82 research outputs found

    A message passing framework with multiple data integration for miRNA-disease association prediction

    Get PDF
    Micro RNA or miRNA is a highly conserved class of non-coding RNA that plays an important role in many diseases. Identifying miRNA-disease associations can pave the way for better clinical diagnosis and finding potential drug targets. We propose a biologically-motivated data-driven approach for the miRNA-disease association prediction, which overcomes the data scarcity problem by exploiting information from multiple data sources. The key idea is to enrich the existing miRNA/disease-protein-coding gene (PCG) associations via a message passing framework, followed by the use of disease ontology information for further feature filtering. The enriched and filtered PCG associations are then used to construct the inter-connected miRNA-PCG-disease network to train a structural deep network embedding (SDNE) model. Finally, the pre-trained embeddings and the biologically relevant features from the miRNA family and disease semantic similarity are concatenated to form the pair input representations to a Random Forest classifier whose task is to predict the miRNA-disease association probabilities. We present large-scale comparative experiments, ablation, and case studies to showcase our approach’s superiority. Besides, we make the model prediction results for 1618 miRNAs and 3679 diseases, along with all related information, publicly available at http://software.mpm.leibniz-ai-lab.de/ to foster assessments and future adoption

    Integration of multi-scale protein interactions for biomedical data analysis

    Get PDF
    With the advancement of modern technologies, we observe an increasing accumulation of biomedical data about diseases. There is a need for computational methods to sift through and extract knowledge from the diverse data available in order to improve our mechanistic understanding of diseases and improve patient care. Biomedical data come in various forms as exemplified by the various omics data. Existing studies have shown that each form of omics data gives only partial information on cells state and motivated jointly mining multi-omics, multi-modal data to extract integrated system knowledge. The interactome is of particular importance as it enables the modelling of dependencies arising from molecular interactions. This Thesis takes a special interest in the multi-scale protein interactome and its integration with computational models to extract relevant information from biomedical data. We define multi-scale interactions at different omics scale that involve proteins: pairwise protein-protein interactions, multi-protein complexes, and biological pathways. Using hypergraph representations, we motivate considering higher-order protein interactions, highlighting the complementary biological information contained in the multi-scale interactome. Based on those results, we further investigate how those multi-scale protein interactions can be used as either prior knowledge, or auxiliary data to develop machine learning algorithms. First, we design a neural network using the multi-scale organization of proteins in a cell into biological pathways as prior knowledge and train it to predict a patient's diagnosis based on transcriptomics data. From the trained models, we develop a strategy to extract biomedical knowledge pertaining to the diseases investigated. Second, we propose a general framework based on Non-negative Matrix Factorization to integrate the multi-scale protein interactome with multi-omics data. We show that our approach outperforms the existing methods, provide biomedical insights and relevant hypotheses for specific cancer types

    Joint learning from multiple information sources for biological problems

    Get PDF
    Thanks to technological advancements, more and more biological data havebeen generated in recent years. Data availability offers unprecedented opportunities to look at the same problem from multiple aspects. It also unveils a more global view of the problem that takes into account the intricated inter-play between the involved molecules/entities. Nevertheless, biological datasets are biased, limited in quantity, and contain many false-positive samples. Such challenges often drastically downgrade the performance of a predictive model on unseen data and, thus, limit its applicability in real biological studies. Human learning is a multi-stage process in which we usually start with simple things. Through the accumulated knowledge over time, our cognition ability extends to more complex concepts. Children learn to speak simple words before being able to formulate sentences. Similarly, being able to speak correct sentences supports our learning to speak correct and meaningful paragraphs, etc. Generally, knowledge acquired from related learning tasks would help boost our learning capability in the current task. Motivated by such a phenomenon, in this thesis, we study supervised machine learning models for bioinformatics problems that can improve their performance through exploiting multiple related knowledge sources. More specifically, we concern with ways to enrich the supervised models’ knowledge base with publicly available related data to enhance the computational models’ prediction performance. Our work shares commonality with existing works in multimodal learning, multi-task learning, and transfer learning. Nevertheless, there are certain differences in some cases. Besides the proposed architectures, we present large-scale experiment setups with consensus evaluation metrics along with the creation and release of large datasets to showcase our approaches’ superiority. Moreover, we add case studies with detailed analyses in which we place no simplified assumptions to demonstrate the systems’ utilities in realistic application scenarios. Finally, we develop and make available an easy-to-use website for non-expert users to query the model’s generated prediction results to facilitate field experts’ assessments and adaptation. We believe that our work serves as one of the first steps in bridging the gap between “Computer Science” and “Biology” that will open a new era of fruitful collaboration between computer scientists and biological field experts

    Opportunities and obstacles for deep learning in biology and medicine

    Get PDF
    Deep learning describes a class of machine learning algorithms that are capable of combining raw inputs into layers of intermediate features. These algorithms have recently shown impressive results across a variety of domains. Biology and medicine are data-rich disciplines, but the data are complex and often ill-understood. Hence, deep learning techniques may be particularly well suited to solve problems of these fields. We examine applications of deep learning to a variety of biomedical problems-patient classification, fundamental biological processes and treatment of patients-and discuss whether deep learning will be able to transform these tasks or if the biomedical sphere poses unique challenges. Following from an extensive literature review, we find that deep learning has yet to revolutionize biomedicine or definitively resolve any of the most pressing challenges in the field, but promising advances have been made on the prior state of the art. Even though improvements over previous baselines have been modest in general, the recent progress indicates that deep learning methods will provide valuable means for speeding up or aiding human investigation. Though progress has been made linking a specific neural network\u27s prediction to input features, understanding how users should interpret these models to make testable hypotheses about the system under study remains an open challenge. Furthermore, the limited amount of labelled data for training presents problems in some domains, as do legal and privacy constraints on work with sensitive health records. Nonetheless, we foresee deep learning enabling changes at both bench and bedside with the potential to transform several areas of biology and medicine

    Generation and Applications of Knowledge Graphs in Systems and Networks Biology

    Get PDF
    The acceleration in the generation of data in the biomedical domain has necessitated the use of computational approaches to assist in its interpretation. However, these approaches rely on the availability of high quality, structured, formalized biomedical knowledge. This thesis has the two goals to improve methods for curation and semantic data integration to generate high granularity biological knowledge graphs and to develop novel methods for using prior biological knowledge to propose new biological hypotheses. The first two publications describe an ecosystem for handling biological knowledge graphs encoded in the Biological Expression Language throughout the stages of curation, visualization, and analysis. Further, the second two publications describe the reproducible acquisition and integration of high-granularity knowledge with low contextual specificity from structured biological data sources on a massive scale and support the semi-automated curation of new content at high speed and precision. After building the ecosystem and acquiring content, the last three publications in this thesis demonstrate three different applications of biological knowledge graphs in modeling and simulation. The first demonstrates the use of agent-based modeling for simulation of neurodegenerative disease biomarker trajectories using biological knowledge graphs as priors. The second applies network representation learning to prioritize nodes in biological knowledge graphs based on corresponding experimental measurements to identify novel targets. Finally, the third uses biological knowledge graphs and develops algorithmics to deconvolute the mechanism of action of drugs, that could also serve to identify drug repositioning candidates. Ultimately, the this thesis lays the groundwork for production-level applications of drug repositioning algorithms and other knowledge-driven approaches to analyzing biomedical experiments

    Big Data Analytics for Complex Systems

    Get PDF
    The evolution of technology in all fields led to the generation of vast amounts of data by modern systems. Using data to extract information, make predictions, and make decisions is the current trend in artificial intelligence. The advancement of big data analytics tools made accessing and storing data easier and faster than ever, and machine learning algorithms help to identify patterns in and extract information from data. The current tools and machines in health, computer technologies, and manufacturing can generate massive raw data about their products or samples. The author of this work proposes a modern integrative system that can utilize big data analytics, machine learning, super-computer resources, and industrial health machines’ measurements to build a smart system that can mimic the human intelligence skills of observations, detection, prediction, and decision-making. The applications of the proposed smart systems are included as case studies to highlight the contributions of each system. The first contribution is the ability to utilize big data revolutionary and deep learning technologies on production lines to diagnose incidents and take proper action. In the current digital transformational industrial era, Industry 4.0 has been receiving researcher attention because it can be used to automate production-line decisions. Reconfigurable manufacturing systems (RMS) have been widely used to reduce the setup cost of restructuring production lines. However, the current RMS modules are not linked to the cloud for online decision-making to take the proper decision; these modules must connect to an online server (super-computer) that has big data analytics and machine learning capabilities. The online means that data is centralized on cloud (supercomputer) and accessible in real-time. In this study, deep neural networks are utilized to detect the decisive features of a product and build a prediction model in which the iFactory will make the necessary decision for the defective products. The Spark ecosystem is used to manage the access, processing, and storing of the big data streaming. This contribution is implemented as a closed cycle, which for the best of our knowledge, no one in the literature has introduced big data analysis using deep learning on real-time applications in the manufacturing system. The code shows a high accuracy of 97% for classifying the normal versus defective items. The second contribution, which is in Bioinformatics, is the ability to build supervised machine learning approaches based on the gene expression of patients to predict proper treatment for breast cancer. In the trial, to personalize treatment, the machine learns the genes that are active in the patient cohort with a five-year survival period. The initial condition here is that each group must only undergo one specific treatment. After learning about each group (or class), the machine can personalize the treatment of a new patient by diagnosing the patients’ gene expression. The proposed model will help in the diagnosis and treatment of the patient. The future work in this area involves building a protein-protein interaction network with the selected genes for each treatment to first analyze the motives of the genes and target them with the proper drug molecules. In the learning phase, a couple of feature-selection techniques and supervised standard classifiers are used to build the prediction model. Most of the nodes show a high-performance measurement where accuracy, sensitivity, specificity, and F-measure ranges around 100%. The third contribution is the ability to build semi-supervised learning for the breast cancer survival treatment that advances the second contribution. By understanding the relations between the classes, we can design the machine learning phase based on the similarities between classes. In the proposed research, the researcher used the Euclidean matrix distance among each survival treatment class to build the hierarchical learning model. The distance information that is learned through a non-supervised approach can help the prediction model to select the classes that are away from each other to maximize the distance between classes and gain wider class groups. The performance measurement of this approach shows a slight improvement from the second model. However, this model reduced the number of discriminative genes from 47 to 37. The model in the second contribution studies each class individually while this model focuses on the relationships between the classes and uses this information in the learning phase. Hierarchical clustering is completed to draw the borders between groups of classes before building the classification models. Several distance measurements are tested to identify the best linkages between classes. Most of the nodes show a high-performance measurement where accuracy, sensitivity, specificity, and F-measure ranges from 90% to 100%. All the case study models showed high-performance measurements in the prediction phase. These modern models can be replicated for different problems within different domains. The comprehensive models of the newer technologies are reconfigurable and modular; any newer learning phase can be plugged-in at both ends of the learning phase. Therefore, the output of the system can be an input for another learning system, and a newer feature can be added to the input to be considered for the learning phase
    • …
    corecore