Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article in English | MEDLINE | ID: mdl-33723053

ABSTRACT

Metal ions are vital to metabolism, as they can act as cofactors on enzymes and thus modulate individual enzymatic reactions. Although many enzymes have been reported to interact with metal ions, the quantitative relationships between metal ions and metabolism are lacking. Here, we reconstructed a genome-scale metabolic model of the yeast Saccharomyces cerevisiae to account for proteome constraints and enzyme cofactors such as metal ions, named CofactorYeast. The model is able to estimate abundances of metal ions binding on enzymes in cells under various conditions, which are comparable to measured metal ion contents in biomass. In addition, the model predicts distinct metabolic flux distributions in response to reduced levels of various metal ions in the medium. Specifically, the model reproduces changes upon iron deficiency in metabolic and gene expression levels, which could be interpreted by optimization principles (i.e., yeast optimizes iron utilization based on metabolic network and enzyme kinetics rather than preferentially targeting iron to specific enzymes or pathways). At last, we show the potential of using the model for understanding cell factories that harbor heterologous iron-containing enzymes to synthesize high-value compounds such as p-coumaric acid. Overall, the model demonstrates the dependence of enzymes on metal ions and links metal ions to metabolism on a genome scale.


Subject(s)
Metabolic Engineering , Metabolic Networks and Pathways , Metals/metabolism , Saccharomyces cerevisiae/physiology , Coenzymes/metabolism , Ions/metabolism , Proteome
2.
J Anim Ecol ; 92(6): 1203-1215, 2023 06.
Article in English | MEDLINE | ID: mdl-37158280

ABSTRACT

Metabolic scaling provides valuable information about the physiological and ecological functions of organisms, although few studies have quantified the metabolic scaling exponent (b) of communities under natural conditions. Maximum entropy theory of ecology (METE) is a constraint-based unified theory with the potential to empirically assess the spatial variation of the metabolic scaling. Our main goal is to develop a novel method of estimating b within a community by integrating metabolic scaling and METE. We also aim to study the relationships between the estimated b and environmental variables across communities. We developed a new METE framework to estimate b in 118 stream fish communities in the north-eastern Iberian Peninsula. We first extended the original maximum entropy model by parameterizing b in the model prediction of the community-level individual size distributions and compared our results with empirical and theoretical predictions. We then tested the effects of abiotic conditions, species composition and human disturbance on the spatial variation of community-level b. We found that community-level b of the best maximum entropy models showed great spatial variability, ranging from 0.25 to 2.38. The mean exponent (b = 0.93) resembled the community-aggregated mean values from three previous metabolic scaling meta-analyses, all of which were greater than the theoretical predictions of 0.67 and 0.75. Furthermore, the generalized additive model showed that b reached maximum at the intermediate mean annual precipitation level and declined significantly as human disturbance intensified. The parameterized METE is proposed here as a novel framework for estimating the metabolic pace of life of stream fish communities. The large spatial variation of b may reflect the combined effects of environmental constraints and species interactions, which likely have important feedback on the structure and function of natural communities. Our newly developed framework can also be applied to study the impact of global environmental pressures on metabolic scaling and energy use in other ecosystems.


Subject(s)
Ecosystem , Rivers , Animals , Humans , Entropy , Models, Biological
3.
Proc Natl Acad Sci U S A ; 117(15): 8494-8502, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32229570

ABSTRACT

Human tuberculosis is caused by members of the Mycobacterium tuberculosis complex (MTBC) that vary in virulence and transmissibility. While genome-wide association studies have uncovered several mutations conferring drug resistance, much less is known about the factors underlying other bacterial phenotypes. Variation in the outcome of tuberculosis infection and diseases has been attributed primarily to patient and environmental factors, but recent evidence indicates an additional role for the genetic diversity among MTBC clinical strains. Here, we used metabolomics to unravel the effect of genetic variation on the strain-specific metabolic adaptive capacity and vulnerability. To define the functionality of single-nucleotide polymorphisms (SNPs) systematically, we developed a constraint-based approach that integrates metabolomic and genomic data. Our model-based predictions correctly classify SNP effects in pyruvate kinase and suggest a genetic basis for strain-specific inherent baseline susceptibility to the antibiotic para-aminosalicylic acid. Our method is broadly applicable across microbial life, opening possibilities for the development of more selective treatment strategies.


Subject(s)
Antitubercular Agents/pharmacology , Genomics/methods , Host-Pathogen Interactions , Metabolome , Mycobacterium tuberculosis/genetics , Polymorphism, Single Nucleotide , Tuberculosis/genetics , Aminosalicylic Acid/pharmacology , Genome, Bacterial , Genome-Wide Association Study , Humans , Models, Molecular , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Phenotype , Phylogeny , Pyruvate Kinase/metabolism , Tuberculosis/drug therapy , Tuberculosis/microbiology , Virulence
4.
Mar Drugs ; 20(2)2022 Feb 01.
Article in English | MEDLINE | ID: mdl-35200644

ABSTRACT

Docosahexaenoic acid (DHA) is one of the most important long-chain polyunsaturated fatty acids (LC-PUFAs), with numerous health benefits. Crypthecodinium cohnii, a marine heterotrophic dinoflagellate, is successfully used for the industrial production of DHA because it can accumulate DHA at high concentrations within the cells. Glycerol is an interesting renewable substrate for DHA production since it is a by-product of biodiesel production and other industries, and is globally generated in large quantities. The DHA production potential from glycerol, ethanol and glucose is compared by combining fermentation experiments with the pathway-scale kinetic modeling and constraint-based stoichiometric modeling of C. cohnii metabolism. Glycerol has the slowest biomass growth rate among the tested substrates. This is partially compensated by the highest PUFAs fraction, where DHA is dominant. Mathematical modeling reveals that glycerol has the best experimentally observed carbon transformation rate into biomass, reaching the closest values to the theoretical upper limit. In addition to our observations, the published experimental evidence indicates that crude glycerol is readily consumed by C. cohnii, making glycerol an attractive substrate for DHA production.


Subject(s)
Dinoflagellida/metabolism , Docosahexaenoic Acids/metabolism , Models, Theoretical , Biomass , Ethanol/metabolism , Fermentation , Glucose/metabolism , Glycerol/metabolism
5.
Cell Mol Life Sci ; 77(3): 489-495, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31748916

ABSTRACT

Genome-scale metabolic models have been successfully applied to study the metabolism of multiple plant species in the past decade. While most existing genome-scale modelling studies have focussed on studying the metabolic behaviour of individual plant metabolic systems, there is an increasing focus on combining models of multiple tissues or organs to produce multi-tissue models that allow the investigation of metabolic interactions between tissues and organs. Multi-tissue metabolic models were constructed for multiple plants including Arabidopsis, barley, soybean and Setaria. These models were applied to study various aspects of plant physiology including the division of labour between organs, source and sink tissue relationship, growth of different tissues and organs and charge and proton balancing. In this review, we outline the process of constructing multi-tissue genome-scale metabolic models, discuss the strengths and challenges in using multi-tissue models, review the current status of plant multi-tissue and whole plant metabolic models and explore the approaches for integrating genome-scale metabolic models into multi-scale plant models.


Subject(s)
Metabolic Networks and Pathways/genetics , Plants/genetics , Plants/metabolism , Genome, Plant/genetics , Humans , Models, Biological
6.
BMC Bioinformatics ; 21(1): 67, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32085724

ABSTRACT

BACKGROUND: Constraint-based metabolic modeling has been applied to understand metabolism related disease mechanisms, to predict potential new drug targets and anti-metabolites, and to identify biomarkers of complex diseases. Although the state-of-art modeling toolbox, COBRA 3.0, is powerful, it requires substantial computing time conducting flux balance analysis, knockout analysis, and Markov Chain Monte Carlo (MCMC) sampling, which may limit its application in large scale genome-wide analysis. RESULTS: Here, we rewrote the underlying code of COBRA 3.0 using C/C++, and developed a toolbox, termed FastMM, to effectively conduct constraint-based metabolic modeling. The results showed that FastMM is 2~400 times faster than COBRA 3.0 in performing flux balance analysis and knockout analysis and returns consistent outputs. When applied to MCMC sampling, FastMM is 8 times faster than COBRA 3.0. FastMM is also faster than some efficient metabolic modeling applications, such as Cobrapy and Fast-SL. In addition, we developed a Matlab/Octave interface for fast metabolic modeling. This interface was fully compatible with COBRA 3.0, enabling users to easily perform complex applications for metabolic modeling. For example, users who do not have deep constraint-based metabolic model knowledge can just type one command in Matlab/Octave to perform personalized metabolic modeling. Users can also use the advance and multiple threading parameters for complex metabolic modeling. Thus, we provided an efficient and user-friendly solution to perform large scale genome-wide metabolic modeling. For example, FastMM can be applied to the modeling of individual cancer metabolic profiles of hundreds to thousands of samples in the Cancer Genome Atlas (TCGA). CONCLUSION: FastMM is an efficient and user-friendly toolbox for large-scale personalized constraint-based metabolic modeling. It can serve as a complementary and invaluable improvement to the existing functionalities in COBRA 3.0. FastMM is under GPL license and can be freely available at GitHub site: https://github.com/GonghuaLi/FastMM.


Subject(s)
Metabolic Networks and Pathways , Software , Genome , Humans , Models, Biological , Neoplasms/genetics , Neoplasms/metabolism
7.
Biotechnol Bioeng ; 117(11): 3545-3558, 2020 11.
Article in English | MEDLINE | ID: mdl-32648961

ABSTRACT

Lactic acid is widely used in many industries, especially in the production of poly-lactic acid. Bacillus coagulans is a promising lactic acid producer in industrial fermentation due to its thermophilic property. In this study, we developed the first genome-scale metabolic model (GEM) of B. coagulans iBag597, together with an enzyme-constrained model ec-iBag597. We measured strain-specific biomass composition and integrated the data into a biomass equation. Then, we validated iBag597 against experimental data generated in this study, including amino acid requirements and carbon source utilization, showing that simulations were generally consistent with the experimental results. Subsequently, we carried out chemostats to investigate the effects of specific growth rate and culture pH on metabolism of B. coagulans. Meanwhile, we used iBag597 to estimate the intracellular metabolic fluxes for those conditions. The results showed that B. coagulans was capable of generating ATP via multiple pathways, and switched among them in response to various conditions. With ec-iBag597, we estimated the protein cost and protein efficiency for each ATP-producing pathway to investigate the switches. Our models pave the way for systems biology of B. coagulans, and our findings suggest that maintaining a proper growth rate and selecting an optimal pH are beneficial for lactate fermentation.


Subject(s)
Bacillus coagulans , Genome, Bacterial/genetics , Lactic Acid/metabolism , Models, Biological , Bacillus coagulans/genetics , Bacillus coagulans/metabolism , Fermentation , Hydrogen-Ion Concentration , Metabolic Engineering
8.
BMC Bioinformatics ; 20(1): 386, 2019 Jul 10.
Article in English | MEDLINE | ID: mdl-31291905

ABSTRACT

BACKGROUND: Mathematical models of biological networks can provide important predictions and insights into complex disease. Constraint-based models of cellular metabolism and probabilistic models of gene regulatory networks are two distinct areas that have progressed rapidly in parallel over the past decade. In principle, gene regulatory networks and metabolic networks underly the same complex phenotypes and diseases. However, systematic integration of these two model systems remains a fundamental challenge. RESULTS: In this work, we address this challenge by fusing probabilistic models of gene regulatory networks into constraint-based models of metabolism. The novel approach utilizes probabilistic reasoning in BN models of regulatory networks serves as the "glue" that enables a natural interface between the two systems. Probabilistic reasoning is used to predict and quantify system-wide effects of perturbation to the regulatory network in the form of constraints for flux variability analysis. In this setting, both regulatory and metabolic networks inherently account for uncertainty. Applications leverage constraint-based metabolic models of brain metabolism and gene regulatory networks parameterized by gene expression data from the hippocampus to investigate the role of the HIF-1 pathway in Alzheimer's disease. Integrated models support HIF-1A as effective target to reduce the effects of hypoxia in Alzheimer's disease. However, HIF-1A activation is far less effective in shifting metabolism when compared to brain metabolism in healthy controls. CONCLUSIONS: The direct integration of probabilistic regulatory networks into constraint-based models of metabolism provides novel insights into how perturbations in the regulatory network may influence metabolic states. Predictive modeling of enzymatic activity can be facilitated using probabilistic reasoning, thereby extending the predictive capacity of the network. This framework for model integration is generalizable to other systems.


Subject(s)
Alzheimer Disease/metabolism , Metabolic Networks and Pathways , Models, Biological , Models, Statistical , Bayes Theorem , Enzymes/metabolism , Gene Regulatory Networks , Humans , Phenotype , Signal Transduction
9.
Curr Genomics ; 20(4): 252-259, 2019 May.
Article in English | MEDLINE | ID: mdl-32030085

ABSTRACT

BACKGROUND: Constraint-based metabolic network models have been widely used in pheno-typic prediction and metabolic engineering design. In recent years, researchers have attempted to im-prove prediction accuracy by integrating regulatory information and multiple types of "omics" data into this constraint-based model. The transcriptome is the most commonly used data type in integration, and a large number of FBA (flux balance analysis)-based integrated algorithms have been developed. METHODS AND RESULTS: We mapped the Kcat values to the tree structure of GO terms and found that the Kcat values under the same GO term have a higher similarity. Based on this observation, we developed a new method, called iMTBGO, to predict metabolic flux distributions by constraining reaction bounda-ries based on gene expression ratios normalized by marker genes under the same GO term. We applied this method to previously published data and compared the prediction results with other metabolic flux analysis methods which also utilize gene expression data. The prediction errors of iMTBGO for both growth rates and fluxes in the central metabolic pathways were smaller than those of earlier published methods. CONCLUSION: Considering the fact that reaction rates are not only determined by genes/expression levels, but also by the specific activities of enzymes, the iMTBGO method allows us to make more precise pre-dictions of metabolic fluxes by using expression values normalized based on GO.

10.
BMC Bioinformatics ; 19(1): 325, 2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30217144

ABSTRACT

BACKGROUND: Constraint-based metabolic flux analysis of knockout strategies is an efficient method to simulate the production of useful metabolites in microbes. Owing to the recent development of technologies for artificial DNA synthesis, it may become important in the near future to mathematically design minimum metabolic networks to simulate metabolite production. RESULTS: We have developed a computational method where parsimonious metabolic flux distribution is computed for designated constraints on growth and production rates which are represented by grids. When the growth rate of this obtained parsimonious metabolic network is maximized, higher production rates compared to those noted using existing methods are observed for many target metabolites. The set of reactions used in this parsimonious flux distribution consists of reactions included in the original genome scale model iAF1260. The computational experiments show that the grid size affects the obtained production rates. Under the conditions that the growth rate is maximized and the minimum cases of flux variability analysis are considered, the developed method produced more than 90% of metabolites, while the existing methods produced less than 50%. Mathematical explanations using examples are provided to demonstrate potential reasons for the ability of the proposed algorithm to identify design strategies that the existing methods could not identify. CONCLUSION: We developed an efficient method for computing the design of minimum metabolic networks by using constraint-based flux balance analysis to simulate the production of useful metabolites. The source code is freely available, and is implemented in MATLAB and COBRA toolbox.


Subject(s)
Algorithms , Computational Biology/methods , Carbon/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Genome, Bacterial , Metabolic Flux Analysis , Metabolic Networks and Pathways
11.
J Biol Chem ; 292(24): 10250-10261, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28446608

ABSTRACT

Whereas genomes can be rapidly sequenced, the functions of many genes are incompletely or erroneously annotated because of a lack of experimental evidence or prior functional knowledge in sequence databases. To address this weakness, we describe here a model-enabled gene search (MEGS) approach that (i) identifies metabolic functions either missing from an organism's genome annotation or incorrectly assigned to an ORF by using discrepancies between metabolic model predictions and experimental culturing data; (ii) designs functional selection experiments for these specific metabolic functions; and (iii) selects a candidate gene(s) responsible for these functions from a genomic library and directly interrogates this gene's function experimentally. To discover gene functions, MEGS uses genomic functional selections instead of relying on correlations across large experimental datasets or sequence similarity as do other approaches. When applied to the bioluminescent marine bacterium Vibrio fischeri, MEGS successfully identified five genes that are responsible for four metabolic and transport reactions whose absence from a draft metabolic model of V. fischeri caused inaccurate modeling of high-throughput experimental data. This work demonstrates that MEGS provides a rapid and efficient integrated computational and experimental approach for annotating metabolic genes, including those that have previously been uncharacterized or misannotated.


Subject(s)
Aliivibrio fischeri/genetics , Aquatic Organisms/genetics , Bacterial Proteins/genetics , Expert Systems , Genomics/methods , Models, Genetic , Aliivibrio fischeri/growth & development , Aliivibrio fischeri/metabolism , Animals , Aquaculture , Aquatic Organisms/metabolism , Bacterial Proteins/metabolism , Computer Simulation , Decapodiformes/growth & development , Decapodiformes/microbiology , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/metabolism , Gene Deletion , Genetic Complementation Test , Genomic Library , Hawaii , High-Throughput Nucleotide Sequencing , Molecular Sequence Annotation , Open Reading Frames , Pacific Ocean , Recombinant Proteins/metabolism , Reproducibility of Results , Species Specificity
12.
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
13.
Biomolecules ; 14(5)2024 May 20.
Article in English | MEDLINE | ID: mdl-38786009

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) is a ubiquitous molecule found within all cells, acting as a crucial coenzyme in numerous metabolic reactions. It plays a vital role in energy metabolism, cellular signaling, and DNA repair. Notably, NAD levels decline naturally with age, and this decline is associated with the development of various age-related diseases. Despite this established link, current genome-scale metabolic models, which offer powerful tools for understanding cellular metabolism, do not account for the dynamic changes in NAD concentration. This impedes our understanding of a fluctuating NAD level's impact on cellular metabolism and its contribution to age-related pathologies. To bridge this gap in our knowledge, we have devised a novel method that integrates altered NAD concentration into genome-scale models of human metabolism. This approach allows us to accurately reflect the changes in fatty acid metabolism, glycolysis, and oxidative phosphorylation observed experimentally in an engineered human cell line with a compromised level of subcellular NAD.


Subject(s)
Glycolysis , Models, Biological , NAD , NAD/metabolism , Humans , Oxidative Phosphorylation , Fatty Acids/metabolism , Energy Metabolism
14.
Biomolecules ; 13(3)2023 03 09.
Article in English | MEDLINE | ID: mdl-36979435

ABSTRACT

BACKGROUND: Whole-genome models (GEMs) have become a versatile tool for systems biology, biotechnology, and medicine. GEMs created by automatic and semi-automatic approaches contain a lot of redundant reactions. At the same time, the nonlinearity of the model makes it difficult to evaluate the significance of the reaction for cell growth or metabolite production. METHODS: We propose a new way to apply the global sensitivity analysis (GSA) to GEMs in a straightforward parallelizable fashion. RESULTS: We have shown that Partial Rank Correlation Coefficient (PRCC) captures key steps in the metabolic network despite the network distance from the product synthesis reaction. CONCLUSIONS: FBA-PRCC is a fast, interpretable, and reliable metric to identify the sign and magnitude of the reaction contribution to various cellular functions.


Subject(s)
Genome , Systems Biology , Metabolic Networks and Pathways , Biotechnology , Models, Biological
15.
FEBS Open Bio ; 13(12): 2172-2186, 2023 12.
Article in English | MEDLINE | ID: mdl-37734920

ABSTRACT

Computational systems biology plays a key role in the discovery of suitable antiviral targets. We designed a cell-specific, constraint-based modeling technique for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected lungs. We used the gene sequence of the alpha variant of SARS-CoV-2 to build a viral biomass reaction (VBR). We also used the mass proportion of lipids between the viral biomass and its host cell to estimate the stoichiometric coefficients of viral lipids in the reaction. We then integrated the VBR, the gene expression of the alpha variant of SARS-CoV-2, and the generic human metabolic network Recon3D to reconstruct a cell-specific genome-scale metabolic model. An antiviral target discovery (AVTD) platform was introduced using this model to identify therapeutic drug targets for combating COVID-19. The AVTD platform not only identified antiviral genes for eliminating viral replication but also predicted side effects of treatments. Our computational results revealed that knocking out dihydroorotate dehydrogenase (DHODH) might reduce the synthesis rate of cytidine-5'-triphosphate and uridine-5'-triphosphate, which terminate the viral building blocks of DNA and RNA for SARS-CoV-2 replication. Our results also indicated that DHODH is a promising antiviral target that causes minor side effects, which is consistent with the results of recent reports. Moreover, we discovered that the genes that participate in the de novo biosynthesis of glycerophospholipids and ceramides become unidentifiable if the VBR does not involve the stoichiometry of lipids.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , COVID-19/genetics , Dihydroorotate Dehydrogenase , Antiviral Agents/pharmacology , Lung , Lipids
16.
Methods Mol Biol ; 2513: 271-290, 2022.
Article in English | MEDLINE | ID: mdl-35781211

ABSTRACT

Genome-scale metabolic models (GEMs) provide a useful framework for modeling the metabolism of microorganisms. While the applications of GEMs are wide and far reaching, the reconstruction and continuous curation of such models can be perceived as a tedious and time-consuming task. Using RAVEN, a MATLAB-based toolbox designed to facilitate the reconstruction analysis of metabolic networks, this protocol practically demonstrates how researchers can create their own GEMs using a homology-based approach. To provide a complete example, a draft GEM for the industrially relevant yeast Hansenula polymorpha is reconstructed.


Subject(s)
Metabolic Networks and Pathways , Saccharomycetales , Genome, Fungal , Metabolic Networks and Pathways/genetics , Models, Biological , Saccharomycetales/genetics , Saccharomycetales/metabolism
17.
J Fungi (Basel) ; 8(5)2022 May 20.
Article in English | MEDLINE | ID: mdl-35628779

ABSTRACT

Ustilago maydis is an important plant pathogen that causes corn smut disease and serves as an effective biotechnological production host. The lack of a comprehensive metabolic overview hinders a full understanding of the organism's environmental adaptation and a full use of its metabolic potential. Here, we report the first genome-scale metabolic model (GSMM) of Ustilago maydis (iUma22) for the simulation of metabolic activities. iUma22 was reconstructed from sequencing and annotation using PathwayTools, and the biomass equation was derived from literature values and from the codon composition. The final model contains over 25% annotated genes (6909) in the sequenced genome. Substrate utilization was corrected by BIOLOG phenotype arrays, and exponential batch cultivations were used to test growth predictions. The growth data revealed a decrease in glucose uptake rate with rising glucose concentration. A pangenome of four different U. maydis strains highlighted missing metabolic pathways in iUma22. The new model allows for studies of metabolic adaptations to different environmental niches as well as for biotechnological applications.

18.
Front Bioinform ; 1: 716112, 2021.
Article in English | MEDLINE | ID: mdl-36303731

ABSTRACT

Flux balance analysis (FBA) is a crucial method to analyze large-scale constraint-based metabolic networks and computing design strategies for strain production in metabolic engineering. However, as it is often non-straightforward to obtain such design strategies to produce valuable metabolites, many tools have been proposed based on FBA. Among them, GridProd, which divides the solution space into small squares by focusing on the cell growth rate and the target metabolite production rate to efficiently find the reaction deletion strategies, was extended to CubeProd, which divides the solution space into small cubes. However, as GridProd and CubeProd naively divide the solution space into equal sizes, even places where solutions are unlikely to exist are examined. To address this issue, we introduce dynamic solution space division methods based on CubeProd for faster computing by avoiding searching in places where the solutions do not exist. We applied the proposed method DynCubeProd to iJO1366, which is a genome-scale constraint-based model of Escherichia coli. Compared with CubeProd, DynCubeProd significantly accelerated the calculation of the reaction deletion strategy for each target metabolite production. In addition, under the anaerobic condition of iJO1366, DynCubeProd could obtain the reaction deletion strategies for almost 40% of the target metabolites that the elementary flux vector-based method, which is one of the most effective methods in existence, could not. The developed software is available on https://github.com/Ma-Yier/DynCubeProd.

19.
Front Microbiol ; 11: 596626, 2020.
Article in English | MEDLINE | ID: mdl-33281796

ABSTRACT

Advances in nanopore-based sequencing techniques have enabled rapid characterization of genomes and transcriptomes. An emerging application of this sequencing technology is point-of-care characterization of pathogenic bacteria. However, genome assessments alone are unable to provide a complete understanding of the pathogenic phenotype. Genome-scale metabolic reconstruction and analysis is a bottom-up Systems Biology technique that has elucidated the phenotypic nuances of antimicrobial resistant (AMR) bacteria and other human pathogens. Combining these genome-scale models (GEMs) with point-of-care nanopore sequencing is a promising strategy for combating the emerging health challenge of AMR pathogens. However, the sequencing errors inherent to the nanopore technique may negatively affect the quality, and therefore the utility, of GEMs reconstructed from nanopore assemblies. Here we describe and validate a workflow for rapid construction of GEMs from nanopore (MinION) derived assemblies. Benchmarking the pipeline against a high-quality reference GEM of Escherichia coli K-12 resulted in nanopore-derived models that were >99% complete even at sequencing depths of less than 10× coverage. Applying the pipeline to clinical isolates of pathogenic bacteria resulted in strain-specific GEMs that identified canonical AMR genome content and enabled simulations of strain-specific microbial growth. Additionally, we show that treating the sequencing run as a mock metagenome did not degrade the quality of models derived from metagenome assemblies. Taken together, this study demonstrates that combining nanopore sequencing with GEM construction pipelines enables rapid, in situ characterization of microbial metabolism.

20.
mSystems ; 5(5)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32900868

ABSTRACT

Gene essentiality is altered during polymicrobial infections. Nevertheless, most studies rely on single-species infections to assess pathogen gene essentiality. Here, we use genome-scale metabolic models (GEMs) to explore the effect of coinfection of the diarrheagenic pathogen Vibrio cholerae with another enteric pathogen, enterotoxigenic Escherichia coli (ETEC). Model predictions showed that V. cholerae metabolic capabilities were increased due to ample cross-feeding opportunities enabled by ETEC. This is in line with increased severity of cholera symptoms known to occur in patients with dual infections by the two pathogens. In vitro coculture systems confirmed that V. cholerae growth is enhanced in cocultures relative to single cultures. Further, expression levels of several V. cholerae metabolic genes were significantly perturbed as shown by dual RNA sequencing (RNAseq) analysis of its cocultures with different ETEC strains. A decrease in ETEC growth was also observed, probably mediated by nonmetabolic factors. Single gene essentiality analysis predicted conditionally independent genes that are essential for the pathogen's growth in both single-infection and coinfection scenarios. Our results reveal growth differences that are of relevance to drug targeting and efficiency in polymicrobial infections.IMPORTANCE Most studies proposing new strategies to manage and treat infections have been largely focused on identifying druggable targets that can inhibit a pathogen's growth when it is the single cause of infection. In vivo, however, infections can be caused by multiple species. This is important to take into account when attempting to develop or use current antibacterials since their efficacy can change significantly between single infections and coinfections. In this study, we used genome-scale metabolic models (GEMs) to interrogate the growth capabilities of Vibrio cholerae in single infections and coinfections with enterotoxigenic E. coli (ETEC), which cooccur in a large fraction of diarrheagenic patients. Coinfection model predictions showed that V. cholerae growth capabilities are enhanced in the presence of ETEC relative to V. cholerae single infection, through cross-fed metabolites made available to V. cholerae by ETEC. In vitro, cocultures of the two enteric pathogens further confirmed model predictions showing an increased growth of V. cholerae in coculture relative to V. cholerae single cultures while ETEC growth was suppressed. Dual RNAseq analysis of the cocultures also confirmed that the transcriptome of V. cholerae was distinct during coinfection compared to single-infection scenarios where processes related to metabolism were significantly perturbed. Further, in silico gene-knockout simulations uncovered discrepancies in gene essentiality for V. cholerae growth between single infections and coinfections. Integrative model-guided analysis thus identified druggable targets that would be critical for V. cholerae growth in both single infections and coinfections; thus, designing inhibitors against those targets would provide a broader spectrum of coverage against cholera infections.

SELECTION OF CITATIONS
SEARCH DETAIL