Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Drug Metab Dispos ; 38(7): 1083-93, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20400660

RESUMEN

The study was initiated as an observation of incomplete extraction recovery of N-(4-(3-chloro-4-(2-pyridinylmethoxy)anilino)-3-cyano-7-ethoxy-6-quinolyl)-4-(dimethylamino)-2-butenamide (HKI-272) from human plasma. The objective of this study was to 1) identify the binding site(s) of HKI-272 to human plasma protein(s); 2) characterize the nature of the binding; and 3) evaluate the potential reversibility of the covalent binding. After incubation of [(14)C]HKI-272 with human plasma, the mixture was directly injected on liquid chromatography/mass spectrometry (LC/MS), and an intact molecular mass of HKI-272 human serum albumin (HSA) adduct was determined to be 66,999 Da, which is 556 Da (molecular mass of HKI-272) larger than the measured molecular mass of HSA (66,443 Da). For peptide mapping, the incubation mixture was separated with SDS-polyacrylamide gel electrophoresis followed by tryptic digestion combined with LC/tandem MS. A radioactive peptide fragment, LDELRDEGKASSAK [amino acid (AA) residue 182-195 of albumin], was confirmed to covalently bind to HKI-272. In addition, after HCl hydrolysis, a radioactive HKI-272-lysine adduct was identified by LC/MS. After combining the results of tryptic digestion and HCl hydrolysis, the AA residue of Lys190 of HSA was confirmed to covalently bind to HKI-272. A standard HKI-272-lysine was synthesized and characterized by NMR. The data showed that the adduct was formed via Michael addition with the epsilon-amine of lysine attacking to the beta-carbon of the amide moiety of HKI-272. Furthermore, reversibility of the covalent binding of HKI-272 to HSA was shown when a gradual release of HKI-272 was observed from protein pellet of HKI-272-treated human plasma after resuspension in phosphate buffer, pH 7.4, at 37 degrees C for 18 h.


Asunto(s)
Química Farmacéutica/métodos , Quinolinas/sangre , Albúmina Sérica/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Radioisótopos de Carbono/sangre , Humanos , Mapeo Peptídico/métodos , Péptidos/metabolismo , Ensayo de Unión Radioligante/métodos
2.
Chem Res Toxicol ; 23(1): 159-70, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19961160

RESUMEN

Previous studies suggested that lamotrigene (LTG) underwent bioactivation to a reactive aryl epoxide intermediate in rats. Nevertheless, definitive structures of these thioether conjugates, which are often needed to substantiate the mechanism of bioactivation and identity of reactive intermediate(s), were not fully established. In the present study, GSH, cysteinylglycine, and N-acetyl cysteine conjugates of LTG were isolated from bile of rats orally dosed with LTG (100 mg/kg), and their structures were fully elucidated by LC/MS and NMR. The definitive structural characterization of these metabolites provided evidence for the existence of a reactive aryl epoxide that was trapped as a GSH adduct. In vitro studies using various hepatic cellular and subcellular fractions obtained from human and rat were performed to demonstrate that LTG underwent bioactivation to form a GSH conjugate that was identical to the one initially characterized from in vivo studies. Human P450 2A6 and rat P450 2C11 appeared to be the primary enzymes activating LTG in human and rat liver microsomes, respectively. Interindividual variation in the bioactivation of LTG was demonstrated with 20 individual human liver microsomes. Furthermore, it was shown that human epidermal keratinocytes were capable of forming the same GSH conjugate, suggesting that LTG could be bioactivated in skin cells. The results from these studies suggest that LTG has the potential to undergo hepatic and nonhepatic bioactivation, leading to a reactive aryl epoxide intermediate in human. The bioactivation of LTG in epidermal cells provides a possible explanation for the idiosyncratic cutaneous reactions associated with LTG therapy.


Asunto(s)
Anticonvulsivantes/metabolismo , Hepatocitos/metabolismo , Queratinocitos/metabolismo , Microsomas Hepáticos/metabolismo , Triazinas/metabolismo , Administración Oral , Animales , Anticonvulsivantes/química , Anticonvulsivantes/farmacología , Hidrocarburo de Aril Hidroxilasas/metabolismo , Línea Celular , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP2A6 , Familia 2 del Citocromo P450 , Femenino , Glutatión/metabolismo , Humanos , Cinética , Lamotrigina , Espectroscopía de Resonancia Magnética , Masculino , Conformación Molecular , Ratas , Espectrometría de Masa por Ionización de Electrospray , Esteroide 16-alfa-Hidroxilasa/metabolismo , Sulfuros/química , Triazinas/química , Triazinas/farmacología
3.
Chem Res Toxicol ; 22(2): 299-310, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18980340

RESUMEN

Nuclear magnetic resonance (NMR) spectroscopy has traditionally been considered as an indispensable tool in elucidating structures of metabolites. With the advent of Fourier transform (FT) spectrometers, along with improvements in software and hardware (such as high-field magnets, cryoprobes, versatile pulse sequences, and solvent suppression techniques), NMR is increasingly being considered as a critical quantitative tool, despite its lower sensitivity as compared to mass spectrometry. A specific quantitative application of NMR is in determining the concentrations of biologically isolated metabolites, which could potentially be used as reference standards for further quantitative work by liquid chromatography/mass spectrometry. With the recent demands from regulatory agencies on quantitative information on metabolites, it is proposed that NMR will play a significant role in strategies aimed at addressing metabolite coverage in toxicological species. Traditionally, biologically isolated metabolites have not been considered as a way of generating "reference standards" for further quantitative work. However, because of the recent FDA guidance on safety testing of metabolites, one has to consider means of authenticating and quantitating biologically or nonbiologically generated metabolites. 1H NMR is being proposed as the method of choice, as it is able to be used as both a qualitative and a quantitative tool, hence allowing structure determination, purity check, and quantitative measurement of the isolated metabolite. In this publication, the application of NMR as a powerful and robust analytical technique in determining the concentrations of in vitro or in vivo isolated metabolites is discussed. Furthermore, to demonstrate the reliability and accuracy of metabolite concentrations determined by NMR, validation and cross-validation with gravimetric and mass spectrometric methods were conducted.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Preparaciones Farmacéuticas/análisis , Pruebas de Toxicidad/métodos , Acetaminofén/análisis , Acetaminofén/química , Acetaminofén/metabolismo , Animales , Cromatografía Liquida , Espectrometría de Masas , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Fenacetina/análisis , Fenacetina/química , Fenacetina/metabolismo , Ratas
4.
Chem Res Toxicol ; 22(2): 311-22, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19067650

RESUMEN

The recent guidance on "Safety Testing of Drug Metabolites" issued by the U.S. Food and Drug Administration, Center for Drug Evaluation and Research (CDER) has highlighted the importance of identifying and characterizing drug metabolites as early as possible in drug discovery and development. Furthermore, upon identifying significant circulating metabolites in human plasma, it has become important to demonstrate that these metabolites are present at an equal or greater exposure level (area under the curve, AUC) in any one of the preclinical species used in safety testing. Frequently, synthetic standards of metabolites are not available, and hence, obtaining their AUC values can be a challenge. In this report, we demonstrate how combinations of nuclear magnetic resonance (NMR) spectroscopy, liquid chromatography/ultraviolet/mass spectrometry (LC/UV/MS), and plasma pooling methods were used to obtain reliable AUC values of metabolites present in the plasma of preclinical species from short-term safety studies. Plasma pooling methods were compared to the traditional approaches of obtaining quantitative information on the levels of circulating metabolites in preclinical species. The exposure values obtained via sample pooling were comparable to those obtained by traditional methods of analyzing samples individually. In the absence of synthetic chemical standards, calculations of AUC values of metabolites, using either sample pooling or traditional approaches, were achieved through the use of UV detectors. In cases where the UV properties of metabolites were significantly different from their parent compounds, NMR was used as a quantitative tool to obtain exposure values. NMR was found to be useful in quantitating biologically produced metabolites, which could subsequently be used as reference compounds for further quantitative studies. The limitations of UV detectors to obtain exposure estimates are discussed. A practical solution is presented that will enable us to obtain a quantitative assessment of metabolite exposure in humans and coverage in toxicology species, hence, circumventing the use of radiolabeled compounds or authentic chemically synthesized standards of metabolites.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Preparaciones Farmacéuticas/sangre , Pruebas de Toxicidad/métodos , Algoritmos , Animales , Área Bajo la Curva , Cromatografía Líquida de Alta Presión , Perros , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Radioisótopos/química , Ratas , Estándares de Referencia , Espectrofotometría Ultravioleta , Espectrometría de Masas en Tándem , Pruebas de Toxicidad/normas
5.
Chem Biol Drug Des ; 70(4): 354-9, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17937780

RESUMEN

Major metabolites of dimethylaminoantipyrine have been synthesized using iron ortho-nitrophenylporphyrin chloride as biomimetic catalyst. Reactivity of iron tetrakis-ortho-nitrophenylporphyrin chloride [Fe(TNO2PP)Cl] has been compared with iron tetrakis-pentafluorophenylporphyrin chloride and iron tetrakis-2,6-dichlorophenylporphyrin chloride using various oxidants such as hydrogen peroxide, iodosobenzene, and cumene hydroperoxide in either protic or aprotic solvent. Effect of imidazole has been showed on the reactivity of Fe(TNO2PP)Cl/cumene hydroperoxide system.


Asunto(s)
Aminopirina/química , Cloruros/química , Hierro/química , Metaloporfirinas/química , Aminopirina/metabolismo , Peróxido de Hidrógeno/química , Estructura Molecular , Oxidantes/química , Oxidación-Reducción
6.
Chem Res Toxicol ; 20(12): 1954-65, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17935300

RESUMEN

The current study examined the bioactivation potential of a nonpeptidyl thrombopoietin receptor agonist, 1-(3-chloro-5-((4-(4-fluoro-3-(trifluoromethyl)phenyl)thiazol-2-yl)carbamoyl)pyridine-2-yl)piperidine-4-carboxylic acid (1), containing a 2-carboxamido-4-arylthiazole moiety in the core structure. Toxicological risks arising from P450-catalyzed C4-C5 thiazole ring opening in 1 via the epoxidation-->diol sequence were alleviated, since mass spectrometric analysis of human liver microsome and/or hepatocyte incubations of 1 did not reveal the formation of reactive acylthiourea and/or glyoxal metabolites, which are prototypic products derived from thiazole ring scission. However, 4-(4-fluoro-3-(trifluoromethyl)phenyl)thiazol-2-amine (2), the product of hydrolysis of 1 in human liver microsomes, hepatocytes, and plasma, underwent oxidative bioactivation in human liver microsomes, since trapping studies with glutathione led to the formation of two conjugates derived from the addition of the thiol nucleophile to 2 and a thiazole- S-oxide metabolite of 2. Mass spectral fragmentation and NMR analysis indicated that the site of attachment of the glutathionyl moiety in both conjugates was the C5 position in the thiazole ring. Based on the structures of the glutathione conjugates, two bioactivation pathways are proposed, one involving beta-elimination of an initially formed hydroxylamine metabolite and the other involving direct two-electron oxidation of the electron-rich 2-aminothiazole system to electrophilic intermediates. This mechanistic insight into the bioactivation process allowed the development of a rational chemical intervention strategy that involved blocking the C5 position with a fluorine atom or replacing the thiazole ring with a 1,2,4-thiadiazole group. These structural changes not only abrogated the bioactivation liability associated with 1 but also resulted in compounds that retained the attractive pharmacological and pharmacokinetic attributes of the prototype agent.


Asunto(s)
Piridinas/farmacología , Receptores de Trombopoyetina/agonistas , Tiazoles/química , Animales , Disponibilidad Biológica , Biotransformación , Línea Celular , Estabilidad de Medicamentos , Glutatión/metabolismo , Hepatocitos/metabolismo , Humanos , Masculino , Ratones , Microsomas Hepáticos/metabolismo , Estructura Molecular , Piridinas/sangre , Piridinas/química , Piridinas/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores de Trombopoyetina/genética , Tiazoles/sangre , Tiazoles/metabolismo , Tiazoles/farmacología , Transfección
7.
Mol Pharm ; 3(1): 55-61, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16686369

RESUMEN

Sulfasalazine is used in the treatment of ulcerative colitis, Crohn's disease, and rheumatoid arthritis. When administered orally, sulfasalazine is poorly absorbed with an estimated bioavailability of 3-12%. Recent studies using the T-cell line (CEM) have shown that sulfasalazine is a substrate for the ATP-binding cassette (ABC) efflux pump ABCG2. ABCG2 is known to efflux a number of xenobiotics and appears to be a key determinant of efficacy and toxicity of ABCG2 substrates. To date, there has not been any systematic study on the mechanisms involved in the transport of sulfasalazine in vivo. Accordingly, we investigated whether Bcrp (abcg2) is involved in the disposition of sulfasalazine. After oral administration of 20 mg/kg sulfasalazine, the area under the plasma concentration (AUC) time profile in Bcrp1 (abcg2)-/- knockout (KO) mice was approximately 111-fold higher than that in FVB wild-type (WT) mice. After intravenous administration of 5 mg/kg sulfasalazine, the AUC in Bcrp1 (abcg2)-/- KO mice was approximately 13-fold higher than that in WT mice. Moreover, treatment of WT mice with a single oral dose of gefitinib (Iressa; 50 mg/kg), a known inhibitor of Bcrp, given 2 h prior to administering a single oral dose of sulfasalazine (20 mg/kg), resulted in a 13-fold increase in the AUC of sulfasalazine compared to the AUC in vehicle-treated mice. Since gefitinib is also an inhibitor of P-glycoprotein (P-gp), the impact of P-gp on sulfasalazine absorption in vivo was also examined. The sulfasalazine AUC in mdr1a-/- KO versus WT mice did not differ significantly after either an oral (20 mg/kg) or an intravenous dose (5 mg/kg). We conclude that Bcrp (abcg2) is an important determinant for the oral bioavailability and the elimination of sulfasalazine in the mouse, and that sulfasalazine has the potential to be utilized as a specific in vivo probe of Bcrp (abcg2).


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Absorción/efectos de los fármacos , Quinazolinas/farmacología , Sulfasalazina/metabolismo , Sulfasalazina/farmacocinética , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/efectos de los fármacos , Transportadoras de Casetes de Unión a ATP/genética , Administración Oral , Animales , Área Bajo la Curva , Disponibilidad Biológica , Gefitinib , Inyecciones Intravenosas , Masculino , Ratones , Ratones Noqueados , Sulfasalazina/administración & dosificación , Factores de Tiempo
8.
Chem Res Toxicol ; 17(7): 879-88, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15257612

RESUMEN

Raloxifene was approved in 1997 by the FDA for the treatment of osteoporosis in postmenopausal women, and it is currently in clinical trials for the chemoprevention of breast cancer. Before widespread use as a chemopreventive agent in healthy women, the potential cytotoxic mechanisms of raloxifene should be investigated. In the current study, raloxifene was incubated with GSH and either rat or human liver microsomes, and the metabolites and GSH conjugates were characterized using liquid chromatography-tandem mass spectrometry. Raloxifene was converted to raloxifene diquinone methide GSH conjugates, raloxifene o-quinone GSH conjugates, and raloxifene catechols. For comparison, three raloxifene catechols were synthesized and characterized. In particular, 7-hydroxyraloxifene was found to oxidize to the 6,7-o-quinone. As compared with raloxifene diquinone methide, which has a half-life of less than 1 s in phosphate buffer, the half-life of raloxifene 6,7-o-quinone was much longer at t(1/2) = 69 +/- 2.5 min. The stability offered by raloxifene 6,7-o-quinone implies that it may be more toxic than raloxifene diquinone methide. Cytotoxicity studies in the human breast cancer cell lines S30 and MDA-MB-231 showed that 7-hydroxyraloxifene was more toxic than raloxifene in both cell lines. These results suggest that raloxifene could be metabolized to electrophilic and redox active quinoids, which have the potential to cause toxicity in vivo.


Asunto(s)
Benzoquinonas/metabolismo , Indolquinonas/metabolismo , Clorhidrato de Raloxifeno/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/metabolismo , Animales , Benzoquinonas/toxicidad , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Glutatión/análogos & derivados , Glutatión/metabolismo , Humanos , Indolquinonas/análisis , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Oxidación-Reducción , Clorhidrato de Raloxifeno/análogos & derivados , Clorhidrato de Raloxifeno/toxicidad , Ratas , Ratas Sprague-Dawley , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad
9.
Mutat Res ; 550(1-2): 109-21, 2004 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-15135645

RESUMEN

Long-term exposure to synthetic and endogenous estrogens has been associated with the development of cancer in several tissues. One potential mechanism of estrogen carcinogenesis involves catechol formation and these catechols are further oxidized to electrophilic/redox active o-quinones, which have the potential to both initiate and promote the carcinogenic process. Previously we showed that 4-hydroxyequilenin (4-OHEN) autoxidized to an o-quinone and caused a variety of damage to DNA. Since these deleterious effects could contribute to gene mutations, we investigated the Chinese hamster V79 cells to ascertain the relative ability of estradiol, 4-hydroxyestradiol, 17beta-hydroxyequilenin, 4,17beta-hydroxyequilenin, estrone, 4-hydroxyestrone, equilenin, and 4-hydroxyequilenin to induce the mutation of the hypoxanthine-guanine phosphoribosyltransferase (hprt) gene. All the 4-hydroxylated catechols induced significantly more colony formations in V79 cells as compared to the parent phenols at 100nM, suggesting that the catechol estrogen metabolites are more mutagenic towards the hprt gene than estrogens. Since 4-OHEN induced the highest mutation frequency, we examined a biomarker for transformation potential of this compound in MCF-10A cells using an anchorage-independent growth assay. Although 4-OHEN induced anchorage-independent growth of these cells, the isolated clones were not able to grow as tumors in vivo when injected into nude mice. These cells were assayed for genetic changes using cDNA microarrays. Real time RT-PCR confirmation of some of the differentially expressed genes showed down-regulation of metallothionein 2A, p53, BRCA1, and c-myc. Moreover, we showed the involvement of other genes important in cell transformation and oxidative stress, strengthening the hypothesis that this mechanism plays a considerable role in 4-OHEN-induced anchorage-independent growth.


Asunto(s)
Equilenina/análogos & derivados , Equilenina/farmacología , Congéneres del Estradiol/farmacología , Estrógenos/metabolismo , Regulación Neoplásica de la Expresión Génica , Animales , Adhesión Celular , División Celular , Línea Celular , Línea Celular Tumoral , Cricetinae , Análisis Mutacional de ADN , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Caballos , Masculino , Metalotioneína/metabolismo , Modelos Químicos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidación-Reducción , Estrés Oxidativo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
10.
Chem Res Toxicol ; 17(4): 512-20, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15089093

RESUMEN

Catechol-O-methyltransferase (COMT) plays an important role in the inactivation of biologically active and toxic catechols. It has been shown that COMT is genetically polymorphic with a wild-type and variant form where a valine has been substituted with a methionine. Several, but not all, epidemiological studies have shown that women, homozygous with the variant form, have an increased risk of developing breast cancer. Previously, we showed that 4-hydroxyequilenin (4-OHEN), a cytotoxic/genotoxic equine catechol estrogen metabolite, is both a substrate of COMT and an irreversible inhibitor of the methylation activity of COMT in vitro. To further understand the mechanism(s) of the association between the breast cancer risk and the COMT polymorphism, it was of interest to study the effect of the Val/Met polymorphism on COMT-catalyzed catechol estrogen methylation and 4-OHEN-mediated inhibition. In the present study, Michaelis-Menten analysis showed no difference between the relative ability of each form to methylate 4-OHEN. However, we found that the COMT variant form was more susceptible to 4-OHEN-mediated irreversible inactivation. Electrospray ionization mass spectrometry and SDS-gel analysis of COMT modified by 4-OHEN revealed that inhibition mechanisms include alkylation and/or oxidation of certain amino acids. In addition, site-directed mutagenesis experiments showed that Cys33 played a more important role in the variant form of COMT demonstrated by the fact that the C33A mutant of the variant form of COMT decreased its catalytic capability more dramatically as compared with that of wild type. Furthermore, thermotropic studies indicated that the variant form was more thermolabile, which suggested that the valine to methionine substitution may have changed the secondary/tertiary structure of the variant form of COMT, making it more susceptible to 4-OHEN and heat inactivation. These data suggest that 4-OHEN-mediated inhibition of the variant form of COMT in vivo might affect the detoxification efficiency of endogenous and/or exogenous catechol estrogens and play a role in the association between breast cancer risk and COMT polymorphism.


Asunto(s)
Neoplasias de la Mama/etiología , Neoplasias de la Mama/genética , Catecol O-Metiltransferasa/genética , Catecol O-Metiltransferasa/farmacología , Equilenina/análogos & derivados , Equilenina/farmacología , Equilenina/toxicidad , Inhibidores de Catecol O-Metiltransferasa , Congéneres del Estradiol , Femenino , Humanos , Metilación , Factores de Riesgo , Temperatura
12.
Chem Res Toxicol ; 16(10): 1264-76, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14565768

RESUMEN

The regulation of estrogenic and antiestrogenic effects by selective estrogen receptor modulators (SERMs) provides the basis for use in long-term therapy in cancer chemoprevention and postmenopausal osteoporosis. However, the evidence for carcinogenic properties within this class requires study of potential pathways of toxicity. There is strong evidence for the elevation of cellular levels of NO in tissue treated with SERMs, including the benzothiophene derivative, raloxifene, in part via up-regulation of nitric oxide synthases. Therefore, the reactions of 17beta-estradiol (E(2)), raloxifene, and an isomer with NO, peroxynitrite, and reactive nitrogen/oxygen species (RNOS) generated from NO(2)(-)/H(2)O(2) systems were examined. Peroxynitrite from bolus injection or slow release from higher concentrations of 3-morpholinosydnonimine (SIN-1) reacted with the benzothiophenes and E(2) to give aromatic ring nitration, whereas peroxynitrite, produced from the slow decomposition of lower concentrations of SIN-1, was relatively unreactive toward E(2) and yielded oxidation and nitrosation products with raloxifene and its isomer. The oxidation and nitrosation products formed were characterized as a dimer and quinone oxime derivative. Interestingly, the reaction of the benzothiophenes with NO in aerobic solution efficiently generated the same oxidation products. Stable quinone oximes are not unprecedented but have not been previously reported as products of RNOS-mediated metabolism. The reaction of glutathione (GSH) with the quinone oxime gave both GSH adducts from Michael addition and reduction to the corresponding o-aminophenol. The ready autoxidation of raloxifene, observed in the presence of NO, is the first such observation on the reactivity of SERMs and is potentially a general phenomenon of significance to SERM chemical toxicology.


Asunto(s)
Óxido Nítrico/química , Ácido Peroxinitroso/química , Clorhidrato de Raloxifeno/química , Especies de Nitrógeno Reactivo/química , Especies Reactivas de Oxígeno/química , Moduladores Selectivos de los Receptores de Estrógeno/química , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Cromatografía Líquida de Alta Presión , Cromatografía Liquida , Dimerización , Estradiol/química , Glutatión/química , Peróxido de Hidrógeno/química , Espectrometría de Masas , Estructura Molecular , Dióxido de Nitrógeno/química , Nitrosación , Oxidación-Reducción , Clorhidrato de Raloxifeno/toxicidad
13.
Chem Res Toxicol ; 16(6): 733-40, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12807356

RESUMEN

Dietary supplements containing Piper methysticum Forst. (kava) have been implicated in multiple cases of liver injury in humans, including 10 recently reviewed cases in which patients required liver transplantation following the usage of kava-containing products (Centers for Disease Control and Prevention, reprinted. (2003) J. Am. Med. Assoc. 289, 36-37). To investigate a possible mechanism(s) of kava-induced hepatotoxicity, an extract of kava was incubated in vitro with hepatic microsomes, NADPH, and GSH. Electrophilic intermediates that were generated via metabolic activation were trapped as GSH conjugates and removed from the protein mixture using ultrafiltration. Positive ion electrospray LC-MS/MS with precursor ion scanning was used for the selective detection of GSH conjugates, and LC-MS(n) product ion scanning was used to elucidate their structures. Using this in vitro MS-based screening assay, two novel electrophilic metabolites of kava, 11,12-dihydroxy-7,8-dihydrokavain-o-quinone and 11,12-dihydroxykavain-o-quinone, were identified. Mercapturic acids of these quinoid species were not detected in the urine of a human volunteer following ingestion of a dietary supplement that contained kava; instead, the corresponding catechols were metabolized extensively to glucuronic acid and sulfate conjugates. These observations indicate that quinoid metabolites, under most circumstances, are probably not formed in substantial quantities following the ingestion of moderate doses of kava. However, the formation of electrophilic quinoid metabolites by hepatic microsomes in vitro suggests that such metabolites might contribute to hepatotoxicity in humans when metabolic pathways are altered (e.g., because of a drug interaction, genetic difference in enzyme expression, etc.) or if conjugation pathways become saturated.


Asunto(s)
Kava/metabolismo , Extractos Vegetales/metabolismo , Acetilcisteína/orina , Adulto , Animales , Suplementos Dietéticos/efectos adversos , Glutatión/metabolismo , Humanos , Kava/química , Kava/toxicidad , Masculino , Microsomas Hepáticos/metabolismo , Extractos Vegetales/farmacocinética , Extractos Vegetales/toxicidad , Ratas , Ratas Sprague-Dawley , Espectrometría de Masa por Ionización de Electrospray
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA