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.

Constraint-based models predict metabolic and associated cellular functions

Key Points

  • Understanding the genotype–phenotype relationship of a cell has been a long-standing goal in biology. The availability of genome sequencing and of other high-throughput 'omic' data sets provides an opportunity to parameterize various statistical and mechanistic models.

  • Genome-scale metabolic network reconstructions contain comprehensively curated and systematized information on the cellular metabolism of an organism. These networks can be converted into a mathematical format that is amenable to constraint-based modelling.

  • Biological phenotype is constrained by the complex relationships between the genotype of a cell, its environment and physico-chemical laws. Rather than deriving a single solution to a problem, the philosophy of constraint-based modelling is to impose known constraints on physiological function to study the set of possible solutions.

  • Constraint-based modelling has a 30-year history, which can be separated into four phases — from initial theoretical interest and conception to maturing to predictive biological practice. In the current fourth phase, there is a critical mass of studies that combine high-throughput data and constraint-based models (CBMs) to answer relevant biological questions in a prospective manner.

  • Key successes in this field of research include deriving underlying principles for both optimal flux states and protein evolution; experimentally discovering cancer drug targets and antibiotics; and designing organisms that overproduce metabolic precursors of commodity chemicals.

  • Integrated modelling efforts combine CBMs with alternative modelling frameworks that are more amenable to capture other cellular processes in order to expand predictive scope outside metabolism. Expansions for other cellular features — such as transcription and translation machinery, transcriptional regulation and structural protein properties — have been constructed.

  • The formulation of high-dimensional models is enabled by the availability of genome-sequences (that is, a parts list), high-throughput omic data (that is, a functional readout) and CBMs (that is, a mechanistic framework to reconcile network topology, primary literature and omic data). These models can be used to quantitatively study the genotype–phenotype relationship for cellular metabolism, which will lead to a new generation of genome-scale science.

Abstract

The prediction of cellular function from a genotype is a fundamental goal in biology. For metabolism, constraint-based modelling methods systematize biochemical, genetic and genomic knowledge into a mathematical framework that enables a mechanistic description of metabolic physiology. The use of constraint-based approaches has evolved over ~30 years, and an increasing number of studies have recently combined models with high-throughput data sets for prospective experimentation. These studies have led to validation of increasingly important and relevant biological predictions. As reviewed here, these recent successes have tangible implications in the fields of microbial evolution, interaction networks, genetic engineering and drug discovery.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: The multiple uses of high-throughput data in constraint-based models.
Figure 2: Predictive case studies in understanding underlying principles of interaction networks.
Figure 3: Predictive case studies in metabolic engineering and drug targeting.
Figure 4: Expanding predictive scope through integrative modelling.

Similar content being viewed by others

References

  1. Feist, A. M., Herrgard, M. J., Thiele, I., Reed, J. L. & Palsson, B. O. Reconstruction of biochemical networks in microorganisms. Nature Rev. Microbiol. 7, 129–143 (2009). This is a review on constructing and validating a genome-scale metabolic network.

    CAS  Google Scholar 

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

    CAS  Google Scholar 

  3. Lewis, N. E., Nagarajan, H. & Palsson, B. O. Constraining the metabolic genotype–phenotype relationship using a phylogeny of in silico methods. Nature Rev. Microbiol. 10, 291–305 (2012). This is a thorough review of the various constraint-based modelling methodologies.

    CAS  Google Scholar 

  4. 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).

    PubMed  Google Scholar 

  5. Klitgord, N. & Segre, D. Environments that induce synthetic microbial ecosystems. PLoS Comput. Biol. 6, e1001002 (2010).

    PubMed  PubMed Central  Google Scholar 

  6. 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).

    PubMed  PubMed Central  Google Scholar 

  7. Bordbar, A., Lewis, N. E., Schellenberger, J., Palsson, B. O. & Jamshidi, N. Insight into human alveolar macrophage and M. tuberculosis interactions via metabolic reconstructions. Mol. Syst. Biol. 6, 422 (2010).

    PubMed  PubMed Central  Google Scholar 

  8. Lewis, N. E. et al. Large-scale in silico modeling of metabolic interactions between cell types in the human brain. Nature Biotech. 28, 1279–1285 (2010).

    CAS  Google Scholar 

  9. Papin, J. A. & Palsson, B. O. The JAK–STAT signaling network in the human B-cell: an extreme signaling pathway analysis. Biophys. J. 87, 37–46 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Li, F., Thiele, I., Jamshidi, N. & Palsson, B. O. Identification of potential pathway mediation targets in Toll-like receptor signaling. PLoS Comput. Biol. 5, e1000292 (2009).

    PubMed  PubMed Central  Google Scholar 

  11. Gianchandani, E. P., Joyce, A. R., Palsson, B. O. & Papin, J. A. Functional states of the genome-scale Escherichia coli transcriptional regulatory system. PLoS Comput. Biol. 5, e1000403 (2009).

    PubMed  PubMed Central  Google Scholar 

  12. Thiele, I., Jamshidi, N., Fleming, R. M. & Palsson, B. O. 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).

    PubMed  PubMed Central  Google Scholar 

  13. Fell, D. A. & Small, J. R. Fat synthesis in adipose tissue. An examination of stoichiometric constraints. Biochem. J. 238, 781–786 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Majewski, R. A. & Domach, M. M. Simple constrained optimization view of acetate overflow in E. coli. Biotechnol. Bioeng. 35, 732–738 (1990).

    CAS  PubMed  Google Scholar 

  15. Savinell, J. M. & Palsson, B. O. Optimal selection of metabolic fluxes for in vivo measurement. II. Application to Escherichia coli and hybridoma cell metabolism. J. Theor. Biol. 155, 215–242 (1992).

    CAS  PubMed  Google Scholar 

  16. Varma, A. & Palsson, B. O. Stoichiometric flux balance models quantitatively predict growth and metabolic by-product secretion in wild-type Escherichia coli W3110. Appl. Environ. Microbiol. 60, 3724–3731 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Schuster, S. & Hilgetag, C. On elementary flux modes in biochemical reaction systems at steady state. J. Biol. Systems 2, 165–182 (1994).

    Google Scholar 

  18. Schilling, C. H., Letscher, D. & Palsson, B. O. 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).

    CAS  PubMed  Google Scholar 

  19. Clarke, B. L. in Advances in Chemical Physics Vol. 43 (eds. Prigogine, I. & Rice, S. A.) 1–215 (Wiley, 1980).

    Google Scholar 

  20. Dandekar, T., Schuster, S., Snel, B., Huynen, M. & Bork, P. Pathway alignment: application to the comparative analysis of glycolytic enzymes. Biochem. J. 343, 115–124 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Liao, J. C., Hou, S. Y. & Chao, Y. P. Pathway analysis, engineering and physiological considerations for redirecting central metabolism. Biotechnol. Bioeng. 52, 129–140 (1996).

    CAS  PubMed  Google Scholar 

  22. Fleischmann, R. D. et al. Whole-genome random sequencing and assembly of Haemophilus influenzae Rd. Science 269, 496–512 (1995).

    CAS  PubMed  Google Scholar 

  23. Edwards, J. S. & Palsson, B. O. Systems properties of the Haemophilus influenzae Rd metabolic genotype. J. Biol. Chem. 274, 17410–17416 (1999).

    CAS  PubMed  Google Scholar 

  24. Edwards, J. S., Ibarra, R. U. & Palsson, B. O. In silico predictions of Escherichia coli metabolic capabilities are consistent with experimental data. Nature Biotech. 19, 125–130 (2001).

    CAS  Google Scholar 

  25. Segre, D., Vitkup, D. & Church, G. M. Analysis of optimality in natural and perturbed metabolic networks. Proc. Natl Acad. Sci. USA 99, 15112–15117 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Stelling, J., Klamt, S., Bettenbrock, K., Schuster, S. & Gilles, E. D. Metabolic network structure determines key aspects of functionality and regulation. Nature 420, 190–193 (2002).

    CAS  PubMed  Google Scholar 

  27. Ibarra, R. U., Edwards, J. S. & Palsson, B. O. Escherichia coli K-12 undergoes adaptive evolution to achieve in silico predicted optimal growth. Nature 420, 186–189 (2002).

    CAS  PubMed  Google Scholar 

  28. Almaas, E., Kovacs, B., Vicsek, T., Oltvai, Z. N. & Barabasi, A. L. Global organization of metabolic fluxes in the bacterium Escherichia coli. Nature 427, 839–843 (2004).

    CAS  PubMed  Google Scholar 

  29. Papp, B., Pal, C. & Hurst, L. D. Metabolic network analysis of the causes and evolution of enzyme dispensability in yeast. Nature 429, 661–664 (2004).

    CAS  PubMed  Google Scholar 

  30. Pal, C., Papp, B. & Lercher, M. J. Adaptive evolution of bacterial metabolic networks by horizontal gene transfer. Nature Genet. 37, 1372–1375 (2005).

    CAS  PubMed  Google Scholar 

  31. Hyduke, D. R., Lewis, N. E. & Palsson, B. O. Analysis of omics data with genome-scale models of metabolism. Mol. Biosyst 9, 167–174 (2013). This is a review of techniques to integrate omic data with CBMs.

    CAS  PubMed  Google Scholar 

  32. Patil, K. R. & Nielsen, J. Uncovering transcriptional regulation of metabolism by using metabolic network topology. Proc. Natl Acad. Sci. USA 102, 2685–2689 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kharchenko, P., Church, G. M. & Vitkup, D. Expression dynamics of a cellular metabolic network. Mol Syst Biol 1, 2005.0016 (2005).

    PubMed  PubMed Central  Google Scholar 

  34. Shlomi, T., Cabili, M. N., Herrgard, M. J., Palsson, B. O. & Ruppin, E. Network-based prediction of human tissue-specific metabolism. Nature Biotech. 26, 1003–1010 (2008).

    CAS  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  36. Carlson, R. & Srienc, F. Fundamental Escherichia coli biochemical pathways for biomass and energy production: creation of overall flux states. Biotechnol. Bioeng. 86, 149–162 (2004).

    CAS  PubMed  Google Scholar 

  37. Carlson, R. & Srienc, F. Fundamental Escherichia coli biochemical pathways for biomass and energy production: identification of reactions. Biotechnol. Bioeng. 85, 1–19 (2004).

    CAS  PubMed  Google Scholar 

  38. Harcombe, W. R., Delaney, N. F., Leiby, N., Klitgord, N. & Marx, C. J. The ability of flux balance analysis to predict evolution of central metabolism scales with the initial distance to the optimum. PLoS Comput. Biol. 9, e1003091 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Schuetz, R., Kuepfer, L. & Sauer, U. Systematic evaluation of objective functions for predicting intracellular fluxes in Escherichia coli. Mol Syst Biol. 3, 119 (2007).

    PubMed  PubMed Central  Google Scholar 

  40. Molenaar, D., van Berlo, R., de Ridder, D. & Teusink, B. Shifts in growth strategies reflect tradeoffs in cellular economics. Mol. Syst. Biol. 5, 323 (2009).

    PubMed  PubMed Central  Google Scholar 

  41. Schuetz, R., Zamboni, N., Zampieri, M., Heinemann, M. & Sauer, U. Multidimensional optimality of microbial metabolism. Science 336, 601–604 (2012).

    CAS  PubMed  Google Scholar 

  42. 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).

    PubMed  PubMed Central  Google Scholar 

  43. Khersonsky, O. & Tawfik, D. S. Enzyme promiscuity: a mechanistic and evolutionary perspective. Annu. Rev. Biochem. 79, 471–505 (2010).

    CAS  PubMed  Google Scholar 

  44. Nam, H. et al. Network context and selection in the evolution to enzyme specificity. Science 337, 1101–1104 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. 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).

    PubMed  PubMed Central  Google Scholar 

  46. Baba, T. et al. Construction of Escherichia coli K-12 in-frame, single-gene knockout mutants: the Keio collection. Mol Syst Biol. 2, 2006.0008 (2006).

    PubMed  PubMed Central  Google Scholar 

  47. Scheer, M. et al. BRENDA, the enzyme information system in 2011. Nucleic Acids Res. 39, D670–D676 (2011).

    CAS  PubMed  Google Scholar 

  48. Lobel, L., Sigal, N., Borovok, I., Ruppin, E. & Herskovits, A. A. Integrative genomic analysis identifies isoleucine and CodY as regulators of Listeria monocytogenes virulence. PLoS Genet. 8, e1002887 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Costanzo, M. et al. The genetic landscape of a cell. Science 327, 425–431 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Uetz, P. et al. A comprehensive analysis of protein–protein interactions in Saccharomyces cerevisiae. Nature 403, 623–627 (2000).

    CAS  PubMed  Google Scholar 

  51. Gama-Castro, S. et al. RegulonDB version 7.0: transcriptional regulation of Escherichia coli K-12 integrated within genetic sensory response units (Gensor Units). Nucleic Acids Res. 39, D98–D105 (2011).

    CAS  PubMed  Google Scholar 

  52. Segre, D., DeLuna, A., Church, G. M. & Kishnoy, R. Modular epistasis in yeast metabolism. Nature Genet. 37, 77–83 (2005).

    CAS  PubMed  Google Scholar 

  53. Harrison, R., Papp, B., Pal, C., Oliver, S. G. & Delneri, D. Plasticity of genetic interactions in metabolic networks of yeast. Proc. Natl Acad. Sci. USA 104, 2307–2312 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. He, X., Qian, W., Wang, Z., Li, Y. & Zhang, J. Prevalent positive epistasis in Escherichia coli and Saccharomyces cerevisiae metabolic networks. Nature Genet. 42, 272–276 (2010).

    CAS  PubMed  Google Scholar 

  55. Szappanos, B. et al. An integrated approach to characterize genetic interaction networks in yeast metabolism. Nature Genet. 43, 656–662 (2011).

    CAS  PubMed  Google Scholar 

  56. Mo, M. L., Palsson, B. O. & Herrgard, M. J. Connecting extracellular metabolomic measurements to intracellular flux states in yeast. BMC Syst. Biol. 3, 37 (2009).

    PubMed  PubMed Central  Google Scholar 

  57. Wessely, F. et al. Optimal regulatory strategies for metabolic pathways in Escherichia coli depending on protein costs. Mol. Syst. Biol. 7, 515 (2011).

    PubMed  PubMed Central  Google Scholar 

  58. Notebaart, R. A., Teusink, B., Siezen, R. J. & Papp, B. Co-regulation of metabolic genes is better explained by flux coupling than by network distance. PLoS Comput. Biol. 4, e26 (2008).

    PubMed  PubMed Central  Google Scholar 

  59. Kaleta, C., de Figueiredo, L. F. & Schuster, S. Can the whole be less than the sum of its parts? Pathway analysis in genome-scale metabolic networks using elementary flux patterns. Genome Res. 19, 1872–1883 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Faith, J. J. et al. Many Microbe Microarrays Database: uniformly normalized Affymetrix compendia with structured experimental metadata. Nucleic Acids Res. 36, D866–D870 (2008).

    CAS  PubMed  Google Scholar 

  61. Orth, J. D. & Palsson, B. O. Systematizing the generation of missing metabolic knowledge. Biotechnol. Bioeng. 107, 403–412 (2010). This is a review on techniques and applications of CBMs for a targeted expansion of biochemical knowledge.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  63. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  65. Kanehisa, M., Goto, S., Sato, Y., Furumichi, M. & Tanabe, M. KEGG for integration and interpretation of large-scale molecular data sets. Nucleic Acids Res. 40, D109–D114 (2012).

    CAS  PubMed  Google Scholar 

  66. Nakahigashi, K. et al. Systematic phenome analysis of Escherichia coli multiple-knockout mutants reveals hidden reactions in central carbon metabolism. Mol. Syst. Biol. 5, 306 (2009).

    PubMed  PubMed Central  Google Scholar 

  67. Lee, S. Y., Lee, D. Y. & Kim, T. Y. Systems biotechnology for strain improvement. Trends Biotechnol. 23, 349–358 (2005).

    CAS  PubMed  Google Scholar 

  68. Park, J. H. & Lee, S. Y. Towards systems metabolic engineering of microorganisms for amino acid production. Curr. Opin. Biotechnol. 19, 454–460 (2008). This is a review of using systems biology methodologies for metabolic engineering applications.

    CAS  PubMed  Google Scholar 

  69. Caspeta, L. & Nielsen, J. Economic and environmental impacts of microbial biodiesel. Nature Biotech. 31, 789–793 (2013).

    CAS  Google Scholar 

  70. Yim, H. et al. Metabolic engineering of Escherichia coli for direct production of 1,4-butanediol. Nature Chem. Biol. 7, 445–452 (2011).

    CAS  Google Scholar 

  71. Hatzimanikatis, V. et al. Exploring the diversity of complex metabolic networks. Bioinformatics 21, 1603–1609 (2005).

    CAS  PubMed  Google Scholar 

  72. Constantinou, L. & Gani, R. New group-contribution method for estimating properties of pure compounds. AIChE J. 40, 1697–1710 (1994).

    CAS  Google Scholar 

  73. Khatri, P., Sirota, M. & Butte, A. J. Ten years of pathway analysis: current approaches and outstanding challenges. PLoS Comput. Biol. 8, e1002375 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. 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).

    CAS  PubMed  Google Scholar 

  75. Oberhardt, M. A., Yizhak, K. & Ruppin, E. Metabolically re-modeling the drug pipeline. Curr. Opin. Pharmacol. 13, 778–785 (2013). This is a review on using constraint-based modelling for drug discovery.

    CAS  PubMed  Google Scholar 

  76. Hsu, P. P. & Sabatini, D. M. Cancer cell metabolism: Warburg and beyond. Cell 134, 703–707 (2008).

    CAS  PubMed  Google Scholar 

  77. Folger, O. et al. Predicting selective drug targets in cancer through metabolic networks. Mol. Syst. Biol. 7, 501 (2011).

    PubMed  PubMed Central  Google Scholar 

  78. Frezza, C. et al. Haem oxygenase is synthetically lethal with the tumour suppressor fumarate hydratase. Nature 477, 225–228 (2011).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  80. Kim, P. J. et al. Metabolite essentiality elucidates robustness of Escherichia coli metabolism. Proc. Natl Acad. Sci. USA 104, 13638–13642 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Kim, H. U. et al. Integrative genome-scale metabolic analysis of Vibrio vulnificus for drug targeting and discovery. Mol. Syst. Biol. 7, 460 (2011).

    PubMed  PubMed Central  Google Scholar 

  82. Brynildsen, M. P., Winkler, J. A., Spina, C. S., MacDonald, I. C. & Collins, J. J. Potentiating antibacterial activity by predictably enhancing endogenous microbial ROS production. Nature Biotech. 31, 160–165 (2013).

    CAS  Google Scholar 

  83. Lerman, J. A. et al. In silico method for modelling metabolism and gene product expression at genome scale. Nature Commun. 3, 929 (2012).

    Google Scholar 

  84. Zhang, Y. et al. Three-dimensional structural view of the central metabolic network of Thermotoga maritima. Science 325, 1544–1549 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Thiele, I., Fleming, R. M., Bordbar, A., Schellenberger, J. & Palsson, B. O. Functional characterization of alternate optimal solutions of Escherichia coli's transcriptional and translational machinery. Biophys. J. 98, 2072–2081 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Pramanik, J. & Keasling, J. D. Effect of Escherichia coli biomass composition on central metabolic fluxes predicted by a stoichiometric model. Biotechnol. Bioeng. 60, 230–238 (1998).

    CAS  PubMed  Google Scholar 

  87. Rodionova, I. A. et al. Diversity and versatility of the Thermotoga maritima sugar kinome. J. Bacteriol. 194, 5552–5563 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. O'Brien, E. J., Lerman, J. A., Chang, R. L., Hyduke, D. R. & Palsson, B. O. Genome-scale models of metabolism and gene expression extend and refine growth phenotype prediction. Mol. Syst. Biol. 9, 693 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Chandrasekaran, S. & Price, N. D. Probabilistic integrative modeling of genome-scale metabolic and regulatory networks in Escherichia coli and Mycobacterium tuberculosis. Proc. Natl Acad. Sci. USA 107, 17845–17850 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Covert, M. W., Knight, E. M., Reed, J. L., Herrgard, M. J. & Palsson, B. O. Integrating high-throughput and computational data elucidates bacterial networks. Nature 429, 92–96 (2004).

    CAS  PubMed  Google Scholar 

  91. Chang, R. L. et al. Structural systems biology evaluation of metabolic thermotolerance in Escherichia coli. Science 340, 1220–1223 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Gu, J. & Bourne, P. E. Structural bioinformatics (Wiley-Blackwell, 2009).

    Google Scholar 

  93. Marr, A. G. & Ingraham, J. L. Effect of temperature on the composition of fatty acids in Escherichia coli. J. Bacteriol. 84, 1260–1267 (1962).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Tenaillon, O. et al. The molecular diversity of adaptive convergence. Science 335, 457–461 (2012).

    CAS  PubMed  Google Scholar 

  95. Mörters, P., Peres, Y., Schramm, O. & Werner, W. Brownian motion (Cambridge Univ. Press, 2010).

    Google Scholar 

  96. Karr, J. R. et al. A whole-cell computational model predicts phenotype from genotype. Cell 150, 389–401 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Thiele, I. et al. A community-driven global reconstruction of human metabolism. Nature Biotech. 31, 419–425 (2013).

    CAS  Google Scholar 

  98. Borenstein, E. Computational systems biology and in silico modeling of the human microbiome. Brief Bioinform. 13, 769–780 (2012).

    PubMed  Google Scholar 

  99. Levy, R. & Borenstein, E. Metabolic modeling of species interaction in the human microbiome elucidates community-level assembly rules. Proc. Natl Acad. Sci. USA 110, 12804–12809 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Atkinson, D. E. The energy charge of the adenylate pool as a regulatory parameter. Interaction with feedback modifiers. Biochemistry 7, 4030–4034 (1968).

    CAS  PubMed  Google Scholar 

  101. Weisz, P. B. Diffusion and chemical transformation. Science 179, 433–440 (1973).

    CAS  PubMed  Google Scholar 

  102. Reed, J. L. Shrinking the metabolic solution space using experimental datasets. PLoS Comput. Biol. 8, e1002662 (2012). This is a review of the potential constraints that have been placed on CBMs.

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Colijn, C. et al. Interpreting expression data with metabolic flux models: predicting Mycobacterium tuberculosis mycolic acid production. PLoS Comput. Biol. 5, e1000489 (2009).

    PubMed  PubMed Central  Google Scholar 

  104. Orth, J. D., Thiele, I. & Palsson, B. O. What is flux balance analysis? Nature Biotech. 28, 245–248 (2010). This paper presents a primer on the theory, applications and software toolboxes for FBA.

    CAS  Google Scholar 

  105. Mahadevan, R. & Schilling, C. H. The effects of alternate optimal solutions in constraint-based genome-scale metabolic models. Metab. Eng. 5, 264–276 (2003).

    CAS  PubMed  Google Scholar 

  106. Wilkinson, D. J. Stochastic modelling for quantitative description of heterogeneous biological systems. Nature Rev. Genet. 10, 122–133 (2009).

    CAS  PubMed  Google Scholar 

  107. Steuer, R. Computational approaches to the topology, stability and dynamics of metabolic networks. Phytochemistry 68, 2139–2151 (2007).

    CAS  PubMed  Google Scholar 

  108. de Jong, H. Modeling and simulation of genetic regulatory systems: a literature review. J. Comput. Biol. 9, 67–103 (2002).

    CAS  PubMed  Google Scholar 

  109. Friedman, N., Linial, M., Nachman, I. & Pe'er, D. Using Bayesian networks to analyze expression data. J. Computat. Biol. 7, 601–620 (2000).

    CAS  Google Scholar 

  110. Stephens, M. & Balding, D. J. Bayesian statistical methods for genetic association studies. Nature Rev. Genet. 10, 681–690 (2009).

    CAS  PubMed  Google Scholar 

  111. Ideker, T. & Krogan, N. J. Differential network biology. Mol. Syst. Biol. 8, 565 (2012).

    PubMed  PubMed Central  Google Scholar 

  112. Califano, A., Butte, A. J., Friend, S., Ideker, T. & Schadt, E. Leveraging models of cell regulation and GWAS data in integrative network-based association studies. Nature Genet. 44, 841–847 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank D. Zielinski, J. Lerman, N. E. Lewis and H. Nagarajan for their criticisms and comments on the manuscript. This work was supported by the US National Institutes of Health grants GM068837 and GM057089, and by the Novo Nordisk Foundation. Z.A.K. is supported through the US National Science Foundation Graduate Research Fellowship (DGE-1144086).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bernhard O. Palsson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

PowerPoint slides

Glossary

Metabolite overflows

Biological phenomena whereby the rate of substrate use by a cell for growth is lower than the rates of uptake and conversion of the substrate, which results in production of side metabolites (for example, acetate in Escherichia coli).

Metabolic fluxes

The rates of turnover or movement of metabolites through a reaction or a pathway.

Objective functions

The particular variables, or metabolic reactions, that are being maximized or minimized for by the linear programme. In flux-balance analysis, the objective function is often a pseudoreaction for biomass generation that represents cellular growth.

Metabolic pathways

In the context of this Review, sets of pathways that are calculated by metabolic network-based pathway analysis tools such as Extreme Pathways and Elementary Flux Modes.

Metabolic engineering

The practice of improving cellular production of target compounds of interest by modifying and optimizing genetic, regulatory and environmental parameters of cellular metabolism.

Genome-scale models

The formulation, using mathematical models, of genome-scale metabolic network reconstructions. They are synonymous with constraint-based models in the context of this Review.

Pathway enrichment analysis

A high-throughput data analysis technique to understand more global changes in an experiment by grouping individual measurements of biological components (for example, genes and proteins) into a context that is based on various pathway databases (for example, Kyoto Encyclopedia of Genes and Genomes, BioCyc and Gene Ontology).

Metabolic flux analysis

An experimental approach to identify metabolic fluxes using isotopically labelled metabolites and computational software that reconciles experimental data with network topology.

Flux distributions

Sets of calculated flux values for all reactions in a constraint-based model.

Pareto surface

The space that is formed when multiple objective functions are modelled at once; it represents a set of optimal solutions, in which increasing the value of one of the objectives results in a trade-off with other objective values.

Central carbon metabolism

The metabolic pathways and reactions that convert sugars into the metabolic precursors that are required for growth. It is typically comprised of glycolysis, pentose phosphate pathways and the tricarboxylic acid cycle.

Solution space

The range of all feasible values for variables in a constraint-based model, which represents all potential metabolic reaction flux distributions on the basis of the given constraints.

Machine learning method

A method that applies statistical methods to discover generalizable rules and patterns in complex data sets.

Gap-filling

Pertaining to a procedure for targeted expansion of metabolic knowledge, whereby prospective experiments are designed on the basis of discrepancies in experimental data and model predictions.

Auxotrophies

Metabolic limitations that impair the ability of a cell or organism to synthesize a particular metabolite that is essential for growth, which force the cell or organism to rely on an exogenous source of the nutrient.

Reaction bounds

User-defined constraints on the minimum and maximum allowable flux values for a particular metabolic reaction in a constraint-based model.

Metabolite essentiality analysis

A metabolite-centric approach to determine essential components for cellular growth. To computationally test the essentiality of a metabolite, the consuming reactions of the particular metabolites are constrained to zero.

Coupling constraints

Constraints that enforce strict relationships between model biochemical transformations, thereby connecting the fluxes for different cellular processes (such as transcription, translation, and tRNA and protein use for a metabolic reaction).

Linear programming

A mathematical optimization technique that calculates the maximum or minimum value of a particular variable (that is, the objective function) on the basis of a set of linear constraints; an example of this is flux-balance analysis.

Consensus sequences

Conserved sequences of nucleotides or amino acids that represent the target for a biomolecular event, often for proteins binding to the genome.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Bordbar, A., Monk, J., King, Z. et al. Constraint-based models predict metabolic and associated cellular functions. Nat Rev Genet 15, 107–120 (2014). https://doi.org/10.1038/nrg3643

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3643

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

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