Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Intervalo de ano de publicação
1.
Clin Cancer Res ; 15(22): 6780-9, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19903783

RESUMO

PURPOSE: Colon cancer is a major cause of cancer deaths. Dietary factors contribute substantially to the risk of this malignancy. Western-style diets promote development of azoxymethane-induced colon cancer. Although we showed that epidermal growth factor receptors (EGFR) controlled azoxymethane tumorigenesis in standard fat conditions, the role of EGFR in tumor promotion by high dietary fat has not been examined. EXPERIMENTAL DESIGN: A/J x C57BL6/J mice with wild-type Egfr (Egfr(wt)) or loss-of-function waved-2 Egfr (Egfr(wa2)) received azoxymethane followed by standard (5% fat) or western-style (20% fat) diet. As F(1) mice were resistant to azoxymethane, we treated mice with azoxymethane followed by one cycle of inflammation-inducing dextran sulfate sodium to induce tumorigenesis. Mice were sacrificed 12 weeks after dextran sulfate sodium. Tumors were graded for histology and assessed for EGFR ligands and proto-oncogenes by immunostaining, Western blotting, and real-time PCR. RESULTS: Egfr(wt) mice gained significantly more weight and had exaggerated insulin resistance compared with Egfr(wa2) mice on high-fat diet. Dietary fat promoted tumor incidence (71.2% versus 36.7%; P < 0.05) and cancer incidence (43.9% versus 16.7%; P < 0.05) only in Egfr(wt) mice. The lipid-rich diet also significantly increased tumor and cancer multiplicity only in Egfr(wt) mice. In tumors, dietary fat and Egfr(wt) upregulated transforming growth factor-alpha, amphiregulin, CTNNB1, MYC, and CCND1, whereas PTGS2 was only increased in Egfr(wt) mice and further upregulated by dietary fat. Notably, dietary fat increased transforming growth factor-alpha in normal colon. CONCLUSIONS: EGFR is required for dietary fat-induced weight gain and tumor promotion. EGFR-dependent increases in receptor ligands and PTGS2 likely drive diet-related tumor promotion.


Assuntos
Azoximetano/farmacologia , Neoplasias do Colo/metabolismo , Ciclo-Oxigenase 2/metabolismo , Sulfato de Dextrana/farmacologia , Receptores ErbB/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Animais , Glicemia/metabolismo , Regulação Neoplásica da Expressão Gênica , Insulina/sangue , Resistência à Insulina , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Mutação Puntual
2.
J Neurosci ; 28(45): 11537-49, 2008 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-18987190

RESUMO

Aspartoacylase (ASPA) is an oligodendrocyte-restricted enzyme that catalyzes the hydrolysis of neuronally derived N-acetylaspartate (NAA) to acetate and aspartic acid. ASPA deficiency leads to the fatal childhood autosomal recessive leukodystrophy Canavan disease (CD). Here we demonstrate that the previously described ENU-induced nur7 mouse mutant is caused by a nonsense mutation, Q193X, in the Aspa gene (Aspa(nur7)). Homozygous Aspa(nur7nur7) mice do not express detectable Aspa protein and display an early-onset spongy degeneration of CNS myelin with increased NAA levels similar to that observed in CD patients. In addition, CNS regions rich in neuronal cell bodies also display vacuolization. Interestingly, distinct myelin rich areas, such as the corpus callosum, optic nerve, and spinal cord white matter appear normal in Aspa(nur7/nur7) mice. Reduced cerebroside synthesis has been demonstrated in CD patients and animal models. To determine the potential relevance of this observation in disease pathogenesis, we generated Aspa(nur7/nur7) mice that were heterozygous for a null allele of the gene that encodes the enzyme UDP-galactose:ceramide galactosyltransferase (Cgt), which is responsible for catalyzing the synthesis of the abundant myelin galactolipids. Despite reduced amounts of cerebrosides, the Aspa(nur7/nur7);Cgt(+/-) mice were not more severely affected than the Aspa(nur7) mutants, suggesting that diminished cerebroside synthesis is not a major contributing factor in disease pathogenesis. Furthermore, we found that myelin degeneration leads to significant axonal loss in the cerebellum of older Aspa(nur7) mutants. This finding suggests that axonal pathology caused by CNS myelin defects may underlie the neurological disabilities that CD patients develop at late stages of the disease.


Assuntos
Amidoidrolases/genética , Doença de Canavan/genética , Sistema Nervoso Central/patologia , Códon sem Sentido , Modelos Animais de Doenças , Fatores Etários , Animais , Animais Recém-Nascidos , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Axônios/patologia , Comportamento Animal/fisiologia , Doença de Canavan/patologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/ultraestrutura , Mapeamento Cromossômico/métodos , Progressão da Doença , Glutamina/genética , Metabolismo dos Lipídeos/genética , Espectroscopia de Ressonância Magnética/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , Bainha de Mielina/ultraestrutura , N-Acilesfingosina Galactosiltransferase/genética , Oligodendroglia/patologia , Oligodendroglia/ultraestrutura
3.
J Steroid Biochem Mol Biol ; 111(1-2): 37-40, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18515093

RESUMO

Lithocholic acid (LCA), a secondary bile acid, is a vitamin D receptor (VDR) ligand. 1,25-Dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), the hormonal form of vitamin D, is involved in the anti-inflammatory action through VDR. Therefore, we hypothesize that LCA acts like 1,25(OH)(2)D(3) to drive anti-inflammatory signals. In present study, we used human colonic cancer cells to assess the role of LCA in regulation of the pro-inflammatory NF-kappaB pathway. We found that LCA treatment increased VDR levels, mimicking the effect of 1,25(OH)(2)D(3). LCA pretreatment inhibited the IL-1beta-induced IkappaBalpha degradation and decreased the NF-kappaB p65 phosphorylation. We also measured the production of IL-8, a well-known NF-kappaB target gene, as a read-out of the biological effect of LCA expression on NF-kappaB pathway. LCA significantly decreased IL-8 secretion induced by IL-1beta. These LCA-induced effects were very similar to those of 1,25(OH)(2)D(3.) Thus, LCA recapitulated the effects of 1,25(OH)(2)D(3) on IL-1beta stimulated cells. Mouse embryonic fibroblast (MEF) cells lacking VDR have intrinsically high NF-kappaB activity. LCA pretreatment was not able to prevent TNFalpha-induced IkappaBalpha degradation in MEF VDR (-/-), whereas LCA stabilized IkappaBalpha in MEF VDR (+/-) cells. Collectively, our data indicated that LCA activated the VDR to block inflammatory signals in colon cells.


Assuntos
Neoplasias do Colo/metabolismo , Regulação para Baixo/efeitos dos fármacos , Ácido Litocólico/farmacologia , NF-kappa B/metabolismo , Receptores de Calcitriol/efeitos dos fármacos , Células CACO-2 , Linhagem Celular Tumoral , Células Clonais , Neoplasias do Colo/genética , Células HT29 , Humanos , Ácido Litocólico/metabolismo , NF-kappa B/genética , Receptores de Calcitriol/metabolismo
4.
Dig Dis Sci ; 53(12): 3055-64, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18512153

RESUMO

BACKGROUND AND PURPOSE: Current therapies offer scant benefit to patients with advanced esophageal adenocarcinoma. We investigated the effects of Sorafenib, a multifunctional kinase inhibitor, on several growth regulatory pathways that control cell growth and survival in SEG-1 cells derived from Barrett's adenocarcinoma. METHODS: SEG-1 cells were exposed to acidified medium or taurocholic acid, with and without pre-incubation with Sorafenib. Cyclin D1 and E, c-Myc, and Bcl-2 expression levels as well as STAT3 activations were determined by Western blotting. Cyclin D1 mRNA was measured by real-time PCR. Apoptosis was assessed by TUNEL assay. RESULTS: Sorafenib significantly inhibited SEG-1 cell proliferation stimulated by acid or bile acid treatments and reduced cell survival. This drug significantly reduced the up-regulations of cyclin D1, cyclin E, c-Myc, and Bcl-2 as well as the activation of STAT3 in SEG-1 cells. CONCLUSIONS: These results support a rational basis for future clinical studies to assess the therapeutic benefit of Sorafenib in esophageal adenocarcinoma.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzenossulfonatos/farmacologia , Proliferação de Células/efeitos dos fármacos , Neoplasias Esofágicas/patologia , Piridinas/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Ciclina E/metabolismo , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Niacinamida/análogos & derivados , Compostos de Fenilureia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator de Transcrição STAT3/metabolismo , Sorafenibe , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Nutr Cancer ; 60(3): 389-400, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18444174

RESUMO

In the azoxymethane (AOM) model of experimental rodent colon cancer, cholic acid and its colonic metabolite deoxycholic acid (DCA) strongly promote tumorigenesis. In contrast, we showed that ursodeoxycholic acid (UDCA), a low abundance bile acid, inhibited AOM tumorigenesis. Dietary UDCA also blocked the development of tumors with activated Ras and suppressed cyclooxygenase-2 (Cox-2) upregulation in AOM tumors. In this study, we compared the effect of dietary supplementation with tumor-promoting cholic acid to chemopreventive UDCA on Cox-2 expression in AOM tumors. Cholic acid enhanced Cox-2 upregulation in AOM tumors, whereas UDCA inhibited this increase and concomitantly decreased CCAAT/enhancer binding protein beta (C/EBPbeta), a transcriptional regulator of Cox-2. In HCA-7 colon cancer cells, DCA activated Ras and increased C/EBPbeta and Cox-2 by a mechanism requiring the mitogen-activated protein kinase p38. UDCA inhibited DCA-induced p38 activation and decreased C/EBPbeta and Cox-2 upregulation. Using transient transfections, UDCA inhibited Cox-2 promoter and C/EBP reporter activation by DCA. Transfection with dominant-negative (17)N-Ras abolished DCA-induced p38 activation and C/EBPbeta and Cox-2 upregulation. Taken together, these studies have identified a transcriptional pathway regulating Cox-2 expression involving Ras, p38, and C/EBPbeta that is inhibited by UDCA. These signal transducers are novel targets of UDCA's chemopreventive actions.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Neoplasias do Colo/enzimologia , Ciclo-Oxigenase 2/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes ras , Ácido Ursodesoxicólico/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Azoximetano/toxicidade , Células Cultivadas , Quimioprevenção , Ácido Cólico/toxicidade , Neoplasias do Colo/induzido quimicamente , Ciclo-Oxigenase 2/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Suplementos Nutricionais , Genes ras/efeitos dos fármacos , Genes ras/fisiologia , Transdução de Sinais , Fatores de Transcrição/metabolismo , Transfecção
6.
Clin Cancer Res ; 14(8): 2253-62, 2008 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-18413814

RESUMO

PURPOSE: Colonic carcinogenesis deranges growth-regulating epidermal growth factor receptors (EGFR). We previously showed that EGFR signals were up-regulated in human aberrant crypt foci (ACF), putative colon cancer precursors. The azoxymethane model of colon cancer recapitulates many aspects of human colonic tumors. Recent studies indicate that flat dysplastic ACF with increased beta-catenin are tumor precursors in this model. We asked, therefore, if EGFR signals are required for flat dysplastic ACF development and cancer progression. EXPERIMENTAL DESIGN: Rats received azoxymethane or saline, and standard chow or chow supplemented with gefitinib, an EGFR inhibitor, for 44 weeks. EGFR signals were quantified in normal colon, flat ACF, and tumors by computerized analysis of immunostains and Western blots. K-ras mutations were assessed by PCR and mRNA for egfr ligands by quantitative real-time PCR. RESULTS: EGFR inhibition with gefitinib decreased the incidence of flat dysplastic ACF from 66% to 36% and tumors from 71% to 22% (P < 0.05). This inhibitor also reduced the overexpressions of cyclin D1 and Cox-2 in flat ACF. Furthermore, in flat ACF, EGFR blockade decreased the up-regulation of c-Jun, FosB, phosphorylated active signal transducers and activators of transcription 3, and CCAAT/enhancer binding protein-beta, potential regulators of cyclin D1 and Cox-2. In colonic tumors, EGFR blockade significantly decreased angiogenesis, proliferation, and progression while also increasing apoptosis (P < 0.05). Gefitinib also inhibited the activations of extracellular signal-regulated kinase, Src, and AKT pathways in tumors. CONCLUSIONS: We have shown for the first time that EGFR promotes the development of flat dysplastic ACF and the progression of malignant colonic tumors. Furthermore, we have mechanistically identified several transcription factors and their targets as EGFR effectors in colonic carcinogenesis.


Assuntos
Neoplasias do Colo/etiologia , Receptores ErbB/fisiologia , Lesões Pré-Cancerosas/etiologia , Animais , Azoximetano/toxicidade , Proteína beta Intensificadora de Ligação a CCAAT/fisiologia , Neoplasias do Colo/prevenção & controle , Ciclina D , Ciclinas/análise , Progressão da Doença , Receptores ErbB/antagonistas & inibidores , Gefitinibe , Masculino , Lesões Pré-Cancerosas/prevenção & controle , Quinazolinas/uso terapêutico , Ratos , Ratos Endogâmicos F344 , Fator de Transcrição STAT3/fisiologia , Fator de Transcrição AP-1/fisiologia
7.
J Surg Res ; 142(2): 239-45, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17574271

RESUMO

BACKGROUND: The azoxymethane (AOM) model recapitulates many features of human colon cancer, lacking an inflammatory component. Dextran sulfate sodium (DSS) induces colitis and promotes AOM-induced colon cancer in mice. Vitamin D analogues are anti-inflammatory and chemopreventive in models of colon cancer. Our aim was to evaluate the anti-inflammatory and chemopreventive efficacy of the vitamin D analogue Ro26-2198 in the AOM/DSS model and in vitro in HCA-7 colon cancer cells. MATERIALS AND METHODS: A/J mice received Ro26-2198 (0.01 microg/kg body wt/day x 28 days) or vehicle by mini-osmotic pump. Animals were treated with a single dose of AOM (5 mg/kg body wt) or vehicle 1 week after pump insertion. Mice received 3% DSS or water x 7 days beginning week 3. Animals were sacrificed after 8 weeks and colon segments were fixed in formalin or flash-frozen. Hematoxylin and eosin colonic sections were examined for dysplasia and colonic lysates were assessed for c-Myc, cyclooxygenase 2, and phospho-(active) extracellular signal regulated kinase (ERK) by Western blotting. For in vitro studies, HCA-7 cells were treated with Ro26-2198 followed by interleukin-1beta (IL-1beta). Proliferation was measured by WST-1 assay. RESULTS: Ro26-2198 delayed the onset of clinical colitis. Several dysplastic foci were present in the AOM/DSS group; none were found in the Ro26-2198 group. Compared with control, AOM/DSS significantly increased c-Myc (15-fold), cyclooxygenase 2 (COX-2) (2.5-fold), and pERK (10-fold), and Ro26-2198 abolished these increases. In vitro, Ro26-2198 inhibited IL-1beta-induced ERK activation and COX-2 induction and decreased HCA-7 cell proliferation. CONCLUSIONS: Ro26-2198 inhibited proliferative (ERK, c-Myc) and pro-inflammatory (COX-2) signals and progression to dysplasia, suggesting chemopreventive efficacy in this model of colitis-associated carcinogenesis.


Assuntos
Adenocarcinoma/prevenção & controle , Colecalciferol/análogos & derivados , Colite/prevenção & controle , Neoplasias do Colo/prevenção & controle , Adenocarcinoma/complicações , Adenocarcinoma/patologia , Animais , Anti-Inflamatórios/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Colecalciferol/farmacologia , Colite/complicações , Colite/patologia , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/complicações , Neoplasias do Colo/patologia , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Interleucina-1beta/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos A , Proteínas Proto-Oncogênicas c-myc/metabolismo , Regulação para Cima/efeitos dos fármacos
8.
Cancer Res ; 67(2): 827-35, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234795

RESUMO

Colonic carcinogenesis involves the progressive dysregulation of homeostatic mechanisms that control growth. The epidermal growth factor (EGF) receptor (EGFR) regulates colonocyte growth and differentiation and is overexpressed in many human colon cancers. A requirement for EGFR in colonic premalignancy, however, has not been shown. In the current study, we used a specific EGFR antagonist, gefitinib, to investigate this role of the receptor in azoxymethane colonic premalignancy. The azoxymethane model shares many clinical, histologic, and molecular features of human colon cancer. Mice received azoxymethane i.p. (5 mg/kg/wk) or saline for 6 weeks. Animals were also gavaged with gefitinib (10 mg/kg body weight) or vehicle (DMSO) thrice weekly for 18 weeks, a dose schedule that inhibited normal receptor activation by exogenous EGF. Compared with control colonocytes [bromodeoxyuridine (BrdUrd), 2.2+/-1.2%], azoxymethane significantly increased proliferation (BrdUrd, 12.6+/-2.8%), whereas gefitinib inhibited this hyperproliferation (BrdUrd, 6.2+/-4.0%; <0.005). Azoxymethane significantly induced pro-transforming growth factor-alpha (6.4+/-1.3-fold) and increased phospho-(active) EGFR (5.9+/-1.1-fold), phospho-(active) ErbB2 (2.3+/-0.2-fold), and phospho-(active) extracellular signal-regulated kinase (3.3+/-0.4-fold) in premalignant colonocytes. Gefitinib inhibited activations of these kinases by >75% (P<0.05). Gefitinib also significantly reduced the number of large aberrant crypt foci and decreased the incidence of colonic microadenomas from 75% to 33% (P<0.05). Gefitinib concomitantly decreased cell cycle-regulating cyclin D1 and prostanoid biosynthetic enzyme cyclooxygenase-2 in microadenomas, suggesting that these regulators are key targets of EGFR in colonic carcinogenesis. These results show for the first time that EGFR signaling is required for early stages of colonic carcinogenesis. Our findings suggest, moreover, that inhibitors of EGFR might be useful in chemopreventive strategies in individuals at increased risk for colonic malignancies.


Assuntos
Adenoma/enzimologia , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/enzimologia , Receptores ErbB/metabolismo , Adenoma/induzido quimicamente , Adenoma/genética , Adenoma/prevenção & controle , Animais , Azoximetano/antagonistas & inibidores , Carcinógenos , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/genética , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Neoplasias do Colo/prevenção & controle , Ciclina D1/antagonistas & inibidores , Ciclina D1/biossíntese , Ciclo-Oxigenase 2/biossíntese , Inibidores de Ciclo-Oxigenase 2/farmacologia , Modelos Animais de Doenças , Receptores ErbB/antagonistas & inibidores , Gefitinibe , Genes ras/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos A , Mutação/efeitos dos fármacos , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , beta Catenina/biossíntese , beta Catenina/genética
9.
Mol Cancer Res ; 4(9): 683-94, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16940160

RESUMO

Colonic carcinogenesis is accompanied by abnormalities in multiple signal transduction components, including alterations in protein kinase C (PKC). The expression level of PKC-zeta, an atypical PKC isoform, increases from the crypt base to the luminal surface and parallels crypt cell differentiation in normal colon. In prior studies in the azoxymethane model of colon cancer, we showed that PKC-zeta was down-regulated in rat colonic tumors. In this study, we showed that PKC-zeta is expressed predominantly in colonic epithelial and not stromal cells, and loss of PKC-zeta occurs as early as the adenoma stage in human colonic carcinogenesis. To assess the regulation of growth and differentiation by PKC-zeta, we altered this isoform in human Caco-2 colon cancer cells using stable constitutive or inducible expression vectors, specific peptide inhibitors or small interfering RNA. In ecdysone-regulated transfectants grown on collagen I, ponasterone A significantly induced PKC-zeta expression to 135% of empty vector cells, but did not alter nontargeted PKC isoforms. This up-regulation was accompanied by a 2-fold increase in basal and 4-fold increase in insulin-stimulated PKC-zeta biochemical activity. Furthermore, PKC-zeta up-regulation caused >50% inhibition of cell proliferation on collagen I (P < 0.05). Increased PKC-zeta also significantly enhanced Caco-2 cell differentiation, nearly doubling alkaline phosphatase activity, while inducing a 3-fold increase in the rate of apoptosis (P < 0.05). In contrast, knockdown of this isoform by small interfering RNA or kinase inhibition by myristoylated pseudosubstrate significantly and dose-dependently increased Caco-2 cell growth on collagen I. In transformation assays, constitutively up-regulated wild-type PKC-zeta significantly inhibited Caco-2 cell growth in soft agar, whereas a kinase-dead mutant caused a 3-fold increase in soft agar growth (P < 0.05). Taken together, these studies indicate that PKC-zeta inhibits colon cancer cell growth and enhances differentiation and apoptosis, while inhibiting the transformed phenotype of these cells. The observed down-regulation of this growth-suppressing PKC isoform in colonic carcinogenesis would be predicted to contribute to tumorigenesis.


Assuntos
Apoptose/fisiologia , Colágeno Tipo I/antagonistas & inibidores , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Proteína Quinase C/metabolismo , Células CACO-2 , Adesão Celular/fisiologia , Diferenciação Celular/fisiologia , Processos de Crescimento Celular/fisiologia , Colágeno Tipo I/metabolismo , Neoplasias do Colo/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/biossíntese , Proteína Quinase C/genética , RNA Interferente Pequeno/genética , Transfecção
10.
Cancer Res ; 66(11): 5656-64, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16740703

RESUMO

Aberrant crypt foci (ACF) are collections of abnormal colonic crypts with heterogeneous molecular and pathologic characteristics. Large and dysplastic ACF are putative precursors of colon cancer with neoplastic risk related to increased proliferation. In this study, we examined the role of epidermal growth factor receptor (EGFR) signaling in regulating ACF proliferation. Using magnification chromoendoscopy, we collected large ACF with endoscopic features of dysplasia and separately biopsied adjacent mucosa. Transcript levels were measured by real-time PCR, proteins were assessed by Western blotting, and levels were expressed as fold changes of adjacent mucosa. K-ras and B-Raf mutations were assessed by PCR and Ras activation by the ratio Ras-GTP / (Ras-GTP + Ras-GDP). At the RNA level, 38% of ACF were hyperproliferative, with proliferating cell nuclear antigen (PCNA) mRNA >/=2-fold of adjacent mucosa. Hyperproliferative ACF had significantly increased mRNA levels of EGFR (6.0 +/- 1.7-fold), transforming growth factor-alpha (14.4 +/- 5.0-fold), heparin-binding EGF-like growth factor (4.5 +/- 1.4-fold), cyclin D1 (4.6 +/- 0.7-fold), and cyclooxygenase-2 (COX-2; 9.3 +/- 4.2-fold; P < 0.05). At the protein level, 46% of ACF were hyperproliferative (PCNA, 3.2 +/- 1.2-fold). In hyperproliferative ACF, 44% possessed significant increases in four EGFR signaling components: EGFR (9.5 +/- 1.3-fold), phosphoactive ErbB2 (2.6 +/- 0.4-fold), phosphoactive extracellular signal-regulated kinase (3.7 +/- 1.1-fold), and cyclin D1 (3.4 +/- 0.8-fold; P < 0.05). Ras was activated in 46% of ACF (3.2 +/- 0.4-fold; P < 0.05), but K-ras mutations were present in only 7% of ACF. In contrast to COX-2 mRNA, the protein was not increased in hyperproliferative ACF. In summary, we have shown that ACF with up-regulated PCNA possess increased EGFR signaling components that likely contribute to the enhanced proliferative state of dysplastic-appearing ACF.


Assuntos
Neoplasias do Colo/metabolismo , Receptores ErbB/metabolismo , Lesões Pré-Cancerosas/metabolismo , Processos de Crescimento Celular/fisiologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Ciclina D1/biossíntese , Ciclina D1/genética , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Feminino , Genes ras , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Antígeno Nuclear de Célula em Proliferação/biossíntese , Antígeno Nuclear de Célula em Proliferação/genética , Proteínas Proto-Oncogênicas B-raf/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais , Regulação para Cima
11.
Cancer Res ; 63(13): 3517-23, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12839936

RESUMO

K-ras mutations occur frequently in colon cancer and contribute to autonomous growth. In the azoxymethane (AOM) model of colon cancer, in addition to K-ras mutations, we have shown that wild-type (WT) Ras can be activated by upstream pathways, including, e.g., signaling by ErbB receptors. Tumors with mutant or activated WT Ras had increased cyclooxygenase-2 (Cox-2) expression. We have also shown that ursodeoxycholic acid (UDCA) prevented AOM-induced colon cancer and suppressed Cox-2 induction. In this study, we examined the role of Ras in Cox-2 inhibition by UDCA. Rats were fed AIN-76A chow alone, or supplemented with 0.4% UDCA, and received 20 mg/kg AOM i.p. weekly x 2 weeks. At 40 weeks, rats were sacrificed, and tumors were harvested. K-ras mutations were assessed by primer-mediated RFLP, allele-specific oligonucleotide hybridization, and direct DNA sequencing. Ras was immunoprecipitated and defined as activated if [Ras - GTP/(Ras - GTP + Ras - GDP)] was >3 SD above normal colonocytes. Cox-2 mRNA was determined by reverse transcription-PCR, and protein expression was assessed by Western blotting and immunostaining. In the AOM alone group, Ras was activated by mutations in 8 of 30 (27%) tumors, and WT Ras was activated in 7 of 30 (23%) tumors. UDCA significantly suppressed the incidence of tumors with mutant Ras (1 of 31, 3.2%; P < 0.05) and totally abolished the development of tumors with activated WT Ras (0 of 10; P < 0.05). In the AOM alone group, Cox-2 was up-regulated >50-fold in tumors with normal Ras activity and further enhanced in tumors with mutant or signaling-activated Ras. UDCA significantly inhibited Cox-2 protein and mRNA levels in tumors with normal Ras activity. In summary, we have shown for the first time that UDCA suppressed the development of tumors with Ras mutations and blocked activation of WT Ras. Furthermore, UDCA inhibited Cox-2 induction by Ras-dependent and -independent mechanisms.


Assuntos
Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes ras , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintases/genética , Transcrição Gênica/efeitos dos fármacos , Ácido Ursodesoxicólico/farmacologia , Animais , Azoximetano , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/enzimologia , Ciclo-Oxigenase 2 , Genes ras/efeitos dos fármacos , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA