Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Ann Appl Stat ; 7(2): 989-1009, 2013.
Article in English | MEDLINE | ID: mdl-24052809

ABSTRACT

We develop a Bayesian model for the alignment of two point configurations under the full similarity transformations of rotation, translation and scaling. Other work in this area has concentrated on rigid body transformations, where scale information is preserved, motivated by problems involving molecular data; this is known as form analysis. We concentrate on a Bayesian formulation for statistical shape analysis. We generalize the model introduced by Green and Mardia for the pairwise alignment of two unlabeled configurations to full similarity transformations by introducing a scaling factor to the model. The generalization is not straight-forward, since the model needs to be reformulated to give good performance when scaling is included. We illustrate our method on the alignment of rat growth profiles and a novel application to the alignment of protein domains. Here, scaling is applied to secondary structure elements when comparing protein folds; additionally, we find that one global scaling factor is not in general sufficient to model these data and, hence, we develop a model in which multiple scale factors can be included to handle different scalings of shape components.

2.
J Phys Chem B ; 116(35): 10856-69, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22920218

ABSTRACT

We use molecular docking and free energy calculations to estimate the relative free energy of binding of six arylamide compounds designed to inhibit the hDM2-p53 interaction. We show that using docking methods to predict or rank the binding affinity of a series of arylamide inhibitors of the hDM2-p53 interaction is problematic. However, using free energy calculations, we show that we can achieve levels of accuracy that can guide the development of novel arylamide compounds. We perform alchemical free energy calculations using the Desmond molecular dynamics package with the same arylamide inhibitors of the hDM2-p53 system and illustrate the challenges of performing accurate free energy calculations for realistic systems. To our knowledge, these are the first calculations for inhibitors of the hDM2 system that employ a full treatment of statistical mechanics including explicit water representation and full protein flexibility. We show that mutating three functional groups in a single transformation can be more efficient than mutating the groups one by one if proper intermediates are used. We also show that Hamiltonian exchanges can improve the efficiency of the calculation compared to standard alchemical methods, with a novel use of the phase space overlap to monitor sampling extent. We show that, despite sampling limitations, this approach can achieve levels of accuracy sufficient to bias further inhibitor modification toward binding, and identifies antiparallel configurations as stable or more stable than the parallel configurations that are typically considered.


Subject(s)
Amides/chemistry , RNA-Binding Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Amides/metabolism , Humans , Molecular Docking Simulation , Protein Binding , RNA-Binding Proteins/metabolism , Thermodynamics , Tumor Suppressor Protein p53/metabolism
3.
PLoS One ; 7(8): e43253, 2012.
Article in English | MEDLINE | ID: mdl-22916232

ABSTRACT

The design of novel α-helix mimetic inhibitors of protein-protein interactions is of interest to pharmaceuticals and chemical genetics researchers as these inhibitors provide a chemical scaffold presenting side chains in the same geometry as an α-helix. This conformational arrangement allows the design of high affinity inhibitors mimicking known peptide sequences binding specific protein substrates. We show that GAFF and AutoDock potentials do not properly capture the conformational preferences of α-helix mimetics based on arylamide oligomers and identify alternate parameters matching solution NMR data and suitable for molecular dynamics simulation of arylamide compounds. Results from both docking and molecular dynamics simulations are consistent with the arylamides binding in the p53 peptide binding pocket. Simulations of arylamides in the p53 binding pocket of hDM2 are consistent with binding, exhibiting similar structural dynamics in the pocket as simulations of known hDM2 binders Nutlin-2 and a benzodiazepinedione compound. Arylamide conformations converge towards the same region of the binding pocket on the 20 ns time scale, and most, though not all dihedrals in the binding pocket are well sampled on this timescale. We show that there are two putative classes of binding modes for arylamide compounds supported equally by the modeling evidence. In the first, the arylamide compound lies parallel to the observed p53 helix. In the second class, not previously identified or proposed, the arylamide compound lies anti-parallel to the p53 helix.


Subject(s)
Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Binding Sites , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Piperazines/chemistry , Piperazines/metabolism
5.
J Chem Inf Model ; 51(3): 624-34, 2011 Mar 28.
Article in English | MEDLINE | ID: mdl-21361385

ABSTRACT

Ligand promiscuity, which is now recognized as an extremely common phenomenon, is a major underlying cause of drug toxicity. We have developed a new reverse virtual screening (VS) method called ReverseScreen3D, which can be used to predict the potential protein targets of a query compound of interest. The method uses a 2D fingerprint-based method to select a ligand template from each unique binding site of each protein within a target database. The target database contains only the structurally determined bioactive conformations of known ligands. The 2D comparison is followed by a 3D structural comparison to the selected query ligand using a geometric matching method, in order to prioritize each target binding site in the database. We have evaluated the performance of the ReverseScreen2D and 3D methods using a diverse set of small molecule protein inhibitors known to have multiple targets, and have shown that they are able to provide a highly significant enrichment of true targets in the database. Furthermore, we have shown that the 3D structural comparison improves early enrichment when compared with the 2D method alone, and that the 3D method performs well even in the absence of 2D similarity to the template ligands. By carrying out further experimental screening on the prioritized list of targets, it may be possible to determine the potential targets of a new compound or determine the off-targets of an existing drug. The ReverseScreen3D method has been incorporated into a Web server, which is freely available at http://www.modelling.leeds.ac.uk/ReverseScreen3D .


Subject(s)
Proteins/chemistry , Ligands , Models, Molecular , Molecular Structure
6.
J Chem Inf Model ; 51(2): 408-19, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21291174

ABSTRACT

Docking scoring functions are notoriously weak predictors of binding affinity. They typically assign a common set of weights to the individual energy terms that contribute to the overall energy score; however, these weights should be gene family dependent. In addition, they incorrectly assume that individual interactions contribute toward the total binding affinity in an additive manner. In reality, noncovalent interactions often depend on one another in a nonlinear manner. In this paper, we show how the use of support vector machines (SVMs), trained by associating sets of individual energy terms retrieved from molecular docking with the known binding affinity of each compound from high-throughput screening experiments, can be used to improve the correlation between known binding affinities and those predicted by the docking program eHiTS. We construct two prediction models: a regression model trained using IC(50) values from BindingDB, and a classification model trained using active and decoy compounds from the Directory of Useful Decoys (DUD). Moreover, to address the issue of overrepresentation of negative data in high-throughput screening data sets, we have designed a multiple-planar SVM training procedure for the classification model. The increased performance that both SVMs give when compared with the original eHiTS scoring function highlights the potential for using nonlinear methods when deriving overall energy scores from their individual components. We apply the above methodology to train a new scoring function for direct inhibitors of Mycobacterium tuberculosis (M.tb) InhA. By combining ligand binding site comparison with the new scoring function, we propose that phosphodiesterase inhibitors can potentially be repurposed to target M.tb InhA. Our methodology may be applied to other gene families for which target structures and activity data are available, as demonstrated in the work presented here.


Subject(s)
Drug Discovery/methods , Drug Repositioning , Algorithms , Binding Sites , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/chemistry , Humans , Ligands , Models, Molecular , Phosphodiesterase Inhibitors/chemistry , Phosphoric Diester Hydrolases/chemistry , Software , Support Vector Machine
7.
Biometrics ; 67(2): 611-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20618307

ABSTRACT

One of the key ingredients in drug discovery is the derivation of conceptual templates called pharmacophores. A pharmacophore model characterizes the physicochemical properties common to all active molecules, called ligands, bound to a particular protein receptor, together with their relative spatial arrangement. Motivated by this important application, we develop a Bayesian hierarchical model for the derivation of pharmacophore templates from multiple configurations of point sets, partially labeled by the atom type of each point. The model is implemented through a multistage template hunting algorithm that produces a series of templates that capture the geometrical relationship of atoms matched across multiple configurations. Chemical information is incorporated by distinguishing between atoms of different elements, whereby different elements are less likely to be matched than atoms of the same element. We illustrate our method through examples of deriving templates from sets of ligands that all bind structurally related protein active sites and show that the model is able to retrieve the key pharmacophore features in two test cases.


Subject(s)
Bayes Theorem , Computational Biology/methods , Drug Design , Algorithms , Biometry/methods , Catalytic Domain , Drug Discovery , Proteins/chemistry , Structure-Activity Relationship
8.
PLoS Comput Biol ; 6(11): e1000976, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-21079673

ABSTRACT

We report a computational approach that integrates structural bioinformatics, molecular modelling and systems biology to construct a drug-target network on a structural proteome-wide scale. The approach has been applied to the genome of Mycobacterium tuberculosis (M.tb), the causative agent of one of today's most widely spread infectious diseases. The resulting drug-target interaction network for all structurally characterized approved drugs bound to putative M.tb receptors, we refer to as the 'TB-drugome'. The TB-drugome reveals that approximately one-third of the drugs examined have the potential to be repositioned to treat tuberculosis and that many currently unexploited M.tb receptors may be chemically druggable and could serve as novel anti-tubercular targets. Furthermore, a detailed analysis of the TB-drugome has shed new light on the controversial issues surrounding drug-target networks [1]-[3]. Indeed, our results support the idea that drug-target networks are inherently modular, and further that any observed randomness is mainly caused by biased target coverage. The TB-drugome (http://funsite.sdsc.edu/drugome/TB) has the potential to be a valuable resource in the development of safe and efficient anti-tubercular drugs. More generally the methodology may be applied to other pathogens of interest with results improving as more of their structural proteomes are determined through the continued efforts of structural biology/genomics.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Computational Biology/methods , Molecular Targeted Therapy/methods , Mycobacterium tuberculosis/metabolism , Antitubercular Agents/chemistry , Binding Sites , Cluster Analysis , Computer Simulation , Databases, Factual , Humans , Models, Biological , Reproducibility of Results , Tuberculosis/drug therapy , Tuberculosis/microbiology
9.
J Chem Inf Model ; 50(10): 1806-20, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-20883031

ABSTRACT

Inhibition of amyloid fibril formation by stabilization of the native form of the protein transthyretin (TTR) is a viable approach for the treatment of familial amyloid polyneuropathy that has been gaining momentum in the field of amyloid research. The TTR stabilizer molecules discovered to date have shown efficacy at inhibiting fibrilization in vitro but display impairing issues of solubility, affinity for TTR in the blood plasma and/or adverse effects. In this study we present a benchmark of four protein- and ligand-based virtual screening (VS) methods for identifying novel TTR stabilizers: (i) two-dimensional (2D) similarity searches with chemical hashed, pharmacophore, and UNITY fingerprints, (ii) 3D searches based on shape, chemical, and electrostatic similarity, (iii) LigMatch, a new ligand-based method which uses multiple templates and combines 3D geometric hashing with a 2D preselection process, and (iv) molecular docking to consensus X-ray crystal structures of TTR. We illustrate the potential of the best-performing VS protocols to retrieve promising new leads by ranking a tailored library of 2.3 million commercially available compounds. Our predictions show that the top-scoring molecules possess distinctive features from the known TTR binders, holding better solubility, fraction of halogen atoms, and binding affinity profiles. To the best of our knowledge, this is the first attempt to rationalize the utilization of a large battery of in silico screening techniques toward the identification of a new generation of TTR amyloid inhibitors.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Amyloid/antagonists & inhibitors , Drug Design , Prealbumin/antagonists & inhibitors , Prealbumin/metabolism , Amyloid/metabolism , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Prealbumin/chemistry , Protein Binding , Protein Conformation
10.
J Mol Biol ; 399(4): 645-61, 2010 Jun 18.
Article in English | MEDLINE | ID: mdl-20434455

ABSTRACT

Current homology-modelling methods do not consider small molecules in their automated processes. Therefore, the development of a reliable tool for protein-ligand homology modelling is an important next step in generating plausible models for molecular interactions. Two automated protein-ligand homology-modelling strategies, requiring no expert knowledge from the user, are investigated here. Both employ the "induced fit" concept with flexibility in side chains and ligand. The most successful strategy superimposes the new ligand over the original ligand before homology modelling, allowing the new ligand to be taken into consideration during protein modelling (rather than after), facilitating conformational change in the local backbone if necessary. We show that this approach results in successful modelling of the ligand and key binding-site residues of angiotensin-converting enzyme 2 (ACE2) from its homologue ACE, which is not possible via conventional homology modelling or by homology modelling followed by docking. Several other difficult target complexes are also successfully modelled, reproducing native protein-ligand contacts with significantly different biological substrates and different binding-site conformations. These include the modelling of Cdk5 (cyclin-dependent kinase 5) from Cdk2, thymidine phosphorylase from a bacterial homologue, and dihydrofolate reductase from a recombinant variant with a markedly different inhibitor. In terms of average modelling quality across 82 targets, the ligand RMSD with respect to the experimental structure is 1.4 A (and 2.0 A for the protein binding site) for "easy" cases and 2.9 A for the ligand (and 2.7 A for the protein binding site) in "hard" cases. This demonstrates the importance of selecting an optimal template. Ligand-modelling accuracy is strongly dependent on target-template ligand structural similarity, rather than target-template sequence identity. However, protein-modelling accuracy is dependent on both. Our automated protein-ligand homology-modelling strategy generates a higher degree of accuracy than homology modelling followed by docking, generating an average ligand RMSD that is 1-2 A better than docking with homology models.


Subject(s)
Models, Molecular , Structural Homology, Protein , Algorithms , Binding Sites , Databases, Protein , Ligands , Protein Conformation , Sequence Alignment , Software
11.
J Mol Graph Model ; 28(3): 297-303, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19762259

ABSTRACT

Carbohydrate binding sites are considered important for cellular recognition and adhesion and are important targets for drug design. In this paper we present a new method called InCa-SiteFinder for predicting non-covalent inositol and carbohydrate binding sites on the surface of protein structures. It uses the van der Waals energy of a protein-probe interaction and amino acid propensities to locate and predict carbohydrate binding sites. The protein surface is searched for continuous volume envelopes that correspond to a favorable protein-probe interaction. These volumes are subsequently analyzed to demarcate regions of high cumulative propensity for binding a carbohydrate moiety based on calculated amino acid propensity scores. InCa-SiteFinder(1) was tested on an independent test set of 80 protein-ligand complexes. It efficiently identifies carbohydrate binding sites with high specificity and sensitivity. It was also tested on a second test set of 80 protein-ligand complexes containing 40 known carbohydrate binders (having 40 carbohydrate binding sites) and 40 known drug-like compound binders (having 58 known drug-like compound binding sites) for the prediction of the location of the carbohydrate binding sites and to distinguish these from the drug-like compound binding sites. At 73% sensitivity the method showed 98% specificity. Almost all of the carbohydrate and drug-like compound binding sites were correctly identified with an overall error rate of 12%.


Subject(s)
Carbohydrates/chemistry , Computational Biology/methods , Inositol/chemistry , Inositol/metabolism , Proteins/chemistry , Proteins/metabolism , Binding Sites , Protein Binding , Protein Conformation
12.
J Chem Inf Model ; 49(9): 2056-66, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19685924

ABSTRACT

We have developed a new virtual screening (VS) method called LigMatch and evaluated its performance on 13 protein targets using a filtered and clustered version of the directory of useful decoys (DUD). The method uses 3D structural comparison to a crystallographically determined ligand in a bioactive 'template' conformation, using a geometric hashing method, in order to prioritize each database compound. We show that LigMatch outperforms several other widely used VS methods on the 13 DUD targets. We go on to demonstrate that improved VS performance can be gained from using multiple, structurally diverse templates rather than a single template ligand for a particular protein target. In this case, a 2D fingerprint-based method is used to select a ligand template from a set of known bioactive conformations. Furthermore, we show that LigMatch performs well even in the absence of 2D similarity to the template ligands, thereby demonstrating its robustness with respect to purely 2D methods and its potential for scaffold hopping.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Molecular , Molecular Conformation , User-Computer Interface , Databases, Factual , Ligands , ROC Curve
13.
Nat Genet ; 41(7): 829-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19525956

ABSTRACT

Aicardi-Goutières syndrome is a mendelian mimic of congenital infection and also shows overlap with systemic lupus erythematosus at both a clinical and biochemical level. The recent identification of mutations in TREX1 and genes encoding the RNASEH2 complex and studies of the function of TREX1 in DNA metabolism have defined a previously unknown mechanism for the initiation of autoimmunity by interferon-stimulatory nucleic acid. Here we describe mutations in SAMHD1 as the cause of AGS at the AGS5 locus and present data to show that SAMHD1 may act as a negative regulator of the cell-intrinsic antiviral response.


Subject(s)
Brain Diseases, Metabolic, Inborn/genetics , Immunity, Innate , Monomeric GTP-Binding Proteins/genetics , Amino Acid Substitution , Brain Diseases, Metabolic, Inborn/immunology , Humans , Monomeric GTP-Binding Proteins/immunology , SAM Domain and HD Domain-Containing Protein 1
14.
J Chem Inf Model ; 49(2): 318-29, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19434833

ABSTRACT

Methods for analyzing complete gene families are becoming of increasing importance to the drug discovery process, because similarities and differences within a family are often the key to understanding functional differences that can be exploited in drug design. We undertake a large-scale structural comparison of protein kinase ATP-binding sites using a geometric hashing method. Subsequently, we propose a relevant classification of the protein kinase family based on the structural similarity of its binding sites. Our classification is not only able to reveal the great diversity of different protein kinases and therefore their different potential for inhibitor selectivity but it is also able to distinguish subtle differences within binding site conformation reflecting the protein activation state. Furthermore, using experimental inhibition profiling, we demonstrate that our classification can be used to identify protein kinase binding sites that are known experimentally to bind the same drug, demonstrating that it has potential as an inverse (protein) virtual screening tool, by identifying which other sites have the potential to bind a given drug. In this way the cross-reactivities of the anticancer drugs Tarceva and Gleevec are rationalized.


Subject(s)
Protein Kinases/metabolism , Binding Sites , Models, Molecular , Protein Conformation , Protein Kinases/chemistry , Protein Kinases/classification
15.
Drug Discov Today ; 14(3-4): 155-61, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19041415

ABSTRACT

Protein-protein interfaces are highly attractive targets for drug discovery because they are involved in a large number of disease pathways where therapeutic intervention would bring widespread benefit. Recent successes have challenged the widely held belief that these targets are 'undruggable'. The pocket finding algorithms described here show marked differences between the binding pockets that define protein-protein interactions (PPIs) and those that define protein-ligand interactions (PLIs) of currently marketed drugs. In the case of PPIs, drug discovery methods that simultaneously target several small pockets at the protein-protein interface are likely to increase the chances of success in this new and important field of therapeutics.


Subject(s)
Drug Delivery Systems , Drug Discovery/methods , Proteins/metabolism , Algorithms , Binding Sites , Humans , Ligands , Protein Binding
16.
J Chem Inf Model ; 48(10): 1990-8, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18767831

ABSTRACT

The development and validation of a new knowledge based scoring function (SIScoreJE) to predict binding energy between proteins and ligands is presented. SIScoreJE efficiently predicts the binding energy between a small molecule and its protein receptor. Protein-ligand atomic contact information was derived from a Non-Redundant Data set (NRD) of over 3000 X-ray crystal structures of protein-ligand complexes. This information was classified for individual "atom contact pairs" (ACP) which is used to calculate the atomic contact preferences. In addition to the two schemes generated in this study we have assessed a number of other common atom-type classification schemes. The preferences were calculated using an information theoretic relationship of joint entropy. Among 18 different atom-type classification schemes "ScoreJE Atom Type set2" (SATs2) was found to be the most suitable for our approach. To test the sensitivity of the method to the inclusion of solvent, Single-body Solvation Potentials (SSP) were also derived from the atomic contacts between the protein atom types and water molecules modeled using AQUARIUS2. Validation was carried out using an evaluation data set of 100 protein-ligand complexes with known binding energies to test the ability of the scoring functions to reproduce known binding affinities. In summary, it was found that a combined SSP/ScoreJE (SIScoreJE) performed significantly better than ScoreJE alone, and SIScoreJE and ScoreJE performed better than GOLD::GoldScore, GOLD::ChemScore, and XScore.


Subject(s)
Information Theory , Ligands , Proteins/chemistry , Structure-Activity Relationship , Adenosine Deaminase/chemistry , Adenosine Deaminase/drug effects , Algorithms , Computer Simulation , Databases, Protein , Drug Evaluation, Preclinical , Entropy , Phosphoribosylglycinamide Formyltransferase/chemistry , Phosphoribosylglycinamide Formyltransferase/drug effects , Protein Binding , Protein Conformation , Reproducibility of Results , Software , X-Ray Diffraction
17.
BMC Genomics ; 8: 194, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17597519

ABSTRACT

BACKGROUND: Mammalian angiotensin converting enzyme (ACE) plays a key role in blood pressure regulation. Although multiple ACE-like proteins exist in non-mammalian organisms, to date only one other ACE homologue, ACE2, has been identified in mammals. RESULTS: Here we report the identification and characterisation of the gene encoding a third homologue of ACE, termed ACE3, in several mammalian genomes. The ACE3 gene is located on the same chromosome downstream of the ACE gene. Multiple sequence alignment and molecular modelling have been employed to characterise the predicted ACE3 protein. In mouse, rat, cow and dog, the predicted protein has mutations in some of the critical residues involved in catalysis, including the catalytic Glu in the HEXXH zinc binding motif which is Gln, and ESTs or reverse-transcription PCR indicate that the gene is expressed. In humans, the predicted ACE3 protein has an intact HEXXH motif, but there are other deletions and insertions in the gene and no ESTs have been identified. CONCLUSION: In the genomes of several mammalian species there is a gene that encodes a novel, single domain ACE-like protein, ACE3. In mouse, rat, cow and dog ACE3, the catalytic Glu is replaced by Gln in the putative zinc binding motif, indicating that in these species ACE3 would lack catalytic activity as a zinc metalloprotease. In humans, no evidence was found that the ACE3 gene is expressed and the presence of deletions and insertions in the sequence indicate that ACE3 is a pseudogene.


Subject(s)
Gene Expression Profiling , Genomics/methods , Peptidyl-Dipeptidase A/genetics , Amino Acid Sequence , Animals , Cattle , Dogs , Expressed Sequence Tags , Humans , Metalloproteases/chemistry , Mice , Molecular Sequence Data , Peptidyl-Dipeptidase A/chemistry , Rats , Sequence Homology, Amino Acid , Species Specificity
18.
Bioinformatics ; 23(15): 1901-8, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17510171

ABSTRACT

MOTIVATION: There are two main areas of difficulty in homology modelling that are particularly important when sequence identity between target and template falls below 50%: sequence alignment and loop building. These problems become magnified with automatic modelling processes, as there is no human input to correct mistakes. As such we have benchmarked several stand-alone strategies that could be implemented in a workflow for automated high-throughput homology modelling. These include three new sequence-structure alignment programs: 3D-Coffee, Staccato and SAlign, plus five homology modelling programs and their respective loop building methods: Builder, Nest, Modeller, SegMod/ENCAD and Swiss-Model. The SABmark database provided 123 targets with at least five templates from the same SCOP family and sequence identities

Subject(s)
Algorithms , Models, Chemical , Proteins/chemistry , Sequence Alignment/methods , Sequence Analysis, Protein/methods , Software Validation , Software , Amino Acid Sequence , Computer Simulation , Models, Molecular , Molecular Sequence Data , Reproducibility of Results , Sensitivity and Specificity
19.
Bioinformatics ; 23(5): 573-81, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17237047

ABSTRACT

MOTIVATION: Due to the limitations in experimental methods for determining binary interactions and structure determination of protein complexes, the need exists for computational models to fill the increasing gap between genome sequence information and protein annotation. Here we describe a novel method that uses structural models to reduce a large number of in silico predictions to a high confidence subset that is amenable to experimental validation. RESULTS: A two-stage evaluation procedure was developed, first, a sequence-based method assessed the conservation of protein interface patches used in the original in silico prediction method, both in terms of position within the primary sequence, and in terms of sequence conservation. When applying the most stringent conditions it was found that 20.5% of the data set being assessed passed this test. Secondly, a high-throughput structure-based docking evaluation procedure assessed the soundness of three dimensional models produced for the putative interactions. Of the data set being assessed, 8264 interactions or over 70% could be modelled in this way, and 27% of these can be considered 'valid' by the applied criteria. In all, 6.9% of the interactions passed both the tests and can be considered to be a high confidence set of predicted interactions, several of which are described. AVAILABILITY: http://bioinformatics.leeds.ac.uk/~bmb4sjc. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Models, Molecular , Protein Conformation , Protein Interaction Mapping/methods , Azotobacter vinelandii/enzymology , Computer Simulation , Databases, Protein , Dihydrolipoamide Dehydrogenase/chemistry , Insulin Receptor Substrate Proteins , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Proteins/chemistry , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Structure-Activity Relationship
20.
Curr Protein Pept Sci ; 7(5): 395-406, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17073692

ABSTRACT

Structure Based Drug Design (SBDD) is a computational approach to lead discovery that uses the three-dimensional structure of a protein to fit drug-like molecules into a ligand binding site to modulate function. Identifying the location of the binding site is therefore a vital first step in this process, restricting the search space for SBDD or virtual screening studies. The detection and characterisation of functional sites on proteins has increasingly become an area of interest. Structural genomics projects are increasingly yielding protein structures with unknown functions and binding sites. Binding site prediction was pioneered by pocket detection, since the binding site is often found in the largest pocket. More recent methods involve phylogenetic analysis, identifying structural similarity with proteins of known function and identifying regions on the protein surface with a potential for high binding affinity. Binding site prediction has been used in several SBDD projects and has been incorporated into several docking tools. We discuss different methods of ligand binding site prediction, their strengths and weaknesses, and how they have been used in SBDD.


Subject(s)
Drug Design , Drug Evaluation, Preclinical/methods , Animals , Binding Sites , HIV Protease/chemistry , Ligands , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...