SBMLBioModels: G - H

G


Gould2013 - Temperature Sensitive Circadian Clock: BIOMD0000000564v0.0.1

Gould2011 - Temperature Sensitive Circadian ClockThis model is a temperature sensitive version of Pokhilko *et al*.  20…

Details

Circadian clocks exhibit 'temperature compensation', meaning that they show only small changes in period over a broad temperature range. Several clock genes have been implicated in the temperature-dependent control of period in Arabidopsis. We show that blue light is essential for this, suggesting that the effects of light and temperature interact or converge upon common targets in the circadian clock. Our data demonstrate that two cryptochrome photoreceptors differentially control circadian period and sustain rhythmicity across the physiological temperature range. In order to test the hypothesis that the targets of light regulation are sufficient to mediate temperature compensation, we constructed a temperature-compensated clock model by adding passive temperature effects into only the light-sensitive processes in the model. Remarkably, this model was not only capable of full temperature compensation and consistent with mRNA profiles across a temperature range, but also predicted the temperature-dependent change in the level of LATE ELONGATED HYPOCOTYL, a key clock protein. Our analysis provides a systems-level understanding of period control in the plant circadian oscillator. link: http://identifiers.org/pubmed/23511208

Parameters:

NameDescription
p10 = 0.36Reaction: => cNI; cNI_m, cNI_m, Rate Law: def*p10*cNI_m/def
m10 = 0.3Reaction: cY => ; cY, Rate Law: def*m10*cY/def
g3 = 0.4; m3 = 0.2; p3 = 0.1; c = 3.0Reaction: cL => ; cL, Rate Law: def*(m3*cL+p3*cL^c/(cL^c+g3^c))/def
m13 = 0.32; m22 = 2.0; D = 0.5; L = 0.5Reaction: cP9 => ; cP9, Rate Law: def*(m13*L+m22*D)*cP9/def
m25 = 0.280006253789297; D = 0.5; m26 = 0.14; L = 0.5Reaction: cTm => ; cTm, Rate Law: def*(m25*L+m26*D)*cTm/def
m7 = 0.5; D = 0.5; p5 = 1.0; m8 = 0.1; m6 = 0.250006065831407; L = 0.5Reaction: cT => ; cZG, cZTL, cT, cZG, cZTL, Rate Law: def*((m6*L+m7*D)*cT*(p5*cZTL+cZG)+m8*cT)/def
n8 = 0.42; g11 = 0.7; j = 3.0; n9 = 0.26; k = 3.0; g10 = 0.7Reaction: => cP7_m; cL, cLm, cP9, cL, cLm, cP9, Rate Law: def*(n8*(cLm+cL)^j/((cLm+cL)^j+g10^j)+n9*cP9^k/(cP9^k+g11^k))/def
m12 = 1.0Reaction: cP9_m => ; cP9_m, Rate Law: def*m12*cP9_m/def
p14 = 0.45Reaction: => cZTL, Rate Law: def*p14/def
g3 = 0.4; p3 = 0.1; c = 3.0Reaction: => cLm; cL, cL, Rate Law: def*p3*cL^c/(cL^c+g3^c)/def
m14 = 0.28Reaction: cP7_m => ; cP7_m, Rate Law: def*m14*cP7_m/def
n0 = 0.400002792587441; n1 = 1.8; g2 = 0.28; g1 = 0.1; q1 = 0.8; a = 2.0; b = 3.0; L = 0.5Reaction: => cL_m; cNI, cP, cP7, cP9, cTm, cNI, cP, cP7, cP9, cTm, Rate Law: def*(n0*L+L*q1*cP+n1*cTm^b/(cTm^b+g2^b))*g1^a/((cP9+cP7+cNI)^a+g1^a)/def
g9 = 0.3; n7 = 0.2; i = 3.0; g8 = 0.14; q3 = 2.9; h = 2.0; n4 = 0.0; L = 0.5Reaction: => cP9_m; cL, cP, cT, cL, cP, cT, Rate Law: def*(L*q3*cP+(n4*L+n7*cL^i/(cL^i+g9^i))*g8^h/(cT^h+g8^h))/def
m4 = 0.2Reaction: cLm => ; cLm, Rate Law: def*m4*cLm/def
p4 = 0.268Reaction: => cT; cT_m, cT_m, Rate Law: def*p4*cT_m/def
m18 = 1.0Reaction: cG_m => ; cG_m, Rate Law: def*m18*cG_m/def
p6 = 0.44Reaction: => cY; cY_m, cY_m, Rate Law: def*p6*cY_m/def
p8 = 0.7Reaction: => cP9; cP9_m, cP9_m, Rate Law: def*p8*cP9_m/def
m20 = 1.2Reaction: cZTL => ; cZTL, Rate Law: def*m20*cZTL/def
g16 = 0.2; n5 = 3.4; D = 0.5; q2 = 0.5; s = 3.0; g7 = 0.18; n6 = 1.25; L = 0.5; g = 2.0Reaction: => cY_m; cL, cP, cT, cL, cP, cT, Rate Law: def*(L*q2*cP+(n5*L+n6*D)*g7^s/(cT^s+g7^s)*g16^g/(cL^g+g16^g))/def
m1 = 0.54001322056037; D = 0.5; m2 = 0.24; L = 0.5Reaction: cL_m => ; cL_m, Rate Law: def*(m1*L+m2*D)*cL_m/def
g14 = 0.17; g15 = 0.4; n = 1.0; q4 = 0.6; o = 2.0; L = 0.5; n12 = 2.3499889284595Reaction: => cG_m; cL, cP, cT, cL, cP, cT, Rate Law: def*(L*q4*cP+n12*L*g15^o/(cL^o+g15^o)*g14^n/(cT^n+g14^n))/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
m21 = 0.2Reaction: cZG => ; cZG, Rate Law: def*m21*cZG/def
m24 = 0.405; D = 0.5; m17 = 0.3; L = 0.5Reaction: cNI => ; cNI, Rate Law: def*(m17*L+m24*D)*cNI/def
D = 0.5; m15 = 0.310006139862489; L = 0.5; m23 = 1.0Reaction: cP7 => ; cP7, Rate Law: def*(m15*L+m23*D)*cP7/def
g4 = 0.91; n2 = 0.7; g5 = 0.3; e = 2.0; n3 = 0.06; d = 2.5Reaction: => cT_m; cL, cY, cL, cY, Rate Law: def*(n2*cY^d/(cY^d+g4^d)+n3)*g5^e/(cL^e+g5^e)/def
p9 = 0.4Reaction: => cP7; cP7_m, cP7_m, Rate Law: def*p9*cP7_m/def
D = 0.5; p13 = 0.4; p12 = 30.0; L = 0.5Reaction: cG + cZTL => cZG; cG, cZG, cZTL, Rate Law: def*(p12*L*cZTL*cG-p13*D*cZG)/def
m5 = 0.3Reaction: cT_m => ; cT_m, Rate Law: def*m5*cT_m/def
m9 = 1.0Reaction: cY_m => ; cY_m, Rate Law: def*m9*cY_m/def
D = 0.5; p7 = 0.3Reaction: => cP; cP, Rate Law: def*p7*D*(1-cP)/def
p15 = 0.05; f = 3.0; g6 = 0.3Reaction: => cTm; cT, cT, Rate Law: def*p15*cT^f/(cT^f+g6^f)/def
p11 = 0.23Reaction: => cG; cG_m, cG_m, Rate Law: def*p11*cG_m/def
D = 0.5; p2 = 0.27; p1 = 0.400007560981732; L = 0.5Reaction: => cL; cL_m, cL_m, Rate Law: def*cL_m*(p1*L+p2*D)/def
m19 = 0.2Reaction: cG => ; cG, Rate Law: def*m19*cG/def
n10 = 0.18; g12 = 0.5; m = 2.0; n11 = 0.71; g13 = 0.6; l = 2.0Reaction: => cNI_m; cLm, cP7, cLm, cP7, Rate Law: def*(n10*cLm^l/(cLm^l+g12^l)+n11*cP7^m/(cP7^m+g13^m))/def

States:

NameDescription
cL m[messenger RNA]
cNI[inhibitor]
cG[Protein GIGANTEA]
cP9[Two-component response regulator-like APRR9]
cP9 m[messenger RNA]
cZTL[Adagio protein 1]
cP7 m[messenger RNA]
cNI m[inhibitor; messenger RNA]
cG m[messenger RNA]
cY[protein]
cT m[messenger RNA]
cY m[messenger RNA; RNA]
cPcP
cLm[Protein CCA1; protein modification; Protein LHY]
cP7[Two-component response regulator-like APRR7]
cT[Two-component response regulator-like APRR1]
cZG[Protein GIGANTEA; Adagio protein 1]
cTm[protein modification; Two-component response regulator-like APRR1]
cL[Protein CCA1; Protein LHY]

Goyal2014 - Genome-scale metabolic model of M.maripaludis S2: MODEL1304120000v0.0.1

Constraint-based genome-scale Metabolic Model of Methanococcus maripaludis S2

Details

Methane is a major energy source for heating and electricity. Its production by methanogenic bacteria is widely known in nature. M. maripaludis S2 is a fully sequenced hydrogenotrophic methanogen and an excellent laboratory strain with robust genetic tools. However, a quantitative systems biology model to complement these tools is absent in the literature. To understand and enhance its methanogenesis from CO2, this work presents the first constraint-based genome-scale metabolic model (iMM518). It comprises 570 reactions, 556 distinct metabolites, and 518 genes along with gene-protein-reaction (GPR) associations, and covers 30% of open reading frames (ORFs). The model was validated using biomass growth data and experimental phenotypic studies from the literature. Its comparison with the in silico models of Methanosarcina barkeri, Methanosarcina acetivorans, and Sulfolobus solfataricus P2 shows M. maripaludis S2 to be a better organism for producing methane. Using the model, genes essential for growth were identified, and the efficacies of alternative carbon, hydrogen and nitrogen sources were studied. The model can predict the effects of reengineering M. maripaludis S2 to guide or expedite experimental efforts. link: http://identifiers.org/pubmed/24553424

Grandi2009_VentricularMyocyte: MODEL1006230092v0.0.1

This a model from the article: A novel computational model of the human ventricular action potential and Ca transient.…

Details

We have developed a detailed mathematical model for Ca handling and ionic currents in the human ventricular myocyte. Our aims were to: (1) simulate basic excitation-contraction coupling phenomena; (2) use realistic repolarizing K current densities; (3) reach steady-state. The model relies on the framework of the rabbit myocyte model previously developed by our group, with subsarcolemmal and junctional compartments where ion channels sense higher [Ca] vs. bulk cytosol. Ion channels and transporters have been modeled on the basis of the most recent experimental data from human ventricular myocytes. Rapidly and slowly inactivating components of I(to) have been formulated to differentiate between endocardial and epicardial myocytes. Transmural gradients of Ca handling proteins and Na pump were also simulated. The model has been validated against a wide set of experimental data including action potential duration (APD) adaptation and restitution, frequency-dependent increase in Ca transient peak and Na. Interestingly, Na accumulation at fast heart rate is a major determinant of APD shortening, via outward shifts in Na pump and Na-Ca exchange currents. We investigated the effects of blocking K currents on APD and repolarization reserve: I(Ks) block does not affect the former and slightly reduces the latter; I(K1) blockade modestly increases APD and more strongly reduces repolarization reserve; I(Kr) blockers significantly prolong APD, an effect exacerbated as pacing frequency is decreased, in good agreement with experimental results in human myocytes. We conclude that this model provides a useful framework to explore excitation-contraction coupling mechanisms and repolarization abnormalities at the single myocyte level. link: http://identifiers.org/pubmed/19835882

Grange2001 - L Dopa PK model: BIOMD0000000321v0.0.1

Grange2001 - L-dopa PK modelA pharmacokinetics of L-dopa in rats after administration of L-dopa alone (this model: BIOMD…

Details

PURPOSE: To study the PK interaction of L-dopa/benserazide in rats. METHODS: Male rats received a single oral dose of 80 mg/kg L-dopa or 20 mg/kg benserazide or 80/20 mg/kg L-dopa/benserazide. Based on plasma concentrations the kinetics of L-dopa, 3-O-methyldopa (3-OMD), benserazide, and its metabolite Ro 04-5127 were characterized by noncompartmental analysis and a compartmental model where total L-dopa clearance was the sum of the clearances mediated by amino-acid-decarboxylase (AADC), catechol-O-methyltransferase and other enzymes. In the model Ro 04-5127 inhibited competitively the L-dopa clearance by AADC. RESULTS: The coadministration of L-dopa/benserazide resulted in a major increase in systemic exposure to L-dopa and 3-OMD and a decrease in L-dopa clearance. The compartmental model allowed an adequate description of the observed L-dopa and 3-OMD concentrations in the absence and presence of benserazide. It had an advantage over noncompartmental analysis because it could describe the temporal change of inhibition and recovery of AADC. CONCLUSIONS: Our study is the first investigation where the kinetics of benserazide and Ro 04-5127 have been described by a compartmental model. The L-dopa/benserazide model allowed a mechanism-based view of the L-dopa/benserazide interaction and supports the hypothesis that Ro 04-5127 is the primary active metabolite of benserazide. link: http://identifiers.org/pubmed/11587490

Parameters:

NameDescription
CL_rest = NaN l_per_hReaction: C_dopa =>, Rate Law: CL_rest*C_dopa
F_b = NaN dimensionless; ka_b = 2.11 per_hReaction: A_dopa => C_dopa, Rate Law: ka_b*A_dopa*F_b
CL_COMT = NaN l_per_hReaction: C_dopa => C_OMD, Rate Law: CL_COMT*C_dopa
CL_AADC = NaN l_per_hReaction: C_dopa =>, Rate Law: CL_AADC*C_dopa
CL_OMD = 0.012Reaction: C_OMD =>, Rate Law: CL_OMD*C_OMD

States:

NameDescription
C OMD[metabolite; 9307]
C dopa[L-dopa; 3,4-Dihydroxy-L-phenylalanine; 6047]
A dopa[L-dopa; 6047; 3,4-Dihydroxy-L-phenylalanine]

Grange2001 - PK interaction of L-dopa and benserazide: BIOMD0000000320v0.0.1

Grange2001 - PK interaction of L-dopa and benserazideA pharmacokinetics of L-dopa in rats after administration of L-dopa…

Details

PURPOSE: To study the PK interaction of L-dopa/benserazide in rats. METHODS: Male rats received a single oral dose of 80 mg/kg L-dopa or 20 mg/kg benserazide or 80/20 mg/kg L-dopa/benserazide. Based on plasma concentrations the kinetics of L-dopa, 3-O-methyldopa (3-OMD), benserazide, and its metabolite Ro 04-5127 were characterized by noncompartmental analysis and a compartmental model where total L-dopa clearance was the sum of the clearances mediated by amino-acid-decarboxylase (AADC), catechol-O-methyltransferase and other enzymes. In the model Ro 04-5127 inhibited competitively the L-dopa clearance by AADC. RESULTS: The coadministration of L-dopa/benserazide resulted in a major increase in systemic exposure to L-dopa and 3-OMD and a decrease in L-dopa clearance. The compartmental model allowed an adequate description of the observed L-dopa and 3-OMD concentrations in the absence and presence of benserazide. It had an advantage over noncompartmental analysis because it could describe the temporal change of inhibition and recovery of AADC. CONCLUSIONS: Our study is the first investigation where the kinetics of benserazide and Ro 04-5127 have been described by a compartmental model. The L-dopa/benserazide model allowed a mechanism-based view of the L-dopa/benserazide interaction and supports the hypothesis that Ro 04-5127 is the primary active metabolite of benserazide. link: http://identifiers.org/pubmed/11587490

Parameters:

NameDescription
CL_M = 4.29 l_per_hReaction: C1_M =>, Rate Law: CL_M*C1_M
CL_rest = NaN l_per_hReaction: C_dopa =>, Rate Law: CL_rest*C_dopa
CL_dM = 1.06 l_per_hReaction: C1_M => C2_M, Rate Law: CL_dM*(C1_M-C2_M)
fm = 0.15 dimensionless; CL_B = 1.67 l_per_hReaction: C1_B => C1_M, Rate Law: fm/(1-fm)*CL_B*C1_B
CL_COMT = NaN l_per_hReaction: C_dopa => C_OMD, Rate Law: CL_COMT*C_dopa
ka_c = 1.29; F_c = NaN dimensionlessReaction: A_dopa => C_dopa, Rate Law: ka_c*A_dopa*F_c
ka_M = 2.47Reaction: A_M => C1_M, Rate Law: ka_M*A_M
CL_OMD = 0.00895 l_per_hReaction: C_OMD =>, Rate Law: CL_OMD*C_OMD
ka_B = 0.94; F_B = 0.022 dimensionlessReaction: A_B =>, Rate Law: ka_B*A_B*(1-F_B)
CL_B = 1.67 l_per_hReaction: C1_B =>, Rate Law: CL_B*C1_B
CL_AADC = NaN l_per_hReaction: C_dopa =>, Rate Law: CL_AADC*C_dopa
CL_dB = 0.072 l_per_hReaction: C1_B => C2_B, Rate Law: CL_dB*(C1_B-C2_B)

States:

NameDescription
C1 M[metabolite; 188973]
C OMD[9307; metabolite]
C2 B[Benserazide (USAN/INN); 2327]
A B[Benserazide (USAN/INN); 2327]
C1 B[Benserazide (USAN/INN); 2327]
C dopa[L-dopa; 3,4-Dihydroxy-L-phenylalanine; 6047]
A dopa[L-dopa; 3,4-Dihydroxy-L-phenylalanine; 6047]
A M[metabolite; 188973]
C2 M[metabolite; 188973]

Gray2016 - The Akt switch model: BIOMD0000000854v0.0.1

This is a simple, linear, four-compartment ordinary differential equation (ODE) model Akt activation that tracks both th…

Details

Akt/PKB is a biochemical regulator that functions as an important cross-talk node between several signalling pathways in the mammalian cell. In particular, Akt is a key mediator of glucose transport in response to insulin. The phosphorylation (activation) of only a small percentage of the Akt pool of insulin-sensitive cells results in maximal translocation of glucose transporter 4 (GLUT4) to the plasma membrane (PM). This enables the diffusion of glucose into the cell. The dysregulation of Akt signalling is associated with the development of diabetes, cancer and cardiovascular disease. Akt is synthesised in the cytoplasm in the inactive state. Under the influence of insulin, it moves to the PM, where it is phosphorylated to form pAkt. Although phosphorylation occurs only at the PM, pAkt is found in many cellular locations, including the PM, the cytoplasm, and the nucleus. Indeed, the spatial distribution of pAkt within the cell appears to be an important determinant of downstream regulation. Here we present a simple, linear, four-compartment ordinary differential equation (ODE) model of Akt activation that tracks both the biochemical state and the physical location of Akt. This model embodies the main features of the activation of this important cross-talk node and is consistent with the experimental data. In particular, it allows different downstream signalling motifs without invoking separate feedback pathways. Moreover, the model is computationally tractable, readily analysed, and elucidates some of the apparent anomalies in insulin signalling via Akt. link: http://identifiers.org/pubmed/26992575

Parameters:

NameDescription
beta1 = 2.2; koff = 0.35Reaction: Ap => Pp, Rate Law: compartment*beta1*koff*Ap
kin = 0.55Reaction: Pp => Pc, Rate Law: compartment*kin*Pp
koff = 0.35Reaction: Pc => Ac, Rate Law: compartment*koff*Pc
alpha1 = 0.014; kin = 0.55Reaction: Pc => Pp, Rate Law: compartment*alpha1*kin*Pc

States:

NameDescription
Ap[AKT kinase; plasma membrane]
Pp[AKT kinase; phosphoprotein; plasma membrane]
Ac[AKT kinase; cytosol]
Pc[AKT kinase; phosphoprotein; cytosol]

Greene2019 - Differentiate Spontaneous and Induced Evolution to Drug Resistance During Cancer Treatment: BIOMD0000000825v0.0.1

This model is built by COPASI 4.24(Build 197), based on paper: Mathematical Approach to Differentiate Spontaneous and I…

Details

PURPOSE:Drug resistance is a major impediment to the success of cancer treatment. Resistance is typically thought to arise from random genetic mutations, after which mutated cells expand via Darwinian selection. However, recent experimental evidence suggests that progression to drug resistance need not occur randomly, but instead may be induced by the treatment itself via either genetic changes or epigenetic alterations. This relatively novel notion of resistance complicates the already challenging task of designing effective treatment protocols. MATERIALS AND METHODS:To better understand resistance, we have developed a mathematical modeling framework that incorporates both spontaneous and drug-induced resistance. RESULTS:Our model demonstrates that the ability of a drug to induce resistance can result in qualitatively different responses to the same drug dose and delivery schedule. We have also proven that the induction parameter in our model is theoretically identifiable and propose an in vitro protocol that could be used to determine a treatment's propensity to induce resistance. link: http://identifiers.org/pubmed/30969799

Parameters:

NameDescription
alpha = 0.01; epsilon = 1.0E-6; u = 0.0Reaction: Sensitive_tumor_S => Resistant_tumor_R, Rate Law: compartment*(epsilon+alpha*u)*Sensitive_tumor_S
p_r = 0.2Reaction: => Resistant_tumor_R; Sensitive_tumor_S, Rate Law: compartment*p_r*(1-(Sensitive_tumor_S+Resistant_tumor_R))*Resistant_tumor_R
d = 1.0; u = 0.0Reaction: Sensitive_tumor_S =>, Rate Law: compartment*d*u*Sensitive_tumor_S

States:

NameDescription
Resistant tumor R[resistant to; cancer]
Tumor Volume V[cancer; tumor size; Tumor Size]
Sensitive tumor S[cancer; 0000516]

Guisoni2016 - Cis-regulatory system (CRS) can drive sustained oscillations: MODEL1611030000v0.0.1

Guisoni2016 - Cis-regulatory system (CRS) can drive sustained oscillationsThis model is described in the article: [Prom…

Details

It is well known that single-gene circuits with negative feedback loop can lead to oscillatory gene expression when they operate with time delay. In order to generate these oscillations many processes can contribute to properly timing such delay. Here we show that the time delay coming from the transitions between internal states of the cis-regulatory system (CRS) can drive sustained oscillations in an auto-repressive single-gene circuit operating in a small volume like a cell. We found that the cooperative binding of repressor molecules is not mandatory for a oscillatory behavior if there are enough binding sites in the CRS. These oscillations depend on an adequate balance between the CRS kinetic, and the synthesis/degradation rates of repressor molecules. This finding suggest that the multi-site CRS architecture can play a key role for oscillatory behavior of gene expression. Finally, our results can also help to synthetic biologists on the design of the promoters architecture for new genetic oscillatory circuits. link: http://identifiers.org/pubmed/26958852

Gulati2014 - Simplified model of fibrinogen recovery following brown snake bite_1: MODEL1805090001v0.0.1

Bridging systems biology and pharmacokinetics–pharmacodynamics has resulted in models that are highly complex and compli…

Details

Bridging systems biology and pharmacokinetics-pharmacodynamics has resulted in models that are highly complex and complicated. They usually contain large numbers of states and parameters and describe multiple input-output relationships. Based on any given data set relating to a specific input-output process, it is possible that some states of the system are either less important or have no influence at all. In this study, we explore a simplification of a systems pharmacology model of the coagulation network for use in describing the time course of fibrinogen recovery after a brown snake bite. The technique of proper lumping is used to simplify the 62-state systems model to a 5-state model that describes the brown snake venom-fibrinogen relationship while maintaining an appropriate mechanistic relationship. The simplified 5-state model explains the observed decline and recovery in fibrinogen concentrations well. The techniques used in this study can be applied to other multiscale models. link: http://identifiers.org/pubmed/24402117

Gulbudak2019 - Heterogeneous viral strategies promote coexistence in virus-microbe systems: BIOMD0000000845v0.0.1

This is a mathematical model describing describing the population dynamics of microbes infected by lytic viruses.

Details

Viral infections of microbial cells often culminate in lysis and the release of new virus particles. However, viruses of microbes can also initiate chronic infections, in which new viruses particles are released via budding and without cell lysis. In chronic infections, viral genomes may also be passed on from mother to daughter cells during division. The consequences of chronic infections for the population dynamics of viruses and microbes remains under-explored. In this paper we present a model of chronic infections as well as a model of interactions between lytic and chronic viruses competing for the same microbial population. In the chronic only model, we identify conditions underlying complex bifurcations such as saddle-node, backward and Hopfbifurcations, leading to parameter regions with multiple attractors and/or oscillatory behavior. We then utilize invasion analysis to examine the coupled nonlinear system of microbes, lytic viruses, and chronic viruses. In so doing we find unexpected results, including a regime in which the chronic virus requires the lytic virus for survival, invasion, and persistence. In this regime, lytic viruses decrease total cell densities, so that a subpopulation of chronically infected cells experience decreased niche competition. As such, even when chronically infected cells have a growth disadvantage, lytic viruses can, paradoxically, enable the proliferation of both chronically infected cells and chronic viruses. We discuss the implications of our results for understanding the ecology and long-term evolution of heterogeneous viral strategies. link: http://identifiers.org/pubmed/30389532

Parameters:

NameDescription
eta = 1.5Reaction: I =>, Rate Law: compartment*eta*I
phi = 1.73925271309261E-10Reaction: S + V_L => I, Rate Law: compartment*phi*S*V_L
mu = 0.0866Reaction: V_L =>, Rate Law: compartment*mu*V_L
r = 0.339; N = 8.3E8; K = 8.947E8Reaction: => S, Rate Law: compartment*r*S*(1-N/K)
d = 0.0416666666666667Reaction: I =>, Rate Law: compartment*d*I
beta = 20.0; eta = 1.5Reaction: => V_L; I, Rate Law: compartment*beta*eta*I

States:

NameDescription
I[infected cell]
S[C14187]
V L[C14368]

Gulbudak2019.2 - Heterogeneous viral strategies promote coexistence in virus-microbe systems (Chronic): BIOMD0000000846v0.0.1

This is a mathematical model describing describing the population dynamics of microbes infected by chronically infecting…

Details

Viral infections of microbial cells often culminate in lysis and the release of new virus particles. However, viruses of microbes can also initiate chronic infections, in which new viruses particles are released via budding and without cell lysis. In chronic infections, viral genomes may also be passed on from mother to daughter cells during division. The consequences of chronic infections for the population dynamics of viruses and microbes remains under-explored. In this paper we present a model of chronic infections as well as a model of interactions between lytic and chronic viruses competing for the same microbial population. In the chronic only model, we identify conditions underlying complex bifurcations such as saddle-node, backward and Hopfbifurcations, leading to parameter regions with multiple attractors and/or oscillatory behavior. We then utilize invasion analysis to examine the coupled nonlinear system of microbes, lytic viruses, and chronic viruses. In so doing we find unexpected results, including a regime in which the chronic virus requires the lytic virus for survival, invasion, and persistence. In this regime, lytic viruses decrease total cell densities, so that a subpopulation of chronically infected cells experience decreased niche competition. As such, even when chronically infected cells have a growth disadvantage, lytic viruses can, paradoxically, enable the proliferation of both chronically infected cells and chronic viruses. We discuss the implications of our results for understanding the ecology and long-term evolution of heterogeneous viral strategies. link: http://identifiers.org/pubmed/30389532

Parameters:

NameDescription
r_tilde = 0.2; N = 8.3E8; K = 8.947E8Reaction: => C, Rate Law: compartment*r_tilde*C*(1-N/K)
alpha = 0.1Reaction: => V_C; C, Rate Law: compartment*alpha*C
mu = 0.0866Reaction: V_C =>, Rate Law: compartment*mu*V_C
r = 0.339; N = 8.3E8; K = 8.947E8Reaction: => S, Rate Law: compartment*r*S*(1-N/K)
d = 0.0416666666666667Reaction: S =>, Rate Law: compartment*d*S
d_tilde = 0.05Reaction: C =>, Rate Law: compartment*d_tilde*C
phi_tilde = 5.0E-12Reaction: S + V_C => C, Rate Law: compartment*phi_tilde*S*V_C

States:

NameDescription
V C[C14368]
S[C14187]
C[C14283; C14141]

Gupta2007_HypothalamicPituitaryAdrenal_ModelA: MODEL1006230111v0.0.1

This a model from the article: Inclusion of the glucocorticoid receptor in a hypothalamic pituitary adrenal axis model…

Details

The body's primary stress management system is the hypothalamic pituitary adrenal (HPA) axis. The HPA axis responds to physical and mental challenge to maintain homeostasis in part by controlling the body's cortisol level. Dysregulation of the HPA axis is implicated in numerous stress-related diseases.We developed a structured model of the HPA axis that includes the glucocorticoid receptor (GR). This model incorporates nonlinear kinetics of pituitary GR synthesis. The nonlinear effect arises from the fact that GR homodimerizes after cortisol activation and induces its own synthesis in the pituitary. This homodimerization makes possible two stable steady states (low and high) and one unstable state of cortisol production resulting in bistability of the HPA axis. In this model, low GR concentration represents the normal steady state, and high GR concentration represents a dysregulated steady state. A short stress in the normal steady state produces a small perturbation in the GR concentration that quickly returns to normal levels. Long, repeated stress produces persistent and high GR concentration that does not return to baseline forcing the HPA axis to an alternate steady state. One consequence of increased steady state GR is reduced steady state cortisol, which has been observed in some stress related disorders such as Chronic Fatigue Syndrome (CFS).Inclusion of pituitary GR expression resulted in a biologically plausible model of HPA axis bistability and hypocortisolism. High GR concentration enhanced cortisol negative feedback on the hypothalamus and forced the HPA axis into an alternative, low cortisol state. This model can be used to explore mechanisms underlying disorders of the HPA axis. link: http://identifiers.org/pubmed/17300722

Gupta2007_HypothalamicPituitaryAdrenal_ModelB: MODEL1006230036v0.0.1

This a model from the article: Inclusion of the glucocorticoid receptor in a hypothalamic pituitary adrenal axis model…

Details

The body's primary stress management system is the hypothalamic pituitary adrenal (HPA) axis. The HPA axis responds to physical and mental challenge to maintain homeostasis in part by controlling the body's cortisol level. Dysregulation of the HPA axis is implicated in numerous stress-related diseases.We developed a structured model of the HPA axis that includes the glucocorticoid receptor (GR). This model incorporates nonlinear kinetics of pituitary GR synthesis. The nonlinear effect arises from the fact that GR homodimerizes after cortisol activation and induces its own synthesis in the pituitary. This homodimerization makes possible two stable steady states (low and high) and one unstable state of cortisol production resulting in bistability of the HPA axis. In this model, low GR concentration represents the normal steady state, and high GR concentration represents a dysregulated steady state. A short stress in the normal steady state produces a small perturbation in the GR concentration that quickly returns to normal levels. Long, repeated stress produces persistent and high GR concentration that does not return to baseline forcing the HPA axis to an alternate steady state. One consequence of increased steady state GR is reduced steady state cortisol, which has been observed in some stress related disorders such as Chronic Fatigue Syndrome (CFS).Inclusion of pituitary GR expression resulted in a biologically plausible model of HPA axis bistability and hypocortisolism. High GR concentration enhanced cortisol negative feedback on the hypothalamus and forced the HPA axis into an alternative, low cortisol state. This model can be used to explore mechanisms underlying disorders of the HPA axis. link: http://identifiers.org/pubmed/17300722

Gupta2009 - Eicosanoid Metabolism: BIOMD0000000436v0.0.1

Gupta2009 - Eicosanoid MetabolismIntegrated model of eicosanoid metabolism and signaling based on lipidomics flux analys…

Details

There is increasing evidence for a major and critical involvement of lipids in signal transduction and cellular trafficking, and this has motivated large-scale studies on lipid pathways. The Lipid Metabolites and Pathways Strategy consortium is actively investigating lipid metabolism in mammalian cells and has made available time-course data on various lipids in response to treatment with KDO(2)-lipid A (a lipopolysaccharide analog) of macrophage RAW 264.7 cells. The lipids known as eicosanoids play an important role in inflammation. We have reconstructed an integrated network of eicosanoid metabolism and signaling based on the KEGG pathway database and the literature and have developed a kinetic model. A matrix-based approach was used to estimate the rate constants from experimental data and these were further refined using generalized constrained nonlinear optimization. The resulting model fits the experimental data well for all species, and simulated enzyme activities were similar to their literature values. The quantitative model for eicosanoid metabolism that we have developed can be used to design experimental studies utilizing genetic and pharmacological perturbations to probe fluxes in lipid pathways. link: http://identifiers.org/pubmed/19486676

Parameters:

NameDescription
k9 = 0.187 (0.0002778*s)^(-1)Reaction: HETE =>, Rate Law: k9*HETE
k21 = 0.034 (0.0002778*s)^(-1)Reaction: PGJ2 => dPGJ2, Rate Law: k21*PGJ2
DGactivity = 1.0 dimensionless; DNA = 1.0 μg; k10 = 0.024 μg*(3600*s)^(-1)Reaction: AA => PGH2; DG, Rate Law: k10*DGactivity*AA/DNA
k16 = 1.0E-15 (0.0002778*s)^(-1)Reaction: PGF2a =>, Rate Law: k16*PGF2a
k8 = 0.007 (0.0002778*s)^(-1)Reaction: AA => HETE, Rate Law: k8*AA
k20 = 0.014 (0.0002778*s)^(-1)Reaction: dPGD2 =>, Rate Law: k20*dPGD2
DGactivity = 1.0 dimensionless; GPChoratio = 1.0 dimensionless; DNA = 1.0 μg; k5 = 1.0E-15 μg*(3600*s)^(-1)Reaction: GPCho => AA; DG, Rate Law: k5*DGactivity*GPChoratio*GPCho/DNA
k17 = 3.116 (0.0002778*s)^(-1)Reaction: PGH2 => PGD2, Rate Law: k17*PGH2
LPSactivity = 0.0 dimensionless; GPChoratio = 1.0 dimensionless; k6 = 0.33 (0.0002778*s)^(-1)Reaction: GPCho => AA; LPS, Rate Law: k6*GPCho*GPChoratio*LPSactivity
k22 = 0.116 (0.0002778*s)^(-1)Reaction: dPGJ2 =>, Rate Law: k22*dPGJ2
k2 = 1.0E-15 (0.0002778*s)^(-1)Reaction: FA => AA, Rate Law: k2*FA
k14 = 1.0E-15 (0.0002778*s)^(-1)Reaction: PGE2 =>, Rate Law: k14*PGE2
k19 = 0.029 (0.0002778*s)^(-1)Reaction: PGD2 => dPGD2, Rate Law: k19*PGD2
k11 = 0.111 (0.0002778*s)^(-1)Reaction: AA => PGH2; LPS, Rate Law: k11*AA
k18 = 0.054 (0.0002778*s)^(-1)Reaction: PGD2 => PGJ2, Rate Law: k18*PGD2
LPSactivity = 0.0 dimensionless; k1 = 355.637 (0.0002778*s)^(-1); onepmol = 1.0 pmolReaction: FA => AA; LPS, Rate Law: k1*onepmol*LPSactivity
LPSactivity = 0.0 dimensionless; k12 = 0.098 (0.0002778*s)^(-1)Reaction: AA => PGH2, Rate Law: k12*AA*LPSactivity
k3 = 1.0E-15 (0.0002778*s)^(-1); DGactivity = 1.0 dimensionless; DNA = 1.0 μg; DGperDNA = 1.0 pmol*μg^(-1)Reaction: DG => AA, Rate Law: k3*DGactivity*DGperDNA*DNA
k4 = 1.0E-15 (0.0002778*s)^(-1)Reaction: AA =>, Rate Law: k4*AA
k15 = 0.061 (0.0002778*s)^(-1)Reaction: PGH2 => PGF2a, Rate Law: k15*PGH2
k13 = 0.204 (0.0002778*s)^(-1)Reaction: PGH2 => PGE2, Rate Law: k13*PGH2
k7 = 1.0E-15 (0.0002778*s)^(-1); GPChoratio = 1.0 dimensionlessReaction: GPCho => AA, Rate Law: k7*GPChoratio*GPCho

States:

NameDescription
PGJ2[Prostaglandin J2; LMFA03010019; prostaglandin J2]
FA[fatty acid]
HETE[LMFA03060085]
dPGJ2[15-Deoxy-Delta12,14-PGJ2; LMFA03010021; 15-deoxy-Delta(12,14)-prostaglandin J2]
PGD2[Prostaglandin D2; LMFA03010004; prostaglandin D2]
dPGD2[LMFA03010051]
GPCho[phosphatidylcholine(1+)]
PGF2a[Prostaglandin F2alpha; LMFA03010002; prostaglandin F2alpha]
PGH2[prostaglandin H2]
AA[Arachidonate; LMFA01030001; arachidonic acid]
DG[1,2-diglyceride]
PGE2[LMFA03010003]

Gupta2019 - Restoration of cytosolic calcium inhibits Mycobacterium tuberculosis intracellular growth: MODEL1911120004v0.0.1

This is a four dimensionsal ordinary differential equation mathematical model that explores the contribution of PI3P dur…

Details

Mycobacterium tuberculosis (Mtb) is a highly successful intracellular pathogen because of its ability to modulate host's anti-microbial pathways. Phagocytosis acts as the first line of defence against microbial infection. However, Mtb inhibits Phosphatidylinositol 3-phosphate (PI3P) oscillations which is required for phagolysosomal fusion. Here we attempted to understand the mechanisms by which Mtb eliminates phagosome-lysosome fusion. To address this, we built a four dimensional ordinary differential equation model and explored the contribution of PI3P during Mtb phagocytosis. Using this model, we identified some sensitive parameters that influence the dynamics of host-pathogen interactions. We observed that PI3P dynamics can be controlled by regulating the intracellular calcium oscillations. Some plausible methods to restore PI3P oscillations are ER flux rate, recruitment rate of proteins, like Rab GTPase, and cooperativity coefficient of calcium dependent consumption of calmodulin. Further, we investigated whether modulation of these pathways is a potential therapeutic intervention strategy. Here we showed that RyR2 agonist caffeine stimulated calcium influx and inhibited growth of intracellular Mtb in macrophages. Taken together, we demonstrate that modulation of host calcium level is a plausible strategy for killing of intracellular Mtb. link: http://identifiers.org/pubmed/31002776

Guyton1972_Aldosterone: MODEL0911376350v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Guyton1972_Angiotensin: MODEL0911342562v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Guyton1972_Autonomics: MODEL0911270005v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Guyton1972_Electrolytes: MODEL0912160001v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Guyton1972_HeartHypertrophy: MODEL0911231713v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Guyton1972_StressRelaxation: MODEL0910896131v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Guyton1972_volumeReceptors: MODEL0909931851v0.0.1

This a model from the article: Circulation: overall regulation. Guyton AC, Coleman TG, Granger HJ. Annu Rev Physiol…

Details

link: http://identifiers.org/pubmed/4334846

Gérard - 2019 - Coupling the cell cycle and the cell transformation networks (SBML): MODEL1906070001v0.0.1

Model for the Let-7-mediated coupling between the CDK network driving the cell cycle and the malignant cell transformati…

Details

The microRNA Let-7 controls the expression of proteins that belong to two distinct gene regulatory networks, namely, a cyclin-dependent kinase (Cdk) network driving the cell cycle and a cell transformation network that can undergo an epigenetic switch between a non-transformed and a malignant transformed cell state. Using mathematical modeling and transcriptomic data analysis, we here investigate how Let-7 controls the Cdk-dependent cell cycle network, and how it couples the latter with the transformation network. We also assess the consequence of this coupling on cancer progression. Our analysis shows that the switch from a quiescent to a proliferative state depends on the relative levels of Let-7 and several cell cycle activators. Numerical simulations further indicate that the Let-7-coupled cell cycle and transformation networks mutually control each other, and our model identifies key players for this mutual control. Transcriptomic data analysis from The Cancer Genome Atlas (TCGA) suggests that the two networks are activated in cancer, in particular in gastrointestinal cancers, and that the activation levels vary significantly among patients affected by a same cancer type. Our mathematical model, when applied to a heterogeneous cell population, suggests that heterogeneity among tumors may in part result from stochastic switches between a non-transformed cell state with low proliferative capability and a transformed cell state with high proliferative property. The model further predicts that Let-7 may reduce tumor heterogeneity by decreasing the occurrence of stochastic switches towards a transformed, proliferative cell state. In conclusion, we identified the key components responsible for the qualitative dynamics of two networks interconnected by Let-7. The two networks are heterogeneously activated in several cancers, thereby stressing the need to consider patient’s specific characteristics to optimize therapeutic strategies. link: http://identifiers.org/doi/10.3389/fphys.2019.00848

Görlich2003_RanGTP_gradient: BIOMD0000000192v0.0.1

This model represents a concentration gradient of RanGTP across the nuclear envelope. This gradient is generated by dist…

Details

Here, we analyse the RanGTPase system and its coupling to receptor-mediated nuclear transport. Our simulations predict nuclear RanGTP levels in HeLa cells to be very sensitive towards the cellular energy charge and to exceed the cytoplasmic concentration approximately 1000-fold. The steepness of the RanGTP gradient appears limited by both the cytoplasmic RanGAP concentration and the imperfect retention of nuclear RanGTP by nuclear pore complexes (NPCs), but not by the nucleotide exchange activity of RCC1. Neither RanBP1 nor the NPC localization of RanGAP has a significant direct impact on the RanGTP gradient. NTF2-mediated import of Ran appears to be the bottleneck for maximal capacity of Ran-driven nuclear transport. We show that unidirectional nuclear transport can be faithfully simulated without the assumption of a vectorial NPC passage; transport receptors only need to reversibly cross NPCs and switch their affinity for cargo in response to the RanGTP gradient. A significant RanGTP gradient after nuclear envelope (NE) breakdown can apparently exist only in large cytoplasm. This indicates that RanGTP gradients can provide positional information for mitotic spindle and NE assembly in early embryonic cells, but hardly any in small somatic cells. link: http://identifiers.org/pubmed/12606574

Parameters:

NameDescription
kpermRanGDP=0.12 psecReaction: RanGDP_nuc => RanGDP_cy, Rate Law: kpermRanGDP*nucleus*(RanGDP_nuc-RanGDP_cy)
kon=0.3 pmicroMpsec; koff=4.0E-4 psecReaction: RanGTP_cy + RanBP1 => RanGTP_RanBP1, Rate Law: (kon*RanGTP_cy*RanBP1-koff*RanGTP_RanBP1)*cytoplasm
r7=11.0 pmicroMpsec; r2=21.0 psecReaction: RCC1_RanGDP => RCC1_Ran + GDP, Rate Law: nucleus*(r2*RCC1_RanGDP-r7*RCC1_Ran*GDP)
r5=100.0 pmicroMpsec; r4=55.0 psecReaction: RCC1_RanGTP => RanGTP_nuc + RCC1, Rate Law: nucleus*(r4*RCC1_RanGTP-r5*RanGTP_nuc*RCC1)
r3=0.6 pmicroMpsec; r6=19.0 psecReaction: RCC1_Ran + GTP => RCC1_RanGTP, Rate Law: nucleus*(r3*RCC1_Ran*GTP-r6*RCC1_RanGTP)
kpermRanGTP=0.03 psecReaction: RanGTP_nuc => RanGTP_cy, Rate Law: kpermRanGTP*nucleus*(RanGTP_nuc-RanGTP_cy)
Km_GAP=0.7 microM; kcat_GAP=10.6 psecReaction: RanGTP_cy => RanGDP_cy; RanGAP, Rate Law: cytoplasm*kcat_GAP*RanGTP_cy*RanGAP/(Km_GAP+RanGTP_cy)
kcat=10.8 psec; Km=0.1 microMReaction: RanGTP_RanBP1 => RanGDP_cy + RanBP1; RanGAP, Rate Law: cytoplasm*kcat*RanGTP_RanBP1*RanGAP/(RanGTP_RanBP1+Km)
r8=55.0 psec; r1=74.0 pmicroMpsecReaction: RanGDP_nuc + RCC1 => RCC1_RanGDP, Rate Law: nucleus*(r1*RanGDP_nuc*RCC1-r8*RCC1_RanGDP)

States:

NameDescription
RCC1 RanGTP[GTP; GTP-binding nuclear protein Ran; Regulator of chromosome condensation; GTP]
RanGDP nuc[GTP-binding nuclear protein Ran; GDP]
GDP[GDP; GDP]
RCC1[RCC1; Regulator of chromosome condensation]
RanGDP cy[GTP-binding nuclear protein Ran; GDP]
RanGTP nuc[GTP-binding nuclear protein Ran; GTP]
GTP[GTP; GTP]
RCC1 RanGDP[GDP; Regulator of chromosome condensation; GTP-binding nuclear protein Ran; GDP]
RanGTP cy[GTP-binding nuclear protein Ran; GTP]
RanBP1[IPR000156; Ran-specific GTPase-activating protein]
RanGTP RanBP1[GTP-binding nuclear protein Ran; Ran-specific GTPase-activating protein; GTP]
RCC1 Ran[Regulator of chromosome condensation; GTP-binding nuclear protein Ran]

H


Haberichter2007_cellcycle: BIOMD0000000109v0.0.1

This model is according to the paper *A systems biology dynamical model of mammalian G1 cell cycle progression.* Supple…

Details

The current dogma of G(1) cell-cycle progression relies on growth factor-induced increase of cyclin D:Cdk4/6 complex activity to partially inactivate pRb by phosphorylation and to sequester p27(Kip1)-triggering activation of cyclin E:Cdk2 complexes that further inactivate pRb. pRb oscillates between an active, hypophosphorylated form associated with E2F transcription factors in early G(1) phase and an inactive, hyperphosphorylated form in late G(1), S and G(2)/M phases. However, under constant growth factor stimulation, cells show constitutively active cyclin D:Cdk4/6 throughout the cell cycle and thereby exclude cyclin D:Cdk4/6 inactivation of pRb. To address this paradox, we developed a mathematical model of G(1) progression using physiological expression and activity profiles from synchronized cells exposed to constant growth factors and included a metabolically responsive, activating modifier of cyclin E:Cdk2. Our mathematical model accurately simulates G(1) progression, recapitulates observations from targeted gene deletion studies and serves as a foundation for development of therapeutics targeting G(1) cell-cycle progression. link: http://identifiers.org/pubmed/17299420

Parameters:

NameDescription
kbYCyclinEYYCdk2 = 5.01E-5Reaction: Cdk2Y010 + CyclinE => Cdk2Y011, Rate Law: kbYCyclinEYYCdk2*Cdk2Y010*CyclinE*X
kuYD4YYpRb = 0.1Reaction: Cdk4Y01YpRbY00YpRbY10YInt => pRbY00 + Cdk4Y01, Rate Law: X*kuYD4YYpRb*Cdk4Y01YpRbY00YpRbY10YInt
kbYAPCCYYCyclinA = 1.61E-5Reaction: CyclinA + APCC => APCCYCyclinAYInt, Rate Law: X*kbYAPCCYYCyclinA*CyclinA*APCC
kuYp27YYCdk4 = 0.1Reaction: Cdk4Y11 => Cdk4Y01 + p27, Rate Law: kuYp27YYCdk4*Cdk4Y11*X
kdYE2F = 0.006465Reaction: pRbY01 => pRbY00, Rate Law: kdYE2F*pRbY01*X
kbYCyclinAYYCdk2 = 9.52E-5Reaction: Cdk2Y010 + CyclinA => Cdk2Y012, Rate Law: X*kbYCyclinAYYCdk2*Cdk2Y010*CyclinA
ktYpRbYYDephos = 0.023194Reaction: pRbY20 => pRbY00, Rate Law: X*ktYpRbYYDephos*pRbY20
kuYE2FYYpRb = 0.1Reaction: pRbY01 => pRbY00 + E2F, Rate Law: X*kuYE2FYYpRb*pRbY01
kupYE2YYpRb = 4.78271Reaction: Cdk2Y011YpRbY10YpRbY20YInt => pRbY20 + Cdk2Y011, Rate Law: X*kupYE2YYpRb*Cdk2Y011YpRbY10YpRbY20YInt
kdYCyclinE = 0.05Reaction: Cdk2Y011 => Cdk2Y010, Rate Law: kdYCyclinE*Cdk2Y011*X
kbYA2YYpRb = 6.25E-5Reaction: pRbY11 + Cdk2Y012 => Cdk2Y012YpRbY11YpRbY21YInt, Rate Law: X*kbYA2YYpRb*pRbY11*Cdk2Y012
kuYp27YYCdk2 = 0.1Reaction: Cdk2Y101 => Cdk2Y001 + p27, Rate Law: kuYp27YYCdk2*Cdk2Y101*X
kupYA2YYpRb = 0.200091Reaction: Cdk2Y012YpRbY11YpRbY21YInt => pRbY21 + Cdk2Y012, Rate Law: X*kupYA2YYpRb*Cdk2Y012YpRbY11YpRbY21YInt
kdYCyclinA = 0.05Reaction: Cdk2Y012 => Cdk2Y010, Rate Law: kdYCyclinA*Cdk2Y012*X
kYact = 0.0Reaction: Cdk2Y000 => Cdk2Y010, Rate Law: kYact*Cdk2Y000*X
kdYEmi1 = 0.018158Reaction: APCCYEmi1 => APCC, Rate Law: kdYEmi1*APCCYEmi1*X
kdYp27 = 0.001575Reaction: Cdk2Y102 => Cdk2Y002, Rate Law: kdYp27*Cdk2Y102*X
kuYCyclinAYYCdk2 = 0.1Reaction: Cdk2Y012 => Cdk2Y010 + CyclinA, Rate Law: X*kuYCyclinAYYCdk2*Cdk2Y012
kbYp27YYCdk2 = 1.23E-5Reaction: Cdk2Y000 + p27 => Cdk2Y100, Rate Law: kbYp27YYCdk2*Cdk2Y000*p27*X
kbYE2YYpRb = 5.74E-5Reaction: pRbY10 + Cdk2Y011 => Cdk2Y011YpRbY10YpRbY20YInt, Rate Law: X*kbYE2YYpRb*pRbY10*Cdk2Y011
kuYCyclinEYYCdk2 = 0.1Reaction: Cdk2Y001 => Cdk2Y000 + CyclinE, Rate Law: kuYCyclinEYYCdk2*Cdk2Y001*X
kudYAPCCYYCyclinA = 4.999555Reaction: APCCYCdk2Y000YCdk2Y002YInt => Cdk2Y000 + APCC, Rate Law: X*kudYAPCCYYCyclinA*APCCYCdk2Y000YCdk2Y002YInt
kuYAPCCYYCyclinA = 0.1Reaction: APCCYCyclinAYInt => CyclinA + APCC, Rate Law: X*kuYAPCCYYCyclinA*APCCYCyclinAYInt
kbYD4YYpRb = 3.15E-5Reaction: pRbY01 + Cdk4Y01 => Cdk4Y01YpRbY01YpRbY11YInt, Rate Law: X*kbYD4YYpRb*pRbY01*Cdk4Y01
kuYA2YYpRb = 0.1Reaction: Cdk2Y012YpRbY10YpRbY20YInt => pRbY10 + Cdk2Y012, Rate Law: X*kuYA2YYpRb*Cdk2Y012YpRbY10YpRbY20YInt
kupYA1YYpRb = 0.202132Reaction: Cdk1Y11YpRbY11YpRbY21YInt => pRbY21 + Cdk1Y11, Rate Law: X*kupYA1YYpRb*Cdk1Y11YpRbY11YpRbY21YInt
kuYE2YYpRb = 0.1Reaction: Cdk2Y011YpRbY10YpRbY20YInt => pRbY10 + Cdk2Y011, Rate Law: X*kuYE2YYpRb*Cdk2Y011YpRbY10YpRbY20YInt
kbYE2FYYpRb = 9.66E-6Reaction: pRbY00 + E2F => pRbY01, Rate Law: X*kbYE2FYYpRb*pRbY00*E2F
kd1Yp27 = 0.071149Reaction: Cdk2Y111 => Cdk2Y011, Rate Law: kd1Yp27*Cdk2Y111*X

States:

NameDescription
APCCYCdk1Y10YCdk1Y11YInt[Cyclin-dependent kinase 1; Anaphase-promoting complex subunit 2; IPR015453]
hyperphosphorylatedYpRbhyperphosphorylatedYpRb
Cdk2Y011YpRbY11YpRbY21YInt[G1/S-specific cyclin-E1; Cyclin-dependent kinase 2; Retinoblastoma-associated protein; IPR015633]
APCC[anaphase-promoting complex; Anaphase-promoting complex subunit 2]
Cdk2Y100[Cyclin-dependent kinase 2; IPR015456]
Cdk2Y010[Cyclin-dependent kinase 2]
pRbY01[Rb-E2F complex]
Cdk2Y011[Cyclin-dependent kinase 2; G1/S-specific cyclin-E1]
pRbY00[Retinoblastoma-associated protein]
Cdk2Y012[Cyclin-dependent kinase 2; IPR015453]
Cdk2Y012YpRbY10YpRbY20YInt[Cyclin-dependent kinase 2; Retinoblastoma-associated protein; IPR015453]
hypophosphorylatedYpRbhypophosphorylatedYpRb
pRbY11[Retinoblastoma-associated protein; Rb-E2F complex]
p27[IPR015456]
pRbY10[Retinoblastoma-associated protein]
Cdk2Y001[Cyclin-dependent kinase 2; G1/S-specific cyclin-E1]
pRbY21[Retinoblastoma-associated protein; Rb-E2F complex]
Cdk2Y011YpRbY10YpRbY20YInt[G1/S-specific cyclin-E1; Cyclin-dependent kinase 2; Retinoblastoma-associated protein]
Cdk2Y012YpRbY11YpRbY21YInt[Cyclin-dependent kinase 2; Retinoblastoma-associated protein; IPR015453; IPR015633]
APCCYCdk1Y00YCdk1Y01YInt[Anaphase-promoting complex subunit 2; Cyclin-dependent kinase 1; IPR015453]
Cdk2Y002[Cyclin-dependent kinase 2]

Haffez2017 - RAR interaction with synthetic analogues: BIOMD0000000629v0.0.1

Haffez2017 - RAR interaction with synthetic analogues This model is described in the article: [The molecular basis of…

Details

All-trans-retinoic acid (ATRA) and its synthetic analogues EC23 and EC19 direct cellular differentiation by interacting as ligands for the retinoic acid receptor (RARα, β and γ) family of nuclear receptor proteins. To date, a number of crystal structures of natural and synthetic ligands complexed to their target proteins have been solved, providing molecular level snap-shots of ligand binding. However, a deeper understanding of receptor and ligand flexibility and conformational freedom is required to develop stable and effective ATRA analogues for clinical use. Therefore, we have used molecular modelling techniques to define RAR interactions with ATRA and two synthetic analogues, EC19 and EC23, and compared their predicted biochemical activities to experimental measurements of relative ligand affinity and recruitment of coactivator proteins. A comprehensive molecular docking approach that explored the conformational space of the ligands indicated that ATRA is able to bind the three RAR proteins in a number of conformations with one extended structure being favoured. In contrast the biologically-distinct isomer, 9-cis-retinoic acid (9CRA), showed significantly less conformational flexibility in the RAR binding pockets. These findings were used to inform docking studies of the synthetic retinoids EC23 and EC19, and their respective methyl esters. EC23 was found to be an excellent mimic for ATRA, and occupied similar binding modes to ATRA in all three target RAR proteins. In comparison, EC19 exhibited an alternative binding mode which reduces the strength of key polar interactions in RARα/γ but is well-suited to the larger RARβ binding pocket. In contrast, docking of the corresponding esters revealed the loss of key polar interactions which may explain the much reduced biological activity. Our computational results were complemented using an in vitro binding assay based on FRET measurements, which showed that EC23 was a strongly binding, pan-agonist of the RARs, while EC19 exhibited specificity for RARβ, as predicted by the docking studies. These findings can account for the distinct behaviour of EC23 and EC19 in cellular differentiation assays, and additionally, the methods described herein can be further applied to the understanding of the molecular basis for the selectivity of different retinoids to RARα, β and γ. link: http://identifiers.org/doi/10.1039/C6MD00680A

Parameters:

NameDescription
k2=0.2; k1=0.014Reaction: LR + CA => LRCA, Rate Law: RAR_retinoids*(k1*LR*CA-k2*LRCA)
k1=0.6; k2=0.1Reaction: L + R => LR, Rate Law: RAR_retinoids*(k1*L*R-k2*LR)

States:

NameDescription
LR[all-trans-retinoic acid; Retinoic acid receptor alpha]
LRCA[all-trans-retinoic acid; Retinoic acid receptor alpha]
CA[fluorescin; peptide]
L[all-trans-retinoic acid]
R[Retinoic acid receptor alpha]

Halloy2002_FollicularAutomaton: MODEL1006230014v0.0.1

This a model from the article: The follicular automaton model: effect of stochasticity and of synchronization of hair…

Details

Human scalp hair consists of a set of about 10(5)follicles which progress independently through developmental cycles. Each hair follicle successively goes through the anagen (A), catagen (C), telogen (T) and latency (L) phases that correspond, respectively, to growth, arrest and hair shedding before a new anagen phase is initiated. Long-term experimental observations in a group of ten male, alopecic and non-alopecic volunteers allowed determination of the characteristics of hair follicle cycles. On the basis of these observations, we previously proposed a follicular automaton model to simulate the dynamics of human hair cycles and the development of different patterns of alopecia [Halloy et al. (2000) Proc. Natl Acad. Sci. U.S.A.97, 8328-8333]. The automaton model is defined by a set of rules that govern the stochastic transitions of each follicle between the successive states A, T, L and the subsequent return to A. These transitions occur independently for each follicle, after time intervals given stochastically by a distribution characterized by a mean and a standard deviation. The follicular automaton model was shown to account both for the dynamical transitions observed in a single follicle, and for the behaviour of an ensemble of independently cycling follicles. Here, we extend these results and investigate additional properties of the model. We present a deterministic version of the follicular automaton. We show that numerical simulations of the stochastic version of the automaton yield steady-state level of follicles in the different phases which approach the levels predicted by the deterministic equations as the number of follicles progressively increases. Only the stochastic version can successfully reproduce the fluctuations of the fractions of follicles in each of the three phases, observed in small follicle populations. When the standard deviation is reduced or when the follicles become otherwise synchronized, e.g. by a periodic external signal inducing the transition of anagen follicles into telogen phase, large-amplitude oscillations occur in the fractions of follicles in the three phases. These oscillations are not observed in humans but are reminiscent of the phenomenon of moulting observed in a number of mammalian species. link: http://identifiers.org/pubmed/11846603

Hamey2017 - Blood stem cell regulatory network: MODEL1610060000v0.0.1

Hamey2017 - Blood stem cell regulatory networkThis model is described in the article: [Reconstructing blood stem cell r…

Details

Adult blood contains a mixture of mature cell types, each with specialized functions. Single hematopoietic stem cells (HSCs) have been functionally shown to generate all mature cell types for the lifetime of the organism. Differentiation of HSCs toward alternative lineages must be balanced at the population level by the fate decisions made by individual cells. Transcription factors play a key role in regulating these decisions and operate within organized regulatory programs that can be modeled as transcriptional regulatory networks. As dysregulation of single HSC fate decisions is linked to fatal malignancies such as leukemia, it is important to understand how these decisions are controlled on a cell-by-cell basis. Here we developed and applied a network inference method, exploiting the ability to infer dynamic information from single-cell snapshot expression data based on expression profiles of 48 genes in 2,167 blood stem and progenitor cells. This approach allowed us to infer transcriptional regulatory network models that recapitulated differentiation of HSCs into progenitor cell types, focusing on trajectories toward megakaryocyte–erythrocyte progenitors and lymphoid-primed multipotent progenitors. By comparing these two models, we identified and subsequently experimentally validated a difference in the regulation of nuclear factor, erythroid 2 (Nfe2) and core-binding factor, runt domain, alpha subunit 2, translocated to, 3 homolog (Cbfa2t3h) by the transcription factor Gata2. Our approach confirms known aspects of hematopoiesis, provides hypotheses about regulation of HSC differentiation, and is widely applicable to other hierarchical biological systems to uncover regulatory relationships. link: http://identifiers.org/doi/10.1073/pnas.1610609114

Hamey2017 - Blood stem cell regulatory network (LMPP network): MODEL1610060001v0.0.1

Hamey2017 - Blood stem cell regulatory network (LMPP network)This model is described in the article: [Reconstructing bl…

Details

Adult blood contains a mixture of mature cell types, each with specialized functions. Single hematopoietic stem cells (HSCs) have been functionally shown to generate all mature cell types for the lifetime of the organism. Differentiation of HSCs toward alternative lineages must be balanced at the population level by the fate decisions made by individual cells. Transcription factors play a key role in regulating these decisions and operate within organized regulatory programs that can be modeled as transcriptional regulatory networks. As dysregulation of single HSC fate decisions is linked to fatal malignancies such as leukemia, it is important to understand how these decisions are controlled on a cell-by-cell basis. Here we developed and applied a network inference method, exploiting the ability to infer dynamic information from single-cell snapshot expression data based on expression profiles of 48 genes in 2,167 blood stem and progenitor cells. This approach allowed us to infer transcriptional regulatory network models that recapitulated differentiation of HSCs into progenitor cell types, focusing on trajectories toward megakaryocyte–erythrocyte progenitors and lymphoid-primed multipotent progenitors. By comparing these two models, we identified and subsequently experimentally validated a difference in the regulation of nuclear factor, erythroid 2 (Nfe2) and core-binding factor, runt domain, alpha subunit 2, translocated to, 3 homolog (Cbfa2t3h) by the transcription factor Gata2. Our approach confirms known aspects of hematopoiesis, provides hypotheses about regulation of HSC differentiation, and is widely applicable to other hierarchical biological systems to uncover regulatory relationships. link: http://identifiers.org/doi/10.1073/pnas.1610609114

Hansen2019 - Nine species reduced model of blood coagulation: BIOMD0000000755v0.0.1

its a nine species reduced model of Hockin 2002. Model uses different level of reduction (5,7,9,11) and testing the best…

Details

Mathematical modeling of thrombosis typically involves modeling the coagulation cascade. Models of coagulation generally involve the reaction kinetics for dozens of proteins. The resulting system of equations is difficult to parameterize and its numerical solution is challenging when coupled to blood flow or other physics important to clotting. Prior research suggests that essential aspects of coagulation may be reproduced by simpler models. This evidence motivates a systematic approach to model reduction. We herein introduce an automated framework to generate reduced-order models of blood coagulation. The framework consists of nested optimizations, where an outer optimization selects the optimal species for the reduced-order model and an inner optimization selects the optimal reaction rates for the new coagulation network. The framework was tested on an established 34-species coagulation model to rigorously consider what level of model fidelity is necessary to capture essential coagulation dynamics. The results indicate that a nine-species reduced-order model is sufficient to reproduce the thrombin dynamics of the benchmark 34-species model for a range of tissue factor concentrations, including those not included in the optimization process. Further model reduction begins to compromise the ability to capture the thrombin generation process. The framework proposed herein enables automated development of reduced-order models of coagulation that maintain essential dynamics used to model thrombosis. link: http://identifiers.org/pubmed/31161687

Parameters:

NameDescription
k1=121.267Reaction: TF + X => Xa_Va, Rate Law: compartment*k1*TF*X
k1=0.0201671Reaction: IIa => IIa_ATIII, Rate Law: compartment*k1*IIa
k1=77540.2Reaction: Xa_Va_II => Xa_Va + mIIa, Rate Law: compartment*k1*Xa_Va_II
k1=2.56984E12Reaction: Xa_Va + II => Xa_Va_II, Rate Law: compartment*k1*Xa_Va*II
k1=4.74645E-16Reaction: TF + X + II => TF + X + IIa, Rate Law: compartment*k1*TF*X*II
k1=6.96794E10Reaction: Xa_Va + mIIa => Xa_Va + IIa, Rate Law: compartment*k1*Xa_Va*mIIa
k1=0.00472749Reaction: mIIa => mIIa_ATIII, Rate Law: compartment*k1*mIIa

States:

NameDescription
mIIa[Prothrombin]
mIIa ATIII[Prothrombin; Antithrombin-III]
IIa[Prothrombin]
IIa ATIII[Antithrombin-III; Prothrombin]
X[Coagulation factor X]
Xa Va[Coagulation factor X; Coagulation factor V]
Xa Va II[Coagulation factor V; Coagulation factor X; Prothrombin]
TF[Tissue factor]
II[Prothrombin]

Hansen2019 - Seven species reduced model of blood coagulation: MODEL1907180003v0.0.1

its a seven species reduced model of Hockin 2002. Model uses different level of reduction (5,7,9,11) and testing the bes…

Details

Mathematical modeling of thrombosis typically involves modeling the coagulation cascade. Models of coagulation generally involve the reaction kinetics for dozens of proteins. The resulting system of equations is difficult to parameterize and its numerical solution is challenging when coupled to blood flow or other physics important to clotting. Prior research suggests that essential aspects of coagulation may be reproduced by simpler models. This evidence motivates a systematic approach to model reduction. We herein introduce an automated framework to generate reduced-order models of blood coagulation. The framework consists of nested optimizations, where an outer optimization selects the optimal species for the reduced-order model and an inner optimization selects the optimal reaction rates for the new coagulation network. The framework was tested on an established 34-species coagulation model to rigorously consider what level of model fidelity is necessary to capture essential coagulation dynamics. The results indicate that a nine-species reduced-order model is sufficient to reproduce the thrombin dynamics of the benchmark 34-species model for a range of tissue factor concentrations, including those not included in the optimization process. Further model reduction begins to compromise the ability to capture the thrombin generation process. The framework proposed herein enables automated development of reduced-order models of coagulation that maintain essential dynamics used to model thrombosis. link: http://identifiers.org/pubmed/31161687

Hanson2016 - Toxicity Management in CAR T cell therapy for B-ALL: BIOMD0000000837v0.0.1

This model provides an in silico mathematical platform to explore the interactions between chimeric antigen receptor-mod…

Details

Advances in genetic engineering have made it possible to reprogram individual immune cells to express receptors that recognise markers on tumour cell surfaces. The process of re-engineering T cell lymphocytes to express Chimeric Antigen Receptors (CARs), and then re-infusing the CAR-modified T cells into patients to treat various cancers is referred to as CAR T cell therapy. This therapy is being explored in clinical trials - most prominently for B Cell Acute Lymphoblastic Leukaemia (B-ALL), a common B cell malignancy, for which CAR T cell therapy has led to remission in up to 90% of patients. Despite this extraordinary response rate, however, potentially fatal inflammatory side effects occur in up to 10% of patients who have positive responses. Further, approximately 50% of patients who initially respond to the therapy relapse. Significant improvement is thus necessary before the therapy can be made widely available for use in the clinic.

To inform future development, we develop a mathematical model to explore interactions between CAR T cells, inflammatory toxicity, and individual patients’ tumour burdens in silico. This paper outlines the underlying system of coupled ordinary differential equations designed based on well-known immunological principles and widely accepted views on the mechanism of toxicity development in CAR T cell therapy for B-ALL - and reports in silico outcomes in relationship to standard and recently conjectured predictors of toxicity in a heterogeneous, randomly generated patient population. Our initial results and analyses are consistent with and connect immunological mechanisms to the clinically observed, counterintuitive hypothesis that initial tumour burden is a stronger predictor of toxicity than is the dose of CAR T cells administered to patients.

We outline how the mechanism of action in CAR T cell therapy can give rise to such non-standard trends in toxicity development, and demonstrate the utility of mathematical modelling in understanding the relationship between predictors of toxicity, mechanism of action, and patient outcomes. link: http://identifiers.org/doi/10.1101/049908

Parameters:

NameDescription
p_1 = 0.002Reaction: => Inflam; B, C_e, H_e, Rate Law: compartment*p_1*B*(C_e+H_e)
r_4 = 0.1; Lymphocyte_Term = 0.0Reaction: => L; L, Rate Law: compartment*r_4*L*Lymphocyte_Term
p_0 = 200.0Reaction: => Inflam, Rate Law: compartment*p_0
d_3 = 0.004Reaction: C_e =>, Rate Law: compartment*d_3*C_e
d_4 = 0.004Reaction: H_e =>, Rate Law: compartment*d_4*H_e
d_1 = 2.0E-4Reaction: B => ; C_e, Rate Law: compartment*d_1*B*C_e
Lymphocyte_Term = 0.0; r_3 = 0.1Reaction: => H_m; H_m, Rate Law: compartment*r_3*H_m*Lymphocyte_Term
k = 4800.0; r_1 = 0.003Reaction: => B; B, Rate Law: compartment*r_1*B*(1-B/k)
b = 800.0; a_3 = 8.0E-5Reaction: H_m => ; B, Inflam, Rate Law: compartment*a_3*B*H_m*Inflam^2/(Inflam^2+b^2)
n = 6.0; b = 800.0; a_1 = 4.0E-7; a_2 = 2.0Reaction: => C_e; B, C_m, Inflam, H_e, Rate Law: compartment*2^n*a_1*B*C_m*Inflam^2/(Inflam^2+b^2)*(1+a_2*H_e)
b = 800.0; a_1 = 4.0E-7; a_2 = 2.0Reaction: C_m => ; B, H_e, Inflam, Rate Law: compartment*a_1*B*C_m*(1+a_2*H_e)*Inflam^2/(Inflam^2+b^2)
p_2 = 0.4Reaction: => L, Rate Law: compartment*p_2
d_2 = 1.5Reaction: Inflam =>, Rate Law: compartment*d_2*Inflam
n = 6.0; b = 800.0; a_3 = 8.0E-5Reaction: => H_e; B, H_m, Inflam, Rate Law: compartment*2^n*a_3*B*H_m*Inflam^2/(Inflam^2+b^2)
d_5 = 2.0E-4Reaction: L =>, Rate Law: compartment*d_5*L
r_2 = 0.1; Lymphocyte_Term = 0.0Reaction: => C_m; C_m, Inflam, Rate Law: compartment*r_2*C_m*Lymphocyte_Term

States:

NameDescription
H m[helper T cell]
B[BTO:0000731]
C m[cytotoxic T cell]
Inflam[inflammatory response]
H e[helper T cell]
C e[cytotoxic T cell]
L[lymphocyte]

Hartmann2016 - Blood coagulation model for simulating anticoagulants: MODEL1807180004v0.0.1

Mathematical model of blood coagulation. Reused Wajima2009 model with modifications to reactions 27 (formation of Va:Xa…

Details

Warfarin is the anticoagulant of choice for venous thromboembolism (VTE) treatment, although its suppression of the endogenous clot-dissolution complex APC:PS may ultimately lead to longer time-to-clot dissolution profiles, resulting in increased risk of re-thrombosis. This detrimental effect might not occur during VTE treatment using other anticoagulants, such as rivaroxaban or enoxaparin, given their different mechanisms of action within the coagulation network. A quantitative systems pharmacology model was developed describing the coagulation network to monitor clotting factor levels under warfarin, enoxaparin, and rivaroxaban treatment. The model allowed for estimation of all factor rate constants and production rates. Predictions of individual coagulation factor time courses under steady-state warfarin, enoxaparin, and rivaroxaban treatment reflected the suppression of protein C and protein S under warfarin compared to rivaroxaban and enoxaparin. The model may be used as a tool during clinical practice to predict effects of anticoagulants on individual clotting factor time courses and optimize antithrombotic therapy. link: http://identifiers.org/pubmed/27647667

Hat2016 - Reponse of p53 System to irradiation in cell fate decision making: BIOMD0000000943v0.0.1

The p53 transcription factor is a regulator of key cellular processes including DNA repair, cell cycle arrest, and apopt…

Details

The p53 transcription factor is a regulator of key cellular processes including DNA repair, cell cycle arrest, and apoptosis. In this theoretical study, we investigate how the complex circuitry of the p53 network allows for stochastic yet unambiguous cell fate decision-making. The proposed Markov chain model consists of the regulatory core and two subordinated bistable modules responsible for cell cycle arrest and apoptosis. The regulatory core is controlled by two negative feedback loops (regulated by Mdm2 and Wip1) responsible for oscillations, and two antagonistic positive feedback loops (regulated by phosphatases Wip1 and PTEN) responsible for bistability. By means of bifurcation analysis of the deterministic approximation we capture the recurrent solutions (i.e., steady states and limit cycles) that delineate temporal responses of the stochastic system. Direct switching from the limit-cycle oscillations to the "apoptotic" steady state is enabled by the existence of a subcritical Neimark-Sacker bifurcation in which the limit cycle loses its stability by merging with an unstable invariant torus. Our analysis provides an explanation why cancer cell lines known to have vastly diverse expression levels of Wip1 and PTEN exhibit a broad spectrum of responses to DNA damage: from a fast transition to a high level of p53 killer (a p53 phosphoform which promotes commitment to apoptosis) in cells characterized by high PTEN and low Wip1 levels to long-lasting p53 level oscillations in cells having PTEN promoter methylated (as in, e.g., MCF-7 cell line). link: http://identifiers.org/pubmed/26928575

Parameters:

NameDescription
u6 = 1.0E-4Reaction: Cyclin_E_p21_complex => p21__free + Cyclin_E__free, Rate Law: nuclear*u6*Cyclin_E_p21_complex
g16 = 1.0E-4Reaction: Mdm2_nuc_S166S186p_S395p =>, Rate Law: nuclear*g16*Mdm2_nuc_S166S186p_S395p
b2 = 0.003Reaction: BclXL__free + Bad_0__free =>, Rate Law: cytoplasm*b2*BclXL__free*Bad_0__free
s1 = 0.1; q0_wip1 = 1.0E-5; q1_wip1 = 3.0E-13; h = 2.0; q2 = 0.003Reaction: => Wip1_mRNA; p53_arrester, Rate Law: nuclear*s1*(q0_wip1+q1_wip1*p53_arrester^h)/(q2+q0_wip1+q1_wip1*p53_arrester^h)
b3 = 0.003Reaction: Bad_phosphorylated__free => ; Fourteen33_free, Rate Law: b3*Fourteen33_free*Bad_phosphorylated__free
p3 = 3.0E-8Reaction: p53_0phosphorylated => p53_arrester; ATM_phosphorylated, Rate Law: nuclear*p3*ATM_phosphorylated*p53_0phosphorylated
g17 = 3.0E-4Reaction: proCaspase =>, Rate Law: nuclear*g17*proCaspase
g6 = 3.0E-5Reaction: PTEN =>, Rate Law: nuclear*g6*PTEN
g14 = 1.0E-4Reaction: Mdm2_cyt_0phosphorylated =>, Rate Law: cytoplasm*g14*Mdm2_cyt_0phosphorylated
t5 = 0.1Reaction: => p21__free; p21_mRNA, Rate Law: nuclear*t5*p21_mRNA
d9 = 3.0E-5Reaction: Bad_phosphorylated__free => Bad_0__free, Rate Law: cytoplasm*d9*Bad_phosphorylated__free
d4 = 1.0E-10Reaction: p53_killer => p53_arrester; Wip1, Rate Law: nuclear*d4*Wip1*p53_killer
s8 = 30.0Reaction: => HIPK2, Rate Law: nuclear*s8
d8 = 1.0E-4Reaction: AKT_phosphorylated =>, Rate Law: cytoplasm*d8*AKT_phosphorylated
h = 2.0; p1 = 3.0E-4; M1 = 5.0Reaction: => ATM_phosphorylated; ATM_0, DNA_double_strand_break, Rate Law: nuclear*p1*ATM_0*DNA_double_strand_break^h/(DNA_double_strand_break^h+M1^h)
s9 = 30.0; M3 = 200000.0Reaction: => Cyclin_E__free; E2F1, Rate Law: nuclear*s9*E2F1^2/(M3^2+E2F1^2)
g7 = 1.0E-13Reaction: HIPK2 => ; SIAH1_0, Mdm2_nuc_S166S186phosphorylated, Rate Law: nuclear*g7*HIPK2*(SIAH1_0+Mdm2_nuc_S166S186phosphorylated)^2
s10 = 3.0Reaction: => Cyclin_E__free, Rate Law: nuclear*s10
g20 = 1.0E-4Reaction: Cyclin_E__free =>, Rate Law: nuclear*g20*Cyclin_E__free
q1_p21 = 3.0E-13; q0_p21 = 1.0E-5; s5 = 0.1; h = 2.0; q2 = 0.003Reaction: => p21_mRNA; p53_arrester, Rate Law: nuclear*s5*(q0_p21+q1_p21*p53_arrester^h)/(q2+q0_p21+q1_p21*p53_arrester^h)
p8 = 3.0E-9Reaction: => PIP3; PIP2, PI3K_tot, Rate Law: nuclear*p8*PIP2*PI3K_tot
p7 = 3.0E-9Reaction: Bad_0__free => Bad_phosphorylated__free; AKT_phosphorylated, Rate Law: cytoplasm*p7*AKT_phosphorylated*Bad_0__free
t1 = 0.1Reaction: => Wip1; Wip1_mRNA, Rate Law: nuclear*t1*Wip1_mRNA
u3 = 0.001Reaction: => Bad_phosphorylated__free; Bad_phosphorylated_Fourteen33_complex, Rate Law: cytoplasm*u3*Bad_phosphorylated_Fourteen33_complex
s7 = 30.0Reaction: => proCaspase, Rate Law: nuclear*s7
p11 = 1.0E-10Reaction: p53_0phosphorylated => p53_S46phosphorylated; HIPK2, Rate Law: nuclear*p11*HIPK2*p53_0phosphorylated
d7 = 3.0E-7Reaction: PIP3 => ; PTEN, Rate Law: nuclear*d7*PTEN*PIP3
d10 = 1.0E-4Reaction: p53_killer => p53_S46phosphorylated, Rate Law: nuclear*d10*p53_killer
t3 = 0.1Reaction: => Mdm2_cyt_0phosphorylated; Mdm2_mRNA, Rate Law: t3*Mdm2_mRNA
u2 = 0.001Reaction: => Bad_0__free; BclXL_Bad_complex, Rate Law: cytoplasm*u2*BclXL_Bad_complex
s6 = 300.0Reaction: => p53_0phosphorylated, Rate Law: nuclear*s6
DNA_DSB_due_to_IR = 0.0333333333333333; h1 = 1.0E-6; is_IR_switched_on = 0.0; h2 = 1.0E-13; DNA_DSB_max = 1000000.0Reaction: => DNA_double_strand_break; Caspase, DNA_double_strand_break, Rate Law: nuclear*(h1*DNA_DSB_due_to_IR*is_IR_switched_on+h2*Caspase)*(DNA_DSB_max-DNA_double_strand_break)
g18 = 3.0E-4Reaction: Caspase =>, Rate Law: nuclear*g18*Caspase
p5 = 1.0E-8Reaction: Mdm2_cyt_0phosphorylated => Mdm2_cyt_S166S186phosphorylated; AKT_phosphorylated, Rate Law: cytoplasm*p5*AKT_phosphorylated*Mdm2_cyt_0phosphorylated
d5 = 1.0E-4Reaction: Mdm2_cyt_S166S186phosphorylated => Mdm2_cyt_0phosphorylated, Rate Law: cytoplasm*d5*Mdm2_cyt_S166S186phosphorylated
d11 = 1.0E-10Reaction: p53_S46phosphorylated => p53_0phosphorylated; Wip1, Rate Law: nuclear*d11*Wip1*p53_S46phosphorylated
t2 = 0.1Reaction: => PTEN; PTEN_mRNA, Rate Law: nuclear*t2*PTEN_mRNA
a1 = 3.0E-10Reaction: proCaspase => Caspase; Bax__free, Rate Law: nuclear*a1*Bax__free*proCaspase
rep = 0.001; DNA_DSB_RepairCplx_total = 20.0Reaction: DNA_double_strand_break =>, Rate Law: nuclear*DNA_double_strand_break*rep/(DNA_double_strand_break+DNA_DSB_RepairCplx_total)
g101 = 1.0E-5; h = 2.0; g11 = 1.0E-11Reaction: p53_0phosphorylated => ; Mdm2_nuc_S166S186phosphorylated, Rate Law: nuclear*(g101+g11*Mdm2_nuc_S166S186phosphorylated^h)*p53_0phosphorylated
g3 = 3.0E-4Reaction: Mdm2_mRNA =>, Rate Law: nuclear*g3*Mdm2_mRNA
p12 = 1.0E-9Reaction: => AKT_phosphorylated; AKT_0, PIP3, Rate Law: p12*AKT_0*PIP3
g5 = 3.0E-4Reaction: p21_mRNA =>, Rate Law: nuclear*g5*p21_mRNA
p2 = 1.0E-8Reaction: SIAH1_0 => ; ATM_phosphorylated, Rate Law: nuclear*p2*ATM_phosphorylated*SIAH1_0
d6 = 1.0E-10Reaction: Mdm2_nuc_S166S186p_S395p => Mdm2_nuc_S166S186phosphorylated; Wip1, Rate Law: nuclear*d6*Wip1*Mdm2_nuc_S166S186p_S395p
q1_pten = 3.0E-13; s2 = 0.03; h = 2.0; q0_pten = 1.0E-5; q2 = 0.003Reaction: => PTEN_mRNA; p53_killer, Rate Law: nuclear*s2*(q0_pten+q1_pten*p53_killer^h)/(q2+q0_pten+q1_pten*p53_killer^h)
d3 = 1.0E-4Reaction: p53_arrester => p53_0phosphorylated, Rate Law: nuclear*d3*p53_arrester
a2 = 1.0E-12Reaction: proCaspase => Caspase, Rate Law: nuclear*a2*Caspase^2*proCaspase
g8 = 3.0E-4Reaction: Wip1 =>, Rate Law: nuclear*g8*Wip1
g15 = 3.0E-5Reaction: Mdm2_nuc_S166S186phosphorylated =>, Rate Law: nuclear*g15*Mdm2_nuc_S166S186phosphorylated
d1 = 1.0E-8Reaction: ATM_phosphorylated => ; Wip1, Rate Law: nuclear*d1*Wip1*ATM_phosphorylated
g101 = 1.0E-5; h = 2.0; g12 = 1.0E-13Reaction: p53_arrester => ; Mdm2_nuc_S166S186phosphorylated, Rate Law: nuclear*(g101+g12*Mdm2_nuc_S166S186phosphorylated^h)*p53_arrester
g19 = 3.0E-4Reaction: p21__free =>, Rate Law: nuclear*g19*p21__free
i1 = 0.001Reaction: Mdm2_cyt_S166S186phosphorylated => Mdm2_nuc_S166S186phosphorylated, Rate Law: i1*Mdm2_cyt_S166S186phosphorylated
h = 2.0; g10 = 1.0E-5; g13 = 1.0E-13Reaction: p53_S46phosphorylated => ; Mdm2_nuc_S166S186phosphorylated, Rate Law: nuclear*(g10+g13*Mdm2_nuc_S166S186phosphorylated^h)*p53_S46phosphorylated
g2 = 3.0E-4Reaction: PTEN_mRNA =>, Rate Law: nuclear*g2*PTEN_mRNA
b5 = 1.0E-5Reaction: p21__free + Cyclin_E__free => Cyclin_E_p21_complex, Rate Law: nuclear*b5*p21__free*Cyclin_E__free
q0_mdm2 = 1.0E-4; q1_mdm2 = 3.0E-13; h = 2.0; s3 = 0.1; q2 = 0.003Reaction: => Mdm2_mRNA; p53_arrester, Rate Law: nuclear*s3*(q0_mdm2+q1_mdm2*p53_arrester^h)/(q2+q0_mdm2+q1_mdm2*p53_arrester^h)
p4 = 1.0E-10Reaction: p53_arrester => p53_killer; HIPK2, Rate Law: nuclear*p4*HIPK2*p53_arrester
d2 = 3.0E-5Reaction: => SIAH1_0; SIAH1_phosphorylated, Rate Law: nuclear*d2*SIAH1_phosphorylated
g1 = 3.0E-4Reaction: Wip1_mRNA =>, Rate Law: nuclear*g1*Wip1_mRNA
p6 = 1.0E-8Reaction: Mdm2_nuc_S166S186phosphorylated => Mdm2_nuc_S166S186p_S395p; ATM_phosphorylated, Rate Law: nuclear*p6*ATM_phosphorylated*Mdm2_nuc_S166S186phosphorylated

States:

NameDescription
Mdm2 nuc S166S186p S395p[E3 ubiquitin-protein ligase Mdm2; phosphorylated]
Caspase[Caspase-2]
Wip1[Protein phosphatase 1D]
BclXL free[Bcl-2-like protein 1]
Bad phosphorylated free[Bcl2-associated agonist of cell death; phosphorylated]
PTEN[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN]
proCaspase[Precursor]
p53 S46phosphorylated[Cellular tumor antigen p53; phosphorylated]
p53 0phosphorylated[Cellular tumor antigen p53; phosphorylated]
p53 arrester[Cellular tumor antigen p53]
Wip1 mRNA[Protein phosphatase 1D; ribonucleic acid]
PTEN mRNA[Phosphatidylinositol 3,4,5-trisphosphate 3-phosphatase and dual-specificity protein phosphatase PTEN; ribonucleic acid]
PIP2PIP2
Bad phosphorylated Fourteen33 complex[14-3-3 protein sigma; Bcl2-associated agonist of cell death; phosphorylated]
p53 killer[Cellular tumor antigen p53]
Mdm2 cyt S166S186phosphorylated[E3 ubiquitin-protein ligase Mdm2; phosphorylated]
PIP3PIP3
p21 free[p21 RAS Protein]
Bad 0 free[Bcl2-associated agonist of cell death]
ATM 0[Serine-protein kinase ATM]
Rb phosphorylated[Retinoblastoma-associated protein; phosphorylated]
Cyclin E p21 complex[p21 RAS Protein; G1/S-specific cyclin-E2]
SIAH1 phosphorylated[E3 ubiquitin-protein ligase SIAH1; phosphorylated]
AKT phosphorylated[RAC-alpha serine/threonine-protein kinase; phosphorylated]
E2F1[Transcription factor E2F1]
AKT 0[RAC-alpha serine/threonine-protein kinase]
Fourteen33 free[14-3-3 protein sigma]
Mdm2 nuc S166S186phosphorylated[E3 ubiquitin-protein ligase Mdm2; phosphorylated]
Cyclin E free[G1/S-specific cyclin-E2]
Mdm2 cyt 0phosphorylated[E3 ubiquitin-protein ligase Mdm2; phosphorylated]
BclXL Bad complex[Bcl2-associated agonist of cell death; Bcl-2-like protein 1]
SIAH1 0[E3 ubiquitin-protein ligase SIAH1]
HIPK2[Homeodomain-interacting protein kinase 2]
DNA double strand break[site of double-strand break]
ATM phosphorylated[Serine-protein kinase ATM; phosphorylated]
Mdm2 mRNA[E3 ubiquitin-protein ligase Mdm2; ribonucleic acid]
p21 mRNA[p21 RAS Protein; ribonucleic acid]

Hatakeyama2003_MAPK: BIOMD0000000146v0.0.1

Figure4 and Figure5 can be simulated by Copasi. Figure4 can be simulated in MathSBML as well. There are some typos in th…

Details

ErbB tyrosine kinase receptors mediate mitogenic signal cascade by binding a variety of ligands and recruiting the different cassettes of adaptor proteins. In the present study, we examined heregulin (HRG)-induced signal transduction of ErbB4 receptor and found that the phosphatidylinositol 3'-kinase (PI3K)-Akt pathway negatively regulated the extracellular signal-regulated kinase (ERK) cascade by phosphorylating Raf-1 on Ser(259). As the time-course kinetics of Akt and ERK activities seemed to be transient and complex, we constructed a mathematical simulation model for HRG-induced ErbB4 receptor signalling to explain the dynamics of the regulation mechanism in this signal transduction cascade. The model reflected well the experimental results observed in HRG-induced ErbB4 cells and in other modes of growth hormone-induced cell signalling that involve Raf-Akt cross-talk. The model suggested that HRG signalling is regulated by protein phosphatase 2A as well as Raf-Akt cross-talk, and protein phosphatase 2A modulates the kinase activity in both the PI3K-Akt and MAPK (mitogen-activated protein kinase) pathways. link: http://identifiers.org/pubmed/12691603

Parameters:

NameDescription
k_7 = 546.0; k7 = 60.0Reaction: RShP + GS => RShGS, Rate Law: compartment_0000001*(k7*RShP*GS-k_7*RShGS)
k1 = 0.0012; k_1 = 7.6E-4Reaction: R + HRG => RHRG, Rate Law: compartment_0000001*(k1*R*HRG-k_1*RHRG)
k_6 = 5.0; k6 = 20.0Reaction: RShc => RShP, Rate Law: compartment_0000001*(k6*RShc-k_6*RShP)
K22 = 60.0; K20 = 160.0; k20 = 0.3Reaction: ERKP => ERK; ERKPP, MKP3, Rate Law: compartment_0000001*k20*MKP3*ERKP/(K20*(1+ERKPP/K22)+ERKP)
V28 = 17000.0; K28 = 9.02Reaction: PIP3 => P_I, Rate Law: compartment_0000001*V28*PIP3/(K28+PIP3)
k17 = 2.9; K15 = 317.0; K17 = 317.0Reaction: MEKP => MEKPP; MEK, Rafstar, Rate Law: compartment_0000001*k17*Rafstar*MEKP/(K17*(1+MEK/K15)+MEKP)
k18 = 0.058; K16 = 2200.0; K33 = 12.0; K18 = 60.0; K31 = 4.35Reaction: MEKPP => MEKP; AktPIP, AktPIPP, PP2A, Rate Law: compartment_0000001*k18*PP2A*MEKPP/(K18*(1+MEKP/K16+AktPIPP/K31+AktPIPP/K33)+MEKPP)
K16 = 2200.0; K33 = 12.0; K18 = 60.0; k16 = 0.058; K31 = 4.35Reaction: MEKP => MEK; MEKPP, AktPIP, AktPIPP, PP2A, Rate Law: compartment_0000001*k16*PP2A*MEKP/(K16*(1+MEKPP/K18+AktPIP/K31+AktPIPP/K33)+MEKP)
k2 = 0.01; k_2 = 0.1Reaction: RHRG => RHRG2, Rate Law: compartment_0000001*(k2*RHRG^2-k_2*RHRG2)
K4 = 50.0; V4 = 62.5Reaction: RP => RHRG2, Rate Law: compartment_0000001*V4*RP/(K4+RP)
k29 = 507.0; k_29 = 234.0Reaction: PIP3 + Akt => AktPIP3, Rate Law: compartment_0000001*(k29*PIP3*Akt-k_29*AktPIP3)
K19 = 146000.0; K21 = 146000.0; k19 = 9.5Reaction: ERK => ERKP; MEKPP, Rate Law: compartment_0000001*k19*MEKPP*ERK/(K19*(1+ERKP/K21)+ERK)
K16 = 2200.0; K33 = 12.0; K18 = 60.0; k31 = 0.107; K31 = 4.35Reaction: AktPIP => AktPIP3; MEKP, MEKPP, AktPIPP, PP2A, Rate Law: compartment_0000001*k31*PP2A*AktPIP/(K31*(1+MEKP/K16+MEKPP/K18+AktPIPP/K33)+AktPIP)
k_9 = 0.0; k9 = 40.8Reaction: ShGS => GS + ShP, Rate Law: compartment_0000001*(k9*ShGS-k_9*GS*ShP)
K12 = 0.0571; V12 = 0.289Reaction: RasGTP => RasGDP, Rate Law: compartment_0000001*V12*RasGTP/(K12+RasGTP)
k8 = 2040.0; k_8 = 15700.0Reaction: RShGS => ShGS + RP, Rate Law: compartment_0000001*(k8*RShGS-k_8*ShGS*RP)
K26 = 3680.0; V26 = 2620.0Reaction: PI3Kstar => PI3K, Rate Law: compartment_0000001*V26*PI3Kstar/(K26+PI3Kstar)
K22 = 60.0; K20 = 160.0; k22 = 0.27Reaction: ERKPP => ERKP; MKP3, Rate Law: compartment_0000001*k22*MKP3*ERKPP/(K22*(1+ERKP/K20)+ERKPP)
V30 = 20000.0; K30 = 80000.0; K32 = 80000.0Reaction: AktPIP3 => AktPIP, Rate Law: compartment_0000001*V30*AktPIP3/(K30*(1+AktPIP/K32)+AktPIP3)
K33 = 12.0; K16 = 2200.0; K18 = 60.0; K31 = 4.35; k33 = 0.211Reaction: AktPIPP => AktPIP; MEKP, MEKPP, PP2A, Rate Law: compartment_0000001*k33*PP2A*AktPIPP/(K33*(1+MEKP/K16+MEKPP/K18+AktPIP/K31)+AktPIPP)
k_3 = 0.01; k3 = 1.0Reaction: RHRG2 => RP, Rate Law: compartment_0000001*(k3*RHRG2-k_3*RP)
k5 = 0.1; k_5 = 1.0Reaction: RP + Shc => RShc, Rate Law: compartment_0000001*(k5*RP*Shc-k_5*RShc)
K19 = 146000.0; k21 = 16.0; K21 = 146000.0Reaction: ERKP => ERKPP; MEKPP, ERK, Rate Law: compartment_0000001*k21*MEKPP*ERKP/(K21*(1+ERK/K19)+ERKP)
V10 = 0.0154; K10 = 340.0Reaction: ShP => Shc, Rate Law: compartment_0000001*V10*ShP/(K10+ShP)
k24 = 9.85; k_24 = 0.0985Reaction: RPI3K => RPI3Kstar, Rate Law: compartment_0000001*(k24*RPI3K-k_24*RPI3Kstar)
k11 = 0.222; K11 = 0.181Reaction: RasGDP => RasGTP; ShGS, Rate Law: compartment_0000001*k11*ShGS*RasGDP/(K11+RasGDP)
k13 = 1.53; K13 = 11.7Reaction: Raf => Rafstar; RasGTP, Rate Law: compartment_0000001*k13*RasGTP*Raf/(K13+Raf)
K30 = 80000.0; K32 = 80000.0; V32 = 20000.0Reaction: AktPIP => AktPIPP; AktPIP3, Rate Law: compartment_0000001*V32*AktPIP/(K32*(1+AktPIP3/K30)+AktPIP)
k15 = 3.5; K15 = 317.0; K17 = 317.0Reaction: MEK => MEKP; Rafstar, Rate Law: compartment_0000001*k15*Rafstar*MEK/(K15*(1+MEKP/K17)+MEK)
k_23 = 2.0; k23 = 0.1Reaction: RP + PI3K => RPI3K, Rate Law: compartment_0000001*(k23*RP*PI3K-k_23*RPI3K)
k34 = 0.001; k_34 = 0.0Reaction: RP => internalization, Rate Law: compartment_0000001*(k34*RP-k_34*internalization)
k_25 = 0.047; k25 = 45.8Reaction: RPI3Kstar => RP + PI3Kstar, Rate Law: compartment_0000001*(k25*RPI3Kstar-k_25*RP*PI3Kstar)
K27 = 39.1; k27 = 16.9Reaction: P_I => PIP3; PI3Kstar, Rate Law: compartment_0000001*k27*PI3Kstar*P_I/(K27+P_I)
k14 = 0.00673; K14 = 8.07Reaction: Rafstar => Raf; AktPIPP, E, Rate Law: compartment_0000001*k14*(AktPIPP+E)*Rafstar/(K14+Rafstar)

States:

NameDescription
Rafstar[Serine/threonine-protein kinase B-raf]
RHRG2[IPR002154; receptor complex]
Shc[IPR001452; IPR000980]
Akt[IPR015744]
MEKP[phosphate(3-); IPR003527]
ShGSShGS
RasGDP[GDP; IPR015592; IPR013753]
RP[receptor complex]
RShPRShP
AktPIPP[1-phosphatidyl-1D-myo-inositol 3,4-bisphosphate; IPR015744]
PIP3[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate]
RPI3K[receptor complex; phosphatidylinositol 3-kinase complex]
PI3Kstar[phosphatidylinositol 3-kinase complex]
RShGSRShGS
internalizationinternalization
AktPIP[phosphatidylinositol 3-phosphate; IPR015744]
MEK[IPR003527]
PI3K[phosphatidylinositol 3-kinase complex]
ERKPP[diphosphate(4-); IPR008349; diphosphate(4-); IPR008350]
P I[phosphatidylinositol 3-phosphate]
ERKP[phosphate(3-); IPR008349; phosphate(3-); IPR008350]
GSGS
RPI3Kstar[phosphatidylinositol 3-kinase complex; receptor complex]
AktPIP3[1-phosphatidyl-1D-myo-inositol 3,4,5-trisphosphate; IPR015744]
ShPShP
HRG[IPR002154]
RHRG[IPR002154; receptor complex]
MEKPP[diphosphate ion; IPR003527]
Raf[Serine/threonine-protein kinase B-raf]
RasGTP[GTP; IPR015592; IPR013753]
ERK[IPR003527; IPR008349; IPR008350]
RShcRShc
R[receptor complex; Receptor tyrosine-protein kinase erbB-4]

Hatzimanikatis1999-Regulation of the G1-S transition of the mammalian cell cycle.: MODEL2005070001v0.0.1

A mathematical model of regulation of the G1-S transition of the mammalian cell cycle has been formulated to organize av…

Details

A mathematical model of regulation of the G1-S transition of the mammalian cell cycle has been formulated to organize available experimental molecular-level information in a systematic quantitative framework and to evaluate the ability of this manifestation of current knowledge to calculate correctly experimentally observed phenotypes. This model includes nine components and includes cyclin-cdk complexes, a pocket protein (pRb), a transcription factor (E2F-1), and a cyclin-cdk complex inhibitor. Simulation of the model equations yields stable oscillatory solutions corresponding to cell proliferation and asymptotically stable solutions corresponding to cell cycle arrest (quiescence). Bifurcation analysis of the system suggests changes in the intracellular concentrations of either E2F or cyclin E can activate cell proliferation and that co-overexpression of these molecules can prevent cell proliferation. Further analysis suggests that the amount of inhibitor necessary to prevent cell proliferation is independent of the concentrations of cyclin E and E2F and depends only on the equilibrium ratio between the bound and unbound forms of the inhibitor to the complex. link: http://identifiers.org/pubmed/10550769

Haugh2004_hGH: MODEL0848676877v0.0.1

This a model from the article: Mathematical model of human growth hormone (hGH)-stimulated cell proliferation explains…

Details

Human growth hormone (hGH) is a therapeutically important endocrine factor that signals various cell types. Structurally and functionally, the interactions of hGH with its receptor have been resolved in fine detail, such that hGH and hGH receptor variants can be practically engineered by either random or rational approaches to achieve significant changes in the free energies of binding. A somewhat unique feature of hGH action is its homodimerization of two hGH receptors, which is required for intracellular signaling and stimulation of cell proliferation, yet the potencies of hGH mutants in cell-based assays rarely correlate with their overall receptor-binding avidities. Here, a mathematical model of hGH-stimulated cell signaling is posed, accounting not only for binding interactions at the cell surface but induction of receptor endocytosis and downregulation as well. Receptor internalization affects ligand potency by imposing a limit on the lifetime of an active receptor complex, irrespective of ligand-receptor binding properties. The model thus explains, in quantitative terms, the numerous published observations regarding hGH receptor agonism and antagonism and challenges the interpretations of previous studies that have not considered receptor trafficking as a central regulatory mechanism in hGH signaling. link: http://identifiers.org/pubmed/15458315

Haut1974_Pentose_Cycle_Rat: MODEL1004070000v0.0.1

This is the Unlabelled model as described in: Simulation of the Pentose Cycle in Lactating Rat Mammary Gland Haut M…

Details

A computer model representing the pentose cycle, the tricarboxylic acid cycle and glycolysis in slices of lactating rat mammary glands has been constructed. This model is based primarily on the studies, with radioactive chemicals, of Abraham & Chaikoff (1959) [although some of the discrepant data of Katz & Wals (1972) could be accommodated by changing one enzyme activity]. Data obtained by using [1-(14)C]-, [6-(14)C]- and [3,4-(14)C]-glucose were simulated, as well as data obtained by using unlabelled glucose (for which some new experimental data are presented). Much past work on the pentose cycle has been mainly concerned with the division of glucose flow between the pentose cycle and glycolysis, and has relied on the assumption that the system is in steady state (both labelled and unlabelled). This assumption may not apply to lactating rat mammary glands, since the model shows that the percentage flow through the shunt progressively decreased for the first 2h of a 3h experiment, and we were unable to construct a completely steady-state model. The model allows examination of many quantitative features of the system, especially the amount of material passing through key enzymes, some of which appear to be regulated by NADP(+) concentrations as proposed by McLean (1960). Supplementary information for this paper has been deposited as Supplementary Publication SUP 50023 at the British Museum (Lending Division) (formerly the National Lending Library for Science and Technology), Boston Spa, Yorks. LS23 7BQ, U.K., from whom copies can be obtained on the terms indicated in Biochem. J. (1973) 131, 5. link: http://identifiers.org/pubmed/4154746

Hayashi1999_NOSynth_Phospho: MODEL4780784080v0.0.1

This model features the phosphorylation of rat brain neuronal NOS expressed in E. coli or Sf9 cells, which leads to a de…

Details

Phosphorylation of neuronal nitric-oxide synthase (nNOS) by Ca2+/calmodulin (CaM)-dependent protein kinases (CaM kinases) including CaM kinase Ialpha (CaM-K Ialpha), CaM kinase IIalpha (CaM-K IIalpha), and CaM kinase IV (CaM-K IV), was studied. It was found that purified recombinant nNOS was phosphorylated by CaM-K Ialpha, CaM-K IIalpha, and CaM-K IV at Ser847 in vitro. Replacement of Ser847 with Ala (S847A) prevented phosphorylation by CaM kinases. Phosphorylated recombinant wild-type nNOS at Ser847 (approximately 0.5 mol of phosphate incorporation into nNOS) exhibited a 30% decrease of Vmax with little change of both the Km for L-arginine and Kact for CaM relative to unphosphorylated enzyme. The activity of mutant S847D was decreased to a level 50-60% as much as the wild-type enzyme. The decreased NOS enzyme activity of phosphorylated nNOS at Ser847 and mutant S847D was partially due to suppression of CaM binding, but not to impairment of dimer formation which is thought to be essential for enzyme activation. Inactive nNOS lacking CaM-binding ability was generated by mutation of Lys732-Lys-Leu to Asp732-Asp-Glu (Watanabe, Y., Hu, Y., and Hidaka, H. (1997) FEBS Lett. 403, 75-78). It was phosphorylated by CaM kinases, as was the wild-type enzyme, indicating that CaM-nNOS binding was not required for the phosphorylation reaction. We developed antibody NP847, which specifically recognize nNOS in its phosphorylated state at Ser847. Using the antibody NP847, we obtained evidence that nNOS is phosphorylated at Ser847 in rat brain. Thus, our results suggest that CaM kinase-induced phosphorylation of nNOS at Ser847 alters the activity control of this enzyme. link: http://identifiers.org/pubmed/10400690

Hayer2005_AMPAR_CaMKII_strong_coupling: MODEL9087255381v0.0.1

This is a model of tight coupling between the AMPAR trafficking bistability, and the CaMKII autophosphorylation bistabil…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

Hayer2005_AMPAR_CaMKII_weak_coupling: MODEL9087474843v0.0.1

This is a model of weak coupling between the AMPAR traffikcing bistability, and the CaMKII autophosphorylation bistabili…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

Hayer2005_AMPAR_traff_model0: MODEL9086207764v0.0.1

This is model 0 from Hayer and Bhalla, PLoS Comput Biol 2005. It has a bistable model of AMPAR traffic, plus a non-bista…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

Hayer2005_AMPAR_traff_model1: MODEL9086518048v0.0.1

This is the basic model of AMPAR trafficking bistability. It is based on Hayer and Bhalla, PLoS Comput. Biol. 2005. It i…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

Hayer2005_CaMKII_model3: MODEL9086953089v0.0.1

This is the complete model of CaMKII bistability, model 3. It exhibits bistability in CaMKII activation due to autophosp…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

Hayer2005_CaMKII_noPKA_model3: MODEL9086926384v0.0.1

This is the model of CaMKII bistability, model 3. It exhibits bistability in CaMKII activation due to autophosphorylatio…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

Hayer2005_simple_AMPAR_traff_model2: MODEL9086628127v0.0.1

This is a highly simplified model of the AMPAR trafficking cycle that exhibits bistability. It is model 2 from Hayer and…

Details

Changes in the synaptic connection strengths between neurons are believed to play a role in memory formation. An important mechanism for changing synaptic strength is through movement of neurotransmitter receptors and regulatory proteins to and from the synapse. Several activity-triggered biochemical events control these movements. Here we use computer models to explore how these putative memory-related changes can be stabilised long after the initial trigger, and beyond the lifetime of synaptic molecules. We base our models on published biochemical data and experiments on the activity-dependent movement of a glutamate receptor, AMPAR, and a calcium-dependent kinase, CaMKII. We find that both of these molecules participate in distinct bistable switches. These simulated switches are effective for long periods despite molecular turnover and biochemical fluctuations arising from the small numbers of molecules in the synapse. The AMPAR switch arises from a novel self-recruitment process where the presence of sufficient receptors biases the receptor movement cycle to insert still more receptors into the synapse. The CaMKII switch arises from autophosphorylation of the kinase. The switches may function in a tightly coupled manner, or relatively independently. The latter case leads to multiple stable states of the synapse. We propose that similar self-recruitment cycles may be important for maintaining levels of many molecules that undergo regulated movement, and that these may lead to combinatorial possible stable states of systems like the synapse. link: http://identifiers.org/pubmed/16110334

He2017 - A mathematical model of pancreatic cancer with two kinds of treatments: BIOMD0000000811v0.0.1

This is a mathematical model of pancreatic cancer which includes descriptions of regulatory T cell activity and inhibiti…

Details

In this paper, we investigate a mathematical model of pancreatic cancer, which extends the existing pancreatic cancer models with regulatory T cells (Tregs) and Treg inhibitory therapy. The model consists of tumor-immune interaction and immune suppression from Tregs. In the absence of treatments, we first characterize the system dynamics by locating equilibrium points and determining stability properties. Next, cytokine induced killer (CIK) immunotherapy is incorporated. Numerical simulations of prognostic results illustrate that the median overall survival associated with treatment can be prolonged approximately from 7 to 13 months, which is consistent with the clinical data. Furthermore, we consider cyclophosphamide (CTX) therapy as well as the combined therapy with CIK and CTX. Intensive simulation results suggest that both CTX therapy and the combined CIK/CTX therapy can reduce the number of Tregs and increase the overall survival (OS), but Tregs and tumor cells will gradually rise to equilibrium state as long as therapies are ceased. link: http://identifiers.org/doi/10.1142/S021833901750005X

Parameters:

NameDescription
r_e = 5.0E-12Reaction: => E_CD8; N_Killer, C_PCC, Rate Law: compartment*r_e*N_Killer*C_PCC
b_e = 0.02Reaction: E_CD8 =>, Rate Law: compartment*b_e*E_CD8
b_c = 1.5E-11Reaction: C_PCC => ; N_Killer, Rate Law: compartment*b_c*N_Killer*C_PCC
g_h = 0.3; tau_1_alpha_1 = 2.2483E11; p_h = 0.125Reaction: => H_T_Helper, Rate Law: compartment*p_h*H_T_Helper*H_T_Helper/(g_h*tau_1_alpha_1+H_T_Helper)
b_h = 0.0012Reaction: H_T_Helper =>, Rate Law: compartment*b_h*H_T_Helper
delta_e = 1.0E-10Reaction: E_CD8 => ; R_T_Regulatory, Rate Law: compartment*delta_e*R_T_Regulatory*E_CD8
gamma_2_tau_3 = 4.4691E-13; r_2 = 0.286; d_c = 7.87E-5; r_1 = 0.345Reaction: C_PCC => ; E_CD8, R_T_Regulatory, Rate Law: compartment*d_c*E_CD8*C_PCC/((1+r_1*R_T_Regulatory)*(1+r_2*gamma_2_tau_3*C_PCC))
a_n = 130000.0Reaction: => N_Killer, Rate Law: compartment*a_n
c_n = 1.0E-13Reaction: N_Killer => ; C_PCC, Rate Law: compartment*c_n*N_Killer*C_PCC
c_e = 3.42E-12Reaction: E_CD8 => ; C_PCC, Rate Law: compartment*c_e*E_CD8*C_PCC
beta_3_tau_2 = 4.4691E-13; f_n = 0.125; h_n = 0.3; beta_1_tau_2 = 4.4691E-13; beta_2_tau_2 = 4.4691E-13Reaction: => N_Killer; E_CD8, H_T_Helper, Rate Law: compartment*f_n*N_Killer*(beta_1_tau_2*E_CD8+beta_2_tau_2*N_Killer+beta_3_tau_2*H_T_Helper)/(h_n+beta_1_tau_2*E_CD8+beta_2_tau_2*N_Killer+beta_3_tau_2*H_T_Helper)
tau_1_alpha_1 = 2.2483E11; p_e = 0.125; g_e = 0.3Reaction: => E_CD8; H_T_Helper, Rate Law: compartment*p_e*H_T_Helper*E_CD8/(g_e*tau_1_alpha_1+H_T_Helper)
a_r = 2.0E-4Reaction: => R_T_Regulatory; E_CD8, Rate Law: compartment*a_r*E_CD8
lambda_p = 0.015Reaction: P_PSC =>, Rate Law: compartment*lambda_p*P_PSC
a_c = 1.02E-11; k_c = 0.0195; mu_c = 1.821414E-21Reaction: => C_PCC; P_PSC, Rate Law: compartment*(k_c+mu_c*P_PSC)*C_PCC*(1-a_c*C_PCC)
a = 560000.0Reaction: => R_T_Regulatory, Rate Law: compartment*a
tau_1_alpha_1 = 2.2483E11; g_r = 0.3; p_r = 0.125Reaction: => R_T_Regulatory; H_T_Helper, Rate Law: compartment*p_r*H_T_Helper*R_T_Regulatory/(g_r*tau_1_alpha_1+H_T_Helper)
a_h = 360000.0Reaction: => H_T_Helper, Rate Law: compartment*a_h
beta_3_tau_2 = 4.4691E-13; f_h = 0.125; h_h = 0.3; beta_1_tau_2 = 4.4691E-13; beta_2_tau_2 = 4.4691E-13Reaction: => H_T_Helper; E_CD8, N_Killer, Rate Law: compartment*f_h*H_T_Helper*(beta_1_tau_2*E_CD8+beta_2_tau_2*N_Killer+beta_3_tau_2*H_T_Helper)/(h_h+beta_1_tau_2*E_CD8+beta_2_tau_2*N_Killer+beta_3_tau_2*H_T_Helper)
delta_r = 0.023Reaction: R_T_Regulatory =>, Rate Law: compartment*delta_r*R_T_Regulatory
beta_3_tau_2 = 4.4691E-13; h_e = 0.3; f_e = 0.125; beta_1_tau_2 = 4.4691E-13; beta_2_tau_2 = 4.4691E-13Reaction: => E_CD8; N_Killer, H_T_Helper, Rate Law: compartment*f_e*E_CD8*(beta_1_tau_2*E_CD8+beta_2_tau_2*N_Killer+beta_3_tau_2*H_T_Helper)/(h_e+beta_1_tau_2*E_CD8+beta_2_tau_2*N_Killer+beta_3_tau_2*H_T_Helper)
r = 1.0E-11Reaction: R_T_Regulatory => ; N_Killer, Rate Law: compartment*r*N_Killer*R_T_Regulatory
k_p = 0.00195; f_p = 0.125; mu_p = 5.6E7; a_p = 1.7857E-9Reaction: => P_PSC; C_PCC, Rate Law: compartment*(k_p+f_p*C_PCC/(mu_p+C_PCC))*P_PSC*(1-a_p*P_PSC)
b_r = 4.0E-4Reaction: => R_T_Regulatory; H_T_Helper, Rate Law: compartment*b_r*H_T_Helper
g_n = 0.3; tau_1_alpha_1 = 2.2483E11; p_n = 0.125Reaction: => N_Killer; H_T_Helper, Rate Law: compartment*p_n*H_T_Helper*N_Killer/(g_n*tau_1_alpha_1+H_T_Helper)
a_e = 13000.0Reaction: => E_CD8, Rate Law: compartment*a_e
delta_h = 1.0E-10Reaction: H_T_Helper => ; R_T_Regulatory, Rate Law: compartment*delta_h*R_T_Regulatory*H_T_Helper
b_n = 0.015Reaction: N_Killer =>, Rate Law: compartment*b_n*N_Killer
delta_n = 1.0E-10Reaction: N_Killer =>, Rate Law: compartment*delta_n*N_Killer

States:

NameDescription
N Killer[natural killer cell]
P PSC[pancreatic stellate cell]
E CD8[CD8-Positive T-Lymphocyte]
H T Helper[helper T-lymphocyte]
R T Regulatory[regulatory T-lymphocyte]
C PCC[pancreatic cancer cell]

Heavner2012 - Metabolic Network of S.cerevisiae: MODEL1209060000v0.0.1

Heavner2012 - Metabolic Network of S.cerevisiaeThis SBML representation of the yeast metabolic network is made available…

Details

Efforts to improve the computational reconstruction of the Saccharomyces cerevisiae biochemical reaction network and to refine the stoichiometrically constrained metabolic models that can be derived from such a reconstruction have continued since the first stoichiometrically constrained yeast genome scale metabolic model was published in 2003. Continuing this ongoing process, we have constructed an update to the Yeast Consensus Reconstruction, Yeast 5. The Yeast Consensus Reconstruction is a product of efforts to forge a community-based reconstruction emphasizing standards compliance and biochemical accuracy via evidence-based selection of reactions. It draws upon models published by a variety of independent research groups as well as information obtained from biochemical databases and primary literature.Yeast 5 refines the biochemical reactions included in the reconstruction, particularly reactions involved in sphingolipid metabolism; updates gene-reaction annotations; and emphasizes the distinction between reconstruction and stoichiometrically constrained model. Although it was not a primary goal, this update also improves the accuracy of model prediction of viability and auxotrophy phenotypes and increases the number of epistatic interactions. This update maintains an emphasis on standards compliance, unambiguous metabolite naming, and computer-readable annotations available through a structured document format. Additionally, we have developed MATLAB scripts to evaluate the model's predictive accuracy and to demonstrate basic model applications such as simulating aerobic and anaerobic growth. These scripts, which provide an independent tool for evaluating the performance of various stoichiometrically constrained yeast metabolic models using flux balance analysis, are included as Additional files 1, 2 and 3.Yeast 5 expands and refines the computational reconstruction of yeast metabolism and improves the predictive accuracy of a stoichiometrically constrained yeast metabolic model. It differs from previous reconstructions and models by emphasizing the distinction between the yeast metabolic reconstruction and the stoichiometrically constrained model, and makes both available as Additional file 4 and Additional file 5 and at http://yeast.sf.net/ as separate systems biology markup language (SBML) files. Through this separation, we intend to make the modeling process more accessible, explicit, transparent, and reproducible. link: http://identifiers.org/pubmed/22663945

Heberle-Razquin Navas-2019 - PI3K-MAPK/p38-mTOR Model V: MODEL1902140002v0.0.1

Model V simulating stress induction with stress inputs on PI3K, Akt-pS473 and mTORC1. The model scheme is depicted in Fi…

Details

All cells and organisms exhibit stress-coping mechanisms to ensure survival. Cytoplasmic protein-RNA assemblies termed stress granules are increasingly recognized to promote cellular survival under stress. Thus, they might represent tumor vulnerabilities that are currently poorly explored. The translationinhibitory eIF2α kinases are established as main drivers of stress granule assembly. Using a systems approach, we identify the translation enhancers PI3K and MAPK/p38 as pro-stressgranule- kinases. They act through the metabolic master regulator mammalian target of rapamycin complex 1 (mTORC1) to promote stress granule assembly.When highly active, PI3K is the main driver of stress granules; however, the impact of p38 becomes apparent as PI3K activity declines. PI3K and p38 thus act in a hierarchical manner to drive mTORC1 activity and stress granule assembly. Of note, this signaling hierarchy is also present in human breast cancer tissue. Importantly, only the recognition of the PI3K-p38 hierarchy under stress enabled the discovery of p38’s role in stress granule formation. In summary, we assign a new prosurvival function to the key oncogenic kinases PI3K link: http://identifiers.org/doi/10.26508/lsa.201800257

HeberleRazquinNavas2019 - The PI3K and MAPK/p38 pathways control stress granuleassembly in a hierarchical manner model 2: MODEL2001030002v0.0.1

All cells and organisms exhibit stress-coping mechanisms to ensure survival. Cytoplasmic protein-RNA assemblies termed s…

Details

All cells and organisms exhibit stress-coping mechanisms to ensure survival. Cytoplasmic protein-RNA assemblies termed stress granules are increasingly recognized to promote cellular survival under stress. Thus, they might represent tumor vulnerabilities that are currently poorly explored. The translation-inhibitory eIF2α kinases are established as main drivers of stress granule assembly. Using a systems approach, we identify the translation enhancers PI3K and MAPK/p38 as pro-stress-granule-kinases. They act through the metabolic master regulator mammalian target of rapamycin complex 1 (mTORC1) to promote stress granule assembly. When highly active, PI3K is the main driver of stress granules; however, the impact of p38 becomes apparent as PI3K activity declines. PI3K and p38 thus act in a hierarchical manner to drive mTORC1 activity and stress granule assembly. Of note, this signaling hierarchy is also present in human breast cancer tissue. Importantly, only the recognition of the PI3K-p38 hierarchy under stress enabled the discovery of p38's role in stress granule formation. In summary, we assign a new pro-survival function to the key oncogenic kinases PI3K and p38, as they hierarchically promote stress granule formation. link: http://identifiers.org/pubmed/30923191

HeberleRazquinNavas2019 - The PI3K and MAPK/p38 pathways control stress granuleassembly in a hierarchical manner model 3: BIOMD0000000907v0.0.1

All cells and organisms exhibit stress-coping mechanisms toensure survival. Cytoplasmic protein-RNA assemblies termedstr…

Details

All cells and organisms exhibit stress-coping mechanisms to ensure survival. Cytoplasmic protein-RNA assemblies termed stress granules are increasingly recognized to promote cellular survival under stress. Thus, they might represent tumor vulnerabilities that are currently poorly explored. The translation-inhibitory eIF2α kinases are established as main drivers of stress granule assembly. Using a systems approach, we identify the translation enhancers PI3K and MAPK/p38 as pro-stress-granule-kinases. They act through the metabolic master regulator mammalian target of rapamycin complex 1 (mTORC1) to promote stress granule assembly. When highly active, PI3K is the main driver of stress granules; however, the impact of p38 becomes apparent as PI3K activity declines. PI3K and p38 thus act in a hierarchical manner to drive mTORC1 activity and stress granule assembly. Of note, this signaling hierarchy is also present in human breast cancer tissue. Importantly, only the recognition of the PI3K-p38 hierarchy under stress enabled the discovery of p38's role in stress granule formation. In summary, we assign a new pro-survival function to the key oncogenic kinases PI3K and p38, as they hierarchically promote stress granule formation. link: http://identifiers.org/pubmed/30923191

Parameters:

NameDescription
a1_X11_0 = 0.106711200647841; a2_X11_0 = 1.00000017501247E-5; a_X6_Y2 = 1.00000114154884E-5; b_X11_1 = 0.182804161260864; b_X11_2 = 0.224858434757367; Y2 = 1.0Reaction: => X11_3; X10_0, X10_1, X10_2, X10_3, X11_1, X11_2, X11_3, X5_0, X9_0, X9_2, Rate Law: default*(((X11_2*(Y2*a_X6_Y2+2*a2_X11_0*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1))-X11_3*b_X11_2)-X11_3*b_X11_1)+2*X11_1*X5_0*a1_X11_0)/default
ModelValue_114 = 0.996685919963556Reaction: fourEBP1_pT37_46_obs = ModelValue_114*X12_1, Rate Law: missing
a1_X11_0 = 0.106711200647841; a_X6_Y2 = 1.00000114154884E-5; b_X11_1 = 0.182804161260864; b_X11_2 = 0.224858434757367; Y2 = 1.0Reaction: => X11_1; X10_0, X10_1, X10_2, X10_3, X11_0, X11_1, X11_3, X5_0, X9_0, X9_2, Rate Law: default*(((X11_3*b_X11_2-X11_1*b_X11_1)+X11_0*(Y2*a_X6_Y2+a1_X11_0*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1)))-2*X11_1*X5_0*a1_X11_0)/default
b_X2_2 = 0.106214679132925; a1_X2_0 = 0.0014976539751451Reaction: => X2_0; X11_1, X11_3, X1_1, X2_0, X2_2, Rate Law: default*((X2_2*b_X2_2-X1_1*X2_0)-X2_0*a1_X2_0*(X11_1+X11_3))/default
Y5 = 0.0; a2_X8_0 = 0.210752496177883; k_stress_2 = 0.00999999977724154; Y3 = 1.0; b_X8_1 = 0.0462909157235242; b_X8_2 = 0.0100376101872374Reaction: => X8_2; X4_1, X5_1, X8_0, X8_2, X8_3, Rate Law: default*(((X8_3*b_X8_1-X8_2*b_X8_2)-X8_0*(0.83*Y5-1)*(X4_1*a2_X8_0+Y3*k_stress_2))+2*X5_1*X8_2*a2_X8_0*(0.83*Y5-1))/default
a_X6_Y2 = 1.00000114154884E-5; b_X12_1 = 0.0102134541960737; a_X12_0 = 0.198339568602839; Y2 = 1.0Reaction: => X12_0; X10_0, X10_1, X10_2, X10_3, X12_0, X12_1, X9_0, X9_2, Rate Law: default*(X12_1*b_X12_1-X12_0*(Y2*a_X6_Y2+a_X12_0*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1)))/default
Y5 = 0.0; a1_X8_0 = 0.584037889511307; a2_X8_0 = 0.210752496177883; k_stress_2 = 0.00999999977724154; Y3 = 1.0; b_X8_1 = 0.0462909157235242; b_X8_2 = 0.0100376101872374Reaction: => X8_0; X4_1, X5_1, X8_0, X8_1, X8_2, Rate Law: default*(X8_1*b_X8_1+X8_2*b_X8_2+X8_0*(0.83*Y5-1)*(X4_1*a2_X8_0+Y3*k_stress_2)+X5_1*X8_0*a1_X8_0*(0.83*Y5-1))/default
b_X5_1 = 0.077833118821602; a_X5_0 = 9.99999969718096Reaction: => X5_0; X4_1, X5_0, X5_1, Rate Law: default*(X5_1*b_X5_1-X4_1*X5_0*a_X5_0)/default
b_X10_2 = 0.011959597261903; b_X10_1 = 0.00263737900398121; a_X6_Y2 = 1.00000114154884E-5; a1_X10_0 = 1.04880466121365E-5; a2_X10_0 = 0.196797907822297; Y2 = 1.0Reaction: => X10_0; X10_0, X10_1, X10_2, X10_3, X8_1, X8_3, X9_0, X9_2, Rate Law: default*(((X10_1*b_X10_1+X10_2*b_X10_2)-X10_0*(Y2*a_X6_Y2+a1_X10_0*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1)))-X10_0*a2_X10_0*(X8_1+X8_3))/default
ModelValue_113 = 3.98428884870299Reaction: PRAS40_pS183_obs = ModelValue_113*X10_1+ModelValue_113*X10_3, Rate Law: missing
k_stress_1 = 9.99999999995476; Y3 = 1.0; Y4 = 1.0; b_X4_1 = 1.08358100911056E-5; a_X4_0 = 1.11095303548777E-4Reaction: => X4_0; X2_1, X4_0, X4_1, Rate Law: default*(X4_1*b_X4_1+X4_0*(X2_1*a_X4_0+Y3*k_stress_1)*(Y4-1))/default
ModelValue_116 = 74.7402331598434Reaction: p70_S6K_pT229_obs = ModelValue_116*X11_2+ModelValue_116*X11_3, Rate Law: missing
b_X10_2 = 0.011959597261903; b_X10_1 = 0.00263737900398121; a_X6_Y2 = 1.00000114154884E-5; a2_X10_0 = 0.196797907822297; a_X10_2 = 9.99999999991509; Y2 = 1.0Reaction: => X10_2; X10_0, X10_1, X10_2, X10_3, X8_1, X8_3, X9_0, X9_2, Rate Law: default*(((X10_3*b_X10_1-X10_2*b_X10_2)-X10_2*(Y2*a_X6_Y2+a_X10_2*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1)))+X10_0*a2_X10_0*(X8_1+X8_3))/default
ModelValue_117 = 997.421063173575Reaction: IRS1_pS636_639_obs = ModelValue_117*X2_2, Rate Law: missing
ModelValue_109 = 1.54625898449999Reaction: Akt_pT308_obs = ModelValue_109*X8_1+ModelValue_109*X8_3, Rate Law: missing
a2_X11_0 = 1.00000017501247E-5; a_X6_Y2 = 1.00000114154884E-5; b_X11_1 = 0.182804161260864; b_X11_2 = 0.224858434757367; Y2 = 1.0Reaction: => X11_2; X10_0, X10_1, X10_2, X10_3, X11_0, X11_2, X11_3, X5_0, X9_0, X9_2, Rate Law: default*(((X11_3*b_X11_1-X11_2*b_X11_2)-X11_2*(Y2*a_X6_Y2+2*a2_X11_0*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1)))+X11_0*X5_0*a2_X11_0)/default
ModelValue_112 = 10.1154012696402Reaction: PRAS40_pT246_obs = ModelValue_112*X10_2+ModelValue_112*X10_3, Rate Law: missing
a_X10_1 = 1.00000000000206E-5; b_X10_2 = 0.011959597261903; b_X10_1 = 0.00263737900398121; a_X6_Y2 = 1.00000114154884E-5; a1_X10_0 = 1.04880466121365E-5; Y2 = 1.0Reaction: => X10_1; X10_0, X10_1, X10_2, X10_3, X8_1, X8_3, X9_0, X9_2, Rate Law: default*(((X10_3*b_X10_2-X10_1*b_X10_1)+X10_0*(Y2*a_X6_Y2+a1_X10_0*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1)))-X10_1*a_X10_1*(X8_1+X8_3))/default
a1_X8_0 = 0.584037889511307; Y5 = 0.0; a2_X8_0 = 0.210752496177883; b_X8_1 = 0.0462909157235242; b_X8_2 = 0.0100376101872374Reaction: => X8_3; X4_1, X5_1, X8_1, X8_2, X8_3, Rate Law: default*((((-X8_3)*b_X8_1-X8_3*b_X8_2)-2*X4_1*X8_1*a1_X8_0*(0.83*Y5-1))-2*X5_1*X8_2*a2_X8_0*(0.83*Y5-1))/default
a1_X8_0 = 0.584037889511307; Y5 = 0.0; b_X8_1 = 0.0462909157235242; b_X8_2 = 0.0100376101872374Reaction: => X8_1; X4_1, X5_1, X8_0, X8_1, X8_3, Rate Law: default*(((X8_3*b_X8_2-X8_1*b_X8_1)+2*X4_1*X8_1*a1_X8_0*(0.83*Y5-1))-X5_1*X8_0*a1_X8_0*(0.83*Y5-1))/default
a2_X9_0 = 0.0216220006084923; b_X9_2 = 0.0369559223359753Reaction: => X9_0; X8_1, X8_3, X9_0, X9_2, Rate Law: default*(X9_2*b_X9_2-X9_0*a2_X9_0*(X8_1+X8_3))/default
ModelValue_115 = 86.0602161862265Reaction: p70_S6K_pT389_obs = ModelValue_115*X11_1+ModelValue_115*X11_3, Rate Law: missing
a_X10_1 = 1.00000000000206E-5; b_X10_2 = 0.011959597261903; a_X6_Y2 = 1.00000114154884E-5; b_X10_1 = 0.00263737900398121; a_X10_2 = 9.99999999991509; Y2 = 1.0Reaction: => X10_3; X10_0, X10_1, X10_2, X10_3, X8_1, X8_3, X9_0, X9_2, Rate Law: default*(((X10_2*(Y2*a_X6_Y2+a_X10_2*(X10_0/(X10_0+X10_1+X10_2+X10_3)-1)*(X9_0/(X9_0+X9_2)-1))-X10_3*b_X10_2)-X10_3*b_X10_1)+X10_1*a_X10_1*(X8_1+X8_3))/default
Y1 = 0.0Reaction: => X1_1; X1_0, X1_1, Rate Law: default*(X1_0*Y1-X1_1)/default
ModelValue_111 = 2.71349287061239Reaction: TSC1_TSC2_pT1462_obs = ModelValue_111*X9_2, Rate Law: missing
ModelValue_110 = 11.9261080736157Reaction: Akt_pS473_obs = ModelValue_110*X8_2+ModelValue_110*X8_3, Rate Law: missing

States:

NameDescription
X2 2X2_2
X8 2X8_2
X9 2X9_2
X1 1X1_1
X9 0X9_0
X1 0X1_0
X12 1X12_1
IRS1 pS636 639 obsIRS1_pS636-639_obs
TSC1 TSC2 pT1462 obsTSC1_TSC2_pT1462_obs
PRAS40 pT246 obsPRAS40_pT246_obs
X8 1X8_1
X10 0X10_0
X2 1X2_1
X8 3X8_3
X4 0X4_0
X10 2X10_2
X10 3X10_3
p70 S6K pT229 obsp70_S6K_pT229_obs
X5 0X5_0
Akt pS473 obsAkt_pS473_obs
X8 0X8_0
PRAS40 pS183 obsPRAS40_pS183_obs
X11 1X11_1
X4 1X4_1
X12 0X12_0
fourEBP1 pT37 46 obsfourEBP1_pT37_46_obs
X5 1X5_1
p70 S6K pT389 obsp70_S6K_pT389_obs
X10 1X10_1
X11 3X11_3
X2 0X2_0
X11 2X11_2
Akt pT308 obsAkt_pT308_obs
X11 0X11_0

Heiland2012_CircadianClock_C.reinhardtii: BIOMD0000000411v0.0.1

This model is from the article: Modeling temperature entrainment of circadian clocks using the Arrhenius equation and…

Details

Endogenous circadian rhythms allow living organisms to anticipate daily variations in their natural environment. Temperature regulation and entrainment mechanisms of circadian clocks are still poorly understood. To better understand the molecular basis of these processes, we built a mathematical model based on experimental data examining temperature regulation of the circadian RNA-binding protein CHLAMY1 from the unicellular green alga Chlamydomonas reinhardtii, simulating the effect of temperature on the rates by applying the Arrhenius equation. Using numerical simulations, we demonstrate that our model is temperature-compensated and can be entrained to temperature cycles of various length and amplitude. The range of periods that allow entrainment of the model depends on the shape of the temperature cycles and is larger for sinusoidal compared to rectangular temperature curves. We show that the response to temperature of protein (de)phosphorylation rates play a key role in facilitating temperature entrainment of the oscillator in Chlamydomonas reinhardtii. We systematically investigated the response of our model to single temperature pulses to explain experimentally observed phase response curves. link: http://identifiers.org/pubmed/23729908

Parameters:

NameDescription
T = 291.0; T2 = 296.0; k=0.4; parameter_3 = 8.31447; v=2.6; h=2.0; parameter_7 = 84000.0Reaction: s2 => s9; s11, Rate Law: default*v*exp(parameter_7/parameter_3*(T2-T)/(T*T2))/(k+s11^h)
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; v=3.0; parameter_6 = 50000.0; Km=2.0Reaction: s9 => species_12, Rate Law: default*v*exp(parameter_6/parameter_3*(T2-T)/(T*T2))*s9/(Km+s9)
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; E=67000.0; v=30.0; Km=2.0Reaction: species_1 => species_12, Rate Law: default*v*exp(E/parameter_3*(T2-T)/(T*T2))*species_1/(Km+species_1)
T = 291.0; T2 = 296.0; parameter_1 = 1.0; parameter_3 = 8.31447; parameter_9 = 60000.0Reaction: species_1 => species_3, Rate Law: default*parameter_1*exp(parameter_9/parameter_3*(T2-T)/(T*T2))*species_1
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; E=67000.0; v=20.0; Km=4.0Reaction: species_4 => species_12, Rate Law: default*v*exp(E/parameter_3*(T2-T)/(T*T2))*species_4/(Km+species_4)
T = 291.0; T2 = 296.0; v=2.2; parameter_3 = 8.31447; parameter_6 = 50000.0; Km=0.2Reaction: s10 => species_12, Rate Law: default*v*exp(parameter_6/parameter_3*(T2-T)/(T*T2))*s10/(Km+s10)
T = 291.0; T2 = 296.0; parameter_10 = 67000.0; parameter_3 = 8.31447; parameter_2 = 0.5Reaction: species_3 => species_1, Rate Law: default*parameter_2*exp(parameter_10/parameter_3*(T2-T)/(T*T2))*species_3
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; v=10.0; parameter_7 = 84000.0; a=2.0Reaction: species_3 + s11 => species_4, Rate Law: default*v*exp(parameter_7/parameter_3*(T2-T)/(T*T2))*species_3*s11^a
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; E=67000.0; parameter_8 = 1.0; Km=1.0Reaction: species_3 => species_12, Rate Law: default*parameter_8*exp(E/parameter_3*(T2-T)/(T*T2))*species_3/(Km+species_3)
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; v=0.1; parameter_6 = 50000.0Reaction: s10 => s11, Rate Law: default*v*exp(parameter_6/parameter_3*(T2-T)/(T*T2))*s10
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; E=67000.0; v=19.0Reaction: species_2 => species_1, Rate Law: default*v*exp(E/parameter_3*(T2-T)/(T*T2))*species_2
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; parameter_6 = 50000.0; v=1.5; Km=1.4Reaction: s11 => species_12, Rate Law: default*v*exp(parameter_6/parameter_3*(T2-T)/(T*T2))*s11/(Km+s11)
T = 291.0; T2 = 296.0; parameter_3 = 8.31447; parameter_7 = 84000.0; v=5.0Reaction: s13 => s10; s9, Rate Law: default*v*exp(parameter_7/parameter_3*(T2-T)/(T*T2))*s9

States:

NameDescription
species 2[RNA binding protein]
s11[RNA binding protein; Phosphoprotein]
s13[RNA binding protein]
species 1[RNA binding protein]
species 3[RNA binding protein; Phosphoprotein]
s2[RNA binding protein]
species 4[RNA binding protein; RNA binding protein]
s9[RNA binding protein]
species 12junk
s10[RNA binding protein]

Heiland2019 - NAD pathway model analysing the impact of NNMT on pathway dynamics and evolution: MODEL1905220001v0.0.1

This is a detailed model of NAD biosynthesis and consumption representing pathway variances across kingdoms.

Details

Nicotinamide adenine dinucleotide (NAD) provides an important link between metabolism and signal transduction and has emerged as central hub between bioenergetics and all major cellular events. NAD-dependent signaling (e.g., by sirtuins and poly–adenosine diphosphate [ADP] ribose polymerases [PARPs]) consumes considerable amounts of NAD. To maintain physiological functions, NAD consumption and biosynthesis need to be carefully balanced. Using extensive phylogenetic analyses, mathematical modeling of NAD metabolism, and experimental verification, we show that the diversification of NAD-dependent signaling in vertebrates depended on 3 critical evolutionary events: 1) the transition of NAD biosynthesis to exclusive usage of nicotinamide phosphoribosyltransferase (NamPT); 2) the occurrence of nicotinamide N-methyltransferase (NNMT), which diverts nicotinamide (Nam) from recycling into NAD, preventing Nam accumulation and inhibition of NAD-dependent signaling reactions; and 3) structural adaptation of NamPT, providing an unusually high affinity toward Nam, necessary to maintain NAD levels. Our results reveal an unexpected coevolution and kinetic interplay between NNMT and NamPT that enables extensive NAD signaling. This has implications for therapeutic strategies of NAD supplementation and the use of NNMT or NamPT inhibitors in disease treatment. link: http://identifiers.org/doi/10.1073/pnas.1902346116

Heiland2019 - Two compartment model of NAD biosynthesis and consumption: MODEL1905220002v0.0.1

The model is based on MODEL1905220001 but has two compartments that have different composition of the biosynthetic enzym…

Details

Nicotinamide adenine dinucleotide (NAD) provides an important link between metabolism and signal transduction and has emerged as central hub between bioenergetics and all major cellular events. NAD-dependent signaling (e.g., by sirtuins and poly–adenosine diphosphate [ADP] ribose polymerases [PARPs]) consumes considerable amounts of NAD. To maintain physiological functions, NAD consumption and biosynthesis need to be carefully balanced. Using extensive phylogenetic analyses, mathematical modeling of NAD metabolism, and experimental verification, we show that the diversification of NAD-dependent signaling in vertebrates depended on 3 critical evolutionary events: 1) the transition of NAD biosynthesis to exclusive usage of nicotinamide phosphoribosyltransferase (NamPT); 2) the occurrence of nicotinamide N-methyltransferase (NNMT), which diverts nicotinamide (Nam) from recycling into NAD, preventing Nam accumulation and inhibition of NAD-dependent signaling reactions; and 3) structural adaptation of NamPT, providing an unusually high affinity toward Nam, necessary to maintain NAD levels. Our results reveal an unexpected coevolution and kinetic interplay between NNMT and NamPT that enables extensive NAD signaling. This has implications for therapeutic strategies of NAD supplementation and the use of NNMT or NamPT inhibitors in disease treatment. link: http://identifiers.org/doi/10.1073/pnas.1902346116

Heinemann2005 - Genome-scale reconstruction of Staphylococcus aureus (iMH551): MODEL1507180072v0.0.1

Heinemann2005 - Genome-scale reconstruction of Staphylococcus aureus (iMH551)This model is described in the article: [I…

Details

A genome-scale metabolic model of the Gram-positive, facultative anaerobic opportunistic pathogen Staphylococcus aureus N315 was constructed based on current genomic data, literature, and physiological information. The model comprises 774 metabolic processes representing approximately 23% of all protein-coding regions. The model was extensively validated against experimental observations and it correctly predicted main physiological properties of the wild-type strain, such as aerobic and anaerobic respiration and fermentation. Due to the frequent involvement of S. aureus in hospital-acquired bacterial infections combined with its increasing antibiotic resistance, we also investigated the clinically relevant phenotype of small colony variants and found that the model predictions agreed with recent findings of proteome analyses. This indicates that the model is useful in assisting future experiments to elucidate the interrelationship of bacterial metabolism and resistance. To help directing future studies for novel chemotherapeutic targets, we conducted a large-scale in silico gene deletion study that identified 158 essential intracellular reactions. A more detailed analysis showed that the biosynthesis of glycans and lipids is rather rigid with respect to circumventing gene deletions, which should make these areas particularly interesting for antibiotic development. The combination of this stoichiometric model with transcriptomic and proteomic data should allow a new quality in the analysis of clinically relevant organisms and a more rationalized system-level search for novel drug targets. link: http://identifiers.org/pubmed/16155945

Heinze1998_GnRH_LH: MODEL0848507209v0.0.1

This a model from the article: A mathematical model of luteinizing hormone release from ovine pituitary cells in perif…

Details

We model the effect of gonadotropin-releasing hormone (GnRH) on the production of luteinizing hormone (LH) by the ovine pituitary. GnRH, released by the hypothalamus, stimulates the secretion of LH from the pituitary. If stimulus pulses are regular, LH response will follow a similar pattern. However, during application of GnRH at high frequencies or concentrations or with continuous application, the pituitary delivers a decreased release of LH (termed desensitization). The proposed mathematical model consists of a system of nonlinear differential equations and incorporates two possible mechanisms to account for this observed behavior: desensitized receptor and limited, available LH. Desensitization was provoked experimentally in vitro by using ovine pituitary cells in a perifusion system. The model was fit to resulting experimental data by using maximum-likelihood estimation. Consideration of smaller models revealed that the desensitized receptor is significant. Limited, available LH was significant in three of four chambers. Throughout, the proposed model was in excellent agreement with experimental data. link: http://identifiers.org/pubmed/9843750

Heitzler2012 - GPCR signalling: BIOMD0000000842v0.0.1

This model is from the article: Competing G protein-coupled receptor kinases balance G protein and β-arrestin signalin…

Details

Seven-transmembrane receptors (7TMRs) are involved in nearly all aspects of chemical communications and represent major drug targets. 7TMRs transmit their signals not only via heterotrimeric G proteins but also through β-arrestins, whose recruitment to the activated receptor is regulated by G protein-coupled receptor kinases (GRKs). In this paper, we combined experimental approaches with computational modeling to decipher the molecular mechanisms as well as the hidden dynamics governing extracellular signal-regulated kinase (ERK) activation by the angiotensin II type 1A receptor (AT(1A)R) in human embryonic kidney (HEK)293 cells. We built an abstracted ordinary differential equations (ODE)-based model that captured the available knowledge and experimental data. We inferred the unknown parameters by simultaneously fitting experimental data generated in both control and perturbed conditions. We demonstrate that, in addition to its well-established function in the desensitization of G-protein activation, GRK2 exerts a strong negative effect on β-arrestin-dependent signaling through its competition with GRK5 and 6 for receptor phosphorylation. Importantly, we experimentally confirmed the validity of this novel GRK2-dependent mechanism in both primary vascular smooth muscle cells naturally expressing the AT(1A)R, and HEK293 cells expressing other 7TMRs. link: http://identifiers.org/pubmed/22735336

Parameters:

NameDescription
k14 = 0.0311Reaction: Hbarr2RP1 => barr2 + HRP1; Hbarr2RP1, Rate Law: compartmentOne*k14*Hbarr2RP1/compartmentOne
k8 = 1.77Reaction: PKC_a => PKC; PKC_a, Rate Law: compartmentOne*k8*PKC_a/compartmentOne
k24 = 0.347Reaction: Hbarr2RP2 => barr2 + HRP2; Hbarr2RP2, Rate Law: compartmentOne*k24*Hbarr2RP2/compartmentOne
k18 = 0.59; GRK56 = 1.5180818Reaction: HR => HRP2; GRK5_6, Rate Law: compartmentOne*k18*GRK56*HR/compartmentOne
k5 = 2.65Reaction: ERK + PKC_a => GpERK + PKC_a; ERK, PKC_a, Rate Law: compartmentOne*k5*ERK*PKC_a/compartmentOne
k23 = 1.05Reaction: HRbarr2 => barr2 + HR; HRbarr2, Rate Law: compartmentOne*k23*HRbarr2/compartmentOne
k12 = 2.59Reaction: barr2 + HRP1 => Hbarr2RP1; barr2, HRP1, Rate Law: compartmentOne*k12*barr2*HRP1/compartmentOne
k21 = 4.2E-4Reaction: ERK + HRbarr2 => bpERK + HRbarr2; ERK, HRbarr2, Rate Law: compartmentOne*k21*ERK*HRbarr2/compartmentOne
k25 = 0.762Reaction: bpERK => ERK; bpERK, Rate Law: compartmentOne*k25*bpERK/compartmentOne
k22 = 14.44Reaction: ERK + Hbarr2RP2 => bpERK + Hbarr2RP2; ERK, Hbarr2RP2, Rate Law: compartmentOne*k22*ERK*Hbarr2RP2/compartmentOne
k3 = 4.63Reaction: G_a + PIP2 => DAG + G_a; G_a, PIP2, Rate Law: compartmentOne*k3*G_a*PIP2/compartmentOne
k17 = 0.0665Reaction: HRP2 => HR; HRP2, Rate Law: compartmentOne*k17*HRP2/compartmentOne
k0 = 3.11E-4Reaction: G => G_a; G, Rate Law: compartmentOne*k0*G/compartmentOne
k2 = 7.6Reaction: G + HR => G_a + HR; G, HR, Rate Law: compartmentOne*k2*G*HR/compartmentOne
k1 = 0.0178Reaction: G + HRP1 => G_a + HRP1; G, HRP1, Rate Law: compartmentOne*k1*G*HRP1/compartmentOne
k16 = 0.0723Reaction: Hbarr2RP1 => barr2 + HR; Hbarr2RP1, Rate Law: compartmentOne*k16*Hbarr2RP1/compartmentOne
k9 = 3.04Reaction: GpERK => ERK; GpERK, Rate Law: compartmentOne*k9*GpERK/compartmentOne
k20 = 1.04Reaction: barr2 + HRP2 => Hbarr2RP2; barr2, HRP2, Rate Law: compartmentOne*k20*barr2*HRP2/compartmentOne
k11 = 2.61Reaction: barr1 + HRP1 => Hbarr1RP1; barr1, HRP1, Rate Law: compartmentOne*k11*barr1*HRP1/compartmentOne
GRK23 = 0.899447579; k10 = 2.27Reaction: HR => HRP1; GRK2_3, Rate Law: compartmentOne*k10*GRK23*HR/compartmentOne
k6 = 5.0985Reaction: G_a => G; G_a, Rate Law: compartmentOne*k6*G_a/compartmentOne
k7 = 0.461Reaction: DAG => PIP2; DAG, Rate Law: compartmentOne*k7*DAG/compartmentOne
k4 = 0.0787Reaction: DAG + PKC => DAG + PKC_a; DAG, PKC, Rate Law: compartmentOne*k4*DAG*PKC/compartmentOne
k19 = 0.205Reaction: barr2 + HR => HRbarr2; barr2, HR, Rate Law: compartmentOne*k19*barr2*HR/compartmentOne
k15 = 6.54E-5Reaction: Hbarr1RP1 => barr1 + HR; Hbarr1RP1, Rate Law: compartmentOne*k15*Hbarr1RP1/compartmentOne
k13 = 0.00619Reaction: Hbarr1RP1 => barr1 + HRP1; Hbarr1RP1, Rate Law: compartmentOne*k13*Hbarr1RP1/compartmentOne

States:

NameDescription
DAG[CDP-diacylglycerol]
GpERK[Mitogen-activated protein kinase 3; phosphorylated]
HR[P01019; P30556]
PIP2[CHEBI:83417]
barr2[Beta-arrestin-2]
G[Guanine nucleotide-binding protein G(s) subunit alpha isoforms short]
barr1[P49407]
Hbarr2RP2[P30556; P01019; Beta-arrestin-2; phosphorylated]
Hbarr2RP1[P01019; P30556; Beta-arrestin-2; phosphorylated]
HRbarr2[P01019; P30556; Beta-arrestin-2]
G a[Guanine nucleotide-binding protein G(s) subunit alpha isoforms short; active]
pERK[Mitogen-activated protein kinase 3; phosphorylated]
bpERK[Mitogen-activated protein kinase 3; phosphorylated]
ERK[Mitogen-activated protein kinase 3]
HRP1[P30556; P01019; phosphorylated]
Hbarr1RP1[P30556; P49407; P01019; phosphorylated]
PKC a[Protein kinase C alpha type; active]
HRP2[P01019; P30556; phosphorylated]
PKC[Protein kinase C alpha type]

Heldt2002_OrthostaticStress_circpbpk: MODEL1006230084v0.0.1

This a model from the article: Computational modeling of cardiovascular response to orthostatic stress. Heldt T, Shi…

Details

The objective of this study is to develop a model of the cardiovascular system capable of simulating the short-term (< or = 5 min) transient and steady-state hemodynamic responses to head-up tilt and lower body negative pressure. The model consists of a closed-loop lumped-parameter representation of the circulation connected to set-point models of the arterial and cardiopulmonary baroreflexes. Model parameters are largely based on literature values. Model verification was performed by comparing the simulation output under baseline conditions and at different levels of orthostatic stress to sets of population-averaged hemodynamic data reported in the literature. On the basis of experimental evidence, we adjusted some model parameters to simulate experimental data. Orthostatic stress simulations are not statistically different from experimental data (two-sided test of significance with Bonferroni adjustment for multiple comparisons). Transient response characteristics of heart rate to tilt also compare well with reported data. A case study is presented on how the model is intended to be used in the future to investigate the effects of post-spaceflight orthostatic intolerance. link: http://identifiers.org/pubmed/11842064

Heldt2002_OrthostaticStress_heart: MODEL1006230103v0.0.1

This a model from the article: Computational modeling of cardiovascular response to orthostatic stress. Heldt T, Shi…

Details

The objective of this study is to develop a model of the cardiovascular system capable of simulating the short-term (< or = 5 min) transient and steady-state hemodynamic responses to head-up tilt and lower body negative pressure. The model consists of a closed-loop lumped-parameter representation of the circulation connected to set-point models of the arterial and cardiopulmonary baroreflexes. Model parameters are largely based on literature values. Model verification was performed by comparing the simulation output under baseline conditions and at different levels of orthostatic stress to sets of population-averaged hemodynamic data reported in the literature. On the basis of experimental evidence, we adjusted some model parameters to simulate experimental data. Orthostatic stress simulations are not statistically different from experimental data (two-sided test of significance with Bonferroni adjustment for multiple comparisons). Transient response characteristics of heart rate to tilt also compare well with reported data. A case study is presented on how the model is intended to be used in the future to investigate the effects of post-spaceflight orthostatic intolerance. link: http://identifiers.org/pubmed/11842064

Heldt2002_OrthostaticStress_lpc: MODEL1006230113v0.0.1

This a model from the article: Computational modeling of cardiovascular response to orthostatic stress. Heldt T, Shi…

Details

The objective of this study is to develop a model of the cardiovascular system capable of simulating the short-term (< or = 5 min) transient and steady-state hemodynamic responses to head-up tilt and lower body negative pressure. The model consists of a closed-loop lumped-parameter representation of the circulation connected to set-point models of the arterial and cardiopulmonary baroreflexes. Model parameters are largely based on literature values. Model verification was performed by comparing the simulation output under baseline conditions and at different levels of orthostatic stress to sets of population-averaged hemodynamic data reported in the literature. On the basis of experimental evidence, we adjusted some model parameters to simulate experimental data. Orthostatic stress simulations are not statistically different from experimental data (two-sided test of significance with Bonferroni adjustment for multiple comparisons). Transient response characteristics of heart rate to tilt also compare well with reported data. A case study is presented on how the model is intended to be used in the future to investigate the effects of post-spaceflight orthostatic intolerance. link: http://identifiers.org/pubmed/11842064

Heldt2012 - Influenza Virus Replication: BIOMD0000000463v0.0.1

Heldt2012 - Influenza Virus ReplicationThe model describes the life cycle of influenza A virus in a mammalian cell inclu…

Details

Influenza viruses transcribe and replicate their negative-sense RNA genome inside the nucleus of host cells via three viral RNA species. In the course of an infection, these RNAs show distinct dynamics, suggesting that differential regulation takes place. To investigate this regulation in a systematic way, we developed a mathematical model of influenza virus infection at the level of a single mammalian cell. It accounts for key steps of the viral life cycle, from virus entry to progeny virion release, while focusing in particular on the molecular mechanisms that control viral transcription and replication. We therefore explicitly consider the nuclear export of viral genome copies (vRNPs) and a recent hypothesis proposing that replicative intermediates (cRNA) are stabilized by the viral polymerase complex and the nucleoprotein (NP). Together, both mechanisms allow the model to capture a variety of published data sets at an unprecedented level of detail. Our findings provide theoretical support for an early regulation of replication by cRNA stabilization. However, they also suggest that the matrix protein 1 (M1) controls viral RNA levels in the late phase of infection as part of its role during the nuclear export of viral genome copies. Moreover, simulations show an accumulation of viral proteins and RNA toward the end of infection, indicating that transport processes or budding limits virion release. Thus, our mathematical model provides an ideal platform for a systematic and quantitative evaluation of influenza virus replication and its complex regulation. link: http://identifiers.org/pubmed/22593159

Parameters:

NameDescription
parameter_17 = 1.0Reaction: species_28 + species_29 + species_30 => species_11; species_28, species_29, species_30, Rate Law: compartment_1*parameter_17*species_28*species_29*species_30
parameter_13 = 3.01E-4Reaction: species_19 + species_13 => species_9; species_19, species_13, Rate Law: compartment_1*parameter_13*species_19*species_13
parameter_43 = 20.2922077922078Reaction: species_9 => species_9 + species_24; species_9, Rate Law: compartment_1*parameter_43*species_9
parameter_20 = 0.09Reaction: species_9 => ; species_9, Rate Law: compartment_1*parameter_20*species_9
parameter_40 = 13.4698275862069Reaction: species_9 => species_9 + species_21; species_9, Rate Law: compartment_1*parameter_40*species_9
parameter_8 = 6.0Reaction: species_8 => species_9; species_8, Rate Law: compartment_1*parameter_8*species_8
parameter_10 = 13.86Reaction: species_17 => species_17 + species_18; species_17, Rate Law: compartment_1*parameter_10*species_17
parameter_28 = 8.1Reaction: species_26 => species_26 + species_34; species_26, Rate Law: compartment_1*parameter_28*species_26
parameter_14 = 1.39E-6Reaction: species_9 + species_14 => species_15; species_9, species_14, Rate Law: compartment_1*parameter_14*species_9*species_14
parameter_21 = 0.33Reaction: species_22 => ; species_22, Rate Law: compartment_1*parameter_21*species_22
parameter_15 = 1.0E-6Reaction: species_15 + species_31 => species_16; species_15, species_31, Rate Law: compartment_1*parameter_15*species_15*species_31
parameter_16 = 405.0Reaction: species_22 => species_22 + species_30; species_22, Rate Law: compartment_1*parameter_16*species_22
parameter_11 = 1.38Reaction: species_9 => species_9 + species_10; species_9, Rate Law: compartment_1*parameter_11*species_9
parameter_29 = 50.625Reaction: species_27 => species_27 + species_31; species_27, Rate Law: compartment_1*parameter_29*species_27
parameter_27 = 396.9Reaction: species_26 => species_26 + species_14; species_26, Rate Law: compartment_1*parameter_27*species_26
parameter_39 = 13.4698275862069Reaction: species_9 => species_9 + species_20; species_9, Rate Law: compartment_1*parameter_39*species_9
parameter_19 = 36.36Reaction: species_10 => ; species_10, Rate Law: compartment_1*parameter_19*species_10
parameter_2 = 4.55E-4; parameter_4 = 5.46218487394958Reaction: species_3 + species_4 => species_5; species_3, species_4, species_5, Rate Law: compartment_1*(parameter_2*species_3*species_4-parameter_4*species_5)
parameter_22 = 4.25Reaction: species_19 => ; species_19, Rate Law: compartment_1*parameter_22*species_19
parameter_7 = 3.08411764705882Reaction: species_6 => ; species_6, Rate Law: compartment_1*parameter_7*species_6
parameter_45 = 31.0945273631841Reaction: species_9 => species_9 + species_26; species_9, Rate Law: compartment_1*parameter_45*species_9
parameter_6 = 3.21Reaction: species_6 => species_7 + species_8; species_6, Rate Law: compartment_1*parameter_6*species_6
parameter_44 = 22.4497126436782Reaction: species_9 => species_9 + species_25; species_9, Rate Law: compartment_1*parameter_44*species_9
parameter_1 = 0.0809; parameter_3 = 7.15929203539823Reaction: species_3 + species_1 => species_2; species_3, species_1, species_2, Rate Law: compartment_1*(parameter_1*species_3*species_1-parameter_3*species_2)
parameter_42 = 17.7859988616961Reaction: species_9 => species_9 + species_23; species_9, Rate Law: compartment_1*parameter_42*species_9
parameter_46 = 36.0023041474654Reaction: species_9 => species_9 + species_27; species_9, Rate Law: compartment_1*parameter_46*species_9
parameter_12 = 1.0Reaction: species_18 + species_11 => species_19; species_18, species_11, Rate Law: compartment_1*parameter_12*species_18*species_11
parameter_41 = 14.1338760741746Reaction: species_9 => species_9 + species_22; species_9, Rate Law: compartment_1*parameter_41*species_9
KmB=450.0; KmE=1000.0; KmC=5000.0; KmD=10000.0; KmF=30000.0; parameter_18 = 0.0037; KmH=1650.0; KmG=400.0Reaction: species_16 + species_11 + species_13 + species_14 + species_31 + species_32 + species_33 + species_34 => species_35; species_16, species_11, species_32, species_13, species_33, species_14, species_34, species_31, Rate Law: compartment_1*parameter_18*species_16*species_11*species_32*species_13*species_33*species_14*species_34*species_31/((species_11+KmB)*(species_32+KmC)*(species_13+KmD)*(species_33+KmE)*(species_14+KmF)*(species_34+KmG)*(species_31+KmH))
parameter_5 = 4.8Reaction: species_5 => species_6 + species_4; species_5, Rate Law: compartment_1*parameter_5*species_5

States:

NameDescription
species 9[nucleus; intracellular ribonucleoprotein complex]
species 27Rm8
species 31[Nuclear export protein]
species 1[sialic acid]
species 20Rm1
species 4Blo
species 16[cytoplasm; Matrix protein 1; intracellular ribonucleoprotein complex]
species 18[Influenza A virus (strain A/Puerto Rico/8/1934 H1N1)]
species 28[RNA-directed RNA polymerase catalytic subunit]
species 39total vRNA of a segment
species 34[Matrix protein 2Matrix protein 2]
species 21Rm2
species 8[cytoplasm; intracellular ribonucleoprotein complex]
species 17[intracellular ribonucleoprotein complex]
species 12[RNA viral genome; viral RNA-directed RNA polymerase complex]
species 25Rm6
species 5[virion attachment to host cell; virion]
species 15[nucleus; Matrix protein 1; intracellular ribonucleoprotein complex]
species 29[Polymerase basic protein 2]
species 2[virion attachment to host cell; virion]
species 30[Polymerase acidic protein]
species 6[endosome; virion]
species 38total vRNA
species 19[RNA viral genome; viral RNA-directed RNA polymerase complex]
species 10[RNA viral genome; transcription, RNA-templated]
species 33[Neuraminidase]
species 11[viral RNA-directed RNA polymerase complex]
species 24Rm5
species 14[Matrix protein 1]
species 22Rm3
species 3[extracellular region; virion]
species 23Rm4
species 7[fusion of virus membrane with host endosome membrane; viral membrane; endosome membrane]
species 26Rm7
species 13[Nucleoprotein]

Heldt2018 - Budding yeast size control by titration of nuclear sites: MODEL1803220002v0.0.1

This model is decribed in the article: Dilution and titration of cell-cycle regulators may control cell size in budding…

Details

The size of a cell sets the scale for all biochemical processes within it, thereby affecting cellular fitness and survival. Hence, cell size needs to be kept within certain limits and relatively constant over multiple generations. However, how cells measure their size and use this information to regulate growth and division remains controversial. Here, we present two mechanistic mathematical models of the budding yeast (S. cerevisiae) cell cycle to investigate competing hypotheses on size control: inhibitor dilution and titration of nuclear sites. Our results suggest that an inhibitor-dilution mechanism, in which cell growth dilutes the transcriptional inhibitor Whi5 against the constant activator Cln3, can facilitate size homeostasis. This is achieved by utilising a positive feedback loop to establish a fixed size threshold for the START transition, which efficiently couples cell growth to cell cycle progression. Yet, we show that inhibitor dilution cannot reproduce the size of mutants that alter the cell’s overall ploidy and WHI5 gene copy number. By contrast, size control through titration of Cln3 against a constant number of genomic binding sites for the transcription factor SBF recapitulates both size homeostasis and the size of these mutant strains. Moreover, this model produces an imperfect ‘sizer’ behaviour in G1 and a ‘timer’ in S/G2/M, which combine to yield an ‘adder’ over the whole cell cycle; an observation recently made in experiments. Hence, our model connects these phenomenological data with the molecular details of the cell cycle, providing a systems-level perspective of budding yeast size control. link: http://identifiers.org/doi/10.1371/journal.pcbi.1006548

Heldt2018 - Proliferation-quiescence decision in response to DNA damage: BIOMD0000000700v0.0.1

Heldt2018 - Proliferation-quiescence decision in response to DNA damageThis model is described in the article: [A compr…

Details

Human cells that suffer mild DNA damage can enter a reversible state of growth arrest known as quiescence. This decision to temporarily exit the cell cycle is essential to prevent the propagation of mutations, and most cancer cells harbor defects in the underlying control system. Here we present a mechanistic mathematical model to study the proliferation-quiescence decision in nontransformed human cells. We show that two bistable switches, the restriction point (RP) and the G1/S transition, mediate this decision by integrating DNA damage and mitogen signals. In particular, our data suggest that the cyclin-dependent kinase inhibitor p21 (Cip1/Waf1), which is expressed in response to DNA damage, promotes quiescence by blocking positive feedback loops that facilitate G1 progression downstream of serum stimulation. Intriguingly, cells exploit bistability in the RP to convert graded p21 and mitogen signals into an all-or-nothing cell-cycle response. The same mechanism creates a window of opportunity where G1 cells that have passed the RP can revert to quiescence if exposed to DNA damage. We present experimental evidence that cells gradually lose this ability to revert to quiescence as they progress through G1 and that the onset of rapid p21 degradation at the G1/S transition prevents this response altogether, insulating S phase from mild, endogenous DNA damage. Thus, two bistable switches conspire in the early cell cycle to provide both sensitivity and robustness to external stimuli. link: http://identifiers.org/pubmed/29463760

Parameters:

NameDescription
kReDamP53 = 0.005; kReDam = 0.001; jDam = 0.5Reaction: Dam => ; P53, Rate Law: Cell*(kReDam+kReDamP53*P53/(jDam+Dam))*Dam
kSyDna = 0.0093Reaction: aRc => aRc + Dna, Rate Law: Cell*kSyDna*aRc
kDeE2f = 0.05Reaction: RbE2f => Rb, Rate Law: Cell*kDeE2f*RbE2f
kDsRcPc = 0.001; kAsRcPc = 0.01Reaction: iPcna + pRc => iRc, Rate Law: Cell*(kAsRcPc*iPcna*pRc-kDsRcPc*iRc)
kDpRb = 0.05Reaction: pRb => Rb, Rate Law: Cell*kDpRb*pRb
kDeE1C1 = 0.005Reaction: E1C1 => C1, Rate Law: Cell*kDeE1C1*E1C1
kDsRbE2f = 0.005; kAsRbE2f = 5.0Reaction: Rb + E2f => RbE2f, Rate Law: Cell*(kAsRbE2f*Rb*E2f-kDsRbE2f*RbE2f)
kPhC1Ca = 1.0; kPhC1 = 0.0; kPhC1Ce = 0.01Reaction: C1 => pC1; Ce, Ca, Rate Law: Cell*(kPhC1+kPhC1Ce*Ce+kPhC1Ca*Ca)*C1
kDeE1 = 5.0E-4Reaction: E1 =>, Rate Law: Cell*kDeE1*E1
kExPc = 0.006Reaction: aPcna =>, Rate Law: Cell*kExPc*aPcna
kImPc = 0.003Reaction: => aPcna, Rate Law: Cell*kImPc
n = 6.0; kPhRc = 0.1; jCy = 1.8Reaction: Rc => pRc; Ce, Ca, Rate Law: Cell*kPhRc*(Ce+Ca)^n/(jCy^n+(Ce+Ca)^n)*Rc
kDsE1C1 = 0.01; kAsE1C1 = 10.0Reaction: E1 + C1 => E1C1, Rate Law: Cell*(kAsE1C1*E1*C1-kDsE1C1*E1C1)
kDeCaC1 = 2.0; kDeCa = 0.01Reaction: CaP21 => P21; C1, Rate Law: Cell*(kDeCa+kDeCaC1*C1)*CaP21
kGeDam = 0.001Reaction: => Dam, Rate Law: Cell*kGeDam
kPhRbCe = 0.3; Cd = 0.65; kPhRbCd = 0.2; kPhRbCa = 0.3Reaction: RbE2f => pRb + E2f; Ce, Ca, Rate Law: Cell*(kPhRbCd*Cd+kPhRbCe*Ce+kPhRbCa*Ca)*RbE2f
Skp2 = 1.0; kDeP21 = 0.0025; kDeP21aRc = 1.0; Cdt2 = 1.0; kDeP21Cy = 0.007Reaction: iRc => aRc; Ce, Ca, aRc, Rate Law: Cell*(kDeP21+kDeP21Cy*Skp2*(Ce+Ca)+kDeP21aRc*Cdt2*aRc)*iRc
kSyCe = 0.01Reaction: E2f => E2f + Ce, Rate Law: Cell*kSyCe*E2f
kDsCyP21 = 0.05; kAsCyP21 = 1.0Reaction: Ce + P21 => CeP21, Rate Law: Cell*(kAsCyP21*Ce*P21-kDsCyP21*CeP21)
kDsPcP21 = 0.01; kAsPcP21 = 100.0Reaction: aRc + P21 => iRc, Rate Law: Cell*(kAsPcP21*aRc*P21-kDsPcP21*iRc)
kDeCeCa = 0.015; kDeCe = 0.004Reaction: CeP21 => P21; Ca, Rate Law: Cell*(kDeCe+kDeCeCa*Ca)*CeP21
kDePr = 1.0E-4; kDeCaC1 = 2.0Reaction: Pr => ; C1, Rate Law: Cell*(kDePr+kDeCaC1*C1)*Pr
kSyE2f = 0.03; jSyE2f = 0.2; kSyE2fE2f = 0.04Reaction: => E2f; E2f, Rate Law: Cell*(kSyE2f+kSyE2fE2f*E2f/(jSyE2f+E2f))
kSyP53 = 0.05Reaction: => P53, Rate Law: Cell*kSyP53
kDpC1 = 0.05Reaction: pC1 => C1, Rate Law: Cell*kDpC1*pC1
kSyCa = 0.02Reaction: E2f => E2f + Ca, Rate Law: Cell*kSyCa*E2f
kSyPr = 0.01Reaction: => Pr, Rate Law: Cell*kSyPr
kSyE1 = 0.005Reaction: E2f => E2f + E1, Rate Law: Cell*kSyE1*E2f
kSyP21 = 0.002; kSyP21P53 = 0.008Reaction: => P21; P53, Rate Law: Cell*(kSyP21+kSyP21P53*P53)
kGeDamArc = 0.012Reaction: aRc => aRc + Dam, Rate Law: Cell*kGeDamArc*aRc
kDpRc = 0.05Reaction: pRc => Rc, Rate Law: Cell*kDpRc*pRc
kDeP53 = 0.05; jP53 = 0.01Reaction: P53 => ; Dam, Rate Law: Cell*kDeP53/(jP53+Dam)*P53

States:

NameDescription
tE1[F-box only protein 5]
tC1[anaphase-promoting complex]
E2f[Transcription factor E2F1; active]
Ce[Cyclin-dependent kinase 2; protein-containing complex; G1/S-specific cyclin-E1; active]
aRc[Pre-Replication Complex; active]
Rc[Pre-Replication Complex]
C1[anaphase-promoting complex; active]
tP21[Cyclin-dependent kinase inhibitor 1]
PrActivity_probe_of_APC_C_Cdh1
P53[Cellular tumor antigen p53]
P21[Cyclin-dependent kinase inhibitor 1]
aPcna[Proliferating cell nuclear antigen; active]
E1[F-box only protein 5]
CeP21[Cyclin-dependent kinase 2; inactive; G1/S-specific cyclin-E1; protein-containing complex]
pC1[anaphase-promoting complex; phosphorylated; inactive]
RbE2f[Retinoblastoma-like protein 2; protein-containing complex; Transcription factor E2F1; inactive]
tRb[Retinoblastoma-like protein 2]
iRc[Pre-Replication Complex; inactive]
Rb[Retinoblastoma-like protein 2]
E1C1[F-box only protein 5; protein-containing complex; anaphase-promoting complex; inactive]
CaP21[Cyclin-A2; inactive; Cyclin-dependent kinase inhibitor 1; protein-containing complex; Cyclin-dependent kinase 2]
iPcna[Proliferating cell nuclear antigen; inactive]
Dam[DNA; damaged]
tCe[Cyclin-dependent kinase 2; protein-containing complex; G1/S-specific cyclin-E1]
Dna[DNA]
tE2f[Transcription factor E2F1]
pRb[Retinoblastoma-like protein 2; increased phosphorylation]
tCa[Cyclin-A2; protein-containing complex; Cyclin-dependent kinase 2]
Ca[Cyclin-dependent kinase 2; Cyclin-A2; protein-containing complex]
pRc[Pre-Replication Complex; phosphorylated; urn:miriam:sbo:SBO%3A0000643]

Heldt2019 - Chlamydomonas multiple-fission cycles: MODEL1904020001v0.0.1

This model is described in the article: A single light-responsive sizer can control multiple-fission cycles in Chlamydom…

Details

Most eukaryotic cells execute binary division after each mass doubling in order to maintain size homeostasis by coordinating cell growth and division. By contrast, the photosynthetic green alga Chlamydomonas can grow more than 8-fold during daytime and then, at night, undergo rapid cycles of DNA replication, mitosis, and cell division, producing up to 16 daughter cells. Here, we propose a mechanistic model for multiple-fission cycles and cell-size control in Chlamydomonas. The model comprises a light-sensitive and size-dependent biochemical toggle switch that acts as a sizer, guarding transitions into and exit from a phase of cell-division cycle oscillations. This simple “sizer-oscillator” arrangement reproduces the experimentally observed features of multiple-fission cycles and the response of Chlamydomonas cells to different light-dark regimes. Our model also makes specific predictions about the size dependence of the time of onset of cell division after cells are transferred from light to dark conditions, and we confirm these predictions by single-cell experiments. Collectively, our results provide a new perspective on the concept of a “commitment point” during the growth of Chlamydomonas cells and hint at intriguing similarities of cell-size control in different eukaryotic lineages. link: http://identifiers.org/doi/10.1016/j.cub.2019.12.026

Henry2009 - Genome-scale metabolic network of Bacillus subtilis (iBsu1103): MODEL1507180015v0.0.1

Henry2009 - Genome-scale metabolic network of Bacillus subtilis (iBsu1103)This model is described in the article: [iBsu…

Details

BACKGROUND: Bacillus subtilis is an organism of interest because of its extensive industrial applications, its similarity to pathogenic organisms, and its role as the model organism for Gram-positive, sporulating bacteria. In this work, we introduce a new genome-scale metabolic model of B. subtilis 168 called iBsu1103. This new model is based on the annotated B. subtilis 168 genome generated by the SEED, one of the most up-to-date and accurate annotations of B. subtilis 168 available. RESULTS: The iBsu1103 model includes 1,437 reactions associated with 1,103 genes, making it the most complete model of B. subtilis available. The model also includes Gibbs free energy change (DeltarG' degrees ) values for 1,403 (97%) of the model reactions estimated by using the group contribution method. These data were used with an improved reaction reversibility prediction method to identify 653 (45%) irreversible reactions in the model. The model was validated against an experimental dataset consisting of 1,500 distinct conditions and was optimized by using an improved model optimization method to increase model accuracy from 89.7% to 93.1%. CONCLUSIONS: Basing the iBsu1103 model on the annotations generated by the SEED significantly improved the model completeness and accuracy compared with the most recent previously published model. The enhanced accuracy of the iBsu1103 model also demonstrates the efficacy of the improved reaction directionality prediction method in accurately identifying irreversible reactions in the B. subtilis metabolism. The proposed improved model optimization methodology was also demonstrated to be effective in minimally adjusting model content to improve model accuracy. link: http://identifiers.org/pubmed/19555510

Henze 2017: MODEL1812210002v0.0.1

The spindle assembly checkpoint (SAC) is an evolutionarily conserved mechanism, exclusively sensitive to the states of k…

Details

The spindle assembly checkpoint (SAC) is an evolutionarily conserved mechanism, exclusively sensitive to the states of kinetochores attached to microtubules. During metaphase, the anaphase-promoting complex/cyclosome (APC/C) is inhibited by the SAC but it rapidly switches to its active form following proper attachment of the final spindle. It had been thought that APC/C activity is an all-or-nothing response, but recent findings have demonstrated that it switches steadily. In this study, we develop a detailed mathematical model that considers all 92 human kinetochores and all major proteins involved in SAC activation and silencing. We perform deterministic and spatially-stochastic simulations and find that certain spatial properties do not play significant roles. Furthermore, we show that our model is consistent with in-vitro mutation experiments of crucial proteins as well as the recently-suggested rheostat switch behavior, measured by Securin or CyclinB concentration. Considering an autocatalytic feedback loop leads to an all-or-nothing toggle switch in the underlying core components, while the output signal of the SAC still behaves like a rheostat switch. The results of this study support the hypothesis that the SAC signal varies with increasing number of attached kinetochores, even though it might still contain toggle switches in some of its components. link: http://identifiers.org/pubmed/28634351

Hermansen2015 - denovo biosynthesis of pyrimidines in yeast: BIOMD0000000590v0.0.1

Hermansen2015 - denovo biosynthesis of pyrimidines in yeastThis model is described in the article: [Characterizing sele…

Details

Selection on proteins is typically measured with the assumption that each protein acts independently. However, selection more likely acts at higher levels of biological organization, requiring an integrative view of protein function. Here, we built a kinetic model for de novo pyrimidine biosynthesis in the yeast Saccharomyces cerevisiae to relate pathway function to selective pressures on individual protein-encoding genes.Gene families across yeast were constructed for each member of the pathway and the ratio of nonsynonymous to synonymous nucleotide substitution rates (dN/dS) was estimated for each enzyme from S. cerevisiae and closely related species. We found a positive relationship between the influence that each enzyme has on pathway function and its selective constraint.We expect this trend to be locally present for enzymes that have pathway control, but over longer evolutionary timescales we expect that mutation-selection balance may change the enzymes that have pathway control. link: http://identifiers.org/pubmed/26511837

Parameters:

NameDescription
K_Mp = 5.48714446027226; g_pyr = 0.198306450999093Reaction: utp => ; utp, Rate Law: compartment*g_pyr*utp/(K_Mp+utp)/compartment
prpp = 0.181644900226225; vmax5 = 5227.49670547203; K_m5 = 0.0195216150005324Reaction: oro => omp; oro, Rate Law: compartment*vmax5*oro*prpp/(K_m5+oro*prpp)/compartment
d = 0.1Reaction: cp => ; cp, Rate Law: compartment*d*cp/compartment
K_m7 = 0.166382738667754; vmax7 = 5.83104141997666Reaction: udp => utp; udp, Rate Law: compartment*vmax7*udp/(K_m7+udp)/compartment
vmax3 = 28.6613123782585; K_m3 = 1.27179181717468Reaction: ca => dho; ca, Rate Law: compartment*vmax3*ca/(K_m3+ca)/compartment
K_m6 = 20.3406449182435; vmax6 = 34.9720846528477Reaction: omp => ump; omp, Rate Law: compartment*vmax6*omp/(K_m6+omp)/compartment
K_m4 = 0.0160033122150611; vmax4 = 91.7802875108298Reaction: dho => oro; dho, Rate Law: compartment*vmax4*dho/(K_m4+dho)/compartment
K_asp = 0.168308889432487; vmax2 = 2.44590712912244; K_m2 = 2.00489353757245; asp = 0.0972544685826559; K_utp = 1.413855257896Reaction: cp => ca; utp, cp, utp, Rate Law: compartment*vmax2*cp*asp/((1+utp/K_utp)*(K_m2+cp)*(K_asp+asp))/compartment
K_q = 0.05784981576165; K_bc = 2.3716657188714; atp = 0.150650172583633; bc = 1.52264278250403; glu = 0.54620785996429; vmax1 = 3.61602627459517; K_utp = 1.413855257896; K_atp = 1.28940553329954Reaction: => cp; utp, utp, Rate Law: compartment*vmax1*bc*glu*atp/((1+utp/K_utp)*(K_atp+atp)*(K_bc+bc)*(K_q+glu))/compartment
K_m8 = 0.00435621076587497; vmax8 = 0.162943604164789Reaction: utp => ctp; utp, Rate Law: compartment*vmax8*utp/(K_m8+utp)/compartment
vmax10 = 6.55543523218919; K_m10 = 0.0267841313759584Reaction: ump => udp; ump, Rate Law: compartment*vmax10*ump/(K_m10+ump)/compartment

States:

NameDescription
udp[UDP; UDP]
utp[UTP; UTP]
dho[(S)-Dihydroorotate; (S)-dihydroorotic acid]
omp[Orotidine 5'-phosphate; orotidine 5'-phosphate]
ctp[CTP; CTP]
ump[UMP; UMP]
ca[N-Carbamoyl-L-aspartate; N-carbamoyl-L-aspartic acid]
oro[Orotate; orotic acid]
cp[Carbamoyl phosphate; carbamoyl phosphate]

Hernjak2005_Calcium_Signaling: BIOMD0000000162v0.0.1

The model reproduces the time profiles of Calcium in the spine and dendrites as depicted in Fig 8 and Fig 9 of the paper…

Details

Modeling and simulation of the calcium signaling events that precede long-term depression of synaptic activity in cerebellar Purkinje cells are performed using the Virtual Cell biological modeling framework. It is found that the unusually high density and low sensitivity of inositol-1,4,5-trisphosphate receptors (IP3R) are critical to the ability of the cell to generate and localize a calcium spike in a single dendritic spine. The results also demonstrate the model's capability to simulate the supralinear calcium spike observed experimentally during coincident activation of the parallel and climbing fibers. The sensitivity of the calcium spikes to certain biological and geometrical effects is investigated as well as the mechanisms that underlie the cell's ability to generate the supralinear spike. The sensitivity of calcium release rates from the IP3R to calcium concentrations, as well as IP3 concentrations, allows the calcium spike to form. The diffusion barrier caused by the small radius of the spine neck is shown to be important, as a threshold radius is observed above which a spike cannot be formed. Additionally, the calcium buffer capacity and diffusion rates from the spine are found to be important parameters in shaping the calcium spike. link: http://identifiers.org/pubmed/16169982

Parameters:

NameDescription
I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Jmax2=21000.0 1E-57*dimensionless*item^(-4)*m^6*mol*s^(-1); dI=20.0 0.001*dimensionless*m^(-3)*mol; Kact=0.3 0.001*dimensionless*m^(-3)*molReaction: Ca_Cytosol => Ca_ER; IP3_Cytosol, h_ERM, ERDensity_ERM, Rate Law: (-ERDensity_ERM*Jmax2*(1+(-0.00166112956810631*Ca_Cytosol*1/(0.00166112956810631*Ca_ER)))*(h_ERM*0.00166112956810631*IP3_Cytosol*0.00166112956810631*Ca_Cytosol*1/(0.00166112956810631*IP3_Cytosol+dI)*1/(0.00166112956810631*Ca_Cytosol+Kact))^3)*ERM*1*1/KMOLE
D=28.0 1E-12*dimensionless*m^2*s^(-1); D28kB_F=4.16951 0.001*dimensionless*m^(-3)*mol; r_n=0.1 μm; r_D=1.0 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; lc=5.6265 μmReaction: D28kB_D_Cytosol => ; D28kB_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*D28kB_D_Cytosol+(-0.00166112956810631*D28kB_Cytosol))*1/l_n*1/r_D^2*1/l_star+D*(0.00166112956810631*D28kB_D_Cytosol+(-D28kB_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
r_d=1.0 μm; r_n=0.1 μm; PABMg_F=60.47222 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; D=43.0 1E-12*dimensionless*m^2*s^(-1); lc=5.6265 μmReaction: PABMg_D_Cytosol => ; PABMg_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*PABMg_D_Cytosol+(-0.00166112956810631*PABMg_Cytosol))*1/l_n*1/r_d^2*1/l_star+D*(0.00166112956810631*PABMg_D_Cytosol+(-PABMg_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
r_n=0.1 μm; r_D=1.0 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; Ca_F=0.045 0.001*dimensionless*m^(-3)*mol; D=223.0 1E-12*dimensionless*m^2*s^(-1); lc=5.6265 μmReaction: Ca_D_Cytosol => ; Ca_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*Ca_D_Cytosol+(-0.00166112956810631*Ca_Cytosol))*1/l_n*1/r_D^2*1/l_star+D*(0.00166112956810631*Ca_D_Cytosol+(-Ca_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
Kf=430.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kr=140.0 s^(-1)Reaction: Ca_D_Cytosol + CG_D_Cytosol => CGB_D_Cytosol, Rate Law: (Kf*0.00166112956810631*Ca_D_Cytosol*0.00166112956810631*CG_D_Cytosol+(-Kr*0.00166112956810631*CGB_D_Cytosol))*Cytosol*1*1/KMOLE
l=0.66 μm; r_neck=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; r_spine=0.288 μm; D=223.0 1E-12*dimensionless*m^2*s^(-1)Reaction: Ca_Cytosol => ; Ca_D_Cytosol, Rate Law: 0.75*D*(0.00166112956810631*Ca_Cytosol+(-0.00166112956810631*Ca_D_Cytosol))*r_neck^2*1/l*1/r_spine^3*Cytosol*1*1/KMOLE
r_d=1.0 μm; r_n=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; CG_F=140.47567 0.001*dimensionless*m^(-3)*mol; D=15.0 1E-12*dimensionless*m^2*s^(-1); lc=5.6265 μmReaction: CG_D_Cytosol => ; CG_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*CG_D_Cytosol+(-0.00166112956810631*CG_Cytosol))*1/l_n*1/r_d^2*1/l_star+D*(0.00166112956810631*CG_D_Cytosol+(-CG_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
r_d=1.0 μm; r_n=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; PABCa_F=16.32481 0.001*dimensionless*m^(-3)*mol; D=43.0 1E-12*dimensionless*m^2*s^(-1); lc=5.6265 μmReaction: PABCa_D_Cytosol => ; PABCa_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*PABCa_D_Cytosol+(-0.00166112956810631*PABCa_Cytosol))*1/l_n*1/r_d^2*1/l_star+D*(0.00166112956810631*PABCa_D_Cytosol+(-PABCa_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
SVR=3.0 μm^(-1); Js=0.0 1E-9*dimensionless*m^(-2)*mol*s^(-1); Rs=0.288 dimensionless; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; pulses_ar = 0.0Reaction: => IP3_Cytosol, Rate Law: SVR*Js*pulses_ar*1/Rs*Cytosol*1*1/KMOLE
KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; Kf=43.5 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=35.8 s^(-1)Reaction: D28k_D_Cytosol + Ca_D_Cytosol => D28kB_D_Cytosol, Rate Law: (Kf*0.00166112956810631*D28k_D_Cytosol*0.00166112956810631*Ca_D_Cytosol+(-Kr*0.00166112956810631*D28kB_D_Cytosol))*Cytosol*1*1/KMOLE
l=0.66 μm; r_neck=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; r_spine=0.288 μm; D=43.0 1E-12*dimensionless*m^2*s^(-1)Reaction: PA_Cytosol => ; PA_D_Cytosol, Rate Law: 0.75*D*(0.00166112956810631*PA_Cytosol+(-0.00166112956810631*PA_D_Cytosol))*r_neck^2*1/l*1/r_spine^3*Cytosol*1*1/KMOLE
D=28.0 1E-12*dimensionless*m^2*s^(-1); r_n=0.1 μm; r_D=1.0 μm; D28k_F=75.83049 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; lc=5.6265 μmReaction: D28k_D_Cytosol => ; D28k_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*D28k_D_Cytosol+(-0.00166112956810631*D28k_Cytosol))*1/l_n*1/r_D^2*1/l_star+D*(0.00166112956810631*D28k_D_Cytosol+(-D28k_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
D=283.0 1E-12*dimensionless*m^2*s^(-1); r_d=1.0 μm; r_n=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; IP3_F=0.16 0.001*dimensionless*m^(-3)*mol; lc=5.6265 μmReaction: IP3_D_Cytosol => ; IP3_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*IP3_D_Cytosol+(-0.00166112956810631*IP3_Cytosol))*1/l_n*1/r_d^2*1/l_star+D*(0.00166112956810631*IP3_D_Cytosol+(-IP3_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
Kf=107.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=0.95 s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*molReaction: PA_Cytosol + Ca_Cytosol => PABCa_Cytosol, Rate Law: (Kf*0.00166112956810631*PA_Cytosol*0.00166112956810631*Ca_Cytosol+(-Kr*0.00166112956810631*PABCa_Cytosol))*Cytosol*1*1/KMOLE
vP=3.75 1E-21*dimensionless*item^(-1)*mol*s^(-1); I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; kP=0.27Reaction: Ca_D_Cytosol => Ca_D_ER; ERDensity_D_ERM, Rate Law: ERDensity_D_ERM*vP*0.00166112956810631*Ca_D_Cytosol*0.00166112956810631*Ca_D_Cytosol*1/(kP*kP+0.00166112956810631*Ca_D_Cytosol*0.00166112956810631*Ca_D_Cytosol)*ERM*1*1/KMOLE
Kf=0.8 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=25.0 s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*molReaction: PA_Cytosol + Mg_Cytosol => PABMg_Cytosol, Rate Law: (Kf*0.00166112956810631*PA_Cytosol*0.00166112956810631*Mg_Cytosol+(-Kr*0.00166112956810631*PABMg_Cytosol))*Cytosol*1*1/KMOLE
D=28.0 1E-12*dimensionless*m^2*s^(-1); l=0.66 μm; r_neck=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; r_spine=0.288 μmReaction: D28k_Cytosol => ; D28k_D_Cytosol, Rate Law: 0.75*D*(0.00166112956810631*D28k_Cytosol+(-0.00166112956810631*D28k_D_Cytosol))*r_neck^2*1/l*1/r_spine^3*Cytosol*1*1/KMOLE
vL=0.12396 1E-21*dimensionless*item^(-1)*mol*s^(-1); I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*molReaction: Ca_Cytosol => Ca_ER; ERDensity_ERM, Rate Law: (-ERDensity_ERM*vL*(1+(-0.00166112956810631*Ca_Cytosol*1/(0.00166112956810631*Ca_ER))))*ERM*1*1/KMOLE
D=28.0 1E-12*dimensionless*m^2*s^(-1); r_n=0.1 μm; r_D=1.0 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; D28kB_high_F=6.98896 0.001*dimensionless*m^(-3)*mol; lc=5.6265 μmReaction: D28kB_high_D_Cytosol => ; D28kB_high_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*D28kB_high_D_Cytosol+(-0.00166112956810631*D28kB_high_Cytosol))*1/l_n*1/r_D^2*1/l_star+D*(0.00166112956810631*D28kB_high_D_Cytosol+(-D28kB_high_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
Kr=2.6 s^(-1); Kf=5.5 1000*dimensionless*m^3*mol^(-1)*s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*molReaction: Ca_Cytosol + D28k_high_Cytosol => D28kB_high_Cytosol, Rate Law: (Kf*0.00166112956810631*Ca_Cytosol*0.00166112956810631*D28k_high_Cytosol+(-Kr*0.00166112956810631*D28kB_high_Cytosol))*Cytosol*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; flux1_ar = 0.0Reaction: Ca_D_Extracellular => Ca_D_Cytosol, Rate Law: flux1_ar*PM*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kon=2.7 1E15*dimensionless*item*m*mol^(-1)*s^(-1); Kinh=0.2 0.001*dimensionless*m^(-3)*molReaction: h_ERM => ; Ca_Cytosol, Rate Law: (-(Kinh+(-(0.00166112956810631*Ca_Cytosol+Kinh)*h_ERM))*Kon)*ERM
r_d=1.0 μm; r_n=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; CGB_F=19.5243 0.001*dimensionless*m^(-3)*mol; D=15.0 1E-12*dimensionless*m^2*s^(-1); lc=5.6265 μmReaction: CGB_D_Cytosol => ; CGB_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*CGB_D_Cytosol+(-0.00166112956810631*CGB_Cytosol))*1/l_n*1/r_d^2*1/l_star+D*(0.00166112956810631*CGB_D_Cytosol+(-CGB_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; flux0_ar = 0.0Reaction: Ca_Extracellular => Ca_Cytosol, Rate Law: flux0_ar*PM*1*1/KMOLE
D=28.0 1E-12*dimensionless*m^2*s^(-1); r_n=0.1 μm; r_D=1.0 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; D28k_high_F=73.01104 0.001*dimensionless*m^(-3)*mol; lc=5.6265 μmReaction: D28k_high_D_Cytosol => ; D28k_high_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*D28k_high_D_Cytosol+(-0.00166112956810631*D28k_high_Cytosol))*1/l_n*1/r_D^2*1/l_star+D*(0.00166112956810631*D28k_high_D_Cytosol+(-D28k_high_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
l=0.66 μm; D=283.0 1E-12*dimensionless*m^2*s^(-1); r_neck=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; r_spine=0.288 μmReaction: IP3_Cytosol => ; IP3_D_Cytosol, Rate Law: 0.75*D*(0.00166112956810631*IP3_Cytosol+(-0.00166112956810631*IP3_D_Cytosol))*r_neck^2*1/l*1/r_spine^3*Cytosol*1*1/KMOLE
PA_F=3.20298 0.001*dimensionless*m^(-3)*mol; r_d=1.0 μm; r_n=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; l_n=0.66 μm; l_star=27.9812 μm; D=43.0 1E-12*dimensionless*m^2*s^(-1); lc=5.6265 μmReaction: PA_D_Cytosol => ; PA_Cytosol, Rate Law: (D*r_n^2*(0.00166112956810631*PA_D_Cytosol+(-0.00166112956810631*PA_Cytosol))*1/l_n*1/r_d^2*1/l_star+D*(0.00166112956810631*PA_D_Cytosol+(-PA_F))*1/l_star*1/lc)*Cytosol*1*1/KMOLE
l=0.66 μm; r_neck=0.1 μm; KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; r_spine=0.288 μm; D=15.0 1E-12*dimensionless*m^2*s^(-1)Reaction: CGB_Cytosol => ; CGB_D_Cytosol, Rate Law: 0.75*D*(0.00166112956810631*CGB_Cytosol+(-0.00166112956810631*CGB_D_Cytosol))*r_neck^2*1/l*1/r_spine^3*Cytosol*1*1/KMOLE
Kdegr=0.14 s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*mol; IP3_CytosolD=0.16 0.001*dimensionless*m^(-3)*molReaction: IP3_D_Cytosol =>, Rate Law: Kdegr*(0.00166112956810631*IP3_D_Cytosol+(-IP3_CytosolD))*Cytosol*1*1/KMOLE
IP3_CytosolS=0.16 0.001*dimensionless*m^(-3)*mol; Kdegr=0.14 s^(-1); KMOLE = 0.00166112956810631 1E-21*dimensionless*item^(-1)*molReaction: IP3_Cytosol =>, Rate Law: Kdegr*(0.00166112956810631*IP3_Cytosol+(-IP3_CytosolS))*Cytosol*1*1/KMOLE

States:

NameDescription
Ca D Extracellular[calcium(2+); Calcium cation]
CGB CytosolCGB_Cytosol
PABCa D Cytosol[Parvalbumin alpha; Calcium cation; calcium(2+); Parvalbumin alpha]
D28kB high D Cytosol[Calbindin]
D28k D Cytosol[Calbindin]
Ca D ER[calcium(2+); Calcium cation]
CG D CytosolCG_D_Cytosol
PA Cytosol[Parvalbumin alpha]
h D ERMh_D_ERM
D28k Cytosol[Calbindin]
D28kB high Cytosol[Calbindin]
D28kB Cytosol[Calbindin]
Ca ER[calcium(2+); Calcium cation]
D28k high D Cytosol[Calbindin]
D28k high Cytosol[Calbindin]
IP3 D Cytosol[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
Ca D Cytosol[calcium(2+); Calcium cation]
D28kB D Cytosol[Calbindin]
CG CytosolCG_Cytosol
PABMg Cytosol[Parvalbumin alpha; Magnesium cation; magnesium atom; Parvalbumin alpha]
h ERMh_ERM
PA D Cytosol[Parvalbumin alpha]
Mg Cytosol[magnesium(2+); Magnesium cation]
CGB D CytosolCGB_D_Cytosol
PABMg D Cytosol[Parvalbumin alpha; Magnesium cation; magnesium atom; Parvalbumin alpha]
Ca Cytosol[calcium(2+); Calcium cation]
IP3 Cytosol[1D-myo-inositol 1,4,5-trisphosphate; D-myo-Inositol 1,4,5-trisphosphate]
Ca Extracellular[calcium(2+); Calcium cation]
Mg D Cytosol[magnesium(2+); Magnesium cation]
PABCa Cytosol[Parvalbumin alpha; Calcium cation; calcium(2+); Parvalbumin alpha]

Herrgård2008_MetabolicNetwork_Yeast: MODEL0072364382v0.0.1

This is a reconstruction of the biochemical network of the yeast *Saccharomyces cerevisiae* carried out at a jamboree o…

Details

Genomic data allow the large-scale manual or semi-automated assembly of metabolic network reconstructions, which provide highly curated organism-specific knowledge bases. Although several genome-scale network reconstructions describe Saccharomyces cerevisiae metabolism, they differ in scope and content, and use different terminologies to describe the same chemical entities. This makes comparisons between them difficult and underscores the desirability of a consolidated metabolic network that collects and formalizes the 'community knowledge' of yeast metabolism. We describe how we have produced a consensus metabolic network reconstruction for S. cerevisiae. In drafting it, we placed special emphasis on referencing molecules to persistent databases or using database-independent forms, such as SMILES or InChI strings, as this permits their chemical structure to be represented unambiguously and in a manner that permits automated reasoning. The reconstruction is readily available via a publicly accessible database and in the Systems Biology Markup Language (http://www.comp-sys-bio.org/yeastnet). It can be maintained as a resource that serves as a common denominator for studying the systems biology of yeast. Similar strategies should benefit communities studying genome-scale metabolic networks of other organisms. link: http://identifiers.org/pubmed/18846089

Hettling2011_CreatineKinase: BIOMD0000000408v0.0.1

This model is from the article: Analyzing the functional properties of the creatine kinase system with multiscale '…

Details

In this study the function of the two isoforms of creatine kinase (CK; EC 2.7.3.2) in myocardium is investigated. The 'phosphocreatine shuttle' hypothesis states that mitochondrial and cytosolic CK plays a pivotal role in the transport of high-energy phosphate (HEP) groups from mitochondria to myofibrils in contracting muscle. Temporal buffering of changes in ATP and ADP is another potential role of CK. With a mathematical model, we analyzed energy transport and damping of high peaks of ATP hydrolysis during the cardiac cycle. The analysis was based on multiscale data measured at the level of isolated enzymes, isolated mitochondria and on dynamic response times of oxidative phosphorylation measured at the whole heart level. Using 'sloppy modeling' ensemble simulations, we derived confidence intervals for predictions of the contributions by phosphocreatine (PCr) and ATP to the transfer of HEP from mitochondria to sites of ATP hydrolysis. Our calculations indicate that only 15±8% (mean±SD) of transcytosolic energy transport is carried by PCr, contradicting the PCr shuttle hypothesis. We also predicted temporal buffering capabilities of the CK isoforms protecting against high peaks of ATP hydrolysis (3750 µMs(-1)) in myofibrils. CK inhibition by 98% in silico leads to an increase in amplitude of mitochondrial ATP synthesis pulsation from 215±23 to 566±31 µMs(-1), while amplitudes of oscillations in cytosolic ADP concentration double from 77±11 to 146±1 µM. Our findings indicate that CK acts as a large bandwidth high-capacity temporal energy buffer maintaining cellular ATP homeostasis and reducing oscillations in mitochondrial metabolism. However, the contribution of CK to the transport of high-energy phosphate groups appears limited. Mitochondrial CK activity lowers cytosolic inorganic phosphate levels while cytosolic CK has the opposite effect. link: http://identifiers.org/pubmed/21912519

Parameters:

NameDescription
j_ck_mm = 0.0 μmol*l^(-1)*s^(-1)Reaction: Cr + ATP => PCr + ADP, Rate Law: j_ck_mm
Kadp = 25.0 μmol*l^(-1); Vmaxsyn = 1503.74 μmol*l^(-1)*s^(-1); Kpi = 800.0 μmol*l^(-1)Reaction: P_ii + ADPi => ATPi, Rate Law: Vmaxsyn*ADPi*P_ii/(Kadp*Kpi*(1+ADPi/Kadp+P_ii/Kpi+ADPi*P_ii/(Kadp*Kpi)))
j_ck_mi = 0.0 μmol*l^(-1)*s^(-1)Reaction: ATPi + Cri => PCri + ADPi, Rate Law: j_ck_mi
j_diff_adp = 0.0 μmol*l^(-1)*s^(-1)Reaction: ADPi => ADP, Rate Law: j_diff_adp
j_diff_pcr = 1.0 μmol*l^(-1)*s^(-1)Reaction: PCri => PCr, Rate Law: j_diff_pcr
j_diff_atp = 1.0 μmol*l^(-1)*s^(-1)Reaction: ATPi => ATP, Rate Law: j_diff_atp
j_diff_cr = 0.0 μmol*l^(-1)*s^(-1)Reaction: Cri => Cr, Rate Law: j_diff_cr
Jhyd = 486.5 μmol*l^(-1)*s^(-1)Reaction: ATP => ADP + P_i, Rate Law: Jhyd
j_diff_pi = 0.0 μmol*l^(-1)*s^(-1)Reaction: P_ii => P_i, Rate Law: j_diff_pi

States:

NameDescription
P ii[inorganic phosphate]
PCr[N-phosphocreatine; Phosphocreatine; B00422; 5359]
PCri[N-phosphocreatine; Phosphocreatine; 5359; B00422]
ATP[ATP; ATP; 3304; B01125]
Cr[creatine; Creatine; 3594; B00084]
ATPi[ATP; ATP; 3304; B01125]
ADPi[ADP; ADP; 3310; B01130]
Cri[creatine; Creatine; 3594; B00084]
ADP[ADP; ADP; 3310; B01130]
P i[inorganic phosphate]

Hilgemann1987_CalciumTransients: MODEL0848444339v0.0.1

This a model from the article: Excitation-contraction coupling and extracellular calcium transients in rabbit atrium:…

Details

Interactions of electrogenic sodium-calcium exchange, calcium channel and sarcoplasmic reticulum in the mammalian heart have been explored by simulation of extracellular calcium transients measured with tetramethylmurexide in rabbit atrium. The approach has been to use the simplest possible formulations of these mechanisms, which together with a minimum number of additional mechanisms allow reconstruction of action potentials, intracellular calcium transients and extracellular calcium transients. A 3:1 sodium-calcium exchange stoichiometry is assumed. Calcium-channel inactivation is assumed to take place by a voltage-dependent mechanism, which is accelerated by a rise in intracellular calcium; intracellular calcium release becomes a major physiological regulator of calcium influx via calcium channels. A calcium release mechanism is assumed, which is both calcium- and voltage-sensitive, and which undergoes prolonged inactivation. 200 microM cytosolic calcium buffer is assumed. For most simulations only instantaneous potassium conductances are simulated so as to study the other mechanisms independently of time- and calcium-dependent outward current. Thus, the model reconstructs extracellular calcium transients and typical action-potential configuration changes during steady-state and non-steady-state stimulation from the mechanisms directly involved in trans-sarcolemmal calcium movements. The model predicts relatively small trans-sarcolemmal calcium movements during regular stimulation (ca. 2 mumol kg-1 fresh mass per excitation); calcium current is fully activated within 2 ms of excitation, inactivation is substantially complete within 30 ms, and sodium-calcium exchange significantly resists repolarization from approximately -30 mV. Net calcium movements many times larger are possible during non-steady-state stimulation. Long action potentials at premature excitations or after inhibition of calcium release can be supported almost exclusively by calcium current (net calcium influx 5-30 mumol kg-1 fresh mass); action potentials during potentiated post-stimulatory contractions can be supported almost exclusively by sodium-calcium exchange (net calcium efflux 4-20 mumol kg-1 fresh mass). Large calcium movements between the extracellular space and the sarcoplasmic reticulum can take place through the cytosol with virtually no contractile activation. The simulations provide integrated explanations of electrical activity, contractile function and trans-sarcolemmal calcium movements, which were outside the explanatory range of previous models. link: http://identifiers.org/pubmed/2884668

Hinch2004_VentricularMyocytes: MODEL0848342500v0.0.1

This a model from the article: A simplified local control model of calcium-induced calcium release in cardiac ventricu…

Details

Calcium (Ca2+)-induced Ca2+ release (CICR) in cardiac myocytes exhibits high gain and is graded. These properties result from local control of Ca2+ release. Existing local control models of Ca2+ release in which interactions between L-Type Ca2+ channels (LCCs) and ryanodine-sensitive Ca2+ release channels (RyRs) are simulated stochastically are able to reconstruct these properties, but only at high computational cost. Here we present a general analytical approach for deriving simplified models of local control of CICR, consisting of low-dimensional systems of coupled ordinary differential equations, from these more complex local control models in which LCC-RyR interactions are simulated stochastically. The resulting model, referred to as the coupled LCC-RyR gating model, successfully reproduces a range of experimental data, including L-Type Ca2+ current in response to voltage-clamp stimuli, inactivation of LCC current with and without Ca2+ release from the sarcoplasmic reticulum, voltage-dependence of excitation-contraction coupling gain, graded release, and the force-frequency relationship. The model does so with low computational cost. link: http://identifiers.org/pubmed/15465866

Hingant2014 - Micellar On-Pathway Intermediate in Prion Amyloid Formation: MODEL1409230001v0.0.1

Hingant2014 - Micellar On-Pathway Intermediate in Prion Amyloid FormationHingant2014 - Micellar On-Pathway Intermediate…

Details

In a previous work by Alvarez-Martinez et al. (2011), the authors pointed out some fallacies in the mainstream interpretation of the prion amyloid formation. It appeared necessary to propose an original hypothesis able to reconcile the in vitro data with the predictions of a mathematical model describing the problem. Here, a model is developed accordingly with the hypothesis that an intermediate on-pathway leads to the conformation of the prion protein into an amyloid competent isoform thanks to a structure, called micelles, formed from hydrodynamic interaction. The authors also compare data to the prediction of their model and propose a new hypothesis for the formation of infectious prion amyloids. link: http://identifiers.org/pubmed/25101755

Ho2019 - Mathematical models of transmission dynamics and vaccine strategies in Hong Kong during the 2017-2018 winter influenza season (Simple): BIOMD0000000851v0.0.1

This is the simple version of the two mathematical models presented by Ho et al. It is a model comprised of simple ordin…

Details

Two mathematical models described by simple ordinary differential equations are developed to investigate the Hong Kong influenza epidemic during 2017-2018 winter, based on overall epidemic dynamics and different influenza subtypes. The first model, describing the overall epidemic dynamics, provides the starting data for the second model which different influenza subtypes, and whose dynamics is further investigated. Weekly data from December 2017 to May 2018 are obtained from the data base of the Centre of Health Protection in Hong Kong, and used to parametrise the models. With the help of these models, we investigate the impact of different vaccination strategies and determine the corresponding critical vaccination coverage for different vaccine efficacies. The results suggest that at least 72% of Hong Kong population should have been vaccinated during 2017-2018 winter to prevent the seasonal epidemic by herd immunity (while data showed that only a maximum of 11.6% of the population were vaccinated). Our results also show that the critical vaccination coverage decreases with increasing vaccine efficacy, and the increase in one influenza subtype vaccine efficacy may lead to an increase in infections caused by a different subtype. link: http://identifiers.org/pubmed/31128142

Parameters:

NameDescription
r = 0.0155; A = 0.1155Reaction: S => V + V_e, Rate Law: compartment*r*(1-V_e/A)
beta = 2.7516Reaction: S => I, Rate Law: compartment*beta*I*S
k = 1.51338Reaction: V => I, Rate Law: compartment*k*I*V
gamma = 2.1272Reaction: I => R, Rate Law: compartment*gamma*I

States:

NameDescription
I[C17005; influenza infection]
S[C17005; Susceptibility]
V e[C17005; C49287; C28385]
V[C17005; C28385]
R[C17005; 0009785]

Hockin2002_BloodCoagulation: BIOMD0000000335v0.0.1

This model is from the article: A model for the stoichiometric regulation of blood coagulation. Hockin MF, Jones…

Details

We have developed a model of the extrinsic blood coagulation system that includes the stoichiometric anticoagulants. The model accounts for the formation, expression, and propagation of the vitamin K-dependent procoagulant complexes and extends our previous model by including: (a) the tissue factor pathway inhibitor (TFPI)-mediated inactivation of tissue factor (TF).VIIa and its product complexes; (b) the antithrombin-III (AT-III)-mediated inactivation of IIa, mIIa, factor VIIa, factor IXa, and factor Xa; (c) the initial activation of factor V and factor VIII by thrombin generated by factor Xa-membrane; (d) factor VIIIa dissociation/activity loss; (e) the binding competition and kinetic activation steps that exist between TF and factors VII and VIIa; and (f) the activation of factor VII by IIa, factor Xa, and factor IXa. These additions to our earlier model generate a model consisting of 34 differential equations with 42 rate constants that together describe the 27 independent equilibrium expressions, which describe the fates of 34 species. Simulations are initiated by "exposing" picomolar concentrations of TF to an electronic milieu consisting of factors II, IX, X, VII, VIIa, V, and VIIII, and the anticoagulants TFPI and AT-III at concentrations found in normal plasma or associated with coagulation pathology. The reaction followed in terms of thrombin generation, proceeds through phases that can be operationally defined as initiation, propagation, and termination. The generation of thrombin displays a nonlinear dependence upon TF, AT-III, and TFPI and the combination of these latter inhibitors displays kinetic thresholds. At subthreshold TF, thrombin production/expression is suppressed by the combination of TFPI and AT-III; for concentrations above the TF threshold, the bolus of thrombin produced is quantitatively equivalent. A comparison of the model with empirical laboratory data illustrates that most experimentally observable parameters are captured, and the pathology that results in enhanced or deficient thrombin generation is accurately described. link: http://identifiers.org/pubmed/11893748

Parameters:

NameDescription
k15 = 1.8Reaction: TF_VIIa_IX => TF_VIIa + IXa, Rate Law: compartment_1*k15*TF_VIIa_IX
k6 = 1.3E7Reaction: Xa + VII => Xa + VIIa, Rate Law: compartment_1*k6*Xa*VII
k27 = 0.2; k28 = 4.0E8Reaction: Xa + Va => Xa_Va, Rate Law: compartment_1*(k28*Xa*Va-k27*Xa_Va)
k21 = 1.0E8; k20 = 0.001Reaction: IXa_VIIIa + X => IXa_VIIIa_X, Rate Law: compartment_1*(k21*IXa_VIIIa*X-k20*IXa_VIIIa_X)
k29 = 103.0; k30 = 1.0E8Reaction: Xa_Va + II => Xa_Va_II, Rate Law: compartment_1*(k30*Xa_Va*II-k29*Xa_Va_II)
k40 = 490.0Reaction: IXa + ATIII => IXa_ATIII, Rate Law: compartment_1*k40*IXa*ATIII
k37 = 5.0E7Reaction: TF_VIIa + Xa_TFPI => TF_VIIa_Xa_TFPI, Rate Law: compartment_1*k37*TF_VIIa*Xa_TFPI
k41 = 7100.0Reaction: IIa + ATIII => IIa_ATIII, Rate Law: compartment_1*k41*IIa*ATIII
k9 = 2.5E7; k8 = 1.05Reaction: TF_VIIa + X => TF_VIIa_X, Rate Law: compartment_1*(k9*TF_VIIa*X-k8*TF_VIIa_X)
k17 = 2.0E7Reaction: IIa + VIII => IIa + VIIIa, Rate Law: compartment_1*k17*IIa*VIII
k23 = 22000.0; k24 = 0.006Reaction: VIIIa => VIIIa1_L + VIIIa2, Rate Law: compartment_1*(k24*VIIIa-k23*VIIIa1_L*VIIIa2)
k25 = 0.001Reaction: IXa_VIIIa_X => VIIIa1_L + VIIIa2 + X + IXa, Rate Law: compartment_1*k25*IXa_VIIIa_X
k35 = 1.1E-4; k36 = 3.2E8Reaction: TF_VIIa_Xa + TFPI => TF_VIIa_Xa_TFPI, Rate Law: compartment_1*(k36*TF_VIIa_Xa*TFPI-k35*TF_VIIa_Xa_TFPI)
k32 = 1.5E7Reaction: mIIa + Xa_Va => IIa + Xa_Va, Rate Law: compartment_1*k32*mIIa*Xa_Va
k16 = 7500.0Reaction: Xa + II => Xa + IIa, Rate Law: compartment_1*k16*Xa*II
k5 = 440000.0Reaction: TF_VIIa + VII => TF_VIIa + VIIa, Rate Law: compartment_1*k5*TF_VIIa*VII
k10 = 6.0Reaction: TF_VIIa_X => TF_VIIa_Xa, Rate Law: compartment_1*k10*TF_VIIa_X
k4 = 2.3E7; k3 = 0.0031Reaction: TF + VIIa => TF_VIIa, Rate Law: compartment_1*(k4*TF*VIIa-k3*TF_VIIa)
k38 = 1500.0Reaction: Xa + ATIII => Xa_ATIII, Rate Law: compartment_1*k38*Xa*ATIII
k7 = 23000.0Reaction: IIa + VII => IIa + VIIa, Rate Law: compartment_1*k7*IIa*VII
k26 = 2.0E7Reaction: IIa + V => IIa + Va, Rate Law: compartment_1*k26*IIa*V
k39 = 7100.0Reaction: mIIa + ATIII => mIIa_ATIII, Rate Law: compartment_1*k39*mIIa*ATIII
k31 = 63.5Reaction: Xa_Va_II => Xa_Va + mIIa, Rate Law: compartment_1*k31*Xa_Va_II
k34 = 900000.0; k33 = 3.6E-4Reaction: Xa + TFPI => Xa_TFPI, Rate Law: compartment_1*(k34*Xa*TFPI-k33*Xa_TFPI)
k2 = 3200000.0; k1 = 0.0031Reaction: TF + VII => TF_VII, Rate Law: compartment_1*(k2*TF*VII-k1*TF_VII)
k22 = 8.2Reaction: IXa_VIIIa_X => IXa_VIIIa + Xa, Rate Law: compartment_1*k22*IXa_VIIIa_X
k12 = 2.2E7; k11 = 19.0Reaction: TF_VIIa + Xa => TF_VIIa_Xa, Rate Law: compartment_1*(k12*TF_VIIa*Xa-k11*TF_VIIa_Xa)
k42 = 230.0Reaction: TF_VIIa + ATIII => TF_VIIa_ATIII, Rate Law: compartment_1*k42*TF_VIIa*ATIII
k19 = 1.0E7; k18 = 0.005Reaction: IXa + VIIIa => IXa_VIIIa, Rate Law: compartment_1*(k19*IXa*VIIIa-k18*IXa_VIIIa)
k14 = 1.0E7; k13 = 2.4Reaction: TF_VIIa + IX => TF_VIIa_IX, Rate Law: compartment_1*(k14*TF_VIIa*IX-k13*TF_VIIa_IX)

States:

NameDescription
IIa ATIII[Antithrombin-III; Prothrombin]
VIII[Coagulation factor VIII]
TFPI[Tissue factor pathway inhibitor]
Xa ATIII[Coagulation factor X; Antithrombin-III]
V[Coagulation factor V]
Xa Va II[Prothrombin; Coagulation factor V; Coagulation factor X]
ATIII[Antithrombin-III]
Xa[Coagulation factor X]
VIIIa1 L[Coagulation factor VIII]
TF VIIa ATIII[Tissue factor; Antithrombin-III; Coagulation factor VII]
IXa ATIII[Coagulation factor IX; Antithrombin-III]
TF VIIa X[Tissue factor; Coagulation factor X; Coagulation factor VII]
TF VIIa Xa[Tissue factor; Coagulation factor X; Coagulation factor VII]
TF[Tissue factor]
TF VIIa Xa TFPI[Tissue factor; Coagulation factor X; Coagulation factor VII; Tissue factor pathway inhibitor]
mIIa ATIII[Antithrombin-III; Prothrombin]
TF VII[Tissue factor; Coagulation factor VII]
X[Coagulation factor X]
Xa Va[Coagulation factor V; Coagulation factor X]
VIIIa2[Coagulation factor VIII]
TF VIIa[Tissue factor; Coagulation factor VII]
VIIIa[Coagulation factor VIII]
Va[Coagulation factor V]
IIa[Prothrombin]
mIIa[Prothrombin]
VIIa[Coagulation factor VII]
IXa VIIIa X[Coagulation factor X; Coagulation factor IX; Coagulation factor VIII]
Xa TFPI[Coagulation factor X; Tissue factor pathway inhibitor]
TF VIIa IX[Tissue factor; Coagulation factor IX; Coagulation factor VII]
IXa[Coagulation factor IX]
VII[Coagulation factor VII]
II[Prothrombin]
IX[Coagulation factor IX]
IXa VIIIa[Coagulation factor IX; Coagulation factor VIII]

Hoefnagel2002_PyruvateBranches: BIOMD0000000017v0.0.1

This a model from the article: Metabolic engineering of lactic acid bacteria, the combined approach: kinetic modelli…

Details

Everyone who has ever tried to radically change metabolic fluxes knows that it is often harder to determine which enzymes have to be modified than it is to actually implement these changes. In the more traditional genetic engineering approaches 'bottle-necks' are pinpointed using qualitative, intuitive approaches, but the alleviation of suspected 'rate-limiting' steps has not often been successful. Here the authors demonstrate that a model of pyruvate distribution in Lactococcus lactis based on enzyme kinetics in combination with metabolic control analysis clearly indicates the key control points in the flux to acetoin and diacetyl, important flavour compounds. The model presented here (available at http://jjj.biochem.sun.ac.za/wcfs.html) showed that the enzymes with the greatest effect on this flux resided outside the acetolactate synthase branch itself. Experiments confirmed the predictions of the model, i.e. knocking out lactate dehydrogenase and overexpressing NADH oxidase increased the flux through the acetolactate synthase branch from 0 to 75% of measured product formation rates. link: http://identifiers.org/pubmed/11932446

Parameters:

NameDescription
Kaclac_9=10.0; V_9=106.0; Kacet_9=100.0Reaction: AcLac => AcetoinIn, Rate Law: V_9*AcLac/Kaclac_9/(1+AcLac/Kaclac_9+AcetoinIn/Kacet_9)
k_14=3.0E-4Reaction: AcLac => AcetoinIn, Rate Law: k_14*AcLac
Kaccoa_3=0.008; Knad_3=0.4; Kcoa_3=0.014; Kpyr_3=1.0; V_3=259.0; Knadh_3=0.1; Ki_3=46.4159Reaction: NAD + pyruvate + CoA => NADH + AcCoA, Rate Law: V_3*pyruvate/Kpyr_3*NAD/Knad_3*CoA/Kcoa_3*NAD/(NAD+Ki_3*NADH)/((1+pyruvate/Kpyr_3)*(1+NAD/Knad_3+NADH/Knadh_3)*(1+CoA/Kcoa_3+AcCoA/Kaccoa_3))
V_10=200.0; Kacet_10=5.0Reaction: AcetoinIn => AcetoinOut, Rate Law: V_10*AcetoinIn/Kacet_10/(1+AcetoinIn/Kacet_10)
n_12=2.58; Katp_12=6.196; V_12=900.0Reaction: ATP => ADP, Rate Law: V_12*(ATP/(ADP*Katp_12))^n_12/(1+(ATP/(ADP*Katp_12))^n_12)
Keq_11=1400.0; Knad_11=0.16; V_11=105.0; Knadh_11=0.02; Kacet_11=0.06; Kbut_11=2.6Reaction: NADH + AcetoinIn => NAD + Butanediol, Rate Law: V_11*(AcetoinIn*NADH-Butanediol*NAD/Keq_11)/(Kacet_11*Knadh_11)/((1+AcetoinIn/Kacet_11+Butanediol/Kbut_11)*(1+NADH/Knadh_11+NAD/Knad_11))
V_13=118.0; Knadh_13=0.041; Ko_13=0.2; Knad_13=1.0Reaction: NADH + O2 => NAD, Rate Law: V_13*NADH*O2/(Knadh_13*Ko_13)/((1+NADH/Knadh_13+NAD/Knad_13)*(1+O2/Ko_13))
Kadp_1=0.04699; V_1=2397.0; Kglc_1=0.1; Knad_1=0.1412; Katp_1=0.01867; Kpyr_1=2.5; Knadh_1=0.08999Reaction: ADP + NAD + halfglucose => ATP + NADH + pyruvate, Rate Law: 2*V_1*halfglucose/(2*Kglc_1)*NAD/Knad_1*ADP/Kadp_1/((1+halfglucose/(2*Kglc_1)+pyruvate/Kpyr_1)*(1+NAD/Knad_1+NADH/Knadh_1)*(1+ADP/Kadp_1+ATP/Katp_1))
Knadh_7=0.05; V_7=162.0; Ketoh_7=1.0; Knad_7=0.08; Keq_7=12354.9; Kaco_7=0.03Reaction: NADH + AcO => NAD + EtOH, Rate Law: V_7*(AcO*NADH-EtOH*NAD/Keq_7)/(Kaco_7*Knadh_7)/((1+NAD/Knad_7+NADH/Knadh_7)*(1+AcO/Kaco_7+EtOH/Ketoh_7))
Kaccoa_6=0.007; Kaco_6=10.0; V_6=97.0; Keq_6=1.0; Kcoa_6=0.008; Knad_6=0.08; Knadh_6=0.025Reaction: NADH + AcCoA => NAD + CoA + AcO, Rate Law: V_6*(AcCoA*NADH-CoA*NAD*AcO/Keq_6)/(Kaccoa_6*Knadh_6)/((1+NAD/Knad_6+NADH/Knadh_6)*(1+AcCoA/Kaccoa_6+CoA/Kcoa_6)*(1+AcO/Kaco_6))
V_5=2700.0; Kac_5=7.0; Keq_5=174.217; Kacp_5=0.16; Kadp_5=0.5; Katp_5=0.07Reaction: ADP + AcP => ATP + Ac, Rate Law: V_5*(AcP*ADP-Ac*ATP/Keq_5)/(Kadp_5*Kacp_5)/((1+AcP/Kacp_5+Ac/Kac_5)*(1+ADP/Kadp_5+ATP/Katp_5))
Kaclac_8=100.0; Kpyr_8=50.0; n_8=2.4; V_8=600.0; Keq_8=9.0E12Reaction: pyruvate => AcLac, Rate Law: V_8*pyruvate/Kpyr_8*(1-AcLac/(pyruvate*Keq_8))*(pyruvate/Kpyr_8+AcLac/Kaclac_8)^(n_8-1)/(1+(pyruvate/Kpyr_8+AcLac/Kaclac_8)^n_8)
Knadh_2=0.08; Knad_2=2.4; Klac_2=100.0; V_2=5118.0; Kpyr_2=1.5; Keq_2=21120.69Reaction: NADH + pyruvate => NAD + lactate, Rate Law: V_2*(pyruvate*NADH-lactate*NAD/Keq_2)/(Kpyr_2*Knadh_2)/((1+pyruvate/Kpyr_2+lactate/Klac_2)*(1+NADH/Knadh_2+NAD/Knad_2))
Kpi_4=2.6; Kacp_4=0.7; Kipi_4=2.6; Kicoa_4=0.029; V_4=42.0; Keq_4=0.0065; Kiaccoa_4=0.2; Kiacp_4=0.2Reaction: AcCoA + PO4 => CoA + AcP, Rate Law: V_4*(AcCoA*PO4-AcP*CoA/Keq_4)/(Kiaccoa_4*Kpi_4)/(1+AcCoA/Kiaccoa_4+PO4/Kipi_4+AcP/Kiacp_4+CoA/Kicoa_4+AcCoA*PO4/(Kiaccoa_4*Kpi_4)+AcP*CoA/(Kacp_4*Kicoa_4))

States:

NameDescription
CoA[coenzyme A; CoA]
halfglucosehalfglucose
ATP[ATP; ATP]
NADH[NADH; NADH]
lactate[lactate; (S)-Lactate]
AcetoinOut[acetoin; Acetoin]
EtOH[ethanol; Ethanol]
AcO[acetaldehyde; Acetaldehyde]
PO4[phosphate(3-); Orthophosphate]
AcCoA[acetyl-CoA; Acetyl-CoA]
AcLac[2-acetyllactic acid; 2-Acetolactate]
pyruvate[pyruvate; Pyruvate]
AcP[acetyl dihydrogen phosphate; Acetyl phosphate]
ADP[ADP; ADP]
NAD[NAD(+); NAD+]
Ac[acetate; Acetate]
AcetoinIn[acetoin; Acetoin]
Butanediol[(S,S)-butane-2,3-diol; (R,R)-butane-2,3-diol; (S,S)-Butane-2,3-diol; (R,R)-Butane-2,3-diol]
O2[dioxygen; Oxygen]

Hoffman2018- ADCC against cancer: BIOMD0000000802v0.0.1

The paper describes a model of ADCC. Created by COPASI 4.26 (Build 213) This model is described in the article:…

Details

Immunotherapies exploit the immune system to target and kill cancer cells, while sparing healthy tissue. Antibody therapies, an important class of immunotherapies, involve the binding to specific antigens on the surface of the tumour cells of antibodies that activate natural killer (NK) cells to kill the tumour cells. Preclinical assessment of molecules that may cause antibody-dependent cellular cytotoxicity (ADCC) involves co-culturing cancer cells, NK cells and antibody in vitro for several hours and measuring subsequent levels of tumour cell lysis. Here we develop a mathematical model of such an in vitro ADCC assay, formulated as a system of time-dependent ordinary differential equations and in which NK cells kill cancer cells at a rate which depends on the amount of antibody bound to each cancer cell. Numerical simulations generated using experimentally-based parameter estimates reveal that the system evolves on two timescales: a fast timescale on which antibodies bind to receptors on the surface of the tumour cells, and NK cells form complexes with the cancer cells, and a longer time-scale on which the NK cells kill the cancer cells. We construct approximate model solutions on each timescale, and show that they are in good agreement with numerical simulations of the full system. Our results show how the processes involved in ADCC change as the initial concentration of antibody and NK-cancer cell ratio are varied. We use these results to explain what information about the tumour cell kill rate can be extracted from the cytotoxicity assays. link: http://identifiers.org/pubmed/28970093

Parameters:

NameDescription
y = 1.0 1; a2 = 1.44 1Reaction: => A; R, S, Rate Law: tme*a2*y*R*S
a1 = 0.001 1Reaction: A => ; R, S, Rate Law: tme*a1*(1-R)*A*S
v1 = 120.0 1; u = 20.0 1Reaction: => C; S, Rate Law: tme*v1*(u-C)*(S-C)
f = 0.0 1Reaction: S => ; C, Rate Law: tme*f*C
v2 = 14.4 1Reaction: C =>, Rate Law: tme*v2*C
y = 1.0 1; a1 = 0.001 1Reaction: => R; A, Rate Law: tme*a1/y*(1-R)*A
a2 = 1.44 1Reaction: R =>, Rate Law: tme*a2*R

States:

NameDescription
S[malignant cell]
A[Antibody]
C[Complex]
R[Complex]

Hoffmann2002_KnockOut_IkBNFkB_Signaling: BIOMD0000000139v0.0.1

The model corresponds to the knock out model of beta-/-, epsilon -/- and reproduces the upper panel in Fig 2C. In order…

Details

Nuclear localization of the transcriptional activator NF-kappaB (nuclear factor kappaB) is controlled in mammalian cells by three isoforms of NF-kappaB inhibitor protein: IkappaBalpha, -beta, and - epsilon. Based on simplifying reductions of the IkappaB-NF-kappaB signaling module in knockout cell lines, we present a computational model that describes the temporal control of NF-kappaB activation by the coordinated degradation and synthesis of IkappaB proteins. The model demonstrates that IkappaBalpha is responsible for strong negative feedback that allows for a fast turn-off of the NF-kappaB response, whereas IkappaBbeta and - epsilon function to reduce the system's oscillatory potential and stabilize NF-kappaB responses during longer stimulations. Bimodal signal-processing characteristics with respect to stimulus duration are revealed by the model and are shown to generate specificity in gene expression. link: http://identifiers.org/pubmed/12424381

Parameters:

NameDescription
r6 = 0.66Reaction: IKK_IkBeps_NFkB => NFkB + IKK, Rate Law: cytoplasm*r6*IKK_IkBeps_NFkB
a1 = 1.35; d1 = 0.075Reaction: IKK + IkBalpha => IKK_IkBalpha, Rate Law: cytoplasm*(a1*IkBalpha*IKK-d1*IKK_IkBalpha)
r2 = 0.09Reaction: IKK_IkBbeta => IKK, Rate Law: cytoplasm*r2*IKK_IkBbeta
r5 = 0.45Reaction: IKK_IkBbeta_NFkB => NFkB + IKK, Rate Law: cytoplasm*r5*IKK_IkBbeta_NFkB
tr1 = 0.2448Reaction: => IkBeps; IkBeps_transcript, Rate Law: nucleus*tr1*IkBeps_transcript
deg4 = 0.00135Reaction: IkBeps_NFkB => NFkB, Rate Law: cytoplasm*deg4*IkBeps_NFkB
tr2a = 9.25E-5Reaction: => IkBalpha_transcript, Rate Law: nucleus*tr2a
tr3 = 0.0168Reaction: IkBalpha_transcript =>, Rate Law: nucleus*tr3*IkBalpha_transcript
tp2 = 0.012; tp1 = 0.018Reaction: IkBalpha => IkBalpha_nuc, Rate Law: cytoplasm*tp1*IkBalpha-nucleus*tp2*IkBalpha_nuc
k1 = 5.4; k01 = 0.0048Reaction: NFkB => NFkB_nuc, Rate Law: cytoplasm*k1*NFkB-nucleus*k01*NFkB_nuc
tr2 = 0.99Reaction: => IkBalpha_transcript; NFkB_nuc, Rate Law: nucleus*tr2*NFkB_nuc^2
tr2b = 0.0Reaction: => IkBbeta_transcript, Rate Law: nucleus*tr2b
flag_for_after_trigger = 0.5; k2_IkBbeta_nuc_NFkB_nuc=0.0069; fr_after_trigger = 0.5Reaction: IkBbeta_nuc_NFkB_nuc => IkBbeta_NFkB, Rate Law: nucleus*k2_IkBbeta_nuc_NFkB_nuc*(fr_after_trigger+flag_for_after_trigger)*IkBbeta_nuc_NFkB_nuc
d6 = 0.03; a6 = 30.0Reaction: NFkB + IKK_IkBeps => IKK_IkBeps_NFkB, Rate Law: cytoplasm*(a6*IKK_IkBeps*NFkB-d6*IKK_IkBeps_NFkB)
tr2e = 0.0Reaction: => IkBeps_transcript, Rate Law: nucleus*tr2e
a8 = 2.88; d2 = 0.105Reaction: IKK + IkBbeta_NFkB => IKK_IkBbeta_NFkB, Rate Law: cytoplasm*(a8*IKK*IkBbeta_NFkB-d2*IKK_IkBbeta_NFkB)
a2 = 0.36; d2 = 0.105Reaction: IKK + IkBbeta => IKK_IkBbeta, Rate Law: cytoplasm*(a2*IkBbeta*IKK-d2*IKK_IkBbeta)
r4 = 1.224Reaction: IKK_IkBalpha_NFkB => NFkB + IKK, Rate Law: cytoplasm*r4*IKK_IkBalpha_NFkB
r3 = 0.132Reaction: IKK_IkBeps => IKK, Rate Law: cytoplasm*r3*IKK_IkBeps
k2 = 0.828Reaction: IkBalpha_nuc_NFkB_nuc => IkBalpha_NFkB, Rate Law: nucleus*k2*IkBalpha_nuc_NFkB_nuc
r1 = 0.2442Reaction: IKK_IkBalpha => IKK, Rate Law: cytoplasm*r1*IKK_IkBalpha
d5 = 0.03; a5 = 30.0Reaction: NFkB_nuc + IkBbeta_nuc => IkBbeta_nuc_NFkB_nuc, Rate Law: nucleus*(a5*IkBbeta_nuc*NFkB_nuc-d5*IkBbeta_nuc_NFkB_nuc)
a9 = 4.2; d3 = 0.105Reaction: IKK + IkBeps_NFkB => IKK_IkBeps_NFkB, Rate Law: cytoplasm*(a9*IKK*IkBeps_NFkB-d3*IKK_IkBeps_NFkB)
deg1 = 0.00678Reaction: IkBeps =>, Rate Law: cytoplasm*deg1*IkBeps
d4 = 0.03; a4 = 30.0Reaction: NFkB_nuc + IkBalpha_nuc => IkBalpha_nuc_NFkB_nuc, Rate Law: nucleus*(a4*IkBalpha_nuc*NFkB_nuc-d4*IkBalpha_nuc_NFkB_nuc)
k02 = 0.0072Reaction: IKK =>, Rate Law: cytoplasm*k02*IKK
a7 = 11.1; d1 = 0.075Reaction: IKK + IkBalpha_NFkB => IKK_IkBalpha_NFkB, Rate Law: cytoplasm*(a7*IKK*IkBalpha_NFkB-d1*IKK_IkBalpha_NFkB)
a3 = 0.54; d3 = 0.105Reaction: IKK + IkBeps => IKK_IkBeps, Rate Law: cytoplasm*(a3*IkBeps*IKK-d3*IKK_IkBeps)
k2_eps = 0.624Reaction: IkBeps_nuc_NFkB_nuc => IkBeps_NFkB, Rate Law: nucleus*0.5*k2_eps*IkBeps_nuc_NFkB_nuc

States:

NameDescription
IkBalpha nuc[NF-kappa-B inhibitor alpha]
IkBeps nuc[NF-kappa-B inhibitor epsilon]
IkBbeta nuc NFkB nuc[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor beta]
IKK[Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta]
IkBeps nuc NFkB nuc[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor epsilon]
IKK IkBbeta NFkB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkBbeta transcriptIkBbeta_transcript
IkBbeta NFkB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor beta]
IkBalpha NFkB[NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
IKK IkBbeta[NF-kappa-B inhibitor beta; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta]
IkBbeta nuc[NF-kappa-B inhibitor beta]
IkBeps transcriptIkBeps_transcript
IKK IkBalpha NFkB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta]
IkBalpha[NF-kappa-B inhibitor alpha]
IkBalpha nuc NFkB nuc[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
IKK IkBeps[NF-kappa-B inhibitor epsilon; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta]
IKK IkBeps NFkB[Nuclear factor NF-kappa-B p105 subunit; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B inhibitor epsilon]
IkBeps NFkB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor epsilon]
IkBeps[NF-kappa-B inhibitor epsilon]
IkBbeta[NF-kappa-B inhibitor beta]
IKK IkBalpha[NF-kappa-B inhibitor alpha; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta]
NFkB nuc[Nuclear factor NF-kappa-B p105 subunit]
NFkB[Nuclear factor NF-kappa-B p105 subunit]
IkBalpha transcriptIkBalpha_transcript

Hoffmann2002_WT_IkBNFkB_Signaling: BIOMD0000000140v0.0.1

This model corresponds to the IkB-NFkB signaling in wild type cells and reproduces the dynamics of the species as depict…

Details

Nuclear localization of the transcriptional activator NF-kappaB (nuclear factor kappaB) is controlled in mammalian cells by three isoforms of NF-kappaB inhibitor protein: IkappaBalpha, -beta, and - epsilon. Based on simplifying reductions of the IkappaB-NF-kappaB signaling module in knockout cell lines, we present a computational model that describes the temporal control of NF-kappaB activation by the coordinated degradation and synthesis of IkappaB proteins. The model demonstrates that IkappaBalpha is responsible for strong negative feedback that allows for a fast turn-off of the NF-kappaB response, whereas IkappaBbeta and - epsilon function to reduce the system's oscillatory potential and stabilize NF-kappaB responses during longer stimulations. Bimodal signal-processing characteristics with respect to stimulus duration are revealed by the model and are shown to generate specificity in gene expression. link: http://identifiers.org/pubmed/12424381

Parameters:

NameDescription
r6 = 0.66Reaction: IKK_IkBeps_NFkB => NFkB + IKK, Rate Law: cytoplasm*r6*IKK_IkBeps_NFkB
r2 = 0.09Reaction: IKK_IkBbeta => IKK, Rate Law: cytoplasm*r2*IKK_IkBbeta
a1 = 1.35; d1 = 0.075Reaction: IKK + IkBalpha => IKK_IkBalpha, Rate Law: cytoplasm*(a1*IkBalpha*IKK-d1*IKK_IkBalpha)
r5 = 0.45Reaction: IKK_IkBbeta_NFkB => NFkB + IKK, Rate Law: cytoplasm*r5*IKK_IkBbeta_NFkB
tr1 = 0.2448Reaction: => IkBalpha; IkBalpha_transcript, Rate Law: nucleus*tr1*IkBalpha_transcript
tr2b = 1.068E-5Reaction: => IkBbeta_transcript, Rate Law: nucleus*tr2b
a7 = 11.1; d1 = 0.075Reaction: IKK + IkBalpha_NFkB => IKK_IkBalpha_NFkB, Rate Law: cytoplasm*(a7*IKK*IkBalpha_NFkB-d1*IKK_IkBalpha_NFkB)
tr2a = 9.25E-5Reaction: => IkBalpha_transcript, Rate Law: nucleus*tr2a
tr2e = 7.62E-6Reaction: => IkBeps_transcript, Rate Law: nucleus*tr2e
tr3 = 0.0168Reaction: IkBbeta_transcript =>, Rate Law: nucleus*tr3*IkBbeta_transcript
tp2 = 0.012; tp1 = 0.018Reaction: IkBalpha => IkBalpha_nuc, Rate Law: cytoplasm*tp1*IkBalpha-nucleus*tp2*IkBalpha_nuc
k1 = 5.4; k01 = 0.0048Reaction: NFkB => NFkB_nuc, Rate Law: cytoplasm*k1*NFkB-nucleus*k01*NFkB_nuc
tr2 = 0.99Reaction: => IkBalpha_transcript; NFkB_nuc, Rate Law: nucleus*tr2*NFkB_nuc^2
flag_for_after_trigger = 0.5; k2_IkBbeta_nuc_NFkB_nuc=0.0069; fr_after_trigger = 0.5Reaction: IkBbeta_nuc_NFkB_nuc => IkBbeta_NFkB, Rate Law: nucleus*k2_IkBbeta_nuc_NFkB_nuc*(fr_after_trigger+flag_for_after_trigger)*IkBbeta_nuc_NFkB_nuc
d6 = 0.03; a6 = 30.0Reaction: NFkB + IKK_IkBeps => IKK_IkBeps_NFkB, Rate Law: cytoplasm*(a6*IKK_IkBeps*NFkB-d6*IKK_IkBeps_NFkB)
a8 = 2.88; d2 = 0.105Reaction: IKK + IkBbeta_NFkB => IKK_IkBbeta_NFkB, Rate Law: cytoplasm*(a8*IKK*IkBbeta_NFkB-d2*IKK_IkBbeta_NFkB)
a2 = 0.36; d2 = 0.105Reaction: IKK + IkBbeta => IKK_IkBbeta, Rate Law: cytoplasm*(a2*IkBbeta*IKK-d2*IKK_IkBbeta)
r4 = 1.224Reaction: IKK_IkBalpha_NFkB => NFkB + IKK, Rate Law: cytoplasm*r4*IKK_IkBalpha_NFkB
r3 = 0.132Reaction: IKK_IkBeps => IKK, Rate Law: cytoplasm*r3*IKK_IkBeps
k2 = 0.828Reaction: IkBalpha_nuc_NFkB_nuc => IkBalpha_NFkB, Rate Law: nucleus*k2*IkBalpha_nuc_NFkB_nuc
r1 = 0.2442Reaction: IKK_IkBalpha => IKK, Rate Law: cytoplasm*r1*IKK_IkBalpha
d5 = 0.03; a5 = 30.0Reaction: NFkB_nuc + IkBbeta_nuc => IkBbeta_nuc_NFkB_nuc, Rate Law: nucleus*(a5*IkBbeta_nuc*NFkB_nuc-d5*IkBbeta_nuc_NFkB_nuc)
a9 = 4.2; d3 = 0.105Reaction: IKK + IkBeps_NFkB => IKK_IkBeps_NFkB, Rate Law: cytoplasm*(a9*IKK*IkBeps_NFkB-d3*IKK_IkBeps_NFkB)
deg1 = 0.00678Reaction: IkBalpha =>, Rate Law: cytoplasm*deg1*IkBalpha
deg4 = 0.00135Reaction: IkBeps_NFkB => NFkB, Rate Law: cytoplasm*deg4*IkBeps_NFkB
a3 = 0.54; d3 = 0.105Reaction: IKK + IkBeps => IKK_IkBeps, Rate Law: cytoplasm*(a3*IkBeps*IKK-d3*IKK_IkBeps)
d4 = 0.03; a4 = 30.0Reaction: NFkB_nuc + IkBalpha_nuc => IkBalpha_nuc_NFkB_nuc, Rate Law: nucleus*(a4*IkBalpha_nuc*NFkB_nuc-d4*IkBalpha_nuc_NFkB_nuc)
k02 = 0.0072Reaction: IKK =>, Rate Law: cytoplasm*k02*IKK
k2_eps = 0.624Reaction: IkBeps_nuc_NFkB_nuc => IkBeps_NFkB, Rate Law: nucleus*0.5*k2_eps*IkBeps_nuc_NFkB_nuc

States:

NameDescription
IkBalpha nuc[NF-kappa-B inhibitor alpha]
IkBeps nuc[NF-kappa-B inhibitor epsilon]
IkBbeta nuc NFkB nuc[NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IKK[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
IkBeps nuc NFkB nuc[NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit]
IKK IkBbeta NFkB[Inhibitor of nuclear factor kappa-B kinase subunit alpha; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IkBbeta transcriptIkBbeta_transcript
IkBbeta NFkB[NF-kappa-B inhibitor beta; Nuclear factor NF-kappa-B p105 subunit]
IkBalpha NFkB[Nuclear factor NF-kappa-B p105 subunit; NF-kappa-B inhibitor alpha]
IKK IkBbeta[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor beta]
IkBbeta nuc[NF-kappa-B inhibitor beta]
IkBeps transcriptIkBeps_transcript
IKK IkBalpha NFkB[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
IkBalpha[NF-kappa-B inhibitor alpha]
IKK IkBeps[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor epsilon]
IkBalpha nuc NFkB nuc[NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
IKK IkBeps NFkB[NF-kappa-B inhibitor epsilon; Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; Nuclear factor NF-kappa-B p105 subunit]
IkBeps NFkB[NF-kappa-B inhibitor epsilon; Nuclear factor NF-kappa-B p105 subunit]
IkBeps[NF-kappa-B inhibitor epsilon]
IkBbeta[NF-kappa-B inhibitor beta]
IKK IkBalpha[Inhibitor of nuclear factor kappa-B kinase subunit beta; NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit alpha; NF-kappa-B inhibitor alpha]
NFkB nuc[Nuclear factor NF-kappa-B p105 subunit]
NFkB[Nuclear factor NF-kappa-B p105 subunit]
IkBalpha transcriptIkBalpha_transcript

Hofmeyer1986_SeqFb_Proc_AA_Synthesis: BIOMD0000000284v0.0.1

This model is the reaction sequence SEQFB, a model pathway of a branched system with sequential feedback interactions fo…

Details

METAMOD, a BBC microcomputer-based software package for steady-state modelling and control analysis of model metabolic pathways, is described, The package consists of two programs. METADEF allows the user to define the pathway in terms of reactions, rate equations and initial concentrations of metabolites. METACAL uses one of two algorithms to calculate the steady-state concentrations and fluxes. One algorithm uses the current ratio of production and consumption rates of variable metabolites to adjust iteratively their concentrations in such a way that they converge towards the steady state. The other algorithm solves the roots of the system equations by means of a quasi-Newtonian procedure. Control analysis allows the calculation of elasticity, control and response coefficients, by means of finite difference approximation. METAMOD is interactive and easy to use, and suitable for teaching and research purposes. link: http://identifiers.org/pubmed/3450367

States:

NameDescription
YY
BB
ZZ
AA
XX
CC
DD
EE
FF

Hofmeyr1996 - metabolic control analysis: MODEL1304300000v0.0.1

Hofmeyr1996 - metabolic control analysisUnderstanding of genetic mechanisms would not be possible by studying the proper…

Details

The formulation of the standard summation and connectivity relationships as a statement that the matrix of all the elasticities in a system is the inverse of the matrix of all the control coefficients is completely general, provided that only control coefficients for independent fluxes and concentrations are considered, and that the elasticity matrix is written to take account of the stoichiometry of the pathway and the implied dependences between concentrations. This generally implies that co-response analysis is also general, i.e. that all of the elasticities and all of the control coefficients in any system, regardless of branching, feedback effects, moiety conservation or other complications, can be determined by comparing the effects of perturbations of the enzyme activities on the steady-state fluxes and concentrations of the pathway. The approach requires no quantitative information about the magnitudes of the effects on the individual enzyme activities, and consequently no enzymes need to be studied in isolation from the pathway. link: http://identifiers.org/pubmed/8944170

Holmes2006 - Hill's model of muscle contraction: BIOMD0000000677v0.0.1

Holmes2006 - Hill's model of muscle contractionThis model is described in the article: [Teaching from classic papers: H…

Details

A. V. Hill's 1938 paper "The heat of shortening and the dynamic constants of muscle" is an enduring classic, presenting detailed methods, meticulous experiments, and the model of muscle contraction that now bears Hill's name. Pairing a simulation based on Hill's model with a reading of his paper allows students to follow his thought process to discover key principles of muscle physiology and gain insight into how to develop quantitative models of physiological processes. In this article, the experience of the author using this approach in a graduate biomedical engineering course is outlined, along with suggestions for adapting this approach to other audiences. link: http://identifiers.org/pubmed/16709736

Holzhutter2004_Erythrocyte_Metabolism: BIOMD0000000070v0.0.1

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

Details

Cellular functions are ultimately linked to metabolic fluxes brought about by thousands of chemical reactions and transport processes. The synthesis of the underlying enzymes and membrane transporters causes the cell a certain 'effort' of energy and external resources. Considering that those cells should have had a selection advantage during natural evolution that enabled them to fulfil vital functions (such as growth, defence against toxic compounds, repair of DNA alterations, etc.) with minimal effort, one may postulate the principle of flux minimization, as follows: given the available external substrates and given a set of functionally important 'target' fluxes required to accomplish a specific pattern of cellular functions, the stationary metabolic fluxes have to become a minimum. To convert this principle into a mathematical method enabling the prediction of stationary metabolic fluxes, the total flux in the network is measured by a weighted linear combination of all individual fluxes whereby the thermodynamic equilibrium constants are used as weighting factors, i.e. the more the thermodynamic equilibrium lies on the right-hand side of the reaction, the larger the weighting factor for the backward reaction. A linear programming technique is applied to minimize the total flux at fixed values of the target fluxes and under the constraint of flux balance (= steady-state conditions) with respect to all metabolites. The theoretical concept is applied to two metabolic schemes: the energy and redox metabolism of erythrocytes, and the central metabolism of Methylobacterium extorquens AM1. The flux rates predicted by the flux-minimization method exhibit significant correlations with flux rates obtained by either kinetic modelling or direct experimental determination. Larger deviations occur for segments of the network composed of redundant branches where the flux-minimization method always attributes the total flux to the thermodynamically most favourable branch. Nevertheless, compared with existing methods of structural modelling, the principle of flux minimization appears to be a promising theoretical approach to assess stationary flux rates in metabolic systems in cases where a detailed kinetic model is not yet available. link: http://identifiers.org/pubmed/15233787

Parameters:

NameDescription
kATPasev15=1.68 hour_inverseReaction: MgATP => Phi + MgADP, Rate Law: compartment*kATPasev15*MgATP
KR5Pv22=2.2 mM; Vmaxv22=730.0 mM_per_hour; KRu5Pv22=0.78 mM; Keqv22=3.0 dimensionlessReaction: Rul5P => Rib5P, Rate Law: compartment*Vmaxv22*(Rul5P-Rib5P/Keqv22)/(Rul5P+KRu5Pv22*(1+Rib5P/KR5Pv22))
K2PGv10=1.0 mM; Keqv10=0.145 dimensionless; K3PGv10=5.0 mM; Vmaxv10=2000.0 mM_per_hourReaction: Gri3P => Gri2P, Rate Law: compartment*Vmaxv10*(Gri3P-Gri2P/Keqv10)/(Gri3P+K3PGv10*(1+Gri2P/K2PGv10))
Kv20=0.03 hour_inverseReaction: GSH => GSSG, Rate Law: compartment*Kv20*GSH
Keqv7=1455.0 dimensionless; Vmaxv7=5000.0 mM_per_hour; KMgADPv7=0.35 mM; K3PGv7=1.2 mM; K13P2Gv7=0.002 mM; KMgATPv7=0.48 mMReaction: MgADP + Gri13P2 => MgATP + Gri3P, Rate Law: compartment*Vmaxv7/(KMgADPv7*K13P2Gv7)*(MgADP*Gri13P2-MgATP*Gri3P/Keqv7)/(((1+MgADP/KMgADPv7)*(1+Gri13P2/K13P2Gv7)+(1+MgATP/KMgATPv7)*(1+Gri3P/K3PGv7))-1)
Vmaxv13=2800000.0 per_mM_hour; Keqv13=9090.0 dimensionlessReaction: NADH + Pyr => Lac + NAD, Rate Law: compartment*Vmaxv13*(Pyr*NADH-Lac*NAD/Keqv13)
KAMPv3=0.033 mM; KFru6Pv3=0.1 mM; Keqv3=100000.0 dimensionless; KATPv3=0.01 mM; Vmaxv3=239.0 mM_per_hour; KMgATPv3=0.068 mM; L0v3=0.001072 dimensionless; KMgv3=0.44 mMReaction: MgATP + Fru6P => Fru16P2 + MgADP; ATPf, Mgf, AMPf, MgAMP, Rate Law: compartment*Vmaxv3*(Fru6P*MgATP-Fru16P2*MgADP/Keqv3)/((Fru6P+KFru6Pv3)*(MgATP+KMgATPv3)*(1+L0v3*((1+ATPf/KATPv3)*(1+Mgf/KMgv3)/((1+(AMPf+MgAMP)/KAMPv3)*(1+Fru6P/KFru6Pv3)))^4))
KGSHv19=20.0 mM; Vmaxv19=90.0 mM_per_hour; KGSSGv19=0.0652 mM; KNADPHv19=0.00852 mM; KNADPv19=0.07 mM; Keqv19=1.04 dimensionlessReaction: GSSG + NADPHf => GSH + NADPf, Rate Law: compartment*Vmaxv19*(GSSG*NADPHf/(KGSSGv19*KNADPHv19)-GSH^2/KGSHv19^2*NADPf/(KNADPv19*Keqv19))/(1+NADPHf*(1+GSSG/KGSSGv19)/KNADPHv19+NADPf/KNADPv19*(1+GSH*(1+GSH/KGSHv19)/KGSHv19))
KNADPv17=0.00367 mM; KG6Pv17=0.0667 mM; Keqv17=2000.0 dimensionless; KATPv17=0.749 mM; Vmaxv17=162.0 mM_per_hour; KPGA23v17=2.289 mM; KNADPHv17=0.00312 mMReaction: Glc6P + NADPf => GlcA6P + NADPHf; ATPf, MgATP, Gri23P2f, MgGri23P2, Rate Law: compartment*Vmaxv17/KG6Pv17/KNADPv17*(Glc6P*NADPf-GlcA6P*NADPHf/Keqv17)/(1+NADPf*(1+Glc6P/KG6Pv17)/KNADPv17+(ATPf+MgATP)/KATPv17+NADPHf/KNADPHv17+(Gri23P2f+MgGri23P2)/KPGA23v17)
KNADPHv18=0.0045 mM; Keqv18=141.7 dimensionless; KATPv18=0.154 mM; K6PG1v18=0.01 mM; KNADPv18=0.018 mM; K6PG2v18=0.058 mM; KPGA23v18=0.12 mM; Vmaxv18=1575.0 mM_per_hourReaction: GlcA6P + NADPf => Rul5P + NADPHf; Gri23P2f, MgGri23P2, ATPf, MgATP, Rate Law: compartment*Vmaxv18/K6PG1v18/KNADPv18*(GlcA6P*NADPf-Rul5P*NADPHf/Keqv18)/((1+NADPf/KNADPv18)*(1+GlcA6P/K6PG1v18+(Gri23P2f+MgGri23P2)/KPGA23v18)+(ATPf+MgATP)/KATPv18+NADPHf*(1+GlcA6P/K6PG2v18)/KNADPHv18)
KdATP=0.072 mM; EqMult=1.0E7 hour_inverseReaction: MgATP => Mgf + ATPf, Rate Law: compartment*EqMult*(MgATP-Mgf*ATPf/KdATP)
EqMult=1.0E7 hour_inverse; Kd2=1.0E-5 mMReaction: P2NADP => P2f + NADPf, Rate Law: compartment*EqMult*(P2NADP-P2f*NADPf/Kd2)
Keqv8=100000.0 dimensionless; K23P2Gv8=0.04 mM; kDPGMv8=76000.0 hour_inverseReaction: Gri13P2 => Gri23P2f; MgGri23P2, Rate Law: compartment*kDPGMv8*(Gri13P2-(Gri23P2f+MgGri23P2)/Keqv8)/(1+(Gri23P2f+MgGri23P2)/K23P2Gv8)
Kd23P2G=1.667 mM; EqMult=1.0E7 hour_inverseReaction: MgGri23P2 => Mgf + Gri23P2f, Rate Law: compartment*EqMult*(MgGri23P2-Mgf*Gri23P2f/Kd23P2G)
KR5Pv25=0.57 mM; Keqv25=100000.0 dimensionless; Vmaxv25=1.1 mM_per_hour; KATPv25=0.03 mMReaction: MgATP + Rib5P => MgAMP + PRPP, Rate Law: compartment*Vmaxv25*(Rib5P*MgATP-PRPP*MgAMP/Keqv25)/((KATPv25+MgATP)*(KR5Pv25+Rib5P))
K6v23=0.00774 dimensionless; K7v23=48.8 dimensionless; K4v23=0.00496 mM; K1v23=0.4177 mM; K5v23=0.41139 dimensionless; Vmaxv23=23.5 mM_per_hour; Keqv23=1.05 dimensionless; K2v23=0.3055 mM; K3v23=12.432 mMReaction: Xul5P + Rib5P => GraP + Sed7P, Rate Law: compartment*Vmaxv23*(Rib5P*Xul5P-GraP*Sed7P/Keqv23)/((K1v23+Rib5P)*Xul5P+(K2v23+K6v23*Sed7P)*Rib5P+(K3v23+K5v23*Sed7P)*GraP+K4v23*Sed7P+K7v23*Xul5P*GraP)
EqMult=1.0E7 hour_inverse; Kd4=2.0E-4 mMReaction: P2NADPH => P2f + NADPHf, Rate Law: compartment*EqMult*(P2NADPH-P2f*NADPHf/Kd4)
KMGlcv1=0.1 mM; Keqv1=3900.0 mM; Vmax1v1=15.8 mM_per_hour; Vmax2v1=33.2 mM_per_hour; Inhibv1=1.0 dimensionless; KMgATPv1=1.44 mM; KGlc6Pv1=0.0045 mM; K23P2Gv1=2.7 mM; KMgATPMgv1=1.14 mM; KMgv1=1.03 mM; KMg23P2Gv1=3.44 mMReaction: Glcin + MgATP => Glc6P + MgADP; Mgf, Gri23P2f, MgGri23P2, Rate Law: compartment*Inhibv1*Glcin/(Glcin+KMGlcv1)*Vmax1v1/KMgATPv1*((MgATP+Vmax2v1/Vmax1v1*MgATP*Mgf/KMgATPMgv1)-Glc6P*MgADP/Keqv1)/(1+MgATP/KMgATPv1*(1+Mgf/KMgATPMgv1)+Mgf/KMgv1+(1.55+Glc6P/KGlc6Pv1)*(1+Mgf/KMgv1)+(Gri23P2f+MgGri23P2)/K23P2Gv1+Mgf*(Gri23P2f+MgGri23P2)/(KMgv1*KMg23P2Gv1))
Vmaxv28=10000.0 hour_inverse; Keqv28=1.0 dimensionlessReaction: Lacex => Lac, Rate Law: compartment*Vmaxv28*(Lacex-Lac/Keqv28)
Keqv2=0.3925 dimensionless; KGlc6Pv2=0.182 mM; Vmaxv2=935.0 mM_per_hour; KFru6Pv2=0.071 mMReaction: Glc6P => Fru6P, Rate Law: compartment*Vmaxv2*(Glc6P-Fru6P/Keqv2)/(Glc6P+KGlc6Pv2*(1+Fru6P/KFru6Pv2))
Vmaxv26=23.5 mM_per_hour; K6v26=0.122 dimensionless; K5v26=0.0287 dimensionless; Keqv26=1.2 dimensionless; K4v26=3.0E-4 mM; K3v26=0.0548 mM; K2v26=0.3055 mM; K7v26=0.215 dimensionless; K1v26=0.00184 mMReaction: Xul5P + E4P => GraP + Fru6P, Rate Law: compartment*Vmaxv26*(E4P*Xul5P-GraP*Fru6P/Keqv26)/((K1v26+E4P)*Xul5P+(K2v26+K6v26*Fru6P)*E4P+(K3v26+K5v26*Fru6P)*GraP+K4v26*Fru6P+K7v26*Xul5P*GraP)
Vmaxv29=10000.0 hour_inverse; Keqv29=1.0 dimensionlessReaction: Pyrex => Pyr, Rate Law: compartment*Vmaxv29*(Pyrex-Pyr/Keqv29)
Vmaxv21=4634.0 mM_per_hour; KX5Pv21=0.5 mM; KRu5Pv21=0.19 mM; Keqv21=2.7 dimensionlessReaction: Rul5P => Xul5P, Rate Law: compartment*Vmaxv21*(Rul5P-Xul5P/Keqv21)/(Rul5P+KRu5Pv21*(1+Xul5P/KX5Pv21))
Keqv27=1.0 dimensionless; Vmaxv27=100.0 hour_inverseReaction: Phiex => Phi, Rate Law: compartment*Vmaxv27*(Phiex-Phi/Keqv27)
L0v12=19.0 dimensionless; Vmaxv12=570.0 mM_per_hour; KMgADPv12=0.474 mM; KPEPv12=0.225 mM; Keqv12=13790.0 dimensionless; KATPv12=3.39 mM; KFru16P2v12=0.005 mMReaction: PEP + MgADP => MgATP + Pyr; ATPf, Fru16P2, Rate Law: compartment*Vmaxv12*(PEP*MgADP-Pyr*MgATP/Keqv12)/((PEP+KPEPv12)*(MgADP+KMgADPv12)*(1+L0v12*(1+(ATPf+MgATP)/KATPv12)^4/((1+PEP/KPEPv12)^4*(1+Fru16P2/KFru16P2v12)^4)))
EqMult=1.0E7 hour_inverse; Kd1=2.0E-4 mMReaction: P1NADP => P1f + NADPf, Rate Law: compartment*EqMult*(P1NADP-P1f*NADPf/Kd1)
KdAMP=16.64 mM; EqMult=1.0E7 hour_inverseReaction: MgAMP => Mgf + AMPf, Rate Law: compartment*EqMult*(MgAMP-Mgf*AMPf/KdAMP)
K13P2Gv6=0.0035 mM; Keqv6=1.92E-4 dimensionless; KNADHv6=0.0083 mM; KGraPv6=0.005 mM; Vmaxv6=4300.0 mM_per_hour; KNADv6=0.05 mM; KPv6=3.9 mMReaction: GraP + Phi + NAD => NADH + Gri13P2, Rate Law: compartment*Vmaxv6/(KNADv6*KGraPv6*KPv6)*(NAD*GraP*Phi-Gri13P2*NADH/Keqv6)/(((1+NAD/KNADv6)*(1+GraP/KGraPv6)*(1+Phi/KPv6)+(1+NADH/KNADHv6)*(1+Gri13P2/K13P2Gv6))-1)
Vmaxv4=98.91000366 mM_per_hour; KiGraPv4=0.0572 mM; KiiGraPv4=0.176 mM; KGraPv4=0.1906 mM; KFru16P2v4=0.0071 mM; Keqv4=0.114 mM; KDHAPv4=0.0364 mMReaction: Fru16P2 => GraP + DHAP, Rate Law: compartment*Vmaxv4/KFru16P2v4*(Fru16P2-GraP*DHAP/Keqv4)/(1+Fru16P2/KFru16P2v4+GraP/KiGraPv4+DHAP*(GraP+KGraPv4)/(KDHAPv4*KiGraPv4)+Fru16P2*GraP/(KFru16P2v4*KiiGraPv4))
KAMPv16=0.08 mM; KADPv16=0.11 mM; KATPv16=0.09 mM; Keqv16=0.25 dimensionless; Vmaxv16=1380.0 mM_per_hourReaction: MgATP + AMPf => ADPf + MgADP, Rate Law: compartment*Vmaxv16/(KATPv16*KAMPv16)*(MgATP*AMPf-MgADP*ADPf/Keqv16)/((1+MgATP/KATPv16)*(1+AMPf/KAMPv16)+(MgADP+ADPf)/KADPv16+MgADP*ADPf/KADPv16^2)
Keqv5=0.0407 dimensionless; KGraPv5=0.428 mM; KDHAPv5=0.838 mM; Vmaxv5=5456.600098 mM_per_hourReaction: DHAP => GraP, Rate Law: compartment*Vmaxv5*(DHAP-GraP/Keqv5)/(DHAP+KDHAPv5*(1+GraP/KGraPv5))
KdADP=0.76 mM; EqMult=1.0E7 hour_inverseReaction: MgADP => Mgf + ADPf, Rate Law: compartment*EqMult*(MgADP-Mgf*ADPf/KdADP)
Keqv14=14181.8 dimensionless; kLDHv14=243.4 per_mM_hourReaction: Pyr + NADPHf => Lac + NADPf, Rate Law: compartment*kLDHv14*(Pyr*NADPHf-Lac*NADPf/Keqv14)
Keqv0=1.0 dimensionless; KMoutv0=1.7 mM; alfav0=0.54 dimensionless; Vmaxv0=33.6 mM_per_hour; KMinv0=6.9 mMReaction: Glcout => Glcin, Rate Law: compartment*Vmaxv0/KMoutv0*(Glcout-Glcin/Keqv0)/(1+Glcout/KMoutv0+Glcin/KMinv0+alfav0*Glcout*Glcin/KMoutv0/KMinv0)
Keqv11=1.7 dimensionless; K2PGv11=1.0 mM; Vmaxv11=1500.0 mM_per_hour; KPEPv11=1.0 mMReaction: Gri2P => PEP, Rate Law: compartment*Vmaxv11*(Gri2P-PEP/Keqv11)/(Gri2P+K2PGv11*(1+PEP/KPEPv11))
K6v24=0.4653 dimensionless; Keqv24=1.05 dimensionless; K5v24=0.8683 dimensionless; Vmaxv24=27.2 mM_per_hour; K1v24=0.00823 mM; K2v24=0.04765 mM; K7v24=2.524 dimensionless; K3v24=0.1733 mM; K4v24=0.006095 mMReaction: GraP + Sed7P => E4P + Fru6P, Rate Law: compartment*Vmaxv24*(Sed7P*GraP-E4P*Fru6P/Keqv24)/((K1v24+GraP)*Sed7P+(K2v24+K6v24*Fru6P)*GraP+(K3v24+K5v24*Fru6P)*E4P+K4v24*Fru6P+K7v24*Sed7P*E4P)
Keqv9=100000.0 dimensionless; Vmaxv9=0.53 mM_per_hour; K23P2Gv9=0.2 mMReaction: Gri23P2f => Gri3P + Phi; MgGri23P2, Rate Law: compartment*Vmaxv9*((Gri23P2f+MgGri23P2)-Gri3P/Keqv9)/(Gri23P2f+MgGri23P2+K23P2Gv9)
Kd3=1.0E-5 mM; EqMult=1.0E7 hour_inverseReaction: P1NADPH => P1f + NADPHf, Rate Law: compartment*EqMult*(P1NADPH-P1f*NADPHf/Kd3)

States:

NameDescription
ADPf[ADP; ADP]
Rib5P[aldehydo-D-ribose 5-phosphate; D-Ribose 5-phosphate]
NADPf[NADP(+); NADP+]
Glc6P[alpha-D-glucose 6-phosphate; alpha-D-Glucose 6-phosphate]
Fru16P2[beta-D-fructofuranose 1,6-bisphosphate; beta-D-Fructose 1,6-bisphosphate]
Rul5P[D-ribulose 5-phosphate; D-Ribulose 5-phosphate]
GraP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
DHAP[dihydroxyacetone phosphate; Glycerone phosphate]
GSSG[glutathione disulfide; Glutathione disulfide]
MgATP[magnesium atom; ATP; Magnesium cation; ATP; magnesium(2+)]
AMPf[AMP; AMP]
Phi[phosphate ion]
NADPHf[NADPH; NADPH]
Gri3P[3-phospho-D-glyceric acid; 3-Phospho-D-glycerate]
Gri13P2[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
Sed7P[Sedoheptulose 7-phosphate; sedoheptulose 7-phosphate]
GSH[glutathione; Glutathione]
Gri23P2f[2,3-bisphospho-D-glyceric acid; 2,3-Bisphospho-D-glycerate]
GlcA6P[6-O-phosphono-D-glucono-1,5-lactone; D-Glucono-1,5-lactone 6-phosphate]
NADH[NADH; NADH]
Xul5P[D-xylulose 5-phosphate; D-Xylulose 5-phosphate]
MgADP[magnesium atom; ADP; Magnesium cation; ADP; magnesium(2+)]
Mgf[magnesium atom; Magnesium cation]
Pyr[pyruvate; Pyruvate; pyruvic acid]
E4P[D-erythrose 4-phosphate(2-); D-Erythrose 4-phosphate]
Lac[(R)-Lactate; (R)-lactic acid]
MgAMP[magnesium atom; AMP; Magnesium cation; AMP; magnesium(2+)]
Gri2P[2-phospho-D-glyceric acid; 2-Phospho-D-glycerate]
PEP[Phosphoenolpyruvate; phosphoenolpyruvate; phosphoenolpyruvate]
Glcin[glucose; C00293]
NAD[NAD(+); NAD+]
Fru6P[beta-D-fructofuranose 6-phosphate; beta-D-Fructose 6-phosphate]

Hong2004 - Genome-scale metabolic network of Mannheimia succiniciproducens (iSH335): MODEL1507180025v0.0.1

Hong2004 - Genome-scale metabolic network of Mannheimia succiniciproducens (iSH335)This model is described in the articl…

Details

The rumen represents the first section of a ruminant animal's stomach, where feed is collected and mixed with microorganisms for initial digestion. The major gas produced in the rumen is CO(2) (65.5 mol%), yet the metabolic characteristics of capnophilic (CO(2)-loving) microorganisms are not well understood. Here we report the 2,314,078 base pair genome sequence of Mannheimia succiniciproducens MBEL55E, a recently isolated capnophilic Gram-negative bacterium from bovine rumen, and analyze its genome contents and metabolic characteristics. The metabolism of M. succiniciproducens was found to be well adapted to the oxygen-free rumen by using fumarate as a major electron acceptor. Genome-scale metabolic flux analysis indicated that CO(2) is important for the carboxylation of phosphoenolpyruvate to oxaloacetate, which is converted to succinic acid by the reductive tricarboxylic acid cycle and menaquinone systems. This characteristic metabolism allows highly efficient production of succinic acid, an important four-carbon industrial chemical. link: http://identifiers.org/pubmed/15378067

Hong2009_CircadianClock: BIOMD0000000216v0.0.1

This a model from the article: Minimum criteria for DNA damage-induced phase advances in circadian rhythms. Hong CI…

Details

Robust oscillatory behaviors are common features of circadian and cell cycle rhythms. These cyclic processes, however, behave distinctively in terms of their periods and phases in response to external influences such as light, temperature, nutrients, etc. Nevertheless, several links have been found between these two oscillators. Cell division cycles gated by the circadian clock have been observed since the late 1950s. On the other hand, ionizing radiation (IR) treatments cause cells to undergo a DNA damage response, which leads to phase shifts (mostly advances) in circadian rhythms. Circadian gating of the cell cycle can be attributed to the cell cycle inhibitor kinase Wee1 (which is regulated by the heterodimeric circadian clock transcription factor, BMAL1/CLK), and possibly in conjunction with other cell cycle components that are known to be regulated by the circadian clock (i.e., c-Myc and cyclin D1). It has also been shown that DNA damage-induced activation of the cell cycle regulator, Chk2, leads to phosphorylation and destruction of a circadian clock component (i.e., PER1 in Mus or FRQ in Neurospora crassa). However, the molecular mechanism underlying how DNA damage causes predominantly phase advances in the circadian clock remains unknown. In order to address this question, we employ mathematical modeling to simulate different phase response curves (PRCs) from either dexamethasone (Dex) or IR treatment experiments. Dex is known to synchronize circadian rhythms in cell culture and may generate both phase advances and delays. We observe unique phase responses with minimum delays of the circadian clock upon DNA damage when two criteria are met: (1) existence of an autocatalytic positive feedback mechanism in addition to the time-delayed negative feedback loop in the clock system and (2) Chk2-dependent phosphorylation and degradation of PERs that are not bound to BMAL1/CLK. link: http://identifiers.org/pubmed/19424508

Parameters:

NameDescription
ka = 100.0Reaction: CP => CP2, Rate Law: system*ka*CP^2
kica = 20.0Reaction: CP2 + TF => IC, Rate Law: system*kica*CP2*TF
chk2 = 0.0Reaction: CP =>, Rate Law: system*chk2*CP
kicd = 0.01Reaction: IC => CP2 + TF, Rate Law: system*kicd*IC
Dex = 0.0Reaction: => M, Rate Law: system*Dex/system
kms = 1.0; n = 2.0; J = 0.3Reaction: => M; TF, Rate Law: system*kms*TF^n/(J^n+TF^n)/system
Jp = 0.05; kp1 = 10.0Reaction: CP => ; CP2, IC, Rate Law: system*kp1*CP/(Jp+CP+2*CP2+2*IC)/system
kmd = 0.1Reaction: M =>, Rate Law: system*kmd*M
kcpd = 0.525Reaction: CP =>, Rate Law: system*kcpd*CP
kd = 0.01Reaction: CP2 => CP, Rate Law: system*kd*CP2
chk2c = 0.0Reaction: IC => TF, Rate Law: system*chk2c*IC
kcp2d = 0.0525Reaction: IC => TF, Rate Law: system*kcp2d*IC
kcps = 0.5Reaction: => CP; M, Rate Law: system*kcps*M
Jp = 0.05; kp2 = 0.1Reaction: IC => TF; CP2, CP, Rate Law: system*kp2*IC/(Jp+CP+2*CP2+2*IC)/system

States:

NameDescription
CP[Circadian locomoter output cycles protein kaput]
IC[Dual specificity protein kinase CLK1; Aryl hydrocarbon receptor nuclear translocator-like protein 1; Circadian locomoter output cycles protein kaput]
M[messenger RNA; RNA]
TF[Dual specificity protein kinase CLK1; Aryl hydrocarbon receptor nuclear translocator-like protein 1]
CPtotCPtot
CP2[Circadian locomoter output cycles protein kaput]

Hoppe2012 - Predicting changes in metabolic function using transcript profiles (HepatoNet1b_mouse): MODEL1208060000v0.0.1

Hoppe2012 - Predicting changes in metabolic function using transcript profilesMeasuring metabolite concentrations, react…

Details

Genome-wide transcript profiles are often the only available quantitative data for a particular perturbation of a cellular system and their interpretation with respect to the metabolism is a major challenge in systems biology, especially beyond on/off distinction of genes. We present a method that predicts activity changes of metabolic functions by scoring reference flux distributions based on relative transcript profiles, providing a ranked list of most regulated functions. Then, for each metabolic function, the involved genes are ranked upon how much they represent a specific regulation pattern. Compared with the naïve pathway-based approach, the reference modes can be chosen freely, and they represent full metabolic functions, thus, directly provide testable hypotheses for the metabolic study. In conclusion, the novel method provides promising functions for subsequent experimental elucidation together with outstanding associated genes, solely based on transcript profiles. link: http://identifiers.org/doi/10.4230/OASIcs.GCB.2012.1

Hornberg2005 - MAPKsignalling: BIOMD0000000667v0.0.1

Hornberg2005 - MAPKsignallingLarge model of the ERK signalling network. Results from this model were used to generate a…

Details

Oncogenesis results from changes in kinetics or in abundance of proteins in signal transduction networks. Recently, it was shown that control of signalling cannot reside in a single gene product, and might well be dispersed over many components. Which of the reactions in these complex networks are most important, and how can the existing molecular information be used to understand why particular genes are oncogenes whereas others are not? We implement a new method to help address such questions. We apply control analysis to a detailed kinetic model of the epidermal growth factor-induced mitogen-activated protein kinase network. We determine the control of each reaction with respect to three biologically relevant characteristics of the output of this network: the amplitude, duration and integrated output of the transient phosphorylation of extracellular signal-regulated kinase (ERK). We confirm that control is distributed, but far from randomly: a small proportion of reactions substantially control signalling. In particular, the activity of Raf is in control of all characteristics of the transient profile of ERK phosphorylation, which may clarify why Raf is an oncogene. Most reactions that really matter for one signalling characteristic are also important for the other characteristics. Our analysis also predicts the effects of mutations and changes in gene expression. link: http://identifiers.org/pubmed/16007170

Parameters:

NameDescription
kd55 = 5.7Reaction: ERK_P_MEKPP => ERK_PP + MEK_PP, Rate Law: Compartment*kd55*ERK_P_MEKPP
k18 = 2.5E-5; kd18 = 1.3Reaction: Ras_GDP + _EGF_EGFR__2_GAP_SHC__Grb2_Sos => _EGF_EGFR__2_GAP_SHC__Grb2_Sos_Ras_GDP, Rate Law: Compartment*(k18*Ras_GDP*_EGF_EGFR__2_GAP_SHC__Grb2_Sos-kd18*_EGF_EGFR__2_GAP_SHC__Grb2_Sos_Ras_GDP)
kd19 = 0.5; k19 = 1.66E-7Reaction: Ras_GTP + _EGF_EGFR__2_GAP_Grb2_Sos => _EGF_EGFR__2_GAP_Grb2_Sos_Ras_GDP, Rate Law: Compartment*(k19*Ras_GTP*_EGF_EGFR__2_GAP_Grb2_Sos-kd19*_EGF_EGFR__2_GAP_Grb2_Sos_Ras_GDP)
k37 = 1.5E-6; kd37 = 0.3Reaction: _EGF_EGFRi__2_GAP + Shc_0 => _EGF_EGFRi__2_GAP_SHC_0, Rate Law: Compartment*(k37*_EGF_EGFRi__2_GAP*Shc_0-kd37*_EGF_EGFRi__2_GAP_SHC_0)
kd20 = 0.4; k20 = 3.5E-6Reaction: _EGF_EGFR__2_GAP_SHC__Grb2_Sos + Ras_GTP_ => _EGF_EGFR__2_GAP_SHC__Grb2_Sos_Ras_GTP, Rate Law: Compartment*(k20*_EGF_EGFR__2_GAP_SHC__Grb2_Sos*Ras_GTP_-kd20*_EGF_EGFR__2_GAP_SHC__Grb2_Sos_Ras_GTP)
kd57 = 0.246Reaction: ERKi_P_phosphatase3 => ERK + phosphatase3, Rate Law: Compartment*kd57*ERKi_P_phosphatase3
k40 = 5.0E-5; kd40 = 0.064Reaction: Sos + Shc__Grb2 => Shc__Grb2_Sos, Rate Law: Compartment*(k40*Sos*Shc__Grb2-kd40*Shc__Grb2_Sos)
k21 = 3.66E-7; kd21 = 0.023Reaction: _EGF_EGFR__2_GAP_SHC__Grb2_Sos + Ras_GDP => _EGF_EGFR__2_GAP_SHC__Grb2_Sos_Ras_GTP, Rate Law: Compartment*(k21*_EGF_EGFR__2_GAP_SHC__Grb2_Sos*Ras_GDP-kd21*_EGF_EGFR__2_GAP_SHC__Grb2_Sos_Ras_GTP)
kd50 = 0.5; k50 = 4.5E-7Reaction: phosphatse2 + MEKi_P => MEKi_P_phosphatase2, Rate Law: Compartment*(k50*phosphatse2*MEKi_P-kd50*MEKi_P_phosphatase2)
k6 = 5.0E-4Reaction: _EGF_EGFR__2_GAP_Grb2 => _EGF_EGFRi__2_GAP_Grb2, Rate Law: Compartment*k6*_EGF_EGFR__2_GAP_Grb2
k44 = 1.95E-5; kd52 = 0.033Reaction: MEK_P + Raf_0 => MEK_P_Raf, Rate Law: Compartment*(k44*MEK_P*Raf_0-kd52*MEK_P_Raf)
kd127 = 1.0E-4; k127 = 0.0Reaction: ERK_PP + _EGF_EGFR__2_GAP_Grb2_Sos_deg => _EGF_EGFR__2_GAP_Grb2_Sos_ERK_PP, Rate Law: Compartment*(k127*ERK_PP*_EGF_EGFR__2_GAP_Grb2_Sos_deg-kd127*_EGF_EGFR__2_GAP_Grb2_Sos_ERK_PP)
kd25 = 0.0214; k25 = 1.66E-5Reaction: Sos + _EGF_EGFR__2_GAP_SHC__Grb2 => _EGF_EGFR__2_GAP_SHC__Grb2_Sos, Rate Law: Compartment*(k25*Sos*_EGF_EGFR__2_GAP_SHC__Grb2-kd25*_EGF_EGFR__2_GAP_SHC__Grb2_Sos)
k32 = 4.0E-7; kd32 = 0.1Reaction: _EGF_EGFRi__2_GAP + Shc__Grb2_Sos => _EGF_EGFRi__2_GAP_SHC__Grb2_Sos, Rate Law: Compartment*(k32*_EGF_EGFRi__2_GAP*Shc__Grb2_Sos-kd32*_EGF_EGFRi__2_GAP_SHC__Grb2_Sos)
kd17 = 0.06; k17 = 1.66E-5Reaction: Sos + _EGF_EGFR__2_GAP_Grb2 => _EGF_EGFR__2_GAP_Grb2_Sos, Rate Law: Compartment*(k17*Sos*_EGF_EGFR__2_GAP_Grb2-kd17*_EGF_EGFR__2_GAP_Grb2_Sos)
k60 = 0.0055Reaction: _EGF_EGFRi__2_GAP_SHC => _EGF_EGFRi___2deg, Rate Law: Compartment*k60*_EGF_EGFRi__2_GAP_SHC
k34 = 7.5E-6; kd34 = 0.03Reaction: _EGF_EGFRi__2_GAP + Grb2_Sos => _EGF_EGFRi__2_GAP_Grb2_Sos, Rate Law: Compartment*(k34*_EGF_EGFRi__2_GAP*Grb2_Sos-kd34*_EGF_EGFRi__2_GAP_Grb2_Sos)
k126 = 1.66E-7; kd126 = 2.0Reaction: ERKi_PP + Sos => Sos_ERKi_PP, Rate Law: Compartment*(k126*ERKi_PP*Sos-kd126*Sos_ERKi_PP)
kd23 = 0.06; k23 = 6.0Reaction: _EGF_EGFRi__2_GAP_SHC => _EGF_EGFRi__2_GAP_SHC_0, Rate Law: Compartment*(k23*_EGF_EGFRi__2_GAP_SHC-kd23*_EGF_EGFRi__2_GAP_SHC_0)
kd63 = 0.275; k16 = 1.66E-5Reaction: _EGF_EGFRi__2_GAP + Grb2 => _EGF_EGFRi__2_GAP_Grb2, Rate Law: Compartment*(k16*_EGF_EGFRi__2_GAP*Grb2-kd63*_EGF_EGFRi__2_GAP_Grb2)
kd10 = 0.011; k10b = 0.0543Reaction: EGFRi + EGFi => EGF_EGFRi, Rate Law: Compartment*(k10b*EGFRi*EGFi-kd10*EGF_EGFRi)
kd45 = 3.5Reaction: MEK_Raf => MEK_P + Raf_0, Rate Law: Compartment*kd45*MEK_Raf
k56 = 2.35E-5; kd56 = 0.6Reaction: ERK_PP + phosphatase3 => ERK_PP_phosphatase3, Rate Law: Compartment*(k56*ERK_PP*phosphatase3-kd56*ERK_PP_phosphatase3)
k58 = 8.33E-6; kd58 = 0.5Reaction: phosphatase3 + ERK_P => ERK_P_phosphatase3, Rate Law: Compartment*(k58*phosphatase3*ERK_P-kd58*ERK_P_phosphatase3)
k13 = 2.17Reaction: => EGFR, Rate Law: Compartment*k13
k61 = 6.7E-4Reaction: EGFi => EGFideg, Rate Law: Compartment*k61*EGFi
kd35 = 0.0015; k35 = 7.5E-6Reaction: Sos + Grb2 => Grb2_Sos, Rate Law: Compartment*(k35*Sos*Grb2-kd35*Grb2_Sos)
kd49 = 0.0568Reaction: MEK_P_phosphatase2 => MEK + phosphatse2, Rate Law: Compartment*kd49*MEK_P_phosphatase2
k22 = 3.5E-5; kd22 = 0.1Reaction: Shc + _EGF_EGFRi__2_GAP => _EGF_EGFRi__2_GAP_SHC, Rate Law: Compartment*(k22*Shc*_EGF_EGFRi__2_GAP-kd22*_EGF_EGFRi__2_GAP_SHC)
kd53 = 16.0Reaction: ERK_MEK_PP => MEK_PP + ERK_P, Rate Law: Compartment*kd53*ERK_MEK_PP
kd41 = 0.0429; k41 = 5.0E-5Reaction: Grb2_Sos + _EGF_EGFR__2_GAP_SHC_0 => _EGF_EGFR__2_GAP_SHC__Grb2_Sos, Rate Law: Compartment*(k41*Grb2_Sos*_EGF_EGFR__2_GAP_SHC_0-kd41*_EGF_EGFR__2_GAP_SHC__Grb2_Sos)
k16 = 1.66E-5; kd24 = 0.55Reaction: Grb2 + _EGF_EGFRi__2_GAP_SHC_0 => _EGF_EGFRi__2_GAP_SHC__Grb2, Rate Law: Compartment*(k16*Grb2*_EGF_EGFRi__2_GAP_SHC_0-kd24*_EGF_EGFRi__2_GAP_SHC__Grb2)
kd4 = 0.00166; k4 = 1.73E-7Reaction: _EGF_EGFR__2_GAP_Grb2 + Prot => _EGF_EGFR__2_GAP_Grb2_Prot, Rate Law: Compartment*(k4*_EGF_EGFR__2_GAP_Grb2*Prot-kd4*_EGF_EGFR__2_GAP_Grb2_Prot)
k52 = 8.91E-5; kd44 = 0.0183Reaction: ERK + MEKi_PP => ERKi_MEKi_PP_0, Rate Law: Compartment*(k52*ERK*MEKi_PP-kd44*ERKi_MEKi_PP_0)
kd8 = 0.2; k8 = 1.66E-6Reaction: _EGF_EGFRi__2 + GAP => _EGF_EGFRi__2_GAP, Rate Law: Compartment*(k8*_EGF_EGFRi__2*GAP-kd8*_EGF_EGFRi__2_GAP)
kd36 = 0.0; k36 = 0.005Reaction: Shc_0 => Shc, Rate Law: Compartment*(k36*Shc_0-kd36*Shc)

States:

NameDescription
MEK Raf[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1]
EGF EGFRi 2 GAP SHC[Shc-EGFR complex]
EGF EGFR 2 GAP SHC Grb2 Sos Ras GTP[EGFR-Shc-Grb2-Sos complex; Cell division control protein 42 homolog; GTPase KRas]
EGFR[Epidermal growth factor receptor]
Shc[SHC-transforming protein 1]
phosphatse2[Dual specificity protein phosphatase 3]
EGF EGFR 2 GAP SHC Grb2[Shc-EGFR complex; Growth factor receptor-bound protein 2]
EGF EGFR 2 GAP Grb2[Grb2-EGFR complex]
EGF EGFR 2 GAP SHC 0[Shc-EGFR complex]
EGF EGFR 2 GAP SHC Grb2 Sos Ras GDP[EGFR-Shc-Grb2-Sos complex; GTPase KRas; Cell division control protein 42 homolog]
Prot[Interleukin-4 receptor subunit alpha]
ERK P[Phosphoprotein; Mitogen-activated protein kinase 1]
EGF EGFRi 2 GAP SHC Grb2 Sos[EGFR-Shc-Grb2-Sos complex]
phosphatase3[Dual specificity protein phosphatase 3]
ERK P phosphatase3[Phosphoprotein; Dual specificity protein phosphatase 3; Mitogen-activated protein kinase 1]
EGF EGFRi 2 GAP SHC 0[Shc-EGFR complex]
Grb2 Sos[Growth factor receptor-bound protein 2; Son of sevenless homolog 1]
EGF EGFR 2 GAP SHC[Shc-EGFR complex]
Sos[Son of sevenless homolog 1]
EGF EGFRi 2 GAP[Pro-epidermal growth factor; Epidermal growth factor receptor; Ras GTPase-activating protein 1]
ERK PP phosphatase3[Phosphoprotein; Mitogen-activated protein kinase 1; Dual specificity protein phosphatase 3]
ERK MEK PP[Mitogen-activated protein kinase 1; Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]
Grb2[Growth factor receptor-bound protein 2]
MEK P phosphatase2[Phosphoprotein; Dual specificity mitogen-activated protein kinase kinase 1; Dual specificity protein phosphatase 3]
EGFi[Pro-epidermal growth factor]
ERK P MEKPP[Dual specificity mitogen-activated protein kinase kinase 1; Mitogen-activated protein kinase 1; Phosphoprotein]
EGF EGFR 2 GAP SHC Grb2 Sos[EGFR-Shc-Grb2-Sos complex]
MEK P Raf[RAF proto-oncogene serine/threonine-protein kinase; Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]
ERK[Mitogen-activated protein kinase 1]
EGF EGFR 2 GAP Grb2 Sos[EGFR-Grb2-Sos complex]
MEK P[Dual specificity mitogen-activated protein kinase kinase 1; Phosphoprotein]
ERK PP[Phosphoprotein; Mitogen-activated protein kinase 1]

Hornberg2005_ERKcascade: BIOMD0000000084v0.0.1

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

Details

General and simple principles are identified that govern signal transduction. The effects of kinase and phosphatase inhibition on a MAP kinase pathway are first examined in silico. Quantitative measures for the control of signal amplitude, duration and integral strength are introduced. We then identify and prove new principles, such that total control on signal amplitude and on final signal strength must amount to zero, and total control on signal duration and on integral signal intensity must equal -1. Collectively, kinases control amplitudes more than duration, whereas phosphatases tend to control both. We illustrate and validate these principles experimentally: (a) a kinase inhibitor affects the amplitude of EGF-induced ERK phosphorylation much more than its duration and (b) a phosphatase inhibitor influences both signal duration and signal amplitude, in particular long after EGF administration. Implications for the cellular decision between growth and differentiation are discussed. link: http://identifiers.org/pubmed/15634347

Parameters:

NameDescription
Vm4=0.3; Km4=1.0Reaction: x1p => x1, Rate Law: Vm4*x1p/(Km4+x1p)
Km5=0.1; k5=1.0Reaction: x2 => x2p; x1p, Rate Law: k5*x1p*x2/(Km5+x2)
Vm2=0.01; Km2=0.1Reaction: Rin => R, Rate Law: Vm2*Rin/(Km2+Rin)
k7=1.0; Km7=0.1Reaction: x3 => x3p; x2p, Rate Law: k7*x2p*x3/(Km7+x3)
Vm1=1.0; Km1=0.1Reaction: R => Rin, Rate Law: Vm1*R/(Km1+R)
Km3=0.1; k3=1.0Reaction: x1 => x1p; R, Rate Law: k3*R*x1/(Km3+x1)
Km6=1.0; Vm6=0.3Reaction: x2p => x2, Rate Law: Vm6*x2p/(Km6+x2p)
Vm8=0.3; Ki8=1.0; Km8=1.0; Inh=0.0Reaction: x3p => x3, Rate Law: Vm8*x3p/Km8/(1+x3p/Km8+Inh/Ki8)

States:

NameDescription
x1p[RAF proto-oncogene serine/threonine-protein kinase]
x1[RAF proto-oncogene serine/threonine-protein kinase]
x2[Dual specificity mitogen-activated protein kinase kinase 1]
Rin[Receptor protein-tyrosine kinase]
x3p[Mitogen-activated protein kinase 1]
R[Receptor protein-tyrosine kinase]
x3[Mitogen-activated protein kinase 1]
x2p[Dual specificity mitogen-activated protein kinase kinase 1]