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.
Biochemistry ; 52(26): 4531-40, 2013 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-23731208

RESUMO

Soluble epidermal growth factor receptor (sEGFR) is a circulating serum biomarker in cancer patients. Recent studies suggest that baseline serum sEGFR concentrations may predict responsiveness to EGFR-targeted therapy. Here, we demonstrate that sEGFR is generated through proteolytic cleavage of a cell surface precursor of an alternately spliced EGF receptor isoform and that sEGFR binds to EGF with high affinity. Proteolytic cleavage is stimulated by an anti-α5/ß1 integrin antibody and 4-aminophenylmercuric acetate, and inhibited by fibronectin. Two FDA-approved therapeutic anti-EGFR antibodies also inhibit shedding of sEGFR, thus implicating the cell surface precursor of sEGFR as a competing target for anti-EGFR antibodies in human tissues. These observations parallel trastuzumab regulation of HER2 shedding and have implications for patient stratification in future clinical trials of EGFR-targeted antibodies.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Fator de Crescimento Epidérmico , Receptores ErbB , Integrinas/química , Células Neoplásicas Circulantes/química , Processamento Alternativo/efeitos dos fármacos , Animais , Anticorpos Anti-Idiotípicos/administração & dosagem , Biomarcadores Tumorais/química , Células CHO , Cetuximab , Ensaios Clínicos como Assunto , Cricetinae , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/sangue , Receptores ErbB/química , Humanos , Integrinas/antagonistas & inibidores , Integrinas/imunologia , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Isoformas de Proteínas/química
2.
Oncol Rep ; 27(3): 861-6, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22200873

RESUMO

Obesity is a significant risk factor for post-menopausal women to develop and die from breast cancer. Leptin, an adipokine is produced in high levels in obese individuals, and its receptor is overexpressed in breast tumors and lymph node metastases. Previously, we demonstrated that leptin stimulates breast cancer cell invasion, which is correlated with breast cancer metastasis. Programmed cell death 4 (PDCD4) has been shown to block cancer cell invasion. However, whether PDCD4 blocks leptin-induced breast cancer cell invasion is not known. Here, we report the novel findings that leptin failed to induce invasion in MCF-7 breast cancer cells overexpressing PDCD4 (MCF-7/PDCD4). Tissue inhibitor of metalloproteinase-2 (TIMP-2) was essential to the anti-invasive effect of PDCD4, as leptin stimulated the invasion of MCF-7/PDCD4 cells pretreated with TIMP-2 siRNA. Furthermore, TIMP-2 knockdown allowed leptin to augment phosphorylation of extracellular signal-regulated kinases 1,2 and signal transducer and activator of transcription 3, but not that of Jun N-terminal kinases. These data indicate that PDCD4 utilizes TIMP-2 to exert its anti-invasive effect by suppressing leptin-induced activation of extracellular signal-regulated kinases 1,2 and signal transducer and activator of transcription 3. Novel therapeutic strategies aiming at enhancing PDCD4 expression in breast tumors may be able to stop obesity-related breast tumor progression and prolong the life of patients.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Leptina/antagonistas & inibidores , Proteínas de Ligação a RNA/biossíntese , Proteínas Reguladoras de Apoptose/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Técnicas de Silenciamento de Genes/métodos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leptina/metabolismo , Leptina/farmacologia , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Fosforilação/genética , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Transfecção
3.
Oncotarget ; 2(4): 298-304, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21666286

RESUMO

Recent developments in multi-functional nanoparticles offer a great potential for targeted delivery of therapeutic compounds and imaging contrast agents to specific cell types, in turn, enhancing therapeutic effect and minimizing side effects. Despite the promise, site specific delivery carriers have not been translated into clinical reality. In this study, we have developed long circulating liposomes with the outer surface decorated with thioated oligonucleotide aptamer (thioaptamer) against E-selectin (ESTA) and evaluated the targeting efficacy and PK parameters. In vitro targeting studies using Human Umbilical Cord Vein Endothelial Cell (HUVEC) demonstrated efficient and rapid uptake of the ESTA conjugated liposomes (ESTA-lip). In vivo, the intravenous administration of ESTA-lip resulted in their accumulation at the tumor vasculature of breast tumor xenografts without shortening the circulation half-life. The study presented here represents an exemplary use of thioaptamer and liposome and opens the door to testing various combinations of thioaptamer and nanocarriers that can be constructed to target multiple cancer types and tumor components for delivery of both therapeutics and imaging agent.


Assuntos
Antineoplásicos/administração & dosagem , Aptâmeros de Nucleotídeos/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Lipossomos/uso terapêutico , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacocinética , Animais , Antineoplásicos/farmacocinética , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacocinética , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Carcinoma/irrigação sanguínea , Carcinoma/tratamento farmacológico , Carcinoma/patologia , Células Cultivadas , Portadores de Fármacos/uso terapêutico , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Humanos , Lipossomos/química , Lipossomos/farmacocinética , Camundongos , Camundongos Nus , Modelos Biológicos , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Compostos de Sulfidrila/química , Compostos de Sulfidrila/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Int J Oncol ; 38(4): 963-71, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21271218

RESUMO

Targeted therapy with reduced side effects is a major goal in cancer research. We investigated the effects of JS-K, a nitric oxide (NO) prodrug designed to release high levels of NO when suitably activated, on human breast cancer cell lines, on non-transformed human MCF-10A mammary cells, and on normal human mammary epithelial cells (HMECs). Cell viability assay, flow cytometry, electron microscopy, and Western blot analysis were used to study the effects of JS-K on breast cancer and on mammary epithelial cells. After a 3-day incubation, the IC50s of JS-K against the breast cancer cells ranged from 0.8 to 3 µM. However, JS-K decreased the viability of the MCF-10A cells by only 20% at 10-µM concentration, and HMECs were unaffected by 10 µM JS-K. Flow cytometry indicated that JS-K increased the percentages of breast cancer cells under-going apoptosis. Interestingly, flow cytometry indicated that JS-K increased acidic vesicle organelle formation in breast cancer cells, suggesting that JS-K induced autophagy in breast cancer cells. Electron microscopy confirmed that JS-K-treated breast cancer cells underwent autophagic cell death. Western blot analysis showed that JS-K induced the expression of microtubule light chain 3-II, another autophagy marker, in breast cancer cells. However, JS-K did not induce apoptosis or autophagy in normal human mammary epithelial cells. These data indicate that JS-K selectively induces programmed cell death in breast cancer cells while sparing normal mammary epithelial cells under the same conditions. The selective anti-tumor activity of JS-K warrants its further investigation in breast tumors.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Compostos Azo/farmacologia , Neoplasias da Mama/patologia , Glândulas Mamárias Humanas/efeitos dos fármacos , Óxido Nítrico/metabolismo , Piperazinas/farmacologia , Pró-Fármacos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Feminino , Heme Oxigenase-1/metabolismo , Humanos , Glândulas Mamárias Humanas/patologia , Proteínas Associadas aos Microtúbulos/metabolismo , Regulação para Cima
5.
Mol Cancer Ther ; 9(11): 3090-9, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21045138

RESUMO

The epidermal growth factor receptor (EGFR) signaling pathway has emerged as a promising target for cancer therapy. EGFR tyrosine kinase inhibitors (TKI) such as erlotinib have been approved for cancer treatment but have shown very limited activity in breast cancer patients. Clarifying the molecular mechanism underlying resistance to EGFR TKIs could lead to more effective treatment against breast cancer. We previously reported that the sensitivity of breast cancer cells to erlotinib is partially dependent on p27 and that cytoplasmic localization of p27 is associated with erlotinib resistance. In the present study, we found that erlotinib induces p27 phosphorylation at Ser¹° (S10), and S10 p27 phosphorylation leads to erlotinib resistance in EGFR-expressing breast cancer. Inhibiting S10 phosphorylation of p27 by knocking down human kinase-interacting stathmin (KIS), a nuclear protein that can phosphorylate p27 at S10, led to p27 accumulation in the nucleus and enhanced erlotinib-mediated cytotoxicity. Further, in vivo KIS gene silencing enhanced the antitumor activity of erlotinib in an orthotopic breast cancer xenograft model. KIS depletion also enhanced erlotinib sensitivity in erlotinib-resistant EGFR-expressing triple-negative breast cancer cells. Our study provides a rationale for the development of combinations of erlotinib with KIS inhibition to overcome EGFR TKI resistance in EGFR-expressing breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Carcinoma/tratamento farmacológico , Inibidor de Quinase Dependente de Ciclina p27/antagonistas & inibidores , Quinazolinas/uso terapêutico , Estatmina/genética , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Cloridrato de Erlotinib , Feminino , Inativação Gênica/fisiologia , Genes erbB-1 , Humanos , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Quinases/metabolismo , Quinazolinas/administração & dosagem , Quinazolinas/farmacologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Serina/metabolismo , Estatmina/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
6.
PLoS One ; 5(9)2010 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-20927342

RESUMO

Active targeting of a drug carrier to a specific target site is crucial to provide a safe and efficient delivery of therapeutics and imaging contrast agents. E-selectin expression is induced on the endothelial cell surface of vessels in response to inflammatory stimuli but is absent in the normal vessels. Thus, E-selectin is an attractive molecular target, and high affinity ligands for E-selectin could be powerful tools for the delivery of therapeutics and/or imaging agents to inflamed vessels. In this study, we identified a thiophosphate modified aptamer (thioaptamer, TA) against E-selectin (ESTA-1) by employing a two-step selection strategy: a recombinant protein-based TA binding selection from a combinatorial library followed by a cell-based TA binding selection using E-selectin expressing human microvascular endothelial cells. ESTA-1 selectively bound to E-selectin with nanomolar binding affinity (K(D) = 47 nM) while exhibiting minimal cross reactivity to P- and L-selectin. Furthermore, ESTA-1 binding to E-selectin on the endothelial cells markedly antagonized the adhesion (over 75% inhibition) of sLe(x) positive HL-60 cells at nanomolar concentration. ESTA-1 also bound specifically to the inflamed tumor-associated vasculature of human carcinomas derived from breast, ovarian, and skin but not to normal organs, and this binding was highly associated with the E-selectin expression level. Similarly, intravenously injected ESTA-1 demonstrated distinct binding to the tumor vasculature in a breast cancer xenograft model. Together, our data substantiates the discovery of a thioaptamer (ESTA-1) that binds to E-selectin with high affinity and specificity, thereby highlighting the potential application of ESTA-1 for E-selectin targeted delivery.


Assuntos
Aptâmeros de Nucleotídeos/farmacologia , Circulação Sanguínea/efeitos dos fármacos , Selectina E/metabolismo , Células Endoteliais/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Animais , Aptâmeros de Nucleotídeos/síntese química , Aptâmeros de Nucleotídeos/química , Sequência de Bases , Ensaios de Seleção de Medicamentos Antitumorais , Selectina E/química , Selectina E/genética , Células Endoteliais/química , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células HL-60 , Humanos , Cinética , Ligantes , Camundongos , Dados de Sequência Molecular , Neoplasias/imunologia , Neoplasias/fisiopatologia , Conformação de Ácido Nucleico , Fosfatos/química , Ligação Proteica
7.
Cancer Res ; 70(9): 3687-96, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20430760

RESUMO

RNA interference (RNAi) is a powerful approach for silencing genes associated with a variety of pathologic conditions; however, in vivo RNAi delivery has remained a major challenge due to lack of safe, efficient, and sustained systemic delivery. Here, we report on a novel approach to overcome these limitations using a multistage vector composed of mesoporous silicon particles (stage 1 microparticles, S1MP) loaded with neutral nanoliposomes (dioleoyl phosphatidylcholine, DOPC) containing small interfering RNA (siRNA) targeted against the EphA2 oncoprotein, which is overexpressed in most cancers, including ovarian. Our delivery methods resulted in sustained EphA2 gene silencing for at least 3 weeks in two independent orthotopic mouse models of ovarian cancer following a single i.v. administration of S1MP loaded with EphA2-siRNA-DOPC. Furthermore, a single administration of S1MP loaded with-EphA2-siRNA-DOPC substantially reduced tumor burden, angiogenesis, and cell proliferation compared with a noncoding control siRNA alone (SKOV3ip1, 54%; HeyA8, 57%), with no significant changes in serum chemistries or in proinflammatory cytokines. In summary, we have provided the first in vivo therapeutic validation of a novel, multistage siRNA delivery system for sustained gene silencing with broad applicability to pathologies beyond ovarian neoplasms.


Assuntos
Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Silício/administração & dosagem , Animais , Linhagem Celular Tumoral , Feminino , Inativação Gênica , Terapia Genética/métodos , Humanos , Lipossomos/administração & dosagem , Lipossomos/química , Lipossomos/farmacocinética , Camundongos , Camundongos Nus , Nanopartículas/química , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/terapia , Fosfatidilcolinas/administração & dosagem , Fosfatidilcolinas/química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacocinética , Receptor EphA2/genética , Silício/química , Silício/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Clin Exp Metastasis ; 26(3): 197-204, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19112600

RESUMO

In breast tumors, high levels of leptin have been associated with increased incidence of breast cancer metastasis. Breast cancer metastasis is directly associated with breast cancer cell invasion. However, whether leptin could augment breast cancer cell invasion is not known. Here we showed that leptin increased the invasiveness and the matrix metallo-proteinase-2 (MMP-2) activity of the MCF-7 breast cancer cell line. Leptin stimulated the phosphorylation of extracellular signals regulated kinases, signal transducers and activators of transcription 3 and Jun N-terminal kinases (JNK); however, only inhibition of JNK decreased leptin-mediated activation of MMP-2. Furthermore, inhibition of JNK suppressed leptin-mediated breast cancer cell invasion. Here we report the novel findings that leptin increased invasion of breast cancer cells by activating JNK, resulting in increased MMP-2 activity.


Assuntos
Neoplasias da Mama/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Leptina/fisiologia , Metaloproteinase 2 da Matriz/metabolismo , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Feminino , Humanos , Leptina/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Invasividade Neoplásica , Fosforilação , Transdução de Sinais
9.
Breast Cancer Res Treat ; 114(2): 203-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18386173

RESUMO

High levels of the cyclooxygenase-2 (COX-2) protein have been associated with invasion and metastasis of breast tumors. Both prostaglandin E(2) (PGE(2)) and interleukin-8 (IL-8) have been shown to mediate the invasive activity of COX-2 in breast cancer cells. Here we expand these studies to determine how COX-2 uses PGE(2) and IL-8 to induce breast cancer cell invasion. We demonstrated that PGE(2) and IL-8 decreased the expression of the tumor suppressor protein Programmed Cell Death 4 (PDCD4). We hypothesized that suppression of PDCD4 expression is vital to the invasive activity of PGE(2) and IL-8. In MCF-7 cells overexpressing PDCD4 (MCF-7/PDCD4), PGE(2) and IL-8 failed to induce invasion, in contrast to the parental MCF-7 cells, thus indicating that PDCD4 blocks breast cancer cell invasion. MCF-7/PDCD4 cells produced higher levels of the Tissue Inhibitor of Metalloproteinases-2 (TIMP-2) than the parental cells. Silencing TIMP-2 mRNA in MCF-7/PDCD4 cells reversed the anti-invasive effects of PDCD4, allowing PGE(2) and IL-8 to induce the invasion of these cells. Here we report the novel findings that suppression of PDCD4 expression is vital for the invasive activity of COX-2 mediated by PGE(2) and IL-8, and that PDCD4 increases TIMP-2 expression to inhibit breast cancer cell invasion.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Neoplasias da Mama/patologia , Proteínas de Ligação a RNA/genética , Inibidor Tecidual de Metaloproteinase-2/genética , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Invasividade Neoplásica , Interferência de RNA/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Células Tumorais Cultivadas
10.
Exp Cell Res ; 314(16): 2907-18, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18687326

RESUMO

Soluble isoforms of the epidermal growth factor receptor (sEGFR) previously have been identified in the conditioned culture media (CCM) of the vulvar adenocarcinoma cell line, A431 and within exosomes of the keratinocyte cell line HaCaT. Here, we report that the extracellular domain (ECD) of EGFR is shed from the cell surface of human carcinoma cell lines that express 7x10(5) receptors/cell or more. We purified this proteolytic isoform of EGFR (PI-sEGFR) from the CCM of MDA-MB-468 breast cancer cells. The amino acid sequence of PI-sEGFR was determined by reverse-phase HPLC nano-electrospray tandem mass spectrometry of peptides generated by trypsin, chymotrypsin or GluC digestion. The PI-sEGFR protein is identical in amino acid sequence to the EGFR ECD. The release of PI-sEGFR from MDA-MB-468 cells is enhanced by phorbol 12-myristate 13-acetate, heat-inactivated fetal bovine serum, pervanadate, and EGFR ligands (i.e., EGF and TGF-alpha). In addition, 4-aminophenylmercuric acetate, an activator of metalloproteases, increased PI-sEGFR levels in the CCM of MDA-MB-468 cells. Inhibitors of metalloproteases decreased the constitutive shedding of EGFR while the PMA-induced shedding was inhibited by metalloprotease inhibitors, by the two serine protease inhibitors leupeptin and 3,4-dichloroisocoumarin (DCI), and by the aspartyl inhibitor pepstatin. These results suggest that PI-sEGFR arises by proteolytic cleavage of EGFR via a mechanism that is regulated by both PKC- and phosphorylation-dependent pathways. Our results further suggest that when proteolytic shedding of EGFR does occur, it is correlated with a highly malignant phenotype.


Assuntos
Receptores ErbB/metabolismo , Neoplasias/metabolismo , Isoformas de Proteínas/metabolismo , Sequência de Aminoácidos , Animais , Bovinos , Linhagem Celular Tumoral , Ativação Enzimática , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/química , Receptores ErbB/genética , Humanos , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Acetato de Fenilmercúrio/análogos & derivados , Acetato de Fenilmercúrio/metabolismo , Inibidores de Proteases/metabolismo , Isoformas de Proteínas/genética , Proteína Quinase C/metabolismo , Estrutura Terciária de Proteína , Alinhamento de Sequência , Espectrometria de Massas por Ionização por Electrospray , Acetato de Tetradecanoilforbol/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Vanadatos/metabolismo
11.
Breast Cancer Res ; 10(3): R44, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18474097

RESUMO

INTRODUCTION: Tumor invasion and metastasis remain a major cause of mortality in breast cancer patients. High concentrations of nitric oxide (NO) suppress tumor invasion and metastasis in vivo. NO prodrugs generate large amounts of NO upon metabolism by appropriate intracellular enzymes, and therefore could have potential in the prevention and therapy of metastatic breast cancer. METHODS: The present study was designed to determine the effects of the NO-releasing prodrug O2-(2,4-dinitrophenyl) 1- [(4-ethoxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate (JS-K) on breast cancer invasion and the mechanisms involved. MDA-MB-231, MDA-MB-231/F10, and MCF-7/COX-2 were the three breast cancer cell lines tested. NO levels were determined spectrophotometrically using a NO assay kit. Invasion and the expression of matrix metalloproteinases (MMPs) and tissue inhibitor of MMPs were determined using Matrigel invasion assays, an MMP array kit and ELISAs. The activity and expression of extracellular signal-regulated kinase 1/2, p38, and c-Jun N-terminal kinase mitogen-activated protein kinases were determined using western blot analyses. RESULTS: Under conditions by which JS-K was not cytotoxic, JS-K significantly decreased (P < 0.05) the invasiveness of breast cancer cells across the Matrigel basement membrane, which was directly correlated with NO production. JS-43-126, a non-NO-releasing analog of JS-K, had no effect on NO levels or invasion. JS-K increased (P < 0.05) TIMP-2 production, and blocking TIMP-2 activity with a neutralizing antibody significantly increased (P < 0.05) the invasive activity of JS-K-treated cells across Matrigel. JS-K decreased p38 activity, whereas the activity and the expression of extracellular signal-regulated kinase 1/2 and c-Jun N-terminal kinase were unaffected. CONCLUSION: We report the novel findings that JS-K inhibits breast cancer invasion across the Matrigel basement membrane, and NO production is vital for this activity. Upregulation of TIMP-2 production is one mechanism by which JS-K mediates its anti-invasive effects. JS-K and other NO prodrugs may represent an innovative biological approach in the prevention and treatment of metastatic breast cancer.


Assuntos
Antineoplásicos/farmacologia , Compostos Azo/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Invasividade Neoplásica , Óxido Nítrico/metabolismo , Piperazinas/farmacologia , Pró-Fármacos/farmacologia , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Membrana Basal/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Colágeno/química , Combinação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Humanos , Laminina/química , Modelos Químicos , Metástase Neoplásica , Proteoglicanas/química
12.
Int J Oncol ; 30(4): 785-92, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17332916

RESUMO

Cyclooxygenase-2 (COX-2) increases breast cancer cell invasion. Expression of various pro-angiogenic and pro-invasive factors has been correlated with high expression of COX-2. However, whether these factors are essential to COX-2-mediated breast cancer invasion, and the mechanisms by which COX-2 increases the expression of these factors are unknown. Our microarray results indicate that higher COX-2 expression was associated with increased levels of interleukin-8 (IL-8), a key factor in breast cancer invasion and metastasis. COX-2 overexpressing cells (MCF-7/COX-2), generated by transfecting COX-2-encoding plasmids into the poorly invasive MCF-7 breast cancer cells, were more invasive and produced higher IL-8 levels than the parental cells. To investigate the role of IL-8 in COX-2-mediated invasion, MCF-7 parental cells were incubated with IL-8. Exogenous IL-8 increased the invasiveness of MCF-7 cells. IL-8 is one pathway by which COX-2 mediates breast cancer invasion. Protein kinase A (PKA) and protein kinase C (PKC) are activated by COX-2 and are involved in IL-8 regulation. Inhibition of PKC, not PKA, decreased IL-8 production and invasion in MCF-7/COX-2 cells. Activation of PKC, not PKA, increased IL-8 production and invasion in MCF-7 cells. Thus, the invasive effects of COX-2 are mediated by PKC, not PKA. Activity of the urokinase-type plasminogen activator (uPA) was increased in MCF-7 cells by COX-2 overexpression or by the addition of a PKC activator or by IL-8. Inhibition of PKC decreased uPA activity in MCF-7/COX-2 cells. Furthermore, inhibition of uPA activity decreased the invasiveness of MCF-7/COX-2 cells, indicating that uPA was essential to COX-2-mediated invasion. Herein we demonstrate for the first time a detailed mechanism by which COX-2 increases breast cancer invasion: the PKC/IL-8/uPA pathway.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/patologia , Ciclo-Oxigenase 2/metabolismo , Interleucina-8/metabolismo , Proteínas de Membrana/metabolismo , Proteína Quinase C/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/genética , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Membrana/genética , Invasividade Neoplásica , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...