Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 14(4): e0214862, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30951542

RESUMEN

The effects of hyperuricemia on the expression of kidney drug transporters and on the pharmacokinetics of several substrate drugs were examined. We first established a rat model of hyperuricemia without marked symptoms of chronic kidney failure by 10-day co-administration of oxonic acid (uricase inhibitor) and adenine (biosynthetic precursor of uric acid). These hyperuricemic rats showed plasma uric acid concentrations of up to 6 mg/dL, which is similar to the serum uric acid level in hyperuricemic humans, with little change of inulin clearance. The mRNA levels of multidrug and toxin extrusion 1 (Mate1, Slc47a1), organic anion transporter 1 (Oat1, Slc22a6), organic cation transporter 2 (Oct2, Slc22a2), urate transporter 1 (Urat1, Slc22a12) and peptide transporter 1 (Pept1, Slc15a1) were significantly decreased in kidney of hyperuricemic rats. Since Oct2, Mate1 and Oat1 are important for renal drug elimination, we next investigated whether the pharmacokinetics of their substrates, metformin, cephalexin and creatinine, were altered. The plasma concentration of metformin was not affected, while its kidney tissue accumulation was significantly increased. The plasma concentration and kidney tissue accumulation of cephalexin and the plasma concentration of creatinine were also increased. Furthermore, the protein expression of kidney Mate1 was decreased in hyperuricemic rats. Accordingly, although multiple factors may influence renal handling of these drugs, these observations can be accounted for, at least in part, by downregulation of Mate1-mediated apical efflux from tubular cells and Oct2-mediated basolateral uptake. Our results suggest that hyperuricemia could alter the disposition of drugs that are substrates of Mate1 and/or Oct2.


Asunto(s)
Antiportadores/genética , Antiportadores/metabolismo , Hiperuricemia/genética , Hiperuricemia/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Transportador 2 de Cátion Orgánico/genética , Transportador 2 de Cátion Orgánico/metabolismo , Adenina/administración & dosificación , Animales , Cefalexina/sangre , Cefalexina/farmacocinética , Creatinina/sangre , Creatinina/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Hiperuricemia/sangre , Riñón/efectos de los fármacos , Masculino , Metformina/sangre , Metformina/farmacocinética , Ácido Oxónico/administración & dosificación , Farmacocinética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Ácido Úrico/administración & dosificación
2.
Eur J Pharm Biopharm ; 122: 49-53, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28974435

RESUMEN

The purpose of this study was to evaluate the intestinal metabolism and absorption in a mini-Ussing chamber equipped with animal intestinal tissues, based on the transport index (TI). TI value was defined as the sum of drug amounts transported to the basal-side component (Xcorr) and drug amounts accumulated in the tissue (Tcorr), which are normalized by AUC of a drug in the apical compartment, as an index for drug absorption. Midazolam was used as a test compound for the evaluation of intestinal metabolism and absorption. The metabolite formulation of midazolam was observed in both rats and dogs. Ketoconazole inhibited the intestinal metabolism of midazolam in rats and improved its intestinal absorption to a statistically significant extent. Therefore, the mini-Ussing chamber, equipped with animal intestinal tissues, showed potential to use the evaluation of the intestinal metabolism and absorption, including the assessment of species differences.


Asunto(s)
Absorción Intestinal/fisiología , Mucosa Intestinal/metabolismo , Cetoconazol/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Perros , Masculino , Permeabilidad/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
3.
Biopharm Drug Dispos ; 38(4): 301-312, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28092695

RESUMEN

Delamanid (OPC-67683, Deltyba™, nitro-dihydro-imidazooxazoles derivative) is approved for the treatment of adult pulmonary multidrug-resistant tuberculosis. The absorption, distribution and excretion of delamanid-derived radioactivity were investigated after a single oral administration of 14 C-delamanid at 3 mg/kg to rats. In both male and female rats, radioactivity in blood and all tissues reached peak levels by 8 or 24 h post-dose, and thereafter decreased slowly. Radioactivity levels were 3- to 5-fold higher in lung tissue at time to maximum concentration compared with plasma. In addition, radioactivity was broadly distributed in various tissues, including the central nervous system, eyeball, placenta and fetus, indicating that 14 C-delamanid permeated the brain, retinal and placental blood barriers. By 168 h post-dose, radioactivity in almost all the tissues was higher than that in the plasma. Radioactivity was also transferred into the milk of lactating rats. Approximately 6% and 92% of radioactivity was excreted in the urine and feces, respectively, indicating that the absorbed radioactivity was primarily excreted via the biliary route. No significant differences in the absorption, distribution and excretion of 14 C-delamanid were observed between male and female rats. The pharmacokinetic results suggested that delamanid was broadly distributed to the lungs and various tissues for a prolonged duration of time at concentrations expected to effectively target tuberculosis bacteria. These data indicate that delamanid, in addition to its previously demonstrated efficacy in pulmonary tuberculosis, might be an effective therapeutic approach to treating extrapulmonary tuberculosis. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Antituberculosos/farmacocinética , Antituberculosos/uso terapéutico , Nitroimidazoles/farmacocinética , Nitroimidazoles/uso terapéutico , Oxazoles/farmacocinética , Oxazoles/uso terapéutico , Tuberculosis/tratamiento farmacológico , Animales , Antituberculosos/orina , Bilis/química , Bilis/metabolismo , Heces/química , Femenino , Absorción Intestinal , Hígado/metabolismo , Masculino , Intercambio Materno-Fetal , Leche/química , Nitroimidazoles/orina , Oxazoles/orina , Placenta/metabolismo , Embarazo , Ratas , Ratas Sprague-Dawley , Distribución Tisular
4.
Eur J Pharm Sci ; 96: 373-380, 2017 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-27721045

RESUMEN

An adequate evaluation system for drug intestinal absorption is essential in the pharmaceutical industry. Previously, we established a novel prediction system of drug intestinal absorption in humans, using the mini-Ussing chamber equipped with human intestinal tissues. In this system, the TI value was defined as the sum of drug amounts transported to the basal-side component (Xcorr) and drug amounts accumulated in the tissue (Tcorr), which are normalized by AUC of a drug in the apical compartment, as an index for drug absorption. In order to apply this system to the screening assay, it is important to understand the differences between animal and human tissues in the intestinal absorption of drugs. In this study, the transport index (TI) values of three drugs, with different levels of membrane permeability, were determined to evaluate the rank order of drug absorbability in intestinal tissues from rats, dogs, and monkeys. The TI values in small intestinal tissues in rats and dogs showed a good correlation with those in humans. On the other hand, the correlation of TI values in monkeys was lower compared to rats and dogs. The rank order of the correlation coefficient between human and investigated animal tissues was as follows: dog (r2=0.978), rat (r2=0.955), and monkey (r2=0.620). TI values in large intestinal tissues from rats (r2=0.929) and dogs (r2=0.808) also showed a good correlation. The obtained TI values in small intestinal tissues in rats and dogs were well correlated with the fraction of drug absorbed (Fa) in humans. From these results, the mini-Ussing chamber, equipped with intestinal tissues in rats and dogs, would be useful as a screening tool in the drug discovery stage. In addition, the obtained TI values can be used for the prediction of the Fa in humans.


Asunto(s)
Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Animales , Atenolol/metabolismo , Atenolol/farmacología , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Cámaras de Difusión de Cultivos , Perros , Predicción , Haplorrinos , Humanos , Masculino , Metoprolol/metabolismo , Metoprolol/farmacología , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie
5.
Biol Pharm Bull ; 38(9): 1425-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26094899

RESUMEN

The direct inhibitory potential of twenty five anti-tuberculosis drugs on eight CYP-specific reactions in human liver microsomes was investigated to predict in vivo drug-drug interactions (DDIs) from in vitro data. Rifampicin, rifabutin, and thioacetazone inhibited one CYP reaction. Isoniazid and clofazimine had inhibitory effects on four CYP reactions, and rifapentine, ethionamide, and prothionamide widely inhibited CYP reactions. Based on the inhibition constant (Ki) and the therapeutic total inhibitor concentrations [I]max of eight drugs in human plasma, [I]max/Ki values were calculated to evaluate clinical DDIs. The [I]max/Ki values were 0.20 or less for rifampicin, rifabutin, and thioacetazone; 0.15-2.0 for isoniazid; 0.14-1.5 for rifapentine; 0.29-1.4 for ethionamide; 0.41-2.2 for prothionamide; and 0.12-6.3 for clofazimine. The highest [I]max/Ki values were 2.0 for isoniazid on CYP3A4 [testosterone (T)]; 1.5 for rifapentine on CYP3A4 [midazolam (M)]; 1.4 for ethionamide on CYP2C8; 2.2, 1.8, and 1.3 for prothionamide on CYP2B6, CYP2C19, and CYP2C8, respectively; and 6.3 and 5.7 for clofazimine on CYP3A4 (M) and CYP3A4 (T), respectively. These drugs with high [I]max/Ki values lead to clinical DDIs. Considering the drug regimens for tuberculosis (TB) and co-infection with TB and human immunodeficiency virus, the inhibitory potential for CYP3A4 and CYP2B6 is particularly important. These results suggest that clofazimine and prothionamide are likely to cause clinically relevant DDIs when co-administered with products metabolized by CYP3A4 and CYP2B6, respectively. Isoniazid and rifapentine may cause DDIs with drugs metabolized by CYP3A4.


Asunto(s)
Antituberculosos/farmacología , Inhibidores Enzimáticos del Citocromo P-450/farmacología , Microsomas Hepáticos/efectos de los fármacos , Sistema Enzimático del Citocromo P-450/metabolismo , Interacciones Farmacológicas , Humanos , Microsomas Hepáticos/metabolismo
6.
Drug Metab Dispos ; 43(8): 1277-83, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26055621

RESUMEN

The metabolism of delamanid (OPC-67683, Deltyba), a novel treatment of multidrug-resistant tuberculosis, was investigated in vitro using plasma and purified protein preparations from humans and animals. Delamanid was rapidly degraded by incubation in the plasma of all species tested at 37°C, with half-life values (hours) of 0.64 (human), 0.84 (dog), 0.87 (rabbit), 1.90 (mouse), and 3.54 (rat). A major metabolite, (R)-2-amino-4,5-dihydrooxazole derivative (M1), was formed in the plasma by cleavage of the 6-nitro-2,3-dihydroimidazo(2,1-b)oxazole moiety of delamanid. The rate of M1 formation increased with temperature (0-37°C) and pH (6.0-8.0). Delamanid was not converted to M1 in plasma filtrate, with a molecular mass cutoff of 30 kDa, suggesting that bioconversion is mediated by plasma proteins of higher molecular weight. When delamanid was incubated in plasma protein fractions separated by gel filtration chromatography, M1 was observed in the fraction consisting of albumin, γ-globulin, and α1-acid glycoprotein. In pure preparations of these proteins, only human serum albumin (HSA) metabolized delamanid to M1. The formation of M1 followed Michaelis-Menten kinetics in both human plasma and the HSA solution, with similar Km values: 67.8 µM in plasma and 51.5 µM in HSA. The maximum velocity and intrinsic clearance values for M1 were also comparable in plasma and HSA. These results strongly suggest that albumin is predominantly responsible for metabolizing delamanid to M1. We propose that delamanid degradation by albumin begins with a nucleophilic attack of amino acid residues on the electron-poor carbon at the 5 position of nitro-dihydro-imidazooxazole, followed by cleavage of the imidazooxazole moiety to form M1.


Asunto(s)
Antituberculosos/sangre , Nitroimidazoles/sangre , Oxazoles/sangre , Animales , Antituberculosos/farmacocinética , Biotransformación , Perros , Semivida , Humanos , Concentración de Iones de Hidrógeno , Ratones , Peso Molecular , Nitroimidazoles/farmacocinética , Oxazoles/farmacocinética , Conejos , Ratas , Albúmina Sérica/metabolismo , Temperatura
7.
Biol Pharm Bull ; 37(11): 1727-35, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25366478

RESUMEN

Delamanid is a new drug for the treatment of multidrug-resistant tuberculosis. Individuals who are co-infected with human immunodeficiency virus and Mycobacterium tuberculosis may require treatment with a number of medications that might interact significantly with the CYP enzyme system as inhibitors or inducers. It is therefore important to understand how drugs in development for the treatment of tuberculosis will affect CYP enzyme metabolism. The ability of delamanid to inhibit or induce CYP enzymes was investigated in vitro using human liver microsomes or human hepatocytes. Delamanid (100 µM) had little potential for mechanism-based inactivation on eight CYP isoforms (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4). Delamanid's metabolites were noted to inhibit the metabolism of some CYP isoforms, but these effects were observed only at metabolite concentrations that were well above those observed in human plasma during clinical trials. Delamanid (≤10 µM) did not induce CYP1A2, CYP2C9, and CYP3A4 activities in human hepatocytes, and there were no increases in CYP1A2, CYP2B6, CYP2C9, and CYP3A4 mRNA levels. Taken together, these data suggest that delamanid is unlikely to cause clinically relevant drug-drug interactions when co-administered with products that are metabolized by the CYP enzyme system.


Asunto(s)
Antituberculosos/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Nitroimidazoles/farmacología , Oxazoles/farmacología , Células Cultivadas , Sistema Enzimático del Citocromo P-450/genética , Interacciones Farmacológicas , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , ARN Mensajero/metabolismo
8.
Curr Drug Metab ; 14(2): 239-52, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22963393

RESUMEN

Cynomolgus monkeys are used to predict human pharmacokinetic and/or toxic profiles in the drug developmental stage. Cynomolgus P450s exhibit a high degree of identity (more than 90%) in both cDNA and amino acid sequences with corresponding human P450s. CYP3A protein predominantly exists in cynomolgus monkey liver microsomes, followed by CYP2A, CYP2C, CYP2B6, CYP2E1, and CYP2D. There are many similarities of metabolic properties in cytochrome P450s between cynomolgus monkeys and humans, but the species differences between cynomolgus monkey and human P450s are clearly present in substrate specificity and inhibitor selectivity. Diclofenac 4'-hydroxylation (DFOH) in monkey liver and intestinal microsomes shows much lower activities compared with those in human liver and intestinal microsomes. Sulfaphenazole strongly inhibits DFOH in human liver microsomes, but does not effectively inhibit DFOH in monkey liver and intestinal microsomes. Cynomolgus CYP2C19 exhibits higher activity for DFOH than cynomolgus CYP2C9 although this reaction is a marker reaction of human CYP2C9. On the other hand, cynomolgus CYP2C76 orthologue is not expressed in humans and shows 70-72% identity in amino acid sequences of human CYP2C subfamilies. Cynomolgus CYP2C76 metabolizes non-CYP2C substrates, 7-ethoxyresorufin (human CYP1A substrate) and bufuralol (human CYP2D6 substrate). In addition, cynomolgus CYP3A4 and CYP3A5 also exhibits wider substrate selectivity toward human CYP2D6 and CYP2E1 substrates. These enzymes may be responsible for species difference in drug metabolism between cynomolgus monkeys and humans. The comparative data presented here can be helpful for designing in vivo metabolic assays using cynomolgus monkeys in terms of substrate specificity and inhibitor selectivity.


Asunto(s)
Sistema Enzimático del Citocromo P-450/metabolismo , Animales , Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/química , Humanos , Cinética , Macaca fascicularis , Preparaciones Farmacéuticas/metabolismo , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
9.
Xenobiotica ; 42(8): 719-30, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22324396

RESUMEN

Cynomolgus monkeys are widely used to predict human pharmacokinetic and/or toxic profiles in the drug developmental stage. Characterization of cynomolgus monkey P450s such as the mRNA expression level, substrate specificity, and inhibitor selectivity were conducted to provide helpful information in designing monkey in vivo studies and monkey-to-human extrapolation. The expression levels of 12 monkey P450 mRNAs, which are considered to be important P450 subfamilies in drug metabolism, were investigated in the liver, small intestine (duodenum, jejunum, and ileum), and colon of individual monkeys. 3. In vitro activities and intrinsic clearance values were determined in monkey intestinal and liver microsomes (MIM and MLM, respectively) using nine typical oxidative reactions for human P450s. Paclitaxel 6α-hydroxylation, diclofenac 4'-hydroxylation, and S-mephenytoin 4'-hydroxylation showed low activities in MIM and MLM. IC50 values of eight selective inhibitors of human P450s were determined in MIM and MLM. Inhibitory effects of furafylline and sulfaphenazole were weak in monkeys on phenacetin O-deethylation and diclofenac 4'-hydroxylation, respectively. These results show profiles of monkey P450s in both the intestine and liver in detail and contribute to a better understanding of the species difference in substrate specificity and inhibitor selectivity between cynomolgus monkeys and humans.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450 , Sistema Enzimático del Citocromo P-450/metabolismo , Inhibidores Enzimáticos/farmacología , Intestinos/enzimología , Hígado/enzimología , Macaca fascicularis/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Sistema Enzimático del Citocromo P-450/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Inactivación Metabólica , Concentración 50 Inhibidora , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Masculino , Espectrometría de Masas , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/enzimología , Oxidación-Reducción/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estándares de Referencia , Especificidad por Sustrato/efectos de los fármacos , Teofilina/análogos & derivados , Teofilina/farmacología , Factores de Tiempo
10.
Biochem Pharmacol ; 79(2): 261-9, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19682433

RESUMEN

Phenobarbital has long been known to increase cellular levels of CYP1A1 and CYP1A2 possibly through a pathway(s) independent of aryl hydrocarbon receptor. We have investigated the role of constitutive androstane receptor (CAR), a xenobiotic-responsive nuclear receptor, in the transactivation of human CYP1A1 and CYP1A2. These genes are located in a head-to-head orientation, sharing a 5'-flanking region. Reporter assays were thus performed with dual-reporter constructs, containing the whole or partially deleted human CYP1A promoter between two different reporter genes. In this system, human CAR (hCAR) enhanced the transcription of both genes through common promoter regions from -461 to -554 and from -18089 to -21975 of CYP1A1. With reporter assays using additional deleted and mutated constructs, electrophoresis mobility shift assays and chromatin immunoprecipitation assays, an ER8 motif (everted repeat separated by eight nucleotides), located at around -520 of CYP1A1, was identified as an hCAR-responsive element and a binding motif of hCAR/human retinoid X receptor alpha heterodimer. hCAR enhanced the transcription of both genes also in the presence of an aryl hydrocarbon receptor ligand. Finally, hCAR activation increased CYP1A1 and CYP1A2 mRNA levels in cultured human hepatocytes. Our results indicate that CAR transactivates human CYP1A1 and CYP1A2 in human hepatocytes through the common cis-element ER8. Interestingly, the ER8 motif is highly conserved in the CYP1A1 proximal promoter sequences of various species, suggesting a fundamental role of CAR in the xenobiotic-induced expression of CYP1A1 and CYP1A2 independent of aryl hydrocarbon receptor.


Asunto(s)
Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Receptores Citoplasmáticos y Nucleares/fisiología , Secuencias Reguladoras de Ácidos Nucleicos , Transcripción Genética/fisiología , Animales , Secuencia de Bases , Inmunoprecipitación de Cromatina , Receptor de Androstano Constitutivo , Cartilla de ADN , Dimerización , Ensayo de Cambio de Movilidad Electroforética , Genes Reporteros , Hepatocitos/metabolismo , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...