28 research outputs found

    Linking Proteomic and Transcriptional Data through the Interactome and Epigenome Reveals a Map of Oncogene-induced Signaling

    Get PDF
    Cellular signal transduction generally involves cascades of post-translational protein modifications that rapidly catalyze changes in protein-DNA interactions and gene expression. High-throughput measurements are improving our ability to study each of these stages individually, but do not capture the connections between them. Here we present an approach for building a network of physical links among these data that can be used to prioritize targets for pharmacological intervention. Our method recovers the critical missing links between proteomic and transcriptional data by relating changes in chromatin accessibility to changes in expression and then uses these links to connect proteomic and transcriptome data. We applied our approach to integrate epigenomic, phosphoproteomic and transcriptome changes induced by the variant III mutation of the epidermal growth factor receptor (EGFRvIII) in a cell line model of glioblastoma multiforme (GBM). To test the relevance of the network, we used small molecules to target highly connected nodes implicated by the network model that were not detected by the experimental data in isolation and we found that a large fraction of these agents alter cell viability. Among these are two compounds, ICG-001, targeting CREB binding protein (CREBBP), and PKF118–310, targeting β-catenin (CTNNB1), which have not been tested previously for effectiveness against GBM. At the level of transcriptional regulation, we used chromatin immunoprecipitation sequencing (ChIP-Seq) to experimentally determine the genome-wide binding locations of p300, a transcriptional co-regulator highly connected in the network. Analysis of p300 target genes suggested its role in tumorigenesis. We propose that this general method, in which experimental measurements are used as constraints for building regulatory networks from the interactome while taking into account noise and missing data, should be applicable to a wide range of high-throughput datasets.National Science Foundation (U.S.) (DB1-0821391)National Institutes of Health (U.S.) (Grant U54-CA112967)National Institutes of Health (U.S.) (Grant R01-GM089903)National Institutes of Health (U.S.) (P30-ES002109

    Probing the chemical–biological relationship space with the Drug Target Explorer

    No full text
    Abstract Modern phenotypic high-throughput screens (HTS) present several challenges including identifying the target(s) that mediate the effect seen in the screen, characterizing ‘hits’ with a polypharmacologic target profile, and contextualizing screen data within the large space of drugs and screening models. To address these challenges, we developed the Drug–Target Explorer. This tool allows users to query molecules within a database of experimentally-derived and curated compound-target interactions to identify structurally similar molecules and their targets. It enables network-based visualizations of the compound-target interaction space, and incorporates comparisons to publicly-available in vitro HTS datasets. Furthermore, users can identify molecules using a query target or set of targets. The Drug Target Explorer is a multifunctional platform for exploring chemical space as it relates to biological targets, and may be useful at several steps along the drug development pipeline including target discovery, structure–activity relationship, and lead compound identification studies

    Network-Based Interpretation of Diverse High-Throughput Datasets through the Omics Integrator Software Package.

    Get PDF
    High-throughput, 'omic' methods provide sensitive measures of biological responses to perturbations. However, inherent biases in high-throughput assays make it difficult to interpret experiments in which more than one type of data is collected. In this work, we introduce Omics Integrator, a software package that takes a variety of 'omic' data as input and identifies putative underlying molecular pathways. The approach applies advanced network optimization algorithms to a network of thousands of molecular interactions to find high-confidence, interpretable subnetworks that best explain the data. These subnetworks connect changes observed in gene expression, protein abundance or other global assays to proteins that may not have been measured in the screens due to inherent bias or noise in measurement. This approach reveals unannotated molecular pathways that would not be detectable by searching pathway databases. Omics Integrator also provides an elegant framework to incorporate not only positive data, but also negative evidence. Incorporating negative evidence allows Omics Integrator to avoid unexpressed genes and avoid being biased toward highly-studied hub proteins, except when they are strongly implicated by the data. The software is comprised of two individual tools, Garnet and Forest, that can be run together or independently to allow a user to perform advanced integration of multiple types of high-throughput data as well as create condition-specific subnetworks of protein interactions that best connect the observed changes in various datasets. It is available at http://fraenkel.mit.edu/omicsintegrator and on GitHub at https://github.com/fraenkel-lab/OmicsIntegrator

    Anticipated results: In silico EGFR knock-out experiment in network modeling.

    No full text
    <p>Blue nodes represent ‘Steiner nodes’ that were not measured as changing in the original experiment but are identified through network reconstruction; yellow nodes represent ‘terminal nodes’ that are the phosphoproteomic hits. The original network and the network with EGFR knock-out have been merged to clearly show the common and different nodes and edges in the two conditions. Common edges in two conditions are black lines, edges only present in EGFR knock-out condition are red dotted lines and edges only present in the wild-type condition are blue dashed lines. Cell surface receptors are arrow-shaped. The parameters are μ = 0.002, ω = 2, β = 150, and D = 10.</p

    Summary of features differentiating Omics Integrator from existing tools and which features are available when Garnet and Forest are used individually.

    No full text
    <p><sup>1</sup> Some network algorithms model TFs by including protein-DNA interactions in the network or generating TF scores for the protein nodes. <sup>2</sup> Some network algorithms optimize the transmission of information from source nodes to target nodes and require the sources to be identified in advance. <sup>3</sup> Time series analysis algorithms require omic data from three or more time points. <sup>4</sup> Intermediate proteins, like the Steiner nodes predicted by Forest, are not assigned condition-specific scores but are important for connecting other scored nodes in the subnetwork. <sup>5</sup> Negative evidence discourages network algorithms from selecting particular nodes due to prior knowledge or a bias, such as node degree.</p

    The final PCSF reconstructed from the terminal set formed by the members of mRNA splicing pathway, pyruvate metabolism pathway, and Rho cell motility pathway in ConsensusPathDB.

    No full text
    <p>Each node is colored according to the pathway to which it belongs, and Steiner nodes are colored gray. The parameters are μ = 0.009, ω = 3, β = 5, and D = 5.</p

    The flowchart of the software.

    No full text
    <p>Step 1 requires downloading and unzipping the scripts and data files. Step 2 consists of the installation of the necessary tools to run Omics Integrator. Step 3 describes how to prepare input files. Step 4 and 5 are designed for data collection and formatting for Garnet and Forest modules, respectively. At Step 6, configuration files are prepared where parameters are defined for Garnet and Forest separately. Garnet and Forest scripts are run at Step 7. If the initial data contains transcriptional data, then Garnet must be run before Forest. Otherwise Forest can be run independently. Detailed instructions of these steps are in the ‘Procedure’ section of the <a href="http://www.ploscompbiol.org/article/info:doi/10.1371/journal.pcbi.1004879#pcbi.1004879.s001" target="_blank">S1 Text</a>.</p

    Anticipated results: Network reconstructed from changes in phosphoproteomic measurements (circles) and gene expression measurements (triangles) in lung cancer cell lines stimulated with Tgf-β.

    No full text
    <p>Blue hexagons represent ‘Steiner nodes’ that were not measured as changing in the original experimental measurements but identified through network reconstruction. Nodes that are not blue were measured in the phosphoproteomic data, with color indicating the degree of change in phosphoproteomic measurements: grey indicates no change and yellow indicates a large amount of change. Network robustness was measured by adding noise to the edges using the --noisyEdges flag. The shade of the edge is correlated with the number of times the edge was selected over all perturbations, and the size of a node represents number of times the node was selected. The width of the edge represents the weight assigned to the interaction in the original interactome.</p

    Systems Approaches to Cancer Biology.

    No full text
    Cancer systems biology aims to understand cancer as an integrated system of genes, proteins, networks, and interactions rather than an entity of isolated molecular and cellular components. The inaugural Systems Approaches to Cancer Biology Conference, cosponsored by the Association of Early Career Cancer Systems Biologists and the National Cancer Institute of the NIH, focused on the interdisciplinary field of cancer systems biology and the challenging cancer questions that are best addressed through the combination of experimental and computational analyses. Attendees found that elucidating the many molecular features of cancer inevitably reveals new forms of complexity and concluded that ensuring the reproducibility and impact of cancer systems biology studies will require widespread method and data sharing and, ultimately, the translation of important findings to the clinic. Cancer Res; 76(23); 6774-7. ©2016 AACR
    corecore