Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 23(9)2022 Apr 26.
Article in English | MEDLINE | ID: mdl-35563148

ABSTRACT

The prediction of how a ligand binds to its target is an essential step for Structure-Based Drug Design (SBDD) methods. Molecular docking is a standard tool to predict the binding mode of a ligand to its macromolecular receptor and to quantify their mutual complementarity, with multiple applications in drug design. However, docking programs do not always find correct solutions, either because they are not sampled or due to inaccuracies in the scoring functions. Quantifying the docking performance in real scenarios is essential to understanding their limitations, managing expectations and guiding future developments. Here, we present a fully automated pipeline for pose prediction validated by participating in the Continuous Evaluation of Ligand Pose Prediction (CELPP) Challenge. Acknowledging the intrinsic limitations of the docking method, we devised a strategy to automatically mine and exploit pre-existing data, defining-whenever possible-empirical restraints to guide the docking process. We prove that the pipeline is able to generate predictions for most of the proposed targets as well as obtain poses with low RMSD values when compared to the crystal structure. All things considered, our pipeline highlights some major challenges in the automatic prediction of protein-ligand complexes, which will be addressed in future versions of the pipeline.


Subject(s)
Drug Design , Binding Sites , Crystallography, X-Ray , Ligands , Molecular Docking Simulation , Protein Binding , Protein Conformation
2.
Sensors (Basel) ; 22(6)2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35336249

ABSTRACT

The Sunrise missions consist of observing the magnetic field of the sun continuously for a few days from the stratosphere. In these missions, a balloon supporting a telescope and associated instrumentation, including a Tunable Magnetograph (TuMag), is lifted into the stratosphere. In the camera of this instrument, the image sensor sends its data to a Field Programmable Gate Array (FPGA) using eight transmission channels. These channels must be previously calibrated for a correct delivery of the image. For this mission, the FPGA has been exchanged for a newer and larger one, so the firmware has been adapted to the new device. In addition, the calibration algorithm has been parallelized as the main innovation of this work, taking advantage of the increase in logic resources of the new FPGA, in order to minimize the calibration time of the channels. The algorithm has been implemented specifically for this instrument without using the Input Serial Deserializer (ISERDES) Intellectual Property (IP), since this IP does not support the deserialization of the data sent by the image sensor to the FPGA.


Subject(s)
Algorithms , Magnetic Fields , Calibration
3.
Curr Drug Discov Technol ; 19(2): 62-68, 2022.
Article in English | MEDLINE | ID: mdl-34951392

ABSTRACT

BACKGROUND: Mixed solvents MD (MDmix) simulations have proved to be a useful and increasingly accepted technique with several applications in structure-based drug discovery. One of the assumptions behind the methodology is the transferability of free energy values from the simulated cosolvent molecules to larger drug-like molecules. However, the binding free energy maps (ΔGbind) calculated for the different moieties of the cosolvent molecules (e.g. a hydroxyl map for the ethanol) are largely influenced by the rest of the solvent molecule and do not reflect the intrinsic affinity of the moiety in question. As such, they are hardly transferable to different molecules. METHOD: To achieve transferable energies, we present here a method for decomposing the molecular binding free energy into accurate atomic contributions. RESULT: We demonstrate with two qualitative visual examples how the corrected energy maps better match known binding hotspots and how they can reveal hidden hotspots with actual drug design potential. CONCLUSION: Atomic decomposition of binding free energies derived from MDmix simulations provides transferable and quantitative binding free energy maps.


Subject(s)
Drug Design , Molecular Dynamics Simulation , Drug Discovery , Solvents/chemistry
5.
J Chem Inf Model ; 57(4): 846-863, 2017 04 24.
Article in English | MEDLINE | ID: mdl-28318252

ABSTRACT

One of the most important biological processes at the molecular level is the formation of protein-ligand complexes. Therefore, determining their structure and underlying key interactions is of paramount relevance and has direct applications in drug development. Because of its low cost relative to its experimental sibling, molecular dynamics (MD) simulations in the presence of different solvent probes mimicking specific types of interactions have been increasingly used to analyze protein binding sites and reveal protein-ligand interaction hot spots. However, a systematic comparison of different probes and their real predictive power from a quantitative and thermodynamic point of view is still missing. In the present work, we have performed MD simulations of 18 different proteins in pure water as well as water mixtures of ethanol, acetamide, acetonitrile and methylammonium acetate, leading to a total of 5.4 µs simulation time. For each system, we determined the corresponding solvent sites, defined as space regions adjacent to the protein surface where the probability of finding a probe atom is higher than that in the bulk solvent. Finally, we compared the identified solvent sites with 121 different protein-ligand complexes and used them to perform molecular docking and ligand binding free energy estimates. Our results show that combining solely water and ethanol sites allows sampling over 70% of all possible protein-ligand interactions, especially those that coincide with ligand-based pharmacophoric points. Most important, we also show how the solvent sites can be used to significantly improve ligand docking in terms of both accuracy and precision, and that accurate predictions of ligand binding free energies, along with relative ranking of ligand affinity, can be performed.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Proteins/chemistry , Proteins/metabolism , Solvents/chemistry , Hydrophobic and Hydrophilic Interactions , Ligands , Protein Binding , Protein Conformation , Thermodynamics , Water/chemistry
6.
PLoS One ; 10(7): e0131462, 2015.
Article in English | MEDLINE | ID: mdl-26154497

ABSTRACT

As previously reported, P. aeruginosa genes PA2077 and PA2078 code for 10S-DOX (10S-Dioxygenase) and 7,10-DS (7,10-Diol Synthase) enzymes involved in long-chain fatty acid oxygenation through the recently described oleate-diol synthase pathway. Analysis of the amino acid sequence of both enzymes revealed the presence of two heme-binding motifs (CXXCH) on each protein. Phylogenetic analysis showed the relation of both proteins to bacterial di-heme cytochrome c peroxidases (Ccps), similar to Xanthomonas sp. 35Y rubber oxidase RoxA. Structural homology modelling of PA2077 and PA2078 was achieved using RoxA (pdb 4b2n) as a template. From the 3D model obtained, presence of significant amino acid variations in the predicted heme-environment was found. Moreover, the presence of palindromic repeats located in enzyme-coding regions, acting as protein evolution elements, is reported here for the first time in P. aeruginosa genome. These observations and the constructed phylogenetic tree of the two proteins, allow the proposal of an evolutionary pathway for P. aeruginosa oleate-diol synthase operon. Taking together the in silico and in vivo results obtained we conclude that enzymes PA2077 and PA2078 are the first described members of a new subfamily of bacterial peroxidases, designated as Fatty acid-di-heme Cytochrome c peroxidases (FadCcp).


Subject(s)
Computer Simulation , Cytochrome-c Peroxidase/genetics , Evolution, Molecular , Heme/metabolism , Multigene Family , Oleic Acid/metabolism , Pseudomonas aeruginosa/enzymology , Amino Acid Motifs , Amino Acid Sequence , Genes, Bacterial , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Mutant Proteins/metabolism , Operon/genetics , Oxygenases/metabolism , Phylogeny , Pseudomonas aeruginosa/genetics , Sequence Alignment , Structural Homology, Protein
7.
J Med Chem ; 57(20): 8530-9, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25275946

ABSTRACT

Binding sites present well-defined interaction patterns that putative ligands must meet. Knowing them is essential to guide structure-based drug discovery projects. However, complex aspects of molecular recognition-such as protein flexibility or the effect of aqueous solvation-hinder accurate predictions. This is particularly true for polar contacts, which are heavily influenced by the local environment and the behavior of discrete water molecules. Here we present and validate MDmix (Molecular Dynamics simulations with mixed solvents) as a method that provides much more accurate interaction maps than ordinary potentials (e.g., GRID). Additionally, MDmix also affords water displaceability predictions, with advantages over methods that use pure water as solvent (e.g., inhomogeneous fluid solvation theory). With current MD software and hardware solutions, predictions can be obtained in a matter of hours and visualized in a very intuitive manner. Thus, MDmix is an ideal complement in everyday structure-based drug discovery projects.


Subject(s)
Molecular Dynamics Simulation , Proteins/chemistry , Proteins/metabolism , Software , Binding Sites , HIV Protease/chemistry , HIV Protease/metabolism , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Reproducibility of Results , Solvents/chemistry , Water
8.
PLoS Comput Biol ; 10(4): e1003571, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24722481

ABSTRACT

Identification of chemical compounds with specific biological activities is an important step in both chemical biology and drug discovery. When the structure of the intended target is available, one approach is to use molecular docking programs to assess the chemical complementarity of small molecules with the target; such calculations provide a qualitative measure of affinity that can be used in virtual screening (VS) to rank order a list of compounds according to their potential to be active. rDock is a molecular docking program developed at Vernalis for high-throughput VS (HTVS) applications. Evolved from RiboDock, the program can be used against proteins and nucleic acids, is designed to be computationally very efficient and allows the user to incorporate additional constraints and information as a bias to guide docking. This article provides an overview of the program structure and features and compares rDock to two reference programs, AutoDock Vina (open source) and Schrödinger's Glide (commercial). In terms of computational speed for VS, rDock is faster than Vina and comparable to Glide. For binding mode prediction, rDock and Vina are superior to Glide. The VS performance of rDock is significantly better than Vina, but inferior to Glide for most systems unless pharmacophore constraints are used; in that case rDock and Glide are of equal performance. The program is released under the Lesser General Public License and is freely available for download, together with the manuals, example files and the complete test sets, at http://rdock.sourceforge.net/


Subject(s)
Nucleic Acids/chemistry , Proteins/chemistry , Drug Discovery , Ligands
9.
J Chem Theory Comput ; 10(6): 2608-14, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-26580781

ABSTRACT

Conceptually, the simplistic lock and key model has been superseded by more realistic views of molecular recognition that take into account the intrinsic dynamics of biological macromolecules. However, it is still common for structure-based drug discovery methods to represent the receptor as static structures. The practical advantages of this approximation, the notable success attained over the past few decades with such simple models and the absence of clear guidelines for weighing the pros and cons of accounting for flexibility may prompt some investigators to stretch the rigid model beyond its scope. Here, we investigate the relationship between protein flexibility and binding free energy and present some useful hints for understanding when, and to what extent, flexibility should be considered. Using molecular dynamics simulations of hen egg-white lysozyme (HEWL) with explicit aqueous/organic solvent mixtures and a range of restraint conditions, we find out how artificially restricted mobility affects binding hot spots. Barring sampling errors or an inappropriate choice of reference structure, we find that decreased mobility (measured as B-factors) leads to artifactually more negative binding free energies, but a logarithmic relationship between both terms attenuates the errors. Consequently, ignoring flexibility may be an acceptable approximation for intrinsically rigid regions (such as the active site of enzymes) but may lead to larger errors elsewhere. For the same reason, local conformational sampling yields very accurate predictions and, owing to its practical advantages, may be preferable to full conformational sampling for many applications.

SELECTION OF CITATIONS
SEARCH DETAIL
...