Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Biol Chem ; 298(8): 102161, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35724964

RESUMEN

Recent studies have shown that human solute carrier SLC19A3 (hSLC19A3) can transport pyridoxine (vitamin B6) in addition to thiamine (vitamin B1), its originally identified substrate, whereas rat and mouse orthologs of hSLC19A3 can transport thiamine but not pyridoxine. This finding implies that some amino acid residues required for pyridoxine transport, but not for thiamine transport, are specific to hSLC19A3. Here, we sought to identify these residues to help clarify the unique operational mechanism of SLC19A3 through analyses comparing hSLC19A3 and mouse Slc19a3 (mSlc19a3). For our analyses, hSLC19A3 mutants were prepared by replacing selected amino acid residues with their counterparts in mSlc19a3, and mSlc19a3 mutants were prepared by substituting selected residues with their hSLC19A3 counterparts. We assessed pyridoxine and thiamine transport by these mutants in transiently transfected human embryonic kidney 293 cells. Our analyses indicated that the hSLC19A3-specific amino acid residues of Gln86, Gly87, Ile91, Thr93, Trp94, Ser168, and Asn173 are critical for pyridoxine transport. These seven amino acid residues were found to be mostly conserved in the SLC19A3 orthologs that can transport pyridoxine but not in orthologs that are unable to transport pyridoxine. In addition, these residues were also found to be conserved in several SLC19A2 orthologs, including rat, mouse, and human orthologs, which were all found to effectively transport both pyridoxine and thiamine, exhibiting no species-dependent differences. Together, these findings provide a molecular basis for the unique functional characteristics of SLC19A3 and also of SLC19A2.


Asunto(s)
Aminoácidos , Proteínas de Transporte de Membrana/metabolismo , Aminoácidos/metabolismo , Animales , Transporte Biológico , Células Epiteliales/metabolismo , Humanos , Ratones , Ratas , Tiamina/genética , Tiamina/metabolismo
2.
Drug Metab Dispos ; 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37963658

RESUMEN

Our recent study revealed that SLC49A4, known as disrupted in renal carcinoma 2, is a H+-coupled lysosomal exporter for pyridoxine (vitamin B6), a cationic compound, and involved in the regulation of its lysosomal and cellular levels. We here examined a possibility that this transporter might also transport cationic amphiphilic drugs (CADs) that are known to undergo lysosomal trapping, using pyrilamine, an H1-antagonist, as a model CAD and the COS-7 cell line as a model cell system for transient introduction of human SLC49A4 and a recombinant SLC49A4 protein (SLC49A4-AA), in which the N-terminal dileucine motif involved in lysosomal localization was removed by replacing with dialanine for redirected localization to the plasma membrane. The introduction of SLC49A4 into COS-7 cells induced a significant decrease in the accumulation of pyrilamine in the intracellular compartments in the cells treated with digitonin for permeabilization of plasma membranes, suggesting its operation for lysosomal pyrilamine export. Accordingly, functional analysis using the SLC49A4-AA mutant, which operates for cellular uptake at the plasma membrane, in transiently transfected COS-7 cells demonstrated its H+-coupled operation for pyrilamine transport, which was saturable with a Michaelis constant of 132 µM at pH 5.5. In addition, many CADs that may potentially undergo lysosomal trapping, which include imipramine, propranolol, verapamil, and some others, were found to inhibit SLC49A4-AA-mediated pyrilamine transport, suggesting their affinity for SLC49A4. These results suggest that SLC49A4 is involved in the lysosomal trapping of pyrilamine, operating for its exit. The CADs that inhibited SLC49A4-AA-mediated pyrilamine transport could also be SLC49A4 substrate candidates. Significance Statement SLC49A4 mediates the transport of pyrilamine in a H+-coupled manner at the lysosomal membrane. This could be a newly identified mechanism for lysosomal export involved in its lysosomal trapping.

3.
J Biol Chem ; 295(50): 16998-17008, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33008889

RESUMEN

SLC19A2 and SLC19A3, also known as thiamine transporters (THTR) 1 and 2, respectively, transport the positively charged thiamine (vitamin B1) into cells to enable its efficient utilization. SLC19A2 and SLC19A3 are also known to transport structurally unrelated cationic drugs, such as metformin, but whether this charge selectivity extends to other molecules, such as pyridoxine (vitamin B6), is unknown. We tested this possibility using Madin-Darby canine kidney II (MDCKII) cells and human embryonic kidney 293 (HEK293) cells for transfection experiments, and also using Caco-2 cells as human intestinal epithelial model cells. The stable expression of SLC19A2 and SLC19A3 in MDCKII cells (as well as their transient expression in HEK293 cells) led to a significant induction in pyridoxine uptake at pH 5.5 compared with control cells. The induced uptake was pH-dependent, favoring acidic conditions over neutral to basic conditions, and protonophore-sensitive. It was saturable as a function of pyridoxine concentration, with an apparent Km of 37.8 and 18.5 µm, for SLC19A2 and SLC19A3, respectively, and inhibited by the pyridoxine analogs pyridoxal and pyridoxamine as well as thiamine. We also found that silencing the endogenous SLC19A3, but not SLC19A2, of Caco-2 cells with gene-specific siRNAs lead to a significant reduction in carrier-mediated pyridoxine uptake. These results show that SLC19A2 and SLC19A3 are capable of recognizing/transporting pyridoxine, favoring acidic conditions for operation, and suggest a possible role for these transporters in pyridoxine transport mainly in tissues with an acidic environment like the small intestine, which has an acidic surface microclimate.


Asunto(s)
Ácidos/metabolismo , Células Epiteliales/metabolismo , Intestino Delgado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Microclima , Animales , Transporte Biológico , Perros , Humanos , Concentración de Iones de Hidrógeno , Células de Riñón Canino Madin Darby , Tiamina/metabolismo
4.
Biol Pharm Bull ; 43(9): 1293-1300, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32879202

RESUMEN

It has long been suggested that a Na+-dependent carrier-mediated transport system is involved in the absorption of nucleobases and analogs, including some drugs currently in therapeutic use, for their uptake at the brush border membrane of epithelial cells in the small intestine, mainly based on studies in non-primate experimental animals. The presence of this transport system was indeed proved by the recent identification of sodium-dependent nucleobase transporter 1 (SNBT1/Slc23a4) as its molecular entity in rats. However, this transporter has been found to be genetically deficient in humans and higher primates. Aware of this deficiency, we need to revisit the issue of the absorption of these compounds in the human small intestine so that we can understand the mechanisms and gain information to assure the more rational use and development of drugs analogous to nucleobases. Here, we review the current understanding of the intestinal absorption of nucleobases and analogs. This includes recent knowledge about the efflux transport of those compounds across the basolateral membrane when exiting epithelial cells, following brush border uptake, in order to complete the overall absorption process; the facilitative transporters of equilibrative nucleoside transporter 1 (ENT1/SLC29A1) and equilibrative nucleobase transporter 1 (ENBT1/SLC43A3) may be involved in that in many animal species, including human and rat, without any major species differences.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Absorción Intestinal/genética , Purinas/farmacocinética , Pirimidinas/farmacocinética , Sistemas de Transporte de Aminoácidos/genética , Animales , Membrana Celular , Tranportador Equilibrativo 1 de Nucleósido/genética , Humanos , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Proteínas de Transporte de Nucleobases/metabolismo , Ratas , Especificidad de la Especie
5.
Biochem Biophys Res Commun ; 495(3): 2152-2157, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29273507

RESUMEN

Bioluminescence (BL) imaging based on d-luciferin (d-luc)-luciferase reaction allows noninvasive and real-time monitoring of luciferase-expressing cells. Because BL intensity depends on photons generated through the d-luc-luciferase reaction, an approach to increase intracellular levels of d-luc could improve the detection sensitivity. In the present study, we showed that organic anion transporter 1 (OAT1) is useful, as a d-luc transporter, in boosting the BL intensity in luciferase-expressing cells. Functional screening of several transporters showed that the expression of OAT1 in HEK293 cells stably expressing Pyrearinus termitilluminans luciferase (HEK293/eLuc) markedly enhanced BL intensity in the presence of d-luc. When OAT1 was transiently expressed in HEK293 cells, intracellular accumulation of d-luc was higher than that in control cells, and the specific d-luc uptake mediated by OAT1 was saturable with a Michaelis constant (Km) of 0.23 µM. The interaction between OAT1 and d-luc was verified using 6-carboxyfluorescein, a typical substrate of OAT1, which showed that d-luc inhibited the uptake of 6-carboxyfluorescein mediated by OAT1. BL intensity was concentration-dependent at steady states in HEK293/eLuc cells stably expressing OAT1, and followed Michaelis-Menten kinetics with an apparent Km of 0.36 µM. In addition, the enhanced BL was significantly inhibited by OAT1-specific inhibitors. Thus, OAT1-mediated transport of d-luc could be a rate-limiting step in the d-luc-luciferase reaction. Furthermore, we found that expressing OAT1 in HEK293/eLuc cells implanted subcutaneously in mice also significantly increased the BL after intraperitoneal injection of d-luc. Our findings suggest that because OAT1 is capable of transporting d-luc, it can also be used to improve visualization and monitoring of luciferase-expressing cells.


Asunto(s)
Benzotiazoles/metabolismo , Aumento de la Imagen/métodos , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Genes Reporteros/genética , Células HEK293 , Humanos , Luciferasas/genética , Imagen Molecular/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
6.
J Pharmacol Exp Ther ; 360(1): 59-68, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27807008

RESUMEN

A suicide gene therapy using herpes simplex virus thymidine kinase (HSV-TK) with ganciclovir (GCV) has been under development as a tumor-targeted therapy; however, the mechanism of cellular GCV uptake, which is prerequisite in the therapy, has not been clarified. In an attempt to resolve this situation and gain information to optimize HSV-TK/GCV system for cancer therapy, we found that human equilibrative nucleobase transporter 1 (ENBT1) can transport GCV with a Michaelis constant of 2.75 mM in Madin-Darby canine kidney II (MDCKII) cells stably transfected with this transporter. In subsequent experiments using green fluorescent protein (GFP)-tagged ENBT1 (GFP-ENBT1) and HSV-TK, the uptake of GCV (30 µM), which was minimal in MDCKII cells and unchanged by their transfection with HSV-TK alone, was increased extensively by their transfection with GFP-ENBT1, together with HSV-TK. Accordingly, cytotoxicity, which was assessed by the WST-8 cell viability assay after the treatment of those cells with GCV (30 µM) for 72 hours, was induced in those transfected with GFP-ENBT1, together with HSV-TK but not in those transfected with HSV-TK alone. These results suggest that ENBT1 could facilitate GCV uptake and thereby enhance cytotoxicity in HSV-TK/GCV system. We also identified Helacyton gartleri (HeLa) and HepG2 as cancer cell lines that are rich with ENBT1 and A549, HCT-15 and MCF-7 as those poor with ENBT1. Accordingly, the HSV-TK/GCV system was effective in inducing cytotoxicity in the former but not in the latter. Thus, ENBT1 was found to be a GCV transporter that could enhance the performance of HSV-TK/GCV suicide gene therapy.


Asunto(s)
Sistemas de Transporte de Aminoácidos/metabolismo , Apoptosis/genética , Ganciclovir/metabolismo , Ganciclovir/farmacología , Terapia Genética , Simplexvirus/enzimología , Timidina Quinasa/genética , Animales , Transporte Biológico , Línea Celular , Perros , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Simplexvirus/genética
7.
Ann Rheum Dis ; 76(5): 869-877, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27899376

RESUMEN

OBJECTIVE: A genome-wide association study (GWAS) of gout and its subtypes was performed to identify novel gout loci, including those that are subtype-specific. METHODS: Putative causal association signals from a GWAS of 945 clinically defined gout cases and 1213 controls from Japanese males were replicated with 1396 cases and 1268 controls using a custom chip of 1961 single nucleotide polymorphisms (SNPs). We also first conducted GWASs of gout subtypes. Replication with Caucasian and New Zealand Polynesian samples was done to further validate the loci identified in this study. RESULTS: In addition to the five loci we reported previously, further susceptibility loci were identified at a genome-wide significance level (p<5.0×10-8): urate transporter genes (SLC22A12 and SLC17A1) and HIST1H2BF-HIST1H4E for all gout cases, and NIPAL1 and FAM35A for the renal underexcretion gout subtype. While NIPAL1 encodes a magnesium transporter, functional analysis did not detect urate transport via NIPAL1, suggesting an indirect association with urate handling. Localisation analysis in the human kidney revealed expression of NIPAL1 and FAM35A mainly in the distal tubules, which suggests the involvement of the distal nephron in urate handling in humans. Clinically ascertained male patients with gout and controls of Caucasian and Polynesian ancestries were also genotyped, and FAM35A was associated with gout in all cases. A meta-analysis of the three populations revealed FAM35A to be associated with gout at a genome-wide level of significance (p meta =3.58×10-8). CONCLUSIONS: Our findings including novel gout risk loci provide further understanding of the molecular pathogenesis of gout and lead to a novel concept for the therapeutic target of gout/hyperuricaemia.


Asunto(s)
Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Gota/genética , Adulto , Anciano , Pueblo Asiatico/genética , Estudios de Casos y Controles , Proteínas de Transporte de Catión/genética , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Sitios Genéticos , Genotipo , Gota/clasificación , Histonas/genética , Humanos , Japón , Masculino , Persona de Mediana Edad , Nativos de Hawái y Otras Islas del Pacífico/genética , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Polimorfismo de Nucleótido Simple , Proteínas/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Población Blanca/genética
8.
Drug Metab Dispos ; 44(10): 0, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27417181

RESUMEN

We previously demonstrated that differentiated enterocytes from human induced pluripotent stem (iPS) cells exhibited drug-metabolizing activities and cytochrome P450 CYP3A4 inducibility. The aim of this study was to apply human iPS cell-derived enterocytes in pharmacokinetic studies by investigating the characteristics of drug transport into enterocyte-like cells. Human iPS cells cultured on feeder cells were differentiated into endodermal cells using activin A. These endodermal-like cells were then differentiated into intestinal stem cells by fibroblast growth factor 2. Finally, epidermal growth factor and small-molecule compounds induced the maturation of the intestinal stem cell-like cells. After differentiation, we performed transepithelial electrical resistance (TEER) measurements, immunofluorescence staining, and transport studies. TEER values increased in a time-dependent manner and reached approximately 100 Ω × cm(2) Efflux transport of Hoechst 33342, a substrate of breast cancer resistance protein (BCRP), was observed and inhibited by the BCRP inhibitor Ko143. The uptake of peptide transporter 1 substrate glycylsarcosine was also confirmed and suppressed when the temperature was lowered to 4°C. Using immunofluorescence staining, villin and Na(+)-K(+) ATPase were expressed. These results suggest that human iPS cell-derived enterocytes had loose tight junctions, polarity, as well as uptake and efflux transport functions. In addition, the rank order of apparent membrane permeability coefficient (Papp) values of these test compounds across the enterocyte-like cell membrane corresponded to the fraction absorbance (Fa) values. Therefore, differentiated enterocytes from human iPS cells may provide a useful comprehensive evaluation model of drug transport and metabolism in the small intestine.


Asunto(s)
Enterocitos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Mucosa Intestinal/metabolismo , Bencimidazoles/metabolismo , Transporte Biológico , Técnica del Anticuerpo Fluorescente , Humanos , Intestinos/citología
9.
J Biol Chem ; 289(7): 4405-16, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24379411

RESUMEN

Colonic microbiota synthesize a considerable amount of thiamine in the form of thiamine pyrophosphate (TPP). Recent functional studies from our laboratory have shown the existence of a specific, high-affinity, and regulated carrier-mediated uptake system for TPP in human colonocytes. Nothing, however, is known about the molecular identity of this system. Here we report on the molecular identification of the colonic TPP uptake system as the product of the SLC44A4 gene. We cloned the cDNA of SLC44A4 from human colonic epithelial NCM460 cells, which, upon expression in ARPE19 cells, led to a significant (p < 0.01, >5-fold) induction in [(3)H]TPP uptake. Uptake by the induced system was also found to be temperature- and energy-dependent; Na(+)-independent, slightly higher at acidic buffer pH, and highly sensitive to protonophores; saturable as a function of TPP concentration, with an apparent Km of 0.17 ± 0.064 µM; and highly specific for TPP and not affected by free thiamine, thiamine monophosphate, or choline. Expression of the human TPP transporter was found to be high in the colon and negligible in the small intestine. A cell surface biotinylation assay and live cell confocal imaging studies showed the human TPP transporter protein to be expressed at the apical membrane domain of polarized epithelia. These results show, for the first time, the molecular identification and characterization of a specific and high-affinity TPP uptake system in human colonocytes. The findings further support the hypothesis that the microbiota-generated TPP is absorbable and could contribute toward host thiamine homeostasis, especially toward cellular nutrition of colonocytes.


Asunto(s)
Colon/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana/biosíntesis , Tiamina Pirofosfato/biosíntesis , Animales , Transporte Biológico Activo/fisiología , Clonación Molecular , Colon/citología , ADN Complementario , Perros , Humanos , Concentración de Iones de Hidrógeno , Intestino Delgado/citología , Intestino Delgado/metabolismo , Células de Riñón Canino Madin Darby , Proteínas de Transporte de Membrana/genética , Especificidad de Órganos/fisiología , Tiamina Pirofosfato/genética
10.
Pharm Res ; 31(1): 136-47, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23907530

RESUMEN

PURPOSE: Multidrug and toxin extrusion proteins (MATEs) are multispecific organic cation transporters mediating the efflux of various cationic drugs into the urine. The present study aimed at identifying endogenous compounds in human plasma and urine specimens as biomarkers to evaluate drug interactions involving MATEs in the kidney without administration of their exogenous probe drugs. METHODS: An untargeted metabolomic analysis was performed using urine and plasma samples from healthy volunteers and mice treated with or without the potent MATE inhibitor, pyrimethamine. Plasma and urinary concentrations of candidate markers were measured using liquid chromatography-mass spectrometry. Transport activities were determined in MATE- or OCT2-expressing HEK293 cells. The deuterium-labeled compounds of candidates were administered to mice for pharmacokinetics study. RESULTS: Urinary excretion of eleven compounds including thiamine and carnitine was significantly lower in the pyrimethamine-treatment group in humans and mice, whereas no endogenous compound was noticeably accumulated in the plasma. The renal clearance of thiamine and carnitine was decreased by 70%-84% and 90%-94% (p < 0.05), respectively, in human. The specific uptake of thiamine was observed in MATE1-, MATE2-K- or OCT2-expressing HEK293 cells with Km of 3.5 ± 1.0, 3.9 ± 0.8 and 59.9 ± 6.7 µM, respectively. The renal clearance of carnitine-d 3 was decreased by 62% in mice treated with pyrimethamine. CONCLUSIONS: Our findings indicate that MATEs account for the efflux of thiamine and perhaps carnitine as well as drugs into the urine. The urinary excretion of thiamine is useful to detect drug interaction involving MATEs in the kidney.


Asunto(s)
Biomarcadores/sangre , Biomarcadores/orina , Interacciones Farmacológicas/fisiología , Proteínas de Transporte de Catión Orgánico/metabolismo , Adulto , Animales , Transporte Biológico/fisiología , Línea Celular , Células HEK293 , Humanos , Riñón/metabolismo , Riñón/fisiología , Masculino , Ratones , Adulto Joven
12.
Biochem Biophys Res Commun ; 431(2): 158-63, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23313509

RESUMEN

Proton-coupled folate transporter (PCFT), which is responsible for the intestinal uptake of folates and analogs, is expressed only in the proximal region in the small intestine. The present study was to examine its transcriptional regulation, which may be involved in such a unique expression profile and potentially in its alteration, using dual-luciferase reporter assays in human embryonic kidney (HEK) 293 cells. The luciferase activity derived from the reporter construct containing the 5'-flanking sequence of -1695/+96 of the human PCFT gene was enhanced most extensively by the introduction of Krüppel-like factor 4 (KLF4). The KLF4-induced luciferase activity was further enhanced by hepatocyte nuclear factor 4α (HNF4α) synergistically. To the contrary, caudal-type homeobox transcription factor 2 (CDX2) and CCAAT/enhancer-binding protein α (C/EBPα) extensively suppressed the luciferase activity induced by KLF4 alone and also that induced by KLF4 and HNF4α. Western blot analysis using the rat small intestine indicated uniform expression of KLF4 along the intestinal tract, proximal-oriented expression of HNF4α, distal-oriented expression of CDX2 and C/EBPα. These results suggest that the activity of PCFT promoter is basically induced by KLF4 and the gradiented expression profile of PCFT may be at least in part accounted for by those of HNF4α, CDX2 and C/EBPα.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Transportador de Folato Acoplado a Protón/genética , Activación Transcripcional , Animales , Factor de Transcripción CDX2 , Metilación de ADN , Genes Reporteros , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Luciferasas/genética , Masculino , Regiones Promotoras Genéticas , Ratas , Ratas Wistar
13.
Pharm Res ; 30(2): 447-57, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23073666

RESUMEN

PURPOSE: To examine the effect of the fluoroquinolone DX-619 on CYP3A4 and urinary excretion of 6ß-hydroxycortisol, an endogenous probe of hepatic CYP3A4 activity, in healthy subjects. METHODS: The effect of DX-619 on CYP3A4 was examined in human liver microsomes. The apparent formation and renal clearance of 6ß-hydroxycortisol (CL(6ß-OHF) and CL(renal,6ß-OHF), respectively) were determined in placebo- and DX-619-treated subjects. 6ß-hydroxycortisol uptake was determined in HEK293 cells expressing OAT1, OAT3, OCT2, MATE1, and MATE2-K. RESULTS: DX-619 was a mechanism-based inhibitor of CYP3A4, with K(I) and k(inact) of 67.9 ± 7.3 µmol/l and 0.0730 ± 0.0033 min(-1), respectively. Pharmacokinetic simulation suggested in vivo relevance of CYP3A4 inhibition by DX-619. CL(6ß-OHF) and CL(renal,6ß-OHF) were decreased 72% and 70%, respectively, on day 15 in DX-619-treated group compared with placebo (P < 0.05). 6ß-hydroxycortisol was a substrate of OAT3 (K(m) = 183 ± 25 µmol/l), OCT2, MATE1, and MATE2-K. Maximum unbound concentration of DX-619 (9.1 ± 0.4 µmol/l) was above K(i) of DX-619 for MATE1 (4.32 ± 0.79 µmol/l). CONCLUSIONS: DX-619 caused a moderate inhibition of hepatic CYP3A4-mediated formation and significant inhibition of MATE-mediated efflux of 6ß-hydroxycortisol into urine. Caution is needed in applying CL(6ß-OHF) as an index of hepatic CYP3A4 activity without evaluating CL(renal,6ß-OHF).


Asunto(s)
Antibacterianos/metabolismo , Citocromo P-450 CYP3A/metabolismo , Hidrocortisona/análogos & derivados , Microsomas Hepáticos/metabolismo , Pirrolidinas/metabolismo , Quinolonas/metabolismo , Femenino , Células HEK293 , Humanos , Hidrocortisona/metabolismo , Hidrocortisona/orina , Masculino , Microsomas Hepáticos/efectos de los fármacos , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico
14.
Life Sci Alliance ; 6(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36456177

RESUMEN

Disrupted in renal carcinoma 2 (DIRC2) has gained interest because of its association with the development of renal cancer and cosegregation with a chromosomal translocation. It is a member of the SLC49 family (SLC49A4) and is considered to be an electrogenic lysosomal metabolite transporter; however, its molecular function has not been fully defined. To perform a detailed functional analysis of human DIRC2, we used a recombinant DIRC2 protein (DIRC2-AA), in which the N-terminal dileucine motif involved in its lysosomal localization was removed by replacing with dialanine for redirected localization to the plasma membrane, exposing intralysosomal segments to the extracellular space. The DIRC2-AA mutant induced the cellular uptake of pyridoxine (vitamin B6) under acidic conditions when expressed transiently in COS-7 cells. In addition, uptake was markedly inhibited by protonophores, indicating its function through an H+-coupled mechanism. In separate experiments, the transient overexpression of unmodified DIRC2 (tagged with HA) in human embryonic kidney 293 cells reduced cellular pyridoxine accumulation induced by transiently introduced human thiamine transporter 2/SLC19A3 (tagged with FLAG), a plasma membrane thiamine transporter that also transports pyridoxine. The cellular accumulation of pyridoxine in Caco-2 cells as a cell model was increased by the knockdown of endogenous DIRC2. Overall, the results indicate that DIRC2 is an H+-driven lysosomal pyridoxine exporter. Its overexpression leads to a reduction in cellular pyridoxine accumulation associated with reduced lysosomal accumulation and, conversely, its suppression results in an increase in lysosomal and cellular pyridoxine accumulation.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Proteínas de Transporte de Membrana , Humanos , Células CACO-2 , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Lisosomas , Proteínas de Transporte de Membrana/genética , Piridoxina , Tiamina
15.
J Pharmacol Exp Ther ; 340(2): 393-403, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22072731

RESUMEN

Cimetidine, an H2 receptor antagonist, has been used to investigate the tubular secretion of organic cations in human kidney. We report a systematic comprehensive analysis of the inhibition potency of cimetidine for the influx and efflux transporters of organic cations [human organic cation transporter 1 (hOCT1) and hOCT2 and human multidrug and toxin extrusion 1 (hMATE1) and hMATE2-K, respectively]. Inhibition constants (K(i)) of cimetidine were determined by using five substrates [tetraethylammonium (TEA), metformin, 1-methyl-4-phenylpyridinium, 4-(4-(dimethylamino)styryl)-N-methylpyridinium, and m-iodobenzylguanidine]. They were 95 to 146 µM for hOCT2, providing at most 10% inhibition based on its clinically reported plasma unbound concentrations (3.6-7.8 µM). In contrast, cimetidine is a potent inhibitor of MATE1 and MATE2-K with K(i) values (µM) of 1.1 to 3.8 and 2.1 to 6.9, respectively. The same tendency was observed for mouse Oct1 (mOct1), mOct2, and mouse Mate1. Cimetidine showed a negligible effect on the uptake of metformin by mouse kidney slices at 20 µM. Cimetidine was administered to mice by a constant infusion to achieve a plasma unbound concentration of 21.6 µM to examine its effect on the renal disposition of Mate1 probes (metformin, TEA, and cephalexin) in vivo. The kidney- and liver-to-plasma ratios of metformin both were increased 2.4-fold by cimetidine, whereas the renal clearance was not changed. Cimetidine also increased the kidney-to-plasma ratio of TEA and cephalexin 8.0- and 3.3-fold compared with a control and decreased the renal clearance from 49 to 23 and 11 to 6.6 ml/min/kg, respectively. These results suggest that the inhibition of MATEs, but not OCT2, is a likely mechanism underlying the drug-drug interactions with cimetidine in renal elimination.


Asunto(s)
Cimetidina/farmacología , Riñón/efectos de los fármacos , Proteínas de Transporte de Catión Orgánico/efectos de los fármacos , 1-Metil-4-fenilpiridinio/metabolismo , 3-Yodobencilguanidina/metabolismo , Animales , Unión Competitiva/fisiología , Transporte Biológico/efectos de los fármacos , Cefalexina/administración & dosificación , Cefalexina/sangre , Cefalexina/metabolismo , Cefalexina/farmacocinética , Cefalexina/orina , Cimetidina/administración & dosificación , Cimetidina/metabolismo , Cimetidina/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Células HEK293 , Humanos , Riñón/metabolismo , Cinética , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Metformina/administración & dosificación , Metformina/sangre , Metformina/metabolismo , Metformina/farmacocinética , Metformina/orina , Ratones , Ratones Endogámicos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 1 de Catión Orgánico/antagonistas & inhibidores , Transportador 1 de Catión Orgánico/efectos de los fármacos , Transportador 1 de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico , Piridinas/metabolismo , Tetraetilamonio/administración & dosificación , Tetraetilamonio/sangre , Tetraetilamonio/metabolismo , Tetraetilamonio/farmacocinética , Tetraetilamonio/orina , Transfección
16.
Drug Metab Pharmacokinet ; 44: 100456, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35512554

RESUMEN

The thiamine transporters, SLC19A2 and SLC19A3, have recently been shown to transport pyridoxine in addition to thiamine, the originally identified substrate, in our study on human orthologs. Based on these results, we characterized the rat and mouse orthologs for pyridoxine transport function. Through the assessment of pyridoxine uptake in human embryonic kidney 293 cells transiently expressing the SLC19A2/3 orthologs, we found that both rat and mouse Slc19a2 can transport pyridoxine, but rat or mouse Slc19a3 cannot. However, all SLC19A2/3 orthologs were capable of thiamine transport. We subsequently demonstrated in the rat small intestine that a carrier-mediated mechanism exists for thiamine uptake, but not for pyridoxine uptake. This is supported by the finding that rat Slc19a3, for which the human ortholog operates for the intestinal uptake of both pyridoxine and thiamine, lacks the pyridoxine transport function. Thus, SLC19A3s from different animal species exhibit differences in pyridoxine transport. Rats and mice, in which Slc19a3 lacks this function, are not suitable model animals for studies involving pyridoxine disposition and related issues.


Asunto(s)
Intestino Delgado , Proteínas de Transporte de Membrana , Piridoxina , Tiamina , Animales , Transporte Biológico , Intestino Delgado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Piridoxina/metabolismo , Ratas , Especificidad de la Especie , Tiamina/metabolismo
17.
Drug Metab Pharmacokinet ; 43: 100443, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35144162

RESUMEN

Orotate, a nutritional compound typically utilized as an intermediate in pyrimidine synthesis, has been suggested to undergo renal reabsorption. However, the detailed mechanisms involved in the process remain unclear, with only urate transporter 1 (URAT1/SLC22A12) being indicated as a transporter involved in its tubular uptake. As an attempt to identify transporters involved in that to help clarify the mechanisms, we examined a possibility that organic anion transporter 10 (OAT10/SLC22A13), which is present at the brush border membrane in renal tubular epithelial cells, could transport orotate. The operation of human OAT10 for orotate transport was demonstrated indeed and analyzed in detail in Madin-Darby canine kidney II cells introduced with this transporter by stable transfection. Orotate transport by OAT10 was found to be kinetically saturable with a biphasic characteristic and dependent on Cl-. These are unique characteristics previously unknown in its operation for the other substrates. Orotate transport by OAT10 was, on the other hand, inhibited by several anionic compounds known as OAT10 inhibitors. Finally, the rat ortholog of OAT10 was found not to be able to transport orotate, indicating animal species differences in that function. Thus, human OAT10 has been demonstrated to operate for orotate transport with unique characteristics.


Asunto(s)
Transportadores de Anión Orgánico , Animales , Transporte Biológico , Perros , Humanos , Riñón/metabolismo , Células de Riñón Canino Madin Darby , Microvellosidades/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Ratas
18.
J Biol Chem ; 285(9): 6522-31, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20042597

RESUMEN

Nucleobases are important compounds that constitute nucleosides and nucleic acids. Although it has long been suggested that specific transporters are involved in their intestinal absorption and uptake in other tissues, none of their molecular entities have been identified in mammals to date. Here we describe identification of rat Slc23a4 as the first sodium-dependent nucleobase transporter (rSNBT1). The mRNA of rSNBT1 was expressed highly and only in the small intestine. When transiently expressed in HEK293 cells, rSNBT1 could transport uracil most efficiently. The transport of uracil mediated by rSNBT1 was sodium-dependent and saturable with a Michaelis constant of 21.2 microM. Thymine, guanine, hypoxanthine, and xanthine were also transported, but adenine was not. It was also suggested by studies of the inhibitory effect on rSNBT1-mediated uracil transport that several nucleobase analogs such as 5-fluorouracil are recognized by rSNBT1, but cytosine and nucleosides are not or only poorly recognized. Furthermore, rSNBT1 fused with green fluorescent protein was mainly localized at the apical membrane, when stably expressed in polarized Madin-Darby canine kidney II cells. These characteristics of rSNBT1 were almost fully in agreement with those of the carrier-mediated transport system involved in intestinal uracil uptake. Therefore, it is likely that rSNBT1 is its molecular entity or at least in part responsible for that. It was also found that the gene orthologous to the rSNBT1 gene is genetically defective in humans. This may have a biological and evolutional meaning in the transport and metabolism of nucleobases. The present study provides novel insights into the specific transport and metabolism of nucleobases and their analogs for therapeutic use.


Asunto(s)
Absorción Intestinal , Proteínas de Transporte de Nucleobases/metabolismo , Purinas/metabolismo , Pirimidinas/metabolismo , Animales , Línea Celular , Perros , Humanos , Cinética , Datos de Secuencia Molecular , Ratas , Sodio , Especificidad de la Especie , Especificidad por Sustrato , Uracilo/metabolismo
19.
Cell Physiol Biochem ; 27(6): 749-56, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21691092

RESUMEN

BACKGROUND/AIMS: Although aquaglyceroporins have been generally believed to operate in a channel mode, which is of nonsaturable nature, for glycerol as well as for water, we recently found that human aquaporin 9 (hAQP9) operates in a carrier-mediated mode, which is of saturable nature, for glycerol. Based on the finding, we assumed that such a characteristic might be shared by the other aquaglyceroporins and examined the functional characteristics of hAQP10, which is an intestine-specific aquaglyceroporin. METHODS: Transport assays were conducted using Xenopus laevis oocytes expressing hAQP10 derived from the microinjected cRNA. RESULTS: The transport of glycerol by hAQP10 was found to be highly saturable with a Michaelis constant of 10.4 µM and specifically inhibited by several glycerol analogs such as monoacetin. Furthermore, when glycerol was preloaded in hAQP10-expressing oocytes, its efflux was trans-stimulated by extracellular glycerol. These results indicate the involvement of a carrier-mediated mechanism in glycerol transport by hAQP10. Interestingly, a channel mechanism was also found to be involved in part in hAQP10-mediated glycerol transport. CONCLUSION: The present study unveiled the uniquely dual functional characteristic of hAQP10 as a carrier/channel for solute transport, providing a novel insight into its operation mechanism, which would help further elucidate its physiological role.


Asunto(s)
Acuaporinas/fisiología , Animales , Acuaporinas/genética , Secuencia de Bases , Transporte Biológico , Cartilla de ADN , ADN Complementario , Femenino , Glicerol/metabolismo , Humanos , Cinética , Xenopus laevis
20.
J Pharmacol Exp Ther ; 333(1): 341-50, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20065018

RESUMEN

This report describes a potent and selective inhibitor of multidrug and toxin extrusion (MATE) protein, pyrimethamine (PYR), and examines its effect on the urinary and biliary excretion of typical Mate1 substrates in mice. In vitro inhibition studies demonstrated that PYR is a potent inhibitor of mouse (m)Mate1 (K(i) = 145 nM) among renal organic cation transporters mOctn1 and mOctn2 (K(i) > 30 microM), mOct1 (K(i) = 3.6 microM), and mOct2 (K(i) = 6.0 microM). PYR inhibited the uptake of metformin by kidney brush-border membrane vesicles (BBMVs) (K(i) = 41 nM) and canalicular membrane vesicles in the presence of outward gradient of H+. PYR treatment significantly increased the kidney-to-plasma ratio of tetraethylammonium, and both the liver- and kidney-to-plasma ratios of metformin in mice, whereas it did not affect their plasma concentrations and urinary excretion rates. Furthermore, the plasma lactate concentration, a biomarker for inhibition of gluconeogenesis by metformin, was significantly higher in the PYR-treated group than in the control group. These results not only suggest the importance of mMate1 in the efflux of organic cations into the urine and bile in mice but also the importance of canalicular efflux mediated by MATE proteins for the therapeutic efficacy of metformin. PYR is a potent inhibitor of human (h)MATE1 and hMATE2-K (K(i) = 77 and 46 nM, respectively) and H+ and organic cation exchanger in human kidney BBMVs (K(i) = 31 nM) in the presence of outward gradient of H+. Taken together, PYR can be used as a potent probe inhibitor of human MATE transporters.


Asunto(s)
Riñón/efectos de los fármacos , Hígado/efectos de los fármacos , Metformina/farmacocinética , Proteínas de Transporte de Catión Orgánico/fisiología , Pirimetamina/farmacología , Tetraetilamonio/farmacocinética , Animales , Canalículos Biliares/metabolismo , Transporte Biológico , Línea Celular , Humanos , Técnicas In Vitro , Riñón/metabolismo , Riñón/ultraestructura , Ácido Láctico/sangre , Hígado/metabolismo , Hígado/ultraestructura , Masculino , Metformina/farmacología , Ratones , Microvellosidades/metabolismo , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Pirimetamina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA