Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
2.
Bioelectromagnetics ; 35(6): 406-13, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24839179

RESUMO

Previously, we found that extremely low frequency (ELF) electric fields were able to elicit an approximate 3.5-fold increase in heat shock gene expression, a response which may have applicability to cancer therapy. Based on recent studies demonstrating the ability of magnetic fields to influence gene expression, we hypothesized that low level static magnetic fields may be able to affect heat shock gene expression while avoiding some of the clinical difficulties that arise with electric fields. Transfected rat primary cells in monolayer were exposed to magnetic fields of 1 to 440 mT for 16, 24, or 48 h starting at 24 and 48 h post transfection. Heat shock protein (HSP70) expression, as indicated by a promoter linked luciferase reporter, was followed for up to 96 h and showed a dependence on flux density, exposure duration, and start time post transfection. A nonlinear response was observed for increasing flux density with a maximum of a 3.5-fold increase in expression for 48 h of exposure starting 48 h after transfection. These results demonstrate an enhancement of gene expression similar in magnitude to that observed with external electric field exposure, while eliminating many of the clinical complications.


Assuntos
Campos Eletromagnéticos , Proteínas de Choque Térmico HSP70/metabolismo , Animais , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Expressão Gênica/efeitos da radiação , Proteínas de Choque Térmico HSP70/genética , Dinâmica não Linear , Ratos , Fatores de Tempo , Transfecção
3.
Bioelectromagnetics ; 34(5): 405-13, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23457046

RESUMO

Recent studies have demonstrated that the Ku70 gene fragment can be placed in the anti-sense orientation under the control of a heat-inducible heat shock protein 70 (HSP70) promoter and activated through heat shock exposure. This results in attenuation of the Ku70 protein expression, inhibiting cellular repair processes, and sensitizing the transfected cells to exposures such as the ionizing radiation exposures used clinically. However, achieving the tissue temperatures necessary to thermally induce the HSP70 response presents significant limitations to the clinical application of this strategy. Previous findings suggest an alternative approach to inducing a heat shock response, specifically through the use of extremely low frequency (ELF) electrical field stimulation. To further pursue this approach, we investigated HSP70 responses in transfected rat primary fibroblast (RAT1) cells exposed to 10 Hz electric fields at intensities of 20-500 V/m. We confirmed that low frequency electric fields can induce HSP70 heat shock expression, with peak responses obtained at 8 h following a 2 h field exposure. However, the approximate threefold increase in expression is substantially lower than that obtained using thermal stimulation, raising questions of the clinical utility of the response.


Assuntos
Campos Eletromagnéticos , Proteínas de Choque Térmico HSP70/efeitos da radiação , Animais , Antígenos Nucleares/genética , Técnicas de Cultura de Células , Linhagem Celular , Proteínas de Ligação a DNA/genética , Fibroblastos/efeitos da radiação , Autoantígeno Ku , Luciferases , Substâncias Luminescentes , Regiões Promotoras Genéticas/genética , Doses de Radiação , Ratos , Fatores de Tempo , Ativação Transcricional/efeitos da radiação , Transfecção
4.
Int J Hyperthermia ; 26(3): 224-31, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20230250

RESUMO

Clinically achievable mild temperature local hyperthermia (<43 degrees C) has been demonstrated to be an effective adjuvant to radiotherapy in pre-clinical and clinical studies. In this article, we briefly review the recent progress in the following areas: (1) the effect of mild temperature hyperthermia (MTH) on tumour hypoxia and blood perfusion as assessed by dual marker immunohistochemistry (IHC); (2) the kinetics of MTH induced changes in tumour hypoxia; (3) the potential role of heat-induced tumour reoxygenation on radio- and chemo-sensitisation; (4) the potential role of MTH in combination with vascular targeting agents (VTAs) on tumour response; and (5) non-invasive detection of changes in tumour oxygenation and blood perfusion. It is shown that MTH, by itself or in combination with VTAs, leads to changes in tumour perfusion and oxygenation with potential for radio- and chemo-sensitisation.


