Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Arch Biochem Biophys ; 312(1): 234-9, 1994 Jul.
Article in English | MEDLINE | ID: mdl-8031133

ABSTRACT

Sexually dimorphic regulation of phenobarbital-induced cytochromes P450 2B1 and 2B2 (collectively referred to as P450 2B) as well as P450 2B-dependent monooxygenase activities was studied in multi-hormone-depleted hypophysectomized rats and in growth hormone (GH)-deficient monosodium glutamate (MSG)-treated rats. Our results indicate that endogenous GH suppresses phenobarbital induction of P450 2B and that the feminine pattern of continuous GH secretion is more suppressive than the masculine profile of episodic secretion. Moreover, we have found that it is the height of the GH pulse, and not necessarily its frequency nor the interpulse trough periods, that signals the suppressive effects of GH on P450 2B expression in male rats. Last, irrespective of the presence or absence of circulating GH, the magnitude of phenobarbital induction of P450 2B and associated monooxygenases was consistently lower in female rats, suggesting the presence of some degree of sex-dependent, but GH-independent regulation.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Enzyme Induction/drug effects , Growth Hormone/pharmacology , Microsomes, Liver/enzymology , Phenobarbital/pharmacology , Sex Characteristics , Steroid Hydroxylases/biosynthesis , Animals , Female , Growth Hormone/blood , Hypophysectomy , Male , Periodicity , Rats , Rats, Sprague-Dawley , Sodium Glutamate/pharmacology
2.
Cancer Res ; 54(11): 2878-86, 1994 Jun 01.
Article in English | MEDLINE | ID: mdl-8187072

ABSTRACT

Dehydroepiandrosterone (DHEA) given to rodents in pharmacological doses induces several hepatic enzymes including cytochromes P4504A, NADPH:P450 oxidoreductase, palmitoyl coenzyme A oxidase, and other enzymes associated with the peroxisomal beta-oxidation pathway, leading to peroxisome proliferation and development of hepatocellular carcinoma in rodents. Comparison of the inductive potency of DHEA and other intermediates of the steroid biosynthetic path demonstrated that only DHEA, 5-ene-androstene-3 beta,17 beta-diol (ADIOL), and to a lesser extent, 17 alpha-hydroxypregnenolone, a precursor of DHEA, induce cytochromes P4504A protein and other enzymes associated with the peroxisome proliferative response in vivo. ADIOL exerted its inductive response at a somewhat lower dosage than DHEA, whereas ADIOL and DHEA both induced the microsomal enzymes (P4504A and its oxidoreductase) at somewhat lower dosages than those required to induce peroxisomal enzymes. Northern analysis demonstrated increases in the mRNAs encoding the cytochromes P4504A (> 20-fold) and NADPH:P450 oxidoreductase (> 10-fold) in the livers of DHEA- and ADIOL-treated rats. Run-on transcription analysis demonstrated that DHEA induces CYP4A gene expression 11-fold at the level of transcription initiation. Comparison of the responsiveness of individual rat CYP4A genes (4A1, 4A2, and 4A3) to DHEA and ADIOL in immature versus mature male rats revealed 2-3-fold higher levels of induced CYP4A1 and 4A3 mRNAs in immature rat livers. In contrast, hepatic CYP4A2 mRNA was induced to 6-10-fold higher levels in mature rats. No basal or significant inducible expression of mRNA for CYP4A1 and 4A3 was noted in rat kidney. Significant basal levels of kidney CYP4A2 mRNA were observed only in mature animals, where they were inducible by ADIOL and DHEA to a 3-5-fold greater extent than in the kidneys of immature rats. These studies demonstrate developmental differences in the responsiveness of CYP4A mRNA levels to DHEA and ADIOL in rat kidney and liver. Moreover, the striking inducibility of CYP4A protein and mRNAs, together with the increased rates of synthesis of nascent CYP4A mRNA transcripts in hepatic nuclei from DHEA-treated rats, establish that DHEA increases the expression of these microsomal enzymes at the transcriptional level.


Subject(s)
Androstenediol/pharmacology , Catalase/biosynthesis , Cytochrome P-450 Enzyme System/biosynthesis , Dehydroepiandrosterone/pharmacology , Microbodies/enzymology , Microsomes, Liver/enzymology , Oxidoreductases/biosynthesis , Animals , Dehydroepiandrosterone/analogs & derivatives , Dose-Response Relationship, Drug , Enzyme Induction , Male , NADP/biosynthesis , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
3.
Cancer Res ; 52(20): 5797-802, 1992 Oct 15.
Article in English | MEDLINE | ID: mdl-1394205

ABSTRACT

Glutathione S-transferases (GSTs) play an important role in the detoxification of diverse electrophilic chemicals, including anticancer drugs. Gene-specific oligonucleotide probes were developed to monitor the expression of individual GST mRNAs in livers of adult male rats treated with drugs and other chemical modulators of GST expression. Northern blot analysis of total liver RNA using probes specific for individual GSTs belonging to classes alpha (GSTs Ya1, Ya2, Yc), mu (GSTs Yb1, Yb2, Yb3), pi (GST Yp), and GSTms demonstrated the expression in liver of all but Yp mRNA. Kidney GST expression was at least as high as that in liver for GSTs Ya1, Yc, and Yp, while it was substantially lower but still detectable for GSTs Ya2, Yb2, and GSTms. Several of the liver GST class alpha mRNAs, in particular Ya2, were inducible by pretreatment of rats with phenobarbital or isosafrole. In contrast, dexamethasone preferentially induced Yb1, Yb2, and Ya2, while two other inducers of liver drug metabolism, isoniazid and clofibrate, were less effective with respect to GST induction. GSTms mRNA was induced to a small extent or not at all by the agents tested. Treatment of adult male rats with the anticancer drug cisplatin increased liver expression of GST Yc mRNA and suppressed Ya1 mRNA levels with little or no major effect on several other GST mRNAs. Western blot analysis of liver cytosols prepared from the cisplatin-treated rats revealed corresponding changes in GST Yc and Ya protein levels. Comparable changes in liver GST Ya1 and Yc expression were effected by the cisplatin analogue iproplatin but not by carboplatin or transplatin. This pattern of response to these platinum drugs is comparable to that seen with respect to platinum drug-induced gonadal toxicity and modulation of liver cytochrome P450 expression, suggesting a common mechanistic basis for these diverse effects of platinum anticancer drugs on hepatic enzymes of drug metabolism. Together, these studies demonstrate the utility of oligonucleotide probes for phenotyping liver tissue for the expression of GST enzymes that can contribute to anticancer drug metabolism and resistance. They also raise the possibility of drug-drug interactions involving cisplatin and alkylating agent anticancer drugs that can be metabolized in liver by alpha-class GSTs.


Subject(s)
Cisplatin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glutathione Transferase/genetics , Microsomes, Liver/enzymology , Oligonucleotide Probes , RNA Probes , Animals , Base Sequence , Cisplatin/analogs & derivatives , Cisplatin/toxicity , Enzyme Induction , Female , Glutathione Transferase/biosynthesis , Kidney/enzymology , Liver/enzymology , Male , Molecular Sequence Data , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344
4.
Biochem Pharmacol ; 43(10): 2201-8, 1992 May 28.
Article in English | MEDLINE | ID: mdl-1599506

ABSTRACT

Hybridomas obtained by the fusion of spleen cells from rat cytochrome b5-immunized mice with mouse myeloma cells produced five groups of monoclonal antibodies (MAbs) with three mouse immunoglobulin subtypes: IgG1, IgG2b and IgM. All of the MAbs bound strongly to rat cytochrome b5 as measured by radioimmunoassay (RIA). Four clones of MAbs were also strongly immunoreactive with cytochrome b5 when tested by Western blotting, but only one of the MAbs (1-39-2) weakly immunoprecipitated cytochrome b5 in an Ouchterlony double-immunodiffusion test. Two of the MAbs partially inhibited cytochrome b5-mediated NADH cytochrome c reduction catalyzed by liver microsomes (24-36%). Expression of immunodetectable cytochrome b5 was highest in the liver, next highest in the kidney, and quite low in the other tissues examined with MAb 1-17-1 by Western blotting. This MAb recognized homologous cytochrome b5 of human liver microsomes and in homogenates of TK- cells infected with recombinant vaccinia virus encoding human cytochrome b5. These MAbs to cytochrome b5 will be useful for the identification, quantification, and purification of cytochrome b5 from animal and human tissues, and for understanding its role in cytochrome P450 catalyzed drug metabolism and carcinogen activation with respect to tissue, organ and individual differences.


Subject(s)
Antibodies, Monoclonal , Cytochromes b5/immunology , Microsomes, Liver/enzymology , Age Factors , Animals , Antibodies, Monoclonal/immunology , Cytochromes b5/metabolism , Female , Immunodiffusion , Kidney/enzymology , Male , NADH Dehydrogenase/immunology , Radioimmunoassay , Rats , Rats, Inbred Strains
5.
Arch Biochem Biophys ; 290(1): 160-6, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1898086

ABSTRACT

Steroid hydroxylation specificities were determined for 11 forms of human cytochrome P450, representing four gene families and eight subfamilies, that were synthesized in human hepatoma Hep G2 cells by means of cDNA-directed expression using vaccinia virus. Microsomes isolated from the P450-expressing Hep G2 cells were isolated and then assayed for their regioselectivity of hydroxylation toward testosterone, androstenedione, and progesterone. Four of the eleven P450s exhibited high steroid hydroxylase activity (150-900 pmol hydroxysteroid/min/mg Hep G2 microsomal protein), one was moderately active (30-50 pmol/min/mg) and six were inactive. In contrast, 10 of the P450s effectively catalyzed O-deethylation of 7-ethoxycoumarin, a model drug substrate, while only one (P450 2A6) catalyzed significant coumarin 7-hydroxylation. Human P450 4B1, which is expressed in lung but not liver, catalyzed the 6 beta-hydroxylation of all three steroids at similar rates and with only minor formation of other hydroxylated products. Three members of human P450 family 3A, which are expressed in liver and other tissues, also catalyzed steroid 6 beta-hydroxylation as their major activity but, additionally, formed several minor products that include 2 beta-hydroxy and 15 beta-hydroxy derivatives in the case of testosterone. These patterns are similar to those exhibited by rat family 3A P450s. Although several rodent P450s belonging to subfamilies 2A, 2B, 2C, 2D are active steroid hydroxylases, four of five human P450s belonging to these subfamilies exhibited very low activity or were inactive, as were the human 1A and 2E P450s examined in the present study. These studies demonstrate that individual human cytochrome P450 enzymes can hydroxylate endogenous steroid hormones with a high degree of stereospecificity and regioselectivity, and that some, but not all of the human cytochromes exhibit metabolite profiles similar to their rodent counterparts.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Steroid Hydroxylases/metabolism , Cytochrome P-450 Enzyme System/genetics , DNA/genetics , Genetic Vectors , Humans , Kinetics , Multigene Family , Steroid Hydroxylases/genetics , Steroids , Substrate Specificity , Tumor Cells, Cultured/enzymology , Vaccinia virus/genetics
6.
Biochem J ; 265(1): 187-94, 1990 Jan 01.
Article in English | MEDLINE | ID: mdl-2302165

ABSTRACT

Rat hepatic cytochrome P-450 form 3 (testosterone 7 alpha-hydroxylase; P-450 gene IIA1) and P-450 form RLM2 (testosterone 15 alpha-hydroxylase; P-450 gene IIA2) are 88% identical in primary structure, yet they hydroxylate testosterone with distinct and apparently unrelated regioselectivities. In this study, androstenedione and progesterone were used to assess the regioselectivity and stereospecificity of these two P-450 enzymes towards other steroid substrates. Although P-450 RLM2 exhibited low 7 alpha-hydroxylase activity with testosterone or progesterone as substrate (turnover number less than or equal to 1-2 nmol of metabolite/min per nmol of P-450), it did catalyse androstenedione 7 alpha-hydroxylation at a high rate (21 min-1) which exceeded that of P-450 3 (7 min-1). However, whereas P-450 3 exhibited a high specificity for hydroxylation of these steroids at the 7 alpha position (95-97% of total activity), P-450 RLM2 actively metabolized these compounds at four or more major sites including the nearby C-15 position, which dominated in the case of testosterone and progesterone. The observation that androstenedione is actively 7 alpha-hydroxylated by purified P-450 RLM2 suggested that this P-450 enzyme might make significant contributions to microsomal androstenedione 7 alpha-hydroxylation, an activity that was previously reported to be associated with immunoreactive P-450 3. Antibody inhibition experiments were therefore carried out in liver microsomes using polyclonal anti-(P-450 3) antibodies which cross-react with P-450 RLM2, and using a monoclonal antibody that is reactive with and inhibitory towards P-450 3 but not P-450 RLM2. P-450 3 was thus shown to catalyse only around 35% of the total androstenedione 7 alpha-hydroxylase activity in uninduced adult male rat liver microsomes, with the balance attributed to P-450 RLM2. The P-450-3-dependent 7 alpha-hydroxylase activity was increased to approximately 65% of the total in phenobarbital-induced adult male microsomes, and to greater than 90% of the total in untreated adult female rat liver microsomes. These observations are consistent with the inducibility of P-450 3 by phenobarbital and with the absence of P-450 RLM2 from adult female rat liver respectively. These findings establish that P-450 RLM2 and P-450 3 can both contribute significantly to microsomal androstenedione 7 alpha-hydroxylation, thus demonstrating that the 7 alpha-hydroxylation of this androgen does not serve as a specific catalytic monitor for microsomal P-450 3.


Subject(s)
Androstenedione/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Isoenzymes/metabolism , Microsomes, Liver/metabolism , Steroid Hydroxylases/metabolism , Animals , Blotting, Western , Catalysis , Chromatography, Gas , Chromatography, High Pressure Liquid , Chromatography, Thin Layer , Female , Hydroxylation , Male , Microsomes, Liver/enzymology , Progesterone/metabolism , Protein Conformation , Rats , Rats, Inbred Strains , Substrate Specificity
7.
Endocrinology ; 125(6): 2935-44, 1989 Dec.
Article in English | MEDLINE | ID: mdl-2510988

ABSTRACT

Neonatal male rats were treated with monosodium glutamate (MSG) at either 2 or 4 mg/g BW on alternate days during the first 10 days of life. As adults, the 4 mg MSG-treated rats displayed the obesity, growth retardation, and reduced pituitary weights that typify this syndrome. These animals had no detectable plasma GH as determined from serial blood samples taken every 20 min for 8 consecutive h. Associated with this loss of circulating GH was an induction of the female-specific hepatic cytochrome P450 2d (gene IIC12) and the disappearance of the male-specific form of cytochrome P450 2c (gene IIC11). The catalytic activities of cytochrome P450 2c (i.e. androgen 16 alpha- and 2 alpha-hydroxylase), sex-dependent hexobarbital hydroxylase and total cytochrome P450 were similarly feminized. Rats exposed to the 2-mg dose of MSG were also obese, but their growth rates and pituitary sizes were not as severely affected as in the 4 mg MSG-treated rats. Circulating GH in these lower dosed males was secreted in a pulsatile pattern similar to that in normal males, except that the pulse amplitude was reduced as much as 90%. In spite of this profound decline in GH peak heights in the 2 mg MSG-treated males, liver metabolism was characteristically masculine. That is, female-specific cytochrome P450 2d remained undetectable, while the male forms of cytochrome P450 2c, 2a (gene IIIA2), and RLM2 (gene IIA2), their respective catalytic steroid hydroxylase activities, and associated sex-dependent drug-metabolizing enzymes were expressed at the level of or greater than that in normal males. Thus, while an ultradian pulse of circulating GH is necessary for the characteristically masculine profile of sex-specific forms of hepatic cytochrome P450, neither the amplitude of the secretory peaks nor their total GH content is critical, and these can be greatly reduced from the normal male levels.


Subject(s)
Chronobiology Phenomena/physiology , Cytochrome P-450 Enzyme System/genetics , Gene Expression/physiology , Growth Hormone/blood , Liver/enzymology , Sex Characteristics , Adrenal Glands/anatomy & histology , Animals , Animals, Newborn/physiology , Cytochrome P-450 Enzyme System/metabolism , Female , Male , NADPH-Ferrihemoprotein Reductase/metabolism , Organ Size/drug effects , Pituitary Gland/anatomy & histology , Rats , Rats, Inbred Strains , Sodium Glutamate/pharmacology , Weight Gain/drug effects
8.
J Biol Chem ; 264(35): 21327-33, 1989 Dec 15.
Article in English | MEDLINE | ID: mdl-2574176

ABSTRACT

The phenobarbital-inducible P-450 forms IIB1 and IIB2 are identical in sequence except for 14 amino acid differences within the carboxyl-terminal half of the molecule. IIB1 has about a 5-10-fold higher turnover number for most monooxygenase substrates examined although the substrate specificities of both enzymes are virtually identical. Both P-450s oxygenate testosterone to yield the 16 alpha-hydroxy, 16 beta-hydroxy, 17-keto, and 16 beta-hydroxy, 17-keto metabolites as major products. A variant IIB2 cDNA, isolated from an uninduced rat liver lambda gt11 library, and when expressed in Hep G2 cells using a vaccinia virus vector, was found to code for a protein that produced the 16 alpha-hydroxy and 17-keto metabolites of testosterone but no 16 beta-hydroxylated products. Although the published sequences of IIB1 and IIB2 are identical within the N-terminal halves of the proteins, sequence analysis of the variant cDNA revealed two amino acid substitutions in this region; Leu58----Phe and I1e114----Phe. When these two amino acid changes were incorporated into IIB1, via construction of a chimeric cDNA, the resultant expressed enzyme did not catalyze the 16 beta-hydroxylation of testosterone or androstenedione. Formation of the 16 alpha-hydroxy and 17-keto metabolites, however, was only slightly reduced compared with the parent IIB1. A IIB1 protein that possessed only the I1e114----Phe replacement catalyzed the production of all four testosterone metabolites with only slightly different product ratios compared with the parent enzyme. The substrate specificity of a IIB1 variant containing only the Leu58----Phe replacement could not be determined, since that protein did not accumulate in cells infected with the corresponding recombinant vaccinia virus. These data suggest that two distinct amino acid residues located within the amino-terminal fourth of IIB1 and IIB2 can affect substrate orientation at the active site.


Subject(s)
Isoleucine , Leucine , Mutation , Phenylalanine , Steroid 11-beta-Hydroxylase/genetics , Steroid Hydroxylases/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Chimera , Cloning, Molecular/methods , Genetic Vectors , Liver/enzymology , Molecular Sequence Data , Poly A/genetics , RNA, Messenger/genetics , Rats , Rats, Inbred Strains , Steroid 11-beta-Hydroxylase/metabolism , Testosterone/isolation & purification , Testosterone/metabolism , Vaccinia virus/genetics
9.
Biochem Pharmacol ; 38(18): 3067-74, 1989 Sep 15.
Article in English | MEDLINE | ID: mdl-2783161

ABSTRACT

Hybridomas were formed from myeloma cells and spleen cells derived from BALB/c female mice immunized with purified liver microsomal cytochrome P-450 2c/RLM5 (P-450 gene IIC11) isolated from untreated adult male rats. Six hybridoma clones produced monoclonal antibodies (MAbs) of the IgM(kappa) type. All the MAbs bound strongly to P-450 2c/RLM5 when measured by radioimmunoassay, and four of the six specifically immunoprecipitated P-450 2c/RLM5 in an Ouchterlony double-immunodiffusion test. These four MAbs also bound but did not immunoprecipitate P-450 RLM3. The MAbs that precipitated P-450 2c/RLM5 neither bound nor precipitated P-450 PB-B (gene IIB1) and P-450 BNF-B (gene IA1) of rats or P-450 LM2 and P-450 LM4 of rabbits. In contrast, mouse polyclonal anti-P-450 2c/RLM5 antibody strongly immunoprecipitated P-450 RLM3 as well as P-450 2c/RLM5 and to a lesser extent P-450 PB-B and P-450 LM2. The MAbs that precipitated P-450 2c/RLM5 also inhibited by more than 90% androstenedione 16 alpha-hydroxylase activity of untreated rat microsomes, but did not inhibit microsomal 6 beta- or 7 alpha-hydroxylation. In addition, complete inhibition of both androstenedione 16 alpha-hydroxylation and testosterone 16 alpha-hydroxylation was observed in a reconstituted system with P-450 2c/RLM5. Androstenedione 6 beta-hydroxylation catalyzed by P-450 2c/RLM5 was also inhibited, whereas P-450 3-catalyzed 7 alpha-hydroxylation was not inhibited by the MAbs. P-450 2c/RLM5 catalyzed 2 alpha-, 16 alpha- and 6 beta-hydroxylation of progesterone in a reconstituted system were also inhibited by the MAb by 60-80%. These MAbs should prove useful for "reaction phenotyping," i.e. for defining the contribution of microsomal P-450 2c/RLM5 to the oxidative metabolism of endogenous steroids and other P-450 substrates in animal and human tissues.


Subject(s)
Antibodies, Monoclonal/immunology , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/immunology , Microsomes, Liver/enzymology , Steroids/metabolism , Androstenedione/metabolism , Animals , Antibody Specificity , Cytochrome P-450 CYP2C8 , Cytochrome P-450 CYP2C9 , Cytochrome P450 Family 2 , Female , Hydroxylation , Male , Mice , Mice, Inbred BALB C , Progesterone/metabolism , Rats , Steroid 16-alpha-Hydroxylase
10.
J Biol Chem ; 264(18): 10388-95, 1989 Jun 25.
Article in English | MEDLINE | ID: mdl-2732228

ABSTRACT

Immunoblotting analysis of human liver microsome preparations revealed that human cytochrome P-450 PCN1 (hPCN1, Mr approximately 52,000) was expressed in each of 40 individual specimens examined. In about 10-20% of the livers, an immunologically related protein having a lower electrophoretic mobility (Mr approximately 52,500) was also detected. A single liver was found that expressed only the lower mobility protein, designated hPCN3, and RNA isolated from this liver was used to construct a lambda gt11 library. The library was screened with an hPCN1 cDNA probe resulting in the isolation of a unique full-length cDNA that was sequenced and shown to encode hPCN3. The deduced amino acid sequence of this cDNA contained 502 residues, a calculated molecular mass of 57,115 daltons, and displayed 84% similarity with hPCN1. The deduced amino-terminal sequence of hPCN3 was identical to that of HFLa, a major cytochrome P-450 expressed in human fetal liver that is immunologically cross-reactive with several family III cytochrome P-450s. hPCN1 and hPCN3 cDNAs were expressed in Hep G2 cells using a vaccinia virus expression system and shown to encode active enzymes, both characterized by reduced CO-binding spectra with lambda max at 450 nm. Enzymatic analysis revealed that both cytochrome P-450s were similarly active in catalyzing oxidation of the calcium channel blocking drug nifedipine. Both enzymes also catalyzed 6 beta-hydroxylation of the steroid hormones testosterone, progesterone, and androstenedione, although hPCN1 exhibited several-fold higher expressed activity than hPCN3. Several minor oxidation products of these steroids (e.g. 15 beta-hydroxytestosterone), comprising up to approximately 20% of the total metabolites, were formed by hPCN1 but not hPCN3, indicating that hPCN3 is a more highly regiospecific monooxygenase catalyst with steroid substrates. Clear differences were also detected in their catalytic activities toward the immunosuppressive drug cyclosporine, with two hydroxylated metabolites (M1 and M17) and one demethylated metabolite (M21) formed by hPCN1 but only one metabolite (M1) formed by hPCN3. These studies establish that hPCN3 is a newly described cytochrome P-450 that is differentially expressed in the adult human population and that has overlapping substrate specificity compared to hPCN1 for metabolism of steroid and drug substrates.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , DNA/genetics , Genes , Microsomes, Liver/enzymology , Mixed Function Oxygenases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , Cyclosporins/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Humans , Immunoblotting , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , Species Specificity , Substrate Specificity , Vaccinia virus/genetics
11.
Biochem J ; 260(1): 81-5, 1989 May 15.
Article in English | MEDLINE | ID: mdl-2789039

ABSTRACT

Androgen hydroxylation catalysed by Chinese hamster fibroblast SD1 cells, which stably express cytochrome P-450 form PB-4, the rat P450IIB1 gene product, was assessed and compared to that catalysed by purified cytochrome P-450 PB-4 isolated from rat liver. SD1 cell homogenates catalysed the NADPH-dependent hydroxylation of androstenedione and testosterone with a regioselectivity very similar to that purified by P-450 PB-4 (16 beta-hydroxylation/16 alpha-hydroxylation = 6.0-6.8 for androstenedione; 16 beta/16 alpha = 0.9 for testosterone). Homogenates prepared from the parental cell line V79, which does not express detectable levels of P-450 PB-4 or any other cytochrome P-450, exhibited no androgen 16 beta- or 16 alpha-hydroxylase activity. The hydroxylase activities catalysed by the SD1 cell homogenate were selectively and quantitatively inhibited (greater than 90%) by a monoclonal antibody to P-450 PB-4 at a level of antibody (40 pmol of antibody binding sites/mg of SD1 homogenate) that closely corresponds to the P-450 PB-4 content of the cells (48 pmol of PB-4/mg of SD1 homogenate). Fractionation of cell homogenates into cytosol and microsomes revealed that the P-450 PB-4-mediated activities are associated with the membrane fraction. Although the P-450 PB-4-specific content of the SD1 microsomes was 15% of that present in phenobarbital-induced rat liver microsomes, the P-450 PB-4-dependent androstenedione 16 beta-hydroxylase activity of the SD1 membrane fraction was only 2-3% of that present in the liver microsomes. This activity could be stimulated several-fold, however, by supplementation of SD1 microsomes with purified rat NADPH P-450 reductase. These studies establish that a single P-450 gene product (IIB1) can account for the hydroxylation of androgen substrates at multiple sites, and suggest that SD1 cells can be used to assess the catalytic specificity of P-450 PB-4 with other substrates as well.


Subject(s)
Androstenedione/metabolism , Aryl Hydrocarbon Hydroxylases , Liver/enzymology , Steroid 11-beta-Hydroxylase/metabolism , Steroid Hydroxylases/metabolism , Testosterone/metabolism , Animals , Catalysis , Cell Line , Cricetinae , Cricetulus , Fibroblasts/metabolism , Hydroxylation , Rats , Steroid 16-alpha-Hydroxylase
12.
J Biol Chem ; 263(34): 17995-8002, 1988 Dec 05.
Article in English | MEDLINE | ID: mdl-3192524

ABSTRACT

Previous studies on regulation of the rat hepatic P-450 IIA1 cDNA have provided evidence for a second gene closely related to but regulated in a manner quite distinct from P-450 IIA1. Experiments were carried out to isolate the cDNA for this second P-450 gene, designated IIA2, in order to study more directly its regulation and relationship to IIA1. A full length cDNA to IIA2 was isolated from an adult male rat liver lambda gt11 library and sequenced completely. The IIA2 cDNA shared 93% nucleotide and 88% deduced amino acid similarities with the previously characterized IIA1 cDNA (Nagata, K., Matsunaga, T., Gillette, J., Gelboin, H. V., and Gonzalez, F. J. (1987) J. Biol. Chem. 262, 2787-2793). The protein, deduced from the cDNA, contained 492 amino acids and a calculated Mr of 56,352. Comparison of the IIA1 and IIA2 cDNAs revealed areas of low nucleotide similarity interspersed with areas of absolute identity, suggesting that gene conversions have played a role in the evolution of the IIA subfamily. Expression of IIA1 and IIA2 mRNAs in rat liver during development was studied with use of specific oligonucleotide probes. IIA1 mRNA was increased within 1 week after birth in both male and female rats; however, its postpubertal expression was decreased in males yet remained elevated in females. In contrast, IIA2 mRNA was markedly induced in male rat liver at puberty but was not detectable in females at any age examined. Furthermore, only IIA1 mRNA was induced by treatment of rats with 3-methylcholanthrene. Although IIA1 and IIA2 mRNAs were actively expressed in hepatic tissue, no evidence for their expression was found in lung, kidney, or intestine, suggesting that the IIA genes have tissue-specific promoters. Reconstituted enzyme assays on the purified protein products P-450 IIA1 and P-450 IIA2 showed that, although both enzymes share considerable sequence similarity, their positional specificities toward the prototype substrate testosterone are strikingly different.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/genetics , Gene Conversion , Gene Expression Regulation , Genes , Microsomes, Liver/enzymology , Steroid Hydroxylases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cytochrome P-450 Enzyme System/isolation & purification , Cytochrome P-450 Enzyme System/metabolism , Humans , Male , Molecular Sequence Data , Rats , Rats, Inbred Strains , Sequence Homology, Nucleic Acid , Sex Factors , Species Specificity , Steroid Hydroxylases/isolation & purification , Steroid Hydroxylases/metabolism
13.
Gastroenterology ; 95(5): 1326-31, 1988 Nov.
Article in English | MEDLINE | ID: mdl-3049217

ABSTRACT

Anti-liver/kidney microsome1-positive sera from children with chronic active hepatitis were studied in an effort to identify the microsomal antigens selected during induction and progression of this autoimmune disease. Immunoblot analysis of sodium dodecyl sulfate gel-resolved microsomal proteins from human and rat liver using anti-liver/kidney microsome1-positive sera revealed a single polypeptide of 48 kilodaltons (human microsomes) or 50 kilodaltons (rat microsomes). Levels of the 50-kilodalton rat microsomal polypeptide were suppressed in vivo by several drugs known to modulate expression of individual forms (enzymes) of hepatic cytochrome P-450, with the largest decrease effected by phenobarbital. Dot blot analysis using a panel of 10 electrophoretically homogeneous rat liver cytochrome P-450 forms under nondenaturing conditions established that the two methylcholanthrene-inducible forms, P-450 BNF-B and P-450 ISF-G (P-450 gene subfamily IA), are selectively recognized by the anti-liver/kidney microsome1 antibodies. These findings demonstrate that sera associated with autoimmune (anti-liver/kidney microsome1) chronic active hepatitis are specifically reactive with select rat hepatic P-450 forms and suggest that these autoantibodies may be principally directed against one or more constitutive forms of the corresponding human liver cytochromes.


Subject(s)
Cytochrome P-450 Enzyme System/analysis , Hepatitis, Chronic/enzymology , Kidney/enzymology , Microsomes, Liver/enzymology , Microsomes/enzymology , Autoantibodies , Blotting, Western , Child , Fluorescent Antibody Technique , Hepatitis, Chronic/immunology , Humans , Immunoblotting , Kidney/immunology , Microsomes/immunology , Microsomes, Liver/immunology
14.
J Biol Chem ; 263(23): 11396-406, 1988 Aug 15.
Article in English | MEDLINE | ID: mdl-3403535

ABSTRACT

Rat hepatic cytochrome P-450 form RLM2 is a testosterone 15 alpha-hydroxylase reported to be male-specific on the basis of purification studies (Jansson, I., Mole, J., and Schenkman, J. B. (1985) J. Biol. Chem. 260, 7084-7093). The sex dependence, developmental regulation, xenobiotic induction, and hormonal control of P-450 RLM2 expression were studied using P-450 form-specific immunochemical and catalytic assays. Polyclonal antibodies raised to rat hepatic P-450 3 (P-450 gene IIA1) were found to cross-react strongly with P-450 RLM2, but not with 10 other rat P-450 forms, suggesting that P-450 3 and P-450 RLM2 are highly conserved in primary structure. Western blotting of liver microsomes under conditions where P-450s 3 and RLM2 are resolved electrophoretically revealed that P-450 RLM2 is markedly induced at puberty in male rats, with no protein detected (less than or equal to 5% of adult male levels) in adult females or immature animals of either sex. A similar developmental dependence was observed for hepatic microsomal testosterone 15 alpha-hydroxylase activity, which was found to be catalyzed primarily by P-450 RLM2. P-450 RLM2 was resistant to induction by several xenobiotics and in the case of phenobarbital and beta-naphthoflavone, was suppressed by 50-60%. Studies on the steroid hormonal regulation of P-450 RLM2 revealed that its adult male-specific expression is imprinted (programmed) in response to neonatal testosterone exposure. Ovariectomy studies demonstrated that suppression by estrogen does not contribute significantly to the absence of P-450 RLM2 in adult female rats. Although the male-specific developmental induction of P-450 RLM2 in response to neonatal testosterone is strikingly similar to that of P-450 2c (testosterone 2 alpha/16 alpha-hydroxylase; gene IIC11), P-450 RLM2 expression is not dependent on the pulsatile pituitary growth hormone secretion required for P-450 2c synthesis. Rather, hypophysectomy of adult male rats increased P-450 RLM2 and its associated testosterone 15 alpha-hydroxylase activity by 50-100%.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Growth Hormone/pharmacology , Isoenzymes/biosynthesis , Liver/enzymology , Animals , Cross Reactions , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Male , Rats , Sex Characteristics , Steroid Hydroxylases/antagonists & inhibitors
15.
Arch Biochem Biophys ; 263(2): 424-36, 1988 Jun.
Article in English | MEDLINE | ID: mdl-3259858

ABSTRACT

Cytochrome P-450-dependent steroid hormone metabolism was studied in isolated human liver microsomal fractions. 6 beta hydroxylation was shown to be the major route of NADPH-dependent oxidative metabolism (greater than or equal to 75% of total hydroxylated metabolites) with each of three steroid substrates, testosterone, androstenedione, and progesterone. With testosterone, 2 beta and 15 beta hydroxylation also occurred, proceeding at approximately 10% and 3-4% the rate of microsomal 6 beta hydroxylation, respectively, in each of the liver samples examined. Rates for the three steroid 6 beta-hydroxylase activities were highly correlated with each other (r = 0.95-0.97 for 25 individual microsomal preparations), suggesting that a single human liver P-450 enzyme is the principal microsomal 6 beta-hydroxylase catalyst with all three steroid substrates. Steroid 6 beta-hydroxylase rates correlated well with the specific content of human P-450NF (r = 0.69-0.83) and with its associated nifedipine oxidase activity (r = 0.80), but not with the rates for debrisoquine 4-hydroxylase, phenacetin O-deethylase, or S-mephenytoin 4-hydroxylase activities or the specific contents of their respective associated P-450 forms in these same liver microsomes (r less than 0.2). These correlative observations were supported by the selective inhibition of human liver microsomal 6 beta hydroxylation by antibody raised to either human P-450NF or a rat homolog, P-450 PB-2a. Anti-P-450NF also inhibited human microsomal testosterone 2 beta and 15 beta hydroxylation in parallel to the 6 beta-hydroxylation reaction. This antibody also inhibited rat P-450 2a-dependent steroid hormone 6 beta hydroxylation in uninduced adult male rat liver microsomes but not the steroid 2 alpha, 16 alpha, or 7 alpha hydroxylation reactions catalyzed by other rat P-450 forms. Finally, steroid 6 beta hydroxylation catalyzed by either human or rat liver microsomes was selectively inhibited by NADPH-dependent complexation of the macrolide antibiotic triacetyloleandomycin, a reaction that is characteristic of members of the P-450NF gene subfamily (P-450 IIIA subfamily). These observations establish that P-450NF or a closely related enzyme is the major catalyst of steroid hormone 6 beta hydroxylation in human liver microsomes, and furthermore suggest that steroid 6 beta hydroxylation may provide a useful, noninvasive monitor for the monooxygenase activity of this hepatic P-450 form.


Subject(s)
Cytochrome P-450 Enzyme System , Microsomes, Liver/enzymology , Steroid Hydroxylases/metabolism , Androstenedione/metabolism , Animals , Cross Reactions , Cytochrome P-450 CYP3A , Female , Humans , Male , NADP/metabolism , Progesterone/metabolism , Rats , Steroid Hydroxylases/antagonists & inhibitors , Steroid Hydroxylases/immunology , Substrate Specificity , Testosterone/metabolism , Troleandomycin/metabolism
16.
Mol Pharmacol ; 32(5): 615-24, 1987 Nov.
Article in English | MEDLINE | ID: mdl-3119986

ABSTRACT

Cytochrome P-450 (P-450) form specificities were established for a total of nine monoclonal antibodies (MAbs) raised to four distinct rat hepatic P-450 enzymes (P-450s 2c, PB-2a, PB-4, and BNF-B), using a combination of enzyme-linked immunosorbent analysis, dot immunoblotting, Western blotting, Ouchterlony immunodiffusion, and immunoinhibition analyses. Four of the MAbs were fully (greater than or equal to 85%) inhibitory toward the corresponding immunoreactive P-450s when assayed in purified, reconstituted enzyme systems, while two of the MAbs were partially inhibitory, with a maximum of 50 or 80% inhibition achieved in the presence of saturating MAb. Inhibitory MAbs reactive with P-450s 2c, 3, and PB-4, respectively, were used to demonstrate that the formation of multiple hydroxytestosterone metabolites by each of the respective purified P-450 enzymes is reflective of their inherent catalytic specificities and not due to the presence of immunochemical distinguishable P-450 enzyme contaminants. P-450 form-specific contributions to rat hepatic microsomal steroid hormone hydroxylase activities were then assessed using the inhibitory MAbs as probes. MAb-reactive P-450 2c was shown to be the major (greater than or equal to 85%) catalyst of microsomal testosterone and androstenedione 16 alpha-hydroxylation in both untreated and beta-naphthoflavone-induced rats. However, this P-450 form catalyzed only approximately 30% of hepatic microsomal steroid 16 alpha-hydroxylase activity in phenobarbital-induced adult males, and less than or equal to 10% of steroid 16 alpha-hydroxylase activity in (phenobarbital-induced immature males or adult females, where the balance of 16 alpha-hydroxylase activity is catalyzed by MAb-reactive P-450 PB-4. Although MAb-reactive P-450 PB-4 catalyzed the majority (greater than or equal to 90%) of microsomal androstenedione 16 beta-hydroxylation in phenobarbital-induced rats, this P-450 enzyme did not contribute to the low level 16 beta-hydroxylase activity of uninduced liver samples. Finally, MAb-reactive P-450 3 catalyzed at least 85% of microsomal androstenedione 7 alpha-hydroxylation, independent of the age, sex, or induction status of the animals used as source of liver microsomes. These findings demonstrate the usefulness of MAbs as probes for the contributions of individual P-450 enzymes to the metabolism of steroid hormones susceptible to hydroxylation at multiple sites.


Subject(s)
Antibodies, Monoclonal , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme Inhibitors , Microsomes, Liver/enzymology , Steroid Hydroxylases/antagonists & inhibitors , 7-Alkoxycoumarin O-Dealkylase , Animals , Cytochrome P-450 Enzyme System/immunology , Cytochrome P450 Family 2 , Immunodiffusion , Kinetics , Male , Oxygenases/antagonists & inhibitors , Oxygenases/immunology , Rats , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/immunology
17.
J Immunol ; 135(1): 355-61, 1985 Jul.
Article in English | MEDLINE | ID: mdl-3998467

ABSTRACT

We previously purified a potent serum suppressor factor from malignant ascites fluid and showed that it had serologic cross-reactivity with E receptor of human T lymphocytes. We termed this factor "suppressive E receptor factor" (SER). Subsequent studies on SER showed that SER interfered with the production of interleukin 1 and 2 as well as interfering with their activities on target cells. However, SER was not directly cytotoxic to lymphocytes. In this study, we compared the inhibitor of DNA-polymerase (IDP) activity with the suppressive activity on phytohemagglutinin-induced DNA synthesis on intact cells. These two activities were closely correlated (with a linear correlation coefficient of 0.988) even with the whole plasmas derived from cancer patients. Fractionation and purification of IDP activity identified it with SER of a similar potency. Therefore, SER appeared to exhibit its potent immunosuppressive effect via its direct interference on DNA-polymerase activity. Furthermore, the DNA-polymerase inhibitory activity of SER appeared to be specific to DNA and it did not affect the RNA-polymerase activity. SER inhibition of DNA polymerase activity with respect to DNA primer as well as with the nucleotide substrate. Direct inhibition on DNA-polymerase-alpha activity may be one of the possible mechanisms of action of SER on lymphocyte proliferation.


Subject(s)
DNA Polymerase II/antagonists & inhibitors , Glycoproteins/physiology , Pleural Effusion/immunology , Receptors, Antigen, T-Cell , Binding Sites , Chemical Fractionation , Chromatography, Gel , DNA Polymerase II/blood , Glycoproteins/blood , Glycoproteins/isolation & purification , Humans , In Vitro Techniques , Kinetics , Lymphocyte Activation , Neoplasm Proteins , Phytohemagglutinins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...