Your browser does not support JavaScript!
http://iet.metastore.ingenta.com
1887

Proper lumping in systems biology models

Proper lumping in systems biology models

For access to this article, please select a purchase option:

Buy article PDF
£12.50
(plus tax if applicable)
Buy Knowledge Pack
10 articles for £75.00
(plus taxes if applicable)

IET members benefit from discounts to all IET publications and free access to E&T Magazine. If you are an IET member, log in to your account and the discounts will automatically be applied.

Learn more about IET membership 

Recommend Title Publication to library

You must fill out fields marked with: *

Librarian details
Name:*
Email:*
Your details
Name:*
Email:*
Department:*
Why are you recommending this title?
Select reason:
 
 
 
 
 
IET Systems Biology — Recommend this title to your library

Thank you

Your recommendation has been sent to your librarian.

An algorithm for automatic order reduction of models defined by large systems of differential equations is presented. The algorithm was developed with systems biology models in mind and the motivation behind it is to develop mechanistic pharmacokinetic/pharmacodynamic models from already available systems biology models. The approach used for model reduction is proper lumping of the system's states and is based on a search through the possible combinations of lumps. To avoid combinatorial explosion, a heuristic, greedy search strategy is employed and comparison with the full exhaustive search provides evidence that it performs well. The method takes advantage of an apparent property of this kind of systems that lumps remain consistent over different levels of order reduction. Advantages of the method presented include: the variables and parameters of the reduced model retain a specific physiological meaning; the algorithm is automatic and easy to use; it can be used for nonlinear models and can handle parameter uncertainty and constraints. The algorithm was applied to a model of NF-κB signalling pathways in order to demonstrate its use and performance. Significant reduction was achieved for the example model, while agreement with the original model was proportional to the size of the reduced model, as expected. The results of the model reduction were compared with a published, intuitively reduced model of NF-κB signalling pathways and were found to be in agreement, in terms of the identified key species for the system's kinetic behaviour. The method may provide useful insights which are complementary to the intuitive reduction approach, especially in large systems.

References

    1. 1)
      • A. Hoffmann , A. Levchenko , M.L. Scott , D. Baltimore . The I kappa B-NF-kappa B signaling module: temporal control and selective gene activation. Science , 1241 - 1245
    2. 2)
      • S.M. Keating . SBMLToolbox.
    3. 3)
      • M.S. Okino , M.L. Mavrovouniotis . Simplification of mathematical models of chemical reaction systems. Chem. Rev. , 391 - 408
    4. 4)
      • C. Brochot , J. Toth , F.Y. Bois . Lumping in pharmacokinetics. J. Pharmacokinet. Pharmacodyn. , 719 - 736
    5. 5)
      • D.E. Nelson , A.E. Ihekwaba . Oscillations in NF-kappaB signaling control the dynamics of gene expression. Science , 704 - 708
    6. 6)
      • M.D. McKay , R.J. Beckman , W.J. Conover . A comparison of three methods for selecting values of input variables in the analysis of output from a computer code. Technometrics , 239 - 245
    7. 7)
      • A. Gelman , J.B. Carlin , H.S. Stern , D.B. Rubin . (2004) Bayesian data analysis.
    8. 8)
      • G. Li , H. Rabitz . A general-analysis of approximate lumping in chemical-kinetics. Chem. Eng. Sci. , 977 - 1002
    9. 9)
      • G.Y. Li , H. Rabitz . A general lumping analysis of a reaction system coupled with diffusion. Chem. Eng. Sci. , 2041 - 2053
    10. 10)
    11. 11)
      • S.J. Wilkinson , N. Benson , D.B. Kell . Proximate parameter tuning for biochemical networks with uncertain kinetic parameters. Mol. Biosyst. , 74 - 97
    12. 12)
      • I.A. Nestorov , L.J. Aarons , P.A. Arundel , M. Rowland . Lumping of whole-body physiologically based pharmacokinetic models. J. Pharmacokinet. Biopharm. , 21 - 46
    13. 13)
      • B. Bhattacharjee , D.A. Schwer , P.I. Barton , W.H. Green . Optimally-reduced kinetic models: reaction elimination in large-scale kinetic mechanisms. Combust. Flame , 191 - 208
    14. 14)
      • G. Astarita , R. Ocone . Lumping nonlinear kinetics. AICHE J. , 1299 - 1309
    15. 15)
      • W. Liebermeister , U. Baur , E. Klipp . Biochemical network models simplified by balanced truncation. FEBS J. , 4034 - 4043
    16. 16)
      • J. Wei , J.C.W. Kuo . A lumping analysis in monomolecular reaction systems – analysis of exactly lumpable system. Ind. Eng. Chem. Fund. , 114 - 123
    17. 17)
      • S. Krishna , M.H. Jensen , K. Sneppen . Minimal model of spiky oscillations in NF-kappaB signaling. Proc. Natl. Acad. Sci. USA , 10840 - 10845
    18. 18)
      • B.C. Moore . Principal component analysis in linear systems: controllability, observability and model reduction. IEEE Trans. Autom. Control , 17 - 32
    19. 19)
      • L.B. Sheiner , S. Beal , B. Rosenberg , V.V. Marathe . Forecasting individual pharmacokinetics. Clin. Pharmacol. Ther. , 294 - 305
    20. 20)
      • L.B. Sheiner , J.L. Steimer . Pharmacokinetic/pharmacodynamic modeling in drug development. Annu. Rev. Pharmacol. Toxicol. , 67 - 95
    21. 21)
      • G. Li , H. Rabitz . A general-analysis of exact lumping in chemical-kinetics. Chem. Eng. Sci. , 1413 - 1430
    22. 22)
      • N. Friedman , M. Linial , I. Nachman , D. Pe'er . Using Bayesian networks to analyze expression data. J. Comput. Biol. , 601 - 620
http://iet.metastore.ingenta.com/content/journals/10.1049/iet-syb_20070055
Loading

Related content

content/journals/10.1049/iet-syb_20070055
pub_keyword,iet_inspecKeyword,pub_concept
6
6
Loading
This is a required field
Please enter a valid email address