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










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 10(7): e0131716, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26147105

RESUMO

Histone methyltransferases are epigenetic regulators that modify key lysine and arginine residues on histones and are believed to play an important role in cancer development and maintenance. These epigenetic modifications are potentially reversible and as a result this class of enzymes has drawn great interest as potential therapeutic targets of small molecule inhibitors. Previous studies have suggested that the histone lysine methyltransferase G9a (EHMT2) is required to perpetuate malignant phenotypes through multiple mechanisms in a variety of cancer types. To further elucidate the enzymatic role of G9a in cancer, we describe herein the biological activities of a novel peptide-competitive histone methyltransferase inhibitor, A-366, that selectively inhibits G9a and the closely related GLP (EHMT1), but not other histone methyltransferases. A-366 has significantly less cytotoxic effects on the growth of tumor cell lines compared to other known G9a/GLP small molecule inhibitors despite equivalent cellular activity on methylation of H3K9me2. Additionally, the selectivity profile of A-366 has aided in the discovery of a potentially important role for G9a/GLP in maintenance of leukemia. Treatment of various leukemia cell lines in vitro resulted in marked differentiation and morphological changes of these tumor cell lines. Furthermore, treatment of a flank xenograft leukemia model with A-366 resulted in growth inhibition in vivo consistent with the profile of H3K9me2 reduction observed. In summary, A-366 is a novel and highly selective inhibitor of G9a/GLP that has enabled the discovery of a role for G9a/GLP enzymatic activity in the growth and differentiation status of leukemia cells.


Assuntos
Inibidores Enzimáticos/farmacologia , Epigênese Genética , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Indóis/farmacologia , Leucemia/enzimologia , Compostos de Espiro/farmacologia , Animais , Feminino , Xenoenxertos , Humanos , Leucemia/genética , Leucemia/patologia , Células MCF-7 , Camundongos , Camundongos SCID
2.
Clin Cancer Res ; 17(3): 459-71, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21149615

RESUMO

PURPOSE: To understand the functional and preclinical efficacy of targeting the urokinase plasminogen activator receptor (u-PAR) in ovarian cancer. EXPERIMENTAL DESIGN: Expression of u-PAR was studied in 162 epithelial ovarian cancers, including 77 pairs of corresponding primary and metastatic tumors. The effect of an antibody against u-PAR (ATN-658) on proliferation, adhesion, invasion, apoptosis, and migration was assessed in 3 (SKOV3ip1, HeyA8, and CaOV3) ovarian cancer cell lines. The impact of the u-PAR antibody on tumor weight, number, and survival was examined in corresponding ovarian cancer xenograft models and the mechanism by which ATN-658 blocks metastasis was explored. RESULTS: Only 8% of all ovarian tumors were negative for u-PAR expression. Treatment of SKOV3ip1, HeyA8, and CaOV3 ovarian cancer cell lines with the u-PAR antibody inhibited cell invasion, migration, and adhesion. In vivo, anti-u-PAR treatment reduced the number of tumors and tumor weight in CaOV3 and SKOV3ip1 xenografts and reduced tumor weight and increased survival in HeyA8 xenografts. Immunostaining of CaOV3 xenograft tumors and ovarian cancer cell lines showed an increase in active-caspase 3 and TUNEL staining. Treatment with u-PAR antibody inhibited α(5)-integrin and u-PAR colocalization on primary human omental extracellular matrix. Anti-u-PAR treatment also decreased the expression of urokinase, u-PAR, ß(3)-integrin, and fibroblast growth factor receptor-1 both in vitro and in vivo. CONCLUSIONS: This study shows that an antibody against u-PAR reduces metastasis, induces apoptosis, and reduces the interaction between u-PAR and α(5)-integrin. This provides a rationale for targeting the u-PAR pathway in patients with ovarian cancer and for further testing of ATN-658 in this indication.


Assuntos
Metástase Neoplásica/prevenção & controle , Neoplasias Ovarianas/patologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Animais , Anticorpos Monoclonais/farmacologia , Apoptose , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Camundongos , Invasividade Neoplásica , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Am J Pathol ; 175(5): 2184-96, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19808644

RESUMO

The role of the vitronectin receptor (alpha(v)beta(3)-integrin) as a tumor promoter seems well established, and, consequently, therapies that block this integrin are currently in clinical testing. We undertook the current study to determine whether alpha(v)beta(3)-integrin is an appropriate target in ovarian cancer treatment. Expression of beta(3)-integrin in SKOV3ip1 ovarian cancer cells led to the overexpression of alpha(v)beta(3)-integrin on the cell surface and increased adhesion. However, alpha(v)beta(3)-integrin-overexpressing cells showed impaired invasion, protease expression, and colony formation. These results were recapitulated in xenograft studies: alpha(v)beta(3)-integrin-expressing cells showed increased adhesion to mouse peritoneum, but the overall number of metastatic nodules (105 versus 68 tumors) and tumor weight were significantly lower than those in the parental SKOV3ip1 cells. The alpha(v)beta(3)-integrin-overexpressing cells had a decreased proliferation rate mediated by inhibition of cyclin B1 and induction of phospho-Cdc2 and p53 expression, consistent with a G(2)M cell cycle arrest. Confirming the above results, inhibition of beta(3)-integrin in cultured or primary OvCa cells decreased adhesion but increased invasion and proliferation. Patients with tumors expressing high beta(3)-integrin had significantly better disease-free and overall survival (52 months versus 27 months, P < 0.05). This study shows that alpha(v)beta(3)-integrin expression on tumor cells actually slows tumor progression and acts as a tumor suppressor. Therefore, the vitronectin receptor might not be an appropriate therapeutic target in ovarian cancer.


Assuntos
Integrina alfaVbeta3/metabolismo , Metástase Neoplásica/patologia , Neoplasias Ovarianas , Células Tumorais Cultivadas , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Biomarcadores/metabolismo , Adesão Celular/fisiologia , Ciclo Celular/fisiologia , Progressão da Doença , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Integrina alfaVbeta3/genética , Estimativa de Kaplan-Meier , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Transplante de Neoplasias , Omento/metabolismo , Neoplasias Ovarianas/diagnóstico , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Prognóstico
4.
Cancer Prev Res (Phila) ; 2(9): 792-9, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19737983

RESUMO

Although epidemiologic evidence for the ability of combined oral contraception (OC) to reduce the risk of ovarian cancer (OvCa) is convincing, the biological mechanisms underlying this effect are largely unknown. We conducted the present study to determine if OC also influences ovarian carcinogenesis in a genetic mouse model and, if so, to investigate the mechanism underlying the protective effect. LSL-K-ras(G12D/+)Pten(loxP/loxP) mice were treated with ethinyl estradiol plus norethindrone, contraceptive hormones commonly used in combined OC, or norethindrone alone, or a gonadotropin-releasing hormone agonist. The combined OC had a 29% reduction in mean total tumor weight compared with placebo (epithelial tumor weight, -80%). Norethindrone alone reduced mean total tumor weight by 42% (epithelial tumor weight, -46%), and the gonadotropin-releasing hormone agonist increased mean total tumor weight by 71% (epithelial tumor weight, +150%). Large variations in tumor size affected the P values for these changes, which were not statistically significant. Nonetheless, the OC reductions are consistent with the epidemiologic data indicating a protective effect of OC. Matrix metalloproteinase-2 activity was decreased in association with OC, indicating that OC may affect ovarian carcinogenesis by decreasing proteolytic activity, an important early event in the pathogenesis of OvCa. In contrast, OC increased invasion in a K-ras/Pten OvCa cell line established from the mouse tumors, suggesting that OC hormones, particularly estrogen, may have a detrimental effect after the disease process is under way. Our study results support further investigation of OC effects and mechanisms for OvCa prevention.


Assuntos
Anticoncepcionais Orais Sintéticos/administração & dosagem , Modelos Animais de Doenças , Etinilestradiol/administração & dosagem , Hormônio Liberador de Gonadotropina/agonistas , Noretindrona/administração & dosagem , Neoplasias Ovarianas/prevenção & controle , Animais , Apoptose , Western Blotting , Estrogênios/administração & dosagem , Feminino , Gonadotropinas/metabolismo , Técnicas Imunoenzimáticas , Integrases/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Knockout , Invasividade Neoplásica , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , PTEN Fosfo-Hidrolase/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Células Tumorais Cultivadas
5.
Cancer Res ; 68(7): 2329-39, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18381440

RESUMO

E-cadherin loss is frequently associated with ovarian cancer metastasis. Given that adhesion to the abdominal peritoneum is the first step in ovarian cancer dissemination, we reasoned that down-regulation of E-cadherin would affect expression of cell matrix adhesion receptors. We show here that inhibition of E-cadherin in ovarian cancer cells causes up-regulation of alpha(5)-integrin protein expression and transcription. When E-cadherin was blocked, RMUG-S ovarian cancer cells were able to attach and invade more efficiently. This greater efficiency could, in turn, be inhibited both in vitro and in vivo with an alpha(5)beta(1)-integrin-blocking antibody. When E-cadherin is silenced, alpha(5)-integrin is up-regulated through activation of an epidermal growth factor receptor/FAK/Erk1-mitogen-activated protein kinase-dependent signaling pathway and not through the canonical E-cadherin/beta-catenin signaling pathway. In SKOV-3ip1 ovarian cancer xenografts, which express high levels of alpha(5)-integrin, i.p. treatment with an alpha(5)beta(1)-integrin antibody significantly reduced tumor burden, ascites, and number of metastasis and increased survival by an average of 12 days when compared with IgG treatment (P < 0.0005). alpha(5)-Integrin expression was detected by immunohistochemistry in 107 advanced stage ovarian cancers using a tissue microarray annotated with disease-specific patient follow-up. Ten of 107 tissues (9%) had alpha(5)-integrin overexpression, and 39% had some level of alpha(5)-integrin expression. The median survival for patients with high alpha(5)-integrin levels was 26 months versus 35 months for those with low integrin expression (P < 0.05). Taken together, we have identified alpha(5)-integrin up-regulation as a molecular mechanism by which E-cadherin loss promotes tumor progression, providing an explanation for how E-cadherin loss increases metastasis. Targeting this integrin could be a promising therapy for a subset of ovarian cancer patients.


Assuntos
Caderinas/antagonistas & inibidores , Integrina alfa5/metabolismo , Neoplasias Ovarianas/patologia , Neoplasias Peritoneais/secundário , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/metabolismo , Western Blotting , Caderinas/biossíntese , Caderinas/metabolismo , Adesão Celular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Integrina alfa5/biossíntese , Integrina alfa5/genética , Integrina alfa5beta1/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Nus , Invasividade Neoplásica , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Neoplasias Peritoneais/enzimologia , Neoplasias Peritoneais/metabolismo , Prognóstico , 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.
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
8.
J Biol Chem ; 283(5): 2822-34, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18048360

RESUMO

The procoagulatory serine protease, thrombin, is known to induce invasion and metastasis in various cancers, but the mechanisms by which it promotes tumorigenesis are poorly understood. Because the 92-kDa gelatinase (MMP-9) is a known mediator of tumor cell invasion, we sought to determine whether and how thrombin regulates MMP-9. The thrombin receptor, PAR-1, and MMP-9 are expressed in osteosarcomas, as determined by immunohistochemistry. Stimulation of U2-OS osteosarcoma cells with thrombin and a thrombin receptor-activating peptide induced pro-MMP-9 secretion as well as cell surface-associated pro-MMP-9 expression and proteolytic activity. This was paralleled by an increase in MMP-9 mRNA and MMP-9 promoter activity. Thrombin-induced invasion of U2-OS cells through Matrigel was mediated by the phosphatidylinositol 3-kinase signaling pathway and could be inhibited with an MMP-9 antibody. The stimulation of MMP-9 by thrombin was paralleled by an increase in beta1-integrin mRNA and beta1-integrin expression on the cell surface, which was also mediated by phosphatidylinositol 3-kinase and was required for invasion. Thrombin activation induced and co-localized both beta1-integrin and pro-MMP-9 on the cell membrane, as evidenced by co-immunoprecipitation, confocal microscopy, and a protein binding assay. The thrombin-mediated association of these two proteins, as well as thrombin-mediated invasion of U2-OS cells, could be blocked with a cyclic peptide and with an antibody preventing binding of the MMP-9 hemopexin domain to beta1-integrin. These results suggest that thrombin induces expression and association of beta1-integrin with MMP-9 and that the cell surface localization of the protease by the integrin promotes tumor cell invasion.


Assuntos
Integrina beta1/fisiologia , Metaloproteinase 9 da Matriz/fisiologia , Invasividade Neoplásica/fisiopatologia , Trombina/farmacologia , Trombina/fisiologia , Sequência de Aminoácidos , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Neoplasias Ósseas/fisiopatologia , Linhagem Celular Tumoral , Membrana Celular/fisiologia , Expressão Gênica/efeitos dos fármacos , Humanos , Metaloproteinase 9 da Matriz/genética , Modelos Biológicos , Dados de Sequência Molecular , Osteossarcoma/genética , Osteossarcoma/patologia , Osteossarcoma/fisiopatologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Transdução de Sinais/efeitos dos fármacos
9.
Am J Physiol Lung Cell Mol Physiol ; 292(6): L1488-94, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17322284

RESUMO

Small cell lung cancer (SCLC) is a difficult disease to treat and sometimes has overexpression or mutation of c-Met receptor tyrosine kinase. The effects of c-Met/hepatocyte growth factor (c-Met/HGF, ligand for c-Met) on activation of reactive oxygen species (ROS) was determined. HGF stimulation of c-Met-overexpressing H69 SCLC cells (40 ng/ml, 15 min) resulted in an increase of ROS, measured with fluorescent probe 2'-7'-dichlorofluorescein diacetate (DCFH-DA) or dihydroethidine (DHE) but not in c-Met-null H446 cells. ROS was increased in juxtamembrane (JM)-mutated variants (R988C and T1010I) of c-Met compared with wild-type c-Met-expressing cells. ROS was significantly inhibited by preincubation of SCLC cells with pyrrolidine dithiocarbamate (PDTC, 100 microM) and/or SU11274 (small molecule c-Met tyrosine kinase inhibitor, 2 microM) for 3 h. PDTC and SU11274 also abrogated the HGF proliferative signal and cell motility in a cooperative fashion. H(2)O(2) treatment of SCLC cells (over 15 min) led to phosphorylation of c-Met receptor tyrosine kinase and further upregulated downstream phosphorylation of phospho-AKT, ERK1/2, and paxillin in a dose-dependent manner (125 microM to 500 microM). c-Met is an important target in lung cancer, and the pathways responsible for ROS generation together may provide novel therapeutic intervention.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Antioxidantes/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Indóis/farmacologia , Neoplasias Pulmonares/patologia , Muridae , Fosforilação , Piperazinas/farmacologia , Prolina/análogos & derivados , Prolina/farmacologia , Proteínas Proto-Oncogênicas c-met/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Sulfonamidas/farmacologia , Tiocarbamatos/farmacologia , Tirosina/metabolismo
10.
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
11.
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
12.
Cancer Res ; 66(1): 352-61, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16397249

RESUMO

c-Met receptor tyrosine kinase (RTK) has not been extensively studied in malignant pleural mesothelioma (MPM). In this study, c-Met was overexpressed and activated in most of the mesothelioma cell lines tested. Expression in MPM tissues by immunohistochemistry was increased (82%) in MPM in general compared with normal. c-Met was internalized with its ligand hepatocyte growth factor (HGF) in H28 MPM cells, with robust expression of c-Met. Serum circulating HGF was twice as high in mesothelioma patients as in healthy controls. There was a differential growth response and activation of AKT and extracellular signal-regulated kinase 1/2 in response to HGF for the various cell lines. Dose-dependent inhibition (IC50 < 2.5 micromol/L) of cell growth in mesothelioma cell lines, but not in H2052, H2452, and nonmalignant MeT-5A (IC50 > 10 micromol/L), was observed with the small-molecule c-Met inhibitor SU11274. Furthermore, migration of H28 cells was blocked with both SU11274 and c-Met small interfering RNA. Abrogation of HGF-induced c-Met and downstream signaling was seen in mesothelioma cells. Of the 43 MPM tissues and 7 cell lines, we have identified mutations within the semaphorin domain (N375S, M431V, and N454I), the juxtamembrane domain (T1010I and G1085X), and an alternative spliced product with deletion of the exon 10 of c-Met in some of the samples. Interestingly, we observed that the cell lines H513 and H2596 harboring the T1010I mutation exhibited the most dramatic reduction of cell growth with SU11274 when compared with wild-type H28 and nonmalignant MeT-5A cells. Ultimately, c-Met would be an important target for therapy against MPM.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Mesotelioma/metabolismo , Neoplasias Pleurais/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Ativação Enzimática , Fator de Crescimento Epidérmico/sangue , Fator de Crescimento de Hepatócito/sangue , Humanos , Indóis/farmacologia , Mesotelioma/tratamento farmacológico , Mesotelioma/genética , Mesotelioma/patologia , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Piperazinas/farmacologia , Neoplasias Pleurais/tratamento farmacológico , Neoplasias Pleurais/genética , Neoplasias Pleurais/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Interferente Pequeno/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/genética , Transdução de Sinais , Sulfonamidas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...