262 research outputs found

    To metabolomics and beyond: a technological portfolio to investigate cancer metabolism

    Get PDF
    Tumour cells have exquisite flexibility in reprogramming their metabolism in order to support tumour initiation, progression, metastasis and resistance to therapies. These reprogrammed activities include a complete rewiring of the bioenergetic, biosynthetic and redox status to sustain the increased energetic demand of the cells. Over the last decades, the cancer metabolism field has seen an explosion of new biochemical technologies giving more tools than ever before to navigate this complexity. Within a cell or a tissue, the metabolites constitute the direct signature of the molecular phenotype and thus their profiling has concrete clinical applications in oncology. Metabolomics and fluxomics, are key technological approaches that mainly revolutionized the field enabling researchers to have both a qualitative and mechanistic model of the biochemical activities in cancer. Furthermore, the upgrade from bulk to single-cell analysis technologies provided unprecedented opportunity to investigate cancer biology at cellular resolution allowing an in depth quantitative analysis of complex and heterogenous diseases. More recently, the advent of functional genomic screening allowed the identification of molecular pathways, cellular processes, biomarkers and novel therapeutic targets that in concert with other technologies allow patient stratification and identification of new treatment regimens. This review is intended to be a guide for researchers to cancer metabolism, highlighting current and emerging technologies, emphasizing advantages, disadvantages and applications with the potential of leading the development of innovative anti-cancer therapies

    Ovarian Cancer Data Analysis using Deep Learning: A Systematic Review from the Perspectives of Key Features of Data Analysis and AI Assurance

    Full text link
    Background and objectives: By extracting this information, Machine or Deep Learning (ML/DL)-based autonomous data analysis tools can assist clinicians and cancer researchers in discovering patterns and relationships from complex data sets. Many DL-based analyses on ovarian cancer (OC) data have recently been published. These analyses are highly diverse in various aspects of cancer (e.g., subdomain(s) and cancer type they address) and data analysis features. However, a comprehensive understanding of these analyses in terms of these features and AI assurance (AIA) is currently lacking. This systematic review aims to fill this gap by examining the existing literature and identifying important aspects of OC data analysis using DL, explicitly focusing on the key features and AI assurance perspectives. Methods: The PRISMA framework was used to conduct comprehensive searches in three journal databases. Only studies published between 2015 and 2023 in peer-reviewed journals were included in the analysis. Results: In the review, a total of 96 DL-driven analyses were examined. The findings reveal several important insights regarding DL-driven ovarian cancer data analysis: - Most studies 71% (68 out of 96) focused on detection and diagnosis, while no study addressed the prediction and prevention of OC. - The analyses were predominantly based on samples from a non-diverse population (75% (72/96 studies)), limited to a geographic location or country. - Only a small proportion of studies (only 33% (32/96)) performed integrated analyses, most of which used homogeneous data (clinical or omics). - Notably, a mere 8.3% (8/96) of the studies validated their models using external and diverse data sets, highlighting the need for enhanced model validation, and - The inclusion of AIA in cancer data analysis is in a very early stage; only 2.1% (2/96) explicitly addressed AIA through explainability

    Integration of epigenetic regulatory mechanisms in heart failure

    Get PDF
    The number of “omics” approaches is continuously growing. Among others, epigenetics has appeared as an attractive area of investigation by the cardiovascular research community, notably considering its association with disease development. Complex diseases such as cardiovascular diseases have to be tackled using methods integrating different omics levels, so called “multi-omics” approaches. These approaches combine and co-analyze different levels of disease regulation. In this review, we present and discuss the role of epigenetic mechanisms in regulating gene expression and provide an integrated view of how these mechanisms are interlinked and regulate the development of cardiac disease, with a particular attention to heart failure. We focus on DNA, histone, and RNA modifications, and discuss the current methods and tools used for data integration and analysis. Enhancing the knowledge of these regulatory mechanisms may lead to novel therapeutic approaches and biomarkers for precision healthcare and improved clinical outcomes

    Integration of multi-scale protein interactions for biomedical data analysis

    Get PDF
    With the advancement of modern technologies, we observe an increasing accumulation of biomedical data about diseases. There is a need for computational methods to sift through and extract knowledge from the diverse data available in order to improve our mechanistic understanding of diseases and improve patient care. Biomedical data come in various forms as exemplified by the various omics data. Existing studies have shown that each form of omics data gives only partial information on cells state and motivated jointly mining multi-omics, multi-modal data to extract integrated system knowledge. The interactome is of particular importance as it enables the modelling of dependencies arising from molecular interactions. This Thesis takes a special interest in the multi-scale protein interactome and its integration with computational models to extract relevant information from biomedical data. We define multi-scale interactions at different omics scale that involve proteins: pairwise protein-protein interactions, multi-protein complexes, and biological pathways. Using hypergraph representations, we motivate considering higher-order protein interactions, highlighting the complementary biological information contained in the multi-scale interactome. Based on those results, we further investigate how those multi-scale protein interactions can be used as either prior knowledge, or auxiliary data to develop machine learning algorithms. First, we design a neural network using the multi-scale organization of proteins in a cell into biological pathways as prior knowledge and train it to predict a patient's diagnosis based on transcriptomics data. From the trained models, we develop a strategy to extract biomedical knowledge pertaining to the diseases investigated. Second, we propose a general framework based on Non-negative Matrix Factorization to integrate the multi-scale protein interactome with multi-omics data. We show that our approach outperforms the existing methods, provide biomedical insights and relevant hypotheses for specific cancer types

    Proteogenomic profiling of metastatic melanoma. From protein expression to patient stratification.

    Get PDF

    Incorporating standardised drift-tube ion mobility to enhance non-targeted assessment of the wine metabolome (LC×IM-MS)

    Get PDF
    Liquid chromatography with drift-tube ion mobility spectrometry-mass spectrometry (LCxIM-MS) is emerging as a powerful addition to existing LC-MS workflows for addressing a diverse range of metabolomics-related questions [1,2]. Importantly, excellent precision under repeatability and reproducibility conditions of drift-tube IM separations [3] supports the development of non-targeted approaches for complex metabolome assessment such as wine characterisation [4]. In this work, fundamentals of this new analytical metabolomics approach are introduced and application to the analysis of 90 authentic red and white wine samples originating from Macedonia is presented. Following measurements, intersample alignment of metabolites using non-targeted extraction and three-dimensional alignment of molecular features (retention time, collision cross section, and high-resolution mass spectra) provides confidence for metabolite identity confirmation. Applying a fingerprinting metabolomics workflow allows statistical assessment of the influence of geographic region, variety, and age. This approach is a state-of-the-art tool to assess wine chemodiversity and is particularly beneficial for the discovery of wine biomarkers and establishing product authenticity based on development of fingerprint libraries

    Algorithms and Methods for Robust Processing and Analysis of Mass Spectrometry Data

    Get PDF
    Liquid chromatography-mass spectrometry (LC-MS) and mass spectrometry imaging (MSI) are two techniques that are routinely used to study proteins, peptides, and metabolites at a large scale. Thousands of biological compounds can be identified and quantified in a single experiment with LC-MS, but many studies fail to convert this data to a better understanding of disease biology. One of the primary reasons for this is low reproducibility, which in turn is partially due to inaccurate and/or inconsistent data processing. Protein biomarkers and signatures for various types of cancer are frequently discovered with LC-MS, but their behavior in independent cohorts is often inconsistent to that in the discovery cohort. Biomarker candidates must be thoroughly validated in independent cohorts, which makes the ability to share data across different laboratories crucial to the future success of the MS-based research fields. The emergence and growth of public repositories for MSI data is a step in the rightdirection. Still, many of those data sets remain incompatible one another due to inaccurate or incompatible preprocessing strategies. Ensuring compatibility between data generated in different labs is therefore necessary to gain access to the full potential of MS-based research. In two of the studies that I present in this thesis, we used LC-MS to characterize lymph node metastases from individuals with melanoma. Furthermore, my thesis work has resulted in two novel preprocessing methods for MSI data sets. The first one is a peak detection method that achieves considerably higher sensitivity for faintly expressed compounds than existing methods, and the second one is a accurate, robust, and general approach to mass alignment. Both algorithms deliberately rely on centroid spectra, which makes them compatible with most shared data sets. I believe that the improvements demonstrated by these methods can lead to a higher reproducibility in the MS-based research fields, and, ultimately, to a better understanding of disease processes
    corecore