862 research outputs found

    Representability of algebraic topology for biomolecules in machine learning based scoring and virtual screening

    Full text link
    This work introduces a number of algebraic topology approaches, such as multicomponent persistent homology, multi-level persistent homology and electrostatic persistence for the representation, characterization, and description of small molecules and biomolecular complexes. Multicomponent persistent homology retains critical chemical and biological information during the topological simplification of biomolecular geometric complexity. Multi-level persistent homology enables a tailored topological description of inter- and/or intra-molecular interactions of interest. Electrostatic persistence incorporates partial charge information into topological invariants. These topological methods are paired with Wasserstein distance to characterize similarities between molecules and are further integrated with a variety of machine learning algorithms, including k-nearest neighbors, ensemble of trees, and deep convolutional neural networks, to manifest their descriptive and predictive powers for chemical and biological problems. Extensive numerical experiments involving more than 4,000 protein-ligand complexes from the PDBBind database and near 100,000 ligands and decoys in the DUD database are performed to test respectively the scoring power and the virtual screening power of the proposed topological approaches. It is demonstrated that the present approaches outperform the modern machine learning based methods in protein-ligand binding affinity predictions and ligand-decoy discrimination

    Connectable Components for Protein Design

    Get PDF
    Protein design requires reusable, trustworthy, and connectable parts in order to scale to complex challenges. The recent explosion of protein structures stored within the Protein Data Bank provides a wealth of small motifs we can harvest, but we still lack tools to combine them into larger proteins. Here I explore two approaches for connecting reusable protein components on two different length scales. On the atomic scale, I build an interactive search engine for connecting chemical fragments together. Protein fragments built using this search engine recapitulate native-like protein assemblies that can be integrated into existing protein scaffolds using backbone search engines such as MaDCaT. On the protein domain scale, I quantitatively dissect structural variations in two-component systems in order to extract general principles for engineering interfacial flexibility between modular four-helix bundles. These bundles exhibit large scissoring motions where helices move towards or away from the bundle axis and these motions propagate across domain boundaries. Together, these two approaches form the beginnings of a multiscale methodology for connecting reusable protein fragments where there is a constant interplay and feedback between design of atomic structure, secondary structure, and tertiary structure. Rapid iteration, visualization, and search glue these diverse length scales together into a cohesive whole

    Machine Learning for Kinase Drug Discovery

    Get PDF
    Cancer is one of the major public health issues, causing several million losses every year. Although anti-cancer drugs have been developed and are globally administered, mild to severe side effects are known to occur during treatment. Computer-aided drug discovery has become a cornerstone for unveiling treatments of existing as well as emerging diseases. Computational methods aim to not only speed up the drug design process, but to also reduce time-consuming, costly experiments, as well as in vivo animal testing. In this context, over the last decade especially, deep learning began to play a prominent role in the prediction of molecular activity, property and toxicity. However, there are still major challenges when applying deep learning models in drug discovery. Those challenges include data scarcity for physicochemical tasks, the difficulty of interpreting the prediction made by deep neural networks, and the necessity of open-source and robust workflows to ensure reproducibility and reusability. In this thesis, after reviewing the state-of-the-art in deep learning applied to virtual screening, we address the previously mentioned challenges as follows: Regarding data scarcity in the context of deep learning applied to small molecules, we developed data augmentation techniques based on the SMILES encoding. This linear string notation enumerates the atoms present in a compound by following a path along the molecule graph. Multiplicity of SMILES for a single compound can be reached by traversing the graph using different paths. We applied the developed augmentation techniques to three different deep learning models, including convolutional and recurrent neural networks, and to four property and activity data sets. The results show that augmentation improves the model accuracy independently of the deep learning model, as well as of the data set size. Moreover, we computed the uncertainty of a model by using augmentation at inference time. In this regard, we have shown that the more confident the model is in its prediction, the smaller is the error, implying that a given prediction can be trusted and is close to the target value. The software and associated documentation allows making predictions for novel compounds and have been made freely available. Trusting predictions blindly from algorithms may have serious consequences in areas of healthcare. In this context, better understanding how a neural network classifies a compound based on its input features is highly beneficial by helping to de-risk and optimize compounds. In this research project, we decomposed the inner layers of a deep neural network to identify the toxic substructures, the toxicophores, of a compound that led to the toxicity classification. Using molecular fingerprints —vectors that indicate the presence or absence of a particular atomic environment —we were able to map a toxicity score to each of these substructures. Moreover, we developed a method to visualize in 2D the toxicophores within a compound, the so- called cytotoxicity maps, which could be of great use to medicinal chemists in identifying ways to modify molecules to eliminate toxicity. Not only does the deep learning model reach state-of-the-art results, but the identified toxicophores confirm known toxic substructures, as well as expand new potential candidates. In order to speed up the drug discovery process, the accessibility to robust and modular workflows is extremely advantageous. In this context, the fully open-source TeachOpenCADD project was developed. Significant tasks in both cheminformatics and bioinformatics are implemented in a pedagogical fashion, allowing the material to be used for teaching as well as the starting point for novel research. In this framework, a special pipeline is dedicated to kinases, a family of proteins which are known to be involved in diseases such as cancer. The aim is to gain insights into off-targets, i.e. proteins that are unintentionally affected by a compound, and that can cause adverse effects in treatments. Four measures of kinase similarity are implemented, taking into account sequence, and structural information, as well as protein-ligand interaction, and ligand profiling data. The workflow provides clustering of a set of kinases, which can be further analyzed to understand off-target effects of inhibitors. Results show that analyzing kinases using several perspectives is crucial for the insight into off-target prediction, and gaining a global perspective of the kinome. These novel methods can be exploited in the discovery of new drugs, and more specifically diseases involved in the dysregulation of kinases, such as cancer

    Leveraging Structural Flexibility to Predict Protein Function

    Get PDF
    Proteins are essentially versatile and flexible molecules and understanding protein function plays a fundamental role in understanding biological systems. Protein structure comparisons are widely used for revealing protein function. However,with rigidity or partial rigidity assumption, most existing comparison methods do not consider conformational flexibility in protein structures. To address this issue, this thesis seeks to develop algorithms for flexible structure comparisons to predict one specific aspect of protein function, binding specificity. Given conformational samples as flexibility representation, we focus on two predictive problems related to specificity: aggregate prediction and individual prediction.For aggregate prediction, we have designed FAVA (Flexible Aggregate Volumetric Analysis). FAVA is the first conformationally general method to compare proteins with identical folds but different specificities. FAVA is able to correctly categorize members of protein superfamilies and to identify influential amino acids that cause different specificities. A second method PEAP (Point-based Ensemble for Aggregate Prediction) employs ensemble clustering techniques from many base clustering to predict binding specificity. This method incorporates structural motions of functional substructures and is capable of mitigating prediction errors.For individual prediction, the first method is an atomic point representation for representing flexibilities in the binding cavity. This representation is able to predict binding specificity on each protein conformation with high accuracy, and it is the first to analyze maps of binding cavity conformations that describe proteins with different specificities. Our second method introduces a volumetric lattice representation. This representation localizes solvent-accessible shape of the binding cavity by computing cavity volume in each user-defined space. It proves to be more informative than point-based representations. Last but not least, we discuss a structure-independent representation. This representation builds a lattice model on protein electrostatic isopotentials. This is the first known method to predict binding specificity explicitly from the perspective of electrostatic fields.The methods presented in this thesis incorporate the variety of protein conformations into the analysis of protein ligand binding, and provide more views on flexible structure comparisons and structure-based function annotation of molecular design

    Structure based drug design for the discovery of promising inhibitors of human Bcl-2 and Streptococcus dysgalactiae LytR proteins

    Get PDF
    Drug research has evolved significantly in the last decades toward the concept of the rational design of drugs. The capability to study molecular interactions at the atomic level and to rationalize this knowledge to construct and improve drug candidates provided the premises of structure-based drug design (SBDD). This approach allied to the computational methods available nowadays yields the opportunity to expedite the intricate process of drug discovery. In the present thesis, the SBDD approach was implemented to study promising candidate inhibitors of the human Bcl-2 and the Streptococcus dysgalactiae LytR proteins. Half of the cancers in humans are estimated to be related with overexpression of Bcl-2 protein. This macromolecule is responsible for the inhibition of the apoptotic process, which is pivotal for the elimination of abnormal cells. When Bcl-2 is overexpressed, these abnormal cells don’t respond to death stimuli, either endogenous or exogenous, such as chemotherapeutic, and become immortal. Promising 4H-chromene and indole derivatives were studied regarding their potential to inhibit Bcl-2. Molecular docking studies revealed sub-micromolar binding of the 4Hchromene activemethine and the indole derivatives in the binding groove essential for Bcl-2 biological function. Biophysical characterization did not demonstrate significant evidence of binding between Bcl-2 and the compounds under study, probably due to their small network of interactions with the binding pocket residues. The structure determination process of the proteinligand complexes achieved preliminary co-crystallization conditions that require further optimization. Numerous infectious diseases are associated to the bacterial biofilm phenotype, which consists of agglomerates of cells enclosed in a self-produced matrix. Biofilms confer bacteria improved resistance to the host’s innate immune system and to conventional antibiotics. LytR belongs to the LCP family of proteins, which are thought to be responsible for the attachment of anionic polymers to the peptidoglycan, protecting the Gram-positive bacteria from phagocytosis and lysis. Previous virtual screening studies yielded ellagic acid and fisetin has promising inhibitors of LytR, displaying anti-biofilm activity. Molecular docking revealed binding of these compounds in the hypothetical active site of LytR, with micromolar affinities, and specific interactions with crucial protein residues for catalysis. Biophysical techniques failed to provide evidence of protein-ligand interactions, although this may be related to the possible co-purification with a lipidic substrate, which has been reported before. Mass spectrometry or structural determination, through X-ray crystallography or NMR, should be pivotal to establish evidence of this molecule’s accommodation in the binding pocket

    Genome-wide Protein-chemical Interaction Prediction

    Get PDF
    The analysis of protein-chemical reactions on a large scale is critical to understanding the complex interrelated mechanisms that govern biological life at the cellular level. Chemical proteomics is a new research area aimed at genome-wide screening of such chemical-protein interactions. Traditional approaches to such screening involve in vivo or in vitro experimentation, which while becoming faster with the application of high-throughput screening technologies, remains costly and time-consuming compared to in silico methods. Early in silico methods are dependant on knowing 3D protein structures (docking) or knowing binding information for many chemicals (ligand-based approaches). Typical machine learning approaches follow a global classification approach where a single predictive model is trained for an entire data set, but such an approach is unlikely to generalize well to the protein-chemical interaction space considering its diversity and heterogeneous distribution. In response to the global approach, work on local models has recently emerged to improve generalization across the interaction space by training a series of independant models localized to each predict a single interaction. This work examines current approaches to genome-wide protein-chemical interaction prediction and explores new computational methods based on modifications to the boosting framework for ensemble learning. The methods are described and compared to several competing classification methods. Genome-wide chemical-protein interaction data sets are acquired from publicly available resources, and a series of experimental studies are performed in order to compare the the performance of each method under a variety of conditions

    Allo-network drugs: Extension of the allosteric drug concept to protein-protein interaction and signaling networks

    Get PDF
    Allosteric drugs are usually more specific and have fewer side effects than orthosteric drugs targeting the same protein. Here, we overview the current knowledge on allosteric signal transmission from the network point of view, and show that most intra-protein conformational changes may be dynamically transmitted across protein-protein interaction and signaling networks of the cell. Allo-network drugs influence the pharmacological target protein indirectly using specific inter-protein network pathways. We show that allo-network drugs may have a higher efficiency to change the networks of human cells than those of other organisms, and can be designed to have specific effects on cells in a diseased state. Finally, we summarize possible methods to identify allo-network drug targets and sites, which may develop to a promising new area of systems-based drug design
    • …
    corecore