99 results on '"Gorboulev V"'
Search Results
2. Nucleoside transporters and human organic cation transporter 1 determine the cellular handling of DNA-methyltransferase inhibitors
- Author
-
Arimany-Nardi, C, Errasti-Murugarren, E, Minuesa, G, Martinez-Picado, J, Gorboulev, V, Koepsell, H, and Pastor-Anglada, M
- Published
- 2014
- Full Text
- View/download PDF
3. Downregulation of the Na+-D-glucose Cotransporter SGLT1 by Protein RS1 (RSC1A1) is Dependent on Dynamin and Protein Kinase C
- Author
-
Veyhl, M., Wagner, C. A., Gorboulev, V., Schmitt, B. M., Lang, F., and Koepsell, H.
- Published
- 2003
- Full Text
- View/download PDF
4. Molecular Pharmacology of Organic Cation Transporters in Kidney
- Author
-
Koepsell, H., Gorboulev, V., and Arndt, P.
- Published
- 1999
- Full Text
- View/download PDF
5. Phosphorylation of RS1 (RSC1A1) Steers Inhibition of Different Exocytotic Pathways for Glucose Transporter SGLT1 and Nucleoside Transporter CNT1, and an RS1-Derived Peptide Inhibits Glucose Absorption
- Author
-
Veyhl-Wichmann M, Friedrich A, Vernaleken A, Singh S, Kipp H, Gorboulev V, Keller T, Chintalapati C, Pipkorn R, Pastor-Anglada M, Groll J, and Koepsell H
- Published
- 2016
6. Roct1, a rat polyspecific transporter gene for the excretion of cationic drugs, maps to Chromosome 1q11-12
- Author
-
Koehler, M. R., Gorboulev, V., Koepsell, H., Steinlein, C., and Schmid, M.
- Published
- 1996
- Full Text
- View/download PDF
7. Erratum
- Author
-
Koehler, M. R., Gorboulev, V., Koepsell, H., Steinlein, C., and Schmid, M.
- Published
- 1996
- Full Text
- View/download PDF
8. Transport of lamivudine [(-)-beta-L-2',3'-dideoxy-3'-thiacytidine] and high-affinity interaction of nucleoside reverse transcriptase inhibitors with human organic cation transporters 1, 2, and 3
- Author
-
Minuesa G, Volk C, Molina-Arcas M, Gorboulev V, Itziar Erkizia, Arndt P, Clotet B, Pastor-Anglada M, Koepsell H, and Martinez-Picado J
- Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) need to enter cells to act against the HIV-1. Human organic cation transporters (hOCT1-3) are expressed and active in CD4+ T cells, the main target of HIV-1, and have been associated with antiviral uptake in different tissues. In this study, we examined whether NRTIs interact and are substrates of hOCT in cells stably expressing these transporters. Using [(3)H]N-methyl-4-phenylpyridinium, we found a high-affinity interaction among abacavir [[(1S,4R)-4-[2-amino-6-(cyclopropylamino)purin-9-yl]-cyclopent-2-enyl]methanol sulfate] (ABC)
- Published
- 2009
9. Organic cation transporters
- Author
-
Koepsell, H., primary, Schmitt, B. M., additional, and Gorboulev, V., additional
- Full Text
- View/download PDF
10. Downregulation of the Na + -D-glucose Cotransporter SGLT1 by Protein RS1 (RSC1A1) is Dependent on Dynamin and Protein Kinase C
- Author
-
Veyhl, M., primary, Wagner, C. A., additional, Gorboulev, V., additional, Schmitt, B. M., additional, Lang, F., additional, and Koepsell, H., additional
- Published
- 2003
- Full Text
- View/download PDF
11. Organic cation transporter capable of transporting serotonin is up-regulated in serotonin transporter-deficient mice
- Author
-
Schmitt, A., primary, Mössner, R., additional, Gossmann, A., additional, Fischer, I.G., additional, Gorboulev, V., additional, Murphy, D.L., additional, Koepsell, H., additional, and Lesch, K.P., additional
- Published
- 2002
- Full Text
- View/download PDF
12. Polyspecific cation transporters in the proximal tubule
- Author
-
Koepsell, H., primary, Gorboulev, V., additional, Karbach, U., additional, and Arndt, P., additional
- Published
- 2000
- Full Text
- View/download PDF
13. Molecular pharmacology of organic cation transport
- Author
-
Koepsell, H., primary, Gorboulev, V., additional, Volk, C., additional, and Arndt, P., additional
- Published
- 2000
- Full Text
- View/download PDF
14. The two human organic cation transporter genes SLC22A1 and SLC22A2 are located on chromosome 6q26
- Author
-
Koehler, M.R., primary, Wissinger, B., additional, Gorboulev, V., additional, Koepsell, H., additional, and Schmid, M., additional
- Published
- 1997
- Full Text
- View/download PDF
15. Monoamine neurotransmitter transport mediated by the polyspecific cation transporter rOCT1
- Author
-
Busch, A.E, primary, Quester, S, additional, Ulzheimer, J.C, additional, Gorboulev, V, additional, Akhoundova, A, additional, Waldegger, S, additional, Lang, F, additional, and Koepsell, H, additional
- Published
- 1996
- Full Text
- View/download PDF
16. Na(+)-D-glucose cotransporter SGLT1 is pivotal for intestinal glucose absorption and glucose-dependent incretin secretion.
- Author
-
Gorboulev V, Schürmann A, Vallon V, Kipp H, Jaschke A, Klessen D, Friedrich A, Scherneck S, Rieg T, Cunard R, Veyhl-Wichmann M, Srinivasan A, Balen D, Breljak D, Rexhepaj R, Parker HE, Gribble FM, Reimann F, Lang F, and Wiese S
- Abstract
To clarify the physiological role of Na(+)-D-glucose cotransporter SGLT1 in small intestine and kidney, Sglt1(-/-) mice were generated and characterized phenotypically. After gavage of d-glucose, small intestinal glucose absorption across the brush-border membrane (BBM) via SGLT1 and GLUT2 were analyzed. Glucose-induced secretion of insulinotropic hormone (GIP) and glucagon-like peptide 1 (GLP-1) in wild-type and Sglt1(-/-) mice were compared. The impact of SGLT1 on renal glucose handling was investigated by micropuncture studies. It was observed that Sglt1(-/-) mice developed a glucose-galactose malabsorption syndrome but thrive normally when fed a glucose-galactose-free diet. In wild-type mice, passage of D-glucose across the intestinal BBM was predominantly mediated by SGLT1, independent the glucose load. High glucose concentrations increased the amounts of SGLT1 and GLUT2 in the BBM, and SGLT1 was required for upregulation of GLUT2. SGLT1 was located in luminal membranes of cells immunopositive for GIP and GLP-1, and Sglt1(-/-) mice exhibited reduced glucose-triggered GIP and GLP-1 levels. In the kidney, SGLT1 reabsorbed ∼3% of the filtered glucose under normoglycemic conditions. The data indicate that SGLT1 is 1) pivotal for intestinal mass absorption of d-glucose, 2) triggers the glucose-induced secretion of GIP and GLP-1, and 3) triggers the upregulation of GLUT2. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
17. Organic cation transporters.
- Author
-
Koepsell, H., Schmitt, B. M., and Gorboulev, V.
- Abstract
Over the last 15 years, a number of transporters that translocate organic cations were characterized functionally and also identified on the molecular level. Organic cations include endogenous compounds such as monoamine neurotransmitters, choline, and coenzymes, but also numerous drugs and xenobiotics. Some of the cloned organic cation transporters accept one main substrate or structurally similar compounds (oligospecific transporters), while others translocate a variety of structurally diverse organic cations (polyspecific transporters). This review provides a survey of cloned organic cation transporters and tentative models that illustrate how different types of organic cation transporters, expressed at specific subcellular sites in hepatocytes and renal proximal tubular cells, are assembled into an integrated functional framework. We briefly describe oligospecific Na
+ -and Cl- dependent monoamine neurotransmitter transporters (SLC6-family), high-affinity choline transporters (SLC5-family), and high-affinity thiamine transporters (SLC19-family), as well as polyspecific transporters that translocate some organic cations next to their preferred, noncationic substrates. The polyspecific cation transporters of the SLC22 family including the subtypes OCT1-3 and OCTN1-2 are presented in detail, covering the current knowledge about distribution, substrate specificity, and recent data on their electrical properties and regulation. Moreover, we discuss artificial and spontaneous mutations of transporters of the SLC22 family that provide novel insight as to the function of specific protein domains. Finally, we discuss the clinical potential of the increasing knowledge about polymorphisms and mutations in polyspecific organic cation transporters. [ABSTRACT FROM AUTHOR]- Published
- 2003
- Full Text
- View/download PDF
18. The plasma membrane-associated protein RS1 decreases transcription of the transporter SGLT1 in confluent LLC-PK1 cells.
- Author
-
Korn, T, Kühlkamp, T, Track, C, Schatz, I, Baumgarten, K, Gorboulev, V, and Koepsell, H
- Abstract
Previously we cloned RS1, a 67-kDa polypeptide that is associated with the intracellular side of the plasma membrane. Upon co-expression in Xenopus laevis oocytes, human RS1 decreased the concentration of the Na(+)-D-glucose co-transporter hSGLT1 in the plasma membrane (Valentin, M., Kühlkamp, T., Wagner, K., Krohne, G., Arndt, P., Baumgarten, K., Weber, W.-M., Segal, A., Veyhl, M., and Koepsell, H. (2000) Biochim. Biophys. Acta 1468, 367-380). Here, the porcine renal epithelial cell line LLC-PK1 was used to investigate whether porcine RS1 (pRS1) plays a role in transcriptional up-regulation of SGLT1 after confluence and in down-regulation of SGLT1 by high extracellular D-glucose concentrations. Western blots indicated a dramatic decrease of endogenous pRS1 protein at the plasma membrane after confluence but no significant effect of D-glucose. In confluent LLC-PK1 cells overexpressing pRS1, SGLT1 mRNA, protein, and methyl-alpha-D-glucopyranoside uptakes were drastically decreased; however, the reduction of methyl-alpha-D-glucopyranoside uptake after cultivation with 25 mm D-glucose remained. In confluent pRS1 antisense cells, the expression of SGLT1 mRNA and protein was strongly increased, whereas the reduction of SGLT1 expression during cultivation with high D-glucose was not influenced. Nuclear run-on assays showed that the transcription of SGLT1 was 10-fold increased in the pRS1 antisense cells. The data suggest that RS1 participates in transcriptional up-regulation of SGLT1 after confluence but not in down-regulation by D-glucose.
- Published
- 2001
- Full Text
- View/download PDF
19. Electrogenic properties and substrate specificity of the polyspecific rat cation transporter rOCT1.
- Author
-
Busch, A E, Quester, S, Ulzheimer, J C, Waldegger, S, Gorboulev, V, Arndt, P, Lang, F, and Koepsell, H
- Abstract
The previously cloned rat cation transporter rOCT1 detected in renal proximal tubules and hepatocytes (Gründemann, D., Gorboulev, V., Gambaryan, S., Veyhl, M., and Koepsell, H. (1994) Nature 372, 549-552) was expressed in Xenopus oocytes, and transport properties were analyzed using tracer uptake studies and electrophysiological measurements. rOCT1 induced highly active transport of a variety of cations, including the classical substrates for cation transport, such as N-1-methylnicotinamide, 1-methyl-4-phenylpyridinium (MPP), and tetraethylammonium (TEA), but also the physiologically important choline. In oocytes rOCT1 also mediated efflux of MPP, which could be trans-stimulated by MPP and TEA. Cation transport via rOCT1 was electrogenic. In voltage-clamped oocytes, transport of TEA and choline via rOCT1 produced inwardly directed currents, which were independent of extracellular ion composition or pH. The choline- and TEA-induced currents were voltage-dependent at nonsaturating concentrations, and the apparent affinity of these cations was decreased at depolarized voltages. Other substrates transported by rOCT1 were the polyamines spermine and spermidine. Interestingly, the previously described potent inhibitors of rOCT1, cyanine 863, quinine, and D-tubocurarine were substrates themselves. The data indicate that rOCT1 is an effective transport system that is responsible for electrogenic uptake of a wide variety of organic cations into epithelial cells of renal proximal tubules and hepatocytes.
- Published
- 1996
20. The two human organic cation transporter genes SLC22A1 and SLC22A2 are located on chromosome 6q26.
- Author
-
Koehler, M.R., Wissinger, B., Gorboulev, V., Koepsell, H., and Schmid, M.
- Published
- 1997
- Full Text
- View/download PDF
21. Rat Organic Cation Transporter 1 Contains Three Binding Sites for Substrate 1-Methyl-4-phenylpyridinium per Monomer.
- Author
-
Keller T, Gorboulev V, Mueller TD, Dötsch V, Bernhard F, and Koepsell H
- Subjects
- Animals, Binding Sites, Green Fluorescent Proteins metabolism, Mutagenesis physiology, Phospholipids metabolism, Proteolipids metabolism, Rats, 1-Methyl-4-phenylpyridinium metabolism, Catecholamine Plasma Membrane Transport Proteins metabolism
- Abstract
Organic cation transporters OCT1 ( SLC22A1 ) and OCT2 ( SLC22A2 ) are critically involved in absorption and excretion of diverse cationic drugs. Because drug-drug interactions at these transporters may induce adverse drug effects in patients, in vitro testing during drug development for interaction with the human transporters is mandatory. Recent data performed with rat OCT1 (rOCT1) suggest that currently performed in vitro tests assuming one polyspecific binding site are insufficient. Here we measured the binding and transport of model substrate 1-methyl-4-phenylpyridinium
+ (MPP+ ) by cell-free-expressed fusion proteins of rOCT1 and rOCT1 mutants with green fluorescent protein that had been reconstituted into nanodiscs or proteoliposomes. The nanodiscs were formed with major scaffold protein (MSP) and different phospholipids, whereas the proteoliposomes were formed with a mixture of cholesterol, phosphatidylserine, and phosphatidylcholine. In nanodiscs formed with 1-palmitoyl-2-oleoyl- sn- glycero-3-phosphocholine or cholesterol, phosphatidylserine, and phosphatidylcholine, two low-affinity MPP+ binding sites and one high-affinity MPP+ binding site per transporter monomer were determined. Mutagenesis revealed that tryptophan 218 and aspartate 475 in neighboring positions in the modeled outward-open cleft contribute to one low-affinity binding site, whereas arginine 440 located distantly in the cleft is critical for MPP+ binding to another low-affinity site. Comparing MPP+ binding with MPP+ transport suggests that the low-affinity sites are involved in MPP+ transport, whereas high-affinity MPP+ binding influences transport allosterically. The data will be helpful in the interpretation of future crystal structures and provides a rationale for future in vitro testing that is more sophisticated and reliable, leading to the generation of pharmacophore models with high predictive power., (Copyright © 2019 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2019
- Full Text
- View/download PDF
22. Assay Conditions Influence Affinities of Rat Organic Cation Transporter 1: Analysis of Mutagenesis in the Modeled Outward-Facing Cleft by Measuring Effects of Substrates and Inhibitors on Initial Uptake.
- Author
-
Gorboulev V, Rehman S, Albert CM, Roth U, Meyer MJ, Tzvetkov MV, Mueller TD, and Koepsell H
- Subjects
- 1-Methyl-4-phenylpyridinium metabolism, 1-Methyl-4-phenylpyridinium pharmacology, Animals, Dose-Response Relationship, Drug, Female, HEK293 Cells, Humans, Mutagenesis physiology, Quaternary Ammonium Compounds metabolism, Quaternary Ammonium Compounds pharmacology, Rats, Substrate Specificity drug effects, Substrate Specificity physiology, Xenopus laevis, Catecholamine Plasma Membrane Transport Proteins antagonists & inhibitors, Catecholamine Plasma Membrane Transport Proteins metabolism, Mutagenesis drug effects
- Abstract
The effects of mutations in the modeled outward-open cleft of rat organic cation transporter 1 (rOCT1) on affinities of substrates and inhibitors were investigated. Human embryonic kidney 293 cells were stably transfected with rOCT1 or rOCT1 mutants, and uptake of the substrates 1-methyl-4-phenylpyridinium
+ (MPP+ ) and tetraethylammonium+ (TEA+ ) or inhibition of MPP+ uptake by the nontransported inhibitors tetrabutylammonium+ (TBuA+ ), tetrapentylammonium+ (TPeA+ ), and corticosterone was measured. Uptake measurements were performed on confluent cell layers using a 2-minute incubation or in dissociated cells using incubation times of 1, 5, or 10 seconds. With both methods, different apparent Michaelis-Menten constant ( Km ) values, different IC50 values, and varying effects of mutations were determined. In addition, varying IC50 values for the inhibition of MPP+ uptake and varying effects of mutations were obtained when different MPP+ concentrations far below the apparent Km value were used for uptake measurements. Eleven mutations were investigated by measuring initial uptake in dissociated cells and employing 0.1 µ M MPP+ for uptake during inhibition experiments. Altered affinities for substrates and/or inhibitors were observed when Phe160, Trp218, Arg440, Leu447, and Asp475 were mutated. The mutations resulted in changes of apparent Km values for TEA+ and/or MPP+ Mutation of Trp218 and Asp475 led to altered IC50 values for TBuA+ , TPeA+ , and corticosterone, whereas the mutation of Phe160 and Leu447 changed the IC50 values for two inhibitors. Thereby amino acids in the outward-facing conformation of rOCT1 could be identified that interact with structurally different inhibitors and probably also with different substrates., (Copyright © 2018 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2018
- Full Text
- View/download PDF
23. Protein RS1 (RSC1A1) Downregulates the Exocytotic Pathway of Glucose Transporter SGLT1 at Low Intracellular Glucose via Inhibition of Ornithine Decarboxylase.
- Author
-
Chintalapati C, Keller T, Mueller TD, Gorboulev V, Schäfer N, Zilkowski I, Veyhl-Wichmann M, Geiger D, Groll J, and Koepsell H
- Subjects
- Animals, Biological Transport drug effects, Caco-2 Cells, Cell Membrane drug effects, Cell Membrane metabolism, Eflornithine pharmacology, Electrophoresis, Polyacrylamide Gel, HEK293 Cells, Humans, Immunoprecipitation, Intracellular Space metabolism, Kinetics, Methylglucosides pharmacology, Models, Biological, Monosaccharide Transport Proteins chemistry, Oocytes drug effects, Oocytes metabolism, Phlorhizin pharmacology, Protein Binding drug effects, Protein Domains, Recombinant Proteins metabolism, Subcellular Fractions drug effects, Subcellular Fractions metabolism, Surface Plasmon Resonance, Xenopus laevis, Down-Regulation drug effects, Exocytosis drug effects, Glucose pharmacology, Monosaccharide Transport Proteins metabolism, Ornithine Decarboxylase metabolism, Sodium-Glucose Transporter 1 metabolism
- Abstract
Na
+ -d-glucose cotransporter 1 (SGLT1) is rate-limiting for glucose absorption in the small intestine. Shortly after intake of glucose-rich food, SGLT1 abundance in the luminal membrane of the small intestine is increased. This upregulation occurs via glucose-induced acceleration of the release of SGLT1-containing vesicles from the trans-Golgi network (TGN), which is regulated by a domain of protein RS1 (RSC1A1) named RS1-Reg. Dependent on phosphorylation, RS1-Reg blocks release of vesicles containing SGLT1 or concentrative nucleoside transporter 1. The hypothesis has been raised that RS1-Reg binds to different receptor proteins at the TGN, which trigger release of vesicles with different transporters. To identify the presumed receptor proteins, two-hybrid screening was performed. Interaction with ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme of polyamine synthesis, was observed and verified by immunoprecipitation. Binding of RS1-Reg mutants to ODC1 was characterized using surface plasmon resonance. Inhibition of ODC1 activity by RS1-Reg mutants and the ODC1 inhibitor difluoromethylornithine (DFMO) was measured in the absence and presence of glucose. In addition, short-term effects of DFMO, RS1-Reg mutants, the ODC1 product putrescine, and/or glucose on SGLT1 expressed in oocytes of Xenopus laevis were investigated. High-affinity binding of RS1-Reg to ODC1 was demonstrated, and evidence for a glucose binding site in ODC1 was provided. Binding of RS1-Reg to ODC1 inhibits the enzymatic activity at low intracellular glucose, which is blunted at high intracellular glucose. The data suggest that generation of putrescine by ODC1 at the TGN stimulates release of SGLT1-containing vesicles. This indicates a biomedically important role of ODC1 in regulation of glucose homeostasis., (Copyright © 2016 by The American Society for Pharmacology and Experimental Therapeutics.)- Published
- 2016
- Full Text
- View/download PDF
24. Central nervous system infiltrates are characterized by features of ongoing B cell-related immune activity in MP4-induced experimental autoimmune encephalomyelitis.
- Author
-
Batoulis H, Wunsch M, Birkenheier J, Rottlaender A, Gorboulev V, and Kuerten S
- Subjects
- Animals, Cerebellum immunology, Cerebellum metabolism, Chemokine CXCL13 immunology, Disease Models, Animal, Mice, Myelin Basic Protein immunology, Myelin Proteolipid Protein immunology, Neprilysin immunology, Spleen immunology, Spleen metabolism, Autoantigens immunology, B-Lymphocytes immunology, Central Nervous System immunology, Encephalomyelitis, Autoimmune, Experimental immunology, Multiple Sclerosis immunology, Myelin Sheath immunology, RNA, Messenger metabolism
- Abstract
In multiple sclerosis (MS) lymphoid follicle-like aggregates have been reported in the meninges of patients. Here we investigated the functional relevance of B cell infiltration into the central nervous system (CNS) in MP4-induced experimental autoimmune encephalomyelitis (EAE), a B cell-dependent mouse model of MS. In chronic EAE, B cell aggregates were characterized by the presence of CXCL13(+) and germinal center CD10(+) B cells. Germline transcripts were expressed in the CNS and particularly related to TH17-associated isotypes. We also observed B cells with restricted VH gene usage that differed from clones found in the spleen. Finally, we detected CNS-restricted spreading of the antigen-specific B cell response towards a myelin and a neuronal autoantigen. These data imply the development of autonomous B cell-mediated autoimmunity in the CNS in EAE - a concept that might also apply to MS itself., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
25. Substrate- and cell contact-dependent inhibitor affinity of human organic cation transporter 2: studies with two classical organic cation substrates and the novel substrate cd2+.
- Author
-
Thévenod F, Ciarimboli G, Leistner M, Wolff NA, Lee WK, Schatz I, Keller T, Al-Monajjed R, Gorboulev V, and Koepsell H
- Subjects
- Animals, Apoptosis drug effects, Cadmium pharmacology, Catecholamine Plasma Membrane Transport Proteins metabolism, Cell Line, Cimetidine pharmacology, Corticosterone pharmacology, Humans, Kidney Tubules, Proximal metabolism, Organic Cation Transporter 2, Rats, Tetraethylammonium pharmacology, Cadmium metabolism, Organic Cation Transport Proteins antagonists & inhibitors, Organic Cation Transport Proteins metabolism
- Abstract
Polyspecific organic cation transporter Oct2 from rat (gene Slc22A2) has been previously shown to transport Cs(+). Here we report that human OCT2 (hOCT2) is able to transport Cd(2+) showing substrate saturation with a Michaelis-Menten constant (Km) of 54 ± 5.8 μM. Uptake of Cd(2+) by hOCT2 was inhibited by typical hOCT2 ligands (unlabeled substrates and inhibitors), and the rate of uptake was decreased by a point mutation in a substrate binding domain of hOCT2. Incubation of hOCT2 overexpressing human embryonic kidney 293 cells (HEK-hOCT2-C) or rat renal proximal tubule cells expressing rOct2 (NRK-52E-C) with Cd(2+) resulted in an increased level of apoptosis that was reduced by OCT2 inhibitory ligand cimetidine(+). HEK-hOCT2-C exhibited different functional properties when they were confluent or had been dissociated by removal of Ca(2+) and Mg(2+). Only confluent HEK-hOCT2-C transported Cd(2+), and confluent and dissociated cells exhibited different potencies for inhibition of uptake of 1-methyl-4-phenylpyridinium(+) (MPP(+)) by Cd(2+), MPP(+), tetraethylammonium(+), cimetidine(+), and corticosterone. In confluent HEK-hOCT2-C, largely different inhibitor potencies were obtained upon comparison of inhibition of Cd(2+) uptake, 4-[4-(dimethylamino)styryl]-N-methylpyridinium(+) (ASP(+)) uptake, and MPP(+) uptake using substrate concentrations far below the respective Km values. Employing a point mutation in the previously identified substrate binding site of rat Oct1 produced evidence that short distance allosteric effects between binding sites for substrates and inhibitors are involved in substrate-dependent inhibitor potency. Substrate-dependent inhibitor affinity is probably a common property of OCTs. To predict interactions between drugs that are transported by OCTs and inhibitory drugs, it is necessary to employ the specific transported drug rather than a model substrate for in vitro measurements.
- Published
- 2013
- Full Text
- View/download PDF
26. A substrate binding hinge domain is critical for transport-related structural changes of organic cation transporter 1.
- Author
-
Egenberger B, Gorboulev V, Keller T, Gorbunov D, Gottlieb N, Geiger D, Mueller TD, and Koepsell H
- Subjects
- Animals, Binding Sites, Catecholamine Plasma Membrane Transport Proteins chemistry, Catecholamine Plasma Membrane Transport Proteins genetics, HEK293 Cells, Humans, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, Rats, Substrate Specificity physiology, Xenopus laevis, Catecholamine Plasma Membrane Transport Proteins metabolism, Ion Channel Gating physiology
- Abstract
Organic cation transporters are membrane potential-dependent facilitative diffusion systems. Functional studies, extensive mutagenesis, and homology modeling indicate the following mechanism. A transporter conformation with a large outward-open cleft binds extracellular substrate, passes a state in which the substrate is occluded, turns to a conformation with an inward-open cleft, releases substrate, and subsequently turns back to the outward-open state. In the rat organic cation transporter (rOct1), voltage- and ligand-dependent movements of fluorescence-labeled cysteines were measured by voltage clamp fluorometry. For fluorescence detection, cysteine residues were introduced in extracellular parts of cleft-forming transmembrane α-helices (TMHs) 5, 8, and 11. Following expression of the mutants in Xenopus laevis oocytes, cysteines were labeled with tetramethylrhodamine-6-maleimide, and voltage-dependent conformational changes were monitored by voltage clamp fluorometry. One cysteine was introduced in the central domain of TMH 11 replacing glycine 478. This domain contains two amino acids that are involved in substrate binding and two glycine residues (Gly-477 and Gly-478) allowing for helix bending. Cys-478 could be modified with the transported substrate analog [2-(trimethylammonium)-ethyl]methanethiosulfonate but was inaccessible to tetramethylrhodamine-6-maleimide. Voltage-dependent movements at the indicator positions of TMHs 5, 8, and 11 were altered by substrate applications indicating large conformational changes during transport. The G478C exchange decreased transporter turnover and blocked voltage-dependent movements of TMHs 5 and 11. [2-(Trimethylammonium)-ethyl]methanethiosulfonate modification of Cys-478 blocked substrate binding, transport activity, and movement of TMH 8. The data suggest that Gly-478 is located within a mechanistically important hinge domain of TMH 11 in which substrate binding induces transport-related structural changes.
- Published
- 2012
- Full Text
- View/download PDF
27. The large extracellular loop of organic cation transporter 1 influences substrate affinity and is pivotal for oligomerization.
- Author
-
Keller T, Egenberger B, Gorboulev V, Bernhard F, Uzelac Z, Gorbunov D, Wirth C, Koppatz S, Dötsch V, Hunte C, Sitte HH, and Koepsell H
- Subjects
- Animals, Catecholamine Plasma Membrane Transport Proteins chemistry, Catecholamine Plasma Membrane Transport Proteins genetics, Dithiothreitol chemistry, Dithiothreitol pharmacology, HEK293 Cells, Humans, Ion Transport drug effects, Ion Transport physiology, Mutation, Organic Anion Transport Protein 1 chemistry, Organic Anion Transport Protein 1 genetics, Organic Anion Transport Protein 1 metabolism, Protein Multimerization drug effects, Protein Structure, Quaternary, Protein Structure, Secondary, Rats, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Xenopus laevis, Catecholamine Plasma Membrane Transport Proteins metabolism, Protein Multimerization physiology
- Abstract
Polyspecific organic anion transporters (OATs) and organic cation transporters (OCTs) of the SLC22 transporter family play a pivotal role in absorption, distribution, and excretion of drugs. Polymorphisms in these transporters influence therapeutic effects. On the basis of functional characterizations, homology modeling, and mutagenesis, hypotheses for how OCTs bind and translocate structurally different cations were raised, assuming functionally competent monomers. However, homo-oligomerization has been described for OATs and OCTs. In the present study, evidence is provided that the large extracellular loops (EL) of rat Oct1 (rOct1) and rat Oat1 (rOat1) mediate homo- but not hetero-oligomerization. Replacement of the cysteine residues in the EL of rOct1 by serine residues (rOct1(6ΔC-l)) or breaking disulfide bonds with dithiothreitol prevented oligomerization. rOct1 chimera containing the EL of rOat1 (rOct1(rOat1-l)) showed oligomerization but reduced transporter amount in the plasma membrane. For rOct1(6ΔC-l) and rOct1(rOat1-l), similar K(m) values for 1-methyl-4-phenylpyridinium(+) (MPP(+)) and tetraethylammonium(+) (TEA(+)) were obtained that were higher compared with rOct1 wild type. The increased K(m) of rOct1(rOat1-l) indicates an allosteric effect of EL on the cation binding region. The similar substrate affinity of the oligomerizing and non-oligomerizing loop mutants suggests that oligomerization does not influence transport function. Independent transport function of rOct1 monomers was also demonstrated by showing that K(m) values for MPP(+) and TEA(+) were not changed after treatment with dithiothreitol and that a tandem protein with two rOct1 monomers showed about 50% activity with unchanged K(m) values for MPP(+) and TEA(+) when one monomer was blocked. The data help to understand how OCTs work and how mutations in patients may affect their functions.
- Published
- 2011
- Full Text
- View/download PDF
28. Novel shuttling domain in a regulator (RSC1A1) of transporter SGLT1 steers cell cycle-dependent nuclear location.
- Author
-
Filatova A, Leyerer M, Gorboulev V, Chintalapati C, Reinders Y, Müller TD, Srinivasan A, Hübner S, and Koepsell H
- Subjects
- Active Transport, Cell Nucleus, Animals, Cell Cycle, Culture Media, Serum-Free, Genetic Vectors, Glucose metabolism, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Karyopherins metabolism, LLC-PK1 Cells, Monosaccharide Transport Proteins genetics, Nuclear Localization Signals metabolism, Phosphorylation, Protein Kinase C metabolism, Protein Structure, Tertiary, Protein Transport physiology, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Ribonucleoproteins, Small Nuclear metabolism, Sodium metabolism, Swine, Time Factors, Transfection, trans-Golgi Network metabolism, Cell Nucleus metabolism, Membrane Transport Proteins metabolism, Monosaccharide Transport Proteins chemistry, Monosaccharide Transport Proteins metabolism, Sodium-Glucose Transporter 1 metabolism
- Abstract
The gene product of RSC1A1, RS1, participates in the regulation of the Na(+)-D-glucose cotransporter SGLT1. RS1 inhibits release of SGLT1 from the trans Golgi network. In subconfluent LLC-PK(1) cells, RS1 migrates into the nucleus and modulates transcription of SGLT1, whereas most confluent cells do not contain RS1 in the nuclei. We showed that confluence-dependent nuclear location of RS1 is because of different phases of the cell cycle and identified a RS1 nuclear shuttling domain (RNS) with an associated protein kinase C (PKC) phosphorylation site (RNS-PKC) that mediates cell cycle-dependent nuclear location. RNS-PKC contains a novel non-conventional nuclear localization signal interacting with importin beta1, a nuclear export signal mediating export via protein CRM1 and a Ca(2+)-dependent calmodulin binding site. PKC and calmodulin compete for binding to RNS-PKC. Mutagenesis experiments and analyses of the phosphorylation status suggest the following sequences of events. Subconfluent cells without and with synchronization to the G2/M phase contain non-phosphorylated RNS-PKC that mediates nuclear import of RS1 but not its export. During confluence or synchronization of subconfluent cells to the G2/M phase, phosphorylation of RNS-PKC mediates rapid nuclear export of RS1.
- Published
- 2009
- Full Text
- View/download PDF
29. Five amino acids in the innermost cavity of the substrate binding cleft of organic cation transporter 1 interact with extracellular and intracellular corticosterone.
- Author
-
Volk C, Gorboulev V, Kotzsch A, Müller TD, and Koepsell H
- Subjects
- Alanine metabolism, Amino Acid Sequence genetics, Amino Acid Substitution, Amino Acids genetics, Animals, Corticosterone pharmacology, Dose-Response Relationship, Drug, Female, Hydrogen Bonding, Hydrophobic and Hydrophilic Interactions, Inhibitory Concentration 50, Models, Molecular, Molecular Sequence Data, Oocytes, Organic Cation Transporter 1 genetics, Patch-Clamp Techniques, Phenylalanine metabolism, Point Mutation, Protein Structure, Secondary, Rats, Sequence Homology, Amino Acid, Substrate Specificity physiology, Tyrosine metabolism, Xenopus laevis, Amino Acids metabolism, Corticosterone metabolism, Organic Cation Transporter 1 metabolism
- Abstract
We have shown previously that Leu447 and Gln448 in the transmembrane helix (TMH) 10 of rat organic cation transporter rOCT1 are critical for inhibition of cation uptake by corticosterone. Here, we tested whether the affinity of corticosterone is different when applied from the extracellular or intracellular side. The affinity of corticosterone was determined by measuring the inhibition of currents induced by tetraethylammonium(+) (TEA(+)) in Xenopus laevis oocytes expressing rOCT1. Either corticosterone and TEA(+) were added to the bath simultaneously or the oocytes were preincubated with corticosterone, washed, and TEA(+)-induced currents were determined subsequently. In mutant L447Y, K(i) values for extracellular and intracellular corticosterone were decreased, whereas in mutant Q448E, only the K(i) for intracellular corticosterone was changed. Modeling of the interaction of corticosterone with rOCT1 in the inward- or outward-facing conformation predicted direct binding to Leu447, Phe160 (TMH2), Trp218 (TMH4), Arg440 (TMH10), and Asp475 (TM11) from both sides. In mutant F160A, affinities for extracellular and intracellular corticosterone were increased, whereas maximal inhibition was reduced in W218F and R440K. In stably transfected epithelial cells, the affinities for inhibition of 1-methyl-4-phenyl-pyridinium(+) (MPP(+)) uptake by extracellular and intracellular corticosterone were decreased when Asp475 was replaced by glutamate. In mutants F160A, W218Y, R440K, and L447F, the affinities for MPP(+) uptake were changed, and in mutant D475E, the affinity for TEA(+) uptake was changed. The data suggest that Phe160, Trp218, Arg440, Leu447, and Asp475 are located within an innermost cavity of the binding cleft that is alternatingly exposed to the extracellular or intracellular side during substrate transport.
- Published
- 2009
- Full Text
- View/download PDF
30. Charge-to-substrate ratio during organic cation uptake by rat OCT2 is voltage dependent and altered by exchange of glutamate 448 with glutamine.
- Author
-
Schmitt BM, Gorbunov D, Schlachtbauer P, Egenberger B, Gorboulev V, Wischmeyer E, Müller T, and Koepsell H
- Subjects
- Animals, Binding Sites, Cations, Dicarboxylic Acids metabolism, Gluconates metabolism, Glutamic Acid, Glutamine, Hydrogen-Ion Concentration, Ion Transport, Lactic Acid metabolism, Membrane Potentials, Models, Molecular, Mutation, Oocytes, Organic Cation Transport Proteins chemistry, Organic Cation Transport Proteins genetics, Organic Cation Transporter 2, Permeability, Protein Conformation, Rats, Structure-Activity Relationship, Tetraethylammonium metabolism, Xenopus laevis, Organic Cation Transport Proteins metabolism
- Abstract
Uptake of substrate and electric charge was measured simultaneously in voltage-clamped Xenopus laevis oocytes expressing rat organic cation transporter 2 (rOCT2). At 0 mV, saturating substrate concentrations induced uptake of more positive elementary charges than monovalent organic cations, with charge-to-substrate ratios of 1.5 for guanidinium(+), 3.5 for tetraethylammonium(+), and 4.0 for 1-methyl-4-phenylpyridinium(+). At negative holding potentials, the charge-to-substrate ratios decreased toward unity. At 0 mV, charge-to-substrate ratios higher than unity were observed at different extracellular pH and after replacement of extracellular Na(+), K(+), Ca(2+), Mg(2+), and/or Cl(-). Charge-to-substrate ratios were not influenced by intracellular succinate(2-) or glutarate(2-). The effects of membrane potential and ion substitution strongly suggest that the surplus of transported positive charge is not generated by passive ion permeabilities. Rather, we hypothetize that small cations are taken up together with organic cation substrates whereas the outward reorientation of the empty transporter is electroneutral. Nonselective cotransport of small cations was supported by the three-dimensional structures of rOCT2 in its inward-facing and outward-facing conformations, which we determined by homology modeling based on known corresponding structures of H(+)-lactose permease of E. coli, and by functional analysis of OCT mutants. In our model, the innermost cavity of the outward-open binding cleft is negatively charged by Glu448 and Asp475, whereas the inward-open innermost cavity is electroneutral, containing Asp379, Asp475, Lys215, and Arg440. Substitution of Glu448 by glutamine reduced the charge-to-TEA(+) ratio at 0 mV to unity. The observed charge excess associated with organic cation uptake into depolarized cells may contribute to tubular damage in renal failure.
- Published
- 2009
- Full Text
- View/download PDF
31. Cell free expression and functional reconstitution of eukaryotic drug transporters.
- Author
-
Keller T, Schwarz D, Bernhard F, Dötsch V, Hunte C, Gorboulev V, and Koepsell H
- Subjects
- 1-Methyl-4-phenylpyridinium metabolism, Animals, Catecholamine Plasma Membrane Transport Proteins genetics, Catecholamine Plasma Membrane Transport Proteins isolation & purification, Cell-Free System, Ketoglutaric Acids metabolism, Organic Anion Transport Protein 1 genetics, Organic Anion Transport Protein 1 isolation & purification, Organic Cation Transport Proteins genetics, Organic Cation Transport Proteins isolation & purification, Organic Cation Transporter 2, Proteolipids metabolism, Rats, Recombinant Proteins biosynthesis, Recombinant Proteins isolation & purification, Recombinant Proteins metabolism, p-Aminohippuric Acid metabolism, Catecholamine Plasma Membrane Transport Proteins metabolism, Organic Anion Transport Protein 1 metabolism, Organic Cation Transport Proteins metabolism
- Abstract
Polyspecific organic cation and anion transporters of the SLC22 protein family are critically involved in absorption and excretion of drugs. To elucidate transport mechanisms, functional and biophysical characterization of purified transporters is required and tertiary structures must be determined. Here, we synthesized rat organic cation transporters OCT1 and OCT2 and rat organic anion transporter OAT1 in a cell free system in the absence of detergent. We solubilized the precipitates with 2% 1-myristoyl-2-hydroxy- sn-glycero-3-[phospho- rac-(1-glycerol)] (LMPG), purified the transporters in the presence of 1% 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate (CHAPS) or octyl glucoside, and reconstituted them into proteoliposomes. From 1 mL reaction vessels 0.13-0.36 mg of transporter proteins was purified. Thus, from five to ten 1 mL reaction vessels sufficient protein for crystallization was obtained. In the presence of 1% LMPG and 0.5% CHAPS, OCT1 and OAT1 formed homo-oligomers but no hetero-oligomers. After reconstitution of OCT1, OCT2, and OAT1 into proteoliposomes, similar Michaelis-Menten K m values were measured for uptake of 1-methyl-4-phenylpyridinium and p-aminohippurate (PAH (-)) by the organic cation and anion transporters, respectively, as after expression of the transporters in cells. Using the reconstituted system, evidence was obtained that OAT1 operates as obligatory and electroneutral PAH (-)/dicarboxylate antiporter and contains a low-affinity chloride binding site that stimulates turnover. PAH (-) uptake was observed only with alpha-ketoglutarate (KG (2-)) on the trans side, and trans-KG (2-) increased the PAH (-) concentration in voltage-clamped proteoliposomes transiently above equilibrium. The V max of PAH (-)/KG (2-) antiport was increased by Cl (-) in a manner independent of gradients, and PAH (-)/KG (2-) antiport was independent of membrane potential in the absence or presence of Cl (-).
- Published
- 2008
- Full Text
- View/download PDF
32. High-affinity cation binding to organic cation transporter 1 induces movement of helix 11 and blocks transport after mutations in a modeled interaction domain between two helices.
- Author
-
Gorbunov D, Gorboulev V, Shatskaya N, Mueller T, Bamberg E, Friedrich T, and Koepsell H
- Subjects
- Animals, Catecholamine Plasma Membrane Transport Proteins chemistry, Catecholamine Plasma Membrane Transport Proteins genetics, Cations, Protein Binding, Rats, Catecholamine Plasma Membrane Transport Proteins metabolism, Mutation
- Abstract
Voltage-clamp fluorometry was performed with a cysteine-deprived mutant of rat organic cation transporter 1 (rOCT1) in which Phe483 in transmembrane alpha-helix (TMH) 11 close to the extracellular surface was replaced by cysteine and labeled with tetramethylrhodamine-6-maleimide. Potential-dependent fluorescence changes were observed that were sensitive to presence of substrates choline, tetraethylammonium (TEA), and 1-methyl-4-phenylpyridinium (MPP) and of the nontransported inhibitor tetrabutylammonium (TBuA). Using potential-dependent fluorescence changes as readout, one high-affinity binding site per substrate and two high-affinity binding sites for TBuA were identified in addition to the previously described single interaction sites. In a structure model of rOCT1 with an inward open cleft that was derived from a known crystal structure of lacY permease, Phe483 is close to Trp147 in TMH 2. In contrast, in a model with an outward open cleft these amino acids are far apart. After replacement of Phe483 or Trp147 by cysteine or serine, high-affinity binding of TBuA leads to inhibition of MPP or TEA uptake, whereas it has no effect on cation uptake by wild-type rOCT1. Coexisting high-affinity cation binding sites in organic cation transporters may collect low concentration xenobiotics and drugs; however, translocation including transitions between outward- and inward-oriented conformations may only be induced when a low-affinity cation binding site is loaded. We propose that cations bound to high-affinity sites may be translocated together with cations bound to low-affinity sites or that they may block the translocation mechanism.
- Published
- 2008
- Full Text
- View/download PDF
33. Tripeptides of RS1 (RSC1A1) inhibit a monosaccharide-dependent exocytotic pathway of Na+-D-glucose cotransporter SGLT1 with high affinity.
- Author
-
Vernaleken A, Veyhl M, Gorboulev V, Kottra G, Palm D, Burckhardt BC, Burckhardt G, Pipkorn R, Beier N, van Amsterdam C, and Koepsell H
- Subjects
- Animals, Antimetabolites pharmacology, Biological Transport, Active drug effects, Biological Transport, Active physiology, Caco-2 Cells, Deoxyglucose pharmacology, Diabetes Mellitus drug therapy, Diabetes Mellitus genetics, Diabetes Mellitus metabolism, Female, Gastrointestinal Motility drug effects, Gastrointestinal Motility physiology, Gene Expression, Humans, Intestinal Absorption physiology, Liver metabolism, Male, Methylglucosides pharmacology, Monosaccharide Transport Proteins chemistry, Monosaccharide Transport Proteins genetics, Oligopeptides chemistry, Oligopeptides therapeutic use, Oocytes cytology, Peptide Transporter 1, Protein Processing, Post-Translational physiology, Rats, Rats, Wistar, Sodium-Glucose Transporter 1 genetics, Symporters genetics, Symporters metabolism, Xenopus laevis, trans-Golgi Network metabolism, Glucose metabolism, Intestinal Absorption drug effects, Monosaccharide Transport Proteins metabolism, Oligopeptides pharmacology, Protein Processing, Post-Translational drug effects, Sodium-Glucose Transporter 1 metabolism
- Abstract
The human gene RSC1A1 codes for a 67-kDa protein named RS1 that mediates transcriptional and post-transcriptional regulation of Na(+)-D-glucose cotransporter SGLT1. The post-transcriptional regulation occurs at the trans-Golgi network (TGN). We identified two tripeptides in human RS1 (Gln-Cys-Pro (QCP) and Gln-Ser-Pro (QSP)) that induce posttranscriptional down-regulation of SGLT1 at the TGN leading to 40-50% reduction of SGLT1 in plasma membrane. For effective intracellular concentrations IC(50) values of 2.0 nM (QCP) and 0.16 nm (QSP) were estimated. Down-regulation of SGLT1 by tripeptides was attenuated by intracellular monosaccharides including non-metabolized methyl-alpha-D-glucopyranoside and 2-deoxyglucose. In small intestine post-transcriptional regulation of SGLT1 may contribute to glucose-dependent regulation of liver metabolism and intestinal mobility. QCP and QSP are transported by the H(+)-peptide cotransporter PepT1 that is colocated with SGLT1 in small intestinal enterocytes. Using coexpression of SGLT1 and PepT1 in Xenopus oocytes or polarized Caco-2 cells that contain both transporters we demonstrated that the tripeptides were effective when applied to the extracellular compartment. After a 1-h perfusion of intact rat small intestine with QSP, glucose absorption was reduced by 30%. The data indicate that orally applied tripeptides can be used to down-regulate small intestinal glucose absorption, e.g. in diabetes mellitus.
- Published
- 2007
- Full Text
- View/download PDF
34. Identification of cysteines in rat organic cation transporters rOCT1 (C322, C451) and rOCT2 (C451) critical for transport activity and substrate affinity.
- Author
-
Sturm A, Gorboulev V, Gorbunov D, Keller T, Volk C, Schmitt BM, Schlachtbauer P, Ciarimboli G, and Koepsell H
- Subjects
- Animals, Biological Transport, Active, Catecholamine Plasma Membrane Transport Proteins chemistry, Catecholamine Plasma Membrane Transport Proteins metabolism, Electric Capacitance, Electrophysiology, Exocytosis, Methyl Methanesulfonate pharmacology, Models, Molecular, Oocytes drug effects, Oocytes metabolism, Organic Cation Transport Proteins chemistry, Organic Cation Transport Proteins metabolism, Organic Cation Transporter 2, Protein Binding, Protein Conformation, Rats, Substrate Specificity, Xenopus laevis, Catecholamine Plasma Membrane Transport Proteins genetics, Cysteine chemistry, Organic Cation Transport Proteins genetics
- Abstract
Effects of the sulfhydryl reagent methylmethanethiosulfonate (MMTS) on functions of organic cation transporters (OCTs) were investigated. Currents induced by 10 mM choline [I(max(choline))] in Xenopus laevis oocytes expressing rat OCT1 (rOCT1) were increased four- to ninefold after 30-s incubation with 5 mM MMTS whereas I(max(choline)) by rat OCT2 was 70% decreased. MMTS activated the rOCT1 transporter within the plasma membrane without changing stoichiometry between translocated charge and cation. After modification of oocytes expressing rOCT1 or rOCT2 with MMTS, I(0.5(choline)) values for choline-induced currents were increased. For rOCT1 it was shown that MMTS increased I(0.5) values for different cations by different degrees. Mutagenesis of individual cysteine residues in rOCT1 revealed that modification of cysteine 322 in the large intracellular loop, and of cysteine 451 at the transition of the transmembrane alpha-helix (TMH) 10 to the short intracellular loop between the TMH 10 and 11 is responsible for the observed effects of MMTS. After replacement of cysteine 451 by methionine, the IC(50(choline)) for choline to inhibit MPP uptake by rOCT1 was increased whereas the I(0.5(choline)) value for choline-induced current remained unchanged. At variance, in double mutant Cys322Ser, Cys451Met, I(0.5(choline)) was increased compared with rOCT1 wild-type whereas in the single mutant Cys322Ser I(0.5(choline)) was not changed. The data suggest that modification of rOCT1 at cysteines 322 and 451 leads to an increase in turnover. They indicate that cysteine 451 in rOCT1 interacts with the large intracellular loop and that cysteine 451 in both rOCT1 and rOCT2 is critical for the affinity of choline.
- Published
- 2007
- Full Text
- View/download PDF
35. Transporter regulator RS1 (RSC1A1) coats the trans-Golgi network and migrates into the nucleus.
- Author
-
Kroiss M, Leyerer M, Gorboulev V, Kühlkamp T, Kipp H, and Koepsell H
- Subjects
- Animals, Brefeldin A pharmacology, Clathrin metabolism, Dynamin II metabolism, Green Fluorescent Proteins genetics, Humans, Kidney cytology, LLC-PK1 Cells, Monosaccharide Transport Proteins genetics, Protein Synthesis Inhibitors pharmacology, Protein Transport drug effects, Sodium-Glucose Transporter 1 metabolism, Species Specificity, Swine, Transfection, Cell Nucleus metabolism, Monosaccharide Transport Proteins metabolism, Protein Transport physiology, trans-Golgi Network metabolism
- Abstract
The product of gene RSC1A1, named RS1, is involved in transcriptional and posttranscriptional regulation of sodium-d-glucose cotransporter SGLT1, and removal of RS1 in mice led to an increase of SGLT1 expression in small intestine and to obesity (Osswald C, Baumgarten K, Stümpel F, Gorboulev V, Akimjanova M, Knobeloch K-P, Horak I, Kluge R, Joost H-G, and Koepsell H. Mol Cell Biol 25: 78-87, 2005). Previous data showed that RS1 inhibits transcription of SGLT1 in LLC-PK1 cells derived from porcine kidney. A decrease of the intracellular amount of RS1 protein was observed during cell confluence, which was paralleled by transcriptional upregulation of SGLT1. In the present study, the subcellular distributions of endogenously expressed RS1 and SGLT1 were compared in LLC-PK1 cells and human embryonic kidney (HEK)-293 cells using immunofluorescence microscopy. RS1 was located at the plasma membrane, at the entire trans-Golgi network (TGN), and within the nucleus. Treatment of LLC-PK1 cells with brefeldin A induced rapid release of RS1 from the TGN, and confluence of LLC-PK1 cells was accompanied by reduction of nuclear location of RS1; 84-90% of subconfluent cells and 5-34% of confluent cells contained RS1 in the nuclei. This suggests that confluence-dependent transcriptional inhibition by RS1 is partially regulated by nuclear migration. Furthermore, we assigned SGLT1 to microtubule-associated tubulovesicular structures and dynamin-containing parts of the TGN. The data indicate that RS1 inhibits the dynamin-dependent release of SGLT1-containing vesicles from the TGN.
- Published
- 2006
- Full Text
- View/download PDF
36. RS1 (RSC1A1) regulates the exocytotic pathway of Na+-D-glucose cotransporter SGLT1.
- Author
-
Veyhl M, Keller T, Gorboulev V, Vernaleken A, and Koepsell H
- Subjects
- Animals, Biological Transport drug effects, Biological Transport physiology, Brefeldin A pharmacology, Carbon Radioisotopes, Dynamins metabolism, Exocytosis drug effects, Glucose metabolism, Humans, Insecta, Methylglucosides pharmacokinetics, Monosaccharide Transport Proteins genetics, Monosaccharide Transport Proteins pharmacology, Oocytes cytology, Oocytes metabolism, Organic Cation Transport Proteins metabolism, Organic Cation Transporter 2, Protein Kinase C metabolism, Protein Synthesis Inhibitors pharmacology, Rats, Sodium metabolism, Xenopus laevis, trans-Golgi Network metabolism, Exocytosis physiology, Monosaccharide Transport Proteins metabolism, Sodium-Glucose Transporter 1 metabolism
- Abstract
The product of gene RSC1A1, named RS1, participates in transcriptional and posttranscriptional regulation of the sodium-d-glucose cotransporter SGLT1. Using coexpression in oocytes of Xenopus laevis, posttranscriptional inhibition of human SGLT1 (hSGLT1) and some other transporters by human RS1 (hRS1) was demonstrated previously. In the present study, histidine-tagged hRS1 was expressed in oocytes or Sf9 cells and purified using nickel(II)-charged nitrilotriacetic acid-agarose. hRS1 protein was injected into oocytes expressing hSGLT1 or the human organic cation transporter hOCT2, and the effect on hSGLT1-mediated uptake of methyl-alpha-D-[14C]glucopyranoside ([14C]AMG) or hOCT2-mediated uptake of [14C]tetraethylammonium ([14C]TEA) was measured. Within 30 min after the injection of hRS1 protein, hSGLT1-expressed AMG uptake or hOCT2-expressed TEA uptake was inhibited by approximately 50%. Inhibition of AMG uptake was decreased when a dominant negative mutant of dynamin I was coexpressed and increased after stimulation of PKC. Inhibition remained unaltered when endocytosis was inhibited by chlorpromazine, imipramine, or filipin but was prevented when exocytosis was inhibited by botulinum toxin B or when the release of vesicles from the TGN and endosomes was inhibited by brefeldin A. Inhibition of hSGLT1-mediated AMG uptake and hOCT2-mediated TEA uptake by hRS1 protein were decreased at an enhanced intracellular AMG concentration. The data suggest that hRS1 protein exhibits glucose-dependent, short-term inhibition of hSGLT1 and hOCT2 by inhibiting the release of vesicles from the trans-Golgi network.
- Published
- 2006
- Full Text
- View/download PDF
37. Characterization of regulatory mechanisms and states of human organic cation transporter 2.
- Author
-
Biermann J, Lang D, Gorboulev V, Koepsell H, Sindic A, Schröter R, Zvirbliene A, Pavenstädt H, Schlatter E, and Ciarimboli G
- Subjects
- Amiloride metabolism, Binding, Competitive, Biological Transport, Blotting, Western, Calcium metabolism, Calmodulin metabolism, Flow Cytometry, Humans, Immunohistochemistry, Microscopy, Confocal, Microscopy, Fluorescence, Organic Cation Transporter 2, Patch-Clamp Techniques, Protein Transport physiology, Pyridines metabolism, Substrate Specificity, Models, Biological, Organic Cation Transport Proteins metabolism, Signal Transduction physiology
- Abstract
Polyspecific organic cation transporters (OCTs) have a large substrate binding pocket with different interaction domains. To determine whether OCT regulation is substrate specific, suitable fluorescent organic cations were selected by comparing their uptake in wild-type (WT) human embryonic kidney (HEK)-293 cells and in HEK-293 cells stably transfected with hOCT2. N-amidino-3,5-diamino-6-chloropyrazine-carboxamide (amiloride) and 4-[4-(dimethylamino)-styryl]-N-methylpyridinium (ASP) showed concentration-dependent uptake in hOCT2 at 37 degrees C. After subtraction of unspecific uptake determined in WT at 37 degrees C or in hOCT2 at 8 degrees C saturable specific uptake of both substrates was measured. Km values of hOCT2-mediated uptake of 95 microM amiloride and 24 microM ASP were calculated. Inhibition of amiloride and ASP uptake by several organic cations was also measured [IC50 (in microM) for amiloride and ASP, respectively, tetraethylammonium (TEA) 98 and 30, cimetidine 14 and 26, and tetrapentylammonium (TPA) 7 and 2]. Amiloride and ASP uptake were significantly reduced by inhibition of Ca2+/CaM complex (-55 +/- 5%, n = 10 and -63 +/- 2%, n = 15, for amiloride and ASP, respectively) and stimulation of PKC (-54 +/- 5%, n = 14, and -31 +/- 6%, n = 26) and PKA (-16 +/- 5%, n = 16, and -18 +/- 4%, n = 40), and they were increased by inhibition of phosphatidylinositol 3-kinase (+28 +/- 6%, n = 8, and +55 +/- 17%, n = 16). Inhibition of Ca2+/CaM complex resulted in a significant decrease of Vmax (160-99 photons/s) that can be explained in part by a reduction of the membrane-associated hOCT2 (-22 +/- 6%, n = 9) as determined using FACScan flow cytometry. The data indicate that saturable transport by hOCT2 can be measured by the fluorescent substrates amiloride and ASP and that transport activity for both substrates is regulated similarly. Inhibition of the Ca2+/CaM complex causes changes in transport capacity via hOCT2 trafficking.
- Published
- 2006
- Full Text
- View/download PDF
38. Rat renal glucose transporter SGLT1 exhibits zonal distribution and androgen-dependent gender differences.
- Author
-
Sabolić I, Skarica M, Gorboulev V, Ljubojević M, Balen D, Herak-Kramberger CM, and Koepsell H
- Subjects
- Androgens physiology, Animals, Castration, Down-Regulation, Female, Gene Expression Profiling, Immunoblotting, Kidney Glomerulus physiology, Kidney Tubules, Proximal physiology, Male, Ovariectomy, RNA, Messenger analysis, Rats, Rats, Wistar, Sex Factors, Sodium-Glucose Transporter 1 analysis, Sodium-Glucose Transporter 1 physiology, Kidney Glomerulus chemistry, Kidney Tubules, Proximal chemistry, Sodium-Glucose Transporter 1 biosynthesis
- Abstract
SGLT1 (SLC5A1) mediates a part of glucose and galactose reabsorption in the mammalian proximal tubule (PT), but the detailed localization of the transporter along the tubule is still disputable. Here, we used several methods to localize rat SGLT1 (rSGLT1) in the kidneys of intact and variously treated male (M) and female (F) rats. In immunoblots of isolated cortical (C) and outer stripe (OS) brush-border membranes (BBM), a peptide-specific polyclonal antibody for rSGLT1 labeled a sharp inzone-, and gender-dependent approximately 40-kDa protein and a broad approximately 75-kDa band that exhibited strong zonal (OS > C) and gender differences (F > M). In tissue cryosections, the antibody strongly stained BBM of the S3 PT segments in the OS and medullary rays (F > M) and smooth muscles of the blood vessels and renal capsule (F approximately M) and weakly stained the apical domain of other PT segments in the C (F approximately M). The phlorizin-sensitive uptake of d-[(3)H]galactose in BBM vesicles, as well as the tissue abundance of rSGLT1-specific mRNA, matched the immunoblotting data related to the 75-kDa protein and the immunostaining in S3, proving zonal and gender differences in the functional transporter. Ovariectomy had no effect, castration upregulated, whereas treatment of castrated rats with testosterone, but not with estradiol or progesterone, downregulated the 75-kDa protein and the immunostaining in S3. We conclude that in the rat kidney, the expression of SGLT1 is represented by a 75-kDa protein localized largely in the PT S3 segments, where it exhibits gender differences (F > M) at both the protein and mRNA levels that are caused by androgen inhibition.
- Published
- 2006
- Full Text
- View/download PDF
39. Purification and functional reconstitution of the rat organic cation transporter OCT1.
- Author
-
Keller T, Elfeber M, Gorboulev V, Reiländer H, and Koepsell H
- Subjects
- 1-Methyl-4-phenylpyridinium pharmacology, Animals, Catecholamine Plasma Membrane Transport Proteins antagonists & inhibitors, Catecholamine Plasma Membrane Transport Proteins genetics, Cell Line, Gene Expression, Proteolipids drug effects, Proteolipids genetics, Proteolipids metabolism, Rats, Spodoptera, Substrate Specificity, Catecholamine Plasma Membrane Transport Proteins isolation & purification, Catecholamine Plasma Membrane Transport Proteins metabolism
- Abstract
The rat organic cation transporter rOCT1 with six histidine residues added to the C-terminus was expressed in Sf9 insect cells, and expression of organic cation transport was demonstrated. To purify rOCT1 protein, Sf9 cells were lysed with 1% (w/v) CHAPS [3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate], centrifuged, and subjected to sequential affinity chromatography using lentil-lectin Sepharose and nickel(II)-charged nitrilotriacetic acid-agarose. This procedure yielded approximately 70 microg of purified rOCT1 protein from 10 standard culture plates. Using a freeze-thaw procedure, purified rOCT1 was reconstituted into proteoliposomes formed from phosphatidylcholine, phosphatidylserine, and cholesterol. Proteoliposomes exhibited uptake of [3H]-1-Methyl-4-phenylpyridinium ([3H]MPP) that was inhibited by quinine and stimulated by an inside-negative membrane potential. MPP uptake was saturable with an apparent K(m) of 30 +/- 17 microM. MPP uptake (0.1 microM) was inhibited by tetraethylammonium, tetrabutylammonium, and tetrapentylammonium with IC50 values of 197 +/- 11, 19 +/- 1, and 1.8 +/- 0.03 microM, respectively. With membrane potential clamped to 0 mV using valinomycin in the presence of 100 mM potassium on both sides of the membrane, uptake of 0.1 microM MPP was trans stimulated 3-fold by 2.5 mM intracellular choline, and efflux of 0.1 microM MPP was trans stimulated 4-fold by 9.5 mM extracellular choline. The data show that rOCT1 is capable and sufficient to mediate transport of organic cations. The observed trans stimulation under voltage-clamp conditions shows that rOCT1 operates as a transporter rather than a channel. Purification and reconstitution of functional active rOCT1 protein is an important step toward the biophysical characterization and crystallization.
- Published
- 2005
- Full Text
- View/download PDF
40. Individual PKC-phosphorylation sites in organic cation transporter 1 determine substrate selectivity and transport regulation.
- Author
-
Ciarimboli G, Koepsell H, Iordanova M, Gorboulev V, Dürner B, Lang D, Edemir B, Schröter R, Van Le T, and Schlatter E
- Subjects
- Animals, Cells, Cultured, Humans, Ion Transport, Mutation, Organic Cation Transporter 1 metabolism, Phosphorylation, Polymorphism, Single Nucleotide, Protein Kinase C metabolism, Rats, Substrate Specificity, Organic Cation Transporter 1 genetics, Protein Kinase C genetics
- Abstract
To elucidate the molecular mechanisms underlying stimulation of rat organic cation transporter type 1 (rOCT1) by protein kinase C (PKC) activation, functional properties and regulation of rOCT1 stably expressed in HEK293 cells after site-directed mutagenesis of putative PKC phosphorylation-sites were compared with wild-type (WT) rOCT1 using microfluorometric measurements with the fluorescence organic cation 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP(+)). Either substitutions of single (S286A, S292A, T296A, S328A, and T550A) or of all five PKC-sites (5x-PKC) with alanine suppressed PKC-induced stimulation of ASP(+) uptake, whereas regulation by p56(lck) tyrosine kinase was conserved in all mutants. Remarkably, the apparent affinities for TEA(+), TPA(+), and quinine were changed differently in each mutant (EC(50) in WT, S286A, S292A, T296A, S328A, T550A, and 5x-PKC in mumol: TEA(+): 105, 153, 56, 1135, 484, 498, 518; TPA(+): 0.1, 2.1, 0.3, 1.0, 43, 0.3, 2.2; quinine: 1.5, 3.0, 2.5, 4.8, 81, 7.6, 8.9, respectively). After mutations, no effects of PKC activation on apparent affinity of rOCT1 for these substrates could be detected, in contrast to what was observed in WT. PKC activation had no significant effect on rOCT1 trafficking from intracellular pools to the cell membrane. Substitution of all PKC sites suppressed PKC-induced phosphorylation of rOCT1. In conclusion, it was found that the presence of all five potential PKC phosphorylation sites is necessary for the PKC-induced stimulation of rOCT1. The different effects on the EC(50) values by the different mutations suggest that the large intracellular loop participates in building the substrate binding pocket of rOCT1 or specifically modulates its structure.
- Published
- 2005
- Full Text
- View/download PDF
41. 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.
- Author
-
Popp C, Gorboulev V, Müller TD, Gorbunov D, Shatskaya N, and Koepsell H
- Subjects
- Amino Acid Sequence, Amino Acids chemistry, Animals, Female, Membrane Transport Proteins chemistry, Molecular Sequence Data, Organic Anion Transport Protein 1 chemistry, Protein Structure, Tertiary physiology, Substrate Specificity physiology, Xenopus laevis, Amino Acids metabolism, Membrane Transport Proteins metabolism, Models, Molecular, Organic Anion Transport Protein 1 metabolism
- Abstract
To identify functionally relevant amino acids in the rat organic cation transporter 1 (rOCT1), 18 consecutive amino acids in the presumed fourth transmembrane alpha helix (TMH) were mutated and functionally characterized after expression in Xenopus laevis oocytes. After mutation of three amino acids on successive turns of the alpha helix, K(m) values for tetraethylammonium (TEA) and/or 1-methyl-4-phenylpyridinium (MPP) were decreased. After replacement of Trp218 by tyrosine (W218Y) and Tyr222 by leucine (Y222L), the K(m) values for both TEA and MPP were decreased. In mutants Y222F and T226A, only the K(m) values for TEA and MPP were decreased, respectively. The data suggest that amino acids Trp218 and Tyr222 participate in the binding of both TEA and MPP, whereas Thr226 is only involved in the binding of MPP. Using the crystal structure of the lactose permease LacY from Escherichia coli that belongs to the same major facilitator superfamily as rOCT1, we modeled the tertiary structure of the presumed 12 transmembrane alpha helices. The validity of the model was suggested because seven amino acids that have been shown to participate in the binding of cations by mutagenesis experiments [fourth TMH Trp218, Tyr222, and Thr226 (this work); 10th TMH Ala443, Leu447, and Gln448 (companion work in this issue of Molecular Pharmacology); 11th TMH Asp475 (previous report)] are located in one region surrounding a large cleft that opens to the intracellular side. The dimensions of TEA in comparison with the interacting amino acids in the modeled cleft suggest that more than one TEA molecule can bind in parallel to the modeled conformation of the transporter.
- Published
- 2005
- Full Text
- View/download PDF
42. Subtype-specific affinity for corticosterone of rat organic cation transporters rOCT1 and rOCT2 depends on three amino acids within the substrate binding region.
- Author
-
Gorboulev V, Shatskaya N, Volk C, and Koepsell H
- Subjects
- Amino Acid Sequence genetics, Amino Acids genetics, Animals, Binding Sites physiology, Catecholamine Plasma Membrane Transport Proteins, Corticosterone pharmacology, Dose-Response Relationship, Drug, Female, Membrane Transport Proteins genetics, Molecular Sequence Data, Organic Cation Transport Proteins genetics, Organic Cation Transporter 2, Rats, Substrate Specificity physiology, Xenopus laevis, Amino Acids metabolism, Corticosterone metabolism, Membrane Transport Proteins metabolism, Organic Cation Transport Proteins metabolism
- Abstract
The affinity of corticosterone to organic cation transporters (OCTs) is subtype- and species-dependent. For example, the IC50 values for corticosterone inhibition of cation uptake by transporters rOCT1 and rOCT2 are approximately 150 and approximately 4 microM, respectively. By introducing domains and amino acids from rOCT2 into rOCT1, we found that the exchange of three amino acids in the presumed 10th transmembrane alpha helix is sufficient to increase the affinity of rOCT1 for corticosterone to that of rOCT2. Replacement of these amino acids in rOCT2 decreased the affinity for corticosterone. These amino acids (Ala443, Leu447, and Gln448 in rOCT1 and Ile443, Tyr447, and Glu448 in rOCT2) are probably located within the substrate binding region because in rOCT1 mutants, the K(m) values for uptake of tetraethylammonium (TEA) and 1-methyl-4-phenylpyridinium (MPP) were decreased in parallel with a decrease of the IC50 values for the inhibition of cation uptake by corticosterone. In mutant rOCT1(L447Y/Q448E), the IC50 value for the inhibition of [3H]MPP (0.1 microM) uptake by corticosterone (24 +/- 4 microM) was significantly higher compared with the IC50 value for inhibition of [14C]TEA (10 microM) uptake (5.3 +/- 1.7 microM). This finding suggests an allosteric interaction between transported cation and corticosterone. Because this substrate-specific effect cannot be explained by differential replacement of corticosterone by MPP versus TEA and was observed after point mutations within the presumed substrate region, the data suggest that MPP or TEA bind to the substrate binding region simultaneously with corticosterone and cause a short-range allosteric effect on the corticosterone binding site.
- Published
- 2005
- Full Text
- View/download PDF
43. Mice without the regulator gene Rsc1A1 exhibit increased Na+-D-glucose cotransport in small intestine and develop obesity.
- Author
-
Osswald C, Baumgarten K, Stümpel F, Gorboulev V, Akimjanova M, Knobeloch KP, Horak I, Kluge R, Joost HG, and Koepsell H
- Subjects
- Animals, Biological Transport, Blotting, Northern, Blotting, Southern, Blotting, Western, Cholesterol blood, Cloning, Molecular, Enzyme-Linked Immunosorbent Assay, Female, Glucose Transporter Type 2, Insulin metabolism, Intestinal Mucosa metabolism, Introns, Leptin metabolism, Male, Mice, Mice, Knockout, Microscopy, Fluorescence, Models, Genetic, Phenotype, Polymerase Chain Reaction, RNA Processing, Post-Transcriptional, Sex Factors, Sodium-Glucose Transporter 1, Time Factors, Transcription, Genetic, Transfection, Glucose metabolism, Intestine, Small metabolism, Membrane Glycoproteins metabolism, Monosaccharide Transport Proteins genetics, Monosaccharide Transport Proteins metabolism, Monosaccharide Transport Proteins physiology, Obesity genetics, Sodium metabolism, Up-Regulation
- Abstract
The product of the intronless single copy gene RSC1A1, named RS1, is an intracellular 617-amino-acid protein that is involved in the regulation of the Na(+)-d-glucose cotransporter SGLT1. We generated and characterized RS1 knockout (RS1(-/-) mice. In the small intestines of RS1(-/-) mice, the SGLT1 protein was up-regulated sevenfold compared to that of wild-type mice but was not changed in the kidneys. The up-regulation of SGLT1 was posttranscriptional. Small intestinal d-glucose uptake measured in jointly perfused small bowel and liver was increased twofold compared to that of the wild-type, with increased peak concentrations of d-glucose in the portal vein. At birth, the weights of RS1(-/-) and wild-type mice were similar. At the age of 3 months, male RS1(-/-) mice had 5% higher weights and 15% higher food intakes, whereas their energy expenditures and serum leptin concentrations were similar to those of wild-type mice. At the age of 5 months, male and female RS1(-/-) mice were obese, with 30% increased body weight, 80% increased total fat, and 30% increased serum cholesterol. At this age, serum leptin was increased, whereas food intake was the same as for wild-type mice. The data suggest that the removal of RS1 leads to leptin-independent up-regulation of food intake, which causes obesity.
- Published
- 2005
- Full Text
- View/download PDF
44. Regulation of the human organic cation transporter hOCT1.
- Author
-
Ciarimboli G, Struwe K, Arndt P, Gorboulev V, Koepsell H, Schlatter E, and Hirsch JR
- Subjects
- Animals, Calmodulin antagonists & inhibitors, Calmodulin metabolism, Cell Line, Cricetinae, Cyclic GMP metabolism, Enzyme Activation, Fluorescence, Gene Expression Regulation, Humans, Organic Cation Transporter 1 antagonists & inhibitors, Organic Cation Transporter 1 genetics, Protein Kinases metabolism, Pyridinium Compounds pharmacology, Receptors, G-Protein-Coupled metabolism, Spectrometry, Fluorescence, Substrate Specificity, Organic Cation Transporter 1 metabolism
- Abstract
The human organic cation transporter type 1 (hOCT1) is an important transport system for small organic cations in the liver. Organic cation transporters are regulated by different signaling pathways, but the regulation of hOCT1 has not yet been studied. In this work, we have for the first time investigated the regulation of hOCT1. hOCT1 was expressed in Chinese hamster ovary cells (CHO-hOCT1) and in human embryonic kidney cells (HEK293-hOCT1). Its activity was monitored using microfluorimetry with the fluorescent organic cation 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP(+)) as substrate. hOCT1 expressed in CHO-cells was inhibited by protein kinase A (PKA) activation (1 microM forskolin, -58 +/- 6%, n = 12), calmodulin inhibition (0.1 microM calmidazolium, -68 +/- 3%, n = 6; 10 microM ophiobolin A, -48 +/- 10%, n = 7), calmodulin-dependent kinase II inhibition (1 microM KN62, -78 +/- 4%, n = 12), and inhibition of p56(lck) tyrosine kinase (10 microM aminogenistein, -35 +/- 7%, n = 12). The apparent affinities for TEA(+) were lower in CHO-hOCT1 than in HEK293-hOCT1, while those for TPA(+) and quinine were almost identical; the rank order of EC(50) values (TPA(+) > quinine > TEA(+)) was independent of the expression system. EC(50) values for TEA(+) in CHO-hOCT1 or HEK293-hOCT1 were increased under calmidazolium incubation (6.3 and 1.4 mM, respectively). hOCT1 was inhibited by PKA and endogenously activated by calmodulin, calmodulin-dependent kinase II, and p56(lck) tyrosine kinase. Regulation pathways were the same in the two expression systems. Since apparent substrate affinities depend on activity of regulatory pathways, the expression system plays a role in determining the substrate affinities.
- Published
- 2004
- Full Text
- View/download PDF
45. Na(+)-D-glucose cotransporter in muscle capillaries increases glucose permeability.
- Author
-
Elfeber K, Stümpel F, Gorboulev V, Mattig S, Deussen A, Kaissling B, and Koepsell H
- Subjects
- Animals, Biological Transport, Blotting, Western, Capillary Permeability, Cell Membrane metabolism, Glucose Transporter Type 4, Immunohistochemistry, Insulin metabolism, Intestine, Small blood supply, Male, Membrane Glycoproteins chemistry, Microscopy, Fluorescence, Monosaccharide Transport Proteins biosynthesis, Monosaccharide Transport Proteins chemistry, Muscle, Skeletal metabolism, Myocardium cytology, Myocardium metabolism, Perfusion, Polymerase Chain Reaction, Rats, Rats, Wistar, Reverse Transcriptase Polymerase Chain Reaction, Sodium-Glucose Transporter 1, Submandibular Gland blood supply, Time Factors, Capillaries metabolism, Glucose metabolism, Membrane Glycoproteins metabolism, Monosaccharide Transport Proteins metabolism, Muscle Proteins, Muscles blood supply
- Abstract
By immunohistochemistry, we demonstrated the localization of the Na(+)-D-glucose cotransporter SGLT1 in capillaries of rat heart and skeletal muscle, but not in capillaries of small intestine and submandibular gland. mRNA of SGLT1 was identified in skeletal muscle and primary cultured coronary endothelial cells. The functional relevance of SGLT1 for glucose transport across capillary walls in muscle was tested by measuring the extraction of D-glucose from the perfusate during non-recirculating perfusion of isolated rat hindlimbs. In this model, D-glucose extraction from the perfusate is increased by insulin which accelerates D-glucose uptake into myocytes by increasing the concentration of glucose transporter GLUT4 in the plasma membrane. The insulin-induced increase of D-glucose extraction from the perfusate was abolished after blocking SGLT1 with the specific inhibitor phlorizin. The data show that SGLT1 in capillaries of skeletal muscle is required for the action of insulin on D-glucose supply of myocytes.
- Published
- 2004
- Full Text
- View/download PDF
46. Different affinities of inhibitors to the outwardly and inwardly directed substrate binding site of organic cation transporter 2.
- Author
-
Volk C, Gorboulev V, Budiman T, Nagel G, and Koepsell H
- Subjects
- Animals, Biological Transport, Corticosterone pharmacology, Electrophysiology, Membrane Transport Proteins metabolism, Oocytes, Organic Cation Transport Proteins antagonists & inhibitors, Organic Cation Transport Proteins genetics, Organic Cation Transporter 2, Rats, Transfection, Xenopus laevis, Membrane Transport Modulators, Membrane Transport Proteins antagonists & inhibitors, Organic Cation Transport Proteins metabolism, Quaternary Ammonium Compounds pharmacology
- Abstract
The rat organic cation transporter 2 (rOCT2) was expressed in Xenopus laevis oocytes and cation-induced outward and inward currents were measured in whole cells and giant patches using voltage clamp techniques. Tetrabutylammonium (TBuA) and corticosterone were identified as nontransported inhibitors that bind to the substrate binding site of rOCT2. They inhibited cation-induced currents from both membrane sides. Increased substrate concentrations could partially overcome the inhibition. At 0 mV, the affinity of TBuA from the extracellular side compared with the intracellular side of the membrane was 4-fold higher, whereas the affinity of corticosterone was 20-fold lower. The data suggest that the substrate binding site of rOCT2 is like a pocket containing overlapping binding domains for ligands. These binding domains may undergo separate structural changes. From the extracellular surface, the affinity for uncharged corticosterone was increased by making membrane potential more negative. This implies potential-dependent structural changes in the extracellular binding pocket and existence of a voltage sensor. Interestingly, at 0 mV, an 18-fold higher affinity was determined for trans-inhibition of choline efflux by corticosterone compared with cis-inhibition of choline uptake. This suggests an additional high affinity-conformation of the empty outwardly oriented substrate binding pocket. A model is proposed that describes how substrates and inhibitors might interact with rOCT2. The data provide a theoretical basis to understand drug-drug interactions at polyspecific transporters for organic cations.
- Published
- 2003
- Full Text
- View/download PDF
47. A glucose sensor hiding in a family of transporters.
- Author
-
Diez-Sampedro A, Hirayama BA, Osswald C, Gorboulev V, Baumgarten K, Volk C, Wright EM, and Koepsell H
- Subjects
- Animals, Base Sequence, Chromosomes, Human, Pair 22, Cloning, Molecular, DNA Primers, Humans, Immunohistochemistry, Monosaccharide Transport Proteins genetics, Reverse Transcriptase Polymerase Chain Reaction, Sodium-Glucose Transport Proteins, Xenopus laevis, Monosaccharide Transport Proteins physiology
- Abstract
We have examined the expression and function of a previously undescribed human member (SGLT3/SLC5A4) of the sodium/glucose cotransporter gene family (SLC5) that was first identified by the chromosome 22 genome project. The cDNA was cloned and sequenced, confirming that the gene coded for a 659-residue protein with 70% amino acid identity to the human SGLT1. RT-PCR and Western blotting showed that the gene was transcribed and mRNA was translated in human skeletal muscle and small intestine. Immunofluorescence microscopy indicated that in the small intestine the protein was expressed in cholinergic neurons in the submucosal and myenteric plexuses, but not in enterocytes. In skeletal muscle SGLT3 immunoreactivity colocalized with the nicotinic acetylcholine receptor. Functional studies using the Xenopus laevis oocyte expression system showed that hSGLT3 was incapable of sugar transport, even though SGLT3 was efficiently inserted into the plasma membrane. Electrophysiological assays revealed that glucose caused a specific, phlorizin-sensitive, Na+-dependent depolarization of the membrane potential. Uptake assays under voltage clamp showed that the glucose-induced inward currents were not accompanied by glucose transport. We suggest that SGLT3 is not a Na+/glucose cotransporter but instead a glucose sensor in the plasma membrane of cholinergic neurons, skeletal muscle, and other tissues. This points to an unexpected role of glucose and SLC5 proteins in physiology, and highlights the importance of determining the tissue expression and function of new members of gene families.
- Published
- 2003
- Full Text
- View/download PDF
48. Organic cation transporter capable of transporting serotonin is up-regulated in serotonin transporter-deficient mice.
- Author
-
Schmitt A, Mössner R, Gossmann A, Fischer IG, Gorboulev V, Murphy DL, Koepsell H, and Lesch KP
- Subjects
- Animals, Brain Stem metabolism, Carrier Proteins genetics, Hippocampus metabolism, In Situ Hybridization, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Organ Specificity, Organic Cation Transport Proteins genetics, Organic Cation Transporter 1 genetics, Organic Cation Transporter 1 metabolism, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Serotonin Plasma Membrane Transport Proteins, Up-Regulation genetics, Brain metabolism, Membrane Glycoproteins deficiency, Membrane Transport Proteins, Nerve Tissue Proteins, Organic Cation Transport Proteins metabolism, Serotonin metabolism
- Abstract
The serotonin (5HT) transporter (5HTT) regulates serotonergic neurotransmission by mediating the reuptake of 5HT from the synaptic cleft. Although lacking the high affinity and selectivity of the 5HTT, the brain expresses a large number of other transporters, including the polyspecific organic cation transporters (OCTs). OCT1 and OCT3, members of the potential-sensitive organic cation transporter gene family, physiologically transport a wide spectrum of organic cations. In addition, both transporters mediate low-affinity 5HT transport and, therefore, may participate in the clearance of excessive 5HT. Because concentrations of extracellular 5HT are increased in the brain of 5HTT-deficient mice, they are a model for investigating the role of OCTs in 5HT system homeostasis. Here, we analyzed OCT1 and OCT3 gene expression in the brain of 5HTT knockout mice by semiquantitative competitive polymerase chain reaction and in situ hybridization. We demonstrate that, in 5HTT-deficient mice, OCT3 mRNA concentrations were significantly increased in the hippocampus, but not in other brain regions, including cortex, striatum, cerebellum, and brainstem. In contrast, no difference in OCT1 expression was detected between 5HTT knockout and control mice. Up-regulation of OCT3 expression and enhanced low-affinity 5HT uptake may limit the adverse effects of elevated extracellular 5HT and may play a critical role in maintaining 5HT-dependent functions of the hippocampus in the absence of 5HTT., (Copyright 2002 Wiley-Liss, Inc.)
- Published
- 2003
- Full Text
- View/download PDF
49. Regulation of human organic cation transporter hOCT2 by PKA, PI3K, and calmodulin-dependent kinases.
- Author
-
Cetinkaya I, Ciarimboli G, Yalçinkaya G, Mehrens T, Velic A, Hirsch JR, Gorboulev V, Koepsell H, and Schlatter E
- Subjects
- Binding, Competitive, Calcium physiology, Cell Line, Electrochemistry, Enzyme Activation physiology, Humans, Organic Cation Transport Proteins antagonists & inhibitors, Organic Cation Transporter 2, Protein Kinase C metabolism, Pyridinium Compounds pharmacokinetics, Substrate Specificity, Calcium-Calmodulin-Dependent Protein Kinases physiology, Cyclic AMP-Dependent Protein Kinases physiology, Organic Cation Transport Proteins metabolism, Phosphatidylinositol 3-Kinases physiology
- Abstract
Properties and regulation of the human organic cation (OC) transporter type 2 (hOCT2) expressed in HEK-293 cells were extensively characterized using the fluorescent OC 4-[4-(dimethylamino)styryl]-N-methylpyridinium (ASP(+)). ASP(+) uptake was electrogenic and inhibited by TPA(+) (EC(50) = 2.7 microM), tetraethylammonium (EC(50) = 35 microM), cimetidine (EC(50) = 36 microM), or quinine (EC(50) = 6.7 microM). Stimulation with carbachol or ATP decreased initial uptake by 44 +/- 3 (n = 14) and 34 +/- 4% (n = 21), respectively, independently of PKC but dependent on phosphatidylinositol 3-kinase (PI3K). PKA stimulation decreased uptake by 18 +/- 4% (n = 40). Inhibition of calmodulin (CaM), Ca(2+)/CaM-dependent kinase II, or myosin light chain kinase decreased uptake by 63 +/- 2 (n = 15), 40 +/- 4 (n = 30), and 31 +/- 4% (n = 16), respectively. Inhibition of CaM resulted in a significant change in the EC(50) value for the inhibition of ASP(+) uptake by tetraethylammonium. In conclusion, we demonstrate that the hOCT2 is inhibited by PI3K and PKA and activated by a CaM-dependent signaling pathway, probably via a change in substrate affinity.
- Published
- 2003
- Full Text
- View/download PDF
50. The cation transporters rOCT1 and rOCT2 interact with bicarbonate but play only a minor role for amantadine uptake into rat renal proximal tubules.
- Author
-
Goralski KB, Lou G, Prowse MT, Gorboulev V, Volk C, Koepsell H, and Sitar DS
- Subjects
- Animals, Cell Line, Female, Humans, Kidney Tubules, Proximal cytology, Kidney Tubules, Proximal drug effects, Male, Organic Cation Transport Proteins antagonists & inhibitors, Organic Cation Transporter 1 antagonists & inhibitors, Organic Cation Transporter 2, Rats, Rats, Sprague-Dawley, Xenopus laevis, Amantadine metabolism, Bicarbonates metabolism, Kidney Tubules, Proximal metabolism, Organic Cation Transport Proteins metabolism, Organic Cation Transporter 1 metabolism
- Abstract
In renal proximal tubules, the organic cation transporters rOCT1 and rOCT2 are supposed to mediate the first step in organic cation secretion. We investigated whether previously described differences in amantadine and tetraethylammonium (TEA) uptake into isolated renal proximal tubules could be explained by differences in their transport by rOCT1 and rOCT2. By expressing rOCT1 and rOCT2 in Xenopus oocytes and HEK 293 cells, we demonstrated that both transporters translocated amantadine. In Xenopus oocytes, the inhibitory potency of several rOCT1/2 inhibitors was similar for amantadine compared to TEA uptake and supports amantadine transport by rOCT1 and rOCT2. In proximal tubules, procainamide, quinine, cyanine(863), choline, and guanidine in concentrations that inhibit rOCT1/2-mediated TEA or amantadine uptake in Xenopus oocytes exhibited no effect on amantadine uptake. At variance, these inhibitors blocked TEA uptake into proximal tubules. Amantadine and TEA transport were sensitive to modulation by 25 mM bicarbonate. The effect of bicarbonate on organic cation transport was dependent on substrate (amantadine or TEA), cell system (oocytes, HEK 293 cells, or proximal tubules), and transporter (rOCT1 or rOCT2). In proximal tubules, only amantadine uptake was stimulated by bicarbonate. The data suggested that rat renal proximal tubules contain an organic cation transporter in addition to rOCT1 and rOCT2 that mediates amantadine uptake and requires bicarbonate for optimal function. TEA uptake by the basolateral membrane may be mediated mainly by rOCT1 and rOCT2, but these transporters may be in a different functional or regulatory state when expressed in cells or oocytes compared with expression in vivo.
- Published
- 2002
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.