1,035 research outputs found

    Cross-reactivity virtual profiling of the human kinome by X-React \u3csup\u3eKIN\u3c/sup\u3e: A chemical systems biology approach

    Get PDF
    Many drug candidates fail in clinical development due to their insufficient selectivity that may cause undesired side effects. Therefore, modern drug discovery is routinely supported by computational techniques, which can identify alternate molecular targets with a significant potential for cross-reactivity. In particular, the development of highly selective kinase inhibitors is complicated by the strong conservation of the ATP-binding site across the kinase family. In this paper, we describe X-ReactKIN, a new machine learning approach that extends the modeling and virtual screening of individual protein kinases to a system level in order to construct a cross-reactivity virtual profile for the human kinome. To maximize the coverage of the kinome, X-ReactKIN relies solely on the predicted target structures and employs state-of-the-art modeling techniques. Benchmark tests carried out against available selectivity data from high-throughput kinase profiling experiments demonstrate that, for almost 70% of the inhibitors, their alternate molecular targets can be effectively identified in the human kinome with a high (\u3e0.5) sensitivity at the expense of a relatively low false positive rate (\u3c0.5). Furthermore, in a case study, we demonstrate how X-React KIN can support the development of selective inhibitors by optimizing the selection of kinase targets for small-scale counter-screen experiments. The constructed cross-reactivity profiles for the human kinome are freely available to the academic community at http://cssb.biology.gatech.edu/kinomelhm/. © 2010 American Chemical Society

    Comprehensive structural and functional characterization of the human kinome by protein structure modeling and ligand virtual screening

    Get PDF
    The growing interest in the identification of kinase inhibitors, promising therapeutics in the treatment of many diseases, has created a demand for the structural characterization of the entire human kinome. At the outset of the drug development process, the lead-finding stage, approaches that enrich the screening library with bioactive compounds are needed. Here, protein structure based methods can play an important role, but despite structural genomics efforts, it is unlikely that the three-dimensional structures of the entire kinome will be available soon. Therefore, at the proteome level, structure-based approaches must rely on predicted models, with a key issue being their utility in virtual ligand screening. In this study, we employ the recently developed FINDSITE/Q-Dock ligand homology modeling approach, which is well-suited for proteome-scale applications using predicted structures, to provide extensive structural and functional characterization of the human kinome. Specifically, we construct structure models for the human kinome; these are subsequently subject to virtual screening against a library of more than 2 million compounds. To rank the compounds, we employ a hierarchical approach that combines ligand- and structure-based filters. Modeling accuracy is carefully validated using available experimental data with particularly encouraging results found for the ability to identify, without prior knowledge, specific kinase inhibitors. More generally, the modeling procedure results in a large number of predicted molecular interactions between kinases and small ligands that should be of practical use in the development of novel inhibitors. The data set is freely available to the academic community via a user-friendly Web interface at http://cssb.biology.gatech.edu/kinomelhm/ as well as at the ZINC Web site (http://zinc.docking.org/applications/2010Apr/Brylinski-2010.tar.gz). © 2010 American Chemical Society

    Structural Cheminformatics for Kinase-Centric Drug Design

    Get PDF
    Drug development is a long, expensive, and iterative process with a high failure rate, while patients wait impatiently for treatment. Kinases are one of the main drug targets studied for the last decades to combat cancer, the second leading cause of death worldwide. These efforts resulted in a plethora of structural, chemical, and pharmacological kinase data, which are collected in the KLIFS database. In this thesis, we apply ideas from structural cheminformatics to the rich KLIFS dataset, aiming to provide computational tools that speed up the complex drug discovery process. We focus on methods for target prediction and fragment-based drug design that study characteristics of kinase binding sites (also called pockets). First, we introduce the concept of computational target prediction, which is vital in the early stages of drug discovery. This approach identifies biological entities such as proteins that may (i) modulate a disease of interest (targets or on-targets) or (ii) cause unwanted side effects due to their similarity to on-targets (off-targets). We focus on the research field of binding site comparison, which lacked a freely available and efficient tool to determine similarities between the highly conserved kinase pockets. We fill this gap with the novel method KiSSim, which encodes and compares spatial and physicochemical pocket properties for all kinases (kinome) that are structurally resolved. We study kinase similarities in the form of kinome-wide phylogenetic trees and detect expected and unexpected off-targets. To allow multiple perspectives on kinase similarity, we propose an automated and production-ready pipeline; user-defined kinases can be inspected complementarily based on their pocket sequence and structure (KiSSim), pocket-ligand interactions, and ligand profiles. Second, we introduce the concept of fragment-based drug design, which is useful to identify and optimize active and promising molecules (hits and leads). This approach identifies low-molecular-weight molecules (fragments) that bind weakly to a target and are then grown into larger high-affinity drug-like molecules. With the novel method KinFragLib, we provide a fragment dataset for kinases (fragment library) by viewing kinase inhibitors as combinations of fragments. Kinases have a highly conserved pocket with well-defined regions (subpockets); based on the subpockets that they occupy, we fragment kinase inhibitors in experimentally resolved protein-ligand complexes. The resulting dataset is used to generate novel kinase-focused molecules that are recombinations of the previously fragmented kinase inhibitors while considering their subpockets. The KinFragLib and KiSSim methods are published as freely available Python tools. Third, we advocate for open and reproducible research that applies FAIR principles ---data and software shall be findable, accessible, interoperable, and reusable--- and software best practices. In this context, we present the TeachOpenCADD platform that contains pipelines for computer-aided drug design. We use open source software and data to demonstrate ligand-based applications from cheminformatics and structure-based applications from structural bioinformatics. To emphasize the importance of FAIR data, we dedicate several topics to accessing life science databases such as ChEMBL, PubChem, PDB, and KLIFS. These pipelines are not only useful to novices in the field to gain domain-specific skills but can also serve as a starting point to study research questions. Furthermore, we show an example of how to build a stand-alone tool that formalizes reoccurring project-overarching tasks: OpenCADD-KLIFS offers a clean and user-friendly Python API to interact with the KLIFS database and fetch different kinase data types. This tool has been used in this thesis and beyond to support kinase-focused projects. We believe that the FAIR-based methods, tools, and pipelines presented in this thesis (i) are valuable additions to the toolbox for kinase research, (ii) provide relevant material for scientists who seek to learn, teach, or answer questions in the realm of computer-aided drug design, and (iii) contribute to making drug discovery more efficient, reproducible, and reusable

    Structure-Based Target-Specific Screening Leads to Small-Molecule CaMKII Inhibitors

    Get PDF
    Target-specific scoring methods are more commonly used to identify small-molecule inhibitors among compounds docked to a target of interest. Top candidates that emerge from these methods have rarely been tested for activity and specificity across a family of proteins. In this study we docked a chemical library into CaMKIIÎŽ, a member of the Ca2+ /calmodulin (CaM)-dependent protein kinase (CaMK) family, and re-scored the resulting protein-compound structures using Support Vector Machine SPecific (SVMSP), a target-specific method that we developed previously. Among the 35 selected candidates, three hits were identified, such as quinazoline compound 1 (KIN-1; N4-[7-chloro-2-[(E)-styryl]quinazolin-4-yl]-N1,N1-diethylpentane-1,4-diamine), which was found to inhibit CaMKIIÎŽ kinase activity at single-digit micromolar IC50 . Activity across the kinome was assessed by profiling analogues of 1, namely 6 (KIN-236; N4-[7-chloro-2-[(E)-2-(2-chloro-4,5-dimethoxyphenyl)vinyl]quinazolin-4-yl]-N1,N1-diethylpentane-1,4-diamine), and an analogue of hit compound 2 (KIN-15; 2-[4-[(E)-[(5-bromobenzofuran-2-carbonyl)hydrazono]methyl]-2-chloro-6-methoxyphenoxy]acetic acid), namely 14 (KIN-332; N-[(E)-[4-(2-anilino-2-oxoethoxy)-3-chlorophenyl]methyleneamino]benzofuran-2-carboxamide), against 337 kinases. Interestingly, for compound 6, CaMKIIÎŽ and homologue CaMKIIÎł were among the top ten targets. Among the top 25 targets of 6, IC50 values ranged from 5 to 22 Όm. Compound 14 was found to be not specific toward CaMKII kinases, but it does inhibit two kinases with sub-micromolar IC50 values among the top 25. Derivatives of 1 were tested against several kinases including several members of the CaMK family. These data afforded a limited structure-activity relationship study. Molecular dynamics simulations with explicit solvent followed by end-point MM-GBSA free-energy calculations revealed strong engagement of specific residues within the ATP binding pocket, and also changes in the dynamics as a result of binding. This work suggests that target-specific scoring approaches such as SVMSP may hold promise for the identification of small-molecule kinase inhibitors that exhibit some level of specificity toward the target of interest across a large number of proteins

    The kinase polypharmacology landscape of clinical PARP inhibitors.

    Get PDF
    Polypharmacology plays an important role in defining response and adverse effects of drugs. For some mechanisms, experimentally mapping polypharmacology is commonplace, although this is typically done within the same protein class. Four PARP inhibitors have been approved by the FDA as cancer therapeutics, yet a precise mechanistic rationale to guide clinicians on which to choose for a particular patient is lacking. The four drugs have largely similar PARP family inhibition profiles, but several differences at the molecular and clinical level have been reported that remain poorly understood. Here, we report the first comprehensive characterization of the off-target kinase landscape of four FDA-approved PARP drugs. We demonstrate that all four PARP inhibitors have a unique polypharmacological profile across the kinome. Niraparib and rucaparib inhibit DYRK1s, CDK16 and PIM3 at clinically achievable, submicromolar concentrations. These kinases represent the most potently inhibited off-targets of PARP inhibitors identified to date and should be investigated further to clarify their potential implications for efficacy and safety in the clinic. Moreover, broad kinome profiling is recommended for the development of PARP inhibitors as PARP-kinase polypharmacology could potentially be exploited to modulate efficacy and side-effect profiles

    Bioinformatics Approaches for Predicting Kinase–Substrate Relationships

    Get PDF
    Protein phosphorylation, catalyzed by protein kinases, is the main posttranslational modification in eukaryotes, regulating essential aspects of cellular function. Using mass spectrometry techniques, a profound knowledge has been achieved in the localization of phosphorylated residues at proteomic scale. Although it is still largely unknown, the protein kinases are responsible for such modifications. To fill this gap, many computational algorithms have been developed, which are capable to predict kinase–substrate relationships. The greatest difficulty for these approaches is to model the complex nature that determines kinase–substrate specificity. The vast majority of predictors is based on the linear primary sequence pattern that surrounds phosphorylation sites. However, in the intracellular environment the protein kinase specificity is influenced by contextual factors, such as protein–protein interactions, substrates co-expression patterns, and subcellular localization. Only recently, the development of phosphorylation predictors has begun to incorporate these variables, significantly improving specificity of these methods. An accurate modeling of kinase–substrate relationships could be the greatest contribution of bioinformatics to understand physiological cell signaling and its pathological impairment

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

    Get PDF

    Analysis of mitotic phosphoproteomes using SILAC based mass spectrometry

    Get PDF

    Selectivity Profiling and Biological Activity of Novel beta-Carbolines as Potent and Selective DYRK1 Kinase Inhibitors

    Get PDF
    DYRK1A is a pleiotropic protein kinase with diverse functions in cellular regulation, including cell cycle control, neuronal differentiation, and synaptic transmission. Enhanced activity and overexpression of DYRK1A have been linked to altered brain development and function in Down syndrome and neurodegenerative diseases such as Alzheimer's disease. The beta-carboline alkaloid harmine is a high affinity inhibitor of DYRK1A but suffers from the drawback of inhibiting monoamine oxidase A (MAO-A) with even higher potency. Here we characterized a series of novel harmine analogs with minimal or absent MAO-A inhibitory activity. We identified several inhibitors with submicromolar potencies for DYRK1A and selectivity for DYRK1A and DYRK1B over the related kinases DYRK2 and HIPK2. An optimized inhibitor, AnnH75, inhibited CLK1, CLK4, and haspin/GSG2 as the only off-targets in a panel of 300 protein kinases. In cellular assays, AnnH75 dose-dependently reduced the phosphorylation of three known DYRK1A substrates (SF3B1, SEPT4, and tau) without negative effects on cell viability. AnnH75 inhibited the cotranslational tyrosine autophosphorylation of DYRK1A and threonine phosphorylation of an exogenous substrate protein with similar potency. In conclusion, we have characterized an optimized beta-carboline inhibitor as a highly selective chemical probe that complies with desirable properties of drug-like molecules and is suitable to interrogate the function of DYRK1A in biological studies
    • 

    corecore