Comprehensive Physiology Wiley Online Library

Renal Transport of Organic Anions and Cations

Full Article on Wiley Online Library



Abstract

Organic anions and cations (OAs and OCs, respectively) comprise an extraordinarily diverse array of compounds of physiological, pharmacological, and toxicological importance. The kidney, primarily the renal proximal tubule, plays a critical role in regulating the plasma concentrations of these organic electrolytes and in clearing the body of potentially toxic xenobiotics agents, a process that involves active, transepithelial secretion. This transepithelial transport involves separate entry and exit steps at the basolateral and luminal aspects of renal tubular cells. Basolateral and luminal OA and OC transport reflects the concerted activity of a suite of separate proteins arranged in parallel in each pole of proximal tubule cells. The cloning of multiple members of several distinct transport families, the subsequent characterization of their activity, and their subcellular localization within distinct regions of the kidney, now allows the development of models describing the molecular basis of the renal secretion of OAs and OCs. New information on naturally occurring genetic variation of many of these processes provides insight into the basis of observed variability of drug efficacy and unwanted drug‐drug interactions in human populations. The present review examines recent work on these issues. © 2011 American Physiological Society. Compr Physiol 1:1795‐1835, 2011.

Comprehensive Physiology offers downloadable PowerPoint presentations of figures for non-profit, educational use, provided the content is not modified and full credit is given to the author and publication.

Download a PowerPoint presentation of all images


Figure 1. Figure 1.

Schematic model of the transport processes involved in secretion of organic anions (OAs) by renal proximal tubule cells. The direction of the arrows indicates the direction of net substrate transport. The uptake of OAs across the basolateral membrane by organic anion transporter 1 and 3 (OAT1 and OAT3) occurs via a tertiary active mechanism where (i) Na+,K+‐ATPase creates the sodium gradient that (ii) drives Na+‐dependent αKG2− (Kreb's cycle intermediate) uptake by Na+‐dicarboxylate cotransporter 1 (NaDC1) and NaDC3, and in turn, (iii) the outwardly directed αKG2− gradient energizes OA uptake by OAT1 and OAT3. Although organic anion transporter 2 is suspected of mediating OA uptake, the intracellular counteranion involved in the exchange process has not been clearly established. Na+‐phosphate cotransporter 1 and 4 (NPT1 and NPT4) operate via electrogenic facilitated diffusion, and multidrug resistance‐associated proteins 2 and 4 are ATPases. URAT1 is most noted for its involvement in urate reabsorption via exchange with intracellular monocarboxylates (MC). The outwardly directed monocarboxylate gradient is maintained by Na+‐dependent monocarboxylate transporters 1 and 2 (SMCT1 and 2). The energetic mechanism of organic anion transporter 4 and 10 (OAT4 and OAT10) has not been clearly established, and thus, it is not clear if they would predominate in OA secretion or reabsorption. However, OA transport by OAT4 is trans‐stimulated by αKG2− and OAT10 by monocarboxylates; the outwardly directed mono‐ and di‐carboxylate gradients may drive OA reabsorption by these transporters, as depicted in the figure.

Figure 2. Figure 2.

Schematic model of the transport processes involved in the secretion or reabsorption of urate by renal proximal tubule (RPT) cells. The direction of the arrows indicates the direction of net substrate transport. Secretion: Urate uptake across the basolateral membrane of RPT cells by organic anion transporter 1 and 3 is driven by the outwardly directed αKG2− gradient, which is maintained by Na+‐dicarboxylate cotransporter 1 (NaDC1) and NaDC3. Although organic anion transporter 2 is suspected of mediating urate uptake, the intracellular counteranion involved in the exchange process has not been clearly established. Na+‐phosphate cotransporter 1 (NPT1), NPT4, and glucose transporter 9 (GLUT9) operate via electrogenic facilitated diffusion, whereas multidrug resistance‐associated proteins 4 is an ATPase. Reabsorption: URAT1 is an exchanger suspected of playing a major role in the uptake of urate across the luminal membrane of RPT cells. The outwardly directed monocarboxylate (MC) gradient, maintained by the Na+‐dependent monocarboxylate transporters 1 and 2 (SMCT1 and 2), is suspected of providing the energetic basis for urate uptake by urate transporter 1. The energetic mechanism of organic anion transporter 4 and 10 (OAT4 and OAT10) has not been clearly established, and thus, it is not clear if they would predominate in urate secretion or reabsorption. However, OA transport by OAT4 is trans‐stimulated by αKG2− and OAT10 by monocarboxylates; the outwardly directed mono‐ and di‐carboxylate gradients may drive urate reabsorption by these transporters, as depicted in the figure. GLUT9 operates via electrogenic facilitated diffusion, and is suspected of mediating urate efflux across the basolateral membrane.

Figure 3. Figure 3.

(A) Uptake of [3H]ochratoxin A (OTA) by rabbit organic anion transporter 1 (rbOAT1) and rabbit organic anion transporter 3 (rbOAT3) stably expressed in Chinese hamster ovary cells. OTA uptake was expressed as a percentage of the uptake into Chinese hamster ovary cells not expressing the respective transporters (Control). OTA uptake was conducted in the absence or presence of the OAT1 inhibitor para‐aminohippurate (PAH; 2.5 mM) or the OAT3 inhibitor estrone‐3‐sulfate (ES; 1 mM). (B) Effect of PAH and ES either alone or in combination on OTA uptake into freshly isolated rabbit renal proximal tubules in suspension. For comparison, OTA uptake was also conducted in the presence of the dual OAT1 and OAT3 inhibitor, probenecid (Prob).

Modified from 468.
Figure 4. Figure 4.

Shown are two views of a three‐dimensional homology model of human organic anion transporter 1 structure. The model was built using the crystal structure of GlpT (bacterial glycerol‐3‐phosphate/Pi exchanger) as a template 287. (A) View of the model from the cytoplasmic face. (B) Side view of the homology model, with transmembrane spanning helices 2 and 4 omitted to highlight key residues involved in substrate binding. Key residues in substrate binding (orange = Y230; yellow = K431, purple = F438, and red = R466) are exposed to the hydrophilic cleft that is presumed to comprise the translocation pathway in these proteins.

Figure 5. Figure 5.

Model of the regulatory cascade involved in the acute stimulation of OA secretion, presumably through increased expression of organic anion transporter 1 and 3 (OAT1 and OAT3) at the basolateral membranes of renal proximal tubule cells. The arrows indicate the sequence of events beginning with epidermal growth factor (EGF) or insulin binding to their receptors. MEK = mitogen‐activated/extracellular signal‐regulated kinase; ERK1/2 = extracellular signal‐regulated kinase isoforms 1 and 2; PLA2 = phospholipase A2; AA = arachidonic acid; COX1 = cyclooxygenase 1; PGE2 = prostaglandin E2; AC = adenylate cyclase. The dashed arrow indicates trafficking of OAT1 and OAT3 to the basolateral membrane.

Figure 6. Figure 6.

Topology of short and long multidrug resistance‐associated proteins (MRP). All MRPs consist of the core functional unit, multispanning domains (MSD) 1 and 2, Walker A and Walker B motifs, and intracellular nucleotide‐binding domains (NBD) 1 and 2. Long MRPs, such as MRP2, contain a third MSD (MSD0) that is linked to the core functional unit by a cytoplasmic loop (CL3). Topologies predicted by different computer‐assisted algorithms differ, with some favoring MSD comprising four versus six transmembrane spanning helices.

Modified from 94.
Figure 7. Figure 7.

Dependence of substrate concentration on cGMP uptake by multidrug resistance‐associated proteins 4 and estradiol 17β‐D‐glucuronide (E217βG) uptake by multidrug resistance‐associated proteins 2 into membrane vesicles prepared from Sf9 cells that heterologously expressed the transport proteins. In both cases, substrate uptake was sigmoidal with concentration, indicating positive cooperativity.

Data were adapted from 460 and 417.
Figure 8. Figure 8.

Schematic model of the transport processes associated with the secretion of organic cations (OCs) by renal proximal tubule cells. Circles indicate carrier‐mediated transport processes. Arrows indicate the direction of net substrate transport that occurs under physiological conditions. Solid lines depict what are believed to be principal pathways of substrate transport; dotted lines indicate pathways that are believed to be of secondary importance; the dashed line indicates diffusive movement. Following is a brief description of each of the numbered processes currently believed to play a role, direct or indirect, in transepithelial OC secretion. Principal basolateral processes include (i) Na,K‐ATPase; maintains the inwardly directed Na+ gradient and the K+ gradient associated with the inside‐negative membrane potential, both of which represent driving forces associated with active OC secretion. (ii) OCT1, OCT2, and OCT3; support electrogenic uniport (facilitated diffusion) from the blood of type I OCs (these processes are also believed to support electroneutral OC/OC exchange). Note: basolateral OCT expression (both amount and location) is species dependent; for example, in the human it appears that expression of OCT1 is restricted to the luminal membrane (see text). (iii) Diffusive flux of type II OCs. Principal apical transport processes include: (i) MDR1; supports the ATP‐dependent, active luminal export of type II OCs. (ii) NHE3 and NHE8; support the Na+/H+ exchange activity that sustains the inwardly directed electrochemical gradient for H+ that, in turn, supports activity of multidrug and toxin extrusion (MATE) transporters. (iii) MATE1 and MATE2‐K, support the mediated electroneutral exchange of luminal H+ for intracellular type I OCs. (iv) OCT1 (in human kidney); suspected to support reabsorption of selected OCs; (v) OCTN1 and OCTN2; support electrogenic Na‐dependent reabsorptive fluxes of ergothioneine and carnitine, respectively (secondary, postulated, role in the reabsorptive flux of selected OCs). (vi) CTL1?; postulated to support electrogenic choline reabsorption.

Figure 9. Figure 9.

Influence of MATE1 on the renal handling of metformin. (A) Plasma concentration profile of metformin in Mate1 (+/+) (○) and Mate1 (−/−) (•) mice. Metformin and mannitol were administered as a bolus via the jugular vein *w/1% mannitol administered to maintain a sufficient and constant urine flow. Blood samples were collected at the time points indicated. Each point represents the mean ± S.D. for six mice of each genotype. *P <0.05; ***P <0.001, significantly different from Mate1 (+/+) mice. (B) Urinary excretion of metformin in Mate1 (+/+) (open column) and Mate1 (−/−) (closed column) mice. Urine was collected for 60 min after the drug administration. Each column represents the mean ± S.D. for six mice. ***P <0.001, significantly different from Mate1 (+/+) mice. (C) Tissue distribution (a, b) and tissue clearance (c, d) of metformin in Mate1 (+/+) (□) and Mate1 (−/−) (▪) mice.

Tissue concentrations of metformin were determined 60 min after the drug administration. Each column represents the mean ± S.D. for six mice. **P <0.01; ***P <0.001, significantly different from Mate1 (+/+) mice.

[Modified, with permission, from 397, in which experimental details are outlined.]
Figure 10. Figure 10.

Mean plasma concentration‐time profiles for procainamide (□○) and N‐acetylprocainamide (▪•) when given alone (▪□) and when co‐administered orally with 150 mg ranitidine (•○) in six subjects.

[Modified from 356.]
Figure 11. Figure 11.

rOCT2‐mediated inward current and current under conditions of organic cation (OC) exchange in rOCT2‐expressing Xenopus oocytes. (A) The patch pipette (in detached patch mode) was filled with solution containing 20 mM choline, and the bath was superfused with 2 or 20 mM choline, 2 mM tetraethylammonium (TEA), 100 μM quinine, or no substrate, and voltage pulses from −60 to 60 mV were applied after complete solution exchange (indicated by lowercase letters). Under “symmetric” conditions, that is, 20 mM choline in pipette and bath (see a and g), zero net current was observed (indicated by broken line). Complete removal of substrate from the bath lead to an inward‐directed current (downward, see b and e). Superfusion with 2 mM choline (f) lead to a reduced inward current. Addition of 100 μM quinine (c) or 2 mm TEA (h) inhibited the inward current almost completely. (B) An analogous experiment with 2 mM choline in the pipette. The bath was superfused with 1, 2, 5, or 20 mM choline, 100 μM quinine, or no substrate. rOCT2‐mediated currents are seen below (inward directed) and above (outward‐directed) the broken line. Lowercase letters indicate application of voltage pulses.

[Modified, with permission, from 47].
Figure 12. Figure 12.

Charge distribution within the outward‐ and inward‐facing cleft of structure models of rOCT2. (A) Surface representation of rOCT2 with the substrate binding cleft in the outward‐facing (left) and inward‐facing conformation with superimposed contour grid showing isopotential surfaces at (negative, red; positive, blue). (B) Distribution of electrical charges (surface potential map) on the surface of rOCT2 wild‐type and the rOCT2 (E448Q) mutant (site of mutation is indicated by an arrow). In the outward‐facing conformations, negative potentials are shown in red and positive potentials in blue, viewed from the outside. (C) Electrostatic potential mapped onto the surface of rOCT2 wild‐type and rOCT2 (E448Q) mutant (site of mutation is indicated by an arrow) in the inward‐facing conformation; presentation and color coding are as in B. The negative charges within the outward‐facing innermost cavity may “trap” small inorganic cations and translocate them together with the organic cation substrate, whereas the reorientation may be electroneutral (nonselective‐cotransport‐of‐small‐cations hypothesis).

[Modified from 333].
Figure 13. Figure 13.

Structural alignment of 10 most potent OCT2 inhibitors (left) and the pharmacophore model (right), in which the blue sphere represents a positive ion interaction site and the red sphere represents a hydrophobic aromatic interaction site.

[Modified, with permission, from 476].
Figure 14. Figure 14.

Homology models of OCT 3D structure. The model of rOCT1 292 used the crystal structure of LacY (bacterial sugar‐H+ cotransporter) as its template, whereas the model of rbOCT2 469 used the crystal structure of GlpT (bacterial glycerol‐3‐phosphate/Pi exchanger). Both models are viewed from the cytoplasmic aspect of the proteins. In both models, key residues in the binding of substrates (Q448 and D475 in rOCT1 and their OCT2 homologs, E447 and D474, in rbOCT2) are exposed to the hydrophilic cleft between the N‐ and C‐terminal halves that is presumed to comprise the translocation pathway in these proteins.

Figure 15. Figure 15.

(A) hMATE1 model showing side view (top) and the extracellular face (bottom). The principal cleft helices (1, 2, 7, and 8) are labeled in the bottom view. (B) Topological comparison (from the cytoplasmic aspect) of a representative MATE family member (NorM; left) and a representative MFS member (EmrD; right). Below the simplified topology diagram are the x‐ray structures with the TM helices (1‐12 marked).



Figure 1.

Schematic model of the transport processes involved in secretion of organic anions (OAs) by renal proximal tubule cells. The direction of the arrows indicates the direction of net substrate transport. The uptake of OAs across the basolateral membrane by organic anion transporter 1 and 3 (OAT1 and OAT3) occurs via a tertiary active mechanism where (i) Na+,K+‐ATPase creates the sodium gradient that (ii) drives Na+‐dependent αKG2− (Kreb's cycle intermediate) uptake by Na+‐dicarboxylate cotransporter 1 (NaDC1) and NaDC3, and in turn, (iii) the outwardly directed αKG2− gradient energizes OA uptake by OAT1 and OAT3. Although organic anion transporter 2 is suspected of mediating OA uptake, the intracellular counteranion involved in the exchange process has not been clearly established. Na+‐phosphate cotransporter 1 and 4 (NPT1 and NPT4) operate via electrogenic facilitated diffusion, and multidrug resistance‐associated proteins 2 and 4 are ATPases. URAT1 is most noted for its involvement in urate reabsorption via exchange with intracellular monocarboxylates (MC). The outwardly directed monocarboxylate gradient is maintained by Na+‐dependent monocarboxylate transporters 1 and 2 (SMCT1 and 2). The energetic mechanism of organic anion transporter 4 and 10 (OAT4 and OAT10) has not been clearly established, and thus, it is not clear if they would predominate in OA secretion or reabsorption. However, OA transport by OAT4 is trans‐stimulated by αKG2− and OAT10 by monocarboxylates; the outwardly directed mono‐ and di‐carboxylate gradients may drive OA reabsorption by these transporters, as depicted in the figure.



Figure 2.

Schematic model of the transport processes involved in the secretion or reabsorption of urate by renal proximal tubule (RPT) cells. The direction of the arrows indicates the direction of net substrate transport. Secretion: Urate uptake across the basolateral membrane of RPT cells by organic anion transporter 1 and 3 is driven by the outwardly directed αKG2− gradient, which is maintained by Na+‐dicarboxylate cotransporter 1 (NaDC1) and NaDC3. Although organic anion transporter 2 is suspected of mediating urate uptake, the intracellular counteranion involved in the exchange process has not been clearly established. Na+‐phosphate cotransporter 1 (NPT1), NPT4, and glucose transporter 9 (GLUT9) operate via electrogenic facilitated diffusion, whereas multidrug resistance‐associated proteins 4 is an ATPase. Reabsorption: URAT1 is an exchanger suspected of playing a major role in the uptake of urate across the luminal membrane of RPT cells. The outwardly directed monocarboxylate (MC) gradient, maintained by the Na+‐dependent monocarboxylate transporters 1 and 2 (SMCT1 and 2), is suspected of providing the energetic basis for urate uptake by urate transporter 1. The energetic mechanism of organic anion transporter 4 and 10 (OAT4 and OAT10) has not been clearly established, and thus, it is not clear if they would predominate in urate secretion or reabsorption. However, OA transport by OAT4 is trans‐stimulated by αKG2− and OAT10 by monocarboxylates; the outwardly directed mono‐ and di‐carboxylate gradients may drive urate reabsorption by these transporters, as depicted in the figure. GLUT9 operates via electrogenic facilitated diffusion, and is suspected of mediating urate efflux across the basolateral membrane.



Figure 3.

(A) Uptake of [3H]ochratoxin A (OTA) by rabbit organic anion transporter 1 (rbOAT1) and rabbit organic anion transporter 3 (rbOAT3) stably expressed in Chinese hamster ovary cells. OTA uptake was expressed as a percentage of the uptake into Chinese hamster ovary cells not expressing the respective transporters (Control). OTA uptake was conducted in the absence or presence of the OAT1 inhibitor para‐aminohippurate (PAH; 2.5 mM) or the OAT3 inhibitor estrone‐3‐sulfate (ES; 1 mM). (B) Effect of PAH and ES either alone or in combination on OTA uptake into freshly isolated rabbit renal proximal tubules in suspension. For comparison, OTA uptake was also conducted in the presence of the dual OAT1 and OAT3 inhibitor, probenecid (Prob).

Modified from 468.


Figure 4.

Shown are two views of a three‐dimensional homology model of human organic anion transporter 1 structure. The model was built using the crystal structure of GlpT (bacterial glycerol‐3‐phosphate/Pi exchanger) as a template 287. (A) View of the model from the cytoplasmic face. (B) Side view of the homology model, with transmembrane spanning helices 2 and 4 omitted to highlight key residues involved in substrate binding. Key residues in substrate binding (orange = Y230; yellow = K431, purple = F438, and red = R466) are exposed to the hydrophilic cleft that is presumed to comprise the translocation pathway in these proteins.



Figure 5.

Model of the regulatory cascade involved in the acute stimulation of OA secretion, presumably through increased expression of organic anion transporter 1 and 3 (OAT1 and OAT3) at the basolateral membranes of renal proximal tubule cells. The arrows indicate the sequence of events beginning with epidermal growth factor (EGF) or insulin binding to their receptors. MEK = mitogen‐activated/extracellular signal‐regulated kinase; ERK1/2 = extracellular signal‐regulated kinase isoforms 1 and 2; PLA2 = phospholipase A2; AA = arachidonic acid; COX1 = cyclooxygenase 1; PGE2 = prostaglandin E2; AC = adenylate cyclase. The dashed arrow indicates trafficking of OAT1 and OAT3 to the basolateral membrane.



Figure 6.

Topology of short and long multidrug resistance‐associated proteins (MRP). All MRPs consist of the core functional unit, multispanning domains (MSD) 1 and 2, Walker A and Walker B motifs, and intracellular nucleotide‐binding domains (NBD) 1 and 2. Long MRPs, such as MRP2, contain a third MSD (MSD0) that is linked to the core functional unit by a cytoplasmic loop (CL3). Topologies predicted by different computer‐assisted algorithms differ, with some favoring MSD comprising four versus six transmembrane spanning helices.

Modified from 94.


Figure 7.

Dependence of substrate concentration on cGMP uptake by multidrug resistance‐associated proteins 4 and estradiol 17β‐D‐glucuronide (E217βG) uptake by multidrug resistance‐associated proteins 2 into membrane vesicles prepared from Sf9 cells that heterologously expressed the transport proteins. In both cases, substrate uptake was sigmoidal with concentration, indicating positive cooperativity.

Data were adapted from 460 and 417.


Figure 8.

Schematic model of the transport processes associated with the secretion of organic cations (OCs) by renal proximal tubule cells. Circles indicate carrier‐mediated transport processes. Arrows indicate the direction of net substrate transport that occurs under physiological conditions. Solid lines depict what are believed to be principal pathways of substrate transport; dotted lines indicate pathways that are believed to be of secondary importance; the dashed line indicates diffusive movement. Following is a brief description of each of the numbered processes currently believed to play a role, direct or indirect, in transepithelial OC secretion. Principal basolateral processes include (i) Na,K‐ATPase; maintains the inwardly directed Na+ gradient and the K+ gradient associated with the inside‐negative membrane potential, both of which represent driving forces associated with active OC secretion. (ii) OCT1, OCT2, and OCT3; support electrogenic uniport (facilitated diffusion) from the blood of type I OCs (these processes are also believed to support electroneutral OC/OC exchange). Note: basolateral OCT expression (both amount and location) is species dependent; for example, in the human it appears that expression of OCT1 is restricted to the luminal membrane (see text). (iii) Diffusive flux of type II OCs. Principal apical transport processes include: (i) MDR1; supports the ATP‐dependent, active luminal export of type II OCs. (ii) NHE3 and NHE8; support the Na+/H+ exchange activity that sustains the inwardly directed electrochemical gradient for H+ that, in turn, supports activity of multidrug and toxin extrusion (MATE) transporters. (iii) MATE1 and MATE2‐K, support the mediated electroneutral exchange of luminal H+ for intracellular type I OCs. (iv) OCT1 (in human kidney); suspected to support reabsorption of selected OCs; (v) OCTN1 and OCTN2; support electrogenic Na‐dependent reabsorptive fluxes of ergothioneine and carnitine, respectively (secondary, postulated, role in the reabsorptive flux of selected OCs). (vi) CTL1?; postulated to support electrogenic choline reabsorption.



Figure 9.

Influence of MATE1 on the renal handling of metformin. (A) Plasma concentration profile of metformin in Mate1 (+/+) (○) and Mate1 (−/−) (•) mice. Metformin and mannitol were administered as a bolus via the jugular vein *w/1% mannitol administered to maintain a sufficient and constant urine flow. Blood samples were collected at the time points indicated. Each point represents the mean ± S.D. for six mice of each genotype. *P <0.05; ***P <0.001, significantly different from Mate1 (+/+) mice. (B) Urinary excretion of metformin in Mate1 (+/+) (open column) and Mate1 (−/−) (closed column) mice. Urine was collected for 60 min after the drug administration. Each column represents the mean ± S.D. for six mice. ***P <0.001, significantly different from Mate1 (+/+) mice. (C) Tissue distribution (a, b) and tissue clearance (c, d) of metformin in Mate1 (+/+) (□) and Mate1 (−/−) (▪) mice.

Tissue concentrations of metformin were determined 60 min after the drug administration. Each column represents the mean ± S.D. for six mice. **P <0.01; ***P <0.001, significantly different from Mate1 (+/+) mice.

[Modified, with permission, from 397, in which experimental details are outlined.]


Figure 10.

Mean plasma concentration‐time profiles for procainamide (□○) and N‐acetylprocainamide (▪•) when given alone (▪□) and when co‐administered orally with 150 mg ranitidine (•○) in six subjects.

[Modified from 356.]


Figure 11.

rOCT2‐mediated inward current and current under conditions of organic cation (OC) exchange in rOCT2‐expressing Xenopus oocytes. (A) The patch pipette (in detached patch mode) was filled with solution containing 20 mM choline, and the bath was superfused with 2 or 20 mM choline, 2 mM tetraethylammonium (TEA), 100 μM quinine, or no substrate, and voltage pulses from −60 to 60 mV were applied after complete solution exchange (indicated by lowercase letters). Under “symmetric” conditions, that is, 20 mM choline in pipette and bath (see a and g), zero net current was observed (indicated by broken line). Complete removal of substrate from the bath lead to an inward‐directed current (downward, see b and e). Superfusion with 2 mM choline (f) lead to a reduced inward current. Addition of 100 μM quinine (c) or 2 mm TEA (h) inhibited the inward current almost completely. (B) An analogous experiment with 2 mM choline in the pipette. The bath was superfused with 1, 2, 5, or 20 mM choline, 100 μM quinine, or no substrate. rOCT2‐mediated currents are seen below (inward directed) and above (outward‐directed) the broken line. Lowercase letters indicate application of voltage pulses.

[Modified, with permission, from 47].


Figure 12.

Charge distribution within the outward‐ and inward‐facing cleft of structure models of rOCT2. (A) Surface representation of rOCT2 with the substrate binding cleft in the outward‐facing (left) and inward‐facing conformation with superimposed contour grid showing isopotential surfaces at (negative, red; positive, blue). (B) Distribution of electrical charges (surface potential map) on the surface of rOCT2 wild‐type and the rOCT2 (E448Q) mutant (site of mutation is indicated by an arrow). In the outward‐facing conformations, negative potentials are shown in red and positive potentials in blue, viewed from the outside. (C) Electrostatic potential mapped onto the surface of rOCT2 wild‐type and rOCT2 (E448Q) mutant (site of mutation is indicated by an arrow) in the inward‐facing conformation; presentation and color coding are as in B. The negative charges within the outward‐facing innermost cavity may “trap” small inorganic cations and translocate them together with the organic cation substrate, whereas the reorientation may be electroneutral (nonselective‐cotransport‐of‐small‐cations hypothesis).

[Modified from 333].


Figure 13.

Structural alignment of 10 most potent OCT2 inhibitors (left) and the pharmacophore model (right), in which the blue sphere represents a positive ion interaction site and the red sphere represents a hydrophobic aromatic interaction site.

[Modified, with permission, from 476].


Figure 14.

Homology models of OCT 3D structure. The model of rOCT1 292 used the crystal structure of LacY (bacterial sugar‐H+ cotransporter) as its template, whereas the model of rbOCT2 469 used the crystal structure of GlpT (bacterial glycerol‐3‐phosphate/Pi exchanger). Both models are viewed from the cytoplasmic aspect of the proteins. In both models, key residues in the binding of substrates (Q448 and D475 in rOCT1 and their OCT2 homologs, E447 and D474, in rbOCT2) are exposed to the hydrophilic cleft between the N‐ and C‐terminal halves that is presumed to comprise the translocation pathway in these proteins.



Figure 15.

(A) hMATE1 model showing side view (top) and the extracellular face (bottom). The principal cleft helices (1, 2, 7, and 8) are labeled in the bottom view. (B) Topological comparison (from the cytoplasmic aspect) of a representative MATE family member (NorM; left) and a representative MFS member (EmrD; right). Below the simplified topology diagram are the x‐ray structures with the TM helices (1‐12 marked).

References
 1. Abla N, Chinn LW, Nakamura T, Liu L, Huang CC, Johns SJ, Kawamoto M, Stryke D, Taylor TR, Ferrin TE, Giacomini KM, Kroetz DL. The human multidrug resistance protein 4 (MRP4, ABCC4): Functional analysis of a highly polymorphic gene. J Pharmacol Exp Ther 325: 859‐868, 2008.
 2. Abramson J, Smirnova I, Kasho V, Verner G, Kaback HR, Iwata S. Structure and mechanism of the lactose permease of Escherichia coli. Science 301: 610‐615, 2003.
 3. Acara M. The kidney in regulation of plasma choline in the chicken. Am J Physiol 228: 645‐649, 1975.
 4. Acara M, Rennick B. Regulation of plasma choline by the renal tubule: Bidirectional transport of choline. Am J Physiol 225: 1123‐1128, 1973.
 5. Acara M, Roch‐Ramel F, Rennick B. Bidirectional renal tubular transport of free choline: A micropuncture study. Am J Physiol 236: F112‐F118, 1979.
 6. Ahlin G, Karlsson J, Pedersen JM, Gustavsson L, Larsson R, Matsson P, Norinder U, Bergstrom CA, Artursson P. Structural requirements for drug inhibition of the liver specific human organic cation transport protein. J Med Chem 51: 5932‐5942, 2008.
 7. Ahn SY, Eraly SA, Tsigelny I, Nigam SK. Interaction of organic cations with organic anion transporters. J Biol Chem 284: 31422‐31430, 2009.
 8. Ambudkar SV, Dey S, Hrycyna CA, Ramachandra M, Pastan I, Gottesman MM. Biochemical, cellular, and pharmacological aspects of the multidrug transporter. Annu Rev Pharmacol Toxicol 39: 361‐398, 1999.
 9. Anzai N, Ichida K, Jutabha P, Kimura T, Babu E, Jin CJ, Srivastava S, Kitamura K, Hisatome I, Endou H, Sakurai H. Plasma urate level is directly regulated by a voltage‐driven urate efflux transporter URATv1 (SLC2A9) in humans. J Biol Chem 283: 26834‐26838, 2008.
 10. Anzai N, Jutabha P, Kanai Y, Endou H. Integrated physiology of proximal tubular organic anion transport. Curr Opin Nephrol Hypertens 14: 472‐479, 2005.
 11. Anzai N, Kanai Y, Endou H. New insights into renal transport of urate. Curr Opin Rheumatol 19: 151‐157, 2007.
 12. Apparsundaram S, Ferguson SM, Blakely RD. Molecular cloning and characterization of a murine hemicholinium‐3‐sensitive choline transporter. Biochem Soc Trans 29: 711‐716, 2001.
 13. Arndt P, Volk C, Gorboulev V, Budiman T, Popp C, Ulzheimer‐Teuber I, Akhoundova A, Koppatz S, Bamberg E, Nagel G, Koepsell H. Interaction of cations, anions, and weak base quinine with rat renal cation transporter rOCT2 compared with rOCT1. Am J Physiol Renal Physiol 281: F454‐F468, 2001.
 14. Asaka J, Terada T, Tsuda M, Katsura T, Inui K. Identification of essential histidine and cysteine residues of the H+/organic cation antiporter multidrug and toxin extrusion (MATE). Mol Pharmacol 71: 1487‐1493, 2007.
 15. Asaka JI, Terada T, Okuda M, Katsura T, Inui KI. Androgen receptor is responsible for Rat organic cation transporter 2 gene regulation but not for rOCT1 and rOCT3. Pharm Res 23: 697‐704, 2006.
 16. Aslamkhan A, Han YH, Walden R, Sweet DH, Pritchard JB. Stoichiometry of organic anion/dicarboxylate exchange in membrane vesicles from rat renal cortex and hOAT1‐expressing cells. Am J Physiol Renal Physiol 285: F775‐F783, 2003.
 17. Aslamkhan AG, Thompson DM, Perry JL, Bleasby K, Wolff NA, Barros S, Miller DS, Pritchard JB. The flounder organic anion transporter fOat has sequence, function, and substrate specificity similarity to both mammalian Oat1 and Oat3. Am J Physiol Regul Integr Comp Physiol 291: R1773‐R1780, 2006.
 18. Augustin R, Carayannopoulos MO, Dowd LO, Phay JE, Moley JF, Moley KH. Identification and characterization of human glucose transporter‐like protein‐9 (GLUT9): Alternative splicing alters trafficking. J Biol Chem 279: 16229‐16236, 2004.
 19. Babu E, Takeda M, Narikawa S, Kobayashi Y, Enomoto A, Tojo A, Cha SH, Sekine T, Sakthisekaran D, Endou H. Role of human organic anion transporter 4 in the transport of ochratoxin A. Biochim Biophys Acta 1590: 64‐75, 2002.
 20. Bahn A, Hagos Y, Reuter S, Balen D, Brzica H, Krick W, Burckhardt BC, Sabolic I, Burckhardt G. Identification of a new urate and high affinity nicotinate transporter, hOAT10 (SLC22A13). J Biol Chem 283: 16332‐16341, 2008.
 21. Bahn A, Hagos Y, Rudolph T, Burckhardt G. Mutation of amino acid 475 of rat organic cation transporter 2 (rOCT2) impairs organic cation transport. Biochimie 86: 133‐136, 2004.
 22. Bahn A, Hauss A, Appenroth D, Ebbinghaus D, Hagos Y, Steinmetzer P, Burckhardt G, Fleck C. RT‐PCR‐based evidence for the in vivo stimulation of renal tubular p‐aminohippurate (PAH) transport by triiodothyronine (T3) or dexamethasone (DEXA) in kidney tissue of immature and adult rats. Exp Toxicol Pathol 54: 367‐373, 2003.
 23. Bahn A, Prawitt D, Buttler D, Reid G, Enklaar T, Wolff NA, Ebbinghaus C, Hillemann A, Schulten HJ, Gunawan B, Fuzesi L, Zabel B, Burckhardt G. Genomic structure and in vivo expression of the human organic anion transporter 1 (hOAT1) gene. Biochem Biophys Res Commun 275: 623‐630, 2000.
 24. Bai J, Lai L, Yeo HC, Goh BC, Tan TM. Multidrug resistance protein 4 (MRP4/ABCC4) mediates efflux of bimane‐glutathione. Int J Biochem Cell Biol 36: 247‐257, 2004.
 25. Bakhiya A, Bahn A, Burckhardt G, Wolff NA. Human organic anion transporter 3 (hOAT3) can operate as an exchanger and mediate secretory urate flux. Cell Physiol Biochem 13: 249‐256, 2003.
 26. Bandler PE, Westlake CJ, Grant CE, Cole SP, Deeley RG. Identification of regions required for apical membrane localization of human multidrug resistance protein 2. Mol Pharmacol 74: 9‐19, 2008.
 27. Barendt WM, Wright SH. The human organic cation transporter (hOCT2) recognizes the degree of substrate ionization. J Biol Chem 277: 22491‐22496, 2002.
 28. Barros SA, Srimaroeng C, Perry JL, Walden R, Dembla‐Rajpal N, Sweet DH, Pritchard JB. Activation of protein kinase Czeta increases OAT1 (SLC22A6)‐ and OAT3 (SLC22A8)‐mediated transport. J Biol Chem 284: 2672‐2679, 2009.
 29. Bataille AM, Goldmeyer J, Renfro JL. Avian renal proximal tubule epithelium urate secretion is mediated by Mrp4. Am J Physiol Regul Integr Comp Physiol 295: R2024‐R2033, 2008.
 30. Becker ML, Kallewaard M, Caspers PW, Visser LE, Leufkens HG, Stricker BH. Hospitalisations and emergency department visits due to drug‐drug interactions: A literature review. Pharmacoepidemiol Drug Saf 16: 641‐651, 2007.
 31. Becker ML, Visser LE, van Schaik RH, Hofman A, Uitterlinden AG, Stricker BH. Interaction between polymorphisms in the OCT1 and MATE1 transporter and metformin response. Pharmacogenet Genomics 20: 38‐44, 2010.
 32. Bednarczyk D, Ekins S, Wikel JH, Wright SH. Influence of molecular structure on substrate binding to the human organic cation transporter, hOCT1. Mol Pharm 63: 489‐498, 2003.
 33. Bello‐Reuss E. Electrical properties of the basolateral membrane of the straight portion of the rabbit proximal tubule. J Physiol 326: 49‐63, 1982.
 34. Beretta GL, Benedetti V, Cossa G, Assaraf YG, Bram E, Gatti L, Corna E, Carenini N, Colangelo D, Howell SB, Zunino F, Perego P. Increased levels and defective glycosylation of MRPs in ovarian carcinoma cells resistant to oxaliplatin. Biochem Pharmacol 79: 1108‐1117, 2010.
 35. Bergwerk AJ, Shi X, Ford AC, Kanai N, Jacquemin E, Burk RD, Bai S, Novikoff PM, Stieger B, Meier PJ, Schuster VL, Wolkoff AW. Immunologic distribution of an organic anion transport protein in rat liver and kidney. Am J Physiol 271: G231‐G238, 1996.
 36. Besseghir K, Pearce LB, Rennick B. Renal tubular transport and metabolism of organic cations by the rabbit. Am J Physiol 241: F308‐F314, 1981.
 37. Bibert S, Hess SK, Firsov D, Thorens B, Geering K, Horisberger JD, Bonny O. Mouse GLUT9: Evidences for a urate uniporter. Am J Physiol Renal Physiol 297: F612‐F619, 2009.
 38. Biermann J, Lang D, Gorboulev V, Koepsell H, Sindic A, Schroter R, Zvirbliene A, Pavenstadt H, Schlatter E, Ciarimboli G. Characterization of regulatory mechanisms and states of human organic cation transporter 2. Am J Physiol Cell Physiol 290: C1521‐C1531, 2006.
 39. Blackmore CG, McNaughton PA, van Veen HW. Multidrug transporters in prokaryotic and eukaryotic cells: Physiological functions and transport mechanisms. Mol Membr Biol 18: 97‐103, 2001.
 40. Bleasby K, Castle JC, Roberts CJ, Cheng C, Bailey WJ, Sina JF, Kulkarni AV, Hafey MJ, Evers R, Johnson JM, Ulrich RG, Slatter JG. Expression profiles of 50 xenobiotic transporter genes in humans and pre‐clinical species: A resource for investigations into drug disposition. Xenobiotica 36: 963‐988, 2006.
 41. Bleasby K, Hall LA, Perry JL, Mohrenweiser HW, Pritchard JB. Functional consequences of single nucleotide polymorphisms in the human organic anion transporter hOAT1 (SLC22A6). J Pharmacol Exp Ther 314: 923‐931, 2005.
 42. Borst P, de WC, van de Wetering K. Multidrug resistance‐associated proteins 3, 4, and 5. Pflugers Arch 453: 661‐673, 2007.
 43. Boumendil‐Podevin EF, Podevin RA. Nicotinic acid transport by brush border membrane vesicles from rabbit kidney. Am J Physiol 240: F185‐F191, 1981.
 44. Bräunlich H, Meinig T, Grosch U. Postnatal development of sex differences in renal tubular transport of p‐aminohippurate (PAH) in rats. Exp Toxicol Pathol 45: 309‐313, 1993.
 45. Bräunlich H, Rassbach H, Vogelsang S. Stimulation of renal tubular transport of p‐aminohippurate in rats of different ages by treatment with adrenocortical steroids. Dev Pharmacol Ther 19: 1‐5, 1992.
 46. Brzica H, Breljak D, Ljubojevic M, Balen D, Micek V, Anzai N, Sabolic I. Optimal methods of antigen retrieval for organic anion transporters in cryosections of the rat kidney. Arh Hig Rada Toksikol 60: 7‐17, 2009.
 47. Budiman T, Bamberg E, Koepsell H, Nagel G. Mechanism of electrogenic cation transport by the cloned organic cation transporter 2 from rat. J Biol Chem 275: 29413‐29420, 2000.
 48. Buist SC, Cherrington NJ, Choudhuri S, Hartley DP, Klaassen CD. Gender‐specific and developmental influences on the expression of rat organic anion transporters. J Pharmacol Exp Ther 301: 145‐151, 2002.
 49. Buist SC, Klaassen CD. Rat and mouse differences in gender‐predominant expression of organic anion transporter (Oat1‐3; Slc22a6‐8) mRNA levels. Drug Metab Dispos 32: 620‐625, 2004.
 50. Burckhardt BC, Brai S, Wallis S, Krick W, Wolff NA, Burckhardt G. Transport of cimetidine by flounder and human renal organic anion transporter 1. Am J Physiol Renal Physiol 284: F503‐F509, 2003.
 51. Burckhardt BC, Wolff NA, Burckhardt G. Electrophysiologic characterization of an organic anion transporter cloned from winter flounder kidney (fROAT). J Am Soc Nephrol 11: 9‐17, 2000.
 52. Burckhardt G, Wolff NA. Structure of renal organic anion and cation transporters. Am J Physiol Renal Physiol 278: F853‐F866, 2000.
 53. Burger H, Zoumaro‐Djayoon A, Boersma AW, Helleman J, Berns EM, Mathijssen RH, Loos WJ, Wiemer EA. Differential transport of platinum compounds by the human organic cation transporter hOCT2 (hSLC22A2). Br J Pharmacol 159: 898‐908, 2010.
 54. Busch AE, Quester S, Ulzheimer JC, Waldegger S, Gorboulev V, Arndt P, Lang F, Koepsell H. Electrogenic properties and substrate specificity of the polyspecific rat cation transporter rOCT1. J Biol Chem 271: 32599‐32604, 1996.
 55. Cardinal J, Lapointe JY, Laprade R. Luminal and peritubular ionic substitutions and intracellular potential of the rabbit proximal convoluted tubule. Am J Physiol 247: F352‐F364, 1984.
 56. Caulfield MJ, Munroe PB, O'Neill D, Witkowska K, Charchar FJ, Doblado M, Evans S, Eyheramendy S, Onipinla A, Howard P, Shaw‐Hawkins S, Dobson RJ, Wallace C, Newhouse SJ, Brown M, Connell JM, Dominiczak A, Farrall M, Lathrop GM, Samani NJ, Kumari M, Marmot M, Brunner E, Chambers J, Elliott P, Kooner J, Laan M, Org E, Veldre G, Viigimaa M, Cappuccio FP, Ji C, Iacone R, Strazzullo P, Moley KH, Cheeseman C. SLC2A9 is a high‐capacity urate transporter in humans. PLoS Med 5: e197, 2008.
 57. Cerrutti JA, Brandoni A, Quaglia NB, Torres AM. Sex differences in p‐aminohippuric acid transport in rat kidney: Role of membrane fluidity and expression of OAT1. Mol Cell Biochem 233: 175‐179, 2002.
 58. Cerrutti JA, Quaglia NB, Brandoni A, Torres AM. Effects of gender on the pharmacokinetics of drugs secreted by the renal organic anions transport systems in the rat. Pharmacol Res 45: 107‐112, 2002.
 59. Cerrutti JA, Quaglia NB, Torres AM. Characterization of the mechanisms involved in the gender differences in p‐aminohippurate renal elimination in rats. Can J Physiol Pharmacol 79: 805‐813, 2001.
 60. Cetinkaya I, Ciarimboli G, Yalcinkaya G, Mehrens T, Velic A, Hirsch JR, Gorboulev V, Koepsell H, Schlatter E. Regulation of human organic cation transporter hOCT2 by PKA, PI3K, and calmodulin‐dependent kinases. Am J Physiol Renal Physiol 284: F293‐F302, 2003.
 61. Cha SH, Sekine T, Fukushima JI, Kanai Y, Kobayashi Y, Goya T, Endou H. Identification and characterization of human organic anion transporter 3 expressing predominantly in the kidney. Mol Pharmacol 59: 1277‐1286, 2001.
 62. Cha SH, Sekine T, Kusuhara H, Yu E, Kim JY, Kim DK, Sugiyama Y, Kanai Y, Endou H. Molecular cloning and characterization of multispecific organic anion transporter 4 expressed in the placenta. J Biol Chem 275: 4507‐4512, 2000.
 63. Chandra P, Brouwer KL. The complexities of hepatic drug transport: Current knowledge and emerging concepts. Pharm Res 21: 719‐735, 2004.
 64. Chang XB. Molecular mechanism of ATP‐dependent solute transport by multidrug resistance‐associated protein 1. Methods Mol Biol 596: 223‐249, 2010.
 65. Chatsudthipong V, Jutabha P, Evans KK, Dantzler WH. Effects of inhibitors and substitutes for chloride in lumen on p‐aminohippurate transport by isolated perfused rabbit renal proximal tubules. J Pharmacol Exp Ther 288: 993‐1001, 1999.
 66. Chen C, Klaassen CD. Rat multidrug resistance protein 4 (Mrp4, Abcc4): Molecular cloning, organ distribution, postnatal renal expression, and chemical inducibility. Biochem Biophys Res Comm 317: 46‐53, 2004.
 67. Chen C, Liu X, Smith BJ. Utility of Mdr1‐gene deficient mice in assessing the impact of P‐glycoprotein on pharmacokinetics and pharmacodynamics in drug discovery and development. Curr Drug Metab 4: 272‐291, 2003.
 68. Chen L, Takizawa M, Chen E, Schlessinger A, Choi JH, Segenthelar J, Sali A, Kubo M, Nakamura S, Iwamoto Y, Iwasaki N, Giacomini KM. Genetic polymorphisms in the organic cation transporter 1, OCT1, in Chinese and Japanese populations, exhibit altered function. J Pharmacol Exp Ther 335: 42‐50, 2010.
 69. Chen Y, Li S, Brown C, Cheatham S, Castro RA, Leabman MK, Urban TJ, Chen L, Yee SW, Choi JH, Huang Y, Brett CM, Burchard EG, Giacomini KM. Effect of genetic variation in the organic cation transporter 2 on the renal elimination of metformin. Pharmacogenet Genomics 19: 497‐504, 2009.
 70. Chen Y, Teranishi K, Li S, Yee SW, Hesselson S, Stryke D, Johns SJ, Ferrin TE, Kwok P, Giacomini KM. Genetic variants in multidrug and toxic compound extrusion‐1, hMATE1, alter transport function. Pharmacogenomics J 9: 127‐136, 2009.
 71. Chen ZS, Lee K, Kruh GD. Transport of cyclic nucleotides and estradiol 17‐β‐D‐glucuronide by multidrug resistance protein 4. Resistance to 6‐mercaptopurine and 6‐thioguanine. J Biol Chem 276: 33747‐33754, 2001.
 72. Chen ZS, Lee K, Walther S, Raftogianis RB, Kuwano M, Zeng H, Kruh GD. Analysis of methotrexate and folate transport by multidrug resistance protein 4 (ABCC4): MRP4 is a component of the methotrexate efflux system. Cancer Res 62: 3144‐3150, 2002.
 73. Chinn LW, Kroetz DL. ABCB1 pharmacogenetics: Progress, pitfalls, and promise. Clin Pharmacol Ther 81: 265‐269, 2007.
 74. Christian CD Jr, Meredith CG, Speeg KV Jr. Cimetidine inhibits renal procainamide clearance. Clin Pharmacol Ther 36: 221‐227, 1984.
 75. Chu XY, Liang Y, Cai X, Cuevas‐Licea K, Rippley RK, Kassahun K, Shou M, Braun MP, Doss GA, Anari MR, Evers R. Metabolism and renal elimination of gaboxadol in humans: Role of UDP‐glucuronosyltransferases and transporters. Pharm Res 26: 459‐468, 2009.
 76. Ciarimboli G, Schlatter E. Regulation of organic cation transport. Pflugers Arch 449: 423‐441, 2005.
 77. Ciarimboli G, Struwe K, Arndt P, Gorboulev V, Koepsell H, Schlatter E, Hirsch JR. Regulation of the human organic cation transporter hOCT1. J Cell Physiol 201: 420‐428, 2004.
 78. Cihlar T, Lin DC, Pritchard JB, Fuller MD, Mendel DB, Sweet DH. The antiviral nucleotide analogs cidofovir and adefovir are novel substrates for human and rat renal organic anion transporter 1. Mol Pharmacol 56: 570‐580, 1999.
 79. Clavell AL, Burnett JC Jr. Physiologic and pathophysiologic roles of endothelin in the kidney. Curr Opin Nephrol Hypertens 3: 66‐72, 1994.
 80. Cortney MA, Mylle M, Lassiter WE, Gottschalk CW. Renal tubular transport of water, solute, and PAH in rats loaded with isotonic saline. Am J Physiol 209: 1199‐1205, 1965.
 81. Cropp CD, Komori T, Shima JE, Urban TJ, Yee SW, More SS, Giacomini KM. Organic anion transporter 2 (SLC22A7) is a facilitative transporter of cGMP. Mol Pharmacol 73: 1151‐1158, 2008.
 82. Cui Y, Konig J, Buchholz JK, Spring H, Leier I, Keppler D. Drug resistance and ATP‐dependent conjugate transport mediated by the apical multidrug resistance protein, MRP2, permanently expressed in human and canine cells. Mol Pharmacol 55: 929‐937, 1999.
 83. Cundy KC. Clinical pharmacokinetics of the antiviral nucleotide analogues cidofovir and adefovir. Clin Pharmacokinet 36: 127‐143, 1999.
 84. Daly AK, Aithal GP, Leathart JB, Swainsbury RA, Dang TS, Day CP. Genetic susceptibility to diclofenac‐induced hepatotoxicity: Contribution of UGT2B7, CYP2C8, and ABCC2 genotypes. Gastroenterology 132: 272‐281, 2007.
 85. Dantzler WH. Comparative aspects of renal urate transport. Kidney Int 49: 1549‐1551, 1996.
 86. Dantzler WH. Renal tubular secretion of organic anions. Adv Drug Del Rev 25: 217‐230, 1997.
 87. Dantzler WH, Braun EJ. Comparative nephron function in reptiles, birds, and mammals. Am J Physiol 239: R197‐R213, 1980.
 88. Dantzler WH, Brokl O, Wright SH. Brush‐border TEA transport in intact proximal tubules and isolated membrane vesicles. Am J Physiol 256: F290‐F297, 1989.
 89. Dantzler WH, Evans KK, Wright SH. Basolateral choline transport in isolated rabbit renal proximal tubule. Pflugers Arch 436: 899‐905, 1998.
 90. Dantzler WH, Wright SH. Renal tubular transport of organic anions and cations. In: Goldstein RS, editor. Comprehensive Toxicology. V. 7. Renal Toxicology. New York: Pergamon, 61‐75, 1997.
 91. Dantzler WH, Wright SH. The molecular and cellular physiology of basolateral organic anion transport in mammalian renal tubules. Biochim Biophys Acta 1618: 185‐193, 2003.
 92. Dantzler WH, Wright SH, Chatsudthipong V, Brokl O. Basolateral tetraethylammonium transport in intact tubules: Specificity and trans‐stimulation. Am J Physiol 261: F386‐F392, 1991.
 93. David C, Rumrich G, Ullrich KJ. Luminal transport system for H+/organic cations in the rat proximal tubule. Kinetics, dependence on pH; specificity as compared with the contraluminal organic cation‐transport system. Pflugers Arch 430: 477‐492, 1995.
 94. Deeley RG, Westlake C, Cole SP. Transmembrane transport of endo‐ and xenobiotics by mammalian ATP‐binding cassette multidrug resistance proteins. Physiol Rev 86: 849‐899, 2006.
 95. Deguchi T, Kouno Y, Terasaki T, Takadate A, Otagiri M. Differential contributions of rOat1 (Slc22a6) and rOat3 (Slc22a8) to the in vivo renal uptake of uremic toxins in rats. Pharm Res 22: 619‐627, 2005.
 96. Deguchi T, Kusuhara H, Takadate A, Endou H, Otagiri M, Sugiyama Y. Characterization of uremic toxin transport by organic anion transporters in the kidney. Kidney Int 65: 162‐174, 2004.
 97. Deguchi T, Ohtsuki S, Otagiri M, Takanaga H, Asaba H, Mori S, Terasaki T. Major role of organic anion transporter 3 in the transport of indoxyl sulfate in the kidney. Kidney Int 61: 1760‐1768, 2002.
 98. Deguchi T, Takemoto M, Uehara N, Lindup WE, Suenaga A, Otagiri M. Renal clearance of endogenous hippurate correlates with expression levels of renal organic anion transporters in uremic rats. J Pharmacol Exp Ther 314: 932‐938, 2005.
 99. Demeule M, Jodoin J, Beaulieu E, Brossard M, Beliveau R. Dexamethasone modulation of multidrug transporters in normal tissues. FEBS Lett 442: 208‐214, 1999.
 100. Dinour D, Gray NK, Campbell S, Shu X, Sawyer L, Richardson W, Rechavi G, Amariglio N, Ganon L, Sela BA, Bahat H, Goldman M, Weissgarten J, Millar MR, Wright AF, Holtzman EJ. Homozygous SLC2A9 mutations cause severe renal hypouricemia. J Am Soc Nephrol 21: 64‐72, 2010.
 101. Dresser MJ, Gray AT, Giacomini KM. Kinetic and selectivity differences between rodent, rabbit, and human organic cation transporters (OCT1). J Pharmacol Exp Ther 292: 1146‐1152, 2000.
 102. Dudas PL, Pelis RM, Braun EJ, Renfro JL. Transepithelial urate transport by avian renal proximal tubule epithelium in primary culture. J Exp Biol 208: 4305‐4315, 2005.
 103. Ekaratanawong S, Anzai N, Jutabha P, Miyazaki H, Noshiro R, Takeda M, Kanai Y, Sophasan S, Endou H. Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules. J Pharmacol Sci 94: 297‐304, 2004.
 104. El Sheikh AA, Van Den Heuvel JJ, Koenderink JB, Russel FG. Interaction of nonsteroidal anti‐inflammatory drugs with multidrug resistance protein (MRP) 2/A. J Pharmacol Exp Ther 320: 229‐235, 2007.
 105. El‐Sheikh AA, Van Den Heuvel JJ, Krieger E, Russel FG, Koenderink JB. Functional role of arginine 375 in transmembrane helix 6 of multidrug resistance protein 4 (MRP4/ABCC4). Mol Pharmacol 74: 964‐971, 2008.
 106. Endres CJ, Hsiao P, Chung FS, Unadkat JD. The role of transporters in drug interactions. Eur J Pharm Sci 27: 501‐517, 2006.
 107. Enomoto A, Kimura H, Chairoungdua A, Shigeta Y, Jutabha P, Cha SH, Hosoyamada M, Takeda M, Sekine T, Igarashi T, Matsuo H, Kikuchi Y, Oda T, Ichida K, Hosoya T, Shimokata K, Niwa T, Kanai Y, Endou H. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature 417: 447‐452, 2002.
 108. Enomoto A, Takeda M, Shimoda M, Narikawa S, Kobayashi Y, Kobayashi Y, Yamamoto T, Sekine T, Cha SH, Niwa T, Endou H. Interaction of human organic anion transporters 2 and 4 with organic anion transport inhibitors. J Pharmacol Exp Ther 301: 797‐802, 2002.
 109. Enomoto A, Takeda M, Taki K, Takayama F, Noshiro R, Niwa T, Endou H. Interactions of human organic anion as well as cation transporters with indoxyl sulfate. Eur J Pharmacol 466: 13‐20, 2003.
 110. Enomoto A, Takeda M, Tojo A, Sekine T, Cha SH, Khamdang S, Takayama F, Aoyama I, Nakamura S, Endou H, Niwa T. Role of organic anion transporters in the tubular transport of indoxyl sulfate and the induction of its nephrotoxicity. J Am Soc Nephrol 13: 1711‐1720, 2002.
 111. Eraly SA, Hamilton BA, Nigam SK. Organic anion and cation transporters occur in pairs of similar and similarly expressed genes. Biochem Biophys Res Commun 300: 333‐342, 2003.
 112. Eraly SA, Monte JC, Nigam SK. Novel slc22 transporter homologs in fly, worm, and human clarify the phylogeny of organic anion and cation transporters. Physiol Genomics 18: 12‐24, 2004.
 113. Eraly SA, Vallon V, Rieg T, Gangoiti JA, Wikoff WR, Siuzdak G, Barshop BA, Nigam SK. Multiple organic anion transporters contribute to net renal excretion of uric acid. Physiol Genomics 33: 180‐192, 2008.
 114. Eraly SA, Vallon V, Vaughn DA, Gangoiti JA, Nigam SK, Nagle M, Monte JC, Rieg T, Truong DM, Long JM, Barshop BA, Kaler G, Nigam SK. Decreased renal organic anion secretion and plasma accumulation of endogenous organic anions in OAT1 knock‐out mice. J Biol Chem 281: 5072‐5083, 2006.
 115. Erdman AR, Mangravite LM, Urban TJ, Lagpacan LL, Castro RA, de la Cruz M, Chan W, Huang CC, Johns SJ, Kawamoto M, Stryke D, Taylor TR, Carlson EJ, Ferrin TE, Brett CM, Burchard EG, Giacomini KM. The human organic anion transporter 3 (OAT3; SLC22A8): Genetic variation and functional genomics. Am J Physiol Renal Physiol 290: F905‐F912, 2006.
 116. Ernest S, Bello‐Reuss E. Expression and function of P‐glycoprotein in a mouse kidney cell line. Am J Physiol 269: C323‐C333, 1995.
 117. Ernest S, Rajaraman S, Megyesi J, Bello‐Reuss EN. Expression of MDR1 (multidrug resistance) gene and its protein in normal human kidney. Nephron 77: 284‐289, 1997.
 118. Evers R, de HM, Sparidans R, Beijnen J, Wielinga PR, Lankelma J, Borst P. Vinblastine and sulfinpyrazone export by the multidrug resistance protein MRP2 is associated with glutathione export. Br J Cancer 83: 375‐383, 2000.
 119. Fernandez SB, Hollo Z, Kern A, Bakos E, Fischer PA, Borst P, Evers R. Role of the N‐terminal transmembrane region of the multidrug resistance protein MRP2 in routing to the apical membrane in MDCKII cells. J Biol Chem 277: 31048‐31055, 2002.
 120. Filipski KK, Mathijssen RH, Mikkelsen TS, Schinkel AH, Sparreboom A. Contribution of organic cation transporter 2 (OCT2) to cisplatin‐induced nephrotoxicity. Clin Pharmacol Ther 86: 396‐402, 2009.
 121. Fisher MB, Paine MF, Strelevitz TJ, Wrighton SA. The role of hepatic and extrahepatic UDP‐glucuronosyltransferases in human drug metabolism. Drug Metab Rev 33: 273‐297, 2001.
 122. Franke RM, Gardner ER, Sparreboom A. Pharmacogenetics of drug transporters. Curr Pharm Des 16: 220‐230, 2010.
 123. Fujita T, Brown C, Carlson EJ, Taylor T, de la Cruz M, Johns SJ, Stryke D, Kawamoto M, Fujita K, Castro R, Chen CW, Lin ET, Brett CM, Burchard EG, Ferrin TE, Huang CC, Leabman MK, Giacomini KM. Functional analysis of polymorphisms in the organic anion transporter, SLC22A6 (OAT1). Pharmacogenet Genomics 15: 201‐209, 2005.
 124. Fujita T, Shimada A, Okada N, Yamamoto A. Functional characterization of Na+‐independent choline transport in primary cultures of neurons from mouse cerebral cortex. Neurosci Lett 393: 216‐221, 2006.
 125. Gao M, Yamazaki M, Loe DW, Westlake CJ, Grant CE, Cole SP, Deeley RG. Multidrug resistance protein. Identification of regions required for active transport of leukotriene C4. J Biol Chem 273: 10733‐10740, 1998.
 126. Garcia ML, Benavides J, Valdivieso F. Ketone body transport in renal brush border membrane vesicles. Biochim Biophys Acta 600: 922‐930, 1980.
 127. Gekle M, Mildenberger S, Sauvant C, Bednarczyk D, Wright SH, Dantzler WH. Inhibition of initial transport rate of basolateral organic anion carrier in renal PT by BK and phenylephrine. Am J Physiol 277: F251‐F256, 1999.
 128. Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, Chu X, Dahlin A, Evers R, Fischer V, Hillgren KM, Hoffmaster KA, Ishikawa T, Keppler D, Kim RB, Lee CA, Niemi M, Polli JW, Sugiyama Y, Swaan PW, Ware JA, Wright SH, Yee SW, Zamek‐Gliszczynski MJ, Zhang L. Membrane transporters in drug development. Nat Rev Drug Discov 9: 215‐236, 2010.
 129. Gluck S, Nelson R. The role of the V‐ATPase in renal epithelial H+ transport. J Exp Biol 172: 205‐218, 1992.
 130. Goralski KB, Lou G, Prowse MT, Gorboulev V, Volk C, Koepsell H, Sitar DS. The cation transporters rOCT1 and rOCT2 interact with bicarbonate but play only a minor role for amantadine uptake into rat renal proximal tubules. J Pharmacol Exp Ther 303: 959‐968, 2002.
 131. Gorboulev V, Shatskaya N, Volk C, Koepsell H. Subtype‐specific affinity for corticosterone of rat organic cation transporters rOCT1 and rOCT2 depends on three amino acids within the substrate binding region. Mol Pharmacol 67: 1612‐1619, 2005.
 132. Gorboulev V, Volk C, Arndt P, Akhoundova A, Koepsell H. Selectivity of the polyspecific cation transporter rOCT1 is changed by mutation of aspartate 475 to glutamate. Mol Pharmacol 56: 1254‐1261, 1999.
 133. Gorbunov D, Gorboulev V, Shatskaya N, Mueller T, Bamberg E, Friedrich T, Koepsell H. High‐affinity cation binding to transporter OCT1 induces movement of helix 11 and blocks transport after mutations in a modelled interaction domain between two helices. Mol Pharmacol 73: 50‐61, 2008.
 134. Grigat S, Fork C, Bach M, Golz S, Geerts A, Schomig E, Grundemann D. The carnitine transporter SLC22A5 is not a general drug transporter, but it efficiently translocates mildronate. Drug Metab Dispos 37: 330‐337, 2009.
 135. Groves CE, Evans K, Dantzler WH, Wright SH. Peritubular organic cation transport in isolated rabbit proximal tubules. Am J Physiol 266: F450‐F458, 1994.
 136. Groves CE, Suhre WB, Cherrington NJ, Wright SH. Sex differences in the mRNA, protein, and functional expression of organic anion transporter (Oat) 1, Oat3, and organic cation transporter (Oct) 2 in rabbit renal proximal tubules. J Pharmacol Exp Ther 316: 743‐752, 2006.
 137. Gründemann D, Gorboulev V, Gambaryan S, Veyhl M, Koepsell H. Drug excretion mediated by a new prototype of polyspecific transporter. Nature 372: 549‐552, 1994.
 138. Gründemann D, Harlfinger S, Golz S, Geerts A, Lazar A, Berkels R, Jung N, Rubbert A, Schömig E. Discovery of the ergothioneine transporter. Proc Natl Acad Sci U S A 102: 5256‐5261, 2005.
 139. Gutmann H, Miller DS, Droulle A, Drewe J, Fahr A, Fricker G. P‐glycoprotein‐ and mrp2‐mediated octreotide transport in renal proximal tubule. Br J Pharmacol 129: 251‐256, 2000.
 140. Hagos Y, Stein D, Ugele B, Burckhardt G, Bahn A. Human renal organic anion transporter 4 operates as an asymmetric urate transporter. J Am Soc Nephrol 18: 430‐439, 2007.
 141. Haisa M, Matsumoto T, Komatsu T, Mariyama Y. Wide variety of locations for rodent MATE1, a transporter protein that mediates the final excretion step for toxic organic cations. Am J Physiol Cell Physiol 291: C678‐C86, 2006.
 142. Han YH, Busler D, Hong Y, Tian Y, Chen C, Rodrigues AD. Transporter studies with the 3‐O‐sulfate conjugate of 17α‐ethinylestradiol: Assessment of human kidney drug transporters. Drug Metab Dispos 38: 1064‐1071, 2010.
 143. Hartmann G, Vassileva V, Piquette‐Miller M. Impact of endotoxin‐induced changes in P‐glycoprotein expression on disposition of doxorubicin in mice. Drug Metab Dispos 33: 820‐828, 2005.
 144. Hawk CT, Dantzler WH. Tetraethylammonium transport by isolated perfused snake renal tubules. Am J Physiol 246: F476‐F487, 1984.
 145. Hayer M, Bonisch H, Bruss M. Molecular cloning, functional characterization and genomic organization of four alternatively spliced isoforms of the human organic cation transporter 1 (hOCT1/SLC22A1). Ann Hum Genet 63: 473‐482, 1999.
 146. He X, Szewczyk P, Karyakin A, Evin M, Chang G. Structure of a cation‐bound multidrug and toxin compound extrusion (MATE) transporter. Nature 467: 991‐994, 2010.
 147. Hediger MA, Johnson RJ, Miyazaki H, Endou H. Molecular physiology of urate transport. Physiology (Bethesda) 20: 125‐133, 2005.
 148. Hiasa M, Matsumoto T, Komatsu T, Omote H, Moriyama Y. Functional characterization of testis‐specific rodent multidrug and toxic compound extrusion 2, a class III MATE‐type polyspecific H+/organic cation exporter. Am J Physiol Cell Physiol 293: C1437‐C1444, 2007.
 149. Higgins CF, Callaghan R, Linton KJ, Rosenberg MF, Ford RC. Structure of the multidrug resistance P‐glycoprotein. Semin Cancer Biol 8: 135‐142, 1997.
 150. Hirono S, Nakagome I, Imai R, Maeda K, Kusuhara H, Sugiyama Y. Estimation of the three‐dimensional pharmacophore of ligands for rat multidrug‐resistance‐associated protein 2 using ligand‐based drug design techniques. Pharm Res 22: 260‐269, 2005.
 151. Ho ES, Lin DC, Mendel DB, Cihlar T. Cytotoxicity of antiviral nucleotides adefovir and cidofovir is induced by the expression of human renal organic anion transporter 1. J Am Soc Nephrol 11: 383‐393, 2000.
 152. Hocher B, Thone‐Reineke C, Bauer C, Raschack M, Neumayer HH. The paracrine endothelin system: Pathophysiology and implications in clinical medicine. Eur J Clin Chem Clin Biochem 35: 175‐189, 1997.
 153. Holohan PD, Pessah NI, Ross CR. Binding of N1‐methylnicotinamide and p‐aminohippuric acid to a particulate fraction from dog kidney. J Pharmacol Exp Ther 195: 22‐33, 1975.
 154. Holohan PD, Ross CR. Mechanisms of organic cation transport in kidney plasma membrane vesicles: 1. Countertransport studies. J Pharmacol Exp Ther 215: 191‐197, 1980.
 155. Holohan PD, Ross CR. Mechanisms of organic cation transport in kidney plasma membrane vesicles: 2. DpH studies. J Pharmacol Exp Ther 216: 294‐298, 1981.
 156. Hong M, Xu W, Yoshida T, Tanaka K, Wolff DJ, Zhou F, Inouye M, You G. Human organic anion transporter hOAT1 forms homooligomers. J Biol Chem 280: 32285‐32290, 2005.
 157. Hong M, Zhou F, Lee K, You G. The putative transmembrane segment 7 of human organic anion transporter hOAT1 dictates transporter substrate binding and stability. J Pharmacol Exp Ther 320: 1209‐1215, 2007.
 158. Hong M, Zhou F, You G. Critical amino acid residues in transmembrane domain 1 of the human organic anion transporter hOAT1. J Biol Chem 279: 31478‐31482, 2004.
 159. Hooijberg JH, Broxterman HJ, Kool M, Assaraf YG, Peters GJ, Noordhuis P, Scheper RJ, Borst P, Pinedo HM, Jansen G. Antifolate resistance mediated by the multidrug resistance proteins MRP1 and MRP2. Cancer Res 59: 2532‐2535, 1999.
 160. Hosoyamada M, Sekine T, Kanai Y, Endou H. Molecular cloning and functional expression of a multispecific organic anion transporter from human kidney. Am J Physiol 276: F122‐F128, 1999.
 161. Huang Y, Lemieux MJ, Song J, Auer M, Wang DN. Structure and mechanism of the glycerol‐3‐phosphate transporter from Escherichia coli. Science 301: 616‐620, 2003.
 162. Hulot JS, Villard E, Maguy A, Morel V, Mir L, Tostivint I, William‐Faltaos D, Fernandez C, Hatem S, Deray G, Komajda M, Leblond V, Lechat P. A mutation in the drug transporter gene ABCC2 associated with impaired methotrexate elimination. Pharmacogenet Genomics 15: 277‐285, 2005.
 163. Ichida K, Hosoyamada M, Hisatome I, Enomoto A, Hikita M, Endou H, Hosoya T. Clinical and molecular analysis of patients with renal hypouricemia in Japan‐influence of URAT1 gene on urinary urate excretion. J Am Soc Nephrol 15: 164‐173, 2004.
 164. Ichida K, Hosoyamada M, Kimura H, Takeda M, Utsunomiya Y, Hosoya T, Endou H. Urate transport via human PAH transporter hOAT1 and its gene structure. Kidney Int 63: 143‐155, 2003.
 165. Ieiri I, Higuchi S, Sugiyama Y. Genetic polymorphisms of uptake (OATP1B1, 1B3) and efflux (MRP2, BCRP) transporters: Implications for inter‐individual differences in the pharmacokinetics and pharmacodynamics of statins and other clinically relevant drugs. Expert Opin Drug Metab Toxicol 5: 703‐729, 2009.
 166. Iharada M, Miyaji T, Fujimoto T, Hiasa M, Anzai N, Omote H, Moriyama Y. Type 1 sodium‐dependent phosphate transporter (SLC17A1 Protein) is a Cl‐‐dependent urate exporter. J Biol Chem 285: 26107‐26113, 2010.
 167. Imaoka T, Kusuhara H, Adachi M, Schuetz JD, Takeuchi K, Sugiyama Y. Functional involvement of multidrug resistance‐associated protein 4 (MRP4/ABCC4) in the renal elimination of the antiviral drugs adefovir and tenofovir. Mol Pharmacol 71: 619‐627, 2007.
 168. Inazu M, Takeda H, Matsumiya T. Molecular and functional characterization of an Na+‐independent choline transporter in rat astrocytes. J Neurochem 94: 1427‐1437, 2005.
 169. Ito K, Oleschuk CJ, Westlake C, Vasa MZ, Deeley RG, Cole SP. Mutation of Trp1254 in the multispecific organic anion transporter, multidrug resistance protein 2 (MRP2) (ABCC2), alters substrate specificity and results in loss of methotrexate transport activity. J Biol Chem 276: 38108‐38114, 2001.
 170. Ito K, Suzuki H, Sugiyama Y. Charged amino acids in the transmembrane domains are involved in the determination of the substrate specificity of rat Mrp2. Mol Pharmacol 59: 1077‐1085, 2001.
 171. Itoda M, Saito Y, Maekawa K, Hichiya H, Komamura K, Kamakura S, Kitakaze M, Tomoike H, Ueno K, Ozawa S, Sawada J. Seven novel single nucleotide polymorphisms in the human SLC22A1 gene encoding organic cation transporter 1 (OCT1). Drug Metab Pharmacokinet 19: 308‐312, 2004.
 172. Iwanaga T, Kobayashi D, Hirayama M, Maeda T, Tamai I. Involvement of uric acid transporter in increased renal clearance of the xanthine oxidase inhibitor oxypurinol induced by a uricosuric agent, benzbromarone. Drug Metab Dispos 33: 1791‐1795, 2005.
 173. Izzedine H, Hulot JS, Villard E, Goyenvalle C, Dominguez S, Ghosn J, Valantin MA, Lechat P, Deray AG. Association between ABCC2 gene haplotypes and tenofovir‐induced proximal tubulopathy. J Infect Dis 194: 1481‐1491, 2006.
 174. Jedlitschky G, Hoffmann U, Kroemer HK. Structure and function of the MRP2 (ABCC2) protein and its role in drug disposition. Expert Opin Drug Metab Toxicol 2: 351‐366, 2006.
 175. Jemnitz K, Heredi‐Szabo K, Janossy J, Ioja E, Vereczkey L, Krajcsi P. ABCC2/Abcc2: A multispecific transporter with dominant excretory functions. Drug Metab Rev 42: 402‐436, 2010.
 176. Johnson AG, Seideman P, Day RO. Adverse drug interactions with nonsteroidal anti‐inflammatory drugs (NSAIDs). Recognition, management and avoidance. Drug Saf 8: 99‐127, 1993.
 177. Jonker JW, Wagenaar E, Mol CA, Buitelaar M, Koepsell H, Smit JW, Schinkel AH. Reduced hepatic uptake and intestinal excretion of organic cations in mice with a targeted disruption of the organic cation transporter 1 (Oct1 [Slc22a1]) gene. Mol Cell Biol 21: 5471‐5477, 2001.
 178. Jonker JW, Wagenaar E, Van Eijl S, Schinkel AH. Deficiency in the organic cation transporters 1 and 2 (Oct1/Oct2 [Slc22a1/Slc22a2]) in mice abolishes renal secretion of organic cations. Mol Cell Biol 23: 7902‐7908, 2003.
 179. Jutabha P, Anzai N, Kitamura K, Taniguchi A, Kaneko S, Yan K, Yamada H, Shimada H, Kimura T, Katada T, Fukutomi T, Tomita K, Urano W, Yamanaka H, Seki G, Fujita T, Moriyama Y, Yamada A, Uchida S, Wempe MF, Endou H, Sakurai H. Human sodium‐phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate. J Biol Chem 285: 35123‐35132, 2010.
 180. Kaewmokul S, Chatsudthipong V, Evans KK, Dantzler WH, Wright SH. Functional mapping of rbOCT1 and rbOCT2 activity in the S2 segment of rabbit proximal tubule. Am J Physiol Renal Physiol 285: F1149‐F1159, 2003.
 181. Karbach U, Kricke J, Meyer‐Wentrup F, Gorboulev V, Volk C, Loffing‐Cueni D, Kaissling B, Bachmann S, Koepsell H. Localization of organic cation transporters OCT1 and OCT2 in rat kidney. Am J Physiol Renal Physiol 279: F679‐F687, 2000.
 182. Kato Y, Kubo Y, Iwata D, Kato S, Sudo T, Sugiura T, Kagaya T, Wakayama T, Hirayama A, Sugimoto M, Sugihara K, Kaneko S, Soga T, Asano M, Tomita M, Matsui T, Wada M, Tsuji A. Gene knockout and metabolome analysis of carnitine/organic cation transporter OCTN1. Pharm Res 27: 832‐840, 2010.
 183. Kato Y, Yoshida K, Watanabe C, Sai Y, Tsuji A. Screening of the interaction between xenobiotic transporters and PDZ proteins. Pharm Res 21: 1886‐1894, 2004.
 184. Kauffmann HM, Schrenk D. Sequence analysis and functional characterization of the 5′‐ flanking region of the rat multidrug resistance protein 2 (mrp2) gene. Biochem Biophys Res Commun 245: 325‐331, 1998.
 185. Keitel V, Kartenbeck J, Nies AT, Spring H, Brom M, Keppler D. Impaired protein maturation of the conjugate export pump multidrug resistance protein 2 as a consequence of a deletion mutation in Dubin‐Johnson syndrome. Hepatology 32: 1317‐1328, 2000.
 186. Kekuda R, Prasad PD, Wu X, Wang H, Fei YJ, Leibach FH, Ganapathy V. Cloning and functional characterization of a potential‐sensitive, polyspecific organic cation transporter (OCT3) most abundantly expressed in placenta. J Biol Chem 273: 15971‐15979, 1998.
 187. Keller T, Schwarz D, Bernhard F, Dotsch V, Hunte C, Gorboulev V, Koepsell H. Cell free expression and functional reconstitution of eukaryotic drug transporters. Biochemistry 47: 4552‐4564, 2008.
 188. Khamdang S, Takeda M, Shimoda M, Noshiro R, Narikawa S, Huang XL, Enomoto A, Piyachaturawat P, Endou H. Interactions of human‐ and rat‐organic anion transporters with pravastatin and cimetidine. J Pharmacol Sci 94: 197‐202, 2004.
 189. Kikuchi R, Kusuhara H, Hattori N, Shiota K, Kim I, Gonzalez FJ, Sugiyama Y. Regulation of the expression of human organic anion transporter 3 by hepatocyte nuclear factor 1alpha/beta and DNA methylation. Mol Pharmacol 70: 887‐896, 2006.
 190. Kikuchi Y, Koga H, Yasutomo Y, Kawabata Y, Shimizu E, Naruse M, Kiyama S, Nonoguchi H, Tomita K, Sasatomi Y, Takebayashi S. Patients with renal hypouricemia with exercise‐induced acute renal failure and chronic renal dysfunction. Clin Nephrol 53: 467‐472, 2000.
 191. Kim YK, Dantzler WH. Effects of pH on basolateral tetraethylammonium transport in snake renal proximal tubules. Am J Physiol 272: R955‐R961, 1997.
 192. Kinsella JL, Holohan PD, Pessah NI, Ross CR. Transport of organic ions in renal cortical luminal and antiluminal membrane vesicles. J Pharmacol Exp Ther 209: 443‐450, 1979.
 193. Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: Function and regulation. Pharmacol Rev 62: 1‐96, 2010.
 194. Kobara A, Hiasa M, Matsumoto T, Otsuka M, Omote H, Moriyama Y. A novel variant of mouse MATE‐1 H+/organic cation antiporter with a long hydrophobic tail. Arch Biochem Biophys 469: 195‐199, 2008.
 195. Kobayashi Y, Ohshiro N, Sakai R, Ohbayashi M, Kohyama N, Yamamoto T. Transport mechanism and substrate specificity of human organic anion transporter 2 (hOat2 [SLC22A7]). J Pharm Pharmacol 57: 573‐578, 2005.
 196. Koepsell H. Substrate recognition and translocation by polyspecific organic cation transporters. Biol Chem 392: 95‐101, 2011.
 197. Koepsell H, Endou H. The SLC22 drug transporter family. Pflugers Arch 447: 666‐676, 2004.
 198. Kojima R, Sekine T, Kawachi M, Cha SH, Suzuki Y, Endou H. Immunolocalization of multispecific organic anion transporters, OAT1, OAT2, and OAT3, in rat kidney. J Am Soc Nephrol 13: 848‐857, 2002.
 199. Konig J, Nies AT, Cui Y, Leier I, Keppler D. Conjugate export pumps of the multidrug resistance protein (MRP) family: Localization, substrate specificity, and MRP2‐mediated drug resistance. Biochim Biophys Acta 1461: 377‐394, 1999.
 200. Kristufek D, Rudorfer W, Pifl C, Huck S. Organic cation transporter mRNA and function in the rat superior cervical ganglion. J Physiol 543: 117‐134, 2002.
 201. Kusuhara H, Sekine T, Utsunomiya‐Tate N, Tsuda M, Kojima R, Cha SH, Sugiyama Y, Kanai Y, Endou H. Molecular cloning and characterization of a new multispecific organic anion transporter from rat brain. J Biol Chem 274: 13675‐13680, 1999.
 202. Lahjouji K, Mitchell GA, Qureshi IA. Carnitine transport by organic cation transporters and systemic carnitine deficiency. Mol Genet Metab 73: 287‐297, 2001.
 203. Lai L, Tan TM. Role of glutathione in the multidrug resistance protein 4 (MRP4/ABCC4)‐mediated efflux of cAMP and resistance to purine analogues. Biochem J 361: 497‐503, 2002.
 204. Lalezari JP, Stagg RJ, Kuppermann BD, Holland GN, Kramer F, Ives DV, Youle M, Robinson MR, Drew WL, Jaffe HS. Intravenous cidofovir for peripheral cytomegalovirus retinitis in patients with AIDS. A randomized, controlled trial. Ann Intern Med 126: 257‐263, 1997.
 205. Leabman MK, Huang CC, DeYoung J, Carlson EJ, Taylor TR, De La CM, Johns SJ, Stryke D, Kawamoto M, Urban TJ, Kroetz DL, Ferrin TE, Clark AG, Risch N, Herskowitz I, Giacomini KM and Pharmacogenetics of Membrane Transporters Investigators. Natural variation in human membrane transporter genes reveals evolutionary and functional constraints. Proc Natl Acad Sci U S A 100: 5896‐5901, 2003.
 206. Leabman MK, Huang CC, Kawamoto M, Johns SJ, Stryke D, Ferrin TE, DeYoung J, Taylor T, Clark AG, Herskowitz I, Giacomini KM. Polymorphisms in a human kidney xenobiotic transporter, OCT2, exhibit altered function. Pharmacogenetics 12: 395‐405, 2002.
 207. Lee W, Glaeser H, Smith LH, Roberts RL, Moeckel GW, Gervasini G, Leake BF, Kim RB. Polymorphisms in human organic anion‐transporting polypeptide 1A2 (OATP1A2): Implications for altered drug disposition and central nervous system drug entry. J Biol Chem 280: 9610‐9617, 2005.
 208. Leier I, Hummel‐Eisenbeiss J, Cui Y, Keppler D. ATP‐dependent para‐aminohippurate transport by apical multidrug resistance protein MRP2. Kidney Int 57: 1636‐1642, 2000.
 209. Leslie E, Ito K, Upadhyaya P, Hecht S, Deeley R, Cole S. Transport of the β‐O‐Glucuronide conjugate of the tobacco‐specific carcinogen 4‐(methylnitrosamino)‐1‐(3‐pyridyl)‐1‐butanol (NNAL) by the multidrug resistance protein 1 (MRP1). J Biol Chem 276: 27846‐27854, 2001.
 210. Li N, Hartley DP, Cherrington NJ, Klaassen CD. Tissue expression, ontogeny, and inducibility of rat organic anion transporting polypeptide 4. J Pharmacol Exp Ther 301: 551‐560, 2002.
 211. Lickteig AJ, Cheng X, Augustine LM, Klaassen CD, Cherrington NJ. Tissue distribution, ontogeny and induction of the transporters Multidrug and toxin extrusion (MATE) 1 and MATE2 mRNA expression levels in mice. Life Sci 83: 59‐64, 2008.
 212. Ljubojevic M, Balen D, Breljak D, Kusan M, Anzai N, Bahn A, Burckhardt G, Sabolic I. Renal expression of organic anion transporter OAT2 in rats and mice is regulated by sex hormones. Am J Physiol Renal Physiol 292: F361‐F372, 2007.
 213. Ljubojevic M, Herak‐Kramberger CM, Hagos Y, Bahn A, Endou H, Burckhardt G, Sabolic I. Rat renal cortical OAT1 and OAT3 exhibit gender differences determined by both androgen stimulation and estrogen inhibition. Am J Physiol Renal Physiol 287: F124‐F138, 2004.
 214. Lopez‐Nieto CE, You G, Bush KT, Barros EJ, Beier DR, Nigam SK. Molecular cloning and characterization of NKT, a gene product related to the organic cation transporter family that is almost exclusively expressed in the kidney. J Biol Chem 272: 6471‐6478, 1997.
 215. Lu K, Nishimori H, Nakamura Y, Shima K, Kuwajima M. A missense mutation of mouse OCTN2, a sodium‐dependent carnitine cotransporter, in the juvenile visceral steatosis mouse. Biochem Biophys Res Commun 252: 590‐594, 1998.
 216. Lu R, Chan BS, Schuster VL. Cloning of the human kidney PAH transporter: Narrow substrate specificity and regulation by protein kinase C. Am J Physiol 276: F295‐F303, 1999.
 217. MacDonald TT, Di SA, Gordon JN. Immunopathogenesis of Crohn's disease. JPEN J Parenter Enteral Nutr 29: S118‐S124, 2005.
 218. Maddrell SH, Gardiner BO, Pilcher DE, Reynolds SE. Active transport by insect malpighian tubules of acidic dyes and of acylamides. J Exp Biol 61: 357‐377, 1974.
 219. Maeda A, Tsuruoka S, Kanai Y, Endou H, Saito K, Miyamoto E, Fujimura A. Evaluation of the interaction between nonsteroidal anti‐inflammatory drugs and methotrexate using human organic anion transporter 3‐transfected cells. Eur J Pharmacol 596: 166‐172, 2008.
 220. Maggs JL, Grimmer SF, Orme ML, Breckenridge AM, Park BK, Gilmore IT. The biliary and urinary metabolites of [3H]17 α‐ethynylestradiol in women. Xenobiotica 13: 421‐431, 1983.
 221. Maher JM, Slitt AL, Callaghan TN, Cheng X, Cheung C, Gonzalez FJ, Klaassen CD. Alterations in transporter expression in liver, kidney, and duodenum after targeted disruption of the transcription factor HNF1alpha. Biochem Pharmacol 72: 512‐522, 2006.
 222. Maher JM, Slitt AL, Cherrington NJ, Cheng X, Klaassen CD. Tissue distribution and hepatic and renal ontogeny of the multidrug resistance‐associated protein (Mrp) family in mice. Drug Metab Dispos 33: 947‐955, 2005.
 223. Malvin RL, Wilde WS, Sullivan LP. Localization of nephron transport by stop flow analysis. Am J Physiol 194: 135‐142, 1958.
 224. Manganel M, Roch‐Ramel F, Murer H. Sodium‐pyrazinoate cotransport in rabbit renal brush border membrane vesicles. Am J Physiol 249: F400‐F408, 1985.
 225. Marshall E, Grafflin AL. The structure and function of the kidney of Lophius piscatorius. Bull Johns Hopkins Hosp 43: 205‐235, 1928.
 226. Masereeuw R, Moons MM, Toomey BH, Russel FG, Miller DS. Active lucifer yellow secretion in renal proximal tubule: Evidence for organic anion transport system crossover. J Pharmacol Exp Ther 289: 1104‐1111, 1999.
 227. Masereeuw R, Notenboom S, Smeets PH, Wouterse AC, Russel FG. Impaired renal secretion of substrates for the multidrug resistance protein 2 in mutant transport‐deficient (TR−) rats. J Am Soc Nephrol 14: 2741‐2749, 2003.
 228. Masereeuw R, Russel FG. Therapeutic implications of renal anionic drug transporters. Pharmacol Ther 126: 200‐216, 2010.
 229. Masereeuw R, Terlouw SA, van Aubel RA, Russel FG, Miller DS. Endothelin B receptor‐mediated regulation of ATP‐driven drug secretion in renal proximal tubule. Mol Pharmacol 57: 59‐67, 2000.
 230. Masuda S, Saito H, Nonoguchi H, Tomita K, Inui K‐I. mRNA distribution and membrane localization of the OAT‐K1 organic anion transporter in rat renal tubules. FEBS Lett 407: 127‐131, 1997.
 231. Masuda S, Terada T, Yonezawa A, Tanihara Y, Kishimoto K, Katsura T, Ogawa O, Inui KI. Identification and functional characterization of a new human kidney‐specific H+/organic cation antiporter, kidney‐specific multidrug and toxin extrusion 2. J Am Soc Nephrol 17: 2127‐2135, 2006.
 232. Matsumoto T, Kanamoto T, Otsuka M, Omote H, Moriyama Y. Role of glutamate residues in substrate recognition by human MATE1 polyspecific H+/organic cation exporter. Am J Physiol Cell Physiol 294: C1074‐C1078, 2008.
 233. Matsushima S, Maeda K, Inoue K, Ohta KY, Yuasa H, Kondo T, Nakayama H, Horita S, Kusuhara H, Sugiyama Y. The inhibition of human multidrug and toxin extrusion 1 (hMATE1) is involved in the drug‐drug interaction caused by cimetidine. Drug Metab Dispos 37: 555‐559, 2008.
 234. McKinney TD. Procainamide uptake by rabbit proximal tubules. J Pharmacol Exp Ther 224: 302‐306, 1983.
 235. McKinney TD. Further studies of organic base secretion by rabbit proximal tubules. Am J Physiol 246: F282‐F289, 1984.
 236. McKinney TD, Myers P, Speeg KV Jr. Cimetidine secretion by rabbit renal tubules in vitro. Am J Physiol 241: F69‐F76, 1981.
 237. McKinney TD, Speeg KV Jr. Cimetidine and procainamide secretion by proximal tubules in vitro. Am J Physiol 242: F672‐F680, 1982.
 238. Meetam P, Srimaroeng C, Soodvilai S, Chatsudthipong V. Regulatory role of testosterone in organic cation transport: In vivo and in vitro studies. Biol Pharm Bull 32: 982‐987, 2009.
 239. Mehrens T, Lelleck S, Cetinkaya I, Knollmann M, Hohage H, Gorboulev V, Boknik P, Koepsell H, Schlatter E. The affinity of the organic cation transporter rOCT1 is increased by protein kinase C‐dependent phosphorylation. J Am Soc Nephrol 11: 1216‐1224, 2000.
 240. Meijer DKF, Mol WEM, Müller M, Kurz G. Carrier‐mediated transport in the hepatic distribution and elimination of drugs, with special reference to the category of organic cations. J Pharmacokinet Biopharm 18: 35‐70, 1990.
 241. Michel V, Yuan Z, Ramsubir S, Bakovic M. Choline transport for phospholipid synthesis. Exp Biol Med (Maywood) 231: 490‐504, 2006.
 242. Mikkaichi T, Suzuki T, Onogawa T, Tanemoto M, Mizutamari H, Okada M, Chaki T, Masuda S, Tokui T, Eto N, Abe M, Satoh F, Unno M, Hishinuma T, Inui K, Ito S, Goto J, Abe T. Isolation and characterization of a digoxin transporter and its rat homologue expressed in the kidney. Proc Natl Acad Sci U S A 101: 3569‐3574, 2004.
 243. Miller DS. Protein kinase C regulation of organic anion transport in renal proximal tubule. Am J Physiol 274: F156‐F164, 1998.
 244. Miller DS. Nucleoside phosphonate interactions with multiple organic anion transporters in renal proximal tubule. J Pharmacol Exp Ther 299: 567‐574, 2001.
 245. Miller DS, Fricker G, Drewe J. p‐Glycoprotein‐mediated transport of a fluorescent rapamycin derivative in renal proximal tubule. J Pharmacol Exp Ther 282: 440‐444, 1997.
 246. Miller DS, Sussman CR, Renfro JL. Protein kinase C regulation of p‐glycoprotein‐mediated xenobiotic secretion in renal proximal tubule. Am J Physiol 275: F785‐F795, 1998.
 247. Miyamoto Y, Tiruppathi C, Ganapathy V, Leibach FH. Multiple transport systems for organic cations in renal brush‐border membrane vesicles. Am J Physiol 256: F540‐F548, 1989.
 248. Miyazaki H, Anzai N, Ekaratanawong S, Sakata T, Shin HJ, Jutabha P, Hirata T, He X, Nonoguchi H, Tomita K, Kanai Y, Endou H. Modulation of renal apical organic anion transporter 4 function by two PDZ domain‐containing proteins. J Am Soc Nephrol 16: 3498‐3506, 2005.
 249. Moaddel R, Ravichandran S, Bighi F, Yamaguchi R, Wainer IW. Pharmacophore modelling of stereoselective binding to the human organic cation transporter (hOCT1). Br J Pharmacol 151: 1305‐1314, 2007.
 250. Montrose‐Rafizadeh C, Mingard F, Murer H, Roch‐Ramel F. Carrier‐mediated transport of tetraethylammonium across rabbit renal basolateral membrane. Am J Physiol 257: F243‐F251, 1989.
 251. Montrose‐Rafizadeh C, Roch‐Ramel F, Schäli C. Axial heterogeneity of organic cation transport along the rabbit renal proximal tubule: Studies with brush‐border membrane vesicles. Biochim Biophys Acta 904: 175‐177, 1987.
 252. Moseley RH, Jarose SM, Permoad P. Organic cation transport by rat liver plasma membrane vesicles: Studies with tetraethylammonium. Am J Physiol 263: G775‐G785, 1992.
 253. Moseley RH, Morrissette J, Johnson TR. Transport of N1‐methylnicotinamide by organic cation‐proton exchange in rat liver membrane vesicles. Am J Physiol 259: G973‐G982, 1990.
 254. Motohashi H, Sakurai Y, Saito H, Masuda S, Urakami Y, Goto M, Fukatsu A, Ogawa O, Inui K. Gene expression levels and immunolocalization of organic ion transporters in the human kidney. J Am Soc Nephrol 13: 866‐874, 2002.
 255. Mount DB, Kwon CY, Zandi‐Nejad K. Renal urate transport. Rheum Dis Clin North Am 32: 313‐331, vi, 2006.
 256. Nagel G, Volk C, Friedrich T, Ulzheimer JC, Bamberg E, Koepsell H. A reevaluation of substrate specificity of the rat cation transporter rOCT1. J Biol Chem 272: 31953‐31956, 1997.
 257. Nagle MA, Truong DM, Dnyanmote AV, Eraly SA, Wu W, Nigam SK. Analysis of 3‐dimensional systems for developing and mature kidney clarifies the role of OAT1 and OAT3 in antiviral handling. J Biol Chem 286: 243‐251, 2010.
 258. Nakamura T, Yonezawa A, Hashimoto S, Katsura T, Inui KI. Disruption of multidrug and toxin extrusion MATE1 potentiates cisplatin‐induced nephrotoxicity. Biochem Pharmacol 80: 1762‐1767, 2010.
 259. Niemi M. Role of OATP transporters in the disposition of drugs. Pharmacogenomics 8: 787‐802, 2007.
 260. Nies AT, Keppler D. The apical conjugate efflux pump ABCC2 (MRP2). Pflugers Arch 453: 643‐659, 2007.
 261. Nies AT, Koepsell H, Damme K, Schwab M. Organic cation transporters (OCTs, MATEs), in vitro and in vivo evidence for the importance in drug therapy. Handb Exp Pharmacol 201: 105‐167, 2011.
 262. Notenboom S, Miller DS, Kuik LH, Smits P, Russel FG, Masereeuw R. Short‐term exposure of renal proximal tubules to gentamicin increases long‐term multidrug resistance protein 2 (Abcc2) transport function and reduces nephrotoxicant sensitivity. J Pharmacol Exp Ther 315: 912‐920, 2005.
 263. Notenboom S, Miller DS, Smits P, Russel FG, Masereeuw R. Role of NO in endothelin‐regulated drug transport in the renal proximal tubule. Am J Physiol Renal Physiol 282: F458‐F464, 2002.
 264. Notenboom S, Miller DS, Smits P, Russel FG, Masereeuw R. Involvement of guanylyl cyclase and cGMP in the regulation of Mrp2‐mediated transport in the proximal tubule. Am J Physiol Renal Physiol 287: F33‐F38, 2004.
 265. Notenboom S, Wouterse AC, Peters B, Kuik LH, Heemskerk S, Russel FG, Masereeuw R. Increased apical insertion of the multidrug resistance protein 2 (MRP2/ABCC2) in renal proximal tubules following gentamicin exposure. J Pharmacol Exp Ther 318: 1194‐1202, 2006.
 266. O'Donnell MJ, Ianowski JP, Linton SM, Rheault MR. Inorganic and organic anion transport by insect renal epithelia. Biochim Biophys Acta 1618: 194‐206, 2003.
 267. O'Regan S, Meunier FM. Selection and characterization of the choline transport mutation suppressor from Torpedo electric lobe, CTL1. Neurochem Res 28: 551‐555, 2003.
 268. O'Regan S, Traiffort E, Ruat M, Cha N, Compaore D, Meunier FM. An electric lobe suppressor for a yeast choline transport mutation belongs to a new family of transporter‐like proteins. Proc Natl Acad Sci U S A 97: 1835‐1840, 2000.
 269. Ogasawara K, Terada T, Asaka J, Katsura T, Inui K. Human organic anion transporter 3 gene is regulated constitutively and inducibly via a cAMP‐response element. J Pharmacol Exp Ther 319: 317‐322, 2006.
 270. Ogasawara K, Terada T, Asaka J, Katsura T, Inui K. Hepatocyte nuclear factor‐4α regulates the human organic anion transporter 1 gene in the kidney. Am J Physiol Renal Physiol 292: F1819‐F1826, 2007.
 271. Ohashi R, Tamai I, Nezu JJ, Nikaido H, Hashimoto N, Oku A, Sai Y, Shimane M, Tsuji A. Molecular and physiological evidence for multifunctionality of carnitine/organic cation transporter OCTN2. Mol Pharm 59: 358‐366, 2001.
 272. Ohtsu N, Anzai N, Fukutomi T, Kimura T, Sakurai H, Endou H. [Human renal urate transpoter URAT1 mediates the transport of salicylate]. Nippon Jinzo Gakkai Shi 52: 499‐504, 2010.
 273. Okuda M, Saito H, Urakami Y, Takano M, Inui K‐I. cDNA cloning and functional expression of a novel rat kidney organic cation transporter, OCT2. Biochem Biophys Res Comm 224: 500‐507, 1996.
 274. Okuda T, Haga T. High‐affinity choline transporter. Neurochem Res 28: 483‐488, 2003.
 275. Omote H, Hiasa M, Matsumoto T, Otsuka M, Moriyama Y. The MATE proteins as fundamental transporters of metabolic and xenobiotic organic cations. Trends Pharmacol Sci 27: 587‐593, 2006.
 276. Otsuka M, Matsumoto T, Morimoto R, Arioka S, Omote H, Moriyama Y. A human transporter protein that mediates the final excretion step for toxic organic cations. Proc Natl Acad Sci U S A 102: 17923‐17928, 2005.
 277. Otsuka M, Yasuda M, Morita Y, Otsuka C, Tsuchiya T, Omote H, Moriyama Y. Identification of essential amino acid residues of the NorM Na+/multidrug antiporter in Vibrio parahaemolyticus. J Bacteriol 187: 1552‐1558, 2005.
 278. Ott RJ, Hui AC, Yuan G, Giacomini KM. Organic cation transport in human renal brush‐border membrane vesicles. Am J Physiol 261: F443‐F451, 1991.
 279. Palmieri O, Latiano A, Valvano R, D'Inca R, Vecchi M, Sturniolo GC, Saibeni S, Peyvandi F, Bossa F, Zagaria C, Andriulli A, Devoto M, Annese V. Variants of OCTN1‐2 cation transporter genes are associated with both Crohn's disease and ulcerative colitis. Aliment Pharmacol Ther 23: 497‐506, 2006.
 280. Pao SS, Paulsen IT, Saier MH Jr. Major facilitator superfamily. Microbiol Mol Biol Rev 62: 1‐34, 1998.
 281. Paulusma CC, van Geer MA, Evers R, Heijn M, Ottenhoff R, Borst P, Oude Elferink RP. Canalicular multispecific organic anion transporter/multidrug resistance protein 2 mediates low‐affinity transport of reduced glutathione. Biochem J 338: 393‐401, 1999.
 282. Pelis RM, Dangprapai Y, Wunz TM, Wright SH. Inorganic mercury interacts with cysteine residues (C451 and C474) of hOCT2 to reduce its transport activity. Am J Physiol Renal Physiol 292: F1583‐F1591, 2007.
 283. Pelis RM, Hartman RC, Wright SH, Wunz TM, Groves CE. Influence of estrogen and xenoestrogens on basolateral uptake of tetraethylammonium by opossum kidney cells in culture. J Pharmacol Exp Ther 323: 555‐561, 2007.
 284. Pelis RM, Suhre WM, Wright SH. Functional influence of N‐glycosylation in OCT2‐mediated tetraethylammonium transport. Am J Physiol Renal Physiol 290: F1118‐F1126, 2006.
 285. Pelis RM, Zhang X, Dangprapai Y, Wright SH. Cysteine accessibility in the hydrophilic cleft of the human organic cation transporter 2. J Biol Chem 281: 35272‐35280, 2006.
 286. Peltekova VD, Wintle RF, Rubin LA, Amos CI, Huang Q, Gu X, Newman B, Van Oene M, Cescon D, Greenberg G, Griffiths AM, George‐Hyslop PH, Siminovitch KA. Functional variants of OCTN cation transporter genes are associated with Crohn disease. Nat Genet 36: 471‐475, 2004.
 287. Perry JL, Dembla‐Rajpal N, Hall LA, Pritchard JB. A three‐dimensional model of human organic anion transporter 1: Aromatic amino acids required for substrate transport. J Biol Chem 281: 38071‐38079, 2006.
 288. Phillips RA, Hamilton PB. Effect of 20, 60 and 120 minutes of renal ischemia on glomerular and tubular function. Am J Physiol 152, 523‐530, 1948.
 289. Pietig G, Mehrens T, Hirsch JR, Cetinkaya I, Piechota H, Schlatter E. Properties and regulation of organic cation transport in freshly isolated human proximal tubules. J Biol Chem 276: 33741‐33746, 2001.
 290. Polkowski CA, Grassl SM. Uric acid transport in rat renal basolateral vesicles. Biochim Biophys Acta 1146: 145‐152, 1993.
 291. Popowski K, Eloranta JJ, Saborowski M, Fried M, Meier PJ, Kullak‐Ublick GA. The human organic anion transporter 2 gene is transactivated by hepatocyte nuclear factor‐4α and suppressed by bile acids. Mol Pharmacol 67: 1629‐1638, 2005.
 292. Popp C, Gorboulev V, Muller TD, Gorbunov D, Shatskaya N, Koepsell H. Amino acids critical for substrate affinity of rat organic cation transporter 1 line the substrate binding region in a model derived from the tertiary structure of lactose permease. Mol Pharmacol 67: 1600‐1611, 2005.
 293. Pratt S, Chen V, Perry WI III, Starling JJ, Dantzig AH. Kinetic validation of the use of carboxydichlorofluorescein as a drug surrogate for MRP5‐mediated transport. Eur J Pharm Sci 27: 524‐532, 2006.
 294. Pritchard JB. Luminal and peritubular steps in the renal transport of p‐aminohippurate. Biochim Biophys Acta 906: 295‐308, 1987.
 295. Pritchard JB. Coupled transport of p‐aminohippurate by rat kidney basolateral membrane vesicles. Am J Physiol 255: F597‐F604, 1988.
 296. Pritchard JB, Miller DS. Mechanisms mediating renal secretion of organic anions and cations. Physiol Rev 73: 765‐796, 1993.
 297. Pritchard JB, Miller DS. Renal secretion of organic anions and cations. Kidney Int 49: 1649‐1654, 1996.
 298. Qian YM, Grant CE, Westlake CJ, Zhang DW, Lander PA, Shepard RL, Dantzig AH, Cole SP, Deeley RG. Photolabeling of human and murine multidrug resistance protein 1 with the high affinity inhibitor [125I]LY475776 and azidophenacyl‐[35S]glutathione. J Biol Chem 277: 35225‐35231, 2002.
 299. Race JE, Grassl SM, Williams WJ, Holtzman EJ. Molecular cloning and characterization of two novel human renal organic anion transporters (hOAT1 and hOAT3). Biochem Biophys Res Commun 255: 508‐514, 1999.
 300. Rasmussen SN, Shalmi M, Hansen M, Christensen S. Tetraethylammonium and p‐aminohippurate as clearance markers for renal plasma flow in the rat during saline and glucose infusion. Ren Physiol Biochem 13: 314‐323, 1990.
 301. Rau T, Erney B, Gores R, Eschenhagen T, Beck J, Langer T. High‐dose methotrexate in pediatric acute lymphoblastic leukemia: Impact of ABCC2 polymorphisms on plasma concentrations. Clin Pharmacol Ther 80: 468‐476, 2006.
 302. Ravna AW, Sager G. Molecular model of the outward facing state of the human multidrug resistance protein 4 (MRP4/ABCC4). Bioorg Med Chem Lett 18: 3481‐3483, 2008.
 303. Reid G, Wielinga P, Zelcer N, de Haas M, van Deemter L, Wijnholds J, Balzarini J, Borst P. Characterization of the transport of nucleoside analog drugs by the human multidrug resistance proteins MRP4 and MRP5. Mol Pharmacol 63: 1094‐1103, 2003.
 304. Reid G, Wielinga P, Zelcer N, van der Heijden I, Kuil A, de Haas M, Wijnholds J, Borst P. The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc Natl Acad Sci U S A 100: 9244‐9249, 2003.
 305. Reid G, Wolff NA, Dautzenberg FM, Burckhardt G. Cloning of a human renal p‐aminohippurate transporter, hROAT1. Kidney Blood Press Res 21: 233‐237, 1998.
 306. Reidenberg MM, Camacho M, Kluger J, Drayer DE. Aging and renal clearance of procainamide and acetylprocainamide. Clin Pharmacol Ther 28: 732‐735, 1980.
 307. Rennick B, Moe GK. Stop‐flow localization of renal tubular excretion of tetraethylammonium. Am J Physiol 198: 1267‐1270, 1960.
 308. Rius M, Hummel‐Eisenbeiss J, Hofmann AF, Keppler D. Substrate specificity of human ABCC4 (MRP4)‐mediated cotransport of bile acids and reduced glutathione. Am J Physiol Gastrointest Liver Physiol 290: G640‐G649, 2006.
 309. Rius M, Hummel‐Eisenbeiss J, Keppler D. ATP‐dependent transport of leukotrienes B4 and C4 by the multidrug resistance protein ABCC4 (MRP4). J Pharmacol Exp Ther 324: 86‐94, 2008.
 310. Rizwan AN, Burckhardt G. Organic anion transporters of the SLC22 family: Biopharmaceutical, physiological, and pathological roles. Pharm Res 24: 450‐470, 2007.
 311. Rizwan AN, Krick W, Burckhardt G. The chloride dependence of the human organic anion transporter 1 (hOAT1) is blunted by mutation of a single amino acid. J Biol Chem 282: 13402‐13409, 2007.
 312. Roch‐Ramel F, Besseghir K, Murer H. Renal excretion and tubular transport of organic anions and cations. In: Windhager EE, editor. Handbook of Physiology. Section 8. Renal Physiology, Vol. II. New York: Oxford Univ. Press, 1992.
 313. Roch‐Ramel F, Guisan B, Schild L. Indirect coupling of urate and p‐aminohippurate transport to sodium in human brush‐border membrane vesicles. Am J Physiol 270: F61‐F68, 1996.
 314. Roch‐Ramel F, Werner D, Guisan B. Urate transport in brush‐border membrane of human kidney. Am J Physiol 266: F797‐F805, 1994.
 315. Rodvold KA, Paloucek FP, Jung D, Gallastegui J. Interaction of steady‐state procainamide with H2‐receptor antagonists cimetidine and ranitidine. Ther Drug Monit 9: 378‐383, 1987.
 316. Russel FG, Koenderink JB, Masereeuw R. Multidrug resistance protein 4 (MRP4/ABCC4): A versatile efflux transporter for drugs and signalling molecules. Trends Pharmacol Sci 29: 200‐207, 2008.
 317. Russel FG, Masereeuw R, van Aubel RA. Molecular aspects of renal anionic drug transport. Annu Rev Physiol 64: 563‐594, 2002.
 318. Russell RK, Drummond HE, Nimmo ER, Anderson NH, Noble CL, Wilson DC, Gillett PM, McGrogan P, Hassan K, Weaver LT, Bisset WM, Mahdi G, Satsangi J. Analysis of the influence of OCTN1/2 variants within the IBD5 locus on disease susceptibility and growth indices in early onset inflammatory bowel disease. Gut 55: 1114‐1123, 2006.
 319. Ryu S, Kawabe T, Nada S, Yamaguchi A. Identification of basic residues involved in drug export function of human multidrug resistance‐associated protein 2. J Biol Chem 275: 39617‐39624, 2000.
 320. Saito H. Pathophysiological regulation of renal SLC22A organic ion transporters in acute kidney injury: Pharmacological and toxicological implications. Pharmacol Ther 125: 79‐91, 2010.
 321. Saji T, Kikuchi R, Kusuhara H, Kim I, Gonzalez FJ, Sugiyama Y. Transcriptional regulation of human and mouse organic anion transporter 1 by hepatocyte nuclear factor 1 α/β. J Pharmacol Exp Ther 324: 784‐790, 2008.
 322. Sata R, Ohtani H, Tsujimoto M, Murakami H, Koyabu N, Nakamura T, Uchiumi T, Kuwano M, Nagata H, Tsukimori K, Nakano H, Sawada Y. Functional analysis of organic cation transporter 3 expressed in human placenta. J Pharmacol Exp Ther 315: 888‐895, 2005.
 323. Sato M, Mamada H, Anzai N, Shirasaka Y, Nakanishi T, Tamai I. Renal secretion of uric acid by organic anion transporter 2 (OAT2/SLC22A7) in human. Biol Pharm Bull 33: 498‐503, 2010.
 324. Sato T, Masuda S, Yonezawa A, Tanihara Y, Katsura T, Inui K. Transcellular transport of organic cations in double‐transfected MDCK cells expressing human organic cation transporters hOCT1/hMATE1 and hOCT2/hMATE1. Biochem Pharmacol 76: 894‐903, 2008.
 325. Sauvant C, Hesse D, Holzinger H, Evans KK, Dantzler WH, Gekle M. Action of EGF and PGE2 on basolateral organic anion uptake in rabbit proximal renal tubules and hOAT1 expressed in human kidney epithelial cells. Am J Physiol Renal Physiol 286: F774‐F783, 2004.
 326. Sauvant C, Holzinger H, Gekle M. Short‐term regulation of basolateral organic anion uptake in proximal tubular OK cells: EGF acts via MAPK, PLA(2), and COX1. J Am Soc Nephrol 13: 1981‐1991, 2002.
 327. Sauvant C, Holzinger H, Gekle M. Short‐term regulation of basolateral organic anion uptake in proximal tubular OK cells: Protaglandin E2 acts via EP4‐receptor and activation of PKA. J Am Soc Nephrol 14: 3017‐3026, 2003.
 328. Sauvant C, Holzinger H, Gekle M. Prostaglandin E2 inhibits its own renal transport by downregulation of organic anion transporters rOAT1 and rOAT3. J Am Soc Nephrol 17: 46‐53, 2006.
 329. Schäli C, Schild L, Overney J, Roch‐Ramel F. Secretion of tetraethylammonium by proximal tubules of rabbit kidneys. Am J Physiol 245: F238‐F246, 1983.
 330. Schaub TP, Kartenbeck J, König J, Spring H, Dorsam J, Staehler G, Storkel S, Thon WF, Keppler D. Expression of the MRP2 gene‐encoded conjugate export pump in human kidney proximal tubules and in renal cell carcinoma. J Am Soc Nephrol 10: 1159‐1169, 1999.
 331. Schaub TP, Kartenbeck J, König J, Vogel O, Witzgall R, Kriz W, Keppler D. Expression of the conjugate export pump encoded by the mrp2 gene in the apical membrane of kidney proximal tubules. J Am Soc Nephrol 8: 1213‐1221, 1997.
 332. Scheen AJ. New therapeutic approaches in type 2 diabetes. Acta Clin Belg 63: 402‐407, 2008.
 333. Schmitt BM, Gorbunov D, Schlachtbauer P, Egenberger B, Gorboulev V, Wischmeyer E, Muller T, Koepsell H. Charge‐to‐substrate ratio during organic cation uptake by rat OCT2 is voltage dependent and altered by exchange of glutamate 448 with glutamine. Am J Physiol Renal Physiol 296: F709‐F722, 2009.
 334. Schomig E, Lazar A, Grundemann D. Extraneuronal monoamine transporter and organic cation transporters 1 and 2: A review of transport efficiency. Handb Exp Pharmacol 175: 151‐180, 2006.
 335. Schomig E, Spitzenberger F, Engelhardt M, Martel F, Ording N, Gründemann D. Molecular cloning and characterization of two novel transport proteins from rat kidney. FEBS Lett 425: 79‐86, 1998.
 336. Schuetz JD, Connelly MC, Sun D, Paibir SG, Flynn PM, Srinivas RV, Kumar A, Fridland A. MRP4: A previously unidentified factor in resistance to nucleoside‐based antiviral drugs. Nat Med 5: 1048‐1051, 1999.
 337. Sekine T, Watanabe N, Hosoyamada M, Kanaim Y, Endou H. Expression cloning and characterization of a novel multispecific organic anion transporter. J Biol Chem 272: 18526‐18529, 1997.
 338. Seliskar M, Rozman D. Mammalian cytochromes P450–importance of tissue specificity. Biochim Biophys Acta 1770: 458‐466, 2007.
 339. Sheikh MI. Renal handling of phenol red. I. A comparative study on the accumulation of phenol red and p‐aminohippurate in rabbit kidney tubules in vitro. J Physiol 227: 565‐590, 1972.
 340. Shekhawat PS, Srinivas SR, Matern D, Bennett MJ, Boriack R, George V, Xu H, Prasad PD, Roon P, Ganapathy V. Spontaneous development of intestinal and colonic atrophy and inflammation in the carnitine‐deficient jvs (OCTN2−/−) mice. Mol Genet Metab 92: 315‐324, 2007.
 341. Shikata E, Yamamoto R, Takane H, Shigemasa C, Ikeda T, Otsubo K, Ieiri I. Human organic cation transporter (OCT1 and OCT2) gene polymorphisms and therapeutic effects of metformin. J Hum Genet 52: 117‐122, 2007.
 342. Shima JE, Komori T, Taylor TR, Stryke D, Kawamoto M, Johns SJ, Carlson EJ, Ferrin TE, Giacomini KM. Genetic variants of human organic anion transporter 4 demonstrate altered transport of endogenous substrates. Am J Physiol Renal Physiol 299: F767‐F775, 2010.
 343. Shimada H, Moewes B, Burckhardt G. Indirect coupling to Na+ of p‐aminohippuric acid uptake into rat renal basolateral membrane vesicles. Am J Physiol 253: F795‐F801, 1987.
 344. Shu Y, Brown C, Castro RA, Shi RJ, Lin ET, Owen RP, Sheardown SA, Yue L, Burchard EG, Brett CM, Giacomini KM. Effect of genetic variation in the organic cation transporter 1, OCT1, on metformin pharmacokinetics. Clin Pharmacol Ther 83: 273‐280, 2007.
 345. Shu Y, Leabman MK, Feng B, Mangravite LM, Huang CC, Stryke D, Kawamoto M, Johns SJ, DeYoung J, Carlson E, Ferrin TE, Herskowitz I, Giacomini KM and Pharmacogenetics Of Membrane Transporters Investigators. Evolutionary conservation predicts function of variants of the human organic cation transporter, OCT1. Proc Natl Acad Sci U S A 100: 5902‐5907, 2003.
 346. Shu Y, Sheardown SA, Brown C, Owen RP, Zhang S, Castro RA, Ianculescu AG, Yue L, Lo JC, Burchard EG, Brett CM, Giacomini KM. Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action. J Clin Invest 117: 1422‐1431, 2007.
 347. Shuprisha A, Lynch RM, Wright SH, Dantzler WH. PKC regulation of organic anion secretion in perfused S2 segments of rabbit proximal tubules. Am J Physiol 278: F104‐F109, 2000.
 348. Shuprisha A, Wright SH, Dantzler WH. Method for measuring luminal efflux of fluorescent organic compounds in isolated, perfused renal tubules. Am J Physiol Renal Physiol 279: F960‐F964, 2000.
 349. Silverberg MS. OCTNs: Will the real IBD5 gene please stand up? World J Gastroenterol 12: 3678‐3681, 2006.
 350. Slitt AL, Cherrington NJ, Hartley DP, Leazer TM, Klaassen CD. Tissue distribution and renal developmental changes in rat organic cation transporter mRNA levels. Drug Metab Dispos 30: 212‐219, 2002.
 351. Smeets PH, van Aubel RA, Wouterse AC, Van Den Heuvel JJ, Russel FG. Contribution of multidrug resistance protein 2 (MRP2/ABCC2) to the renal excretion of p‐aminohippurate (PAH) and identification of MRP4 (ABCC4) as a novel PAH transporter. J Am Soc Nephrol 15: 2828‐2835, 2004.
 352. Smith PM, Pritchard JB, Miller DS. Membrane potential drives organic cation transport into teleost renal proximal tubules. Am J Physiol 255: R492‐R499, 1988.
 353. Sokol PP, Gates SB. Effect of endogenous and exogenous polyamines on organic cation transport in rabbit renal plasma membrane vesicles. J Pharmacol Exp Ther 255: 52‐58, 1990.
 354. Sokol PP, Holohan PD, Ross CR. Electroneutral transport of organic cations in canine renal brush border membrane vesicles (BBMV). J Pharmacol Exp Ther 233: 694‐699, 1985.
 355. Sokol PP, McKinney TD. Mechanism of organic cation transport in rabbit renal basolateral membrane vesicles. Am J Physiol 258: F1599‐F1607, 1990.
 356. Somogyi A, Bochner F. Dose and concentration dependent effect of ranitidine on procainamide disposition and renal clearance in man. Br J clin Pharmacol 18: 175‐181, 1984.
 357. Somogyi A, Heinzow B. Cimetidine reduces procainamide elimination. New Eng J Med 307: 1080, 1982.
 358. Somogyi A, McLean A, Heinzow B. Cimetidine‐procainamide pharmacokinetic interaction on man: Evidence of competition for tubular secretion of basic drugs. Eur J Clin Pharmacol 25: 339‐345, 1983.
 359. Somogyi A, Stockley C, Keal J, Rolan P, Bochner F. Reduction of metformin renal tubular secretion by cimetidine in man. Br J Clin Pharmacol 23: 545‐551, 1987.
 360. Somogyi AA, Rumrich G, Fritzsch G, Ullrich KJ. Stereospecificity in contraluminal and luminal transporters of organic cations in the rat renal proximal tubule. J Pharmacol Exp Ther 278: 31‐36, 1996.
 361. Song I, Shin H, Shim E, Jung I, Kim W, Shon J, Shin J. Genetic variants of the organic cation transporter 2 influence the disposition of metformin. Clin Pharmacol Ther 84: 559‐562, 2008.
 362. Song IS, Shin HJ, Shin JG. Genetic variants of organic cation transporter 2 (OCT2) significantly reduce metformin uptake in oocytes. Xenobiotica 38: 1252‐1262, 2008.
 363. Soodvilai S, Chatsudthipong A, Chatsudthipong V. Role of MAPK and PKA in regulation of rbOCT2‐mediated renal organic cation transport. Am J Physiol Renal Physiol 293: F21‐F27, 2007.
 364. Soodvilai S, Chatsudthipong V, Evans KK, Wright SH, Dantzler WH. Acute regulation of OAT3‐mediated estrone sulfate transport in isolated rabbit renal proximal tubules. Am J Physiol Renal Physiol 287: F1021‐F1029, 2004.
 365. Srimaroeng C, Perry JL, Pritchard JB. Physiology, structure, and regulation of the cloned organic anion transporters. Xenobiotica 38: 889‐935, 2008.
 366. Suhre WM, Ekins S, Chang C, Swaan PW, Wright SH. Molecular determinants of substrate/inhibitor binding to the human and rabbit renal organic cation transporters, hOCT2 and rbOCT2. Mol Pharmacol 67: 1067‐1077, 2005.
 367. Sweet DH, Chan LM, Walden R, Yang XP, Miller DS, Pritchard JB. Organic anion transporter 3 [Slc22a8] is a dicarboxylate exchanger indirectly coupled to the Na+ gradient. Am J Physiol Renal Physiol 284: F763‐F769, 2003.
 368. Sweet DH, Miller DS, Pritchard JB, Fujiwara Y, Beier DR, Nigam SK. Impaired organic anion transport in kidney and choroid plexus of organic anion transporter 3 (Oat3 [Slc22a8]) knockout mice. J Biol Chem 277: 26934‐26943, 2002.
 369. Sweet DH, Wolff NA, Pritchard JB. Expression cloning and characterization of ROAT1: The renal basolateral organic anion transporter in rat kidney. J Biol Chem 272: 30088‐30095, 1997.
 370. Taggart JV. Protein binding of p‐aminohippurate in human and dog plasma. Am J Physiol 167, 248‐254, 1951.
 371. Takane H, Shikata E, Otsubo K, Higuchi S, Ieiri I. Polymorphism in human organic cation transporters and metformin action. Pharmacogenomics 9: 415‐422, 2008.
 372. Takano M, Inui K‐I, Okano T, Saito H, Hori R. Carrier‐mediated transport systems of tetraethylammonium in rat renal brush‐border and basolateral membrane vesicles. Biochim Biophys Acta 773: 113‐124, 1984.
 373. Takeda M, Khamdang S, Narikawa S, Kimura H, Kobayashi Y, Yamamoto T, Cha SH, Sekine T, Endou H. Human organic anion transporters and human organic cation transporters mediate renal antiviral transport. J Pharmacol Exp Ther 300: 918‐924, 2002.
 374. Takeda M, Sekine T, Endou H. Regulation by protein kinase C of organic anion transport driven by rat organic anion transporter 3 (rOAT3). Life Sci 67: 1087‐1093, 2000.
 375. Tamai I, Nakanishi T, Kobayashi D, China K, Kosugi Y, Nezu J, Sai Y, Tsuji A. Involvement of OCTN1 (SLC22A4) in pH‐dependent transport of organic cations. Mol Pharm 1: 57‐66, 2004.
 376. Tamai I, Ohashi R, Nezu J, Yabuuchi H, Oku A, Shimane M, Sai Y, Tsuji A. Molecular and functional identification of sodium ion‐dependent, high affinity human carnitine transporter OCTN2. J Biol Chem 273: 20378‐20382, 1998.
 377. Tamai I, Yabuuchi H, Nezu J‐I, Sai Y, Oku A, Shimane M, Tsuji A. Cloning and characterization of a novel human pH‐dependent organic cation transporter, OCTN1. FEBS Lett 419: 107‐111, 1997.
 378. Tanaka K, Xu W, Zhou F, You G. Role of glycosylation in the organic anion transporter OAT1. J Biol Chem 279: 14961‐14966, 2004.
 379. Tanaka K, Zhou F, Kuze K, You G. Cysteine residues in the organic anion transporter mOAT1. Biochem J 380: 283‐287, 2004.
 380. Tang NL, Ganapathy V, Wu X, Hui J, Seth P, Yuen PM, Wanders RJ, Fok TF, Hjelm NM. Mutations of OCTN2, an organic cation/carnitine transporter, lead to deficient cellular carnitine uptake in primary carnitine deficiency. Hum Mol Genet 8: 655‐660, 1999.
 381. Taniguchi A, Kamatani N. Control of renal uric acid excretion and gout. Curr Opin Rheumatol 20: 192‐197, 2008.
 382. Taniguchi A, Urano W, Yamanaka M, Yamanaka H, Hosoyamada M, Endou H, Kamatani N. A common mutation in an organic anion transporter gene, SLC22A12, is a suppressing factor for the development of gout. Arthritis Rheum 52: 2576‐2577, 2005.
 383. Taniguchi K, Wada M, Kohno K, Nakamura T, Kawabe T, Kawakami M, Kagotani K, Okumura K, Akiyama S, Kuwano M. A human canalicular multispecific organic anion transporter (cMOAT) gene is overexpressed in cisplatin‐resistant human cancer cell lines with decreased drug accumulation. Cancer Res 56: 4124‐4129, 1996.
 384. Tanihara Y, Masuda S, Katsura T, Inui K. Protective effect of concomitant administration of imatinib on cisplatin‐induced nephrotoxicity focusing on renal organic cation transporter OCT2. Biochem Pharmacol 78: 1263‐1271, 2009.
 385. Tanihara Y, Masuda S, Sato T, Katsura T, Ogawa O, Inui KI. Substrate specificity of MATE1 and MATE2‐K, human multidrug and toxin extrusions/H+‐organic cation antiporters. Biochem Pharmacol 74: 359‐371, 2007.
 386. Tein I. Carnitine transport: Pathophysiology and metabolism of known molecular defects. J Inherit Metab Dis 26: 147‐169, 2003.
 387. Terada T, Inui K. Physiological and pharmacokinetic roles of H+/organic cation antiporters (MATE/SLC47A). Biochem Pharmacol 75: 1689‐1696, 2008.
 388. Terada T, Masuda S, Asaka JI, Tsuda M, Katsura T, Inui KI. Molecular cloning, functional characterization and tissue distribution of rat H(+)/organic cation antiporter MATE1. Pharm Res 23: 1696‐1701, 2006.
 389. Terlouw SA, Graeff C, Smeets PH, Fricker G, Russel FG, Masereeuw R, Miller DS. Short‐ and long‐term influences of heavy metals on anionic drug efflux from renal proximal tubule. J Pharmacol Exp Ther 301: 578‐585, 2002.
 390. Terlouw SA, Masereeuw R, Russel FG, Miller DS. Nephrotoxicants induce endothelin release and signaling in renal proximal tubules: Effect on drug efflux. Mol Pharmacol 59: 1433‐1440, 2001.
 391. Terlouw SA, Masereeuw R, Van Den Broek PH, Notenboom S, Russel FG. Role of multidrug resistance protein 2 (MRP2) in glutathione‐bimane efflux from Caco‐2 and rat renal proximal tubule cells. Br J Pharmacol 134: 931‐938, 2001.
 392. Thangaraju M, Ananth S, Martin PM, Roon P, Smith SB, Sterneck E, Prasad PD, Ganapathy V. c/ebpδ Null mouse as a model for the double knock‐out of slc5a8 and slc5a12 in kidney. J Biol Chem 281: 26769‐26773, 2006.
 393. Thiebaut F, Tsuruo T, Hamada H, Gottesman MM, Pastan I. Cellular localization of multidrug resistance gene product P‐glycoprotein in normal human tissues. Proc Natl Acad Sci U S A 84: 7735‐7738, 1987.
 394. Tian Q, Zhang J, Tan TM, Chan E, Duan W, Chan SY, Boelsterli UA, Ho PC, Yang H, Bian JS, Huang M, Zhu YZ, Xiong W, Li X, Zhou S. Human multidrug resistance associated protein 4 confers resistance to camptothecins. Pharm Res 22: 1837‐1853, 2005.
 395. Toh S, Wada M, Uchiumi T, Inokuchi A, Makino Y, Horie Y, Adachi Y, Sakisaka S, Kuwano M. Genomic structure of the canalicular multispecific organic anion‐transporter gene (MRP2/cMOAT) and mutations in the ATP‐binding‐cassette region in Dubin‐Johnson syndrome. Am J Hum Genet 64: 739‐746, 1999.
 396. Tsuda M, Terada T, Asaka J, Ueba M, Katsura T, Inui K. Oppositely directed H+ gradient functions as a driving force of rat H+/organic cation antiporter MATE1. Am J Physiol Renal Physiol 292: F593‐F598, 2007.
 397. Tsuda M, Terada T, Mizuno T, Katsura T, Shimakura J, Inui KI. Targeted disruption of the multidrug and toxin extrusion 1 (Mate1) gene in mice reduces renal secretion of metformin. Mol Pharmacol 75: 1280‐1286, 2009.
 398. Tsuda M, Terada T, Ueba M, Sato T, Masuda S, Katsura T, Inui K. Involvement of human multidrug and toxin extrusion 1 in the drug interaction between cimetidine and metformin in renal epithelial cells. J Pharmacol Exp Ther 329: 185‐191, 2009.
 399. Tsujii H, König J, Rost D, Stockel B, Leuschner U, Keppler D. Exon‐intron organization of the human multidrug‐resistance protein 2 (MRP2) gene mutated in Dubin‐Johnson syndrome. Gastroenterology 117: 653‐660, 1999.
 400. Tzvetkov MV, Vormfelde SV, Balen D, Meineke I, Schmidt T, Sehrt D, Sabolic I, Koepsell H, Brockmoller J. The effects of genetic polymorphisms in the organic cation transporters OCT1, OCT2, and OCT3 on the renal clearance of metformin. Clin Pharmacol Ther 86: 299‐306, 2009.
 401. Uchino H, Tamai I, Yamashita K, Minemoto Y, Sai Y, Yabuuchi H, Miyamoto K, Takeda E, Tsuji A. p‐Aminohippuric acid transport at renal apical membrane mediated by human inorganic phosphate transporter NPT1. Biochem Biophys Res Commun 270: 254‐259, 2000.
 402. Ullrich KJ. Renal transporters for organic anions and cations. Structural requirements for substrates. J Membrane Biol 158: 95‐107, 1997.
 403. Ullrich KJ, Fritzsch G, Rumrich G, David C. Polysubstrates: Substances that interact with renal contraluminal PAH, sulfate, and NMeN transport: Sulfamoyl‐, sulfonylurea‐, thiazide‐ and benzeneamino‐carboxylate (nicotinate) compounds. J Pharmacol Exp Ther 269: 684‐692, 1994.
 404. Ullrich KJ, Papavassiliou F, David C, Rumrich G, Fritzsch G. Contraluminal transport of organic cations in the proximal tubule of the rat kidney. I. Kinetics of N1‐methylnicotinamide and tetraethylammonium, influence of K+, HCO3, pH; inhibition by aliphatic primary, secondary and tertiary amines, and mono‐ and bisquaternary compounds. Pflugers Arch 419: 84‐92, 1991.
 405. Ullrich KJ, Rumrich G. Morphine analogues: Relationship between chemical structure and interaction with proximal tubular transporters – contraluminal organic cation and anion transporter, luminal H+/organic cation exchanger, and luminal choline transporter. Cell Physiol Biochem 5: 290‐298, 1995.
 406. Ullrich KJ, Rumrich G. Luminal transport system for choline+ in relation to the other organic cation transport systems in the rat proximal tubule. Kinetics, specificity: Alkyl/arylamines, alkylamines with OH, O, SH, NH2, ROCO, RSCO and H2PO4‐groups, methylaminostyryl, rhodamine, acridine, phenanthrene and cyanine compounds. Pflügers Arch 432: 471‐485, 1996.
 407. Ullrich KJ, Rumrich G, David C, Fritzsch G. Bisubstrates: Substances that interact with both, renal contraluminal organic anion and organic cation transport systems. II. Zwitterionic substrates: Dipeptides, cephalosporins, quinolone‐carboxylate gyrase inhibitors and phosphoamide thiazine carboxylates; nonionizable substrates: Steroid hormones and cyclophosphamides. Pflugers Arch 425: 300‐312, 1993a.
 408. Ullrich KJ, Rumrich G, David C, Fritzsch G. Bisubstrates: Substances that interact with renal organic anion and organic cation transport systems. I. Amines, piperidines, piperazines, azepines, pyridines, quinolines, imadazoles, thiazoles, guanidines, and hydrazines. Pflugers Arch 425: 280‐299, 1993b.
 409. Ullrich KJ, Rumrich G, Fritzsch G. Contraluminal transport of organic cations in the proximal tubule of the rat kidney. II. Specificity: Anilines, phenylalkylamines (catecholamines), heterocyclic compounds (pyridines, quinolines, acridines). Pflugers Arch 420: 29‐38, 1992.
 410. Urakami Y, Akazawa M, Saito H, Okuda M, Inui K. cDNA cloning, functional characterization, and tissue distribution of an alternatively spliced variant of organic cation transporter hOCT2 Predominantly Expressed in the Human Kidney. J Am Soc Nephrol 13: 1703‐1710, 2002.
 411. Urakami Y, Nakamura N, Takahashi K, Okuda M, Saito H, Hashimoto Y, Inui K. Gender differences in expression of organic cation transporter OCT2 in rat kidney. FEBS Lett 461: 339‐342, 1999.
 412. Urakami Y, Okuda M, Saito H, Inui K. Hormonal regulation of organic cation transporter OCT2 expression in rat kidney. FEBS Lett 473: 173‐176, 2000.
 413. Urano W, Taniguchi A, Anzai N, Inoue E, Kanai Y, Yamanaka M, Endou H, Kamatani N, Yamanaka H. Sodium‐dependent phosphate cotransporter type 1 sequence polymorphisms in male patients with gout. Ann Rheum Dis 69: 1232‐1234, 2010.
 414. van Aubel RA, Koenderink JB, Peters JG, van Os CH, Russel FG. Mechanisms and interaction of vinblastine and reduced glutathione transport in membrane vesicles by the rabbit multidrug resistance protein MRP2 expressed in insect cells. Mol Pharmacol 56: 714‐719, 1999.
 415. van Aubel RA, Peters JG, Masereeuw R, van Os CH, Russel FG. Multidrug resistance protein MRP2 mediates ATP‐dependent transport of classic renal organic anion p‐aminohippurate. Am J Physiol Renal Physiol 279: F713‐F717, 2000.
 416. van Aubel RA, Smeets PH, Peters JG, Bindels RJ, Russel FG. The MRP4/ABCC4 gene encodes a novel apical organic anion transporter in human kidney proximal tubules: Putative efflux pump for urinary cAMP and cGMP. J Am Soc Nephrol 13: 595‐603, 2002.
 417. van Aubel RA, Smeets PH, Van Den Heuvel JJ, Russel FG. Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate binding sites. Am J Physiol Renal Physiol 288: F327‐F333, 2005.
 418. van Aubel RAMH, van Kuijck MA, Koenderink JB, Deen PMT, van Os CH, Russel FGM. Adenosine triphosphate‐dependent transport of anionic conjugates by the rabbit multidrug resistance‐associated protein MRP2 expressed in insect cells. Mol Pharm 53: 1062‐1067, 1998.
 419. van de Water FM, Masereeuw R, Russel FG. Function and regulation of multidrug resistance proteins (MRPs) in the renal elimination of organic anions. Drug Metab Rev 37: 443‐471, 2005.
 420. van Montfoort JE, Muller M, Groothuis GM, Meijer DK, Koepsell H, Meier PJ. Comparison of “type I” and “type II” organic cation transport by organic cation transporters and organic anion‐transporting polypeptides. J Pharmacol Exp Ther 298: 110‐115, 2001.
 421. Vanwert AL, Gionfriddo MR, Sweet DH. Organic anion transporters: Discovery, pharmacology, regulation and roles in pathophysiology. Biopharm Drug Dispos 31: 1‐71, 2010.
 422. Vardy E, Arkin IT, Gottschalk KE, Kaback HR, Schuldiner S. Structural conservation in the major facilitator superfamily as revealed by comparative modeling. Protein Sci 13: 1832‐1840, 2004.
 423. Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CN, Knott SA, Kolcic I, Polasek O, Graessler J, Wilson JF, Marinaki A, Riches PL, Shu X, Janicijevic B, Smolej‐Narancic N, Gorgoni B, Morgan J, Campbell S, Biloglav Z, Barac‐Lauc L, Pericic M, Klaric IM, Zgaga L, Skaric‐Juric T, Wild SH, Richardson WA, Hohenstein P, Kimber CH, Tenesa A, Donnelly LA, Fairbanks LD, Aringer M, McKeigue PM, Ralston SH, Morris AD, Rudan P, Hastie ND, Campbell H, Wright AF. SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nat Genet 40: 437‐442, 2008.
 424. Volk C, Gorboulev V, Budiman T, Nagel G, Koepsell H. Different affinities of inhibitors to the outwardly and inwardly directed substrate binding site of organic cation transporter 2. Mol Pharmacol 64: 1037‐1047, 2003.
 425. Wagner CA, Lukewille U, Kaltenbach S, Moschen I, Broer A, Risler T, Broer S, Lang F. Functional and pharmacological characterization of human Na+‐carnitine cotransporter hOCTN2. Am J Physiol Renal Physiol 279: F584‐F591, 2000.
 426. Wang Y, Ye J, Ganapathy V, Longo N. Mutations in the organic cation/carnitine transporter OCTN2 in primary carnitine deficiency. Proc Natl Acad Sci U S A 96: 2356‐2360, 1999.
 427. Wang ZJ, Yin OQ, Tomlinson B, Chow MS. OCT2 polymorphisms and in‐vivo renal functional consequence: Studies with metformin and cimetidine. Pharmacogenet Genomics 18: 637‐645, 2008.
 428. Watanabe S, Tsuda M, Terada T, Katsura T, Inui KI. Reduced renal clearance of a zwitterionic substrate cephalexin in MATE1‐deficient mice. J Pharmacol Exp Ther 334: 651‐656, 2010.
 429. Weiner IM. Urate transport in the nephron. Am J Physiol 237: F85‐F92, 1979.
 430. Welborn JR, Shpun S, Dantzler WH, Wright SH. Effect of a‐ketoglutarate on organic anion transport in single rabbit renal proximal tubules. Am J Physiol 274: F165‐F174, 1998.
 431. Westlake CJ, Cole SP, Deeley RG. Role of the NH2‐terminal membrane spanning domain of multidrug resistance protein 1/ABCC1 in protein processing and trafficking. Mol Biol Cell 16: 2483‐2492, 2005.
 432. Westlake CJ, Qian YM, Gao M, Vasa M, Cole SP, Deeley RG. Identification of the structural and functional boundaries of the multidrug resistance protein 1 cytoplasmic loop 3. Biochemistry 42: 14099‐14113, 2003.
 433. Wever KE, Masereeuw R, Miller DS, Hang XM, Flik G. Endothelin and calciotropic hormones share regulatory pathways in multidrug resistance protein 2‐mediated transport. Am J Physiol Renal Physiol 292: F38‐F46, 2007.
 434. Wielinga PR, van der Heijden I, Reid G, Beijnen JH, Wijnholds J, Borst P. Characterization of the MRP4‐ and MRP5‐mediated transport of cyclic nucleotides from intact cells. J Biol Chem 278: 17664‐17671, 2003.
 435. Wilde S, Schlatter E, Koepsell H, Edemir B, Reuter S, Pavenstadt H, Neugebauer U, Schroter R, Brast S, Ciarimboli G. Calmodulin‐associated post‐translational regulation of rat organic cation transporter 2 in the kidney is gender dependent. Cell Mol Life Sci 66: 1729‐1740, 2009.
 436. Wolff NA, Grunwald B, Friedrich B, Lang F, Godehardt S, Burckhardt G. Cationic amino acids involved in dicarboxylate binding of the flounder renal organic anion transporter. J Am Soc Nephrol 12: 2012‐2018, 2001.
 437. Wolff NA, Thies K, Kuhnke N, Reid G, Friedrich B, Lang F, Burckhardt G. Protein kinase C activation downregulates human organic anion transporter 1‐mediated transport through carrier internalization. J Am Soc Nephrol 14: 1959‐1968, 2003.
 438. Wright AF, Rudan I, Hastie ND, Campbell H. A ‘complexity’ of urate transporters. Kidney Int 78: 446‐452, 2010.
 439. Wright SH. Transport of N1‐methylnicotinamide across brush border membrane vesicles from rabbit kidney. Am J Physiol 249: F903‐F911, 1985.
 440. Wright SH, Dantzler WH. Molecular and cellular physiology of renal organic cation and anion transport. Physiol Rev 84: 987‐1049, 2004.
 441. Wright SH, Evans KK, Zhang X, Cherrington NJ, Sitar DS, Dantzler WH. Functional map of TEA transport activity in isolated rabbit renal proximal tubules. Am J Physiol Renal Physiol 287: F442‐F451, 2004.
 442. Wright SH, Wunz TM. Transport of tetraethylammonium by rabbit renal brush‐border and basolateral membrane vesicles. Am J Physiol 253: F1040‐F1050, 1987.
 443. Wright SH, Wunz TM. Influence of substrate structure on turnover of the renal organic cation/H+ exchanger of the renal luminal membrane. Pflügers Arch 436: 469‐477, 1998.
 444. Wright SH, Wunz TM. Influence of substrate structure on substrate binding to the renal organic cation/H+ exchanger. Pflügers Arch 437: 603‐610, 1999.
 445. Wright SH, Wunz TM, Wunz TP. A choline transporter in renal brush‐border membrane vesicles: Energetics and structural specificity. J Membr Biol 126: 51‐65, 1992.
 446. Wright SH, Wunz TM, Wunz TP. Structure and interaction of inhibitors with the TEA/H+ exchanger of rabbit renal brush border membranes. Pflugers Arch 429: 313‐324, 1995.
 447. Wu X, George RL, Huang W, Wang H, Conway SJ, Leibach FH, Ganapathy V. Structural and functional characteristics and tissue distribution pattern of rat OCTN1, an organic cation transporter, cloned from placenta. Biochim Biophys Acta 1466: 315‐327, 2000.
 448. Wu X, Huang W, Ganapathy ME, Wang H, Kekuda R, Conway SJ, Leibach FH, Ganapathy V. Structure, function, and regional distribution of the organic cation transporter OCT3 in the kidney. Am J Physiol Renal Physiol 279: F449‐F458, 2000.
 449. Wu X, Prasad PD, Leibach FH, Ganapathy V. cDNA sequence, transport function, and genomic organization of human OCTN2, a new member of the organic cation transporter family. Biochem Biophys Res Commun 246: 589‐595, 1998.
 450. Wu XW, Muzny DM, Lee CC, Caskey CT. Two independent mutational events in the loss of urate oxidase during hominoid evolution. J Mol Evol 34: 78‐84, 1992.
 451. Xu W, Tanaka K, Sun AQ, You G. Functional role of the C terminus of human organic anion transporter hOAT1. J Biol Chem 281: 31178‐31183, 2006.
 452. Yabuki M, Inazu M, Yamada T, Tajima H, Matsumiya T. Molecular and functional characterization of choline transporter in rat renal tubule epithelial NRK‐52E cells. Arch Biochem Biophys 485: 88‐96, 2009.
 453. Yabuuchi H, Tamai I, Nezu JI, Sakamoto K, Oku A, Shimane M, Sai Y, Tsuji A. Novel membrane transporter OCTN1 mediates multispecific, bidirectional, and pH‐dependent transport of organic cations. J Pharmacol Exp Ther 289: 768‐773, 1999.
 454. Yang CH, Glover KP, Han X. Characterization of cellular uptake of perfluorooctanoate via organic anion‐transporting polypeptide 1A2, organic anion transporter 4, and urate transporter 1 for their potential roles in mediating human renal reabsorption of perfluorocarboxylates. Toxicol Sci 117: 294‐302, 2010.
 455. Yokoo S, Yonezawa A, Masuda S, Fukatsu A, Katsura T, Inui K. Differential contribution of organic cation transporters, OCT2 and MATE1, in platinum agent‐induced nephrotoxicity. Biochem Pharmacol 74: 477‐487, 2007.
 456. Yoshitomi K, Frömter E. Cell pH of rat renal proximal tubule in vivo and the conductive nature of peritubular HCO3− (OH−) exit. Pflugers Arch 402: 300‐305, 1984.
 457. You G, Kuze K, Kohanski RA, Amsler K, Henderson S. Regulation of mOAT‐mediated organic anion transport by okadaic acid and protein kinase C in LLC‐PK(1) cells. J Biol Chem 275: 10278‐10284, 2000.
 458. Yuan Z, Tie A, Tarnopolsky M, Bakovic M. Genomic organization, promoter activity, and expression of the human choline transporter‐like protein 1. Physiol Genomics 26: 76‐90, 2006.
 459. Zaman GJ, Lankelma J, van TO, Beijnen J, Dekker H, Paulusma C, Oude Elferink RP, Baas F, Borst P. Role of glutathione in the export of compounds from cells by the multidrug‐resistance‐associated protein. Proc Natl Acad Sci U S A 92: 7690‐7694, 1995.
 460. Zelcer N, Huisman MT, Reid G, Wielinga P, Breedveld P, Kuil A, Knipscheer P, Schellens JHM, Schinkel AH, Borst P. Evidence for two interacting ligand binding sites in human multidrug resistance protein 2 (ATP binding cassette C2). J Biol Chem 278: 23538‐23544, 2003.
 461. Zelcer N, Reid G, Wielinga P, Kuil A, Van Der Heijden I, Schuetz J, Borst P. Steroid and bile acid conjugates are substrates of human multidrug‐resistance protein (MRP) 4 (ATP‐binding cassette C4). Biochem J 371: 361‐367, 2003.
 462. Zhang L, Dresser MJ, Chun JK, Babbitt PC, Giacomini KM. Cloning and functional characterization of a rat renal organic cation transporter isoform (rOCT1A). J Biol Chem 272: 16548‐16554, 1997.
 463. Zhang L, Gorset W, Dresser MJ, Giacomini KM. The interaction of n‐tetraalkylammonium compounds with a human organic cation transporter, hOCT1. J Pharmacol Exp Ther 288: 1192‐1198, 1999.
 464. Zhang L, Gorset W, Washington CB, Blaschke TF, Kroetz DL, Giacomini KM. Interactions of HIV protease inhibitors with a human organic cation transporter in a mammalian expression system. Drug Metab Dispos 28: 329‐334, 2000.
 465. Zhang L, Schaner ME, Giacomini KM. Functional characterization of an organic cation transporter (hOCT1) in a transiently transfected human cell line (HeLa). J Pharmacol Exp Ther 286: 354‐361, 1998.
 466. Zhang P, Tian X, Chandra P, Brouwer KL. Role of glycosylation in trafficking of MRP2 in sandwich‐cultured rat hepatocytes. Mol Pharmacol 67: 1334‐1341, 2005.
 467. Zhang X, Cherrington NJ, Wright SH. Molecular Identification and functional characterization of rabbit MATE1 and MATE2‐K. Am J Physiol Renal Physiol 293: F360, 2007.
 468. Zhang X, Groves CE, Bahn A, Barendt WM, Prado MD, Rödiger M, Chatsudthipong V, Burckhardt G, Wright SH. Relative contribution of OAT and OCT transporters to organic electrolyte transport in rabbit proximal tubule. Am J Physiol Renal Physiol 287: F999‐F1010, 2004.
 469. Zhang X, Shirahatti NV, Mahadevan D, Wright SH. A conserved glutamate residue in transmembrane helix 10 influences substrate specificity of rabbit OCT2 (SLC22A2). J Biol Chem 280: 34813‐34822, 2005.
 470. Zhang X, Wright SH. MATE1 has an external COOH terminus, consistent with a 13‐helix topology. Am J Physiol Renal Physiol 297: F263‐F271, 2009.
 471. Zhao X, Imig JD. Kidney CYP450 enzymes: Biological actions beyond drug metabolism. Curr Drug Metab 4: 73‐84, 2003.
 472. Zhou F, You G. Molecular insights into the structure‐function relationship of organic anion transporters OATs. Pharm Res 24: 28‐36, 2007.
 473. Zimmerman WB, Byun E, McKinney TD, Sokol PP. Sulfhydryl groups are essential for organic cation exchange in rabbit renal basolateral membrane vesicles. J Biol Chem 266: 5459‐5463, 1991.
 474. Zlender V, Breljak D, Ljubojevic M, Flajs D, Balen D, Brzica H, Domijan AM, Peraica M, Fuchs R, Anzai N, Sabolic I. Low doses of ochratoxin A upregulate the protein expression of organic anion transporters Oat1, Oat2, Oat3 and Oat5 in rat kidney cortex. Toxicol Appl Pharmacol 239: 284‐296, 2009.
 475. Zolk O. Current understanding of the pharmacogenomics of metformin. Clin Pharmacol Ther 86: 595‐598, 2009.
 476. Zolk O, Solbach TF, Konig J, Fromm MF. Structural determinants of inhibitor interaction with the human organic cation transporter OCT2 (SLC22A2). Naunyn Schmiedebergs Arch Pharmacol 379: 337‐348, 2008.
 477. Zolk O, Solbach TF, Konig J, Fromm MF. Functional characterization of the human organic cation transporter 2 variant p.270Ala>Ser. Drug Metab Dispos 37: 1312‐1318, 2009.
 478. Zwart R, Verhaagh S, Buitelaar M, Popp‐Snijders C, Barlow DP. Impaired activity of the extraneuronal monoamine transporter system known as uptake‐2 in Orct3/Slc22a3‐deficient mice. Mol Cell Biol 21: 4188‐4196, 2001.

Related Articles:

Hepatic Transport of Organic Solutes
Transport and Metabolism in the Hepatobiliary System
Renal Plasma Membranes: Isolation, General Properties, and Biochemical Components
Epithelial Transport
Proximal Nephron

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Ryan M. Pelis, Stephen H. Wright. Renal Transport of Organic Anions and Cations. Compr Physiol 2011, 1: 1795-1835. doi: 10.1002/cphy.c100084