Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Pflugers Arch ; 468(11-12): 1909-1918, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27812757

RESUMO

Dantrolene is the only available drug for the treatment of malignant hyperthermia, a life-threatening inborn sensitivity of the ryanodine receptor (RyR1) in skeletal muscles to volatile anesthetics. Dantrolene is metabolized in the liver to 5-OH dantrolene. Both compounds are zwitterions or net negatively charged. Here, we investigated interactions of dantrolene and 5-OH dantrolene with solute carrier (SLC) family members occurring in skeletal muscle cells, hepatocytes, and renal proximal tubule cells. SLC22A8 (organic anion transporter 3, OAT3) was very sensitive to both compounds exhibiting IC50 values of 0.35 ± 0.03 and 1.84 ± 0.34 µM, respectively. These IC50 concentrations are well below the plasma concentration in patients treated with dantrolene (3-28 µM). SLC22A7 (OAT2) was less sensitive to dantrolene and 5-OH dantrolene with IC50 values of 15.6 ± 2.1 and 15.8 ± 3.2 µM, respectively. SLCO1B1 (OATP1B1), SLCO1B3 (OATP1B3), and SLCO2B1 (OATP2B1) mainly interacted with 5-OH dantrolene albeit with higher IC50 values than those observed for OAT2 and OAT3. Dantrolene and 5-OH dantrolene failed to inhibit uptake of 1-methyl-4-phenylpyrimidinium (MPP) by OCT1 and of carnitine by OCTN2. In counter-flow experiments on OAT3, dantrolene and 5-OH dantrolene decreased pre-equilibrated cellular [3H]estrone-3-sulfate (ES) content as did the transported substrates glutarate, furosemide, and bumetanide. With OAT2, dantrolene and 5-OH dantrolene slightly decreased the pre-equilibrated [3H]cGMP content. If no other transporter markedly contributes to uptake or release of ES or cGMP, respectively, these data suggest that OAT3 and OAT2 may be involved in absorption, metabolism, and excretion of dantrolene and its metabolite 5-OH dantrolene.


Assuntos
Dantroleno/farmacologia , Relaxantes Musculares Centrais/farmacologia , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Transporte Biológico , Células HEK293 , Humanos , Ligação Proteica
2.
Am J Physiol Renal Physiol ; 311(1): F227-38, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27053689

RESUMO

The initial step in renal secretion of organic anions (OAs) is mediated by transporters in the basolateral membrane (BLM). Contributors to this process are primary active Na(+)-K(+)-ATPase (EC 3.6.3.9), secondary active Na(+)-dicarboxylate cotransporter 3 (NaDC3/SLC13A3), and tertiary active OA transporters (OATs) OAT1/SLC22A6, OAT2/SLC22A7, and OAT3/SLC22A8. In human kidneys, we analyzed the localization of these transporters by immunochemical methods in tissue cryosections and isolated membranes. The specificity of antibodies was validated with human embryonic kidney-293 cells stably transfected with functional OATs. Na(+)-K(+)-ATPase was immunolocalized to the BLM along the entire human nephron. NaDC3-related immunostaining was detected in the BLM of proximal tubules and in the BLM and/or luminal membrane of principal cells in connecting segments and collecting ducts. The thin and thick ascending limbs, macula densa, and distal tubules exhibited no reactivity with the anti-NaDC3 antibody. OAT1-OAT3-related immunostaining in human kidneys was detected only in the BLM of cortical proximal tubules; all three OATs were stained more intensely in S1/S2 segments compared with S3 segment in medullary rays, whereas the S3 segment in the outer stripe remained unstained. Expression of NaDC3, OAT1, OAT2, and OAT3 proteins exhibited considerable interindividual variability in both male and female kidneys, and sex differences in their expression could not be detected. Our experiments provide a side-by-side comparison of basolateral transporters cooperating in renal OA secretion in the human kidney.


Assuntos
Transportadores de Ácidos Dicarboxílicos/metabolismo , Néfrons/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Simportadores/metabolismo , Adulto , Feminino , Células HEK293 , Humanos , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Túbulos Renais Distais/metabolismo , Masculino , Membranas/metabolismo , Pessoa de Meia-Idade , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Caracteres Sexuais , ATPase Trocadora de Sódio-Potássio/metabolismo
3.
Croat Med J ; 56(5): 447-59, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26526882

RESUMO

AIM: To investigate whether the sex-dependent expression of hepatic and renal oxalate transporter sat-1 (Slc26a1) changes in a rat model of ethylene glycol (EG)-induced hyperoxaluria. METHODS: Rats were given tap water (12 males and 12 females; controls) or EG (12 males and 12 females; 0.75% v/v in tap water) for one month. Oxaluric state was confirmed by biochemical parameters in blood plasma, urine, and tissues. Expression of sat-1 and rate-limiting enzymes of oxalate synthesis, alcohol dehydrogenase 1 (Adh1) and hydroxy-acid oxidase 1 (Hao1), was determined by immunocytochemistry (protein) and/or real time reverse transcription polymerase chain reaction (mRNA). RESULTS: EG-treated males had significantly higher (in µmol/L; mean±standard deviation) plasma (59.7±27.2 vs 12.9±4.1, P<0.001) and urine (3716±1726 vs 241±204, P<0.001) oxalate levels, and more abundant oxalate crystaluria than controls, while the liver and kidney sat-1 protein and mRNA expression did not differ significantly between these groups. EG-treated females, in comparison with controls had significantly higher (in µmol/L) serum oxalate levels (18.8±2.9 vs 11.6±4.9, P<0.001), unchanged urine oxalate levels, low oxalate crystaluria, and significantly higher expression (in relative fluorescence units) of the liver (1.59±0.61 vs 0.56±0.39, P=0.006) and kidney (1.77±0.42 vs 0.69±0.27, P<0.001) sat-1 protein, but not mRNA. The mRNA expression of Adh1 was female-dominant and that of Hao1 male-dominant, but both were unaffected by EG treatment. CONCLUSIONS: An increased expression of hepatic and renal oxalate transporting protein sat-1 in EG-treated female rats could protect from hyperoxaluria and oxalate urolithiasis.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Antiporters/metabolismo , Etilenoglicol/uso terapêutico , Hiperoxalúria/prevenção & controle , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Álcool Desidrogenase/genética , Álcool Desidrogenase/metabolismo , Animais , Proteínas de Transporte de Ânions/genética , Antiporters/genética , Western Blotting , Oxalato de Cálcio/sangue , Oxalato de Cálcio/urina , Cromatografia Líquida de Alta Pressão , Feminino , Hiperoxalúria/metabolismo , Rim/metabolismo , Fígado/metabolismo , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Fatores Sexuais , Transportadores de Sulfato
4.
Eur J Med Chem ; 92: 723-31, 2015 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-25618019

RESUMO

Antineoplastic compounds are used in the treatment of a variety of cancers. The effectiveness of an antineoplastic compound to exert its activity is largely dependent on transport proteins involved in the entry of the compound into the cells, and those which drive it out of the cell. Organic anion transporting polypeptide 1B1 (OATP1B1) and organic anion transporting polypeptide 1B3 (OATP1B3), belonging to the SLCO family of proteins, are specifically expressed in the sinusoidal membranes of the liver, and are known to interact with a variety of drugs. The present study deals with the interaction of these proteins with antineoplastic compounds routinely used in cancer chemotherapy. The proteins OATP1B1 and OATP1B3 were functionally characterized in stably transfected human embryonic kidney cells using [(3)H] labeled estrone 3-sulfate and [(3)H] labeled cholecystokinin octapeptide (CCK-8) as substrates, respectively. Substrate uptake experiments performed in the presence of antineoplastic compounds showed that vinblastine and paclitaxel strongly interacted with the OATP1B1 with Ki values of 10.2 µM and 0.84 µM, respectively. OATP1B3 showed highly significant interactions with a variety of antineoplastic compounds including chlorambucil, mitoxantrone, vinblastine, vincristine, paclitaxel and etoposide, with Ki values of 40.6 µM, 3.2 µM, 15.9 µM, 30.6 µM, 1.8 µM and 13.5 µM, respectively. We report several novel interactions of the transporter proteins OATP1B1 and OATP1B3 highlighting the need to investigate their role in drug-drug interactions and cancer chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Peptídeos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/síntese química , Protocolos de Quimioterapia Combinada Antineoplásica/química , Linhagem Celular , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado , Estrutura Molecular , Transportadores de Ânions Orgânicos/química , Transportadores de Ânions Orgânicos Sódio-Independentes/química , Peptídeos/química , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto
5.
Pharmacol Res ; 91: 78-87, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25481222

RESUMO

The ability of an antineoplastic drug to exert its cytostatic effect depends largely on the balance between its uptake into and extrusion from the cancer cells. ATP driven efflux transporter proteins drive the export of antineoplastic drugs and play a pivotal role in the development of chemoresistance. As regards uptake transporters, comparably less is known on their impact in drug action. In the current study, we characterized the interactions of two uptake transporter proteins, expressed mainly in the liver; the organic anion transporter 2 (OAT2, encoded by the SLC22A7 gene) and the sodium taurocholate cotransporting polypeptide (NTCP, encoded by the SLC10A1 gene), stably transfected in human embryonic kidney cells, with some antineoplastic agents that are routinely being used in cancer chemotherapy. Whereas NTCP did not show any strong interactions with the cytostatics tested, we observed a very strong inhibition of OAT2 mediated [(3)H] cGMP uptake in the presence of bendamustine, irinotecan and paclitaxel. The Ki values of OAT2 for bendamustine, irinotecan and paclitaxel were determined to be 43.3±4.33µM, 26.4±2.34µM and 10.4±0.45µM, respectively. Incubation of bendamustine with OAT2 expressing cells increased the caspase-3 activity, and this increase was inhibited by simultaneous incubation with bendamustine and probenecid, a well-known inhibitor of OATs, suggesting that bendamustine is a substrate of OAT2. A higher accumulation of irinotecan was observed in OAT2 expressing cells compared to control pcDNA cells by HPLC analysis of cell lysates. The accumulation was diminished in the presence of cGMP, the substrate we used to functionally characterize OAT2, suggesting specificity of this uptake and the fact that OAT2 mediates uptake of irinotecan.


Assuntos
Antineoplásicos/farmacologia , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Simportadores/metabolismo , Apoptose/efeitos dos fármacos , Cloridrato de Bendamustina , Transporte Biológico , Camptotecina/análogos & derivados , Camptotecina/farmacologia , GMP Cíclico/metabolismo , Estrona/análogos & derivados , Estrona/metabolismo , Células HEK293 , Humanos , Irinotecano , Compostos de Mostarda Nitrogenada/farmacologia , Paclitaxel/farmacologia
6.
Am J Physiol Renal Physiol ; 308(4): F330-8, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25477469

RESUMO

Chronic lymphatic leukemia (CLL) is often associated with nephritic syndrome. Effective treatment of CLL by chlorambucil and bendamustine leads to the restoration of renal function. In this contribution, we sought to elucidate the impact of organic anion transporters (OATs) on the uptake of bendamustine and chlorambucil as a probable reason for the superior efficacy of bendamustine over chlorambucil in the treatment of CLL. We examined the effects of structural analogs of p-aminohippurate (PAH), melphalan, chlorambucil, and bendamustine, on OAT1-mediated [(3)H]PAH uptake and OAT3- and OAT4-mediated [(3)H]estrone sulfate (ES) uptake in stably transfected human embryonic kidney-293 cells. Melphalan had no significant inhibitory effect on any OAT, whereas chlorambucil reduced OAT1-, OAT3-, and OAT4-mediated uptake of PAH or ES down to 14.6%, 16.3%, and 66.0% of control, respectively. Bendamustine inhibited only OAT3-mediated ES uptake, which was reduced down to 14.3% of control cells, suggesting that it interacts exclusively with OAT3. The IC50 value for OAT3 was calculated to be 0.8 µM. Real-time PCR experiments demonstrated a high expression of OAT3 in lymphoma cell lines as well as primary CLL cells. OAT3-mediated accumulation of bendamustine was associated with reduced cell proliferation and an increased rate of apoptosis. We conclude that the high efficacy of bendamustine in treating CLL might be partly contributed to the expression of OAT3 in lymphoma cells and the high affinity of bendamustine for this transporter.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Leucemia Linfocítica Crônica de Células B/metabolismo , Linfoma de Células T/metabolismo , Compostos de Mostarda Nitrogenada/farmacologia , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Antineoplásicos Alquilantes/metabolismo , Apoptose/efeitos dos fármacos , Cloridrato de Bendamustina , Proliferação de Células/efeitos dos fármacos , Clorambucila/metabolismo , Clorambucila/farmacologia , Relação Dose-Resposta a Droga , Estrona/análogos & derivados , Estrona/metabolismo , Células HEK293 , Humanos , Células Jurkat , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Linfoma de Células T/genética , Linfoma de Células T/patologia , Melfalan/metabolismo , Melfalan/farmacologia , Compostos de Mostarda Nitrogenada/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/antagonistas & inibidores , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/antagonistas & inibidores , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transfecção , Células Tumorais Cultivadas , Ácido p-Aminoipúrico/metabolismo
7.
Am J Physiol Renal Physiol ; 307(12): F1373-9, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25354943

RESUMO

Inborn defects in N-acetylglutamate (NAG) synthase (NAGS) cause a reduction of NAG, an essential cofactor for the initiation of the urea cycle. As a consequence, blood ammonium concentrations are elevated, leading to severe neurological disorders. The orphan drug N-carbamoylglutamate (NCG; Carbaglu), efficiently overcomes NAGS deficiency. However, not much is known about the transporters involved in the uptake, distribution, and elimination of the divalent organic anion NCG. Organic anion-transporting polypeptides (OATPs) as well as organic anion transporters (OATs) working in cooperation with sodium dicarboxylate cotransporter 3 (NaDC3) accept a wide variety of structurally unrelated drugs. To test for possible interactions with OATPs and OATs, the impact of NCG on these transporters in stably transfected human embryonic kidney-293 cells was measured. The two-electrode voltage-clamp technique was used to monitor NCG-mediated currents in Xenopus laevis oocytes that expressed NaDC3. Neither OATPs nor OAT2 and OAT3 interacted with NCG, but OAT1 transported NCG. In addition, NCG was identified as a high-affinity substrate of NaDC3. Preincubation of OAT4-transfected human embryonic kidney-293 cells with NCG showed an increased uptake of estrone sulfate, the reference substrate of OAT4, indicating efflux of NCG by OAT4. In summary, NaDC3 and, to a lesser extent, OAT1 are likely to be responsible for the uptake of NCG from the blood. Efflux of NCG across the luminal membrane into the tubular lumen probably occurs by OAT4 completing renal secretion of this drug.


Assuntos
Transportadores de Ácidos Dicarboxílicos/metabolismo , Glutamatos/metabolismo , Túbulos Renais Proximais/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Eliminação Renal , Simportadores/metabolismo , Distúrbios Congênitos do Ciclo da Ureia/tratamento farmacológico , Aminoácido N-Acetiltransferase , Animais , Transportadores de Ácidos Dicarboxílicos/genética , Células HEK293 , Humanos , Potenciais da Membrana , Proteína 1 Transportadora de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , Simportadores/genética , Transfecção , Distúrbios Congênitos do Ciclo da Ureia/enzimologia , Xenopus laevis
8.
Am J Physiol Renal Physiol ; 307(11): F1283-91, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25234312

RESUMO

The human organic anion transporter 1 (OAT1) is crucial for the excretion of organic anions in renal proximal tubular cells and has been classified as a clinically relevant transporter in the kidneys. Our previous study indicated that renal male-predominant expression of rat Oat1 and Oat3 appears to be regulated by transcription factor B-cell CLL/lymphoma 6 (BCL6). The aim of this study was to characterize the effect of BCL6 on human OAT1 promoter and on the transcription of OAT1 mediated by hepatocyte nuclear factor-1α (HNF-1α). Luciferase assays were carried out in opossum kidney (OK) cells transiently transfected with promoter constructs of OAT1, expression vectors for BCL6 and HNF-1α, and the empty control vectors. BCL6 and HNF-1α binding on OAT1 promoter was analyzed using electrophoretic mobility shift assay (EMSA). Protein expression of HNF-1α was investigated by Western blot analysis. Site-directed mutagenesis was used to introduce mutations into BCL6 and HNF-1α binding sites within the OAT1 promoter. BCL6 enhanced the promoter activity of OAT1 independently of predicted BCL6 binding sites but was dependent on HNF-1α response element and HNF-1α protein. Coexpression of BCL6 and HNF-1α induced an additive effect on OAT1 promoter activation compared with BCL6 or HNF-1α alone. BCL6 does not bind directly or indirectly to OAT1 promoter but increases the protein expression of HNF-1α and thereby indirectly enhances OAT1 gene transcription. BCL6 constitutes a promising candidate gene for the regulation of human OAT1 transcription and other renal and/or hepatic drug transporters that have been already shown to be activated by HNF-1α.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/genética , Gambás/fisiologia , Proteína 1 Transportadora de Ânions Orgânicos/biossíntese , Animais , Linhagem Celular , DNA/metabolismo , Fator 1-alfa Nuclear de Hepatócito/biossíntese , Fator 1-alfa Nuclear de Hepatócito/genética , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Humanos , Rim/metabolismo , Mutagênese Sítio-Dirigida , Mutação/genética , Mutação/fisiologia , Proteína 1 Transportadora de Ânions Orgânicos/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-bcl-6
9.
J Pharm Sci ; 103(10): 3326-34, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25175346

RESUMO

Increased expression of transporters-mediating uptake of antineoplastic drugs could render renal cell carcinoma (RCC) more sensitive to chemotherapy. Here, we studied the effect of hepatocyte nuclear factor 4α (HNF4α) on the expression of selected uptake transporters in RCC lines. Organic cation transporters (OCTs) and organic anion transporters (OATs) mRNA levels in HNF4α-transfected RCCs were measured by real-time PCR. Expression of HNF4α, ß-catenin, N-cadherin, and E-cadherin was detected by immunofluorescence. OCT1, OAT2, and concentrative nucleoside transporter 3 (CNT3) were tested using tritium-labeled substrates and an apoptosis assay. Most RCC did not express uptake transporters in the absence or presence of HNF4α. In RCCNG1 cells, HNF4α-expression increased the chemosensitivity to oxaliplatin and enhanced the accumulation of methyl-4-phenylpyridinium acetate, a model substrate for OCT1. Furthermore, HNF4α enhanced OAT2 mRNA and increased caspase-3 activity upon incubation with a purported OAT2 substrate, 5-fluorouracil (5-FU). However, functional OAT2 protein was not upregulated. CNT3 mRNA was significantly elevated by HNF4α. Inhibition of CNT3-mediated uridine uptake by 5-FU metabolite 5-fluoro-2'-deoxyuridine suggested the involvement of CNT3 in increased caspase-3 activity. Our data suggest that HNF4α increases the expression of OCT1 and CNT3 in RCCNG1 cells, thereby increasing the chemosensitivity of tumor cells to oxaliplatin and 5-FU.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Renais/patologia , Fluoruracila/farmacologia , Fator 4 Nuclear de Hepatócito/fisiologia , Neoplasias Renais/patologia , Proteínas de Membrana Transportadoras/fisiologia , Transportador 1 de Cátions Orgânicos/fisiologia , Compostos Organoplatínicos/farmacologia , Linhagem Celular Tumoral , Humanos , Oxaliplatina , Reação em Cadeia da Polimerase em Tempo Real
10.
Nephron Physiol ; 124(1-2): 1-5, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24247155

RESUMO

BACKGROUND/AIMS: During a single pass through the kidneys, more than 80% of glutathione (GSH) is excreted, indicating not only glomerular filtration, but also tubular secretion. The first step in tubular secretion is the uptake of a substance across the basolateral membrane of proximal tubule cells by sodium-dependent and -independent transporters. Due to the dicarboxylate-like structure, we postulated that GSH uptake across the basolateral membrane is mediated by the sodium-dependent dicarboxylate transporter 3 (NaDC3). METHODS: Tracer uptake and electrophysiologic measurements using a two-electrode voltage clamp device were performed in Xenopus laevis oocytes expressing the human (h)NaDC3. RESULTS: Uptake of succinate, the reference substrate of hNaDC3, was inhibited by GSH in a dose-dependent manner with an IC50 of 1.88 mM. GSH evoked potential-dependent inward currents, which were abolished under sodium-free conditions. At -60 mV, GSH currents showed saturation kinetics with a KM of 1.65 mM. CONCLUSION: hNaDC3 present at the basolateral membrane of proximal tubule cells mediates sodium-dependent GSH uptake. The kinetic data show that NaDC3 is a low-affinity GSH transporter.


Assuntos
Glutationa/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Ácido Succínico/metabolismo , Simportadores/metabolismo , Animais , Humanos , Túbulos Renais Proximais/metabolismo , Fígado/metabolismo , Oócitos/metabolismo , Transportadores de Ânions Orgânicos Dependentes de Sódio/genética , RNA Complementar/genética , Simportadores/genética , Xenopus laevis/genética
11.
Am J Physiol Renal Physiol ; 304(4): F403-9, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23255614

RESUMO

Due to their clearance function, the kidneys are exposed to high concentrations of oxidants and potentially toxic substances. To maintain cellular integrity, renal cells have to be protected by sufficient concentrations of the antioxidant glutathione (GSH). We tested whether GSH or its precursors are taken up by human organic anion transporters 1 (OAT1) and 3 (OAT3) stably expressed in HEK293 cells. GSH did not inhibit uptake of p-aminohippurate (PAH) or of estrone sulfate (ES) in OAT3-transfected HEK293 cells. In OAT1-transfected cells, GSH reduced the uptake of PAH marginally. Among the GSH constituent amino acids, glutamate, cysteine, and glycine, only glutamate inhibited OAT1, but labeled glutamate was not taken up by a probenecid-inhibitable transport system. Thus OAT1 binds glutamate but is unable to translocate it. The GSH precursor dipeptide, cysteinyl glycine (cysgly), and the glutamate derivative N-acetyl glutamate (NAG), inhibited uptake of PAH when present in the medium and trans-stimulated uptake of PAH from the intracellular side, indicating that they are hitherto unrecognized transported substrates of OAT1. N-acetyl aspartate weakly interacted with OAT1, but aspartate did not. NAG inhibited also OAT3, albeit with much lower affinity compared with OAT1, and glutamate did not interact with OAT3 at all. Taken together, human OAT3 and OAT1 cannot be involved in renal GSH extraction from the blood. However, OAT1 could support intracellular GSH synthesis by taking up cysteinyl glycine.


Assuntos
Glutamatos/metabolismo , Glutationa/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Ácidos Aminoipúricos/metabolismo , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Transporte Biológico , Cisteína/metabolismo , Dipeptídeos/metabolismo , Estrona/análogos & derivados , Estrona/metabolismo , Glicina/metabolismo , Células HEK293 , Humanos , Rim/efeitos dos fármacos , Rim/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Probenecid/farmacologia , Transfecção , Uricosúricos/farmacologia
12.
Pflugers Arch ; 464(4): 367-74, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22875277

RESUMO

2-Oxoglutarate or α-ketoglutarate (αKG) is a substrate of HIF prolyl hydroxylases 1-3 that decrease cellular levels of the hypoxia-inducible factor 1α (HIF-1α) in the presence of oxygen. αKG analogs are applied to stabilize HIF-1α even in the presence of oxygen and thus provide a novel therapeutic option in treating kidney diseases. In the kidneys, the organic anion transporters 1 and 3 (OAT1 and OAT3, respectively) in cooperation with the sodium-dependent dicarboxylate transporter 3 (NaDC3) and the OAT4 might be responsible for the uptake of αKG analogs into and the efflux out of the tubular cells. Using the radiolabelled substrates p-aminohippurate (PAH, OAT1), estrone-3-sulfate (ES; OAT3, OAT4), and succinate (NaDC3), N-oxalylglycine (NOG), dimethyloxalyl glycine (DMOG), 2,4-diethylpyridine dicarboxylate (2,4-DPD), and pyridine-2,4-dicarboxylic acid (PDCA) were tested in cis-inhibition and trans-stimulation experiments. None of these αKG analogs interacted with NaDC3. 2,4-DPD and PDCA inhibited ES uptake by OAT3 moderately. NOG, 2,4-DPD and PDCA, but not DMOG, inhibited PAH uptake by OAT1 significantly. trans-Stimulation experiments and experiments demonstrating stabilization of HIF-1α revealed that NOG and PDCA, but not 2,4-DPD, are translocated by OAT1. All compounds trans-stimulated ES uptake by OAT4, but only PDCA stabilized HIF-1α. The data suggest that OAT1 is involved in the uptake of NOG and PDCA across the basolateral membrane of proximal tubule cells, whereas OAT4 may release these compounds into the primary urine.


Assuntos
Dioxigenases/antagonistas & inibidores , Ácidos Cetoglutáricos/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Aminoácidos Dicarboxílicos/metabolismo , Transporte Biológico Ativo , Estrona/análogos & derivados , Estrona/metabolismo , Células HEK293 , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Transportadores de Ânions Orgânicos/efeitos dos fármacos , Transportadores de Ânions Orgânicos Dependentes de Sódio/efeitos dos fármacos , Transportadores de Ânions Orgânicos Dependentes de Sódio/metabolismo , Piridinas/metabolismo , Ácido Succínico/metabolismo , Simportadores/efeitos dos fármacos , Simportadores/metabolismo , Ácido p-Aminoipúrico/metabolismo
13.
Pharmacol Ther ; 136(1): 106-30, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22841915

RESUMO

Common to all so far functionally characterized Organic Anion Transporters (OATs) is their broad substrate specificity and their ability to exchange extracellular against intracellular organic anions. Many OATs occur in renal proximal tubules, the site of active drug secretion. Exceptions are murine Oat6 (nasal epithelium), human OAT7 (liver), and rat Oat8 (renal collecting ducts). In human kidneys, OAT1, OAT2, and OAT3 are localized in the basolateral membrane, and OAT4, OAT10, and URAT1 in the apical cell membrane of proximal tubule cells, respectively. In rats and mice, Oat1 and Oat3 are located basolaterally, and Oat2, Oat5, Oat9, Oat10, and Urat1 apically. Several classes of drugs interact with human OAT1-3, including ACE inhibitors, angiotensin II receptor antagonists, diuretics, HMG CoA reductase inhibitors, ß-lactam antibiotics, antineoplastic and antiviral drugs, and uricosuric drugs. For most drugs, interaction was demonstrated in vitro by inhibition of OAT-mediated transport of model substrates; for some drugs, transport by OATs was directly proven. Based on IC50 values reported in the literature, OAT1 and OAT3 show comparable affinities for diuretics, cephalosporins, and nonsteroidal anti-inflammatory drugs whereas OAT2 has a lower affinity to most of these compounds. Drug-drug interactions at OAT1 and OAT3 may retard renal drug secretion and cause untoward effects. OAT4, OAT10, and URAT1 in the apical membrane contribute to proximal tubular urate absorption, and OAT10 to nicotinate absorption. OAT4 is in addition able to release drugs, e.g. diuretics, into the tubule lumen.


Assuntos
Transportadores de Ânions Orgânicos/fisiologia , Preparações Farmacêuticas/metabolismo , Animais , Transporte Biológico , Interações Medicamentosas , Regulação da Expressão Gênica , Humanos , Transportadores de Ânions Orgânicos/genética , Polimorfismo de Nucleotídeo Único , Caracteres Sexuais , Especificidade da Espécie , Especificidade por Substrato
14.
PLoS One ; 7(4): e35556, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22530049

RESUMO

BACKGROUND: Organic anion transporters 1 (Oat1) and 3 (Oat3) mediate the transport of organic anions, including frequently prescribed drugs, across cell membranes in kidney proximal tubule cells. In rats, these transporters are known to be male-dominant and testosterone-dependently expressed. The molecular mechanisms that are involved in the sex-dependent expression are unknown. Our aim was to identify genes that show a sex-dependent expression and could be involved in male-dominant regulation of Oat1 and Oat3. METHODOLOGY/PRINCIPAL FINDINGS: Promoter activities of Oat1 and Oat3 were analyzed using luciferase assays. Expression profiling was done using a SurePrint G3 rat GE 8 × 60K microarray. RNA was isolated from renal cortical slices of four adult rats per sex. To filter the achieved microarray data for genes expressed in proximal tubule cells, transcription database alignment was carried out. We demonstrate that predicted androgen response elements in the promoters of Oat1 and Oat3 are not functional when the promoters were expressed in OK cells. Using microarray analyses we analyzed 17,406 different genes. Out of these genes, 56 exhibit a sex-dependent expression in rat proximal tubule cells. As genes potentially involved in the regulation of Oat1 and Oat3 expression, we identified, amongst others, the male-dominant hydroxysteroid (17-beta) dehydrogenase 1 (Hsd17b1), B-cell CLL/lymphoma 6 (BCL6), and polymerase (RNA) III (DNA directed) polypeptide G (Polr3g). Moreover, our results revealed that the transcription factor BCL6 activates promoter constructs of Oat1 and Oat3. CONCLUSION: The results indicate that the male-dominant expression of both transporters, Oat1 and Oat3, is possibly not directly regulated by the classical androgen receptor mediated transcriptional pathway but appears to be regulated by the transcription factor BCL6.


Assuntos
Túbulos Renais Proximais/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos Sódio-Independentes/genética , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Ativação Transcricional , Animais , Sítios de Ligação , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Túbulos Renais Proximais/efeitos dos fármacos , Masculino , Regiões Promotoras Genéticas/efeitos dos fármacos , Ratos , Ratos Wistar , Fatores Sexuais , Testosterona/farmacologia
15.
J Pharmacol Exp Ther ; 341(1): 16-23, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22202118

RESUMO

Antineoplastic agents directed at nuclear and cytoplasmic targets in tumor cells represent the current mainstay of treatment for patients with disseminated malignant diseases. Cellular uptake of antineoplastics is a prerequisite for their efficacy. Five of six lymphoma cell lines as well as primary samples from chronic lymphocytic leukemia patients demonstrated significant expression of SLC22A1 mRNA coding for organic cation transporter 1 (OCT1). Functionally, the antineoplastic agents irinotecan, mitoxantrone, and paclitaxel inhibited the uptake of the organic cation [(3)H]1-methyl-4-pyridinium iodide into OCT1-transfected Chinese hamster ovary model cells, with K(i) values of 1.7, 85, and 50 µM, respectively. Correspondingly, OCT1-positive cell lines and transfectants exhibited significantly higher susceptibilities to the cytotoxic effects of irinotecan and paclitaxel compared with those of OCT1-negative controls. We hypothesize that OCT1 can contribute to the susceptibility of cancer cells to selected antineoplastic drugs. In the future, an expression analysis of the transporters and the application of transporter-specific antineoplastic agents could help to tailor cancer therapy.


Assuntos
Camptotecina/análogos & derivados , Regulação Neoplásica da Expressão Gênica , Linfoma/metabolismo , Transportador 1 de Cátions Orgânicos/biossíntese , Paclitaxel/metabolismo , Animais , Células CHO , Camptotecina/metabolismo , Camptotecina/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Cricetinae , Cricetulus , Humanos , Irinotecano , Paclitaxel/farmacologia
16.
Drug Metabol Drug Interact ; 26(4): 181-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22149660

RESUMO

BACKGROUND: The transport of endogenous and exogenous organic cations across the plasma membrane of cells is mediated by multispecific organic cation transporters (OCTs), and the multidrug and toxin extrusion (MATE) transporters. MATE belongs to the SLC47 transporter family consisting of only two members, MATE1 and MATE2-K. MATE2-K is exclusively expressed in the kidney at the apical membrane of proximal tubular epithelial cells. MATE1 is highly expressed in the kidney, liver, skeletal muscle and also in adrenal glands, testes and heart. MATE1 exchanges organic cations against protons both in influx as well as in efflux modes. METHODS: Here, we examined the interaction of 25 antineoplastic agents with human MATE1. We generated stably transfected MATE1-HEK293 cells and determined the inhibition of MATE1-mediated [(3)H]1-methyl-4-phenylpyridinium (MPP) uptake by the antineoplastic agents. RESULTS: We found a significant inhibition of MATE1-mediated MPP uptake by several antineoplastic agents and pH dependent IC(50)values for mitoxantrone (7.8 µM at pH 7.4 and 0.6 µM at pH 8.5) as well as for irinotecan (4.4 µM at pH 7.4 and 1.1 µM at pH 8.5), respectively. CONCLUSIONS: We suggest that hMATE1 could play a role in chemosensitivity of tumor cells. In addition, hepatic and renal MATE1 could potentially be involved in drug-drug-interactions as well as in drug metabolism and excretion during chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Irinotecano , Mitoxantrona/farmacologia , Proteínas de Transporte de Cátions Orgânicos/fisiologia , Transfecção
17.
Biol Chem ; 392(1-2): 117-24, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21194368

RESUMO

The response to chemotherapy by tumor cells depends on the concentration of cytostatics accumulated inside the cells. The accumulation of anticancer drugs in tumor cells is mainly dependent on functional expression of efflux and influx transporters and to a minor extent on passive diffusion through the membrane. Efflux transporters of the ABC family are partially responsible for the chemoresistance of cancer cells by secreting these cytostatics. Over the past decades, the role of ABC transporters in the chemoresistance of various malignant tumors has been very well documented. By contrast, very little is known about the impact on tumor therapy of influx transporters belonging to the solute carrier transporters (SLC family). In this review, we focus on the interaction of SLC22 transporters with cytostatics, the expression of these transporters in tumor cells as well as their impact on the chemosensitivity of cancer cells.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Neoplasias/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Linhagem Celular Tumoral , Células/efeitos dos fármacos , Humanos
18.
Nephrol Dial Transplant ; 26(7): 2175-81, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21148271

RESUMO

BACKGROUND: Hereditary renal hypouricemia may be complicated by nephrolithiasis or exercise-induced acute renal failure. Most patients described so far are of Japanese origin and carry the truncating mutation W258X in the uric acid transporter URAT1 encoded by SLC22A12. Recently, we described severe renal hypouricemia in Israeli patients with uric acid transporter GLUT9 (SLC2A9) loss-of-function mutations. Renal hypouricemia in Iraqi Jews has been previously reported, but its molecular basis has not been ascertained. METHODS: Three Jewish Israeli families of Iraqi origin with hereditary hypouricemia and hyperuricosuria were clinically characterized. DNA was extracted and the URAT1 gene was sequenced. Transport studies into Xenopus laevis oocytes were utilized to evaluate the function of URAT1 mutants found. RESULTS: A missense URAT1 mutation, R406C, was detected in all three families. Two affected siblings were found to carry in addition a homozygous missense URAT1 mutation, G444R. Both mutations dramatically impaired urate uptake into X. laevis oocytes. Moreover, we demonstrate for the first time that URAT1 facilitates urate efflux, which was abolished in the mutants, indicating also a secretion defect. Homozygous patients had serum uric acid concentrations of 0.5-0.8 mg% and a fractional excretion of uric acid of 50-85%. Most individuals studied were asymptomatic, two had nephrolithiasis and none developed exercise-induced acute renal failure. CONCLUSIONS: The URAT1 R406C mutation detected in all three families is likely to be the founder mutation in Iraqi Jews. Our findings contribute to a better definition of the different types of hereditary renal hypouricemia and suggest that the phenotype of this disorder depends mainly on the degree of inhibition of uric acid transport.


Assuntos
Judeus/genética , Mutação de Sentido Incorreto/genética , Transportadores de Ânions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/genética , Erros Inatos do Transporte Tubular Renal/etiologia , Cálculos Urinários/etiologia , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Animais , Células Cultivadas , Família , Feminino , Homozigoto , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Oócitos/citologia , Oócitos/metabolismo , Fenótipo , Erros Inatos do Transporte Tubular Renal/patologia , Homologia de Sequência de Aminoácidos , Cálculos Urinários/patologia , Xenopus laevis/metabolismo
19.
Mol Pharm ; 8(1): 270-9, 2011 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-21077648

RESUMO

Anticancer treatment with ifosfamide but not with its structural isomer cyclophosphamide is associated with development of renal Fanconi syndrome leading to diminished growth in children and bone problems in adults. Since both cytotoxics share the same principal metabolites, we investigated whether a specific renal uptake of ifosfamide is the basis for this differential effect. First we studied the interaction of these cytotoxics using cells transfected with organic anion or cation transporters and freshly isolated murine and human proximal tubules with appropriate tracers. Next we determined changes in membrane voltage in proximal tubular cells to understand their differentiated nephrotoxicity. Ifosfamide but not cyclophosphamide was significantly transported into cells expressing human organic cation transporter 2 (hOCT2) while both did not interact with organic anion transporters. This points toward a specific interaction of ifosfamide with hOCT2, which is the main OCT isoform in human kidney. In isolated human proximal tubules ifosfamide also interacted with organic cation transport. This interaction was also seen in isolated mouse proximal tubules; however, it was absent in tubules from OCT-deficient mice, illustrating the biological importance of this selective transport. Ifosfamide decreased the viability of cells expressing hOCT2, but not that of control cells. Coadministration of cimetidine, a known competitive substrate of hOCT2, completely prevented this ifosfamide-induced toxicity. Finally, ifosfamide but not cyclophosphamide depolarized proximal tubular cells. We propose that the nephrotoxicity of ifosfamide is due to its selective uptake by hOCT2 into renal proximal tubular cells, and that coadministration of cimetidine may be used to prevent ifosfamide-induced nephrotoxicity.


Assuntos
Ifosfamida/farmacocinética , Rim/efeitos dos fármacos , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Cimetidina/farmacocinética , Cimetidina/uso terapêutico , Feminino , Humanos , Ifosfamida/uso terapêutico , Técnicas In Vitro , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Modelos Biológicos , Proteínas de Transporte de Cátions Orgânicos/genética , Transportador 1 de Cátions Orgânicos/genética , Transportador 1 de Cátions Orgânicos/metabolismo , Transportador 2 de Cátion Orgânico
20.
J Hepatol ; 54(3): 513-20, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21093948

RESUMO

BACKGROUND & AIMS: Hyperoxaluria is a major problem causing nephrolithiasis. Little is known about the regulation of oxalate transport from the liver, the main organ for oxalate synthesis, into the circulation. Since the sulfate anion transporter-1(sat-1) is present in the sinusoidal membrane of hepatocytes and translocates oxalate, its impact on increased oxalate synthesis was studied. METHODS: Sat-1 expressing oocytes were used for cis-inhibition, trans-stimulation, and efflux experiments with labelled sulfate and oxalate to demonstrate the interactions of oxalate, glyoxylate, and glycolate with sat-1. HepG2 cells were incubated with oxalate and its precursors (glycine, hydroxyproline, glyoxylate, and glycolate). Changes in endogenous sat-1 mRNA-expression were examined using real-time PCR. After incubation of HepG2 cells in glyoxylate, sat-1 protein-expression was analysed by Western blotting, and sulfate uptake into HepG2 cells was measured. RT-PCR was used to screen for mRNA of other transporters. RESULTS: While oxalate and glyoxylate inhibited sulfate uptake, glycolate did not. Sulfate and oxalate uptake were trans-stimulated by glyoxylate but not by glycolate. Glyoxylate enhanced sulfate efflux. Glyoxylate was the only oxalate precursor stimulating sat-1 mRNA-expression. After incubation of HepG2 cells in glyoxylate, both sat-1 protein-expression and sulfate uptake into the cells increased. mRNA-expression of other transporters in HepG2 cells was not affected by glyoxylate treatment. CONCLUSIONS: The oxalate precursor glyoxylate was identified as a substrate of sat-1. Upregulated expression of sat-1 mRNA and of a functional sat-1 protein indicates that glyoxylate may be responsible for the elevated oxalate release from hepatocytes observed in hyperoxaluria.


Assuntos
Proteínas de Transporte de Ânions/genética , Proteínas de Transporte de Ânions/metabolismo , Glioxilatos/metabolismo , Animais , Sequência de Bases , Primers do DNA/genética , Feminino , Glicolatos/metabolismo , Glicolatos/farmacologia , Glioxilatos/farmacologia , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hiperoxalúria/complicações , Hiperoxalúria/metabolismo , Técnicas In Vitro , Modelos Biológicos , Nefrolitíase/etiologia , Nefrolitíase/metabolismo , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Oxalatos/metabolismo , Oxalatos/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transportadores de Sulfato , Sulfatos/metabolismo , Regulação para Cima/efeitos dos fármacos , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA