Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 42(Web Server issue): W32-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24792161

ABSTRACT

Bioactive small molecules, such as drugs or metabolites, bind to proteins or other macro-molecular targets to modulate their activity, which in turn results in the observed phenotypic effects. For this reason, mapping the targets of bioactive small molecules is a key step toward unraveling the molecular mechanisms underlying their bioactivity and predicting potential side effects or cross-reactivity. Recently, large datasets of protein-small molecule interactions have become available, providing a unique source of information for the development of knowledge-based approaches to computationally identify new targets for uncharacterized molecules or secondary targets for known molecules. Here, we introduce SwissTargetPrediction, a web server to accurately predict the targets of bioactive molecules based on a combination of 2D and 3D similarity measures with known ligands. Predictions can be carried out in five different organisms, and mapping predictions by homology within and between different species is enabled for close paralogs and orthologs. SwissTargetPrediction is accessible free of charge and without login requirement at http://www.swisstargetprediction.ch.


Subject(s)
Drug Discovery , Proteins/chemistry , Software , Algorithms , Animals , Cattle , Humans , Internet , Ligands , Mice , Pharmaceutical Preparations/chemistry , Proteins/drug effects , Rats , Sequence Homology, Amino Acid , User-Computer Interface
2.
Nucleic Acids Res ; 40(Web Server issue): W597-603, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22661580

ABSTRACT

ExPASy (http://www.expasy.org) has worldwide reputation as one of the main bioinformatics resources for proteomics. It has now evolved, becoming an extensible and integrative portal accessing many scientific resources, databases and software tools in different areas of life sciences. Scientists can henceforth access seamlessly a wide range of resources in many different domains, such as proteomics, genomics, phylogeny/evolution, systems biology, population genetics, transcriptomics, etc. The individual resources (databases, web-based and downloadable software tools) are hosted in a 'decentralized' way by different groups of the SIB Swiss Institute of Bioinformatics and partner institutions. Specifically, a single web portal provides a common entry point to a wide range of resources developed and operated by different SIB groups and external institutions. The portal features a search function across 'selected' resources. Additionally, the availability and usage of resources are monitored. The portal is aimed for both expert users and people who are not familiar with a specific domain in life sciences. The new web interface provides, in particular, visual guidance for newcomers to ExPASy.


Subject(s)
Computational Biology , Proteomics , Software , Computer Graphics , Genomics , Internet , Systems Integration , User-Computer Interface
3.
J Med Chem ; 55(11): 5270-90, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22616902

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is an important therapeutic target for the treatment of diseases such as cancer that involve pathological immune escape. Starting from the scaffold of our previously discovered IDO1 inhibitor 4-phenyl-1,2,3-triazole, we used computational structure-based methods to design more potent ligands. This approach yielded highly efficient low molecular weight inhibitors, the most active being of nanomolar potency both in an enzymatic and in a cellular assay, while showing no cellular toxicity and a high selectivity for IDO1 over tryptophan 2,3-dioxygenase (TDO). A quantitative structure-activity relationship based on the electrostatic ligand-protein interactions in the docked binding modes and on the quantum chemically derived charges of the triazole ring demonstrated a good explanatory power for the observed activities.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Triazoles/chemical synthesis , Animals , Catalytic Domain , Cell Line , Drug Design , Enzyme Assays , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Ligands , Mice , Models, Molecular , Protein Binding , Protein Conformation , Quantitative Structure-Activity Relationship , Quantum Theory , Static Electricity , Triazoles/chemistry , Triazoles/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors
4.
Eur J Med Chem ; 46(12): 6098-103, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22000921

