Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Med ; 3(11): 1233-41, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9359698

RESUMEN

Colorectal cancer (CRC) is the second leading cause of cancer deaths in the United States. Five-fluorouracil (5FU) remains the single most effective treatment for advanced disease, despite a response rate of only 20%. Herein, we show that the antioxidants pyrrolidinedithiocarbamate and vitamin E induce apoptosis in CRC cells. This effect is mediated by induction of p21WAF1/CIP1, a powerful inhibitor of the cell cycle, through a mechanism involving C/EBPbeta (a member of the CCAAT/enhancer binding protein family of transcription factors), independent of p53. Antioxidants significantly enhance CRC tumor growth inhibition by cytotoxic chemotherapy in vitro (5FU and doxorubicin) and in vivo (5FU). Thus, chemotherapeutic agents administered in the presence of antioxidants may provide a novel therapy for colorectal cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Antioxidantes/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Ciclinas/biosíntesis , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Acetilcisteína/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT , Neoplasias Colorrectales/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Doxorrubicina/uso terapéutico , Interacciones Farmacológicas , Inhibidores Enzimáticos/metabolismo , Fluorouracilo/uso terapéutico , Humanos , Pirrolidinas/uso terapéutico , Tiocarbamatos/uso terapéutico , Células Tumorales Cultivadas , Vitamina E/uso terapéutico
2.
J Clin Invest ; 84(3): 1017-23, 1989 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2760208

RESUMEN

Transforming growth factor alpha (TGF alpha) shares with epidermal growth factor (EGF) structural homology (35%), a common cell-surface membrane receptor (TGF alpha/EGF receptor), and a nearly identical spectrum of biological activity, including inhibition of gastric acid secretion. Herein, we report expression of TGF alpha mRNA in normal gastric mucosa of the adult guinea pig, rat, and dog. TGF alpha mRNA was also detected in matched surgically resected gastric mucosa and adjacent gastric carcinoma from 10 patients, and in gastric mucosa adjacent to a benign ulcer from an additional patient. TGF alpha protein was quantitated by radioimmunoassay and was present in tumor and adjacent mucosa. TGF alpha/EGF receptor mRNA was also detected in gastric mucosa from all species studied. Localization of TGF alpha and TGF alpha/EGF receptor mRNA expression was examined in samples of unfractionated guinea pig gastric mucosa and from chief cell-enriched and parietal cell-enriched fractions. All samples exhibited TGF alpha and TGF alpha/EGF receptor expression. The TGF alpha signal was greatest in the parietal cell fraction (5.8-fold increase), but was also enhanced in the chief cell fraction (1.9-fold increase) relative to the unfractionated gastric mucosa. Like TGF alpha expression, TGF alpha/EGF receptor mRNA expression was most intense in the parietal cell-enriched fraction (7.8-fold increase), but was also increased in the chief cell-enriched fraction (2.7-fold increase) relative to the unfractionated guinea pig gastric mucosa. We conclude that TGF alpha and TGF alpha/EGF receptor genes are expressed in normal adult mammalian gastric mucosa. These findings, when interpreted in light of described actions of TGF alpha and EGF, provide evidence that local production of TGF alpha could play an important role in the regulation of acid secretion and mucosal renewal in the stomach.


Asunto(s)
Receptores ErbB/aislamiento & purificación , Ácido Gástrico/metabolismo , Mucosa Gástrica/metabolismo , Factores de Crecimiento Transformadores/aislamiento & purificación , Adulto , Anciano , Anciano de 80 o más Años , Animales , Northern Blotting , Carcinoma/análisis , Carcinoma/metabolismo , Sondas de ADN , Perros , Receptores ErbB/fisiología , Mucosa Gástrica/fisiología , Cobayas , Humanos , Persona de Mediana Edad , ARN Mensajero/aislamiento & purificación , Ratas , Neoplasias Gástricas/análisis , Neoplasias Gástricas/metabolismo , Factores de Crecimiento Transformadores/metabolismo , Factores de Crecimiento Transformadores/fisiología
3.
J Clin Invest ; 99(9): 2254-9, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9151799

RESUMEN

A considerable amount of evidence collected from several different experimental systems indicates that cyclooxygenase-2 (COX-2) may play a role in colorectal tumorigenesis. Large epidemiologic studies have shown a 40-50% reduction in mortality from colorectal cancer in persons taking aspirin or other nonsteroidal antiinflammatory drugs on a regular basis. One property shared by all of these drugs is their ability to inhibit COX, a key enzyme in the conversion of arachidonic acid to prostaglandins. Two isoforms of COX have been characterized, COX-1 and COX-2. COX-2 is expressed at high levels in intestinal tumors in humans and rodents. In this study, we selected two transformed human colon cancer cell lines for studies on the role of COX-2 in intestinal tumorigenesis. We evaluated HCA-7 cells which express high levels of COX-2 protein constitutively and HCT-116 cells which lack COX-2 protein. Treatment of nude mice implanted with HCA-7 cells with a selective COX-2 inhibitor (SC-58125), reduced tumor formation by 85-90%. SC-58125 also inhibited colony formation of cultured HCA-7 cells. Conversely, SC-58125 had no effect on HCT-116 implants in nude mice or colony formation in culture. Here we provide evidence that there may be a direct link between inhibition of intestinal cancer growth and selective inhibition of the COX-2 pathway.


Asunto(s)
Neoplasias Colorrectales/enzimología , Inhibidores de la Ciclooxigenasa/farmacología , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Pirazoles/farmacología , Animales , Northern Blotting , Western Blotting , División Celular , Neoplasias Colorrectales/tratamiento farmacológico , Ciclooxigenasa 1 , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/uso terapéutico , Humanos , Isoenzimas/genética , Isoenzimas/inmunología , Proteínas de la Membrana , Ratones , Ratones Desnudos , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/inmunología , Prostaglandinas/inmunología , Prostaglandinas/metabolismo , Pirazoles/uso terapéutico , ARN/análisis , ARN/metabolismo , Trasplante Heterólogo , Células Tumorales Cultivadas
4.
Cancer Res ; 56(4): 733-7, 1996 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-8631005

RESUMEN

Colorectal cancer is the second leading cause of death from cancer in the United States. Continuous use of aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) has been shown to reduce the risk of colorectal cancer in humans by 40-50%. Patients with familial adenomatous polyposis who take NSAIDs, such as sulindac, undergo a regression of intestinal adenomas. Rodents exposed to carcinogens that cause colon cancer have a 50-60% reduction in the size and number of colonic tumors when treated continuously with NSAIDs. One common target for these drugs is prostaglandin endoperoxide synthase, also referred to as cyclooxygenase (COX). We and others have shown recently that COX-2 levels are increased dramatically in 85-90% of human colorectal adenocarcinomas and in 40-50% of colonic adenomas. We prepared intestinal epithelial cells that express the COX-2 gene permanently and found that they have altered adhesion properties and resist undergoing apoptosis. We report here that these cells also have a 3-fold increase in the duration of G1, lower levels of cyclin D1 protein, and a marked decrease in retinoblastoma kinase activity associated with cyclin-dependent kinase 4. The delay in G1 transit may relate to the resistance of these cells to undergo programmed cell death, which could affect their tumorigenic potential.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Anticarcinógenos/uso terapéutico , Aspirina/uso terapéutico , Neoplasias Colorrectales/prevención & control , Fase G1 , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Proteínas Proto-Oncogénicas , Adenoma/prevención & control , Animales , Línea Celular , Neoplasias del Colon/prevención & control , Neoplasias Colorrectales/mortalidad , Ciclina D1 , Quinasa 4 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/biosíntesis , Ciclinas/biosíntesis , ADN/biosíntesis , ADN/efectos de los fármacos , Epitelio , Humanos , Intestinos , Cinética , Proteínas Oncogénicas/biosíntesis , Sistemas de Lectura Abierta , Ratas , Proteínas Recombinantes/biosíntesis , Proteína de Retinoblastoma/biosíntesis , Factores de Tiempo , Transfección , Estados Unidos/epidemiología
5.
Cancer Res ; 47(19): 5000-4, 1987 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-3621187

RESUMEN

We have recently demonstrated that gastrin stimulates growth of mouse colon cancer (MC-26) in vivo by regulation of gastrin receptors (GR). In the present study, we have tested the effect of proglumide (PGL), a GR antagonist, on the trophic and GR-regulatory effects of gastrin on MC-26 tumors. Four groups of 12 mice each were inoculated with 5 X 10(4) MC-26 cells and given injections of either normal saline (control), pentagastrin (PG), PGL, or both PG + PGL for 21 days. At the end of the treatment period, body, tumor, fundic, and colon weights were noted and GR measured. Two types of specific gastrin-binding sites were found on tumor cell membranes of control mice, one with high binding affinity (Kd = less than 1.0 nM) and low capacity (GR), and the other with a very high capacity and a low affinity (Kd = greater than 0.1 microM) (type 2 gastrin-binding sites). Only the type 1 GR were observed on the fundic mucosal and colon membranes. PG treatment resulted in a significant weight increase of the tumors with an up-regulation of only type 1 GR. On the other hand, PG had no significant effect on fundic mucosal and colonic GR levels, but caused a significant increase in fundic mucosal weights. PGL completely inhibited both the trophic and GR up-regulatory effects of PG on tumors, but incompletely reduced the PG-stimulated fundic mucosal weight gain, indicating differential sensitivity of tumor and normal tissues to PGL. PGL, in the absence of PG, was slightly trophic for normal fundic mucosa, but had no effect on MC-26 tumors and normal colon. The one striking effect of PGL, in the presence of PG, was the significant lowering of the binding affinity of type 1 GR for gastrin on both the tumor and normal gastrointestinal tissues. This effect may be another mechanism by which PGL interferes with the actions of PG on MC-26 tumors and fundic mucosa of mice.


Asunto(s)
Neoplasias del Colon/patología , Glutamina/análogos & derivados , Pentagastrina/farmacología , Proglumida/farmacología , Receptores de Colecistoquinina/efectos de los fármacos , Animales , Colon/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Gastrinas/metabolismo , Masculino , Ratones
6.
Cancer Res ; 58(2): 362-6, 1998 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9443418

RESUMEN

Previously, we have shown that forced expression of prostaglandin endoperoxide synthase-2 [also called cyclooxygenase (COX) 2] leads to inhibition of programmed cell death in intestinal epithelial cells. More recently, we have demonstrated that growth of human colonic cancer xenografts is inhibited by treatment with a highly selective COX-2 inhibitor in tumors that express COX-2 (HCA-7) but not in those that lack COX-2 expression (HCT-116). To explore the biochemical mechanisms involved in these effects, we have evaluated the role of COX-2-derived eicosanoid products on programmed cell death in human colon cancer cells. Here we report that PGE2 treatment of human colon cancer cells leads to increased clonogenicity of HCA-7, but not HCT-116 cells. Treatment with a highly selective COX-2 inhibitor (SC-58125) decreases colony formation in monolayer culture and this growth inhibition was reversed by treatment with PGE2. Additionally, PGE2 inhibits programmed cell death caused by SC-58125 and induces Bcl-2 expression, but did not affect Bcl-x or Bax expression in human colon cancer (HCA-7) cells. Therefore, decreased cell death caused by PGE2 would enhance the tumorigenic potential of intestinal epithelial cells. Thus, these results may help to explain a component of the mechanism by which COX inhibitors prevent colorectal cancer in humans.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias del Colon/metabolismo , Dinoprostona/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Animales , Células Clonales/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Immunoblotting , Isoenzimas/metabolismo , Proteínas de la Membrana , Ratones , Ratones Desnudos , Neoplasias Experimentales/patología , Prostaglandina-Endoperóxido Sintasas/metabolismo , Pirazoles/farmacología , Células Madre/efectos de los fármacos , Trasplante Heterólogo , Células Tumorales Cultivadas/efectos de los fármacos
7.
Cancer Res ; 58(11): 2323-7, 1998 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-9622066

RESUMEN

Increased expression of cyclooxygenase (COX) and overproduction of prostaglandins (PGs) have been implicated in the development and progression of colorectal cancer (CRC). Recent observations suggest that reactive oxygen intermediates play a role in tumor cell growth regulation and expression of the inducible COX, COX-2. We therefore evaluated the effects of various antioxidants on COX expression and cellular growth in the human CRC cell line HCA-7. The antioxidants pyrrolidinedithiocarbamate (PDTC), N-acetylcysteine, 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid (Trolox), and U74006 decreased PG production, intracellular redox status, and cellular growth in a concentration-dependent manner. The decrease in cellular growth was associated with the induction of apoptosis. Unlike the selective COX inhibitors 1-[(4-methylsulfonyl)phenyl]-3-trifluoromethyl-5-[(4-fluoro)phenyl]pyraz ole (SC 58125) and (2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide (NS 398) that inhibit COX-2 catalytic activity, these antioxidants decreased COX-2 expression at the transcriptional level. Combined treatment of HCA-7 cells with PDTC and SC 58125 resulted in an additive decrease in PG levels and anchorage-dependent and -independent growth. Furthermore, whereas antioxidants or SC 58125 reduced tumor growth in vivo, coadministration of PDTC and SC 58125 resulted in actual tumor regression. These results suggest that combined therapy with NSAIDs and antioxidants might be useful in the prevention and/or treatment of CRC.


Asunto(s)
Antioxidantes/farmacología , Neoplasias Colorrectales/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Isoenzimas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Prostaglandinas/biosíntesis , Pirazoles/farmacología , Animales , División Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Humanos , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos BALB C , Oxidación-Reducción , Transcripción Genética , Células Tumorales Cultivadas
8.
Cancer Res ; 51(19): 5253-60, 1991 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-1913648

RESUMEN

A serotonin-secreting human pancreatic carcinoid cell line (BON) is demonstrated to express transcripts for all three mammalian types of transforming growth factor beta (TGF beta 1, 2, and 3). Similarly, freshly excised carcinoid tumors from six patients were also found to express mRNA for all three of the type-beta TGFs. Medium conditioned by BON cells had detectable TGF beta activity, although most of the activity was latent as determined by radioreceptor assay with and without prior acid treatment. However, nonactivated BON-conditioned medium stimulated DNA synthesis, soft agar growth, and an increase in TGF beta 1 and fibronectin mRNA expression in AKR-2B fibroblasts. In addition, BON-conditioned medium had a potent endothelial cell growth-stimulatory activity. Since the TGF beta s inhibit growth of endothelial cells, the presence of other growth factors was suspected. TGF alpha, c-sis, and basic fibroblast growth factor transcripts were also found to be expressed by the BON carcinoid cells. These data indicate that multiple peptide growth factors may have a paracrine role in the desmoplastic reaction accompanying carcinoid tumors.


Asunto(s)
Tumor Carcinoide/metabolismo , Sustancias de Crecimiento/biosíntesis , Neoplasias Pancreáticas/metabolismo , Isomerasas de Aminoácido/biosíntesis , Northern Blotting , Proteínas Portadoras/biosíntesis , División Celular/fisiología , ADN/biosíntesis , Relación Dosis-Respuesta a Droga , Endotelio/parasitología , Fibroblastos/patología , Fibronectinas/biosíntesis , Humanos , Insulina/farmacología , Isomerasa de Peptidilprolil , Procolágeno/biosíntesis , ARN Mensajero/biosíntesis , Factores de Tiempo , Transcripción Genética , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis
9.
Cancer Res ; 48(11): 3265-9, 1988 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-3130188

RESUMEN

We studied the effect of inhibition of polyamine biosynthesis by alpha-difluoromethylornithine on the growth of a human gastric adenocarcinoma (CLEES) xenotransplanted in nude mice. CLEES is a well-differentiated gastric adenocarcinoma of the intestinal type. The doubling time has ranged from 7 to 10 days through 11 passages. Electron microscopic and immunohistochemical studies comparing the original tumor and xenotransplants showed similar structure and similar amounts of carcinoembryonic antigen. Polyamine biosynthesis is required for cell division. alpha-Difluoromethylornithine inhibits ornithine decarboxylase, the rate-limiting enzyme in polyamine biosynthesis. In this study, 48 athymic mice were used in two experiments. In the first experiment, two groups of 12 mice each were inoculated with CLEES tumor cells and received either tap water or a 3% alpha-difluoromethylornithine solution as drinking water. Tumor size was measured twice weekly. Tumor size was significantly decreased from controls by the fourth week of treatment and at all points of analysis thereafter for 7 wk. In the second experiment, alpha-difluoromethylornithine significantly reduced tumor concentrations of the polyamines putrescine and spermidine. In addition, the tumor content of DNA was significantly reduced in treated mice (0.64 +/- 0.16 mg) compared to controls (4.76 +/- 0.92 mg). Our data suggest that inhibition of polyamine biosynthesis may be a useful component of multidrug chemotherapy for human gastric adenocarcinoma. Establishment of tumor lines such as this gastric adenocarcinoma will facilitate further studies on the biological behavior of human gastric cancer and its response to chemotherapeutic manipulation in vivo.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Eflornitina/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Membrana Celular/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Poliaminas/análisis , Receptores de Colecistoquinina/análisis , Neoplasias Gástricas/patología , Trasplante Heterólogo , Células Tumorales Cultivadas
10.
Cancer Res ; 57(1): 169-75, 1997 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-8988060

RESUMEN

We postulated that increased expression of the cell cycle regulators cyclin D1 and cyclin-dependent kinase (Cdk) 4 may be involved in the development of intestinal adenomas associated with familial adenomatous polyposis (FAP). In the present study of multiple intestinal neoplasia (Min) mice and human FAP patients, the expression and distribution of cyclin D1, Cdk4, and cell proliferative activity (5-bromo-2'-deoxyuridine incorporation) in normal and adenomatous intestinal epithelium were investigated. Immunohistochemical analysis of Min mouse intestine revealed that cyclin D1 immunoreactivity in the intestinal epithelium was restricted to the adenomatous areas, with a significantly higher percentage of positively staining nuclei in high-grade dysplasia versus low-grade dysplasia (54.8 +/- 18.4% versus 34.6 +/- 16.9%, P = 0.016). Morphologically normal areas of intestinal epithelia were uniformly negative for cyclin D1 immunoreactivity. Cdk4 nuclear immunoreactivity was restricted to the crypt areas in morphologically normal small intestine and colon. Conversely, Cdk4 immunoreactivity was uniformly abundant in adenomatous areas regardless of the degree of dysplasia. Increased expression of cyclin D1 and Cdk4 in adenomas was accompanied by a significantly increased 5-bromo-2'-deoxyuridine incorporation rate in the same areas. Immunoblot analysis of lysates from surgical specimens revealed increased levels of cyclin D1 and Cdk4 in the majority of intestinal adenomas from human FAP patients in comparison to the adjacent grossly normal colonic mucosa. Our results indicate that overexpression of cyclin D1 and Cdk4 occurs in intestinal adenomas and is associated with increased cell proliferative activity in premalignant neoplastic cells. Increased cyclin D1 immunoreactivity is associated with more severe dysplasia. These data suggest that abnormal up-regulation of these important G1 cell cycle proteins is a relatively early event in intestinal carcinogenesis and that these changes may contribute to malignant progression within those lesions.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Neoplasias Intestinales/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Primarias Múltiples/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas , Poliposis Adenomatosa del Colon/patología , Animales , División Celular , Ciclina D1 , Quinasa 4 Dependiente de la Ciclina , Humanos , Neoplasias Intestinales/patología , Ratones , Ratones Endogámicos AKR , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Primarias Múltiples/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo
11.
Cancer Res ; 57(9): 1638-43, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9134999

RESUMEN

Tumor cells often become resistant to the growth-inhibitory effects of transforming growth factor beta (TGF-beta). Recent studies have identified TGF-beta type II receptor (RII) mutations in a subset of cancers, including colon cancer. To evaluate the expression of TGF-beta RII in premalignant intestinal adenomas and the relationship with cell cycle regulation, we investigated the expression of TGF-beta RII, cyclin D1, and cyclin-dependent kinase 4 (Cdk4) in Min/+ mouse intestinal adenomas. Immunohistochemistry indicated that TGF-beta RII cytoplasmic immunoreactivity was undetectable in the proliferative crypt zones of the normal small intestinal and normal colonic epithelium but was abundant toward the villus tips of the normal small intestine and the lumenal third of the colonic glands. As was observed in the proliferating crypt zones, TGF-beta RII immunoreactivity was dramatically decreased or undetectable in all adenomas examined in comparison to the abundant levels in adjacent normal differentiated intestinal epithelium. TGF-beta RII mRNA was also reduced in the adenomas in comparison to normal mucosa as determined by reverse transcription-PCR. In an inverse distribution to TGF-beta RII, Cdk4 nuclear immunoreactivity was restricted to the crypt regions of the small and large intestine, whereas cyclin D1 immunoreactivity was uniformly absent in normal intestinal epithelium. For both cyclin D1 and Cdk4, protein and mRNA levels were increased in intestinal adenomas but not in normal intestinal epithelium as determined by immunohistochemistry, in situ hybridization, and reverse transcription-PCR. In summary, the lack of TGF-beta RII expression was associated with increased cyclin D1 and Cdk4 expression in Min/+ mouse intestinal adenomas. We hypothesize that the former may enable tumor cells to escape from the normal growth-constraining influence of TGF-beta, whereas the latter promotes inappropriate cell proliferation and adenoma progression.


Asunto(s)
Adenoma/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Neoplasias Intestinales/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína de la Poliposis Adenomatosa del Colon , Animales , Ciclina D1 , Quinasa 4 Dependiente de la Ciclina , Proteínas del Citoesqueleto/genética , Hibridación in Situ , Mucosa Intestinal/metabolismo , Ratones , Ratones Mutantes , Proteínas Serina-Treonina Quinasas , ARN Mensajero/genética , ARN Neoplásico/genética , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética
12.
Cancer Res ; 61(14): 5389-95, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11454681

RESUMEN

Cyclin D1 is a known oncogene and a key regulator of cell cycle progression. Amplification of the cyclin D1 gene and its overexpression have been associated with aggressive forms of human hepatocellular carcinoma (HCC). In this study, two independent lines of transgenic mice have been generated that express cyclin D1 under the control of the rat liver fatty acid binding protein promoter. This transgene specifically directs expression in the liver and the intestines. RNA and protein analysis demonstrated increased expression of the cyclin D1 gene product in the liver and bowel when compared with wild-type siblings. Both transgenic lines developed progressive liver disease. Examination of H&E stained sections of the liver and bowel revealed hyperplastic changes in the liver by 3 months of age. By 6 months of age, transgenic mice had obvious hepatomegaly and histological evidence of dysplasia in the liver. These early changes were significantly more dramatic in male animals when compared with female animals. By 9 months of age adenomas of the liver appeared, progressing to HCC over the ensuing 6-month period. By 15-17 months of age, 87% of male and 69% of female animals had either adenomatous nodules or HCCs. By 17 months of age, 31% of male and female animals had disease that had progressed to HCC. These animals represent a unique and significant new model for the study of human HCC. This study demonstrates that overexpression of cyclin D1 is sufficient to initiate hepatocellular carcinogenesis.


Asunto(s)
Carcinoma Hepatocelular/genética , Ciclina D1/genética , Neoplasias Hepáticas/genética , Animales , Apoptosis/genética , Carcinoma Hepatocelular/patología , ADN Complementario/genética , ADN Complementario/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Hepatomegalia/genética , Hepatomegalia/patología , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ratas , Factores Sexuales , Factores de Tiempo , Transgenes/genética
13.
Oncogene ; 10(1): 177-84, 1995 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-7824270

RESUMEN

Transforming growth factor-beta 1 (TGF-beta 1) inhibits most epithelial cell types by blocking cell cycle progression during the G1 phase. D cyclins are normally expressed during G1 and are regulators of G1 progression. One of the crucial functions of D cyclins is their ability to bind to a cyclin-dependent kinase (Cdk4). In mink lung epithelial cells, TGF-beta 1 inhibits Cdk4 expression. We have measured cell cycle progression and D cyclins and Cdk4 expression in non-transformed rat intestinal epithelial cell lines (IEC-6 and RIE-1) after TGF-beta 1 treatment. In exponentially growing cultures, TGF-beta 1 blocked DNA synthesis and suppressed cyclin D1 mRNA and protein expression, whereas the levels of cyclins D2, D3 and Cdk4 remained relatively unchanged. TGF-beta 1 was also added to G0-synchronized IEC-6 cells after serum stimulation. TGF-beta 1 prevention of G1 progression was associated with an inhibition of cyclin D1 protein expression. Cyclin D3 levels were not affected by TGF-beta 1 during G1 traverse. Our results suggest that cyclin D/Cdk4 is a crucial target of TGF-beta 1 and that regulation of this kinase is mediated through cyclin D1 in intestinal epithelial cells.


Asunto(s)
Ciclinas/antagonistas & inhibidores , Intestino Delgado/metabolismo , Proteínas Oncogénicas/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología , Animales , División Celular , Ciclina D1 , Ciclinas/genética , Células Epiteliales , Epitelio/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Intestino Delgado/citología , Proteínas Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas
14.
Oncogene ; 16(26): 3445-54, 1998 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-9692552

RESUMEN

Transforming growth factor-beta 1 (TGF-beta1) arrests intestinal epithelial cells (RIE-1 and IEC-6) in the G1 phase of the cell cycle and inhibits cyclin D1 expression. This report describes experiments designed to elucidate the mechanism of cyclin D1 inhibition and to determine whether inhibition of cyclin D1 expression is the cause, rather than the result, of TGF-beta1-mediated cell cycle arrest. TGF-beta1 inhibition of IEC-6 cell proliferation was associated with a decrease in the abundance of cyclin D1/Cdk4 complexes and a corresponding decrease in Cdk4-dependent phosphorylation of the retinoblastoma protein. Metabolic labeling studies indicated that TGF-beta1 inhibited cyclin D1 synthesis without altering the rate of cyclin D1 protein degradation. Cyclin D1 antisense oligonucleotides blocked serum-stimulated induction of cyclin D1 and DNA synthesis, whereas cyclin D1 sense oligonucleotides had no effect. RIE-1 cells were engineered to overexpress human cyclin D1 under the control of a tetracycline-repressible promoter. These cells entered S phase in the presence of TGF-beta1 only when human cyclin D1 was derepressed by the withdrawal of tetracycline. These data indicate that TGF-beta1 inhibits the synthesis of cyclin D1 in gut epithelial cells and that this inhibition is the cause, rather than the result, of TGF-beta1-mediated arrest of intestinal epithelial cell proliferation.


Asunto(s)
Ciclina D1/biosíntesis , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Proteínas Proto-Oncogénicas , Factor de Crecimiento Transformador beta/farmacología , Animales , Línea Celular , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Células Epiteliales/citología , Fase G1/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Mucosa Intestinal/citología , Oligonucleótidos Antisentido/farmacología , Fosforilación , Unión Proteica , Ratas , Proteínas Recombinantes/biosíntesis , Proteína de Retinoblastoma/metabolismo
15.
Oncogene ; 19(7): 841-9, 2000 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-10702792

RESUMEN

Oncogenic mutations in ras lead to constitutive activation of downstream signaling pathways that modulate the activities of transcription factors. In turn, these factors control the expression of a subset of genes responsible for neoplastic cell transformation. Recent studies suggest that transcription factor NF-kappa B contributes to cell transformation by inhibiting the cell death signal activated by oncogenic Ras. In this study, inhibition of NF-kappa B activity by forced expression of a super-repressor form of I kappa B alpha, the major inhibitor of NF-kappa B, markedly decreased the growth rate, saturation density and tumorigenicity of oncogenic H-Ras transformed rat embryo fibroblasts. Such clonally isolated cells overexpressing I kappa B alpha super-repressor not only were viable but also exhibited no sign of spontaneous apoptosis. Inhibition of NF-kappa B in these cells was functionally demonstrated by both the loss of cytokine induced DNA binding activity and a profoundly increased sensitivity to cell death in response to TNF-alpha treatment. In contrast, inhibition of NF-kappa B activity in non-transformed fibroblasts had minimal effect on growth, but rendered the cells resistant to a subsequent transformation by H-ras oncogene. Similar results were also obtained with rat intestinal epithelial cells harboring an inducible ras oncogene. Taken together, these findings suggest that NF-kappa B activity is essential for abnormal cell proliferation and tumorigenicity activated by the ras oncogene and highlight an alternative functional role for NF-kappa B in oncogenic Ras-mediated cell transformation that is distinct from its anti-apoptotic activity. Oncogene (2000) 19, 841 - 849.


Asunto(s)
Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Genes ras/fisiología , FN-kappa B/fisiología , Animales , Apoptosis/genética , División Celular/genética , Línea Celular Transformada , Supervivencia Celular/genética , Transformación Celular Neoplásica/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Desnudos , Ratas
16.
Oncogene ; 18(4): 855-67, 1999 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-10023661

RESUMEN

The precise role of TGF-beta in colorectal carcinogenesis is not clear. The purpose of this study was to determine the phenotypic alterations caused by chronic exposure to TGF-beta in non-transformed intestinal epithelial (RIE-1) cells. Growth of RIE-1 cells was inhibited by >75% following TGF-beta1 treatment for 7 days, after which the cells resumed a normal growth despite the presence of TGF-beta1. These 'TGF-beta-resistant' cells (RIE-Tr) were continuously exposed to TGF-beta for >50 days. Unlike the parental RIE cells, RIE-Tr cells lost contact inhibition, formed foci in culture, grew in soft agarose. RIE-Tr cells demonstrated TGF-beta-dependent invasive potential in an in vitro assay and were resistant to Matrigel and Na-butyrate-induced apoptosis. The RIE-Tr cells were also tumorigenic in nude mice. The transformed phenotype of RIE-Tr cells was associated with a 95% decrease in the level of the type II TGF-beta receptor (TbetaRII) protein, a 40-fold increase in cyclooxygenase-2 (COX-2) protein, and 5.9-fold increase in the production of prostacyclin. Most RIE-Tr subclones that expressed low levels of TbetaRII and high levels of COX-2 were tumorigenic. Those subclones that express abundant TbetaRII and low levels of COX-2 were not tumorigenic in nude mice. A selective COX-2 inhibitor inhibited RIE-Tr cell growth in culture and tumor growth in nude mice. The reduced expression of TbetaRII, increased expression of COX-2, and the ability to form colonies in Matrigel were all reversible upon withdrawal of exogenous TGF-beta1 for the RIE-Tr cells.


Asunto(s)
Transformación Celular Neoplásica/inducido químicamente , Regulación hacia Abajo , Células Epiteliales/efectos de los fármacos , Intestinos/efectos de los fármacos , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Apoptosis , Recuento de Células , División Celular/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ciclooxigenasa 2 , Resistencia a Medicamentos , Inducción Enzimática , Células Epiteliales/metabolismo , Intestinos/citología , Fenotipo , Proteínas Serina-Treonina Quinasas , Ratas , Receptor Tipo II de Factor de Crecimiento Transformador beta
17.
J Clin Oncol ; 18(19): 3384-9, 2000 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11013279

RESUMEN

PURPOSE: We performed a phase I trial of protracted venous infusion (PVI) fluorouracil (5-FU) plus weekly gemcitabine with concurrent radiation therapy in patients with locally advanced pancreas cancer to determine the maximum-tolerated dose of gemcitabine that could be safely administered. We also sought to identify the toxicities associated with this treatment protocol. PATIENTS AND METHODS: Seven patients with locally advanced pancreas cancer were treated with planned doses of radiation (59.4 Gy) and PVI of 5-FU (200 mg/m(2)/d) with gemcitabine doses of 50 to 100 mg/m(2)/wk. RESULTS: Two of three patients at the 100-mg/m(2)/wk dose level experienced dose-limiting toxicity (DLT), as did three of four at the 50-mg/m(2)/wk dose level. One patient experienced a mucocutaneous reaction described as a Stevens-Johnson syndrome that was attributed to chemotherapy. Three patients developed gastric or duodenal ulcers with severe bleeding requiring transfusion. One patient developed severe thrombocytopenia lasting longer than 4 weeks. Three of the five episodes of DLT developed at radiation doses < or = 36 Gy. CONCLUSION: Based on this experience, we cannot recommend further investigation of regimens incorporating gemcitabine into regimens of radiation with PVI 5-FU. The mechanism of this synergistic toxicity remains to be determined.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Terapia Combinada , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Esquema de Medicación , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Radioterapia/efectos adversos , Análisis de Supervivencia , Gemcitabina
18.
Mol Endocrinol ; 6(11): 1789-96, 1992 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-1282669

RESUMEN

Transforming growth factor-beta (TGF beta) has been implicated in the regulation of hepatocyte function. We have examined TGF beta 1 regulation of albumin and alpha-fetoprotein (AFP) mRNA levels in a well differentiated mouse hepatoma cell line (BWTG3). TGF beta 1 reversibly decreased steady state mRNA levels of both albumin and AFP. By nuclear run-on assays, we found that TGF beta 1 caused no significant change in transcription rates for albumin or AFP. Pretreatment with actinomycin-D prevented the TGF beta 1-induced decrease in albumin and AFP mRNA levels. Also, if cells were treated with actinomycin-D after a 12-h exposure to TGF beta 1, actinomycin-D abrogated the further decrease in albumin and AFP mRNA levels that occurred after treatment with TGF beta 1 alone. Cycloheximide pretreatment blocked the TGF beta 1-induced decrease in albumin and AFP mRNA levels. TGF beta 1 altered neither the rate of BWTG3 cell growth nor the levels of mRNA for the growth-associated protooncogene c-myc. These data suggest that TGF beta 1 has regulatory effects on specific hepatocyte functions that are independent of growth regulatory effects. The decrease in albumin and AFP mRNAs caused by TGF beta 1 is posttranscriptional and dependent upon de novo RNA and protein synthesis.


Asunto(s)
Albúminas/biosíntesis , Hígado/efectos de los fármacos , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/farmacología , alfa-Fetoproteínas/biosíntesis , Albúminas/genética , Animales , Depresión Química , Regulación Neoplásica de la Expresión Génica , Hígado/metabolismo , Neoplasias Hepáticas Experimentales/patología , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , ARN Mensajero/metabolismo , ARN Neoplásico/metabolismo , Células Tumorales Cultivadas , alfa-Fetoproteínas/genética
19.
Oncogene ; 34(35): 4570-80, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25500541

RESUMEN

In normal colon, claudin-7 is one of the highly expressed claudin proteins and its knockdown in mice results in altered epithelial cell homeostasis and neonatal death. Notably, dysregulation of the epithelial homeostasis potentiates oncogenic transformation and growth. However, the role of claudin-7 in the regulation of colon tumorigenesis remains poorly understood. Using a large colorectal cancer (CRC) patient database and mouse models of colon cancer, we found claudin-7 expression to be significantly downregulated in cancer samples. Most notably, forced claudin-7 expression in poorly differentiated and highly metastatic SW620 colon cancer cells induced epithelial characteristics and inhibited their growth in soft agar and tumor growth in vivo. By contrast, knockdown of claudin-7 in HT-29 or DLD-1 cells induced epithelial-to-mesenchymal transition (EMT), colony formation, xenograft-tumor growth in athymic mice and invasion. Importantly, a claudin-7 signature gene profile generated by overlapping the DEGs (differentially expressed genes in a high-throughput transcriptome analysis using claudin-7-manipulated cells) with human claudin-7 signature genes identified high-risk CRC patients. Furthermore, Rab25, a colon cancer suppressor and regulator of the polarized cell trafficking constituted one of the highly upregulated DEGs in claudin-7 overexpressing cells. Notably, silencing of Rab25 expression counteracted the effects of claudin-7 expression and not only increased proliferation and cell invasion but also increased the expression of p-Src and mitogen-activated protein kinase-extracellular signal-regulated kinase 1/2 that were suppressed upon claudin-7 overexpression. Of interest, CRC cell lines, which exhibited decreased claudin-7 expression, also exhibited promoter DNA hypermethylation, a modification associated with transcriptional silencing. Taken together, our data demonstrate a previously undescribed role of claudin-7 as a colon cancer suppressor and suggest that loss of claudin-7 potentiates EMT to promote colon cancer, in a manner dependent on Rab25.


Asunto(s)
Adenocarcinoma/patología , Adenoma/patología , Carcinogénesis/metabolismo , Claudinas/fisiología , Neoplasias del Colon/patología , Transición Epitelial-Mesenquimal , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Adenoma/metabolismo , Adenoma/mortalidad , Animales , Neoplasias del Colon/metabolismo , Neoplasias del Colon/mortalidad , Células Epiteliales/metabolismo , Células HT29 , Humanos , Sistema de Señalización de MAP Quinasas , Ratones Desnudos , Trasplante de Neoplasias , Transcriptoma , Carga Tumoral , Proteínas de Unión al GTP rab/metabolismo
20.
Neoplasia ; 2(6): 523-30, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11228545

RESUMEN

Modulation of cyclooxygenase-2 (COX-2) mRNA stability plays an important role in the regulation of its expression by oncogenic Ras. Here, we evaluate COX-2 mRNA stability in response to treatment with two known endogenous promoters of gastrointestinal cancer, the bile acid (chenodeoxycholate; CD) and ceramide. Treatment with CD and ceramide resulted in a 10-fold increase in the level of COX-2 protein and a four-fold lengthening of the half-life of COX-2 mRNA. COX-2 mRNA stability was assessed by Northern blot analysis and by evaluating the AU-rich element located in the COX-2 3'-UTR. A known inhibitor of mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK), PD98059, reversed the effects of CD or ceramide to stabilize COX-2 mRNA. Overexpression of a dominant-negative ERK-1 or ERK-2 protein also led to destabilization of COX-2 mRNA. Treatment with a p38 MAPK inhibitor, PD169316, or transfection with a dominant-negative p38 MAPK construct reversed the effect of CD or ceramide to stabilize COX-2 mRNA. Expression of a dominant-negative c-Jun N-terminal kinase (JNK) had no effect on COX-2 mRNA stability in cells treated with CD or ceramide. We conclude that posttranscriptional mechanisms play an important role in the regulation of COX-2 expression during carcinogenesis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Mucosa Intestinal/efectos de los fármacos , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintasas/genética , Procesamiento Postranscripcional del ARN/genética , ARN Mensajero/metabolismo , Animales , Línea Celular , Ceramidas/farmacología , Ácido Quenodesoxicólico/farmacología , Ciclooxigenasa 2 , Genes Dominantes , Mucosa Intestinal/enzimología , Isoenzimas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA