Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
BMC Bioinformatics ; 24(1): 492, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129786

ABSTRACT

BACKGROUND: Flux Balance Analysis (FBA) is a key metabolic modeling method used to simulate cellular metabolism under steady-state conditions. Its simplicity and versatility have led to various strategies incorporating transcriptomic and proteomic data into FBA, successfully predicting flux distribution and phenotypic results. However, despite these advances, the untapped potential lies in leveraging gene-related connections like co-expression patterns for valuable insights. RESULTS: To fill this gap, we introduce ICON-GEMs, an innovative constraint-based model to incorporate gene co-expression network into the FBA model, facilitating more precise determination of flux distributions and functional pathways. In this study, transcriptomic data from both Escherichia coli and Saccharomyces cerevisiae were integrated into their respective genome-scale metabolic models. A comprehensive gene co-expression network was constructed as a global view of metabolic mechanism of the cell. By leveraging quadratic programming, we maximized the alignment between pairs of reaction fluxes and the correlation of their corresponding genes in the co-expression network. The outcomes notably demonstrated that ICON-GEMs outperformed existing methodologies in predictive accuracy. Flux variabilities over subsystems and functional modules also demonstrate promising results. Furthermore, a comparison involving different types of biological networks, including protein-protein interactions and random networks, reveals insights into the utilization of the co-expression network in genome-scale metabolic engineering. CONCLUSION: ICON-GEMs introduce an innovative constrained model capable of simultaneous integration of gene co-expression networks, ready for board application across diverse transcriptomic data sets and multiple organisms. It is freely available as open-source at https://github.com/ThummaratPaklao/ICOM-GEMs.git .


Subject(s)
Proteomics , Systems Biology , Genome , Metabolic Engineering , Gene Expression Profiling , Escherichia coli/genetics , Escherichia coli/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Models, Biological , Metabolic Networks and Pathways/genetics , Metabolic Flux Analysis/methods
2.
BMC Bioinformatics ; 19(1): 368, 2018 Oct 10.
Article in English | MEDLINE | ID: mdl-30305012

ABSTRACT

BACKGROUND: Synthetic biology and related techniques enable genome scale high-throughput investigation of the effect on organism fitness of different gene knock-downs/outs and of other modifications of genomic sequence. RESULTS: We develop statistical and computational pipelines and frameworks for analyzing high throughput fitness data over a genome scale set of sequence variants. Analyzing data from a high-throughput knock-down/knock-out bacterial study, we investigate differences and determinants of the effect on fitness in different conditions. Comparing fitness vectors of genes, across tens of conditions, we observe that fitness consequences strongly depend on genomic location and more weakly depend on gene sequence similarity and on functional relationships. In analyzing promoter sequences, we identified motifs associated with conditions studied in bacterial media such as Casaminos, D-glucose, Sucrose, and other sugars and amino-acid sources. We also use fitness data to infer genes associated with orphan metabolic reactions in the iJO1366 E. coli metabolic model. To do this, we developed a new computational method that integrates gene fitness and gene expression profiles within a given reaction network neighborhood to associate this reaction with a set of genes that potentially encode the catalyzing proteins. We then apply this approach to predict candidate genes for 107 orphan reactions in iJO1366. Furthermore - we validate our methodology with known reactions using a leave-one-out approach. Specifically, using top-20 candidates selected based on combined fitness and expression datasets, we correctly reconstruct 39.7% of the reactions, as compared to 33% based on fitness and to 26% based on expression separately, and to 4.02% as a random baseline. Our model improvement results include a novel association of a gene to an orphan cytosine nucleosidation reaction. CONCLUSION: Our pipeline for metabolic modeling shows a clear benefit of using fitness data for predicting genes of orphan reactions. Along with the analysis pipelines we developed, it can be used to analyze similar high-throughput data.


Subject(s)
Exercise Test/methods , Genome-Wide Association Study/methods , Genomics/methods , Humans , Models, Biological
3.
Mol Biol Rep ; 45(5): 961-972, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30019152

ABSTRACT

In the present study, the effects of individual as well as multiple genes of pentose phosphate pathway (PPP) on human interferon gamma (hIFN-γ) production were analyzed. With overexpression of 6-phosphogluconate dehydrogenase (GND2), 1.9-fold increase in hIFN-γ was achieved, while synergetic effect of 6-phosphogluconolactonase (SOL3) and D-ribulose-5-phosphate 3-epimerase (RPE1) resulted in 2.56-fold increase in hIFN-γ as compared to control. Fed batch fermentation using mixed feeding of gluconate and methanol (carbon source) was carried out, resulting in 80 and 123 mg L-1 of hIFN-γ enhancement in recombinant Pichia GS115 strain encoding codon optimized hIFN-γ (GS115/hIFN-γ) and Pichia GS115 strain encoding codon optimized hIFN-γ with co-expressed 6-phosphogluconolactonase(SOL3) and D-ribulose-5-phosphate 3-epimerase (RPE1) (GS115/hIFN-γ/SR) respectively. To get more insight of the flux distribution towards hIFN-γ, studies were carried out by applying flux balance analysis during methanol fed batch phase for both strains. In both strains (GS115/hIFN-γ and GS115/hIFN-γ/SR) more than 95% of formaldehyde flux is directed towards assimilatory pathway. The analysis revealed that with the overexpression of SOL3 and RPE1 the flux towards PPP triggering the alleviation in hIFN-γ production.


Subject(s)
Interferon-gamma/genetics , Metabolic Engineering/methods , Pentose Phosphate Pathway , Pichia/growth & development , Batch Cell Culture Techniques , Carbohydrate Epimerases/genetics , Carboxylic Ester Hydrolases/genetics , Fermentation , Humans , Interferon-gamma/metabolism , Phosphogluconate Dehydrogenase/genetics , Pichia/genetics , Recombinant Proteins/metabolism
4.
New Phytol ; 213(4): 1726-1739, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27861943

ABSTRACT

Tomato is a model organism to study the development of fleshy fruit including ripening initiation. Unfortunately, few studies deal with the brief phase of accelerated ripening associated with the respiration climacteric because of practical problems involved in measuring fruit respiration. Because constraint-based modelling allows predicting accurate metabolic fluxes, we investigated the respiration and energy dissipation of fruit pericarp at the breaker stage using a detailed stoichiometric model of the respiratory pathway, including alternative oxidase and uncoupling proteins. Assuming steady-state, a metabolic dataset was transformed into constraints to solve the model on a daily basis throughout tomato fruit development. We detected a peak of CO2 released and an excess of energy dissipated at 40 d post anthesis (DPA) just before the onset of ripening coinciding with the respiration climacteric. We demonstrated the unbalanced carbon allocation with the sharp slowdown of accumulation (for syntheses and storage) and the beginning of the degradation of starch and cell wall polysaccharides. Experiments with fruits harvested from plants cultivated under stress conditions confirmed the concept. We conclude that modelling with an accurate metabolic dataset is an efficient tool to bypass the difficulty of measuring fruit respiration and to elucidate the underlying mechanisms of ripening.


Subject(s)
Fruit/cytology , Fruit/physiology , Models, Biological , Solanum lycopersicum/cytology , Solanum lycopersicum/physiology , Adenosine Triphosphate/metabolism , Carbohydrate Metabolism , Carbon/metabolism , Carbon Dioxide/metabolism , Cell Respiration , Fruit/growth & development , Fruit/metabolism , Solanum lycopersicum/growth & development , Solanum lycopersicum/metabolism , Nitrogen/metabolism , Stress, Physiological , Sucrose/metabolism , Thermogenesis , Time Factors
5.
Brief Bioinform ; 15(1): 108-22, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23131418

ABSTRACT

Flux balance analysis (FBA) is a widely used computational method for characterizing and engineering intrinsic cellular metabolism. The increasing number of its successful applications and growing popularity are possibly attributable to the availability of specific software tools for FBA. Each tool has its unique features and limitations with respect to operational environment, user-interface and supported analysis algorithms. Presented herein is an in-depth evaluation of currently available FBA applications, focusing mainly on usability, functionality, graphical representation and inter-operability. Overall, most of the applications are able to perform basic features of model creation and FBA simulation. COBRA toolbox, OptFlux and FASIMU are versatile to support advanced in silico algorithms to identify environmental and genetic targets for strain design. SurreyFBA, WEbcoli, Acorn, FAME, GEMSiRV and MetaFluxNet are the distinct tools which provide the user friendly interfaces in model handling. In terms of software architecture, FBA-SimVis and OptFlux have the flexible environments as they enable the plug-in/add-on feature to aid prospective functional extensions. Notably, an increasing trend towards the implementation of more tailored e-services such as central model repository and assistance to collaborative efforts was observed among the web-based applications with the help of advanced web-technologies. Furthermore, most recent applications such as the Model SEED, FAME, MetaFlux and MicrobesFlux have even included several routines to facilitate the reconstruction of genome-scale metabolic models. Finally, a brief discussion on the future directions of FBA applications was made for the benefit of potential tool developers.


Subject(s)
Metabolic Flux Analysis/statistics & numerical data , Software , Algorithms , Computational Biology , Computer Simulation , Genomics/statistics & numerical data , Metabolic Flux Analysis/trends , Models, Biological , Models, Genetic , Phenotype , User-Computer Interface
6.
J Ind Microbiol Biotechnol ; 42(10): 1401-14, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26254041

ABSTRACT

Optimizing the overall NADPH turnover is one of the key challenges in various value-added biochemical syntheses. In this work, we first analyzed the NADPH regeneration potentials of common cell factories, including Escherichia coli, Saccharomyces cerevisiae, Bacillus subtilis, and Pichia pastoris across multiple environmental conditions and determined E. coli and glycerol as the best microbial chassis and most suitable carbon source, respectively. In addition, we identified optimal cofactor specificity engineering (CSE) enzyme targets, whose cofactors when switched from NAD(H) to NADP(H) improve the overall NADP(H) turnover. Among several enzyme targets, glyceraldehyde-3-phosphate dehydrogenase was recognized as a global candidate since its CSE improved the NADP(H) regeneration under most of the conditions examined. Finally, by analyzing the protein structures of all CSE enzyme targets via homology modeling, we established that the replacement of conserved glutamate or aspartate with serine in the loop region could change the cofactor dependence from NAD(H) to NADP(H).


Subject(s)
Bacillus subtilis/metabolism , Bioreactors/microbiology , Computer Simulation , Escherichia coli/metabolism , NADP/metabolism , Pichia/metabolism , Saccharomyces cerevisiae/metabolism , Bacillus subtilis/enzymology , Enzymes/chemistry , Enzymes/metabolism , Escherichia coli/enzymology , Glycerol/metabolism , NAD/metabolism , Oxidation-Reduction , Saccharomyces cerevisiae/enzymology , Serine/metabolism
7.
Comput Struct Biotechnol J ; 21: 3736-3745, 2023.
Article in English | MEDLINE | ID: mdl-37547082

ABSTRACT

The biomass equation is a critical component in genome-scale metabolic models (GEMs): it is used as the de facto objective function in flux balance analysis (FBA). This equation accounts for the quantities of all known biomass precursors that are required for cell growth based on the macromolecular and monomer compositions measured at certain conditions. However, it is often reported that the macromolecular composition of cells could change across different environmental conditions and thus the use of the same single biomass equation in FBA, under multiple conditions, is questionable. Herein, we first investigated the qualitative and quantitative variations of macromolecular compositions of three representative host organisms, Escherichia coli, Saccharomyces cerevisiae and Cricetulus griseus, across different environmental/genetic variations. While macromolecular building blocks such as RNA, protein, and lipid composition vary notably, changes in fundamental biomass monomer units such as nucleotides and amino acids are not appreciable. We also observed that flux predictions through FBA is quite sensitive to macromolecular compositions but not the monomer compositions. Based on these observations, we propose ensemble representations of biomass equation in FBA to account for the natural variation of cellular constituents. Such ensemble representations of biomass better predicted the flux through anabolic reactions as it allows for the flexibility in the biosynthetic demands of the cells. The current study clearly highlights that certain component of the biomass equation indeed vary across different conditions, and the ensemble representation of biomass equation in FBA by accounting for such natural variations could avoid inaccuracies that may arise from in silico simulations.

8.
Front Genet ; 13: 1084727, 2022.
Article in English | MEDLINE | ID: mdl-36726720

ABSTRACT

Metabolism of an organism underlies its phenotype, which depends on many factors, such as the genetic makeup, habitat, and stresses to which it is exposed. This is particularly important for the prokaryotes, which undergo significant vertical and horizontal gene transfers. In this study we have used the energy-intensive Aromatic Amino Acid (Tryptophan, Tyrosine and Phenylalanine, TTP) biosynthesis pathway, in a large number of prokaryotes, as a model system to query the different levels of organization of metabolism in the whole intracellular biochemical network, and to understand how perturbations, such as mutations, affects the metabolic flux through the pathway - in isolation and in the context of other pathways connected to it. Using an agglomerative approach involving complex network analysis and Flux Balance Analyses (FBA), of the Tryptophan, Tyrosine and Phenylalanine and other pathways connected to it, we identify several novel results. Using the reaction network analysis and Flux Balance Analyses of the Tryptophan, Tyrosine and Phenylalanine and the genome-scale reconstructed metabolic pathways, many common hubs between the connected networks and the whole genome network are identified. The results show that the connected pathway network can act as a proxy for the whole genome network in Prokaryotes. This systems level analysis also points towards designing functional smaller synthetic pathways based on the reaction network and Flux Balance Analyses analysis.

9.
ACS Synth Biol ; 11(8): 2672-2684, 2022 08 19.
Article in English | MEDLINE | ID: mdl-35801944

ABSTRACT

Flux balance analysis (FBA) and ordinary differential equation models have been instrumental in depicting the metabolic functioning of a cell. Nevertheless, they demonstrate a population's average behavior (summation of individuals), thereby portraying homogeneity. However, living organisms such as Escherichia coli contain more biochemical reactions than engaging metabolites, making them an underdetermined and degenerate system. This results in a heterogeneous population with varying metabolic patterns. We have formulated a population systems biology model that predicts this degeneracy by emulating a diverse metabolic makeup with unique biochemical signatures. The model mimics the universally accepted experimental view that a subpopulation of bacteria, even under normal growth conditions, renders a unique biochemical state, leading to the synthesis of metabolites and persister progenitors of antibiotic resistance and biofilms. We validate the platform's predictions by producing commercially important heterologous (isobutanol) and homologous (shikimate) metabolites. The predicted fluxes are tested in vitro resulting in 32- and 42-fold increased product of isobutanol and shikimate, respectively. Moreover, we authenticate the platform by mimicking a bacterial population in the presence of glyphosate, a metabolic pathway inhibitor. Here, we observe a fraction of subsisting persisters despite inhibition, thus affirming the signature of a heterogeneous populace. The platform has multiple uses based on the disposition of the user.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Computer Simulation , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Humans , Metabolic Networks and Pathways , Models, Biological
10.
Front Genet ; 12: 586293, 2021.
Article in English | MEDLINE | ID: mdl-33633777

ABSTRACT

Microbial life in the oceans impacts the entire marine ecosystem, global biogeochemistry and climate. The marine cyanobacterium Prochlorococcus, an abundant component of this ecosystem, releases a significant fraction of the carbon fixed through photosynthesis, but the amount, timing and molecular composition of released carbon are still poorly understood. These depend on several factors, including nutrient availability, light intensity and glycogen storage. Here we combine multiple computational approaches to provide insight into carbon storage and exudation in Prochlorococcus. First, with the aid of a new algorithm for recursive filling of metabolic gaps (ReFill), and through substantial manual curation, we extended an existing genome-scale metabolic model of Prochlorococcus MED4. In this revised model (iSO595), we decoupled glycogen biosynthesis/degradation from growth, thus enabling dynamic allocation of carbon storage. In contrast to standard implementations of flux balance modeling, we made use of forced influx of carbon and light into the cell, to recapitulate overflow metabolism due to the decoupling of photosynthesis and carbon fixation from growth during nutrient limitation. By using random sampling in the ensuing flux space, we found that storage of glycogen or exudation of organic acids are favored when the growth is nitrogen limited, while exudation of amino acids becomes more likely when phosphate is the limiting resource. We next used COMETS to simulate day-night cycles and found that the model displays dynamic glycogen allocation and exudation of organic acids. The switch from photosynthesis and glycogen storage to glycogen depletion is associated with a redistribution of fluxes from the Entner-Doudoroff to the Pentose Phosphate pathway. Finally, we show that specific gene knockouts in iSO595 exhibit dynamic anomalies compatible with experimental observations, further demonstrating the value of this model as a tool to probe the metabolic dynamic of Prochlorococcus.

11.
Genes (Basel) ; 12(6)2021 05 24.
Article in English | MEDLINE | ID: mdl-34073716

ABSTRACT

The current SARS-CoV-2 pandemic is still threatening humankind. Despite first successes in vaccine development and approval, no antiviral treatment is available for COVID-19 patients. The success is further tarnished by the emergence and spreading of mutation variants of SARS-CoV-2, for which some vaccines have lower efficacy. This highlights the urgent need for antiviral therapies even more. This article describes how the genome-scale metabolic model (GEM) of the host-virus interaction of human alveolar macrophages and SARS-CoV-2 was refined by incorporating the latest information about the virus's structural proteins and the mutant variants B.1.1.7, B.1.351, B.1.28, B.1.427/B.1.429, and B.1.617. We confirmed the initially identified guanylate kinase as a potential antiviral target with this refined model and identified further potential targets from the purine and pyrimidine metabolism. The model was further extended by incorporating the virus' lipid requirements. This opened new perspectives for potential antiviral targets in the altered lipid metabolism. Especially the phosphatidylcholine biosynthesis seems to play a pivotal role in viral replication. The guanylate kinase is even a robust target in all investigated mutation variants currently spreading worldwide. These new insights can guide laboratory experiments for the validation of identified potential antiviral targets. Only the combination of vaccines and antiviral therapies will effectively defeat this ongoing pandemic.


Subject(s)
COVID-19/metabolism , COVID-19/virology , Energy Metabolism , Genome, Viral , Guanylate Kinases/metabolism , Host-Pathogen Interactions , Mutation , SARS-CoV-2/genetics , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , COVID-19/genetics , Gene Knockdown Techniques , Humans , Lipid Metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , SARS-CoV-2/drug effects , Viral Load , Virus Replication , COVID-19 Drug Treatment
12.
Biotechnol Genet Eng Rev ; 36(1): 32-55, 2020 Apr.
Article in English | MEDLINE | ID: mdl-33292061

ABSTRACT

Systems biology is one of the integrated ways to study biological systems and is more favourable than the earlier used approaches. It includes metabolic pathway analysis, modelling, and regulatory as well as signal transduction for getting insights into cellular behaviour. Among the various techniques of modelling, simulation, analysis of networks and pathways, flux-based analysis (FBA) has been recognised because of its extensibility as well as simplicity. It is widely accepted because it is not like a mechanistic simulation which depends on accurate kinetic data. The study of fluxes through the network is informative and can give insights even in the absence of kinetic data. FBA is one of the widely used tools to study biochemical networks and needs information of reaction stoichiometry, growth requirements, specific measurement parameters of the biological system, in particular the reconstruction of the metabolic network for the genome-scale, many of which have already been built previously. It defines the boundaries of flux distributions which are possible and achievable with a defined set of genes. This review article gives an insight into FBA, from the extension of flux balancing to mathematical representation followed by a discussion about the formulation of flux-balance analysis problems, defining constraints for the stoichiometry of the pathways and the tools that can be used in FBA such as FASIMA, COBRA toolbox, and OptFlux. It also includes broader areas in terms of applications which can be covered by FBA as well as the queries which can be addressed through FBA.


Subject(s)
Computational Biology/trends , Models, Biological , Physical Phenomena , Systems Biology/trends , Algorithms , Computer Simulation , Gene Regulatory Networks/genetics , Kinetics , Metabolic Networks and Pathways/genetics , Signal Transduction/genetics
13.
Front Cell Dev Biol ; 8: 566702, 2020.
Article in English | MEDLINE | ID: mdl-33251208

ABSTRACT

Pathogens give rise to a wide range of diseases threatening global health and hence drawing public health agencies' attention to establish preventative and curative solutions. Genome-scale metabolic modeling is ever increasingly used tool for biomedical applications including the elucidation of antibiotic resistance, virulence, single pathogen mechanisms and pathogen-host interaction systems. With this approach, the sophisticated cellular system of metabolic reactions inside the pathogens as well as between pathogen and host cells are represented in conjunction with their corresponding genes and enzymes. Along with essential metabolic reactions, alternate pathways and fluxes are predicted by performing computational flux analyses for the growth of pathogens in a very short time. The genes or enzymes responsible for the essential metabolic reactions in pathogen growth are regarded as potential drug targets, as a priori guide to researchers in the pharmaceutical field. Pathogens alter the key metabolic processes in infected host, ultimately the objective of these integrative constraint-based context-specific metabolic models is to provide novel insights toward understanding the metabolic basis of the acute and chronic processes of infection, revealing cellular mechanisms of pathogenesis, identifying strain-specific biomarkers and developing new therapeutic approaches including the combination drugs. The reaction rates predicted during different time points of pathogen development enable us to predict active pathways and those that only occur during certain stages of infection, and thus point out the putative drug targets. Among others, fatty acid and lipid syntheses reactions are recent targets of new antimicrobial drugs. Genome-scale metabolic models provide an improved understanding of how intracellular pathogens utilize the existing microenvironment of the host. Here, we reviewed the current knowledge of genome-scale metabolic modeling in pathogen cells as well as pathogen host interaction systems and the promising applications in the extension of curative strategies against pathogens for global preventative healthcare.

14.
Methods Mol Biol ; 2088: 315-330, 2020.
Article in English | MEDLINE | ID: mdl-31893380

ABSTRACT

The drug development pipeline has stalled because of the difficulty in identifying new drug targets while minimizing off-target effects. Computational methods, such as the use of metabolic network reconstructions, may provide a cost-effective platform to test new hypotheses for drug targets and prevent off-target effects. Here, we summarize available methods to identify drug targets and off-target effects using either reaction-centric, gene-centric, or metabolite-centric approaches with genome-scale metabolic network reconstructions.


Subject(s)
Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Pharmaceutical Preparations/administration & dosage , Computational Biology/methods , Drug Delivery Systems/methods , Genome/physiology , Humans
15.
Metabolites ; 10(5)2020 May 06.
Article in English | MEDLINE | ID: mdl-32384607

ABSTRACT

Folate deficiency in the critical developmental period has been repeatedly associated with an increased risk of Autism spectrum disorders (ASD), but the key pathophysiological mechanism has not yet been identified. In this work, we focused on identifying genes whose defect has similar consequences to folate depletion in the metabolic network. Within the Flux Balance Analysis (FBA) framework, we developed a method of blocked metabolites that allowed us to define the metabolic consequences of various gene defects and folate depletion. We identified six genes (GART, PFAS, PPAT, PAICS, ATIC, and ADSL) whose blocking results in nearly the same effect in the metabolic network as folate depletion. All of these genes form the purine biosynthetic pathway. We found that, just like folate depletion, the blockade of any of the six genes mentioned above results in a blockage of purine metabolism. We hypothesize that this can lead to decreased adenosine triphosphate (ATP) and subsequently, an S-adenosyl methionine (SAM) pool in neurons in the case of rapid cell division. Based on our results, we consider the methylation defect to be a potential cause of ASD, due to the depletion of purine, and consequently S-adenosyl methionine (SAM), biosynthesis.

16.
Nat Metab ; 1(1): 125-132, 2019 01.
Article in English | MEDLINE | ID: mdl-32694810

ABSTRACT

The principles governing cellular metabolic operation are poorly understood. Because diverse organisms show similar metabolic flux patterns, we hypothesized that a fundamental thermodynamic constraint might shape cellular metabolism. Here, we develop a constraint-based model for Saccharomyces cerevisiae with a comprehensive description of biochemical thermodynamics including a Gibbs energy balance. Non-linear regression analyses of quantitative metabolome and physiology data reveal the existence of an upper rate limit for cellular Gibbs energy dissipation. By applying this limit in flux balance analyses with growth maximization as the objective function, our model correctly predicts the physiology and intracellular metabolic fluxes for different glucose uptake rates as well as the maximal growth rate. We find that cells arrange their intracellular metabolic fluxes in such a way that, with increasing glucose uptake rates, they can accomplish optimal growth rates but stay below the critical rate limit on Gibbs energy dissipation. Once all possibilities for intracellular flux redistribution are exhausted, cells reach their maximal growth rate. This principle also holds for Escherichia coli and different carbon sources. Our work proposes that metabolic reaction stoichiometry, a limit on the cellular Gibbs energy dissipation rate, and the objective of growth maximization shape metabolism across organisms and conditions.


Subject(s)
Energy Metabolism , Models, Biological , Biochemical Phenomena , Escherichia coli/metabolism , Glucose/metabolism , Phenotype , Saccharomyces cerevisiae/metabolism , Thermodynamics
17.
Methods Mol Biol ; 1975: 305-320, 2019.
Article in English | MEDLINE | ID: mdl-31062316

ABSTRACT

Stem cell metabolism is intrinsically tied to stem cell pluripotency and function. Yet, understanding metabolic rewiring in stem cells has been challenging due to the complex and highly interconnected nature of the metabolic network. Genome-scale metabolic network models are increasingly used to holistically model the metabolic behavior of various cells and tissues using transcriptomics data. However, these powerful approaches that model steady-state behavior have limited utility for studying dynamic stem cell state transitions. To address this complexity, we recently developed the dynamic flux activity (DFA) approach; DFA is a genome-scale modeling approach that uses time-course metabolic data to predict metabolic flux rewiring. This protocol outlines the steps for modeling steady-state and dynamic metabolic behavior using transcriptomics and time-course metabolomics data, respectively. Using data from naive and primed pluripotent stem cells, we demonstrate how we can use genome-scale modeling and DFA to comprehensively characterize the metabolic differences between these states.


Subject(s)
Cell Differentiation , Cell Lineage , Computational Biology/methods , Gene Regulatory Networks , Metabolome , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Transcriptome
18.
Biotechnol Biofuels ; 12: 65, 2019.
Article in English | MEDLINE | ID: mdl-30962820

ABSTRACT

BACKGROUND: l-Histidine biosynthesis is embedded in an intertwined metabolic network which renders microbial overproduction of this amino acid challenging. This is reflected in the few available examples of histidine producers in literature. Since knowledge about the metabolic interplay is limited, we systematically perturbed the metabolism of Corynebacterium glutamicum to gain a holistic understanding in the metabolic limitations for l-histidine production. We, therefore, constructed C. glutamicum strains in a modularized metabolic engineering approach and analyzed them with LC/MS-QToF-based systems metabolic profiling (SMP) supported by flux balance analysis (FBA). RESULTS: The engineered strains produced l-histidine, equimolar amounts of glycine, and possessed heavily decreased intracellular adenylate concentrations, despite a stable adenylate energy charge. FBA identified regeneration of ATP from 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) as crucial step for l-histidine production and SMP identified strong intracellular accumulation of inosine monophosphate (IMP) in the engineered strains. Energy engineering readjusted the intracellular IMP and ATP levels to wild-type niveau and reinforced the intrinsic low ATP regeneration capacity to maintain a balanced energy state of the cell. SMP further indicated limitations in the C1 supply which was overcome by expression of the glycine cleavage system from C. jeikeium. Finally, we rerouted the carbon flux towards the oxidative pentose phosphate pathway thereby further increasing product yield to 0.093 ± 0.003 mol l-histidine per mol glucose. CONCLUSION: By applying the modularized metabolic engineering approach combined with SMP and FBA, we identified an intrinsically low ATP regeneration capacity, which prevents to maintain a balanced energy state of the cell in an l-histidine overproduction scenario and an insufficient supply of C1 units. To overcome these limitations, we provide a metabolic engineering strategy which constitutes a general approach to improve the production of ATP and/or C1 intensive products.

19.
Methods Mol Biol ; 1835: 229-242, 2018.
Article in English | MEDLINE | ID: mdl-30109656

ABSTRACT

Synthetic biology is an emergent field of research whose aim is to make biology an engineering discipline, thus permitting to design, control, and standardize biological processes. Synthetic biology is therefore expected to boost the development of biotechnological processes such as protein production and enzyme engineering, which can be significantly relevant for lipases and esterases.


Subject(s)
Esterases/biosynthesis , Lipase/biosynthesis , Metabolic Engineering/methods , Synthetic Biology/methods , Biotechnology
20.
BMC Syst Biol ; 11(1): 50, 2017 04 19.
Article in English | MEDLINE | ID: mdl-28420402

ABSTRACT

BACKGROUND: Essential reactions are vital components of cellular networks. They are the foundations of synthetic biology and are potential candidate targets for antimetabolic drug design. Especially if a single reaction is catalyzed by multiple enzymes, then inhibiting the reaction would be a better option than targeting the enzymes or the corresponding enzyme-encoding gene. The existing databases such as BRENDA, BiGG, KEGG, Bio-models, Biosilico, and many others offer useful and comprehensive information on biochemical reactions. But none of these databases especially focus on essential reactions. Therefore, building a centralized repository for this class of reactions would be of great value. DESCRIPTION: Here, we present a species-specific essential reactions database (SSER). The current version comprises essential biochemical and transport reactions of twenty-six organisms which are identified via flux balance analysis (FBA) combined with manual curation on experimentally validated metabolic network models. Quantitative data on the number of essential reactions, number of the essential reactions associated with their respective enzyme-encoding genes and shared essential reactions across organisms are the main contents of the database. CONCLUSION: SSER would be a prime source to obtain essential reactions data and related gene and metabolite information and it can significantly facilitate the metabolic network models reconstruction and analysis, and drug target discovery studies. Users can browse, search, compare and download the essential reactions of organisms of their interest through the website http://cefg.uestc.edu.cn/sser .


Subject(s)
Computational Biology/methods , Databases, Factual , Metabolic Flux Analysis
SELECTION OF CITATIONS
SEARCH DETAIL