ABSTRACT

Human inhibitor NF-κB kinase 2 (hIKK-2) is the primary component responsible for activating NF-κB in response to various inflammatory stimuli. Thus, synthetic ATP-competitive inhibitors for hIKK-2 have been developed as anti-inflammatory compounds. We recently reported a virtual screening protocol (doi:10.1371/journal.pone.0016903) that is able to identify hIKK-2 inhibitors that are not structurally related to any known molecule that inhibits hIKK-2 and that have never been reported to have anti-inflammatory activity. In this study, a stricter version of this protocol was applied to an in-house database of 29,779 natural products annotated with their natural source. The search identified 274 molecules (isolated from 453 different natural extracts) predicted to inhibit hIKK-2. An exhaustive bibliographic search revealed that anti-inflammatory activity has been previously described for: (a) 36 out of these 453 extracts; and (b) 17 out of 30 virtual screening hits present in these 36 extracts. Only one of the remaining 13 hit molecules in these extracts shows chemical similarity with known synthetic hIKK-2 inhibitors. Therefore, it is plausible that a significant portion of the remaining 12 hit molecules are lead-hopping candidates for the development of new hIKK-2 inhibitors.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Discovery/methods , I-kappa B Kinase/antagonists & inhibitors , Plant Extracts/pharmacology , Protein Kinase Inhibitors/pharmacology , Anti-Inflammatory Agents/chemistry , Biological Products/chemistry , Biological Products/pharmacology , Databases, Factual , Humans , I-kappa B Kinase/metabolism , Plant Extracts/chemistry , Plants, Medicinal/chemistry , Protein Kinase Inhibitors/chemistry , Workflow
5.
Nucleic Acids Res ; 39(Web Server issue): W270-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21624888

ABSTRACT

Most life science processes involve, at the atomic scale, recognition between two molecules. The prediction of such interactions at the molecular level, by so-called docking software, is a non-trivial task. Docking programs have a wide range of applications ranging from protein engineering to drug design. This article presents SwissDock, a web server dedicated to the docking of small molecules on target proteins. It is based on the EADock DSS engine, combined with setup scripts for curating common problems and for preparing both the target protein and the ligand input files. An efficient Ajax/HTML interface was designed and implemented so that scientists can easily submit dockings and retrieve the predicted complexes. For automated docking tasks, a programmatic SOAP interface has been set up and template programs can be downloaded in Perl, Python and PHP. The web site also provides an access to a database of manually curated complexes, based on the Ligand Protein Database. A wiki and a forum are available to the community to promote interactions between users. The SwissDock web site is available online at http://www.swissdock.ch. We believe it constitutes a step toward generalizing the use of docking tools beyond the traditional molecular modeling community.


Subject(s)
Ligands , Protein Conformation , Software , Drug Design , Internet , Models, Molecular , Protein Engineering
6.
J Comput Chem ; 32(10): 2149-59, 2011 Jul 30.
Article in English | MEDLINE | ID: mdl-21541955

ABSTRACT

The prediction of binding modes (BMs) occurring between a small molecule and a target protein of biological interest has become of great importance for drug development. The overwhelming diversity of needs leaves room for docking approaches addressing specific problems. Nowadays, the universe of docking software ranges from fast and user friendly programs to algorithmically flexible and accurate approaches. EADock2 is an example of the latter. Its multiobjective scoring function was designed around the CHARMM22 force field and the FACTS solvation model. However, the major drawback of such a software design lies in its computational cost. EADock dihedral space sampling (DSS) is built on the most efficient features of EADock2, namely its hybrid sampling engine and multiobjective scoring function. Its performance is equivalent to that of EADock2 for drug-like ligands, while the CPU time required has been reduced by several orders of magnitude. This huge improvement was achieved through a combination of several innovative features including an automatic bias of the sampling toward putative binding sites, and a very efficient tree-based DSS algorithm. When the top-scoring prediction is considered, 57% of BMs of a test set of 251 complexes were reproduced within 2 Å RMSD to the crystal structure. Up to 70% were reproduced when considering the five top scoring predictions. The success rate is lower in cross-docking assays but remains comparable with that of the latest version of AutoDock that accounts for the protein flexibility.


Subject(s)
Algorithms , Ligands , Molecular Docking Simulation/methods , Proteins/chemistry , Software
7.
J Comput Chem ; 32(11): 2359-68, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21541964

ABSTRACT

The drug discovery process has been deeply transformed recently by the use of computational ligand-based or structure-based methods, helping the lead compounds identification and optimization, and finally the delivery of new drug candidates more quickly and at lower cost. Structure-based computational methods for drug discovery mainly involve ligand-protein docking and rapid binding free energy estimation, both of which require force field parameterization for many drug candidates. Here, we present a fast force field generation tool, called SwissParam, able to generate, for arbitrary small organic molecule, topologies, and parameters based on the Merck molecular force field, but in a functional form that is compatible with the CHARMM force field. Output files can be used with CHARMM or GROMACS. The topologies and parameters generated by SwissParam are used by the docking software EADock2 and EADock DSS to describe the small molecules to be docked, whereas the protein is described by the CHARMM force field, and allow them to reach success rates ranging from 56 to 78%. We have also developed a rapid binding free energy estimation approach, using SwissParam for ligands and CHARMM22/27 for proteins, which requires only a short minimization to reproduce the experimental binding free energy of 214 ligand-protein complexes involving 62 different proteins, with a standard error of 2.0 kcal mol(-1), and a correlation coefficient of 0.74. Together, these results demonstrate the relevance of using SwissParam topologies and parameters to describe small organic molecules in computer-aided drug design applications, together with a CHARMM22/27 description of the target protein. SwissParam is available free of charge for academic users at www.swissparam.ch.


Subject(s)
Computer Simulation , Organic Chemicals , Proteins/metabolism , Small Molecule Libraries , Drug Discovery , Protein Binding , Proteins/chemistry , Thermodynamics
8.
PLoS One ; 6(2): e16903, 2011 Feb 24.
Article in English | MEDLINE | ID: mdl-21390216

ABSTRACT

BACKGROUND: Their large scaffold diversity and properties, such as structural complexity and drug similarity, form the basis of claims that natural products are ideal starting points for drug design and development. Consequently, there has been great interest in determining whether such molecules show biological activity toward protein targets of pharmacological relevance. One target of particular interest is hIKK-2, a serine-threonine protein kinase belonging to the IKK complex that is the primary component responsible for activating NF-κB in response to various inflammatory stimuli. Indeed, this has led to the development of synthetic ATP-competitive inhibitors for hIKK-2. Therefore, the main goals of this study were (a) to use virtual screening to identify potential hIKK-2 inhibitors of natural origin that compete with ATP and (b) to evaluate the reliability of our virtual-screening protocol by experimentally testing the in vitro activity of selected natural-product hits. METHODOLOGY/PRINCIPAL FINDINGS: We thus predicted that 1,061 out of the 89,425 natural products present in the studied database would inhibit hIKK-2 with good ADMET properties. Notably, when these 1,061 molecules were merged with the 98 synthetic hIKK-2 inhibitors used in this study and the resulting set was classified into ten clusters according to chemical similarity, there were three clusters that contained only natural products. Five molecules from these three clusters (for which no anti-inflammatory activity has been previously described) were then selected for in vitro activity testing, in which three out of the five molecules were shown to inhibit hIKK-2. CONCLUSIONS/SIGNIFICANCE: We demonstrated that our virtual-screening protocol was successful in identifying lead compounds for developing new inhibitors for hIKK-2, a target of great interest in medicinal chemistry. Additionally, all the tools developed during the current study (i.e., the homology model for the hIKK-2 kinase domain and the pharmacophore) will be made available to interested readers upon request.


Subject(s)
Enzyme Assays/methods , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays/methods , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/chemistry , User-Computer Interface , Amino Acid Sequence , Biological Products/analysis , Biological Products/isolation & purification , Biological Products/pharmacology , Catalytic Domain/physiology , Computational Biology/methods , Enzyme Inhibitors/pharmacology , Humans , I-kappa B Kinase/metabolism , Libraries, Digital , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Protein Structure, Tertiary/physiology , Reproducibility of Results , Sequence Homology
9.
J Mol Recognit ; 23(5): 457-61, 2010.
Article in English | MEDLINE | ID: mdl-20101644

ABSTRACT

Protein-ligand docking has made important progress during the last decade and has become a powerful tool for drug development, opening the way to virtual high throughput screening and in silico structure-based ligand design. Despite the flattering picture that has been drawn, recent publications have shown that the docking problem is far from being solved, and that more developments are still needed to achieve high successful prediction rates and accuracy. Introducing an accurate description of the solvation effect upon binding is thought to be essential to achieve this goal. In particular, EADock uses the Generalized Born Molecular Volume 2 (GBMV2) solvent model, which has been shown to reproduce accurately the desolvation energies calculated by solving the Poisson equation. Here, the implementation of the Fast Analytical Continuum Treatment of Solvation (FACTS) as an implicit solvation model in small molecules docking calculations has been assessed using the EADock docking program. Our results strongly support the use of FACTS for docking. The success rates of EADock/FACTS and EADock/GBMV2 are similar, i.e. around 75% for local docking and 65% for blind docking. However, these results come at a much lower computational cost: FACTS is 10 times faster than GBMV2 in calculating the total electrostatic energy, and allows a speed up of EADock by a factor of 4. This study also supports the EADock development strategy relying on the CHARMM package for energy calculations, which enables straightforward implementation and testing of the latest developments in the field of Molecular Modeling.


Subject(s)
Computer Simulation , Proteins/chemistry , Algorithms , Drug Discovery , Ligands , Models, Molecular , Protein Binding , Proteins/metabolism , Solvents/chemistry , Static Electricity , Thermodynamics
10.
J Med Chem ; 53(3): 1172-89, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20055453

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is an important therapeutic target for the treatment of diseases such as cancer that involve pathological immune escape. We have used the evolutionary docking algorithm EADock to design new inhibitors of this enzyme. First, we investigated the modes of binding of all known IDO inhibitors. On the basis of the observed docked conformations, we developed a pharmacophore model, which was then used to devise new compounds to be tested for IDO inhibition. We also used a fragment-based approach to design and to optimize small organic molecule inhibitors. Both approaches yielded several new low-molecular weight inhibitor scaffolds, the most active being of nanomolar potency in an enzymatic assay. Cellular assays confirmed the potential biological relevance of four different scaffolds.


Subject(s)
Cell Proliferation/drug effects , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Animals , Cells, Cultured , Enzyme Inhibitors/chemical synthesis , Humans , Kynurenine/metabolism , Mice , Models, Molecular , Small Molecule Libraries , Structure-Activity Relationship , Tryptophan/metabolism
11.
J Comput Chem ; 30(14): 2305-15, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19288474

ABSTRACT

In silico screening has become a valuable tool in drug design, but some drug targets represent real challenges for docking algorithms. This is especially true for metalloproteins, whose interactions with ligands are difficult to parametrize. Our docking algorithm, EADock, is based on the CHARMM force field, which assures a physically sound scoring function and a good transferability to a wide range of systems, but also exhibits difficulties in case of some metalloproteins. Here, we consider the therapeutically important case of heme proteins featuring an iron core at the active site. Using a standard docking protocol, where the iron-ligand interaction is underestimated, we obtained a success rate of 28% for a test set of 50 heme-containing complexes with iron-ligand contact. By introducing Morse-like metal binding potentials (MMBP), which are fitted to reproduce density functional theory calculations, we are able to increase the success rate to 62%. The remaining failures are mainly due to specific ligand-water interactions in the X-ray structures. Testing of the MMBP on a second data set of non iron binders (14 cases) demonstrates that they do not introduce a spurious bias towards metal binding, which suggests that they may reliably be used also for cross-docking studies.


Subject(s)
Computer Simulation , Hemeproteins/chemistry , Models, Chemical , Algorithms , Catalytic Domain , Databases, Factual , Ligands , Quantum Theory
12.
J Cell Mol Med ; 13(2): 238-48, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19183238

ABSTRACT

The drug discovery process has been profoundly changed recently by the adoption of computational methods helping the design of new drug candidates more rapidly and at lower costs. In silico drug design consists of a collection of tools helping to make rational decisions at the different steps of the drug discovery process, such as the identification of a biomolecular target of therapeutical interest, the selection or the design of new lead compounds and their modification to obtain better affinities, as well as pharmacokinetic and pharmacodynamic properties. Among the different tools available, a particular emphasis is placed in this review on molecular docking, virtual high-throughput screening and fragment-based ligand design.


Subject(s)
Computational Biology/methods , Drug Design , Drug Evaluation, Preclinical/methods , Ligands , Algorithms , Humans , Models, Molecular , Molecular Structure , Protein Conformation , Structure-Activity Relationship
13.
J Comput Chem ; 30(13): 2021-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19130502

ABSTRACT

Molecular docking softwares are one of the important tools of modern drug development pipelines. The promising achievements of the last 10 years emphasize the need for further improvement, as reflected by several recent publications (Leach et al., J Med Chem 2006, 49, 5851; Warren et al., J Med Chem 2006, 49, 5912). Our initial approach, EADock, showed a good performance in reproducing the experimental binding modes for a set of 37 different ligand-protein complexes (Grosdidier et al., Proteins 2007, 67, 1010). This article presents recent improvements regarding the scoring and sampling aspects over the initial implementation, as well as a new seeding procedure based on the detection of cavities, opening the door to blind docking with EADock. These enhancements were validated on 260 complexes taken from the high quality Ligand Protein Database [LPDB, (Roche et al., J Med Chem 2001, 44, 3592)]. Two issues were identified: first, the quality of the initial structures cannot be assumed and a manual inspection and/or a search in the literature are likely to be required to achieve the best performance. Second the description of interactions involving metal ions still has to be improved. Nonetheless, a remarkable success rate of 65% was achieved for a large scale blind docking assay, when considering only the top ranked binding mode and a success threshold of 2 A RMSD to the crystal structure. When looking at the five-top ranked binding modes, the success rate increases up to 76%. In a standard local docking assay, success rates of 75 and 83% were obtained, considering only the top ranked binding mode, or the five top binding modes, respectively.


Subject(s)
Algorithms , Drug Design , Proteins/chemistry , Software , Ligands , Protein Binding , Proteins/metabolism
14.
PLoS One ; 3(7): e2645, 2008 Jul 09.
Article in English | MEDLINE | ID: mdl-18612410

ABSTRACT

Homology modeling is the most commonly used technique to build a three-dimensional model for a protein sequence. It heavily relies on the quality of the sequence alignment between the protein to model and related proteins with a known three dimensional structure. Alignment quality can be assessed according to the physico-chemical properties of the three dimensional models it produces. In this work, we introduce fifteen predictors designed to evaluate the properties of the models obtained for various alignments. They consist of an energy value obtained from different force fields (CHARMM, ProsaII or ANOLEA) computed on residue selected around misaligned regions. These predictors were evaluated on ten challenging test cases. For each target, all possible ungapped alignments are generated and their corresponding models are computed and evaluated. The best predictor, retrieving the structural alignment for 9 out of 10 test cases, is based on the ANOLEA atomistic mean force potential and takes into account residues around misaligned secondary structure elements. The performance of the other predictors is significantly lower. This work shows that substantial improvement in local alignments can be obtained by careful assessment of the local structure of the resulting models.


Subject(s)
Structural Homology, Protein , Algorithms , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Proteins/chemistry , Sequence Alignment
15.
Biopharm Drug Dispos ; 29(2): 103-18, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18188833

ABSTRACT

The main objective of the study was to examine the biotransformation of the anticancer drug imatinib in target cells by incubating it with oxidoreductases expressed in tumor cells. The second objective was to obtain an in silico prediction of the potential activity of imatinib metabolites. An in vitro enzyme kinetic study was performed with cDNA expressed human oxidoreductases and LC-MS/MS analysis. The kinetic parameters (Km and Vmax) were determined for six metabolites. A molecular modeling approach was used to dock these metabolites to the target Abl or Bcr-Abl kinases. CYP3A4 isozyme showed the broadest metabolic capacity, whereas CYP1A1, CYP1B1 and FMO3 isozymes biotransformed imatinib with a high intrinsic clearance. The predicted binding modes for the metabolites to Abl were comparable to that of the parent drug, suggesting potential activity. These findings indicate that CYP1A1 and CYP1B1, which are known to be overexpressed in a wide range of tumors, are involved in the biotransformation of imatinib. They could play a role in imatinib disposition in the targeted stem, progenitor and differentiated cancer cells, with a possible contribution of the metabolites toward the activity of the drug.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Aryl Hydrocarbon Hydroxylases/physiology , Cytochrome P-450 CYP1A1/physiology , Neoplasms/metabolism , Piperazines/pharmacokinetics , Pyrimidines/pharmacokinetics , Benzamides , Biotransformation , Cytochrome P-450 CYP1B1 , Humans , Imatinib Mesylate , Kinetics , Mass Spectrometry , Proto-Oncogene Proteins c-abl/physiology
16.
J Biol Chem ; 282(26): 19152-66, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17468099

ABSTRACT

The ability of pollutants to affect human health is a major concern, justified by the wide demonstration that reproductive functions are altered by endocrine disrupting chemicals. The definition of endocrine disruption is today extended to broader endocrine regulations, and includes activation of metabolic sensors, such as the peroxisome proliferator-activated receptors (PPARs). Toxicology approaches have demonstrated that phthalate plasticizers can directly influence PPAR activity. What is now missing is a detailed molecular understanding of the fundamental basis of endocrine disrupting chemical interference with PPAR signaling. We thus performed structural and functional analyses that demonstrate how monoethyl-hexyl-phthalate (MEHP) directly activates PPARgamma and promotes adipogenesis, albeit to a lower extent than the full agonist rosiglitazone. Importantly, we demonstrate that MEHP induces a selective activation of different PPARgamma target genes. Chromatin immunoprecipitation and fluorescence microscopy in living cells reveal that this selective activity correlates with the recruitment of a specific subset of PPARgamma coregulators that includes Med1 and PGC-1alpha, but not p300 and SRC-1. These results highlight some key mechanisms in metabolic disruption but are also instrumental in the context of selective PPAR modulation, a promising field for new therapeutic development based on PPAR modulation.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Diethylhexyl Phthalate/analogs & derivatives , Environmental Pollutants/toxicity , PPAR gamma/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/physiology , Animals , Binding Sites , COS Cells , Chlorocebus aethiops , Diethylhexyl Phthalate/chemistry , Diethylhexyl Phthalate/metabolism , Diethylhexyl Phthalate/toxicity , Environmental Pollutants/chemistry , Environmental Pollutants/metabolism , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Mice , PPAR gamma/chemistry , PPAR gamma/genetics , Promoter Regions, Genetic/physiology , Protein Structure, Tertiary , RNA, Small Interfering/pharmacology , Rosiglitazone , Thiazolidinediones/chemistry , Thiazolidinediones/metabolism
17.
Proteins ; 67(4): 1010-25, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17380512

ABSTRACT

In recent years, protein-ligand docking has become a powerful tool for drug development. Although several approaches suitable for high throughput screening are available, there is a need for methods able to identify binding modes with high accuracy. This accuracy is essential to reliably compute the binding free energy of the ligand. Such methods are needed when the binding mode of lead compounds is not determined experimentally but is needed for structure-based lead optimization. We present here a new docking software, called EADock, that aims at this goal. It uses an hybrid evolutionary algorithm with two fitness functions, in combination with a sophisticated management of the diversity. EADock is interfaced with the CHARMM package for energy calculations and coordinate handling. A validation was carried out on 37 crystallized protein-ligand complexes featuring 11 different proteins. The search space was defined as a sphere of 15 A around the center of mass of the ligand position in the crystal structure, and on the contrary to other benchmarks, our algorithm was fed with optimized ligand positions up to 10 A root mean square deviation (RMSD) from the crystal structure, excluding the latter. This validation illustrates the efficiency of our sampling strategy, as correct binding modes, defined by a RMSD to the crystal structure lower than 2 A, were identified and ranked first for 68% of the complexes. The success rate increases to 78% when considering the five best ranked clusters, and 92% when all clusters present in the last generation are taken into account. Most failures could be explained by the presence of crystal contacts in the experimental structure. Finally, the ability of EADock to accurately predict binding modes on a real application was illustrated by the successful docking of the RGD cyclic pentapeptide on the alphaVbeta3 integrin, starting far away from the binding pocket.


Subject(s)
Computer Simulation , Evolution, Molecular , Proteins/chemistry , Proteins/metabolism , Algorithms , Binding Sites , Crystallography, X-Ray , Ligands , Models, Molecular , Probability , Protein Binding , Protein Structure, Tertiary , Proteins/genetics , Static Electricity
18.
Biochim Biophys Acta ; 1771(8): 915-25, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17317294

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) compose a family of nuclear receptors that mediate the effects of lipidic ligands at the transcriptional level. In this review, we highlight advances in the understanding of the PPAR ligand binding domain (LBD) structure at the atomic level. The overall structure of PPARs LBD is described, and important protein ligand interactions are presented. Structure-activity relationships between isotypes structures and ligand specificity are addressed. It is shown that the numerous experimental three-dimensional structures available, together with in silico simulations, help understanding the role played by the activating function-2 (AF-2) in PPARs activation and its underlying molecular mechanism. The relation between the PPARs constitutive activity and the intrinsic stability of the active conformation is discussed. Finally, the interactions of PPARs LBD with co-activators or co-repressors, as well as with the retinoid X receptor (RXR) are described and considered in relation to PPARs activation.


Subject(s)
Peroxisome Proliferator-Activated Receptors/chemistry , Peroxisome Proliferator-Activated Receptors/physiology , Animals , Humans , Ligands , Models, Molecular , PPAR alpha/chemistry , PPAR alpha/physiology , Peroxisome Proliferator-Activated Receptors/genetics , Protein Conformation , Substrate Specificity
19.
J Biol Chem ; 282(13): 9666-9677, 2007 Mar 30.
Article in English | MEDLINE | ID: mdl-17200111

ABSTRACT

The dynamic properties of helix 12 in the ligand binding domain of nuclear receptors are a major determinant of AF-2 domain activity. We investigated the molecular and structural basis of helix 12 mobility, as well as the involvement of individual residues with regard to peroxisome proliferator-activated receptor alpha (PPARalpha) constitutive and ligand-dependent transcriptional activity. Functional assays of the activity of PPARalpha helix 12 mutants were combined with free energy molecular dynamics simulations. The agreement between the results from these approaches allows us to make robust claims concerning the mechanisms that govern helix 12 functions. Our data support a model in which PPARalpha helix 12 transiently adopts a relatively stable active conformation even in the absence of a ligand. This conformation provides the interface for the recruitment of a coactivator and results in constitutive activity. The receptor agonists stabilize this conformation and increase PPARalpha transcription activation potential. Finally, we disclose important functions of residues in PPARalpha AF-2, which determine the positioning of helix 12 in the active conformation in the absence of a ligand. Substitution of these residues suppresses PPARalpha constitutive activity, without changing PPARalpha ligand-dependent activation potential.


Subject(s)
Computer Simulation , Models, Biological , Mutagenesis, Site-Directed , PPAR alpha/chemistry , PPAR alpha/genetics , Point Mutation , Amino Acid Sequence , Animals , Computational Biology , HeLa Cells , Humans , Ligands , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/physiology , PPAR alpha/physiology , Protein Structure, Secondary , Thermodynamics , Transcription, Genetic , Xenopus laevis
20.
J Biol Chem ; 279(37): 38895-902, 2004 Sep 10.
Article in English | MEDLINE | ID: mdl-15234969

ABSTRACT

Several members of the FXYD protein family are tissue-specific regulators of Na,K-ATPase that produce distinct effects on its apparent K(+) and Na(+) affinity. Little is known about the interaction sites between the Na,K-ATPase alpha subunit and FXYD proteins that mediate the efficient association and/or the functional effects of FXYD proteins. In this study, we have analyzed the role of the transmembrane segment TM9 of the Na,K-ATPase alpha subunit in the structural and functional interaction with FXYD2, FXYD4, and FXYD7. Mutational analysis combined with expression in Xenopus oocytes reveals that Phe(956), Glu(960), Leu(964), and Phe(967) in TM9 of the Na,K-ATPase alpha subunit represent one face interacting with the three FXYD proteins. Leu(964) and Phe(967) contribute to the efficient association of FXYD proteins with the Na,K-ATPase alpha subunit, whereas Phe(956) and Glu(960) are essential for the transmission of the functional effect of FXYD proteins on the apparent K(+) affinity of Na,K-ATPase. The relative contribution of Phe(956) and Glu(960) to the K(+) effect differs for different FXYD proteins, probably reflecting the intrinsic differences of FXYD proteins on the apparent K(+) affinity of Na,K-ATPase. In contrast to the effect on the apparent K(+) affinity, Phe(956) and Glu(960) are not involved in the effect of FXYD2 and FXYD4 on the apparent Na(+) affinity of Na,K-ATPase. The mutational analysis is in good agreement with a docking model of the Na,K-ATPase/FXYD7 complex, which also predicts the importance of Phe(956), Glu(960), Leu(964), and Phe(967) in subunit interaction. In conclusion, by using mutational analysis and modeling, we show that TM9 of the Na,K-ATPase alpha subunit exposes one face of the helix that interacts with FXYD proteins and contributes to the stable interaction with FXYD proteins, as well as mediating the effect of FXYD proteins on the apparent K(+) affinity of Na,K-ATPase.


Subject(s)
Sodium-Potassium-Exchanging ATPase/chemistry , Alanine/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Membrane/metabolism , DNA Mutational Analysis , Electrophysiology , Glutamine/chemistry , Leucine/chemistry , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Oocytes/metabolism , Phenylalanine/chemistry , Potassium/chemistry , Precipitin Tests , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Structure-Activity Relationship , Time Factors , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...