Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Pharmacol Toxicol ; 69(6): 400-9, 1991 Dec.
Article in English | MEDLINE | ID: mdl-1766914

ABSTRACT

In the female Sprague-Dawley rat uterus 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and related compounds exhibited a broad spectrum of antioestrogenic responses. For example 2,3,7,8-TCDD inhibited the 17 beta-oestradiol-induced uterine wet weight increase, peroxidase activity, oestrogen and progesterone receptor levels, epidermal growth factor (EGF) receptor binding, and EGF receptor and c-fos protooncogene mRNA levels. The aryl hydrocarbon (Ah) receptor was identified in the rat uterus and the antioestrogenic activities of TCDD and related compounds were structure-dependent. In parallel studies, the effects of TCDD as an antioestrogen in MCF-7 human breast cancer cells was also investigated. TCDD inhibited the 17 beta-oestradiol-induced proliferation of these cells and the secretion of the 34-, 52- and 160-kDa proteins. Treatment of MCF-7 cells with 1 nM [3H]-17 beta-oestradiol resulted in a rapid accumulation of nuclear oestrogen receptor (ER) complexes. Pretreatment of the cells with TCDD caused a rapid decrease in nuclear ER binding activity and immunoreactive protein; moreover, the structure-dependent potencies of TCDD and related compounds as antioestrogens were similar to their Ah receptor binding affinities. TCDD also caused a decrease in nuclear ER levels in wild-type Ah-responsive Hepa 1c1c7 cells but was inactive in Ah non-responsive mutant Hepa 1c1c7 cells. Moreover, in the wild-type cells, both actinomycin D and cycloheximide blocked the effects of TCDD. 6-Methyl-1,3,8-trichlorodibenzofuran (MCDF) has previously been characterized as a TCDD antagonist in rodents and in transformed rodent cell lines. However, like TCDD, MCDF also exhibited a broad spectrum of antioestrogenic activities in both the female Sprague-Dawley rat uterus and MCF-7 cells. MCDF is relatively non-toxic compared to TCDD and is being investigated as a compound which may be clinically useful for the treatment of mammary cancer.


Subject(s)
Estrogen Antagonists/pharmacology , Polychlorinated Dibenzodioxins/pharmacology , Animals , Female , Humans , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred Strains , Tumor Cells, Cultured
3.
Toxicol Lett ; 56(3): 305-15, 1991 May.
Article in English | MEDLINE | ID: mdl-1903567

ABSTRACT

Acute administration of 17 beta-estradiol (5 micrograms/rat) to 25-day-old female Sprague-Dawley rats resulted in an increase of uterine mRNA for the cellular oncogene c-fos. The c-fos mRNA levels were significantly elevated 12 and 24 h after exposure to the hormone (232 and 164% of control values) and the elevation was not observed after 48 h. In contrast, treatment of the animals with either 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 6-methyl-1,3,8-trichlorodibenzofuran (MCDF) resulted in a dose-dependent decrease in constitutive uterine c-fos mRNA levels. In rats co-treated with 17 beta-estradiol plus TCDD or MCDF, it was apparent from the results that the halogenated aromatic hydrocarbons significantly inhibited the estrogen-induced increases in uterine c-fos mRNA levels. These observations further extend the diverse spectrum of antiestrogenic effects caused by TCDD and related compounds and also show an interaction between TCDD and the constitutive expression of the c-fos proto-oncogene in the female rat uterus.


Subject(s)
Estradiol/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogenes/genetics , Uterus/drug effects , Animals , Benzofurans/pharmacology , Dose-Response Relationship, Drug , Female , Gene Expression/drug effects , Gene Expression/genetics , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-fos , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Time Factors , Uterus/metabolism , Uterus/physiology
4.
Toxicology ; 69(2): 187-97, 1991.
Article in English | MEDLINE | ID: mdl-1949047

ABSTRACT

The comparative antiestrogenic effects of 6-methyl-1,3,8-trichlorodibenzofuran (MCDF), 6-t-butyl-1,3,8-trichlorodibenzofuran (triCDF) and 6-cyclohexyl-1,3,8-triCDF were determined in immature female Sprague-Dawley rats. Treatment of the animals with 17 beta-estradiol (0.33 mumol/kg X 2) caused an increase in uterine cytosolic and nuclear estrogen and progesterone receptor levels, uterine peroxidase activity, uterine wet weights and uterine epidermal growth factor (EGF) receptor binding activity and steady state EGF receptor mRNA levels. MCDF and 6-t-butyl-1,3,8-triCDF, two compounds which exhibit moderate aryl hydrocarbon (Ah) receptor binding affinity were also administered (100 mumol/kg) to the female rats in the presence or absence of 17 beta-estradiol. The results of these studies show that both compounds decrease the constitutive and 17 beta-estradiol-induced responses noted above. In contrast, 6-cyclohexyl-1,3,8-triCDF, a congener which exhibits low Ah receptor binding, was inactive as an antiestrogen. These studies clearly demonstrate that selected 6-alkyl-1,3,8-triCDFs elicit a broad spectrum of antiestrogenic activity in immature female rats. Moreover, in contrast to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) which also is a potent antiestrogen, the 6-alkyl-1,3,8-triCDFs are relatively non-toxic and can serve as prototypes for the future development of a new class of antiestrogens with potential for clinical applications.


Subject(s)
Benzofurans/pharmacology , Estrogen Antagonists/pharmacology , Animals , Cell Nucleus/metabolism , Cytosol/metabolism , Estradiol/pharmacology , Female , Rats , Rats, Inbred Strains , Receptors, Estrogen/metabolism , Receptors, Estrogen/physiology , Receptors, Progesterone/drug effects , Receptors, Progesterone/metabolism
5.
Arch Biochem Biophys ; 284(1): 193-200, 1991 Jan.
Article in English | MEDLINE | ID: mdl-1846513

ABSTRACT

6-Methyl-8-iodo-1,3,-dichlorodibenzofuran (I-MCDF) and its radiolabeled analog [125I]MCDF have been synthesized and used to investigate the mechanism of action of 1,3,6,8-substituted dibenzofurans as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) antagonists. Like 6-methyl-1,3,8-trichlorodibenzofuran (MCDF), I-MCDF partially antagonized the induction by TCDD of microsomal aryl hydrocarbon hydroxylase (AHH) and ethoxyresorufin O-deethylase (EROD) activities in rat hepatoma H-4-II E cells and male Long-Evans rat liver. Incubation of rat liver cytosol with [125I]MCDF followed by velocity sedimentation analysis on sucrose gradients gave a specifically bound peak which sedimented at 9.6 S. This radioactive peak was displaced by coincubation with a 200-fold excess of unlabeled I-MCDF, 6-methyl-1,3,8-trichlorodibenzofuran (MCDF), 2,3,7,8-tetrachlorodibenzofuran (TCDF), and benzo [a]pyrene. Based on the velocity sedimentation results and the elution profile from a Sephacryl S-300 gel permeation column, the Stokes radius and apparent molecular weights of the cytosolic [125I]MCDF-Ah receptor complex were 6.5 nm and 259,200, respectively. In addition, the nuclear [125I]MCDF-receptor complex eluted at a salt concentration of 0.29 M KCl from a DNA-Sepharose column. Velocity sediment analysis of the nuclear [125I]MCDF-Ah receptor complex from rat hepatoma H-4-II E cells gave a specifically bound peak at 5.6 +/- 0.8 S. All of these properties were similar to those observed using [3H]TCDD as the radioligand. In addition, there were several ligand-dependent differences observed in the properties of the I-MCDF and TCDD receptor complexes; for example, the [125I]MCDF rat cytosolic receptor complex was unstable in high salt buffer and was poorly transformed into a form with increased binding affinity on DNA-Sepharose columns; Scatchard plot analysis of the saturation binding of [3H]TCDD and [125I]MCDF with rat hepatic cytosol gave KD values of 1.07 and 0.13 nM and Bmax values of 137 and 2.05 fmol/mg protein, respectively. The nuclear extract from rat hepatoma H-4-II E cells treated with I-MCDF or TCDD interacted with a dioxin-responsive element in a gel retardation assay. These results suggest that the mechanism of antagonism may be associated with competition of the antagonist receptor complex for nuclear binding sites.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Benzofurans/metabolism , DNA-Binding Proteins/metabolism , Polychlorinated Dibenzodioxins/antagonists & inhibitors , Transcription Factors/metabolism , Animals , Base Sequence , Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , Enzyme Induction/drug effects , Kinetics , Microsomes, Liver/enzymology , Molecular Sequence Data , Oligonucleotides/metabolism , Oxidoreductases/metabolism , Polychlorinated Dibenzodioxins/pharmacology , Protein Binding , Rats , Receptors, Aryl Hydrocarbon , Receptors, Drug/metabolism , Regulatory Sequences, Nucleic Acid
6.
Biotechnol Appl Biochem ; 12(6): 621-9, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2128736

ABSTRACT

Adriamycin was internalized in canine red blood cells (RBC) by two procedures involving (a) simple diffusion of the drug into cells and (b) hypotonic dialysis followed by isotonic resealing. The two procedures yielded comparable amounts of encapsulated adriamycin, around 35 micrograms/10(9) RBC. Exposure of adriamycin-loaded RBC to 0.16% glutaraldehyde consistently slowed down the rate of efflux of the drug as compared with non-glutaraldehyde-treated cells: after 1 h of incubation at 37 degrees C, greater than 80% of adriamycin was still present inside the glutaraldehyde-treated RBC, while at 24 h it was 66%, compared to 10% and 1%, respectively, in the adriamycin-loaded, non-glutaraldehyde-treated cells. Canine RBC showed a higher rate of transformation of adriamycin than the human cells, the only intracellular metabolite being adriamycinol, which is apparently formed by the NADPH-dependent enzyme aldehyde reductase. Production of adriamycinol was remarkably lower in the glutaraldehyde-treated RBC, as a result of progressive and extensive inactivation of hexose monophosphate shunt activity responsible for NADPH formation. These results, coupled with the known selective targeting of glutaraldehyde-treated RBC to liver, hold promise as to in vivo applications of this drug delivery system in antineoplastic therapy.


Subject(s)
Doxorubicin/administration & dosage , Animals , Biotransformation , Chromatography, High Pressure Liquid , Dogs , Doxorubicin/blood , Drug Carriers , Erythrocyte Membrane , Erythrocytes/metabolism , Glutaral , Hexosephosphates/blood , In Vitro Techniques , Osmotic Fragility/drug effects
7.
Mol Cell Endocrinol ; 72(3): 247-52, 1990 Sep 10.
Article in English | MEDLINE | ID: mdl-2289633

ABSTRACT

Treatment of immature female Sprague-Dawley rats with 17 beta-estradiol (5 micrograms/animal) resulted in an increase in uterine epidermal growth factor (EGF) receptor binding activity. Moreover, in a separate study it was also shown that 17 beta-estradiol increased steady-state levels of rat uterine EGF receptor mRNA as determined by Northern analysis. In contrast, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) caused a dose-response decrease in constitutive rat uterine EGF receptor binding activity and this was paralleled by a decrease in steady-state levels of uterine EGF receptor mRNA. Cotreatment of the animals with both TCDD (16 nmol/kg) and 17 beta-estradiol (5 micrograms/rat) gave results which showed that TCDD significantly inhibited the estrogen-induced increases in rat uterine EGF receptor binding activity and EGF receptor mRNA levels. These results further extend the range of antiestrogenic properties of TCDD and suggest that the inhibition of growth factor expression may play a role in the growth-inhibiting properties of TCDD in estrogen-responsive tissues or cells.


Subject(s)
Epidermal Growth Factor/metabolism , ErbB Receptors/drug effects , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Polychlorinated Dibenzodioxins/pharmacology , Uterus/drug effects , Animals , Depression, Chemical , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation/drug effects , RNA, Messenger/biosynthesis , Rats , Rats, Inbred Strains , Recombinant Proteins/biosynthesis , Uterus/metabolism
8.
Biochem Pharmacol ; 39(3): 485-8, 1990 Feb 01.
Article in English | MEDLINE | ID: mdl-2154986

ABSTRACT

Treatment of 25-day-old female Sprague-Dawley rats with 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) significantly lowered constitutive uterine peroxidase activity and decreased uterine wet weights in a dose-response fashion. In cotreatment studies with 17 beta-estradiol, 2,3,7,8-TCDD antagonized the increase in uterine peroxidase activity and uterine wet weights, and these effects persisted for up to 156 hr. In the rat uterus, the antiestrogenic affects of two potent Ah receptor agonists, 2,3,7,8-TCDD and 2,3,4,7,8-pentachlorodibenzofuran, were comparable at a dose of 80 micrograms/kg, whereas the weaker Ah receptor agonist, 1,2,4,7,8-pentachlorodibenzo-p-dioxin, was relatively inactive at this dose. These results show that 2,3,7,8-TCDD antagonizes a well-characterized estrogen-induced response (uterine peroxidase activity), and the structure-activity data suggest that the Ah receptor is involved in mediating the antiestrogenic responses in target cells/organs.


Subject(s)
Dioxins/pharmacology , Estrogen Antagonists/pharmacology , Peroxidase/antagonists & inhibitors , Polychlorinated Dibenzodioxins/pharmacology , Uterus/enzymology , Animals , Benzofurans/pharmacology , Estradiol/pharmacology , Female , Kinetics , Organ Size/drug effects , Polychlorinated Dibenzodioxins/analogs & derivatives , Rats , Rats, Inbred Strains , Receptors, Aryl Hydrocarbon , Receptors, Drug/physiology , Structure-Activity Relationship , Uterus/anatomy & histology , Uterus/drug effects
9.
Toxicology ; 59(3): 285-96, 1989 Dec 15.
Article in English | MEDLINE | ID: mdl-2631297

ABSTRACT

The activities of several 6-substituted-1,3,8-trichlorodibenzofurans (CDFs) as partial antagonists of the induction of hepatic microsomal aryl hydrocarbon hydroxylase (AHH) and ethoxyresorufin O-deethylase (EROD) activities in the rat by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) were structure-dependent. Treatment of the rats with TCDD (16 nmol/kg), the 6-substituted-1,3,8-triCDFs (50 mumol/kg) and TCDD plus the 6-substituted-1,3,8-triCDFs showed that most of the substituted congeners were either inactive (6-methyl, ethyl, propyl, i-propyl, t-butyl) or weak (6-cyclohexyl, nitro) inducers of AHH and EROD activities, whereas TCDD caused an 8.1- and 58-fold induction of these enzyme activities respectively. In the co-administration studies, the 6-methyl, propyl, ethyl, isopropyl and t-butyl analogs partially antagonized the induction of the monooxygenase enzyme activities by TCDD, whereas, the 6-cyclohexyl and 6-nitro-1,3,8-triCDFs exhibited minimal activity as TCDD antagonists. The Ah receptor binding affinities of the 6-substituted compounds were determined in a series of in vitro competitive binding studies using [3H]TCDD as the radioligand. Analysis of the data by Scatchard and Dixon plots showed that the avidities for the Ah receptor by the 6-substituted-1,3,8-triCDFs followed the order 6-methyl greater than 6-t-butyl greater than 6-i-propyl greater than 6-propyl approximately 6-ethyl greater than 6-cyclohexyl greater than 6-nitro-1,3,8-triCDF. In addition there was a good correlation between the in vitro binding avidities and Ki values for these compounds and their in vivo activity as partial antagonists of the induction of AHH and EROD activities by TCDD. The results suggested that the 6-substituted-1,3,8-triCDFs competitively displayed TCDD from the Ah receptor and this interaction may play a role in the mechanism of action of this class of TCDD antagonists.


Subject(s)
Benzofurans/pharmacology , Dioxins/antagonists & inhibitors , Polychlorinated Dibenzodioxins/antagonists & inhibitors , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Induction/drug effects , Male , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Oxidoreductases/biosynthesis , Rats , Structure-Activity Relationship
10.
Mol Pharmacol ; 35(5): 729-35, 1989 May.
Article in English | MEDLINE | ID: mdl-2542761

ABSTRACT

6-Methyl-1,3,8-trichlorodibenzofuran (MCDF) binds with moderate affinity to the aryl hydrocarbon (Ah) receptor protein (4.9 x 10(-8) M) but is a weak Ah receptor agonist. Cotreatment of male Long Evans rats with MCDF (50 mumol/kg) and a dose of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) that causes a near-maximal induction of hepatic microsomal aryl hydrocarbon hydroxylase and ethoxyresorufin O-deethylase activities resulted in a significant inhibition of these activities for up to 96 hr. Comparable results were obtained with MCDF (10(-7) M) and TCDD (10(-8) M) in rat hepatoma H-4-II E cells in culture over 36 hr. TCDD treatment of rats resulted in an initial decrease of hepatic cytosolic Ah receptor within 6 hr and this was followed by a subsequent 138% increase in cytosolic receptor levels 72 hr after treatment. Although MCDF (50 mumol/kg) did not significantly alter rat hepatic cytosolic Ah receptor levels in animals cotreated with TCDD plus MCDF, the latter compound significantly inhibited TCDD-mediated replenishment of the cytosolic Ah receptor. In contrast, treatment of rat hepatoma H-4-II E cells with TCDD (10(-8) M) resulted in the rapid (within 1 hr) depletion of cytosolic Ah receptor, which remained undetectable for up to 36 hr; cotreatment of the cells with MCDF (10(-7) M) and TCDD (10(-8) M) resulted in cytosolic Ah receptor levels that were similar to those observed after treatment with TCDD alone. The effects of MCDF on the uptake and persistence of nuclear [3H]TCDD-Ah receptor complex levels were also determined in rat liver and rat hepatoma H-4-II E cells in culture. MCDF did not significantly decrease levels of occupied nuclear Ah receptor complexes in the rat or rat hepatoma cells. Moreover, using the sucrose density gradient assay procedure, the sedimentation coefficients of the cytosolic and nuclear TCDD-Ah receptor complexes in the presence or absence of MCDF were comparable. The results of these and other related studies with 6-substituted-1,3,8-trichlorodibenzofurans suggest that MCDF may act as a partial TCDD antagonist by competing with TCDD for nuclear binding sites.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Benzofurans/pharmacology , Dioxins/antagonists & inhibitors , Polychlorinated Dibenzodioxins/antagonists & inhibitors , Animals , Enzyme Induction/drug effects , Male , Polychlorinated Dibenzodioxins/metabolism , Rats , Receptors, Aryl Hydrocarbon , Receptors, Drug/analysis , Receptors, Drug/drug effects
11.
Toxicology ; 54(2): 139-50, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2922766

ABSTRACT

6-Methyl-1,3,8-trichlorodibenzofuran (MCDF), 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and TCDD plus MCDF were administered to C57BL/6 mice and their effects on several aryl hydrocarbon (Ah) receptor-mediated responses including hepatic microsomal aryl hydrocarbon hydroxylase (AHH) and ethoxyresorufin O-deethylase (EROD) induction, immunotoxicity and teratogenicity were determined. MCDF did not induce hepatic microsomal AHH and EROD at doses up to 500 mumol/kg, however, co-administration of MCDF (50 mumol/kg) with a dose of TCDD which elicited a submaximal induction response (i.e. ED80-100, 15 nmol/kg) resulted in some small but significant inhibition of the induction of hepatic microsomal AHH and EROD (14 and 17%, respectively) compared to that observed with TCDD alone. Co-administration of TCDD and other doses of MCDF (10, 100, 200 or 500 mumol/kg) did not effect the induction response. These results were in contrast to the effectiveness of MCDF as an antagonist of the induction of AHH and EROD by TCDD in the rat (up to 50% inhibition of monooxygenase induction). Administration of MCDF (4, 20 and 40 mumol/kg) to C57BL/6 mice caused some inhibition of the splenic plaque-forming cell response to sheep erythrocytes only at the highest dose (26% decrease); the interaction of MCDF (4, 20 and 40 mumol/kg) and an immunotoxic dose of TCDD (3.7 nmol/kg) resulted in significant protection from the immunotoxic effects of TCDD at the 2 higher dose levels of MCDF. Similarly, MCDF (400 mumol/kg) did not cause cleft palate in mice but at this dose level MCDF afforded some protection from TCDD (20 micrograms/kg)-mediated cleft palate in mice. However, studies utilizing [3H]TCDD suggested that the protective effects may be due to modulation of TCDD reaching the palate in the co-treated animals (MCDF plus TCDD). Although both MCDF and Aroclor 1254 were both weak Ah receptor agonists in C57BL/6 mice, the former compound was much less effective as a TCDD antagonist. The observed species-specific effects for these 2 TCDD antagonists may be related species-dependent differences in receptor structure and receptor-ligand (i.e. agonist or antagonist) interactions.


Subject(s)
Benzofurans/pharmacology , Dioxins/antagonists & inhibitors , Polychlorinated Dibenzodioxins/antagonists & inhibitors , Abnormalities, Drug-Induced/etiology , Animals , Antibody Formation/drug effects , Aroclors/pharmacology , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Induction/drug effects , Male , Mice , Mice, Inbred C57BL , Oxidoreductases/biosynthesis , Species Specificity
12.
Toxicol Appl Pharmacol ; 95(3): 435-43, 1988 Sep 30.
Article in English | MEDLINE | ID: mdl-2847362

ABSTRACT

The ED50s for the dose-response induction of hepatic microsomal aryl hydrocarbon hydroxylase (AHH) and ethoxyresorufin O-deethylase (EROD) by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in the female Sprague-Dawley rat were 3.3 and 2.7 nmol/kg, respectively. In contrast, the corresponding ED50 values for induction by the nontoxic 6-methyl-1,3,8-trichlorodibenzofuran (MCDF) were 524 and 578 mumol/kg for AHH and EROD, respectively, and TCDD was greater than 1.5 X 10(5) more potent than MCDF as an agonist for this response. Cotreatment of the female rats with MCDF (20, 50, or 100 mumol/kg) and TCDD (6.4 nmol/kg) showed that MCDF partially antagonized the induction of AHH and EROD by TCDD and this corresponded with results previously reported in the male rat. Like TCDD, MCDF also caused a dose-response decrease in uterine and hepatic cytosolic and nuclear estrogen (ERc and ERn) and progesterone (PRc and PRn) receptor levels. The relative TCDD/MCDF potencies for the reduction of uterine ERc, ERn, PRc, and PRn levels were 293, 569, 560, and 459, respectively, and comparable potency ratios (693, 409, 405, and 424, respectively) were observed in the liver. Since MCDF was active as an antiestrogen at dose levels which caused only minimal induction of hepatic monooxygenases, it is unlikely that induction of these enzymes and the subsequent increased metabolism of estradiol play a role in the antiestrogenic effects of MCDF (or TCDD). The reasons for the differences in the relative potency of MCDF for the "traditional" Ah receptor-mediated response (i.e., AHH induction) and the modulation of steroid hormone receptor binding levels are unknown. MCDF, a compound which exhibits relatively high TCDD receptor binding activity and low toxicity, represents a new class of nontoxic halogenated aromatic antiestrogens that can be utilized to further probe the mechanism of this response in model systems.


Subject(s)
Benzofurans/pharmacology , Dioxins/pharmacology , Estrogen Antagonists/pharmacology , Polychlorinated Dibenzodioxins/pharmacology , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , Estradiol/pharmacology , Female , Mesothelin , Microsomes, Liver/enzymology , Rats , Rats, Inbred Strains , Receptors, Aryl Hydrocarbon , Receptors, Drug/metabolism , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
13.
Mol Pharmacol ; 33(2): 231-6, 1988 Feb.
Article in English | MEDLINE | ID: mdl-2828916

ABSTRACT

In addition to being one of the most toxic chemicals known, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is the most potent inducer of rat liver microsomal cytochrome P-4501A1 (P-450c). Previous studies have demonstrated that a high affinity, low capacity cytosolic receptor (the Ah receptor) mediates the activity of TCDD to induce cytochrome P-4501A1, which catalyzes benzo[a]pyrene hydroxylation [aryl hydrocarbon hydroxylase (AHH]) and 7-ethoxyresorufin O-dealkylation (EROD). The results of the present study indicate that 6-methyl-1,3,8-trichlorodibenzofuran (MCDF) effectively competes with [3H]TCDD for binding to the Ah receptor in rat liver cytosol. The concentration of MCDF effecting 50% displacement of [3H]TCDD was 4.9 X 10(-8) M, which is approximately 50 times greater than the EC50 for unlabeled TCDD (approximately 1 X 10(-9) M). However, in contrast to TCDD, MCDF was only a weak inducer of AHH and EROD activity in rat hepatoma H-4-II cells in culture. When co-incubated, MCDF diminished in a concentration-dependent manner the ability of TCDD to induce AHH and EROD activity in vitro. Treatment of rats with 20-200 mumol/kg MCDF in vivo had little or no effect on liver microsomal AHH and EROD activity, whereas treatment of rats with 16 nmol/kg TCDD caused a 6- and a 70-fold induction of AHH and EROD activity, respectively. When co-administered, MCDF diminished by approximately 50% the ability of TCDD to induce AHH and EROD activity in vivo. The partial antagonism produced by 50 mumol/kg MCDF could be partially overcome by doubling the dosage of TCDD from 16 to 32 nmol/kg. Immunochemical analysis of rat liver microsomes revealed that treatment of rats with 20-200 mumol/kg MCDF caused little or no induction of cytochromes P-4501A1 and P-4501A2 (P-450d), whereas these isozymes were induced 33- and 5-fold, respectively, in rats treated with 16 nmol/kg TCDD. When co-administered, MCDF diminished by approximately 50% the ability of TCDD to induce cytochrome P-4501A1 in vivo, which established that MCDF was not simply acting as an inhibitor of AHH and EROD activity. MCDF also antagonized the ability of TCDD to induce cytochrome P-4501A2, which suggests that the induction of both cytochromes P-4501A1 and P-4501A2 is regulated by the Ah receptor. These results indicate that MCDF binds with high affinity to the Ah receptor in rat liver cytosol and competitively blocks the binding of TCDD.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Benzofurans/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Isoenzymes/biosynthesis , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Cytochrome P-450 CYP1A1 , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/drug effects , Male , Oxidoreductases/biosynthesis , RNA, Messenger/analysis , Rats , Receptors, Aryl Hydrocarbon , Receptors, Drug/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...