6,873 research outputs found

    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

    High-throughput plant phenotyping: a role for metabolomics?

    Get PDF
    High-throughput (HTP) plant phenotyping approaches are developing rapidly and are already helping to bridge the genotype–phenotype gap. However, technologies should be developed beyond current physico-spectral evaluations to extend our analytical capacities to the subcellular level. Metabolites define and determine many key physiological and agronomic features in plants and an ability to integrate a metabolomics approach within current HTP phenotyping platforms has huge potential for added value. While key challenges remain on several fronts, novel technological innovations are upcoming yet under-exploited in a phenotyping context. In this review, we present an overview of the state of the art and how current limitations might be overcome to enable full integration of metabolomics approaches into a generic phenotyping pipeline in the near future.info:eu-repo/semantics/publishedVersio

    Pathway and network analysis in proteomics

    Get PDF
    Proteomics is inherently a systems science that studies not only measured protein and their expressions in a cell, but also the interplay of proteins, protein complexes, signaling pathways, and network modules. There is a rapid accumulation of Proteomics data in recent years. However, Proteomics data are highly variable, with results sensitive to data preparation methods, sample condition, instrument types, and analytical methods. To address the challenge in Proteomics data analysis, we review current tools being developed to incorporate biological function and network topological information. We categorize these tools into four types: tools with basic functional information and little topological features (e.g., GO category analysis), tools with rich functional information and little topological features (e.g., GSEA), tools with basic functional information and rich topological features (e.g., Cytoscape), and tools with rich functional information and rich topological features (e.g., PathwayExpress). We first review the potential application of these tools to Proteomics; then we review tools that can achieve automated learning of pathway modules and features, and tools that help perform integrated network visual analytics

    Modeling cancer metabolism on a genome scale

    Get PDF
    Cancer cells have fundamentally altered cellular metabolism that is associated with their tumorigenicity and malignancy. In addition to the widely studied Warburg effect, several new key metabolic alterations in cancer have been established over the last decade, leading to the recognition that altered tumor metabolism is one of the hallmarks of cancer. Deciphering the full scope and functional implications of the dysregulated metabolism in cancer requires both the advancement of a variety of omics measurements and the advancement of computational approaches for the analysis and contextualization of the accumulated data. Encouragingly, while the metabolic network is highly interconnected and complex, it is at the same time probably the best characterized cellular network. Following, this review discusses the challenges that genome‐scale modeling of cancer metabolism has been facing. We survey several recent studies demonstrating the first strides that have been done, testifying to the value of this approach in portraying a network‐level view of the cancer metabolism and in identifying novel drug targets and biomarkers. Finally, we outline a few new steps that may further advance this field

    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

    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

    Metabolic profiling on 2D NMR TOCSY spectra using machine learning

    Get PDF
    Due to the dynamicity of biological cells, the role of metabolic profiling in discovering biological fingerprints of diseases, and their evolution, as well as the cellular pathway of different biological or chemical stimuli is most significant. Two-dimensional nuclear magnetic resonance (2D NMR) is one of the fundamental and strong analytical instruments for metabolic profiling. Though, total correlation spectroscopy (2D NMR 1H -1H TOCSY) can be used to improve spectral overlap of 1D NMR, strong peak shift, signal overlap, spectral crowding and matrix effects in complex biological mixtures are extremely challenging in 2D NMR analysis. In this work, we introduce an automated metabolic deconvolution and assignment based on the deconvolution of 2D TOCSY of real breast cancer tissue, in addition to different differentiation pathways of adipose tissue-derived human Mesenchymal Stem cells. A major alternative to the common approaches in NMR based machine learning where images of the spectra are used as an input, our metabolic assignment is based only on the vertical and horizontal frequencies of metabolites in the 1H-1H TOCSY. One- and multi-class Kernel null foley–Sammon transform, support vector machines, polynomial classifier kernel density estimation, and support vector data description classifiers were tested in semi-supervised learning and novelty detection settings. The classifiers’ performance was evaluated by comparing the conventional human-based methodology and automatic assignments under different initial training sizes settings. The results of our novel metabolic profiling methods demonstrate its suitability, robustness, and speed in automated nontargeted NMR metabolic analysis

    Metabolomics : a tool for studying plant biology

    Get PDF
    In recent years new technologies have allowed gene expression, protein and metabolite profiles in different tissues and developmental stages to be monitored. This is an emerging field in plant science and is applied to diverse plant systems in order to elucidate the regulation of growth and development. The goal in plant metabolomics is to analyze, identify and quantify all low molecular weight molecules of plant organisms. The plant metabolites are extracted and analyzed using various sensitive analytical techniques, usually mass spectrometry (MS) in combination with chromatography. In order to compare the metabolome of different plants in a high through-put manner, a number of biological, analytical and data processing steps have to be performed. In the work underlying this thesis we developed a fast and robust method for routine analysis of plant metabolite patterns using Gas Chromatography-Mass Spectrometry (GC/MS). The method was performed according to Design of Experiment (DOE) to investigate factors affecting the extraction and derivatization of the metabolites from leaves of the plant Arabidopsis thaliana. The outcome of metabolic analysis by GC/MS is a complex mixture of approximately 400 overlapping peaks. Resolving (deconvoluting) overlapping peaks is time-consuming, difficult to automate and additional processing is needed in order to compare samples. To avoid deconvolution being a major bottleneck in high through-put analyses we developed a new semi-automated strategy using hierarchical methods for processing GC/MS data that can be applied to all samples simultaneously. The two methods include base-line correction of the non-processed MS-data files, alignment, time-window determinations, Alternating Regression and multivariate analysis in order to detect metabolites that differ in relative concentrations between samples. The developed methodology was applied to study the effects of the plant hormone GA on the metabolome, with specific emphasis on auxin levels in Arabidopsis thaliana mutants defective in GA biosynthesis and signalling. A large series of plant samples was analysed and the resulting data were processed in less than one week with minimal labour; similar to the time required for the GC/MS analyses of the samples
    corecore