Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Toxicol Appl Pharmacol ; 169(1): 102-13, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11076702

ABSTRACT

Efavirenz, a potent nonnucleoside reverse transcriptase inhibitor widely prescribed for the treatment of HIV infection, produces renal tubular epithelial cell necrosis in rats but not in cynomolgus monkeys or humans. This species selectivity in nephrotoxicity could result from differences in the production or processing of reactive metabolites, or both. A detailed comparison of the metabolites produced by rats, monkeys, and humans revealed that rats produce a unique glutathione adduct. The mechanism of formation and role of this glutathione adduct in the renal toxicity were investigated using both chemical and biochemical probes. Efavirenz was labeled at the methine position on the cyclopropyl ring with the stable isotope deuterium, effectively reducing the formation of the cyclopropanol metabolite, an obligate precursor to the glutathione adduct. This substitution markedly reduced both the incidence and severity of nephrotoxicity as measured histologically. Further processing of this glutathione adduct was also important in producing the lesion and was demonstrated by inhibiting gamma-glutamyltranspeptidase with acivicin pretreatment (10 mg/kg, IV) prior to dosing with efavirenz. Again, both the incidence and severity of the nephrotoxicity were reduced, such that four of nine rats given acivicin were without detectable lesions. These studies provide compelling evidence that a species-specific formation of glutathione conjugate(s) from efavirenz is involved in producing nephrotoxicity in rats. Mechanisms are proposed for the formation of reactive metabolites that could be responsible for the renal toxicity observed in rats.


Subject(s)
Anti-HIV Agents/metabolism , Glutathione/drug effects , Kidney Diseases/metabolism , Kidney Tubules/drug effects , Oxazines/metabolism , Reverse Transcriptase Inhibitors/metabolism , Alkynes , Animals , Benzoxazines , Cyclopropanes , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gas Chromatography-Mass Spectrometry , Glutathione/metabolism , Haplorhini , Humans , Isoxazoles/pharmacology , Kidney Diseases/chemically induced , Kidney Tubules/pathology , Male , Necrosis , Oxazines/toxicity , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Inhibitors/toxicity , Species Specificity
2.
Arch Biochem Biophys ; 307(2): 386-90, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8274026

ABSTRACT

The present study determined the effect of genetic obesity and phenobarbital (PB) treatment on the expression and regulation of the hepatic cytochrome P450 enzyme (CYP2C11) in Fa/? and fa/fa Zucker rats. Hepatic CYP2C11 levels as determined by Western immunoblotting and associated enzymatic activity (testosterone oxidation at the 2 alpha position) were significantly lower in untreated fa/fa Zucker rats compared with that observed in Fa/? Zucker rats. There was no significant difference in the constitutive CYP2C11 steady-state mRNA level hybridizable to the cDNA (P450 16 alpha) or specific oligonucleotide probe (Northern and slot blot analyses) between fa/fa and Fa/? Zucker rats. The depressed constitutive CYP2C11 protein levels in fa/fa rats may be attributed to their low plasma testosterone and growth hormone levels; however, lack of differences in CYP2C11 steady-state mRNA suggest post-transcriptional regulatory mechanism(s). Treatment with PB further suppressed hepatic CYP2C11 protein levels and activities in both fa/fa and Fa/? Zucker rats in comparison with that seen in controls. The level of CYP2C11 steady-state mRNA was significantly higher after treatment with PB in Fa/? Zucker rats, while no change was observed in fa/fa animals. The mechanism by which PB treatment fails to increase CYP2C11 steady-state mRNA levels in the fa/fa Zucker rat is unknown; however, it may share a common molecular basis with the defect in nuclear transcription rate previously observed with CYP2B1/2B2.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Gene Expression Regulation, Enzymologic , Isoenzymes/metabolism , Obesity/enzymology , Phenobarbital/pharmacology , Sex Characteristics , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/biosynthesis , Animals , Base Sequence , Body Weight/drug effects , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 2 , DNA , Gene Expression Regulation, Enzymologic/drug effects , Heterozygote , Homozygote , Isoenzymes/genetics , Male , Microsomes, Liver/drug effects , Molecular Sequence Data , Obesity/genetics , Organ Size/drug effects , RNA, Messenger/analysis , Rats , Rats, Zucker , Steroid Hydroxylases/genetics , Testosterone/metabolism
3.
Arch Biochem Biophys ; 303(2): 313-20, 1993 Jun.
Article in English | MEDLINE | ID: mdl-8512318

ABSTRACT

The present study describes the mechanism of the dampened induction of the CYP2B1 and CYP2B2 genes following phenobarbital treatment in the phenotypically obese fa/fa Zucker rat. The fa/fa Zucker rat demonstrated a threefold lower level of CYP2B1/2B2 enzyme induction, as indicated by reduced testosterone oxidation at the 16 beta position and resorufin formation from pentoxy- and benzyloxyresorufin, protein concentration (Western blot analysis), and steady-state mRNA levels (Northern and slot blot analyses) following in vivo treatment with phenobarbital than the Fa/? littermate controls. A primary hepatocyte cell culture system was used to determine if the dampened induction of the CYP2B1/2B2 enzyme is dependent on hormonal influences. Phenobarbital-treated (0.75 mM) hepatocytes from fa/fa Zucker rats showed approximately a three-fold lower induction response based on measurements of CYP2B1/2B2 (R-17 cDNA probe) and CYP2B1 (oligo probe) mRNAs. In order to evaluate whether this dampened response was at the level of transcriptional activation or initiation, as opposed to altered message stability, we measured the rate of transcription of CYP2B1/2B2 genes in nuclei from cultured hepatocytes during run-off experiments. Compared to Fa/? controls, the fa/fa Zucker rat had a greater than threefold lower nuclear transcription rate of CYP2B1/2B2 mRNA. These results suggest that the defective induction of the CYP2B1 and CYP2B2 genes exists at the transcriptional level in the mutant obese fa/fa Zucker rat. These data provide strong evidence that at least two genes are involved. Multiple gene involvement would suggest that the defect is not due to a mutation of the CYP2B gene cis-acting sequence. Instead, the lack of binding of a trans-acting factor, the presence of a repressor, or a defect in transcriptional activation is more likely the molecular mechanism(s) for this enzyme induction defect.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Mutation , Obesity/enzymology , Oxidoreductases/genetics , Phenobarbital/pharmacology , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Cytochrome P-450 CYP2B1 , DNA Probes , Liver/drug effects , Liver/enzymology , Male , Oxazines/metabolism , Oxidation-Reduction , RNA, Messenger/metabolism , Rats , Rats, Zucker , Testosterone/metabolism , Transcription, Genetic/drug effects
4.
Arch Biochem Biophys ; 296(2): 374-83, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1378714

ABSTRACT

The 6 alpha-, 7 alpha-, and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by rat prostate microsomes appears to be catalyzed by a single, high-affinity cytochrome P450 enzyme. In the present study we have examined the hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by prostate microsomes from cynomolgus monkeys and from normal subjects and patients with benign prostatic hyperplasia. Our results suggest that although rat, monkey, and human prostate microsomes catalyze the 6 alpha-, 7 alpha-, and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol, these pathways of oxidation in monkeys and humans are not catalyzed by a single cytochrome P450 enzyme. The ratio of the three metabolites was not uniform among prostate microsomal samples from individual humans or monkeys. The 6 alpha-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol varied independently of both the 7 alpha- and 7 beta-hydroxylation, which varied in unison. The 6 alpha-, 7 alpha-, and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by monkey prostate microsomes appeared to be differentially affected by in vivo treatment of monkeys with beta-naphthoflavone or dexamethasone. Treatment of a monkey with dexamethasone appeared to cause a 2.5-fold increase in both the 7 alpha- and the 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol without increasing the 6 alpha-hydroxylation. The 7 alpha- and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by human and monkey prostate microsomes, but not the 6 alpha-hydroxylation, was inhibited by antibody against rat liver NADPH-cytochrome P450 reductase. Similarly, the 7 alpha- and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by human prostate microsomes, but not the 6 alpha-hydroxylation, was markedly inhibited (greater than 85%) by equimolar concentrations of the imidazole-containing antimycotic drugs ketoconazole, clotrimazole, and miconazole. These results suggest that the 7 alpha- and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by monkey and human prostate microsomes is catalyzed by a cytochrome P450 enzyme, whereas the 6 alpha-hydroxylation is catalyzed by a different enzyme which may or may not be a cytochrome P450 monooxygenase. The hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by prostate microsomes from normal human subjects was quantitatively and qualitatively similar to its hydroxylation by prostate microsomes from patients with benign prostatic hyperplasia.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Androstane-3,17-diol/metabolism , Cytochrome P-450 Enzyme System/metabolism , Microsomes/enzymology , Prostate/enzymology , Animals , Antibodies , Antifungal Agents/pharmacology , Benzoflavones/pharmacology , Carbon Monoxide/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Dexamethasone/pharmacology , Humans , Hydroxylation , Macaca fascicularis , Male , NADPH-Ferrihemoprotein Reductase/antagonists & inhibitors , NADPH-Ferrihemoprotein Reductase/immunology , Prostate/ultrastructure , Prostatic Hyperplasia/enzymology , Rats , Species Specificity , beta-Naphthoflavone
5.
Arch Biochem Biophys ; 296(2): 355-65, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1632629

ABSTRACT

The purpose of the present study was to test the hypothesis that rat prostate microsomes contain a single cytochrome P450 enzyme responsible for the conversion of 5 alpha-androstane-3 beta,17 beta-diol to a series of trihydroxylated products. The three major metabolites formed by in vitro incubation of 5 alpha-[3H]androstane-3 beta,17 beta-diol with rat prostate microsomes were apparently 5 alpha-androstane-3 beta,6 alpha,17 beta-triol, 5 alpha-androstane-3 beta,7 alpha,17 beta-triol, and 5 alpha-androstane-3 beta,7 beta,17 beta-triol, which were resolved and quantified by reverse-phase HPLC with a flow through radioactivity detector. The ratio of the three metabolites remained constant as a function of incubation time, microsomal protein concentration, ionic strength, and substrate concentration. The ratio of the three metabolites was dependent on pH, apparently because the hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol shifted from the 6 alpha- to the 7 alpha-position with increasing pH (6.8-8.0). The V(max) values were 380, 160, and 60 pmol/mg microsomal protein/min for the rate of 6 alpha-, 7 alpha-, and 7 beta-hydroxylation, respectively. Similar Km values (0.5-0.7 microM) were measured for enzymatic formation of all three metabolites, which suggests that formation of all three metabolites was catalyzed by a single, high-affinity enzyme. Testosterone, 5 alpha-dihydrotestosterone, and 5 alpha-androstane-3 alpha,17 beta-diol did not appreciably inhibit the hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol, suggesting that this enzyme exhibits a high degree of substrate specificity. Formation of all three metabolites was inhibited by antibody against rat liver NADPH-cytochrome P450 reductase (85%) and by a 9:1 mixture of carbon monoxide and oxygen (60%). Several chemical inhibitors of cytochrome P450 enzymes, especially the antimycotic drug clotrimazole, also inhibited the formation of all three metabolites. Polyclonal antibodies that recognize liver cytochrome P450 1A, 2A, 2B, 2C, and 3A enzymes did not inhibit 5 alpha-androstane-3 beta,17 beta-diol hydroxylase activity. Overall, these results are consistent with the hypothesis that the 6 alpha-, 7 alpha-, and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol by rat prostate microsomes is catalyzed by a single, high-affinity P450 enzyme. This cytochrome P450 enzyme appears to be structurally distinct from those in the 1A, 2A, 2B, 2C, and 3A gene families.


Subject(s)
Androstane-3,17-diol/metabolism , Cytochrome P-450 Enzyme Inhibitors , Microsomes/enzymology , Potassium Compounds , Prostate/enzymology , Animals , Antibodies , Carbon Monoxide/pharmacology , Chromatography, High Pressure Liquid , Clotrimazole/pharmacology , Cytochrome P-450 Enzyme System/immunology , Cytochrome P-450 Enzyme System/metabolism , Gas Chromatography-Mass Spectrometry , Hydrogen-Ion Concentration , Hydroxylation , Male , NADPH-Ferrihemoprotein Reductase/immunology , Osmolar Concentration , Phosphates/pharmacology , Potassium/pharmacology , Prostate/ultrastructure , Rats , Rats, Inbred Strains
6.
Arch Biochem Biophys ; 296(2): 366-73, 1992 Aug 01.
Article in English | MEDLINE | ID: mdl-1632630

ABSTRACT

5 alpha-Dihydrotestosterone, the principal androgen mediating prostate growth and function in the rat, is formed from testosterone by steroid 5 alpha-reductase. The inactivation of 5 alpha-dihydrotestosterone involves reversible reduction to 5 alpha-androstane-3 beta,17 beta-diol by 3 beta-hydroxysteroid oxidoreductase followed by 6 alpha-, 7 alpha-, or 7 beta-hydroxylation. 5 alpha-Androstane-3 beta,17 beta-diol hydroxylation represents the ultimate inactivation step of dihydrotestosterone in rat prostate and is apparently catalyzed by a single, high-affinity (Km approximately 0.5 microM) microsomal cytochrome P450 enzyme. The present studies were designed to determine if 5 alpha-androstane-3 beta,17 beta-diol hydroxylation by rat prostate microsomes is inhibited by agents that are known inhibitors of androgen-metabolizing enzymes. Inhibitors of steroid 5 alpha-reductase (4-azasteroid analogs; 10 microM) or inhibitors of 3 beta-hydroxysteroid oxidoreductase (trilostane, azastene, and cyanoketone; 10 microM) had no appreciable effect on the 6 alpha-, 7 alpha-, or 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol (10 microM) by rat prostate microsomes. Imidazole-type antimycotic drugs (ketoconazole, clotrimazole, and miconazole; 0.1-10 microM) all markedly inhibited 5 alpha-androstane-3 beta,17 beta-diol hydroxylation in a concentration-dependent manner, whereas triazole-type antimycotic drugs (fluconazole and itraconazole; 0.1-10 microM) had no inhibitory effect. The rank order of inhibitory potency of the imidazole-type antimycotic drugs was miconazole greater than clotrimazole greater than ketoconazole. In the case of clotrimazole, the inhibition was shown to be competitive in nature, with a Ki of 0.03 microM. The imidazole-type antimycotic drugs inhibited all three pathways of 5 alpha-androstane-3 beta,17 beta-diol hydroxylation to the same extent, which provides further evidence that, in rat prostate microsomes, a single cytochrome P450 enzyme catalyzes the 6 alpha-, 7 alpha-, and 7 beta-hydroxylation of 5 alpha-androstane-3 beta,17 beta-diol. These studies demonstrate that certain imidazole-type compounds are potent, competitive inhibitors of 5 alpha-androstane-3 beta,17 beta-diol hydroxylation by rat prostate microsomes, which is consistent with the effect of these antimycotic drugs on cytochrome P450 enzymes involved in the metabolism of other androgens and steroids.


Subject(s)
5-alpha Reductase Inhibitors , Androstane-3,17-diol/metabolism , Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Imidazoles/pharmacology , Microsomes/enzymology , Prostate/enzymology , 3-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Animals , Binding, Competitive , Clotrimazole/pharmacology , Hydroxylation , Male , Prostate/ultrastructure , Rats , Rats, Inbred Strains , Triazoles/pharmacology
7.
J Biochem Toxicol ; 7(1): 43-52, 1992.
Article in English | MEDLINE | ID: mdl-1588573

ABSTRACT

We recently reported that antibody against purified P450 3A1 (P450p) recognizes two electrophoretically distinct proteins (50 and 51 kDa) in liver microsomes from male and female rats, as determined by Western immunoblotting. Depending on the source of the liver microsomes, the 51-kDa protein corresponded to 3A1 and/or 3A2 which could not be resolved by sodium dodecyl sulfate (SDS)polyacrylamide gel electrophoresis. The other protein (50 kDa) appears to be another member of the P450 IIIA gene family. Both proteins were markedly intensified in liver microsomes from male or female rats treated with pregnenolone-16 alpha-carbonitrile, dexamethasone, troleandomycin, or chlordane. In contrast, treatment of male or female rats with phenobarbital intensified only the 51-kDa protein. Treatment of male rats with Aroclor 1254 induced the 51-kDa protein, but suppressed the 50-kDa form. In addition to their changes in response to inducers, the 50- and 51-kDa proteins also differed in their developmental expression. For example, the 50-kDa protein was not expressed until weaning (3 weeks), whereas the 51-kDa protein was expressed even in 1-week-old rats. At puberty (between weeks 5 and 6), the levels of the 50-kDa and 51-kDa proteins markedly declined in female but not in male rats, which introduced a large sex difference (male greater than female) in the levels of both proteins. Changes in the level of the 51-kDa protein were paralleled by changes in the rate of testosterone 2 beta-, 6 beta-, and 15 beta-hydroxylation. In male rats, the marked increase in the levels of the 50-kDa protein between weeks 2 and 3 coincided with a three- to four fold increase in the rate of testosterone 2 beta-, 6 beta-, and 15 beta-hydroxylation, which suggests that the 50-kDa protein catalyzes the same pathways of testosterone oxidation as the 51-kDa protein. However, this developmental increase in testosterone oxidation may have resulted from an activation of the 51-kDa 3A protein. These results indicate that the two electrophoretically distinct proteins recognized by antibody against P450 3A1 are regulated in a similar but not identical manner, and suggest that the 51-kDa 3A protein is the major microsomal enzyme responsible for catalyzing the 2 beta-, 6 beta-, and 15 beta-hydroxylation of testosterone.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Microsomes, Liver/enzymology , Testosterone/metabolism , Animals , Antibodies/immunology , Blotting, Western , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P-450 Enzyme System/immunology , Cytochromes b5/pharmacology , Enzyme Induction , Female , Hydroxylation , Male , NADPH-Ferrihemoprotein Reductase/metabolism , NADPH-Ferrihemoprotein Reductase/pharmacology , Oxidation-Reduction , Phosphatidylcholines/pharmacology , Rats , Rats, Inbred Strains , Species Specificity
8.
Mol Pharmacol ; 40(5): 859-67, 1991 Nov.
Article in English | MEDLINE | ID: mdl-1944247

ABSTRACT

In rats, cytochrome P450 (P450) IIIA enzymes are an important determinant of digitoxin toxicity. Induction of these liver microsomal enzymes decreases the toxicity of digitoxin by increasing its oxidative cleavage to digitoxigenin bis- and monodigitoxoside (dt2 and dt1). The present study shows that the susceptibility of different mammalian species to digitoxin toxicity is inversely related to liver microsomal P450 IIIA activity (measured as testosterone 6 beta-hydroxylase activity). Based on this correlation, we correctly predicted that hamsters, which have the highest P450 IIIA activity, are extremely resistant to digitoxin toxicity. To further examine the relationship between digitoxin toxicity and P450 IIIA activity, the pathways of digitoxin metabolism catalyzed by liver microsomes from nine mammalian species were examined by high performance liquid chromatography. The overall rate of digitoxin metabolism varied approximately 90-fold and followed the rank order: hamster greater than rat greater than guinea pig greater than dog greater than mouse approximately monkey greater than rabbit approximately cat greater than human. The qualitative differences in digitoxin metabolism were as striking as the quantitative differences. Formation of 16- and/or 17-hydroxydigitoxin was the major pathway of digitoxin oxidation catalyzed by liver microsomes from hamster, guinea pig, rabbit, cat, dog, and cynomolgus monkey. Guinea pig and, to a lesser extent, hamster liver microsomes also converted digitoxin to an unknown metabolite, the formation of which was catalyzed by P450. None of the species examined catalyzed the 12-hydroxylation of digitoxin to digoxin at a high rate. Similarly, none of the species examined catalyzed a high rate of conversion of digitoxin to dt2, with the notable exception of the rat. However, dt2 formation was the major pathway of digitoxin metabolism catalyzed by human liver microsomes, although humans were much less active (approximately 2%) than rats in this regard. The rate of dt2 formation varied approximately 41-fold among 22 samples of human liver microsomes, which was highly correlated (r = 0.841) with the rate of testosterone 6 beta-hydroxylation. Antibody against rat P450 IIIA1 inhibited the high rate of dt2 formation by rat liver microsomes and the low rate catalyzed by mouse, guinea pig, dog, monkey, and human liver microsomes. In contrast, anti-P450 IIIA1 did not inhibit the 12-, 16-, or 17-hydroxylation of digitoxin (or the formation of the unknown metabolite), despite the fact that anti-P450 IIIA1 strongly inhibited (greater than 70%) the 6 beta-hydroxylation of testosterone by liver microsomes from each of the species examined (except rabbit liver microsomes, which were inhibited only approximately 30%).(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Digitoxin/metabolism , Adolescent , Adult , Animals , Cats , Child , Cricetinae , Cytochrome P-450 Enzyme System/analysis , Digitoxin/toxicity , Dogs , Female , Guinea Pigs , Humans , Hydroxytestosterones/metabolism , Macaca fascicularis , Male , Mesocricetus , Mice , Mice, Inbred C3H , Microsomes, Liver/metabolism , Middle Aged , Rabbits , Rats , Species Specificity
9.
Proc Natl Acad Sci U S A ; 88(12): 5227-31, 1991 Jun 15.
Article in English | MEDLINE | ID: mdl-2052602

ABSTRACT

Differences in the pattern of growth hormone (GH) secretion in mature rats (i.e., "continuous" secretion in females versus "pulsatile" secretion in males) are thought to be the underlying cause of sex-dependent differences in a subpopulation of liver microsomal P450 enzymes and steroid 5 alpha-reductase. A new strain of dwarf rats (NIMR/AS) has recently been shown to have low or undetectable levels of circulating GH due to a selective defect in pituitary GH synthesis. We have measured the levels and/or activity of IIA1 (P450a), IIA2 (P450m), IIC11 (P450h), IIC12 (P450i), IIIA2 (a P450p isozyme), and steroid 5 alpha-reductase in liver microsomes from male and female dwarf rats, to test the hypothesis that the expression of these sexually dimorphic enzymes is regulated by GH. In mature rats, the levels of liver microsomal IIA2, IIC11, and IIIA2 were higher in male than in female dwarf rats, whereas the levels of activity of IIA1, IIC12, and steroid 5 alpha-reductase were greater in female than in male dwarf rats. These sex differences resulted from age-related changes in either male dwarf rats (i.e., an increase in IIC11 and IIA2 and a decrease in IIA1) or female dwarf rats (i.e., an increase in IIC12 and 5 alpha-reductase and a decrease in IIIA2). The magnitudes of these sex-dependent, age-related changes were essentially indistinguishable from those observed in normal rats. These unexpected results suggest that GH is not the pituitary factor responsible for regulating the levels of sexually dimorphic, steroid-metabolizing enzymes in rat liver. Alternatively, it is possible that these enzymes are regulated by extremely low levels of GH. In either case, the current model of how steroid-metabolizing enzymes are regulated in rats must be revised to account for the normal sexual differentiation of these enzymes in dwarf rats.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Cytochrome P-450 Enzyme System/metabolism , Growth Hormone/physiology , Microsomes, Liver/enzymology , Sex Differentiation , Animals , Blotting, Western , Female , Growth Hormone/biosynthesis , Male , Oxidation-Reduction , Rats , Sex Factors , Thyroid Hormones/analysis
11.
J Steroid Biochem ; 35(3-4): 429-40, 1990 Mar.
Article in English | MEDLINE | ID: mdl-2325409

ABSTRACT

The aim of this study was to determine the effects of ionic strength and pH on the different pathways of testosterone oxidation catalyzed by rat liver microsomes. The catalytic activity of cytochromes P-450a (IIA1), P-450b (IIB1), P-450h (IIC11) and P-450p (IIIA1) was measured in liver microsomes from mature male rats and phenobarbital-treated rats as testosterone 7 alpha-, 16 beta-, 2 alpha- and 6 beta-hydroxylase activity, respectively. An increase in the concentration of potassium phosphate (from 25 to 250 mM) caused a marked decrease in the catalytic activity of cytochromes P-450a (to 8%), P-450b (to 22%) and P-450h (to 23%), but caused a pronounced increase in the catalytic activity of cytochrome P-450p (up to 4.2-fold). These effects were attributed to changes in ionic strength, because similar but less pronounced effects were observed with Tris-HCl (which has approximately 1/3 the ionic strength of phosphate buffer at pH 7.4). Testosterone oxidation by microsomal cytochromes P-450a, P-450b, P-450h and P-450p was also differentially affected by pH (over the range 6.8-8.0). The pH optima ranged from 7.1 (for P-450a and P-450h) to 8.0 (for P-450p), with an intermediate value of 7.4 for cytochrome P-450b. Increasing the pH from 6.8 to 8.0 unexpectedly altered the relative amounts of the 3 major metabolites produced by cytochrome P-450h. The decline in testosterone oxidation by cytochromes P-450a, P-450b and P-450h that accompanied an increase in ionic strength or pH could be duplicated in reconstitution systems containing purified P-450a, P-450b or P-450h, equimolar amounts of NADPH-cytochrome P-450 reductase and optimal amounts of dilauroylphosphatidylcholine. This result indicated that the decline in testosterone oxidation by cytochromes P-450a, P-450b and P-450h was a direct effect of ionic strength and pH on these enzymes, rather than a secondary effect related to the increase in testosterone oxidation by cytochrome P-450p. Similar studies with purified cytochrome P-450p were complicated by the atypical conditions needed to reconstitute this enzyme. However, studies on the conversion of digitoxin to digitoxigenin bisdigitoxoside by liver microsomes, which is catalyzed specifically by cytochrome P-450p, provided indirect evidence that the increase in catalytic activity of cytochrome P-450p was also a direct effect of ionic strength and pH on this enzyme.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Cytochrome P-450 Enzyme System/physiology , Microsomes, Liver/enzymology , Potassium Compounds , Testosterone/metabolism , Animals , Buffers , Digitoxin/metabolism , Enzyme Induction , Hydrogen-Ion Concentration , Male , Osmolar Concentration , Oxidation-Reduction , Phosphates/pharmacology , Potassium/pharmacology , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL