Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 202
Filter
1.
EFORT Open Rev ; 8(1): 45-51, 2023 Jan 27.
Article in English | MEDLINE | ID: mdl-36705616

ABSTRACT

Purpose: Total elbow arthroplasty (TEA) is rarely performed compared to other arthroplasties. For many surgical procedures, literature shows better outcomes when they are performed by experienced surgeons and in so-called 'high-volume' hospitals. We systematically reviewed the literature on the relationship between surgical volume and outcomes following TEA. Methods: A literature search was performed using the MEDLINE, EMBASE and CINAHL databases. The literature was systematically reviewed for original studies comparing TEA outcomes among hospitals or surgeons with different annual or career volumes. For each study, data were collected on study design, indications for TEA, number of included patients, implant types, cut-off values for volume, number and types of complications, revision rate and functional outcome measures. The methodological quality of the included studies was assessed using the Newcastle-Ottawa Scale. Results: Two studies, which included a combined 2301 TEAs, found that higher surgeon volumes were associated with lower revision rates. The examined complication rates did not differ between high- and low-volume surgeons. In one study, low-hospital volume is associated with an increased risk of revision compared to high-volume hospitals, but for other complication types, no difference was found. Conclusions: Based on the results, the evidence suggests that high-volume centers have a lower revision rate in the long term. No minimum amount of procedures per year can be advised, as the included studies have different cut-off values between groups. As higher surgeon- and center-volume, (therefore presumably experience) appear to yield better outcomes, centralization of total elbow arthroplasty should be encouraged.

2.
Curr Med Chem ; 22(4): 500-27, 2015.
Article in English | MEDLINE | ID: mdl-25245506

ABSTRACT

The endocrine system is a major communication system in the body and is involved in maintenance of the reproductive system, fetal development, growth, maturation, energy production, and metabolism,. The endocrine system responds to the needs of an organism by secreting a wide variety of hormones that enable the body to maintain homeostasis, to respond to external stimuli, and to follow various developmental programs. This occurs through complex signalling cascades,with multiple sites at which the signals can be regulated. Endocrine disrupting compounds (EDCs) affect the endocrine system by simulating the action of the naturally produced hormones, by inhibiting the action of natural hormones, by changing the function and synthesis of hormone receptors, or by altering the synthesis, transport, metabolism, and elimination of hormones. It has been established that exposure to environmental EDCs is a risk factor for disruption of reproductive development and oncogenesis in both humans and wildlife. For accurate risk assessment of EDCs, the possibility of bioactivation through biotransformation processes needs to be included since neglecting these mechanisms may lead to undervaluation of adverse effects on human health caused by EDCs and/or their metabolites. This accurate risk assessment should include: (1) possibility of EDCs to be bioactivated into metabolites with enhanced endocrine disruption (ED) effects, and (2) possibility of EDCs to be biotransformed into reactive metabolites that may cause DNA damage. Here, we present an overview of different metabolic enzymes that are involved in the biotransformation of EDCs. In addition, we describe how biotransformation by Cytochromes P450 (CYPs), human estrogen sulfotransferase 1E1 (SULT1E1) and selected other phase II enzymes, can lead to the formation of bioactive metabolites. This review mainly focuses on CYP- and SULT-mediated bioactivation of estrogenic EDCs and summarizes our views on this topic while also showing the importance of including bioactivation and biotransformation processes for improved risk assessment strategies.


Subject(s)
Endocrine Disruptors/pharmacokinetics , Estrogens/pharmacokinetics , Animals , Biotransformation , Cytochrome P-450 Enzyme System/metabolism , Humans , Sulfotransferases/metabolism
3.
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
4.
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
5.
Pharmacogenomics J ; 10(6): 478-88, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20125119

ABSTRACT

Inter-individual variability in cytochrome P450 (CYP)-mediated xenobiotic metabolism is extensive. CYP1A2 is involved in the metabolism of drugs and in the bioactivation of carcinogens. The objective of this study was to functionally characterize eight polymorphic forms of human CYP1A2, namely T83M, S212C, S298R, G299S, I314V, I386F, C406Y and R456H. cDNAs of these variants were constructed and coexpressed in Escherichia coli with human NADPH cytochrome P450 oxidoreductase (CYPOR). All variants showed similar levels of apoprotein and holoprotein expression, except for I386F and R456H, which showed only apoprotein, and both were functionally inactive. The activity of CYP1A2 variants was investigated using 8 substrates, measuring 16 different activity parameters. The resulting heterogeneous activity data set was analyzed together with CYP1A2 wild-type (WT) form, applying multivariate analysis. This analysis indicated that variant G299S is substantially altered in catalytic properties in comparison with WT, whereas variant T83M is slightly but significantly different from the WT. Among CYP1A2 variants, out of the heterogeneous set of eight substrates, carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) was the most discriminative compound. In addition, R456 could be identified as an important residue for proper heme binding and stabilization.


Subject(s)
Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP1A2/metabolism , Cloning, Molecular , Escherichia coli/enzymology , Heme/metabolism , Humans , Mutagenesis, Site-Directed , NADPH-Ferrihemoprotein Reductase/metabolism , Nitrosamines/metabolism , Polymorphism, Genetic , Recombinant Proteins/metabolism
6.
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
7.
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
8.
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
9.
Drug Metab Dispos ; 34(9): 1640-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16790557

ABSTRACT

We have developed a fully automated bioreactor coupled to an on-line receptor affinity detection system. This analytical system provides detailed information on pharmacologically active metabolites of selective estrogen receptor modulators (SERMs) generated by cytochromes P450 (P450s). We demonstrated this novel concept by investigating the metabolic activation of tamoxifen and raloxifene by P450-containing pig and rat liver microsomes. The high resolution screening (HRS) system is based on the coupling of a P450-bioreactor to an HPLC-based estrogen receptor alpha (ERalpha) affinity assay. P450-derived metabolites of the SERMs were generated in the bioreactor, subsequently trapped on-line with solid phase extraction, and finally separated with gradient HPLC. Upon elution, the metabolites were screened on affinity for ERalpha with an on-line HRS assay. With this HRS system, we were able to follow, in a time-dependent manner, the formation of ERalpha-binding metabolites of tamoxifen and raloxifene. By analyzing the bioaffinity chromatograms with liquid chromatography-tandem mass spectrometry, structural information of the pharmacologically active metabolites was obtained as well. For tamoxifen, 15 active and 6 nonactive metabolites were observed, of which 5 were of primary, 10 of secondary, and 6 of an as yet unknown order of metabolism. Raloxifene was biotransformed in three primary and three secondary metabolites. MS/MS analysis revealed that three of the observed active metabolites of raloxifene were not described before. The present automated on-line HRS system coupled to a P450-containing bioreactor and an ERalpha-affinity detector proved very efficient, sensitive, and selective in metabolic profiling of SERMs.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Estrogen Receptor alpha/metabolism , Selective Estrogen Receptor Modulators/metabolism , Animals , Automation , Bioreactors , Biotransformation , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical/methods , In Vitro Techniques , Kinetics , Liver/enzymology , Mass Spectrometry , Microsomes, Liver , Molecular Structure , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/metabolism , Rats , Reproducibility of Results , Selective Estrogen Receptor Modulators/chemistry , Swine , Tamoxifen/chemistry , Tamoxifen/metabolism
10.
Toxicol In Vitro ; 20(6): 1005-13, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16488111

ABSTRACT

We recently developed two biomarker sets for oxidative damage: one for determination of lipid peroxidation (LPO) degradation products; acetaldehyde, propanal, butanal, pentanal, hexanal, heptanal, octanal, nonanal, malondialdehyde and acetone, by a gas chromatography-electron capture detection method, and the other for protein oxidation products such as o,o'-dityrosine, by an isotope dilution high performance liquid chromatography-tandem mass spectrometry method. In the present study, we explored the possibility to utilize these biomarkers for determining the oxidative damage in liver mammalian cells in vitro. Two different treatments were chosen for inducing oxidative stress in Chinese Hamster ovary cells: menadione and copper plus hydrogen peroxide (Cu2+/H2O2). Cells were incubated with the model compounds in the presence or absence of vitamin E and C, and cytotoxicity was evaluated by a nuclear-dye method. Results were compared to two fluorescent probes, H2DCF-DA and C11 -BODIPY581/591, which have been used for determining the formation of free radicals in the cells. From ten LPO degradation products, eight were increased significantly following incubation with menadione in cell lysate or incubation media. Menadione-induced oxidative stress was also confirmed by oxidation of fluorescent probes. However, no increased formation of protein oxidation products was observed. Vitamin E and C did not diminish the formation of LPO degradation products that were increased by menadione. Although Cu2+/H2O2 did not induce oxidation of fluorescent probes, it induced formation of six out of ten LPO degradation products. Vitamin E and C did not diminish the formation of LPO degradation products; vitamin C even substantially increased the formation of acetaldehyde and propanal, which is in line with its reported prooxidant action under certain conditions. Vitamin C also caused two-fold increase in Cu2+/H2O2-induced o,o'-dityrosine formation when applied simultaneously. In conclusion, our present results show that the LPO biomarker set can be used for evaluation of oxidant capacity and the toxic potential of various chemicals in an in vitro cell model. These biomarkers might even be more sensitive than measuring protein oxidation products or oxidation of fluorescent probes.


Subject(s)
Boron Compounds , Fluoresceins , Fluorescent Dyes , Lipid Peroxidation , Proteins/metabolism , Animals , Ascorbic Acid/pharmacology , Biomarkers , CHO Cells , Cell Survival , Cricetinae , Malondialdehyde/analysis , Oxidation-Reduction , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Vitamin E/pharmacology
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
Environ Toxicol Pharmacol ; 10(1-2): V, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11382557
18.
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
19.
Crit Rev Toxicol ; 31(1): 55-138, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11215692

ABSTRACT

An overview is presented on the molecular aspects of toxicity due to paracetamol (acetaminophen) and structural analogues. The emphasis is on four main topics, that is, bioactivation, detoxication, chemoprevention, and chemoprotection. In addition, some pharmacological and clinical aspects are discussed briefly. A general introduction is presented on the biokinetics, biotransformation, and structural modification of paracetamol. Phase II biotransformation in relation to marked species differences and interorgan transport of metabolites are described in detail, as are bioactivation by cytochrome P450 and peroxidases, two important phase I enzyme families. Hepatotoxicity is described in depth, as it is the most frequent clinical observation after paracetamol-intoxication. In this context, covalent protein binding and oxidative stress are two important initial (Stage I) events highlighted. In addition, the more recently reported nuclear effects are discussed as well as secondary events (Stage II) that spread over the whole liver and may be relevant targets for clinical treatment. The second most frequent clinical observation, renal toxicity, is described with respect to the involvement of prostaglandin synthase, N-deacetylase, cytochrome P450 and glutathione S-transferase. Lastly, mechanism-based developments of chemoprotective agents and progress in the development of structural analogues with an improved therapeutic index are outlined.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/etiology , Kidney/drug effects , Liver/drug effects , Acetaminophen/analogs & derivatives , Acetaminophen/pharmacokinetics , Analgesics, Non-Narcotic/pharmacokinetics , Animals , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Chemoprevention , Kidney/metabolism , Liver/metabolism , Mice , Rats , Species Specificity , Toxicity Tests
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...