Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Drug Metab Dispos ; 45(6): 604-611, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28325716

RESUMEN

Inside-out-oriented membrane vesicles are useful tools to investigate whether a compound can be an inhibitor of efflux transporters such as multidrug resistance-associated protein 2 (MRP2). However, because of technical limitations of substrate diffusion and low dynamic uptake windows for interacting drugs used in the clinic, estradiol-17ß-glucuronide (E17ßG) remains the probe substrate that is frequently used in MRP2 inhibition assays. Here we recapitulated the sigmoidal kinetics of MRP2-mediated transport of E17ßG, with apparent Michaelis-Menten constant (Km) and Vmax values of 170 ±17 µM and 1447 ± 137 pmol/mg protein/min, respectively. The Hill coefficient (2.05 ± 0.1) suggests multiple substrate binding sites for E17ßG transport with cooperative interactions. Using E17ßG as a probe substrate, 51 of 97 compounds tested (53%) showed up to 6-fold stimulatory effects. Here, we demonstrate for the first time that coproporphyrin-I (CP-I) is a MRP2 substrate in membrane vesicles. The uptake of CP-I followed a hyperbolic relationship, adequately described by the standard Michaelis-Menten equation (apparent Km and Vmax values were 7.7 ± 0.7 µM and 48 ± 11 pmol/mg protein/min, respectively), suggesting the involvement of a single binding site. Of the 47 compounds tested, 30 compounds were inhibitors of human MRP2 and 8 compounds (17%) stimulated MRP2-mediated CP-I transport. The stimulators were found to share the basic backbone structure of the physiologic steroids, which suggests a potential in vivo relevance of in vitro stimulation of MRP2 transport. We concluded that CP-I could be an alternative in vitro probe substrate replacing E17ßG for appreciating MRP2 interactions while minimizing potential false-negative results for MRP2 inhibition due to stimulatory effects.


Asunto(s)
Coproporfirinas/farmacología , Colorantes Fluorescentes/farmacología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/análisis , Sitios de Unión/efectos de los fármacos , Transporte Biológico/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/metabolismo , Humanos , Cinética , Proteínas de Transporte de Membrana/metabolismo , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos
2.
J Mol Recognit ; 27(10): 609-17, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25178856

RESUMEN

The present study involves molecular docking, molecular dynamics (MD) simulation studies, and Caco-2 cell monolayer permeability assay to investigate the effect of structural modifications on PepT1-mediated transport of thyrotropin releasing hormone (TRH) analogs. Molecular docking of four TRH analogs was performed using a homology model of human PepT1 followed by subsequent MD simulation studies. Caco-2 cell monolayer permeability studies of four TRH analogs were performed at apical to basolateral and basolateral to apical directions. Inhibition experiments were carried out using Gly-Sar, a typical PepT1 substrate, to confirm the PepT1-mediated transport mechanism of TRH analogs. Papp of the four analogs follows the order: NP-1894 < NP-2378 < NP-1896 < NP-1895. Higher absorptive transport was observed in the case of TRH analogs, indicating the possibility of a carrier-mediated transport mechanism. Further, the significant inhibition of the uptake of Gly-Sar by TRH analogs confirmed the PepT1-mediated transport mechanism. Glide docking scores of all the four analogues were in good agreement with their transport rates, suggesting the role of substrate binding affinity in the PepT1-mediated transport of TRH analogs. MD simulation studies revealed that the polar interactions with amino acid residues present in the active site are primarily responsible for substrate binding, and a downward trend was observed with the increase in bulkiness at the N-histidyl moiety of TRH analogs.


Asunto(s)
Mucosa Intestinal/metabolismo , Modelos Moleculares , Simportadores/química , Hormona Liberadora de Tirotropina/química , Sitios de Unión , Transporte Biológico , Células CACO-2 , Cromatografía Líquida de Alta Presión , Biología Computacional , Simulación por Computador , Humanos , Simulación de Dinámica Molecular , Transportador de Péptidos 1 , Permeabilidad , Simportadores/metabolismo , Simportadores/fisiología , Hormona Liberadora de Tirotropina/análogos & derivados , Hormona Liberadora de Tirotropina/farmacocinética
3.
Int J Pharm ; 464(1-2): 104-10, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24463070

RESUMEN

11-Keto-ß-boswellic acid (KBA) is an important and potent boswellic acids responsible for anti-inflammatory action of Boswellia extract. However, its pharmaceutical development has been severely limited by its poor oral bioavailability. The present work aims to investigate the permeability related hurdles in oral delivery of KBA. Gastrointestinal stability, gastrointestinal metabolism, adsorption-desorption kinetics and Caco-2 permeability studies have been carried out. KBA was found poorly permeable with Papp value of 2.85 ± 0.14 × 10(-6)cm/s. Higher absorptive transport indicated role of carrier mediated transport. Moreover, KBA transport across monolayer showed saturation kinetics at higher concentrations. KBA exposed to 1α,25-(OH)2 vitamin D3 treated cell monolayer showed the lowest Papp value of 2.01×10(-6) ± 0.02 × 10(-6)cm/s indicating role of CYP3A4 mediated metabolism during KBA transport. Metabolic stability experiments in jejunum S9 fractions further confirmed this. KBA was found unstable in simulated gastrointestinal fluids and also got accumulated in the enterocytes. Sorption and desorption kinetic studies using Caco-2 cells further confirmed accumulation of KBA inside the enterocytes. KBA also showed pH dependent permeability with higher flux at gradient pH condition of pH 6.5 at apical and 7.4 at basolateral. Taken as whole, the major permeability related hurdles that hampered oral bioavailability of KBA included its gastrointestinal instability, CYP3A4 mediated intestinal metabolism, accumulation within the enterocytes and saturable kinetics. The present investigation may help in designing novel drug delivery system for KBA.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Triterpenos/administración & dosificación , Triterpenos/metabolismo , Administración Oral , Animales , Células CACO-2 , Humanos , Absorción Intestinal/efectos de los fármacos , Absorción Intestinal/fisiología , Masculino , Permeabilidad/efectos de los fármacos , Ratas Sprague-Dawley
4.
J Comput Aided Mol Des ; 27(4): 347-63, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23612916

RESUMEN

P-glycoprotein (P-gp) is a plasma membrane efflux transporter belonging to ATP-binding cassette superfamily, responsible for multidrug resistance in tumor cells. Over-expression of P-gp in cancer cells limits the efficacy of many anticancer drugs. A clear understanding of P-gp substrate binding will be advantageous in early drug discovery process. However, substrate poly-specificity of P-gp is a limiting factor in rational drug design. In this investigation, we report a dynamic trans-membrane model of P-gp that accurately identified the substrate binding residues of known anticancer agents. The study included homology modeling of human P-gp based on the crystal structure of C. elegans P-gp, molecular docking, molecular dynamics analyses and binding free energy calculations. The model was further utilized to speculate substrate propensity of in-house anticancer compounds. The model demonstrated promising results with one anticancer compound (NSC745689). As per our observations, the molecule could be a potential lead for anticancer agents devoid of P-gp mediated multiple drug resistance. The in silico results were further validated experimentally using Caco-2 cell lines studies, where NSC745689 exhibited poor permeability (P app 1.03 ± 0.16 × 10(-6) cm/s) and low efflux ratio of 0.26.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Neoplasias/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Animales , Antineoplásicos/farmacocinética , Células CACO-2 , Caenorhabditis elegans/química , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Simulación por Computador , Cristalografía por Rayos X , Humanos , Modelos Biológicos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Homología Estructural de Proteína , Especificidad por Sustrato , Termodinámica
5.
Mol Pharm ; 9(9): 2458-68, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22779445

RESUMEN

The present study, in general, is aimed to uncover the properties of the transport mechanism or mechanisms responsible for the uptake of NP-647 into Caco-2 cells and, in particular, to understand whether it is a substrate for the intestinal oligopeptide transporter, PEPT1 (SLC15A1). NP-647 showed a carrier-mediated, saturable transport with Michaelis-Menten parameters K(m) = 1.2 mM and V(max) = 2.2 µM/min. The effect of pH, sodium ion (Na(+)), glycylsarcosine and amoxicillin (substrates of PEPT1), and sodium azide (Na(+)/K(+)-ATPase inhibitor) on the flux rate of NP-647 was determined. Molecular docking and molecular dynamics simulation studies were carried out to investigate molecular interactions of NP-647 with transporter using homology model of human PEPT1. The permeability coefficient (P(appCaco-2)) of NP-647 (32.5 × 10(-6) cm/s) was found to be four times higher than that of TRH. Results indicate that NP-647 is transported into Caco-2 cells by means of a carrier-mediated, proton-dependent mechanism that is inhibited by Gly-Sar and amoxicillin. In turn, NP-647 also inhibits the uptake of Gly-Sar into Caco-2 cells and, together, this evidence suggests that PEPT1 is involved in the process. Docking and molecular dynamics simulation studies indicate high affinity of NP-647 toward PEPT1 binding site as compared to TRH. High permeability of NP-647 over TRH is attributed to its increased hydrophobicity which increases its affinity toward PEPT1 by interacting with the hydrophobic pocket of the transporter through hydrophobic forces.


Asunto(s)
Anticonvulsivantes/farmacocinética , Simportadores/metabolismo , Hormona Liberadora de Tirotropina/análogos & derivados , Amoxicilina/farmacología , Anticonvulsivantes/química , Anticonvulsivantes/metabolismo , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Línea Celular Tumoral , Dipéptidos/farmacología , Estabilidad de Medicamentos , Humanos , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Absorción Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Modelos Moleculares , Simulación de Dinámica Molecular , Transportador de Péptidos 1 , Sodio/metabolismo , Azida Sódica/metabolismo , Simportadores/química , Hormona Liberadora de Tirotropina/química , Hormona Liberadora de Tirotropina/metabolismo , Hormona Liberadora de Tirotropina/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...