Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
3.
J Prosthet Dent ; 75(4): 453-6, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8642537

ABSTRACT

Silicone putty casts are useful for articulating existing dentures for evaluation and problem solving before relining procedures are performed or a new prosthesis is made. Tissue health may require significant modification of the existing dentures and treatment with tissue-conditioning material before final impressions are made. This practical procedure uses a functional impression that substitutes for a quality preliminary impression and combines tissue conditioning, a functional impression, silicone putty cast, custom final impression tray, and a final impression for a complete denture.


Subject(s)
Dental Impression Technique , Denture, Complete , Models, Dental , Silicone Elastomers , Dental Impression Technique/instrumentation , Humans , Technology, Dental/methods , Tissue Conditioning, Dental
4.
J Pharm Biomed Anal ; 14(3): 317-24, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8851756

ABSTRACT

Metabolically active forms of the radioprotective and chemoprotective drug S-3-(3-methylaminopropylamino)propylphosphorothioic acid (WR-151327) are S-3-(3-methylaminopropylamino)propanethiol (WR-151326) and its symmetrical disulfide (WR-25595501). This paper describes applications of sensitive and specific procedures such as capillary column gas chromatography with flame ionization detection, electron impact mass spectrometry and liquid chromatography with electrochemical detection for structural characterization and analysis of the active forms of WR-151327. These chromatographic procedures provide reproducible linear calibration graphs for a relatively wide range of concentrations of the active forms of WR-151327. The described procedures will further facilitate in vivo and in vitro investigations of chemoprotective and radioprotective properties of WR-151327 and its active metabolites.


Subject(s)
Radiation-Protective Agents/analysis , Sulfhydryl Compounds/analysis , Chromatography, Liquid , Electrochemistry , Gas Chromatography-Mass Spectrometry , Indicators and Reagents , Iodine/chemistry , Metals/chemistry , Oxidation-Reduction
5.
Int J Hyperthermia ; 10(3): 309-13, 1994.
Article in English | MEDLINE | ID: mdl-7930796

ABSTRACT

The C3H 10T1/2 mouse embryo cell line was used to determine the effect of hyperthermia on the in vitro oncogenic transforming potential of radiation. Heat exposures at 45 degrees C/15 min or at 43 degrees C/60 min administered alone yielded no significant transformation as previously reported. However, our recent results repeat our earlier findings that there is an increase in the in vitro transformation frequency after the combined treatment of hyperthermia and radiation, if foci/flask or foci/surviving cell are used to calculate transformation frequency, if high temperature exposures are used (e.g. 43 degrees C/60 min or 45 degrees C/15 min) and if the time between the combined treatments of hyperthermia and 200 cGy of 60Co radiation is < or = 5 min at ambient temperature. As can be seen in this and past reports whether the combination of hyperthermia and radiation show an increase, a decrease, or no change in in vitro oncogenic transformation, a number of factors are critical. These critical factors are (1) temperature/exposure time and radiation dose as expected; (2) stage of the cell cycle and growth conditions at each exposure; (3) time between treatments; and (4) method of data analysis, i.e. whether the transformation frequency was based on the foci/viable cells, foci/flask or the foci/total cells at risk (total cells plated x plating efficiency of the untreated cells). Recent publications have shown that the position of cells in the cell cycle determine the frequency of cell transformation (Cao et al. 1992, Miller et al. 1992). Factors 1-3 affect the cells position in the cell cycle.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Hot Temperature/adverse effects , Animals , Cell Cycle , Cell Line , Cell Transformation, Neoplastic/pathology , Cocarcinogenesis , Hyperthermia, Induced/adverse effects , Mice , Time Factors , Trypsin
6.
Int J Radiat Biol ; 64(5): 547-54, 1993 Nov.
Article in English | MEDLINE | ID: mdl-7902394

ABSTRACT

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.


Subject(s)
Micronucleus Tests , Osteosarcoma/genetics , Cell Line , Genes, ras , Humans , Lovastatin/pharmacology , Radiation Genetics
7.
Mol Cell Biol ; 13(7): 4416-22, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8321241

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, ras , Glutathione/metabolism , RNA Processing, Post-Transcriptional , 3T3 Cells , Animals , Blotting, Northern , Buthionine Sulfoximine , Cell Cycle , Cycloheximide/pharmacology , Down-Regulation , Humans , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology , Mice , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , Proto-Oncogene Mas , Transcription, Genetic , Tumor Cells, Cultured
8.
Int J Cancer ; 53(2): 302-7, 1993 Jan 21.
Article in English | MEDLINE | ID: mdl-8425769

ABSTRACT

Alterations in ras oncogene expression have been associated with increased cellular resistance to ionizing radiation. As an extension of studies with murine cell models, we have now explored the radioresponses of human osteosarcoma (HOS) sub-clones that differ in their EJras expression. Quantitative analysis revealed a tight correlation between the amounts of ras-encoded mRNA and p21 produced, and the degree of cell radioresistance. Interestingly, treatment of the ras-transformed cells with lovastatin, an inhibitor of p21ras post-translational processing via the mevalonate pathway, markedly decreased their radioresistance. Under the experimental conditions used, lovastatin prevented the membrane association, but not the biosynthesis, of p21. The decline in radiation resistance following lovastatin treatment could not be attributed to perturbation of cholesterol metabolism or to non-specific cell-cycle effects. In agreement, lovastatin did not alter the radiation responses of control HOS cells that do not express EJras, or those with an activated met oncogene. The results indicate that elevation in ras gene expression can lead to increased radioresistance of human tumor cells. It appears, however, that p21ras membrane localization is critical for maintenance of the radioresistant phenotype, thus providing a target for pharmacological intervention.


Subject(s)
Genes, ras/physiology , Lovastatin/pharmacology , Osteosarcoma/radiotherapy , Proto-Oncogene Proteins p21(ras)/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Animals , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Transformed , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Osteosarcoma/genetics , Osteosarcoma/metabolism , Protein Prenylation/drug effects , Proto-Oncogene Proteins p21(ras)/drug effects , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects
9.
Radiat Res ; 130(3): 366-71, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1594764

ABSTRACT

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).


Subject(s)
Chromosomes/ultrastructure , Escherichia coli/radiation effects , Mutation , Radiation Genetics , Radiation Tolerance/genetics , Ultraviolet Rays , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Escherichia coli/cytology , Escherichia coli/genetics
10.
Int J Radiat Biol ; 60(4): 613-26, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1680142

ABSTRACT

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.


Subject(s)
DNA Repair , DNA, Bacterial/radiation effects , Mutation , Radiation Tolerance/genetics , DNA/radiation effects , DNA Damage , Electrophoresis , Gram-Positive Bacteria/radiation effects
11.
Radiat Res ; 126(2): 244-50, 1991 May.
Article in English | MEDLINE | ID: mdl-2023995

ABSTRACT

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.


Subject(s)
Cell Transformation, Neoplastic/genetics , Genes, ras , Proto-Oncogenes , Radiation Tolerance/genetics , Animals , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Mice , Proto-Oncogene Mas
12.
Int J Radiat Biol ; 57(4): 709-22, 1990 Apr.
Article in English | MEDLINE | ID: mdl-1969903

ABSTRACT

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.


Subject(s)
Radiation-Protective Agents/therapeutic use , Animals , Drug Therapy, Combination , Male , Mice , Radiation-Protective Agents/administration & dosage
13.
Int J Radiat Oncol Biol Phys ; 16(5): 1219-23, 1989 May.
Article in English | MEDLINE | ID: mdl-2715073

ABSTRACT

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.


Subject(s)
DNA Repair , Sulfhydryl Compounds/metabolism , Animals , Cells/radiation effects , Models, Biological , Oxygen/physiology , Radiation Tolerance
14.
Int J Radiat Oncol Biol Phys ; 16(5): 1311-4, 1989 May.
Article in English | MEDLINE | ID: mdl-2715084

ABSTRACT

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.


Subject(s)
Cell Survival/radiation effects , Glutathione/physiology , Radiation Tolerance , Aerobiosis , Buthionine Sulfoximine , Cell Line , Cell Survival/drug effects , Humans , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology , Peroxides/pharmacology , tert-Butylhydroperoxide
16.
Radiat Res ; 115(3): 495-502, 1988 Sep.
Article in English | MEDLINE | ID: mdl-3174933

ABSTRACT

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.


Subject(s)
Fumarates/pharmacology , Glutathione/metabolism , Radiation-Sensitizing Agents , Animals , Cell Line , Cell Survival/radiation effects , Cricetinae , Dimethyl Fumarate , Female , Kinetics , Ovary , Oxygen/pharmacology
19.
Radiat Res ; 108(3): 238-50, 1986 Dec.
Article in English | MEDLINE | ID: mdl-3797633

ABSTRACT

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.


Subject(s)
Glutathione/physiology , Radiation Tolerance , Aerobiosis , Buthionine Sulfoximine , Cell Line , Cell Membrane/metabolism , Culture Media , Glutathione/analysis , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology
20.
Int J Radiat Oncol Biol Phys ; 12(7): 1161-4, 1986 Jul.
Article in English | MEDLINE | ID: mdl-2943713

ABSTRACT

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.


Subject(s)
Methionine Sulfoximine/analogs & derivatives , Misonidazole/toxicity , Nitroimidazoles/toxicity , Pyrazoles/pharmacology , Radiation-Sensitizing Agents/toxicity , Buthionine Sulfoximine , Cell Line , Cell Survival/drug effects , Etanidazole , Glutathione/metabolism , Humans , Hydrogen Peroxide/biosynthesis , In Vitro Techniques , Methionine Sulfoximine/pharmacology , Oxygen/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...