Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Sci Rep ; 11(1): 20563, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663856

RESUMEN

Brain age can be estimated using different Magnetic Resonance Imaging (MRI) modalities including diffusion MRI. Recent studies demonstrated that white matter (WM) tracts that share the same function might experience similar alterations. Therefore, in this work, we sought to investigate such issue focusing on five WM bundles holding that feature that is Association, Brainstem, Commissural, Limbic and Projection fibers, respectively. For each tract group, we estimated brain age for 15,335 healthy participants from United Kingdom Biobank relying on diffusion MRI data derived endophenotypes, Bayesian ridge regression modeling and 10 fold-cross validation. Furthermore, we estimated brain age for an Ensemble model that gathers all the considered WM bundles. Association analysis was subsequently performed between the estimated brain age delta as resulting from the six models, that is for each tract group as well as for the Ensemble model, and 38 daily life style measures, 14 cardiac risk factors and cardiovascular magnetic resonance imaging features and genetic variants. The Ensemble model that used all tracts from all fiber groups (FG) performed better than other models to estimate brain age. Limbic tracts based model reached the highest accuracy with a Mean Absolute Error (MAE) of 5.08, followed by the Commissural ([Formula: see text]), Association ([Formula: see text]), and Projection ([Formula: see text]) ones. The Brainstem tracts based model was the less accurate achieving a MAE of 5.86. Accordingly, our study suggests that the Limbic tracts experience less brain aging or allows for more accurate estimates compared to other tract groups. Moreover, the results suggest that Limbic tract leads to the largest number of significant associations with daily lifestyle factors than the other tract groups. Lastly, two SNPs were significantly (p value [Formula: see text]) associated with brain age delta in the Projection fibers. Those SNPs are mapped to HIST1H1A and SLC17A3 genes.


Asunto(s)
Encéfalo/fisiología , Sustancia Blanca/diagnóstico por imagen , Factores de Edad , Envejecimiento , Teorema de Bayes , Encéfalo/patología , Bases de Datos Genéticas , Imagen de Difusión por Resonancia Magnética/métodos , Imagen de Difusión Tensora/métodos , Femenino , Cardiopatías , Histonas/genética , Histonas/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Modelos Biológicos , Factores de Riesgo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Reino Unido/epidemiología , Sustancia Blanca/patología , Sustancia Blanca/fisiología
2.
Biol Pharm Bull ; 43(11): 1653-1659, 2020 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-32863294

RESUMEN

Hyperuricemia is mainly the result of relative underexcretion of urate. Urate is mainly eliminated by kidney and several important transporters expressed on the membrane of renal tubular cells involved in urate excretion. Olsalazine sodium was screened from 3167 authorized small compounds/drugs, targeting xanthine oxidoreductase. In previous study, we reported that olsalazine sodium significantly reduced the serum urate levels, and the anti-hyperuricemic activity linked with inhibiting urate formation by reducing the activity of xanthine oxidoreductase. The current research aimed to assess olsalazine sodium renal urate excretion and likely molecular mechanism. The results showed that administration of olsalazine sodium 5.0 mg/kg decreased the levels of serum urate in hyperuricemic rats, and noticeably improved the fractional excretion of urate and urate clearance, exhibiting an uricosuric action. Moreover, olsalazine sodium (2.5, 5.0, 10.0 mg/kg) reduced the level of blood urea nitrogen in rats. Further study showed that olsalazine sodium reduced the mRNA expression of urate reabsorptive transporter glucose transporter 9 (GLUT9), increased the mRNA expression of urate secretory transporters, organic anion transporter 1 (OAT1), OAT3 and type 1 sodium-dependent phosphate transporter (NPT1) as well as the protein expression of OAT3 in the kidney in hyperuricemic mice. In conclusion, olsalazine sodium exhibited a promotion of urate excretion in kidney by increasing the expression of OAT3.


Asunto(s)
Ácidos Aminosalicílicos/farmacología , Hiperuricemia/tratamiento farmacológico , Transportadores de Anión Orgánico Sodio-Independiente/agonistas , Eliminación Renal/efectos de los fármacos , Ácido Úrico/metabolismo , Ácidos Aminosalicílicos/uso terapéutico , Animales , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Creatinina/orina , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Proteínas Facilitadoras del Transporte de la Glucosa/antagonistas & inhibidores , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Humanos , Hiperuricemia/sangre , Hiperuricemia/fisiopatología , Hiperuricemia/orina , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/fisiopatología , Masculino , Ratones , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , Proteínas de Transporte de Monosacáridos/metabolismo , Proteína 1 de Transporte de Anión Orgánico/agonistas , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Ratas , Ratas Sprague-Dawley , Eliminación Renal/fisiología , Reabsorción Renal/efectos de los fármacos , Reabsorción Renal/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/agonistas , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Ácido Úrico/sangre , Ácido Úrico/orina
3.
Nat Commun ; 9(1): 4455, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30367059

RESUMEN

Thyroid dysfunction is an important public health problem, which affects 10% of the general population and increases the risk of cardiovascular morbidity and mortality. Many aspects of thyroid hormone regulation have only partly been elucidated, including its transport, metabolism, and genetic determinants. Here we report a large meta-analysis of genome-wide association studies for thyroid function and dysfunction, testing 8 million genetic variants in up to 72,167 individuals. One-hundred-and-nine independent genetic variants are associated with these traits. A genetic risk score, calculated to assess their combined effects on clinical end points, shows significant associations with increased risk of both overt (Graves' disease) and subclinical thyroid disease, as well as clinical complications. By functional follow-up on selected signals, we identify a novel thyroid hormone transporter (SLC17A4) and a metabolizing enzyme (AADAT). Together, these results provide new knowledge about thyroid hormone physiology and disease, opening new possibilities for therapeutic targets.


Asunto(s)
2-Aminoadipato-Transaminasa/metabolismo , Regulación de la Expresión Génica/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Hormonas Tiroideas/genética , Tirotropina/metabolismo , 2-Aminoadipato-Transaminasa/genética , Animales , Transporte Biológico , Células COS , Chlorocebus aethiops , Estudio de Asociación del Genoma Completo , Humanos , Hipertiroidismo/genética , Hipertiroidismo/fisiopatología , Hipotiroidismo/genética , Hipotiroidismo/fisiopatología , Polimorfismo de Nucleótido Simple , Factores de Riesgo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Glándula Tiroides/metabolismo , Glándula Tiroides/fisiopatología , Hormonas Tiroideas/metabolismo , Población Blanca
4.
Biopharm Drug Dispos ; 39(7): 328-334, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29975986

RESUMEN

Indoxyl sulfate (IS) is a protein-bound uremic toxin that progressively accumulates in plasma during chronic kidney disease (CKD), and its accumulation is associated with the progression of CKD. This study examined the intestinal secretion of IS using in situ single-pass intestinal perfusion in a rat model of renal insufficiency, MRP2- and BCRP-overexpressing Sf9 membrane vesicles, and Caco-2 cell monolayers. An in situ single-pass perfusion study in CKD model rats demonstrated that a small amount of IS is secreted into intestinal lumen after iv administration of IS, and the clearance increased AUC-dependently. An excess amount of IS (3 mm) partially inhibited the MRP2- and BCRP-mediated uptake of specific fluorescent substrates, CDCF and Lucifer yellow, respectively, into the membrane vesicles, although IS was not taken up at a physiological concentration, 10 µm. In the Caco-2 cell monolayers, the IS transport was higher in the absorptive direction than in the secretory direction (p < 0.05). p-Aminohippuric acid (PAH) strongly inhibited IS transport in both directions (absorptive, p = 0.142; secretory, p < 0.01). Given that the blood IS levels are much higher than those in the intestinal lumen, it is possible that this unknown PAH-sensitive system contributes to the intestinal IS secretion. Although in situ inhibition study is needed to confirm that this unknown transporter mediates the in vivo intestinal secretion of IS, we speculate that this unknown active efflux system works as a compensatory excretion pathway for excess organic anions such as IS especially in end-stage renal disease.


Asunto(s)
Indicán/metabolismo , Yeyuno/metabolismo , Insuficiencia Renal Crónica/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Células CACO-2 , Humanos , Secreciones Intestinales/metabolismo , Masculino , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Ratas Sprague-Dawley , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo
5.
Sci Rep ; 8(1): 3848, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29497127

RESUMEN

The aims of this study were to identify candidate pathways associated with serum urate and to explore the genetic effect of those pathways on the risk of gout. Pathway analysis of the loci identified in genome-wide association studies (GWASs) showed that the ion transmembrane transporter activity pathway (GO: 0015075) and the secondary active transmembrane transporter activity pathway (GO: 0015291) were both associated with serum urate concentrations, with PFDR values of 0.004 and 0.007, respectively. In a Chinese population of 4,332 individuals, the two pathways were also found to be associated with serum urate (PFDR = 1.88E-05 and 3.44E-04, separately). In addition, these two pathways were further associated with the pathogenesis of gout (PFDR = 1.08E-08 and 2.66E-03, respectively) in the Chinese population and a novel gout-associated gene, SLC17A2, was identified (OR = 0.83, PFDR = 0.017). The mRNA expression of candidate genes also showed significant differences among different groups at pathway level. The present study identified two transmembrane transporter activity pathways (GO: 0015075 and GO: 0015291) were associations with serum urate concentrations and the risk of gout. SLC17A2 was identified as a novel gene that influenced the risk of gout.


Asunto(s)
Gota/genética , Ácido Úrico/análisis , Pueblo Asiatico/genética , Estudios de Casos y Controles , China , Femenino , Sitios Genéticos , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Genotipo , Gota/metabolismo , Humanos , Hiperuricemia/genética , Transporte Iónico/genética , Transporte Iónico/fisiología , Masculino , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Ácido Úrico/sangre
6.
Chem Biol Interact ; 276: 15-22, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28254523

RESUMEN

ALDH16A1 is a novel member of the ALDH superfamily that is enzymatically-inactive and highly expressed in the kidney. Recent studies identified an association between a rare missense single nucleotide variant (SNV) in the ALDH16A1 gene and elevated serum uric acid levels and gout. The present study explores the mechanisms by which ALDH16A1 influences uric acid homeostasis in the kidney. We generated and validated a mouse line with global disruption of the Aldh16a1 gene through gene targeting and performed RNA-seq analyses in the kidney of wild-type (WT) and Aldh16a1 knockout (KO) mice, along with plasma metabolomics. We found that ALDH16A1 is expressed in proximal and distal convoluted tubule cells in the cortex of the kidney and in zone 3 hepatocytes. RNA-seq and gene ontology enrichment analyses showed that cellular lipid and lipid metabolic processes are up-regulated. Three transporters localized in the apical membrane of the proximal convoluted tubule of the kidney known to influence urate/uric acid homeostasis were found to be up-regulated (Abcc4, Slc16a9) or down-regulated (Slc17a3). An initial metabolomics analysis in plasma revealed an altered lipid profile in KO mice that is in agreement with our RNA-seq analysis. This is the first study demonstrating a functional role of ALDH16A1 in the kidney.


Asunto(s)
Aldehído Deshidrogenasa/genética , Riñón/metabolismo , Aldehído Deshidrogenasa/deficiencia , Animales , Regulación hacia Abajo , Perfilación de la Expresión Génica , Riñón/patología , Lípidos/sangre , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Mutación Missense , Análisis de Secuencia de ARN , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Regulación hacia Arriba
7.
Nucleosides Nucleotides Nucleic Acids ; 35(10-12): 536-542, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27906618

RESUMEN

Human sodium-dependent phosphate cotransporter type 1 (NPT1/SLC17A1) is one of the urate transporters in the kidney. Our recent study revealed that a common missense variant, I269T (rs1165196), of NPT1 decreases the risk of renal underexcretion gout. Moreover, we demonstrated that human NPT1 is localized to the apical membrane of the renal proximal tubule, and that I269T is the gain-of-function variant which increases the NPT1-mediated urate export. However, the mechanism by which I269T variant increases the urate export remains to be clarified. Thus, we performed immunostaining and functional analysis of human NPT1 using the Xenopus oocyte expression system. For comparison of human NPT1 expression levels of oocyte membrane between 269I (wild type) and 269T (variant), immunostaining was performed with anti-human NPT1 antibodies. As a result, we showed that NPT1 I269T variant did not change the human NPT1 membrane expression levels, although NPT1 I269T variant increased the urate transport compared with NPT1 wild type. Combined with the previous report that I269T variant did not induce Km changes but increased the Vmax of urate transport in a proteoliposome system, our findings suggest that I269T variant increases NPT1-mediated urate export without increase of NPT1 expression levels on the membrane. Thus, I269T, a common missense variant of NPT1, might have faster conformation changes than NPT1 wild type in terms of the alternating-access model of transporters, and increases renal urate export in humans.


Asunto(s)
Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Ácido Úrico/metabolismo , Animales , Transporte Biológico , Células Cultivadas , Estudios de Asociación Genética , Humanos , Mutación Missense , Oocitos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Xenopus laevis
8.
Am J Physiol Cell Physiol ; 309(2): C71-80, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25972451

RESUMEN

Membrane potential (Δψ)-driven and Cl(-)-dependent organic anion transport is a primary function of the solute carrier family 17 (SLC17) transporter family. Although the transport substrates and physiological relevance of the major members are well understood, SLC17A2 protein known to be Na(+)-phosphate cotransporter 3 (NPT3) is far less well characterized. In the present study, we investigated the transport properties and expression patterns of mouse SLC17A2 protein (mNPT3). Proteoliposomes containing the purified mNPT3 protein took up radiolabeled p-aminohippuric acid (PAH) in a Δψ- and Cl(-)-dependent manner. The mNPT3-mediated PAH uptake was inhibited by 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDs) and Evans blue, common inhibitors of SLC17 family members. The PAH uptake was also inhibited by various anionic compounds, such as hydrophilic nonsteroidal anti-inflammatory drugs (NSAIDs) and urate. Consistent with these observations, the proteoliposome took up radiolabeled urate in a Δψ- and Cl(-)-dependent manner. Immunohistochemistry with specific antibodies against mNPT3 combined with RT-PCR revealed that mNPT3 is present in various tissues, including the hepatic bile duct, luminal membranes of the renal urinary tubules, maternal side of syncytiotrophoblast in the placenta, apical membrane of follicle cells in the thyroid, bronchiole epithelial cells in the lungs, and astrocytes around blood vessels in the cerebrum. These results suggested that mNPT3 is a polyspecific organic anion transporter that is involved in circulation of urate throughout the body.


Asunto(s)
Membrana Celular/metabolismo , Cloruros/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Ácido Úrico/metabolismo , Animales , Transporte Biológico , Membrana Celular/efectos de los fármacos , Regulación de la Expresión Génica , Hipuratos/metabolismo , Cinética , Potenciales de la Membrana , Ratones Endogámicos C57BL , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/antagonistas & inhibidores , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética
11.
Curr Mol Pharmacol ; 6(2): 88-94, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23876149

RESUMEN

SLC17A1 protein (NPT1) was the first identified member of the SLC17 phosphate transporter family, and is known to mediate Na(+)/inorganic phosphate (Pi) co-transport when expressed in Xenopus oocytes. Although this protein was suggested to be a renal polyspecific anion exporter, its transport properties were not well characterized. The clean biochemical approach revealed that proteoliposomes comprising purified NPT1 as the only protein source transport various organic anions such as urate, p-aminohippuric acid (PAH), and acetylsalicylic acid (aspirin) in a membrane potential (Δψ)-driven and Cl(-) -dependent manner. Human NPT1 carrying an SNP mutation, Thr269Ile, known to increase the risk of gout, exhibited 32% lower urate transport activity compared to the wild type protein, leading to the conclusion that NPT1 is the long searched for transporter responsible for renal urate excretion. In the present article, we summarized the history of identification of the urate exporter and its possible involvement in the dynamism of urate under physiological and pathological conditions.


Asunto(s)
Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Ácido Úrico/metabolismo , Secuencia de Aminoácidos , Animales , Gota/genética , Gota/metabolismo , Humanos , Potenciales de la Membrana , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/análisis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética
12.
Poult Sci ; 92(8): 2101-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23873558

RESUMEN

Two 21-d broiler experiments were conducted to assess the efficacy of a bacterial 6-phytase expressed in Aspergillus oryzae on growth performance, nutrient utilization, and intestinal molecular markers. Two hundred forty birds in 5 treatments (experiment 1) or 256 birds in 4 treatments (experiment 2) were used. The treatments included a negative control diet that was marginally deficient in P (NC) or NC plus tricalcium phosphate, 500, 1,000, or 2,000 phytase units/kg (experiment 1), and NC or NC plus monocalcium phosphate, 500 or 1,000 phytase units/kg (experiment 2). In both experiments, excreta were collected on d 19 to 21, whereas birds and feed were weighed and ileal digesta collected on d 21. For experiment 1, mucosa scraping was collected from the duodenum, jejunum, and ileum from all birds for quantification of expression level of gut level inflammatory cytokines, Toll-like receptors, and phosphate transporter (NaPi-IIb). In both experiments, tricalcium phosphate, monocalcium phosphate, and phytase supplementation improved (P < 0.05) weight gain and percentage tibia ash. Phosphorus and Ca retention and phytic acid disappearance improved (P < 0.05) with phytase supplementation (experiment 1) and there was an increase (P < 0.01) in Ca and P retention in response to phytase supplementation (experiment 2). Diets did not affect the expression of gut level cytokines, Toll-like receptors, or the mucin gene. Phytase supplementation tended to decrease (P < 0.10) the expression of NaPi-IIb. It was concluded from these studies that the bacterial 6-phytase was effective in enhancing growth of broilers receiving low-P diets as well as in increasing efficiency of P utilization and phytic acid degradation.


Asunto(s)
6-Fitasa/farmacología , Pollos/crecimiento & desarrollo , 6-Fitasa/metabolismo , Alimentación Animal/análisis , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Aspergillus oryzae/metabolismo , Calcio/metabolismo , Dieta/veterinaria , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Masculino , Mucina 2/genética , Mucina 2/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo
13.
Cell ; 154(2): 267-8, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23870116

RESUMEN

The wiring of the nervous system requires that axons navigate to the correct targets and maintain their correct positions during developmental growth. In this issue, Shao et al. (2013) now reveal a crucial new role for glia in preserving correct synaptic connectivity during developmental growth.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Neuroglía/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Sinapsis , Animales
14.
Cell ; 154(2): 337-50, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23870123

RESUMEN

Synaptic contacts are largely established during embryogenesis and are then maintained during growth. To identify molecules involved in this process, we conducted a forward genetic screen in C. elegans and identified cima-1. In cima-1 mutants, synaptic contacts are correctly established during embryogenesis, but ectopic synapses emerge during postdevelopmental growth. cima-1 encodes a solute carrier in the SLC17 family of transporters that includes sialin, a protein that when mutated in humans results in neurological disorders. cima-1 does not function in neurons but rather functions in the nearby epidermal cells to correctly position glia during postlarval growth. Our findings indicate that CIMA-1 antagonizes the FGF receptor (FGFR), and does so most likely by inhibiting FGFR's role in epidermal-glia adhesion rather than signaling. Our data suggest that epidermal-glia crosstalk, in this case mediated by a transporter and the FGF receptor, is vital to preserve embryonically derived circuit architecture during postdevelopmental growth.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Neuroglía/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Sinapsis , Animales , Tamaño Corporal , Caenorhabditis elegans/embriología , Proteínas de Caenorhabditis elegans/genética , Desarrollo Embrionario , Células Epidérmicas , Epidermis/metabolismo , Mutación , Neuritas/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética
15.
Am J Physiol Cell Physiol ; 302(11): C1652-60, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22460716

RESUMEN

The SLC17 anion transporter family comprises nine members that transport various organic anions in membrane potential (Δψ)- and Cl(-)-dependent manners. Although the transport substrates and physiological relevance of the majority of the members have already been determined, little is known about SLC17A4 proteins known to be Na(+)-phosphate cotransporter homologue (NPT homologue). In the present study, we investigated the expression and transport properties of human SLC17A4 protein. Using specific antibodies, we found that a human NPT homologue is specifically expressed and present in the intestinal brush border membrane. Proteoliposomes containing the purified protein took up radiolabeled p-aminohippuric acid (PAH) in a Cl(-)-dependent manner at the expense of an electrochemical gradient of protons, especially Δψ, across the membrane. The Δψ- and Cl(-)-dependent PAH uptake was inhibited by diisothiocyanostilbene-2,2'-disulfonic acid and Evans blue, common inhibitors of SLC17 family members. cis-Inhibition studies revealed that various anionic compounds, such as hydrophilic nonsteroidal anti-inflammatory drugs, pravastatin, and urate inhibited the PAH uptake. Proteoliposomes took up radiolabeled urate, with the uptake having properties similar to those of PAH uptake. These results strongly suggested that the human NPT homologue acts as a polyspecific organic anion exporter in the intestines. Since SLC17A1 protein (NPT1) and SLC17A3 protein (NPT4) are responsible for renal urate extrusion, our results reveal the possible involvement of a NPT homologue in urate extrusion from the intestinal duct.


Asunto(s)
Intestino Delgado/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Animales , Transporte Biológico , Humanos , Intestino Delgado/fisiología , Masculino , Potenciales de la Membrana , Ratones , Pravastatina/farmacología , Proteolípidos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/inmunología , Ácido Úrico/metabolismo , Ácido Úrico/farmacología , Ácido p-Aminohipúrico/metabolismo
16.
J Pharmacol Sci ; 116(4): 392-6, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21778665

RESUMEN

In the present study, we investigated the transport of nephrotoxic mycotoxin ochratoxin A (OTxA) by a novel human organic anion transporter hNPT4 using the Xenopus oocyte expression system. hNPT4 mediated time- and concentration-dependent uptake of OTxA (K(m): 802.8 µM) in a pH- and voltage-sensitive manner. Cis-inhibition experiments suggest that the substrate selectivity of hNPT4 is similar to that of hOAT4. The fact that the K(m) of OTxA for the efflux transporter hNPT4 was much higher than those for the uptake transporters hOAT1 and hOAT3 may favor the accumulation of OTxA in the tubular cell and lead to nephrotoxicity.


Asunto(s)
Ocratoxinas/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Animales , Transporte Biológico/fisiología , Humanos , Oocitos , Xenopus
17.
Br J Nutr ; 105(11): 1619-26, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21443813

RESUMEN

In goats, the combination of dietary N and Ca reduction caused hypocalcaemia and further changes in Ca homeostasis. The aim of the present study was to characterise the effects of dietary N reduction under normocalcaemia on mineral and bone metabolism in young goats. Young male goats of the Saanen breed were fed a diet reduced in N (8 %) for about 7 weeks (ten animals per group) and were compared with goats fed with an adequate N (14 %) diet. When N intake was reduced in young goats, plasma urea concentrations as well as renal elimination of urea were reduced. This was inversely related to creatinine in plasma and urine, which increased during a dietary N reduction as a function of reduced renal activity to save urea during N scarcity. During this decrease in renal function, associated with declined insulin-like growth factor 1 concentrations, a reduction in calcidiol and calcitriol concentrations could be observed. Meanwhile, carboxyterminal cross-linked telopeptide of type I collagen values and activity of total alkaline phosphatase were both elevated, indicating some bone remodelling processes taking place during a reduced N diet in young goats. The concentrations of inorganic phosphate (Pi) and total Ca were changed in several body fluids, indicating that Pi and Ca homeostasis was perturbed in goats fed a reduced N diet. Therefore, more research is needed to find the balance between reduction of environmental N pollution by reducing dietary N in ruminant feeding and maintaining the animal's health.


Asunto(s)
Alimentación Animal/análisis , Dieta/veterinaria , Electrólitos/metabolismo , Cabras/crecimiento & desarrollo , Homeostasis/fisiología , Nitrógeno/administración & dosificación , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Transporte Biológico , Western Blotting , Líquidos Corporales/química , Líquidos Corporales/metabolismo , Calcio/metabolismo , AMP Cíclico/metabolismo , Ingestión de Alimentos , Regulación de la Expresión Génica , Cabras/sangre , Intestino Delgado/metabolismo , Masculino , Microvellosidades/metabolismo , Nitrógeno/química , Nitrógeno/metabolismo , Fosfatos/química , Fósforo/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Aumento de Peso/efectos de los fármacos
18.
J Pharmacol Sci ; 115(2): 249-53, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21282933

RESUMEN

We analyzed the functional properties of five nonsynonymous single nucleotide polymorphisms (SNPs) in the sodium-phosphate transporter NPT4 gene (SLC17A3) using the Xenopus oocyte expression system. NPT4 variants carrying SNP V257F, G279R, or P378L exhibited reduced transport of [(14)C]para-aminohippurate, [(3)H]bumetanide, [(3)H]estrone sulfate, and [(14)C]urate, when each variant clone was expressed in the plasma membrane of oocytes. This study suggests the possibility that the genetic variation of NPT4 contributes to inter-individual differences in disposition of anionic drugs such as diuretics as well as certain endogenous organic anions such as urate.


Asunto(s)
Polimorfismo de Nucleótido Simple , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Animales , Antioxidantes/farmacocinética , Transporte Biológico , Bumetanida/farmacocinética , Membrana Celular/metabolismo , Diuréticos/farmacocinética , Estrona/análogos & derivados , Estrona/farmacocinética , Humanos , Oocitos , Ácido Úrico/farmacocinética , Xenopus , Ácido p-Aminohipúrico/metabolismo
19.
J Biol Chem ; 285(45): 35123-32, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20810651

RESUMEN

The evolutionary loss of hepatic urate oxidase (uricase) has resulted in humans with elevated serum uric acid (urate). Uricase loss may have been beneficial to early primate survival. However, an elevated serum urate has predisposed man to hyperuricemia, a metabolic disturbance leading to gout, hypertension, and various cardiovascular diseases. Human serum urate levels are largely determined by urate reabsorption and secretion in the kidney. Renal urate reabsorption is controlled via two proximal tubular urate transporters: apical URAT1 (SLC22A12) and basolateral URATv1/GLUT9 (SLC2A9). In contrast, the molecular mechanism(s) for renal urate secretion remain unknown. In this report, we demonstrate that an orphan transporter hNPT4 (human sodium phosphate transporter 4; SLC17A3) was a multispecific organic anion efflux transporter expressed in the kidneys and liver. hNPT4 was localized at the apical side of renal tubules and functioned as a voltage-driven urate transporter. Furthermore, loop diuretics, such as furosemide and bumetanide, substantially interacted with hNPT4. Thus, this protein is likely to act as a common secretion route for both drugs and may play an important role in diuretics-induced hyperuricemia. The in vivo role of hNPT4 was suggested by two hyperuricemia patients with missense mutations in SLC17A3. These mutated versions of hNPT4 exhibited reduced urate efflux when they were expressed in Xenopus oocytes. Our findings will complete a model of urate secretion in the renal tubular cell, where intracellular urate taken up via OAT1 and/or OAT3 from the blood exits from the cell into the lumen via hNPT4.


Asunto(s)
Bumetanida/farmacocinética , Diuréticos/farmacocinética , Furosemida/farmacocinética , Túbulos Renales Proximales/metabolismo , Modelos Biológicos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Ácido Úrico/metabolismo , Animales , Bumetanida/efectos adversos , Bumetanida/farmacología , Diuréticos/efectos adversos , Diuréticos/farmacología , Furosemida/efectos adversos , Furosemida/farmacología , Predisposición Genética a la Enfermedad , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Gota/genética , Gota/metabolismo , Humanos , Hiperuricemia/inducido químicamente , Hiperuricemia/genética , Hiperuricemia/metabolismo , Transporte Iónico/efectos de los fármacos , Transporte Iónico/genética , Hígado/metabolismo , Mutación Missense , Proteína 1 de Transporte de Anión Orgánico/genética , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/genética , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Urato Oxidasa/genética , Urato Oxidasa/metabolismo , Xenopus laevis
20.
Bone ; 47(5): 938-47, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20709201

RESUMEN

As chondrocytes mature, the concentration of inorganic phosphate (Pi) increases in the extracellular milieu. It was demonstrated that the progressive accumulation of Pi started from the proliferative zone and peaked in the hypertrophic zone of growth plate. Although extracellular Pi is reported to be involved in the apoptosis and mineralization of mature chondrocytes, its role in proliferating chondrocytes remains unclear. Here we investigated this role utilizing ATDC5, an established cell model of chondrocytic differentiation. In proliferating ATDC5 cells, we found that the expression of cyclin D1 was up-regulated, and that of alkaline phosphatase (ALP) was down-regulated in response to an increase in extracellular Pi within 24h. Moreover, an increase in extracellular Pi-induced activation of the Raf/MEK/ERK pathway, and treatment with a MEK inhibitor PD98059 abolished the effects on the expression of cyclin D1 and ALP, indicating that extracellular Pi regulates the expression of these genes through the Raf/MEK/ERK pathway. Consistent with its up-regulation of cyclin D1 expression, the extracellular Pi facilitated the proliferation of ATDC5 cells. Treatment with phosphonoformic acid (PFA), an inhibitor of sodium/phosphate (Na(+)/Pi) cotransporters, abrogated the activation of the Raf/MEK/ERK pathway and gene expression induced by the increase in extracellular Pi. Knocking down of the type III Na(+)/Pi cotransporter Pit-1 diminished the responsiveness of ATDC5 cells to the increase in extracellular Pi. Interestingly, the increased extracellular Pi induced the phosphorylation of fibroblast growth factor receptor substrate 2α (FRS2α), which was also cancelled by knocking down of the expression of Pit-1. In primary chondrocytes isolated from mouse rib cages as well, increased extracellular Pi induced the phosphorylation of ERK1/2 and alterations in the expression of cyclin D1 and ALP, both of which were abolished by treatment with PFA. These results suggest that signaling by extracellular Pi is mediated by Pit-1 and FRS2α, and leads to activation of the Raf/MEK/ERK pathway and increased expression of cyclin D1, which facilitates the proliferation of immature chondrocytes.


Asunto(s)
Condrocitos/metabolismo , Ciclina D1/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosfatos/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/metabolismo , Quinasas raf/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Western Blotting , Línea Celular , Condrocitos/citología , Condrocitos/efectos de los fármacos , Ciclina D1/genética , Electroforesis en Gel Bidimensional , Inhibidores Enzimáticos , Quinasas MAP Reguladas por Señal Extracelular/genética , Flavonoides/farmacología , Foscarnet/farmacología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Quinasas raf/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...