Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Med Sci Monit ; 7(5): 861-8, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11535925

RESUMEN

BACKGROUND: Oxidative stress is a major causative agent of carcinogenesis, aging, and a number of diseases. 8-oxoG is the most stable and deleterious lesion of oxidative DNA damage. The 8-oxoG lesions can be eliminated by human repair systems consisting of three enzymes hMTH1, hOGG1, and hMYH homologous to E. coli MutT, MutM, and MutY proteins, respectively. MATERIAL AND METHODS: Human cells (P1, P2, and P3) resistant to H(2)O(2) were derived from the non-tumorigenic human breast cell line MCF10A by sequential treatment of the cells with H(2)O(2). The protein expression levels of DNA repair enzymes were analyzed by Western blotting. The DNA binding and glycosylase activities of hMYH and hOGG1 were measured in the extracts of the H(2)O(2) resistant cells. RESULTS: The H(2)O(2) resistant cells displayed tremendously greater anchorage-independent growth capability and higher expression of the anti-apoptotic protein BCL-2 than the parental cells. H(2)O(2) detoxification ability was elevated in P1 and P2 cells, but not in P3 cells, suggesting P3 cells might employ a different defense mechanism from P1 and P2 cells. In P3 cells, both hOGG1 and hMYH glycosylase activities were reduced but their protein levels increased. Two A/8-oxoG binding complexes were detected with cell extracts: the fast-migrating complex (bottom form) was dominated in MCF10A cells, and was greatly reduced in P3 cells. Interesting, the P3 cells showing the least amount of bottom form had the weakest hMYH glycosylase activity. CONCLUSIONS: Our results demonstrated, for the first time, that alteration of base excision repair pathways is correlated to cell resistance to oxidative stress.


Asunto(s)
Mama/efectos de los fármacos , Transformación Celular Neoplásica , ADN Glicosilasas , ADN Ligasas/metabolismo , Reparación del ADN , Peróxido de Hidrógeno/farmacología , N-Glicosil Hidrolasas/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Mama/citología , Mama/enzimología , Fraccionamiento Celular , Línea Celular , Proteínas de Unión al ADN/metabolismo , ADN-Formamidopirimidina Glicosilasa , Desoxiguanosina/análogos & derivados , Desoxiguanosina/genética , Desoxiguanosina/metabolismo , Femenino , Humanos , Oxidantes/farmacología , Estrés Oxidativo
3.
Redox Rep ; 6(6): 351-62, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11865975

RESUMEN

It has been hypothesized that programmed cell death is mediated, in part, through the formation of free radicals via oxidative pathways. Furthermore, it has been proposed that BCL-2 acts to inhibit cell death by interfering with the production of oxygen-derived free radicals induced by a wide variety of stimuli. In order to examine the antioxidant function of BCL-2, we transfected mouse epidermal cells JB6 clone 41 with the expression vector pD5-Neo-BCL-2 and studied the effect of BCL-2 overexpression on oxidant-induced cell death and on the production of reactive oxygen species. Compared to Neo control cells, BCL-2-expressing cells are more resistant to the killing and growth retardation induced by hydrogen peroxide, superoxide, or by the oxygen radical-generating quinone-containing compounds menadione, diaziquone and adriamycin. The latter compounds generate reactive oxygen species during bioreductive metabolism. In addition, the exposed cells die by necrosis rather than apoptosis. Hydroxyl radical levels generated by the quinone-containing agents were low in BCL-2-expressing JB6 cells compared to control Neo cells. BCL-2, however, does not change the activities of the major cellular antioxidant enzymes superoxide dismutase, catalase or glutathione peroxidase. On the other hand, the glutathione concentrations increased in BCL-2 overexpressing cells after oxidative challenge, while the opposite was true for control cells. Thus, our results suggest that BCL-2 inhibition of oxidant-induced cell death is mediated, at least in part, through an antioxidant pathway, and that this pathway involves glutathione.


Asunto(s)
Oxidantes/metabolismo , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Animales , Antifibrinolíticos/farmacología , Antineoplásicos/farmacología , Antioxidantes/farmacología , Apoptosis , Western Blotting , Caspasa 3 , Caspasas/metabolismo , Muerte Celular , Supervivencia Celular , Células Cultivadas , Relación Dosis-Respuesta a Droga , Doxorrubicina/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Glutatión/metabolismo , Humanos , Células Jurkat , Ratones , Necrosis , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno , Superóxidos/metabolismo , Transfección , Vitamina K 3/farmacología
4.
Free Radic Biol Med ; 29(3-4): 263-75, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11035255

RESUMEN

Bioactivation of quinone-containing anticancer agents has been studied extensively within the context of the chemistry and structure of the individual quinones which may result in various mechanisms of bioactivation and activity. In this review we focus on the two electron enzymatic reduction/activation of quinone-containing anticancer agents by DT Diaphorase (DTD). This enzyme has become important in oncopharmacology because its activity varies with tissues and it has been found to be elevated in tumors. Thus, a selective tumor cell kill can exist for agents that are good substrates for this enzyme. In addition, the enzyme can be induced by a variety of agents, a fact that can be used in chemotherapy. That is induction by a nontoxic agent followed by treatment with a good DT-Diaphorase substrate. A wide variety of anticancer drugs are discussed some of which are not good substrates such as Adriamycin, and some of which are excellent substrates. The latter category includes a variety of quinone containing alkylating agents.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Quinonas/análisis , Animales , Antineoplásicos/metabolismo , Inducción Enzimática , Radicales Libres/metabolismo , Humanos , Lactante , Oxidación-Reducción , Quinonas/metabolismo , Células Tumorales Cultivadas
5.
Free Radic Biol Med ; 29(6): 548-56, 2000 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-11025198

RESUMEN

Quantifying oxygen radicals that arise during the redox cycling of quinone-containing anticancer agents such as diaziquone (AZQ) has been difficult, as has been their detection at low drug concentrations. This is due to the fact that EPR spin trapping, the method most often used for *OH detection, requires the use of high drug concentrations. Using a new highly sensitive technique that employs a fluorescamine-derivatized nitroxide, we show that low levels of NADPH-cytochrome P450 reductase (4.25 microg/ml) catalyze the production of hydroxyl radicals at very low, clinically relevant AZQ concentrations. Thus, at this enzyme concentration, we were able to detect a rate of 0.10 nM s(-1) hydroxyl radical production by 5 microM AZQ, a clinically relevant concentration. The Michaelis-Menten constants for AZQ-mediated hydroxyl radical production are: K(M) = 10.7 +/- 1.4 microM, and V(max) = 5.2 +/- 0.9 x 10(-8) M s(-1) (mg protein)(-1). Experiments employing catalase, superoxide dismutase, and NADPH-cytochrome P450 reductase, confirm the previously deduced conclusions from high drug concentrations, that is, that at low concentrations, AZQ acts to shuttle reducing equivalents from the enzyme to oxygen, thus generating the redox cycle. The data presented here suggest that the levels and locations of redox active metal ions may be the principal controlling factor in the pathway of AZQ activity that involves oxidative stress.


Asunto(s)
Antineoplásicos/metabolismo , Aziridinas/metabolismo , Benzoquinonas/metabolismo , Radical Hidroxilo/metabolismo , Animales , Antineoplásicos/química , Aziridinas/química , Benzoquinonas/química , Cromatografía Líquida de Alta Presión , Dimetilsulfóxido/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Radicales Libres/química , Radicales Libres/metabolismo , Radical Hidroxilo/análisis , Hierro/metabolismo , Cinética , Hígado/enzimología , Estructura Molecular , NADPH-Ferrihemoproteína Reductasa/metabolismo , Oxidación-Reducción , Oxígeno/metabolismo , Ratas , Superóxido Dismutasa/metabolismo
6.
Front Biosci ; 5: D629-38, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10877994

RESUMEN

The metabolism of quinone-containing antitumor agents involves enzymatic reduction of the quinone by one or two electrons. This reduction results in the formation of the semiquinone or the hydroquinone of the anticancer drug. The consequence of these enzymatic reductions is that the semiquinone yields its extra electron to oxygen with the formation of superoxide radical anion and the original quinone. This reduction by a reductase followed by oxidation by molecular oxygen (dioxygen) is known as redox-cycling and continues until the system becomes anaerobic. In the case of a two electron reduction, the hydroquinone could become stable, and as such, excreted by the organism in a detoxification pathway. In some cases such as aziridine quinones, the hydroquinone can be oxidized by one electron at a time resulting in the production of superoxide, the semiquinone and the parental quinone. Quinone anticancer agents upon reduction can also set up an equilibrium between the hydroquinone, the parental quinone and the semiquinone which results in a long-lived semiquinone. Depending on the compound, aziridine quinones, for example, this equilibrium is long lasting thus allowing for the detection of the semiquinone under aerobic conditions. This phenomenon is known as comproportionation-disporportionation equilibrium. The series of reviews in this Special Issue address the consequences of bioreduction of quinone alkylators used in the treatment of cancer. In this particular review we are interested in describing the phenomenon of redox-cycling, how it is measured, and the biological consequences of the presence of the semiquinone and the oxygen radicals generated.


Asunto(s)
Alquilantes/metabolismo , Radicales Libres/análisis , NADP/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Quinonas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Chem Res Toxicol ; 12(11): 1042-9, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10563829

RESUMEN

The effect of quinone anti-cancer compounds, 3,6-diaziridinyl-2, 5-bis(carboethoxyamino)-1,4-benzoquinone (diaziquone or AZQ) and 3, 6-dihydro-2,5-bis(aziridinyl)-1,4-benzoquinone (DZQ), on the production of hydroxyl radical ((*)OH) in a series of mouse epidermal cell lines was examined using a new, highly sensitive method that employs a fluorescamine-derivatized nitroxide probe. Cell lines that were examined included the mouse epidermal cell line, JB6 clone 41, and JB6 cells transfected with the human Cu-Zn superoxide dismutase (SOD) genes (SOD3 and SOD15) and human catalase (CAT) genes (CAT13 and CAT10). Bioreduction of the nitroxide probe by these cell lines was insignificant at the cell densities employed in these experiments, and thus did not interfere with the (*)OH measurements. In the presence of low concentrations of AZQ or DZQ (20-200 microM), (*)OH production rates were highest in the JB6 cells, intermediate in the SOD-transfected cells, and lowest in the CAT-transfected cells, illustrating that both superoxide and hydrogen peroxide are involved in the production of (*)OH in these systems. Further experiments in which the addition of exogenous SOD and CAT was employed, as well as measurements of probe incorporation into the cells, indicated that this probe can cross cell membranes and detect (*)OH generated intracellularly. In the presence of 100 microM diethylenetriaminepentaacetic acid (DTPA), the rate of (*)OH production in the presence of 100 microM AZQ is approximately 4.7 x 10(4) molecules s(-)(1) cell(-)(1); as much as 45% of this production appears to originate within the cells. This new method should be broadly applicable to the rapid screening of compounds or treatments thought to induce oxidative stress in mammalian cells.


Asunto(s)
Antineoplásicos/farmacología , Epidermis/metabolismo , Quinonas/farmacología , Animales , Catalasa/genética , Catalasa/metabolismo , Línea Celular , Células Epidérmicas , Espacio Extracelular/metabolismo , Humanos , Radical Hidroxilo , Cinética , Ratones , Oxidación-Reducción , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Transfección
9.
Mol Cell Biol ; 18(11): 6493-504, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9774665

RESUMEN

Interferons (IFNs) and retinoids are potent biological response modifiers. By using JAK-STAT pathways, IFNs regulate the expression of genes involved in antiviral, antitumor, and immunomodulatory actions. Retinoids exert their cell growth-regulatory effects via nuclear receptors, which also function as transcription factors. Although these ligands act through distinct mechanisms, several studies have shown that the combination of IFNs and retinoids synergistically inhibits cell growth. We have previously reported that IFN-beta-all-trans-retinoic acid (RA) combination is a more potent growth suppressor of human tumor xenografts in vivo than either agent alone. Furthermore, the IFN-RA combination causes cell death in several tumor cell lines in vitro. However, the molecular basis for these growth-suppressive actions is unknown. It has been suggested that certain gene products, which mediate the antiviral actions of IFNs, are also responsible for the antitumor actions of the IFN-RA combination. However, we did not find a correlation between their activities and cell death. Therefore, we have used an antisense knockout approach to directly identify the gene products that mediate cell death and have isolated several genes associated with retinoid-IFN-induced mortality (GRIM). In this investigation, we characterized one of the GRIM cDNAs, GRIM-12. Sequence analysis suggests that the GRIM-12 product is identical to human thioredoxin reductase (TR). TR is posttranscriptionally induced by the IFN-RA combination in human breast carcinoma cells. Overexpression of GRIM-12 causes a small amount of cell death and further enhances the susceptibility of cells to IFN-RA-induced death. Dominant negative inhibitors directed against TR inhibit its cell death-inducing functions. Interference with TR enzymatic activity led to growth promotion in the presence of the IFN-RA combination. Thus, these studies identify a novel function for TR in cell growth regulation.


Asunto(s)
Apoptosis/efectos de los fármacos , Interferones/farmacología , Reductasa de Tiorredoxina-Disulfuro/fisiología , Tretinoina/farmacología , Secuencia de Aminoácidos , Neoplasias de la Mama/enzimología , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/química , Oligonucleótidos Antisentido/farmacología , Análisis de Secuencia , Células Tumorales Cultivadas
10.
Chem Res Toxicol ; 11(4): 360-8, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9548807

RESUMEN

The biologic functions attributed to the nucleophosphoprotein p53 have been increasing in recent years. Some studies suggested that wild type p53 is responsible for cell cycle arrest brought about as a response to exposure of mammalian cells to DNA-damaging agents. This cell cycle arrest occurs in order for cells to repair the damaged macromolecules. Extensively damaged cells are also thought to undergo apoptosis via the p53-dependent or -independent signal transduction pathways. In this study, we investigated the ability of diaziridinylbenzoquinones to increase p53 levels in the human breast cancer cell line MCF-7. Diaziquone (AZQ), an anticancer agent, and its derivatives, diaziridinequinone (DZQ) and methyldiaziridinequinone (MeDZQ), induced p53 in a dose- and time-dependent manner as measured by the electrophoretic mobility shift assay. Wild type p53 induction by AZQ was suppressed when DT-diaphorase activity was inhibited by pretreating the cells with dicumarol. Aside from their potent alkylating activity, these agents also undergo redox cycling as evidenced by oxygen consumption and the production of reactive oxygen species (ROS). Inhibition of ROS production by the antioxidant enzyme catalase reduced AZQ- and DZQ-mediated p53 induction by about 45%. Thiotepa, a non-quinone aziridine-containing agent, and 1,4-benzoquinone (p-BQ), a redox cycling quinone, increased p53 levels. The nonalkylator oxygen-radical-generating agent menadione (MD) caused p53 induction only when MCF-7 cells were allowed to recover in drug-free media. On the basis of these data, we propose that the bioreductive activation of AZQ is a prerequisite for p53 induction. Moreover, the induction of p53 by AZQ requires both the quinone and the aziridine moieties of the AZQ molecule. Although AZQ and its analogues increased p53 levels in MCF-7 cells, p53 induction in these cells may not be responsible for the apoptosis seen upon treatment of MCF-7 cells with these agents. The uncoupling of p53 induction and apoptosis is evidenced by the generation of nucleosomal DNA laddering in aziridinequinone-treated T47D cells, a breast cancer cell line bearing a p53 mutation.


Asunto(s)
Antineoplásicos/farmacología , Aziridinas/farmacología , Benzoquinonas/farmacología , Proteína p53 Supresora de Tumor/biosíntesis , Adenocarcinoma , Neoplasias de la Mama , Humanos , Células Tumorales Cultivadas/efectos de los fármacos
11.
Anal Chem ; 69(21): 4295-302, 1997 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9360488

RESUMEN

A simple and highly sensitive method to quantify the rates of production of OH in biological systems is described. This method employs the reaction between OH and dimethyl sulfoxide to generate quantitatively a methyl radical, which then reacts with a fluorescamine-derivatized nitroxide to produce the stable O-methylhydroxylamine. This O-methylhydroxylamine is separated by reversed-phase high-performance liquid chromatography and quantified fluorometrically. The estimated detection limit of the O-methylhydroxylamine is 3.5 nM for a 50 microL injection at a signal to noise ratio of 2. The method is applied to the determination of the rates of OH production in a biologically relevant model system and in a mouse epidermal cell line treated with a quinone anticancer compound.


Asunto(s)
Radical Hidroxilo/análisis , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Aziridinas/química , Aziridinas/farmacología , Benzoquinonas/química , Benzoquinonas/farmacología , Línea Celular , Ratones , Estructura Molecular
12.
Radiat Res ; 145(1): 98-101, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8532844

RESUMEN

We compared the ability of continuous-wave ultrasound to enhance cytotoxicity from X irradiation, hyperthermia or exposure to adriamycin. The survival of CHO cells exposed in culture medium to these agents was determined with and without continuous-wave ultrasound (1.62 or 1.765 MHz). In water-filled transmission exposure vessels with 2-cm-diameter Mylar end windows, 10-min insonation not producing cytotoxicity could produce .OH radicals (measured by electron paramagnetic resonance) even at 0.4 W/cm2. Ultrasound at intensities ranging between 1 and 2.5 W/cm2 increased the clonogenic cytotoxicity of adriamycin (P = 0.0023 by paired t test) but not of X rays (2-10 Gy) or hyperthermia (44 degrees C for 10-50 min). The only significant action of continuous-wave ultrasound under similar test conditions was the potentiation of adriamycin-induced clonogenic cytotoxicity, possibly mediated by cavitational activity.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Doxorrubicina/toxicidad , Calor , Ultrasonografía , Animales , Células CHO , Supervivencia Celular/fisiología , Cricetinae , Relación Dosis-Respuesta en la Radiación , Espectroscopía de Resonancia por Spin del Electrón , Radical Hidroxilo/análisis , Radical Hidroxilo/metabolismo , Hipertermia Inducida , Cinética , Rayos X
13.
Ultrasound Med Biol ; 22(3): 355-62, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8783468

RESUMEN

We investigated in vitro enhancement of cytotoxicity of chemotherapeutic agents by tone-burst ultrasound. Survival of CHO cell exposed to chemotherapeutic agents in culture medium was determined with and without insonation (1.62 and 0.29 MHz, 10% duty cycle). Insonations up to 0.4 MPa peak pressure (5 kW/m2 spatial and temporal average) occurred in the middle of 1 h drug exposures. Cytotoxicity in ultrasound control groups was never observed. Ultrasound increased the clonogenic cytotoxicity of adriamycin (p = 0.00027 by paired t test) and diaziquone but not of cisplatin or mitomycin C. Potentiation of adriamycin depended on exposure time and tone-burst frequency. .OH production in water occurred at intensities as low as 0.4 kW/m2, but did not increase with added adriamycin. Ultrasound did not affect membrane fluidity, but moderately increased cellular adriamycin accumulation, possibly explaining the observed drug potentiation.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos de la radiación , Células CHO/efectos de los fármacos , Ultrasonografía , Animales , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Aziridinas/farmacología , Benzoquinonas/farmacología , Células CHO/diagnóstico por imagen , Células CHO/metabolismo , Técnicas de Cultivo de Célula , Supervivencia Celular , Cisplatino/farmacología , Cricetinae , Doxorrubicina/farmacología , Sinergismo Farmacológico , Citometría de Flujo , Radical Hidroxilo/metabolismo , Fluidez de la Membrana , Mitomicina/farmacología
14.
Chem Res Toxicol ; 8(3): 455-64, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7578933

RESUMEN

The antitumor agent diaziquone (AZQ) reacts with reduced glutathione (GSH) in aqueous solutions and under aerobic conditions to give rise to the glutathionyl and hydroxyl free radicals, as well as the AZQ semiquinone. Under anaerobic conditions, the only radical observed was the glutathionyl radical. These radicals are quickly abrogated when superoxide dismutase and catalase are coincubated. Separately, superoxide dismutase favors the formation of thiyl radicals while catalase favors the formation of hydroxyl radicals and AZQ semiquinone. The metal chelator diethylenetriaminepentaacetic acid favors the production of hydroxyl radicals and AZQ semiquinone. The reaction of AZQ with GSH at pH 7.2 and 5.5 results in a variety of conjugates. These conjugates include addition of glutathione to both aziridines, displacement of the aziridines by GSH, and a combination of both. The majority of the conjugates are formed by nucleophilic attack of GSH to the AZQ aziridines or by 1,4-Michael addition to the AZQ quinone or a combination of both. There may be a small free radical component in conjugate formation, but the majority of the free radicals observed are from redox reactions that involve the oxidation of glutathione and the reduction and autoxidation of AZQ to produce oxygen radicals and hydrogen peroxide, a process that is enhanced by trace metal ions.


Asunto(s)
Antineoplásicos/química , Aziridinas/química , Benzoquinonas/química , Glutatión/química , Antioxidantes/farmacología , Catalasa/farmacología , Interacciones Farmacológicas , Espectroscopía de Resonancia por Spin del Electrón , Radicales Libres/química , Consumo de Oxígeno , Superóxido Dismutasa/farmacología
15.
Cancer Chemother Pharmacol ; 35(5): 377-86, 1995.
Artículo en Inglés | MEDLINE | ID: mdl-7850918

RESUMEN

PURPOSE: It has been recognized that enhanced antioxidant defenses can contribute to the resistance of cancer cells displaying multidrug resistance (MDR) that arises in conjunction with the overexpression of P-glycoprotein (Pgp). The purpose of this study was to determine if the defenses against oxidant stress in MDR human leukemia cells (HL-60/AR) that overexpress multidrug-resistance-associated protein (MRP), but not Pgp, contribute to the mechanism of drug resistance in this cell line. METHODS: HL-60/AR cells were evaluated in comparison with wild-type cells with respect to sensitivity to the oxidants hydrogen peroxide (H2O2) and tert-butyl hydroperoxide (t-BuOOH), the activities and amounts of the antioxidant enzymes catalase and glutathione peroxidase (GSH-Px), and the effects that manipulation of the activities of these enzymes may have on cellular sensitivity to the oxidants and to daunorubicin. We also evaluated the ability of the cells to generate daunorubicin semiquinone free radical as measured by electron spin resonance (ESR) spectroscopy. RESULTS: HL-60/AR cells were > 10-fold resistant to the cytotoxic effects of the H2O2 or t-BuOOH as compared with parental, drug-sensitive HL-60 cells. This phenomenon could be attributed largely to elevated activity and protein levels of catalase in HL-60/AR cells. Furthermore, inhibition of catalase by 3-amino-1,2,4-triazole (AT) diminished the resistance of HL-60/AR to these oxidants by > 80% or > 50%, respectively. Despite these findings, AT was incapable of causing sensitization of HL-60/AR cells to the cytotoxic effects of daunorubicin. We found that the activity and amount of selenium-dependent glutathione peroxidase (GSH-Px) was no greater in HL-60/AR cells than in HL-60 cells. Cultivation of cells in selenium-deficient medium caused a marked reduction in GSH-Px activity in HL-60/AR cells and a profound inhibition of GSH-redox cycling manifested by a decrease in baseline hexose monophosphate shunt activity (HMPS) and markedly blunted stimulation of the HMPS by the oxidant t-BuOOH in both wild-type and resistant cells. These variations in GSH-Px activity and GSH-redox cycling, however, were not associated with an alteration in cellular sensitivity to daunorubicin. The failure of catalase inhibition or selenium manipulation of GSH-Px activity to affect daunorubicin cytotoxicity was not due to the inability of these cells to produce free-radical species of daunorubicin, since ESR studies revealed that the generation of daunorubicin semiquinone free radical by HL-60/AR cells was equal to and, in fact, 3-fold that obtained with HL-60 cells. CONCLUSIONS: In comparison with parental HL-60 cells, MRP-overexpressing HL-60/AR cells have demonstrable alterations in antioxidant defenses that are manifested by cellular resistance to the cytotoxic effects of H2O2 and t-BuOOH and by elevated protein levels and activity of catalase. Whether these alterations are epiphenomena or are related to overexpression of MRP remains to be determined. However, it does appear that the enhanced antioxidant defenses observed in HL-60/AR cells do not contribute to the resistance to daunorubicin manifested by this cell line. Although HL-60/AR cells generate daunorubicin semiquinone free radical to an extent equal to or greater than that observed in HL-60 cells, the failure of alterations in GSH-Px activity or inhibition of catalase to change the sensitivity of HL-60/AR cells to daunorubicin suggests that the cytotoxicity of daunorubicin in these cells in not mediated through H2O2 or other peroxide species detoxified by these enzymes.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Catalasa/metabolismo , Daunorrubicina/toxicidad , Oxidantes/metabolismo , Western Blotting , Catalasa/antagonistas & inhibidores , Catalasa/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Simulación por Computador , Daunorrubicina/metabolismo , Resistencia a Múltiples Medicamentos , Espectroscopía de Resonancia por Spin del Electrón , Glutatión Peroxidasa/antagonistas & inhibidores , Glutatión Peroxidasa/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/toxicidad , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Oxidantes/toxicidad , Vía de Pentosa Fosfato/efectos de los fármacos , Vía de Pentosa Fosfato/fisiología , Peróxidos/metabolismo , Peróxidos/toxicidad , Especies Reactivas de Oxígeno , Células Tumorales Cultivadas , terc-Butilhidroperóxido
16.
Free Radic Biol Med ; 17(5): 379-88, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7835744

RESUMEN

Nitroxides are cell-permeable, stable radicals that react readily with paramagnetic species such as transition metals or short-lived free radicals, though not generally with diamagnetic molecules. Nitroxides can undergo one-electron selective redox reactions and thereby potentially modify the activity of cytotoxic drugs. Streptonigrin (SN) toxicity requires bioreduction to yield the semiquinone radical, and the toxicity is reportedly mediated by transition metals and oxygen-derived reactive species via redox-cycling of the semiquinone intermediate. The present study shows that (1) nitroxides protected isolated DNA and also aerated or hypoxic bacterial cells from SN toxicity; (2) H2O2 potentiated the hypoxic cytotoxicity of the drug but inhibited the damage to aerated cells; (3) pretreatment of cells with H2O2 conferred some protection, but not when the drug alone was preexposed to H2O2; and (4) desferrioxamine and 2,2-dipyridyl, though neither diethylenetriamino pentaacetate, exogenous catalase, or superoxide dismutase, decreased SN-induced cell killing. The mechanisms by which nitroxides protect from SN toxicity involve both a selective radical-radical reaction with SN semiquinone and the reoxidation of reduced cellular transition metal ions. On the other hand, H2O2 appears to exert two opposing effects: (1) facilitation of cell killing by the Fenton reaction and (2) lowering the cellular level of reducing equivalents, thus inhibiting the bioreductive activation of SN.


Asunto(s)
Óxidos N-Cíclicos/farmacología , Daño del ADN , Escherichia coli/efectos de los fármacos , Estreptonigrina/toxicidad , Superóxido Dismutasa , Aerobiosis , Anaerobiosis , Espectroscopía de Resonancia por Spin del Electrón , Escherichia coli/crecimiento & desarrollo , Radicales Libres , Peróxido de Hidrógeno/toxicidad , Radical Hidroxilo/análisis , Cinética , Marcadores de Spin , Superóxidos/análisis
17.
Chem Res Toxicol ; 7(1): 23-8, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-8155821

RESUMEN

The o-quinone forms of 2,3- and 3,4-catechol estrogens have been implicated in the carcinogenicity of these hormones. The concomitant production of reactive oxygen species during reduction of the o-quinone estrogens has been inferred to play a mechanistic role in their mutagenic potential. Conclusive evidence documenting the production of hydrogen peroxide, the hydroxyl radical, and the estrone 3,4-semiquinone in estrone 3,4-quinone (3,4-EQ)-treated human breast cancer subcellular fractions was demonstrated in the absence of exogenously added catalysts. Subcellular fractions of MCF-7 cells treated with 3,4-EQ and NADPH, including nuclei, mitochondria, and microsomes, were shown to support significant amounts of hydrogen peroxide production. Hydrogen peroxide production in 3,4-EQ-treated cellular fractions and the chromosomal DNA damage induced in 3,4-EQ-treated MCF-7 cells were abolished by the addition of catalase. A significant and potentially physiologically relevant spontaneous reduction of 3,4-EQ by NADPH resulting in hydrogen peroxide production was demonstrated. The results unequivocally demonstrate that free radicals are produced during the metabolism of estrone 3,4-quinone in human cells.


Asunto(s)
Neoplasias de la Mama/genética , Daño del ADN , ADN de Neoplasias/efectos de los fármacos , Estrenos/toxicidad , Estrógenos/toxicidad , Neoplasias de la Mama/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Estrenos/metabolismo , Estrógenos/metabolismo , Radicales Libres , Humanos , Peróxido de Hidrógeno/análisis , Quinonas/metabolismo , Especies Reactivas de Oxígeno , Fracciones Subcelulares/efectos de los fármacos , Células Tumorales Cultivadas
18.
Chem Biol Interact ; 88(2-3): 137-53, 1993 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8403076

RESUMEN

Electron paramagnetic resonance (EPR/ESR) spin trapping studies with DMPO revealed that purified rat liver NAD(P)H (quinone-acceptor) oxidoreductase (QAO) mediated hydroxyl radical formation by a diverse range of quinone-based antitumour agents. However, when MCF-7 S9 cell fraction was the source of QAO, EPR studies distinguished four different interactions by these agents and QAO with respect to hydroxyl radical formation: (i) hydroxyl radical formation by diaziquone (AZQ), menadione, 1AQ; 1,5AQ and 1,8AQ was mediated entirely or partially by QAO in MCF-7 S9 fraction; (ii) hydroxyl radical formation by daunorubicin and Adriamycin was not mediated by QAO in MCF-7 S9 fraction; (iii) hydroxyl radical formation by mitomycin C was stimulated in MCF-7 S9 fraction when QAO was inhibited by dicumarol; (iv) no hydroxyl radical formation was detected for 1,4AQ or mitoxantrone in MCF-7 S9 fraction. This study shows that purified rat liver QAO can mediate hydroxyl radical formation by a variety of diverse quinone antitumour agents. However, QAO did not necessarily contribute to hydroxyl radical formation by these agents in MCF-7 S9 fraction and in the case of mitomycin C, QAO played a protective role against hydroxyl radical formation.


Asunto(s)
Antineoplásicos/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/fisiología , Quinonas/metabolismo , Animales , Espectroscopía de Resonancia por Spin del Electrón , Radicales Libres , Ratas , Fracciones Subcelulares , Especificidad por Sustrato , Células Tumorales Cultivadas
19.
Infect Immun ; 60(12): 5113-8, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1333449

RESUMEN

Human endothelial cells infected with Rickettsia rickettsii, the etiological agent of Rocky Mountain spotted fever, undergo striking morphological changes to the endoplasmic reticulum-outer nuclear envelope complex. These changes are accompanied by concurrent accumulation of intracellular peroxides. Both of these findings are consistent with the notion that cells undergo some form of oxidative stress. Since oxidant injury is often initiated or mediated through oxygen radicals, we examined superoxide radical generation when endothelial cells were exposed to R. rickettsii. We also examined the levels of superoxide dismutase, an enzyme induced in response to increased superoxide formation. The levels of both superoxide and superoxide dismutase increased when endothelial cells were exposed to R. rickettsii. These results, together with our previous findings, support our hypothesis that cells infected by this intracellular bacterium experience oxidant-mediated injury that may eventually contribute to cell death.


Asunto(s)
Endotelio Vascular/metabolismo , Rickettsia rickettsii/fisiología , Superóxido Dismutasa/biosíntesis , Superóxidos/metabolismo , Células Cultivadas , Inducción Enzimática , Radicales Libres , Humanos , Oxidación-Reducción
20.
Biochem Pharmacol ; 44(8): 1625-35, 1992 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-1301071

RESUMEN

The alkylating activity of reduced diaziquone was studied by the nitrobenzylpyridine (NBP) assay and was compared to those of the parent compound and aziridine-containing N,N',N"-triethylenethiophosphoramide (Thio-TEPA). Diaziquone (AZQ) was reduced enzymatically by 2e- using S9 cell fraction from MCF-7 cells which is rich in NAA(P)H:quinone-acceptor oxidoreductase (DT-diaphorase) (QAO) activity. One electron enzymatic reduction was performed with NADPH-cytochrome c reductase. The alkylating activity of AZQ increased 3-fold when reduced by 2e-. This increase was inhibited by dicumarol, an inhibitor of QAO. In contrast, the alkylating activity of AZQ did not increase beyond that of the parent compound when reduced by 1e- using purified NADPH-cytochrome c reductase. Similar results were obtained when AZQ was reduced chemically with borohydride (2e-) and with NADPH (1e-). Anaerobic incubations of AZQ with the S9 fraction of MCF-7 cells (2e- reduction) resulted in an increase in NBP alkylation over its aerobic counterpart (1.8-fold) while maintaining the near 3-fold increase in alkylation over untreated AZQ. In contrast, AZQ incubations with NADPH-cytochrome c reductase (1e- reduction) under the same conditions did not result in an NBP alkylation increase over untreated AZQ. These results indicate that AZQ hydroquinone is most likely the responsible species for the observed alkylation of this antitumor agent to DNA and other nucleophiles. The results also suggest that NAD(P)H:quinone-acceptor oxidoreductase is a very important enzyme in the bioactivation of AZQ.


Asunto(s)
Aziridinas/metabolismo , Benzoquinonas/metabolismo , Hígado/metabolismo , Alquilación , Animales , Aziridinas/química , Aziridinas/farmacología , Benzoquinonas/química , Benzoquinonas/farmacología , Biotransformación , Línea Celular , Óxidos N-Cíclicos , Humanos , NADH Deshidrogenasa/metabolismo , NADP/metabolismo , Oxidación-Reducción , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA