Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol Update
  • Published:

Creation and analysis of biochemical constraint-based models using the COBRA Toolbox v.3.0

Abstract

Constraint-based reconstruction and analysis (COBRA) provides a molecular mechanistic framework for integrative analysis of experimental molecular systems biology data and quantitative prediction of physicochemically and biochemically feasible phenotypic states. The COBRA Toolbox is a comprehensive desktop software suite of interoperable COBRA methods. It has found widespread application in biology, biomedicine, and biotechnology because its functions can be flexibly combined to implement tailored COBRA protocols for any biochemical network. This protocol is an update to the COBRA Toolbox v.1.0 and v.2.0. Version 3.0 includes new methods for quality-controlled reconstruction, modeling, topological analysis, strain and experimental design, and network visualization, as well as network integration of chemoinformatic, metabolomic, transcriptomic, proteomic, and thermochemical data. New multi-lingual code integration also enables an expansion in COBRA application scope via high-precision, high-performance, and nonlinear numerical optimization solvers for multi-scale, multi-cellular, and reaction kinetic modeling, respectively. This protocol provides an overview of all these new features and can be adapted to generate and analyze constraint-based models in a wide variety of scenarios. The COBRA Toolbox v.3.0 provides an unparalleled depth of COBRA methods.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of key constraint-based reconstruction and analysis concepts.
Fig. 2: Continuous integration of newly developed code is performed on a dedicated server running Jenkins.
Fig. 3: Unsteady-state flux balance analysis.
Fig. 4: Solution spaces from steady-state fluxes are anisotropic, that is, long in some directions and short in others.
Fig. 5: In the OptForce procedure, the MUST sets are determined by contrasting the flux ranges obtained using flux variability analysis (FVA) of a wild-type (blue bars) and an overproducing strain (red bars).
Fig. 6: Development branching model of the COBRA Toolbox.
Fig. 7
Fig. 8: An energy-generating stoichiometrically balanced cycle.
Fig. 9: The interventions predicted by the OptForce method for succinate overproduction in E. coli (AntCore model) under aerobic conditions.
Fig. 10: Qualitatively forward, quantitatively reverse reactions in a multi-compartmental, genome-scale model.
Fig. 11: Human metabolic network visualization.
Fig. 12: Selective scope visualization of the E. coli core model by Paint4Net.

Similar content being viewed by others

References

  1. Palsson, B. Ø. Systems Biology: Constraint-Based Reconstruction and Analysis (Cambridge University Press, Cambridge, 2015).

    Book  Google Scholar 

  2. O’Brien, E. J., Monk, J. M. & Palsson, B. O. Using genome-scale models to predict biological capabilities. Cell 161, 971–987 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Becker, S. A. et al. Quantitative prediction of cellular metabolism with constraint-based models: the COBRA Toolbox. Nat. Protoc. 2, 727–738 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Schellenberger, J. et al. Quantitative prediction of cellular metabolism with constraint-based models: the COBRA Toolbox v2.0. Nat. Protoc. 6, 1290–1307 (2011).

  5. Lewis, N. E., Nagarajan, H. & Palsson, B. O. Constraining the metabolic genotype–phenotype relationship using a phylogeny of in silico methods. Nat. Rev. Microbiol. 10, 291–305 (2012).

  6. Thiele, I. & Palsson, B. Ø. A protocol for generating a high-quality genome-scale metabolic reconstruction. Nat. Protoc. 5, 93–121 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kitano, H., Ghosh, S. & Matsuoka, Y. Social engineering for virtual ‘big science’ in systems biology. Nat. Chem. Biol. 7, 323–326 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Bordbar, A., Monk, J. M., King, Z. A. & Palsson, B. O. Constraint-based models predict metabolic and associated cellular functions. Nat. Rev. Genet. 15, 107–120 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Maia, P., Rocha, M. & Rocha, I. In silico constraint-based strain optimization methods: the quest for optimal cell factories. Microbiol. Mol. Biol. Rev. 80, 45–67 (2016).

    Article  PubMed  Google Scholar 

  10. Hefzi, H. et al. A consensus genome-scale reconstruction of Chinese hamster ovary cell metabolism. Cell Syst. 3, 434–443.e8 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Yusufi, F. N. K. et al. Mammalian systems biotechnology reveals global cellular adaptations in a recombinant CHO cell line. Cell Syst. 4, 530–542.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Zhuang, K. et al. Genome-scale dynamic modeling of the competition between Rhodoferax and Geobacter in anoxic subsurface environments. ISME J 5, 305–316 (2011).

    Article  PubMed  Google Scholar 

  13. Jamshidi, N. & Palsson, B. Ø. Systems biology of the human red blood cell. Blood Cells Mol. Dis. 36, 239–247 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Yizhak, K., Gabay, O., Cohen, H. & Ruppin, E. Model-based identification of drug targets that revert disrupted metabolism and its application to ageing. Nat. Commun. 4, 2632 (2013).

    Article  PubMed  CAS  Google Scholar 

  15. Shlomi, T., Cabili, M. N. & Ruppin, E. Predicting metabolic biomarkers of human inborn errors of metabolism. Mol. Syst. Biol. 5, 263 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Sahoo, S., Franzson, L., Jonsson, J. J. & Thiele, I. A compendium of inborn errors of metabolism mapped onto the human metabolic network. Mol. Biosyst. 8, 2545–2558 (2012).

    Article  CAS  PubMed  Google Scholar 

  17. Thiele, I. et al. A community-driven global reconstruction of human metabolism. Nat. Biotechnol. 31, 419–425 (2013).

    Article  CAS  PubMed  Google Scholar 

  18. Pagliarini, R. & di Bernardo, D. A genome-scale modeling approach to study inborn errors of liver metabolism: toward an in silico patient. J. Comput. Biol. 20, 383–397 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Shaked, I., Oberhardt, M. A., Atias, N., Sharan, R. & Ruppin, E. Metabolic network prediction of drug side effects. Cell Syst. 2, 209–213 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Chang, R. L., Xie, L., Xie, L., Bourne, P. E. & Palsson, B. Drug off-target effects predicted using structural analysis in the context of a metabolic network model. PLoS Comput. Biol. 6, e1000938 (2010).

  21. Kell, D. B. Systems biology, metabolic modelling and metabolomics in drug discovery and development. Drug Discov. Today 11, 1085–1092 (2006).

    Article  CAS  PubMed  Google Scholar 

  22. Duarte, N. C. et al. Global reconstruction of the human metabolic network based on genomic and bibliomic data. Proc. Natl. Acad. Sci. USA 104, 1777–1782 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Swainston, N. et al. Recon 2.2: from reconstruction to model of human metabolism. Metabolomics 12, 109 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Pornputtapong, N., Nookaew, I. & Nielsen, J. Human metabolic atlas: an online resource for human metabolism. Database 2015, bav068 (2015).

  25. Zielinski, D. C. et al. Systems biology analysis of drivers underlying hallmarks of cancer cell metabolism. Sci. Rep. 7, 41241 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mardinoglu, A. et al. Genome-scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non-alcoholic fatty liver disease. Nat. Commun. 5, 3083 (2014).

    Article  PubMed  CAS  Google Scholar 

  27. Karlstädt, A. et al. CardioNet: a human metabolic network suited for the study of cardiomyocyte metabolism. BMC Syst. Biol. 6, 114 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Gille, C. et al. HepatoNet1: a comprehensive metabolic reconstruction of the human hepatocyte for the analysis of liver physiology. Mol. Syst. Biol. 6, 411 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Martins Conde Pdo, R., Sauter, T. & Pfau, T. Constraint based modeling going multicellular. Front. Mol. Biosci. 3, 3 (2016).

  30. Bordbar, A. et al. A multi-tissue type genome-scale metabolic network for analysis of whole-body systems physiology. BMC Syst. Biol. 5, 180 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Yizhak, K. et al. Phenotype-based cell-specific metabolic modeling reveals metabolic liabilities of cancer. Elife 3, e03641 (2014).

    Article  PubMed Central  Google Scholar 

  32. Mardinoglu, A. et al. Integration of clinical data with a genome-scale metabolic model of the human adipocyte. Mol. Syst. Biol. 9, 649 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Bordbar, A. et al. Personalized whole-cell kinetic models of metabolism for discovery in genomics and pharmacodynamics. Cell Syst. 1, 283–292 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Shoaie, S. et al. Quantifying diet-induced metabolic changes of the human gut microbiome. Cell Metab. 22, 320–331 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Nogiec, C. D. & Kasif, S. To supplement or not to supplement: a metabolic network framework for human nutritional supplements. PLoS ONE 8, e68751 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Heinken, A., Sahoo, S., Fleming, R. M. T. & Thiele, I. Systems-level characterization of a host-microbe metabolic symbiosis in the mammalian gut. Gut Microbes 4, 28–40 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Heinken, A. et al. Functional metabolic map of Faecalibacterium prausnitzii, a beneficial human gut microbe. J. Bacteriol. 196, 3289–3302 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Magnúsdóttir, S. et al. Generation of genome-scale metabolic reconstructions for 773 members of the human gut microbiota. Nat. Biotechnol. 35, 81–89 (2017).

    Article  PubMed  CAS  Google Scholar 

  39. Lakshmanan, M., Koh, G., Chung, B. K. S. & Lee, D.-Y. Software applications for flux balance analysis. Brief Bioinform. 15, 108–122 (2014).

    Article  PubMed  Google Scholar 

  40. Ebrahim, A., Lerman, J. A., Palsson, B. O. & Hyduke, D. R. COBRApy: constraints-based reconstruction and analysis for Python. BMC Syst. Biol. 7, 74 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Arkin, A. P. et al. The United States Department of Energy Systems Biology Knowledgebase. Nat. Biotechnol. 36, 566–569 (2018).

  42. Heirendt, L., Thiele, I. & Fleming, R. M. T. DistributedFBA.jl: high-level, high-performance flux balance analysis in Julia. Bioinformatics 33, 1421–1423 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Latendresse, M., Krummenacker, M., Trupp, M. & Karp, P. D. Construction and completion of flux balance models from pathway databases. Bioinformatics 28, 388–396 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Karp, P. D. et al. Pathway Tools version 19.0 update: software for pathway/genome informatics and systems biology. Brief Bioinform. 17, 877–890 (2016).

    Article  CAS  PubMed  Google Scholar 

  45. Sandve, G. K., Nekrutenko, A., Taylor, J. & Hovig, E. Ten simple rules for reproducible computational research. PLoS Comput. Biol. 9, e1003285 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Ince, D. C., Hatton, L. & Graham-Cumming, J. The case for open computer programs. Nature 482, 485–488 (2012).

    Article  CAS  PubMed  Google Scholar 

  47. Gevorgyan, A., Bushell, M. E., Avignone-Rossa, C. & Kierzek, A. M. SurreyFBA: a command line tool and graphics user interface for constraint-based modeling of genome-scale metabolic reaction networks. Bioinformatics 27, 433–434 (2011).

    Article  CAS  PubMed  Google Scholar 

  48. Thorleifsson, S. G. & Thiele, I. rBioNet: a COBRA toolbox extension for reconstructing high-quality biochemical networks. Bioinformatics 27, 2009–2010 (2011).

  49. Sauls, J. T. & Buescher, J. M. Assimilating genome-scale metabolic reconstructions with modelBorgifier. Bioinformatics 30, 1036–1038 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Noronha, A. et al. ReconMap: an interactive visualization of human metabolism. Bioinformatics 33, 605–607 (2017).

    CAS  PubMed  Google Scholar 

  51. Gawron, P. et al. MINERVA—a platform for visualization and curation of molecular interaction networks. npj Syst. Biol. Appl. 2, 16020 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Olivier, B. G., Rohwer, J. M. & Hofmeyr, J.-H. S. Modelling cellular systems with PySCeS. Bioinformatics 21, 560–561 (2005).

    Article  CAS  PubMed  Google Scholar 

  53. Gelius-Dietrich, G., Desouki, A. A., Fritzemeier, C. J. & Lercher, M. J. Sybil—efficient constraint-based modelling in R. BMC Syst. Biol. 7, 125 (2013).

  54. Ma, D. et al. Reliable and efficient solution of genome-scale models of metabolism and macromolecular expression. Sci. Rep. 7, 40863 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Klamt, S., Saez-Rodriguez, J. & Gilles, E. D. Structural and functional analysis of cellular networks with CellNetAnalyzer. BMC Syst. Biol. 1, 2 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Klamt, S. & von Kamp, A. An application programming interface for CellNetAnalyzer. Biosystems 105, 162–168 (2011).

    Article  CAS  PubMed  Google Scholar 

  57. Apaolaza, I. et al. An in-silico approach to predict and exploit synthetic lethality in cancer metabolism. Nat. Commun. 8, 459 (2017).

  58. Maranas, C. D. & Zomorrodi, A. R. Optimization Methods in Metabolic Networks (Wiley, New York, 2016).

    Book  Google Scholar 

  59. Chowdhury, A., Zomorrodi, A. R. & Maranas, C. D. Bilevel optimization techniques in computational strain design. Comp. Chem. Eng. 72, 363–372 (2015).

    Article  CAS  Google Scholar 

  60. Thiele, I. et al. Multiscale modeling of metabolism and macromolecular synthesis in E. coli and its application to the evolution of codon usage. PLoS ONE 7, e45635 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Feist, A. M. et al. A genome-scale metabolic reconstruction for Escherichia coli K-12 MG1655 that accounts for 1260 ORFs and thermodynamic information. Mol. Syst. Biol. 3, 121 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Thiele, I., Jamshidi, N., Fleming, R. M. T. & Palsson, B. Ø. Genome-scale reconstruction of Escherichia coli’s transcriptional and translational machinery: a knowledge base, its mathematical formulation, and its functional characterization. PLoS Comput. Biol. 5, e1000312 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Yang, L. et al. Systems biology definition of the core proteome of metabolism and expression is consistent with high-throughput data. Proc. Natl. Acad. Sci. USA 112, 10810–10815 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Bornstein, B. J., Keating, S. M., Jouraku, A. & Hucka, M. LibSBML: an API library for SBML. Bioinformatics 24, 880–881 (2008).

    Article  CAS  PubMed  Google Scholar 

  65. Aurich, M. K., Fleming, R. M. T. & Thiele, I. MetaboTools: a comprehensive toolbox for analysis of genome-scale metabolic models. Front. Physiol. 7, 327 (2016).

  66. Brunk, E. et al. Recon 3D: a resource enabling a three-dimensional view of gene variation in human metabolism. Nat. Biotechnol. 36, 272–281 (2018).

  67. Ma, D. & Saunders, M. A. Solving multiscale linear programs using the simplex method in quadruple precision. in Numerical Analysis and Optimization, Vol. 134 (eds. Al-Baali, M., Grandinetti, L. & Purnama, A.) 223–235 (Springer International Publishing, Cham, Switzerland, 2015).

  68. Fleming, R. M. T. & Thiele, I. Mass conserved elementary kinetics is sufficient for the existence of a non-equilibrium steady state concentration. J. Theor. Biol. 314, 173–181 (2012).

  69. Gevorgyan, A., Poolman, M. G. & Fell, D. A. Detection of stoichiometric inconsistencies in biomolecular models. Bioinformatics 24, 2245–2251 (2008).

    Article  CAS  PubMed  Google Scholar 

  70. Orth, J. D., Thiele, I. & Palsson, B. Ø. What is flux balance analysis? Nat. Biotechnol. 28, 245–248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Feist, A. M. & Palsson, B. O. The biomass objective function. Curr. Opin. Microbiol. 13, 344–349 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Meléndez-Hevia, E. & Isidoro, A. The game of the pentose phosphate cycle. J. Theor. Biol. 117, 251–263 (1985).

    Article  PubMed  Google Scholar 

  73. Orth, J. D. & Palsson, B. Ø. Systematizing the generation of missing metabolic knowledge. Biotechnol. Bioeng. 107, 403–412 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yamada, T. et al. Prediction and identification of sequences coding for orphan enzymes using genomic and metagenomic neighbours. Mol. Syst. Biol. 8, 581 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Liberal, R. & Pinney, J. W. Simple topological properties predict functional misannotations in a metabolic network. Bioinformatics 29, i154–i161 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Reed, J. L. et al. Systems approach to refining genome annotation. Proc. Natl. Acad. Sci. USA 103, 17480–17484 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Orth, J. D. & Palsson, B. Gap-filling analysis of the iJO1366 Escherichia coli metabolic network reconstruction for discovery of metabolic functions. BMC Syst. Biol. 6, 30 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Chang, R. L. et al. Metabolic network reconstruction of Chlamydomonas offers insight into light-driven algal metabolism. Mol. Syst. Biol. 7, 518 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Rolfsson, O., Palsson, B. Ø. & Thiele, I. The human metabolic reconstruction Recon 1 directs hypotheses of novel human metabolic functions. BMC Syst. Biol. 5, 155 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Rolfsson, Ó., Paglia, G., Magnusdóttir, M., Palsson, B. Ø. & Thiele, I. Inferring the metabolism of human orphan metabolites from their metabolic network context affirms human gluconokinase activity. Biochem. J. 449, 427–435 (2013).

    Article  CAS  PubMed  Google Scholar 

  81. Satish Kumar, V., Dasika, M. S. & Maranas, C. D. Optimization based automated curation of metabolic reconstructions. BMC Bioinformatics 8, 212 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Thiele, I., Vlassis, N. & Fleming, R. M. T. fastGapFill: efficient gap filling in metabolic networks. Bioinformatics 30, 2529–2531 (2014).

  83. Willemsen, A. M. et al. MetDFBA: incorporating time-resolved metabolomics measurements into dynamic flux balance analysis. Mol. Biosyst. 11, 137–145 (2014).

    Article  PubMed  CAS  Google Scholar 

  84. Kleessen, S., Irgang, S., Klie, S., Giavalisco, P. & Nikoloski, Z. Integration of transcriptomics and metabolomics data specifies the metabolic response of Chlamydomonas to rapamycin treatment. Plant J. 81, 822–835 (2015).

    Article  CAS  PubMed  Google Scholar 

  85. Bordbar, A. et al. Elucidating dynamic metabolic physiology through network integration of quantitative time-course metabolomics. Sci. Rep. 7, 46249 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  86. Blazier, A. S. & Papin, J. A. Integration of expression data in genome-scale metabolic network reconstructions. Front. Physiol. 3, 299 (2012).

  87. Opdam, S. et al. A systematic evaluation of methods for tailoring genome-scale metabolic models. Cell Syst. 4, 318–329.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Estévez, S. R. & Nikoloski, Z. Generalized framework for context-specific metabolic model extraction methods. Front. Plant Sci. 5, 491 (2014).

  89. Vlassis, N., Pacheco, M. P. & Sauter, T. Fast reconstruction of compact context-specific metabolic network models. PLoS Comput. Biol. 10, e1003424 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  90. Becker, S. A. & Palsson, B. O. Context-specific metabolic networks are consistent with experiments. PLoS Comput. Biol. 4, e1000082 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  91. Zur, H., Ruppin, E. & Shlomi, T. iMAT: an integrative metabolic analysis tool. Bioinformatics 26, 3140–3142 (2010).

    Article  CAS  PubMed  Google Scholar 

  92. Agren, R. et al. Reconstruction of genome-scale active metabolic networks for 69 human cell types and 16 cancer types using INIT. PLoS Comp. Biol. 8, e1002518 (2012).

    Article  CAS  Google Scholar 

  93. Jerby, L., Shlomi, T. & Ruppin, E. Computational reconstruction of tissue-specific metabolic models: application to human liver metabolism. Mol. Syst. Biol. 6, 401 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  94. Wang, Y., Eddy, J. A. & Price, N. D. Reconstruction of genome-scale metabolic models for 126 human tissues using mCADRE. BMC Syst. Biol. 6, 153 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Kuhar, M. J. On the use of protein turnover and half-lives. Neuropsychopharmacology 34, 1172–1173 (2008).

    Article  PubMed  CAS  Google Scholar 

  96. Lajtha, A. & Sylvester, V. Handbook of Neurochemistry and Molecular Neurobiology (Springer, Boston, 2008).

  97. Schuster, S. & Hilgetag, C. On elementary flux modes in biochemical reaction systems at steady state. J. Biol. Syst. 02, 165–182 (1994).

    Article  Google Scholar 

  98. Schilling, C. H., Letscher, D. & Palsson, B. Ø. Theory for the systemic definition of metabolic pathways and their use in interpreting metabolic function from a pathway-oriented perspective. J. Theor. Biol. 203, 229–248 (2000).

    Article  CAS  PubMed  Google Scholar 

  99. Klamt, S. et al. From elementary flux modes to elementary flux vectors: metabolic pathway analysis with arbitrary linear flux constraints. PLoS Comput. Biol. 13, e1005409 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Bordbar, A. et al. Minimal metabolic pathway structure is consistent with associated biomolecular interactions. Mol. Syst. Biol. 10, 737 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Gudmundsson, S. & Thiele, I. Computationally efficient flux variability analysis. BMC Bioinformatics 11, 489 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  102. Haraldsdóttir, H. S., Cousins, B., Thiele, I., Fleming, R. M. T. & Vempala, S. CHRR: coordinate hit-and-run with rounding for uniform sampling of constraint-based models. Bioinformatics 33, 1741–1743 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Cousins, B. & Vempala, S. Gaussian cooling and algorithms for volume and Gaussian volume. SIAM J. Comput. 47, 1237–1273 (2018).

  104. Cousins, B. & Vempala, S. A practical volume algorithm. Math. Prog. Comp. 8, 1–28 (2015).

    Google Scholar 

  105. Burgard, A. P., Pharkya, P. & Maranas, C. D. Optknock: a bilevel programming framework for identifying gene knockout strategies for microbial strain optimization. Biotechnol. Bioeng. 84, 647–657 (2003).

    Article  CAS  PubMed  Google Scholar 

  106. Patil, K. R., Rocha, I., Förster, J. & Nielsen, J. Evolutionary programming as a platform for in silico metabolic engineering. BMC Bioinformatics 6, 308 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  107. Lun, D. S. et al. Large-scale identification of genetic design strategies using local search. Mol. Syst. Biol. 5, 296 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Ranganathan, S., Suthers, P. F. & Maranas, C. D. OptForce: an optimization procedure for identifying all genetic manipulations leading to targeted overproductions. PLoS Comput. Biol. 6, e1000744 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Antoniewicz, M. R. et al. Metabolic flux analysis in a nonstationary system: fed-batch fermentation of a high yielding strain of E. coli producing 1,3-propanediol. Metab. Eng. 9, 277–292 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Haraldsdóttir, H. S., Thiele, I. & Fleming, R. M. T. Comparative evaluation of open source software for mapping between metabolite identifiers in metabolic network reconstructions: application to Recon 2. J. Cheminform. 6, 2 (2014).

  111. Preciat Gonzalez, G. A. et al. Comparative evaluation of atom mapping algorithms for balanced metabolic reactions: application to Recon 3D. J. Cheminform. 9, 39 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Kim, S. et al. PubChem substance and compound databases. Nucleic Acids Res. 44, D1202–D1213 (2016).

    Article  CAS  PubMed  Google Scholar 

  113. Kanehisa, M. & Goto, S. KEGG: Kyoto Encyclopedia of Genes and Genomes. Nucleic Acids Res. 28, 27–30 (2000).

  114. Hastings, J. et al. The ChEBI reference database and ontology for biologically relevant chemistry: enhancements for 2013. Nucleic Acids Res. 41, D456–D463 (2013).

    Article  CAS  PubMed  Google Scholar 

  115. Sud, M. et al. LMSD: LIPID MAPS structure database. Nucleic Acids Res. 35, D527–D532 (2007).

    Article  CAS  PubMed  Google Scholar 

  116. Forster, M., Pick, A., Raitner, M., Schreiber, F. & Brandenburg, F. J. The system architecture of the BioPath system. In Silico Biol. 2, 415–426 (2002).

    CAS  PubMed  Google Scholar 

  117. Williams, A. J., Tkachenko, V., Golotvin, S., Kidd, R. & McCann, G. ChemSpider—building a foundation for the semantic web by hosting a crowd sourced databasing platform for chemistry. J. Cheminform. 2, O16 (2010).

  118. Wishart, D. S. et al. HMDB: the Human Metabolome Database. Nucleic Acids Res. 35, D521–D526 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Rahman, S. A. et al. Reaction Decoder Tool (RDT): extracting features from chemical reactions. Bioinformatics 32, 2065–2066 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Kumar, A. & Maranas, C. D. CLCA: maximum common molecular substructure queries within the MetRxn Database. J. Chem. Inf. Model. 54, 3417–3438 (2014).

    Article  CAS  PubMed  Google Scholar 

  121. Shimizu, Y., Hattori, M., Goto, S. & Kanehisa, M. Generalized reaction patterns for prediction of unknown enzymatic reactions. Genome Inform. 20, 149–158 (2008).

    CAS  PubMed  Google Scholar 

  122. Haraldsdóttir, H. S. & Fleming, R. M. T. Identification of conserved moieties in metabolic networks by graph theoretical analysis of atom transition networks. PLoS Comput. Biol. 12, e1004999 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  123. Klamt, S., Haus, U.-U. & Theis, F. Hypergraphs and cellular networks. PLoS Comput. Biol. 5, e1000385 (2009).

  124. Fleming, R. M. T. & Thiele, I. von Bertalanffy 1.0: a COBRA toolbox extension to thermodynamically constrain metabolic models. Bioinformatics 27, 142–143 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Fleming, R. M. T., Thiele, I. & Nasheuer, H. P. Quantitative assignment of reaction directionality in constraint-based models of metabolism: application to Escherichia coli. Biophys. Chem. 145, 47–56 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Haraldsdóttir, H. S., Thiele, I. & Fleming, R. M. T. Quantitative assignment of reaction directionality in a multicompartmental human metabolic reconstruction. Biophys. J. 102, 1703–1711 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  127. Noor, E., Haraldsdóttir, H. S., Milo, R. & Fleming, R. M. T. Consistent estimation of Gibbs energy using component contributions. PLoS Comput. Biol. 9, e1003098 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Fleming, R. M. T., Maes, C. M., Saunders, M. A., Ye, Y. & Palsson, B. Ø. A variational principle for computing nonequilibrium fluxes and potentials in genome-scale biochemical networks. J. Theor. Biol. 292, 71–77 (2012).

    Article  CAS  PubMed  Google Scholar 

  129. Beard, D. A., Liang, S.-D. & Qian, H. Energy balance for analysis of complex metabolic networks. Biophys. J. 83, 79–86 (2002).

  130. Qian, H. & Beard, D. A. Thermodynamics of stoichiometric biochemical networks in living systems far from equilibrium. Biophys. Chem. 114, 213–220 (2005).

    Article  CAS  PubMed  Google Scholar 

  131. Fleming, R. M. T., Thiele, I., Provan, G. & Nasheuer, H. P. Integrated stoichiometric, thermo- dynamic and kinetic modelling of steady state metabolism. J. Theor. Biol. 264, 683–692 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Schellenberger, J., Lewis, N. E. & Palsson, B. Ø. Elimination of thermodynamically infeasible loops in steady-state metabolic models. Biophys. J. 100, 544–553 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Soh, K. C. & Hatzimanikatis, V. Network thermodynamics in the post-genomic era. Curr. Opin. Microbiol. 13, 350–357 (2010).

    Article  CAS  PubMed  Google Scholar 

  134. Fleming, R. M. T., Vlassis, N., Thiele, I. & Saunders, M. A. Conditions for duality between fluxes and concentrations in biochemical networks. J. Theor. Biol. 409, 1–10 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. ​Aragón Artacho, F.J., Fleming, R. M. T. & Vuong, P. T. Accelerating the DC algorithm for smooth functions. Math. Program. 169, 95–118 (2018).

  136. Artacho, F. J. A. & Fleming, R. M. T. Globally convergent algorithms for finding zeros of duplomonotone mappings. Optim. Lett. 9, 1–16 (2014).

    Google Scholar 

  137. Ahookhosh, M., Aragón, F. J., Fleming, R. M. T. & Vuong, P. T. Local convergence of Levenberg-Marquardt methods under Hölder metric subregularity. Preprint at https://arxiv.org/abs/1703.07461 (2017).

  138. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. King, Z. A. et al. Escher: a web application for building, sharing, and embedding data-rich visualizations of biological pathways. PLoS Comput. Biol. 11, e1004321 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  140. Kuperstein, I. et al. NaviCell: a web-based environment for navigation, curation and maintenance of large molecular interaction maps. BMC Syst. Biol. 7, 100 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  141. Kostromins, A. & Stalidzans, E. Paint4net: COBRA Toolbox extension for visualization of stoichiometric models of metabolism. Biosystems 109, 233–239 (2012).

    Article  CAS  PubMed  Google Scholar 

  142. Aurich, M. K. et al. Prediction of intracellular metabolic states from extracellular metabolomic data. Metabolomics 11, 603–619 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Guebila, M. B. & Thiele, I. Model-based dietary optimization for late-stage, levodopa-treated, Parkinson’s disease patients. npj Syst. Biol. Appl. 2, 16013 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  144. Sun, Y., Fleming, R. M. T., Thiele, I. & Saunders, M. A. Robust flux balance analysis of multiscale biochemical reaction networks. BMC Bioinformatics 14, 240 (2013).

  145. Lewis, N. E. et al. Omic data from evolved E. coli are consistent with computed optimal growth from genome-scale models. Mol. Syst. Biol. 6, 390 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  146. Thiele, I., Fleming, R. M. T., Bordbar, A., Schellenberger, J. & Palsson, B. Ø. Functional characterization of alternate optimal solutions of Escherichia coli’s transcriptional and translational machinery. Biophys. J. 98, 2072–2081 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Ballerstein, K., von Kamp, A., Klamt, S. & Haus, U.-U. Minimal cut sets in a metabolic network are elementary modes in a dual network. Bioinformatics 28, 381–387 (2012).

    Article  CAS  PubMed  Google Scholar 

  148. von Kamp, A. & Klamt, S. Enumeration of smallest intervention strategies in genome-scale metabolic networks. PLoS Comput. Biol. 10, e1003378 (2014).

    Article  CAS  Google Scholar 

  149. Fujita, K. A. et al. Integrating pathways of Parkinson’s disease in a molecular interaction map. Mol. Neurobiol. 49, 88–102 (2014).

    Article  CAS  PubMed  Google Scholar 

  150. Agren, R. et al. The RAVEN Toolbox and its use for generating a genome-scale metabolic model for Penicillium chrysogenum. PLoS Comput. Biol. 9, e1002980 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Grafahrend-Belau, E., Klukas, C., Junker, B. H. & Schreiber, F. FBA-SimVis: interactive visualization of constraint-based metabolic models. Bioinformatics 25, 2755–2757 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Rocha, I. et al. OptFlux: an open-source software platform for in silico metabolic engineering. BMC Syst. Biol. 4, 45 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  153. Poolman, M. G. ScrumPy: metabolic modelling with Python. Syst. Biol. 153, 375–378 (2006).

  154. Hoppe, A., Hoffmann, S., Gerasch, A., Gille, C. & Holzhütter, H.-G. FASIMU: flexible software for flux-balance computation series in large metabolic networks. BMC Bioinformatics 12, 28 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  155. Boele, J., Olivier, B. G. & Teusink, B. FAME, the flux analysis and modeling environment. BMC Syst. Biol. 6, 8 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The Reproducible Research Results (R3) team, in particular, C. Trefois and Y. Jarosz, of the Luxembourg Centre for Systems Biomedicine, is acknowledged for their help in setting up the virtual machine and the Jenkins server. This study was funded by the National Centre of Excellence in Research (NCER) on Parkinson’s disease, the U.S. Department of Energy, Offices of Advanced Scientific Computing Research and the Biological and Environmental Research as part of the Scientific Discovery Through Advanced Computing program, grant no. DE-SC0010429. This project also received funding from the European Union’s HORIZON 2020 Research and Innovation Programme under grant agreement no. 668738 and the Luxembourg National Research Fund (FNR) ATTRACT program (FNR/A12/01) and OPEN (FNR/O16/11402054) grants. N.E.L. was supported by NIGMS (R35 GM119850) and the Novo Nordisk Foundation (NNF10CC1016517). M.A.P.O. was supported by the Luxembourg National Research Fund (FNR) grant AFR/6669348. A.R. was supported by the Lilly Innovation Fellows Award. F.J.P. was supported by the Minister of Economy and Competitiveness of Spain (BIO2016-77998-R) and the ELKARTEK Programme of the Basque Government (KK-2016/00026). I.A. was supported by a Basque Government predoctoral grant (PRE_2016_2_0044). B.Ø.P. was supported by the Novo Nordisk Foundation through the Center for Biosustainability at the Technical University of Denmark (NNF10CC1016517).

Author information

Authors and Affiliations

Authors

Contributions

S.A.: continuous integration, code review, opencobra.github.io/cobratoolbox, Jenkins, Documenter.py, changeCobraSolver, pull request support, tutorials, tests, coordination, manuscript, and initCobraToolbox. L.H.: continuous integration, code review, fastFVA (new version, test, and integration), MATLAB.devTools, opencobra.github.io, tutorials, tests, pull request support, coordination, manuscript, initCobraToolbox, and forum support. T.P.: input–output and transcriptomic integration, tutorials, tutorial reviews, input–output and transcriptomic integration sections of manuscript, forum support, pull request support, and code review. S.N.M.: development and update of strain design algorithms, GAMS and MATLAB integration, and tutorials. A.R.: transcriptomic data integration methods, tutorials, transcriptomic integration section of manuscript, RuMBA, pFBA, metabolic tasks, and tutorial review. A.H.: multispecies modeling code contribution, tutorial review, and testing. H.S. Haraldsdóttir: thermodynamics, conserved moiety, and sampling methods. J.W.: documentation. S.M.K.: SBML input–output support. V.V.: tutorials. S.M.: multispecies modeling, tutorial review, and testing. C.Y.N.: strain design code review, tutorial review, and manuscript (OptForce/biotech introduction). G.P.: tutorials and chemoinformatics for metabolite structures and atom mapping data. A.Ž.: metabolic cartography. S.H.J.C.: solution navigation, multispecies modeling code, and tutorial review. M.K.A.: metabolomic data integration. C.M.C.: tutorials and testing. J.M.: metabolic cartography and human metabolic network visualization tutorials. J.T.S.: modelBorgifier code and tutorial. A.N.: virtual metabolic human interoperability. A.B.: MinSpan method and tutorial, supervision on uFBA method and tutorial. B.C.: CHRR uniform sampling. D.C.E.A.: tutorials. L.V.V.: tutorials and genetic MCS implementation. I.A.: tutorials and genetic MCS implementation. S.G.: interoperability with CellNetAnalyzer. M.A.: adaptive Levenberg–Marquardt solver. M.B.G.: tutorial reviews. A.K.: Paint4Net code and tutorial. N.S.: development of metabolomic cartography tool and tutorial. H.M.L.: cardinality optimization solver. D.M.: quadruple-precision solvers. Y.S.: multiscale FBA reformulation. L.W.: strain design code review, tutorial review, and manuscript (OptForce). J.T.Y.: uFBA method and tutorial. M.A.P.O.: tutorial. P.T.V.: adaptive Levenberg–Marquardt solvers and boosted difference of convex optimization solver. L.P.E.A.: chemoinformatic data integration and documentation. I.K.: development of metabolomic cartography tool and tutorial. A.Z.: development of metabolomic cartography tool and tutorial. H.S. Hinton: E. coli core tutorials. W.A.B.: code refinement. F.J.A.A.: duplomonotone equation solver, boosted difference of convex optimization solver, and adaptive Levenberg–Marquardt solvers. F.J.P.: academic supervision, tutorials, and genetic MCS implementation. E.S.: academic supervision, Paint4Net, and tutorial. A.M.: academic supervision. S.V.: academic supervision and CHRR uniform sampling algorithm. M.H.: academic supervision and SBML input–output support. M.A.S.: academic supervision, quadruple-precision solvers, nullspace computation, and convex optimization. C.D.M.: academic supervision and strain design algorithms. N.E.L.: academic supervision and coding, and transcriptomic data integration, RuMBA, pFBA, metabolic tasks, and tutorial review. T.S.: academic supervision and FASTCORE algorithm. B.Ø.P.: academic supervision and openCOBRA stewardship. I.T.: academic supervision, tutorials, code contribution, and manuscript. R.M.T.F.: conceptualization, lead developer, academic supervision, software architecture, code review, sparse optimization, nullspace computation, thermodynamics, variational kinetics, fastGapFill, sampling, conserved moieties, network visualization, forum support, tutorials, and manuscript.

Corresponding author

Correspondence to Ronan M. T. Fleming.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Schellenberger, J. et al. Nat. Protocols 6, 1290–1307 (2011): https://www.nature.com/articles/nprot.2011.308

Becker, S. A. et al. Nat. Protocols 2, 727–738 (2007): https://www.nature.com/articles/nprot.2007.99

This protocol is an update to Nat. Protoc. 2, 727–738 (2007): https://doi.org/10.1038/nprot.2007.99 and Nat. Protoc. 6, 1290–1307 (2011): https://doi.org/10.1038/protex.2011.234

Supplementary information

Supplementary Data 1

Supplementary Manual 1

MATLAB basics

Supplementary Manual 2

Shell or Terminal basics

Supplementary Manual 3

Contributing to the COBRA Toolbox using git

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Heirendt, L., Arreckx, S., Pfau, T. et al. Creation and analysis of biochemical constraint-based models using the COBRA Toolbox v.3.0. Nat Protoc 14, 639–702 (2019). https://doi.org/10.1038/s41596-018-0098-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0098-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing AI and Robotics

Sign up for the Nature Briefing: AI and Robotics newsletter — what matters in AI and robotics research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: AI and Robotics