Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
J Natl Cancer Inst ; 59(6): 1737-9, 1977 Dec.
Article in English | MEDLINE | ID: mdl-200761

ABSTRACT

In vitro drug metabolism in the Hartley guinea pig was compared with that in two inbred guinea pig strains used as carriers for the line 10 hepatoma. We observed minor differences in enzyme specific activity among the three strains. Three weeks after intradermal inoculation of Strain 2 guinea pigs with line 10 hepatoma cells, cytochrome P450 levels and aminopyrine demethylase activity were significantly decreased. Seven to 10 days after inoculation with the ascites form of the tumor, the activities of aniline and biphenyl hydroxylases, p-aminobenzoic acid N-acetyltransferase, and dichloronitrobenzene glutathione S-aryltransferase, in addition to those of cytochrome P450 and aminopyrine N-demethylase, were probably also described.


Subject(s)
Antineoplastic Agents/metabolism , Carcinoma, Hepatocellular/metabolism , Guinea Pigs/metabolism , Liver Neoplasms/metabolism , Microsomes, Liver/metabolism , Aminopyrine N-Demethylase/metabolism , Animals , Cytochrome P-450 Enzyme System/metabolism , In Vitro Techniques , Male , Microsomes, Liver/enzymology , Neoplasms, Experimental/metabolism , Species Specificity
2.
J Natl Cancer Inst ; 81(24): 1904-9, 1989 Dec 20.
Article in English | MEDLINE | ID: mdl-2593167

ABSTRACT

To investigate the possibility of increased activity of cytotoxic anticancer drugs combined with cytokines, we treated human melanoma cells with combinations of etoposide (VP-16) and human recombinant interleukin-1 alpha (rIL-1 alpha). We evaluated the combined cytotoxic effects of VP-16 and rIL-1 alpha using A375-C6 cells, which are sensitive to rIL-1 alpha, and A375-C5 cells, a clonal variant line resistant to rIL-1 alpha. We used the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium-bromid e) assay and the inhibition of [3H]thymidine incorporation into DNA. We analyzed data using the median effects principle of Chou and Talalay (Chou's analysis). The calculated combination index values, at a dose ratio of VP-16 to rIL-1 alpha of 12:1 in simultaneous exposure, indicated synergistic cytotoxicity toward both A375-C6 cells and A375-C5 cells. We observed more pronounced synergism with VP-16 and rIL-1 alpha toward the A375-C5 IL-1 alpha-resistant melanoma cells. These results suggest that rIL-1 alpha combined with cytotoxic antitumor drugs may provide increased benefit in the treatment of malignant melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Etoposide/pharmacology , Interleukin-1/pharmacology , Melanoma/pathology , Cell Survival/drug effects , DNA/biosynthesis , DNA Damage , Drug Synergism , Etoposide/administration & dosage , Humans , Interleukin-1/administration & dosage , Recombinant Proteins/pharmacology , Tumor Cells, Cultured
3.
Cancer Res ; 51(3): 769-74, 1991 Feb 01.
Article in English | MEDLINE | ID: mdl-1824825

ABSTRACT

To investigate the possibility that anticancer drugs combined with cytokines may show increased activity, human tumor cells were treated with combinations of human recombinant interleukin 1 alpha (rIL-1 alpha) and etoposide (VP-16). The cytotoxicity of these combinations was evaluated by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide assay using rIL-1 alpha-sensitive A375-C6 melanoma cells and A375-C5 cells, a clonal variant line that is resistant to IL-1 alpha. Data were analyzed for synergism by the median effect principle of T-C. Chou and P. Talalay (J. Biol. Chem., 252: 6438-6442, 1977). At a dose ratio of VP-16 to rIL-1 alpha of 12 nM:1 unit/ml in either simultaneous or sequential exposure (VP-16 first), the calculated combination index values indicated synergistic cytotoxicity toward both A375-C6 cells and A375-C5 cells. IL-1 alpha treatment 24 h prior to VP-16 exposure had no advantage over simultaneous treatment. Surface IL-1 alpha receptors on both A375-C6 and A375-C5 cells were measured using 125I-radiolabeled rIL-1 alpha binding; A375-C6 cells had 701 +/- 128 (SD) receptor molecules/cell and A375-C5 cells only had 58 +/- 33 receptor molecules/cell. The dissociation constants for IL-1 alpha were similar in both cell types (19 +/- 6 pM for A375-C6 and 17 +/- 2 pM for A375-C5). The specific binding of rIL-1 alpha to the surface IL-1 alpha receptors of both sensitive and resistant cells was significantly increased in a dose-dependent fashion by the prior treatment with VP-16 (1.75-fold on A375-C6 cells and 3.5-fold on A375-C5 cells). VP-16 also enhanced the internalization of receptor-bound rIL-1 alpha, suggesting that a possible mechanism of the synergistic cytotoxicity of rIL-1 alpha and VP-16 might be related to the modulation of rIL-1 alpha receptors by VP-16, resulting in increased internalization of rIL-1 alpha.


Subject(s)
Etoposide/pharmacology , Interleukin-1/pharmacology , Receptors, Immunologic/metabolism , Drug Administration Schedule , Drug Synergism , Etoposide/administration & dosage , Etoposide/metabolism , Humans , Interleukin-1/administration & dosage , Interleukin-1/metabolism , Melanoma/metabolism , Receptors, Immunologic/drug effects , Receptors, Interleukin-1 , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Tumor Cells, Cultured/metabolism
4.
Cancer Res ; 38(3): 787-92, 1978 Mar.
Article in English | MEDLINE | ID: mdl-75060

ABSTRACT

Bleomycin treatment produced dose-dependent changes in lung collagen content and in several measurable histopathological parameters. NIH/Swiss mice were treated twice weekly for 6 weeks with bleomycin, 0, 1, 20, or 40 mg/kg s.c. The two highest doses produced mortalities of 35 and 100%, respectively, as well as loss of body weight and increase in lung wet weight. Lung hydroxyproline content, an index of collagen, increased to 40 to 50% above control levels at 6 and 8 weeks after initiation of treatment with bleomycin 20 mg/kg. Morphometric analysis was applied to the following parameters at light microscopy: number of intraalveolar macrophages and leukocytes, total pulmonary cell count, alveolar wall thickness, and percentage of consolidation of lung parenchyma. The two highest doses produced increases in all of these parameters as compared to controls. The most marked changes occurred in the number of intraalveolar cells, which in the group given 20 mg/kg rose to 150, 190, and 210% of controls at 4, 6, and 8 weeks. The lowest dose of bleomycin, 1 mg/kg twice weekly for 6 weeks, evoked no pulmonary or other toxicity by the parameters examined. This model of chronic pulmonary toxicity may be useful in analog development, in testing potential antidotes, and in examining the effects of other factors that might modify the pulmonary toxicity of bleomycin.


Subject(s)
Bleomycin/toxicity , Hydroxyproline/metabolism , Lung/drug effects , Animals , Bleomycin/administration & dosage , Cell Count , Collagen/metabolism , Leukocytes/pathology , Lung/metabolism , Lung/pathology , Macrophages/pathology , Male , Mice , Pulmonary Alveoli/pathology
5.
Cancer Res ; 45(9): 4071-6, 1985 Sep.
Article in English | MEDLINE | ID: mdl-2411396

ABSTRACT

Mitomycin C (MC) activation to a reactive species was studied in nuclei isolated from rat liver and EMT6 tumor cells. Both preparations were similar in the rate of 4-(p-nitrobenzyl)pyridine (NBP) alkylation by MC and the levels of NADPH-cytochrome P-450 reductase. MC activation by both hepatic and EMT6 cell nuclei was inhibited by the presence of O2 and by heat inactivation. NADPH was preferred over NADH as the source of reducing equivalents by both types of isolated nuclei. MC activation to alkylating metabolites was not affected when EDTA or diethylenetriaminepentaacetic acid, two Fe2+ chelating agents, was present in the incubation system with either preparation of isolated nuclei. Glutathione (1 and 5 mM) and N-acetylcysteine (1 and 10 mM) both inhibited MC alkylation of NBP in nuclear preparations from rat liver and EMT6 tumor cells by 50-60%. Ethylxanthate (1 mM) effectively inhibited the MC alkylation of NBP by hepatic nuclei but was unable to inhibit MC alkylation of NBP by tumor cell nuclei. At 100 mM, ethylxanthate produced a slight stimulation in the rate of MC alkylation of NBP. These data are consistent with the hypothesis that MC activation in EMT6 tumor cells proceeds via a one electron reduction pathway which is inhibitable by glutathione but not inhibitable by ethylxanthate. Hepatic nuclei are apparently able to activate MC by either a one- or two-electron pathway.


Subject(s)
Cell Nucleus/metabolism , Glutathione/pharmacology , Liver/metabolism , Mammary Neoplasms, Experimental/metabolism , Mitomycins/metabolism , Thiones/pharmacology , Animals , Biotransformation , Dicumarol/pharmacology , Edetic Acid/pharmacology , Hot Temperature , In Vitro Techniques , Male , Mice , Mitomycin , NAD/pharmacology , Oxygen/pharmacology , Pentetic Acid/pharmacology , Rats , Rats, Inbred Strains
6.
Cancer Res ; 44(7): 2892-6, 1984 Jul.
Article in English | MEDLINE | ID: mdl-6327028

ABSTRACT

Rat liver microsomal activation of the anthracycline antitumor drug, Adriamycin, in the presence of reduced pyridine nucleotide under anaerobic conditions produces reactive species that bind covalently to cellular macromolecules including DNA. Since the nuclear membrane contains enzymes capable of activating Adriamycin, we have examined activation of Adriamycin by isolated nuclei. The anaerobic incubation of Adriamycin with rat hepatic nuclei resulted in the formation of the Adriamycin semiquinone free radical. Moreover, this activation resulted in the covalent binding of Adriamycin to nuclear DNA. The binding of Adriamycin to DNA was reduced pyridine nucleotide and time dependent and was significantly decreased in the presence of reduced glutathione or ethylxanthate . Dicumerol , an inhibitor of DT-diaphorase, in contrast, had no effect on this binding. When the incubation was carried out in the presence of oxygen, no semiquinone radical was detected; however, superoxide and hydroxyl radicals were readily detected by a spin-trapping technique. Furthermore, little binding of Adriamycin to nuclear DNA was observed under aerobic conditions. These observations suggest that the nuclear activation and covalent binding of Adriamycin to DNA may be important in the biochemical actions of this drug.


Subject(s)
Cell Nucleus/metabolism , DNA/metabolism , Doxorubicin/metabolism , Liver/metabolism , Microsomes, Liver/metabolism , Anaerobiosis , Animals , Biotransformation , Daunorubicin/metabolism , Electron Spin Resonance Spectroscopy , Kinetics , Oxidation-Reduction , Rats
7.
Cancer Res ; 45(7): 3296-304, 1985 Jul.
Article in English | MEDLINE | ID: mdl-2988766

ABSTRACT

Isolated rat liver nuclei enzymatically activated Adriamycin to the electron spin resonance-detectable semiquinone free radical in the presence of reduced nicotinamide adenine dinucleotide phosphate (NADPH). This process resulted in the enhancement of oxyradical-mediated peroxidation of the nuclear envelope membrane unsaturated phospholipids, measured as malonaldehyde equivalents by the thiobarbituric acid method. Peroxidation required the inclusion of NADPH and catalytically active protein, presumably NADPH:cytochrome P-450 reductase, and was enhanced more than 5-fold by Adriamycin. It was observed that Adriamycin-stimulated nuclear membrane peroxidation was diminished by superoxide dismutase, reduced glutathione, the hydroxyl radical scavenger, 1,3-dimethylurea, and by the metal ion chelator, diethylenetriaminepentaacetic acid, indicating that multiple species of reactive oxygen and trace amounts of metal ions (iron) were required in the peroxidation reaction. The generation of superoxide and hydroxyl radicals was confirmed by 5,5-dimethyl-1-pyrroline-N-oxide spin trap electron spin resonance spectroscopy. Calf thymus DNA added to incubations containing nuclei and NADPH caused a pronounced concentration-dependent inhibition of Adriamycin-stimulated lipid peroxidation. It was found that nuclei incubated with Adriamycin (300 microM) accumulated 128 nmol of the drug per mg of nuclear protein within 1 h, apparently because the Adriamycin was internalized and bound to the DNA and nuclear protein. These results suggest that some of the toxic effects of Adriamycin observed in the nucleus could result, directly or indirectly, through the peroxidation of the unsaturated lipids of the nuclear membrane.


Subject(s)
Cell Nucleus/drug effects , Doxorubicin/pharmacology , Lipid Peroxides/metabolism , Animals , Cell Nucleus/metabolism , Cyclic N-Oxides/metabolism , DNA/metabolism , DNA/pharmacology , Electron Spin Resonance Spectroscopy , Free Radicals , In Vitro Techniques , Male , NADP/pharmacology , NADPH-Ferrihemoprotein Reductase/analysis , Oxygen/metabolism , Rats , Rats, Inbred Strains
8.
Cancer Res ; 49(1): 8-15, 1989 Jan 01.
Article in English | MEDLINE | ID: mdl-2535695

ABSTRACT

Recent evidence supports the concept that Adriamycin cytotoxicity may be mediated by drug semiquinone free radical and oxyradical generation. We tested this hypothesis further by exposing drug-sensitive (WT) and 500-fold Adriamycin-resistant MCF-7 human breast tumor cells (ADRR) to exogenous superoxide- and hydrogen peroxide-generating systems and subsequently monitored cell proliferation as a measure of cytotoxicity. The ADRR tumor cells tolerated a 4-fold greater exposure than sensitive cells to superoxide generated by the xanthine/xanthine oxidase system. Likewise, exposure to hydrogen peroxide produced by the action of glucose oxidase on glucose revealed a 4-fold diminished susceptibility of the drug-resistant cells to this reduced form of oxygen. Similar results were obtained by the direct application of hydrogen peroxide to cells. For both cell lines, cytotoxicity was dependent upon the magnitude and the duration of reactive oxygen exposure. When WT and ADRR cells were cultured under hyperoxia (95% O2:5% CO2), in order to stimulate the intracellular production of oxyradicals, proliferation was inhibited to a greater extent in the drug-sensitive cell line. Additionally, hyperoxia potentiated the cytotoxicity of Adriamycin to both sensitive and drug-resistant cells, but the effect depended upon the concentration of the drug. Under hyperoxic conditions, Adriamycin caused oxygen radical-dependent cytotoxicity to the WT tumor cells at clinically relevant drug concentrations as low as 2 to 3 nM. With ADRR tumor cells, hyperoxia increased the cytotoxicity of Adriamycin at concentrations above 5 microM. Paradoxically, both the WT and the ADRR tumor cells were equally susceptible to the cytotoxic effects of gamma irradiation. It is known that the Adriamycin-resistant MCF-7 cells greatly overexpress glutathione peroxidase and glutathione transferase activities; however, other biochemical defenses against reactive drug intermediates and oxygen radicals have been reported to be similar in the two cell lines. We have reexamined those observations in this report. The resistance of ADRR breast tumor cells to Adriamycin appears to be associated with a developed tolerance to superoxide, most likely because of a twofold increase in superoxide dismutase activity, and a decreased susceptibility to hydrogen peroxide, most likely because of 12-fold augmented selenium-dependent glutathione peroxidase activity. Acting in concert, these two enzymes would decrease the formation of hydroxyl radical from reduced molecular oxygen intermediates.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Breast Neoplasms/pathology , Doxorubicin/pharmacology , Hydrogen Peroxide/pharmacology , Superoxides/pharmacology , Cell Survival/drug effects , Drug Resistance , Female , Free Radicals , Glucose Oxidase/pharmacology , Humans , Oxygen/toxicity , Tumor Cells, Cultured/drug effects
9.
Cancer Res ; 49(3): 511-5, 1989 Feb 01.
Article in English | MEDLINE | ID: mdl-2535960

ABSTRACT

Resistance to antineoplastic drugs is a major problem in the clinical management of cancer. Previous studies have demonstrated that the cytotoxicity of certain anticancer drugs is increased by lowering the glutathione (GSH) levels with buthionine sulfoximine (BSO), a specific inhibitor of gamma-glutamylcysteine synthetase. In this study we report that the resistance to doxorubicin, an anthracycline antibiotic and the most active agent in the treatment of breast cancer, can be partially reversed by exposing MCF-7 doxorubicin-resistant breast tumor cells (MCF-7/ADRR) to minimally cytotoxic doses of BSO. We found that the BSO treatment (50 microM, 48 h) of MCF-7/ADRR cells resulted in 80 to 90% depletion in total GSH concentrations. The toxicity of doxorubicin, as determined by growth inhibition and clonogenic assays, was significantly potentiated in the BSO-treated GSH-depleted cells relative to control breast tumor cells, and a dose-modifying factor of 5 to 7 was observed. Since the cytotoxicity of doxorubicin has been associated with its ability to undergo enzymatic activation and to form hydroxyl (OH) radicals in this cell line, we also quantitated the OH formation in the BSO-treated and untreated MCF-7/ADRR cells using electron spin resonance spintrapping techniques. These results show that doxorubicin stimulated at least 2-fold more OH formation in the tumor cells after GSH levels were decreased by 90%. These results indicate that GSH plays an important role in modulating doxorubicin-induced OH formation via the scavenging of hydrogen peroxide by glutathione peroxidase and thus partially protects MCF-7/ADRR cells from the cytotoxic effect of doxorubicin.


Subject(s)
Breast Neoplasms/pathology , Doxorubicin/pharmacology , Methionine Sulfoximine/analogs & derivatives , Buthionine Sulfoximine , Cell Line/drug effects , Cell Survival/drug effects , Drug Resistance , Drug Synergism , Electron Spin Resonance Spectroscopy , Free Radicals , Glutathione Peroxidase/metabolism , Humans , Hydrogen Peroxide/metabolism , Methionine Sulfoximine/pharmacology
10.
Cancer Res ; 49(14): 3844-8, 1989 Jul 15.
Article in English | MEDLINE | ID: mdl-2544260

ABSTRACT

Previous studies with Adriamycin-sensitive and -resistant (ADRR) MCF-7 human breast tumor cell lines indicated that Adriamycin formed significantly less hydroxyl radical (.OH) as the result of enhanced detoxification of reactive oxygen intermediates in the ADRR cell line. In order to further define the sites of drug activation and the role of detoxification mechanisms in free radical levels, subcellular fractions were isolated from these two cell lines and free radical formation in the presence of Adriamycin was examined by using electron spin resonance spectroscopy. Studies reported here show that considerable NADPH-cytochrome P-450 reductase and NADH dehydrogenase activities were present in microsomes and mitochondria, respectively, and in nuclei obtained from these cells, and the relative activity of NADH dehydrogenase was 2-fold higher in the mitochondrial fraction of ADRR cells compared to the mitochondrial fraction from the parental wild type cells. In the presence of Adriamycin and a reducing cofactor (NADPH or NADH), Adriamycin semiquinone free radical, superoxide anion, and .OH were detected in all these fractions. Although only a small difference in the relative amount of oxy radical formation was detected in tumor microsomes, both mitochondria and nuclei of ADRR cells showed an overall 2-fold decreased formation of oxy radicals. The formation of the free radicals was significantly inhibited by superoxide dismutase, catalase, and dimethyl sulfoxide, indicating that free .OH generation was both superoxide and hydrogen peroxide dependent. The addition of purified glutathione peroxidase likewise inhibited .OH formation in a dose-dependent fashion. Similarly, when the lysate from ADRR cells, which contains 12- to 14-fold more glutathione peroxidase than Adriamycin-sensitive cells, was added to reaction mixtures containing Adriamycin-sensitive cells and Adriamycin, the .OH formation was diminished. Decreased free radical formation in nuclei and mitochondria, as a result of detoxification of hydrogen peroxide by glutathione peroxidase, may be significant in the protection of ADRR cells from Adriamycin-induced cell killing.


Subject(s)
Breast Neoplasms/metabolism , Doxorubicin/metabolism , Glutathione Peroxidase/metabolism , Tumor Cells, Cultured/metabolism , Biotransformation , Cell Line , Cell Nucleus/enzymology , Drug Resistance , Electron Spin Resonance Spectroscopy , Female , Free Radicals , Humans , Microsomes/enzymology , Mitochondria/enzymology , NADH Dehydrogenase/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism
11.
Cancer Res ; 55(20): 4623-6, 1995 Oct 15.
Article in English | MEDLINE | ID: mdl-7553639

ABSTRACT

Taxol stabilizes microtubules, prevents tubulin depolymerization, and promotes tubulin bundling and is one of the most effective drugs for the treatment of metastatic breast and ovarian cancer. Although its interaction with tubulin has been well characterized, the mechanism by which taxol induces growth arrest and cytotoxicity is not well understood. Herein, we show that taxol induced dose- and time-dependent accumulation of the cyclin inhibitor p21WAF1 in both p53 wild-type and p53-null cells, although the degree of induction was greater in cells expressing wild-type p53. In MCF7 cells, wild-type p53 protein was also induced after taxol treatment, and this induction was mediated primarily by increased protein stability. Taxol induced both p21WAF1 and wild-type p53 optimally in MCF7 cells after 20-24-h exposure with an EC50(3) of 5 nM. In p53-null PC3M cells, p21WAF1 was similarly induced after 24-h exposure to taxol. Coincident with these biochemical effects, taxol altered the electrophoretic mobility of c-raf-1 and stimulated mitogen activated protein kinase. Previous depletion of c-raf-1 inhibited both the p21WAF1- and p53-inducing properties of taxol, as well as the activation of MAP kinase. These data suggest that induction of p21WAF1 by taxol requires c-raf-1 activity, but that it is not strictly dependent on wild-type p53. Furthermore, the ability of taxol to both induce wild-type p53 in MCF7 cells and activate MAP kinase is also dependent on c-raf-1 expression.


Subject(s)
Cyclins/biosynthesis , Paclitaxel/pharmacology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Tumor Suppressor Protein p53/biosynthesis , 3T3 Cells , Animals , Benzoquinones , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic , Mice , Microtubules/drug effects , Proto-Oncogene Proteins c-raf , Quinones/pharmacology , Signal Transduction/drug effects , Tumor Cells, Cultured
12.
Biochim Biophys Acta ; 1180(3): 231-5, 1993 Jan 22.
Article in English | MEDLINE | ID: mdl-8422427

ABSTRACT

Several possible mechanisms of the synergistic interactions of IL-1 alpha and VP-16 against A375-C6 human melanoma cells were investigated. Studies indicate that IL-1 alpha did not increase topoisomerase II-dependent VP-16-mediated DNA damage, nor did IL-1 alpha inhibit the repair of VP-16-induced DNA damage in these cells. Furthermore, IL-1 alpha by itself or in combination with VP-16 did not cause significant fragmentation of cellular DNA into oligomers, indicating programmed cell death (apoptosis) was not involved in the mechanism of synergy. In contrast, an IL-1-specific receptor antagonist significantly decreased IL-1 alpha toxicity toward the melanoma cells and nearly eliminated the synergistic interactions of IL-1 alpha with VP-16. These results strongly indicate that synergism of IL-1 alpha with VP-16 was dependent upon an IL-1-receptor-mediated processes. DNA-strand breakage was unlikely to be a primary intracellular target for IL-1 alpha cytotoxicity and synergism with VP-16.


Subject(s)
DNA Damage , Etoposide/toxicity , Interleukin-1/toxicity , Cytotoxins/metabolism , Cytotoxins/toxicity , DNA/drug effects , DNA Repair/drug effects , Drug Synergism , Etoposide/metabolism , Humans , Interleukin-1/metabolism , Melanoma , Receptors, Interleukin-1/metabolism , Tumor Cells, Cultured
13.
Cell Death Differ ; 8(12): 1182-96, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11753566

ABSTRACT

Enzymatic deubiquitination of mono-ubiquitinated nucleosomal histone H2A (uH2A) and H2B (uH2B) is closely associated with mitotic chromatin condensation, although the function of this histone modification in cell division remains ambiguous. Here we show that rapid and extensive deubiquitination of nucleosomal uH2A occurs in Jurkat cells undergoing apoptosis initiated by anti-Fas activating antibody, staurosporine, etoposide, doxorubicin and the proteasome inhibitor, N-acetyl-leucyl-leucyl-norlucinal. These diverse apoptosis inducers also promoted the accumulation of slowly migrating, high molecular weight ubiquitinated proteins and depleted the cellular pool of unconjugated ubiquitin. In apoptotic cells, ubiquitin was cleaved from uH2A subsequent to the appearance of plasma membrane blebbing, and deubiquitination of uH2A closely coincided with the onset of nuclear pyknosis and chromatin condensation. Nucleosomal uH2A deubiquitination, poly (ADP-ribose)polymerase (PARP) cleavage and chromatin condensation were prevented in cells challenged with apoptosis inducers by pretreatment with the pan-caspase inhibitor, zVAD-fmk, or by over-expressing anti-apoptotic Bcl-xL protein. These results implicate a connection between caspase cascade activation and nucleosomal uH2A deubiquitination. Transient transfection of 293 cells with the gene encoding Ubp-M, a human deubiquitinating enzyme, promoted uH2A deubiquitination, while an inactive mutated Ubp-M enzyme did not. However, Ubp-M-promoted deubiquitination of uH2A was insufficient to initiate apoptosis in these cells. We conclude that uH2A deubiquitination is a down-stream consequence of procaspase activation and that unscheduled cleavage of ubiquitin from uH2A is a consistent feature of the execution phase of apoptosis rather than a determining or initiating apoptogenic event. Nucleosomal uH2A deubiquitination may function as a cellular sensor of stress in situations like apoptosis through which cells attempt to preserve genomic integrity.


Subject(s)
Apoptosis/physiology , Caspases/pharmacology , Chromatin/physiology , Histones/drug effects , Histones/metabolism , Nucleosomes/metabolism , Ubiquitins/drug effects , Ubiquitins/metabolism , Caspase 3 , Caspases/metabolism , Cell Membrane/chemistry , Cells, Cultured , Doxorubicin/pharmacology , Gene Expression , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transfection , bcl-X Protein
14.
J Immunother (1991) ; 10(3): 165-73, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1651105

ABSTRACT

To evaluate whether cells selected for doxorubicin resistance were cross-resistant to tumor necrosis factor, the effects of doxorubicin and recombinant human tumor necrosis factor-alpha (TNF) on doxorubicin-sensitive (WT) and 40-fold doxorubicin-resistant (40F) MCF-7 cell proliferation were assessed. The median dose (MD) for doxorubicin was 14.5 nM for WT cells and 474 nM for 40F cells. The MD for TNF was 0.18 nM for WT cells, while 40F cells were highly resistant to TNF concentrations up to 60 nM. Doxorubicin and TNF in combination were synergistic against WT cells, but not 40F cells. Glutathione depletion by buthionine sulfoxamine sensitized WT cells threefold to TNF, with no change in their response to doxorubicin, while 40F cells showed a twofold increase in doxorubicin sensitivity, with no apparent change in their resistance to TNF. No significant differences in TNF receptor number, Kd, or capacity for TNF internalization were noted between the two cell types. WT cells produced a single 15 kDa TNF degradation product, while the 40F cells produced three lower molecular weight degradation products. We conclude that cross-resistance to TNF in doxorubicin-resistant MCF-7 cells may be explained in part by altered TNF degradation.


Subject(s)
Doxorubicin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Buthionine Sulfoximine , Drug Resistance/physiology , Drug Screening Assays, Antitumor , Drug Synergism , Glutathione/metabolism , Humans , Methionine Sulfoximine/analogs & derivatives , Methionine Sulfoximine/pharmacology , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor , Tumor Cells, Cultured
15.
Free Radic Biol Med ; 8(6): 567-81, 1990.
Article in English | MEDLINE | ID: mdl-2113883

ABSTRACT

Certain anticancer agents form free radical intermediates during enzymatic activation. Recent studies have indicated that free radicals generated from adriamycin and mitomycin C may play a critical role in their toxicity to human tumor cells. Furthermore, it is becoming increasingly apparent that reduced drug activation and or enhanced detoxification of reactive oxygen species may be related to the resistance to these anticancer agents by certain tumor cell lines. The purposes of this review are to summarize the evidence pointing toward the significance of free radicals formation in drug toxicity and to evaluate the role of decreased free radical formation and enhanced free radical scavenging and detoxification in the development of anticancer drug resistance by a spectrum of tumor cell types. Studies failing to support the participation of oxyradicals in the cytotoxicity and resistance of adriamycin are also discussed.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance , Free Radicals , Neoplasms/drug therapy , Oxygen/pharmacology , Animals , Doxorubicin/pharmacology , Humans , Mitomycin , Mitomycins/pharmacology , Neoplasms/metabolism
16.
Biochem Pharmacol ; 45(10): 2099-107, 1993 May 25.
Article in English | MEDLINE | ID: mdl-8512591

ABSTRACT

Interleukin-1 alpha (IL-1 alpha) exerts antiproliferative effects on a human ovarian carcinoma cell line, NIH:OVCAR-3, which is resistant to clinically relevant concentrations of doxorubicin (DOX) and other chemotherapeutic agents. This action of IL-1 alpha depends on the presence of type I (80 kDa) receptors, although no quantitative relationship has been established between receptor occupancy and inhibition of cell growth. When NIH:OVCAR-3 cells were exposed to IL-1 alpha and DOX in combination, a mutual potentiation of the antiproliferative effects of the two agents was observed. This synergistic effect was not due to IL-1 receptor expression up-regulation by DOX, and receptor-dependent internalization of the cytokine was also unaffected. The involvement of IL-1 receptors is supported by the observation that synergism between the two agents was diminished (but not abolished) in the presence of a specific IL-1 receptor antagonist at concentrations blocking more than 75% of IL-1 alpha binding. DOX was found to significantly increase IL-1 alpha accumulation by NIH:OVCAR-3 cells after long-term (48 hr) exposure to the cytokine at 37 degrees, which might be due to increased nonspecific fluid phase uptake or to interference with cytokine degradation and/or release processes. The potent synergy of IL-1 alpha and DOX against ovarian carcinoma cells in vitro suggests that this drug combination may be effective against this disease in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ovarian Neoplasms/drug therapy , Cell Division/drug effects , Doxorubicin/administration & dosage , Doxorubicin/pharmacology , Drug Interactions , Drug Synergism , Female , Humans , Interleukin-1/administration & dosage , Interleukin-1/metabolism , Interleukin-1/pharmacokinetics , Intracellular Fluid/metabolism , Iodine Radioisotopes , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Binding , Receptors, Interleukin-1/drug effects , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1/physiology , Tumor Cells, Cultured/drug effects , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Biochem Pharmacol ; 31(5): 805-14, 1982 Mar 01.
Article in English | MEDLINE | ID: mdl-7082349

ABSTRACT

In vitro experiments were performed to evaluate the capacity of the redox cycling compounds mitomycin C (MC), nitrofurantoin (NF) and paraquat (PQ) to stimulate pulmonary microsomal lipid peroxidation. It was observed that the interaction of MC, NF or PQ with rat or mouse lung microsomes in the presence of an NADPH-generating system and an O2 atmosphere resulted in significant lipid peroxidation. All three compounds demonstrated similar concentration dependency, similar time courses and the ability to generate lipid peroxidation-associated chemiluminescence. The stimulation of lipid peroxidation by MC, NF or PQ was inhibited significantly by superoxide dismutase, glutathione, ascorbic acid, catalase and EDTA, agents which either scavenge reactive oxygen and/or prevent the generation of secondary reactive oxygen metabolites. In addition, the ability of MC or NF, but not PQ, to stimulate lipid peroxidation was reduced significantly following preincubation with microsomes and NADPH under N2 (15-20 min) prior to incubation under O2. During this period under N2. MC and NF underwent reductive metabolism of their quinone and nitro moieties respectively. Thus, it appears that under aerobic conditions the pulmonary microsomal-mediated redox cycling of MC, NF and PQ is accompanied by the stimulation of reactive oxygen-dependent lipid peroxidation.


Subject(s)
Lipid Peroxides/metabolism , Lung/metabolism , Microsomes/metabolism , Mitomycins/toxicity , Nitrofurantoin/toxicity , Oxygen/pharmacology , Paraquat/toxicity , Animals , In Vitro Techniques , Luminescent Measurements , Lung/drug effects , Male , Mice , Mice, Inbred Strains , Rats , Rats, Inbred Strains
18.
Biochem Pharmacol ; 34(6): 847-56, 1985 Mar 15.
Article in English | MEDLINE | ID: mdl-3977958

ABSTRACT

Mitochondrial degeneration is a consistently prominent morphological alteration associated with adriamycin toxicity which may be the consequence of adriamycin-enhanced peroxidative damage to unsaturated mitochondrial membrane lipids. Using isolated rat liver mitochondria as an in vitro model system to study the effects of the anticancer drug adriamycin on lipid peroxidation, we found that NADH-dependent mitochondrial peroxidation--measured by the 2-thiobarbituric acid method--was stimulated by adriamycin as much as 4-fold. Marker enzyme analysis indicated that the mitochondria were substantially free of contaminating microsomes (less than 5%). Lipid peroxidation in mitochondria incubated in KCl-Tris-HCl buffer (pH 7.4) under an oxygen atmosphere was optimal at 1-2 mg of mitochondrial protein/ml and with NADH at 2.5 mM. Malonaldehyde production was linear with time to beyond 60 min, and the maximum enhancement of peroxidation was observed with adriamycin at 50-100 microM. Interestingly, in contrast to its stimulatory effect on NADH-supported mitochondrial peroxidation, adriamycin markedly diminished ascorbate-promoted lipid peroxidation in mitochondria. Superoxide dismutase, catalase, 1,3-dimethylurea, reduced glutathione, alpha-tocopherol and EDTA added to incubation mixtures inhibited endogenous and adriamycin-augmented NADH-dependent peroxidation of mitochondrial lipids, indicating that multiple species of reactive oxygen (superoxide anion radical, hydrogen peroxide and hydroxyl radical) and possibly trace amounts of endogenous ferric iron participated in the peroxidation reactions. In submitochondrial particles freed of endogenous defenses against oxyradicals, lipid peroxidation was increased 7-fold by adriamycin. These observations suggest that some of the effects of adriamycin on mitochondrial morphology and biochemical function may be mediated by adriamycin-enhanced reactive oxygen-dependent mitochondrial lipid peroxidation.


Subject(s)
Doxorubicin/pharmacology , Lipid Peroxides/metabolism , Membrane Lipids/metabolism , Mitochondria/metabolism , Oxygen/metabolism , Animals , Ascorbic Acid/pharmacology , Free Radicals , In Vitro Techniques , Male , Mitochondria/drug effects , NAD/pharmacology , Rats , Rats, Inbred Strains
19.
Biochem Pharmacol ; 35(23): 4327-35, 1986 Dec 01.
Article in English | MEDLINE | ID: mdl-3024646

ABSTRACT

Adriamycin causes both glomerular and tubular lesions in kidney, which can be severe enough to progress to irreversible renal failure. This drug-caused nephrotoxicity may result from the metabolic reductive activation of Adriamycin to a semiquinone free radical intermediate by oxidoreductive enzymes such as NADPH-cytochrome P-450 reductase and NADH-dehydrogenase. The drug semiquinone, in turn, autoxidizes and efficiently generates highly reactive and toxic oxyradicals. We report here that the reductive activation of Adriamycin markedly enhanced both NADPH- and NADH-dependent kidney microsomal membrane lipid peroxidation, measured as malonaldehyde by the thiobarbituric acid method. Adriamycin-enhanced kidney microsomal lipid peroxidation was diminished by the inclusion of the oxyradical scavengers, superoxide dismutase and 1,3-dimethylurea, and by the chelating agents, EDTA and diethylenetriamine-pentaacetic acid (DETPAC), implicating an obligatory role for reactive oxygen species and metal ions in the peroxidation mechanism. Furthermore, the inclusion of exogenous ferric and ferrous iron salts more than doubled Adriamycin-stimulated peroxidation. Lipid peroxidation was prevented by the sulfhydryl-reacting agent, p-chloromercuribenzenesulfonic acid, by omitting NAD(P)H, or by heat-inactivating the kidney microsomes, indicating the requirement for active pyridine-nucleotide linked enzymes. Several analogs of Adriamycin as well as mitomycin C, drugs which are capable of oxidation-reduction cycling, greatly increased NADPH-dependent kidney microsomal peroxidation. Carminomycin and 4-demethoxydaunorubicin were noteworthy in this respect because they were three to four times as potent as Adriamycin. In isolated kidney mitochondria, Adriamycin promoted a 12-fold increase in NADH-supported (NADH-dehydrogenase-dependent) peroxidation. These observations clearly indicate that anthracyclines enhance oxyradical-mediated membrane lipid peroxidation in vitro, and suggest that peroxidation-caused damage to kidney endoplasmic reticulum and mitochondrial membranes in vivo could contribute to the development of anthracycline-caused nephrotoxicity.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Kidney/drug effects , Lipid Peroxides/metabolism , Membrane Lipids/metabolism , Animals , Catalase/pharmacology , Chelating Agents/pharmacology , Doxorubicin/toxicity , Hydroxides , Hydroxyl Radical , Kidney/metabolism , Male , Mice , Mice, Inbred Strains , NADP/metabolism , Naphthacenes/toxicity , Oxidation-Reduction , Rats , Rats, Inbred Strains
20.
Biochem Pharmacol ; 31(4): 499-502, 1982 Feb 15.
Article in English | MEDLINE | ID: mdl-6802138

ABSTRACT

Previous work revealed that portacaval anastomosis (PCA) in rats results in hepatic atrophy and marked decreases in components of the microsomal monooxygenase system such as cytochrome P-450. In the present study, the effects of PCA on hepatic monooxygenase activity were studied in more detail. We report that PCA, in general, produces effects resembling those of castration. Thus, in male rats, PCA depressed the activity of highly sex-dependent enzymes such as ethylmorphine and aminopyrine demethylases. Similar effects were produced by castration, and the combination of PCA and castration produced the same effect as either treatment alone. In male rats, non-sex-dependent enzymes such as aniline hydroxylase and NADPH-cytochrome c reductase were unaffected by either PCA or castration. By contrast, in female rats, neither PCA nor castration significantly affected microsomal monooxygenase activities. In male rats, PCA was accompanied by a 75% reduction in serum testosterone levels and a 6-fold increase in total estrogen levels. We conclude that these effects of PCA in male rats were due, in large measure, to a demasculinizing effect.


Subject(s)
Microsomes, Liver/enzymology , Oxidoreductases/metabolism , Portacaval Shunt, Surgical , Animals , Castration , Cytochromes/metabolism , Estrogens/blood , Female , Male , Mixed Function Oxygenases/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Oxidoreductases, N-Demethylating/metabolism , Rats , Rats, Inbred Strains , Sex Factors , Testosterone/blood
SELECTION OF CITATIONS
SEARCH DETAIL