8,583 research outputs found

    Knowledge transfer via classification rules using functional mapping for integrative modeling of gene expression data

    Get PDF
    Background: Most 'transcriptomic' data from microarrays are generated from small sample sizes compared to the large number of measured biomarkers, making it very difficult to build accurate and generalizable disease state classification models. Integrating information from different, but related, 'transcriptomic' data may help build better classification models. However, most proposed methods for integrative analysis of 'transcriptomic' data cannot incorporate domain knowledge, which can improve model performance. To this end, we have developed a methodology that leverages transfer rule learning and functional modules, which we call TRL-FM, to capture and abstract domain knowledge in the form of classification rules to facilitate integrative modeling of multiple gene expression data. TRL-FM is an extension of the transfer rule learner (TRL) that we developed previously. The goal of this study was to test our hypothesis that "an integrative model obtained via the TRL-FM approach outperforms traditional models based on single gene expression data sources". Results: To evaluate the feasibility of the TRL-FM framework, we compared the area under the ROC curve (AUC) of models developed with TRL-FM and other traditional methods, using 21 microarray datasets generated from three studies on brain cancer, prostate cancer, and lung disease, respectively. The results show that TRL-FM statistically significantly outperforms TRL as well as traditional models based on single source data. In addition, TRL-FM performed better than other integrative models driven by meta-analysis and cross-platform data merging. Conclusions: The capability of utilizing transferred abstract knowledge derived from source data using feature mapping enables the TRL-FM framework to mimic the human process of learning and adaptation when performing related tasks. The novel TRL-FM methodology for integrative modeling for multiple 'transcriptomic' datasets is able to intelligently incorporate domain knowledge that traditional methods might disregard, to boost predictive power and generalization performance. In this study, TRL-FM's abstraction of knowledge is achieved in the form of functional modules, but the overall framework is generalizable in that different approaches of acquiring abstract knowledge can be integrated into this framework

    A robust and efficient method for estimating enzyme complex abundance and metabolic flux from expression data

    Get PDF
    A major theme in constraint-based modeling is unifying experimental data, such as biochemical information about the reactions that can occur in a system or the composition and localization of enzyme complexes, with highthroughput data including expression data, metabolomics, or DNA sequencing. The desired result is to increase predictive capability resulting in improved understanding of metabolism. The approach typically employed when only gene (or protein) intensities are available is the creation of tissue-specific models, which reduces the available reactions in an organism model, and does not provide an objective function for the estimation of fluxes, which is an important limitation in many modeling applications. We develop a method, flux assignment with LAD (least absolute deviation) convex objectives and normalization (FALCON), that employs metabolic network reconstructions along with expression data to estimate fluxes. In order to use such a method, accurate measures of enzyme complex abundance are needed, so we first present a new algorithm that addresses quantification of complex abundance. Our extensions to prior techniques include the capability to work with large models and significantly improved run-time performance even for smaller models, an improved analysis of enzyme complex formation logic, the ability to handle very large enzyme complex rules that may incorporate multiple isoforms, and depending on the model constraints, either maintained or significantly improved correlation with experimentally measured fluxes. FALCON has been implemented in MATLAB and ATS, and can be downloaded from: https://github.com/bbarker/FALCON. ATS is not required to compile the software, as intermediate C source code is available, and binaries are provided for Linux x86-64 systems. FALCON requires use of the COBRA Toolbox, also implemented in MATLAB.Comment: 30 pages, 12 figures, 4 table

    Integration and mining of malaria molecular, functional and pharmacological data: how far are we from a chemogenomic knowledge space?

    Get PDF
    The organization and mining of malaria genomic and post-genomic data is highly motivated by the necessity to predict and characterize new biological targets and new drugs. Biological targets are sought in a biological space designed from the genomic data from Plasmodium falciparum, but using also the millions of genomic data from other species. Drug candidates are sought in a chemical space containing the millions of small molecules stored in public and private chemolibraries. Data management should therefore be as reliable and versatile as possible. In this context, we examined five aspects of the organization and mining of malaria genomic and post-genomic data: 1) the comparison of protein sequences including compositionally atypical malaria sequences, 2) the high throughput reconstruction of molecular phylogenies, 3) the representation of biological processes particularly metabolic pathways, 4) the versatile methods to integrate genomic data, biological representations and functional profiling obtained from X-omic experiments after drug treatments and 5) the determination and prediction of protein structures and their molecular docking with drug candidate structures. Progresses toward a grid-enabled chemogenomic knowledge space are discussed.Comment: 43 pages, 4 figures, to appear in Malaria Journa

    Updates in metabolomics tools and resources: 2014-2015

    Get PDF
    Data processing and interpretation represent the most challenging and time-consuming steps in high-throughput metabolomic experiments, regardless of the analytical platforms (MS or NMR spectroscopy based) used for data acquisition. Improved machinery in metabolomics generates increasingly complex datasets that create the need for more and better processing and analysis software and in silico approaches to understand the resulting data. However, a comprehensive source of information describing the utility of the most recently developed and released metabolomics resources—in the form of tools, software, and databases—is currently lacking. Thus, here we provide an overview of freely-available, and open-source, tools, algorithms, and frameworks to make both upcoming and established metabolomics researchers aware of the recent developments in an attempt to advance and facilitate data processing workflows in their metabolomics research. The major topics include tools and researches for data processing, data annotation, and data visualization in MS and NMR-based metabolomics. Most in this review described tools are dedicated to untargeted metabolomics workflows; however, some more specialist tools are described as well. All tools and resources described including their analytical and computational platform dependencies are summarized in an overview Table

    Deep Learning for Genomics: A Concise Overview

    Full text link
    Advancements in genomic research such as high-throughput sequencing techniques have driven modern genomic studies into "big data" disciplines. This data explosion is constantly challenging conventional methods used in genomics. In parallel with the urgent demand for robust algorithms, deep learning has succeeded in a variety of fields such as vision, speech, and text processing. Yet genomics entails unique challenges to deep learning since we are expecting from deep learning a superhuman intelligence that explores beyond our knowledge to interpret the genome. A powerful deep learning model should rely on insightful utilization of task-specific knowledge. In this paper, we briefly discuss the strengths of different deep learning models from a genomic perspective so as to fit each particular task with a proper deep architecture, and remark on practical considerations of developing modern deep learning architectures for genomics. We also provide a concise review of deep learning applications in various aspects of genomic research, as well as pointing out potential opportunities and obstacles for future genomics applications.Comment: Invited chapter for Springer Book: Handbook of Deep Learning Application

    In-silico-Systemanalyse von Biopathways

    Get PDF
    Chen M. In silico systems analysis of biopathways. Bielefeld (Germany): Bielefeld University; 2004.In the past decade with the advent of high-throughput technologies, biology has migrated from a descriptive science to a predictive one. A vast amount of information on the metabolism have been produced; a number of specific genetic/metabolic databases and computational systems have been developed, which makes it possible for biologists to perform in silico analysis of metabolism. With experimental data from laboratory, biologists wish to systematically conduct their analysis with an easy-to-use computational system. One major task is to implement molecular information systems that will allow to integrate different molecular database systems, and to design analysis tools (e.g. simulators of complex metabolic reactions). Three key problems are involved: 1) Modeling and simulation of biological processes; 2) Reconstruction of metabolic pathways, leading to predictions about the integrated function of the network; and 3) Comparison of metabolism, providing an important way to reveal the functional relationship between a set of metabolic pathways. This dissertation addresses these problems of in silico systems analysis of biopathways. We developed a software system to integrate the access to different databases, and exploited the Petri net methodology to model and simulate metabolic networks in cells. It develops a computer modeling and simulation technique based on Petri net methodology; investigates metabolic networks at a system level; proposes a markup language for biological data interchange among diverse biological simulators and Petri net tools; establishes a web-based information retrieval system for metabolic pathway prediction; presents an algorithm for metabolic pathway alignment; recommends a nomenclature of cellular signal transduction; and attempts to standardize the representation of biological pathways. Hybrid Petri net methodology is exploited to model metabolic networks. Kinetic modeling strategy and Petri net modeling algorithm are applied to perform the processes of elements functioning and model analysis. The proposed methodology can be used for all other metabolic networks or the virtual cell metabolism. Moreover, perspectives of Petri net modeling and simulation of metabolic networks are outlined. A proposal for the Biology Petri Net Markup Language (BioPNML) is presented. The concepts and terminology of the interchange format, as well as its syntax (which is based on XML) are introduced. BioPNML is designed to provide a starting point for the development of a standard interchange format for Bioinformatics and Petri nets. The language makes it possible to exchange biology Petri net diagrams between all supported hardware platforms and versions. It is also designed to associate Petri net models and other known metabolic simulators. A web-based metabolic information retrieval system, PathAligner, is developed in order to predict metabolic pathways from rudimentary elements of pathways. It extracts metabolic information from biological databases via the Internet, and builds metabolic pathways with data sources of genes, sequences, enzymes, metabolites, etc. The system also provides a navigation platform to investigate metabolic related information, and transforms the output data into XML files for further modeling and simulation of the reconstructed pathway. An alignment algorithm to compare the similarity between metabolic pathways is presented. A new definition of the metabolic pathway is proposed. The pathway defined as a linear event sequence is practical for our alignment algorithm. The algorithm is based on strip scoring the similarity of 4-hierarchical EC numbers involved in the pathways. The algorithm described has been implemented and is in current use in the context of the PathAligner system. Furthermore, new methods for the classification and nomenclature of cellular signal transductions are recommended. For each type of characterized signal transduction, a unique ST number is provided. The Signal Transduction Classification Database (STCDB), based on the proposed classification and nomenclature, has been established. By merging the ST numbers with EC numbers, alignments of biopathways are possible. Finally, a detailed model of urea cycle that includes gene regulatory networks, metabolic pathways and signal transduction is demonstrated by using our approaches. A system biological interpretation of the observed behavior of the urea cycle and its related transcriptomics information is proposed to provide new insights for metabolic engineering and medical care

    Artificial intelligence for dementia genetics and omics

    Get PDF
    Genetics and omics studies of Alzheimer's disease and other dementia subtypes enhance our understanding of underlying mechanisms and pathways that can be targeted. We identified key remaining challenges: First, can we enhance genetic studies to address missing heritability? Can we identify reproducible omics signatures that differentiate between dementia subtypes? Can high-dimensional omics data identify improved biomarkers? How can genetics inform our understanding of causal status of dementia risk factors? And which biological processes are altered by dementia-related genetic variation? Artificial intelligence (AI) and machine learning approaches give us powerful new tools in helping us to tackle these challenges, and we review possible solutions and examples of best practice. However, their limitations also need to be considered, as well as the need for coordinated multidisciplinary research and diverse deeply phenotyped cohorts. Ultimately AI approaches improve our ability to interrogate genetics and omics data for precision dementia medicine

    Mapping the proteome with data-driven methods: A cycle of measurement, modeling, hypothesis generation, and engineering

    Get PDF
    The living cell exhibits emergence of complex behavior and its modeling requires a systemic, integrative approach if we are to thoroughly understand and harness it. The work in this thesis has had the more narrow aim of quantitatively characterizing and mapping the proteome using data-driven methods, as proteins perform most functional and structural roles within the cell. Covered are the different parts of the cycle from improving quantification methods, to deriving protein features relying on their primary structure, predicting the protein content solely from sequence data, and, finally, to developing theoretical protein engineering tools, leading back to experiment.\ua0\ua0\ua0\ua0 High-throughput mass spectrometry platforms provide detailed snapshots of a cell\u27s protein content, which can be mined towards understanding how the phenotype arises from genotype and the interplay between the various properties of the constituent proteins. However, these large and dense data present an increased analysis challenge and current methods capture only a small fraction of signal. The first part of my work has involved tackling these issues with the implementation of a GPU-accelerated and distributed signal decomposition pipeline, making factorization of large proteomics scans feasible and efficient. The pipeline yields individual analyte signals spanning the majority of acquired signal, enabling high precision quantification and further analytical tasks.\ua0\ua0\ua0 Having such detailed snapshots of the proteome enables a multitude of undertakings. One application has been to use a deep neural network model to learn the amino acid sequence determinants of temperature adaptation, in the form of reusable deep model features. More generally, systemic quantities may be predicted from the information encoded in sequence by evolutionary pressure. Two studies taking inspiration from natural language processing have sought to learn the grammars behind the languages of expression, in one case predicting mRNA levels from DNA sequence, and in the other protein abundance from amino acid sequence. These two models helped build a quantitative understanding of the central dogma and, furthermore, in combination yielded an improved predictor of protein amount. Finally, a mathematical framework relying on the embedded space of a deep model has been constructed to assist guided mutation of proteins towards optimizing their abundance
    • …
    corecore