Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Fundam Clin Pharmacol ; 37(4): 833-842, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36843181

RESUMO

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.


Assuntos
Buspirona , Proteínas de Transporte de Cátions Orgânicos , Humanos , Buspirona/farmacologia , Células CACO-2 , Transportador 2 de Cátion Orgânico , Transportador 1 de Cátions Orgânicos/metabolismo , Cátions/metabolismo
2.
Biomed Pharmacother ; 148: 112732, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35217281

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Chalconas/farmacologia , Colistina/efeitos adversos , Nefropatias/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Animais , Antibacterianos/efeitos adversos , Linhagem Celular , Colistina/farmacologia , Células Epiteliais/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Rim/lesões , Nefropatias/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Zingiberaceae/química
3.
Genetics ; 214(4): 1079-1090, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32005656

RESUMO

Gout is a common arthritis caused by monosodium urate crystals. The heritability of serum urate levels is estimated to be 30-70%; however, common genetic variants account for only 7.9% of the variance in serum urate levels. This discrepancy is an example of "missing heritability." The "missing heritability" suggests that variants associated with uric acid levels are yet to be found. By using genomic sequences of the ToMMo cohort, we identified rare variants of the SLC22A12 gene that affect the urate transport activity of URAT1. URAT1 is a transporter protein encoded by the SLC22A12 gene. We grouped the participants with variants affecting urate uptake by URAT1 and analyzed the variance of serum urate levels. The results showed that the heritability explained by the SLC22A12 variants of men and women exceeds 10%, suggesting that rare variants underlie a substantial portion of the "missing heritability" of serum urate levels.


Assuntos
Gota/genética , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Polimorfismo de Nucleotídeo Único , Ácido Úrico/sangue , Animais , Feminino , Interação Gene-Ambiente , Humanos , Masculino , Pessoa de Meia-Idade , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Ácido Úrico/metabolismo , Xenopus
4.
Bioorg Med Chem Lett ; 28(10): 1915-1918, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29655983

RESUMO

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.


Assuntos
Compostos de Boro/química , Fenilalanina/análogos & derivados , Alquilação , Compostos de Boro/síntese química , Catálise , Complexos de Coordenação/química , Paládio/química , Fenilalanina/síntese química , Fenilalanina/química , Estereoisomerismo
5.
J Pharmacol Sci ; 136(4): 196-202, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29656005

RESUMO

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.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Di-Hidropiridinas/farmacologia , Modelos Animais , Uricosúricos , Animais , Creatinina/sangue , Creatinina/urina , Proteínas de Ligação a DNA , Masculino , Camundongos Endogâmicos ICR , Transportadores de Ânions Orgânicos , Ácido Úrico/sangue , Ácido Úrico/urina
6.
Tumour Biol ; 39(3): 1010428317694545, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28347255

RESUMO

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.


Assuntos
Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/genética , Transportador 1 de Aminoácidos Neutros Grandes/biossíntese , Animais , Apoptose/efeitos dos fármacos , Benzoxazóis/administração & dosagem , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Colangiocarcinoma/patologia , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/genética , Leucina/metabolismo , Camundongos , Terapia de Alvo Molecular , Tirosina/administração & dosagem , Tirosina/análogos & derivados , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Parasitol Int ; 66(4): 471-478, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26657242

RESUMO

L-type amino acid transporter 1 (LAT1) is highly expressed in various human cancers, including cholangiocarcinoma (CCA), the most common cancer in Northeast Thailand. Chronic inflammation and oxidative stress induced by liver fluke, Opisthorchis viverrini, infection has been recognized as the major cause of CCA in this area. We show here that an increased expression of LAT1 and its co-functional protein CD98 are found during carcinogenesis induced by Ov in hamster CCA tissues. We also demonstrate that oxidative stress induced by H2O2 is time-dependent and dramatically activates LAT1 and CD98 expression in immortal cholangiocytes (MMNK1). In addition, H2O2 treatment increased LAT1 and CD98 expression, as well as an activated form of AKT and mTOR in MMNK1 and CCA cell lines (KKU-M055 and KKU-M213). We also show that suppression of PI3K/AKT pathway activity with a dual PI3K/mTOR inhibitor, BEZ235, causes a reduction in LAT1 and CD98 expression in KKU-M055 and KKU-M213 in parallel with a reduction of activated AKT and mTOR. Interestingly, high expression of LAT1 in human CCA tissues is a significant prognostic factor for shorter survival. Taken together, our data show that LAT1 expression is significantly associated with CCA progression and cholangiocarcinogenesis induced by oxidative stress. Moreover, the expression of LAT1 and CD98 in CCA is possibly regulated by the PI3K/AKT signaling pathway.


Assuntos
Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Proteína-1 Reguladora de Fusão/genética , Transportador 1 de Aminoácidos Neutros Grandes/genética , Opistorquíase/metabolismo , Opisthorchis/fisiologia , Adulto , Idoso , Animais , Neoplasias dos Ductos Biliares/diagnóstico , Linhagem Celular , Colangiocarcinoma/diagnóstico , Cricetinae , Feminino , Proteína-1 Reguladora de Fusão/metabolismo , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Masculino , Mesocricetus , Pessoa de Meia-Idade , Opistorquíase/complicações , Opistorquíase/diagnóstico , Estresse Oxidativo , Prognóstico , Análise Serial de Tecidos , Regulação para Cima
8.
Anat Sci Int ; 92(2): 200-206, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27614971

RESUMO

Organic anions (OAs) are secreted in renal proximal tubules in two steps. In the first step, OAs are transported from the blood through basolateral membranes into proximal tubular cells. The prototypical substrate for renal organic anion transport systems, para-aminohippurate (PAH), is transported across basolateral membranes of proximal tubular cells via OAT1 (SLC22A6) and OAT3 (SLC22A8) against an electrochemical gradient in exchange for intracellular dicarboxylates. In the second step, OAs exit into urine through apical membranes of proximal tubules. This step is thought to be performed by multidrug efflux transporters and a voltage-driven organic anion transporter. However, the molecular nature and precise functional properties of these efflux systems are largely unknown. Recently, we characterized an orphan transporter known as human type I sodium-phosphate transporter 4, hNPT4 (SLC17A3), using the Xenopus oocyte expression system. hNPT4 acts as a voltage-driven efflux transporter ("human OATv1") for several OAs such as PAH, estrone sulfate, diuretic drugs, and urate. Here, we describe a model for an OA secretory pathway in renal tubular cells in which OAs exit cells and enter the tubular lumen via hOATv1 (hNPT4). Additionally, hOATv1 functions as a common renal secretory pathway for both urate and drugs, indicating that hOATv1 may be a leak pathway for excess urate that is reabsorbed via apical URAT1 to control the intracellular urate levels. Therefore, we propose a molecular mechanism for the induction of hyperuricemia by diuretics: the diuretics enter proximal tubular cells via basolateral OAT1 and/or OAT3 and may then interfere with the NPT4-mediated apical urate efflux in the renal proximal tubule.


Assuntos
Túbulos Renais Proximais/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Transporte Biológico , Humanos , Rim/metabolismo
9.
J Pharmacol Sci ; 132(3): 201-204, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27567475

RESUMO

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.


Assuntos
Aminoácidos/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Timoma/metabolismo , Neoplasias do Timo/metabolismo , Benzoxazóis/farmacologia , Linhagem Celular Tumoral , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/biossíntese , Leucina/metabolismo , Timoma/tratamento farmacológico , Neoplasias do Timo/tratamento farmacológico , Tirosina/análogos & derivados , Tirosina/farmacologia
10.
Drug Des Devel Ther ; 6: 323-39, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23152669

RESUMO

Human uric acid transporter 1 (hURAT1; SLC22A12) is a very important urate anion exchanger. Elevated urate levels are known to play a pivotal role in cardiovascular diseases, chronic renal disease, diabetes, and hypertension. Therefore, the development of potent uric acid transport inhibitors may lead to novel therapeutic agents to combat these human diseases. The current study investigates small molecular weight compounds and their ability to inhibit 14C-urate uptake in oocytes expressing hURAT1. Using the most promising drug candidates generated from our structure-activity relationship findings, we subsequently conducted in vitro hepatic metabolism and pharmacokinetic (PK) studies in male Sprague-Dawley rats. Compounds were incubated with rat liver microsomes containing cofactors nicotinamide adenine dinucleotide phosphate and uridine 5'-diphosphoglucuronic acid. In vitro metabolism and PK samples were analyzed using liquid chromatography/mass spectrometry-mass spectrometry methods. Independently, six different inhibitors were orally (capsule dosing) or intravenously (orbital sinus) administered to fasting male Sprague-Dawley rats. Blood samples were collected and analyzed; these data were used to compare in vitro and in vivo metabolism and to compute noncompartmental model PK values. Mono-oxidation (Phase I) and glucuronidation (Phase II) pathways were observed in vitro and in vivo. The in vitro data were used to compute hepatic intrinsic clearance, and the in vivo data were used to compute peak blood concentration, time after administration to achieve peak blood concentration, area under the curve, and orally absorbed fraction. The experimental data provide additional insight into the hURAT1 inhibitor structure-activity relationship and in vitro-in vivo correlation. Furthermore, the results illustrate that one may successfully prepare potent inhibitors that exhibit moderate to good oral bioavailability.


Assuntos
Benzobromarona/análogos & derivados , Benzobromarona/metabolismo , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Animais , Área Sob a Curva , Disponibilidade Biológica , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
11.
Oncol Rep ; 28(3): 862-6, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22736142

RESUMO

Tumor cells take up a massive amount of nutrition compared to normal cells for increased metabolism. Therefore, special transporters for organic materials are required to satisfy the powerful consumption of nutrition in tumor cells. L-type amino acid transporter 1 (LAT1) incorporates large neutral amino acids, most of which are also categorized as essential amino acids, into cells in a Na+-independent manner. Because of its high expression levels in a variety of cancer cells, it is speculated that LAT1 functions as a key transporter for highly effective delivery of essential amino acids into cancer cells. In this regard, LAT1 inhibitor is expected to have clinical benefit for cancer therapy. However, the molecular mechanism of enrichment of LAT1 in cancer cells remains poorly understood. Here, we show that a proto-oncogene, c-Myc, is a critical positive regulator of LAT1 expression in MIA Paca-2 human pancreatic cancer cells. The uptake of leucine, a representative neutral amino acid, was strictly dependent on LAT1 in MIA Paca-2 cells, and siRNA-mediated knockdown of LAT1 inhibited cell proliferation. Diminished c-Myc expression with siRNA resulted in severe reduction of LAT1 protein levels as well as mRNA levels, which, in turn, led to a significant defect of leucine incorporation. The LAT1 promoter has a canonical c-Myc binding sequence and overexpression of c-Myc increased LAT1 promoter activity, whereas mutation of c-Myc binding site diminished this effect. Our results suggest biological significance of LAT1 in tumor growth and molecular machinery that could explain why LAT1 is preferentially expressed in cancer cells.


Assuntos
Regulação Neoplásica da Expressão Gênica , Expressão Gênica , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular Tumoral , Proliferação de Células , Sequência Consenso , Regulação para Baixo , Técnicas de Silenciamento de Genes , Genes Reporter , Humanos , Transportador 1 de Aminoácidos Neutros Grandes/genética , Leucina/metabolismo , Luciferases/biossíntese , Luciferases/genética , Neoplasias Pancreáticas , Regiões Promotoras Genéticas , Proto-Oncogene Mas , RNA Interferente Pequeno
12.
Drug Metab Pharmacokinet ; 27(1): 155-61, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21914964

RESUMO

Many primary human tumors and tumor cell lines highly express human L-type amino acid transporter 1 (hLAT1); cancerous cells in vivo are strongly linked to LAT1 expression. Synthetic chemistry and in vitro screening efforts have afforded a variety of novel and highly hLAT1 selective compounds, such as JPH203 1. In a recent report, we demonstrated that 1 has potent in vitro and in vivo activity. JPH203 was intravenously administered to produce significant growth inhibition against HT-29 tumors transplanted in nude mice. The current work develops a robust LC/MS-MS method to monitor 1 and its major Phase II metabolite N-acetyl-JPH203 2 from biological samples. We have conducted in vitro and in vivo experiments and the major scientific findings are: i) the major route of biotransformation of 1 is Phase II metabolism to produce 2; ii) metabolite 2 is formed in various organs/tissues (i.e. blood, liver, kidney); and iii) as dogs, which are deficient in NAT genes, do not produce 2, the dog will not be an appropriate toxicological model to evaluate 1.


Assuntos
Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Benzoxazóis/farmacocinética , Transportador 1 de Aminoácidos Neutros Grandes/química , Moduladores de Transporte de Membrana/metabolismo , Moduladores de Transporte de Membrana/farmacocinética , Microssomos/metabolismo , Tirosina/análogos & derivados , Acetilação , Animais , Antineoplásicos/análise , Antineoplásicos/sangue , Benzoxazóis/análise , Benzoxazóis/sangue , Benzoxazóis/metabolismo , Biotransformação , Cães , Humanos , Intestino Delgado/metabolismo , Rim/química , Rim/metabolismo , Fígado/química , Fígado/metabolismo , Macaca fascicularis , Masculino , Moduladores de Transporte de Membrana/análise , Moduladores de Transporte de Membrana/sangue , Camundongos , Microssomos Hepáticos/metabolismo , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual , Tirosina/análise , Tirosina/sangue , Tirosina/metabolismo , Tirosina/farmacocinética
13.
Clin Exp Nephrol ; 16(1): 89-95, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22038265

RESUMO

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.


Assuntos
Hiperuricemia/metabolismo , Túbulos Renais Proximais/metabolismo , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Ácido Úrico/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Estudo de Associação Genômica Ampla , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Proteínas de Membrana/fisiologia , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas de Neoplasias/genética , Polimorfismo de Nucleotídeo Único , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/genética , Ácido Úrico/sangue
14.
PLoS One ; 6(12): e28641, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22194875

RESUMO

Idiopathic renal hypouricaemia is an inherited form of hypouricaemia, associated with abnormal renal handling of uric acid. There is excessive urinary wasting of uric acid resulting in hypouricaemia. Patients may be asymptomatic, but the persistent urinary abnormalities may manifest as renal stone disease, and hypouricaemia may manifest as exercise induced acute kidney injury. Here we have identified Macedonian and British patients with hypouricaemia, who presented with a variety of renal symptoms and signs including renal stone disease, hematuria, pyelonephritis and nephrocalcinosis. We have identified heterozygous missense mutations in SLC22A12 encoding the urate transporter protein URAT1 and correlate these genetic findings with functional characterization. Urate handling was determined using uptake experiments in HEK293 cells. This data highlights the importance of the URAT1 renal urate transporter in determining serum urate concentrations and the clinical phenotypes, including nephrolithiasis, that should prompt the clinician to suspect an inherited form of renal hypouricaemia.


Assuntos
Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Adolescente , Adulto , Sequência de Aminoácidos , Sequência de Bases , Criança , Biologia Computacional , Sequência Conservada/genética , Demografia , Feminino , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Mutação/genética , Transportadores de Ânions Orgânicos/química , Proteínas de Transporte de Cátions Orgânicos/química , Erros Inatos do Transporte Tubular Renal/genética , Cálculos Urinários/genética
15.
J Med Chem ; 54(8): 2701-13, 2011 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-21449597

RESUMO

The kidneys are a vital organ in the human body. They serve several purposes including homeostatic functions such as regulating extracellular fluid volume and maintaining acid-base and electrolyte balance and are essential regarding the excretion of metabolic waste. Furthermore, the kidneys play an important role in uric acid secretion/reabsorption. Abnormalities associated with kidney transporters have been associated with various diseases, such as gout. The current study utilized Xenopus oocytes expressing human uric acid transporter 1 (hURAT1; SLC22A12) as an in vitro method to investigate novel compounds and their ability to inhibit (14)C-uric acid uptake via hURAT1. We have prepared and tested a series of 2-ethyl-benzofuran compounds and probed the hURAT1 in vitro inhibitor structure-activity relationship. As compared to dimethoxy analogues, monophenols formed on the C ring showed the best in vitro inhibitory potential. Compounds with submicromolar (i.e., IC(50) < 1000 nM) inhibitors were prepared by brominating the corresponding phenols to produce compounds with potent uricosuric activity.


Assuntos
Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Animais , Desenho de Fármacos , Humanos , Rim/metabolismo , Espectroscopia de Ressonância Magnética , Xenopus
16.
J Physiol Sci ; 61(3): 253-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21350910

RESUMO

Orotate is a precursor of pyrimidine synthesis. The kidney uses exogenous orotate for the synthesis of uridine diphosphosugars, which are used in the glycosylation of collagen in glomerular and tubular basement membranes. Orotate uptake occurs in the liver and kidney, but its molecular mechanism is largely unknown. Since orotate has been shown to be a substrate of the renal urate/anion exchanger in brush border membrane vesicle studies, we investigated whether human URAT1 (hURAT1) mediates the transport of orotate using HEK293 cells expressing hURAT1 (HEK-hURAT1). hURAT1 mediated a time- and dose-dependent uptake of orotate (K (m) 5.2 µM). hURAT1-mediated [(3)H]orotate transport was inhibited strongly by non-labeled (cold) orotate and the uricouric agent benzbromarone, and moderately inhibited by urate, nicotinate, and another uricouric agent, probenecid. This is the first report demonstrating that hURAT1 mediates the transport of orotate. hURAT1 may function as one of the entrance pathways in renal proximal tubular cells.


Assuntos
Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Ácido Orótico/metabolismo , Animais , Benzobromarona/farmacologia , Transporte Biológico/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Oócitos , Ácido Orótico/antagonistas & inibidores , Ácido Úrico/metabolismo , Uricosúricos/farmacologia , Xenopus
17.
Comp Med ; 61(5): 412-8, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22330348

RESUMO

Epidemiologic studies indicate that the incidence of gastric cancer is higher in males than in females. Although the mechanisms mediating this difference are unclear, a role for estrogens has been proposed. We used Western blotting to evaluate the role of estrogen receptor (ER) subtypes ERα and ERß and proliferating cell nuclear antigen (PCNA) in N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced gastric carcinogenesis in Wistar rats; ERα and ERß mRNA levels also were analyzed by quantitative real-time RT-PCR analysis. The incidence of gastric cancer was significantly higher in male than female rats. In both sexes, ERα expression was similar in MNNG-treated cancerous and noncancerous tissues and normal gastric tissue. However, ERß expression in MNNG-treated cancerous and noncancerous tissues was significantly lower in male rats and higher in female rats than that in normal gastric tissue; MNNG-induced cancerous tissue showed the highest ERß expression. PCNA expression in MNNG-treated cancerous tissues was higher than that in noncancerous tissues, and was higher in male rats than female rats. Western blotting results were consistent with the mRNA changes determined by quantitative real-time RT-PCR. The present study provides evidence of a sex-associated difference in ERß and PCNA expression in MNNG-induced gastric cancers in Wistar rats.


Assuntos
Receptor beta de Estrogênio/metabolismo , Metilnitronitrosoguanidina/toxicidade , Neoplasias Gástricas/induzido quimicamente , Neoplasias Gástricas/metabolismo , Animais , Western Blotting , Feminino , Masculino , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais
18.
J Biol Chem ; 285(45): 35123-32, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20810651

RESUMO

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.


Assuntos
Bumetanida/farmacocinética , Diuréticos/farmacocinética , Furosemida/farmacocinética , Túbulos Renais Proximais/metabolismo , Modelos Biológicos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/metabolismo , Ácido Úrico/metabolismo , Animais , Bumetanida/efeitos adversos , Bumetanida/farmacologia , Diuréticos/efeitos adversos , Diuréticos/farmacologia , Furosemida/efeitos adversos , Furosemida/farmacologia , Predisposição Genética para Doença , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Gota/genética , Gota/metabolismo , Humanos , Hiperuricemia/induzido quimicamente , Hiperuricemia/genética , Hiperuricemia/metabolismo , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/genética , Fígado/metabolismo , Mutação de Sentido Incorreto , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/genética , Urato Oxidase/genética , Urato Oxidase/metabolismo , Xenopus laevis
19.
J Pharmacol Sci ; 114(1): 115-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20736511

RESUMO

Hyperuricemia is currently recognized as a risk factor for cardiovascular diseases. It has been reported that the angiotensin II-receptor blocker (ARB) losartan decreases serum uric acid level. In this study, the effects of another ARB, irbesartan, on [(14)C]uric acid-transport activity of renal uric acid reabsorptive transporters URAT1 and URATv1 were examined with Xenopus oocytes expressing each transporter. The results showed that irbesartan (100-500 µM) inhibited the uptake of uric acid via both transporters. The inhibitory effects of irbesartan exceeded those of losartan and other ARBs, and the results suggest that irbesartan can reduce serum uric acid level.


Assuntos
Compostos de Bifenilo/farmacologia , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Tetrazóis/farmacologia , Ácido Úrico/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Feminino , Irbesartana , Rim/efeitos dos fármacos , Rim/metabolismo , Ácido Úrico/metabolismo , Xenopus
20.
J Biol Chem ; 285(29): 22141-51, 2010 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-20448048

RESUMO

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.


Assuntos
Hidroxiprostaglandina Desidrogenases/metabolismo , Túbulos Renais Proximais/enzimologia , Transportadores de Ânions Orgânicos/metabolismo , Prostaglandinas/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Dinoprosta/análogos & derivados , Dinoprosta/farmacologia , Dinoprostona/análogos & derivados , Dinoprostona/metabolismo , Dinoprostona/farmacologia , Perfilação da Expressão Gênica , Humanos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Cinética , Camundongos , Dados de Sequência Molecular , Transportadores de Ânions Orgânicos/química , Transportadores de Ânions Orgânicos/genética , Prostaglandinas/farmacologia , Relação Estrutura-Atividade , Especificidade por Substrato/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA