207 research outputs found

    Reverse Engineering of Biological Systems

    Get PDF
    Gene regulatory network (GRN) consists of a set of genes and regulatory relationships between the genes. As outputs of the GRN, gene expression data contain important information that can be used to reconstruct the GRN to a certain degree. However, the reverse engineer of GRNs from gene expression data is a challenging problem in systems biology. Conventional methods fail in inferring GRNs from gene expression data because of the relative less number of observations compared with the large number of the genes. The inherent noises in the data make the inference accuracy relatively low and the combinatorial explosion nature of the problem makes the inference task extremely difficult. This study aims at reconstructing the GRNs from time-course gene expression data based on GRN models using system identification and parameter estimation methods. The main content consists of three parts: (1) a review of the methods for reverse engineering of GRNs, (2) reverse engineering of GRNs based on linear models and (3) reverse engineering of GRNs based on a nonlinear model, specifically S-systems. In the first part, after the necessary background and challenges of the problem are introduced, various methods for the inference of GRNs are comprehensively reviewed from two aspects: models and inference algorithms. The advantages and disadvantages of each method are discussed. The second part focus on inferring GRNs from time-course gene expression data based on linear models. First, the statistical properties of two sparse penalties, adaptive LASSO and SCAD, with an autoregressive model are studied. It shows that the proposed methods using these two penalties can asymptotically reconstruct the underlying networks. This provides a solid foundation for these methods and their extensions. Second, the integration of multiple datasets should be able to improve the accuracy of the GRN inference. A novel method, Huber group LASSO, is developed to infer GRNs from multiple time-course data, which is also robust to large noises and outliers that the data may contain. An efficient algorithm is also developed and its convergence analysis is provided. The third part can be further divided into two phases: estimating the parameters of S-systems with system structure known and inferring the S-systems without knowing the system structure. Two methods, alternating weighted least squares (AWLS) and auxiliary function guided coordinate descent (AFGCD), have been developed to estimate the parameters of S-systems from time-course data. AWLS takes advantage of the special structure of S-systems and significantly outperforms one existing method, alternating regression (AR). AFGCD uses the auxiliary function and coordinate descent techniques to get the smart and efficient iteration formula and its convergence is theoretically guaranteed. Without knowing the system structure, taking advantage of the special structure of the S-system model, a novel method, pruning separable parameter estimation algorithm (PSPEA) is developed to locally infer the S-systems. PSPEA is then combined with continuous genetic algorithm (CGA) to form a hybrid algorithm which can globally reconstruct the S-systems

    Unique networks: a method to identity disease-specific regulatory networks from microarray data

    Get PDF
    This thesis was submitted for the degree of Doctor of Philosophy and awarded by Brunel University.The survival of any organismis determined by the mechanisms triggered in response to the inputs received. Underlying mechanisms are described by graphical networks that can be inferred from different types of data such as microarrays. Deriving robust and reliable networks can be complicated due to the microarray structure of the data characterized by a discrepancy between the number of genes and samples of several orders of magnitude, bias and noise. Researchers overcome this problem by integrating independent data together and deriving the common mechanisms through consensus network analysis. Different conditions generate different inputs to the organism which reacts triggering different mechanisms with similarities and differences. A lot of effort has been spent into identifying the commonalities under different conditions. Highlighting similarities may overshadow the differences which often identify the main characteristics of the triggered mechanisms. In this thesis we introduce the concept of study-specific mechanism. We develop a pipeline to semiautomatically identify study-specific networks called unique-networks through a combination of consensus approach, graphical similarities and network analysis. The main pipeline called UNIP (Unique Networks Identification Pipeline) takes a set of independent studies, builds gene regulatory networks for each of them, calculates an adaptation of the sensitivity measure based on the networks graphical similarities, applies clustering to group the studies who generate the most similar networks into study-clusters and derives the consensus networks. Once each study-cluster is associated with a consensus-network, we identify the links that appear only in the consensus network under consideration but not in the others (unique-connections). Considering the genes involved in the unique-connections we build Bayesian networks to derive the unique-networks. Finally, we exploit the inference tool to calculate each gene prediction-accuracy across all studies to further refine the unique-networks. Biological validation through different software and the literature are explored to validate our method. UNIP is first applied to a set of synthetic data perturbed with different levels of noise to study the performance and verify its reliability. Then, wheat under stress conditions and different types of cancer are explored. Finally, we develop a user-friendly interface to combine the set of studies by using AND and NOT logic operators. Based on the findings, UNIP is a robust and reliable method to analyse large sets of transcriptomic data. It easily detects the main complex relationships between transcriptional expression of genes specific for different conditions and also highlights structures and nodes that could be potential targets for further research

    Predictive network modeling of the high-resolution dynamic plant transcriptome in response to nitrate

    Get PDF
    International audienceABSTRACT: BACKGROUND: Nitrate, acting as both a nitrogen source and a signaling molecule, controls many aspects of plant development. However, gene networks involved in plant adaptation to fluctuating nitrate environments have not yet been identified. RESULTS: Here we use time-series transcriptome data to decipher gene relationships and consequently to build core regulatory networks involved in Arabidopsis root adaptation to nitrate provision. The experimental approach has been to monitor genome-wide responses to nitrate at 3, 6, 9, 12, 15 and 20 minutes, using Affymetrix ATH1 gene chips. This high-resolution time course analysis demonstrated that the previously known primary nitrate response is actually preceded by a very fast gene expression modulation, involving genes and functions needed to prepare plants to use or reduce nitrate. A state-space model inferred from this microarray time-series data successfully predicts gene behavior in unlearnt conditions. CONCLUSIONS: The experiments and methods allow us to propose a temporal working model for nitrate-driven gene networks. This network model is tested both in silico and experimentally. For example, the over-expression of a predicted gene hub encoding a transcription factor induced early in the cascade indeed leads to the modification of the kinetic nitrate response of sentinel genes such as NIR, NIA2, and NRT1.1, and several other transcription factors. The potential nitrate /hormone connections implicated by this time-series data is also evaluated

    INTEGRATIVE ANALYSIS OF OMICS DATA IN ADULT GLIOMA AND OTHER TCGA CANCERS TO GUIDE PRECISION MEDICINE

    Get PDF
    Transcriptomic profiling and gene expression signatures have been widely applied as effective approaches for enhancing the molecular classification, diagnosis, prognosis or prediction of therapeutic response towards personalized therapy for cancer patients. Thanks to modern genome-wide profiling technology, scientists are able to build engines leveraging massive genomic variations and integrating with clinical data to identify “at risk” individuals for the sake of prevention, diagnosis and therapeutic interventions. In my graduate work for my Ph.D. thesis, I have investigated genomic sequencing data mining to comprehensively characterise molecular classifications and aberrant genomic events associated with clinical prognosis and treatment response, through applying high-dimensional omics genomic data to promote the understanding of gene signatures and somatic molecular alterations contributing to cancer progression and clinical outcomes. Following this motivation, my dissertation has been focused on the following three topics in translational genomics. 1) Characterization of transcriptomic plasticity and its association with the tumor microenvironment in glioblastoma (GBM). I have integrated transcriptomic, genomic, protein and clinical data to increase the accuracy of GBM classification, and identify the association between the GBM mesenchymal subtype and reduced tumorpurity, accompanied with increased presence of tumor-associated microglia. Then I have tackled the sole source of microglial as intrinsic tumor bulk but not their corresponding neurosphere cells through both transcriptional and protein level analysis using a panel of sphere-forming glioma cultures and their parent GBM samples.FurthermoreI have demonstrated my hypothesis through longitudinal analysis of paired primary and recurrent GBM samples that the phenotypic alterations of GBM subtypes are not due to intrinsic proneural-to-mesenchymal transition in tumor cells, rather it is intertwined with increased level of microglia upon disease recurrence. Collectively I have elucidated the critical role of tumor microenvironment (Microglia and macrophages from central nervous system) contributing to the intra-tumor heterogeneity and accurate classification of GBM patients based on transcriptomic profiling, which will not only significantly impact on clinical perspective but also pave the way for preclinical cancer research. 2) Identification of prognostic gene signatures that stratify adult diffuse glioma patientsharboring1p/19q co-deletions. I have compared multiple statistical methods and derived a gene signature significantly associated with survival by applying a machine learning algorithm. Then I have identified inflammatory response and acetylation activity that associated with malignant progression of 1p/19q co-deleted glioma. In addition, I showed this signature translates to other types of adult diffuse glioma, suggesting its universality in the pathobiology of other subset gliomas. My efforts on integrative data analysis of this highly curated data set usingoptimizedstatistical models will reflect the pending update to WHO classification system oftumorsin the central nervous system (CNS). 3) Comprehensive characterization of somatic fusion transcripts in Pan-Cancers. I have identified a panel of novel fusion transcripts across all of TCGA cancer types through transcriptomic profiling. Then I have predicted fusion proteins with kinase activity and hub function of pathway network based on the annotation of genetically mobile domains and functional domain architectures. I have evaluated a panel of in -frame gene fusions as potential driver mutations based on network fusion centrality hypothesis. I have also characterised the emerging complexity of genetic architecture in fusion transcripts through integrating genomic structure and somatic variants and delineating the distinct genomic patterns of fusion events across different cancer types. Overall my exploration of the pathogenetic impact and clinical relevance of candidate gene fusions have provided fundamental insights into the management of a subset of cancer patients by predicting the oncogenic signalling and specific drug targets encoded by these fusion genes. Taken together, the translational genomic research I have conducted during my Ph.D. study will shed new light on precision medicine and contribute to the cancer research community. The novel classification concept, gene signature and fusion transcripts I have identified will address several hotly debated issues in translational genomics, such as complex interactions between tumor bulks and their adjacent microenvironments, prognostic markers for clinical diagnostics and personalized therapy, distinct patterns of genomic structure alterations and oncogenic events in different cancer types, therefore facilitating our understanding of genomic alterations and moving us towards the development of precision medicine

    RGBM: regularized gradient boosting machines for identification of the transcriptional regulators of discrete glioma subtypes

    Get PDF
    We propose a generic framework for gene regulatory network (GRN) inference approached as a feature selection problem. GRNs obtained using Machine Learning techniques are often dense, whereas real GRNs are rather sparse. We use a Tikonov regularization inspired optimal L-curve criterion that utilizes the edge weight distribution for a given target gene to determine the optimal set of TFs associated with it. Our proposed framework allows to incorporate a mechanistic active biding network based on cis-regulatory motif analysis. We evaluate our regularization framework in conjunction with two non-linear ML techniques, namely gradient boosting machines (GBM) and random-forests (GENIE), resulting in a regularized feature selection based method specifically called RGBM and RGENIE respectively. RGBM has been used to identify the main transcription factors that are causally involved as master regulators of the gene expression signature activated in the FGFR3-TACC3-positive glioblastoma. Here, we illustrate that RGBM identifies the main regulators of the molecular subtypes of brain tumors. Our analysis reveals the identity and corresponding biological activities of the master regulators characterizing the difference between G-CIMP-high and G-CIMP-low subtypes and between PA-like and LGm6-GBM, thus providing a clue to the yet undetermined nature of the transcriptional events among these subtypes

    Identifying Biological Network Structure, Predicting Network Behavior, and Classifying Network State With High Dimensional Model Representation (HDMR)

    Get PDF
    This work presents an adapted Random Sampling - High Dimensional Model Representation (RS-HDMR) algorithm for synergistically addressing three key problems in network biology: (1) identifying the structure of biological networks from multivariate data, (2) predicting network response under previously unsampled conditions, and (3) inferring experimental perturbations based on the observed network state. RS-HDMR is a multivariate regression method that decomposes network interactions into a hierarchy of non-linear component functions. Sensitivity analysis based on these functions provides a clear physical and statistical interpretation of the underlying network structure. The advantages of RS-HDMR include efficient extraction of nonlinear and cooperative network relationships without resorting to discretization, prediction of network behavior without mechanistic modeling, robustness to data noise, and favorable scalability of the sampling requirement with respect to network size. As a proof-of-principle study, RS-HDMR was applied to experimental data measuring the single-cell response of a protein-protein signaling network to various experimental perturbations. A comparison to network structure identified in the literature and through other inference methods, including Bayesian and mutual-information based algorithms, suggests that RS-HDMR can successfully reveal a network structure with a low false positive rate while still capturing non-linear and cooperative interactions. RS-HDMR identified several higher-order network interactions that correspond to known feedback regulations among multiple network species and that were unidentified by other network inference methods. Furthermore, RS-HDMR has a better ability to predict network response under unsampled conditions in this application than the best statistical inference algorithm presented in the recent DREAM3 signaling-prediction competition. RS-HDMR can discern and predict differences in network state that arise from sources ranging from intrinsic cell-cell variability to altered experimental conditions, such as when drug perturbations are introduced. This ability ultimately allows RS-HDMR to accurately classify the experimental conditions of a given sample based on its observed network state

    Integrative approaches for systematic reconstruction of regulatory circuits in mammals

    Get PDF
    Thesis (Ph. D.)--Massachusetts Institute of Technology, Computational and Systems Biology Program, 2012.Cataloged from PDF version of thesis.Includes bibliographical references (p. 141-149).The reconstruction of regulatory networks is one of the most challenging tasks in systems biology. Although some models for inferring regulatory networks can make useful predictions about the wiring and mechanisms of molecular interactions, these approaches are still limited and there is a strong need to develop increasingly universal and accurate approaches for network reconstruction. This problem is particularly challenging in mammals, due to the higher complexity of mammalian regulatory networks and limitations in experimental manipulation. In this thesis, I present three systematic approachs to reconstruct, analyse and refine models of gene regulation. In Chapter 1, I devise a method for deriving an observational model from temporal genomic profiles. I use it to choose targets for perturbation experiments in order to determine a network controlling the responses of mouse primary dendritic cells to stimulation with pathogen components. In Chapter 2, I introduce the algorithm Exigo, for identifying essential interactions in regulatory networks reconstructed from experimental data where regulators have been silenced, using a network reduction strategy. Exigo outperforms previous approaches on simulated data, uncovers the core network structure when applied to real networks derived from perturbation studies in mammals, and improves the performance of network inference methods. Lastly, I introduce in Chapter 3 an approach to learn a module network from multiple highthroughput assays. Analysis of a diffuse large B-cell lymphoma dataset identifies candidate regulator genes, microRNAs and copy number aberrations with biological, and possibly therapeutic, importance.by Ana Paula Santos Botelho Oliveira Leite.Ph.D
    corecore