Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogenesis ; 6(8): e369, 2017 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-28785073

RESUMO

The coupling between DNA methylation and histone modification contributes to aberrant expression of oncogenes or tumor suppressor genes that leads to tumor development. Our previous study demonstrated that lysine demethylase 2A (KDM2A) functions as an oncogene in breast cancer by promoting cancer stemness and angiogenesis via activation of the Notch signaling. Here, we demonstrate that knockdown of KDM2A significantly increases the 5'-hydroxymethylcytosine (5'-hmc) level in genomic DNA and expression of tet-eleven translocation 2 (TET2) in various breast cancer cell lines. Conversely, ectopic expression of KDM2A inhibits TET2 expression in KDM2A-depleted cells suggesting TET2 is a transcriptional repression target of KDM2A. Our results show that KDM2A interacts with RelA to co-occupy at the TET2 gene promoter to repress transcription and depletion of RelA or KDM2A restores TET2 expression. Upregulation of TET2 in the KDM2A-depleted cells induces the re-activation of two TET downstream tumor suppressor genes, epithelial cell adhesion molecule (EpCAM) and E-cadherin, and inhibits migration and invasion. On the contrary, knockdown of TET2 in these cells decreases EpCAM and E-cadherin and increases cell invasiveness. More importantly, TET2 expression is negatively associated KDM2A in triple-negative breast tumor tissues, and its expression predicts a better survival. Taken together, we demonstrate for the first time that TET2 is a direct repression target of KDM2A and reveal a novel mechanism by which KDM2A promotes DNA methylation and breast cancer progression via the inhibition of a DNA demethylase.

2.
Oncogene ; 36(39): 5532-5543, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28581520

RESUMO

Krüppel-like transcription factor 10 (KLF10), also named as TIEG1, plays essential roles in mediating transforming growth factor beta (TGFß) signaling and has been shown to function as a tumor suppressor in multiple cancer types. However, its roles in mediating cancer progression in vivo have yet to be fully characterized. Here, we have employed two well-characterized Pdx-1CreLSL-KrasG12D and Pdx-1CreLSL-KrasG12Dp53L/L pancreatic cancer models to ablate KLF10 expression and determine the impact of KLF10 deletion on tumor development and progression. We show that loss of KLF10 cooperates with KrasG12D leading to an invasive and widely metastatic phenotype of pancreatic ductal adenocarcinoma (PDAC). Mechanistically, loss of KLF10 in PDAC is shown to increase distant metastases and cancer stemness through activation of SDF-1/CXCR4 and AP-1 pathways. Furthermore, we demonstrate that targeting the SDF-1/CXCR4 pathway in the context of KLF10 deletion substantially suppresses PDAC progression suggesting that inhibition of this pathway represents a novel therapeutic strategy for PDAC treatment.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Quimiocina CXCL12/metabolismo , Fatores de Transcrição de Resposta de Crescimento Precoce/deficiência , Fatores de Transcrição Kruppel-Like/deficiência , Neoplasias Pancreáticas/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Quimiocina CXCL12/genética , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Taxa de Sobrevida , Proteína Supressora de Tumor p53/genética
3.
Oncogene ; 35(17): 2223-34, 2016 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-26411367

RESUMO

Adenomatous polyposis coli (APC), a tumor-suppressor gene critically involved in familial adenomatous polyposis, is integral in Wnt/ß-catenin signaling and is implicated in the development of sporadic tumors of the distal gastrointestinal tract including pancreatic cancer (PC). Here we report for the first time that functional APC is required for the growth and maintenance of pancreatic islets and maturation. Subsequently, a non-Kras mutation-induced premalignancy mouse model was developed; in this model, APC haploinsufficiency coupled with p53 deletion resulted in the development of a distinct type of pancreatic premalignant precursors, mucinous cystic neoplasms (MCNs), exhibiting pathomechanisms identical to those observed in human MCNs, including accumulation of cystic fluid secreted by neoplastic and ovarian-like stromal cells, with 100% penetrance and the presence of hepatic and gastric metastases in >30% of the mice. The major clinical implications of this study suggest targeting the Wnt signaling pathway as a novel strategy for managing MCN.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Pancreáticas/genética , Proteína Supressora de Tumor p53/genética , Animais , Modelos Animais de Doenças , Feminino , Haploinsuficiência/genética , Humanos , Perda de Heterozigosidade , Camundongos , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Pancreáticas/patologia , Via de Sinalização Wnt/genética , Neoplasias Pancreáticas
4.
Blood Cancer J ; 5: e313, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25978433

RESUMO

Epigenetic modifying enzymes have a crucial role in the pathogenesis of acute myeloid leukemia (AML). Methylation of lysine 9 on histone H3 by the methyltransferase G9a and SUV39H1 is associated with inhibition of tumor suppressor genes. We studied the effect of G9a and SUV39H1 inhibitors on viability and differentiation of AML cells and tested the cytotoxicity induced by combination of G9a and SUV39H1 inhibitors and various epigenetic drugs. The SUV39H1 inhibitor (chaetocin) and the G9a inhibitor (UNC0638) caused cell death in AML cells at high concentrations. However, only chaetocin-induced CD11b expression and differentiation of AML cells at non-cytotoxic concentration. HL-60 and KG-1a cells were more sensitive to chaetocin than U937 cells. Long-term incubation of chaetocin led to downregulation of SUV39H1 and reduction of H3K9 tri-methylation in HL-60 and KG-1a cells. Combination of chaetocin with suberoylanilide hydroxamic acid (SAHA, a histone deacetylase inhibitor) or JQ (a BET (bromodomain extra terminal) bromodomain inhibitor) showed synergistic cytotoxicity. Conversely, no synergism was found by combining chaetocin and UNC0638. More importantly, chaetocin-induced differentiation and combined cytotoxicity were also found in the primary cells of AML patients. Collectively, the SUV39H1 inhibitor chaetocin alone or in combination with other epigenetic drugs may be effective for the treatment of AML.


Assuntos
Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Metiltransferases/antagonistas & inibidores , Proteínas Repressoras/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Células Cultivadas , Sinergismo Farmacológico , Epigênese Genética , Humanos , Immunoblotting , Piperazinas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Oncogene ; 34(35): 4558-69, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-25435370

RESUMO

Interstitial fluid flow in and around the tumor tissue is a physiologically relevant mechanical signal that regulates intracellular signaling pathways throughout the tumor. Yet, the effects of interstitial flow and associated fluid shear stress on the tumor cell function have been largely overlooked. Using in vitro bioengineering models in conjunction with molecular cell biology tools, we found that fluid shear (2 dyn/cm(2)) markedly upregulates matrix metalloproteinase 12 (MMP-12) expression and its activity in human chondrosarcoma cells. MMP-12 expression is induced in human chondrocytes during malignant transformation. However, the signaling pathway regulating MMP-12 expression and its potential role in human chondrosarcoma cell invasion and metastasis have yet to be delineated. We discovered that fluid shear stress induces the synthesis of insulin growth factor-2 (IGF-2) and vascular endothelial growth factor (VEGF) B and D, which in turn transactivate MMP-12 via PI3-K, p38 and JNK signaling pathways. IGF-2-, VEGF-B- or VEGF-D-stimulated chondrosarcoma cells display markedly higher migratory and invasive potentials in vitro, which are blocked by inhibiting MMP-12, PI3-K, p38 or JNK activity. Moreover, recombinant human MMP-12 or MMP-12 overexpression can potentiate chondrosarcoma cell invasion in vitro and the lung colonization in vivo. By reconstructing and delineating the signaling pathway regulating MMP-12 activation, potential therapeutic strategies that interfere with chondrosarcoma cell invasion may be identified.


Assuntos
Neoplasias Ósseas/enzimologia , Condrossarcoma/enzimologia , Fator de Crescimento Insulin-Like II/fisiologia , Neoplasias Pulmonares/enzimologia , Metaloproteinase 12 da Matriz/metabolismo , Fator B de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Animais , Fenômenos Biomecânicos , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular , Condrossarcoma/secundário , Ativação Enzimática , Indução Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/secundário , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Invasividade Neoplásica , Transplante de Neoplasias
6.
Br J Cancer ; 109(3): 731-8, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23820254

RESUMO

BACKGROUND: Signal transducer and activator of transcription 3 (STAT3) activation is frequently found in human lung cancer and is associated with increased metastasis and reduced survival. How STAT3 enhances invasiveness is unclear. METHODS: The expression of microRNAs and target genes was measured by real-time RT-PCR. Protein level was studied by western blotting. Luciferase reporter assay was used to confirm the direct targeting of microRNAs. Gelatin zymography was used to study matrix metalloproteinase (MMP) activity. Transwell assay was used to investigate cell migration and invasion. RESULTS: Enforced expression of STAT3 decreases the endogenous MMP inhibitor RECK protein but not mRNA level in H460 cells. Conversely, STAT3 inhibitor S3I-201 increases RECK protein in STAT3-activating H1299 cells. We demonstrate that STAT3 upregulates miR-92a to repress RECK via post-transcriptional inhibition. The RECK 3'-untranslated region (3'UTR) reporter activity assay suggests that RECK is a direct repression target of miR-92a. Delivery of pre-miR-92a reduces RECK protein level whereas transfection of anti-miR-92a restores STAT3-induced downregulation of RECK. Anti-miR-92a attenuates MMP activity, migration and invasion of H1299 cells and STAT3-overexpressing H460 cells, suggesting miR-92a is critical for STAT3-induced invasiveness. CONCLUSION: The STAT3-induced miR-92a promotes cancer invasion by suppressing RECK and targeting of the STAT3/miR-92a axis may be helpful for cancer treatment.


Assuntos
Proteínas Ligadas por GPI/antagonistas & inibidores , MicroRNAs/genética , Fator de Transcrição STAT3/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proteínas Ligadas por GPI/biossíntese , Proteínas Ligadas por GPI/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/metabolismo , Invasividade Neoplásica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fator de Transcrição STAT3/metabolismo , Transcrição Gênica , Transfecção , Regulação para Cima
7.
Curr Pharm Biotechnol ; 12(5): 839-46, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21446905

RESUMO

Three blending methods were introduced to combine a biodegradable cationic- polyurethane (PUg3) and polyethylenimine (PEI) together with DNA by different mixing sequences. Results of gel electrophoresis assays and particle size measurements show that complexes prepared by method 1 and 3 bear an ability to condense DNA into small nanoparticles. On the contrary, the use of method 2 in making complexes produces significantly large particles because of the weaker interaction with DNA and lack of DNA condensation. Moreover, cell proliferation assays show that no cytotoxicity of the DNA/blended-polymers complexes (exhibited by method 1) was found and due to a result of the outer coating of PUg3, reducing cytotoxic PEI exposure outside the complexes. With a new technique in pharmaceutics, the complexes prepared for DNA delivery by mixing of PEI and PUg3 with DNA in a sequence (method 1) could achieve an even better transfection efficiency (reaching 40% higher) than using PEI alone as well as reduce the cytotoxicity substantially. In conclusion, a new class of complexes (non-viral combo-system) made by a skillful blending sequence (method 1) has been designed and demonstrated to obtain the beneficial properties from two useful and individual polymers for gene delivery. This method can be used in greatly improving the transfection efficiency of polymer-based gene vectors. The blended polymers with DNA also have a better biocompatibility and no cytotoxicity, which are the requirements and critical points for great success in performing gene therapy in vivo.


Assuntos
DNA/administração & dosagem , Polietilenoimina/química , Poliuretanos/química , Transfecção/métodos , Animais , Células COS , Cátions/administração & dosagem , Cátions/química , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , DNA/química , DNA/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Nanopartículas/administração & dosagem , Nanopartículas/química , Tamanho da Partícula , Polietilenoimina/administração & dosagem , Poliuretanos/administração & dosagem , Células Tumorais Cultivadas
8.
Lasers Surg Med ; 41(5): 391-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19533759

RESUMO

BACKGROUND AND OBJECTIVES: Multidrug-resistant (MDR) Pseudomonas aeruginosa infection is becoming a critical problem worldwide. Currently, only limited therapeutic options are available for the treatment of infections caused by MDR P. aeruginosa, therefore, the development of new alternative treatments is needed. Toluidine blue O (TBO) is an effective antibacterial photosensitizing agent against various bacteria. However, reports on antibacterial photosensitization of MDR bacteria are limited. This study aims to determine the in vitro photobactericidal activity of TBO against MDR P. aeruginosa. STUDY DESIGN/MATERIALS AND METHODS: The efficacy of antibacterial photodynamic inactivation, DNA fragmentation and protein carbonylation of three MDR P. aeruginosa strains and one susceptible strain was compared using TBO as the photosensitizer followed by red light irradiation (630 nm, 90 J/cm(2)) from a light-emitting diode light source. Subsequently, the efficacy of TBO photodynamic inactivation (TBO-PDI) on 60 MDR strains, including 11 with the efflux pump phenotype and 49 with no pump activity, was tested using the minimum lethal drug concentration (MLC) assay. RESULTS: TBO-PDI caused similar bactericidal effect (6-7 logs of killing effect), DNA fragmentation and protein carbonylation in three MDR and one susceptible P. aeruginosa strains. Although the TBO accumulation assay indicated that TBO is a substrate for the efflux pump, TBO-PDI produce similar photobactericidal activity against 60 MDR P. aeruginosa strains, either with or without efflux-pump phenotype, and 19 susceptible strains. CONCLUSION: MDR did not affect the susceptibility of P. aeruginosa strains to TBO-PDI. The efflux pump played an insignificant role in TBO-PDI of MDR P. aeruginosa.


Assuntos
Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla/efeitos da radiação , Fotoquimioterapia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos da radiação , Cloreto de Tolônio/farmacologia , Humanos , Testes de Sensibilidade Microbiana
9.
Cancer Gene Ther ; 15(6): 393-401, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18369382

RESUMO

Increasing the specificity of chemotherapy may improve the efficacy of cancer treatment. Toward this aim, we developed a strain of bacteria to express enzymes for selective prodrug activation and non-invasive imaging in tumors. beta-glucuronidase and the luxCDABE gene cluster were expressed in the DH5alpha strain of Escherichia coli to generate DH5alpha-lux/betaG. These bacteria emitted light for imaging and hydrolyzed the glucuronide prodrug 9ACG to the topoisomerase I inhibitor 9-aminocamptothecin (9AC). By optical imaging, colony-forming units (CFUs) and staining for betaG activity, we found that DH5alpha-lux/betaG preferentially localized and replicated within CL1-5 human lung tumors in mice. The intensity of luminescence, CFU and betaG activity increased with time, indicating bacterial replication occurred in tumors. In comparison with DH5alpha-lux/betaG, 9AC or 9ACG treatment, combined systemic administration of DH5alpha-lux/betaG followed by 9ACG prodrug treatment significantly (P<0.005) delayed the growth of CL1-5 tumors. Our results demonstrate that prodrug-activating bacteria may be useful for selective cancer chemotherapy.


Assuntos
Bactérias/metabolismo , Neoplasias/terapia , Pró-Fármacos/uso terapêutico , Animais , Bactérias/genética , Glucuronidase/genética , Glucuronidase/metabolismo , Glucuronídeos/metabolismo , Humanos , Modelos Biológicos , Neoplasias/microbiologia , Neoplasias/patologia , Pró-Fármacos/metabolismo
10.
J Clin Pathol ; 61(5): 658-64, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-17908805

RESUMO

BACKGROUND: Hypoxia inducible factor (HIF)-1alpha is a critical regulatory protein of cellular response to hypoxia and is closely related to angiogenic process. AIMS: To explore the potential role and the prognostic value of HIF-1alpha in urothelial carcinoma (UC). METHODS: Clinicopathological and follow-up data on 99 UC cases were reviewed and immunostained for HIF-1alpha, CD68, vascular endothelial growth factor (VEGF) and CD34 antigen. Tumour-associated macrophage (TAM) counts and HIF-1alpha expression were compared with clinicopathologic characteristics, overall survival (OS) and disease-free survival rates (DFS). RESULTS: High expression of HIF-1alpha was detected in 55 of 99 (55.6%) tumours. HIF-1alpha expression was correlated with tumour size, histological grade, tumour invasiveness and recurrence. VEGF and microvessel density (MVD) demonstrated their positive correlation with HIF-1alpha overexpression, supporting the correlation of HIF-1alpha up-regulation with tumour angiogenesis. Higher TAM infiltration was identified in high expression of HIF-1alpha cases rather than HIF-1alpha low expression cases (p = 0.002). Kaplan-Meier analysis found that HIF-1alpha overexpression and high TAM count was only associated with worse DFS (p = 0.009, p = 0.023) but was not associated with OS (p = 0.696, p = 0.141). Multivariate analyses indicated only tumour size (p = 0.038) to be an independently significant prognostic factor for OS, in addition, HIF-1alpha expression (p = 0.011), as well as histological grade (p = 0.038), and MVD (p = 0.004), to be independently significant prognostic factors for DFS. CONCLUSIONS: Our results indicate that HIF-1alpha is a key regulator of the angiogenic cascade. We show that HIF-1alpha is an independent prognostic factor for disease-free survival.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células de Transição/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Macrófagos/patologia , Neovascularização Patológica/metabolismo , Neoplasias da Bexiga Urinária/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células de Transição/irrigação sanguínea , Carcinoma de Células de Transição/patologia , Progressão da Doença , Feminino , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Prognóstico , Análise de Sobrevida , Neoplasias da Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Histopathology ; 51(6): 785-92, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17953697

RESUMO

AIMS: The mechanisms of urothelial carcinogenesis in areas highly contaminated with arsenic remain unclear. The aim was to determine whether hypermethylation of death-associated protein kinase (DAPK) gene is associated with chronic arsenic exposure. METHODS AND RESULTS: The frequency of aberrant promoter methylation of DAPK in 17 urothelial carcinomas from an arsenic-contaminated area and 21 urothelial carcinomas from a non-arsenic-contaminated area was determined by methylation-specific polymerase chain reaction. DAPK hypermethylation status was significantly higher in urothelial cancers arising in arsenic-contaminated areas when compared with tumours from patients from non-contaminated areas (P = 0.018). In the subset of patients from living environments which were contaminated with arsenic, there was a statistically significant association between DAPK hypermethylation and patient's age, tumour invasiveness, histological grade and recurrence. This was not seen for urothelial carcinoma from patients from non-contaminated areas. A close correlation was also found between DAPK promoter methylation and low-intensity DAPK expression, as detected by immunohistochemistry (P = 0.037). CONCLUSION: Exposure to arsenic may induce DAPK promoter hypermethylation and inactivate the function of DAPK in urothelial carcinoma. This could prove to be a key molecular event contributing to the malignant phenotype of tumour arising in patients from arsenic-contaminated environments.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Intoxicação por Arsênico/genética , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Carcinoma/genética , Metilação de DNA , Neoplasias da Bexiga Urinária/genética , Idoso , Biomarcadores Tumorais/genética , Carcinoma/induzido quimicamente , Proteínas Quinases Associadas com Morte Celular , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Taiwan/epidemiologia , Neoplasias da Bexiga Urinária/induzido quimicamente , Urotélio/efeitos dos fármacos , Urotélio/patologia
12.
FEBS Lett ; 508(3): 365-8, 2001 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-11728453

RESUMO

Recent studies show that up-regulation of cyclooxygenase-2 (COX-2) in human cancer cells induces activation of matrix metalloproteinases (MMPs) and increase of metastatic potential. In this study, we investigate the effect of a COX-2 selective inhibitor, NS398, on the expression and enzymatic activity of MMPs in human lung cancer cells. We found that NS398 inhibited MMP-2, not MMP-9, mRNA expression. NS398 also reduced the amount of MMP-2, not MMP-9, released into the medium. Additionally, this COX-2 inhibitor attenuated the degrading activity of MMP-2 as demonstrated by gelatin zymography. Investigation of cellular MMP-2 by Western blotting indicated that synthesis and processing of MMP-2 was significantly suppressed by NS398. We performed promoter activity assay to address whether NS398 might affect MMP-2 gene transcription. Our results indicated that NS398 directly inhibited MMP-2 promoter activity. However, the inhibitory effect of NS398 is not fully dependent on inhibition of COX-2 because a high concentration of NS398 was needed to suppress MMP-2 expression and addition of prostaglandin E2 only partially reversed the action of NS398. Moreover, a non-selective COX inhibitor indomethacin also suppressed the expression of MMP-2. Taken together, these results indicate that non-steroidal anti-inflammatory drugs suppress MMP-2 expression via repression of transcription and support the notion that COX inhibitors may be useful in inhibition and/or prevention of metastasis.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/enzimologia , Metaloproteinase 2 da Matriz/genética , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Meios de Cultivo Condicionados , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/farmacologia , Relação Dose-Resposta a Droga , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Células Tumorais Cultivadas
13.
Am J Kidney Dis ; 38(5): 1096-104, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11684565

RESUMO

Advanced glycation end products (AGEs) are important in the pathogenesis of diabetic nephropathy, which leads to renal fibrosis. Previously, we found that the janus kinase (JAK)/signal transducers and activators of transcription (STAT) signaling pathway is necessary for AGE-induced cellular proliferation in normal rat kidney interstitial fibroblast (NRK-49F) cells. However, a direct link between JAK/STAT and cell-cycle progression has not been well established. In this regard, STAT5 has been found to induce cyclin D1 and proliferation in hematopoietic cells. Therefore, we examined effects of AGE on STAT5 and cell-cycle-dependent mitogenesis in NRK-49F cells. We found that AGE increased cyclin D1 expression and cyclin-dependent kinase (cdk)4 activity while decreasing p21(WAF1/CIP1) expression. We also found that AGE (100 microg/mL) induced STAT5 tyrosine phosphorylation. Meanwhile, AGE induced STAT5 protein-DNA binding activity, which was reversed by AG-490 (a specific JAK2 inhibitor) and STAT5 decoy oligodeoxynucleotide (ODN). In addition, STAT5 decoy ODN reversed AGE-induced cell-cycle-dependent cellular proliferation and cyclin D1 protein expression. We concluded that AGE induced cell-cycle-dependent cellular proliferation by inducing the JAK2-STAT5-cyclin D1 and cdk4 pathways in NRK-49F cells.


Assuntos
Divisão Celular/efeitos dos fármacos , Ciclina D1/efeitos dos fármacos , Proteínas de Ligação a DNA/fisiologia , Produtos Finais de Glicação Avançada/farmacologia , Proteínas do Leite , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas , Transativadores/fisiologia , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/fisiologia , Linhagem Celular , Ciclina D1/metabolismo , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/efeitos dos fármacos , Quinases Ciclina-Dependentes/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Janus Quinase 2 , Oligonucleotídeos/genética , Oligonucleotídeos/farmacologia , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Fator de Transcrição STAT5 , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transativadores/genética , Transativadores/metabolismo , Tirosina/metabolismo
14.
J Cell Physiol ; 188(2): 188-93, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11424085

RESUMO

Our previous work has shown that a number of sphingolipid metabolites including sphingosine, sphinganine, and other long-chain bases potently induced apoptosis in human hepatoma cells. In this study, we examined the possibility that sphingosine may trigger apoptosis in human hepatoma cells via inhibition of anti-apoptotic pathways. We investigated the effect of sphingosine on AKT kinase, a serine/threonine kinase which was found to protect cells from apoptosis induced by a variety of extracellular stresses. Our results indicated that sphingosine inhibited basal and serum-stimulated AKT kinase activity in a dose-dependent manner in hepatoma cells. Additionally, sphingosine-induced inhibition of AKT kinase was correlated with induction of apoptosis in these cells. Pretreatment of insulin, a potent stimulator of AKT kinase, partially reversed the inhibition of AKT kinase by sphingosine and counteracted the apoptotic action of this sphingolipid. Expression of activated AKT kinase partially protected cells from sphingosine-induced apoptosis, whereas expression of kinase-dead AKT kinase had no effect. The molecular mechanism by which AKT kinase suppressed the apoptotic action of sphingosine was investigated. Our results showed that increased release of cytochrome C from mitochondria and subsequent activation of caspase-3 were detected in sphingosine-treated hepatoma cells. On the contrary, expression of activated AKT kinase in Hep3B cells attenuated cytochrome C release and caspase-3 activation induced by sphingosine. Taken together, these findings suggest that suppression of AKT kinase is one of the mechanisms by which sphingosine induces apoptosis in hepatoma cells and activation of AKT kinase may inhibit sphingosine-induced apoptosis by blocking a step upstream of cytochrome C release and caspase-3 activation.


Assuntos
Apoptose/fisiologia , Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Esfingosina/toxicidade , Apoptose/efeitos dos fármacos , Proteínas Sanguíneas/farmacologia , Grupo dos Citocromos c/metabolismo , Ativação Enzimática/fisiologia , Humanos , Ácido Mirístico/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Células Tumorais Cultivadas
15.
J Cell Biochem ; 81(1): 102-13, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11180401

RESUMO

Advanced glycation end-product (AGE) is important in the pathogenesis of diabetic nephropathy (DN), and captopril (an angiotensin converting enzyme inhibitor) is effective in treating this disorder. We have shown that the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) cascade is responsible for AGE-induced mitogenesis in NRK-49F (normal rat kidney fibroblast) cells, but its role in renal fibrosis in DN remains unknown. Therefore, we have sought to determine whether JAK/STAT is involved in AGE-regulated collagen production in NRK-49F cells. We found that AGE time (1-7 days) and dose (10-200 microg/ml)-dependently increased collagen production in these cells. Additionally, AGE increased RAGE (receptor for AGE) protein expression. AGE-induced RAGE expression was dose-dependently inhibited by antisense RAGE oligodeoxynucleotide (ODN) and captopril. AGE-induced type I collagen production and JAK2-STAT1/STAT3 activation were decreased by AG-490 (a specific JAK2 inhibitor), antisense RAGE ODN and captopril. Meanwhile, STAT1 and STAT3 decoy ODNs also suppressed the induction of collagen by AGE. We concluded that RAGE and the JAK2-STAT1/STAT3 pathway were involved in AGE-induced collagen production in NRK-49F cells. Furthermore, captopril was found to reverse AGE-induced collagen production, probably by attenuating RAGE expression and JAK2-STAT1/STAT3 activities.


Assuntos
Colágeno/biossíntese , Proteínas de Ligação a DNA/metabolismo , Produtos Finais de Glicação Avançada , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Receptores Imunológicos/metabolismo , Transdução de Sinais , Transativadores/metabolismo , Sequência de Bases , Captopril/farmacologia , Linhagem Celular , Primers do DNA , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Janus Quinase 2 , Cinética , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Tirosina/metabolismo , Tirfostinas/farmacologia
16.
Mol Pharmacol ; 58(6): 1398-403, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11093779

RESUMO

Increased expression of cyclooxygenase-2 (COX-2) causes enhanced production of prostaglandins, which are emerging as important mediators of growth stimulation of cancer cells. Overexpression of COX-2 has been found in human non-small cell lung cancer tissues and cell lines. In vitro and in vivo studies showed that nonselective cyclooxygenase inhibitors (like aspirin and indomethacin) may suppress growth of lung cancer cells and may prevent lung tumorigenesis induced by the tobacco-specific carcinogens. However, the molecular mechanisms that mediated the anticancer action of these inhibitors are not well defined. In this study, we examined the effect of a specific COX-2 inhibitor, N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide (NS398), on high COX-2-expressing A549 lung cancer cells. Our results indicated that NS398 inhibited prostaglandin E(2) synthesis and induced G(1) growth arrest in these cells. NS398 specifically up-regulated cyclin-dependent kinase inhibitor p27(KIP1), whereas the expressions of G(1)-acting cyclins and cyclin-dependent kinases were not changed. Additionally, NS398 effectively suppressed cyclin E-associated kinase activity in A549 cells. The molecular mechanism responsible for the induction of p27(KIP1) by NS398 was characterized. We found that NS398 did not induce p27(KIP1) through transcriptional activation because this drug could not stimulate the p27(KIP1) promoter. Metabolic labeling experiments showed that the synthesis rate of p27(KIP1) protein was not altered by NS398. Conversely, pulse-chase assays demonstrated that degradation of p27(KIP1) protein was obviously reduced in NS398-treated cells. We conclude that NS398 enhances p27(KIP1) expression via post-translational regulation, and our results provide a new mechanism by which specific COX-2 inhibitors suppress proliferation of cancer cells.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular , Fase G1/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/biossíntese , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Proteínas Supressoras de Tumor , Divisão Celular/efeitos dos fármacos , Ciclina G , Ciclina G1 , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/biossíntese , Neoplasias Pulmonares , Proteínas de Membrana , Proteínas Associadas aos Microtúbulos/genética , Prostaglandina-Endoperóxido Sintases/biossíntese , Biossíntese de Proteínas/efeitos dos fármacos , Células Tumorais Cultivadas , Regulação para Cima
17.
Int J Oncol ; 17(4): 659-65, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10995875

RESUMO

Our previous results demonstrated that the plant amino acid mimosine blocked cell cycle progression and suppressed proliferation of human lung cancer cells in vitro by multiple mechanisms. Inhibition of cyclin D1 expression or induction of cyclin-dependent kinase inhibitor p21WAF1 expression was found in mimosine-treated lung cancer cells. However, whether mimosine may modulate the expression of these cell cycle regulatory proteins and suppress tumor growth in vivo is unknown. In this study, we examined the anti-cancer effect of mimosine on human H226 lung cancer cells grown in nude mice. Our results demonstrated that mimosine inhibits cyclin D1 and induces p21WAF1 expression in vivo. Furthermore, results of TUNEL analysis indicated that mimosine may induce apoptosis to suppress tumor growth in nude mice. Collectively, these results suggest that mimosine exerts anti-cancer effect in vivo and might be useful in the therapy of lung cancer.


Assuntos
Proteínas de Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Mimosina/farmacologia , Neoplasias Experimentais/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Proteínas de Ciclo Celular/biossíntese , Ciclina D1/biossíntese , Ciclina D1/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/biossíntese , Ciclinas/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Fatores de Tempo , Transplante Heterólogo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Br J Haematol ; 110(1): 110-5, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10930986

RESUMO

In this study, platelet thrombi formation was induced by irradiation of mesenteric venules with filtered light in mice pretreated intravenously with fluorescein sodium. Rutaecarpine (200 microg/g) significantly prolonged the latent period of inducing platelet plug formation in mesenteric venules when it was intravenously injected. Rutaecarpine (200 microg/g) prolonged occlusion time by approximately 1.5-fold (control 127 +/- 29 vs. taecarpine 188 +/- 23 s). Furthermore, aspirin (250 microg/g) also showed a similar prolongation of the occlusion time in this experiment. On a molar basis, rutaecarpine was approximately twofold more potent than aspirin at prolonging the occlusion time. Furthermore, rutaecarpine was also effective in reducing the mortality of ADP-induced acute pulmonary thromboembolism in mice when administered intravenously at doses of 25 and 50 microg/g. Intravenous injection of rutaecarpine (50 microg/g) significantly prolonged the bleeding time by approximately 1.5-fold compared with normal saline in the severed mesenteric arteries of rats. Continuous infusion of rutaecarpine (5 microg/g/min) also significantly increased the bleeding time 1. 5-fold, and the bleeding time returned to baseline within 60 min after cessation of rutaecarpine infusion. These results suggest that rutaecarpine has an effective anti-platelet effect in vivo and that it may be a potential therapeutic agent for arterial thrombosis, but it must be assessed further for toxicity.


Assuntos
Alcaloides/farmacologia , Medicamentos de Ervas Chinesas , Agregação Plaquetária/efeitos dos fármacos , Difosfato de Adenosina , Animais , Aspirina/farmacologia , Tempo de Sangramento , Pressão Sanguínea/efeitos dos fármacos , Fluoresceína , Alcaloides Indólicos , Camundongos , Camundongos Endogâmicos ICR , Inibidores da Agregação Plaquetária/farmacologia , Embolia Pulmonar/tratamento farmacológico , Quinazolinas , Ratos , Ratos Sprague-Dawley
19.
Oncogene ; 19(33): 3766-73, 2000 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-10949931

RESUMO

Although originally synthesized as an anti-estrogen, tamoxifen (Tam) was found to be able to inhibit proliferation of estrogen receptor (ER)-negative cancer cells in vitro. However, the molecular basis of such ER-independent growth inhibition is largely unknown. We have previously demonstrated that Tam induces p21WAF1 and p27KIP1 expression in human lung cancer cells which lack ER-alpha and -beta. We found that Tam induced p21WAF1 expression via transcriptional activation. In order to determine the molecular mechanism responsible for p21WAF1 induction by Tam, we performed a deletion analysis on the p21WAF1 promoter. The minimal region in the p21WAF1 promoter required for Tam-activated induction was mapped to a contiguous stretch of 10 bp located 83 bases upstream of the transcription initiation site. Our results showed that transcription factor Sp1 and Sp3 bound to this GC-rich region and mutation of Sp1-binding sites dramatically attenuated Tam-induced p21WAF1 promoter activity. We also tried to elucidate the signaling pathway that mediated the activation of p21WAF1 by Tam. Inhibition of mitogen-activated protein kinase pathways did not block Tam-induced p21WAF1. Similarly, protein kinase C inhibitor calphostin C could not suppress Tam-induced p21WAF1. Conversely, pretreatment of a specific protein kinase A inhibitor H89 significantly attenuated the induction of p21WAF1 by Tam. Furthermore, PKA activators forskolin and dibutyryl-cAMP activated p21WAFI promoter activity and increased p21wAF1 protein level in lung cancer cells. Taken together, these results demonstrate that Tam activates the p21WAF1 promoter via Sp1-binding sites and suggest that PKA may be involved in the induction of p21wAF1 by Tam in ER-negative lung cancer cells.


Assuntos
Antineoplásicos Hormonais/farmacologia , Ciclinas/genética , Antagonistas de Estrogênios/farmacologia , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Fator de Transcrição Sp1/metabolismo , Tamoxifeno/farmacologia , Antineoplásicos Hormonais/metabolismo , Sítios de Ligação , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Luciferases/genética , Luciferases/metabolismo , Neoplasias Pulmonares , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Mutagênese , Proteínas Nucleares/metabolismo , Proteína Quinase C/antagonistas & inibidores , Elementos de Resposta , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3 , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
20.
Cancer Lett ; 145(1-2): 1-8, 1999 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-10530763

RESUMO

The plant amino acid mimosine has been reported to block cell cycle progression in the late G1 phase. A recent study showed that mimosine might induce growth arrest by activating the expression of p21CIP1, a cyclin-dependent kinase inhibitor (CDKI), and by inhibiting the activity of cyclin E-associated kinases in human breast cancer cells. However, mimosine at higher concentrations also blocked proliferation of p21-/- cells by unknown mechanisms. In this study, we investigated the effect of mimosine on the expression of cyclins and CDKIs in human lung cancer cells. We found that mimosine specifically inhibited cyclin D1 expression in H226 cells. The expression of another G1 cyclin, cyclin E, was not regulated by mimosine in all lung cancer cell lines examined. Moreover, mimosine induced p21CIP1 expression in H226 and H358 cells, while it activated p27KIP1 expression in H322 cells. However, mimosine does not affect transcription of these genes directly because significant changes in cyclin D1 or CDKI expression were observed at 12-24 h after drug addition. Our results indicate that mimosine may block cell proliferation by multiple mechanisms and this amino acid is a useful agent for the study of cell cycle control.


Assuntos
Ciclinas/biossíntese , Inibidores Enzimáticos/metabolismo , Neoplasias Pulmonares/tratamento farmacológico , Mimosina/uso terapêutico , Proteínas de Neoplasias/biossíntese , Divisão Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21 , Fase G1/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/patologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA