Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 309(10): F843-51, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26377792

ABSTRACT

Phylogentically, organic anion transporter (OAT)1 and OAT3 are closely related, whereas OAT2 is more distant. Experiments with human embryonic kidney-293 cells stably transfected with human OAT1, OAT2, or OAT3 were performed to compare selected transport properties. Common to OAT1, OAT2, and OAT3 is their ability to transport cGMP. OAT2 interacted with prostaglandins, and cGMP uptake was inhibited by PGE2 and PGF2α with IC50 values of 40.8 and 12.7 µM, respectively. OAT1 (IC50: 23.7 µM), OAT2 (IC50: 9.5 µM), and OAT3 (IC50: 1.6 µM) were potently inhibited by MK571, an established multidrug resistance protein inhibitor. OAT2-mediated cGMP uptake was not inhibited by short-chain monocarboxylates and, as opposed to OAT1 and OAT3, not by dicarboxylates. Consequently, OAT2 showed no cGMP/glutarate exchange. OAT1 and OAT3 exhibited a pH and a Cl- dependence with higher substrate uptake at acidic pH and lower substrate uptake in the absence of Cl-, respectively. Such pH and Cl- dependencies were not observed with OAT2. Depolarization of membrane potential by high K+ concentrations in the presence of the K+ ionophore valinomycin left cGMP uptake unaffected. In addition to cGMP, OAT2 transported urate and glutamate, but cGMP/glutamate exchange could not be demonstrated. These experiments suggest that OAT2-mediated cGMP uptake does not occur via exchange with monocarboxylates, dicarboxylates, and hydroxyl ions. The counter anion for electroneutral cGMP uptake remains to be identified.


Subject(s)
Biological Transport/physiology , Kidney/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Uric Acid/metabolism , Anions/metabolism , HEK293 Cells , Humans , Organic Anion Transport Protein 1/metabolism
2.
Nephron Physiol ; 124(1-2): 1-5, 2013.
Article in English | MEDLINE | ID: mdl-24247155

ABSTRACT

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.


Subject(s)
Glutathione/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Succinic Acid/metabolism , Symporters/metabolism , Animals , Humans , Kidney Tubules, Proximal/metabolism , Liver/metabolism , Oocytes/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , RNA, Complementary/genetics , Symporters/genetics , Xenopus laevis/genetics
3.
Pflugers Arch ; 464(4): 367-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875277

ABSTRACT

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.


Subject(s)
Dioxygenases/antagonists & inhibitors , Ketoglutaric Acids/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Amino Acids, Dicarboxylic/metabolism , Biological Transport, Active , Estrone/analogs & derivatives , Estrone/metabolism , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1/metabolism , Organic Anion Transporters/drug effects , Organic Anion Transporters, Sodium-Dependent/drug effects , Organic Anion Transporters, Sodium-Dependent/metabolism , Pyridines/metabolism , Succinic Acid/metabolism , Symporters/drug effects , Symporters/metabolism , p-Aminohippuric Acid/metabolism
4.
Am J Physiol Renal Physiol ; 301(5): F1026-34, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21865262

ABSTRACT

Organic anions are taken up from the blood into proximal tubule cells by organic anion transporters 1 and 3 (OAT1 and OAT3) in exchange for dicarboxylates. The released dicarboxylates are recycled by the sodium dicarboxylate cotransporter 3 (NaDC3). In this study, we tested the substrate specificities of human NaDC3, OAT1, and OAT3 to identify those dicarboxylates for which the three cooperating transporters have common high affinities. All transporters were stably expressed in HEK293 cells, and extracellularly added dicarboxylates were used as inhibitors of [(14)C]succinate (NaDC3), p-[(3)H]aminohippurate (OAT1), or [(3)H]estrone-3-sulfate (OAT3) uptake. Human NaDC3 was stably expressed as proven by immunochemical methods and by sodium-dependent uptake of succinate (K(0.5) for sodium activation, 44.6 mM; Hill coefficient, 2.1; K(m) for succinate, 18 µM). NaDC3 was best inhibited by succinate (IC(50) 25.5 µM) and less by α-ketoglutarate (IC(50) 69.2 µM) and fumarate (IC(50) 95.2 µM). Dicarboxylates with longer carbon backbones (adipate, pimelate, suberate) had low or no affinity for NaDC3. OAT1 exhibited the highest affinity for glutarate, α-ketoglutarate, and adipate (IC(50) between 3.3 and 6.2 µM), followed by pimelate (18.6 µM) and suberate (19.3 µM). The affinity of OAT1 to succinate and fumarate was low. OAT3 showed the same dicarboxylate selectivity with ∼13-fold higher IC(50) values compared with OAT1. The data 1) reveal α-ketoglutarate as a common high-affinity substrate of NaDC3, OAT1, and OAT3 and 2) suggest potentially similar molecular structures of the binding sites in OAT1 and OAT3 for dicarboxylates.


Subject(s)
Dicarboxylic Acid Transporters/metabolism , Dicarboxylic Acids/metabolism , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Symporters/metabolism , Amino Acid Sequence , Binding Sites , Blotting, Western , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acids/chemistry , Electrophoresis, Polyacrylamide Gel , Estrone/pharmacology , HEK293 Cells , Humans , Immunohistochemistry , Ketoglutaric Acids/metabolism , Molecular Sequence Data , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Independent/genetics , Structure-Activity Relationship , Succinates/metabolism , Symporters/genetics , Transfection , p-Aminohippuric Acid/metabolism
5.
Am J Physiol Renal Physiol ; 297(1): F145-54, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19369292

ABSTRACT

Tubular reabsorption of sulfate is achieved by the sodium-dependent sulfate transporter, NaSi-1, located at the apical membrane, and the sulfate-anion exchanger, sat-1, located at the basolateral membrane. To delineate the physiological role of rat sat-1, [(35)S]sulfate and [(14)C]oxalate uptake into sat-1-expressing oocytes was determined under various experimental conditions. Influx of [(35)S]sulfate was inhibited by bicarbonate, thiosulfate, sulfite, and oxalate, but not by sulfamate and sulfide, in a competitive manner with K(i) values of 2.7 +/- 1.3 mM, 101.7 +/- 9.7 microM, 53.8 +/- 10.9 microM, and 63.5 +/- 38.7 microM, respectively. Vice versa, [(14)C]oxalate uptake was inhibited by sulfate with a K(i) of 85.9 +/- 9.5 microM. The competitive type of inhibition indicates that these compounds are most likely substrates of sat-1. Physiological plasma bicarbonate concentrations (25 mM) reduced sulfate and oxalate uptake by more than 75%. Simultaneous application of sulfate, bicarbonate, and oxalate abolished sulfate as well as oxalate uptake. These data and electrophysiological studies using a two-electrode voltage-clamp device provide evidence that sat-1 preferentially works as an electroneutral sulfate-bicarbonate or oxalate-bicarbonate exchanger. In kidney proximal tubule cells, sat-1 likely completes sulfate reabsorption from the ultrafiltrate across the basolateral membrane in exchange for bicarbonate. In hepatocytes, oxalate extrusion is most probably mediated either by an exchange for sulfate or bicarbonate.


Subject(s)
Amino Acid Transport System A/metabolism , Bicarbonates/pharmacokinetics , Oocytes/metabolism , Oxalates/pharmacokinetics , Sulfates/pharmacokinetics , Amino Acid Transport System A/genetics , Animals , Biological Transport/physiology , Female , Models, Biological , Oocytes/cytology , Patch-Clamp Techniques , Rats , Transfection , Xenopus laevis
6.
Pflugers Arch ; 457(6): 1381-92, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19002488

ABSTRACT

The sulfate anion transporter (sat-1, Slc26a1) has been cloned from rat liver, functionally characterized, and localized to the sinusoidal membrane in hepatocytes and basolateral membrane (BLM) in proximal tubules (PT). Here, we confirm previously described localization of sat-1 protein in rat liver and kidneys and report on gender differences (GD) in its expression by immunochemical, transport, and excretion studies in rats. The approximately 85-kDa sat-1 protein was localized to the sinusoidal membrane in hepatocytes and BLM in renal cortical PT, with the male-dominant expression. However, the real-time reverse-transcription polymerase chain reaction data indicated no GD at the level of sat-1 mRNA. In agreement with the protein data, isolated membranes from both organs exhibited the male-dominant exchange of radiolabeled sulfate for oxalate, whereas higher oxalate in plasma and 24-h urine indicated higher oxalate production and excretion in male rats. Furthermore, the expression of liver, but not renal, sat-1 protein was: unaffected by castration, upregulated by ovariectomy, and downregulated by estrogen or progesterone treatment in males. Therefore, GD (males > females) in the expression of sat-1 protein in rat liver (and, possibly, kidneys) are caused by the female sex-hormone-driven inhibition at the posttranscriptional level. The male-dominant abundance of sat-1 protein in liver may conform to elevated uptake of sulfate and extrusion of oxalate, causing higher plasma oxalate in males. Oxalate is then excreted by the kidneys via the basolateral sat-1 (males > females) and the apical CFEX (Slc26a6; GD unknown) in PT and eliminated in the urine (males > females), where it may contribute to the male-prevailing development of oxalate urolithiasis.


Subject(s)
Anion Transport Proteins/biosynthesis , Antiporters/biosynthesis , Kidney/metabolism , Liver/metabolism , Animals , Castration , Cell Membrane/metabolism , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Gene Expression , Immunohistochemistry , Liver/drug effects , Male , Oxalates/urine , Progesterone/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors , Sulfate Transporters , Sulfates/metabolism
7.
J Biol Chem ; 283(24): 16332-41, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18411268

ABSTRACT

The orphan transporter hORCTL3 (human organic cation transporter like 3; SLC22A13) is highly expressed in kidneys and to a weaker extent in brain, heart, and intestine. hORCTL3-expressing Xenopus laevis oocytes showed uptake of [(3)H]nicotinate, [(3)H]p-aminohippurate, and [(14)C]urate. Hence, hORCTL3 is an organic anion transporter, and we renamed it hOAT10. [(3)H]Nicotinate transport by hOAT10 into X. laevis oocytes and into Caco-2 cells was saturable with Michaelis constants (K(m)) of 22 and 44 microm, respectively, suggesting that hOAT10 may be the molecular equivalent of the postulated high affinity nicotinate transporter in kidneys and intestine. The pH dependence of hOAT10 suggests p-aminohippurate(-)/OH(-), urate(-)/OH(-), and nicotinate(-)/OH(-) exchange as possible transport modes. Urate inhibited [(3)H]nicotinate transport by hOAT10 with an IC(50) value of 759 microm, assuming that hOAT10 represents a low affinity urate transporter. hOAT10-mediated [(14)C]urate uptake was elevated by an exchange with l -lactate, pyrazinoate, and nicotinate. Surprisingly, we have detected urate(-)/glutathione exchange by hOAT10, consistent with an involvement of hOAT10 in the renal glutathione cycle. Uricosurics, diuretics, and cyclosporine A showed substantial interactions with hOAT10, of which cyclosporine A enhanced [(14)C]urate uptake, providing the first molecular evidence for cyclosporine A-induced hyperuricemia.


Subject(s)
Niacin/metabolism , Organic Anion Transporters/physiology , Amino Acid Sequence , Animals , Caco-2 Cells , Female , Humans , Male , Models, Biological , Molecular Sequence Data , Niacin/chemistry , Oocytes/metabolism , Organic Anion Transporters/chemistry , Organic Anion Transporters/metabolism , Rats , Rats, Wistar , Tissue Distribution , Xenopus laevis
8.
Pflugers Arch ; 457(1): 223-31, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18365245

ABSTRACT

Glutaric acidurias are rare inherited neurodegenerative disorders accompanied by accumulation of the metabolites glutarate (GA) and 3-hydroxyglutarate (3OHGA), glutaconate, L: -, or D: -2-hydroxyglutarate (L: -2OHGA, D: -2OHGA) in all body fluids. Oocytes expressing the human (h) sodium-dicarboxylate cotransporter (NaDC3) showed sodium-dependent inward currents mediated by GA, 3OHGA, L: -, and D: -2OHGA. The organic anion transporters (OATs) were examined as additional transporters for GA derivatives. The uptake of [(3)H]p-aminohippurate in hOAT1-transfected human embryonic kidney (HEK293) cells was inhibited by GA, 3OHGA, D: -, or L: -2OHGA in a concentration-dependent manner. None of these compounds affected the hOAT3-mediated uptake of [(3)H]estrone sulfate (ES). In hOAT4-expressing cells and oocytes, ES uptake was strongly increased by intracellular GA derivatives. The data provide a model for the concerted action of OAT1 and NaDC3 mediating the basolateral uptake, and OAT4 mediating apical secretion of GA derivatives from proximal tubule cells and therefore contribute to the renal clearance of these compounds.


Subject(s)
Glutarates/metabolism , Glutarates/urine , Organic Anion Transport Protein 1/physiology , Organic Anion Transporters, Sodium-Independent/physiology , Animals , Cell Line , Dicarboxylic Acid Transporters/metabolism , Estrone/physiology , Female , Glutarates/pharmacology , Humans , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Kinetics , Oocytes/drug effects , Oocytes/metabolism , Organic Anion Transport Protein 1/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , RNA, Complementary/biosynthesis , RNA, Complementary/genetics , Sodium Channels/drug effects , Sodium Channels/metabolism , Sodium Channels/physiology , Substrate Specificity/physiology , Symporters/metabolism , Transfection , Xenopus laevis , p-Aminohippuric Acid/metabolism
9.
J Biol Chem ; 282(18): 13402-9, 2007 May 04.
Article in English | MEDLINE | ID: mdl-17353191

ABSTRACT

Organic anion transporter 1 (OAT1) is key for the secretion of organic anions in renal proximal tubules. These organic anions comprise endogenous as well as exogenous compounds including frequently used drugs of various chemical structures. The molecular basis for the polyspecificity of OAT1 is not known. Here we mutated a conserved positively charged arginine residue (Arg(466)) in the 11(th) transmembrane helix of human OAT1. The replacement by the positively charged lysine (R466K) did not impair expression of hOAT1 at the plasma membrane of Xenopus laevis oocytes but decreased the transport of p-aminohippurate (PAH) considerably. Extracellular glutarate inhibited and intracellular glutarate trans-stimulated wild type and mutated OAT1, suggesting for the mutant R466K an unimpaired interaction with dicarboxylates. However, when Arg(466) was replaced by the negatively charged aspartate (R466D), glutarate no longer interacted with the mutant. PAH uptake by wild type hOAT1 was stimulated in the presence of chloride, whereas the R466K mutant was chloride-insensitive. Likewise, the uptake of labeled glutarate or ochratoxin A was chloride-dependent in the wild type but not in R466K. Kinetic experiments revealed that chloride did not alter the apparent K(m) for PAH but influenced V(max) in wild type OAT1-expressing oocytes. In R466K mutants the apparent K(m) for PAH was similar to that of the wild type, but V(max) was not changed by chloride removal. We conclude that Arg(466) influences the binding of glutarate, but not interaction with PAH, and interacts with chloride, which is a major determinant in substrate translocation.


Subject(s)
Amino Acid Substitution , Chlorides/metabolism , Glutarates/metabolism , Mutation, Missense , Organic Anion Transport Protein 1/metabolism , Animals , Calcium Channel Blockers/pharmacology , Gene Expression , Humans , Ion Transport/drug effects , Ion Transport/genetics , Kinetics , Ochratoxins/pharmacology , Oocytes/metabolism , Organic Anion Transport Protein 1/genetics , Protein Structure, Secondary/genetics , Xenopus laevis
10.
Am J Physiol Gastrointest Liver Physiol ; 290(5): G1075-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16357056

ABSTRACT

Although the sulfate/anion transporter (sat-1; SLC26A1) was isolated from a rat liver cDNA library by expression cloning, localization of sat-1 within the liver and its contribution to the transport of sulfate and organo sulfates have remained unresolved. In situ hybridization and immunohistochemical studies were undertaken to demonstrate the localization of sat-1 in liver tissue. RT-PCR studies on isolated hepatocytes and liver endothelial and stellate cells in culture were performed to test for the presence of sat-1 in these cells. In sulfate uptake and efflux experiments, the substrate specificity of sat-1 was evaluated. Sat-1 mRNA was found in hepatocytes and endothelial cells. Sat-1 protein was localized in sinusoidal membranes and along the borders of hepatocytes. The canalicular region and bile capillaries were not stained. Sulfate uptake was only slightly affected by sulfamoyl diuretics or organo sulfates. Sulfate efflux from sat-1-expressing oocytes was enhanced in the presence of bicarbonate, indicating sulfate/bicarbonate exchange. Estrone sulfate was not transported by sat-1. Sat-1 may be responsible for the uptake of inorganic sulfate from the blood into hepatocytes to enable sulfation reactions. In hepatocytes and endothelial cells, sat-1 may also supply sulfate for proteoglycan synthesis.


Subject(s)
Amino Acid Transport System A/analysis , Liver/chemistry , Sulfates/metabolism , Amino Acid Transport System A/physiology , Animals , Bicarbonates/pharmacology , Dehydroepiandrosterone Sulfate/pharmacokinetics , Endothelial Cells/metabolism , Estrone/analogs & derivatives , Estrone/metabolism , Hepatocytes/metabolism , Liver/metabolism , Male , Rats , Unithiol/pharmacology
11.
Biochimie ; 87(5): 421-4, 2005 May.
Article in English | MEDLINE | ID: mdl-15820748

ABSTRACT

With the cloning of pig renal organic anion transporter 1 (pOAT1) (Biochimie 84 (2002) 1219) we set up a model system for comparative studies of cloned and natively isolated membrane located transport proteins. Meanwhile, another transport protein involved in p-aminohippurate (PAH) uptake on the basolateral side of the proximal tubule cells was identified, designated organic anion transporter 3 (OAT3). To explore the contribution of pOAT1 to the PAH clearance in comparison to OAT3, it was the aim of this study to extend our model by cloning of the pig ortholog of OAT3. Sequence comparisons of human organic anion transporter 3 (hOAT3) with the expressed sequence tag (EST) database revealed a clone and partial sequence of the pig renal organic anion transporter 3 (pOAT3) ortholog. Sequencing of the entire open reading frame resulted in a protein of 543 amino acid residues encoded by 1632 base pairs (EMBL Acc. No. AJ587003). It showed high homologies of 81%, 80%, 76%, and 77% to the human, rabbit, rat, and mouse OAT3, respectively. A functional characterization of pOAT3 in Xenopus laevis oocytes yielded an apparent Km (Kt) for [3H]estrone sulfate of 7.8 +/- 1.3 microM. Moreover, pOAT3 mediated [3H]estrone sulfate uptake was almost abolished by 0.5 mM of glutarate, dehydroepiandosterone sulfate, or probenecid consistent with the hallmarks of OAT3 function.


Subject(s)
Estrone/analogs & derivatives , Kidney/metabolism , Organic Anion Transporters, Sodium-Independent/genetics , Amino Acid Sequence , Animals , DNA, Complementary , Dehydroepiandrosterone Sulfate/pharmacology , Estrone/metabolism , Glutarates/pharmacology , Humans , Kidney/drug effects , Mice , Molecular Sequence Data , Oocytes/drug effects , Oocytes/physiology , Organic Anion Transporters, Sodium-Independent/metabolism , Probenecid/pharmacology , Rabbits , Rats , Sequence Homology, Amino Acid , Swine , Uricosuric Agents/pharmacology , Xenopus laevis/metabolism , p-Aminohippuric Acid/pharmacokinetics
12.
Am J Physiol Renal Physiol ; 284(3): F503-9, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12429554

ABSTRACT

The H(2)-receptor antagonist cimetidine is efficiently excreted by the kidneys. In vivo studies indicated an interaction of cimetidine not only with transporters for basolateral uptake of organic cations but also with those involved in excretion of organic anions. We therefore tested cimetidine as a possible substrate of the organic anion transporters cloned from winter flounder (fROAT) and from human kidney (hOAT1). Uptake of [(3)H]cimetidine into fROAT-expressing Xenopus laevis oocytes exceeded uptake into control oocytes. At -60-mV clamp potential, 1 mM cimetidine induced an inward current, which was smaller than that elicited by 0.1 mM PAH. Cimetidine concentrations exceeding 0.1 mM decreased PAH-induced inward currents, indicating interaction with the same transporter. At pH 6.6, no current was seen with 0.1 mM cimetidine, whereas at pH 8.6 a current was readily detectable, suggesting preferential translocation of uncharged cimetidine by fROAT. Oocytes expressing hOAT1 also showed [(3)H]cimetidine uptake. These data reveal cimetidine as a substrate for fROAT/hOAT1 and suggest that organic anion transporters contribute to cimetidine excretion in proximal tubules.


Subject(s)
Cimetidine/pharmacokinetics , Histamine H2 Antagonists/pharmacokinetics , Kidney/metabolism , Organic Anion Transport Protein 1/metabolism , Animals , Dose-Response Relationship, Drug , Flounder , Humans , Hydrogen-Ion Concentration , Membrane Potentials/drug effects , Membrane Potentials/physiology , Microinjections , Oocytes/metabolism , Organic Anion Transport Protein 1/genetics , Patch-Clamp Techniques , RNA, Complementary/metabolism , Xenopus laevis
13.
Biochimie ; 84(12): 1221-4, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12628298

ABSTRACT

A pig kidney cDNA library was screened for the porcine ortholog of the multispecific organic anion transporter 1 (pOAT1). Several positive clones were isolated resulting in two alternatively spliced cDNA clones of pOAT1 (pOAT1 and pOAT1A). pOAT1-cDNAs consist of 2126 or 1895 base pairs (EMBL Acc. No. AJ308234 and AJ308235) encoding 547 or 533 amino acid residue proteins with 89, 87, 83 and 81% homology to the human, rabbit, rat, and mouse OAT1, respectively. Heterologous expression of pOAT1 in Xenopus laevis oocytes revealed an apparent K(m) for [3H]PAH of 3.75 +/- 1.6 microM. [3H]PAH uptake mediated by pOAT1 was abolished by 0.5 mM glutarate or 1 mM probenecid. Functional characterization of pOAT1A did not show any affinity for [3H]PAH. In summary, we cloned two alternative splice variants of the pig ortholog of organic anion transporter 1. One splice form (pOAT1) showed typical functional characteristics of organic anion transporter 1, whereas the second form appears not to transport PAH.


Subject(s)
Kidney/metabolism , Organic Anion Transport Protein 1/genetics , Swine/genetics , Alternative Splicing , Amino Acid Sequence , Animals , Anions/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Conserved Sequence/genetics , DNA, Complementary/chemistry , DNA, Complementary/genetics , Gene Library , Kinetics , Molecular Sequence Data , Oocytes/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Xenopus laevis , p-Aminohippuric Acid/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...