Cargando…

Automatic construction of metabolic models with enzyme constraints

BACKGROUND: In order to improve the accuracy of constraint-based metabolic models, several approaches have been developed which intend to integrate additional biological information. Two of these methods, MOMENT and GECKO, incorporate enzymatic (kcat) parameters and enzyme mass constraints to furthe...

Descripción completa

Detalles Bibliográficos
Autores principales: Bekiaris, Pavlos Stephanos, Klamt, Steffen
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6961255/
https://www.ncbi.nlm.nih.gov/pubmed/31937255
http://dx.doi.org/10.1186/s12859-019-3329-9
_version_ 1783487952036298752
author Bekiaris, Pavlos Stephanos
Klamt, Steffen
author_facet Bekiaris, Pavlos Stephanos
Klamt, Steffen
author_sort Bekiaris, Pavlos Stephanos
collection PubMed
description BACKGROUND: In order to improve the accuracy of constraint-based metabolic models, several approaches have been developed which intend to integrate additional biological information. Two of these methods, MOMENT and GECKO, incorporate enzymatic (kcat) parameters and enzyme mass constraints to further constrain the space of feasible metabolic flux distributions. While both methods have been proven to deliver useful extensions of metabolic models, they may considerably increase size and complexity of the models and there is currently no tool available to fully automate generation and calibration of such enzyme-constrained models from given stoichiometric models. RESULTS: In this work we present three major developments. We first conceived short MOMENT (sMOMENT), a simplified version of the MOMENT approach, which yields the same predictions as MOMENT but requires significantly fewer variables and enables direct inclusion of the relevant enzyme constraints in the standard representation of a constraint-based model. When measurements of enzyme concentrations are available, these can be included as well leading in the extreme case, where all enzyme concentrations are known, to a model representation that is analogous to the GECKO approach. Second, we developed the AutoPACMEN toolbox which allows an almost fully automated creation of sMOMENT-enhanced stoichiometric metabolic models. In particular, this includes the automatic read-out and processing of relevant enzymatic data from different databases and the reconfiguration of the stoichiometric model with embedded enzymatic constraints. Additionally, tools have been developed to adjust (kcat and enzyme pool) parameters of sMOMENT models based on given flux data. We finally applied the new sMOMENT approach and the AutoPACMEN toolbox to generate an enzyme-constrained version of the E. coli genome-scale model iJO1366 and analyze its key properties and differences with the standard model. In particular, we show that the enzyme constraints improve flux predictions (e.g., explaining overflow metabolism and other metabolic switches) and demonstrate, for the first time, that these constraints can markedly change the spectrum of metabolic engineering strategies for different target products. CONCLUSIONS: The methodological and tool developments presented herein pave the way for a simplified and routine construction and analysis of enzyme-constrained metabolic models.
format Online
Article
Text
id pubmed-6961255
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-69612552020-01-17 Automatic construction of metabolic models with enzyme constraints Bekiaris, Pavlos Stephanos Klamt, Steffen BMC Bioinformatics Methodology Article BACKGROUND: In order to improve the accuracy of constraint-based metabolic models, several approaches have been developed which intend to integrate additional biological information. Two of these methods, MOMENT and GECKO, incorporate enzymatic (kcat) parameters and enzyme mass constraints to further constrain the space of feasible metabolic flux distributions. While both methods have been proven to deliver useful extensions of metabolic models, they may considerably increase size and complexity of the models and there is currently no tool available to fully automate generation and calibration of such enzyme-constrained models from given stoichiometric models. RESULTS: In this work we present three major developments. We first conceived short MOMENT (sMOMENT), a simplified version of the MOMENT approach, which yields the same predictions as MOMENT but requires significantly fewer variables and enables direct inclusion of the relevant enzyme constraints in the standard representation of a constraint-based model. When measurements of enzyme concentrations are available, these can be included as well leading in the extreme case, where all enzyme concentrations are known, to a model representation that is analogous to the GECKO approach. Second, we developed the AutoPACMEN toolbox which allows an almost fully automated creation of sMOMENT-enhanced stoichiometric metabolic models. In particular, this includes the automatic read-out and processing of relevant enzymatic data from different databases and the reconfiguration of the stoichiometric model with embedded enzymatic constraints. Additionally, tools have been developed to adjust (kcat and enzyme pool) parameters of sMOMENT models based on given flux data. We finally applied the new sMOMENT approach and the AutoPACMEN toolbox to generate an enzyme-constrained version of the E. coli genome-scale model iJO1366 and analyze its key properties and differences with the standard model. In particular, we show that the enzyme constraints improve flux predictions (e.g., explaining overflow metabolism and other metabolic switches) and demonstrate, for the first time, that these constraints can markedly change the spectrum of metabolic engineering strategies for different target products. CONCLUSIONS: The methodological and tool developments presented herein pave the way for a simplified and routine construction and analysis of enzyme-constrained metabolic models. BioMed Central 2020-01-14 /pmc/articles/PMC6961255/ /pubmed/31937255 http://dx.doi.org/10.1186/s12859-019-3329-9 Text en © The Author(s). 2020 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Methodology Article
Bekiaris, Pavlos Stephanos
Klamt, Steffen
Automatic construction of metabolic models with enzyme constraints
title Automatic construction of metabolic models with enzyme constraints
title_full Automatic construction of metabolic models with enzyme constraints
title_fullStr Automatic construction of metabolic models with enzyme constraints
title_full_unstemmed Automatic construction of metabolic models with enzyme constraints
title_short Automatic construction of metabolic models with enzyme constraints
title_sort automatic construction of metabolic models with enzyme constraints
topic Methodology Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6961255/
https://www.ncbi.nlm.nih.gov/pubmed/31937255
http://dx.doi.org/10.1186/s12859-019-3329-9
work_keys_str_mv AT bekiarispavlosstephanos automaticconstructionofmetabolicmodelswithenzymeconstraints
AT klamtsteffen automaticconstructionofmetabolicmodelswithenzymeconstraints