Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Commun Biol ; 4(1): 55, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33420418

RESUMEN

Cytochrome P450 (CYP) heme monooxygenases require two electrons for their catalytic cycle. For mammalian microsomal CYPs, key enzymes for xenobiotic metabolism and steroidogenesis and important drug targets and biocatalysts, the electrons are transferred by NADPH-cytochrome P450 oxidoreductase (CPR). No structure of a mammalian CYP-CPR complex has been solved experimentally, hindering understanding of the determinants of electron transfer (ET), which is often rate-limiting for CYP reactions. Here, we investigated the interactions between membrane-bound CYP 1A1, an antitumor drug target, and CPR by a multiresolution computational approach. We find that upon binding to CPR, the CYP 1A1 catalytic domain becomes less embedded in the membrane and reorients, indicating that CPR may affect ligand passage to the CYP active site. Despite the constraints imposed by membrane binding, we identify several arrangements of CPR around CYP 1A1 that are compatible with ET. In the complexes, the interactions of the CPR FMN domain with the proximal side of CYP 1A1 are supplemented by more transient interactions of the CPR NADP domain with the distal side of CYP 1A1. Computed ET rates and pathways agree well with available experimental data and suggest why the CYP-CPR ET rates are low compared to those of soluble bacterial CYPs.


Asunto(s)
Membrana Celular/metabolismo , Citocromo P-450 CYP1A1/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Transporte de Electrón , Humanos , Simulación de Dinámica Molecular , Dominios Proteicos
2.
Sci Rep ; 10(1): 7284, 2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32350331

RESUMEN

The simulation of membrane proteins requires compatible protein and lipid force fields that reproduce the properties of both the protein and the lipid bilayer. Cytochrome P450 enzymes are bitopic membrane proteins with a transmembrane helical anchor and a large cytosolic globular domain that dips into the membrane. As such, they are representative and challenging examples of membrane proteins for simulations, displaying features of both peripheral and integral membrane proteins. We performed molecular dynamics simulations of three cytochrome P450 isoforms (2C9, 2C19 and 1A1) in a 2-oleoyl-1-palmitoyl-sn-glycerol-3-phosphocholine bilayer using two AMBER force field combinations: GAFF-LIPID with ff99SB for the protein, and LIPID14 with ff14SB for the protein. Comparison of the structural and dynamic properties of the proteins, the lipids and the protein-membrane interactions shows differing sensitivity of the cytochrome P450 isoforms to the choice of force field, with generally better agreement with experiment for the LIPID14 + ff14SB combination.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Membrana Dobles de Lípidos/química , Lípidos de la Membrana/química , Simulación de Dinámica Molecular , Animales , Humanos , Estructura Secundaria de Proteína
3.
Int J Mol Sci ; 20(18)2019 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-31487853

RESUMEN

The human cytochrome P450 (CYP) 2C9 and 2C19 enzymes are two highly similar isoforms with key roles in drug metabolism. They are anchored to the endoplasmic reticulum membrane by their N-terminal transmembrane helix and interactions of their cytoplasmic globular domain with the membrane. However, their crystal structures were determined after N-terminal truncation and mutating residues in the globular domain that contact the membrane. Therefore, the CYP-membrane interactions are not structurally well-characterized and their dynamics and the influence of membrane interactions on CYP function are not well understood. We describe herein the modeling and simulation of CYP 2C9 and CYP 2C19 in a phospholipid bilayer. The simulations revealed that, despite high sequence conservation, the small sequence and structural differences between the two isoforms altered the interactions and orientations of the CYPs in the membrane bilayer. We identified residues (including K72, P73, and I99 in CYP 2C9 and E72, R73, and H99 in CYP 2C19) at the protein-membrane interface that contribute not only to the differing orientations adopted by the two isoforms in the membrane, but also to their differing substrate specificities by affecting the substrate access tunnels. Our findings provide a mechanistic interpretation of experimentally observed effects of mutagenesis on substrate selectivity.


Asunto(s)
Citocromo P-450 CYP2C19/química , Citocromo P-450 CYP2C9/química , Fosfolípidos/metabolismo , Sitios de Unión , Citocromo P-450 CYP2C19/metabolismo , Citocromo P-450 CYP2C9/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica
4.
Biophys J ; 116(3): 419-432, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30658838

RESUMEN

Human cytochrome P450 (CYP) enzymes play an important role in the metabolism of drugs, steroids, fatty acids, and xenobiotics. Microsomal CYPs are anchored in the endoplasmic reticulum membrane by an N-terminal transmembrane (TM) helix that is connected to the globular catalytic domain by a flexible linker sequence. However, the structural and functional importance of the TM-helix is unclear because it has been shown that CYPs can still associate with the membrane and have enzymatic activity in reconstituted systems after truncation or modification of the N-terminal sequence. Here, we investigated the effect of mutations in the N-terminal TM-helix residues of two human steroidogenic enzymes, CYP 17A1 and CYP 19A1, that are major drug targets for cancer therapy. These mutations were originally introduced to increase the expression of the proteins in Escherichia coli. To investigate the effect of the mutations on protein-membrane interactions and function, we carried out coarse-grained and all-atom molecular dynamics simulations of the CYPs in a phospholipid bilayer. We confirmed the orientations of the globular domain in the membrane observed in the simulations by linear dichroism measurements in a Nanodisc. Whereas the behavior of CYP 19A1 was rather insensitive to truncation of the TM-helix, mutations in the TM-helix of CYP 17A1, especially W2A and E3L, led to a gradual drifting of the TM-helix out of the hydrophobic core of the membrane. This instability of the TM-helix could affect interactions with the allosteric redox partner, cytochrome b5, required for CYP 17A1's lyase activity. Furthermore, the simulations showed that the mutant TM-helix influenced the membrane interactions of the CYP 17A1 globular domain. In some simulations, the mutated TM-helix obstructed the substrate access tunnel from the membrane to the CYP active site, indicating a possible effect on enzyme function.


Asunto(s)
Aromatasa/química , Aromatasa/metabolismo , Membrana Celular/metabolismo , Mutación , Esteroide 17-alfa-Hidroxilasa/química , Esteroide 17-alfa-Hidroxilasa/metabolismo , Secuencia de Aminoácidos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Esteroide 17-alfa-Hidroxilasa/genética
5.
Biomed Chromatogr ; 30(10): 1556-72, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27006091

RESUMEN

The aim of the present study was to evaluate the contribution of metabolites to drug-drug interaction and drug-herb interaction using the inhibition of CYP2D6 and CYP3A4 by metoprolol (MET) and its metabolites. The peak concentrations of unbound plasma concentration of MET, α-hydroxy metoprolol (HM), O-desmethyl metoprolol (ODM) and N-desisopropyl metoprolol (DIM) were 90.37 ± 2.69, 33.32 ± 1.92, 16.93 ± 1.70 and 7.96 ± 0.94 ng/mL, respectively. The metabolites identified, HM and ODM, had a ratio of metabolic area under the concentration-time curve (AUC) to parent AUC of ≥0.25 when either total or unbound concentration of metabolite was considered. In vitro CYP2D6 and CYP3A4 inhibition by MET, HM and ODM study revealed that MET, HM and ODM were not inhibitors of CYP3A4-catalyzed midazolam metabolism and CYP2D6-catalyzed dextromethorphan metabolism. However, DIM only met the criteria of >10% of the total drug related material and <25% of the parent using unbound concentrations. If CYP inhibition testing is solely based on metabolite exposure, DIM metabolite would probably not be considered. However, the present study has demonstrated that DIM contributes significantly to in vitro drug-drug interaction. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Inhibidores del Citocromo P-450 CYP2D6/farmacología , Citocromo P-450 CYP2D6/efectos de los fármacos , Inhibidores del Citocromo P-450 CYP3A/farmacología , Citocromo P-450 CYP3A/efectos de los fármacos , Metoprolol/farmacología , Espectrometría de Masa por Ionización de Electrospray/métodos , Espectrometría de Masas en Tándem/métodos , Área Bajo la Curva , Inhibidores del Citocromo P-450 CYP2D6/metabolismo , Inhibidores del Citocromo P-450 CYP3A/metabolismo , Interacciones Farmacológicas , Humanos , Metoprolol/metabolismo
6.
Eur J Med Chem ; 115: 82-93, 2016 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-26994845

RESUMEN

CYP1A1 is a potential target for anticancer drug development due to its overexpression in certain cancer cells and role in cancer progression. To identify new leads for CYP1A1 mediated anticancer action, we attempted ligand based pharmacophore mapping, virtual screening of databases, molecular docking, MetaSite based filtering, and molecular dynamics simulations. Initial computational and in vitro screening identified 11 compounds from which we identified two lead compounds, ZINC33468944 and ZINC32101539, showed potential antitumor activity on MDA-MB-435 cell lines (GI50 < 0.1 µM) and CYP1A1 inhibition of 0.13 and 0.3 µM, respectively. Furthermore, the lead compounds were evaluated for CYP1A1 mediated metabolism, showing N-hydroxylated metabolites, which have potential of DNA adduct formation and cause cancerous cell death. Analysis of molecular dynamics simulations provided important guidelines for the further modification of the lead compounds. Hence, we claim the lead molecules for further development in anticancer drug discovery.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Citocromo P-450 CYP1A1/antagonistas & inhibidores , Indoles/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Indoles/síntesis química , Indoles/química , Estructura Molecular , Relación Estructura-Actividad
7.
J Mol Recognit ; 29(8): 370-90, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26916064

RESUMEN

Recent trends in new drug discovery of anticancer drugs have made oncologists more aware of the fact that the new drug discovery must target the developing mechanism of tumorigenesis to improve the therapeutic efficacy of antineoplastic drugs. The drugs designed are expected to have high affinity towards the novel targets selectively. Current research highlights overexpression of CYP450s, particularly cytochrome P450 1A1 (CYP1A1), in tumour cells, representing a novel target for anticancer therapy. However, the CYP1 family is identified as posing significant problems in selectivity of anticancer molecules towards CYP1A1. Three members have been identified in the human CYP1 family: CYP1A1, CYP1A2 and CYP1B1. Although sequences of the three isoform have high sequence identity, they have distinct substrate specificities. The understanding of macromolecular features that govern substrate specificity is required to understand the interplay between the protein function and dynamics, design novel antitumour compounds that could be specifically metabolized by only CYP1A1 to mediate their antitumour activity and elucidate the reasons for differences in substrate specificity profile among the three proteins. In the present study, we employed a combination of computational methodologies: molecular docking and molecular dynamics simulations. We utilized eight substrates for elucidating the difference in substrate specificity of the three isoforms. Lastly, we conclude that the substrate specificity of a particular substrate depends upon the type of the active site residues, the dynamic motions in the protein structure upon ligand binding and the physico-chemical characteristics of a particular ligand. Copyright © 2016 John Wiley & Sons, Ltd.


Asunto(s)
Antineoplásicos/farmacología , Citocromo P-450 CYP1A1/química , Citocromo P-450 CYP1A2/química , Citocromo P-450 CYP1B1/química , Antineoplásicos/química , Dominio Catalítico/efectos de los fármacos , Citocromo P-450 CYP1A1/metabolismo , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP1B1/metabolismo , Bases de Datos de Compuestos Químicos , Diseño de Fármacos , Humanos , Ligandos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Especificidad por Sustrato
8.
Eur J Med Chem ; 111: 72-83, 2016 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-26854379

RESUMEN

Thyrotropin-releasing hormone (TRH) analogues bearing halogen groups (Cl, Br and I) at the C-2 and/or C-5 position, and the alkyl group (CH3, C2H5, C3H7, CH2C6H5) at the N-1 position of the imidazole ring of the central histidine residue were synthesized and evaluated for the receptor binding, calcium mobilization (FLIPR), and IP-1 assay at the HEK mTRHR1 and HEK mTRHR2 expressing cell lines. The most promising analogue 7k showed 925-fold selectivity for HEK mTRH-R2 receptor subtype in the IP-1 assay, 272-fold selectivity for HEK mTRH-R2 receptor subtype in the FLIPR assay, and 21-fold receptor binding specificity at HEK TRH-R2 receptor subtype. The peptide 7k was evaluated in vitro in a brain membrane competitive binding assay, and for stability analysis in the presence of TRH-DE, in vivo. The analogue 7k showed decrease in the sleeping time by more than 76% in a pentobarbital-induced sleeping assay, and showed comparatively less elevation in the TSH level in the blood, in vivo. The computational homology modeling of TRH-R1 and TRH-R2 and docking study with the most potent peptide 7k provide impetus to design CNS specific TRH analogues.


Asunto(s)
Histidina/metabolismo , Hormona Liberadora de Tirotropina/análogos & derivados , Hormona Liberadora de Tirotropina/metabolismo , Células HEK293 , Histidina/química , Humanos , Modelos Moleculares , Conformación Molecular , Receptores de Hormona Liberadora de Tirotropina/química , Receptores de Hormona Liberadora de Tirotropina/metabolismo , Hormona Liberadora de Tirotropina/química
9.
Bioorg Med Chem ; 23(17): 5641-53, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26216015

RESUMEN

Thyrotropin-releasing hormone (TRH)-like peptides were synthesized by replacing critical histidine and pGlu residues in the native peptide. The peptides were evaluated in vitro for receptor binding activity assay and in the cell functional assay; the peptides exhibit selective basal signaling agonist behavior toward TRH-R2. For example, peptides 8a, 8b, 8c, 8 f, 8 h, 8 l and 12 d activated TRH-R2 with potency (EC50) of 0.53 µM, 0.048 µM, 0.05 µM, 0.006 µM, 0.31 µM, 0.034 µM and 0.004 µM, respectively. In contrast for signaling activation of TRH-R1, the same peptide required higher concentration of 19.35 µM, 3.98 µM, 2.54 µM, 0.287 µM, 11.28 µM, 0.986 µM and 0.944 µM, respectively. The results showed that peptides were 36.5, 82.9, 50.8, 47.8, 36.3, 32.6 and 235-fold selective to TRH-R2 receptor subtype. The peptides were investigated for CNS activity at 10 µmol/kg in pentobarbital-induced sleep assay study. Peptides 8c (16.5 ± 1.4 min) and 8l (16.5 ± 2.1 min) displayed excellent CNS activity. In an in vivo study, peptide 8c did not cause significant change in the rat plasma TSH levels. The peptide 8c was further investigated for neuroprotective potential, and significantly reduced infracts volume and neurological score in the focal cerebral ischemia model in mice. Peptide 8c also significantly lowered MDA levels, indicating reduction of oxidative and enhanced percentage cell survival in CA1 region, when compared to ischemic brain.


Asunto(s)
Trastornos del Conocimiento/genética , Péptidos/metabolismo , Receptores de Hormona Liberadora de Tirotropina/química , Hormona Liberadora de Tirotropina/síntesis química , Animales , Isquemia Encefálica , Ratones , Estructura Molecular , Ratas
10.
J Chem Phys ; 143(24): 243139, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26723624

RESUMEN

An important step in the simulation of a membrane protein in a phospholipid bilayer is the correct immersion of the protein in the bilayer. Crystal structures are determined without the bilayer. Particularly for proteins with monotopic domains, it can be unclear how deeply and in which orientation the protein is being inserted in the membrane. We have previously developed a procedure combining coarse-grain (CG) with all-atom (AA) molecular dynamics (MD) simulations to insert and simulate a cytochrome P450 (CYP) possessing an N-terminal transmembrane helix connected by a flexible linker region to a globular domain that dips into the membrane. The CG simulations provide a computationally efficient means to explore different orientations and conformations of the CYP in the membrane. Converged configurations obtained in the CG simulations are then refined in AA simulations. Here, we tested different variants of the MARTINI CG model, differing in the water model, the treatment of long-range non-bonded interactions, and the implementation (GROMACS 4.5.5 vs 5.0.4), for this purpose. We examined the behavior of the models for simulating a 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayer in water and for the immersion of CYP3A4 in a POPC bilayer, and compared the CG-MD results with the previously reported experimental and simulation results. We also tested the methodology on a set of four other CYPs. Finally, we propose an optimized protocol for modeling such protein-membrane systems that provides the most plausible configurations and is computationally efficient; this incorporates the standard non-polar water model and the GROMACS 5.0.4 implementation with a reaction field treatment of long-range interactions.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Membrana Dobles de Lípidos/química , Proteínas de la Membrana/química , Simulación de Dinámica Molecular , Fosfatidilcolinas/química , Sistema Enzimático del Citocromo P-450/metabolismo , Difusión , Interacciones Hidrofóbicas e Hidrofílicas , Membrana Dobles de Lípidos/metabolismo , Proteínas de la Membrana/metabolismo , Fosfatidilcolinas/metabolismo
11.
J Pharm Biomed Anal ; 102: 386-99, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25459938

RESUMEN

Fidarestat, an aldose reductase inhibitor, has been used for the treatment of the diabetic associated complications such as retinopathy, neuropathy and nephropathy. To better understand the metabolism and pharmacokinetics of fidarestat, we have evaluated plasma protein binding, pharmacokinetics, tissue distribution of the drug and its conjugated metabolites and CYP450 biotransformation by liquid chromatography-high resolution mass spectrometry. Effective chromatographic separation of fidarestat and hydrochlorothiazide (IS) in rat plasma and tissues was achieved on Hypersil gold C-18 column in an isocratic elution mode. For detection, a high-resolution Orbitrap mass spectrometer with heated electrospray ionization inlet in the negative ion mode was used. High-resolution extracted ion chromatograms for each analyte were obtained by processing the full-scan MS mode with 5 ppm mass tolerance. The impact of plasma protein binding with the drug and conjugated metabolites of the drug on pharmacokinetics has been determined. The study indicated that 9.5% of free form of fidarestat may be pharmacologically active and the Cmax for free fidarestat was found to be 80.30 ± 6.78 ng/mL. The AUC0-t and AUC0-∞ were found to be 185.46 ± 32 and 195.92 ± 15.06 ng h/mL, respectively. Among tissues, the maximum observed distribution was found to be in kidney followed by liver and heart. Docking experiments and in vitro CYP450 reaction phenotyping revealed that two CYP1A2 and CYP2D6 are involved in the phase I metabolism of fidarestat. Oxidative deamination and N/O glucuronidation are the major phase I and phase II metabolites, respectively. In vitro CYP450 inhibition assay of fidarestat for drug-drug interaction showed weak inhibition and may not alter pharmacokinetics, distribution or clearance of other co-administered drug.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Imidazolidinas/metabolismo , Espectrometría de Masas en Tándem/métodos , Animales , Biotransformación/fisiología , Proteínas Sanguíneas/análisis , Cromatografía Liquida/métodos , Sistema Enzimático del Citocromo P-450/análisis , Femenino , Imidazolidinas/análisis , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Distribución Tisular/fisiología
12.
Protein J ; 33(6): 536-48, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25331835

RESUMEN

Apart from playing key roles in drug metabolism and adverse drug-drug interactions, CYPs are potential drug targets to treat a variety of diseases. The intervention of over expression of P450 1A1 (CYP1A1) in tumor cells is identified as a novel strategy for anticancer therapy. We investigated three isoforms of CYP1 family (CYP1A1, CYP1A2, and CYP1B1) for their substrate specificity. The understanding of macromolecular features that govern substrate specificity is required to understand the interplay between the protein function and dynamics. This can help in design of new antitumor molecule specifically metabolized by CYP1A1 to mediate their antitumor activity. In the present study, we carried out the comparative protein structure analysis of the three isoforms. Sequence alignment, root mean square deviation (RMSD) analysis, B-factor analysis was performed to give a better understanding of the macromolecular features involved in substrate specificity and to understand the interplay between protein dynamics and functions which will have important implications on rational design of anticancer drugs. We identified the differences in amino acid residues among the three isoforms of CYP1 family, which may account for differential substrate specificity. Six putative substrate recognition sequences are characterized along with the regions they form in the protein structure. Further the RMSD and B-factor analysis provides the information about the identified residues having the maximum RMSD and B-factor deviations.


Asunto(s)
Sistema Enzimático del Citocromo P-450/química , Proteómica/métodos , Alineación de Secuencia/métodos , Especificidad por Sustrato/fisiología , Secuencia de Aminoácidos , Dominio Catalítico , Análisis de Secuencia de Proteína
13.
Mol Divers ; 18(4): 895-909, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25213397

RESUMEN

The translational failure between preclinical animal models and clinical outcome has alarmed us to search for a new strategy in the treatment of Alzheimer's disease (AD). Interlink between Pregnane X Receptor (PXR) and P-glycoprotein (Pgp) at the blood brain barrier (BBB) has raised hope toward a new disease modifying therapy in AD. Pgp is a major efflux transporter for beta amyloid (Aß) at human BBB. A literature survey reveals diminished expression of Pgp transporter at the BBB in AD patients. Pregnane X Receptor is a major transcriptional regulator of Pgp. Restoration of Pgp at the BBB enhances the elimination of the Aß from brain alongside and inhibits the apical to basolateral movement of Aß from the circulatory blood. This review concentrates on in vitro, in vivo, and in silico advancements on the study of the PXR in context to Pgp and discusses the substrate and inhibitor specificity between PXR and Pgp.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Enfermedad de Alzheimer/metabolismo , Receptores de Esteroides/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/etiología , Animales , Manejo de la Enfermedad , Humanos , Inflamación/etiología , Inflamación/metabolismo , Modelos Moleculares , Terapia Molecular Dirigida , Receptor X de Pregnano , Unión Proteica , Conformación Proteica , Receptores de Esteroides/química , Receptores de Esteroides/genética
14.
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
15.
Mol Divers ; 18(4): 865-78, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25028215

RESUMEN

Recently, CYP1 enzymes are documented for selective metabolism of anticancer leads in cancer prevention and/or progression. Elucidation of specificity of substrates/inhibitors of CYP1 isoforms plays a vital role in design of more selective and potent anticancer leads. However, an area of concern is the broad range of substrate specificities and planar nature of substrates with limited dataset which makes it difficult to predict their site of metabolism (SOM) accurately. In the present study, various models for prediction of site of metabolism in case of CYP1A1, CYP1A2, and CYP1B1 substrates were developed using MetaSite, molecular docking, and quantum chemical descriptors. The predictive accuracy of MetaSite, molecular docking, and quantum chemical descriptors in identifying experimental site of metabolism was analyzed at three levels; top rank, top three ranks, and top five ranks. Two quantum chemical descriptors, chemical hardness and local nucleophilicity are proposed for the prediction of CYP-mediated SOM for the first time. The predictive accuracy shown by chemical hardness at top three ranks was 83.3, 85.7, and 84.6 % for CYP1A1, CYP1A2 and CYP1B1, respectively, whereas local nucleophilicity gave poor predictions of 50, 42.8, and 46.2 %, respectively. The predictability of chemical hardness descriptor outperformed at all three levels of ranks for CYP1A1, CYP1A2, and CYP1B1. Hence, we propose chemical hardness as an useful quantum chemical descriptor for prediction of metabolically vulnerable prints in CYP1A1, CYP1A2, and CYP1B1 mediated metabolism and support the optimization efforts in drug discovery and development programs.


Asunto(s)
Citocromo P-450 CYP1A1/química , Citocromo P-450 CYP1A2/química , Citocromo P-450 CYP1B1/química , Modelos Químicos , Modelos Moleculares , Sitios de Unión , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Teoría Cuántica , Reproducibilidad de los Resultados , Especificidad por Sustrato
16.
J Mass Spectrom ; 49(5): 380-91, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24809899

RESUMEN

Ketorolac, a nonsteroidal anti-inflammatory drug, was subjected to forced degradation studies as per International Conference on Harmonization guidelines. A simple, rapid, precise, and accurate high-performance liquid chromatography combined with electrospray ionization quadrupole time-of-flight tandem mass spectrometry (LC/ESI/Q/TOF/MS/MS) method has been developed for the identification and structural characterization of stressed degradation products of ketorolac. The drug was found to degrade in hydrolytic (acidic, basic, and neutral), photolytic (acidic, basic, and neutral solution), and thermal conditions, whereas the solid form of the drug was found to be stable under photolytic conditions. The method has shown adequate separation of ketorolac tromethamine and its degradation products on a Grace Smart C-18 (250 mm × 4.6 mm i.d., 5 µm) column using 20 mM ammonium formate (pH = 3.2): acetonitrile as a mobile phase in gradient elution mode at a flow rate of 1.0 ml/min. A total of nine degradation products were identified and characterized by LC/ESI/MS/MS. The most probable mechanisms for the formation of degradation products have been proposed on the basis of a comparison of the fragmentation of the [M + H](+) ions of ketorolac and its degradation products. In silico toxicity of the drug and degradation products was investigated by using topkat and derek softwares. The method was validated in terms of specificity, linearity, accuracy, precision, and robustness as per International Conference on Harmonization guidelines.


Asunto(s)
Cromatografía Liquida/métodos , Simulación por Computador , Ketorolaco Trometamina/química , Ketorolaco Trometamina/toxicidad , Espectrometría de Masas en Tándem/métodos , Animales , Femenino , Masculino , Ratones , Oxidación-Reducción , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Pruebas de Toxicidad
17.
Gene ; 539(1): 82-90, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24491504

RESUMEN

Epidermal growth factor receptor tyrosine kinase (EGFR-TK) is an attractive target for cancer therapy. Despite a number of effective EGFR inhibitors that are constantly expanding and different methods being employed to obtain novel compounds, the search for newer EGFR inhibitors is still a major scientific challenge. In the present study, a molecular docking and molecular dynamics investigation has been carried out with an ensemble of EGFR-TK structures against a synthetically feasible library of curcumin analogs to discover potent EGFR inhibitors. To resolve protein flexibility issue we have utilized 5 EGFR wild type crystal structures during docking as this gives improved possibility of identifying an active compound as compared to using a single crystal structure. We then identified five curcumin analogs representing different scaffolds that can serve as lead molecules. Finally, the 5 ns molecular dynamics simulation shows that knoevenagel condensate of curcumin specifically C29 and C30 can be used as starting blocks for developing effective leads capable of inhibiting EGFR.


Asunto(s)
Curcumina/análogos & derivados , Curcumina/metabolismo , Receptores ErbB/antagonistas & inhibidores , Simulación del Acoplamiento Molecular/métodos , Simulación de Dinámica Molecular , Antineoplásicos/farmacología , Dominio Catalítico/genética , Cristalografía por Rayos X , Descubrimiento de Drogas , Receptores ErbB/ultraestructura , Humanos , Neoplasias/tratamiento farmacológico , Relación Estructura-Actividad
18.
J Mol Recognit ; 27(3): 138-50, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24446378

RESUMEN

The discovery of novel anticancer molecules 5F-203 (NSC703786) and 5-aminoflavone (5-AMF, NSC686288) has addressed the issues of toxicity and reduced efficacy by targeting over expressed Cytochrome P450 1A1 (CYP1A1) in cancer cells. CYP1A1 metabolizes these compounds into their reactive metabolites, which are proven to mediate their anticancer effect through DNA adduct formation. However, the drug metabolite-DNA binding has not been explored so far. Hence, understanding the binding characteristics and molecular recognition for drug metabolites with DNA is of practical and fundamental interest. The present study is aimed to model binding preference shown by reactive metabolites of 5F-203 and 5-AMF with DNA in forming DNA adducts. To perform this, three different DNA crystal structures covering sequence diversity were selected, and 12 DNA-reactive metabolite complexes were generated. Molecular dynamics simulations for all complexes were performed using AMBER 11 software after development of protocol for DNA-reactive metabolite system. Furthermore, the MM-PBSA/GBSA energy calculation, per-nucleotide energy decomposition, and Molecular Electrostatic Surface Potential analysis were performed. The results obtained from present study clearly indicate that minor groove in DNA is preferable for binding of reactive metabolites of anticancer compounds. The binding preferences shown by reactive metabolites were also governed by specific nucleotide sequence and distribution of electrostatic charges in major and minor groove of DNA structure. Overall, our study provides useful insights into the initial step of mechanism of reactive metabolite binding to the DNA and the guidelines for designing of sequence specific DNA interacting anticancer agents.


Asunto(s)
Antineoplásicos/química , Citocromo P-450 CYP1A1/metabolismo , ADN/química , Flavonoides/química , Sustancias Intercalantes/química , Tiazoles/química , Antineoplásicos/metabolismo , Sitios de Unión , Biotransformación , Línea Celular Tumoral , Cristalografía por Rayos X , Aductos de ADN/química , Flavonoides/metabolismo , Humanos , Sustancias Intercalantes/metabolismo , Cinética , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Conformación de Ácido Nucleico , Electricidad Estática , Termodinámica , Tiazoles/metabolismo
19.
Bioorg Med Chem Lett ; 22(24): 7566-72, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23122866

RESUMEN

The DNA molecule is a target for plethora of anticancer and antiviral drugs that forms covalent and non-covalent adducts with major or minor groove of DNA. In present study we synthesized series of novel Pyrazolo [1,5-a]pyrimidine derivatives. The newly synthesized compounds were characterized by elemental analysis, IR, (1)H NMR, and mass spectral data. The selected compounds were studied for interaction with Calf thymus DNA (CT-DNA) using electronic spectra, viscosity measurement and thermal denaturation studies. Further, molecular interactions were revealed for compound IIIa and IVa by computational methodologies. The preferred mode of ligand binding with double helical DNA as well as preferable DNA groove were explored by molecular docking in different DNA models.


Asunto(s)
ADN/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Animales , Sitios de Unión/efectos de los fármacos , Bovinos , Ligandos , Modelos Moleculares , Estructura Molecular , Pirazoles/síntesis química , Pirazoles/química , Pirimidinas/síntesis química , Pirimidinas/química
20.
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