Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters











Publication year range
1.
J Surg Res ; 99(2): 294-300, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11469900

ABSTRACT

BACKGROUND: The absorptive function of the intestinal graft is one of the most important factors for successful intestinal transplantation. To clarify whether the intestinal H(+)/peptide cotransporter (PEPT1) was expressed in the transplanted intestine, we examined the expression of PEPT1 in an experimental model of rat small intestinal transplantation in comparison with expression of Na(+)/glucose cotransporter (SGLT1). MATERIALS AND METHODS: Heterotopic intestinal transplantation was performed in allogeneic and syngeneic rat strain combinations. An additional group of allogeneic recipients was treated with tacrolimus (1 mg/kg) prior to transplantation, then daily for 7 days. Intestinal grafts were examined for histopathology and PEPT1 and SGLT1 expression. RESULTS: In the isografts, the levels of messenger RNA (mRNA) encoding both transporters were not changed, while the amount of SGLT1 protein was decreased and that of PEPT1 protein was increased. In the allografts, mRNA level and protein amount of both transporters and the amount of villin protein were decreased, and microscopic examination revealed histopathological features of rejection on day 7. Tacrolimus treatment ameliorated the histopathological features and prevented the decrease in villin protein expression. However, the decreases in PEPT1 and SGLT1 expression (both mRNA and protein) were partially prevented by tacrolimus treatment. CONCLUSION: This study indicated that the expression of transporters should be determined to evaluate intestinal graft function in addition to histopathological examination of the mucosa and that the levels of mRNA encoding intestinal nutrient transporters in biopsy specimens may be useful for evaluating the intestinal graft function for intestinal transplant patients.


Subject(s)
Carrier Proteins/genetics , Jejunum/transplantation , Symporters , Animals , Blotting, Western , Carrier Proteins/analysis , Gene Expression , Graft Rejection/drug therapy , Graft Rejection/physiopathology , Immunosuppressive Agents/pharmacology , Jejunum/chemistry , Jejunum/pathology , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/genetics , Monosaccharide Transport Proteins/analysis , Monosaccharide Transport Proteins/genetics , Peptide Transporter 1 , RNA, Messenger/analysis , Rats , Rats, Inbred Lew , Sodium-Glucose Transporter 1 , Sodium-Potassium-Exchanging ATPase/analysis , Tacrolimus/pharmacology
2.
Am J Physiol Renal Physiol ; 281(2): F273-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11457718

ABSTRACT

We investigated expression of the Na(+)-L-carnitine cotransport system and its role in transport of tetraethylammonium in a kidney epithelial cell line, LLC-PK(1). L-Carnitine uptake in the LLC-PK(1) cells was markedly stimulated in the presence of Na(+). The uptake was saturable, with Michaelis constant and maximal uptake velocity values of 7.8 microM and 153.7 pmol x mg protein(-1) x 15 min(-1), respectively. Cationic drugs such as tetraethylammonium, cimetidine, and quinidine inhibited L-carnitine uptake. The basolateral-to-apical transport of [(14)C]tetraethylammonium was enhanced markedly in the presence of an H(+) gradient on the apical side at a pH of 5.9. Under the conditions in which Na(+)/L-carnitine cotransport activity was saturable by the addition of 100 microM L-carnitine to the apical-side medium, the basolateral-to-apical transcellular transport of [(14)C]tetraethylammonium was unaffected. These results suggested that the Na(+)-L-carnitine cotransporter is expressed in the apical membranes of LLC-PK(1) cells, and is not responsible for efflux of tetraethylammonium from the cells. Transport of tetraethylammonium appeared to be mediated predominantly by an H(+)/organic cation antiporter in the apical membranes.


Subject(s)
Carnitine/metabolism , Carrier Proteins/metabolism , Kidney Tubules, Proximal/metabolism , Sodium/metabolism , Tetraethylammonium/metabolism , Animals , Carnitine/pharmacology , Cations/metabolism , Cell Line , Cell Polarity , Cephaloridine/pharmacology , Cephalosporins/pharmacology , Cimetidine/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hydrogen-Ion Concentration , Ion Transport/drug effects , Kidney Tubules, Proximal/cytology , Kinetics , Quinidine/pharmacology , Swine
3.
Kidney Int ; 58(3): 944-58, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10972658

ABSTRACT

The kidney plays an important role in the elimination of numerous hydrophilic xenobiotics, including drugs, toxins, and endogenous compounds. It has developed high-capacity transport systems to prevent urinary loss of filtered nutrients, as well as electrolytes, and simultaneously to facilitate tubular secretion of a wide range of organic ions. Transport systems for organic anions and cations are primarily involved in the secretion of drugs in renal tubules. The identification and characterization of organic anion and cation transporters have been progressing at the molecular level. To date, many members of the organic anion transporter (OAT), organic cation transporter (OCT), and organic anion-transporting polypeptide (oatp) gene families have been found to mediate the transport of diverse organic anions and cations. It has also been suggested that ATP-dependent primary active transporters such as MDR1/P-glycoprotein and the multidrug resistance-associated protein (MRP) gene family function as efflux pumps of renal tubular cells for more hydrophobic molecules and anionic conjugates. Tubular reabsorption of peptide-like drugs such as beta-lactam antibiotics across the brush-border membranes appears to be mediated by two distinct H+/peptide cotransporters: PEPT1 and PEPT2. Renal disposition of drugs is the consequence of interaction and/or transport via these diverse secretory and absorptive transporters in renal tubules. Studies of the functional characteristics, such as substrate specificity and transport mechanisms, and of the localization of cloned drug transporters could provide information regarding the cellular network involved in renal handling of drugs. Detailed information concerning molecular and cellular aspects of drug transporters expressed in the kidney has facilitated studies of the mechanisms underlying renal disposition as well as transporter-mediated drug interactions.


Subject(s)
Carrier Proteins/metabolism , Kidney Tubules/metabolism , Pharmacokinetics , Animals , Humans , Kidney Tubules/chemistry
4.
J Pharmacol Exp Ther ; 295(1): 261-5, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10991988

ABSTRACT

The renal tubular secretion of thiazides and loop diuretics via the organic anion transport system in renal tubules is required for them to reach their principal sites of action. Similarly, acetazolamide, a diuretic clinically administered for glaucoma, is excreted from the kidney by glomerular filtration and tubular secretion. In this study, we investigated the interaction and transport of these diuretics via the rat renal organic anion transporter rOAT1 by using Xenopus laevis oocyte expression system. p-[(14)C]Aminohippurate (PAH) uptake by rOAT1-expressing oocytes was inhibited in the presence of a thiazide (chlorothiazide, cyclothiazide, hydrochlorothiazide), a loop diuretic (bumetanide, ethacrynic acid, furosemide), or a carbonic anhydrase inhibitor (acetazolamide, ethoxzolamide, methazolamide). Dixon plot analysis demonstrated that the inhibition constant (K(i)) value was 1.1 mM for acetazolamide, 150 microM for hydrochlorothiazide, 9.5 microM for furosemide, and 5. 5 microM for bumetanide. Kinetic analysis revealed that acetazolamide inhibited rOAT1 competitively and that inhibition style of furosemide was a mixture of competitive and noncompetitive. [(14)C]PAH efflux was significantly enhanced when the rOAT1-expressing oocytes were incubated in the presence of unlabeled PAH, alpha-ketoglutarate, acetazolamide, chlorothiazide, or hydrochlorothiazide. rOAT1 stimulated acetazolamide uptake, which was inhibited by probenecid. Although the loop diuretics had little trans-stimulation effect on [(14)C]PAH efflux via rOAT1, the rOAT1-mediated furosemide uptake was observed. These findings suggest that rOAT1 contributes, at least in part, to the renal tubular secretion of acetazolamide, thiazides, and loop diuretics.


Subject(s)
Acetazolamide/pharmacokinetics , Carrier Proteins/physiology , Diuretics/pharmacokinetics , Kidney Tubules/metabolism , Animals , Anion Transport Proteins , Biological Transport , Bumetanide/pharmacokinetics , Furosemide/pharmacokinetics , Hydrochlorothiazide/pharmacokinetics , Organotechnetium Compounds/pharmacokinetics , Rats
5.
J Pharmacol Exp Ther ; 295(1): 360-6, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10992002

ABSTRACT

Grepafloxacin and levofloxacin transport by Caco-2 cell monolayers was examined to characterize the intestinal behavior of these quinolones. The levels of transcellular transport of [(14)C]grepafloxacin and [(14)C]levofloxacin from the basolateral to the apical side were greater than those in the opposite direction. The unidirectional transport was inhibited by the presence of excess unlabeled quinolones, accompanied by increased accumulation. The inhibitory effects of cyclosporin A plus grepafloxacin on basolateral-to-apical transcellular transport and cellular accumulation of [(14)C]grepafloxacin were comparable to those of cyclosporin A alone, indicating that the transport of grepafloxacin across the apical membrane was mainly mediated by P-glycoprotein. On the other hand, basolateral-to-apical transcellular transport of [(14)C]levofloxacin in the presence of cyclosporin A was decreased by unlabeled levofloxacin, grepafloxacin, and enoxacin, accompanied by significantly increased cellular accumulation. The organic cation cimetidine, organic anion p-aminohippurate, and the multidrug resistance-related protein (MRP) modulator probenecid did not affect the transcellular transport of [(14)C]grepafloxacin or [(14)C]levofloxacin in the presence of cyclosporin A. The basolateral-to-apical transcellular transport of levofloxacin in the presence of cyclosporin A showed concentration-dependent saturation with an apparent Michaelis constant of 5.6 mM. In conclusion, these results suggested that basolateral-to-apical flux of quinolones was mediated by P-glycoprotein and a specific transport system distinct from organic cation and anion transporters and MRP.


Subject(s)
Anti-Infective Agents/pharmacokinetics , Fluoroquinolones , Intestinal Mucosa/metabolism , Levofloxacin , Ofloxacin/pharmacokinetics , Piperazines/pharmacokinetics , Biological Transport , Caco-2 Cells , Cyclosporine/pharmacology , Dose-Response Relationship, Drug , Humans , Metabolic Clearance Rate
6.
Am J Physiol Gastrointest Liver Physiol ; 279(4): G799-805, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11005768

ABSTRACT

Transport characteristics of procainamide in the brush-border membrane isolated from rabbit small intestine were studied by a rapid-filtration technique. Procainamide uptake by brush-border membrane vesicles was stimulated by an outward H(+) gradient (pH(in) = 6.0, pH(out) = 7.5) against a concentration gradient (overshoot phenomenon), and this stimulation was reduced when the H(+) gradient was subjected to rapid dissipation by the presence of a protonophore, FCCP. An outward H(+) gradient-dependent procainamide uptake was not caused by H(+) diffusion potential. The initial uptake of procainamide was inhibited by other tertiary amines with N-dimethyl or N-diethyl moieties in their structures, such as triethylamine, dimethylaminoethyl chloride, and diphenhydramine, but not by tetraethylammonium and thiamine. Furthermore, procainamide uptake was stimulated by preloading the vesicles with these tertiary amines (trans-stimulation effect), indicating the existence of a specific transport system for tertiary amines. These findings indicate that procainamide transport in the intestinal brush-border membrane is mediated by the H(+)/tertiary amine antiport system that recognizes N-dimethyl or N-diethyl moieties in the structures of tertiary amines.


Subject(s)
Amines/metabolism , Intestinal Mucosa/physiology , Microvilli/physiology , Procainamide/pharmacokinetics , Amines/pharmacology , Animals , Biological Transport/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Cimetidine/pharmacology , Ethylamines/pharmacology , Guanidine/pharmacology , Hydrogen-Ion Concentration , Intestine, Small , Kinetics , Male , Mannitol/pharmacokinetics , Microvilli/drug effects , Potassium/metabolism , Rabbits , Sodium/metabolism
7.
J Pharm Pharmacol ; 52(12): 1467-72, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11197074

ABSTRACT

The effects of renal failure on the pharmacokinetics and bioavailability of tacrolimus were investigated in rats. Experimental renal dysfunction was induced by intraperitoneal injection of cisplatin (5 mg kg(-1)) into rats. The blood concentration of tacrolimus was measured after intravenous and intra-intestinal administration of the drug. The blood concentration of tacrolimus after intravenous administration (1 mg kg(-1)) was slightly increased (up to 1.3 fold) by induction of renal dysfunction. In contrast, the peak tacrolimus concentration after intra-intestinal administration (1 mg kg(-1) or 3 mg kg(-1)) in rats with renal failure was about 2-fold higher than that in normal controls. The bioavailability was increased by about 35% in rats with impaired renal function as compared with normal controls. These results suggested that the bioavailability of tacrolimus, which is mainly metabolized in the liver and intestine after oral administration, is also influenced by renal function.


Subject(s)
Immunosuppressive Agents/pharmacokinetics , Renal Insufficiency/metabolism , Tacrolimus/pharmacokinetics , Alanine Transaminase/blood , Alanine Transaminase/drug effects , Animals , Area Under Curve , Aspartate Aminotransferases/blood , Aspartate Aminotransferases/drug effects , Biological Availability , Blood Urea Nitrogen , Cisplatin/adverse effects , Creatinine/blood , Infusions, Intravenous , Intestinal Absorption , Male , Metabolic Clearance Rate , Rats , Rats, Wistar , Renal Insufficiency/chemically induced , Renal Insufficiency/physiopathology , Tacrolimus/blood
8.
J Pharmacol Exp Ther ; 291(2): 705-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10525090

ABSTRACT

Peptide transporters (PEPT1 and PEPT2) in epithelia play an important role in the absorption of small peptides and peptide-like drugs. Recently, it was demonstrated that various nonpeptidic compounds can be transported by these transporters. In the present study, we focused on the L-amino acid ester compounds and examined the mechanisms of their interaction with rat PEPTs (rPEPTs) using stable transfectants. Valacyclovir, the L-valyl ester prodrug of the antiherpetic agent acyclovir, competitively inhibited [(14)C]glycylsarcosine uptake in the rPEPT1- or rPEPT2-expressing cells. Dixon plot analyses showed that the inhibition constant (K(i)) values of valacyclovir were 2.7 and 0.22 mM for rPEPT1 and rPEPT2, respectively, suggesting that rPEPT2 had higher affinity for this agent. Various L-valine alkyl esters significantly inhibited [(14)C]glycylsarcosine uptake. L-Valine methyl ester (Val-OMe) competitively inhibited [(14)C]glycylsarcosine uptake with K(i) values of 3.6 and 0.83 mM for rPEPT1 and rPEPT2, respectively, indicating that Val-OMe is also a high-affinity substrate for rPEPT2. Val-OMe had a trans-stimulation effect on [(14)C]glycylsarcosine efflux from both transfectants, suggesting the translocation of L-valine methyl ester via rPEPTs. Val-OMe showed the most potent inhibitory effect among the several L-amino acid methyl esters examined. We conclude that Val-OMe, as well as valacyclovir, could be recognized and transported by rPEPT1 and rPEPT2 and that these L-valyl esters showed higher affinity for rPEPT2 as do most substrates of these transporters. Our results suggest that L-valine is a desirable L-amino acid for the esterification of poorly permeable drugs to enhance their oral bioavailability targeting intestinal PEPT1.


Subject(s)
Acyclovir/analogs & derivatives , Amino Acids/physiology , Carrier Proteins/physiology , Dipeptides/pharmacokinetics , Valine/analogs & derivatives , Valine/pharmacology , Acyclovir/pharmacology , Animals , Carrier Proteins/classification , Dose-Response Relationship, Drug , LLC-PK1 Cells , Prodrugs/pharmacology , Rats , Swine , Transfection , Valacyclovir
9.
J Pharm Pharmacol ; 51(7): 777-82, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10467951

ABSTRACT

A new derivative of recombinant human granulocyte colony-stimulating factor (rhG-CSF) has been synthesized by conjugating rhG-CSF to poly(styrene-co-maleic acid) (poly(styrene-co-maleic acid)-rhG-CSF) to try to avoid glomerular filtration and thus potentiate the neutrophil-proliferating activity of rhG-CSF. Poly(styrene-co-maleic acid)-rhG-CSF was highly bound to bovine serum albumin (BSA) and the molecular weight of the poly(styrene-co-maleic acid)-rhG-CSF-BSA complex was estimated to be about 90000 by gel filtration. Intravenous administration of poly(styrene-co-maleic acid)-rhG-CSF to normal rats resulted in a dose-dependent increase in neutrophil count. The neutrophil-proliferating activity of poly(styrene-co-maleic acid)-rhG-CSF was about 10 times greater than that of rhG-CSF. After intravenous injection at a dose of 5 microg protein kg(-1) the total clearance of rhG-CSF fell from 71.0 to 32.1 mLh(-1) kg(-1) following poly(styrene-co-maleic acid) modification. An isolated perfusion study in rat kidney showed that the filtered fraction of rhG-CSF was reduced by conjugation with poly(styrene-co-maleic acid). These results suggest that poly(styrene-co-maleic acid)-conjugation can potentiate the neutrophil-proliferating activity of rhG-CSF by reducing, at least in part, its renal clearance.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/pharmacokinetics , Maleates/administration & dosage , Polystyrenes/administration & dosage , Animals , Area Under Curve , Cross-Linking Reagents , Granulocyte Colony-Stimulating Factor/blood , Hematopoiesis/drug effects , Humans , Injections, Intravenous , Kidney/metabolism , Male , Maleates/blood , Metabolic Clearance Rate , Polystyrenes/blood , Rats , Rats, Wistar , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Tissue Distribution
10.
J Pharmacol Exp Ther ; 290(1): 388-92, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10381804

ABSTRACT

Transport characteristics of diphenhydramine, an antihistamine, were studied in cultured human intestinal Caco-2 cell monolayers to elucidate the mechanisms of its intestinal absorption. Diphenhydramine accumulation in the monolayers increased rapidly and was influenced by extracellular pH (pH 7.4 > 6.5 > 5.5). Diphenhydramine uptake was temperature dependent, saturable, and not potential sensitive. Kinetic analysis revealed that the apparent Km values were constant (0.8-1.0 mM) in all pH conditions tested, whereas Vmax values decreased at the lower pH. The initial uptake of diphenhydramine was competitively inhibited by another antihistamine, chlorpheniramine, with a Ki value of 1.3 mM. On the other hand, cimetidine and tetraethylammonium, typical substrates for the renal organic cation transport system, had no effect. Moreover, biological amines and neurotransmitters, such as histamine, dopamine, serotonin, and choline, also had no effect on the diphenhydramine accumulation. Finally, diphenhydramine uptake was stimulated by preloading monolayers with chlorpheniramine (trans-stimulation effect). These findings indicate that diphenhydramine transport in Caco-2 cells is mediated by a specific transport system. This pH-dependent transport system may contribute to the intestinal absorption of diphenhydramine.


Subject(s)
Diphenhydramine/metabolism , Histamine H1 Antagonists/metabolism , Intestinal Absorption/physiology , Biological Transport, Active , Caco-2 Cells , Cations , Chlorpheniramine/pharmacology , Chromatography, High Pressure Liquid , Humans , Hydrogen-Ion Concentration , Kinetics , Membrane Potentials/drug effects , Spectrophotometry, Ultraviolet , Stereoisomerism
11.
J Ocul Pharmacol Ther ; 15(2): 97-105, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10229487

ABSTRACT

Although acetazolamide, a carbonic anhydrase inhibitor, has an effect of lowering intraocular pressure, a number of side effects have been reported. Therefore, we investigated the relationship between the concentration of acetazolamide and its side effects, including plasma electrolyte imbalance. This study was conducted on 23 glaucomatous patients who received repeated doses of oral acetazolamide for one week or more. The concentrations of total and unbound plasma acetazolamide, as well as in the whole blood from the patients, were measured by high-performance liquid chromatography. The serum creatinine concentration, electrolyte concentrations, and adverse reactions were monitored. We found that plasma concentrations of chloride ion after repeated doses became higher than the normal range. This chloride ion concentration significantly correlated with the acetazolamide concentration in the erythrocytes, but not with the plasma concentration. The patients with erythrocyte acetazolamide concentration more than 20 microg/ml had higher incidences of the side effects. Periodical monitoring of erythrocyte acetazolamide concentration and plasma chloride ion can be easily and safely applied to elderly glaucomatous patients treated with acetazolamide for long periods to prevent overdosage and side effects.


Subject(s)
Acetazolamide/therapeutic use , Carbonic Anhydrase Inhibitors/therapeutic use , Glaucoma/drug therapy , Acetazolamide/adverse effects , Acetazolamide/blood , Adolescent , Adult , Aged , Aged, 80 and over , Electrolytes/blood , Female , Humans , Male , Middle Aged
12.
J Pharm Pharmacol ; 51(2): 227-32, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10217324

ABSTRACT

The nephrotoxicity of vancomycin and cisplatin and the protective effects of fosfomycin and imipenem-cilastatin on renal function have been studied in rats. The renal clearance of vancomycin after the induction of renal dysfunction was also evaluated by calculating the glomerular filtration rate (GFR) and its secretory clearance. Plasma concentrations of creatinine and urea nitrogen increased dose-dependently after vancomycin injection. No such increases were observed after co-treatment with fosfomycin or imipenem-cilastatin. Changes of N-acetyl-beta-D-glucosaminidase activity in the urine of vancomycin-treated rats were not remarkable compared with those in cisplatin-treated animals. The reduced renal clearance of vancomycin in rats with acute renal failure induced by vancomycin was because of a decrease in both GFR and secretory clearance. However, the changes in GFR and secretory clearance were not proportional-the change in GFR was more pronounced than that of secretory clearance in the experimental groups. In addition, the renal clearance of vancomycin was maintained at the control level after co-administration of fosfomycin or imipenem-cilastatin with vancomycin. These results suggest that vancomycin impairs glomerular filtration more markedly than renal tubular function as compared with cisplatin. Co-administration with fosfomycin or imipenem-cilastatin confers significant protection against the nephrotoxic effects of vancomycin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Kidney/drug effects , Acetylglucosaminidase/drug effects , Acetylglucosaminidase/urine , Animals , Antineoplastic Agents/pharmacology , Body Weight/drug effects , Cilastatin/pharmacology , Cilastatin, Imipenem Drug Combination , Cisplatin/pharmacology , Creatinine/blood , Dose-Response Relationship, Drug , Drug Combinations , Fosfomycin/pharmacology , Glomerular Filtration Rate/drug effects , Imipenem/pharmacology , Kidney/pathology , Kidney/physiopathology , Male , Nitrogen/urine , Rats , Rats, Wistar , Vancomycin/adverse effects , Vancomycin/blood , Vancomycin/pharmacology , Weight Gain/drug effects
13.
Mol Pharmacol ; 55(4): 743-52, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10101033

ABSTRACT

We have isolated a cDNA coding a new organic anion transporter, OAT-K2, expressed specifically in rat kidney. The OAT-K2 cDNA had an open reading frame encoding a 498-amino acid protein (calculated molecular mass of 55 kDa) that shows 91% identity with the rat kidney-specific organic anion transporter, OAT-K1. Reverse transcription-coupled polymerase chain reaction analyses revealed that the OAT-K2 mRNA was expressed predominantly in the proximal convoluted tubules, proximal straight tubules, and cortical collecting ducts. When expressed in Xenopus oocytes, OAT-K2 stimulated the uptake of hydrophobic organic anions, such as taurocholate, methotrexate, folate, and prostaglandin E2, although its homolog OAT-K1 transported methotrexate and folate, but not taurocholate and prostaglandin E2. In MDCK cells stably transfected with the OAT-K1 and OAT-K2 cDNAs, each transporter was localized functionally to the apical membranes and showed transport activity similar to that in the oocyte. Moreover, the efflux of preloaded taurocholate was also enhanced across the apical membrane in OAT-K2 transfectant. The taurocholate transport by OAT-K2-expressing cells showed saturability (Km = 10.3 microM). Several organic anions, bile acids, cardiac glycosides, and steroids had potent inhibitory effects on the OAT-K2-mediated taurocholate transport in the transfectant. These findings suggest that the OAT-K2 participates in epithelial transport of hydrophobic anionic compounds in the kidney.


Subject(s)
Carrier Proteins/genetics , Kidney/metabolism , Membrane Proteins/genetics , Multidrug Resistance-Associated Proteins , Organic Anion Transporters , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/metabolism , Cloning, Molecular , DNA, Complementary/analysis , DNA, Complementary/genetics , Male , Membrane Proteins/metabolism , Molecular Sequence Data , Oocytes/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Tissue Distribution , Transfection , Xenopus laevis
14.
J Pharmacol Exp Ther ; 287(2): 800-5, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9808712

ABSTRACT

We have isolated a kidney-specific organic cation transporter, rat OCT2, which is distinct from rat OCT1 (Okuda M, Saito H, Urakami Y, Takano M and Inui K (1996) Biochem Biophys Res Commun 224:500-507). In our study, the functional characteristics and membrane localization of OCT1 and OCT2 were investigated by uptake studies using MDCK cells transfected with rat OCT1 or OCT2 cDNA (MDCK-OCT1 or MDCK-OCT2) and immunological studies. Tetraethylammonium (TEA) uptake by both MDCK-OCT1 and MDCK-OCT2 cells was markedly elevated when TEA was added to the basolateral medium, but not to the apical medium. Efflux of TEA from MDCK-OCT1 and MDCK-OCT2 cells was not changed by extracellular pH from 5.4 to 8.4, whereas TEA uptake by both transfectants was decreased by acidification of extracellular medium. Apparent Km values for TEA uptake by MDCK-OCT1 and MDCK-OCT2 cells were 38 and 45 microM, respectively. Although various hydrophilic organic cations such as 1-methyl-4-phenylpyridinium, cimetidine, quinidine, nicotine, N1-methylnicotinamide and guanidine markedly inhibited TEA uptake by both MDCK-OCT1 and MDCK-OCT2 cells, there were no significant differences in the apparent inhibition constants (Ki) against these organic cations between both transfectants. Furthermore, immunological studies using a polyclonal antibody against OCT1 revealed that OCT1 was expressed in the basolateral membranes but not in the brush-border membranes of the rat kidney. These results suggested that both OCT1 and OCT2 are basolateral-type organic cation transporters with broad substrate specificities, mediating tubular secretion of cationic drugs.


Subject(s)
Carrier Proteins/metabolism , Kidney Tubules/metabolism , Membrane Proteins/metabolism , Organic Cation Transport Proteins , Tetraethylammonium/metabolism , Animals , Base Sequence , Cations , Cell Line , Cell Membrane/metabolism , DNA Primers , Dogs , Organic Cation Transporter 1 , Organic Cation Transporter 2 , Rats
15.
J Pharmacol Exp Ther ; 286(2): 676-80, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9694920

ABSTRACT

To clarify the mechanisms of the renal tubular secretion of nicotine, we studied transport of nicotine in the kidney epithelial cell line LLC-PK1. The transcellular transport of nicotine from the basolateral side to the apical side of the LLC-PK1 monolayers grown on membrane filters was much greater than that of tetraethylammonium. The basolateral-to-apical transport of nicotine was stimulated by lowering the pH of the apical side, accompanied by a decrease in the accumulation of nicotine. The accumulation of nicotine from the basolateral side was inhibited by unlabeled nicotine, cotinine, tetraethylammonium, cimetidine and quinidine. The uptake of nicotine across the apical membrane was inhibited by unlabeled nicotine and quinidine but not by tetraethylammonium or cimetidine. Pretreatment with p-chloromercuribenzene sulfonate caused a decrease in the transcellular transport of tetraethylammonium but not of nicotine. These results suggest that nicotine undergoes vectorial transport from basolateral side to the apical side of LLC-PK1 monolayers in a H+ gradient-dependent manner, corresponding to the secretion in the renal tubules. Nicotine transport in LLC-PK1 cells could be mediated by a transport system that is distinct from the transport system for tetraethylammonium.


Subject(s)
Nicotine/metabolism , Nicotinic Agonists/metabolism , Tetraethylammonium/metabolism , 4-Chloromercuribenzenesulfonate/pharmacology , Animals , Biological Transport, Active , Hydrogen-Ion Concentration , LLC-PK1 Cells , Sulfhydryl Reagents/pharmacology , Swine
16.
J Pharmacol Exp Ther ; 286(2): 1037-42, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9694966

ABSTRACT

Two H+/peptide cotransporters, PEPT1 and PEPT2, are expressed in the kidney, mediating the renal tubular reabsorption of oligopeptides and beta-lactam antibiotics. We examined the interactions of beta-lactam antibiotics with peptide transporters in rat renal brush-border membranes by evaluating the inhibitory potencies of the antibiotics against glycylsarcosine transport. Western blot analysis revealed that PEPT1 and PEPT2 were expressed in the renal brush-border membranes with the apparent molecular masses of 75 and 105 kDa, respectively. Using renal brush-border membrane vesicles, the uphill transport of glycylsarcosine was observed in the presence of an inward H+ gradient and an inside-negative membrane potential. Two transport systems with high affinity (Km of 50 microM) and low affinity (Km of 1.2 mM) appeared kinetically to mediate the glycylsarcosine uptake. The inhibition constants of the antibiotics for glycylsarcosine transport were more closely correlated with those in stable LLC-PK1 cells transfected with rat PEPT2 rather than PEPT1 cDNA. The beta-lactam antibiotics with an alpha-amino group showed trans-stimulation effects on the glycylsarcosine uptake, suggesting that these antibiotics and glycylsarcosine share a common peptide transporter. However, the antibiotics lacking an alpha-amino group failed to show the trans-stimulation effect. It is concluded that amino-beta-lactam antibiotics at therapeutic concentrations interact predominantly with PEPT2 localized in the brush-border membranes of rat kidney.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carrier Proteins/pharmacology , Kidney/metabolism , Symporters , Animals , Blotting, Western , Dipeptides/metabolism , In Vitro Techniques , Kidney/drug effects , Kinetics , Male , Membranes/drug effects , Membranes/metabolism , Microvilli/drug effects , Microvilli/metabolism , Peptide Transporter 1 , Rats , Rats, Wistar , beta-Lactams
17.
Pharm Res ; 15(5): 734-8, 1998 May.
Article in English | MEDLINE | ID: mdl-9619782

ABSTRACT

PURPOSE: The effects of fosfomycin and imipenem/cilastatin on the nephrotoxicity of vancomycin were studied in rats, and those on the renal handling of vancomycin were also investigated in perfused kidneys. METHODS: The protective effects of fosfomycin and imipenem/cilastatin on vancomycin nephrotoxicity were evaluated by increases in plasma concentration of creatinine and urea nitrogen in rats. The urinary excretion of vancomycin was measured and analyzed kinetically in the perfused rat kidney. RESULTS: The nephrotoxicity induced by vancomycin (500 mg/kg, i.v.) was inhibited almost completely by co-administration of fosfomycin or imipenem/cilastatin. In the perfused rat kidney, the excretion ratio of vancomycin was less than those of p-aminohippurate and cimetidine, and greater than that of arbekacin, suggesting the secretion and reabsorption of vancomycin in renal tubules. The tissue/perfusate ratios of unbound vancomycin were not significantly changed by co-treatment with fosfomycin or imipenem/cilastatin. Imipenem/cilastatin significantly decreased the excretion ratio of vancomycin. Fosfomycin also decreased vancomycin excretion ratio, although this effect was not significant. CONCLUSIONS: The renal handling of vancomycin was different from those of organic anions and cations and an aminoglycoside antibiotic. The protective effects of fosfomycin and imipenem/cilastatin against the nephrotoxicity of vancomycin might be partly due to the change in renal handling of vancomycin, probably in its tubular secretion/ reabsorption, in rats.


Subject(s)
Drug Therapy, Combination/pharmacology , Fosfomycin/pharmacology , Kidney/drug effects , Vancomycin/metabolism , Animals , Cilastatin/pharmacology , Cilastatin, Imipenem Drug Combination , Drug Combinations , Drug Interactions , Drug Therapy, Combination/pharmacokinetics , Imipenem/pharmacology , Kidney/metabolism , Male , Metabolic Clearance Rate , Rats , Rats, Wistar , Vancomycin/pharmacology
18.
J Pharmacol Exp Ther ; 282(2): 955-60, 1997 Aug.
Article in English | MEDLINE | ID: mdl-9262363

ABSTRACT

The purpose of this study was to characterize the transport mechanisms involved in the renal tubular secretion of quinolones. The contribution of P-glycoprotein to the transport of quinolones was elucidated using a kidney epithelial cell line, LLC-PK1, and its transfectant derivative cell line, LLC-GA5-COL150, which expresses human P-glycoprotein on the apical membrane. The transcellular transport of levofloxacin, a quinolone antibacterial drug, from the basolateral to apical side was increased in LLC-GA5-COL150 compared with that in LLC-PK1 monolayers. The apparent Michaelis constant and maximum velocity values for the saturable transcellular transport of levofloxacin from the basolateral to apical side in LLC-GA5-COL150 monolayers were 3.0 mM and 45 nmol/mg protein per 15 min, respectively. The increased basolateral-to-apical transport in LLC-GA5-COL150 monolayers was completely inhibited by cyclosporin A and quinidine to the level observed in LLC-PK1 monolayers. In addition, 3 mM levofloxacin inhibited the basolateral-to-apical transport of daunorubicin in LLC-GA5-COL150 monolayers. The basolateral-to-apical transport of another quinolone antibacterial drug, DU-6859a, in LLC-GA5-COL150 monolayers greatly exceeded than that in LLC-PK1 monolayers, and was inhibited by levofloxacin. These findings suggest that quinolone antibacterial drugs are transported by P-glycoprotein, and that P-glycoprotein may contribute at least in part to the renal tubular secretion of quinolones.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Anti-Infective Agents/pharmacokinetics , Fluoroquinolones , Kidney/metabolism , Levofloxacin , Ofloxacin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Biological Transport , Daunorubicin/antagonists & inhibitors , Daunorubicin/pharmacokinetics , Epithelial Cells , Epithelium/metabolism , Humans , Kidney/cytology , LLC-PK1 Cells , Quinolones/pharmacokinetics , Swine
19.
J Pharmacol Exp Ther ; 283(3): 1039-42, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9399974

ABSTRACT

We recently cloned and characterized the rat kidney-specific organic anion transporter, OAT-K1, which was suggested to mediate renal tubular transport of methotrexate. In this study, we investigated the interactions of nonsteroidal anti-inflammatory drugs (NSAIDs) with OAT-K1 by evaluating the effects of these drugs on renal distribution of methotrexate in vivo, and on methotrexate accumulation in the stably transfected LLC-PK1 cells expressing OAT-K1 (LLC-OAT-K1). NSAIDs such as indomethacin and ketoprofen had significant inhibitory effects on renal accumulation of methotrexate in rats after coadministration. Indomethacin and ketoprofen inhibited methotrexate accumulation by LLC-OAT-K1 cells in a competitive manner with the apparent inhibition constant values of 1. 0 mM and 1.9 mM, respectively. Other NSAIDs including ibuprofen, flufenamate and phenylbutazone also showed potent inhibitory effects on methotrexate accumulation. However, indomethacin was not transported via OAT-K1. These results indicate that NSAIDs have potent inhibitory effects against the OAT-K1-mediated methotrexate transport, which suggests that the OAT-K1 may be one of interaction sites for methotrexate and NSAIDs in the kidney.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carrier Proteins/drug effects , Kidney/metabolism , Animals , Anion Transport Proteins , Drug Interactions , Indomethacin/pharmacokinetics , Indomethacin/pharmacology , Ketoprofen/pharmacology , Kidney/drug effects , Male , Methotrexate/pharmacokinetics , Rats , Rats, Wistar , Tetraethylammonium Compounds/pharmacokinetics
20.
FEBS Lett ; 394(2): 196-200, 1996 Sep 30.
Article in English | MEDLINE | ID: mdl-8843163

ABSTRACT

The LLC-PK1 cells stably transfected with a rat PEPT1 cDNA transported ceftibuten (anion) and cephradine (zwitterion), both oral beta-lactam antibiotics, in a H+-gradient-dependent manner. Diethylpyrocarbonate, a histidine residue modifier, abolished ceftibuten uptake. This inhibition was prevented in the presence of glycylsarcosine or cephradine. When expressed in Xenopus oocytes, replacement of either histidine 57 or histidine 121 of the rat PEPT1 with glutamine by site-directed mutagenesis eliminated ceftibuten and [14C]glycylsarcosine transport activities. Immunostaining of oocyte sections indicated that insertion of the mutant transporters in the plasma membranes was not impaired. These findings suggest that both histidine 57 and histidine 121, which are conserved in the rat, rabbit and human PEPT1, are involved in substrate recognition of this molecule.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cephalosporins/metabolism , Histidine/metabolism , Symporters , Animals , Blotting, Western , Carrier Proteins/genetics , Ceftibuten , Cephradine/metabolism , Diethyl Pyrocarbonate/pharmacology , Dipeptides/metabolism , Histidine/genetics , Hydrogen-Ion Concentration , LLC-PK1 Cells , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Oocytes/metabolism , Peptide Transporter 1 , Plasmids , Rats , Recombinant Proteins/metabolism , Swine , Transfection , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL