1,890 research outputs found

    Co-clustering algorithm for the identification of cancer subtypes from gene expression data

    Get PDF
    Cancer has been classified as a heterogeneous genetic disease comprising various different subtypes based on gene expression data. Early stages of diagnosis and prognosis for cancer type have become an essential requirement in cancer informatics research because it is helpful for the clinical treatment of patients. Besides this, gene network interaction which is the significant in order to understand the cellular and progressive mechanisms of cancer has been barely considered in current research. Hence, applications of machine learning methods become an important area for researchers to explore in order to categorize cancer genes into high and low risk groups or subtypes. Presently co-clustering is an extensively used data mining technique for analyzing gene expression data. This paper presents an improved network assisted co-clustering for the identification of cancer subtypes (iNCIS) where it combines gene network information with gene expression data to obtain co-clusters. The effectiveness of iNCIS was evaluated on large-scale Breast Cancer (BRCA) and Glioblastoma Multiforme (GBM). This weighted co-clustering approach in iNCIS delivers a distinctive result to integrate gene network into the clustering procedure

    Identifying Cancer Subtypes Using Unsupervised Deep Learning

    Get PDF
    Glioblastoma multiforme (GBM) is the most fatal malignant type of brain tumor with a very poor prognosis with a median survival of around one year. Numerous studies have reported tumor subtypes that consider different characteristics on individual patients, which may play important roles in determining the survival rates in GBM. In this study, we present a pathway-based clustering method using Restricted Boltzmann Machine (RBM), called R-PathCluster, for identifying unknown subtypes with pathway markers of gene expressions. In order to assess the performance of R-PathCluster, we conducted experiments with several clustering methods such as k-means, hierarchical clustering, and RBM models with different input data. R-PathCluster showed the best performance in clustering longterm and short-term survivals, although its clustering score was not the highest among them in experiments. R-PathCluster provides a solution to interpret the model in biological sense, since it takes pathway markers that represent biological process of pathways. We discussed that our findings from R-PathCluster are supported by many biological literatures. Keywords. Glioblastoma multiforme, tumor subtypes, clustering, Restricted Boltzmann Machin

    Development of a simple artificial intelligence method to accurately subtype breast cancers based on gene expression barcodes

    Get PDF
    >Magister Scientiae - MScINTRODUCTION: Breast cancer is a highly heterogeneous disease. The complexity of achieving an accurate diagnosis and an effective treatment regimen lies within this heterogeneity. Subtypes of the disease are not simply molecular, i.e. hormone receptor over-expression or absence, but the tumour itself is heterogeneous in terms of tissue of origin, metastases, and histopathological variability. Accurate tumour classification vastly improves treatment decisions, patient outcomes and 5-year survival rates. Gene expression studies aided by transcriptomic technologies such as microarrays and next-generation sequencing (e.g. RNA-Sequencing) have aided oncology researcher and clinician understanding of the complex molecular portraits of malignant breast tumours. Mechanisms governing cancers, which include tumorigenesis, gene fusions, gene over-expression and suppression, cellular process and pathway involvementinvolvement, have been elucidated through comprehensive analyses of the cancer transcriptome. Over the past 20 years, gene expression signatures, discovered with both microarray and RNA-Seq have reached clinical and commercial application through the development of tests such as Mammaprint®, OncotypeDX®, and FoundationOne® CDx, all which focus on chemotherapy sensitivity, prediction of cancer recurrence, and tumour mutational level. The Gene Expression Barcode (GExB) algorithm was developed to allow for easy interpretation and integration of microarray data through data normalization with frozen RMA (fRMA) preprocessing and conversion of relative gene expression to a sequence of 1's and 0's. Unfortunately, the algorithm has not yet been developed for RNA-Seq data. However, implementation of the GExB with feature-selection would contribute to a machine-learning based robust breast cancer and subtype classifier. METHODOLOGY: For microarray data, we applied the GExB algorithm to generate barcodes for normal breast and breast tumour samples. A two-class classifier for malignancy was developed through feature-selection on barcoded samples by selecting for genes with 85% stable absence or presence within a tissue type, and differentially stable between tissues. A multi-class feature-selection method was employed to identify genes with variable expression in one subtype, but 80% stable absence or presence in all other subtypes, i.e. 80% in n-1 subtypes. For RNA-Seq data, a barcoding method needed to be developed which could mimic the GExB algorithm for microarray data. A z-score-to-barcode method was implemented and differential gene expression analysis with selection of the top 100 genes as informative features for classification purposes. The accuracy and discriminatory capability of both microarray-based gene signatures and the RNA-Seq-based gene signatures was assessed through unsupervised and supervised machine-learning algorithms, i.e., K-means and Hierarchical clustering, as well as binary and multi-class Support Vector Machine (SVM) implementations. RESULTS: The GExB-FS method for microarray data yielded an 85-probe and 346-probe informative set for two-class and multi-class classifiers, respectively. The two-class classifier predicted samples as either normal or malignant with 100% accuracy and the multi-class classifier predicted molecular subtype with 96.5% accuracy with SVM. Combining RNA-Seq DE analysis for feature-selection with the z-score-to-barcode method, resulted in a two-class classifier for malignancy, and a multi-class classifier for normal-from-healthy, normal-adjacent-tumour (from cancer patients), and breast tumour samples with 100% accuracy. Most notably, a normal-adjacent-tumour gene expression signature emerged, which differentiated it from normal breast tissues in healthy individuals. CONCLUSION: A potentially novel method for microarray and RNA-Seq data transformation, feature selection and classifier development was established. The universal application of the microarray signatures and validity of the z-score-to-barcode method was proven with 95% accurate classification of RNA-Seq barcoded samples with a microarray discovered gene expression signature. The results from this comprehensive study into the discovery of robust gene expression signatures holds immense potential for further R&F towards implementation at the clinical endpoint, and translation to simpler and cost-effective laboratory methods such as qtPCR-based tests

    Integration of molecular network data reconstructs Gene Ontology.

    Get PDF
    Motivation: Recently, a shift was made from using Gene Ontology (GO) to evaluate molecular network data to using these data to construct and evaluate GO. Dutkowski et al. provide the first evidence that a large part of GO can be reconstructed solely from topologies of molecular networks. Motivated by this work, we develop a novel data integration framework that integrates multiple types of molecular network data to reconstruct and update GO. We ask how much of GO can be recovered by integrating various molecular interaction data. Results: We introduce a computational framework for integration of various biological networks using penalized non-negative matrix tri-factorization (PNMTF). It takes all network data in a matrix form and performs simultaneous clustering of genes and GO terms, inducing new relations between genes and GO terms (annotations) and between GO terms themselves. To improve the accuracy of our predicted relations, we extend the integration methodology to include additional topological information represented as the similarity in wiring around non-interacting genes. Surprisingly, by integrating topologies of bakers’ yeasts protein–protein interaction, genetic interaction (GI) and co-expression networks, our method reports as related 96% of GO terms that are directly related in GO. The inclusion of the wiring similarity of non-interacting genes contributes 6% to this large GO term association capture. Furthermore, we use our method to infer new relationships between GO terms solely from the topologies of these networks and validate 44% of our predictions in the literature. In addition, our integration method reproduces 48% of cellular component, 41% of molecular function and 41% of biological process GO terms, outperforming the previous method in the former two domains of GO. Finally, we predict new GO annotations of yeast genes and validate our predictions through GIs profiling. Availability and implementation: Supplementary Tables of new GO term associations and predicted gene annotations are available at http://bio-nets.doc.ic.ac.uk/GO-Reconstruction/. Contact: [email protected] Supplementary information: Supplementary data are available at Bioinformatics online

    Development of Biclustering Techniques for Gene Expression Data Modeling and Mining

    Get PDF
    The next-generation sequencing technologies can generate large-scale biological data with higher resolution, better accuracy, and lower technical variation than the arraybased counterparts. RNA sequencing (RNA-Seq) can generate genome-scale gene expression data in biological samples at a given moment, facilitating a better understanding of cell functions at genetic and cellular levels. The abundance of gene expression datasets provides an opportunity to identify genes with similar expression patterns across multiple conditions, i.e., co-expression gene modules (CEMs). Genomescale identification of CEMs can be modeled and solved by biclustering, a twodimensional data mining technique that allows clustering of rows and columns in a gene expression matrix, simultaneously. Compared with traditional clustering that targets global patterns, biclustering can predict local patterns. This unique feature makes biclustering very useful when applied to big gene expression data since genes that participate in a cellular process are only active in specific conditions, thus are usually coexpressed under a subset of all conditions. The combination of biclustering and large-scale gene expression data holds promising potential for condition-specific functional pathway/network analysis. However, existing biclustering tools do not have satisfied performance on high-resolution RNA-Seq data, majorly due to the lack of (i) a consideration of high sparsity of RNA-Seq data, especially for scRNA-Seq data, and (ii) an understanding of the underlying transcriptional regulation signals of the observed gene expression values. QUBIC2, a novel biclustering algorithm, is designed for large-scale bulk RNA-Seq and single-cell RNA-seq (scRNA-Seq) data analysis. Critical novelties of the algorithm include (i) used a truncated model to handle the unreliable quantification of genes with low or moderate expression; (ii) adopted the Gaussian mixture distribution and an information-divergency objective function to capture shared transcriptional regulation signals among a set of genes; (iii) utilized a Dual strategy to expand the core biclusters, aiming to save dropouts from the background; and (iv) developed a statistical framework to evaluate the significances of all the identified biclusters. Method validation on comprehensive data sets suggests that QUBIC2 had superior performance in functional modules detection and cell type classification. The applications of temporal and spatial data demonstrated that QUBIC2 could derive meaningful biological information from scRNA-Seq data. Also presented in this dissertation is QUBICR. This R package is characterized by an 82% average improved efficiency compared to the source C code of QUBIC. It provides a set of comprehensive functions to facilitate biclustering-based biological studies, including the discretization of expression data, query-based biclustering, bicluster expanding, biclusters comparison, heatmap visualization of any identified biclusters, and co-expression networks elucidation. In the end, a systematical summary is provided regarding the primary applications of biclustering for biological data and more advanced applications for biomedical data. It will assist researchers to effectively analyze their big data and generate valuable biological knowledge and novel insights with higher efficiency

    Genomic and proteomic analysis with dynamically growing self organising tree (DGSOT) for measuring clinical outcomes of cancer

    Get PDF
    Genomics and proteomics microarray technologies are used for analysing molecular and cellular expressions of cancer. This creates a challenge for analysis and interpretation of the data generated as it is produced in large volumes. The current review describes a combined system for genetic, molecular interpretation and analysis of genomics and proteomics technologies that offers a wide range of interpreted results. Artificial neural network systems technology has the type of programmes to best deal with these large volumes of analytical data. The artificial system to be recommended here is to be determined from the analysis and selection of the best of different available technologies currently being used or reviewed for microarray data analysis. The system proposed here is a tree structure, a new hierarchical clustering algorithm called a dynamically growing self-organizing tree (DGSOT) algorithm, which overcomes drawbacks of traditional hierarchical clustering algorithms. The DGSOT algorithm combines horizontal and vertical growth to construct a mutlifurcating hierarchical tree from top to bottom to cluster the data. They are designed to combine the strengths of Neural Networks (NN), which have speed and robustness to noise, and hierarchical clustering tree structure which are minimum prior requirement for number of clusters specification and training in order to output results of interpretable biological context. The combined system will generate an output of biological interpretation of expression profiles associated with diagnosis of disease (including early detection, molecular classification and staging), metastasis (spread of the disease to non-adjacent organs and/or tissues), prognosis (predicting clinical outcome) and response to treatment; it also gives possible therapeutic options ranking them according to their benefits for the patient.Key words: Genomics, proteomics, microarray, dynamically growing self-organizing tree (DGSOT)

    Analysis of large-scale molecular biological data using self-organizing maps

    Get PDF
    Modern high-throughput technologies such as microarrays, next generation sequencing and mass spectrometry provide huge amounts of data per measurement and challenge traditional analyses. New strategies of data processing, visualization and functional analysis are inevitable. This thesis presents an approach which applies a machine learning technique known as self organizing maps (SOMs). SOMs enable the parallel sample- and feature-centered view of molecular phenotypes combined with strong visualization and second-level analysis capabilities. We developed a comprehensive analysis and visualization pipeline based on SOMs. The unsupervised SOM mapping projects the initially high number of features, such as gene expression profiles, to meta-feature clusters of similar and hence potentially co-regulated single features. This reduction of dimension is attained by the re-weighting of primary information and does not entail a loss of primary information in contrast to simple filtering approaches. The meta-data provided by the SOM algorithm is visualized in terms of intuitive mosaic portraits. Sample-specific and common properties shared between samples emerge as a handful of localized spots in the portraits collecting groups of co-regulated and co-expressed meta-features. This characteristic color patterns reflect the data landscape of each sample and promote immediate identification of (meta-)features of interest. It will be demonstrated that SOM portraits transform large and heterogeneous sets of molecular biological data into an atlas of sample-specific texture maps which can be directly compared in terms of similarities and dissimilarities. Spot-clusters of correlated meta-features can be extracted from the SOM portraits in a subsequent step of aggregation. This spot-clustering effectively enables reduction of the dimensionality of the data in two subsequent steps towards a handful of signature modules in an unsupervised fashion. Furthermore we demonstrate that analysis techniques provide enhanced resolution if applied to the meta-features. The improved discrimination power of meta-features in downstream analyses such as hierarchical clustering, independent component analysis or pairwise correlation analysis is ascribed to essentially two facts: Firstly, the set of meta-features better represents the diversity of patterns and modes inherent in the data and secondly, it also possesses the better signal-to-noise characteristics as a comparable collection of single features. Additionally to the pattern-driven feature selection in the SOM portraits, we apply statistical measures to detect significantly differential features between sample classes. Implementation of scoring measurements supplements the basal SOM algorithm. Further, two variants of functional enrichment analyses are introduced which link sample specific patterns of the meta-feature landscape with biological knowledge and support functional interpretation of the data based on the ‘guilt by association’ principle. Finally, case studies selected from different ‘OMIC’ realms are presented in this thesis. In particular, molecular phenotype data derived from expression microarrays (mRNA, miRNA), sequencing (DNA methylation, histone modification patterns) or mass spectrometry (proteome), and also genotype data (SNP-microarrays) is analyzed. It is shown that the SOM analysis pipeline implies strong application capabilities and covers a broad range of potential purposes ranging from time series and treatment-vs.-control experiments to discrimination of samples according to genotypic, phenotypic or taxonomic classifications

    Towards generalizable machine learning models for computer-aided diagnosis in medicine

    Get PDF
    Hidden stratification represents a phenomenon in which a training dataset contains unlabeled (hidden) subsets of cases that may affect machine learning model performance. Machine learning models that ignore the hidden stratification phenomenon--despite promising overall performance measured as accuracy and sensitivity--often fail at predicting the low prevalence cases, but those cases remain important. In the medical domain, patients with diseases are often less common than healthy patients, and a misdiagnosis of a patient with a disease can have significant clinical impacts. Therefore, to build a robust and trustworthy CAD system and a reliable treatment effect prediction model, we cannot only pursue machine learning models with high overall accuracy, but we also need to discover any hidden stratification in the data and evaluate the proposing machine learning models with respect to both overall performance and the performance on certain subsets (groups) of the data, such as the ‘worst group’. In this study, I investigated three approaches for data stratification: a novel algorithmic deep learning (DL) approach that learns similarities among cases and two schema completion approaches that utilize domain expert knowledge. I further proposed an innovative way to integrate the discovered latent groups into the loss functions of DL models to allow for better model generalizability under the domain shift scenario caused by the data heterogeneity. My results on lung nodule Computed Tomography (CT) images and breast cancer histopathology images demonstrate that learning homogeneous groups within heterogeneous data significantly improves the performance of the computer-aided diagnosis (CAD) system, particularly for low-prevalence or worst-performing cases. This study emphasizes the importance of discovering and learning the latent stratification within the data, as it is a critical step towards building ML models that are generalizable and reliable. Ultimately, this discovery can have a profound impact on clinical decision-making, particularly for low-prevalence cases
    • …
    corecore