Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochem Pharmacol ; 159: 25-31, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30423313

RESUMO

The activities of hundreds, perhaps thousands, of metabolites are regulated by human cytosolic sulfotransferases (SULTs) - a 13-member family of disease relevant enzymes that catalyze transfer of the sulfuryl moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfonate) to the hydroxyls and amines of acceptors. SULTs harbor two independent allosteric sites, one of which, the focus of this work, binds non-steroidal anti-inflammatory drugs (NSAIDs). The structure of the first NSAID-binding site - that of SULT1A1 - was elucidated recently and homology modeling suggest that variants of the site are present in all SULT isoforms. The objective of the current study was to assess whether the NSAID-binding site can be used to regulate sulfuryl transfer in humans in an isoform specific manner. Mefenamic acid (Mef) is a potent (Ki 27 nM) NSAID-inhibitor of SULT1A1 - the predominant SULT isoform in small intestine and liver. Acetaminophen (APAP), a SULT1A1 specific substrate, is extensively sulfonated in humans. Dehydroepiandrosterone (DHEA) is specific for SULT2A1, which we show here is insensitive to Mef inhibition. APAP and DHEA sulfonates are readily quantified in urine and thus the effects of Mef on APAP and DHEA sulfonation could be studied non-invasively. Compounds were given orally in a single therapeutic dose to a healthy, adult male human with a typical APAP-metabolite profile. Mef profoundly decreased APAP sulfonation during first pass metabolism and substantially decreased systemic APAP sulfonation without influencing DHEA sulfonation; thus, it appears the NSAID site can be used to control sulfonation in humans in a SULT-isoform specific manner.


Assuntos
Acetaminofen/farmacocinética , Arilsulfotransferase/metabolismo , Ácido Mefenâmico/farmacocinética , Sulfotransferases/metabolismo , Acetaminofen/metabolismo , Acetaminofen/urina , Sítio Alostérico , Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacocinética , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/química , Sítios de Ligação , Desidroepiandrosterona/administração & dosagem , Desidroepiandrosterona/metabolismo , Desidroepiandrosterona/urina , Interações Medicamentosas , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Espectroscopia de Ressonância Magnética , Ácido Mefenâmico/metabolismo , Ácido Mefenâmico/urina , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/química
2.
J Biol Chem ; 292(49): 20305-20312, 2017 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-29038294

RESUMO

Non-steroidal anti-inflammatory drugs (NSAIDs) are among the most commonly prescribed drugs worldwide-more than 111 million prescriptions were written in the United States in 2014. NSAIDs allosterically inhibit cytosolic sulfotransferases (SULTs) with high specificity and therapeutically relevant affinities. This study focuses on the interactions of SULT1A1 and mefenamic acid (MEF)-a potent, highly specific NSAID inhibitor of 1A1. Here, the first structure of an NSAID allosteric site-the MEF-binding site of SULT1A1-is determined using spin-label triangulation NMR. The structure is confirmed by site-directed mutagenesis and provides a molecular framework for understanding NSAID binding and isoform specificity. The mechanism of NSAID inhibition is explored using molecular dynamics and equilibrium and pre-steady-state ligand-binding studies. MEF inhibits SULT1A1 turnover through an indirect (helix-mediated) stabilization of the closed form of the active-site cap of the enzyme, which traps the nucleotide and slows its release. Using the NSAID-binding site structure of SULT1A1 as a comparative model, it appears that 11 of the 13 human SULT isoforms harbor an NSAID-binding site. We hypothesize that these sites evolved to enable SULT isoforms to respond to metabolites that lie within their metabolic domains. Finally, the NSAID-binding site structure offers a template for developing isozyme-specific allosteric inhibitors that can be used to regulate specific areas of sulfuryl-transfer metabolism.


Assuntos
Sítio Alostérico , Anti-Inflamatórios não Esteroides/metabolismo , Citosol/enzimologia , Sulfotransferases/química , Arilsulfotransferase/antagonistas & inibidores , Humanos , Isoenzimas/metabolismo , Espectroscopia de Ressonância Magnética , Ácido Mefenâmico/metabolismo , Ácido Mefenâmico/farmacologia , Ligação Proteica , Sulfotransferases/antagonistas & inibidores
3.
Toxicol Lett ; 262: 27-38, 2016 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-27633141

RESUMO

Melatonin (Mel) as an endogenous hormone, has been widely used in clinic for multiple therapeutic purposes. Further, the natural anthraquinones were widespread in various plants including herbs, foods, and some flavoring agents. The present work aims to evaluate the metabolic disorder of Mel caused by various common herbs and further identify their underlying mechanism. More importantly, the relationships between inhibitory activity and their structures were also investigated. Our results demonstrate that some herbs containing anthraquinone derivatives exhibited strong inhibition on Mel metabolism. Additionally, five anthraquinones from R. palmatum could inhibit phase I and II metabolism of Mel with a mixed inhibition kinetic model based on the mechanism of inhibiting human CYP1A1, 1A2, and SULT1A1. At last, the influence of R. palmatum and its five major components on the Mel metabolism were verified in human primary hepatocytes. In conclusion, our studies elucidated that herbs or foods containing abundant anthraquinones such as R. palmatum will cause a metabolic disorder of Mel, and should be avoided to combined application with Mel in clinic.


Assuntos
Antraquinonas/farmacologia , Arilsulfotransferase/antagonistas & inibidores , Inibidores das Enzimas do Citocromo P-450/farmacologia , Melatonina/metabolismo , Rheum/química , Antraquinonas/química , Arilsulfotransferase/metabolismo , Citocromo P-450 CYP1A1/efeitos dos fármacos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/efeitos dos fármacos , Citocromo P-450 CYP1A2/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Doenças Metabólicas , Modelos Moleculares , Raízes de Plantas/química , Cultura Primária de Células , Relação Estrutura-Atividade
4.
Carcinogenesis ; 37(7): 647-655, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27207664

RESUMO

Aristolochic acids (AA) are implicated in the development of chronic renal disease and upper urinary tract carcinoma in humans. Using in vitro approaches, we demonstrated that N-hydroxyaristolactams, metabolites derived from partial nitroreduction of AA, require sulfotransferase (SULT)-catalyzed conjugation with a sulfonyl group to form aristolactam-DNA adducts. Following up on this observation, bioactivation of AA-I and N-hydroxyaristolactam I (AL-I-NOH) was studied in human kidney (HK-2) and skin fibroblast (GM00637) cell lines. Pentachlorophenol, a known SULT inhibitor, significantly reduced cell death and aristolactam-DNA adduct levels in HK-2 cells following exposure to AA-I and AL-I-NOH, suggesting a role for Phase II metabolism in AA activation. A gene knockdown, siRNA approach was employed to establish the involvement of selected SULTs and nitroreductases in AA-I bioactivation. Silencing of SULT1A1 and PAPSS2 led to a significant decrease in aristolactam-DNA levels in both cell lines following exposure to AA-I, indicating the critical role for sulfonation in the activation of AA-I in vivo Since HK-2 cells proved relatively resistant to knockdown with siRNAs, gene silencing of xanthine oxidoreductase, cytochrome P450 oxidoreductase and NADPH:quinone oxidoreductase was conducted in GM00637 cells, showing a significant increase, decrease and no effect on aristolactam-DNA levels, respectively. In GM00637 cells exposed to AL-I-NOH, suppressing the SULT pathway led to a significant decrease in aristolactam-DNA formation, mirroring data obtained for AA-I. We conclude from these studies that SULT1A1 is involved in the bioactivation of AA-I through the sulfonation of AL-I-NOH, contributing significantly to the toxicities of AA observed in vivo.


Assuntos
Ácidos Aristolóquicos/metabolismo , Arilsulfotransferase/genética , Complexos Multienzimáticos/genética , Sulfato Adenililtransferase/genética , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/metabolismo , Carcinógenos/metabolismo , Carcinógenos/toxicidade , DNA/genética , DNA/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Rim/metabolismo , Rim/patologia , Complexos Multienzimáticos/metabolismo , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Pentaclorofenol/farmacologia , RNA Interferente Pequeno , Sulfato Adenililtransferase/metabolismo , Xantina Desidrogenase/metabolismo
5.
Toxicol Lett ; 243: 98-110, 2016 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-26739637

RESUMO

Rat B-13 progenitor cells are readily converted into functional hepatocyte-like B-13/H cells capable of phase I cytochrome P450-dependent activation of pro-carcinogens and induction of DNA damage. The aim of the present study was to investigate whether the cells are also capable of Phase II sulphotransferase (SULT)-dependent activation of a pro-carcinogen to an ultimate carcinogen. To this end we therefore examined the bioactivation of the model hepatic (hepato- and cholangio-) carcinogen estragole and its proximate SULT1A1-activated genotoxic metabolite 1'-hydroxyestragole. Exposing B-13 or B-13/H cells to estragole (at concentrations up to 1mM) resulted in the production of low levels of 1'-hydroxyestragole, but did not result in detectable DNA damage. Exposing B-13/H cells - but not B-13 cells - to 1'-hydroxyestragole resulted in a dose-dependent increase in DNA damage in comet assays, confirmed by detection of N(2)-(trans-isoestragol-3'-yl)-2'-deoxyguanosine adducts. Genotoxicity was inhibited by general SULT inhibitors, supporting a role for SULTS in the activation of 1-hydroxyestragole in B-13/H cells. However, B-13 and B-13/H cells did not express biologically significant levels of SULT1A1 as determined by qRT-PCR, Western blotting and its associated 7-hydroxycoumarin sulphation activity. B-13 and B-13/H cells expressed - relative to intact rat liver - high levels of SULT2B1 (primarily the b isoform) and SULT4A1 mRNAs and proteins. B-13 and B-13/H cells also expressed the 3'-phosphoadenosine 5'-phosphosulphate synthase 1 required for the generation of activated sulphate cofactor 3'-phosphoadenosine 5'-phosphosulphate. However, only B-13/H cells expressed functional SULT activities towards SULT2B1 substrates DHEA, pregnenolone and 4 methylumbelliferone. Since liver progenitor cells are bi-potential and also form cholangiocytes, we therefore hypothesised that B-13 cells express a cholangiocyte-like SULT profile. To test this hypothesis, the expression of SULTs was examined in liver by RT-PCR and immunohistochemistry. SULT2B1 - but not SULT1A1 - was determined to be expressed in both rat and human cholangiocytes. Since 1'-hydroxyestragole exposure readily produced DNA injury in B-13/H cells, these data suggest that cholangiocarcinomas generated in rats fed estragole may be dependent, in part, on SULT2B1 activation of the 1'-hydroxyestragole metabolite.


Assuntos
Anisóis/toxicidade , Dano ao DNA/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Sulfotransferases/genética , Derivados de Alilbenzenos , Animais , Anisóis/administração & dosagem , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Carcinógenos/administração & dosagem , Carcinógenos/toxicidade , Linhagem Celular , Ensaio Cometa , Desidroepiandrosterona/farmacologia , Regulação da Expressão Gênica , Humanos , Himecromona/farmacologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Pregnenolona/farmacologia , Ratos , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/metabolismo
6.
Biochem Pharmacol ; 98(1): 203-14, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26291395

RESUMO

Efflux transport is a critical determinant to the pharmacokinetics of sulfate conjugates. Here we aimed to establish SULT1A3 stably transfected HEK293 cells, and to determine the contributions of BCRP and MRP transporters to excretion of chrysin and apigenin sulfates. The cDNA of SULT1A3 was stably introduced into HEK293 cells using a lentiviral vector, generating a sulfonation active cell line (i.e., SULT293 cells). Identification of sulfate transporters was achieved through chemical inhibition (using chemical inhibitors) and biological inhibition (using short-hairpin RNAs (shRNAs)) methods. Sulfated metabolites were rapidly generated and excreted upon incubation of SULT293 cells with chrysin and apigenin. Ko143 (a selective BCRP inhibitor) did not show inhibitory effects on sulfate disposition, whereas the pan-MRP inhibitor MK-571 caused significant reductions (38.5-64.3%, p<0.001) in sulfate excretion and marked elevations (160-243%, p<0.05) in sulfate accumulation. Further, two efflux transporters (BCRP and MRP4) expressed in the cells were knocked-down by shRNA-mediated silencing. Neither sulfate excretion nor sulfate accumulation was altered in BCRP knocked-down cells as compared to scramble cells. By contrast, MRP4 knock-down led to moderate decreases (17.1-20.6%, p<0.05) in sulfate excretion and increases (125-135%, p<0.05) in sulfate accumulation. In conclusion, MRP4 was identified as an exporter for chrysin and apigenin sulfates. The SULT1A3 modified HEK293 cells were an appropriate tool to study SULT1A3-mediated sulfonation and to characterize BCRP/MRP4-mediated sulfate transport.


Assuntos
Apigenina/química , Apigenina/metabolismo , Arilsulfotransferase/metabolismo , Flavonoides/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Dicetopiperazinas , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Compostos Heterocíclicos de 4 ou mais Anéis , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Propionatos/farmacologia , Quinolinas/farmacologia , Sulfatos/metabolismo
7.
Drug Metab Dispos ; 43(6): 843-50, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25819444

RESUMO

Tamoxifen is successfully used for both treatment and prevention of estrogen-dependent breast cancer, yet side effects and development of resistance remain problematic. Endoxifen is a major active metabolite of tamoxifen that is being investigated for clinical use. We hypothesized that endoxifen and perhaps other major metabolites of tamoxifen may affect the ability of human estrogen sulfotransferase 1E1 (hSULT1E1) and human phenol sulfotransferase 1A1 isoform 1 (hSULT1A1*1) to catalyze the sulfation of estradiol, an important mechanism in termination of estrogen signaling through loss of activity at estrogen receptors. Our results indicated that endoxifen, N-desmethyltamoxifen (N-desTAM), 4-hydroxytamoxifen (4-OHTAM), and tamoxifen-N-oxide were weak inhibitors of hSULT1E1 with Ki values ranging from 10 µM to 38 µM (i.e., over 1000 times higher than the 8.1 nM Km value for estradiol as substrate for the enzyme). In contrast to the results with hSULT1E1, endoxifen and 4-OHTAM were significant inhibitors of the sulfation of 2.0 µM estradiol catalyzed by hSULT1A1*1, with IC50 values (9.9 µM and 1.6 µM, respectively) that were similar to the Km value (1.5 µM) for estradiol as substrate for this enzyme. Additional investigation of the interaction of these metabolites with the two sulfotransferases revealed that endoxifen, 4-OHTAM, and N-desTAM were substrates for hSULT1E1 and hSULT1A1*1, although the relative catalytic efficiencies varied with both the substrate and the enzyme. These results may assist in future elucidation of cell- and tissue-specific effects of tamoxifen and its metabolites.


Assuntos
Antineoplásicos Hormonais/farmacologia , Arilsulfotransferase/antagonistas & inibidores , Drogas em Investigação/farmacologia , Inibidores Enzimáticos/farmacologia , Estradiol/metabolismo , Sulfotransferases/antagonistas & inibidores , Tamoxifeno/análogos & derivados , Antineoplásicos Hormonais/metabolismo , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Drogas em Investigação/metabolismo , Inibidores Enzimáticos/metabolismo , Humanos , Cinética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Sulfotransferases/genética , Sulfotransferases/metabolismo , Tamoxifeno/metabolismo , Tamoxifeno/farmacologia
8.
Environ Mol Mutagen ; 56(4): 404-11, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25243916

RESUMO

1-Methylpyrene (1-MP) is a widespread pollutant that is carcinogenic in animals following metabolic activation. Previous studies have shown that benzylic hydroxylation of 1-MP, catalyzed by multiple CYP isoforms, gives rise to 1-hydroxymethylpyrene (1-HMP), which becomes bioreactive following further metabolism by various sulfotransferase (SULT) isoforms. However, the mutagenic and chromosome damaging effects of 1-MP and 1-HMP in mammalian cells have not been investigated. In this study a Chinese hamster V79-derived cell line expressing both human CYP2E1 and human SULT1A1 was used to investigate the ability of 1-MP and 1-HMP to induce cytotoxicity (using the CCK-8 assay), micronuclei and Hprt gene mutations. The role of each enzyme was investigated through co-exposure in the presence of an enzyme inhibitor. We found that at concentrations of 0.5-4 µM and 5-20 µM, under conditions where no reduction in cell viability/growth occurred, 1-HMP and 1-MP induced micronuclei in V79-hCYP2E1-hSULT1A1 cells in a concentration-dependent manner; however, both compounds were inactive in V79 cells. Similarly, they both caused an increase in Hprt mutant frequency in V79-hCYP2E1-hSULT1A1 cells in these concentration ranges, with 1-MP impairing cell viability/growth at 10 µM and above in the mutagenicity assay. The compounds were again both inactive in V79 cells. The effects of 1-HMP in V79-hCYP2E1-hSULT1A1 cells were blocked or reduced by addition of pentachlorophenol (PCP), a SULT1 inhibitor; the genotoxicity of 1-MP was significantly reduced by either 1-aminobenotrazole, a CYP2E1 inhibitor, or PCP. The results suggest that human CYP2E1 and SULT1A1 cooperate to activate 1-MP and cause genotoxicity in mammalian cells.


Assuntos
Arilsulfotransferase/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Testes de Mutagenicidade/métodos , Pirenos/toxicidade , Animais , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Linhagem Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Sistema Enzimático do Citocromo P-450/genética , Família 2 do Citocromo P450 , Relação Dose-Resposta a Droga , Humanos , Hipoxantina Fosforribosiltransferase/genética , Testes para Micronúcleos , Mutagênicos/toxicidade , Taxa de Mutação , Pentaclorofenol/farmacologia
9.
Horm Mol Biol Clin Investig ; 20(3): 81-90, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25418972

RESUMO

Drug-drug interactions (DDI) with oral contraceptives containing 17α-ethinylestradiol (EE2) have been well characterized with regard to interactions with phase I drug metaolizing enzymes; however, DDI with EE2 and phase II enzymes have not been as thoroughly addressed. Our laboratory recently reported that in vitro EE2 potently inhibits human cytosolic sulfotransferase (SULT) 1A1 while EE2 was not sulfated until micromolar concentrations. Molecular docking studies demonstrated that Tyr169 and isoleucine 89 (Ile89) may play a role in the inhibitory and/or catalytic positioning of EE2 within the active site of SULT1A1. Therefore, the current study focused on determining the role of Ile89 in the inhibition of SULT1A1 utilizing site-directed mutagenesis. Ile89 was mutated to an alanine and the effect of the mutation was characterized using kinetic and binding assays. SULT1A1-Ile89Ala was found to have a Km for EE2 that was 11-fold greater than wild-type enzyme. A decreased affinity (Kd) of EE2 for SULT1A1-Ile89Ala was apparently responsible for the increase in Km, and also resulted in the loss of the potent inhibition. Molecular modeling was used in an attempt to determine the atomic level changes in binding of EE2 to SULT1A1-Ile89Ala. However, analysis of the effect of the single Ile89 mutation on both the open and closed homology models was not consistent with the docking and kinetic results. Overall, the mechanism of inhibition of EE2 for SULT1A1 is apparently the result of interactions of Ile89 with EE2 holding it in a potent inhibitory conformation, and mutation of the Ile89 significantly decreases the inhibition.


Assuntos
Arilsulfotransferase/antagonistas & inibidores , Estrogênios/química , Etinilestradiol/química , Isoleucina/química , Substituição de Aminoácidos , Arilsulfotransferase/química , Arilsulfotransferase/genética , Citosol/enzimologia , Humanos , Simulação de Acoplamento Molecular , Mutação , Tirosina/química
10.
J Biol Chem ; 288(48): 34494-501, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24129576

RESUMO

Predicting enzymatic behavior in silico is an integral part of our efforts to understand biology. Hundreds of millions of compounds lie in targeted in silico libraries waiting for their metabolic potential to be discovered. In silico "enzymes" capable of accurately determining whether compounds can inhibit or react is often the missing piece in this endeavor. This problem has now been solved for the cytosolic sulfotransferases (SULTs). SULTs regulate the bioactivities of thousands of compounds--endogenous metabolites, drugs and other xenobiotics--by transferring the sulfuryl moiety (SO3) from 3'-phosphoadenosine 5'-phosphosulfate to the hydroxyls and primary amines of these acceptors. SULT1A1 and 2A1 catalyze the majority of sulfation that occurs during human Phase II metabolism. Here, recent insights into the structure and dynamics of SULT binding and reactivity are incorporated into in silico models of 1A1 and 2A1 that are used to identify substrates and inhibitors in a structurally diverse set of 1,455 high value compounds: the FDA-approved small molecule drugs. The SULT1A1 models predict 76 substrates. Of these, 53 were known substrates. Of the remaining 23, 21 were tested, and all were sulfated. The SULT2A1 models predict 22 substrates, 14 of which are known substrates. Of the remaining 8, 4 were tested, and all are substrates. The models proved to be 100% accurate in identifying substrates and made no false predictions at Kd thresholds of 100 µM. In total, 23 "new" drug substrates were identified, and new linkages to drug inhibitors are predicted. It now appears to be possible to accurately predict Phase II sulfonation in silico.


Assuntos
Arilsulfotransferase/química , Simulação por Computador , Bibliotecas de Moléculas Pequenas/química , Sulfotransferases/química , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Citosol/enzimologia , Inibidores Enzimáticos/química , Humanos , Desintoxicação Metabólica Fase II , Simulação de Dinâmica Molecular , Ligação Proteica , Especificidade por Substrato , Sulfatos/química , Sulfatos/metabolismo , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/genética
11.
J Biol Chem ; 288(48): 34364-74, 2013 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-24136195

RESUMO

Dopamine neurotoxicity is associated with several neurodegenerative diseases, and neurons utilize several mechanisms, including uptake and metabolism, to protect them from injury. Metabolism of dopamine involves three enzymes: monoamine oxidase, catechol O-methyltransferase, and sulfotransferase. In primates but not lower order animals, a sulfotransferase (SULT1A3) is present that can rapidly metabolize dopamine to dopamine sulfate. Here, we show that SULT1A3 and a closely related protein SULT1A1 are highly inducible by dopamine. This involves activation of the D1 and NMDA receptors. Both ERK1/2 phosphorylation and calcineurin activation are required for induction. Pharmacological agents that inhibited induction or siRNA targeting SULT1A3 significantly increased the susceptibility of cells to dopamine toxicity. Taken together, these results show that dopamine can induce its own metabolism and protect neuron-like cells from damage, suggesting that SULT1A3 activity may be a risk factor for dopamine-dependent neurodegenerative diseases.


Assuntos
Arilsulfotransferase/metabolismo , Dopamina/metabolismo , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Benzazepinas/farmacologia , Calcineurina/metabolismo , Linhagem Celular , Dopamina/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/genética , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Fosforilação , RNA Interferente Pequeno/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/genética , Fatores de Risco , Transdução de Sinais
12.
Biofactors ; 39(6): 644-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24038617

RESUMO

Hydroxycinnamic acids and flavonoids are dietary phenolic antioxidants that are abundant in our diet. Hydroxycinnamic acids are highly sulfated in vivo, and sulfotransferases (SULTs), in particular SULT1A1, play a major role in their metabolism. Flavonoids are potent inhibitors of human SULTs. In this study, the potential metabolic interaction between dietary hydroxycinnamic acids and flavonoids was investigated. Flavonoids, such as luteolin, quercetin, daidzein, and genistein, are identified as potent inhibitors of hydroxycinnamic acid sulfation in human liver S9 homogenate with IC50 values <1 µM. The inhibitory activity was less potent in the human intestinal S9 homogenate. We also demonstrate that quercetin conjugates found in vivo (quercetin-3-O-glucuronide, quercetin-7-O-glucuronide, and quercetin-3'-O-sulfate) moderately inhibited the sulfation of hydroxycinnamic acids in human liver S9. In an intact cellular system, human HepG2 cells, caffeic acid and ferulic acid sulfation was inhibited by luteolin and quercetin (IC50 : 1.6-3.9 µM). Quercetin-3'-O-sulfate weakly inhibited sulfation. Quercetin glucuronides, limited by their low cellular uptake, were ineffective. These data suggest that the inhibition of SULTs by flavonoids and in vivo flavonoid conjugates may modify the bioavailability of dietary hydroxycinnamic acids by suppressing their conversion to sulfated metabolites.


Assuntos
Arilsulfotransferase/antagonistas & inibidores , Ácidos Cumáricos/metabolismo , Quercetina/farmacologia , Arilsulfotransferase/metabolismo , Ácidos Cafeicos/metabolismo , Genisteína/farmacologia , Células Hep G2 , Humanos , Concentração Inibidora 50 , Intestinos/enzimologia , Isoflavonas/farmacologia , Fígado/enzimologia , Luteolina/farmacologia
13.
Mutagenesis ; 28(5): 609-19, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23894158

RESUMO

The benzylic alcohols 1- and 2-hydroxy-3-methylcholanthrene (OH-MC) are major primary metabolites of the carcinogen 3-methylcholanthrene (MC). We investigated them for mutagenicity in TA1538-derived Salmonella typhimurium strains expressing mammalian sulphotransferases (SULTs). 1-OH-MC was efficiently activated by human (h) SULT1B1 (2400 revertants/nmol), weakly activated by hSULT1C3 and hSULT2A1 (2-9 revertants/nmol), but not activated by the other hSULTs studied (1A2, 1A3, 1C2 and 1E1). Mouse, rat and dog SULT1B1 activated 1-OH-MC (8-100 revertants/nmol) with much lower efficiency than their human orthologue. The other isomer, 2-OH-MC, was activated to a potent mutagen by hSULT1A1 (4000-5400 revertants/nmol), weakly activated by hSULT1A2 or hSULT2A1 (1-12 revertants/nmol), but not activated by the other hSULTs. In contrast to their human orthologue, mouse, rat and dog SULT1A1 did not appreciably activate 2-OH-MC (<1 to 6 revertants/nmol), either. Instead, mouse and rat SULT1B1, unlike their human and canine orthologues, demonstrated some activation of 2-OH-MC (15-100 revertants/nmol). Docking analyses indicated that 1- and 2-OH-MC might bind to the active site of hSULT1A1 and hSULT1B1, but only for (S)-2-OH-MC/hSULT1A1 and (R)-1-OH-MC/hSULT1B1 with an orientation suitable for catalysis. Indeed, 1- and 2-OH-MC were potent inhibitors of the hSULT1A1-mediated sulphation of acetaminophen [concentration inhibiting the enzyme activity by 50% (IC50) 15 and 13nM, respectively]. This inhibition was weak with mouse, rat and dog SULT1A1 (IC50 ≥ 4 µM). Inhibition of the SULT1B1 enzymes was moderate, strongest for 1-OH-MC/hSULT1B1. In conclusion, this study provides examples for high selectivity of bioactivation of promutagens by an individual form of human SULT and for pronounced differences in activation capacity between orthologous SULTs from different mammalian species. These characteristics make the detection and evaluation of such mutagens extremely difficult, in particular as the critical form may even differ for positional isomers, such as 1- and 2-OH-MC. Moreover, the species-dependent differences will complicate the verification of in vitro results in animal studies.


Assuntos
Metilcolantreno/análogos & derivados , Mutagênicos/farmacocinética , Salmonella typhimurium/genética , Sulfotransferases/metabolismo , Acetaminofen/química , Acetaminofen/metabolismo , Animais , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Cães , Inibidores Enzimáticos/farmacologia , Humanos , Isomerismo , Metilcolantreno/química , Metilcolantreno/farmacocinética , Metilcolantreno/toxicidade , Camundongos , Simulação de Acoplamento Molecular , Testes de Mutagenicidade , Mutagênicos/metabolismo , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/enzimologia , Especificidade da Espécie , Sulfotransferases/química , Sulfotransferases/genética
14.
J Appl Toxicol ; 32(10): 751-5, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22678655

RESUMO

Sudden cardiac death is a significant health issue, causing millions of deaths worldwide annually. Studies have found that the likelihood of such death is higher in winter. Further studies identified that the highest likelihood occurs on Christmas Day and New Years Day, but not the interim period. Thanksgiving, Independence Day and the Islamic holiday Eid Al-Fitr also show significant increases in the rate of cardiac events or death. A number of mechanisms have been proposed, but none have satisfactorily explained the evidence. This article reviews the data supporting the existence of a holiday cardiac death phenomenon, the involvement of catecholamines and the normal modes of human catecholamine deactivation. Further evidence is reviewed that supports a hypothesized mechanism whereby critical SULT1A catecholamine deactivation enzymes can in some patients be inhibited by naturally-occurring phenols and polyphenols in foods and alcohols. If deactivation is inhibited by holiday consumption excesses, holiday stress or excitement could lead to a buildup of catecholamines that can cause fatal arrhythmias. Awareness of this mechanism could reduce deaths, both through doctor/patient education leading to a moderation in consumption and through the potential identification of patients with a predisposition to SULT1A inhibition. This hypothesis also raises parallels between sudden cardiac death in adults and Sudden Infant Death Syndrome (SIDS). The possible involvement of SULT1A inhibition in SIDS is discussed.


Assuntos
Consumo de Bebidas Alcoólicas/efeitos adversos , Arilsulfotransferase/metabolismo , Morte Súbita Cardíaca/etiologia , Dieta/efeitos adversos , Férias e Feriados , Adulto , Idoso , Alelos , Animais , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Catecolaminas/metabolismo , Suscetibilidade a Doenças , Humanos , Lactente , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Polimorfismo de Nucleotídeo Único , Morte Súbita do Lactente/etiologia , Morte Súbita do Lactente/genética , Sulfotransferases/antagonistas & inibidores , Sulfotransferases/genética , Sulfotransferases/metabolismo
15.
Drug Metab Dispos ; 40(8): 1588-95, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593037

RESUMO

Sulfotransferase (SULT) 1A1 is the major drug/xenobiotic-conjugating SULT isoform in human liver because of its broad substrate reactivity and high expression level. SULT1A1 sulfates estrogens with low micromolar K(m) values consistent with its affinity for sulfation of many small phenolic compounds. Binding studies showed the unexpected ability of 17α-ethinylestradiol (EE2) to bind and inhibit SULT1A1 activity toward p-nitrophenol and ß-naphthol at low nanomolar concentrations, whereas EE2 was not sulfated until significantly higher concentrations were reached. EE2 had a K(i) of 10 nM for inhibiting p-nitrophenol and ß-naphthol sulfation and inhibited 17ß-estradiol (E2) sulfation in intact human MCF-7 breast cancer cells with a K(i) of 19 nM. In contrast, the K(m) for EE2 sulfation by SULT1A1 was 700 nM. The K(d) for EE2 binding of pure SULT1A1 was 0.5 ± 0.15 µM; however, the K(d) for EE2 binding to the SULT1A1-PAP complex was >100-fold lower (4.3 ± 1.7 nM). The K(d) for E2 binding to SULT1A1 changed from 2.3 ± 0.9 to 1.2 ± 0.56 µM in the presence of PAP. Docking studies with E2 indicate that E2 binds in a competent orientation in the resolved structure of SULT1A1 in the both presence and absence of 3'-phosphoadenosine 5'-phosphosulfate (PAPS). However, EE2 binds in a catalytically competent orientation in the absence of PAPS but in a noncompetent orientation via formation of a charge interaction with Tyr108 if PAPS is bound first. In conclusion, EE2 is a potent inhibitor, but not a substrate, of SULT1A1 at low nanomolar concentrations, indicating the possibility of drug-drug interactions during contraceptive therapy.


Assuntos
Arilsulfotransferase/antagonistas & inibidores , Etinilestradiol/farmacologia , Fosfoadenosina Fosfossulfato/farmacologia , Humanos
16.
Food Chem Toxicol ; 50(6): 2243-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22433984

RESUMO

SULT1A enzymes protect humans from catecholamines, but natural substances in many foods have been found to inhibit these enzymes in vitro. Given the hormonal roles of catecholamines, any in vivo SULT1A inhibition could have serious consequences. This paper uses a re-analysis of published data to confirm that SULT1A inhibitors have effect in vivo in at least some patients. Nineteen studies are cited that show ingestion of SULT1A inhibitors leading to catecholamine increases, blood pressure changes, migraine headaches, or atrial fibrillation. SULT1A inhibition does not create the catecholamines, but prevents normal catecholamine deactivation. Susceptible patients probably have lower-activity SULT1A alleles. The paper discusses new hypotheses that SULT1A inhibition can cause "holiday heart" arrhythmias and type 2 diabetes in susceptible patients. Subgroup analysis based on SULT1A alleles, and addition of a catecholamine source, should improve the consistency of results from tests of SULT1A inhibitors. SULT1A inhibition may be a key contributor to cheese-induced migraines (via annatto), false positives in metanephrine testing, and the cardiovascular impacts of recreational alcohols.


Assuntos
Arilsulfotransferase/antagonistas & inibidores , Bebidas/toxicidade , Catecolaminas/sangue , Citrus sinensis/toxicidade , Vinho/toxicidade , Bebidas Alcoólicas/toxicidade , Fibrilação Atrial/induzido quimicamente , Pressão Sanguínea/efeitos dos fármacos , Café , Dieta , Dopamina/sangue , Epinefrina/sangue , Frutas/química , Humanos , Transtornos de Enxaqueca/induzido quimicamente , Norepinefrina/sangue , Polifenóis/química , Polifenóis/farmacologia , Quercetina/farmacologia , Chá/química
17.
Toxicol In Vitro ; 25(2): 500-4, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21144891

RESUMO

Intake of high doses of vitamin C has known to modulate sulfoconjugation of drugs in the intestine, but the underlying mechanisms for this effect remain to be elucidated. In the present study, we investigated the effects of vitamin C (l-ascorbic acid (AA)) on sulfation of 1-naphthol using Caco-2 cells, a model of human intestinal cells. We found that high dose of AA inhibited the accumulation of 1-naphthyl sulfate in Caco-2 culture medium within 24h in a dose-dependent manner (IC(50)=42 mM). Dehydroascorbic acid (DA), an oxidized form of AA, showed no inhibition. AA did not inhibit the in vitro sulfotransferase (SULT) activity toward 1-naphthol, whereas it reduced the expression of genes belonging to SULT1A family, SULT1A1 and SULT1A3. DA showed no effect on SULT1A gene expression. Consistent with the reduction in gene expression, AA reduced the cytosolic SULT activity towards 1-naphthol in the AA-treated Caco-2 cells. In addition, cAMP exerted an additive effect on AA-mediated repression of SULT1A gene expression. Our results suggest that megadose AA suppresses sulfoconjugation in the intestine mainly by downregulating the expression of SULT1A genes.


Assuntos
Ácido Ascórbico/farmacologia , Mucosa Intestinal/metabolismo , Naftóis/metabolismo , Ésteres do Ácido Sulfúrico/metabolismo , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Células CACO-2 , AMP Cíclico/farmacologia , Humanos , Mucosa Intestinal/efeitos dos fármacos
18.
Arch Biochem Biophys ; 506(2): 137-41, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21111704

RESUMO

Cytosolic sulfotransferase (SULT)-catalyzed sulfation regulates the activity of bio-signaling molecules and aids in metabolizing hydroxyl-containing xenobiotics. The sulfuryl donor for the SULT reaction is adenosine 3'-phosphate 5'-phosphosulfate (PAPS), while products are adenosine 3',5'-diphosphate (PAP) and a sulfated alcohol. Human phenol sulfotransferase (SULT1A1) is one of the major detoxifying enzymes for phenolic xenobiotics. The mechanism of SULT1A1-catalyzed sulfation of PAP by pNPS was investigated. PAP was sulfated by para-nitrophenyl sulfate (pNPS) in a concentration-dependent manner. 2-Naphthol inhibited sulfation of PAP, competing with pNPS, while phenol activated the sulfation reaction. At saturating PAP, a ping pong kinetic mechanism is observed with pNPS and phenol as substrates, consistent with phenol intercepting the E-PAPS complex prior to dissociation of PAPS. At high concentrations, phenol competes with pNPS, consistent with formation of the E-PAP-phenol dead-end complex. Data are consistent with the previously reported mechanism for sulfation of 2-naphthol by PAPS, and its activation by pNPS. Overall, data are consistent with release of PAP from E-PAP and PAPS from E-PAPS contributing to rate-limitation in both reaction directions.


Assuntos
Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Difosfato de Adenosina/metabolismo , Arilsulfotransferase/antagonistas & inibidores , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Técnicas In Vitro , Cinética , Modelos Biológicos , Naftóis/farmacologia , Nitrobenzenos/metabolismo , Fenol/farmacologia , Fosfoadenosina Fosfossulfato/metabolismo , Proteínas Recombinantes de Fusão/antagonistas & inibidores , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato
19.
Biochem Biophys Res Commun ; 396(2): 429-34, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20417180

RESUMO

The cytosolic sulfotransferases (SULTs) in vertebrates catalyze the sulfonation of endogenous thyroid/steroid hormones and catecholamine neurotransmitters, as well as a variety of xenobiotics, using 3'-phosphoadenosine 5'-phosphosulfate (PAPS) as the sulfonate donor. In this study, we determined the structures of SULT1A2 and an allozyme of SULT1A1, SULT1A1 *3, bound with 3'-phosphoadenosine 5'-phosphate (PAP), at 2.4 and 2.3A resolution, respectively. The conformational differences between the two structures revealed a plastic substrate-binding pocket with two channels and a switch-like substrate selectivity residue Phe247, providing clearly a structural basis for the substrate inhibition. In SULT1A2, Tyr149 extends approximately 2.1A further to the inside of the substrate-binding pocket, compared with the corresponding His149 residue in SULT1A1 *3. Site-directed mutagenesis study showed that, compared with the wild-type SULT1A2, mutant Tyr149Phe SULT1A2 exhibited a 40 times higher K(m) and two times lower V(max) with p-nitrophenol as substrate. These latter data imply a significant role of Tyr149 in the catalytic mechanism of SULT1A2.


Assuntos
Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/química , Arilsulfotransferase/genética , Catálise , Cristalografia por Raios X , Humanos , Mutagênese Sítio-Dirigida , Mutação , Nitrofenóis/química , Fosfoadenosina Fosfossulfato/química , Conformação Proteica , Especificidade por Substrato , Tirosina/química , Tirosina/genética
20.
Environ Mol Mutagen ; 51(1): 23-30, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19484729

RESUMO

Genetic engineering of target cells for investigating the genotoxicity associated with specific xenobiotic-metabolizing enzymes is useful for elucidating metabolic activation and inactivation processes. We constructed a V79-derived cell line expressing both human cytochrome P450 (CYP) 2E1 and human sulfotransferase (SULT) 1A1. We previously reported that this cell line (V79-hCYP2E1-hSULT1A1) efficiently activates various important pro-genotoxicants. Here we present data on the expression level and stability of the heterologous enzymes, measured by immunoblotting, enzyme activities, and mutagenic responses to CYP2E1- and SULT1A1-dependent promutagens. Unexpectedly, these cells demonstrated greatly elevated spontaneous gene mutation frequencies (determined at the Hprt locus), and elevated frequencies of sister chromatid exchange, as compared with control V79 cells and V79-derived lines engineered for other enzymes. Therefore, V79-hCYP2E1-hSULT1A1 cells require regular cleansing in aminopterin-containing medium when used for Hprt gene mutation assays. In a 4-week time course without such selection, V79-hCYP2E1-hSULT1A1 demonstrated a progressive increase in the spontaneous mutant frequency from 2.9 to 155 x 10(-6). This phenomenon was moderately, strongly, and completely prohibited in the presence of CYP2E1 inhibitor 1-aminobenzotriazole, SULT1A1 inhibitor pentachlorophenol and both in combination, respectively. This protection indicates that the enhanced spontaneous mutagenicity involves the activity of the expressed enzymes rather than being caused by an accidental genetic alteration that might have occurred during transfection. We postulate that human CYP2E1 and SULT1A1 activate an endogenous cellular molecule or a medium component to become mutagenic. It will be challenging to identify this compound and to see whether it is involved in spontaneous mutagenesis and carcinogenesis in vivo.


Assuntos
Arilsulfotransferase/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Mutagênese/genética , Proteínas Recombinantes de Fusão/metabolismo , Animais , Arilsulfotransferase/antagonistas & inibidores , Arilsulfotransferase/genética , Células CHO , Linhagem Celular , Células Cultivadas , Cricetinae , Cricetulus , Citocromo P-450 CYP2E1/genética , Inibidores do Citocromo P-450 CYP2E1 , Inibidores Enzimáticos/farmacologia , Engenharia Genética , Vetores Genéticos , Humanos , Mutagênese/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Triazóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...