Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 20(4): 1061-1065, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29171930

RESUMEN

An integrated analysis was performed with data from 4 phase 2 and phase 3 studies of tofogliflozin in which patients with type 2 diabetes mellitus received the sodium-glucose cotransporter 2 inhibitor tofogliflozin for up to 24 weeks. Sex differences, baseline haemoglobin A1c (HbA1c) and serum uric acid (UA) levels, and log10 -transformed urinary N-acetyl-ß-D-glucosaminidase ratio were significantly correlated with the reduction in serum UA levels at both 4 and 24 weeks in multivariate analysis (respectively, P < .0001). The decrease in HbA1c levels was greatest in the group with the highest baseline HbA1c level (quartile 4; HbA1c > 8.6%) and lowest in the group with the lowest baseline HbA1c level (quartile 1; HbA1c ≤ 7.4%). The decrease in serum UA levels was greatest in the quartile 1 group and lowest in the quartile 4 group. In most groups, the maximum decrease in serum UA levels was seen in the first 4 weeks, while the maximum decrease in HbA1c was seen at week 24. Thus, serum UA levels were significantly decreased in patients with moderate HbA1c levels.


Asunto(s)
Compuestos de Bencidrilo/uso terapéutico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucósidos/uso terapéutico , Hemoglobina Glucada/efectos de los fármacos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Ácido Úrico/sangre , Adulto , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Ensayos Clínicos Fase II como Asunto/estadística & datos numéricos , Ensayos Clínicos Fase III como Asunto/estadística & datos numéricos , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/epidemiología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
2.
Bioorg Med Chem Lett ; 28(10): 1915-1918, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29655983

RESUMEN

p-Boronophenylalanine (l-BPA) is applied in clinical settings as a boron carrier for boron neutron capture therapy (BNCT) to cure malignant melanomas. Structural modification or derivatization of l-BPA, however, to improve its uptake efficiency into tumor cells has scarcely been investigated. We successfully synthesized (S)-2-amino-3-(4-boronophenyl)-2-methylpropanoic acid in enantioenriched form as a novel candidate molecule for BNCT. Key steps to enhance the efficiency of this synthesis were enantioselective alkylation of N-protected alanine tert-butyl ester with a Maruoka catalyst and Miyaura borylation reaction to install the boron functionality.


Asunto(s)
Compuestos de Boro/química , Fenilalanina/análogos & derivados , Alquilación , Compuestos de Boro/síntesis química , Catálisis , Complejos de Coordinación/química , Paladio/química , Fenilalanina/síntesis química , Fenilalanina/química , Estereoisomerismo
3.
J Pharmacol Sci ; 136(4): 196-202, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29656005

RESUMEN

The purpose of this study was to create novel urate under-excretion animal models using pyrazinamide and to evaluate whether dihydropyridine calcium channel blockers (CCBs) have uricosuric effects in vivo. Adult male ICR mice were treated with pyrazinamide, vehicle (dimethyl sulfoxide: DMSO), or tap water. Thirty minutes later, pyrazinamide-treated mice were given benzbromarone, losartan, nilvadipine, nitrendipine, nifedipine or azelnidipine. Six hours after the second administration, urine (by urinary bladder puncture) and plasma were collected to measure uric acid and creatinine levels, and fractional excretion of uric acid (FEUA) and creatinine clearance (Ccr) were calculated and evaluated. There was no significant difference in the levels of plasma uric acid, plasma creatinine, Ccr, urinary N-acetyl-ß-d-glucosaminidase (NAG) and urinary NAG-creatinine ratio between water, DMSO, and pyrazinamide-treated mice. But the FEUA of pyrazinamide-treated mice was significantly lower than water mice. The FEUA was significantly higher in mice taking the dihydropyridine CCBs (nilvadipine, nitrendipine, nifedipine, and high-dose azelnidipine) than in pyrazinamide-treated mice. There was no significant difference in Ccr. Thus, a novel animal model created with PZA administration was useful as a urate under-excretion animal model that was probably URAT1-mediated, and the uricosuric effects of dihydropyridine CCBs were confirmed in vivo.


Asunto(s)
Bloqueadores de los Canales de Calcio/farmacología , Dihidropiridinas/farmacología , Modelos Animales , Uricosúricos , Animales , Creatinina/sangre , Creatinina/orina , Proteínas de Unión al ADN , Masculino , Ratones Endogámicos ICR , Transportadores de Anión Orgánico , Ácido Úrico/sangre , Ácido Úrico/orina
4.
Tumour Biol ; 39(3): 1010428317694545, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28347255

RESUMEN

Unlike normal cells, cancer cells undergo unlimited growth and multiplication, causing them to require massive amounts of amino acid to support their continuous metabolism. Among the amino acid transporters expressed on the plasma membrane, l-type amino acid transporter-1, a Na+-independent neutral amino acid transporter, is highly expressed in many types of human cancer including cholangiocarcinoma. Our previous study reported that l-type amino acid transporter-1 and its co-functional protein CD98 were highly expressed and implicated in cholangiocarcinoma progression and carcinogenesis. Therefore, this study determined the effect of JPH203, a selective inhibitor of l-type amino acid transporter-1 activity, on cholangiocarcinoma cell inhibition both in vitro and in vivo. JPH203 dramatically suppressed [14C]l-leucine uptake as well as cell growth in cholangiocarcinoma cell lines along with altering the expression of l-type amino acid transporter-1 and CD98 in response to amino acid depletion. We also demonstrated that JPH203 induced both G2/M and G0/G1 cell cycle arrest, as well as reduced the S phase accompanied by altered expression of the proteins in cell cycle progression: cyclin D1, CDK4, and CDK6. There was also cell cycle arrest of the related proteins, P21 and P27, in KKU-055 and KKU-213 cholangiocarcinoma cells. Apoptosis induction, detected by an increase in trypan blue-stained cells along with a cleaved caspase-3/caspase-3 ratio, occurred in JPH203-treated cholangiocarcinoma cells at the highest concentration tested (100 µM). As expected, daily intravenous administration of JPH203 (12.5 and 25 mg/kg) significantly inhibited tumor growth in KKU-213 cholangiocarcinoma cell xenografts in the nude mice model in a dose-dependent manner with no statistically significant change in the animal's body weight and with no differences in the histology and appearance of the internal organs compared with the control group. Our study demonstrates that suppression of l-type amino acid transporter-1 activity using JPH203 might be used as a new therapeutic strategy for cholangiocarcinoma treatment.


Asunto(s)
Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Benzoxazoles/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colangiocarcinoma/patología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Transportador de Aminoácidos Neutros Grandes 1/genética , Leucina/metabolismo , Ratones , Terapia Molecular Dirigida , Tirosina/administración & dosificación , Tirosina/análogos & derivados , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Pharmacol Sci ; 132(3): 201-204, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27567475

RESUMEN

L-type amino acid transporter 1 (LAT1, SLC7A5) incorporates essential amino acids into cells. Recent studies have shown that LAT1 is a predominant transporter in various human cancers. However, the function of LAT1 in thymic carcinoma remains unknown. Here we demonstrate that LAT1 is a critical transporter for human thymic carcinoma cells. LAT1 was strongly expressed in human thymic carcinoma tissues. LAT1-specific inhibitor significantly suppressed leucine uptake and growth of Ty82 human thymic carcinoma cell lines, suggesting that thymic carcinoma takes advantage of LAT1 as a quality transporter and that LAT1-specific inhibitor might be clinically beneficial in therapy for thymic carcinoma.


Asunto(s)
Aminoácidos/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Timoma/metabolismo , Neoplasias del Timo/metabolismo , Benzoxazoles/farmacología , Línea Celular Tumoral , Humanos , Transportador de Aminoácidos Neutros Grandes 1/biosíntesis , Leucina/metabolismo , Timoma/tratamiento farmacológico , Neoplasias del Timo/tratamiento farmacológico , Tirosina/análogos & derivados , Tirosina/farmacología
6.
Xenobiotica ; 46(7): 641-650, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26576923

RESUMEN

1. Green tea extract (GTE) and EGCG have previously shown to increase the uptake of MPP+ into Caco-2 cells. However, whether GTE and its derivatives interact with renal basolateral organic cation transporter 2 (Oct2) which plays a crucial role for cationic clearance remains unknown. Thus, this study assessed the potential of drug-green tea (GT) catechins and its derivatives interactions with rat Oct2 using renal cortical slices and S2 stably expressing rat Oct2 (S2rOct2). 2. Both GTE and ECG inhibited MPP+ uptake in renal slices in a concentration-dependent manner (IC50 = 2.71 ± 0.360 mg/ml and 0.87 ± 0.151 mM), and this inhibitory effect was reversible. Inhibition of [3H]MPP+ transport in S2rOct2 by either GTE or ECG (IC50 = 1.90 ± 0.087 mg/ml and 1.67 ± 0.088 mM) was also observed. 3. The weak and reversible interactions of GTE and ECG with rOct2 indicate that consumption of GT beverages could not interfere with cationic drugs secreted via renal OCT2 in humans. However, the rise of therapeutic use of GTE and ECG might have to take into account the significant possibility of adverse drug-green tea catechins interactions which could alter renal organic cation drug clearance.

8.
J Immunol ; 191(8): 4080-5, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24038088

RESUMEN

Activation of T cells accompanies remarkable enhancement of metabolism. Sufficient and continuous nutrient supply is therefore important to support immune reaction in T cells. However, the mechanism of the promotion of nutrient incorporation in activated T cells has not been elucidated. In this study, we show that L-type amino acid transporter 1 (LAT1) is a major transporter for essential amino acids into activated human T cells. CD3/CD28 stimulation in primary human T cells triggered dramatic induction of LAT1 expression mediated by NF-κB and AP-1. Functional disturbance of LAT1 by a specific inhibitor and by small interfering RNA in human T cells suppressed essential amino acid uptake and induced a stress response mediated by DNA damage-inducible transcript 3 to attenuate cytokine production via inhibition of NF-κB and NFAT activities. These results uncover the previously unknown mechanism by which T cells accelerate essential amino acid uptake upon activation and adapt to essential amino acid starvation. Our results also raise the possibility for application of an LAT1 inhibitor as a new drug for therapy of disease caused by exaggerated immune response.


Asunto(s)
Aminoácidos Esenciales/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Activación de Linfocitos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Sistemas de Transporte de Aminoácidos , Transporte Biológico , Antígenos CD28/metabolismo , Complejo CD3/metabolismo , Células Cultivadas , Humanos , Transportador de Aminoácidos Neutros Grandes 1/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción CHOP/metabolismo
9.
J Pharmacol Sci ; 124(4): 511-3, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24671056

RESUMEN

Endothelial cell proliferation supporting angiogenesis requires sufficient nutrient supply because of facilitated intracellular metabolism. However, little is known about the mechanism for the promotion of nutrient incorporation in proliferating endothelial cells. Here we show that L-type amino acid transporter 1 (LAT1) is a major transporter of essential amino acids in human umbilical vein endothelial cells (HUVECs). Growing HUVECs express a certain level of LAT1. A LAT1-specific inhibitor suppressed leucine uptake, cell proliferation, and tube formation of HUVECs. Therefore, LAT1 acts to support effective uptake of amino acids, which is critical for the optimal function of HUVECs for angiogenesis.


Asunto(s)
Aminoácidos Esenciales/metabolismo , Células Endoteliales/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/fisiología , Venas Umbilicales/citología , Venas Umbilicales/metabolismo , Proliferación Celular , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/fisiología , Humanos , Neovascularización Fisiológica
10.
Toxicol Pathol ; 41(3): 480-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22968287

RESUMEN

The present study describes atypical Leydig cell (LC) hyperplasia in 20-week-old Sprague-Dawley rats with low testosterone and high luteinizing hormone levels after prenatal administration of 100 mg/kg/day di(n-butyl) phthalate on days 12 to 21 postconception. Light microscopy revealed LC hyperplasia surrounded by severely degenerated seminiferous tubules. Aggregated LCs had large ovoid nuclei with nucleoli and abundant eosinophilic cytoplasm. Immunohistochemical analysis showed expression of proliferating cell nuclear antigen and vimentin in many hyperplastic LCs. Electron microscopy revealed atypical nuclei, abundant free ribosomes, stripped rough endoplasmic reticulum, intermediate-size filaments, elongated cytoplasmic filopodia, atypical tight junctions, and cilia formations, but smooth endoplasmic reticulum was scarcely observed.


Asunto(s)
Dibutil Ftalato/toxicidad , Células Intersticiales del Testículo/efectos de los fármacos , Células Intersticiales del Testículo/patología , Hormona Luteinizante/sangre , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/patología , Testosterona/metabolismo , Animales , Femenino , Histocitoquímica , Hiperplasia/inducido químicamente , Hiperplasia/patología , Masculino , Embarazo , Ratas , Ratas Sprague-Dawley , Testículo/química , Testículo/efectos de los fármacos , Testículo/patología
11.
Nat Genet ; 36(9): 999-1002, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15286787

RESUMEN

Hartnup disorder, an autosomal recessive defect named after an English family described in 1956 (ref. 1), results from impaired transport of neutral amino acids across epithelial cells in renal proximal tubules and intestinal mucosa. Symptoms include transient manifestations of pellagra (rashes), cerebellar ataxia and psychosis. Using homozygosity mapping in the original family in whom Hartnup disorder was discovered, we confirmed that the critical region for one causative gene was located on chromosome 5p15 (ref. 3). This region is homologous to the area of mouse chromosome 13 that encodes the sodium-dependent amino acid transporter B(0)AT1 (ref. 4). We isolated the human homolog of B(0)AT1, called SLC6A19, and determined its size and molecular organization. We then identified mutations in SLC6A19 in members of the original family in whom Hartnup disorder was discovered and of three Japanese families. The protein product of SLC6A19, the Hartnup transporter, is expressed primarily in intestine and renal proximal tubule and functions as a neutral amino acid transporter.


Asunto(s)
Enfermedad de Hartnup/genética , Mutación , Secuencia de Aminoácidos , Sistemas de Transporte de Aminoácidos Neutros/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Linaje
12.
Fundam Clin Pharmacol ; 37(4): 833-842, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36843181

RESUMEN

Buspirone, a cationic drug, is an anxiolytic and antidepressant drug. However, whether buspirone and its metabolites are interacted with organic cationic transporter remains uncertain. In this study, we examined the interaction of buspirone and its major metabolites 1-(2-pyrimidinyl)piperazine (1-PP) and 6-hydroxybuspirone (6'-OH-Bu) with hOCTs using human hepatocellular carcinoma (HepG2), human colorectal adenocarcinoma (Caco-2) cells, and S2 cells expressing OCT1 (S2hOCT1), 2 (S2hOCT2), or 3 (S2hOCT3). Coadministration of buspirone and fluorescent 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+ ) was examined using HepG2 cells, and [3 H]-1-methyl-4-phenylpyridinium (MPP+ ) transport was assessed in S2 cell overexpressing hOCTs. The results showed that ASP+ transport was suppressed by buspirone with an IC50 of 26.3 ± 2.9 µM without any cytotoxic effects in HepG2 expressing hOCTs cells. Consistently, buspirone strongly inhibited [3 H]-MPP+ uptake by S2hOCT1, S2hOCT2, and S2hOCT3 cells with an IC50s of 89.0 ± 1.3 µM, 43.7 ± 7.5 µM, and 20.4 ± 1.0 µM, respectively. Nonetheless, 6'-OH-Bu and 1-PP caused weak or no inhibition on ASP+ and [3 H]-MPP+ transport. These findings suggest the potential interaction of buspirone with organic cation drugs that are handled by hOCT3. However, further clinical relevance is needed to support these findings for preventing drug-drug interaction in patients who take prescribed drugs together with buspirone.


Asunto(s)
Buspirona , Proteínas de Transporte de Catión Orgánico , Humanos , Buspirona/farmacología , Células CACO-2 , Transportador 2 de Cátion Orgánico , Transportador 1 de Catión Orgánico/metabolismo , Cationes/metabolismo
13.
Biomed Pharmacother ; 165: 115098, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37437378

RESUMEN

As a final common pathway of renal injuries, renal fibrosis leads to chronic kidney disease (CKD). Currently, there is no safe and effective therapy to prevent the progression of renal fibrosis to CKD. Inhibition of transforming growth factor-ß1 (TGF-ß1) pathway is proposed as one of the most promising approaches for anti-renal fibrosis therapies. This study aimed to identify novel anti-fibrotic agents using the TGF-ß1-induced fibrosis in renal proximal tubule epithelial cells (RPTEC) and characterize their mechanism of action as well as in vivo efficacy. By screening 362 natural product-based compounds for their ability to reduce collagen accumulation assessed by picro-sirius red (PSR) staining in RPTEC cells, a chalcone derivative AD-021 was identified as an anti-fibrotic agent with IC50 of 14.93 µM. AD-021 suppressed TGF-ß1-induced collagen production, expression of pro-fibrotic proteins (fibronectin and α-smooth muscle actin (αSMA)), and Smad-dependent and Smad-independent signaling pathways via suppression of TGF-ß receptor II (TGFßRII) phosphorylation in RPTEC cells. Furthermore, TGF-ß1-induced mitochondrial fission in RPTEC cells was ameliorated by AD-021 via mechanisms involving inhibition of Drp1 phosphorylation. In a mouse model of unilateral ureteral obstruction (UUO)-induced renal fibrosis, AD-021 reduced plasma TGF-ß1, ameliorated renal fibrosis and improved renal function. Collectively, AD-021 represents a novel class of natural product-based anti-fibrotic agent that has therapeutic potential in the prevention of fibrosis-associated renal disorders including CKD.


Asunto(s)
Chalcona , Chalconas , Enfermedades Renales , Insuficiencia Renal Crónica , Obstrucción Ureteral , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Antifibróticos , Chalconas/farmacología , Chalconas/uso terapéutico , Chalconas/metabolismo , Chalcona/farmacología , Chalcona/uso terapéutico , Enfermedades Renales/metabolismo , Riñón , Insuficiencia Renal Crónica/tratamiento farmacológico , Insuficiencia Renal Crónica/metabolismo , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/tratamiento farmacológico , Fibrosis
14.
Clin Exp Nephrol ; 16(1): 89-95, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22038265

RESUMEN

Humans have higher serum uric acid levels than other mammalian species owing to the genetic silencing of the hepatic enzyme uricase that metabolizes uric acid into allantoin. Urate (the ionized form of uric acid) is generated from purine metabolism and it may provide antioxidant defense in the human body. Despite its potential advantage, sustained hyperuricemia has pathogenetic causes in gout and renal diseases, and putative roles in hypertension and cardiovascular diseases. Since the kidney plays a dominant role in maintaining plasma urate levels through the excretion process, it is important to understand the molecular mechanism of renal urate handling. Although the molecular identification of a kidney-specific urate/anion exchanger URAT1 in 2002 paved the way for successive identification of several urate transport-related proteins, the entire picture of effective renal urate handling in humans has not yet been clarified. Recently, several genome-wide association studies identified a substantial association between uric acid concentration and single nucleotide polymorphisms in at least ten genetic loci including eight transporter-coding genes. In 2008, we functionally characterized the facilitatory glucose transporter family member SLC2A9 (GLUT9), one of the candidate genes for urate handling, as a voltage-driven urate transporter URATv1 at the basolateral side of renal proximal tubules that comprises the main route of the urate reabsorption pathway, in tandem with URAT1 at the apical side. In this review, recent findings concerning these candidate molecules are presented.


Asunto(s)
Hiperuricemia/metabolismo , Túbulos Renales Proximales/metabolismo , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Ácido Úrico/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Estudio de Asociación del Genoma Completo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Humanos , Proteínas de la Membrana/fisiología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo I/genética , Ácido Úrico/sangre
15.
Biomed Pharmacother ; 148: 112732, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35217281

RESUMEN

Colistin is a last-resort polypeptide antibiotic widely used to treat against multidrug-resistant Gram-negative bacterial infections. However, this treatment is associated with nephrotoxicity. The aim of this study was to examine the potential protective effect of panduratin A, a bioactive compound of Boesenbergia rotunda, on colistin-induced nephrotoxicity in both in vivo and in vitro models. Intraperitoneal injection of 15 mg/kg colistin for 7 days markedly promoted renal tubular degeneration, increased blood urea nitrogen (BUN) levels, and upregulated the expression of renal injury biomarker and apoptosis proteins. In addition, treatment with colistin increased oxidative stress and apoptosis in mice kidney tissues. Interestingly, these defects were attenuated when co-administered of colistin with panduratin A (2.5 or 25 mg/kg). The underlying mechanisms of panduratin A attenuating colistin toxicity was investigated in human renal proximal tubular cells (RPTEC/TERT1). The mechanisms by which colistin-triggered cytotoxicity was determined by analysis of cell death, reactive oxygen species (ROS) levels, mitochondria function as well as the expression of proteins related to apoptosis pathway. Colistin treatment (200 µg/ml) significantly increased cell apoptosis, elevated ROS production, reduced mitochondrial membrane potential, and decreased anti-apoptotic protein (Bcl-2) expression. These effects were notably suppressed by co-treatment with panduratin A (5 µM). Collectively, panduratin A exerts as a novel nephroprotective agent to protect against colistin-induced renal injury by attenuating mitochondrial damage and renal cell apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Chalconas/farmacología , Colistina/efectos adversos , Enfermedades Renales/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Sustancias Protectoras/farmacología , Animales , Antibacterianos/efectos adversos , Línea Celular , Colistina/farmacología , Células Epiteliales/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Riñón/lesiones , Enfermedades Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Zingiberaceae/química
16.
J Biol Chem ; 285(29): 22141-51, 2010 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-20448048

RESUMEN

We identified a novel prostaglandin (PG)-specific organic anion transporter (OAT) in the OAT group of the SLC22 family. The transporter designated OAT-PG from mouse kidney exhibited Na(+)-independent and saturable transport of PGE(2) when expressed in a proximal tubule cell line (S(2)). Unusual for OAT members, OAT-PG showed narrow substrate selectivity and high affinity for a specific subset of PGs, including PGE(2), PGF(2alpha), and PGD(2). Similar to PGE(2) receptor and PGT, a structurally distinct PG transporter, OAT-PG requires for its substrates an alpha-carboxyl group, with a double bond between C13 and C14 as well as a (S)-hydroxyl group at C15. Unlike the PGE(2) receptor, however, the hydroxyl group at C11 in a cyclopentane ring is not essential for OAT-PG substrates. Addition of a hydroxyl group at C19 or C20 impairs the interaction with OAT-PG, whereas an ethyl group at C20 enhances the interaction, suggesting the importance of hydrophobicity around the omega-tail tip forming a "hydrophobic core" accompanied by a negative charge, which is essential for substrates of OAT members. OAT-PG-mediated transport is concentrative in nature, although OAT-PG mediates both facilitative and exchange transport. OAT-PG is kidney-specific and localized on the basolateral membrane of proximal tubules where a PG-inactivating enzyme, 15-hydroxyprostaglandin dehydrogenase, is expressed. Because of the fact that 15-keto-PGE(2), the metabolite of PGE(2) produced by 15-hydroxyprostaglandin dehydrogenase, is not a substrate of OAT-PG, the transport-metabolism coupling would make unidirectional PGE(2) transport more efficient. By removing extracellular PGE(2), OAT-PG is proposed to be involved in the local PGE(2) clearance and metabolism for the inactivation of PG signals in the kidney cortex.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas/metabolismo , Túbulos Renales Proximales/enzimología , Transportadores de Anión Orgánico/metabolismo , Prostaglandinas/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Dinoprost/análogos & derivados , Dinoprost/farmacología , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Dinoprostona/farmacología , Perfilación de la Expresión Génica , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Cinética , Ratones , Datos de Secuencia Molecular , Transportadores de Anión Orgánico/química , Transportadores de Anión Orgánico/genética , Prostaglandinas/farmacología , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos
17.
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
18.
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
19.
J Pharmacol Sci ; 115(2): 249-253, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-32272545

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 [14C]para-aminohippurate, [3H]bumetanide, [3H]estrone sulfate, and [14C]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.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA