Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Phytomedicine ; 80: 153374, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33075645

RESUMEN

BACKGROUND: Insufficient renal urate excretion and/or overproduction of uric acid (UA) are the dominant causes of hyperuricemia. Baicalein (BAL) is widely distributed in dietary plants and has extensive biological activities, including antioxidative, anti-inflammatory and antihypertensive activities. PURPOSE: To investigate the anti-hyperuricemic effects of BAL and the underlying mechanisms in vitro and in vivo. METHODS: We investigated the inhibitory effects of BAL on GLUT9 and URAT1 in vitro through electrophysiological experiments and 14C-urate uptake assays. To evaluate the impact of BAL on serum and urine UA, the expression of GLUT9 and URAT1, and the activity of xanthine oxidase (XOD), we developed a mouse hyperuricemia model by potassium oxonate (PO) injection. Molecular docking analysis based on homology modeling was performed to explain the predominant efficacy of BAL compared with the other test compounds. RESULTS: BAL dose-dependently inhibited GLUT9 and URAT1 in a noncompetitive manner with IC50 values of 30.17 ± 8.68 µM and 31.56 ± 1.37 µM, respectively. BAL (200 mg/kg) significantly decreased serum UA and enhanced renal urate excretion in PO-induced hyperuricemic mice. Moreover, the expression of GLUT9 and URAT1 in the kidney was downregulated, and XOD activity in the serum and liver was suppressed. The docking analysis revealed that BAL potently interacted with Trp336, Asp462, Tyr71 and Gln328 of GLUT9 and Ser35 and Phe241 of URAT1. CONCLUSION: These results indicated that BAL exerts potent antihyperuricemic efects through renal UA excretal promotion and serum UA production. Thus, we propose that BAL may be a promising treatment for the prevention of hyperuricemia owing to its multitargeted inhibitory activity.


Asunto(s)
Flavanonas/farmacología , Hiperuricemia/tratamiento farmacológico , Ácido Úrico/orina , Xantina Oxidasa/antagonistas & inhibidores , Animales , Antioxidantes/farmacología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Flavanonas/química , Flavanonas/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/química , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Células HEK293 , Humanos , Hiperuricemia/inducido químicamente , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/química , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Ácido Oxónico/toxicidad , Ácido Úrico/sangre
2.
J Chem Inf Model ; 57(6): 1402-1413, 2017 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-28562037

RESUMEN

Human organic anion transporters (OATPs) are vital for the uptake and efflux of drugs and endogenous compounds. Current identification of inhibitors of these transporters is based on experimental screening. Virtual screening remains a challenge due to a lack of experimental three-dimensional protein structures. Here, we describe a workflow to identify inhibitors of the OATP2B1 transporter in the DrugBank library of over 5,000 drugs and druglike molecules. OATP member 2B1 transporter is highly expressed in the intestine, where it participates in oral absorption of drugs. Predictions from a Random forest classifier, prioritized by docking against multiple comparative protein structure models of OATP2B1, indicated that 33 of the 5,000 compounds were putative inhibitors of OATP2B1. Ten predicted inhibitors that are prescription drugs were tested experimentally in cells overexpressing the OATP2B1 transporter. Three of these ten were validated as potent inhibitors of estrone-3-sulfate uptake (defined as more than 50% inhibition at 20 µM) and tested in multiple concentrations to determine exact IC50. The IC50 values of bicalutamide, ticagrelor, and meloxicam suggest that they might inhibit intestinal OATP2B1 at clinically relevant concentrations and therefore modulate the absorption of other concomitantly administered drugs.


Asunto(s)
Descubrimiento de Drogas/métodos , Transportadores de Anión Orgánico/antagonistas & inhibidores , Animales , Células CHO , Simulación por Computador , Cricetinae , Cricetulus , Evaluación Preclínica de Medicamentos , Humanos , Concentración 50 Inhibidora , Simulación del Acoplamiento Molecular , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/metabolismo , Conformación Proteica
3.
Mol Cell Endocrinol ; 428: 133-41, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27033324

RESUMEN

The sodium-dependent organic anion transporter SOAT specifically transports sulfated steroid hormones and is supposed to play a role in testicular steroid regulation and male fertility. The present study aimed to identify novel specific SOAT inhibitors for further in vitro and in vivo studies on SOAT function. More than 100 compounds of different molecular structures were screened for inhibition of the SOAT-mediated transport of dehydroepiandrosterone sulfate in stably transfected SOAT-HEK293 cells. Twenty-five of these with IC50 values covering four orders of magnitude were selected as training set for 3D pharmacophore modelling. The SOAT pharmacophore features were calculated by CATALYST and consist of three hydrophobic sites and two hydrogen bond acceptors. By substrate database screening, compound T 0511-1698 was predicted as a novel SOAT inhibitor with an IC50 of 15 µM. This value was confirmed by cell-based transport assays. Therefore, the developed SOAT pharmacophore model demonstrated its suitability in predicting novel SOAT inhibitors.


Asunto(s)
Sulfato de Deshidroepiandrosterona/metabolismo , Evaluación Preclínica de Medicamentos , Modelos Moleculares , Transportadores de Anión Orgánico/antagonistas & inhibidores , Transportadores de Anión Orgánico/química , Relación Estructura-Actividad Cuantitativa , Ácidos y Sales Biliares/química , Ácidos y Sales Biliares/farmacología , Células HEK293 , Humanos , Concentración 50 Inhibidora , Reproducibilidad de los Resultados
4.
Curr Top Membr ; 73: 233-61, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24745985

RESUMEN

Transporters within the SLC22, SLC44, and SLC47 families of solute carriers mediate transport of a structurally diverse array of organic electrolytes, that is, molecules that are generally charged (cationic, anionic, or zwitterionic) at physiological pH. Transporters in the SLC22 family--all of which are members of the major facilitator superfamily (MFS) of transporters--represent a mechanistically diverse set of processes, including the organic anion transporters (OATs and URAT1) that physiologically operate as organic anion (OA) exchangers, the organic cation transporters (OCTs) that operate as electrogenic uniporters of organic cations (OCs), and the so-called "novel" organic cation transporters (OCTNs) that support Na-cotransport of selected zwitterions. Whereas the OCTNs display a high degree of substrate selectivity, the physiological hallmark of the OATs and OCTs is their multiselectivity--consistent with a principal role in renal and hepatic clearance of a wide array of both endogenous and xenobiotic compounds. SLC47 consists of members of the multidrug and toxin extruder (MATE) family, which are carriers that are obligatory exchangers and that physiologically support electroneutral H⁺ exchange. The MATEs also display a characteristic multiselectivity and are frequently paired with OCTs to mediate transepithelial OC secretion, with the OCTs typically supporting basolateral OC entry and the MATEs supporting apical OC efflux. The SLC44 family contains the choline transporter-like (CTL) transporters. Largely restricted to choline and a limited set of structural congeners, the CTLs appear to support the Na-independent, electrogenic uniport of choline, thereby providing choline for membrane biogenesis. The solution of X-ray crystal structures of representative prokaryotic MFS and MATE transporters has led to the development of homology models of mammalian OAT, OCT, and MATE transporters that, in turn, have supplemented studies of the molecular basis of the complex interactions of ligands with these multiselective proteins.


Asunto(s)
Células/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Animales , Humanos , Transportadores de Anión Orgánico/química , Proteínas de Transporte de Catión Orgánico/química
5.
J Biol Chem ; 287(14): 11489-97, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22334707

RESUMEN

Secondary active transporters from the SLC17 protein family are required for excitatory and purinergic synaptic transmission, sialic acid metabolism, and renal function, and several members are associated with inherited neurological or metabolic diseases. However, molecular tools to investigate their function or correct their genetic defects are limited or absent. Using structure-activity, homology modeling, molecular docking, and mutagenesis studies, we have located the substrate-binding site of sialin (SLC17A5), a lysosomal sialic acid exporter also recently implicated in exocytotic release of aspartate. Human sialin is defective in two inherited sialic acid storage diseases and is responsible for metabolic incorporation of the dietary nonhuman sialic acid N-glycolylneuraminic acid. We built cytosol-open and lumen-open three-dimensional models of sialin based on weak, but significant, sequence similarity with the glycerol-3-phosphate and fucose permeases from Escherichia coli, respectively. Molecular docking of 31 synthetic sialic acid analogues to both models was consistent with inhibition studies. Narrowing the sialic acid-binding site in the cytosol-open state by two phenylalanine to tyrosine mutations abrogated recognition of the most active analogue without impairing neuraminic acid transport. Moreover, a pilot virtual high-throughput screening of the cytosol-open model could identify a pseudopeptide competitive inhibitor showing >100-fold higher affinity than the natural substrate. This validated model of human sialin and sialin-guided models of other SLC17 transporters should pave the way for the identification of inhibitors, glycoengineering tools, pharmacological chaperones, and fluorescent false neurotransmitters targeted to these proteins.


Asunto(s)
Biología Computacional , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/metabolismo , Simportadores/química , Simportadores/metabolismo , Azepinas/metabolismo , Sitios de Unión , Evaluación Preclínica de Medicamentos , Células HEK293 , Humanos , Indoles/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación Missense , Transportadores de Anión Orgánico/genética , Proyectos Piloto , Unión Proteica , Conformación Proteica , Homología de Secuencia de Aminoácido , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Relación Estructura-Actividad , Simportadores/genética
6.
J Neurochem ; 119(1): 1-5, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21781115

RESUMEN

Sialin, the protein coded by SLC17A5, is responsible for membrane potential (Δψ)-driven aspartate and glutamate transport into synaptic vesicles in addition to H+/sialic acid co-transport in lysosomes. Rodent sialin mutants harboring the mutations associated with Salla disease in humans did not transport aspartate and glutamate whereas H+/sialic acid co-transport activity was about one-third of the wild-type protein. In this study, we investigate the effects of various mutations on the transport activities of human sialin. Proteoliposomes containing purified heterologously expressed human sialin exhibited both Δψ-driven aspartate and glutamate transport activity and H+/sialic acid co-transport activity. Aspartate and glutamate transport was not detected in the R39C and K136E mutant forms of SLC17A5 protein associated with Salla disease, whereas H+/sialic acid co-transport activity corresponded to 30-50% of the recombinant wild-type protein. In contrast, SLC17A5 protein harboring the mutations associated with infantile sialic acid storage disease, H183R and Δ268SSLRN272 still showed normal levels of Δψ-driven aspartate and glutamate transport even though H+/sialic acid co-transport activity was absent. Human sialin carrying the G328E mutation that causes both phenotypes, and P334R and G378V mutations that cause infantile sialic acid storage disease showed no transport activity. These results support the idea that people suffering from Salla disease have been defective in aspartergic and glutamatergic neurotransmissions.


Asunto(s)
Transportadores de Anión Orgánico/metabolismo , Simportadores/metabolismo , Proteínas de Transporte Vesicular de Aminas Biógenas/metabolismo , Animales , Ácido Aspártico/metabolismo , Baculoviridae/metabolismo , Transporte Biológico Activo/fisiología , ADN Complementario/biosíntesis , ADN Complementario/genética , Ácido Glutámico/metabolismo , Humanos , Liposomas/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis , Mutación/fisiología , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/genética , Ratas , Proteínas Recombinantes/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Enfermedad por Almacenamiento de Ácido Siálico/genética , Simportadores/química , Simportadores/genética
7.
FEBS Lett ; 581(12): 2255-62, 2007 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-17418140

RESUMEN

Soluble aluminium (Al(3+)) is the major constraint to plant growth on acid soils. Plants have evolved mechanisms to tolerate Al(3+) and one type of mechanism relies on the efflux of organic anions that protect roots by chelating the Al(3+). Al(3+) resistance genes of several species have now been isolated and found to encode membrane proteins that facilitate organic anion efflux from roots. These proteins belong to the Al(3+)-activated malate transporter (ALMT) and multi-drug and toxin extrusion (MATE) families. We review the roles of these proteins in Al(3+) resistance as well as their roles in other aspects of mineral nutrition.


Asunto(s)
Aluminio/toxicidad , Proteínas de Transporte de Membrana/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteínas de Plantas/metabolismo , Plantas/efectos de los fármacos , Plantas/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Evolución Biológica , Resistencia a Medicamentos/genética , Genes de Plantas , Transporte Iónico/genética , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Minerales/metabolismo , Modelos Biológicos , Modelos Moleculares , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/genética , Fósforo/metabolismo , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/genética , Plantas/genética , Estructura Secundaria de Proteína
8.
J Steroid Biochem Mol Biol ; 97(4): 369-75, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16150593

RESUMEN

A family of organic anion transporters (OAT) recently identified has important roles for the excretion or reabsorption of endogenous and exogenous compounds, and several new isoforms have been reported in this decade. Although the transepithelial transport properties of organic anions are gradually being understood, many portions of their functional characteristics in functions remain to be elucidated. A recently reported new cDNA encoding a mouse OAT5 (mOAT5) was constructed, using 3'-RACE PCR, with the total RNA isolated from a mouse kidney. When mOAT5 was expressed in Xenopus oocytes, mOAT5 transported estrone sulfate, dehydroepiandrosterone sulfate and ochratoxin A. Estrone sulfate uptake by mOAT5 displayed a time-dependent and sodium-independent manner. The Km values of estrone sulfate and dehydroepiandrosterone sulfate were 2.2 and 3.8 microM, respectively. mOAT5 interacted with chemically heterogeneous steroid or organic sulfates, such as nitrophenyl sulfate, methylumbelliferyl sulfate and estradiol sulfates. In contrast to the sulfate conjugates, mOAT5-mediated estrone sulfate uptake was not inhibited by the steroid or organic glucuronides. The mOAT5 protein having about 85 kDa molecular weight was shown to be mainly localized in the apical membrane of the proximal tubules of the outer medulla. These results suggest an important role of mOAT5 for the excretion or reabsorption of steroid sulfates in the kidney.


Asunto(s)
Riñón/metabolismo , Transportadores de Anión Orgánico/química , Sulfatos/metabolismo , Animales , Transporte Biológico , Western Blotting , Proteínas Portadoras/química , Clonación Molecular , ADN Complementario/metabolismo , Sulfato de Deshidroepiandrosterona/metabolismo , Relación Dosis-Respuesta a Droga , Estrona/análogos & derivados , Estrona/metabolismo , Glucurónidos/química , Immunoblotting , Inmunohistoquímica , Túbulos Renales/metabolismo , Cinética , Ratones , Ocratoxinas/metabolismo , Oocitos/metabolismo , ARN/metabolismo , Sulfatos/química , Factores de Tiempo , Xenopus
9.
J Biol Chem ; 278(30): 27930-8, 2003 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-12740363

RESUMEN

A novel transport protein with the properties of voltage-driven organic anion transport was isolated from pig kidney cortex by expression cloning in Xenopus laevis oocytes. A cDNA library was constructed from size-fractionated poly(A)+ RNA and screened for p-aminohippurate (PAH) transport in high potassium medium. A 1856-base pair cDNA encoding a 467-amino acid peptide designated as OATV1 (voltage-driven organic anion transporter 1) was isolated. The predicted amino acid sequence of OATV1 exhibited 60-65% identity to those of human, rat, rabbit, and mouse sodium-dependent phosphate cotransporter type 1 (NPT1), although OATV1 did not transport phosphate. The homology of this transporter to known members of the organic anion transporter family (OAT family) was about 25-30%. OATV1-mediated PAH transport was affected by the changes in membrane potential. The transport was Na+-independent and enhanced at high concentrations of extracellular potassium and low concentrations of extracellular chloride. Under the voltage clamp condition, extracellularly applied PAH induced outward currents in oocytes expressing OATV1. The current showed steep voltage dependence, consistent with the voltage-driven transport of PAH by OATV1. The PAH transport was inhibited by various organic anions but not by organic cations, indicating the multispecific nature of OATV1 for anionic compounds. This transport protein is localized at the apical membrane of renal proximal tubule, consistent with the proposed localization of a voltage-driven organic anion transporter. Therefore, it is proposed that OATV1 plays an important role to excrete drugs, xenobiotics, and their metabolites driven by membrane voltage through the apical membrane of the tubular epithelial cells into the urine.


Asunto(s)
Membrana Celular/metabolismo , Transporte Iónico , Túbulos Renales/metabolismo , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/fisiología , Animales , Transporte Biológico , Northern Blotting , Western Blotting , Cloro/farmacología , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Electrofisiología , Células Epiteliales/metabolismo , Biblioteca de Genes , Humanos , Concentración de Iones de Hidrógeno , Inmunohistoquímica , Iones , Riñón/metabolismo , Cinética , Potenciales de la Membrana , Ratones , Oocitos/metabolismo , Técnicas de Placa-Clamp , Péptidos/química , Poli A , Potasio/metabolismo , Potasio/farmacología , ARN Complementario/metabolismo , Conejos , Ratas , Sodio/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III , Especificidad por Sustrato , Porcinos , Simportadores/química , Factores de Tiempo , Xenopus , Xenopus laevis , Ácido p-Aminohipúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA