1,989 research outputs found

    Integrative analysis of complex genomic and epigenomic maps

    Full text link
    Modern healthcare research demands collaboration across disciplines to build preventive measures and innovate predictive capabilities for curing diseases. Along with the emergence of cutting-edge computational and statistical methodologies, data generation and analysis has become cheaper in the last ten years. However, the complexity of big data due to its variety, volume, and velocity creates new challenges for biologists, physicians, bioinformaticians, statisticians, and computer scientists. Combining data from complex multiple profiles is useful to better understand cellular functions and pathways that regulates cell function to provide insights that could not have been obtained using the individual profiles alone. However, current normalization and artifact correction methods are platform and data type specific, and may require both the training and test sets for any application (e.g. biomarker development). This often leads to over-fitting and reduces the reproducibility of genomic findings across studies. In addition, many bias correction and integration approaches require renormalization or reanalysis if additional samples are later introduced. The motivation behind this research was to develop and evaluate strategies for addressing data integration issues across data types and profiling platforms, which should improve healthcare-informatics research and its application in personalized medicine. We have demonstrated a comprehensive and coordinated framework for data standardization across tissue types and profiling platforms. This allows easy integration of data from multiple data generating consortiums. The main goal of this research was to identify regions of genetic-epigenetic co-ordination that are independent of tissue type and consistent across epigenomics profiling data platforms. We developed multi-‘omic’ therapeutic biomarkers for epigenetic drug efficacy by combining our biomarker regions with drug perturbation data generated in our previous studies. We used an adaptive Bayesian factor analysis approach to develop biomarkers for multiple HDACs simultaneously, allowing for predictions of comparative efficacy between the drugs. We showed that this approach leads to different predictions across breast cancer subtypes compared to profiling the drugs separately. We extended this approach on patient samples from multiple public data resources containing epigenetic profiling data from cancer and normal tissues (The Cancer Genome Atlas, TCGA; NIH Roadmap epigenomics data)

    Multi-Omics Integrative Approach of Extracellular Vesicles: A Future Challenging Milestone

    Get PDF
    In the era of multi-omic sciences, dogma on singular cause-effect in physio-pathological processes is overcome and system biology approaches have been providing new perspectives to see through. In this context, extracellular vesicles (EVs) are offering a new level of complexity, given their role in cellular communication and their activity as mediators of specific signals to target cells or tissues. Indeed, their heterogeneity in terms of content, function, origin and potentiality contribute to the cross-interaction of almost every molecular process occurring in a complex system. Such features make EVs proper biological systems being, therefore, optimal targets of omic sciences. Currently, most studies focus on dissecting EVs content in order to either characterize it or to explore its role in various pathogenic processes at transcriptomic, proteomic, metabolomic, lipidomic and genomic levels. Despite valuable results being provided by individual omic studies, the categorization of EVs biological data might represent a limit to be overcome. For this reason, a multi-omic integrative approach might contribute to explore EVs function, their tissue-specific origin and their potentiality. This review summarizes the state-of-the-art of EVs omic studies, addressing recent research on the integration of EVs multi-level biological data and challenging developments in EVs origin

    Toward a Standardized Strategy of Clinical Metabolomics for the Advancement of Precision Medicine

    Get PDF
    Despite the tremendous success, pitfalls have been observed in every step of a clinical metabolomics workflow, which impedes the internal validity of the study. Furthermore, the demand for logistics, instrumentations, and computational resources for metabolic phenotyping studies has far exceeded our expectations. In this conceptual review, we will cover inclusive barriers of a metabolomics-based clinical study and suggest potential solutions in the hope of enhancing study robustness, usability, and transferability. The importance of quality assurance and quality control procedures is discussed, followed by a practical rule containing five phases, including two additional "pre-pre-" and "post-post-" analytical steps. Besides, we will elucidate the potential involvement of machine learning and demonstrate that the need for automated data mining algorithms to improve the quality of future research is undeniable. Consequently, we propose a comprehensive metabolomics framework, along with an appropriate checklist refined from current guidelines and our previously published assessment, in the attempt to accurately translate achievements in metabolomics into clinical and epidemiological research. Furthermore, the integration of multifaceted multi-omics approaches with metabolomics as the pillar member is in urgent need. When combining with other social or nutritional factors, we can gather complete omics profiles for a particular disease. Our discussion reflects the current obstacles and potential solutions toward the progressing trend of utilizing metabolomics in clinical research to create the next-generation healthcare system.11Ysciescopu

    Key biological processes driving metastatic spread of pancreatic cancer as identified by multi-omics studies.

    Get PDF
    Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive malignancy, characterized by a high metastatic burden, already at the time of diagnosis. The metastatic potential of PDAC is one of the main reasons for the poor outcome next to lack of significant improvement in effective treatments in the last decade. Key mutated driver genes, such as activating KRAS mutations, are concordantly expressed in primary and metastatic tumors. However, the biology behind the metastatic potential of PDAC is not fully understood. Recently, large-scale omic approaches have revealed new mechanisms by which PDAC cells gain their metastatic potency. In particular, genomic studies have shown that multiple heterogeneous subclones reside in the primary tumor with different metastatic potential. The development of metastases may be correlated to a more mesenchymal transcriptomic subtype. However, for cancer cells to survive in a distant organ, metastatic sites need to be modulated into pre-metastatic niches. Proteomic studies identified the influence of exosomes on the Kuppfer cells in the liver, which could function to prepare this tissue for metastatic colonization. Phosphoproteomics adds an extra layer to the established omic techniques by unravelling key functional signaling. Future studies integrating results from these large-scale omic approaches will hopefully improve PDAC prognosis through identification of new therapeutic targets and patient selection tools. In this article, we will review the current knowledge on the biology of PDAC metastasis unravelled by large scale multi-omic approaches

    Machine Learning and Integrative Analysis of Biomedical Big Data.

    Get PDF
    Recent developments in high-throughput technologies have accelerated the accumulation of massive amounts of omics data from multiple sources: genome, epigenome, transcriptome, proteome, metabolome, etc. Traditionally, data from each source (e.g., genome) is analyzed in isolation using statistical and machine learning (ML) methods. Integrative analysis of multi-omics and clinical data is key to new biomedical discoveries and advancements in precision medicine. However, data integration poses new computational challenges as well as exacerbates the ones associated with single-omics studies. Specialized computational approaches are required to effectively and efficiently perform integrative analysis of biomedical data acquired from diverse modalities. In this review, we discuss state-of-the-art ML-based approaches for tackling five specific computational challenges associated with integrative analysis: curse of dimensionality, data heterogeneity, missing data, class imbalance and scalability issues

    Supervised Methods for Biomarker Detection from Microarray Experiments

    Get PDF
    Biomarkers are valuable indicators of the state of a biological system. Microarray technology has been extensively used to identify biomarkers and build computational predictive models for disease prognosis, drug sensitivity and toxicity evaluations. Activation biomarkers can be used to understand the underlying signaling cascades, mechanisms of action and biological cross talk. Biomarker detection from microarray data requires several considerations both from the biological and computational points of view. In this chapter, we describe the main methodology used in biomarkers discovery and predictive modeling and we address some of the related challenges. Moreover, we discuss biomarker validation and give some insights into multiomics strategies for biomarker detection.Non peer reviewe

    Computational Models for Transplant Biomarker Discovery.

    Get PDF
    Translational medicine offers a rich promise for improved diagnostics and drug discovery for biomedical research in the field of transplantation, where continued unmet diagnostic and therapeutic needs persist. Current advent of genomics and proteomics profiling called "omics" provides new resources to develop novel biomarkers for clinical routine. Establishing such a marker system heavily depends on appropriate applications of computational algorithms and software, which are basically based on mathematical theories and models. Understanding these theories would help to apply appropriate algorithms to ensure biomarker systems successful. Here, we review the key advances in theories and mathematical models relevant to transplant biomarker developments. Advantages and limitations inherent inside these models are discussed. The principles of key -computational approaches for selecting efficiently the best subset of biomarkers from high--dimensional omics data are highlighted. Prediction models are also introduced, and the integration of multi-microarray data is also discussed. Appreciating these key advances would help to accelerate the development of clinically reliable biomarker systems
    corecore