SBMLBioModels: F - G

F


Feist2006_methanogenesis_OptiH2-CO2: MODEL5662398146v0.0.1

This model originates from BioModels Database: A Database of Annotated Published Models (http://www.ebi.ac.uk/biomodels/…

Details

We present a genome-scale metabolic model for the archaeal methanogen Methanosarcina barkeri. We characterize the metabolic network and compare it to reconstructions from the prokaryotic, eukaryotic and archaeal domains. Using the model in conjunction with constraint-based methods, we simulate the metabolic fluxes and resulting phenotypes induced by different environmental and genetic conditions. This represents the first large-scale simulation of either a methanogen or an archaeal species. Model predictions are validated by comparison to experimental growth measurements and phenotypes of M. barkeri on different substrates. The predicted growth phenotypes for wild type and mutants of the methanogenic pathway have a high level of agreement with experimental findings. We further examine the efficiency of the energy-conserving reactions in the methanogenic pathway, specifically the Ech hydrogenase reaction, and determine a stoichiometry for the nitrogenase reaction. This work demonstrates that a reconstructed metabolic network can serve as an analysis platform to predict cellular phenotypes, characterize methanogenic growth, improve the genome annotation and further uncover the metabolic characteristics of methanogenesis. link: http://identifiers.org/pubmed/16738551

Feist2006_methanogenesis_OptiMethanol: MODEL5662324959v0.0.1

This model originates from BioModels Database: A Database of Annotated Published Models (http://www.ebi.ac.uk/biomodels/…

Details

We present a genome-scale metabolic model for the archaeal methanogen Methanosarcina barkeri. We characterize the metabolic network and compare it to reconstructions from the prokaryotic, eukaryotic and archaeal domains. Using the model in conjunction with constraint-based methods, we simulate the metabolic fluxes and resulting phenotypes induced by different environmental and genetic conditions. This represents the first large-scale simulation of either a methanogen or an archaeal species. Model predictions are validated by comparison to experimental growth measurements and phenotypes of M. barkeri on different substrates. The predicted growth phenotypes for wild type and mutants of the methanogenic pathway have a high level of agreement with experimental findings. We further examine the efficiency of the energy-conserving reactions in the methanogenic pathway, specifically the Ech hydrogenase reaction, and determine a stoichiometry for the nitrogenase reaction. This work demonstrates that a reconstructed metabolic network can serve as an analysis platform to predict cellular phenotypes, characterize methanogenic growth, improve the genome annotation and further uncover the metabolic characteristics of methanogenesis. link: http://identifiers.org/pubmed/16738551

Feist2006_methanogenesis_OptiPyruvate: MODEL5662425708v0.0.1

This model originates from BioModels Database: A Database of Annotated Published Models (http://www.ebi.ac.uk/biomodels/…

Details

We present a genome-scale metabolic model for the archaeal methanogen Methanosarcina barkeri. We characterize the metabolic network and compare it to reconstructions from the prokaryotic, eukaryotic and archaeal domains. Using the model in conjunction with constraint-based methods, we simulate the metabolic fluxes and resulting phenotypes induced by different environmental and genetic conditions. This represents the first large-scale simulation of either a methanogen or an archaeal species. Model predictions are validated by comparison to experimental growth measurements and phenotypes of M. barkeri on different substrates. The predicted growth phenotypes for wild type and mutants of the methanogenic pathway have a high level of agreement with experimental findings. We further examine the efficiency of the energy-conserving reactions in the methanogenic pathway, specifically the Ech hydrogenase reaction, and determine a stoichiometry for the nitrogenase reaction. This work demonstrates that a reconstructed metabolic network can serve as an analysis platform to predict cellular phenotypes, characterize methanogenic growth, improve the genome annotation and further uncover the metabolic characteristics of methanogenesis. link: http://identifiers.org/pubmed/16738551

Feist2007_EcMetabol_flux1: MODEL3023609334v0.0.1

organism E. coli K-12 MG1655 model iAF1260 Biomass Objective Function (BOF) Ec_biomass_iAF1260_core_59p81M (E. coli biom…

Details

An updated genome-scale reconstruction of the metabolic network in Escherichia coli K-12 MG1655 is presented. This updated metabolic reconstruction includes: (1) an alignment with the latest genome annotation and the metabolic content of EcoCyc leading to the inclusion of the activities of 1260 ORFs, (2) characterization and quantification of the biomass components and maintenance requirements associated with growth of E. coli and (3) thermodynamic information for the included chemical reactions. The conversion of this metabolic network reconstruction into an in silico model is detailed. A new step in the metabolic reconstruction process, termed thermodynamic consistency analysis, is introduced, in which reactions were checked for consistency with thermodynamic reversibility estimates. Applications demonstrating the capabilities of the genome-scale metabolic model to predict high-throughput experimental growth and gene deletion phenotypic screens are presented. The increased scope and computational capability using this new reconstruction is expected to broaden the spectrum of both basic biology and applied systems biology studies of E. coli metabolism. link: http://identifiers.org/pubmed/17593909

Feist2007_EcMetabol_flux2: MODEL3023641273v0.0.1

organism E. coli K-12 MG1655 model iAF1260 Biomass Objective Function (BOF) Ec_biomass_iAF1260_core_59p81M (E. coli biom…

Details

An updated genome-scale reconstruction of the metabolic network in Escherichia coli K-12 MG1655 is presented. This updated metabolic reconstruction includes: (1) an alignment with the latest genome annotation and the metabolic content of EcoCyc leading to the inclusion of the activities of 1260 ORFs, (2) characterization and quantification of the biomass components and maintenance requirements associated with growth of E. coli and (3) thermodynamic information for the included chemical reactions. The conversion of this metabolic network reconstruction into an in silico model is detailed. A new step in the metabolic reconstruction process, termed thermodynamic consistency analysis, is introduced, in which reactions were checked for consistency with thermodynamic reversibility estimates. Applications demonstrating the capabilities of the genome-scale metabolic model to predict high-throughput experimental growth and gene deletion phenotypic screens are presented. The increased scope and computational capability using this new reconstruction is expected to broaden the spectrum of both basic biology and applied systems biology studies of E. coli metabolism. link: http://identifiers.org/pubmed/17593909

Feizabadi2011/1 - immunodeficiency in cancer core model: BIOMD0000000760v0.0.1

The paper describes a basic model of immune-cancer interaction. Created by COPASI 4.25 (Build 207) This model is desc…

Details

In this paper, we develop a theoretical contribution towards the understanding of the complex behavior of conjoint tumor-normal cell growth under the influence of immuno-chemotherapeutic agents under simple immune system response. In particular, we consider a core model for the interaction of tumor cells with the surrounding normal cells. We then add the effects of a simple immune system, and both immune-suppression factors and immuno-chemotherapeutic agents as well. Through a series of numerical simulations, we illustrate that the interdependency of tumor-normal cells, together with choice of drug and the nature of the immunodeficiency, leads to a variety of interesting patterns in the evolution of both the tumor and the normal cell populations. link: http://identifiers.org/pubmed/21647303

Parameters:

NameDescription
t = 300000.0 1; k = 0.028 1Reaction: => N; T, Rate Law: tumor_microenvironment*k*T*(1-T/t)
r0 = 1.0 1; r1 = 1000.0 1; b = 1.0 1Reaction: T => ; N, Rate Law: tumor_microenvironment*b*r0*N/(r1+N)
rt = 0.3 1; kt = 1200000.0 1Reaction: => T, Rate Law: tumor_microenvironment*rt*T*(1-T/kt)
rn = 0.4 1; kn = 1000000.0 1Reaction: => N, Rate Law: tumor_microenvironment*rn*N*(1-N/kn)

States:

NameDescription
T[malignant cell]
N[cell]

FelixGarza2017 - Blue Light Treatment of Psoriasis (simplified): BIOMD0000000695v0.0.1

FelixGarza2017 - Blue Light Treatment of Psoriasis (simplified)This model is described in the article: [A Dynamic Model…

Details

Clinical investigations prove that blue light irradiation reduces the severity of psoriasis vulgaris. Nevertheless, the mechanisms involved in the management of this condition remain poorly defined. Despite the encouraging results of the clinical studies, no clear guidelines are specified in the literature for the irradiation scheme regime of blue light-based therapy for psoriasis. We investigated the underlying mechanism of blue light irradiation of psoriatic skin, and tested the hypothesis that regulation of proliferation is a key process. We implemented a mechanistic model of cellular epidermal dynamics to analyze whether a temporary decrease of keratinocytes hyper-proliferation can explain the outcome of phototherapy with blue light. Our results suggest that the main effect of blue light on keratinocytes impacts the proliferative cells. They show that the decrease in the keratinocytes proliferative capacity is sufficient to induce a transient decrease in the severity of psoriasis. To study the impact of the therapeutic regime on the efficacy of psoriasis treatment, we performed simulations for different combinations of the treatment parameters, i.e., length of treatment, fluence (also referred to as dose), and intensity. These simulations indicate that high efficacy is achieved by regimes with long duration and high fluence levels, regardless of the chosen intensity. Our modeling approach constitutes a framework for testing diverse hypotheses on the underlying mechanism of blue light-based phototherapy, and for designing effective strategies for the treatment of psoriasis. link: http://identifiers.org/pubmed/28184200

Parameters:

NameDescription
k4 = 0.0556Reaction: xFinal_4 => xFinal_5, Rate Law: compartmentOne*k4*xFinal_4/compartmentOne
apopFBL = 0.0; k3 = 0.216; AIh = 0.0012Reaction: xFinal_3 =>, Rate Law: compartmentOne*(k3*xFinal_3*AIh/(1-AIh)+apopFBL*xFinal_3)/compartmentOne
apopFBL = 0.0; alpha = 0.0714Reaction: xFinal_6 =>, Rate Law: compartmentOne*(alpha*xFinal_6+apopFBL*xFinal_6)/compartmentOne
rhoTA = 4.0; k2s = 0.0173Reaction: xFinal_8 => xFinal_9, Rate Law: Psoriatic*k2s*rhoTA*xFinal_8/compartmentOne
Pscmax = 4500.0; lambda = 3.5; bProl = -0.003404; doseBL = 52.11; aProl = 1.0; rhoSC = 4.0; gamma1h = 0.0033Reaction: xFinal_6 + xFinal_7 => xFinal_6, Rate Law: aProl*gamma1h*rhoSC*exp(bProl*doseBL)*xFinal_7*xFinal_6/(lambda*Pscmax)/compartmentOne
apopFBL = 0.0; alpha = 0.0714; rhoDe = 4.0Reaction: xFinal_12 =>, Rate Law: Psoriatic*(alpha*rhoDe*xFinal_12+apopFBL*xFinal_12)/compartmentOne
apopFBL = 0.0; k4 = 0.0556; AIh = 0.0012Reaction: xFinal_4 =>, Rate Law: compartmentOne*(k4*xFinal_4*AIh/(1-AIh)+apopFBL*xFinal_4)/compartmentOne
k2a = 0.138Reaction: xFinal_2 => xFinal_2 + xFinal_3, Rate Law: compartmentOne*k2a*xFinal_2/compartmentOne
k2a = 0.138; rhoTA = 4.0Reaction: xFinal_8 => xFinal_8 + xFinal_9, Rate Law: Psoriatic*k2a*rhoTA*xFinal_8/compartmentOne
apopFBL = 0.0; AId = 3.5E-4; rhoTA = 4.0; k2s = 0.0173Reaction: xFinal_8 =>, Rate Law: Psoriatic*(AId*k2s*xFinal_8*rhoTA/(1-AId)+apopFBL*xFinal_8)/compartmentOne
apopFBL = 0.0; k1sh = 0.00164; AIh = 0.0012Reaction: xFinal_1 =>, Rate Law: compartmentOne*(k1sh*xFinal_1*AIh/(1-AIh)+apopFBL*xFinal_1)/compartmentOne
k5 = 0.111Reaction: xFinal_5 => xFinal_6, Rate Law: compartmentOne*k5*xFinal_5/compartmentOne
bProl = -0.003404; doseBL = 52.11; Ptah = 11184.7844353585; aProl = 1.0; k1sh = 0.00164; Pscmax = 4500.0; n = 3.0; gamma1h = 0.0033; omega = 100.0Reaction: xFinal_1 + xFinal_2 + xFinal_8 => xFinal_2 + xFinal_8, Rate Law: (gamma1h*aProl*exp(bProl*doseBL)*xFinal_1*omega*xFinal_1/(1+(omega-1)*((xFinal_2+xFinal_8)/Ptah)^n)/Pscmax+k1sh*xFinal_1*omega/(1+(omega-1)*((xFinal_2+xFinal_8)/Ptah)^n))/compartmentOne
k1sh = 0.00164; rhoSC = 4.0Reaction: xFinal_7 => xFinal_8, Rate Law: Psoriatic*k1sh*rhoSC*xFinal_7/compartmentOne
n = 3.0; Ptah = 11184.7844353585; k1sh = 0.00164; omega = 100.0Reaction: xFinal_1 + xFinal_2 + xFinal_8 => xFinal_1 + xFinal_2 + xFinal_8, Rate Law: omega*xFinal_1*k1sh/(1+(omega-1)*((xFinal_2+xFinal_8)/Ptah)^n)/compartmentOne
rhoTr = 5.0; apopFBL = 0.0; k4 = 0.0556; AId = 3.5E-4Reaction: xFinal_10 =>, Rate Law: Psoriatic*(AId*k4*xFinal_10*rhoTr/(1-AId)+apopFBL*xFinal_10)/compartmentOne
rhoTr = 5.0; apopFBL = 0.0; k3 = 0.216; AId = 3.5E-4Reaction: xFinal_9 =>, Rate Law: Psoriatic*(AId*k3*xFinal_9*rhoTr/(1-AId)+apopFBL*xFinal_9)/compartmentOne
k1ah = 0.0131; rhoSC = 4.0Reaction: xFinal_7 => xFinal_7 + xFinal_8, Rate Law: Psoriatic*k1ah*rhoSC*xFinal_7/compartmentOne
gamma2 = 0.014; bProl = -0.003404; doseBL = 52.11; aProl = 1.0Reaction: xFinal_2 => xFinal_2, Rate Law: compartmentOne*aProl*gamma2*exp(bProl*doseBL)*xFinal_2/compartmentOne
apopFBL = 0.0; k5 = 0.111; AIh = 0.0012Reaction: xFinal_5 =>, Rate Law: compartmentOne*(k5*xFinal_5*AIh/(1-AIh)+apopFBL*xFinal_5)/compartmentOne
gamma2 = 0.014; bProl = -0.003404; doseBL = 52.11; aProl = 1.0; rhoTA = 4.0Reaction: xFinal_8 => xFinal_8, Rate Law: Psoriatic*aProl*aProl*gamma2*rhoTA*exp(bProl*doseBL)*exp(bProl*doseBL)*xFinal_8/compartmentOne
n = 3.0; bProl = -0.003404; doseBL = 52.11; Ptah = 11184.7844353585; aProl = 1.0; gamma1h = 0.0033; omega = 100.0Reaction: xFinal_1 + xFinal_2 + xFinal_8 => xFinal_1 + xFinal_2 + xFinal_8, Rate Law: aProl*gamma1h*exp(bProl*doseBL)*xFinal_1*omega/(1+(omega-1)*((xFinal_2+xFinal_8)/Ptah)^n)/compartmentOne
k2s = 0.0173Reaction: xFinal_2 => xFinal_3, Rate Law: compartmentOne*k2s*xFinal_2/compartmentOne
Pscmax = 4500.0; n = 3.0; lambda = 3.5; bProl = -0.003404; doseBL = 52.11; Ptah = 11184.7844353585; aProl = 1.0; gamma1h = 0.0033; omega = 100.0Reaction: xFinal_1 + xFinal_2 + xFinal_7 + xFinal_8 => xFinal_2 + xFinal_7 + xFinal_8, Rate Law: gamma1h*aProl*exp(bProl*doseBL)*xFinal_1*omega*xFinal_7/(1+(omega-1)*((xFinal_2+xFinal_8)/Ptah)^n)/lambda/Pscmax/compartmentOne
bProl = -0.003404; doseBL = 52.11; aProl = 1.0; rhoSC = 4.0; gamma1h = 0.0033Reaction: xFinal_7 => xFinal_7, Rate Law: Psoriatic*aProl*gamma1h*rhoSC*exp(bProl*doseBL)*xFinal_7/compartmentOne
n = 3.0; k1ah = 0.0131; Ptah = 11184.7844353585; omega = 100.0Reaction: xFinal_1 + xFinal_2 + xFinal_8 => xFinal_1 + xFinal_2 + xFinal_8, Rate Law: omega*xFinal_1*k1ah/(1+(omega-1)*((xFinal_2+xFinal_8)/Ptah)^n)/compartmentOne
rhoTr = 5.0; k3 = 0.216Reaction: xFinal_9 => xFinal_10, Rate Law: Psoriatic*k3*rhoTr*xFinal_9/compartmentOne
rhoTr = 5.0; k4 = 0.0556Reaction: xFinal_10 => xFinal_12, Rate Law: Psoriatic*k4*rhoTr*xFinal_10/compartmentOne
Kp = 6.0; apopFBL = 0.0; bProl = -0.003404; doseBL = 52.11; aProl = 1.0; k1sh = 0.00164; Pscmax = 4500.0; Ka = 392.772887665773; lambda = 3.5; rhoSC = 4.0; AId = 3.5E-4; gamma1h = 0.0033Reaction: xFinal_7 =>, Rate Law: Psoriatic*(aProl*gamma1h*rhoSC*exp(bProl*doseBL)*xFinal_7*xFinal_7/(lambda*Pscmax)+AId*k1sh*xFinal_7*rhoSC/(1-AId)+apopFBL*xFinal_7+Kp*1/(Ka^2+xFinal_7^2)*xFinal_7^2)/compartmentOne
km1 = 1.0E-6Reaction: xFinal_2 => xFinal_1, Rate Law: compartmentOne*km1*xFinal_2/compartmentOne
apopFBL = 0.0; AIh = 0.0012; k2s = 0.0173Reaction: xFinal_2 =>, Rate Law: compartmentOne*(k2s*xFinal_2*AIh/(1-AIh)+apopFBL*xFinal_2)/compartmentOne
km2 = 1.0E-6Reaction: xFinal_3 => xFinal_2, Rate Law: compartmentOne*km2*xFinal_3/compartmentOne
k3 = 0.216Reaction: xFinal_3 => xFinal_4, Rate Law: compartmentOne*k3*xFinal_3/compartmentOne

States:

NameDescription
xFinal 3[keratinocyte; cell cycle arrest]
xFinal 5[keratinocyte; granular cell of epidermis]
xFinal 4[keratinocyte]
xFinal 12[keratinocyte; corneocyte]
xFinal 9[keratinocyte; cell cycle arrest]
xFinal 1[keratinocyte; stem cell]
xFinal 8[keratinocyte]
xFinal 7[keratinocyte; stem cell]
xFinal 2[keratinocyte]
xFinal 10[keratinocyte]
xFinal 6[keratinocyte; corneocyte]

Fenton1998_MyocardiumVortexDynamics: MODEL0911989198v0.0.1

This a model from the article: Vortex dynamics in three-dimensional continuous myocardium with fiber rotation: Filamen…

Details

Wave propagation in ventricular muscle is rendered highly anisotropic by the intramural rotation of the fiber. This rotational anisotropy is especially important because it can produce a twist of electrical vortices, which measures the rate of rotation (in degree/mm) of activation wavefronts in successive planes perpendicular to a line of phase singularity, or filament. This twist can then significantly alter the dynamics of the filament. This paper explores this dynamics via numerical simulation. After a review of the literature, we present modeling tools that include: (i) a simplified ionic model with three membrane currents that approximates well the restitution properties and spiral wave behavior of more complex ionic models of cardiac action potential (Beeler-Reuter and others), and (ii) a semi-implicit algorithm for the fast solution of monodomain cable equations with rotational anisotropy. We then discuss selected results of a simulation study of vortex dynamics in a parallelepipedal slab of ventricular muscle of varying wall thickness (S) and fiber rotation rate (theta(z)). The main finding is that rotational anisotropy generates a sufficiently large twist to destabilize a single transmural filament and cause a transition to a wave turbulent state characterized by a high density of chaotically moving filaments. This instability is manifested by the propagation of localized disturbances along the filament and has no previously known analog in isotropic excitable media. These disturbances correspond to highly twisted and distorted regions of filament, or "twistons," that create vortex rings when colliding with the natural boundaries of the ventricle. Moreover, when sufficiently twisted, these rings expand and create additional filaments by further colliding with boundaries. This instability mechanism is distinct from the commonly invoked patchy failure or wave breakup that is not observed here during the initial instability. For modified Beeler-Reuter-like kinetics with stable reentry in two dimensions, decay into turbulence occurs in the left ventricle in about one second above a critical wall thickness in the range of 4-6 mm that matches experiment. However this decay is suppressed by uniformly decreasing excitability. Specific experiments to test these results, and a method to characterize the filament density during fibrillation are discussed. Results are contrasted with other mechanisms of fibrillation and future prospects are summarized. (c)1998 American Institute of Physics. link: http://identifiers.org/pubmed/12779708

Fernandez2006_ModelA: BIOMD0000000152v0.0.1

To the extent possible under law, all copyright and related or neighbouring rights to this encoded model have been dedic…

Details

Integration of neurotransmitter and neuromodulator signals in the striatum plays a central role in the functions and dysfunctions of the basal ganglia. DARPP-32 is a key actor of this integration in the GABAergic medium-size spiny neurons, in particular in response to dopamine and glutamate. When phosphorylated by cAMP-dependent protein kinase (PKA), DARPP-32 inhibits protein phosphatase-1 (PP1), whereas when phosphorylated by cyclin-dependent kinase 5 (CDK5) it inhibits PKA. DARPP-32 is also regulated by casein kinases and by several protein phosphatases. These complex and intricate regulations make simple predictions of DARPP-32 dynamic behaviour virtually impossible. We used detailed quantitative modelling of the regulation of DARPP-32 phosphorylation to improve our understanding of its function. The models included all the combinations of the three best-characterized phosphorylation sites of DARPP-32, their regulation by kinases and phosphatases, and the regulation of those enzymes by cAMP and Ca(2+) signals. Dynamic simulations allowed us to observe the temporal relationships between cAMP and Ca(2+) signals. We confirmed that the proposed regulation of protein phosphatase-2A (PP2A) by calcium can account for the observed decrease of Threonine 75 phosphorylation upon glutamate receptor activation. DARPP-32 is not simply a switch between PP1-inhibiting and PKA-inhibiting states. Sensitivity analysis showed that CDK5 activity is a major regulator of the response, as previously suggested. Conversely, the strength of the regulation of PP2A by PKA or by calcium had little effect on the PP1-inhibiting function of DARPP-32 in these conditions. The simulations showed that DARPP-32 is not only a robust signal integrator, but that its response also depends on the delay between cAMP and calcium signals affecting the response to the latter. This integration did not depend on the concentration of DARPP-32, while the absolute effect on PP1 varied linearly. In silico mutants showed that Ser137 phosphorylation affects the influence of the delay between dopamine and glutamate, and that constitutive phosphorylation in Ser137 transforms DARPP-32 in a quasi-irreversible switch. This work is a first attempt to better understand the complex interactions between cAMP and Ca(2+) regulation of DARPP-32. Progressive inclusion of additional components should lead to a realistic model of signalling networks underlying the function of striatal neurons. link: http://identifiers.org/pubmed/17194217

Parameters:

NameDescription
kcat28=3.0Reaction: D34_75_137_PP2C => D34_75 + PP2C, Rate Law: Spine*D34_75_137_PP2C*kcat28
kcat44=10.0Reaction: cAMP_PDE => AMP + PDE, Rate Law: Spine*cAMP_PDE*kcat44
kon29=3.0E7Reaction: CK1P + PP2B => CK1P_PP2B, Rate Law: Spine*CK1P*PP2B*kon29
kon7=4400000.0Reaction: D75 + CK1 => D75CK1, Rate Law: Spine*D75*CK1*kon7
kon36=3.0E15Reaction: PP2BinactiveCa2 + Ca => PP2B, Rate Law: Spine*PP2BinactiveCa2*Ca*Ca*kon36
kon38=5.4E7Reaction: cAMP_R2C2 + cAMP => cAMP2_R2C2, Rate Law: Spine*cAMP_R2C2*cAMP*kon38
kon4=5600000.0Reaction: D34 + CDK5 => D34_CDK5, Rate Law: Spine*D34*CDK5*kon4
kcat17=4.0Reaction: D34_75_PP2B => D75 + PP2B, Rate Law: Spine*D34_75_PP2B*kcat17
koff39=110.0Reaction: cAMP3_R2C2 => cAMP2_R2C2 + cAMP, Rate Law: Spine*cAMP3_R2C2*koff39
koff37=33.0Reaction: cAMP_R2C2 => R2C2 + cAMP, Rate Law: Spine*cAMP_R2C2*koff37
kon43=60.0Reaction: cAMP4_R2C => cAMP4_R2 + PKA, Rate Law: Spine*cAMP4_R2C*kon43
koff9=24.0Reaction: D75_PP2A => D75 + PP2A, Rate Law: Spine*D75_PP2A*koff9
koff5=12.0Reaction: D34_CK1 => D34 + CK1, Rate Law: Spine*D34_CK1*koff5
kcat34=5.0Reaction: PP2AP => PP2A, Rate Law: Spine*PP2AP*kcat34
kcat6=4.0Reaction: D34_PP2B => D + PP2B, Rate Law: Spine*D34_PP2B*kcat6
kcat5=3.0Reaction: D34_CK1 => D34_137 + CK1, Rate Law: Spine*D34_CK1*kcat5
kcat29=6.0Reaction: CK1P_PP2B => CK1 + PP2B, Rate Law: Spine*CK1P_PP2B*kcat29
koff36=1.0Reaction: PP2B => PP2BinactiveCa2 + Ca, Rate Law: Spine*PP2B*koff36
kon27=75000.0Reaction: D34_75_137 + PP2B => D34_75_137_PP2B, Rate Law: Spine*D34_75_137*PP2B*kon27
kon39=7.5E7Reaction: cAMP2_R2C2 + cAMP => cAMP3_R2C2, Rate Law: Spine*cAMP2_R2C2*cAMP*kon39
kcat14=3.0Reaction: D34_75_CK1 => D34_75_137 + CK1, Rate Law: Spine*D34_75_CK1*kcat14
kon14=4400000.0Reaction: D34_75 + CK1 => D34_75_CK1, Rate Law: Spine*D34_75*CK1*kon14
kcat15=10.0Reaction: D34_75_PP2A => D34 + PP2A, Rate Law: Spine*D34_75_PP2A*kcat15
kon3=5600000.0Reaction: D + PKA => D_PKA, Rate Law: Spine*D*PKA*kon3
koff4=12.0Reaction: D34_CDK5 => D34 + CDK5, Rate Law: Spine*D34_CDK5*koff4
koff27=120.0Reaction: D34_75_137_PP2B => D34_75_137 + PP2B, Rate Law: Spine*D34_75_137_PP2B*koff27
kon19=75000.0Reaction: D34_137 + PP2B => D34_137_PP2B, Rate Law: Spine*D34_137*PP2B*kon19
kon45=5040000.0Reaction: cAMP + PDEP => cAMP_PDEP, Rate Law: Spine*cAMP*PDEP*kon45
kon24=7500000.0Reaction: D75_137 + PP2C => D75_137_PP2C, Rate Law: Spine*D75_137*PP2C*kon24
koff40=32.5Reaction: cAMP4_R2C2 => cAMP3_R2C2 + cAMP, Rate Law: Spine*cAMP4_R2C2*koff40
koff20=12.0Reaction: D34_137_PP2C => D34_137 + PP2C, Rate Law: Spine*D34_137_PP2C*koff20
kcat20=3.0Reaction: D34_137_PP2C => D34 + PP2C, Rate Law: Spine*D34_137_PP2C*kcat20
koff14=12.0Reaction: D34_75_CK1 => D34_75 + CK1, Rate Law: Spine*D34_75_CK1*koff14
koff45=80.0Reaction: cAMP_PDEP => cAMP + PDEP, Rate Law: Spine*cAMP_PDEP*koff45
kcat26=24.0Reaction: D34_75_137_PP2AP => D34_137 + PP2AP, Rate Law: Spine*D34_75_137_PP2AP*kcat26
koff16=40.0Reaction: D34_75_PP2AP => D34_75 + PP2AP, Rate Law: Spine*D34_75_PP2AP*koff16
koff24=12.0Reaction: D75_137_PP2C => D75_137 + PP2C, Rate Law: Spine*D75_137_PP2C*koff24
kcat8=0.0Reaction: D75_PKA => D34_75 + PKA, Rate Law: Spine*D75_PKA*kcat8
kon5=4400000.0Reaction: D34 + CK1 => D34_CK1, Rate Law: Spine*D34*CK1*kon5
kon10=1.7E7Reaction: D75 + PP2AP => D75_PP2AP, Rate Law: Spine*D75*PP2AP*kon10
kon23=1.7E7Reaction: D75_137 + PP2AP => D75_137_PP2AP, Rate Law: Spine*D75_137*PP2AP*kon23
kon15=3800000.0Reaction: D34_75 + PP2A => D34_75_PP2A, Rate Law: Spine*D34_75*PP2A*kon15
kon28=7500000.0Reaction: D34_75_137 + PP2C => D34_75_137_PP2C, Rate Law: Spine*D34_75_137*PP2C*kon28
k57 = 2.5E-8Reaction: Empty => Ca, Rate Law: Spine*k57
kon9=3800000.0Reaction: D75 + PP2A => D75_PP2A, Rate Law: Spine*D75*PP2A*kon9
kcat23=24.0Reaction: D75_137_PP2AP => D137 + PP2AP, Rate Law: Spine*D75_137_PP2AP*kcat23
kcat4=3.0Reaction: D34_CDK5 => D34_75 + CDK5, Rate Law: Spine*D34_CDK5*kcat4
kon17=1.0E7Reaction: D34_75 + PP2B => D34_75_PP2B, Rate Law: Spine*D34_75*PP2B*kon17
kcat16=24.0Reaction: D34_75_PP2AP => D34 + PP2AP, Rate Law: Spine*D34_75_PP2AP*kcat16
koff28=12.0Reaction: D34_75_137_PP2C => D34_75_137 + PP2C, Rate Law: Spine*D34_75_137_PP2C*koff28
kcat10=24.0Reaction: D75_PP2AP => D + PP2AP, Rate Law: Spine*D75_PP2AP*kcat10
kon42=1.8E7Reaction: cAMP4_R2 + PKA => cAMP4_R2C, Rate Law: Spine*cAMP4_R2*PKA*kon42
kcat33=4.0Reaction: PP2A_PKA => PP2AP + PKA, Rate Law: Spine*PP2A_PKA*kcat33
kon1=5600000.0Reaction: D + CDK5 => D_CDK5, Rate Law: Spine*kon1*D*CDK5
k58=1.7Reaction: Ca => Empty, Rate Law: Spine*Ca*k58
koff15=24.0Reaction: D34_75_PP2A => D34_75 + PP2A, Rate Law: Spine*D34_75_PP2A*koff15
kcat24=3.0Reaction: D75_137_PP2C => D75 + PP2C, Rate Law: Spine*D75_137_PP2C*kcat24
koff19=0.12Reaction: D34_137_PP2B => D34_137 + PP2B, Rate Law: Spine*D34_137_PP2B*koff19
koff17=1600.0Reaction: D34_75_PP2B => D34_75 + PP2B, Rate Law: Spine*D34_75_PP2B*koff17
koff26=40.0Reaction: D34_75_137_PP2AP => D34_75_137 + PP2AP, Rate Law: Spine*D34_75_137_PP2AP*koff26
koff1=12.0Reaction: D_CDK5 => D + CDK5, Rate Law: Spine*D_CDK5*koff1
koff29=24.0Reaction: CK1P_PP2B => CK1P + PP2B, Rate Law: Spine*CK1P_PP2B*koff29
koff6=16.0Reaction: D34_PP2B => D34 + PP2B, Rate Law: Spine*D34_PP2B*koff6
kcat1=3.0Reaction: D_CDK5 => D75 + CDK5, Rate Law: Spine*D_CDK5*kcat1
koff44=40.0Reaction: cAMP_PDE => cAMP + PDE, Rate Law: Spine*cAMP_PDE*koff44
kon6=1.0E7Reaction: D34 + PP2B => D34_PP2B, Rate Law: Spine*D34*PP2B*kon6
kcat27=0.03Reaction: D34_75_137_PP2B => D75_137 + PP2B, Rate Law: Spine*D34_75_137_PP2B*kcat27
kcat19=0.03Reaction: D34_137_PP2B => D137 + PP2B, Rate Law: Spine*D34_137_PP2B*kcat19
koff38=33.0Reaction: cAMP2_R2C2 => cAMP_R2C2 + cAMP, Rate Law: Spine*cAMP2_R2C2*koff38
kon11=5600000.0Reaction: D137 + CDK5 => D137_CDK5, Rate Law: Spine*D137*CDK5*kon11
kcat45=20.0Reaction: cAMP_PDEP => AMP + PDEP, Rate Law: Spine*cAMP_PDEP*kcat45
koff41=60.0Reaction: cAMP4_R2C2 => cAMP4_R2C + PKA, Rate Law: Spine*cAMP4_R2C2*koff41

States:

NameDescription
D34 CDK5[Cyclin-dependent-like kinase 5; Protein phosphatase 1 regulatory subunit 1B]
D34 75 CK1[Casein kinase 1, epsilonCasein kinase1 epsilon-2; Protein phosphatase 1 regulatory subunit 1B]
D75[Protein phosphatase 1 regulatory subunit 1B]
D PKA[Protein phosphatase 1 regulatory subunit 1B; PIRSF000582]
cAMP4 R2[protein complex; 3',5'-cyclic AMP; PIRSF000548]
AMP[AMP]
cAMP PDEP[3',5'-cyclic AMP; IPR000396]
cAMP[3',5'-cyclic AMP]
cAMP4 R2C2[3',5'-cyclic AMP; PIRSF000548; PIRSF000582; protein complex]
cAMP4 R2C[3',5'-cyclic AMP; PIRSF000548; PIRSF000582; protein complex]
PP2B[PIRSF000911]
PP2C[IPR015655]
D34 75[Protein phosphatase 1 regulatory subunit 1B]
D75 PP2A[Protein phosphatase 1 regulatory subunit 1B; IPR006186]
cAMP R2C2[protein complex; 3',5'-cyclic AMP; PIRSF000582; PIRSF000548]
cAMP PDE[3',5'-cyclic AMP; IPR000396]
cAMP2 R2C2[3',5'-cyclic AMP; PIRSF000582; PIRSF000548; protein complex]
D34 PP2B[Protein phosphatase 1 regulatory subunit 1B; PIRSF000911]
PKA[PIRSF000582]
D75 PP2AP[Protein phosphatase 1 regulatory subunit 1B; IPR006186]
CDK5[Cyclin-dependent-like kinase 5]
EmptyEmpty
D34 CK1[Casein kinase 1, epsilonCasein kinase1 epsilon-2; Protein phosphatase 1 regulatory subunit 1B]
PP2AP[IPR006186]
D CDK5[Cyclin-dependent-like kinase 5; Protein phosphatase 1 regulatory subunit 1B]
D34[Protein phosphatase 1 regulatory subunit 1B]

Fernandez2006_ModelB: BIOMD0000000153v0.0.1

To the extent possible under law, all copyright and related or neighbouring rights to this encoded model have been dedic…

Details

Integration of neurotransmitter and neuromodulator signals in the striatum plays a central role in the functions and dysfunctions of the basal ganglia. DARPP-32 is a key actor of this integration in the GABAergic medium-size spiny neurons, in particular in response to dopamine and glutamate. When phosphorylated by cAMP-dependent protein kinase (PKA), DARPP-32 inhibits protein phosphatase-1 (PP1), whereas when phosphorylated by cyclin-dependent kinase 5 (CDK5) it inhibits PKA. DARPP-32 is also regulated by casein kinases and by several protein phosphatases. These complex and intricate regulations make simple predictions of DARPP-32 dynamic behaviour virtually impossible. We used detailed quantitative modelling of the regulation of DARPP-32 phosphorylation to improve our understanding of its function. The models included all the combinations of the three best-characterized phosphorylation sites of DARPP-32, their regulation by kinases and phosphatases, and the regulation of those enzymes by cAMP and Ca(2+) signals. Dynamic simulations allowed us to observe the temporal relationships between cAMP and Ca(2+) signals. We confirmed that the proposed regulation of protein phosphatase-2A (PP2A) by calcium can account for the observed decrease of Threonine 75 phosphorylation upon glutamate receptor activation. DARPP-32 is not simply a switch between PP1-inhibiting and PKA-inhibiting states. Sensitivity analysis showed that CDK5 activity is a major regulator of the response, as previously suggested. Conversely, the strength of the regulation of PP2A by PKA or by calcium had little effect on the PP1-inhibiting function of DARPP-32 in these conditions. The simulations showed that DARPP-32 is not only a robust signal integrator, but that its response also depends on the delay between cAMP and calcium signals affecting the response to the latter. This integration did not depend on the concentration of DARPP-32, while the absolute effect on PP1 varied linearly. In silico mutants showed that Ser137 phosphorylation affects the influence of the delay between dopamine and glutamate, and that constitutive phosphorylation in Ser137 transforms DARPP-32 in a quasi-irreversible switch. This work is a first attempt to better understand the complex interactions between cAMP and Ca(2+) regulation of DARPP-32. Progressive inclusion of additional components should lead to a realistic model of signalling networks underlying the function of striatal neurons. link: http://identifiers.org/pubmed/17194217

Parameters:

NameDescription
kon25=3800000.0Reaction: D34_75_137 + PP2A => D34_75_137_PP2A, Rate Law: Spine*D34_75_137*PP2A*kon25
kcat3=2.7Reaction: D_PKA => D34 + PKA, Rate Law: Spine*D_PKA*kcat3
kon7=4400000.0Reaction: D75 + CK1 => D75CK1, Rate Law: Spine*D75*CK1*kon7
koff2=12.0Reaction: D_CK1 => D + CK1, Rate Law: Spine*koff2*D_CK1
kcat50=24.0Reaction: D34_75_PP2APCa => D34 + PP2APCa, Rate Law: Spine*D34_75_PP2APCa*kcat50
kon2=4400000.0Reaction: D + CK1 => D_CK1, Rate Law: Spine*D*CK1*kon2
kon4=5600000.0Reaction: D34 + CDK5 => D34_CDK5, Rate Law: Spine*D34*CDK5*kon4
kon56=200000.0Reaction: PP2AP + Ca => PP2APCa, Rate Law: Spine*PP2AP*Ca*kon56
kcat55=4.0Reaction: PP2ACa_PKA => PP2APCa + PKA, Rate Law: Spine*PP2ACa_PKA*kcat55
kon43=60.0Reaction: cAMP4_R2C => cAMP4_R2 + PKA, Rate Law: Spine*cAMP4_R2C*kon43
koff9=24.0Reaction: D75_PP2A => D75 + PP2A, Rate Law: Spine*D75_PP2A*koff9
kcat34=5.0Reaction: PP2AP => PP2A, Rate Law: Spine*PP2AP*kcat34
koff5=12.0Reaction: D34_CK1 => D34 + CK1, Rate Law: Spine*D34_CK1*koff5
kcat48=10.0Reaction: D75_PP2ACa => D + PP2ACa, Rate Law: Spine*D75_PP2ACa*kcat48
kcat6=4.0Reaction: D34_PP2B => D + PP2B, Rate Law: Spine*D34_PP2B*kcat6
kcat29=6.0Reaction: CK1P_PP2B => CK1 + PP2B, Rate Law: Spine*CK1P_PP2B*kcat29
koff47=6.0Reaction: D34_75_137_PP2ACa => D34_75_137 + PP2ACa, Rate Law: Spine*D34_75_137_PP2ACa*koff47
kcat5=3.0Reaction: D34_CK1 => D34_137 + CK1, Rate Law: Spine*D34_CK1*kcat5
kon54=200000.0Reaction: Ca + PP2A => PP2ACa, Rate Law: Spine*PP2A*Ca*kon54
koff55=16.0Reaction: PP2ACa_PKA => PP2ACa + PKA, Rate Law: Spine*PP2ACa_PKA*koff55
kon26=1.7E7Reaction: D34_75_137 + PP2AP => D34_75_137_PP2AP, Rate Law: Spine*D34_75_137*PP2AP*kon26
kon8=5600000.0Reaction: D75 + PKA => D75_PKA, Rate Law: Spine*D75*PKA*kon8
kcat14=3.0Reaction: D34_75_CK1 => D34_75_137 + CK1, Rate Law: Spine*D34_75_CK1*kcat14
kon3=5600000.0Reaction: D + PKA => D_PKA, Rate Law: Spine*D*PKA*kon3
kcat18=3.0Reaction: D34_137_CDK5 => D34_75_137 + CDK5, Rate Law: Spine*D34_137_CDK5*kcat18
koff4=12.0Reaction: D34_CDK5 => D34 + CDK5, Rate Law: Spine*D34_CDK5*koff4
kon45=5040000.0Reaction: cAMP + PDEP => cAMP_PDEP, Rate Law: Spine*cAMP*PDEP*kon45
kcat22=10.0Reaction: D75_137_PP2A => D137 + PP2A, Rate Law: Spine*D75_137_PP2A*kcat22
kcat26=24.0Reaction: D34_75_137_PP2AP => D34_137 + PP2AP, Rate Law: Spine*D34_75_137_PP2AP*kcat26
koff14=12.0Reaction: D34_75_CK1 => D34_75 + CK1, Rate Law: Spine*D34_75_CK1*koff14
koff45=80.0Reaction: cAMP_PDEP => cAMP + PDEP, Rate Law: Spine*cAMP_PDEP*koff45
koff16=40.0Reaction: D34_75_PP2AP => D34_75 + PP2AP, Rate Law: Spine*D34_75_PP2AP*koff16
koff23=40.0Reaction: D75_137_PP2AP => D75_137 + PP2AP, Rate Law: Spine*D75_137_PP2AP*koff23
kcat47=10.0Reaction: D34_75_137_PP2ACa => D34_137 + PP2ACa, Rate Law: Spine*D34_75_137_PP2ACa*kcat47
kcat2=3.0Reaction: D_CK1 => D137 + CK1, Rate Law: Spine*kcat2*D_CK1
kon5=4400000.0Reaction: D34 + CK1 => D34_CK1, Rate Law: Spine*D34*CK1*kon5
kcat52=24.0Reaction: D75_PP2APCa => D + PP2APCa, Rate Law: Spine*D75_PP2APCa*kcat52
kcat46=10.0Reaction: D34_75_PP2ACa => D34 + PP2ACa, Rate Law: Spine*D34_75_PP2ACa*kcat46
kon10=1.7E7Reaction: D75 + PP2AP => D75_PP2AP, Rate Law: Spine*D75*PP2AP*kon10
koff31=36.0Reaction: PDE_PKA => PDE + PKA, Rate Law: Spine*PDE_PKA*koff31
kcat53=24.0Reaction: D75_137_PP2APCa => D137 + PP2APCa, Rate Law: Spine*D75_137_PP2APCa*kcat53
kon33=1.0E7Reaction: PP2A + PKA => PP2A_PKA, Rate Law: Spine*PP2A*PKA*kon33
kon48=3800000.0Reaction: D75 + PP2ACa => D75_PP2ACa, Rate Law: Spine*D75*PP2ACa*kon48
kcat49=10.0Reaction: D75_137_PP2ACa => D137 + PP2ACa, Rate Law: Spine*D75_137_PP2ACa*kcat49
koff7=12.0Reaction: D75CK1 => D75 + CK1, Rate Law: Spine*D75CK1*koff7
koff11=12.0Reaction: D137_CDK5 => D137 + CDK5, Rate Law: Spine*D137_CDK5*koff11
koff54=1.0Reaction: PP2ACa => PP2A + Ca, Rate Law: Spine*PP2ACa*koff54
kon18=5600000.0Reaction: D34_137 + CDK5 => D34_137_CDK5, Rate Law: Spine*D34_137*CDK5*kon18
kcat13=3.0Reaction: D137_PP2C => D + PP2C, Rate Law: Spine*D137_PP2C*kcat13
koff56=1.0Reaction: PP2APCa => PP2AP + Ca, Rate Law: Spine*PP2APCa*koff56
kcat23=24.0Reaction: D75_137_PP2AP => D137 + PP2AP, Rate Law: Spine*D75_137_PP2AP*kcat23
kcat4=3.0Reaction: D34_CDK5 => D34_75 + CDK5, Rate Law: Spine*D34_CDK5*kcat4
kon12=5600000.0Reaction: D137 + PKA => D137_PKA, Rate Law: Spine*D137*PKA*kon12
kcat16=24.0Reaction: D34_75_PP2AP => D34 + PP2AP, Rate Law: Spine*D34_75_PP2AP*kcat16
koff3=10.8Reaction: D_PKA => D + PKA, Rate Law: Spine*D_PKA*koff3
koff49=6.0Reaction: D75_137_PP2ACa => D75_137 + PP2ACa, Rate Law: Spine*D75_137_PP2ACa*koff49
kcat10=24.0Reaction: D75_PP2AP => D + PP2AP, Rate Law: Spine*D75_PP2AP*kcat10
kon42=1.8E7Reaction: cAMP4_R2 + PKA => cAMP4_R2C, Rate Law: Spine*cAMP4_R2*PKA*kon42
kon1=5600000.0Reaction: D + CDK5 => D_CDK5, Rate Law: Spine*kon1*D*CDK5
koff25=24.0Reaction: D34_75_137_PP2A => D34_75_137 + PP2A, Rate Law: Spine*D34_75_137_PP2A*koff25
koff26=40.0Reaction: D34_75_137_PP2AP => D34_75_137 + PP2AP, Rate Law: Spine*D34_75_137_PP2AP*koff26
kon49=3800000.0Reaction: D75_137 + PP2ACa => D75_137_PP2ACa, Rate Law: Spine*D75_137*PP2ACa*kon49
koff17=1600.0Reaction: D34_75_PP2B => D34_75 + PP2B, Rate Law: Spine*D34_75_PP2B*koff17
koff52=10.0Reaction: D75_PP2APCa => D75 + PP2APCa, Rate Law: Spine*D75_PP2APCa*koff52
koff1=12.0Reaction: D_CDK5 => D + CDK5, Rate Law: Spine*D_CDK5*koff1
koff46=6.0Reaction: D34_75_PP2ACa => D34_75 + PP2ACa, Rate Law: Spine*D34_75_PP2ACa*koff46
kcat1=3.0Reaction: D_CDK5 => D75 + CDK5, Rate Law: Spine*D_CDK5*kcat1
kcat9=10.0Reaction: D75_PP2A => D + PP2A, Rate Law: Spine*D75_PP2A*kcat9
kon52=1.7E7Reaction: D75 + PP2APCa => D75_PP2APCa, Rate Law: Spine*D75*PP2APCa*kon52
kcat30=1.0Reaction: CK1 => CK1P, Rate Law: Spine*CK1*kcat30
kon6=1.0E7Reaction: D34 + PP2B => D34_PP2B, Rate Law: Spine*D34*PP2B*kon6
koff53=10.0Reaction: D75_137_PP2APCa => D75_137 + PP2APCa, Rate Law: Spine*D75_137_PP2APCa*koff53

States:

NameDescription
D137[Protein phosphatase 1 regulatory subunit 1B]
D34 CDK5[Cyclin-dependent-like kinase 5; Protein phosphatase 1 regulatory subunit 1B]
D75 137 PP2ACa[calcium(2+); Protein phosphatase 1 regulatory subunit 1B; IPR006186]
D75[Protein phosphatase 1 regulatory subunit 1B]
D34 75 CK1[Casein kinase 1, epsilonCasein kinase1 epsilon-2; Protein phosphatase 1 regulatory subunit 1B]
cAMP PDEP[3',5'-cyclic AMP; IPR000396]
PP2ACa[IPR006186]
cAMP4 R2[3',5'-cyclic AMP; PIRSF000548]
PP2B[calcium(2+); PIRSF000911]
D75 PP2ACa[calcium(2+); Protein phosphatase 1 regulatory subunit 1B; IPR006186]
D34 75 137 PP2AP[Protein phosphatase 1 regulatory subunit 1B; IPR006186]
D75 PP2A[Protein phosphatase 1 regulatory subunit 1B; IPR006186]
D CK1[Casein kinase 1, epsilonCasein kinase1 epsilon-2; Protein phosphatase 1 regulatory subunit 1B]
D75 PP2AP[Protein phosphatase 1 regulatory subunit 1B; IPR006186]
PKA[PIRSF000582]
D[Protein phosphatase 1 regulatory subunit 1B]
CDK5[Cyclin-dependent-like kinase 5]
D34 75 137 PP2ACa[calcium(2+); Protein phosphatase 1 regulatory subunit 1B; IPR006186]
PP2A[IPR006186]
PP2AP[IPR006186]
D34 CK1[Casein kinase 1, epsilonCasein kinase1 epsilon-2; Protein phosphatase 1 regulatory subunit 1B]
CK1[Casein kinase 1, epsilonCasein kinase1 epsilon-2]
PP2APCa[calcium(2+); IPR006186]
D75 137 PP2AP[Protein phosphatase 1 regulatory subunit 1B; IPR006186]
D CDK5[Cyclin-dependent-like kinase 5; Protein phosphatase 1 regulatory subunit 1B]
D34[Protein phosphatase 1 regulatory subunit 1B]

Ferreira2003_CML_generation2: BIOMD0000000053v0.0.1

The model should reproduce the figure 2F of the article. The equation 7 has been split into equations 7a-7c, in order t…

Details

The Maillard reaction between reducing sugars and amino groups of biomolecules generates complex structures known as AGEs (advanced glycation endproducts). These have been linked to protein modifications found during aging, diabetes and various amyloidoses. To investigate the contribution of alternative routes to the formation of AGEs, we developed a mathematical model that describes the generation of CML [ N(epsilon)-(carboxymethyl)lysine] in the Maillard reaction between glucose and collagen. Parameter values were obtained by fitting published data from kinetic experiments of Amadori compound decomposition and glycoxidation of collagen by glucose. These raw parameter values were subsequently fine-tuned with adjustment factors that were deduced from dynamic experiments taking into account the glucose and phosphate buffer concentrations. The fine-tuned model was used to assess the relative contributions of the reaction between glyoxal and lysine, the Namiki pathway, and Amadori compound degradation to the generation of CML. The model suggests that the glyoxal route dominates, except at low phosphate and high glucose concentrations. The contribution of Amadori oxidation is generally the least significant at low glucose concentrations. Simulations of the inhibition of CML generation by aminoguanidine show that this compound effectively blocks the glyoxal route at low glucose concentrations (5 mM). Model results are compared with literature estimates of the contributions to CML generation by the three pathways. The significance of the dominance of the glyoxal route is discussed in the context of possible natural defensive mechanisms and pharmacological interventions with the goal of inhibiting the Maillard reaction in vivo. link: http://identifiers.org/pubmed/12911334

Parameters:

NameDescription
k5b=0.0017Reaction: Glyoxal =>, Rate Law: compartment*k5b*Glyoxal
k2b=0.0012; p2=0.75Reaction: Amadori => Schiff, Rate Law: compartment*p2*k2b*Amadori
k3=7.92E-7; p7=60.0; ox=1.0Reaction: Schiff =>, Rate Law: compartment*ox*p7*k3*(Schiff/0.25)^0.36
k1b=0.36Reaction: Schiff => Lysine + Glucose, Rate Law: compartment*k1b*Schiff
p4=1.0; k4=8.6E-5; ox=1.0Reaction: Amadori => CML, Rate Law: compartment*ox*p4*k4*Amadori
p5=1.0; ox=1.0; k5=0.019Reaction: Lysine + Glyoxal => CML, Rate Law: compartment*ox*p5*k5*Glyoxal*Lysine
p6=2.7; k3=7.92E-7; ox=1.0Reaction: Schiff => CML, Rate Law: compartment*ox*p6*k3*(Schiff/0.25)^0.36
p1=0.115; k1a=0.09Reaction: Lysine + Glucose => Schiff, Rate Law: compartment*p1*k1a*Glucose*Lysine
p2=0.75; k2a=0.033Reaction: Schiff => Amadori, Rate Law: compartment*p2*k2a*Schiff
p3=1.0; k3=7.92E-7; ox=1.0Reaction: Glucose => Glyoxal, Rate Law: compartment*ox*p3*k3*(Glucose/0.25)^0.36

States:

NameDescription
Glucose[glucose; C00293]
SchiffSchiff
CMLCML
Lysine[lysine]
AmadoriAmadori
Glyoxal[Glyoxal; glyoxal]

ferrel2011 - autonomous biochemical oscillator in cell cycle in Xenopus laevis v2: BIOMD0000000936v0.0.1

Computational modeling and the theory of nonlinear dynamical systems allow one to not simply describe the events of the…

Details

Computational modeling and the theory of nonlinear dynamical systems allow one to not simply describe the events of the cell cycle, but also to understand why these events occur, just as the theory of gravitation allows one to understand why cannonballs fly in parabolic arcs. The simplest examples of the eukaryotic cell cycle operate like autonomous oscillators. Here, we present the basic theory of oscillatory biochemical circuits in the context of the Xenopus embryonic cell cycle. We examine Boolean models, delay differential equation models, and especially ordinary differential equation (ODE) models. For ODE models, we explore what it takes to get oscillations out of two simple types of circuits (negative feedback loops and coupled positive and negative feedback loops). Finally, we review the procedures of linear stability analysis, which allow one to determine whether a given ODE model and a particular set of kinetic parameters will produce oscillations. link: http://identifiers.org/pubmed/21414480

Parameters:

NameDescription
n1 = 8.0; b1 = 1.0; k1 = 0.5Reaction: CDK1_active =>, Rate Law: compartment*b1*CDK1_active^(n1+1)/(k1^n1+CDK1_active^n1)
a1 = 0.1Reaction: => CDK1_active, Rate Law: compartment*a1

States:

NameDescription
CDK1 active[Cyclin-dependent kinase 1-A; active]

Ferrel2011 - Autonomous biochemical oscillator in regulation of CDK1, Plk1, and APC in Xenopus Laevis cell cycle: BIOMD0000000937v0.0.1

Computational modeling and the theory of nonlinear dynamical systems allow one to not simply describe the events of the…

Details

Computational modeling and the theory of nonlinear dynamical systems allow one to not simply describe the events of the cell cycle, but also to understand why these events occur, just as the theory of gravitation allows one to understand why cannonballs fly in parabolic arcs. The simplest examples of the eukaryotic cell cycle operate like autonomous oscillators. Here, we present the basic theory of oscillatory biochemical circuits in the context of the Xenopus embryonic cell cycle. We examine Boolean models, delay differential equation models, and especially ordinary differential equation (ODE) models. For ODE models, we explore what it takes to get oscillations out of two simple types of circuits (negative feedback loops and coupled positive and negative feedback loops). Finally, we review the procedures of linear stability analysis, which allow one to determine whether a given ODE model and a particular set of kinetic parameters will produce oscillations. link: http://identifiers.org/pubmed/21414480

Parameters:

NameDescription
n1 = 8.0; b1 = 3.0; k1 = 0.5Reaction: CDK1_active => ; APC_active, Rate Law: compartment*b1*CDK1_active*APC_active^n1/(k1^n1+APC_active^n1)
a3 = 3.0; n3 = 8.0; k3 = 0.5Reaction: => APC_active; Plk1_active, Rate Law: compartment*a3*(1-APC_active)*Plk1_active^n3/(k3^n3+Plk1_active^n3)
k2 = 0.5; n2 = 8.0; a2 = 3.0Reaction: => Plk1_active; CDK1_active, Rate Law: compartment*a2*(1-Plk1_active)*CDK1_active^n2/(k2^n2+CDK1_active^n2)
b2 = 1.0Reaction: Plk1_active =>, Rate Law: compartment*b2*Plk1_active
a1 = 0.1Reaction: => CDK1_active, Rate Law: compartment*a1
b3 = 1.0Reaction: APC_active =>, Rate Law: compartment*b3*APC_active

States:

NameDescription
APC active[Adenomatous polyposis coli homolog; active]
Plk1 active[Serine/threonine-protein kinase PLK1; active]
CDK1 active[Cyclin-dependent kinase 1-A; active]

Ferrel2011 - Cdk1 and APC regulation in cell cycle in Xenopus laevis: BIOMD0000000935v0.0.1

Mathematical model of the regulation of Cdk1 and APC in cell cycle in Xenopus Laevis

Details

Computational modeling and the theory of nonlinear dynamical systems allow one to not simply describe the events of the cell cycle, but also to understand why these events occur, just as the theory of gravitation allows one to understand why cannonballs fly in parabolic arcs. The simplest examples of the eukaryotic cell cycle operate like autonomous oscillators. Here, we present the basic theory of oscillatory biochemical circuits in the context of the Xenopus embryonic cell cycle. We examine Boolean models, delay differential equation models, and especially ordinary differential equation (ODE) models. For ODE models, we explore what it takes to get oscillations out of two simple types of circuits (negative feedback loops and coupled positive and negative feedback loops). Finally, we review the procedures of linear stability analysis, which allow one to determine whether a given ODE model and a particular set of kinetic parameters will produce oscillations. link: http://identifiers.org/pubmed/21414480

Parameters:

NameDescription
n1 = 8.0; b1 = 3.0; k1 = 0.5Reaction: CDK1_active => ; APC_active, Rate Law: nuclear*b1*CDK1_active*APC_active^n1/(k1^n1+APC_active^n1)
k2 = 0.5; n2 = 8.0; a2 = 3.0Reaction: => APC_active; CDK1_active, Rate Law: nuclear*a2*(1-APC_active)*CDK1_active^n2/(k2^n2+CDK1_active^n2)
a1 = 0.1Reaction: => CDK1_active, Rate Law: nuclear*a1
b2 = 1.0Reaction: APC_active =>, Rate Law: nuclear*b2*APC_active

States:

NameDescription
APC active[Adenomatous polyposis coli homolog; active]
CDK1 active[Cyclin-dependent kinase 1-A; active]

Field1974_Oregonator: BIOMD0000000040v0.0.1

# Field-Noyes Model of BZ Reaction CitationR.J.Field and R.M.Noyes,J.Chem.Phys.60,1877 (1974)DescriptionField Noyes Vers…

Details

The chemical mechanism of Field, Körös, and Noyes for the oscillatory Belousov reaction has been generalized by a model composed of five steps involving three independent chemical intermediates. The behavior of the resulting differential equations has been examined numerically, and it has been shown that the system traces a stable closed trajectory in three dimensional phase space. The same trajectory is attained from other phase points and even from the point corresponding to steady state solution of the differential equations. The model appears to exhibit limit cycle behavior. By stiffly coupling the concentrations of two of the intermediates, the limit cycle model can be simplified to a system described by two independent variables; this coupled system is amenable to analysis by theoretical techniques already developed for such systems. ©1974 American Institute of Physics link: http://identifiers.org/doi/10.1063/1.1681288

Parameters:

NameDescription
k5=1.0Reaction: Ce => Br, Rate Law: Ce*k5*BZ
k3=8000.0Reaction: BrO3 + HBrO2 => Ce + HBrO2, Rate Law: BrO3*HBrO2*k3*BZ
k2=1.6E9Reaction: Br + HBrO2 => HOBr, Rate Law: Br*HBrO2*k2*BZ
k4=4.0E7Reaction: HBrO2 => BrO3 + HOBr, Rate Law: HBrO2^2*k4*BZ
k1=1.34Reaction: Br + BrO3 => HBrO2 + HOBr, Rate Law: Br*BrO3*k1*BZ

States:

NameDescription
CeCe4+
Br[bromide]
BrO3[bromate]
HOBrCe4+
HBrO2Ce4+

Figueredo2013/1 - immunointeraction base model: BIOMD0000000753v0.0.1

The paper describes a basic model of immune-itumor interaction. Created by COPASI 4.25 (Build 207) This model is de…

Details

Many advances in research regarding immuno-interactions with cancer were developed with the help of ordinary differential equation (ODE) models. These models, however, are not effectively capable of representing problems involving individual localisation, memory and emerging properties, which are common characteristics of cells and molecules of the immune system. Agent-based modelling and simulation is an alternative paradigm to ODE models that overcomes these limitations. In this paper we investigate the potential contribution of agent-based modelling and simulation when compared to ODE modelling and simulation. We seek answers to the following questions: Is it possible to obtain an equivalent agent-based model from the ODE formulation? Do the outcomes differ? Are there any benefits of using one method compared to the other? To answer these questions, we have considered three case studies using established mathematical models of immune interactions with early-stage cancer. These case studies were re-conceptualised under an agent-based perspective and the simulation results were then compared with those from the ODE models. Our results show that it is possible to obtain equivalent agent-based models (i.e. implementing the same mechanisms); the simulation output of both types of models however might differ depending on the attributes of the system to be modelled. In some cases, additional insight from using agent-based modelling was obtained. Overall, we can confirm that agent-based modelling is a useful addition to the tool set of immunologists, as it has extra features that allow for simulations with characteristics that are closer to the biological phenomena. link: http://identifiers.org/pubmed/23734575

Parameters:

NameDescription
m = 0.00311 1Reaction: E => ; T, Rate Law: tumor_microenvironment*m*E*T
d = 2.0 1Reaction: E =>, Rate Law: tumor_microenvironment*d*E
a = 1.636 1Reaction: => T, Rate Law: tumor_microenvironment*a*T
a = 1.636 1; b = 0.004 1Reaction: T =>, Rate Law: tumor_microenvironment*a*b*T*T
n = 1.0 1Reaction: T => ; E, Rate Law: tumor_microenvironment*n*T*E
s = 0.318 1Reaction: => E, Rate Law: tumor_microenvironment*s
g = 20.19 1; p = 1.131 1Reaction: => E; T, Rate Law: tumor_microenvironment*p*T*E/(g+T)

States:

NameDescription
T[malignant cell]
E[Effector Immune Cell]

Figueredo2013/2 - immunointeraction model with IL2: BIOMD0000000754v0.0.1

The paper describes a model of immune-itumor interaction with IL2. Created by COPASI 4.25 (Build 207) This model is…

Details

Many advances in research regarding immuno-interactions with cancer were developed with the help of ordinary differential equation (ODE) models. These models, however, are not effectively capable of representing problems involving individual localisation, memory and emerging properties, which are common characteristics of cells and molecules of the immune system. Agent-based modelling and simulation is an alternative paradigm to ODE models that overcomes these limitations. In this paper we investigate the potential contribution of agent-based modelling and simulation when compared to ODE modelling and simulation. We seek answers to the following questions: Is it possible to obtain an equivalent agent-based model from the ODE formulation? Do the outcomes differ? Are there any benefits of using one method compared to the other? To answer these questions, we have considered three case studies using established mathematical models of immune interactions with early-stage cancer. These case studies were re-conceptualised under an agent-based perspective and the simulation results were then compared with those from the ODE models. Our results show that it is possible to obtain equivalent agent-based models (i.e. implementing the same mechanisms); the simulation output of both types of models however might differ depending on the attributes of the system to be modelled. In some cases, additional insight from using agent-based modelling was obtained. Overall, we can confirm that agent-based modelling is a useful addition to the tool set of immunologists, as it has extra features that allow for simulations with characteristics that are closer to the biological phenomena. link: http://identifiers.org/pubmed/23734575

Parameters:

NameDescription
g2 = 100000.0 1; aa = 1.0 1Reaction: T => ; E, Rate Law: tumor_microenvironment*aa*E*T/(g2+T)
u2 = 0.03 1Reaction: E =>, Rate Law: tumor_microenvironment*u2*E
p2 = 5.0 1; g3 = 1000.0 1Reaction: => I; E, T, Rate Law: tumor_microenvironment*p2*E*T/(g3+T)
s2 = 0.0 1Reaction: => I, Rate Law: tumor_microenvironment*s2
a = 0.18 1Reaction: => T, Rate Law: tumor_microenvironment*a*T
c = 0.05 1Reaction: => E; T, Rate Law: tumor_microenvironment*c*T
s1 = 0.0 1Reaction: => E, Rate Law: tumor_microenvironment*s1
g1 = 2.0E7 1; p1 = 0.1245 1Reaction: => E; I, Rate Law: tumor_microenvironment*p1*E*I/(g1+I)
a = 0.18 1; b = 1.0E-9 1Reaction: T =>, Rate Law: tumor_microenvironment*a*b*T*T
u3 = 10.0 1Reaction: I =>, Rate Law: tumor_microenvironment*u3*I

States:

NameDescription
I[Interleukin-2]
T[malignant cell]
E[Effector Immune Cell]

Figueredo2013/3 - immunointeraction full model: BIOMD0000000756v0.0.1

The paper describes a full model of immune-itumor interaction. Created by COPASI 4.25 (Build 207) This model is des…

Details

Many advances in research regarding immuno-interactions with cancer were developed with the help of ordinary differential equation (ODE) models. These models, however, are not effectively capable of representing problems involving individual localisation, memory and emerging properties, which are common characteristics of cells and molecules of the immune system. Agent-based modelling and simulation is an alternative paradigm to ODE models that overcomes these limitations. In this paper we investigate the potential contribution of agent-based modelling and simulation when compared to ODE modelling and simulation. We seek answers to the following questions: Is it possible to obtain an equivalent agent-based model from the ODE formulation? Do the outcomes differ? Are there any benefits of using one method compared to the other? To answer these questions, we have considered three case studies using established mathematical models of immune interactions with early-stage cancer. These case studies were re-conceptualised under an agent-based perspective and the simulation results were then compared with those from the ODE models. Our results show that it is possible to obtain equivalent agent-based models (i.e. implementing the same mechanisms); the simulation output of both types of models however might differ depending on the attributes of the system to be modelled. In some cases, additional insight from using agent-based modelling was obtained. Overall, we can confirm that agent-based modelling is a useful addition to the tool set of immunologists, as it has extra features that allow for simulations with characteristics that are closer to the biological phenomena. link: http://identifiers.org/pubmed/23734575

Parameters:

NameDescription
g4 = 1000.0 1; p3 = 5.0 1; alpha = 0.001 1Reaction: => I; E, T, S, Rate Law: tumor_microenvironment*p3*E*T/((g4+T)*(1+alpha*S))
a = 0.18 1; k = 1.0E10 1Reaction: T =>, Rate Law: tumor_microenvironment*a*T^2/k
g3 = 2.0E7 1; p2 = 0.27 1Reaction: => T; S, Rate Law: tumor_microenvironment*p2*S*T/(g3+S)
g2 = 100000.0 1; aa = 1.0 1Reaction: T => ; E, Rate Law: tumor_microenvironment*aa*E*T/(g2+T)
p4 = 2.84 1; theta = 1000000.0 1Reaction: => S; T, Rate Law: tumor_microenvironment*p4*T^2/(theta^2+T^2)
a = 0.18 1Reaction: => T, Rate Law: tumor_microenvironment*a*T
u2 = 10.0 1Reaction: I =>, Rate Law: tumor_microenvironment*u2*I
gamma = 10.0 1; c = 0.035 1Reaction: => E; T, S, Rate Law: tumor_microenvironment*c*T/(1+gamma*S)
g1 = 2.0E7 1; q1 = 10.0 1; p1 = 0.1245 1; q2 = 0.1121 1Reaction: => E; I, S, Rate Law: tumor_microenvironment*p1*E*I/(g1+I)*(p1-q1*S/(q2+S))
u3 = 10.0 1Reaction: S =>, Rate Law: tumor_microenvironment*u3*S
u1 = 0.03 1Reaction: E =>, Rate Law: tumor_microenvironment*u1*E

States:

NameDescription
I[Interleukin-2]
S[Transforming growth factor beta-1]
T[malignant cell]
E[Effector Immune Cell]

Fink2008_VentricularActionPotential: MODEL1006230009v0.0.1

This a model from the article: Contributions of HERG K+ current to repolarization of the human ventricular action pote…

Details

Action potential repolarization in the mammalian heart is governed by interactions of a number of time- and voltage-dependent channel-mediated currents, as well as contributions from the Na+/Ca2+ exchanger and the Na+/K+ pump. Recent work has shown that one of the K+ currents (HERG) which contributes to repolarization in mammalian ventricle is a locus at which a number of point mutations can have significant functional consequences. In addition, the remarkable sensitivity of this K+ channel isoform to inhibition by a variety of pharmacological agents and clinical drugs has resulted in HERG being a major focus for Safety Pharmacology requirements. For these reasons we and others have attempted to define the functional role for HERG-mediated K+ currents in repolarization of the action potential in the human ventricle. Here, we describe and evaluate changes in the formulations for two K+ currents, IK1 and HERG (or IK,r), within the framework of ten Tusscher model of the human ventricular action potential. In this computational study, new mathematical formulations for the two nonlinear K+ conductances, IK1 and HERG, have been developed based upon experimental data obtained from electrophysiological studies of excised human ventricular tissue and/or myocytes. The resulting mathematical model provides much improved simulations of the relative sizes and time courses of the K+ currents which modulate repolarization. Our new formulation represents an important first step in defining the mechanism(s) of repolarization of the membrane action potential in the human ventricle. Our overall goal is to understand the genesis of the T-wave of the human electrocardiogram. link: http://identifiers.org/pubmed/17919688

Firczuk2013 - Eukaryotic mRNA translation machinery: BIOMD0000000457v0.0.1

Firczuk2013 - Eukaryotic mRNA translation machineryThis is a model of *Saccharomyces cerevisiae* mRNA translation which…

Details

Rate control analysis defines the in vivo control map governing yeast protein synthesis and generates an extensively parameterized digital model of the translation pathway. Among other non-intuitive outcomes, translation demonstrates a high degree of functional modularity and comprises a non-stoichiometric combination of proteins manifesting functional convergence on a shared maximal translation rate. In exponentially growing cells, polypeptide elongation (eEF1A, eEF2, and eEF3) exerts the strongest control. The two other strong control points are recruitment of mRNA and tRNA(i) to the 40S ribosomal subunit (eIF4F and eIF2) and termination (eRF1; Dbp5). In contrast, factors that are found to promote mRNA scanning efficiency on a longer than-average 5'untranslated region (eIF1, eIF1A, Ded1, eIF2B, eIF3, and eIF5) exceed the levels required for maximal control. This is expected to allow the cell to minimize scanning transition times, particularly for longer 5'UTRs. The analysis reveals these and other collective adaptations of control shared across the factors, as well as features that reflect functional modularity and system robustness. Remarkably, gene duplication is implicated in the fine control of cellular protein synthesis. link: http://identifiers.org/pubmed/23340841

Parameters:

NameDescription
k=1.3072E13; parameter_1 = 7.16464328895E-7Reaction: species_34 + species_33 => species_46 + species_11 + species_14 + species_1 + species_7 + species_24 + species_25 + species_17 + species_18 + species_8 + species_31 + species_21 + species_20 + species_29; species_46, species_47, species_48, species_49, species_50, species_51, species_52, species_53, species_54, species_55, species_56, species_57, species_58, species_59, species_60, species_61, species_62, species_63, species_64, species_65, species_66, species_67, species_68, species_69, species_70, species_71, species_72, species_73, species_74, species_75, species_76, species_77, species_78, species_79, species_80, species_81, species_82, species_83, species_84, species_85, species_86, species_87, species_88, species_89, species_90, species_91, species_92, species_93, species_94, species_95, species_96, species_97, species_98, species_99, species_100, species_101, species_102, species_103, species_104, species_105, species_106, species_107, species_108, species_109, species_110, species_111, species_112, species_113, species_114, species_115, species_116, species_117, species_118, species_119, species_120, species_121, species_122, species_123, species_124, species_125, species_126, species_127, species_128, species_129, species_130, species_131, species_132, species_133, species_34, species_33, species_46, species_47, species_48, species_49, species_50, species_51, species_52, species_53, species_54, species_55, species_56, species_57, species_58, species_59, species_60, species_61, species_62, species_63, species_64, species_65, species_66, species_67, species_68, species_69, species_70, species_71, species_72, species_73, species_74, species_75, species_76, species_77, species_78, species_79, species_80, species_81, species_82, species_83, species_84, species_85, species_86, species_87, species_88, species_89, species_90, species_91, species_92, species_93, species_94, species_95, species_96, species_97, species_98, species_99, species_100, species_101, species_102, species_103, species_104, species_105, species_106, species_107, species_108, species_109, species_110, species_111, species_112, species_113, species_114, species_115, species_116, species_117, species_118, species_119, species_120, species_121, species_122, species_123, species_124, species_125, species_126, species_127, species_128, species_129, species_130, species_131, species_132, species_133, Rate Law: compartment_1*k*species_34*species_33*(parameter_1-(species_46+species_47+species_48+species_49+species_50+species_51+species_52+species_53+species_54+species_55+species_56+species_57+species_58+species_59+species_60+species_61+species_62+species_63+species_64+species_65+species_66+species_67+species_68+species_69+species_70+species_71+species_72+species_73+species_74+species_75+species_76+species_77+species_78+species_79+species_80+species_81+species_82+species_83+species_84+species_85+species_86+species_87+species_88+species_89+species_90+species_91+species_92+species_93+species_94+species_95+species_96+species_97+species_98+species_99+species_100+species_101+species_102+species_103+species_104+species_105+species_106+species_107+species_108+species_109+species_110+species_111+species_112+species_113+species_114+species_115+species_116+species_117+species_118+species_119+species_120+species_121+species_122+species_123+species_124+species_125+species_126+species_127+species_128+species_129+species_130+species_131+species_132+species_133))
parameter_2 = 8.10035535716195E9; parameter_3 = 0.284007213965168Reaction: species_40 + species_98 => species_99; species_40, species_98, species_99, Rate Law: compartment_1*(parameter_2*species_40*species_98-parameter_3*species_99)
parameter_6 = 0.00322599; parameter_5 = 3.10377169466493E9Reaction: species_42 + species_94 => species_95; species_42, species_94, species_95, Rate Law: compartment_1*(parameter_5*species_42*species_94-parameter_6*species_95)
k1=1.06204E9Reaction: species_16 + species_27 => species_28; species_16, species_27, Rate Law: compartment_1*k1*species_16*species_27
k1=3.5208E14; k2=0.785013Reaction: species_25 + species_166 => species_27; species_25, species_166, species_27, Rate Law: compartment_1*(k1*species_25*species_166-k2*species_27)
k2=47.8215; k1=5.61005E8Reaction: species_30 + species_32 => species_33; species_30, species_32, species_33, Rate Law: compartment_1*(k1*species_30*species_32-k2*species_33)
k1=1.80542; k2=1.29513Reaction: species_41 => species_42; species_41, species_42, Rate Law: compartment_1*(k1*species_41-k2*species_42)
parameter_7 = 2306950.0; parameter_1 = 7.16464328895E-7Reaction: species_53 => species_60 + species_41; species_56, species_57, species_58, species_59, species_60, species_61, species_62, species_63, species_64, species_65, species_66, species_67, species_68, species_69, species_70, species_71, species_72, species_73, species_74, species_75, species_76, species_77, species_78, species_79, species_80, species_81, species_82, species_83, species_84, species_85, species_86, species_87, species_88, species_89, species_90, species_91, species_92, species_93, species_94, species_95, species_96, species_97, species_98, species_99, species_100, species_101, species_102, species_103, species_104, species_105, species_106, species_107, species_108, species_109, species_110, species_111, species_112, species_113, species_114, species_115, species_116, species_117, species_118, species_119, species_120, species_121, species_122, species_123, species_124, species_125, species_126, species_127, species_128, species_129, species_130, species_131, species_132, species_133, species_134, species_135, species_136, species_137, species_138, species_139, species_140, species_141, species_142, species_143, species_144, species_145, species_53, species_56, species_57, species_58, species_59, species_60, species_61, species_62, species_63, species_64, species_65, species_66, species_67, species_68, species_69, species_70, species_71, species_72, species_73, species_74, species_75, species_76, species_77, species_78, species_79, species_80, species_81, species_82, species_83, species_84, species_85, species_86, species_87, species_88, species_89, species_90, species_91, species_92, species_93, species_94, species_95, species_96, species_97, species_98, species_99, species_100, species_101, species_102, species_103, species_104, species_105, species_106, species_107, species_108, species_109, species_110, species_111, species_112, species_113, species_114, species_115, species_116, species_117, species_118, species_119, species_120, species_121, species_122, species_123, species_124, species_125, species_126, species_127, species_128, species_129, species_130, species_131, species_132, species_133, species_134, species_135, species_136, species_137, species_138, species_139, species_140, species_141, species_142, species_143, species_144, species_145, Rate Law: compartment_1*parameter_7*species_53*(parameter_1-(species_56+species_57+species_58+species_59+species_60+species_61+species_62+species_63+species_64+species_65+species_66+species_67+species_68+species_69+species_70+species_71+species_72+species_73+species_74+species_75+species_76+species_77+species_78+species_79+species_80+species_81+species_82+species_83+species_84+species_85+species_86+species_87+species_88+species_89+species_90+species_91+species_92+species_93+species_94+species_95+species_96+species_97+species_98+species_99+species_100+species_101+species_102+species_103+species_104+species_105+species_106+species_107+species_108+species_109+species_110+species_111+species_112+species_113+species_114+species_115+species_116+species_117+species_118+species_119+species_120+species_121+species_122+species_123+species_124+species_125+species_126+species_127+species_128+species_129+species_130+species_131+species_132+species_133+species_134+species_135+species_136+species_137+species_138+species_139+species_140+species_141+species_142+species_143+species_144+species_145))/(parameter_1-(species_56+species_57+species_58+species_59+species_60+species_61+species_62+species_63+species_64+species_65+species_66+species_67+species_68+species_69+species_70+species_71+species_72+species_73+species_74+species_75+species_76+species_77+species_78+species_79+species_80+species_81+species_82+species_83+species_84+species_85+species_86+species_87+species_88+species_89+species_90+species_91+species_92+species_93+species_94+species_95+species_96+species_97+species_98+species_99+species_100+species_101+species_102+species_103+species_104+species_105+species_106+species_107+species_108+species_109+species_110+species_111+species_112+species_113+species_114+species_115+species_116+species_117+species_118+species_119+species_120+species_121+species_122+species_123+species_124+species_125+species_126+species_127+species_128+species_129+species_130+species_131+species_132+species_133+species_134+species_135+species_136+species_137+species_138+species_139))
parameter_9 = 72911.6740026381Reaction: species_97 => species_92 + species_43 + species_45; species_97, Rate Law: compartment_1*parameter_9*species_97
k1=8.7134E10; k2=1.2395Reaction: species_28 + species_29 => species_30; species_28, species_29, species_30, Rate Law: compartment_1*(k1*species_28*species_29-k2*species_30)
k1=93.5995; k2=43714.4Reaction: species_43 => species_44; species_43, species_44, Rate Law: compartment_1*(k1*species_43-k2*species_44)
k2=45.4082; k1=304.768Reaction: species_31 => species_32; species_31, species_32, Rate Law: compartment_1*(k1*species_31-k2*species_32)
k2=2.70026; k1=5.79912E7Reaction: species_19 + species_22 => species_23; species_19, species_22, species_23, Rate Law: compartment_1*(k1*species_19*species_22-k2*species_23)
k1=4.33274E7; k2=1977.92Reaction: species_17 + species_18 => species_19; species_17, species_18, species_19, Rate Law: compartment_1*(k1*species_17*species_18-k2*species_19)
parameter_8 = 2.24052E9Reaction: species_96 + species_44 => species_97; species_96, species_44, Rate Law: compartment_1*parameter_8*species_96*species_44
k2=0.00774034; k1=5026500.0Reaction: species_20 + species_21 => species_22; species_20, species_21, species_22, Rate Law: compartment_1*(k1*species_20*species_21-k2*species_22)
parameter_4 = 28324.3562938545Reaction: species_99 => species_100 + species_35; species_99, Rate Law: compartment_1*parameter_4*species_99
parameter_7 = 2306950.0Reaction: species_101 => species_108 + species_41; species_101, Rate Law: compartment_1*parameter_7*species_101
k1=1.97254E7Reaction: species_12 + species_15 => species_16; species_12, species_15, Rate Law: compartment_1*k1*species_12*species_15
k1=3865650.0; k2=31.1969Reaction: species_11 + species_10 => species_12; species_11, species_10, species_12, Rate Law: compartment_1*(k1*species_11*species_10-k2*species_12)

States:

NameDescription
species 50[cytosolic ribosome]
species 101[alpha-aminoacyl-tRNA; GTP; Elongation factor 2]
species 27[messenger RNA; ATP-dependent RNA helicase eIF4A; Eukaryotic initiation factor 4F subunit p150; Eukaryotic translation initiation factor 4B; Eukaryotic initiation factor 4F subunit p130]
species 100[alpha-aminoacyl-tRNA; cytosolic ribosome]
species 31[GDP; Eukaryotic translation initiation factor 5B]
species 45[transfer RNA]
species 98[cytosolic ribosome]
species 51[alpha-aminoacyl-tRNA; GTP; Elongation factor 1-alpha; cytosolic ribosome]
species 20[messenger RNA; capped_mRNA]
species 28[eukaryotic 48S preinitiation complex]
species 97[GTP; alpha-aminoacyl-tRNA; Elongation factor 3A; cytosolic ribosome]
species 16[eukaryotic 43S preinitiation complex]
species 61[alpha-aminoacyl-tRNA; GTP; Elongation factor 3A; cytosolic ribosome]
species 60[transfer RNA; cytosolic ribosome]
species 102[transfer RNA; cytosolic ribosome]
species 25[Eukaryotic translation initiation factor 4B]
species 29[ATP-dependent RNA helicase DED1]
species 32[GTP; Eukaryotic translation initiation factor 5B]
species 30[ATP-dependent RNA helicase DED1; eukaryotic 48S preinitiation complex]
species 12[tRNA(Met); GTP; Eukaryotic translation initiation factor 3 subunit C; Eukaryotic translation initiation factor 2 subunit alpha; Eukaryotic translation initiation factor 3 subunit G; Eukaryotic translation initiation factor 3 subunit A; Eukaryotic translation initiation factor 5; Eukaryotic translation initiation factor eIF-1; Eukaryotic translation initiation factor 2 subunit gamma; Eukaryotic translation initiation factor 3 subunit B; Eukaryotic translation initiation factor 3 subunit I; Eukaryotic translation initiation factor 2 subunit beta; multi-eIF complex]
species 17[Eukaryotic translation initiation factor 4E]
species 21[Polyadenylate-binding protein, cytoplasmic and nuclear]
species 94[alpha-aminoacyl-tRNA; cytosolic ribosome]
species 42[GTP; Elongation factor 2]
species 44[GTP; Elongation factor 3A]
species 123[alpha-aminoacyl-tRNA; GTP; Elongation factor 1-alpha; cytosolic ribosome]
species 19[Eukaryotic initiation factor 4F subunit p150; Eukaryotic initiation factor 4F subunit p130; Eukaryotic translation initiation factor 4E]
species 129[alpha-aminoacyl-tRNA; GTP; Elongation factor 1-alpha; cytosolic ribosome]
species 11[Eukaryotic translation initiation factor eIF-1]
species 96[transfer RNA; cytosolic ribosome]
species 24[ATP-dependent RNA helicase eIF4A]
species 77[alpha-aminoacyl-tRNA; GTP; Elongation factor 2]
species 95[GTP; alpha-aminoacyl-tRNA; Elongation factor 2]
species 43[GDP; Elongation factor 3A]
species 130[alpha-aminoacyl-tRNA; cytosolic ribosome]
species 122[cytosolic ribosome]
species 41[GDP; Elongation factor 2]
species 99[GTP; alpha-aminoacyl-tRNA; Elongation factor 1-alpha; cytosolic ribosome]
species 46[cytosolic ribosome; translation initiation complex]
species 40[GTP; alpha-aminoacyl-tRNA; Elongation factor 1-alpha]
species 65[GTP; alpha-aminoacyl-tRNA; Elongation factor 2]

Fisher2006_Ca_Oscillation_dpdnt_NFAT_dynamics: BIOMD0000000122v0.0.1

The model reproduces the calcium oscillation dependent activation-deactivation kinetics of nuclear factor of activated T…

Details

Mathematical models for the regulation of the Ca(2+)-dependent transcription factors NFAT and NFkappaB that are involved in the activation of the immune and inflammatory responses in T lymphocytes have been developed. These pathways are important targets for drugs, which act as powerful immunosuppressants by suppressing activation of NFAT and NFkappaB in T cells. The models simulate activation and deactivation over physiological concentrations of Ca(2+), diacyl glycerol (DAG), and PKCtheta using single and periodic step increases. The model suggests the following: (1) the activation NFAT does not occur at low frequencies as NFAT requires calcineurin activated by Ca(2+) to remain dephosphorylated and in the nucleus; (2) NFkappaB is activated at lower Ca(2+) oscillation frequencies than NFAT as IkappaB is degraded in response to elevations in Ca(2+) allowing free NFkappaB to translocate into the nucleus; and (3) the degradation of IkappaB is essential for efficient translocation of NFkappaB to the nucleus. Through sensitivity analysis, the model also suggests that the largest controlling factor for NFAT activation is the dissociation/reassociation rate of the NFAT:calcineurin complex and the translocation rate of the complex into the nucleus and for NFkappaB is the degradation/resynthesis rate of IkappaB and the import rate of IkappaB into the nucleus. link: http://identifiers.org/pubmed/17031595

Parameters:

NameDescription
k19 = 1.0; k20 = 1.0Reaction: Ca_Nuc + Inact_C_Nuc => Act_C_Nuc, Rate Law: nucleus*(k19*Inact_C_Nuc*Ca_Nuc^3-k20*Act_C_Nuc)
k8 = 0.5; k7 = 0.005Reaction: NFAT_Pi_Act_C_Cyt => NFAT_Pi_Act_C_Nuc, Rate Law: cytosol*k7*NFAT_Pi_Act_C_Cyt-nucleus*k8*NFAT_Pi_Act_C_Nuc
k1 = 2.56E-5; k2 = 0.00256Reaction: NFAT_Pi_Nuc + Act_C_Nuc => Act_C_Nuc + NFAT_Nuc, Rate Law: nucleus*(k1*NFAT_Pi_Nuc-k2*NFAT_Nuc)
k17 = 0.0015; k18 = 9.6E-4Reaction: NFAT_Nuc => NFAT_Cyt, Rate Law: nucleus*k18*NFAT_Nuc-cytosol*k17*NFAT_Cyt
k5 = 0.0019; k6 = 9.2E-4Reaction: Inact_C_Cyt => Inact_C_Nuc, Rate Law: cytosol*k5*Inact_C_Cyt-nucleus*k6*Inact_C_Nuc
k12 = 0.00168; k11 = 6.63Reaction: NFAT_Pi_Act_C_Cyt => Act_C_Cyt + NFAT_Pi_Cyt, Rate Law: cytosol*(k12*NFAT_Pi_Act_C_Cyt-k11*NFAT_Pi_Cyt*Act_C_Cyt)
k9 = 0.5; k10 = 0.005Reaction: NFAT_Act_C_Nuc => NFAT_Act_C_Cyt, Rate Law: nucleus*k10*NFAT_Act_C_Nuc-cytosol*k9*NFAT_Act_C_Cyt
k3 = 0.005; k4 = 0.5Reaction: NFAT_Pi_Cyt => NFAT_Pi_Nuc, Rate Law: cytosol*k3*NFAT_Pi_Cyt-nucleus*k4*NFAT_Pi_Nuc
k21 = 0.21; k22 = 0.5Reaction: Ca_Cyt => Ca_Nuc, Rate Law: cytosol*k21*Ca_Cyt-nucleus*k22*Ca_Nuc
k15 = 0.00168; k16 = 6.63Reaction: Act_C_Nuc + NFAT_Nuc => NFAT_Act_C_Nuc, Rate Law: nucleus*(k16*NFAT_Nuc*Act_C_Nuc-k15*NFAT_Act_C_Nuc)
k13 = 0.5; k14 = 0.00256Reaction: NFAT_Act_C_Cyt => NFAT_Pi_Act_C_Cyt, Rate Law: cytosol*(k14*NFAT_Act_C_Cyt-k13*NFAT_Pi_Act_C_Cyt)

States:

NameDescription
Act C Cyt[Calcineurin B homologous protein 1]
NFAT Act C Nuc[Calcineurin B homologous protein 1; Nuclear factor of activated T-cells 5]
NFAT Pi Act C Nuc[Calcineurin B homologous protein 1; Nuclear factor of activated T-cells 5]
Inact C Cyt[Calcineurin B homologous protein 1]
NFAT Act C Cyt[Calcineurin B homologous protein 1; Nuclear factor of activated T-cells 5]
NFAT Pi Nuc[Nuclear factor of activated T-cells 5]
NFAT Pi Act C Cyt[Calcineurin B homologous protein 1; Nuclear factor of activated T-cells 5]
Inact C Nuc[Calcineurin B homologous protein 1]
NFAT Pi Cyt[Nuclear factor of activated T-cells 5]
Ca Nuc[calcium(2+); Calcium cation]
NFAT Cyt[Nuclear factor of activated T-cells 5]
Act C Nuc[Calcineurin B homologous protein 1]
NFAT Nuc[Nuclear factor of activated T-cells 5]
Ca Cyt[calcium(2+); Calcium cation]

Fisher2006_NFAT_Activation: BIOMD0000000123v0.0.1

The model reproduces the kinetics of the nuclear factor of activated cells (NFAT) as depicted in Figure 3a of the paper.…

Details

Mathematical models for the regulation of the Ca(2+)-dependent transcription factors NFAT and NFkappaB that are involved in the activation of the immune and inflammatory responses in T lymphocytes have been developed. These pathways are important targets for drugs, which act as powerful immunosuppressants by suppressing activation of NFAT and NFkappaB in T cells. The models simulate activation and deactivation over physiological concentrations of Ca(2+), diacyl glycerol (DAG), and PKCtheta using single and periodic step increases. The model suggests the following: (1) the activation NFAT does not occur at low frequencies as NFAT requires calcineurin activated by Ca(2+) to remain dephosphorylated and in the nucleus; (2) NFkappaB is activated at lower Ca(2+) oscillation frequencies than NFAT as IkappaB is degraded in response to elevations in Ca(2+) allowing free NFkappaB to translocate into the nucleus; and (3) the degradation of IkappaB is essential for efficient translocation of NFkappaB to the nucleus. Through sensitivity analysis, the model also suggests that the largest controlling factor for NFAT activation is the dissociation/reassociation rate of the NFAT:calcineurin complex and the translocation rate of the complex into the nucleus and for NFkappaB is the degradation/resynthesis rate of IkappaB and the import rate of IkappaB into the nucleus. link: http://identifiers.org/pubmed/17031595

Parameters:

NameDescription
k19 = 1.0; k20 = 1.0Reaction: Ca_Nuc + Inact_C_Nuc => Act_C_Nuc, Rate Law: nucleus*(k19*Inact_C_Nuc*Ca_Nuc^3-k20*Act_C_Nuc)
k8 = 0.5; k7 = 0.005Reaction: NFAT_Pi_Act_C_Cyt => NFAT_Pi_Act_C_Nuc, Rate Law: cytosol*k7*NFAT_Pi_Act_C_Cyt-nucleus*k8*NFAT_Pi_Act_C_Nuc
k1 = 2.56E-5; k2 = 0.00256Reaction: NFAT_Pi_Cyt + Act_C_Cyt => Act_C_Cyt + NFAT_Cyt, Rate Law: cytosol*(k1*NFAT_Pi_Cyt-k2*NFAT_Cyt)
k17 = 0.0015; k18 = 9.6E-4Reaction: NFAT_Nuc => NFAT_Cyt, Rate Law: nucleus*k18*NFAT_Nuc-cytosol*k17*NFAT_Cyt
k5 = 0.0019; k6 = 9.2E-4Reaction: Act_C_Nuc => Act_C_Cyt, Rate Law: nucleus*k6*Act_C_Nuc-cytosol*k5*Act_C_Cyt
k12 = 0.00168; k11 = 6.63Reaction: NFAT_Pi_Act_C_Cyt => Act_C_Cyt + NFAT_Pi_Cyt, Rate Law: cytosol*(k12*NFAT_Pi_Act_C_Cyt-k11*NFAT_Pi_Cyt*Act_C_Cyt)
k9 = 0.5; k10 = 0.005Reaction: NFAT_Act_C_Nuc => NFAT_Act_C_Cyt, Rate Law: nucleus*k10*NFAT_Act_C_Nuc-cytosol*k9*NFAT_Act_C_Cyt
k3 = 0.005; k4 = 0.5Reaction: NFAT_Pi_Cyt => NFAT_Pi_Nuc, Rate Law: cytosol*k3*NFAT_Pi_Cyt-nucleus*k4*NFAT_Pi_Nuc
k21 = 0.21; k22 = 0.5Reaction: Ca_Cyt => Ca_Nuc, Rate Law: cytosol*k21*Ca_Cyt-nucleus*k22*Ca_Nuc
k15 = 0.00168; k16 = 6.63Reaction: NFAT_Act_C_Cyt => Act_C_Cyt + NFAT_Cyt, Rate Law: cytosol*(k15*NFAT_Act_C_Cyt-k16*NFAT_Cyt*Act_C_Cyt)
k13 = 0.5; k14 = 0.00256Reaction: NFAT_Act_C_Cyt => NFAT_Pi_Act_C_Cyt, Rate Law: cytosol*(k14*NFAT_Act_C_Cyt-k13*NFAT_Pi_Act_C_Cyt)

States:

NameDescription
Act C Cyt[calcineurin complex; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform]
NFAT Act C Nuc[Nuclear factor of activated T-cells 5; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform; calcineurin complex]
Inact C Cyt[calcineurin complex; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform]
NFAT Pi Act C Nuc[Nuclear factor of activated T-cells 5; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform; calcineurin complex]
NFAT Act C Cyt[Nuclear factor of activated T-cells 5; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform; calcineurin complex]
NFAT Pi Nuc[Nuclear factor of activated T-cells 5]
NFAT Pi Act C Cyt[Nuclear factor of activated T-cells 5; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform; calcineurin complex]
Inact C Nuc[calcineurin complex; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform]
NFAT Pi Cyt[Nuclear factor of activated T-cells 5]
Ca Nuc[calcium(2+); Calcium cation]
NFAT Cyt[Nuclear factor of activated T-cells 5]
Act C Nuc[calcineurin complex; Serine/threonine-protein phosphatase 2B catalytic subunit beta isoform]
NFAT Nuc[Nuclear factor of activated T-cells 5]
Ca Cyt[calcium(2+); Calcium cation]

FitzHugh1961_NerveMembrane: BIOMD0000000346v0.0.1

This is the original model from Richard FitzHugh, which led the famous FitzHugh–Nagumo model, still used for instance in…

Details

Van der Pol's equation for a relaxation oscillator is generalized by the addition of terms to produce a pair of non-linear differential equations with either a stable singular point or a limit cycle. The resulting "BVP model" has two variables of state, representing excitability and refractoriness, and qualitatively resembles Bonhoeffer's theoretical model for the iron wire model of nerve. This BVP model serves as a simple representative of a class of excitable-oscillatory systems including the Hodgkin-Huxley (HH) model of the squid giant axon. The BVP phase plane can be divided into regions corresponding to the physiological states of nerve fiber (resting, active, refractory, enhanced, depressed, etc.) to form a "physiological state diagram," with the help of which many physiological phenomena can be summarized. A properly chosen projection from the 4-dimensional HH phase space onto a plane produces a similar diagram which shows the underlying relationship between the two models. Impulse trains occur in the BVP and HH models for a range of constant applied currents which make the singular point representing the resting state unstable. link: http://identifiers.org/pubmed/19431309

Parameters:

NameDescription
a = 0.7 dimensionless; c = 3.0 dimensionless; b = 0.8 dimensionlessReaction: y = (-1/c)*(x+(-a)+b*y), Rate Law: (-1/c)*(x+(-a)+b*y)
z = -0.4 dimensionless; c = 3.0 dimensionlessReaction: x = c*(x+(-x^3/3)+y+z), Rate Law: c*(x+(-x^3/3)+y+z)

States:

NameDescription
xx
yy

Flahaut2013 - Genome-scale metabolic model of L.lactis (MG1363): MODEL1310300000v0.0.1

Flahaut2013 - Genome-scale metabolic model of L.lactis (MG1363)Genome-scale metabolic model for *Lactococcus lactis* MG…

Details

Lactococcus lactis subsp. cremoris MG1363 is a paradigm strain for lactococci used in industrial dairy fermentations. However, despite of its importance for process development, no genome-scale metabolic model has been reported thus far. Moreover, current models for other lactococci only focus on growth and sugar degradation. A metabolic model that includes nitrogen metabolism and flavor-forming pathways is instrumental for the understanding and designing new industrial applications of these lactic acid bacteria. A genome-scale, constraint-based model of the metabolism and transport in L. lactis MG1363, accounting for 518 genes, 754 reactions, and 650 metabolites, was developed and experimentally validated. Fifty-nine reactions are directly or indirectly involved in flavor formation. Flux Balance Analysis and Flux Variability Analysis were used to investigate flux distributions within the whole metabolic network. Anaerobic carbon-limited continuous cultures were used for estimating the energetic parameters. A thorough model-driven analysis showing a highly flexible nitrogen metabolism, e.g., branched-chain amino acid catabolism which coupled with the redox balance, is pivotal for the prediction of the formation of different flavor compounds. Furthermore, the model predicted the formation of volatile sulfur compounds as a result of the fermentation. These products were subsequently identified in the experimental fermentations carried out. Thus, the genome-scale metabolic model couples the carbon and nitrogen metabolism in L. lactis MG1363 with complete known catabolic pathways leading to flavor formation. The model provided valuable insights into the metabolic networks underlying flavor formation and has the potential to contribute to new developments in dairy industries and cheese-flavor research. link: http://identifiers.org/pubmed/23974365

Flis2015 - Plant clock gene circuit (P2011.1.2 PLM_71 ver 1): BIOMD0000000597v0.0.1

Flis2015 - Plant clock gene circuit (P2011.1.2 PLM_71 ver 1)This model is described in the article: [Defining the robus…

Details

Our understanding of the complex, transcriptional feedback loops in the circadian clock mechanism has depended upon quantitative, timeseries data from disparate sources. We measure clock gene RNA profiles in Arabidopsis thaliana seedlings, grown with or without exogenous sucrose, or in soil-grown plants and in wild-type and mutant backgrounds. The RNA profiles were strikingly robust across the experimental conditions, so current mathematical models are likely to be broadly applicable in leaf tissue. In addition to providing reference data, unexpected behaviours included co-expression of PRR9 and ELF4, and regulation of PRR5 by GI. Absolute RNA quantification revealed low levels of PRR9 transcripts (peak approx. 50 copies cell(-1)) compared with other clock genes, and threefold higher levels of LHY RNA (more than 1500 copies cell(-1)) than of its close relative CCA1. The data are disseminated from BioDare, an online repository for focused timeseries data, which is expected to benefit mechanistic modelling. One data subset successfully constrained clock gene expression in a complex model, using publicly available software on parallel computers, without expert tuning or programming. We outline the empirical and mathematical justification for data aggregation in understanding highly interconnected, dynamic networks such as the clock, and the observed design constraints on the resources required to make this approach widely accessible. link: http://identifiers.org/pubmed/26468131

Parameters:

NameDescription
p16 = 0.62Reaction: => cE3; cE3_m, cE3_m, Rate Law: def*p16*cE3_m/def
m16 = 0.5Reaction: cNI_m => ; cNI_m, Rate Law: def*m16*cNI_m/def
m11 = 1.0; L = 0.5Reaction: cP => ; cP, Rate Law: def*m11*cP*L/def
p8 = 0.6Reaction: => cP9; cP9_m, cP9_m, Rate Law: def*p8*cP9_m/def
n2 = 0.64; g5 = 0.15; g4 = 0.01; e = 2.0Reaction: => cT_m; cEC, cL, cEC, cL, Rate Law: def*n2*g4/(cEC+g4)*g5^e/(cL^e+g5^e)/def
p6 = 0.6Reaction: cCOP1c => cCOP1n; cCOP1c, Rate Law: def*p6*cCOP1c/def
a = 2.0; n1 = 2.6; g1 = 0.1; q1 = 1.2; L = 0.5Reaction: => cL_m; cNI, cP, cP7, cP9, cT, cNI, cP, cP7, cP9, cT, Rate Law: def*(L*q1*cP+n1*g1^a/((cP9+cP7+cNI+cT)^a+g1^a))/def
g16 = 0.3; e = 2.0; n3 = 0.29Reaction: => cE3_m; cL, cL, Rate Law: def*n3*g16^e/(cL^e+g16^e)/def
p23 = 0.37Reaction: => cE4; cE4_m, cE4_m, Rate Law: def*p23*cE4_m/def
p3 = 0.1; c = 2.0; g3 = 0.6Reaction: => cLm; cL, cL, Rate Law: def*p3*cL^c/(cL^c+g3^c)/def
n5 = 0.23Reaction: => cCOP1c, Rate Law: def*n5/def
p11 = 0.51Reaction: => cG; cG_m, cG_m, Rate Law: def*p11*cG_m/def
p17 = 4.8Reaction: cE3 + cG => cEG; cE3, cG, Rate Law: def*p17*cE3*cG/def
p27 = 0.8Reaction: => cLUX; cLUX_m, cLUX_m, Rate Law: def*p27*cLUX_m/def
m20 = 0.6Reaction: cZTL => ; cZTL, Rate Law: def*m20*cZTL/def
m14 = 0.4Reaction: cP7_m => ; cP7_m, Rate Law: def*m14*cP7_m/def
p9 = 0.8Reaction: => cP7; cP7_m, cP7_m, Rate Law: def*p9*cP7_m/def
m12 = 1.0Reaction: cP9_m => ; cP9_m, Rate Law: def*m12*cP9_m/def
g14 = 0.004; n12 = 12.5; q2 = 1.56; g15 = 0.4; e = 2.0; L = 0.5Reaction: => cG_m; cEC, cL, cP, cEC, cL, cP, Rate Law: def*(L*q2*cP+n12*g14/(cEC+g14)*g15^e/(cL^e+g15^e))/def
m19 = 0.2; p26 = 0.3; p28 = 2.0; m30 = 3.0; m29 = 5.0; p25 = 8.0; m37 = 0.8; p29 = 0.1; m36 = 0.1; p17 = 4.8; p21 = 1.0Reaction: cE3n => ; cCOP1d, cCOP1n, cE4, cG, cLUX, cCOP1d, cCOP1n, cE3n, cE4, cG, cLUX, Rate Law: def*(((m29*cE3n*cCOP1n+m30*cE3n*cCOP1d+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/def
m5 = 0.3Reaction: cT_m => ; cT_m, Rate Law: def*m5*cT_m/def
m21 = 0.08Reaction: cZG => ; cZG, Rate Law: def*m21*cZG/def
m13 = 0.32; D = 0.5; m22 = 0.1Reaction: cP9 => ; cP9, Rate Law: def*(m13+m22*D)*cP9/def
p1 = 0.13; p2 = 0.27; L = 0.5Reaction: => cL; cL_m, cL_m, Rate Law: def*cL_m*(p1*L+p2)/def
m1 = 0.3; m2 = 0.24; L = 0.5Reaction: cL_m => ; cL_m, Rate Law: def*(m1*L+m2)*cL_m/def
m26 = 0.5Reaction: cE3_m => ; cE3_m, Rate Law: def*m26*cE3_m/def
m3 = 0.2; p3 = 0.1; c = 2.0; g3 = 0.6Reaction: cL => ; cL, Rate Law: def*(m3*cL+p3*cL^c/(cL^c+g3^c))/def
p4 = 0.56Reaction: => cT; cT_m, cT_m, Rate Law: def*p4*cT_m/def
p25 = 8.0; m37 = 0.8; m36 = 0.1; p26 = 0.3; m39 = 0.3; p21 = 1.0Reaction: cLUX => ; cCOP1d, cCOP1n, cE3n, cE4, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*(m39*cLUX+p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))/def
m4 = 0.2Reaction: cLm => ; cLm, Rate Law: def*m4*cLm/def
p14 = 0.14Reaction: => cZTL, Rate Law: def*p14/def
m34 = 0.6Reaction: cE4_m => ; cE4_m, Rate Law: def*m34*cE4_m/def
D = 0.5; m33 = 13.0; m31 = 0.3Reaction: cCOP1d => ; cCOP1d, Rate Law: def*m31*(1+m33*D)*cCOP1d/def
m32 = 0.2; m19 = 0.2; m10 = 1.0; p29 = 0.1; m36 = 0.1; p17 = 4.8; p31 = 0.1; L = 0.5; d = 2.0; p24 = 10.0; p18 = 4.0; p28 = 2.0; g7 = 0.6; m37 = 0.8; m9 = 1.1Reaction: cEC => ; cCOP1d, cCOP1n, cE3n, cEG, cG, cCOP1d, cCOP1n, cE3n, cEC, cEG, cG, Rate Law: def*(m36*cCOP1n*cEC+m37*cCOP1d*cEC+m32*cEC*(1+p24*L*(p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))^d/((p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))^d+g7^d)))/def
p25 = 8.0; m37 = 0.8; m36 = 0.1; p26 = 0.3; p21 = 1.0; m35 = 0.3Reaction: cE4 => ; cCOP1d, cCOP1n, cE3n, cLUX, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*((m35*cE4+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))/def
q3 = 2.8; g8 = 0.01; g9 = 0.3; n7 = 0.2; e = 2.0; L = 0.5; n4 = 0.07Reaction: => cP9_m; cEC, cL, cP, cEC, cL, cP, Rate Law: def*(L*q3*cP+(n4+n7*cL^e/(cL^e+g9^e))*g8/(cEC+g8))/def
n10 = 0.4; g12 = 0.2; n11 = 0.6; b = 2.0; e = 2.0; g13 = 1.0Reaction: => cNI_m; cLm, cP7, cLm, cP7, Rate Law: def*(n10*cLm^e/(cLm^e+g12^e)+n11*cP7^b/(cP7^b+g13^b))/def
m18 = 3.4Reaction: cG_m => ; cG_m, Rate Law: def*m18*cG_m/def
m19 = 0.2; p29 = 0.1; p17 = 4.8; p28 = 2.0Reaction: cG => ; cE3n, cE3n, cG, Rate Law: def*((m19*cG+p28*cG)-p29*p28*cG/(p29+m19+p17*cE3n))/def
D = 0.5; p7 = 0.3Reaction: => cP; cP, Rate Law: def*p7*D*(1-cP)/def
D = 0.5; m23 = 1.8; m15 = 0.7Reaction: cP7 => ; cP7, Rate Law: def*(m15+m23*D)*cP7/def
p12 = 3.4; D = 0.5; L = 0.5; p13 = 0.1Reaction: cG + cZTL => cZG; cG, cZG, cZTL, Rate Law: def*(p12*L*cZTL*cG-p13*D*cZG)/def
m9 = 1.1Reaction: cE3 => ; cCOP1c, cCOP1c, cE3, Rate Law: def*m9*cE3*cCOP1c/def
m27 = 0.1; p15 = 3.0; L = 0.5Reaction: cCOP1c => ; cCOP1c, Rate Law: def*m27*cCOP1c*(1+p15*L)/def
n14 = 0.1; n6 = 20.0; L = 0.5Reaction: cCOP1n => cCOP1d; cP, cCOP1n, cP, Rate Law: def*(n6*L*cP*cCOP1n+n14*cCOP1n)/def
D = 0.5; m24 = 0.1; m17 = 0.5Reaction: cNI => ; cNI, Rate Law: def*(m17+m24*D)*cNI/def
m19 = 0.2; p18 = 4.0; p28 = 2.0; m10 = 1.0; p29 = 0.1; m9 = 1.1; p17 = 4.8; p31 = 0.1Reaction: cEG => ; cCOP1c, cCOP1d, cCOP1n, cE3n, cG, cCOP1c, cCOP1d, cCOP1n, cE3n, cEG, cG, Rate Law: def*((m9*cEG*cCOP1c+p18*cEG)-p31*(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))/def
p20 = 0.1; p19 = 1.0Reaction: cE3 => cE3n; cE3, cE3n, Rate Law: def*(p19*cE3-p20*cE3n)/def
p10 = 0.54Reaction: => cNI; cNI_m, cNI_m, Rate Law: def*p10*cNI_m/def
p25 = 8.0; m37 = 0.8; m36 = 0.1; p26 = 0.3; p21 = 1.0Reaction: => cEC; cCOP1d, cCOP1n, cE3n, cE4, cLUX, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n)/def
g11 = 0.7; n9 = 0.2; f = 2.0; g10 = 0.5; n8 = 0.5; e = 2.0Reaction: => cP7_m; cL, cLm, cP9, cL, cLm, cP9, Rate Law: def*(n8*(cLm+cL)^e/((cLm+cL)^e+g10^e)+n9*cP9^f/(cP9^f+g11^f))/def
n13 = 1.3; g2 = 0.01; e = 2.0; g6 = 0.3Reaction: => cE4_m; cEC, cL, cEC, cL, Rate Law: def*n13*g2/(cEC+g2)*g6^e/(cL^e+g6^e)/def
D = 0.5; p5 = 4.0; m7 = 0.7; m6 = 0.3; m8 = 0.4Reaction: cT => ; cZG, cZTL, cT, cZG, cZTL, Rate Law: def*((m6+m7*D)*cT*(p5*cZTL+cZG)+m8*cT)/def

States:

NameDescription
cE4cE4
cNIcNI
cLUXcLUX
cP9cP9
cP9 mcP9_m
cZTLcZTL
cCOP1ncCOP1n
cNI mcNI_m
cE4 mcE4_m
cG mcG_m
cEGcEG
cCOP1dcCOP1d
cE3ncE3n
cPcP
cP7cP7
cZGcZG
cE3 mcE3_m
cECcEC
cGcG
cE3cE3
cL mcL_m
cP7 mcP7_m
cCOP1ccCOP1c
cLUX mcLUX_m
cT mcT_m
cLmcLm
cTcT
cLcL

Flis2015 - Plant clock gene circuit (P2011.2.1 PLM_71 ver 2): BIOMD0000000598v0.0.1

cL_m_degr, param m1, modified to ensure light rate > dark rate. Parameter set from PLM_67v2_LDLLLDs_newFFT_1, with modif…

Details

Our understanding of the complex, transcriptional feedback loops in the circadian clock mechanism has depended upon quantitative, timeseries data from disparate sources. We measure clock gene RNA profiles in Arabidopsis thaliana seedlings, grown with or without exogenous sucrose, or in soil-grown plants and in wild-type and mutant backgrounds. The RNA profiles were strikingly robust across the experimental conditions, so current mathematical models are likely to be broadly applicable in leaf tissue. In addition to providing reference data, unexpected behaviours included co-expression of PRR9 and ELF4, and regulation of PRR5 by GI. Absolute RNA quantification revealed low levels of PRR9 transcripts (peak approx. 50 copies cell(-1)) compared with other clock genes, and threefold higher levels of LHY RNA (more than 1500 copies cell(-1)) than of its close relative CCA1. The data are disseminated from BioDare, an online repository for focused timeseries data, which is expected to benefit mechanistic modelling. One data subset successfully constrained clock gene expression in a complex model, using publicly available software on parallel computers, without expert tuning or programming. We outline the empirical and mathematical justification for data aggregation in understanding highly interconnected, dynamic networks such as the clock, and the observed design constraints on the resources required to make this approach widely accessible. link: http://identifiers.org/pubmed/26468131

Parameters:

NameDescription
n8 = 0.46468005656595; f = 2.0; n9 = 0.12054287502747; g10 = 0.59800649651902; e = 2.0; g11 = 0.97065591755812Reaction: => cP7_m; cL, cLm, cP9, cL, cLm, cP9, Rate Law: def*(n8*(cLm+cL)^e/((cLm+cL)^e+g10^e)+n9*cP9^f/(cP9^f+g11^f))/def
p3 = 0.0738150022314; c = 2.0; g3 = 0.6; m3 = 0.17565464903571Reaction: cL => ; cL, Rate Law: def*(m3*cL+p3*cL^c/(cL^c+g3^c))/def
p11 = 0.49350029121361Reaction: => cG; cG_m, cG_m, Rate Law: def*p11*cG_m/def
p17 = 4.32998167851186; m29 = 6.5829611214384; m19 = 0.47083189258762; p26 = 0.3; m30 = 3.12936002914913; m37 = 0.43830433763055; p28 = 2.0; p25 = 8.0; m36 = 0.09362464249722; p29 = 0.1; p21 = 1.0Reaction: cE3n => ; cCOP1d, cCOP1n, cE4, cG, cLUX, cCOP1d, cCOP1n, cE3n, cE4, cG, cLUX, Rate Law: def*(((m29*cE3n*cCOP1n+m30*cE3n*cCOP1d+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/def
g8 = 0.02785533720284; q3 = 2.91645248092752; n4 = 0.04557059014918; n7 = 0.14205317472212; g9 = 0.32642600662781; e = 2.0; L = 0.5Reaction: => cP9_m; cEC, cL, cP, cEC, cL, cP, Rate Law: def*(L*q3*cP+(n4+n7*cL^e/(cL^e+g9^e))*g8/(cEC+g8))/def
m14 = 0.58317183194053Reaction: cP7_m => ; cP7_m, Rate Law: def*m14*cP7_m/def
m11 = 1.0; L = 0.5Reaction: cP => ; cP, Rate Law: def*m11*cP*L/def
p6 = 0.6Reaction: cCOP1c => cCOP1n; cCOP1c, Rate Law: def*p6*cCOP1c/def
m18 = 2.38992856366188Reaction: cG_m => ; cG_m, Rate Law: def*m18*cG_m/def
p17 = 4.32998167851186Reaction: cE3 + cG => cEG; cE3, cG, Rate Law: def*p17*cE3*cG/def
p27 = 1.04800925749369Reaction: => cLUX; cLUX_m, cLUX_m, Rate Law: def*p27*cLUX_m/def
a = 2.0; g1 = 0.08672864809113; q1 = 0.6; n1 = 1.99252254640817; L = 0.5Reaction: => cL_m; cNI, cP, cP7, cP9, cT, cNI, cP, cP7, cP9, cT, Rate Law: def*(L*q1*cP+n1*g1^a/((cP9+cP7+cNI+cT)^a+g1^a))/def
n3 = 0.16472770747976; g16 = 0.21835306363087; e = 2.0Reaction: => cE3_m; cL, cL, Rate Law: def*n3*g16^e/(cL^e+g16^e)/def
n5 = 0.23Reaction: => cCOP1c, Rate Law: def*n5/def
p2 = 0.20262717003844; p1 = 0.07150399789214; L = 0.5Reaction: => cL; cL_m, cL_m, Rate Law: def*cL_m*(p1*L+p2)/def
p9 = 0.85704792589418Reaction: => cP7; cP7_m, cP7_m, Rate Law: def*p9*cP7_m/def
p14 = 0.10935964554573Reaction: => cZTL, Rate Law: def*p14/def
m20 = 0.6Reaction: cZTL => ; cZTL, Rate Law: def*m20*cZTL/def
p25 = 8.0; m36 = 0.09362464249722; p26 = 0.3; m37 = 0.43830433763055; p21 = 1.0Reaction: => cEC; cCOP1d, cCOP1n, cE3n, cE4, cLUX, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n)/def
g4 = 0.00503234997631; n2 = 0.68148116717556; g5 = 0.20247194961847; e = 2.0Reaction: => cT_m; cEC, cL, cEC, cL, Rate Law: def*n2*g4/(cEC+g4)*g5^e/(cL^e+g5^e)/def
m9 = 1.42873823342205; m32 = 0.2; p17 = 4.32998167851186; m37 = 0.43830433763055; p24 = 14.5984045217467; m10 = 1.0; m36 = 0.09362464249722; p29 = 0.1; p31 = 0.1; L = 0.5; d = 2.0; g7 = 0.45632674147836; m19 = 0.47083189258762; p18 = 3.48112987474967; p28 = 2.0Reaction: cEC => ; cCOP1d, cCOP1n, cE3n, cEG, cG, cCOP1d, cCOP1n, cE3n, cEC, cEG, cG, Rate Law: def*(m36*cCOP1n*cEC+m37*cCOP1d*cEC+m32*cEC*(1+p24*L*(p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))^d/((p28*cG/(p29+m19+p17*cE3n)+(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))^d+g7^d)))/def
m12 = 1.0Reaction: cP9_m => ; cP9_m, Rate Law: def*m12*cP9_m/def
p25 = 8.0; m36 = 0.09362464249722; m35 = 0.18382557500265; p26 = 0.3; m37 = 0.43830433763055; p21 = 1.0Reaction: cE4 => ; cCOP1d, cCOP1n, cE3n, cLUX, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*((m35*cE4+p25*cE4*cE3n)-p21*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))/def
n13 = 1.18471991918001; g6 = 0.28604744186645; e = 2.0; g2 = 0.01109625947768Reaction: => cE4_m; cEC, cL, cEC, cL, Rate Law: def*n13*g2/(cEC+g2)*g6^e/(cL^e+g6^e)/def
m9 = 1.42873823342205Reaction: cE3 => ; cCOP1c, cCOP1c, cE3, Rate Law: def*m9*cE3*cCOP1c/def
p16 = 0.9855875650128Reaction: => cE3; cE3_m, cE3_m, Rate Law: def*p16*cE3_m/def
m5 = 0.3Reaction: cT_m => ; cT_m, Rate Law: def*m5*cT_m/def
p3 = 0.0738150022314; c = 2.0; g3 = 0.6Reaction: => cLm; cL, cL, Rate Law: def*p3*cL^c/(cL^c+g3^c)/def
m21 = 0.08Reaction: cZG => ; cZG, Rate Law: def*m21*cZG/def
m24 = 0.11119364985807; D = 0.5; m17 = 0.5Reaction: cNI => ; cNI, Rate Law: def*(m17+m24*D)*cNI/def
p10 = 0.88102987349092Reaction: => cNI; cNI_m, cNI_m, Rate Law: def*p10*cNI_m/def
m26 = 0.5Reaction: cE3_m => ; cE3_m, Rate Law: def*m26*cE3_m/def
g14 = 0.00518249003042; q2 = 0.57336977424479; g15 = 0.49185301792787; e = 2.0; L = 0.5; n12 = 8.43921672276903Reaction: => cG_m; cEC, cL, cP, cEC, cL, cP, Rate Law: def*(L*q2*cP+n12*g14/(cEC+g14)*g15^e/(cL^e+g15^e))/def
p29 = 0.1; p17 = 4.32998167851186; m19 = 0.47083189258762; p28 = 2.0Reaction: cG => ; cE3n, cE3n, cG, Rate Law: def*((m19*cG+p28*cG)-p29*p28*cG/(p29+m19+p17*cE3n))/def
p8 = 0.4098375626616Reaction: => cP9; cP9_m, cP9_m, Rate Law: def*p8*cP9_m/def
p4 = 0.51783935402389Reaction: => cT; cT_m, cT_m, Rate Law: def*p4*cT_m/def
p12 = 2.43270583452351; D = 0.5; p13 = 0.16471437958494; L = 0.5Reaction: cG + cZTL => cZG; cG, cZG, cZTL, Rate Law: def*(p12*L*cZTL*cG-p13*D*cZG)/def
p20 = 0.1940717319972; p19 = 1.74107843497564Reaction: cE3 => cE3n; cE3, cE3n, Rate Law: def*(p19*cE3-p20*cE3n)/def
m2 = 0.45186541768694; m1 = 0.04813458231306; L = 0.5Reaction: cL_m => ; cL_m, Rate Law: def*(m1*L+m2)*cL_m/def
m23 = 0.54491969619247; D = 0.5; m15 = 0.7Reaction: cP7 => ; cP7, Rate Law: def*(m15+m23*D)*cP7/def
m4 = 0.2Reaction: cLm => ; cLm, Rate Law: def*m4*cLm/def
m34 = 0.74619776125315Reaction: cE4_m => ; cE4_m, Rate Law: def*m34*cE4_m/def
D = 0.5; m33 = 13.0; m31 = 0.3Reaction: cCOP1d => ; cCOP1d, Rate Law: def*m31*(1+m33*D)*cCOP1d/def
p25 = 8.0; m36 = 0.09362464249722; p26 = 0.3; m37 = 0.43830433763055; p21 = 1.0; m39 = 0.36610515263739Reaction: cLUX => ; cCOP1d, cCOP1n, cE3n, cE4, cCOP1d, cCOP1n, cE3n, cE4, cLUX, Rate Law: def*(m39*cLUX+p26*cLUX*p25*cE4*cE3n/(p26*cLUX+p21+m37*cCOP1d+m36*cCOP1n))/def
p23 = 0.74Reaction: => cE4; cE4_m, cE4_m, Rate Law: def*p23*cE4_m/def
m7 = 0.49132441826399; D = 0.5; m6 = 0.1718885396183; m8 = 0.33013479704789; p5 = 3.69349002161811Reaction: cT => ; cZG, cZTL, cT, cZG, cZTL, Rate Law: def*((m6+m7*D)*cT*(p5*cZTL+cZG)+m8*cT)/def
m16 = 0.54342221617699Reaction: cNI_m => ; cNI_m, Rate Law: def*m16*cNI_m/def
D = 0.5; p7 = 0.3Reaction: => cP; cP, Rate Law: def*p7*D*(1-cP)/def
m27 = 0.1; p15 = 3.0; L = 0.5Reaction: cCOP1n => ; cCOP1n, Rate Law: def*m27*cCOP1n*(1+p15*L)/def
n14 = 0.1; n6 = 20.0; L = 0.5Reaction: cCOP1n => cCOP1d; cP, cCOP1n, cP, Rate Law: def*(n6*L*cP*cCOP1n+n14*cCOP1n)/def
g12 = 0.2; n11 = 1.04887048285294; n10 = 0.53104365301892; b = 2.0; e = 2.0; g13 = 1.0Reaction: => cNI_m; cLm, cP7, cLm, cP7, Rate Law: def*(n10*cLm^e/(cLm^e+g12^e)+n11*cP7^b/(cP7^b+g13^b))/def
m13 = 0.32; D = 0.5; m22 = 0.09605427710298Reaction: cP9 => ; cP9, Rate Law: def*(m13+m22*D)*cP9/def
m9 = 1.42873823342205; p17 = 4.32998167851186; p18 = 3.48112987474967; m19 = 0.47083189258762; p28 = 2.0; m10 = 1.0; p29 = 0.1; p31 = 0.1Reaction: cEG => ; cCOP1c, cCOP1d, cCOP1n, cE3n, cG, cCOP1c, cCOP1d, cCOP1n, cE3n, cEG, cG, Rate Law: def*((m9*cEG*cCOP1c+p18*cEG)-p31*(p18*cEG+p17*cE3n*p28*cG/(p29+m19+p17*cE3n))/(m9*cCOP1n+m10*cCOP1d+p31))/def

States:

NameDescription
cE4cE4
cNIcNI
cLUXcLUX
cP9cP9
cP9 mcP9_m
cZTLcZTL
cCOP1ncCOP1n
cE4 mcE4_m
cNI mcNI_m
cEGcEG
cG mcG_m
cCOP1dcCOP1d
cPcP
cE3ncE3n
cP7cP7
cZGcZG
cE3 mcE3_m
cECcEC
cGcG
cE3cE3
cL mcL_m
cP7 mcP7_m
cCOP1ccCOP1c
cT mcT_m
cLUX mcLUX_m
cLmcLm
cTcT
cLcL

Flis2015 - Plant clock gene circuit (P2011.3.1 PLM_1041 ver 1): MODEL1510190002v0.0.1

cL_m_degr, param m1, modified to ensure light rate > dark rate.

Details

Our understanding of the complex, transcriptional feedback loops in the circadian clock mechanism has depended upon quantitative, timeseries data from disparate sources. We measure clock gene RNA profiles in Arabidopsis thaliana seedlings, grown with or without exogenous sucrose, or in soil-grown plants and in wild-type and mutant backgrounds. The RNA profiles were strikingly robust across the experimental conditions, so current mathematical models are likely to be broadly applicable in leaf tissue. In addition to providing reference data, unexpected behaviours included co-expression of PRR9 and ELF4, and regulation of PRR5 by GI. Absolute RNA quantification revealed low levels of PRR9 transcripts (peak approx. 50 copies cell(-1)) compared with other clock genes, and threefold higher levels of LHY RNA (more than 1500 copies cell(-1)) than of its close relative CCA1. The data are disseminated from BioDare, an online repository for focused timeseries data, which is expected to benefit mechanistic modelling. One data subset successfully constrained clock gene expression in a complex model, using publicly available software on parallel computers, without expert tuning or programming. We outline the empirical and mathematical justification for data aggregation in understanding highly interconnected, dynamic networks such as the clock, and the observed design constraints on the resources required to make this approach widely accessible. link: http://identifiers.org/pubmed/26468131

Flis2015 - Plant clock gene circuit (P2011.4.1 PLM_1042 ver 1): MODEL1510190003v0.0.1

cL_m_degr, param m1, modified to ensure light rate > dark rate.

Details

Our understanding of the complex, transcriptional feedback loops in the circadian clock mechanism has depended upon quantitative, timeseries data from disparate sources. We measure clock gene RNA profiles in Arabidopsis thaliana seedlings, grown with or without exogenous sucrose, or in soil-grown plants and in wild-type and mutant backgrounds. The RNA profiles were strikingly robust across the experimental conditions, so current mathematical models are likely to be broadly applicable in leaf tissue. In addition to providing reference data, unexpected behaviours included co-expression of PRR9 and ELF4, and regulation of PRR5 by GI. Absolute RNA quantification revealed low levels of PRR9 transcripts (peak approx. 50 copies cell(-1)) compared with other clock genes, and threefold higher levels of LHY RNA (more than 1500 copies cell(-1)) than of its close relative CCA1. The data are disseminated from BioDare, an online repository for focused timeseries data, which is expected to benefit mechanistic modelling. One data subset successfully constrained clock gene expression in a complex model, using publicly available software on parallel computers, without expert tuning or programming. We outline the empirical and mathematical justification for data aggregation in understanding highly interconnected, dynamic networks such as the clock, and the observed design constraints on the resources required to make this approach widely accessible. link: http://identifiers.org/pubmed/26468131

Flis2015 - Plant clock gene circuit (P2011.5.1 PLM_1043 ver 1): MODEL1510190004v0.0.1

cL_m_degr, param m1, modified to ensure light rate > dark rate.

Details

Our understanding of the complex, transcriptional feedback loops in the circadian clock mechanism has depended upon quantitative, timeseries data from disparate sources. We measure clock gene RNA profiles in Arabidopsis thaliana seedlings, grown with or without exogenous sucrose, or in soil-grown plants and in wild-type and mutant backgrounds. The RNA profiles were strikingly robust across the experimental conditions, so current mathematical models are likely to be broadly applicable in leaf tissue. In addition to providing reference data, unexpected behaviours included co-expression of PRR9 and ELF4, and regulation of PRR5 by GI. Absolute RNA quantification revealed low levels of PRR9 transcripts (peak approx. 50 copies cell(-1)) compared with other clock genes, and threefold higher levels of LHY RNA (more than 1500 copies cell(-1)) than of its close relative CCA1. The data are disseminated from BioDare, an online repository for focused timeseries data, which is expected to benefit mechanistic modelling. One data subset successfully constrained clock gene expression in a complex model, using publicly available software on parallel computers, without expert tuning or programming. We outline the empirical and mathematical justification for data aggregation in understanding highly interconnected, dynamic networks such as the clock, and the observed design constraints on the resources required to make this approach widely accessible. link: http://identifiers.org/pubmed/26468131

Flis2015 - Plant clock gene circuit (P2011.6.1 PLM_1044 ver 1): MODEL1510190005v0.0.1

cL_m_degr, param m1, modified to ensure light rate > dark rate.

Details

Our understanding of the complex, transcriptional feedback loops in the circadian clock mechanism has depended upon quantitative, timeseries data from disparate sources. We measure clock gene RNA profiles in Arabidopsis thaliana seedlings, grown with or without exogenous sucrose, or in soil-grown plants and in wild-type and mutant backgrounds. The RNA profiles were strikingly robust across the experimental conditions, so current mathematical models are likely to be broadly applicable in leaf tissue. In addition to providing reference data, unexpected behaviours included co-expression of PRR9 and ELF4, and regulation of PRR5 by GI. Absolute RNA quantification revealed low levels of PRR9 transcripts (peak approx. 50 copies cell(-1)) compared with other clock genes, and threefold higher levels of LHY RNA (more than 1500 copies cell(-1)) than of its close relative CCA1. The data are disseminated from BioDare, an online repository for focused timeseries data, which is expected to benefit mechanistic modelling. One data subset successfully constrained clock gene expression in a complex model, using publicly available software on parallel computers, without expert tuning or programming. We outline the empirical and mathematical justification for data aggregation in understanding highly interconnected, dynamic networks such as the clock, and the observed design constraints on the resources required to make this approach widely accessible. link: http://identifiers.org/pubmed/26468131

Floc'hlay2020 - SeaUrchin_model_ginsim: MODEL2002190001v0.0.1

Multilevel logical model encompassing the Nodal and BMP pathways together with key transcription factors setting the dor…

Details

During sea urchin development, secretion of Nodal and BMP2/4 ligands and their antagonists Lefty and Chordin from a ventral organizer region specifies the ventral and dorsal territories. This process relies on a complex interplay between the Nodal and BMP pathways through numerous regulatory circuits. To decipher the interplay between these pathways, we used a combination of treatments with recombinant Nodal and BMP2/4 proteins and a computational modelling approach. We assembled a logical model focusing on cell responses to signalling inputs along the dorsal-ventral axis, which was extended to cover ligand diffusion and enable multicellular simulations. Our model simulations accurately recapitulate gene expression in wild type embryos, accounting for the specification of ventral ectoderm, ciliary band and dorsal ectoderm. Our model simulations further recapitulate various morphant phenotypes, reveals a dominance of the BMP pathway over the Nodal pathway, and stresses the crucial impact of the rate of Smad activation in D/V patterning. These results emphasise the key role of the mutual antagonism between the Nodal and BMP2/4 pathways in driving early dorsal-ventral patterning of the sea urchin embryo. link: http://identifiers.org/pubmed/33298464

Fox2002_IonicMechanism_CardiacMyocytes: MODEL0911665321v0.0.1

This a model from the article: Ionic mechanism of electrical alternans. Fox JJ, McHarg JL, Gilmour RF Jr. Am J Physi…

Details

Although alternans of action potential duration (APD) is a robust feature of the rapidly paced canine ventricle, currently available ionic models of cardiac myocytes do not recreate this phenomenon. To address this problem, we developed a new ionic model using formulations of currents based on previous models and recent experimental data. Compared with existing models, the inward rectifier K(+) current (I(K1)) was decreased at depolarized potentials, the maximum conductance and rectification of the rapid component of the delayed rectifier K(+) current (I(Kr)) were increased, and I(Kr) activation kinetics were slowed. The slow component of the delayed rectifier K(+) current (I(Ks)) was increased in magnitude and activation shifted to less positive voltages, and the L-type Ca(2+) current (I(Ca)) was modified to produce a smaller, more rapidly inactivating current. Finally, a simplified form of intracellular calcium dynamics was adopted. In this model, APD alternans occurred at cycle lengths = 150-210 ms, with a maximum alternans amplitude of 39 ms. APD alternans was suppressed by decreasing I(Ca) magnitude or calcium-induced inactivation and by increasing the magnitude of I(K1), I(Kr), or I(Ks). These results establish an ionic basis for APD alternans, which should facilitate the development of pharmacological approaches to eliminating alternans. link: http://identifiers.org/pubmed/11788399

Frascoli2014 - A dynamical model of tumour immunotherapy: BIOMD0000000787v0.0.1

This is a coupled ordinary differential equation model of tumour-immune dynamics, accounting for biological and clinical…

Details

A coupled ordinary differential equation model of tumour-immune dynamics is presented and analysed. The model accounts for biological and clinical factors which regulate the interaction rates of cytotoxic T lymphocytes on the surface of the tumour mass. A phase plane analysis demonstrates that competition between tumour cells and lymphocytes can result in tumour eradication, perpetual oscillations, or unbounded solutions. To investigate the dependence of the dynamic behaviour on model parameters, the equations are solved analytically and conditions for unbounded versus bounded solutions are discussed. An analytic characterisation of the basin of attraction for oscillatory orbits is given. It is also shown that the tumour shape, characterised by a surface area to volume scaling factor, influences the size of the basin, with significant consequences for therapy design. The findings reveal that the tumour volume must surpass a threshold size that depends on lymphocyte parameters for the cancer to be completely eliminated. A semi-analytic procedure to calculate oscillation periods and determine their sensitivity to model parameters is also presented. Numerical results show that the period of oscillations exhibits notable nonlinear dependence on biologically relevant conditions. link: http://identifiers.org/pubmed/24759513

Parameters:

NameDescription
rho = 4.83597586204941; min_C = 0.1; k = 0.2Reaction: V_Tumor_Volume => ; C_Cytotoxic_T_Lymphocytes_Coverage, Rate Law: compartment*rho*k*V_Tumor_Volume^(2/3)*min_C
d_c = 0.2Reaction: C_Cytotoxic_T_Lymphocytes_Coverage =>, Rate Law: compartment*d_c*C_Cytotoxic_T_Lymphocytes_Coverage
rho = 4.83597586204941; r_t = 0.1Reaction: => V_Tumor_Volume, Rate Law: compartment*rho*r_t*V_Tumor_Volume^(2/3)
r_c = 0.001; rho = 4.83597586204941Reaction: => C_Cytotoxic_T_Lymphocytes_Coverage; V_Tumor_Volume, Rate Law: compartment*rho*r_c*V_Tumor_Volume^(2/3)*C_Cytotoxic_T_Lymphocytes_Coverage

States:

NameDescription
C Cytotoxic T Lymphocytes Coverage[cytotoxic T cell; T cell mediated cytotoxicity directed against tumor cell target]
V Tumor Volume[Tumor Volume]

Fribourg2014 - Dynamics of viral antagonism and innate immune response (H1N1 influenza A virus - Cal/09): BIOMD0000000528v0.0.1

Fribourg2014 - Dynamics of viral antagonism and innate immune response (H1N1 influenza A virus - Cal/09) The dynamics o…

Details

Viral antagonism of host responses is an essential component of virus pathogenicity. The study of the interplay between immune response and viral antagonism is challenging due to the involvement of many processes acting at multiple time scales. Here we develop an ordinary differential equation model to investigate the early, experimentally measured, responses of human monocyte-derived dendritic cells to infection by two H1N1 influenza A viruses of different clinical outcomes: pandemic A/California/4/2009 and seasonal A/New Caledonia/20/1999. Our results reveal how the strength of virus antagonism, and the time scale over which it acts to thwart the innate immune response, differs significantly between the two viruses, as is made clear by their impact on the temporal behavior of a number of measured genes. The model thus sheds light on the mechanisms that underlie the variability of innate immune responses to different H1N1 viruses. link: http://identifiers.org/pubmed/24594370

Parameters:

NameDescription
k26 = 0.360085 substance; tao12 = 1.0 substance; r4 = 1.0E-5 substance; IC2 = 0.0; IC1 = 0.0Reaction: w => STATm; STATP2n, Rate Law: (r4*IC1+k26*STATP2n)*IC2-STATm*ln(2)/tao12
C = 500000.0 substance; K19 = 0.004 substance; vmax19 = 154800.0 substance; NA = 6.023E23 substanceReaction: w => TNFenv; TNFam, Rate Law: 1000000000*C*vmax19/NA*TNFam/(K19+TNFam)
TJ = 0.0; tao3 = 0.56 substance; K5 = 0.01 substanceReaction: w => STATP2n; STAT, Rate Law: K5*TJ*STAT/2/(K5+STAT)-STATP2n*ln(2)/tao3
tao1 = 2.5 substance; IC2 = 0.0; k15 = 3.6E-8 substance; r0 = 0.001 substance; IC1 = 0.0Reaction: w => IFNb_mRNA; IRF7Pn, Rate Law: (r0*IC1+k15*IRF7Pn)*IC2-IFNb_mRNA*ln(2)/tao1
tao8 = 2.0 substance; IC2ifa = 0.0; k16 = 0.36 substanceReaction: w => IFNa_mRNA; IRF7Pn, Rate Law: k16*IRF7Pn*IC2ifa-IFNa_mRNA*ln(2)/tao8
k28 = 360.0 substance; tao13 = 15.0 substanceReaction: w => STAT; STATm, Rate Law: k28*STATm-STAT*ln(2)/tao13
C = 500000.0 substance; K17 = 0.002 substance; vmax17 = 72000.0 substance; NA = 6.023E23 substanceReaction: w => IFNa_env; IFNa_mRNA, Rate Law: 1000000000*C*vmax17/NA*IFNa_mRNA/(K17+IFNa_mRNA)
C = 500000.0 substance; K2 = 0.002 substance; vmax2 = 72000.0 substance; NA = 6.023E23 substanceReaction: w => IFNb_env; IFNb_mRNA, Rate Law: 1000000000*C*vmax2/NA*IFNb_mRNA/(K2+IFNb_mRNA)
K20 = 6.0E-4 substance; tao9 = 2.0 substance; r1 = 1.0E-4 substance; IC2 = 0.0; rmax20 = 0.001 substance; IC1 = 0.0Reaction: w => TNFam; TNFenv, Rate Law: (r1*IC1+rmax20*TNFenv/(K20+TNFenv))*IC2-TNFam*ln(2)/tao9
k8 = 0.0036 substance; IC2 = 0.0; r3 = 1.0E-7 substance; tao4 = 0.46 substance; IC1 = 0.0Reaction: w => SOCS1m; STATP2n, Rate Law: (r3*IC1+k8*STATP2n)*IC2-SOCS1m*ln(2)/tao4
k12 = 360.0 substance; IC1 = 0.0Reaction: w => IRF7Pn; IRF7m, Rate Law: k12*IC1*IRF7m
k11 = 3.6E-4 substance; k14 = 3.204E-7 substance; IC2 = 0.0; tao6 = 1.0Reaction: w => IRF7m; STATP2n, IRF7Pn, Rate Law: (k11*STATP2n+k14*IRF7Pn)*IC2-IRF7m*ln(2)/tao6

States:

NameDescription
SOCS1m[IPR028411]
IFNa mRNA[IPR015589]
IFNa env[IPR015589]
IRF7Pn[Interferon regulatory factor 7]
IRF7m[Interferon regulatory factor 7]
TNFam[Tumor necrosis factor]
ww
STATm[IPR001217]
IFNb env[Interferon beta]
IFNb mRNA[Interferon beta]
TNFenv[Tumor necrosis factor]
STATP2n[IPR001217]
STAT[IPR001217]

Fribourg2014 - Dynamics of viral antagonism and innate immune response (H1N1 influenza A virus - NC/99): BIOMD0000000529v0.0.1

Fribourg2014 - Dynamics of viral antagonism and innate immune response (H1N1 influenza A virus - NC/99) The dynamics of…

Details

Viral antagonism of host responses is an essential component of virus pathogenicity. The study of the interplay between immune response and viral antagonism is challenging due to the involvement of many processes acting at multiple time scales. Here we develop an ordinary differential equation model to investigate the early, experimentally measured, responses of human monocyte-derived dendritic cells to infection by two H1N1 influenza A viruses of different clinical outcomes: pandemic A/California/4/2009 and seasonal A/New Caledonia/20/1999. Our results reveal how the strength of virus antagonism, and the time scale over which it acts to thwart the innate immune response, differs significantly between the two viruses, as is made clear by their impact on the temporal behavior of a number of measured genes. The model thus sheds light on the mechanisms that underlie the variability of innate immune responses to different H1N1 viruses. link: http://identifiers.org/pubmed/24594370

Parameters:

NameDescription
C = 500000.0 substance; K19 = 0.004 substance; vmax19 = 154800.0 substance; NA = 6.023E23 substanceReaction: w => TNFenv; TNFam, Rate Law: 1000000000*C*vmax19/NA*TNFam/(K19+TNFam)
C = 500000.0 substance; K2 = 72000.0 substance; vmax2 = 72000.0 substance; NA = 6.023E23 substanceReaction: w => IFNb_env; IFNb_mRNA, Rate Law: 1000000000*C*vmax2/NA*IFNb_mRNA/(K2+IFNb_mRNA)
tao8 = 2.0 substance; k16 = 3600.0 substance; IC2ifa = 0.0Reaction: w => IFNa_mRNA; IRF7Pn, Rate Law: k16*IRF7Pn*IC2ifa-IFNa_mRNA*ln(2)/tao8
k15 = 3.6E-5 substance; tao1 = 2.5 substance; IC2 = 0.0; r0 = 0.003 substance; IC1 = 0.0Reaction: w => IFNb_mRNA; IRF7Pn, Rate Law: (r0*IC1+k15*IRF7Pn)*IC2-IFNb_mRNA*ln(2)/tao1
TJ = 0.0; tao3 = 0.56 substance; K5 = 0.01 substanceReaction: w => STATP2n; STAT, Rate Law: K5*TJ*STAT/2/(K5+STAT)-STATP2n*ln(2)/tao3
k28 = 360.0 substance; tao13 = 15.0 substanceReaction: w => STAT; STATm, Rate Law: k28*STATm-STAT*ln(2)/tao13
k26 = 0.360085 substance; tao12 = 1.0 substance; r4 = 1.0E-6 substance; IC2 = 0.0; IC1 = 0.0Reaction: w => STATm; STATP2n, Rate Law: (r4*IC1+k26*STATP2n)*IC2-STATm*ln(2)/tao12
K20 = 6.0E-4 substance; tao9 = 2.0 substance; IC2 = 0.0; rmax20 = 0.001 substance; r1 = 2.5E-4 substance; IC1 = 0.0Reaction: w => TNFam; TNFenv, Rate Law: (r1*IC1+rmax20*TNFenv/(K20+TNFenv))*IC2-TNFam*ln(2)/tao9
C = 500000.0 substance; K17 = 0.002 substance; vmax17 = 72000.0 substance; NA = 6.023E23 substanceReaction: w => IFNa_env; IFNa_mRNA, Rate Law: 1000000000*C*vmax17/NA*IFNa_mRNA/(K17+IFNa_mRNA)
k8 = 0.0036 substance; IC2 = 0.0; r3 = 1.0E-7 substance; tao4 = 0.46 substance; IC1 = 0.0Reaction: w => SOCS1m; STATP2n, Rate Law: (r3*IC1+k8*STATP2n)*IC2-SOCS1m*ln(2)/tao4
k12 = 3600.0 substance; IC1 = 0.0Reaction: w => IRF7Pn; IRF7m, Rate Law: k12*IC1*IRF7m
k11 = 3.6E-4 substance; k14 = 3.204E-7 substance; IC2 = 0.0; tao6 = 1.0Reaction: w => IRF7m; STATP2n, IRF7Pn, Rate Law: (k11*STATP2n+k14*IRF7Pn)*IC2-IRF7m*ln(2)/tao6

States:

NameDescription
SOCS1m[IPR028411]
IFNa mRNA[IPR015589]
IFNa env[IPR015589]
IRF7m[Interferon regulatory factor 7]
IRF7Pn[Interferon regulatory factor 7]
TNFam[Tumor necrosis factor]
ww
STATm[IPR001217]
IFNb env[IPR015588]
IFNb mRNA[IPR015588]
TNFenv[Tumor necrosis factor]
STATP2n[IPR001217]
STAT[IPR001217]

Fribourg2014 - Model of influenza A virus infection dynamics of viral antagonism and innate immune response.: BIOMD0000000889v0.0.1

This is an ordinary differential equation mathematical model investigating the early responses of human monocyte-derived…

Details

Viral antagonism of host responses is an essential component of virus pathogenicity. The study of the interplay between immune response and viral antagonism is challenging due to the involvement of many processes acting at multiple time scales. Here we develop an ordinary differential equation model to investigate the early, experimentally measured, responses of human monocyte-derived dendritic cells to infection by two H1N1 influenza A viruses of different clinical outcomes: pandemic A/California/4/2009 and seasonal A/New Caledonia/20/1999. Our results reveal how the strength of virus antagonism, and the time scale over which it acts to thwart the innate immune response, differs significantly between the two viruses, as is made clear by their impact on the temporal behavior of a number of measured genes. The model thus sheds light on the mechanisms that underlie the variability of innate immune responses to different H1N1 viruses. link: http://identifiers.org/pubmed/24594370

Parameters:

NameDescription
t_6 = 1.0Reaction: IRF7m =>, Rate Law: compartment*IRF7m*ln(2)/t_6
K_20 = 6.0E-4; r_20 = 0.001; IC2 = 1.0; r_1 = 1.0E-4; IC1 = 1.0Reaction: => TNFam; TFNenv, Rate Law: compartment*IC2*(r_1*IC1+r_20*TFNenv/(K_20+TFNenv))
k_26 = 0.018; IC2 = 1.0; r_4 = 1.0E-6; IC1 = 1.0Reaction: => STATm; STATP2n, Rate Law: compartment*(r_4*IC1+k_26*STATP2n)*IC2
k_8 = 0.0036; IC2 = 1.0; r_3 = 1.0E-7; IC1 = 1.0Reaction: => SOCSm; STATP2n, Rate Law: compartment*(r_3*IC1+k_8*STATP2n)*IC2
k_12 = 360.0; IC1 = 1.0Reaction: => IRF7Pn; IRF7m, Rate Law: compartment*k_12*IRF7m*IC1
t_4 = 0.46Reaction: SOCSm =>, Rate Law: compartment*SOCSm*ln(2)/t_4
t_8 = 2.0Reaction: IFNam =>, Rate Law: compartment*IFNam*ln(2)/t_8
k_16 = 0.36; IC2 = 1.0Reaction: => IFNam; IRF7Pn, Rate Law: compartment*k_16*IRF7Pn*IC2
t_3 = 0.56Reaction: STATP2n =>, Rate Law: compartment*STATP2n*ln(2)/t_3
t_13 = 15.0Reaction: STAT =>, Rate Law: compartment*STAT*ln(2)/t_13
C = 500000.0; v_max217 = 72360.0; K_217 = 0.002; gamma = 1.66030217499585E-15Reaction: => IFNBenv, Rate Law: compartment*gamma*C*v_max217*IFNBenv/(K_217+IFNBenv)
t_9 = 2.0Reaction: TNFam =>, Rate Law: compartment*TNFam*ln(2)/t_9
t_12 = 1.0Reaction: STATm =>, Rate Law: compartment*STATm*ln(2)/t_12
k_5 = 3600.0; K_5 = 0.01; TJ = 3.83790087997253E-11Reaction: => STATP2n; STAT, Rate Law: compartment*k_5*TJ*STAT/(2*(K_5+STAT))
r_0 = 0.001; IC2 = 1.0; k_15 = 3.6E-5; IC1 = 1.0Reaction: => IFNBm; IRF7Pn, Rate Law: compartment*(r_0*IC1+k_15*IRF7Pn)*IC2
C = 500000.0; K_19 = 0.004; v_max19 = 165600.0; gamma = 1.66030217499585E-15Reaction: => TFNenv; TNFam, Rate Law: compartment*gamma*C*v_max19*TNFam/(K_19+TNFam)
k_28 = 360.0Reaction: => STAT; STATm, Rate Law: compartment*k_28*STATm
t_1 = 2.5Reaction: IFNBm =>, Rate Law: compartment*IFNBm*ln(2)/t_1
IC2 = 1.0; k_11 = 3.6E-4; k_14 = 3.204E-7Reaction: => IRF7m; STATP2n, IRF7Pn, Rate Law: compartment*(k_11*STATP2n+k_14*IRF7Pn)*IC2

States:

NameDescription
IRF7m[C128883]
IRF7Pn[C128883]
TNFam[PR:000000134]
IFNBm[C20495]
TFNenv[PR:000000134]
IFNBenv[C20495]
STATm[C19618]
SOCSm[C97796]
IFNam[C20494]
IFNaenv[C20494]
STATP2n[C19618; SBO:0000607]
STAT[C19618]

Fridlyand2003_Calcium_flux: BIOMD0000000059v0.0.1

The model reproduces block A of Fig 5 and also Fig 3 (without the inclusion of Tg action). The model was successfully te…

Details

We have developed a detailed mathematical model of ionic flux in beta-cells that includes the most essential channels and pumps in the plasma membrane. This model is coupled to equations describing Ca2+, inositol 1,4,5-trisphosphate (IP3), ATP, and Na+ homeostasis, including the uptake and release of Ca2+ by the endoplasmic reticulum (ER). In our model, metabolically derived ATP activates inward Ca2+ flux by regulation of ATP-sensitive K+ channels and depolarization of the plasma membrane. Results from the simulations support the hypothesis that intracellular Na+ and Ca2+ in the ER can be the main variables driving both fast (2-7 osc/min) and slow intracellular Ca2+ concentration oscillations (0.3-0.9 osc/min) and that the effect of IP3 on Ca2+ leak from the ER contributes to the pattern of slow calcium oscillations. Simulations also show that filling the ER Ca2+ stores leads to faster electrical bursting and Ca2+ oscillations. Specific Ca2+ oscillations in isolated beta-cell lines can also be simulated. link: http://identifiers.org/pubmed/12644446

Parameters:

NameDescription
kadp = 3.7E-4 Time inverseReaction: => ATP_cyt; ADP_cyt, Rate Law: Cytoplasm*kadp*ADP_cyt
F = 9.6485E16 Faraday constant; I_CRAN = 0.0 CurrentReaction: => Na_cyt, Rate Law: (-I_CRAN)/F
katp = 5.0E-5 Time inverseReaction: ATP_cyt =>, Rate Law: Cytoplasm*katp*ATP_cyt
kip = 3.0E-4 concentration per time; Kipca = 0.4 ConcentrationReaction: => IP3_cyt; Ca_cyt, Rate Law: Cytoplasm*kip*Ca_cyt^2/(Ca_cyt^2+Kipca^2)
kdip = 4.0E-5 Time inverseReaction: IP3_cyt =>, Rate Law: Cytoplasm*kdip*IP3_cyt
I_NaCa = 0.0 Current; F = 9.6485E16 Faraday constantReaction: Na_cyt =>, Rate Law: 3*I_NaCa/F
katpca = 5.0E-5 per concentration per timeReaction: ATP_cyt => ; Ca_cyt, Rate Law: Cytoplasm*katpca*Ca_cyt*ATP_cyt
F = 9.6485E16 Faraday constant; I_CaPump = 0.0 CurrentReaction: ATP_cyt =>, Rate Law: I_CaPump/F
Jout = 0.0 amount per timeReaction: Ca_er => Ca_cyt, Rate Law: Jout
fi = 0.01 dimensionless; F = 9.6485E16 Faraday constant; I_CaPump = 0.0 CurrentReaction: Ca_cyt =>, Rate Law: fi*2*I_CaPump/(2*F)
I_Na = 0.0 Current; F = 9.6485E16 Faraday constantReaction: => Na_cyt, Rate Law: (-I_Na)/F
ksg = 1.0E-4 Time inverseReaction: Ca_cyt =>, Rate Law: Cytoplasm*ksg*Ca_cyt
F = 9.6485E16 Faraday constant; I_NaK = 0.0 CurrentReaction: Na_cyt =>, Rate Law: 3*I_NaK/F
Jerp = 0.0 concentration per timeReaction: ATP_cyt =>, Rate Law: Cytoplasm*Jerp/2
I_NaCa = 0.0 Current; fi = 0.01 dimensionless; F = 9.6485E16 Faraday constantReaction: => Ca_cyt, Rate Law: fi*2*I_NaCa/(2*F)
fi = 0.01 dimensionless; I_Vca = 0.0 Current; F = 9.6485E16 Faraday constantReaction: => Ca_cyt, Rate Law: fi*(-I_Vca)/(2*F)

States:

NameDescription
Na cyt[sodium(1+); Sodium cation]
ADP cyt[ADP; ADP]
ATP cyt[ATP; ATP]
Ca cyt[calcium(2+); Calcium cation]
IP3 cyt[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
Ca er[calcium(2+); Calcium cation]

Fridlyand2010_GlucoseSensitivity_A: BIOMD0000000348v0.0.1

This a model from the article: Glucose sensing in the pancreatic beta cell: a computational systems analysis. Fridl…

Details

Pancreatic beta-cells respond to rising blood glucose by increasing oxidative metabolism, leading to an increased ATP/ADP ratio in the cytoplasm. This leads to a closure of KATP channels, depolarization of the plasma membrane, influx of calcium and the eventual secretion of insulin. Such mechanism suggests that beta-cell metabolism should have a functional regulation specific to secretion, as opposed to coupling to contraction. The goal of this work is to uncover contributions of the cytoplasmic and mitochondrial processes in this secretory coupling mechanism using mathematical modeling in a systems biology approach.We describe a mathematical model of beta-cell sensitivity to glucose. The cytoplasmic part of the model includes equations describing glucokinase, glycolysis, pyruvate reduction, NADH and ATP production and consumption. The mitochondrial part begins with production of NADH, which is regulated by pyruvate dehydrogenase. NADH is used in the electron transport chain to establish a proton motive force, driving the F1F0 ATPase. Redox shuttles and mitochondrial Ca2+ handling were also modeled.The model correctly predicts changes in the ATP/ADP ratio, Ca2+ and other metabolic parameters in response to changes in substrate delivery at steady-state and during cytoplasmic Ca2+ oscillations. Our analysis of the model simulations suggests that the mitochondrial membrane potential should be relatively lower in beta cells compared with other cell types to permit precise mitochondrial regulation of the cytoplasmic ATP/ADP ratio. This key difference may follow from a relative reduction in respiratory activity. The model demonstrates how activity of lactate dehydrogenase, uncoupling proteins and the redox shuttles can regulate beta-cell function in concert; that independent oscillations of cytoplasmic Ca2+ can lead to slow coupled metabolic oscillations; and that the relatively low production rate of reactive oxygen species in beta-cells under physiological conditions is a consequence of the relatively decreased mitochondrial membrane potential.This comprehensive model predicts a special role for mitochondrial control mechanisms in insulin secretion and ROS generation in the beta cell. The model can be used for testing and generating control hypotheses and will help to provide a more complete understanding of beta-cell glucose-sensing central to the physiology and pathology of pancreatic beta-cells. link: http://identifiers.org/pubmed/20497556

Parameters:

NameDescription
Jgpd = NaN; kgpd = 1.0E-5; JGlu = NaN; Vi = 0.53Reaction: G3P = (2*JGlu+(-Jgpd))*1/Vi+(-kgpd*G3P), Rate Law: (2*JGlu+(-Jgpd))*1/Vi+(-kgpd*G3P)
Jtnadh = NaN; Jgpd = NaN; JLDH = NaN; Vi = 0.53; knadhc = 1.0E-4Reaction: NADHc = (Jgpd+(-Jtnadh)+(-JLDH))*1/Vi+(-knadhc*NADHc), Rate Law: (Jgpd+(-Jtnadh)+(-JLDH))*1/Vi+(-knadhc*NADHc)
fm = 3.0E-4; Vmmit = 0.0144; Juni = NaN; JNCa = NaNReaction: Cam = fm*(Juni+(-JNCa))*1/Vmmit, Rate Law: fm*(Juni+(-JNCa))*1/Vmmit
Jtnadh = NaN; knadhm = 1.0E-4; Vmmit = 0.0144; JPYR = NaN; Jhres = NaNReaction: NADHm = (4.6*JPYR+(-0.1*Jhres)+Jtnadh)*1/Vmmit+(-knadhm*NADHm), Rate Law: (4.6*JPYR+(-0.1*Jhres)+Jtnadh)*1/Vmmit+(-knadhm*NADHm)
Jph = NaN; Juni = NaN; Cmit = 5200.0; JNCa = NaN; Jhres = NaN; F = 96480.0; Jhl = NaNReaction: Vm = (Jhres+(-Jph)+(-Jhl)+(-2*Juni)+(-JNCa))*F*1/Cmit, Rate Law: (Jhres+(-Jph)+(-Jhl)+(-2*Juni)+(-JNCa))*F*1/Cmit
Jgpd = NaN; Vmmit = 0.0144; JPYR = NaN; JLDH = NaN; Vi = 0.53Reaction: PYR = (Jgpd+(-JPYR)+(-JLDH))*1/(Vi+Vmmit), Rate Law: (Jgpd+(-JPYR)+(-JLDH))*1/(Vi+Vmmit)
Jph = NaN; kATPCa = 9.0E-5; Cac = NaN; kATP = 4.0E-5; JGlu = NaN; Vi = 0.53Reaction: ATP = (-(kATP+kATPCa*Cac)*ATP)+(2*JGlu+0.231*Jph)*1/Vi, Rate Law: (-(kATP+kATPCa*Cac)*ATP)+(2*JGlu+0.231*Jph)*1/Vi

States:

NameDescription
G3P[glyceraldehyde 3-phosphate]
ATP[ATP]
PYR[pyruvate]
NADHm[NADH]
Vm[membrane potential]
NADHc[NADH]
Cam[calcium(2+)]

Fridlyand2010_GlucoseSensitivity_B: BIOMD0000000349v0.0.1

This a model from the article: Glucose sensing in the pancreatic beta cell: a computational systems analysis. Fridl…

Details

Pancreatic beta-cells respond to rising blood glucose by increasing oxidative metabolism, leading to an increased ATP/ADP ratio in the cytoplasm. This leads to a closure of KATP channels, depolarization of the plasma membrane, influx of calcium and the eventual secretion of insulin. Such mechanism suggests that beta-cell metabolism should have a functional regulation specific to secretion, as opposed to coupling to contraction. The goal of this work is to uncover contributions of the cytoplasmic and mitochondrial processes in this secretory coupling mechanism using mathematical modeling in a systems biology approach.We describe a mathematical model of beta-cell sensitivity to glucose. The cytoplasmic part of the model includes equations describing glucokinase, glycolysis, pyruvate reduction, NADH and ATP production and consumption. The mitochondrial part begins with production of NADH, which is regulated by pyruvate dehydrogenase. NADH is used in the electron transport chain to establish a proton motive force, driving the F1F0 ATPase. Redox shuttles and mitochondrial Ca2+ handling were also modeled.The model correctly predicts changes in the ATP/ADP ratio, Ca2+ and other metabolic parameters in response to changes in substrate delivery at steady-state and during cytoplasmic Ca2+ oscillations. Our analysis of the model simulations suggests that the mitochondrial membrane potential should be relatively lower in beta cells compared with other cell types to permit precise mitochondrial regulation of the cytoplasmic ATP/ADP ratio. This key difference may follow from a relative reduction in respiratory activity. The model demonstrates how activity of lactate dehydrogenase, uncoupling proteins and the redox shuttles can regulate beta-cell function in concert; that independent oscillations of cytoplasmic Ca2+ can lead to slow coupled metabolic oscillations; and that the relatively low production rate of reactive oxygen species in beta-cells under physiological conditions is a consequence of the relatively decreased mitochondrial membrane potential.This comprehensive model predicts a special role for mitochondrial control mechanisms in insulin secretion and ROS generation in the beta cell. The model can be used for testing and generating control hypotheses and will help to provide a more complete understanding of beta-cell glucose-sensing central to the physiology and pathology of pancreatic beta-cells. link: http://identifiers.org/pubmed/20497556

Parameters:

NameDescription
Jgpd = NaN; kgpd = 1.0E-5; JGlu = NaN; Vi = 0.53Reaction: G3P = (2*JGlu+(-Jgpd))*1/Vi+(-kgpd*G3P), Rate Law: (2*JGlu+(-Jgpd))*1/Vi+(-kgpd*G3P)
Jtnadh = NaN; Jgpd = NaN; JLDH = NaN; Vi = 0.53; knadhc = 1.0E-4Reaction: NADHc = (Jgpd+(-Jtnadh)+(-JLDH))*1/Vi+(-knadhc*NADHc), Rate Law: (Jgpd+(-Jtnadh)+(-JLDH))*1/Vi+(-knadhc*NADHc)
fm = 3.0E-4; Vmmit = 0.0144; Juni = NaN; JNCa = NaNReaction: Cam = fm*(Juni+(-JNCa))*1/Vmmit, Rate Law: fm*(Juni+(-JNCa))*1/Vmmit
Jtnadh = NaN; knadhm = 1.0E-4; Vmmit = 0.0144; JPYR = NaN; Jhres = NaNReaction: NADHm = (4.6*JPYR+(-0.1*Jhres)+Jtnadh)*1/Vmmit+(-knadhm*NADHm), Rate Law: (4.6*JPYR+(-0.1*Jhres)+Jtnadh)*1/Vmmit+(-knadhm*NADHm)
Jph = NaN; Cmit = 1.82; Juni = NaN; JNCa = NaN; Jhres = NaN; Jhl = NaNReaction: Vm = (Jhres+(-Jph)+(-Jhl)+(-2*Juni)+(-JNCa))*1/Cmit, Rate Law: (Jhres+(-Jph)+(-Jhl)+(-2*Juni)+(-JNCa))*1/Cmit
Jgpd = NaN; Vmmit = 0.0144; JPYR = NaN; JLDH = NaN; Vi = 0.53Reaction: PYR = (Jgpd+(-JPYR)+(-JLDH))*1/(Vi+Vmmit), Rate Law: (Jgpd+(-JPYR)+(-JLDH))*1/(Vi+Vmmit)
Jph = NaN; kATPCa = 9.0E-5; Cac = NaN; kATP = 4.0E-5; JGlu = NaN; Vi = 0.53Reaction: ATP = (-(kATP+kATPCa*Cac)*ATP)+(2*JGlu+0.231*Jph)*1/Vi, Rate Law: (-(kATP+kATPCa*Cac)*ATP)+(2*JGlu+0.231*Jph)*1/Vi

States:

NameDescription
Cam[calcium(2+)]
ATP[ATP]
PYR[pyruvate]
NADHm[NADH]
Vm[membrane potential]
NADHc[NADH]
G3P[glyceraldehyde 3-phosphate]

Friedland2009_Ara_RTC3_counter: BIOMD0000000301v0.0.1

This is the model of the RTC3 counter described in the article: **Synthetic gene networks that count.** Friedland AE…

Details

Synthetic gene networks can be constructed to emulate digital circuits and devices, giving one the ability to program and design cells with some of the principles of modern computing, such as counting. A cellular counter would enable complex synthetic programming and a variety of biotechnology applications. Here, we report two complementary synthetic genetic counters in Escherichia coli that can count up to three induction events: the first, a riboregulated transcriptional cascade, and the second, a recombinase-based cascade of memory units. These modular devices permit counting of varied user-defined inputs over a range of frequencies and can be expanded to count higher numbers. link: http://identifiers.org/pubmed/19478183

Parameters:

NameDescription
sT = 0.8467; k_ara = 0.0571; s0_taRNA = 8.0E-4Reaction: => taRNA; ara, Rate Law: cell*(sT*ara/(ara+k_ara)+s0_taRNA)
d_pT3 = 0.0069Reaction: pT3 =>, Rate Law: cell*d_pT3*pT3
d_mGFP = 0.07Reaction: mGFPcr =>, Rate Law: cell*d_mGFP*mGFPcr
k_pT7 = 3.8009; n7 = 2.602; km7 = 3.0455; s0_mT3cr = 3.0E-4Reaction: => mT3cr; pT7, Rate Law: cell*(s0_mT3cr+k_pT7*pT7^n7/(km7^n7+pT7^n7))
s0_pT3 = 0.0; s_pT3k = 0.0115Reaction: => pT3; taRNA, mT3cr, Rate Law: cell*(s0_pT3*mT3cr+s_pT3k*taRNA*mT3cr)
s_pT7k = 0.0766; s0_pT7 = 3.0E-4Reaction: => pT7; taRNA, mT7cr, Rate Law: cell*(s0_pT7*mT7cr+s_pT7k*mT7cr*taRNA)
d_pGFP = 0.003Reaction: pGFP =>, Rate Law: cell*d_pGFP*pGFP
pulse_flag = 0.0; cAra = 3.0E-4; dAra = 0.1201Reaction: ara =>, Rate Law: cell*piecewise(cAra, (pulse_flag == 1) && (ara > 0), dAra*ara)
d_taRNA = 0.1177Reaction: taRNA =>, Rate Law: cell*d_taRNA*taRNA
s0_pGFP = 0.1007; s_pGFPk = 0.9923Reaction: => pGFP; taRNA, mGFPcr, Rate Law: cell*(s0_pGFP*mGFPcr+s_pGFPk*mGFPcr*taRNA)
km3 = 7.9075; k_pT3 = 3.006; s0_mGFPcr = 0.0123; n3 = 0.8892Reaction: => mGFPcr; pT3, Rate Law: cell*(s0_mGFPcr+k_pT3*pT3^n3/(km3^n3+pT3^n3))
s0_mT7cr = 0.0252Reaction: => mT7cr, Rate Law: cell*s0_mT7cr
d_mT3 = 0.0701Reaction: mT3cr =>, Rate Law: cell*d_mT3*mT3cr
d_pT7 = 0.0056Reaction: pT7 =>, Rate Law: cell*d_pT7*pT7
d_mT7 = 0.0706Reaction: mT7cr =>, Rate Law: cell*d_mT7*mT7cr

States:

NameDescription
pGFP[Green fluorescent protein; IPR000786]
mT7cr[T7 RNA polymerase; messenger RNA]
ara[L-Arabinose; L-arabinopyranose]
mGFPcr[IPR000786; messenger RNA]
pT3[DNA-directed RNA polymerase; DNA-directed RNA polymerase complex]
pT7[T7 RNA polymerase; DNA-directed RNA polymerase complex]
mT3cr[DNA-directed RNA polymerase; messenger RNA]
taRNA[positive regulation of translation, ncRNA-mediated; ribonucleic acid]

Fuentealb2016 - Genome-scale metabolic reconstruction (iPF215) of Piscirickettsia salmonis LF-89: MODEL1610250000v0.0.1

Fuentealb2016 - Genome-scale metabolic reconstruction (iPF215) of Piscirickettsia salmonis LF-89This model is described…

Details

Piscirickettsia salmonis is a fish bacterium that causes the disease piscirickettsiosis in salmonids. This pathology is partially controlled by vaccines. The lack of knowledge has hindered its culture on laboratory and industrial scale. The study describes the metabolic phenotype of P. salmonis in culture. This study presents the first genome-scale model (iPF215) of the LF-89 strain of P. salmonis, describing the central metabolic pathway, biosynthesis and molecule degradation and transport mechanisms. The model was adjusted with experiment data, allowing the identification of the capacities that were not predicted by the automatic annotation of the genome sequences. The iPF215 model is comprised of 417 metabolites, 445 reactions and 215 genes, was used to reproduce the growth of P. salmonis (μmax 0.052±0.005h-1). The metabolic reconstruction of the P. salmonis LF-89 strain obtained in this research provides a baseline that describes the metabolic capacities of the bacterium and is the basis for developing improvements to its cultivation for vaccine formulation. link: http://identifiers.org/pubmed/27788423

Fuentes2005_ZymogenActivation: BIOMD0000000092v0.0.1

. . . **[SBML](http://www.sbml.org/) level 2 code generated for the JWS Online project by Jacky Snoep using [PySCeS]…

Details

A mathematical description was made of an autocatalytic zymogen activation mechanism involving both intra- and intermolecular routes. The reversible formation of an active intermediary enzyme-zymogen complex was included in the intermolecular activation route, thus allowing a Michaelis-Menten constant to be defined for the activation of the zymogen towards the active enzyme. Time-concentration equations describing the evolution of the species involved in the system were obtained. In addition, we have derived the corresponding kinetic equations for particular cases of the general model studied. Experimental design and kinetic data analysis procedures to evaluate the kinetic parameters, based on the derived kinetic equations, are suggested. The validity of the results obtained were checked by using simulated progress curves of the species involved. The model is generally good enough to be applied to the experimental kinetic study of the activation of different zymogens of physiological interest. The system is illustrated by following the transformation kinetics of pepsinogen into pepsin. link: http://identifiers.org/pubmed/15634334

Parameters:

NameDescription
k1=0.004 sec_invReaction: z => w + e, Rate Law: compartment*k1*z
k3=5.4E-4 sec_invReaction: ez => w + e, Rate Law: compartment*k3*ez
k21=1000.0 M_inv_sec_inv; k22=2.1E-4 sec_invReaction: z + e => ez, Rate Law: compartment*(k21*e*z-k22*ez)

States:

NameDescription
wPeptide
eEnzyme
z[zymogen granule]
ez[protein complex]

Fujita2010_Akt_Signalling_EGF: BIOMD0000000262v0.0.1

EGF dependent Akt pathway model made by Kazuhiro A. Fujita. This is the EGF dependent Akt pathway model described in:…

Details

In cellular signal transduction, the information in an external stimulus is encoded in temporal patterns in the activities of signaling molecules; for example, pulses of a stimulus may produce an increasing response or may produce pulsatile responses in the signaling molecules. Here, we show how the Akt pathway, which is involved in cell growth, specifically transmits temporal information contained in upstream signals to downstream effectors. We modeled the epidermal growth factor (EGF)-dependent Akt pathway in PC12 cells on the basis of experimental results. We obtained counterintuitive results indicating that the sizes of the peak amplitudes of receptor and downstream effector phosphorylation were decoupled; weak, sustained EGF receptor (EGFR) phosphorylation, rather than strong, transient phosphorylation, strongly induced phosphorylation of the ribosomal protein S6, a molecule downstream of Akt. Using frequency response analysis, we found that a three-component Akt pathway exhibited the property of a low-pass filter and that this property could explain decoupling of the peak amplitudes of receptor phosphorylation and that of downstream effectors. Furthermore, we found that lapatinib, an EGFR inhibitor used as an anticancer drug, converted strong, transient Akt phosphorylation into weak, sustained Akt phosphorylation, and, because of the low-pass filter characteristics of the Akt pathway, this led to stronger S6 phosphorylation than occurred in the absence of the inhibitor. Thus, an EGFR inhibitor can potentially act as a downstream activator of some effectors. link: http://identifiers.org/pubmed/20664065

Parameters:

NameDescription
EGFR_turnover = 1.06386129269658E-4 per secondReaction: pro_EGFR => EGFR, Rate Law: Cell*EGFR_turnover*pro_EGFR
k1=0.00121498 per secondReaction: pAkt_S6 => pAkt + pS6, Rate Law: Cell*k1*pAkt_S6
k1=2.10189E-6 per conc per second; k2=5.1794E-15 per secondReaction: pAkt + S6 => pAkt_S6, Rate Law: Cell*(k1*pAkt*S6-k2*pAkt_S6)
k1=0.0305684 per secondReaction: pEGFR_Akt => pEGFR + pAkt, Rate Law: Cell*k1*pEGFR_Akt
EGF_conc_ramp = 30.0 ng_per_ml; EGF_conc_impulse = 0.0 ng_per_ml; EGF_conc_step = 0.0 ng_per_ml; ramp_time = 3600.0 seconds; pulse_time = 60.0 secondsReaction: EGF = EGF_conc_step+piecewise(EGF_conc_impulse, time <= pulse_time, 0)+EGF_conc_ramp*time/ramp_time, Rate Law: missing
k1=0.0327962 per secondReaction: pAkt => Akt, Rate Law: Cell*k1*pAkt
k1=0.0997194 per secondReaction: pEGFR =>, Rate Law: Cell*k1*pEGFR
k1=0.00113102 per secondReaction: pS6 => S6, Rate Law: Cell*k1*pS6
k1=0.00673816; k2=0.040749 per secondReaction: EGF + EGFR => EGF_EGFR, Rate Law: Cell*(k1*EGF*EGFR-k2*EGF_EGFR)
k2=0.00517473 per second; k1=1.5543E-5 per conc per secondReaction: pEGFR + Akt => pEGFR_Akt, Rate Law: Cell*(k1*pEGFR*Akt-k2*pEGFR_Akt)
k1=0.0192391 per secondReaction: EGF_EGFR => pEGFR, Rate Law: Cell*k1*EGF_EGFR

States:

NameDescription
EGFR[Egfr]
EGF EGFR[Egfr; Pro-epidermal growth factor]
Akt[RAC-gamma serine/threonine-protein kinase]
EGF[IPR006209]
pro EGFR[Egfr]
pS6[40S ribosomal protein S6; Phosphoprotein]
pAkt[RAC-gamma serine/threonine-protein kinase; Phosphoprotein]
pEGFR[Egfr]
S6[40S ribosomal protein S6]
pEGFR Akt[Egfr; RAC-gamma serine/threonine-protein kinase; Phosphoprotein]
pAkt S6[40S ribosomal protein S6; RAC-gamma serine/threonine-protein kinase; Phosphoprotein]

Fujita2010_Akt_Signalling_EGFRinhib: BIOMD0000000264v0.0.1

Akt pathway model with EGFR inhibitor made by Kazuhiro A. Fujita. This is the Akt pathway model with an EGFR inhibitor…

Details

In cellular signal transduction, the information in an external stimulus is encoded in temporal patterns in the activities of signaling molecules; for example, pulses of a stimulus may produce an increasing response or may produce pulsatile responses in the signaling molecules. Here, we show how the Akt pathway, which is involved in cell growth, specifically transmits temporal information contained in upstream signals to downstream effectors. We modeled the epidermal growth factor (EGF)-dependent Akt pathway in PC12 cells on the basis of experimental results. We obtained counterintuitive results indicating that the sizes of the peak amplitudes of receptor and downstream effector phosphorylation were decoupled; weak, sustained EGF receptor (EGFR) phosphorylation, rather than strong, transient phosphorylation, strongly induced phosphorylation of the ribosomal protein S6, a molecule downstream of Akt. Using frequency response analysis, we found that a three-component Akt pathway exhibited the property of a low-pass filter and that this property could explain decoupling of the peak amplitudes of receptor phosphorylation and that of downstream effectors. Furthermore, we found that lapatinib, an EGFR inhibitor used as an anticancer drug, converted strong, transient Akt phosphorylation into weak, sustained Akt phosphorylation, and, because of the low-pass filter characteristics of the Akt pathway, this led to stronger S6 phosphorylation than occurred in the absence of the inhibitor. Thus, an EGFR inhibitor can potentially act as a downstream activator of some effectors. link: http://identifiers.org/pubmed/20664065

Parameters:

NameDescription
k1=0.00121498 per secondReaction: pAkt_S6 => pAkt + pS6, Rate Law: Cell*k1*pAkt_S6
k1=2.10189E-6 per conc per second; k2=5.1794E-15 per secondReaction: pAkt + S6 => pAkt_S6, Rate Law: Cell*(k1*pAkt*S6-k2*pAkt_S6)
k1=0.0327962 per secondReaction: pAkt => Akt, Rate Law: Cell*k1*pAkt
EGF_conc_pulse = 0.0 ng_per_ml; EGF_conc_ramp = 0.0 ng_per_ml; EGF_conc_step = 30.0 ng_per_ml; ramp_time = 3600.0 seconds; pulse_time = 60.0 secondsReaction: EGF = EGF_conc_step+piecewise(EGF_conc_pulse, time <= pulse_time, 0)+EGF_conc_ramp*time/ramp_time, Rate Law: missing
EGFR_turnover = 1.06386E-4 per secondReaction: pro_EGFR => EGFR, Rate Law: Cell*EGFR_turnover*pro_EGFR
k1=0.0997194 per secondReaction: pEGFR =>, Rate Law: Cell*k1*pEGFR
k1=0.0528141 per secondReaction: pEGFR_Akt => pEGFR + pAkt, Rate Law: Cell*k1*pEGFR_Akt
inhibitor_binding_kf = 2.43466029020655E-5 per conc per second; inhibitor_binding_kb = 5.25096686262403E-5 per secondReaction: Inhibitor + EGFR => EGFR_i, Rate Law: Cell*(inhibitor_binding_kf*Inhibitor*EGFR-inhibitor_binding_kb*EGFR_i)
k1=0.00113102 per secondReaction: pS6 => S6, Rate Law: Cell*k1*pS6
k2=0.00517473 per second; k1=1.5543E-5 per conc per secondReaction: pEGFR + Akt => pEGFR_Akt, Rate Law: Cell*(k1*pEGFR*Akt-k2*pEGFR_Akt)
k1=0.0192391 per secondReaction: EGF_EGFR => pEGFR, Rate Law: Cell*k1*EGF_EGFR
EGF_binding_kf = 0.00673816 ml_per_ng_per_sec; EGF_binding_kb = 0.040749 per secondReaction: EGF + EGFR_i => EGF_EGFR_i, Rate Law: Cell*(EGF_binding_kf*EGF*EGFR_i-EGF_binding_kb*EGF_EGFR_i)

States:

NameDescription
Inhibitor[Lapatinib (INN); epidermal growth factor receptor binding]
EGFR[Egfr]
EGF EGFR[Egfr; Pro-epidermal growth factor]
Akt[RAC-gamma serine/threonine-protein kinase]
EGF EGFR i[lapatinib; Egfr; Pro-epidermal growth factor]
EGFR i[lapatinib; Egfr]
pro EGFR[Egfr]
EGF[Pro-epidermal growth factor; epidermal growth factor receptor binding]
pS6[Phosphoprotein; 40S ribosomal protein S6]
pAkt[Phosphoprotein; RAC-gamma serine/threonine-protein kinase]
pEGFR[Egfr; Phosphoprotein]
S6[40S ribosomal protein S6]
pEGFR Akt[RAC-gamma serine/threonine-protein kinase; Egfr; Phosphoprotein]
pAkt S6[RAC-gamma serine/threonine-protein kinase; 40S ribosomal protein S6; Phosphoprotein]

Fujita2010_Akt_Signalling_NGF: BIOMD0000000263v0.0.1

NGF dependent Akt pathway model made by Kazuhiro A. Fujita. This is the NGF dependent Akt pathway model described in:…

Details

In cellular signal transduction, the information in an external stimulus is encoded in temporal patterns in the activities of signaling molecules; for example, pulses of a stimulus may produce an increasing response or may produce pulsatile responses in the signaling molecules. Here, we show how the Akt pathway, which is involved in cell growth, specifically transmits temporal information contained in upstream signals to downstream effectors. We modeled the epidermal growth factor (EGF)-dependent Akt pathway in PC12 cells on the basis of experimental results. We obtained counterintuitive results indicating that the sizes of the peak amplitudes of receptor and downstream effector phosphorylation were decoupled; weak, sustained EGF receptor (EGFR) phosphorylation, rather than strong, transient phosphorylation, strongly induced phosphorylation of the ribosomal protein S6, a molecule downstream of Akt. Using frequency response analysis, we found that a three-component Akt pathway exhibited the property of a low-pass filter and that this property could explain decoupling of the peak amplitudes of receptor phosphorylation and that of downstream effectors. Furthermore, we found that lapatinib, an EGFR inhibitor used as an anticancer drug, converted strong, transient Akt phosphorylation into weak, sustained Akt phosphorylation, and, because of the low-pass filter characteristics of the Akt pathway, this led to stronger S6 phosphorylation than occurred in the absence of the inhibitor. Thus, an EGFR inhibitor can potentially act as a downstream activator of some effectors. link: http://identifiers.org/pubmed/20664065

Parameters:

NameDescription
NGF_conc_step = 0.0 ng_per_ml; NGF_conc_pulse = 0.0 ng_per_ml; NGF_conc_ramp = 30.0 ng_per_ml; ramp_time = 3600.0 seconds; pulse_time = 60.0 secondsReaction: NGF = NGF_conc_step+piecewise(NGF_conc_pulse, time <= pulse_time, 0)+NGF_conc_ramp*time/ramp_time, Rate Law: missing
k2=1.47518E-10 per second; k1=0.0882701 per conc per secondReaction: pTrkA + Akt => pTrkA_Akt, Rate Law: Cell*(k1*pTrkA*Akt-k2*pTrkA_Akt)
k1=0.0202517Reaction: pTrkA_Akt => pTrkA + pAkt, Rate Law: Cell*k1*pTrkA_Akt
k1=0.0056515 per secondReaction: pAkt_S6 => pAkt + pS6, Rate Law: Cell*k1*pAkt_S6
k2=5.23519 per second; k1=68.3666 per conc per secondReaction: pAkt + S6 => pAkt_S6, Rate Law: Cell*(k1*pAkt*S6-k2*pAkt_S6)
k1=2.93167E-4 per secondReaction: pS6 => S6, Rate Law: Cell*k1*pS6
TrkA_turnover = 0.0011032440769796 per secondReaction: pro_TrkA => TrkA, Rate Law: Cell*TrkA_turnover*pro_TrkA
k2=0.0133747; k1=0.00269408Reaction: NGF + TrkA => NGF_TrkA, Rate Law: Cell*(k1*NGF*TrkA-k2*NGF_TrkA)
k1=0.00833178 per secondReaction: NGF_TrkA => pTrkA, Rate Law: Cell*k1*NGF_TrkA
k1=1.28135 per secondReaction: pAkt => Akt, Rate Law: Cell*k1*pAkt
k1=0.0684084 per secondReaction: pTrkA =>, Rate Law: Cell*k1*pTrkA

States:

NameDescription
NGF TrkA[High affinity nerve growth factor receptor; Beta-nerve growth factor]
TrkA[High affinity nerve growth factor receptor]
NGF[Beta-nerve growth factor]
Akt[RAC-gamma serine/threonine-protein kinase]
pTrkA[High affinity nerve growth factor receptor]
pTrkA Akt[High affinity nerve growth factor receptor; RAC-gamma serine/threonine-protein kinase; Phosphoprotein]
pS6[Phosphoprotein; 40S ribosomal protein S6]
pro TrkA[High affinity nerve growth factor receptor]
pAkt[Phosphoprotein; Active AKT [cytosol]]
S6[40S ribosomal protein S6]
pAkt S6[RAC-gamma serine/threonine-protein kinase; 40S ribosomal protein S6; Phosphoprotein]

Fung2005_Metabolic_Oscillator: BIOMD0000000067v0.0.1

# A Synthetic Gene-Metabolic Oscillator **Reference:**[*Fung et al; Nature (2005) 435:118-122*](http://www.nature.com/na…

Details

Autonomous oscillations found in gene expression and metabolic, cardiac and neuronal systems have attracted significant attention both because of their obvious biological roles and their intriguing dynamics. In addition, de novo designed oscillators have been demonstrated, using components that are not part of the natural oscillators. Such oscillators are useful in testing the design principles and in exploring potential applications not limited by natural cellular behaviour. To achieve transcriptional and metabolic integration characteristic of natural oscillators, here we designed and constructed a synthetic circuit in Escherichia coli K12, using glycolytic flux to generate oscillation through the signalling metabolite acetyl phosphate. If two metabolite pools are interconverted by two enzymes that are placed under the transcriptional control of acetyl phosphate, the system oscillates when the glycolytic rate exceeds a critical value. We used bifurcation analysis to identify the boundaries of oscillation, and verified these experimentally. This work demonstrates the possibility of using metabolic flux as a control factor in system-wide oscillation, as well as the predictability of a de novo gene-metabolic circuit designed using nonlinear dynamic analysis. link: http://identifiers.org/pubmed/15875027

Parameters:

NameDescription
S0 = 0.5Reaction: => AcCoA, Rate Law: compartment*S0
kTCA = 10.0Reaction: AcCoA =>, Rate Law: compartment*kTCA*AcCoA
KM2 = 0.1; k2 = 0.8Reaction: OAc => AcCoA; Acs, Rate Law: compartment*k2*Acs*OAc/(KM2+OAc)
kAck_f = 1.0; kAck_r = 1.0Reaction: AcP => OAc, Rate Law: compartment*(kAck_f*AcP-kAck_r*OAc)
k3 = 0.01Reaction: HOAc => HOAc_E, Rate Law: compartment*k3*(HOAc-HOAc_E)
KM1 = 0.06; k1 = 80.0Reaction: AcCoA => AcP; Pta, Rate Law: compartment*k1*Pta*AcCoA/(KM1+AcCoA)
Kg3 = 0.001; alpha3 = 2.0; n = 2.0; alpha0 = 0.0Reaction: => Pta; LacI, Rate Law: alpha3/(1+(LacI/Kg3)^n)+alpha0
kd = 0.06Reaction: LacI =>, Rate Law: compartment*kd*LacI
Kg1 = 10.0; alpha1 = 0.1; n = 2.0; alpha0 = 0.0Reaction: => LacI; AcP, Rate Law: compartment*(alpha1*(AcP/Kg1)^n/(1+(AcP/Kg1)^n)+alpha0)
alpha2 = 2.0; n = 2.0; alpha0 = 0.0; Kg2 = 10.0Reaction: => Acs; AcP, Rate Law: compartment*(alpha2*(AcP/Kg2)^n/(1+(AcP/Kg2)^n)+alpha0)
H = 1.0E-7; Keq = 5.0E-4; C = 100.0Reaction: OAc => HOAc, Rate Law: compartment*C*(OAc*H-Keq*HOAc)

States:

NameDescription
LacI[transcriptional repressor complex]
HOAc E[acetate; Acetate]
OAc[acetate; Acetate]
AcCoA[acetyl-CoA; Acetyl-CoA]
Pta[Phosphate acetyltransferase]
HOAc[acetate; Acetate]
AcP[acetyl dihydrogen phosphate; Acetyl phosphate]
Acs[Acetyl-coenzyme A synthetase]

Fuss2006_MitoticActivation: BIOMD0000000069v0.0.1

The model was curated with XPP. The figure 3 was successfully reproduced.

Details

MOTIVATION: The protein tyrosine kinase Src is involved in a multitude of biochemical pathways and cellular functions. A complex network of interactions with other kinases and phosphatases obscures its precise mode of operation. RESULTS: We have constructed a semi-quantitative computational dynamic systems model of the activation of Src at mitosis based on protein interactions described in the literature. Through numerical simulation and bifurcation analysis we show that Src regulation involves a bistable switch, a pattern increasingly recognised as essential to biochemical signalling. The switch is operated by the tyrosine kinase CSK, which itself is involved in a negative feedback loop with Src. Negative feedback generates an excitable system, which produces transient activation of Src. One of the system parameters, which is linked to the cyclin dependent kinase cdc2, controls excitability via a second bistable switch. This topology allows for differentiated responses to a multitude of signals. The model offers explanations for the existence of the positive and negative feedback loops involving protein tyrosine phosphatase alpha (PTPalpha) and translocation of CSK and predicts a specific relationship between Src phosphorylation and activity. link: http://identifiers.org/pubmed/16873466

Parameters:

NameDescription
k1 = 1.0; k2 = 0.8; ptp_activity = NaNReaction: srci => srco; Cbp_P_CSK, Rate Law: (k2*ptp_activity*srci-k1*Cbp_P_CSK*srco)*default
kCSKoff = 0.01; kCSKon = 0.1Reaction: CSK_cytoplasm + Cbp_P => Cbp_P_CSK, Rate Law: (Cbp_P*kCSKon*CSK_cytoplasm-kCSKoff*Cbp_P_CSK)*default
src_activity = NaN; kCbp = 1.0Reaction: Cbp => Cbp_P, Rate Law: kCbp*src_activity*Cbp*default
src_activity = NaN; p1 = 0.05; k3 = 1.0Reaction: srco => srca, Rate Law: (k3*src_activity*srco-p1*srca)*default
p1 = 0.05; k4 = 10.0Reaction: srcc => srci, Rate Law: default*k4*p1*srcc
p2 = 0.15; kPTP = 1.0; src_activity = NaN; p3 = 0.035Reaction: PTP => PTP_pY789, Rate Law: default*((kPTP*src_activity+p3)*PTP-p2*PTP_pY789)

States:

NameDescription
srcc[Proto-oncogene tyrosine-protein kinase Src]
PTP pY789[Receptor-type tyrosine-protein phosphatase alpha]
Cbp P CSK[Tyrosine-protein kinase CSK; Phosphoprotein associated with glycosphingolipid-enriched microdomains 1]
Cbp P[Phosphoprotein associated with glycosphingolipid-enriched microdomains 1]
srco[Proto-oncogene tyrosine-protein kinase Src]
srci[Proto-oncogene tyrosine-protein kinase Src]
PTP[Receptor-type tyrosine-protein phosphatase alpha]
Cbp[Phosphoprotein associated with glycosphingolipid-enriched microdomains 1]
CSK cytoplasm[Tyrosine-protein kinase CSK]
srca[Proto-oncogene tyrosine-protein kinase Src]

Förster2008 - Genome-scale metabolic network of Saccharamyces cerevisiae (iFF708): MODEL1507180012v0.0.1

Förster2008 - Genome-scale metabolic network of Saccharamyces cerevisiae (iFF708)This model is described in the article:…

Details

The metabolic network in the yeast Saccharomyces cerevisiae was reconstructed using currently available genomic, biochemical, and physiological information. The metabolic reactions were compartmentalized between the cytosol and the mitochondria, and transport steps between the compartments and the environment were included. A total of 708 structural open reading frames (ORFs) were accounted for in the reconstructed network, corresponding to 1035 metabolic reactions. Further, 140 reactions were included on the basis of biochemical evidence resulting in a genome-scale reconstructed metabolic network containing 1175 metabolic reactions and 584 metabolites. The number of gene functions included in the reconstructed network corresponds to approximately 16% of all characterized ORFs in S. cerevisiae. Using the reconstructed network, the metabolic capabilities of S. cerevisiae were calculated and compared with Escherichia coli. The reconstructed metabolic network is the first comprehensive network for a eukaryotic organism, and it may be used as the basis for in silico analysis of phenotypic functions. link: http://identifiers.org/pubmed/12566402

G


Gaetano2008_DiabetesProgressionModel: MODEL1112110003v0.0.1

This a model from the article: Mathematical models of diabetes progression. De Gaetano A, Hardy T, Beck B, Abu-Radda…

Details

Few attempts have been made to model mathematically the progression of type 2 diabetes. A realistic representation of the long-term physiological adaptation to developing insulin resistance is necessary for effectively designing clinical trials and evaluating diabetes prevention or disease modification therapies. Writing a good model for diabetes progression is difficult because the long time span of the disease makes experimental verification of modeling hypotheses extremely awkward. In this context, it is of primary importance that the assumptions underlying the model equations properly reflect established physiology and that the mathematical formulation of the model give rise only to physically plausible behavior of the solutions. In the present work, a model of the pancreatic islet compensation is formulated, its physiological assumptions are presented, some fundamental qualitative characteristics of its solutions are established, the numerical values assigned to its parameters are extensively discussed (also with reference to available cross-sectional epidemiologic data), and its performance over the span of a lifetime is simulated under various conditions, including worsening insulin resistance and primary replication defects. The differences with respect to two previously proposed models of diabetes progression are highlighted, and therefore, the model is proposed as a realistic, robust description of the evolution of the compensation of the glucose-insulin system in healthy and diabetic individuals. Model simulations can be run from the authors' web page. link: http://identifiers.org/pubmed/18780774

Galante2012 - B7-H1 and a Mathematical Model for Cytotoxic T Cell and Tumor Cell Interaction: BIOMD0000000812v0.0.1

This is a mathematical model that describes the interactions between cytotoxic T cells and tumor cells as influenced by…

Details

The surface protein B7-H1, also called PD-L1 and CD274, is found on carcinomas of the lung, ovary, colon, and melanomas but not on most normal tissues. B7-H1 has been experimentally determined to be an antiapoptotic receptor on cancer cells, where B7-H1-positive cancer cells have been shown to be immune resistant, and in vitro experiments and mouse models have shown that B7-H1-negative tumor cells are significantly more susceptible to being repressed by the immune system. We derive a new mathematical model for studying the interaction between cytotoxic T cells and tumor cells as affected by B7-H1. By integrating experimental data into the model, we isolate the parameters that control the dynamics and obtain insights on the mechanisms that control apoptosis. link: http://identifiers.org/pubmed/21656310

Parameters:

NameDescription
k_p = 0.097; k_m_2 = 80.0; k_m_1 = 2.2Reaction: => P_Perforin; E_CTL, C_Cancer_Uncomplexed, Rate Law: compartment*k_p*E_CTL/((k_m_1+E_CTL)*k_m_2*C_Cancer_Uncomplexed)
ModelValue_11 = 1.0Reaction: E_CTL = ModelValue_11-X_Complex, Rate Law: missing
k_2 = 1.0E-4Reaction: X_Complex => C_Cancer_Uncomplexed, Rate Law: compartment*k_2*X_Complex
k_3 = 1.0E-4Reaction: X_Complex =>, Rate Law: compartment*k_3*X_Complex
k = 0.035Reaction: => C_Cancer_Uncomplexed, Rate Law: compartment*k*C_Cancer_Uncomplexed
k_1 = 1.0E-4Reaction: C_Cancer_Uncomplexed => X_Complex; E_CTL, Rate Law: compartment*k_1*C_Cancer_Uncomplexed*E_CTL
k_5 = 0.003Reaction: C_Cancer_Uncomplexed =>, Rate Law: compartment*k_5*C_Cancer_Uncomplexed^2
k_4 = 3.0Reaction: P_Perforin + C_Cancer_Uncomplexed =>, Rate Law: compartment*k_4*P_Perforin*C_Cancer_Uncomplexed

States:

NameDescription
P Perforin[Perforin]
X Complex[cytotoxic T cell; neoplastic cell; Complex]
T ast[neoplastic cell]
E CTL[cytotoxic T cell]
T Cancer Cell Total[neoplastic cell]
C Cancer Uncomplexed[neoplastic cell]

Galazzo1990_FermentationPathwayKinetics: BIOMD0000000063v0.0.1

This a model from the article: Fermentation pathway kinetics and metabolic flux control in suspended and immobilize…

Details

Measurements of rates of glucose uptake and of glycerol and ethanol formation combined with knowledge of the metabolic pathways involved in S. cerevisiae were employed to obtain in vivo rates of reaction catalysed by pathway enzymes for suspended and alginate-entrapped cells at pH 4.5 and 5.5. Intracellular concentrations of substrates and effectors for most key pathway enzymes were estimated from in vivo phosphorus-31 nuclear magnetic resonance measurements. These data show the validity in vivo of kinetic models previously proposed for phosphofructokinase and pyruvate kinase based on in vitro studies. Kinetic representations of hexokinase, glycogen synthetase, and glyceraldehyde 3-phosphate dehydrogenase, which incorporate major regulatory properties of these enzymes, are all consistent with the in vivo data. This detailed model of pathway kinetics and these data on intracellular metabolite concentrations allow evaluation of flux-control coefficients for all key enzymes involved in glucose catabolism under the four different cell environments examined. This analysis indicates that alginate entrapment increases the glucose uptake rate and shifts the step most influencing ethanol production from glucose uptake to phosphofructokinase. The rate of ATP utilization in these nongrowing cells strongly limits ethanol production at pH 5.5 but is relatively insignificant at pH 4.5. link: http://identifiers.org/doi/10.1016/0141-0229(90)90033-M

Parameters:

NameDescription
Ks2Glc=0.0062 milliMolar; Km2Glc=0.11 milliMolar; Vm2=68.5 mM per minute; Km2ATP=0.1 milliMolarReaction: ATP + Glci => G6P, Rate Law: cytoplasm*Vm2/(1+Km2Glc/Glci+Km2ATP/ATP+Ks2Glc*Km2ATP/(Glci*ATP))
g6R=0.1 dimensionless; K6ADP=5.0 milliMolar; g6T=1.0 dimensionless; K6FDP=0.2 milliMolar; c6FDP=0.01 dimensionless; c6PEP=1.58793E-4 dimensionless; c6ADP=1.0 dimensionless; h6=1.14815E-7 dimensionless; Vm6=3440.0 mM per minute; K6PEP=0.00793966 milliMolar; q6=1.0 dimensionless; L60=164.084 dimensionlessReaction: PEP => ATP + EtOH; FDP, Rate Law: cytoplasm*Vm6*PEP/K6PEP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP*(g6R*(1+PEP/K6PEP+0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6R*PEP/K6PEP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP)+q6*L60*((1+c6FDP*FDP/K6FDP)/(1+FDP/K6FDP))^2*g6T*c6PEP*PEP/K6PEP*c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP*(1+c6PEP*PEP/K6PEP+c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6T*c6PEP*PEP/K6PEP*c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP))/((1+9.55*10^-9/h6)*((1+PEP/K6PEP+0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6R*PEP/K6PEP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP)^2+L60*((1+c6FDP*FDP/K6FDP)/(1+FDP/K6FDP))^2*(1+c6PEP*PEP/K6PEP+c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6T*c6PEP*PEP/K6PEP*c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP)^2))
Vm8=25.1 minute_inverseReaction: ATP =>, Rate Law: cytoplasm*Vm8*ATP
g6R=0.1 dimensionless; K6ADP=5.0 milliMolar; g6T=1.0 dimensionless; K6FDP=0.2 milliMolar; c6FDP=0.01 dimensionless; c6PEP=1.58793E-4 dimensionless; c6ADP=1.0 dimensionless; h6=1.14815E-7 dimensionless; Vm7=203.0 mM per minute; K6PEP=0.00793966 milliMolar; q6=1.0 dimensionless; L60=164.084 dimensionlessReaction: FDP => Gly; PEP, ATP, Rate Law: Vm7*cytoplasm*PEP/K6PEP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP*(g6R*(1+PEP/K6PEP+0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6R*PEP/K6PEP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP)+q6*L60*((1+c6FDP*FDP/K6FDP)/(1+FDP/K6FDP))^2*g6T*c6PEP*PEP/K6PEP*c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP*(1+c6PEP*PEP/K6PEP+c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6T*c6PEP*PEP/K6PEP*c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP))/((1+9.55*10^-9/h6)*((1+PEP/K6PEP+0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6R*PEP/K6PEP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP)^2+L60*((1+c6FDP*FDP/K6FDP)/(1+FDP/K6FDP))^2*(1+c6PEP*PEP/K6PEP+c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP+g6T*c6PEP*PEP/K6PEP*c6ADP*0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K6ADP)^2))
NADH=0.0806142 milliMolar; K5NADH=3.0E-4 milliMolar; K5AMP=1.1 milliMolar; K5ADP=1.5 milliMolar; Vm5=49.9 mM per minute; K5NAD=0.18 dimensionless; NAD=1.91939 milliMolar; K5ATP=2.5 milliMolar; K5G3P=0.0025 milliMolarReaction: FDP => ATP + PEP, Rate Law: cytoplasm*Vm5/(1+K5G3P/(0.01*FDP)+(K5NAD/NAD+K5G3P*K5NAD/(NAD*0.01*FDP)+K5G3P*K5NAD*NADH/(NAD*0.01*FDP*K5NADH))*(1+0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5)/K5ADP+((3-ATP)-0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5))/K5AMP+ATP/K5ATP))
Vm1=19.7 mM per minute; Ki1G6P=3.7 minute_inverseReaction: Glco => Glci; G6P, Rate Law: cytoplasm*(Vm1-Ki1G6P*G6P)
c4AMP=0.019 dimensionless; K4F6P=1.0 milliMolar; K4AMP=0.025 milliMolar; L40=3342.0 dimensionless; gT=1.0 dimensionless; c4F6P=5.0E-4 dimensionless; Vm4=31.7 mM per minute; K4ATP=0.06 milliMolar; c4ATP=1.0 dimensionless; g4R=10.0 dimensionlessReaction: ATP + G6P => FDP, Rate Law: cytoplasm*Vm4*g4R*0.3*G6P/K4F6P*ATP/K4ATP*(1+0.3*G6P/K4F6P+ATP/K4ATP+g4R*0.3*G6P/K4F6P*ATP/K4ATP)/((1+0.3*G6P/K4F6P+ATP/K4ATP+g4R*0.3*G6P/K4F6P*ATP/K4ATP)^2+L40*((1+c4AMP*((3-ATP)-0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5))/K4AMP)/(1+((3-ATP)-0.5*((-ATP)+(12*ATP-3*ATP^2)^0.5))/K4AMP))^2*(1+c4F6P*0.3*G6P/K4F6P+c4ATP*ATP/K4ATP+gT*c4F6P*0.3*G6P/K4F6P*c4ATP*ATP/K4ATP)^2)
n3=8.25 dimensionless; K3Gly=2.0 milliMolar; Km30=1.0 milliMolar; Vm3=14.31 mM per minute; Km3G6P=1.1 milliMolarReaction: ATP + G6P => Carbo, Rate Law: cytoplasm*1.1*Vm3*G6P^n3/(K3Gly^n3+G6P^n3)/(1+Km30/0.7*(1+Km3G6P/G6P))

States:

NameDescription
ATP[ATP; ATP]
Gly[glycerol; Glycerol]
Glco[glucose; C00293]
EtOH[ethanol; Ethanol]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]
Carbo[glycogen; trehalose; Glycogen; alpha,alpha-Trehalose; glycogen]
Glci[glucose; C00293]
G6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
FDP[keto-D-fructose 1,6-bisphosphate; D-Fructose 1,6-bisphosphate]

Gall1999_CalciumBursting_BetaCellModel_A: MODEL1201070000v0.0.1

This a model from the article: Effect of Na/Ca exchange on plateau fraction and [Ca]i in models for bursting in pancre…

Details

In the presence of an insulinotropic glucose concentration, beta-cells, in intact pancreatic islets, exhibit periodic bursting electrical activity consisting of an alternation of active and silent phases. The fraction of time spent in the active phase over a period is called the plateau fraction and is correlated with the rate of insulin release. However, the mechanisms that regulate the plateau fraction remain unclear. In this paper we investigate the possible role of the plasma membrane Na+/Ca2+ exchange of the beta-cell in controlling the plateau fraction. We have extended different single-cell models to incorporate this Ca2+-activated electrogenic Ca2+ transporter. We find that the Na+/Ca2+ exchange can provide a physiological mechanism to increase the plateau fraction as the glucose concentration is raised. In addition, we show theoretically that the Na+/Ca2+ exchanger is a key regulator of the cytoplasmic calcium concentration in clusters of heterogeneous cells with gap-junctional electrical coupling. link: http://identifiers.org/pubmed/10388739

Gall1999_CalciumBursting_BetaCellModel_B: MODEL1201070001v0.0.1

This a model from the article: Effect of Na/Ca exchange on plateau fraction and [Ca]i in models for bursting in pancre…

Details

In the presence of an insulinotropic glucose concentration, beta-cells, in intact pancreatic islets, exhibit periodic bursting electrical activity consisting of an alternation of active and silent phases. The fraction of time spent in the active phase over a period is called the plateau fraction and is correlated with the rate of insulin release. However, the mechanisms that regulate the plateau fraction remain unclear. In this paper we investigate the possible role of the plasma membrane Na+/Ca2+ exchange of the beta-cell in controlling the plateau fraction. We have extended different single-cell models to incorporate this Ca2+-activated electrogenic Ca2+ transporter. We find that the Na+/Ca2+ exchange can provide a physiological mechanism to increase the plateau fraction as the glucose concentration is raised. In addition, we show theoretically that the Na+/Ca2+ exchanger is a key regulator of the cytoplasmic calcium concentration in clusters of heterogeneous cells with gap-junctional electrical coupling. link: http://identifiers.org/pubmed/10388739

Gall1999_CalciumBursting_BetaCellModel_C: MODEL1201140000v0.0.1

This a model from the article: Effect of Na/Ca exchange on plateau fraction and [Ca]i in models for bursting in pancre…

Details

In the presence of an insulinotropic glucose concentration, beta-cells, in intact pancreatic islets, exhibit periodic bursting electrical activity consisting of an alternation of active and silent phases. The fraction of time spent in the active phase over a period is called the plateau fraction and is correlated with the rate of insulin release. However, the mechanisms that regulate the plateau fraction remain unclear. In this paper we investigate the possible role of the plasma membrane Na+/Ca2+ exchange of the beta-cell in controlling the plateau fraction. We have extended different single-cell models to incorporate this Ca2+-activated electrogenic Ca2+ transporter. We find that the Na+/Ca2+ exchange can provide a physiological mechanism to increase the plateau fraction as the glucose concentration is raised. In addition, we show theoretically that the Na+/Ca2+ exchanger is a key regulator of the cytoplasmic calcium concentration in clusters of heterogeneous cells with gap-junctional electrical coupling. link: http://identifiers.org/pubmed/10388739

Gallaher2018 - Tumor–Immune dynamics in multiple myeloma: BIOMD0000000743v0.0.1

The paper describes a model on the key components for tumor–immune dynamics in multiple myeloma. Created by COPASI 4.25…

Details

In this work, we analyze a mathematical model we introduced previously for the dynamics of multiple myeloma and the immune system. We focus on four main aspects: (1) obtaining and justifying ranges and values for all parameters in the model; (2) determining a subset of parameters to which the model is most sensitive; (3) determining which parameters in this subset can be uniquely estimated given cer- tain types of data; and (4) exploring the model numerically. Using global sensitivity analysis techniques, we found that the model is most sensitive to certain growth, loss, and efficacy parameters. This anal- ysis provides the foundation for a future application of the model: prediction of optimal combination regimens in patients with multiple myeloma. link: http://identifiers.org/doi/10.1016/j.jtbi.2018.08.037

Parameters:

NameDescription
kr = 80.0 1; rr = 0.0831 1/dReaction: => Tr, Rate Law: compartment*rr*(1-Tr/kr)*Tr
dr = 0.0757 1/dReaction: Tr =>, Rate Law: compartment*dr*Tr
dc = 0.02 1/dReaction: Tc =>, Rate Law: compartment*dc*Tc
kc = 800.0 1; rc = 0.013 1/dReaction: => Tc, Rate Law: compartment*rc*(1-Tc/kc)*Tc
sm = 0.001 1/dReaction: => M, Rate Law: compartment*sm
rn = 0.04 1/d; kn = 450.0 1Reaction: => N, Rate Law: compartment*rn*(1-N/kn)*N
bnm = 150.0 1; amm = 0.5 1; bmm = 3.0 1; brm = 25.0 1; anm = 5.0 1; acnm = 8.0 1; arm = 0.5 1; bcm = 375.0 1; dm = 0.002 1/d; acm = 5.0 1Reaction: M => ; N, Tc, Tr, Rate Law: compartment*M*(anm*N/(bnm+N)+acm*Tc/(bcm+Tc)+acnm*N*Tc/((bnm+N)*(bcm+Tc)))*((1-amm*M/(bmm+M))-arm*Tr/(brm+Tr))*dm
bmc = 3.0 1; kc = 800.0 1; anc = 1.0 1; rc = 0.013 1/d; amc = 5.0 1; bnc = 150.0 1Reaction: => Tc; N, M, Rate Law: compartment*rc*(1-Tc/kc)*(amc*M/(bmc+M)+anc*M/(bnc+M))*Tc
dm = 0.002 1/dReaction: M =>, Rate Law: compartment*M*dm
rn = 0.04 1/d; kn = 450.0 1; bcn = 375.0 1; acn = 1.0 1Reaction: => N; Tc, Rate Law: compartment*rn*(1-N/kn)*acn*Tc/(bcn+Tc)*N
amr = 2.0 1; kr = 80.0 1; bmr = 3.0 1; rr = 0.0831 1/dReaction: => Tr; M, Rate Law: compartment*rr*(1-Tr/kr)*amr*M/(bmr+M)*Tr
km = 10.0 1; rm = 0.0175 1/dReaction: => M, Rate Law: compartment*rm*(1-M/km)*M
dn = 0.025 1/dReaction: N =>, Rate Law: compartment*dn*N
sn = 0.03 1/dReaction: => N, Rate Law: compartment*sn

States:

NameDescription
M[M Protein]
Tc[Activated Mature Cytotoxic T-Lymphocyte; CD8-positive, alpha-beta cytotoxic T cell]
N[Natural Killer Cell; mature natural killer cell]
Tr[regulatory T cell; CD4+ CD25+ Regulatory T Cells]

Ganguli2018-immuno regulatory mechanisms in tumor microenvironment: BIOMD0000000810v0.0.1

This model describes the concept of Cancer Stem Cells(CSC) differentiation and tumor-immune interaction into a generic m…

Details

The tumor microenvironment comprising of the immune cells and cytokines acts as the 'soil' that nourishes a developing tumor. Lack of a comprehensive study of the interactions of this tumor microenvironment with the heterogeneous sub-population of tumor cells that arise from the differentiation of Cancer Stem Cells (CSC), i.e. the 'seed', has limited our understanding of the development of drug resistance and treatment failures in Cancer. Based on this seed and soil hypothesis, for the very first time, we have captured the concept of CSC differentiation and tumor-immune interaction into a generic model that has been validated with known experimental data. Using this model we report that as the CSC differentiation shifts from symmetric to asymmetric pattern, resistant cancer cells start accumulating in the tumor that makes it refractory to therapeutic interventions. Model analyses unveiled the presence of feedback loops that establish the dual role of M2 macrophages in regulating tumor proliferation. The study further revealed oscillations in the tumor sub-populations in the presence of TH1 derived IFN-γ that eliminates CSC; and the role of IL10 feedback in the regulation of TH1/TH2 ratio. These analyses expose important observations that are indicative of Cancer prognosis. Further, the model has been used for testing known treatment protocols to explore the reasons of failure of conventional treatment strategies and propose an improvised protocol that shows promising results in suppressing the proliferation of all the cellular sub-populations of the tumor and restoring a healthy TH1/TH2 ratio that assures better Cancer remission. link: http://identifiers.org/pubmed/30183728

Parameters:

NameDescription
delta_S = 2.0E-7 1/dReaction: Resistant_Stem_Cells_S_R =>, Rate Law: compartment*delta_S*Resistant_Stem_Cells_S_R
k9 = 0.001 1/ml; myu_Th1Ck3 = 0.1245 1/dReaction: => Type_I_T_helper_Cell_T_H1; Cytokine_IL2, Rate Law: compartment*myu_Th1Ck3*Cytokine_IL2*Type_I_T_helper_Cell_T_H1/(Cytokine_IL2+k9)
myu_TcTreg = 1.5E-5 1/d; lambda_Tc3 = 5.0E10 1/mlReaction: Cytotoxic_T_Cells_T_C => ; Regulatory_T_Cells_T_reg, Rate Law: compartment*myu_TcTreg*Cytotoxic_T_Cells_T_C*Regulatory_T_Cells_T_reg/(lambda_Tc3+Regulatory_T_Cells_T_reg)
delta_Th2 = 2.0 1/dReaction: Type_II_T_helper_cells_T_H2 =>, Rate Law: compartment*delta_Th2*Type_II_T_helper_cells_T_H2
gamma_C = 0.1282 1/d; m_C = 0.01; K_tumor = 2.0E10; r_1 = 1.0E-4Reaction: => Cancer_Cells_C, Rate Law: compartment*gamma_C*(1-m_C)*ln(0.5*K_tumor/(Cancer_Cells_C+r_1))
delta_Treg = 1.0 1/dReaction: Regulatory_T_Cells_T_reg =>, Rate Law: compartment*delta_Treg*Regulatory_T_Cells_T_reg
lambda_Tc4 = 100000.0 1/ml; gamma_Tc = 1.0 1/dReaction: => Cytotoxic_T_Cells_T_C; Type_I_T_helper_Cell_T_H1, Rate Law: compartment*gamma_Tc*Cytotoxic_T_Cells_T_C*Type_I_T_helper_Cell_T_H1/(Cytotoxic_T_Cells_T_C+lambda_Tc4)
p_1 = 0.2; p_2 = 0.05; gamma_S = 0.15 1/dReaction: => Resistant_Stem_Cells_S_R, Rate Law: compartment*gamma_S*((1-p_1)-p_2)*Resistant_Stem_Cells_S_R
delta_CR = 5.37E-5 1/dReaction: Resistant_Cancer_Cells_C_R =>, Rate Law: compartment*delta_CR*Resistant_Cancer_Cells_C_R
m_S = 4.0E-7; p_1 = 0.2; p_2 = 0.05; gamma_S = 0.15 1/dReaction: Cancer_Stem_Cells_S => Resistant_Stem_Cells_S_R, Rate Law: compartment*gamma_S*m_S*((1-p_1/2)-p_2)*Cancer_Stem_Cells_S
myu_C2 = 0.9 1/d; k4 = 3.02 1/mlReaction: Cancer_Cells_C => ; Interferon_gamma, Rate Law: compartment*myu_C2*Cancer_Cells_C*Interferon_gamma/(Interferon_gamma+k4)
tck = 0.1 1/d; ktc2 = 1.0E8 1/mlReaction: Resistant_Stem_Cells_S_R => ; Cytotoxic_T_Cells_T_C, Rate Law: compartment*tck*Resistant_Stem_Cells_S_R*Cytotoxic_T_Cells_T_C/(ktc2+Cytotoxic_T_Cells_T_C)
myu_SR = 0.18 1/d; k2 = 10.0 1/mlReaction: Resistant_Stem_Cells_S_R => ; Interferon_gamma, Rate Law: compartment*myu_SR*Resistant_Stem_Cells_S_R*Interferon_gamma/(Interferon_gamma+k2)
beta_Tc = 1.0E-8 1/dReaction: => Interferon_gamma; Cytotoxic_T_Cells_T_C, Rate Law: compartment*beta_Tc*Cytotoxic_T_Cells_T_C
tck = 0.1 1/d; ktc3 = 1.0E9 1/mlReaction: Cancer_Cells_C => ; Cytotoxic_T_Cells_T_C, Rate Law: compartment*tck*Cancer_Cells_C*Cytotoxic_T_Cells_T_C/(ktc3+Cytotoxic_T_Cells_T_C)
delta_Ck3 = 8.664339 1/dReaction: Cytokine_IL2 =>, Rate Law: compartment*delta_Ck3*Cytokine_IL2
beta_M2 = 1.0E-15 1/dReaction: => Cytokine_IL10; M2_Tumor_Associated_Macrophages, Rate Law: compartment*beta_M2*M2_Tumor_Associated_Macrophages
k6 = 6.9937 1/ml; myu_C2 = 0.9 1/dReaction: Resistant_Cancer_Cells_C_R => ; Interferon_gamma, Rate Law: compartment*myu_C2*Resistant_Cancer_Cells_C_R*Interferon_gamma/(Interferon_gamma+k6)
beta_Th1CK3 = 1.0E-8 1/dReaction: => Cytokine_IL2; Type_I_T_helper_Cell_T_H1, Rate Law: compartment*beta_Th1CK3*Type_I_T_helper_Cell_T_H1
myu_TcS = 1.0E-10 1/d; lambda_Tc2 = 500000.0 1/mlReaction: Cytotoxic_T_Cells_T_C => ; Cancer_Stem_Cells_S, Resistant_Stem_Cells_S_R, Rate Law: compartment*myu_TcS*Cytotoxic_T_Cells_T_C*(Cancer_Stem_Cells_S+Resistant_Stem_Cells_S_R)/(Cytotoxic_T_Cells_T_C+lambda_Tc2)
delta_M1 = 1.02 1/dReaction: M1_Tumor_Associated_Macrophages =>, Rate Law: compartment*delta_M1*M1_Tumor_Associated_Macrophages
beta_Th2 = 1.0E-9 1/dReaction: => Cytokine_IL10; Type_II_T_helper_cells_T_H2, Rate Law: compartment*beta_Th2*Type_II_T_helper_cells_T_H2
tck = 0.1 1/d; ktc4 = 1.0E9 1/mlReaction: Resistant_Cancer_Cells_C_R => ; Cytotoxic_T_Cells_T_C, Rate Law: compartment*tck*Resistant_Cancer_Cells_C_R*Cytotoxic_T_Cells_T_C/(ktc4+Cytotoxic_T_Cells_T_C)
delta_Ck2 = 6.1212 1/dReaction: Interferon_gamma =>, Rate Law: compartment*delta_Ck2*Interferon_gamma
beta_Th1CK2 = 1.0E-7 1/dReaction: => Interferon_gamma; Type_I_T_helper_Cell_T_H1, Rate Law: compartment*beta_Th1CK2*Type_I_T_helper_Cell_T_H1
p_1 = 0.2; gamma_S = 0.15 1/dReaction: Resistant_Stem_Cells_S_R => Resistant_Stem_Cells_S_R + Resistant_Cancer_Cells_C_R, Rate Law: compartment*p_1*gamma_S*Resistant_Stem_Cells_S_R
p_2 = 0.05; gamma_S = 0.15 1/dReaction: Cancer_Stem_Cells_S => Cancer_Cells_C, Rate Law: compartment*p_2*gamma_S*Cancer_Stem_Cells_S
ktc1 = 1.0E9 1/ml; tck = 0.1 1/dReaction: Cancer_Stem_Cells_S => ; Cytotoxic_T_Cells_T_C, Rate Law: compartment*tck*Cancer_Stem_Cells_S*Cytotoxic_T_Cells_T_C/(ktc1+Cytotoxic_T_Cells_T_C)
m_C = 0.01; gamma_C = 0.1282 1/dReaction: Cancer_Cells_C => Resistant_Cancer_Cells_C_R, Rate Law: compartment*m_C*gamma_C*Cancer_Cells_C
myu_C1 = 0.75 1/d; k3 = 2.0531 1/mlReaction: => Cancer_Cells_C; Cytokine_IL10, Rate Law: compartment*myu_C1*Cancer_Cells_C*Cytokine_IL10/(Cytokine_IL10+k3)
k8 = 0.01 1/ml; myu_Th1Ck1 = 1.0E-9 1/dReaction: Type_I_T_helper_Cell_T_H1 => ; Cytokine_IL10, Rate Law: compartment*myu_Th1Ck1*Cytokine_IL10*Type_I_T_helper_Cell_T_H1/(Cytokine_IL10+k8)
delta_Tc = 5.2939 1/dReaction: Cytotoxic_T_Cells_T_C =>, Rate Law: compartment*delta_Tc*Cytotoxic_T_Cells_T_C
gamma_Th2 = 2.0 2/d; lambda_Th2 = 100000.0 1/mlReaction: => Type_II_T_helper_cells_T_H2; M2_Tumor_Associated_Macrophages, Rate Law: compartment*gamma_Th2*Type_II_T_helper_cells_T_H2*M2_Tumor_Associated_Macrophages/(lambda_Th2+Type_II_T_helper_cells_T_H2)
myu_M2Ck1 = 0.01 1/d; k10 = 0.01 1/mlReaction: => M2_Tumor_Associated_Macrophages; Cytokine_IL10, Rate Law: compartment*myu_M2Ck1*M2_Tumor_Associated_Macrophages*Cytokine_IL10/(Cytokine_IL10+k10)
lambda_M2 = 1000000.0 1/ml; gamma_M2 = 0.01 1/dReaction: => M2_Tumor_Associated_Macrophages; Cancer_Cells_C, Resistant_Cancer_Cells_C_R, Rate Law: compartment*gamma_M2*M2_Tumor_Associated_Macrophages*(Cancer_Cells_C+Resistant_Cancer_Cells_C_R)/(M2_Tumor_Associated_Macrophages+lambda_M2)
delta_Ck1 = 19.757 1/dReaction: Cytokine_IL10 =>, Rate Law: compartment*delta_Ck1*Cytokine_IL10
gamma_Tc = 1.0 1/d; lambda_Tc1 = 100000.0 1/mlReaction: => Cytotoxic_T_Cells_T_C; Cancer_Cells_C, Resistant_Cancer_Cells_C_R, Rate Law: compartment*gamma_Tc*Cytotoxic_T_Cells_T_C*(Cancer_Cells_C+Resistant_Cancer_Cells_C_R)/(Cytotoxic_T_Cells_T_C+lambda_Tc1)
gamma_Treg = 0.3 1/d; lambda_Treg2 = 1.0E7 1/mlReaction: => Regulatory_T_Cells_T_reg; M2_Tumor_Associated_Macrophages, Rate Law: compartment*gamma_Treg*Regulatory_T_Cells_T_reg*M2_Tumor_Associated_Macrophages/(Regulatory_T_Cells_T_reg+lambda_Treg2)
lambda_M1 = 1.0E8 1/ml; gamma_M1 = 0.7 1/dReaction: => M1_Tumor_Associated_Macrophages; Cancer_Cells_C, Resistant_Cancer_Cells_C_R, Rate Law: compartment*gamma_M1*M1_Tumor_Associated_Macrophages*(Cancer_Cells_C+Resistant_Cancer_Cells_C_R)/(M1_Tumor_Associated_Macrophages+lambda_M1)
delta_Th1 = 2.0 1/dReaction: Type_I_T_helper_Cell_T_H1 =>, Rate Law: compartment*delta_Th1*Type_I_T_helper_Cell_T_H1
lambda_Th1 = 100000.0 1/ml; gamma_Th1 = 2.0 1/dReaction: => Type_I_T_helper_Cell_T_H1; M1_Tumor_Associated_Macrophages, Rate Law: compartment*gamma_Th1*Type_I_T_helper_Cell_T_H1*M1_Tumor_Associated_Macrophages/(lambda_Th1+Type_I_T_helper_Cell_T_H1)
gamma_C = 0.1282 1/d; K_tumor = 2.0E10; r_2 = 1.0E-5Reaction: => Resistant_Cancer_Cells_C_R, Rate Law: compartment*gamma_C*Resistant_Cancer_Cells_C_R*ln(0.5*K_tumor/(Resistant_Cancer_Cells_C_R+r_2))
myu_M1Ck2 = 0.01 1/d; k7 = 0.2 1/mlReaction: => M1_Tumor_Associated_Macrophages; Interferon_gamma, Rate Law: compartment*myu_M1Ck2*M1_Tumor_Associated_Macrophages*Interferon_gamma/(Interferon_gamma+k7)
k11 = 0.001 1/ml; myu_TregCk1 = 1.0E-7 1/dReaction: => Regulatory_T_Cells_T_reg; Cytokine_IL10, Rate Law: compartment*myu_TregCk1*Cytokine_IL10*Regulatory_T_Cells_T_reg/(Regulatory_T_Cells_T_reg+k11)
myu_C1 = 0.75 1/d; k5 = 6.7979 1/mlReaction: => Resistant_Cancer_Cells_C_R; Cytokine_IL10, Rate Law: compartment*myu_C1*Resistant_Cancer_Cells_C_R*Cytokine_IL10/(Cytokine_IL10+k5)
beta_Treg = 1.0E-10 1/dReaction: => Cytokine_IL10; Regulatory_T_Cells_T_reg, Rate Law: compartment*beta_Treg*Regulatory_T_Cells_T_reg
myu_S = 0.17 1/d; k1 = 10.0 1/mlReaction: Cancer_Stem_Cells_S => ; Interferon_gamma, Rate Law: compartment*myu_S*Cancer_Stem_Cells_S*Interferon_gamma/(Interferon_gamma+k1)
delta_M2 = 0.05 1/dReaction: M2_Tumor_Associated_Macrophages =>, Rate Law: compartment*delta_M2*M2_Tumor_Associated_Macrophages
delta_C = 0.8055 1/dReaction: Cancer_Cells_C =>, Rate Law: compartment*delta_C*Cancer_Cells_C

States:

NameDescription
Interferon gamma[Interferon Gamma; Cytokine]
Cytokine IL10[Cytokine; Interleukin-10]
Regulatory T Cells T reg[Cytotoxic and Regulatory T-Cell Molecule]
Cancer Cells C[Malignant Cell]
Cancer Stem Cells S[Cancer Stem Cell]
Type I T helper Cell T H1[T-helper cell type 1; T-helper 1 cell differentiation]
Cytokine IL2[Cytokine; Interleukin-2]
M1 Tumor Associated Macrophages[Tumor-Associated Macrophage; M1 Macrophage]
Resistant Stem Cells S R[Drug Resistance Status; Cancer Stem Cell]
Resistant Cancer Cells C R[Malignant Cell; Drug Resistance Status]
M2 Tumor Associated Macrophages[M2 Macrophage; Tumor-Associated Macrophage]
Cytotoxic T Cells T C[Cytotoxic and Regulatory T-Cell Molecule]
Type II T helper cells T H2[T-helper 2 cell differentiation; T-helper cell type 2]
100000 SR100000*SR

Garcia2018basic - cancer and immune cell count basic model: BIOMD0000000742v0.0.1

The paper describes a basic model of immune-tumor cell interactions. Created by COPASI 4.25 (Build 207) This model is…

Details

Cancer immunotherapies rely on how interactions between cancer and immune system cells are constituted. The more essential to the emergence of the dynamical behavior of cancer growth these are, the more effectively they may be used as mechanisms for interventions. Mathematical modeling can help unearth such connections, and help explain how they shape the dynamics of cancer growth. Here, we explored whether there exist simple, consistent properties of cancer-immune system interaction (CISI) models that might be harnessed to devise effective immunotherapy approaches. We did this for a family of three related models of increasing complexity. To this end, we developed a base model of CISI, which captures some essential features of the more complex models built on it. We find that the base model and its derivates can reproduce biologically plausible behavior. This behavior is consistent with situations in which the suppressive effects exerted by cancer cells on immune cells dominate their proliferative effects. Under these circumstances, the model family may display a pattern of bistability, where two distinct, stable states (a cancer-free, and a full-grown cancer state) are possible, consistent with the notion of an immunological barrier. Increasing the effectiveness of immune-caused cancer cell killing may remove the basis for bistability, and abruptly tip the dynamics of the system into cancer-free state. In combination with the administration of immune effector cells, modifications in cancer cell killing may also be harnessed for immunotherapy without resolving the bistability. We use these ideas to test immunotherapeutic interventions in silico in a stochastic version of the base model. This bistability-reliant approach to cancer interventions might offer advantages over those that comprise gradual declines in cancer cell numbers. link: http://identifiers.org/doi/10.1101/498741

Parameters:

NameDescription
a = 0.514 1/ksReaction: => T, Rate Law: Tumor*a*T
k = 1.0E-4 1/ksReaction: T => ; E, Rate Law: Tumor*k*T*E
m = -1.0E-6 1/ksReaction: => E; T, Rate Law: Tumor*m*E*T
a = 0.514 1/ks; b = 1.02E-9 1Reaction: T =>, Rate Law: Tumor*a*b*T*T
d = 0.02 1/ksReaction: E =>, Rate Law: Tumor*d*E
s = 10.0 1/ksReaction: => E, Rate Law: Tumor*s

States:

NameDescription
T[neoplastic cell]
E[Effector Immune Cell; leukocyte]

Garde2020-Minimal model describing metabolic oscillations in Bacillus subtilis biofilms: BIOMD0000000932v0.0.1

Biofilms offer an excellent example of ecological interaction among bacteria. Temporal and spatial oscillations in biofi…

Details

Biofilms offer an excellent example of ecological interaction among bacteria. Temporal and spatial oscillations in biofilms are an emerging topic. In this paper, we describe the metabolic oscillations in Bacillus subtilis biofilms by applying the smallest theoretical chemical reaction system showing Hopf bifurcation proposed by Wilhelm and Heinrich in 1995. The system involves three differential equations and a single bilinear term. We specifically select parameters that are suitable for the biological scenario of biofilm oscillations. We perform computer simulations and a detailed analysis of the system including bifurcation analysis and quasi-steady-state approximation. We also discuss the feedback structure of the system and the correspondence of the simulations to biological observations. Our theoretical work suggests potential scenarios about the oscillatory behaviour of biofilms and also serves as an application of a previously described chemical oscillator to a biological system. link: http://identifiers.org/pubmed/32257302

Parameters:

NameDescription
k4 = 2.0Reaction: Gp => Gi, Rate Law: compartment*k4*Gp
k5 = 2.3Reaction: Gi => A, Rate Law: compartment*k5*Gi
b = 0.1Reaction: => B; A, Gp, Rate Law: compartment*b*A*Gp*B
k3 = 4.0Reaction: A =>, Rate Law: compartment*k3*A
GE = 30.0; k2 = 5.3; k1 = 0.3426Reaction: => Gp; A, Rate Law: compartment*(k1*GE*Gp-k2*A*Gp)

States:

NameDescription
B[biomass production]
A[ammonia]
Gi[CHEBI:32484; C25234]
Gp[CHEBI:32484; C25233]

Gardner1998 - Cell Cycle Goldbeter: BIOMD0000000008v0.0.1

Gardner1998 - Cell Cycle GoldbeterMathematical modeling of cell division cycle (CDC) dynamics. The SBML file has been…

Details

We demonstrate, by using mathematical modeling of cell division cycle (CDC) dynamics, a potential mechanism for precisely controlling the frequency of cell division and regulating the size of a dividing cell. Control of the cell cycle is achieved by artificially expressing a protein that reversibly binds and inactivates any one of the CDC proteins. In the simplest case, such as the checkpoint-free situation encountered in early amphibian embryos, the frequency of CDC oscillations can be increased or decreased by regulating the rate of synthesis, the binding rate, or the equilibrium constant of the binding protein. In a more complex model of cell division, where size-control checkpoints are included, we show that the same reversible binding reaction can alter the mean cell mass in a continuously dividing cell. Because this control scheme is general and requires only the expression of a single protein, it provides a practical means for tuning the characteristics of the cell cycle in vivo. link: http://identifiers.org/pubmed/9826676

Parameters:

NameDescription
k1=0.5; K5=0.02Reaction: C => ; X, Rate Law: C*k1*X*(C+K5)^-1
a2=0.05Reaction: Z => C + Y, Rate Law: a2*Z
K4=0.1; V4=0.1Reaction: X =>, Rate Law: V4*X*(K4+X)^-1
kd=0.02Reaction: C =>, Rate Law: C*kd
kd=0.02; alpha=0.1Reaction: Z => Y, Rate Law: alpha*kd*Z
a1=0.05Reaction: C + Y => Z, Rate Law: a1*C*Y
alpha=0.1; d1=0.05Reaction: Z => C, Rate Law: alpha*d1*Z
K3=0.2; V3 = NaNReaction: => X, Rate Law: V3*(1+-1*X)*(K3+-1*X+1)^-1
K1=0.1; V1 = NaNReaction: => M, Rate Law: (1+-1*M)*V1*(K1+-1*M+1)^-1
d1=0.05Reaction: Y =>, Rate Law: d1*Y
vi=0.1Reaction: => C, Rate Law: vi
K2=0.1; V2=0.25Reaction: M =>, Rate Law: M*V2*(K2+M)^-1
vs=0.2Reaction: => Y, Rate Law: vs

States:

NameDescription
Ycyclin inhibitor
Z[IPR006670]
X[peptidase activity]
C[IPR006670]
M[Cyclin-dependent kinase 1-A; Cyclin-dependent kinase 1-B]

Gardner2000 - genetic toggle switch in E.coli: BIOMD0000000507v0.0.1

Gardner2000 - genetic toggle switch in E.coliThe behaviour of the genetic toggle switch and the conditions for bistabili…

Details

It has been proposed' that gene-regulatory circuits with virtually any desired property can be constructed from networks of simple regulatory elements. These properties, which include multistability and oscillations, have been found in specialized gene circuits such as the bacteriophage lambda switch and the Cyanobacteria circadian oscillator. However, these behaviours have not been demonstrated in networks of non-specialized regulatory components. Here we present the construction of a genetic toggle switch-a synthetic, bistable gene-regulatory network-in Escherichia coli and provide a simple theory that predicts the conditions necessary for bistability. The toggle is constructed from any two repressible promoters arranged in a mutually inhibitory network. It is flipped between stable states using transient chemical or thermal induction and exhibits a nearly ideal switching threshold. As a practical device, the toggle switch forms a synthetic, addressable cellular memory unit and has implications for biotechnology, biocomputing and gene therapy. link: http://identifiers.org/pubmed/10659857

Parameters:

NameDescription
parameter_7 = 0.0; parameter_2 = 15.6; parameter_4 = 1.0Reaction: => species_2, Rate Law: compartment_1*parameter_2/(1+parameter_7^parameter_4)
k1=1.0Reaction: species_1 => ; species_1, Rate Law: compartment_1*k1*species_1
parameter_1 = 156.25; parameter_3 = 2.5Reaction: => species_1; species_2, species_2, Rate Law: compartment_1*parameter_1/(1+species_2^parameter_3)

States:

NameDescription
species 2[Tetracycline repressor protein class B from transposon Tn10]
species 1[Lactose operon repressor]

Garira2019 - A coupled multiscale model to guide malaria control and elimination: MODEL2003190008v0.0.1

This is a coupled multiscale mathematical model of malaria control and elimination containing four submodels: mosquito-t…

Details

In this paper, we share with the biomathematics community a new coupled multiscale model which has the potential to inform policy and guide malaria control and elimination. The formulation of this multiscale model is based on integrating four submodels which are: (i) a sub-model for the mosquito-to-human transmission of malaria parasite, (ii) a sub-model for the human-to-mosquito transmission of malaria parasite, (iii) a within-mosquito malaria parasite population dynamics sub-model and (iv) a within-human malaria parasite population dynamics sub-model. The integration of the four submodels is achieved by assuming that the transmission parameters of the sub-model for the mosquito-to-human transmission of malaria at the epidemiological scale are functions of the dependent variables of the within-mosquito sporozoite population dynamics while the transmission parameters of the sub-model for the human-to-mosquito transmission of malaria are functions of the dependent variables of the within-human gametocyte population dynamics. This establishes a unidirectionally coupled multiscale model where the within-human and within-mosquito submodels are unidirectionally coupled to the human-to-mosquito and mosquito-to-human submodels. A fast and slow time scale analysis is performed on this system. The result is a simple multiscale model which describes the mechanics of malaria transmission in terms of the major components of the complete malaria parasite life-cycle. This multiscale modelling approach may be found useful in guiding malaria control and elimination. link: http://identifiers.org/pubmed/31128139

Gebauer2016 - Genome-scale model of Caenorhabditis elegans metabolism (with bacteria): MODEL1704200001v0.0.1

Gebauer2016 - Genome-scale model of Caenorhabditis elegans metabolism (with bacteria)This model is one of the two versio…

Details

We present a genome-scale model of Caenorhabditis elegans metabolism along with the public database ElegCyc (http://elegcyc.bioinf.uni-jena.de:1100), which represents a reference for metabolic pathways in the worm and allows for the visualization as well as analysis of omics datasets. Our model reflects the metabolic peculiarities of C. elegans that make it distinct from other higher eukaryotes and mammals, including mice and humans. We experimentally verify one of these peculiarities by showing that the lifespan-extending effect of L-tryptophan supplementation is dose dependent (hormetic). Finally, we show the utility of our model for analyzing omics datasets through predicting changes in amino acid concentrations after genetic perturbations and analyzing metabolic changes during normal aging as well as during two distinct, reactive oxygen species (ROS)-related lifespan-extending treatments. Our analyses reveal a notable similarity in metabolic adaptation between distinct lifespan-extending interventions and point to key pathways affecting lifespan in nematodes. link: http://identifiers.org/pubmed/27211858

Gebauer2016 - Genome-scale model of Caenorhabditis elegans metabolism (without bacteria): MODEL1704200000v0.0.1

Gebauer2016 - Genome-scale model of Caenorhabditis elegans metabolism (without bacteria)This model is one of the two ver…

Details

We present a genome-scale model of Caenorhabditis elegans metabolism along with the public database ElegCyc (http://elegcyc.bioinf.uni-jena.de:1100), which represents a reference for metabolic pathways in the worm and allows for the visualization as well as analysis of omics datasets. Our model reflects the metabolic peculiarities of C. elegans that make it distinct from other higher eukaryotes and mammals, including mice and humans. We experimentally verify one of these peculiarities by showing that the lifespan-extending effect of L-tryptophan supplementation is dose dependent (hormetic). Finally, we show the utility of our model for analyzing omics datasets through predicting changes in amino acid concentrations after genetic perturbations and analyzing metabolic changes during normal aging as well as during two distinct, reactive oxygen species (ROS)-related lifespan-extending treatments. Our analyses reveal a notable similarity in metabolic adaptation between distinct lifespan-extending interventions and point to key pathways affecting lifespan in nematodes. link: http://identifiers.org/pubmed/27211858

Geier2011 - Integrin activation: MODEL1208280001v0.0.1

Geier2011 - Integrin activationRule based model that integrates the available data to test the biololical hypotheses reg…

Details

Integrin signaling regulates cell migration and plays a pivotal role in developmental processes and cancer metastasis. Integrin signaling has been studied extensively and much data is available on pathway components and interactions. Yet the data is fragmented and an integrated model is missing. We use a rule-based modeling approach to integrate available data and test biological hypotheses regarding the role of talin, Dok1 and PIPKI in integrin activation. The detailed biochemical characterization of integrin signaling provides us with measured values for most of the kinetics parameters. However, measurements are not fully accurate and the cellular concentrations of signaling proteins are largely unknown and expected to vary substantially across different cellular conditions. By sampling model behaviors over the physiologically realistic parameter range we find that the model exhibits only two different qualitative behaviors and these depend mainly on the relative protein concentrations, which offers a powerful point of control to the cell. Our study highlights the necessity to characterize model behavior not for a single parameter optimum, but to identify parameter sets that characterize different signaling modes. link: http://identifiers.org/pubmed/22110576

Geißert2020 - Yersinia enterocolitica co-infection in mice: MODEL2002070001v0.0.1

This model simulates the colonization of the mouse gut with different strains of Yersinia enterocolitica. Thereby it tak…

Details

The complex interplay of a pathogen with the host immune response and the endogenous microbiome determines the course and outcome of gastrointestinal infection (GI). Expansion of a pathogen within the gastrointestinal tract implies an increased risk to develop systemic infection. Through computational modeling, we aimed to calculate bacterial population dynamics in GI in order to predict infection course and outcome. For the implementation and parameterization of the model, oral mouse infection experiments with Yersinia enterocolitica were used. Our model takes into account pathogen specific characteristics, such as virulence, as well as host properties, such as microbial colonization resistance or immune responses. We were able to confirm the model calculations in these scenarios by experimental mouse infections and show that it is possible to computationally predict the infection course. Far future clinical application of computational modeling of infections may pave the way for personalized treatment and prevention strategies of GI. link: http://identifiers.org/doi/10.1101/2020.08.11.244202

Gerard2009 - An Integrated Mammalian Cell Cycle Model: BIOMD0000000730v0.0.1

We propose an integrated computational model for the network of cyclin-dependent kinases (Cdks) that controls the dynami…

Details

We propose an integrated computational model for the network of cyclin-dependent kinases (Cdks) that controls the dynamics of the mammalian cell cycle. The model contains four Cdk modules regulated by reversible phosphorylation, Cdk inhibitors, and protein synthesis or degradation. Growth factors (GFs) trigger the transition from a quiescent, stable steady state to self-sustained oscillations in the Cdk network. These oscillations correspond to the repetitive, transient activation of cyclin D/Cdk4-6 in G(1), cyclin E/Cdk2 at the G(1)/S transition, cyclin A/Cdk2 in S and at the S/G(2) transition, and cyclin B/Cdk1 at the G(2)/M transition. The model accounts for the following major properties of the mammalian cell cycle: (i) repetitive cell cycling in the presence of suprathreshold amounts of GF; (ii) control of cell-cycle progression by the balance between antagonistic effects of the tumor suppressor retinoblastoma protein (pRB) and the transcription factor E2F; and (iii) existence of a restriction point in G(1), beyond which completion of the cell cycle becomes independent of GF. The model also accounts for endoreplication. Incorporating the DNA replication checkpoint mediated by kinases ATR and Chk1 slows down the dynamics of the cell cycle without altering its oscillatory nature and leads to better separation of the S and M phases. The model for the mammalian cell cycle shows how the regulatory structure of the Cdk network results in its temporal self-organization, leading to the repetitive, sequential activation of the four Cdk modules that brings about the orderly progression along cell-cycle phases. link: http://identifiers.org/pubmed/20007375

Parameters:

NameDescription
kpc3 = 0.025; eps = 17.0Reaction: pRBp + E2F => pRBc2; pRBp, E2F, Rate Law: cell*kpc3*pRBp*E2F*eps
kdpb = 0.1; eps = 17.0Reaction: Pb => ; Pb, Rate Law: cell*kdpb*Pb*eps
eps = 17.0; vs1p27 = 0.8Reaction: => p27, Rate Law: cell*vs1p27*eps
V1 = 2.2; K1 = 0.1; eps = 17.0Reaction: pRB => pRBp; pRB, Md, Mdp27, Rate Law: cell*V1*pRB/(K1+pRB)*(Md+Mdp27)*eps
Cdk4_tot = 1.5; eps = 17.0; kcom1 = 0.175Reaction: Cd => Mdi; Mdi, Md, Mdp27, Rate Law: cell*kcom1*Cd*(Cdk4_tot-(Mdi+Md+Mdp27))*eps
eps = 17.0; K4b = 0.1; Vm4b = 0.7Reaction: Cdc20a => Cdc20i; Cdc20a, Rate Law: cell*Vm4b*Cdc20a/(K4b+Cdc20a)*eps
K2b = 0.1; ib3 = 0.5; eps = 17.0; Vm2b = 2.1Reaction: Mb => Mbi; Wee1, Mb, Rate Law: cell*Vm2b*(Wee1+ib3)*Mb/(K2b+Mb)*eps
V4 = 2.0; eps = 17.0; K4 = 0.1Reaction: pRBpp => pRBp; pRBpp, Rate Law: cell*V4*pRBpp/(K4+pRBpp)*eps
kdecom3 = 0.1; eps = 17.0Reaction: Mai => Ca; Mai, Rate Law: cell*kdecom3*Mai*eps
eps = 17.0; vsprb = 0.8Reaction: => pRB, Rate Law: cell*vsprb*eps
eps = 17.0; kpc2 = 0.5Reaction: pRBc1 => pRB + E2F; pRBc1, Rate Law: cell*kpc2*pRBc1*eps
kdpe = 0.075; eps = 17.0Reaction: Pe => ; Pe, Rate Law: cell*kdpe*Pe*eps
Vm1a = 2.0; K1a = 0.1; eps = 17.0Reaction: Mai => Ma; Mai, Pa, Rate Law: cell*Vm1a*Mai/(K1a+Mai)*Pa*eps
Ki13 = 0.1; eps = 17.0; Ki14 = 2.0; vs2p27 = 0.1Reaction: => p27; E2F, pRB, pRBp, Rate Law: cell*vs2p27*E2F*Ki13/(Ki13+pRB)*Ki14/(Ki14+pRBp)*eps
GF = 1.0; vsap1 = 1.0; Kagf = 0.1; eps = 17.0Reaction: => AP1, Rate Law: cell*vsap1*GF/(Kagf+GF)*eps
K2d = 0.1; Vm2d = 0.2; eps = 17.0Reaction: Md => Mdi; Md, Rate Law: cell*Vm2d*Md/(K2d+Md)*eps
kc7 = 0.12; eps = 17.0Reaction: Mb + p27 => Mbp27; Mb, p27, Rate Law: cell*kc7*Mb*p27*eps
kc6 = 0.125; eps = 17.0Reaction: Map27 => Ma + p27; Map27, Rate Law: cell*kc6*Map27*eps
kdwee1 = 0.1; eps = 17.0Reaction: Wee1 => ; Wee1, Rate Law: cell*kdwee1*Wee1*eps
kdap1 = 0.15; eps = 17.0Reaction: AP1 => ; AP1, Rate Law: cell*kdap1*AP1*eps
K2 = 0.1; eps = 17.0; V2 = 2.0Reaction: pRBp => pRB; pRBp, Rate Law: cell*V2*pRBp/(K2+pRBp)*eps
kdprbp = 0.06; eps = 17.0Reaction: pRBp => ; pRBp, Rate Law: cell*kdprbp*pRBp*eps
eps = 17.0; Kdd = 0.1; Vdd = 5.0Reaction: Cd => ; Cd, Rate Law: cell*Vdd*Cd/(Kdd+Cd)*eps
kc2 = 0.05; eps = 17.0Reaction: Mdp27 => Md + p27; Mdp27, Rate Law: cell*kc2*Mdp27*eps
K8b = 0.1; Vm8b = 1.0; eps = 17.0Reaction: Wee1p => Wee1; Wee1p, Rate Law: cell*Vm8b*Wee1p/(K8b+Wee1p)*eps
Kdp27p = 0.1; eps = 17.0; Kdp27skp2 = 0.1; Vdp27p = 5.0Reaction: p27p => ; Skp2, p27p, Rate Law: cell*Vdp27p*Skp2/(Kdp27skp2+Skp2)*p27p/(Kdp27p+p27p)*eps
Cdk2_tot = 2.0; eps = 17.0; kcom2 = 0.2Reaction: Ce => Mei; Mei, Me, Mep27, Mai, Ma, Map27, Rate Law: cell*kcom2*Ce*(Cdk2_tot-(Mei+Me+Mep27+Mai+Ma+Map27))*eps
Kde = 0.1; Vde = 3.0; Kdceskp2 = 2.0; eps = 17.0Reaction: Ce => ; Skp2, Ce, Rate Law: cell*Vde*Skp2/(Kdceskp2+Skp2)*Ce/(Kde+Ce)*eps
kc1 = 0.15; eps = 17.0Reaction: Md + p27 => Mdp27; Md, p27, Rate Law: cell*kc1*Md*p27*eps
kc5 = 0.15; eps = 17.0Reaction: Ma + p27 => Map27; Ma, p27, Rate Law: cell*kc5*Ma*p27*eps
eps = 17.0; kc3 = 0.2Reaction: Me + p27 => Mep27; Me, p27, Rate Law: cell*kc3*Me*p27*eps
kpc1 = 0.05; eps = 17.0Reaction: pRB + E2F => pRBc1; pRB, E2F, Rate Law: cell*kpc1*pRB*E2F*eps
Ki8 = 2.0; eps = 17.0; Ki7 = 0.1; kcd2 = 0.005Reaction: => Cd; E2F, pRB, pRBp, Rate Law: cell*kcd2*E2F*Ki7/(Ki7+pRB)*Ki8/(Ki8+pRBp)*eps
K1cdh1 = 0.01; eps = 17.0; V1cdh1 = 1.25Reaction: Cdh1i => Cdh1a; Cdh1i, Rate Law: cell*V1cdh1*Cdh1i/(K1cdh1+Cdh1i)*eps
eps = 17.0; Ki9 = 0.1; Ki10 = 2.0; kce = 0.25Reaction: => Ce; E2F, pRB, pRBp, Rate Law: cell*kce*E2F*Ki9/(Ki9+pRB)*Ki10/(Ki10+pRBp)*eps
eps = 17.0; K3b = 0.1; Vm3b = 8.0Reaction: Cdc20i => Cdc20a; Cdc20i, Mb, Rate Law: cell*Vm3b*Cdc20i/(K3b+Cdc20i)*Mb*eps
kddp27p = 0.01; eps = 17.0Reaction: p27p => ; p27p, Rate Law: cell*kddp27p*p27p*eps
kdecom4 = 0.1; eps = 17.0Reaction: Mbi => Cb; Mbi, Rate Law: cell*kdecom4*Mbi*eps
eps = 17.0; kdecom2 = 0.1Reaction: Mei => Ce; Mei, Rate Law: cell*kdecom2*Mei*eps
eps = 17.0; kcd1 = 0.4Reaction: => Cd; AP1, Rate Law: cell*kcd1*AP1*eps
V2cdh1 = 8.0; K2cdh1 = 0.01; eps = 17.0Reaction: Cdh1a => Cdh1i; Cdh1a, Ma, Mb, Rate Law: cell*V2cdh1*Cdh1a/(K2cdh1+Cdh1a)*(Ma+Mb)*eps
kdda = 0.005; eps = 17.0Reaction: Ca => ; Ca, Rate Law: cell*kdda*Ca*eps
Vm1e = 2.0; eps = 17.0; K1e = 0.1Reaction: Mei => Me; Mei, Pe, Rate Law: cell*Vm1e*Mei/(K1e+Mei)*Pe*eps
ATR_tot = 0.5; eps = 17.0; kaatr = 0.022Reaction: Primer => ATR; ATR, Primer, Rate Law: cell*kaatr*(ATR_tot-ATR)*Primer*eps
kdde = 0.005; eps = 17.0Reaction: Ce => ; Ce, Rate Law: cell*kdde*Ce*eps
K2p27 = 0.5; eps = 17.0; V2p27 = 0.1Reaction: p27p => p27; p27p, Rate Law: cell*V2p27*p27p/(K2p27+p27p)*eps
eps = 17.0; Vda = 2.5; Kda = 1.1; Kacdc20 = 2.0Reaction: Ca => ; Ca, Cdc20a, Rate Law: cell*Vda*Ca/(Kda+Ca)*Cdc20a/(Kacdc20+Cdc20a)*eps
K3 = 0.1; V3 = 1.0; eps = 17.0Reaction: pRBp => pRBpp; pRBp, Me, Rate Law: cell*V3*pRBp/(K3+pRBp)*Me*eps
eps = 17.0; kdcdc20a = 0.05Reaction: Cdc20a => ; Cdc20a, Rate Law: cell*kdcdc20a*Cdc20a*eps
eps = 17.0; kpc4 = 0.5Reaction: pRBc2 => pRBp + E2F; pRBc2, Rate Law: cell*kpc4*pRBc2*eps
ib1 = 0.5; eps = 17.0; Vm2e = 1.4; K2e = 0.1Reaction: Me => Mei; Wee1, Me, Rate Law: cell*Vm2e*(Wee1+ib1)*Me/(K2e+Me)*eps
Vm5e = 5.0; eps = 17.0; ae = 0.25; K5e = 0.1Reaction: Pei => Pe; Me, Pei, Rate Law: cell*Vm5e*(Me+ae)*Pei/(K5e+Pei)*eps
kdecom1 = 0.1; eps = 17.0Reaction: Mdi => Cd; Mdi, Rate Law: cell*kdecom1*Mdi*eps
kdpei = 0.15; eps = 17.0Reaction: Pei => ; Pei, Rate Law: cell*kdpei*Pei*eps
V1cdc45 = 0.8; Cdc45_tot = 0.5; eps = 17.0; K1cdc45 = 0.02Reaction: => Cdc45; Me, Rate Law: cell*V1cdc45*Me*(Cdc45_tot-Cdc45)/((K1cdc45+Cdc45_tot)-Cdc45)*eps
ksprim = 0.05; eps = 17.0Reaction: => Primer; Pol, Rate Law: cell*ksprim*Pol*eps
kdprb = 0.01; eps = 17.0Reaction: pRB => ; pRBp, Rate Law: cell*kdprb*pRBp*eps
kc4 = 0.1; eps = 17.0Reaction: Mep27 => Me + p27; Mep27, Rate Law: cell*kc4*Mep27*eps
K7b = 0.1; eps = 17.0; ib = 0.75; Vm7b = 1.2Reaction: Wee1 => Wee1p; Mb, Wee1, Rate Law: cell*Vm7b*(Mb+ib)*Wee1/(K7b+Wee1)*eps
V6e = 0.8; xe1 = 1.0; eps = 17.0; xe2 = 1.0; K6e = 0.1Reaction: Pe => Pei; Chk1, Pe, Rate Law: cell*V6e*(xe1+xe2*Chk1)*Pe/(K6e+Pe)*eps
K2a = 0.1; Vm2a = 1.85; eps = 17.0; ib2 = 0.5Reaction: Ma => Mai; Wee1, Ma, Rate Law: cell*Vm2a*(Wee1+ib2)*Ma/(K2a+Ma)*eps
kddd = 0.005; eps = 17.0Reaction: Cd => ; Cd, Rate Law: cell*kddd*Cd*eps
kdcdc20i = 0.14; eps = 17.0Reaction: Cdc20i => ; Cdc20i, Rate Law: cell*kdcdc20i*Cdc20i*eps
kdatr = 0.15; eps = 17.0Reaction: ATR => Primer; ATR, Rate Law: cell*kdatr*ATR*eps
K1p27 = 0.5; eps = 17.0; V1p27 = 100.0Reaction: p27 => p27p; p27, Me, Rate Law: cell*V1p27*p27/(K1p27+p27)*Me*eps
K1d = 0.1; Vm1d = 1.0; eps = 17.0Reaction: Mdi => Md; Mdi, Rate Law: cell*Vm1d*Mdi/(K1d+Mdi)*eps
kc8 = 0.2; eps = 17.0Reaction: Mbp27 => Mb + p27; Mbp27, Rate Law: cell*kc8*Mbp27*eps
kdprim = 0.15; eps = 17.0Reaction: Primer => ; Primer, Rate Law: cell*kdprim*Primer*eps
eps = 17.0; kddp27 = 0.06Reaction: p27 => ; p27, Rate Law: cell*kddp27*p27*eps
kca = 0.0375; Ki11 = 0.1; eps = 17.0; Ki12 = 2.0Reaction: => Ca; E2F, pRB, pRBp, Rate Law: cell*kca*E2F*Ki11/(Ki11+pRB)*Ki12/(Ki12+pRBp)*eps
Cdk2_tot = 2.0; kcom3 = 0.2; eps = 17.0Reaction: Ca => Mai; Mei, Me, Mep27, Mai, Ma, Map27, Rate Law: cell*kcom3*Ca*(Cdk2_tot-(Mei+Me+Mep27+Mai+Ma+Map27))*eps

States:

NameDescription
Ce[G1/S-specific cyclin-E1; G1/S-specific cyclin-E2]
pRBc2[Retinoblastoma-associated protein; Transcription factor E2F1]
Pe[M-phase inducer phosphatase 2; M-phase inducer phosphatase 3; M-phase inducer phosphatase 1; Phosphorylated Peptide]
Pei[M-phase inducer phosphatase 3; M-phase inducer phosphatase 2; M-phase inducer phosphatase 1]
Mbi[G2/mitotic-specific cyclin-B1; Cyclin-dependent kinase 1]
p27p[Cyclin-dependent kinase inhibitor 1B; Phosphorylated Peptide]
Cdc20a[Cell division cycle protein 20 homolog]
Mbp27[G2/mitotic-specific cyclin-B1; Cyclin-dependent kinase 1; Cyclin-dependent kinase inhibitor 1B]
Map27[Cyclin-A2; Cyclin-dependent kinase 2; Cyclin-dependent kinase inhibitor 1B]
PrimerPrimer
pRB[Retinoblastoma-associated protein]
AP1[1,4-beta-D-Mannooligosaccharide]
Wee1[Wee1-like protein kinase]
Mdi[G1/S-specific cyclin-D2; G1/S-specific cyclin-D3; Cyclin-dependent kinase 6; G1/S-specific cyclin-D1; Cyclin-dependent kinase 4]
Mdp27[Cyclin-dependent kinase 4; Dehydrin Rab25; G1/S-specific cyclin-D1; G1/S-specific cyclin-D2; Cyclin-dependent kinase 6; Cyclin-dependent kinase inhibitor 1B]
ATR[Serine/threonine-protein kinase ATR]
Ma[Cyclin-A2; Cyclin-dependent kinase 2]
pRBc1[Retinoblastoma-associated protein; Transcription factor E2F1]
Md[Cyclin-dependent kinase 6; G1/S-specific cyclin-D1; G1/S-specific cyclin-D2; Cyclin-dependent kinase 4; G1/S-specific cyclin-D3]
Cdc45[Cell division control protein 45 homolog]
pRBpp[Retinoblastoma-associated protein; Phosphorylated Peptide]
Wee1p[Wee1-like protein kinase; Phosphorylated Peptide]
Mei[G1/S-specific cyclin-E1; G1/S-specific cyclin-E2; Cyclin-dependent kinase 2]
p27[Cyclin-dependent kinase inhibitor 1B]
Cdh1i[Cadherin-1]
Mai[Cyclin-dependent kinase 2; Cyclin-A2]
Ca[Cyclin-A2]
pRBp[Retinoblastoma-associated protein; Phosphorylated Peptide]
Cd[G1/S-specific cyclin-D2; G1/S-specific cyclin-D3; G1/S-specific cyclin-D1]
Cdc20i[Cell division cycle protein 20 homolog]
Pb[M-phase inducer phosphatase 3; M-phase inducer phosphatase 1; M-phase inducer phosphatase 2; Phosphorylated Peptide]

Gerard2010 - Progression of mammalian cell cycle by successive activation of various cyclin cdk complexes: BIOMD0000000941v0.0.1

We previously proposed a detailed, 39-variable model for the network of cyclin-dependent kinases (Cdks) that controls pr…

Details

We previously proposed a detailed, 39-variable model for the network of cyclin-dependent kinases (Cdks) that controls progression along the successive phases of the mammalian cell cycle. Here, we propose a skeleton, 5-variable model for the Cdk network that can be seen as the backbone of the more detailed model for the mammalian cell cycle. In the presence of sufficient amounts of growth factor, the skeleton model also passes from a stable steady state to sustained oscillations of the various cyclin/Cdk complexes. This transition corresponds to the switch from quiescence to cell proliferation. Sequential activation of the cyclin/Cdk complexes allows the ordered progression along the G1, S, G2 and M phases of the cell cycle. The 5-variable model can also account for the existence of a restriction point in G1, and for endoreplication. Like the detailed model, it contains multiple oscillatory circuits and can display complex oscillatory behaviour such as quasi-periodic oscillations and chaos. We compare the dynamical properties of the skeleton model with those of the more detailed model for the mammalian cell cycle. link: http://identifiers.org/pubmed/22419972

Parameters:

NameDescription
vsa = 0.175Reaction: => cyclin_A_Cdk2; transcription_factor_E2F_active, Rate Law: nuclear*vsa*transcription_factor_E2F_active
Vdb = 0.28; Kdb = 0.005Reaction: cyclin_B_Cdk1 => ; Cdc20_active, Rate Law: nuclear*Vdb*Cdc20_active*cyclin_B_Cdk1/(Kdb+cyclin_B_Cdk1)
Vda = 0.245; Kda = 0.1Reaction: cyclin_A_Cdk2 => ; Cdc20_active, Rate Law: nuclear*Vda*Cdc20_active*cyclin_A_Cdk2/(Kda+cyclin_A_Cdk2)
K2cdc20 = 1.0; V2cdc20 = 0.35Reaction: Cdc20_active =>, Rate Law: nuclear*V2cdc20*Cdc20_active/(K2cdc20+Cdc20_active)
vse = 0.21Reaction: => cyclin_E_Cdk2; transcription_factor_E2F_active, Rate Law: nuclear*vse*transcription_factor_E2F_active
K1cdc20 = 1.0; V1cdc20 = 0.21Reaction: => Cdc20_active; cyclin_B_Cdk1, Cdc20_total, Rate Law: nuclear*V1cdc20*cyclin_B_Cdk1*(Cdc20_total-Cdc20_active)/(K1cdc20+(Cdc20_total-Cdc20_active))
Kdd = 0.1; Vdd = 0.245Reaction: cyclin_D_Cdk4_6 => ; cyclin_D_Cdk4_6, Rate Law: nuclear*Vdd*cyclin_D_Cdk4_6/(Kdd+cyclin_D_Cdk4_6)
V2e2f = 0.7; K2e2f = 0.01Reaction: transcription_factor_E2F_active => ; cyclin_A_Cdk2, Rate Law: nuclear*V2e2f*transcription_factor_E2F_active/(K2e2f+transcription_factor_E2F_active)*cyclin_A_Cdk2
GF = 1.0; Kgf = 0.1; vsd = 0.175Reaction: => cyclin_D_Cdk4_6, Rate Law: nuclear*vsd*GF/(Kgf+GF)
K1e2f = 0.01; V1e2f = 0.805Reaction: => transcription_factor_E2F_active; E2F_total, cyclin_D_Cdk4_6, cyclin_E_Cdk2, Rate Law: nuclear*V1e2f*(E2F_total-transcription_factor_E2F_active)/((K1e2f+E2F_total)-transcription_factor_E2F_active)*(cyclin_D_Cdk4_6+cyclin_E_Cdk2)
Kde = 0.1; Vde = 0.35Reaction: cyclin_E_Cdk2 => ; cyclin_A_Cdk2, Rate Law: nuclear*Vde*cyclin_A_Cdk2*cyclin_E_Cdk2/(Kde+cyclin_E_Cdk2)
vsb = 0.21Reaction: => cyclin_B_Cdk1; cyclin_A_Cdk2, Rate Law: nuclear*vsb*cyclin_A_Cdk2

States:

NameDescription
Cdc20 active[Cell division cycle protein 20 homolog; active; phosphorylated]
cyclin D Cdk4 6[G1/S-specific cyclin-D1; Cyclin-dependent kinase 4; protein-containing complex]
transcription factor E2F active[Transcription factor E2F1; active]
cyclin B Cdk1[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin-B1; protein-containing complex]
cyclin E Cdk2[G1/S-specific cyclin-E1; Cyclin-dependent kinase 2; protein-containing complex]
cyclin A Cdk2[Cyclin-dependent kinase 2; Cyclin-A2; protein-containing complex]

Gerard2013 - Model 3 - Embryonic-type eukaryotic Cell Cycle regulation based on negative feedback between Cdk/cyclin and APC and competitive inhibition between Cdk/cyclin and securin for polyubiquitylation_1: BIOMD0000000938v0.0.1

The eukaryotic cell cycle is characterized by alternating oscillations in the activities of cyclin-dependent kinase (Cdk…

Details

The eukaryotic cell cycle is characterized by alternating oscillations in the activities of cyclin-dependent kinase (Cdk) and the anaphase-promoting complex (APC). Successful completion of the cell cycle is dependent on the precise, temporally ordered appearance of these activities. A modest level of Cdk activity is sufficient to initiate DNA replication, but mitosis and APC activation require an elevated Cdk activity. In present-day eukaryotes, this temporal order is provided by a complex network of regulatory proteins that control both Cdk and APC activities via sharp thresholds, bistability, and time delays. Using simple computational models, we show here that these dynamical features of cell-cycle organization could emerge in a control system driven by a single Cdk/cyclin complex and APC wired in a negative-feedback loop. We show that ordered phosphorylation of cellular proteins could be explained by multisite phosphorylation/dephosphorylation and competition of substrates for interconverting kinase (Cdk) and phosphatase. In addition, the competition of APC substrates for ubiquitylation can create and maintain sustained oscillations in cyclin levels. We propose a sequence of models that gets closer and closer to a realistic model of cell-cycle control in yeast. Since these models lack the elaborate control mechanisms characteristic of modern eukaryotes, they suggest that bistability and time delay may have characterized eukaryotic cell divisions before the current cell-cycle control network evolved in all its complexity. link: http://identifiers.org/pubmed/23528096

Parameters:

NameDescription
k_d1cdk = 0.01Reaction: Cdk =>, Rate Law: nuclear*k_d1cdk*Cdk
K_dsec = 0.001; K_dcdk = 0.01; k_dsec = 0.4Reaction: Securin => ; Anaphase_promoting_complex_Phosphorylated, Cdk, Rate Law: nuclear*k_dsec*Anaphase_promoting_complex_Phosphorylated*Securin/(K_dsec*(1+Cdk/K_dcdk)+Securin)
V_ssec = 0.1Reaction: => Securin, Rate Law: nuclear*V_ssec
V_scdk = 0.06Reaction: => Cdk, Rate Law: nuclear*V_scdk
K_dsec = 0.001; K_dcdk = 0.01; k_dcdk = 0.35Reaction: Cdk => ; Anaphase_promoting_complex_Phosphorylated, Securin, Rate Law: nuclear*k_dcdk*Anaphase_promoting_complex_Phosphorylated*Cdk/(K_dcdk*(1+Securin/K_dsec)+Cdk)
K_1APC = 0.01; V_1APC = 0.15Reaction: Anaphase_promoting_complex_Phosphorylated =>, Rate Law: nuclear*V_1APC*Anaphase_promoting_complex_Phosphorylated/(K_1APC+Anaphase_promoting_complex_Phosphorylated)
k_d1sec = 0.01Reaction: Securin =>, Rate Law: nuclear*k_d1sec*Securin
k_2APC = 0.3; K_2APC = 0.01Reaction: => Anaphase_promoting_complex_Phosphorylated; Cdk, Anaphase_promoting_complex, Rate Law: nuclear*k_2APC*Cdk*Anaphase_promoting_complex/(K_2APC+Anaphase_promoting_complex)

States:

NameDescription
Anaphase promoting complex[anaphase-promoting complex]
Securin[Securin]
Anaphase promoting complex Phosphorylated[anaphase-promoting complex; phosphorylated]
Cdk[Cyclin-dependent kinase 1]

Gerlin2020 - Genome scale metabolic model of bacterial growth: MODEL2003100001v0.0.1

High proliferation rate and robustness are vital characteristics of bacterial pathogens to successfully colonize their h…

Details

High proliferation rate and robustness are vital characteristics of bacterial pathogens that successfully colonize their hosts. The observation of drastically slow growth in some pathogens is thus paradoxical and remains unexplained. In this study, we sought to understand the slow (fastidious) growth of the plant pathogen Xylella fastidiosa. Using genome-scale metabolic network reconstruction, modeling, and experimental validation, we explored its metabolic capabilities. Despite genome reduction and slow growth, the pathogen’s metabolic network is complete but strikingly minimalist and lacking in robustness. Most alternative reactions were missing, especially those favoring fast growth, and were replaced by less efficient paths. We also found that the production of some virulence factors imposes a heavy burden on growth. Interestingly, some specific determinants of fastidious growth were also found in other slow-growing pathogens, enriching the view that these metabolic peculiarities are a pathogenicity strategy to remain at a low population level. link: http://identifiers.org/doi/10.1128/mSystems.00698-19

Gevertz2018 - cancer treatment with oncolytic viruses and dendritic cell injections minimal model: BIOMD0000000817v0.0.1

The model is based on 'Developing a Minimally Structured Mathematical Model of Cancer Treatment with Oncolytic Viruses a…

Details

Mathematical models of biological systems must strike a balance between being sufficiently complex to capture important biological features, while being simple enough that they remain tractable through analysis or simulation. In this work, we rigorously explore how to balance these competing interests when modeling murine melanoma treatment with oncolytic viruses and dendritic cell injections. Previously, we developed a system of six ordinary differential equations containing fourteen parameters that well describes experimental data on the efficacy of these treatments. Here, we explore whether this previously developed model is the minimal model needed to accurately describe the data. Using a variety of techniques, including sensitivity analyses and a parameter sloppiness analysis, we find that our model can be reduced by one variable and three parameters and still give excellent fits to the data. We also argue that our model is not too simple to capture the dynamics of the data, and that the original and minimal models make similar predictions about the efficacy and robustness of protocols not considered in experiments. Reducing the model to its minimal form allows us to increase the tractability of the system in the face of parametric uncertainty. link: http://identifiers.org/pubmed/30510594

Parameters:

NameDescription
C_T = 1.428064Reaction: => Tumor_targeting_T_cells_T; Infected_Cancer_Cell_I, Rate Law: compartment*C_T*Infected_Cancer_Cell_I
delta_I = 1.0Reaction: Infected_Cancer_Cell_I =>, Rate Law: compartment*delta_I*Infected_Cancer_Cell_I
delta_V = 2.3Reaction: Oncolytic_Adenovirus_V =>, Rate Law: compartment*delta_V*Oncolytic_Adenovirus_V
chi_D = 4.901894Reaction: => Tumor_targeting_T_cells_T; Dendritic_Cells_D, Rate Law: compartment*chi_D*Dendritic_Cells_D
U_V = 0.0Reaction: => Oncolytic_Adenovirus_V, Rate Law: compartment*U_V
r = 0.3198Reaction: => Uninfected_Tumor_Cell_U, Rate Law: compartment*r*Uninfected_Tumor_Cell_U
c_kill = 0.623397; k0 = 2.0Reaction: Uninfected_Tumor_Cell_U => ; Infected_Cancer_Cell_I, Tumor_targeting_T_cells_T, Total_cells_N, Rate Law: compartment*(k0+c_kill*Infected_Cancer_Cell_I)*Uninfected_Tumor_Cell_U*Tumor_targeting_T_cells_T/Total_cells_N
delta_T = 0.35Reaction: Tumor_targeting_T_cells_T =>, Rate Law: compartment*delta_T*Tumor_targeting_T_cells_T
U_D = 0.0Reaction: => Dendritic_Cells_D, Rate Law: compartment*U_D
beta = 1.008538Reaction: Uninfected_Tumor_Cell_U => Infected_Cancer_Cell_I; Oncolytic_Adenovirus_V, Total_cells_N, Rate Law: compartment*beta*Uninfected_Tumor_Cell_U*Oncolytic_Adenovirus_V/Total_cells_N
alpha = 3.0; delta_I = 1.0Reaction: => Oncolytic_Adenovirus_V; Infected_Cancer_Cell_I, Rate Law: compartment*alpha*delta_I*Infected_Cancer_Cell_I
delta_D = 0.35Reaction: Dendritic_Cells_D =>, Rate Law: compartment*delta_D*Dendritic_Cells_D

States:

NameDescription
total tumor cells[cancer; B16-F10 cell]
Infected Cancer Cell I[B16-F10 cell; cancer; Abnormal]
Uninfected Tumor Cell U[cancer; B16-F10 cell]
Dendritic Cells D[dendritic cell; Dendritic Cell]
Total cells N[B16-F10 cell; cancer; Natural Killer T-Cell]
Oncolytic Adenovirus V[Oncolytic; Adenoviridae]
Tumor targeting T cells T[Natural Killer T-Cell; Targeting]

Gevertz2018 - Cancer Treatment with Oncolytic Viruses and Dendritic Cell injections original model: BIOMD0000000816v0.0.1

The model is based on 'Developing a Minimally Structured Mathematical Model of Cancer Treatment with Oncolytic Viruses a…

Details

Mathematical models of biological systems must strike a balance between being sufficiently complex to capture important biological features, while being simple enough that they remain tractable through analysis or simulation. In this work, we rigorously explore how to balance these competing interests when modeling murine melanoma treatment with oncolytic viruses and dendritic cell injections. Previously, we developed a system of six ordinary differential equations containing fourteen parameters that well describes experimental data on the efficacy of these treatments. Here, we explore whether this previously developed model is the minimal model needed to accurately describe the data. Using a variety of techniques, including sensitivity analyses and a parameter sloppiness analysis, we find that our model can be reduced by one variable and three parameters and still give excellent fits to the data. We also argue that our model is not too simple to capture the dynamics of the data, and that the original and minimal models make similar predictions about the efficacy and robustness of protocols not considered in experiments. Reducing the model to its minimal form allows us to increase the tractability of the system in the face of parametric uncertainty. link: http://identifiers.org/pubmed/30510594

Parameters:

NameDescription
C_T = 1.698362Reaction: => Tumor_targeting_T_cells_T; Infected_Cancer_Cell_I, Rate Law: compartment*C_T*Infected_Cancer_Cell_I
delta_I = 1.0Reaction: Infected_Cancer_Cell_I =>, Rate Law: compartment*delta_I*Infected_Cancer_Cell_I
k0 = 2.0; c_kill = 0.595397Reaction: Uninfected_Tumor_Cell_U => ; Infected_Cancer_Cell_I, Tumor_targeting_T_cells_T, Total_cells_N, Rate Law: compartment*(k0+c_kill*Infected_Cancer_Cell_I)*Uninfected_Tumor_Cell_U*Tumor_targeting_T_cells_T/Total_cells_N
chi_D = 4.675397Reaction: => Tumor_targeting_T_cells_T; Dendritic_Cells_D, Rate Law: compartment*chi_D*Dendritic_Cells_D
delta_V = 2.3Reaction: Oncolytic_Adenovirus_V =>, Rate Law: compartment*delta_V*Oncolytic_Adenovirus_V
U_V = 0.0Reaction: => Oncolytic_Adenovirus_V, Rate Law: compartment*U_V
delta_A = 0.35Reaction: Naive_T_cells_A =>, Rate Law: compartment*delta_A*Naive_T_cells_A
chi_A = 1.0Reaction: => Tumor_targeting_T_cells_T; Naive_T_cells_A, Rate Law: compartment*chi_A*Naive_T_cells_A
r = 0.3198Reaction: => Uninfected_Tumor_Cell_U, Rate Law: compartment*r*Uninfected_Tumor_Cell_U
delta_T = 0.35Reaction: Tumor_targeting_T_cells_T =>, Rate Law: compartment*delta_T*Tumor_targeting_T_cells_T
C_A = 5.17E-4Reaction: => Naive_T_cells_A; Infected_Cancer_Cell_I, Rate Law: compartment*C_A*Infected_Cancer_Cell_I
beta = 1.008538Reaction: Uninfected_Tumor_Cell_U => Infected_Cancer_Cell_I; Oncolytic_Adenovirus_V, Total_cells_N, Rate Law: compartment*beta*Uninfected_Tumor_Cell_U*Oncolytic_Adenovirus_V/Total_cells_N
U_D = 0.0Reaction: => Dendritic_Cells_D, Rate Law: compartment*U_D
alpha = 3.0; delta_I = 1.0Reaction: => Oncolytic_Adenovirus_V; Infected_Cancer_Cell_I, Rate Law: compartment*alpha*delta_I*Infected_Cancer_Cell_I
delta_D = 0.35Reaction: Dendritic_Cells_D =>, Rate Law: compartment*delta_D*Dendritic_Cells_D

States:

NameDescription
total tumor cells[B16-F10 cell; cancer]
Infected Cancer Cell I[cancer; B16-F10 cell; Abnormal]
Naive T cells A[Natural Killer T-Cell]
Uninfected Tumor Cell U[cancer; B16-F10 cell]
Dendritic Cells D[Dendritic Cell; dendritic cell]
Total cells N[cancer; B16-F10 cell; Natural Killer T-Cell]
Oncolytic Adenovirus V[Adenoviridae; Oncolytic]
Tumor targeting T cells T[Natural Killer T-Cell; Targeting]

Gex-Fabry1984 - model of receptor-mediated endocytosis of EGF in BALB/c 3T3 cells: BIOMD0000000985v0.0.1

We present a mathematical model for analyzing, simulating, and quantitating the dynamic and steady-state characteristics…

Details

We present a mathematical model for analyzing, simulating, and quantitating the dynamic and steady-state characteristics of receptor-mediated endocytosis. The basic processes considered by the model are ligand-receptor binding, diffusion of receptors and ligand-receptor complexes in the plane of the membrane toward and away from coated pits, binding of ligand-receptor complexes to coated pit proteins, endocytosis of coated pit contents, degradation of ligand, and recycling of undegraded receptors. The model accounts quantitatively for a wide variety of kinetic data and makes new predictions about steady-state characteristics. We show that for homogeneous receptors the slope of the Scatchard plot is not necessarily constant but can have a positive or negative derivative, depending on the concentration of coated pit proteins and their reactivity. This finding suggests that binding data, which show linear and concave curves, might be explainable be a simple coated pit-related mechanism. Similarly the relationship between the x-intercept and the number of receptors is also affected by kinetic parameters controlling endocytosis. We briefly discuss these results in terms of possible mechanisms for the action of tumor promoters, the large variations in receptor number and affinity in the literature, and methods for quantitative characterization of parameters. link: http://identifiers.org/pubmed/6149699

Ghaffari2019 - Thrombomodulin Gene Expression after Retinoic Acid Treatment for Cancer Patients with Coagulation Disorders: MODEL1907140001v0.0.1

Publication includes a mathematical model for how retinoic acid affects thrombomodulin gene and mRNA expression as well…

Details

BACKGROUND:Clinical studies have shown that all-trans retinoic acid (RA), which is often used in treatment of cancer patients, improves hemostatic parameters and bleeding complications such as disseminated intravascular coagulation (DIC). However, the mechanisms underlying this improvement have yet to be elucidated. In vitro studies have reported that RA upregulates thrombomodulin (TM) expression on the endothelial cell surface. The objective of this study was to investigate how and to what extent the TM concentration changes after RA treatment in cancer patients, and how this variation influences the blood coagulation cascade. RESULTS:In this study, we introduced an ordinary differential equation (ODE) model of gene expression for the RA-induced upregulation of TM concentration. Coupling the gene expression model with a two-compartment pharmacokinetic model of RA, we obtained the time-dependent changes in TM and thrombomodulin-mRNA (TMR) concentrations following oral administration of RA. Our results indicated that the TM concentration reached its peak level almost 14 h after taking a single oral dose (110 [Formula: see text]) of RA. Continuous treatment with RA resulted in oscillatory expression of TM on the endothelial cell surface. We then coupled the gene expression model with a mechanistic model of the coagulation cascade, and showed that the elevated levels of TM over the course of RA therapy with a single daily oral dose (110 [Formula: see text]) of RA, reduced the peak thrombin levels and endogenous thrombin potential (ETP) up to 50 and 49%, respectively. We showed that progressive reductions in plasma levels of RA, observed in continuous RA therapy with a once-daily oral dose (110 [Formula: see text]) of RA, did not affect TM-mediated reduction of thrombin generation significantly. This finding prompts the hypothesis that continuous RA treatment has more consistent therapeutic effects on coagulation disorders than on cancer. CONCLUSIONS:Our results indicate that the oscillatory upregulation of TM expression on the endothelial cells over the course of RA therapy could potentially contribute to the treatment of coagulation abnormalities in cancer patients. Further studies on the impacts of RA therapy on the procoagulant activity of cancer cells are needed to better elucidate the mechanisms by which RA therapy improves hemostatic abnormalities in cancer. link: http://identifiers.org/pubmed/30764845

Ghanbari2020 - forecasting the second wave of COVID-19 in Iran: BIOMD0000000976v0.0.1

One of the common misconceptions about COVID-19 disease is to assume that we will not see a recurrence after the first w…

Details

One of the common misconceptions about COVID-19 disease is to assume that we will not see a recurrence after the first wave of the disease has subsided. This completely wrong perception causes people to disregard the necessary protocols and engage in some misbehavior, such as routine socializing or holiday travel. These conditions will put double pressure on the medical staff and endanger the lives of many people around the world. In this research, we are interested in analyzing the existing data to predict the number of infected people in the second wave of out-breaking COVID-19 in Iran. For this purpose, a model is proposed. The mathematical analysis corresponded to the model is also included in this paper. Based on proposed numerical simulations, several scenarios of progress of COVID-19 corresponding to the second wave of the disease in the coming months, will be discussed. We predict that the second wave of will be most severe than the first one. From the results, improving the recovery rate of people with weak immune systems via appropriate medical incentives is resulted as one of the most effective prescriptions to prevent the widespread unbridled outbreak of the second wave of COVID-19. link: http://identifiers.org/pubmed/32834656

Giani2019 - Computational modeling to predict MAP3K8 effects as mediator of resistance to vemurafenib in thyroid cancer stem cells: BIOMD0000000883v0.0.1

Computational modeling to predict MAP3K8 effects as mediator of resistance to vemurafenib in thyroid cancer stem cells

Details

MOTIVATION: Val600Glu (V600E) mutation is the most common BRAF mutation detected in thyroid cancer. Hence, recent research efforts have been performed trying to explore several inhibitors of the V600E mutation-containing BRAF kinase as potential therapeutic options in thyroid cancer refractory to standard interventions. Among them, vemurafenib is a selective BRAF inhibitor approved by FDA for clinical practice. Unfortunately, vemurafenib often displays limited efficacy in poorly differentiated and anaplastic thyroid carcinomas probably because of intrinsic and/or acquired resistance mechanisms. In this view, cancer stem cells may represent a possible mechanism of resistance to vemurafenib, due to their self-renewal and chemo resistance properties.

RESULTS: We present a computational framework to suggest new potential targets to overcome drug resistance. It has been validated with an in vitro model based upon a spheroid-forming method able to isolate thyroid cancer stem cells that may mimic resistance to vemurafenib. Indeed, vemurafenib did not inhibit cell proliferation of BRAF V600E thyroid cancer stem cells, but rather stimulated cell proliferation along with a paradoxical overactivation of ERK and AKT pathways. The computational model identified a fundamental role of mitogen-activated protein kinase 8 (MAP3K8), a serine/threonine kinase expressed in thyroid cancer stem cells, in mediating this drug resistance. To confirm model prediction, we set a suitable in vitro experiment revealing that the treatment with MAP3K8 inhibitor restored the effect of vemurafenib in terms of both DNA fragmentation and PARP cleavage (apoptosis) in thyroid cancer stem cells. Moreover, MAP3K8 expression levels may be a useful marker to predict the response to vemurafenib. link: http://identifiers.org/pubmed/30481266

Parameters:

NameDescription
km=0.1; Kcat=0.096Reaction: species_9 => species_8; species_6, Rate Law: compartment_0*Kcat*species_6*species_9/(km+species_9)
Kcat=0.1; km=0.1Reaction: IKKbeta_IKKalfa_IKKgamma_bRafINH_Active => species_10; bRafMutated, Rate Law: compartment_0*piecewise(Kcat*bRafMutated*IKKbeta_IKKalfa_IKKgamma_bRafINH_Active/(km+IKKbeta_IKKalfa_IKKgamma_bRafINH_Active), bRafMutated <= 1, 0)
k1=0.1Reaction: PDK1Active => PDK1Inactive, Rate Law: compartment_0*k1*PDK1Active
Kcat=0.12; km=0.1Reaction: species_7 => species_6; species_4, Rate Law: compartment_0*Kcat*species_4*species_7/(km+species_7)

States:

NameDescription
species 9[Dual specificity mitogen-activated protein kinase kinase 1; inactive]
FGF[Fibroblast growth factor 1]
TRAF1 TRAF2 TRAF3 Active[TNF receptor-associated factor 2; Q13114; Q13077; Active]
species 1[Epidermal growth factor receptor]
mTORC2Active[Active; Rapamycin-insensitive companion of mTOR; Serine/threonine-protein kinase mTOR; Active]
species 16[RAC-alpha serine/threonine-protein kinase; phosphorylated]
PDK1Inactive[3-phosphoinositide-dependent protein kinase 1; inactive]
species 0[Epidermal growth factor receptor; phosphorylated]
species 25[Pro-epidermal growth factor]
species 8[Dual specificity mitogen-activated protein kinase kinase 1; Active]
species 17[RAC-alpha serine/threonine-protein kinase]
species 15[Phosphatidylinositol 3-kinase regulatory subunit alpha; inactive]
species 2[Active; Son of sevenless homolog 1]
species 6[RAF proto-oncogene serine/threonine-protein kinase; Active]
IKKbeta IKKalfa IKKgamma Inactive[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; inactive]
Tpl2 NF kB bRafINH Inactive[P41279; CHEBI:75047; Nuclear factor NF-kappa-B p105 subunit; inactive]
species 10[Mitogen-activated protein kinase 1; phosphorylated]
freeFGFR[Fibroblast growth factor receptor 1]
species 11[Mitogen-activated protein kinase 1]
pTNFR2[P20333; phosphorylated]
Tpl2 NF kB RasINH Active[Nuclear factor NF-kappa-B p105 subunit; C1902; P41279; Active]
PDK1Active[3-phosphoinositide-dependent protein kinase 1; Active]
bRafMutated[Serine/threonine-protein kinase B-raf; Mutation Abnormality]
species 3[Son of sevenless homolog 1; inactive]
PIP3Inactive[1-phosphatidyl-1D-myo-inositol 4,5-bisphosphate]
IKKbeta IKKalfa IKKgamma Active[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; Active]
TNF[Q5STB3]
freeTNFR1[Tumor necrosis factor receptor superfamily member 1A]
species 4[GTPase HRas; Active]
NIKActive[Mitogen-activated protein kinase kinase kinase 14; Active]
Grb2Inactive[Growth factor receptor-bound protein 2; inactive]
TRADD TRAF2 TRAF5 RIP1 Inactive[TNF receptor-associated factor 2; O00463; Receptor-interacting serine/threonine-protein kinase 1; Tumor necrosis factor receptor type 1-associated DEATH domain protein; inactive]
TRADD TRAF2 TRAF5 RIP1 Active[O00463; Receptor-interacting serine/threonine-protein kinase 1; TNF receptor-associated factor 2; Tumor necrosis factor receptor type 1-associated DEATH domain protein; Active]
TAB1 TAB2 TAB3 TAK1 Active[TGF-beta-activated kinase 1 and MAP3K7-binding protein 3; TGF-beta-activated kinase 1 and MAP3K7-binding protein 2; Mitogen-activated protein kinase kinase kinase 7; Q15750; Active]
pTNFR1[Tumor necrosis factor receptor superfamily member 1A; phosphorylated]
mTORC1Inactive[inactive; Regulatory-associated protein of mTOR; Serine/threonine-protein kinase mTOR; inactive]
freeTNFR2[P20333]
species 5[GTPase HRas; inactive]
pFGFR[Fibroblast growth factor receptor 1; phosphorylated]
mTORC1Active[Active; Regulatory-associated protein of mTOR; Serine/threonine-protein kinase mTOR; Active]
TRAF1 TRAF2 TRAF3 bRafINH Inactive[CHEBI:75047; Q13077; Q13114; TNF receptor-associated factor 2; inactive]
TRAF1 TRAF2 TRAF3 Inactive[Q13114; Q13077; TNF receptor-associated factor 2; inactive]
species 14[Phosphatidylinositol 3-kinase regulatory subunit alpha; Active]
species 7[RAF proto-oncogene serine/threonine-protein kinase; inactive]

Giantsos-Adams2013 - Growth of glycocalyx under static conditions: MODEL1609100001v0.0.1

Giantsos-Adams2013 - Growth of glycocalyx under static conditionsThis model is described in the article: [Heparan Sulfa…

Details

The local hemodynamic shear stress waveforms present in an artery dictate the endothelial cell phenotype. The observed decrease of the apical glycocalyx layer on the endothelium in atheroprone regions of the circulation suggests that the glycocalyx may have a central role in determining atherosclerotic plaque formation. However, the kinetics for the cells' ability to adapt its glycocalyx to the environment have not been quantitatively resolved. Here we report that the heparan sulfate component of the glycocalyx of HUVECs increases by 1.4-fold following the onset of high shear stress, compared to static cultured cells, with a time constant of 19 h. Cell morphology experiments show that 12 h are required for the cells to elongate, but only after 36 h have the cells reached maximal alignment to the flow vector. Our findings demonstrate that following enzymatic degradation, heparan sulfate is restored to the cell surface within 12 h under flow whereas the time required is 20 h under static conditions. We also propose a model describing the contribution of endocytosis and exocytosis to apical heparan sulfate expression. The change in HS regrowth kinetics from static to high-shear EC phenotype implies a differential in the rate of endocytic and exocytic membrane turnover. link: http://identifiers.org/pubmed/23805169

GiantsosAdams2013 - Growth of glycocalyx under static conditions: BIOMD0000000830v0.0.1

Giantsos-Adams2013 - Growth of glycocalyx under static conditionsThis model is described in the article: [Heparan Sulfa…

Details

The local hemodynamic shear stress waveforms present in an artery dictate the endothelial cell phenotype. The observed decrease of the apical glycocalyx layer on the endothelium in atheroprone regions of the circulation suggests that the glycocalyx may have a central role in determining atherosclerotic plaque formation. However, the kinetics for the cells' ability to adapt its glycocalyx to the environment have not been quantitatively resolved. Here we report that the heparan sulfate component of the glycocalyx of HUVECs increases by 1.4-fold following the onset of high shear stress, compared to static cultured cells, with a time constant of 19 h. Cell morphology experiments show that 12 h are required for the cells to elongate, but only after 36 h have the cells reached maximal alignment to the flow vector. Our findings demonstrate that following enzymatic degradation, heparan sulfate is restored to the cell surface within 12 h under flow whereas the time required is 20 h under static conditions. We also propose a model describing the contribution of endocytosis and exocytosis to apical heparan sulfate expression. The change in HS regrowth kinetics from static to high-shear EC phenotype implies a differential in the rate of endocytic and exocytic membrane turnover. link: http://identifiers.org/pubmed/23805169

Parameters:

NameDescription
k3=0.96Reaction: s6 => s1, Rate Law: default*Function_for_k3(default, k3, s6)
k2=0.05Reaction: s2 => s1, Rate Law: default*Function_for_k2(default, k2, s2)
k1=0.05Reaction: s1 => s2, Rate Law: default*Function_for_k1(default, k1, s1)
k8=0.005Reaction: s4 => release, Rate Law: default*Function_for_k8(default, k8, s4)
k7=0.01Reaction: s3 => s4, Rate Law: default*Function_for_k7(default, k7, s3)
k4=0.033Reaction: s1 => shedding, Rate Law: default*Function_for_k4(default, k4, s1)
k6=0.01Reaction: s2 => s3, Rate Law: default*Function_for_k6(default, k6, s2)

States:

NameDescription
s1[Heparan Sulfate]
release[Release]
sheddingshedding
s6[Golgi Apparatus]
s2[early endosome]
s4[lysosome]
s3[late endosome]

Gidvani2012 - DNA replication initiation model in S. Cevevisiae: MODEL2003180001v0.0.1

A quantitative model of the initiation of DNA replication in Saccharomyces cerevisiae predicts the effects of system per…

Details

Eukaryotic cell proliferation involves DNA replication, a tightly regulated process mediated by a multitude of protein factors. In budding yeast, the initiation of replication is facilitated by the heterohexameric origin recognition complex (ORC). ORC binds to specific origins of replication and then serves as a scaffold for the recruitment of other factors such as Cdt1, Cdc6, the Mcm2-7 complex, Cdc45 and the Dbf4-Cdc7 kinase complex. While many of the mechanisms controlling these associations are well documented, mathematical models are needed to explore the network's dynamic behaviour. We have developed an ordinary differential equation-based model of the protein-protein interaction network describing replication initiation.The model was validated against quantified levels of protein factors over a range of cell cycle timepoints. Using chromatin extracts from synchronized Saccharomyces cerevisiae cell cultures, we were able to monitor the in vivo fluctuations of several of the aforementioned proteins, with additional data obtained from the literature. The model behaviour conforms to perturbation trials previously reported in the literature, and accurately predicts the results of our own knockdown experiments. Furthermore, we successfully incorporated our replication initiation model into an established model of the entire yeast cell cycle, thus providing a comprehensive description of these processes.This study establishes a robust model of the processes driving DNA replication initiation. The model was validated against observed cell concentrations of the driving factors, and characterizes the interactions between factors implicated in eukaryotic DNA replication. Finally, this model can serve as a guide in efforts to generate a comprehensive model of the mammalian cell cycle in order to explore cancer-related phenotypes. link: http://identifiers.org/pubmed/22738223

Gilbert2008_ElectrochemicalBiosensor: MODEL1173105855v0.0.1

This a model from the article: A Case Study in Model-driven Synthetic Biology David Gilbert, Monika Heiner, Susan Ro…

Details

We report on a case study in synthetic biology, demonstrating the model-driven design of a self-powering electrochemical biosensor. An essential result of the design process is a general template of a biosensor, which can be instantiated to be adapted to specific pollutants. This template represents a gene expression network extended by metabolic activity. We illustrate the model-based analysis of this template using qualitative, stochastic and continuous Petri nets and related analysis techniques, contributing to a reliable and robust design. link: http://identifiers.org/doi/10.1007/978-0-387-09655-1_15

Gille2010_HepatoNet1_Metabolic_Network: MODEL1009150000v0.0.1

This is the genome-scale metabolic network of a hepatocyte described in the article: HepatoNet1: a comprehensive metab…

Details

We present HepatoNet1, the first reconstruction of a comprehensive metabolic network of the human hepatocyte that is shown to accomplish a large canon of known metabolic liver functions. The network comprises 777 metabolites in six intracellular and two extracellular compartments and 2539 reactions, including 1466 transport reactions. It is based on the manual evaluation of >1500 original scientific research publications to warrant a high-quality evidence-based model. The final network is the result of an iterative process of data compilation and rigorous computational testing of network functionality by means of constraint-based modeling techniques. Taking the hepatic detoxification of ammonia as an example, we show how the availability of nutrients and oxygen may modulate the interplay of various metabolic pathways to allow an efficient response of the liver to perturbations of the homeostasis of blood compounds. link: http://identifiers.org/pubmed/20823849

Giordano2020 - SIDARTHE model of COVID-19 spread in Italy: BIOMD0000000955v0.0.1

In Italy, 128,948 confirmed cases and 15,887 deaths of people who tested positive for SARS-CoV-2 were registered as of 5…

Details

In Italy, 128,948 confirmed cases and 15,887 deaths of people who tested positive for SARS-CoV-2 were registered as of 5 April 2020. Ending the global SARS-CoV-2 pandemic requires implementation of multiple population-wide strategies, including social distancing, testing and contact tracing. We propose a new model that predicts the course of the epidemic to help plan an effective control strategy. The model considers eight stages of infection: susceptible (S), infected (I), diagnosed (D), ailing (A), recognized (R), threatened (T), healed (H) and extinct (E), collectively termed SIDARTHE. Our SIDARTHE model discriminates between infected individuals depending on whether they have been diagnosed and on the severity of their symptoms. The distinction between diagnosed and non-diagnosed individuals is important because the former are typically isolated and hence less likely to spread the infection. This delineation also helps to explain misperceptions of the case fatality rate and of the epidemic spread. We compare simulation results with real data on the COVID-19 epidemic in Italy, and we model possible scenarios of implementation of countermeasures. Our results demonstrate that restrictive social-distancing measures will need to be combined with widespread testing and contact tracing to end the ongoing COVID-19 pandemic. link: http://identifiers.org/pubmed/32322102

Gnügge2016 - Synthetic switch Open loop circuit in yeast: MODEL1804240001v0.0.1

This model was used to describe the behaviour of the synthetic open loop circuit in yeast, published in [1]. The accompa…

Details

Feedback loops in biological networks, among others, enable differentiation and cell cycle progression, and increase robustness in signal transduction. In natural networks, feedback loops are often complex and intertwined, making it challenging to identify which loops are mainly responsible for an observed behavior. However, minimal synthetic replicas could allow for such identification. Here, we engineered a synthetic permease-inducer-repressor system in Saccharomyces cerevisiae to analyze if a transport-mediated positive feedback loop could be a core mechanism for the switch-like behavior in the regulation of metabolic gene networks such as the S. cerevisiae GAL system or the Escherichia coli lac operon. We characterized the synthetic circuit using deterministic and stochastic mathematical models. Similar to its natural counterparts, our synthetic system shows bistable and hysteretic behavior, and the inducer concentration range for bistability as well as the switching rates between the two stable states depend on the repressor concentration. Our results indicate that a generic permease-inducer-repressor circuit with a single feedback loop is sufficient to explain the experimentally observed bistable behavior of the natural systems. We anticipate that the approach of reimplementing natural systems with orthogonal parts to identify crucial network components is applicable to other natural systems such as signaling pathways. link: http://identifiers.org/pubmed/27148753

Goffin2010_L_plantarum_Metabolism: MODEL1011090000v0.0.1

This is the genome scale metabolic reconstruction of Lactobacillus plantarum described in the article: Understanding t…

Details

Situations of extremely low substrate availability, resulting in slow growth, are common in natural environments. To mimic these conditions, Lactobacillus plantarum was grown in a carbon-limited retentostat with complete biomass retention. The physiology of extremely slow-growing L. plantarum–as studied by genome-scale modeling and transcriptomics–was fundamentally different from that of stationary-phase cells. Stress resistance mechanisms were not massively induced during transition to extremely slow growth. The energy-generating metabolism was remarkably stable and remained largely based on the conversion of glucose to lactate. The combination of metabolic and transcriptomic analyses revealed behaviors involved in interactions with the environment, more particularly with plants: production of plant hormones or precursors thereof, and preparedness for the utilization of plant-derived substrates. Accordingly, the production of compounds interfering with plant root development was demonstrated in slow-growing L. plantarum. Thus, conditions of slow growth and limited substrate availability seem to trigger a plant environment-like response, even in the absence of plant-derived material, suggesting that this might constitute an intrinsic behavior in L. plantarum. link: http://identifiers.org/pubmed/20865006

Goldbeter1990_CalciumSpike_CICR: BIOMD0000000098v0.0.1

In a variety of cells, hormonal or neurotransmitter signals elicit a train of intracellular Ca2+ spikes. The analysis of…

Details

The model reproduces the time profile of cytosolic and intracellular calcium as depicted in the upper panel of Fig 2 in the paper. The model was successfully tested on MathSBML and Jarnac.

This model originates from BioModels Database: A Database of Annotated Published Models. It is copyright (c) 2005-2009 The BioModels Team.

For more information see the terms of use.

To cite BioModels Database, please use Le Novère N., Bornstein B., Broicher A., Courtot M., Donizelli M., Dharuri H., Li L., Sauro H., Schilstra M., Shapiro B., Snoep J.L., Hucka M. (2006) BioModels Database: A Free, Centralized Database of Curated, Published, Quantitative Kinetic Models of Biochemical and Cellular Systems Nucleic Acids Res., 34: D689-D691.

Parameters:

NameDescription
v0 = 1.0 uM_per_secReaction: => Z, Rate Law: cytosol*v0
beta = 0.301 dimensionless; v1 = 7.3 uM_per_secReaction: => Z, Rate Law: cytosol*v1*beta
k = 10.0 sec_invReaction: Z =>, Rate Law: cytosol*k*Z
kf = 1.0 sec_invReaction: Y => Z, Rate Law: store*kf*Y
Kr = 2.0 uM; m = 2.0 dimensionless; Ka = 0.9 uM; Vm3 = 500.0 uM_per_sec; p = 4.0 dimensionlessReaction: Y => Z, Rate Law: store*Vm3*Y^m*Z^p/((Kr^m+Y^m)*(Ka^p+Z^p))
K2 = 1.0 uM; n = 2.0 dimensionless; Vm2 = 65.0 uM_per_secReaction: Z => Y, Rate Law: cytosol*Vm2*Z^n/(K2^n+Z^n)

States:

NameDescription
Y[calcium(2+); Calcium cation]
Z[calcium(2+); Calcium cation]

Goldbeter1991 - Min Mit Oscil: BIOMD0000000003v0.0.1

Goldbeter1991 - Min Mit OscilMinimal cascade model for the mitotic oscillator involving cyclin and cdc2 kinase. This mo…

Details

A minimal model for the mitotic oscillator is presented. The model, built on recent experimental advances, is based on the cascade of post-translational modification that modulates the activity of cdc2 kinase during the cell cycle. The model pertains to the situation encountered in early amphibian embryos, where the accumulation of cyclin suffices to trigger the onset of mitosis. In the first cycle of the bicyclic cascade model, cyclin promotes the activation of cdc2 kinase through reversible dephosphorylation, and in the second cycle, cdc2 kinase activates a cyclin protease by reversible phosphorylation. That cyclin activates cdc2 kinase while the kinase triggers the degradation of cyclin has suggested that oscillations may originate from such a negative feedback loop [Félix, M. A., Labbé, J. C., Dorée, M., Hunt, T. & Karsenti, E. (1990) Nature (London) 346, 379-382]. This conjecture is corroborated by the model, which indicates that sustained oscillations of the limit cycle type can arise in the cascade, provided that a threshold exists in the activation of cdc2 kinase by cyclin and in the activation of cyclin proteolysis by cdc2 kinase. The analysis shows how miototic oscillations may readily arise from time lags associated with these thresholds and from the delayed negative feedback provided by cdc2-induced cyclin degradation. A mechanism for the origin of the thresholds is proposed in terms of the phenomenon of zero-order ultrasensitivity previously described for biochemical systems regulated by covalent modification. link: http://identifiers.org/pubmed/1833774

Parameters:

NameDescription
K2=0.005; V2=1.5Reaction: M =>, Rate Law: cell*M*V2*(K2+M)^-1
K1=0.005; V1 = NaNReaction: => M, Rate Law: cell*(1+-1*M)*V1*(K1+-1*M+1)^-1
kd=0.01Reaction: C =>, Rate Law: C*cell*kd
vd=0.25; Kd=0.02Reaction: C => ; X, Rate Law: C*cell*vd*X*(C+Kd)^-1
vi=0.025Reaction: => C, Rate Law: cell*vi
K3=0.005; V3 = NaNReaction: => X, Rate Law: cell*V3*(1+-1*X)*(K3+-1*X+1)^-1
V4=0.5; K4=0.005Reaction: X =>, Rate Law: cell*V4*X*(K4+X)^-1

States:

NameDescription
M[Cyclin-dependent kinase 1-B; Cyclin-dependent kinase 1-A]
C[Cyclin-C; IPR006670]
X[anaphase-promoting complex; CDC20:p-APC/C [cytosol]]

Goldbeter1991 - Min Mit Oscil, Expl Inact: BIOMD0000000004v0.0.1

Goldbeter1991 - Min Mit Oscil, Expl InactThis model represents the inactive forms of CDC-2 Kinase and Cyclin Protease as…

Details

A minimal model for the mitotic oscillator is presented. The model, built on recent experimental advances, is based on the cascade of post-translational modification that modulates the activity of cdc2 kinase during the cell cycle. The model pertains to the situation encountered in early amphibian embryos, where the accumulation of cyclin suffices to trigger the onset of mitosis. In the first cycle of the bicyclic cascade model, cyclin promotes the activation of cdc2 kinase through reversible dephosphorylation, and in the second cycle, cdc2 kinase activates a cyclin protease by reversible phosphorylation. That cyclin activates cdc2 kinase while the kinase triggers the degradation of cyclin has suggested that oscillations may originate from such a negative feedback loop [Félix, M. A., Labbé, J. C., Dorée, M., Hunt, T. & Karsenti, E. (1990) Nature (London) 346, 379-382]. This conjecture is corroborated by the model, which indicates that sustained oscillations of the limit cycle type can arise in the cascade, provided that a threshold exists in the activation of cdc2 kinase by cyclin and in the activation of cyclin proteolysis by cdc2 kinase. The analysis shows how miototic oscillations may readily arise from time lags associated with these thresholds and from the delayed negative feedback provided by cdc2-induced cyclin degradation. A mechanism for the origin of the thresholds is proposed in terms of the phenomenon of zero-order ultrasensitivity previously described for biochemical systems regulated by covalent modification. link: http://identifiers.org/pubmed/1833774

Parameters:

NameDescription
K2=0.005; V2=1.5Reaction: M => MI, Rate Law: cell*M*V2*(K2+M)^-1
K1=0.005; V1 = NaNReaction: MI => M, Rate Law: cell*MI*V1*(K1+MI)^-1
kd=0.01Reaction: C =>, Rate Law: C*cell*kd
vd=0.25; Kd=0.02Reaction: C => ; X, Rate Law: C*cell*vd*X*(C+Kd)^-1
vi=0.025Reaction: => C, Rate Law: cell*vi
K3=0.005; V3 = NaNReaction: XI => X, Rate Law: cell*V3*XI*(K3+XI)^-1
V4=0.5; K4=0.005Reaction: X => XI, Rate Law: cell*V4*X*(K4+X)^-1

States:

NameDescription
MI[Cyclin-dependent kinase 1-B; Cyclin-dependent kinase 1-A]
M[Cyclin-dependent kinase 1-B; Cyclin-dependent kinase 1-A]
C[IPR006670]
XIInactive Cyclin Protease
XActive Cyclin Protease

Goldbeter1995_CircClock: BIOMD0000000016v0.0.1

To the extent possible under law, all copyright and related or neighbouring rights to this encoded model have been dedic…

Details

The mechanism of circadian oscillations in the period protein (PER) in Drosophila is investigated by means of a theoretical model. Taking into account recent experimental observations, the model for the circadian clock is based on multiple phosphorylation of PER and on the negative feedback exerted by PER on the transcription of the period (per) gene. This minimal biochemical model provides a molecular basis for circadian oscillations of the limit cycle type. During oscillations, the peak in per mRNA precedes by several hours the peak in total PER protein. The results support the view that multiple PER phosphorylation introduces times delays which strengthen the capability of negative feedback to produce oscillations. The analysis shows that the rhythm only occurs in a range bounded by two critical values of the maximum rate of PER degradation. A similar result is obtained with respect to the rate of PER transport into the nucleus. The results suggest a tentative explanation for the altered period of per mutants, in terms of variations in the rate of PER degradation. link: http://identifiers.org/pubmed/8587874

Parameters:

NameDescription
K3=2.0; V3=5.0Reaction: P1 => P2, Rate Law: CYTOPLASM*V3*P1/(K3+P1)
V4=2.5; K4=2.0Reaction: P2 => P1, Rate Law: CYTOPLASM*V4*P2/(K4+P2)
ks=0.38Reaction: EmptySet => P0; M, Rate Law: ks*M*default
Vm=0.65; Km=0.5Reaction: M => EmptySet, Rate Law: Vm*M*CYTOPLASM/(Km+M)
k2=1.3Reaction: Pn => P2, Rate Law: k2*Pn*compartment_0000004
K2=2.0; V2=1.58Reaction: P1 => P0, Rate Law: CYTOPLASM*V2*P1/(K2+P1)
KI=1.0; Vs=0.76; n=4.0Reaction: EmptySet => M; Pn, Rate Law: default*Vs*KI^n/(KI^n+Pn^n)
k1=1.9Reaction: P2 => Pn, Rate Law: k1*P2*CYTOPLASM
Vd=0.95; Kd=0.2Reaction: P2 => EmptySet, Rate Law: CYTOPLASM*Vd*P2/(Kd+P2)
K1=2.0; V1=3.2Reaction: P0 => P1, Rate Law: CYTOPLASM*V1*P0/(K1+P0)

States:

NameDescription
M[messenger RNA; RNA]
Pn[Period circadian protein]
Pt[Period circadian protein]
P2[Period circadian protein]
P1[Period circadian protein]
P0[Period circadian protein]

Goldbeter1996 - Cyclin Cdc2 kinase Oscillations: BIOMD0000000729v0.0.1

We consider a minimal cascade model previously proposed for the mitotic oscillator driving the embryonic cell division c…

Details

We consider a minimal cascade model previously proposed for the mitotic oscillator driving the embryonic cell division cycle. The model is based on a bicyclic phosphorylation-dephosphorylation cascade involving cyclin and cdc2 kinase. By constructing stability diagrams showing domains of periodic behavior as a function of the maximum rates of the kinases and phosphatases involved in the two cycles of the cascade, we investigate the role of these converter enzymes in the oscillatory mechanism. Oscillations occur when the balance of kinase and phosphatase rates in each cycle is in a range bounded by two critical values. The results suggest ways to arrest the mitotic oscillator by altering the maximum rates of the converter enzymes. These results bear on the control of cell proliferation. link: http://identifiers.org/pubmed/8631387

Parameters:

NameDescription
V3 = 0.0; K3 = 0.01Reaction: => X, Rate Law: compartment*V3*(1-X)/((K3+1)-X)
K4 = 0.01; V4 = 0.5Reaction: X =>, Rate Law: compartment*V4*X/(K4+X)
Kd = 0.02; vd = 0.25Reaction: C => ; X, Rate Law: compartment*vd*X*C/(Kd+C)
V2 = 1.5; K2 = 0.01Reaction: M =>, Rate Law: compartment*V2*M/(K2+M)
vi = 0.05Reaction: => C, Rate Law: compartment*vi
K1 = 0.01; V1 = 0.0Reaction: => M, Rate Law: compartment*V1*(1-M)/((K1+1)-M)
kd = 0.01Reaction: C =>, Rate Law: compartment*kd*C

States:

NameDescription
M[Kinase]
C[Rate Constant; Guanidine]
X[Phosphatase]

Goldbeter2006_weightCycling: BIOMD0000000079v0.0.1

This model is according to the paper of *A model for the dynamics of human weight cycling* by A. Goldbeter 2006.The fig…

Details

The resolution to lose weight by cognitive restraint of nutritional intake often leads to repeated bouts of weight loss and regain, a phenomenon known as weight cycling or "yo-yo dieting". A simple mathematical model for weight cycling is presented. The model is based on a feedback of psychological nature by which a subject decides to reduce dietary intake once a threshold weight is exceeded. The analysis of the model indicates that sustained oscillations in body weight occur in a parameter range bounded by critical values. Only outside this range can body weight reach a stable steady state. The model provides a theoretical framework that captures key facets of weight cycling and suggests ways to control the phenomenon. The view that weight cycling represents self-sustained oscillations has indeed specific implications. In dynamical terms, to bring weight cycling to an end, parameter values should change in such a way as to induce the transition of body weight from sustained oscillations around an unstable steady state to a stable steady state. Maintaining weight under a critical value should prevent weight cycling and allow body weight to stabilize below the oscillatory range. link: http://identifiers.org/pubmed/16595882

Parameters:

NameDescription
V=0.1; Km=0.2Reaction: P =>, Rate Law: V*P/(Km+P)
k3=0.01; V3=6.0Reaction: => R; P, Rate Law: P*V3*(1-R)/(k3+(1-R))
K1=0.01; V1=1.0Reaction: => Q, Rate Law: V1*(1-Q)/(K1+(1-Q))
K2=0.01; V2=1.5Reaction: Q => ; R, Rate Law: V2*R*Q/(K2+Q)
a=0.1Reaction: => P; Q, Rate Law: body*a*Q
V=2.5; Km=0.01Reaction: R =>, Rate Law: V*R/(Km+R)

States:

NameDescription
QQ
PP
RR

Goldbeter2007_Somitogenesis_Switch: BIOMD0000000275v0.0.1

This is the simple model without diffusion described in th epublication Sharp developmental thresholds defined through…

Details

The establishment of thresholds along morphogen gradients in the embryo is poorly understood. Using mathematical modeling, we show that mutually inhibitory gradients can generate and position sharp morphogen thresholds in the embryonic space. Taking vertebrate segmentation as a paradigm, we demonstrate that the antagonistic gradients of retinoic acid (RA) and Fibroblast Growth Factor (FGF) along the presomitic mesoderm (PSM) may lead to the coexistence of two stable steady states. Here, we propose that this bistability is associated with abrupt switches in the levels of FGF and RA signaling, which permit the synchronized activation of segmentation genes, such as mesp2, in successive cohorts of PSM cells in response to the segmentation clock, thereby defining the future segments. Bistability resulting from mutual inhibition of RA and FGF provides a molecular mechanism for the all-or-none transitions assumed in the "clock and wavefront" somitogenesis model. Given that mutually antagonistic signaling gradients are common in development, such bistable switches could represent an important principle underlying embryonic patterning. link: http://identifiers.org/pubmed/17497689

Parameters:

NameDescription
kd4 = 1.0 per minReaction: F =>, Rate Law: PSM*kd4*F
m = 2.0 dimensionless; Ki = 0.2 nM; ks3 = 1.0 per minReaction: => F; RA, M_F, Rate Law: PSM*ks3*M_F*Ki^m/(Ki^m+RA^m)
vs1 = NaN nM per minReaction: => RA, Rate Law: PSM*vs1
ks2 = 1.0 per minReaction: => C; M_C, Rate Law: PSM*ks2*M_C
kd5 = 0.0 per minReaction: RA =>, Rate Law: PSM*kd5*RA
kd1 = 1.0 per nM per minReaction: RA => ; C, Rate Law: PSM*kd1*RA*C
kd3 = 1.0 per minReaction: M_C =>, Rate Law: PSM*kd3*M_C
kd2 = 0.28 per minReaction: C =>, Rate Law: PSM*kd2*C
Ka = 0.2 nM; Vsc = 7.1 nM per min; n = 2.0 dimensionless; V0 = 0.365 nM per minReaction: => M_C; F, Rate Law: PSM*(V0+Vsc*F^n/(Ka^n+F^n))
L = 50.0 arbit. length; M_0 = 5.0 nM; x = 15.0 arbit. lengthReaction: M_F = M_0*x/L, Rate Law: missing

States:

NameDescription
RA[retinoic acid]
C[retinoic acid 4-hydroxylase activity; CYP26C; CYP26B; CYP26A; Cytochrome P450 26B1; Cytochrome P450 26C1; Cytochrome P450 26A1; IPR001128]
M F[messenger RNA]
M C[IPR001128; m7G(5')pppR-mRNA; messenger RNA]
F[type 2 fibroblast growth factor receptor binding; type 1 fibroblast growth factor receptor binding; growth factor activity; Fibroblast growth factor 8; IPR002348]

Goldbeter2008_Somite_Segmentation_Clock_Notch_Wnt_FGF: BIOMD0000000201v0.0.1

This is a model of the coupled Natch, Wnt and FGF modules as described in: **A. Goldbeter and O. Pourquié** , Modelin…

Details

The formation of somites in the course of vertebrate segmentation is governed by an oscillator known as the segmentation clock, which is characterized by a period ranging from 30 min to a few hours depending on the organism. This oscillator permits the synchronized activation of segmentation genes in successive cohorts of cells in the presomitic mesoderm in response to a periodic signal emitted by the segmentation clock, thereby defining the future segments. Recent microarray experiments [Dequeant, M.L., Glynn, E., Gaudenz, K., Wahl, M., Chen, J., Mushegian, A., Pourquie, O., 2006. A complex oscillating network of signaling genes underlies the mouse segmentation clock. Science 314, 1595-1598] indicate that the Notch, Wnt and Fibroblast Growth Factor (FGF) signaling pathways are involved in the mechanism of the segmentation clock. By means of computational modeling, we investigate the conditions in which sustained oscillations occur in these three signaling pathways. First we show that negative feedback mediated by the Lunatic Fringe protein on intracellular Notch activation can give rise to periodic behavior in the Notch pathway. We then show that negative feedback exerted by Axin2 on the degradation of beta-catenin through formation of the Axin2 destruction complex can produce oscillations in the Wnt pathway. Likewise, negative feedback on FGF signaling mediated by the phosphatase product of the gene MKP3/Dusp6 can produce oscillatory gene expression in the FGF pathway. Coupling the Wnt, Notch and FGF oscillators through common intermediates can lead to synchronized oscillations in the three signaling pathways or to complex periodic behavior, depending on the relative periods of oscillations in the three pathways. The phase relationships between cycling genes in the three pathways depend on the nature of the coupling between the pathways and on their relative autonomous periods. The model provides a framework for analyzing the dynamics of the segmentation clock in terms of a network of oscillating modules involving the Wnt, Notch and FGF signaling pathways. link: http://identifiers.org/pubmed/18308339

Parameters:

NameDescription
theta = 1.5 dimensionless; vsB = 0.087 fluxReaction: => B, Rate Law: theta*cytosol*vsB
theta = 1.5 dimensionless; VMK = 5.08 flux; K1 = 0.28 nanomolar; KID = 0.5 nanomolarReaction: B => Bp; AK, D, Kt, Rate Law: theta*cytosol*VMK*KID/(KID+D)*B/(K1+B)*AK/Kt
KaErk = 0.05 nanomolar; eta = 0.3 dimensionless; VMaErk = 3.3 fluxReaction: => ERKa; ERKi, Rasa, Rast, Rate Law: eta*cytosol*VMaErk*Rasa/Rast*ERKi/(KaErk+ERKi)
v0 = 0.06 flux; theta = 1.5 dimensionlessReaction: => MAx; BN, Rate Law: theta*cytosol*v0
theta = 1.5 dimensionless; vmd = 0.8 flux; Kmd = 0.48 nanomolarReaction: MAx =>, Rate Law: theta*cytosol*vmd*MAx/(Kmd+MAx)
vdDusp = 2.0 flux; eta = 0.3 dimensionless; KdDusp = 0.5 nanomolarReaction: Dusp =>, Rate Law: eta*cytosol*vdDusp*Dusp/(KdDusp+Dusp)
VMaX = 1.6 flux; eta = 0.3 dimensionless; KaX = 0.05 nanomolarReaction: => Xa; ERKa, ERKt, Xi, Rate Law: eta*cytosol*VMaX*ERKa/ERKt*Xi/(KaX+Xi)
theta = 1.5 dimensionless; a1 = 1.8 second_order_rate_constant; d1 = 0.1 first_order_rate_constantReaction: AK => A + K, Rate Law: theta*cytosol*(d1*AK-a1*A*K)
theta = 1.5 dimensionless; ksAx = 0.02 first_order_rate_constantReaction: => A; MAx, Rate Law: theta*cytosol*ksAx*MAx
KdNa = 0.001 nanomolar; VdNa = 0.01 flux; epsilon = 0.3 dimensionlessReaction: Na =>, Rate Law: epsilon*cytosol*VdNa*Na/(KdNa+Na)
theta = 1.5 dimensionless; K2 = 0.03 nanomolar; VMP = 1.0 fluxReaction: Bp => B, Rate Law: theta*cytosol*VMP*Bp/(K2+Bp)
q = 2.0 dimensionless; KaMDusp = 0.5 nanomolar; eta = 0.3 dimensionless; VMsMDusp = 0.9 fluxReaction: => MDusp; Xa, Rate Law: eta*cytosol*VMsMDusp*Xa^q/(KaMDusp^q+Xa^q)
VdNan = 0.1 flux; KdNan = 0.001 nanomolar; epsilon = 0.3 dimensionlessReaction: Nan =>, Rate Law: epsilon*cytosol*VdNan*Nan/(KdNan+Nan)
VMaRas = 4.968 flux; KaFgf = 0.5 nanomolar; eta = 0.3 dimensionless; r = 2.0 dimensionless; KaRas = 0.103 nanomolarReaction: => Rasa; Rasi, Fgf, Rate Law: eta*cytosol*VMaRas*Fgf^r/(KaFgf^r+Fgf^r)*Rasi/(KaRas+Rasi)
vdN = 2.82 flux; KdN = 1.4 nanomolar; epsilon = 0.3 dimensionlessReaction: N =>, Rate Law: epsilon*cytosol*vdN*N/(KdN+N)
KdErk = 0.05 nanomolar; eta = 0.3 dimensionless; kcDusp = 1.35 first_order_rate_constantReaction: ERKa => ; Dusp, Rate Law: eta*cytosol*kcDusp*Dusp*ERKa/(KdErk+ERKa)
theta = 1.5 dimensionless; kd1 = 0.0 first_order_rate_constantReaction: B =>, Rate Law: theta*cytosol*kd1*B
VMdMDusp = 0.5 flux; eta = 0.3 dimensionless; KdMDusp = 0.5 nanomolarReaction: MDusp =>, Rate Law: eta*cytosol*VMdMDusp*MDusp/(KdMDusp+MDusp)
vdF = 0.39 flux; KdF = 0.37 nanomolar; epsilon = 0.3 dimensionlessReaction: F =>, Rate Law: epsilon*cytosol*vdF*F/(KdF+F)
ksF = 0.3 first_order_rate_constant; epsilon = 0.3 dimensionlessReaction: => F; MF, Rate Law: epsilon*cytosol*ksF*MF
vmF = 1.92 flux; KdMF = 0.768 nanomolar; epsilon = 0.3 dimensionlessReaction: MF =>, Rate Law: epsilon*cytosol*vmF*MF/(KdMF+MF)
theta = 1.5 dimensionless; m = 2.0 dimensionless; vMXa = 0.5 flux; KaXa = 0.05 nanomolarReaction: => MAx; Xa, Rate Law: theta*cytosol*vMXa*Xa^m/(KaXa^m+Xa^m)
kt2 = 0.1 first_order_rate_constant; kt1 = 0.1 first_order_rate_constant; epsilon = 0.3 dimensionlessReaction: Na => Nan, Rate Law: epsilon*cytosol*(kt1*Na-kt2*Nan)
theta = 1.5 dimensionless; KaB = 0.7 nanomolar; vMB = 1.64 flux; n = 2.0 dimensionlessReaction: => MAx; BN, Rate Law: theta*cytosol*vMB*BN^n/(KaB^n+BN^n)
KdAx = 0.63 nanomolar; theta = 1.5 dimensionless; vdAx = 0.6 fluxReaction: A =>, Rate Law: theta*cytosol*vdAx*A/(KdAx+A)
VMdX = 0.5 flux; eta = 0.3 dimensionless; KdX = 0.05 nanomolarReaction: Xa =>, Rate Law: eta*cytosol*VMdX*Xa/(KdX+Xa)
ksDusp = 0.5 first_order_rate_constant; eta = 0.3 dimensionlessReaction: => Dusp; MDusp, Rate Law: eta*cytosol*ksDusp*MDusp
theta = 1.5 dimensionless; kd2 = 7.062 first_order_rate_constantReaction: Bp =>, Rate Law: theta*cytosol*kd2*Bp
vsFK = NaN flux; p = 2.0 dimensionless; KA = 0.05 nanomolar; epsilon = 0.3 dimensionlessReaction: => MF; Nan, Rate Law: epsilon*cytosol*vsFK*Nan^p/(KA^p+Nan^p)
j = 2.0 dimensionless; KIF = 0.5 nanomolar; epsilon = 0.3 dimensionless; kc = 3.45 first_order_rate_constantReaction: N => Na; F, Rate Law: epsilon*cytosol*kc*N*KIF^j/(KIF^j+F^j)
theta = 1.5 dimensionless; kt3 = 0.7 first_order_rate_constant; kt4 = 1.5 first_order_rate_constantReaction: BN => B, Rate Law: theta*cytosol*(kt4*BN-kt3*B)
vsN = 0.23 flux; epsilon = 0.3 dimensionlessReaction: => N, Rate Law: cytosol*epsilon*vsN
VMdRas = 0.41 flux; KdRas = 0.1 nanomolar; eta = 0.3 dimensionlessReaction: Rasa =>, Rate Law: eta*cytosol*VMdRas*Rasa/(KdRas+Rasa)

States:

NameDescription
Na[IPR008297]
Xi[IPR006715]
Dusp[Dual specificity protein phosphatase 6]
A[Axin-2]
Rasi[GDP; IPR001806]
Nan[IPR008297]
ERKa[IPR008349]
Xa[IPR006715]
MF[messenger RNA; Beta-1,3-N-acetylglucosaminyltransferase lunatic fringe; IPR017374]
Bp[Catenin beta-1; IPR013284; Phosphoprotein]
B[IPR013284]
MAx[messenger RNA; AXIN2; Axin-2]
BN[Catenin beta-1; IPR013284]
MDusp[Dual specificity protein phosphatase 6; DUSP6]
N[Neurogenic locus notch homolog protein 1; IPR008297]
AK[Axin-2; Glycogen synthase kinase-3 beta]
K[Glycogen synthase kinase-3 beta]
ERKi[IPR008349]
F[IPR017374; Beta-1,3-N-acetylglucosaminyltransferase lunatic fringe]
Rasa[GTP; IPR001806]

Goldbeter2013-Oscillatory activity of cyclin-dependent kinases in the cell cycle: BIOMD0000000944v0.0.1

A model for oscillations of Cdc2 kinase in embryonic cell cycles based on Michaelis–Menten phosphorylation–dephosphoryla…

Details

Oscillations occur in a number of enzymatic systems as a result of feedback regulation. How Michaelis-Menten kinetics influences oscillatory behavior in enzyme systems is investigated in models for oscillations in the activity of phosphofructokinase (PFK) in glycolysis and of cyclin-dependent kinases in the cell cycle. The model for the PFK reaction is based on a product-activated allosteric enzyme reaction coupled to enzymatic degradation of the reaction product. The Michaelian nature of the product decay term markedly influences the period, amplitude and waveform of the oscillations. Likewise, a model for oscillations of Cdc2 kinase in embryonic cell cycles based on Michaelis-Menten phosphorylation-dephosphorylation kinetics shows that the occurrence and amplitude of the oscillations strongly depend on the ultrasensitivity of the enzymatic cascade that controls the activity of the cyclin-dependent kinase. link: http://identifiers.org/pubmed/23892075

Parameters:

NameDescription
vs = 0.06Reaction: => Cyclin, Rate Law: compartment*vs
V4 = 0.7; K4 = 0.01Reaction: Active_APC =>, Rate Law: compartment*V4*Active_APC/(K4+Active_APC)
kd = 0.046; vd = 0.25; Kd = 0.001Reaction: Cyclin => ; Active_APC, Rate Law: compartment*(vd*Active_APC*Cyclin/(Kd+Cyclin)+kd*Cyclin)
K1 = 0.002; V1 = 0.0784313725490196Reaction: => Active_Cdc2_kinase, Rate Law: compartment*V1*(1-Active_Cdc2_kinase)/(K1+(1-Active_Cdc2_kinase))
K2 = 0.002; V2 = 2.0Reaction: Active_Cdc2_kinase =>, Rate Law: compartment*V2*Active_Cdc2_kinase/(K2+Active_Cdc2_kinase)
V3 = 0.01; K3 = 0.01Reaction: => Active_APC, Rate Law: compartment*V3*(1-Active_APC)/(K3+(1-Active_APC))

States:

NameDescription
Cyclin[Guanidine]
Active Cdc2 kinase[0016746]
Active APC[anaphase-promoting complex]

Golomb2006_SomaticBursting: BIOMD0000000118v0.0.1

Model is according to the paper *Contribution of Persistent Na+ Current and M-Type K+ Current to Somatic Bursting in CA1…

Details

The intrinsic firing modes of adult CA1 pyramidal cells vary along a continuum of "burstiness" from regular firing to rhythmic bursting, depending on the ionic composition of the extracellular milieu. Burstiness is low in neurons exposed to a normal extracellular Ca(2+) concentration (Ca(2+)), but is markedly enhanced by lowering Ca(2+), although not by blocking Ca(2+) and Ca(2+)-activated K(+) currents. We show, using intracellular recordings, that burstiness in low Ca(2+) persists even after truncating the apical dendrites, suggesting that bursts are generated by an interplay of membrane currents at or near the soma. To study the mechanisms of bursting, we have constructed a conductance-based, one-compartment model of CA1 pyramidal neurons. In this neuron model, reduced Ca(2+) is simulated by negatively shifting the activation curve of the persistent Na(+) current (I(NaP)) as indicated by recent experimental results. The neuron model accounts, with different parameter sets, for the diversity of firing patterns observed experimentally in both zero and normal Ca(2+). Increasing I(NaP) in the neuron model induces bursting and increases the number of spikes within a burst but is neither necessary nor sufficient for bursting. We show, using fast-slow analysis and bifurcation theory, that the M-type K(+) current (I(M)) allows bursting by shifting neuronal behavior between a silent and a tonically active state provided the kinetics of the spike generating currents are sufficiently, although not extremely, fast. We suggest that bursting in CA1 pyramidal cells can be explained by a single compartment "square bursting" mechanism with one slow variable, the activation of I(M). link: http://identifiers.org/pubmed/16807352

Golomb2006_SomaticBursting_nonzero[Ca]: BIOMD0000000119v0.0.1

Model is according to the paper *Contribution of Persistent Na+ Current and M-Type K+ Current to Somatic Bursting in CA1…

Details

The intrinsic firing modes of adult CA1 pyramidal cells vary along a continuum of "burstiness" from regular firing to rhythmic bursting, depending on the ionic composition of the extracellular milieu. Burstiness is low in neurons exposed to a normal extracellular Ca(2+) concentration (Ca(2+)), but is markedly enhanced by lowering Ca(2+), although not by blocking Ca(2+) and Ca(2+)-activated K(+) currents. We show, using intracellular recordings, that burstiness in low Ca(2+) persists even after truncating the apical dendrites, suggesting that bursts are generated by an interplay of membrane currents at or near the soma. To study the mechanisms of bursting, we have constructed a conductance-based, one-compartment model of CA1 pyramidal neurons. In this neuron model, reduced Ca(2+) is simulated by negatively shifting the activation curve of the persistent Na(+) current (I(NaP)) as indicated by recent experimental results. The neuron model accounts, with different parameter sets, for the diversity of firing patterns observed experimentally in both zero and normal Ca(2+). Increasing I(NaP) in the neuron model induces bursting and increases the number of spikes within a burst but is neither necessary nor sufficient for bursting. We show, using fast-slow analysis and bifurcation theory, that the M-type K(+) current (I(M)) allows bursting by shifting neuronal behavior between a silent and a tonically active state provided the kinetics of the spike generating currents are sufficiently, although not extremely, fast. We suggest that bursting in CA1 pyramidal cells can be explained by a single compartment "square bursting" mechanism with one slow variable, the activation of I(M). link: http://identifiers.org/pubmed/16807352

Parameters:

NameDescription
ICa = NaN; tauCa = 13.0; uuCa = 0.13Reaction: => Ca, Rate Law: compartment_0000001*((-uuCa)*ICa-Ca)/tauCa

States:

NameDescription
Ca[calcium(2+); Calcium cation]

Gomez-Cabrero2011_Atherogenesis: MODEL1002160000v0.0.1

This model is from the article: Workflow for generating competing hypothesis from models with parameter uncertainty.…

Details

Mathematical models are increasingly used in life sciences. However, contrary to other disciplines, biological models are typically over-parametrized and loosely constrained by scarce experimental data and prior knowledge. Recent efforts on analysis of complex models have focused on isolated aspects without considering an integrated approach-ranging from model building to derivation of predictive experiments and refutation or validation of robust model behaviours. Here, we develop such an integrative workflow, a sequence of actions expanding upon current efforts with the purpose of setting the stage for a methodology facilitating an extraction of core behaviours and competing mechanistic hypothesis residing within underdetermined models. To this end, we make use of optimization search algorithms, statistical (machine-learning) classification techniques and cluster-based analysis of the state variables' dynamics and their corresponding parameter sets. We apply the workflow to a mathematical model of fat accumulation in the arterial wall (atherogenesis), a complex phenomena with limited quantitative understanding, thus leading to a model plagued with inherent uncertainty. We find that the mathematical atherogenesis model can still be understood in terms of a few key behaviours despite the large number of parameters. This result enabled us to derive distinct mechanistic predictions from the model despite the lack of confidence in the model parameters. We conclude that building integrative workflows enable investigators to embrace modelling of complex biological processes despite uncertainty in parameters. link: http://identifiers.org/pubmed/22670212

Gonzalez2010_N_pharaonis_metabolism: MODEL1011080001v0.0.1

This is metabolic network reconstruction of Natronomonas pharaonis described in the article Characterization of growt…

Details

Natronomonas pharaonis is an archaeon adapted to two extreme conditions: high salt concentration and alkaline pH. It has become one of the model organisms for the study of extremophilic life. Here, we present a genome-scale, manually curated metabolic reconstruction for the microorganism. The reconstruction itself represents a knowledge base of the haloalkaliphile's metabolism and, as such, would greatly assist further investigations on archaeal pathways. In addition, we experimentally determined several parameters relevant to growth, including a characterization of the biomass composition and a quantification of carbon and oxygen consumption. Using the metabolic reconstruction and the experimental data, we formulated a constraints-based model which we used to analyze the behavior of the archaeon when grown on a single carbon source. Results of the analysis include the finding that Natronomonas pharaonis, when grown aerobically on acetate, uses a carbon to oxygen consumption ratio that is theoretically near-optimal with respect to growth and energy production. This supports the hypothesis that, under simple conditions, the microorganism optimizes its metabolism with respect to the two objectives. We also found that the archaeon has a very low carbon efficiency of only about 35%. This inefficiency is probably due to a very low P/O ratio as well as to the other difficulties posed by its extreme environment. link: http://identifiers.org/pubmed/20543878

GonzalezHeydrich1994_HPAaxisRegulation_CortisolProduction: MODEL0911270004v0.0.1

This a model from the article: A computer simulation of the hypothalamic-pituitary-adrenal axis. Gonzalez-Heydrich J…

Details

This paper describes the construction of a computer model that simulates the hypothalamic-pituitary-adrenal axis (HPA axis) regulation of cortisol production. It is presented to illustrate the process of physiological modeling using standard "off the shelf" technologies. The model simulates components of the HPA axis involved in the continuous secretion and elimination of cortisol, adrenocorticotropin (ACTH), and corticotropin releasing hormone (CRH). The physiological relations of these component pieces were modeled based on the current knowledge of their functioning. Rate constants, half lives, and receptor affinities were assigned values derived from the experimental literature. At its current level of development the model is able to accurately simulate the timing, magnitude and decay of the ACTH and cortisol concentration peaks resulting from the ovine-CRH stimulation test in normal and hypercortisolemic patients. The model will be used to predict the effects of lesions in different components of the HPA axis on the time course of cortisol and ACTH levels. We plan to use the model to explore the experimental conditions required to distinguish mechanisms underlying various disorders of the HPA axis, particularly depression. Efforts are currently underway to validate the model for a large variety of normal and pathological perturbations of the HPA axis. link: http://identifiers.org/pubmed/7949852

GonzalezMiranda2013 - The effect of circadian oscillations on biochemical cell signaling by NF-κB: BIOMD0000000893v0.0.1

This is a mathematical model for NF-κB oscillations, described by a set of ordinary nonlinear differential equations, wh…

Details

We report the results of a numerical investigation of a mathematical model for NF-κB oscillations, described by a set of ordinary nonlinear differential equations, when perturbed by a circadian oscillation. The main result is that a circadian rhythm, even when it represents a weak perturbation, enhances the signaling capabilities of NF-κB oscillations. This is done by turning rest states into periodic oscillations, and periodic oscillations into quasiperiodic oscillations. Strong perturbations result in complex periodic oscillations and even in chaos. Circadian rhythms would then result in a NF-κB dynamics that is more complex than the simple oscillations and rest states, initially reported for this model. This renders it more amenable for information coding. link: http://identifiers.org/pubmed/23820037

Parameters:

NameDescription
A = 0.007; epsilon = 2.0E-5Reaction: => x; z, Rate Law: compartment*A*(1-x)/(epsilon+z)
k1=1.0Reaction: y =>, Rate Law: compartment*k1*y
epsilon = 2.0E-5; C = 0.035Reaction: z => ; x, Rate Law: compartment*C*z*(1-x)/(epsilon+z)
delta = 0.029; B = 954.5Reaction: x => ; z, Rate Law: compartment*B*z*x/(delta+x)

States:

NameDescription
x[NF-kB]
z[C104199]
y[C104199]

González-Domenech2012_MetabolicNetwork_iCG230: MODEL1110130001v0.0.1

This model is from the article: Metabolic stasis in an ancient symbiosis: genome-scale metabolic networks from two Bla…

Details

BACKGROUND: Cockroaches are terrestrial insects that strikingly eliminate waste nitrogen as ammonia instead of uric acid. Blattabacterium cuenoti (Mercier 1906) strains Bge and Pam are the obligate primary endosymbionts of the cockroaches Blattella germanica and Periplaneta americana, respectively. The genomes of both bacterial endosymbionts have recently been sequenced, making possible a genome-scale constraint-based reconstruction of their metabolic networks. The mathematical expression of a metabolic network and the subsequent quantitative studies of phenotypic features by Flux Balance Analysis (FBA) represent an efficient functional approach to these uncultivable bacteria. RESULTS: We report the metabolic models of Blattabacterium strains Bge (iCG238) and Pam (iCG230), comprising 296 and 289 biochemical reactions, associated with 238 and 230 genes, and 364 and 358 metabolites, respectively. Both models reflect both the striking similarities and the singularities of these microorganisms. FBA was used to analyze the properties, potential and limits of the models, assuming some environmental constraints such as aerobic conditions and the net production of ammonia from these bacterial systems, as has been experimentally observed. In addition, in silico simulations with the iCG238 model have enabled a set of carbon and nitrogen sources to be defined, which would also support a viable phenotype in terms of biomass production in the strain Pam, which lacks the first three steps of the tricarboxylic acid cycle. FBA reveals a metabolic condition that renders these enzymatic steps dispensable, thus offering a possible evolutionary explanation for their elimination. We also confirm, by computational simulations, the fragility of the metabolic networks and their host dependence. CONCLUSIONS: The minimized Blattabacterium metabolic networks are surprisingly similar in strains Bge and Pam, after 140 million years of evolution of these endosymbionts in separate cockroach lineages. FBA performed on the reconstructed networks from the two bacteria helps to refine the functional analysis of the genomes enabling us to postulate how slightly different host metabolic contexts drove their parallel evolution. link: http://identifiers.org/pubmed/22376077

González-Domenech2012_MetabolicNetwork_iCG238: MODEL1110130000v0.0.1

This model is from the article: Metabolic stasis in an ancient symbiosis: genome-scale metabolic networks from two Bla…

Details

BACKGROUND: Cockroaches are terrestrial insects that strikingly eliminate waste nitrogen as ammonia instead of uric acid. Blattabacterium cuenoti (Mercier 1906) strains Bge and Pam are the obligate primary endosymbionts of the cockroaches Blattella germanica and Periplaneta americana, respectively. The genomes of both bacterial endosymbionts have recently been sequenced, making possible a genome-scale constraint-based reconstruction of their metabolic networks. The mathematical expression of a metabolic network and the subsequent quantitative studies of phenotypic features by Flux Balance Analysis (FBA) represent an efficient functional approach to these uncultivable bacteria. RESULTS: We report the metabolic models of Blattabacterium strains Bge (iCG238) and Pam (iCG230), comprising 296 and 289 biochemical reactions, associated with 238 and 230 genes, and 364 and 358 metabolites, respectively. Both models reflect both the striking similarities and the singularities of these microorganisms. FBA was used to analyze the properties, potential and limits of the models, assuming some environmental constraints such as aerobic conditions and the net production of ammonia from these bacterial systems, as has been experimentally observed. In addition, in silico simulations with the iCG238 model have enabled a set of carbon and nitrogen sources to be defined, which would also support a viable phenotype in terms of biomass production in the strain Pam, which lacks the first three steps of the tricarboxylic acid cycle. FBA reveals a metabolic condition that renders these enzymatic steps dispensable, thus offering a possible evolutionary explanation for their elimination. We also confirm, by computational simulations, the fragility of the metabolic networks and their host dependence. CONCLUSIONS: The minimized Blattabacterium metabolic networks are surprisingly similar in strains Bge and Pam, after 140 million years of evolution of these endosymbionts in separate cockroach lineages. FBA performed on the reconstructed networks from the two bacteria helps to refine the functional analysis of the genomes enabling us to postulate how slightly different host metabolic contexts drove their parallel evolution. link: http://identifiers.org/pubmed/22376077