scholarly journals On the inconsistent treatment of gene-protein-reaction rules in context-specific metabolic models

2019 ◽  
Author(s):  
Miguel Ponce-de-León ◽  
Iñigo Apaolaza ◽  
Alfonso Valencia ◽  
Francisco J. Planes

ABSTRACTWith the publication of high-quality genome-scale metabolic models for several organisms, the Systems Biology community has developed a number of algorithms for their analysis making use of ever growing –omics data. In particular, the reconstruction of the first genome-scale human metabolic model, Recon1, promoted the development of Context-Specific Model (CS-Model) reconstruction methods. This family of algorithms aims to identify the set of metabolic reactions that are active in a cell in a given condition using omics data, such as gene expression levels. Different CS-Model reconstruction algorithms have their own strengths and weaknesses depending on the problem under study and omics data available. However, after careful inspection, we found that all of these algorithms share common issues in the way GPR rules and gene expression data are treated. The first issue is related with how gapfilling reactions are managed after the reconstruction is conducted. The second issue concerns the molecular context, which is used to build the CS-model but neglected for posterior analyses. To evaluate the effect of these issues, we reconstructed ∼400 CS-Models of cancer cell lines and conducted gene essentiality analysis, using CRISPR–Cas9 essentiality data for validation purposes. Altogether, our results illustrate the importance of correcting the errors introduced during the GPR translation in many of the published metabolic reconstructions.

2021 ◽  
Author(s):  
Francisco Zorrilla ◽  
Kiran R. Patil ◽  
Aleksej Zelezniak

AbstractAdvances in genome-resolved metagenomic analysis of complex microbial communities have revealed a large degree of interspecies and intraspecies genetic diversity through the reconstruction of metagenome assembled genomes (MAGs). Yet, metabolic modeling efforts still tend to rely on reference genomes as the starting point for reconstruction and simulation of genome scale metabolic models (GEMs), neglecting the immense intra- and inter-species diversity present in microbial communities. Here we present metaGEM (https://github.com/franciscozorrilla/metaGEM), an end-to-end highly scalable pipeline enabling metabolic modeling of multi-species communities directly from metagenomic samples. The pipeline automates all steps from the extraction of context-specific prokaryotic GEMs from metagenome assembled genomes to community level flux balance simulations. To demonstrate the capabilities of the metaGEM pipeline, we analyzed 483 samples spanning lab culture, human gut, plant associated, soil, and ocean metagenomes, to reconstruct over 14 000 prokaryotic GEMs. We show that GEMs reconstructed from metagenomes have fully represented metabolism comparable to the GEMs reconstructed from reference genomes. We further demonstrate that metagenomic GEMs capture intraspecies metabolic diversity by identifying the differences between pathogenicity levels of type 2 diabetes at the level of gut bacterial metabolic exchanges. Overall, our pipeline enables simulation-ready metabolic model reconstruction directly from individual metagenomes, provides a resource of all reconstructed metabolic models, and showcases community-level modeling of microbiomes associated with disease conditions allowing generation of mechanistic hypotheses.


2018 ◽  
Vol 46 (4) ◽  
pp. 931-936 ◽  
Author(s):  
José P. Faria ◽  
Miguel Rocha ◽  
Isabel Rocha ◽  
Christopher S. Henry

In the era of next-generation sequencing and ubiquitous assembly and binning of metagenomes, new putative genome sequences are being produced from isolate and microbiome samples at ever-increasing rates. Genome-scale metabolic models have enormous utility for supporting the analysis and predictive characterization of these genomes based on sequence data. As a result, tools for rapid automated reconstruction of metabolic models are becoming critically important for supporting the analysis of new genome sequences. Many tools and algorithms have now emerged to support rapid model reconstruction and analysis. Here, we are comparing and contrasting the capabilities and output of a variety of these tools, including ModelSEED, Raven Toolbox, PathwayTools, SuBliMinal Toolbox and merlin.


2021 ◽  
Vol 17 (11) ◽  
pp. e1009589
Author(s):  
Sudharshan Ravi ◽  
Rudiyanto Gunawan

Genome-scale metabolic models (GEMs) provide a powerful framework for simulating the entire set of biochemical reactions in a cell using a constraint-based modeling strategy called flux balance analysis (FBA). FBA relies on an assumed metabolic objective for generating metabolic fluxes using GEMs. But, the most appropriate metabolic objective is not always obvious for a given condition and is likely context-specific, which often complicate the estimation of metabolic flux alterations between conditions. Here, we propose a new method, called ΔFBA (deltaFBA), that integrates differential gene expression data to evaluate directly metabolic flux differences between two conditions. Notably, ΔFBA does not require specifying the cellular objective. Rather, ΔFBA seeks to maximize the consistency and minimize inconsistency between the predicted flux differences and differential gene expression. We showcased the performance of ΔFBA through several case studies involving the prediction of metabolic alterations caused by genetic and environmental perturbations in Escherichia coli and caused by Type-2 diabetes in human muscle. Importantly, in comparison to existing methods, ΔFBA gives a more accurate prediction of flux differences.


2019 ◽  
Author(s):  
Vikash Pandey ◽  
Daniel Hernandez Gardiol ◽  
Anush Chiappino-Pepe ◽  
Vassily Hatzimanikatis

AbstractA large number of genome-scale models of cellular metabolism are available for various organisms. These models include all known metabolic reactions based on the genome annotation. However, the reactions that are active are dependent on the cellular metabolic function or environmental condition. Constraint-based methods that integrate condition-specific transcriptomics data into models have been used extensively to investigate condition-specific metabolism. Here, we present a method (TEX-FBA) for modeling condition-specific metabolism that combines transcriptomics and reaction thermodynamics data to generate a thermodynamically-feasible condition-specific metabolic model. TEX-FBA is an extension of thermodynamic-based flux balance analysis (TFA), which allows the simultaneous integration of different stages of experimental data (e.g., absolute gene expression, metabolite concentrations, thermodynamic data, and fluxomics) and the identification of alternative metabolic states that maximize consistency between gene expression levels and condition-specific reaction fluxes. We applied TEX-FBA to a genome-scale metabolic model ofEscherichia coliby integrating available condition-specific experimental data and found a marked reduction in the flux solution space. Our analysis revealed a marked correlation between actual gene expression profile and experimental flux measurements compared to the one obtained from a randomly generated gene expression profile. We identified additional essential reactions from the membrane lipid and folate metabolism when we integrated transcriptomics data of the given condition on the top of metabolomics and thermodynamics data. These results show TEX-FBA is a promising new approach to study condition-specific metabolism when different types of experimental data are available.Author summaryCells utilize nutrients via biochemical reactions that are controlled by enzymes and synthesize required compounds for their survival and growth. Genome-scale models of metabolism representing these complex reaction networks have been reconstructed for a wide variety of organisms ranging from bacteria to human cells. These models comprise all possible biochemical reactions in a cell, but cells choose only a subset of reactions for their immediate needs and functions. Usually, these models allow for a large flux solution space and one can integrate experimental data in order to reduce it and potentially predict the physiology for a specific condition. We developed a method for integrating different types of omics data, such as fluxomics, transcriptomics, metabolomics into genome-scale metabolic models that reduces the flux solution space. Using gene expression data, the algorithm maximizes the consistency between the predicted and experimental flux for the reactions and predicts biologically relevant flux ranges for the remaining reactions in the network. This method is useful for determining fluxes of metabolic reactions with reduced uncertainty and suitable for performing context- and condition-specific analysis in metabolic models using different types of experimental data.


2021 ◽  
Author(s):  
German Preciat ◽  
Agnieszka B. Wegrzyn ◽  
Ines Thiele ◽  
Thomas Hankemeier ◽  
Ronan MT Fleming

Constraint-based modelling can mechanistically simulate the behaviour of a biochemical system, permitting hypotheses generation, experimental design and interpretation of experimental data, with numerous applications, including modelling of metabolism. Given a generic model, several methods have been developed to extract a context-specific, genome-scale metabolic model by incorporating information used to identify metabolic processes and gene activities in a given context. However, existing model extraction algorithms are unable to ensure that the context-specific model is thermodynamically feasible. This protocol introduces XomicsToModel, a semi-automated pipeline that integrates bibliomic, transcriptomic, proteomic, and metabolomic data with a generic genome-scale metabolic reconstruction, or model, to extract a context-specific, genome-scale metabolic model that is stoichiometrically, thermodynamically and flux consistent. The XomicsToModel pipeline is exemplified for extraction of a specific metabolic model from a generic metabolic model, but it enables multi-omic data integration and extraction of physicochemically consistent mechanistic models from any generic biochemical network. With all input data fully prepared, algorithmic completion of the pipeline takes ~10 min, however manual review of intermediate results may also be required, e.g., when inconsistent input data lead to an infeasible model.


2021 ◽  
Vol 20 (1) ◽  
Author(s):  
Jingru Zhou ◽  
Yingping Zhuang ◽  
Jianye Xia

Abstract Background Genome-scale metabolic model (GSMM) is a powerful tool for the study of cellular metabolic characteristics. With the development of multi-omics measurement techniques in recent years, new methods that integrating multi-omics data into the GSMM show promising effects on the predicted results. It does not only improve the accuracy of phenotype prediction but also enhances the reliability of the model for simulating complex biochemical phenomena, which can promote theoretical breakthroughs for specific gene target identification or better understanding the cell metabolism on the system level. Results Based on the basic GSMM model iHL1210 of Aspergillus niger, we integrated large-scale enzyme kinetics and proteomics data to establish a GSMM based on enzyme constraints, termed a GEM with Enzymatic Constraints using Kinetic and Omics data (GECKO). The results show that enzyme constraints effectively improve the model’s phenotype prediction ability, and extended the model’s potential to guide target gene identification through predicting metabolic phenotype changes of A. niger by simulating gene knockout. In addition, enzyme constraints significantly reduced the solution space of the model, i.e., flux variability over 40.10% metabolic reactions were significantly reduced. The new model showed also versatility in other aspects, like estimating large-scale $$k_{{cat}}$$ k cat values, predicting the differential expression of enzymes under different growth conditions. Conclusions This study shows that incorporating enzymes’ abundance information into GSMM is very effective for improving model performance with A. niger. Enzyme-constrained model can be used as a powerful tool for predicting the metabolic phenotype of A. niger by incorporating proteome data. In the foreseeable future, with the fast development of measurement techniques, and more precise and rich proteomics quantitative data being obtained for A. niger, the enzyme-constrained GSMM model will show greater application space on the system level.


2020 ◽  
Author(s):  
Bonnie V. Dougherty ◽  
Kristopher D. Rawls ◽  
Glynis L. Kolling ◽  
Kalyan C. Vinnakota ◽  
Anders Wallqvist ◽  
...  

SummaryThe heart is a metabolic omnivore, known to consume many different carbon substrates in order to maintain function. In diseased states, the heart’s metabolism can shift between different carbon substrates; however, there is some disagreement in the field as to the metabolic shifts seen in end-stage heart failure and whether all heart failure converges to a common metabolic phenotype. Here, we present a new, validated cardiomyocyte-specific GEnome-scale metabolic Network REconstruction (GENRE), iCardio, and use the model to identify common shifts in metabolic functions across heart failure omics datasets. We demonstrate the utility of iCardio in interpreting heart failure gene expression data by identifying Tasks Inferred from Differential Expression (TIDEs) which represent metabolic functions associated with changes in gene expression. We identify decreased NO and Neu5Ac synthesis as common metabolic markers of heart failure across datasets. Further, we highlight the differences in metabolic functions seen across studies, further highlighting the complexity of heart failure. The methods presented for constructing a tissue-specific model and identifying TIDEs can be extended to multiple tissue and diseases of interest.


2019 ◽  
Author(s):  
Chintan J. Joshi ◽  
Song-Min Schinn ◽  
Anne Richelle ◽  
Isaac Shamie ◽  
Eyleen J. O’Rourke ◽  
...  

AbstractDiverse algorithms can integrate transcriptomics with genome-scale metabolic models (GEMs) to build context-specific metabolic models. These algorithms require identification of a list of high confidence (core) reactions from transcriptomics, but parameters related to identification of core reactions, such as thresholding of expression profiles, can significantly change model content. Importantly, current thresholding approaches are burdened with setting singular arbitrary thresholds for all genes; thus, resulting in removal of enzymes needed in small amounts and even many housekeeping genes. Here, we describe StanDep, a novel heuristic method for using transcriptomics to identify core reactions prior to building context-specific metabolic models. StanDep clusters gene expression data based on their expression pattern across different contexts and determines thresholds for each cluster using data-dependent statistics, specifically standard deviation and mean. To demonstrate the use of StanDep, we built hundreds of models for the NCI-60 cancer cell lines. These models successfully increased the inclusion of housekeeping reactions, which are often lost in models built using standard thresholding approaches. Further, StanDep also provided a transcriptomic explanation for inclusion of lowly expressed reactions that were otherwise only supported by model extraction methods. Our study also provides novel insights into how cells may deal with context-specific and ubiquitous functions. StanDep, as a MATLAB toolbox, is available at https://github.com/LewisLabUCSD/StanDep


Sign in / Sign up

Export Citation Format

Share Document