Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Pharmacol Exp Ther ; 369(1): 78-87, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30674560

RESUMEN

Dopamine exerts various effects including movement coordination and reward. It is useful to understand the quantitative relationship between drug pharmacokinetics and target engagement such as the change in occupancy and dopamine level in brain for the proper treatment of dopamine-related diseases. This study was aimed at developing a pharmacokinetic-pharmacodynamic (PK-PD) model based on dopamine transporter (DAT) occupancies that could describe changes in extracellular dopamine levels in brain after administration of methylphenidate (a DAT inhibitor) to rat. First, uptake of fluorescent substrates was studied in DAT-expressing human embryonic kidney 293 cells and concentration dependently inhibited by methylphenidate. By analyzing the uptake of fluorescent substrates in the presence or absence of methylphenidate, a mathematical model could estimate the association and dissociation rate constants of methylphenidate for DAT. Next, we measured the concentrations of methylphenidate in plasma and cerebrospinal fluid (CSF) and extracellular dopamine levels in the nucleus accumbens after single intraperitoneal administration of methylphenidate. The concentrations of methylphenidate in plasma increased almost dose proportionally and the CSF-to-plasma concentration ratio was similar among evaluated dose. The extracellular dopamine levels also increased with dose. These data were analyzed using the mechanism-based PK-PD model, which incorporates dopamine biosynthesis, release from a synapse, reuptake via DAT into a synapse, and elimination from a synapse. Methylphenidate concentrations in plasma and dopamine profiles predicted by the PK-PD model were close to in vivo observations. In conclusion, our mechanism-based PK-PD model can accurately describe dopamine levels in the brain after administration of methylphenidate to rats.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Dopamina/metabolismo , Metilfenidato/farmacología , Metilfenidato/farmacocinética , Animales , Transporte Biológico/efectos de los fármacos , Masculino , Modelos Animales , Ratas
2.
Xenobiotica ; 49(9): 1044-1053, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30351180

RESUMEN

1. Naldemedine is a peripherally acting µ-opioid receptor antagonist for the treatment of opioid-induced constipation. 2. This phase 1 study investigated the absorption, distribution, metabolism and excretion of naldemedine, following a single oral 2-mg dose of [oxadiazole-14C]-naldemedine or [carbonyl-14C]-naldemedine to 12 healthy adult male subjects. Pharmacokinetic assessments were performed on blood, urine and fecal samples collected at defined intervals. 3. Naldemedine was the major circulating component in plasma with a median Tmax of approximately 0.8-0.9 h and a geometric mean t1/2,z of approximately 11 h. Total systemic exposures, AUC, of metabolites nor-naldemedine were less abundant than those of naldemedine (9% or 13% of AUC of naldemedine) and 16.2% or 18.1% of naldemedine was excreted as unchanged in urine after administration of [oxadiazole-14C]-naldemedine or [carbonyl-14C]-naldemedine, respectively, and benzamidine was the major radioactive component after administration of [oxadiazole-14C]-naldemedine (32.5% of administered dose). Overall, the recovery of total radioactivity was 92% (57.3% in urine; 34.8% in feces) after administration of [oxadiazole-14C]-naldemedine and 85% (20.4% in urine; 64.3% in feces) after administration of [carbonyl-14C]-naldemedine. 4. Our findings suggest that naldemedine is mainly metabolized to nor-naldemedine. Naldemedine was rapidly absorbed and well tolerated, with no major safety signals observed.


Asunto(s)
Naltrexona/análogos & derivados , Administración Oral , Adulto , Área Bajo la Curva , Radioisótopos de Carbono/farmacocinética , Estreñimiento/inducido químicamente , Voluntarios Sanos , Humanos , Inactivación Metabólica , Absorción Intestinal , Masculino , Naltrexona/administración & dosificación , Naltrexona/efectos adversos , Naltrexona/sangre , Naltrexona/farmacocinética , Náusea/inducido químicamente , Oxadiazoles/química , Oxadiazoles/farmacocinética , Distribución Tisular
3.
Drug Metab Dispos ; 46(11): 1497-1506, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30135242

RESUMEN

Cell models to investigate intestinal absorption functions, such as those of transporters and metabolic enzymes, are essential for oral drug discovery and development. The purpose of this study was to generate intestinal epithelial cells from human induced pluripotent stem cells (hiPSC-IECs) and then clarify whether the functions of hydrolase and transporters in them reflect oral drug absorption in the small intestine. The hiPSC-IECs showed the transport activities of P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and peptide transporter 1 (PEPT1), revealed by using their probe substrates ([3H]digoxin, sulfasalazine, and [14C]glycylsarcosine), and the metabolic activities of CYP3A4, CES2, and CES1, which were clarified using their probe substrates (midazolam, irinotecan, and temocapril). The intrinsic clearance by hydrolysis of six ester prodrugs into the active form in hiPSC-IECs was correlated with the plasma exposure (Cmax , AUC, and bioavailability) of the active form after oral administration of these prodrugs to rats. Also, the permeability coefficients of 14 drugs, containing two substrates of P-gp (doxorubicin and [3H]digoxin), one substrate of BCRP (sulfasalazine), and 11 nonsubstrates of transporters (ganciclovir, [14C]mannitol, famotidine, sulpiride, atenolol, furosemide, ranitidine, hydrochlorothiazide, acetaminophen, propranolol, and antipyrine) in hiPSC-IECs were correlated with their values of the fraction of intestinal absorption (Fa) in human clinical studies. These findings suggest that hiPSC-IECs would be a useful cell model to investigate the hydrolysis of ester prodrugs and to predict drug absorption in the small intestine.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Absorción Intestinal/fisiología , Intestino Delgado/metabolismo , Preparaciones Farmacéuticas/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Línea Celular Tumoral , Células Cultivadas , Humanos , Hidrólisis , Células Madre Pluripotentes Inducidas/metabolismo , Intestino Delgado/fisiología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Permeabilidad , Preparaciones Farmacéuticas/administración & dosificación , Profármacos/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Biochemistry ; 53(14): 2261-70, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24673529

RESUMEN

Organic anion transporting polypeptide (OATP) 1B1 is an important drug transporter expressed in human hepatocytes. Previous studies have indicated that transmembrane (TM) domain 2, 6, 8, 9, and in particular 10 might be part of the substrate binding site/translocation pathway. To explore which amino acids in TM10 are important for substrate transport, we mutated 34 amino acids individually to cysteines, expressed them in HEK293 cells, and determined their surface expression. Transport activity of the two model substrates estrone-3-sulfate and estradiol-17ß-glucuronide as well as of the drug substrate valsartan for selected mutants was measured. Except for F534C and F537C, all mutants were expressed at the plasma membrane of HEK293 cells. Mutants Q541C and A549C did not transport estradiol-17ß-glucuronide and showed negligible estrone-3-sulfate transport. However, A549C showed normal valsartan transport. Pretreatment with the anionic and cell impermeable sodium (2-sulfonatoethyl)methanethiosulfonate (MTSES) affected the transport of each substrate differently. Pretreatment of L545C abolished estrone-3-sulfate uptake almost completely, while it stimulated estradiol-17ß-glucuronide uptake. Further analyses revealed that mutant L545C in the absence of MTSES showed biphasic kinetics for estrone-3-sulfate that was converted to monophasic kinetics with a decreased apparent affinity, explaining the previously seen inhibition. In contrast, the apparent affinity for estradiol-17ß-glucuronide was not changed by MTSES treatment, but the Vmax value was increased about 4-fold, explaining the previously seen stimulation. Maleimide labeling of L545C was affected by preincubation with estrone-3-sulfate but not with estradiol-17ß-glucuronide. These results strongly suggest that L545C is part of the estrone-3-sulfate binding site/translocation pathway but is not directly involved in binding/translocation of estradiol-17ß-glucuronide.


Asunto(s)
Cisteína/genética , Mutagénesis , Transportadores de Anión Orgánico/genética , Células HEK293 , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado
5.
J Pharm Sci ; 109(4): 1605-1614, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31884012

RESUMEN

None of the physiologically based pharmacokinetic (PBPK) approaches using preclinical data show high predictability of human pharmacokinetic (PK) profiles for drugs affected by the intestinal first-pass effect. Here we report a novel PBPK approach that incorporated the findings of a permeation study using human induced pluripotent stem cell-derived intestinal epithelial cells (hiPSC-IECs) to predict human PK profiles after oral administration of drugs. In hiPSC-IECs, gene expression levels of cytochrome P450 3A4 (CYP3A4) and P-glycoprotein (P-gp) are enhanced by rifampicin and 1,25-dihydroxyvitamin D3. The permeability of 24 drugs (10 test drugs and 14 reference drugs), including CYP3A4 and P-gp substrates, correlated highly with gastrointestinal availability (Fa × Fg), and could be converted to the apparent absorption rate constant (ka, app) based on the correlation between Fa × Fg and in vivo ka of 27 drugs. The ka, app was input into the PBPK model which contained the optimized calculation processes of metabolism and tissue distribution. The absolute average fold error of PK parameters such as maximum plasma concentration and bioavailability for test drugs was less than 2, suggesting that human PK profiles could be predicted with high accuracy. Our robust PBPK approach enables quick decision-making in drug discovery based on human PK profiles.


Asunto(s)
Células Madre Pluripotentes Inducidas , Administración Oral , Simulación por Computador , Células Epiteliales , Humanos , Modelos Biológicos , Farmacocinética
6.
J Pharm Sci ; 109(7): 2345-2355, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32283068

RESUMEN

Proper prediction of human pharmacokinetic (PK) profiles can accelerate the compound selection in drug discovery. Recently, we reported a robust bottom-up physiologically-based pharmacokinetic (PBPK) approach (J Pharm Sci. 2019 Aug; 108(8):2718-2727), which uses the in vivo rat distribution volume at the steady state (Vss) to determine human tissue-to-plasma partition coefficients (Kptissue). Here, we report on a bottom-up PBPK approach that can simulate the PK profile with both high-throughput and high-predictive accuracy only using in vitro data. In this study, as an alternative parameter of in vivo rat Vss which was used for the correction of human Kptissue, Vss, in vitro was obtained from protein binding data in rats, and the values of Vss, in vitro for 31 reference compounds showed good correlation with the observed rat Vss (R2 = 0.859). Next, rat and human PK profiles of reference compounds were predicted by the bottom-up PBPK approach using Kptissue corrected by rat Vss, in vitro. As a result, the absolute average fold errors for pharmacokinetic parameters were almost less than 2, showing that these PK profiles could be accurately predicted using in vitro data. This method enables the screening of promising compounds with good PK profiles in humans at an early stage of drug discovery.


Asunto(s)
Modelos Biológicos , Plasma , Animales , Descubrimiento de Drogas , Humanos , Fenómenos Físicos , Unión Proteica , Ratas
7.
J Pharm Sci ; 108(8): 2718-2727, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30876861

RESUMEN

The physiologically based pharmacokinetics (PBPK) model is a major mechanistic approach for predicting human pharmacokinetics (PK) using drug-specific and physiological parameters but has been difficult to use for human PK prediction with acceptable accuracy. Here, we report a newly developed PBPK approach that incorporates the mechanism of albumin-mediated membrane penetration in the liver and interspecies correlation for unbound tissue fractions. To verify the utility of our PBPK approach, we used 12 drugs that are mainly eliminated by hepatic metabolism to compare the prediction accuracy with a conventional PBPK approach and to observe human PK parameters. We found the predictive accuracy for total clearance (CLtot), distribution volume at the steady state (Vss), elimination half-life (t1/2), and plasma concentration at the last measurable time point (Clast) of our PBPK approach to show better absolute average fold error and percentage within 2-fold error (1.6-1.8 and 67%-92%, respectively) compared with values obtained from the conventional PBPK approach (2.1-2.4 and 42%-67%, respectively). As our approach can use parameters obtained in early drug screening, it could help accelerate successful nomination of drug candidates by optimizing the pharmacokinetics of new chemical entities by directly using predicted human PK profiles.


Asunto(s)
Albúminas/metabolismo , Hígado/metabolismo , Modelos Biológicos , Farmacocinética , Membrana Celular/metabolismo , Humanos , Tasa de Depuración Metabólica , Preparaciones Farmacéuticas/sangre , Preparaciones Farmacéuticas/metabolismo
8.
Drug Metab Pharmacokinet ; 34(2): 126-133, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30770183

RESUMEN

Naldemedine tosylate, a peripherally acting µ-opioid receptor antagonist, is indicated for treatment of opioid induced constipation in both Japan and US. Naldemedine has limited ability to affect the central analgesic effect of opioid analgesics. In this study, we investigated the contribution of P-glycoprotein (P-gp) on the brain distribution of naldemedine. Naldemedine tosylate showed acceptable oral absorption in rats. Following a single oral administration of [14C]-naldemedine tosylate to rats and ferrets, little radioactivity was detected in the region protected by the blood-brain barrier (BBB). In the assessment using Caco-2 cells, it was determined that naldemedine is a substrate for P-gp. The contribution of P-gp to the brain distribution of naldemedine was assessed using multidrug resistance 1a/b (mdr1a/b) knockout mice. While the brain-to-plasma concentration ratio (brain Kp) of naldemedine in the mdr1a/b knockout mice was 4-fold of that in the wild-type mice, the brain Kp in the mdr1a/b knockout mice was quite low (brain Kp < 0.1). These results suggest that the low brain distribution of naldemedine was due to the limited ability to cross the BBB rather than efflux by P-gp and therefore brain distribution of naldemedine would not be affected by concomitant administration of P-gp inhibitors or functional disorder of P-gp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Encéfalo/metabolismo , Naltrexona/análogos & derivados , Receptores Opioides mu/antagonistas & inhibidores , Animales , Células CACO-2 , Humanos , Masculino , Ratones , Ratones Noqueados , Estructura Molecular , Naltrexona/química , Naltrexona/farmacología , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/metabolismo , Distribución Tisular
9.
J Pharm Sci ; 107(5): 1443-1453, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29331382

RESUMEN

Accurate prediction of human pharmacokinetics (PK) is important for the choice of promising compounds in humans. As the predictability of human PK by an empirical approach is low for drugs with species-specific PK, the utility of a physiologically based pharmacokinetic (PBPK) model was verified using 16 hepatically metabolized reference drugs. After the prediction method for total clearance (CLtot) and distribution volume at steady state (Vdss) in the conventional PBPK model had been optimized, plasma concentrations following a single oral administration of each reference drug to healthy volunteers were simulated, and the prediction accuracy for human PK was compared between empirical approaches and the optimized PBPK model. In the drugs with low species-specific CLtot, there was little difference in predictability for maximum concentration (Cmax), time to maximum plasma concentration (Tmax), and area under the curve (AUC) (absolute average fold error: 1.3-2.4). In contrast, the optimized PBPK model predicted Cmax and AUC of the drugs with high species-specific CLtot with lower absolute average fold error (Cmax and AUC: 2.8 and 3.2, respectively) than those of the empirical approach (Cmax and AUC: 2.6-4.9 and 3.9-10.7, respectively). Therefore, the optimized PBPK model is useful for human PK prediction of drugs with species-specific CLtot.


Asunto(s)
Hígado/metabolismo , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Administración Oral , Área Bajo la Curva , Simulación por Computador , Humanos , Tasa de Depuración Metabólica , Modelos Biológicos , Preparaciones Farmacéuticas/administración & dosificación , Preparaciones Farmacéuticas/sangre
10.
J Med Chem ; 61(12): 5122-5137, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29733614

RESUMEN

Accumulation of Aß peptides is a hallmark of Alzheimer's disease (AD) and is considered a causal factor in the pathogenesis of AD. ß-Secretase (BACE1) is a key enzyme responsible for producing Aß peptides, and thus agents that inhibit BACE1 should be beneficial for disease-modifying treatment of AD. Here we describe the discovery and optimization of novel oxazine-based BACE1 inhibitors by lowering amidine basicity with the incorporation of a double bond to improve brain penetration. Starting from a 1,3-dihydrooxazine lead 6 identified by a hit-to-lead SAR following HTS, we adopted a p Ka lowering strategy to reduce the P-gp efflux and the high hERG potential leading to the discovery of 15 that produced significant Aß reduction with long duration in pharmacodynamic models and exhibited wide safety margins in cardiovascular safety models. This compound improved the brain-to-plasma ratio relative to 6 by reducing P-gp recognition, which was demonstrated by a P-gp knockout mouse model.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Oxazinas/química , Fragmentos de Péptidos/metabolismo , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Secretasas de la Proteína Precursora del Amiloide/química , Animales , Ácido Aspártico Endopeptidasas/química , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Cristalografía por Rayos X , Perros , Diseño de Fármacos , Canal de Potasio ERG1/metabolismo , Cobayas , Humanos , Células de Riñón Canino Madin Darby , Ratones Endogámicos C57BL , Ratones Noqueados , Oxazinas/farmacología , Inhibidores de Proteasas/farmacocinética , Relación Estructura-Actividad
11.
J Med Chem ; 61(13): 5525-5546, 2018 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-29775538

RESUMEN

ß-Secretase (BACE1) has an essential role in the production of amyloid ß peptides that accumulate in patients with Alzheimer's disease (AD). Thus, inhibition of BACE1 is considered to be a disease-modifying approach for the treatment of AD. Our hit-to-lead efforts led to a cellular potent 1,3-dihydro-oxazine 6, which however inhibited hERG and showed high P-gp efflux. The close analogue of 5-fluoro-oxazine 8 reduced P-gp efflux; further introduction of electron withdrawing groups at the 6-position improved potency and also mitigated P-gp efflux and hERG inhibition. Changing to a pyrazine followed by optimization of substituents on both the oxazine and the pyrazine culminated in 24 with robust Aß reduction in vivo at low doses as well as reduced CYP2D6 inhibition. On the basis of the X-ray analysis and the QM calculation of given dihydro-oxazines, we reasoned that the substituents at the 6-position as well as the 5-fluorine on the oxazine would stabilize a bioactive conformation to increase potency.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Oxazinas/química , Oxazinas/farmacología , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Humanos , Simulación del Acoplamiento Molecular , Oxazinas/metabolismo , Oxazinas/farmacocinética , Conformación Proteica , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Distribución Tisular
12.
Biochim Biophys Acta ; 1609(2): 218-24, 2003 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-12543384

RESUMEN

We investigated the contribution of the Na(+)/L-carnitine cotransporter in the transport of tetraethylammonium (TEA) by rat renal brush-border membrane vesicles. The transient uphill transport of L-carnitine was observed in the presence of a Na(+) gradient. The uptake of L-carnitine was of high affinity (K(m)=21 microM) and pH dependent. Various compounds such as TEA, cephaloridine, and p-chloromercuribenzene sulfonate (PCMBS) had potent inhibitory effects for L-carnitine uptake. Therefore, we confirmed the Na(+)/L-carnitine cotransport activity in rat renal brush-border membranes. Levofloxacin and PCMBS showed different inhibitory effects for TEA and L-carnitine uptake. The presence of an outward H(+) gradient induced a marked stimulation of TEA uptake, whereas it induced no stimulation of L-carnitine uptake. Furthermore, unlabeled TEA preloaded in the vesicles markedly enhanced [14C]TEA uptake, but unlabeled L-carnitine did not stimulate [14C]TEA uptake. These results suggest that transport of TEA across brush-border membranes is independent of the Na(+)/L-carnitine cotransport activity, and organic cation secretion across brush-border membranes is predominantly mediated by the H(+)/organic cation antiporter.


Asunto(s)
Carnitina/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Tetraetilamonio/metabolismo , 4-Cloromercuribencenosulfonato/farmacología , Animales , Transporte Biológico Activo , Carnitina/antagonistas & inhibidores , Concentración de Iones de Hidrógeno , Levofloxacino , Masculino , Microvellosidades/metabolismo , Ofloxacino/farmacología , Ratas , Ratas Wistar , Reactivos de Sulfhidrilo/farmacología , Tetraetilamonio/antagonistas & inhibidores
13.
Drug Metab Pharmacokinet ; 23(4): 293-303, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18762717

RESUMEN

Pivalic acid and valproic acid decreases L-carnitine concentration in the body via urinary excretion of their acylcarnitines, pivaloylcarnitine (PC) and valproylcarnitine (VC). To obtain an information about the mechanism of the physiological response, we investigated the renal handling of these acylcarnitines by Na+/L-carnitine cotransporter, OCTN2 using the isolated perfused rat kidney, rat OCTN2 (rOCTN2) and human OCTN2 (hOCTN2) expressing cells. In the perfused rat kidney, PC and VC were strongly reabsorbed with an efficiency comparable to L-carnitine, and these reabsorption were inhibited by 1 mM L-carnitine, suggesting that the interaction of L-carnitine with PC and VC reabsorption would be responsible for renal handling of these acylcarnitines in rats. The rOCTN2-mediated uptake of PC was lower than that of L-carnitine, whereas rOCTN2-mediated uptake of VC was as high as that of L-carnitine, indicating that contribution of rOCTN2 in renal handling of PC and VC would be different. Furthermore, hOCTN2-mediated uptake of these acylcarnitines was markedly lower than that of L-carnitine. On the other hand, both PC and VC inhibited L-carnitine reabsorption in the perfused rat kidney and their concentration-dependent inhibition was also observed for rOCTN2 and hOCTN2. These results suggest that low renal reabsorption and interaction of hOCTN2 for these acylcarnitines might possibly affect the decrease of L-carnitine concentration in humans.


Asunto(s)
Carnitina/análogos & derivados , Riñón/metabolismo , Proteínas de Transporte de Catión Orgánico/fisiología , Ácidos Pentanoicos/metabolismo , Profármacos/metabolismo , Ácido Valproico/metabolismo , Animales , Células CHO , Carnitina/metabolismo , Cricetinae , Cricetulus , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos
14.
Pharm Res ; 23(8): 1729-35, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16826461

RESUMEN

PURPOSE: Prodrugs with pivalic acid and valproic acid decrease L-carnitine concentration in plasma and tissues by urinary excretion of acylcarnitine as pivaloylcarnitine (PC) and valproylcarnitine (VC), respectively. We investigated the role of the Na+/L-carnitine cotransporter in the porcine kidney epithelial cell line, LLC-PK1 for the decrease of L-carnitine concentration. METHODS: The uptake of L-[3H]carnitine, acetyl-L-[3H]carnitine (AC), L-[3H]PC and L-[3H]VC were investigated in LLC-PK1 cells seeded in a 6-well culture plate. RESULTS: L-Carnitine and AC uptake in LLC-PK1 cells exhibited Na+ dependency. The Km values for L-carnitine and AC uptake were 11.0 and 8.18 microM, respectively. These results indicated expression of Na+/ L-carnitine cotransporter in LLC-PK1 cells. PC and VC inhibited Na+/L-carnitine cotransporter in the competitive (Ki = 90.4 microM) and noncompetitive (Ki = 41.6 microM) manners, respectively. PC and VC uptake by Na+/L-carnitine cotransporter were not observed in LLC-PK1 cells. CONCLUSIONS: These data suggested that PC and VC formed in the body could not be reabsorbed in the kidney, resulting in the decrease of L-carnitine concentration. In addition, inhibition of L-carnitine reabsorption by VC with lower Ki value could induce the decrease of L-carnitine concentration. Collectively, the recognition and interaction of Na+/L-carnitine cotransporter are important factors for carnitine homeostasis.


Asunto(s)
Carnitina/metabolismo , Proteínas Portadoras/metabolismo , Inhibidores Enzimáticos/farmacología , Ácidos Pentanoicos/farmacología , Ácido Valproico/farmacología , Acetilcarnitina/metabolismo , Algoritmos , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Carnitina/análogos & derivados , Células LLC-PK1 , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA