Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 293
Filter
1.
Can J Physiol Pharmacol ; 87(10): 821-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20052008

ABSTRACT

Previous studies showed that amantadine transport increased while tetraethylammonium (TEA) transport decreased in kidney tissue from diabetic rats. Changes in transport activity were reversed by exogenous insulin. We hypothesized that this difference in transport regulation is due to differential regulation of different transport systems. Native human embryonic kidney cortex cells (HEK293 cell line) and rat organic cation transporter (rOCT)-transfected cells were used to test the hypothesis. In support of differential regulation, short-term glucose starvation stimulated amantadine transport and inhibited TEA transport, but the effect was bicarbonate-modulated only for amantadine. cAMP analogues inhibited TEA transport while stimulating amantadine transport. This effect was additive to the effect of insulin, and the presence of bicarbonate affected the extent of the change. Our findings indicated that regulation of rOCT 1 and 2 was mediated by transmembrane adenylyl cyclase, and regulation of amantadine transport was mediated by soluble adenylyl cyclase, suggesting that intracellular microdomains of cAMP may be important in determining overall cellular transport for organic cations. Soluble adenylyl cyclase activity is known to be modulated by bicarbonate and lactate. These observations support our hypothesis and reconcile our previous studies demonstrating increased transport affinity for amantadine in the presence of bicarbonate and decreased transport affinity in the presence of lactate.


Subject(s)
Organic Anion Transporters/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Adenylyl Cyclases/metabolism , Amantadine/pharmacology , Bicarbonates/pharmacology , Biological Transport, Active/drug effects , Blood Glucose/metabolism , Cell Line , Colforsin/pharmacology , Cyclic AMP/metabolism , Glucose/deficiency , Humans , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Organic Anion Transport Protein 1/metabolism , Organic Anion Transporters, Sodium-Independent/metabolism , Recombinant Proteins/metabolism , Tetraethylammonium Compounds/metabolism
2.
Mol Pharm ; 4(4): 600-7, 2007.
Article in English | MEDLINE | ID: mdl-17616214

ABSTRACT

Throughout the mammalian spermatogenic pathway, differentiating spermatogenic cells remain in close contact with somatic Sertoli cells, and this has been considered to be essential for the proliferation, differentiation, and survival of spermatogenic cells. It is thought that Sertoli cells form tight junctions to protect developing spermatogenic cells against harmful agents and provide several nutrients for spermatogenesis from the blood stream. Accordingly, Sertoli cells should regulate the movement of various nutritious and xenobiotic compounds by selective membrane transporters. However, the information on membrane transporters in Sertoli cells is limited. In the present study, we characterized the transport systems of organic cations in Sertoli cells. Uptake of [14C]tetraethylammonium (TEA) was measured by primary-cultured rat Sertoli cells. Initial uptake of TEA was concentration dependent, and an Eadie-Hofstee plot indicated the involvement of two saturable transport systems. The apparent Km values of high- and low-affinity components were comparable to those of previously known organic cation transporter (OCT) or organic cation/carnitine transporter (OCTN). In addition, OCT1, OCT3, OCTN1, and OCTN2 were expressed in Sertoli cells. In conclusion, multiple organic cation transporters, OCTs and OCTNs are expressed in Sertoli cells and the cells regulate transport of cationic compounds to protect and/or maintain the spermatogenesis in testis as the blood-testis barrier.


Subject(s)
Blood-Testis Barrier/metabolism , Organic Cation Transport Proteins/metabolism , Testis/metabolism , Animals , Biological Transport, Active , Carnitine/metabolism , Carnitine/pharmacology , Cell Culture Techniques , Cell Polarity/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Hydrogen-Ion Concentration , Male , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Sertoli Cells/metabolism , Tetraethylammonium Compounds/antagonists & inhibitors , Tetraethylammonium Compounds/metabolism , Tetraethylammonium Compounds/pharmacology
3.
Biochem Pharmacol ; 74(2): 359-71, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17509534

ABSTRACT

The substrate specificities of human (h) multidrug and toxin extrusion (MATE) 1 and hMATE2-K were examined to find functional differences between these two transporters by the transfection of the cDNA of hMATE1 and hMATE2-K into HEK293 cells. Western blotting revealed specific signals for hMATE1 and hMATE2-K consistent with a size of 50 and 40kDa, respectively, in the transfectants as well as human renal brush-border membranes under reducing conditions. In the presence of oppositely directed H(+)-gradient, the transport activities of various compounds such as tetraethylammonium, 1-methyl-4-phenylpyridinium, cimetidine, metformin, creatinine, guanidine, procainamide, and topotecan were stimulated in hMATE1- and hMATE2-K-expressing cells. In addition to cationic compounds, anionic estrone sulfate, acyclovir, and ganciclovir were also recognized as substrates of these transporters. Kinetic analyses demonstrated the Michaelis-Menten constants for the hMATE1-mediated transport of tetraethylammonium, 1-methyl-4-phenylpyridinium, cimetidine, metformin, guanidine, procainamide, topotecan, estrone sulfate, acycrovir, and ganciclovir to be (in mM) 0.38, 0.10, 0.17, 0.78, 2.10, 1.23, 0.07, 0.47, 2.64, and 5.12, respectively. Those for hMATE2-K were 0.76, 0.11, 0.12, 1.98, 4.20, 1.58, 0.06, 0.85, 4.32, and 4.28, respectively. Although their affinity for hMATE1 and hMATE2-K was similar, the zwitterionic cephalexin and cephradine were revealed to be specific substrates of hMATE1, but not of hMATE2-K. Levofloxacin and ciprofloxacin were not transported, but were demonstrated to be potent inhibitors of these transporters. These results suggest that hMATE1 and hMATE2-K function together as a detoxication system, by mediating the tubular secretion of intracellular ionic compounds across the brush-border membranes of the kidney.


Subject(s)
Kidney/metabolism , Organic Cation Transport Proteins/physiology , Toxins, Biological/metabolism , Cells, Cultured , Cephalexin/metabolism , Humans , Protons , Substrate Specificity , Tetraethylammonium Compounds/metabolism
4.
Am J Physiol Renal Physiol ; 293(1): F21-7, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17327500

ABSTRACT

The effects of protein kinases MAPK and PKA on the regulation of organic cation transporter 2 (OCT2) were investigated both in a heterologous cell system [Chinese hamster ovary (CHO-K1) cells stably transfected with rabbit (rb)OCT2] and in native intact rabbit renal proximal S2 segments. Inhibition of MEK (by U-0126) or PKA (by H-89) reduced transport activity of rbOCT2 in CHO-K1 cells. The inhibitory effect of U-0126 combined with H-89 produced no additive effect, indicating that the action of PKA and MAPK in the regulation of rbOCT2 is in a common pathway. Activation of PKA by forskolin stimulated rbOCT2 activity, and this stimulatory effect was eliminated by H-89, indicating that the stimulation required PKA activation. In S2 segments of rabbit renal proximal tubules, activation of MAPK (by EGF) and PKA (by forskolin) stimulated activity of rbOCT2, and this activation was abolished by U-0126 and H-89, respectively. This is the first study to show that MAPK and PKA are involved, apparently in a common pathway, in the regulation of OCT2 activity in both a heterologous cell system and intact renal proximal tubules.


Subject(s)
Cations/metabolism , Cyclic AMP-Dependent Protein Kinases/physiology , Kidney/metabolism , Mitogen-Activated Protein Kinases/physiology , Organic Cation Transport Proteins/physiology , Algorithms , Animals , Biological Transport, Active/physiology , Butadienes/pharmacology , CHO Cells , Colforsin/pharmacology , Cricetinae , Cricetulus , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Enzyme Activators/metabolism , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Isoquinolines/pharmacology , Kidney Tubules, Proximal/enzymology , Kidney Tubules, Proximal/metabolism , Kinetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Nitriles/pharmacology , Organic Cation Transport Proteins/antagonists & inhibitors , Rabbits , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , Tetraethylammonium Compounds/metabolism
5.
Am J Physiol Renal Physiol ; 292(2): F593-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17047166

ABSTRACT

Recently, we have isolated the rat (r) H(+)/organic cation antiporter multidrug and toxin extrusion 1 (MATE1) and reported its tissue distribution and transport characteristics. Functional characterization suggested that an oppositely directed H(+) gradient serves as a driving force for the transport of a prototypical organic cation, tetraethylammonium, by MATE1, but there is no direct evidence to prove this. In the present study, therefore, we elucidated the driving force of tetraethylammonium transport via rMATE1 using plasma membrane vesicles isolated from HEK293 cells stably expressing rMATE1 (HEK-rMATE1 cells). A 70-kDa rMATE1 protein was confirmed to exist in HEK-rMATE1 cells, and the transport of various organic cations including [(14)C]tetraethylammonium was stimulated in intracellular acidified HEK-rMATE1 cells but not mock cells. The transport of [(14)C]tetraethylammonium in membrane vesicles from HEK-rMATE1 cells exhibited the overshoot phenomenon only when there was an outwardly directed H(+) gradient, as observed in rat renal brush-border membrane vesicles. The overshoot phenomenon was not observed in the vesicles from mock cells. The stimulated [(14)C]tetraethylammonium uptake by an H(+) gradient [intravesicular H(+) concentration ([H(+)](in)) > extravesicular H(+) concentration ([H(+)](out))] was significantly reduced in the presence of a protonophore, carbonyl cyanide p-trifluoromethoxyphenylhydrazone (FCCP). [(14)C]tetraethylammonium uptake was not changed in the presence of valinomycin-induced membrane potential. These findings definitively indicate that an oppositely directed H(+) gradient serves as a driving force of tetraethylammonium transport via rMATE1, and this is the first demonstration to identify the driving force of the MATE family. The present experimental strategy is very useful in identifying the driving force of cloned transporters whose driving force has not been evaluated.


Subject(s)
Antiporters/physiology , Hydrogen/physiology , Organic Cation Transport Proteins/physiology , Animals , Cell Line , Cell Membrane/metabolism , Humans , Rats , Tetraethylammonium Compounds/metabolism
6.
J Pharmacol Exp Ther ; 303(2): 574-80, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12388638

ABSTRACT

The secretory transport of the H(2)-antagonists, ranitidine and famotidine, across Caco-2 cell monolayers was found to be a saturable process. Both drugs exhibited greater permeability in the basolateral (BL) to apical (AP) direction than in the AP to BL direction, indicating apically directed secretion; BL to AP transport was inhibited by P-glycoprotein (P-gp) inhibitors verapamil and cyclosporin A. The cellular uptake of ranitidine across the BL membrane was saturable and temperature dependent, indicative of carrier-mediated transport. The K(m) and V(max) for the uptake process were estimated to be 66.9 mM and 20.9 nmol/mg of protein/min, respectively. The uptake of [(14)C]ranitidine across the BL membrane was inhibited by unlabeled ranitidine and structurally diverse organic cations. The tetraethylammonium (TEA)-sensitive organic cation transporters are not involved in the uptake of ranitidine and famotidine across the BL membrane of Caco-2. This conclusion was based on the evidence that functionally active TEA-sensitive organic cation transporters did not exist in the BL membranes of the Caco-2 cells, whereas the functionally active TEA-sensitive organic cation transporter(s) in LLC-PK(1) cells did not contribute to the transport of ranitidine or famotidine across the cell monolayers. Thus, we conclude that the secretory transport of ranitidine and famotidine across Caco-2 cell monolayers is mediated by 1) a carrier in the BL membrane that is distinct from the TEA-sensitive organic cation transporter(s) and 2) P-gp in the apical membrane.


Subject(s)
Famotidine/metabolism , Histamine H2 Antagonists/metabolism , Ranitidine/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Biological Transport, Active/drug effects , Caco-2 Cells , Humans , LLC-PK1 Cells , Organic Cation Transport Proteins/metabolism , Swine , Tetraethylammonium Compounds/metabolism , Tight Junctions/drug effects
7.
J Biol Chem ; 274(47): 33388-92, 1999 Nov 19.
Article in English | MEDLINE | ID: mdl-10559218

ABSTRACT

Novel organic cation transporter (OCTN2) is an organic cation/carnitine transporter, and two missense mutations, L352R and P478L, in OCTN2 have been identified as the cause for primary carnitine deficiency. In the present study, we assessed the influence of these two mutations on the carnitine transport function and the organic cation transport function of OCTN2. The L352R mutation resulted in a complete loss of both transport functions. In contrast, the P478L mutation resulted in a complete loss of only the carnitine transport function but significantly stimulated the organic cation transport function. Studies with human OCTN2/rat OCTN2 chimeric transporters indicated that the carnitine transport site and the organic cation transport site were not identical. Because carnitine transport is Na(+)-dependent whereas organic cation transport is Na(+)-independent, we investigated the possibility that the P478L mutation affected Na(+) binding. The Na(+) activation kinetics were found to be similar for the P478L mutant and wild type OCTN2. We then mutated nine different tyrosine residues located in or near transmembrane domains and assessed the transport function of these mutants. One of these mutations, Y211F, was found to have differential influence on the two transport activities of OCTN2 as did the P478L mutation. However, the Na(+) activation kinetics were not affected. These findings are of clinical relevance to patients with primary carnitine deficiency because whereas each and every mutation in these patients is expected to result in the loss of the carnitine transport function, all of these mutations may not interfere with the organic cation transport function.


Subject(s)
Carnitine/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Organic Cation Transport Proteins , Animals , Carrier Proteins/genetics , Cations , HeLa Cells , Humans , Ion Transport , Kinetics , Membrane Proteins/genetics , Mutagenesis, Site-Directed , Rats , Solute Carrier Family 22 Member 5 , Tetraethylammonium Compounds/metabolism , Tyrosine/metabolism
8.
J Membr Biol ; 172(3): 215-23, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10568791

ABSTRACT

P2X(2) purinoceptors are cation-selective channels activated by ATP and its analogues. Using single channel measurements we studied the channel's selectivity for the alkali metal ions and organic monovalent cations NMDG(+), Tris(+), TMA(+), and TEA(+). The selectivity sequence for currents carried by alkali metal ions is: K(+) > Rb(+) > Cs(+) > Na(+) > Li(+), which is Eisenman sequence IV. This is different from the mobility sequence of the ions in free solution suggesting there is weak interaction between the ions and the channel interior. The relative conductance for alkali ions increases linearly in relation to the Stokes radius. The organic ions NMDG(+), Tris(+), TMA(+) and TEA(+) were virtually impermeant. The divalent ions (Mn(2+), Mg(2+), Ca(2+) and Ba(2+)) induced a fast block visible as a reduction in amplitude of the unitary currents. Using a single-site binding model, the divalent ions exhibited an equilibrium affinity sequence of Mn(2+) > Mg(2+) > Ca(2+) > Ba(2+).


Subject(s)
Metals, Alkali/metabolism , Receptors, Purinergic P2/physiology , Adenosine Triphosphate/metabolism , Cell Line , Cesium/metabolism , Electrophysiology , Humans , Lithium/metabolism , Meglumine/metabolism , Potassium/metabolism , Quaternary Ammonium Compounds/metabolism , Receptors, Purinergic P2X2 , Rubidium/metabolism , Sodium/metabolism , Tetraethylammonium Compounds/metabolism , Tromethamine/metabolism
9.
J Pharmacol Exp Ther ; 289(2): 768-73, 1999 May.
Article in English | MEDLINE | ID: mdl-10215651

ABSTRACT

In the present study, functional characteristics of organic cation transporter (OCTN)1, which was cloned as the pH-dependent tetraethylammonium (TEA) transporter when expressed in mammalian human embryonic kidney (HEK)293 cells, were further investigated using Xenopus oocytes as well as HEK293 cells as gene expression systems. When OCTN1-derived complementary RNA was injected into Xenopus oocytes, pH-dependent transport of [14C]TEA was observed as the same in HEK293 cells. In contrast, a replacement of sodium ions with potassium ions in the surrounding medium did not cause any change in [14C]TEA uptake in Xenopus oocytes expressed with OCTN1. In addition, when OCTN1 was expressed in HEK293 cells, efflux of TEA from the cells was pH dependent, with an accelerated rate at acidic external medium pH. Accordingly, membrane potential or sodium ions are suggested to have no influence on [14C]TEA transport and the transport activity of OCTN1 is directly affected by pH itself. Furthermore, addition of the unlabeled TEA in external medium enhanced the efflux of preloaded [14C]TEA. These observations suggest that OCTN1 is a pH-dependent and bidirectional TEA transporter. OCTN1-mediated [14C]TEA uptake was inhibited by various organic cations such as cimetidine, procainamide, pyrilamine, quinidine, quinine, and verapamil. In addition, uptakes of cationic compounds such as [3H]pyrilamine, [3H]quinidine, and [3H]verapamil and zwitterionic L-[3H]carnitine were increased by expression of OCTN1 in Xenopus oocytes. Accordingly, OCTN1 was functionally demonstrated to be a multispecific and pH-dependent organic cation transporter, which presumably functions as a proton/organic cation antiporter at the renal apical membrane and other tissues.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins , Animals , Biological Transport, Active , Carrier Proteins/biosynthesis , Cell Line , Gene Expression , Humans , Hydrogen-Ion Concentration , Membrane Potentials , Membrane Proteins/biosynthesis , Oocytes , Organic Cation Transport Proteins , Quinidine/pharmacology , RNA, Messenger/biosynthesis , Symporters , Tetraethylammonium Compounds/metabolism , Tetraethylammonium Compounds/pharmacology , Xenopus laevis
10.
Toxicology ; 132(1): 43-55, 1999 Jan 01.
Article in English | MEDLINE | ID: mdl-10199580

ABSTRACT

The agricultural fungicide N-(3,5-dichlorophenyl)succinimide (NDPS) is an acute nephrotoxicant in rats. Our previous studies have strongly suggested that glucuronide conjugation of NDPS metabolites might be a bioactivation step mediating NDPS nephrotoxicity. In this study, effects of substrates and/or inhibitors of primarily glucuronidation on NDPS nephrotoxicity were examined to explore further the role of glucuronidation in NDPS nephrotoxicity. Male Fischer 344 rats (4-6/group) were administered one of the following intraperitoneal (i.p.) pretreatments (dose, pretreatment time) prior to NDPS (0.4 mmol/kg) or NDPS vehicle (sesame oil, 2.5 ml/kg): (1) no pretreatment; (2) borneol (900 mg/kg, 30 min); (3) eugenol (500 mg/kg per day, 3 days); (4) clofibric acid (400 mg/kg, 15 min before (1/2 dose) and 3 h after (1/2 dose)), or (5) valproic acid, sodium salt (1.0 mmol/kg, 15 min). Following NDPS or NDPS vehicle administration, renal function was monitored at 24 and 48 h. Pretreatment with borneol or eugenol, substrates for ether glucuronidation and sulfation (mainly glucuronidation), afforded complete protection against NDPS nephrotoxicity. Substrates for acyl glucuronidation, clofibric acid or valproic acid, mildly reduced or had little effect on NDPS nephrotoxicity, respectively. These results suggest that ether glucuronide conjugates of NDPS metabolites, rather than acyl glucuronide conjugates, may be the primary ultimate nephrotoxicant species mediating NDPS nephrotoxicity.


Subject(s)
Fungicides, Industrial/toxicity , Glucuronates/metabolism , Kidney Diseases/chemically induced , Kidney/drug effects , Succinimides/toxicity , Animals , Biotransformation , Blood Urea Nitrogen , Camphanes/pharmacology , Clofibric Acid/pharmacology , Drinking/drug effects , Eating/drug effects , Eugenol/pharmacology , Fungicides, Industrial/chemistry , Fungicides, Industrial/metabolism , In Vitro Techniques , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Male , Organ Size/drug effects , Proteinuria/chemically induced , Rats , Rats, Inbred F344 , Succinimides/chemistry , Succinimides/metabolism , Tetraethylammonium Compounds/metabolism , Valproic Acid/pharmacology , p-Aminohippuric Acid/metabolism
11.
Toxicol Sci ; 47(2): 181-6, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10220855

ABSTRACT

A cDNA for the organic cation transporter (rOCT2) of the rat kidney was inserted into the retroviral plasmid pLXSN. This plasmid was used to stably transfect NIH3T3 cells. The transfected cell line exhibited an enhanced rate of tetraethylammonium (TEA) uptake and efflux compared to wild-type NIH3T3 cells. Uptake of TEA by the transfected cells was markedly reduced upon incubation at 4 degrees C. When the extracellular pH was lowered from 8.1 to 5.9, uptake was also reduced, suggesting inhibition of rOCT2 by extracellular protons. The apparent K(m) for TEA in the transfected cells was 141 microM. The classical organic cation transport inhibitors, cyanine 863 and cimetidine, produced noncompetitive inhibition with apparent Ki values of 0.81 and 198 microM, respectively. Daunomycin, vinblastine, and the deoxyadenosine analogs, 2'-deoxytubercidin and 2-chlorodeoxyadenosine, did not appear to be substrates for rOCT2. However, the anticancer drug, cisplatin, competitively inhibited TEA uptake by rOCT2 with an apparent Ki value of 925 microM, suggesting that rOCT2 may play a role in its renal secretion. In summary, transfected NIH3T3 cells provide a facile system by which this and other organic ion transporters can be studied.


Subject(s)
Carrier Proteins/genetics , DNA, Complementary/isolation & purification , Kidney/metabolism , Models, Genetic , Organic Cation Transport Proteins , Transfection , 3T3 Cells , Animals , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Daunorubicin/pharmacology , Deoxyadenosines/pharmacology , Kinetics , Mice , Organic Cation Transporter 2 , Rats , Reverse Transcriptase Polymerase Chain Reaction , Tetraethylammonium Compounds/metabolism , Vinblastine/pharmacology
12.
J Pharmacol Exp Ther ; 288(3): 1192-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10027858

ABSTRACT

Polyspecific organic cation transporters in epithelia play an important role in the elimination of many endogenous bioactive amines and therapeutically important drugs. Recently, the first human organic cation transporter (hOCT1) was cloned from liver. The purpose of the current study was to determine the effect of molecular size and hydrophobicity on the transport of organic cations by hOCT1. We studied the interaction of a series of n-tetraalkylammonium (n-TAA) compounds (alkyl chain length, N, ranging from 1 to 6 carbons) with hOCT1 in a transiently transfected human cell line, HeLa. [14C]tetraethylammonium (TEA) uptake was measured under different experimental conditions. Both cis-inhibition and trans-stimulation studies were carried out. With the exception of tetramethylammonium, all of the n-TAAs significantly inhibited [14C]TEA uptake. A reversed correlation of IC50 values (range, 3.0-260 microM) with alkyl chain lengths or partition coefficients (LogP) was observed. trans-Stimulation studies revealed that TEA, tetrapropylammonium, tetrabutylammonium, as well as tributylmethylammonium trans-stimulated TEA uptake mediated by hOCT1. In contrast, tetramethylammonium and tetrapentylammonium did not trans-stimulate [14C]TEA uptake, and tetrahexylammonium demonstrated an apparent "trans-inhibition" effect. These data indicate that with increasing alkyl chain lengths (N >/= 2), n-TAA compounds are more poorly translocated by hOCT1 although their potency of inhibition increases. Similar findings were obtained with nonaliphatic hydrocarbons. These data suggest that a balance between hydrophobic and hydrophilic properties is necessary for binding and subsequent translocation by hOCT1.


Subject(s)
Carrier Proteins/metabolism , Membrane Proteins/metabolism , Quaternary Ammonium Compounds/metabolism , Biological Transport , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , HeLa Cells , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Molecular Weight , Organic Cation Transporter 1 , Quaternary Ammonium Compounds/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Solubility , Tetraethylammonium Compounds/metabolism , Transfection
13.
Circ Res ; 81(6): 1053-64, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9400387

ABSTRACT

Enantiomers of local anesthetics are useful probes of ion channel structure that can reveal three-dimensional relations for drug binding in the channel pore and may have important clinical consequences. Bupivacaine block of open hKv1.5 channels is stereoselective, with the R(+)-enantiomer being 7-fold more potent than the S(-)-enantiomer (Kd = 4.1 mumol/L versus 27.3 mumol/L). Using whole-cell voltage clamp of hKv1.5 channels and site-directed mutants stably expressed in Ltk- cells, we have identified a set of amino acids that determine the stereoselectivity of bupivacaine block. Replacement of threonine 505 by hydrophobic amino acids (isoleucine, valine, or alanine) abolished stereoselective block, whereas a serine substitution preserved it [Kd = 60 mumol/L and 7.4 mumol/L for S(-)- and R(+)-bupivacaine, respectively]. A similar substitution at the internal tetraethylammonium binding site (T477S) reduced the affinity for both enantiomers similarly, thus preserving the stereoselectivity [Kd = 45.5 mumol/L and 7.8 mumol/L for S(-)- and R(+)-bupivacaine, respectively]. Replacement of L508 or V512 by a methionine (L508M and V512M) abolished stereoselective block, whereas substitution of V512 by an alanine (V512A) preserved it. Block of Kv2.1 channels, which carry valine, leucine, and isoleucine residues at T505, L508, and V512 equivalent sites, respectively, was not stereoselective [Kd = 8.3 mumol/L and 13 mumol/L for S(-)- and R(+)-bupivacaine, respectively]. These results suggest that (1) the bupivacaine binding site is located in the inner mouth of the pore, (2) stereoselective block displays subfamily selectivity, and (3) a polar interaction with T505 combined with hydrophobic interactions with L508 and V512 are required for stereoselective block.


Subject(s)
Anesthetics, Local/pharmacology , Bupivacaine/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/drug effects , Amino Acid Sequence , Binding Sites , Dose-Response Relationship, Drug , Humans , Kv1.5 Potassium Channel , Molecular Sequence Data , Mutagenesis, Site-Directed , Stereoisomerism , Structure-Activity Relationship , Tetraethylammonium Compounds/metabolism , Time Factors
14.
J Neuroimmunol ; 78(1-2): 8-18, 1997 Sep.
Article in English | MEDLINE | ID: mdl-9307224

ABSTRACT

Beta-endorphin and naloxone bind to Jurkat cell membrane preparations and can mutually displace each other from membrane binding sites. Tetraethylammonium ion, a potassium channel blocker, competitively displaces beta-endorphin and naloxone from membrane binding sites. Mitogen stimulated calcium ion flux is inhibited by tetraethyl ammonium and this inhibition is relieved by naloxone. With data derived from whole cell calcium ion flux studies, we accurately calculated the competitive displacement of beta-endorphin and naloxone from membrane preparations by tetraethylammonium thus showing that the action of these agents on potassium channels does not require second messengers. Using the resuspension induced ion flux technique, we find that beta-endorphin competes against naloxone for binding to Jurkat cells and naloxone competes against charybdotoxin, a potassium channel inhibitor, which like tetraethylammonium, is known to bind to the outer vestibule of the channel. Patch clamp electrophysiological studies show that beta-endorphin and naloxone exert complex actions on potassium channels in the presence or absence of mitogens. We conclude that one molecule of beta-endorphin or naloxone, but not both at the same time, bind to an area near the charybdotoxin/tetraethylammonium binding locus of Jurkat potassium channels.


Subject(s)
Naloxone/metabolism , Narcotic Antagonists/metabolism , Potassium Channel Blockers , T-Lymphocytes/metabolism , Tetraethylammonium Compounds/metabolism , beta-Endorphin/metabolism , Binding Sites , Binding, Competitive , Calcium Channel Blockers/pharmacology , Charybdotoxin/pharmacology , Electrophysiology , Mitogens/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Potassium Channels/drug effects , Potassium Channels/physiology , Proto-Oncogene Proteins c-myc/metabolism , Quaternary Ammonium Compounds/pharmacology , Tetraethylammonium , Tetraethylammonium Compounds/pharmacology , Tumor Cells, Cultured , beta-Endorphin/pharmacology
15.
Toxicology ; 122(1-2): 123-32, 1997 Sep 26.
Article in English | MEDLINE | ID: mdl-9274808

ABSTRACT

Paraquat (PQ) induces lung, liver and kidney damage. Since PQ mainly is eliminated by the kidney, the kidney damage is of particular importance to the outcome of PQ poisoning. The exact toxic mechanism of PQ is still unclear but it is assumed to involve redox cycling and formation of reactive oxygen species. In this study, further investigations on the toxic mechanism and metabolic effects of PQ were performed using isolated renal proximal tubules from rabbits. Proximal tubules were isolated using a combined iron perfusion and collagenase method. Suspended tubules were incubated for varying periods and concentrations of PQ at 25 or 37 degrees C in Krebs-Ringer phosphate buffer or HCO3-/CO2 buffer. The cytotoxic effect of PQ was evaluated by (1) markers of oxidative stress: status of glutathione (GSH/GSSG) and formation of malondialdehyde (MDA); and (2) markers of tubular metabolism: oxygen consumption (QO2), transport of 14C-p-aminohippuric acid (PAH) and 14C-tetraethylammonium (TEA). Using 0.5 and 5 mM PQ, the GSH/GSSG ratio decreased whereas formation of MDA increased indicating oxidative stress. PQ reduced the accumulation of PAH and TEA, the basal QO2 and the ouabain sensitive QO2 indicating inhibition of the Na/K-ATPase. Nystatin-stimulated QO2 was reduced by PQ, excluding inhibition of Na+ entry as a possible cytotoxic mechanism and suggesting mitochondrial injury. This was confirmed by measuring FCCP-uncoupled QO2. Thus high concentrations of PQ appear to disrupt mitochondrial electron chain transfer resulting in reduction of metabolic functions.


Subject(s)
Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Paraquat/toxicity , Aminohippuric Acids/metabolism , Animals , Biological Transport/drug effects , Female , In Vitro Techniques , Kidney Tubules, Proximal/metabolism , Male , Malondialdehyde/metabolism , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Rabbits , Tetraethylammonium , Tetraethylammonium Compounds/metabolism
17.
Biochim Biophys Acta ; 1327(2): 249-58, 1997 Jul 25.
Article in English | MEDLINE | ID: mdl-9271267

ABSTRACT

A large conductance, Ca2+-activated K+ channel in a human lung epithelial cell line (A549) was identified using the single channel patch clamp technique. Channel conductance was 242 +/- 33 pS (n = 67) in symmetrical KCl (140 mM). The channel was activated by membrane depolarization and increased cytosolic Ca2+. High selectivity was observed for K+ over Rb+(0.49) > Cs+(0.14) > Na+(0.09). Open probability was significantly decreased by Ba2+ (5 mM) and quinidine (5 mM) to either surface, but TEA (5 mM) was only effective when added to the external surface. All effects were reversible. Increasing cytosolic Ca2+ concentration from 10(-7) to 10(-6) M caused an increase in open probability from near zero to fully activated. ATP decreased open probability at approximately 2 mM, but the effect was variable. The channel was almost always observed together with a smaller conductance channel, although they could both be seen individually. We conclude that A549 cells contain large conductance Ca2+-activated K+ channels which could explain a major fraction of the K+ conductance in human alveolar epithelial membranes.


Subject(s)
Lung/chemistry , Potassium Channels, Calcium-Activated , Potassium Channels/analysis , Adenosine Triphosphate/metabolism , Cell Line , Epithelium/chemistry , Humans , Large-Conductance Calcium-Activated Potassium Channels , Patch-Clamp Techniques , Tetraethylammonium Compounds/metabolism
18.
J Biol Chem ; 272(26): 16548-54, 1997 Jun 27.
Article in English | MEDLINE | ID: mdl-9195965

ABSTRACT

Polyspecific organic cation transporters in the renal proximal tubule mediate the secretion of many clinically used drugs as well as endogenous metabolites. Recently, two organic cation transporters (rOCT1 and rOCT2) were cloned from rat kidney. In this study, we report the cloning and functional expression of an rOCT1 isoform, rOCT1A, from rat kidney. Genomic DNA cloning and sequencing demonstrated that rOCT1A is an alternatively spliced variant of rOCT1 with a deletion of 104 base pairs near the 5'-end. The uptake of [14C]tetraethylammonium (TEA) in oocytes injected with the cRNA-encoding rOCT1A was increased 16-fold over that in water-injected oocytes (29 +/- 2.8 pmol/oocyte/h versus 1.8 +/- 0.13 pmol/oocyte/h, mean +/- S.E., p < 0.05). [14C]TEA uptake in the cRNA-injected oocytes was saturable (Km = 42 +/- 11 microM) and was inhibited significantly by organic cations, including cimetidine and N1-methylnicotinamide. The amino acid sequence was deduced from the cDNA after examination of all three reading frames. Two overlapping open reading frames were found. Studies with synthetic constructs suggest that a functional organic cation transporter is encoded by the larger open reading frame. The larger open reading frame encodes a 430-amino acid protein (termed rOCT1A) that is 92% identical to rOCT1 and 57% identical to rOCT2. From hydropathy analysis, rOCT1A is predicted to have 10 transmembrane domains with both amino and carboxyl termini intracellular. RNase protection assays demonstrate the presence of rOCT1A mRNA transcripts in rat kidney cortex, medulla, and intestine. These studies demonstrate the presence of a functional, alternatively spliced organic cation transporter (rOCT1A) in rat kidney.


Subject(s)
Carrier Proteins/physiology , Kidney/metabolism , Membrane Proteins/physiology , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/genetics , Cloning, Molecular , Membrane Proteins/genetics , Molecular Sequence Data , Organic Cation Transporter 1 , Protein Biosynthesis , RNA, Messenger/analysis , Rats , Tetraethylammonium Compounds/metabolism , Xenopus laevis
19.
J Gen Physiol ; 109(5): 527-35, 1997 May.
Article in English | MEDLINE | ID: mdl-9154902

ABSTRACT

Small organic molecules, like quaternary ammonium compounds, have long been used to probe both the permeation and gating of voltage-dependent K+ channels. For most K+ channels, intracellularly applied quaternary ammonium (QA) compounds such as tetraethylammonium (TEA) and decyltriethylammonium (C10) behave primarily as open channel blockers: they can enter the channel only when it is open, and they must dissociate before the channel can close. In some cases, it is possible to force the channel to close with a QA blocker still bound, with the result that the blocker is "trapped." Armstrong (J. Gen. Physiol. 58:413-437) found that at very negative voltages, squid axon K+ channels exhibited a slow phase of recovery from QA blockade consistent with such trapping. In our studies on the cloned Shaker channel, we find that wild-type channels can trap neither TEA nor C10, but channels with a point mutation in S6 can trap either compound very efficiently. The trapping occurs with very little change in the energetics of channel gating, suggesting that in these channels the gate may function as a trap door or hinged lid that occludes access from the intracellular solution to the blocker site and to the narrow ion-selective pore.


Subject(s)
Ion Channel Gating/physiology , Potassium Channels/metabolism , Animals , Cell Line , Electrophysiology , Energy Metabolism , Humans , Ion Channel Gating/drug effects , Membrane Potentials/physiology , Mice , Potassium Channels/drug effects , Potassium Channels/genetics , Recombinant Proteins/metabolism , Tetraethylammonium Compounds/metabolism
20.
J Physiol ; 499 ( Pt 2): 403-16, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9080370

ABSTRACT

1. The delayed rectifier (DR) K+ channel pore was probed using different permeant and blocking ions applied intra- and extracellularly. Currents were recorded from bullfrog sympathetic neurons using whole-cell patch-clamp techniques. 2. With intra- and extracellular Cs+ (0 K+), there were large, tetraethylammonium (TEA)-sensitive currents. Adding K+ back to the extracellular solution revealed that the current with Cs+i was K+ selective (permeability ratio PCs/PK = 0.17 +/- 0.02, n = 4) and showed a strong anomalous mole fraction effect. 3. There were also large non-inactivating currents with Na+i and Na+o (0 K+). The current with Na+i was K+ selective (Na+o vs. K+o: PNa/PK = 0.022 +/- 0.005, n = 5), and was TEA sensitive with K+o but not with Na+o. 4. Permeant ions affected gating kinetics. DR currents activated faster in K+ than in Cs+, and activated faster with increasing concentrations of either K+ or Cs+. Deactivation was slowed by increased K+ or Cs+ concentration, with no difference between K+ and Cs+. 5. The pore was also characterized using intracellular blocking ions. A wide variety of monovalent cations (TEA, N-methyl-D-glucamine, arginine, choline, CH3NH3+, Li+, Cs+ and Na+) blocked DR channels from the inside in a voltage-dependent manner: KD at 0 mV was 2.9 mM for TEA and 134-487 mM for the others, at apparent electrical distances (delta) of 0.33-0.79. There was no detectable block by 10 mM Mgi2+. Apart from TEA, the organic cations did not block from the outside. 6. The permeability to Na+ in the absence of K+, and the strong anomalous mole fraction effects observed for Cs+o + K+o mixtures, suggest that DR channels select for K+ using ion-ion competition. The block by large intracellular cations shows that the pore is asymmetrical. The loss of high affinity TEAo block with Na+i and Na+o, and the effects of permeant ions on gating, suggest that channel conformation may be affected by ions in the pore.


Subject(s)
Ganglia, Sympathetic/cytology , Ion Channel Gating , Neurons/metabolism , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , Arginine/metabolism , Cesium/metabolism , Choline/metabolism , Delayed Rectifier Potassium Channels , Magnesium/metabolism , Meglumine/metabolism , Potassium/metabolism , Rana catesbeiana , Sodium/metabolism , Tetraethylammonium , Tetraethylammonium Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...