Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Drug Metab Dispos ; 47(9): 949-953, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31266751

RESUMEN

Sulfotransferase 4A1 (SULT4A1), a member of cytosolic sulfotransferases (SULT), is exclusively expressed in neurons with no known function. Severe phenotype and early postnatal death in SULT4A1 knockout mice revealed that SULT4A1 is an essential neuronal protein. Localization of SULT4A1 in different cytosolic compartments, including mitochondria, suggests multiple roles for this protein. We observed that knockdown of SULT4A1 results in the accumulation of reactive oxygen species in primary cortical neurons, suggesting a potential role of SULT4A1 in regulating redox homeostasis. Expression of SULT4A1 in the human neuroblastoma SH-SY5Y cells revealed a defused but nonuniform staining pattern in the cytoplasm, with increased density around mitochondria. Subcellular fractionation of SULT4A1 expressing SH-SY5Y cells confirms the presence of SULT4A1 in mitochondrial fractions. SULT4A1 expressing cells display significant protection against H2O2-mediated defects in mitochondrial function and loss of mitochondrial membrane potential. Expression of SULT4A1 in SH-SY5Y cells also protects against H2O2-induced cell death. These data indicate that SULT4A1 protects mitochondria against oxidative damage and may serve as a potential pharmacological target in neural diseases involving mitochondrial dysfunction and oxidative stress. SIGNIFICANCE STATEMENT: Studies on SULT4A1 knockout mice suggest that SULT4A1 plays a vital role in neuronal function and survival via yet undefined mechanisms. Our data demonstrate that depletion of SULT4A1 induces oxidative stress in neurons and expression of SULT4A1 in SH-SY5Y cells protects against oxidative-stress-induced mitochondrial dysfunction and cell death. These results suggest that SULT4A1 may have a crucial protective function against mitochondrial dysfunction and oxidative stress, and may serve a potential therapeutic target in different neurological diseases involving mitochondrial dysfunction and oxidative stress.


Asunto(s)
Mitocondrias/patología , Neuronas/patología , Sulfotransferasas/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Corteza Cerebral/citología , Clonación Molecular , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Neuronas/citología , Estrés Oxidativo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sulfotransferasas/genética
2.
Drug Metab Dispos ; 46(1): 41-45, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29109113

RESUMEN

Sulfotransferase 4A1 (SULT4A1) belongs to the cytosolic sulfotransferase (SULT) superfamily of enzymes that catalyze sulfonation reactions with a variety of endogenous and exogenous substrates. Of the SULTs, SULT4A1 was shown to have the highest sequence homology between vertebrate species, yet no known function or enzymatic activity has been identified for this orphan SULT. To better understand SULT4A1 function in mammalian brain, two mutant SULT4A1 mouse strains were generated utilizing clustered regulatory interspaced short palindromic repeats (CRISPR)-content-addressable storage (Cas) 9 technology. The first strain possessed a 28-base pair (bp) deletion (Δ28) in exon 1 that resulted in a frameshift mutation with premature termination. The second strain possessed a 12-bp in-frame deletion (Δ12) immediately preceding an active site histidine111 common to the SULT family. Homozygous pups of both strains present with severe and progressive neurologic symptoms, including tremor, absence seizures, abnormal gait, ataxia, decreased weight gain compared with littermates, and death around postnatal days 21-25. SULT4A1 immunostaining was decreased in brains of heterozygous pups and not detectable in homozygous pups of both SULT4A1 mutants. SULT4A1 localization in subcellular fractions of mouse brain showed SULT4A1 associated with mitochondrial, cytosolic, and microsomal fractions, a novel localization pattern for SULTs. Finally, primary cortical neurons derived from embryonic (E15) CD-1 mice expressed high levels of SULT4A1 throughout the cell except in nuclei. Taken together, SULT4A1 appears to be an essential neuronal protein required for normal brain function, at least in mammals. Mouse models will be valuable in future studies to investigate the regulation and functions of SULT4A1 in the mammalian brain.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Sulfotransferasas/metabolismo , Animales , Conducta Animal , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Sistemas CRISPR-Cas/genética , Citosol/metabolismo , Exones/genética , Femenino , Mutación del Sistema de Lectura/genética , Homocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Proteínas del Tejido Nervioso/genética , Cultivo Primario de Células , Sulfotransferasas/genética
3.
Xenobiotica ; 48(1): 79-88, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28084139

RESUMEN

1. Human cytosolic sulfotransferase 1B1 (SULT1B1) sulfates small phenolic compounds and bioactivates polycyclic aromatic hydrocarbons. To date, no SULT1B1 allelic variants have been well-characterized. 2. While cloning SULT1B1 from human endometrial specimens, an allelic variant resulting in valine instead of leucine at the 145th amino acid position (L145V) was detected. NCBI reported this alteration as the highest frequency SULT1B1 allelic variant. 3. L145V frequency comprised 9% of 37 mixed-population human patients and was specific to African Americans with an allelic frequency of 25%. Structurally, replacement of leucine with valine potentially destabilizes a conserved helix (α8) that forms the "floor" of both the substrate and PAPS binding domains. This destabilization results in altered kinetic properties including a four-fold decrease in affinity for PAP (3', 5'-diphosphoadenosine). Kms for 3'-phosphoadenosine- 5'-phosphosulfate (PAPS) are similar; however, maximal turnover rate of the variant isoform (0.86 pmol/(min*µg)) is slower than wild-type (WT) SULT1B1 (1.26 pmol/(min*µg)). The L145V variant also displays altered kinetics toward small phenolic substrates, including a diminished p-nitrophenol Km and increased susceptibility to 1-naphthol substrate inhibition. 4. No significant correlation between genotype and prostate or colorectal cancer was observed in patients; however, the variant isoform could underlie specific pathologies in sub-Saharan African carriers.


Asunto(s)
Sulfotransferasas/genética , Negro o Afroamericano , Humanos , Mutación Missense
4.
J Lipid Res ; 57(7): 1133-43, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27230263

RESUMEN

Bile acid CoA:amino acid N-acyltransferase (BAAT) is the terminal enzyme in the synthesis of bile salts from cholesterol and catalyzes the conjugation of taurine or glycine to bile acid CoA thioesters to form bile acid N-acylamidates. BAAT has a dual localization to the cytosol and peroxisomes, possibly due to an inefficient carboxy-terminal peroxisomal targeting signal (PTS), -serine-glutamine-leucine (-SQL). Mutational analysis was used to define the role of the carboxy terminus in peroxisomal localization and kinetic activity. Amidation activity of BAAT and BAAT lacking the final two amino acids (AAs) (BAAT-S) were similar, whereas the activity of BAAT with a canonical PTS sequence (BAAT-SKL) was increased >2.5-fold. Kinetic analysis of BAAT and BAAT-SKL showed that BAAT-SKL had a lower Km for taurine and glycine as well as a greater Vmax There was no difference in the affinity for cholyl-CoA. In contrast to BAAT, BAAT-SKL forms bile acid N-acylamidates with ß-alanine. BAAT-S immunoprecipitated when incubated with peroxisomal biogenesis factor 5 (Pex5) and rabbit anti-Pex5 antibodies; however, deleting the final 12 AAs prevented coimmunoprecipitation with Pex5, indicating the Pex5 interaction involves more than the -SQL sequence. These results indicate that even small changes in the carboxy terminus of BAAT can have significant effects on activity and substrate specificity.


Asunto(s)
Ácidos y Sales Biliares/genética , Hígado/enzimología , Esfingosina N-Aciltransferasa/genética , Ácidos y Sales Biliares/metabolismo , Citosol/enzimología , Análisis Mutacional de ADN , Humanos , Cinética , Mutación , Receptor de la Señal 1 de Direccionamiento al Peroxisoma , Peroxisomas/enzimología , Receptores Citoplasmáticos y Nucleares/metabolismo , Esfingosina N-Aciltransferasa/química , Esfingosina N-Aciltransferasa/metabolismo , Especificidad por Sustrato
5.
J Biol Chem ; 289(38): 26474-26480, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25056952

RESUMEN

Human cytosolic sulfotransferases (SULTs) regulate the activities of thousands of signaling small molecules via transfer of the sulfuryl moiety (-SO3) from 3'-phosphoadenosine 5'-phosphosulfate (PAPS) to the hydroxyls and primary amines of acceptors. Sulfonation controls the affinities of ligands for their targets, and thereby regulates numerous receptors, which, in turn, regulate complex cellular responses. Despite their biological and medical relevance, basic SULT mechanism issues remain unresolved. To settle these issues, and to create an in-depth model of SULT catalysis, the complete kinetic mechanism of a representative member of the human SULT family, SULT2A1, was determined. The mechanism is composed of eight enzyme forms that interconvert via 22 rate constants, each of which was determined independently. The result is a complete quantitative description of the mechanism that accurately predicts complex enzymatic behavior. This is the first description of a SULT mechanism at this resolution, and it reveals numerous principles of SULT catalysis and resolves previously ambiguous issues. The structures and catalytic behaviors SULTs are highly conserved; hence, the mechanism presented here should prove paradigmatic for the family.


Asunto(s)
Sulfotransferasas/química , Biocatálisis , Deshidroepiandrosterona/química , Humanos , Cinética , Modelos Químicos , Fosfoadenosina Fosfosulfato/química , Unión Proteica , Sulfotransferasas/antagonistas & inhibidores
6.
Drug Metab Dispos ; 43(3): 418-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25534770

RESUMEN

Human sulfotransferases (SULTs) comprise a small, 13-member enzyme family that regulates the activities of thousands of compounds-endogenous metabolites, drugs, and other xenobiotics. SULTs transfer the sulfuryl-moiety (-SO3) from a nucleotide donor, PAPS (3'-phosphoadenosine 5'-phosphosulfate), to the hydroxyls and primary amines of acceptors. SULT1A1, a progenitor of the family, has evolved to sulfonate compounds that are remarkably structurally diverse. SULT1A1, which is found in many tissues, is the predominant SULT in liver, where it is a major component of phase II metabolism. Early work demonstrated that catechins and nonsteroidal anti-inflammatory drugs inhibit SULT1A1 and suggested that the inhibition was not competitive versus substrates. Here, the mechanism of inhibition of a single, high affinity representative from each class [epigallocatechin gallate (EGCG) and mefenamic acid] is determined using initial-rate and equilibrium-binding studies. The findings reveal that the inhibitors bind at sites separate from those of substrates, and at saturation turnover of the enzyme is reduced to a nonzero value. Further, the EGCG inhibition patterns suggest a molecular explanation for its isozyme specificity. Remarkably, the inhibitors bind at sites that are separate from one another, and binding at one site does not affect affinity at the other. For the first time, it is clear that SULT1A1 is allosterically regulated, and that it contains at least two, functionally distinct allosteric sites, each of which responds to a different class of compounds.


Asunto(s)
Sitio Alostérico/fisiología , Arilsulfotransferasa/metabolismo , Unión Proteica/fisiología , Catequina/análogos & derivados , Catequina/metabolismo , Humanos , Ácido Mefenámico/metabolismo
7.
Drug Metab Dispos ; 43(7): 1037-44, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25934576

RESUMEN

Since its identification in 2000, sulfotransferase (SULT) 4A1 has presented an enigma to the field of cytosolic SULT biology. SULT4A1 is exclusively expressed in neural tissue, is highly conserved, and has been identified in every vertebrate studied to date. Despite this singular level of conservation, no substrate or function for SULT4A1 has been identified. Previous studies demonstrated that SULT4A1 does not bind the obligate sulfate donor, 3'-phosphoadenosine-5'-phosphosulfate, yet SULT4A1 is classified as a SULT superfamily member based on sequence and structural similarities to the other SULTs. In this study, transcription activator-like effector nucleases were used to generate heritable mutations in the SULT4A1 gene of zebrafish. The mutation (SULT4A1(Δ8)) consists of an 8-nucleotide deletion within the second exon of the gene, resulting in a frameshift mutation and premature stop codon after 132 AA. During early adulthood, casual observations were made that mutant zebrafish were exhibiting excessively sedentary behavior during the day. These observations were inconsistent with published reports on activity in zebrafish that are largely diurnal organisms and are highly active during the day. Thus, a decrease in activity during the day represents an abnormal behavior and warranted further systematic analysis. EthoVision video tracking software was used to monitor activity levels in wild-type (WT) and SULT4A1(Δ8/Δ8) fish over 48 hours of a normal light/dark cycle. SULT4A1(Δ8/Δ8) fish were shown to exhibit increased inactivity bout length and frequency as well as a general decrease in daytime activity levels when compared with their WT counterparts.


Asunto(s)
Mutación del Sistema de Lectura/genética , Actividad Motora/genética , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/metabolismo , Secuencia de Aminoácidos , Animales , Ansiedad/genética , Ansiedad/psicología , Secuencia de Bases , Desoxirribonucleasas/administración & dosificación , Desoxirribonucleasas/farmacología , Embrión no Mamífero , Exones , Microinyecciones , Datos de Secuencia Molecular , Mutación , Conducta Social
8.
J Biol Chem ; 288(48): 34494-501, 2013 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-24129576

RESUMEN

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.


Asunto(s)
Arilsulfotransferasa/química , Simulación por Computador , Bibliotecas de Moléculas Pequeñas/química , Sulfotransferasas/química , Arilsulfotransferasa/antagonistas & inhibidores , Arilsulfotransferasa/genética , Citosol/enzimología , Inhibidores Enzimáticos/química , Humanos , Fase II de la Desintoxicación Metabólica , Simulación de Dinámica Molecular , Unión Proteica , Especificidad por Sustrato , Sulfatos/química , Sulfatos/metabolismo , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/genética
9.
J Biol Chem ; 288(12): 8619-8626, 2013 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-23362278

RESUMEN

Human cytosolic sulfotransferases (SULTs) regulate the activities of hundreds of signaling metabolites via transfer of the sulfuryl moiety (-SO3) from activated sulfate (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and primary amines of xeno- and endobiotics. How SULTs select substrates from the scores of competing ligands present in a cytosolic milieu is an important issue in the field. Selectivity appears to be sterically controlled by a molecular pore that opens and closes in response to nucleotide binding. This point of view is fostered by structures showing nucleotide-dependent pore closure and the fact that nucleotide binding induces an isomerization that restricts access to the acceptor-binding pocket. Molecular dynamics models underscore the importance of pore isomerization in selectivity and predict that specific molecular linkages stabilize the closed pore in response to nucleotide binding. To test the pore model, these linkages were disrupted in SULT2A1 via mutagenesis, and the effects on selectivity were determined. The mutations uncoupled nucleotide binding from selectivity and produced enzymes that no longer discriminated between large and small substrates. The mutations did not affect the affinity or turnover of small substrates but resulted in a 183-fold gain in catalytic efficiently toward large substrates. Models predict that an 11-residue "flap" covering the acceptor-binding pocket can open and admit large substrates when nucleotide is bound; a mutant structure demonstrated that this is so. In summary, the model was shown to be a robust, accurate predictor of SULT structure and selectivity whose general features will likely apply to other members of the SULT family.


Asunto(s)
Modelos Moleculares , Sulfotransferasas/química , Sustitución de Aminoácidos , Dominio Catalítico , Cristalografía por Rayos X , Deshidroepiandrosterona/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Mutagénesis Sitio-Dirigida , Unión Proteica , Clorhidrato de Raloxifeno/química , Sulfotransferasas/genética , Termodinámica
10.
Drug Metab Dispos ; 42(5): 947-53, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24553382

RESUMEN

Sulfotransferase (SULT) 4A1 is an orphan enzyme that shares distinct structure and sequence similarities with other cytosolic SULTs. SULT4A1 is primarily expressed in neuronal tissue and is also the most conserved SULT, having been identified in every vertebrate investigated to date. Certain haplotypes of the SULT4A1 gene are correlated with higher baseline psychopathology in schizophrenic patients, but no substrate or function for SULT4A1 has yet been identified despite its high level of sequence conservation. In this study, deep RNA sequencing was used to search for alterations in gene expression in 72-hour postfertilization zebrafish larvae following transient SULT4A1 knockdown (KD) utilizing splice blocking morpholino oligonucleotides. This study demonstrates that transient inhibition of SULT4A1 expression in developing zebrafish larvae results in the up-regulation of several genes involved in phototransduction. SULT4A1 KD was verified by immunoblot analysis and quantitative real-time polymerase chain reaction (qPCR). Gene regulation changes identified by deep RNA sequencing were validated by qPCR. This study is the first identification of a cellular process whose regulation appears to be associated with SULT4A1 expression.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Fototransducción/genética , Sulfotransferasas/fisiología , Transcriptoma , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Encéfalo/embriología , Encéfalo/metabolismo , Ojo/embriología , Ojo/metabolismo , Fertilización , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Larva , Datos de Secuencia Molecular , Morfolinos/farmacología , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ARN , Homología de Secuencia de Aminoácido , Sulfotransferasas/genética , Regulación hacia Arriba , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
11.
Drug Metab Dispos ; 42(3): 352-60, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24335392

RESUMEN

The cystolic sulfotransferse 1C3 (SULT1C3) gene was identified by computational analysis of the human genome and suggested to contain duplications of its last two exons (7a/b and 8a/b). Although the SULT1C3 isoform containing the more downstream exons 7b and 8b (SULT1C3d) has been expressed in Escherichia coli, crystallized, and characterized for activity, there is currently no evidence that SULT1C3 is expressed in any human tissue. Using reverse-transcription polymerase chain reaction, we detected SULT1C3 mRNA in the colorectal adenocarcinoma cell line (LS180), colon, and small intestine, but the amplified fragment contained the more upstream exons 7a and 8a. 3'-Rapid amplification of cDNA ends (RACE) confirmed that the SULT1C3 transcript expressed in LS180 cells contained exons 7a/8a, whereas 5'-RACE identified a noncoding exon 1. Full-length SULT1C3 transcript containing exons 7a/8a was amplified from LS180 and intestinal RNA, and in vitro transcription-translation of the cloned cDNA indicated that translation primarily began at the first of three in-frame ATG codons. Since SULT1C3 containing exons 7a/8a (SULT1C3a) would differ by 30 amino acids from SULT1C3d containing exons 7b/8b, we considered the functional implications of expressing one or the other isoform by generating structural models based on the reported crystal structure for SULT1C3d. Comparison of the structures indicated that five of the residues forming the substrate-binding pocket differed between the two isoforms, resulting in a change in both electron density and charge distribution along the inner wall of the substrate-binding pocket. These data indicate that SULT1C3 is expressed in human intestine but suggest that the expressed isoform is likely to differ functionally from the isoform that has been previously characterized.


Asunto(s)
Colon/enzimología , Intestino Delgado/enzimología , Sulfotransferasas/genética , Transcripción Genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular Tumoral , Codón , Exones , Humanos , Datos de Secuencia Molecular , Isoformas de Proteínas , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfotransferasas/biosíntesis , Sulfotransferasas/química
12.
Biochemistry ; 52(2): 415-24, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23256751

RESUMEN

Human cytosolic sulfotransferases (SULTs) transfer the sulfuryl moiety (-SO(3)) from activated sulfate [3'-phosphoadenosine 5'-phosphosulfate (PAPS)] to the hydroxyls and primary amines of numerous metabolites, drugs, and xenobiotics. Receipt of the sulfuryl group often radically alters acceptor-target interactions. How these enzymes select particular substrates from the hundreds of candidates in a complex cytosol remains an important question. Recent work reveals PAPS binding causes SULT2A1 to undergo an isomerization that controls selectivity by constricting the opening through which acceptors must pass to enter the active site. The enzyme maintains an affinity for large substrates by isomerizing between the open and closed states with nucleotide bound. Here, the molecular basis of the nucleotide-induced closure is explored in equilibrium and nonequilibrium molecular dynamics simulations. The simulations predict that the active-site "cap," which covers both the nucleotide and acceptor binding sites, opens and closes in response to nucleotide. The cap subdivides into nucleotide and acceptor halves whose motions, while coupled, exhibit an independence that can explain the isomerization. In silico weakening of electrostatic interactions between the cap and base of the active site causes the acceptor half of the cap to open and close while the nucleotide lid remains shut. Simulations predict that SULT1A1, the most abundant SULT in human liver, will utilize a similar selection mechanism. This prediction is tested using fulvestrant, an anti-estrogen too large to pass through the closed pore, and estradiol, which is not restricted by closure. Equilibrium and pre-steady-state binding studies confirm that SULT1A1 undergoes a nucleotide-induced isomerzation that controls substrate selection.


Asunto(s)
Arilsulfotransferasa/química , Arilsulfotransferasa/metabolismo , Nucleótidos/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , Conformación Proteica , Especificidad por Sustrato
13.
Drug Metab Rev ; 45(1): 15-33, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23330539

RESUMEN

The cytosolic sulfotransferases (SULTs) are a multigene family of enzymes that catalyze the transfer of a sulfonate group from the physiologic sulfate donor, 3'-phosphoadenosine-5'-phosphosulfate, to a nucleophilic substrate to generate a polar product that is more amenable to elimination from the body. As catalysts of both xenobiotic and endogenous metabolism, the SULTs are major points of contact between the external and physiological environments, and modulation of SULT-catalyzed metabolism can not only affect xenobiotic disposition, but it can also alter endogenous metabolic processes. Therefore, it is not surprising that SULT expression is regulated by numerous members of the nuclear receptor (NR) superfamily that function as sensors of xenobiotics as well as endogenous molecules, such as fatty acids, bile acids, and oxysterols. These NRs include the peroxisome proliferator-activated receptors, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, liver X receptors, farnesoid X receptor, retinoid-related orphan receptors, and estrogen-related receptors. This review summarizes current information about NR regulation of SULT expression. Because species differences in SULT subfamily composition and tissue-, sex-, development-, and inducer-dependent regulation are prominent, these differences will be emphasized throughout the review. In addition, because of the central role of the SULTs in cellular physiology, the effect of NR-mediated SULT regulation on physiological and pathophysiological processes will be discussed. Gaps in current knowledge that require further investigation are also highlighted.


Asunto(s)
Citosol/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Sulfotransferasas/metabolismo , Animales , Citosol/enzimología , Humanos , Receptores Citoplasmáticos y Nucleares/genética , Sulfotransferasas/genética , Xenobióticos/farmacocinética
14.
Drug Metab Rev ; 45(4): 388-400, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24020383

RESUMEN

The SULT2b gene family consists of a single gene capable of generating two functional transcripts utilizing different transcriptional start sites in the first exon. This results in the translation of two separate proteins, SULT2B1a and SULT2B1b, with different amino-terminal peptides and approximately 95% identical sequences. The second distinguishing feature of the SULT2B isoforms is the proline/serine-rich carboxy-terminal sequence. To date, presence of the SULT2B gene appears limited to mammals and there is also only limited conservation of structure or sequence of the carboxy-terminal peptide. Although both SULT2B1 messages are present in human tissues, to date, only the SULT2B1b protein has been detected in the tissues investigated. In contrast, selective expression of SULT2B1a has been detected in rodent brain, whereas SULT2B1b was expressed in skin and intestine. Characterization of the SULT2B1 isoforms has been limited by the inability to isolate reliably active SULT2B1b from tissues or cells. SULT2B1 cDNAs can be expressed in Escherichia coli and the expressed active enzymes show selectivity for sulfation of 3ß-hydroxysteroids. SULT2B1b due to the binding properties of the amino-terminal peptides also shows high cholesterol sulfation activity. Although human SULT2B1b displays significant substrate cross-reactivity with SULT2A1, the isoforms have different tissue expression patterns. Human SULT2B1b also shows nuclear localization in selected tissues that appears related to serine phosphorylation of the carboxy-terminal peptide. Overall, the understanding of the properties and function of the SULT2B1 isoforms is limited and the structural variability of the unique amino- and carboxy-sequences suggests significant species differences that need to be investigated.


Asunto(s)
Sulfotransferasas/genética , Sulfotransferasas/fisiología , Secuencia de Aminoácidos , Animales , Núcleo Celular/enzimología , Colesterol/metabolismo , Citosol/enzimología , Humanos , Isoenzimas , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Prolina/genética , Prolina/metabolismo , Unión Proteica , Alineación de Secuencia , Serina/genética , Serina/metabolismo , Especificidad de la Especie , Sulfotransferasas/metabolismo
15.
Mutagenesis ; 28(5): 609-19, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23894158

RESUMEN

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.


Asunto(s)
Metilcolantreno/análogos & derivados , Mutágenos/farmacocinética , Salmonella typhimurium/genética , Sulfotransferasas/metabolismo , Acetaminofén/química , Acetaminofén/metabolismo , Animales , Arilsulfotransferasa/antagonistas & inhibidores , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Perros , Inhibidores Enzimáticos/farmacología , Humanos , Isomerismo , Metilcolantreno/química , Metilcolantreno/farmacocinética , Metilcolantreno/toxicidad , Ratones , Simulación del Acoplamiento Molecular , Pruebas de Mutagenicidad , Mutágenos/metabolismo , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Salmonella typhimurium/enzimología , Especificidad de la Especie , Sulfotransferasas/química , Sulfotransferasas/genética
16.
Methods Enzymol ; 689: 332-352, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37802577

RESUMEN

Conjugation of steroids and sterol compounds with a sulfonate group is a major pathway in the regulation of their activity, synthesis and excretion. Three human cytosolic sulfotransferases are highly involved in the sulfonation of sterol compounds. SULT1E1 has a low nM affinity for estrogen sulfonation and also conjugates non-aromatic steroids with a significantly lower affinity. SULT2A1 is responsible for the high levels of fetal and adult dehydroepiandrosterone (DHEA) sulfate synthesis in the adrenal gland as well as many 3α and 3ß-hydroxysteroids and bile acids. SULT2B1b is responsible for the majority of cholesterol sulfation in tissues as well as conjugating 3ß-hydroxysteroids. Although there are multiple methods for assaying cytosolic SULT activity, two relatively simple, rapid and versatile assays for steroid sulfonation are described. The first method utilizes radiolabeled substrates and organic solvent extraction to isolate the radiolabeled product from the aqueous phase. The second assay utilizes 35S-3'-phosphoadenosine 5'-phosphosulfate (PAPS) to generate 35S-conjugated products that are resolved by thin layer chromatography. Both assays useful in situations requiring measurement of SULT activity in a timely manner.


Asunto(s)
Esteroides , Sulfotransferasas , Adulto , Humanos , Hidroxiesteroides , Sulfotransferasas/metabolismo , Esteroles
17.
Sci Rep ; 13(1): 1377, 2023 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-36697463

RESUMEN

Tyrosyl-DNA phosphodiesterase I (Tdp1) hydrolyzes phosphodiester-linked adducts from both ends of DNA. This includes the topoisomerase I (TOP1)-DNA covalent reaction intermediate that is the target of the camptothecin class of chemotherapeutics. Tdp1 two-step catalysis is centered on the formation of a Tdp1-DNA covalent complex (Tdp1cc) using two catalytic histidines. Here, we examined the role of the understudied, structurally undefined, and poorly conserved N-terminal domain (NTD) of Tdp1 in context of full-length protein in its ability to remove TOP1cc in cells. Using toxic Tdp1 mutants, we observed that the NTD is critical for Tdp1's ability to remove TOP1-DNA adducts in yeast. Full-length and N-terminal truncated Tdp1 mutants showed similar expression levels and cellular distribution yet an inversed TOP1-dependent toxicity. Single turnover catalysis was significantly different between full-length and truncated catalytic mutants but not wild-type enzyme, suggesting that Tdp1 mutants depend on the NTD for catalysis. These observations suggest that the NTD plays a critical role in the regulation of Tdp1 activity and interaction with protein-DNA adducts such as TOP1cc in cells. We propose that the NTD is a regulatory domain and coordinates stabilization of the DNA-adducted end within the catalytic pocket to access the phosphodiester linkage for hydrolysis.


Asunto(s)
Aductos de ADN , ADN-Topoisomerasas de Tipo I , Hidrolasas Diéster Fosfóricas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , ADN , Reparación del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
Biochemistry ; 51(28): 5674-83, 2012 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-22703301

RESUMEN

Human SULT2A1 is one of two predominant sulfotransferases in liver and catalyzes transfer of the sulfuryl moiety (-SO(3)) from activated sulfate (PAPS, 3'-phosphoadenosine 5-phosphosulfate) to hundreds of acceptors (metabolites and xenobiotics). Sulfation recodes the biologic activity of acceptors by altering their receptor interactions. The molecular basis on which these enzymes select and sulfonate specific acceptors from complex mixtures of competitors in vivo is a long-standing issue in the SULT field. Raloxifene, a synthetic steroid used in the prevention of osteoporosis, and dehydroepiandrosterone (DHEA), a ubiquitous steroid precusor, are reported to be sulfated efficiently by SULT2A1 in vitro, yet unlike DHEA, raloxifene is not sulfated in vivo. This selectivity was explored in initial rate and equilibrium binding studies that demonstrate pronounced binding antisynergy (21-fold) between PAPS and raloxifene, but not DHEA. Analysis of crystal structures suggests that PAP binding restricts access to the acceptor-binding pocket by restructuring a nine-residue segment of the pocket edge that constricts the active site opening, or "pore", that sieves substrates on the basis of their geometries. In silico docking predicts that raloxifene, which is considerably larger than DHEA, can bind only to the unliganded (open) enzyme, whereas DHEA binds both the open and closed forms. The predictions of these structures with regard to substrate binding are tested using equilibrium and pre-steady-state ligand binding studies, and the results confirm that a nucleotide-driven isomerization controls access to the acceptor-binding pocket and plays an important role in substrate selection by SULT2A1 and possibly other sulfotransferases.


Asunto(s)
Conservadores de la Densidad Ósea/química , Deshidroepiandrosterona/química , Fosfoadenosina Fosfosulfato/química , Clorhidrato de Raloxifeno/química , Sulfotransferasas/química , Simulación por Computador , Cinética , Simulación del Acoplamiento Molecular , Unión Proteica , Proteínas Recombinantes/química , Especificidad por Sustrato
19.
Drug Metab Dispos ; 40(8): 1588-95, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22593037

RESUMEN

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.


Asunto(s)
Arilsulfotransferasa/antagonistas & inhibidores , Etinilestradiol/farmacología , Fosfoadenosina Fosfosulfato/farmacología , Humanos
20.
Sci Rep ; 12(1): 1625, 2022 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-35102205

RESUMEN

Sulfotransferase 4A1 (SULT4A1) is an orphan member of the cytosolic SULT superfamily that contains enzymes that catalyze the sulfonation of hydrophobic drugs and hormones. SULT4A1 has been assessed through all classical SULT approaches yet no SULT activity has been reported. To ascertain SULT4A1 function and activity, we utilized Saccharomyces cerevisiae as a model system, which exhibits no endogenous SULT activity nor possesses SULT-related genes. We observed that ectopic SULT4A1 expression in yeast displays similar subcellular localization as reported in mouse neurons and observed that SULT4A1 is associated with the outer mitochondria membrane. SULT4A1 expression stimulates colony formation and protects these cells from hydrogen peroxide and metabolism-associated oxidative stress. These SULT4A1-mediated phenotypes are dependent on extracellular sulfate that is converted in yeast to PAPS, the universal sulfonate donor for SULT activity. Thus, heterologous SULT4A1 expression in yeast is correctly distributed and functional, and SULT4A1 antioxidant activity is sulfate dependent supporting the concept that SULT4A1 has sulfate-associated activity.


Asunto(s)
Sulfatos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA