Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 13129, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162941

RESUMO

Sulfotransferases (SULTs) are phase II drug-metabolizing enzymes catalyzing the sulfoconjugation from the co-factor 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to a substrate. It has been previously suggested that a considerable shift of SULT structure caused by PAPS binding could control the capability of SULT to bind large substrates. We employed molecular dynamics (MD) simulations and the recently developed approach of MD with excited normal modes (MDeNM) to elucidate molecular mechanisms guiding the recognition of diverse substrates and inhibitors by SULT1A1. MDeNM allowed exploring an extended conformational space of PAPS-bound SULT1A1, which has not been achieved up to now by using classical MD. The generated ensembles combined with docking of 132 SULT1A1 ligands shed new light on substrate and inhibitor binding mechanisms. Unexpectedly, our simulations and analyses on binding of the substrates estradiol and fulvestrant demonstrated that large conformational changes of the PAPS-bound SULT1A1 could occur independently of the co-factor movements that could be sufficient to accommodate large substrates as fulvestrant. Such structural displacements detected by the MDeNM simulations in the presence of the co-factor suggest that a wider range of drugs could be recognized by PAPS-bound SULT1A1 and highlight the utility of including MDeNM in protein-ligand interactions studies where major rearrangements are expected.


Assuntos
Arilsulfotransferase/química , Simulação de Dinâmica Molecular , Sítios de Ligação , Humanos , Fosfoadenosina Fosfossulfato/metabolismo , Ligação Proteica , Especificidade por Substrato
2.
Arch Biochem Biophys ; 695: 108621, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33049293

RESUMO

Structural determinations of members of the sulfotransferase (SULT) family suggest a direct interaction between a conserved tryptophanyl side chain and bound 3'-phosphoadenosine-5'-phosphate (PAP). We have prepared and purified mutants of the bovine SULT1A1, a very conserved homolog to the human SULT1A1, in which tryptophanyl-53 was sequentially trimmed to tyrosine, leucine, and alanine. Differential scanning fluorimetry indicated structural stabilities of the mutant proteins comparable to the wild type SULT1A1; however, less thermal stabilizations by PAP plus pentachlorophenol were observed with the mutants, suggesting weakened ligand binding. Protein fluorescence of the wild type enzyme decreased 6.5% upon binding PAP, whereas no changes occurred with the mutant enzymes. This reveals that W53, or its positional counterpart, has been the source of emission intensity changes used in previous investigations of other SULTs. Fluorescence resonance energy transfer from excited tryptophans to bound 7-hydroxycoumarin, as induced by PAP, indicated weakened binding of ligands to the mutant SULTs. This was also encountered and quantified in initial rate kinetic analyses. Ablation of the PAPS adenine-to-W53 ring interaction, shown by the W53A mutant enzyme, resulted in a 6.4-fold increase in KPAPS and a 92% decrease in kcat/KPAPS. Measured KPAPS values reveal the W53 indole ring contribution to PAPS binding to be 1.1 kcal/mol (4.6 kJ/mol). These results verify the structurally-inferred role for the π-π stacking interaction between PAP(S) and the conserved tryptophanyl residue in SULT1A1 and other members of the SULT family.


Assuntos
Arilsulfotransferase/química , Substituição de Aminoácidos , Arilsulfotransferase/genética , Sítios de Ligação , Catálise , Transferência Ressonante de Energia de Fluorescência , Humanos , Mutação de Sentido Incorreto , Triptofano/química , Triptofano/genética
3.
J Agric Food Chem ; 68(40): 11197-11206, 2020 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-32910657

RESUMO

Authentic standards of food flavonoids are important for human metabolic studies. Their isolation from biological materials is impracticable; however, they can be prepared in vitro. Twelve sulfated metabolites of luteolin, myricetin, and ampelopsin were obtained with arylsulfotransferase from Desulfitobacterium hafniense and fully characterized by high-performance liquid chromatography, MS, and NMR. The compounds were tested for their ability to scavenge 1,1-diphenyl-2-picrylhydrazyl, 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulphonic acid), and N,N-dimethyl-p-phenylenediamine radicals, to reduce ferric ions and Folin-Ciocalteu reagent, and to inhibit tert-butyl hydroperoxide-induced lipid peroxidation of rat liver microsomes. The activity differed considerably even between monosulfate isomers. The parent compounds and myricetin-3'-O-sulfate were the most active while other compounds displayed significantly lower activity, particularly luteolin sulfates. No mutagenic activity of the parent compounds and their main metabolites was observed; only myricetin showed minor pro-mutagenicity. The prepared sulfated metabolites are now available as authentic standards for future in vitro and in vivo metabolic studies.


Assuntos
Arilsulfotransferase/química , Proteínas de Bactérias/química , Desulfitobacterium/enzimologia , Flavonoides/química , Luteolina/química , Sulfatos/química , Animais , Antioxidantes/química , Antioxidantes/farmacologia , Biocatálise , Fenômenos Biofísicos , Flavonoides/metabolismo , Flavonoides/farmacologia , Isomerismo , Peroxidação de Lipídeos/efeitos dos fármacos , Luteolina/metabolismo , Luteolina/farmacologia , Masculino , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Estrutura Molecular , Ratos , Sulfatos/metabolismo
4.
Int J Mol Sci ; 20(24)2019 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-31835852

RESUMO

Sulfotransferase 1A1 (SULT1A1) is responsible for catalyzing various types of endogenous and exogenous compounds. Accumulating data indicates that the polymorphism rs9282861 (R213H) is responsible for inefficient enzymatic activity and associated with cancer progression. To characterize the detailed functional consequences of this mutation behind the loss-of-function of SULT1A1, the present study deployed molecular dynamics simulation to get insights into changes in the conformation and binding energy. The dynamics scenario of SULT1A1 in both wild and mutated types as well as with and without ligand showed that R213H induced local conformational changes, especially in the substrate-binding loop rather than impairing overall stability of the protein structure. The higher conformational changes were observed in the loop3 (residues, 235-263), turning loop conformation to A-helix and B-bridge, which ultimately disrupted the plasticity of the active site. This alteration reduced the binding site volume and hydrophobicity to decrease the binding affinity of the enzyme to substrates, which was highlighted by the MM-PBSA binding energy analysis. These findings highlight the key insights of structural consequences caused by R213H mutation, which would enrich the understanding regarding the role of SULT1A1 mutation in cancer development and also xenobiotics management to individuals in the different treatment stages.


Assuntos
Arilsulfotransferase/genética , Polimorfismo de Nucleotídeo Único/genética , Arilsulfotransferase/química , Sítios de Ligação , Simulação por Computador , Estabilidade Enzimática , Humanos , Ligantes , Modelos Moleculares , Mutação/genética , Análise de Componente Principal , Estrutura Secundária de Proteína , Especificidade por Substrato , Termodinâmica
5.
J Agric Food Chem ; 67(26): 7281-7288, 2019 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-31198027

RESUMO

Potential metabolites of bioactive compounds are important for their biological activities and as authentic standards for metabolic studies. The phenolic compounds contained in olive oil are an important part of the human diet, and therefore their potential metabolites are of utmost interest. We developed a convenient, scalable, one-pot chemoenzymatic method using the arylsulfotransferase from Desulfitobacterium hafniense for the sulfation of the natural olive oil phenols tyrosol, hydroxytyrosol, and of their monoacetylated derivatives. Respective monosulfated (tentative) metabolites were fully structurally characterized using LC-MS, NMR, and HRMS. In addition, Folin-Ciocalteu reduction, 1,1-diphenyl-2-picrylhydrazyl radical scavenging, and antilipoperoxidant activity in rat liver microsomes damaged by tert-butylhydroperoxide were measured and compared to the parent compounds. As expected, the sulfation diminished the radical scavenging properties of the prepared compounds. These compounds will serve as authentic standards of phase II metabolites.


Assuntos
Arilsulfotransferase/química , Proteínas de Bactérias/química , Sequestradores de Radicais Livres/química , Álcool Feniletílico/análogos & derivados , Acetilação , Biocatálise , Cromatografia Líquida de Alta Pressão , Desulfitobacterium/enzimologia , Sequestradores de Radicais Livres/síntese química , Espectrometria de Massas , Estrutura Molecular , Azeite de Oliva/química , Fenóis/química , Álcool Feniletílico/síntese química , Álcool Feniletílico/química , Sulfatos/química
6.
Pharmacogenet Genomics ; 29(5): 99-105, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31145702

RESUMO

OBJECTIVES: Phenylephrine and salbutamol are drugs that are used widely to treat diseases/disorders, such as nasal congestion, hypotension, and asthma, in individuals of different age groups. Human cytosolic sulfotransferase (SULT) SULT1A3 has been shown to be critically involved in the metabolism of these therapeutic agents. This study was carried out to investigate the effects of single nucleotide polymorphisms of human SULT1A3 and SULT1A4 genes on the sulfation of phenylephrine and salbutamol by SULT1A3 allozymes. MATERIALS AND METHODS: Wild-type and SULT1A3 allozymes, prepared previously by site-directed mutagenesis in conjunction with bacterial expression and affinity purification, were analyzed for sulfating activity using an established assay procedure. RESULTS: Purified SULT1A3 allozymes, in comparison with the wild-type enzyme, showed differential sulfating activities toward phenylephrine and salbutamol. Kinetic studies showed further significant variations in their substrate-binding affinity and catalytic activity toward phenylephrine and salbutamol. CONCLUSION: The results obtained showed clearly the differential enzymatic characteristics of SULT1A3 allozymes in mediating the sulfation of phenylephrine and salbutamol. This information may contribute toward a better understanding of the pharmacokinetics of these two drugs in individuals with distinct SULT1A3 and/or SULT1A4 genotypes.


Assuntos
Albuterol/metabolismo , Arilsulfotransferase/genética , Fenilefrina/metabolismo , Sulfotransferases/genética , Albuterol/uso terapêutico , Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Asma/tratamento farmacológico , Asma/genética , Genótipo , Humanos , Hipotensão/tratamento farmacológico , Hipotensão/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Mutagênese Sítio-Dirigida , Fenilefrina/uso terapêutico , Polimorfismo de Nucleotídeo Único/genética , Sulfatos/metabolismo , Sulfotransferases/química , Sulfotransferases/metabolismo
7.
Electrophoresis ; 40(18-19): 2271-2276, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30882918

RESUMO

Sulfotransferases are categorized as phase II metabolic enzymes. Human sulfotransferase 1A1 (SULT1A1) is involved in the sulfonation of xenobiotics with aid from the cofactor 3'-phosphoadenosine-5'-phosphosulfate that acts as a sulfonate donor. In this study, we have attempted to immobilize SULT1A1 on magnetic microparticles (MMs). Different functionalized MMs were used to immobilize SULT1A1 and their enzyme activity was compared to the control (enzyme in solution). Paracetamol was used as model substrate. Separation of paracetamol and paracetamol sulfate by CE-UV was optimized and validated. MMs with epoxy based immobilization of SULT1A1 showed better enzyme activity. Hence, they were tested for repeated usage to allow their implementation for the development of a CE immobilized micro enzyme reactor.


Assuntos
Arilsulfotransferase , Eletroforese Capilar/métodos , Enzimas Imobilizadas , Imãs , Acetaminofen/análogos & derivados , Acetaminofen/análise , Acetaminofen/metabolismo , Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Limite de Detecção , Modelos Lineares , Microesferas , Reprodutibilidade dos Testes
8.
Appl Microbiol Biotechnol ; 103(9): 3761-3771, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30830250

RESUMO

Sulfation is an important way for detoxifying xenobiotics and endobiotics including catechols. Enzymatic sulfation occurs usually with high chemo- and/or regioselectivity under mild reaction conditions. In this study, a two-step p-NPS-4-AAP screening system for laboratory evolution of aryl sulfotransferase B (ASTB) was developed in 96-well microtiter plates to improve the sulfate transfer efficiency toward catechols. Increased transfer efficiency and improved sulfation stoichiometry are achieved through the two-step screening procedure in a one-pot reaction. In the first step, the p-NPS assay is used (detection of the colorimetric by-product, p-nitrophenol) to determine the apparent ASTB activity. The sulfated product, 3-chlorocatechol-1-monosulfate, is quantified by the 4-aminoantipyrine (4-AAP) assay in the second step. Comparison of product formation to p-NPS consumption ensures successful directed evolution campaigns of ASTB. Optimization yielded a coefficient of variation below 15% for the two-step screening system (p-NPS-4-AAP). In total, 1760 clones from an ASTB-SeSaM library were screened toward the improved sulfation activity of 3-chlorocatechol. The turnover number (kcat = 41 ± 2 s-1) and catalytic efficiency (kcat/KM = 0.41 µM-1 s-1) of the final variant ASTB-M5 were improved 2.4- and 2.3-fold compared with ASTB-WT. HPLC analysis confirmed the improved sulfate stoichiometry of ASTB-M5 with a conversion of 58% (ASTB-WT 29%; two-fold improvement). Mass spectrometry (MS) and nuclear magnetic resonance spectroscopy (NMR) confirmed the chemo- and regioselectivity, which yielded exclusively 3-chlorocatechol-1-monosulfate. For all five additionally investigated catechols, the variant ASTB-M5 achieved an improved kcat value of up to 4.5-fold and sulfate transfer efficiency was also increased (up to 2.3-fold).


Assuntos
Arilsulfotransferase/genética , Proteínas de Bactérias/genética , Catecóis/metabolismo , Desulfitobacterium/enzimologia , Sulfatos/metabolismo , Ampirona/química , Ampirona/metabolismo , Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Catecóis/química , Desulfitobacterium/química , Desulfitobacterium/genética , Evolução Molecular Direcionada , Cinética , Espectroscopia de Ressonância Magnética , Especificidade por Substrato , Sulfatos/química
9.
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
10.
J Biol Chem ; 294(7): 2293-2301, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30545938

RESUMO

Catecholamine neurotransmitter levels in the synapses of the brain shape human disposition-cognitive flexibility, aggression, depression, and reward seeking-and manipulating these levels is a major objective of the pharmaceutical industry. Certain neurotransmitters are extensively sulfonated and inactivated by human sulfotransferase 1A3 (SULT1A3). To our knowledge, sulfonation as a therapeutic means of regulating transmitter activity has not been explored. Here, we describe the discovery of a SULT1A3 allosteric site that can be used to inhibit the enzyme. The structure of the new site is determined using spin-label-triangulation NMR. The site forms a cleft at the edge of a conserved ∼30-residue active-site cap that must open and close during the catalytic cycle. Allosteres anchor into the site via π-stacking interactions with two residues that sandwich the planar core of the allostere and inhibit the enzyme through cap-stabilizing interactions with substituents attached to the core. Changes in cap free energy were calculated ab initio as a function of core substituents and used to design and synthesize a series of inhibitors intended to progressively stabilize the cap and slow turnover. The inhibitors bound tightly (34 nm to 7.4 µm) and exhibited progressive inhibition. The cap-stabilizing effects of the inhibitors were experimentally determined and agreed remarkably well with the theoretical predictions. These studies establish a reliable heuristic for the design of SULT1A3 allosteric inhibitors and demonstrate that the free-energy changes of a small, dynamic loop that is critical for SULT substrate selection and turnover can be calculated accurately.


Assuntos
Arilsulfotransferase/química , Neurotransmissores/química , Regulação Alostérica , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Domínio Catalítico , Humanos , Neurotransmissores/genética , Neurotransmissores/metabolismo , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Marcadores de Spin
11.
Arch Biochem Biophys ; 648: 44-52, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29705271

RESUMO

Sulfoconjugation has been shown to be critically involved in the metabolism of acetaminophen (APAP), morphine, tapentadol and O-desmethyl tramadol (O-DMT). The objective of this study was to investigate the effects of single nucleotide polymorphisms (SNPs) of human SULT1A3 and SULT1A4 genes on the sulfating activity of SULT1A3 allozymes toward these analgesic compounds. Twelve non-synonymous coding SNPs (cSNPs) of SULT1A3/SULT1A4 were investigated, and the corresponding cDNAs were generated by site-directed mutagenesis. SULT1A3 allozymes, bacterially expressed and purified, exhibited differential sulfating activity toward each of the four analgesic compounds tested as substrates. Kinetic analyses of SULT1A3 allozymes further revealed significant differences in binding affinity and catalytic activity toward the four analgesic compounds. Collectively, the results derived from the current study showed clearly the impact of cSNPs of the coding genes, SULT1A3 and SULT1A4, on the sulfating activity of the coded SULT1A3 allozymes toward the tested analgesic compounds. These findings may have implications in the pharmacokinetics as well as the toxicity profiles of these analgesics administered in individuals with distinct SULT1A3 and/or SULT1A4 genotypes.


Assuntos
Acetaminofen/metabolismo , Analgésicos Opioides/metabolismo , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Citosol/enzimologia , Polimorfismo de Nucleotídeo Único , Sulfatos/metabolismo , Sulfotransferases/genética , Arilsulfotransferase/química , Humanos , Cinética , Modelos Moleculares , Conformação Proteica
12.
Proc Natl Acad Sci U S A ; 114(27): E5317-E5324, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28630292

RESUMO

Monoamine neurotransmitters are among the hundreds of signaling small molecules whose target interactions are switched "on" and "off" via transfer of the sulfuryl-moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and amines of their scaffolds. These transfer reactions are catalyzed by a small family of broad-specificity enzymes-the human cytosolic sulfotransferases (SULTs). The first structure of a SULT allosteric-binding site (that of SULT1A1) has recently come to light. The site is conserved among SULT1 family members and is promiscuous-it binds catechins, a naturally occurring family of flavanols. Here, the catechin-binding site of SULT1A3, which sulfonates monoamine neurotransmitters, is modeled on that of 1A1 and used to screen in silico for endogenous metabolite 1A3 allosteres. Screening predicted a single high-affinity allostere, tetrahydrobiopterin (THB), an essential cofactor in monoamine neurotransmitter biosynthesis. THB is shown to bind and inhibit SULT1A3 with high affinity, 23 (±2) nM, and to bind weakly, if at all, to the four other major SULTs found in brain and liver. The structure of the THB-bound binding site is determined and confirms that THB binds the catechin site. A structural comparison of SULT1A3 with SULT1A1 (its immediate evolutionary progenitor) reveals how SULT1A3 acquired high affinity for THB and that the majority of residue changes needed to transform 1A1 into 1A3 are clustered at the allosteric and active sites. Finally, sequence records reveal that the coevolution of these sites played an essential role in the evolution of simian neurotransmitter metabolism.


Assuntos
Aminas/química , Biopterinas/análogos & derivados , Neurotransmissores/química , Sítio Alostérico , Arilsulfotransferase/química , Sítios de Ligação , Biopterinas/química , Escherichia coli/metabolismo , Vetores Genéticos , Humanos , Isoenzimas/química , Cinética , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Ligação Proteica , Software , Enxofre/química
13.
J Steroid Biochem Mol Biol ; 172: 46-54, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28552400

RESUMO

Celecoxib is known to alter the preferred position of SULT2A1-catalyzed sulfonation of 17ß-estradiol (17ß-E2) and other estrogens from the 3- to the 17-position. Understanding the effects of celecoxib on estrogen sulfonation is of interest in the context of the investigational use of celecoxib to treat breast cancer. This study examined the effects on celecoxib on cytosolic sulfotransferases in human and rat liver and on SULT enzymes known to be expressed in liver. Celecoxib's effects on the sulfonation of several steroids catalyzed by human liver cytosol were similar but not identical to those observed previously for SULT2A1. Celecoxib was shown to inhibit recombinant SULT1A1-catalyzed sulfonation of 10nM estrone and 4µM p-nitrophenol with IC50 values of 2.6 and 2.1µM, respectively, but did not inhibit SULT1E1-catalyzed estrone sulfonation. In human liver cytosol, the combined effect of celecoxib and known SULT1A1 and 1E1 inhibitors, quercetin and triclosan, resulted in inhibition of 17ß-E2-3-sulfonation such that the 17-sulfate became the major metabolite: this is of interest because the 17-sulfate is not readily hydrolyzed by steroid sulfatase to 17ß-E2. Investigation of hepatic cytosolic steroid sulfonation in rat revealed that celecoxib did not stimulate 17ß-E2 17-sulfonation in male or female rat liver as it does with human SULT2A1 and human liver cytosol, demonstrating that rat is not a useful model of this effect. In silico studies suggested that the presence of the bulky tryptophan residue in the substrate-binding site of the rat SULT2A homolog instead of glycine as in human SULT2A1 may explain this species difference.


Assuntos
Arilsulfotransferase/metabolismo , Celecoxib/farmacologia , Estradiol/metabolismo , Estrona/metabolismo , Hepatócitos/enzimologia , Adulto , Animais , Arilsulfotransferase/química , Arilsulfotransferase/genética , Celecoxib/química , Estradiol/farmacologia , Estrona/farmacologia , Feminino , Expressão Gênica , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Modelos Moleculares , Simulação de Acoplamento Molecular , Nitrofenóis/química , Nitrofenóis/farmacologia , Cultura Primária de Células , Quercetina/química , Quercetina/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Homologia Estrutural de Proteína , Sulfatos/química , Sulfatos/metabolismo , Sulfotransferases/química , Sulfotransferases/genética , Sulfotransferases/metabolismo , Triclosan/química , Triclosan/farmacologia
14.
ChemSusChem ; 10(10): 2267-2273, 2017 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-28425669

RESUMO

We introduce the concept of using site-specific sulfation of various lignins for increasing their aqueous solubility and thereby their processability. Using p-nitrophenylsulfate as a sulfate source and an aryl sulfotransferase enzyme as catalyst, lignins are easily sulfated at ambient conditions. We demonstrate the specific sulfation of phenolic hydroxyl groups on five different lignins: Indulin AT (Kraft softwood), Protobind 1000 (mixed wheat straw/Sarkanda grass soda) and three organosolv lignins. The reaction proceeds smoothly and the increase in solubility is visible to the naked eye. We then examine the reaction kinetics, and show that these are easily monitored qualitatively and quantitatively using UV/Vis spectroscopy. The UV/Vis results are validated with 31 P NMR spectroscopy of the lignin phenol groups after derivatization with phosphorylation reagent II. In general, the results are more significant with organosolv lignins, as Kraft and soda lignins are produced from aqueous lignocellulose extraction processes.


Assuntos
Arilsulfotransferase/química , Lignina/química , Espectroscopia de Ressonância Magnética , Espectrofotometria Ultravioleta , Água/química
15.
Proc Natl Acad Sci U S A ; 113(50): 14312-14317, 2016 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-27911811

RESUMO

We are just beginning to understand the allosteric regulation of the human cytosolic sulfotransferase (SULTs) family-13 disease-relevant enzymes that regulate the activities of hundreds, if not thousands, of signaling small molecules. SULT1A1, the predominant isoform in adult liver, harbors two noninteracting allosteric sites, each of which binds a different molecular family: the catechins (naturally occurring flavonols) and nonsteroidal antiinflammatory drugs (NSAIDs). Here, we present the structure of an SULT allosteric binding site-the catechin-binding site of SULT1A1 bound to epigallocatechin gallate (EGCG). The allosteric pocket resides in a dynamic region of the protein that enables EGCG to control opening and closure of the enzyme's active-site cap. Furthermore, the structure offers a molecular explanation for the isozyme specificity of EGCG, which is corroborated experimentally. The binding-site structure was obtained without X-ray crystallography or multidimensional NMR. Instead, a SULT1A1 apoprotein structure was used to guide positioning of a small number of spin-labeled single-Cys mutants that coat the entire enzyme surface with a paramagnetic field of sufficient strength to determine its contribution to the bound ligand's transverse (T2) relaxation from its 1D solution spectrum. EGCG protons were mapped to the protein surface by triangulation using the T2 values to calculate their distances to a trio of spin-labeled Cys mutants. The final structure was obtained using distance-constrained molecular dynamics docking. This approach, which is readily extensible to other systems, is applicable over a wide range of ligand affinities, requires little protein, avoids the need for isotopically labeled protein, and has no protein molecular weight limitations.


Assuntos
Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Catequina/metabolismo , Sítio Alostérico , Arilsulfotransferase/genética , Catequina/análogos & derivados , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Modelos Moleculares , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Marcadores de Spin , Especificidade por Substrato
16.
Biochemistry ; 55(29): 4036-46, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27356022

RESUMO

The human cytosolic sulfotransferases (SULTs) comprise a 13-member enzyme family that regulates the activities of hundreds, perhaps thousands, of signaling small molecules via regiospecific transfer of the sulfuryl moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and amines of acceptors. Signaling molecules regulated by sulfonation include numerous steroid and thyroid hormones, epinephrine, serotonin, and dopamine. SULT1A1, a major phase II metabolism SULT isoform, is found at a high concentration in liver and has recently been show to harbor two allosteric binding sites, each of which binds a separate and complex class of compounds: the catechins (naturally occurring polyphenols) and nonsteroidal anti-inflammatory drugs. Among catechins, epigallocatechin gallate (EGCG) displays high affinity and specificity for SULT1A1. The allosteric network associated with either site has yet to be defined. Here, using equilibrium binding and pre-steady state studies, the network is shown to involve 14 distinct complexes. ECGG binds both the allosteric site and, relatively weakly, the active site of SULT1A1. It is not a SULT1A1 substrate but is sulfonated by SULT2A1. EGCG binds 17-fold more tightly when the active-site cap of the enzyme is closed by the binding of the nucleotide. When nucleotide is saturating, EGCG binds in two phases. In the first, it binds to the cap-open conformer; in the second, it traps the cap in the closed configuration. Cap closure encapsulates the nucleotide, preventing its release; hence, the EGCG-induced cap stabilization slows nucleotide release, inhibiting turnover. Finally, a comprehensive quantitative model of the network is presented.


Assuntos
Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Regulação Alostérica , Sítio Alostérico , Catequina/análogos & derivados , Catequina/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Ligantes , Fosfoadenosina Fosfossulfato/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
17.
Drug Metab Dispos ; 44(4): 481-4, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26658224

RESUMO

The human sulfotransferases (SULTs) regulate the activities of hundreds, if not thousands, of small molecule metabolites via transfer of the sulfuryl-moiety (-SO3) from the nucleotide donor, 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to the hydroxyls and amines of the recipients. Our understanding of the molecular basis of SULT catalysis has expanded considerably in recent years. The basic kinetic mechanism of these enzymes, previously thought to be ordered, has been redefined as random for SULT2A1, a representative member of the superfamily. An active-site cap whose structure and dynamics are highly responsive to nucleotides was discovered and shown to be critical in determining SULT selectivity, a topic of longstanding interest to the field. We now realize that a given SULT can operate in two specificity modes-broad and narrow-depending on the disposition of the cap. More recent work has revealed that the caps of the SULT1A1 are controlled by homotropic allosteric interactions between PAPS molecules bound at the dimer's active sites. These interactions cause the catalytic efficiency of SULT1A1 to vary in a substrate-dependent fashion by as much as two orders of magnitude over a range of PAPS concentrations that spans those found in human tissues. SULT catalysis is further complicated by the fact that these enzymes are frequently inhibited by their substrates. This review provides an overview of the mechanistic features of SULT1A1 that are important for the design and interpretation of SULT1A1 assays.


Assuntos
Arilsulfotransferase/química , Arilsulfotransferase/metabolismo , Ensaios Enzimáticos/métodos , Animais , Domínio Catalítico/fisiologia , Humanos , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , Especificidade por Substrato/fisiologia
18.
Biochemistry ; 54(39): 6114-22, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26340710

RESUMO

The human cytosolic sulfotransferases (SULTs) regulate hundreds, perhaps thousands, of small molecule metabolites and xenobiotics via transfer of a sulfuryl moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and primary amines of the recipients. In liver, where it is abundant, SULT1A1 engages in modifying metabolites and neutralizing toxins. The specificity of 1A1 is the broadest of any SULT, and understanding its selectivity is fundamental to understanding its biology. Here, for the first time, we show that SULT1A1 substrates separate naturally into two classes: those whose affinities are either enhanced ∼20-fold (positive synergy) or unaffected (neutral synergy) by the presence of a saturating nucleotide. kcat for the positive-synergy substrates is shown to be ∼100-fold greater than that of neutral-synergy compounds; consequently, the catalytic efficiency (kcat/Km) is approximately 3 orders of magnitude greater for the positive-synergy species. All-atom dynamics modeling suggests a molecular mechanism for these observations in which the binding of only positive-synergy compounds causes two phenylalanine residues (F81 and 84) to reposition and "sandwich" the phenolic moiety of the substrates, thus enhancing substrate affinity and positioning the nucleophilic oxygen for attack. Molecular dynamics movies reveal that the neutral-synergy compounds "wander" about the active site, infrequently achieving a reactive position. In-depth analysis of select point mutants strongly supports the model and provides an intimate view of the interdependent catalytic functions of subsections of the active site.


Assuntos
Arilsulfotransferase/química , Simulação de Dinâmica Molecular , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Domínio Catalítico , Humanos , Mutação Puntual , Relação Estrutura-Atividade , Especificidade por Substrato
19.
Biochem Pharmacol ; 94(4): 282-96, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25738837

RESUMO

Exogenous melatonin (Mel) is widely used in clinic for multiple therapeutic purposes. In metabolism pathways of Mel, 6-hydroxymelatonin-sulfate (S-O-Mel) and N-acetylserotonin sulfate (S-NAS) are the most abundant metabolites account for over 90% of total Mel metabolites in humans, indicating that sulfation plays an important role in reflecting the functions and clearance of Mel in vivo. In the present study, we characterized Mel sulfation using various human organ cytosols (liver, lung, kidney, small intestine and brain), liver cytosols from five different animal species, and cDNA-expressed human sulfotransferase (SULT) for the first time. Our results demonstrated that liver, lung, kidney and small intestine of humans had high catalytic efficiency for Mel sulfation, however, brain contained a very low reaction rate. Interestingly, organ cytosols prepared from females exhibited higher sulfation activity than those of males. SULT isoforms 1A1, 1A2, 1A3, 1B1 and 1E1 exhibited metabolic activities toward Mel. According to kinetic parameters (Km and Vmax), chemical inhibition, correlation analysis, molecular docking and sulfation assays with recombinant human SULTs isoforms, SULT1A1 was determined as the major enzyme responsible for Mel sulfation. Furthermore, considerable species differences in Mel sulfation were observed, and the total intrinsic clearance rate of Mel sulfation was as follows: monkey>rat>dog>human>pig>mouse. Additionally, the anti-inflammatory effects of Mel and its sulfated metabolites were evaluated by inhibiting nitric oxide (NO) production in RAW264.7 cells, and S-O-Mel as a bioactive form, exhibited potent bioactivity. Our investigation provided a global view of the enzyme-dependent sulfation of Mel that can guide biomedical research on Mel.


Assuntos
Arilsulfotransferase/metabolismo , Melatonina/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Arilsulfotransferase/química , Linhagem Celular , Cães , Feminino , Haplorrinos , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Masculino , Melatonina/química , Melatonina/farmacologia , Camundongos , Simulação de Acoplamento Molecular , Óxido Nítrico/biossíntese , Especificidade de Órgãos , Ratos , Especificidade da Espécie , Suínos
20.
Xenobiotica ; 45(6): 495-502, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25539458

RESUMO

1. Cattle are an important component of the human food chain. Drugs used either legally or illegally in cattle may therefore enter the food chain and it is thus important to understand pathways of drug metabolism in this species, including sulfation catalyzed by the sulfotransferases (SULTs). 2. In this study, we have analyzed the sulfation of 4-nitrophenol and other compounds in male and female bovine liver and characterized recombinant bovine SULT isoforms 1A1 and 1B1 expressed in Escherichia coli. 3. We found that, in contrast to most other mammalian species, the major phenol sulfotransferase SULT1A1 is not expressed in bovine liver. Rather SULT1B1 seems to be a major form in both male and female bovine liver. 4. We also identified kinetic differences between bovine and human SULT1A1 and, using the human SULT1A1 crystal structure, identified two amino acid positions in the active site of bovine SULT1A1 (Ile89Val and Phe247Val) that may be responsible for these differences.


Assuntos
Fígado/enzimologia , Sulfotransferases/química , Sulfotransferases/metabolismo , Animais , Arilsulfotransferase/química , Arilsulfotransferase/genética , Arilsulfotransferase/metabolismo , Bovinos , Cristalografia por Raios X , Feminino , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Nitrofenóis/farmacocinética , Nitrofenóis/farmacologia , Sulfotransferases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...