Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Mol Genet Metab ; 132(4): 215-219, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33610470

RESUMEN

BACKGROUND: A subset of patients with phenylketonuria benefit from treatment with tetrahydrobiopterin (BH4), although there is no consensus on the definition of BH4 responsiveness. The aim of this study therefore was to gain insight into the definitions of long-term BH4 responsiveness being used around the world. METHODS: We performed a web-based survey targeting healthcare professionals involved in the treatment of PKU patients. Data were analysed according to geographical region (Europe, USA/Canada, other). RESULTS: We analysed 166 responses. Long-term BH4 responsiveness was commonly defined using natural protein tolerance (95.6%), improvement of metabolic control (73.5%) and increase in quality of life (48.2%). When a specific value for a reduction in phenylalanine concentrations was reported (n = 89), 30% and 20% were most frequently used as cut-off values (76% and 19% of respondents, respectively). When a specific relative increase in natural protein tolerance was used to define long-term BH4 responsiveness (n = 71), respondents most commonly reported cut-off values of 30% and 100% (28% of respondents in both cases). Respondents from USA/Canada (n = 50) generally used less strict cut-off values compared to Europe (n = 96). Furthermore, respondents working within the same center answered differently. CONCLUSION: The results of this study suggest a very heterogeneous situation on the topic of defining long-term BH4 responsiveness, not only at a worldwide level but also within centers. Developing a strong evidence- and consensus-based definition would improve the quality of BH4 treatment.


Asunto(s)
Biopterinas/análogos & derivados , Fenilalanina/genética , Fenilcetonurias/tratamiento farmacológico , Biopterinas/efectos adversos , Biopterinas/uso terapéutico , Canadá/epidemiología , Europa (Continente)/epidemiología , Humanos , Fenilalanina/sangre , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/sangre , Fenilcetonurias/epidemiología , Fenilcetonurias/patología , Estados Unidos/epidemiología
2.
Clin Pharmacol Ther ; 101(4): 519-530, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27943276

RESUMEN

A microdose cocktail containing midazolam, dabigatran etexilate, pitavastatin, rosuvastatin, and atorvastatin has been established to allow simultaneous assessment of a perpetrator impact on the most common drug metabolizing enzyme, cytochrome P450 (CYP)3A, and the major transporters organic anion-transporting polypeptides (OATP)1B, breast cancer resistance protein (BCRP), and MDR1 P-glycoprotein (P-gp). The clinical utility of these microdose cocktail probe substrates was qualified by conducting clinical drug interaction studies with three inhibitors with different in vitro inhibitory profiles (rifampin, itraconazole, and clarithromycin). Generally, the pharmacokinetic profiles of the probe substrates, in the absence and presence of the inhibitors, were comparable to their reported corresponding pharmacological doses, and/or in agreement with theoretical expectations. The exception was dabigatran, which resulted in an approximately twofold higher magnitude for microdose compared to conventional dosing, and, thus, can be used to flag a worst-case scenario for P-gp. Broader application of the microdose cocktail will facilitate a more comprehensive understanding of the roles of drug transporters in drug disposition and drug interactions.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocromo P-450 CYP3A/metabolismo , Combinación de Medicamentos , Interacciones Farmacológicas , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Adulto , Área Bajo la Curva , Proteínas Portadoras/antagonistas & inhibidores , Línea Celular , Inhibidores del Citocromo P-450 CYP3A/efectos adversos , Inhibidores del Citocromo P-450 CYP3A/uso terapéutico , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/enzimología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/metabolismo , Femenino , Voluntarios Sanos , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Farmacocinética , Distribución Tisular , Adulto Joven
3.
Clin Pharmacol Ther ; 100(5): 437-440, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27509262

RESUMEN

Creatinine is eliminated by the kidneys through a combination of glomerular filtration and active transport. Drug-induced increases in serum creatinine (SCr) and/or reduced creatinine renal clearance are used as a marker for acute kidney injury. However, inhibition of active transport of creatinine can result in reversible and, therefore, benign increases in SCr levels. Herein, the transporters involved in creatinine clearance are discussed, in addition to limitations of using creatinine as a biomarker for kidney damage.


Asunto(s)
Lesión Renal Aguda/sangre , Creatinina/sangre , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/sangre , Proteínas de Transporte de Catión Orgánico/metabolismo , Lesión Renal Aguda/diagnóstico , Animales , Biomarcadores/sangre , Humanos , Modelos Biológicos , Proteínas de Transporte de Catión Orgánico/efectos de los fármacos
4.
Clin Pharmacol Ther ; 100(4): 362-70, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27301780

RESUMEN

Protein expression of major hepatic uptake and efflux drug transporters in human pediatric (n = 69) and adult (n = 41) livers was quantified by liquid chromatography / tandem mass spectroscopy (LC-MS/MS). Transporter protein expression of OCT1, OATP1B3, P-gp, and MRP3 was age-dependent. Particularly, significant differences were observed in transporter expression (P < 0.05) between the following age groups: neonates vs. adults (OCT1, OATP1B3, P-gp), neonates or infants vs. adolescents and/or adults (OCT1, OATP1B3, and P-gp), infants vs. children (OATP1B3 and P-gp), and adolescents vs. adults (MRP3). OCT1 showed the largest increase, of almost 5-fold, in protein expression with age. Ontogenic expression of OATP1B1 was confounded by genotype and was revealed only in livers harboring SLCO1B1*1A/*1A. In livers >1 year, tissues harboring SLCO1B1*14/*1A showed 2.5-fold higher (P < 0.05) protein expression than SLCO1B1*15/*1A. Integration of these ontogeny data in physiologically based pharmacokinetic (PBPK) models will be a crucial step in predicting hepatic drug disposition in children.


Asunto(s)
Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Envejecimiento/metabolismo , Transportador 1 de Anión Orgánico Específico del Hígado/biosíntesis , Hígado/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Transportadores de Anión Orgánico Sodio-Independiente/biosíntesis , Transportador 1 de Catión Orgánico/biosíntesis , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Adolescente , Niño , Preescolar , Genotipo , Humanos , Lactante , Recién Nacido , Transportador 1 de Anión Orgánico Específico del Hígado/genética , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple/genética , Proteómica , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos
5.
Clin Pharmacol Ther ; 96(3): 291-5, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25141952

RESUMEN

Understanding genetic variations that influence pharmacokinetics (PK) in humans is important for optimal clinical use of drugs. Guidances for making decisions on when to conduct pharmacogenetic research during drug development have been proposed by regulatory agencies, but their uniform adoption presents problems due to an inherent lack of flexibility. A questions-based approach (QBA) was developed to enable drug development teams at Merck to iteratively and flexibly evaluate the potential impact of pharmacogenetics (PGx) on clinical pharmacokinetic variability.


Asunto(s)
Transporte Biológico/genética , Biotransformación/genética , Descubrimiento de Drogas/métodos , Variación Genética , Farmacogenética , Farmacocinética , Algoritmos , Animales , Genotipo , Humanos , Fenotipo , Medición de Riesgo
7.
Clin Pharmacol Ther ; 94(1): 95-112, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23588315

RESUMEN

This white paper addresses current approaches and knowledge gaps concerning methods to assess the role of transport proteins in drug/metabolite disposition in humans. The discussion focuses on in vitro tools to address key questions in drug development, including vesicle- and cell-based systems. How these methods can be used to assess the liability of compounds for transporter-based drug-drug interactions (DDIs) in vivo is also explored. Existing challenges and approaches to examine the involvement of transporters in drug disposition are discussed.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Descubrimiento de Drogas/métodos , Interacciones Farmacológicas , Proteínas de Transporte de Membrana/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Humanos
8.
Clin Pharmacol Ther ; 89(2): 234-42, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21191377

RESUMEN

We studied the time course for the reversal of rifampin's effect on the pharmacokinetics of oral midazolam (a cytochrome P450 (CYP) 3A4 substrate) and digoxin (a P-glycoprotein (P-gp) substrate). Rifampin increased midazolam metabolism, greatly reducing the area under the concentration-time curve (AUC(0-∞)). The midazolam AUC(0-∞) returned to baseline with a half-life of ~8 days. Rifampin's effect on the AUC(0-3 h) of digoxin was biphasic: the AUC(0-3 h) increased with concomitant dosing of the two drugs but decreased when digoxin was administered after rifampin. Digoxin was found to be a weak substrate of organic anion-transporting polypeptide (OATP) 1B3 in transfected cells. Although the drug was transported into isolated hepatocytes, it is not likely that this transport was through OATP1B3 because the transport was not inhibited by rifampin. However, rifampin did inhibit the P-gp-mediated transport of digoxin with a half-maximal inhibitory concentration (IC(50)) below anticipated gut lumen concentrations, suggesting that rifampin inhibits digoxin efflux from the enterocyte to the intestinal lumen. Pharmacokinetic modeling suggested that the effects on digoxin are consistent with a combination of inhibitory and inductive effects on gut P-gp. These results suggest modifications to drug-drug interaction (DDI) trial designs.


Asunto(s)
Digoxina/farmacocinética , Midazolam/farmacocinética , Proyectos de Investigación , Rifampin/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Adulto , Área Bajo la Curva , Transporte Biológico , Interacciones Farmacológicas , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Transportadores de Anión Orgánico Sodio-Independiente/fisiología , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos
9.
Xenobiotica ; 40(12): 826-39, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20925584

RESUMEN

Compound A ((1aR,5S,8S,10R,22aR)-5-tert-butyl-N-{(1R,2S)-1-[(cyclopropylsulfonyl)carbamoyl]-2-ethenylcyclopropyl}-14-methoxy-3,6-dioxo-1,1a,3,4,5,6,9,10,18,19,20,21,22,22a-tetradecahydro-8H-7,10-methanocyclopropa[18,19][1,10,3,6]dioxadiazacyclononadecino[12,11-b]quinoline-8-carboxamide) is a prototype of a series of subnanomolar inhibitors of genotypes 1, 2, and 3 hepatitis C virus (HCV) NS3/4A proteases. HCV NS3/4A protease inhibitors have demonstrated high antiviral effects in patients with chronic HCV infection and are likely to form a key component of future HCV therapy. Compound A showed excellent liver exposure in rats, which is essential for compounds intended to treat HCV. The compound was mainly eliminated intact in bile and showed greater than dose proportional systemic exposure in rats. Compound A demonstrated time- and temperature-dependent uptake into rat and human hepatocytes and proved to be a substrate for rat hepatic uptake transporter Oatp1b2 and for human hepatic uptake transporters OATP1B1 and OATP1B3. The liver selectivity observed for this compound is likely to be due to transporter-mediated hepatic uptake together with moderate passive permeability. Metabolism was mainly CYP3A-mediated and generated a reactive epoxide on the vinylcyclopropyl sulfonamide moiety that could be quenched by glutathione. Similar metabolic profiles of Compound A were obtained in liver microsomes of rats and humans. The oral bioavailability at 5 mg/kg was low due to extensive hepatic first-pass effect but clearly the intestinal absorption was enough to deliver a high amount of the compound to the liver. The metabolism and disposition properties of Compound A are particularly attractive to support its evaluation as a drug candidate for the treatment of hepatitis C.


Asunto(s)
Antivirales/metabolismo , Antivirales/farmacocinética , Hepacivirus/enzimología , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacocinética , Proteínas no Estructurales Virales/antagonistas & inhibidores , Animales , Antivirales/administración & dosificación , Antivirales/sangre , Transporte Biológico/efectos de los fármacos , Perros , Hepacivirus/efectos de los fármacos , Humanos , Inyecciones Intravenosas , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Microsomas Hepáticos/efectos de los fármacos , Inhibidores de Proteasas/administración & dosificación , Inhibidores de Proteasas/sangre , Ratas , Ratas Sprague-Dawley , Espectrometría de Masas en Tándem , Factores de Tiempo , Proteínas no Estructurales Virales/metabolismo
10.
Xenobiotica ; 38(2): 223-37, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18197560

RESUMEN

N-(1-(3,5-dichlorobenzenesulfonyl)-2S-methyl-azetidine-2-carbonyl)-L-4-(2',6'-dimethoxyphenyl)phenylalanine (1) is a potent antagonist of the very late activating (VLA) antigen-4. During initial screening, 1 exhibited an apparent plasma clearance (CL) of 227 ml min(-1) kg(-1) in Sprague-Dawley rats following an intravenous bolus dose formulated in an aqueous solution containing 40% polyethylene glycol. Such a high CL value led to speculation that the elimination of compound 1 involved extra-hepatic tissues. However, the apparent plasma CL was reduced to 97 ml in(-1) kg(-1) when a 2-min time point was added to sample collections, and further decreased to 48 ml min(-1) kg(-1) after the dose was formulated in rat plasma. The lung extraction of 1 in rats was negligible whereas the hepatic extraction was > or =90%, based on comparison of area under the curve (AUC) values derived from intra-artery, intravenous, and portal vein administration. In rats dosed intravenously with [(14)C]-1, approximately 91% of the radioactivity was recovered in bile over 48 h, with 85% accounted for in the first 4-h samples. The biliary radioactivity profile consisted of approximately 30% intact parent compound, 20% 1-glucuronide, and 50% oxidation products resulting from O-demethylation or hydroxylation reactions. When incubated with rat liver microsomes, oxidative metabolism of 1 was inhibited completely by 1-aminobenzotriazole (ABT), whereas the oxidation and glucuronidation reactions were little affected in the presence of cyclosporin A (CsA). In contrast, the hepatic extraction of 1 in rats was unperturbed in animals pre-dosed with ABT, but was reduced approximately 60% following treatment with CsA. In vitro, 1 was a substrate of the rat organic anion transporter Oatp1b2, and its cellular uptake was inhibited by CsA. In addition, the hepatic extraction of 1 was approximately 30% lower in Eisai hyperbilirubinaemic rats which lack functional multidrug resistant protein-2 (MRP2). Collectively, these data suggest that the clearance of 1 in rats likely is a result of the combined processes of hepatic oxidation, glucuronidation and biliary excretion, all of which are facilitated by active hepatic uptake of parent compound and subsequent active efflux of both unchanged parent and its metabolites into bile. It was concluded, therefore, that multiple mechanisms contribute to the clearance of 1 in rats, and suggest that appropriate pharmacokinetic properties might be difficult to achieve for this class of compounds. This case study demonstrates that an integrated strategy, incorporating both rapid screening and mechanistic investigations, is of particular value in supporting drug discovery efforts and decision-making processes.


Asunto(s)
Integrina alfa4beta1/antagonistas & inhibidores , Fenilalanina/análogos & derivados , Animales , Células Cultivadas , Ciclosporina/metabolismo , Perros , Inactivación Metabólica , Microsomas Hepáticos/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Fenilalanina/metabolismo , Fenilalanina/fisiología , Ratas , Ratas Sprague-Dawley , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos , Factores de Tiempo , Triazoles/farmacología
11.
Bioresour Technol ; 99(6): 1503-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17570662

RESUMEN

The adsorption kinetics of methylene blue on pyrolyzed petrified sediment (PPS) has been performed using a batch-adsorption technique. The effects of various experimental parameters, such as initial dye concentration, contact time, and temperature were investigated. The pseudo-first-order and pseudo-second-order kinetic models were used to describe the kinetic data. The best correlation coefficient was obtained using the pseudo first-order kinetic model, which shows that the adsorption of methylene blue followed the pseudo-first-order rate expression and the rate constants were evaluated. The Langmuir and Freundlich adsorption isotherm models were applied to describe the equilibrium isotherms and the isotherm constants were determined. It was found that the data fitted well to Langmuir and Freundlich models. The activation energy of adsorption was also evaluated for the adsorption of methylene blue onto pyrolyzed sediment. It was found about 8.5 kJ mol(-1). Thermodynamics parameters DeltaG(o), DeltaH(o), DeltaS(o) were calculated, indicating that this process can be spontaneous and endothermic. The adsorption enthalpy and entropy were found as 14-18.5 kJ mol(-1) and 52.8-67 J mol(-1) K(-1), respectively. The results obtained from the adsorption process using PPS as adsorbent was subjected to student's t-test.


Asunto(s)
Química Física/métodos , Azul de Metileno/química , Eliminación de Residuos Líquidos , Adsorción , Difusión , Residuos Industriales , Cinética , Modelos Químicos , Temperatura , Termodinámica , Factores de Tiempo , Contaminantes Químicos del Agua , Purificación del Agua
12.
Xenobiotica ; 37(2): 124-38, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17484516

RESUMEN

Recent studies have demonstrated that the pregnane X receptor (PXR) is a key regulator of cytochromes P450 3A (e.g. CYP3A4 in human) gene expression. As a result, activation of PXR may lead to CYP3A4 protein over-expression. Because induction of CYP3A4 could result in clinically important drug drug interactions, there has been a great interest in reducing the possibility of PXR activation by drug candidates in drug-discovery programmes. In order to provide structural insight for attenuating drug candidate-mediated PXR activation, we used a docking approach to study the structure activity relationship for PXR activators. Based on our docking models, it is proposed that introducing polar groups to the end of an activator should reduce its human PXR (hPXR) activity via destabilizing interactions in the hydrophobic areas of the PXR ligand-binding pocket. A number of analogues that incorporate these structural features then were designed and synthesized, and they exhibited significantly lower hPXR activation in a transactivation assay and decreased CYP3A4 induction in a human hepatocytes-based assay. In addition, an example in which attenuating hPXR activation was achieved by sterically destabilizing the helices 11 and 12 of the receptor is presented.


Asunto(s)
Receptores de Esteroides/química , Receptores de Esteroides/metabolismo , Adulto , Sitios de Unión , Línea Celular , Citocromo P-450 CYP3A , Sistema Enzimático del Citocromo P-450/biosíntesis , Sistema Enzimático del Citocromo P-450/genética , Inducción Enzimática , Femenino , Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Técnicas In Vitro , Ligandos , Masculino , Persona de Mediana Edad , Modelos Moleculares , Receptor X de Pregnano , Relación Estructura-Actividad , Xenobióticos/metabolismo , Xenobióticos/farmacología
13.
Xenobiotica ; 36(10-11): 902-37, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17118914

RESUMEN

To examine species-specific aspects of the induction of absorption, distribution, metabolism and excretion (ADME)-related genes, we used 25 000 gene oligonucleotide microarrays to construct a rodent gene-response compendium that compared hepatic gene expression profiles and developed consensus aryl hydrocarbon receptor (AhR), constitutive androstane receptor (CAR) and pregnane X-receptor (PXR) ligand signatures relevant to drug clearance. Twenty-six inducer compounds were chosen from the literature. Rats and mice received one of six dose levels (log2 dose escalation, 32-fold dose range) of each compound daily for 3 days. Animals were necropsied 6-9 h after the last dose, and tissues were collected for RNA analysis. Hepatic gene expression profiles were obtained using Rosetta Resolver expression analysis system, and ADME-related genes were extracted. Cross-talk among nuclear receptors or hepatoxicity at high dose levels resulted in large signatures (usually >1000 genes at p < 0.01) for most compounds. After ADME gene transcript enrichment, agglomerative clustering separated AhR ligands from CAR/PXR ligands, but it was difficult to distinguish CAR from PXR ligands. Consensus signatures were derived from groups of AhR, CAR and PXR ligands; and cross-talk among responding genes was determined. Many compounds had distinct log dose-response profiles, and relative potencies for ligands were established. Robust responses by CYP1A1, CYP2B10 (CAR responsive in mice) and CYP2B15 (CAR responsive in rats) and CYP3A1 (PXR responsive in rats) were used to benchmark the relative potency of different ligands and to determine the relative selectivity for AhR, CAR or PXR. By using a compendium of gene expression profiles, we defined species-specific induction patterns across the ADME transcriptome.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hígado/metabolismo , Análisis por Micromatrices/métodos , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Análisis por Conglomerados , Receptor de Androstano Constitutivo , Relación Dosis-Respuesta a Droga , Femenino , Ligandos , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Receptor X de Pregnano , Ratas , Ratas Sprague-Dawley , Receptores de Hidrocarburo de Aril/genética , Receptores Citoplasmáticos y Nucleares/genética , Receptores de Esteroides/genética , Factores de Transcripción/genética
14.
Xenobiotica ; 36(10-11): 963-88, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17118916

RESUMEN

Carrier-mediated transporters play a critical role in xenobiotic disposition and transporter research is complicated by species differences and their selective tissue expression. The purpose of this study was to generate a comprehensive data set of xenobiotic transporter gene expression profiles in humans and the pre-clinical species mouse, rat, beagle dog and cynomolgus monkey. mRNA expression profiles of 50 genes from the ABC, SLC and SLCO transporter superfamilies were examined in 40 human tissues by microarray analyses. Transporter genes that were identified as enriched in the liver or kidney, or that were selected for their known roles in xenobiotic disposition, were then compared in 22 tissues across the five species. Finally, as clinical variability in drug response and adverse reactions may be the result of variability in transporter gene expression, variability in the expression of selected transporter genes in 75 human liver donors were examined and compared with the highly variable drug metabolizing enzyme CYP3A4.


Asunto(s)
Perfilación de la Expresión Génica , Proteínas de Transporte de Membrana/genética , Xenobióticos/metabolismo , Animales , Femenino , Expresión Génica , Humanos , Inactivación Metabólica , Riñón/metabolismo , Hígado/metabolismo , Masculino , Especificidad de la Especie , Donantes de Tejidos
15.
J Cell Sci ; 113 Pt 24: 4451-61, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11082039

RESUMEN

The human multidrug resistance protein (MRP1) contributes to drug resistance in cancer cells. In addition to an MDR1-like core, MRP1 contains an N-terminal membrane-bound (TMD(0)) region and a cytoplasmic linker (L(0)), both characteristic of several members of the MRP family. In order to study the role of the TMD(0) and L(0) regions, we constructed various truncated and mutated MRP1, and chimeric MRP1-MDR1 molecules, which were expressed in insect (Sf9) and polarized mammalian (MDCKII) cells. The function of the various proteins was examined in isolated membrane vesicles by measuring the transport of leukotriene C(4) and other glutathione conjugates, and by vanadate-dependent nucleotide occlusion. Cellular localization, and glutathione-conjugate and drug transport, were also studied in MDCKII cells. We found that chimeric proteins consisting of N-terminal fragments of MRP1 fused to the N terminus of MDR1 preserved the transport, nucleotide occlusion and apical membrane routing of wild-type MDR1. As shown before, MRP1 without TMD(0)L(0) (Delta MRP1), was non-functional and localized intracellularly, so we investigated the coexpression of Delta MRP1 with the isolated L(0) region. Coexpression yielded a functional MRP1 molecule in Sf9 cells and routing to the lateral membrane in MDCKII cells. Interestingly, the L(0) peptide was found to be associated with membranes in Sf9 cells and could only be solubilized by urea or detergent. A 10-amino-acid deletion in a predicted amphipathic region of L(0) abolished its attachment to the membrane and eliminated MRP1 transport function, but did not affect membrane routing. Taken together, these experiments suggest that the L(0) region forms a distinct domain within MRP1, which interacts with hydrophobic membrane regions and with the core region of MRP1.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Animales , Línea Celular , Perros , Expresión Génica , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Mutagénesis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Spodoptera/citología
16.
Br J Cancer ; 83(3): 366-74, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10917553

RESUMEN

The human multidrug transporter MDR1 P-glycoprotein and the multidrug resistance proteins MRP1 and MRP2 transport a range of cytotoxic drugs, resulting in multidrug resistance in tumour cells. To overcome this form of drug resistance in patients, several inhibitors (reversal agents) of these transporters have been isolated. Using polarized cell lines stably expressing human MDR1, MRP1 or MRP2cDNA, and 2008 ovarian carcinoma cells stably expressing MRP1 cDNA, we have investigated in this study the specificity of the reversal agents V-104 (a pipecolinate derivative), GF120918 (an acridone carboxamide derivative also known as GG918), and Pluronic L61 (a (poly)oxypropethylene and (poly)oxypropylene block copolymer). Transport experiments with cytotoxic drugs with polarized cell lines indicate that all three compounds efficiently inhibit MDR1 Pgp. Furthermore, V-104 partially inhibits daunorubicin transport by MRP1 but not vinblastine transport by MRP2. V-104 reverses etoposide resistance of 2008/MRP1 cells, whereas GF120918 does not reverse resistance due to MRP1. V-104 partially inhibits the export of the organic anion dinitrophenyl S-glutathione by MDCKII-MRP1 but not by MDCKII-MRP2 cells. Unexpectedly, export of the organic anion calcein by MDCKII-MRP1 and MDCKII-MRP2 cells is stimulated by Pluronic L61, probably because it relieves the block on entry of calcein AM into the cell by endogenous MDR1 Pgp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Acridinas/farmacología , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Isoquinolinas/farmacología , Metanfetamina/análogos & derivados , Poloxámero/farmacología , Tetrahidroisoquinolinas , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/efectos de los fármacos , Proteínas de Transporte de Anión , Antibióticos Antineoplásicos/farmacocinética , Antineoplásicos Fitogénicos/farmacocinética , Transporte Biológico Activo/efectos de los fármacos , Proteínas Portadoras/farmacología , Daunorrubicina/farmacocinética , Etopósido/farmacocinética , Fluoresceínas/farmacología , Humanos , Metanfetamina/farmacología , Factores de Tiempo , Vinblastina/farmacocinética
17.
Br J Cancer ; 83(3): 375-83, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10917554

RESUMEN

The multidrug resistance proteins MRP1 and MRP2 are members of the same subfamily of ATP-binding cassette transporters. Besides organic molecules conjugated to negatively charged ligands, these proteins also transport cytotoxic drugs for which no negatively charged conjugates are known to exist. In polarized MDCKII cells, MRP1 routes to the lateral plasma membrane, and MRP2 to the apical plasma membrane. In these cells MRP1 transports daunorubicin, and MRP2 vinblastine; both transporters export reduced glutathione (GSH) into the medium. We demonstrate that glutathione transport in MDCKII-MRP1 cells is inhibited by the inhibitors of organic anion transporters sulfinpyrazone, indomethacin, probenecid and benzbromarone. In MDCKII-MRP2 cells, GSH export is stimulated by low concentrations of sulfinpyrazone or indomethacin, whereas export is inhibited down to control levels at high concentrations. We find that unmodified sulfinpyrazone is a substrate for MRP2, also at concentrations where GSH export is inhibited. We also show that GSH export in MDCKII-MRP2 cells increases in the presence of vinblastine, and that the stoichiometry between drug and GSH exported is between two and three. Our data indicate that transport of sulfinpyrazone and vinblastine is associated with GSH export. However, at high sulfinpyrazone concentrations this compound is transported without GSH. Models of MRP action are discussed that could explain these results.


Asunto(s)
Antineoplásicos Fitogénicos/metabolismo , Proteínas Portadoras/metabolismo , Glutatión/metabolismo , Sulfinpirazona/metabolismo , Uricosúricos/metabolismo , Vinblastina/metabolismo , Proteínas de Transporte de Anión , Antiinflamatorios no Esteroideos/farmacología , Benzbromarona/farmacología , Transporte Biológico Activo/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Indometacina/farmacología , Probenecid/farmacología , Células Tumorales Cultivadas
18.
J Natl Cancer Inst ; 92(16): 1295-302, 2000 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-10944550

RESUMEN

The human multidrug resistance-associated protein (MRP) family currently has seven members. The ability of several of these membrane proteins to transport a wide range of anticancer drugs out of cells and their presence in many tumors make them prime suspects in unexplained cases of drug resistance, although proof that they contribute to clinical drug resistance is still lacking. Recent studies have begun to clarify the function of the MRP family members. MRPs are organic anion transporters; i.e., they transport anionic drugs, exemplified by methotrexate, and neutral drugs conjugated to acidic ligands, such as glutathione (GSH), glucuronate, or sulfate. However, MRP1, MRP2, and MRP3 can also cause resistance to neutral organic drugs that are not known to be conjugated to acidic ligands by transporting these drugs together with free GSH. MRP1 can even confer resistance to arsenite and MRP2 to cisplatin, again probably by transporting these compounds in complexes with GSH. MRP4 overexpression is associated with high-level resistance to the nucleoside analogues 9-(2-phosphonylmethoxyethyl) adenine and azidothymidine, both of which are used as anti-human immunodeficiency virus drugs. MRPs may, therefore, also have a role in resistance against nucleoside analogues used in cancer chemotherapy. Mice without Mrp1, a high-affinity leukotriene C(4) transporter, have an altered response to inflammatory stimuli but are otherwise healthy and fertile. MRP2 is the major transporter responsible for the secretion of bilirubin glucuronides into bile, and humans without MRP2 develop a mild liver disease known as the Dubin-Johnson syndrome. The physiologic functions of the other MRPs are not known. Whether long-term inhibition of MRPs in humans can be tolerated (assuming that suitable inhibitors will be found) remains to be determined.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Regulación hacia Arriba
19.
Mol Pharmacol ; 57(4): 760-8, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10727523

RESUMEN

The human multidrug resistance protein MRP1 and its homolog, MRP2, are both suggested as being involved in cancer drug resistance and the transport of organic anions. We expressed MRP1 and MRP2 in Spodoptera frugiperda ovarian cells and compared their ATP-dependent transport properties and vanadate-sensitive ATPase activities in isolated membrane vesicles. Both MRP1 and MRP2 actively transported leukotriene C(4) and N-ethylmaleimide glutathione (NEM-GS), although the relative affinity of MRP2 for these substrates was found to be significantly lower than that of MRP1. Methotrexate was actively transported by both proteins, although more efficiently by MRP2. ATP-dependent NEM-GS transport by MRP1 and MRP2 was variably modulated by organic anions. Probenecid and furosemide inhibited, whereas under certain conditions sulfinpyrazone, penicillin G, and indomethacin greatly stimulated, MRP2-mediated NEM-GS uptake. Vanadate-sensitive ATPase activity in isolated membranes containing MRP1 or MRP2 was significantly stimulated by NEM-GS and reduced GS, although these compounds acted only at higher concentrations in MRP2. ATP hydrolysis by MRP2 was also effectively stimulated by methotrexate. Probenecid, sulfinpyrazone, indomethacin, furosemide, and penicillin G all significantly increased MRP2-ATPase activity, whereas these compounds acted more as ATPase inhibitors on MRP1. These results indicate that MRP1 is a more efficient transporter of glutathione conjugates and free glutathione than MRP2, whereas several anions are preferred substrates for MRP2. Our data suggest that MRP2 may be responsible for the active secretion of pharmacologically relevant organic anions, such as diuretics and antibiotics, and indicate different modulation possibilities for MRP1 or MRP2 in drug-resistant tumor cells.


Asunto(s)
Aniones/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Transporte de Membrana , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Adenosina Trifosfatasas/metabolismo , Animales , Transporte Biológico , Membrana Celular/metabolismo , Células Cultivadas , Resistencia a Múltiples Medicamentos , Glutatión/metabolismo , Humanos , Insectos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteína 3 Homóloga de MutS
20.
Biochim Biophys Acta ; 1461(2): 347-57, 1999 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-10581366

RESUMEN

The human multidrug resistance protein (MRP) family contains at least six members: MRP1, the godfather of the family and well known as the multidrug resistance protein, and five homologs, called MRP2-6. In this review, we summarize what is known about the protein structure, the expression in tissues, the routing in cells, the physiological functions, the substrate specificity, and the role in multidrug resistance of the individual members of the MRP family.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/genética , Resistencia a Múltiples Medicamentos/genética , Proteínas de Transporte de Membrana , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Proteínas de Transporte de Anión , Proteínas Portadoras/genética , Línea Celular , Membrana Celular/metabolismo , Glutatión/metabolismo , Humanos , Modelos Moleculares , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Filogenia , Terminología como Asunto , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA