Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
Add more filters










Publication year range
1.
Biochemistry ; 51(3): 750-60, 2012 Jan 24.
Article in English | MEDLINE | ID: mdl-22208729

ABSTRACT

Cytochrome P450 BM3 from Bacillus megaterium is a monooxygenase with great potential for biotechnological applications. In this paper, we present engineered drug-metabolizing P450 BM3 mutants as a novel tool for regioselective hydroxylation of steroids at position 16ß. In particular, we show that by replacing alanine at position 82 with a tryptophan in P450 BM3 mutants M01 and M11, the selectivity toward 16ß-hydroxylation for both testosterone and norethisterone was strongly increased. The A82W mutation led to a ≤42-fold increase in V(max) for 16ß-hydroxylation of these steroids. Moreover, this mutation improves the coupling efficiency of the enzyme, which might be explained by a more efficient exclusion of water from the active site. The substrate affinity for testosterone increased at least 9-fold in M11 with tryptophan at position 82. A change in the orientation of testosterone in the M11 A82W mutant as compared to the orientation in M11 was observed by T(1) paramagnetic relaxation nuclear magnetic resonance. Testosterone is oriented in M11 with both the A- and D-ring protons closest to the heme iron. Substituting alanine at position 82 with tryptophan results in increased A-ring proton-iron distances, consistent with the relative decrease in the level of A-ring hydroxylation at position 2ß.


Subject(s)
Amino Acid Substitution/genetics , Bacillus megaterium/enzymology , Bacillus megaterium/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , NADPH-Ferrihemoprotein Reductase/chemistry , NADPH-Ferrihemoprotein Reductase/genetics , Norethindrone/metabolism , Testosterone/metabolism , Alanine/genetics , Bacillus megaterium/metabolism , Bacterial Proteins/metabolism , Biotransformation/genetics , Catalytic Domain/genetics , Cytochrome P-450 Enzyme System/metabolism , Hydroxylation/genetics , Mutagenesis, Site-Directed , NADPH-Ferrihemoprotein Reductase/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Spectrophotometry, Ultraviolet , Tryptophan/genetics
2.
Chem Res Toxicol ; 24(8): 1263-74, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21639118

ABSTRACT

Covalent binding of reactive metabolites (RMs) to proteins is considered to be one of the important mechanisms by which drugs can cause tissue damage. To facilitate the study of drug-protein adducts, we developed a potentially generic method for producing high levels of covalently modified proteins. A highly active drug metabolizing P450 BM3 mutant (CYP102A1M11H) is used for drug bioactivation. Because of its His-tag, CYP102A1M11H is easily removed by nickel affinity chromatography, facilitating subsequent characterization of the modified target protein. The applicability of our procedure is demonstrated by the trapping of RMs of acetaminophen (APAP), clozapine (CLOZ), and troglitazone (TGZ) with human glutathione-S-transferase P1-1 (hGST P1-1) as the model target protein. Tryptic digests of hGST P1-1 were subjected to analysis by LC-MS/MS and modified peptides identified by the comparative analysis of tryptic peptides of adducted and nonadducted hGST P1-1. Characteristic MS/MS ions of drug-modified peptides were identified by first searching for expected adduct-masses. Unanticipated drug-peptide adducts were subsequently identified in an unbiased manner by screening for diagnostic MS/MS ions of modified peptides. Reactive intermediates of APAP and CLOZ adducted to cysteine-47 and mass shifts corresponded to the alkylation of N-acetyl-p-benzoquinone imine (NAPQI) and the CLOZ nitrenium ion, respectively. Adduction of TGZ appeared more complex, yielding three different types of adducts to cysteine-47, two adducts to cysteine-14, and a single adduct to cysteine-101. Together, these findings show that P450 BM3 mutants with high capacity to activate drugs into relevant RMs can be employed to produce protein adducts to study the nucleophilic selectivity of highly reactive electrophiles.


Subject(s)
Acetaminophen/chemistry , Bacterial Proteins/metabolism , Chromans/chemistry , Clozapine/chemistry , Cytochrome P-450 Enzyme System/metabolism , Glutathione Transferase/chemistry , NADPH-Ferrihemoprotein Reductase/metabolism , Thiazolidinediones/chemistry , Acetaminophen/metabolism , Bacterial Proteins/genetics , Chromans/metabolism , Chromatography, High Pressure Liquid , Clozapine/metabolism , Cysteine/metabolism , Cytochrome P-450 Enzyme System/genetics , Glutathione Transferase/metabolism , Humans , Inactivation, Metabolic , Microsomes, Liver/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , Peptides/analysis , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tandem Mass Spectrometry , Thiazolidinediones/metabolism , Troglitazone , Trypsin/metabolism
3.
Xenobiotica ; 39(4): 302-11, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19350453

ABSTRACT

Glutathione S-transferases (GSTs) are important phase II drug-metabolizing enzymes that play a major role in protecting cells from the toxic insults of electrophilic compounds. Curcumin, a promising chemotherapeutic agent, inhibits human GSTA1-1, GSTM1-1, and GSTP1-1 isoenzymes. In the present study, the effect of three series of curcumin analogues, 2,6-dibenzylidenecyclohexanone (A series), 2,5-dibenzylidenecyclopentanone (B series), and 1,4-pentadiene-3-one (C series) substituted analogues (n = 34), on these three human GST isoenzymes, and on human and rat liver cytosolic GSTs, was investigated using 1-chloro-2,4-dinitrobenzene (CDNB) as a substrate. Most of the 34 curcumin analogues showed less potent inhibitory activities towards GSTA1-1, GSTM1-1, and GSTP1-1 than the parent curcumin. Compounds B14 and C10 were the most potent inhibitors of GSTA1-1 and human liver cytosolic GSTs, with IC(50) values of 0.2-0.6 microM. The most potent inhibitors of GSTM1-1 were C1, C3 and C10, with IC(50) values of 0.2-0.7 microM. Similarly, GSTP1-1 was predominantly strongly inhibited by compounds of the C series C0, C1, C2 C10 and A0, with IC(50) values of 0.4-4.6 microM. Compounds in the B series showed no significant inhibition of GSTP1-1. Molecular Operating Environment (MOE) program-based quantitative structure-activity relationship (QSAR) analyses have also suggested the relevance of Van der Waals surface area and compound lipophilicity factors for the inhibition of GSTA1-1 and GSTM1-1 and partial charge factors for GSTP1-1. These results may be useful in the design and synthesis of curcumin analogues with either more or less potency for GST inhibition.


Subject(s)
Curcumin/pharmacology , Glutathione S-Transferase pi/antagonists & inhibitors , Glutathione Transferase/antagonists & inhibitors , Liver/metabolism , Animals , Dinitrochlorobenzene , Humans , Inhibitory Concentration 50 , Isoenzymes/antagonists & inhibitors , Molecular Structure , Quantitative Structure-Activity Relationship , Rats
4.
Xenobiotica ; 36(9): 763-71, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16971342

ABSTRACT

Previous studies have shown the critical roles residues F120 and F483 play in the oxidative metabolism of 7-methoxy-4-(aminomethyl)-coumarin (MAMC) by cytochrome P450 2D6 (CYP2D6). In the present study, a series of N-alkyl-7-methoxy-4-(aminomethyl)-coumarins (MAMC analogues) were used as substrates for the F120A and F483A mutants in order to probe the CYP2D6 active site. The F120A and F483A mutants of CYP2D6 displayed significant activity towards the MAMC analogues. Automated docking studies of the MAMC analogues in a CYP2D6 homology model suggested a distal hydrophobic active site binding cleft for the substrate N-alkyl chains, consisting of the residues L213 and V308.


Subject(s)
Coumarins/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Mutant Proteins/metabolism , Alkylation , Binding Sites , Catalysis , Coumarins/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Methylation , Models, Molecular , Mutant Proteins/genetics , Mutation/genetics , Substrate Specificity
5.
Xenobiotica ; 36(7): 645-57, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16864509

ABSTRACT

Enzyme kinetic parameters of the bioactivation of thiourea-containing compounds by human flavin-containing monooxygenase enzymes (FMOs) FMO1 and FMO3 were investigated. A microtitre-based adaptation of methodology described for the thiourea-dependent oxidation of thiocholine was used to determine the turnover of thiourea-containing compounds by human FMO1 and FMO3. The results show that major differences in enzyme kinetic parameters for N-substituted N'-(4-imidazole-ethyl)thiourea exist between human FMO3 and human FMO1. Whereas Km values of N-substituted N'-(4-imidazole-ethyl)thioureas for human FMO3 are all in the millimolar range, the Km values for human FMO1 range from the low micromolar to the low millimolar range. Furthermore, among a series of N-p-phenyl-substituted N'-(4-imidazole-ethyl)thioureas an interesting structure-activity relationship is evident with both FMO1 and FMO3. Where the Km decreases with increasing electron-withdrawing capacity of the p-substituent in the case of FMO1, the opposite phenomenon may be the case with FMO3. The kcat values of the compounds were all comparable for FMO1, averaging 3.03 +/- 0.56 min-1, whereas more variation was found for FMO3 (3.71 +/- 2.01 min-1). Enzyme kinetic parameters Km and kcat/Km of human FMO1 for N-substituted N'-(4-imidazole-ethyl)thioureas show a high degree of correlation with the results obtained in rat liver microsomes, in which rat FMO1 is the most abundant form, whereas those of human FMO3 do not.


Subject(s)
Imidazoles/pharmacokinetics , Oxygenases/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacokinetics , Animals , Biotransformation , Humans , Oxygenases/genetics , Rats , Recombinant Proteins/genetics , Structure-Activity Relationship
6.
Xenobiotica ; 35(4): 391-404, 2005 Apr.
Article in English | MEDLINE | ID: mdl-16019959

ABSTRACT

A previous study showed that the cytotoxicity of a series of N-p-phenyl-substituted N'-(4-imidazole-ethyl)thiourea in precision-cut rat liver slices increased with increasing electron-withdrawing capacity of the p-substituent and may be related to the Vmax/Km values of bioactivation of the thiourea-moiety by hepatic flavin-containing monooxygenases (FMOs). However, differences in the uptake of xenobiotics into precision-cut liver slices can also have consequences for the rates of metabolism of xenobiotics. In the present study, therefore, we investigated the rate and nature of uptake of 9 N-substituted N'-(4-imidazole-ethyl)thiourea into precision-cut rat liver slices. It was found that a five-fold difference exists among a series of N-substituted N'-(4-imidazole-ethyl)thiourea both in the initial rate of uptake and in the steady-state levels ultimately achieved in the precision-cut rat liver slices. It appeared that the most cytotoxic compounds were also the most readily absorbed compounds. The concentration-dependent initial rate of uptake could be described by a carrier-mediated saturable component and a non-saturable component. At cytotoxic concentrations, the non-saturable component accounted for more than 95% of the total uptake. From this study, it is concluded that differences in rate of uptake of thiourea-containing compounds may be a contributing factor to the differences in bioactivation by FMOs as the basis of the structure-toxicity relationships observed in precision-cut rat liver slices.


Subject(s)
Liver/metabolism , Thiourea/toxicity , Animals , Dose-Response Relationship, Drug , Male , Microdissection , Organ Culture Techniques , Rats , Rats, Wistar , Structure-Activity Relationship , Thiourea/analogs & derivatives
7.
Xenobiotica ; 34(3): 301-16, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15204701

ABSTRACT

1: The use of fluorine-19 nuclear magnetic resonance (19F-NMR) and gas chromatography-electron capture detection (GC-ECD) in the analysis of fluorine-containing products in the urine of sevoflurane-exposed patients was explored. 2: Ten patients were anaesthetized by sevoflurane for 135-660 min at a flow rate of 6 l min(-1). Urine samples were collected before, directly after and 24 h after discontinuation of anaesthesia. 3: 19F-NMR analysis of the urines showed the presence of several fluorine-containing metabolites. The main oxidative metabolite, hexafluoroisopropanol (HFIP)-glucuronide, showed two strong quartet signals in the 19F-NMR spectrum. HFIP concentrations after beta-glucuronidase treatment were quantified by (19)F-nuclear magnetic resonance. Concentrations directly after and 24 h after discontinuation of anaesthesia were 131 +/- 41 (mean +/- SEM) and 61 +/- 19 mol mg(-1) creatinine, respectively. Urinary HFIP excretions correlated with sevoflurane exposure. 4: Longer scanning times enabled the measurement of signals from two compound A-derived metabolites, i.e. compound A mercapturic acid I (CAMA-I) and compound A mercapturic acid II (CAMA-II), as well as products from beta-lyase activation of the respective cysteine conjugates of compound A. The signals of the mercapturic acids, 3,3,3-trifluoro-2-(fluoromethoxy)-propanoic acid and 3,3,3-trifluorolactic acid were visible after combining and concentrating the patient urines. CAMA-I and -II excretions in patients were completed after 24 h. 5: Since 19F-nuclear magnetic resonance is not sensitive enough, urinary mercapturic acids concentrations were quantified by gas chromatography-electron capture detection. CAMA-I and -II urinary concentrations were 2.3 +/- 0.7 and 1.4 +/- 0.4 mol mg(-1) creatinine, respectively. Urinary excretion of CAMA-I showed a correlation with sevoflurane exposure, whereas CAMA-II did not. 6. The results show that 19F-nuclear magnetic resonance is a very selective and convenient technique to detect and quantify HFIP in non-concentrated human urine. 19F-nuclear magnetic resonance can also be used to monitor the oxidative biotransformation of sevoflurane in anaesthetized patients. Compound A-derived mercapturic acids and 3,3,3-trifluoro-2-(fluoromethoxy)-propanoic acid and 3,3,3-trifluorolactic acid, however, require more sensitive techniques such as gas chromatography-electron capture detection and/or gas chromatography-mass spectrometry for quantification.


Subject(s)
Anesthetics, Inhalation/metabolism , Fluorine Compounds/urine , Methyl Ethers/metabolism , Anesthesia, Inhalation , Anesthetics, Inhalation/urine , Chromatography, Gas , Fluorine Radioisotopes , Humans , Magnetic Resonance Spectroscopy , Methyl Ethers/urine , Sensitivity and Specificity , Sevoflurane
8.
Xenobiotica ; 33(1): 57-72, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12519694

ABSTRACT

1. To investigate whether cytochrome P450 (P450) inhibition can contribute to the chemopreventive activity of selenocysteine Se-conjugates (SeCys conjugates), 21 SeCys conjugates were screened for their inhibitory potency towards seven of the most important human P450s. 2. The majority of the SeCys conjugates produced near complete inhibition of CYP1A1 at a concentration of 250 microm. The most potent inhibitor, Se-benzyl-L-selenocysteine, displayed an IC50 of 12.8 +/- 1.2 microm. CYP2C9, -2C19 and -2D6 were moderately (50-60%) inhibited by the SeCys conjugates. CYP1A2, -2E1 and -3A4 were least inhibited. 3. Studies on the susceptibility of CYP1A1 to SeCys conjugates implicated a thiol-reactive intermediate, as evidenced by reduced inhibition levels in the presence of glutathione and N-acetyl cysteine. Uncoupling of the P450-catalytic cycle was of no importance as ROS scavengers did not influence inhibition levels. 4. P450 inhibition by two physiologically relevant metabolite classes of SeCys conjugates was also studied. N-acetylation of SeCys conjugates consistently increased the inhibitory potency towards CYP1A2, -2C19, -2E1 and -3A4. Beta-lyase catalysed bioactivation of alkyl-substituted SeCys conjugates or Se-benzyl-L-selenocysteine produced little or no additional inhibition of P450 activity. For Se-phenyl-L-selenocysteine, however, significant increases in P450 inhibition were obtained by beta-lyase pre-incubation. 5. It is concluded that the potent and relatively selective CYP1A1 inhibition exerted by SeCys conjugates may contribute to their chemopreventive activity.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Selenium/metabolism , Selenocysteine/metabolism , Acetylation , Binding, Competitive , Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 Enzyme Inhibitors , Fluorescent Dyes , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Kinetics , Selenocysteine/chemical synthesis , Selenocysteine/pharmacology
9.
J Pharmacol Exp Ther ; 299(3): 921-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11714877

ABSTRACT

Midazolam is almost exclusively metabolized by cytochrome P450 3A (CYP3A) isoenzymes. Therefore, midazolam is used as a probe to determine CYP3A levels in humans and rats. A prerequisite for longitudinal determination of CYP3A expression levels using midazolam as a probe is that midazolam itself has no effect on the expression of CYP3A. In the present study, we analyzed the mRNA levels and enzyme activities of the major CYP isoforms in the rat liver after intraperitoneal injection of midazolam (50 mg/kg) for 3 consecutive days. CYP3A1 mRNA levels were increased 4-fold in midazolam-treated animals compared with controls, whereas the mRNA levels of CYP3A2, CYP3A9, and CYP3A18 were not altered. The increase in CYP3A1 mRNA was accompanied by a 25% increase in microsomal testosterone 6beta-hydroxylation activity. More strikingly, CYP2B1/2 mRNA levels were increased 22-fold upon midazolam treatment, leading to an 11- to 95-fold enhancement of CYP2B enzyme activity. CYP2C6 mRNA levels were 4 times higher in midazolam-treated animals. Formation of 2alpha-hydroxy-testosterone, mainly catalyzed by CYP2C11, was 2.6-fold lower in liver microsomes from midazolam-treated animals. Midazolam induced CYP2E enzyme activity 2.5-fold at the post-transcriptional level. The induction of CYP2B1/2 mRNA levels by midazolam was dose-dependent (4.5-fold increase at 10 mg/kg). Induction of CYP3A1 and CYP2B expression was also observed in isolated rat hepatocytes cultured with 100 microM midazolam. We conclude that midazolam is a phenobarbital-like CYP inducer in rats. Induction of CYP3A1 by midazolam may have implications for the longitudinal use of midazolam as a probe for analysis of CYP3A expression levels in rats.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Midazolam/pharmacology , Phenobarbital/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Enzyme Induction/drug effects , Hypnotics and Sedatives/pharmacology , Male , Oxidoreductases, N-Demethylating/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley
10.
Chem Res Toxicol ; 14(8): 996-1005, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11511173

ABSTRACT

Several selenocysteine Se-conjugates have been shown to possess potent chemopreventive activity in animal models for chemical carcinogenesis. As a mechanism of action, beta-elimination reactions to form chemopreventive selenols, ammonia, and pyruvate has been proposed. The enzymes involved in these beta-elimination reactions, however, have been partially elucidated. Next to cysteine conjugate beta-lyases, as yet unidentified non-pyridoxal-5'-phosphate-dependent enzymes also appear to be involved in cytosolic beta-elimination reactions. In the present study, it was investigated whether amino acid oxidases contribute to the bioactivation of selenocysteine Se-conjugates. Using purified L-amino acid oxidase from Crotalus adamanteus as a model enzyme, significant beta-elimination activities were indeed observed upon incubation with Se-methylselenocysteine (K(m), 195 microM; k(cat), 48 min(-1)), Se-allylselenocysteine (K(m), 608 microM; k(cat), 34 min(-1)), Se-phenylselenocysteine (K(m), 107 microM; k(cat), 57 min(-1)) and Se-benzylselenocysteine (K(m), 59 microM; k(cat), 13 min(-1)). For all selenocysteine Se-conjugates tested, the rate of pyruvate formation was comparable to that of hydrogen peroxide, one of the products of oxidative deamination. The fact that addition of catalase did not alter pyruvate formation indicated that the beta-elimination reaction observed was not mediated by selenoxidation/syn-elimination due to the hydrogen peroxide formed via the oxidative deamination pathway. Using D-amino acid oxidase from porcine kidney and D-SeCys conjugates similar results were obtained. To delineate whether mammalian L-amino acid oxidases are also able to catalyze beta-elimination of selenocysteine Se-conjugates, rat renal cytosol was fractionated and screened for beta-elimination and oxidative deamination activities. One of the fractions isolated displayed oxidative deamination activity with several amino acids and cysteine S-conjugates. With selenocysteine Se-conjugates as substrates, however, this fraction displayed both oxidative deamination and beta-elimination activities, when incubated in the presence of aminoxyacetic acid to block contribution of pyridoxal-5'-phosphate-dependent enzymes. The potential significance of this novel bioactivation route for the chemopreventive activity of selenocysteine Se-conjugates is discussed.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Chemoprevention , Selenium/chemistry , Selenocysteine/metabolism , Animals , Biotransformation , Catalase/metabolism , Chromatography, High Pressure Liquid , Kidney/chemistry , Oxidation-Reduction , Rats , Selenocysteine/chemistry
11.
Anesthesiology ; 95(1): 165-75, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11465554

ABSTRACT

BACKGROUND: Renal toxicity has occasionally been observed after enflurane anesthesia. Although originally attributed to its oxidative metabolism to inorganic fluoride, serum levels of inorganic fluoride appear to be small to explain these renal effects. Formation of potentially nephrotoxic halogenated alkenes during alkaline degradation in carbon dioxide absorbers and subsequent bioactivation via the glutathione conjugation pathway may be considered as an alternative mechanism for renal toxicity. The aim of this study was to characterize the thioethers formed chemically and biosynthetically. METHODS: Alkaline degradation of enflurane was achieved by stirring with pulverized potassium hydroxide. Volatile degradation products were analyzed by 19F nuclear magnetic resonance (NMR) analysis of head space gasses trapped in dimethyl sulfoxide (DMSO). Thioethers were generated chemically by trapping head space gasses in DMSO containing N-acetyl-L-cysteine or 2-mercaptoacetic acid as model thiol compounds. Glutathione conjugates were generated biosynthetically by passing head space through rat liver fractions in presence of glutathione. Products formed were analyzed by gas chromatography-mass spectroscopy and 19F-NMR. RESULTS: Direct analysis of head space gasses showed formation of 1-chloro-1,2-difluorovinyl difluoromethyl ether and two unidentified fluorine-containing products as alkaline degradation products of enflurane. When trapped in DMSO-N-acetyl-L-cysteine-triethylamine, N-acetyl-S-(2-chloro-1,2-difluoro-1-(difluoromethoxy)ethyl)-L-cysteine was identified as the major product. Another N-acetyl-L-cysteine S-conjugate formed was N-acetyl-S-(2-chloro-1,1,2-trifluoroethyl)-L-cysteine, a potent nephrotoxin in rats. 19F-NMR analysis of glutathione conjugates formed after incubation with rat liver fractions resulted in formation of corresponding S-conjugates. CONCLUSIONS: The current study demonstrates that alkaline degradation products of enflurane can be conjugated to thiol compounds, forming S-conjugates that could theoretically contribute to adverse renal effects observed occasionally with enflurane anesthesia. The N-acetyl-L-cysteine S-conjugates identified may be biomarkers to assess exposure of humans to alkaline degradation products of enflurane.


Subject(s)
Anesthetics, Inhalation/chemistry , Enflurane/chemistry , Sulfides/chemistry , Acetylcysteine/chemistry , Alkalies , Animals , Cytosol/metabolism , Gas Chromatography-Mass Spectrometry , Glutathione/chemistry , Magnetic Resonance Spectroscopy , Male , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Rats , Rats, Wistar , Thioglycolates/chemistry
12.
Int Arch Occup Environ Health ; 74(2): 102-8, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11317702

ABSTRACT

OBJECTIVES: To investigate in humans the contribution of the cytochrome P-450- and glutathione-dependent biotransformation of trichloroethylene (TRI) under controlled repeated exposure in volunteers, and under occupational conditions. METHODS: Volunteers were exposed to TRI, using repeated 15 min exposures at 50 and 100 ppm. This exposure schedule resulted in internal doses of 1.30 and 2.40 mmol of TRI respectively. Urine samples were collected for a minimum of 45 h. Urine samples were also collected from occupationally exposed workers. The samples were analysed for the known human metabolites of TRI, trichloroethanol (TCE), trichloroacetic acid (TCA) and both regio-isomeric forms of the mercapturic acid N-acetyl-S-(dichlorovinyl)-L-cysteine (DCV-NAC), and for (dichlorovinyl)-L-cysteine (DCVC). In order to further elucidate the metabolism of TRI in humans, we analysed samples for dichloroacetic acid and for the proposed break-down products of 1,2 and 2,2-dichlorovinyl-L-cysteine after deamination: the S-conjugates of 3-mercaptolactic acid, 3-mercaptopyruvic acid and 2-mercaptoacetic acid. RESULTS: None of the glutathione metabolites was found in urine of volunteers. In workers occupationally exposed to TRI at levels between 0.4 and 21 ppm [8-h time-weighted average (TWA)], levels of DCV-NAC in urine samples collected at the end of the 4th working day and also next morning were below detection limit (0.04 mumol/l). This confirms the findings of Bernauer et al. (1996) that these metabolites are excreted at very low levels in humans. Urinary levels of DCVC and six postulated metabolites of dichlorovinyl-S-cysteine conjugates via deamination were also below 0.04 mumol/l, indicating that at most 0.05% of the dose is excreted in the form of these metabolites. These data further strengthen the argument for a very low activity of glutathione-mediated metabolism for chronically exposed workers. CONCLUSIONS: This study gives additional data which indicate that glutathione-mediated metabolism is of minor importance in humans exposed to TRI. In spite of indications to the contrary, significant metabolism of the cysteine conjugate via beta-lyase, which could result in a toxic metabolite, cannot be ruled out completely.


Subject(s)
Air Pollutants, Occupational/analysis , Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Occupational Exposure/analysis , Trichloroethylene/pharmacokinetics , Adult , Biomarkers , Biotransformation/physiology , Ethylene Chlorohydrin/analogs & derivatives , Ethylene Chlorohydrin/urine , Humans , Male , Middle Aged , Trichloroacetic Acid/urine
13.
Chem Biol Interact ; 134(2): 167-90, 2001 Apr 16.
Article in English | MEDLINE | ID: mdl-11311212

ABSTRACT

The proximal tubule is a frequent target for nephrotoxic compounds due to it's ability to transport and accumulate xenobiotics and their metabolites, as well as by the presence of an organ-selective set of biotransformation enzymes. The aim of the present study was to characterize the activities of different biotransformation enzymes during primary culturing of rat proximal tubular cells (PT cells). Specific marker substrates for determining cytochrome P450 (CYP450) activity of primary cultured PT cells include 7-ethoxyresorufin (CYP1A1), caffeine (CYP1A), testosterone (CY2B/C, CYP3A), tolbutamide (CYP2C) and dextromethorphan (CYP2D1). Activities of the CYP450 isoenzymes decreased considerably during culture with the greatest loss in activity within 24 h of culture. In addition, expression of CYP450 apoprotein, including CYP1A, CYP2C, CYP2D, CYP2E and CYP4A, was detected in microsomes from freshly isolated PT cells by immunoblotting using specific antibodies. CYP2B and CYP3A apoprotein could not be detected. Activity of the phase II biotransformation enzymes GST, GGT, beta-lyase and UGT was determined with 1-chloro-2,4-dinitrobenzene, L-glutamic acid gamma-(7-amido-4-methyl-coumarin), S-(1,1,2,2-tetrafluoroethyl)-L-cysteine and 1-naphthol, respectively, as marker substrates. Activity of the phase II enzymes remained more stable and, in contrast to CYP450 activity, significant activity was still expressed after 1 week of PT cell culture. Thus, despite the obvious advantages of PT cells as an in-vitro model for studies of biotransformation mediated toxicity, the strong time dependency of especially phase I and, to a lesser extent, phase II biotransformation activities confers limitations to their application.


Subject(s)
Kidney Tubules, Proximal/enzymology , Animals , Biotransformation , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Glucuronosyltransferase/metabolism , Glutathione Transferase/metabolism , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Rats , Substrate Specificity , Xenobiotics/pharmacokinetics , Xenobiotics/toxicity , gamma-Glutamyltransferase/metabolism
14.
Chem Res Toxicol ; 14(1): 127-34, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11170516

ABSTRACT

Previously, it was shown that beta-elimination of selenocysteine Se-conjugates by rat renal cytosol leading to pyruvate formation was not solely catalyzed by pyridoxal phosphate-dependent enzymes. It was hypothesized that selenoxidation of the selenocysteine Se-conjugates, followed by syn-elimination, may be an alternative mechanism for pyruvate formation. In this study, selenoxidation of selenocysteine Se-conjugates was studied using rat liver microsomes and recombinant human oxidative enzymes. For all six selenocysteine Se-conjugates that were tested, it was found that rat liver microsomal incubations led to the formation of pyruvate, whereas the corresponding selenoxides were not observed. Microsomal pyruvate formation from Se-benzyl-L-selenocysteine (SeBC) was NADPH-dependent, but only marginally inhibited by several P450 inhibitors. Inhibition by methimazole and by heat pretreatment and stimulation by n-octylamine indicated that flavin-containing monooxygenases are mainly responsible for pyruvate formation from the selenocysteine Se-conjugates in rat liver microsomes. In the case of S-benzyl-L-cysteine, the sulfur analogue of SeBC, pyruvate formation was not observed. For this substrate, a chemically stable sulfoxide could be observed, as previously described. By using recombinantly expressed human flavin-containing monooxygenases and P450 enzymes, it was delineated that SeBC is selenoxidized by human FMOs, but not by human P450s. The k(cat)/K(m) of selenoxidation was 3.8-fold higher for FMO-1 than for FMO-3. In conclusion, selenoxidation of selenocysteine Se-conjugates catalyzed by FMOs and subsequently syn-elimination has taken place as an alternative route for the formation of pyruvate from selenocysteine Se-conjugates. Although selenoxides are known to be easily reduced by thiol compounds, microsomal pyruvate formation from SeBC was only 75% inhibited in the presence of an excess of glutathione. This indicates that even in the presence of physiological concentrations of reducing thiol compounds, selenoxides of selenocysteine Se-conjugates may undergo syn-elimination to some extent. Whether selenoxides and/or selenenic acids that are formed are involved in the activity of chemopreventive selenocysteine Se-conjugates remains to be established.


Subject(s)
Cysteine/analogs & derivatives , Oxygenases/metabolism , Selenocysteine/metabolism , Animals , Cysteine/chemistry , Cysteine/metabolism , Cysteine/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Humans , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Oxidation-Reduction , Oxygenases/antagonists & inhibitors , Pyruvic Acid/metabolism , Rats , Rats, Wistar , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Selenium Compounds/metabolism , Selenium Oxides , Selenocysteine/analogs & derivatives , Selenocysteine/pharmacokinetics
16.
Eur J Pharm Sci ; 12(2): 151-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11102743

ABSTRACT

We recently reported on the design, synthesis and characterisation of a novel and selective substrate of human cytochrome P450 2D6 (CYP2D6), 7-methoxy-4-(aminomethyl)-coumarin (MAMC). Here, we describe a high-throughput microplate reader assay, which makes use of MAMC as a fluorescent probe for determining the inhibition and activity of CYP2D6 in heterologously expressed systems and human liver microsomes. The high-throughput screening (HTS) assay can be used both in an end-point and real-time configuration, and is easy to use, rapid and sensitive. In addition, end-point measurements by means of flow injection analysis have also successfully been performed. The HTS-assay was validated by performing inhibition experiments for several low- and high-affinity ligands (n=6) of CYP2D6, and comparing the findings to those obtained with the standard O-demethylation assay of dextromethorphan. The results indicate that all compounds tested display competitive inhibition in both the MAMC and dextromethorphan assay, and the K(i) values reveal a very good correlation (R(2)=0.984) between the two assays. To further demonstrate the usefulness of the HTS-assay, IC(50) values of a series of five N-substituted analogs of 3, 4-methylenedioxyamphetamine for CYP2D6 have been determined. The results obtained demonstrate that the current HTS-assay represents a significant improvement over previous assays, with a higher turnover of MAMC and a higher selectivity for CYP2D6.


Subject(s)
Coumarins/pharmacokinetics , Cytochrome P-450 CYP2D6/metabolism , Dextromethorphan/pharmacokinetics , Microsomes, Liver/enzymology , Microsomes/enzymology , Calibration , Cell Line , Cytochrome P-450 CYP2D6/analysis , Humans , Kinetics , Recombinant Proteins/analysis , Recombinant Proteins/metabolism , Sensitivity and Specificity , Spectrometry, Fluorescence/methods , Substrate Specificity
17.
Drug Metab Dispos ; 28(12): 1524-32, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11095593

ABSTRACT

A series of six structural analogs of 7-methoxy-4-(aminomethyl)-coumarin (MAMC), a recently developed high-throughput substrate of P450 2D6 (CYP2D6), was synthesized to investigate the influence of N-substitution on the metabolism by cytochrome P450s, as well as on P450 selectivity. The analogs were obtained by introducing alkyl substituents at the amino group of MAMC and by replacing this moiety with a pyridine group. Competition experiments using heterologously expressed CYP2D6 demonstrated that the introduction and elongation of alkyl substituents strongly decreased the IC(50) values toward dextromethorphan O-demethylation. Metabolism studies showed that the regioselectivity of metabolism was unaffected by the varying N substituents, as only O-dealkylation of the analogs and no N-dealkylation was observed. In excellent agreement with the competition experiments, metabolism studies also showed that elongation of the alkyl chain dramatically increased the affinity of the compounds toward CYP2D6, as indicated by an up to 100-fold decrease in K(m) values. The V(max) values displayed a much less pronounced decrease with an increasing N-alkyl chain, resulting in as much as a 30-fold increase in the V(max)/K(m) value. Interestingly, due to the higher fluorescent yield of the N-alkyl metabolites compared with the metabolite of MAMC, O-dealkylation of N-methyl MAMC by CYP2D6 can be measured with a more than 3-fold higher sensitivity. Studies on P450 selectivity showed that only CYP1A2 and CYP2D6 contribute to the O-dealkylation of the N-alkyl analogs in both heterologously expressed P450s and human liver microsomes. In sharp contrast to CYP2D6, N-alkylation of MAMC did not significantly affect the K(m) values of O-dealkylation by CYP1A2, but it did result in higher V(max) values. Finally, CYP1A2 also N-dealkylated the analogs.


Subject(s)
Coumarins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Alkylation , Binding, Competitive/drug effects , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2D6/metabolism , Dealkylation , Humans , Hydroxylation , In Vitro Techniques , Isoenzymes/metabolism , Kinetics , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Models, Biological , Protein Conformation , Proteins/metabolism
18.
Biochem Pharmacol ; 60(10): 1509-18, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11020454

ABSTRACT

The study of drug metabolism in cultured rat hepatocytes is hampered by the rapid loss of the expression of cytochrome P450 enzymes. Nevertheless, the activity of cytochrome P450 3A (CYP3A), one of the most important isoenzymes for drug metabolism, can be elevated by chemical inducers. In the present study, we investigated in cultured rat hepatocytes the induction of all four currently identified CYP3A isoforms by dexamethasone, and compared the results obtained in vitro with the induction profile of dexamethasone in vivo. To this end, CYP3A mRNA levels were quantified with a novel, radioactive reverse transcriptase-polymerase chain reaction (RT-PCR) assay, and CYP3A enzymatic activity was measured by a testosterone hydroxylation assay. In the RT-PCR assay, CYP3A isoforms were co-amplified with glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in the presence of radioactively labeled nucleotides. This resulted in an extremely sensitive and accurate determination of CYP3A expression levels, relative to those of GAPDH. Using this RT-PCR assay, it was found that the expression of all CYP3A isoforms in rat hepatocytes, cultured on a collagen matrix, was decreased by 80-90% within one day of cultivation. After addition of dexamethasone, at one day after isolation, CYP3A1 mRNA levels were elevated to levels comparable to those in freshly isolated hepatocytes within two days. In contrast, CYP3A2, CYP3A9, and CYP3A18 mRNA levels were not affected by dexamethasone treatment, and were hardly detectable after three days of cultivation. CYP3A enzymatic activity was also induced in cultured hepatocytes (approximately 6-fold) after addition of dexamethasone. In vivo, CYP3A1 mRNA levels increased 45-fold after dexamethasone administration. However, in contrast to the situation in cultured hepatocytes, CYP3A2 and CYP3A18 were also induced, albeit to a lesser extent (4- and 7-fold elevated mRNA levels, respectively). We conclude that the selective induction of CYP3A1 in dexamethasone-treated rat hepatocytes allows the study of biotransformation reactions by CYP3A1, without interference by any of the other CYP3A isoenzymes.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/biosynthesis , Dexamethasone/pharmacology , Liver/drug effects , Mixed Function Oxygenases/biosynthesis , Animals , Anti-Inflammatory Agents/pharmacology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Enzyme Induction/drug effects , In Vitro Techniques , Liver/enzymology , Male , Mixed Function Oxygenases/genetics , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/metabolism
19.
J Pharmacol Exp Ther ; 294(2): 753-61, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10900257

ABSTRACT

Selenocysteine Se-conjugates have recently been proposed as potential prodrugs to target pharmacologically active selenol compounds to the kidney. Although rat renal cytosol displayed a high activity of beta-elimination activity toward these substrates, the enzymes involved in this activation pathway as yet have not been identified. In the present study, the possible involvement of cysteine conjugate beta-lyase/glutamine transaminase K (beta-lyase/GTK) in cytosolic activity was investigated. To this end, the enzyme kinetics of 15 differentially substituted selenocysteine Se-conjugates and 11 cysteine S-conjugates was determined using highly purified rat renal beta-lyase/GTK. The results demonstrate that most selenocysteine Se-conjugates are beta-eliminated at a very high activity by purified beta-lyase/GTK, implicating an important role of this protein in the previously reported beta-elimination reactions in rat renal cytosol. As indicated by the rapid consumption of alpha-keto-gamma-methiolbutyric acid, purified beta-lyase/GTK also catalyzed transamination reactions, which appeared to even exceed that of beta-elimination. The corresponding sulfur analogs also showed significant transamination but were beta-eliminated at an extremely low rate. Comparison of the obtained enzyme kinetic data of purified beta-lyase/GTK with previously obtained data from rat renal cytosol showed a poor correlation. By determining the activity profiles of cytosolic fractions applied to anion exchange fast protein liquid chromatography and gel filtration chromatography, the involvement of multiple enzymes in the beta-elimination of selenocysteine Se-conjugates in rat renal cytosol was demonstrated. The identity and characteristics of these alternative selenocysteine conjugate beta-lyases, however, remain to be established.


Subject(s)
Carbon-Sulfur Lyases/metabolism , Lyases/metabolism , Selenocysteine/analogs & derivatives , Selenocysteine/pharmacokinetics , Transaminases/metabolism , Animals , Anions , Biotransformation , Carbon-Sulfur Lyases/isolation & purification , Chromatography, Gel/methods , Chromatography, Ion Exchange/methods , Cytosol/enzymology , Kidney/enzymology , Kinetics , Lyases/isolation & purification , Male , Rats , Rats, Wistar , Substrate Specificity , Transaminases/isolation & purification
20.
J Pharmacol Exp Ther ; 294(2): 762-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10900258

ABSTRACT

This study was performed to evaluate whether selenocysteine Se-conjugates are substrates for human cysteine conjugate beta-lyase enzymes. By testing kidney cytosols of three different humans, we studied interindividual differences in beta-lyase enzymes in humans. A series of 22 selenocysteine Se-conjugates were tested in rat and human kidney cytosols to compare their ability to form selenol compounds by beta-elimination. All compounds appeared to be good substrates for rat and human cysteine conjugate beta-lyase enzymes. The beta-lyase activity toward the selenocysteine Se-conjugates was comparable with those of the known nephrotoxic cysteine S-conjugate S-(2-chloro-1,1,2-trifluoroethyl)-L-cysteine in rats and humans. In rat kidney cytosol, between 22- and 877-fold higher beta-elimination rates were observed compared with human kidney cytosol. Significant correlations (P <.0001) between three human kidney cytosols in beta-lyase activities were found within the tested series of 22 compounds. Specific beta-lyase activities and intrinsic clearances of beta-elimination reactions ranged up to 3-fold, indicating that there are quantitative rather than qualitative interindividual differences in beta-eliminating enzymes in humans. Furthermore, Se-alkyl selenocysteine conjugates showed a sterically dependent bioactivation to selenol compounds in humans but not in rats. The present study supports the hypothesis that selenocysteine Se-conjugates may be useful as prodrugs to target pharmacologically active selenol compounds (e.g., antitumor or chemoprotective) to the kidney in humans.


Subject(s)
Kidney/enzymology , Prodrugs/pharmacokinetics , Selenocysteine/analogs & derivatives , Selenocysteine/pharmacokinetics , Aged , Animals , Cytosol/enzymology , Humans , Kinetics , Lyases/metabolism , Male , Middle Aged , Rats , Rats, Wistar , Selenocysteine/chemical synthesis , Structure-Activity Relationship , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...