Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Phytomedicine ; 62: 152975, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31181404

RESUMO

BACKGROUND: Alpinia oxyphylla is a well-known traditional medicine used in China and Korea to treat intestinal disorders, urosis, diuresis, and chronic glomerulonephritis. PURPOSE: We investigated the anti-hyperuricemic effects of Alpinia oxyphylla seed extract (AE), and the underlying mechanisms of action through in vitro and in vivo studies. METHODS: We evaluated levels of uric acid in the serum and urine, the expression of renal urate transport proteins, and levels of inflammatory cytokines in potassium oxonate (PO)-induced hyperuricemic rats. Xanthine oxidase activity was analyzed in vitro, while cellular uric acid uptake was assessed in oocytes expressing the human urate transporter 1 (hURAT1). Moreover, the main components of AE were analyzed using UPLC. RESULTS: In PO-induced hyperuricemic rats, 200 and 400 mg/kg of AE significantly decreased levels of uric acid in serum, while 400 mg/kg of AE increased uric acid levels in urine. AE did not inhibit xanthine oxidase in vitro; however, 1, 10, and 100 µg/ml of AE significantly decreased uric acid uptake into oocytes expressing hURAT1. Furthermore, 400 mg/kg of AE increased levels of organic anion transporter (OAT) 1 protein, while 200 and 400 mg/kg of AE decreased the protein content of urate transporter, URAT1 and inflammatory cytokines in the kidneys. Nootkatone was identified as one the main chemical components in AE from UPLC analysis. CONCLUSIONS: These findings suggest that AE exerts anti-hyperuricemic and uricosuric effects, which are related to the promotion of uric acid excretion via enhanced secretion and inhibition of uric acid reabsorption in the kidneys. Thus, AE may be a potential treatment for hyperuricemia and gout.


Assuntos
Alpinia/química , Hiperuricemia/tratamento farmacológico , Extratos Vegetais/administração & dosagem , Ácido Úrico/urina , Xantina Oxidase/metabolismo , Animais , China/epidemiologia , Gota , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Masculino , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos/metabolismo , Ácido Oxônico , Extratos Vegetais/química , Ratos , República da Coreia/epidemiologia , Xantina Oxidase/genética
2.
Nephrology (Carlton) ; 22(8): 642-648, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27282888

RESUMO

AIM: Furosemide is a loop diuretic. Different authors demonstrated that continuous administration of furosemide modulates the expression of organic anion transporters. This study was undertaken to simultaneously evaluate the effects of furosemide pretreatment on organic anion transporter 1 (Oat1) and multidrug resistance protein 2 (Mrp2) renal expressions, on p-aminohippurate (PAH) pharmacokinetics and on renal and urinary PAH levels in rats. METHODS: Male Wistar rats were treated with furosemide (6 mg/100 g body weight per day, subcutaneously, 4 days) (treated group) or saline (control group). On the fifth day, PAH was administered as a bolus infusion in the femoral vein, and plasma samples were obtained from femoral artery at different time points. PAH levels in renal tissue and urine were also assessed. Renal Oat1 and Mrp2 expressions were evaluated by western blotting. RESULTS: Furosemide pretreatment increased both the expression of Oat1 and Mrp2. PAH plasma concentrations decreased following a biexponential function. The furosemide-treated group showed higher PAH plasma levels, a lower systemic clearance and elimination rate constant from the peripheral compartment, indicating that PAH renal elimination was decreased. PAH levels in renal tissue were significantly elevated and in urine appeared to be significantly lower as compared with control animals. CONCLUSIONS: Furosemide pretreatment caused a significant decrease of PAH renal elimination, despite Oat1 and Mrp2 augmented renal expression. The goal of the present study is the addition of important information in the wide gap of knowledge that exists about drug-drug interactions. Because of furosemide worldwide use, the data obtained are interesting and useful in terms of translation to clinical practice.


Assuntos
Furosemida/farmacologia , Rim/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Ácido p-Aminoipúrico/farmacocinética , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Interações Medicamentosas , Furosemida/administração & dosagem , Injeções Intravenosas , Injeções Subcutâneas , Rim/metabolismo , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Ratos Wistar , Eliminação Renal/efeitos dos fármacos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/administração & dosagem , Regulação para Cima , Ácido p-Aminoipúrico/administração & dosagem , Ácido p-Aminoipúrico/sangue , Ácido p-Aminoipúrico/urina
3.
J Pharm Sci ; 103(11): 3793-3805, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25266751

RESUMO

Evidence in the literature suggests that 1α,25-dihydroxyvitamin D3 [1,25(OH)2 D3 ], the vitamin D receptor ligand, down-regulated the expression of the rat renal organic anion (renal organic anion transporter, rOAT) and oligopeptide (rPEPT) transporters, but increased intestinal rPEPT1 expression. We investigated, in rats, the intravenous and oral pharmacokinetics of 2 mg/kg cefdinir and cefadroxil, two cephalosporins that are eliminated via renal OAT1/OAT3 and are substrates of PEPT1/PEPT2, with and without 1,25(OH)2 D3 treatment. The area under the plasma concentration-time curve (AUC) of cefdinir or cefadroxil after 1,25(OH)2 D3 treatment was increased significantly because of decreased clearance (CL). Both kidney uptake and cumulative urinary recovery were significantly decreased, whereas liver uptake and fecal recovery remained unchanged in 1,25(OH)2 D3 -treated rats. Similar changes in AUC and CL were observed for both drugs upon coadministration of probenecid, the OAT inhibitor. Oral availability of cefdinir and cefadroxil remained unchanged with 1,25(OH)2 D3 treatment, suggesting lack of a role for intestinal rPEPT1. Rather, reduction of rOAT1/rOAT3 mRNA expression in kidney with 1,25(OH)2 D3 -treatment was observed, confirmed by decreased function in MDCKII cells overexpressing human OAT1 and OAT3. These composite results suggest that 1,25(OH)2 D3 treatment reduces cefdinir and cefadroxil clearances by diminution of renal OAT1/OAT3 expression, implicating a role for 1,25(OH)2 D3 in eliciting transporter-based drug interactions.


Assuntos
Calcitriol/administração & dosagem , Cefadroxila/farmacocinética , Cefalosporinas/farmacocinética , Rim/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos Sódio-Independentes/efeitos dos fármacos , Receptores de Calcitriol/agonistas , Administração Oral , Animais , Área Sob a Curva , Disponibilidade Biológica , Cefadroxila/administração & dosagem , Cefadroxila/urina , Cefdinir , Cefalosporinas/administração & dosagem , Cefalosporinas/urina , Cães , Regulação para Baixo , Interações Medicamentosas , Humanos , Injeções Intravenosas , Rim/metabolismo , Ligantes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Células Madin Darby de Rim Canino , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Transportador 1 de Peptídeos , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Eliminação Renal/efeitos dos fármacos , Simportadores/metabolismo , Distribuição Tecidual , Transfecção
4.
Am J Nephrol ; 40(1): 1-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24943263

RESUMO

BACKGROUND/AIMS: We have previously shown that 1 mg/kg indomethacin improves expression and functionality of renal organic anion transporters Oat1 and Oat3 after renal ischemia and furthermore improves renal outcome after ischemia. As we detected differential effects of COX1 or COX2 inhibitors on organic anion transport after ischemia and reperfusion in culture, we investigated the effect of the SC560 (COX1 inhibitor) and SC58125 (COX2 inhibitor) on expression of Oat1/3 and renal outcome after ischemic acute kidney injury (iAKI). METHODS: iAKI was induced in rats by bilateral clamping of renal arteries for 45 min. SC560 or SC58125 (1 mg/kg each) were given intraperitoneally as soon as reperfusion started. Sham-treated animals served as controls. Oat1/3 were determined by qPCR and Western blot. Glomerular filtration rate (GFR), p-aminohippurate (PAH) clearance and PAH extraction ratio was determined. All parameters were detected 24 h after ischemia. Renal plasma flow was calculated. RESULTS: In clamped animals SC560 (COX1 inhibitor) restored expression of Oat1/3, as well as renal perfusion. Additionally, SC560 substantially improved kidney function as measured by GFR. Application of the COX2 inhibitor SC58125 did not exert these beneficial effects. CONCLUSION: Our study indicates that COX1 inhibitor SC560 applied after ischemia prevents ischemia-induced downregulation of Oat1/3 during reperfusion and has a substantial protective effect on kidney function. Whether and to what particular extent this apparent improvement of function is mechanistically due to beneficial effects on tubular function, renal perfusion or glomerular filtration will be the scope of future studies.


Assuntos
Injúria Renal Aguda/genética , Inibidores de Ciclo-Oxigenase 2/farmacologia , Rim/efeitos dos fármacos , Pirazóis/farmacologia , RNA Mensageiro/efeitos dos fármacos , Traumatismo por Reperfusão/genética , Injúria Renal Aguda/metabolismo , Animais , Western Blotting , Inibidores de Ciclo-Oxigenase/farmacologia , Regulação para Baixo , Taxa de Filtração Glomerular/efeitos dos fármacos , Rim/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/efeitos dos fármacos , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , RNA Mensageiro/metabolismo , Ratos , Traumatismo por Reperfusão/metabolismo , Ácido p-Aminoipúrico/metabolismo
5.
Antimicrob Agents Chemother ; 58(3): 1294-301, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24295974

RESUMO

Raltegravir (RAL) is a human immunodeficiency virus type 1 (HIV-1) integrase inhibitor approved to treat HIV infection in adults in combination with other antiretrovirals. The potential of RAL to cause transporter-related drug-drug interactions (DDIs) as an inhibitor has not been well described to date. In this study, a series of in vitro experiments were conducted to assess the inhibitory effects of RAL on major human drug transporters known to be involved in clinically relevant drug interactions, including hepatic and renal uptake transporters and efflux transporters. For hepatic uptake transporters, RAL showed no inhibition of organic anion-transporting polypeptide 1B1 (OATP1B1), weak inhibition of OATP1B3 (40% inhibition at 100 µM), and no inhibition of organic cation transporter 1 (OCT1). Studies of renal uptake transporters showed that RAL inhibited organic anion transporters 1 and 3 (OAT1 and OAT3) with 50% inhibitory concentrations (IC50s) (108 µM and 18.8 µM, respectively) well above the maximum concentration of drug in plasma (Cmax) at the clinical 400-mg dose and did not inhibit organic cation transporter 2 (OCT2). As for efflux transporters, RAL did not inhibit breast cancer resistance protein (BCRP) and showed weak inhibition of multidrug and toxin extrusion protein 1 (MATE1) (52% inhibition at 100 µM) and MATE2-K (29% inhibition at 100 µM). These studies indicate that at clinically relevant exposures, RAL does not inhibit or only weakly inhibits hepatic uptake transporters OATP1B1, OATP1B3, and OCT1, renal uptake transporters OCT2, OAT1, and OAT3, as well as efflux transporters BCRP, MATE1, and MATE2-K. The propensity for RAL to cause DDIs via inhibition of these transporters is therefore considered low.


Assuntos
Inibidores de Integrase de HIV/farmacologia , Proteínas de Membrana Transportadoras/efeitos dos fármacos , Pirrolidinonas/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Animais , Cães , Interações Medicamentosas , Humanos , Técnicas In Vitro , Transportador 1 de Ânion Orgânico Específico do Fígado , Células Madin Darby de Rim Canino , Proteínas de Neoplasias/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos Sódio-Independentes/efeitos dos fármacos , Proteínas de Transporte de Cátions Orgânicos/efeitos dos fármacos , Transportador 1 de Cátions Orgânicos/efeitos dos fármacos , Transportador 2 de Cátion Orgânico , Raltegravir Potássico , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto
6.
Am J Physiol Renal Physiol ; 294(4): F867-73, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18216144

RESUMO

Organic anion transporter (OAT) genes have been implicated in renal secretion of organic anions, but the individual in vivo contributions of OAT1 (first identified as NKT) and OAT3 remain unclear. Potential substrates include loop diuretics (e.g., furosemide) and thiazide diuretics (e.g., bendroflumethiazide), which reach their tubular sites of action mainly by proximal tubular secretion. Previous experiments in Oat1 knockout (-/-) mice revealed an almost complete loss of renal secretion of the prototypic organic anion p-aminohippurate (PAH) and a role of OAT1 in tubular secretion of furosemide (Eraly SA, Vallon V, Vaughn D, Gangoiti JA, Richter K, Nagle M, Monte JC, Rieg T, Truong DM, Long JM, Barshop BA, Kaler G, Nigam SK. J Biol Chem 281: 5072-5083, 2006). In this study we found that both furosemide and bendroflumethiazide inhibited mOat1- and mOat3-mediated uptake of a labeled tracer in Xenopus oocytes injected with cRNA, consistent with their being substrates for mouse OAT1 and OAT3. Experiments in Oat3(-/-) mice revealed intact renal secretion of PAH, but the dose-natriuresis curves for furosemide and bendroflumethiazide were shifted to the right and urinary furosemide excretion was impaired similar to the defect in Oat1(-/-) mice. Thus, whereas OAT1 (in contrast to OAT3) is the classic basolateral PAH transporter of the proximal tubule, both OAT1 and OAT3 contribute similarly to normal renal secretion of furosemide and bendroflumethiazide, and a lack of either one is not fully compensated by the other. Although microarray expression analysis in the kidneys of Oat1(-/-) and Oat3(-/-) mice revealed somewhat altered expression of a small number of transport-related genes, none were common to both knockout models. When searching for polymorphisms involved in human diuretic responsiveness, it may be necessary to consider both OAT1 and OAT3, among other genes.


Assuntos
Oócitos/fisiologia , Proteína 1 Transportadora de Ânions Orgânicos/fisiologia , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , Inibidores de Simportadores de Cloreto de Sódio/farmacologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Animais , Bendroflumetiazida/farmacologia , Diuréticos/farmacologia , Feminino , Furosemida/farmacologia , Insulina/farmacologia , Oócitos/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos Sódio-Independentes/efeitos dos fármacos , Xenopus , Ácido p-Aminoipúrico/farmacologia
7.
Drug Metab Dispos ; 32(4): 424-30, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15039295

RESUMO

The human organic anion transporter 1 (hOAT1) facilitates the basolateral entry of organic anions such as endogenous metabolites, xenobiotics, and drugs into the proximal tubule cells. In the present study we investigated the general occurrence of hOAT1 isoforms in the kidneys and performed functional characterizations. Kidney specimens of 10 patients were analyzed by reverse transcription-polymerase chain reaction. We detected hOAT1-2 as the main transcript in almost all patients, and weak transcripts of hOAT1-1, hOAT1-3, and hOAT1-4 in many of them. An evaluation of the renal distribution showed all four mRNAs mostly restricted to the cortex. Western blot analysis of membrane fractions from two kidney specimens yielded two bands corresponding to the observed mRNA expression, suggesting hOAT1-3 and hOAT1-4 to be expressed on the protein level in vivo. This observation is further supported by immunofluorescence analyses of all four cloned hOAT1 isoforms transiently transfected in COS 7 cells. Functional characterizations did not show any transport activity of hOAT1-3 and hOAT1-4 for the tested substrates. Cotransfection studies of each of them with hOAT1-1 did not alter fluorescein uptake indicating no regulatory impact of these isoforms. Further functional comparisons of hOAT1-1 and hOAT1-2 in fluorescein uptake studies exhibited almost identical affinities for fluorescein with Michaelis constants of 11.6 +/- 3.7 microM (hOAT1-1) and 11.9 +/- 6.4 microM (hOAT1-2), and similar sensitivities to inhibition by p-aminohippurate [IC(50): 16 microM (hOAT1-1), 10 microM (hOAT1-2)], urate [IC(50): 440 microM (hOAT1-1), 385 microM (hOAT1-2)], and furosemide (IC(50): 14 microM (hOAT1-1), 20 microM (hOAT1-2)], implying functional equivalence.


Assuntos
Córtex Renal/citologia , Proteína 1 Transportadora de Ânions Orgânicos/genética , Isoformas de Proteínas/genética , Regulação para Cima/genética , Animais , Western Blotting/métodos , Células COS , Membrana Celular/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Chlorocebus aethiops , Clonagem de Organismos/métodos , Fluoresceína/metabolismo , Imunofluorescência/métodos , Furosemida/farmacocinética , Humanos , Córtex Renal/efeitos dos fármacos , Córtex Renal/fisiologia , Modelos Moleculares , Proteína 1 Transportadora de Ânions Orgânicos/química , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Isoformas de Proteínas/química , RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Frações Subcelulares/química , Frações Subcelulares/efeitos dos fármacos , Transfecção/métodos , Ácido Úrico/química , Ácido Úrico/farmacocinética , Ácido p-Aminoipúrico/farmacocinética
8.
Mol Pharmacol ; 62(5): 1128-36, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12391276

RESUMO

The metal chelator DMPS (2,3-dimercapto-1-propanesulfonate) is used to treat heavy metal intoxication because it increases renal excretion of these toxins, which are accumulated in proximal tubule cells. To evaluate the involvement of the organic anion transporter 1 (OAT1) in the renal flux of DMPS, we examined the effect of DMPS on transport mediated by the rabbit ortholog of OAT1 and compared these characteristics with those observed in intact isolated rabbit proximal tubules. The rabbit OAT1 (rbOAT1) cDNA consisted of 2124 base pairs encoding a protein of 551 amino acids. Heterologous expression in COS-7 cells revealed rbOAT1-mediated transport of p-aminohippurate (PAH; K(t) = 16 microM). A 1 mM concentration of unlabeled PAH, alpha-ketoglutarate, urate, or probenecid inhibited [(3)H]PAH uptake by 70 to 90%. cis-Inhibition and trans-stimulation experiments using several Krebs cycle intermediates implicated alpha-ketoglutarate as the main intracellular exchange anion. Reduced DMPS inhibited rbOAT1-mediated fluorescein transport with an apparent K(i) of 102 microM. These characteristics paralleled those observed in isolated rabbit proximal tubules. PAH was transported into nonperfused single proximal tubule S(2) segments with a K(t) of 76 microM. DMPS inhibited FL uptake into single tubule segments with a K(i-app) of 71 microM. Fluorescein efflux from preloaded tubules was trans-stimulated by 1 mM PAH and 1 mM DMPS, consistent with DMPS entry into tubule cells by rbOAT1. In summary, rbOAT1 mediates basolateral uptake of DMPS into proximal tubule cells, implicating this process in the detoxification process of heavy metals in the kidneys.


Assuntos
Quelantes/farmacologia , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Unitiol/farmacologia , Sequência de Aminoácidos , Animais , Ânions/metabolismo , Células COS , Clonagem Molecular , Haplorrinos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Dados de Sequência Molecular , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Coelhos , Homologia de Sequência de Aminoácidos , Ácido p-Aminoipúrico/farmacologia
9.
Eur J Pharmacol ; 438(3): 137-42, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11909604

RESUMO

Cephalosporin antibiotics are thought to be excreted into the urine via organic anion transporters (OATs). The purpose of this study was to elucidate the interaction of human-OATs with various cephalosporin antibiotics, using proximal tubule cells stably expressing human-OAT1, human-OAT3 and human-OAT4. Human-OAT1 and human-OAT3 are localized to the basolateral side of the proximal tubule, whereas human-OAT4 is localized to the apical side. The cephalosporin antibiotics tested were cephalothin, cefoperazone, cefazolin, ceftriaxone, cephaloridine, cefotaxime, cefadroxil and cefamandole. All of these cephalosporin antibiotics significantly inhibited organic anion uptake mediated by human-OAT1, human-OAT3 and human-OAT4. Kinetic analysis revealed that these inhibitions were competitive. The inhibition constant (K(i)) values of cefoperazone, cefazolin, ceftriaxone and cephaloridine for human-OAT1 were much lower than those for human-OAT3 and human-OAT4, whereas the K(i) values of cephalothin and cefotaxime for human-OAT3 were much lower than those for human-OAT1 and human-OAT4. Human-OAT4 mediated the bidirectional transport of estrone sulfate, an optimal substrate for human-OAT4. These results suggest that human-OAT1, human-OAT3 and human-OAT4 interact with various cephalosporin antibiotics, and that human-OAT1 and human-OAT3 play a distinct role in the basolateral uptake of cephalosporin antibiotics. Since the K(i) value of cephaloridine for human-OAT4-mediated organic uptake was much higher than that for human-OAT1, the results indicate the possibility that human-OAT4 limits the efflux of cephaloridine, leading to the accumulation of cephaloridine and the induction of nephrotoxicity.


Assuntos
Cefalosporinas/farmacologia , Estrona/análogos & derivados , Transportadores de Ânions Orgânicos/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Cefadroxila/farmacologia , Cefotaxima/farmacologia , Ceftriaxona/farmacologia , Linhagem Celular , Cefaloridina/farmacologia , Cefalotina/farmacologia , Estrona/farmacocinética , Humanos , Cinética , Transportador 1 de Ânion Orgânico Específico do Fígado , Camundongos , Camundongos Transgênicos , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Proteína 1 Transportadora de Ânions Orgânicos/genética , Proteína 1 Transportadora de Ânions Orgânicos/fisiologia , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/fisiologia , Transportadores de Ânions Orgânicos Sódio-Independentes/efeitos dos fármacos , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/fisiologia , Trítio
10.
J Pharmacol Exp Ther ; 299(2): 741-7, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11602689

RESUMO

The kidney is the primary target organ in which inorganic mercury (Hg2+) accumulates and expresses its toxic effects. The chelating agent 2,3-dimercapto-1-propanesulfonic acid (DMPS) can rapidly reduce the renal burden of mercury and increase the urinary excretion of mercury. However, the cellular and molecular basis of its efficacy is still unknown. A number of previous studies implicated the "classical organic anion secretory pathway" in the secretion of DMPS and its chelation products. In this study we used the human ortholog of the organic anion transporter (hOAT1) expressed in the Xenopus oocyte expression system to study the interaction of DMPS and its mercury chelates with hOAT1. [3H]PAH was used to show the transport activity of hOAT1 (Km = 3.9 +/-1.3 microM). Uptake of [3H]para-aminohippuric acid (PAH) was inhibited by reduced DMPS (K(i) = 22.4 +/- 8.4 microM). We also investigated the interaction of oxidized DMPS with hOAT1 because it has been shown that at least 80% of DMPS in the blood is oxidized within 30 min. Oxidized DMPS also inhibited uptake of [3H]PAH (K(i) = 66 +/-13.6 microM). In contrast, we found no interaction of the DMPS-Hg chelate with hOAT1. To determine whether DMPS and oxidized DMPS are transported by hOAT1 we examined the effect of inwardly directed gradients these two compounds on efflux of [3H]PAH from HeLa cells transiently transfected with hOAT1. Gradients of both DMPS and oxidized DMPS significantly trans-stimulated efflux of [3H]PAH. These data suggest that hOAT1 can transport DMPS and oxidized DMPS, whereas the DMPS-Hg chelate has no significant affinity for the transporter.


Assuntos
Quelantes/farmacologia , Proteína 1 Transportadora de Ânions Orgânicos/efeitos dos fármacos , Unitiol/farmacologia , Albuminas/farmacologia , Células HeLa , Humanos , Cinética , Mercúrio/farmacologia , Oxirredução , Ácido p-Aminoipúrico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA