Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 25(1): 85-89, 2017 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-28245380

RESUMO

OBJECTIVE: To investigate the allo-NK cell-mediated killing effect enhanced by decitabine on leukemia stem cells(LSC) and the underlying mechanisms. METHODS: LSC were separated from KG1a cells by using immunomagnetic beads. Allo-NK cells were isolated and purified from PBMC of healthy donors. Cytotoxicity of allo-NK cells against LSC were measured by LDH releasing assay. The apoptosis induced by allo-NK cells in LSC and the expressions of NKG2D ligands including MICA/B and ULBP1-3 on LSC were detected by flow cytometry. RESULTS: The killing rate of allo-NK cells to LSC treated with 10 µmol/L decitabine for 24 hours was significant higher than that to LSC without treatment(60.52%±3.52% vs 22.08%±2.07%, 73.93%±2.33% vs 28. 99%±3.13%, 83.08%±1.32% vs 36.44%±2.40%, respectively)at the effector-target ratios of 5:1, 10:1, 20:1 (P<0.05). At the effector-target ratio of 10:1, decitabine significantly enhanced the apoptosis of LSC induced by allo-NK cells (7.84%±0.34% vs 3.33%±0.64%)(P<0.05). The expressions of NKG2D ligands(MICA/B,ULBP1,ULBP2,ULBP3) on LSC treated with decitabine 10 µmol/L for 24 hours were significantly increased (P<0.05). CONCLUSION: Decitabine may enhance the allo-NK cell-mediated killing effects on LSC by up-regulation of the expressions of NKG2D ligands on LSC.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , Leucemia/tratamento farmacológico , Células-Tronco/efeitos dos fármacos , Azacitidina/farmacologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Decitabina , Antígenos de Histocompatibilidade Classe I , Humanos , Células Matadoras Naturais , Leucócitos Mononucleares , Subfamília K de Receptores Semelhantes a Lectina de Células NK
2.
Onco Targets Ther ; 7: 771-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24899815

RESUMO

BACKGROUND: Manumycin exhibits an antitumor effect in a variety of cancer cell lines, including prostate cancer cell lines (DU145 and PC-3). Our previous studies demonstrated that manumycin induced the apoptosis of anaplastic thyroid cancer cells and leukemia cells via the intrinsic apoptosis pathway. In the current study, we further evaluated the effect of manumycin in two prostate cancer cell lines (LNCaP and 22Rv1), and here we elucidate some of the underlying mechanisms. MATERIALS AND METHODS: The cell viability of prostate cancer cells was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay after treatment with manumycin for 48 hours. Apoptosis was detected by flow cytometry using annexin V and propidium iodide. The expressions of B-cell lymphoma (Bcl)-2 family members and the activations of caspase-9 and caspase-3 were detected by Western blotting. RESULTS: Manumycin treatment resulted in significant decreases in the viabilities of the two prostate cancer cell lines in a dose-dependent manner through apoptosis, and this apoptosis involved caspase-9 activation. A specific inhibitor of caspase-9 protected cells from caspase-3 activation, apoptosis, and cytotoxicity induced by manumycin. We also found that manumycin downregulated Bcl-2 expression and upregulated Bax expression. CONCLUSION: Our data suggest that manumycin induces apoptosis in prostate cancer cells through regulation of the Bcl-2 family involving caspase-9 activation. These results suggest that manumycin may be beneficial for the treatment of prostate cancer.

3.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(1): 250-3, 2013 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-23484730

RESUMO

Drug resistance and relapse are the major challenge for current treatment of acute leukemia. It is critical for ultimately curing leukemia to overcome chemoresistance of leukemic stem cells (LSC) and to eradicate LSC. Recent studies have found that abnormal activated Hedgehog (HH) signaling pathway plays an important role in a wide variety of tumors and regulates multi-drug resistance of LSC. This review briefly summarizes the molecular mechanism of HH signal pathway inducing drug resistance of LSC and leading to novel strategies for eradicating LSC.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Proteínas Hedgehog/metabolismo , Leucemia/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Transdução de Sinais , Humanos
4.
Cancer Lett ; 318(2): 173-9, 2012 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-22198207

RESUMO

Leukemic stem cells (LSCs) play the central role in the relapse and refractory of acute myeloid leukemia (AML) and highlight the critical need for the new therapeutic strategies to directly target the LSC population. However, relatively little is known about the unique molecular mechanisms of drug and natural killer cells (NK)-killing resistance of LSCs because of very small number of LSCs in bone marrow. In this study, we investigated whether established leukemia cell line contains LSCs. We showed that KG1a leukemia cell line contained leukemic stem-like cells, which have been phenotypically restricted within the CD34(+)CD38(-) fractions. CD34(+)CD38(-) cells could generate CD34(+)CD38(+) cells in culture medium and had renewal function. Moreover, CD34(+)CD38(-) cells had self-renewal potential. We found that leukemic stem-like cells from KG1a cells were resistant to chemotherapy and NK-mediated cytotoxicity. NKG2D ligands involve in protecting LSCs from NK-mediated attack. Taken together, our studies provide a novel cell model for leukemic stem cells research. Our data also shed light on mechanism of double resistant to chemotherapy and NK cell immunotherapy, which was helpful for developing novel effective strategies for LSCs.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/patologia , ADP-Ribosil Ciclase 1/imunologia , Antígenos CD34/imunologia , Linhagem Celular Tumoral , Proliferação de Células , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Leucemia Mieloide Aguda/imunologia , Células-Tronco Neoplásicas/imunologia
5.
Arch Med Res ; 41(5): 315-23, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20851287

RESUMO

BACKGROUND AND AIMS: Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide. Aberrant activation of sonic hedgehog (Shh) signaling pathway plays important roles in tumorigenesis and progression of several tumors. Cyclopamine, an important inhibitor of Shh signaling pathway, can induce cell apoptosis. However, the mechanisms underlying cyclopamine-induced apoptosis are not well understood. The aim of this study is to determine the expression of the Shh signaling pathway components in HCC and to investigate the mechanisms underlying cyclopamine-induced apoptosis in HCC cells. METHODS: Shh signaling components (Shh, Ptch, Smo and Gli-1) expression levels were evaluated by immunohistochemistry on tissue microarrays containing 98 HCCs with paired adjacent noncancerous liver tissues. The relationships between sonic hedgehog signal pathway and clinicopathological factors were analyzed in HCC. Cell viability was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. Apoptosis was detected by flow cytometry. mRNA and protein levels were analyzed by RT-PCR and Western blot, respectively. RESULTS: Shh, Ptch, Smo and Gli-1 were overexpressed in HCC tissues compared with paired adjacent noncancerous liver tissue. Activated Shh signaling pathway was associated with tumor size, capsular invasion and vascular invasion in HCC. Cyclopamine remarkably decreased cell viability, induced apoptosis and downregulated Bcl-2 expression in HCC cells. CONCLUSIONS: Shh signaling pathway plays an important role in HCC tumorigenesis and progression, indicating that Shh signaling pathway is a potential therapeutic target for HCC. Cyclopamine induces apoptosis through downregulating Bcl-2 in HCC.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Regulação para Baixo , Proteínas Hedgehog/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais , Alcaloides de Veratrum/farmacologia , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Ai Zheng ; 27(8): 835-9, 2008 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-18710617

RESUMO

BACKGROUND & OBJECTIVE: Repair of DNA damage is important to cell survival. Our previous study showed DNA damage response induced by manumycin in cancer cells. We hypothesized that methoxyamine, an inhibitor of base-excision repair, can enhance the antineoplastic effect of manumycin. This study was to investigate apoptosis induced by manumycin combined with methoxyamine in myeloid leukemia cell line U937, and to explore the role of mitochondrial apoptotic pathway in apoptosis induction of the two drugs. METHODS: U937 cells were treated with various concentrations of manumycin and/or methoxyamine for 48 h. The cell viability was analyzed by MTT assay. Colony formation was evaluated by soft agar clonogenic assay. Cell apoptosis was investigated by flow cytometry. Protein expressions of cytochrome c, caspase-9, and poly ADP-ribose polymerase (PARP) were determined by Western blot. RESULTS: The dose-response curve of manumycin was shifted to the left after addition of methoxyamine. The combination index (CI) was less than 1 (P<0.05) in U937 cells (P<0.05), indicating a synergistic effect of manumycin and methoxyamine. Rates of colony formation of U937 cells treated with 1 micromol/L manumycin, or 5 mmol/L methoxyamine, or the combination of the two were 0.3641+/-0.0463, 0.7541+/-0.0379, and 0.0473+/-0.0024, respectively compared with that of control cells (P<0.05). Moreover, the drug combination resulted in enhanced apoptosis in U937 cells. The apoptotic rates of the control, manumycin, methoxyamine and combination group were (2.34+/-0.30)%, (8.80+/-0.95)%, (2.21+/-0.19)%, and (13.37+/-0.91)%, respectively. The combination of manumycin with methoxyamine also promoted the release of cytochrome c from mitochondria into the cytosol, activated caspase-9, and led appearance of specific cleavage of PARP in U937 cells. CONCLUSION: Methoxyamine enhances manumycin-induced apoptosis in U937 myeloid leukemia cells.


Assuntos
Apoptose/efeitos dos fármacos , Hidroxilaminas/farmacologia , Polienos/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Antineoplásicos/farmacologia , Caspase 9/metabolismo , Citocromos c/metabolismo , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Farnesiltranstransferase/antagonistas & inibidores , Humanos , Mitocôndrias/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Células U937
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 28(2): 173-5, 2008 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-18250035

RESUMO

OBJECTIVE: To study the cytotoxic effect of allogenetic natural killer (NK) cells in vitro on human CD34+ acute myelogenous leukemia cells. METHODS: CD34 expression on acute myelogenous leukemia KG1a cells was detected by flow cytometry. KG1a cells were co-cultured at different effector-to-target (E:T) ratios with NK cells isolated from 5 healthy individuals using magnetic cell sorting. Lactate dehydrogenase (LDH) release assay was employed to examine the cytolysis of KG1a cells in the co-culture, and the inhibition rate of the KG1a cell colony formation in methylcellulose was determined with K562 cells sensitive to NK cells as the control. RESULTS: A expression rate as much as (98.0-/+1.1)% was detected for CD34 antigen on KG1a cells, and the isolated NK cells (CD3(-)CD16+CD56+ cells) had a purity of (93.2-/+3.7)% after magnetic cell sorting. Allogenetic NK cells exhibited obvious cytotoxicity and colony inhibition in vitro against KG1a cells at different E:T ratios, and the effects were significantly enhanced as the E:T ratios increased (P<0.05). At the same E:T ratio, the cytotoxicity and colony inhibition rate of allogenetic NK cells against KG1a cells was lower than those against K562 cells (P<0.05). CONCLUSION: Allogenetic NK cells exhibit obvious cytotoxicity and colony formation against CD34+ acute myelogenous leukemia cells.


Assuntos
Citotoxicidade Imunológica , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/imunologia , Antígenos CD34/imunologia , Técnicas de Cocultura , Citometria de Fluxo , Humanos , Células K562
9.
World J Gastroenterol ; 14(4): 582-9, 2008 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-18203291

RESUMO

AIM: To investigate the effects of Gli-1 small interference RNA (siRNA) on Huh7 cells, and the change of Bcl-2 expression in Huh7 cells. METHODS: Human hepatocellular carcinoma cells Huh7 were used. Cell viability was analyzed by 3-(4, 5-Dimethylthiazol-2-yl)-2, 5-diphenyl tetrazolium bromide (MTT) assay. The expressions of Gli-1 and Bcl-2 family members were detected by RT-PCR and Western blot. Apoptosis was detected by Flow cytometry using propidium iodide, measured by Hoechst 33258 staining using Advanced Fluorescence Microscopy and caspase-3 enzymatic assay. Cell growth was analyzed after treatment with Gli-1 siRNA and 5-fluorouracil (5-Fu). RESULTS: Inhibition of Gli-1 mRNA in Huh7 cells through Gli-1 siRNA reduced cell viability. Gli-1 siRNA treatment also induced apoptosis by three criteria, increase in the sub-G1 cell cycle fraction, nuclear condensation, a morphologic change typical of apoptosis, and activation of caspase-3. Gli-1 siRNA was also able to down-regulate Bcl-2. However, Gli-1 siRNA resulted in no significant changes in Bcl-xl, Bax, Bad, and Bid. Furthermore, Gli-1 siRNA increased the cytotoxic effect of 5-Fu on Huh7 cell. CONCLUSION: Down-regulation of Bcl-2 plays an important role in apoptosis induced by Gli-1 siRNA in HCC cells. Combination Gli-1 siRNA with chemotherapeutic drug could represent a more promising strategy against HCC. The effects of the strategies need further investigation in vivo and may have potential clinical application.


Assuntos
Apoptose/fisiologia , Carcinoma Hepatocelular/patologia , Hepatócitos/fisiologia , Neoplasias Hepáticas/patologia , Fatores de Transcrição/genética , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Genes bcl-2/fisiologia , Terapia Genética/métodos , Hepatócitos/citologia , Humanos , Neoplasias Hepáticas/terapia , RNA Interferente Pequeno/farmacologia , Proteína GLI1 em Dedos de Zinco
10.
Zhonghua Xue Ye Xue Za Zhi ; 28(6): 404-6, 2007 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-17939408

RESUMO

OBJECTIVE: To investigate the apoptosis induced by manumycin in U937 and HL-60 cell lines, and to explore the role of mitochondria apoptotic pathway in manumycin-inducing apoptosis. METHODS: Leukemic cells line U937 and HL-60 were treated by manumycin at 2 micromol/L for different time. Apoptosis of leukemia cells was detected by flow cytometry. The cytosolic proteins were extracted using a digitonin buffer. The protein expression of cytochrome C, caspase-9, caspase-8, and caspase-3 were determined by western blot. Mitochondrial membrane potential was detected by JC-1. RESULTS: In U937 and HL-60 cells, manumycin induced mitochondrial depolarization after 6 h treatment. The average red/green fluorescence ratios at 6 h were significantly (P < 0.01) lower than those at time 0, being 0.51 +/- 0.07 and 0.41 +/- 0.06 for control group respectively. Manumycin induced cytochrome C release from the mitochondria into the cytosol after 6 h treatment, and activated caspase-9, caspase-8, and caspase-3 after a 16h treatment. The broad-spectrum caspase-inhibitor Z-VAD-fmk at 50 micromol/L was able to inhibit caspase cleavage completely, but only reduced the manumycin-induced apoptosis rates by 51.69% and 56.47% in U937 and HL-60, respectively. CONCLUSION: Manumycin induced apoptosis in U937 and HL-60 cell lines via mitochondria apoptotic pathway.


Assuntos
Apoptose/efeitos dos fármacos , Mitocôndrias/fisiologia , Polienos/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Citocromos c/metabolismo , Células HL-60 , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo
11.
Acta Pharmacol Sin ; 28(7): 1037-44, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17588341

RESUMO

AIM: To investigate the effects of 2-methoxyestradiol (2-ME) on 2 myeloid leukemia cell lines HL-60 and U937, and to explore its mechanisms. METHODS: Human myeloid leukemia cells HL-60 and U937 were used. Measurement of mitochondrial membrane potential (Dym) was performed using 5,5',6,6'-Tetrachloro-1,1',3,3'- tetraethylbenzimidazolylcarbocyanine iodide ( JC-1). Apoptosis and cellular nitric oxide (NO) were detected by flow cytometry using Annexin V and NO sensor dye. Superoxide anion was measured with a fluorescent plate reader by dihydroethidium (DHE). Cytotoxicity was analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay. RESULTS: 2-ME resulted in viability decrease in a dose-dependent manner. 2-ME treatment also generated reactive oxygen species (ROS), including NO and superoxide anions, which resulted in mitochondria damage. 2-ME-induced apoptosis was correlated with an increase in ROS. The quenching of ROS with N-acetyl-L-cysteine protected leukemia cells from 2-ME cytotoxicity and prevented apoptosis induction by 2-ME. Furthermore, the addition of manumycin, a farnesyltransferase inhibitor, significantly enhanced apoptosis induced by 2-ME. CONCLUSION: Cellular ROS generation plays an important role in the cytotoxic effect of 2-ME. It is possible to use ROS generation agents, such as manumycin, to enhance the antileukemic effect. The combination strategy needs further in vivo justification and may have potential clinical application.


Assuntos
Apoptose , Estradiol/análogos & derivados , Leucemia/metabolismo , Espécies Reativas de Oxigênio/metabolismo , 2-Metoxiestradiol , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Estradiol/farmacologia , Células HL-60 , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Óxido Nítrico/metabolismo , Polienos/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Superóxidos/metabolismo , Moduladores de Tubulina/farmacologia , Células U937
12.
J Clin Endocrinol Metab ; 92(8): 2902-9, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17550961

RESUMO

CONTEXT: Anaplastic thyroid cancer (ATC) is extremely aggressive, and no effective treatment is available. Combretastatin A4 phosphate (CA4P), a vascular disrupting agent, has limited activity against ATC in a clinical trial, and so does paclitaxel. OBJECTIVE: We hypothesized that a triple-drug combination including CA4P and paclitaxel would improve efficacy against ATC. Therefore, we evaluated two such combinations in vivo. SETTING: We used a nude mouse xenograft model with ARO and KAT-4 cells. INTERVENTIONS: The first combination consisted of CA4P, paclitaxel, and manumycin A (a farnesyltransferase inhibitor), and the second, CA4P, paclitaxel, and carboplatin. MAIN OUTCOME MEASURES: Main outcome measures included tumor growth curves and tumor weights. RESULTS: Tumor growth curve analysis (linear mixed models, P < 0.05) and xenograft weight analysis (Kruskal-Wallis one-way ANOVA on ranks, post hoc pairwise comparison, Dunn's test, P < 0.05) demonstrated that both triple-drug combinations were significantly better than placebo for both cell lines. Anti-bromodeoxyuridine immunostaining of xenograft sections from animals injected with bromodeoxyuridine before being killed showed that CA4P alone did not inhibit DNA synthesis, but manumycin A and paclitaxel did. CA4P decreased the depth of the viable outer rim of tumor cells on xenograft sections. Using electron microscopy, we found blebbing/budding of endothelial cells into capillary lumens and autophagy of tumor cells in CA4P-treated xenografts. CONCLUSIONS: Both triple-drug combinations demonstrated excellent antineoplastic activity against ATC. The correlative findings in xenografts were consistent with vascular disruption but not direct inhibition of cell proliferation as the primary antineoplastic mechanism contributed by CA4P. These regimens warrant further investigation in clinical trials for ATC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma/tratamento farmacológico , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Antibióticos Antineoplásicos/uso terapêutico , Antimetabólitos Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos/administração & dosagem , Bromodesoxiuridina/metabolismo , Carboplatina/uso terapêutico , Carcinoma/patologia , Linhagem Celular Tumoral , Forma Celular , Células Endoteliais/patologia , Células Endoteliais/ultraestrutura , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Transplante de Neoplasias , Paclitaxel/administração & dosagem , Polienos/uso terapêutico , Alcamidas Poli-Insaturadas/uso terapêutico , Estilbenos/administração & dosagem , Neoplasias da Glândula Tireoide/patologia , Transplante Heterólogo
13.
Cancer Biol Ther ; 5(3): 275-80, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16410725

RESUMO

Our previous studies demonstrated that manumycin A, a farnesyltransferase inhibitor, induced apoptosis of anaplastic thyroid cancer cells via the intrinsic apoptosis pathway and induced reactive oxygen species (ROS), which mediated DNA damage. In this study, we investigated the hypothesis that the mechanism of apoptosis induced by manumycin in anaplastic thyroid cancer cells fits the general pattern of the "xenobiotic apoptosis pathway," the hallmarks of which are induction of oxidative stress, mitogen-activated protein kinase (MAPK) signaling, and cytochrome c release, which activates the intrinsic apoptosis pathway. We found that manumycin reduced intracellular glutathione and generated ROS: nitric oxide and superoxide anions. Manumycin-induced apoptosis correlated with increase in ROS. Quenching of ROS with N-acetyl-L-cysteine prevented cytochrome c release by manumycin. Manumycin induced phosphorylation of p38 MAPK, which was blocked by N-acetyl-L-cysteine. p38 MAPK may be an important signaling mediator in the activation of the intrinsic apoptotic pathway by manumycin because the p38 MAPK inhibitor SB203580 inhibited cytochrome c release and activation of caspase-3 by manumycin. In conclusion, manumycin activated the intrinsic apoptosis pathway via activation of p38 MAPK by oxidative stress. The mechanism of apoptosis induced by manumycin fits the emerging general pattern for apoptosis induced by xenobiotics.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Polienos/metabolismo , Polienos/farmacologia , Alcamidas Poli-Insaturadas/metabolismo , Alcamidas Poli-Insaturadas/farmacologia , Neoplasias da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/patologia , Antineoplásicos/metabolismo , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Glutationa/metabolismo , Humanos , Immunoblotting , Óxido Nítrico/metabolismo , Oxirredução , Estresse Oxidativo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Cancer Lett ; 238(2): 197-201, 2006 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16154259

RESUMO

We previously showed that the in vivo anticancer effects of a combination of manumycin (a farnesyltransferase inhibitor) and paclitaxel (a microtubule inhibitor) against anaplastic thyroid carcinoma (ATC) were partially due to inhibition of angiogenesis. In this study, we investigated the effect of adding minocycline (a matrix metalloproteinase inhibitor) to manumycin and paclitaxel against human ATC cells xenografted in nude mice. The triple-drug combination resulted in the lowest average tumor growth rate, and it conferred significantly better survival than manumycin alone, paclitaxel alone, or manumycin plus paclitaxel. In conclusion, this novel combination deserves further investigation in the treatment of ATC.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Inibidores Enzimáticos/administração & dosagem , Farnesiltranstransferase/antagonistas & inibidores , Inibidores de Metaloproteinases de Matriz , Neoplasias da Glândula Tireoide/tratamento farmacológico , Animais , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Minociclina/administração & dosagem , Transplante de Neoplasias , Paclitaxel/administração & dosagem , Paclitaxel/efeitos adversos , Polienos/administração & dosagem , Alcamidas Poli-Insaturadas , Neoplasias da Glândula Tireoide/mortalidade , Neoplasias da Glândula Tireoide/patologia , Transplante Heterólogo
15.
Cancer Res ; 65(9): 3671-81, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15867362

RESUMO

Farnesyltransferase inhibitors (FTIs) possess antitumor activity. Based on recent findings, we hypothesized that FTIs induce reactive oxygen species (ROS) that damage DNA, leading to DNA damage responses. To test this hypothesis, we investigated the effects of FTIs on the generation of ROS, DNA double-strand breaks (DSB), DNA damage responses, and RhoB, and the effects of quenching ROS on these FTI effects. We evaluated four FTIs in human cancer cell lines of different tissue origins. We found that FTIs induced ROS and DSBs. Suppressing expression of the beta-subunit of farnesyltransferase with siRNA did not induce ROS, but slightly attenuated the ROS induced by FTIs. N-acetyl-L-cysteine (NAC), but not caspase inhibitors, blocked FTI-induced DSBs, suggesting that the DSBs were caused by ROS and did not result from apoptosis. The DSBs led to DNA damage responses. H2AX became phosphorylated and formed nuclear foci. The DNA-damage-sensing molecules involved were probably ataxia-telangiectasia mutated protein (ATM) and DNA-dependent protein kinase (DNA-PK) but not ATM- and Rad3-related protein (ATR). Key components of the homologous recombination and nonhomologous end joining repair pathways (DNA-PK, BRCA1, and NBS1) underwent phosphorylation and formed nuclear foci. RhoB, a mediator of the antineoplastic effect of FTIs and a protein inducible by DNA damage, was increased by FTIs. This increase was blocked by NAC. We concluded that FTIs induced oxidative DNA damage by inducing ROS and initiated DNA damage responses, including RhoB induction, and there was a complex relationship among FTIs, farnesyltransferase, ROS, and RhoB. Our data also imply that inhibitors of DNA repair may accentuate the clinical efficacy of FTIs.


Assuntos
Alquil e Aril Transferases/antagonistas & inibidores , Dano ao DNA , Inibidores Enzimáticos/farmacologia , Neoplasias/genética , Neoplasias/metabolismo , Polienos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , DNA de Neoplasias/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/metabolismo , Farnesiltranstransferase , Células HCT116 , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Proteínas Nucleares , Alcamidas Poli-Insaturadas , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína rhoB de Ligação ao GTP/biossíntese , Proteína rhoB de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA