinase CDC2/antagonistas & inibidores , Ciclo Celular/efeitos dos fármacos , Quinase 2 Dependente de Ciclina , DNA/genética , Humanos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptor de Interferon alfa e beta , Receptores de Interferon/fisiologia , Transdução de Sinais , Ativação Transcricional , Células Tumorais Cultivadas
9.
J Cell Biol ; 150(6): 1467-78, 2000 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-10995450

RESUMO

The tumor suppressor gene p16(INK4a) inhibits the kinase activity of the cyclin-dependent kinase 4-6/cyclin D complexes and subsequent phosphorylation of critical substrates necessary for transit through the G1 phase of the cell cycle. Recent studies suggested that control of the G1/S boundary might not be the sole biological function of p16(INK4a). We hypothesized that p16(INK4a) might influence hitherto unknown critical features of a malignant epithelial phenotype, such as anchorage dependence. Here we provide evidence that stable transfection of p16(INK4a) restitutes apoptosis induction upon loss of anchorage (anoikis) in a variety of human cancer cells. Anoikis in p16(INK4a)-transfected cells was evidenced by DNA fragmentation and poly(ADP-ribose) polymerase cleavage upon cultivation on polyhydroxyethylmethacrylate-coated dishes and was associated with suppression of anchorage-independent growth as well as complete loss of tumorigenicity. p16(INK4a)-mediated anoikis was due to selective transcriptional upregulation of the alpha(5) integrin chain of the alpha(5)beta(1) fibronectin receptor as detected by FACS((R)) analysis, immunoprecipitation, Northern blotting, and nuclear run-on assays. Addition of soluble fibronectin and inhibitory alpha(5) antibodies to nonadherent cells completely abolished p16(INK4a)-mediated anoikis, whereas laminin was ineffective. Furthermore, antisense-induced downregulation of the alpha(5) integrin chain in p16(INK4a)-transfected cells restored resistance to anoikis. These data suggest a novel functional interference between a cell cycle-regulating tumor suppressor gene and membrane-bound integrins, thus regulating a hallmark feature of an epithelial transformed phenotype: susceptibility to anoikis.


Assuntos
Apoptose/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Genes Supressores de Tumor/fisiologia , Proteínas Proto-Oncogênicas , Receptores de Fibronectina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Divisão Celular/fisiologia , Quinase 4 Dependente de Ciclina , Inibidor p16 de Quinase Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Regulação para Baixo/fisiologia , Fibronectinas/farmacologia , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas In Vitro , Rim/citologia , Fígado/citologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas , Fenótipo , Pele/citologia , Transfecção , Células Tumorais Cultivadas , Regulação para Cima/fisiologia
10.
J Cell Sci ; 113 ( Pt 17): 3025-35, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10934041

RESUMO

We have analyzed human pancreatic cancer cells to explore the growth regulatory function of protein kinase C (PKC)alpha. PKCalpha subcellular redistribution, activation kinetics and downregulation were examined in detail and correlated to immediate and delayed effects on cell-cycle regulatory pathways. TPA treatment resulted in transient PKC(&agr;) activation accompanied by translocation of the enzyme into membrane and nuclear compartments, and was followed by subsequent downregulation. TPA-induced inhibition of DNA synthesis was prevented by a PKC-antagonist and was reproduced by microinjection of recombinant PKCalpha, indicating that activation of this isoenzyme was required and sufficient for growth inhibitory effects. PKC(&agr;) activation arrested cells in the G(1) phase of the cell cycle as a consequence of selective inhibition of cyclin dependent kinase (CDK)2 activity with concomitant hypophosphorylation of Rb. The inhibition of CDK2 activity resulted from induction of p21(cip1) cyclin-dependent kinase inhibitors. Levels of p21(cip1) remained elevated and CDK2 activity repressed in spite of PKCalpha downregulation, indicating that downstream effectors of PKCalpha are the primary determinants for the duration of PKC-mediated growth inhibition. The PKCalpha-induced block in cell proliferation persisted even though cells were kept in the presence of growth factors, suggesting that induction of PKCalpha results in a permanent withdrawal of pancreatic cancer cells from the cell cycle.


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Ciclo Celular , Ciclinas/metabolismo , Isoenzimas/fisiologia , Neoplasias Pancreáticas/metabolismo , Proteína Quinase C/fisiologia , Apoptose , Transporte Biológico/efeitos dos fármacos , Divisão Celular , Núcleo Celular/metabolismo , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p21 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Replicação do DNA , Humanos , Isoenzimas/efeitos dos fármacos , Isoenzimas/genética , Isoenzimas/metabolismo , Neoplasias Pancreáticas/enzimologia , Fosforilação , Proteína Quinase C/efeitos dos fármacos , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteína Quinase C-alfa , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Proteína do Retinoblastoma/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Células Tumorais Cultivadas
11.
Gastroenterology ; 118(4): 735-48, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10734025

RESUMO

BACKGROUND & AIMS: Although human neuroendocrine tumors respond to interferon (IFN)-alpha treatment in vivo, the underlying mechanisms of growth inhibition are poorly understood. To characterize the antiproliferative effects at a molecular level, we explored the growth-regulatory action of IFN-alpha in the human neuroendocrine tumor cell lines BON and QGP1. METHODS: IFN-alpha receptor expression and signal transduction were examined by reverse-transcription polymerase chain reaction, immunoblotting, subcellular fractionation, and transactivation assays. Growth regulation was evaluated by cell numbers, soft agar assays, and cell cycle analysis using flow cytometry. Expression and activity of cell cycle-regulatory molecules were determined by immunoblotting and histone H1-kinase assays. RESULTS: Both cell lines expressed IFN-alpha receptor mRNA transcripts. Ligand binding initiated phosphorylation of Jak kinases and Stat transcription factors, resulting in Stat activation, nuclear translocation, and transcription from an ISRE-reporter construct. Prolonged IFN-alpha treatment dose-dependently inhibited both anchorage-dependent and -independent growth. Cell cycle analysis of IFN-alpha-treated, unsynchronized cultures revealed an increased S-phase population, which was further substantiated in G(1) synchronized QGP1 cells. IFN-alpha-treated cells entered S phase in parallel to control cultures, but their progress into G(2)/M phase was delayed. Both cellular cyclin B levels and CDC 2 activity were substantially reduced. The extent and time course of this reduction corresponded to the observed S-phase accumulation. CONCLUSIONS: IFN-alpha directly inhibits growth of human neuroendocrine tumor cells by specifically delaying progression through S phase and into G(2)/M. These cell cycle changes are associated with inhibition of cyclin B expression, resulting in reduced CDC2 activity.


Assuntos
Antineoplásicos/farmacologia , Interferon-alfa/farmacologia , Tumores Neuroendócrinos/patologia , Proteína Quinase CDC2/antagonistas & inibidores , Divisão Celular/efeitos dos fármacos , Ciclina B/antagonistas & inibidores , Humanos , Interferon-alfa/metabolismo , RNA Mensageiro/metabolismo , Receptores de Interferon/genética , Receptores de Interferon/metabolismo , Fase S , Células Tumorais Cultivadas
13.
J Biol Chem ; 274(31): 21701-6, 1999 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-10419481

RESUMO

Activation of G(q) protein-coupled receptors can either stimulate or inhibit cell growth. Previously, these opposite effects were explained by differences in the cell models. Here we show that activation of m3 muscarinic acetylcholine receptors ectopically expressed in NIH3T3 cells can cause stimulation and inhibition of growth in the same cell. A clonal cell line was selected from cells that formed foci agonist dependently (3T3/m3 cells). In quiescent 3T3/m3 cells, carbachol stimulated DNA synthesis. In contrast, when 3T3/m3 cells were growing, either due to the presence of serum or after transformation with oncogenic v-src, carbachol inhibited growth. This inhibition was not due to reduction of extracellular signal-regulated kinase activity because carbachol induced extracellular signal-regulated kinase phosphorylation in both quiescent and growing 3T3/m3 cells. Investigating the cell cycle mechanisms involved in growth inhibition, we found that carbachol treatment decreased cyclin D1 levels, increased p21(cip1) expression, and led to hypophosphorylation of the retinoblastoma gene product (Rb). Proteasome inhibitors blocked the carbachol-induced degradation of cyclin D1. Effects on p21(cip1) were blocked by a protein kinase C inhibitor. Thus, m3 muscarinic acetylcholine receptors couple to both growth-stimulatory and -inhibitory signaling pathways in NIH3T3 cells, and the observed effects of receptor activation depend on the context of cellular growth.


Assuntos
Carbacol/farmacologia , Divisão Celular/fisiologia , Proteínas Quinases Ativadas por Mitógeno , Receptores Muscarínicos/fisiologia , Células 3T3 , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica , Células Clonais , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , DNA/biossíntese , Genes src , Camundongos , Proteína Quinase 1 Ativada por Mitógeno , Proteína Quinase 3 Ativada por Mitógeno , N-Metilescopolamina/farmacocinética , Fosforilação , Receptor Muscarínico M3 , Receptores Muscarínicos/efeitos dos fármacos , Proteína do Retinoblastoma/metabolismo
14.
J Cell Sci ; 112 ( Pt 1): 137-46, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9841910

RESUMO

Cytokines are supposed to be mediators in diarrhoeal diseases. The aim of this study is to characterize the effect of tumor necrosis factor-alpha (TNFalpha) on epithelial barrier function in the colonic epithelial cell line HT-29/B6. Active ion transport and barrier function were measured as short-circuit current and transepithelial electrical resistance (Rt), respectively. In parallel, freeze-fracture electron microscopy (EM) of tight junctions (TJ) and immunofluorescence microscopy of the zonula occludens protein-1 (ZO-1) were performed. Serosal addition of TNF(alpha) (100 ng/ml) decreased Rt by 81%. This effect was dose-dependent and could be mimicked by antibodies against the p55 form of the TNF receptor. Cytotoxic effects were excluded by a negative lactate dehydrogenase (LDH) assay. Immunofluorescence localization with anti-ZO-1 antibodies revealed no evidence for disruption of the monolayer after TNFalpha treatment. In freeze-fracture EM, TJ complexity was decreased by TNFalpha, as indicated by a decrease in the number of strands from 4.7 to 3.4. The tyrosine kinase blocker genistein and the protein kinase A inhibitor H-8 reduced the effect of TNFalpha. A combination of TNFalpha with interferon-gamma acted synergistically on the epithelial barrier. In conclusion, TNFalpha impairs epithelial barrier function by altering structure and function of the tight junction, which could be of pathogenic relevance in intestinal inflammation.


Assuntos
Mucosa Intestinal/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Apoptose , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Impedância Elétrica , Inibidores Enzimáticos/farmacologia , Técnica de Fratura por Congelamento , Células HT29 , Humanos , Imuno-Histoquímica , Interferon gama/farmacologia , Mucosa Intestinal/citologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/ultraestrutura , L-Lactato Desidrogenase/metabolismo , Manitol/metabolismo , Microscopia Eletrônica , Fosfotransferases/antagonistas & inibidores , Fosfotransferases/fisiologia , Receptores do Fator de Necrose Tumoral/imunologia , Sódio/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores
15.
Br J Cancer ; 78(10): 1288-95, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9823968

RESUMO

During carcinogenesis, pancreatic acinar cells can dedifferentiate into ductal adenocarcinoma of the pancreas. DSL-6A/C1 cells represent an in vitro model of this carcinogenic sequence. This study was designed to examine the effects of retinoids on cell growth in DSL-6A/C1 cells and to characterize further the molecular mechanisms underlying the antiproliferative actions of retinoids. Treatment of DSL-6A/C1 cells with retinoids results in a time- and dose-dependent inhibition of cell growth, paralleled by a retinoid-mediated transactivation of a pTK::betaRAREx2-luciferase reporter construct transiently transfected into DSL-6A/C1 cells. Retinoid receptor expression was evaluated by reverse transcriptase polymerase chain reaction (RT-PCR) using subtype-specific primers and demonstrated expression of retinoic acid receptor alpha (RAR-alpha), RAR-beta and retinoid X receptor alpha (RXR-alpha). Using a panel of receptor subtype-specific agonists, the RAR-alpha specific agonist Ro 40-6055 was the most potent retinoid in terms of growth inhibition. Furthermore, all-trans-retinoic acid-mediated growth inhibition and transactivation was completely blocked by the RAR-alpha-specific antagonist Ro 41-5253. In summary, the RAR-alpha subtype predominantly mediates the antiproliferative effects of retinoids in DSL-6A/C1 cells. Furthermore, this cell system provides a feasible tool to study the molecular mechanisms underlying the growth inhibitory effects of retinoids in ductal pancreatic carcinoma cells derived from a primary acinar cell phenotype.


Assuntos
Transformação Celular Neoplásica , Receptores do Ácido Retinoico/fisiologia , Retinoides/farmacologia , Animais , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Divisão Celular/efeitos dos fármacos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ratos , Receptores do Ácido Retinoico/biossíntese , Células Tumorais Cultivadas
16.
Am J Physiol ; 273(5): C1449-57, 1997 11.
Artigo em Inglês | MEDLINE | ID: mdl-9374628

RESUMO

Rat cholecystokinin-B (CCK-B) receptors were transfected into Chinese hamster ovary (CHO)-K1 (CHO-CCK-B) and Swiss 3T3 (Swiss 3T3-CCK-B) cells, and the effects of receptor activation on cell proliferation and intracellular signaling were investigated. CCK octapeptide (CCK-8) treatment had no effect on cell growth in quiescent CHO-CCK-B cells but inhibited DNA synthesis, proliferation, and colony formation when the cells were grown in fetal bovine serum (FBS). In contrast, CCK-8 stimulated DNA synthesis in quiescent Swiss 3T3-CCK-B cells and had no effect when the cells were grown in FBS. These differences in growth responses were not due to differences in the level of receptor expression, as similar numbers of receptors were present in both cell types. To determine whether the different growth effects were due to differences in receptor coupling to common second messenger pathways, we investigated the effects of CCK-8 on several known intracellular signals. In both cell types, CCK-8 stimulated increases in intracellular Ca2+ concentration and polyphosphoinositide hydrolysis with similar potencies and efficacies. CCK-8 also stimulated arachidonate release from both cell types, although the potency was higher in the CHO cells. Adenosine 3',5'-cyclic monophosphate generation was observed at high agonist concentrations in both cell types and was much greater in cells with higher receptor density. In summary, receptor activation had opposite effects on growth parameters in CHO and Swiss 3T3 cells, but only minor differences were observed in the characteristics of CCK-B receptor coupling to specific second messengers in the two cell types. Thus cellular context is a principal determinant of the biological effects of CCK-B receptor activation, and differences in biological responses may occur independently of major differences in receptor coupling.


Assuntos
Receptores da Colecistocinina/fisiologia , Sincalida/farmacologia , Células 3T3 , Animais , Ácido Araquidônico/metabolismo , Células CHO , Cálcio/metabolismo , Bovinos , Divisão Celular/efeitos dos fármacos , Cricetinae , AMP Cíclico/metabolismo , DNA/biossíntese , Cinética , Camundongos , Fosfatidilinositóis/metabolismo , Receptor de Colecistocinina B , Receptores da Colecistocinina/biossíntese , Proteínas Recombinantes/biossíntese , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Sistemas do Segundo Mensageiro/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Transfecção
17.
Gastroenterology ; 112(3): 952-9, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9041258

RESUMO

BACKGROUND & AIMS: Cholecystokinin (CCK) acting via CCK(A) receptors and gastrin acting via CCK(B) receptors exert trophic effects on a variety of nontransformed tissues. However, their role as hormonal regulators of pancreatic cancer is controversial. The aim of this study was to determine the effects of activation of CCK(A) and CCK(B) receptors on the growth of human pancreatic cancer cells in vitro. METHODS: Two human pancreatic cell lines MiaPaca-2 and Panc-1 were transfected stably with both CCK receptor subtypes. Effects of CCK on various growth parameters including DNA synthesis, nuclear labeling, and colony formation were evaluated. RESULTS: Cells expressing either receptor subtype, but not untransfected cells, bound ligand and mobilized Ca2+ in response to CCK. CCK treatment caused a sustained pronounced inhibition of anchorage-independent growth. Similarly, CCK treatment inhibited anchorage-dependent growth. Receptor activation caused a concentration and time-dependent reduction in [3H]thymidine incorporation and nuclear labeling in cells cultured anchored to a plastic substrate. However, these effects on anchorage-dependent growth were transient, suggesting cellular desensitization. CONCLUSIONS: These data indicate that both CCK receptor subtypes can mediate growth inhibitory responses in pancreatic cancer cell lines and raise the possibility that CCK exerts a predominant growth inhibitory action on human pancreatic cancer cells.


Assuntos
Neoplasias Pancreáticas/patologia , Receptores da Colecistocinina/fisiologia , Divisão Celular/efeitos dos fármacos , DNA/biossíntese , Relação Dose-Resposta a Droga , Humanos , Receptores da Colecistocinina/análise , Receptores da Colecistocinina/genética , Sincalida/farmacologia , Células Tumorais Cultivadas
18.
Digestion ; 58(4): 361-7, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9324163

RESUMO

It was recently found that cholecystokinin (CCK) activates mitogen-activated protein kinases (MAPK) in isolated rat pancreatic acini. The present study evaluates whether one or both types of CCK receptors are capable of MAPK activation in pancreatic AR42J acinar cells as well as CHO cells transfected with CCK-A or CCK-B receptors. CCK significantly increased p44 MAPK and p42 MAPK activities in AR42J cells. Minimal, half-maximal, and maximal responses were observed at 30 and 500 pM and 10 nM, respectively, after CCK-8 stimulation and at 100 pM and 1.5 and 30 nM, respectively, after gastrin stimulation. Glycine-extended gastrin had no effect at 100 nM and a small but significant effect at 1 microM. The CCK-B receptor antagonist L365,260 almost totally blocked MAPK activation in AR42J cells after stimulation with gastrin and glycine-extended gastrin and substantially reduced the activation of both kinases by CCK-8, while the CCK-A receptor antagonist L364,718 was much less effective. The CCK-A-selective agonist A71376, however, was an effective stimulant of MAPK activity. In an alternative approach, stably transfected CHO cells bearing either CCK-A or CCK-B receptors were stimulated with CCK-8. Each receptor induced a time-dependent increase in activity of both MAPKs by five- to sixfold in CCK-A- and CCK-B-bearing cells. In conclusion, both CCK-A and CCK-B receptors activate MAPK in AR42J cells and in transfected CHO cells.


Assuntos
MAP Quinase Quinase Quinases , Compostos de Fenilureia , Proteínas Quinases/metabolismo , Receptores da Colecistocinina/fisiologia , Animais , Benzodiazepinonas/farmacologia , Western Blotting , Células CHO/efeitos dos fármacos , Células CHO/metabolismo , Carcinoma de Células Acinares/fisiopatologia , Células Cultivadas , Cricetinae , Devazepida , Relação Dose-Resposta a Droga , Gastrinas/farmacologia , MAP Quinase Quinase Quinase 4 , Oligopeptídeos/farmacologia , Neoplasias Pancreáticas/fisiopatologia , Proteínas Quinases/efeitos dos fármacos , Ratos , Receptor de Colecistocinina A , Receptor de Colecistocinina B , Receptores da Colecistocinina/antagonistas & inibidores , Receptores da Colecistocinina/efeitos dos fármacos , Valores de Referência , Sincalida/farmacologia , Transfecção , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo
19.
Proc Natl Acad Sci U S A ; 92(24): 10929-33, 1995 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-7479912

RESUMO

Cellular desensitization is believed to be important for growth control but direct evidence is lacking. In the current study we compared effects of wild-type and down-regulation-resistant mutant m3 muscarinic receptors on Chinese hamster ovary (CHO-K1) cell desensitization, proliferation, and transformation. We found that down-regulation of m3 muscarinic acetylcholine receptors was the principal mechanism of desensitization of receptor-activated inositol phosphate phospholipid hydrolysis in these cells. Activation of wild-type and mutant receptors inhibited anchorage-independent growth as assayed by colony formation in agar. However, the potency for inhibition of anchorage-independent growth was greater for cells expressing the mutant receptor. Activation of either receptor also initially inhibited anchorage-dependent cell proliferation in randomly growing populations. Rates of DNA synthesis and cell division were profoundly reduced by carbachol in cells expressing either receptor at early time points. Analysis of cell cycle parameters indicated that cell cycle progression was inhibited at transitions from G1 to S and G2/M to G1 phases. However, mutant receptor effects on anchorage-dependent growth were sustained, whereas wild-type receptor effects were transient. Thus, receptor down-regulation restored cell cycle progression. In contrast, activation of either receptor blocked entry into the cell cycle from quiescence, and this response was not reduced by receptor down-regulation. Therefore, activation of m3 muscarinic acetylcholine receptors inhibited CHO cell anchorage-dependent and -independent growth. In anchored cells carbachol inhibited the cell cycle at three distinct points. Inhibitions at two of these points were eliminated by wild-type receptor down-regulation while the other was not. These results directly demonstrate that desensitization mechanisms can act as principal determinants of cellular growth responses.


Assuntos
Ciclo Celular/efeitos dos fármacos , Receptores Muscarínicos/metabolismo , Animais , Células CHO , Carbacol/farmacologia , Cricetinae , DNA/biossíntese , Regulação para Baixo , Agonistas Muscarínicos/farmacologia , Fosfatidilinositóis/metabolismo , Transdução de Sinais
20.
J Biol Chem ; 270(32): 18858-64, 1995 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-7642540

RESUMO

To investigate the role of the carboxy terminus in the regulation of the bombesin (BN) receptor, we constructed two chimeric receptors with carboxyl termini transferred from either m3 muscarinic cholinergic (m3 ACh) (BMC) or cholecystokinin A (CCKA) (BCC) receptors and expressed them in Chinese hamster ovary cells. Previous studies showed that agonist treatment caused rapid internalization of CCKA but not m3 ACh receptors in these cells. In the current study we conducted separate analyses of ligand and receptor internalization and analyzed receptor recycling. Ligand internalization was assessed using acid washing. BN and CCKA receptors internalized ligand with 80 +/- 3 and 85 +/- 7% in an acid-resistant compartment at equilibrium. Ligand internalization of chimeric receptors generally assumed the properties of the donor receptors. Thus, BCC receptors internalized ligand to a similar extent as wild-type CCKA receptors (75 +/- 3%), whereas, BMC receptors showed reduced ligand internalization (38 +/- 1%). Receptor internalization was more directly assessed by determining agonist-induced loss of surface binding. BN and CCKA receptors were largely internalized (56 +/- 8 and 50 +/- 7%, respectively). BCC receptors were also extensively internalized (82 +/- 3%). In contrast, BMC receptors were minimally internalized (22 +/- 8%). Receptor recycling was assessed as recovery from agonist induced loss of binding. BN, CCKA, and BMC receptors showed rapid recycling. In contrast, BCC receptors did not recycle. These data indicate that carboxyl-terminal structures determine both internalization of ligand-receptor complexes and subsequent receptor recycling.


Assuntos
Receptores da Bombesina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células CHO , Cricetinae , Dados de Sequência Molecular , Receptores da Bombesina/química , Receptores da Colecistocinina/metabolismo , Receptores Muscarínicos/metabolismo , Proteínas Recombinantes de Fusão/química , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...