Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 430
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(23)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38069221

RESUMEN

Sulfotransferases (SULTs) are phase II metabolizing enzymes catalyzing the sulfoconjugation from the co-factor 3'-Phosphoadenosine 5'-Phosphosulfate (PAPS) to a wide variety of endogenous compounds, drugs and natural products. Although SULT1A1 and SULT1A3 share 93% identity, SULT1A1, the most abundant SULT isoform in humans, exhibits a broad substrate range with specificity for small phenolic compounds, while SULT1A3 displays a high affinity toward monoamine neurotransmitters like dopamine. To elucidate the factors determining the substrate specificity of the SULT1 isoenzymes, we studied the dynamic behavior and structural specificities of SULT1A1 and SULT1A3 by using molecular dynamics (MD) simulations and ensemble docking of common and specific substrates of the two isoforms. Our results demonstrated that while SULT1A1 exhibits a relatively rigid structure by showing lower conformational flexibility except for the lip (loop L1), the loop L2 and the cap (L3) of SULT1A3 are extremely flexible. We identified protein residues strongly involved in the recognition of different substrates for the two isoforms. Our analyses indicated that being more specific and highly flexible, the structure of SULT1A3 has particularities in the binding site, which are crucial for its substrate selectivity.


Asunto(s)
Isoenzimas , Sulfotransferasas , Humanos , Sulfotransferasas/metabolismo , Especificidad por Sustrato , Sitios de Unión , Isoenzimas/metabolismo , Arilsulfotransferasa/metabolismo
2.
J Biol Chem ; 296: 100094, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33485192

RESUMEN

Controlling unmodified serotonin levels in brain synapses is a primary objective when treating major depressive disorder-a disease that afflicts ∼20% of the world's population. Roughly 60% of patients respond poorly to first-line treatments and thus new therapeutic strategies are sought. To this end, we have constructed isoform-specific inhibitors of the human cytosolic sulfotransferase 1A3 (SULT1A3)-the isoform responsible for sulfonating ∼80% of the serotonin in the extracellular brain fluid. The inhibitor design includes a core ring structure, which anchors the inhibitor into a SULT1A3-specific binding pocket located outside the active site, and a side chain crafted to act as a latch to inhibit turnover by fastening down the SULT1A3 active-site cap. The inhibitors are allosteric, they bind with nanomolar affinity and are highly specific for the 1A3 isoform. The cap-stabilizing effects of the latch can be accurately calculated and are predicted to extend throughout the cap and into the surrounding protein. A free-energy correlation demonstrates that the percent inhibition at saturating inhibitor varies linearly with cap stabilization - the correlation is linear because the rate-limiting step of the catalytic cycle, nucleotide release, scales linearly with the fraction of enzyme in the cap-open form. Inhibitor efficacy in cultured cells was studied using a human mammary epithelial cell line that expresses SULT1A3 at levels comparable with those found in neurons. The inhibitors perform similarly in ex vivo and in vitro studies; consequently, SULT1A3 turnover can now be potently suppressed in an isoform-specific manner in human cells.


Asunto(s)
Células Epiteliales/metabolismo , Neurotransmisores/metabolismo , Sitio Alostérico , Arilsulfotransferasa/metabolismo , Catecolaminas/metabolismo , Trastorno Depresivo Mayor/metabolismo , Humanos , Cinética , Simulación de Dinámica Molecular , Estructura Molecular , Serotonina/metabolismo , Relación Estructura-Actividad , Sulfotransferasas/metabolismo
3.
Arch Toxicol ; 96(2): 673-687, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34921608

RESUMEN

Breast cancer etiology is associated with both proliferation and DNA damage induced by estrogens. Breast cancer risk factors (BCRF) such as body mass index (BMI), smoking, and intake of estrogen-active drugs were recently shown to influence intratissue estrogen levels. Thus, the aim of the present study was to investigate the influence of BCRF on estrogen-induced proliferation and DNA damage in 41 well-characterized breast glandular tissues derived from women without breast cancer. Influence of intramammary estrogen levels and BCRF on estrogen receptor (ESR) activation, ESR-related proliferation (indicated by levels of marker transcripts), oxidative stress (indicated by levels of GCLC transcript and oxidative derivatives of cholesterol), and levels of transcripts encoding enzymes involved in estrogen biotransformation was identified by multiple linear regression models. Metabolic fluxes to adducts of estrogens with DNA (E-DNA) were assessed by a metabolic network model (MNM) which was validated by comparison of calculated fluxes with data on methoxylated and glucuronidated estrogens determined by GC- and UHPLC-MS/MS. Intratissue estrogen levels significantly influenced ESR activation and fluxes to E-DNA within the MNM. Likewise, all BCRF directly and/or indirectly influenced ESR activation, proliferation, and key flux constraints influencing E-DNA (i.e., levels of estrogens, CYP1B1, SULT1A1, SULT1A2, and GSTP1). However, no unambiguous total effect of BCRF on proliferation became apparent. Furthermore, BMI was the only BCRF to indeed influence fluxes to E-DNA (via congruent adverse influence on levels of estrogens, CYP1B1 and SULT1A2).


Asunto(s)
Neoplasias de la Mama/metabolismo , Daño del ADN , Estrógenos/metabolismo , Glándulas Mamarias Humanas/metabolismo , Adulto , Arilsulfotransferasa/metabolismo , Índice de Masa Corporal , Neoplasias de la Mama/etiología , Proliferación Celular/fisiología , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP1B1/metabolismo , Femenino , Humanos , Glándulas Mamarias Humanas/patología , Estrés Oxidativo/fisiología , Factores de Riesgo , Espectrometría de Masas en Tándem
4.
Int J Mol Sci ; 22(4)2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33578912

RESUMEN

Endogenous factors involved in the progression of cisplatin nephropathy remain undetermined. Here, we demonstrate the toxico-pathological roles of indoxyl sulfate (IS), a sulfate-conjugated uremic toxin, and sulfotransferase 1A1 (SULT1A1), an enzyme involved in its synthesis, in cisplatin-induced acute kidney injury using Sult1a1-deficient (Sult1a1-/- KO) mice. With cisplatin administration, severe kidney dysfunction, tissue damage, and apoptosis were attenuated in Sult1a1-/- (KO) mice. Aryl hydrocarbon receptor (AhR) expression was increased by treatment with cisplatin in mouse kidney tissue. Moreover, the downregulation of antioxidant stress enzymes in wild-type (WT) mice was not observed in Sult1a1-/- (KO) mice. To investigate the effect of IS on the reactive oxygen species (ROS) levels, HK-2 cells were treated with cisplatin and IS. The ROS levels were significantly increased compared to cisplatin or IS treatment alone. IS-induced increases in ROS were reversed by downregulation of AhR, xanthine oxidase (XO), and NADPH oxidase 4 (NOX4). These findings suggest that SULT1A1 plays toxico-pathological roles in the progression of cisplatin-induced acute kidney injury, while the IS/AhR/ROS axis brings about oxidative stress.


Asunto(s)
Lesión Renal Aguda/inducido químicamente , Antineoplásicos/efectos adversos , Arilsulfotransferasa/genética , Cisplatino/efectos adversos , Indicán/metabolismo , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Arilsulfotransferasa/metabolismo , Línea Celular , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo
5.
Bull Exp Biol Med ; 170(5): 645-648, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788099

RESUMEN

The study examined peculiarities of immune regulation and associated polymorphic variants of candidate genes in men with atherosclerosis in Perm region. The revealed deficiency of CD127 lymphocytes and Annexin V-FITC+7AAD- cells, as well as enhanced level of CD3+CD4+ lymphocytes against the background of variant alleles of candidate genes FAS (rs1159120), CPOX (rs1131857) and wild-type alleles SULT1A1 (rs9282861), MMP9 (rs17576) are responsible for peculiar features of hereditary determination and pathogenesis of atherosclerosis in examined sample (p<0.05). The genetically determined degradation of extracellular matrix in vascular wall and implication of regulated Fas/APO1 apoptosis in the development of progressive atherosclerotic lesions indicate important role of immune system in atherogenesis. The revealed immunological and genetic features are recommended as the markers for early diagnosis of atherosclerosis and its prevention in men of Perm region.


Asunto(s)
Aterosclerosis/genética , Polimorfismo Genético/genética , Adulto , Alelos , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Aterosclerosis/inmunología , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Humanos , Linfocitos/metabolismo , Masculino , Persona de Mediana Edad
6.
J Biol Chem ; 294(7): 2293-2301, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30545938

RESUMEN

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.


Asunto(s)
Arilsulfotransferasa/química , Neurotransmisores/química , Regulación Alostérica , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Dominio Catalítico , Humanos , Neurotransmisores/genética , Neurotransmisores/metabolismo , Resonancia Magnética Nuclear Biomolecular , Estructura Secundaria de Proteína , Marcadores de Spin
7.
Breast Cancer Res ; 22(1): 80, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32727562

RESUMEN

BACKGROUND: The estrogen receptor (ER)-positive breast cancer represents over 80% of all breast cancer cases. Even though adjuvant hormone therapy with tamoxifen (TMX) is saving lives of patients with ER-positive breast cancer, the acquired resistance to TMX anti-estrogen therapy is the main hurdle for successful TMX therapy. Here we address the mechanism for TMX resistance and explore the ways to eradicate TMX-resistant breast cancer in both in vitro and ex vivo experiments. EXPERIMENTAL DESIGN: To identify compounds able to overcome TMX resistance, we used short-term and long-term viability assays in cancer cells in vitro and in patient samples in 3D ex vivo, analysis of gene expression profiles and cell line pharmacology database, shRNA screen, CRISPR-Cas9 genome editing, real-time PCR, immunofluorescent analysis, western blot, measurement of oxidative stress using flow cytometry, and thioredoxin reductase 1 enzymatic activity. RESULTS: Here, for the first time, we provide an ample evidence that a high level of the detoxifying enzyme SULT1A1 confers resistance to TMX therapy in both in vitro and ex vivo models and correlates with TMX resistance in metastatic samples in relapsed patients. Based on the data from different approaches, we identified three anticancer compounds, RITA (Reactivation of p53 and Induction of Tumor cell Apoptosis), aminoflavone (AF), and oncrasin-1 (ONC-1), whose tumor cell inhibition activity is dependent on SULT1A1. We discovered thioredoxin reductase 1 (TrxR1, encoded by TXNRD1) as a target of bio-activated RITA, AF, and ONC-1. SULT1A1 depletion prevented the inhibition of TrxR1, induction of oxidative stress, DNA damage signaling, and apoptosis triggered by the compounds. Notably, RITA efficiently suppressed TMX-unresponsive patient-derived breast cancer cells ex vivo. CONCLUSION: We have identified a mechanism of resistance to TMX via hyperactivated SULT1A1, which renders selective vulnerability to anticancer compounds RITA, AF, and ONC-1, and provide a rationale for a new combination therapy to overcome TMX resistance in breast cancer patients. Our novel findings may provide a strategy to circumvent TMX resistance and suggest that this approach could be developed further for the benefit of relapsed breast cancer patients.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/farmacología , Tamoxifeno/farmacología , Antineoplásicos Hormonales/química , Antineoplásicos Hormonales/farmacología , Apoptosis , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Humanos , Tamoxifeno/química , Células Tumorales Cultivadas
8.
Biochem Biophys Res Commun ; 528(4): 691-697, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32513533

RESUMEN

Bisphenol A (BPA) is one of the most common toxic endocrine disruptors in the environment. A fast, efficient and environmental-friendly method for BPA detoxification is urgently needed. In this study, we show that the enzymatic transformation of BPA into a non-estrogenic BPA sulfate can be performed by the aryl sulfotransferase (ASTB) from Desulfitobacterium hafniense. We developed and compared two Escherichia coli ASTB cell-surface displaying systems using the outer membrane porin F (OprF) and the lipoprotein outer membrane A (Lpp-OmpA) as carriers. The surface localization of both fusion proteins was confirmed by Western blot and flow cytometry analysis as well as the enzymatic activity assay of the outer membrane fractions. Unfortunately, Lpp-OmpA-ASTB cells had an adverse effect on cell growth. In contrast, the OprF-ASTB cell biocatalyst was stable, expressing 70% of enzyme activity for 7 days. It also efficiently sulfated 90% of 5 mM BPA (1 mg/mL) in wastewater within 6 h.


Asunto(s)
Arilsulfotransferasa/metabolismo , Compuestos de Bencidrilo/metabolismo , Desulfitobacterium/enzimología , Disruptores Endocrinos/metabolismo , Fenoles/metabolismo , Contaminantes Químicos del Agua/metabolismo , Compuestos de Bencidrilo/aislamiento & purificación , Biotransformación , Disruptores Endocrinos/aislamiento & purificación , Escherichia coli/enzimología , Fenoles/aislamiento & purificación , Contaminantes Químicos del Agua/aislamiento & purificación , Purificación del Agua/métodos
9.
Drug Metab Dispos ; 48(5): 337-344, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32152050

RESUMEN

Sulfotransferase (SULT) 4A1 is a brain-selective sulfotransferase-like protein that has recently been shown to be essential for normal neuronal development in mice. In the present study, SULT4A1 was found to colocalize with SULT1A1/3 in human brain neurons. Using immunoprecipitation, SULT4A1 was shown to interact with both SULT1A1 and SULT1A3 when expressed in human cells. Mutation of the conserved dimerization motif located in the C terminus of the sulfotransferases prevented this interaction. Both ectopically expressed and endogenous SULT4A1 decreased SULT1A1/3 protein levels in neuronal cells, and this was also prevented by mutation of the dimerization motif. During differentiation of neuronal SH-SY5Y cells, there was a loss in SULT1A1/3 protein but an increase in SULT4A1 protein. This resulted in an increase in the toxicity of dopamine, a substrate for SULT1A3. Inhibition of SULT4A1 using small interference RNA abrogated the loss in SULT1A1/3 and reversed dopamine toxicity. These results show a reciprocal relationship between SULT4A1 and the other sulfotransferases, suggesting that it may act as a chaperone to control the expression of SULT1A1/3 in neuronal cells. SIGNIFICANCE STATEMENT: The catalytically inactive sulfotransferase (SULT) 4A1 may regulate the function of other SULTs by interacting with them via a conserved dimerization motif. In neuron-like cells, SULT4A1 is able to modulate dopamine toxicity by interacting with SULT1A3, potentially decreasing the metabolism of dopamine.


Asunto(s)
Arilsulfotransferasa/genética , Encéfalo/enzimología , Regulación del Desarrollo de la Expresión Génica , Sulfotransferasas/metabolismo , Arilsulfotransferasa/metabolismo , Encéfalo/citología , Diferenciación Celular , Línea Celular Tumoral , Dopamina/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Mutación , Neuronas/enzimología , Multimerización de Proteína/genética , Sulfotransferasas/genética
10.
Pharmacogenet Genomics ; 29(5): 99-105, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31145702

RESUMEN

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.


Asunto(s)
Albuterol/metabolismo , Arilsulfotransferasa/genética , Fenilefrina/metabolismo , Sulfotransferasas/genética , Albuterol/uso terapéutico , Arilsulfotransferasa/química , Arilsulfotransferasa/metabolismo , Asma/tratamiento farmacológico , Asma/genética , Genotipo , Humanos , Hipotensión/tratamiento farmacológico , Hipotensión/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Mutagénesis Sitio-Dirigida , Fenilefrina/uso terapéutico , Polimorfismo de Nucleótido Simple/genética , Sulfatos/metabolismo , Sulfotransferasas/química , Sulfotransferasas/metabolismo
11.
Electrophoresis ; 40(18-19): 2271-2276, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30882918

RESUMEN

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.


Asunto(s)
Arilsulfotransferasa , Electroforesis Capilar/métodos , Enzimas Inmovilizadas , Imanes , Acetaminofén/análogos & derivados , Acetaminofén/análisis , Acetaminofén/metabolismo , Arilsulfotransferasa/química , Arilsulfotransferasa/metabolismo , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Límite de Detección , Modelos Lineales , Microesferas , Reproducibilidad de los Resultados
12.
Appl Microbiol Biotechnol ; 103(9): 3761-3771, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30830250

RESUMEN

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).


Asunto(s)
Arilsulfotransferasa/genética , Proteínas Bacterianas/genética , Catecoles/metabolismo , Desulfitobacterium/enzimología , Sulfatos/metabolismo , Ampirona/química , Ampirona/metabolismo , Arilsulfotransferasa/química , Arilsulfotransferasa/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Catecoles/química , Desulfitobacterium/química , Desulfitobacterium/genética , Evolución Molecular Dirigida , Cinética , Espectroscopía de Resonancia Magnética , Especificidad por Sustrato , Sulfatos/química
13.
Xenobiotica ; 49(3): 270-275, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29436892

RESUMEN

Sulfotransferase (SULT) has been found in the brain; however, the details of its function remain unclear. The present study aimed to elucidate the regional differences in the expression of SULT1 and SULT2 mRNA and SULT activities in the eight functional regions of the rat brain (cerebellum, cortex, hippocampus, medulla oblongata, midbrain, olfactory bulb, striatum, and thalamus). All SULT1 isoforms were detected in the medulla oblongata and thalamus. SULT2A1 mRNA was not observed in any of the eight regions, whereas SULT2B1a and SULT2B1b were found in all regions. The SULT2B1b mRNA expression level in the medulla oblongata was 1.7-fold higher than that in the liver. The sulfonation of p-nitrophenol and pregnenolone was detected in all regions. The kinetics of p-nitrophenol sulfonation in the cerebellum fitted to the substrate inhibition model (Km = 37.6 nM, Vmax = 2.72 pmol/min/mg, Vinh = 1.60 pmol/min/mg, and Ki = 0.87 µM). The pregnenolone sulfonation also exhibited substrate inhibition kinetics (Km = 0.99 µM, Vmax = 1.53 pmol/min/mg, and Ki = 54.67 µM). We clarified that SULT1 and SULT2 were expressed and had metabolizing capacities in the rat brain, suggesting that brain SULTs may be involved in metabolism of endogenous compounds and drugs.


Asunto(s)
Arilsulfotransferasa/metabolismo , Encéfalo/enzimología , Animales , Arilsulfotransferasa/genética , Cinética , Hígado/metabolismo , Masculino , ARN Mensajero/metabolismo , Ratas Sprague-Dawley
14.
Arch Toxicol ; 93(6): 1515-1527, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30993378

RESUMEN

We previously showed that purified 1-methoxy-3-indolylmethyl (1-MIM) glucosinolate, a secondary plant metabolite in Brassica species, is mutagenic in various in vitro systems and forms DNA and protein adducts in mouse models. In the present study, we administered 1-MIM glucosinolate in a natural matrix to mice, by feeding a diet containing pak choi powder and extract. Groups of animals were killed after 1, 2, 4 and 8 days of pak choi diet, directly or, in the case of the 8-day treatment, after 0, 8 and 16 days of recovery with pak choi-free diet. DNA adducts [N2-(1-MIM)-dG, N6-(1-MIM)-dA] in six tissues, as well as protein adducts [τN-(1-MIM)-His] in serum albumin (SA) and hemoglobin (Hb) were determined using UPLC-MS/MS with isotopically labeled internal standards. None of the samples from the 12 control animals under standard diet contained any 1-MIM adducts. All groups receiving pak choi diet showed DNA adducts in all six tissues (exception: lung of mice treated for a single day) as well as SA and Hb adducts. During the feeding period, all adduct levels continuously increased until day 8 (in the jejunum until day 4). During the 14-day recovery period, N2-(1-MIM)-dG in liver, kidney, lung, jejunum, cecum and colon decreased to 52, 41, 59, 11, 7 and 2%, respectively, of the peak level. The time course of N6-(1-MIM)-dA was similar. Immunohistochemical analyses indicated that cell turnover is a major mechanism of DNA adduct elimination in the intestine. In the same recovery period, protein adducts decreased more rapidly in SA than in Hb, to 0.7 and 37%, respectively, of the peak level, consistent with the differential turnover of these proteins. In conclusion, the pak choi diet lead to the formation of high levels of adducts in mice. Cell and protein turnover was a major mechanism of adduct elimination, at least in gut and blood.


Asunto(s)
Proteínas Sanguíneas/análisis , Brassica/química , Aductos de ADN/análisis , Dieta , Glucosinolatos/análisis , Indoles/análisis , Animales , Arilsulfotransferasa/análisis , Arilsulfotransferasa/metabolismo , Peso Corporal/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Hemoglobinas/análisis , Masculino , Espectrometría de Masas , Ratones , Albúmina Sérica/análisis , Espectrometría de Masas en Tándem , Distribución Tisular
15.
PLoS Genet ; 12(2): e1005709, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26872257

RESUMEN

The 16p11.2 600 kb BP4-BP5 deletion and duplication syndromes have been associated with developmental delay; autism spectrum disorders; and reciprocal effects on the body mass index, head circumference and brain volumes. Here, we explored these relationships using novel engineered mouse models carrying a deletion (Del/+) or a duplication (Dup/+) of the Sult1a1-Spn region homologous to the human 16p11.2 BP4-BP5 locus. On a C57BL/6N inbred genetic background, Del/+ mice exhibited reduced weight and impaired adipogenesis, hyperactivity, repetitive behaviors, and recognition memory deficits. In contrast, Dup/+ mice showed largely opposite phenotypes. On a F1 C57BL/6N × C3B hybrid genetic background, we also observed alterations in social interaction in the Del/+ and the Dup/+ animals, with other robust phenotypes affecting recognition memory and weight. To explore the dosage effect of the 16p11.2 genes on metabolism, Del/+ and Dup/+ models were challenged with high fat and high sugar diet, which revealed opposite energy imbalance. Transcriptomic analysis revealed that the majority of the genes located in the Sult1a1-Spn region were sensitive to dosage with a major effect on several pathways associated with neurocognitive and metabolic phenotypes. Whereas the behavioral consequence of the 16p11 region genetic dosage was similar in mice and humans with activity and memory alterations, the metabolic defects were opposite: adult Del/+ mice are lean in comparison to the human obese phenotype and the Dup/+ mice are overweight in comparison to the human underweight phenotype. Together, these data indicate that the dosage imbalance at the 16p11.2 locus perturbs the expression of modifiers outside the CNV that can modulate the penetrance, expressivity and direction of effects in both humans and mice.


Asunto(s)
Deleción Cromosómica , Duplicación Cromosómica/genética , Cognición , Adiposidad , Alelos , Animales , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Conducta Animal , Peso Corporal , Encéfalo/metabolismo , Encéfalo/fisiopatología , Cromosomas de los Mamíferos/genética , Anomalías Craneofaciales/genética , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Reordenamiento Génico/genética , Hipocampo/fisiopatología , Memoria , Ratones Endogámicos C57BL , Actividad Motora , Fenotipo , Transmisión Sináptica/genética , Síndrome , Destete
16.
Proc Natl Acad Sci U S A ; 113(50): 14312-14317, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911811

RESUMEN

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.


Asunto(s)
Arilsulfotransferasa/química , Arilsulfotransferasa/metabolismo , Catequina/metabolismo , Sitio Alostérico , Arilsulfotransferasa/genética , Catequina/análogos & derivados , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Marcadores de Spin , Especificidad por Sustrato
17.
Arch Biochem Biophys ; 648: 44-52, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29705271

RESUMEN

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.


Asunto(s)
Acetaminofén/metabolismo , Analgésicos Opioides/metabolismo , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Citosol/enzimología , Polimorfismo de Nucleótido Simple , Sulfatos/metabolismo , Sulfotransferasas/genética , Arilsulfotransferasa/química , Humanos , Cinética , Modelos Moleculares , Conformación Proteica
18.
Biotechnol Bioeng ; 115(5): 1106-1115, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29288579

RESUMEN

Bacterial aryl sulfotransferases (AST) utilize p-nitrophenylsulfate (pNPS) as a phenolic donor to sulfurylate typically a phenolic acceptor. Interest in aryl sulfotransferases is growing because of their broad variety of acceptors and cost-effective sulfuryl-donors. For instance, aryl sulfotransferase A (ASTA) from Desulfitobacterium hafniense was recently reported to sulfurylate d-glucose. In this study, a directed evolution protocol was developed and validated for aryl sulfotransferase B (ASTB). Thereby the well-known pNPS quantification system was advanced to operate efficiently as a continuous screening system in 96-well MTP format with a true coefficient of variation of 14.3%. A random mutagenesis library (SeSaM library) of ASTB was screened (1,760 clones) to improve sulfurylation of the carbohydrate building block N-acetylglucosamine (GlcNAc). The beneficial variant ASTB-V1 (Val579Asp) showed an up to 3.4-fold increased specific activity toward GlcNAc when compared to ASTB-WT. HPLC- and MS-analysis confirmed ASTB-V1's increased GlcNAc monosulfurylation (2.4-fold increased product formation) representing the validation of the first successful directed evolution round of an AST for a saccharide substrate.


Asunto(s)
Acetilglucosamina/metabolismo , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Desulfitobacterium/enzimología , Evolución Molecular Dirigida/métodos , Pruebas Genéticas , Mutagénesis
19.
Biotechnol Bioeng ; 115(6): 1561-1570, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29484646

RESUMEN

Chondroitin sulfate has been widely used in both medical and clinical applications. Commercial chondroitin sulfate has been mainly acquired from animal tissue extraction. Here we report a new two-step biological strategy for producing chondroitin sulfate A and chondroitin sulfate C. First, the chondroitin biosynthesis pathway in a recombinant Bacillus subtilis strain using sucrose as carbon source was systematically optimized and the titer of chondroitin was significantly enhanced to 7.15 g/L. Then, specific sulfation transformation systems were successfully constructed and optimized by combining the purified aryl sulfotransferase IV (ASST IV), chondroitin 4-sulfotransferase (C4ST) and chondroitin 6-sulfotransferase (C6ST). Chondroitin sulfate A and C were enzymatically transformed from chondroitin at conversion rates of 98% and 96%, respectively. The present biological strategy has great potential to be scaled up for biosynthesis of chondroitin sulfate A and C from cheap carbon sources.


Asunto(s)
Arilsulfotransferasa/metabolismo , Bacillus subtilis/metabolismo , Biotecnología/métodos , Sulfatos de Condroitina/metabolismo , Sulfotransferasas/metabolismo , Bacillus subtilis/crecimiento & desarrollo , Carbono/metabolismo , Medios de Cultivo/química , Ingeniería Metabólica/métodos , Sacarosa/metabolismo , Carbohidrato Sulfotransferasas
20.
Hum Reprod ; 32(7): 1465-1473, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28482050

RESUMEN

STUDY QUESTION: Are bisphenol A (BPA) and BPA analogs (BPA-A) safe for male human reproductive function? SUMMARY ANSWER: The endocrine function of human testes explants [assessed by measuring testosterone and insulin-like factor 3 (INSL3)] was impacted by exposure of the human adult testis explants to BPA/BPA-A. WHAT IS KNOWN ALREADY: The few epidemiologic studies performed suggest that bisphenols have potential endocrine disruptive properties, but they did not identify clear and direct patterns of endocrine disruption. STUDY DESIGN, SIZE, DURATION: Adult human testis explants in culture were exposed to BPA and the analogs bisphenol F (BPF), bisphenol S (BPS), bisphenol E (BPE), bisphenol B (BPB) and bisphenol A diglycidyl ether (BADGE) at 10-9-10-5 M for 24 or 48 h. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human adult testes were obtained from prostate cancer patients who had no hormone therapy, or from multiorgan donors. After ex vivo exposure to the investigated bisphenols, the measured outcomes were related to histopathology (gross morphology and germ cell viability determined by anti-caspase three immunohistochemistry), and the levels of testosterone, INSL3 and inhibin B were measured using immunoassays. The levels of mRNA encoding key enzymes of bisphenol biotransformation were investigated by quantitative PCR: UGT2B15 UDP (glucuronosyltransferase two family, polypeptide B15), GUSB (glucuronidase beta), SULT1A1 and 3 (sulfotransferase family 1 A member 1 and 3) and STS (steroid sulfatase). MAIN RESULTS AND THE ROLE OF CHANCE: A significant dose-dependent inhibition was found between testosterone levels measured in the culture medium and concentrations of BPA (P = 0.00778 at 24 h and P = 0.0291 at 48 h), BPE (P = 0.039) and BPF (P = 0.00663). The observed BPA and BPA-A-induced inhibition of testosterone production varied according to duration of exposure and BPA/BPA-A concentrations. BPA (10-9 M; P < 0.05), BPB (10-9 M; P < 0.05), BPS (10-9 and 10-8 M; P < 0.05) and BADGE (10-5 M; P < 0.05) increased Leydig cell INSL3 production. By contrast, BPE dose dependently inhibited INSL3 (P = 0.0372). Conversely, Sertoli cell function (inhibin B) and germ cell viability were not significantly affected by either bisphenols. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Environmental compounds cannot be deliberately administered to men, justifying the use of an ex vivo approach. A relatively low number of testes samples were available for analysis (n = 3, except for testosterone secretion with n = 5). The active concentrations of BPA and BPA-A used in the study were higher than those found in human biological fluids. WIDER IMPLICATIONS OF THE FINDINGS: Under our experimental conditions, direct exposure to BPA or BPA-A can result in endocrine disturbance in the adult human testis. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by Inserm (Institut National de la Santé et de la Recherche Médicale), EHESP-School of Public Health, University of Rennes1, by grants from the Agence Nationale de la Recherche (ANR; grant#ANR-13-CESA-0012-03 NEWPLAST) and Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES; grant#EST-2010/2/046 (BPATESTIS)). All authors declare they have no current or potential competing financial interests.


Asunto(s)
Compuestos de Bencidrilo/toxicidad , Disruptores Endocrinos/toxicidad , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Insulina/agonistas , Antiandrógenos no Esteroides/toxicidad , Fenoles/toxicidad , Proteínas/agonistas , Testículo/efectos de los fármacos , Testosterona/antagonistas & inhibidores , Adulto , Apoptosis/efectos de los fármacos , Arilsulfotransferasa/genética , Arilsulfotransferasa/metabolismo , Compuestos de Bencidrilo/química , Disruptores Endocrinos/química , Compuestos Epoxi/toxicidad , Glucuronidasa/genética , Glucuronidasa/metabolismo , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Humanos , Insulina/metabolismo , Células Intersticiales del Testículo/citología , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/metabolismo , Masculino , Antiandrógenos no Esteroides/química , Fenoles/química , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Reproducibilidad de los Resultados , Células de Sertoli/citología , Células de Sertoli/efectos de los fármacos , Células de Sertoli/metabolismo , Esteril-Sulfatasa/genética , Esteril-Sulfatasa/metabolismo , Sulfonas/toxicidad , Testículo/citología , Testículo/metabolismo , Testosterona/metabolismo , Técnicas de Cultivo de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA