1,750 research outputs found

    Inferring Gene Regulatory Networks from Time Series Microarray Data

    Get PDF
    The innovations and improvements in high-throughput genomic technologies, such as DNA microarray, make it possible for biologists to simultaneously measure dependencies and regulations among genes on a genome-wide scale and provide us genetic information. An important objective of the functional genomics is to understand the controlling mechanism of the expression of these genes and encode the knowledge into gene regulatory network (GRN). To achieve this, computational and statistical algorithms are especially needed. Inference of GRN is a very challenging task for computational biologists because the degree of freedom of the parameters is redundant. Various computational approaches have been proposed for modeling gene regulatory networks, such as Boolean network, differential equations and Bayesian network. There is no so called golden method which can generally give us the best performance for any data set. The research goal is to improve inference accuracy and reduce computational complexity. One of the problems in reconstructing GRN is how to deal with the high dimensionality and short time course gene expression data. In this work, some existing inference algorithms are compared and the limitations lie in that they either suffer from low inference accuracy or computational complexity. To overcome such difficulties, a new approach based on state space model and Expectation-Maximization (EM) algorithms is proposed to model the dynamic system of gene regulation and infer gene regulatory networks. In our model, GRN is represented by a state space model that incorporates noises and has the ability to capture more various biological aspects, such as hidden or missing variables. An EM algorithm is used to estimate the parameters based on the given state space functions and the gene interaction matrix is derived by decomposing the observation matrix using singular value decomposition, and then it is used to infer GRN. The new model is validated using synthetic data sets before applying it to real biological data sets. The results reveal that the developed model can infer the gene regulatory networks from large scale gene expression data and significantly reduce the computational time complexity without losing much inference accuracy compared to dynamic Bayesian network

    Modeling multiple time units delayed gene regulatory network using dynamic Bayesian network.

    Get PDF

    Inference of the genetic network regulating lateral root initiation in Arabidopsis thaliana

    Get PDF
    Regulation of gene expression is crucial for organism growth, and it is one of the challenges in Systems Biology to reconstruct the underlying regulatory biological networks from transcriptomic data. The formation of lateral roots in Arabidopsis thaliana is stimulated by a cascade of regulators of which only the interactions of its initial elements have been identified. Using simulated gene expression data with known network topology, we compare the performance of inference algorithms, based on different approaches, for which ready-to-use software is available. We show that their performance improves with the network size and the inclusion of mutants. We then analyse two sets of genes, whose activity is likely to be relevant to lateral root initiation in Arabidopsis, by integrating sequence analysis with the intersection of the results of the best performing methods on time series and mutants to infer their regulatory network. The methods applied capture known interactions between genes that are candidate regulators at early stages of development. The network inferred from genes significantly expressed during lateral root formation exhibits distinct scale-free, small world and hierarchical properties and the nodes with a high out-degree may warrant further investigation

    Comparative study of discretization methods of microarray data for inferring transcriptional regulatory networks

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Microarray data discretization is a basic preprocess for many algorithms of gene regulatory network inference. Some common discretization methods in informatics are used to discretize microarray data. Selection of the discretization method is often arbitrary and no systematic comparison of different discretization has been conducted, in the context of gene regulatory network inference from time series gene expression data.</p> <p>Results</p> <p>In this study, we propose a new discretization method "bikmeans", and compare its performance with four other widely-used discretization methods using different datasets, modeling algorithms and number of intervals. Sensitivities, specificities and total accuracies were calculated and statistical analysis was carried out. Bikmeans method always gave high total accuracies.</p> <p>Conclusions</p> <p>Our results indicate that proper discretization methods can consistently improve gene regulatory network inference independent of network modeling algorithms and datasets. Our new method, bikmeans, resulted in significant better total accuracies than other methods.</p

    Multi-scale genetic network inference based on time series gene expression profiles

    Get PDF
    This work integrates multi-scale clustering and short-time correlation to estimate genetic networks with different time resolutions and detail levels. Gene expression data are noisy and large scale. Clustering is widely used to group genes with similar pattern. The cluster centers can be used to infer the genetic networks among these clusters. This work introduces the Multi-scale Fuzzy K-means clustering algorithm to uncover groups of coregulated genes and capture the networks in different levels of detail.;Time series expression profiles provide dynamic information for inferring gene regulatory relationships. Large scale network inference, identifying the transient interactions and feedback loops as well as differentiating direct and indirect interactions are among the major challenges of genetic network inference. Time correlation can estimate the time delay and edge direction. Partial correlation and directed-separation theory help differentiate direct and indirect interactions and identify feedback loops. This work introduces the constraint-based time-correlation (CBTC) network inference algorithm that combines these methods with time correlation estimation to more fully characterize genetic networks. Gene expression regulation can happen in specific time periods and conditions instead of across the whole expression profile. Short-time correlation can capture transient interactions.;The network discovery algorithm was mainly validated using yeast cell cycle data. The algorithm successfully identified the yeast cell cycle development stages, cell cycle and negative feedback loops, and indicated how the networks dynamically changes over time. The inferred networks reflect most interactions previously identified by genome-wide location analysis and match the extant literature. At detailed network level, the inferred networks provide more detailed information about genes (or clusters) and the interactions among them. Interesting genes, clusters and interactions were identified, which match the literature and the gene ontology information and provide hypotheses for further studies

    Defining a robust biological prior from Pathway Analysis to drive Network Inference

    Get PDF
    Inferring genetic networks from gene expression data is one of the most challenging work in the post-genomic era, partly due to the vast space of possible networks and the relatively small amount of data available. In this field, Gaussian Graphical Model (GGM) provides a convenient framework for the discovery of biological networks. In this paper, we propose an original approach for inferring gene regulation networks using a robust biological prior on their structure in order to limit the set of candidate networks. Pathways, that represent biological knowledge on the regulatory networks, will be used as an informative prior knowledge to drive Network Inference. This approach is based on the selection of a relevant set of genes, called the "molecular signature", associated with a condition of interest (for instance, the genes involved in disease development). In this context, differential expression analysis is a well established strategy. However outcome signatures are often not consistent and show little overlap between studies. Thus, we will dedicate the first part of our work to the improvement of the standard process of biomarker identification to guarantee the robustness and reproducibility of the molecular signature. Our approach enables to compare the networks inferred between two conditions of interest (for instance case and control networks) and help along the biological interpretation of results. Thus it allows to identify differential regulations that occur in these conditions. We illustrate the proposed approach by applying our method to a study of breast cancer's response to treatment

    Comparison of Probabilistic Boolean Network and Dynamic Bayesian Network Approaches for Inferring Gene Regulatory Networks

    Get PDF
    Background: The regulation of gene expression is achieved through gene regulatory networks (GRNs) in which collections of genes interact with one another and other substances in a cell. In order to understand the underlying function of organisms, it is necessary to study the behavior of genes in a gene regulatory network context. Several computational approaches are available for modeling gene regulatory networks with different datasets. In order to optimize modeling of GRN, these approaches must be compared and evaluated in terms of accuracy and efficiency. Results: In this paper, two important computational approaches for modeling gene regulatory networks, probabilistic Boolean network methods and dynamic Bayesian network methods, are compared using a biological time-series dataset from the Drosophila Interaction Database to construct a Drosophila gene network. A subset of time points and gene samples from the whole dataset is used to evaluate the performance of these two approaches. Conclusions: The comparison indicates that both approaches had good performance in modeling the gene regulatory networks. The accuracy in terms of recall and precision can be improved if a smaller subset of genes is selected for inferring GRNs. The accuracy of both approaches is dependent upon the number of selected genes and time points of gene samples. In all tested cases, DBN identified more gene interactions and gave better recall than PBN

    Bayesian network learning and applications in Bioinformatics

    Get PDF
    Abstract A Bayesian network (BN) is a compact graphic representation of the probabilistic re- lationships among a set of random variables. The advantages of the BN formalism include its rigorous mathematical basis, the characteristics of locality both in knowl- edge representation and during inference, and the innate way to deal with uncertainty. Over the past decades, BNs have gained increasing interests in many areas, including bioinformatics which studies the mathematical and computing approaches to under- stand biological processes. In this thesis, I develop new methods for BN structure learning with applications to bi- ological network reconstruction and assessment. The first application is to reconstruct the genetic regulatory network (GRN), where each gene is modeled as a node and an edge indicates a regulatory relationship between two genes. In this task, we are given time-series microarray gene expression measurements for tens of thousands of genes, which can be modeled as true gene expressions mixed with noise in data generation, variability of the underlying biological systems etc. We develop a novel BN structure learning algorithm for reconstructing GRNs. The second application is to develop a BN method for protein-protein interaction (PPI) assessment. PPIs are the foundation of most biological mechanisms, and the knowl- edge on PPI provides one of the most valuable resources from which annotations of genes and proteins can be discovered. Experimentally, recently-developed high- throughput technologies have been carried out to reveal protein interactions in many organisms. However, high-throughput interaction data often contain a large number of iv spurious interactions. In this thesis, I develop a novel in silico model for PPI assess- ment. Our model is based on a BN that integrates heterogeneous data sources from different organisms. The main contributions are: 1. A new concept to depict the dynamic dependence relationships among random variables, which widely exist in biological processes, such as the relationships among genes and genes' products in regulatory networks and signaling pathways. This con- cept leads to a novel algorithm for dynamic Bayesian network learning. We apply it to time-series microarray gene expression data, and discover some missing links in a well-known regulatory pathway. Those new causal relationships between genes have been found supportive evidences in literature. 2. Discovery and theoretical proof of an asymptotic property of K2 algorithm ( a well-known efficient BN structure learning approach). This property has been used to identify Markov blankets (MB) in a Bayesian network, and further recover the BN structure. This hybrid algorithm is evaluated on a benchmark regulatory pathway, and obtains better results than some state-of-art Bayesian learning approaches. 3. A Bayesian network based integrative method which incorporates heterogeneous data sources from different organisms to predict protein-protein interactions (PPI) in a target organism. The framework is employed in human PPI prediction and in as- sessment of high-throughput PPI data. Furthermore, our experiments reveal some interesting biological results. 4. We introduce the learning of a TAN (Tree Augmented Naïve Bayes) based net- work, which has the computational simplicity and robustness to high-throughput PPI assessment. The empirical results show that our method outperforms naïve Bayes and a manual constructed Bayesian Network, additionally demonstrate sufficient informa- tion from model organisms can achieve high accuracy in PPI prediction

    Integrate qualitative biological knowledge for gene regulatory network reconstruction with dynamic Bayesian networks

    Get PDF
    Reconstructing gene regulatory networks, especially the dynamic gene networks that reveal the temporal program of gene expression from microarray expression data, is essential in systems biology. To overcome the challenges posed by the noisy and under-sampled microarray data, developing data fusion methods to integrate legacy biological knowledge for gene network reconstruction is a promising direction. However, large amount of qualitative biological knowledge accumulated by previous research, albeit very valuable, has received less attention for reconstructing dynamic gene networks due to its incompatibility with the quantitative computational models.;In this dissertation, I introduce a novel method to fuse qualitative gene interaction information with quantitative microarray data under the Dynamic Bayesian Networks framework. This method extends the previous data integration methods by its capabilities of both utilizing qualitative biological knowledge by using Bayesian Networks without the involvement of human experts, and taking time-series data to produce dynamic gene networks. The experimental study shows that when compared with standard Dynamic Bayesian Networks method which only uses microarray data, our method excels by both accuracy and consistency

    Discovering time-lagged rules from microarray data using gene profile classifiers

    Get PDF
    Background: Gene regulatory networks have an essential role in every process of life. In this regard, the amount of genome-wide time series data is becoming increasingly available, providing the opportunity to discover the time-delayed gene regulatory networks that govern the majority of these molecular processes.Results: This paper aims at reconstructing gene regulatory networks from multiple genome-wide microarray time series datasets. In this sense, a new model-free algorithm called GRNCOP2 (Gene Regulatory Network inference by Combinatorial OPtimization 2), which is a significant evolution of the GRNCOP algorithm, was developed using combinatorial optimization of gene profile classifiers. The method is capable of inferring potential time-delay relationships with any span of time between genes from various time series datasets given as input. The proposed algorithm was applied to time series data composed of twenty yeast genes that are highly relevant for the cell-cycle study, and the results were compared against several related approaches. The outcomes have shown that GRNCOP2 outperforms the contrasted methods in terms of the proposed metrics, and that the results are consistent with previous biological knowledge. Additionally, a genome-wide study on multiple publicly available time series data was performed. In this case, the experimentation has exhibited the soundness and scalability of the new method which inferred highly-related statistically-significant gene associations.Conclusions: A novel method for inferring time-delayed gene regulatory networks from genome-wide time series datasets is proposed in this paper. The method was carefully validated with several publicly available data sets. The results have demonstrated that the algorithm constitutes a usable model-free approach capable of predicting meaningful relationships between genes, revealing the time-trends of gene regulation. © 2011 Gallo et al; licensee BioMed Central Ltd.Fil: Gallo, Cristian Andrés. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Universidad Nacional del Sur. Departamento de Ciencias e Ingeniería de la Computación; ArgentinaFil: Carballido, Jessica Andrea. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Universidad Nacional del Sur. Departamento de Ciencias e Ingeniería de la Computación; ArgentinaFil: Ponzoni, Ignacio. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro Científico Tecnológico Conicet - Bahía Blanca. Planta Piloto de Ingeniería Química. Universidad Nacional del Sur. Planta Piloto de Ingeniería Química; Argentina. Universidad Nacional del Sur. Departamento de Ciencias e Ingeniería de la Computación; Argentin
    corecore