Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
Clin Genet ; 76(1): 63-8, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19659761

ABSTRACT

Sudden death of an infant is a devastating event that needs an explanation. When an explanation cannot be found, the case is labeled as sudden infant death syndrome or unclassified sudden infant death. The influence of genetic factors has been recognized for sudden infant death, but copy number variations (CNVs) as potential risk factors have not been evaluated yet. Twenty-seven families were enrolled in this study. The tissue specimens from deceased children were obtained and array-based comparative genomic hybridization (array-CGH) experiments were performed on the genomic DNA isolated from these specimens using Agilent Technologies Custom 4 x 44K arrays. Quantitative polymerase chain reaction experiments were performed to confirm the overlapping duplication and deletion region in two different cases. A de novo CNV is detected in 3 of 27 cases (11%). In case 1, an approximately 3-Mb (chr 8: 143,211,215-qter) duplication on 8q24.3-qter and a 4.4-Mb deletion on the 22q13.3-qter (chr 22: 45,047,068-qter) were detected. Subtelomeric chromosome analysis of the father and the surviving sibling of case 1 showed a balanced reciprocal translocation, 46,XY,t(8;22)(q24.3;q13.3). A 240-kb (chr 6: 26,139,810-26,380,787) duplication and a 1.9-Mb deletion (chr 6: 26,085,971-27,966,150) at chromosome 6p22 were found in cases 2 and 3, respectively. Array-CGH and conventional cytogenetic studies did not reveal the observed CNVs in the parents and the siblings of cases 2 and 3. The detected CNVs in cases 2 and 3 encompassed several genes including the major histone cluster genes. Array-CGH analysis may be beneficial during the investigations after sudden infant death.


Subject(s)
Gene Dosage , Sudden Infant Death/genetics , Child , Child, Preschool , Comparative Genomic Hybridization , Databases, Genetic , Fatal Outcome , Genome, Human/genetics , Humans , Infant , Infant, Newborn , Software
2.
Curr Drug Metab ; 2(1): 17-36, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11465149

ABSTRACT

The most common drug-drug interactions may be understood in terms of alterations of metabolism, associated primarily with changes in the activity of cytochrome P450 (CYP) enzymes. Kinetic parameters such as Km, Vmax, Ki and Ka, which describe metabolism-based drug interactions, are usually determined by appropriate kinetic models and may be used to predict the pharmacokinetic consequences of exposure to one or multiple drugs. According to classic Michaelis-Menten (M-M) kinetics, one binding site models can be employed to simply interpret inhibition (pure competitive, non-competitive and uncompetitive) or activation of the enzyme. However, some cytochromes P450, in particular CYP3A4, exhibit unusual kinetic characteristics. In this instance, the changes in apparent kinetic constants in the presence of inhibitor or activator or second substrate do not obey the rules of M-M kinetics, and the resulting kinetics are not straightforward and hamper mechanistic interpretation of the interaction in question. These unusual kinetics include substrate activation (autoactivation), substrate inhibition, partial inhibition, activation, differential kinetics and others. To address this problem, several kinetic models can be proposed, based upon the assumption that multiple substrate binding sites exist at the active site of a particular P450, and the resulting kinetic constants are, therefore, solved to adequately describe the observed interaction between multiple drugs. The following is an overview of some cytochrome P450-mediated classic and atypical enzyme kinetics, and the associated kinetic models. Applications of these kinetic models can provide some new insights into the mechanism of P450-mediated drug-drug interactions.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Algorithms , Animals , Cytochrome P-450 Enzyme Inhibitors , Enzyme Activation/physiology , Humans , Kinetics
3.
Chem Res Toxicol ; 14(1): 62-70, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11170509

ABSTRACT

Therapy with the oral antidiabetic agent troglitazone (Rezulin) has been associated with cases of severe hepatotoxicity and drug-induced liver failure, which led to the recent withdrawal of the product from the U.S. market. While the mechanism of this toxicity remains unknown, it is possible that chemically reactive metabolites of the drug play a causative role. In an effort to address this possibility, this study was undertaken to determine whether troglitazone undergoes metabolism in human liver microsomal preparations to electrophilic intermediates. Following incubation of troglitazone with human liver microsomes and with cDNA-expressed cytochrome P450 isoforms in the presence of glutathione (GSH), a total of five GSH conjugates (M1-M5) were detected and identified tentatively by LC-MS/MS analysis. In two cases (M1 and M5), the structures of the adducts were confirmed by NMR spectroscopy and/or by comparison with an authentic standard prepared by synthesis. The formation of GSH conjugates M1-M5 revealed the operation of two distinct metabolic activation pathways for troglitazone, one of which involves oxidation of the substituted chromane ring system to a reactive o-quinone methide derivative, while the second involves a novel oxidative cleavage of the thiazolidinedione (TZD) ring, potentially generating highly electrophilic alpha-ketoisocyanate and sulfenic acid intermediates. When troglitazone was administered orally to a rat, samples of bile were found to contain GSH conjugates which reflected the operation of these same metabolic pathways in vivo. The finding that metabolism of the TZD ring of troglitazone was catalyzed selectively by P450 3A enzymes is significant in light of the recent report that troglitazone is an inducer of this isoform in human hepatocytes. The implications of these results are discussed in the context of the potential for troglitazone to covalently modify hepatic proteins and to cause oxidative stress through redox cycling processes, either of which may play a role in drug-induced liver injury.


Subject(s)
Chromans/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Thiazoles/pharmacokinetics , Thiazolidinediones , Animals , Bile/metabolism , Biotransformation , Catalysis , Chromans/metabolism , Chromans/toxicity , Chromatography, Liquid , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Glutathione/pharmacology , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/toxicity , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Ketoconazole/pharmacology , Kinetics , Mass Spectrometry , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , NADP/metabolism , Rats , Steroid Hydroxylases/antagonists & inhibitors , Steroid Hydroxylases/metabolism , Thiazoles/metabolism , Thiazoles/toxicity , Troglitazone
4.
Drug Metab Dispos ; 28(11): 1274-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11038152

ABSTRACT

Caspofungin acetate (MK-0991) is a semisynthetic pneumocandin derivative being developed as a parenteral antifungal agent with broad-spectrum activity against systemic infections such as those caused by Candida and Aspergillus species. Following a 1-h i.v. infusion of 70 mg of [(3)H]MK-0991 to healthy subjects, excretion of drug-related material was very slow, such that 41 and 35% of the dosed radioactivity was recovered in urine and feces, respectively, over 27 days. Plasma and urine samples collected around 24 h postdose contained predominantly unchanged MK-0991, together with trace amounts of a peptide hydrolysis product, M0, a linear peptide. However, at later sampling times, M0 proved to be the major circulating component, whereas corresponding urine specimens contained mainly the hydrolytic metabolites M1 and M2, together with M0 and unchanged MK-0991, whose cumulative urinary excretion over the first 16 days postdose represented 13, 71, 1, and 9%, respectively, of the urinary radioactivity. The major metabolite, M2, was highly polar and extremely unstable under acidic conditions when it was converted to a less polar product identified as N-acetyl-4(S)-hydroxy-4-(4-hydroxyphenyl)-L-threonine gamma-lactone. Derivatization of M2 in aqueous media led to its identification as the corresponding gamma-hydroxy acid, N-acetyl-4(S)-hydroxy-4-(4-hydroxyphenyl)-L-threonine. Metabolite M1, which was extremely polar, eluting from HPLC column just after the void volume, was identified by chemical derivatization as des-acetyl-M2. Thus, the major urinary and plasma metabolites of MK-0991 resulted from peptide hydrolysis and/or N-acetylation.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Antifungal Agents/pharmacokinetics , Peptides, Cyclic , Peptides , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Antifungal Agents/blood , Antifungal Agents/urine , Caspofungin , Chromatography, High Pressure Liquid , Echinocandins , Humans , Lipopeptides , Magnetic Resonance Spectroscopy , Mass Spectrometry
5.
J Clin Pharmacol ; 39(3): 260-7, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10073325

ABSTRACT

This study assessed whether the previously reported difference in tirilazad clearance between pre- and postmenopausal women is reversed by hormone replacement and whether this observation can be explained by differences in CYP3A4 activity. Ten healthy women from each group were enrolled: premenopausal (ages 18-35), postmenopausal (ages 50-70), postmenopausal receiving estrogen, and postmenopausal women receiving estrogen and progestin. Volunteers received 0.0145 mg/kg midazolam and 3.0 mg/kg tirilazad mesylate intravenously on separate days. Plasma tirilazad and midazolam were measured by HPLC/dual mass spectrophotometry (MS/MS) assays. Tirilazad clearance was significantly higher in premenopausal women (0.51 +/- 0.09 L/hr/kg) than in postmenopausal groups (0.34 +/- 0.07, 0.32 +/- 0.06, and 0.36 +/- 0.08 L/hr/kg, respectively) (p = 0.0001). Midazolam clearance (0.64 +/- 0.12 L/hr/kg) was significantly higher in premenopausal women compared to postmenopausal groups (0.47 +/- 0.11, 0.49 +/- 0.11, and 0.53 +/- 0.19 L/hr/kg, respectively) (p = 0.037). Tirilazad clearance was weakly correlated with midazolam clearance (r2 = 0.129, p = 0.02). Tirilazad clearance is faster in premenopausal women than in postmenopausal women, but the effect of menopause on clearance is not reversed by hormone replacement. Tirilazad clearance in these women is weakly related to midazolam clearance, a marker of CYP3A activity.


Subject(s)
Antioxidants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases , Estrogens/pharmacology , Hormone Replacement Therapy , Pregnatrienes/pharmacokinetics , Progesterone/pharmacology , Adolescent , Adult , Age Factors , Aged , Area Under Curve , Body Weight , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/physiology , Drug Therapy, Combination , Estrogens/therapeutic use , Female , Humans , Metabolic Clearance Rate , Midazolam/analogs & derivatives , Midazolam/blood , Midazolam/pharmacokinetics , Middle Aged , Oxidoreductases, N-Demethylating/physiology , Postmenopause/metabolism , Pregnatrienes/blood , Premenopause/metabolism , Progesterone/therapeutic use
6.
J Pharmacol Exp Ther ; 287(2): 583-90, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808684

ABSTRACT

Tirilazad mesylate (FREEDOX), a potent inhibitor of membrane lipid peroxidation in vitro, is under clinical development for the treatment of subarachnoid hemorrhage. In humans, tirilazad is cleared almost exclusively via hepatic elimination with a medium-to-high extraction ratio. In human liver microsomal preparations, tirilazad is biotransformed to multiple oxidative products and one reduced, pharmacologically active metabolite, U-89678. Characterization of the reduced metabolite by mass spectrometry and cochromatography with an authentic standard demonstrated that U-89678 was formed via stereoselective reduction of the Delta4 bond in the steroid A-ring. Kinetic analysis of tirilazad reduction in human liver microsomes revealed that kinetically distinct type 1 and type 2 5alpha-reductase enzymes were responsible for U-89678 formation; the apparent KM values for type 2 and type 1 were approximately 15 and approximately 0.5 microM, respectively. Based on pH dependence and finasteride inhibition studies, it was inferred that 5alpha-reductase type 1 was the high affinity/low capacity microsomal reductase that contributed to tirilazad clearance in vivo. In addition, a role for CYP3A4 in the metabolism of U-89678 was established using cDNA expressed CYP3A4 and correlation studies comparing U-89678 consumption with cytochrome P450 activities across a population of human liver microsomes. Collectively, these data suggest that formation of U-89678, a circulating pharmacologically active metabolite, contributes to the total metabolic elimination of tirilazad in humans and that clearance of U-89678 is mediated primarily via CYP3A4 metabolism. Therefore, concurrent administration of therapeutic agents that modulate 5alpha-reductase type 1 or CYP3A activity are anticipated to affect the pharmacokinetics of PNU-89678.


Subject(s)
Isoenzymes/metabolism , Microsomes, Liver/enzymology , Oxidoreductases/metabolism , Pregnatrienes/pharmacokinetics , Biotransformation , Cholestenone 5 alpha-Reductase , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Neuroprotective Agents/pharmacokinetics , Oxidoreductases/antagonists & inhibitors
7.
J Pharmacol Exp Ther ; 287(2): 591-7, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808685

ABSTRACT

The effect of oral finasteride, an inhibitor of 5alpha-reductase, on the clearance of tirilazad, a membrane lipid peroxidation inhibitor, was assessed in eight healthy men who received: 1) 10 mg/kg tirilazad mesylate solution orally on the 7th day of a 10-day regimen of 5 mg finasteride once daily, 2) 10 mg/kg tirilazad mesylate orally, 3) 2 mg/kg tirilazad mesylate i.v. on the 7th day of a 10-day regimen of 5 mg finasteride once daily and 4) 2 mg/kg tirilazad mesylate i.v., in a four-way cross-over design. Plasma concentrations of tirilazad and its active reduced metabolites (U-89678 and U-87999) were measured by liquid chromatography with tandem mass spectrometry (LC-MS-MS). Finasteride increased mean tirilazad areas under the curve by 21 and 29% for i.v. and p.o. tirilazad, respectively. Mean U-89678 areas under the curve were decreased 92 and 75% by finasteride administration with i.v. and p.o. tirilazad, respectively, and decreases of 94 and 85% in mean U-87999 area under the curve values were observed. These differences were statistically significant. These results indicate that finasteride inhibits the metabolism of tirilazad to U-89678. However, this inhibition has only a moderate effect on the overall clearance of tirilazad. These results thus confirm earlier in vitro work that showed that tirilazad is predominantly metabolized by CYP3A4. Although the major circulating metabolites of tirilazad are formed via reduction, this represents a minor route of tirilazad elimination in man.


Subject(s)
Enzyme Inhibitors/pharmacology , Finasteride/pharmacology , Oxidoreductases/antagonists & inhibitors , Pregnatrienes/pharmacokinetics , Adolescent , Adult , Area Under Curve , Biotransformation , Cholestenone 5 alpha-Reductase , Free Radical Scavengers/pharmacology , Half-Life , Humans , Male , Neuroprotective Agents/pharmacology , Reference Values
8.
Xenobiotica ; 27(11): 1131-45, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9413917

ABSTRACT

1. We have determined that 2,4-dipyrrolidinylpyrimidine (2,4-DPP), used as a model for studies of the metabolism of therapeutic agents containing this moiety, undergoes three characteristic hydroxylations when incubated with male rat liver microsomes. Analysis of microsomal incubates of stable isotope labelled analogues of 2,4-DPP by particle beam-liquid chromatography-mass spectrometry (LC-PB-MS) has shown that the three metabolites are 4-(3-hydroxypyrrolidinyl)-2-(pyrrolidinyl)-pyrimidine (M1), 4-(2-hydroxypyrrolidinyl)-2-(pyrrolidinyl)-pyrimidine (M2) and 2-(2-hydroxypyrrolidinyl)-4-(pyrrolidinyl)-pyrimidine (M3). 2. We determined that enzymes of the cytochrome P450 family are responsible for the in vitro hydroxylations of 2,4-DPP. 3. We observed that in microsomal incubations carried out in the presence of cyanide, a single cyanide adduct is formed implicating an iminium ion intermediate in the oxidation of the 2-pyrrolidine ring. 4. We also determined the intermolecular deuterium isotope effects for the formation of each of the three products. For M1, kH/kD = 14.55 +/- 0.54; for M2, kH/kD = 6.01 +/- 0.65; and for M3, kH/kD = 5.35 +/- 1.18. 5. We interpret these data as suggesting that M2 and M3 are formed by the same mechanism, probably including the formation of an iminium ion, and that M1 is formed by direct hydrogen abstraction.


Subject(s)
Antioxidants/metabolism , Cytochrome P-450 Enzyme System/metabolism , Free Radical Scavengers , Pregnatrienes/metabolism , Pyrimidines/metabolism , Pyrroles/metabolism , Animals , Biotransformation , Chromatography, Liquid , Cyanides/metabolism , Deuterium , Hydroxylation , Male , Mass Spectrometry , Oxidation-Reduction , Pregnatrienes/pharmacokinetics , Rats , Rats, Sprague-Dawley
9.
Drug Metab Dispos ; 24(7): 697-701, 1996 Jul.
Article in English | MEDLINE | ID: mdl-8818564

ABSTRACT

After administration of a mixed dose of both radioisotope and stable-isotope-labeled tirilazad, we carried out a parallel set of HPLC analyses for drug metabolites in bile samples from monkeys and dogs using either radioactivity monitoring (RAM) for 14C or the chemical reaction interface mass spectrometry technique (CRIMS) to detect 13C or 15N. CRIMS is a novel method where analytes are decomposed in a microwave-induced plasma and the elements contained in the analytes are reformulated into small gaseous species that are detected by a mass spectrometer. The comprehensiveness of detection, chromatographic resolution, sensitivity, signal/noise, and quantitative abilities of CRIMS were compared with RAM and in no case was RAM superior. This implies that stable isotopes may be substituted for radioisotopes in studies of drug metabolism where the ability of the latter approach to detect a label independent of the structures in which the label appears has been the primary reason for continuing to use a hazardous and expensive tracer. With HPLC-CRIMS, stable isotopes such as 13C and 15N can be comprehensively detected and quantitative patterns of drug metabolism from biological fluids can be produced that mirror the results when 14C is used.


Subject(s)
Bile/metabolism , Carbon Radioisotopes/chemistry , Free Radical Scavengers/analysis , Mass Spectrometry/methods , Pregnatrienes/analysis , Animals , Carbon Isotopes , Dogs , Macaca fascicularis , Male , Nitrogen Isotopes , Pregnatrienes/administration & dosage , Pregnatrienes/metabolism
10.
J Pharmacol Exp Ther ; 277(2): 982-90, 1996 May.
Article in English | MEDLINE | ID: mdl-8627581

ABSTRACT

Tirilazad mesylate (Freedox), a potent inhibitor of membrane lipid peroxidation in vitro, is under clinical development for the treatment of subarachnoid hemorrhage. In humans, tirilazad is cleared almost exclusively via hepatic elimination. Characterization of three major microsomal metabolites of tirilazad by mass spectrometry indicated that hydroxylation had occurred in the pyrrolidine ring(s) and at the 6 beta-position of the steroid domain. A role for CYP3A4 in the formation of the three major metabolites (tirilazad hydroxylase activity) was established in human liver microsomal preparations: 1) Tirilazad hydroxylation was potently inhibited by troleandomycin and ketoconazole, specific inhibitors of CYP3A4. 2) The rates of tirilazad hydroxylation within a population of 14 human livers displayed a 9-fold interindividual variation and a significant correlation (r2 = .95) between tirilazad hydroxylation and testosterone 6 beta-hydroxylation. 3) Kinetic analysis of tirilazad hydroxylase activity in three human livers resulted in an apparent Km of 2.12, 1.68 and 1.66 microM, and Vmax = 0.85, 0.44 and 3.45 (nmol/mg protein/min) for HL14, HL17 and HL21, respectively. In addition, an apparent Km of 2.07 microM was established for tirilazad hydroxylation in a cDNA-expressed CYP3A4 microsomal system. Collectively, these data indicate that the metabolic clearance of tirilazad in humans is catalyzed primarily by CYP3A4 and provide an insight into factors (i.e., age, sex, drug-drug interactions) that modulate the metabolic clearance of tirilazad in vivo.


Subject(s)
Antioxidants/pharmacokinetics , Cytochrome P-450 Enzyme System/physiology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/physiology , Pregnatrienes/pharmacokinetics , Biotransformation , Cytochrome P-450 CYP3A , Humans , Hydroxylation , Steroid Hydroxylases/physiology
11.
J Pharmacol Exp Ther ; 277(2): 991-8, 1996 May.
Article in English | MEDLINE | ID: mdl-8627582

ABSTRACT

The effect of ketoconazole, a CYP3A inhibitor, on the oral bioavailability of tirilazad mesylate was assessed in 12 healthy subjects, who received the following treatments in a crossover design: a) 10 mg/kg tirilazad mesylate solution orally on the fourth day of a 7-day regimen of 200 mg ketoconazole once daily, b) 10 mg/kg tirilazad mesylate solution orally, c) 2 mg/kg i.v. tirilazad mesylate solution on the fourth day of a 7-day regimen of 200 mg ketoconazole once daily and d) 2mg/kg i.v. tirilazad mesylate solution. Plasma concentrations of tirilazad mesylate and its active reduced metabolites (U-89678 and U-87999) were measured by high-performance liquid chromatography. Urinary ratios of 6 beta-hydroxycortisol to cortisol (6 beta-OHC/C) were measured as an index of hepatic CYP3A activity. Ketoconazole increased mean tirilazad mesylate area under the curve (AUC) values by 67% and 309% for i.v. and oral administration, respectively. Mean AUC values for U-89678 were increased 472% and 720% by ketoconazole coadministration with i.v. and oral tirilazad, respectively, whereas increases of > 10-fold in mean U-87999 AUC values were observed. These differences were statistically significant. These results indicate that ketoconazole inhibits the metabolism of these three compounds, which suggests that all of the compounds are substrates for CYP3A. Urinary 6 beta-OHC/C ratios did not reflect this level of effect of ketoconazole on CYP3A; this probe may not be useful for assessing the effect of CYP3A inhibitors. The absolute bioavailability of oral tirilazad was 8.7 +/- 4.8%; ketoconazole increased the bioavailability to 20.9 +/- 6.5%. Ketoconazole increased tirilazad mesylate bioavailability by decreasing the first-pass liver and gut wall metabolism of tirilazad mesylate to similar degrees.


Subject(s)
Antioxidants/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Pregnatrienes/pharmacokinetics , Adult , Biological Availability , Biotransformation , Cross-Over Studies , Female , Humans , Male , Metabolic Clearance Rate , Middle Aged
12.
Fundam Appl Toxicol ; 28(1): 111-7, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8566475

ABSTRACT

The metabolism of tris(2,3-dibromopropyl) phosphate (Tris-BP) was compared with that of completely deuterated Tris-BP (D15-Tris-BP) in an isolated, recirculating rat liver perfusion system in order to determine the relative quantitative importance of two different biotransformation pathways of Tris-BP: (i) cytochrome P450-mediated metabolism and (ii) GSH S-transferase-mediated metabolism. To accomplish this we quantitated the biliary excretion of S-(3-hydroxypropyl)glutathione (GSOH) as a marker metabolite for cytochrome P450-mediated metabolism and that of S-(2,3-dihydroxypropyl) glutathione (GSOHOH) as a marker metabolite for GSH S-transferase-mediated metabolism. Complete deuterium substitution of Tris-BP significantly decreased the formation of GSOH, whereas there was no effect on the formation of GSOHOH. Because our previous studies showed a large decrease in genotoxicity of D15-Tris-BP compared to Tris-BP, the present results support our hypothesis that cytochrome P450-mediated metabolism is responsible for the genotoxic effects of Tris-BP in the rat liver.


Subject(s)
Deuterium/adverse effects , Flame Retardants/metabolism , Liver/metabolism , Organophosphates/metabolism , Animals , Bile/metabolism , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme System/metabolism , Electrochemistry , Glutathione/metabolism , Glutathione Transferase/metabolism , In Vitro Techniques , Isotope Labeling , Liver/drug effects , Liver/enzymology , Male , Rats , Rats, Wistar
13.
Chem Res Toxicol ; 8(4): 560-73, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7548736

ABSTRACT

The haloalkane 1,2-dibromo-3-chloropropane (DBCP) is a carcinogen, mutagen, nephrotoxin, and testicular toxin. The identification of N-acetylcysteine conjugates of DBCP provides information on GSH mediated and cytochrome P450 mediated bioactivation pathways in the expression of DBCP-induced toxicities. N-Acetylcysteine conjugates excreted in the urine of male Sprague-Dawley rats administered DBCP, C1D2-DBCP, C2D1-DBCP, C3D2-DBCP, or D5-DBCP (80 mg/kg) were purified by reverse-phase HPLC as their methyl ester derivatives and characterized by fast atom bombardment tandem mass spectrometry. These metabolites were also converted to tert-butyldimethylsilyl ether derivatives and analyzed by gas chromatography-mass spectrometry (GC-MS) to facilitate the identification of N-acetyl-S-(2,3-dihydroxypropyl)cysteine (Ia), an apparent regioisomer of Ia, 2-(S-(N-acetylcysteinyl))-1,3-propanediol (Ib), N-acetyl-S-(3-hydroxypropyl)cysteine (IIa), and N-acetyl-S-(3-chloro-2-hydroxypropyl)-cysteine (III). Metabolites Ia, Ib, and III displayed quantitative retention of deuterium, an observation consistent with the formation of episulfonium ion intermediate(s) in their biogenesis. Mercapturate IIa retained three atoms of deuterium from D5-DBCP, and two atoms of deuterium from the dideuterio analogs (C1D2-DBCP and C3D2-DBCP), thus invoking P450 mediated formation of 2-bromoacrolein (2-BA) as an intermediate in the biogenesis of IIa. A mechanism is proposed in which conjugate addition of GSH to 2-BA, subsequent episulfonium ion formation, and addition of GSH afford 1,2-(diglutathion-S-yl)propanal. Glutathione mediated reduction is invoked to afford S-(3-hydroxypropyl)GSH which would be excreted in the urine as IIa. The quantitative retention of deuterium from C1D2-DBCP or C3D2-DBCP was indicative of isotopically sensitive branching of P450 metabolism at either C1 or C3 to afford 2-BA. C2D1-DBCP showed a 30% retention of 1 deuterium atom in IIa; the loss of the deuterium is consistent with 2-BA formation, whereas the retention of one deuterium atom is indicative of the formation of metabolite IIa through GSH conjugation of either 2,3-dibromopropanal or 2-bromo-3-chloropropanal. These data indicate that IIa is a marker metabolite for the potent direct-acting mutagen, 2-BA, or its metabolic precursors 2,3-dibromopropanal or 2-bromo-3-chloropropanal. Therefore, evidence has been presented for bioactivation of DBCP by glutathione and cytochrome P450 mediated mechanisms.


Subject(s)
Acetylcysteine/chemistry , Cytochrome P-450 Enzyme System/metabolism , Glutathione/metabolism , Insecticides/chemistry , Propane/analogs & derivatives , Acetylcysteine/metabolism , Animals , Biotransformation , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Insecticides/metabolism , Isotope Labeling , Magnetic Resonance Spectroscopy , Male , Propane/chemistry , Propane/metabolism , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Fast Atom Bombardment
14.
Drug Metab Dispos ; 23(3): 383-92, 1995 Mar.
Article in English | MEDLINE | ID: mdl-7628305

ABSTRACT

Tirilazad mesylate, a potent inhibitor of membrane lipid peroxidation in vitro, is under clinical development for the treatment of subarachnoid hemorrhage and head injury. In rat, tirilazad seems to be highly extracted and is cleared almost exclusively via hepatic elimination. The biotransformation of tirilazad has been investigated in liver microsomal preparations from adult male and female Sprague-Dawley rats. Tirilazad metabolism in male rat liver microsomes resulted in the formation of two primary metabolites: M1 and M2. In incubations with female rat liver microsomes, M2 was the only primary metabolite detected. Structural characterization of M1 and M2 by mass spectrometry demonstrated that M2 was formed by reduction of the delta 4-double bond in the steroid A-ring, whereas M1 arose from oxidative desaturation of one pyrrolidine ring. Further structural analysis of M2 by proton NMR demonstrated that reduction at C-5 had occurred by addition of hydrogen in the alpha-configuration. Using metabolic probes and antibodies specific to individual hepatic microsomal enzymes, CYP2C11 and 3-oxo-5 alpha-steroid:NADP+ delta 4-oxidoreductase (5 alpha-reductase) were identified as responsible for the formation of M1 and M2, respectively. The formation of M1 was inhibited by testosterone, nicotine, cimetidine, and anti-CYP2C11 IgG. The formation of M2 was inhibited by finasteride, a potent inhibitor of 5 alpha-reductase. Kinetic analysis of CYP2C11-mediated M1 formation in male rat liver microsomal incubations revealed that M1 formation occurred through a low-affinity/low-capacity process (KM = 16.67 microM, Vmax = 0.978 nmol/mg microsomal protein/min); the formation of M2 was mediated by 5 alpha-reductase in a high-affinity/low-capacity process (KM = 3.07 microM, Vmax = 1.06 nmol/mg microsomal protein/min). In contrast, the formation of M2 in female rat liver microsomes was mediated by 5 alpha-reductase in a high-affinity/high-capacity process (KM = 2.72 microM, Vmax = 4.11 nmol/mg microsomal protein/min). Comparison of calculated intrinsic formation clearances (Vmax/KM) for M1 and M2 indicated that the female rat possessed a greater in vitro metabolic capacity for tirilazad biotransformation than the male rat. Therefore, the clearance of tirilazad mesylate in the rat is mediated primarily by rat liver 5 alpha-reductase, and the capacity in the female rat is 5-fold the capacity in the male. These observations correlate with documented differences in 5 alpha-reductase activity and predict a gender difference in tirilazad hepatic clearance in vivo.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/metabolism , Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/metabolism , Lipid Peroxides/metabolism , Microsomes, Liver/metabolism , Pregnatrienes/metabolism , Steroid 16-alpha-Hydroxylase , Steroid Hydroxylases/metabolism , Animals , Biotransformation , Cytochrome P450 Family 2 , Female , Gas Chromatography-Mass Spectrometry , Lipid Peroxides/pharmacokinetics , Magnetic Resonance Spectroscopy , Male , Metabolic Clearance Rate , Microsomes, Liver/enzymology , Molecular Structure , Pregnatrienes/pharmacokinetics , Protons , Rats , Rats, Sprague-Dawley
15.
Cancer Res ; 54(3): 679-84, 1994 Feb 01.
Article in English | MEDLINE | ID: mdl-8306329

ABSTRACT

Calf thymus single-stranded DNA was modified with 2-bromoacrolein (2BA), a genotoxic metabolite of tris(2,3-dibromopropyl)phosphate. This DNA was used as a template for in vitro DNA replication by T7-polymerase and Klenow fragment of Escherichia coli DNA polymerase I. Increasing levels of 2BA modification led to decreased DNA synthesis as measured by [methyl-3H]dTTP incorporation. M13 mp19 single-stranded DNA template modified with 2BA was used to determine the sites of termination of DNA replication by T7 polymerase and Klenow fragment of Escherichia coli DNA polymerase I. It was found that DNA replication stopped frequently before and occasionally opposite deoxycytidine nucleotides. These results indicated that an as yet unidentified deoxycytidine:2BA adduct may have been formed in the reaction of 2BA with M13 DNA. To investigate if such adducts were formed, we reacted 2BA with deoxycytidine in vitro at pH 4.4, and putative deoxycytidine:2BA adducts were isolated by high-performance liquid chromatography. They were characterized by 1H and 13C nuclear magnetic resonance and with fast atom bombardment mass spectrometry as two diastereomeric 3-bromo-7-(beta-D-deoxyribofuranosyl)- 3,4-dihydro-2-hydroxy-(2H,7H)[1,6-a]pyrimidin-6-one adducts and a 3-bromo-7-(beta-deoxyribofuranosyl)-(4H,7H)-pyrimido[1,6-a]pyrimidin-6 -one adduct. Only the latter adduct, however, was formed in the reaction of 2BA with calf thymus single-stranded DNA in vitro. Tris(2,3-dibromopropyl)phosphate is clastogenic. Because clastogenicity may result from DNA adducts that block replication, the role of the presently identified deoxycytidine adducts of the reaction metabolite 2BA in the clastogenicity of tris(2,3-dibromopropyl)phosphate is discussed.


Subject(s)
Acrolein/analogs & derivatives , DNA Damage , DNA Replication/drug effects , DNA, Single-Stranded/drug effects , DNA/drug effects , Deoxycytidine/metabolism , Mutagens/toxicity , Acrolein/metabolism , Acrolein/toxicity , Animals , Base Sequence , Cattle , DNA/metabolism , DNA/physiology , DNA, Single-Stranded/metabolism , DNA, Single-Stranded/physiology , Magnetic Resonance Spectroscopy/methods , Molecular Sequence Data , Mutagens/metabolism , Spectrometry, Mass, Fast Atom Bombardment , Thymus Gland/chemistry
16.
Biochem Pharmacol ; 46(5): 877-84, 1993 Sep 01.
Article in English | MEDLINE | ID: mdl-8373438

ABSTRACT

In a previous report (Ryan and Petry, Arch Biochem Biophys 300: 699-704, 1993), the effects of two 21-aminosteroids (U-74500A and U-74006F) on the oxidation and reduction of iron in a buffer/organic solvent system were investigated. In those studies, U-74500A was found to be an efficient iron reductant and potential iron chelator, whereas U-74006F had little effect on iron redox chemistry. As an extension of those studies, we now report the effects of U-74006F and U-74500A on lipid peroxidation in systems that are dependent upon iron oxidation/reduction. In liposomes, U-74500A inhibited ADP:Fe(II)-dependent lipid peroxidation in a concentration-dependent manner, whereas U-74006F was minimally effective in this system. The mechanism of U-74500A-dependent inhibition probably involved interactions with iron, as iron oxidation was inhibited in the presence of this compound. No effects on iron oxidation were observed in the presence of U-74006F. Addition of Ferrozine to liposomal incubation mixtures indicated that at least two iron pools were present in samples containing U-74500A, one immediately bound by Ferrozine, and another that was bound more slowly. Furthermore, ADP:Fe(III)/ascorbate-dependent lipid peroxidation was blocked completely by U-74500A, presumably by formation of a redox inert complex upon reduction of the iron. U-74500A partially protected ADP:Fe(II) from oxidation by H2O2 and lipid hydroperoxides, indicating that the U-74500A:iron complex was stable in the presence of biologically relevant oxidants. U-74006F did not markedly affect iron oxidation or reduction when incorporated into phospholipid liposomes. In microsomal lipid peroxidation systems containing ADP:Fe(III) and NADPH, both U-74500A and U-74006F inhibited lipid peroxidation. U-74006F-dependent inhibition of microsomal lipid peroxidation was dependent on both NADPH and Fe(III). Further, it was enhanced when U-74006F was allowed to preincubate in this system prior to iron addition. Preincubation of U-74006F with microsomes, NADPH, and ADP:Fe(III) produced several metabolites detectable by HPLC. These results suggest that U-74500A inhibits lipid peroxidation by directly affecting iron redox chemistry, whereas U-74006F-mediated inhibition is enhanced by preincubation with a metabolically competent microsomal system.


Subject(s)
Lipid Peroxidation/drug effects , Pregnatrienes/pharmacology , Animals , Dose-Response Relationship, Drug , Down-Regulation , Ferric Compounds/pharmacology , Ferrous Compounds/pharmacology , Ferrozine , Liposomes , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , NADP/pharmacology , Oxidation-Reduction , Rats , Rats, Sprague-Dawley , Thiobarbituric Acid Reactive Substances/analysis , Time Factors
17.
Chem Res Toxicol ; 6(3): 335-40, 1993.
Article in English | MEDLINE | ID: mdl-8318655

ABSTRACT

Following administration to rats of a single ip dose (6.6 mg kg-1) of the investigational antitumor agent caracemide (N-acetyl-N,O-bis[methylcarbamoyl]hydroxylamine), the mercapturic acid derivative N-acetyl-S-(N-methylcarbamoyl)cysteine (AMCC) was identified in urine by thermospray LC-MS. Quantification of this conjugate was carried out by stable isotope dilution thermospray LC-MS, which indicated that the fraction of the caracemide dose recovered as AMCC in 24-h urine collections was 54.0 +/- 5.5% (n = 4). Since AMCC is known to represent a major urinary metabolite of methyl isocyanate (MIC) in the rat, the results of this study support the contention that caracemide yields MIC as a toxic intermediate in vivo. Furthermore, with the aid of a specifically deuterium-labeled analog of caracemide ([carbamoyloxy-C2H3]caracemide), it was shown that the methylcarbamoyl group of AMCC derived from both the O-methylcarbamoyl (72%) and N-methylcarbamoyl (28%) side chains of the drug. In view of these findings, it is concluded that caracemide acts as a latent form of MIC in vivo and that this reactive isocyanate (or labile S-linked conjugates thereof) may contribute to the antitumor properties and/or adverse side-effects of caracemide.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Cyanates/pharmacokinetics , Hydroxyurea/analogs & derivatives , Isocyanates , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacokinetics , Acetylcysteine/urine , Animals , Antineoplastic Agents/urine , Cyanates/toxicity , Cyanates/urine , Hydroxyurea/pharmacokinetics , Hydroxyurea/urine , Male , Rats , Rats, Sprague-Dawley
18.
Toxicol Appl Pharmacol ; 118(2): 186-95, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8441997

ABSTRACT

Analogs of tris(2,3-dibromopropyl)phosphate (Tris-BP) either labeled at specific positions with carbon-14, phosphorus-32, or oxygen-18 or dual-labeled with both deuterium and tritium were used as metabolic probes to study the chemical and metabolic events in the bioactivation of Tris-BP to chemically reactive metabolites in liver microsomal preparations. Oxidation at the terminal (C-3) carbon atom of the propyl groups of Tris-BP yielded the direct-acting mutagen 2-bromoacrolein as the major metabolite that binds to DNA. Although this reactive metabolite also appears to bind to microsomal protein, the rate of binding of radiolabeled Tris-BP to protein is 15-20x greater than binding to DNA, and some metabolites that retain the phosphate group are bound. Studies with deuterated analogs of Tris-BP implicate oxidation at C-2 of the propyl group as a major pathway that leads to protein binding which is enhanced by phenobarbital pretreatment of rats. Moreover, investigations with 18O-Tris-BP and H2(18)O show that Bis-BP that is formed from oxidation of Tris-BP incorporates one atom of oxygen from water. Deuterium isotope studies suggest that most of the Bis-BP arises from initial oxidation at C-2. Taken together these studies indicate that P-450 oxidation of Tris-BP at C-2 of the propyl group yields a reactive alpha-bromoketone metabolite of Tris-BP that can either alkylate proteins directly or be hydrolyzed to Bis-BP and an alpha-bromo-alpha'-hydroxyketone that can alkylate microsomal proteins.


Subject(s)
Flame Retardants/pharmacokinetics , Organophosphates/pharmacokinetics , Acrolein/analogs & derivatives , Acrolein/metabolism , Animals , Biotransformation , Cytochrome P-450 Enzyme System/physiology , DNA/metabolism , In Vitro Techniques , Male , Microsomes, Liver/metabolism , Organophosphates/metabolism , Oxidation-Reduction , Protein Binding , Rats , Rats, Sprague-Dawley
19.
Toxicol Appl Pharmacol ; 118(2): 196-204, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8441998

ABSTRACT

Analogs of tris(2,3-dibromopropyl)phosphate (Tris-BP) either labeled at specific positions with carbon-14 and phosphorus-32 or dual-labeled with both deuterium and tritium were administered to male Wistar rats at a nephrotoxic dose of 360 mumol/kg. The covalent binding of Tris-BP metabolites to hepatic, renal, and testicular proteins was determined after 9 and 24 hr, and plasma concentrations of bis(2,3-dibromopropyl)-phosphate (Bis-BP) formed metabolically from Tris-BP were measured at intervals throughout the initial 9-hr postdosing period. The covalent binding of 14C-Tris-BP metabolites in the kidney (2495 +/- 404 pmol/mg protein) was greater than that in the liver (476 +/- 123 pmol/mg protein) or testes (94 +/- 11 pmol/mg protein); the extent of renal covalent protein binding of Tris-BP metabolites was decreased by 82 and 84% when deuterium was substituted at carbon-2 and carbon-3, respectively. Substitution of Tris-BP with deuterium at carbon-2 or carbon-3 also decreased the mean area under the curve for Bis-BP plasma concentration by 48 and 57%, respectively. The mechanism of Tris-BP-induced renal and hepatic DNA damage was evaluated in Wistar rats by an automated alkaline elution procedure after the administration of analogs of Tris-BP or Bis-BP labeled at specific positions with deuterium. Renal DNA damage was decreased when Tris-BP was substituted with deuterium at either carbon-2 or carbon-3; the magnitude of the change correlated with both a decrease in the area under the Bis-BP plasma curve and a decrease in renal covalent binding of Tris-BP metabolites for each of the deuterated analogs. In marked contrast, analogs of Bis-BP labeled with deuterium at carbon-2 or carbon-3 did not show a decrease in the severity of renal DNA damage compared to unlabeled Bis-BP. On the basis of these observations a metabolic scheme for hepatic P-450-mediated oxidation at either carbon-2 or carbon-3 of Tris-BP affording Bis-BP by two alternate pathways that are susceptible to primary deuterium kinetic isotope effects is proposed. The Tris-BP metabolite, Bis-BP, is subsequently metabolized to reactive intermediates that cause DNA damage and bind to kidney proteins in a mechanism independent of cytochrome P-450.


Subject(s)
DNA Damage , DNA/drug effects , Flame Retardants/pharmacokinetics , Organophosphates/pharmacokinetics , Animals , Biotransformation , Cytochrome P-450 Enzyme System/physiology , Deuterium , In Vitro Techniques , Male , Organophosphates/metabolism , Organophosphates/toxicity , Oxidation-Reduction , Rats , Rats, Wistar
20.
Neurobiol Aging ; 13(2): 333-7, 1992.
Article in English | MEDLINE | ID: mdl-1522948

ABSTRACT

2-Amino-3-(methylamino)-propanoic acid (BMAA) is a low potency excitatory amino acid present in the cycad plant that has been proposed as a factor in the high incidence of amyotrophic lateral sclerosis-parkinsonism dementia (ALS-PD) in the western Pacific region. We employed stable isotopic forms of BMAA to assess the oral bioavailability of this compound in cynomolgous monkeys (n = 3). The stable isotope labeled BMAA ([15N]-BMAA) was injected i.v. at the same time that the unlabeled compound was administered orally. Both forms of BMAA were then quantified in a 48h urine sample by gas chromatography-mass spectrometry (GC/MS). Following oral dosing, 80% of the administered BMAA was absorbed into the systemic circulation; thus, oral bioavailability was high and other routes of administration could not result in significantly higher circulating levels of BMAA for a given administered dose.


Subject(s)
Amino Acids, Diamino/pharmacokinetics , Administration, Oral , Amino Acids, Diamino/administration & dosage , Amino Acids, Diamino/urine , Animals , Biological Availability , Cyanobacteria Toxins , Gas Chromatography-Mass Spectrometry , Injections, Intravenous , Macaca fascicularis
SELECTION OF CITATIONS
SEARCH DETAIL
...