Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
1.
Poult Sci ; 96(12): 4409-4418, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29053815

RESUMEN

In mammals, insulin regulates blood glucose levels and plays a key regulatory role in appetite via the hypothalamus. In contrast, chickens are characterized by atypical glucose homeostasis, with relatively high blood glucose levels, reduced glucose sensitivity of pancreatic beta cells, and large resistance to exogenous insulin. The aim of the present study was to investigate in chickens the effects of 5 h fasting and 5 h insulin immuno-neutralization on hypothalamic mRNA levels of 23 genes associated with food intake, energy balance, and glucose metabolism. We observed that insulin immune-neutralization by administration of anti-porcine insulin guinea pig serum (AI) significantly decreased food intake and increased plasma glucose levels in chickens, while 5 h fasting produced a limited and non-significant reduction in plasma glucose. In addition, 5 h fasting increased levels of NPY, TAS1R1, DIO2, LEPR, GLUT1, GLUT3, GLUT8, and GCK mRNA. In contrast, AI had no impact on the levels of any selected mRNA. Therefore, our results demonstrate that in chickens, food intake inhibition or satiety mechanisms induced by insulin immuno-neutralization do not rely on hypothalamic abundance of the 23 transcripts analyzed. The hypothalamic transcripts that were increased in the fasted group are likely components of a mechanism of adaptation to fasting in chickens.


Asunto(s)
Pollos/fisiología , Ingestión de Alimentos/fisiología , Ayuno/fisiología , Expresión Génica , Insulina/metabolismo , Animales , Pollos/genética , Hipotálamo/metabolismo , Insulina/deficiencia , ARN Mensajero/genética , ARN Mensajero/metabolismo
2.
Leukemia ; 28(2): 338-48, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23689514

RESUMEN

Extranodal, nasal-type natural killer (NK)/T-cell lymphoma (NKCL) is an aggressive malignancy with poor prognosis in which, usually, signal transducer and activator of transcription 3 (STAT3) is constitutively activated and oncogenic. Here, we demonstrate that STAT3 activation mostly results from constitutive Janus kinase (JAK)3 phosphorylation on tyrosine 980, as observed in three of the four tested NKCL cell lines and in 20 of the 23 NKCL tumor samples under study. In one of the cell lines and in 4 of 19 (21%) NKCL primary tumor samples, constitutive JAK3 activation was related to an acquired mutation (A573V or V722I) in the JAK3 pseudokinase domain. We then show that constitutive activation of the JAK3/STAT3 pathway has a major role in NKCL cell growth and survival and in the invasive phenotype. Indeed, NKCL cell growth was slowed down in vitro by targeting JAK3 with chemical inhibitors or small-interfering RNAs. In a human NKCL xenograft mouse model, tumor growth was significantly delayed by the JAK3 inhibitor CP-690550. Altogether, the constitutive activation of JAK3, which can result from JAK3-activating mutations, is a frequent feature of NKCL that deserves to be tested as a therapeutic target.


Asunto(s)
Janus Quinasa 3/genética , Linfoma Extranodal de Células NK-T/genética , Linfoma Extranodal de Células NK-T/patología , Mutación , Adulto , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Janus Quinasa 3/antagonistas & inhibidores , Janus Quinasa 3/metabolismo , Linfoma Extranodal de Células NK-T/tratamiento farmacológico , Linfoma Extranodal de Células NK-T/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Fosforilación , Piperidinas/administración & dosificación , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Pirroles/administración & dosificación , Pirroles/farmacología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Gastroenterol Clin Biol ; 34(12): 653-61, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21051167

RESUMEN

The fine-tuning between cell proliferation and differentiation of self-renewing stem cells and pluripotent progenitors in gastric glands and colon epithelial crypts is coordinated by the mechanisms that regulate colon epithelial cell migration and guidance along the crypt axis. This leads to the acquisition of specialized cellular functions and the exfoliation of desquamated senescent and apoptotic epithelial cells at the apical mucosa interface with the gut lumen. Self-renewing stem cells and pluripotent progenitors are involved in the clonal and polyclonal growth of digestive tumors. Several lines of evidence support the existence of a subpopulation of cancer cells with stem cell-like (SCL) phenotypes in solid tumors of breast and digestive system. Consistently, epithelial cancer cell lines in long-term culture are phenotypically and functionally heterogeneous. It is suggested that only a small proportion of transformed cells are clonogenic in vivo and ex vivo to form colonies and to initiate tumor growth in immunodeficient mice. A discrete subpopulation of tumor -initiating SCL cancer cells are highly competent to survive, propagate and spread through the invasive and metastatic cascade. A better understanding of the mechanisms driving the plasticity and pluripotency of stem cells, their derived progenitors and SCL colon cancer initiating cells during tumor progression will open new avenues for the early detection and treatment of local and distant tumors of the digestive tract.


Asunto(s)
Neoplasias del Colon/etiología , Neoplasias del Colon/patología , Células Madre , Animales , Humanos , Células Madre Neoplásicas , Células Madre/fisiología
4.
Oncogene ; 27(29): 4024-33, 2008 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-18317448

RESUMEN

From the conditioned medium of the human colon carcinoma cells, HT-29 5M21 (CM-5M21), expressing a spontaneous invasive phenotype, tumor-associated trypsin inhibitor (TATI) was identified and characterized by proteomics, cDNA microarray approaches and functional analyses. Both CM-5M21 and recombinant TATI, but not the K18Y-TATI mutant at the protease inhibitor site, trigger collagen type I invasion by several human adenoma and carcinoma cells of the colon and breast, through phosphoinositide-3-kinase, protein kinase C and Rho-GTPases/Rho kinase-dependent pathways. Conversely, the proinvasive action of TATI in parental HT29 cells was alleviated by the TATI antibody PSKAN2 and the K18Y-TATI mutant. Stable expression of K18Y-TATI in HT-29 5M21 cells downregulated tumor growth, angiogenesis and the expression of several metastasis-related genes, including CSPG4 (13.8-fold), BMP-7 (9.7-fold), the BMP antagonist CHORDIN (5.2-fold), IGFBP-2 and IGF2 (9.6- and 4.6-fold). Accordingly, ectopic expression of KY-TATI inhibited the development of lung metastases from HT-29 5M21 tumor xenografts in immunodeficient mice. These findings identify TATI as an autocrine transforming factor potentially involved in early and late events of colon cancer progression, including local invasion of the primary tumor and its metastatic spread. Targeting TATI, its molecular partners and effectors may bring novel therapeutic applications for high-grade human solid tumors in the digestive and urogenital systems.


Asunto(s)
Comunicación Autocrina , Neoplasias del Colon/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Inhibidor de Tripsina Pancreática de Kazal/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Anticuerpos Antineoplásicos/farmacología , Comunicación Autocrina/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones SCID , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Inhibidor de Tripsina Pancreática de Kazal/genética , Inhibidor de Tripsina Pancreática de Kazal/farmacología
5.
Oncogene ; 26(41): 6071-81, 2007 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-17667937

RESUMEN

The recently identified subfamily of WNK protein kinases is characterized by a unique sequence variation in the catalytic domain and four related human WNK genes were identified. Here, we describe the cloning and functional analysis of the human family member WNK2. We show that the depletion of endogenous WNK2 expression by RNA interference in human cervical HeLa cancer cells led to the activation of the extracellular signal-regulated kinase (ERK)1/2 mitogen-activated protein kinases but, in contrast to the depletion of WNK1, had no effect on ERK5. Furthermore, expression of a kinase-dead WNK2-K207M mutant also activated ERK1/2 suggesting that WNK2 catalytic activity is required. Depletion of WNK2 expression increased G1/S progression and potentiated the cellular response to low epidermal growth factor concentrations. The molecular mechanism of ERK1/2 activation in WNK2-depleted cells lies downstream of the Raf kinases and involves MEK1 phosphorylation at serine 298 in both HeLa and HT29 colon cancer cells. This modification is linked to the upregulation of MEK1 activity toward ERK1/2. Together, these results provide evidence that WNK2 is involved in the modulation of growth factor-induced cancer cell proliferation through the MEK1/ERK1/2 pathway. The data identify WNK2 as a candidate tumor suppressor gene and suggest a coordinated activity of WNK kinases in the regulation of cell proliferation.


Asunto(s)
División Celular/efectos de los fármacos , MAP Quinasa Quinasa 1/metabolismo , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Empalme Alternativo , Clonación Molecular , Replicación del ADN , Activación Enzimática , Células HeLa , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/deficiencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
6.
Br J Surg ; 94(8): 1020-7, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17440950

RESUMEN

BACKGROUND: Hereditary non-polyposis colorectal cancer (HNPCC) arises mostly from germline mutations of the mismatch repair genes MSH2 and MLH1. The diagnosis of HNPCC is based on a set of clinical criteria that may be too restrictive to identify all affected patients. Immunohistochemical staining (IHC) for the mismatch repair proteins, MutS homologue 2 (MSH2) and MutL homologue 1 (MLH1), reliably identifies the microsatellite instability phenotype. This study evaluated the ability of IHC to detect germline mutations in an unselected group of patients with colorectal cancer (CRC). METHODS: All patients with CRC operated on between July 2000 and March 2003, and demonstrating a loss of protein, were contacted. Following informed consent, searchs for germline mutation and methylation of the promoter were performed on normal and tumoral DNA. RESULTS: Thirty patients agreed to participate, four of whom fulfilled the Amsterdam II criteria. Loss of expression of MLH1 was found in 20 patients, and loss of expression of MSH2 in ten patients. Four of the MLH1-deficient patients had a germline MLH1 point mutation (positive predictive value (PPV) 20 (95 per cent confidence interval (c.i.) 2 to 38 per cent) and 11 had promoter methylation. Seven of the MSH2-deficient patients had a germline MSH2 point mutation (PPV 70 (95 per cent c.i. 54 to 96 per cent), and none showed promoter methylation. CONCLUSION: MLH1-deficient patients who are young or have a positive family history of cancer should be referred for genetic testing and counselling, whereas MSH2-deficient patients should be counselled in the same way as patients with HNPCC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales Hereditarias sin Poliposis/diagnóstico , Reparación de la Incompatibilidad de ADN , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Nucleares/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Disparidad de Par Base/genética , Metilación de ADN , Femenino , Técnicas Genéticas , Humanos , Inmunohistoquímica/métodos , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Reacción en Cadena de la Polimerasa/métodos
7.
Oncogene ; 26(38): 5615-25, 2007 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-17334389

RESUMEN

Deleted in colon cancer (DCC) and UNC5 function as netrin dependence receptors by inducing apoptosis in the absence of their ligand and accordingly were recently designated as putative conditional tumor suppressors. Herein, we determined whether netrin-1 and its receptors are implicated in cancer cell invasion and tumor progression. Expression of DCC, UNC5 and adenosine A2B-receptors (A2B-Rs) was investigated by reverse transcription polymerase chain reaction in human colon cancer cells. The impact of DCC restitution and netrin-1 was evaluated on collagen type I invasion, tumor growth and metastasis in nude mice, cancer cell survival and gene expression profiling. Flow cytometry, poly(ADP-ribose)polymerase-1 and caspase-8 activation were used to evaluate the impact of DCC on cell death. Both netrin-1 and A2B-R activation induced the invasive phenotype through the Rho-Rho kinase axis in DCC-deficient human colorectal cancer cells. Restitution of wild-type DCC blocked invasion induced by netrin-1, A2B-R agonist and other agents. Ectopic expression of netrin-1 led to increased growth of human colon tumor xenografts in athymic mice. Conversely, introduction of wt-DCC in kidney MDCKts.src-ggl cells strongly inhibited metastasis in lymph nodes and lungs and increased sensitivity to apoptosis in hypoxia. DNA microarrays revealed that netrin and DCC had common and divergent impacts on gene expression linked to cell cycle, survival, surface signaling and adhesion. Our findings underscore that netrin is a potent invasion and tumor growth-promoting agent and that DCC is a metastasis suppressor gene targeting both proinvasive and survival pathways in a cumulative manner.


Asunto(s)
Neoplasias/patología , Factores de Crecimiento Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Western Blotting , Hipoxia de la Célula , Línea Celular Transformada , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/fisiología , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Receptor DCC , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factores de Crecimiento Nervioso/genética , Netrina-1 , Receptor de Adenosina A2B/genética , Receptor de Adenosina A2B/metabolismo , Receptores de Superficie Celular/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Trasplante Heterólogo , Proteínas Supresoras de Tumor/genética
8.
Oncogene ; 25(50): 6628-36, 2006 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-16715141

RESUMEN

TFF1 is overexpressed in inflammatory diseases and human cancers of the digestive and urogenital systems. To examine the transforming potential of TFF1 in human colon epithelial cells, premalignant PC/AA/C1 adenoma cells (PC) derived from a patient with familial adenomatous polyposis (FAP) were transformed by the TFF1 cDNA and used as a model of the adenoma-carcinoma transition. Constitutive expression of TFF1 increased anchorage-independent cell growth in soft agar, and induced or potentiated the growth of colon PC-TFF1 and kidney MDCKts.src-TFF1 tumor xenografts in athymic mice. This resulted in reduction of thapsigargin-induced apoptosis and promotion of collagen type I invasion through several oncogenic pathways. Using the differential display approach to identify TFF1 target genes, we found that the dual specific phosphatases Cdc25A and B implicated in cell cycle transitions are strongly upregulated under active forms in both PC-TFF1 and HCT8/S11-TFF1 colon cancer cells. Accordingly, TFF1 expression is absent in normal human colon crypts but is induced in correlation with Cdc25a and b transcript levels and tumor grade in familial and sporadic colon adenomas and carcinomas. We propose that TFF1 and Cdc25A-B cooperate with other dominant oncogenic pathways to induce the adenoma and adenocarcinoma transitions. Agents that target TFF1/Cdc25 signaling pathways may be useful for treating patients with TFF1-positive solid tumors.


Asunto(s)
Adenoma/patología , Carcinoma/patología , Proteínas de Ciclo Celular/metabolismo , Colon/citología , Neoplasias del Colon/patología , Mucosa Intestinal/metabolismo , Proteínas Supresoras de Tumor/fisiología , Fosfatasas cdc25/metabolismo , Adenoma/enzimología , Adenoma/metabolismo , Animales , Carcinoma/enzimología , Carcinoma/metabolismo , Adhesión Celular/genética , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Neoplasias del Colon/enzimología , Neoplasias del Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Progresión de la Enfermedad , Perros , Inducción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Fenotipo , Lesiones Precancerosas/patología , Transfección , Trasplante Heterólogo , Factor Trefoil-1 , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
9.
J Fr Ophtalmol ; 26(10): 1007-14, 2003 Dec.
Artículo en Francés | MEDLINE | ID: mdl-14691392

RESUMEN

PURPOSE: Trefoil factor family (TFF) peptides (formerly P-domain peptides; trefoil factor) are small (7-12 kDa) protease-resistant secreted peptides designated pS2 (or TFF1), SP (TFF2) and ITF (TFF3). Human conjunctival goblet cells (GCs) are known to synthesize TFF, but TFF expression by these cells has not been studied in pathological conditions. We quantified trefoil factor family (TFF) gene transcripts in pterygium, and we immunolocalized TFF protein. METHODS: Eleven pterygium specimens were studied, together with 19 biopsy specimens of normal human conjunctiva as controls. TFF1 (pS2), TFF2 (spasmolytic peptide) and TFF3 (intestinal trefoil factor) mRNA expression was semiquantified by means of reverse-transcription polymerase chain reaction amplification (RT-PCR). TFF1, TFF2 and TFF3 mRNA levels were determined individually, relative to beta2 microglobulin housekeeping gene mRNA (internal standard), by coamplification of the target fragments and beta2 microglobulin in the same tube. Five pterygia and five normal human conjunctival biopsy specimens were also analyzed for TFF1 and mucin (MUC5AC) protein expression by immunostaining with monoclonal antibodies. Anti-PS2 (Zymed Laboratories, San Francisco), a mouse monoclonal antibody (MAb) against the 30 C-terminal amino acids of human TFF1, and P2802 (provided by Doctor Marie-Christine Rio, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM, Strasbourg, France), a mouse MAb directed against a synthetic peptide corresponding to the last 28 amino acids of TFF1, were used at 1/20 dilution. A mouse monoclonal antibody directed against the peptidic core of gastric M1 mucin was used as previously described. M1 immunoreactivity is encoded by the MUC5AC gene. RESULTS: TFF1 and TFF3 mRNA was expressed in all normal conjunctival and pterygium specimens. TFF2 mRNA was not expressed by either sample type, but was expressed by the positive control (human stomach cDNA). TFF1 mRNA expression was stronger in pterygium than in controls (p=0.02). TFF3 mRNA expression was similar in the two sample types (p=0.89). TFF are coexpressed and act in concert with mucins to protect mucous epithelia and trigger wound-healing responses. Inflammation and ulceration of the gastrointestinal tract are associated with increased TFF expression. Conjunctival GCs secrete TFF in both pigs and humans. We found that TFF1 mRNA was overexpressed in pterygium relative to healthy conjunctiva, whereas the TFF1 immunostaining patterns were similar. TFF1 protein expression was confined to goblet cells. However, whereas all GCs were positive for MUC5AC, not all GC were labeled by anti-TFF1 mAbs in either normal conjunctiva or pterygium. The observed TFF1 mRNA overexpression in pterygium was not associated with abnormal TFF1 peptide localization. Increased MUC5AC protein expression would be expected in pterygium, because of increased GC density. Indeed, in conjunctival diseases such as dry-eye syndrome in which GC density is decreased, mucin secretion is also decreased. This could explain the increased expression of TFF1 mRNA in pterygium, although not all GCs expressed TFF1 protein. TFF proteins are copackaged within mucous cell granules; TFF1 preferentially colocalizes with MUC5AC, and TFF3 with MUC2. However, we found some cell granules containing MUC5AC but not TFF1. The proportion of TFF1-negative GCs was similar in pterygium and normal conjunctiva. The normal TFF3 mRNA expression in pterygium was unexpected and suggests that only GCs involved in TFF1 secretion are overrepresented in this pathological tissue. TFF2 mRNA was undetectable in both normal conjunctiva and pterygium, possibly because of its copackaging in mucous cell granules and its preferential cosecretion with MUC6, which is not expressed in the conjunctiva. CONCLUSION: As in normal conjunctiva, the TFF1 and TFF3 genes are expressed by conjunctival goblet cells in pterygium, contrary to the TFF2 gene. Only TFF1 gene expression was elevated in pterygium compared to normal conjunctiva.


Asunto(s)
Mucinas/genética , Proteínas Musculares/genética , Neuropéptidos , Péptidos/genética , Pterigion/genética , Adulto , Anciano , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Mucinas/análisis , Proteínas Musculares/análisis , Péptidos/análisis , Pterigion/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor Trefoil-2 , Factor Trefoil-3
10.
Oncogene ; 21(44): 6740-50, 2002 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-12360401

RESUMEN

Bile acids are implicated in colorectal carcinogenesis as evidenced by epidemiological and experimental studies. We examined whether bile acids stimulate cellular invasion of human colorectal and dog kidney epithelial cells at different stages of tumor progression. Colon PC/AA/C1, PCmsrc, and HCT-8/E11 cells and kidney MDCKT23 cells were seeded on top of collagen type I gels and invasive cells were counted after 24 h incubation. Activation of the Rac1 and RhoA small GTPases was investigated by pull-down assays. Haptotaxis was analysed with modified Boyden chambers. Lithocholic acid, chenodeoxycholic acid, cholic acid and deoxycholic acid stimulated cellular invasion of SRC- and RhoA-transformed PCmsrc and MDCKT23-RhoAV14 cells, and of HCT-8/E11 cells originating from a sporadic tumor, but were ineffective in premalignant PC/AA/C1 and MDCKT23 cells. Bile acid-stimulated invasion occurred through stimulation of haptotaxis and was dependent on the RhoA/Rho-kinase pathway and signaling cascades using protein kinase C, mitogen-activated protein kinase, and cyclooxygenase-2. Accordingly, BA-induced invasion was associated with activation of the Rac1 and RhoA GTPases and expression of the farnesoid X receptor. We conclude that bile acids stimulate invasion and haptotaxis in colorectal cancer cells via several cancer invasion signaling pathways.


Asunto(s)
Ácidos y Sales Biliares/farmacología , Neoplasias Colorrectales/patología , Genes src/fisiología , Proteína de Unión al GTP rhoA/fisiología , Ciclooxigenasa 2 , Relación Dosis-Respuesta a Droga , Guanosina Trifosfato/metabolismo , Humanos , Integrina beta1/fisiología , Isoenzimas/fisiología , Proteínas de la Membrana , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/fisiología , Lesiones Precancerosas/patología , Prostaglandina-Endoperóxido Sintasas/fisiología , Células Tumorales Cultivadas
12.
Bull Cancer ; 88 Spec No: S26-34, 2001 Aug.
Artículo en Francés | MEDLINE | ID: mdl-11567911

RESUMEN

Therapeutic innovation in oncology often requires the optimization of combinations with active drugs based on in vitro and in vivo data. This is exemplified by oxaliplatin for which several preclinical studies of combinations have been realized. Oxaliplatin has been combined with 5-fluoro-uracile, gemcitabine, topoisomerase I inhibitors, taxanes demonstrating synergy or additivity. Synergistic and additive effects are often due to the optimization in the use of distinct mechanism of action or resistance and might be associated with no overlapping toxicity when combined in clinical trials.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Área Bajo la Curva , Carboplatino/administración & dosificación , Cisplatino/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Interacciones Farmacológicas , Ensayos de Selección de Medicamentos Antitumorales , Fluorouracilo/administración & dosificación , Humanos , Ratones , Modelos Biológicos , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Células Tumorales Cultivadas/efectos de los fármacos , Gemcitabina
13.
Mutat Res ; 480-481: 359-69, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11506828

RESUMEN

Bile acids play a role in colorectal carcinogenesis as evidenced by epidemiological and experimental studies. Some bile acids stimulate growth of normal colonic and adenoma cells, but not of colorectal cancer cells. Moreover, bile acids stimulate invasion of colorectal cancer cells, at least in vitro. One possible mechanism of action is bile acid-induced DNA binding and transactivation of the activator protein-1 (AP-1) by co-operate activation of extracellular signal-regulated kinases (ERKs) and PKC signaling. In the present paper, we review the mechanisms by which bile acids influence carcinogenesis.


Asunto(s)
Ácidos y Sales Biliares/toxicidad , Transformación Celular Neoplásica/inducido químicamente , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/epidemiología , Animales , Ácidos y Sales Biliares/metabolismo , División Celular/efectos de los fármacos , ADN/metabolismo , Humanos , Invasividad Neoplásica , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional/efectos de los fármacos
14.
Mol Pharmacol ; 60(2): 363-72, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11455024

RESUMEN

It was shown previously that platelet-activating factor receptors (PAF-Rs) inhibit invasiveness of colonic and kidney epithelial cells induced by the src and Met oncogenes via a pertussis toxin-sensitive mechanism. Therefore, Madin-Darby canine kidney (MDCKts.src) cells were stably transfected with constitutively activated forms of Galphao, Galphai1, Galphai2, Galphai3 (AGalphao/i), two Gbetagamma sequestering proteins [C-terminal end of beta-adrenergic receptor kinase (ct-betaARK) and the Galphat subunit of retinal G-protein transducin], and Gbeta1-Ggamma2 subunits alone or in combination. Cellular invasion induced by src, Met, and leptin was abrogated by the AGalphao/i, ct-betaARK, and Galphat-positive clones, but was induced by coexpression of Gbeta1gamma2. In contrast, invasion stimulated by the trefoil factors (TFFs) pS2 and intestinal trefoil factor in MDCKts.src cells or human colonic epithelial cells PCmsrc and HCT8/S11 was insensitive to PAF, AGalphao, AGalphai1, and AGalphai2, but was abolished by AGalphai3 and the protease-activated receptor-1 (PAR-1) agonist thrombin receptor-activating peptide. Depletion of free Gbetagamma heterodimers by ct-betaARK resulted in a remarkable decrease of cellular adhesion and spreading on collagen matrix. Our data demonstrate the following: 1) PAF-Rs impair cellular invasion induced by src, Met, and leptin via the activation of Galphao and Galphai1 to -3; 2) invasion induced by TFFs is selectively inhibited by PAR-1 receptors and Galphai3 activation; and 3) Gbetagamma dimers are required as positive effectors of invasion pathways induced by oncogenes and epigenetic factors. Thus, redistribution of Galphao/Galphai and Gbeta/gamma heterotrimeric G-proteins by PAF-R and PAR-1 exert differential functions on positive and negative signaling pathways involved in cellular invasion and may serve as potential targets for anticancer therapy.


Asunto(s)
Proteínas de Caenorhabditis elegans , Movimiento Celular/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/farmacología , Proteínas de Unión al GTP Heterotriméricas/farmacología , Proteínas Proto-Oncogénicas/farmacología , Receptores de Superficie Celular , Receptores Acoplados a Proteínas G , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Transformada , Células Cultivadas , Perros , Subunidad alfa de la Proteína de Unión al GTP Gi2 , Proteínas de Unión al GTP Heterotriméricas/biosíntesis , Ratones , Ratones Desnudos , Invasividad Neoplásica , Péptidos/síntesis química , Péptidos/química , Péptidos/farmacología , Toxina del Pertussis , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transfección , Factores de Virulencia de Bordetella/farmacología
15.
FASEB J ; 15(9): 1517-28, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11427483

RESUMEN

We have investigated the possible functional relationships between cellular invasion pathways induced by trefoil factors (TFFs), src, and the cyclooxygenases COX-1 and COX-2. Pharmacological inhibitors of the Rho small GTPase (C3 exoenzyme), phospholipase C (U-73122), cyclooxygenases (SC-560, NS-398), and the thromboxane A2 receptor (TXA2-R) antagonist SQ-295 completely abolished invasion induced by intestinal trefoil factor, pS2, and src in kidney and colonic epithelial cells MDCKts.src and PCmsrc. In contrast, invasion was induced by the TXA2-R mimetic U-46619, constitutively activated forms of the heterotrimeric G-proteins Galphaq (AGalphaq), Galpha12, Galpha13 (AGalpha12/13), which are signaling elements downstream of TXA2-R. Ectopic overexpression of pS2 cDNA and protein in MDCKts.src-pS2 cells and human colorectal cancer cells HCT8/S11-pS2 initiate distinct invasion signals that are Rho independent and COX and TXA2-R dependent. We detected a marked induction of COX-2 protein and accumulation of the stable PGH2/TXA2 metabolite TXB2 in the conditioned medium from cells transformed by src. This led to activation of the TXA2-R-dependent invasion pathway, which is monitored via a Rho- and Galpha12/Galpha13-independent mechanism using the Galphaq/PKC signaling cascade. These findings identify a new intracrine/paracrine loop that can be monitored by TFFs and src in inflammatory diseases and progression of colorectal cancers.


Asunto(s)
Sustancias de Crecimiento/farmacología , Mucinas , Proteínas Musculares , Neuropéptidos , Péptidos/farmacología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/farmacología , Receptores de Tromboxanos/metabolismo , Línea Celular Transformada , Células Cultivadas , Medios de Cultivo Condicionados , Ciclooxigenasa 1 , Ciclooxigenasa 2 , ADN Complementario/genética , Proteínas de Unión al ADN/metabolismo , GTP Fosfohidrolasas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/enzimología , Isoenzimas/metabolismo , Riñón/citología , Riñón/enzimología , Proteínas de la Membrana , Invasividad Neoplásica , Proteínas/genética , Proteínas/farmacología , Transducción de Señal , Transfección , Factor Trefoil-1 , Factor Trefoil-2 , Factor Trefoil-3 , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Fosfolipasas de Tipo C/metabolismo
16.
Cell Mol Biol (Noisy-le-grand) ; 47(1): 167-78, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11292252

RESUMEN

To establish new tools for studying human thymic stromal cells, we transfected adherent cells from a human postnatal thymus using a plasmid encoding SV40 large T antigen. Among the cell lines obtained, we characterized four epithelial cell lines (LT-TEC1 to LT-TEC4) and one thymic myoid cell line (MITC). Several morphological, functional and phenotypic differences were observed between these 2 cell types. Epithelial cells were heterogeneous and larger than myoid cells. Untreated LT-TEC lines expressed MHC class I, ICAM-1 and LFA-3 antigens and not MHC class II antigens, similarly to primary thymic epithelial cells (PTEC), while MITC line expressed only class I and LFA-3 antigens. After IFN-gamma treatment, MHC class II and ICAM-1 antigens were markedly upregulated in LT-TEC lines but not in MITC, indicating the absence or a dysfunction of regulatory factors in MITC line. Myoid cells expressed mRNA for all the subunits of the acetylcholine receptor (AChR) while epithelial cells expressed only the alpha, beta and epsilon subunits. Strikingly, LT-TEC produced much more C-C chemokines and IL-6 than MITC cells, while these latter produced higher levels of IL-8 and TNF-alpha. Altogether, these results reveal phenotypic and functional differences between these two stromal cell types, suggesting a potential involvement of myoid cells in the thymic function.


Asunto(s)
Timo/citología , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos CD58/biosíntesis , Línea Celular Transformada , Células Cultivadas , Citocinas/biosíntesis , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Humanos , Lactante , Molécula 1 de Adhesión Intercelular/biosíntesis , Interferón gamma/inmunología , Interferón gamma/farmacología , Ratones , Ratones Endogámicos BALB C , Fenotipo , Receptores Colinérgicos/biosíntesis , Células del Estroma/clasificación , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/fisiología
17.
FASEB J ; 15(2): 351-61, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11156951

RESUMEN

Trefoil factors (TFFs) are protease-resistant peptides that promote epithelial cell migration and mucosal restitution during inflammatory conditions and wound healing in the gastrointestinal tract. To date, the molecular mechanism of TFFs action and their possible role in tumor progression are unclear. In the present study, we observed that premalignant human colonic PC/AA/C1 and canine kidney MDCK epithelial cells are not competent to invade collagen gels in response to exogenously added TFFs (pS2, spasmolytic polypeptide, and intestinal trefoil factor). In contrast, activated src and RhoA exert permissive induction of invasion by the TFFs that produce similar parallel dose-response curves in src-transformed MDCKts.src and PCmsrc cells (EC50=20-40 nM). Cell scattering is also induced by TFFs in MDCKts.src cells. Stable expression of the pS2 cDNA promotes constitutive invasiveness in MDCKts.src-pS2 cells and human colonic HCT8/S11-pS2 cells established from a sporadic tumor. Furthermore, we found that TFF-mediated cellular invasion is dependent of several signaling pathways implicated in cell transformation and survival, including phosphoinositide PI3'-kinase, phospholipase C, protein kinase C, and the rapamycin target TOR. Constitutive and intense expression of pS2 was revealed by Western blot analyses and immunohistochemistry in human colorectal tumors and their adjacent control mucosa during the neoplastic progression, from the adenoma to the liver metastases. Our studies indicated that TFFs can be involved in cell scattering and tumor invasion via autocrine loops and may serve as potential targets in the control of colon cancer progression.


Asunto(s)
Neoplasias Colorrectales/patología , Genes src , Sustancias de Crecimiento/farmacología , Mucinas , Proteínas Musculares , Invasividad Neoplásica , Neuropéptidos , Péptidos/farmacología , Proteína de Unión al GTP rhoA/fisiología , Animales , Línea Celular Transformada , Movimiento Celular , Colágeno , Neoplasias del Colon , Perros , Inhibidores Enzimáticos/farmacología , Humanos , Mucosa Intestinal/patología , Mucosa Intestinal/fisiopatología , Riñón , Lesiones Precancerosas , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor Trefoil-2 , Factor Trefoil-3 , Urotelio , Proteína de Unión al GTP rhoA/genética
18.
Biochem Biophys Res Commun ; 277(3): 741-51, 2000 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-11062023

RESUMEN

Rac1 is a member of the Rho family of small GTPases involved in signal transduction pathways that control proliferation, adhesion, and migration of cells during embryonic development and invasiveness of tumor cells. Here we present the complete structure of the human RAC1 gene and characterize its expression. The gene comprises 7 exons over a length of 29 kb and is localized to chromosome 7p22. The GC-rich gene promoter shows characteristics of a housekeeping gene and Northern blot studies revealed ubiquitous expression of two rac1 transcripts, 1.2 and 2.5 kb in size. The two transcripts are expressed in tissue-specific ratios, reflecting competition between two alternative polyadenylation sites. The RAC1 but not RAC2 gene contains an additional exon 3b that is included by alternative splicing into the variant Rac1b, a constitutively active mutant which induces the formation of lamellipodia in fibroblasts. These data indicate that the RAC1 gene encodes two signaling GTPases. The gene structure reported here will enable studies on the regulation of RAC1 expression during tumorigenesis and development.


Asunto(s)
Regulación de la Expresión Génica , Regiones Promotoras Genéticas/genética , Proteína de Unión al GTP rac1/genética , Regiones no Traducidas 5'/genética , Actinas/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Mapeo Cromosómico , Cromosomas Humanos Par 7 , Citoesqueleto/fisiología , ADN/análisis , Exones , Fibroblastos/fisiología , Genoma Humano , Humanos , Intrones , Cariotipificación , Datos de Secuencia Molecular , Peso Molecular , Transfección , Proteína de Unión al GTP rac1/aislamiento & purificación
19.
FASEB J ; 14(14): 2329-38, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11053255

RESUMEN

Leptin plays a key role regulating food intake, body weight and fat mass. These critical parameters are associated with an increased risk for digestive and mammary gland cancer in the Western population. Here we determined whether leptin contributes to the invasive phenotype of colonic and kidney epithelial cells at various stages of the neoplastic progression. First, leptin potently (EC50 = 10-30 ng/ml) induces invasion of collagen gels by premalignant familial adenomatous colonic cells PC/AA/C1 and nontumorigenic MDCK kidney epithelial cells, their src-transformed counterparts, and the human adenocarcinoma colonic cells LoVo and HCT-8/S11. Leptin and its Ob-Rb receptors were consistently identified by RT-PCR and immunoblotting in these cell lines, as well as in human colonic epithelial crypts, polyps, colonic tumor resections, and adjacent mucosa. Leptin-induced invasion was effectively blocked by pharmacological inhibitors of several downstream signaling pathways involved in cell transformation, namely, JAK2 tyrosine kinase (AG490), phosphoinositide PI3'-kinase (wortmannin and LY294002), mTOR kinase (rapamycin), and protein kinases C (GF109203X, Gö6976). Accordingly, leptin induces transient elevation of the PI3'-kinase lipid products in JAK2 immunoprecipitates prepared from parental MDCK cells. The leptin effect on invasion was potentiated by the activated form of the small GTPase RhoA and was abrogated by dominant negative mutants of RhoA, Rac1, and the p110alpha of PI3'-K. Our data indicate that leptin may exert a local and beneficial effect on migration of normal colonic epithelial cells and reparation of the inflamed or wounded digestive mucosa. We also emphasize a new role for leptin, linking the nutritional and body fat status to digestive cancer susceptibility by stimulating the invasive capacity of colonic epithelial cells at early stages of neoplasia. This finding has potential clinical implications for colon cancer progression and management of obesity.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Neoplasias del Colon/patología , Enzimas/metabolismo , Riñón/efectos de los fármacos , Leptina/farmacología , Receptores de Superficie Celular , Transducción de Señal , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Expresión Génica , Humanos , Immunoblotting , Riñón/citología , Leptina/genética , Leptina/metabolismo , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Receptores de Leptina , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
20.
Anticancer Drugs ; 11(7): 579-82, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11036962

RESUMEN

This study was designed to assess the inhibition of tumor growth by oxaliplatin combined with UFT and folinic acid (FA). Growth inhibition was studied in nude mice transplanted with human colorectal HT29 tumor cell xenografts and treated for 28 days with oral UFT (20 mg/kg/day) and FA (4 mg/kg/day), i.p. oxaliplatin (10 mg/kg on day 1) or a combination of oxaliplatin, UFT and FA, or else not treated (controls). Tumor surface area and weight were recorded twice a week, and mice were sacrificed at day 28. Two separate experiments were performed for each group of 25 mice. At day 28, mean tumor weights (g) were 2.89+/-0.22 (controls), 2.03+/-0.14 (oxaliplatin), 2.02+/-0.21 (UFT/FA) and 1.23+/-0.17 (oxaliplatin+UFT/FA). For the three treatment groups, tumor weight decreases were 30.1% (p<0.05), 29.9% (p<0.05) and 57.5% (p<0.001), respectively. Combined treatment (UFT/FA+oxaliplatin) reduced tumor weight by 39% compared to oxaliplatin alone (p<0.05) or UFT/FA (p<0.05). These results demonstrate the synergistic effect of the combination of oxaliplatin, UFT and FA in this HT29 cell xenograft model, and warrant further investigations in patients with metastatic colorectal cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Leucovorina/uso terapéutico , Compuestos Organoplatinos/uso terapéutico , Tegafur/uso terapéutico , Uracilo/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , División Celular , Neoplasias Colorrectales/patología , Combinación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Células HT29 , Humanos , Leucovorina/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Compuestos Organoplatinos/efectos adversos , Oxaliplatino , Tegafur/efectos adversos , Trasplante Heterólogo , Uracilo/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA