Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31730853

RESUMO

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Assuntos
Genoma de Protozoário , Estágios do Ciclo de Vida/genética , Fígado/metabolismo , Fígado/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/genética , Alelos , Amino Açúcares/biossíntese , Animais , Culicidae/parasitologia , Eritrócitos/parasitologia , Ácido Graxo Sintases/metabolismo , Ácidos Graxos/metabolismo , Técnicas de Inativação de Genes , Genótipo , Modelos Biológicos , Mutação/genética , Parasitos/genética , Parasitos/crescimento & desenvolvimento , Fenótipo , Plasmodium berghei/metabolismo , Ploidias , Reprodução
2.
Proc Natl Acad Sci U S A ; 119(46): e2211197119, 2022 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-36343249

RESUMO

Advances in medicine and biotechnology rely on a deep understanding of biological processes. Despite the increasingly available types and amounts of omics data, significant knowledge gaps remain, with current approaches to identify and curate missing annotations being limited to a set of already known reactions. Here, we introduce Network Integrated Computational Explorer for Gap Annotation of Metabolism (NICEgame), a workflow to identify and curate nonannotated metabolic functions in genomes using the ATLAS of Biochemistry and genome-scale metabolic models (GEMs). To resolve gaps in GEMs, NICEgame provides alternative sets of known and hypothetical reactions, assesses their thermodynamic feasibility, and suggests candidate genes to catalyze these reactions. We identified metabolic gaps and applied NICEgame in the latest GEM of Escherichia coli, iML1515, and enhanced the E. coli genome annotation by resolving 47% of these gaps. NICEgame, applicable to any GEM and functioning from open-source software, should thus enhance all GEM-based predictions and subsequent biotechnological and biomedical applications.


Assuntos
Escherichia coli , Redes e Vias Metabólicas , Escherichia coli/genética , Escherichia coli/metabolismo , Fluxo de Trabalho , Software , Genoma , Modelos Biológicos
3.
Bioinformatics ; 39(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36495209

RESUMO

MOTIVATION: Large-scale kinetic models are an invaluable tool to understand the dynamic and adaptive responses of biological systems. The development and application of these models have been limited by the availability of computational tools to build and analyze large-scale models efficiently. The toolbox presented here provides the means to implement, parameterize and analyze large-scale kinetic models intuitively and efficiently. RESULTS: We present a Python package (SKiMpy) bridging this gap by implementing an efficient kinetic modeling toolbox for the semiautomatic generation and analysis of large-scale kinetic models for various biological domains such as signaling, gene expression and metabolism. Furthermore, we demonstrate how this toolbox is used to parameterize kinetic models around a steady-state reference efficiently. Finally, we show how SKiMpy can implement multispecies bioreactor simulations to assess biotechnological processes. AVAILABILITY AND IMPLEMENTATION: The software is available as a Python 3 package on GitHub: https://github.com/EPFL-LCSB/SKiMpy, along with adequate documentation. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Modelos Biológicos , Software , Cinética , Documentação
4.
Metab Eng ; 84: 109-116, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38880390

RESUMO

The production of recombinant proteins in a host using synthetic constructs such as plasmids comes at the cost of detrimental effects such as reduced growth, energetic inefficiencies, and other stress responses, collectively known as metabolic burden. Increasing the number of copies of the foreign gene increases the metabolic load but increases the expression of the foreign protein. Thus, there is a trade-off between biomass and product yield in response to changes in heterologous gene copy number. This work proposes a computational method, rETFL (recombinant Expression and Thermodynamic Flux), for analyzing and predicting the responses of recombinant organisms to the introduction of synthetic constructs. rETFL is an extension to the ETFL formulations designed to reconstruct models of metabolism and expression (ME-models). We have illustrated the capabilities of the method in four studies to (i) capture the growth reduction in plasmid-containing E. coli and recombinant protein production; (ii) explore the trade-off between biomass and product yield as plasmid copy number is varied; (iii) predict the emergence of overflow metabolism in recombinant E. coli in agreement with experimental data; and (iv) investigate the individual pathways and enzymes affected by the presence of the plasmid. We anticipate that rETFL will serve as a comprehensive platform for integrating available omics data for recombinant organisms and making context-specific predictions that can help optimize recombinant expression systems for biopharmaceutical production and gene therapy.

5.
Proc Natl Acad Sci U S A ; 118(8)2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33602812

RESUMO

Diauxie, or the sequential consumption of carbohydrates in bacteria such as Escherichia coli, has been hypothesized to be an evolutionary strategy which allows the organism to maximize its instantaneous specific growth-giving the bacterium a competitive advantage. Currently, the computational techniques used in industrial biotechnology fall short of explaining the intracellular dynamics underlying diauxic behavior. In particular, the understanding of the proteome dynamics in diauxie can be improved. We developed a robust iterative dynamic method based on expression- and thermodynamically enabled flux models to simulate the kinetic evolution of carbohydrate consumption and cellular growth. With minimal modeling assumptions, we couple kinetic uptakes, gene expression, and metabolic networks, at the genome scale, to produce dynamic simulations of cell cultures. The method successfully predicts the preferential uptake of glucose over lactose in E. coli cultures grown on a mixture of carbohydrates, a manifestation of diauxie. The simulated cellular states also show the reprogramming in the content of the proteome in response to fluctuations in the availability of carbon sources, and it captures the associated time lag during the diauxie phenotype. Our models suggest that the diauxic behavior of cells is the result of the evolutionary objective of maximization of the specific growth of the cell. We propose that genetic regulatory networks, such as the lac operon in E. coli, are the biological implementation of a robust control system to ensure optimal growth.


Assuntos
Escherichia coli/crescimento & desenvolvimento , Escherichia coli/metabolismo , Redes e Vias Metabólicas , Modelos Biológicos , Acetatos/metabolismo , Enzimas/metabolismo , Escherichia coli/citologia , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Genoma Bacteriano , Glucose/metabolismo , Cinética , Lactose/metabolismo , Termodinâmica
6.
Bioinformatics ; 37(20): 3560-3568, 2021 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-34003971

RESUMO

MOTIVATION: Finding biosynthetic pathways is essential for metabolic engineering of organisms to produce chemicals, biodegradation prediction of pollutants and drugs, and for the elucidation of bioproduction pathways of secondary metabolites. A key step in biosynthetic pathway design is the extraction of novel metabolic pathways from big networks that integrate known biological, as well as novel, predicted biotransformations. However, the efficient analysis and the navigation of big biochemical networks remain a challenge. RESULTS: Here, we propose the construction of searchable graph representations of metabolic networks. Each reaction is decomposed into pairs of reactants and products, and each pair is assigned a weight, which is calculated from the number of conserved atoms between the reactant and the product molecule. We test our method on a biochemical network that spans 6546 known enzymatic reactions to show how our approach elegantly extracts biologically relevant metabolic pathways from biochemical networks, and how the proposed network structure enables the application of efficient graph search algorithms that improve navigation and pathway identification in big metabolic networks. The weighted reactant-product pairs of an example network and the corresponding graph search algorithm are available online. The proposed method extracts metabolic pathways fast and reliably from big biochemical networks, which is inherently important for all applications involving the engineering of metabolic networks. AVAILABILITY AND IMPLEMENTATION: https://github.com/EPFL-LCSB/nicepath. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

7.
Metab Eng ; 72: 259-274, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35381376

RESUMO

Synthetic biology and metabolic engineering rely on computational search tools for predictions of novel biosynthetic pathways to industrially important compounds, many of which are derived from aromatic amino acids. Pathway search tools vary in their scope of covered reactions and compounds, as well as in metrics for ranking and evaluation. In this work, we present a new computational resource called ARBRE: Aromatic compounds RetroBiosynthesis Repository and Explorer. It consists of a comprehensive biochemical reaction network centered around aromatic amino acid biosynthesis and a computational toolbox for navigating this network. ARBRE encompasses over 33'000 known and 390'000 novel reactions predicted with generalized enzymatic reactions rules and over 74'000 compounds, of which 19'000 are known to biochemical databases and 55'000 only to PubChem. Over 1'000 molecules that were solely part of the PubChem database before and were previously impossible to integrate into a biochemical network are included into the ARBRE reaction network by assigning enzymatic reactions. ARBRE can be applied for pathway search, enzyme annotation, pathway ranking, visualization, and network expansion around known biochemical pathways and products of lignin degradation to predict valuable compound derivations. In line with the standards of open science, we have made the toolbox freely available to the scientific community on git (https://github.com/EPFL-LCSB/ARBRE) and we provide the web-version at http://lcsb-databases.epfl.ch/arbre/. We envision that ARBRE will provide the community with a new computational resource and comprehensive search tool to predict and rank pathways towards industrially important aromatic compounds.


Assuntos
Engenharia Metabólica , Redes e Vias Metabólicas , Aminoácidos Aromáticos/genética , Vias Biossintéticas , Biologia Sintética
8.
PLoS Comput Biol ; 17(7): e1009140, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34292935

RESUMO

The metabolic capabilities of the species and the local environment shape the microbial interactions in a community either through the exchange of metabolic products or the competition for the resources. Cells are often arranged in close proximity to each other, creating a crowded environment that unevenly reduce the diffusion of nutrients. Herein, we investigated how the crowding conditions and metabolic variability among cells shape the dynamics of microbial communities. For this, we developed CROMICS, a spatio-temporal framework that combines techniques such as individual-based modeling, scaled particle theory, and thermodynamic flux analysis to explicitly incorporate the cell metabolism and the impact of the presence of macromolecular components on the nutrients diffusion. This framework was used to study two archetypical microbial communities (i) Escherichia coli and Salmonella enterica that cooperate with each other by exchanging metabolites, and (ii) two E. coli with different production level of extracellular polymeric substances (EPS) that compete for the same nutrients. In the mutualistic community, our results demonstrate that crowding enhanced the fitness of cooperative mutants by reducing the leakage of metabolites from the region where they are produced, avoiding the resource competition with non-cooperative cells. Moreover, we also show that E. coli EPS-secreting mutants won the competition against the non-secreting cells by creating less dense structures (i.e. increasing the spacing among the cells) that allow mutants to expand and reach regions closer to the nutrient supply point. A modest enhancement of the relative fitness of EPS-secreting cells over the non-secreting ones were found when the crowding effect was taken into account in the simulations. The emergence of cell-cell interactions and the intracellular conflicts arising from the trade-off between growth and the secretion of metabolites or EPS could provide a local competitive advantage to one species, either by supplying more cross-feeding metabolites or by creating a less dense neighborhood.


Assuntos
Biologia Computacional/métodos , Interações Microbianas/fisiologia , Microbiota/fisiologia , Modelos Biológicos , Escherichia coli/metabolismo , Escherichia coli/fisiologia , Salmonella enterica/metabolismo , Salmonella enterica/fisiologia , Análise Espaço-Temporal
9.
PLoS Comput Biol ; 17(7): e1009158, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34292941

RESUMO

Microorganisms are frequently organized into crowded structures that affect the nutrients diffusion. This reduction in metabolite diffusion could modify the microbial dynamics, meaning that computational methods for studying microbial systems need accurate ways to model the crowding conditions. We previously developed a computational framework, termed CROMICS, that incorporates the effect of the (time-dependent) crowding conditions on the spatio-temporal modeling of microbial communities, and we used it to demonstrate the crowding influence on the community dynamics. To further identify scenarios where crowding should be considered in microbial modeling, we herein applied and extended CROMICS to simulate several environmental conditions that could potentially boost or dampen the crowding influence in biofilms. We explore whether the nutrient supply (rich- or low-nutrient media), the cell-packing configuration (square or hexagonal spherical cell arrangement), or the cell growing conditions (planktonic state or biofilm) modify the crowding influence on the growth of Escherichia coli. Our results indicate that the growth rate, the abundance and appearance time of different cell phenotypes as well as the amount of by-products secreted to the medium are sensitive to some extent to the local crowding conditions in all scenarios tested, except in rich-nutrient media. Crowding conditions enhance the formation of nutrient gradient in biofilms, but its effect is only appreciated when cell metabolism is controlled by the nutrient limitation. Thus, as soon as biomass (and/or any other extracellular macromolecule) accumulates in a region, and cells occupy more than 14% of the volume fraction, the crowding effect must not be underestimated, as the microbial dynamics start to deviate from the ideal/expected behaviour that assumes volumeless cells or when a homogeneous (reduced) diffusion is applied in the simulation. The modeling and simulation of the interplay between the species diversity (cell shape and metabolism) and the environmental conditions (nutrient quality, crowding conditions) can help to design effective strategies for the optimization and control of microbial systems.


Assuntos
Biofilmes , Biologia Computacional/métodos , Interações Microbianas/fisiologia , Microbiota/fisiologia , Modelos Biológicos , Escherichia coli/fisiologia
10.
Proc Natl Acad Sci U S A ; 116(15): 7298-7307, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30910961

RESUMO

Thousands of biochemical reactions with characterized activities are "orphan," meaning they cannot be assigned to a specific enzyme, leaving gaps in metabolic pathways. Novel reactions predicted by pathway-generation tools also lack associated sequences, limiting protein engineering applications. Associating orphan and novel reactions with known biochemistry and suggesting enzymes to catalyze them is a daunting problem. We propose the method BridgIT to identify candidate genes and catalyzing proteins for these reactions. This method introduces information about the enzyme binding pocket into reaction-similarity comparisons. BridgIT assesses the similarity of two reactions, one orphan and one well-characterized nonorphan reaction, using their substrate reactive sites, their surrounding structures, and the structures of the generated products to suggest enzymes that catalyze the most-similar nonorphan reactions as candidates for also catalyzing the orphan ones. We performed two large-scale validation studies to test BridgIT predictions against experimental biochemical evidence. For the 234 orphan reactions from the Kyoto Encyclopedia of Genes and Genomes (KEGG) 2011 (a comprehensive enzymatic-reaction database) that became nonorphan in KEGG 2018, BridgIT predicted the exact or a highly related enzyme for 211 of them. Moreover, for 334 of 379 novel reactions in 2014 that were later cataloged in KEGG 2018, BridgIT predicted the exact or highly similar enzymes. BridgIT requires knowledge about only four connecting bonds around the atoms of the reactive sites to correctly annotate proteins for 93% of analyzed enzymatic reactions. Increasing to seven connecting bonds allowed for the accurate identification of a sequence for nearly all known enzymatic reactions.


Assuntos
Bases de Dados de Proteínas , Enzimas , Anotação de Sequência Molecular , Análise de Sequência de Proteína , Sítios de Ligação , Enzimas/química , Enzimas/genética
11.
BMC Bioinformatics ; 22(1): 134, 2021 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-33743594

RESUMO

BACKGROUND: Significant efforts have been made in building large-scale kinetic models of cellular metabolism in the past two decades. However, most kinetic models published to date, remain focused around central carbon pathways or are built around ad hoc reduced models without clear justification on their derivation and usage. Systematic algorithms exist for reducing genome-scale metabolic reconstructions to build thermodynamically feasible and consistently reduced stoichiometric models. However, it is important to study how network complexity affects conclusions derived from large-scale kinetic models built around consistently reduced models before we can apply them to study biological systems. RESULTS: We reduced the iJO1366 Escherichia Coli genome-scale metabolic reconstruction systematically to build three stoichiometric models of different size. Since the reduced models are expansions around the core subsystems for which the reduction was performed, the models are nested. We present a method for scaling up the flux profile and the concentration vector reference steady-states from the smallest model to the larger ones, whilst preserving maximum equivalency. Populations of kinetic models, preserving similarity in kinetic parameters, were built around the reference steady-states and their metabolic sensitivity coefficients (MSCs) were computed. The MSCs were sensitive to the model complexity. We proposed a metric for measuring the sensitivity of MSCs to these structural changes. CONCLUSIONS: We proposed for the first time a workflow for scaling up the size of kinetic models while preserving equivalency between the kinetic models. Using this workflow, we demonstrate that model complexity in terms of networks size has significant impact on sensitivity characteristics of kinetic models. Therefore, it is essential to account for the effects of network complexity when constructing kinetic models. The presented metric for measuring MSC sensitivity to structural changes can guide modelers and experimentalists in improving model quality and guide synthetic biology and metabolic engineering. Our proposed workflow enables the testing of the suitability of a kinetic model for answering certain study-specific questions. We argue that the model-based metabolic design targets that are common across models of different size are of higher confidence, while those that are different could be the objective of investigations for model improvement.


Assuntos
Escherichia coli , Engenharia Metabólica , Modelos Biológicos , Algoritmos , Escherichia coli/genética , Cinética , Redes e Vias Metabólicas
12.
Metab Eng ; 66: 191-203, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33895366

RESUMO

The advancements in genome editing techniques over the past years have rekindled interest in rational metabolic engineering strategies. While Metabolic Control Analysis (MCA) is a well-established method for quantifying the effects of metabolic engineering interventions on flows in metabolic networks and metabolite concentrations, it does not consider the physiological limitations of the cellular environment and metabolic engineering design constraints. We report here a constraint-based framework, Network Response Analysis (NRA), for rational genetic strain design. NRA is cast as a Mixed-Integer Linear Programming problem that integrates MCA, Thermodynamically-based Flux Analysis (TFA), biologically relevant constraints, as well as genome editing restrictions into a comprehensive platform for identifying metabolic engineering targets. We show that the NRA formulation and its core constraints are equivalent to the ones of Flux Balance Analysis (FBA) and TFA, which allows it to be used for a wide range of optimization criteria and with various physiological constraints. We also show how the parametrization and introduction of biological constraints enhance the NRA formulation compared to the classical MCA approach, and we demonstrate its features and its ability to generate multiple alternative optimal strategies given several user-defined boundaries and objectives. In summary, NRA is a sophisticated alternative to classical MCA for rational metabolic engineering that accommodates the incorporation of physiological data at metabolic flux, metabolite concentration, and enzyme expression levels.


Assuntos
Escherichia coli , Modelos Biológicos , Escherichia coli/genética , Engenharia Metabólica , Análise do Fluxo Metabólico , Redes e Vias Metabólicas/genética
13.
Bioinformatics ; 35(1): 167-169, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30561545

RESUMO

Summary: pyTFA and matTFA are the first published implementations of the original TFA paper. Specifically, they include explicit formulation of Gibbs energies and metabolite concentrations, which enables straightforward integration of metabolite concentration measurements. Motivation: High-throughput analytic technologies provide a wealth of omics data that can be used to perform thorough analyses for a multitude of studies in the areas of Systems Biology and Biotechnology. Nevertheless, most studies are still limited to constraint-based Flux Balance Analyses (FBA), neglecting an important physicochemical constraint: thermodynamics. Thermodynamics-based Flux Analysis (TFA) in metabolic models enables the integration of quantitative metabolomics data to study their effects on the net-flux directionality of reactions in the network. In addition, it allows us to estimate how far each reaction operates from thermodynamic equilibrium, which provides critical information for guiding metabolic engineering decisions. Results: We present a Python package (pyTFA) and a Matlab toolbox (matTFA) that implement TFA. We show an example of application on both a reduced and a genome-scale model of E. coli., and demonstrate TFA and data integration through TFA reduce the feasible flux space with respect to FBA. Availability and implementation: Documented implementation of TFA framework both in Python (pyTFA) and Matlab (matTFA) are available on www.github.com/EPFL-LCSB/. Supplementary information: Supplementary data are available at Bioinformatics online.


Assuntos
Escherichia coli , Metabolômica , Modelos Biológicos , Software , Biologia Computacional , Redes e Vias Metabólicas , Biologia de Sistemas , Termodinâmica
14.
PLoS Comput Biol ; 15(4): e1006760, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31002661

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is associated with metabolic syndromes spanning a wide spectrum of diseases, from simple steatosis to the more complex nonalcoholic steatohepatitis. To identify the deregulation that occurs in metabolic processes at the molecular level that give rise to these various NAFLD phenotypes, algorithms such as pathway enrichment analysis (PEA) can be used. These analyses require the use of predefined pathway maps, which are composed of reactions describing metabolic processes/subsystems. Unfortunately, the annotation of the metabolic subsystems can differ depending on the pathway database used, making these approaches subject to biases associated with different pathway annotations, and these methods cannot capture the balancing of cofactors and byproducts through the complex nature and interactions of genome-scale metabolic networks (GEMs). Here, we introduce a framework entitled Minimum Network Enrichment Analysis (MiNEA) that is applied to GEMs to generate all possible alternative minimal networks (MiNs), which are possible and feasible networks composed of all the reactions pertaining to various metabolic subsystems that can synthesize a target metabolite. We applied MiNEA to investigate deregulated MiNs and to identify key regulators in different NAFLD phenotypes, such as a fatty liver and liver inflammation, in both humans and mice by integrating condition-specific transcriptomics data from liver samples. We identified key deregulations in the synthesis of cholesteryl esters, cholesterol, and hexadecanoate in both humans and mice, and we found that key regulators of the hydrogen peroxide synthesis network were regulated differently in humans and mice. We further identified which MiNs demonstrate the general and specific characteristics of the different NAFLD phenotypes. MiNEA is applicable to any GEM and to any desired target metabolite, making MiNEA flexible enough to study condition-specific metabolism for any given disease or organism.


Assuntos
Biologia Computacional/métodos , Redes e Vias Metabólicas , Hepatopatia Gordurosa não Alcoólica , Animais , Ceramidas/metabolismo , Bases de Dados Factuais , Perfilação da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Metabolismo dos Lipídeos , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/fisiologia , Camundongos , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/fisiopatologia , Transcriptoma/genética , Transcriptoma/fisiologia
15.
PLoS Comput Biol ; 15(12): e1007536, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31815929

RESUMO

Kinetic models of metabolism can be constructed to predict cellular regulation and devise metabolic engineering strategies, and various promising computational workflows have been developed in recent years for this. Due to the uncertainty in the kinetic parameter values required to build kinetic models, these workflows rely on ensemble modeling (EM) principles for sampling and building populations of models describing observed physiologies. Sensitivity coefficients from metabolic control analysis (MCA) of kinetic models can provide important insight about cellular control around a given physiological steady state. However, despite considering populations of kinetic models and their model outputs, current approaches do not provide adequate tools for statistical inference. To derive conclusions from model outputs, such as MCA sensitivity coefficients, it is necessary to rank/compare populations of variables with each other. Currently existing workflows consider confidence intervals (CIs) that are derived independently for each comparable variable. Hence, it is important to derive simultaneous CIs for the variables that we wish to rank/compare. Herein, we used an existing large-scale kinetic model of Escherichia Coli metabolism to present how univariate CIs can lead to incorrect conclusions, and we present a new workflow that applies three different multivariate statistical approaches. We use the Bonferroni and the exact normal methods to build symmetric CIs using the normality assumptions. We then suggest how bootstrapping can compute asymmetric CIs whilst relaxing this normality assumption. We conclude that the Bonferroni and the exact normal methods can provide simple and efficient ways for constructing reliable CIs, with the exact normal method favored over the Bonferroni when the compared variables present dependencies. Bootstrapping, despite its significantly higher computational cost, is recommended when comparing non-normal distributions of variables. Additionally, we show how the Bonferroni method can readily be used to estimate required sample numbers to attain a certain CI size.


Assuntos
Escherichia coli/metabolismo , Modelos Biológicos , Algoritmos , Células/metabolismo , Biologia Computacional , Simulação por Computador , Intervalos de Confiança , Cinética , Metabolismo , Biologia de Sistemas , Incerteza
16.
PLoS Comput Biol ; 15(5): e1007036, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31083653

RESUMO

The ever-increasing availability of transcriptomic and metabolomic data can be used to deeply analyze and make ever-expanding predictions about biological processes, as changes in the reaction fluxes through genome-wide pathways can now be tracked. Currently, constraint-based metabolic modeling approaches, such as flux balance analysis (FBA), can quantify metabolic fluxes and make steady-state flux predictions on a genome-wide scale using optimization principles. However, relating the differential gene expression or differential metabolite abundances in different physiological states to the differential flux profiles remains a challenge. Here we present a novel method, named REMI (Relative Expression and Metabolomic Integrations), that employs genome-scale metabolic models (GEMs) to translate differential gene expression and metabolite abundance data obtained through genetic or environmental perturbations into differential fluxes to analyze the altered physiology for any given pair of conditions. REMI allows for gene-expression, metabolite abundance, and thermodynamic data to be integrated into a single framework, then uses optimization principles to maximize the consistency between the differential gene-expression levels and metabolite abundance data and the estimated differential fluxes and thermodynamic constraints. We applied REMI to integrate into the Escherichia coli GEM publicly available sets of expression and metabolomic data obtained from two independent studies and under wide-ranging conditions. The differential flux distributions obtained from REMI corresponding to the various perturbations better agreed with the measured fluxomic data, and thus better reflected the different physiological states, than a traditional model. Compared to the similar alternative method that provides one solution from the solution space, REMI was able to enumerate several alternative flux profiles using a mixed-integer linear programming approach. Using this important advantage, we performed a high-frequency analysis of common genes and their associated reactions in the obtained alternative solutions and identified the most commonly regulated genes across any two given conditions. We illustrate that this new implementation provides more robust and biologically relevant results for a better understanding of the system physiology.


Assuntos
Redes e Vias Metabólicas , Modelos Biológicos , Biologia Computacional , Bases de Dados Factuais , Bases de Dados Genéticas , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Genoma Bacteriano , Cinética , Metabolômica , Biologia de Sistemas , Termodinâmica
17.
PLoS Comput Biol ; 15(8): e1007242, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31430276

RESUMO

A persistent obstacle for constructing kinetic models of metabolism is uncertainty in the kinetic properties of enzymes. Currently, available methods for building kinetic models can cope indirectly with uncertainties by integrating data from different biological levels and origins into models. In this study, we use the recently proposed computational approach iSCHRUNK (in Silico Approach to Characterization and Reduction of Uncertainty in the Kinetic Models), which combines Monte Carlo parameter sampling methods and machine learning techniques, in the context of Bayesian inference. Monte Carlo parameter sampling methods allow us to exploit synergies between different data sources and generate a population of kinetic models that are consistent with the available data and physicochemical laws. The machine learning allows us to data-mine the a priori generated kinetic parameters together with the integrated datasets and derive posterior distributions of kinetic parameters consistent with the observed physiology. In this work, we used iSCHRUNK to address a design question: can we identify which are the kinetic parameters and what are their values that give rise to a desired metabolic behavior? Such information is important for a wide variety of studies ranging from biotechnology to medicine. To illustrate the proposed methodology, we performed Metabolic Control Analysis, computed the flux control coefficients of the xylose uptake (XTR), and identified parameters that ensure a rate improvement of XTR in a glucose-xylose co-utilizing S. cerevisiae strain. Our results indicate that only three kinetic parameters need to be accurately characterized to describe the studied physiology, and ultimately to design and control the desired responses of the metabolism. This framework paves the way for a new generation of methods that will systematically integrate the wealth of available omics data and efficiently extract the information necessary for metabolic engineering and synthetic biology decisions.


Assuntos
Modelos Biológicos , Algoritmos , Teorema de Bayes , Fenômenos Bioquímicos , Biologia Computacional , Hexoquinase/metabolismo , Cinética , Aprendizado de Máquina , Engenharia Metabólica , Redes e Vias Metabólicas , Método de Monte Carlo , Saccharomyces cerevisiae/metabolismo , Incerteza , Xilose/metabolismo
18.
PLoS Comput Biol ; 15(6): e1007127, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31216273

RESUMO

Natural soil is characterized as a complex habitat with patchy hydrated islands and spatially variable nutrients that is in a constant state of change due to wetting-drying dynamics. Soil microbial activity is often concentrated in sparsely distributed hotspots that contribute disproportionally to macroscopic biogeochemical nutrient cycling and greenhouse gas emissions. The mechanistic representation of such dynamic hotspots requires new modeling approaches capable of representing the interplay between dynamic local conditions and the versatile microbial metabolic adaptations. We have developed IndiMeSH (Individual-based Metabolic network model for Soil Habitats) as a spatially explicit model for the physical and chemical microenvironments of soil, combined with an individual-based representation of bacterial motility and growth using adaptive metabolic networks. The model uses angular pore networks and a physically based description of the aqueous phase as a backbone for nutrient diffusion and bacterial dispersal combined with dynamic flux balance analysis to calculate growth rates depending on local nutrient conditions. To maximize computational efficiency, reduced scale metabolic networks are used for the simulation scenarios and evaluated strategically to the genome scale model. IndiMeSH was compared to a well-established population-based spatiotemporal metabolic network model (COMETS) and to experimental data of bacterial spatial organization in pore networks mimicking soil aggregates. IndiMeSH was then used to strategically study dynamic response of a bacterial community to abrupt environmental perturbations and the influence of habitat geometry and hydration conditions. Results illustrate that IndiMeSH is capable of representing trophic interactions among bacterial species, predicting the spatial organization and segregation of bacterial populations due to oxygen and carbon gradients, and provides insights into dynamic community responses as a consequence of environmental changes. The modular design of IndiMeSH and its implementation are adaptable, allowing it to represent a wide variety of experimental and in silico microbial systems.


Assuntos
Bactérias/metabolismo , Ecossistema , Redes e Vias Metabólicas/fisiologia , Modelos Biológicos , Microbiologia do Solo , Algoritmos , Biologia Computacional , Oxigênio/metabolismo , Porosidade , Água/metabolismo
19.
Biophys J ; 117(2): 355-368, 2019 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-31311624

RESUMO

Many computational models for analyzing and predicting cell physiology rely on in vitro data collected in dilute and controlled buffer solutions. However, this can mislead models because up to 40% of the intracellular volume-depending on the organism, the physiology, and the cellular compartment-is occupied by a dense mixture of proteins, lipids, polysaccharides, RNA, and DNA. These intracellular macromolecules interfere with the interactions of enzymes and their reactants and thus affect the kinetics of biochemical reactions, making in vivo reactions considerably more complex than the in vitro data indicates. In this work, we present a new, to our knowledge, type of kinetics that captures and quantifies the effect of volume exclusion and other spatial phenomena on the kinetics of elementary reactions. We further developed a framework that allows for the efficient parameterization of these kinetics using particle simulations. Our formulation, entitled generalized elementary kinetics, can be used to analyze and predict the effect of intracellular crowding on enzymatic reactions and was herein applied to investigate the influence of crowding on phosphoglycerate mutase in Escherichia coli, which exhibits prototypical reversible Michaelis-Menten kinetics. Current research indicates that many enzymes are reaction limited and not diffusion limited, and our results suggest that the influence of fractal diffusion is minimal for these reaction-limited enzymes. Instead, increased association rates and decreased dissociation rates lead to a strong decrease in the effective maximal velocities Vmax and the effective Michaelis-Menten constants KM under physiologically relevant volume occupancies. Finally, the effects of crowding were explored in the context of a linear pathway, with the finding that crowding can have a redistributing effect on the effective flux responses in the case of twofold enzyme overexpression. We suggest that this framework, in combination with detailed kinetics models, will improve our understanding of enzyme reaction networks under nonideal conditions.


Assuntos
Simulação por Computador , Substâncias Macromoleculares/química , Algoritmos , Difusão , Escherichia coli/metabolismo , Cinética , Modelos Lineares , Modelos Moleculares , Tamanho da Partícula
20.
Environ Microbiol ; 21(10): 3548-3563, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31020759

RESUMO

Iron is essential for most living organisms. In addition, its biogeochemical cycling influences important processes in the geosphere (e.g., the mobilization or immobilization of trace elements and contaminants). The reduction of Fe(III) to Fe(II) can be catalysed microbially, particularly by metal-respiring bacteria utilizing Fe(III) as a terminal electron acceptor. Furthermore, Gram-positive fermentative iron reducers are known to reduce Fe(III) by using it as a sink for excess reducing equivalents, as a form of enhanced fermentation. Here, we use the Gram-positive fermentative bacterium Clostridium acetobutylicum as a model system due to its ability to reduce heavy metals. We investigated the reduction of soluble and solid iron during fermentation. We found that exogenous (resazurin, resorufin, anthraquinone-2,6-disulfonate) as well as endogenous (riboflavin) electron mediators enhance solid iron reduction. In addition, iron reduction buffers the pH, and elicits a shift in the carbon and electron flow to less reduced products relative to fermentation. This study underscores the role fermentative bacteria can play in iron cycling and provides insights into the metabolic profile of coupled fermentation and iron reduction with laboratory experiments and metabolic network modelling.


Assuntos
Bactérias/metabolismo , Clostridium acetobutylicum/metabolismo , Ferro/metabolismo , Fermentação , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA