8 research outputs found

    DSL: Discriminative Subgraph Learning via Sparse Self-Representation

    Full text link
    The goal in network state prediction (NSP) is to classify the global state (label) associated with features embedded in a graph. This graph structure encoding feature relationships is the key distinctive aspect of NSP compared to classical supervised learning. NSP arises in various applications: gene expression samples embedded in a protein-protein interaction (PPI) network, temporal snapshots of infrastructure or sensor networks, and fMRI coherence network samples from multiple subjects to name a few. Instances from these domains are typically ``wide'' (more features than samples), and thus, feature sub-selection is required for robust and generalizable prediction. How to best employ the network structure in order to learn succinct connected subgraphs encompassing the most discriminative features becomes a central challenge in NSP. Prior work employs connected subgraph sampling or graph smoothing within optimization frameworks, resulting in either large variance of quality or weak control over the connectivity of selected subgraphs. In this work we propose an optimization framework for discriminative subgraph learning (DSL) which simultaneously enforces (i) sparsity, (ii) connectivity and (iii) high discriminative power of the resulting subgraphs of features. Our optimization algorithm is a single-step solution for the NSP and the associated feature selection problem. It is rooted in the rich literature on maximal-margin optimization, spectral graph methods and sparse subspace self-representation. DSL simultaneously ensures solution interpretability and superior predictive power (up to 16% improvement in challenging instances compared to baselines), with execution times up to an hour for large instances.Comment: 9 page

    Machine Learning Approaches for Breast Cancer Survivability Prediction

    Get PDF
    Breast cancer is one of the leading causes of cancer death in women. If not diagnosed early, the 5-year survival rate of patients is just about 26\%. Furthermore, patients with similar phenotypes can respond differently to the same therapies, which means the therapies might not work well for some of them. Identifying biomarkers that can help predict a cancer class with high accuracy is at the heart of breast cancer studies because they are targets of the treatments and drug development. Genomics data have been shown to carry useful information for breast cancer diagnosis and prognosis, as well as uncovering the disease’s mechanism. Machine learning methods are powerful tools to find such information. Feature selection methods are often utilized in supervised learning and unsupervised learning tasks to deal with data containing a large number of features in which only a small portion of them are useful to the classification task. On the other hand, analyzing only one type of data, without reference to the existing knowledge about the disease and the therapies, might mislead the findings. Effective data integration approaches are necessary to uncover this complex disease. In this thesis, we apply and develop machine learning methods to identify meaningful biomarkers for breast cancer survivability prediction after a certain treatment. They include applying feature selection methods on gene-expression data to derived gene-signatures, where the initial genes are collected concerning the mechanism of some drugs used breast cancer therapies. We also propose a new feature selection method, named PAFS, and apply it to discover accurate biomarkers. In addition, it has been increasingly supported that, sub-network biomarkers are more robust and accurate than gene biomarkers. We proposed two network-based approaches to identify sub-network biomarkers for breast cancer survivability prediction after a treatment. They integrate gene-expression data with protein-protein interactions during the optimal sub-network searching process and use cancer-related genes and pathways to prioritize the extracted sub-networks. The sub-network search space is usually huge and many proteins interact with thousands of other proteins. Thus, we apply some heuristics to avoid generating and evaluating redundant sub-networks

    Uncovering packaging features of co-regulated modules based on human protein interaction and transcriptional regulatory networks

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Network co-regulated modules are believed to have the functionality of packaging multiple biological entities, and can thus be assumed to coordinate many biological functions in their network neighbouring regions.</p> <p>Results</p> <p>Here, we weighted edges of a human protein interaction network and a transcriptional regulatory network to construct an integrated network, and introduce a probabilistic model and a bipartite graph framework to exploit human co-regulated modules and uncover their specific features in packaging different biological entities (genes, protein complexes or metabolic pathways). Finally, we identified 96 human co-regulated modules based on this method, and evaluate its effectiveness by comparing it with four other methods.</p> <p>Conclusions</p> <p>Dysfunctions in co-regulated interactions often occur in the development of cancer. Therefore, we focussed on an example co-regulated module and found that it could integrate a number of cancer-related genes. This was extended to causal dysfunctions of some complexes maintained by several physically interacting proteins, thus coordinating several metabolic pathways that directly underlie cancer.</p

    Network-assisted protein identification and data interpretation in shotgun proteomics

    Get PDF
    Protein assembly and biological interpretation of the assembled protein lists are critical steps in shotgun proteomics data analysis. Although most biological functions arise from interactions among proteins, current protein assembly pipelines treat proteins as independent entities. Usually, only individual proteins with strong experimental evidence, that is, confident proteins, are reported, whereas many possible proteins of biological interest are eliminated. We have developed a clique-enrichment approach (CEA) to rescue eliminated proteins by incorporating the relationship among proteins as embedded in a protein interaction network. In several data sets tested, CEA increased protein identification by 8–23% with an estimated accuracy of 85%. Rescued proteins were supported by existing literature or transcriptome profiling studies at similar levels as confident proteins and at a significantly higher level than abandoned ones. Applying CEA on a breast cancer data set, rescued proteins coded by well-known breast cancer genes. In addition, CEA generated a network view of the proteins and helped show the modular organization of proteins that may underpin the molecular mechanisms of the disease

    Investigating the validity of current network analysis on static conglomerate networks by protein network stratification

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>A molecular network perspective forms the foundation of systems biology. A common practice in analyzing protein-protein interaction (PPI) networks is to perform network analysis on a conglomerate network that is an assembly of all available binary interactions in a given organism from diverse data sources. Recent studies on network dynamics suggested that this approach might have ignored the dynamic nature of context-dependent molecular systems.</p> <p>Results</p> <p>In this study, we employed a network stratification strategy to investigate the validity of the current network analysis on conglomerate PPI networks. Using the genome-scale tissue- and condition-specific proteomics data in <it>Arabidopsis thaliana</it>, we present here the first systematic investigation into this question. We stratified a conglomerate <it>A. thaliana </it>PPI network into three levels of context-dependent subnetworks. We then focused on three types of most commonly conducted network analyses, i.e., topological, functional and modular analyses, and compared the results from these network analyses on the conglomerate network and five stratified context-dependent subnetworks corresponding to specific tissues.</p> <p>Conclusions</p> <p>We found that the results based on the conglomerate PPI network are often significantly different from those of context-dependent subnetworks corresponding to specific tissues or conditions. This conclusion depends neither on relatively arbitrary cutoffs (such as those defining network hubs or bottlenecks), nor on specific network clustering algorithms for module extraction, nor on the possible high false positive rates of binary interactions in PPI networks. We also found that our conclusions are likely to be valid in human PPI networks. Furthermore, network stratification may help resolve many controversies in current research of systems biology.</p

    A Knowledge-based Integrative Modeling Approach for <em>In-Silico</em> Identification of Mechanistic Targets in Neurodegeneration with Focus on Alzheimer’s Disease

    Get PDF
    Dementia is the progressive decline in cognitive function due to damage or disease in the body beyond what might be expected from normal aging. Based on neuropathological and clinical criteria, dementia includes a spectrum of diseases, namely Alzheimer's dementia, Parkinson's dementia, Lewy Body disease, Alzheimer's dementia with Parkinson's, Pick's disease, Semantic dementia, and large and small vessel disease. It is thought that these disorders result from a combination of genetic and environmental risk factors. Despite accumulating knowledge that has been gained about pathophysiological and clinical characteristics of the disease, no coherent and integrative picture of molecular mechanisms underlying neurodegeneration in Alzheimer’s disease is available. Existing drugs only offer symptomatic relief to the patients and lack any efficient disease-modifying effects. The present research proposes a knowledge-based rationale towards integrative modeling of disease mechanism for identifying potential candidate targets and biomarkers in Alzheimer’s disease. Integrative disease modeling is an emerging knowledge-based paradigm in translational research that exploits the power of computational methods to collect, store, integrate, model and interpret accumulated disease information across different biological scales from molecules to phenotypes. It prepares the ground for transitioning from ‘descriptive’ to “mechanistic” representation of disease processes. The proposed approach was used to introduce an integrative framework, which integrates, on one hand, extracted knowledge from the literature using semantically supported text-mining technologies and, on the other hand, primary experimental data such as gene/protein expression or imaging readouts. The aim of such a hybrid integrative modeling approach was not only to provide a consolidated systems view on the disease mechanism as a whole but also to increase specificity and sensitivity of the mechanistic model by providing disease-specific context. This approach was successfully used for correlating clinical manifestations of the disease to their corresponding molecular events and led to the identification and modeling of three important mechanistic components underlying Alzheimer’s dementia, namely the CNS, the immune system and the endocrine components. These models were validated using a novel in-silico validation method, namely biomarker-guided pathway analysis and a pathway-based target identification approach was introduced, which resulted in the identification of the MAPK signaling pathway as a potential candidate target at the crossroad of the triad components underlying disease mechanism in Alzheimer’s dementia

    Integrative Biomarker Discovery for Breast Cancer Metastasis from Gene Expression and Protein Interaction Data Using Error-tolerant Pattern Mining

    No full text
    Biomarker discovery for complex diseases is a challenging problem. Most of the existing approaches identify individual genes as disease markers, thereby missing the interactions among genes. Moreover, often only single biological data source is used to discover biomarkers. These factors account for the discovery of inconsistent biomarkers. In this paper, we propose a novel error-tolerant pattern mining approach for integrated analysis of gene expression and protein interaction data. This integrated approach incorporates constraints from protein interaction network and efficiently discovers patterns (groups of genes) in a bottomup fashion from the gene-expression data. We call these patterns active sub-network biomarkers. To illustrate the efficacy of our proposed approach, we used four breast cancer gene expression data sets and a human protein interaction network and showed that active sub-network biomarkers are more biologically plausible and genes discovered are more reproducible across studies. Finally, through pathway analysis, we also showed a substantial enrichment for known cancer genes and hence were able to generate relevant hypotheses for understanding the molecular mechanisms of breast cancer metastasis.
    corecore