19 research outputs found

    Predicting growth of the healthy infant using a genome scale metabolic model

    Get PDF
    An estimated 165 million children globally have stunted growth, and extensive growth data are available. Genome scale metabolic models allow the simulation of molecular flux over each metabolic enzyme, and are well adapted to analyze biological systems. We used a human genome scale metabolic model to simulate the mechanisms of growth and integrate data about breast-milk intake and composition with the infant\u27s biomass and energy expenditure of major organs. The model predicted daily metabolic fluxes from birth to age 6 months, and accurately reproduced standard growth curves and changes in body composition. The model corroborates the finding that essential amino and fatty acids do not limit growth, but that energy is the main growth limiting factor. Disruptions to the supply and demand of energy markedly affected the predicted growth, indicating that elevated energy expenditure may be detrimental. The model was used to simulate the metabolic effect of mineral deficiencies, and showed the greatest growth reduction for deficiencies in copper, iron, and magnesium ions which affect energy production through oxidative phosphorylation. The model and simulation method were integrated to a platform and shared with the research community. The growth model constitutes another step towards the complete representation of human metabolism, and may further help improve the understanding of the mechanisms underlying stunting

    Constraint-based modeling of metabolism - interpreting predictions of growth and ATP synthesis in human and yeast

    Get PDF
    Growth is the primary objective of the cell. Diseases arise when cells diverge from a healthy growth-pattern. An increased understanding of cellular growth may thus be translated into improved human health. The cell requires materials and free energy (in the form of ATP) in order to grow, metabolism supplies the cell with this. The rate of metabolism is ultimately constrained by the biophysical properties of the metabolic enzymes. Interactions between the constraints and the growth-objective gives rise to metabolic trade-offs, e.g. between ATP synthesis from respiration and fermentation. We can gain quantitative insight into these processes by simulating metabolism using mathematical models. In this thesis I simulated the metabolism of four biological systems: the infant, cancer, yeast and muscle. The simulations demonstrated how a shift in metabolic strategy may increase the rates of ATP synthesis and growth. These increased metabolic rates come at the expense of decreased resource efficiency, i.e. ATP produced per carbon consumed. The effect was primarily caused by the low catalytic efficiency of the respiratory enzyme complexes I and V. By shifting from respiratory to fermentative ATP synthesis, the cell was able to bypass these constraints. An intermediate strategy involved bypassing only complex I. The phenomenon was experimentally corroborated in the working muscle, and it is the native state of the yeast Saccharomyces cerevisiae (which lacks complex I). The differences in efficiency between the different metabolic pathways also explained why cells grow faster on some carbon sources, e.g. the specific growth rate for yeast is higher on glucose than on ethanol. These models were extended to predict the world-record running-speeds at different distances, by taking the sizes of the body’s nutrient-deposits into account. A metabolic strategy employed by cancer cells involved excretion of the amino acid glutamate. The simulations showed a mechanistic relation to catabolism of branched-chain amino acids and the localization of amino acid metabolism to different cellular compartments. By experimentally inhibiting glutamate excretion using an off-the-shelf drug (sulfasalazine), the growth rate of a cancer cell line was reduced. The metabolic modeling involved integration of various types of data and thus demonstrated the potential to unify knowledge from different studies and domains. This exposed contradictory claims in literature and highlighted knowledge-gaps that need to be filled to further improve human health

    Artificial neural networks enable genome-scale simulations of intracellular signaling

    Get PDF
    Mammalian cells adapt their functional state in response to external signals in form of ligands that bind receptors on the cell-surface. Mechanistically, this involves signal-processing through a complex network of molecular interactions that govern transcription factor activity patterns. Computer simulations of the information flow through this network could help predict cellular responses in health and disease. Here we develop a recurrent neural network framework constrained by prior knowledge of the signaling network with ligand-concentrations as input and transcription factor-activity as output. Applied to synthetic data, it predicts unseen test-data (Pearson correlation r = 0.98) and the effects of gene knockouts (r = 0.8). We stimulate macrophages with 59 different ligands, with and without the addition of lipopolysaccharide, and collect transcriptomics data. The framework predicts this data under cross-validation (r = 0.8) and knockout simulations suggest a role for RIPK1 in modulating the lipopolysaccharide response. This work demonstrates the feasibility of genome-scale simulations of intracellular signaling. Many diseases are caused by disruptions to the network of biochemical reactions that allow cells to respond to external signals. Here Nilsson et al develop a method to simulate cellular signaling using artificial neural networks to predict cellular responses and activities of signaling molecules

    BioMet Toolbox 2.0: genome-wide analysis of metabolism and omics data

    Get PDF
    Analysis of large data sets using computational and mathematical tools have become a central part of biological sciences. Large amounts of data are being generated each year from different biological research fields leading to a constant development of software and algorithms aimed to deal with the increasing creation of information. The BioMet Toolbox 2.0 integrates a number of functionalities in a user-friendly environment enabling the user to work with biological data in a web interface. The unique and distinguishing feature of the BioMet Toolbox 2.0 is to provide a web user interface to tools for metabolic pathways and omics analysis developed under different platform-dependent environments enabling easy access to these computational tools

    Improving the phenotype predictions of a yeast genome-scale metabolic model by incorporating enzymatic constraints

    Get PDF
    Genome-scale metabolic models (GEMs) are widely used to calculate metabolic phenotypes. They rely on defining a set of constraints, the most common of which is that the production of metabolites and/or growth are limited by the carbon source uptake rate. However, enzyme abundances and kinetics, which act as limitations on metabolic fluxes, are not taken into account. Here, we present GECKO, a method that enhances a GEM to account for enzymes as part of reactions, thereby ensuring that each metabolic flux does not exceed its maximum capacity, equal to the product of the enzyme's abundance and turnover number. We applied GECKO to a Saccharomyces cerevisiae GEM and demonstrated that the new model could correctly describe phenotypes that the previous model could not, particularly under high enzymatic pressure conditions, such as yeast growing on different carbon sources in excess, coping with stress, or overexpressing a specific pathway. GECKO also allows to directly integrate quantitative proteomics data; by doing so, we significantly reduced flux variability of the model, in over 60% of metabolic reactions. Additionally, the model gives insight into the distribution of enzyme usage between and within metabolic pathways. The developed method and model are expected to increase the use of model-based design in metabolic engineering

    Quantitative analysis of amino acid metabolism in liver cancer links glutamate excretion to nucleotide synthesis

    Get PDF
    Many cancer cells consume glutamine at high rates; counterintuitively, they simultaneously excrete glutamate, the first intermediate in glutamine metabolism. Glutamine consumption has been linked to replenishment of tricarboxylic acid cycle (TCA) intermediates and synthesis of adenosine triphosphate (ATP), but the reason for glutamate excretion is unclear. Here, we dynamically profile the uptake and excretion fluxes of a liver cancer cell line (HepG2) and use genome-scale metabolic modeling for in-depth analysis. We find that up to 30% of the glutamine is metabolized in the cytosol, primarily for nucleotide synthesis, producing cytosolic glutamate. We hypothesize that excreting glutamate helps the cell to increase the nucleotide synthesis rate to sustain growth. Indeed, we show experimentally that partial inhibition of glutamate excretion reduces cell growth. Our integrative approach thus links glutamine addiction to glutamate excretion in cancer and points toward potential drug targets

    Recon3D enables a three-dimensional view of gene variation in human metabolism

    Get PDF
    Genome-scale network reconstructions have helped uncover the molecular basis of metabolism. Here we present Recon3D, a computational resource that includes three-dimensional (3D) metabolite and protein structure data and enables integrated analyses of metabolic functions in humans. We use Recon3D to functionally characterize mutations associated with disease, and identify metabolic response signatures that are caused by exposure to certain drugs. Recon3D represents the most comprehensive human metabolic network model to date, accounting for 3,288 open reading frames (representing 17% of functionally annotated human genes), 13,543 metabolic reactions involving 4,140 unique metabolites, and 12,890 protein structures. These data provide a unique resource for investigating molecular mechanisms of human metabolism. Recon3D is available at http://vmh.life

    Genome scale metabolic modeling of cancer

    No full text
    Cancer cells reprogram metabolism to support rapid proliferation and survival. Energy metabolism is particularly important for growth and genes encoding enzymes involved in energy metabolism are frequently altered in cancer cells. A genome scale metabolic model (GEM) is a mathematical formalization of metabolism which allows simulation and hypotheses testing of metabolic strategies. It has successfully been applied to many microorganisms and is now used to study cancer metabolism. Generic models of human metabolism have been reconstructed based on the existence of metabolic genes in the human genome. Cancer specific models of metabolism have also been generated by reducing the number of reactions in the generic model based on high throughput expression data, e.g. transcriptomics and proteomics. Targets for drugs and bio markers for diagnostics have been identified using these models. They have also been used as scaffolds for analysis of high throughput data to allow mechanistic interpretation of changes in expression. Finally, GEMs allow quantitative flux predictions using flux balance analysis (FBA). Here we critically review the requirements for successful FBA simulations of cancer cells and discuss the symmetry between the methods used for modeling of microbial and cancer metabolism. GEMs have great potential for translational research on cancer and will therefore become of increasing importance in the future. \ua9 201
    corecore