12 research outputs found

    Poly-omic statistical methods describe cyanobacterial metabolic adaptation to fluctuating environments

    Get PDF
    In this work, a genome-scale metabolic model of Synechococcus sp. PCC 7002 which utilizes flux balance analysis across multiple layers is analyzed to observe flux response between 23 growth conditions. This is achieved by setting reactions involved in biomass accumulation and energy production as objectives for bi-level linear optimization, thus serving to improve the characterization of mechanisms underlying these processes in photoautotrophic microalgae. Additionally, the incorporation of statistical techniques such as k-means clustering and principal component analysis (PCA) contribute to reducing dimensionality and inferring latent patterns

    Machine and deep learning meet genome-scale metabolic modeling

    Get PDF
    Omic data analysis is steadily growing as a driver of basic and applied molecular biology research. Core to the interpretation of complex and heterogeneous biological phenotypes are computational approaches in the fields of statistics and machine learning. In parallel, constraint-based metabolic modeling has established itself as the main tool to investigate large-scale relationships between genotype, phenotype, and environment. The development and application of these methodological frameworks have occurred independently for the most part, whereas the potential of their integration for biological, biomedical, and biotechnological research is less known. Here, we describe how machine learning and constraint-based modeling can be combined, reviewing recent works at the intersection of both domains and discussing the mathematical and practical aspects involved. We overlap systematic classifications from both frameworks, making them accessible to nonexperts. Finally, we delineate potential future scenarios, propose new joint theoretical frameworks, and suggest concrete points of investigation for this joint subfield. A multiview approach merging experimental and knowledge-driven omic data through machine learning methods can incorporate key mechanistic information in an otherwise biologically-agnostic learning process

    Combining metabolic modelling with machine learning accurately predicts yeast growth rate

    Get PDF
    New metabolic engineering techniques hold great potential for a range of bio-industrial applications. However, their practical use is hindered by the huge number of possible modifications, especially in eukaryotic organisms. To address this challenge, we present a methodology combining genome-scale metabolic modelling and machine learning to precisely predict cellular phenotypes starting from gene expression readouts. Our methodology enables the identification of candidate genetic manipulations that maximise a desired output--potentially reducing the number of in vitro experiments otherwise required. We apply and validate this methodology to a screen of 1,143 Saccharomyces cerevisiae knockout strains. Within the proposed framework, we compare different combinations of feature selection and supervised machine/deep learning approaches to identify the most effective model

    Seeing the wood for the trees: a forest of methods for optimization and omic-network integration in metabolic modelling.

    Get PDF
    Metabolic modelling has entered a mature phase with dozens of methods and software implementations available to the practitioner and the theoretician. It is not easy for a modeller to be able to see the wood (or the forest) for the trees. Driven by this analogy, we here present a 'forest' of principal methods used for constraint-based modelling in systems biology. This provides a tree-based view of methods available to prospective modellers, also available in interactive version at http://modellingmetabolism.net, where it will be kept updated with new methods after the publication of the present manuscript. Our updated classification of existing methods and tools highlights the most promising in the different branches, with the aim to develop a vision of how existing methods could hybridize and become more complex. We then provide the first hands-on tutorial for multi-objective optimization of metabolic models in R. We finally discuss the implementation of multi-view machine learning approaches in poly-omic integration. Throughout this work, we demonstrate the optimization of trade-offs between multiple metabolic objectives, with a focus on omic data integration through machine learning. We anticipate that the combination of a survey, a perspective on multi-view machine learning and a step-by-step R tutorial should be of interest for both the beginner and the advanced user.This work was partially funded by a Teesside University doctoral scholarship, EPSRC, and the EU grant MIMOMICS

    Social Dynamics Modeling of Chrono-nutrition

    Get PDF
    Gut microbiota and human relationships are strictly connected to each other. What we eat reflects our body-mind connection and synchronizes with people around us. However, how this impacts on gut microbiota and, conversely, how gut bacteria influence our dietary behaviors has not been explored yet. To quantify the complex dynamics of this interplay between gut and human behaviors we explore the ``gut-human behavior axis'' and its evolutionary dynamics in a real-world scenario represented by the social multiplex network. We consider a dual type of similarity, homophily and gut similarity, other than psychological and unconscious biases. We analyze the dynamics of social and gut microbial communities, quantifying the impact of human behaviors on diets and gut microbial composition and, backwards, through a control mechanism. Meal timing mechanisms and ``chrono-nutrition'' play a crucial role in feeding behaviors, along with the quality and quantity of food intake. Considering a population of shift workers, we explore the dynamic interplay between their eating behaviors and gut microbiota, modeling the social dynamics of chrono-nutrition in a multiplex network. Our findings allow us to quantify the relation between human behaviors and gut microbiota through the methodological introduction of gut metabolic modeling and statistical estimators, able to capture their dynamic interplay. Moreover, we find that the timing of gut microbial communities is slower than social interactions and shift-working, and the impact of shift-working on the dynamics of chrono-nutrition is a fluctuation of strategies with a major propensity for defection (e.g. high-fat meals). A deeper understanding of the relation between gut microbiota and the dietary behavioral patterns, by embedding also the related social aspects, allows improving the overall knowledge about metabolic models and their implications for human health, opening the possibility to design promising social therapeutic dietary interventions

    A mechanism-aware and multiomic machine-learning pipeline characterizes yeast cell growth

    Get PDF
    Metabolic modeling and machine learning are key components in the emerging next generation of systems and synthetic biology tools, targeting the genotype–phenotype–environment relationship. Rather than being used in isolation, it is becoming clear that their value is maximized when they are combined. However, the potential of integrating these two frameworks for omic data augmentation and integration is largely unexplored. We propose, rigorously assess, and compare machine-learning–based data integration techniques, combining gene expression profiles with computationally generated metabolic flux data to predict yeast cell growth. To this end, we create strain-specific metabolic models for 1,143 Saccharomyces cerevisiae mutants and we test 27 machine-learning methods, incorporating state-of-the-art feature selection and multiview learning approaches. We propose a multiview neural network using fluxomic and transcriptomic data, showing that the former increases the predictive accuracy of the latter and reveals functional patterns that are not directly deducible from gene expression alone. We test the proposed neural network on a further 86 strains generated in a different experiment, therefore verifying its robustness to an additional independent dataset. Finally, we show that introducing mechanistic flux features improves the predictions also for knockout strains whose genes were not modeled in the metabolic reconstruction. Our results thus demonstrate that fusing experimental cues with in silico models, based on known biochemistry, can contribute with disjoint information toward biologically informed and interpretable machine learning. Overall, this study provides tools for understanding and manipulating complex phenotypes, increasing both the prediction accuracy and the extent of discernible mechanistic biological insights

    Protocol for hybrid flux balance, statistical, and machine learning analysis of multi-omic data from the cyanobacterium Synechococcus sp. PCC 7002

    No full text
    Combining a computational framework for flux balance analysis with machine learning improves the accuracy of predicting metabolic activity across conditions, while enabling mechanistic interpretation. This protocol presents a guide to condition-specific metabolic modeling that integrates regularized flux balance analysis with machine learning approaches to extract key features from transcriptomic and fluxomic data. We demonstrate the protocol as applied to Synechococcus sp. PCC 7002; we also outline how it can be adapted to any species or community with available multi-omic data

    Optimization of multi-omic genome-scale models:Methodologies, hands-on tutorial, and perspectives

    Get PDF
    Genome-scale metabolic models are valuable tools for assessing the metabolic potential of living organisms. Being downstream of gene expression, metabolism is being increasingly used as an indicator of the phenotypic outcome for drugs and therapies. We here present a review of the principal methods used for constraint-based modelling in systems biology, and explore how the integration of multi-omic data can be used to improve phenotypic predictions of genome-scale metabolic models. We believe that the large-scale comparison of the metabolic response of an organism to different environmental conditions will be an important challenge for genome-scale models. Therefore, within the context of multi-omic methods, we describe a tutorial for multi-objective optimisation using the metabolic and transcriptomics adaptation estimator (METRADE), implemented in MATLAB. METRADE uses microarray and codon usage data to model bacterial metabolic response to environmental conditions (e.g. antibiotics, temperatures, heat shock). Finally, we discuss key considerations for the integration of multi-omic networks into metabolic models, towards automatically extracting knowledge from such models
    corecore