Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Arch Toxicol ; 96(3): 793-808, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34989853

RESUMO

Sesquiterpene lactone helenalin is used as an antiphlogistic in European and Chinese folk medicine. The pharmacological activities of helenalin have been extensively investigated, yet insufficient information exists about its metabolic properties. The objectives of the present study were (1) to investigate the in vitro NADPH-dependent metabolism of helenalin (5 and 100 µM) using human and rat liver microsomes and liver cytosol, (2) to elucidate the role of human cytochrome P450 (CYP) enzymes in its oxidative metabolism, and (3) to study the inhibition of human CYPs by helenalin. Five oxidative metabolites were detected in NADPH-dependent human and rat liver microsomal incubations, while two reduced metabolites were detected only in NADPH-dependent human microsomal and cytosolic incubations. In human liver microsomes, the main oxidative metabolite was 14-hydroxyhelenalin, and in rat liver microsomes 9-hydroxyhelenalin. The overall oxidation of helenalin was several times more efficient in rat than in human liver microsomes. In humans, CYP3A4 and CYP3A5 followed by CYP2B6 were the main enzymes responsible for the hepatic metabolism of helenalin. The extrahepatic CYP2A13 oxidized helenalin most efficiently among CYP enzymes, possessing the Km value of 0.6 µM. Helenalin inhibited CYP3A4 (IC50 = 18.7 µM) and CYP3A5 (IC50 = 62.6 µM), and acted as a mechanism-based inhibitor of CYP2A13 (IC50 = 1.1 µM, KI = 6.7 µM, and kinact = 0.58 ln(%)/min). It may be concluded that the metabolism of helenalin differs between rats and humans, in the latter its oxidation is catalyzed by hepatic CYP2B6, CYP3A4, CYP3A5, and CYP3A7, and extrahepatic CYP2A13.


Assuntos
Inibidores das Enzimas do Citocromo P-450/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Microssomos Hepáticos/metabolismo , Sesquiterpenos de Guaiano/metabolismo , Animais , Inibidores das Enzimas do Citocromo P-450/administração & dosagem , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/efeitos dos fármacos , Feminino , Humanos , Concentração Inibidora 50 , Masculino , NADP/metabolismo , Ratos , Ratos Wistar , Sesquiterpenos de Guaiano/administração & dosagem , Sesquiterpenos de Guaiano/farmacologia , Especificidade da Espécie
2.
Xenobiotica ; 51(11): 1207-1216, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33703988

RESUMO

CYP2A13 enzyme is expressed in human extrahepatic tissues, while CYP2A6 is a hepatic enzyme. Reactions catalysed by CYP2A13 activate tobacco-specific nitrosamines and some other toxic xenobiotics in lungs.To compare oxidation characteristics and substrate-enzyme active site interactions in CYP2A13 vs CYP2A6, we evaluated CYP2A13 mediated oxidation characteristics of 23 coumarin derivatives and modelled their interactions at the enzyme active site.CYP2A13 did not oxidise six coumarin derivatives to corresponding fluorescent 7-hydroxycoumarins. The Km-values of the other coumarins varied 0.85-97 µM, Vmax-values of the oxidation reaction varied 0.25-60 min-1, and intrinsic clearance varied 26-6190 kL/min*mol CYP2A13). Km of 6-chloro-3-(3-hydroxyphenyl)-coumarin was 0.85 (0.55-1.15 95% confidence limit) µM and Vmax 0.25 (0.23-0.26) min-1, whereas Km of 6-hydroxy-3-(3-hydroxyphenyl)-coumarin was 10.9 (9.9-11.8) µM and Vmax 60 (58-63) min-1. Docking analyses demonstrated that 6-chloro or 6-methoxy and 3-(3-hydroxyphenyl) or 3-(4-trifluoromethylphenyl) substituents of coumarin increased affinity to CYP2A13, whereas 3-triazole or 3-(3-acetate phenyl) or 3-(4-acetate phenyl) substituents decreased it.The active site of CYP2A13 accepts more diversified types of coumarin substrates than the hepatic CYP2A6 enzyme. New sensitive and convenient profluorescent CYP2A13 substrates were identified, such as 6-chloro-3-(3-hydroxyphenyl)-coumarin having high affinity and 6-hydroxy-3-(3-hydroxyphenyl)-coumarin with high intrinsic clearance.


Assuntos
Hidrocarboneto de Aril Hidroxilases , Hidrocarboneto de Aril Hidroxilases/metabolismo , Cumarínicos , Citocromo P-450 CYP2A6 , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Cinética , Simulação de Acoplamento Molecular
3.
Xenobiotica ; 51(3): 268-278, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33289420

RESUMO

Catechol-O-methyltransferase (COMT) methylates both endogenous and exogenous catechol compounds to inactive and safe metabolites. We first optimised conditions for a convenient and sensitive continuous fluorescence-based 6-O-methylation assay of esculetin, which we used for investigating the COMT activity in human, mouse, rat, dog, rabbit, and sheep liver cytosols and microsomes and in ten different rat tissues. Furthermore, we compared the inhibition potencies and mechanisms of two clinically used COMT inhibitors, entacapone and tolcapone, in these species. In most tissues, the COMT activity was at least three times higher in cytosol than in microsomes. In the rat, the highest COMT activity was found in the liver, followed by kidney, ileum, thymus, spleen, lung, pancreas, heart, brain, and finally, skeletal muscle. Entacapone and tolcapone were characterised as highly potent mixed type tight-binding inhibitors. The competitive inhibition type dominated over the uncompetitive inhibition with entacapone, whereas uncompetitive inhibition dominated with tolcapone. Rats, dogs, pigs, and sheep are high COMT activity species, in contrast to humans, mice, and rabbits; COMT activity is highest in the liver. Both entacapone and tolcapone are potent COMT inhibitors, but their inhibition mechanisms differ.


Assuntos
Inibidores de Catecol O-Metiltransferase/farmacologia , Catecol O-Metiltransferase/metabolismo , Catecóis/farmacologia , Nitrilas/farmacologia , Escopoletina/metabolismo , Tolcapona/farmacologia , Umbeliferonas/metabolismo , Animais , Catálise , Cães , Humanos , Metilação , Camundongos , Coelhos , Ratos , Ovinos , Suínos
4.
Xenobiotica ; 50(8): 885-893, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31903849

RESUMO

Sulfonation is an important high affinity elimination pathway for phenolic compounds.In this study sulfonation of 7-hydroxycoumarin and 13 its derivatives were evaluated in liver cytosols of human and six animal species. 7-hydroxycoumarin and its derivatives are strongly fluorescent, and their sulfate conjugates are nonfluorescent at excitation 405 nm and emission 460 nm. A convenient fluorescence based kinetic assay of sulfonation was established.The sulfonation rate of most of the 7-hydroxycoumarin derivatives was low in liver cytosol of human and pig, whereas it was high with most compounds in dog and intermediate in rat, mouse, rabbit, and sheep. Sulfonation of the 7-hydroxycoumarin derivatives followed Michaelis-Menten kinetics with Km values of 0.1-12 µM, Vmax of 0.005-1.7 µmol/(min * g protein) and intrinsic clearance (Vmax/Km) of 0.004-1.9 L/(min * g cytosolic protein).Fluorescence based measurement of sulfonation of 7-hydroxycoumarin derivatives provides a sensitive and convenient high-throughput assay to determine sulfonation rate in different species and tissues and can be applied to evaluate sulfonation kinetics and inhibition.


Assuntos
Citosol/metabolismo , Umbeliferonas/metabolismo , Animais , Cães , Humanos , Camundongos , Coelhos , Ratos , Ovinos , Suínos
5.
Int J Mol Sci ; 21(13)2020 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-32630278

RESUMO

in vivo methods, such as spectrophotometric, fluorometric, mass spectrometric,and radioactivity-based techniques. In fluorescence-based assays, the reaction produces a fluorescentproduct from a nonfluorescent substrate or vice versa. Fluorescence-based enzyme assays areusually highly sensitive and specific, allowing measurements on small specimens of tissues withlow enzyme activities. Fluorescence assays are also amenable to miniaturization of the reactionmixtures and can thus be done in high throughput. 7-Hydroxycoumarin and its derivatives arewidely used as fluorophores due to their desirable photophysical properties. They possess a large -conjugated system with electron-rich and charge transfer properties. This conjugated structure leadsto applications of 7-hydroxycoumarins as fluorescent sensors for biological activities. We describe inthis review historical highlights and current use of coumarins and their derivatives in evaluatingactivities of the major types of xenobiotic-metabolizing enzyme systems. Traditionally, coumarinsubstrates have been used to measure oxidative activities of cytochrome P450 (CYP) enzymes. For thispurpose, profluorescent coumarins are very sensitive, but generally lack selectivity for individual CYPforms. With the aid of molecular modeling, we have recently described several new coumarin-basedsubstrates for measuring activities of CYP and conjugating enzymes with improved selectivity.


Assuntos
Cumarínicos/química , Cumarínicos/metabolismo , Xenobióticos/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Fluorescência , Humanos , Fígado/metabolismo , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Umbeliferonas/metabolismo , Xenobióticos/química
6.
Xenobiotica ; 49(9): 1015-1024, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30272491

RESUMO

Cytochrome P450 (CYP) enzymes constitute an essential xenobiotic metabolizing system that regulates the elimination of lipophilic compounds from the body. Convenient and affordable assays for CYP enzymes are important for assessing these metabolic pathways. In this study, 10 novel profluorescent coumarin derivatives with various substitutions at carbons 3, 6 and 7 were developed. Molecular modeling indicated that 3-phenylcoumarin offers an excellent scaffold for the development of selective substrate compounds for various human CYP forms, as they could be metabolized to fluorescent 7-hydroxycoumarin derivatives. Oxidation of profluorescent coumarin derivatives to fluorescent metabolites by 13 important human liver xenobiotic-metabolizing CYP forms was determined by enzyme kinetic assays. Four of the coumarin derivatives were converted to fluorescent metabolites by CYP1 family enzymes, with 6-methoxy-3-(4-trifluoromethylphenyl)coumarin being oxidized selectively by CYP1A2 in human liver microsomes. Another set of four compounds were metabolized by CYP2A6 and CYP1 enzymes. 7-Methoxy-3-(3-methoxyphenyl)coumarin was oxidized efficiently by CYP2C19 and CYP2D6 in a non-selective fashion. The advantages of the novel substrates were (1) an excellent signal-to-background ratio, (2) selectivity for CYP1 forms, and (3) convenient multiwell plate measurement, allowing for precise determination of potential inhibitors of important human hepatic forms CYP1A2, CYP2C19 and CYP2D6.


Assuntos
Cumarínicos/química , Cumarínicos/metabolismo , Sistema Enzimático do Citocromo P-450/química , Sistema Enzimático do Citocromo P-450/metabolismo , Benzoflavonas/metabolismo , Benzoflavonas/farmacologia , Cumarínicos/síntese química , Inibidores das Enzimas do Citocromo P-450/metabolismo , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Fluorescência , Humanos , Inativação Metabólica , Cinética , Microssomos Hepáticos/metabolismo , Modelos Moleculares , Simulação de Acoplamento Molecular , Oxirredução
7.
Planta Med ; 85(6): 453-464, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30736072

RESUMO

Scoparone, a major constituent of the Chinese herbal medicine Yin Chen Hao, expresses beneficial effects in experimental models of various diseases. The intrinsic doses and effects of scoparone are dependent on its metabolism, both in humans and animals. We evaluated in detail the metabolism of scoparone in human, mouse, rat, pig, dog, and rabbit liver microsomes in vitro and in humans in vivo. Oxidation of scoparone to isoscopoletin via 6-O-demethylation was the major metabolic pathway in liver microsomes from humans, mouse, rat, pig and dog, whereas 7-O-demethylation to scopoletin was the main reaction in rabbit. The scoparone oxidation rates in liver microsomes were 0.8 - 1.2 µmol/(min*g protein) in mouse, pig, and rabbit, 0.2 - 0.4 µmol/(min*g protein) in man and dog, and less than 0.1 µmol/(min*g) in rat. In liver microsomes of all species, isoscopoletin was oxidized to 3-[4-methoxy-ρ-(3, 6)-benzoquinone]-2-propenoate and esculetin, which was formed also in the oxidation of scopoletin. Human CYP2A13 exhibited the highest rate of isoscopoletin and scopoletin oxidation, followed by CYP1A1 and CYP1A2. Glucuronidation of isoscopoletin and scopoletin was catalyzed by the human UGT1A1, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, and UGT2B17. Dog was most similar to man in scoparone metabolism. Isoscopoletin glucuronide and sulfate conjugates were the major scoparone in vivo metabolites in humans, and they were completely excreted within 24 h in urine. Scoparone and its metabolites did not activate key nuclear receptors regulating CYP and UGT enzymes. These results outline comprehensively the metabolic pathways of scoparone in man and key preclinical animal species.


Assuntos
Cumarínicos/metabolismo , Medicamentos de Ervas Chinesas/metabolismo , Animais , Cumarínicos/farmacocinética , Cães , Medicamentos de Ervas Chinesas/farmacocinética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos DBA , Microssomos Hepáticos/metabolismo , Oxirredução , Coelhos , Ratos , Ratos Wistar , Suínos
8.
Mol Pharm ; 15(3): 923-933, 2018 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-29421866

RESUMO

Intestinal and hepatic glucuronidation by the UDP-glucuronosyltransferases (UGTs) greatly affect the bioavailability of phenolic compounds. UGT1A10 catalyzes glucuronidation reactions in the intestine, but not in the liver. Here, our aim was to develop selective, fluorescent substrates to easily elucidate UGT1A10 function. To this end, homology models were constructed and used to design new substrates, and subsequently, six novel C3-substituted (4-fluorophenyl, 4-hydroxyphenyl, 4-methoxyphenyl, 4-(dimethylamino)phenyl, 4-methylphenyl, or triazole) 7-hydroxycoumarin derivatives were synthesized from inexpensive starting materials. All tested compounds could be glucuronidated to nonfluorescent glucuronides by UGT1A10, four of them highly selectively by this enzyme. A new UGT1A10 mutant, 1A10-H210M, was prepared on the basis of the newly constructed model. Glucuronidation kinetics of the new compounds, in both wild-type and mutant UGT1A10 enzymes, revealed variable effects of the mutation. All six new C3-substituted 7-hydroxycoumarins were glucuronidated faster by human intestine than by liver microsomes, supporting the results obtained with recombinant UGTs. The most selective 4-(dimethylamino)phenyl and triazole C3-substituted 7-hydroxycoumarins could be very useful substrates in studying the function and expression of the human UGT1A10.


Assuntos
Desenho de Fármacos , Corantes Fluorescentes/química , Glucuronosiltransferase/metabolismo , Simulação de Acoplamento Molecular , Imagem Molecular/métodos , Corantes Fluorescentes/metabolismo , Glucuronídeos/metabolismo , Glucuronosiltransferase/química , Glucuronosiltransferase/genética , Humanos , Microssomos , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Especificidade por Substrato , Umbeliferonas/química , Umbeliferonas/metabolismo
9.
Rapid Commun Mass Spectrom ; 32(16): 1344-1352, 2018 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-29788543

RESUMO

RATIONALE: The toxic metabolites of pyrrolizidine alkaloids (PAs) are initially formed by cytochrome P450-mediated oxidation reactions and primarily eliminated as glutathione (GSH) conjugates. Although the reaction between the reactive metabolites and GSH can occur spontaneously, the role of the cytosolic enzymes in the process has not been studied. METHODS: The toxic metabolites of selected PAs (retrorsine, monocrotaline, senecionine, lasiocarpine, heliotrine or senkirkine) were generated by incubating them in 100 mM phosphate buffer (pH 7.4) containing liver microsomes of human, pig, rat or sheep, NADPH and reduced GSH in the absence or presence of human, pig, rat or sheep liver cytosolic fraction. The supernatants were analyzed using liquid chromatography connected to Finnigan LTQ ion-trap, Agilent QTOF or Thermo Scientific Q Exactive Focus quadrupole-orbitrap mass spectrometers. RESULTS: Retrorsine, senecionine and lasiocarpine yielded three GSH conjugates producing [M - H]- ions at m/z 439 (7-GSH-DHP (CHO)), m/z 441 (7-GSH-DHP (OH)) and m/z 730 (7,9-diGSH-DHP) in the presence of human liver cytosolic fraction. 7-GSH-DHP (CHO) was a novel metabolite. Monocrotaline, heliotrine and senkirkine did not produce this novel 7-GSH-DHP (CHO) conjugate. 7-GSH-DHP (CHO) disappeared when incubated with hydroxylamine, and a new oxime derivative was formed. This metabolite was formed only by the human liver cytosolic enzymes but not in the presence of rat or sheep liver cytosolic fractions under otherwise identical reaction conditions. CONCLUSIONS: 7-GSH-DHP (CHO) has not been reported before, and thus it was considered as a novel metabolite of PAs. This may clarify the mechanisms involved in PA detoxification and widely observed but less understood species differences in response to PA exposure.


Assuntos
Glutationa/metabolismo , Microssomos Hepáticos/metabolismo , Alcaloides de Pirrolizidina , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Humanos , Oxirredução , Alcaloides de Pirrolizidina/análise , Alcaloides de Pirrolizidina/química , Alcaloides de Pirrolizidina/metabolismo , Ratos , Ovinos , Suínos
10.
Planta Med ; 84(5): 320-328, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28950382

RESUMO

Scoparone is a natural bioactive compound in Chinese herbal medicines. It has numerous pharmacological actions, including liver protective, hypolipidemic, antitumor, and anti-inflammatory effects. The primary metabolism route of scoparone is O-demethylation to scopoletin or isoscopoletin catalyzed by CYP enzymes. The aims of our study were to identify the human CYP enzymes catalyzing scoparone 7-O-demethylation to scopoletin and to compare this oxidation reaction in liver microsomes among different species. A high throughput fluorescent-based assay method was developed to determine the scoparone 7-O-demethylation to scopoletin rate. The rate was 100 - 400 nmol/(min×g protein) in mouse and rabbit liver microsomes, 10 - 20 nmol/(min×g protein) in pig microsomes, 1 - 3 nmol/(min×g protein) in human and less than 1 nmol/(min×g protein) in rat liver microsomes. Human CYP1A1 (Km 13 µM and Vmax 0.8 min-1), CYP1A2 (Km 48 µM and Vmax 0.3 min-1), and CYP2A13 (Km 10 µM and Vmax 22 min-1) were the most efficient catalysts of the reaction. The CYP2A6 selective inhibitor pilocarpine and an antibody against mouse CYP2A5 inhibited scoparone 7-O-demethylation to scopoletin in rabbit, mouse, and pig liver microsomes, indicating involvement of CYP2A enzymes in the reaction. Hepatic scoparone 7-O-demethylation to scopoletin differed between species both with respect to the rate of reaction and catalyzing enzymes. These species differences need to be taken into account when testing scoparone pharmacokinetics in animals and humans.


Assuntos
Cumarínicos/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Escopoletina/análogos & derivados , Escopoletina/metabolismo , Animais , Hidrocarboneto de Aril Hidroxilases/metabolismo , Cumarínicos/química , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Desmetilação , Feminino , Humanos , Masculino , Camundongos , Microssomos Hepáticos/enzimologia , Estrutura Molecular , Oxirredução , Coelhos , Ratos , Escopoletina/química , Suínos
11.
J Enzyme Inhib Med Chem ; 33(1): 743-754, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29620427

RESUMO

A comprehensive set of 3-phenylcoumarin analogues with polar substituents was synthesised for blocking oestradiol synthesis by 17-ß-hydroxysteroid dehydrogenase 1 (HSD1) in the latter part of the sulphatase pathway. Five analogues produced ≥62% HSD1 inhibition at 5 µM and, furthermore, three of them produced ≥68% inhibition at 1 µM. A docking-based structure-activity relationship analysis was done to determine the molecular basis of the inhibition and the cross-reactivity of the analogues was tested against oestrogen receptor, aromatase, cytochrome P450 1A2, and monoamine oxidases. Most of the analogues are only modestly active with 17-ß-hydroxysteroid dehydrogenase 2 - a requirement for lowering effective oestradiol levels in vivo. Moreover, the analysis led to the synthesis and discovery of 3-imidazolecoumarin as a potent aromatase inhibitor. In short, coumarin core can be tailored with specific ring and polar moiety substitutions to block either the sulphatase pathway or the aromatase pathway for treating breast cancer and endometriosis.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Cumarínicos/farmacologia , Inibidores Enzimáticos/farmacologia , Estradiol/biossíntese , 17-Hidroxiesteroide Desidrogenases/metabolismo , Desenho Assistido por Computador , Cumarínicos/síntese química , Cumarínicos/química , Relação Dose-Resposta a Droga , Descoberta de Drogas , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Ligantes , Simulação de Acoplamento Molecular , Estrutura Molecular , Relação Estrutura-Atividade
12.
Xenobiotica ; 46(1): 14-24, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26068522

RESUMO

1. Information about the metabolism of compounds is essential in drug discovery and development, risk assessment of chemicals and further development of predictive methods. 2. In vitro and in silico methods were applied to evaluate the metabolic and inhibitory properties of 6-methylcoumarin, 7-methylcoumarin and 7-formylcoumarin with human CYP2A6, mouse CYP2A5 and pig CYP2A19. 3. 6-Methylcoumarin was oxidized to fluorescent 7-hydroxy-6-methylcoumarin by CYP2A6 (Km: 0.64-0.91 µM; Vmax: 0.81-0.89 min(-1)) and by CYP2A5 and CYP2A19. The reaction was almost completely inhibited at 10 µM 7-methylcoumarin in liver microsomes of human and mouse, but in pig only 40% inhibition was obtained with the anti-CYP2A5 antibody or with methoxsalen and pilocarpine. 7-Methylcoumarin was a mechanism-based inhibitor for CYP2A6, but not for the mouse and pig enzymes. 7-Formylcoumarin was a mechanism-based inhibitor for CYP2As of all species. 4. Docking and molecular dynamics simulations of 6-methylcoumarin and 7-methylcoumarin in the active sites of CYP2A6 and CYP2A5 demonstrated a favorable orientation of the 7-position of 6-methylcoumarin towards the heme moiety. Several orientations of 7-methylcoumarin were possible in CYP2A6 and CYP2A5. 5. These results indicate that the active site of CYP2A6 has unique interaction properties for ligands and differs in this respect from CYP2A5 and CYP2A19.


Assuntos
Cumarínicos/farmacologia , Citocromo P-450 CYP2A6/antagonistas & inibidores , Inibidores das Enzimas do Citocromo P-450/farmacologia , Animais , Citocromo P-450 CYP2A6/metabolismo , Humanos , Hidroxilação , Concentração Inibidora 50 , Cinética , Camundongos , Modelos Moleculares , Oxirredução , Sus scrofa , Fatores de Tempo
13.
Chem Res Toxicol ; 28(10): 2034-44, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26395423

RESUMO

There are species-related differences in the toxicity of pyrrolizidine alkaloids (PAs) partly attributable to the hepatic metabolism of these alkaloids. In this study, the metabolism of lasiocarpine, a potent hepatotoxic and carcinogenic food contaminant, was examined in vitro with human, pig, rat, mouse, rabbit, and sheep liver microsomes. A total of 12 metabolites (M1-M12) were detected with the human liver microsomes, of which M1, M2, M4, and M6 were unstable in the presence of reduced glutathione (GSH). With the exception of M3 and M8, the formation of all metabolites of lasiocarpine was catalyzed by CYP3A4 in humans. Tandem mass spectra (MS/MS) detected several new metabolites, termed M4-M7; their toxicological significance is unknown. M9 (m/z 398), identified as a demethylation product, was the main metabolite in all species, although the relative dominance of this metabolite was lower in humans. The level of the reactive metabolites, as measured by M1 ((3H-pyrrolizin-7-yl)methanol) and the GSH conjugate, was higher with the liver microsomes of susceptible species (human, pig, rat, and mouse) than with the species (rabbit and sheep) resistant to PA intoxication. In general, in addition to the new metabolites (M4-M7) that could make humans more susceptible to lasiocarpine-induced toxicity, the overall metabolite fingerprint detected with the human liver microsomes differed from that of all other species, yielding high levels of GSH-reactive metabolites.


Assuntos
Fígado/metabolismo , Alcaloides de Pirrolizidina/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Citocromo P-450 CYP3A/metabolismo , Glutationa/química , Glutationa/metabolismo , Humanos , Fígado/efeitos dos fármacos , Camundongos , Microssomos Hepáticos/metabolismo , Alcaloides de Pirrolizidina/química , Alcaloides de Pirrolizidina/toxicidade , Coelhos , Ratos , Ovinos , Especificidade da Espécie , Suínos , Espectrometria de Massas em Tandem
14.
Chem Res Toxicol ; 28(4): 702-10, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25651456

RESUMO

In humans, the metabolic bioactivation of pyrrolizidine alkaloids (PAs) is mediated mainly by cytochrome P450 3A4 (CYP3A4) via the hydroxylation of their necine bases at C3 or C8 of heliotridine- and retronecine-type PAs or at the N atom of the methyl substituent of otonecine-type PAs. However, no attempts have been made to identify which C atom is the most favorable site for hydroxylation in silico. Here, in order to determine the site of hydroxylation that eventually leads to the formation of the toxic metabolites produced from lasiocarpine, retrorsine, and senkirkin, we utilized the ligand-based electrophilic Fukui function f(-)(r) and hydrogen-bond dissociation energies (BDEs) as well as structure-based molecular docking. The ligand-based computations revealed that the C3 and C8 atoms of lasiocarpine and retrorsine and the C26 atom of senkirkin were chemically the most susceptible locations for electrophilic oxidizing reactions. Similarly, according to the predicted binding orientation in the active site of the crystal structure of human CYP3A4 (PDB code: 4I4G ), the alkaloids were positioned in such a way that the C3 atom of lasiocarpine and retrorsine and the C26 of senkirkin were closest to the catalytic heme Fe. Thus, it is concluded that the C3 atom of lasiocarpine and retrorsine and C26 of senkirkin are the most favored sites of hydroxylation that lead to the production of their toxic metabolites.


Assuntos
Citocromo P-450 CYP3A/metabolismo , Alcaloides de Pirrolizidina/toxicidade , Simulação por Computador , Humanos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Modelos Moleculares , Oxirredução , Alcaloides de Pirrolizidina/metabolismo , Termodinâmica
15.
Pharmacol Rev ; 64(3): 520-39, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22544865

RESUMO

Aldehyde dehydrogenases (ALDHs) belong to a superfamily of enzymes that play a key role in the metabolism of aldehydes of both endogenous and exogenous derivation. The human ALDH superfamily comprises 19 isozymes that possess important physiological and toxicological functions. The ALDH1A subfamily plays a pivotal role in embryogenesis and development by mediating retinoic acid signaling. ALDH2, as a key enzyme that oxidizes acetaldehyde, is crucial for alcohol metabolism. ALDH1A1 and ALDH3A1 are lens and corneal crystallins, which are essential elements of the cellular defense mechanism against ultraviolet radiation-induced damage in ocular tissues. Many ALDH isozymes are important in oxidizing reactive aldehydes derived from lipid peroxidation and thereby help maintain cellular homeostasis. Increased expression and activity of ALDH isozymes have been reported in various human cancers and are associated with cancer relapse. As a direct consequence of their significant physiological and toxicological roles, inhibitors of the ALDH enzymes have been developed to treat human diseases. This review summarizes known ALDH inhibitors, their mechanisms of action, isozyme selectivity, potency, and clinical uses. The purpose of this review is to 1) establish the current status of pharmacological inhibition of the ALDHs, 2) provide a rationale for the continued development of ALDH isozyme-selective inhibitors, and 3) identify the challenges and potential therapeutic rewards associated with the creation of such agents.


Assuntos
Aldeído Desidrogenase/antagonistas & inibidores , Inibidores Enzimáticos , Aldeído Desidrogenase/química , Animais , Sítios de Ligação , Ensaios Clínicos como Assunto , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Modelos Moleculares , Estrutura Molecular , Especificidade por Substrato
16.
Chem Res Toxicol ; 27(11): 1950-7, 2014 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-25295702

RESUMO

Pyrrolizidine alkaloids (PAs) such as retrorsine are common food contaminants that are known to be bioactivated by cytochrome P450 enzymes to putative hepatotoxic, genotoxic, and carcinogenic metabolites known as dehydropyrrolizidine alkaloids (DHPs). We compared how both electrochemical (EC) and human liver microsomal (HLM) oxidation of retrorsine could produce short-lived intermediate metabolites; we also characterized a toxicologically important metabolite, (3H-pyrrolizin-7-yl)methanol. The EC cell was coupled online or offline to a liquid chromatograph/mass spectrometer (LC/MS), whereas the HLM oxidation was performed in 100 mM potassium phosphate (pH 7.4) in the presence of NADPH at 37 °C. The EC cell oxidation of retrorsine produced 12 metabolites, including dehydroretrorsine (m/z 350, [M + H(+)]), which was degraded to a new reactive metabolite at m/z 136 ([M + H(+)]). The molecular structure of this small metabolite was determined using high-resolution mass spectrometry and NMR spectroscopy followed by chemical synthesis. In addition, we also identified another minor but reactive metabolite at m/z 136, an isomer of (3H-pyrrolizin-7-yl)methanol. Both (3H-pyrrolizin-7-yl)methanol and its minor isomer were also observed after HLM oxidation of retrorsine and other hepatotoxic PAs such as lasiocarpine and senkirkin. In the presence of reduced glutathione (GSH), each isomer formed identical GSH conjugates at m/z 441 and m/z 730 in the negative ESI-MS. Because (3H-pyrrolizine-7-yl)methanol) and its minor isomer subsequently reacted with GSH, it is concluded that (3H-pyrrolizin-7-yl)methanol may be a common toxic metabolite arising from PAs.


Assuntos
Microssomos Hepáticos/efeitos dos fármacos , Alcaloides de Pirrolizidina/metabolismo , Biotransformação , Cromatografia Líquida de Alta Pressão , Técnicas Eletroquímicas , Técnicas In Vitro , Espectroscopia de Ressonância Magnética , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Oxirredução , Alcaloides de Pirrolizidina/química , Alcaloides de Pirrolizidina/toxicidade , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
17.
Bioorg Med Chem ; 22(23): 6655-6664, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25458499

RESUMO

Inhibition of CYP2A6-mediated nicotine metabolism can reduce cigarette smoking. We sought potent and selective CYP2A6 inhibitors to be used as leads for drugs useful in smoking reduction therapy, by evaluating CYP2A6 inhibitory effect of novel formyl, alkyl amine or carbonitrile substituted aromatic core structures. The most potent CYP2A6 inhibitors were thienopyridine-2-carbaldehyde, benzothienophene-3-ylmethanamine, benzofuran-5-carbaldehyde and indole-5-carbaldehyde, with IC50 values below 0.5 µM for coumarin 7-hydroxylation. Nicotine oxidation was effectively inhibited in vitro by two alkyl amine compounds and benzofuran-5-carbonitrile. Some of these molecules could serve as potential lead molecules when designing CYP2A6 inhibitory drugs for smoking reduction therapy.


Assuntos
Citocromo P-450 CYP2A6/antagonistas & inibidores , Inibidores das Enzimas do Citocromo P-450/farmacologia , Desenho de Fármacos , Piridinas/farmacologia , Citocromo P-450 CYP2A6/metabolismo , Inibidores das Enzimas do Citocromo P-450/síntese química , Inibidores das Enzimas do Citocromo P-450/química , Relação Dose-Resposta a Droga , Humanos , Modelos Moleculares , Estrutura Molecular , Piridinas/síntese química , Piridinas/química , Abandono do Hábito de Fumar , Prevenção do Hábito de Fumar , Relação Estrutura-Atividade
18.
Basic Clin Pharmacol Toxicol ; 134(6): 846-857, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38664998

RESUMO

Fungal anthraquinones dermocybin and dermorubin are attractive alternatives for synthetic dyes but their metabolism is largely unknown. We conducted a qualitative in vitro study to identify their metabolism using human liver microsomes and cytosol, as well as recombinant human cytochrome P450 (CYP), UDP-glucuronosyltransferase (UGT) and sulfotransferase (SULT) enzymes. Additionally, liver microsomal and cytosolic fractions from rat, mouse and pig were used. Following incubations of the biocolourants with the enzymes in the presence of nicotinamide adenine dinucleotide phosphate, UDP-glucuronic acid, 3'-phosphoadenosine-5'-phosphosulfate (PAPS) or S-adenosyl methionine (SAM) to enable CYP oxidation, glucuronidation, sulfonation or methylation, we observed several oxidation and conjugation metabolites for dermocybin but none for dermorubin. Human CYP1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4 and 3A7 catalysed dermocybin oxidation. The formation of dermocybin glucuronides was catalysed by human UGT1A1, 1A3, 1A7, 1A8, 1A9, 1A10 and 2B15. Human SULT1B1, 1C2 and 2A1 sulfonated dermocybin. Dermocybin oxidation was faster than conjugation in human liver microsomes. Species differences were seen in dermocybin glucuronidation between human, rat, mouse and pig. In conclusion, many CYP and conjugation enzymes metabolized dermocybin, whereas dermorubin was not metabolized in human liver fractions in vitro. The results indicate that dermocybin would be metabolized in humans in vivo.


Assuntos
Antraquinonas , Sistema Enzimático do Citocromo P-450 , Glucuronosiltransferase , Microssomos Hepáticos , Microssomos Hepáticos/metabolismo , Humanos , Animais , Ratos , Camundongos , Suínos , Glucuronosiltransferase/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Antraquinonas/metabolismo , Masculino , Proteínas Recombinantes/metabolismo , Fígado/metabolismo , Fígado/enzimologia , Citosol/metabolismo , Oxirredução , Glucuronídeos/metabolismo
19.
Mol Pharm ; 10(2): 532-7, 2013 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-22937971

RESUMO

Four novel cyclic phosphates of the anti-inflammatory agent 5-aminosalicylic acid (5-ASA) were designed and synthesized as cytochrome P450 (CYP)-activated prodrugs. These prodrugs can be used for targeting into gut wall, since these types of cyclic phosphates are known to be activated mainly by CYP3A forms, which are expressed not only in the liver but also in the small intestine and to a lesser extent in the colon. The present study shows that aromatic ring activating substituents, like chlorine, are definitely needed to obtain the desired enzymatic cleavage of the cyclic phosphate prodrugs of 5-ASA. However, the position of the activating substituent has also a strong impact on the chemical stability, and therefore, an appropriate balance between the rates of prodrug bioactivation and chemical stability needs to be taken into consideration in future studies on cyclic phosphate prodrugs of 5-ASA.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Mesalamina/química , Mesalamina/síntese química , Pró-Fármacos/química , Pró-Fármacos/síntese química , Ativação Enzimática/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Mesalamina/farmacologia , Estrutura Molecular , Pró-Fármacos/farmacologia
20.
Xenobiotica ; 43(10): 853-61, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23551063

RESUMO

1. UDP-glucuronosyltransferases (UGTs) are versatile and important conjugation enzymes in the metabolism of drugs and other xenobiotics. 2. We have developed a convenient quantitative multi-well plate assay to measure the glucuronidation rate of 7-hydroxy-4-trifluoromethylcoumarin (HFC) for several UGTs. 3. We have used this method to screen 11 recombinant human UGTs for HFC glucuronidation activity and studied the reaction kinetics with the most active enzymes. We have also examined the HFC glucuronidation activity of liver microsomes from human, pig, rabbit and rat. 4. At a substrate concentration of 20 µM, the most active HFC glucuronidation catalysts were UGT1A10 followed by UGT1A6 >UGT1A7 >UGT2A1, whereas at 300 µM UGT1A6 was about 10 times better catalyst than the other recombinant UGTs. The activities of UGTs 1A3, 1A8, 1A9, 2B4 and 2B7 were low, whereas UGT1A1 and UGT2B17 exhibited no HFC glucuronidation activity. UGT1A6 exhibited a significantly higher Vmax and Km values toward both HFC and UDP-glucuronic acid than the other UGTs. 5. Human, pig and rabbit, but not rat liver microsomes, catalyzed HFC glucuronidation at high rates. 6. This new method is particularly suitable for fast activity screenings of UGTs 1A6, 1A7, 1A10 and 2A1 and HFC glucuronidation activity determination from various samples.


Assuntos
Cumarínicos/metabolismo , Glucuronosiltransferase/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Microssomos Hepáticos/metabolismo , Animais , Cumarínicos/análise , Feminino , Glucuronosiltransferase/análise , Glucuronosiltransferase/genética , Ensaios de Triagem em Larga Escala/instrumentação , Humanos , Cinética , Masculino , Coelhos , Ratos , Ratos Wistar , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA