181 research outputs found

    11th German Conference on Chemoinformatics (GCC 2015) : Fulda, Germany. 8-10 November 2015.

    Get PDF

    An enhanced-sampling MD-based protocol for molecular docking

    Get PDF
    Understanding molecular recognition of small molecules by proteins in atomistic detail is key for drug design. Molecular docking is a widely used computational method to mimic ligand-protein association in silico. However, predicting conformational changes occurring in proteins upon ligand binding is still a major challenge. Ensemble docking approaches address this issue by considering a set of different conformations of the protein obtained either experimentally or from computer simulations, e.g. from molecular dynamics. However, holo structures prone to host (the correct) ligands are generally poorly sampled by standard molecular dynamics simulations of the unbound (apo) protein. In order to address this limitation, we introduce a computational approach based on metadynamics simulations called ensemble docking with enhanced sampling of pocket shape (EDES) that allows holo-like conformations of proteins to be generated by exploiting only their apo structures. This is achieved by defining a set of collective variables able to sample different shapes of the binding site, ultimately mimicking the steric effect due to the ligand. In this work, we assessed the method on re-docking and cross-docking calculations. In first case, we selected three different protein targets undergoing different extent of conformational changes upon binding and, for each of them, we docked the experimental ligand conformation into an ensemble of receptor structures generated by EDES. In the second case, in the contest of a blind docking challenge, we generated the 3D structures of a set of different ligands of the same receptor and docked them into a set of EDES-generated conformations of that receptor. In all cases, for both re-docking and cross-docking experiments, our protocol generates a significant fraction of structures featuring a low RMSD from the experimental holo geometry of the receptor. Moreover, ensemble docking calculations using those conformations yielded in almost all cases to native-like poses among the top-ranked ones. Finally, we also tested an improved EDES recipe on a further target, known to be extremely challenging due to its extended binding region and the large extent of conformational changes accompanying the binding of its ligands

    Computational Modeling of Designed Ankyrin Repeat Protein Complexes with their Targets

    Get PDF
    Recombinant therapeutic proteins are playing an ever-increasing role in the clinic. High-affinity binding candidates can be produced in a high-throughput manner through the process of selection and evolution from large libraries, but the structures of the complexes with target protein can only be determined for a small number of them in a costly, low-throughput manner, typically by x-ray crystallography. Reliable modeling of complexes would greatly help to understand their mode of action and improve them by further engineering, for example, by designing bi-paratopic binders. Designed ankyrin repeat proteins (DARPins) are one such class of antibody mimetics that have proven useful in the clinic, in diagnostics and research. Here we have developed a standardized procedure to model DARPin–target complexes that can be used to predict the structures of unknown complexes. It requires only the sequence of a DARPin and a structure of the unbound target. The procedure includes homology modeling of the DARPin, modeling of the flexible parts of a target, rigid body docking to ensembles of the target and docking with a partially flexible backbone. For a set of diverse DARPin–target complexes tested it generated a single model of the complex that well approximates the native state of the complex. We provide a protocol that can be used in a semi-automated way and with tools that are freely available. The presented concepts should help to accelerate the development of novel bio-therapeutics for scaffolds with similar properties

    Structure-based Prediction of Protein-protein Interaction Networks across Proteomes

    Get PDF
    Protein-protein interactions (PPIs) orchestrate virtually all cellular processes, therefore, their exhaustive exploration is essential for the comprehensive understanding of cellular networks. Significant efforts have been devoted to expand the coverage of the proteome-wide interaction space at molecular level. A number of experimental techniques have been developed to discover PPIs, however these approaches have some limitations such as the high costs and long times of experiments, noisy data sets, and often high false positive rate and inter-study discrepancies. Given experimental limitations, computational methods are increasingly becoming important for detection and structural characterization of PPIs. In that regard, we have developed a novel pipeline for high-throughput PPI prediction based on all-to-all rigid body docking of protein structures. We focus on two questions, ‘how do proteins interact?’ and ‘which proteins interact?’. The method combines molecular modeling, structural bioinformatics, machine learning, and functional annotation data to answer these questions and it can be used for genome-wide molecular reconstruction of protein-protein interaction networks. As a proof of concept, 61,913 protein-protein interactions were confidently predicted and modeled for the proteome of E. coli. Further, we validated our method against a few human pathways. The modeling protocol described in this communication can be applied to detect protein-protein interactions in other organisms as well as to construct dimer structures and estimate the confidence of protein interactions experimentally identified with high-throughput techniques

    Systematic Exploitation of Multiple Receptor Conformations for Virtual Ligand Screening

    Get PDF
    The role of virtual ligand screening in modern drug discovery is to mine large chemical collections and to prioritize for experimental testing a comparatively small and diverse set of compounds with expected activity against a target. Several studies have pointed out that the performance of virtual ligand screening can be improved by taking into account receptor flexibility. Here, we systematically assess how multiple crystallographic receptor conformations, a powerful way of discretely representing protein plasticity, can be exploited in screening protocols to separate binders from non-binders. Our analyses encompass 36 targets of pharmaceutical relevance and are based on actual molecules with reported activity against those targets. The results suggest that an ensemble receptor-based protocol displays a stronger discriminating power between active and inactive molecules as compared to its standard single rigid receptor counterpart. Moreover, such a protocol can be engineered not only to enrich a higher number of active compounds, but also to enhance their chemical diversity. Finally, some clear indications can be gathered on how to select a subset of receptor conformations that is most likely to provide the best performance in a real life scenario

    Rational Design of Small-Molecule Inhibitors of Protein-Protein Interactions: Application to the Oncogenic c-Myc/Max Interaction

    Get PDF
    Protein-protein interactions (PPIs) constitute an emerging class of targets for pharmaceutical intervention pursued by both industry and academia. Despite their fundamental role in many biological processes and diseases such as cancer, PPIs are still largely underrepresented in today's drug discovery. This dissertation describes novel computational approaches developed to facilitate the discovery/design of small-molecule inhibitors of PPIs, using the oncogenic c-Myc/Max interaction as a case study.First, we critically review current approaches and limitations to the discovery of small-molecule inhibitors of PPIs and we provide examples from the literature.Second, we examine the role of protein flexibility in molecular recognition and binding, and we review recent advances in the application of Elastic Network Models (ENMs) to modeling the global conformational changes of proteins observed upon ligand binding. The agreement between predicted soft modes of motions and structural changes experimentally observed upon ligand binding supports the view that ligand binding is facilitated, if not enabled, by the intrinsic (pre-existing) motions thermally accessible to the protein in the unliganded form.Third, we develop a new method for generating models of the bioactive conformations of molecules in the absence of protein structure, by identifying a set of conformations (from different molecules) that are most mutually similar in terms of both their shape and chemical features. We show how to solve the problem using an Integer Linear Programming formulation of the maximum-edge weight clique problem. In addition, we present the application of the method to known c-Myc/Max inhibitors.Fourth, we propose an innovative methodology for molecular mimicry design. We show how the structure of the c-Myc/Max complex was exploited to designing compounds that mimic the binding interactions that Max makes with the leucine zipper domain of c-Myc.In summary, the approaches described in this dissertation constitute important contributions to the fields of computational biology and computer-aided drug discovery, which combine biophysical insights and computational methods to expedite the discovery of novel inhibitors of PPIs

    Insights into the Development of Chemotherapeutics Targeting PFKFB Enzymes

    Get PDF
    The PFKFB enzymes control the primary checkpoint in the glycolytic pathway and are implicated in a multitude of diseases: from cancer, to schizophrenia, to diabetes, and heart disease. The inducible isoform, PFKFB3, is known to be associated with the upregulation of glycolysis in many cancers. The first study within this work investigates the potential for using tier-based approaches of virtual screening to target small molecule kinases, with PFKFB3 serving as a case study. For this investigation, bioactive compounds for PFKFB3 were identified from a compound library of 1364 compounds via high-throughput screening, with bioactive compounds being further characterized as either competitive or non-competitive for F6P. Using the F6P-competitive compounds, several structure based docking programs were assessed individually and in conjunction with a pharmacophore screening. The results showed that the tiered virtual screening approach, using pharmacophore screening in addition to structure-based docking, improved enrichments rates in 80% of cases, reduced CPU costs up to 7-fold, and lessened variability among different structure-based docking methods. The second study investigates the structural and kinetic characteristics of citrate inhibition on the heart PFKFB isoenzyme, PFKFB2. High levels of citrate, an intermediate of the TCA cycle, signify an abundance of biosynthetic precursors and that additional glucose need not be degraded for this purpose. Previous studies have noted that citrate acts as an important negative feed-back mechanism to limit glycolytic activity by inhibiting PFKFB enzymes, yet the structural and mechanistic details of citrate’s inhibition had not been determined. To study the molecular basis for citrate inhibition, the three-dimensional structures of the human and bovine PFKFB2 orthologues were solved, each in complex with citrate. For both cases, citrate primarily occupied the binding site of Fructose-6-phosphate (F6P), competitively blocking F6P from binding. Additionally, a carboxy arm of citrate extended into the γ-phosphate binding site of ATP, sterically and electrostatically blocking the catalytic binding mode for ATP. In the human orthologue, which utilized AMPPNP as an ATP analogue, conformational changes were observed in the 2-kinase domain as well as the binding mode for AMPPNP. This study gives new insights as to how the citrate-mediate negative feedback loop influences glycolytic flux through PFKFB enzymes

    Computational strategies to include protein flexibility in Ligand Docking and Virtual Screening

    Get PDF
    The dynamic character of proteins strongly influences biomolecular recognition mechanisms. With the development of the main models of ligand recognition (lock-and-key, induced fit, conformational selection theories), the role of protein plasticity has become increasingly relevant. In particular, major structural changes concerning large deviations of protein backbones, and slight movements such as side chain rotations are now carefully considered in drug discovery and development. It is of great interest to identify multiple protein conformations as preliminary step in a screening campaign. Protein flexibility has been widely investigated, in terms of both local and global motions, in two diverse biological systems. On one side, Replica Exchange Molecular Dynamics has been exploited as enhanced sampling method to collect multiple conformations of Lactate Dehydrogenase A (LDHA), an emerging anticancer target. The aim of this project was the development of an Ensemble-based Virtual Screening protocol, in order to find novel potent inhibitors. On the other side, a preliminary study concerning the local flexibility of Opioid Receptors has been carried out through ALiBERO approach, an iterative method based on Elastic Network-Normal Mode Analysis and Monte Carlo sampling. Comparison of the Virtual Screening performances by using single or multiple conformations confirmed that the inclusion of protein flexibility in screening protocols has a positive effect on the probability to early recognize novel or known active compounds

    IN SILICO METHODS FOR DRUG DESIGN AND DISCOVERY

    Get PDF
    Computer-aided drug design (CADD) methodologies are playing an ever-increasing role in drug discovery that are critical in the cost-effective identification of promising drug candidates. These computational methods are relevant in limiting the use of animal models in pharmacological research, for aiding the rational design of novel and safe drug candidates, and for repositioning marketed drugs, supporting medicinal chemists and pharmacologists during the drug discovery trajectory.Within this field of research, we launched a Research Topic in Frontiers in Chemistry in March 2019 entitled “In silico Methods for Drug Design and Discovery,” which involved two sections of the journal: Medicinal and Pharmaceutical Chemistry and Theoretical and Computational Chemistry. For the reasons mentioned, this Research Topic attracted the attention of scientists and received a large number of submitted manuscripts. Among them 27 Original Research articles, five Review articles, and two Perspective articles have been published within the Research Topic. The Original Research articles cover most of the topics in CADD, reporting advanced in silico methods in drug discovery, while the Review articles offer a point of view of some computer-driven techniques applied to drug research. Finally, the Perspective articles provide a vision of specific computational approaches with an outlook in the modern era of CADD
    corecore