12 research outputs found

    Computational Fragment-Based Binding Site Identification by Ligand Competitive Saturation

    Get PDF
    Fragment-based drug discovery using NMR and x-ray crystallographic methods has proven utility but also non-trivial time, materials, and labor costs. Current computational fragment-based approaches circumvent these issues but suffer from limited representations of protein flexibility and solvation effects, leading to difficulties with rigorous ranking of fragment affinities. To overcome these limitations we describe an explicit solvent all-atom molecular dynamics methodology (SILCS: Site Identification by Ligand Competitive Saturation) that uses small aliphatic and aromatic molecules plus water molecules to map the affinity pattern of a protein for hydrophobic groups, aromatic groups, hydrogen bond donors, and hydrogen bond acceptors. By simultaneously incorporating ligands representative of all these functionalities, the method is an in silico free energy-based competition assay that generates three-dimensional probability maps of fragment binding (FragMaps) indicating favorable fragment∶protein interactions. Applied to the two-fold symmetric oncoprotein BCL-6, the SILCS method yields two-fold symmetric FragMaps that recapitulate the crystallographic binding modes of the SMRT and BCOR peptides. These FragMaps account both for important sequence and structure differences in the C-terminal halves of the two peptides and also the high mobility of the BCL-6 His116 sidechain in the peptide-binding groove. Such SILCS FragMaps can be used to qualitatively inform the design of small-molecule inhibitors or as scoring grids for high-throughput in silico docking that incorporate both an atomic-level description of solvation and protein flexibility

    In Silico Identification of a ÎČ2-Adrenoceptor Allosteric Site That Selectively Augments Canonical ÎČ2AR-Gs Signaling and Function

    Get PDF
    Activation of ÎČ2-adrenoceptors (ÎČ2ARs) causes airway smooth muscle (ASM) relaxation and bronchodilation, and ÎČ2AR agonists (ÎČ-agonists) are front-line treatments for asthma and other obstructive lung diseases. However, the therapeutic efficacy of ÎČ-agonists is limited by agonist-induced ÎČ2AR desensitization and noncanonical ÎČ2AR signaling involving ÎČ-arrestin that is shown to promote asthma pathophysiology. Accordingly, we undertook the identification of an allosteric site on ÎČ2AR that could modulate the activity of ÎČ-agonists to overcome these limitations. We employed the site identification by ligand competitive saturation (SILCS) computational method to comprehensively map the entire 3D structure of in silico-generated ÎČ2AR intermediate conformations and identified a putative allosteric binding site. Subsequent database screening using SILCS identified drug-like molecules with the potential to bind to the site. Experimental assays in HEK293 cells (expressing recombinant wild-type human ÎČ2AR) and human ASM cells (expressing endogenous ÎČ2AR) identified positive and negative allosteric modulators (PAMs and NAMs) of ÎČ2AR as assessed by regulation of ÎČ-agonist-stimulation of cyclic AMP generation. PAMs/NAMs had no effect on ÎČ-agonist-induced recruitment of ÎČ-arrestin to ÎČ2AR- or ÎČ-agonist-induced loss of cell surface expression in HEK293 cells expressing ÎČ2AR. Mutagenesis analysis of ÎČ2AR confirmed the SILCS identified site based on mutants of amino acids R131, Y219, and F282. Finally, functional studies revealed augmentation of ÎČ-agonist-induced relaxation of contracted human ASM cells and bronchodilation of contracted airways. These findings identify a allosteric binding site on the ÎČ2AR, whose activation selectively augments ÎČ-agonist-induced Gs signaling, and increases relaxation of ASM cells, the principal therapeutic effect of ÎČ-agonists

    Next generation 3D pharmacophore modeling

    Get PDF
    3D pharmacophore models are three‐dimensional ensembles of chemically defined interactions of a ligand in its bioactive conformation. They represent an elegant way to decipher chemically encoded ligand information and have therefore become a valuable tool in drug design. In this review, we provide an overview on the basic concept of this method and summarize key studies for applying 3D pharmacophore models in virtual screening and mechanistic studies for protein functionality. Moreover, we discuss recent developments in the field. The combination of 3D pharmacophore models with molecular dynamics simulations could be a quantum leap forward since these approaches consider macromolecule–ligand interactions as dynamic and therefore show a physiologically relevant interaction pattern. Other trends include the efficient usage of 3D pharmacophore information in machine learning and artificial intelligence applications or freely accessible web servers for 3D pharmacophore modeling. The recent developments show that 3D pharmacophore modeling is a vibrant field with various applications in drug discovery and beyond

    Solvents to fragments to drugs: MD applications in drug design

    Get PDF
    Simulations of molecular dynamics (MD) are playing an increasingly important role in structure-based drug discovery (SBDD). Here we review the use of MD for proteins in aqueous solvation, organic/aqueous mixed solvents (MDmix) and with small ligands, to the classic SBDD problems: Binding mode and binding free energy predictions. The simulation of proteins in their condensed state reveals solvent structures and preferential interaction sites (hot spots) on the protein surface. The information provided by water and its cosolvents can be used very effectively to understand protein ligand recognition and to improve the predictive capability of well-established methods such as molecular docking. The application of MD simulations to the study of the association of proteins with drug-like compounds is currently only possible for specific cases, as it remains computationally very expensive and labor intensive. MDmix simulations on the other hand, can be used systematically to address some of the common tasks in SBDD. With the advent of new tools and faster computers we expect to see an increase in the application of mixed solvent MD simulations to a plethora of protein targets to identify new drug candidates.Fil: Defelipe, Lucas Alfredo. Universidad de Buenos Aires; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas. Oficina de CoordinaciĂłn Administrativa Ciudad Universitaria. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales; ArgentinaFil: Arcon, Juan Pablo. Universidad de Buenos Aires; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas. Oficina de CoordinaciĂłn Administrativa Ciudad Universitaria. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales; ArgentinaFil: Modenutti, Carlos Pablo. Universidad de Buenos Aires; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas. Oficina de CoordinaciĂłn Administrativa Ciudad Universitaria. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales; ArgentinaFil: Marti, Marcelo Adrian. Universidad de Buenos Aires; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas. Oficina de CoordinaciĂłn Administrativa Ciudad Universitaria. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales; ArgentinaFil: Turjanski, Adrian. Universidad de Buenos Aires; Argentina. Consejo Nacional de Investigaciones CientĂ­ficas y TĂ©cnicas. Oficina de CoordinaciĂłn Administrativa Ciudad Universitaria. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de QuĂ­mica BiolĂłgica de la Facultad de Ciencias Exactas y Naturales; ArgentinaFil: Barril, Xavier. Institucio Catalana de Recerca I Estudis Avancats

    Solvents to Fragments to Drugs: MD Applications in Drug Design

    Get PDF
    Simulations of molecular dynamics (MD) are playing an increasingly important role in structure-based drug discovery (SBDD). Here we review the use of MD for proteins in aqueous solvation, organic/aqueous mixed solvents (MDmix) and with small ligands, to the classic SBDD problems: Binding mode and binding free energy predictions. The simulation of proteins in their condensed state reveals solvent structures and preferential interaction sites (hot spots) on the protein surface. The information provided by water and its cosolvents can be used very effectively to understand protein ligand recognition and to improve the predictive capability of well-established methods such as molecular docking. The application of MD simulations to the study of the association of proteins with drug-like compounds is currently only possible for specific cases, as it remains computationally very expensive and labor intensive. MDmix simulations on the other hand, can be used systematically to address some of the common tasks in SBDD. With the advent of new tools and faster computers we expect to see an increase in the application of mixed solvent MD simulations to a plethora of protein targets to identify new drug candidates

    Investigating Cryptic Binding Sites by Molecular Dynamics Simulations

    Get PDF
    This Account highlights recent advances and discusses major challenges in investigations of cryptic (hidden) binding sites by molecular simulations. Cryptic binding sites are not visible in protein targets crystallized without a ligand and only become visible crystallographically upon binding events. These sites have been shown to be druggable and might provide a rare opportunity to target difficult proteins. However, due to their hidden nature, they are difficult to find through experimental screening. Computational methods based on atomistic molecular simulations remain one of the best approaches to identify and characterize cryptic binding sites. However, not all methods are equally efficient. Some are more apt at quickly probing protein dynamics but do not provide thermodynamic or druggability information, while others that are able to provide such data are demanding in terms of time and resources. Here, we review the recent contributions of mixed-solvent simulations, metadynamics, Markov state models, and other enhanced sampling methods to the field of cryptic site identification and characterization. We discuss how these methods were able to provide precious information on the nature of the site opening mechanisms, to predict previously unknown sites which were used to design new ligands, and to compute the free energy landscapes and kinetics associated with the opening of the sites and the binding of the ligands. We highlight the potential and the importance of such predictions in drug discovery, especially for difficult (“undruggable”) targets. We also discuss the major challenges in the field and their possible solutions

    Computational Modeling of Protein Structure, Function, and Binding Hotspots

    Full text link
    Mixed-solvent molecular dynamics (MixMD) is a cosolvent mapping technique for structure-based drug design. MixMD simulations are performed with a solvent mixture of small molecule probes and water, which directly compete for binding to the protein’s surface. MixMD has previously been shown to identify active and allosteric sites based on the time-averaged occupancy of the probe molecules over the course of the simulation. Sites with the highest maximal occupancy identified known biologically relevant sites for a wide range of targets. This is consistent with previous experimental work identifying hotspots on protein surfaces based on the occupancy of multiple organic-solvent molecules. However, previous MixMD analysis required extensive manual interpretation to identify and rank sites. MixMD Probeview was introduced to automate this analysis, thereby facilitating the application of MixMD. Implemented as a plugin for the freely available, open-source version of PyMOL, MixMD Probeview successfully identified binding sites for several test systems using three different cosolvent simulation procedures. Following identification of binding sites, the occupancy maps from the MixMD simulations can be converted into pharmacophore models for prospective screening of inhibitors. We have developed a pharmacophore generation procedure to convert MixMD occupancy maps into pharmacophore models. Validation of this procedure on ABL kinase showed good performance. Additionally, we have identified characteristic occupancy levels for non-displaceable water molecules so that these sites may be incorporated into structure-based drug design efforts. Lastly, we have explored the potential for accelerated sampling methods to be used in tandem with MixMD to simultaneously capture conformational changes while mapping favorable interactions within binding sites. These developments greatly extend the utility of MixMD while also simplifying its application. In addition, two exploratory studies were completed. First, traditional MD simulations were performed to understand the dynamics of NSD1. Crystal structures of NSD1 capture the post-SET loop in an autoinhibitory position. MD simulations allow conformational sampling of this loop, yielding insight into its dynamic behavior in solution. Second, an epidemiological study was conducted which was aimed at understanding the transmission and sequence variation of CTX-M-type ÎČ-lactamases, in fulfillment of the clinical research component of the MICHR Translational Research Education Certificate.PHDBiophysicsUniversity of Michigan, Horace H. Rackham School of Graduate Studieshttps://deepblue.lib.umich.edu/bitstream/2027.42/138744/1/sarahgra_1.pd

    3D QSAR Pharmacophore Modeling, in Silico Screening, and Density Functional Theory (DFT) Approaches for Identification of Human Chymase Inhibitors

    Get PDF
    Human chymase is a very important target for the treatment of cardiovascular diseases. Using a series of theoretical methods like pharmacophore modeling, database screening, molecular docking and Density Functional Theory (DFT) calculations, an investigation for identification of novel chymase inhibitors, and to specify the key factors crucial for the binding and interaction between chymase and inhibitors is performed. A highly correlating (r = 0.942) pharmacophore model (Hypo1) with two hydrogen bond acceptors, and three hydrophobic aromatic features is generated. After successfully validating “Hypo1”, it is further applied in database screening. Hit compounds are subjected to various drug-like filtrations and molecular docking studies. Finally, three structurally diverse compounds with high GOLD fitness scores and interactions with key active site amino acids are identified as potent chymase hits. Moreover, DFT study is performed which confirms very clear trends between electronic properties and inhibitory activity (IC50) data thus successfully validating “Hypo1” by DFT method. Therefore, this research exertion can be helpful in the development of new potent hits for chymase. In addition, the combinational use of docking, orbital energies and molecular electrostatic potential analysis is also demonstrated as a good endeavor to gain an insight into the interaction between chymase and inhibitors

    Enumeration, conformation sampling and population of libraries of peptide macrocycles for the search of chemotherapeutic cardioprotection agents

    Get PDF
    Peptides are uniquely endowed with features that allow them to perturb previously difficult to drug biomolecular targets. Peptide macrocycles in particular have seen a flurry of recent interest due to their enhanced bioavailability, tunability and specificity. Although these properties make them attractive hit-candidates in early stage drug discovery, knowing which peptides to pursue is non‐trivial due to the magnitude of the peptide sequence space. Computational screening approaches show promise in their ability to address the size of this search space but suffer from their inability to accurately interrogate the conformational landscape of peptide macrocycles. We developed an in‐silico compound enumerator that was tasked with populating a conformationally laden peptide virtual library. This library was then used in the search for cardio‐protective agents (that may be administered, reducing tissue damage during reperfusion after ischemia (heart attacks)). Our enumerator successfully generated a library of 15.2 billion compounds, requiring the use of compression algorithms, conformational sampling protocols and management of aggregated compute resources in the context of a local cluster. In the absence of experimental biophysical data, we performed biased sampling during alchemical molecular dynamics simulations in order to observe cyclophilin‐D perturbation by cyclosporine A and its mitochondrial targeted analogue. Reliable intermediate state averaging through a WHAM analysis of the biased dynamic pulling simulations confirmed that the cardio‐protective activity of cyclosporine A was due to its mitochondrial targeting. Paralleltempered solution molecular dynamics in combination with efficient clustering isolated the essential dynamics of a cyclic peptide scaffold. The rapid enumeration of skeletons from these essential dynamics gave rise to a conformation laden virtual library of all the 15.2 Billion unique cyclic peptides (given the limits on peptide sequence imposed). Analysis of this library showed the exact extent of physicochemical properties covered, relative to the bare scaffold precursor. Molecular docking of a subset of the virtual library against cyclophilin‐D showed significant improvements in affinity to the target (relative to cyclosporine A). The conformation laden virtual library, accessed by our methodology, provided derivatives that were able to make many interactions per peptide with the cyclophilin‐D target. Machine learning methods showed promise in the training of Support Vector Machines for synthetic feasibility prediction for this library. The synergy between enumeration and conformational sampling greatly improves the performance of this library during virtual screening, even when only a subset is used
    corecore