SBMLBioModels: M - N

M


Mol2013 - Leishmania macrophage signaling network: MODEL1203220000v0.0.1

Created by The MathWorks, Inc. SimBiology tool, Version 3.3

Details

Network of signaling proteins and functional interaction between the infected cell and the leishmanial parasite, though are not well understood, may be deciphered computationally by reconstructing the immune signaling network. As we all know signaling pathways are well-known abstractions that explain the mechanisms whereby cells respond to signals, collections of pathways form networks, and interactions between pathways in a network, known as cross-talk, enables further complex signaling behaviours. In silico perturbations can help identify sensitive crosstalk points in the network which can be pharmacologically tested. In this study, we have developed a model for immune signaling cascade in leishmaniasis and based upon the interaction analysis obtained through simulation, we have developed a model network, between four signaling pathways i.e., CD14, epidermal growth factor (EGF), tumor necrotic factor (TNF) and PI3 K mediated signaling. Principal component analysis of the signaling network showed that EGF and TNF pathways can be potent pharmacological targets to curb leishmaniasis. The approach is illustrated with a proposed workable model of epidermal growth factor receptor (EGFR) that modulates the immune response. EGFR signaling represents a critical junction between inflammation related signal and potent cell regulation machinery that modulates the expression of cytokines. link: http://identifiers.org/pubmed/24432155

Monro2008 - chemotherapy resistance: BIOMD0000000776v0.0.1

The paper describes a model of resistance of cancer to chemotherapy. Created by COPASI 4.25 (Build 207) This model…

Details

The goal of palliative cancer chemotherapy treatment is to prolong survival and improve quality of life when tumour eradication is not feasible. Chemotherapy protocol design is considered in this context using a simple, robust, model of advanced tumour growth with Gompertzian dynamics, taking into account the effects of drug resistance. It is predicted that reduced chemotherapy protocols can readily lead to improved survival times due to the effects of competition between resistant and sensitive tumour cells. Very early palliation is also predicted to quickly yield near total tumour resistance and thus decrease survival duration. Finally, our simulations indicate that failed curative attempts using dose densification, a common protocol escalation strategy, can reduce survival times. link: http://identifiers.org/pubmed/19135065

Parameters:

NameDescription
N = 1.00002E10 1; Ninf = 2.0E12 1; b = 0.005928 1/d; C0 = 2.0 1Reaction: S =>, Rate Law: tme*(-b)*ln(N/Ninf)*C0*S
N = 1.00002E10 1; Ninf = 2.0E12 1; b = 0.005928 1/dReaction: => R, Rate Law: tme*(-b)*ln(N/Ninf)*R
N = 1.00002E10 1; Ninf = 2.0E12 1; t1 = 1.0E-6 1; t2 = 1.0E-6 1; b = 0.005928 1/dReaction: S => R, Rate Law: tme*(-b)*ln(N/Ninf)*(t1*S-t2*R)

States:

NameDescription
S[malignant cell]
R[malignant cell]

Montagne2011_Oligator_optimised: BIOMD0000000315v0.0.1

This is the model of the in vitro DNA oscillator called oligator with the optmized set of parameters described in the ar…

Details

Living organisms perform and control complex behaviours by using webs of chemical reactions organized in precise networks. This powerful system concept, which is at the very core of biology, has recently become a new foundation for bioengineering. Remarkably, however, it is still extremely difficult to rationally create such network architectures in artificial, non-living and well-controlled settings. We introduce here a method for such a purpose, on the basis of standard DNA biochemistry. This approach is demonstrated by assembling de novo an efficient chemical oscillator: we encode the wiring of the corresponding network in the sequence of small DNA templates and obtain the predicted dynamics. Our results show that the rational cascading of standard elements opens the possibility to implement complex behaviours in vitro. Because of the simple and well-controlled environment, the corresponding chemical network is easily amenable to quantitative mathematical analysis. These synthetic systems may thus accelerate our understanding of the underlying principles of biological dynamic modules. link: http://identifiers.org/pubmed/21283142

Parameters:

NameDescription
k0d = 0.0294 nM_per_min; k0r = 3.43457943925 per_minReaction: T1 + alpha => alpha_T1, Rate Law: sample*(k0d*T1*alpha-k0r*alpha_T1)
k26d = 1.7262 per_minReaction: Inh => empty, Rate Law: sample*k26d*Inh
k17d = 0.0171 nM_per_min; k17r = 0.610714285714 per_minReaction: beta + T3_Inh => beta_T3_Inh, Rate Law: sample*(k17d*beta*T3_Inh-k17r*beta_T3_Inh)
k9r = 0.0171 nM_per_min; k9d = 0.610714285714 per_minReaction: T2_beta => T2 + beta, Rate Law: sample*(k9d*T2_beta-k9r*T2*beta)
k6d = 3.34 per_minReaction: alpha_alpha_T1 => alpha_T1_alpha, Rate Law: sample*k6d*alpha_alpha_T1
k19d = 5.566848 per_minReaction: beta_T3_Inh => Inh + beta_Inh_T3, Rate Law: sample*k19d*beta_T3_Inh
k20d = 3.2064 per_minReaction: beta_Inh_T3 => beta_T3_Inh, Rate Law: sample*k20d*beta_Inh_T3
k10r = 0.0294 nM_per_min; k10d = 3.43457943925 per_minReaction: alpha_T2_beta => alpha + T2_beta, Rate Law: sample*(k10d*alpha_T2_beta-k10r*alpha*T2_beta)
k3r = 0.0294 nM_per_min; k3d = 3.43457943925 per_minReaction: alpha_T1_alpha => alpha + T1_alpha, Rate Law: sample*(k3d*alpha_T1_alpha-k3r*alpha*T1_alpha)
k5d = 11.8408 per_minReaction: alpha_T1_alpha => alpha + alpha_alpha_T1, Rate Law: sample*k5d*alpha_T1_alpha
k18d = 17.024 per_minReaction: beta_T3 => beta_Inh_T3, Rate Law: sample*k18d*beta_T3
k2r = 0.0294 nM_per_min; k2d = 3.43457943925 per_minReaction: T1_alpha => T1 + alpha, Rate Law: sample*(k2d*T1_alpha-k2r*T1*alpha)
k11d = 11.8408 per_minReaction: alpha_T2 => alpha_beta_T2, Rate Law: sample*k11d*alpha_T2
k23r = 0.021546 nM_per_min; k23d = 4.15391351351E-5 nM_per_minReaction: alpha + Inh_T1 => alpha_T1 + Inh, Rate Law: sample*(k23d*alpha*Inh_T1-k23r*alpha_T1*Inh)
k21d = 0.027 nM_per_min; k21r = 0.00608108108108 per_minReaction: T1 + Inh => Inh_T1, Rate Law: sample*(k21d*T1*Inh-k21r*Inh_T1)
k1r = 0.0294 nM_per_min; k1d = 3.43457943925 per_minReaction: alpha_T1_alpha => alpha + alpha_T1, Rate Law: sample*(k1d*alpha_T1_alpha-k1r*alpha*alpha_T1)
k12d = 9.2239832 per_minReaction: alpha_T2_beta => beta + alpha_beta_T2, Rate Law: sample*k12d*alpha_T2_beta
k14r = 0.610714285714 per_min; k14d = 0.0171 nM_per_minReaction: beta + T3 => beta_T3, Rate Law: sample*(k14d*beta*T3-k14r*beta_T3)
k13d = 2.60186 per_minReaction: alpha_beta_T2 => alpha_T2_beta, Rate Law: sample*k13d*alpha_beta_T2
k25d = 0.485802 per_minReaction: beta => empty, Rate Law: sample*k25d*beta
k16d = 0.027 nM_per_min; k16r = 0.00186296832954 per_minReaction: T3 + Inh => T3_Inh, Rate Law: sample*(k16d*T3*Inh-k16r*T3_Inh)
k8r = 0.0171 nM_per_min; k8d = 0.610714285714 per_minReaction: alpha_T2_beta => alpha_T2 + beta, Rate Law: sample*(k8d*alpha_T2_beta-k8r*alpha_T2*beta)
k4d = 15.2 per_minReaction: alpha_T1 => alpha_alpha_T1, Rate Law: sample*k4d*alpha_T1
k24d = 0.411 per_minReaction: alpha => empty, Rate Law: sample*k24d*alpha
k7d = 0.0294 nM_per_min; k7r = 3.43457943925 per_minReaction: alpha + T2 => alpha_T2, Rate Law: sample*(k7d*alpha*T2-k7r*alpha_T2)
k15r = 0.027 nM_per_min; k15d = 0.00186296832954 per_minReaction: beta_T3_Inh => beta_T3 + Inh, Rate Law: sample*(k15d*beta_T3_Inh-k15r*beta_T3*Inh)
k22r = 4.15391351351E-5 nM_per_min; k22d = 0.021546 nM_per_minReaction: T1_alpha + Inh => alpha + Inh_T1, Rate Law: sample*(k22d*T1_alpha*Inh-k22r*alpha*Inh_T1)

States:

NameDescription
T3 Inh[deoxyribonucleic acid; DNA]
Inh[deoxyribonucleic acid; DNA]
alpha T2 beta[deoxyribonucleic acid; DNA]
T1 alpha[deoxyribonucleic acid; DNA]
alpha[deoxyribonucleic acid; DNA]
alpha T2[deoxyribonucleic acid; DNA]
Inh T1[deoxyribonucleic acid; DNA]
alpha alpha T1[deoxyribonucleic acid; DNA]
emptyInh_T1
beta[deoxyribonucleic acid; DNA]
T3[deoxyribonucleic acid; DNA]
alpha beta T2[deoxyribonucleic acid; DNA]
beta T3 Inh[deoxyribonucleic acid; DNA]
beta T3[deoxyribonucleic acid; DNA]
T1[deoxyribonucleic acid; DNA]
beta Inh T3[deoxyribonucleic acid; DNA]
T2[deoxyribonucleic acid; DNA]
alpha T1[deoxyribonucleic acid; DNA]
T2 beta[deoxyribonucleic acid; DNA]
alpha T1 alpha[deoxyribonucleic acid; DNA]

Montagud2010 - Genome-scale metabolic network of Synechocystis sp. PCC6803 (iSyn669): MODEL1507180008v0.0.1

Montagud2010 - Genome-scale metabolic network of Synechocystis sp. PCC6803 (iSyn669)This model is described in the artic…

Details

BACKGROUND: Synechocystis sp. PCC6803 is a cyanobacterium considered as a candidate photo-biological production platform–an attractive cell factory capable of using CO2 and light as carbon and energy source, respectively. In order to enable efficient use of metabolic potential of Synechocystis sp. PCC6803, it is of importance to develop tools for uncovering stoichiometric and regulatory principles in the Synechocystis metabolic network. RESULTS: We report the most comprehensive metabolic model of Synechocystis sp. PCC6803 available, iSyn669, which includes 882 reactions, associated with 669 genes, and 790 metabolites. The model includes a detailed biomass equation which encompasses elementary building blocks that are needed for cell growth, as well as a detailed stoichiometric representation of photosynthesis. We demonstrate applicability of iSyn669 for stoichiometric analysis by simulating three physiologically relevant growth conditions of Synechocystis sp. PCC6803, and through in silico metabolic engineering simulations that allowed identification of a set of gene knock-out candidates towards enhanced succinate production. Gene essentiality and hydrogen production potential have also been assessed. Furthermore, iSyn669 was used as a transcriptomic data integration scaffold and thereby we found metabolic hot-spots around which gene regulation is dominant during light-shifting growth regimes. CONCLUSIONS: iSyn669 provides a platform for facilitating the development of cyanobacteria as microbial cell factories. link: http://identifiers.org/pubmed/21083885

Montañez2008_Arginine_catabolism: BIOMD0000000191v0.0.1

SBML creators: Armando Reyes-Palomares * , Raul Montañez *, Carlos Rodriguez-Caso +, Francisca Sanchez-Jimenez * , Migue…

Details

We use a modeling and simulation approach to carry out an in silico analysis of the metabolic pathways involving arginine as a precursor of nitric oxide or polyamines in aorta endothelial cells. Our model predicts conditions of physiological steady state, as well as the response of the system to changes in the control parameter, external arginine concentration. Metabolic flux control analysis allowed us to predict the values of flux control coefficients for all the transporters and enzymes included in the model. This analysis fulfills the flux control coefficient summation theorem and shows that both the low affinity transporter and arginase share the control of the fluxes through these metabolic pathways. link: http://identifiers.org/pubmed/17520329

Parameters:

NameDescription
Kmeffllat=847.0 microM; Vmaxefflhat=160.5 microMpermin; Kiornhat=360.0 microM; Kmhat=70.0 microM; Kmlat=847.0 microM; Vmaxeffllat=420.0 microMperminReaction: ORN => ; ARGex, ARGin, Rate Law: cytosol*(Vmaxefflhat/(1+ARGex/Kmhat)*ORN/(Kiornhat*(1+ARGin/Kmhat)+ORN)+Vmaxeffllat/(1+ARGex/Kmlat)*ORN/(Kmeffllat*(1+ARGin/Kmlat)+ORN))
Vmaxodc=0.013 microMpermin; Kmodc=90.0 microMReaction: ORN =>, Rate Law: cytosol*Vmaxodc*ORN/(Kmodc+ORN)
Kmnos1=16.0 microM; Vmaxnos1=1.33 microMperminReaction: ARGin =>, Rate Law: cytosol*Vmaxnos1*ARGin/(Kmnos1+ARGin)
Kmarg=1500.0 microM; Vmaxarg=110.0 microMpermin; Kioarg=1000.0 microMReaction: ARGin => ORN; ORN, Rate Law: cytosol*Vmaxarg*ARGin/(Kmarg*(1+ORN/Kioarg)+ARGin)
Kiornhat=360.0 microM; Kmhat=70.0 microM; Kmlat=847.0 microM; Vmaxlat=420.0 microMpermin; Vmaxhat=160.5 microMReaction: ARGex => ARGin; ORN, Rate Law: extracellular*(ARGex/(Kmhat+ARGex)*Vmaxhat/(1+ORN/Kiornhat+ARGin/Kmhat)+ARGex/(Kmlat+ARGex)*Vmaxlat/(1+ORN/Kiornhat+ARGin/Kmlat))

States:

NameDescription
ORN[L-ornithine; L-Ornithine]
ARGin[L-arginine; L-Arginine]
ARGex[L-arginine; L-Arginine]

Moore2004 - Chronic Myeloid Leukemic cells and T-lymphocyte interaction: BIOMD0000000662v0.0.1

Moore2004 - Chronic Myeloid Leukemic cells and T-lymphocytes interactionA mathematical model for the interaction of betw…

Details

In this paper, we propose and analyse a mathematical model for chronic myelogenous leukemia (CML), a cancer of the blood. We model the interaction between naive T cells, effector T cells, and CML cancer cells in the body, using a system of ordinary differential equations which gives rates of change of the three cell populations. One of the difficulties in modeling CML is the scarcity of experimental data which can be used to estimate parameters values. To compensate for the resulting uncertainties, we use Latin hypercube sampling (LHS) on large ranges of possible parameter values in our analysis. A major goal of this work is the determination of parameters which play a critical role in remission or clearance of the cancer in the model. Our analysis examines 12 parameters, and identifies two of these, the growth and death rates of CML, as critical to the outcome of the system. Our results indicate that the most promising research avenues for treatments of CML should be those that affect these two significant parameters (CML growth and death rates), while altering the other parameters should have little effect on the outcome. link: http://identifiers.org/pubmed/15038986

Parameters:

NameDescription
gamma_e = 0.0077 0.0864*l/sReaction: T_cell_effector => Sink; CML, Rate Law: COMpartment*gamma_e*CML*T_cell_effector
eta = 43.0 1/Ml; kn = 0.063 1/(0.0115741*ms)Reaction: T_cell_naive => Sink; CML, Rate Law: COMpartment*kn*T_cell_naive*CML/(CML+eta)
gamma_c = 0.047 0.0864*l/sReaction: CML => Sink; T_cell_effector, Rate Law: COMpartment*gamma_c*T_cell_effector*CML
dc = 0.68 1/(0.0115741*ms)Reaction: CML => Sink, Rate Law: COMpartment*dc*CML
de = 0.12 1/(0.0115741*ms)Reaction: T_cell_effector => Sink, Rate Law: COMpartment*de*T_cell_effector
eta = 43.0 1/Ml; alpha_e = 0.53 1/(0.0115741*ms)Reaction: Source => T_cell_effector; CML, Rate Law: COMpartment*alpha_e*T_cell_effector*CML/(CML+eta)
rc = 0.23 1/(0.0115741*ms); Cmax = 190000.0 1/MlReaction: Source => CML, Rate Law: COMpartment*rc*CML*ln(Cmax/CML)
sn = 0.071 1/(11.5741*l*s)Reaction: Source => T_cell_naive, Rate Law: COMpartment*sn*Source
eta = 43.0 1/Ml; alpha_n = 0.56 1; kn = 0.063 1/(0.0115741*ms)Reaction: Source => T_cell_effector; T_cell_naive, CML, Rate Law: COMpartment*alpha_n*kn*T_cell_naive*CML/(CML+eta)
dn = 0.05 1/(0.0115741*ms)Reaction: T_cell_naive => Sink, Rate Law: COMpartment*dn*T_cell_naive

States:

NameDescription
T cell naive[Naive T-Lymphocyte]
SourceSource
CML[leukemia cell]
T cell effector[Effector T-Lymphocyte]
SinkSink

Moore_2004_Mathematical model for CML and T cell interaction: BIOMD0000000733v0.0.1

Its a mathematical model depicting CML (chronic myelogenous leukemia) interaction with T cells and impact of T cell acti…

Details

In this paper, we propose and analyse a mathematical model for chronic myelogenous leukemia (CML), a cancer of the blood. We model the interaction between naive T cells, effector T cells, and CML cancer cells in the body, using a system of ordinary differential equations which gives rates of change of the three cell populations. One of the difficulties in modeling CML is the scarcity of experimental data which can be used to estimate parameters values. To compensate for the resulting uncertainties, we use Latin hypercube sampling (LHS) on large ranges of possible parameter values in our analysis. A major goal of this work is the determination of parameters which play a critical role in remission or clearance of the cancer in the model. Our analysis examines 12 parameters, and identifies two of these, the growth and death rates of CML, as critical to the outcome of the system. Our results indicate that the most promising research avenues for treatments of CML should be those that affect these two significant parameters (CML growth and death rates), while altering the other parameters should have little effect on the outcome. link: http://identifiers.org/pubmed/15038986

Parameters:

NameDescription
Kn = 0.062 1/d; n = 720.0 mmol/l; An = 0.14 dimensionless; Ae = 0.98 1/dReaction: => eff_Tcells; naive_Tcells, tumor_cells, Rate Law: TumorMicroenvr*(An*Kn*naive_Tcells*tumor_cells/(tumor_cells+n)+Ae*eff_Tcells*tumor_cells/(tumor_cells+n))
gamma_E = 0.057 l/(mmol*d); De = 0.3 1/dReaction: eff_Tcells => ; tumor_cells, Rate Law: TumorMicroenvr*(De*eff_Tcells+gamma_E*tumor_cells*eff_Tcells)
Sn = 0.37 mmol/(l*d)Reaction: => naive_Tcells, Rate Law: TumorMicroenvr*Sn
Dn = 0.23 1/d; Kn = 0.062 1/d; n = 720.0 mmol/lReaction: naive_Tcells => ; tumor_cells, Rate Law: TumorMicroenvr*(Dn*naive_Tcells+Kn*naive_Tcells*tumor_cells/(tumor_cells+n))
gamma_C = 0.0034 l/(mmol*d); Dc = 0.024 1/dReaction: tumor_cells => ; eff_Tcells, Rate Law: TumorMicroenvr*(Dc*tumor_cells-gamma_C*tumor_cells*eff_Tcells)
Cmax = 230000.0 mmol/l; Rc = 0.0057 1/dReaction: => tumor_cells, Rate Law: TumorMicroenvr*Rc*tumor_cells*ln(Cmax/tumor_cells)

States:

NameDescription
naive Tcells[Naive T-Lymphocyte]
tumor cells[neoplasm]
eff Tcells[Effector T-Lymphocyte]

Moreno2019 - Stochastic model of G1 arrest due to proteostasis decline delimits replicative lifespan in yeast: MODEL1901210001v0.0.1

This model is described within the paper: A G1 arrest due to proteostasis decline delimits replicative lifespan in yeast…

Details

Loss of proteostasis and cellular senescence are key hallmarks of aging, but direct cause-effect relationships are not well understood. We show that most yeast cells arrest in G1 before death with low nuclear levels of Cln3, a key G1 cyclin extremely sensitive to chaperone status. Chaperone availability is seriously compromised in aged cells, and the G1 arrest coincides with massive aggregation of a metastable chaperone-activity reporter. Moreover, G1-cyclin overexpression increases lifespan in a chaperone-dependent manner. As a key prediction of a model integrating autocatalytic protein aggregation and a minimal Start network, enforced protein aggregation causes a severe reduction in lifespan, an effect that is greatly alleviated by increased expression of specific chaperones or cyclin Cln3. Overall, our data show that proteostasis breakdown, by compromising chaperone activity and G1-cyclin function, causes an irreversible arrest in G1, configuring a molecular pathway postulating proteostasis decay as a key contributing effector of cell senescence. link: http://identifiers.org/pubmed/31518229

Morgan2016 - Dynamics of cholesterol metabolism and ageing: MODEL1508170000v0.0.1

Morgan2016 - Dynamics of cholesterol metabolism and ageingThis model is described in the article: [Mathematically model…

Details

Cardiovascular disease (CVD) is the leading cause of morbidity and mortality in the UK. This condition becomes increasingly prevalent during ageing; 34.1% and 29.8% of males and females respectively, over 75 years of age have an underlying cardiovascular problem. The dysregulation of cholesterol metabolism is inextricably correlated with cardiovascular health and for this reason low density lipoprotein cholesterol (LDL-C) and high density lipoprotein cholesterol (HDL-C) are routinely used as biomarkers of CVD risk. The aim of this work was to use mathematical modelling to explore how cholesterol metabolism is affected by the ageing process. To do this we updated a previously published whole-body mathematical model of cholesterol metabolism to include an additional 96 mechanisms that are fundamental to this biological system. Additional mechanisms were added to cholesterol absorption, cholesterol synthesis, reverse cholesterol transport (RCT), bile acid synthesis, and their enterohepatic circulation. The sensitivity of the model was explored by the use of both local and global parameter scans. In addition, acute cholesterol feeding was used to explore the effectiveness of the regulatory mechanisms which are responsible for maintaining whole-body cholesterol balance. It was found that our model behaves as a hypo-responder to cholesterol feeding, while both the hepatic and intestinal pools of cholesterol increased significantly. The model was also used to explore the effects of ageing in tandem with three different cholesterol ester transfer protein (CETP) genotypes. Ageing in the presence of an atheroprotective CETP genotype, conferring low CETP activity, resulted in a 0.6% increase in LDL-C. In comparison, ageing with a genotype reflective of high CETP activity, resulted in a 1.6% increase in LDL-C. Thus, the model has illustrated the importance of CETP genotypes such as I405V, and their potential role in healthy ageing. link: http://identifiers.org/pubmed/27157786

Moriya2011_CellCycle_FissionYeast: BIOMD0000000406v0.0.1

This model is from the article: Overexpression limits of fission yeast cell-cycle regulators in vivo and in silico.…

Details

Cellular systems are generally robust against fluctuations of intracellular parameters such as gene expression level. However, little is known about expression limits of genes required to halt cellular systems. In this study, using the fission yeast Schizosaccharomyces pombe, we developed a genetic 'tug-of-war' (gTOW) method to assess the overexpression limit of certain genes. Using gTOW, we determined copy number limits for 31 cell-cycle regulators; the limits varied from 1 to >100. Comparison with orthologs of the budding yeast Saccharomyces cerevisiae suggested the presence of a conserved fragile core in the eukaryotic cell cycle. Robustness profiles of networks regulating cytokinesis in both yeasts (septation-initiation network (SIN) and mitotic exit network (MEN)) were quite different, probably reflecting differences in their physiologic functions. Fragility in the regulation of GTPase spg1 was due to dosage imbalance against GTPase-activating protein (GAP) byr4. Using the gTOW data, we modified a mathematical model and successfully reproduced the robustness of the S. pombe cell cycle with the model. link: http://identifiers.org/pubmed/22146300

Parameters:

NameDescription
kini_dash2 = 10.0; kini_dash3 = 0.0; preRC = 0.0; kini_dash = 10.0Reaction: s89 => s90; s67, s56, s63, Rate Law: (kini_dash*s56+kini_dash2*s67+kini_dash3*s63)*preRC
kscig = 0.002; kscig_dash = 0.04; Cdc10T = 1.0Reaction: s55 => s67; s71, Rate Law: kscig*Cdc10T+kscig_dash*s71
kpyp2 = 0.01; k25 = 0.0Reaction: s60 => s56; s83, s64, Rate Law: (kpyp2+k25)*s60
kpyp = 0.6; beta = 10.0; UDNA = 0.0; k25 = 0.0; k255 = 0.1Reaction: s153 => s149; s64, s83, Rate Law: k25*k255*s153+kpyp*s153/(1+beta*UDNA)
kisrw_dash = 40.0; Puc1 = 1.0; kisrw_dash2 = 1.0; kisrw_dash4 = 4.0; kisrw = 1.5; Jisrw = 0.01; kisrw_dash3 = 4.0Reaction: s47 => s65; s56, s49, s75, s67, Rate Law: (kisrw+kisrw_dash*s67+kisrw_dash2*s56+kisrw_dash3*Puc1+kisrw_dash4*s75)*s47/(Jisrw+s47)
kipre = 1.0; n = 4.0; kipre_dash = 1.0; kori = 125.0; Jipre = 0.01Reaction: s91 => s92; s67, s56, s63, Rate Law: kori/(1+((kipre*s56+kipre_dash*s67)/Jipre)^n)*s91
ksrum = 1.0Reaction: s52 => s166, Rate Law: ksrum
Vdc18 = 0.0Reaction: s84 => s88; s130, Rate Law: Vdc18*s84
Vdrum = 0.0Reaction: s161 => s56 + s61; s4, Rate Law: Vdrum*s161
kdci1_dash = 5.0; kdci1 = 0.1; kdci1_dash2 = 0.2Reaction: s75 => s77; s48, s47, Rate Law: (kdci1+kdci1_dash*s48+kdci1_dash2*s47)*s75
kasrw = 1.25; kasrw_dash = 30.0; Jasrw = 0.01; Srw1T = 1.0Reaction: s65 => s47; s48, Rate Law: (kasrw+kasrw_dash*s48)*(Srw1T-s47)/(Jasrw+(Srw1T-s47))
kic10 = 0.01; Jic10 = 0.01; kic10_dash = 3.0Reaction: s71 => s70; s67, Rate Law: (kic10+kic10_dash*s67)*s71/(Jic10+s71)
ksflp = 0.0015; ksflp_dash = 0.015Reaction: s78 => s81; s48, Rate Law: ksflp+ksflp_dash*s48
kac10 = 0.125; Jac10 = 0.01; Cdc10T = 1.0Reaction: s70 => s71, Rate Law: kac10*(Cdc10T-s71)/(Jac10+(Cdc10T-s71))
Vi25 = 0.3; UDNA = 0.0; Vi25_dash2 = 1.0; Ji25 = 0.03; Vi25_dash = 0.24Reaction: s83 => s82; s81, s157, Rate Law: (Vi25_dash+Vi25_dash2*s81+Vi25*UDNA)*s83/(Ji25+s83)
lcm = 1.0; lcp = 3.0Reaction: s166 + s67 => s149, Rate Law: lcp*s67*s166-lcm*s149
kaie = 0.0975; kaie_dash = 0.05; Jaie = 0.01Reaction: s51 => s50; s75, s56, Rate Law: (kaie*s56+kaie_dash*s75)*(1-s50)/(Jaie+(1-s50))
Jawee = 0.04; Vawee_dash = 0.24; Wee1T = 1.0; Vawee_dash2 = 1.0Reaction: s79 => s80; s81, Rate Law: (Vawee_dash+Vawee_dash2*s81)*(Wee1T-s80)/(Jawee+(Wee1T-s80))
ksc18 = 0.005; ksc18_dash = 0.075; Cdc10T = 1.0Reaction: s85 => s84; s71, Rate Law: ksc18*((Cdc10T-s71)+s71)+ksc18_dash*s71
lp = 500.0; lm = 100.0Reaction: s56 + s166 => s161, Rate Law: lp*s56*s166-lm*s161
Cdc25T = 1.0; Ja25 = 0.03; Va25_dash2 = 1.0Reaction: s82 => s83; s56, Rate Law: Va25_dash2*s56*(Cdc25T-s83)/(Ja25+(Cdc25T-s83))
Vamik_dash = 0.75; Vamik = 0.25; Cdc10T = 1.0Reaction: s73 => s72; s71, Rate Law: Vamik*Cdc10T+Vamik_dash*s71
kdcig_dash = 1.0; kdcig = 0.02Reaction: s149 => s166 + s94; s48, Rate Law: (kdcig+kdcig_dash*s48)*s149
kscyc = 0.03Reaction: s57 => s56, Rate Law: kscyc
kdflp = 0.1Reaction: s81 => s93, Rate Law: kdflp*s81
ksci1 = 0.0015Reaction: s76 => s75, Rate Law: ksci1
Vimik_dash3 = 0.25; Vimik_dash2 = 10.0; Vimik = 0.75; Vimik_dash = 10.0Reaction: s72 => s74; s67, s56, s60, Rate Law: (Vimik+Vimik_dash*s67+Vimik_dash2*s56+Vimik_dash3*s60)*s72
Jislp = 0.01; kislp = 0.2Reaction: s48 => s66, Rate Law: kislp*s48/(Jislp+s48)
Jiie = 0.01; kiie = 0.04Reaction: s50 => s51, Rate Law: kiie*s50/(Jiie+s50)
kmik_dash2 = 4.0Reaction: s149 => s153; s72, Rate Law: kmik_dash2*s72*s149
Vdcyc = 0.0Reaction: s161 => s166 + s46; s9, Rate Law: Vdcyc*s161
krepl = 2.0Reaction: s90 => s91, Rate Law: krepl*s90
kaslp = 1.0; Slp1T = 1.0; Jaslp = 0.01Reaction: s66 => s48; s50, Rate Law: kaslp*s50*(Slp1T-s48)/(Jaslp+(Slp1T-s48))
Viwee_dash2 = 1.0; Jiwee = 0.03; Viwee_dash = 0.0Reaction: s80 => s79; s56, Rate Law: (Viwee_dash+Viwee_dash2*s56)*s80/(Jiwee+s80)
kmik_dash = 0.01; kwee = 0.0Reaction: s161 => s137; s80, s72, Rate Law: (kmik_dash*s72+kwee)*s161

States:

NameDescription
s78[mRNA cleavage and polyadenylation factor clp1]
s76[G2/mitotic-specific cyclin cig1]
s83[M-phase inducer phosphatase]
s92[deoxyribonucleic acid]
s57[G2/mitotic-specific cyclin cdc13]
s153[Cyclin-dependent kinase inhibitor rum1; G2/mitotic-specific cyclin cig2]
s50IE
s93sa370_degraded
s71[Start control protein cdc10]
s47[WD repeat-containing protein srw1]
s81[mRNA cleavage and polyadenylation factor clp1]
s52[Cyclin-dependent kinase inhibitor rum1]
s72[Mitosis inhibitor protein kinase mik1]
s46sa4_degraded
s77sa353_degraded
s70[Start control protein cdc10]
s89[nuclear pre-replicative complex]
s51iIE
s166[Cyclin-dependent kinase inhibitor rum1]
s48[WD repeat-containing protein slp1]
s67[G2/mitotic-specific cyclin cig2]
s55[G2/mitotic-specific cyclin cig2]
s84[Cell division control protein 18]
s149[G2/mitotic-specific cyclin cig2; Cyclin-dependent kinase inhibitor rum1]
s91[deoxyribonucleic acid]
s80[Mitosis inhibitor protein kinase wee1]
s75[G2/mitotic-specific cyclin cig1]
s94sa44_degraded
s73[Mitosis inhibitor protein kinase mik1]
s161[Cyclin-dependent kinase inhibitor rum1; G2/mitotic-specific cyclin cdc13]
s56[G2/mitotic-specific cyclin cdc13]
s79[Mitosis inhibitor protein kinase wee1]
s82[M-phase inducer phosphatase]
s137[G2/mitotic-specific cyclin cdc13; Cyclin-dependent kinase inhibitor rum1]
s74sa347_degraded
s90[origin recognition complex]
s88sa386_degraded
s66[WD repeat-containing protein slp1]
s85[Cell division control protein 18]
s60[G2/mitotic-specific cyclin cdc13; Phosphoprotein]
s65[WD repeat-containing protein srw1]

Morris1981_MuscleFibre_Voltage_full: BIOMD0000000324v0.0.1

This is the full model (eq. 1 and 2) of the voltage oscillations in barnacle muscle fibers described in the article: Vo…

Details

Barnacle muscle fibers subjected to constant current stimulation produce a variety of types of oscillatory behavior when the internal medium contains the Ca++ chelator EGTA. Oscillations are abolished if Ca++ is removed from the external medium, or if the K+ conductance is blocked. Available voltage-clamp data indicate that the cell's active conductance systems are exceptionally simple. Given the complexity of barnacle fiber voltage behavior, this seems paradoxical. This paper presents an analysis of the possible modes of behavior available to a system of two noninactivating conductance mechanisms, and indicates a good correspondence to the types of behavior exhibited by barnacle fiber. The differential equations of a simple equivalent circuit for the fiber are dealt with by means of some of the mathematical techniques of nonlinear mechanics. General features of the system are (a) a propensity to produce damped or sustained oscillations over a rather broad parameter range, and (b) considerable latitude in the shape of the oscillatory potentials. It is concluded that for cells subject to changeable parameters (either from cell to cell or with time during cellular activity), a system dominated by two noninactivating conductances can exhibit varied oscillatory and bistable behavior. link: http://identifiers.org/pubmed/7260316

Morris1981_MuscleFibre_Voltage_reduced: BIOMD0000000280v0.0.1

This is the reduced model of the voltage oscillations in barnacle muscle fibers, generally known as the Morris-Lecar mod…

Details

Barnacle muscle fibers subjected to constant current stimulation produce a variety of types of oscillatory behavior when the internal medium contains the Ca++ chelator EGTA. Oscillations are abolished if Ca++ is removed from the external medium, or if the K+ conductance is blocked. Available voltage-clamp data indicate that the cell's active conductance systems are exceptionally simple. Given the complexity of barnacle fiber voltage behavior, this seems paradoxical. This paper presents an analysis of the possible modes of behavior available to a system of two noninactivating conductance mechanisms, and indicates a good correspondence to the types of behavior exhibited by barnacle fiber. The differential equations of a simple equivalent circuit for the fiber are dealt with by means of some of the mathematical techniques of nonlinear mechanics. General features of the system are (a) a propensity to produce damped or sustained oscillations over a rather broad parameter range, and (b) considerable latitude in the shape of the oscillatory potentials. It is concluded that for cells subject to changeable parameters (either from cell to cell or with time during cellular activity), a system dominated by two noninactivating conductances can exhibit varied oscillatory and bistable behavior. link: http://identifiers.org/pubmed/7260316

Morris2002_CellCycle_CDK2Cyclin: BIOMD0000000150v0.0.1

Notes from the original DOCQS curator: In this version of the CDK2/Cyclin A complex activation there is discrepancy i…

Details

Eukaryotic cell cycle progression is controlled by the ordered action of cyclin-dependent kinases, activation of which occurs through the binding of the cyclin to the Cdk followed by phosphorylation of a conserved threonine in the T-loop of the Cdk by Cdk-activating kinase (CAK). Despite our understanding of the structural changes, which occur upon Cdk/cyclin formation and activation, little is known about the dynamics of the molecular events involved. We have characterized the mechanism of Cdk2/cyclin A complex formation and activation at the molecular and dynamic level by rapid kinetics and demonstrate here that it is a two-step process. The first step involves the rapid association between the PSTAIRE helix of Cdk2 and helices 3 and 5 of the cyclin to yield an intermediate complex in which the threonine in the T-loop is not accessible for phosphorylation. Additional contacts between the C-lobe of the Cdk and the N-terminal helix of the cyclin then induce the isomerization of the Cdk into a fully mature form by promoting the exposure of the T-loop for phosphorylation by CAK and the formation of the substrate binding site. This conformational change is selective for the cyclin partner. link: http://identifiers.org/pubmed/11959850

Parameters:

NameDescription
kf=0.813; kb=0.557Reaction: CDK2cycA => CDK2cycA_star_, Rate Law: kf*CDK2cycA*geometry-kb*CDK2cycA_star_*geometry
kb=25.0; kf=1.9E7Reaction: Cdk2 + CyclinA => CDK2cycA, Rate Law: kf*Cdk2*CyclinA*geometry-kb*CDK2cycA*geometry

States:

NameDescription
CyclinA[IPR015453]
CDK2cycA star[Cyclin-dependent kinase 1; IPR015453]
Cdk2[Cyclin-dependent kinase 1]
CDK2cycA[Cyclin-dependent kinase 1; IPR015453]

Morris2008 - Fitting protein aggregation data via F-W 2-step mechanism: BIOMD0000000567v0.0.1

Morris2008 - Fitting protein aggregation data via F-W 2-step mechanismThis model is described in the article: [Fitting…

Details

The aggregation of proteins has been hypothesized to be an underlying cause of many neurological disorders including Alzheimer's, Parkinson's, and Huntington's diseases; protein aggregation is also important to normal life function in cases such as G to F-actin, glutamate dehydrogenase, and tubulin and flagella formation. For this reason, the underlying mechanism of protein aggregation, and accompanying kinetic models for protein nucleation and growth (growth also being called elongation, polymerization, or fibrillation in the literature), have been investigated for more than 50 years. As a way to concisely present the key prior literature in the protein aggregation area, Table 1 in the main text summarizes 23 papers by 10 groups of authors that provide 5 basic classes of mechanisms for protein aggregation over the period from 1959 to 2007. However, and despite this major prior effort, still lacking are both (i) anything approaching a consensus mechanism (or mechanisms), and (ii) a generally useful, and thus widely used, simplest/"Ockham's razor" kinetic model and associated equations that can be routinely employed to analyze a broader range of protein aggregation kinetic data. Herein we demonstrate that the 1997 Finke-Watzky (F-W) 2-step mechanism of slow continuous nucleation, A –> B (rate constant k1), followed by typically fast, autocatalytic surface growth, A + B –> 2B (rate constant k2), is able to quantitatively account for the kinetic curves from all 14 representative data sets of neurological protein aggregation found by a literature search (the prion literature was largely excluded for the purposes of this study in order provide some limit to the resultant literature that was covered). The F-W model is able to deconvolute the desired nucleation, k1, and growth, k2, rate constants from those 14 data sets obtained by four different physical methods, for three different proteins, and in nine different labs. The fits are generally good, and in many cases excellent, with R2 values >or=0.98 in all cases. As such, this contribution is the current record of the widest set of protein aggregation data best fit by what is also the simplest model offered to date. Also provided is the mathematical connection between the 1997 F-W 2-step mechanism and the 2000 3-step mechanism proposed by Saitô and co-workers. In particular, the kinetic equation for Saitô's 3-step mechanism is shown to be mathematically identical to the earlier, 1997 2-step F-W mechanism under the 3 simplifying assumptions Saitô and co-workers used to derive their kinetic equation. A list of the 3 main caveats/limitations of the F-W kinetic model is provided, followed by the main conclusions from this study as well as some needed future experiments. link: http://identifiers.org/pubmed/18247636

Parameters:

NameDescription
k1 = 4.0E-5Reaction: A => B; A, Rate Law: Brain*k1*A
k2 = 1.57E-6; k1 = 4.0E-5; A0 = 184713.375796178Reaction: B = A0-(k1/k2+A0)/(1+k1/(k2*A0)*exp((k1+k2*A0)*time)), Rate Law: missing
k2 = 1.57E-6Reaction: A + B => B; A, B, Rate Law: Brain*k2*A*B

States:

NameDescription
B[PR:P04156]
A[PR:P04156]

Morris2009 - α-Synuclein aggregation variable temperature and pH: BIOMD0000000566v0.0.1

Morris2009 - α-Synuclein aggregation variable temperature and pHThis model is described in the article: [Alpha-synuclei…

Details

The aggregation of proteins is believed to be intimately connected to many neurodegenerative disorders. We recently reported an "Ockham's razor"/minimalistic approach to analyze the kinetic data of protein aggregation using the Finke-Watzky (F-W) 2-step model of nucleation (A–>B, rate constant k(1)) and autocatalytic growth (A+B–>2B, rate constant k(2)). With that kinetic model we have analyzed 41 representative protein aggregation data sets in two recent publications, including amyloid beta, alpha-synuclein, polyglutamine, and prion proteins (Morris, A. M., et al. (2008) Biochemistry 47, 2413-2427; Watzky, M. A., et al. (2008) Biochemistry 47, 10790-10800). Herein we use the F-W model to reanalyze protein aggregation kinetic data obtained under the experimental conditions of variable temperature or pH 2.0 to 8.5. We provide the average nucleation (k(1)) and growth (k(2)) rate constants and correlations with variable temperature or varying pH for the protein alpha-synuclein. From the variable temperature data, activation parameters DeltaG(double dagger), DeltaH(double dagger), and DeltaS(double dagger) are provided for nucleation and growth, and those values are compared to the available parameters reported in the previous literature determined using an empirical method. Our activation parameters suggest that nucleation and growth are energetically similar for alpha-synuclein aggregation (DeltaG(double dagger)(nucleation)=23(3) kcal/mol; DeltaG(double dagger)(growth)=22(1) kcal/mol at 37 degrees C). From the variable pH data, the F-W analyses show a maximal k(1) value at pH approximately 3, as well as minimal k(1) near the isoelectric point (pI) of alpha-synuclein. Since solubility and net charge are minimized at the pI, either or both of these factors may be important in determining the kinetics of the nucleation step. On the other hand, the k(2) values increase with decreasing pH (i.e., do not appear to have a minimum or maximum near the pI) which, when combined with the k(1) vs. pH (and pI) data, suggest that solubility and charge are less important factors for growth, and that charge is important in the k(1), nucleation step of alpha-synuclein. The chemically well-defined nucleation (k(1)) rate constants obtained from the F-W analysis are, as expected, different than the 1/lag-time empirical constants previously obtained. However, k(2)xA (where k(2) is the rate constant for autocatalytic growth and A is the initial protein concentration) is related to the empirical constant, k(app) obtained previously. Overall, the average nucleation and average growth rate constants for alpha-synuclein aggregation as a function of pH and variable temperature have been quantitated. Those values support the previously suggested formation of a partially folded intermediate that promotes aggregation under high temperature or acidic conditions. link: http://identifiers.org/pubmed/19101068

Parameters:

NameDescription
k1 = 8.0E-6; k2 = 0.034; A0 = 3.55Reaction: B = A0-(k1/k2+A0)/(1+k1/(k2*A0)*exp((k1+k2*A0)*time)), Rate Law: missing
k1 = 8.0E-6Reaction: A => B; A, Rate Law: Brain*k1*A
k2 = 0.034Reaction: A + B => B; A, B, Rate Law: Brain*k2*A*B

States:

NameDescription
B[Alpha-synuclein]
A[Alpha-synuclein]

Morrison1989 - Folate Cycle: BIOMD0000000018v0.0.1

Morrison1989 - Folate CycleThe model describes the folate cycle kinetics in breast cancer cells.This model is described…

Details

A mathematical description of polyglutamated folate kinetics for human breast carcinoma cells (MCF-7) has been formulated based upon experimental folate, methotrexate (MTX), purine, and pyrimidine pool sizes as well as reaction rate parameters obtained from intact MCF-7 cells and their enzyme isolates. The schema accounts for the interconversion of highly polyglutamated tetrahydrofolate, 5-methyl-FH4, 5-10-CH2FH4, dihydrofolate (FH2), 10-formyl-FH4 (FFH4), and 10-formyl-FH2 (FFH2), as well as formation and transport of the MTX polyglutamates. Inhibition mechanisms have been chosen to reproduce all observed non-, un-, and pure competition inhibition patterns. Steady state folate concentrations and thymidylate and purine synthesis rates in drug-free intact cells were used to determine normal folate Vmax values. The resulting average-cell folate model, examined for its ability to predict folate pool behavior following exposure to 1 microM MTX over 21 h, agreed well with the experiment, including a relative preservation of the FFH4 and CH2FH4 pools. The results depend strongly on thymidylate synthase (TS) reaction mechanism, especially the assumption that MTX di- and triglutamates inhibit TS synthesis as greatly in the intact cell as they do with purified enzyme. The effects of cell cycle dependence of TS and dihydrofolate reductase activities were also examined by introducing G- to S-phase activity ratios of these enzymes into the model. For activity ratios down to at least 5%, cell population averaged folate pools were only slightly affected, while CH2FH4 pools in S-phase cells were reduced to as little as 10% of control values. Significantly, these folate pool dynamics were indicated to arise from both direct inhibition by MTX polyglutamates as well as inhibition by elevated levels of polyglutamated FH2 and FFH2. link: http://identifiers.org/pubmed/2732237

Parameters:

NameDescription
hp=23.2Reaction: FH4 + HCHO => CH2FH4, Rate Law: cell*hp*FH4*HCHO
Vm=4.65Reaction: MTX1 =>, Rate Law: cell*Vm*MTX1
Vm=0.42Reaction: MTX3b => MTX3 + DHFRf, Rate Law: cell*Vm*MTX3b
Km2=100.0; Km1=100.0; Vm=4656.0Reaction: FGAR => AICAR; glutamine, Rate Law: cell*Vm*glutamine/Km1/(1+glutamine/Km1)*FGAR/Km2/(1+FGAR/Km2)
Vm=0.118Reaction: MTX3 => MTX4, Rate Law: cell*Vm*MTX3
Vm=163000.0Reaction: MTX4 + DHFRf => MTX4b, Rate Law: cell*Vm*DHFRf*MTX4
Ki1=5.0; Ki24=31.0; Km1=4.9; Ki1f=1.0; Ki23=43.0; Vm=4126.0; Km2=52.0; Ki21=84.0; Ki22=60.0; Ki25=22.0Reaction: CHOFH4 + GAR => FGAR + FH4; FH2f, FFH2, MTX1, MTX2, MTX3, MTX4, MTX5, Rate Law: cell*Vm*CHOFH4*GAR/(GAR*CHOFH4+CHOFH4*Km2+(GAR+Km2)*Km1*(1+MTX1/Ki21+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1+FFH2/Ki1f))
Vm=23100.0Reaction: MTX1 + DHFRf => MTX1b, Rate Law: cell*Vm*DHFRf*MTX1
Vm=44300.0Reaction: MTX2 + DHFRf => MTX2b, Rate Law: cell*Vm*DHFRf*MTX2
Km2=210.0; Vm=18330.0; Km1=1.7Reaction: FH4 + serine => CH2FH4, Rate Law: cell*Vm*serine/Km2/(1+serine/Km2)*FH4/Km1/(1+FH4/Km1)
Vm=314000.0Reaction: MTX5 + DHFRf => MTX5b, Rate Law: cell*Vm*DHFRf*MTX5
Ki1=0.4; Vm=224.8; Ki21=59.0; Ki22=21.3; Ki24=2.77; Ki25=1.0; Km1=50.0; Km2=50.0; Ki23=7.68Reaction: CH2FH4 + NADPH => CH3FH4; FH2f, MTX1, MTX2, MTX3, MTX4, MTX5, Rate Law: cell*Vm*CH2FH4*NADPH/(NADPH*CH2FH4+CH2FH4*Km2+(NADPH+Km2)*Km1*(1+MTX1/Ki21+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1))
Vm=0.129Reaction: MTX1 => MTX2, Rate Law: cell*Vm*MTX1
Vm=0.0Reaction: MTX2 =>, Rate Law: cell*Vm*MTX2
Vm=0.03Reaction: DHFRf => ; FH2b, Rate Law: Vm*cell*(DHFRf+FH2b)
Vm=1.22E7; Km1=3200.0; Km2=10000.0Reaction: CH2FH4 => FH4; glycine, Rate Law: cell*Vm*glycine/Km2/(1+glycine/Km2)*CH2FH4/Km1/(1+CH2FH4/Km1)
kter=2109.4Reaction: FH2f => FH4; FH2b, Rate Law: cell*kter*FH2b
Vm=0.195Reaction: MTX2 => MTX1, Rate Law: cell*Vm*MTX2
Ki1=2.89; Ki22=31.5; Ki25=5.89; Ki23=2.33; Km2=24.0; Vm=31675.0; Km1=5.5; Ki1f=5.3; Ki24=3.61; Ki21=40.0Reaction: CHOFH4 + AICAR => FH4; FH2f, FFH2, MTX1, MTX2, MTX3, MTX4, MTX5, Rate Law: cell*Vm*CHOFH4*AICAR/(AICAR*CHOFH4+CHOFH4*Km2+(AICAR+Km2)*Km1*(1+MTX1/Ki21+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1+FFH2/Ki1f))
Vm=0.369Reaction: MTX2 => MTX3, Rate Law: cell*Vm*MTX2
Ki1=2.89; Ki22=31.5; Ki25=5.89; Ki23=2.33; Km2=24.0; Vm=9503.0; Ki24=3.61; Km1=5.3; Ki21=40.0; Ki1f=5.5Reaction: FFH2 + AICAR => FH2f; FH2f, MTX1, MTX2, MTX3, MTX4, MTX5, Rate Law: cell*Vm*FFH2*AICAR/(AICAR*FFH2+FFH2*Km2+(AICAR+Km2)*Km1*(1+MTX1/Ki21+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1+FFH2/Ki1f))
Vm=85100.0Reaction: MTX3 + DHFRf => MTX3b, Rate Law: cell*Vm*DHFRf*MTX3
Vm=0.031Reaction: MTX4 => MTX3, Rate Law: cell*Vm*MTX4
Vm=65.0Reaction: FH2f => FFH2, Rate Law: cell*Vm*FH2f
Vm=22600.0; Km1=125.0; Km2=2900.0Reaction: CH3FH4 + homocysteine => FH4, Rate Law: cell*Vm*homocysteine/Km2/(1+homocysteine/Km2)*CH3FH4/Km1/(1+CH3FH4/Km1)
Vm=0.185Reaction: MTX4 => MTX5, Rate Law: cell*Vm*MTX4
Vm=0.191Reaction: MTX5 => MTX4, Rate Law: cell*Vm*MTX5
Km2=21.8; Km1=3.0; Vm=68500.0Reaction: CH2FH4 + NADP => CHOFH4, Rate Law: cell*Vm*CH2FH4/Km1/(1+CH2FH4/Km1)*NADP/Km2/(1+NADP/Km2)
Ki1f=1.6; Ki24=0.065; Ki1=3.0; Ki25=0.047; Ki22=0.08; Vm=58.0; Km1=2.5; Ki21=13.0; Ki23=0.07; Km2=1.8Reaction: CH2FH4 + dUMP => FH2f; FH2f, FFH2, MTX1, MTX2, MTX3, MTX4, MTX5, Rate Law: cell*Vm*CH2FH4*dUMP/(dUMP*CH2FH4*(1+MTX1/Ki21+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1)+Km1*dUMP*(FFH2/Ki1f*MTX1/Ki21+(1+FFH2/Ki1f)*(1+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1))+Km1*Km2*(1+MTX2/Ki22+MTX3/Ki23+MTX4/Ki24+MTX5/Ki25+FH2f/Ki1))
Km2=56.0; Vm=3600.0; Km1=230.0; Km3=1600.0Reaction: FH4 + formate + ATP => CHOFH4, Rate Law: cell*Vm/((1+Km1/FH4)*(1+Km2/ATP)*(1+Km3/formate))
Vm=82.2; Km=8.2Reaction: EMTX => MTX1, Rate Law: ext*Vm*EMTX/(Km+EMTX)
hl=0.3Reaction: CH2FH4 => FH4 + HCHO, Rate Law: cell*hl*CH2FH4
Vm=0.025Reaction: MTX3 => MTX2, Rate Law: cell*Vm*MTX3
k0=0.0192; k1=0.04416Reaction: => DHFRf; EMTX, Rate Law: cell*(k0+k1*EMTX)
Vm=0.063Reaction: MTX3 =>, Rate Law: cell*Vm*MTX3

States:

NameDescription
CH3FH4[5-methyltetrahydrofolic acid; 5-Methyltetrahydrofolate]
FH4[5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
MTX4b[Methotrexate]
MTX3[Methotrexate]
DHFRf[Dihydrofolate reductase]
AICAR[AICA ribonucleotide; 1-(5'-Phosphoribosyl)-5-amino-4-imidazolecarboxamide]
NADPH[NADPH; NADPH]
MTX5[Methotrexate]
MTX2b[Methotrexate]
MTX1[Methotrexate]
MTX4[Methotrexate]
homocysteine[homocysteine; Homocysteine]
DHFRtot[Dihydrofolate reductase]
FGAR[5'-Phosphoribosyl-N-formylglycinamide]
GAR[5'-Phosphoribosylglycinamide]
CHOFH4[10-formyltetrahydrofolic acid; 10-Formyltetrahydrofolate]
MTX2[Methotrexate]
FFH2[10-formyldihydrofolic acid; 10-Formyldihydrofolate]
MTX3b[Methotrexate]
FH2f[dihydrofolic acid; Dihydrofolate]
CH2FH4[(6R)-5,10-methylenetetrahydrofolate(2-); 5,10-Methylenetetrahydrofolate]
MTX5b[Methotrexate]
MTX1b[Methotrexate]
HCHO[formaldehyde; Formaldehyde]
dUMP[dUMP; dUMP]

Mosca2012 - Central Carbon Metabolism Regulated by AKT: BIOMD0000000426v0.0.1

Mosca2012 - Central Carbon Metabolism Regulated by AKTThe role of the PI3K/Akt/PKB signalling pathway in oncogenesis has…

Details

Signal transduction and gene regulation determine a major reorganization of metabolic activities in order to support cell proliferation. Protein Kinase B (PKB), also known as Akt, participates in the PI3K/Akt/mTOR pathway, a master regulator of aerobic glycolysis and cellular biosynthesis, two activities shown by both normal and cancer proliferating cells. Not surprisingly considering its relevance for cellular metabolism, Akt/PKB is often found hyperactive in cancer cells. In the last decade, many efforts have been made to improve the understanding of the control of glucose metabolism and the identification of a therapeutic window between proliferating cancer cells and proliferating normal cells. In this context, we have modeled the link between the PI3K/Akt/mTOR pathway, glycolysis, lactic acid production, and nucleotide biosynthesis. We used a computational model to compare two metabolic states generated by two different levels of signaling through the PI3K/Akt/mTOR pathway: one of the two states represents the metabolism of a growing cancer cell characterized by aerobic glycolysis and cellular biosynthesis, while the other state represents the same metabolic network with a reduced glycolytic rate and a higher mitochondrial pyruvate metabolism. Biochemical reactions that link glycolysis and pentose phosphate pathway revealed their importance for controlling the dynamics of cancer glucose metabolism. link: http://identifiers.org/pubmed/23181020

Parameters:

NameDescription
Kapp=195172.0; Kadp=0.4; Katp=0.86; parameter_44 = 27.81; Kiatp=2.5; L=1.0; parameter_17 = 1000.0; Kfbp=4.0E-4; Kpyr=10.0; Kpep=0.014Reaction: species_30 + species_3 => species_31 + species_4; species_6, species_3, species_30, species_4, species_6, species_31, Rate Law: compartment_1*parameter_44*(parameter_17*species_3/Kadp/(1+parameter_17*species_3/Kadp)*parameter_17*species_30/Kpep*(1+parameter_17*species_30/Kpep)^3/(L*(1+parameter_17*species_4/Kiatp)^4/(1+parameter_17*species_6/Kfbp)^4+(1+parameter_17*species_30/Kpep)^4)-parameter_17*species_4*parameter_17*species_31/(Katp*Kpyr*Kapp)/(parameter_17*species_4/Katp+parameter_17*species_31/Kpyr+parameter_17*species_4*parameter_17*species_31/(Katp*Kpyr)+1))
Kq=0.0035; parameter_31 = 86.85; Kapp=651.0; Ka=1.0E-4; Kb=0.0011; Kp=2.0E-5Reaction: species_1 + species_4 => species_2 + species_3; species_1, species_4, species_2, species_3, Rate Law: compartment_1*parameter_31/(Ka*Kb)*(species_1*species_4-species_2*species_3/Kapp)/(1+species_1/Ka+species_4/Kb+species_1*species_4/(Ka*Kb)+species_2/Kp+species_3/Kq+species_2*species_3/(Kp*Kq)+species_1*species_3/(Ka*Kq)+species_2*species_4/(Kp*Kb))
alfa=1.0; beta=1.0; parameter_45 = 340.3; parameter_83 = 0.0047; parameter_85 = 2.0E-6; parameter_86 = 3.0E-4; parameter_84 = 7.0E-5; parameter_26 = 54.0471638909003Reaction: species_31 + species_18 => species_32 + species_19; species_18, species_31, species_32, species_19, Rate Law: compartment_1*(parameter_45*species_18*species_31/(alfa*parameter_85*parameter_86)-parameter_26*species_32*species_19/(beta*parameter_83*parameter_84))/(1+species_18/parameter_85+species_31/parameter_86+species_18*species_31/(alfa*parameter_85*parameter_86)+species_32*species_19/(beta*parameter_83*parameter_84)+species_32/parameter_83+species_19/parameter_84)
Kery4p=1.0E-6; parameter_81 = 5.0E-5; Kfbp=6.0E-5; parameter_32 = 7778.0; parameter_82 = 4.0E-4; Kpg=1.5E-5; parameter_13 = 17486.5107913669Reaction: species_2 => species_5; species_7, species_6, species_8, species_2, species_5, species_7, species_6, species_8, Rate Law: compartment_1*(parameter_32*species_2/parameter_82-parameter_13*species_5/parameter_81)/(1+species_2/parameter_82+species_5/parameter_81+species_7/Kery4p+species_6/Kfbp+species_8/Kpg)
Keq=2.26; Vf=141.2Reaction: species_3 => species_4 + species_20; species_3, species_4, species_20, Rate Law: compartment_1*Vf*species_3^2*(1-species_4*species_20/Keq)/(((1+species_3)^2+(1+species_4)*(1+species_20))-1)
K3=1.733E-7; K6=0.4653; K2=4.765E-8; K7=2.524; Vmax=58.27; K5=0.8683; K1=8.23E-9; Keq_TAL=2.703; K4=6.095E-9Reaction: species_17 + species_16 => species_5 + species_7; species_17, species_16, species_7, species_5, Rate Law: compartment_1*Vmax*(species_17*species_16-species_7*species_5/Keq_TAL)/((K1+species_16)*species_17+(K2+K6*species_5)*species_16+(K3+K5*species_5)*species_7+K4*species_5+K7*species_17*species_7)
parameter_30 = 23.03; keq=1.0; KGlc=0.0093; KGlc_e=0.01Reaction: species_9 => species_1; species_9, species_1, Rate Law: compartment_1*parameter_30*(species_9-species_1/keq)/(KGlc_e*(1+species_1/KGlc)+species_9)
KNADP=3.67E-9; KG6P=6.67E-8; KATP=7.49E-7; Kapp=2000.0; KNADPH=3.12E-9; KPGA23=2.289E-6; parameter_33 = 1.008Reaction: species_2 + species_10 => species_8 + species_11; species_4, species_12, species_2, species_10, species_8, species_11, species_4, species_12, Rate Law: compartment_1*parameter_33/KG6P/KNADP*(species_2*species_10-species_8*species_11/Kapp)/(1+species_10*(1+species_2/KG6P)/KNADP+species_4/KATP+species_11/KNADPH+species_12/KPGA23)
KRu5P=1.9E-7; KX5P=5.0E-7; Keq_RUPE=2.7; Vmax=1.471Reaction: species_13 => species_14; species_13, species_14, Rate Law: compartment_1*Vmax*(species_13-species_14/Keq_RUPE)/(species_13+KRu5P*(1+species_14/KX5P))
parameter_57 = 6.3E-5; parameter_15 = 0.203875968992248; parameter_56 = 3.0E-5; parameter_55 = 7.364Reaction: species_22 => species_2; species_22, species_2, Rate Law: compartment_1*(parameter_55*species_22/parameter_57-parameter_15*species_2/parameter_56)/(1+species_22/parameter_57+species_2/parameter_56)
Vmax=0.7646; Keq_R5PI=3.0; KRu5P=7.8E-7; KR5P=2.2E-6Reaction: species_13 => species_15; species_13, species_15, Rate Law: compartment_1*Vmax*(species_13-species_15/Keq_R5PI)/(species_13+KRu5P*(1+species_15/KR5P))
KiPi=0.0047; KGLYb=1.5E-4; parameter_4 = 0.0177545693277311; parameter_60 = 0.0101; parameter_61 = 0.0017; parameter_58 = 0.03347; parameter_59 = 1.5E-4; parameter_62 = 0.004Reaction: species_24 + species_23 => species_24 + species_22; species_24, species_23, species_22, Rate Law: compartment_1*(parameter_58*species_24*species_23/(parameter_61*parameter_62)-parameter_4*species_24*species_22/(KGLYb*parameter_60))/(1+species_24/parameter_61+species_23/KiPi+species_24/parameter_59+species_22/parameter_60+species_24*species_23/(parameter_61*KiPi)+species_24*species_22/(parameter_59*parameter_60))
Kapp=100000.0; KR5P=5.7E-7; Vmax=0.5104; KATP=3.0E-8Reaction: species_15 + species_4 => species_20 + species_21; species_15, species_4, species_21, species_20, Rate Law: compartment_1*Vmax*(species_15*species_4-species_21*species_20/Kapp)/((KATP+species_4)*(KR5P+species_15))
Kf=17400.0; Kr=158.0; Keq=267100.0; parameter_41 = 32040.0; parameter_17 = 1000.0Reaction: species_22 + species_4 => species_24 + species_3 + species_23; species_22, species_4, species_24, species_23, species_3, Rate Law: compartment_1*parameter_41/Kf*parameter_17*species_22*parameter_17*species_4*parameter_17*species_24*(1-(parameter_17*species_23)^2*parameter_17*species_3/(parameter_17*species_22*parameter_17*species_4*Keq))/(1+parameter_17*species_22*parameter_17*species_4*parameter_17*species_24/Kf+parameter_17*species_24*(parameter_17*species_23)^2*parameter_17*species_3/Kr)
Keq_TKL2=29.7; K3=5.48E-8; parameter_36 = 0.1761; K1=1.84E-9; K7=0.215; K6=0.122; K4=3.0E-10; K5=0.0287; K2=3.055E-7Reaction: species_14 + species_7 => species_16 + species_5; species_7, species_14, species_16, species_5, Rate Law: compartment_1*parameter_36*(species_7*species_14-species_16*species_5/Keq_TKL2)/((K1+species_7)*species_14+(K2+K6*species_5)*species_7+(K3+K5*species_5)*species_16+K4*species_5+K7*species_14*species_16)
Katp=2.1E-5; parameter_17 = 1000.0; alfa=0.32; Kfbp=5.0; Kf26bp=8.4E-7; parameter_42 = 107.6; Kf6p=1.0; Kcit=6.8; L=4.1; Kapp=247.0; Kiatp=20.0; Kadp=5.0; beta=0.98Reaction: species_5 + species_4 => species_6 + species_3; species_26, species_25, species_4, species_26, species_5, species_25, species_3, species_6, Rate Law: compartment_1*parameter_42*parameter_17*species_4/Katp/(1+parameter_17*species_4/Katp)*(1+beta*parameter_17*species_26/(alfa*Kf26bp))/(1+parameter_17*species_26/(alfa*Kf26bp))*(parameter_17*species_5*(1+parameter_17*species_26/(alfa*Kf26bp))/(Kf6p*(1+parameter_17*species_26/Kf26bp))*(1+parameter_17*species_5*(1+parameter_17*species_26/(alfa*Kf26bp))/(Kf6p*(1+parameter_17*species_26/Kf26bp)))^3/(L*(1+parameter_17*species_25/Kcit)^4*(1+parameter_17*species_4/Kiatp)^4/(1+parameter_17*species_26/Kf26bp)^4+(1+parameter_17*species_5*(1+parameter_17*species_26/(alfa*Kf26bp))/(Kf6p*(1+parameter_17*species_26/Kf26bp)))^4)-parameter_17*species_3*parameter_17*species_6/(Kadp*Kfbp*Kapp)/(parameter_17*species_3/Kadp+parameter_17*species_6/Kfbp+parameter_17*species_3*parameter_17*species_6/(Kadp*Kfbp)+1))
k1=6210.0Reaction: species_4 => species_3 + species_23; species_4, Rate Law: compartment_1*k1*species_4
parameter_2 = 1.932E-5Reaction: species_10 = parameter_2-species_11, Rate Law: missing
K6=0.00774; K4=4.96E-9; K1=4.177E-7; Keq_TKL=2.08; parameter_35 = 1056.0; K5=0.41139; K2=3.055E-7; K3=1.2432E-5; K7=48.8Reaction: species_15 + species_14 => species_16 + species_17; species_15, species_14, species_16, species_17, Rate Law: compartment_1*parameter_35*(species_15*species_14-species_16*species_17/Keq_TKL)/((K1+species_15)*species_14+(K2+K6*species_17)*species_15+(K3+K5*species_17)*species_16+K4*species_17+K7*species_14*species_16)
parameter_8 = 11.5595061728395; parameter_68 = 8.0E-5; parameter_69 = 1.6E-4; parameter_70 = 9.0E-6; parameter_37 = 14.63Reaction: species_6 => species_16 + species_27; species_6, species_27, species_16, Rate Law: compartment_1*(parameter_37*species_6/parameter_70-parameter_8*species_27*species_16/(parameter_68*parameter_69))/(1+species_6/parameter_70+species_27/parameter_68+species_16/parameter_69+species_27*species_16/(parameter_68*parameter_69))
parameter_9 = 49.2079666512274; parameter_38 = 5.976; parameter_72 = 5.1E-4; parameter_71 = 0.0016Reaction: species_16 => species_27; species_16, species_27, Rate Law: compartment_1*(parameter_38*species_16/parameter_72-parameter_9*species_27/parameter_71)/(1+species_16/parameter_72+species_27/parameter_71)
parameter_46 = 4982000.0; Keq=300.0Reaction: species_18 => species_19; species_18, species_19, Rate Law: compartment_1*parameter_46*species_18*(1-species_19/(species_18*Keq))/((1+species_18+1+species_19)-1)
parameter_49 = 1.3E-4; parameter_51 = 7.9E-5; parameter_50 = 2.7E-4; alfa=1.0; beta=1.0; parameter_11 = 71.7220990679741; parameter_52 = 4.0E-5; parameter_40 = 73.41Reaction: species_12 + species_3 => species_28 + species_4; species_12, species_3, species_28, species_4, Rate Law: compartment_1*(parameter_40*species_12*species_3/(alfa*parameter_51*parameter_52)-parameter_11*species_28*species_4/(beta*parameter_49*parameter_50))/(1+species_12/parameter_51+species_3/parameter_52+species_12*species_3/(alfa*parameter_51*parameter_52)+species_28*species_4/(beta*parameter_49*parameter_50)+species_28/parameter_49+species_4/parameter_50)
parameter_1 = 0.0114Reaction: species_3 = parameter_1-species_4, Rate Law: missing
K6PG1=1.0E-8; Kapp=141.7; KNADPH=4.5E-9; parameter_34 = 31.02; KPGA23=1.2E-7; KATP=1.54E-7; KNADP=1.8E-8; K6PG2=5.8E-8Reaction: species_8 + species_10 => species_13 + species_11; species_12, species_4, species_8, species_10, species_13, species_11, species_12, species_4, Rate Law: compartment_1*parameter_34/K6PG1/KNADP*(species_8*species_10-species_13*species_11/Kapp)/((1+species_10/KNADP)*(1+species_8/K6PG1+species_12/KPGA23)+species_4/KATP+species_11*(1+species_8/K6PG2)/KNADPH)
parameter_78 = 154.0; parameter_80 = 1.9E-4; parameter_79 = 1.2E-4; parameter_22 = 58.9795390787319Reaction: species_28 => species_29; species_28, species_29, Rate Law: compartment_1*(parameter_78*species_28/parameter_80-parameter_22*species_29/parameter_79)/(1+species_28/parameter_80+species_29/parameter_79)
parameter_47 = 127800.0; Keq=0.2Reaction: species_11 => species_10; species_11, species_10, Rate Law: compartment_1*parameter_47*species_11*(1-species_10/(species_11*Keq))/((1+species_11+1+species_10)-1)
y=12.5; Keq=1000000.0; parameter_48 = 9801000.0Reaction: species_31 + species_34 + species_23 + species_3 => species_33 + species_4; species_31, species_23, species_3, species_34, species_4, species_33, Rate Law: compartment_1*parameter_48*species_31^(1/y)*species_23*species_3*species_34^(5/(2*y))*(1-species_4*species_33^(3/y)/(species_31^(1/y)*species_34^(5/(2*y))*species_23*species_3*Keq))/(((1+species_31)^(1/y)*(1+species_34)^(5/(2*y))*(1+species_23)*(1+species_3)+(1+species_4)*(1+species_33)^(3/y))-1)
parameter_73 = 2.2E-5; parameter_10 = 135.42497838741; parameter_75 = 1.9E-4; parameter_77 = 0.029; parameter_76 = 9.0E-5; parameter_74 = 1.0E-5; parameter_39 = 109.1Reaction: species_16 + species_19 + species_23 => species_12 + species_18; species_19, species_16, species_23, species_12, species_18, Rate Law: compartment_1*(parameter_39*species_19*species_16*species_23/(parameter_76*parameter_75*parameter_77)-parameter_10*species_12*species_18/(parameter_73*parameter_74))/(1+species_19/parameter_76+species_19*species_16/(parameter_76*parameter_75)+species_19*species_16*species_23/(parameter_76*parameter_75*parameter_77)+species_12*species_18/(parameter_73*parameter_74)+species_18/parameter_74)
parameter_3 = 0.001345Reaction: species_18 = parameter_3-species_19, Rate Law: missing
parameter_24 = 179.83480680891; parameter_43 = 160.9; parameter_53 = 6.0E-5; parameter_54 = 3.8E-5Reaction: species_29 => species_30; species_29, species_30, Rate Law: compartment_1*(parameter_43*species_29/parameter_54-parameter_24*species_30/parameter_53)/(1+species_29/parameter_54+species_30/parameter_53)
parameter_7 = 6.03725213205671E-5; KiG1P=0.0074; nH=1.75; Kamp=1.9E-12; parameter_66 = 0.015; parameter_27 = 0.00311; parameter_64 = 0.0044; parameter_63 = 0.01049; parameter_67 = 0.0046; parameter_65 = 0.0015; KPi=2.0E-4Reaction: species_24 + species_23 => species_24 + species_22; species_24, species_23, species_22, Rate Law: compartment_1*(parameter_63*species_24*species_23/(parameter_66*KPi)-parameter_7*species_24*species_22/(parameter_64*parameter_65))/(1+species_24/parameter_66+species_23/parameter_67+species_24/parameter_64+species_22/KiG1P+species_24*species_23/(parameter_66*KPi)+species_24*species_22/(parameter_64*parameter_65))*parameter_27^nH/Kamp/(1+parameter_27^nH/Kamp)

States:

NameDescription
species 9[endoplasmic reticulum; glucose]
species 27[glycerone phosphate(2-)]
species 31[pyruvate]
species 1[glucose]
species 18[NADH]
species 4[ATP]
species 16[glyceraldehyde 3-phosphate]
species 20[AMP]
species 28[3-phosphoglyceric acid]
species 34[singlet dioxygen]
species 32[lactate]
species 8[6-O-phosphono-D-glucono-1,5-lactone]
species 30[phosphoenolpyruvate]
species 12[683]
species 17[sedoheptulose 7-phosphate]
species 5[keto-D-fructose 6-phosphate]
species 15[aldehydo-D-ribose 5-phosphate(2-)]
species 21[7339]
species 2[alpha-D-glucose 6-phosphate]
species 29[3-ADP-2-phosphoglyceric acid]
species 6[alpha-D-fructofuranose 1,6-bisphosphate]
species 19[NAD]
species 10[NADP]
species 33[carbon dioxide]
species 11[salicyl alcohol]
species 24[glycogen]
species 14[D-xylulose 5-phosphate(2-)]
species 22[D-glucopyranose 1-phosphate]
species 3[ADP]
species 23[phosphate(3-)]
species 7[D-erythrose 4-phosphate]
species 13[D-ribulose 5-phosphate(2-)]

Mouse Iron Distribution - Adequate iron diet (No Tracer): BIOMD0000000736v0.0.1

# Mouse Iron Distribution Dynamics Dynamic model of iron distribution in mice. This model includes only normal iron with…

Details

Iron is an essential element of most living organisms but is a dangerous substance when poorly liganded in solution. The hormone hepcidin regulates the export of iron from tissues to the plasma contributing to iron homeostasis and also restricting its availability to infectious agents. Disruption of iron regulation in mammals leads to disorders such as anemia and hemochromatosis, and contributes to the etiology of several other diseases such as cancer and neurodegenerative diseases. Here we test the hypothesis that hepcidin alone is able to regulate iron distribution in different dietary regimes in the mouse using a computational model of iron distribution calibrated with radioiron tracer data.A model was developed and calibrated to the data from adequate iron diet, which was able to simulate the iron distribution under a low iron diet. However simulation of high iron diet shows considerable deviations from the experimental data. Namely the model predicts more iron in red blood cells and less iron in the liver than what was observed in experiments.These results suggest that hepcidin alone is not sufficient to regulate iron homeostasis in high iron conditions and that other factors are important. The model was able to simulate anemia when hepcidin was increased but was unable to simulate hemochromatosis when hepcidin was suppressed, suggesting that in high iron conditions additional regulatory interactions are important. link: http://identifiers.org/pubmed/28521769

Parameters:

NameDescription
kInBM = 15.7690636138556Reaction: Fe2Tf => FeBM + Tf, Rate Law: kInBM*Fe2Tf*Plasma
kInLiver = 2.97790545667672Reaction: Fe1Tf => FeLiver + Tf, Rate Law: kInLiver*Fe1Tf*Plasma
VLiverNTBI = 0.0261147638001175; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeLiver => NTBI; Hepcidin, Rate Law: VLiverNTBI*Liver*FeLiver/((Km+FeLiver)*(1+Hepcidin/Ki))
kDuoLoss = 0.0270113302698216Reaction: FeDuo => FeOutside, Rate Law: kDuoLoss*FeDuo*Duodenum
kFe1Tf_Fe2Tf = 1.084322005E9Reaction: Fe1Tf + NTBI => Fe2Tf, Rate Law: Plasma*kFe1Tf_Fe2Tf*Fe1Tf*NTBI
kNTBI_Fe1Tf = 1.084322005E9Reaction: NTBI + Tf => Fe1Tf, Rate Law: Plasma*kNTBI_Fe1Tf*NTBI*Tf
VRestNTBI = 0.0109451335200198; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeRest => NTBI; Hepcidin, Rate Law: VRestNTBI*RestOfBody*FeRest/((Km+FeRest)*(1+Hepcidin/Ki))
kBMSpleen = 0.061902954378781Reaction: FeBM => FeSpleen, Rate Law: kBMSpleen*FeBM*BoneMarrow
vRBCSpleen = 0.0235286Reaction: FeRBC => FeSpleen, Rate Law: vRBCSpleen*FeRBC*RBC
vDiet = 0.00377422331938439Reaction: => FeDuo, Rate Law: Duodenum*vDiet
Km = 0.0159421218669513; VDuoNTBI = 0.200241893786814; Ki = 1.0E-9Reaction: FeDuo => NTBI; Hepcidin, Rate Law: VDuoNTBI*Duodenum*FeDuo/((Km+FeDuo)*(1+Hepcidin/Ki))
v=1.7393E-8Reaction: => Hepcidin, Rate Law: Plasma*v
kInRest = 6.16356235352873Reaction: Fe1Tf => FeRest + Tf, Rate Law: kInRest*Fe1Tf*Plasma
k1=0.75616Reaction: Hepcidin =>, Rate Law: Plasma*k1*Hepcidin
kInDuo = 0.0689984226081531Reaction: Fe1Tf => FeDuo + Tf, Rate Law: kInDuo*Fe1Tf*Plasma
VSpleenNTBI = 1.342204923; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeSpleen => NTBI; Hepcidin, Rate Law: VSpleenNTBI*Spleen*FeSpleen/((Km+FeSpleen)*(1+Hepcidin/Ki))
kInRBC = 1.08447580176706Reaction: FeBM => FeRBC, Rate Law: kInRBC*FeBM*BoneMarrow
kRestLoss = 0.023533240736163Reaction: FeRest => FeOutside, Rate Law: RestOfBody*kRestLoss*FeRest

States:

NameDescription
FeRest[iron cation]
Fe2Tf[iron(3+); Serotransferrin]
NTBI[iron cation]
FeSpleen[iron cation]
FeBM[iron cation]
FeRBC[iron cation]
Fe1Tf[Serotransferrin; iron(3+)]
FeLiver[iron cation]
FeDuo[iron cation]
Tf[Serotransferrin]
Hepcidin[Hepcidin]
FeOutside[iron cation]

Mouse Iron Distribution - Adequate iron diet (tracer): BIOMD0000000735v0.0.1

# Mouse Iron Distribution Dynamics Dynamic model of iron distribution in mice. This model includes normal iron and radio…

Details

Iron is an essential element of most living organisms but is a dangerous substance when poorly liganded in solution. The hormone hepcidin regulates the export of iron from tissues to the plasma contributing to iron homeostasis and also restricting its availability to infectious agents. Disruption of iron regulation in mammals leads to disorders such as anemia and hemochromatosis, and contributes to the etiology of several other diseases such as cancer and neurodegenerative diseases. Here we test the hypothesis that hepcidin alone is able to regulate iron distribution in different dietary regimes in the mouse using a computational model of iron distribution calibrated with radioiron tracer data.A model was developed and calibrated to the data from adequate iron diet, which was able to simulate the iron distribution under a low iron diet. However simulation of high iron diet shows considerable deviations from the experimental data. Namely the model predicts more iron in red blood cells and less iron in the liver than what was observed in experiments.These results suggest that hepcidin alone is not sufficient to regulate iron homeostasis in high iron conditions and that other factors are important. The model was able to simulate anemia when hepcidin was increased but was unable to simulate hemochromatosis when hepcidin was suppressed, suggesting that in high iron conditions additional regulatory interactions are important. link: http://identifiers.org/pubmed/28521769

Parameters:

NameDescription
kInBM = 15.7690636138556Reaction: Fe2Tf_0 => FeBM + Tf, Rate Law: kInBM*Fe2Tf_0*Plasma
kInLiver = 2.97790545667672Reaction: Fe1Tf => FeLiver + Tf, Rate Law: kInLiver*Fe1Tf*Plasma
VLiverNTBI = 0.0261147638001175; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeLiver_0 => NTBI_0; FeLiver, Hepcidin, Rate Law: VLiverNTBI*Liver*FeLiver_0/((Km+FeLiver_0+FeLiver)*(1+Hepcidin/Ki))
kDuoLoss = 0.0270113302698216Reaction: FeDuo => FeOutside, Rate Law: kDuoLoss*FeDuo*Duodenum
kFe1Tf_Fe2Tf = 1.084322005E9Reaction: Fe1Tf + NTBI => Fe2Tf_, Rate Law: Plasma*kFe1Tf_Fe2Tf*Fe1Tf*NTBI
kNTBI_Fe1Tf = 1.084322005E9Reaction: NTBI + Tf => Fe1Tf, Rate Law: Plasma*kNTBI_Fe1Tf*NTBI*Tf
VRestNTBI = 0.0109451335200198; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeRest => NTBI; FeRest_0, Hepcidin, Rate Law: VRestNTBI*RestOfBody*FeRest/((Km+FeRest+FeRest_0)*(1+Hepcidin/Ki))
kBMSpleen = 0.061902954378781Reaction: FeBM_0 => FeSpleen, Rate Law: kBMSpleen*FeBM_0*BoneMarrow
vRBCSpleen = 0.0235286Reaction: FeRBC_0 => FeSpleen_0, Rate Law: vRBCSpleen*FeRBC_0*RBC
vDiet = 0.00377422331938439Reaction: => FeDuo_0, Rate Law: Duodenum*vDiet
Km = 0.0159421218669513; VDuoNTBI = 0.200241893786814; Ki = 1.0E-9Reaction: FeDuo => NTBI; FeDuo_0, Hepcidin, Rate Law: VDuoNTBI*Duodenum*FeDuo/((Km+FeDuo+FeDuo_0)*(1+Hepcidin/Ki))
v=1.7393E-8Reaction: => Hepcidin, Rate Law: Plasma*v
kInRest = 6.16356235352873Reaction: Fe2Tf => FeRest + FeRest_0 + Tf, Rate Law: kInRest*Fe2Tf*Plasma
k1=0.75616Reaction: Hepcidin =>, Rate Law: Plasma*k1*Hepcidin
kInDuo = 0.0689984226081531Reaction: Fe1Tf => FeDuo + Tf, Rate Law: kInDuo*Fe1Tf*Plasma
VSpleenNTBI = 1.342204923; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeSpleen => NTBI; FeSpleen_0, Hepcidin, Rate Law: VSpleenNTBI*Spleen*FeSpleen/((Km+FeSpleen+FeSpleen_0)*(1+Hepcidin/Ki))
kInRBC = 1.08447580176706Reaction: FeBM_0 => FeRBC, Rate Law: kInRBC*FeBM_0*BoneMarrow
kRestLoss = 0.023533240736163Reaction: FeRest => FeOutside, Rate Law: RestOfBody*kRestLoss*FeRest

States:

NameDescription
FeRest[iron cation]
FeOutside 0[iron cation]
NTBI 0[iron cation]
Fe2Tf[iron(3+); Serotransferrin]
NTBI[iron cation]
Fe1Tf 0[iron(3+); Serotransferrin]
Fe2Tf 0[Serotransferrin; iron(3+)]
FeSpleen[iron cation]
FeRBC 0[iron cation]
FeLiver 0[iron cation]
FeBM[iron cation]
FeRBC[iron cation]
FeSpleen 0[iron cation]
Fe1Tf[Serotransferrin; iron(3+)]
FeLiver[iron cation]
FeDuo[iron cation]
Tf[Serotransferrin]
Hepcidin[Hepcidin]
FeBM 0[iron cation]
FeDuo 0[iron cation]

Mouse Iron Distribution - Deficient iron diet (No Tracer): BIOMD0000000737v0.0.1

# Mouse Iron Distribution Dynamics Dynamic model of iron distribution in mice. This model includes only normal iron with…

Details

Iron is an essential element of most living organisms but is a dangerous substance when poorly liganded in solution. The hormone hepcidin regulates the export of iron from tissues to the plasma contributing to iron homeostasis and also restricting its availability to infectious agents. Disruption of iron regulation in mammals leads to disorders such as anemia and hemochromatosis, and contributes to the etiology of several other diseases such as cancer and neurodegenerative diseases. Here we test the hypothesis that hepcidin alone is able to regulate iron distribution in different dietary regimes in the mouse using a computational model of iron distribution calibrated with radioiron tracer data.A model was developed and calibrated to the data from adequate iron diet, which was able to simulate the iron distribution under a low iron diet. However simulation of high iron diet shows considerable deviations from the experimental data. Namely the model predicts more iron in red blood cells and less iron in the liver than what was observed in experiments.These results suggest that hepcidin alone is not sufficient to regulate iron homeostasis in high iron conditions and that other factors are important. The model was able to simulate anemia when hepcidin was increased but was unable to simulate hemochromatosis when hepcidin was suppressed, suggesting that in high iron conditions additional regulatory interactions are important. link: http://identifiers.org/pubmed/28521769

Parameters:

NameDescription
kInBM = 15.7690636138556Reaction: Fe2Tf => FeBM + Tf, Rate Law: kInBM*Fe2Tf*Plasma
kInLiver = 2.97790545667672Reaction: Fe2Tf => FeLiver + Tf, Rate Law: kInLiver*Fe2Tf*Plasma
VLiverNTBI = 0.0261147638001175; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeLiver => NTBI; Hepcidin, Rate Law: VLiverNTBI*Liver*FeLiver/((Km+FeLiver)*(1+Hepcidin/Ki))
kDuoLoss = 0.0270113302698216Reaction: FeDuo => FeOutside, Rate Law: kDuoLoss*FeDuo*Duodenum
kFe1Tf_Fe2Tf = 1.084322005E9Reaction: Fe1Tf + NTBI => Fe2Tf, Rate Law: Plasma*kFe1Tf_Fe2Tf*Fe1Tf*NTBI
kNTBI_Fe1Tf = 1.084322005E9Reaction: NTBI + Tf => Fe1Tf, Rate Law: Plasma*kNTBI_Fe1Tf*NTBI*Tf
VRestNTBI = 0.0109451335200198; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeRest => NTBI; Hepcidin, Rate Law: VRestNTBI*RestOfBody*FeRest/((Km+FeRest)*(1+Hepcidin/Ki))
kBMSpleen = 0.061902954378781Reaction: FeBM => FeSpleen, Rate Law: kBMSpleen*FeBM*BoneMarrow
vRBCSpleen = 0.0235286Reaction: FeRBC => FeSpleen, Rate Law: vRBCSpleen*FeRBC*RBC
Km = 0.0159421218669513; VDuoNTBI = 0.200241893786814; Ki = 1.0E-9Reaction: FeDuo => NTBI; Hepcidin, Rate Law: VDuoNTBI*Duodenum*FeDuo/((Km+FeDuo)*(1+Hepcidin/Ki))
v=8.54927E-9Reaction: => Hepcidin, Rate Law: Plasma*v
kInRest = 6.16356235352873Reaction: Fe2Tf => FeRest + Tf, Rate Law: kInRest*Fe2Tf*Plasma
k1=0.75616Reaction: Hepcidin =>, Rate Law: Plasma*k1*Hepcidin
kInDuo = 0.0689984226081531Reaction: Fe1Tf => FeDuo + Tf, Rate Law: kInDuo*Fe1Tf*Plasma
VSpleenNTBI = 1.342204923; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeSpleen => NTBI; Hepcidin, Rate Law: VSpleenNTBI*Spleen*FeSpleen/((Km+FeSpleen)*(1+Hepcidin/Ki))
vDiet = 0.0Reaction: => FeDuo, Rate Law: Duodenum*vDiet
kInRBC = 1.08447580176706Reaction: FeBM => FeRBC, Rate Law: kInRBC*FeBM*BoneMarrow
kRestLoss = 0.023533240736163Reaction: FeRest => FeOutside, Rate Law: RestOfBody*kRestLoss*FeRest

States:

NameDescription
FeRest[iron cation]
Fe2Tf[Serotransferrin; iron(3+)]
NTBI[iron cation]
FeSpleen[iron cation]
FeBM[iron cation]
FeRBC[iron cation]
Fe1Tf[iron(3+); Serotransferrin]
FeLiver[iron cation]
FeDuo[iron cation]
Tf[Serotransferrin]
Hepcidin[Hepcidin]
FeOutside[iron cation]

Mouse Iron Distribution - Rich and Deficient iron diets (tracer): BIOMD0000000734v0.0.1

# Mouse Iron Distribution Dynamics Dynamic model of iron distribution in mice. This model attempts to fit the radioiron…

Details

Iron is an essential element of most living organisms but is a dangerous substance when poorly liganded in solution. The hormone hepcidin regulates the export of iron from tissues to the plasma contributing to iron homeostasis and also restricting its availability to infectious agents. Disruption of iron regulation in mammals leads to disorders such as anemia and hemochromatosis, and contributes to the etiology of several other diseases such as cancer and neurodegenerative diseases. Here we test the hypothesis that hepcidin alone is able to regulate iron distribution in different dietary regimes in the mouse using a computational model of iron distribution calibrated with radioiron tracer data.A model was developed and calibrated to the data from adequate iron diet, which was able to simulate the iron distribution under a low iron diet. However simulation of high iron diet shows considerable deviations from the experimental data. Namely the model predicts more iron in red blood cells and less iron in the liver than what was observed in experiments.These results suggest that hepcidin alone is not sufficient to regulate iron homeostasis in high iron conditions and that other factors are important. The model was able to simulate anemia when hepcidin was increased but was unable to simulate hemochromatosis when hepcidin was suppressed, suggesting that in high iron conditions additional regulatory interactions are important. link: http://identifiers.org/pubmed/28521769

Parameters:

NameDescription
kInBM = 15.7690636138556Reaction: Fe2Tf => FeBM_0 + FeBM + Tf, Rate Law: kInBM*Fe2Tf*Plasma
kInLiver = 2.97790545667672Reaction: Fe2Tf_ => FeLiver + Tf, Rate Law: kInLiver*Fe2Tf_*Plasma
VLiverNTBI = 0.0261147638001175; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeLiver => NTBI; FeLiver_0, Hepcidin, Rate Law: VLiverNTBI*Liver*FeLiver/((Km+FeLiver+FeLiver_0)*(1+Hepcidin/Ki))
kDuoLoss = 0.0270113302698216Reaction: FeDuo => FeOutside, Rate Law: kDuoLoss*FeDuo*Duodenum
kFe1Tf_Fe2Tf = 1.084322005E9Reaction: Fe1Tf + NTBI => Fe2Tf_, Rate Law: Plasma*kFe1Tf_Fe2Tf*Fe1Tf*NTBI
kNTBI_Fe1Tf = 1.084322005E9Reaction: NTBI_0 + Tf => Fe1Tf_0, Rate Law: Plasma*kNTBI_Fe1Tf*NTBI_0*Tf
VRestNTBI = 0.0109451335200198; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeRest => NTBI; FeRest_0, Hepcidin, Rate Law: VRestNTBI*RestOfBody*FeRest/((Km+FeRest+FeRest_0)*(1+Hepcidin/Ki))
kBMSpleen = 0.061902954378781Reaction: FeBM_0 => FeSpleen, Rate Law: kBMSpleen*FeBM_0*BoneMarrow
vRBCSpleen = 0.0235286Reaction: FeRBC => FeSpleen, Rate Law: vRBCSpleen*FeRBC*RBC
vDiet = 0.00377422331938439Reaction: => FeDuo_0, Rate Law: Duodenum*vDiet
Km = 0.0159421218669513; VDuoNTBI = 0.200241893786814; Ki = 1.0E-9Reaction: FeDuo => NTBI; FeDuo_0, Hepcidin, Rate Law: VDuoNTBI*Duodenum*FeDuo/((Km+FeDuo+FeDuo_0)*(1+Hepcidin/Ki))
v=1.7393E-8Reaction: => Hepcidin, Rate Law: Plasma*v
kInRest = 6.16356235352873Reaction: Fe2Tf => FeRest + FeRest_0 + Tf, Rate Law: kInRest*Fe2Tf*Plasma
k1=0.75616Reaction: Hepcidin =>, Rate Law: Plasma*k1*Hepcidin
kInDuo = 0.0689984226081531Reaction: Fe2Tf_0 => FeDuo_0 + Tf, Rate Law: kInDuo*Fe2Tf_0*Plasma
kInRBC = 1.08447580176706Reaction: FeBM => FeRBC_0, Rate Law: kInRBC*FeBM*BoneMarrow
VSpleenNTBI = 1.342204923; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeSpleen => NTBI; FeSpleen_0, Hepcidin, Rate Law: VSpleenNTBI*Spleen*FeSpleen/((Km+FeSpleen+FeSpleen_0)*(1+Hepcidin/Ki))
kRestLoss = 0.023533240736163Reaction: FeRest => FeOutside, Rate Law: RestOfBody*kRestLoss*FeRest

States:

NameDescription
FeRest[iron cation]
FeRBC 0[iron cation]
FeRBC[iron cation]
FeSpleen 0[iron cation]
Fe1Tf[iron(3+); Serotransferrin]
FeLiver[iron cation]
FeBM 0[iron cation]
FeOutside 0[iron cation]
NTBI 0[iron cation]
Fe2Tf[Serotransferrin; iron(3+)]
NTBI[iron cation]
Fe1Tf 0[Serotransferrin; iron(3+)]
Fe2Tf 0[iron(3+); Serotransferrin]
FeSpleen[iron cation]
FeRest 0[iron cation]
FeLiver 0[iron cation]
FeBM[iron cation]
FeDuo[iron cation]
Tf[Serotransferrin]
Hepcidin[Hepcidin]
FeOutside[iron cation]
FeDuo 0[iron cation]

Mouse Iron Distribution - Rich iron diet (No Tracer): BIOMD0000000738v0.0.1

# Mouse Iron Distribution Dynamics Dynamic model of iron distribution in mice. This model includes only normal iron with…

Details

Iron is an essential element of most living organisms but is a dangerous substance when poorly liganded in solution. The hormone hepcidin regulates the export of iron from tissues to the plasma contributing to iron homeostasis and also restricting its availability to infectious agents. Disruption of iron regulation in mammals leads to disorders such as anemia and hemochromatosis, and contributes to the etiology of several other diseases such as cancer and neurodegenerative diseases. Here we test the hypothesis that hepcidin alone is able to regulate iron distribution in different dietary regimes in the mouse using a computational model of iron distribution calibrated with radioiron tracer data.A model was developed and calibrated to the data from adequate iron diet, which was able to simulate the iron distribution under a low iron diet. However simulation of high iron diet shows considerable deviations from the experimental data. Namely the model predicts more iron in red blood cells and less iron in the liver than what was observed in experiments.These results suggest that hepcidin alone is not sufficient to regulate iron homeostasis in high iron conditions and that other factors are important. The model was able to simulate anemia when hepcidin was increased but was unable to simulate hemochromatosis when hepcidin was suppressed, suggesting that in high iron conditions additional regulatory interactions are important. link: http://identifiers.org/pubmed/28521769

Parameters:

NameDescription
kInBM = 15.7690636138556Reaction: Fe2Tf => FeBM + Tf, Rate Law: kInBM*Fe2Tf*Plasma
kInLiver = 2.97790545667672Reaction: Fe2Tf => FeLiver + Tf, Rate Law: kInLiver*Fe2Tf*Plasma
VLiverNTBI = 0.0261147638001175; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeLiver => NTBI; Hepcidin, Rate Law: VLiverNTBI*Liver*FeLiver/((Km+FeLiver)*(1+Hepcidin/Ki))
kDuoLoss = 0.0270113302698216Reaction: FeDuo => FeOutside, Rate Law: kDuoLoss*FeDuo*Duodenum
kFe1Tf_Fe2Tf = 1.084322005E9Reaction: Fe1Tf + NTBI => Fe2Tf, Rate Law: Plasma*kFe1Tf_Fe2Tf*Fe1Tf*NTBI
kNTBI_Fe1Tf = 1.084322005E9Reaction: NTBI + Tf => Fe1Tf, Rate Law: Plasma*kNTBI_Fe1Tf*NTBI*Tf
VRestNTBI = 0.0109451335200198; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeRest => NTBI; Hepcidin, Rate Law: VRestNTBI*RestOfBody*FeRest/((Km+FeRest)*(1+Hepcidin/Ki))
kBMSpleen = 0.061902954378781Reaction: FeBM => FeSpleen, Rate Law: kBMSpleen*FeBM*BoneMarrow
vRBCSpleen = 0.0235286Reaction: FeRBC => FeSpleen, Rate Law: vRBCSpleen*FeRBC*RBC
Km = 0.0159421218669513; VDuoNTBI = 0.200241893786814; Ki = 1.0E-9Reaction: FeDuo => NTBI; Hepcidin, Rate Law: VDuoNTBI*Duodenum*FeDuo/((Km+FeDuo)*(1+Hepcidin/Ki))
kInRest = 6.16356235352873Reaction: Fe1Tf => FeRest + Tf, Rate Law: kInRest*Fe1Tf*Plasma
k1=0.75616Reaction: Hepcidin =>, Rate Law: Plasma*k1*Hepcidin
kInDuo = 0.0689984226081531Reaction: Fe1Tf => FeDuo + Tf, Rate Law: kInDuo*Fe1Tf*Plasma
VSpleenNTBI = 1.342204923; Km = 0.0159421218669513; Ki = 1.0E-9Reaction: FeSpleen => NTBI; Hepcidin, Rate Law: VSpleenNTBI*Spleen*FeSpleen/((Km+FeSpleen)*(1+Hepcidin/Ki))
kInRBC = 1.08447580176706Reaction: FeBM => FeRBC, Rate Law: kInRBC*FeBM*BoneMarrow
vDiet = 0.00415624Reaction: => FeDuo, Rate Law: Duodenum*vDiet
v=2.30942E-8Reaction: => Hepcidin, Rate Law: Plasma*v
kRestLoss = 0.023533240736163Reaction: FeRest => FeOutside, Rate Law: RestOfBody*kRestLoss*FeRest

States:

NameDescription
FeRest[iron cation]
Fe2Tf[Serotransferrin; iron(3+)]
NTBI[iron cation]
FeSpleen[iron cation]
FeBM[iron cation]
FeRBC[iron cation]
Fe1Tf[Serotransferrin; iron(3+)]
FeLiver[iron cation]
FeDuo[iron cation]
Tf[Serotransferrin]
Hepcidin[Hepcidin]
FeOutside[iron cation]

Mpekris2017 - Role of vascular normalization in benefit from metronomic chemotherapy: MODEL2001200002v0.0.1

Role of vascular normalization in benefit from metronomic chemotherapy. Mpekris F1, Baish JW2, Stylianopoulos T3, Jain R…

Details

Metronomic dosing of chemotherapy-defined as frequent administration at lower doses-has been shown to be more efficacious than maximum tolerated dose treatment in preclinical studies, and is currently being tested in the clinic. Although multiple mechanisms of benefit from metronomic chemotherapy have been proposed, how these mechanisms are related to one another and which one is dominant for a given tumor-drug combination is not known. To this end, we have developed a mathematical model that incorporates various proposed mechanisms, and report here that improved function of tumor vessels is a key determinant of benefit from metronomic chemotherapy. In our analysis, we used multiple dosage schedules and incorporated interactions among cancer cells, stem-like cancer cells, immune cells, and the tumor vasculature. We found that metronomic chemotherapy induces functional normalization of tumor blood vessels, resulting in improved tumor perfusion. Improved perfusion alleviates hypoxia, which reprograms the immunosuppressive tumor microenvironment toward immunostimulation and improves drug delivery and therapeutic outcomes. Indeed, in our model, improved vessel function enhanced the delivery of oxygen and drugs, increased the number of effector immune cells, and decreased the number of regulatory T cells, which in turn killed a larger number of cancer cells, including cancer stem-like cells. Vessel function was further improved owing to decompression of intratumoral vessels as a result of increased killing of cancer cells, setting up a positive feedback loop. Our model enables evaluation of the relative importance of these mechanisms, and suggests guidelines for the optimal use of metronomic therapy. link: http://identifiers.org/pubmed/28174262

Mueller2008_ThrombinGeneration_minimal: BIOMD0000000367v0.0.1

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

Details

This paper focus on the quest for mechanisms that are able to create tolerance and an activation threshold in the extrinsic coagulation cascade. We propose that the interplay of coagulation inhibitor and blood flow creates threshold behavior. First we test this hypothesis in a minimal, four dimensional model. This model can be analysed by means of time scale analysis. We find indeed that only the interplay of blood flow and inhibition together are able to produce threshold behavior. The mechanism relays on a combination of raw substance supply and wash-out effect by the blood flow and a stabilization of the resting state by the inhibition. We use the insight into this minimal model to interpret the simulation results of a large model. Here, we find that the initiating steps (TF that produces together with fVII(a) factor Xa) does not exhibit threshold behavior, but the overall system does. Hence, the threshold behavior appears via the feedback loop (in that fIIa produces indirectly fXa that in turn produces fIIa again) inhibited by ATIII and blood flow. link: http://identifiers.org/pubmed/17936855

Parameters:

NameDescription
zeta = 0.5; b = 1.5; mu_z = 0.4Reaction: z = ((-b)*y*z+zeta*mu_z)-zeta*z, Rate Law: ((-b)*y*z+zeta*mu_z)-zeta*z
zeta = 0.5; mu_x = 4.0; r = 0.2Reaction: x = ((-r)*x*y+zeta*mu_x)-zeta*x, Rate Law: ((-r)*x*y+zeta*mu_x)-zeta*x
zeta = 0.5; r = 0.2; b = 1.5Reaction: y = (r*x*y-b*y*z)-zeta*y, Rate Law: (r*x*y-b*y*z)-zeta*y

States:

NameDescription
xx
zz
yy

Mueller2015 - Hepatocyte proliferation, T160 phosphorylation of CDK2: BIOMD0000000568v0.0.1

Mueller2015 - Hepatocyte proliferation, T160 phosphorylation of CDK2This model is described in the article: [T160-phosp…

Details

Liver regeneration is a tightly controlled process mainly achieved by proliferation of usually quiescent hepatocytes. The specific molecular mechanisms ensuring cell division only in response to proliferative signals such as hepatocyte growth factor (HGF) are not fully understood. Here, we combined quantitative time-resolved analysis of primary mouse hepatocyte proliferation at the single cell and at the population level with mathematical modeling. We showed that numerous G1/S transition components are activated upon hepatocyte isolation whereas DNA replication only occurs upon additional HGF stimulation. In response to HGF, Cyclin:CDK complex formation was increased, p21 rather than p27 was regulated, and Rb expression was enhanced. Quantification of protein levels at the restriction point showed an excess of CDK2 over CDK4 and limiting amounts of the transcription factor E2F-1. Analysis with our mathematical model revealed that T160 phosphorylation of CDK2 correlated best with growth factor-dependent proliferation, which we validated experimentally on both the population and the single cell level. In conclusion, we identified CDK2 phosphorylation as a gate-keeping mechanism to maintain hepatocyte quiescence in the absence of HGF. link: http://identifiers.org/pubmed/25771250

Parameters:

NameDescription
kp_c2cak = 101.599112819407Reaction: S13 => S18; S13, Rate Law: Nucleus*kp_c2cak*S13/cell
ks_e2fe2f = 0.459601740303536; ks_e2fmyc = 2.49174531457788E-6; tf = 0.635098964160441Reaction: => S14; S14, Rate Law: Nucleus*(ks_e2fe2f*S14+ks_e2fmyc)*tf/cell
kd_p21c4 = 1430.78413614709Reaction: S19 => S10 + S12; S19, Rate Law: cell*kd_p21c4*S19/cell
kb_p21c2 = 997.938141166465Reaction: S4 + S12 => S20; S4, S12, Rate Law: cell*kb_p21c2*S4*S12/cell
ks_c2myc = 0.157511710670132; ks_c2e2f = 2.19944932286058; tf = 0.635098964160441Reaction: => S4; S14, S16, Rate Law: cell*(ks_c2myc*tf+ks_c2e2f*(S14+S16))/cell
kdeg_rbbound = 0.0889964132806627Reaction: S16 => S14; S16, Rate Law: Nucleus*kdeg_rbbound*S16/cell
kb_p21c4 = 14.3083360067931Reaction: S10 + S12 => S19; S10, S12, Rate Law: cell*kb_p21c4*S10*S12/cell
kdeg_p21erkskp2 = 2.82976267377082E-4; erk = 0.16; kdeg_p21skp2 = 0.750574831653576; kdeg_p21c2skp2 = 0.040108041739907Reaction: S23 => S18; S18, S14, S23, Rate Law: Nucleus*(kdeg_p21erkskp2*erk+kdeg_p21c2skp2*S18+kdeg_p21skp2)*S14*S23/cell
kcatdp_rbc4 = 2892.0219338341; nrb = 3.0; Km_dprb = 0.118988383643671; kinh_pp1 = 16634.9400020267Reaction: S15 => S1; S18, S15, Rate Law: Nucleus*kcatdp_rbc4*S15^nrb/(Km_dprb^nrb+S15^nrb)*1/(1+kinh_pp1*S18)/cell
kdp_c2cak = 101.282119534273Reaction: S18 => S13; S18, Rate Law: Nucleus*kdp_c2cak*S18/cell
kdeg_e2ffree = 0.100037217670528Reaction: S14 => ; S14, Rate Law: Nucleus*kdeg_e2ffree*S14/cell
kb_rbe2f = 229.976400323907Reaction: S1 + S14 => S2; S1, S14, Rate Law: Nucleus*kb_rbe2f*S1*S14/cell
kd_rbpe2f = 87735.365961809Reaction: S16 => S14 + S15; S16, Rate Law: Nucleus*kd_rbpe2f*S16/cell
nrb = 3.0; Km_prb = 2.03458881189349; kcatp_rbc2 = 7142308.07232621Reaction: S16 => S14 + S21; S18, S16, Rate Law: Nucleus*kcatp_rbc2*S18*S16^nrb/(Km_prb^nrb+S16^nrb)/cell
kdeg_rbfree = 0.346759895758394Reaction: S1 => ; S1, Rate Law: Nucleus*kdeg_rbfree*S1/cell
gsk3b = 0.47; kdeg_c4 = 1.01433121526038; kdeg_c4gsk3b = 0.107637073030656Reaction: S19 => S12; S19, Rate Law: cell*(kdeg_c4+kdeg_c4gsk3b*gsk3b)*S19/cell
kd_rbe2f = 11499.4014796088Reaction: S2 => S1 + S14; S2, Rate Law: Nucleus*kd_rbe2f*S2/cell
kdeg_e2fbound = 0.0999954023364359Reaction: S2 => S1; S2, Rate Law: Nucleus*kdeg_e2fbound*S2/cell
ks_rb = 72.5245257602228; ks_rbe2f = 20.0129834334888Reaction: => S1; S14, Rate Law: Nucleus*(ks_rb+ks_rbe2f*S14)/cell
scale_TotCDK2T160 = 2.728395741944; Vnuc = 0.25; Vcyto = 12.67Reaction: ObsTotCDK2T160_obs = scale_TotCDK2T160*Vnuc*(S18+S23)/(Vnuc+Vcyto), Rate Law: missing
erk = 0.16; gsk3b = 0.47; kdeg_p21erk = 0.736488746268804; kdeg_p21gsk3b = 0.00464010657330714Reaction: S12 => ; S12, Rate Law: cell*(kdeg_p21gsk3b*gsk3b+kdeg_p21erk*erk)*S12/cell
scale_PhosRbS800 = 0.82377467648995; Vnuc = 0.25; Vcyto = 12.67Reaction: ObsPhosRbS800_obs = scale_PhosRbS800*Vnuc*S21/(Vnuc+Vcyto), Rate Law: missing
ks_c4 = 14298.6715905912; tf = 0.635098964160441Reaction: => S10, Rate Law: cell*ks_c4*tf/cell
scale_TotE2F = 28.7418; Vnuc = 0.25; Vcyto = 12.67; scale_TotRb = 0.2605Reaction: ObsTotE2F_obs = (scale_TotE2F+scale_TotRb)*Vnuc*(S2+S14+S16)/(Vnuc+Vcyto), Rate Law: missing
kd_p21c2 = 9.98179979713068Reaction: S20 => S4 + S12; S20, Rate Law: cell*kd_p21c2*S20/cell
Vratio = 0.0197316495659037; kimport = 0.0744777523096695Reaction: S12 => S11; S12, Rate Law: kimport/Vratio*S12/cell
Vnuc = 0.25; Vcyto = 12.67; scale_TotRb = 0.2605Reaction: ObsTotRb_obs = scale_TotRb*Vnuc*(S1+S2+S15+S16+S21)/(Vnuc+Vcyto), Rate Law: missing
Vnuc = 0.25; scale_Totp21CDK2 = 0.339790715037712; Vcyto = 12.67Reaction: ObsCDK2P21_obs = scale_Totp21CDK2*(Vnuc*(S3+S23)+Vcyto*S20)/(Vnuc+Vcyto), Rate Law: missing
nrb = 3.0; kcatdp_rbc2 = 0.00313841707547858; Km_dprb = 0.118988383643671; kinh_pp1 = 16634.9400020267Reaction: S21 => S15; S18, S21, Rate Law: Nucleus*kcatdp_rbc2*S21^nrb/(Km_dprb^nrb+S21^nrb)*1/(1+kinh_pp1*S18)/cell
ks_p21p53 = 3.84136205729286E-6; tfp21 = 0.635098964160441; ks_p21e2f = 0.811617200647839Reaction: => S12; S14, Rate Law: cell*(ks_p21p53+ks_p21e2f*S14)*tfp21/cell
kcatp_rbc4 = 2797.82326282727; nrb = 3.0; Km_prb = 2.03458881189349Reaction: S1 => S15; S24, S1, Rate Law: Nucleus*kcatp_rbc4*S24*S1^nrb/(Km_prb^nrb+S1^nrb)/cell
kb_rbpe2f = 182.218452288549Reaction: S14 + S15 => S16; S14, S15, Rate Law: Nucleus*kb_rbpe2f*S14*S15/cell
k_dna = 0.00949790539669408Reaction: S5 => S17; S18, S14, S5, Rate Law: Nucleus*k_dna*S18*S14*S5/cell
gsk3b = 0.47; kdeg_c2 = 0.225746618767114; kdeg_c2gsk3b = 1.55090179808215E-5Reaction: S3 => S11; S3, Rate Law: Nucleus*(kdeg_c2+kdeg_c2gsk3b*gsk3b)*S3/cell
k_delay = 23.6658781343201Reaction: S27 => S28; S27, Rate Law: Nucleus*k_delay*S27/cell
Vnuc = 0.25; scale_Totp21 = 0.1728; Vcyto = 12.67Reaction: ObsTotP21_obs = scale_Totp21*(Vnuc*(S3+S11+S23+S24)+Vcyto*(S12+S19+S20))/(Vnuc+Vcyto), Rate Law: missing
kdeg_rbp21 = 0.863570809432207Reaction: S16 => S14; S11, S16, Rate Law: Nucleus*kdeg_rbp21*S11*S16/cell
kdeg_c4 = 1.01433121526038Reaction: S24 => ; S24, Rate Law: Nucleus*kdeg_c4*S24/cell

States:

NameDescription
S11[Cyclin-dependent kinase inhibitor 1B]
S14[Transcription factor E2F1]
S16[Transcription factor E2F1; Retinoblastoma-associated protein; phosphorylated]
ObsDNAContent obs[deoxyribonucleic acid]
inhp53[Cellular tumor antigen p53]
S13[phosphorylated; cyclin E1-CDK2 complex]
S23[cyclin E1-CDK2 complex; Cyclin-dependent kinase inhibitor 1B; phosphorylated]
inhakt[RAC-alpha serine/threonine-protein kinase]
inhc4d1[cyclin D1-CDK4 complex]
S17[pre-replicative complex]
S28[pre-replicative complex]
S4[cyclin E1-CDK2 complex; phosphorylated]
S1[Retinoblastoma-associated protein; phosphorylated]
S25[pre-replicative complex]
ObsTotE2F obs[Transcription factor E2F1]
inherk[Mitogen-activated protein kinase 3]
S19[cyclin D1-CDK4 complex; Cyclin-dependent kinase inhibitor 1B]
ObsPhosRbS800 obs[Retinoblastoma-associated protein; phosphorylated]
S5[pre-replicative complex]
S10[cyclin D1-CDK4 complex]
S3[cyclin E1-CDK2 complex; Cyclin-dependent kinase inhibitor 1B; phosphorylated]
S27[pre-replicative complex]
ObsTotCDK2T160 obs[Cyclin-dependent kinase 2; phosphorylated]
S2[Transcription factor E2F1; Retinoblastoma-associated protein; phosphorylated]
S26[cyclin D1-CDK4 complex]
S21[Retinoblastoma-associated protein; phosphorylated]
ObsTotP21 obs[Cyclin-dependent kinase inhibitor 1B]
ObsCDK2P21 obs[Cyclin-dependent kinase inhibitor 1B; Cyclin-dependent kinase 2]
S20[cyclin E1-CDK2 complex; Cyclin-dependent kinase inhibitor 1B; phosphorylated]
hgf[Hepatocyte growth factor]
ObsTotRb obs[Retinoblastoma-associated protein]
S15[Retinoblastoma-associated protein; phosphorylated]
S12[Cyclin-dependent kinase inhibitor 1B]
S24[cyclin D1-CDK4 complex; Cyclin-dependent kinase inhibitor 1B]
S22[pre-replicative complex]

Muenzner2019 - Yeast Cell cycle control network: MODEL2007020001v0.0.1

A mechanistically detailed model of the cell cycle control network of Saccharomyces cerevisiae.

Details

Understanding how cellular functions emerge from the underlying molecular mechanisms is a key challenge in biology. This will require computational models, whose predictive power is expected to increase with coverage and precision of formulation. Genome-scale models revolutionised the metabolic field and made the first whole-cell model possible. However, the lack of genome-scale models of signalling networks blocks the development of eukaryotic whole-cell models. Here, we present a comprehensive mechanistic model of the molecular network that controls the cell division cycle in Saccharomyces cerevisiae. We use rxncon, the reaction-contingency language, to neutralise the scalability issues preventing formulation, visualisation and simulation of signalling networks at the genome-scale. We use parameter-free modelling to validate the network and to predict genotype-to-phenotype relationships down to residue resolution. This mechanistic genome-scale model offers a new perspective on eukaryotic cell cycle control, and opens up for similar models—and eventually whole-cell models—of human cells. link: https://doi.org/10.1038/s41467-019-08903-w

Mufudza2012 - Estrogen effect on the dynamics of breast cancer: BIOMD0000000642v0.0.1

Mufudza2012 - Estrogen effect on the dynamics of breast cancerThis deterministic model shows the dynamics of breast canc…

Details

Worldwide, breast cancer has become the second most common cancer in women. The disease has currently been named the most deadly cancer in women but little is known on what causes the disease. We present the effects of estrogen as a risk factor on the dynamics of breast cancer. We develop a deterministic mathematical model showing general dynamics of breast cancer with immune response. This is a four-population model that includes tumor cells, host cells, immune cells, and estrogen. The effects of estrogen are then incorporated in the model. The results show that the presence of extra estrogen increases the risk of developing breast cancer. link: http://identifiers.org/pubmed/23365616

Parameters:

NameDescription
sigma3 = 0.3; s = 0.4; mu = 0.29; rho = 0.2; gamma3 = 0.085; omega = 0.3; v = 0.4Reaction: I = (((s+rho*I*T/(omega+T))-gamma3*I*T)-mu*I)-sigma3*I*E/(v+E), Rate Law: (((s+rho*I*T/(omega+T))-gamma3*I*T)-mu*I)-sigma3*I*E/(v+E)
beta1 = 0.3; alpha1 = 0.7; sigma1 = 1.2; delta1 = 1.0Reaction: H = H*((alpha1-beta1*H)-delta1*T)-sigma1*H*E, Rate Law: H*((alpha1-beta1*H)-delta1*T)-sigma1*H*E
beta2 = 0.4; alpha3 = 1.0; gamma2 = 0.9; sigma2 = 0.94Reaction: T = (T*(alpha3-beta2*T)-gamma2*I*T)+sigma2*H*E, Rate Law: (T*(alpha3-beta2*T)-gamma2*I*T)+sigma2*H*E

States:

NameDescription
I[immune response; cell]
T[neoplastic cell]
H[cell]

Mukandavire2009 - Model for HIV-Malaria co-infection: MODEL1805230001v0.0.1

Mathematical model for HIV, malaria and HIV-malaria co-infection.

Details

A deterministic model for the co-interaction of HIV and malaria in a community is presented and rigorously analyzed. Two sub-models, namely the HIV-only and malaria-only sub-models, are considered first of all. Unlike the HIV-only sub-model, which has a globally-asymptotically stable disease-free equilibrium whenever the associated reproduction number is less than unity, the malaria-only sub-model undergoes the phenomenon of backward bifurcation, where a stable disease-free equilibrium co-exists with a stable endemic equilibrium, for a certain range of the associated reproduction number less than unity. Thus, for malaria, the classical requirement of having the associated reproduction number to be less than unity, although necessary, is not sufficient for its elimination. It is also shown, using centre manifold theory, that the full HIV-malaria co-infection model undergoes backward bifurcation. Simulations of the full HIV-malaria model show that the two diseases co-exist whenever their reproduction numbers exceed unity (with no competitive exclusion occurring). Further, the reduction in sexual activity of individuals with malaria symptoms decreases the number of new cases of HIV and the mixed HIV-malaria infection while increasing the number of malaria cases. Finally, these simulations show that the HIV-induced increase in susceptibility to malaria infection has marginal effect on the new cases of HIV and malaria but increases the number of new cases of the dual HIV-malaria infection. link: http://identifiers.org/pubmed/19364156

Mukandavire2020 - SEIR model of early COVID-19 transmission in South Africa: BIOMD0000000978v0.0.1

The emergence and fast global spread of COVID-19 has presented one of the greatest public health challenges in modern ti…

Details

The emergence and fast global spread of COVID-19 has presented one of the greatest public health challenges in modern times with no proven cure or vaccine. Africa is still early in this epidemic, therefore the extent of disease severity is not yet clear. We used a mathematical model to fit to the observed cases of COVID-19 in South Africa to estimate the basic reproductive number and critical vaccination coverage to control the disease for different hypothetical vaccine efficacy scenarios. We also estimated the percentage reduction in effective contacts due to the social distancing measures implemented. Early model estimates show that COVID-19 outbreak in South Africa had a basic reproductive number of 2.95 (95% credible interval [CrI] 2.83-3.33). A vaccine with 70% efficacy had the capacity to contain COVID-19 outbreak but at very higher vaccination coverage 94.44% (95% Crl 92.44-99.92%) with a vaccine of 100% efficacy requiring 66.10% (95% Crl 64.72-69.95%) coverage. Social distancing measures put in place have so far reduced the number of social contacts by 80.31% (95% Crl 79.76-80.85%). These findings suggest that a highly efficacious vaccine would have been required to contain COVID-19 in South Africa. Therefore, the current social distancing measures to reduce contacts will remain key in controlling the infection in the absence of vaccines and other therapeutics. link: http://identifiers.org/pubmed/32706790

Mukhopadhyay2013 - T cell receptor proximal signaling reveals emergent ultrasensitivity: MODEL1604100000v0.0.1

Mukhopadhyay2013 - T cell receptor proximal signaling reveals emergent ultrasensitivityThis model is described in the ar…

Details

Receptor phosphorylation is thought to be tightly regulated because phosphorylated receptors initiate signaling cascades leading to cellular activation. The T cell antigen receptor (TCR) on the surface of T cells is phosphorylated by the kinase Lck and dephosphorylated by the phosphatase CD45 on multiple immunoreceptor tyrosine-based activation motifs (ITAMs). Intriguingly, Lck sequentially phosphorylates ITAMs and ZAP-70, a cytosolic kinase, binds to phosphorylated ITAMs with differential affinities. The purpose of multiple ITAMs, their sequential phosphorylation, and the differential ZAP-70 affinities are unknown. Here, we use a systems model to show that this signaling architecture produces emergent ultrasensitivity resulting in switch-like responses at the scale of individual TCRs. Importantly, this switch-like response is an emergent property, so that removal of multiple ITAMs, sequential phosphorylation, or differential affinities abolishes the switch. We propose that highly regulated TCR phosphorylation is achieved by an emergent switch-like response and use the systems model to design novel chimeric antigen receptors for therapy. link: http://identifiers.org/pubmed/23555234

Mukhta2018-the effect of bednet coverage on malaria transmission in South Sudan: MODEL2003160003v0.0.1

A campaign for malaria control, using Long Lasting Insecticide Nets (LLINs) was launched in South Sudan in 2009. The suc…

Details

A campaign for malaria control, using Long Lasting Insecticide Nets (LLINs) was launched in South Sudan in 2009. The success of such a campaign often depends upon adequate available resources and reliable surveillance data which help officials understand existing infections. An optimal allocation of resources for malaria control at a sub-national scale is therefore paramount to the success of efforts to reduce malaria prevalence. In this paper, we extend an existing SIR mathematical model to capture the effect of LLINs on malaria transmission. Available data on malaria is utilized to determine realistic parameter values of this model using a Bayesian approach via Markov Chain Monte Carlo (MCMC) methods. Then, we explore the parasite prevalence on a continued rollout of LLINs in three different settings in order to create a sub-national projection of malaria. Further, we calculate the model's basic reproductive number and study its sensitivity to LLINs' coverage and its efficacy. From the numerical simulation results, we notice a basic reproduction number, [Formula: see text], confirming a substantial increase of incidence cases if no form of intervention takes place in the community. This work indicates that an effective use of LLINs may reduce [Formula: see text] and hence malaria transmission. We hope that this study will provide a basis for recommending a scaling-up of the entry point of LLINs' distribution that targets households in areas at risk of malaria. link: http://identifiers.org/pubmed/29879166

Muller2008 - Simplified MAPK activation Dynamics (Model B): BIOMD0000000664v0.0.1

Muller2008 - Simplified MAPK activation Dynamics (Model B)Simplified mathematical model (model B) for predicting MAPK si…

Details

Activation of the fibroblast growth factor (FGFR) and melanocyte stimulating hormone (MC1R) receptors stimulates B-Raf and C-Raf isoforms that regulate the dynamics of MAPK1,2 signaling. Network topology motifs in mammalian cells include feed-forward and feedback loops and bifans where signals from two upstream molecules integrate to modulate the activity of two downstream molecules. We computationally modeled and experimentally tested signal processing in the FGFR/MC1R/B-Raf/C-Raf/MAPK1,2 network in human melanoma cells; identifying 7 regulatory loops and a bifan motif. Signaling from FGFR leads to sustained activation of MAPK1,2, whereas signaling from MC1R results in transient activation of MAPK1,2. The dynamics of MAPK activation depends critically on the expression level and connectivity to C-Raf, which is critical for a sustained MAPK1,2 response. A partially incoherent bifan motif with a feedback loop acts as a logic gate to integrate signals and regulate duration of activation of the MAPK signaling cascade. Further reducing a 106-node ordinary differential equations network encompassing the complete network to a 6-node network encompassing rate-limiting processes sustains the feedback loops and the bifan, providing sufficient information to predict biological responses. link: http://identifiers.org/pubmed/18171696

Parameters:

NameDescription
E = 10.0; f13 = 0.6 0.06*ml/(mol*s)Reaction: => C_Raf; C_Raf_inactive, FGFR, Rate Law: Compartment*f13*((E-C_Raf)-C_Raf_inactive)*FGFR
E = 10.0; f53 = 1.5 0.06*ml/(mol*s)Reaction: => C_Raf; C_Raf_inactive, MAPK, Rate Law: Compartment*f53*((E-C_Raf)-C_Raf_inactive)*MAPK
f14 = 0.1 1/(59.9999*s)Reaction: => B_Raf; FGFR, Rate Law: Compartment*f14*FGFR
g1 = 0.0Reaction: g1_0 = g1, Rate Law: missing
f24 = 0.8 1/(59.9999*s)Reaction: => B_Raf; MSH, Rate Law: Compartment*f24*MSH
b2 = 10.0; a2 = 10.0 0.06*mmol/(l*s); g2 = 1.0Reaction: => MSH; g2_0, Rate Law: Compartment*a2*g2/(b2+g2)
f45 = 0.1 1/(59.9999*s)Reaction: => MAPK; B_Raf, Rate Law: Compartment*f45*B_Raf
d3 = 1.0 1/(59.9999*s)Reaction: C_Raf =>, Rate Law: Compartment*d3*C_Raf
d6 = 0.001 1/(59.9999*s)Reaction: C_Raf_inactive =>, Rate Law: Compartment*d6*C_Raf_inactive
f35 = 0.3 1/(59.9999*s)Reaction: => MAPK; C_Raf, Rate Law: Compartment*f35*C_Raf
d5 = 1.0 1/(59.9999*s)Reaction: MAPK =>, Rate Law: Compartment*d5*MAPK
h36_y3 = 0.1 0.06*ml/(mol*s)Reaction: C_Raf => C_Raf_inactive; MSH, Rate Law: Compartment*h36_y3*MSH*C_Raf
d1 = 0.2 1/(59.9999*s)Reaction: FGFR =>, Rate Law: Compartment*d1*FGFR
b1 = 10.0; a1 = 10.0 0.06*mmol/(l*s); g1 = 0.0Reaction: => FGFR; g1_0, Rate Law: Compartment*a1*g1/(b1+g1)
d2 = 0.1 1/(59.9999*s)Reaction: MSH =>, Rate Law: Compartment*d2*MSH
g2 = 1.0Reaction: g2_0 = g2, Rate Law: missing
d4 = 1.1 1/(59.9999*s)Reaction: B_Raf =>, Rate Law: Compartment*d4*B_Raf

States:

NameDescription
FGFR[Fibroblast growth factor receptor 1]
C Raf[RAF proto-oncogene serine/threonine-protein kinase]
C Raf inactive[RAF proto-oncogene serine/threonine-protein kinase]
B Raf[Serine/threonine-protein kinase B-raf]
MAPK[Mitogen-activated protein kinase 1]
g2 0[Melanocyte-stimulating hormone; Stimulus]
MSH[melanocyte-stimulating hormone receptor]
g1 0[Fibroblast growth factor 1; Stimulus]

Mulquiney1999_BPG_metabolism: MODEL5950552398v0.0.1

This model is described and analysed in a series of three articles: **Model of 2,3-bisphosphoglycerate metabolism in t…

Details

This is the third of three papers [see also Mulquiney, Bubb and Kuchel (1999) Biochem. J. 342, 565-578; Mulquiney and Kuchel (1999) Biochem. J. 342, 579-594] for which the general goal was to explain the regulation and control of 2,3-bisphosphoglycerate (2,3-BPG) metabolism in human erythrocytes. 2,3-BPG is a major modulator of haemoglobin oxygen affinity and hence is vital in blood oxygen transport. A detailed mathematical model of erythrocyte metabolism was presented in the first two papers. The model was refined through an iterative loop of experiment and simulation and it was used to predict outcomes that are consistent with the metabolic behaviour of the erythrocyte under a wide variety of experimental and physiological conditions. For the present paper, the model was examined using computer simulation and Metabolic Control Analysis. The analysis yielded several new insights into the regulation and control of 2,3-BPG metabolism. Specifically it was found that: (1) the feedback inhibition of hexokinase and phosphofructokinase by 2, 3-BPG are equally as important as the product inhibition of 2,3-BPG synthase in controlling the normal in vivo steady-state concentration of 2,3-BPG; (2) H(+) and oxygen are effective regulators of 2,3-BPG concentration and that increases in 2,3-BPG concentrations are achieved with only small changes in glycolytic rate; (3) these two effectors exert most of their influence through hexokinase and phosphofructokinase; (4) flux through the 2,3-BPG shunt changes in absolute terms in response to different energy demands placed on the cell. This response of the 2,3-BPG shunt contributes an [ATP]-stabilizing effect. A 'cost' of this is that 2, 3-BPG concentrations are very sensitive to the energy demand of the cell and; (5) the flux through the 2,3-BPG shunt does not change in response to different non-glycolytic demands for NADH. link: http://identifiers.org/pubmed/10477270

Munz2009 - Zombi Impulsive Killing: MODEL1008060000v0.0.1

Munz2009 - Zombie Impulsive KillingThis is the basic SZR model with impulsive killing described in the article. This mo…

Details

Zombies are a popular figure in pop culture/entertainment and they are usually portrayed as being brought about through an outbreak or epidemic. Consequently, we model a zombie attack, using biological assumptions based on popular zombie movies. We introduce a basic model for zombie infection, determine equilibria and their stability, and illustrate the outcome with numerical solutions. We then refine the model to introduce a latent period of zombification, whereby humans are infected, but not infectious, before becoming undead. We then modify the model to include the effects of possible quarantine or a cure. Finally, we examine the impact of regular, impulsive reductions in the number of zombies and derive conditions under which eradication can occur. We show that only quick, aggressive attacks can stave off the doomsday scenario: the collapse of society as zombies overtake us all. link: http://www.mathworks.co.uk/matlabcentral/linkexchange/links/1749-when-zombies-attack-mathematical-modelling-of-an-outbreak-of-zombie-infection

Munz2009 - Zombie basic SZR: MODEL1009230000v0.0.1

Munz2009 - Zombie basic SZRThis is the basic SZR model for zombie infection. It is based on a classic mathematical mode…

Details

Zombies are a popular figure in pop culture/entertainment and they are usually portrayed as being brought about through an outbreak or epidemic. Consequently, we model a zombie attack, using biological assumptions based on popular zombie movies. We introduce a basic model for zombie infection, determine equilibria and their stability, and illustrate the outcome with numerical solutions. We then refine the model to introduce a latent period of zombification, whereby humans are infected, but not infectious, before becoming undead. We then modify the model to include the effects of possible quarantine or a cure. Finally, we examine the impact of regular, impulsive reductions in the number of zombies and derive conditions under which eradication can occur. We show that only quick, aggressive attacks can stave off the doomsday scenario: the collapse of society as zombies overtake us all. link: http://www.mathworks.co.uk/matlabcentral/linkexchange/links/1749-when-zombies-attack-mathematical-modelling-of-an-outbreak-of-zombie-infection

Munz2009 - Zombie SIZRC: BIOMD0000000882v0.0.1

Munz2009 - Zombie SIZRC This is the model with an latent infection and cure for zombies described in the article. This…

Details

Zombies are a popular figure in pop culture/entertainment and they are usually portrayed as being brought about through an outbreak or epidemic. Consequently, we model a zombie attack, using biological assumptions based on popular zombie movies. We introduce a basic model for zombie infection, determine equilibria and their stability, and illustrate the outcome with numerical solutions. We then refine the model to introduce a latent period of zombification, whereby humans are infected, but not infectious, before becoming undead. We then modify the model to include the effects of possible quarantine or a cure. Finally, we examine the impact of regular, impulsive reductions in the number of zombies and derive conditions under which eradication can occur. We show that only quick, aggressive attacks can stave off the doomsday scenario: the collapse of society as zombies overtake us all. link: http://www.mathworks.co.uk/matlabcentral/linkexchange/links/1749-when-zombies-attack-mathematical-modelling-of-an-outbreak-of-zombie-infection

Parameters:

NameDescription
delta = 1.0E-4; alpha = 0.005Reaction: => Removal; Susceptible, Zombie, Rate Law: compartment*(alpha*Susceptible*Zombie+delta*Susceptible)
zeta = 1.0E-4; beta = 0.0095Reaction: => Zombie; Susceptible, Removal, Rate Law: compartment*(beta*Susceptible*Zombie+zeta*Removal)
p = 0.05Reaction: => Susceptible, Rate Law: compartment*p
delta = 1.0E-4; beta = 0.0095Reaction: Susceptible => ; Zombie, Rate Law: compartment*(beta*Susceptible*Zombie+delta*Susceptible)
alpha = 0.005Reaction: Zombie => ; Susceptible, Rate Law: compartment*alpha*Susceptible*Zombie
zeta = 1.0E-4Reaction: Removal => ; Susceptible, Zombie, Rate Law: compartment*zeta*Removal

States:

NameDescription
Removal[C64914]
ZombieZombie
Susceptible[Susceptibility]

Munz2009 - Zombie SIZRQ: MODEL1008060002v0.0.1

Munz2009 - Zombie SIZRQThis is the model with latent infection and quarantine described in the article. This model was…

Details

Zombies are a popular figure in pop culture/entertainment and they are usually portrayed as being brought about through an outbreak or epidemic. Consequently, we model a zombie attack, using biological assumptions based on popular zombie movies. We introduce a basic model for zombie infection, determine equilibria and their stability, and illustrate the outcome with numerical solutions. We then refine the model to introduce a latent period of zombification, whereby humans are infected, but not infectious, before becoming undead. We then modify the model to include the effects of possible quarantine or a cure. Finally, we examine the impact of regular, impulsive reductions in the number of zombies and derive conditions under which eradication can occur. We show that only quick, aggressive attacks can stave off the doomsday scenario: the collapse of society as zombies overtake us all. link: http://www.mathworks.co.uk/matlabcentral/linkexchange/links/1749-when-zombies-attack-mathematical-modelling-of-an-outbreak-of-zombie-infection

Muraro2011_Cytokinin-Auxin_CrossRegulation: BIOMD0000000416v0.0.1

This model is from the article: The influence of cytokinin-auxin cross-regulation on cell-fate determination in Arab…

Details

Root growth and development in Arabidopsis thaliana are sustained by a specialised zone termed the meristem, which contains a population of dividing and differentiating cells that are functionally analogous to a stem cell niche in animals. The hormones auxin and cytokinin control meristem size antagonistically. Local accumulation of auxin promotes cell division and the initiation of a lateral root primordium. By contrast, high cytokinin concentrations disrupt the regular pattern of divisions that characterises lateral root development, and promote differentiation. The way in which the hormones interact is controlled by a genetic regulatory network. In this paper, we propose a deterministic mathematical model to describe this network and present model simulations that reproduce the experimentally observed effects of cytokinin on the expression of auxin regulated genes. We show how auxin response genes and auxin efflux transporters may be affected by the presence of cytokinin. We also analyse and compare the responses of the hormones auxin and cytokinin to changes in their supply with the responses obtained by genetic mutations of SHY2, which encodes a protein that plays a key role in balancing cytokinin and auxin regulation of meristem size. We show that although shy2 mutations can qualitatively reproduce the effect of varying auxin and cytokinin supply on their response genes, some elements of the network respond differently to changes in hormonal supply and to genetic mutations, implying a different, general response of the network. We conclude that an analysis based on the ratio between these two hormones may be misleading and that a mathematical model can serve as a useful tool for stimulate further experimental work by predicting the response of the network to changes in hormone levels and to other genetic mutations. link: http://identifiers.org/pubmed/21640126

Parameters:

NameDescription
psiARF = 0.1; psiARFIAA = 0.1; thetaARF = 0.1; thetaARF2 = 0.01; thARFIAA = 0.1; thARRBph = 0.1Reaction: F1 = ARF/thetaARF/(1+ARF/thetaARF+ARF2/thetaARF2+ARFIAA/thARFIAA+ARF*IAAp/psiARFIAA+ARF^2/psiARF+ARRBph/thARRBph), Rate Law: missing
muAux = 0.1; ka = 100.0; eps = 0.01; alphaAux = 1.0; kd = 1.0; etaAuxTIR1 = 10.0Reaction: => Aux; TIR1, AuxTIR1, Rate Law: muAux*(alphaAux-Aux)-1/eps*etaAuxTIR1*(ka*Aux*TIR1-kd*AuxTIR1)
thARRAph = 0.1; thARRBph = 0.1Reaction: F4 = ARRBph/thARRBph/(1+ARRAph/thARRAph+ARRBph/thARRBph), Rate Law: missing
qa = 1.0; qd = 1.0Reaction: => ARF2; ARF, Rate Law: qa*ARF^2-qd*ARF2
eps = 0.01; deltaPINp = 1.0Reaction: => PINp; PINm, Rate Law: 1/eps*(deltaPINp*PINm-PINp)
alphaAHK = 1.0; etaAHKph = 1.0Reaction: CkAHK = alphaAHK-etaAHKph*(AHKph+CkAHKph), Rate Law: missing
lambda1 = 0.1; phiARp = 2.0Reaction: => ARm; F5a, F5b, Rate Law: phiARp*(lambda1*F5a+F5b)-ARm
ud = 1.0; eps = 0.01; ua = 1.0Reaction: => ARRBph; CkAHKph, CkAHK, ARRBp, Rate Law: 1/eps*(ua*CkAHKph*ARRBp-ud*CkAHK*ARRBph)
phiCRp = 2.0Reaction: => CRm; F4, Rate Law: phiCRp*F4-CRm
alphaPH = 1.0Reaction: CkAHKph = ((alphaPH-AHKph)-ARRAph)-ARRBph, Rate Law: missing
deltaARRAp = 1.0; eps = 0.01; sa = 1.0; etaAHKph = 1.0; sd = 1.0Reaction: => ARRAp; ARRAm, CkAHK, ARRAph, CkAHKph, Rate Law: 1/eps*((deltaARRAp*ARRAm-ARRAp)+etaAHKph*(sd*CkAHK*ARRAph-sa*CkAHKph*ARRAp))
alphaTIR1 = 1.0Reaction: TIR1 = (alphaTIR1-AuxTIR1)-AuxTIAA, Rate Law: missing
ka = 100.0; eps = 0.01; la = 0.5; kd = 1.0; ld = 0.1Reaction: => AuxTIR1; Aux, TIR1, AuxTIAA, IAAp, Rate Law: 1/eps*(((ka*Aux*TIR1-kd*AuxTIR1)+(ld+1)*AuxTIAA)-la*AuxTIR1*IAAp)
lambda1 = 0.1; phiIAAp = 100.0; lambda3 = 0.02Reaction: => IAAm; F1, F2, F3, Rate Law: phiIAAp*(lambda1*F1+F2+lambda3*F3)-IAAm
eps = 0.01; sa = 1.0; sd = 1.0Reaction: => ARRAph; CkAHKph, ARRAp, CkAHK, ARRAph, Rate Law: 1/eps*(sa*CkAHKph*ARRAp-sd*CkAHK*ARRAph)
eps = 0.01; la = 0.5; ld = 0.1Reaction: => AuxTIAA; AuxTIAA, IAAp, AuxTIR1, Rate Law: 1/eps*(la*IAAp*AuxTIR1-(ld+1)*AuxTIAA)
phiARRAp = 100.0Reaction: => ARRAm; F6, Rate Law: phiARRAp*F6-ARRAm
eps = 0.01; etaCkPh = 1.0; ra = 1.0; rd = 1.0; muCk = 0.1; alphaCk = 1.0Reaction: => Ck; AHKph, CkAHKph, Rate Law: muCk*(alphaCk-Ck)-etaCkPh/eps*(ra*AHKph*Ck-rd*CkAHKph)
eps = 0.01; ra = 1.0; rd = 1.0Reaction: => AHKph; CkAHKph, Ck, Rate Law: 1/eps*(rd*CkAHKph-ra*AHKph*Ck)
alphaARF = 1.0Reaction: ARF = (alphaARF-2*ARF2)-ARFIAA, Rate Law: missing
psiARF = 0.1; psiARFIAA = 0.1; thetaARF = 0.1; thetaARF2 = 0.01; thARFIAA = 0.1Reaction: F5a = ARF/thetaARF/(1+ARF/thetaARF+ARF2/thetaARF2+ARFIAA/thARFIAA+ARF*IAAp/psiARFIAA+ARF^2/psiARF), Rate Law: missing
thetaARp = 0.1Reaction: F6 = ARp/thetaARp/(1+ARp/thetaARp), Rate Law: missing
eps = 0.01; etaARFIAA = 1.0; la = 0.5; pa = 10.0; ld = 0.1; deltaIAAp = 1.0; pd = 10.0Reaction: => IAAp; IAAm, AuxTIR1, AuxTIAA, ARFIAA, ARF, Rate Law: 1/eps*((deltaIAAp*IAAm-la*IAAp*AuxTIR1)+ld*AuxTIAA)+etaARFIAA*(pd*ARFIAA-pa*IAAp*ARF)
eps = 0.01; deltaCRp = 1.0Reaction: => CRp; CRm, Rate Law: 1/eps*(deltaCRp*CRm-CRp)
eps = 0.01; muIAAs = 1.0Reaction: => IAAs; AuxTIAA, Rate Law: 1/eps*(AuxTIAA-muIAAs*IAAs)
pa = 10.0; pd = 10.0Reaction: => ARFIAA; ARF, IAAp, Rate Law: pa*ARF*IAAp-pd*ARFIAA
eps = 0.01; deltaARp = 1.0Reaction: => ARp; ARm, Rate Law: 1/eps*(deltaARp*ARm-ARp)
lambda1 = 0.1; phiPINp = 100.0Reaction: => PINm; F5a, F5b, Rate Law: phiPINp*(lambda1*F5a+F5b)-PINm
alphaARRB = 2.0; etaAHKph = 1.0Reaction: ARRBp = alphaARRB-etaAHKph*ARRBph, Rate Law: missing

States:

NameDescription
AHKph[Histidine kinase 4; Phosphoprotein]
F3F3
F5bF5b
CkAHK[cytokinin; Histidine kinase 4]
IAAs[Auxin-responsive protein IAA1]
ARF2[Auxin response factor 2]
AuxTIAA[auxin; Protein TRANSPORT INHIBITOR RESPONSE 1; Auxin-responsive protein IAA1]
ARRBph[Two-component response regulator ARR1; Phosphoprotein]
ARFIAA[Auxin response factor 2; Auxin-responsive protein IAA1]
F6F6
AuxTIR1[auxin; Protein TRANSPORT INHIBITOR RESPONSE 1]
F1F1
ARRAph[Two-component response regulator ARR2; Phosphoprotein]
ARRAp[Two-component response regulator ARR1]
ARm[Protein AUXIN RESPONSE 4]
PINm[Peptidyl-prolyl cis-trans isomerase Pin1]
ARRAm[Two-component response regulator ARR1]
Aux[auxin]
CRm[Ethylene-responsive transcription factor CRF1]
CkAHKph[cytokinin; Histidine kinase 4]
PINp[Peptidyl-prolyl cis-trans isomerase Pin1]
F4F4
ARF[Auxin response factor 2]
TIR1[Protein TRANSPORT INHIBITOR RESPONSE 1]
ARp[Protein AUXIN RESPONSE 4]
CRp[Ethylene-responsive transcription factor CRF1]
IAAm[Auxin-responsive protein IAA1; messenger RNA]
ARRBp[Two-component response regulator ARR1]
F2F2
F5aF5a
IAAp[Auxin-responsive protein IAA1]
Ck[cytokinin]

Muraro2014 - Vascular patterning in Arabidopsis roots: BIOMD0000000522v0.0.1

Muraro2014 - Vascular patterning in Arabidopsis rootsUsing a multicellular model, maintanence of vascular patterning in…

Details

As multicellular organisms grow, positional information is continually needed to regulate the pattern in which cells are arranged. In the Arabidopsis root, most cell types are organized in a radially symmetric pattern; however, a symmetry-breaking event generates bisymmetric auxin and cytokinin signaling domains in the stele. Bidirectional cross-talk between the stele and the surrounding tissues involving a mobile transcription factor, SHORT ROOT (SHR), and mobile microRNA species also determines vascular pattern, but it is currently unclear how these signals integrate. We use a multicellular model to determine a minimal set of components necessary for maintaining a stable vascular pattern. Simulations perturbing the signaling network show that, in addition to the mutually inhibitory interaction between auxin and cytokinin, signaling through SHR, microRNA165/6, and PHABULOSA is required to maintain a stable bisymmetric pattern. We have verified this prediction by observing loss of bisymmetry in shr mutants. The model reveals the importance of several features of the network, namely the mutual degradation of microRNA165/6 and PHABULOSA and the existence of an additional negative regulator of cytokinin signaling. These components form a plausible mechanism capable of patterning vascular tissues in the absence of positional inputs provided by the transport of hormones from the shoot. link: http://identifiers.org/pubmed/24381155

Parameters:

NameDescription
lambda_IAA2 = 10.0; mu_m_IAA2 = 10.0; F_IAA2 = 0.0Reaction: IAA2m = lambda_IAA2*F_IAA2-mu_m_IAA2*IAA2m, Rate Law: lambda_IAA2*F_IAA2-mu_m_IAA2*IAA2m
delta_AHP6 = 1.0; mu_p_AHP6 = 1.0Reaction: AHP6p = delta_AHP6*AHP6m-mu_p_AHP6*AHP6p, Rate Law: delta_AHP6*AHP6m-mu_p_AHP6*AHP6p
mu_p_PHB = 1.0; delta_PHB = 1.0Reaction: PHBp = delta_PHB*PHBm-mu_p_PHB*PHBp, Rate Law: delta_PHB*PHBm-mu_p_PHB*PHBp
F_CK = 0.0; p_ck = 2.0; d_ck = 10.0; phloem_rate_ck = 1.0Reaction: Cytokinin = phloem_rate_ck*p_ck*F_CK-d_ck*Cytokinin, Rate Law: phloem_rate_ck*p_ck*F_CK-d_ck*Cytokinin
lambda_ARR5 = 20.0; mu_m_ARR5 = 10.0; F_ARR5 = 0.0Reaction: ARR5m = lambda_ARR5*F_ARR5-mu_m_ARR5*ARR5m, Rate Law: lambda_ARR5*F_ARR5-mu_m_ARR5*ARR5m
mu_m_PIN1 = 0.0; lambda_PIN1 = 0.0; F_PIN1 = 0.0Reaction: PIN1m = lambda_PIN1*F_PIN1-mu_m_PIN1*PIN1m, Rate Law: lambda_PIN1*F_PIN1-mu_m_PIN1*PIN1m
mu_m_AHP6 = 1.0; lambda_AHP6 = 2.0; F_AHP6 = 0.0Reaction: AHP6m = lambda_AHP6*F_AHP6-mu_m_AHP6*AHP6m, Rate Law: lambda_AHP6*F_AHP6-mu_m_AHP6*AHP6m
phloem_rate_ax = 1.0; p_ax = 0.06; d_ax = 1.0Reaction: Auxin = phloem_rate_ax*p_ax-d_ax*Auxin, Rate Law: phloem_rate_ax*p_ax-d_ax*Auxin
lambda_PIN3 = 0.0; F_PIN3 = 0.0; mu_m_PIN3 = 0.0Reaction: PIN3m = lambda_PIN3*F_PIN3-mu_m_PIN3*PIN3m, Rate Law: lambda_PIN3*F_PIN3-mu_m_PIN3*PIN3m
delta_CKX3 = 1.0; mu_p_CKX3 = 1.0Reaction: CKX3p = delta_CKX3*CKX3m-mu_p_CKX3*CKX3p, Rate Law: delta_CKX3*CKX3m-mu_p_CKX3*CKX3p
mu_p_ARR5 = 10.0; delta_ARR5 = 10.0Reaction: ARR5p = delta_ARR5*ARR5m-mu_p_ARR5*ARR5p, Rate Law: delta_ARR5*ARR5m-mu_p_ARR5*ARR5p
d_phb = 1.0; d_mirna_mrna = 10.0; p_phb = 2.0Reaction: PHBm = (p_phb-d_phb*PHBm)-d_mirna_mrna*PHBm*miRNA, Rate Law: (p_phb-d_phb*PHBm)-d_mirna_mrna*PHBm*miRNA
mu_p_IAA2 = 10.0; delta_IAA2 = 10.0Reaction: IAA2p = delta_IAA2*IAA2m-mu_p_IAA2*IAA2p, Rate Law: delta_IAA2*IAA2m-mu_p_IAA2*IAA2p
mu_m_PIN7 = 1.0; lambda_PIN7 = 1.0; F_PIN7 = 0.0Reaction: PIN7m = lambda_PIN7*F_PIN7-mu_m_PIN7*PIN7m, Rate Law: lambda_PIN7*F_PIN7-mu_m_PIN7*PIN7m

States:

NameDescription
AHP6m[Pseudo histidine-containing phosphotransfer protein 6]
Cytokinin[cytokinin]
IAA2p[Auxin-responsive protein IAA2]
PIN3m[Auxin efflux carrier component 3]
ARR5p[Two-component response regulator ARR5]
PHBp[Homeobox-leucine zipper protein ATHB-14]
ARR5m[Two-component response regulator ARR5]
Auxin[Auxin transporter protein 1]
miRNA[SBO:0000316]
PHBm[Homeobox-leucine zipper protein ATHB-14]
IAA2m[Auxin-responsive protein IAA2]
PIN1m[Peptidyl-prolyl cis-trans isomerase Pin1]
CKX3p[Cytokinin dehydrogenase 3]
AHP6p[Pseudo histidine-containing phosphotransfer protein 6]
PIN7m[Auxin efflux carrier component 7]

Murphy2016 - Differences in predictions of ODE models of tumor growth: BIOMD0000000671v0.0.1

Murphy2016 - Differences in predictions of ODE models of tumor growthComparison of 7 ODE models for tumour size. This mo…

Details

While mathematical models are often used to predict progression of cancer and treatment outcomes, there is still uncertainty over how to best model tumor growth. Seven ordinary differential equation (ODE) models of tumor growth (exponential, Mendelsohn, logistic, linear, surface, Gompertz, and Bertalanffy) have been proposed, but there is no clear guidance on how to choose the most appropriate model for a particular cancer.We examined all seven of the previously proposed ODE models in the presence and absence of chemotherapy. We derived equations for the maximum tumor size, doubling time, and the minimum amount of chemotherapy needed to suppress the tumor and used a sample data set to compare how these quantities differ based on choice of growth model.We find that there is a 12-fold difference in predicting doubling times and a 6-fold difference in the predicted amount of chemotherapy needed for suppression depending on which growth model was used.Our results highlight the need for careful consideration of model assumptions when developing mathematical models for use in cancer treatment planning. link: http://identifiers.org/pubmed/26921070

Parameters:

NameDescription
a_exp = 0.0246Reaction: V_exp = a_exp*V_exp, Rate Law: a_exp*V_exp
a_surf = 0.291; b_surf = 708.0Reaction: V_surf = a_surf*V_surf/(V_surf+b_surf)^(1/3), Rate Law: a_surf*V_surf/(V_surf+b_surf)^(1/3)
a_mend = 0.105; b_mend = 0.785Reaction: V_mend = a_mend*V_mend^b_mend, Rate Law: a_mend*V_mend^b_mend
a_bert = 0.2344; b_bert = 3.46E-19Reaction: V_bert = a_bert*V_bert^(2/3)-b_bert*V_bert, Rate Law: a_bert*V_bert^(2/3)-b_bert*V_bert
c_gomp = 10700.0; a_gomp = 0.0919; b_gomp = 15500.0Reaction: V_gomp = a_gomp*V_gomp*ln(b_gomp/(V_gomp+c_gomp)), Rate Law: a_gomp*V_gomp*ln(b_gomp/(V_gomp+c_gomp))
b_lin = 4300.0; a_lin = 132.0Reaction: V_lin = a_lin*V_lin/(V_lin+b_lin), Rate Law: a_lin*V_lin/(V_lin+b_lin)
b_log = 6920.0; a_log = 0.0295Reaction: V_log = a_log*V_log*(1-V_log/b_log), Rate Law: a_log*V_log*(1-V_log/b_log)

States:

NameDescription
V gompV_gomp
V surfV_surf
V linV_lin
V mendV_mend
V logV_log
V bertV_bert
V exp[Exponential Function]

Musante2017 - Switching behaviour of PP2A inhibition by ARPP-16 - mutual inhibitions: BIOMD0000000643v0.0.1

Musante2017 - Switching behaviour of PP2A inhibition by ARPP-16 - mutual inhibitionsThis model is described in the artic…

Details

ARPP-16, ARPP-19, and ENSA are inhibitors of protein phosphatase PP2A. ARPP-19 and ENSA phosphorylated by Greatwall kinase inhibit PP2A during mitosis. ARPP-16 is expressed in striatal neurons where basal phosphorylation by MAST3 kinase inhibits PP2A and regulates key components of striatal signaling. The ARPP-16/19 proteins were discovered as substrates for PKA, but the function of PKA phosphorylation is unknown. We find that phosphorylation by PKA or MAST3 mutually suppresses the ability of the other kinase to act on ARPP-16. Phosphorylation by PKA also acts to prevent inhibition of PP2A by ARPP-16 phosphorylated by MAST3. Moreover, PKA phosphorylates MAST3 at multiple sites resulting in its inhibition. Mathematical modeling highlights the role of these three regulatory interactions to create a switch-like response to cAMP. Together the results suggest a complex antagonistic interplay between the control of ARPP-16 by MAST3 and PKA that creates a mechanism whereby cAMP mediates PP2A disinhibition. link: http://identifiers.org/doi/10.7554/eLife.24998

Parameters:

NameDescription
ModelValue_1 = 2.0; kmpp2a = 0.0161515151515152Reaction: BB = A46+ModelValue_1+kmpp2a, Rate Law: missing
ModelValue_12 = 0.5; ModelValue_14 = 1.0; ModelValue_23 = 2.36; ModelValue_10 = 0.935; ModelValue_11 = 1.6; ModelValue_0 = 10.0; ModelValue_13 = 5.0Reaction: A88 = ModelValue_10*PKA*(ModelValue_0-A88)/((ModelValue_11+ModelValue_23*A46/ModelValue_0+ModelValue_0)-A88)-ModelValue_12*ModelValue_13*A88/(ModelValue_14+A88), Rate Law: ModelValue_10*PKA*(ModelValue_0-A88)/((ModelValue_11+ModelValue_23*A46/ModelValue_0+ModelValue_0)-A88)-ModelValue_12*ModelValue_13*A88/(ModelValue_14+A88)
ModelValue_1 = 2.0Reaction: Complex = (BB-(BB^2-4*A46*ModelValue_1)^(0.5))/2, Rate Law: missing
ModelValue_2 = 2.7; ModelValue_15 = 0.01865; kppx = 0.05Reaction: M = kppx*(ModelValue_2-M)-ModelValue_15*A88*M, Rate Law: kppx*(ModelValue_2-M)-ModelValue_15*A88*M
ModelValue_22 = 0.37526; ModelValue_9 = 0.09; ModelValue_6 = 0.05; ModelValue_0 = 10.0; ModelValue_8 = 0.0988Reaction: A46 = ModelValue_8*M*(ModelValue_0-A46)/(ModelValue_9+ModelValue_22*A88/ModelValue_0+(ModelValue_0-A46))-ModelValue_6*Complex, Rate Law: ModelValue_8*M*(ModelValue_0-A46)/(ModelValue_9+ModelValue_22*A88/ModelValue_0+(ModelValue_0-A46))-ModelValue_6*Complex
ModelValue_3 = 12.0; ModelValue_17 = 2.0; ModelValue_16 = 0.7; ModelValue_20 = 0.0; ModelValue_19 = 0.02335; ModelValue_18 = 10.0Reaction: PKA = ModelValue_16*(ModelValue_3-PKA)*ModelValue_20^ModelValue_17/(ModelValue_18^ModelValue_17+ModelValue_20^ModelValue_17)-ModelValue_19*A46*PKA, Rate Law: ModelValue_16*(ModelValue_3-PKA)*ModelValue_20^ModelValue_17/(ModelValue_18^ModelValue_17+ModelValue_20^ModelValue_17)-ModelValue_19*A46*PKA

States:

NameDescription
BB[urn:miriam:pr:PR%3AP56212-2]
A88[urn:miriam:pr:PR_P56212-2]
M[Microtubule-associated serine/threonine-protein kinase 3]
Complex[protein phosphatase type 2A complex]
PKA[cAMP-dependent protein kinase catalytic subunit alpha]
A46[urn:miriam:pr:PR_P56212-2]

Musante2017 - Switching behaviour of PP2A inhibition by ARPP-16 - mutual inhibitions and PKA inhibits MAST3: BIOMD0000000644v0.0.1

Musante2017 - Switching behaviour of PP2A inhibition by ARPP-16 - mutual inhibitions and PKA inhibits MAST3This model is…

Details

ARPP-16, ARPP-19, and ENSA are inhibitors of protein phosphatase PP2A. ARPP-19 and ENSA phosphorylated by Greatwall kinase inhibit PP2A during mitosis. ARPP-16 is expressed in striatal neurons where basal phosphorylation by MAST3 kinase inhibits PP2A and regulates key components of striatal signaling. The ARPP-16/19 proteins were discovered as substrates for PKA, but the function of PKA phosphorylation is unknown. We find that phosphorylation by PKA or MAST3 mutually suppresses the ability of the other kinase to act on ARPP-16. Phosphorylation by PKA also acts to prevent inhibition of PP2A by ARPP-16 phosphorylated by MAST3. Moreover, PKA phosphorylates MAST3 at multiple sites resulting in its inhibition. Mathematical modeling highlights the role of these three regulatory interactions to create a switch-like response to cAMP. Together the results suggest a complex antagonistic interplay between the control of ARPP-16 by MAST3 and PKA that creates a mechanism whereby cAMP mediates PP2A disinhibition. link: http://identifiers.org/doi/10.7554/eLife.24998

Parameters:

NameDescription
ModelValue_3 = 12.0; ModelValue_18 = 0.7; ModelValue_20 = 10.0; ModelValue_22 = 0.0; ModelValue_19 = 2.0; ModelValue_21 = 0.02335Reaction: PKA = ModelValue_18*(ModelValue_3-PKA)*ModelValue_22^ModelValue_19/(ModelValue_20^ModelValue_19+ModelValue_22^ModelValue_19)-ModelValue_21*A46*PKA, Rate Law: ModelValue_18*(ModelValue_3-PKA)*ModelValue_22^ModelValue_19/(ModelValue_20^ModelValue_19+ModelValue_22^ModelValue_19)-ModelValue_21*A46*PKA
ModelValue_1 = 2.0; kmpp2a = 0.0161515151515152Reaction: BB = A46+ModelValue_1+kmpp2a, Rate Law: missing
ModelValue_1 = 2.0Reaction: Complex = (BB-(BB^2-4*A46*ModelValue_1)^(0.5))/2, Rate Law: missing
ModelValue_12 = 0.5; ModelValue_14 = 1.0; ARPPtot = 10.0; ModelValue_28 = 2.36; ModelValue_10 = 0.935; ModelValue_11 = 1.6; ModelValue_0 = 10.0; ModelValue_13 = 5.0Reaction: A88 = ModelValue_10*PKA*(ARPPtot-A88)/((ModelValue_11+ModelValue_28*A46/ModelValue_0+ARPPtot)-A88)-ModelValue_12*ModelValue_13*A88/(ModelValue_14+A88), Rate Law: ModelValue_10*PKA*(ARPPtot-A88)/((ModelValue_11+ModelValue_28*A46/ModelValue_0+ARPPtot)-A88)-ModelValue_12*ModelValue_13*A88/(ModelValue_14+A88)
ModelValue_2 = 2.7; kpka = 0.097; ModelValue_17 = 0.01865; ModelValue_30 = 0.05Reaction: M = (ModelValue_30*(ModelValue_2-M)-ModelValue_17*A88*M)-kpka*PKA*M, Rate Law: (ModelValue_30*(ModelValue_2-M)-ModelValue_17*A88*M)-kpka*PKA*M
ModelValue_2 = 2.7; ARPPtot = 10.0; ModelValue_9 = 0.09; ModelValue_29 = 1.2; ModelValue_6 = 0.05; ModelValue_0 = 10.0; ModelValue_8 = 0.0988; ModelValue_25 = 0.37526Reaction: A46 = ModelValue_8*M*(ARPPtot-A46)/(ModelValue_9+ModelValue_25*A88/ModelValue_0+ModelValue_29*(ModelValue_2-M)/ModelValue_2+(ARPPtot-A46))-ModelValue_6*Complex, Rate Law: ModelValue_8*M*(ARPPtot-A46)/(ModelValue_9+ModelValue_25*A88/ModelValue_0+ModelValue_29*(ModelValue_2-M)/ModelValue_2+(ARPPtot-A46))-ModelValue_6*Complex

States:

NameDescription
BB[urn:miriam:pr:PR%3AP56212-2]
A88[urn:miriam:pr:PR_P56212-2]
M[Microtubule-associated serine/threonine-protein kinase 3]
Complex[protein phosphatase type 2A complex]
PKA[cAMP-dependent protein kinase catalytic subunit alpha]
A46[urn:miriam:pr:PR_P56212-2]

Musante2017 - Switching behaviour of PP2A inhibition by ARPP-16 - mutual inhibitions and PKA inhibits MAST3 and dominant negative effect: BIOMD0000000645v0.0.1

Musante2017 - Switching behaviour of PP2A inhibition by ARPP-16 - mutual inhibitions and PKA inhibits MAST3 and dominant…

Details

ARPP-16, ARPP-19, and ENSA are inhibitors of protein phosphatase PP2A. ARPP-19 and ENSA phosphorylated by Greatwall kinase inhibit PP2A during mitosis. ARPP-16 is expressed in striatal neurons where basal phosphorylation by MAST3 kinase inhibits PP2A and regulates key components of striatal signaling. The ARPP-16/19 proteins were discovered as substrates for PKA, but the function of PKA phosphorylation is unknown. We find that phosphorylation by PKA or MAST3 mutually suppresses the ability of the other kinase to act on ARPP-16. Phosphorylation by PKA also acts to prevent inhibition of PP2A by ARPP-16 phosphorylated by MAST3. Moreover, PKA phosphorylates MAST3 at multiple sites resulting in its inhibition. Mathematical modeling highlights the role of these three regulatory interactions to create a switch-like response to cAMP. Together the results suggest a complex antagonistic interplay between the control of ARPP-16 by MAST3 and PKA that creates a mechanism whereby cAMP mediates PP2A disinhibition. link: http://identifiers.org/doi/10.7554/eLife.24998

Parameters:

NameDescription
ModelValue_3 = 12.0; ModelValue_18 = 0.7; ModelValue_20 = 10.0; ModelValue_22 = 0.0; ModelValue_19 = 2.0; ModelValue_21 = 0.02335Reaction: PKA = ModelValue_18*(ModelValue_3-PKA)*ModelValue_22^ModelValue_19/(ModelValue_20^ModelValue_19+ModelValue_22^ModelValue_19)-ModelValue_21*A46*PKA, Rate Law: ModelValue_18*(ModelValue_3-PKA)*ModelValue_22^ModelValue_19/(ModelValue_20^ModelValue_19+ModelValue_22^ModelValue_19)-ModelValue_21*A46*PKA
ModelValue_1 = 2.0Reaction: Complex = (BB-(BB^2-4*A46*ModelValue_1)^(0.5))/2, Rate Law: missing
ModelValue_12 = 0.5; ModelValue_14 = 1.0; ARPPtot = 10.0; ModelValue_28 = 2.36; ModelValue_10 = 0.935; ModelValue_11 = 1.6; ModelValue_0 = 10.0; ModelValue_13 = 5.0Reaction: A88 = ModelValue_10*PKA*(ARPPtot-A88)/((ModelValue_11+ModelValue_28*A46/ModelValue_0+ARPPtot)-A88)-ModelValue_12*ModelValue_13*A88/(ModelValue_14+A88), Rate Law: ModelValue_10*PKA*(ARPPtot-A88)/((ModelValue_11+ModelValue_28*A46/ModelValue_0+ARPPtot)-A88)-ModelValue_12*ModelValue_13*A88/(ModelValue_14+A88)
ModelValue_2 = 2.7; kpka = 0.097; ModelValue_17 = 0.01865; ModelValue_33 = 0.05Reaction: M = (ModelValue_33*(ModelValue_2-M)-ModelValue_17*A88*M)-kpka*PKA*M, Rate Law: (ModelValue_33*(ModelValue_2-M)-ModelValue_17*A88*M)-kpka*PKA*M
ModelValue_1 = 2.0; kmpp2a = 0.0484545454545436Reaction: BB = A46+ModelValue_1+kmpp2a, Rate Law: missing
ModelValue_2 = 2.7; ARPPtot = 10.0; ModelValue_9 = 0.09; ModelValue_29 = 1.2; ModelValue_6 = 0.05; ModelValue_0 = 10.0; ModelValue_8 = 0.0988; ModelValue_25 = 0.37526Reaction: A46 = ModelValue_8*M*(ARPPtot-A46)/(ModelValue_9+ModelValue_25*A88/ModelValue_0+ModelValue_29*(ModelValue_2-M)/ModelValue_2+(ARPPtot-A46))-ModelValue_6*Complex, Rate Law: ModelValue_8*M*(ARPPtot-A46)/(ModelValue_9+ModelValue_25*A88/ModelValue_0+ModelValue_29*(ModelValue_2-M)/ModelValue_2+(ARPPtot-A46))-ModelValue_6*Complex

States:

NameDescription
BB[urn:miriam:pr:PR_P56212-2]
A88[urn:miriam:pr:PR_P56212-2]
M[Microtubule-associated serine/threonine-protein kinase 3]
Complex[protein phosphatase type 2A complex]
PKA[cAMP-dependent protein kinase catalytic subunit alpha]
A46[urn:miriam:pr:PR_P56212-2]

Mushayabasa2011- Modeling gonirrhea and HIV co-interaction: MODEL1812040004v0.0.1

A mathematical model was designed to explore the co-interaction of gonorrhea and HIV in the presence of antiretroviral t…

Details

A mathematical model was designed to explore the co-interaction of gonorrhea and HIV in the presence of antiretroviral therapy and gonorrhea treatment. Qualitative and comprehensive mathematical techniques have been used to analyse the model. The gonorrhea-only and HIV-only sub-models are first considered. Analytic expressions for the threshold parameter in each sub-model and the co-interaction model are derived. Global dynamics of this co-interaction shows that whenever the threshold parameter for the respective sub-models and co-interaction model is less than unity, the epidemics dies out, while the reverse results in persistence of the epidemics in the community. The impact of gonorrhea and its treatment on HIV dynamics is also investigated. Numerical simulations using a set of reasonable parameter values show that the two epidemics co-exists whenever their reproduction numbers exceed unity (with no competitive exclusion). Further, simulations of the full HIV-gonorrhea model also suggests that an increase in the number of individuals infected with gonorrhea (either singly or dually with HIV) in the presence of treatment results in a decrease in gonorrhea-only cases, dual-infection cases but increases the number of HIV-only cases. link: http://identifiers.org/pubmed/20869424

Mwalili2020 - SEIR model of COVID-19 transmission and environmental pathogen prevalence: BIOMD0000000964v0.0.1

Objective: Coronavirus disease 2019 (COVID-19) is a pandemic respiratory illness spreading from person-to-person caused…

Details

OBJECTIVE:Coronavirus disease 2019 (COVID-19) is a pandemic respiratory illness spreading from person-to-person caused by a novel coronavirus and poses a serious public health risk. The goal of this study was to apply a modified susceptible-exposed-infectious-recovered (SEIR) compartmental mathematical model for prediction of COVID-19 epidemic dynamics incorporating pathogen in the environment and interventions. The next generation matrix approach was used to determine the basic reproduction number [Formula: see text]. The model equations are solved numerically using fourth and fifth order Runge-Kutta methods. RESULTS:We found an [Formula: see text] of 2.03, implying that the pandemic will persist in the human population in the absence of strong control measures. Results after simulating various scenarios indicate that disregarding social distancing and hygiene measures can have devastating effects on the human population. The model shows that quarantine of contacts and isolation of cases can help halt the spread on novel coronavirus. link: http://identifiers.org/pubmed/32703315

Mwasa2011 - Cholera model with public health interventions: MODEL1812040007v0.0.1

Mathematical analysis of a cholera model with public health interventions

Details

Cholera, an acute gastro-intestinal infection and a waterborne disease continues to emerge in developing countries and remains an important global health challenge. We formulate a mathematical model that captures some essential dynamics of cholera transmission to study the impact of public health educational campaigns, vaccination and treatment as control strategies in curtailing the disease. The education-induced, vaccination-induced and treatment-induced reproductive numbers R(E), R(V), R(T) respectively and the combined reproductive number R(C) are compared with the basic reproduction number R(0) to assess the possible community benefits of these control measures. A Lyapunov functional approach is also used to analyse the stability of the equilibrium points. We perform sensitivity analysis on the key parameters that drive the disease dynamics in order to determine their relative importance to disease transmission and prevalence. Graphical representations are provided to qualitatively support the analytical results. link: http://identifiers.org/pubmed/null

N


Nag2011_ChloroplasticStarchDegradation: BIOMD0000000353v0.0.1

This model is from the article: Kinetic modeling and exploratory numerical simulation of chloroplastic starch degrad…

Details

BACKGROUND: Higher plants and algae are able to fix atmospheric carbon dioxide through photosynthesis and store this fixed carbon in large quantities as starch, which can be hydrolyzed into sugars serving as feedstock for fermentation to biofuels and precursors. Rational engineering of carbon flow in plant cells requires a greater understanding of how starch breakdown fluxes respond to variations in enzyme concentrations, kinetic parameters, and metabolite concentrations. We have therefore developed and simulated a detailed kinetic ordinary differential equation model of the degradation pathways for starch synthesized in plants and green algae, which to our knowledge is the most complete such model reported to date. RESULTS: Simulation with 9 internal metabolites and 8 external metabolites, the concentrations of the latter fixed at reasonable biochemical values, leads to a single reference solution showing β-amylase activity to be the rate-limiting step in carbon flow from starch degradation. Additionally, the response coefficients for stromal glucose to the glucose transporter k(cat) and KM are substantial, whereas those for cytosolic glucose are not, consistent with a kinetic bottleneck due to transport. Response coefficient norms show stromal maltopentaose and cytosolic glucosylated arabinogalactan to be the most and least globally sensitive metabolites, respectively, and β-amylase k(cat) and KM for starch to be the kinetic parameters with the largest aggregate effect on metabolite concentrations as a whole. The latter kinetic parameters, together with those for glucose transport, have the greatest effect on stromal glucose, which is a precursor for biofuel synthetic pathways. Exploration of the steady-state solution space with respect to concentrations of 6 external metabolites and 8 dynamic metabolite concentrations show that stromal metabolism is strongly coupled to starch levels, and that transport between compartments serves to lower coupling between metabolic subsystems in different compartments. CONCLUSIONS: We find that in the reference steady state, starch cleavage is the most significant determinant of carbon flux, with turnover of oligosaccharides playing a secondary role. Independence of stationary point with respect to initial dynamic variable values confirms a unique stationary point in the phase space of dynamically varying concentrations of the model network. Stromal maltooligosaccharide metabolism was highly coupled to the available starch concentration. From the most highly converged trajectories, distances between unique fixed points of phase spaces show that cytosolic maltose levels depend on the total concentrations of arabinogalactan and glucose present in the cytosol. In addition, cellular compartmentalization serves to dampen much, but not all, of the effects of one subnetwork on another, such that kinetic modeling of single compartments would likely capture most dynamics that are fast on the timescale of the transport reactions. link: http://identifiers.org/pubmed/21682905

Parameters:

NameDescription
R06050CY_GlcAG_KM = 2100.0 µmol/l; R06050CY_G1P_KM = 2000.0 µmol/l; R06050CY_GlcAG_Ki = 3800.0 µmol/l; R06050CY_AG_KM = 3800.0 µmol/l; R06050CY_Pi_KM = 5900.0 µmol/l; R06050CY_kcat = 50.0 1/s; R06050CY_Keq = 6.15E-4 1; R06050CY_G1P_Ki = 3100.0 µmol/lReaction: cpd_C00569Glc_CY + cpd_C00009tot_CY => cpd_C00103tot_CY + cpd_C00569_CY; ec_2_4_1_1_CY, Rate Law: Cytosol*R06050CY_kcat*ec_2_4_1_1_CY/Cytosol*(cpd_C00569Glc_CY/Cytosol*cpd_C00009tot_CY/Cytosol-cpd_C00103tot_CY/Cytosol*cpd_C00569_CY/Cytosol/R06050CY_Keq)/(R06050CY_GlcAG_Ki*R06050CY_Pi_KM+R06050CY_Pi_KM*cpd_C00569Glc_CY/Cytosol+R06050CY_GlcAG_KM*cpd_C00009tot_CY/Cytosol+cpd_C00569Glc_CY/Cytosol*cpd_C00009tot_CY/Cytosol+R06050CY_GlcAG_Ki*R06050CY_Pi_KM/(R06050CY_G1P_Ki*R06050CY_AG_KM)*(R06050CY_AG_KM*cpd_C00103tot_CY/Cytosol+R06050CY_G1P_KM*cpd_C00569_CY/Cytosol+cpd_C00103tot_CY/Cytosol*cpd_C00569_CY/Cytosol))
f_bamylase = 0.582 1; conv_gm_umole = 1.0 µg/mol; R02112CS_Gn_KM = 0.5 g/l; f_G3 = 0.13 1; R02112CS_Gn_kcat = 0.073 1/sReaction: cpd_C00369Glc_CS => cpd_C01835_CS; ec_3_2_1_2_CS, cpd_C00369_CS, cpd_C00369db_CS, Rate Law: ChloroplastStroma*R02112CS_Gn_kcat*ec_3_2_1_2_CS/ChloroplastStroma*f_G3*(f_bamylase*cpd_C00369_CS/ChloroplastStroma+cpd_C00369db_CS/ChloroplastStroma)/(conv_gm_umole*(f_G3*(f_bamylase*cpd_C00369_CS/ChloroplastStroma+cpd_C00369db_CS/ChloroplastStroma)+R02112CS_Gn_KM))
R02112CS_G2C_KM = 4.19 g²/l²; f_bamylase = 0.582 1; f_G2 = 0.87 1; conv_gm_umole = 1.0 µg/mol; R02112CS_Keq = 18800.0 g/l; R02112CS_Gn_KM = 0.5 g/l; C00208_MW = 3.42E-4 µg/mol; R02112CS_Gn_kcat = 0.073 1/sReaction: cpd_C00369Glc_CS => cpd_C00208_CS; ec_3_2_1_2_CS, cpd_C00369_CS, cpd_C00369db_CS, Rate Law: ChloroplastStroma*R02112CS_Gn_kcat*ec_3_2_1_2_CS/ChloroplastStroma*(f_G2*(f_bamylase*cpd_C00369_CS/ChloroplastStroma+cpd_C00369db_CS/ChloroplastStroma)-(cpd_C00208_CS/ChloroplastStroma*C00208_MW)^2/R02112CS_Keq)/(conv_gm_umole*(f_G2*(f_bamylase*cpd_C00369_CS/ChloroplastStroma+cpd_C00369db_CS/ChloroplastStroma)+R02112CS_Gn_KM*(1+(cpd_C00208_CS/ChloroplastStroma*C00208_MW)^2/R02112CS_G2C_KM)))
f_bamylase = 0.582 1; ec_3_2_1_68_CS_kcat = 0.0198 1/sReaction: cpd_C00369db_CS = ec_3_2_1_68_CS/ChloroplastStroma*ec_3_2_1_68_CS_kcat*((1-1/(1+exp((-100)*(cpd_C00369db_CS/ChloroplastStroma/(cpd_C00369_CS/ChloroplastStroma*(1-f_bamylase))-0.3))))+1/(1+exp((-100)*(cpd_C00369db_CS/ChloroplastStroma/(cpd_C00369_CS/ChloroplastStroma*(1-f_bamylase))-0.3)))*(1-1.429*(cpd_C00369db_CS/ChloroplastStroma/(cpd_C00369_CS/ChloroplastStroma*(1-f_bamylase))-0.3)))*ChloroplastStroma, Rate Law: ec_3_2_1_68_CS/ChloroplastStroma*ec_3_2_1_68_CS_kcat*((1-1/(1+exp((-100)*(cpd_C00369db_CS/ChloroplastStroma/(cpd_C00369_CS/ChloroplastStroma*(1-f_bamylase))-0.3))))+1/(1+exp((-100)*(cpd_C00369db_CS/ChloroplastStroma/(cpd_C00369_CS/ChloroplastStroma*(1-f_bamylase))-0.3)))*(1-1.429*(cpd_C00369db_CS/ChloroplastStroma/(cpd_C00369_CS/ChloroplastStroma*(1-f_bamylase))-0.3)))*ChloroplastStroma
TC_2_A_1_1_17_KM = 19300.0 µmol/l; TC_2_A_1_1_17_kcat = 240.278 1/sReaction: cpd_C00031_CS => cpd_C00031_CY; tc_2_A_1_1_17_CIMS, Rate Law: ChloroplastStroma*TC_2_A_1_1_17_kcat*tc_2_A_1_1_17_CIMS/ChloroplastIntermembraneSpace*cpd_C00031_CS/ChloroplastStroma/(TC_2_A_1_1_17_KM+cpd_C00031_CS/ChloroplastStroma)
R05196CS_G3_Ki = 746.42 µmol/l; R05196CS_G5_Ki = 100.0 µmol/l; R05196CS_Glc_KM = 11700.0 µmol/l; R05196CS_G3_KM = 3300.0 µmol/l; R05196CS_Keq = 1.0 1; R05196CS_G5_KM = 210.0 µmol/l; R05196CS_kcat = 50.0 1/sReaction: cpd_C01835_CS => cpd_C00031_CS + cpd_G00343_CS; ec_2_4_1_25_CS, Rate Law: ChloroplastStroma*R05196CS_kcat*ec_2_4_1_25_CS/ChloroplastStroma*((cpd_C01835_CS/ChloroplastStroma)^2-cpd_C00031_CS/ChloroplastStroma*cpd_G00343_CS/ChloroplastStroma/R05196CS_Keq)/(R05196CS_G3_KM*cpd_C01835_CS/ChloroplastStroma+(cpd_C01835_CS/ChloroplastStroma)^2+R05196CS_G3_KM*R05196CS_G3_Ki/(R05196CS_Glc_KM*R05196CS_G5_Ki)*(R05196CS_G5_KM*cpd_C00031_CS/ChloroplastStroma*(1+cpd_C01835_CS/ChloroplastStroma/R05196CS_G3_Ki)+R05196CS_Glc_KM*cpd_G00343_CS/ChloroplastStroma*(1+cpd_C01835_CS/ChloroplastStroma/R05196CS_G3_Ki)+cpd_C00031_CS/ChloroplastStroma*cpd_G00343_CS/ChloroplastStroma))
TC_2_A_84_1_2_kcat = 5.963 1/s; TC_2_A_84_1_2_KM = 4000.0 µmol/lReaction: cpd_C00208_CS => cpd_C00208_CY; tc_2_A_84_1_2_CIMS, Rate Law: ChloroplastStroma*TC_2_A_84_1_2_kcat*tc_2_A_84_1_2_CIMS/ChloroplastIntermembraneSpace*cpd_C00208_CS/ChloroplastStroma/(TC_2_A_84_1_2_KM+cpd_C00208_CS/ChloroplastStroma)
AT2G40840CY_Keq = 1.0 1; AT2G40840CY_G2_KM = 4600.0 µmol/l; AT2G40840CY_Glc_KM = 11700.0 µmol/l; AT2G40840CY_G2_Ki = 2190.476 µmol/l; AT2G40840CY_AG_Ki = 1000.0 µmol/l; AT2G40840CY_GlcAG_KM = 1100.0 µmol/l; AT2G40840CY_AG_KM = 1100.0 µmol/l; AT2G40840CY_kcat = 50.0 1/s; AT2G40840CY_GlcAG_Ki = 1000.0 µmol/lReaction: cpd_C00208_CY + cpd_C00569_CY => cpd_C00031_CY + cpd_C00569Glc_CY; ec_2_4_1_25_CY, Rate Law: Cytosol*AT2G40840CY_kcat*ec_2_4_1_25_CY/Cytosol*(cpd_C00208_CY/Cytosol*cpd_C00569_CY/Cytosol-cpd_C00031_CY/Cytosol*cpd_C00569Glc_CY/Cytosol/AT2G40840CY_Keq)/(AT2G40840CY_AG_KM*cpd_C00208_CY/Cytosol+AT2G40840CY_G2_KM*cpd_C00569_CY/Cytosol+cpd_C00208_CY/Cytosol*cpd_C00569_CY/Cytosol+AT2G40840CY_G2_KM*AT2G40840CY_AG_Ki/(AT2G40840CY_Glc_KM*AT2G40840CY_GlcAG_Ki)*(AT2G40840CY_GlcAG_KM*cpd_C00031_CY/Cytosol*(1+cpd_C00208_CY/Cytosol/AT2G40840CY_G2_Ki)+AT2G40840CY_Glc_KM*cpd_C00569Glc_CY/Cytosol*(1+cpd_C00569_CY/Cytosol/AT2G40840CY_AG_Ki)+cpd_C00031_CY/Cytosol*cpd_C00569Glc_CY/Cytosol))
R02112CS_G5_kcat = 0.0913 1/s; G00343_MW = 8.28E-4 µg/mol; conv_gm_umole = 1.0 µg/mol; R02112CS_G5_KM = 1.46 g/lReaction: cpd_G00343_CS => cpd_C00208_CS + cpd_C01835_CS; ec_3_2_1_2_CS, Rate Law: ChloroplastStroma*R02112CS_G5_kcat*ec_3_2_1_2_CS/ChloroplastStroma*cpd_G00343_CS/ChloroplastStroma*G00343_MW/(conv_gm_umole*(cpd_G00343_CS/ChloroplastStroma*G00343_MW+R02112CS_G5_KM))
C00369_MW = 0.27 µg/mol; N_Glc_Starch = 1667.0 1Reaction: cpd_C00369_CS = cpd_C00369Glc_CS/ChloroplastStroma*C00369_MW/N_Glc_Starch*ChloroplastStroma, Rate Law: missing
R00299CY_G6P_KM = 47.0 µmol/l; R00299CY_kfor = 180.0 1/s; R00299CY_G16P_Kip = 30.0 µmol/l; R00299CY_Glc_Ki = 47.0 µmol/l; R00299CY_MgADP_Ki = 1000.0 µmol/l; R00299CY_MgATP_Ki = 1000.0 µmol/l; R00299CY_G6P_Kip = 10.0 µmol/l; R00299CY_krev = 1.16129032258065 1/s; R00299CY_BPG_Kip = 4000.0 µmol/l; R00299CY_GSH_Kip = 3000.0 µmol/l; R00299CY_MgATP_KM = 1000.0 µmol/l; R00299CY_G6P_Ki = 47.0 µmol/lReaction: cpd_C00002tot_CY + cpd_C00031_CY => cpd_C00092tot_CY + cpd_C00008tot_CY + cpd_C00080_CY; ec_2_7_1_1_CY, cpd_C00051_CY, cpd_C00660tot_CY, cpd_C03339tot_CY, Rate Law: Cytosol*ec_2_7_1_1_CY/Cytosol*(R00299CY_kfor*cpd_C00002tot_CY/Cytosol*cpd_C00031_CY/Cytosol/(R00299CY_Glc_Ki*R00299CY_MgATP_KM)-R00299CY_krev*cpd_C00092tot_CY/Cytosol*cpd_C00008tot_CY/Cytosol/(R00299CY_MgADP_Ki*R00299CY_G6P_KM))/(1+cpd_C00002tot_CY/Cytosol/R00299CY_MgATP_Ki+cpd_C00031_CY/Cytosol/R00299CY_Glc_Ki*(1+cpd_C00092tot_CY/Cytosol/R00299CY_G6P_Kip+cpd_C00660tot_CY/Cytosol/R00299CY_G16P_Kip+cpd_C03339tot_CY/Cytosol/R00299CY_BPG_Kip+cpd_C00051_CY/Cytosol/R00299CY_GSH_Kip)+cpd_C00002tot_CY/Cytosol*cpd_C00031_CY/Cytosol/(R00299CY_Glc_Ki*R00299CY_MgATP_KM)+cpd_C00092tot_CY/Cytosol/R00299CY_G6P_Ki+cpd_C00008tot_CY/Cytosol/R00299CY_MgADP_Ki+cpd_C00092tot_CY/Cytosol*cpd_C00008tot_CY/Cytosol/(R00299CY_MgADP_Ki*R00299CY_G6P_KM))

States:

NameDescription
cpd C00569 CY[simple chemical; arabinogalactan; Arabinogalactan]
cpd C00002tot CY[simple chemical; ATP; ATP]
cpd C00080 CY[non-macromolecular ion; H+; proton]
cpd C00103tot CY[simple chemical; C11450; alpha-D-glucose 1-phosphate]
cpd C00208 CY[simple chemical; maltose; Maltose]
cpd C00369Glc CS[simple chemical; MOD:00726; Starch; starch]
cpd C00031 CS[simple chemical; D-glucose]
cpd C00031 CY[simple chemical; D-Glucose; D-glucose]
cpd C00569Glc CY[simple chemical; arabinogalactan; Arabinogalactan; MOD:00726]
cpd C00369db CS[simple chemical; Starch; starch; MOD:00726]
cpd C00009tot CY[simple chemical; Orthophosphate; phosphate(3-)]
cpd C00208 CS[simple chemical; maltose; Maltose]
cpd C00369 CS[simple chemical; Starch; starch]
cpd G00343 CS[simple chemical; maltopentaose]
cpd C00092tot CY[simple chemical; alpha-D-Glucose 6-phosphate; alpha-D-glucose 6-phosphate]
cpd C01835 CS[simple chemical; maltotriose; Maltotriose]
cpd C00008tot CY[simple chemical; ADP; ADP]

Nagashima2002 - Simulating blood coagulation inhibitory effects: BIOMD0000000747v0.0.1

Mathematical model of blood coagulation and the effects of inhibitors of Xa, Va:Xa and IIa.

Details

The present study began with mathematical modeling of how inhibitors of both factor Xa (fXa) and thrombin affect extrinsic pathway-triggered blood coagulation. Numerical simulation demonstrated a stronger inhibition of thrombin generation by a thrombin inhibitor than a fXa inhibitor, but both prolonged clot time to a similar extent when they were given an equal dissociation constant (30 nm) for interaction with their respective target enzymes. These differences were then tested by comparison with the real inhibitors DX-9065a and argatroban, specific competitive inhibitors of fXa and thrombin, respectively, with similar K(i) values. Comparisons were made in extrinsically triggered human citrated plasma, for which endogenous thrombin potential and clot formation were simultaneously measured with a Wallac multilabel counter equipped with both fluorometric and photometric detectors and a fluorogenic reporter substrate. The results demonstrated stronger inhibition of endogenous thrombin potential by argatroban than by DX-9065a, especially when coagulation was initiated at higher tissue factor concentrations, while argatroban appeared to be slightly less potent in its ability to prolong clot time. This study demonstrates differential inhibition of thrombin generation by fXa and thrombin inhibitors and has implications for the pharmacological regulation of blood coagulation by the anticoagulant protease inhibitors. link: http://identifiers.org/pubmed/12496240

Parameters:

NameDescription
k02 = 2.2; k01 = 0.1Reaction: TF_VIIa + IX => TF_VIIa_IX, Rate Law: compartment*(k01*TF_VIIa*IX-k02*TF_VIIa_IX)
k17 = 29.0Reaction: VIIIa_IXa_X => VIIIa_IXa + Xa, Rate Law: compartment*k17*VIIIa_IXa_X
k24 = 0.1; k25 = 0.1Reaction: Xa + Va => Va_Xa, Rate Law: compartment*(k24*Xa*Va-k25*Va_Xa)
k31 = 84.0Reaction: Fibrinogen_IIa => Fibrin + IIa, Rate Law: compartment*k31*Fibrinogen_IIa
k41 = 3.0; k40 = 0.1Reaction: Va_Xa + Xa_Inhibitor => Va_Xa_Xa_Inhibitor, Rate Law: compartment*(k40*Va_Xa*Xa_Inhibitor-k41*Va_Xa_Xa_Inhibitor)
k06 = 1.4Reaction: TF_VIIa_X => TF_VIIa + Xa, Rate Law: compartment*k06*TF_VIIa_X
k36 = 0.1; k37 = 3.0Reaction: Xa + Xa_Inhibitor => Xa_Xa_Inhibitor, Rate Law: compartment*(k36*Xa*Xa_Inhibitor-k37*Xa_Xa_Inhibitor)
k08 = 2.1; k07 = 0.1Reaction: Xa + VIII => Xa_VIII, Rate Law: compartment*(k07*Xa*VIII-k08*Xa_VIII)
k43 = 3.0; k42 = 0.1Reaction: IIa + IIa_Inhibitor => IIa_IIa_Inhibitor, Rate Law: compartment*(k42*IIa*IIa_Inhibitor-k43*IIa_IIa_Inhibitor)
k16 = 19.0; k15 = 0.1Reaction: VIIIa_IXa + X => VIIIa_IXa_X, Rate Law: compartment*(k15*VIIIa_IXa*X-k16*VIIIa_IXa_X)
k30 = 720.0; k29 = 0.1Reaction: Fibrinogen + IIa => Fibrinogen_IIa, Rate Law: compartment*(k29*Fibrinogen*IIa-k30*Fibrinogen_IIa)
k28 = 35.0Reaction: Va_Xa_II => Va_Xa + IIa, Rate Law: compartment*k28*Va_Xa_II
k14 = 0.17; k13 = 0.1Reaction: VIIIa + IXa => VIIIa_IXa, Rate Law: compartment*(k13*VIIIa*IXa-k14*VIIIa_IXa)
k04 = 0.1; k05 = 5.5Reaction: TF_VIIa + X => TF_VIIa_X, Rate Law: compartment*(k04*TF_VIIa*X-k05*TF_VIIa_X)
k12 = 0.9Reaction: IIa_VIII => IIa + VIIIa, Rate Law: compartment*k12*IIa_VIII
k34 = 0.011Reaction: Xa => Xa_inact, Rate Law: compartment*k34*Xa
k20 = 0.043Reaction: Xa_V => Xa + Va, Rate Law: compartment*k20*Xa_V
k18 = 0.1; k19 = 1.0Reaction: Xa + V => Xa_V, Rate Law: compartment*(k18*Xa*V-k19*Xa_V)
k32 = 0.0011Reaction: VIIIa => VIIIa_inact, Rate Law: compartment*k32*VIIIa
k35 = 0.024Reaction: IIa => IIa_inact, Rate Law: compartment*k35*IIa
k11 = 15.0; k10 = 0.1Reaction: IIa + VIII => IIa_VIII, Rate Law: compartment*(k10*IIa*VIII-k11*IIa_VIII)
k33 = 0.0017Reaction: IXa => IXa_inact, Rate Law: compartment*k33*IXa
k23 = 0.26Reaction: IIa_V => IIa + Va, Rate Law: compartment*k23*IIa_V
k03 = 0.47Reaction: TF_VIIa_IX => TF_VIIa + IXa, Rate Law: compartment*k03*TF_VIIa_IX
k38 = 0.1; k39 = 0.1Reaction: Va + Xa_Xa_Inhibitor => Va_Xa_Xa_Inhibitor, Rate Law: compartment*(k38*Va*Xa_Xa_Inhibitor-k39*Va_Xa_Xa_Inhibitor)
k22 = 7.2; k21 = 0.1Reaction: IIa + V => IIa_V, Rate Law: compartment*(k21*IIa*V-k22*IIa_V)
k26 = 0.1; k27 = 100.0Reaction: Va_Xa + II => Va_Xa_II, Rate Law: compartment*(k26*Va_Xa*II-k27*Va_Xa_II)
k09 = 0.023Reaction: Xa_VIII => Xa + VIIIa, Rate Law: compartment*k09*Xa_VIII

States:

NameDescription
IIa inact[Thrombin]
VIIIa IXa X[Coagulation Factor X Human; Coagulation Factor IX Human; Coagulation Factor VIII]
VIII[Coagulation Factor VIII]
Fibrin[Fibrin]
IIa IIa Inhibitor[EC 3.4.21.5 (thrombin) inhibitor; Thrombin]
V[Coagulation Factor V]
Xa VIII[Coagulation Factor VIII; Coagulation Factor X Human]
Xa[Coagulation Factor X Human]
IIa Inhibitor[EC 3.4.21.5 (thrombin) inhibitor]
VIIIa inact[Coagulation Factor VIII]
Va Xa[Coagulation Factor X Human; Coagulation Factor V]
IIa VIII[Coagulation Factor VIII; Thrombin]
TF VIIa X[Coagulation Factor X Human; Coagulation Factor VII Human; Tissue Factor]
Fibrinogen IIa[Thrombin; Fibrinogen]
Xa V[Coagulation Factor V; Coagulation Factor X Human]
Xa Xa Inhibitor[EC 3.4.21.5 (thrombin) inhibitor; Coagulation Factor X Human]
Fibrinogen[Fibrinogen]
X[Coagulation Factor X Human]
Va Xa II[Coagulation Factor X Human; Prothrombin; Coagulation Factor V]
Xa inact[Coagulation Factor X Human]
TF VIIa[Coagulation Factor VII Human; Tissue Factor]
VIIIa[Coagulation Factor VIII]
Xa Inhibitor[EC 3.4.21.6 (coagulation factor Xa) inhibitor]
IIa V[Coagulation Factor V; Thrombin]
Va[Coagulation Factor V]
IIa[Thrombin]
TF VIIa IX[Coagulation Factor IX Human; Tissue Factor; Coagulation Factor VII Human]
Va Xa Xa Inhibitor[Coagulation Factor X Human; Coagulation Factor V; EC 3.4.21.6 (coagulation factor Xa) inhibitor]
IXa[Coagulation Factor IX Human]
VIIIa IXa[Coagulation Factor IX Human; Coagulation Factor VIII]
II[Prothrombin]
IX[Coagulation Factor IX Human]
IXa inact[Coagulation Factor IX Human]

Nair2015 - Interaction between neuromodulators via GPCRs - Effect on cAMP/PKA signaling (D1 Neuron): BIOMD0000000635v0.0.1

Nair2015 - Interaction between neuromodulators via GPCRs - Effect on cAMP/PKA signaling (D1 Neuron)This model is describ…

Details

Transient changes in striatal dopamine (DA) concentration are considered to encode a reward prediction error (RPE) in reinforcement learning tasks. Often, a phasic DA change occurs concomitantly with a dip in striatal acetylcholine (ACh), whereas other neuromodulators, such as adenosine (Adn), change slowly. There are abundant adenylyl cyclase (AC) coupled GPCRs for these neuromodulators in striatal medium spiny neurons (MSNs), which play important roles in plasticity. However, little is known about the interaction between these neuromodulators via GPCRs. The interaction between these transient neuromodulator changes and the effect on cAMP/PKA signaling via Golf- and Gi/o-coupled GPCR are studied here using quantitative kinetic modeling. The simulations suggest that, under basal conditions, cAMP/PKA signaling could be significantly inhibited in D1R+ MSNs via ACh/M4R/Gi/o and an ACh dip is required to gate a subset of D1R/Golf-dependent PKA activation. Furthermore, the interaction between ACh dip and DA peak, via D1R and M4R, is synergistic. In a similar fashion, PKA signaling in D2+ MSNs is under basal inhibition via D2R/Gi/o and a DA dip leads to a PKA increase by disinhibiting A2aR/Golf, but D2+ MSNs could also respond to the DA peak via other intracellular pathways. This study highlights the similarity between the two types of MSNs in terms of high basal AC inhibition by Gi/o and the importance of interactions between Gi/o and Golf signaling, but at the same time predicts differences between them with regard to the sign of RPE responsible for PKA activation.Dopamine transients are considered to carry reward-related signal in reinforcement learning. An increase in dopamine concentration is associated with an unexpected reward or salient stimuli, whereas a decrease is produced by omission of an expected reward. Often dopamine transients are accompanied by other neuromodulatory signals, such as acetylcholine and adenosine. We highlight the importance of interaction between acetylcholine, dopamine, and adenosine signals via adenylyl-cyclase coupled GPCRs in shaping the dopamine-dependent cAMP/PKA signaling in striatal neurons. Specifically, a dopamine peak and an acetylcholine dip must interact, via D1 and M4 receptor, and a dopamine dip must interact with adenosine tone, via D2 and A2a receptor, in direct and indirect pathway neurons, respectively, to have any significant downstream PKA activation. link: http://identifiers.org/pubmed/26468202

Parameters:

NameDescription
mw05f4bef4_5e8d_4a92_bb74_cc0bb4c0260e = 1.0; mw77fab49b_2ba6_4efe_9342_285f4fd3b7fa = 0.01Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw219e8fae_a38b_4620_8726_e6bd1829a351 => mwf46d3666_f0f3_4f05_9603_d7e6bb69005e, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw77fab49b_2ba6_4efe_9342_285f4fd3b7fa*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw219e8fae_a38b_4620_8726_e6bd1829a351-mw05f4bef4_5e8d_4a92_bb74_cc0bb4c0260e*mwf46d3666_f0f3_4f05_9603_d7e6bb69005e)
ModelValue_145 = 100.0; AChdip = 1.0; ModelValue_143 = 100.0; ModelValue_138 = 0.0; ModelValue_144 = 0.001Reaction: mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21 = (1-ModelValue_138)*ModelValue_143+ModelValue_138*(ModelValue_144+(ModelValue_145-ModelValue_144)*AChdip), Rate Law: missing
mw7419e1e3_b601_44a8_93ff_e5b31995791e = 0.08; mw4a930624_fcc1_4d08_8e24_9a0082418629 = 0.04Reaction: mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0 + mw06380287_79c9_4f85_aed6_fa34e7bcdff1 => mwc57c3c2e_69d5_4336_aff5_d1f429420df2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw4a930624_fcc1_4d08_8e24_9a0082418629*mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0*mw06380287_79c9_4f85_aed6_fa34e7bcdff1-mw7419e1e3_b601_44a8_93ff_e5b31995791e*mwc57c3c2e_69d5_4336_aff5_d1f429420df2)
mwf633f298_303f_46d1_b644_ae07ae366f45 = 3.0Reaction: mw6e845d87_603e_4463_874d_866f554303df => mw3d9e6efb_8e12_49c9_a87f_e067914b951d + mw9710c658_a2a1_4f49_b494_af109853f251, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwf633f298_303f_46d1_b644_ae07ae366f45*mw6e845d87_603e_4463_874d_866f554303df
mw515fcf69_b724_40d9_84ba_5f92d75ae5a7 = 1.5E-4Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw7df45520_98cc_4c0b_91a7_c6e7297de98a => mw619502c3_e319_4e29_a677_b2b5f74fc2cf, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw515fcf69_b724_40d9_84ba_5f92d75ae5a7*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw7df45520_98cc_4c0b_91a7_c6e7297de98a
mw269c014a_6379_44c3_813b_52d8145506e7 = 1.0E-4; mwc4c3d33d_b2b7_4ab2_a171_1864ea638ec0 = 0.1Reaction: mw522cacf1_5e61_4b95_8742_cf61cb824893 + mwccd3a17c_e207_4663_9b16_327b78882497 => mw3fcd1ec2_a459_49d4_89f7_361e276096d6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw269c014a_6379_44c3_813b_52d8145506e7*mw522cacf1_5e61_4b95_8742_cf61cb824893*mwccd3a17c_e207_4663_9b16_327b78882497-mwc4c3d33d_b2b7_4ab2_a171_1864ea638ec0*mw3fcd1ec2_a459_49d4_89f7_361e276096d6)
mwdb2a670f_13fb_4bda_8c72_d706c6bc37e9 = 2.8125E-5Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwed1b3928_8d78_44d1_aee7_9d11d6437cfc => mw56dff932_134c_4d88_a611_daad00623fd0, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwdb2a670f_13fb_4bda_8c72_d706c6bc37e9*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mwed1b3928_8d78_44d1_aee7_9d11d6437cfc
mw448bd49f_40ad_46c9_81f6_3494057dc37d = 0.003; mwa466eec8_9bc0_44d5_8027_d5925b378429 = 5.0Reaction: mwe2fc02e6_2684_4071_932a_f7a8bd13b2fe + mw351f6cee_3e64_4b8e_8e60_24b1aca99a92 => mw0b46978f_b522_4cde_97f0_574cd7dbbae7, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw448bd49f_40ad_46c9_81f6_3494057dc37d*mwe2fc02e6_2684_4071_932a_f7a8bd13b2fe*mw351f6cee_3e64_4b8e_8e60_24b1aca99a92-mwa466eec8_9bc0_44d5_8027_d5925b378429*mw0b46978f_b522_4cde_97f0_574cd7dbbae7)
mw0b1ccae3_37fa_4a23_a817_cd8fc458dc79 = 0.1; mw6f753a0e_a7ec_4b4b_bcfc_edb95a3f1296 = 2.0Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw3d9e6efb_8e12_49c9_a87f_e067914b951d => mw6e845d87_603e_4463_874d_866f554303df, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw0b1ccae3_37fa_4a23_a817_cd8fc458dc79*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw3d9e6efb_8e12_49c9_a87f_e067914b951d-mw6f753a0e_a7ec_4b4b_bcfc_edb95a3f1296*mw6e845d87_603e_4463_874d_866f554303df)
mw9510e553_a7fd_4c9a_b284_19b3cc01ae7d = 7.5E-5; mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0Reaction: mw7df45520_98cc_4c0b_91a7_c6e7297de98a + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw619502c3_e319_4e29_a677_b2b5f74fc2cf, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw9510e553_a7fd_4c9a_b284_19b3cc01ae7d*mw7df45520_98cc_4c0b_91a7_c6e7297de98a*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw619502c3_e319_4e29_a677_b2b5f74fc2cf)
mw009f9583_4e96_4672_ab71_0ef4b697aa6f = 6.4; mw2226fa14_2b95_45a6_8705_4b38073fc5f7 = 8.0E-4Reaction: mw522cacf1_5e61_4b95_8742_cf61cb824893 + mw1184c368_03fc_435a_9086_dc6ed3067935 => mw0459271f_3b39_40a4_948f_aed773482cfc, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw2226fa14_2b95_45a6_8705_4b38073fc5f7*mw522cacf1_5e61_4b95_8742_cf61cb824893*mw1184c368_03fc_435a_9086_dc6ed3067935-mw009f9583_4e96_4672_ab71_0ef4b697aa6f*mw0459271f_3b39_40a4_948f_aed773482cfc)
mw1db20a7e_3972_4c3a_83c0_c6fcd7c9cb45 = 3.0E-4; mw4e2575eb_3641_422c_b836_d854958d4d1e = 8.0Reaction: mw68d3f409_9462_4515_8c07_bc105fa0eaf1 + mw24435476_9c30_4878_b26f_4b3c5a0685c6 => mw4179e1ff_9035_4c67_a67c_099e25beb9b0, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw1db20a7e_3972_4c3a_83c0_c6fcd7c9cb45*mw68d3f409_9462_4515_8c07_bc105fa0eaf1*mw24435476_9c30_4878_b26f_4b3c5a0685c6-mw4e2575eb_3641_422c_b836_d854958d4d1e*mw4179e1ff_9035_4c67_a67c_099e25beb9b0)
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mw00f3118f_5d5a_48d0_bcc4_749d5f9dc73a = 1.75E-4Reaction: mw2badefa3_32e8_4b66_9e69_245d9ec74e33 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw07c7392b_8d89_4b94_97c5_59f7e256b6f2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw00f3118f_5d5a_48d0_bcc4_749d5f9dc73a*mw2badefa3_32e8_4b66_9e69_245d9ec74e33*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw07c7392b_8d89_4b94_97c5_59f7e256b6f2)
mwfcfb91ff_a495_41f9_bdff_fcef779112fd = 30.0Reaction: mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 => mw9bcba6bc_9788_4f7f_afb5_1c8f3b33c3d1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwfcfb91ff_a495_41f9_bdff_fcef779112fd*mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3
mwa390f769_ebf1_4023_8af0_1c00e2a9bf82 = 0.002; mw0beb6cc4_36bd_4022_8993_29f981652ebe = 1.0Reaction: mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0 + mw4855b1cd_d7bc_4072_9736_dca30bbe448d => mwcf1bb70c_9d0b_4e82_b58a_6f8e73208af9, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwa390f769_ebf1_4023_8af0_1c00e2a9bf82*mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0*mw4855b1cd_d7bc_4072_9736_dca30bbe448d-mw0beb6cc4_36bd_4022_8993_29f981652ebe*mwcf1bb70c_9d0b_4e82_b58a_6f8e73208af9)
mw5301f7f5_60df_4eb9_ba3b_81e6519d1cbb = 5.0Reaction: mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwd794c746_c826_4ba1_9e09_a9d1e122d925, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw5301f7f5_60df_4eb9_ba3b_81e6519d1cbb*mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e
mwefa9bb47_f13f_4a21_a62d_a4debcf7b52b = 90.0; mw066c69e2_66da_4621_9180_bce71b7077c3 = 1.0Reaction: mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21 + mwd86ce0dc_7329_4b27_9de0_ee6bffee3083 => mwe4e36b8e_18b8_4c76_bd46_13614b71da5c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw066c69e2_66da_4621_9180_bce71b7077c3*mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21*mwd86ce0dc_7329_4b27_9de0_ee6bffee3083-mwefa9bb47_f13f_4a21_a62d_a4debcf7b52b*mwe4e36b8e_18b8_4c76_bd46_13614b71da5c)
mwdcc4ce84_732d_4f5b_84e2_e5b93617200b = 0.3Reaction: mw8825a609_0983_4fb4_a264_e2f7e43d17b3 => mw3fcd1ec2_a459_49d4_89f7_361e276096d6 + mw24435476_9c30_4878_b26f_4b3c5a0685c6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwdcc4ce84_732d_4f5b_84e2_e5b93617200b*mw8825a609_0983_4fb4_a264_e2f7e43d17b3
mw2561b5ab_39c9_4453_99d8_f0f37779b63a = 10.0Reaction: mw4179e1ff_9035_4c67_a67c_099e25beb9b0 => mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0 + mw68d3f409_9462_4515_8c07_bc105fa0eaf1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw2561b5ab_39c9_4453_99d8_f0f37779b63a*mw4179e1ff_9035_4c67_a67c_099e25beb9b0
mwca52f04a_bb5f_4d3f_ba6d_939bbb3895b9 = 2.0; mw326e0065_b4f6_41ae_b1d0_66092dc5ebb2 = 0.13Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw1041345b_f015_436c_9eff_98211008aa1c => mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw326e0065_b4f6_41ae_b1d0_66092dc5ebb2*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw1041345b_f015_436c_9eff_98211008aa1c-mwca52f04a_bb5f_4d3f_ba6d_939bbb3895b9*mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43)
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mw72ceb3da_d538_4f25_8e69_f322eb0b5e57 = 0.00105Reaction: mwfe9ed415_d5af_469c_a549_d8981f1eb01f + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw166e3335_56c3_41ef_af0f_b583860991c1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw72ceb3da_d538_4f25_8e69_f322eb0b5e57*mwfe9ed415_d5af_469c_a549_d8981f1eb01f*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw166e3335_56c3_41ef_af0f_b583860991c1)
mw88c9326a_fbe9_4dd8_aded_b5be3f012691 = 2.3Reaction: mwde741b91_d5bf_44a9_ad45_404d7259d051 => mw081c9f7b_011e_440f_971d_d0316d2a1e6c + mw24435476_9c30_4878_b26f_4b3c5a0685c6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw88c9326a_fbe9_4dd8_aded_b5be3f012691*mwde741b91_d5bf_44a9_ad45_404d7259d051
mw5175a06e_3927_4993_9242_8f76b0aaf42f = 100.0Reaction: mwb80e4fa1_4849_4ed5_b3b0_3e3025c61ad8 + mw9bcba6bc_9788_4f7f_afb5_1c8f3b33c3d1 => mwd8ea533a_c66e_4de4_8c5c_0d4201d8c8a2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw5175a06e_3927_4993_9242_8f76b0aaf42f*mwb80e4fa1_4849_4ed5_b3b0_3e3025c61ad8*mw9bcba6bc_9788_4f7f_afb5_1c8f3b33c3d1
mw65cae8fe_0eac_4792_88bf_2dfb441030e5 = 0.5Reaction: mw619502c3_e319_4e29_a677_b2b5f74fc2cf => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw7df45520_98cc_4c0b_91a7_c6e7297de98a, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw65cae8fe_0eac_4792_88bf_2dfb441030e5*mw619502c3_e319_4e29_a677_b2b5f74fc2cf
mwac1bc66c_2623_47e6_a76d_c1629d962be5 = 10.0Reaction: mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43 => mw1041345b_f015_436c_9eff_98211008aa1c + mw9710c658_a2a1_4f49_b494_af109853f251, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwac1bc66c_2623_47e6_a76d_c1629d962be5*mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43
mw6af7af00_75ac_4f58_8383_7047a5fb5181 = 1.0Reaction: mw0459271f_3b39_40a4_948f_aed773482cfc => mw522cacf1_5e61_4b95_8742_cf61cb824893 + mw24435476_9c30_4878_b26f_4b3c5a0685c6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw6af7af00_75ac_4f58_8383_7047a5fb5181*mw0459271f_3b39_40a4_948f_aed773482cfc
mwe737a297_e5be_46ed_af75_ccc7428c3977 = 0.001; mwddcb8d81_9f5a_457e_a54c_a0c1b1f29f0b = 0.9Reaction: mwccd3a17c_e207_4663_9b16_327b78882497 + mw7086a13a_619e_4069_b163_d8a05fc55f42 => mw619502c3_e319_4e29_a677_b2b5f74fc2cf, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwe737a297_e5be_46ed_af75_ccc7428c3977*mwccd3a17c_e207_4663_9b16_327b78882497*mw7086a13a_619e_4069_b163_d8a05fc55f42-mwddcb8d81_9f5a_457e_a54c_a0c1b1f29f0b*mw619502c3_e319_4e29_a677_b2b5f74fc2cf)
mw8e4e88b6_60b3_43bd_8f5c_923712ee64ea = 5.0; mwb17941e5_1ad5_42b9_98c6_e62b1a697dbb = 0.003Reaction: mwdb9dc389_2bf0_4039_9f09_282f5511958b + mw351f6cee_3e64_4b8e_8e60_24b1aca99a92 => mw6b2f1c44_e0be_4406_bcef_ad5061d519e4, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwb17941e5_1ad5_42b9_98c6_e62b1a697dbb*mwdb9dc389_2bf0_4039_9f09_282f5511958b*mw351f6cee_3e64_4b8e_8e60_24b1aca99a92-mw8e4e88b6_60b3_43bd_8f5c_923712ee64ea*mw6b2f1c44_e0be_4406_bcef_ad5061d519e4)
mwcd307ee9_33da_4303_9c28_644ad2d1630c = 0.1; mw0a255671_d9ca_4384_a153_ce17e1111453 = 0.2Reaction: mw724f1afe_8032_40ae_96ca_808ab7b8b943 + mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c => mwfe9ed415_d5af_469c_a549_d8981f1eb01f, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw0a255671_d9ca_4384_a153_ce17e1111453*mw724f1afe_8032_40ae_96ca_808ab7b8b943*mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c-mwcd307ee9_33da_4303_9c28_644ad2d1630c*mwfe9ed415_d5af_469c_a549_d8981f1eb01f)
mw0dd72d64_80e1_4f76_a444_fd175dbfab0c = 15.0Reaction: mw6b2f1c44_e0be_4406_bcef_ad5061d519e4 => mwaf471bc1_f98a_4115_b0ee_45c189ea20b5 + mwdb9dc389_2bf0_4039_9f09_282f5511958b + mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw0dd72d64_80e1_4f76_a444_fd175dbfab0c*mw6b2f1c44_e0be_4406_bcef_ad5061d519e4
mwaa3af366_350e_4f18_936b_6372dc484f82 = 4.0E-4Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw724f1afe_8032_40ae_96ca_808ab7b8b943 => mw7086a13a_619e_4069_b163_d8a05fc55f42, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwaa3af366_350e_4f18_936b_6372dc484f82*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw724f1afe_8032_40ae_96ca_808ab7b8b943
mwd1b16e73_4fcb_4e4c_9804_3137259ba749 = 1.0E-6; mw36cb62c6_0b3c_4d1b_9001_3b37aa7477e2 = 9.0Reaction: mw3d9e6efb_8e12_49c9_a87f_e067914b951d + mw1c97b02d_169a_4eb8_bc84_1be57c51a255 => mw1041345b_f015_436c_9eff_98211008aa1c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwd1b16e73_4fcb_4e4c_9804_3137259ba749*mw3d9e6efb_8e12_49c9_a87f_e067914b951d*mw1c97b02d_169a_4eb8_bc84_1be57c51a255^2-mw36cb62c6_0b3c_4d1b_9001_3b37aa7477e2*mw1041345b_f015_436c_9eff_98211008aa1c)
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mwb56b5ab7_47cc_4fbc_b68b_dfdc6be6d7a4 = 5.5E-4Reaction: mw42919ead_5972_4151_85ac_fcc88ca105a6 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwb56b5ab7_47cc_4fbc_b68b_dfdc6be6d7a4*mw42919ead_5972_4151_85ac_fcc88ca105a6*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba)
mwb0a6bd5e_87a0_425c_a5c7_ea69903e0bf3 = 10.0Reaction: mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw42919ead_5972_4151_85ac_fcc88ca105a6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwb0a6bd5e_87a0_425c_a5c7_ea69903e0bf3*mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba
mw649b47b3_4c3a_4ac9_ae94_5c38ccf81e39 = 0.002; mwc911f28c_b62f_4269_84ed_d852f6da24f9 = 0.01Reaction: mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c + mw56dff932_134c_4d88_a611_daad00623fd0 => mw07c7392b_8d89_4b94_97c5_59f7e256b6f2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw649b47b3_4c3a_4ac9_ae94_5c38ccf81e39*mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c*mw56dff932_134c_4d88_a611_daad00623fd0-mwc911f28c_b62f_4269_84ed_d852f6da24f9*mw07c7392b_8d89_4b94_97c5_59f7e256b6f2)
mw5623544e_e7e1_439f_88d3_3b0cbea8ccf5 = 30.0Reaction: mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c => mwfed0682b_39f1_4b09_94e8_c45a51744092, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw5623544e_e7e1_439f_88d3_3b0cbea8ccf5*mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c
mwe79f507b_73c9_4056_ae91_6244dcbc49bb = 0.5; mw26206710_ba98_4010_9e5b_c3aae2ce29ec = 1.0Reaction: mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0 + mw3fcd1ec2_a459_49d4_89f7_361e276096d6 => mw8825a609_0983_4fb4_a264_e2f7e43d17b3, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwe79f507b_73c9_4056_ae91_6244dcbc49bb*mw2f3e9c55_e57f_416e_b4b1_cc49a26192c0*mw3fcd1ec2_a459_49d4_89f7_361e276096d6-mw26206710_ba98_4010_9e5b_c3aae2ce29ec*mw8825a609_0983_4fb4_a264_e2f7e43d17b3)
mw62c51fcf_c107_4d3c_849e_9b168df54490 = 10.0Reaction: mwcf1bb70c_9d0b_4e82_b58a_6f8e73208af9 => mw24435476_9c30_4878_b26f_4b3c5a0685c6 + mw4855b1cd_d7bc_4072_9736_dca30bbe448d, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw62c51fcf_c107_4d3c_849e_9b168df54490*mwcf1bb70c_9d0b_4e82_b58a_6f8e73208af9
mwce0df80f_1563_453d_b33d_a88f6b2c93b7 = 90.0; mw80292f32_fd53_4b5d_872a_e21c2d90c52a = 0.01Reaction: mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21 + mwf82770b9_766a_4c4e_851a_d76da19e8517 => mw9d5c5c9d_301d_4e43_ba7b_7d21ccbdc2c2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw80292f32_fd53_4b5d_872a_e21c2d90c52a*mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21*mwf82770b9_766a_4c4e_851a_d76da19e8517-mwce0df80f_1563_453d_b33d_a88f6b2c93b7*mw9d5c5c9d_301d_4e43_ba7b_7d21ccbdc2c2)
mwa4148cd1_a298_447c_aea8_226688c3f526 = 2.0Reaction: mwf46d3666_f0f3_4f05_9603_d7e6bb69005e => mw219e8fae_a38b_4620_8726_e6bd1829a351 + mw9710c658_a2a1_4f49_b494_af109853f251, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwa4148cd1_a298_447c_aea8_226688c3f526*mwf46d3666_f0f3_4f05_9603_d7e6bb69005e
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mwb93138ce_a80b_4b26_b927_6b4a00651b64 = 3.0E-4Reaction: mwd794c746_c826_4ba1_9e09_a9d1e122d925 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwb93138ce_a80b_4b26_b927_6b4a00651b64*mwd794c746_c826_4ba1_9e09_a9d1e122d925*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e)
mw858f28f3_086a_436b_ba23_4fc7372c8884 = 5.0; mw3fc2c1ed_0097_4f7f_bcd5_904dc6ad5a56 = 0.005Reaction: mw0b46978f_b522_4cde_97f0_574cd7dbbae7 + mwbe974953_e869_4622_b4a8_745555c8d7fd => mw6b2f1c44_e0be_4406_bcef_ad5061d519e4, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw3fc2c1ed_0097_4f7f_bcd5_904dc6ad5a56*mw0b46978f_b522_4cde_97f0_574cd7dbbae7*mwbe974953_e869_4622_b4a8_745555c8d7fd-mw858f28f3_086a_436b_ba23_4fc7372c8884*mw6b2f1c44_e0be_4406_bcef_ad5061d519e4)
mwcabc0868_2435_4850_964b_e3ddee39f5ad = 30.0Reaction: mw07c7392b_8d89_4b94_97c5_59f7e256b6f2 => mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba + mw9bcba6bc_9788_4f7f_afb5_1c8f3b33c3d1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwcabc0868_2435_4850_964b_e3ddee39f5ad*mw07c7392b_8d89_4b94_97c5_59f7e256b6f2
mw6ae3f7a6_bf58_475e_930e_6bf7a79f3761 = 5.0; mw1ef56a9a_9d9b_4490_8fcd_53b7e50bf5d6 = 50.0Reaction: mw7086a13a_619e_4069_b163_d8a05fc55f42 + mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 => mw2075d2cf_955e_4150_98b8_847103c53845, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw1ef56a9a_9d9b_4490_8fcd_53b7e50bf5d6*mw7086a13a_619e_4069_b163_d8a05fc55f42*mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3-mw6ae3f7a6_bf58_475e_930e_6bf7a79f3761*mw2075d2cf_955e_4150_98b8_847103c53845)
mwd05b4199_53ad_4807_9a8c_d93ce35be857 = 60.0Reaction: mwe4e36b8e_18b8_4c76_bd46_13614b71da5c => mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 + mw9d5c5c9d_301d_4e43_ba7b_7d21ccbdc2c2 + mwb80e4fa1_4849_4ed5_b3b0_3e3025c61ad8, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwd05b4199_53ad_4807_9a8c_d93ce35be857*mwe4e36b8e_18b8_4c76_bd46_13614b71da5c
mw9c2302f8_3d47_4247_a338_a02c53fc5331 = 1.0E-4; mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0Reaction: mw724f1afe_8032_40ae_96ca_808ab7b8b943 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw7086a13a_619e_4069_b163_d8a05fc55f42, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw9c2302f8_3d47_4247_a338_a02c53fc5331*mw724f1afe_8032_40ae_96ca_808ab7b8b943*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw7086a13a_619e_4069_b163_d8a05fc55f42)
mwc728d91d_7616_43db_bd1d_55e49e9c026a = 0.125Reaction: mw56dff932_134c_4d88_a611_daad00623fd0 => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwed1b3928_8d78_44d1_aee7_9d11d6437cfc, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwc728d91d_7616_43db_bd1d_55e49e9c026a*mw56dff932_134c_4d88_a611_daad00623fd0
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mwa1bc2233_5bb9_4135_88ed_bb51640faec8 = 5.625E-5Reaction: mwed1b3928_8d78_44d1_aee7_9d11d6437cfc + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw56dff932_134c_4d88_a611_daad00623fd0, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwa1bc2233_5bb9_4135_88ed_bb51640faec8*mwed1b3928_8d78_44d1_aee7_9d11d6437cfc*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw56dff932_134c_4d88_a611_daad00623fd0)
mw8db06baf_d8bb_4a1a_b415_2d51fa1e53ba = 0.2Reaction: mw166e3335_56c3_41ef_af0f_b583860991c1 => mw7086a13a_619e_4069_b163_d8a05fc55f42 + mwfed0682b_39f1_4b09_94e8_c45a51744092, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw8db06baf_d8bb_4a1a_b415_2d51fa1e53ba*mw166e3335_56c3_41ef_af0f_b583860991c1
mwc52aebc2_571c_4f96_84ee_0613ae73db89 = 0.01; mw9330e49a_b214_4807_b614_4241a4a12c43 = 0.01Reaction: mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 + mwfe9ed415_d5af_469c_a549_d8981f1eb01f => mwd794c746_c826_4ba1_9e09_a9d1e122d925, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwc52aebc2_571c_4f96_84ee_0613ae73db89*mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3*mwfe9ed415_d5af_469c_a549_d8981f1eb01f-mw9330e49a_b214_4807_b614_4241a4a12c43*mwd794c746_c826_4ba1_9e09_a9d1e122d925)
mw1a6a8649_d7cb_4379_983a_cca2acac3112 = 2.5Reaction: mw07c7392b_8d89_4b94_97c5_59f7e256b6f2 => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw2badefa3_32e8_4b66_9e69_245d9ec74e33, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw1a6a8649_d7cb_4379_983a_cca2acac3112*mw07c7392b_8d89_4b94_97c5_59f7e256b6f2
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mw541807fb_7d9f_4788_9f21_cc62846b5826 = 6.25E-5Reaction: mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a + mw29ba9e7c_6865_4817_8775_be2dbc29651e => mw2075d2cf_955e_4150_98b8_847103c53845, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw541807fb_7d9f_4788_9f21_cc62846b5826*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a*mw29ba9e7c_6865_4817_8775_be2dbc29651e-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw2075d2cf_955e_4150_98b8_847103c53845)

States:

NameDescription
mw8825a609 0983 4fb4 a264 e2f7e43d17b3[calcium(2+); Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; Protein phosphatase 1 regulatory subunit 1B]
mwf46d3666 f0f3 4f05 9603 d7e6bb69005e[3',5'-cyclic AMP; cAMP-specific 3',5'-cyclic phosphodiesterase 4D]
mw8e34c23f 1891 4dc9 8f97 dc2f12a1706c[GTP; Guanine nucleotide-binding protein G(olf) subunit alpha]
mw1041345b f015 436c 9eff 98211008aa1c[cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mw2f3e9c55 e57f 416e b4b1 cc49a26192c0[Protein phosphatase 1 regulatory subunit 1B]
mw46dccec6 6f0f 40f6 a10c 2f34ae7a005a[ATP]
mw0b46978f b522 4cde 97f0 574cd7dbbae7[D(1A) dopamine receptor; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
mwb80e4fa1 4849 4ed5 b3b0 3e3025c61ad8[Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
mw3e1a2fbf 37b1 490c 9528 6cb6bbf11b21[acetylcholine]
mw24435476 9c30 4878 b26f 4b3c5a0685c6[Protein phosphatase 1 regulatory subunit 1B]
mwfed0682b 39f1 4b09 94e8 c45a51744092[GDP; Guanine nucleotide-binding protein G(olf) subunit alpha]
mw3d9e6efb 8e12 49c9 a87f e067914b951d[cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mw07c7392b 8d89 4b94 97c5 59f7e256b6f2[calcium(2+); GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw7086a13a 619e 4069 b163 d8a05fc55f42[ATP; Adenylate cyclase type 5]
mwaf471bc1 f98a 4115 b0ee 45c189ea20b5[Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1]
mw619502c3 e319 4e29 a677 b2b5f74fc2cf[calcium(2+); ATP; Adenylate cyclase type 5]
mw56dff932 134c 4d88 a611 daad00623fd0[calcium(2+); GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw351f6cee 3e64 4b8e 8e60 24b1aca99a92[Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
mwd794c746 c826 4ba1 9e09 a9d1e122d925[GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw0a10f9cb 3f4b 4bfa ace9 0ecd2bd74b5e[GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mwa2c44a01 28c9 4dbd b034 364f9b5b6cc3[GTP; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw9bcba6bc 9788 4f7f afb5 1c8f3b33c3d1[GDP; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw6e845d87 603e 4463 874d 866f554303df[3',5'-cyclic AMP; cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mw081c9f7b 011e 440f 971d d0316d2a1e6c[Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
mwbae3bd11 0ab4 4587 a931 9c5dc5e777ba[calcium(2+); GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha]
mwde741b91 d5bf 44a9 ad45 404d7259d051[Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform; Protein phosphatase 1 regulatory subunit 1B]
mwe4e36b8e 18b8 4c76 bd46 13614b71da5c[acetylcholine; Muscarinic acetylcholine receptor M4; Guanine nucleotide-binding protein G(i) subunit alpha-1; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
mw6b2f1c44 e0be 4406 bcef ad5061d519e4[dopamine; D(1A) dopamine receptor; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
totalActivePKA[3',5'-cyclic AMP; cAMP-dependent protein kinase catalytic subunit alpha; cAMP-dependent protein kinase catalytic subunit beta; cAMP-dependent protein kinase type I-alpha regulatory subunit; cAMP-dependent protein kinase type I-beta regulatory subunit]
mw1f3b8982 3b8c 42b6 8b0f 49b037cbda43[3',5'-cyclic AMP; cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mw522cacf1 5e61 4b95 8742 cf61cb824893[IPR006186; Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
mw4179e1ff 9035 4c67 a67c 099e25beb9b0[cAMP-dependent protein kinase catalytic subunit alpha; cAMP-dependent protein kinase catalytic subunit beta; Protein phosphatase 1 regulatory subunit 1B]

Nair2015 - Interaction between neuromodulators via GPCRs - Effect on cAMP/PKA signaling (D2 Neuron): BIOMD0000000636v0.0.1

Nair2015 - Interaction between neuromodulators via GPCRs - Effect on cAMP/PKA signaling (D2 Neuron)This model is describ…

Details

Transient changes in striatal dopamine (DA) concentration are considered to encode a reward prediction error (RPE) in reinforcement learning tasks. Often, a phasic DA change occurs concomitantly with a dip in striatal acetylcholine (ACh), whereas other neuromodulators, such as adenosine (Adn), change slowly. There are abundant adenylyl cyclase (AC) coupled GPCRs for these neuromodulators in striatal medium spiny neurons (MSNs), which play important roles in plasticity. However, little is known about the interaction between these neuromodulators via GPCRs. The interaction between these transient neuromodulator changes and the effect on cAMP/PKA signaling via Golf- and Gi/o-coupled GPCR are studied here using quantitative kinetic modeling. The simulations suggest that, under basal conditions, cAMP/PKA signaling could be significantly inhibited in D1R+ MSNs via ACh/M4R/Gi/o and an ACh dip is required to gate a subset of D1R/Golf-dependent PKA activation. Furthermore, the interaction between ACh dip and DA peak, via D1R and M4R, is synergistic. In a similar fashion, PKA signaling in D2+ MSNs is under basal inhibition via D2R/Gi/o and a DA dip leads to a PKA increase by disinhibiting A2aR/Golf, but D2+ MSNs could also respond to the DA peak via other intracellular pathways. This study highlights the similarity between the two types of MSNs in terms of high basal AC inhibition by Gi/o and the importance of interactions between Gi/o and Golf signaling, but at the same time predicts differences between them with regard to the sign of RPE responsible for PKA activation.Dopamine transients are considered to carry reward-related signal in reinforcement learning. An increase in dopamine concentration is associated with an unexpected reward or salient stimuli, whereas a decrease is produced by omission of an expected reward. Often dopamine transients are accompanied by other neuromodulatory signals, such as acetylcholine and adenosine. We highlight the importance of interaction between acetylcholine, dopamine, and adenosine signals via adenylyl-cyclase coupled GPCRs in shaping the dopamine-dependent cAMP/PKA signaling in striatal neurons. Specifically, a dopamine peak and an acetylcholine dip must interact, via D1 and M4 receptor, and a dopamine dip must interact with adenosine tone, via D2 and A2a receptor, in direct and indirect pathway neurons, respectively, to have any significant downstream PKA activation. link: http://identifiers.org/pubmed/26468202

Parameters:

NameDescription
mwf633f298_303f_46d1_b644_ae07ae366f45 = 3.0Reaction: mw6e845d87_603e_4463_874d_866f554303df => mw3d9e6efb_8e12_49c9_a87f_e067914b951d + mw9710c658_a2a1_4f49_b494_af109853f251, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwf633f298_303f_46d1_b644_ae07ae366f45*mw6e845d87_603e_4463_874d_866f554303df
mwdad9965c_2334_481f_8544_f1a81385a28e = 0.005; mwc23d8bf6_2a60_4760_8bf5_c1bab432ab52 = 1.0Reaction: A2AR + mwbe974953_e869_4622_b4a8_745555c8d7fd => A2ARAdn, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwdad9965c_2334_481f_8544_f1a81385a28e*A2AR*mwbe974953_e869_4622_b4a8_745555c8d7fd-mwc23d8bf6_2a60_4760_8bf5_c1bab432ab52*A2ARAdn)
mw515fcf69_b724_40d9_84ba_5f92d75ae5a7 = 1.5E-4Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw7df45520_98cc_4c0b_91a7_c6e7297de98a => mw619502c3_e319_4e29_a677_b2b5f74fc2cf, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw515fcf69_b724_40d9_84ba_5f92d75ae5a7*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw7df45520_98cc_4c0b_91a7_c6e7297de98a
mw80292f32_fd53_4b5d_872a_e21c2d90c52a = 0.1; mwce0df80f_1563_453d_b33d_a88f6b2c93b7 = 200.0Reaction: mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21 + mwf82770b9_766a_4c4e_851a_d76da19e8517 => mw9d5c5c9d_301d_4e43_ba7b_7d21ccbdc2c2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw80292f32_fd53_4b5d_872a_e21c2d90c52a*mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21*mwf82770b9_766a_4c4e_851a_d76da19e8517-mwce0df80f_1563_453d_b33d_a88f6b2c93b7*mw9d5c5c9d_301d_4e43_ba7b_7d21ccbdc2c2)
mwfe873584_629a_46c8_aae9_fdacdb9ad266 = 0.1; mwf3c85708_890c_45d1_bcbc_fe90e9ca792f = 10.0Reaction: mwd1171b65_ed6c_4413_bf47_5ed80038a7bd + mwccd3a17c_e207_4663_9b16_327b78882497 => mw4855b1cd_d7bc_4072_9736_dca30bbe448d, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwfe873584_629a_46c8_aae9_fdacdb9ad266*mwd1171b65_ed6c_4413_bf47_5ed80038a7bd*mwccd3a17c_e207_4663_9b16_327b78882497-mwf3c85708_890c_45d1_bcbc_fe90e9ca792f*mw4855b1cd_d7bc_4072_9736_dca30bbe448d)
mwdb2a670f_13fb_4bda_8c72_d706c6bc37e9 = 2.8125E-5Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwed1b3928_8d78_44d1_aee7_9d11d6437cfc => mw56dff932_134c_4d88_a611_daad00623fd0, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwdb2a670f_13fb_4bda_8c72_d706c6bc37e9*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mwed1b3928_8d78_44d1_aee7_9d11d6437cfc
mw0b1ccae3_37fa_4a23_a817_cd8fc458dc79 = 0.1; mw6f753a0e_a7ec_4b4b_bcfc_edb95a3f1296 = 2.0Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw3d9e6efb_8e12_49c9_a87f_e067914b951d => mw6e845d87_603e_4463_874d_866f554303df, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw0b1ccae3_37fa_4a23_a817_cd8fc458dc79*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw3d9e6efb_8e12_49c9_a87f_e067914b951d-mw6f753a0e_a7ec_4b4b_bcfc_edb95a3f1296*mw6e845d87_603e_4463_874d_866f554303df)
mw9510e553_a7fd_4c9a_b284_19b3cc01ae7d = 7.5E-5; mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0Reaction: mw7df45520_98cc_4c0b_91a7_c6e7297de98a + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw619502c3_e319_4e29_a677_b2b5f74fc2cf, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw9510e553_a7fd_4c9a_b284_19b3cc01ae7d*mw7df45520_98cc_4c0b_91a7_c6e7297de98a*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw619502c3_e319_4e29_a677_b2b5f74fc2cf)
mw009f9583_4e96_4672_ab71_0ef4b697aa6f = 6.4; mw2226fa14_2b95_45a6_8705_4b38073fc5f7 = 8.0E-4Reaction: mw522cacf1_5e61_4b95_8742_cf61cb824893 + mw1184c368_03fc_435a_9086_dc6ed3067935 => mw0459271f_3b39_40a4_948f_aed773482cfc, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw2226fa14_2b95_45a6_8705_4b38073fc5f7*mw522cacf1_5e61_4b95_8742_cf61cb824893*mw1184c368_03fc_435a_9086_dc6ed3067935-mw009f9583_4e96_4672_ab71_0ef4b697aa6f*mw0459271f_3b39_40a4_948f_aed773482cfc)
mw066c69e2_66da_4621_9180_bce71b7077c3 = 12.0; mwefa9bb47_f13f_4a21_a62d_a4debcf7b52b = 200.0Reaction: mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21 + mwd86ce0dc_7329_4b27_9de0_ee6bffee3083 => mwe4e36b8e_18b8_4c76_bd46_13614b71da5c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw066c69e2_66da_4621_9180_bce71b7077c3*mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21*mwd86ce0dc_7329_4b27_9de0_ee6bffee3083-mwefa9bb47_f13f_4a21_a62d_a4debcf7b52b*mwe4e36b8e_18b8_4c76_bd46_13614b71da5c)
mw1b9e5266_efac_4696_a213_80f9f83d948a = 9.0E-4; mwa27c20d8_b6ed_4617_a6f6_9af2752d3a33 = 2.0Reaction: mw32351ce4_eaaf_4827_8efa_342224548d8a + mw24435476_9c30_4878_b26f_4b3c5a0685c6 => mw0130a500_18e9_470f_9fac_70af44dc4a9e, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw1b9e5266_efac_4696_a213_80f9f83d948a*mw32351ce4_eaaf_4827_8efa_342224548d8a*mw24435476_9c30_4878_b26f_4b3c5a0685c6-mwa27c20d8_b6ed_4617_a6f6_9af2752d3a33*mw0130a500_18e9_470f_9fac_70af44dc4a9e)
ModelValue_131 = 0.0; ModelValue_124 = 0.0; DAdip = 10.0; DApeak = 10.0; ModelValue_138 = 10.0Reaction: mw3e1a2fbf_37b1_490c_9528_6cb6bbf11b21 = ((1-ModelValue_124)-ModelValue_131)*ModelValue_138+ModelValue_124*DAdip+ModelValue_131*DApeak, Rate Law: missing
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mw00f3118f_5d5a_48d0_bcc4_749d5f9dc73a = 1.75E-4Reaction: mw2badefa3_32e8_4b66_9e69_245d9ec74e33 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw07c7392b_8d89_4b94_97c5_59f7e256b6f2, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw00f3118f_5d5a_48d0_bcc4_749d5f9dc73a*mw2badefa3_32e8_4b66_9e69_245d9ec74e33*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw07c7392b_8d89_4b94_97c5_59f7e256b6f2)
mw0dd72d64_80e1_4f76_a444_fd175dbfab0c = 30.0Reaction: A2ARAdnGolf => mwaf471bc1_f98a_4115_b0ee_45c189ea20b5 + A2ARAdn + mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw0dd72d64_80e1_4f76_a444_fd175dbfab0c*A2ARAdnGolf
mwfcfb91ff_a495_41f9_bdff_fcef779112fd = 30.0Reaction: mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 => mw9bcba6bc_9788_4f7f_afb5_1c8f3b33c3d1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwfcfb91ff_a495_41f9_bdff_fcef779112fd*mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3
mw5301f7f5_60df_4eb9_ba3b_81e6519d1cbb = 5.0Reaction: mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwd794c746_c826_4ba1_9e09_a9d1e122d925, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw5301f7f5_60df_4eb9_ba3b_81e6519d1cbb*mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e
mw034d8151_fae1_4738_b675_39c38a58118d = 0.022Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw42919ead_5972_4151_85ac_fcc88ca105a6 => mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw034d8151_fae1_4738_b675_39c38a58118d*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw42919ead_5972_4151_85ac_fcc88ca105a6
mwca52f04a_bb5f_4d3f_ba6d_939bbb3895b9 = 2.0; mw326e0065_b4f6_41ae_b1d0_66092dc5ebb2 = 0.13Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw1041345b_f015_436c_9eff_98211008aa1c => mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw326e0065_b4f6_41ae_b1d0_66092dc5ebb2*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw1041345b_f015_436c_9eff_98211008aa1c-mwca52f04a_bb5f_4d3f_ba6d_939bbb3895b9*mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43)
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mw72ceb3da_d538_4f25_8e69_f322eb0b5e57 = 0.00105Reaction: mwfe9ed415_d5af_469c_a549_d8981f1eb01f + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw166e3335_56c3_41ef_af0f_b583860991c1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw72ceb3da_d538_4f25_8e69_f322eb0b5e57*mwfe9ed415_d5af_469c_a549_d8981f1eb01f*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw166e3335_56c3_41ef_af0f_b583860991c1)
mw88c9326a_fbe9_4dd8_aded_b5be3f012691 = 2.3Reaction: mwde741b91_d5bf_44a9_ad45_404d7259d051 => mw081c9f7b_011e_440f_971d_d0316d2a1e6c + mw24435476_9c30_4878_b26f_4b3c5a0685c6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw88c9326a_fbe9_4dd8_aded_b5be3f012691*mwde741b91_d5bf_44a9_ad45_404d7259d051
mw65cae8fe_0eac_4792_88bf_2dfb441030e5 = 0.5Reaction: mw619502c3_e319_4e29_a677_b2b5f74fc2cf => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw7df45520_98cc_4c0b_91a7_c6e7297de98a, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw65cae8fe_0eac_4792_88bf_2dfb441030e5*mw619502c3_e319_4e29_a677_b2b5f74fc2cf
mwac1bc66c_2623_47e6_a76d_c1629d962be5 = 10.0Reaction: mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43 => mw1041345b_f015_436c_9eff_98211008aa1c + mw9710c658_a2a1_4f49_b494_af109853f251, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwac1bc66c_2623_47e6_a76d_c1629d962be5*mw1f3b8982_3b8c_42b6_8b0f_49b037cbda43
mwe737a297_e5be_46ed_af75_ccc7428c3977 = 0.001; mwddcb8d81_9f5a_457e_a54c_a0c1b1f29f0b = 0.9Reaction: mw724f1afe_8032_40ae_96ca_808ab7b8b943 + mwccd3a17c_e207_4663_9b16_327b78882497 => mw7df45520_98cc_4c0b_91a7_c6e7297de98a, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwe737a297_e5be_46ed_af75_ccc7428c3977*mw724f1afe_8032_40ae_96ca_808ab7b8b943*mwccd3a17c_e207_4663_9b16_327b78882497-mwddcb8d81_9f5a_457e_a54c_a0c1b1f29f0b*mw7df45520_98cc_4c0b_91a7_c6e7297de98a)
mw2f090a45_946b_4587_a3e3_b29f3bb5c6ae = 100.0Reaction: mwfed0682b_39f1_4b09_94e8_c45a51744092 + mwaf471bc1_f98a_4115_b0ee_45c189ea20b5 => mw351f6cee_3e64_4b8e_8e60_24b1aca99a92, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw2f090a45_946b_4587_a3e3_b29f3bb5c6ae*mwfed0682b_39f1_4b09_94e8_c45a51744092*mwaf471bc1_f98a_4115_b0ee_45c189ea20b5
mwcd307ee9_33da_4303_9c28_644ad2d1630c = 0.1; mw0a255671_d9ca_4384_a153_ce17e1111453 = 0.2Reaction: mw724f1afe_8032_40ae_96ca_808ab7b8b943 + mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c => mwfe9ed415_d5af_469c_a549_d8981f1eb01f, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw0a255671_d9ca_4384_a153_ce17e1111453*mw724f1afe_8032_40ae_96ca_808ab7b8b943*mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c-mwcd307ee9_33da_4303_9c28_644ad2d1630c*mwfe9ed415_d5af_469c_a549_d8981f1eb01f)
mw68039b16_b516_4fba_bedd_d4bbc1a23a02 = 9.1; mw894b221b_266d_4277_ac01_83579ed103e6 = 0.006Reaction: mw4b358131_010c_4545_ac4a_13a6c8bc34c4 + mwccd3a17c_e207_4663_9b16_327b78882497 => mw65a14789_ffcf_4bfd_9d53_d2eb2f4d0896, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw894b221b_266d_4277_ac01_83579ed103e6*mw4b358131_010c_4545_ac4a_13a6c8bc34c4*mwccd3a17c_e207_4663_9b16_327b78882497-mw68039b16_b516_4fba_bedd_d4bbc1a23a02*mw65a14789_ffcf_4bfd_9d53_d2eb2f4d0896)
mwd1b16e73_4fcb_4e4c_9804_3137259ba749 = 1.0E-6; mw36cb62c6_0b3c_4d1b_9001_3b37aa7477e2 = 9.0Reaction: mw3d9e6efb_8e12_49c9_a87f_e067914b951d + mw1c97b02d_169a_4eb8_bc84_1be57c51a255 => mw1041345b_f015_436c_9eff_98211008aa1c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwd1b16e73_4fcb_4e4c_9804_3137259ba749*mw3d9e6efb_8e12_49c9_a87f_e067914b951d*mw1c97b02d_169a_4eb8_bc84_1be57c51a255^2-mw36cb62c6_0b3c_4d1b_9001_3b37aa7477e2*mw1041345b_f015_436c_9eff_98211008aa1c)
mw8186cb1d_66c4_4855_bcbb_82d75173ae8a = 20.0Reaction: mw166e3335_56c3_41ef_af0f_b583860991c1 => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwfe9ed415_d5af_469c_a549_d8981f1eb01f, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw8186cb1d_66c4_4855_bcbb_82d75173ae8a*mw166e3335_56c3_41ef_af0f_b583860991c1
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mwb56b5ab7_47cc_4fbc_b68b_dfdc6be6d7a4 = 5.5E-4Reaction: mw42919ead_5972_4151_85ac_fcc88ca105a6 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwb56b5ab7_47cc_4fbc_b68b_dfdc6be6d7a4*mw42919ead_5972_4151_85ac_fcc88ca105a6*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba)
mwb0a6bd5e_87a0_425c_a5c7_ea69903e0bf3 = 10.0Reaction: mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw42919ead_5972_4151_85ac_fcc88ca105a6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwb0a6bd5e_87a0_425c_a5c7_ea69903e0bf3*mwbae3bd11_0ab4_4587_a931_9c5dc5e777ba
mw649b47b3_4c3a_4ac9_ae94_5c38ccf81e39 = 0.002; mwc911f28c_b62f_4269_84ed_d852f6da24f9 = 0.01Reaction: mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c + mw2075d2cf_955e_4150_98b8_847103c53845 => mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw649b47b3_4c3a_4ac9_ae94_5c38ccf81e39*mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c*mw2075d2cf_955e_4150_98b8_847103c53845-mwc911f28c_b62f_4269_84ed_d852f6da24f9*mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e)
mw6bf18344_b899_4a62_ac8d_5f8bdd4bbe2f = 8.0Reaction: mw3a3e53fb_bbbf_4433_9f75_a12610dbc312 => mw9417144e_14b1_40d9_bd4b_ccd9f4714305 + mw24435476_9c30_4878_b26f_4b3c5a0685c6, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw6bf18344_b899_4a62_ac8d_5f8bdd4bbe2f*mw3a3e53fb_bbbf_4433_9f75_a12610dbc312
mw5623544e_e7e1_439f_88d3_3b0cbea8ccf5 = 30.0Reaction: mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c => mwfed0682b_39f1_4b09_94e8_c45a51744092, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw5623544e_e7e1_439f_88d3_3b0cbea8ccf5*mw8e34c23f_1891_4dc9_8f97_dc2f12a1706c
mwa466eec8_9bc0_44d5_8027_d5925b378429 = 1.0; mw448bd49f_40ad_46c9_81f6_3494057dc37d = 0.005Reaction: A2AR + mw351f6cee_3e64_4b8e_8e60_24b1aca99a92 => mw0b46978f_b522_4cde_97f0_574cd7dbbae7, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw448bd49f_40ad_46c9_81f6_3494057dc37d*A2AR*mw351f6cee_3e64_4b8e_8e60_24b1aca99a92-mwa466eec8_9bc0_44d5_8027_d5925b378429*mw0b46978f_b522_4cde_97f0_574cd7dbbae7)
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mwb93138ce_a80b_4b26_b927_6b4a00651b64 = 3.0E-4Reaction: mwd794c746_c826_4ba1_9e09_a9d1e122d925 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwb93138ce_a80b_4b26_b927_6b4a00651b64*mwd794c746_c826_4ba1_9e09_a9d1e122d925*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw0a10f9cb_3f4b_4bfa_ace9_0ecd2bd74b5e)
mwed967767_31e4_4e9e_8117_5372f9f4f79a = 3.0Reaction: mw0130a500_18e9_470f_9fac_70af44dc4a9e => mw32351ce4_eaaf_4827_8efa_342224548d8a + mw1184c368_03fc_435a_9086_dc6ed3067935, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwed967767_31e4_4e9e_8117_5372f9f4f79a*mw0130a500_18e9_470f_9fac_70af44dc4a9e
mw8db06baf_d8bb_4a1a_b415_2d51fa1e53ba = 0.25Reaction: mwfe9ed415_d5af_469c_a549_d8981f1eb01f => mw724f1afe_8032_40ae_96ca_808ab7b8b943 + mwfed0682b_39f1_4b09_94e8_c45a51744092, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw8db06baf_d8bb_4a1a_b415_2d51fa1e53ba*mwfe9ed415_d5af_469c_a549_d8981f1eb01f
mwcabc0868_2435_4850_964b_e3ddee39f5ad = 30.0Reaction: mw56dff932_134c_4d88_a611_daad00623fd0 => mw619502c3_e319_4e29_a677_b2b5f74fc2cf + mw9bcba6bc_9788_4f7f_afb5_1c8f3b33c3d1, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwcabc0868_2435_4850_964b_e3ddee39f5ad*mw56dff932_134c_4d88_a611_daad00623fd0
mwe4474191_0c92_406c_a6f5_4a167f541d36 = 0.25Reaction: mw2075d2cf_955e_4150_98b8_847103c53845 => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw29ba9e7c_6865_4817_8775_be2dbc29651e, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwe4474191_0c92_406c_a6f5_4a167f541d36*mw2075d2cf_955e_4150_98b8_847103c53845
mw6ae3f7a6_bf58_475e_930e_6bf7a79f3761 = 5.0; mw1ef56a9a_9d9b_4490_8fcd_53b7e50bf5d6 = 50.0Reaction: mw619502c3_e319_4e29_a677_b2b5f74fc2cf + mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 => mw56dff932_134c_4d88_a611_daad00623fd0, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw1ef56a9a_9d9b_4490_8fcd_53b7e50bf5d6*mw619502c3_e319_4e29_a677_b2b5f74fc2cf*mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3-mw6ae3f7a6_bf58_475e_930e_6bf7a79f3761*mw56dff932_134c_4d88_a611_daad00623fd0)
mwd05b4199_53ad_4807_9a8c_d93ce35be857 = 60.0Reaction: mwe4e36b8e_18b8_4c76_bd46_13614b71da5c => mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 + mw9d5c5c9d_301d_4e43_ba7b_7d21ccbdc2c2 + mwb80e4fa1_4849_4ed5_b3b0_3e3025c61ad8, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwd05b4199_53ad_4807_9a8c_d93ce35be857*mwe4e36b8e_18b8_4c76_bd46_13614b71da5c
mw17d612a2_c9d5_4251_8122_5f037fc630e3 = 0.00105Reaction: mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw29ba9e7c_6865_4817_8775_be2dbc29651e => mw2075d2cf_955e_4150_98b8_847103c53845, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw17d612a2_c9d5_4251_8122_5f037fc630e3*mw1c97b02d_169a_4eb8_bc84_1be57c51a255*mw29ba9e7c_6865_4817_8775_be2dbc29651e
mw9c2302f8_3d47_4247_a338_a02c53fc5331 = 1.0E-4; mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0Reaction: mw724f1afe_8032_40ae_96ca_808ab7b8b943 + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw7086a13a_619e_4069_b163_d8a05fc55f42, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw9c2302f8_3d47_4247_a338_a02c53fc5331*mw724f1afe_8032_40ae_96ca_808ab7b8b943*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw7086a13a_619e_4069_b163_d8a05fc55f42)
mwffa5af7e_9155_4942_9424_cd94ac5018ed = 0.055; mw0060906c_a035_468c_aa1c_130959bcf15a = 200.0Reaction: mwf82770b9_766a_4c4e_851a_d76da19e8517 + mwd8ea533a_c66e_4de4_8c5c_0d4201d8c8a2 => mwd86ce0dc_7329_4b27_9de0_ee6bffee3083, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwffa5af7e_9155_4942_9424_cd94ac5018ed*mwf82770b9_766a_4c4e_851a_d76da19e8517*mwd8ea533a_c66e_4de4_8c5c_0d4201d8c8a2-mw0060906c_a035_468c_aa1c_130959bcf15a*mwd86ce0dc_7329_4b27_9de0_ee6bffee3083)
mw2037ae7c_b1dc_4517_a61c_88a1f2bdcd12 = 1.0; mw49e66b9c_64bd_428a_9090_15e4132e9781 = 3.7E-4Reaction: mw1184c368_03fc_435a_9086_dc6ed3067935 + mw68d3f409_9462_4515_8c07_bc105fa0eaf1 => mwb320746f_6a8c_4c8b_ae55_23db454339d8, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw49e66b9c_64bd_428a_9090_15e4132e9781*mw1184c368_03fc_435a_9086_dc6ed3067935*mw68d3f409_9462_4515_8c07_bc105fa0eaf1-mw2037ae7c_b1dc_4517_a61c_88a1f2bdcd12*mwb320746f_6a8c_4c8b_ae55_23db454339d8)
mwc728d91d_7616_43db_bd1d_55e49e9c026a = 0.125Reaction: mw56dff932_134c_4d88_a611_daad00623fd0 => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mwed1b3928_8d78_44d1_aee7_9d11d6437cfc, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mwc728d91d_7616_43db_bd1d_55e49e9c026a*mw56dff932_134c_4d88_a611_daad00623fd0
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mwa1bc2233_5bb9_4135_88ed_bb51640faec8 = 5.625E-5Reaction: mwed1b3928_8d78_44d1_aee7_9d11d6437cfc + mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a => mw56dff932_134c_4d88_a611_daad00623fd0, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwa1bc2233_5bb9_4135_88ed_bb51640faec8*mwed1b3928_8d78_44d1_aee7_9d11d6437cfc*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw56dff932_134c_4d88_a611_daad00623fd0)
mwb494aae2_da19_4ac0_96e2_0dcd9440edc2 = 1.0; mw541807fb_7d9f_4788_9f21_cc62846b5826 = 6.25E-5Reaction: mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a + mw29ba9e7c_6865_4817_8775_be2dbc29651e => mw2075d2cf_955e_4150_98b8_847103c53845, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mw541807fb_7d9f_4788_9f21_cc62846b5826*mw46dccec6_6f0f_40f6_a10c_2f34ae7a005a*mw29ba9e7c_6865_4817_8775_be2dbc29651e-mwb494aae2_da19_4ac0_96e2_0dcd9440edc2*mw2075d2cf_955e_4150_98b8_847103c53845)
mwc52aebc2_571c_4f96_84ee_0613ae73db89 = 0.01; mw9330e49a_b214_4807_b614_4241a4a12c43 = 0.01Reaction: mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3 + mwfe9ed415_d5af_469c_a549_d8981f1eb01f => mwd794c746_c826_4ba1_9e09_a9d1e122d925, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*(mwc52aebc2_571c_4f96_84ee_0613ae73db89*mwa2c44a01_28c9_4dbd_b034_364f9b5b6cc3*mwfe9ed415_d5af_469c_a549_d8981f1eb01f-mw9330e49a_b214_4807_b614_4241a4a12c43*mwd794c746_c826_4ba1_9e09_a9d1e122d925)
mw1a6a8649_d7cb_4379_983a_cca2acac3112 = 2.5Reaction: mw07c7392b_8d89_4b94_97c5_59f7e256b6f2 => mw1c97b02d_169a_4eb8_bc84_1be57c51a255 + mw2badefa3_32e8_4b66_9e69_245d9ec74e33, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw1a6a8649_d7cb_4379_983a_cca2acac3112*mw07c7392b_8d89_4b94_97c5_59f7e256b6f2
mw3a56b314_299f_48d2_a179_97bf6a30f38f = 0.006Reaction: mw9417144e_14b1_40d9_bd4b_ccd9f4714305 => mw081c9f7b_011e_440f_971d_d0316d2a1e6c, Rate Law: mw26af457f_7462_4410_a392_e0bbb6071ea5*mw3a56b314_299f_48d2_a179_97bf6a30f38f*mw9417144e_14b1_40d9_bd4b_ccd9f4714305

States:

NameDescription
mw1041345b f015 436c 9eff 98211008aa1c[cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mw1c97b02d 169a 4eb8 bc84 1be57c51a255[3',5'-cyclic AMP]
mwed1b3928 8d78 44d1 aee7 9d11d6437cfc[calcium(2+); GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw46dccec6 6f0f 40f6 a10c 2f34ae7a005a[ATP]
mw3e1a2fbf 37b1 490c 9528 6cb6bbf11b21[dopamine]
mw2badefa3 32e8 4b66 9e69 245d9ec74e33[calcium(2+); GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mwd86ce0dc 7329 4b27 9de0 ee6bffee3083[D(2) dopamine receptor; Guanine nucleotide-binding protein G(i) subunit alpha-1; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
A2AR[Adenosine receptor A2a]
mw3d9e6efb 8e12 49c9 a87f e067914b951d[cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mwfed0682b 39f1 4b09 94e8 c45a51744092[GDP; Guanine nucleotide-binding protein G(olf) subunit alpha]
mw07c7392b 8d89 4b94 97c5 59f7e256b6f2[calcium(2+); GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mwccd3a17c e207 4663 9b16 327b78882497[calcium(2+)]
mw32351ce4 eaaf 4827 8efa 342224548d8a[Cyclin-dependent-like kinase 5]
mw619502c3 e319 4e29 a677 b2b5f74fc2cf[calcium(2+); ATP; Adenylate cyclase type 5]
mw56dff932 134c 4d88 a611 daad00623fd0[calcium(2+); GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw9417144e 14b1 40d9 bd4b ccd9f4714305[Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
mw0a10f9cb 3f4b 4bfa ace9 0ecd2bd74b5e[GTP; ATP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw724f1afe 8032 40ae 96ca 808ab7b8b943[Adenylate cyclase type 5]
A2ARAdnGolf[adenosine; Adenosine receptor A2a; Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(I)/G(S)/G(O) subunit gamma-2]
mwa2c44a01 28c9 4dbd b034 364f9b5b6cc3[GTP; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw6e845d87 603e 4463 874d 866f554303df[3',5'-cyclic AMP; cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]
mw9bcba6bc 9788 4f7f afb5 1c8f3b33c3d1[GDP; Guanine nucleotide-binding protein G(i) subunit alpha-1]
A2ARAdn[adenosine; Adenosine receptor A2a]
mw29ba9e7c 6865 4817 8775 be2dbc29651e[GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw081c9f7b 011e 440f 971d d0316d2a1e6c[Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform; Serine/threonine-protein phosphatase 2A 55 kDa regulatory subunit B alpha isoform; Serine/threonine-protein phosphatase 2A catalytic subunit alpha isoform]
mwd794c746 c826 4ba1 9e09 a9d1e122d925[GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha; Guanine nucleotide-binding protein G(i) subunit alpha-1]
mw7df45520 98cc 4c0b 91a7 c6e7297de98a[calcium(2+); Adenylate cyclase type 5]
mw0130a500 18e9 470f 9fac 70af44dc4a9e[Cyclin-dependent-like kinase 5; Protein phosphatase 1 regulatory subunit 1B]
mwfe9ed415 d5af 469c a549 d8981f1eb01f[GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha]
mwf82770b9 766a 4c4e 851a d76da19e8517[D(2) dopamine receptor]
mw42919ead 5972 4151 85ac fcc88ca105a6[calcium(2+); GTP; Adenylate cyclase type 5; Guanine nucleotide-binding protein G(olf) subunit alpha]
mw1184c368 03fc 435a 9086 dc6ed3067935[Protein phosphatase 1 regulatory subunit 1B]
mw1f3b8982 3b8c 42b6 8b0f 49b037cbda43[3',5'-cyclic AMP; cAMP and cAMP-inhibited cGMP 3',5'-cyclic phosphodiesterase 10A]

Nakakuki2010_CellFateDecision_Core: BIOMD0000000251v0.0.1

This model describes the activation of immediate early genes such as cFos after EGF or heregulin (HRG) stimulation of th…

Details

Activation of ErbB receptors by epidermal growth factor (EGF) or heregulin (HRG) determines distinct cell-fate decisions, although signals propagate through shared pathways. Using mathematical modeling and experimental approaches, we unravel how HRG and EGF generate distinct, all-or-none responses of the phosphorylated transcription factor c-Fos. In the cytosol, EGF induces transient and HRG induces sustained ERK activation. In the nucleus, however, ERK activity and c-fos mRNA expression are transient for both ligands. Knockdown of dual-specificity phosphatases extends HRG-stimulated nuclear ERK activation, but not c-fos mRNA expression, implying the existence of a HRG-induced repressor of c-fos transcription. Further experiments confirmed that this repressor is mainly induced by HRG, but not EGF, and requires new protein synthesis. We show how a spatially distributed, signaling-transcription cascade robustly discriminates between transient and sustained ERK activities at the c-Fos system level. The proposed control mechanisms are general and operate in different cell types, stimulated by various ligands. link: http://identifiers.org/pubmed/20493519

Parameters:

NameDescription
tau1 = 3.07; L = 1.0; K1 = 1.09Reaction: => x1, Rate Law: compartment*((-x1)/tau1+K1*L/tau1)
k6=0.13; k7 = 0.5; n=1.1Reaction: => cFOSp; ppERKn, pRSKn, Rate Law: compartment*((ppERKn*pRSKn)^n/(k6^n+(ppERKn*pRSKn)^n)-k7*cFOSp)
k8=0.08; k7 = 0.5Reaction: => cFOSm; cFOSp, Rate Law: compartment*(k7*cFOSp-k8*cFOSm)
k2=50.0; k1=15.0; k3=14.0Reaction: => ppERKn; ppERKc, DUSP, Rate Law: compartment*((k1*ppERKc-k2*ppERKn)-k3*DUSP*ppERKn)
tau2 = 472.0; L = 1.0; K2 = 2.89Reaction: => x2, Rate Law: compartment*((-x2)/tau2+K2*L/tau2)
k=1.0Reaction: => DUSP; ppERKn, Rate Law: compartment*k*ppERKn
k4=0.1; k5=0.15Reaction: => pRSKn; ppERKn, Rate Law: compartment*(k4*ppERKn-k5*pRSKn)
k13 = 0.06; k11 = 0.11; k12=0.001Reaction: => pcFOS; cFOS, ppERKc, Rate Law: compartment*((k11*cFOS*ppERKc-k12*pcFOS)-k13*pcFOS)
k9=0.3; k13 = 0.06; k10=0.3; k11 = 0.11Reaction: => cFOS; cFOSm, ppERKc, pcFOS, Rate Law: compartment*(((k9*cFOSm-k10*cFOS)-k11*cFOS*ppERKc)+k13*pcFOS)

States:

NameDescription
ppERKn[Phosphoprotein; Mitogen-activated protein kinase 1; Mitogen-activated protein kinase 3; p-ERK1/2/5 [nucleoplasm]]
cFOSp[FOS [nucleoplasm]; FOS, AP-1, C-FOS, p55]
x1x1
cFOS[Proto-oncogene c-Fos; FOS [nucleoplasm]]
pcFOS[Proto-oncogene c-Fos; p-T325,T331,S362,S374-FOS [nucleoplasm]]
cFOSm[FOS [nucleoplasm]; Proto-oncogene c-Fos]
x2x2
DUSP[DUSP, MKP; IPR014393; ERK-specific DUSP [nucleoplasm]; Dual specificity protein phosphatase 8; Dual specificity protein phosphatase 10; Dual specificity protein phosphatase 5; Dual specificity protein phosphatase 4; Dual specificity protein phosphatase 2; Dual specificity protein phosphatase 1]
pRSKn[Ribosomal protein S6 kinase [nucleoplasm]]
ppERKc[Phosphoprotein; p-T185,Y187-MAPK1 [cytosol]; p-T202,Y204-MAPK3 [cytosol]]

Nakakuki2010_CellFateDecision_Mechanistic: BIOMD0000000250v0.0.1

This mechanistic model describes the activation of immediate early genes such as cFos after EGF or heregulin (HRG) stimu…

Details

Activation of ErbB receptors by epidermal growth factor (EGF) or heregulin (HRG) determines distinct cell-fate decisions, although signals propagate through shared pathways. Using mathematical modeling and experimental approaches, we unravel how HRG and EGF generate distinct, all-or-none responses of the phosphorylated transcription factor c-Fos. In the cytosol, EGF induces transient and HRG induces sustained ERK activation. In the nucleus, however, ERK activity and c-fos mRNA expression are transient for both ligands. Knockdown of dual-specificity phosphatases extends HRG-stimulated nuclear ERK activation, but not c-fos mRNA expression, implying the existence of a HRG-induced repressor of c-fos transcription. Further experiments confirmed that this repressor is mainly induced by HRG, but not EGF, and requires new protein synthesis. We show how a spatially distributed, signaling-transcription cascade robustly discriminates between transient and sustained ERK activities at the c-Fos system level. The proposed control mechanisms are general and operate in different cell types, stimulated by various ligands. link: http://identifiers.org/pubmed/20493519

Parameters:

NameDescription
V14 = 5.636949216; K14 = 34180.48Reaction: DUSP_c => pDUSP_c; ppERK_c, Rate Law: cytoplasm*V14*ppERK_c*DUSP_c/(K14+DUSP_c)
K4 = 60.0; K3 = 160.0; V4 = 0.648Reaction: ppERK_c => pERK_c; pERK_c, Rate Law: cytoplasm*V4*ppERK_c/(K4*(1+pERK_c/K3)+ppERK_c)
p50 = 26.59483436Reaction: DUSP_n_pERK_n => DUSP_n + ERK_n, Rate Law: nucleus*p50*DUSP_n_pERK_n
p38 = 2.57E-4Reaction: c_FOS_c =>, Rate Law: cytoplasm*p38*c_FOS_c
m51 = 9.544308421; p51 = 0.01646825Reaction: DUSP_n + ERK_n => DUSP_n_ERK_n, Rate Law: nucleus*(p51*DUSP_n*ERK_n-m51*DUSP_n_ERK_n)
p49 = 0.314470502; m49 = 2.335459127Reaction: DUSP_n + pERK_n => DUSP_n_pERK_n, Rate Law: nucleus*(p49*DUSP_n*pERK_n-m49*DUSP_n_pERK_n)
KexERKP = 0.018; Vc = 940.0; KimERKP = 0.012; Vn = 220.0Reaction: pERK_c => pERK_n, Rate Law: KimERKP*Vc*pERK_c-KexERKP*Vn*pERK_n
KimRSKP = 0.025925065; KexRSKP = 0.129803956; Vc = 940.0; Vn = 220.0Reaction: pRSK_c => pRSK_n, Rate Law: KimRSKP*Vc*pRSK_c-KexRSKP*Vn*pRSK_n
V115 = 13.74244; K115 = 2122.045Reaction: pMEK => MEK, Rate Law: cytoplasm*V115*pMEK/(K115+pMEK)
V106 = 0.109304; K106 = 606.871Reaction: RsD => RsT; HRG, Rate Law: cytoplasm*V106*HRG*RsD/(K106+RsD)
p47 = 0.001670815; m47 = 15.80783969Reaction: DUSP_n + ppERK_n => DUSP_n_ppERK_n, Rate Law: nucleus*(p47*DUSP_n*ppERK_n-m47*DUSP_n_ppERK_n)
p58 = 2.70488E-4; Vn = 220.0Reaction: PreFmRNA => FmRNA, Rate Law: p58*Vn*PreFmRNA
K102 = 237.2001; V102 = 0.09858154Reaction: A1_2 => A1, Rate Law: cytoplasm*V102*A1_2/(K102+A1_2)
V108 = 0.03436149; K108 = 11.5048Reaction: RsT => RsD; A2_2, Rate Law: cytoplasm*V108*A2_2*RsT/(K108+RsT)
p11 = 1.26129E-4; Vn = 220.0Reaction: PreDUSPmRNA => DUSPmRNA, Rate Law: p11*Vn*PreDUSPmRNA
K31 = 185.9760682; V31 = 0.655214248; KF31 = 0.013844393; nF31 = 2.800340453; n31 = 1.988003164Reaction: => PreFOSmRNA; pCREB_n, pElk1_n, Fn, Rate Law: nucleus*V31*(pCREB_n*pElk1_n)^n31/(K31^n31+(pCREB_n*pElk1_n)^n31+(Fn/KF31)^nF31)
p32 = 0.003284434; Vn = 220.0Reaction: PreFOSmRNA => c_FOSmRNA, Rate Law: p32*Vn*PreFOSmRNA
p45 = 2.57E-4Reaction: FOSn =>, Rate Law: nucleus*p45*FOSn
KimERK = 0.012; KexERK = 0.018; Vc = 940.0; Vn = 220.0Reaction: ERK_c => ERK_n, Rate Law: KimERK*Vc*ERK_c-KexERK*Vn*ERK_n
K105 = 1.027895; V105 = 0.05393704Reaction: RsD => RsT; EGF, Rate Law: cytoplasm*V105*EGF*RsD/(K105+RsD)
K107 = 424.6884; V107 = 5.291093Reaction: RsT => RsD; A1_2, Rate Law: cytoplasm*V107*A1_2*RsT/(K107+RsT)
K57 = 0.637490056; V57 = 1.026834758; n57 = 3.584464176Reaction: => PreFmRNA; FOSn_2, Rate Law: nucleus*V57*FOSn_2^n57/(K57^n57+FOSn_2^n57)
p33 = 6.01234209304622E-4Reaction: c_FOSmRNA =>, Rate Law: cytoplasm*p33*c_FOSmRNA
p55 = 26.59483436Reaction: pDUSP_n_pERK_n => pDUSP_n + ERK_n, Rate Law: nucleus*p55*pDUSP_n_pERK_n
KexDUSP = 0.070467899; Vc = 940.0; KimDUSP = 0.024269764; Vn = 220.0Reaction: DUSP_c => DUSP_n, Rate Law: KimDUSP*Vc*DUSP_c-KexDUSP*Vn*DUSP_n
KimFOS = 0.54528521; Vc = 940.0; KexFOS = 0.133249762; Vn = 220.0Reaction: c_FOS_c => FOSn, Rate Law: KimFOS*Vc*c_FOS_c-KexFOS*Vn*FOSn
V29 = 0.518529841; K29 = 21312.69109Reaction: Elk1_n => pElk1_n; ppERK_n, Rate Law: nucleus*V29*ppERK_n*Elk1_n/(K29+Elk1_n)
KimFOSP = 0.54528521; KexFOSP = 0.133249762; Vc = 940.0; Vn = 220.0Reaction: pc_FOS_c => FOSn_2, Rate Law: KimFOSP*Vc*pc_FOS_c-KexFOSP*Vn*FOSn_2
KimERKPP = 0.011; KexERKPP = 0.013; Vc = 940.0; Vn = 220.0Reaction: ppERK_c => ppERK_n, Rate Law: KimERKPP*Vc*ppERK_c-KexERKPP*Vn*ppERK_n
p59 = 0.001443889Reaction: FmRNA =>, Rate Law: cytoplasm*p59*FmRNA
V112 = 0.8850982; K112 = 4665.217Reaction: Kin_2 => Kin; A3_2, Rate Law: cytoplasm*V112*A3_2*Kin_2/(K112+Kin_2)
m54 = 2.335459127; p54 = 0.314470502Reaction: pDUSP_n + pERK_n => pDUSP_n_pERK_n, Rate Law: nucleus*(p54*pDUSP_n*pERK_n-m54*pDUSP_n_pERK_n)
p23 = 4.81E-5Reaction: pDUSP_n =>, Rate Law: nucleus*p23*pDUSP_n
V27 = 19.23118154; K27 = 441.5834425Reaction: CREB_n => pCREB_n; pRSK_n, Rate Law: nucleus*V27*pRSK_n*CREB_n/(K27+CREB_n)
K111 = 858.3423; V111 = 0.02487469Reaction: Kin => Kin_2; HRG, Rate Law: cytoplasm*V111*HRG*Kin/(K111+Kin)
K20 = 735598.6967; V20 = 0.157678678Reaction: DUSP_n => pDUSP_n; ppERK_n, Rate Law: nucleus*V20*ppERK_n*DUSP_n/(K20+DUSP_n)
p63 = 4.13466150826031E-5Reaction: Fn =>, Rate Law: nucleus*cytoplasm*p63*Fn/nucleus
m56 = 9.544308421; p56 = 0.01646825Reaction: pDUSP_n + ERK_n => pDUSP_n_ERK_n, Rate Law: nucleus*(p56*pDUSP_n*ERK_n-m56*pDUSP_n_ERK_n)
K104 = 4046.71; V104 = 4.635749Reaction: A2_2 => A2, Rate Law: cytoplasm*V104*A2_2/(K104+A2_2)
V6 = 19.4987234631759; K6 = 29.9407371620698; K5 = 29.94073716Reaction: ppERK_n => pERK_n; pERK_n, Rate Law: nucleus*V6*ppERK_n/(K6*(1+pERK_n/K5)+ppERK_n)
K44 = 0.051168202; V44 = 0.078344305Reaction: FOSn_2 => FOSn, Rate Law: nucleus*V44*FOSn_2/(K44+FOSn_2)
p53 = 0.686020478Reaction: pDUSP_n_ppERK_n => pDUSP_n + pERK_n, Rate Law: nucleus*p53*pDUSP_n_ppERK_n
K103 = 1334.132; V103 = 0.3573399Reaction: A2 => A2_2; HRG, Rate Law: cytoplasm*V103*HRG*A2/(K103+A2)
p60 = 0.002448164Reaction: => F; FmRNA, Rate Law: cytoplasm*p60*FmRNA
p61 = 3.49860901414122E-5Reaction: F =>, Rate Law: cytoplasm*p61*F
p46 = 4.81E-5Reaction: FOSn_2 =>, Rate Law: nucleus*p46*FOSn_2
p12 = 0.007875765Reaction: DUSPmRNA =>, Rate Law: cytoplasm*p12*DUSPmRNA
V43 = 0.076717457; K43 = 1157.116021Reaction: FOSn => FOSn_2; pRSK_n, Rate Law: nucleus*V43*pRSK_n*FOSn/(K43+FOSn)
K114 = 7.774197; V114 = 0.03957055Reaction: MEK => pMEK; Kin_2, Rate Law: cytoplasm*V114*Kin_2*MEK/(K114+MEK)
V113 = 0.05377297; K113 = 20.50809Reaction: MEK => pMEK; RsT, Rate Law: cytoplasm*V113*RsT*MEK/(K113+MEK)
V110 = 0.08258693; K110 = 425.5268Reaction: A3_2 => A3, Rate Law: cytoplasm*V110*A3_2/(K110+A3_2)
V42 = 0.909968714; K42 = 3992.061328Reaction: FOSn => FOSn_2; ppERK_n, Rate Law: nucleus*V42*ppERK_n*FOSn/(K42+FOSn)
p48 = 0.686020478Reaction: DUSP_n_ppERK_n => DUSP_n + pERK_n, Rate Law: nucleus*p48*DUSP_n_ppERK_n
p52 = 0.001670815; m52 = 15.80783969Reaction: pDUSP_n + ppERK_n => pDUSP_n_ppERK_n, Rate Law: nucleus*(p52*pDUSP_n*ppERK_n-m52*pDUSP_n_ppERK_n)
K30 = 15.04396629; V30 = 13.79479021Reaction: pElk1_n => Elk1_n, Rate Law: nucleus*V30*pElk1_n/(K30+pElk1_n)
V109 = 0.1374307; K109 = 7424.816Reaction: A3 => A3_2; HRG, Rate Law: cytoplasm*V109*HRG*A3/(K109+A3)
V101 = 0.01807448; K101 = 3475.168Reaction: A1 => A1_2; EGF, Rate Law: cytoplasm*V101*EGF*A1/(K101+A1)
K2 = 350.0; V2 = 0.22; Fct = 0.7485; K1 = 307.041525298866Reaction: pERK_c => ppERK_c; pMEK, ERK_c, Rate Law: cytoplasm*V2*Fct*pMEK*pERK_c/(K2*(1+ERK_c/K1)+pERK_c)
p34 = 7.64816282169636E-5Reaction: => c_FOS_c; c_FOSmRNA, Rate Law: cytoplasm*p34*c_FOSmRNA
K2 = 350.0; V1 = 0.342848369838443; Fct = 0.7485; K1 = 307.041525298866Reaction: ERK_c => pERK_c; pMEK, pERK_c, Rate Law: cytoplasm*V1*Fct*pMEK*ERK_c/(K1*(1+pERK_c/K2)+ERK_c)
V21 = 0.005648117; K21 = 387.8377182Reaction: pDUSP_n => DUSP_n, Rate Law: nucleus*V21*pDUSP_n/(K21+pDUSP_n)
p13 = 0.001245747Reaction: => DUSP_c; DUSPmRNA, Rate Law: cytoplasm*p13*DUSPmRNA
KimF = 0.019898797; KexF = 0.396950616; Vc = 940.0; Vn = 220.0Reaction: F => Fn, Rate Law: KimF*Vc*F-KexF*Vn*Fn
KimDUSPP = 0.024269764; KexDUSPP = 0.070467899; Vc = 940.0; Vn = 220.0Reaction: pDUSP_c => pDUSP_n, Rate Law: KimDUSPP*Vc*pDUSP_c-KexDUSPP*Vn*pDUSP_n
V10 = 29.24109258; n10 = 3.970849295; K10 = 169.0473748Reaction: => PreDUSPmRNA; ppERK_n, Rate Law: nucleus*V10*ppERK_n^n10/(K10^n10+ppERK_n^n10)
K4 = 60.0; K3 = 160.0; V3 = 0.72Reaction: pERK_c => ERK_c; ppERK_c, Rate Law: cytoplasm*V3*pERK_c/(K3*(1+ppERK_c/K4)+pERK_c)

States:

NameDescription
PreFmRNAPreFmRNA
A1 2A1_2
RsTRsT
A2 2A2_2
RsDRsD
Elk1 nElk1_n
DUSP nDUSP_n
pDUSP npDUSP_n
pDUSP n ERK npDUSP_n_ERK_n
pERK cpERK_c
FnFn
DUSP cDUSP_c
pElk1 npElk1_n
MEKMEK
pRSK cpRSK_c
pRSK npRSK_n
pERK npERK_n
A3A3
A1A1
FOSnFOSn
ppERK nppERK_n
A3 2A3_2
CREB nCREB_n
KinKin
FmRNAFmRNA
ERK cERK_c
DUSP n pERK nDUSP_n_pERK_n
A2A2
c FOS cc_FOS_c
DUSPmRNADUSPmRNA
pMEKpMEK
pDUSP n pERK npDUSP_n_pERK_n
c FOSmRNAc_FOSmRNA
FOSn 2FOSn_2
DUSP n ppERK nDUSP_n_ppERK_n
ppERK cppERK_c
PreFOSmRNAPreFOSmRNA
pDUSP n ppERK npDUSP_n_ppERK_n
PreDUSPmRNAPreDUSPmRNA
ERK nERK_n
pCREB npCREB_n
FF
Kin 2Kin_2

Nakano2010_Synaptic_Plasticity: MODEL1101170000v0.0.1

This is an SBML version of the model described in: **A kinetic model of dopamine- and calcium-dependent striatal synapti…

Details

Corticostriatal synapse plasticity of medium spiny neurons is regulated by glutamate input from the cortex and dopamine input from the substantia nigra. While cortical stimulation alone results in long-term depression (LTD), the combination with dopamine switches LTD to long-term potentiation (LTP), which is known as dopamine-dependent plasticity. LTP is also induced by cortical stimulation in magnesium-free solution, which leads to massive calcium influx through NMDA-type receptors and is regarded as calcium-dependent plasticity. Signaling cascades in the corticostriatal spines are currently under investigation. However, because of the existence of multiple excitatory and inhibitory pathways with loops, the mechanisms regulating the two types of plasticity remain poorly understood. A signaling pathway model of spines that express D1-type dopamine receptors was constructed to analyze the dynamic mechanisms of dopamine- and calcium-dependent plasticity. The model incorporated all major signaling molecules, including dopamine- and cyclic AMP-regulated phosphoprotein with a molecular weight of 32 kDa (DARPP32), as well as AMPA receptor trafficking in the post-synaptic membrane. Simulations with dopamine and calcium inputs reproduced dopamine- and calcium-dependent plasticity. Further in silico experiments revealed that the positive feedback loop consisted of protein kinase A (PKA), protein phosphatase 2A (PP2A), and the phosphorylation site at threonine 75 of DARPP-32 (Thr75) served as the major switch for inducing LTD and LTP. Calcium input modulated this loop through the PP2B (phosphatase 2B)-CK1 (casein kinase 1)-Cdk5 (cyclin-dependent kinase 5)-Thr75 pathway and PP2A, whereas calcium and dopamine input activated the loop via PKA activation by cyclic AMP (cAMP). The positive feedback loop displayed robust bi-stable responses following changes in the reaction parameters. Increased basal dopamine levels disrupted this dopamine-dependent plasticity. The present model elucidated the mechanisms involved in bidirectional regulation of corticostriatal synapses and will allow for further exploration into causes and therapies for dysfunctions such as drug addiction. link: http://identifiers.org/pubmed/20169176

Nampala2013 - Liver enzyme elevation in HIV mono-infection: MODEL1812040002v0.0.1

the model depicts a unique endemic equilibrium with a transcritical bifurcation when the basic reproductive number is un…

Details

HIV-infected individuals are increasingly becoming susceptible to liver disease and, hence, liver-related mortality is on a rise. The presence of CD4+ in the liver and the presence of C-X-C chemokine receptor type 4 (CXCR4) on human hepatocytes provide a conducive environment for HIV invasion. In this study, a mathematical model is used to analyse the dynamics of HIV in the liver with the aim of investigating the existence of liver enzyme elevation in HIV mono-infected individuals. In the presence of HIV-specific cytotoxic T-lymphocytes, the model depicts a unique endemic equilibrium with a transcritical bifurcation when the basic reproductive number is unity. Results of the study show that the level of liver enzyme alanine aminotransferase (ALT) increases with increase in the rate of hepatocytes production. Numerical simulations reveal significant elevation of alanine aminotransferase with increase in viral load. The findings presuppose that while liver damage in HIV infection has mostly been associated with HIV/HBV coinfection and use of antiretroviral therapy (ART), it is possible to have liver damage solely with HIV infection. link: http://identifiers.org/pubmed/23291466

Nanda2013 - B cell chronic lymphocytic leukemia A model with immune response: MODEL2001090002v0.0.1

<notes xmlns="http://www.sbml.org/sbml/level2/version4"> <body xmlns="http://www.w3.org/1…

Details

B cell chronic lymphocytic leukemia (B-CLL) is known to havesubstantial clinical heterogeneity. There is no cure, but treatments allow fordisease management. However, the wide range of clinical courses experiencedby B-CLL patients makes prognosis and hence treatment a significant chal-lenge. In an attempt to study disease progression across different patients viaa unified yet flexible approach, we present a mathematical model of B-CLLwith immune response, that can capture both rapid and slow disease progres-sion. This model includes four different cell populations in the peripheral bloodof humans: B-CLL cells, NK cells, cytotoxic T cells and helper T cells. Weanalyze existing data in the medical literature, determine ranges of values forparameters of the model, and compare our model outcomes to clinical patientdata. The goal of this work is to provide a tool that may shed light on factorsaffecting the course of disease progression in patients. This modeling tool canserve as a foundation upon which future treatments can be based. link: http://identifiers.org/doi/10.3934/dcdsb.2013.18.1053

Naski1991 - alpha-Thrombin-catalyzed conversion of Fibrinogen: MODEL1808210002v0.0.1

Mathematical model of blood coagulation factor alpha-thrombin and conversion of fibrinogen to fibrin with ATIII inhibiti…

Details

In this study we report a kinetic model for the alpha-thrombin-catalyzed production of fibrin I and fibrin II at pH 7.4, 37 degrees C, gamma/2 0.17. The fibrin is produced by the action of human alpha-thrombin on plasma levels of human fibrinogen in the presence of the major inhibitor of alpha-thrombin in plasma, antithrombin III (AT). This model quantitatively accounts for the time dependence of alpha-thrombin-catalyzed release of fibrinopeptides A and B concurrent with the inactivation of alpha-thrombin by AT and delineates the concerted interactions of alpha-thrombin, fibrin(ogen), and AT during the production of a fibrin clot. The model also provides a method for estimating the concentration of alpha-thrombin required to produce a clot of known composition and predicts a direct relationship between the plasma concentration of fibrinogen and the amount of fibrin produced by a bolus of alpha-thrombin. The predicted relationship between the concentration of fibrinogen and the amount of fibrin produced in plasma provides a plausible explanation for the observed linkage between plasma concentrations of fibrinogen and the risk for ischemic heart disease. link: http://identifiers.org/pubmed/2071587

Nave2018 - prostate cancer model: MODEL1910030001v0.0.1

This model is based on paper: Combination of singularly perturbed vector field method and method of directly defining t…

Details

We propose a new method to solve a system of complex ordinary differential equations (ODEs) with hidden hierarchy. Given a complex system of the ODE, the hierarchy of the system is generally hidden. Once we reveal the hierarchy of the system, the system can be reduced into subsystems called slow and fast subsystems. This division of slow and fast subsystems reduces the analysis and hence reduces the computation time, which can be expensive. In our new method, we first apply the singularly perturbed vector field method that is the global quasi-linearization method. This method exposes the hierarchy of a given complex system. Subsequently, we apply a version of the homotopy analysis method called the method of directly defining the inverse mapping. We applied our new method to the immunotherapy of advanced prostate cancer. link: http://identifiers.org/pubmed/30384811

Nayak2015 - Blood Coagulation Network - Predicting the Effects of Various Therapies on Biomarkers: BIOMD0000000611v0.0.1

Nayak2015 - Blood Coagulation Network - Predicting the Effects of Various Therapies on BiomarkersNote:The SBML model is…

Details

A number of therapeutics have been developed or are under development aiming to modulate the coagulation network to treat various diseases. We used a systems model to better understand the effect of modulating various components on blood coagulation. A computational model of the coagulation network was built to match in-house in vitro thrombin generation and activated Partial Thromboplastin Time (aPTT) data with various concentrations of recombinant factor VIIa (FVIIa) or factor Xa added to normal human plasma or factor VIII-deficient plasma. Sensitivity analysis applied to the model revealed that lag time, peak thrombin concentration, area under the curve (AUC) of the thrombin generation profile, and aPTT show different sensitivity to changes in coagulation factors' concentrations and type of plasma used (normal or factor VIII-deficient). We also used the model to explore how variability in concentrations of the proteins in coagulation network can impact the response to FVIIa treatment. link: http://identifiers.org/pubmed/26312163

Parameters:

NameDescription
k6 = 0.009975373Reaction: Xa + VII => Xa + VIIa; Xa, VII, Xa, VII, Rate Law: k6*Xa*VII
k30 = 0.10001522; k29 = 149.91541Reaction: Xa_Va + II => Xa_Va_II; Xa_Va, II, Xa_Va_II, Xa_Va, II, Xa_Va_II, Rate Law: k30*Xa_Va*II-k29*Xa_Va_II
k32 = 0.21872155Reaction: mIIa + Xa_Va => IIa + Xa_Va; mIIa, Xa_Va, mIIa, Xa_Va, Rate Law: k32*mIIa*Xa_Va
k10 = 8.9987819Reaction: TF_VIIa_X => TF_VIIa_Xa; TF_VIIa_X, TF_VIIa_X, Rate Law: k10*TF_VIIa_X
k25 = 0.0013357963Reaction: IXa_VIIIa_X => VIIIa1_L + VIIIa2 + X + IXa; IXa_VIIIa_X, IXa_VIIIa_X, Rate Law: k25*IXa_VIIIa_X
k22 = 42.71401Reaction: IXa_VIIIa_X => IXa_VIIIa + Xa; IXa_VIIIa_X, IXa_VIIIa_X, Rate Law: k22*IXa_VIIIa_X
k26 = 0.0013946425Reaction: IXa_VIIIa => VIIIa1_L + VIIIa2 + IXa; IXa_VIIIa, IXa_VIIIa, Rate Law: k26*IXa_VIIIa
mwea0d7c35_f4d2_4205_8c59_11ac05134dde = 1.0958881E-4Reaction: mwbdb849d8_2b25_4551_8de8_adc8bead2303 => mw931f65a6_3967_4ac2_9904_ba791b216fc2; mwbdb849d8_2b25_4551_8de8_adc8bead2303, mwbdb849d8_2b25_4551_8de8_adc8bead2303, Rate Law: mwea0d7c35_f4d2_4205_8c59_11ac05134dde*mwbdb849d8_2b25_4551_8de8_adc8bead2303
mw4fc81076_be53_4fc3_9ade_3587e8d60355 = 0.1857857Reaction: mw3cec90c2_500e_4f30_b6be_325ef5194755 => mwa6be116e_72f1_439e_bca6_eb61f79cc68e + mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f; mw3cec90c2_500e_4f30_b6be_325ef5194755, mw3cec90c2_500e_4f30_b6be_325ef5194755, Rate Law: mw4fc81076_be53_4fc3_9ade_3587e8d60355*mw3cec90c2_500e_4f30_b6be_325ef5194755
k21 = 0.048795021; k20 = 0.0013766033Reaction: IXa_VIIIa + X => IXa_VIIIa_X; IXa_VIIIa, X, IXa_VIIIa_X, IXa_VIIIa, X, IXa_VIIIa_X, Rate Law: k21*IXa_VIIIa*X-k20*IXa_VIIIa_X
mw8482ca53_fca1_4841_ac2f_2469a76a758e = 0.12914436; mw1511789f_5e7b_43bf_b162_d930b027a867 = 0.006Reaction: Xa + Va => Xa_Va; Xa, Va, Xa_Va, Xa, Va, Xa_Va, Rate Law: mw8482ca53_fca1_4841_ac2f_2469a76a758e*Xa*Va-mw1511789f_5e7b_43bf_b162_d930b027a867*Xa_Va
mwa2636601_825e_4846_aa2d_c35bd242ec99 = 0.032359973Reaction: mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43 => mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + mwf5c3f9df_7ccf_4ca7_b241_471a66720da8; mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43, mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43, Rate Law: mwa2636601_825e_4846_aa2d_c35bd242ec99*mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43
k16 = 3.764127E-5Reaction: Xa + II => Xa + IIa + mwbdb849d8_2b25_4551_8de8_adc8bead2303; Xa, II, Xa, II, Rate Law: k16*Xa*II
mw7aeacec0_be36_49bf_8548_7a3e2b5fe3cb = 0.029887563Reaction: mwa4fcfa0c_6944_42fc_8c74_7865f13953c8 => mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + IXa + mwf5c3f9df_7ccf_4ca7_b241_471a66720da8; mwa4fcfa0c_6944_42fc_8c74_7865f13953c8, mwa4fcfa0c_6944_42fc_8c74_7865f13953c8, Rate Law: mw7aeacec0_be36_49bf_8548_7a3e2b5fe3cb*mwa4fcfa0c_6944_42fc_8c74_7865f13953c8
k15 = 2.3887492Reaction: TF_VIIa_IX => TF_VIIa + IXa; TF_VIIa_IX, TF_VIIa_IX, Rate Law: k15*TF_VIIa_IX
mw61fdd721_9193_442c_bc9e_f1058c4720e7 = 1.2943783E-5Reaction: mw6d041b25_87db_4394_9b8b_7ac61e01f359 => VIIa + Xa; mw6d041b25_87db_4394_9b8b_7ac61e01f359, mw6d041b25_87db_4394_9b8b_7ac61e01f359, Rate Law: mw61fdd721_9193_442c_bc9e_f1058c4720e7*mw6d041b25_87db_4394_9b8b_7ac61e01f359
mw7300dcac_9389_4201_88c7_7effa7fdb0f3 = 10.565569Reaction: mwe70b2c96_44b9_48eb_967a_7eb850a916a6 => mw6591152c_8b5a_4c9b_b095_956988a01ba0 + IXa; mwe70b2c96_44b9_48eb_967a_7eb850a916a6, mwe70b2c96_44b9_48eb_967a_7eb850a916a6, Rate Law: mw7300dcac_9389_4201_88c7_7effa7fdb0f3*mwe70b2c96_44b9_48eb_967a_7eb850a916a6
mw6843129b_7601_452f_be5d_977f7203bfb5 = 0.0345Reaction: mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820 => Xa + VIIIa; mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820, mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820, Rate Law: mw6843129b_7601_452f_be5d_977f7203bfb5*mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820
k3 = 0.0019496187; k4 = 0.075680013Reaction: TF + VIIa => TF_VIIa; TF, VIIa, TF_VIIa, TF, VIIa, TF_VIIa, Rate Law: k4*TF*VIIa-k3*TF_VIIa
k17 = 1.44895Reaction: IIa + VIII => IIa + VIIIa; IIa, VIII, IIa, VIII, Rate Law: k17*IIa*VIII
mw234b484f_d2d5_4ae8_a077_217c600588d8 = 0.24027638Reaction: mw2e632a32_3823_4933_95cb_19567cbcc66a => mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + mw18e5caa7_26eb_4521_b217_da75bb3193ad; mw2e632a32_3823_4933_95cb_19567cbcc66a, mw2e632a32_3823_4933_95cb_19567cbcc66a, Rate Law: mw234b484f_d2d5_4ae8_a077_217c600588d8*mw2e632a32_3823_4933_95cb_19567cbcc66a
mwc85f8d37_7f39_41b2_8ea4_00b5adad2eac = 0.07934338; mw807b9a99_fb16_421f_b724_69f29f3fcfb2 = 1.9895374Reaction: mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + VIIIa => mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43; mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, VIIIa, mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43, mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, VIIIa, mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43, Rate Law: mwc85f8d37_7f39_41b2_8ea4_00b5adad2eac*mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f*VIIIa-mw807b9a99_fb16_421f_b724_69f29f3fcfb2*mw8bdbd17d_f542_4b8c_88c6_a82eaf997a43
k38 = 1.0556718E-6Reaction: Xa + ATIII => Xa_ATIII; Xa, ATIII, Xa, ATIII, Rate Law: k38*Xa*ATIII
k11 = 9.5; k12 = 0.032999929Reaction: TF_VIIa + Xa => TF_VIIa_Xa; TF_VIIa, Xa, TF_VIIa_Xa, TF_VIIa, Xa, TF_VIIa_Xa, Rate Law: k12*TF_VIIa*Xa-k11*TF_VIIa_Xa
mw0e80d629_98c1_44a6_bd57_3a4027c87b4c = 2.0869571; mw70d2f292_be41_4999_99cb_9c146808db85 = 0.077518002Reaction: mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + IXa_VIIIa => mwa4fcfa0c_6944_42fc_8c74_7865f13953c8; mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, IXa_VIIIa, mwa4fcfa0c_6944_42fc_8c74_7865f13953c8, mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, IXa_VIIIa, mwa4fcfa0c_6944_42fc_8c74_7865f13953c8, Rate Law: mw70d2f292_be41_4999_99cb_9c146808db85*mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f*IXa_VIIIa-mw0e80d629_98c1_44a6_bd57_3a4027c87b4c*mwa4fcfa0c_6944_42fc_8c74_7865f13953c8
mwaec203ce_06d5_4003_bfdb_7244d3d77255 = 0.0011427258Reaction: mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b => IXa + Xa; mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b, mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b, Rate Law: mwaec203ce_06d5_4003_bfdb_7244d3d77255*mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b
k19 = 0.11749508; k18 = 0.0050724996Reaction: IXa + VIIIa => IXa_VIIIa; IXa, VIIIa, IXa_VIIIa, IXa, VIIIa, IXa_VIIIa, Rate Law: k19*IXa*VIIIa-k18*IXa_VIIIa
k5 = 3.3894832E-4Reaction: TF_VIIa + VII => TF_VIIa + VIIa; TF_VIIa, VII, TF_VIIa, VII, Rate Law: k5*TF_VIIa*VII
k42 = 3.2905257E-7Reaction: TF_VIIa + ATIII => TF_VIIa_ATIII; TF_VIIa, ATIII, TF_VIIa, ATIII, Rate Law: k42*TF_VIIa*ATIII
k37 = 0.025386917Reaction: TF_VIIa + Xa_TFPI => TF_VIIa_Xa_TFPI; TF_VIIa, Xa_TFPI, TF_VIIa, Xa_TFPI, Rate Law: k37*TF_VIIa*Xa_TFPI
k27 = 4.0233556E-4Reaction: IIa + V => IIa + Va; IIa, V, IIa, V, Rate Law: k27*IIa*V
k13 = 20.6708; k14 = 0.010569458Reaction: TF_VIIa + IX => TF_VIIa_IX; TF_VIIa, IX, TF_VIIa_IX, TF_VIIa, IX, TF_VIIa_IX, Rate Law: k14*TF_VIIa*IX-k13*TF_VIIa_IX
mwb01ef86f_18d8_45e7_a452_31878dcb3d49 = 30.668349; mwc0cb654e_d95f_4d4b_8dc2_3a21afd35a19 = 0.13081564Reaction: mw6591152c_8b5a_4c9b_b095_956988a01ba0 + IX => mwe70b2c96_44b9_48eb_967a_7eb850a916a6; mw6591152c_8b5a_4c9b_b095_956988a01ba0, IX, mwe70b2c96_44b9_48eb_967a_7eb850a916a6, mw6591152c_8b5a_4c9b_b095_956988a01ba0, IX, mwe70b2c96_44b9_48eb_967a_7eb850a916a6, Rate Law: mwc0cb654e_d95f_4d4b_8dc2_3a21afd35a19*mw6591152c_8b5a_4c9b_b095_956988a01ba0*IX-mwb01ef86f_18d8_45e7_a452_31878dcb3d49*mwe70b2c96_44b9_48eb_967a_7eb850a916a6
k7 = 1.1527134E-5Reaction: IIa + VII => IIa + VIIa; IIa, VII, IIa, VII, Rate Law: k7*IIa*VII
mw7b89687a_3110_4d5f_a9ec_7ca8761f0d41 = 84.659935; mw05b4111c_4463_4be0_aa1e_5a8f50c7bf67 = 0.059664002Reaction: VIIa + X => mw6d041b25_87db_4394_9b8b_7ac61e01f359; VIIa, X, mw6d041b25_87db_4394_9b8b_7ac61e01f359, VIIa, X, mw6d041b25_87db_4394_9b8b_7ac61e01f359, Rate Law: mw05b4111c_4463_4be0_aa1e_5a8f50c7bf67*VIIa*X-mw7b89687a_3110_4d5f_a9ec_7ca8761f0d41*mw6d041b25_87db_4394_9b8b_7ac61e01f359
k39 = 3.55E-6Reaction: mIIa + ATIII => mIIa_ATIII; mIIa, ATIII, mIIa, ATIII, Rate Law: k39*mIIa*ATIII
mw4d2fe532_2ccd_42c4_9b4b_759022a87484 = 1.4001578; mwb63aa5ed_b6d8_4241_9987_54828945aea3 = 0.1289308Reaction: mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + IXa_VIIIa_X => mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca; mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, IXa_VIIIa_X, mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca, mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, IXa_VIIIa_X, mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca, Rate Law: mwb63aa5ed_b6d8_4241_9987_54828945aea3*mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f*IXa_VIIIa_X-mw4d2fe532_2ccd_42c4_9b4b_759022a87484*mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca
mw44adf04a_f1e2_4ca9_9615_5a9f4d3bbea8 = 0.13304333; mwc189e7ea_7518_4a4f_be0f_03f2d073b29e = 83.206626Reaction: IXa + X => mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b; IXa, X, mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b, IXa, X, mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b, Rate Law: mw44adf04a_f1e2_4ca9_9615_5a9f4d3bbea8*IXa*X-mwc189e7ea_7518_4a4f_be0f_03f2d073b29e*mw64e9cef3_5dd3_43f3_ad04_58e8fc07a91b
mw7be1d52f_926f_47e0_964b_d3303c8453b1 = 0.05Reaction: mw6d041b25_87db_4394_9b8b_7ac61e01f359 + mw6591152c_8b5a_4c9b_b095_956988a01ba0 => VIIa + Xa + mw6591152c_8b5a_4c9b_b095_956988a01ba0; mw6d041b25_87db_4394_9b8b_7ac61e01f359, mw6591152c_8b5a_4c9b_b095_956988a01ba0, mw6d041b25_87db_4394_9b8b_7ac61e01f359, mw6591152c_8b5a_4c9b_b095_956988a01ba0, Rate Law: mw7be1d52f_926f_47e0_964b_d3303c8453b1*mw6d041b25_87db_4394_9b8b_7ac61e01f359*mw6591152c_8b5a_4c9b_b095_956988a01ba0
mwaf2c7981_908c_4f4c_898e_2491a9f04e17 = 0.10523968; mw1ddc2a05_bc78_4434_a2d9_d06701483346 = 19.338228Reaction: mwa6be116e_72f1_439e_bca6_eb61f79cc68e + mw6a8501d2_9479_41ae_8616_1e8d0e1bbfa9 => mw3cec90c2_500e_4f30_b6be_325ef5194755; mwa6be116e_72f1_439e_bca6_eb61f79cc68e, mw6a8501d2_9479_41ae_8616_1e8d0e1bbfa9, mw3cec90c2_500e_4f30_b6be_325ef5194755, mwa6be116e_72f1_439e_bca6_eb61f79cc68e, mw6a8501d2_9479_41ae_8616_1e8d0e1bbfa9, mw3cec90c2_500e_4f30_b6be_325ef5194755, Rate Law: mwaf2c7981_908c_4f4c_898e_2491a9f04e17*mwa6be116e_72f1_439e_bca6_eb61f79cc68e*mw6a8501d2_9479_41ae_8616_1e8d0e1bbfa9-mw1ddc2a05_bc78_4434_a2d9_d06701483346*mw3cec90c2_500e_4f30_b6be_325ef5194755
k41 = 3.917682E-6Reaction: IIa + ATIII => IIa_ATIII; IIa, ATIII, IIa, ATIII, Rate Law: k41*IIa*ATIII
mwd6b996b1_d7fe_42de_b17e_b2482109c54d = 0.1043597; mwc5dc3645_536d_4bb4_88c7_4aeac4f5a241 = 2.0649128Reaction: mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + Va => mw2e632a32_3823_4933_95cb_19567cbcc66a; mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, Va, mw2e632a32_3823_4933_95cb_19567cbcc66a, mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f, Va, mw2e632a32_3823_4933_95cb_19567cbcc66a, Rate Law: mwd6b996b1_d7fe_42de_b17e_b2482109c54d*mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f*Va-mwc5dc3645_536d_4bb4_88c7_4aeac4f5a241*mw2e632a32_3823_4933_95cb_19567cbcc66a
k33 = 1.801577E-4; k34 = 4.5E-4Reaction: Xa + TFPI => Xa_TFPI; Xa, TFPI, Xa_TFPI, Xa, TFPI, Xa_TFPI, Rate Law: k34*Xa*TFPI-k33*Xa_TFPI
mw95e328a0_be5b_4260_b6e4_d85c4c4aae9e = 0.050084768; mw9bcd5c0b_3384_4d5e_92ce_70b13d64e8b8 = 0.11573051Reaction: IIa + mwd68cbf38_9266_4dfb_aa00_f817c3421aec => mwa6be116e_72f1_439e_bca6_eb61f79cc68e; IIa, mwd68cbf38_9266_4dfb_aa00_f817c3421aec, mwa6be116e_72f1_439e_bca6_eb61f79cc68e, IIa, mwd68cbf38_9266_4dfb_aa00_f817c3421aec, mwa6be116e_72f1_439e_bca6_eb61f79cc68e, Rate Law: mw9bcd5c0b_3384_4d5e_92ce_70b13d64e8b8*IIa*mwd68cbf38_9266_4dfb_aa00_f817c3421aec-mw95e328a0_be5b_4260_b6e4_d85c4c4aae9e*mwa6be116e_72f1_439e_bca6_eb61f79cc68e
k9 = 0.036245656; k8 = 1.3800407Reaction: TF_VIIa + X => TF_VIIa_X; TF_VIIa, X, TF_VIIa_X, TF_VIIa, X, TF_VIIa_X, Rate Law: k9*TF_VIIa*X-k8*TF_VIIa_X
k31 = 29.479266Reaction: Xa_Va_II => Xa_Va + mIIa + mwbdb849d8_2b25_4551_8de8_adc8bead2303; Xa_Va_II, Xa_Va_II, Rate Law: k31*Xa_Va_II
mwa4cc6bbe_c310_445f_bba7_a94868342831 = 10740.276; mw3b48c5e7_774a_4dc4_917f_8f8cff8d9c4b = 90.211653Reaction: IIa + mwd3e1ba39_ab10_4702_addd_fb6a7e184a4b => IIa + mwfa9d903a_b5e5_4a38_a649_dfe4719577aa; IIa, mwd3e1ba39_ab10_4702_addd_fb6a7e184a4b, IIa, mwd3e1ba39_ab10_4702_addd_fb6a7e184a4b, Rate Law: mw3b48c5e7_774a_4dc4_917f_8f8cff8d9c4b*IIa*mwd3e1ba39_ab10_4702_addd_fb6a7e184a4b/(mwa4cc6bbe_c310_445f_bba7_a94868342831+mwd3e1ba39_ab10_4702_addd_fb6a7e184a4b)
mw6b555ed1_194e_4fa4_9688_8105aa7c60c0 = 0.013215482Reaction: mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca => mwedf22864_05a0_40c3_a0d5_ede45a3e7e8f + IXa + X + mwf5c3f9df_7ccf_4ca7_b241_471a66720da8; mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca, mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca, Rate Law: mw6b555ed1_194e_4fa4_9688_8105aa7c60c0*mwe0bb059d_deaa_45fa_b7dc_ec1c4409c4ca
mwec1b7289_5544_4c2b_b9f6_bf6524cabda5 = 3.15; mwaa306898_0d0f_4748_b48a_fcd56bdc0b16 = 0.15Reaction: Xa + VIII => mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820; Xa, VIII, mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820, Xa, VIII, mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820, Rate Law: mwaa306898_0d0f_4748_b48a_fcd56bdc0b16*Xa*VIII-mwec1b7289_5544_4c2b_b9f6_bf6524cabda5*mw7a1594c9_f04f_478c_9f5f_ccbe0b95a820

States:

NameDescription
IXIX
mwa4fcfa0c 6944 42fc 8c74 7865f13953c8APC_IXa_VIIIa
mwedf22864 05a0 40c3 a0d5 ede45a3e7e8fAPC
mw6a8501d2 9479 41ae 8616 1e8d0e1bbfa9PC
ATIIIATIII
Xa Va IIXa_Va_II
mw931f65a6 3967 4ac2 9904 ba791b216fc2F12_deg
XaXa
mwf5c3f9df 7ccf 4ca7 b241 471a66720da8VIIIa_deg
TF VIIa XTF_VIIa_X
TF VIIa XaTF_VIIa_Xa
XX
Xa VaXa_Va
mwe0bb059d deaa 45fa b7dc ec1c4409c4caAPC_IXa_VIIIa_X
mw18e5caa7 26eb 4521 b217 da75bb3193adVa_deg
mw7a1594c9 f04f 478c 9f5f ccbe0b95a820Xa_VIII
TF VIIaTF_VIIa
VIIIaVIIIa
VaVa
IIaIIa
Xa TFPIXa_TFPI
VIIaVIIa
IXa VIIIa XIXa_VIIIa_X
TF VIIa IXTF_VIIa_IX
mwd68cbf38 9266 4dfb aa00 f817c3421aecTmod
mw3cec90c2 500e 4f30 b6be 325ef5194755IIa_Tmod_PC
IXaIXa
mwbdb849d8 2b25 4551 8de8 adc8bead2303F12
IXa VIIIaIXa_VIIIa
IIII
mwa6be116e 72f1 439e bca6 eb61f79cc68eIIa_Tmod
mw2e632a32 3823 4933 95cb 19567cbcc66aAPC_Va

Nazaret2008_Dynnik1980_CarbohydrateEnergyMetabolism: MODEL1202170000v0.0.1

This model is from the article: An old paper revisited: ‘‘A mathematical model of carbohydrate energy metabolism. Int…

Details

We revisit an old Russian paper by V.V. Dynnik, R. Heinrich and E.E. Sel'kov (1980a,b) describing: "A mathematical model of carbohydrate energy metabolism. Interaction between glycolysis, the Krebs cycle and the H-transporting shuttles at varying ATPases load". We analyse the model mathematically and calculate the control coefficients as a function of ATPase loads. We also evaluate the structure of the metabolic network in terms of elementary flux modes. We show how this model can respond to an ATPase load as well as to the glucose supply. We also show how this simple model can help in understanding the articulation between the major blocks of energetic metabolism, i.e. glycolysis, the Krebs cycle and the H-transporting shuttles. link: http://identifiers.org/pubmed/18304584

Nazaret2009_TCA_RC_ATP: BIOMD0000000232v0.0.1

This a model from the article: Mitochondrial energetic metabolism: a simplified model of TCA cycle with ATP production…

Details

Mitochondria play a central role in cellular energetic metabolism. The essential parts of this metabolism are the tricarboxylic acid (TCA) cycle, the respiratory chain and the adenosine triphosphate (ATP) synthesis machinery. Here a simplified model of these three metabolic components with a limited set of differential equations is presented. The existence of a steady state is demonstrated and results of numerical simulations are presented. The relevance of a simple model to represent actual in vivo behavior is discussed. link: http://identifiers.org/pubmed/19007794

Parameters:

NameDescription
At = 4.16 millimolarReaction: ADP = At-ATP, Rate Law: missing
k2=0.152 per millimolar per secondReaction: Pyr + NAD => AcCoA + NADH, Rate Law: mitochondrion*k2*Pyr*NAD
JANT = NaN millimolar per secondReaction: ATP => ADP, Rate Law: mitochondrion*JANT
k8=3.6 per secondReaction: OAA =>, Rate Law: mitochondrion*k8*OAA
k7=0.04 per millimolar per secondReaction: Pyr + ATP => OAA + ADP, Rate Law: mitochondrion*k7*Pyr*ATP
Keq=0.3975 dimensionless; k6=0.0032 per secondReaction: OAA => KG, Rate Law: mitochondrion*k6*(OAA-KG/Keq)
k3=57.142 per millimolar per secondReaction: OAA + AcCoA => Cit, Rate Law: mitochondrion*k3*OAA*AcCoA
Jresp = NaN millimolar per secondReaction: NADH + O2 + H => NAD + H2O + He, Rate Law: mitochondrion*Jresp
Nt = 1.07 millimolarReaction: NADH = Nt-NAD, Rate Law: missing
k1=0.038 millimolar per secondReaction: => Pyr, Rate Law: mitochondrion*k1
k5=0.082361 per millimolar squared per second; At = 4.16 millimolarReaction: KG + ADP + NAD => OAA + ATP + NADH, Rate Law: mitochondrion*k5*KG*NAD*(At-ATP)
k4=0.053 per millimolar per secondReaction: Cit + NAD => KG + NADH, Rate Law: mitochondrion*k4*Cit*NAD
JATP = NaN millimolar per secondReaction: ADP + iP + He => ATP + H2O + H, Rate Law: mitochondrion*JATP
Jleak = NaN millimolar per secondReaction: He => H, Rate Law: mitochondrion*Jleak

States:

NameDescription
O2[dioxygen; Oxygen]
iP[phosphate(3-); Orthophosphate]
ATP[ATP; ATP]
NADH[NADH; NADH]
Cit[citrate(3-); Citrate]
Pyr[pyruvate; Pyruvate]
AcCoA[acetyl-CoA; Acetyl-CoA]
H2O[water; H2O]
OAA[oxaloacetate(2-); Oxaloacetate]
ADP[ADP; ADP]
He[proton; H+]
NAD[NAD(+); NAD+]
H[proton; H+]
KG[2-oxoglutarate(2-); 2-Oxoglutarate]

Nazari2018 - IL6 mediated stem cell driven tumor growth and targeted treatment: BIOMD0000000819v0.0.1

This a model from the article: A mathematical model for IL-6-mediated, stem cell driven tumor growth and targeted trea…

Details

Targeting key regulators of the cancer stem cell phenotype to overcome their critical influence on tumor growth is a promising new strategy for cancer treatment. Here we present a modeling framework that operates at both the cellular and molecular levels, for investigating IL-6 mediated, cancer stem cell driven tumor growth and targeted treatment with anti-IL6 antibodies. Our immediate goal is to quantify the influence of IL-6 on cancer stem cell self-renewal and survival, and to characterize the subsequent impact on tumor growth dynamics. By including the molecular details of IL-6 binding, we are able to quantify the temporal changes in fractional occupancies of bound receptors and their influence on tumor volume. There is a strong correlation between the model output and experimental data for primary tumor xenografts. We also used the model to predict tumor response to administration of the humanized IL-6R monoclonal antibody, tocilizumab (TCZ), and we found that as little as 1mg/kg of TCZ administered weekly for 7 weeks is sufficient to result in tumor reduction and a sustained deceleration of tumor growth. link: http://identifiers.org/pubmed/29351275

Parameters:

NameDescription
R_Td = 2.075E-7; P_DD = 0.0133297534723066Reaction: => IL_6R_on_D, Rate Law: compartment*R_Td*P_DD
K_f = 2.35 1/(fmol*d)Reaction: => IL_6__Cell_bound_IL_6R_complex_on_E; IL_6__L, IL_6R_on_E, Rate Law: compartment*K_f*IL_6__L*IL_6R_on_E
P_S = 0.899999967997301; alpha_S = 0.6 1/dReaction: => Cancer_Stem_Cell_S, Rate Law: compartment*alpha_S*P_S*Cancer_Stem_Cell_S
R_Ts = 1.66E-6 fmol; P_phiS = 539.99998079838Reaction: => IL_6R_on_S, Rate Law: compartment*R_Ts*P_phiS
A_out = 2.0; alpha_E = 0.666487673615332 1/dReaction: => Differentiated_tumor_cell_D; Progenitor_tumor_cell_E, Rate Law: compartment*A_out*alpha_E*Progenitor_tumor_cell_E
delta_D = 0.0612 1/d; phi_D = 0.0; gamma_D = 2.38Reaction: Differentiated_tumor_cell_D =>, Rate Law: compartment*delta_D*Differentiated_tumor_cell_D/(1+gamma_D*phi_D)
K_r = 2.24 1/dReaction: IL_6__Cell_bound_IL_6R_complex_on_D => IL_6__L + IL_6R_on_D; IL_6__Cell_bound_IL_6R_complex_on_D, Rate Law: compartment*K_r*IL_6__Cell_bound_IL_6R_complex_on_D
lambda = 0.4152 1/dReaction: IL_6__L =>, Rate Law: compartment*lambda*IL_6__L
R_Te = 2.075E-7; P_etaE = 119.993373526503Reaction: => IL_6R_on_E, Rate Law: compartment*R_Te*P_etaE
R_Te = 2.075E-7; D_etaE = 6.12E-4Reaction: IL_6__Cell_bound_IL_6R_complex_on_E => ; IL_6R_on_E, Rate Law: compartment*IL_6__Cell_bound_IL_6R_complex_on_E*R_Te*D_etaE/(IL_6R_on_E+IL_6__Cell_bound_IL_6R_complex_on_E)
R_Td = 2.075E-7; D_DD = 6.12E-4Reaction: IL_6R_on_D => ; IL_6__Cell_bound_IL_6R_complex_on_D, Rate Law: compartment*IL_6R_on_D*R_Td*D_DD/(IL_6R_on_D+IL_6__Cell_bound_IL_6R_complex_on_D)
R_Ts = 1.66E-6 fmol; D_phiS = 12.6Reaction: IL_6__Cell_bound_IL_6R_complex_on_S => ; IL_6R_on_S, Rate Law: compartment*IL_6__Cell_bound_IL_6R_complex_on_S*R_Ts*D_phiS/(IL_6R_on_S+IL_6__Cell_bound_IL_6R_complex_on_S)
gamma_S = 2.38; phi_S = 0.0; delta_S = 0.0126 1/dReaction: Cancer_Stem_Cell_S =>, Rate Law: compartment*delta_S*Cancer_Stem_Cell_S/(1+gamma_S*phi_S)
alpha_E = 0.666487673615332 1/dReaction: Progenitor_tumor_cell_E =>, Rate Law: compartment*alpha_E*Progenitor_tumor_cell_E
P_S = 0.899999967997301; alpha_S = 0.6 1/d; A_in = 2.0Reaction: => Progenitor_tumor_cell_E; Cancer_Stem_Cell_S, Rate Law: compartment*A_in*alpha_S*(1-P_S)*Cancer_Stem_Cell_S
K_p = 24.95 1/dReaction: IL_6__Cell_bound_IL_6R_complex_on_D => IL_6R_on_D; IL_6__Cell_bound_IL_6R_complex_on_D, Rate Law: compartment*K_p*IL_6__Cell_bound_IL_6R_complex_on_D
delta_E = 0.0612 1/d; gamma_E = 2.38; phi_E = 0.0Reaction: Progenitor_tumor_cell_E =>, Rate Law: compartment*delta_E*Progenitor_tumor_cell_E/(1+gamma_E*phi_E)
rho = 7.0E-7 fmol/dReaction: => IL_6__L; Cancer_Stem_Cell_S, Progenitor_tumor_cell_E, Differentiated_tumor_cell_D, Rate Law: compartment*rho*(Cancer_Stem_Cell_S+Progenitor_tumor_cell_E+Differentiated_tumor_cell_D)

States:

NameDescription
IL 6 Cell bound IL 6R complex on S[Receptor; Interleukin-6; Interleukin-6; Cancer Stem Cell]
IL 6 Cell bound IL 6R complex on E[Interleukin-6; Receptor; Interleukin-6; Ancestor]
tumortumor
Cancer Stem Cell S[Head and Neck Squamous Cell Carcinoma; head and neck squamous cell carcinoma; Cancer Stem Cell]
Progenitor tumor cell E[head and neck squamous cell carcinoma; Head and Neck Squamous Cell Carcinoma; Ancestor]
IL 6R on S[Receptor; Interleukin-6 receptor subunit alpha; Interleukin-6; Cancer Stem Cell]
Differentiated tumor cell D[head and neck squamous cell carcinoma; Head and Neck Squamous Cell Carcinoma; Interleukin-6; differentiated]
IL 6 Cell bound IL 6R complex on D[Interleukin-6; Interleukin-6; Receptor; differentiated]
IL 6R on E[Interleukin-6; Receptor; Interleukin-6 receptor subunit alpha; Ancestor]
IL 6R on D[Interleukin-6; Receptor; Interleukin-6 receptor subunit alpha; differentiated]
IL 6 L[Interleukin-6]

Ndairou2020 - early-stage transmission dynamics of COVID-19 in Wuhan: BIOMD0000000958v0.0.1

We propose a compartmental mathematical model for the spread of the COVID-19 disease with special focus on the transmiss…

Details

We propose a compartmental mathematical model for the spread of the COVID-19 disease with special focus on the transmissibility of super-spreaders individuals. We compute the basic reproduction number threshold, we study the local stability of the disease free equilibrium in terms of the basic reproduction number, and we investigate the sensitivity of the model with respect to the variation of each one of its parameters. Numerical simulations show the suitability of the proposed COVID-19 model for the outbreak that occurred in Wuhan, China. link: http://identifiers.org/pubmed/32341628

Ndii2015-transmission dynamics of dengue in the presence of Wolbachia.: MODEL2003160002v0.0.1

Use of the bacterium Wolbachia is an innovative new strategy designed to break the cycle of dengue transmission. There a…

Details

Use of the bacterium Wolbachia is an innovative new strategy designed to break the cycle of dengue transmission. There are two main mechanisms by which Wolbachia could achieve this: by reducing the level of dengue virus in the mosquito and/or by shortening the host mosquito's lifespan. However, although Wolbachia shortens the lifespan, it also gives a breeding advantage which results in complex population dynamics. This study focuses on the development of a mathematical model to quantify the effect on human dengue cases of introducing Wolbachia into the mosquito population. The model consists of a compartment-based system of first-order differential equations; seasonal forcing in the mosquito population is introduced through the adult mosquito death rate. The analysis focuses on a single dengue outbreak typical of a region with a strong seasonally-varying mosquito population. We found that a significant reduction in human dengue cases can be obtained provided that Wolbachia-carrying mosquitoes persist when competing with mosquitoes without Wolbachia. Furthermore, using the Wolbachia strain WMel reduces the mosquito lifespan by at most 10% and allows them to persist in competition with non-Wolbachia-carrying mosquitoes. Mosquitoes carrying the WMelPop strain, however, are not likely to persist as it reduces the mosquito lifespan by up to 50%. When all other effects of Wolbachia on the mosquito physiology are ignored, cytoplasmic incompatibility alone results in a reduction in the number of human dengue cases. A sensitivity analysis of the parameters in the model shows that the transmission probability, the biting rate and the average adult mosquito death rate are the most important parameters for the outcome of the cumulative proportion of human individuals infected with dengue. link: http://identifiers.org/pubmed/25645184

Nelson1995_HIV1therapy_DIVadministraion_ModelA: MODEL1006230081v0.0.1

This a model from the article: Modeling defective interfering virus therapy for AIDS: conditions for DIV survival. N…

Details

The administration of a genetically engineered defective interfering virus (DIV) that interferes with HIV-1 replication has been proposed as a therapy for HIV-1 infection and AIDS. The proposed interfering virus, which is designed to superinfect HIV-1 infected cells, carries ribozymes that cleave conserved regions in HIV-1 RNA that code for the viral envelope protein. Thus DIV infection of HIV-1 infected cells should reduce or eliminate viral production by these cells. The success of this therapeutic strategy will depend both on the intercellular interaction of DIV and HIV-1, and on the overall dynamics of virus and T cells in the body. To study these dynamical issues, we have constructed a mathematical model of the interaction of HIV-1, DIV, and CD4+ cells in vivo. The results of both mathematical analysis and numerical simulation indicate that survival of the engineered DIV purely on a peripheral blood HIV-1 infection is unlikely. However, analytical results indicate that DIV might well survive on HIV-1 infected CD4+ cells in lymphoid organs such as lymph nodes and spleen, or on other HIV-1 infected cells in these organs. link: http://identifiers.org/pubmed/7881191

Nelson1995_HIV1therapy_DIVadministraion_ModelB: MODEL1006230033v0.0.1

This a model from the article: Modeling defective interfering virus therapy for AIDS: conditions for DIV survival. N…

Details

The administration of a genetically engineered defective interfering virus (DIV) that interferes with HIV-1 replication has been proposed as a therapy for HIV-1 infection and AIDS. The proposed interfering virus, which is designed to superinfect HIV-1 infected cells, carries ribozymes that cleave conserved regions in HIV-1 RNA that code for the viral envelope protein. Thus DIV infection of HIV-1 infected cells should reduce or eliminate viral production by these cells. The success of this therapeutic strategy will depend both on the intercellular interaction of DIV and HIV-1, and on the overall dynamics of virus and T cells in the body. To study these dynamical issues, we have constructed a mathematical model of the interaction of HIV-1, DIV, and CD4+ cells in vivo. The results of both mathematical analysis and numerical simulation indicate that survival of the engineered DIV purely on a peripheral blood HIV-1 infection is unlikely. However, analytical results indicate that DIV might well survive on HIV-1 infected CD4+ cells in lymphoid organs such as lymph nodes and spleen, or on other HIV-1 infected cells in these organs. link: http://identifiers.org/pubmed/7881191

Nelson2000- HIV-1 general model 1: BIOMD0000000875v0.0.1

This is the general model without delay described by the equation system (1) in: **A model of HIV-1 pathogenesis that in…

Details

Mathematical modeling combined with experimental measurements have yielded important insights into HIV-1 pathogenesis. For example, data from experiments in which HIV-infected patients are given potent antiretroviral drugs that perturb the infection process have been used to estimate kinetic parameters underlying HIV infection. Many of the models used to analyze data have assumed drug treatments to be completely efficacious and that upon infection a cell instantly begins producing virus. We consider a model that allows for less then perfect drug effects and which includes a delay in the initiation of virus production. We present detailed analysis of this delay differential equation model and compare the results to a model without delay. Our analysis shows that when drug efficacy is less than 100%, as may be the case in vivo, the predicted rate of decline in plasma virus concentration depends on three factors: the death rate of virus producing cells, the efficacy of therapy, and the length of the delay. Thus, previous estimates of infected cell loss rates can be improved upon by considering more realistic models of viral infection. link: http://identifiers.org/pubmed/10701304

Parameters:

NameDescription
k = 3.43E-8 l/(s*#)Reaction: T => T_i; V_I, Rate Law: plasma*k*V_I*T
delta = 0.5 1/msReaction: T_i =>, Rate Law: plasma*delta*T_i
np = 0.5 1; N = 480.0 1; delta = 0.5 1/msReaction: => V_I; T_i, Rate Law: plasma*(1-np)*N*delta*T_i
delta1 = 0.03 1/msReaction: T =>, Rate Law: plasma*delta1*T
c = 3.0 1/msReaction: V_I =>, Rate Law: plasma*c*V_I
lambda = 10.0 #/(l*s)Reaction: => T, Rate Law: plasma*lambda

States:

NameDescription
V NIV_NI
T[uninfected]
T i[infected cell]
V I[C14283]

Nelson2000_HIV-1_intracellular_delay: MODEL8102792069v0.0.1

described in: **A model of HIV-1 pathogenesis that includes an intracellular delay.** Nelson PW, Murray JD, Perelson A…

Details

Mathematical modeling combined with experimental measurements have yielded important insights into HIV-1 pathogenesis. For example, data from experiments in which HIV-infected patients are given potent antiretroviral drugs that perturb the infection process have been used to estimate kinetic parameters underlying HIV infection. Many of the models used to analyze data have assumed drug treatments to be completely efficacious and that upon infection a cell instantly begins producing virus. We consider a model that allows for less then perfect drug effects and which includes a delay in the initiation of virus production. We present detailed analysis of this delay differential equation model and compare the results to a model without delay. Our analysis shows that when drug efficacy is less than 100%, as may be the case in vivo, the predicted rate of decline in plasma virus concentration depends on three factors: the death rate of virus producing cells, the efficacy of therapy, and the length of the delay. Thus, previous estimates of infected cell loss rates can be improved upon by considering more realistic models of viral infection. link: http://identifiers.org/pubmed/10701304

Neumann2010_CD95Stimulation_NFkB_Apoptosis: BIOMD0000000243v0.0.1

This is the reduced model (model 8) described in: **Dynamics within the CD95 death-inducing signaling complex decide lif…

Details

This study explores the dilemma in cellular signaling that triggering of CD95 (Fas/APO-1) in some situations results in cell death and in others leads to the activation of NF-kappaB. We established an integrated kinetic mathematical model for CD95-mediated apoptotic and NF-kappaB signaling. Systematic model reduction resulted in a surprisingly simple model well approximating experimentally observed dynamics. The model postulates a new link between c-FLIP(L) cleavage in the death-inducing signaling complex (DISC) and the NF-kappaB pathway. We validated experimentally that CD95 stimulation resulted in an interaction of p43-FLIP with the IKK complex followed by its activation. Furthermore, we showed that the apoptotic and NF-kappaB pathways diverge already at the DISC. Model and experimental analysis of DISC formation showed that a subtle balance of c-FLIP(L) and procaspase-8 determines life/death decisions in a nonlinear manner. We present an integrated model describing the complex dynamics of CD95-mediated apoptosis and NF-kappaB signaling. link: http://identifiers.org/pubmed/20212524

Parameters:

NameDescription
k3 = 0.6693316Reaction: L_RF + FL => L_RF_FL, Rate Law: default*k3*L_RF*FL
k2 = 1.277248E-4Reaction: L_RF + C8 => L_RF_C8, Rate Law: default*k2*L_RF*C8
k10 = 0.1205258Reaction: C8 + C3_star => p43_p41 + C3_star, Rate Law: default*k10*C8*C3_star
k5 = 5.946569E-4Reaction: L_RF_FS + C8 => L_RF_C8_FS, Rate Law: default*k5*L_RF_FS*C8
k4 = 1.0E-5Reaction: L_RF + FS => L_RF_FS, Rate Law: default*k4*L_RF*FS
k9 = 0.002249759Reaction: C3 + C8_star => C3_star + C8_star, Rate Law: default*k9*C3*C8_star
k16 = 0.02229912Reaction: p43_FLIP_IKK_star =>, Rate Law: default*k16*p43_FLIP_IKK_star
k14 = 0.3588224Reaction: NF_kB_IkB + p43_FLIP_IKK_star => NF_kB_IkB_P + p43_FLIP_IKK_star, Rate Law: default*k14*NF_kB_IkB*p43_FLIP_IKK_star
k7 = 0.8875063Reaction: L_RF_FL + FS => L_RF_FL_FS, Rate Law: default*k7*L_RF_FL*FS
k13 = 7.204261E-4Reaction: p43_FLIP + IKK => p43_FLIP_IKK_star, Rate Law: default*k13*p43_FLIP*IKK
k11 = 0.02891451Reaction: C8_star =>, Rate Law: default*k11*C8_star
k17 = 0.0064182Reaction: NF_kB_star =>, Rate Law: default*k17*NF_kB_star
k12 = 0.1502914Reaction: C3_star =>, Rate Law: default*k12*C3_star
k1 = 1.0Reaction: L + RF => L_RF, Rate Law: default*k1*L*RF
k6 = 0.9999999Reaction: L_RF_FS + FL => L_RF_FL_FS, Rate Law: default*k6*L_RF_FS*FL
k15 = 3.684162Reaction: NF_kB_IkB_P => NF_kB_star, Rate Law: default*k15*NF_kB_IkB_P
k8 = 8.044378E-4Reaction: p43_p41 + p43_p41 => C8_star, Rate Law: default*k8*p43_p41*p43_p41

States:

NameDescription
C3 star[Caspase-3]
L RF FL[CD95 ligand; CASP8 and FADD-like apoptosis regulator; FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]
C3[Caspase-3]
p43 FLIP[CASP8 and FADD-like apoptosis regulator]
L[CD95 ligand]
IKK[NF-kappa-B essential modulator; Inhibitor of nuclear factor kappa-B kinase subunit beta; Inhibitor of nuclear factor kappa-B kinase subunit alpha]
p43 p41[Caspase-8]
C8[Caspase-8]
L RF C8[CD95 ligand; Caspase-8; Tumor necrosis factor receptor superfamily member 6; FAS-associated death domain protein]
FL[CASP8 and FADD-like apoptosis regulator]
NF kB star[NF-kappaB complex; Nuclear factor NF-kappa-B p105 subunit]
p43 FLIP IKK starp43-FLIP:IKK*
L RF FS[CD95 ligand; CASP8 and FADD-like apoptosis regulator; FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]
NF kB IkB[IkBs:NFkB [cytosol]; NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
C8 star[Caspase-8]
L RF[FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6; CD95 ligand]
L RF FL FS[CD95 ligand; CASP8 and FADD-like apoptosis regulator; FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]
L RF FL FL[CD95 ligand; CASP8 and FADD-like apoptosis regulator; FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]
FS[CASP8 and FADD-like apoptosis regulator]
NF kB IkB P[NFkB Complex [cytosol]; Phospho-NF-kappaB Inhibitor [cytosol]; NF-kappa-B inhibitor alpha; Nuclear factor NF-kappa-B p105 subunit]
L RF C8 FS[CD95 ligand; CASP8 and FADD-like apoptosis regulator; Caspase-8; FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]
RF[FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]
L RF FS FS[CD95 ligand; CASP8 and FADD-like apoptosis regulator; FAS-associated death domain protein; Tumor necrosis factor receptor superfamily member 6]

Neves2008 - Role of cell shape and size in controlling intracellular signalling: BIOMD0000000182v0.0.1

Neves2008 - Role of cell shape and size in controlling intracellular signallingThe role of cell shape and size in the fl…

Details

The role of cell size and shape in controlling local intracellular signaling reactions, and how this spatial information originates and is propagated, is not well understood. We have used partial differential equations to model the flow of spatial information from the beta-adrenergic receptor to MAPK1,2 through the cAMP/PKA/B-Raf/MAPK1,2 network in neurons using real geometries. The numerical simulations indicated that cell shape controls the dynamics of local biochemical activity of signal-modulated negative regulators, such as phosphodiesterases and protein phosphatases within regulatory loops to determine the size of microdomains of activated signaling components. The model prediction that negative regulators control the flow of spatial information to downstream components was verified experimentally in rat hippocampal slices. These results suggest a mechanism by which cellular geometry, the presence of regulatory loops with negative regulators, and key reaction rates all together control spatial information transfer and microdomain characteristics within cells. link: http://identifiers.org/pubmed/18485874

Parameters:

NameDescription
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_PPase_mek = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); Km=15.7 0.001*dimensionless*m^(-3)*molReaction: MEK_active_cyto => MEK_cyto; PP2A_cyto, Rate Law: Vmax_PPase_mek*0.00166112956810631*MEK_active_cyto*1/(Km+0.00166112956810631*MEK_active_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kf_AC_activation=500.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr_AC_activation=1.0 s^(-1)Reaction: G_a_s_cyto + AC_cyto_mem => AC_active_cyto_mem, Rate Law: (Kf_AC_activation*0.00166112956810631*G_a_s_cyto*AC_cyto_mem+(-Kr_AC_activation*AC_active_cyto_mem))*cyto_mem
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Km=0.77 0.001*dimensionless*m^(-3)*mol; Vmax_PPase_MAPK = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: MAPK_active_cyto => MAPK_cyto; PP2A_cyto, Rate Law: Vmax_PPase_MAPK*0.00166112956810631*MAPK_active_cyto*1/(Km+0.00166112956810631*MAPK_active_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kr=0.2 s^(-1); Kf=1.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: BAR_cyto_mem + iso_extra => iso_BAR_cyto_mem, Rate Law: (Kf*BAR_cyto_mem*0.00166112956810631*iso_extra+(-Kr*iso_BAR_cyto_mem))*cyto_mem
Vmax_PPase_Raf = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Km=15.7 0.001*dimensionless*m^(-3)*molReaction: B_Raf_active_cyto => B_Raf_cyto; PP2A_cyto, Rate Law: Vmax_PPase_Raf*0.00166112956810631*B_Raf_active_cyto*1/(Km+0.00166112956810631*B_Raf_active_cyto)*cyto*1*1/KMOLE
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_pde4_p_pde4_p = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); Km_pde4_p=1.3 0.001*dimensionless*m^(-3)*molReaction: cAMP_cyto => AMP_cyto; PDE4_P_cyto, Rate Law: Vmax_pde4_p_pde4_p*0.00166112956810631*cAMP_cyto*1/(Km_pde4_p+0.00166112956810631*cAMP_cyto)*cyto*1*1/KMOLE
Km_PDE4=1.3 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_PDE4_PDE4 = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: cAMP_cyto => AMP_cyto; PDE4_cyto, Rate Law: Vmax_PDE4_PDE4*0.00166112956810631*cAMP_cyto*1/(Km_PDE4+0.00166112956810631*cAMP_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Km_AC_active=32.0 0.001*dimensionless*m^(-3)*mol; Vmax_AC_active_AC_active = NaN item*μm^(-2)*s^(-1)Reaction: ATP_cyto => cAMP_cyto; AC_active_cyto_mem, Rate Law: Vmax_AC_active_AC_active*0.00166112956810631*ATP_cyto*1/(Km_AC_active+0.00166112956810631*ATP_cyto)*cyto_mem
Kf_G_binds_BAR=0.3 1000*dimensionless*m^3*mol^(-1)*s^(-1); I=0.0 dimensionless*A*m^(-2); Kr_G_binds_BAR=0.1 s^(-1)Reaction: BAR_cyto_mem + G_protein_cyto => BAR_G_cyto_mem, Rate Law: (Kf_G_binds_BAR*BAR_cyto_mem*0.00166112956810631*G_protein_cyto+(-Kr_G_binds_BAR*BAR_G_cyto_mem))*cyto_mem
Km=0.046296 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_MEK_activates_MAPK = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: MAPK_cyto => MAPK_active_cyto; MEK_active_cyto, Rate Law: Vmax_MEK_activates_MAPK*0.00166112956810631*MAPK_cyto*1/(Km+0.00166112956810631*MAPK_cyto)*cyto*1*1/KMOLE
Km=6.0 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_pp_ptp = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: PTP_PKA_cyto => PTP_cyto; PTP_PP_cyto, Rate Law: Vmax_pp_ptp*0.00166112956810631*PTP_PKA_cyto*1/(Km+0.00166112956810631*PTP_PKA_cyto)*cyto*1*1/KMOLE
Vmax_highKM_PDE = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); Km=15.0 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3)Reaction: cAMP_cyto => AMP_cyto; PDE_high_km_cyto, Rate Law: Vmax_highKM_PDE*0.00166112956810631*cAMP_cyto*1/(Km+0.00166112956810631*cAMP_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kr_activate_Gs=0.0 1000000*dimensionless*m^6*mol^(-2)*s^(-1); Kf_activate_Gs=0.025 s^(-1)Reaction: iso_BAR_G_cyto_mem => iso_BAR_cyto_mem + bg_cyto + G_a_s_cyto, Rate Law: (Kf_activate_Gs*iso_BAR_G_cyto_mem-Kr_activate_Gs*iso_BAR_cyto_mem*0.00166112956810631*bg_cyto*0.00166112956810631*G_a_s_cyto)*cyto_mem
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Km=0.15909 0.001*dimensionless*m^(-3)*mol; Vmax_Raf_activates_MEK = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: MEK_cyto => MEK_active_cyto; B_Raf_active_cyto, Rate Law: Vmax_Raf_activates_MEK*0.00166112956810631*MEK_cyto*1/(Km+0.00166112956810631*MEK_cyto)*cyto*1*1/KMOLE
Vmax_PKA_P_PDE = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); Km=0.5 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3)Reaction: PDE4_cyto => PDE4_P_cyto; PKA_cyto, Rate Law: Vmax_PKA_P_PDE*0.00166112956810631*PDE4_cyto*1/(Km+0.00166112956810631*PDE4_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Km_grk=15.0 item*μm^(-2); Vmax_grk_GRK = NaN item*μm^(-2)*s^(-1)Reaction: iso_BAR_cyto_mem => iso_BAR_p_cyto_mem; GRK_cyto, Rate Law: Vmax_grk_GRK*iso_BAR_cyto_mem*1/(Km_grk+iso_BAR_cyto_mem)*cyto_mem
Km=0.5 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_PKA_activates_Raf = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: B_Raf_cyto => B_Raf_active_cyto; PKA_cyto, Rate Law: Vmax_PKA_activates_Raf*0.00166112956810631*B_Raf_cyto*1/(Km+0.00166112956810631*B_Raf_cyto)*cyto*1*1/KMOLE
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Kr_GTPase=0.0 s^(-1); Kf_GTPase=0.06667 s^(-1)Reaction: G_a_s_cyto => G_GDP_cyto, Rate Law: (Kf_GTPase*0.00166112956810631*G_a_s_cyto+(-Kr_GTPase*0.00166112956810631*G_GDP_cyto))*cyto*1*1/KMOLE
Kr_G_binds_iso_BAR=0.1 s^(-1); I=0.0 dimensionless*A*m^(-2); Kf_G_binds_iso_BAR=10.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: iso_BAR_cyto_mem + G_protein_cyto => iso_BAR_G_cyto_mem, Rate Law: (Kf_G_binds_iso_BAR*iso_BAR_cyto_mem*0.00166112956810631*G_protein_cyto+(-Kr_G_binds_iso_BAR*iso_BAR_G_cyto_mem))*cyto_mem
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Km=0.46 0.001*dimensionless*m^(-3)*mol; Vmax_PTP = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: MAPK_active_cyto => MAPK_cyto; PTP_cyto, Rate Law: Vmax_PTP*0.00166112956810631*MAPK_active_cyto*1/(Km+0.00166112956810631*MAPK_active_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Km_GRK_bg=4.0 item*μm^(-2); Vmax_GRK_bg_GRK_bg = NaN item*μm^(-2)*s^(-1)Reaction: iso_BAR_cyto_mem => iso_BAR_p_cyto_mem; GRK_bg_cyto, Rate Law: Vmax_GRK_bg_GRK_bg*iso_BAR_cyto_mem*1/(Km_GRK_bg+iso_BAR_cyto_mem)*cyto_mem
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Kr=2.8E-4 s^(-1); Kf=0.0059 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: R2C2_cyto + cAMP_cyto => c_R2C2_cyto, Rate Law: (Kf*0.00166112956810631*R2C2_cyto*0.00166112956810631*cAMP_cyto+(-Kr*0.00166112956810631*c_R2C2_cyto))*cyto*1*1/KMOLE
Km_pp2a_4=8.0 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_pp2a_4_pp2a_4 = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: PDE4_P_cyto => PDE4_cyto; PP_PDE_cyto, Rate Law: Vmax_pp2a_4_pp2a_4*0.00166112956810631*PDE4_P_cyto*1/(Km_pp2a_4+0.00166112956810631*PDE4_P_cyto)*cyto*1*1/KMOLE
I=0.0 dimensionless*A*m^(-2); Kf=1.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); Kr=0.062 s^(-1)Reaction: iso_extra + BAR_G_cyto_mem => iso_BAR_G_cyto_mem, Rate Law: (Kf*0.00166112956810631*iso_extra*BAR_G_cyto_mem+(-Kr*iso_BAR_G_cyto_mem))*cyto_mem
Vmax_PTP_PKA = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1); KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Km=9.0 0.001*dimensionless*m^(-3)*molReaction: MAPK_active_cyto => MAPK_cyto; PTP_PKA_cyto, Rate Law: Vmax_PTP_PKA*0.00166112956810631*MAPK_active_cyto*1/(Km+0.00166112956810631*MAPK_active_cyto)*cyto*1*1/KMOLE
Vmax_AC_basal_AC_basal = NaN item*μm^(-2)*s^(-1); I=0.0 dimensionless*A*m^(-2); Km_AC_basal=1030.0 0.001*dimensionless*m^(-3)*molReaction: ATP_cyto => cAMP_cyto; AC_cyto_mem, Rate Law: Vmax_AC_basal_AC_basal*0.00166112956810631*ATP_cyto*1/(Km_AC_basal+0.00166112956810631*ATP_cyto)*cyto_mem
Kf=8.35 1000*dimensionless*m^3*mol^(-1)*s^(-1); KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Kr=0.0167 s^(-1)Reaction: c3_R2C2_cyto + cAMP_cyto => PKA_cyto, Rate Law: (Kf*0.00166112956810631*c3_R2C2_cyto*0.00166112956810631*cAMP_cyto+(-Kr*0.00166112956810631*PKA_cyto))*cyto*1*1/KMOLE
KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Kr_bg_binds_GRK=0.25 s^(-1); Kf_bg_binds_GRK=1.0 1000*dimensionless*m^3*mol^(-1)*s^(-1)Reaction: GRK_cyto + bg_cyto => GRK_bg_cyto, Rate Law: (Kf_bg_binds_GRK*0.00166112956810631*GRK_cyto*0.00166112956810631*bg_cyto+(-Kr_bg_binds_GRK*0.00166112956810631*GRK_bg_cyto))*cyto*1*1/KMOLE
Kf_trimer=6.0 1000*dimensionless*m^3*mol^(-1)*s^(-1); KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Kr_trimer=0.0 s^(-1)Reaction: bg_cyto + G_GDP_cyto => G_protein_cyto, Rate Law: (Kf_trimer*0.00166112956810631*bg_cyto*0.00166112956810631*G_GDP_cyto+(-Kr_trimer*0.00166112956810631*G_protein_cyto))*cyto*1*1/KMOLE
Km=0.1 0.001*dimensionless*m^(-3)*mol; KMOLE = 0.00166112956810631 item^(-1)*μmol*l^(-1)*μm^(-3); Vmax_PKA_P_PTP = NaN 0.001*dimensionless*m^(-3)*mol*s^(-1)Reaction: PTP_cyto => PTP_PKA_cyto; PKA_cyto, Rate Law: Vmax_PKA_P_PTP*0.00166112956810631*PTP_cyto*1/(Km+0.00166112956810631*PTP_cyto)*cyto*1*1/KMOLE

States:

NameDescription
iso extraiso_extra
PDE4 P cyto[cAMP-specific 3',5'-cyclic phosphodiesterase 4A; IPR003607]
GRK cytoGRK_cyto
bg cytobg_cyto
PKA cyto[Protein kinase, cAMP-dependent, catalytic, alphacAMP-dependent protein kinase catalytic subunit alpha]
BAR cyto mem[Beta-1 adrenergic receptor]
B Raf cyto[V-raf murine sarcoma viral oncogene B1-like protein]
MEK active cyto[Dual specificity mitogen-activated protein kinase kinase 1]
c3 R2C2 cyto[cAMP-dependent protein kinase complex]
R2C2 cyto[cAMP-dependent protein kinase complex]
G a s cyto[Guanine nucleotide-binding protein G(olf) subunit alpha]
AMP cyto[AMP; AMP]
B Raf active cyto[V-raf murine sarcoma viral oncogene B1-like protein]
iso BAR cyto mem[Beta-1 adrenergic receptor]
c R2C2 cyto[cAMP-dependent protein kinase complex]
cAMP cyto[3',5'-cyclic AMP; 3',5'-Cyclic AMP]
GRK bg cytoGRK_bg_cyto
G GDP cyto[GDP; IPR001019; GDP]
PTP PKA cyto[Tyrosine-protein phosphatase non-receptor type 7; Protein kinase, cAMP-dependent, catalytic, alphacAMP-dependent protein kinase catalytic subunit alpha]
MEK cyto[Dual specificity mitogen-activated protein kinase kinase 1]
iso BAR p cyto mem[Beta-1 adrenergic receptor]
G protein cyto[heterotrimeric G-protein complex]
AC cyto mem[Adenylate cyclase type 2; IPR001054]
PDE4 cyto[cAMP-specific 3',5'-cyclic phosphodiesterase 4A; IPR003607]
PTP cyto[Tyrosine-protein phosphatase non-receptor type 7]
AC active cyto mem[Adenylate cyclase type 2; IPR001054]
MAPK cyto[Mitogen-activated protein kinase 1]
BAR G cyto mem[Beta-1 adrenergic receptor]
iso BAR G cyto mem[Beta-1 adrenergic receptor; heterotrimeric G-protein complex]
ATP cyto[ATP; ATP]
c2 R2C2 cyto[cAMP-dependent protein kinase complex]
MAPK active cyto[Mitogen-activated protein kinase 1]

Nguyen2013 - Dynamic model of HIF regulation in hypoxia: MODEL1912100004v0.0.1

Its a mathematcial model explaining regulation of HIF via FIH and oxygen. Model is further validated by Experimental dat…

Details

Activation of the hypoxia-inducible factor (HIF) pathway is a critical step in the transcriptional response to hypoxia. Although many of the key proteins involved have been characterised, the dynamics of their interactions in generating this response remain unclear. In the present study, we have generated a comprehensive mathematical model of the HIF-1α pathway based on core validated components and dynamic experimental data, and confirm the previously described connections within the predicted network topology. Our model confirms previous work demonstrating that the steps leading to optimal HIF-1α transcriptional activity require sequential inhibition of both prolyl- and asparaginyl-hydroxylases. We predict from our model (and confirm experimentally) that there is residual activity of the asparaginyl-hydroxylase FIH (factor inhibiting HIF) at low oxygen tension. Furthermore, silencing FIH under conditions where prolyl-hydroxylases are inhibited results in increased HIF-1α transcriptional activity, but paradoxically decreases HIF-1α stability. Using a core module of the HIF network and mathematical proof supported by experimental data, we propose that asparaginyl hydroxylation confers a degree of resistance upon HIF-1α to proteosomal degradation. Thus, through in vitro experimental data and in silico predictions, we provide a comprehensive model of the dynamic regulation of HIF-1α transcriptional activity by hydroxylases and use its predictive and adaptive properties to explain counter-intuitive biological observations. link: http://identifiers.org/pubmed/23390316

Nguyen2013 - Dynamic model of HIF regulation in hypoxia (reduced model): MODEL1912100005v0.0.1

Its a mathematcial model explaining regulation of HIF via FIH and oxygen. Model is further validated by Experimental dat…

Details

Activation of the hypoxia-inducible factor (HIF) pathway is a critical step in the transcriptional response to hypoxia. Although many of the key proteins involved have been characterised, the dynamics of their interactions in generating this response remain unclear. In the present study, we have generated a comprehensive mathematical model of the HIF-1α pathway based on core validated components and dynamic experimental data, and confirm the previously described connections within the predicted network topology. Our model confirms previous work demonstrating that the steps leading to optimal HIF-1α transcriptional activity require sequential inhibition of both prolyl- and asparaginyl-hydroxylases. We predict from our model (and confirm experimentally) that there is residual activity of the asparaginyl-hydroxylase FIH (factor inhibiting HIF) at low oxygen tension. Furthermore, silencing FIH under conditions where prolyl-hydroxylases are inhibited results in increased HIF-1α transcriptional activity, but paradoxically decreases HIF-1α stability. Using a core module of the HIF network and mathematical proof supported by experimental data, we propose that asparaginyl hydroxylation confers a degree of resistance upon HIF-1α to proteosomal degradation. Thus, through in vitro experimental data and in silico predictions, we provide a comprehensive model of the dynamic regulation of HIF-1α transcriptional activity by hydroxylases and use its predictive and adaptive properties to explain counter-intuitive biological observations. link: http://identifiers.org/pubmed/23390316

Nguyen2016 - Feedback regulation in cell signalling: Lessons for cancer therapeutics: BIOMD0000000651v0.0.1

Feedback regulation in cell signalling: Lessons for cancer therapeuticsThis model is described in the article: [Feedba…

Details

The notion of feedback is fundamental for understanding signal transduction networks. Feedback loops attenuate or amplify signals, change the network dynamics and modify the input-output relationships between the signal and the target. Negative feedback provides robustness to noise and adaptation to perturbations, but as a double-edged sword can prevent effective pathway inhibition by a drug. Positive feedback brings about switch-like network responses and can convert analog input signals into digital outputs, triggering cell fate decisions and phenotypic changes. We show how a multitude of protein-protein interactions creates hidden feedback loops in signal transduction cascades. Drug treatments that interfere with feedback regulation can cause unexpected adverse effects. Combinatorial molecular interactions generated by pathway crosstalk and feedback loops often bypass the block caused by targeted therapies against oncogenic mutated kinases. We discuss mechanisms of drug resistance caused by network adaptations and suggest that development of effective drug combinations requires understanding of how feedback loops modulate drug responses. link: http://identifiers.org/pubmed/26481970

Parameters:

NameDescription
k8r = 0.01; k8f = 0.001Reaction: RasGDP => RasGTP; aRTK, Rate Law: compartment*(k8f*RasGDP*aRTK-k8r*RasGTP)
k4f = 0.001; k4r = 0.01Reaction: mTORC1 => amTORC1; aAkt, Rate Law: compartment*(k4f*mTORC1*aAkt-k4r*amTORC1)
k9f = 0.001; k9r = 0.01Reaction: Raf => aRaf; RasGTP, Rate Law: compartment*(k9f*Raf*RasGTP-k9r*aRaf)
k6f = 0.1; k6r = 0.001Reaction: IRS => iIRS; aS6K, Rate Law: compartment*(k6f*IRS*aS6K-k6r*iIRS)
k13f = 0.1; k13r = 0.001Reaction: RTK => iRTK; aERK, Rate Law: compartment*(k13f*RTK*aERK-k13r*iRTK)
k2fa = 0.001; k2f = 0.001; k2r = 0.01Reaction: PI3K => aPI3K; aIRS, aRTK, Rate Law: compartment*((k2f*aIRS+k2fa*aRTK)*PI3K-k2r*aPI3K)
k10r = 0.01; k10f = 0.001Reaction: MEK => aMEK; aRaf, Rate Law: compartment*(k10f*MEK*aRaf-k10r*aMEK)
k5r = 0.01; k5f = 0.001Reaction: S6K => aS6K; amTORC1, Rate Law: compartment*(k5f*S6K*amTORC1-k5r*aS6K)
k7fa = 0.01; k7r = 0.01; k7f = 0.01Reaction: RTK => aRTK; FOXO, Rate Law: compartment*((k7f+k7fa*FOXO)*RTK-k7r*aRTK)
k3r = 0.01; k3f = 0.001Reaction: Akt => aAkt; aPI3K, Rate Law: compartment*(k3f*Akt*aPI3K-k3r*aAkt)
k16r = 0.001; k16f = 0.01Reaction: MEK + MEKI => iMEK, Rate Law: compartment*(k16f*MEK*MEKI-k16r*iMEK)
k11r = 0.01; k11f = 0.001Reaction: ERK => aERK; aMEK, Rate Law: compartment*(k11f*ERK*aMEK-k11r*aERK)
k14f = 0.1; k14r = 0.001Reaction: FOXO => iFOXO; aAkt, Rate Law: compartment*(k14f*FOXO*aAkt-k14r*iFOXO)
k12r = 0.001; k12f = 0.01Reaction: Raf => iRaf; aERK, Rate Law: compartment*(k12f*Raf*aERK-k12r*iRaf)
k15r = 0.001; k15f = 0.01Reaction: Akt + AktI => iAkt, Rate Law: compartment*(k15f*Akt*AktI-k15r*iAkt)
k1f = 0.01; k1r = 0.01Reaction: IRS => aIRS, Rate Law: compartment*(k1f*IRS-k1r*aIRS)

States:

NameDescription
iAkt[RAC-alpha serine/threonine-protein kinase]
Akt[RAC-alpha serine/threonine-protein kinase]
iIRS[urn:miriam:sbo:SBO%3A0000015]
RasGDP[GTPase HRas]
aERK[Mitogen-activated protein kinase 3]
iRaf[RAF proto-oncogene serine/threonine-protein kinase]
iMEK[Dual specificity mitogen-activated protein kinase kinase 1]
aRaf[RAF proto-oncogene serine/threonine-protein kinase]
amTORC1[Serine/threonine-protein kinase mTOR]
S6K[Ribosomal protein S6 kinase beta-1]
RTK[Epithelial discoidin domain-containing receptor 1]
MEKI[inhibitor]
PI3K[urn:miriam:uniprot:C17270]
MEK[Dual specificity mitogen-activated protein kinase kinase 1]
AktI[inhibitor]
aAkt[RAC-alpha serine/threonine-protein kinase]
IRS[Insulin receptor substrate 1]
iRTK[Epithelial discoidin domain-containing receptor 1]
aS6K[Ribosomal protein S6 kinase beta-1]
aIRS[Insulin receptor substrate 1]
aMEK[Dual specificity mitogen-activated protein kinase kinase 1]
mTORC1[Serine/threonine-protein kinase mTOR]
Raf[RAF proto-oncogene serine/threonine-protein kinase]
FOXO[Forkhead box protein O1]
RasGTP[GTPase HRas]
aRTK[Epithelial discoidin domain-containing receptor 1]
ERK[Mitogen-activated protein kinase 3]
iFOXO[Forkhead box protein O1]
aPI3K[Phosphatidylinositol-4,5-Bisphosphate 3-Kinase]

NguyenLK2011 - Ubiquitination dynamics in Ring1B/Bmi1 system: BIOMD0000000622v0.0.1

NguyenLK2011 - Ubiquitination dynamics in Ring1B-Bmi1 systemThis theoretical model investigates the dynamics of Ring1B/B…

Details

In an active, self-ubiquitinated state, the Ring1B ligase monoubiquitinates histone H2A playing a critical role in Polycomb-mediated gene silencing. Following ubiquitination by external ligases, Ring1B is targeted for proteosomal degradation. Using biochemical data and computational modeling, we show that the Ring1B ligase can exhibit abrupt switches, overshoot transitions and self-perpetuating oscillations between its distinct ubiquitination and activity states. These different Ring1B states display canonical or multiply branched, atypical polyubiquitin chains and involve association with the Polycomb-group protein Bmi1. Bistable switches and oscillations may lead to all-or-none histone H2A monoubiquitination rates and result in discrete periods of gene (in)activity. Switches, overshoots and oscillations in Ring1B catalytic activity and proteosomal degradation are controlled by the abundances of Bmi1 and Ring1B, and the activities and abundances of external ligases and deubiquitinases, such as E6-AP and USP7. link: http://identifiers.org/pubmed/22194680

Parameters:

NameDescription
k1=2.0; k2=0.2Reaction: Bmi1 + R1B => Z, Rate Law: compartment*(k1*Bmi1*R1B-k2*Z)
v=7.5E-6Reaction: => R1B, Rate Law: compartment*v
k1=0.02; k2=0.2Reaction: Z => Zub, Rate Law: compartment*Z*(k1*Z+k2*Zub)
k=0.001Reaction: R1Bubd => R1B; USP7tot, Rate Law: compartment*k*USP7tot*R1Bubd
k1=0.2; k2=0.2Reaction: R1B => R1Bub, Rate Law: compartment*R1B*(k1*R1B+k2*R1Bub)
k1=3.0E-5Reaction: R1Bubd =>, Rate Law: compartment*k1*R1Bubd
k=0.005Reaction: R1Buba => R1B; USP7tot, Rate Law: compartment*k*USP7tot*R1Buba
k1=0.002; k2=2.0; k3=0.2Reaction: H2A => H2Auba; R1Bub, Zub, R1Buba, Rate Law: compartment*H2A*(k1*R1Bub+k2*Zub+k3*R1Buba)
k1=0.01Reaction: R1B => R1Bubd, Rate Law: compartment*k1*R1B
kc=0.005; Km=0.0025Reaction: Zub => Z; USP7tot, Rate Law: compartment*kc*USP7tot*Zub/(Km+Zub)
k1=0.002Reaction: Bmi1 => Bmi1ubd, Rate Law: compartment*k1*Bmi1
k=0.0075Reaction: R1Bub => R1B; USP7tot, Rate Law: compartment*k*USP7tot*R1Bub
k1=0.012; k2=2.0E-5Reaction: Zub => R1Buba + Bmi1, Rate Law: compartment*(k1*Zub-k2*R1Buba*Bmi1)

States:

NameDescription
Bmi1ubd[Polycomb complex protein BMI-1]
R1Bubd[E3 ubiquitin-protein ligase RING1]
Z[Polycomb complex protein BMI-1; E3 ubiquitin-protein ligase RING1]
Bmi1[Polycomb complex protein BMI-1]
R1Buba[E3 ubiquitin-protein ligase RING1]
R1Bub[E3 ubiquitin-protein ligase RING1]
H2A[Histone H2AX]
Zub[E3 ubiquitin-protein ligase RING1; Polycomb complex protein BMI-1]
H2Auba[Histone H2AX]
R1B[E3 ubiquitin-protein ligase RING1]

Niederer2006_CardiacMyocyteRelaxation: MODEL8687196544v0.0.1

This a model from the article: A quantitative analysis of cardiac myocyte relaxation: a simulation study. Niederer S…

Details

The determinants of relaxation in cardiac muscle are poorly understood, yet compromised relaxation accompanies various pathologies and impaired pump function. In this study, we develop a model of active contraction to elucidate the relative importance of the [Ca2+]i transient magnitude, the unbinding of Ca2+ from troponin C (TnC), and the length-dependence of tension and Ca2+ sensitivity on relaxation. Using the framework proposed by one of our researchers, we extensively reviewed experimental literature, to quantitatively characterize the binding of Ca2+ to TnC, the kinetics of tropomyosin, the availability of binding sites, and the kinetics of crossbridge binding after perturbations in sarcomere length. Model parameters were determined from multiple experimental results and modalities (skinned and intact preparations) and model results were validated against data from length step, caged Ca2+, isometric twitches, and the half-time to relaxation with increasing sarcomere length experiments. A factorial analysis found that the [Ca2+]i transient and the unbinding of Ca2+ from TnC were the primary determinants of relaxation, with a fivefold greater effect than that of length-dependent maximum tension and twice the effect of tension-dependent binding of Ca2+ to TnC and length-dependent Ca2+ sensitivity. The affects of the [Ca2+]i transient and the unbinding rate of Ca2+ from TnC were tightly coupled with the effect of increasing either factor, depending on the reference [Ca2+]i transient and unbinding rate. link: http://identifiers.org/pubmed/16339881

Nielsen1998_Glycolysis: BIOMD0000000042v0.0.1

This model was automatically converted from model BIOMD0000000042 by using [libSBML](http://sbml.org/Software/libSBML)…

Details

We report sustained oscillations in glycolysis conducted in an open system (a continuous-flow, stirred tank reactor; CSTR) with inflow of yeast extract as well as glucose. Depending on the operating conditions, we observe simple or complex periodic oscillations or chaos. We report the response of the system to instantaneous additions of small amounts of several substrates as functions of the amount added and the phase of the addition. We simulate oscillations and perturbations by a kinetic model based on the mechanism of glycolysis in a CSTR. We find that the response to particular perturbations forms an efficient tool for elucidating the mechanism of biochemical oscillations. link: http://identifiers.org/pubmed/17029704

Parameters:

NameDescription
V2 = 1.5; K2 = 0.0016; k2 = 0.017; K2ATP = 0.01Reaction: F6P + ATP => FBP + ADP; AMP, Rate Law: compartment*V2*ATP*F6P^2/((K2*(1+k2*(ATP/AMP)^2)+F6P^2)*(K2ATP+ATP))
k9f = 10.0; k9b = 10.0Reaction: AMP + ATP => ADP, Rate Law: compartment*(k9f*AMP*ATP-k9b*ADP^2)
k3b = 50.0; k3f = 1.0Reaction: FBP => GAP, Rate Law: compartment*(k3f*FBP-k3b*GAP^2)
V4 = 10.0; K4GAP = 1.0; K4NAD = 1.0Reaction: GAP + NAD => DPG + NADH, Rate Law: compartment*V4*NAD*GAP/((K4GAP+GAP)*(K4NAD+NAD))
K1GLC = 0.1; V1 = 0.5; K1ATP = 0.063Reaction: GLC + ATP => F6P + ADP, Rate Law: compartment*V1*ATP*GLC/((K1GLC+GLC)*(K1ATP+ATP))
flow = 0.011Reaction: => ATP, Rate Law: compartment*(3.5-ATP)*flow
V6 = 10.0; K6ADP = 0.3; K6PEP = 0.2Reaction: PEP + ADP => PYR + ATP, Rate Law: compartment*V6*ADP*PEP/((K6PEP+PEP)*(K6ADP+ADP))
k5f = 1.0; k5b = 0.5Reaction: DPG + ADP => PEP + ATP, Rate Law: compartment*(k5f*DPG*ADP-k5b*PEP*ATP)
k8b = 1.43E-4; k8f = 1.0Reaction: ACA + NADH => EtOH + NAD, Rate Law: compartment*(k8f*NADH*ACA-k8b*NAD*EtOH)
V7 = 2.0; K7PYR = 0.3Reaction: PYR => ACA, Rate Law: compartment*V7*PYR/(K7PYR+PYR)
k10 = 0.05Reaction: F6P => P, Rate Law: compartment*k10*F6P

States:

NameDescription
ATP[ATP; ATP]
DPG[3-phospho-D-glyceroyl dihydrogen phosphate; 3-Phospho-D-glyceroyl phosphate]
NADH[NADH; NADH]
PP
PYR[pyruvate; Pyruvate]
EtOH[ethanol; Ethanol]
FBP[keto-D-fructose 1,6-bisphosphate; beta-D-Fructose 1,6-bisphosphate]
GLC[glucose; C00293]
F6P[CHEBI_20935; beta-D-Fructose 6-phosphate]
AMP[AMP; AMP]
ACA[acetaldehyde; Acetaldehyde]
GAP[D-glyceraldehyde 3-phosphate; D-Glyceraldehyde 3-phosphate]
PEP[phosphoenolpyruvate; Phosphoenolpyruvate]
ADP[ADP; ADP]
NAD[NAD(+); NAD+]

Nijhout2004_Folate_Cycle: BIOMD0000000213v0.0.1

This is an SBML version of the folate cycle model model from: **A mathematical model of the folate cycle: new insights…

Details

A mathematical model is developed for the folate cycle based on standard biochemical kinetics. We use the model to provide new insights into several different mechanisms of folate homeostasis. The model reproduces the known pool sizes of folate substrates and the fluxes through each of the loops of the folate cycle and has the qualitative behavior observed in a variety of experimental studies. Vitamin B(12) deficiency, modeled as a reduction in the V(max) of the methionine synthase reaction, results in a secondary folate deficiency via the accumulation of folate as 5-methyltetrahydrofolate (the "methyl trap"). One form of homeostasis is revealed by the fact that a 100-fold up-regulation of thymidylate synthase and dihydrofolate reductase (known to occur at the G(1)/S transition) dramatically increases pyrimidine production without affecting the other reactions of the folate cycle. The model also predicts that an almost total inhibition of dihydrofolate reductase is required to significantly inhibit the thymidylate synthase reaction, consistent with experimental and clinical studies on the effects of methotrexate. Sensitivity to variation in enzymatic parameters tends to be local in the cycle and inversely proportional to the number of reactions that interconvert two folate substrates. Another form of homeostasis is a consequence of the nonenzymatic binding of folate substrates to folate enzymes. Without folate binding, the velocities of the reactions decrease approximately linearly as total folate is decreased. In the presence of folate binding and allosteric inhibition, the velocities show a remarkable constancy as total folate is decreased. link: http://identifiers.org/pubmed/15496403

Parameters:

NameDescription
MTCH_VmaxF = 800000.0; MTCH_VmaxR = 20000.0; MTCH_Km_5_10_CHTHF = 250.0; MTCH_Km_10fTHF = 100.0Reaction: _5_10_CHTHF => _10fTHF, Rate Law: MTCH_VmaxF*_5_10_CHTHF/(MTCH_Km_5_10_CHTHF+_5_10_CHTHF)-MTCH_VmaxR*_10fTHF/(MTCH_Km_10fTHF+_10fTHF)
FTD_Vmax = 14000.0; FTD_Km_10fTHF = 20.0Reaction: _10fTHF => THF, Rate Law: FTD_Vmax*_10fTHF/(FTD_Km_10fTHF+_10fTHF)
DHFR_Km_NADPH = 4.0; DHFR_Vmax = 50.0; DHFR_Km_DHF = 0.5Reaction: DHF => THF; NADPH, Rate Law: DHFR_Vmax*NADPH/(DHFR_Km_NADPH+NADPH)*DHF/(DHFR_Km_DHF+DHF)
AICART_Vmax = 45000.0; AICART_Km_10fTHF = 5.9; AICART_Km_AICAR = 100.0Reaction: _10fTHF => THF; AICAR, Rate Law: AICART_Vmax*AICAR/(AICART_Km_AICAR+AICAR)*_10fTHF/(AICART_Km_10fTHF+_10fTHF)
MTD_VmaxR = 594000.0; MTD_Km_5_10_CHTHF = 10.0; MTD_VmaxF = 200000.0; MTD_Km_5_10_CH2THF = 2.0Reaction: _5_10_CH2THF => _5_10_CHTHF, Rate Law: MTD_VmaxF*_5_10_CH2THF/(MTD_Km_5_10_CH2THF+_5_10_CH2THF)-MTD_VmaxR*_5_10_CHTHF/(MTD_Km_5_10_CHTHF+_5_10_CHTHF)
MS_Vmax = 500.0; MS_Km_Hcy = 0.1; MS_Km_5mTHF = 25.0; MS_Kd = 1.0Reaction: _5mTHF => THF; Hcy, Rate Law: MS_Vmax*_5mTHF/MS_Km_5mTHF*Hcy/MS_Km_Hcy/(MS_Kd/MS_Km_5mTHF+_5mTHF/MS_Km_5mTHF+Hcy/MS_Km_Hcy+_5mTHF*Hcy/(MS_Km_5mTHF*MS_Km_Hcy))
PGT_Km_10fTHF = 4.9; PGT_Km_GAR = 520.0; PGT_Vmax = 16200.0Reaction: _10fTHF => THF; GAR, Rate Law: PGT_Vmax*GAR/(PGT_Km_GAR+GAR)*_10fTHF/(PGT_Km_10fTHF+_10fTHF)
TS_Km_dUMP = 6.3; TS_Vmax = 50.0; TS_Km_5_10_CH2THF = 14.0Reaction: _5_10_CH2THF => DHF; dUMP, Rate Law: TS_Vmax*dUMP/(TS_Km_dUMP+dUMP)*_5_10_CH2THF/(TS_Km_5_10_CH2THF+_5_10_CH2THF)
NE_k2 = 12.0; NE_k1 = 0.15Reaction: THF => _5_10_CH2THF; HCOOH, Rate Law: HCOOH*NE_k1*THF-NE_k2*_5_10_CH2THF
FTS_Km_HCOOH = 43.0; FTS_Km_THF = 3.0; FTS_Vmax = 2000.0Reaction: THF => _10fTHF; HCOOH, Rate Law: FTS_Vmax*HCOOH/(FTS_Km_HCOOH+HCOOH)*THF/(FTS_Km_THF+THF)
SHMT_Km_Ser = 600.0; SHMT_Km_THF = 50.0; SHMT_VmaxR = 25000.0; SHMT_VmaxF = 40000.0Reaction: THF => _5_10_CH2THF; Ser, Gly, Rate Law: SHMT_VmaxF*Ser/(SHMT_Km_Ser+Ser)*THF/(SHMT_Km_THF+THF)-SHMT_VmaxR*Gly/(SHMT_Km_Ser+Gly)*_5_10_CH2THF/(SHMT_Km_THF+_5_10_CH2THF)
MTHFR_Km_5_10_CH2THF = 50.0; MTHFR_Vmax = 6000.0; MTHFR_Km_NADPH = 16.0Reaction: _5_10_CH2THF => _5mTHF; NADPH, Rate Law: MTHFR_Vmax*NADPH/(MTHFR_Km_NADPH+NADPH)*_5_10_CH2THF/(MTHFR_Km_5_10_CH2THF+_5_10_CH2THF)

States:

NameDescription
10fTHF[10-formyltetrahydrofolic acid; 10-Formyltetrahydrofolate]
5mTHF[5-methyltetrahydrofolic acid; 5-Methyltetrahydrofolate]
5 10 CHTHF[(6R)-5,10-methenyltetrahydrofolic acid; 5,10-Methenyltetrahydrofolate]
5 10 CH2THF[(6R)-5,10-methylenetetrahydrofolic acid; 5,10-Methylenetetrahydrofolate]
THF[(6S)-5,6,7,8-tetrahydrofolic acid; Tetrahydrofolate]
DHF[dihydrofolic acid; Dihydrofolate]

Nijhout2004_FolateCycle: MODEL6655501972v0.0.1

This is an SBML version of the folate cycle model model from: **A mathematical model of the folate cycle: new insights…

Details

A mathematical model is developed for the folate cycle based on standard biochemical kinetics. We use the model to provide new insights into several different mechanisms of folate homeostasis. The model reproduces the known pool sizes of folate substrates and the fluxes through each of the loops of the folate cycle and has the qualitative behavior observed in a variety of experimental studies. Vitamin B(12) deficiency, modeled as a reduction in the V(max) of the methionine synthase reaction, results in a secondary folate deficiency via the accumulation of folate as 5-methyltetrahydrofolate (the "methyl trap"). One form of homeostasis is revealed by the fact that a 100-fold up-regulation of thymidylate synthase and dihydrofolate reductase (known to occur at the G(1)/S transition) dramatically increases pyrimidine production without affecting the other reactions of the folate cycle. The model also predicts that an almost total inhibition of dihydrofolate reductase is required to significantly inhibit the thymidylate synthase reaction, consistent with experimental and clinical studies on the effects of methotrexate. Sensitivity to variation in enzymatic parameters tends to be local in the cycle and inversely proportional to the number of reactions that interconvert two folate substrates. Another form of homeostasis is a consequence of the nonenzymatic binding of folate substrates to folate enzymes. Without folate binding, the velocities of the reactions decrease approximately linearly as total folate is decreased. In the presence of folate binding and allosteric inhibition, the velocities show a remarkable constancy as total folate is decreased. link: http://identifiers.org/pubmed/15496403

Nijhout2006_Hepatic_Folate_Metab: MODEL1007200000v0.0.1

This is the model described in the article: In silico experimentation with a model of hepatic mitochondrial folate met…

Details

In eukaryotes, folate metabolism is compartmentalized and occurs in both the cytosol and the mitochondria. The function of this compartmentalization and the great changes that occur in the mitochondrial compartment during embryonic development and in rapidly growing cancer cells are gradually becoming understood, though many aspects remain puzzling and controversial.We explore the properties of cytosolic and mitochondrial folate metabolism by experimenting with a mathematical model of hepatic one-carbon metabolism. The model is based on known biochemical properties of mitochondrial and cytosolic enzymes. We use the model to study questions about the relative roles of the cytosolic and mitochondrial folate cycles posed in the experimental literature. We investigate: the control of the direction of the mitochondrial and cytosolic serine hydroxymethyltransferase (SHMT) reactions, the role of the mitochondrial bifunctional enzyme, the role of the glycine cleavage system, the effects of variations in serine and glycine inputs, and the effects of methionine and protein loading.The model reproduces many experimental findings and gives new insights into the underlying properties of mitochondrial folate metabolism. Particularly interesting is the remarkable stability of formate production in the mitochondria in the face of large changes in serine and glycine input. The model shows that in the presence of the bifunctional enzyme (as in embryonic tissues and cancer cells), the mitochondria primarily support cytosolic purine and pyrimidine synthesis via the export of formate, while in adult tissues the mitochondria produce serine for gluconeogenesis. link: http://identifiers.org/pubmed/17150100

NIK-dependent p100 processing into p52 with RelB binding and IkBd degradation, mass action, SBML 2v4: BIOMD0000000871v0.0.1

This model represents NIK-dependent p100 processing into p52 followed by binding to RelB and NIK-dependent IkBd degradat…

Details

Signaling pathways often share molecular components, tying the activity of one pathway to the functioning of another. In the NFκB signaling system, distinct kinases mediate inflammatory and developmental signaling via RelA and RelB, respectively. Although the substrates of the developmental, so-called noncanonical, pathway are induced by inflammatory/canonical signaling, crosstalk is limited. Through dynamical systems modeling, we identified the underlying regulatory mechanism. We found that as the substrate of the noncanonical kinase NIK, the nfkb2 gene product p100, transitions from a monomer to a multimeric complex, it may compete with and inhibit p100 processing to the active p52. Although multimeric complexes of p100 (IκBδ) are known to inhibit preexisting RelA:p50 through sequestration, here we report that p100 complexes can inhibit the enzymatic formation of RelB:p52. We show that the dose–response systems properties of this complex substrate competition motif are poorly accounted for by standard Michaelis–Menten kinetics, but require more detailed mass action formulations. In sum, although tonic inflammatory signaling is required for adequate expression of the noncanonical pathway precursors, the substrate complex competition motif identified here can prevent amplification of the active RelB:p52 dimer in elevated inflammatory conditions to ensure reliable RelB-dependent developmental signaling independent of inflammatory context. link: http://identifiers.org/doi/10.1073/pnas.1816000116

Parameters:

NameDescription
k1=0.0228Reaction: RelB =>, Rate Law: compartment*k1*RelB
k=1000.0; Kd=50.0; canon = 1.0Reaction: => p100, Rate Law: compartment*k*canon/(Kd+canon)
k1=0.05Reaction: p100_NIK => p52 + NIK, Rate Law: compartment*k1*p100_NIK
k2=0.00144; k1=9.6E-4Reaction: RelB + p52 => RelB_p52, Rate Law: compartment*(k1*RelB*p52-k2*RelB_p52)
k1=1.6E-5; k2=2.4E-4Reaction: p100 => IkBd, Rate Law: compartment*(k1*p100^2-k2*IkBd)
k1=3.8E-4Reaction: IkBd =>, Rate Law: compartment*k1*IkBd
k1=0.005; k2=2.4E-4Reaction: IkBd + NIK => IkBd_NIK, Rate Law: compartment*(k1*IkBd*NIK-k2*IkBd_NIK)
k=42.0; Kd=50.0; canon = 1.0Reaction: => RelB, Rate Law: compartment*k*canon/(Kd+canon)

States:

NameDescription
IkBd[Nuclear factor NF-kappa-B p100 subunit]
NIK[Mitogen-activated protein kinase kinase kinase 14]
p100 NIK[Nuclear factor NF-kappa-B p100 subunit; Mitogen-activated protein kinase kinase kinase 14]
p52[Nuclear factor NF-kappa-B p100 subunit]
IkBd NIK[Nuclear factor NF-kappa-B p100 subunit; Mitogen-activated protein kinase kinase kinase 14]
RelB[Transcription factor RelB]
p100[Nuclear factor NF-kappa-B p100 subunit]
RelB p52[Nuclear factor NF-kappa-B p100 subunit; Transcription factor RelB]

Nikolaev2005_AlbuminBilirubinAdsorption: BIOMD0000000291v0.0.1

This a model from the article: Mathematical model of binding of albumin-bilirubin complex to the surface of carbon p…

Details

We proposed a mathematical model and estimated the parameters of adsorption of albumin-bilirubin complex to the surface of carbon pyropolymer. Design data corresponded to the results of experimental studies. Our findings indicate that modeling of this process should take into account fractal properties of the surface of carbon pyropolymer. link: http://identifiers.org/pubmed/16307060

Parameters:

NameDescription
k6 = 3.226E-7; k8 = 1.011E-7; k9 = 0.01685; k10 = 0.1325; n = 1.0Reaction: x2 = (k6*x7*x6-k8*x2)+k9*x1*x7^(n+1)+k10*x4*x7, Rate Law: (k6*x7*x6-k8*x2)+k9*x1*x7^(n+1)+k10*x4*x7
k7 = 0.00301; k5 = 0.005489; k9 = 0.01685; n = 1.0Reaction: x3 = (k5*x7^n*x5-k7*x3)+k9*x1*x7^(n+1), Rate Law: (k5*x7^n*x5-k7*x3)+k9*x1*x7^(n+1)
K_AlB = 95000.0; K_AlB2 = 3000.0; k3 = 5.095E-6; k9 = 0.01685; k10 = 0.1325; k4 = 2.656E-5; n = 1.0Reaction: x1 = (((K_AlB*k3*x5*x6-K_AlB2*k4*x1*x6)-k3*x1)-k9*x1*x7^(n+1))+k4*x4+k10*x4*x7, Rate Law: (((K_AlB*k3*x5*x6-K_AlB2*k4*x1*x6)-k3*x1)-k9*x1*x7^(n+1))+k4*x4+k10*x4*x7
K_AlB2 = 3000.0; k10 = 0.1325; k4 = 2.656E-5Reaction: x4 = (K_AlB2*k4*x1*x6-k4*x4)-k10*x4*x7, Rate Law: (K_AlB2*k4*x1*x6-k4*x4)-k10*x4*x7
n = 1.0Reaction: x7 = (C0-x2)-n*x3, Rate Law: missing

States:

NameDescription
x5[Serum albumin]
x1[Serum albumin; Bilirubin]
x7[macromolecule; carbon atom]
x4[Serum albumin; Bilirubin]
x2[macromolecule; carbon atom; Bilirubin]
x6[bilirubin; Bilirubin]
x3[macromolecule; carbon atom; Serum albumin]

Nikolaev2019 - Immunobiochemical reconstruction of influenza lung infection-melanoma skin cancer interactions: BIOMD0000000865v0.0.1

This is a mathematical mechanistic immunobiochemical model that incorporates T cell pathways that control programmed cel…

Details

It was recently reported that acute influenza infection of the lung promoted distal melanoma growth in the dermis of mice. Melanoma-specific CD8+ T cells were shunted to the lung in the presence of the infection, where they expressed high levels of inflammation-induced cell-activation blocker PD-1, and became incapable of migrating back to the tumor site. At the same time, co-infection virus-specific CD8+ T cells remained functional while the infection was cleared. It was also unexpectedly found that PD-1 blockade immunotherapy reversed this effect. Here, we proceed to ground the experimental observations in a mechanistic immunobiochemical model that incorporates T cell pathways that control PD-1 expression. A core component of our model is a kinetic motif, which we call a PD-1 Double Incoherent Feed-Forward Loop (DIFFL), and which reflects known interactions between IRF4, Blimp-1, and Bcl-6. The different activity levels of the PD-1 DIFFL components, as a function of the cognate antigen levels and the given inflammation context, manifest themselves in phenotypically distinct outcomes. Collectively, the model allowed us to put forward a few working hypotheses as follows: (i) the melanoma-specific CD8+ T cells re-circulating with the blood flow enter the lung where they express high levels of inflammation-induced cell-activation blocker PD-1 in the presence of infection; (ii) when PD-1 receptors interact with abundant PD-L1, constitutively expressed in the lung, T cells loose motility; (iii) at the same time, virus-specific cells adapt to strong stimulation by their cognate antigen by lowering the transiently-elevated expression of PD-1, remaining functional and mobile in the inflamed lung, while the infection is cleared. The role that T cell receptor (TCR) activation and feedback loops play in the underlying processes are also highlighted and discussed. We hope that the results reported in our study could potentially contribute to the advancement of immunological approaches to cancer treatment and, as well, to a better understanding of a broader complexity of fundamental interactions between pathogens and tumors. link: http://identifiers.org/pubmed/30745900

Parameters:

NameDescription
U_a_k_P = 0.0215814688039663; n_b = 2.0; a_b = 100.0; A_b = 10.0; r_b = 2.0; m_b = 2.0; K_b = 1.0; M_b = 10.0; k_b = 25.0Reaction: => B; I, C, Rate Law: compartment*(a_b*U_a_k_P^n_b/(A_b^n_b+U_a_k_P^n_b)+k_b*I^m_b/(K_b^m_b+I^m_b))*M_b^r_b/(M_b^r_b+C^r_b)
mu_b = 1.0Reaction: B =>, Rate Law: compartment*mu_b*B
U_a_k_P = 0.0215814688039663; n_c = 3.0; A_c = 0.01; r_c = 2.0; M_c = 10.0; a_c = 0.75Reaction: => C; B, I, Rate Law: compartment*a_c*U_a_k_P^n_c/(A_c^n_c+U_a_k_P^n_c)*M_c^r_c/(M_c^r_c+B^r_c+I^r_c+C^r_c)
mu_c = 0.1Reaction: C =>, Rate Law: compartment*mu_c*C
mu_p = 0.1Reaction: P =>, Rate Law: compartment*mu_p*P
mu_i = 1.0Reaction: I =>, Rate Law: compartment*mu_i*I
U_a_k_P = 0.0215814688039663; n_p = 3.0; sigma_p_tilde = 0.1; A_p = 0.1; a_p = 0.75; M_p = 10.0; r_p = 4.0Reaction: => P; B, Rate Law: compartment*(sigma_p_tilde+a_p*U_a_k_P^n_p/(A_p^n_p+U_a_k_P^n_p))*M_p^r_p/(M_p^r_p+B^r_p)
U_a_k_P = 0.0215814688039663; Q_i = 1.0; n_i = 2.0; a_i = 75.0; k_i = 7.5; m_i = 2.0; q_i = 7.5; s_i = 2.0; sigma_i = 0.3; K_i = 1.0; Phi_L_P = 1.0; A_i = 1.0Reaction: => I; B, I, Rate Law: compartment*Phi_L_P*(sigma_i+a_i*U_a_k_P^n_i/(A_i^n_i+U_a_k_P^n_i)+k_i*B^m_i/(K_i^m_i+B^m_i)+q_i*I^s_i/(Q_i^s_i+I^s_i))

States:

NameDescription
B[PR:000001831]
I[C17926]
C[C26149]
P[PR:000001919]

Nikolopoulou2018 - Tumour-immune dynamics with an immune checkpoint inhibitor: MODEL1908270001v0.0.1

This is a mathematical model investigating the effects of continuous and intermittent PD-L1 and anti-PD-L1 therapy upon…

Details

The use of immune checkpoint inhibitors is becoming more commonplace in clinical trials across the nation. Two important factors in the tumour-immune response are the checkpoint protein programmed death-1 (PD-1) and its ligand PD-L1. We propose a mathematical tumour-immune model using a system of ordinary differential equations to study dynamics with and without the use of anti-PD-1. A sensitivity analysis is conducted, and series of simulations are performed to investigate the effects of intermittent and continuous treatments on the tumour-immune dynamics. We consider the system without the anti-PD-1 drug to conduct a mathematical analysis to determine the stability of the tumour-free and tumorous equilibria. Through simulations, we found that a normally functioning immune system may control tumour. We observe treatment with anti-PD-1 alone may not be sufficient to eradicate tumour cells. Therefore, it may be beneficial to combine single agent treatments with additional therapies to obtain a better antitumour response. link: http://identifiers.org/doi/10.1080/23737867.2018.1440978

Nishio2008 - Design of the phosphotransferase system for enhanced glucose uptake in E. coli.: BIOMD0000000571v0.0.1

Nishio2008 - Design of the phosphotransferase system for enhanced glucose uptake in E. coli.This model is described in t…

Details

The phosphotransferase system (PTS) is the sugar transportation machinery that is widely distributed in prokaryotes and is critical for enhanced production of useful metabolites. To increase the glucose uptake rate, we propose a rational strategy for designing the molecular architecture of the Escherichia coli glucose PTS by using a computer-aided design (CAD) system and verified the simulated results with biological experiments. CAD supports construction of a biochemical map, mathematical modeling, simulation, and system analysis. Assuming that the PTS aims at controlling the glucose uptake rate, the PTS was decomposed into hierarchical modules, functional and flux modules, and the effect of changes in gene expression on the glucose uptake rate was simulated to make a rational strategy of how the gene regulatory network is engineered. Such design and analysis predicted that the mlc knockout mutant with ptsI gene overexpression would greatly increase the specific glucose uptake rate. By using biological experiments, we validated the prediction and the presented strategy, thereby enhancing the specific glucose uptake rate. link: http://identifiers.org/pubmed/18197177

Parameters:

NameDescription
fast = 1.0E9 (60*s)^(-1); Kb=40000.0 mol^(-1)*l; one_per_M=1.0 mol^(-1)*lReaction: CRP + cAMP => CRP_cAMP; CRP, cAMP, CRP_cAMP, Rate Law: cyt*fast*one_per_M*(Kb^2*(CRP*cAMP)^2-CRP_cAMP^2)
kmd=0.0866 (60*s)^(-1)Reaction: mRNA_crr => ; mRNA_crr, Rate Law: cyt*kmd*mRNA_crr
fast = 1.0E9 (60*s)^(-1); Kb=2.0E8 mol^(-1)*lReaction: Mlc + Mlcsite_ptsGp1 => Mlc_Mlcsite_ptsGp1; Mlc, Mlcsite_ptsGp1, Mlc_Mlcsite_ptsGp1, Rate Law: cyt*fast*(Kb*Mlc*Mlcsite_ptsGp1-Mlc_Mlcsite_ptsGp1)
fast = 1.0E9 (60*s)^(-1); Kb=2.22E7 mol^(-1)*lReaction: CRP_cAMP + CRPsiteI_crp => CRP_cAMP_CRPsiteI_crp; CRP_cAMP, CRPsiteI_crp, CRP_cAMP_CRPsiteI_crp, Rate Law: cyt*fast*(Kb*CRP_cAMP*CRPsiteI_crp-CRP_cAMP_CRPsiteI_crp)
Kmich=9.61 mol*l^(-1); Q=389.0 (60*s)^(-1)Reaction: IICB + Glc6P => IICB_P + Glucose; IICB, Glc6P, Rate Law: cyt*Q*IICB*Glc6P/(Kmich+Glc6P)
kmd=0.0889 (60*s)^(-1)Reaction: mRNA_ptsH => ; mRNA_ptsH, Rate Law: cyt*kmd*mRNA_ptsH
Q=4800.0 (60*s)^(-1); Kmich=2.0E-5 mol*l^(-1)Reaction: IICB_P + Glucose => IICB + Glc6P; IICB_P, Glucose, Rate Law: cyt*Q*IICB_P*Glucose/(Kmich+Glucose)
km=0.892 (60*s)^(-1); TCRPsite_ptsIp1 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_ptsI; CRP_cAMP_CRPsite_ptsIp1, ptsIp1, CRP_cAMP_CRPsite_ptsIp1, ptsIp1, Rate Law: cyt*km*CRP_cAMP_CRPsite_ptsIp1/TCRPsite_ptsIp1*ptsIp1
Kmich=0.001 mol*l^(-1); Q=100.0 (60*s)^(-1)Reaction: ATP => cAMP; CYA, CYA, ATP, Rate Law: cyt*Q*CYA*ATP/(Kmich+ATP)
TCRPsite_ptsGp2 = 2.43E-10 mol*l^(-1); km=2.0 (60*s)^(-1); TMlcsite_ptsGp2 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_ptsG; CRP_cAMP_CRPsite_ptsGp2, Mlc_Mlcsite_ptsGp2, ptsGp2, CRP_cAMP_CRPsite_ptsGp2, Mlc_Mlcsite_ptsGp2, ptsGp2, Rate Law: cyt*km*CRP_cAMP_CRPsite_ptsGp2/TCRPsite_ptsGp2*(1-Mlc_Mlcsite_ptsGp2/TMlcsite_ptsGp2)*ptsGp2
fast = 1.0E9 (60*s)^(-1); Kb=2430000.0 mol^(-1)*lReaction: Mlc + Mlcsite_mlcp1 => Mlc_Mlcsite_mlcp1; Mlc, Mlcsite_mlcp1, Mlc_Mlcsite_mlcp1, Rate Law: cyt*fast*(Kb*Mlc*Mlcsite_mlcp1-Mlc_Mlcsite_mlcp1)
Kb=7000000.0 mol^(-1)*l; fast = 1.0E9 (60*s)^(-1)Reaction: IICB + Mlc => IICB_Mlc; IICB, Mlc, IICB_Mlc, Rate Law: cyt*fast*(Kb*IICB*Mlc-IICB_Mlc)
Kb=1350000.0 mol^(-1)*l; fast = 1.0E9 (60*s)^(-1)Reaction: Mlc + Mlcsite_mlcp2 => Mlc_Mlcsite_mlcp2; Mlc, Mlcsite_mlcp2, Mlc_Mlcsite_mlcp2, Rate Law: cyt*fast*(Kb*Mlc*Mlcsite_mlcp2-Mlc_Mlcsite_mlcp2)
fast = 1.0E9 (60*s)^(-1); Kb=1.0E8 mol^(-2)*l^2Reaction: CYA + IIA_P => IIA_P_CYA; CYA, IIA_P, IIA_P_CYA, Rate Law: cyt*fast*(Kb*CYA*IIA_P^2-IIA_P_CYA)
kx=2.4E8 mol^(-1)*l*(60*s)^(-1)Reaction: IICB_P + IIA => IICB + IIA_P; IIA, IICB_P, Rate Law: cyt*kx*IIA*IICB_P
fast = 1.0E9 (60*s)^(-1); Kb=2700000.0 mol^(-1)*lReaction: CRP_cAMP + CRPsiteII_crp => CRP_cAMP_CRPsiteII_crp; CRP_cAMP, CRPsiteII_crp, CRP_cAMP_CRPsiteII_crp, Rate Law: cyt*fast*(Kb*CRP_cAMP*CRPsiteII_crp-CRP_cAMP_CRPsiteII_crp)
kx=6.6E8 mol^(-1)*l*(60*s)^(-1)Reaction: IICB + IIA_P => IICB_P + IIA; IICB, IIA_P, Rate Law: cyt*kx*IICB*IIA_P
kp=11.0 (60*s)^(-1)Reaction: => CRP; mRNA_crp, mRNA_crp, Rate Law: cyt*kp*mRNA_crp
kpd=0.1 (60*s)^(-1)Reaction: CRP_cAMP_CRPsite_ptsIp1 => CRPsite_ptsIp1; CRP_cAMP_CRPsite_ptsIp1, Rate Law: cyt*kpd*CRP_cAMP_CRPsite_ptsIp1
kmd=0.0797 (60*s)^(-1)Reaction: mRNA_ptsI => ; mRNA_ptsI, Rate Law: cyt*kmd*mRNA_ptsI
Q=480000.0 (60*s)^(-1); Kmich=0.002 mol*l^(-1)Reaction: EI_P + Pyr => EI + PEP; EI_P, Pyr, Rate Law: cyt*2*Q*EI_P*Pyr^2/(Kmich^2+Pyr^2)
TCRPsite_ptsGp1 = 2.43E-10 mol*l^(-1); km=892.0 (60*s)^(-1); TMlcsite_ptsGp1 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_ptsG; CRP_cAMP_CRPsite_ptsGp1, Mlc_Mlcsite_ptsGp1, ptsGp1, CRP_cAMP_CRPsite_ptsGp1, Mlc_Mlcsite_ptsGp1, ptsGp1, Rate Law: cyt*km*CRP_cAMP_CRPsite_ptsGp1/TCRPsite_ptsGp1*(1-Mlc_Mlcsite_ptsGp1/TMlcsite_ptsGp1)*ptsGp1
km=334.5 (60*s)^(-1)Reaction: => mRNA_crr; crr, crr, Rate Law: cyt*km*crr
kpd=400.0 (60*s)^(-1)Reaction: cAMP => ; cAMP, Rate Law: cyt*kpd*cAMP
kx=3.66E9 mol^(-1)*l*(60*s)^(-1)Reaction: IIA + HPr_P => IIA_P + HPr; IIA, HPr_P, Rate Law: cyt*kx*IIA*HPr_P
kx=4.8E8 mol^(-1)*l*(60*s)^(-1)Reaction: HPr_P + EI => HPr + EI_P; EI, HPr_P, Rate Law: cyt*kx*EI*HPr_P
Q=9000.0 (60*s)^(-1); Kmich=0.001 mol*l^(-1)Reaction: ATP => cAMP; IIA_P_CYA, IIA_P_CYA, ATP, Rate Law: cyt*Q*IIA_P_CYA*ATP/(Kmich+ATP)
kmd=0.3014 (60*s)^(-1)Reaction: mRNA_mlc => ; mRNA_mlc, Rate Law: cyt*kmd*mRNA_mlc
kx=1.2E10 mol^(-1)*l*(60*s)^(-1)Reaction: HPr + EI_P => HPr_P + EI; HPr, EI_P, Rate Law: cyt*kx*HPr*EI_P
fast = 1.0E9 (60*s)^(-1); Kb=6.67E7 mol^(-1)*lReaction: CRP_cAMP + CRPsite_cyaA => CRP_cAMP_CRPsite_cyaA; CRP_cAMP, CRPsite_cyaA, CRP_cAMP_CRPsite_cyaA, Rate Law: cyt*fast*(Kb*CRP_cAMP*CRPsite_cyaA-CRP_cAMP_CRPsite_cyaA)
km=1.875 (60*s)^(-1); TMlcsite_mlcp2 = 2.43E-10 mol*l^(-1); TCRPsite_mlcp2 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_mlc; CRP_cAMP_CRPsite_mlcp2, Mlc_Mlcsite_mlcp2, mlcp2, CRP_cAMP_CRPsite_mlcp2, Mlc_Mlcsite_mlcp2, mlcp2, Rate Law: cyt*km*CRP_cAMP_CRPsite_mlcp2/TCRPsite_mlcp2*(1-Mlc_Mlcsite_mlcp2/TMlcsite_mlcp2)*mlcp2
fast = 1.0E9 (60*s)^(-1); Kb=1.0E7 mol^(-1)*lReaction: CRP_cAMP + CRPsite_ptsIp1 => CRP_cAMP_CRPsite_ptsIp1; CRP_cAMP, CRPsite_ptsIp1, CRP_cAMP_CRPsite_ptsIp1, Rate Law: cyt*fast*(Kb*CRP_cAMP*CRPsite_ptsIp1-CRP_cAMP_CRPsite_ptsIp1)
km=17.95 (60*s)^(-1); TCRPsite_ptsHp1 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_ptsH; CRP_cAMP_CRPsite_ptsHp1, ptsHp1, CRP_cAMP_CRPsite_ptsHp1, ptsHp1, Rate Law: cyt*km*CRP_cAMP_CRPsite_ptsHp1/TCRPsite_ptsHp1*ptsHp1
kx=2.82E9 mol^(-1)*l*(60*s)^(-1)Reaction: IIA_P + HPr => IIA + HPr_P; HPr, IIA_P, Rate Law: cyt*kx*HPr*IIA_P
km=6.244 (60*s)^(-1); TCRPsite_ptsIp0 = 2.43E-10 mol*l^(-1); TMlcsite_ptsIp0 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_ptsI; CRP_cAMP_CRPsite_ptsIp0, Mlc_Mlcsite_ptsIp0, ptsIp0, CRP_cAMP_CRPsite_ptsIp0, Mlc_Mlcsite_ptsIp0, ptsIp0, Rate Law: cyt*km*CRP_cAMP_CRPsite_ptsIp0/TCRPsite_ptsIp0*(1-Mlc_Mlcsite_ptsIp0/TMlcsite_ptsIp0)*ptsIp0
km=1.875 (60*s)^(-1); TCRPsite_mlcp1 = 2.43E-10 mol*l^(-1); TMlcsite_mlcp1 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_mlc; CRP_cAMP_CRPsite_mlcp1, Mlc_Mlcsite_mlcp1, mlcp1, CRP_cAMP_CRPsite_mlcp1, Mlc_Mlcsite_mlcp1, mlcp1, Rate Law: cyt*km*(1-CRP_cAMP_CRPsite_mlcp1/TCRPsite_mlcp1)*(1-Mlc_Mlcsite_mlcp1/TMlcsite_mlcp1)*mlcp1
kmd=0.217 (60*s)^(-1)Reaction: mRNA_ptsG => ; mRNA_ptsG, Rate Law: cyt*kmd*mRNA_ptsG
km=71.8 (60*s)^(-1); TCRPsite_ptsHp0 = 2.43E-10 mol*l^(-1); TMlcsite_ptsHp0 = 2.43E-10 mol*l^(-1)Reaction: => mRNA_ptsH; CRP_cAMP_CRPsite_ptsHp0, Mlc_Mlcsite_ptsHp0, ptsHp0, CRP_cAMP_CRPsite_ptsHp0, Mlc_Mlcsite_ptsHp0, ptsHp0, Rate Law: cyt*km*CRP_cAMP_CRPsite_ptsHp0/TCRPsite_ptsHp0*(1-Mlc_Mlcsite_ptsHp0/TMlcsite_ptsHp0)*ptsHp0
Q=108000.0 (60*s)^(-1); Kmich=3.0E-4 mol*l^(-1)Reaction: EI + PEP => EI_P + Pyr; EI, PEP, Rate Law: cyt*2*Q*EI*PEP^2/(Kmich^2+PEP^2)

States:

NameDescription
Mlc Mlcsite ptsIp0[Protein mlc; Protein mlc]
CYA[Adenylate cyclase]
mRNA ptsI[messenger RNA]
HPr P[Phosphocarrier protein HPr]
CRPsite mlcp1[cAMP-activated global transcriptional regulator CRP; Protein mlc]
CRP cAMP[cAMP-activated global transcriptional regulator CRP; 3',5'-cyclic AMP]
IICB[Fused glucose-specific PTS enzymes: IIB component/IIC component2.7.1.69PTS glucose EIICB componentPTS glucose transporter subunit IIBCPTS glucose-specific subunit IIBCPTS system glucose-specific EIIBC component2.7.1.191PTS system glucose-specific EIICB componentPTS system glucose-specific transporter subunit IIBCPTS system, glucose-specific IIBC componentPtsG]
mRNA ptsH[messenger RNA]
cAMP[3',5'-cyclic AMP]
Mlc Mlcsite ptsGp2[Protein mlc; Protein mlc]
Mlc Mlcsite ptsGp1[Protein mlc; Protein mlc]
CRPsiteII crp[cAMP-activated global transcriptional regulator CRP]
CRP cAMP CRPsiteII crp[cAMP-activated global transcriptional regulator CRP; 3',5'-cyclic AMP]
EI[Phosphoenolpyruvate-protein phosphotransferase]
mRNA mlc[messenger RNA]
HPr[Phosphocarrier protein HPr]
Pyr[pyruvic acid]
Mlcsite mlcp1[Protein mlc]
EI P[Phosphoenolpyruvate-protein phosphotransferase]
IICB Mlc[Fused glucose-specific PTS enzymes: IIB component/IIC component2.7.1.69PTS glucose EIICB componentPTS glucose transporter subunit IIBCPTS glucose-specific subunit IIBCPTS system glucose-specific EIIBC component2.7.1.191PTS system glucose-specific EIICB componentPTS system glucose-specific transporter subunit IIBCPTS system, glucose-specific IIBC componentPtsG; Protein mlc]
CRPsiteI crp[cAMP-activated global transcriptional regulator CRP]
IIA[PTS system glucose-specific EIIA component]
Mlc Mlcsite mlcp2[Protein mlc; Protein mlc]
Mlc Mlcsite ptsHp0[Protein mlc; Protein mlc]
IICB P[Fused glucose-specific PTS enzymes: IIB component/IIC component2.7.1.69PTS glucose EIICB componentPTS glucose transporter subunit IIBCPTS glucose-specific subunit IIBCPTS system glucose-specific EIIBC component2.7.1.191PTS system glucose-specific EIICB componentPTS system glucose-specific transporter subunit IIBCPTS system, glucose-specific IIBC componentPtsG]
CRPsite mlcp2[cAMP-activated global transcriptional regulator CRP; Protein mlc]
CRP[cAMP-activated global transcriptional regulator CRP]
Mlc[Protein mlc]
PEP[phosphoenolpyruvic acid]
CRPsite ptsIp1[cAMP-activated global transcriptional regulator CRP; Phosphoenolpyruvate-protein phosphotransferase]
mRNA ptsG[messenger RNA]
Mlc Mlcsite mlcp1[Protein mlc; Protein mlc]
mRNA crr[messenger RNA]
CRP cAMP CRPsite cyaA[cAMP-activated global transcriptional regulator CRP; 3',5'-cyclic AMP]

Noble1984_SinoAtrialNode: MODEL0406151557v0.0.1

This a model from the article: A model of sino-atrial node electrical activity based on a modification of the DiFrance…

Details

DiFrancesco & Noble's (1984) equations (Phil. Trans. R. Soc. Lond. B (in the press.] have been modified to apply to the mammalian sino-atrial node. The modifications are based on recent experimental work. The modified equations successfully reproduce action potential and pacemaker activity in the node. Slightly different versions have been developed for peripheral regions that show a maximum diastolic potential near –75 mV and for central regions that do not hyperpolarize beyond –60 to –65 mV. Variations in extracellular potassium influence the frequency of pacemaker activity in the s.a. node model very much less than they do in the Purkinje fibre model. This corresponds well to the experimental observation that the node is less sensitive to external [K] than are Purkinje fibres. Activation of the Na-K exchange pump in the model by increasing intracellular sodium can suppress pacemaker activity. This phenomenon may contribute to the mechanism of overdrive suppression. link: http://identifiers.org/pubmed/6149553

Noble1998_VentricularCellModel_ModelA: MODEL1006230089v0.0.1

This a model from the article: Improved guinea-pig ventricular cell model incorporating a diadic space, IKr and IKs, a…

Details

The guinea-pig ventricular cell model, originally developed by Noble et al in 1991, has been greatly extended to include accumulation and depletion of calcium in a diadic space between the sarcolemma and the sarcoplasmic reticulum where, according to contempory understanding, the majority of calcium-induced calcium release is triggered. The calcium in this space is also assumed to play the major role in calcium-induced inactivation of the calcium current. Delayed potassium current equations have been developed to include the rapid (IKr) and slow (IKs) components of the delayed rectifier current based on the data of of Heath and Terrar, along with data from Sanguinetti and Jurkiewicz. Length- and tension-dependent changes in mechanical and electrophysiological processes have been incorporated as described recently by Kohl et al. Drug receptor interactions have started to be developed, using the sodium channel as the first target. The new model has been tested against experimental data on action potential clamp, and on force-interval and duration-interval relations; it has been found to reliably reproduce experimental observations. link: http://identifiers.org/pubmed/9487284

Noble1998_VentricularCellModel_ModelB: MODEL1006230080v0.0.1

This a model from the article: Improved guinea-pig ventricular cell model incorporating a diadic space, IKr and IKs, a…

Details

The guinea-pig ventricular cell model, originally developed by Noble et al in 1991, has been greatly extended to include accumulation and depletion of calcium in a diadic space between the sarcolemma and the sarcoplasmic reticulum where, according to contempory understanding, the majority of calcium-induced calcium release is triggered. The calcium in this space is also assumed to play the major role in calcium-induced inactivation of the calcium current. Delayed potassium current equations have been developed to include the rapid (IKr) and slow (IKs) components of the delayed rectifier current based on the data of of Heath and Terrar, along with data from Sanguinetti and Jurkiewicz. Length- and tension-dependent changes in mechanical and electrophysiological processes have been incorporated as described recently by Kohl et al. Drug receptor interactions have started to be developed, using the sodium channel as the first target. The new model has been tested against experimental data on action potential clamp, and on force-interval and duration-interval relations; it has been found to reliably reproduce experimental observations. link: http://identifiers.org/pubmed/9487284

Noble1998_VentricularCellModel_ModelC: MODEL1006230063v0.0.1

This a model from the article: Improved guinea-pig ventricular cell model incorporating a diadic space, IKr and IKs, a…

Details

The guinea-pig ventricular cell model, originally developed by Noble et al in 1991, has been greatly extended to include accumulation and depletion of calcium in a diadic space between the sarcolemma and the sarcoplasmic reticulum where, according to contempory understanding, the majority of calcium-induced calcium release is triggered. The calcium in this space is also assumed to play the major role in calcium-induced inactivation of the calcium current. Delayed potassium current equations have been developed to include the rapid (IKr) and slow (IKs) components of the delayed rectifier current based on the data of of Heath and Terrar, along with data from Sanguinetti and Jurkiewicz. Length- and tension-dependent changes in mechanical and electrophysiological processes have been incorporated as described recently by Kohl et al. Drug receptor interactions have started to be developed, using the sodium channel as the first target. The new model has been tested against experimental data on action potential clamp, and on force-interval and duration-interval relations; it has been found to reliably reproduce experimental observations. link: http://identifiers.org/pubmed/9487284

Nogales2008 - Genome-scale metabolic network of Pseudomonas putida (iJN746): MODEL1507180068v0.0.1

Nogales2008 - Genome-scale metabolic network of Pseudomonas putida (iJN746)This model is described in the article: [A g…

Details

BACKGROUND: Pseudomonas putida is the best studied pollutant degradative bacteria and is harnessed by industrial biotechnology to synthesize fine chemicals. Since the publication of P. putida KT2440's genome, some in silico analyses of its metabolic and biotechnology capacities have been published. However, global understanding of the capabilities of P. putida KT2440 requires the construction of a metabolic model that enables the integration of classical experimental data along with genomic and high-throughput data. The constraint-based reconstruction and analysis (COBRA) approach has been successfully used to build and analyze in silico genome-scale metabolic reconstructions. RESULTS: We present a genome-scale reconstruction of P. putida KT2440's metabolism, iJN746, which was constructed based on genomic, biochemical, and physiological information. This manually-curated reconstruction accounts for 746 genes, 950 reactions, and 911 metabolites. iJN746 captures biotechnologically relevant pathways, including polyhydroxyalkanoate synthesis and catabolic pathways of aromatic compounds (e.g., toluene, benzoate, phenylacetate, nicotinate), not described in other metabolic reconstructions or biochemical databases. The predictive potential of iJN746 was validated using experimental data including growth performance and gene deletion studies. Furthermore, in silico growth on toluene was found to be oxygen-limited, suggesting the existence of oxygen-efficient pathways not yet annotated in P. putida's genome. Moreover, we evaluated the production efficiency of polyhydroxyalkanoates from various carbon sources and found fatty acids as the most prominent candidates, as expected. CONCLUSION: Here we presented the first genome-scale reconstruction of P. putida, a biotechnologically interesting all-surrounder. Taken together, this work illustrates the utility of iJN746 as i) a knowledge-base, ii) a discovery tool, and iii) an engineering platform to explore P. putida's potential in bioremediation and bioplastic production. link: http://identifiers.org/pubmed/18793442

Nogales2012 - Genome-scale metabolic network of Synechocystis sp. PCC6803 (iJN678): MODEL1507180046v0.0.1

Nogales2012 - Genome-scale metabolic network of Synechocystis sp. (iJN678)This model is described in the article: [Deta…

Details

Photosynthesis has recently gained considerable attention for its potential role in the development of renewable energy sources. Optimizing photosynthetic organisms for biomass or biofuel production will therefore require a systems understanding of photosynthetic processes. We reconstructed a high-quality genome-scale metabolic network for Synechocystis sp. PCC6803 that describes key photosynthetic processes in mechanistic detail. We performed an exhaustive in silico analysis of the reconstructed photosynthetic process under different light and inorganic carbon (Ci) conditions as well as under genetic perturbations. Our key results include the following. (i) We identified two main states of the photosynthetic apparatus: a Ci-limited state and a light-limited state. (ii) We discovered nine alternative electron flow pathways that assist the photosynthetic linear electron flow in optimizing the photosynthesis performance. (iii) A high degree of cooperativity between alternative pathways was found to be critical for optimal autotrophic metabolism. Although pathways with high photosynthetic yield exist for optimizing growth under suboptimal light conditions, pathways with low photosynthetic yield guarantee optimal growth under excessive light or Ci limitation. (iv) Photorespiration was found to be essential for the optimal photosynthetic process, clarifying its role in high-light acclimation. Finally, (v) an extremely high photosynthetic robustness drives the optimal autotrophic metabolism at the expense of metabolic versatility and robustness. The results and modeling approach presented here may promote a better understanding of the photosynthetic process. They can also guide bioengineering projects toward optimal biofuel production in photosynthetic organisms. link: http://identifiers.org/pubmed/22308420

Nookaew2008_Yeast_MetabolicNetwork_iIN800: MODEL1002240000v0.0.1

This is a reconstruction of the metabolic network of the yeast Saccharomyces cerevisiae as described in the article:…

Details

BACKGROUND: Up to now, there have been three published versions of a yeast genome-scale metabolic model: iFF708, iND750 and iLL672. All three models, however, lack a detailed description of lipid metabolism and thus are unable to be used as integrated scaffolds for gaining insights into lipid metabolism from multilevel omic measurement technologies (e.g. genome-wide mRNA levels). To overcome this limitation, we reconstructed a new version of the Saccharomyces cerevisiae genome-scale model, iIN800 that includes a more rigorous and detailed description of lipid metabolism. RESULTS: The reconstructed metabolic model comprises 1446 reactions and 1013 metabolites. Beyond incorporating new reactions involved in lipid metabolism, we also present new biomass equations that improve the predictive power of flux balance analysis simulations. Predictions of both growth capability and large scale in silico single gene deletions by iIN800 were consistent with experimental data. In addition, 13C-labeling experiments validated the new biomass equations and calculated intracellular fluxes. To demonstrate the applicability of iIN800, we show that the model can be used as a scaffold to reveal the regulatory importance of lipid metabolism precursors and intermediates that would have been missed in previous models from transcriptome datasets. CONCLUSION: Performing integrated analyses using iIN800 as a network scaffold is shown to be a valuable tool for elucidating the behavior of complex metabolic networks, particularly for identifying regulatory targets in lipid metabolism that can be used for industrial applications or for understanding lipid disease states. link: http://identifiers.org/pubmed/18687109

Norel1990 - MPF and Cyclin Oscillations: BIOMD0000000728v0.0.1

A mathematical model of cell cycle progression is presented, which integrates recent biochemical information on the inte…

Details

A mathematical model of cell cycle progression is presented, which integrates recent biochemical information on the interaction of the maturation promotion factor (MPF) and cyclin. The model retrieves the dynamics observed in early embryos and explains how multiple cycles of MPF activity can be produced and how the internal clock that determines durations and number of cycles can be adjusted by modulating the rate of change in MPF or cyclin concentrations. Experiments are suggested for verifying the role of MPF activity in determining the length of the somatic cell cycle. link: http://identifiers.org/pubmed/1825521

Parameters:

NameDescription
i = 1.2Reaction: => C, Rate Law: cell*i
e = 3.46616Reaction: => M; C, Rate Law: cell*e*C
f = 1.0Reaction: => M; C, Rate Law: cell*f*C*M^2
g = 10.0Reaction: M =>, Rate Law: cell*g*M/(M+1)

States:

NameDescription
M[MPF complex]
C[Guanidine]

Novak1993 - Cell cycle M-phase control: BIOMD0000000107v0.0.1

Novak1993 - Cell cycle M-phase control The model reproduces Figure 9 of the paper. Please note that active MPF and cycli…

Details

To contribute to a deeper understanding of M-phase control in eukaryotic cells, we have constructed a model based on the biochemistry of M-phase promoting factor (MPF) in Xenopus oocyte extracts, where there is evidence for two positive feedback loops (MPF stimulates its own production by activating Cdc25 and inhibiting Wee1) and a negative feedback loop (MPF stimulates its own destruction by indirectly activating the ubiquitin pathway that degrades its cyclin subunit). To uncover the full dynamical possibilities of the control system, we translate the regulatory network into a set of differential equations and study these equations by graphical techniques and computer simulation. The positive feedback loops in the model account for thresholds and time lags in cyclin-induced and MPF-induced activation of MPF, and the model can be fitted quantitatively to these experimental observations. The negative feedback loop is consistent with observed time lags in MPF-induced cyclin degradation. Furthermore, our model indicates that there are two possible mechanisms for autonomous oscillations. One is driven by the positive feedback loops, resulting in phosphorylation and abrupt dephosphorylation of the Cdc2 subunit at an inhibitory tyrosine residue. These oscillations are typical of oocyte extracts. The other type is driven by the negative feedback loop, involving rapid cyclin turnover and negligible phosphorylation of the tyrosine residue of Cdc2. The early mitotic cycles of intact embryos exhibit such characteristics. In addition, by assuming that unreplicated DNA interferes with M-phase initiation by activating the phosphatases that oppose MPF in the positive feedback loops, we can simulate the effect of addition of sperm nuclei to oocyte extracts, and the lengthening of cycle times at the mid-blastula transition of intact embryos. link: http://identifiers.org/pubmed/8126097

Parameters:

NameDescription
kcak = 0.25Reaction: dimer => dimer_p, Rate Law: kcak*dimer
k25 = 0.0Reaction: p_dimer => dimer, Rate Law: k25*p_dimer
k2 = 0.0Reaction: p_dimer_p =>, Rate Law: k2*p_dimer_p
k3 = 0.01Reaction: cyclin + cdc2 => dimer, Rate Law: k3*cyclin*cdc2
kinh = 0.025Reaction: p_dimer_p => p_dimer, Rate Law: kinh*p_dimer_p
total_UbE = 1.0Reaction: UbE = total_UbE-UbE_star, Rate Law: missing
total_IE = 1.0Reaction: IE = total_IE-IE_p, Rate Law: missing
K_a = 0.1; ka = 0.01; total_cdc25 = 1.0Reaction: cdc25 => cdc25_p; dimer_p, Rate Law: ka*dimer_p*(total_cdc25-cdc25_p)/((K_a+total_cdc25)-cdc25_p)
k1AA = 1.0Reaction: => cyclin, Rate Law: k1AA
total_wee1 = 1.0; K_e = 0.3; ke = 0.0133Reaction: wee1 => wee1_p; dimer_p, Rate Law: ke*dimer_p*(total_wee1-wee1_p)/((K_e+total_wee1)-wee1_p)
total_wee1 = 1.0Reaction: wee1 = total_wee1-wee1_p, Rate Law: missing
K_f = 0.3; kfPPase = 0.1Reaction: wee1_p => wee1, Rate Law: kfPPase*wee1_p/(K_f+wee1_p)
kg = 0.0065; K_g = 0.01; total_IE = 1.0Reaction: IE => IE_p; dimer_p, Rate Law: kg*dimer_p*(total_IE-IE_p)/((K_g+total_IE)-IE_p)
K_d = 0.01; kd_anti_IE = 0.095Reaction: UbE_star => UbE, Rate Law: kd_anti_IE*UbE_star/(K_d+UbE_star)
kc = 0.1; K_c = 0.01; total_UbE = 1.0Reaction: UbE => UbE_star; IE_p, Rate Law: kc*IE_p*(total_UbE-UbE_star)/((K_c+total_UbE)-UbE_star)
total_cdc2 = 100.0Reaction: cdc2 = total_cdc2-(dimer+p_dimer+p_dimer_p+dimer_p), Rate Law: missing
kbPPase = 0.125; K_b = 0.1Reaction: cdc25_p => cdc25, Rate Law: kbPPase*cdc25_p/(K_b+cdc25_p)
K_h = 0.01; khPPAse = 0.087Reaction: IE_p => IE, Rate Law: khPPAse*IE_p/(K_h+IE_p)
kwee = 0.0Reaction: dimer_p => p_dimer_p; wee1, Rate Law: kwee*dimer_p
total_cdc25 = 1.0Reaction: cdc25 = total_cdc25-cdc25_p, Rate Law: missing

States:

NameDescription
wee1 p[Wee1-like protein kinase 2-A]
cdc2[Cyclin-dependent kinase 1-A]
p dimer p[Cyclin-dependent kinase 1-A; IPR015454]
cdc25 p[M-phase inducer phosphatase 3]
dimer p[Cyclin-dependent kinase 1-A; IPR015454]
dimer[Cyclin-dependent kinase 1-A; IPR015454]
cdc25[M-phase inducer phosphatase 3]
IEintermediary enzyme
UbE star[ubiquitin conjugating enzyme complex]
UbE[ubiquitin conjugating enzyme complex]
cyclin[IPR015454]
IE pphosphorylated intermediary enzyme
wee1[Wee1-like protein kinase 2-A]
p dimer[Cyclin-dependent kinase 1-A; IPR015454]

Novak1997 - Cell Cycle: BIOMD0000000007v0.0.1

Novak1997 - Cell CycleModeling the control of DNA replication in fission yeast. This model is described in the article:…

Details

A central event in the eukaryotic cell cycle is the decision to commence DNA replication (S phase). Strict controls normally operate to prevent repeated rounds of DNA replication without intervening mitoses ("endoreplication") or initiation of mitosis before DNA is fully replicated ("mitotic catastrophe"). Some of the genetic interactions involved in these controls have recently been identified in yeast. From this evidence we propose a molecular mechanism of "Start" control in Schizosaccharomyces pombe. Using established principles of biochemical kinetics, we compare the properties of this model in detail with the observed behavior of various mutant strains of fission yeast: wee1(-) (size control at Start), cdc13Delta and rum1(OP) (endoreplication), and wee1(-) rum1Delta (rapid division cycles of diminishing cell size). We discuss essential features of the mechanism that are responsible for characteristic properties of Start control in fission yeast, to expose our proposal to crucial experimental tests. link: http://identifiers.org/pubmed/9256450

Parameters:

NameDescription
k4 = 0.1875Reaction: R =>, Rate Law: k4*R
Kmu = 0.01; ku = 0.2; kur = 0.1; Kmur = 0.01Reaction: UbEB => UbE; IE, Rate Law: IE*ku*UbEB/(Kmu+UbEB)-kur*UbE/(Kmur+UbE)
k3 = 0.09375Reaction: => R, Rate Law: k3
k6 = NaNReaction: G1K => ; UbE2, Rate Law: G1K*k6
Kmwr = 0.1; kw = 1.0; Kmw = 0.1; kwr = 0.25Reaction: Wee1B => Wee1; MPF, Rate Law: kwr*Wee1B/(Kmwr+Wee1B)-kw*MPF*Wee1/(Kmw+Wee1)
k7r = 0.1; k7 = 100.0Reaction: G2K + R => G2R, Rate Law: G2K*k7*R-G2R*k7r
kwee = NaN; k25 = NaNReaction: G2K => PG2; Wee1, Cdc25, Rate Law: G2K*kwee-k25*PG2
beta = 0.05Reaction: MPF = G2K+beta*PG2, Rate Law: missing
kur2 = 0.3; Kmu2 = 0.05; ku2 = 1.0; Kmur2 = 0.05Reaction: UbE2B => UbE2; MPF, Rate Law: ku2*MPF*UbE2B/(Kmu2+UbE2B)-kur2*UbE2/(Kmur2+UbE2)
k1 = 0.015Reaction: => G2K, Rate Law: k1
k8 = 10.0; k8r = 0.1Reaction: G1K + R => G1R, Rate Law: G1K*k8*R-G1R*k8r
k2prime = 0.05Reaction: G2R => R, Rate Law: G2R*k2prime
Kmp = 0.001; Mass = 0.49; kp = 3.25Reaction: R => ; SPF, Rate Law: kp*Mass*R*SPF/(Kmp+R)
kc = 1.0; Kmcr = 0.1; Kmc = 0.1; kcr = 0.25Reaction: Cdc25B => Cdc25; MPF, Rate Law: Cdc25B*kc*MPF/(Cdc25B+Kmc)-Cdc25*kcr/(Cdc25+Kmcr)
k6prime = 0.0Reaction: G1R => R, Rate Law: G1R*k6prime
Kmi = 0.01; Kmir = 0.01; ki = 0.4; kir = 0.1Reaction: IEB => IE; MPF, Rate Law: IEB*ki*MPF/(IEB+Kmi)-IE*kir/(IE+Kmir)
k5 = 0.00175Reaction: => G1K, Rate Law: k5
alpha = 0.25; Cig1 = 0.0Reaction: SPF = Cig1+alpha*G1K+MPF, Rate Law: missing
k2 = NaNReaction: G2K => ; UbE, Rate Law: G2K*k2

States:

NameDescription
MPF[G2/mitotic-specific cyclin cdc13; Cyclin-dependent kinase 1]
G2R[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin cdc13; Cyclin-dependent kinase inhibitor rum1]
G2K[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin cdc13]
PG2R[G2/mitotic-specific cyclin cdc13; Cyclin-dependent kinase 1; Cyclin-dependent kinase inhibitor rum1]
IEBBoundIntermediaryEnzyme
UbE[proteasome complex]
Cdc13Total[G2/mitotic-specific cyclin cdc13]
Cig2Total[G2/mitotic-specific cyclin cig2]
G1K[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin cig2]
SPF[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin cdc13; G2/mitotic-specific cyclin cig2]
Wee1[Mitosis inhibitor protein kinase mik1; Mitosis inhibitor protein kinase wee1]
UbE2[proteasome complex]
Cdc25B[M-phase inducer phosphatase]
Wee1B[Mitosis inhibitor protein kinase wee1; Mitosis inhibitor protein kinase mik1]
PG2[G2/mitotic-specific cyclin cdc13; Cyclin-dependent kinase 1]
IEIntermediaryEnzyme
Cdc25[M-phase inducer phosphatase]
G1R[Cyclin-dependent kinase 1; G2/mitotic-specific cyclin cig2; Cyclin-dependent kinase inhibitor rum1]
UbEB[proteasome complex]
R[Cyclin-dependent kinase inhibitor rum1]
Rum1Total[Cyclin-dependent kinase inhibitor rum1]
UbE2B[proteasome complex]

Novak1998 - Mathematical model of fission yeast cell cycle: MODEL2003190004v0.0.1

Mathematical model of the fission yeast cell cycle with checkpoint controls at the G1/S, G2/M and metaphase/anaphase tra…

Details

All events of the fission yeast cell cycle can be orchestrated by fluctuations of a single cyclin-dependent protein kinase, the Cdc13/Cdc2 heterodimer. The G1/S transition is controlled by interactions of Cdc13/Cdc2 and its stoichiometric inhibitor, Rum1. The G2/M transition is regulated by a kinase-phosphatase pair, Wee1 and Cdc25, which determine the phosphorylation state of the Tyr-15 residue of Cdc2. The meta/anaphase transition is controlled by interactions between Cdc13/Cdc2 and the anaphase promoting complex, which labels Cdc13 subunits for proteolysis. We construct a mathematical model of fission yeast growth and division that encompasses all three crucial checkpoint controls. By numerical simulations we show that the model is consistent with a broad selection of cell cycle mutants, and we predict the phenotypes of several multiple-mutant strains that have not yet been constructed. link: http://identifiers.org/pubmed/9652094

Novak1998-Model scenarios for evolution of the eukaryotic cell cycle.: MODEL2005040001v0.0.1

Progress through the division cycle of present day eukaryotic cells is controlled by a complex network consisting of (i)…

Details

Progress through the division cycle of present day eukaryotic cells is controlled by a complex network consisting of (i) cyclin-dependent kinases (CDKs) and their associated cyclins, (ii) kinases and phosphatases that regulate CDK activity, and (iii) stoichiometric inhibitors that sequester cyclin-CDK dimers. Presumably regulation of cell division in the earliest ancestors of eukaryotes was a considerably simpler affair. Nasmyth (1995) recently proposed a mechanism for control of a putative, primordial, eukaryotic cell cycle, based on antagonistic interactions between a cyclin-CDK and the anaphase promoting complex (APC) that labels the cyclin subunit for proteolysis. We recast this idea in mathematical form and show that the model exhibits hysteretic behaviour between alternative steady states: a Gl-like state (APC on, CDK activity low, DNA unreplicated and replication complexes assembled) and an S/M-like state (APC off, CDK activity high, DNA replicated and replication complexes disassembled). In our model, the transition from G1 to S/M ('Start') is driven by cell growth, and the reverse transition ('Finish') is driven by completion of DNA synthesis and proper alignment of chromosomes on the metaphase plate. This simple and effective mechanism for coupling growth and division and for accurately copying and partitioning a genome consisting of numerous chromosomes, each with multiple origins of replication, could represent the core of the eukaryotic cell cycle. Furthermore, we show how other controls could be added to this core and speculate on the reasons why stoichiometric inhibitors and CDK inhibitory phosphorylation might have been appended to the primitive alternation between cyclin accumulation and degradation. link: http://identifiers.org/pubmed/10098216

Novak2001_FissionYeast_CellCycle: BIOMD0000000111v0.0.1

The model reproduces the time evolution of several species as depicted in Fig 4 of the paper. Events have been used to r…

Details

Much is known about the genes and proteins controlling the cell cycle of fission yeast. Can these molecular components be spun together into a consistent mechanism that accounts for the observed behavior of growth and division in fission yeast cells? To answer this question, we propose a mechanism for the control system, convert it into a set of 14 differential and algebraic equations, study these equations by numerical simulation and bifurcation theory, and compare our results to the physiology of wild-type and mutant cells. In wild-type cells, progress through the cell cycle (G1–>S–>G2–>M) is related to cyclic progression around a hysteresis loop, driven by cell growth and chromosome alignment on the metaphase plate. However, the control system operates much differently in double-mutant cells, wee1(-) cdc25Delta, which are defective in progress through the latter half of the cell cycle (G2 and M phases). These cells exhibit "quantized" cycles (interdivision times clustering around 90, 160, and 230 min). We show that these quantized cycles are associated with a supercritical Hopf bifurcation in the mechanism, when the wee1 and cdc25 genes are disabled. (c) 2001 American Institute of Physics. link: http://identifiers.org/pubmed/12779461

Parameters:

NameDescription
k1 = 0.03 min_invReaction: => cdc13T; M, Rate Law: k1*M
k8 = 0.25 min_inv; J8 = 0.001 dimensionlessReaction: slp1 =>, Rate Law: k8*slp1/(J8+slp1)
k6 = 0.1 min_invReaction: slp1T =>, Rate Law: k6*slp1T
J5 = 0.3 dimensionless; k5_double_prime = 0.3 min_inv; k5_prime = 0.005 min_invReaction: => slp1T; MPF, Rate Law: k5_prime+k5_double_prime*MPF^4/(J5^4+MPF^4)
J9 = 0.01 dimensionless; k9 = 0.1 min_invReaction: => IEP; MPF, Rate Law: k9*MPF*(1-IEP)/((J9+1)-IEP)
k3_prime = 1.0 min_inv; k3_double_prime = 10.0 min_inv; J3 = 0.01 dimensionlessReaction: => ste9; slp1, Rate Law: (k3_prime+k3_double_prime*slp1)*(1-ste9)/((J3+1)-ste9)
kwee = 0.0 min_invReaction: => preMPF; cdc13T, Rate Law: kwee*(cdc13T-preMPF)
mu = 0.005 min_invReaction: => M, Rate Law: mu*M
k2_double_prime = 1.0 min_inv; k2_prime = 0.03 min_inv; k2_triple_prime = 0.1 min_invReaction: preMPF => ; ste9, slp1, Rate Law: (k2_prime+k2_double_prime*ste9+k2_triple_prime*slp1)*preMPF
k7 = 1.0 min_inv; J7 = 0.001 dimensionlessReaction: => slp1; IEP, slp1T, Rate Law: k7*IEP*(slp1T-slp1)/((J7+slp1T)-slp1)
k4_prime = 2.0 min_inv; k4 = 35.0 min_inv; J4 = 0.01 dimensionlessReaction: ste9 => ; SK, MPF, Rate Law: (k4_prime*SK+k4*MPF)*ste9/(J4+ste9)
J10 = 0.01 dimensionless; k10 = 0.04 min_invReaction: IEP =>, Rate Law: k10*IEP/(J10+IEP)
k13 = 0.1 min_inv; TF = 0.0 dimensionlessReaction: => SK, Rate Law: k13*TF
Trimer = 0.0 dimensionlessReaction: MPF = (cdc13T-preMPF)*(cdc13T-Trimer)/cdc13T, Rate Law: missing
k14 = 0.1 min_invReaction: SK =>, Rate Law: k14*SK
k11 = 0.1 min_invReaction: => rum1T, Rate Law: k11
k12 = 0.01 min_inv; k12_prime = 1.0 min_inv; k12_double_prime = 3.0 min_invReaction: rum1T => ; SK, MPF, Rate Law: (k12+k12_prime*SK+k12_double_prime*MPF)*rum1T
k25 = 0.0 min_invReaction: preMPF =>, Rate Law: k25*preMPF

States:

NameDescription
preMPF[Cyclin-dependent kinase 1; IPR015454]
MPF[Cyclin-dependent kinase 1; IPR015454]
rum1T[Cyclin-dependent kinase inhibitor rum1]
MCell Mass
IEPIEP
SK[G2/mitotic-specific cyclin cig1]
slp1T[WD repeat-containing protein slp1]
slp1[WD repeat-containing protein slp1]
ste9[WD repeat-containing protein srw1]
cdc13T[IPR015454]

Novak2004-A Model for Restriction Point Control of the Mammalian Cell Cycle: MODEL2006080001v0.0.1

<notes xmlns="http://www.sbml.org/sbml/level2/version4"> <body xmlns="http://www.w3.org/1…

Details

Inhibition of protein synthesis by cycloheximide blocks subsequent division of a mammalian cell, but only if the cell is exposed to the drug before the "restriction point" (i.e. within the first several hours after birth). If exposed to cycloheximide after the restriction point, a cell proceeds with DNA synthesis, mitosis and cell division and halts in the next cell cycle. If cycloheximide is later removed from the culture medium, treated cells will return to the division cycle, showing a complex pattern of division times post-treatment, as first measured by Zetterberg and colleagues. We simulate these physiological responses of mammalian cells to transient inhibition of growth, using a set of nonlinear differential equations based on a realistic model of the molecular events underlying progression through the cell cycle. The model relies on our earlier work on the regulation of cyclin-dependent protein kinases during the cell division cycle of yeast. The yeast model is supplemented with equations describing the effects of retinoblastoma protein on cell growth and the synthesis of cyclins A and E, and with a primitive representation of the signaling pathway that controls synthesis of cyclin D. link: http://identifiers.org/pubmed/15363676

Nowak1996_HostResponse_InfectiousAgents: MODEL1006230050v0.0.1

This a model from the article: Population dynamics of immune responses to persistent viruses. Nowak MA, Bangham CR.…

Details

Mathematical models, which are based on a firm understanding of biological interactions, can provide nonintuitive insights into the dynamics of host responses to infectious agents and can suggest new avenues for experimentation. Here, a simple mathematical approach is developed to explore the relation between antiviral immune responses, virus load, and virus diversity. The model results are compared to data on cytotoxic T cell responses and viral diversity in infections with the human T cell leukemia virus (HTLV-1) and the human immunodeficiency virus (HIV-1). link: http://identifiers.org/pubmed/8600540