Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
J Comput Aided Mol Des ; 30(9): 829-839, 2016 09.
Article in English | MEDLINE | ID: mdl-27699554

ABSTRACT

The D3R Grand Challenge 2015 was focused on two protein targets: Heat Shock Protein 90 (HSP90) and Mitogen-Activated Protein Kinase Kinase Kinase Kinase 4 (MAP4K4). We used a protocol involving a preliminary analysis of the available data in PDB and PubChem BioAssay, and then a docking/scoring step using more computationally demanding parameters that were required to provide more reliable predictions. We could evidence that different docking software and scoring functions can behave differently on individual ligand datasets, and that the flexibility of specific binding site residues is a crucial element to provide good predictions.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Molecular Docking Simulation/methods , Protein Conformation , Protein Serine-Threonine Kinases/chemistry , Algorithms , Binding Sites , Databases, Protein , Drug Design , Humans , Ligands , Protein Binding , Structure-Activity Relationship
2.
J Chem Inf Model ; 56(6): 996-1003, 2016 06 27.
Article in English | MEDLINE | ID: mdl-26391724

ABSTRACT

The 2014 CSAR Benchmark Exercise was focused on three protein targets: coagulation factor Xa, spleen tyrosine kinase, and bacterial tRNA methyltransferase. Our protocol involved a preliminary analysis of the structural information available in the Protein Data Bank for the protein targets, which allowed the identification of the most appropriate docking software and scoring functions to be used for the rescoring of several docking conformations datasets, as well as for pose prediction and affinity ranking. The two key points of this study were (i) the prior evaluation of molecular modeling tools that are most adapted for each target and (ii) the increased search efficiency during the docking process to better explore the conformational space of big and flexible ligands.


Subject(s)
Molecular Docking Simulation , Proteins/chemistry , Proteins/metabolism , Benchmarking , Databases, Protein , Factor Xa/chemistry , Factor Xa/metabolism , Protein Conformation , Syk Kinase/chemistry , Syk Kinase/metabolism , tRNA Methyltransferases/chemistry , tRNA Methyltransferases/metabolism
3.
Bioinformatics ; 31(24): 3930-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26315915

ABSTRACT

MOTIVATION: Cytochrome P450 (CYP) is a superfamily of enzymes responsible for the metabolism of drugs, xenobiotics and endogenous compounds. CYP2D6 metabolizes about 30% of drugs and predicting potential CYP2D6 inhibition is important in early-stage drug discovery. RESULTS: We developed an original in silico approach for the prediction of CYP2D6 inhibition combining the knowledge of the protein structure and its dynamic behavior in response to the binding of various ligands and machine learning modeling. This approach includes structural information for CYP2D6 based on the available crystal structures and molecular dynamic simulations (MD) that we performed to take into account conformational changes of the binding site. We performed modeling using three learning algorithms--support vector machine, RandomForest and NaiveBayesian--and we constructed combined models based on topological information of known CYP2D6 inhibitors and predicted binding energies computed by docking on both X-ray and MD protein conformations. In addition, we identified three MD-derived structures that are capable all together to better discriminate inhibitors and non-inhibitors compared with individual CYP2D6 conformations, thus ensuring complementary ligand profiles. Inhibition models based on classical molecular descriptors and predicted binding energies were able to predict CYP2D6 inhibition with an accuracy of 78% on the training set and 75% on the external validation set.


Subject(s)
Cytochrome P-450 CYP2D6 Inhibitors/chemistry , Cytochrome P-450 CYP2D6/chemistry , Molecular Dynamics Simulation , Algorithms , Binding Sites , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP2D6/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Humans , Ligands , Machine Learning , Protein Conformation
4.
PLoS One ; 8(9): e73587, 2013.
Article in English | MEDLINE | ID: mdl-24039991

ABSTRACT

Drug metabolizing enzymes play a key role in the metabolism, elimination and detoxification of xenobiotics, drugs and endogenous molecules. While their principal role is to detoxify organisms by modifying compounds, such as pollutants or drugs, for a rapid excretion, in some cases they render their substrates more toxic thereby inducing severe side effects and adverse drug reactions, or their inhibition can lead to drug-drug interactions. We focus on sulfotransferases (SULTs), a family of phase II metabolizing enzymes, acting on a large number of drugs and hormones and showing important structural flexibility. Here we report a novel in silico structure-based approach to probe ligand binding to SULTs. We explored the flexibility of SULTs by molecular dynamics (MD) simulations in order to identify the most suitable multiple receptor conformations for ligand binding prediction. Then, we employed structure-based docking-scoring approach to predict ligand binding and finally we combined the predicted interaction energies by using a QSAR methodology. The results showed that our protocol successfully prioritizes potent binders for the studied here SULT1 isoforms, and give new insights on specific molecular mechanisms for diverse ligands' binding related to their binding sites plasticity. Our best QSAR models, introducing predicted protein-ligand interaction energy by using docking, showed accuracy of 67.28%, 78.00% and 75.46%, for the isoforms SULT1A1, SULT1A3 and SULT1E1, respectively. To the best of our knowledge our protocol is the first in silico structure-based approach consisting of a protein-ligand interaction analysis at atomic level that considers both ligand and enzyme flexibility, along with a QSAR approach, to identify small molecules that can interact with II phase dug metabolizing enzymes.


Subject(s)
Protein Structure, Tertiary , Small Molecule Libraries/chemistry , Sulfotransferases/chemistry , Xenobiotics/chemistry , Arylsulfotransferase/chemistry , Arylsulfotransferase/metabolism , Binding Sites , Computer Simulation , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Quantitative Structure-Activity Relationship , Small Molecule Libraries/metabolism , Sulfotransferases/metabolism , Thermodynamics , Xenobiotics/metabolism
5.
J Mol Biol ; 425(21): 3978-92, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23856621

ABSTRACT

Cytochrome P450 (CYP) is a supergene family of metabolizing enzymes involved in the phase I metabolism of drugs and endogenous compounds. CYP oxidation often leads to inactive drug metabolites or to highly toxic or carcinogenic metabolites involved in adverse drug reactions (ADR). During the last decade, the impact of CYP polymorphism in various drug responses and ADR has been demonstrated. Of the drugs involved in ADR, 56% are metabolized by polymorphic phase I metabolizing enzymes, 86% among them being CYP. Here, we review the major CYP polymorphic forms, their impact for drug response and current advances in molecular modeling of CYP polymorphism. We focus on recent studies exploring CYP polymorphism performed by the use of sequence-based and/or protein-structure-based computational approaches. The importance of understanding the molecular mechanisms related to CYP polymorphism and drug response at the atomic level is outlined.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Drug-Related Side Effects and Adverse Reactions , Humans , Metabolic Detoxication, Phase I , Models, Molecular , Polymorphism, Genetic
6.
Drug Discov Today ; 17(1-2): 44-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22056716

ABSTRACT

Quantitative structure-activity relationship (QSAR) methods and related approaches have been used to investigate the molecular features that influence the absorption, distribution, metabolism, excretion and toxicity (ADMET) of drugs. As the three-dimensional structures of several major ADMET proteins become available, structure-based (docking-scoring) computations can be carried out to complement or to go beyond QSAR studies. Applying docking-scoring methods to ADMET proteins is a challenging process because they usually have a large and flexible binding cavity; however, promising results relating to metabolizing enzymes have been reported. After reviewing current trends in the field we applied structure-based methods in the context of receptor flexibility in a case study involving the phase II metabolizing sulfotransferases. Overall, the explored concepts and results suggested that structure-based ADMET profiling will probably join the mainstream during the coming years.


Subject(s)
Drug Discovery/methods , Pharmaceutical Preparations/chemistry , Absorption , Animals , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmacokinetics , Pharmacology , Quantitative Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...