Searching the conformational space for docking

Last updated

In molecular modelling, docking is a method which predicts the preferred orientation of one molecule to another when bound together in a stable complex. In the case of protein docking, the search space consists of all possible orientations of the protein with respect to the ligand. Flexible docking in addition considers all possible conformations of the protein paired with all possible conformations of the ligand. [1]

Contents

With present computing resources, it is impossible to exhaustively explore these search spaces; instead, there are many strategies which attempt to sample the search space with optimal efficiency. Most docking programs in use account for a flexible ligand, and several attempt to model a flexible protein receptor. Each "snapshot" of the pair is referred to as a pose.

Molecular dynamics (MD) simulations

In this approach, proteins are typically held rigid, and the ligand is allowed to freely explore their conformational space. The generated conformations are then docked successively into the protein, and an MD simulation consisting of a simulated annealing protocol is performed. This is usually supplemented with short MD energy minimization steps, and the energies determined from the MD runs are used for ranking the overall scoring. Although this is a computer-expensive method (involving potentially hundreds of MD runs), it has some advantages: for example, no specialized energy/scoring functions are required. MD force-fields can typically be used to find poses that are reasonable and can be compared with experimental structures.

The Distance Constrained Essential Dynamics method (DCED) has been used to generate multiple structures for docking, called eigenstructures. This approach, although avoiding most of the costly MD calculations, can capture the essential motions involved in a flexible receptor, representing a form of coarse-grained dynamics. [2]

Shape-complementarity methods

The most common technique used in many docking programs, shape-complementarity methods focus on the match between the receptor and the ligand in order to find an optimal pose. Programs include DOCK, [3] FRED, [4] GLIDE, [5] SURFLEX, [6] eHiTS [7] and many more. Most methods describe the molecules in terms of a finite number of descriptors that include structural complementarity and binding complementarity. Structural complementarity is mostly a geometric description of the molecules, including solvent-accessible surface area, overall shape and geometric constraints between atoms in the protein and ligand. Binding complementarity takes into account features like hydrogen bonding interactions, hydrophobic contacts and van der Waals interactions to describe how well a particular ligand will bind to the protein. Both kinds of descriptors are conveniently represented in the form of structural templates which are then used to quickly match potential compounds (either from a database or from the user-given inputs) that will bind well at the active site of the protein. Compared to the all-atom molecular dynamics approaches, these methods are very efficient in finding optimal binding poses for the protein and ligand.

Genetic algorithms

Two of the most used docking programs belong to this class: GOLD [8] and AutoDock. [9] Genetic algorithms allow the exploration of a large conformational space – which is basically spanned by the protein and ligand jointly in this case – by representing each spatial arrangement of the pair as a “gene” with a particular energy. The entire genome thus represents the complete energy landscape which is to be explored. The simulation of the evolution of the genome is carried out by cross-over techniques similar to biological evolution, where random pairs of individuals (conformations) are “mated” with the possibility for a random mutation in the offspring. These methods have proven very useful in sampling the vast state-space while maintaining closeness to the actual process involved.

Although genetic algorithms are quite successful in sampling the large conformational space, many docking programs require the protein to remain fixed, while allowing only the ligand to flex and adjust to the active site of the protein. Genetic algorithms also require multiple runs to obtain reliable answers regarding ligands that may bind to the protein. The time it takes to typically run a genetic algorithm in order to allow a proper pose may be longer, hence these methods may not be as efficient as shape complementarity-based approaches in screening large databases of compounds. Recent improvements in using grid-based evaluation of energies, limiting the exploration of the conformational changes at only local areas (active sites) of interest, and improved tabling methods have significantly enhanced the performance of genetic algorithms and made them suitable for virtual screening applications.

Related Research Articles

<span class="mw-page-title-main">Protein</span> Biomolecule consisting of chains of amino acid residues

Proteins are large biomolecules and macromolecules that comprise one or more long chains of amino acid residues. Proteins perform a vast array of functions within organisms, including catalysing metabolic reactions, DNA replication, responding to stimuli, providing structure to cells and organisms, and transporting molecules from one location to another. Proteins differ from one another primarily in their sequence of amino acids, which is dictated by the nucleotide sequence of their genes, and which usually results in protein folding into a specific 3D structure that determines its activity.

<span class="mw-page-title-main">Allosteric regulation</span> Regulation of enzyme activity

In biochemistry, allosteric regulation is the regulation of an enzyme by binding an effector molecule at a site other than the enzyme's active site.

<span class="mw-page-title-main">Molecular dynamics</span> Computer simulations to discover and understand chemical properties

Molecular dynamics (MD) is a computer simulation method for analyzing the physical movements of atoms and molecules. The atoms and molecules are allowed to interact for a fixed period of time, giving a view of the dynamic "evolution" of the system. In the most common version, the trajectories of atoms and molecules are determined by numerically solving Newton's equations of motion for a system of interacting particles, where forces between the particles and their potential energies are often calculated using interatomic potentials or molecular mechanical force fields. The method is applied mostly in chemical physics, materials science, and biophysics.

<span class="mw-page-title-main">Structural bioinformatics</span> Bioinformatics subfield

Structural bioinformatics is the branch of bioinformatics that is related to the analysis and prediction of the three-dimensional structure of biological macromolecules such as proteins, RNA, and DNA. It deals with generalizations about macromolecular 3D structures such as comparisons of overall folds and local motifs, principles of molecular folding, evolution, binding interactions, and structure/function relationships, working both from experimentally solved structures and from computational models. The term structural has the same meaning as in structural biology, and structural bioinformatics can be seen as a part of computational structural biology. The main objective of structural bioinformatics is the creation of new methods of analysing and manipulating biological macromolecular data in order to solve problems in biology and generate new knowledge.

<span class="mw-page-title-main">Molecular mechanics</span> Use of classical mechanics to model molecular systems

Molecular mechanics uses classical mechanics to model molecular systems. The Born–Oppenheimer approximation is assumed valid and the potential energy of all systems is calculated as a function of the nuclear coordinates using force fields. Molecular mechanics can be used to study molecule systems ranging in size and complexity from small to large biological systems or material assemblies with many thousands to millions of atoms.

<span class="mw-page-title-main">Drug design</span> Inventive process of finding new medications based on the knowledge of a biological target

Drug design, often referred to as rational drug design or simply rational design, is the inventive process of finding new medications based on the knowledge of a biological target. The drug is most commonly an organic small molecule that activates or inhibits the function of a biomolecule such as a protein, which in turn results in a therapeutic benefit to the patient. In the most basic sense, drug design involves the design of molecules that are complementary in shape and charge to the biomolecular target with which they interact and therefore will bind to it. Drug design frequently but not necessarily relies on computer modeling techniques. This type of modeling is sometimes referred to as computer-aided drug design. Finally, drug design that relies on the knowledge of the three-dimensional structure of the biomolecular target is known as structure-based drug design. In addition to small molecules, biopharmaceuticals including peptides and especially therapeutic antibodies are an increasingly important class of drugs and computational methods for improving the affinity, selectivity, and stability of these protein-based therapeutics have also been developed.

Protein design is the rational design of new protein molecules to design novel activity, behavior, or purpose, and to advance basic understanding of protein function. Proteins can be designed from scratch or by making calculated variants of a known protein structure and its sequence. Rational protein design approaches make protein-sequence predictions that will fold to specific structures. These predicted sequences can then be validated experimentally through methods such as peptide synthesis, site-directed mutagenesis, or artificial gene synthesis.

<span class="mw-page-title-main">Protein–protein interaction</span> Physical interactions and constructions between multiple proteins

Protein–protein interactions (PPIs) are physical contacts of high specificity established between two or more protein molecules as a result of biochemical events steered by interactions that include electrostatic forces, hydrogen bonding and the hydrophobic effect. Many are physical contacts with molecular associations between chains that occur in a cell or in a living organism in a specific biomolecular context.

<span class="mw-page-title-main">Docking (molecular)</span> Prediction method in molecular modeling

In the field of molecular modeling, docking is a method which predicts the preferred orientation of one molecule to a second when a ligand and a target are bound to each other to form a stable complex. Knowledge of the preferred orientation in turn may be used to predict the strength of association or binding affinity between two molecules using, for example, scoring functions.

Macromolecular docking is the computational modelling of the quaternary structure of complexes formed by two or more interacting biological macromolecules. Protein–protein complexes are the most commonly attempted targets of such modelling, followed by protein–nucleic acid complexes.

<span class="mw-page-title-main">Intrinsically disordered proteins</span> Protein without a fixed 3D structure

In molecular biology, an intrinsically disordered protein (IDP) is a protein that lacks a fixed or ordered three-dimensional structure, typically in the absence of its macromolecular interaction partners, such as other proteins or RNA. IDPs range from fully unstructured to partially structured and include random coil, molten globule-like aggregates, or flexible linkers in large multi-domain proteins. They are sometimes considered as a separate class of proteins along with globular, fibrous and membrane proteins.

Protein–ligand docking is a molecular modelling technique. The goal of protein–ligand docking is to predict the position and orientation of a ligand when it is bound to a protein receptor or enzyme. Pharmaceutical research employs docking techniques for a variety of purposes, most notably in the virtual screening of large databases of available chemicals in order to select likely drug candidates. There has been rapid development in computational ability to determine protein structure with programs such as AlphaFold, and the demand for the corresponding protein-ligand docking predictions is driving implementation of software that can find accurate models. Once the protein folding can be predicted accurately along with how the ligands of various structures will bind to the protein, the ability for drug development to progress at a much faster rate becomes possible.

<span class="mw-page-title-main">Virtual screening</span>

Virtual screening (VS) is a computational technique used in drug discovery to search libraries of small molecules in order to identify those structures which are most likely to bind to a drug target, typically a protein receptor or enzyme.

In the fields of computational chemistry and molecular modelling, scoring functions are mathematical functions used to approximately predict the binding affinity between two molecules after they have been docked. Most commonly one of the molecules is a small organic compound such as a drug and the second is the drug's biological target such as a protein receptor. Scoring functions have also been developed to predict the strength of intermolecular interactions between two proteins or between protein and DNA.

The program UCSF DOCK was created in the 1980s by Irwin "Tack" Kuntz's Group, and was the first docking program. DOCK uses geometric algorithms to predict the binding modes of small molecules. Brian K. Shoichet, David A. Case, and Robert C.Rizzo are codevelopers of DOCK.

Lead Finder is a computational chemistry tool designed for modeling protein-ligand interactions. This application is useful for conducting molecular docking studies and quantitatively assessing ligand binding and biological activity. Lead Finder offers free access to individual users, especially those in non-commercial and academic settings.

Molecular Operating Environment (MOE) is a drug discovery software platform that integrates visualization, modeling and simulations, as well as methodology development, in one package. MOE scientific applications are used by biologists, medicinal chemists and computational chemists in pharmaceutical, biotechnology and academic research. MOE runs on Windows, Linux, Unix, and macOS. Main application areas in MOE include structure-based design, fragment-based design, ligand-based design, pharmacophore discovery, medicinal chemistry applications, biologics applications, structural biology and bioinformatics, protein and antibody modeling, molecular modeling and simulations, virtual screening, cheminformatics & QSAR. The Scientific Vector Language (SVL) is the built-in command, scripting and application development language of MOE.

LeDock is a proprietary, flexible molecular docking software designed for the purpose of docking ligands with target proteins. It is available for Linux, macOS, and Windows.

FlexAID is a molecular docking software that can use small molecules and peptides as ligands and proteins and nucleic acids as docking targets. As the name suggests, FlexAID supports full ligand flexibility as well side-chain flexibility of the target. It does using a soft scoring function based on the complementarity of the two surfaces.

Namasivayam Gautham is a retired Professor Emeritus at the Centre of Advance Study in Crystallography and Biophysics, University of Madras. He is known for his work on DNA Crystallography, protein structure prediction and molecular docking.

References

  1. Halperin I; Ma B; Wolfson H; Nussinov R (June 2002). "Principles of docking: An overview of search algorithms and a guide to scoring functions". Proteins. 47 (4): 409–443. doi:10.1002/prot.10115. PMID   12001221. S2CID   7679220.
  2. Mustard D; Ritchie DW (August 2005). "Docking essential dynamics eigenstructures". Proteins. 60 (2): 269–274. CiteSeerX   10.1.1.134.7903 . doi:10.1002/prot.20569. PMID   15981272. S2CID   15889592.
  3. Shoichet BK; Stroud RM; Santi DV; Kuntz ID; Perry KM (March 1993). "Structure-based discovery of inhibitors of thymidylate synthase". Science. 259 (5100): 1445–50. Bibcode:1993Sci...259.1445S. doi:10.1126/science.8451640. PMID   8451640.
  4. McGann MR; Almond HR; Nicholls A; Grant JA; Brown FK (January 2003). "Gaussian docking functions". Biopolymers. 68 (1): 76–90. CiteSeerX   10.1.1.115.8784 . doi:10.1002/bip.10207. PMID   12579581.
  5. Friesner RA; Banks JL; Murphy RB; Halgren TA; Klicic JJ; Mainz DT; Repasky MP; Knoll EH; Shelley M; Perry JK; Shaw DE; Francis P; Shenkin PS (March 2004). "Glide: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy". J. Med. Chem. 47 (7): 1739–1749. doi:10.1021/jm0306430. PMID   15027865.
  6. Jain AN (February 2003). "Surflex: fully automatic flexible molecular docking using a molecular similarity-based search engine". J. Med. Chem. 46 (4): 499–511. doi:10.1021/jm020406h. PMID   12570372.
  7. Zsoldos Z; Reid D; Simon A; Sadjad SB; Johnson AP (July 2007). "eHiTS: a new fast, exhaustive flexible ligand docking system". J. Mol. Graph. Model. 26 (1): 198–212. doi:10.1016/j.jmgm.2006.06.002. PMID   16860582.
  8. Jones G; Willett P; Glen RC; Leach AR; Taylor R (April 1997). "Development and validation of a genetic algorithm for flexible docking". J. Mol. Biol. 267 (3): 727–748. CiteSeerX   10.1.1.130.3377 . doi:10.1006/jmbi.1996.0897. PMID   9126849.
  9. Goodsell DS; Morris GM; Olson AJ (1996). "Automated docking of flexible ligands: applications of AutoDock". J. Mol. Recognit. 9 (1): 1–5. doi: 10.1002/(SICI)1099-1352(199601)9:1<1::AID-JMR241>3.0.CO;2-6 . PMID   8723313.