Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 586
Filtrar
1.
Curr Radiopharm ; 5(1): 47-58, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21864242

RESUMO

PURPOSE: Chloroquine has demonstrated high affinity for aldehyde dehydrogenase 1A1 (ALDH1), an enzyme expressed in the highly tumorigenic CD133+ brain tumor initiating subpopulation. The purpose of this study is to report the novel synthesis of a chloroquine analogue, n.c.a. iodoquine, and the in vitro and in vivo uptake in cells with high ALDH1 content. METHODS AND MATERIALS: Iodoquine was synthesized in novel no-carrier-added forms (n.c.a.) for both 125I and 123I. I25I IQ and 18F FDG cell uptake assays were performed in the L1210 and L1210cpa (cyclophosphamide resistant), A549, and MG456 glioblastoma cell lines. Uptake was expressed as a percent of the administered activity. 125I IQ biodistribution studies assessed organ uptake at 1, 4, and 24 hours after IV administration (n= 15 total; 5 mice/timepoint). Radiation dosimetry estimates were calculated using standard OLINDA/EXM software. In vivo imaging of 123I IQ uptake in MG456 glioblastoma mouse model (n=10) was performed with small animal high resolution micro-SPECT. Autoradiography and histology co-localized radiotracer and tumor biodistribution. Uptake in MG456 glioblastoma tumors was quantified with gamma counting. RESULTS: L1210 cpa (high ALDH1) showed significantly higher 125I IQ uptake compared to the parental L1210 (low ALDH1) for all time points through 4 hours (20.7% ± 1.4% versus 11.0% ± 0.5%; 21.3% ± 0.9% versus 11.0% ± 0.4%; 20.6% ± 0.7% versus 9.4% ± 0.3%; and 15.7% ± 0.7% versus 7.5% + 0.4% at 30 minutes, and 1, 2 and 4 hours, respectively; p < 0.001 for all time points). In the CD133+ fraction of MG456 glioblastoma cell line, IQ uptake was significantly higher compared to FDG at all time points through 4 hours (81.5% ± 0.9% versus 1.3% ± 0.1%; 88.8% ± 0.4% versus 1.3% ± 0.1%; 87.8% ± 2.1% versus 1.7% ± 0.2%; and 87.0% ± 2.4% versus 1.8% ± 0.1 at 30 minutes, and 1, 2 and 4 hours, respectively; p > 0.001 for all time points). The A549 lung cancer cell line also showed high IQ uptake through 4 hours. IQ normal biodistribution studies showed rapid renal excretion and very low normal background brain activity after IV administration. In vivo micro-SPECT images showed mild uptake in larger MG456 glioblastomas (n=6) as verified with autoradiography and histology. Gamma well counter uptake in large tumors was 2.3% ± 0.48% ID/g (n=5). CONCLUSION: Iodoquine localizes to cells with high ALDH1 content. Cell assays show high 125I IQ uptake in the MG456 cell line, and in vivo micro-SPECT imaging showed mild 123I IQ uptake in MG456 glioblastomas. Further studies are necessary to investigate 131I IQ as a potential therapeutic agent targeting the highly tumorigenic CD133+ brain tumor stem cell subpopulation.


Assuntos
Cloroquina/análogos & derivados , Radioisótopos do Iodo/farmacocinética , Isoenzimas/metabolismo , Neoplasias/diagnóstico por imagem , Compostos Radiofarmacêuticos/farmacocinética , Retinal Desidrogenase/metabolismo , Família Aldeído Desidrogenase 1 , Animais , Western Blotting , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/enzimologia , Linhagem Celular Tumoral , Cloroquina/farmacocinética , Resistencia a Medicamentos Antineoplásicos , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/enzimologia , Leucemia L1210/enzimologia , Masculino , Camundongos , Camundongos Nus , Neoplasias/enzimologia , Doses de Radiação , Compostos Radiofarmacêuticos/síntese química , Tomografia Computadorizada de Emissão de Fóton Único/métodos
2.
J Cell Biochem ; 113(4): 1416-25, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22173742

RESUMO

The acquisition of a multidrug-resistant (MDR) phenotype by tumor cells that renders them unsusceptible to anti-neoplasic agents is one of the main causes of chemotherapy failure in human malignancies. The increased expression of P-glycoprotein (MDR1, P-gp, ABCB1) in tumor cells contributes to drug resistance by extruding chemotherapeutic agents or by regulating programmed cell death. In a study of MDR cell survival under cold stress conditions, it was found that resistant leukemic cells with P-gp over-expression, but not their sensitive counterparts, are hypersensitive to cold-induced cell death when exposed to temperatures below 4 °C. The transfection of parental cells with a P-gp-expressing plasmid makes these cells sensitive to cold stress, demonstrating an association between P-gp expression and cell death at low temperatures. Furthermore, we observed increased basal expression and activity of effector caspase-3 at physiological temperature (37 °C) in MDR cells compared with their parental cell line. Treatment with a caspase-3 inhibitor partially rescues MDR leukemic cells from cold-induced apoptosis, which suggests that the cell death mechanism may require caspase-3 activity. Taken together, these findings demonstrate that P-gp expression plays a role in MDR cell survival, and is accompanied by a collateral sensitivity to death induced by cold stress. These findings may assist in the design of specific therapeutic strategies to complement current chemotherapy treatment against cancer.


Assuntos
Caspase 3/metabolismo , Temperatura Baixa , Resistência a Múltiplos Medicamentos , Leucemia L1210/patologia , Estresse Fisiológico , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Western Blotting , Morte Celular , Linhagem Celular Tumoral , Leucemia L1210/enzimologia , Leucemia L1210/metabolismo , Fenótipo , Fosfatidilserinas/metabolismo
3.
Invest New Drugs ; 29(5): 730-41, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20349265

RESUMO

Quaternary benzo[c]phenanthridines such as fagaronine are natural substances which have been reported to exhibit anticancer and anti-leukemic properties. However, the therapeutic use of these molecules is limited due to the high dose required to exhibit anti-tumor activity and subsequent toxicity. In this study, we describe the therapeutic potential of a new derivative of fagaronine, Ethoxyfagaronine (N-methyl-12-ethoxy-2hydroxy-3, 8, 9-trimethoxybenzo[c]-phenanthridiniumchlorhydrate) as an anti-leukemic agent. Cytotoxic activity and cell growth inhibition of Ethoxyfagaronine (Etxfag) was tested on murine L1210 leukemia cells using trypan blue assay and MTT assay. At the concentration of 10(-7) M, Etxfag induced less than 10% of cell death. Etxfag (10(-7) M) was tested on L1210 cell invasiveness using matrigel™ precoated transwell chambers and efficiently reduces the invasive potential of L1210 cells by more than 50% as compared with untreated cells. Western blot and immunofluorescence experiments showed that Etxfag decreased both MT1-MMP expression and activation at the cell surface, decreased plasmin activity by down-regulating u-PAR and uPA expression at the cell surface and increasing PAI-1 secretion in conditioned media. The set of our findings underscore the therapeutic potential of ethoxyfagaronine as a new potential anticancer agent able to prevent leukemic cell dissemination.


Assuntos
Alcaloides/farmacologia , Benzofenantridinas/farmacologia , Regulação para Baixo/efeitos dos fármacos , Leucemia L1210/enzimologia , Leucemia L1210/patologia , Metaloproteinase 14 da Matriz/metabolismo , Ativadores de Plasminogênio/metabolismo , Animais , Aprotinina/farmacologia , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dipeptídeos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Fibrinolisina/metabolismo , Gelatinases/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Leucemia L1210/genética , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Invasividade Neoplásica
4.
Leuk Res ; 35(1): 110-8, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20510451

RESUMO

New approaches should be sought to treat high-risk acute lymphoblastic leukemia (ALL). Since aberrant DNA methylation plays an important role in leukemogenesis of ALL, it can be targeted by 5-aza-2'-deoxycytidine (5-AZA-CdR), a potent inhibitor of DNA methylation. 5-AZA-CdR is a prodrug that is activated by deoxycytidine kinase (DCK). Leukemic cells lacking DCK are drug-resistant. In a previous phase I study, we reported that 5-AZA-CdR could induce remissions in ALL. However, some patients developed drug-resistance due to deficiency in DCK. These observations aroused our interest in 3-deazauridine (3-DU), a CTP synthetase inhibitor that is effective against leukemic cells deficient in DCK. In this report, we observed that 3-DU enhanced the in vitro antineoplastic action of 5-AZA-CdR on human leukemic cells by increasing its incorporation into DNA. Using an optimized dose-schedule we showed that this combination could cure some mice bearing L1210 leukemia, even in the presence of a subpopulation of drug-resistant (L1210/ARA-C) leukemic cells lacking DCK. 3-DU alone also cured some mice with L1210/ARA-C leukemia. In a pilot study on 3 relapsed patients with advanced ALL, the combination of 5-AZA-CdR and 3-DU produced a marked reduction in leukemic blasts, confirming our preclinical observations. Furthermore, after several treatments with these agents all three patients developed drug-resistance to 5-AZA-CdR as determined by an in vitro drug sensitivity test. In two patients we showed by enzymatic analysis that the drug-resistance was due to deficiency in DCK. Our preclinical and clinical results provide a strong rationale to further investigate the combination of 5-AZA-CdR and 3-DU for the treatment of advanced ALL.


Assuntos
3-Desazauridina/farmacologia , Azacitidina/análogos & derivados , Desoxicitidina Quinase/metabolismo , Resistencia a Medicamentos Antineoplásicos , Leucemia L1210/patologia , Animais , Azacitidina/farmacologia , Linhagem Celular Tumoral , Decitabina , Sinergismo Farmacológico , Humanos , Leucemia L1210/enzimologia , Masculino , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Biomed Khim ; 55(5): 621-34, 2009.
Artigo em Russo | MEDLINE | ID: mdl-20017393

RESUMO

Lysosomal proteases are actively involved in pathogenesis of cancer progression. Alterations in proteases and their inhibitors interaction were suggested to be implicated in the processes of tumor invasion and metastasis. Among proteases connected with malignant growth, cysteine cathepsins B and L and aspartic cathepsin D play the main role in the tumor development. The present study was designed to investigate activity of cathepsins B, L and D activity in the development and treatment of murine experimental leukemias and to determine the correlation of these proteases with tumor malignancy and the chemotherapy effect. P-388 leukemia was characterized by a more aggressive development and unfavorable prognosis than L1210/1 leukemia. The activity of cathepsins B, L and D in tumor tissues of mice infected with P-388 leukemia, as well as in liver and spleen and the activity of cathepsins B and L in serum were lower than their activity in mice infected with L1210/1 leukemia. Changes of cathepsin activity in liver and spleen of mice with leukemias have demonstrated a level of aggressiveness of tumor development and invasion of liver and spleen by neoplastic cells. The treatment resulted in the increase of cathepsin B, L and D activities in tumor tissue, liver, spleen and cathepsin B and L activities in serum. The highest activity of proteases was revealed in the groups of mice characterized by the greatest suppression of tumor growth. These data have shown that lysosomal proteases are involved in progression of murine experimental leukemias and elimination of tumor cells in the result of treatment. Determination of the activity of cysteine and aspartic proteases can be used for evaluation of cancer diseases malignancy, their sensitivity for chemotherapy and efficiency of treatment.


Assuntos
Catepsina B/metabolismo , Catepsina D/metabolismo , Catepsina L/metabolismo , Leucemia L1210/enzimologia , Leucemia P388/enzimologia , Lisossomos/enzimologia , Animais , Leucemia L1210/tratamento farmacológico , Leucemia L1210/patologia , Leucemia P388/tratamento farmacológico , Leucemia P388/patologia , Lisossomos/patologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Invasividade Neoplásica , Metástase Neoplásica
6.
Nucl Med Biol ; 36(8): 919-29, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19875048

RESUMO

INTRODUCTION: To advance the science and clinical application of stem cell therapy, the availability of a highly sensitive, quantitative and translational method for tracking stem cells would be invaluable. Because hematopoetic stem cells express high levels of the cytosolic enzyme aldehyde dehydrogenase-1A1 (ALDH1), we sought to develop an agent that is specific to ALDH1 and thus to cells expressing the enzyme. Such an agent might be also helpful in identifying tumors that are resistant to cyclophosphomide chemotherapy because ALDH1 is known to be responsible for this resistance. METHODS: We developed schemes for the synthesis of two radioiodinated aldehdyes - N-formylmethyl-5-[*I]iodopyridine-3-carboxamide ([*I]FMIC) and 4-diethylamino-3-[*I]iodobenzaldehyde ([*I]DEIBA)-at no-carrier-added levels from their respective tin precursors. These agents were evaluated using pure ALDH1 and tumor cells that expressed the enzyme. RESULTS: The average radiochemical yields for the synthesis of [(125)I]FMIC and [(125)I]DEIBA were 70+/-5% and 47+/-14%, respectively. ALDH1 converted both compounds to respective acids suggesting their suitability as ALDH1 imaging agents. Although ability of ALDH1 within the cells to oxidize one of these substrates was shown, specific uptake in ALDH-expressing tumor cells could not be demonstrated. CONCLUSION: To pursue this approach for ALDH1 imaging, radiolabeled aldehydes need to be designed such that, in addition to being good substrates for ALDH1, the cognate products should be sufficiently polar so as to be retained within the cells.


Assuntos
Aldeído Desidrogenase/metabolismo , Aldeídos/farmacocinética , Leucemia L1210/diagnóstico por imagem , Leucemia L1210/enzimologia , Aldeídos/química , Animais , Sistemas de Liberação de Medicamentos/métodos , Humanos , Radioisótopos do Iodo/química , Radioisótopos do Iodo/farmacocinética , Células K562 , Cintilografia
7.
Acta Pharmacol Sin ; 30(10): 1443-53, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19801998

RESUMO

AIM: To search for novel inhibitors of human polo-like kinase 1 (Plk1), which plays important roles in various aspects of mitotic progression and is believed as a promising anti-cancer drug target, and further investigate the potential inhibition mechanism of active compounds against Plk1, thus developing potent anti-tumor lead compounds. METHODS: Surface plasmon resonance (SPR) technology-based assay and enzymatic inhibition assay were used to screen Plk1 inhibitors. Sulphorhodamine B (SRB)-based assay, flow cytometry, confocal microscopy and Western blotting were used to further identify the potent Plk1 inhibitor. To investigate the inhibitory mechanism of the active compound against Plk1, enzymatic inhibition assay, SPR and yeast two-hybrid technology-based assays were used. RESULTS: Aristolactam AIIIa was identified as a new type of Plk1 inhibitors, targeting the Polo Box domain (PBD) which is another efficient tactic for exploring Plk1 inhibitors. Further studies indicated that it could block the proliferations of HeLa, A549, HGC and the HCT-8/V cells (clinical Navelbine-resistant cancer cell), induce mitotic arrest of HeLa cells at G2/M phase with spindle abnormalities and promote apoptosis in HeLa cells. The results from SPR and yeast two-hybrid technology-based assays suggested that it could target both the catalytic domain of Plk1 (CD) and PBD and enhance the CD/PBD interaction. CONCLUSION: Our current work is expected to shed light on the potential anti-tumor mechanism of Aristolactam AIIIa, and this natural product might be possibly used as a lead compound for further developing anti-tumor drugs.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Neoplasias/genética , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/enzimologia , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Domínio Catalítico/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Relação Dose-Resposta a Droga , Células HT29 , Células HeLa , Humanos , Alcaloides Indólicos , Concentração Inibidora 50 , Lactamas , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Ligantes , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/enzimologia , Camundongos , Estrutura Molecular , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Fuso Acromático/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/enzimologia , Ressonância de Plasmônio de Superfície , Técnicas do Sistema de Duplo-Híbrido , Quinase 1 Polo-Like
8.
J Immunol ; 182(6): 3398-405, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265117

RESUMO

Tumor cell-based vaccines are currently used in clinical trails, but they are in general poorly immunogenic because they are composed of cell extracts or apoptotic cells. Live tumor cells should be much better Ags provided that they are properly processed by the host immune system. We show herein that stable expression of a small hairpin RNA for ERK5 (shERK5) decreases ERK5 levels in human and mouse leukemic cells and leads to their elimination by NK cells in vivo. The shERK5 cells show down-regulation of MHC class I expression at the plasma membrane. Accordingly, ectopic activation of the ERK5 pathway induces MHC class I gene expression. Coinjection of shERK5-expressing cells into the peritoneum diminishes survival of engrafted wild-type tumor cells. Moreover, s.c. injection of shERK5-expressing cells strongly diminishes tumor development by wild-type cells. Our results show that shERK5 expression in leukemia cells effectively attenuates their tumor activity and allows their use as a tumor cell-based vaccine.


Assuntos
Vacinas Anticâncer/imunologia , Técnicas de Silenciamento de Genes , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/imunologia , Leucemia L1210/prevenção & controle , Ativação Linfocitária/imunologia , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/genética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Células Cultivadas , Citotoxicidade Imunológica/genética , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Células Jurkat , Células Matadoras Naturais/metabolismo , Leucemia L1210/enzimologia , Leucemia L1210/genética , Leucemia L1210/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , RNA Interferente Pequeno/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
9.
J Vet Med Sci ; 70(11): 1185-90, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19057136

RESUMO

Humic substances are formed during the decomposition of organic matter in humus, and are found in many natural environments in which organic materials and microorganisms are present. Oral administration of humus extract to mice successfully induced effective protection against experimental challenge by the two subspecies, Trypanosoma brucei brucei and T. brucei gambiense. Mortality was most reduced among mice who received a 3% humus extract for 21 days in drinking water ad libitum. Spleen cells from humus-administered mice exhibited significant non-specific cytotoxic activity against L1210 mouse leukemia target cells. Also, spleen cells produced significantly higher amounts of Interferon-gamma when stimulated in vitro with Concanavalin A than cells from normal controls. These results clearly show that administration to mice of humus extract induced effective resistance against Trypanosoma infection. Enhancement of the innate immune system may be involved in host defense against trypanosomiasis.


Assuntos
Solo , Tripanossomicidas/uso terapêutico , Tripanossomíase Africana/prevenção & controle , Animais , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citocinas/biossíntese , Feminino , L-Lactato Desidrogenase/efeitos dos fármacos , L-Lactato Desidrogenase/metabolismo , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Baço/efeitos dos fármacos , Baço/patologia , Baço/fisiopatologia , Trypanosoma brucei brucei/efeitos dos fármacos , Trypanosoma brucei gambiense/efeitos dos fármacos
10.
Cell Biochem Funct ; 26(5): 609-19, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18508389

RESUMO

Quinolines are known to be multitarget agents with a broad spectrum of biological activity. In a previous study, we showed that newly prepared 4-amino-3-acetylquinoline (AAQ) possesses strong anticancer activities. In this study, we investigated whether AAQ has cytotoxicity in murine L1210 leukemia cells. Results from cell proliferation assays showed that AAQ caused significant decrease in cell number in a dose-dependent manner. The cell death induced by AAQ appeared to involve apoptosis, based on evidence from apoptotic DNA fragmentation, flow cytometry, fluorescence microscopy, and Western blot analyses. We found that AAQ-treated cells had activated p38 MAPK and that apoptosis was processed through a reactive oxygen species (ROS)-dependent mitochondrial pathway. In summary, our results suggest that AAQ can induce apoptosis, at least in part, through the activation of the p38 MAPK pathway in L1210 leukemia cells.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores do Crescimento/farmacologia , Leucemia L1210/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Quinolinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Ativação Enzimática , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Camundongos , Mitocôndrias/enzimologia
11.
Pflugers Arch ; 457(2): 271-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18458940

RESUMO

Nitrosoglutathione [(GSNO), 500 nmol/l] relaxed the norepinephrine precontracted rat aortic rings. The relaxation effect was pronouncedly enhanced by H(2)S- and HS(-)-donor NaHS (30 micromol/l) at 7.5 pH but not at 6.3 pH. To study molecular mechanism of this effect, we investigated whether NaHS can release NO from NO donors. Using an electron paramagnetic resonance spectroscopy method of spin trap and by measuring the NO oxidation product, which is nitrite, by the Griess reaction, we report that NaHS released NO from nitrosothiols, namely from GSNO, S-nitroso-N-acetyl-DL: -penicillamine (SNAP), from metal nitrosyl complex nitroprusside (SNP) and from rat brain homogenate and murine L1210 leukaemia cells. From the observation that the releasing effect was more pronounced at 8.0 pH than 6.0 pH, we suppose that HS(-), rather than H(2)S, is responsible for the NO-releasing effect. Since in mammals, H(2)S and HS(-) are produced endogenously, we assume that their effect to release NO from nitrosothiols and from metal nitrosyl complexes are responsible for some of their biological activities and that this mechanism may be involved in S-nitrosothiol-signalling reactions.


Assuntos
Encéfalo/efeitos dos fármacos , Sulfeto de Hidrogênio/metabolismo , Leucemia L1210/metabolismo , Doadores de Óxido Nítrico , Óxido Nítrico/metabolismo , Nitroprussiato/metabolismo , S-Nitrosotióis/metabolismo , Sulfetos/farmacologia , Vasodilatadores , Animais , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Encéfalo/enzimologia , Encéfalo/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância de Spin Eletrônica , Guanilato Ciclase/metabolismo , Concentração de Íons de Hidrogênio , Leucemia L1210/enzimologia , Masculino , Camundongos , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Nitritos/metabolismo , Ratos , Ratos Wistar , Receptores Citoplasmáticos e Nucleares/metabolismo , S-Nitroso-N-Acetilpenicilamina/metabolismo , S-Nitrosoglutationa/metabolismo , Transdução de Sinais , Guanilil Ciclase Solúvel , Fatores de Tempo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/metabolismo , Vasodilatadores/farmacologia
12.
Cancer Lett ; 259(1): 111-8, 2008 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-18006147

RESUMO

Curcumin, a constituent of turmeric, has anti-inflammatory, anti-carcinogenic, and chemopreventive effects in several animal tumor models. The expression of P-glycoprotein (P-gp), encoded by the mdr gene, is often associated with multidrug resistance (MDR) to unrelated chemotherapeutic drugs in cancer cells. Here, we demonstrate that curcumin down-regulates P-gp expression in multidrug-resistant L1210/Adr cells. Transfection with a series of 5'-deleted constructs of the mdr1b gene promoter indicated that a proximal region between -205 and +42 of the sequence was responsible for the suppression of promoter activity by curcumin. This response might be associated with the inhibition of the phosphatidyinositol 3-kinase (PI3K)/Akt/nuclear factor-kappa B (NF-kappa B) signaling pathway by curcumin. Moreover, curcumin reversed the MDR of the L1210/Adr cells. Thus, curcumin can contribute to the reversal of the MDR phenotype, probably due to the suppression of P-gp expression via the inhibition of the PI3K/Akt/NF-kappa B signaling pathway.


Assuntos
Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Curcumina/farmacologia , Leucemia L1210/tratamento farmacológico , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Animais , Antibióticos Antineoplásicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular Tumoral , Curcumina/uso terapêutico , Relação Dose-Resposta a Droga , Regulação para Baixo , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Leucemia L1210/enzimologia , Leucemia L1210/metabolismo , Camundongos , NF-kappa B/antagonistas & inibidores , Células NIH 3T3 , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Deleção de Sequência , Transdução de Sinais/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transfecção , Membro 4 da Subfamília B de Transportadores de Cassetes de Ligação de ATP
13.
Cancer Res ; 65(24): 11721-8, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16357184

RESUMO

BGC 945 is a cyclopenta[g]quinazoline-based, thymidylate synthase inhibitor specifically transported into alpha-folate receptor (alpha-FR)-overexpressing tumors. Affinity of BGC 945 for the alpha-FR is 70% of the high-affinity ligand folic acid. In contrast to conventional antifolates, BGC 945 has low affinity for the widely expressed reduced-folate carrier (RFC). The K(i) for isolated thymidylate synthase is 1.2 nmol/L and the IC(50) for inhibition of the growth of alpha-FR-negative mouse L1210 or human A431 cells is approximately 7 micromol/L. In contrast, BGC 945 is highly potent in a range of alpha-FR-overexpressing human tumor cell lines (IC(50) approximately 1-300 nmol/L). Pharmacokinetic variables measured following i.v. injection of 100 mg/kg BGC 945 to KB tumor-bearing mice showed rapid plasma clearance (0.021 L/h) and tissue distribution. The terminal half-lives in plasma, liver, kidney, spleen, and tumor were 2, 0.6, 5, 21, and 28 hours, respectively. Tumor BGC 945 concentration at 24 hours was approximately 1 nmol/g tissue, at least 10-fold higher than that in plasma or normal tissues. Inhibition of thymidylate synthase in tissues leads to increased incorporation of 5-[(125)I]-iodo-2'-deoxyuridine ([(125)I]dUrd) into DNA. Forty-eight hours after injection of 100 mg/kg 6RS-BGC 945 ([(125)I]dUrd injected at 24 hours), tumor was the only tissue with incorporation above control level (6-fold). The RFC-mediated thymidylate synthase inhibitor plevitrexed also increased uptake of [(125)I]dUrd in tumor (10-fold) but, in contrast, also caused increased incorporation in other normal tissues such as spleen and small bowel (4.5- and 4.6-fold, respectively). These data suggest that BGC 945 selectively inhibits thymidylate synthase in alpha-FR-overexpressing tumors and should cause minimal toxicity to humans at therapeutic doses.


Assuntos
Proteínas de Transporte/metabolismo , Inibidores Enzimáticos/farmacologia , Quinazolinas/farmacologia , Receptores de Superfície Celular/metabolismo , Timidilato Sintase/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Transporte Biológico , Proliferação de Células/efeitos dos fármacos , Coriocarcinoma/tratamento farmacológico , Coriocarcinoma/enzimologia , Inibidores Enzimáticos/farmacocinética , Feminino , Receptores de Folato com Âncoras de GPI , Ácido Fólico/metabolismo , Humanos , Idoxuridina/metabolismo , Radioisótopos do Iodo , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/enzimologia , Proteínas de Membrana Transportadoras , Camundongos , Camundongos Nus , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Quinazolinas/farmacocinética , Proteína Carregadora de Folato Reduzido , Distribuição Tecidual , Transplante Heterólogo , Células Tumorais Cultivadas
14.
Anticancer Res ; 25(1A): 101-6, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15816525

RESUMO

A mouse leukemia L1210 cell line (Y8), selected for resistance to deoxyadenosine, has a markedly altered phenotypic expression that includes loss of sensitivity to dATP as an allosteric inhibitor of ribonucleotide reductase, increased expression of c-myc, c-fos and WAF1/p21, but decreased expression of p53. In addition, the Y8 cells have a Very strong apoptotic response to a variety of agents under conditions in which the parental wild-type cells do not apoptose. In these studies, we show that flavopiridol (a cdk inhibitor) causes the Y8 cells to undergo apoptosis via a caspase-3 activation process. The apoptotic response to flavopiridol is markedly enhanced by LY294002. Data also show that the apoptotic response of the Y8 cells to roscovitine (a cdk inhibitor) is enhanced by UCN-01 (a PKC inhibitor). These data show that simultaneous blockage of specific pathways leads to increased apoptosis in the Y8 cells with essentially no effects on the parental wild-type L1210 cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Estaurosporina/análogos & derivados , Androstadienos/administração & dosagem , Androstadienos/farmacologia , Animais , Apoptose/fisiologia , Caspase 3 , Caspases/metabolismo , Cromonas/administração & dosagem , Cromonas/farmacologia , Resistência a Múltiplos Medicamentos , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Inibidores Enzimáticos/administração & dosagem , Flavonoides/administração & dosagem , Flavonoides/farmacologia , Leucemia L1210/patologia , Camundongos , Morfolinas/administração & dosagem , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Piperidinas/administração & dosagem , Piperidinas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Purinas/administração & dosagem , Purinas/farmacologia , Roscovitina , Estaurosporina/administração & dosagem , Estaurosporina/farmacologia , Wortmanina
15.
Anticancer Res ; 25(1A): 157-60, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15816533

RESUMO

We investigated the effects of 1-beta-D-arabinofuranosylcytosine (ara-C) on the growth of murine leukemic L1210 cells, which were cultured with high (2.0 x 10(3) ng/ml), middle (100 ng/ml) and low doses (5.0 ng/ml) of ara-C. In the analysis by flow cytometry, high dose ara-C arrested the cell cycle in the G0/G1-phase. Middle and low doses ara-C induced a block in the S-phase, that was not completely blocked by the low dose. Analysis of DNA fragmentation revealed that ara-C dose-dependently induced apoptosis, which was only slightly induced by the low dose. We measured activities of cellular thymidylate synthase (TS) and thymidine kinase (TK) after 24-h culture. Low and middle doses, but not high dose ara-C markedly enhanced TS activity to 2.9- in low and 5.3-fold in middle doses ara-C, and TK activity to 1.3- in low and 2.2-fold in middle doses, respectively, compared with those of the control. The cells accumulated in the S-phase by 48-h culture with low dose ara-C and markedly proliferated compared to that of the control in ara-C-free medium. These results indicate that non-high dose ara-C enhances DNA-synthesizing enzyme activities in L1210 cells, and withdrawal of the non-high dose ara-C results in paradoxical cell proliferation. Thus, daily intramuscular injections with an insufficient dose of ara-C may induce cells into S-phase, resulting in the proliferation of leukemic cells.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Citarabina/administração & dosagem , Leucemia L1210/tratamento farmacológico , Leucemia L1210/patologia , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Leucemia L1210/enzimologia , Camundongos , Fase S/efeitos dos fármacos , Timidina Quinase/metabolismo , Timidilato Sintase/metabolismo
16.
Cancer Res ; 65(6): 2059-64, 2005 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15781612

RESUMO

A naturally occurring gallated polyphenol isolated from green tea leaves, (-)-epigallocatechin gallate (EGCG), has been shown to be an inhibitor of dihydrofolate reductase (DHFR) activity in vitro at concentrations found in the serum and tissues of green tea drinkers (0.1-1.0 micromol/L). These data provide the first evidence that the prophylactic effect of green tea drinking on certain forms of cancer, suggested by epidemiologic studies, is due to the inhibition of DHFR by EGCG and could also explain why tea extracts have been traditionally used in "alternative medicine" as anticarcinogenic/antibiotic agents or in the treatment of conditions such as psoriasis. EGCG exhibited kinetics characteristic of a slow, tight-binding inhibitor of 7,8-dihydrofolate reduction with bovine liver DHFR (K(I) = 0.109 micromol/L), but of a classic, reversible, competitive inhibitor with chicken liver DHFR (K(I) = 10.3 micromol/L). Structural modeling showed that EGCG can bind to human DHFR at the same site and in a similar orientation to that observed for some structurally characterized DHFR inhibitor complexes. The responses of lymphoma cells to EGCG and known antifolates were similar, that is, a dose-dependent inhibition of cell growth (IC50 = 20 micromol/L for EGCG), G0-G1 phase arrest of the cell cycle, and induction of apoptosis. Folate depletion increased the sensitivity of these cell lines to antifolates and EGCG. These effects were attenuated by growing the cells in a medium containing hypoxanthine-thymidine, consistent with DHFR being the site of action for EGCG.


Assuntos
Catequina/análogos & derivados , Catequina/química , Catequina/farmacologia , Antagonistas do Ácido Fólico/farmacologia , Chá/química , Tetra-Hidrofolato Desidrogenase/química , Tetra-Hidrofolato Desidrogenase/metabolismo , Animais , Catequina/metabolismo , Bovinos , Galinhas , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Antagonistas do Ácido Fólico/química , Antagonistas do Ácido Fólico/metabolismo , Humanos , Cinética , Leucemia L1210/tratamento farmacológico , Leucemia L1210/enzimologia , Fígado/enzimologia , Camundongos , Modelos Moleculares , Quinazolinas/química , Quinazolinas/metabolismo
18.
Anticancer Drugs ; 16(3): 301-8, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15711182

RESUMO

Tumor suppressor genes that have been silenced by aberrant DNA methylation are potential targets for reactivation by novel chemotherapeutic agents. The potent inhibitor of DNA methylation and antileukemic agent, 5-aza-2'-deoxycytidine (5-AZA-CdR, Decitabine), can reactivate silent tumor suppressor genes. One hindrance to the curative potential of 5-AZA-CdR is its rapid in vivo inactivation by cytidine deaminase (CD). An approach to overcome this obstacle is to use 5-AZA-CdR in combination with zebularine (Zeb), a potent inhibitor of CD. Zeb also possesses independent antineoplastic activity due to its inhibition of DNA methylation. We tested the capacity of 5-AZA-CdR and Zeb alone and in combination to inhibit growth and colony formation of different leukemic cell lines. 5-AZA-CdR and Zeb in combination produced a greater inhibition of growth against murine L1210 lymphoid leukemic cells, and a greater reduction in colony formation by L1210 and human HL-60 myeloid leukemic cells, than either agent alone. The ability of these agents to reactivate the tumor suppressor gene, p57KIP2, was also tested using RT-PCR. The combination produced a synergistic reactivation of p57KIP2 in HL-60 leukemic cells. A methylation-specific PCR assay showed that this combination also induced a significantly greater demethylation level of the p57KIP2 promoter than either drug alone. The in vivo antineoplastic activity of the agents was evaluated in mice with L1210 leukemia. A greater increase in survival time of mice with L1210 leukemia was observed with the combination than with either agent alone using three different dose schedules. The enhanced activity observed with 5-AZA-CdR plus Zeb in both murine and human leukemic cells lines provides a rationale for the clinical investigation of these drugs in patients with advanced leukemia. The probable mechanism of this drug interaction involves inhibition of CD by Zeb and the complementary inhibition of DNA methylation by both agents.


Assuntos
Antimetabólitos Antineoplásicos/uso terapêutico , Azacitidina/análogos & derivados , Azacitidina/uso terapêutico , Citidina Desaminase/antagonistas & inibidores , Leucemia L1210/tratamento farmacológico , Nucleosídeos de Pirimidina/farmacologia , Animais , Linhagem Celular Tumoral , Citidina/análogos & derivados , Metilação de DNA/efeitos dos fármacos , Decitabina , Relação Dose-Resposta a Droga , Interações Medicamentosas , Humanos , Leucemia L1210/enzimologia , Leucemia L1210/metabolismo , Masculino , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ensaio Tumoral de Célula-Tronco
19.
Cancer Chemother Pharmacol ; 51(4): 284-90, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12721755

RESUMO

PURPOSE: Tamoxifen (TAM), a nonsteroidal anticancer agent, is used in the treatment of breast cancer. In the current study, we investigated whether TAM induces apoptosis in tumor cells by altering the expression of Fas and Fas ligand (FasL). METHODS: Several tumor cell lines were used to test the ability of TAM to induce apoptosis, which was studied using the TUNEL assay. The effect of TAM on the expression of Fas and FasL was analyzed using a flow cytometer. RESULTS: TAM was found to suppress the growth of an estrogen receptor-positive human mammary tumor cell line (T-47D) by inducing apoptosis. Interestingly, TAM also induced apoptosis in an estrogen receptor-negative murine T cell lymphoma cell line, EL-4. The ability of TAM to induce apoptosis in T-47D and EL-4 tumor cells correlated with the increased expression of FasL but not Fas on the tumor cells. Similar to TAM, a metalloproteinase (MP) inhibitor, which is known to increase the expression of membrane-bound FasL, was found to induce apoptosis in both T-47D and EL-4 tumor cells by increasing the expression of FasL but not Fas. Furthermore, both TAM and the MP inhibitor failed to induce apoptosis in L1210 tumor cell lines that failed to express FasL. CONCLUSIONS: The current study demonstrates that TAM can induce apoptosis in Fas(+) tumor cells by upregulating FasL.


Assuntos
Anticarcinógenos/farmacologia , Apoptose/efeitos dos fármacos , Glicoproteínas de Membrana/biossíntese , Neoplasias/metabolismo , Tamoxifeno/farmacologia , Receptor fas/biossíntese , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Dipeptídeos/farmacologia , Proteína Ligante Fas , Humanos , Leucemia L1210/enzimologia , Leucemia L1210/metabolismo , Leucemia L1210/patologia , Linfoma/enzimologia , Linfoma/metabolismo , Linfoma/patologia , Metaloendopeptidases/antagonistas & inibidores , Neoplasias/enzimologia , Neoplasias/patologia , Ratos , Células Tumorais Cultivadas , Regulação para Cima
20.
In Vivo ; 16(3): 175-7, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12182112

RESUMO

Mouse leukemia L1210 cells selected for resistance to drugs targeted specifically at each of the protein subunits of ribonucleotide reductase were studied for their ability to grow in vivo. The life-span of the mice injected with hydroxyurea-resistant L1210 cells, which have elevated levels of the mRNA and protein for the non-heme iron (NHI, R2) subunit of ribonucleotide reductase, was approximately twice that of the mice injected with equal numbers of the parental wild-type L1210 leukemia cells. The life-span of mice injected with the L1210 cells that had alterations in the effector-binding subunit (EB, R1) was considerably shorter than the mice injected with the parental wild-type L1210 cells. These results provide direct evidence that tumor cells with alterations in the properties of ribonucleotide reductase grow differently in vivo, with defined effects on the host mouse that cause either an increased survival time or a decreased survival time compared to the effects of wild-type L1210 leukemia cells on tumor-bearing mice.


Assuntos
Leucemia L1210/patologia , Neoplasias Experimentais/patologia , Ribonucleotídeo Redutases/metabolismo , Animais , Resistencia a Medicamentos Antineoplásicos , Hidroxiureia/farmacologia , Leucemia L1210/enzimologia , Leucemia L1210/mortalidade , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/mortalidade , Ferroproteínas não Heme/genética , Ferroproteínas não Heme/metabolismo , RNA Mensageiro/metabolismo , Ribonucleotídeo Redutases/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...