Extended Robust Boolean Network of Budding Yeast Cell Cycle

Sajad Shafiekhani, Mojtaba Shafiekhani, Sara Rahbar, Amir Homayoun Jafari

DOI: 10.4103/jmss.JMSS_40_19

Abstract


Background: How to explore the dynamics of transition probabilities between phases of budding yeast cell cycle (BYCC) network based on the dynamics of protein activities that control this network? How to identify the robust structure of protein interactions of BYCC Boolean network (BN)? Budding yeast allows scientists to put experiments into effect in order to discover the intracellular cell cycle regulating structures which are well simulated by mathematical modeling. Methods: We extended an available deterministic BN of proteins responsible for the cell cycle to a Markov chain model containing apoptosis besides G1, S, G2, M, and stationary G1. Using genetic algorithm (GA), we estimated the kinetic parameters of the extended BN model so that the subsequent transition probabilities derived using Markov chain model of cell states as normal cell cycle becomes the maximum while the structure of chemical interactions of extended BN of cell cycle becomes more stable. Results: Using kinetic parameters optimized by GA, the probability of the subsequent transitions between cell cycle phases is maximized. The relative basin size of stationary G1 increased from 86% to 96.48% while the number of attractors decreased from 7 in the original model to 5 in the extended one. Hence, an increase in the robustness of the system has been achieved. Conclusion: The structure of interacting proteins in cell cycle network affects its robustness and probabilities of transitions between different cell cycle phases. Markov chain and BN are good approaches to study the stability and dynamics of the cell cycle network.


Keywords


Boolean network, budding yeast cell cycle, genetic algorithm, Markov chain model

Full Text:

PDF

References


Kiyoumarsioskouei A, Shamloo A, Azimi S, Abeddoust M, Saidi M. A computational model for estimation of mechanical parameters in chemotactic endothelial cells. Sci Iran Trans B Mech Eng 2016;23:260. Back to cited text no. 1

Waltemath D, Karr JR, Bergmann FT, Chelliah V, Hucka M, Krantz M, et al. Toward community standards and software for whole-cell modeling. IEEE Trans Biomed Eng 2016;63:2007-14. Back to cited text no. 2

Yaghoubi S, Pishevar A, Saidi M, Shirani E. Modeling self-assembly of the surfactants into biological bilayer membranes with special chemical structures using dissipative particle dynamics method. Sci Iran Trans B Mech Eng 2016;23:942. Back to cited text no. 3

Fisher J, Henzinger TA. Executable cell biology. Nat Biotechnol 2007;25:1239-49. Back to cited text no. 4

Tanay A, Regev A. Scaling single-cell genomics from phenomenology to mechanism. Nature 2017;541:331-8. Back to cited text no. 5

Bargaje R, Trachana K, Shelton MN, McGinnis CS, Zhou JX, Chadick C, et al. Cell population structure prior to bifurcation predicts efficiency of directed differentiation in human induced pluripotent cells. Proc Natl Acad Sci U S A 2017;114:2271-6. Back to cited text no. 6

Davidson EH, Rast JP, Oliveri P, Ransick A, Calestani C, Yuh CH, et al. A genomic regulatory network for development. Science 2002;295:1669-78. Back to cited text no. 7

Huang L, Jiang Y, Chen Y. Predicting drug combination index and simulating the network-regulation dynamics by mathematical modeling of drug-targeted EGFR-ERK signaling pathway. Sci Rep 2017;7:40752. Back to cited text no. 8

Wittwehr C, Aladjov H, Ankley G, Byrne HJ, de Knecht J, Heinzle E, et al. How adverse outcome pathways can aid the development and use of computational prediction models for regulatory toxicology. Toxicol Sci 2017;155:326-36. Back to cited text no. 9

Shariatpanahi SP, Shariatpanahi SP, Madjidzadeh K, Hassan M, Abedi-Valugerdi M. Mathematical modeling of tumor-induced immunosuppression by myeloid-derived suppressor cells: Implications for therapeutic targeting strategies. J Theor Biol 2018;442:1-0. Back to cited text no. 10

Andreozzi S, Chakrabarti A, Soh KC, Burgard A, Yang TH, Van Dien S, et al. Identification of metabolic engineering targets for the enhancement of 1,4-butanediol production in recombinant E. coli using large-scale kinetic models. Metab Eng 2016;35:148-59. Back to cited text no. 11

Sipahi R, Zupanc GK. Stochastic cellular automata model of neurosphere growth: Roles of proliferative potential, contact inhibition, cell death, and phagocytosis. J Theor Biol 2018;445:151-65. Back to cited text no. 12

Lin YT, Hufton PG, Lee EJ, Potoyan DA. A stochastic and dynamical view of pluripotency in mouse embryonic stem cells. PLoS Comput Biol 2018;14:e1006000. Back to cited text no. 13

Zhu X, Yuan R, Hood L, Ao P. Endogenous molecular-cellular hierarchical modeling of prostate carcinogenesis uncovers robust structure. Prog Biophys Mol Biol 2015;117:30-42. Back to cited text no. 14

Bordbar A, Monk JM, King ZA, Palsson BO. Constraint-based models predict metabolic and associated cellular functions. Nat Rev Genet 2014;15:107-20. Back to cited text no. 15

Stewart-Ornstein J, Chen S, Bhatnagar R, Weissman JS, El-Samad H. Model-guided optogenetic study of PKA signaling in budding yeast. Mol Biol Cell 2017;28:221-7. Back to cited text no. 16

Neuert G, Munsky B, Tan RZ, Teytelman L, Khammash M, van Oudenaarden A. Systematic identification of signal-activated stochastic gene regulation. Science 2013;339:584-7. Back to cited text no. 17

Cheng TM, Heeger S, Chaleil RA, Matthews N, Stewart A, Wright J, et al. A simple biophysical model emulates budding yeast chromosome condensation. Elife 2015;4:e05565. Back to cited text no. 18

Serohijos AW, Shakhnovich EI. Merging molecular mechanism and evolution: Theory and computation at the interface of biophysics and evolutionary population genetics. Curr Opin Struct Biol 2014;26:84-91. Back to cited text no. 19

Briat C, Gupta A, Khammash M. Antithetic integral feedback ensures robust perfect adaptation in noisy biomolecular networks. Cell Syst 2016;2:15-26. Back to cited text no. 20

Zickenrott S, Angarica VE, Upadhyaya BB, del Sol A. Prediction of disease-gene-drug relationships following a differential network analysis. Cell Death Dis 2016;7:e2040. Back to cited text no. 21

Teodosiu C, David D, Teodosiu R. Numerical prediction of thermo-aeraulic behavior for a cavity with internal linear heat source. Sci Iran Trans B Mech Eng 2016;23:618. Back to cited text no. 22

O'Brien EJ, Monk JM, Palsson BO. Using genome-scale models to predict biological capabilities. Cell 2015;161:971-87. Back to cited text no. 23

Lee TI, Rinaldi NJ, Robert F, Odom DT, Bar-Joseph Z, Gerber GK, et al. Transcriptional regulatory networks in Saccharomyces cerevisiae. Science 2002;298:799-804. Back to cited text no. 24

Spellman PT, Sherlock G, Zhang MQ, Iyer VR, Anders K, Eisen MB, et al. Comprehensive identification of cell cycle-regulated genes of the yeast Saccharomyces cerevisiae by microarray hybridization. Mol Biol Cell 1998;9:3273-97. Back to cited text no. 25

Nurse P. The Josef Steiner lecture: CDKs and cell-cycle control in fission yeast: Relevance to other eukaryotes and cancer. Int J Cancer 1997;71:707-8. Back to cited text no. 26

Nasmyth K. Viewpoint: Putting the cell cycle in order. Science 1996;274:1643-5. Back to cited text no. 27

Mendenhall MD, Hodge AE. Regulation of Cdc28 cyclin-dependent protein kinase activity during the cell cycle of the yeast Saccharomyces cerevisiae. Microbiol Mol Biol Rev 1998;62:1191-243. Back to cited text no. 28

Hartwell LH, Weinert TA. Checkpoints: Controls that ensure the order of cell cycle events. Science 1989;246:629-34. Back to cited text no. 29

Li F, Long T, Lu Y, Ouyang Q, Tang C. The yeast cell-cycle network is robustly designed. Proc Natl Acad Sci U S A 2004;101:4781-6. Back to cited text no. 30

Kraikivski P, Chen KC, Laomettachit T, Murali TM, Tyson JJ. From START to FINISH: Computational analysis of cell cycle control in budding yeast. NPJ Syst Biol Appl 2015;1:15016. Back to cited text no. 31

Ahmadian M, Tyson J, Cao Y, editors. A Stochastic Model of Size Control in the Budding Yeast Cell Cycle. Proceedings of the 2018 ACM International Conference on Bioinformatics, Computational Biology, and Health Informatics, ACM; 2018 Back to cited text no. 32

Zhang Y, Qian M, Ouyang Q, Deng M, Li F, Tang C. Stochastic model of yeast cell-cycle network. Phys D Nonlin Phenom 2006;219:35-9. Back to cited text no. 33

Davidich MI, Bornholdt S. Boolean network model predicts cell cycle sequence of fission yeast. PLoS One 2008;3:e1672. Back to cited text no. 34

Barik D, Baumann WT, Paul MR, Novak B, Tyson JJ. A model of yeast cell-cycle regulation based on multisite phosphorylation. Mol Syst Biol 2010;6:405. Back to cited text no. 35

Mura I, Csikász-Nagy A. Stochastic petri net extension of a yeast cell cycle model. J Theor Biol 2008;254:850-60. Back to cited text no. 36

Chen KC, Csikasz-Nagy A, Gyorffy B, Val J, Novak B, Tyson JJ. Kinetic analysis of a molecular model of the budding yeast cell cycle. Mol Biol Cell 2000;11:369-91. Back to cited text no. 37

Chaves M, Sontag ED, Albert R. Methods of robustness analysis for boolean models of gene control networks. Syst Biol (Stevenage) 2006;153:154-67. Back to cited text no. 38

Liu F, Heiner M, Gilbert D. Coloured petri nets for multilevel, multiscale and multidimensional modelling of biological systems. Brief Bioinform 2019;20:877-86. Back to cited text no. 39

Liu F, Heiner M, Yang M. Fuzzy stochastic petri nets for modeling biological systems with uncertain kinetic parameters. PLoS One 2016;11:e0149674. Back to cited text no. 40

Werthmann B, Marwan W. Developmental switching in Physarum polycephalum: Petri net analysis of single cell trajectories of gene expression indicates responsiveness and genetic plasticity of the Waddington quasipotential landscape. J Phys D Appl Phys 2017;50:464003. Back to cited text no. 41

Herajy M, Liu F, Heiner M. Efficient modelling of yeast cell cycles based on multisite phosphorylation using coloured hybrid petri nets with marking-dependent arc weights. Nonlin Anal Hybrid Syst 2018;27:191-212. Back to cited text no. 42

Laomettachit T, Chen KC, Baumann WT, Tyson JJ. A Model of yeast cell-cycle regulation based on a standard component modeling strategy for protein regulatory networks. PLoS One 2016;11:e0153738. Back to cited text no. 43

Castro C, Flores DL, Cervantes-Vásquez D, Vargas-Viveros E, Gutiérrez-López E, Muñoz-Muñoz F. An agent-based model of the fission yeast cell cycle. Curr Genet 2019;65:193-200. Back to cited text no. 44

Steggles LJ, Banks R, Wipat A, editors. Modelling and Analysing Genetic Networks: From Boolean Networks to Petri Nets. International Conference on Computational Methods in Systems Biology, Springer; 2006. Back to cited text no. 45

Kaufmann BB, van Oudenaarden A. Stochastic gene expression: From single molecules to the proteome. Curr Opin Genet Dev 2007;17:107-12. Back to cited text no. 46

Elowitz MB, Levine AJ, Siggia ED, Swain PS. Stochastic gene expression in a single cell. Science 2002;297:1183-6. Back to cited text no. 47

McAdams HH, Arkin A. Stochastic mechanisms in gene expression. Proc Natl Acad Sci U S A 1997;94:814-9. Back to cited text no. 48

Paulsson J. Models of stochastic gene expression. Phys Life Rev 2005;2:157-75. Back to cited text no. 49

Soltani M, Vargas-Garcia CA, Antunes D, Singh A. Intercellular variability in protein levels from stochastic expression and noisy cell cycle processes. PLoS Comput Biol 2016;12:e1004972. Back to cited text no. 50

Iyer-Biswas S, Wright CS, Henry JT, Lo K, Burov S, Lin Y, et al. Scaling laws governing stochastic growth and division of single bacterial cells. Proc Natl Acad Sci U S A 2014;111:15912-7. Back to cited text no. 51

Ghusinga KR, Vargas-Garcia CA, Singh A. A mechanistic stochastic framework for regulating bacterial cell division. Sci Rep 2016;6:30229. Back to cited text no. 52

Liepe J, Kirk P, Filippi S, Toni T, Barnes CP, Stumpf MP. A framework for parameter estimation and model selection from experimental data in systems biology using approximate bayesian computation. Nat Protoc 2014;9:439-56. Back to cited text no. 53

Gábor A, Banga JR. Robust and efficient parameter estimation in dynamic models of biological systems. BMC Syst Biol 2015;9:74. Back to cited text no. 54

Moles CG, Mendes P, Banga JR. Parameter estimation in biochemical pathways: A comparison of global optimization methods. Genome Res 2003;13:2467-74. Back to cited text no. 55

Shaw O, Steggles J, Wipat A. Automatic parameterisation of stochastic petri net models of biological networks. Electron Notes Theor Comput Sci 2006;151:111-29. Back to cited text no. 56

Scialdone A, Natarajan KN, Saraiva LR, Proserpio V, Teichmann SA, Stegle O, et al. Computational assignment of cell-cycle stage from single-cell transcriptome data. Methods 2015;85:54-61. Back to cited text no. 57


Refbacks

  • There are currently no refbacks.


 

  https://e-rasaneh.ir/Certificate/22728

https://e-rasaneh.ir/

ISSN : 2228-7477