Assuntos
Hipertermia Induzida/métodos , Microcirculação , Neoplasias/irrigação sanguínea , Neoplasias/terapia , Hemorreologia , Humanos , Hipóxia , Consumo de Oxigênio , Tolerância a Radiação
5.
Int J Hyperthermia ; 25(4): 258-61, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19670094

RESUMO

Of the many heat shock proteins (HSPs), hsp70 appears to correlate best with heat resistance, either permanent or transient. We have investigated various approaches to quantify the concentration of hsp70, and examined the relationship between hsp70 and cells' thermal sensitivity during the development and decay of thermotolerance in model systems. Specifically, experiments were performed to determine the possibility of using the rate of synthesis of hsp70 after a second test heat shock to predict the kinetics of thermotolerance in tumor cells in vitro and in animal tumor models. We found that the cells' ability to re-initiate hsp70 synthesis in response to the test heat shock inversely correlated with retained thermotolerance. These data suggest the level of hsp70 in thermotolerant cells regulates the rate of synthesis of additional hsp70 in response to the subsequent heat challenge. Furthermore, the results showed that the rate of re-induction of hsp70 synthesis after a test heat shock can be used as a rapid measure of retained thermotolerance. This study suggests an approach for quantifying the level of retained thermotolerance during fractionated hyperthermia.


Assuntos
Proteínas de Choque Térmico HSP70/biossíntese , Resposta ao Choque Térmico/fisiologia , Animais , Linhagem Celular Tumoral , Humanos , Hipertermia Induzida , Neoplasias/metabolismo , Neoplasias/terapia
6.
Biotechnol Appl Biochem ; 52(Pt 2): 113-9, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18289126

RESUMO

The apoptotic effects of natural (n-) and synthetic (s-) DADS (diallyl disulfide; 3,3'-thiobisprop-1-ene) on human breast-cancer MCF-7 cells were investigated in vitro. 5-Fu (5-fluorouracil) and CTX (cyclophosphamide; Cytoxan) were used as comparative control anticancer agents. After MCF-7 cells had been treated with the drugs, cell viability, morphological change, apoptosis and changes in the cell cycle were measured. The results indicated that s- and n-DADS had similar cytotoxicities towards human breast-cancer MCF-7 cells and showed dose-dependent and time-dependent inhibitory effects. Morphological observations and flow-cytometric results showed that the four drugs (s-DADS, n-DADS, CTX and 5-Fu) induced apoptosis in MCF-7 cells to different extents. The apoptotic effects of s- and n-DADS were superior to those of 5-Fu and CTX.


Assuntos
Compostos Alílicos/síntese química , Compostos Alílicos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Produtos Biológicos/farmacologia , Neoplasias da Mama/patologia , Dissulfetos/síntese química , Dissulfetos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos
7.
Cancer Res ; 67(2): 634-42, 2007 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17234773

RESUMO

Ku70 is one component of a protein complex, the Ku70/Ku80 heterodimer, which binds to DNA double-strand breaks and activates DNA-dependent protein kinase (DNA-PK), leading to DNA damage repair. Our previous work has confirmed that Ku70 is important for DNA damage repair in that Ku70 deficiency compromises the ability of cells to repair DNA double-strand breaks, increases the radiosensitivity of cells, and enhances radiation-induced apoptosis. Because of the radioresistance of some human cancers, particularly glioblastoma, we examined the use of a radio-gene therapy paradigm to sensitize cells to ionizing radiation. Based on the analysis of the structure-function of Ku70 and the crystal structure of Ku70/Ku80 heterodimer, we designed and identified a candidate dominant negative fragment involving an NH(2)-terminal deletion, and designated it as DNKu70. We generated this mutant construct, stably overexpressed it in Rat-1 cells, and showed that it has a dominant negative effect (i.e., DNKu70 overexpression results in decreased Ku-DNA end-binding activity, and increases radiosensitivity). We then constructed and generated recombinant replication-defective adenovirus, with DNKu70 controlled by the cytomegalovirus promoter, and infected human glioma U-87 MG cells and human colorectal tumor HCT-8 cells. We show that the infected cells significantly express DNKu70 and are greatly radiosensitized under both aerobic and hypoxic conditions. The functional ramification of DNKu70 was further shown in vivo: expression of DNKu70 inhibits radiation-induced DNA-PK catalytic subunit autophosphorylation and prolongs the persistence of gamma-H2AX foci. If radiation-resistant tumor cells could be sensitized by down-regulating the cellular level/activity of Ku/DNA-PK, this approach could be evaluated as an adjuvant to radiation therapy.


Assuntos
Antígenos Nucleares/fisiologia , Neoplasias Colorretais/radioterapia , Proteínas de Ligação a DNA/fisiologia , Glioma/radioterapia , Adenoviridae/genética , Aerobiose , Animais , Antígenos Nucleares/biossíntese , Antígenos Nucleares/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Terapia Combinada , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Terapia Genética/métodos , Glioma/genética , Glioma/metabolismo , Histonas/biossíntese , Humanos , Autoantígeno Ku , Camundongos , Fosforilação/efeitos da radiação , Tolerância a Radiação/genética , Ratos , Transfecção
8.
Int J Radiat Oncol Biol Phys ; 71(2): 533-41, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18374502

RESUMO

PURPOSE: To evaluate the response of cells over-expressing dominant negative (DN) Ku70 to single and multiple small radiation doses. METHODS AND MATERIALS: Clones of fibroblasts over-expressing DNKu70, DNKu70-7, DNKu70-11, and parental Rat-1 cells were irradiated under oxic or hypoxic conditions with single or multiple doses. Cells were trypsinized 0 or 6 h after irradiation to determine surviving fraction (SF). RESULTS: Oxic DNKu70-7 or -11 cells trypsinized 6 h after irradiation were 1.52 or 1.25 and 1.28 or 1.15 times more sensitive than oxic Rat-1 at SF of 0.5 and 0.1, respectively. Hypoxic DNKu70-7 or -11 cells trypsinized 6 h after irradiation were 1.44 or 1.70 and 1.33 or 1.51 times more sensitive than hypoxic Rat-1 at SF of 0.5 and 0.1, respectively. To the multiple doses, oxic and hypoxic DNKu70-7 or -11 cells were 1.35 or 1.37 and 2.23 or 4.61 times more sensitive than oxic and hypoxic Rat-1, respectively, resulting in very small oxygen enhancement ratios. Namely, enhancement caused by DNKu70 under hypoxia after multiple doses was greater than that under oxic conditions and greater than that after single dose. CONCLUSIONS: Over-expression of DNKu70 enhances cells' response to radiation given as a single dose and as multiple small doses. The enhancement after multiple doses was stronger under hypoxic than under oxic conditions. These results encourage the use of DNKu70 fragment in a gene-radiotherapy.


Assuntos
Antígenos Nucleares/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/efeitos da radiação , Tolerância a Radiação/fisiologia , Animais , Antígenos Nucleares/genética , Antígenos Nucleares/uso terapêutico , Hipóxia Celular , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Dano ao DNA , Reparo do DNA , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/uso terapêutico , Relação Dose-Resposta à Radiação , Fibroblastos/metabolismo , Autoantígeno Ku , Modelos Lineares , Doses de Radiação , Ratos , Fatores de Tempo
9.
Radiother Oncol ; 88(2): 269-76, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18538874

RESUMO

PURPOSE: To study the changes in hypoxia resulting from mild temperature hyperthermia (MTH) in a subcutaneous xenograft model using dual-tracer immunohistochemical techniques. MATERIALS AND METHODS: HT29 tumors were locally heated at 41 degrees C. Changes in tumor hypoxia were investigated by pimonidazole and EF5. Pimonidazole was given 1h preheating, EF5 at various times during or after treatment, 1h later the animals were sacrificed. Blood vessels were identified by CD31 staining, and perfusion by Hoechst 33342 injected 1 min pre-sacrifice. RESULTS: The overall hypoxic fraction was significantly decreased by MTH during and immediately after heating. However, MTH induced both increases and decreases in tumor hypoxia in different parts of the tumor. Specifically, MTH decreased hypoxia in the regions with relatively well-perfused blood vessels, but increased hypoxia in regions that were poorly perfused. At 24-h post heating, newly formed hypoxic regions surrounded previously-hypoxic foci, which in turn surrounded pimonidazole-stained debris. Quantitative analysis did not evince changes in tumor oxygenation due to MTH at 24h post-treatment. CONCLUSION: In this xenograft model, the effect of MTH on tumor oxygenation was variable, both spatially and kinetically. Overall tumor oxygenation was improved during and after heating, but the effect was short-lived.


Assuntos
Adenocarcinoma/metabolismo , Neoplasias Colorretais/metabolismo , Hipertermia Induzida , Imuno-Histoquímica/métodos , Oxigênio/metabolismo , Radiossensibilizantes/farmacologia , Adenocarcinoma/irrigação sanguínea , Análise de Variância , Animais , Hipóxia Celular , Neoplasias Colorretais/irrigação sanguínea , Etanidazol/análogos & derivados , Etanidazol/farmacologia , Feminino , Hidrocarbonetos Fluorados/farmacologia , Camundongos , Nitroimidazóis/farmacologia , Transplante Heterólogo
10.
Radiat Res ; 169(1): 67-75, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18159950

RESUMO

Wen, B., Urano, M., Humm, J. L., Seshan, V. E., Li, G. C. and Ling, C. C. Comparison of Helzel and OxyLite Systems in the Measurements of Tumor Partial Oxygen Pressure (pO(2)). Radiat. Res. 168, 67-75 (2008). It has been demonstrated in both experimental and human malignancies that hypoxic tumor cells are linked with aggressive disease phenotype. One of the methods to identify these cells is by direct physical measurement of tumor pO(2). This study compared pO(2) values measured with two systems, the Helzel Hypoximeter (successor of the polarographic Eppendorf electrode) and the Oxford-Optronix OxyLite (fiber-optic probe), in R3327-AT and R3327-AT/tkeGFP tumors. Partial oxygen pressure was measured in individual tumors with either system or in the same tumor with both systems. The similarities and discrepancies in pO(2) measurements between the two systems were also investigated when tumor-bearing animals were breathing pure oxygen. Our data showed a considerable heterogeneity in pO(2) values in each tumor using both the Helzel and OxyLite systems. Similar results were obtained with both systems for the mean and median pO(2) values, and the distributions of pO(2) values within the interval 0 < pO(2) < 40 mmHg (the range important for defining tumor hypoxia) were found to be statistically equivalent. However, the frequencies of high pO(2) values (>40 mmHg) and zero values measured by the two systems were statistically significantly different.


Assuntos
Neoplasias , Oxigênio/metabolismo , Animais , Humanos , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/metabolismo , Neoplasias/patologia , Pressão Parcial
11.
Acta Oncol ; 47(7): 1211-20, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18661431

RESUMO

INTRODUCTION: Increased expression of cytosine deaminase (CD) and uracil phosphoribosyltransferase (UPRT) may improve the antitumoral effect of 5-fluorouracil (5-FU) and 5-fluorocytosine (5-FC), and thereby enhance the potential of gene-directed enzyme prodrug therapy. For the applicability of gene-directed enzyme prodrug therapy in a clinical setting, it is essential to be able to monitor the transgene expression and function in vivo. Thus, we developed a preclinical tumor model to investigate the feasibility of using magnetic resonance spectroscopy and optical imaging to measure non-invasively CD and UPRT expression and function. MATERIALS AND METHODS: Expression vectors of CD or CD/UPRT fused to monomeric DsRed (mDsRed) were constructed and rat prostate carcinoma (R3327-AT) cell lines stably expressing either CD/mDsRed or CD/UPRT/mDsRed were generated. The expression of the fusion proteins was evaluated by flow cytometry, fluorescence microscopy, and Western blot analysis. The function of the fusion protein was confirmed in vitro by assessing 5-FC and 5-FU cytotoxicity. In vivo fluorine-19 magnetic resonance spectroscopy ((19)F MRS) was used to monitor the conversion of 5-FC to 5-FU in mice bearing the R3327-CD/mDsRed and R3327-CD/UPRT/mDsRed tumor xenografts. RESULTS: Sensitivity to 5-FC and 5-FU was higher in cells stably expressing the CD/UPRT/mDsRed fusion gene than in cells stably expressing CD/mDsRed alone or wild-type cells. Whole tumor (19)F MRS measurements showed rapid conversion of 5-FC to 5-FU within 20 min after 5-FC was administered intravenously in both CD/mDsRed and CD/UPRT/mDsRed tumors with subsequent anabolism to cytotoxic fluoronucleotides (FNucs). CD/UPRT/mDsRed tumor was more efficient in these processes. CONCLUSION: This study demonstrates the utility of these tumor models stably expressing CD or CD/UPRT to non-invasively evaluate the efficacy of the transgene expression/activity by monitoring drug metabolism in vivo using MRS, with potential applications in preclinical and clinical settings.


Assuntos
Citosina Desaminase/análise , Proteínas Luminescentes/análise , Espectroscopia de Ressonância Magnética , Pentosiltransferases/análise , Animais , Western Blotting , Linhagem Celular Tumoral , Citometria de Fluxo , Radioisótopos de Flúor , Genes Reporter/fisiologia , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata , Ratos , Sensibilidade e Especificidade , Transfecção , Transgenes/fisiologia , Proteína Vermelha Fluorescente
12.
J Agric Food Chem ; 56(3): 910-5, 2008 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-18193834

RESUMO

The mechanism of enhanced effect of (-)-epigallocatechin-3-gallate (EGCG) on huperzine A's (HUP) inhibition of acetylcholinesterase (AChE) activity in rats was investigated. The inhibitory effects of HUP at 10 and 5 microg/kg on AChE activity were quite weak in the whole phase. In contrast, upon addition of EGCG (100 mg/kg) to the HUP 10 and 5 microg/kg groups, remarkably enhanced inhibitory effects with maximum inhibitory percentages of 90.94 and 88.13% were observed under the same conditions. EGCG also can greatly prolong the inhibitory time. The mechanism of the enhanced effects of EGCG on HUP's inhibition of AChE activity was investigated by steady fluorescence spectroscopy, infrared spectroscopy, and ultraviolet spectroscopy. HUP hardly interacted with the main transport protein, whereas there was a very strong binding interaction between EGCG and bovine serum albumin. The enhanced transport of HUP is a possible cause of the enhanced effect of EGCG on HUP bioactivity.


Assuntos
Acetilcolinesterase/metabolismo , Catequina/análogos & derivados , Inibidores da Colinesterase/farmacologia , Sesquiterpenos/farmacologia , Alcaloides , Animais , Catequina/farmacologia , Cinética , Masculino , Ratos , Ratos Sprague-Dawley , Espectrometria de Fluorescência , Espectrofotometria Ultravioleta , Espectroscopia de Infravermelho com Transformada de Fourier
13.
Oncol Rep ; 39(3): 912-920, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29344644

RESUMO

The DNA-dependent protein kinase (DNA-PK) complex plays a pivotal role in non-homologous end-joining (NHEJ) repair. We investigated the mechanism of NU7441, a highly selective DNA-PK inhibitor, in NHEJ-competent mouse embryonic fibroblast (MEF) cells and NHEJ-deficient cells and explored the feasibility of its application in radiosensitizing nasopharyngeal carcinoma (NPC) cells. We generated wild-type and DNA-PKcs-/- MEF cells. Clonogenic survival assays, flow cytometry, and immunoblotting were performed to study the effect of NU7441 on survival, cell cycle, and DNA repair. NU7441 profoundly radiosensitized wild-type MEF cells and SUNE-1 cells, but not DNA-PKcs-/- MEF cells. NU7441 significantly suppressed radiation-induced DSB repair post-irradiation through unrepaired and lethal DNA damage, the cell cycle arrest. The effect was associated with the activation of cell cycle checkpoints. The present study revealed a mechanism by which inhibition of DNA-PK sensitizes cells to irradiation suggesting that radiotherapy in combination with DNA-PK inhibitor is a promising paradigm for the management of NPC which merits further investigation.


Assuntos
Carcinoma/patologia , Cromonas/farmacologia , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Raios gama/efeitos adversos , Morfolinas/farmacologia , Neoplasias Nasofaríngeas/patologia , Proteínas Nucleares/fisiologia , Radiossensibilizantes/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Carcinoma/genética , Carcinoma/terapia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Células Cultivadas , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibroblastos/efeitos da radiação , Humanos , Camundongos , Camundongos Knockout , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/terapia
14.
Oncotarget ; 8(14): 22662-22673, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-28186989

RESUMO

DNA-dependent protein kinase catalytic subunit (DNA-PKcs) is a distinct factor in the non-homologous end-joining (NHEJ) pathway involved in DNA double-strand break (DSB) repair. We examined the crosstalk between key proteins in the DSB NHEJ repair pathway and cell cycle regulation and found that mouse embryonic fibroblast (MEF) cells deficient in DNA-PKcs or Ku70 were more vulnerable to ionizing radiation (IR) compared with wild-type cells and that DSB repair was delayed. γH2AX was associated with phospho-Ataxia-telangiectasia mutated kinase (Ser1987) and phospho-checkpoint effector kinase 1 (Ser345) foci for the arrest of cell cycle through the G2/M phase. Inhibition of DNA-PKcs prolonged IR-induced G2/M phase arrest because of sequential activation of cell cycle checkpoints. DSBs were introduced, and cell cycle checkpoints were recruited after exposure to IR in nasopharyngeal carcinoma SUNE-1 cells. NU7441 radiosensitized MEF cells and SUNE-1 cells by interfering with DSB repair. Together, these results reveal a mechanism in which coupling of DSB repair with the cell cycle radiosensitizes NHEJ repair-deficient cells, justifying further development of DNA-PK inhibitors in cancer therapy.


Assuntos
Carcinoma/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/genética , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/antagonistas & inibidores , Autoantígeno Ku/fisiologia , Neoplasias Nasofaríngeas/genética , Proteínas Nucleares/antagonistas & inibidores , Tolerância a Radiação/genética , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Carcinoma/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos da radiação , Proliferação de Células , Quinase 1 do Ponto de Checagem/metabolismo , Cromonas/farmacologia , Proteína Quinase Ativada por DNA/fisiologia , Proteínas de Ligação a DNA/fisiologia , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/efeitos da radiação , Camundongos , Camundongos Knockout , Morfolinas/farmacologia , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patologia , Proteínas Nucleares/fisiologia , Radiação Ionizante , Radiossensibilizantes/farmacologia , Células Tumorais Cultivadas
15.
Arch Pharm Res ; 29(12): 1125-31, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17225462

RESUMO

This study was undertaken to elucidate the effect of diallyl disulfide (DADS), an oil-soluble organosulfur compound found in garlic, in suppressing human nasopharyngeal carcinoma cells. A potent increase (of at least 9-fold) in apoptotic cells has accompanied 1) a decrease in cell viability, 2) a increase of the fraction of S-phase cells by up to 63.8%, and 3) a transient increase of the phospho-p38 and phospho-p42/44 (phosphorylated p38 MAPK and phosphorylated p42/44 MAPK) in a time- and concentration-dependent manner. These results indicate that DADS can induce apoptosis in human nasopharyngeal carcinoma cells via, at least partly, S-phase block of the cell cycle, related to a rise in MAPK phosphorylation.


Assuntos
Compostos Alílicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma/tratamento farmacológico , Dissulfetos/farmacologia , Neoplasias Nasofaríngeas/tratamento farmacológico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Alho/química , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Fosforilação , Fase S/efeitos dos fármacos
16.
Cancer Res ; 62(21): 6031-4, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12414624

RESUMO

There are two major pathways for repairing DNA double strand breaks in mammalian cells: nonhomologous end joining (NHEJ) and homologous recombination repair (HRR). The nonhomologous end joining repair is deficient in cells without Ku, whereas HRR is highly efficient in such cells compared with their wild-type counterparts. The mechanism remains unclear. We reported previously that Ku80(-/-) cells show a stronger ATM-dependent S-phase checkpoint response than Ku80(+/+) cells after ionizing radiation (IR; X-Y. Zhou et al., Oncogene, 21:6377-6381, 2002). We report in this study that Ku80(-/-) cells also show a much stronger G(2) accumulation than Ku80(+/+) cells after IR. The stronger G(2) checkpoint response in Ku80(-/-) cells is ATM independent but is accompanied with a higher activity of CHK1 kinase. Treatment with Chk1 antisense oligonucleotide abolishes the stronger G(2) checkpoint response and sensitizes Ku80(-/-) cells to IR. These data indicate that the stronger G(2) checkpoint response shown in Ku80(-/-) cells is CHK1 dependent and suggest that the CHK1-dependent checkpoint response contributes to the highly efficient HRR in such cells.


Assuntos
Antígenos Nucleares , DNA Helicases , Proteínas de Ligação a DNA/fisiologia , Fase G2/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Quinases/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular , Quinase 1 do Ponto de Checagem , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Fase G2/genética , Fase G2/efeitos da radiação , Humanos , Autoantígeno Ku , Camundongos , Proteínas Nucleares/genética , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Transfecção , Proteínas Supressoras de Tumor
17.
Cancer Res ; 62(9): 2483-7, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11980637

RESUMO

Camptothecin (CPT) that targets DNA topoisomerase I is one of the most promising broad-spectrum anticancer drugs in development today. The cytotoxicity of CPT is S phase (S)-specific because the collision of advancing replication forks with CPT-topoisomerase I-DNA complexes results in DNA damage. After DNA damage, proliferating cells could actively slow down the DNA replication through an S checkpoint to provide time for repair. We report now that there is an activated S checkpoint response in CPT-treated mammalian cells. This response is regulated by Ataxia and Rad3-related (ATR)/CHK1 pathway. Compared with their wild-type counterparts, CPT-treated Ku80-/- cells showed stronger inhibition of DNA replication. This stronger inhibition had no relationship with DNA-dependent protein kinase (DNA-PK) activity but correlated with the higher activities of ATR and the higher activities of CHK1 in such cells. Not only caffeine, the nonspecific inhibitor of ATR, or UCN-01, the nonspecific inhibitor of CHK1, but also the specific CHK1 antisense oligonucleotide abolished the stronger inhibition of DNA replication in CPT-treated Ku80-/- cells. These results in aggregate indicated that the stronger S checkpoint in CPT-treated Ku80-/- cells is regulated through the highly activated ATR/CHK1 pathway.


Assuntos
Antígenos Nucleares , Camptotecina/farmacologia , Proteínas de Ciclo Celular/metabolismo , DNA Helicases , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Fase S/efeitos dos fármacos , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Quinase 1 do Ponto de Checagem , DNA/biossíntese , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Humanos , Autoantígeno Ku , Camundongos , Camundongos Knockout , Proteínas Nucleares/genética , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Inibidores de Proteínas Quinases , Proteínas Quinases/genética , Fase S/fisiologia
18.
Cancer Res ; 63(12): 3268-74, 2003 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12810658

RESUMO

Ku70 is one component of a protein complex, Ku70 and Ku80, that functions as a heterodimer to bind DNA double-strand breaks and activates DNA-dependent protein kinase. Our previous study with Ku70-/- and Ku80-/- mice, and cell lines has shown that Ku70- and Ku80-deficiency compromises the ability of cells to repair DNA double-strand breaks, increases radiosensitivity of cells, and enhances radiation-induced apoptosis. In this study, we examined the feasibility of using adenovirus-mediated, heat-activated expression of antisense Ku70 RNA as a gene therapy paradigm to sensitize cells and tumors to ionizing radiation. First, we performed experiments to test the heat inducibility of heat shock protein (hsp) 70 promoter and the efficiency of adenovirus-mediated gene transfer in rodent and human cells. Replication-defective adenovirus vectors were used to introduce a recombinant DNA construct, containing the enhanced green fluorescent protein (EGFP) under the control of an inducible hsp70 promoter, into exponentially growing cells. At 24 h after infection, cells were exposed to heat treatment, and heat-induced EGFP expression at different times was determined by flow cytometry. Our data clearly show that heat shock at 42 degrees C, 43 degrees C, or 44 degrees C appears to be equally effective in activating the hsp70 promoter-driven EGFP expression (>300-fold) in various tumor cells. Second, we have generated adenovirus vectors containing antisense Ku70 under the control of an inducible hsp70 promoter. Exponentially growing cells were infected with the adenovirus vector, heat shocked 24 h later, and the radiosensitivity determined 12 h after heat shock. Our data show that heat shock induces antisense Ku70 RNA, reduces the endogenous Ku70 level, and significantly increases the radiosensitivity of the cells. Third, we have performed studies to test whether Ku70 protein level can be down-regulated in a solid mouse tumor (FSa-II), and whether this results in enhanced radiosensitivity in vivo, as assessed by in vivo/in vitro colony formation and by the tumor growth delay. Our data demonstrate that heat-shock-induced expression of antisense Ku70 RNA attenuates Ku70 protein expression in FSa-II tumors, and significantly sensitizes the FSa-II tumors to ionizing radiation. Taken together, our results suggest that adenovirus-mediated, heat-activated antisense Ku70 expression may provide a novel approach to radiosensitize human tumors.


Assuntos
DNA Helicases , Proteínas de Ligação a DNA/antagonistas & inibidores , Fibrossarcoma/radioterapia , Terapia Genética/métodos , Vetores Genéticos/genética , Temperatura Alta , Mastadenovirus/genética , Proteínas de Neoplasias/antagonistas & inibidores , Oligodesoxirribonucleotídeos Antissenso/genética , Regiões Promotoras Genéticas , Tolerância a Radiação/fisiologia , Adenocarcinoma/patologia , Animais , Antígenos Nucleares/genética , Neoplasias Encefálicas/patologia , Carcinoma de Células Escamosas/patologia , Terapia Combinada , Proteína Quinase Ativada por DNA , Proteínas de Ligação a DNA/genética , Estudos de Viabilidade , Feminino , Fibrossarcoma/patologia , Fibrossarcoma/terapia , Raios gama , Regulação Neoplásica da Expressão Gênica , Genes Reporter , Glioma/patologia , Proteínas de Fluorescência Verde , Proteínas de Choque Térmico HSP70/genética , Humanos , Autoantígeno Ku , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C3H , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Proteínas Nucleares , Neoplasias da Próstata/patologia , Proteínas Serina-Treonina Quinases/fisiologia , Ratos , Transfecção , Células Tumorais Cultivadas/efeitos da radiação
19.
Oncogene ; 21(41): 6377-81, 2002 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12214278

RESUMO

Following exposure to genotoxic stress, proliferating cells actively slow down DNA replication through an S phase checkpoint to provide time for repair. The ATM-dependent pathway plays an important role in the S phase checkpoint response following ionizing irradiation. We report that there is a stronger S phase checkpoint response in irradiated Ku80(-/-) cells as compared with their wild-type counterparts, which has no relationship to DNA-dependent protein kinase (DNA-PK) activity but correlates with a higher ATM activity and with more ATM bound to chromatin DNA in such cells. Wortmannin, a nonspecific inhibitor of ATM, not only reduces the higher activity of ATM kinase, but also abolishes the stronger S phase checkpoint response in Ku80(-/-) cells. Furthermore, a specific ATM antisense oligonucleotide abolishes the stronger S checkpoint response in Ku80(-/-) cells and renders these cells practically indistinguishable from Ku80(+/+) cells for this endpoint. These results in aggregate indicate that the stronger S checkpoint in irradiated Ku80(-/-) cells is due to the higher ATM kinase activity.


Assuntos
Antígenos Nucleares , DNA Helicases , Proteínas de Ligação a DNA/fisiologia , Proteínas Nucleares/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Fase S , Fase S/fisiologia , Androstadienos/farmacologia , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Células CHO , Proteínas de Ciclo Celular , Cricetinae , Dano ao DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Deleção de Genes , Autoantígeno Ku , Proteínas Nucleares/genética , Fase S/efeitos dos fármacos , Fase S/efeitos da radiação , Proteínas Supressoras de Tumor , Wortmanina , Raios X
20.
DNA Repair (Amst) ; 2(2): 151-8, 2003 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-12531386

RESUMO

Ku is an abundant heterodimeric nuclear protein, consisting of 70- and 86-kDa tightly associated subunits that comprise the DNA binding component of DNA-dependent protein kinase. Poly(ADP-ribose) polymerase-1 (PARP-1) is a 113-kDa protein that catalyzes the synthesis of poly(ADP-ribose) on target proteins. Both Ku and PARP-1 recognize and bind to DNA ends. Ku functions in the non-homologous end joining (NHEJ) repair pathway whereas PARP-1 functions in the single strand break repair and base excision repair (BER) pathways. Recent studies have revealed that PARP-1 and Ku80 interact in vitro. To determine whether the association of PARP-1 and Ku80 has any physiological significance or synergistic function in vivo, mice lacking both PARP-1 and Ku80 were generated. The resulting offspring died during embryonic development displaying abnormalities around the gastrulation stage. In addition, PARP-1-/-/Ku80-/- cultured blastocysts had an increased level of apoptosis. These data suggest that the functions of both Ku80 and PARP-1 are essential for normal embryogenesis and that a loss of genomic integrity leading to cell death through apoptosis is likely the cause of the embryonic lethality observed in these mice.


Assuntos
Antígenos Nucleares/genética , DNA Helicases , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/metabolismo , Genes Letais , Poli(ADP-Ribose) Polimerases/genética , Animais , Antígenos Nucleares/metabolismo , Apoptose/genética , Apoptose/fisiologia , Reparo do DNA/genética , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Autoantígeno Ku , Camundongos , Poli(ADP-Ribose) Polimerases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA