Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 17(6): 684-692, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846619

RESUMEN

Heparan sulfate (HS) proteoglycans bind extracellular proteins that participate in cell signaling, attachment and endocytosis. These interactions depend on the arrangement of sulfated sugars in the HS chains generated by well-characterized biosynthetic enzymes; however, the regulation of these enzymes is largely unknown. We conducted genome-wide CRISPR-Cas9 screens with a small-molecule ligand that binds to HS. Screening of A375 melanoma cells uncovered additional genes and pathways impacting HS formation. The top hit was the epigenetic factor KDM2B, a histone demethylase. KDM2B inactivation suppressed multiple HS sulfotransferases and upregulated the sulfatase SULF1. These changes differentially affected the interaction of HS-binding proteins. KDM2B-deficient cells displayed decreased growth rates, which was rescued by SULF1 inactivation. In addition, KDM2B deficiency altered the expression of many extracellular matrix genes. Thus, KDM2B controls proliferation of A375 cells through the regulation of HS structure and serves as a master regulator of the extracellular matrix.


Asunto(s)
Proteínas F-Box/antagonistas & inhibidores , Estudio de Asociación del Genoma Completo , Heparitina Sulfato/metabolismo , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Algoritmos , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Matriz Extracelular/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Unión Proteica/genética , RNA-Seq , Sulfotransferasas/antagonistas & inhibidores
2.
J Biol Chem ; 296: 100353, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33524392

RESUMEN

Polychlorinated bisphenols (PCBs) continue to contaminate food chains globally where they concentrate in tissues and disrupt the endocrine systems of species throughout the ecosphere. Hydroxylated PCBs (OH-PCBs) are major PCB metabolites and high-affinity inhibitors of human estrogen sulfotransferase (SULT1E1), which sulfonates estrogens and thus prevents them from binding to and activating their receptors. OH-PCB inhibition of SULT1E1 is believed to contribute significantly to PCB-based endocrine disruption. Here, for the first time, the molecular basis of OH-PCB inhibition of SULT1E1 is revealed in a structure of SULT1E1 in complex with OH-PCB1 (4'-OH-2,6-dichlorobiphenol) and its substrates, estradiol (E2), and PAP (3'-phosphoadenosine-5-phosphosulfate). OH-PCB1 prevents catalysis by intercalating between E2 and catalytic residues and establishes a new E2-binding site whose E2 affinity and positioning are greater than and competitive with those of the reactive-binding pocket. Such complexes have not been observed previously and offer a novel template for the design of high-affinity inhibitors. Mutating residues in direct contact with OH-PCB weaken its affinity without compromising the enzyme's catalytic parameters. These OH-PCB resistant mutants were used in stable transfectant studies to demonstrate that OH-PCBs regulate estrogen receptors in cultured human cell lines by binding the OH-PCB binding pocket of SULT1E1.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Estrógenos/farmacología , Bifenilos Policlorados/farmacología , Sulfotransferasas/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Estrógenos/química , Humanos , Hidroxilación , Modelos Moleculares , Bifenilos Policlorados/química , Receptores de Estrógenos/metabolismo , Sulfotransferasas/química , Sulfotransferasas/metabolismo
3.
Biochem J ; 478(4): 735-748, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33480417

RESUMEN

Sulfated carbohydrate metabolism is a fundamental process, which occurs in all domains of life. Carbohydrate sulfatases are enzymes that remove sulfate groups from carbohydrates and are essential to the depolymerisation of complex polysaccharides. Despite their biological importance, carbohydrate sulfatases are poorly studied and challenges remain in accurately assessing the enzymatic activity, specificity and kinetic parameters. Most notably, the separation of desulfated products from sulfated substrates is currently a time-consuming process. In this paper, we describe the development of rapid capillary electrophoresis coupled to substrate fluorescence detection as a high-throughput and facile means of analysing carbohydrate sulfatase activity. The approach has utility for the determination of both kinetic and inhibition parameters and is based on existing microfluidic technology coupled to a new synthetic fluorescent 6S-GlcNAc carbohydrate substrate. Furthermore, we compare this technique, in terms of both time and resources, to high-performance anion exchange chromatography and NMR-based methods, which are the two current 'gold standards' for enzymatic carbohydrate sulfation analysis. Our study clearly demonstrates the advantages of mobility shift assays for the quantification of near real-time carbohydrate desulfation by purified sulfatases, and will support the search for small molecule inhibitors of these disease-associated enzymes.


Asunto(s)
Electroforesis Capilar/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Fluorometría/métodos , Ensayos Analíticos de Alto Rendimiento/métodos , Técnicas Analíticas Microfluídicas/métodos , Sulfotransferasas/análisis , Proteínas Bacterianas/análisis , Proteínas Bacterianas/antagonistas & inhibidores , Bacteroides thetaiotaomicron/enzimología , Compuestos de Boro/análisis , Conformación de Carbohidratos , Cromatografía Líquida de Alta Presión , Cromatografía por Intercambio Iónico , Sistemas de Computación , Colorantes Fluorescentes/análisis , Glicosaminoglicanos/metabolismo , Cinética , Resonancia Magnética Nuclear Biomolecular , Proteínas Recombinantes/análisis , Especificidad por Sustrato , Sulfotransferasas/antagonistas & inhibidores
4.
Toxicol Appl Pharmacol ; 425: 115553, 2021 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-33915121

RESUMEN

p-Cresol sulfate, the primary metabolite of p-cresol, is a uremic toxin that has been associated with toxicities and mortalities. The study objectives were to i) characterize the contributions of human sulfotransferases (SULT) catalyzing p-cresol sulfate formation using multiple recombinant SULT enzymes (including the polymorphic variant SULT1A1*2), pooled human liver cytosols, and pooled human kidney cytosols; and ii) determine the potencies and mechanisms of therapeutic inhibitors capable of attenuating the production of p-cresol sulfate. Human recombinant SULT1A1 was the primary enzyme responsible for the formation of p-cresol sulfate (Km = 0.19 ±â€¯0.02 µM [with atypical kinetic behavior at lower substrate concentrations; see text discussion], Vmax = 789.5 ±â€¯101.7 nmol/mg/min, Ksi = 2458.0 ±â€¯332.8 µM, mean ±â€¯standard deviation, n = 3), while SULT1A3, SULT1B1, SULT1E1, and SULT2A1 contributed negligible or minor roles at toxic p-cresol concentrations. Moreover, human recombinant SULT1A1*2 exhibited reduced enzyme activities (Km = 81.5 ±â€¯31.4 µM, Vmax = 230.6 ±â€¯17.7 nmol/mg/min, Ksi = 986.0 ±â€¯434.4 µM) compared to the wild type. The sulfonation of p-cresol was characterized by Michaelis-Menten kinetics in liver cytosols (Km = 14.8 ±â€¯3.4 µM, Vmax = 1.5 ±â€¯0.2 nmol/mg/min) and substrate inhibition in kidney cytosols (Km = 0.29 ±â€¯0.02 µM, Vmax = 0.19 ±â€¯0.05 nmol/mg/min, Ksi = 911.7 ±â€¯278.4 µM). Of the 14 investigated therapeutic inhibitors, mefenamic acid (Ki = 2.4 ±â€¯0.1 nM [liver], Ki = 1.2 ±â€¯0.3 nM [kidney]) was the most potent in reducing the formation of p-cresol sulfate, exhibiting noncompetitive inhibition in human liver cytosols and recombinant SULT1A1, and mixed inhibition in human kidney cytosols. Our novel findings indicated that SULT1A1 contributed an important role in p-cresol sulfonation (hence it can be considered a probe reaction) in liver and kidneys, and mefenamic acid may be utilized as a potential therapeutic agent to attenuate the generation of p-cresol sulfate as an approach to detoxification.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Cresoles/metabolismo , Cresoles/toxicidad , Ácido Mefenámico/farmacología , Sulfotransferasas/metabolismo , Ésteres del Ácido Sulfúrico/metabolismo , Ésteres del Ácido Sulfúrico/toxicidad , Catálisis , Citosol/enzimología , Humanos , Riñón , Hígado , Proteínas Recombinantes , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/genética
5.
J Am Soc Nephrol ; 31(7): 1496-1508, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32424001

RESUMEN

BACKGROUND: Studies have suggested that estrogens may protect mice from AKI. Estrogen sulfotransferase (SULT1E1, or EST) plays an important role in estrogen homeostasis by sulfonating and deactivating estrogens, but studies on the role of SULT1E1 in AKI are lacking. METHODS: We used the renal ischemia-reperfusion model to investigate the role of SULT1E1 in AKI. We subjected wild-type mice, Sult1e1 knockout mice, and Sult1e1 knockout mice with liver-specific reconstitution of SULT1E1 expression to bilateral renal ischemia-reperfusion or sham surgery, either in the absence or presence of gonadectomy. We assessed relevant biochemical, histologic, and gene expression markers of kidney injury. We also used wild-type mice treated with the SULT1E1 inhibitor triclosan to determine the effect of pharmacologic inhibition of SULT1E1 on AKI. RESULTS: AKI induced the expression of Sult1e1 in a tissue-specific and sex-specific manner. It induced expression of Sult1e1 in the liver in both male and female mice, but Sult1e1 induction in the kidney occurred only in male mice. Genetic knockout or pharmacologic inhibition of Sult1e1 protected mice of both sexes from AKI, independent of the presence of sex hormones. Instead, a gene profiling analysis indicated that the renoprotective effect was associated with increased vitamin D receptor signaling. Liver-specific transgenic reconstitution of SULT1E1 in Sult1e1 knockout mice abolished the protection in male mice but not in female mice, indicating that Sult1e1's effect on AKI was also tissue-specific and sex-specific. CONCLUSIONS: SULT1E1 appears to have a novel function in the pathogenesis of AKI. Our findings suggest that inhibitors of SULT1E1 might have therapeutic utility in the clinical management of AKI.


Asunto(s)
Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/prevención & control , Hígado/metabolismo , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Lesión Renal Aguda/etiología , Animales , Calcitriol/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Orquiectomía , Ovariectomía , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Daño por Reperfusión/complicaciones , Factores Sexuales , Transducción de Señal , Sulfotransferasas/antagonistas & inhibidores , Triclosán/farmacología
6.
J Am Chem Soc ; 142(11): 5282-5292, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32083852

RESUMEN

Human endo-O-sulfatases (Sulf-1 and Sulf-2) are extracellular heparan sulfate proteoglycan (HSPG)-specific 6-O-endosulfatases, which regulate a multitude of cell-signaling events through heparan sulfate (HS)-protein interactions and are associated with the onset of osteoarthritis. These endo-O-sulfatases are transported onto the cell surface to liberate the 6-sulfate groups from the internal d-glucosamine residues in the highly sulfated subdomains of HSPGs. In this study, a variety of HS oligosaccharides with different chain lengths and N- and O-sulfation patterns via chemical synthesis were systematically studied about the substrate specificity of human Sulf-1 employing the fluorogenic substrate 4-methylumbelliferyl sulfate (4-MUS) in a competition assay. The trisaccharide sulfate IdoA2S-GlcNS6S-IdoA2S was found to be the minimal-size substrate for Sulf-1, and substitution of the sulfate group at the 6-O position of the d-glucosamine unit with the sulfonamide motif effectively inhibited the Sulf-1 activity with IC50 = 0.53 µM, Ki = 0.36 µM, and KD = 12 nM.


Asunto(s)
Inhibidores Enzimáticos/química , Sulfatasas/antagonistas & inhibidores , Sulfonamidas/química , Sulfotransferasas/antagonistas & inhibidores , Trisacáridos/química , Pruebas de Enzimas , Inhibidores Enzimáticos/síntesis química , Heparitina Sulfato/química , Humanos , Cinética , Especificidad por Sustrato , Sulfatasas/química , Sulfonamidas/síntesis química , Sulfotransferasas/química , Trisacáridos/síntesis química
7.
Med Sci Monit ; 26: e926780, 2020 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-33079922

RESUMEN

BACKGROUND Cervical cancer threatens women's health worldwide. Verteporfin (VP), a small-molecule YAP1 inhibitor, inhibits cancer cell growth. This study investigated whether VP could inhibit the proliferation and promote the apoptosis of cervical cancer cells by decreasing SULT2B1 expression. MATERIAL AND METHODS Normal and cancerous cervical cell proliferation after VP treatment was detected by CCK-8 assay. HeLa cell migration, invasion, and apoptosis after VP treatment and transfection were analyzed by wound healing assay, transwell assay, and TUNEL assay, respectively. The expression of related proteins was determined by western blot analysis. Western blot and RT-qPCR analysis detected mRNA and protein expression of SULT2B1. RESULTS Different VP concentrations (0.5, 1, 2, and 5 µM) inhibited the viability of HeLa cells and had no obvious effect on H8 cells. Therefore, 5 µM VP was selected for subsequent experiments. VP inhibited the proliferation, migration, and invasion of HeLa cells and promoted their apoptosis. Bcl-2 expression decreased, and expression of Bax, caspase-3, and caspase-9 in VP-treated HeLa cells increased. SULT2B1 expression increased in cervical cancer cells compared with normal cervical cells. Furthermore, SULT2B1 expression increased in HeLa cells and VP suppressed SULT2B1 expression. SULT2B1 overexpression reduced the inhibiting effect of VP on the proliferation, migration, and apoptosis of HeLa cells, and reduced VP effect on apoptosis of HeLa cells. SULT2B1 overexpression upregulated the Bcl-2 expression and downregulated the expression of Bax, caspase-3, and caspase-9 in VP-treated HeLa cells. CONCLUSIONS VP inhibited the proliferation, migration, and invasion and promoted apoptosis of cervical cancer cells by decreasing SULT2B1 expression.


Asunto(s)
Antineoplásicos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sulfotransferasas , Neoplasias del Cuello Uterino/tratamiento farmacológico , Verteporfina , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Movimiento Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Invasividad Neoplásica , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/metabolismo , Neoplasias del Cuello Uterino/patología , Verteporfina/farmacología
8.
J Enzyme Inhib Med Chem ; 35(1): 1503-1512, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32657203

RESUMEN

Metachromatic leukodystrophy (MLD) is a rare genetic disease characterised by a dysfunction of the enzyme arylsulphatase A leading to the lysosomal accumulation of cerebroside sulphate (sulphatide) causing subsequent demyelination in patients. The enzyme galactosylceramide (cerebroside) sulphotransferase (CST) catalyses the transfer of a sulphate group from 3'-phosphoadenosine-5'-phosphosulphate (PAPS) to cerebrosides producing sulphatides. Substrate reduction therapy for arylsulphatase A by inhibition of CST was proposed as a promising therapeutic approach. To identify competitive CST inhibitors, we synthesised and investigated analogues of the substrate galactosylceramide with variations at the anomeric position, the acyl substituent and the carbohydrate moiety, and investigated their structure-activity relationships. While most of the compounds behaved as substrates, α-galactosylceramide 16 was identified as the first competitive CST inhibitor. Compound 16 can serve as a new lead structure for the development of drugs for the treatment of this devastating disease, MLD, for which small molecule therapeutics are currently not available.


Asunto(s)
Cerebrósidos/farmacología , Descubrimiento de Drogas , Leucodistrofia Metacromática/tratamiento farmacológico , Sulfotransferasas/antagonistas & inhibidores , Cerebrósidos/síntesis química , Cerebrósidos/química , Relación Dosis-Respuesta a Droga , Humanos , Leucodistrofia Metacromática/enzimología , Estructura Molecular , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , Sulfotransferasas/genética , Sulfotransferasas/metabolismo
9.
J Biol Chem ; 293(27): 10826-10840, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29752409

RESUMEN

Transcellular propagation of protein aggregate "seeds" has been proposed to mediate the progression of neurodegenerative diseases in tauopathies and α-synucleinopathies. We previously reported that tau and α-synuclein aggregates bind heparan sulfate proteoglycans (HSPGs) on the cell surface, promoting cellular uptake and intracellular seeding. However, the specificity and binding mode of these protein aggregates to HSPGs remain unknown. Here, we measured direct interaction with modified heparins to determine the size and sulfation requirements for tau, α-synuclein, and ß-amyloid (Aß) aggregate binding to glycosaminoglycans (GAGs). Varying the GAG length and sulfation patterns, we next conducted competition studies with heparin derivatives in cell-based assays. Tau aggregates required a precise GAG architecture with defined sulfate moieties in the N- and 6-O-positions, whereas the binding of α-synuclein and Aß aggregates was less stringent. To determine the genes required for aggregate uptake, we used CRISPR/Cas9 to individually knock out the major genes of the HSPG synthesis pathway in HEK293T cells. Knockouts of the extension enzymes exostosin 1 (EXT1), exostosin 2 (EXT2), and exostosin-like 3 (EXTL3), as well as N-sulfotransferase (NDST1) or 6-O-sulfotransferase (HS6ST2) significantly reduced tau uptake, consistent with our biochemical findings, and knockouts of EXT1, EXT2, EXTL3, or NDST1, but not HS6ST2 reduced α-synuclein uptake. In summary, tau aggregates display specific interactions with HSPGs that depend on GAG length and sulfate moiety position, whereas α-synuclein and Aß aggregates exhibit more flexible interactions with HSPGs. These principles may inform the development of mechanism-based therapies to block transcellular propagation of amyloid protein-based pathologies.


Asunto(s)
Péptidos beta-Amiloides/química , Glicosaminoglicanos/química , Proteoglicanos de Heparán Sulfato/metabolismo , Azufre/metabolismo , Tauopatías/patología , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo , Péptidos beta-Amiloides/metabolismo , Sistemas CRISPR-Cas , Glicosaminoglicanos/metabolismo , Humanos , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Tauopatías/metabolismo
10.
J Pharmacol Exp Ther ; 369(3): 389-405, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30918069

RESUMEN

Lithocholic acid (LCA) is a bile acid associated with adverse effects, including cholestasis, and it exists in vivo mainly as conjugates known as glyco-LCA (GLCA) and tauro-LCA (TLCA). Tamoxifen has been linked to the development of cholestasis, and it inhibits sulfotransferase 2A1 (SULT2A1)-catalyzed dehydroepiandrosterone (DHEA) sulfonation. The present study was done to characterize the sulfonation of LCA, GLCA, and TLCA and to investigate whether triphenylethylene (clomifene, tamoxifen, toremifene, ospemifene, droloxifene), benzothiophene (raloxifene, arzoxifene), tetrahydronaphthalene (lasofoxifene, nafoxidine), indole (bazedoxifene), and benzopyran (acolbifene) classes of selective estrogen receptor modulator (SERM) inhibit LCA, GLCA, and TLCA sulfonation. Human recombinant SULT2A1, but not SULT2B1b or SULT1E1, catalyzed LCA, GLCA, and TLCA sulfonation, whereas each of these enzymes catalyzed DHEA sulfonation. LCA, GLCA, and TLCA sulfonation is catalyzed by human liver cytosol, and SULT2A1 followed the substrate inhibition model with comparable apparent K m values (≤1 µM). Each of the SERMs inhibited LCA, GLCA, and TLCA sulfonation with varying potency and mode of enzyme inhibition. The potency and extent of inhibition of LCA sulfonation were attenuated or increased by structural modifications to toremifene, bazedoxifene, and lasofoxifene. The inhibitory effect of raloxifene, bazedoxifene, and acolbifene on LCA sulfonation was also observed in HepG2 human hepatocellular carcinoma cells. Overall, among the SERMs investigated, bazedoxifene and raloxifene were the most effective inhibitors of LCA, GLCA, and TLCA sulfonation. These findings provide insight into the structural features of specific SERMs that contribute to their inhibition of SULT2A1-catalyzed LCA sulfonation. Inhibition of LCA, GLCA, and TLCA detoxification by a SERM may provide a biochemical basis for adverse effects associated with a SERM.


Asunto(s)
Biocatálisis/efectos de los fármacos , Ácido Litocólico/análogos & derivados , Moduladores Selectivos de los Receptores de Estrógeno/química , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Ácidos Sulfónicos/metabolismo , Sulfotransferasas/metabolismo , Ácido Taurolitocólico/metabolismo , Citosol/efectos de los fármacos , Citosol/metabolismo , Células Hep G2 , Humanos , Cinética , Ácido Litocólico/metabolismo , Hígado/citología , Oxidación-Reducción , Moduladores Selectivos de los Receptores de Estrógeno/metabolismo , Sulfotransferasas/antagonistas & inhibidores
11.
Chem Res Toxicol ; 32(6): 1281-1288, 2019 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31046239

RESUMEN

Aloe-emodin (AE) is a major anthraquinone ingredient of numerous traditional Chinese medicines with a variety of beneficial biological activities in vitro. Previous studies suggested that AE possessed cytotoxicity and genotoxicity. Nevertheless, the mechanisms of the toxic action of AE have not yet been fully clarified. The present study aimed at characterization of metabolic pathways of AE to better understand the mechanisms of AE-induced cytotoxicity. An AE-derived glutathione conjugate (AE-GSH) was observed in rat liver cytosol incubations containing AE and GSH, along with 3'-phosphoadenosine-5'-phosphosulfate (PAPS). Similar incubation fortified with N-acetylcysteine (NAC) in place of GSH offered an AE-NAC conjugate corresponding to the GSH conjugate. The formation of the two conjugates was found to require PAPS. The two conjugates were respectively detected in bile and urine of rats given AE. Sulfotransferase (SULT) inhibitor pentachlorophenol (PCP) suppressed the production of the observed AE-GSH/NAC conjugates in vivo, which suggested that SULTs participated in the process of the metabolic activation of AE. The presence of PCP attenuated cell susceptibility to AE-induced cytotoxicity. The present study illustrated potential association of sulfation-mediated bioactivation of AE with its cytotoxicity.


Asunto(s)
Antraquinonas/farmacología , Sulfotransferasas/metabolismo , Activación Metabólica/efectos de los fármacos , Animales , Antraquinonas/química , Antraquinonas/metabolismo , Supervivencia Celular/efectos de los fármacos , Citosol/química , Citosol/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Células Hep G2 , Humanos , Hígado/química , Hígado/metabolismo , Masculino , Estructura Molecular , Pentaclorofenol/farmacología , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Sulfotransferasas/antagonistas & inhibidores
12.
Crit Rev Food Sci Nutr ; 59(14): 2308-2320, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29517920

RESUMEN

Migraine is a common multifactorial and polygenic neurological disabling disorder characterized by a genetic background and associated to environmental, hormonal and food stimulations. A large series of evidence suggest a strong correlation between nutrition and migraine and indicates several commonly foods, food additives and beverages that may be involved in the mechanisms triggering the headache attack in migraine-susceptible persons. There are foods and drinks, or ingredients of the same, that can trigger the migraine crisis as well as some foods play a protective function depending on the specific genetic sensitivity of the subject. The recent biotechnological advances have enhanced the identification of some genetic factors involved in onset diseases and the identification of sequence variants of genes responsible for the individual sensitivity to migraine trigger-foods. Therefore many studies are aimed at the analysis of polymorphisms of genes coding for the enzymes involved in the metabolism of food factors in order to clarify the different ways in which people respond to foods based on their genetic constitution. This review discusses the latest knowledge and scientific evidence of the role of gene variants and nutrients, food additives and nutraceuticals interactions in migraine.


Asunto(s)
Bebidas/efectos adversos , Aditivos Alimentarios/efectos adversos , Alimentos/efectos adversos , Trastornos Migrañosos/etiología , Trastornos Migrañosos/genética , Nutrigenómica/métodos , Alcohol Deshidrogenasa/genética , Suplementos Dietéticos/efectos adversos , Histamina/genética , Histamina/metabolismo , Humanos , Trastornos Migrañosos/prevención & control , Fenoles/farmacología , Sulfotransferasas/antagonistas & inhibidores
13.
Lipids Health Dis ; 18(1): 203, 2019 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-31757214

RESUMEN

BACKGROUND: A healthy gastric mucosal epithelium exhibits tumor-suppressive properties. Gastric epithelial cell dysfunction contributes to gastric cancer development. Oxysterols provided from food or cholesterol oxidation in the gastric epithelium may be further sulfated by hydroxysteroid sulfotransferase 2B1 (SULT2B1), which is highly abundant in the gastric epithelium. However, the effects of SULT2B1 on gastric epithelial function and gastric carcinogenesis are unclear. METHODS: A mouse gastric tumor model was established using carcinogenic agent 3-methylcholanthrene (3-MCA). A SULT2B1 deletion (SULT2B1-/-) human gastric epithelial line GES-1 was constructed by CRISPR/CAS9 genome editing system. RESULTS: The gastric tumor incidence was higher in the SULT2B1-/- mice than in the wild-type (WT) mice. In gastric epithelial cells, adenovirus-mediated SULT2B1b overexpression reduced the levels of oxysterols, such as 24(R/S),25-epoxycholesterol (24(R/S),25-EC) and 27-hydroxycholesterol (27HC). This condition also increased PI3K/AKT signaling to promote gastric epithelial cell proliferation, epithelization, and epithelial development. However, SULT2B1 deletion or SULT2B1 knockdown suppressed PI3K/AKT signaling, epithelial cell epithelization, and wound healing and induced gastric epithelial cell malignant transition upon 3-MCA induction. CONCLUSIONS: The abundant SULT2B1 expression in normal gastric epithelium might maintain epithelial function via the PI3K/AKT signaling pathway and suppress gastric carcinogenesis induced by a carcinogenic agent.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Neoplasias Gástricas/genética , Sulfotransferasas/genética , Animales , Secuencia de Bases , Sistemas CRISPR-Cas , Carcinogénesis/efectos de los fármacos , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular , Colesterol/análogos & derivados , Colesterol/metabolismo , Mucosa Gástrica/efectos de los fármacos , Mucosa Gástrica/enzimología , Mucosa Gástrica/patología , Edición Génica , Humanos , Hidroxicolesteroles/metabolismo , Metilcolantreno/administración & dosificación , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Gástricas/inducido químicamente , Neoplasias Gástricas/enzimología , Neoplasias Gástricas/mortalidad , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/deficiencia , Análisis de Supervivencia
14.
Biochem J ; 475(15): 2435-2455, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-29934490

RESUMEN

Protein tyrosine sulfation is a post-translational modification best known for regulating extracellular protein-protein interactions. Tyrosine sulfation is catalysed by two Golgi-resident enzymes termed tyrosylprotein sulfotransferases (TPSTs) 1 and 2, which transfer sulfate from the cofactor PAPS (3'-phosphoadenosine 5'-phosphosulfate) to a context-dependent tyrosine in a protein substrate. A lack of quantitative tyrosine sulfation assays has hampered the development of chemical biology approaches for the identification of small-molecule inhibitors of tyrosine sulfation. In the present paper, we describe the development of a non-radioactive mobility-based enzymatic assay for TPST1 and TPST2, through which the tyrosine sulfation of synthetic fluorescent peptides can be rapidly quantified. We exploit ligand binding and inhibitor screens to uncover a susceptibility of TPST1 and TPST2 to different classes of small molecules, including the anti-angiogenic compound suramin and the kinase inhibitor rottlerin. By screening the Published Kinase Inhibitor Set, we identified oxindole-based inhibitors of the Ser/Thr kinase RAF (rapidly accelerated fibrosarcoma) as low-micromolar inhibitors of TPST1 and TPST2. Interestingly, unrelated RAF inhibitors, exemplified by the dual BRAF/VEGFR2 inhibitor RAF265, were also TPST inhibitors in vitro We propose that target-validated protein kinase inhibitors could be repurposed, or redesigned, as more-specific TPST inhibitors to help evaluate the sulfotyrosyl proteome. Finally, we speculate that mechanistic inhibition of cellular tyrosine sulfation might be relevant to some of the phenotypes observed in cells exposed to anionic TPST ligands and RAF protein kinase inhibitors.


Asunto(s)
Imidazoles/química , Proteínas de la Membrana , Péptidos/química , Proteínas Proto-Oncogénicas B-raf , Piridinas/química , Sulfotransferasas , Tirosina/química , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/química , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/química , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/química
15.
J Appl Toxicol ; 39(4): 650-657, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30874336

RESUMEN

Sulfotransferase 1A (SULT1A) expression is lower in the liver of humans than that of rodents. Therefore, species differences should be taken into consideration when assessing the risk of rodent hepatocarcinogens metabolically activated by SULT1A in humans. Although some renal carcinogens require SULT1A-mediated activation, it is unclear how SULT1A activity in the liver affects renal carcinogens. To explore the effects of SULT1A activity in the liver on genotoxicity induced by SULT1A-activated renal carcinogens, B6C3F1 mice or gpt delta mice of the same strain background were given lucidin-3-O-primeveroside (LuP), a hepatic and renal carcinogen of rodents, for 4 or 13 weeks, respectively, and pentachlorophenol (PCP) as a liver-specific SULT inhibitor, was given from 1 week before LuP treatment to the end of the experiment. A 4 week exposure of LuP induced lucidin-specific DNA adduct formation. The suppression of Sult1a expression was observed only in the liver but not in the kidneys of PCP-treated mice, but co-administration of PCP suppressed LuP-induced DNA adduct formation in both organs. Thirteen-week exposure of LuP increased mutation frequencies and cotreatment with PCP suppressed these increases in both organs. Given that intact levels of SULT activity in the liver were much higher than in the kidneys of rodents, SULT1A may predominantly activate LuP in the liver, consequently leading to genotoxicity not only in the liver but also in the kidney. Thus, species differences should be considered in human risk assessment of renal carcinogens activated by SULT1A as in the case of the corresponding liver carcinogens.


Asunto(s)
Antraquinonas/toxicidad , Disacáridos/toxicidad , Colorantes de Alimentos/toxicidad , Riñón/efectos de los fármacos , Hígado/enzimología , Sulfotransferasas/antagonistas & inhibidores , Animales , Inhibidores Enzimáticos/farmacología , Riñón/patología , Masculino , Ratones , Ratones Endogámicos , Pentaclorofenol/farmacología , Sulfotransferasas/genética
16.
Ecotoxicol Environ Saf ; 180: 146-151, 2019 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31082578

RESUMEN

Thyroid hormones (THs) are essential to proper growth and development of human bodies. Inhibiting the sulfation metabolism of THs has been demonstrated to be an important way for some environmental pollutants, such as halogenated phenolic compounds, to interfere THs homeostasis, thereby causing health problems. However, the important property characteristics that govern the sulfation inhibition of these chemicals are not well understood, and the experimental data on inhibition potential is limited. In this work, an in silico approach was developed to investigate the structure-activity relationship for their sulfotransferases (SULTs) inhibition. A series of quantum chemical descriptors that quantify the electronic and energy properties of 22 halogenated phenolic compounds have been calculated to establish a predictive model and analyzed their corresponding contributions to SULTs inhibition. Density functional theory (DFT) B3LYP/6-31G** has been employed to optimize molecular geometries to obtain a total of 15 descriptors for every compound. The implementation of linear regression shows three descriptors that represent molecular mass, positive charges on hydrogen atoms, and energy of frontier orbitals strongly correlate with SULTs inhibition potential. This indicates molecular size, hydrogen-bond strength, and nucleophilic-electrophilic reactivity may play important roles in SULTs inhibition. The derived regression model has good statistical performance (r2 = 0.84, rms = 0.35), and different validation strategies indicate it can serve as an efficient predictive tool for other chemicals in application domain but with no experimental data, consequently assisting in their THs sulfation inhibition and health risk assessment.


Asunto(s)
Contaminantes Ambientales/farmacología , Fenoles/farmacología , Sulfotransferasas/antagonistas & inhibidores , Hormonas Tiroideas/metabolismo , Simulación por Computador , Contaminantes Ambientales/química , Contaminantes Ambientales/metabolismo , Halogenación , Humanos , Modelos Moleculares , Fenoles/química , Fenoles/metabolismo , Relación Estructura-Actividad , Sulfotransferasas/metabolismo
17.
J Biol Chem ; 292(49): 20305-20312, 2017 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-29038294

RESUMEN

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


Asunto(s)
Sitio Alostérico , Antiinflamatorios no Esteroideos/metabolismo , Citosol/enzimología , Sulfotransferasas/química , Arilsulfotransferasa/antagonistas & inhibidores , Humanos , Isoenzimas/metabolismo , Espectroscopía de Resonancia Magnética , Ácido Mefenámico/metabolismo , Ácido Mefenámico/farmacología , Unión Proteica , Sulfotransferasas/antagonistas & inhibidores
18.
Glycoconj J ; 35(5): 477-491, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30173355

RESUMEN

Chondroitin sulfate E (CS-E) plays a crucial role in diverse processes ranging from viral infection to neuroregeneration. Its regiospecific sulfation pattern, generated by N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase (GalNAc4S-6ST), is the main structural determinant of its biological activity. Inhibitors of GalNAc4S-6ST can serve as powerful tools for understanding physiological functions of CS-E and its potential therapeutic leads for human diseases. A family of new 4-acylamino-ß-GalNAc derivatives and 4-azido-ß-GalNAc derivatives were synthesized for their potential application as inhibitors of GalNAc4S-6ST. The target compounds were evaluated for their inhibitory activities against GalNAc4S-6ST. The results revealed that 4-pivaloylamino- and 4-azido-ß-GalNAc derivatives displayed evident activities against GalNAc4S-6ST with IC50 value ranging from 0.800 to 0.828 mM. They showed higher activities than benzyl D-GalNAc4S that was used as control.


Asunto(s)
Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Galactosamina/síntesis química , Galactosamina/farmacología , Sulfotransferasas/antagonistas & inhibidores , Amidas/química , Animales , Inhibidores Enzimáticos/química , Galactosamina/química , Humanos , Sulfotransferasas/metabolismo
19.
Gastrointest Endosc ; 87(4): 1126-1131, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29122598

RESUMEN

BACKGROUND AND AIMS: Carbohydrate sulfotransferase 15 (CHST15) promotes tumor growth and invasion and is considered to be an emergent therapeutic target for pancreatic cancer. The aim of this study was to evaluate the safety and efficacy of EUS-guided fine-needle injection (EUS-FNI) of STNM01, the double-stranded RNA oligonucleotide that specifically represses CHST15, for use in patients with pancreatic cancer. METHODS: Six patients with unresectable pancreatic cancer, treated at Tokyo Metropolitan Geriatric Hospital, were used in this open-labeled, investigator-initiated trial. A total of 16 mL STNM01 (250 nM) was injected into the tumor through EUS-FNI. The study's primary endpoint was safety, with a secondary endpoint of tumor response 4 weeks after the initial injection. Some patients received a series of infusions as extensions. The local expression of CHST15 and overall survival (OS) were also evaluated. RESULTS: There were no adverse events. The mean tumor diameter changed from 30.7 to 29.3 mm 4 weeks after injection. Four patients exhibited necrosis of tumor in biopsy specimens. CHST15 was highly expressed at baseline, with 2 patients showing large reductions of CHST15 at week 4. The mean OS of these 2 patients was 15 months, whereas it was 5.7 months for the other 4 patients. CONCLUSIONS: EUS-FNI of STNM01 in pancreatic cancer is safe and feasible. The CHST15 reduction could predict tumor progression and OS. Injections of STNM01 during the beginning of treatment may reduce CHST15 and warrants further investigation.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Oligonucleótidos/administración & dosificación , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Anciano , Anciano de 80 o más Años , Endosonografía , Femenino , Humanos , Inyecciones Intralesiones , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/metabolismo , Necrosis , Oligonucleótidos/efectos adversos , Sulfotransferasas/antagonistas & inhibidores , Sulfotransferasas/metabolismo , Tasa de Supervivencia , Carga Tumoral , Ultrasonografía Intervencional
20.
Dermatol Ther ; 31(6): e12741, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30226287

RESUMEN

Topical minoxidil is the only US FDA approved OTC drug for the treatment of androgenetic alopecia (AGA). Minoxidil is a pro-drug converted into its active form, minoxidil sulfate, by the sulfotransferase enzymes in the outer root sheath of hair follicles. Previously, we demonstrated that sulfotransferase activity in hair follicles predicts response to topical minoxidil in the treatment of AGA. In the human liver, sulfotransferase activity is significantly inhibited by salicylic acid. Low-dose OTC aspirin (75-81 mg), a derivative of salicylic acid, is used by millions of people daily for the prevention of coronary heart disease and cancer. It is not known whether oral aspirin inhibits sulfotransferase activity in hair follicles, potentially affecting minoxidil response in AGA patients. In the present study, we determined the follicular sulfotransferase enzymatic activity following 14 days of oral aspirin administration. In our cohort of 24 subjects, 50% were initially predicted to be responders to minoxidil. However, following 14 days of aspirin administration, only 27% of the subjects were predicted to respond to topical minoxidil. To the best of our knowledge, this is the first study to report the effect of low-dose daily aspirin use on the efficacy of topical minoxidil.


Asunto(s)
Alopecia/tratamiento farmacológico , Aspirina/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Folículo Piloso/efectos de los fármacos , Minoxidil/administración & dosificación , Profármacos/administración & dosificación , Sulfotransferasas/antagonistas & inhibidores , Administración Cutánea , Adulto , Alopecia/diagnóstico , Alopecia/fisiopatología , Aspirina/efectos adversos , Interacciones Farmacológicas , Inhibidores Enzimáticos/efectos adversos , Folículo Piloso/enzimología , Folículo Piloso/crecimiento & desarrollo , Humanos , Masculino , Minoxidil/análogos & derivados , Minoxidil/metabolismo , Profármacos/metabolismo , Medición de Riesgo , Sulfotransferasas/metabolismo , Resultado del Tratamiento , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA