Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Science ; 227(4691): 1231-3, 1985 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-3975611

RESUMEN

In a study of the relation between chronic inflammation and carcinogenesis, C3H mouse fibroblasts of the 10T 1/2 clone 8 line (10T 1/2 cells) were exposed to human neutrophils stimulated to synthesize reactive oxygen intermediates or to a cell-free enzymatic system generating superoxide (xanthine oxidase plus hypoxanthine). After exposure, the 10T 1/2 cells were either placed in tissue culture or immediately injected into athymic nude mice. Both malignant and benign tumors developed in the mice injected with treated cells, but not in those injected with control cells; in one instance cells grown from one of the benign tumors subsequently developed a malignant phenotype. Malignant transformation was also observed in treated cells in the experiments in vitro.


Asunto(s)
Transformación Celular Neoplásica/inmunología , Neutrófilos/fisiología , Fagocitos/fisiología , Animales , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Desnudos , Neutrófilos/metabolismo , Oxidación-Reducción/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología
2.
J Clin Invest ; 75(6): 1835-41, 1985 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-3924956

RESUMEN

Stimulated human phagocytes produce sister chromatid exchanges in cultured mammalian cells by a mechanism involving oxygen metabolites. Experiments were designed to determine whether antioxidants inhibit this process. Superoxide dismutase, catalase, and hydroxyl radical scavengers (benzoate, mannitol) protected target Chinese hamster ovary cells from phagocyte-induced sister chromatid exchanges, implicating the involvement of hydroxyl radicals in this chromosomal damage. N-acetylcysteine and beta-carotene were also protective. alpha-Tocopherol (greater than 5 microM) protected target cells exposed to phagocytes but not to enzymatically generated oxidants when the vitamin was added just before the source of oxygen radicals, suggesting, as reported by others, that the principal action of tocopherol in this setting was to inhibit the release of oxidants from phagocytes. On the other hand, cultivation of target cells with supplemental tocopherol protected them from the toxic effects of the enzymatic oxidant-producing system, indicating a role for membrane-associated free radicals in the mechanism of sister chromatid exchange induction. Low concentrations of sodium selenite (0.1-1.0 microM) protected the target cells. However, higher concentrations (10 microM) of selenite had no effect on oxidant-induced sister chromatid exchange formation, and 0.1 mM selenite increased the number of exchanges. Sodium selenite concentrations of 0.1 mM also decreased the intracellular glutathione concentration of target cells during an oxidant stress, and reducing target cell glutathione concentrations with buthionine sulfoximine increased their sensitivity to oxygen-related chromosomal damage. Therefore, the potentiation of oxygen radical-induced chromosomal damage observed with high concentrations of selenite may result from a decrease in the thiol antioxidant defense systems within the cell. The findings suggest that the hydroxyl radical has an important role in the production of phagocyte-induced cytogenetic injury, membrane-derived intermediates may be involved, depletion of intracellular glutathione renders cells more susceptible to this injury, and supplementation of target cells with antioxidants can protect them from oxygen radical-generated chromosomal injury.


Asunto(s)
Oxígeno/toxicidad , Intercambio de Cromátides Hermanas/efectos de los fármacos , Acetilcisteína/farmacología , Animales , Benzoatos/farmacología , Ácido Benzoico , Carotenoides/farmacología , Catalasa/metabolismo , Cricetinae , Cricetulus , Femenino , Radicales Libres , Glutatión/metabolismo , Humanos , Manitol/farmacología , Ovario , Oxígeno/antagonistas & inhibidores , Fagocitos/fisiología , Ácido Selenioso , Selenio/farmacología , Superóxido Dismutasa/metabolismo , Vitamina E/farmacología , Xantina Oxidasa , beta Caroteno
3.
Mol Cell Biol ; 13(7): 4416-22, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8321241

RESUMEN

Alterations in intracellular glutathione (GSH) content are known to affect intrinsic responses to ionizing radiation. More recently, it became apparent that radiation responses may depend also on the expression of specific oncogenes, including ras. These findings, suggesting a possible link between GSH and ras, led us to examine the effect of various GSH modulators on ras expression. Treatment of c-Ha-ras-transformed NIH 3T3 cells with L-buthionine S'R'-sulfoximine, dimethylfumarate, or N',N'-1,3-bis(trans-4-hydroxycyclohexyl)-N'-nitrosourea resulted in dose- and time-dependent reduction in ras mRNA steady-state levels followed by a decrease in ras-encoded p21 protein production. The effect on ras correlated with the extent of GSH decline, was common to different members of the ras family, and was independent of the mode of oncogene activation or cell phenotype. Indeed, similar drug effects were observed with murine cells in which overexpression of the c-Ha-ras proto-oncogene was due to transcriptional activation (PR4, nontumorigenic) or gene amplification (NIH 136, tumorigenic) and with malignant cells expressing a mutated Ha-ras (RS504). Moreover, N-ras, EJras, and Ki-ras in human tumor cells were similarly affected. Molecular analysis revealed a significant decrease in ras mRNA half-life in cells subjected to GSH inhibition, an effect that required de novo protein synthesis, but there was no change in the rate of gene transcription. These results indicate that pharmacological manipulation of cellular GSH content can down-regulate ras expression at the posttranscriptional level by destabilizing ras transcripts. The potential clinical implications are discussed.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genes ras , Glutatión/metabolismo , Procesamiento Postranscripcional del ARN , Células 3T3 , Animales , Northern Blotting , Butionina Sulfoximina , Ciclo Celular , Cicloheximida/farmacología , Regulación hacia Abajo , Humanos , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Ratones , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Proto-Oncogenes Mas , Transcripción Genética , Células Tumorales Cultivadas
4.
Int J Radiat Oncol Biol Phys ; 12(7): 1121-6, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3528088

RESUMEN

There is considerable interest in the role of thiols and redox enzymes as modulators of chemo- and radioresponses from both therapeutic and theoretical perspectives. The metabolism of GSH and the functioning of the GSH redox cycle can have a significant impact on the response of cells to both chemical and radiation stresses. From this rapidly developing research area, attention has been focused on recent progress in several areas: the use of GSH-depletion to reverse chemo-induced resistance to further chemical treatment; the significance of GSH transferase activity in chemoresistant cells; the effectiveness of GSH-depletion in sensitizing radioresistant cells; and the evidence for an alternative GSH-linked recovery pathway. From an examination of these areas, it seems warranted to conclude the following: First, manipulation of the redox state is effective in enhancing the cytotoxic effect of radiation and some chemotherapeutic agents in radio- and chemoresistant cells; second, additional possibilities for a therapeutic advantage may be gained as new redox modifiers are developed; and last, membrane domains may provide important sites for redox alterations by naturally occurring substrates or by drugs specifically designed to alter this interaction.


Asunto(s)
Antineoplásicos/farmacología , Glutatión/metabolismo , Tolerancia a Radiación , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Cricetinae , Resistencia a Medicamentos , Humanos , Técnicas In Vitro , Ratones , Oxidación-Reducción , Ratas
5.
Int J Radiat Oncol Biol Phys ; 16(5): 1219-23, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2715073

RESUMEN

From analytical expressions derived for the radical-repair (competition) model describing the relationship between cellular radiosensitivity and oxygen concentration, "K-curve" behavior has been quantified as a function of the concentration of the species S which restitutes the radiation-induced radicals to their original molecular configuration. If these species are identified with thiols, K-curves modified by fractionally depleting [S] through calculation can be compared with experimental data where cells have their thiols depleted using various means, for example, by chemical agents or by the use of cells with decreased thiols because of genetic deficiency. Families of curves have been calculated related both to the S-depleted and the non-S-depleted hypoxic control, the latter of which is used to calculate enhancement ratios. Comparison of the model with experimental data is made.


Asunto(s)
Reparación del ADN , Compuestos de Sulfhidrilo/metabolismo , Animales , Células/efectos de la radiación , Modelos Biológicos , Oxígeno/fisiología , Tolerancia a Radiación
6.
Int J Radiat Oncol Biol Phys ; 10(8): 1221-7, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6469742

RESUMEN

We have measured the rate of GSH resynthesis in plateau phase cultures of A549 human lung carcinoma cells subjected to a fresh medium change. Buthionine sulfoximine (BSO) blocks this resynthesis. Diethyl maleate (DEM) causes a decrease in accumulation of GSH. If DEM is added concurrently with BSO there is a rapid decline in GSH that is maximal in the presence of 0.5 mM DEM. GSH depletion rapidly occurs when BSO is added to log phase cultures which initially are higher in GSH content. Twenty-four hr treatment of A549 cells with BSO results in cells that are more radiosensitive in air and show a slight hypoxic radiation response. A 2 hr treatment with either 0.25 mM or 0.5 mM DEM results in some hypoxic sensitization and little increase in the aerobic radiation response. The 24 hr BSO + 2 hr DEM treatment sensitizes hypoxic cells to a greater degree than either agent alone but does not increase the aerobic response more than is seen with BSO alone. Cells treated simultaneously with BSO + DEM show little increase in the hypoxic radiation response, compared to DEM alone, but are more sensitive under aerobic conditions. Decreased cell survival for aerobically irradiated log phase A549 cells occurs within minutes after addition of a mixture of BSO + DEM. The decreased cell survival following aerobic irradiation, after prolonged treatment with BSO or acute exposure to BSO + DEM, may be in part due to inhibition of glutathione peroxidases. For example, glutathione-S-transferase, known to have glutathione peroxidase activity (non-selenium), is nearly completely inhibited by the BSO treatments. In addition, cellular capacity to react with peroxide (glutathione peroxidase, selenium containing) was also inhibited. We suggest that the enhanced aerobic radiation response is related to an inability of GSH depleted cells to inactivate either peroxy radicals or hydroperoxides that may be produced during irradiation of BSO treated cells. Furthermore, enhancement of the aerobic radiation response may be useful in vivo if normal tissue responses are not also increased.


Asunto(s)
Glutatión/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacología , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Neoplasias Pulmonares/patología , Maleatos/farmacología , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Factores de Tiempo
7.
Int J Radiat Oncol Biol Phys ; 12(7): 1161-4, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-2943713

RESUMEN

Chronic aerobic exposure of A549 human lung carcinoma cell cultures to 0.1 mM L-buthionine-S,R-sulfoximine and 1 mM misonidazole, or 1 mM SR-2508 results in inhibition of cell growth and decreased clonogenic survival. These patterns are not apparent with the individual drug treatments. Both parameters demonstrate maximum toxicity after 72 hr in culture, which parallels the time required to deplete A549 cells of glutathione with 0.1 mM L-BSO under these growth conditions. Toxicity appears to be related to hydrogen peroxide-produced during 1 electron reduction of the nitro compounds in the presence of oxygen. A549 cells have a lowered capacity to reduce peroxide due to the effect of thiol depletion on the enzymes GSH-peroxidase and GSH-S-transferase, which require the tripeptide as a substrate. The addition of catalase, another important enzyme involved in peroxide reduction, partially reverses the observed toxicity. 4-Hydroxypyrazole, which inhibits endogenous catalase activity, causes an increase in the observed cytotoxicity. Similar effects of L-BSO and 4-hydroxypyrazole are seen for toxicity due to hydrogen peroxide being added directly to cell cultures.


Asunto(s)
Metionina Sulfoximina/análogos & derivados , Misonidazol/toxicidad , Nitroimidazoles/toxicidad , Pirazoles/farmacología , Fármacos Sensibilizantes a Radiaciones/toxicidad , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Etanidazol , Glutatión/metabolismo , Humanos , Peróxido de Hidrógeno/biosíntesis , Técnicas In Vitro , Metionina Sulfoximina/farmacología , Oxígeno/fisiología
8.
Int J Radiat Oncol Biol Phys ; 16(5): 1311-4, 1989 May.
Artículo en Inglés | MEDLINE | ID: mdl-2715084

RESUMEN

We will review the relationships between glutathione (GSH), protein thiols, and cellular responses to radiation, peroxides, and peroxide-producing drugs. Our primary interest involves the behavior of sulfhydryls as electron and hydrogen carriers, and their capacity to protect various target molecules against radiation and peroxidative damage. We used reagents such as L-buthionine sulfoximine (LBSO), alone and in combination with N-ethyl maleimide (NEM), diamide, and dimethylfumarate, to decrease GSH so that it could no longer participate in the electron transfer reactions. Our results indicate that aerobic sensitization produced by GSH depletion can be further enhanced if electron-accepting agents, such as tertiary butyl hydroperoxide (t-BOOH), are present during irradiation. Hydroperoxide is a substrate for glutathione peroxidase and diverts electrons and hydrogen away from target molecules during its reduction. Sensitivity to radiation seems to be due to the inhibition of the mitochondria's capacity to reduce hydroperoxide. We will also report the mitochondria's ability to reduce the oxygen radicals produced by radiation and drugs. Data also indicate that t-BOOH oxidizes protein thiols which are enzymatically involved in repair of DNA damage.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Glutatión/fisiología , Tolerancia a Radiación , Aerobiosis , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Peróxidos/farmacología , terc-Butilhidroperóxido
9.
Int J Radiat Oncol Biol Phys ; 12(7): 1139-42, 1986 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3744931

RESUMEN

Our data show that A549 cells are increasingly radiosensitive with prolonged exposure to L-BSO. The resulting glutathione and protein thiol depleted cells show both loss of shoulder and slope modification. Furthermore, there is an increase in single strand DNA breaks and irrepairable cross-linking. The aerobic radiation damage in the thiol depleted state appears to be different from that obtained with hypoxic cells. Any postulated role for GSH in reducing or preventing peroxidative radiation damage must also include protection against single strand DNA breaks as well as involvement in repairing DNA-protein cross-links. The latter effect may be related to decreased protein thiol content as reflected in a decreased enzyme capacity to repair DNA damage.


Asunto(s)
Metionina Sulfoximina/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/farmacología , Butionina Sulfoximina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Glutatión/metabolismo , Humanos , Técnicas In Vitro , Metionina Sulfoximina/farmacología , Oxígeno/fisiología , Factores de Tiempo
10.
Biochem Pharmacol ; 35(1): 77-90, 1986 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-2934068

RESUMEN

Misonidazole is a metabolically active drug. Its addition to cells causes an immediate alteration in cellular electron transfer pathways. Under aerobic conditions the metabolic alterations can result in futile cycling with electron transfer to oxygen and production of peroxide. Thiol levels are extremely important in protecting the cell against the peroxide formation and potentially hazardous conditions for hydroxyl radical production. Nevertheless such electron shunting out of cellular metabolism will result in alterations in pentose cycle, glycolysis and cellular capacity to reduce metabolites to essential intermediates needed in DNA metabolism (i.e. deoxyribonucleotides). Glutathione must be depleted to very low levels before toxic effects of misonidazole and other nitro compounds are manifested in cell death via peroxidative damage. Under hypoxic conditions misonidazole also diverts the pentose cycle via its own reduction; however, unlike the aerobic conditions, there are a number of reductive intermediates produced that react with non-protein thiols such as GSH as well as protein thiols. The reaction with protein thiols results in the inhibition of glycolysis and other as yet undetermined enzyme systems. The consequences of the hypoxic pretreatment of cells with nitro compounds are increased vulnerability to radiation and chemotherapeutic drugs such as L-PAM, cis-platinum and bleomycin. The role that altered enzyme activity has in the cellular response to misonidazole and chemotherapeutic agents remains to be determined. It is also clear that the GSH depleted state not only makes cells more vulnerable to oxidative stress but also to hypoxic intermediates produced by the reduction of misonidazole beyond the one electron stage. The relevancy of the present work to the proposed use of thiol depletion in vivo to enhance the radiation or chemotherapeutic response of tumor tissue lies with the following considerations. Apparently, spontaneous peroxidative damage to normal tissue such as liver can occur with GSH depletion to 10-20% of control and with other normal tissue when GSH reaches 50% of control. This situation can obviously become more critical if peroxide producing drugs are administered. The only advantage to such combined drug treatments would lie in the possibility that tumors vary in their catalase and peroxidase activity and consequently may be more vulnerable to oxidative stress (cf. review by Meister. Our tumor model, the A549 human lung carcinoma cell in vitro, appears to be an exception because it has catalase, peroxidase and a high content of GSH.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Nitroimidazoles/metabolismo , Animales , Butionina Sulfoximina , Catalasa , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Etanidazol , Radicales Libres , Glucosa/metabolismo , Glutatión/metabolismo , Glucólisis , Hexosafosfatos/metabolismo , Humanos , Hipoxia , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología , Microcuerpos/metabolismo , Microsomas/metabolismo , Misonidazol/metabolismo , Misonidazol/farmacología , Mitocondrias/metabolismo , Oxidación-Reducción , Pentosas/metabolismo , Peroxidasas/metabolismo , Compuestos de Sulfhidrilo/metabolismo
11.
Radiat Res ; 115(3): 495-502, 1988 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-3174933

RESUMEN

Dimethylfumarate (DMF) depletes intracellular glutathione (GSH) by covalent bond formation in a reaction which may be mediated by GSH-S-transferase. In Chinese hamster ovary cells this depletion is rapid; e.g., 0.5 mM DMF depletes GSH to less than 10% of control in 5 min at room temperature. DMF is a very effective hypoxic cell radiosensitizer, with an enhancement ratio (ER) of about 3 obtained by a 5-min exposure of cells at room temperature to 5 mM DMF, without significant toxicity. At this same concentration of drug, there is a small enhancement of aerobic cells (ER = 1.3), but the 5 mM DMF in hypoxia results in nearly a complete collapse of the hypoxic dose-response curve to the same level as seen in air with DMF. It has been suggested previously that DMF sensitizes cells via electron affinic mechanisms. However, this appears not to be the case in this study, as shown by the fact that cells pretreated with DMF and then washed free of the drug remained equally radiosensitive as cells irradiated in the presence of the drug. This large enhancement of radiation sensitivity appears to be related to the drug's ability to deplete thiols; i.e., thiols appear to be a major factor responsible for radioresistance of hypoxic cells.


Asunto(s)
Fumaratos/farmacología , Glutatión/metabolismo , Fármacos Sensibilizantes a Radiaciones , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Cricetinae , Dimetilfumarato , Femenino , Cinética , Ovario , Oxígeno/farmacología
12.
Radiat Res ; 108(3): 238-50, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3797633

RESUMEN

The effect of changes in both the intracellular glutathione (GSH) concentration and the concentration of extracellular reducing equivalents on the aerobic radiosensitization was studied in three cell lines: CHO-10B4, V79, and A549. Intracellular GSH was metabolically depleted after the inhibition of GSH synthesis by buthionine sulfoximine (BSO), while the extracellular environment was controlled through the replacement of growth medium with a thiol-free salt solution and in some experiments by the exogenous addition of either GSH or GSSG. Each of the cell lines examined exhibited an enhanced aerobic radioresponse when the intracellular GSH was extensively depleted (GSH less than 1 nmol GSH/10(6) cells after 1.0 mM BSO/24 h treatment) and the complexity of the extracellular milieu decreased. Although the addition of oxidized glutathione (5 mM GSSG/30 min) to cells prior to irradiation was without effect, much or all of the induced radiosensitivity was overcome by the addition of reduced glutathione (5 mM GSH/15 min). However, the observation that the exogenous GSH addition restores the control radioresponse without increasing the intracellular GSH concentration was entirely unexpected. These results suggest that a number of factors exert an influence on the extent of GSH depletion and determine the extent of aerobic radiosensitization. Furthermore, the interaction of exogenous GSH with--but without penetrating--the cell membrane is sufficient to result in radiorecovery.


Asunto(s)
Glutatión/fisiología , Tolerancia a Radiación , Aerobiosis , Butionina Sulfoximina , Línea Celular , Membrana Celular/metabolismo , Medios de Cultivo , Glutatión/análisis , Metionina Sulfoximina/análogos & derivados , Metionina Sulfoximina/farmacología
13.
Radiat Res ; 130(3): 366-71, 1992 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-1594764

RESUMEN

Cells cope with radiation damage through several mechanisms: (1) increased DNA repair activity, (2) scavenging and inactivation of radiation-induced radical molecules, and (3) entry into a G0-like quiescent state. We have investigated a chromosomal rearrangement to elucidate further the molecular and genetic mechanisms underlying these phenomena. A mutant of Escherichia coli JM83 (phi 80dlacZ delta M15) was isolated that demonstrated significantly increased resistance to both ionizing and ultraviolet radiation. Surviving fractions of mutant and wild-type cells were measured following exposure to standardized doses of radiation. Increased radioresistance was directly related to a chromosomal alteration near the bacteriophage phi 80 attachment site (attB), as initially detected by the LacZ- phenotype of the isolate. Southern hybridization of chromosomal DNA from the mutant and wild-type E. coli JM83 strains indicated that a deletion had occurred. We propose that the deletion near the attB locus produces the radioresistant phenotype of the E. coli JM83 LacZ- mutant, perhaps through the alteration or inactivation of a gene or its controlling element(s).


Asunto(s)
Cromosomas/ultraestructura , Escherichia coli/efectos de la radiación , Mutación , Genética de Radiación , Tolerancia a Radiación/genética , Rayos Ultravioleta , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Escherichia coli/citología , Escherichia coli/genética
14.
Radiat Res ; 126(2): 244-50, 1991 May.
Artículo en Inglés | MEDLINE | ID: mdl-2023995

RESUMEN

The cellular Ha-ras oncogene, activated by missense mutations, has been implicated in intrinsic resistance to ionizing radiation. This study shows that the overexpression of the unmutated gene (proto-oncogene) may also be involved in how the cells respond to radiation. The experimental system consisted of mouse NIH 3T3-derived cell lines which carry multiple copies of a transcriptionally activated human c-Ha-ras proto-oncogene. Both tumorigenic (RS485) and revertant nontumorigenic subclones (PR4 and 4C3) which have high levels of ras expression exhibited a marked increase in radioresistance as measured by D0 compared to control NIH 3T3 cells. Other nontransformed cells with elevated levels of ras (phenotypically revertant line 4C8-A10) also had a significantly increased resistance to radiation, further indicating an association between ras and radioresistance. The increased radioresistance of the RS485 and phenotypic revertants could not be explained by a differential expression of the myc or metallothionein I genes or by variations in cell cycle. The correlation between increased ras proto-oncogene expression and radioresistance suggests that the ras encoded p21, a plasma membrane protein, may participate in the cellular responses to ionizing radiation.


Asunto(s)
Transformación Celular Neoplásica/genética , Genes ras , Proto-Oncogenes , Tolerancia a Radiación/genética , Animales , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Ratones , Proto-Oncogenes Mas
15.
Radiat Res ; 93(3): 479-91, 1983 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-6856753

RESUMEN

The response of cultured CHO cells to ultrahigh-dose-rate radiation (approximately 10(9) Gy/sec) has been previously studied extensively using the thin-layer cell-handling technique developed in this laboratory. When the cells are equilibrated with a low concentration of oxygen, e.g., 0.44% O2, a breaking survival curve, due to radiolytic depletion of the oxygen, is observed. Hypoxic cells irradiated in the presence of the nitroimidazoles (e.g., misonidazole) are sensitized at ultrahigh dose rates in a dose-modifying manner, similar to that observed at conventional dose rates. These radiosensitizer compounds, if present in cells equilibrated with a low concentration of oxygen, prevent the breaking behavior of the survival curve, an observation believed to be due to the sensitizer interfering with the oxygen depletion process, leaving oxygen free to sensitize. Such experiments have recently been extended to studies with diamide, which, unlike the other sensitizers tested, acts primarily as a shoulder-modifying rather than a dose-modifying agent in hypoxic mammalian cells. These data indicate that diamide is active as a sensitizer at ultrahigh dose rates in a manner similar to that observed at conventional dose rates, and does modify the shape of the breaking survival curve observed with low concentrations of oxygen.


Asunto(s)
Compuestos Azo/farmacología , Supervivencia Celular/efectos de la radiación , Diamida/farmacología , Oxígeno , Animales , Línea Celular , Cricetinae , Cricetulus , Reparación del ADN/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Ovario
16.
Radiat Res ; 95(3): 437-55, 1983 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-6684310

RESUMEN

Cellular nonprotein thiols (NPSH) consist of glutathione (GSH) and other low molecular weight species such as cysteine, cysteamine, and coenzyme A. GSH is usually less than the total cellular NPSH, and with thiol reactive agents, such as diethyl maleate (DEM), its rate of depletion is in part dependent upon the cellular capacity for its resynthesis. If resynthesis is blocked by buthionine-S,R-sulfoximine(BSO), the NPSH, including GSH, is depleted more rapidly, Cellular thiol depletion by diamide, N-ethylmaleimide, and BSO may render oxygenated cells more sensitive to radiation. These cells may or may not show a reduction in the oxygen enhancement ratio (OER). Human A549 lung carcinoma cells depleted of their NPSH either by prolonged culture or by BSO treatment do not show a reduced OER but do show increased aerobic responses to radiation. Some nitroheterocyclic radiosensitizing drugs also deplete cellular thiols under aerobic conditions. Such reactivity may be the reason that they show anomalous radiation sensitization (i.e., better than predicted on the basis of electron affinity). Other nitrocompounds, such as misonidazole, are activated under hypoxic conditions to radical intermediates. When cellular thiols are depleted peroxide is formed. Under hypoxic conditions thiols are depleted because metabolically reduced intermediates react with GSH instead of oxygen. Thiol depletion, under hypoxic conditions, may be the reason that misonidazole and other nitrocompounds show an extra enhancement ratio with hypoxic cells. Thiol depletion by DEM or BSO alters the radiation response of hypoxic cells to misonidazole. In conclusion, we propose an altered thiol model which includes a mechanism for thiol involvement in the aerobic radiation response of cells. This mechanism involves both thiol-linked hydrogen donation to oxygen radical adducts to produce hydroperoxides followed by a GSH peroxidase-catalyzed reduction of the hydroperoxides to intermediates entering into metabolic pathways to produce the original molecule.


Asunto(s)
Compuestos Azo/farmacología , Diamida/farmacología , Glutatión/metabolismo , Maleatos/farmacología , Metionina Sulfoximina/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/farmacología , Compuestos de Sulfhidrilo/metabolismo , Animales , Butionina Sulfoximina , Supervivencia Celular , Células Cultivadas , Fenómenos Químicos , Química , Cricetinae , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Neoplasias Pulmonares , Metionina Sulfoximina/farmacología , Ovario , Oxígeno
17.
Radiat Res ; 98(2): 370-80, 1984 May.
Artículo en Inglés | MEDLINE | ID: mdl-6539482

RESUMEN

Buthionine sulfoximine (BSO) inhibits the synthesis of glutathione (GSH), the major nonprotein sulfhydryl (NPSH) present in most mammalian cells. BSO concentrations from 1 microM to 0.1 mM reduced intracellular GSH at different rates, while BSO greater than or equal to 0.1 mM (i.e., 0.1 to 2.0 mM), resulting in inhibitor-enzyme saturation, depleted GSH to less than 10% of control within 10 hr at about equal rates. BSO exposures used in these experiments were not cytotoxic with the one exception that 2.0 mM BSO/24 hr reduced cell viability to approximately 50%. However, alterations in either the cell doubling time(s) or the cell age density distribution(s) were not observed with the BSO exposures used to determine its radiosensitizing effect. BSO significantly radiosensitized (ER = 1.41 with 0.1 mM BSO/24 hr) hypoxic, but not aerobic, CHO cells when the GSH and NPSH concentrations were reduced to less than 10 and 20% of control, respectively, and maximum radiosensitivity was even achieved with microM concentrations of BSO (ER = 1.38 with 10 microM BSO/24 hr). Furthermore, BSO exposure (0.1 mM BSO/24 hr) also enhanced the radiosensitizing effect of various concentrations of misonidazole on hypoxic CHO cells.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Glutatión/fisiología , Metionina Sulfoximina/análogos & derivados , Misonidazol/farmacología , Nitroimidazoles/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Animales , Butionina Sulfoximina , Supervivencia Celular/efectos de los fármacos , Cricetinae , Cricetulus , Relación Dosis-Respuesta en la Radiación , Sinergismo Farmacológico , Femenino , Metionina Sulfoximina/farmacología , Ovario , Oxígeno , Factores de Tiempo
18.
Int J Radiat Biol ; 64(5): 547-54, 1993 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7902394

RESUMEN

Expression of ras has been correlated with increased intrinsic resistance to ionizing radiation. In this study we show that increased EJras expression in human cells is associated with a decrease in the frequency of radiation-induced micronuclei. The experimental system consisted of human osteosarcoma-derived cell lines which quantitatively vary in their EJras expression. There was a dose-dependent relationship between radiation dose and micronuclei formation in all cell lines tested. Human osteosarcoma cells, in which the ras level was undetectable, had the highest frequency of micronuclei production at all radiation doses tested. At 4 Gy the most radioresistant cells exhibited a 41.5 +/- 5% decrease in the production of micronuclei concomitant with high ras expression in comparison with the relatively radiosensitive parental cell line. Cells expressing a low amount of EJras demonstrated a 23 +/- 3% decrease in micronuclei induction compared with parental cells. Treatment of cells with lovastatin, an inhibitor of ras-encoded p21ras post-translational processing via the mevalonate pathway, markedly decreased the yield of micronuclei formation in cells transfected with ras; the drug had no effect on radiation-induced micronuclei formation in parental cells. The use of the in vitro micronuclei assay has provided a convenient way to visualize differences in the genotoxic damage induced by ionizing radiation in cells which express different amount of EJras. The results indicate that elevation of ras expression in human cells can lead to a decrease in the number of radiation-induced micronuclei formed and that this relationship is dependent on membrane association of ras-encoded p21.


Asunto(s)
Pruebas de Micronúcleos , Osteosarcoma/genética , Línea Celular , Genes ras , Humanos , Lovastatina/farmacología , Genética de Radiación
19.
Int J Radiat Biol ; 60(4): 613-26, 1991 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1680142

RESUMEN

Double-strand break (dsb) induction and rejoining after ionizing radiation was analysed in Deinococcus radiodurans and a radiosensitive mutant by pulsed-field gel electrophoresis. Following 2 kGy, migration of genomic DNA (not restriction cleaved) from the plug into the gel was extensive, but was not observed after 90 min postirradiation recovery. By this time D. radiodurans chromosomes were intact, as demonstrated by restoration of the Not I restriction cleavage pattern of 11 bands, which we found to be the characteristic pattern in unirradiated cells. Following the higher exposure of 4 kGy, dsb rejoining took approximately 180 min, twice as long as required following the 2 kGy exposure. Restoration of dsb in the radiosensitive mutant strain 112, which appears to be defective in recombination, was markedly retarded at both 2 and 4 kGy. The Not I restriction fragments of wild-type D. radiodurans and the radiosensitive mutant were identical, totaling 3.58 Mbp, equivalent to 2.36 x 10(9) daltons per chromosome.


Asunto(s)
Reparación del ADN , ADN Bacteriano/efectos de la radiación , Mutación , Tolerancia a Radiación/genética , ADN/efectos de la radiación , Daño del ADN , Electroforesis , Bacterias Grampositivas/efectos de la radiación
20.
Int J Radiat Biol ; 57(4): 709-22, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-1969903

RESUMEN

The most effective radioprotective agents exhibit toxicities that can limit their usefulness. It may be possible to use combinations of agents with different radioprotective mechanisms of action at less toxic doses, or to reduce the toxicity of the major protective compound by adding another agent. With regard to the latter possibility, improved radioprotection and reduced lethal toxicity of the phosphorothioate WR-2721 was observed when it was administered in combination with metals (selenium, zinc or copper). The known mechanisms of action of potential radioprotective agents and varying effects of different doses and times of administration in relation to radiation exposure must be considered when using combined-agent regimens. A number of receptor-mediated protectors and other biological compounds, including endotoxin, eicosanoids and cytokines, have at least an additive effect when administered with thiol protectors. Eicosanoids and other bioactive lipids must be administered before radiation exposure, whereas some immunomodulators have activity when administered either before or after radiation exposure. For example, the cytokine interleukin-1 administered simultaneously with WR-2721 before irradiation or after irradiation enhances the radioprotective efficacy of WR-2721. The most effective single agents or combinations of protectors result in a decrement in locomotor activity, an index of behavioral toxicity. Recent evidence indicates that administration of the CNS stimulant caffeine mitigates the behavioral toxicity of an effective radioprotective dose of the phosphorothioate WR-3689 without altering its radioprotective efficacy. These examples indicate that the use of combinations of agents is a promising approach for maximizing radioprotection with minimal adverse effects.


Asunto(s)
Protectores contra Radiación/uso terapéutico , Animales , Quimioterapia Combinada , Masculino , Ratones , Protectores contra Radiación/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA