Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Biochem Pharmacol ; 77(1): 134-40, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18930712

ABSTRACT

Seleno-L-methionine (SeMet) can be oxidized to L-methionine selenoxide (MetSeO) by flavin-containing monooxygenase 3 (FMO3) and rat liver microsomes in the presence of NADPH. MetSeO can be reduced by GSH to yield SeMet and GSSG. In the present study, the potential reduction of MetSeO to SeMet by other cellular components and antioxidants was investigated. Besides GSH, other thiols (L-cysteine, or N-acetyl-L-cysteine) and antioxidants (ascorbic acid and methimazole) also reduced MetSeO to SeMet. This reduction is unique to MetSeO since methionine sulfoxide was not reduced to methionine under similar conditions. The MetSeO reduction by thiols was instaneous and much faster than the reduction by ascorbic acid or methimazole. However, only one molar equivalent of ascorbic acid or methimazole was needed to complete the reduction, as opposed to two molar equivalents of thiols. Whereas the disulfides produced by the reactions of MetSeO with thiols are chemically stable, methimazole disulfide readily decomposed at pH 7.4, 37 degrees C to yield methimazole, methimazole-sulfenic acid, methimazole sulfinic acid, methimazole S-sulfonate, 1-methylimidazole (MI) and sulfite anion. Collectively, the results demonstrate reduction of MetSeO to SeMet by multiple endogenous thiols, ascorbic acid, and methimazole. Thus, oxidation of SeMet to MetSeO may result in depletion of endogenous thiols and antioxidant molecules. Furthermore, the novel reduction of MetSeO by methimazole provides clear evidence that methimazole should not be used as an alternative FMO substrate when studying FMO-mediated oxidation of SeMet.


Subject(s)
Ascorbic Acid/chemistry , Methimazole/chemistry , Methionine/analogs & derivatives , Organoselenium Compounds/metabolism , Selenomethionine/metabolism , Sulfhydryl Compounds/chemistry , Ascorbic Acid/pharmacology , Methimazole/pharmacology , Methionine/analysis , Methionine/chemistry , Methionine/metabolism , Organoselenium Compounds/analysis , Organoselenium Compounds/chemistry , Oxidation-Reduction , Selenomethionine/chemistry , Sulfhydryl Compounds/pharmacology
2.
J Pharmacol Exp Ther ; 321(3): 1095-101, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17347324

ABSTRACT

Previously, we have provided evidence that cytochromes P450 (P450s) and flavin-containing monooxygenases (FMOs) are involved in the oxidation of S-(1,2,2-trichlorovinyl)-L-cysteine (TCVC) in rabbit liver microsomes to yield the reactive metabolite TCVC sulfoxide (TCVCS). Because TCVC is a known nephrotoxic metabolite of tetrachloroethylene, the nephrotoxic potential of TCVCS in rats and TCVCS formation in rat liver and kidney microsomes were investigated. At 5 mM TCVC, rat liver microsomes formed TCVCS at a rate nearly 5 times higher than the rate measured with rat kidney microsomes, whereas at 1 mM TCVC only the liver activity was detectable. TCVCS formation in liver and kidney microsomes was dependent upon the presence of NADPH and was inhibited by the addition of methimazole or 1-benzylimidazole, but not superoxide dismutase, catalase, KCN, or deferoxamine, consistent with the involvement of both FMOs and P450s. Rats given TCVCS at 230 micromol/kg i.p. exhibited acute tubular necrosis at 2 and 24 h after treatment, and they had elevated blood urea nitrogen levels at 24 h, whereas TCVC was a much less potent nephrotoxicant than TCVCS. Furthermore, pretreatment with aminooxyacetic acid enhanced TCVC toxicity. In addition, reduced nonprotein thiol concentrations in the kidney were decreased by nearly 50% 2 h after TCVCS treatment compared with saline-treated rats, whereas the equimolar dose of TCVC had no effect on kidney nonprotein thiol status. No significant lesions or changes in nonprotein thiol status were observed in liver with either TCVC or TCVCS. Collectively, the results suggest that TCVCS may play a role in TCVC-induced nephrotoxicity.


Subject(s)
Cysteine/analogs & derivatives , Microsomes, Liver/metabolism , Microsomes/metabolism , Sulfoxides/metabolism , Alanine Transaminase/blood , Aminooxyacetic Acid/pharmacology , Animals , Aspartate Aminotransferases/blood , Blood Glucose/analysis , Blood Urea Nitrogen , Cysteine/chemistry , Cysteine/metabolism , Cysteine/toxicity , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Glycosuria/urine , Imidazoles/pharmacology , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Methimazole/pharmacology , Microsomes/drug effects , Microsomes, Liver/drug effects , Microsomes, Liver/pathology , Rats , Rats, Sprague-Dawley , Sulfhydryl Compounds/analysis , Sulfoxides/chemistry , Sulfoxides/toxicity , gamma-Glutamyltransferase/urine
3.
Chem Biol Interact ; 166(1-3): 176-81, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-16735035

ABSTRACT

Hydroxymethylvinyl ketone (HMVK) is a reactive oxidation product of 3-butene-1,2-diol, a metabolite of 1,3-butadiene. The potential for HMVK (0.1 and 1mM) to form hemoglobin (Hb) adducts in erythrocytes from Sprague-Dawley rats was investigated at physiological conditions (pH 7.4, 37 degrees C) using electrospray ionization mass spectrometry (ESI/MS). With the 0.1mM HMVK globin samples, the results indicate HMVK adduction on the alpha2, beta2 and beta3 chains. With the 1.0mM HMVK globin samples, adducts were detected on the beta2 and beta3 chains. However, no correlation was observed between incubation time and the extent of adduct formation, and additional adducts were detected when globin samples were fractionated by HPLC before the ESI/MS analyses. For specific localizations of adducts on the globin chains, trypsin digested peptides from the 1mM HMVK globin samples were subjected to liquid chromatography/mass spectrometry analyses. The results, which are consistent with formation of HMVK adducts on several specific peptides within the alpha- and beta-chains, suggest selectivity in the interaction of HMVK with the different cysteine residues in Hb. Because adducts were also detected in peptides containing no cysteine residues and multiple HMVK moieties were detected on some of the cysteine-containing peptides, the results suggest other amino acids may be also reactive with HMVK. Adduct profiles and their relative intensities were consistent between the 1 and 2h samples providing evidence for the HMVK reactions being fast and selective. The finding that fewer peptides were adducted in the 0.1mM HMVK globin samples provides further evidence for selectivity of the HMVK reaction. Collectively, the results show HMVK readily and selectively forms adducts on Hb. Characterization of these adducts will facilitate development of useful biomarkers of exposure to HMVK and its precursor 1,3-butadiene.


Subject(s)
Butanones/toxicity , Erythrocytes/drug effects , Hemoglobins/analysis , Spectrometry, Mass, Electrospray Ionization , Amino Acid Sequence , Animals , Butanones/analysis , Butanones/chemistry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Erythrocytes/chemistry , Hemoglobins/chemistry , Male , Molecular Sequence Data , Peptides/chemistry , Rats , Rats, Sprague-Dawley
4.
Chem Res Toxicol ; 19(12): 1643-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17173378

ABSTRACT

The roles of flavin-containing monooxygenases (FMOs) in the oxidation of seleno-l-methionine (SeMet) to l-methionine selenoxide (MetSeO) were investigated using cDNA-expressed human FMOs, purified rat liver FMOs, and rat liver microsomes. MetSeO and the N-2,4-dinitrophenyl-derivatives of SeMet and MetSeO were synthesized and characterized by 1H-NMR and ESI/MS. These reference compounds were then used to develop a sensitive HPLC assay to monitor SeMet oxidation to MetSeO. The formation of MetSeO in rat liver microsomes was time-, protein concentration-, SeMet concentration-, and NADPH-dependent. The microsomal activity exhibited a SeMet Km value (mean +/- S.D.; n = 4) of 0.91 +/- 0.29 mM and a Vmax value of 44 +/- 8.0 nmol MetSeO/mg protein/min. The inclusion of 1-benzylimidazole, superoxide dismutase, or deferoxamine caused no inhibition of the rat liver microsomal activity. Because these results suggested the involvement of FMOs in the oxidation of SeMet in rat liver microsomes, the formation of MetSeO was also examined using cDNA-expressed human and purified rat FMOs. The results showed that both rat and human FMO1 and FMO3 but not FMO5 can catalyze the reaction. The SeMet kinetic constants were obtained with purified rat liver FMO3 (Km = 0.11 mM, Vmax = 280 nmol/mg protein/min) and rat liver FMO1 (Km = 7.8 mM, Vmax = 1200 nmol/mg protein/min). Because SeMet has anti-cancer, chemopreventive, and toxic properties, the kinetic results suggest that FMO3 is likely to play a role in the biological activities of SeMet at low exposure conditions.


Subject(s)
Methionine/analogs & derivatives , Microsomes, Liver/metabolism , Organoselenium Compounds/metabolism , Oxygenases/metabolism , Selenomethionine/metabolism , Animals , Chromatography, High Pressure Liquid , DNA, Complementary/genetics , Dinitrobenzenes/chemistry , Humans , In Vitro Techniques , Magnetic Resonance Spectroscopy , Metabolic Detoxication, Phase I , Methionine/chemistry , Methionine/metabolism , Microsomes, Liver/enzymology , Organoselenium Compounds/chemistry , Oxygenases/genetics , Rats , Selenomethionine/chemistry , Spectrometry, Mass, Electrospray Ionization
5.
Biochim Biophys Acta ; 1703(2): 183-9, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15680226

ABSTRACT

Flavin-containing monooxygenases (FMOs) are microsomal enzymes that catalyze the NADPH-dependent oxidation of a large number of sulfur-, selenium-, and nitrogen-containing compounds. Five active isoforms (FMO1-5) have been identified and shown to be differently expressed in various mammalian tissues. Previous work from this laboratory has shown l-methionine to be S-oxidized by rat, rabbit and human FMO1-4, with FMO3 exhibiting the highest stereoselectivity for the formation of the d-diastereomer of methionine sulfoxide. In this report, we describe new studies aimed at determining if N-acetyl-l-methionine and peptides containing l-methionine can be substrates for FMOs. Experiments were carried out using either rabbit liver microsomes or human cDNA-expressed FMOs. The results show that while N-acetyl-l-methionine and peptides with a modified methionine amino group may not function as substrates for FMOs, peptides containing a free N-terminal methionine may act as FMO substrates. With human cDNA-expressed FMO1, FMO3, and FMO5, both FMO1 and FMO3 exhibited activity with the active peptides whereas FMO5 was inactive. With FMO3, the activity measured with methionine was similar (1 mM) or higher (5 mM) than the activity measured with H-Met-Val-OH and H-Met-Phe-OH. With FMO1, H-Met-Phe-OH and methionine exhibited similar activities whereas activity with H-Met-Val-OH was much lower. Collectively, the results show that FMOs can oxidize peptides containing a free N-terminal methionine. Thus, the role of FMOs in the oxidation of methionine in larger peptides or proteins warrants further investigation.


Subject(s)
Methionine/analogs & derivatives , Methionine/metabolism , Oxygenases/metabolism , Peptides/chemistry , Sulfoxides/metabolism , Chromatography, High Pressure Liquid , Humans , Methionine/chemistry
6.
J Pharmacol Exp Ther ; 305(3): 1163-72, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12626654

ABSTRACT

S-(1,2-Dichlorovinyl)-L-cysteine (DCVC) is the penultimate nephrotoxic metabolite of the environmental contaminant trichloroethylene. Although metabolism of DCVC by the cysteine conjugate beta-lyase is the most studied bioactivation pathway, DCVC may also be metabolized by the flavin-containing monooxygenase (FMO) to yield DCVC sulfoxide (DCVCS). Renal cellular injury induced by DCVCS was investigated in primary cultures of human proximal tubular (hPT) cells by assessment of time- and concentration-dependent effects on cellular morphology, acute cellular necrosis, apoptosis, mitochondrial function, and cellular glutathione (GSH) status. Confluent hPT cells incubated with as little as 10 microM DCVCS for 24 h exhibited morphological changes, although at least 100 microM DCVCS was required to produce marked changes. Acute cellular necrosis did not occur until 48 h with at least 200 microM DCVCS, indicating that this is a high-dose, late response. The extent of necrosis was similar to that with DCVC. In contrast, apoptosis occurred as early as 1 h with as little as 10 microM DCVCS and the extent of apoptosis was much less than that with DCVC. Mitochondrial function was maintained with DCVCS concentrations up to 100 microM, consistent with hPT cells only being competent to undergo apoptosis at early time points and relatively low concentrations. Marked depletion (>50%) of cellular GSH content was only observed with 500 microM DCVCS. These results, combined with previous studies showing protection from DCVC-induced necrosis and apoptosis by the FMO inhibitor methimazole, suggest that formation of DCVCS plays a significant role in trichloroethylene-induced renal cellular injury in hPT cells.


Subject(s)
Apoptosis , Cysteine/analogs & derivatives , Cysteine/pharmacology , Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Sulfoxides/pharmacology , Cell Size/drug effects , Cell Survival/drug effects , Cysteine/metabolism , Dose-Response Relationship, Drug , Glutathione/metabolism , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/enzymology , L-Lactate Dehydrogenase/metabolism , Mitochondria/physiology , Necrosis
7.
J Pharmacol Exp Ther ; 304(1): 185-91, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12490590

ABSTRACT

The potential roles of human hepatic and renal flavin-containing monooxygenases (FMOs) in the metabolism of the cysteine S-conjugates S-allyl cysteine (SAC) and S-(1,2-dichlorovinyl)-L-cysteine (DCVC) were investigated. Incubations of human cDNA-expressed FMO1, FMO3, FMO4, and FMO5 with SAC resulted in detection of SAC sulfoxide, with FMO3 exhibiting approximately 3-, 4-, and 10-fold higher activity than FMO1, FMO4, and FMO5, respectively. DCVC sulfoxide formation was only detected with FMO3 and was 59-fold lower than SAC sulfoxide formation. Incubations of human liver microsomes with SAC or DCVC resulted in detection of the corresponding sulfoxides and provided evidence for the involvement of FMO3. Incubations of SAC or DCVC with human kidney microsomes, however, led only to the detection of SAC sulfoxide. Immunoblots with monospecific antibodies to FMO1, FMO3, and FMO5 in kidney microsomes from 26 humans showed that the average expression levels for FMO1, FMO3, and FMO5 were 5.8 +/- 2.3, 0.5 +/- 0.4, and 2.4 +/- 1.4 pmol/mg (means +/- S.D.), respectively. Interestingly, African-American kidney samples (n = 8) exhibited significantly higher FMO1 levels than Caucasian samples (n = 17), whereas no difference in expression level between males and females was observed with any of the examined FMO isoforms. Collectively, the results provide evidence for the expression of three FMO isoforms in the human kidney and show that the contribution of renal FMOs in cysteine S-conjugate metabolism is likely to vary depending upon the cysteine S-conjugate and the relative expression levels of the active FMOs.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/metabolism , Cysteine/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/enzymology , Kidney/enzymology , Monoamine Oxidase/metabolism , Adolescent , Adult , Aged , Blotting, Western , Chromatography, High Pressure Liquid , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Female , Humans , In Vitro Techniques , Isoenzymes/biosynthesis , Isoenzymes/metabolism , Kidney/drug effects , Male , Microsomes/drug effects , Microsomes/enzymology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Middle Aged , Monoamine Oxidase/biosynthesis , Racial Groups , Sex Characteristics , Spectrophotometry, Ultraviolet
8.
Drug Metab Dispos ; 30(10): 1137-42, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12228191

ABSTRACT

S-Allyl-L-cysteine (SAC), a component of garlic and a metabolite of allyl halides, is a known substrate for multiple flavin-containing monooxygenases (FMOs). In the current study, we characterize the in vivo SAC metabolism by investigating the presence of SAC, N-acetyl-S-allyl-L-cysteine (NASAC), and their corresponding sulfoxides in the urine of rats given SAC (200 or 400 mg/kg i.p.). In some experiments, rats were given aminooxyacetic acid (AOAA), an inhibitor of cysteine conjugate beta-lyase, or methimazole, an alternative FMO substrate, 30 min prior to treatment with 200 mg/kg SAC. Nearly 40 to 50% of the dose was recovered in the 24-h collection period. In all treatment groups, the majority of the metabolites were excreted within 8 h. The major metabolites detected were NASAC and NASAC sulfoxide (NASACS; nearly 30-40% and 5-10% of the dose, respectively). Only small amounts of the dose (approximately 1.5%) were recovered as SAC and SAC sulfoxide (SACS). Methimazole pretreatment significantly reduced amounts of both SACS and NASACS detected in the urine when compared with rats given SAC only, whereas AOAA pretreatment had no effect. In vitro assays using rat liver microsomes were also carried out to compare the sulfoxidation rates of SAC and NASAC. The results showed that SAC was much more readily oxidized than NASAC. Collectively, the results provide evidence for the involvement of FMOs in the in vivo metabolism of SAC and that SAC is a much better substrate for FMOs than its corresponding mercapturic acid.


Subject(s)
Cysteine/analogs & derivatives , Cysteine/urine , Oxygenases/physiology , Sulfoxides/urine , Animals , Cysteine/metabolism , Male , Microsomes, Liver/metabolism , Oxygenases/metabolism , Rats , Rats, Sprague-Dawley , Sulfoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...